Kassirer J, Angell M. Losing weight – an ill-fated New Year’s resolution. N Engl J Med. 1998;338(1):52–4. https://pubmed.ncbi.nlm.nih.gov/9414332/
Nelson TD. Promoting healthy aging by confronting ageism. Am Psychol. 2016;71(4):276–82. https://pubmed.ncbi.nlm.nih.gov/27159434/
Binstock RH. Anti-aging medicine and research: a realm of conflict and profound societal implications. J Gerontol A Biol Sci Med Sci. 2004;59(6):B523–33. https://pubmed.ncbi.nlm.nih.gov/15215257/
Reddy SSK, Chaiban JT. The endocrinology of aging: a key to longevity “great expectations.” Endocr Pract. 2017;23(9):1110–9. https://pubmed.ncbi.nlm.nih.gov/28704100/
Kristjuhan Ü. Real aging retardation in humans through diminishing risks to health. Ann N Y Acad Sci. 2007;1119:122–8. https://pubmed.ncbi.nlm.nih.gov/18056961/
Roe DA. Health foods and supplements for the elderly. Who can say no? N Y State J Med. 1993;93(2):109–12. https://pubmed.ncbi.nlm.nih.gov/8455823/
Perls TT. Anti-aging quackery: human growth hormone and tricks of the trade – more dangerous than ever. J Gerontol A Biol Sci Med Sci. 2004;59(7):682–91. https://pubmed.ncbi.nlm.nih.gov/15304532/
United States Senate, Special Committee on Aging. Senate hearing 107–190. Swindlers, hucksters and snake oil salesmen: hype and hope of marketing anti-aging products to seniors. U.S. Government Printing Office. September 10, 2001.; https://www.govinfo.gov/content/pkg/CHRG-107shrg76011/html/CHRG-107shrg76011.htm
United States Congress House of Representatives, Select Committee on Aging. Quackery: a $10 billion scandal. U.S. Government Printing Office. May 31, 1984.; https://centerforinquiry.org/wp-content/uploads/sites/33/quackwatch/pepper-report.pdf
Newton JP. Anti-ageing – fact, fiction or faction? Gerodontology. 2011;28(3):163–4. https://pubmed.ncbi.nlm.nih.gov/21843158/
Anti-aging treatment claims: the promises vs. the science. Consum Rep. 2015;80(8):15–7. https://pubmed.ncbi.nlm.nih.gov/26159004/
McConnel C, Turner L. Medicine, ageing and human longevity: the economics and ethics of anti-ageing interventions. EMBO Rep. 2005;6(S1):S59–62. https://pubmed.ncbi.nlm.nih.gov/15995665/
Anti-aging treatment claims: the promises vs. the science. Consum Rep. 2015;80(8):15–7. https://pubmed.ncbi.nlm.nih.gov/26159004/
Wick G. “Anti-aging” medicine: does it exist? A critical discussion of “anti-aging health products.” Exp Gerontol. 2002;37(8–9):1137–40. https://pubmed.ncbi.nlm.nih.gov/12213565/
Caulfield T. Blinded by science. The Walrus. https://thewalrus.ca/blinded-by-science/. Published September 12, 2011. Updated April 19, 2020. Accessed January 22, 2023.; https://thewalrus.ca/blinded-by-science/
Winslow R. The radium water worked fine until his jaw fell off. Wall Street Journal. August 1, 1990:A1.; https://web.archive.org/web/20170216124222/ https://case.edu/affil/MeMA/MCA/11-20/1991-Nov.pdf
Turner L. The US direct-to-consumer marketplace for autologous stem cell interventions. Perspect Biol Med. 2018;61(1):7–24. https://pubmed.ncbi.nlm.nih.gov/29805145/
Murray IR, Chahla J, Frank RM, et al. Rogue stem cell clinics. Bone Joint J. 2020;102-B(2):148–54. https://pubmed.ncbi.nlm.nih.gov/32009438/
Olshansky SJ, Hayflick L, Carnes BA. No truth to the fountain of youth. Sci Am. 2002;286(6):92–5. https://pubmed.ncbi.nlm.nih.gov/12030096/
Epstein D. Anti-aging doctors sue professors. Inside Higher Ed. https://www.insidehighered.com/news/2005/06/21/anti-aging-doctors-sue-professors. Published June 21, 2005. Accessed January 22, 2023.; https://www.insidehighered.com/news/2005/06/21/anti-aging-doctors-sue-professors
MacGregor C, Petersen A, Parker C. Hyping the market for ‘anti-ageing’ in the news: from medical failure to success in self-transformation. BioSocieties. 2018;13(1):64–80. https://link.springer.com/article/10.1057/s41292-017-0052-5
The American Academy of Anti-Aging Medicine’s official position statement on the truth about human aging intervention. American Academy of Anti-Aging Medicine. https://mail.anme.com.mx/modulacion/extra/official_position_statement.pdf. Published June 2002. Accessed September 26, 2022.; https://mail.anme.com.mx/modulacion/extra/official_position_statement.pdf
Binstock RH. The war on “anti-aging medicine.” Gerontologist. 2003;43(1):4–14. https://pubmed.ncbi.nlm.nih.gov/12604740/
Find an anti-aging product or service. World Health Network. https://web.archive.org/web/20020402011937/http://www.worldhealth.net/cgi-local/DB_Search/db_search.cgi?setup_file=whn_productsa.setup.cgi. Accessed January 31, 2023.; https://web.archive.org/web/20020402011937/http://www.worldhealth.net/cgi-local/DB_Search/db_search.cgi?setup_file=whn_productsa.setup.cgi
Zs-Nagy I. Is consensus in anti-aging medical intervention an elusive expectation or a realistic goal? Arch Gerontol Geriatr. 2009;48(3):271–5. https://pubmed.ncbi.nlm.nih.gov/19269702/
Binstock RH. The war on “anti-aging medicine.” Gerontologist. 2003;43(1):4–14. https://pubmed.ncbi.nlm.nih.gov/12604740/
The American Academy of Anti-Aging Medicine’s official position statement on the truth about human aging intervention. American Academy of Anti-Aging Medicine. https://mail.anme.com.mx/modulacion/extra/official_position_statement.pdf. Published June 2002. Accessed September 26, 2022.; https://mail.anme.com.mx/modulacion/extra/official_position_statement.pdf
Walker RF. On the evolution of anti-aging medicine. Clin Interv Aging. 2006;1(3):201–3. https://pubmed.ncbi.nlm.nih.gov/18046871/
Rattan SIS. Anti-ageing strategies: prevention or therapy? EMBO Rep. 2005;6(Suppl 1):S25–9. https://pubmed.ncbi.nlm.nih.gov/15995657/
Rae MJ. All hype, no hope? Excessive pessimism in the “anti-aging medicine” special sections. J Gerontol A Biol Sci Med Sci. 2005;60(2):139–40. https://academic.oup.com/biomedgerontology/article/60/2/139/563273
Mehra MR, Desai SS, Kuy S, Henry TD, Patel AN. Retraction: cardiovascular disease, drug therapy, and mortality in COVID-19. N Engl J Med. DOI: 10.1056/nejmoa2007621. N Engl J Med. 2020;382(26):2582. https://pubmed.ncbi.nlm.nih.gov/32501665/
Mehra MR, Ruschitzka F, Patel AN. Retraction – Hydroxychloroquine or chloroquine with or without a macrolide for treatment of COVID-19: a multinational registry analysis. Lancet. 2020;395(10240):1820. https://pubmed.ncbi.nlm.nih.gov/32450107/
Miller RA. Extending life: scientific prospects and political obstacles. Milbank Q. 2002;80(1):155–74. https://pubmed.ncbi.nlm.nih.gov/11933792/
Berzlanovich AM, Keil W, Waldhoer T, Sim E, Fasching P, Fazeny-DBerzl B. Do centenarians die healthy? An autopsy study. J Gerontol A Biol Sci Med Sci. 2005;60(7):862–5. https://pubmed.ncbi.nlm.nih.gov/16079208/
Gessert CE, Elliott BA, Haller IV. Dying of old age: an examination of death certificates of Minnesota centenarians. J Am Geriatr Soc. 2002;50(9):1561–5. https://pubmed.ncbi.nlm.nih.gov/12383155/
Wilson DM, Cohen J, Birch S, et al. “No one dies of old age”: implications for research, practice, and policy. J Palliat Care. 2011;27(2):148–56. https://journals.sagepub.com/doi/10.1177/082585971102700211
Berzlanovich AM, Missliwetz J, Sim E, et al. Unexpected out-of-hospital deaths in persons aged 85 years or older: an autopsy study of 1886 patients. Am J Med. 2003;114(5):365–9. https://pubmed.ncbi.nlm.nih.gov/12714125/
John SM, Koelmeyer TD. The forensic pathology of nonagenarians and centenarians: do they die of old age? (The Auckland experience). Am J Forensic Med Pathol. 2001;22(2):150–4. https://pubmed.ncbi.nlm.nih.gov/11394748/
Blagosklonny MV. Answering the ultimate question “what is the proximal cause of aging?” Aging (Albany NY). 2012;4(12):861–77. https://pubmed.ncbi.nlm.nih.gov/23425777/
Murray CJL, Barber RM, Foreman KJ, et al. Global, regional, and national disability-adjusted life years (DALYs) for 306 diseases and injuries and healthy life expectancy (HALE) for 188 countries, 1990–2013: quantifying the epidemiological transition. Lancet. 2015;386(10009):2145–91. https://pubmed.ncbi.nlm.nih.gov/26321261/
Writing Group Members, Roger VL, Go AS, et al. Heart disease and stroke statistics—2012 update: a report from the American Heart Association. Circulation. 2012;125(1): e2-e220. https://pubmed.ncbi.nlm.nih.gov/22179539/
Murphy SL, Kochanek KD, Xu J, Arias E. Mortality in the United States, 2020. NCHS Data Brief. 2021;(427):1–8. https://pubmed.ncbi.nlm.nih.gov/34978528/
Murray CJL, Barber RM, Foreman KJ, et al. Global, regional, and national disability-adjusted life years (DALYs) for 306 diseases and injuries and healthy life expectancy (HALE) for 188 countries, 1990–2013: quantifying the epidemiological transition. Lancet. 2015;386(10009):2145–91. https://pubmed.ncbi.nlm.nih.gov/26321261/
Foreman KJ, Marquez N, Dolgert A, et al. Forecasting life expectancy, years of life lost, and all-cause and cause-specific mortality for 250 causes of death: reference and alternative scenarios for 2016–40 for 195 countries and territories. Lancet. 2018;392(10159):2052–90. https://pubmed.ncbi.nlm.nih.gov/30340847/
Kaeberlein M. The biology of aging: citizen scientists and their pets as a bridge between research on model organisms and human subjects. Vet Pathol. 2016;53(2):291–8. https://pubmed.ncbi.nlm.nih.gov/26077786/
Zainabadi K. A brief history of modern aging research. Exp Gerontol. 2018;104:35–42. https://pubmed.ncbi.nlm.nih.gov/29355705/
Milman S, Barzilai N. Dissecting the mechanisms underlying unusually successful human health span and life span. Cold Spring Harb Perspect Med. 2015;6(1):a025098. https://pubmed.ncbi.nlm.nih.gov/26637439/
Iyen B, Qureshi N, Weng S, et al. Sex differences in cardiovascular morbidity associated with familial hypercholesterolaemia: a retrospective cohort study of the UK Simon Broome register linked to national hospital records. Atherosclerosis. 2020;315:131–7. https://pubmed.ncbi.nlm.nih.gov/33187671/
Tsao CW, Aday AW, Almarzooq ZI, et al. Heart disease and stroke statistics—2022 update: a report from the American Heart Association. Circulation. 2022;145(8):e153–639. https://pubmed.ncbi.nlm.nih.gov/35078371/
Jortveit J, Pripp AH, Langørgen J, Halvorsen S. Incidence, risk factors and outcome of young patients with myocardial infarction. Heart. 2020;106(18):1420–6. https://pubmed.ncbi.nlm.nih.gov/32111640/
Giem P, Beeson WL, Fraser GE. The incidence of dementia and intake of animal products: preliminary findings from the Adventist Health Study. Neuroepidemiology. 1993;12(1):28–36. https://pubmed.ncbi.nlm.nih.gov/8327020/
Wahl D, Cogger VC, Solon-Biet SM, et al. Nutritional strategies to optimise cognitive function in the aging brain. Ageing Res Rev. 2016;31:80–92. https://pubmed.ncbi.nlm.nih.gov/27355990/
Olshansky SJ, Carnes BA, Cassel C. In search of Methuselah: estimating the upper limits to human longevity. Science. 1990;250(4981):634–40. https://pubmed.ncbi.nlm.nih.gov/2237414/
Vaiserman A, Koliada A, Lushchak O, Castillo MJ. Repurposing drugs to fight aging: the difficult path from bench to bedside. Med Res Rev. 2021;41(3):1676–700. https://pubmed.ncbi.nlm.nih.gov/33314257/
Olshansky SJ, Perry D, Miller RA, Butler RN. In pursuit of the longevity dividend. Scientist (Philadelphia, Pa). 2006;20(3):28–36. https://pubmed.ncbi.nlm.nih.gov/17986572/
Blagosklonny MV. Disease or not, aging is easily treatable. Aging (Albany NY). 2018;10(11):3067–78. https://pubmed.ncbi.nlm.nih.gov/30448823/
De Winter G. Aging as disease. Med Health Care Philos. 2015;18(2):237–43. https://pubmed.ncbi.nlm.nih.gov/25240472/
Zhavoronkov A, Bhullar B. Classifying aging as a disease in the context of ICD-11. Front Genet. 2015;6:326. https://pubmed.ncbi.nlm.nih.gov/26583032/
Hodgson J. Consumer, drug firms vie in vitamins. Wall Street Journal. https://www.wsj.com/articles/SB10001424127887323401904578155050445302398. Published December 2, 2012. Accessed January 24, 2023.; https://www.wsj.com/articles/SB10001424127887323401904578155050445302398
Davis B. The link between Big Pharma and the supplement industry. Elsevier: Pharma R&D Today. https://web.archive.org/web/20220930062808/ https:/pharma.elsevier.com/pharma-rd/link-big-pharma-supplement-industry/. Published July 28th, 2017. Accessed February 10, 2023.; https://web.archive.org/web/20220930062808/ https://pharma.elsevier.com/pharma-rd/link-big-pharma-supplement-industry/
Направление, сформированное на стыке косметологии и фармакологии. – Примеч. ред.
Martin KI, Glaser DA. Cosmeceuticals: the new medicine of beauty. Mo Med. 2011;108(1):60–3. https://pubmed.ncbi.nlm.nih.gov/21462614/
Exuviance. Johnson & Johnson. https://www.jnj.com/exuviance. Accessed January 22, 2023.; https://www.jnj.com/exuviance
Spencer M. Coca-Cola, Sanofi in beauty venture. Wall Street Journal. https://www.wsj.com/articles/SB10000872396390443854204578060662301872612. Published October 16, 2012. Accessed January 24, 2023.; https://www.wsj.com/articles/SB10000872396390443854204578060662301872612
Miller RA. Extending life: scientific prospects and political obstacles. Milbank Q. 2002;80(1):155–74. https://pubmed.ncbi.nlm.nih.gov/11933792/
Donner Y, Fortney K, Calimport SRG, Pfleger K, Shah M, Betts-LaCroix J. Great desire for extended life and health amongst the American public. Front Genet. 2016;6:353. https://pubmed.ncbi.nlm.nih.gov/26834780/
Eissenberg JC. Hungering for immortality. Mo Med. 2018;115(1):12–7. https://pubmed.ncbi.nlm.nih.gov/30228670/
Hall WJ. Centenarians: metaphor becomes reality. Arch Intern Med. 2008;168(3):262–3. https://pubmed.ncbi.nlm.nih.gov/18268165/
Faragher RGA. Should we treat aging as a disease? The consequences and dangers of miscategorisation. Front Genet. 2015;6:171. https://pubmed.ncbi.nlm.nih.gov/26236330/
Marengoni A, Angleman S, Melis R, et al. Aging with multimorbidity: a systematic review of the literature. Ageing Res Rev. 2011;10(4):430–9. https://pubmed.ncbi.nlm.nih.gov/21402176/
Barnett K, Mercer SW, Norbury M, Watt G, Wyke S, Guthrie B. Epidemiology of multimorbidity and implications for health care, research, and medical education: a cross-sectional study. Lancet. 2012;380(9836):37–43. https://pubmed.ncbi.nlm.nih.gov/22579043/
Smith-Uffen MES, Johnson SB, Martin AJ, et al. Estimating survival in advanced cancer: a comparison of estimates made by oncologists and patients. Support Care Cancer. 2020;28(7):3399–407. https://pubmed.ncbi.nlm.nih.gov/31781946/
Hole B, Salem J. How long do patients with chronic disease expect to live? A systematic review of the literature. BMJ Open. 2016;6(12):e012248. https://pubmed.ncbi.nlm.nih.gov/28039288/
Kaeberlein M. How healthy is the healthspan concept? GeroScience. 2018;40(4):361–4. https://pubmed.ncbi.nlm.nih.gov/30084059/
Около 400 метров. – Примеч. ред.
Crimmins EM, Beltrán-Sánchez H. Mortality and morbidity trends: is there compression of morbidity? J Gerontol B Psychol Sci Soc Sci. 2011 Jan;66(1):75–86. https://pubmed.ncbi.nlm.nih.gov/21135070/
de Magalhães JP. The scientific quest for lasting youth: prospects for curing aging. Rejuvenation Res. 2014;17(5):458–67. https://pubmed.ncbi.nlm.nih.gov/25132068/
Хуан Понсе де Леон (1460–1521) – испанский конкистадор, который основал первое европейское поселение на Пуэрто-Рико и во время поисков источника вечной молодости в 1513 году первым из европейцев высадился на берега Флориды. – Примеч. ред.
Furrer R, Handschin C. Lifestyle vs. pharmacological interventions for healthy aging. Aging (Albany NY). 2020;12(1):5–7. https://pubmed.ncbi.nlm.nih.gov/31937689/
Barja G. Updating the mitochondrial free radical theory of aging: an integrated view, key aspects, and confounding concepts. Antioxid Redox Signal. 2013;19(12):1420–45. https://pubmed.ncbi.nlm.nih.gov/23642158/
de Magalhães JP. The scientific quest for lasting youth: prospects for curing aging. Rejuvenation Res. 2014;17(5):458–67. https://pubmed.ncbi.nlm.nih.gov/25132068/
Kirkwood T. Why can’t we live forever? Sci Am. 2010;303(3):42–9. https://pubmed.ncbi.nlm.nih.gov/20812478/
Pakkenberg B, Pelvig D, Marner L, et al. Aging and the human neocortex. Exp Gerontol. 2003;38(1–2):95–9. https://pubmed.ncbi.nlm.nih.gov/12543266/
Herculano-Houzel S. The human brain in numbers: a linearly scaled-up primate brain. Front Hum Neurosci. 2009;3:31. https://pubmed.ncbi.nlm.nih.gov/19915731/
Pakkenberg B, Pelvig D, Marner L, et al. Aging and the human neocortex. Exp Gerontol. 2003;38(1–2):95–9. https://pubmed.ncbi.nlm.nih.gov/12543266/
Finlay BB, Pettersson S, Melby MK, Bosch TCG. The microbiome mediates environmental effects on aging. BioEssays. 2019;41(10):1800257. https://pubmed.ncbi.nlm.nih.gov/31157928/
Hayflick L. “Anti-aging” is an oxymoron. J Gerontol A Biol Sci Med Sci. 2004;59(6):B573–8. https://pubmed.ncbi.nlm.nih.gov/15215267/
Underwood M, Bartlett HP, Hall WD. Professional and personal attitudes of researchers in ageing towards life extension. Biogerontology. 2009;10(1):73–81. https://pubmed.ncbi.nlm.nih.gov/18516699/
de Grey ADNJ. Like it or not, life-extension research extends beyond biogerontology. EMBO Rep. 2005;6(11):1000. https://pubmed.ncbi.nlm.nih.gov/16264420/
Richmond CR. Population exposure from the fuel cycle: review and future direction. University of North Texas Libraries Government Documents Department. https://digital.library.unt.edu/ark:/67531/metadc1086292/. Published January 1, 1987. Accessed November 28, 2022.; https://digital.library.unt.edu/ark:/67531/metadc1086292/
de Grey ADNJ. Like it or not, life-extension research extends beyond biogerontology. EMBO Rep. 2005;6(11):1000. https://pubmed.ncbi.nlm.nih.gov/16264420/
Thomson W. Kelvin on science: British lord tells his hopes for wireless telegraphy. The Newark Advocate. https://zapatopi.net/kelvin/papers/interview_aeronautics_and_wireless.html. Published April 26, 1902. Accessed October 24, 2022.; https://zapatopi.net/kelvin/papers/interview_aeronautics_and_wireless.html
Ayyadevara S, Alla R, Thaden JJ, Shmookler Reis RJ. Remarkable longevity and stress resistance of nematode PI3K-null mutants. Aging Cell. 2008;7(1):13–22. https://pubmed.ncbi.nlm.nih.gov/17996009/
Bartke A, Wright JC, Mattison JA, Ingram DK, Miller RA, Roth GS. Extending the lifespan of long-lived mice. Nature. 2001;414(6862):412. https://pubmed.ncbi.nlm.nih.gov/11719795/
Richie JP, Leutzinger Y, Parthasarathy S, Malloy V, Orentreich N, Zimmerman JA. Methionine restriction increases blood glutathione and longevity in F344 rats. FASEB J. 1994;8(15):1302–7. https://pubmed.ncbi.nlm.nih.gov/8001743/
Miller RA. Extending life: scientific prospects and political obstacles. Milbank Q. 2002;80(1):155–74. https://pubmed.ncbi.nlm.nih.gov/11933792/
Campbell S. Will biotechnology stop aging? IEEE Pulse. 2019;10(2):3–7. https://pubmed.ncbi.nlm.nih.gov/31021750/
Faragher RGA. Should we treat aging as a disease? The consequences and dangers of miscategorisation. Front Genet. 2015;6:171. https://pubmed.ncbi.nlm.nih.gov/26236330/
de Grey ADNJ. Escape velocity: why the prospect of extreme human life extension matters now. PLoS Biol. 2004;2(6):e187. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC423155/
Kurzweil R, Grossman T. Fantastic voyage: live long enough to live forever. The science behind radical life extension questions and answers. Stud Health Technol Inform. 2009;149:187–94. https://pubmed.ncbi.nlm.nih.gov/19745481/
Raghavachari N. The impact of apolipoprotein E genetic variability in health and life span. J Gerontol A Biol Sci Med Sci. 2020;75(10):1855–7. https://pubmed.ncbi.nlm.nih.gov/32789475/
Medvedev ZA. An attempt at a rational classification of theories of ageing. Biol Rev Camb Philos Soc. 1990;65(3):375–98. https://pubmed.ncbi.nlm.nih.gov/2205304/
Willcox DC, Willcox BJ, Poon LW. Centenarian studies: important contributors to our understanding of the aging process and longevity. Curr Gerontol Geriatr Res. 2010;2010:484529. https://pubmed.ncbi.nlm.nih.gov/21804821/
Steves CJ, Spector TD, Jackson SHD. Ageing, genes, environment and epigenetics: what twin studies tell us now, and in the future. Age Ageing. 2012;41(5):581–6. https://pubmed.ncbi.nlm.nih.gov/22826292/
Kirkwood T. How can we live forever? BMJ. 1996;313(7072):1571. https://pubmed.ncbi.nlm.nih.gov/8990987/
Milman S, Barzilai N. Dissecting the mechanisms underlying unusually successful human health span and life span. Cold Spring Harb Perspect Med. 2015;6(1):a025098. https://pubmed.ncbi.nlm.nih.gov/26637439/
Ruby JG, Wright KM, Rand KA, et al. Estimates of the heritability of human longevity are substantially inflated due to assortative mating. Genetics. 2018;210(3):1109–24. https://pubmed.ncbi.nlm.nih.gov/30401766/
Herskind AM, McGue M, Holm NV, Sørensen TIA, Harvald B, Vaupel JW. The heritability of human longevity: a population-based study of 2872 Danish twin pairs born 1870–1900. Hum Genet. 1996;97(3):319–23. https://link.springer.com/article/10.1007/bf02185763
Skytthe A, Pedersen NL, Kaprio J, et al. Longevity studies in GenomEUtwin. Twin Res. 2003;6(5):448–54. https://pubmed.ncbi.nlm.nih.gov/14624729/
Ruby JG, Wright KM, Rand KA, et al. Estimates of the heritability of human longevity are substantially inflated due to assortative mating. Genetics. 2018;210(3):1109–24. https://pubmed.ncbi.nlm.nih.gov/30401766/
Lee MB, Hill CM, Bitto A, Kaeberlein M. Antiaging diets: separating fact from fiction. Science. 2021;374(6570):eabe7365. https://pubmed.ncbi.nlm.nih.gov/34793210/
Search results: “the hallmarks of aging.” WebofScience.com. Accessed February 15, 2023.; https://www.webofscience.com/wos/woscc/summary/55559f9d-7ef6-429d-98f8-f41bc4c102d7-84135d71/relevance/1
Levine M, Crimmins E. Not all smokers die young: a model for hidden heterogeneity within the human population. PLoS ONE. 2014;9(2):e87403. https://pubmed.ncbi.nlm.nih.gov/24520332/
Devi AS, Thokchom S, Devi AM. Children living with Progeria. Nurs Care Open Access J. 2017;3(4):275–8. https://medcraveonline.com/NCOAJ/children-living-with-progeria.html
Ahmed MS, Ikram S, Bibi N, Mir A. Hutchinson-Gilford progeria syndrome: a premature aging disease. Mol Neurobiol. 2018;55(5):4417–27. https://pubmed.ncbi.nlm.nih.gov/28660486/
Sosnowska D, Richardson C, Sonntag WE, Csiszar A, Ungvari Z, Ridgway I. A heart that beats for 500 years: age-related changes in cardiac proteasome activity, oxidative protein damage and expression of heat shock proteins, inflammatory factors, and mitochondrial complexes in Arctica islandica, the longest-living noncolonial animal. J Gerontol A Biol Sci Med Sci. 2014;69(12):1448–61. https://pubmed.ncbi.nlm.nih.gov/24347613/
Taormina G, Ferrante F, Vieni S, Grassi N, Russo A, Mirisola MG. Longevity: lesson from model organisms. Genes (Basel). 2019;10(7):518. https://pubmed.ncbi.nlm.nih.gov/31324014/
Концепция проведения научных исследований с привлечением широкого круга добровольцев-любителей (неспециалистов). – Примеч. ред.
Имя Мафусаила, прожившего 960 лет, стало синонимом долгожительства. «Собаками Мафусаила» традиционно называют собак-долгожителей. – Примеч. ред.
Jónás D, Sándor S, Tátrai K, Egyed B, Kubinyi E. A preliminary study to investigate the genetic background of longevity based on whole-genome sequence data of two Methuselah dogs. Front Genet. 2020;11:315. https://pubmed.ncbi.nlm.nih.gov/32373156/
Kaeberlein M, Creevy KE, Promislow DEL. The Dog Aging Project: translational geroscience in companion animals. Mamm Genome. 2016;27(7–8):279–88. https://pubmed.ncbi.nlm.nih.gov/27143112/
Pitt JN, Kaeberlein M. Why is aging conserved and what can we do about it? PLoS Biol. 2015;13(4):e1002131. https://pubmed.ncbi.nlm.nih.gov/25923592/
López M. Hypothalamic AMPK: a golden target against obesity? Eur J Endocrinol. 2017;176(5):R235–46. https://pubmed.ncbi.nlm.nih.gov/28232370/
Steinberg GR, Macaulay SL, Febbraio MA, Kemp BE. AMP-activated protein kinase – the fat controller of the energy railroad. Can J Physiol Pharmacol. 2006;84(7):655–65. https://pubmed.ncbi.nlm.nih.gov/16998529/
Salminen A, Kaarniranta K. AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res Rev. 2012;11(2):230–41. https://pubmed.ncbi.nlm.nih.gov/22186033/
Vazirian M, Nabavi SM, Jafari S, Manayi A. Natural activators of adenosine 5’-monophosphate (AMP)-activated protein kinase (AMPK) and their pharmacological activities. Food Chem Toxicol. 2018;122:69–79. https://pubmed.ncbi.nlm.nih.gov/30290216/
Jiang S, Li T, Yang Z, et al. AMPK orchestrates an elaborate cascade protecting tissue from fibrosis and aging. Ageing Res Rev. 2017;38:18–27. https://pubmed.ncbi.nlm.nih.gov/28709692/
Burkewitz K, Weir HJM, Mair WB. AMPK as a pro-longevity target. In: Cordero MD, Viollet B, eds. AMP-activated Protein Kinase. Experientia Supplementum. Vol 107. Springer; 2016:227–56. https://pubmed.ncbi.nlm.nih.gov/27812983/
Ruiz R, Pérez-Villegas EM, Manuel Carrión Á. AMPK function in aging process. Curr Drug Targets. 2016;17(8):932–41. https://pubmed.ncbi.nlm.nih.gov/26521771/
Salminen A, Kaarniranta K, Kauppinen A. Age-related changes in AMPK activation: role for AMPK phosphatases and inhibitory phosphorylation by upstream signaling pathways. Ageing Res Rev. 2016;28:15–26. https://pubmed.ncbi.nlm.nih.gov/27060201/
Wang S, Kandadi MR, Ren J. Double knockout of Akt2 and AMPK predisposes cardiac aging without affecting lifespan: role of autophagy and mitophagy. Biochim Biophys Acta Mol Basis Dis. 2019;1865(7):1865–75. https://pubmed.ncbi.nlm.nih.gov/31109453/
Ruiz R, Pérez-Villegas EM, Manuel Carrión Á. AMPK function in aging process. Curr Drug Targets. 2016;17(8):932–41. https://pubmed.ncbi.nlm.nih.gov/26521771/
Mair W, Morantte I, Rodrigues APC, et al. Lifespan extension induced by AMPK and calcineurin is mediated by CRTC-1 and CREB. Nature. 2011;470(7334):404–8. https://pubmed.ncbi.nlm.nih.gov/21331044/
Sokolov SS, Severin FF. Manipulating cellular energetics to slow aging of tissues and organs. Biochemistry (Mosc). 2020;85(6):651–9. https://pubmed.ncbi.nlm.nih.gov/32586228/
Burkewitz K, Weir HJM, Mair WB. AMPK as a pro-longevity target. In: Cordero MD, Viollet B, eds. AMP-activated Protein Kinase. Experientia Supplementum. Vol 107. Springer; 2016:227–56. https://pubmed.ncbi.nlm.nih.gov/27812983/
Миметики – это лекарственные вещества, биохимически имитирующие естественное синтезируемое в организме вещество или вызывающие в организме изменения, сходные с теми, которые проявляются под действием какого-либо внешнего фактора. – Примеч. ред.
Burkewitz K, Zhang Y, Mair WB. AMPK at the nexus of energetics and aging. Cell Metab. 2014;20(1):10–25. https://pubmed.ncbi.nlm.nih.gov/24726383/
Musi N, Fujii N, Hirshman MF, et al. AMP-activated protein kinase (AMPK) is activated in muscle of subjects with type 2 diabetes during exercise. Diabetes. 2001;50(5):921–7. https://pubmed.ncbi.nlm.nih.gov/11334434/
Kola B, Grossman AB, Korbonits M. The role of AMP-activated protein kinase in obesity. Front Horm Res. 2008;36:198–211. https://pubmed.ncbi.nlm.nih.gov/18230904/
Narkar VA, Downes M, Yu RT, et al. AMPK and PPARdelta agonists are exercise mimetics. Cell. 2008;134(3):405–15. https://pubmed.ncbi.nlm.nih.gov/18674809/
Benkimoun P. Police find range of drugs after trawling bins used by Tour de France cyclists. BMJ. 2009;339:b4201. https://pubmed.ncbi.nlm.nih.gov/19825964/
Niederberger E, King TS, Russe OQ, Geisslinger G. Activation of AMPK and its impact on exercise capacity. Sports Med. 2015;45(11):1497–509. https://pubmed.ncbi.nlm.nih.gov/26186961/
Niederberger E, King TS, Russe OQ, Geisslinger G. Activation of AMPK and its impact on exercise capacity. Sports Med. 2015;45(11):1497–509. https://pubmed.ncbi.nlm.nih.gov/26186961/
Hawley JA, Joyner MJ, Green DJ. Mimicking exercise: what matters most and where to next? J Physiol. 2021;599(3):791–802. https://pubmed.ncbi.nlm.nih.gov/31749163/
López-Lluch G, Santos-Ocaña C, Sánchez-Alcázar JA, et al. Mitochondrial responsibility in ageing process: innocent, suspect or guilty. Biogerontology. 2015;16(5):599–620. https://pubmed.ncbi.nlm.nih.gov/26105157/
Sharma A, Smith HJ, Yao P, Mair WB. Causal roles of mitochondrial dynamics in longevity and healthy aging. EMBO Rep. 2019;20(12):e48395. https://pubmed.ncbi.nlm.nih.gov/31667999/
Hill S, Van Remmen H. Mitochondrial stress signaling in longevity: a new role for mitochondrial function in aging. Redox Biol. 2014;2:936–44. https://pubmed.ncbi.nlm.nih.gov/25180170/
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–217. https://pubmed.ncbi.nlm.nih.gov/23746838/
Gonzalez-Freire M, de Cabo R, Bernier M, et al. Reconsidering the role of mitochondria in aging. J Gerontol A Biol Sci Med Sci. 2015;70(11):1334–42. https://pubmed.ncbi.nlm.nih.gov/25995290/
Sgarbi G, Matarrese P, Pinti M, et al. Mitochondria hyperfusion and elevated autophagic activity are key mechanisms for cellular bioenergetic preservation in centenarians. Aging (Albany NY). 2014;6(4):296–310. https://pubmed.ncbi.nlm.nih.gov/24799450/
Sengupta P. The laboratory rat: relating its age with human’s. Int J Prev Med. 2013;4(6):624–30. https://pubmed.ncbi.nlm.nih.gov/23930179/
Corbisier P, Remacle J. Influence of the energetic pattern of mitochondria in cell ageing. Mech Ageing Dev. 1993;71(1):47–58. https://pubmed.ncbi.nlm.nih.gov/8309283/
Burkewitz K, Zhang Y, Mair WB. AMPK at the nexus of energetics and aging. Cell Metab. 2014;20(1):10–25. https://pubmed.ncbi.nlm.nih.gov/24726383/
Ruiz R, Pérez-Villegas EM, Manuel Carrión Á. AMPK function in aging process. Curr Drug Targets. 2016;17(8):932–41. https://pubmed.ncbi.nlm.nih.gov/26521771/
Wu S, Zou MH. AMPK, mitochondrial function, and cardiovascular disease. Int J Mol Sci. 2020;21(14):4987. https://pubmed.ncbi.nlm.nih.gov/32679729/
Agency for Healthcare Research and Quality (AHRQ). Medical Expenditure Panel Survey (MEPS) 2013–2019. ClinCalc DrugStats Database version 2021.10. https://clincalc.com/DrugStats/. Accessed May 22, 2023.; https://clincalc.com/DrugStats/
Inzucchi SE, Fonseca V. Dethroning the king?: the future of metformin as first line therapy in type 2 diabetes. J Diabetes Complications. 2019;33(6):462–4. https://pubmed.ncbi.nlm.nih.gov/31003925/
Campbell JM, Bellman SM, Stephenson MD, Lisy K. Metformin reduces all-cause mortality and diseases of ageing independent of its effect on diabetes control: a systematic review and meta-analysis. Ageing Res Rev. 2017;40:31–44. https://pubmed.ncbi.nlm.nih.gov/28802803/
Glucophage® / Glucophage® XR: Response to FDA Comments of 10 12 00. U.S. Food & Drug Administration: Drugs@FDA. https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&ApplNo=021202. Accessed April 25, 2021.; https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&ApplNo=021202
Braun B, Eze P, Stephens BR, et al. Impact of metformin on peak aerobic capacity. Appl Physiol Nutr Metab. 2008;33(1):61–7. https://pubmed.ncbi.nlm.nih.gov/18347654/
Walton RG, Dungan CM, Long DE, et al. Metformin blunts muscle hypertrophy in response to progressive resistance exercise training in older adults: a randomized, double-blind, placebo-controlled, multicenter trial: the MASTERS trial [published correction appears in Aging Cell. 2020;19(3):e13098]. Aging Cell. 2019;18(6):e13039. https://pubmed.ncbi.nlm.nih.gov/31557380/
Burkewitz K, Zhang Y, Mair WB. AMPK at the nexus of energetics and aging. Cell Metab. 2014;20(1):10–25. https://pubmed.ncbi.nlm.nih.gov/24726383/
Knowler WC, Barrett-Connor E, Fowler SE, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346(6):393–403. https://pubmed.ncbi.nlm.nih.gov/11832527/
Iannello S, Camuto M, Cavaleri A, et al. Effects of short-term metformin treatment on insulin sensitivity of blood glucose and free fatty acids. Diabetes Obes Metab. 2004;6(1):8–15. https://pubmed.ncbi.nlm.nih.gov/14686957/
Wen H, Gris D, Lei Y, et al. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat Immunol. 2011;12(5):408–15. https://pubmed.ncbi.nlm.nih.gov/21478880/
Carta G, Murru E, Banni S, Manca C. Palmitic acid: physiological role, metabolism and nutritional implications. Front Physiol. 2017;8:902. https://pubmed.ncbi.nlm.nih.gov/29167646/
Fatima S, Hu X, Gong RH, et al. Palmitic acid is an intracellular signaling molecule involved in disease development. Cell Mol Life Sci. 2019;76(13):2547–57. https://pubmed.ncbi.nlm.nih.gov/30968170/
Kirwan AM, Lenighan YM, O’Reilly ME, McGillicuddy FC, Roche HM. Nutritional modulation of metabolic inflammation. Biochem Soc Trans. 2017;45(4):979–85. https://pubmed.ncbi.nlm.nih.gov/28710289/
Arguello G, Balboa E, Arrese M, Zanlungo S. Recent insights on the role of cholesterol in non-alcoholic fatty liver disease. Biochim Biophys Acta. 2015;1852(9):1765–78. https://pubmed.ncbi.nlm.nih.gov/26027904/
Wang XJ, Malhi H. Nonalcoholic fatty liver disease. Ann Intern Med. 2018;169(9):ITC65–80. https://pubmed.ncbi.nlm.nih.gov/30398639/
Hydes T, Alam U, Cuthbertson DJ. The impact of macronutrient intake on non-alcoholic fatty liver disease (NAFLD): too much fat, too much carbohydrate, or just too many calories? Front Nutr. 2021;8:640557. https://pubmed.ncbi.nlm.nih.gov/33665203/
Luukkonen PK, Sädevirta S, Zhou Y, et al. Saturated fat is more metabolically harmful for the human liver than unsaturated fat or simple sugars. Diabetes Care. 2018;41(8):1732–9. https://pubmed.ncbi.nlm.nih.gov/29844096/
Luukkonen PK, Sädevirta S, Zhou Y, et al. Saturated fat is more metabolically harmful for the human liver than unsaturated fat or simple sugars. Diabetes Care. 2018;41(8):1732–9. https://pubmed.ncbi.nlm.nih.gov/29844096/
Kirwan AM, Lenighan YM, O’Reilly ME, McGillicuddy FC, Roche HM. Nutritional modulation of metabolic inflammation. Biochem Soc Trans. 2017;45(4):979–85. https://pubmed.ncbi.nlm.nih.gov/28710289/
Parry SA, Rosqvist F, Mozes FE, et al. Intrahepatic fat and postprandial glycemia increase after consumption of a diet enriched in saturated fat compared with free sugars. Diabetes Care. 2020;43(5):1134–41. https://pubmed.ncbi.nlm.nih.gov/32165444/
Grahame Hardie D. Regulation of AMP-activated protein kinase by natural and synthetic activators. Acta Pharm Sin B. 2016;6(1):1–19. https://pubmed.ncbi.nlm.nih.gov/26904394/
Wu Y, Song P, Zhang W, et al. Activation of AMPKa2 in adipocytes is essential for nicotine-induced insulin resistance in vivo. Nat Med. 2015;21(4):373–82. https://pubmed.ncbi.nlm.nih.gov/25799226/
Martínez de Morentin PB, Whittle AJ, Fernø J, et al. Nicotine induces negative energy balance through hypothalamic AMP-activated protein kinase. Diabetes. 2012;61(4):807–17. https://pubmed.ncbi.nlm.nih.gov/22315316/
Ferguson SG, Shiffman S, Rohay JM, Gitchell JG, Garvey AJ. Effect of compliance with nicotine gum dosing on weight gained during a quit attempt. Addiction. 2011;106(3):651–6. https://pubmed.ncbi.nlm.nih.gov/21182551/
Novak CM, Gavini CK. Smokeless weight loss. Diabetes. 2012;61(4):776–7. https://pubmed.ncbi.nlm.nih.gov/22442297/
Hadi A, Arab A, Ghaedi E, Rafie N, Miraghajani M, Kafeshani M. Barberry (Berberis vulgaris L.) is a safe approach for management of lipid parameters: a systematic review and meta-analysis of randomized controlled trials. Complement Ther Med. 2019;43:117–24. https://pubmed.ncbi.nlm.nih.gov/30935518/
Fouladi RF. Aqueous extract of dried fruit of Berberis vulgaris L. in acne vulgaris, a clinical trial. J Diet Suppl. 2012;9(4):253–61. https://pubmed.ncbi.nlm.nih.gov/23038982/
Emamat H, Asadian S, Zahedmehr A, Ghanavati M, Nasrollahzadeh J. The effect of barberry (Berberis vulgaris) consumption on flow-mediated dilation and inflammatory biomarkers in patients with hypertension: a randomized controlled trial [published online ahead of print, 2020 Dec 22]. Phytother Res. 2020;10.1002/ptr.7000. https://pubmed.ncbi.nlm.nih.gov/33350540/
Shidfar F, Ebrahimi SS, Hosseini S, Heydari I, Shidfar S, Hajhassani G. The effects of Berberis vulgaris fruit extract on serum lipoproteins, apoB, apoA-I, homocysteine, glycemic control and total antioxidant capacity in type 2 diabetic patients. Iran J Pharm Res. 2012;11(2):643–52. https://pubmed.ncbi.nlm.nih.gov/24250489/
McCarty MF. AMPK activation – protean potential for boosting healthspan. Age (Dordr). 2014;36(2):641–63. https://pubmed.ncbi.nlm.nih.gov/24248330/
Shidfar F, Ebrahimi SS, Hosseini S, Heydari I, Shidfar S, Hajhassani G. The effects of Berberis vulgaris fruit extract on serum lipoproteins, apoB, apoA-I, homocysteine, glycemic control and total antioxidant capacity in type 2 diabetic patients. Iran J Pharm Res. 2012;11(2):643–52. https://pubmed.ncbi.nlm.nih.gov/24250489/
Funk RS, Singh RK, Winefield RD, et al. Variability in potency among commercial preparations of berberine. J Diet Suppl. 2018;15(3):343–51. https://pubmed.ncbi.nlm.nih.gov/28792254/
Arayne MS, Sultana N, Bahadur SS. The berberis story: Berberis vulgaris in therapeutics. Pak J Pharm Sci. 2007;20(1):83–92. https://pubmed.ncbi.nlm.nih.gov/17337435/
Grahame Hardie D. Regulation of AMP-activated protein kinase by natural and synthetic activators. Acta Pharm Sin B. 2016;6(1):1–19. https://pubmed.ncbi.nlm.nih.gov/26904394/
Tavakoli-Rouzbehani OM, Maleki V, Shadnoush M, Taheri E, Alizadeh M. A comprehensive insight into potential roles of Nigella sativa on diseases by targeting AMP-activated protein kinase: a review. Daru. 2020;28(2):779–87. https://pubmed.ncbi.nlm.nih.gov/33140312/
Mousavi SM, Sheikhi A, Varkaneh HK, Zarezadeh M, Rahmani J, Milajerdi A. Effect of Nigella sativa supplementation on obesity indices: a systematic review and meta-analysis of randomized controlled trials. Complement Ther Med. 2018;38:48–57. https://pubmed.ncbi.nlm.nih.gov/29857879/
Sahebkar A, Beccuti G, Simental-Mendía LE, Nobili V, Bo S. Nigella sativa (black seed) effects on plasma lipid concentrations in humans: a systematic review and meta-analysis of randomized placebo-controlled trials. Pharmacol Res. 2016;106:37–50. https://pubmed.ncbi.nlm.nih.gov/26875640/
Sahebkar A, Soranna D, Liu X, et al. A systematic review and meta-analysis of randomized controlled trials investigating the effects of supplementation with Nigella sativa (black seed) on blood pressure. J Hypertens. 2016;34(11):2127–35. https://pubmed.ncbi.nlm.nih.gov/27512971/
Daryabeygi-Khotbehsara R, Golzarand M, Ghaffari MP, Djafarian K. Nigella sativa improves glucose homeostasis and serum lipids in type 2 diabetes: a systematic review and meta-analysis. Complement Ther Med. 2017;35:6–13. https://pubmed.ncbi.nlm.nih.gov/29154069/
Agricultural Research Service, United States Department of Agriculture. Sweet sunnah, whole black seeds nigella sativa. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/food-details/468991/nutrients. Published April 1, 2019. Accessed May 8, 2021.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/468991/nutrients
Montazeri RS, Fatahi S, Sohouli MH, et al. The effect of nigella sativa on biomarkers of inflammation and oxidative stress: a systematic review and meta-analysis of randomized controlled trials. J Food Biochem. 2021;45(4):e13625. https://pubmed.ncbi.nlm.nih.gov/33559935/
He T, Xu X. The influence of Nigella sativa for asthma control: a meta-analysis. Am J Emerg Med. 2020;38(3):589–93. https://pubmed.ncbi.nlm.nih.gov/31892440/
Khabbazi A, Javadivala Z, Seyedsadjadi N, Malek Mahdavi A. A systematic review of the potential effects of Nigella sativa on rheumatoid arthritis. Planta Med. 2020;86(7):457–69. https://pubmed.ncbi.nlm.nih.gov/32274788/
Tajmiri S, Abbasalizad Farhangi M, Dehghan P. Nigella Sativa treatment and serum concentrations of thyroid hormones, transforming growth factor ß (TGF-ß) and interleukin 23 (IL-23) in patients with Hashimoto’s thyroiditis. Eur J Integr Med. 2016;8(4):576–80. https://www.sciencedirect.com/science/article/abs/pii/S1876382016300208
Ardakani Movaghati MR, Yousefi M, Saghebi SA, Sadeghi Vazin M, Iraji A, Mosavat SH. Efficacy of black seed (Nigella sativa L.) on kidney stone dissolution: a randomized, double-blind, placebo-controlled, clinical trial. Phytother Res. 2019;33(5):1404–12. https://pubmed.ncbi.nlm.nih.gov/30873671/
Latiff LA, Parhizkar S, Dollah MA, Hassan ST. Alternative supplement for enhancement of reproductive health and metabolic profile among perimenopausal women: a novel role of Nigella sativa. Iran J Basic Med Sci. 2014;17(12):980–5. https://pubmed.ncbi.nlm.nih.gov/25859301/
Lingesh A, Paul D, Naidu V, Satheeshkumar N. AMPK activating and anti adipogenic potential of Hibiscus rosa sinensis flower in 3T3-L1 cells. J Ethnopharmacol. 2019;233:123–30. https://pubmed.ncbi.nlm.nih.gov/30593890/
Amos A, Khiatah B. Mechanisms of action of nutritionally rich Hibiscus sabdariffa’s therapeutic uses in major common chronic diseases: a literature review [published online ahead of print, 2021 Jan 28]. J Am Coll Nutr. 2021;1–8. https://pubmed.ncbi.nlm.nih.gov/33507846/
Soleimani AR, Akbari H, Soleimani S, Beladi Mousavi SS, Tamadon MR. Effect of sour tea (Lipicom) pill versus captopril on the treatment of hypertension. J Renal Inj Prev. 2015;4(3):73–9. https://pubmed.ncbi.nlm.nih.gov/26468478/
Nwachukwu DC, Aneke EI, Nwachukwu NZ, Azubike N, Obika LF. Does consumption of an aqueous extract of Hibscus sabdariffa affect renal function in subjects with mild to moderate hypertension? J Physiol Sci. 2017;67(1):227–34. https://pubmed.ncbi.nlm.nih.gov/27221151/
Hopkins AL, Lamm MG, Funk JL, Ritenbaugh C. Hibiscus sabdariffa L. in the treatment of hypertension and hyperlipidemia: a comprehensive review of animal and human studies. Fitoterapia. 2013;85:84–94. https://pubmed.ncbi.nlm.nih.gov/23333908/
Bule M, Albelbeisi AH, Nikfar S, Amini M, Abdollahi M. The antidiabetic and antilipidemic effects of Hibiscus sabdariffa: a systematic review and meta-analysis of randomized clinical trials. Food Res Int (Ottawa). 2020;130:108980. https://pubmed.ncbi.nlm.nih.gov/32156406/
Abubakar SM, Ukeyima MT, Spencer JPE, Lovegrove JA. Acute effects of Hibiscus sabdariffa calyces on postprandial blood pressure, vascular function, blood lipids, biomarkers of insulin resistance and inflammation in humans. Nutrients. 2019;11(2):341. https://pubmed.ncbi.nlm.nih.gov/30764582/
Chang HC, Peng CH, Yeh DM, Kao ES, Wang CJ. Hibiscus sabdariffa extract inhibits obesity and fat accumulation, and improves liver steatosis in humans. Food Funct. 2014;5(4):734–9. https://pubmed.ncbi.nlm.nih.gov/24549255/
Wu CH, Huang CC, Hung CH, Yao FY, Wang CJ, Chang YC. Delphinidin-rich extracts of Hibiscus sabdariffa L. trigger mitochondria-derived autophagy and necrosis through reactive oxygen species in human breast cancer cells. J Funct Foods. 2016;25:279–90. https://www.sciencedirect.com/science/article/abs/pii/S175646461630144X?via%3Dihub
Salim LZA, Mohan S, Othman R, et al. Thymoquinone induces mitochondria-mediated apoptosis in acute lymphoblastic leukaemia in vitro. Molecules. 2013;18(9):11219–40. https://pubmed.ncbi.nlm.nih.gov/24036512/
Chen H, Chen T, Giudici P, Chen F. Vinegar functions on health: constituents, sources, and formation mechanisms. Compr Rev Food Sci Food Saf. 2016;15(6):1124–38. https://pubmed.ncbi.nlm.nih.gov/33401833/
Ali Z, Wang Z, Amir RM, et al. Potential uses of vinegar as a medicine and related in vivo mechanisms. Int J Vitam Nutr Res. 2018;86(3–4):1–12. https://pubmed.ncbi.nlm.nih.gov/29580192/
Bagnardi V, Rota M, Botteri E, et al. Alcohol consumption and site-specific cancer risk: a comprehensive dose-response meta-analysis. Br J Cancer. 2015;112(3):580–93. https://pubmed.ncbi.nlm.nih.gov/25422909/
Shield KD, Soerjomataram I, Rehm J. Alcohol use and breast cancer: a critical review. Alcohol Clin Exp Res. 2016;40(6):1166–81. https://pubmed.ncbi.nlm.nih.gov/27130687/
Ceddia RB. The role of AMP-activated protein kinase in regulating white adipose tissue metabolism. Mol Cell Endocrinol. 2013;366(2):194–203. https://pubmed.ncbi.nlm.nih.gov/22750051/
Center for Food Safety and Applied Nutrition, Office of Regulatory Affairs. CPG sec. 525.825 vinegar, definitions – adulteration with vinegar eels. United States Food and Drug Administration. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/cpg-sec-525825-vinegar-definitions-adulteration-vinegar-eels. Published March 1995. Accessed May 8, 2021.; https://www.fda.gov/regulatory-information/search-fda-guidance-documents/cpg-sec-525825-vinegar-definitions-adulteration-vinegar-eels
Park J, Kim J, Kim J, et al. Pomegranate vinegar beverage reduces visceral fat accumulation in association with AMPK activation in overweight women: a double-blind, randomized, and placebo-controlled trial. J Funct Foods. 2014;8:274–81. https://www.sciencedirect.com/science/article/abs/pii/S1756464614001273
Kondo T, Kishi M, Fushimi T, Ugajin S, Kaga T. Vinegar intake reduces body weight, body fat mass, and serum triglyceride levels in obese Japanese subjects. Biosci Biotechnol Biochem. 2009;73(8):1837–43. https://pubmed.ncbi.nlm.nih.gov/19661687/
Johnston C, Quagliano S, White S. Vinegar ingestion at mealtime reduced fasting blood glucose concentrations in healthy adults at risk for type 2 diabetes. J Funct Foods. 2013;5(4):2007–11. https://www.sciencedirect.com/science/article/abs/pii/S1756464613001874
Mitrou P, Petsiou E, Papakonstantinou E, et al. Vinegar consumption increases insulin-stimulated glucose uptake by the forearm muscle in humans with type 2 diabetes. J Diabetes Res. 2015;2015:175204. https://pubmed.ncbi.nlm.nih.gov/26064976/
Hu GX, Chen GR, Xu H, Ge RS, Lin J. Activation of the AMP activated protein kinase by short-chain fatty acids is the main mechanism underlying the beneficial effect of a high fiber diet on the metabolic syndrome. Med Hypotheses. 2010;74(1):123–6. https://pubmed.ncbi.nlm.nih.gov/19665312/
Abid M, Memon Z, Shaheen S, Ahmed F, Shaikh MZ, Agha F. Comparison of apple cider vinegar and metformin combination with metformin alone in newly diagnosed type 2 diabetic patients: a randomized controlled trial. Int J Med Res Health Sci. 2020;9(2):1–7. https://www.ijmrhs.com/abstract/comparison-of-apple-cider-vinegar-and-metformin-combination-with-metformin-alone-in-newly-diagnosed-type-2-diabetic-pati-44684.html
Sakakibara S, Murakami R, Takahashi M, et al. Vinegar intake enhances flow-mediated vasodilatation via upregulation of endothelial nitric oxide synthase activity. Biosci Biotechnol Biochem. 2010;74(5):1055–61. https://pubmed.ncbi.nlm.nih.gov/20460711/
Beheshti Z, Chan YH, Nia HS, et al. Influence of apple cider vinegar on blood lipids. Life Sci J. 2012;9(4):2431–40. https://www.lifesciencesite.com/lsj/life0904/360_10755life0904_2431_2440.pdf
Chuang MH, Chiou SH, Huang CH, Yang WB, Wong CH. The lifespan-promoting effect of acetic acid and Reishi polysaccharide. Bioorg Med Chem. 2009;17(22):7831–40. https://pubmed.ncbi.nlm.nih.gov/19837596/
Hu FB, Stampfer MJ, Manson JE, et al. Dietary intake of alpha-linolenic acid and risk of fatal ischemic heart disease among women. Am J Clin Nutr. 1999;69(5):890–7. https://pubmed.ncbi.nlm.nih.gov/10232627/
Hu GX, Chen GR, Xu H, Ge RS, Lin J. Activation of the AMP activated protein kinase by short-chain fatty acids is the main mechanism underlying the beneficial effect of a high fiber diet on the metabolic syndrome. Med Hypotheses. 2010;74(1):123–6. https://pubmed.ncbi.nlm.nih.gov/19665312/
Koç F, Mills S, Strain C, Ross RP, Stanton C. The public health rationale for increasing dietary fibre: health benefits with a focus on gut microbiota. Nutr Bull. 2020;45:294–308. https://onlinelibrary.wiley.com/doi/10.1111/nbu.12448
Pritchard SE, Marciani L, Garsed KC, et al. Fasting and postprandial volumes of the undisturbed colon: normal values and changes in diarrhea-predominant irritable bowel syndrome measured using serial MRI. Neurogastroenterol Motil. 2014;26(1):124–30. https://pubmed.ncbi.nlm.nih.gov/24131490/
Tang R, Li L. Modulation of short-chain fatty acids as potential therapy method for type 2 diabetes mellitus. Can J Infect Dis Med Microbiol. 2021;2021:6632266. https://pubmed.ncbi.nlm.nih.gov/33488888/
Hu GX, Chen GR, Xu H, Ge RS, Lin J. Activation of the AMP activated protein kinase by short-chain fatty acids is the main mechanism underlying the beneficial effect of a high fiber diet on the metabolic syndrome. Med Hypotheses. 2010;74(1):123–6. https://pubmed.ncbi.nlm.nih.gov/19665312/
Spiller G, ed. Topics in Dietary Fiber Research. Plenum Press; 1978. https://link.springer.com/book/10.1007/978-1-4684-2481-2
Eaton SB, Eaton SB, Konner MJ. Paleolithic nutrition revisited: a twelve-year retrospective on its nature and implications. Eur J Clin Nutr. 1997;51(4):207–16. https://pubmed.ncbi.nlm.nih.gov/9104571/
Usual nutrient intake from food and beverages, by gender and age: what we eat in America, NHANES 2015–2018. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/usual/Usual_Intake_gender_WWEIA_2015_2018.pdf. Published January 2021. Accessed December 25, 2022.; https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/usual/Usual_Intake_gender_WWEIA_2015_2018.pdf
McRorie JW. Evidence-based approach to fiber supplements and clinically meaningful health benefits, part 1: what to look for and how to recommend an effective fiber therapy. Nutr Today. 2015;50(2):82–9. https://pubmed.ncbi.nlm.nih.gov/25972618/
López M. Hypothalamic AMPK: a golden target against obesity? Eur J Endocrinol. 2017;176(5):R235–46. https://pubmed.ncbi.nlm.nih.gov/28232370/
Morgunova GV, Klebanov AA. Age-related AMP-activated protein kinase alterations: from cellular energetics to longevity. Cell Biochem Funct. 2019;37(3):169–76. https://pubmed.ncbi.nlm.nih.gov/30895648/
Американская единица объема «чашка» (cup) равна 240 мл. – Примеч. ред.
There are many different types of autophagy, including chaperone-mediated autophagy and microautophagy. In this book, I’m referring to macroautophagy.
Tschachler E, Eckhart L. Autophagy: how to control your intracellular diet. Br J Dermatol. 2017;176(6):1417–9. https://pubmed.ncbi.nlm.nih.gov/28581245/
Levine B, Klionsky DJ. Autophagy wins the 2016 Nobel Prize in Physiology or Medicine: breakthroughs in baker’s yeast fuel advances in biomedical research. PNAS. 2017;114(2):201–5. https://pubmed.ncbi.nlm.nih.gov/28039434/
Vijayakumar K, Cho G. Autophagy: an evolutionarily conserved process in the maintenance of stem cells and aging. Cell Biochem Funct. 2019;37(6):452–8. https://pubmed.ncbi.nlm.nih.gov/31318072/
Kouda K, Iki M. Beneficial effects of mild stress (hormetic effects): dietary restriction and health. J Physiol Anthropol. 2010;29(4):127–32. https://pubmed.ncbi.nlm.nih.gov/20686325/
Tschachler E, Eckhart L. Autophagy: how to control your intracellular diet. Br J Dermatol. 2017;176(6):1417–9. https://pubmed.ncbi.nlm.nih.gov/28581245/
Cuervo AM. Calorie restriction and aging: the ultimate “cleansing diet.” J Gerontol A Biol Sci Med Sci. 2008;63(6):547–9. https://academic.oup.com/biomedgerontology/article/63/6/547/573952
Madeo F, Zimmermann A, Maiuri MC, Kroemer G. Essential role for autophagy in life span extension. J Clin Invest. 2015;125(1):85–93. https://pubmed.ncbi.nlm.nih.gov/25654554/
Pyo JO, Yoo SM, Ahn HH, et al. Overexpression of Atg5 in mice activates autophagy and extends lifespan. Nat Commun. 2013;4:2300. https://pubmed.ncbi.nlm.nih.gov/23939249/
Wong SQ, Kumar AV, Mills J, Lapierre LR. Autophagy in aging and longevity. Hum Genet. 2020;139(3):277–90. https://pubmed.ncbi.nlm.nih.gov/31144030/
Cuervo AM. Calorie restriction and aging: the ultimate “cleansing diet.” J Gerontol A Biol Sci Med Sci. 2008;63(6):547–9. https://academic.oup.com/biomedgerontology/article/63/6/547/573952
Meijer AJ. Autophagy in practice: stevia and leucine. Autophagy. 2019;15(12):2043. https://pubmed.ncbi.nlm.nih.gov/31455125/
Meijer AJ, Lorin S, Blommaart EF, Codogno P. Regulation of autophagy by amino acids and MTOR-dependent signal transduction. Amino Acids. 2015;47(10):2037–63. https://pubmed.ncbi.nlm.nih.gov/24880909/
Показатель физической работоспособности, определяет максимальное количество кислорода, которое может потреблять организм во время интенсивных упражнений. – Примеч. ред.
Escobar KA, Cole NH, Mermier CM, VanDusseldorp TA. Autophagy and aging: maintaining the proteome through exercise and caloric restriction. Aging Cell. 2019;18(1):e12876. https://pubmed.ncbi.nlm.nih.gov/30430746/
Brandt N, Gunnarsson TP, Bangsbo J, Pilegaard H. Exercise and exercise training – induced increase in autophagy markers in human skeletal muscle. Physiol Rep. 2018;6(7):e13651. https://pubmed.ncbi.nlm.nih.gov/29626392/
Escobar KA, Cole NH, Mermier CM, VanDusseldorp TA. Autophagy and aging: maintaining the proteome through exercise and caloric restriction. Aging Cell. 2019;18(1):e12876. https://pubmed.ncbi.nlm.nih.gov/30430746/
Cuervo AM. Calorie restriction and aging: the ultimate “cleansing diet.” J Gerontol A Biol Sci Med Sci. 2008;63(6):547–9. https://academic.oup.com/biomedgerontology/article/63/6/547/573952
Melnik BC. Leucine signaling in the pathogenesis of type 2 diabetes and obesity. World J Diabetes. 2012;3(3):38. https://pubmed.ncbi.nlm.nih.gov/22442749/
Rittig N, Bach E, Thomsen HH, et al. Anabolic effects of leucine-rich whey protein, carbohydrate, and soy protein with and without ß-hydroxy-ß-methylbutyrate (Hmb) during fasting-induced catabolism: a human randomized crossover trial. Clin Nutr. 2017;36(3):697–705. https://pubmed.ncbi.nlm.nih.gov/27265181/
Tareke E, Rydberg P, Karlsson P, Eriksson S, Törnqvist M. Analysis of acrylamide, a carcinogen formed in heated foodstuffs. J Agric Food Chem. 2002;50:4998–5006. https://pubmed.ncbi.nlm.nih.gov/12166997/
Song D, Xu C, Holck AL, Liu R. Acrylamide inhibits autophagy, induces apoptosis and alters cellular metabolic profiles. Ecotoxicol Environ Saf. 2021;208:111543. https://pubmed.ncbi.nlm.nih.gov/33396091/
Huang M, Jiao J, Wang J, Chen X, Zhang Y. Associations of hemoglobin biomarker levels of acrylamide and all-cause and cardiovascular disease mortality among U.S. adults: National Health and Nutrition Examination Survey 2003–2006. Environ Pollut. 2018;238:852–8. https://pubmed.ncbi.nlm.nih.gov/29627755/
Naruszewicz M, Zapolska-Downar D, Kosmider A, et al. Chronic intake of potato chips in humans increases the production of reactive oxygen radicals by leukocytes and increases plasma C-reactive protein: a pilot study. Am J Clin Nutr. 2009;89(3):773–7. https://pubmed.ncbi.nlm.nih.gov/19158207/
Chase P, Mitchell K, Morley JE. In the steps of giants: the early geriatrics texts. J Am Geriatr Soc. 2000;48(1):89–94. https://pubmed.ncbi.nlm.nih.gov/10642028/
Madeo F, Zimmermann A, Maiuri MC, Kroemer G. Essential role for autophagy in life span extension. J Clin Invest. 2015;125(1):85–93. https://pubmed.ncbi.nlm.nih.gov/25654554/
Arnesen E, Huseby NE, Brenn T, Try K. The Tromsø Heart Study: distribution of, and determinants for, gamma-glutamyltransferase in a free-living population. Scand J Clin Lab Invest. 1986;46(1):63–70. https://pubmed.ncbi.nlm.nih.gov/2869572/
Ruhl CE, Everhart JE. Coffee and tea consumption are associated with a lower incidence of chronic liver disease in the United States. Gastroenterology. 2005;129(6):1928–36. https://pubmed.ncbi.nlm.nih.gov/16344061/
Hayat U, Siddiqui AA, Okut H, Afroz S, Tasleem S, Haris A. The effect of coffee consumption on the non-alcoholic fatty liver disease and liver fibrosis: a meta-analysis of 11 epidemiological studies. Ann Hepatol. 2021;20:100254. https://pubmed.ncbi.nlm.nih.gov/32920163/
Ray K. Caffeine is a potent stimulator of autophagy to reduce hepatic lipid content – a coffee for NAFLD? Nat Rev Gastroenterol Hepatol 2013;10:563. https://pubmed.ncbi.nlm.nih.gov/23982685/
Sinha RA, Farah BL, Singh BK, et al. Caffeine stimulates hepatic lipid metabolism by the autophagy-lysosomal pathway in mice. Hepatology. 2014;59(4):1366–80. https://pubmed.ncbi.nlm.nih.gov/23929677/
Czachor J, Milek M, Galiniak S, Stepien K, Dzugan M, Molon M. Coffee extends yeast chronological lifespan through antioxidant properties. Int J Mol Sci. 2020;21(24):9510. https://pubmed.ncbi.nlm.nih.gov/33327536/
Sutphin GL, Bishop E, Yanos ME, Moller RM, Kaeberlein M. Caffeine extends life span, improves healthspan, and delays age-associated pathology in Caenorhabditis elegans. Longev Healthspan. 2012;1(1):9. https://pubmed.ncbi.nlm.nih.gov/24764514/
Pietrocola F, Malik SA, Mariño G, et al. Coffee induces autophagy in vivo. Cell Cycle. 2014;13(12):1987–94. https://pubmed.ncbi.nlm.nih.gov/24769862/
Takahashi K, Yanai S, Shimokado K, Ishigami A. Coffee consumption in aged mice increases energy production and decreases hepatic mTOR levels. Nutrition. 2017;38:1–8. https://pubmed.ncbi.nlm.nih.gov/28526373/
Известный слоган кофейного бренда Maxwell House. – Примеч. ред.
Saab S, Mallam D, Cox GA, Tong MJ. Impact of coffee on liver diseases: a systematic review. Liver Int. 2014;34(4):495–504. https://pubmed.ncbi.nlm.nih.gov/24102757/
Kanbay M, Siriopol D, Copur S, et al. Effect of coffee consumption on renal outcome: a systematic review and meta-analysis of clinical studies. J Ren Nutr. 2021;31(1):5–20. https://pubmed.ncbi.nlm.nih.gov/32958376/
Grosso G, Godos J, Galvano F, Giovannucci EL. Coffee, caffeine, and health outcomes: an umbrella review. Annu Rev Nutr. 2017;37:131–56. https://pubmed.ncbi.nlm.nih.gov/28826374/
Thomas DR, Hodges ID. Dietary research on coffee: improving adjustment for confounding. Curr Dev Nutr. 2020;4(nzz142). https://pubmed.ncbi.nlm.nih.gov/31938763/
Duregon E, Bernier M, de Cabo R. A glance back at the journal of gerontology – coffee, dietary interventions and life span. J Geront A Biol Sci Med Sci. 2020;75(11):2029–30. https://pubmed.ncbi.nlm.nih.gov/33057720/
Li Q, Liu Y, Sun X, et al. Caffeinated and decaffeinated coffee consumption and risk of all-cause mortality: a dose – response meta-analysis of cohort studies. J Hum Nut Diet. 2019;32(3):279–87. https://pubmed.ncbi.nlm.nih.gov/30786114/
Spiegelhalter D. Using speed of ageing and “microlives” to communicate the effects of lifetime habits and environment. BMJ. 2012 Dec 14;345:e8223. https://pubmed.ncbi.nlm.nih.gov/23247978/
Poole R, Kennedy OJ, Roderick P, Fallowfield JA, Hayes PC, Parkes J. Coffee consumption and health: umbrella review of meta-analyses of multiple health outcomes. BMJ. 2017;359:j5024. https://pubmed.ncbi.nlm.nih.gov/29167102/
Loftfield E, Cornelis MC, Caporaso N, Yu K, Sinha R, Freedman N. Association of coffee drinking with mortality by genetic variation in caffeine metabolism: findings from the UK Biobank. JAMA Intern Med. 2018;178(8):1086. https://pubmed.ncbi.nlm.nih.gov/29971434/
Poole R, Kennedy OJ, Roderick P, Fallowfield JA, Hayes PC, Parkes J. Coffee consumption and health: umbrella review of meta-analyses of multiple health outcomes. BMJ. 2017;359:j5024. https://pubmed.ncbi.nlm.nih.gov/29167102/
Gao LJ, Dai Y, Li XQ, Meng S, Zhong ZQ, Xu SJ. Chlorogenic acid enhances autophagy by upregulating lysosomal function to protect against SH-SY5Y cell injury induced by H2O2. Exp Ther Med. 2021;21(5):426. https://pubmed.ncbi.nlm.nih.gov/33747165/
Ludwig IA, Mena P, Calani L, et al. Variations in caffeine and chlorogenic acid contents of coffees: what are we drinking? Food Funct. 2014;5(8):1718–26. https://pubmed.ncbi.nlm.nih.gov/25014672/
Mills CE, Oruna-Concha MJ, Mottram DS, Gibson GR, Spencer JPE. The effect of processing on chlorogenic acid content of commercially available coffee. Food Chem. 2013;141(4):3335–40. https://pubmed.ncbi.nlm.nih.gov/23993490/
Ludwig IA, Mena P, Calani L, et al. Variations in caffeine and chlorogenic acid contents of coffees: what are we drinking? Food Funct. 2014;5(8):1718–26. https://pubmed.ncbi.nlm.nih.gov/25014672/
Corrêa TAF, Monteiro MP, Mendes TMN, et al. Medium light and medium roast paper-filtered coffee increased antioxidant capacity in healthy volunteers: results of a randomized trial. Plant Foods Hum Nutr. 2012;67(3):277–82. https://pubmed.ncbi.nlm.nih.gov/22766993/
DiBaise JK. A randomized, double-blind comparison of two different coffee-roasting processes on development of heartburn and dyspepsia in coffee-sensitive individuals. Dig Dis Sci. 2003;48(4):652–6. https://pubmed.ncbi.nlm.nih.gov/12741451/
Liu J, Wang Q, Zhang H, Yu D, Jin S, Ren F. Interaction of chlorogenic acid with milk proteins analyzed by spectroscopic and modeling methods. Spectrosc Lett. 2016;49(1):44–50. https://www.tandfonline.com/doi/full/10.1080/00387010.2015.1066826
Duarte GS, Farah A. Effect of simultaneous consumption of milk and coffee on chlorogenic acids’ bioavailability in humans. J Agric Food Chem. 2011;59(14):7925–31. https://pubmed.ncbi.nlm.nih.gov/21627318/
Lorenz M, Jochmann N, von Krosigk A, et al. Addition of milk prevents vascular protective effects of tea. Eur Heart J. 2007;28(2):219–23. https://pubmed.ncbi.nlm.nih.gov/17213230/
Serafini M, Testa MF, Villaño D, et al. Antioxidant activity of blueberry fruit is impaired by association with milk. Free Radic Biol Med. 2009;46(6):769–74. https://pubmed.ncbi.nlm.nih.gov/19135520/
Serafini M, Bugianesi R, Maiani G, Valtuena S, De Santis S, Crozier A. Plasma antioxidants from chocolate. Nature. 2003;424(6952):1013. https://pubmed.ncbi.nlm.nih.gov/12944955/
Budryn G, Palecz B, Rachwal-Rosiak D, et al. Effect of inclusion of hydroxycinnamic and chlorogenic acids from green coffee bean in ß-cyclodextrin on their interactions with whey, egg white and soy protein isolates. Food Chem. 2015;168:276–87. https://pubmed.ncbi.nlm.nih.gov/25172711/
Felberg I, Farah A, Monteiro M, et al. Effect of simultaneous consumption of soymilk and coffee on the urinary excretion of isoflavones, chlorogenic acids and metabolites in healthy adults. J Funct Foods. 2015;19:688–99. https://www.sciencedirect.com/science/article/pii/S1756464615004910?via%3Dihub
Colombo R, Papetti A. An outlook on the role of decaffeinated coffee in neurodegenerative diseases. Crit Rev Food Sci Nutr. 2020;60(5):760–79. https://pubmed.ncbi.nlm.nih.gov/30614247/
Tverdal A, Selmer R, Cohen JM, Thelle DS. Coffee consumption and mortality from cardiovascular diseases and total mortality: does the brewing method matter? Eur J Prev Cardiol. 2020;27(18):1986–93. https://pubmed.ncbi.nlm.nih.gov/32320635/
Aubin HJ, Luquiens A, Berlin I. Letter by Aubin et al regarding article, “Association of coffee consumption with total and cause-specific mortality in 3 large prospective cohorts.” Circulation. 2016;133(20):e659. https://pubmed.ncbi.nlm.nih.gov/27185028/
Sakaki JR, Melough MM, Provatas AA, Perkins C, Chun OK. Evaluation of estrogenic chemicals in capsule and French press coffee using ultra-performance liquid chromatography with tandem mass spectrometry. Toxicol Rep. 2020;7:1020–4. https://pubmed.ncbi.nlm.nih.gov/32874926/
Yang CZ, Yaniger SI, Jordan VC, Klein DJ, Bittner GD. Most plastic products release estrogenic chemicals: a potential health problem that can be solved. Environ Health Perspect. 2011;119(7):989–96. https://pubmed.ncbi.nlm.nih.gov/21367689/
Sakaki JR, Melough MM, Provatas AA, Perkins C, Chun OK. Evaluation of estrogenic chemicals in capsule and French press coffee using ultra-performance liquid chromatography with tandem mass spectrometry. Toxicol Rep. 2020;7:1020–4. https://pubmed.ncbi.nlm.nih.gov/32874926/
Li M, Wang M, Guo W, Wang J, Sun X. The effect of caffeine on intraocular pressure: a systematic review and meta-analysis. Graefes Arch Clin Exp Ophthalmol. 2011;249(3):435–42. https://pubmed.ncbi.nlm.nih.gov/20706731/
Kang JH, Willett WC, Rosner BA, Hankinson SE, Pasquale LR. Caffeine consumption and the risk of primary open-angle glaucoma: a prospective cohort study. Invest Ophthalmol Vis Sci. 2008;49(5):1924–31. https://pubmed.ncbi.nlm.nih.gov/18263806/
Gleason JL, Richter HE, Redden DT, Goode PS, Burgio KL, Markland AD. Caffeine and urinary incontinence in US women. Int Urogynecol J. 2013;24(2):295–302. https://pubmed.ncbi.nlm.nih.gov/22699886/
Davis NJ, Vaughan CP, Johnson TM, et al. Caffeine intake and its association with urinary incontinence in United States men: results from National Health and Nutrition Examination Surveys 2005–2006 and 2007–2008. J Urol. 2013;189(6):2170–4. https://pubmed.ncbi.nlm.nih.gov/23276513/
Bonilha L, Li LM. Heavy coffee drinking and epilepsy. Seizure. 2004;13(4):284–5. https://pubmed.ncbi.nlm.nih.gov/15121141/
Surdea-Blaga T, Negrutiu DE, Palage M, Dumitrascu DL. Food and gastroesophageal reflux disease. Curr Med Chem. 2019;26(19):3497–511. https://pubmed.ncbi.nlm.nih.gov/28521699/
Lloret-Linares C, Lafuente-Lafuente C, Chassany O, et al. Does a single cup of coffee at dinner alter the sleep? A controlled cross-over randomised trial in real-life conditions. Nutr Diet. 2012;69(4):250–5. https://onlinelibrary.wiley.com/doi/10.1111/j.1747–0080.2012.01601.x
Poole R, Kennedy OJ, Roderick P, Fallowfield JA, Hayes PC, Parkes J. Coffee consumption and health: umbrella review of meta-analyses of multiple health outcomes. BMJ. 2017;359:j5024. https://pubmed.ncbi.nlm.nih.gov/29167102/
Son H, Song HJ, Seo HJ, Lee H, Choi SM, Lee S. The safety and effectiveness of self-administered coffee enema: a systematic review of case reports. Medicine. 2020;99(36):e21998. https://pubmed.ncbi.nlm.nih.gov/32899046/
Dirks-Naylor AJ. The benefits of coffee on skeletal muscle. Life Sci. 2015;143:182–6. https://pubmed.ncbi.nlm.nih.gov/26546720/
Juliano LM, Griffiths RR. A critical review of caffeine withdrawal: empirical validation of symptoms and signs, incidence, severity, and associated features. Psychopharmacology (Berl). 2004;176(1):1–29. https://pubmed.ncbi.nlm.nih.gov/15448977/
O’Keefe JH, Bhatti SK, Patil HR, DiNicolantonio JJ, Lucan SC, Lavie CJ. Effects of habitual coffee consumption on cardiometabolic disease, cardiovascular health, and all-cause mortality. J Am Coll Cardiol. 2013;62(12):1043–51. https://pubmed.ncbi.nlm.nih.gov/23871889/
Mendez JD. The other legacy of Antonie van Leeuwenhoek: the polyamines. J Clin Mol Endocrinol. 2017;02(01):e107. https://clinical-and-molecular-endocrinology.imedpub.com/the-other-legacy-of-antonie-van-leeuwenhoek-the-polyamines.php?aid=19400
Bachrach U. The early history of polyamine research. Plant Physiol Biochem. 2010;48(7):490–5. https://pubmed.ncbi.nlm.nih.gov/20219382/
Guerra GP, Rubin MA, Mello CF. Modulation of learning and memory by natural polyamines. Pharmacol Res. 2016;112:99–118. https://pubmed.ncbi.nlm.nih.gov/27015893/
Madeo F, Bauer MA, Carmona-Gutierrez D, Kroemer G. Spermidine: a physiological autophagy inducer acting as an anti-aging vitamin in humans? Autophagy. 2019;15(1):165–8. https://pubmed.ncbi.nlm.nih.gov/30306826/
Madeo F, Eisenberg T, Pietrocola F, Kroemer G. Spermidine in health and disease. Science. 2018;359(6374):eaan2788. https://pubmed.ncbi.nlm.nih.gov/29371440/
Hunter DC, Burritt DJ. Polyamines of plant origin: an important dietary consideration for human health. In: Rao V, ed. Phytochemicals as Nutraceuticals: Global Approaches to Their Role in Nutrition and Health. InTech; 2012:225–44. https://www.intechopen.com/chapters/32904
Kaeberlein M. Spermidine surprise for a long life. Nat Cell Biol. 2009;11(11):1277–8. https://pubmed.ncbi.nlm.nih.gov/19884883/
Hunter DC, Burritt DJ. Polyamines of plant origin: an important dietary consideration for human health. In: Rao V, ed. Phytochemicals as Nutraceuticals: Global Approaches to Their Role in Nutrition and Health. InTech; 2012:225–44. https://www.intechopen.com/chapters/32904
Minois N, Carmona-Gutierrez D, Madeo F. Polyamines in aging and disease. Aging (Albany NY). 2011;3(8):716–32. https://pubmed.ncbi.nlm.nih.gov/21869457/
Soda K, Dobashi Y, Kano Y, Tsujinaka S, Konishi F. Polyamine-rich food decreases age-associated pathology and mortality in aged mice. Exp Gerontol. 2009;44(11):727–32. https://pubmed.ncbi.nlm.nih.gov/19735716/
Yue F, Li W, Zou J, et al. Spermidine prolongs lifespan and prevents liver fibrosis and hepatocellular carcinoma by activating map1s-mediated autophagy. Cancer Res. 2017;77(11):2938–51. https://pubmed.ncbi.nlm.nih.gov/28386016/
Eisenberg T, Knauer H, Schauer A, et al. Induction of autophagy by spermidine promotes longevity. Nat Cell Biol. 2009;11(11):1305–14. https://pubmed.ncbi.nlm.nih.gov/19801973/
Rudman D, Kutner MH, Chawla RK, Goldsmith MA, Blackston RD, Bain R. Serum and urine polyamines in normal and in short children. J Clin Invest. 1979;64(6):1661–8. https://pubmed.ncbi.nlm.nih.gov/500832/
Pucciarelli S, Moreschini B, Micozzi D, et al. Spermidine and spermine are enriched in whole blood of nona/centenarians. Rejuvenation Res. 2012;15(6):590–5. https://pubmed.ncbi.nlm.nih.gov/22950434/
Piore A. Can blood from young people slow aging? Silicon Valley bets it will. Newsweek. April 7, 2021. https://www.newsweek.com/2021/04/16/can-blood-young-people-slow-aging-silicon-valley-has-bet-billions-it-will-1581447.html. Accessed December 25, 2022.; https://www.newsweek.com/2021/04/16/can-blood-young-people-slow-aging-silicon-valley-has-bet-billions-it-will-1581447.html
Viltard M, Durand S, Pérez-Lanzón M, et al. The metabolomic signature of extreme longevity: naked mole rats versus mice. Aging (Albany NY). 2019;11(14):4783–800. https://pubmed.ncbi.nlm.nih.gov/31346149/
Pucciarelli S, Moreschini B, Micozzi D, et al. Spermidine and spermine are enriched in whole blood of nona/centenarians. Rejuvenation Res. 2012;15(6):590–5. https://pubmed.ncbi.nlm.nih.gov/22950434/
Eisenberg T, Abdellatif M, Schroeder S, et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med. 2016;22(12):1428–38. https://pubmed.ncbi.nlm.nih.gov/27841876/
Flurkey K, Currer JM, Harrison DE. 2007. The Mouse in Aging Research. In The Mouse in Biomedical Research 2nd Edition. Fox JG, et al, editors. American College Laboratory Animal Medicine (Elsevier), Burlington, MA. pp. 637–72.; https://www.sciencedirect.com/science/article/abs/pii/B9780123694546500741?via%3Dihub
Eisenberg T, Abdellatif M, Schroeder S, et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med. 2016;22(12):1428–38. https://pubmed.ncbi.nlm.nih.gov/27841876/
Filfan M, Olaru A, Udristoiu I, et al. Long-term treatment with spermidine increases health span of middle-aged Sprague-Dawley male rats. GeroScience. 2020;42(3):937–49. https://pubmed.ncbi.nlm.nih.gov/32285289/
Pekar T, Bruckner K, Pauschenwein-Frantsich S, et al. The positive effect of spermidine in older adults suffering from dementia: first results of a 3-month trial. Wien Klin Wochenschr. 2021;133:484–91. https://pubmed.ncbi.nlm.nih.gov/33211152/
Handa AK, Fatima T, Mattoo AK. Polyamines: bio-molecules with diverse functions in plant and human health and disease. Front Chem. 2018;6. https://pubmed.ncbi.nlm.nih.gov/29468148/
Rinaldi F, Marzani B, Pinto D, Ramot Y. A spermidine-based nutritional supplement prolongs the anagen phase of hair follicles in humans: a randomized, placebo-controlled, double-blind study. Derm Pract Concept. Published online October 31, 2017:17–21.; https://pubmed.ncbi.nlm.nih.gov/29214104/
Metur SP, Klionsky DJ. The curious case of polyamines: spermidine drives reversal of B cell senescence. Autophagy. 2020;16(3):389–90. https://pubmed.ncbi.nlm.nih.gov/31795807/
Zhang H, Alsaleh G, Feltham J, et al. Polyamines control eIF5A hypusination, TFEB translation, and autophagy to reverse B cell senescence. Mol Cell. 2019;76(1):110–25.e9. https://pubmed.ncbi.nlm.nih.gov/31474573/
Metur SP, Klionsky DJ. The curious case of polyamines: spermidine drives reversal of B cell senescence. Autophagy. 2020;16(3):389–90. https://pubmed.ncbi.nlm.nih.gov/31795807/
de Cabo R, Navas P. Spermidine to the rescue for an aging heart. Nat Med. 2016;22(12):1389–90. https://pubmed.ncbi.nlm.nih.gov/27923032/
Eisenberg T, Abdellatif M, Schroeder S, et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med. 2016;22(12):1428–38. https://pubmed.ncbi.nlm.nih.gov/27841876/
Fetterman JL, Holbrook M, Flint N, et al. Restoration of autophagy in endothelial cells from patients with diabetes mellitus improves nitric oxide signaling. Atherosclerosis. 2016;247:207–17. https://pubmed.ncbi.nlm.nih.gov/26926601/
Eisenberg T, Abdellatif M, Schroeder S, et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med. 2016;22(12):1428–38. https://pubmed.ncbi.nlm.nih.gov/27841876/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Madeo F, Bauer MA, Carmona-Gutierrez D, Kroemer G. Spermidine: a physiological autophagy inducer acting as an anti-aging vitamin in humans? Autophagy. 2019;15(1):165–8. https://pubmed.ncbi.nlm.nih.gov/30306826/
Pekar T, Bruckner K, Pauschenwein-Frantsich S, et al. The positive effect of spermidine in older adults suffering from dementia: first results of a 3-month trial. Wien Klin Wochenschr. 2021;133:484–91. https://pubmed.ncbi.nlm.nih.gov/33211152/
Madeo F, Eisenberg T, Pietrocola F, Kroemer G. Spermidine in health and disease. Science. 2018;359(6374):eaan2788. https://pubmed.ncbi.nlm.nih.gov/29371440/
Madeo F, Hofer SJ, Pendl T, et al. Nutritional aspects of spermidine. Annu Rev Nutr. 2020;40(1):135–59. https://pubmed.ncbi.nlm.nih.gov/32634331/
Zoumas-Morse C, Rock CL, Quintana EL, Neuhouser ML, Gerner EW, Meyskens FL. Development of a polyamine database for assessing dietary intake. J Am Diet Assoc. 2007;107(6):1024–7. https://pubmed.ncbi.nlm.nih.gov/17524725/
Kalac P. Health effects and occurrence of dietary polyamines: a review for the period 2005–mid 2013. Food Chem. 2014;161:27–39. https://pubmed.ncbi.nlm.nih.gov/24837918/
Еще раз напомним, что американская единица объема «чашка» (cup) равна 240 мл: здесь и далее во всех рецептах. – Примеч. ред.
Ali MA, Poortvliet E, Strömberg R, Yngve A. Polyamines: total daily intake in adolescents compared to the intake estimated from the Swedish Nutrition Recommendations Objectified (Sno). Food Nutr Res. 2011;55(1):5455. https://pubmed.ncbi.nlm.nih.gov/21249160/
Varghese N, Werner S, Grimm A, Eckert A. Dietary mitophagy enhancer: a strategy for healthy brain aging? Antioxidants (Basel). 2020;9(10). https://pubmed.ncbi.nlm.nih.gov/33003315/
Handa AK, Fatima T, Mattoo AK. Polyamines: bio-molecules with diverse functions in plant and human health and disease. Front Chem. 2018;6. https://pubmed.ncbi.nlm.nih.gov/29468148/
Agricultural Research Service, United States Department of Agriculture. Dill weed, fresh. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=dill&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/172233/nutrients. Published April 1, 2019. Accessed April 30, 2021.; https://fdc.nal.usda.gov/fdc-app.html?query=dill&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/172233/nutrients
Kalac P. Health effects and occurrence of dietary polyamines: a review for the period 2005–mid 2013. Food Chem. 2014;161:27–39. https://pubmed.ncbi.nlm.nih.gov/24837918/
Agricultural Research Service, United States Department of Agriculture. Potato, baked, NFS. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=potato&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1102880/nutrients. Published October 30, 2020. Accessed April 30, 2021.; https://fdc.nal.usda.gov/fdc-app.html?query=potato&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1102880/nutrients
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nut Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Agricultural Research Service, United States Department of Agriculture. Garlic, raw. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=garlic&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1103354/nutrients. Published October 30, 2020. Accessed April 30, 2021.; https://fdc.nal.usda.gov/fdc-app.html?query=apples&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1102644/nutrients
Nishimura K, Shiina R, Kashiwagi K, Igarashi K. Decrease in polyamines with aging and their ingestion from food and drink. J Biochem. 2006;139(1):81–90. https://pubmed.ncbi.nlm.nih.gov/16428322/
Okamoto A, Sugi E, Koizumi Y, Yanagida F, Udaka S. Polyamine content of ordinary foodstuffs and various fermented foods. Biosci Biotechnol Biochem. 1997;61(9):1582–4. https://pubmed.ncbi.nlm.nih.gov/9339564/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Kalac P. Health effects and occurrence of dietary polyamines: a review for the period 2005–mid 2013. Food Chem. 2014;161:27–39. https://pubmed.ncbi.nlm.nih.gov/24837918/
Kalac P. Health effects and occurrence of dietary polyamines: a review for the period 2005–mid 2013. Food Chem. 2014;161:27–39. https://pubmed.ncbi.nlm.nih.gov/24837918/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Nishibori N, Fujihara S, Akatuki T. Amounts of polyamines in foods in Japan and intake by Japanese. Food Chem. 2007;100(2):491–7. https://www.sciencedirect.com/science/article/abs/pii/S0308814605008915?via%3Dihub
Nishimura K, Shiina R, Kashiwagi K, Igarashi K. Decrease in polyamines with aging and their ingestion from food and drink. J Biochem. 2006;139(1):81–90. https://pubmed.ncbi.nlm.nih.gov/16428322/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Cipolla BG, Havouis R, Moulinoux JP. Polyamine contents in current foods: a basis for polyamine reduced diet and a study of its long term observance and tolerance in prostate carcinoma patients. Amino Acids. 2007;33(2):203–12. https://pubmed.ncbi.nlm.nih.gov/17578651/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Nishibori N, Fujihara S, Akatuki T. Amounts of polyamines in foods in Japan and intake by Japanese. Food Chem. 2007;100(2):491–7. https://www.sciencedirect.com/science/article/abs/pii/S0308814605008915?via%3Dihub
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Cipolla BG, Havouis R, Moulinoux JP. Polyamine contents in current foods: a basis for polyamine reduced diet and a study of its long term observance and tolerance in prostate carcinoma patients. Amino Acids. 2007;33(2):203–12. https://pubmed.ncbi.nlm.nih.gov/17578651/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Nishimura K, Shiina R, Kashiwagi K, Igarashi K. Decrease in polyamines with aging and their ingestion from food and drink. J Biochem. 2006;139(1):81–90. https://pubmed.ncbi.nlm.nih.gov/16428322/
Cipolla BG, Havouis R, Moulinoux JP. Polyamine contents in current foods: a basis for polyamine reduced diet and a study of its long term observance and tolerance in prostate carcinoma patients. Amino Acids. 2007;33(2):203–12. https://pubmed.ncbi.nlm.nih.gov/17578651/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Kalac P. Health effects and occurrence of dietary polyamines: a review for the period 2005–mid 2013. Food Chem. 2014;161:27–39. https://pubmed.ncbi.nlm.nih.gov/24837918/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Cipolla BG, Havouis R, Moulinoux JP. Polyamine contents in current foods: a basis for polyamine reduced diet and a study of its long term observance and tolerance in prostate carcinoma patients. Amino Acids. 2007;33(2):203–12. https://pubmed.ncbi.nlm.nih.gov/17578651/
Cipolla BG, Havouis R, Moulinoux JP. Polyamine contents in current foods: a basis for polyamine reduced diet and a study of its long term observance and tolerance in prostate carcinoma patients. Amino Acids. 2007;33(2):203–12. https://pubmed.ncbi.nlm.nih.gov/17578651/
Nishibori N, Fujihara S, Akatuki T. Amounts of polyamines in foods in Japan and intake by Japanese. Food Chem. 2007;100(2):491–7. https://www.sciencedirect.com/science/article/abs/pii/S0308814605008915?via%3Dihub
Nishimura K, Shiina R, Kashiwagi K, Igarashi K. Decrease in polyamines with aging and their ingestion from food and drink. J Biochem. 2006;139(1):81–90. https://pubmed.ncbi.nlm.nih.gov/16428322/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Nishimura K, Shiina R, Kashiwagi K, Igarashi K. Decrease in polyamines with aging and their ingestion from food and drink. J Biochem. 2006;139(1):81–90. https://pubmed.ncbi.nlm.nih.gov/16428322/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Zoumas-Morse C, Rock CL, Quintana EL, Neuhouser ML, Gerner EW, Meyskens FL. Development of a polyamine database for assessing dietary intake. J Am Diet Assoc. 2007;107(6):1024–7. https://pubmed.ncbi.nlm.nih.gov/17524725/
Buyukuslu N, Hizli H, Esin K, Garipagaoglu M. A cross-sectional study: nutritional polyamines in frequently consumed foods of the Turkish population. Foods. 2014;3(4):541–57. https://pubmed.ncbi.nlm.nih.gov/28234336/
Nishibori N, Fujihara S, Akatuki T. Amounts of polyamines in foods in Japan and intake by Japanese. Food Chem. 2007;100(2):491–7. https://www.sciencedirect.com/science/article/abs/pii/S0308814605008915?via%3Dihub
Reis GCL, Dala-Paula BM, Tavano OL, Guidi LR, Godoy HT, Gloria MBA. In vitro digestion of spermidine and amino acids in fresh and processed Agaricus bisporus mushroom. Food Res Int. 2020;137:109616. https://pubmed.ncbi.nlm.nih.gov/33233206/
Pietrocola F, Castoldi F, Kepp O, Carmona-Gutierrez D, Madeo F, Kroemer G. Spermidine reduces cancer-related mortality in humans. Autophagy. 2019;15(2):362–5. https://pubmed.ncbi.nlm.nih.gov/30354939/
Nishimura K, Shiina R, Kashiwagi K, Igarashi K. Decrease in polyamines with aging and their ingestion from food and drink. J Biochem. 2006;139(1):81–90. https://pubmed.ncbi.nlm.nih.gov/16428322/
Eisenberg T, Abdellatif M, Schroeder S, et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med. 2016;22(12):1428–38. https://pubmed.ncbi.nlm.nih.gov/27841876/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Agricultural Research Service, United States Department of Agriculture. Mangos, raw. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/food-details/169910/nutrients. Published April 2018. Accessed February 10, 2023.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/169910/nutrients
Nishimura K, Shiina R, Kashiwagi K, Igarashi K. Decrease in polyamines with aging and their ingestion from food and drink. J Biochem. 2006;139(1):81–90. https://pubmed.ncbi.nlm.nih.gov/16428322/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Soda K, Binh P, Kawakami M. Mediterranean diet and polyamine intake: possible contribution of increased polyamine intake to inhibition of age-associated disease. NDS. Published online December 2010:1.; https://www.dovepress.com/mediterranean-diet-and-polyamine-intake-possible-contribution-of-incre-peer-reviewed-fulltext-article-NDS
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Okamoto A, Sugi E, Koizumi Y, Yanagida F, Udaka S. Polyamine content of ordinary foodstuffs and various fermented foods. Biosci Biotechnol Biochem. 1997;61(9):1582–4. https://pubmed.ncbi.nlm.nih.gov/9339564/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Cipolla BG, Havouis R, Moulinoux JP. Polyamine contents in current foods: a basis for polyamine reduced diet and a study of its long term observance and tolerance in prostate carcinoma patients. Amino Acids. 2007;33(2):203–12. https://pubmed.ncbi.nlm.nih.gov/17578651/
Konakovsky V, Focke M, Hoffmann-Sommergruber K, et al. Levels of histamine and other biogenic amines in high-quality red wines. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 2011;28(4):408–16. https://pubmed.ncbi.nlm.nih.gov/21337238/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Okamoto A, Sugi E, Koizumi Y, Yanagida F, Udaka S. Polyamine content of ordinary foodstuffs and various fermented foods. Biosci Biotechnol Biochem. 1997;61(9):1582–4. https://pubmed.ncbi.nlm.nih.gov/9339564/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Agricultural Research Service, United States Department of Agriculture. Lettuce, raw. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=lettuce&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1103358/nutrients. Published October 30, 2020. Accessed April 30, 2021.; https://fdc.nal.usda.gov/fdc-app.html?query=apples&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1102644/nutrients
Fukushima T, Tanaka K, Ushijima K, Moriyama M. Retrospective study of preventive effect of maize on mortality from Parkinson’s disease in Japan. Asia Pac J Clin Nutr. 2003;12(4):447–50. https://pubmed.ncbi.nlm.nih.gov/14672869/
McCarty MF, Lerner A. Perspective: low risk of Parkinson’s disease in quasi-vegan cultures may reflect GCN2-mediated upregulation of Parkin. Adv Nutr. 2021;12(2):355–62. https://pubmed.ncbi.nlm.nih.gov/32945884/
Rossetto MRM, Vianello F, Saeki MJ, Lima GPP. Polyamines in conventional and organic vegetables exposed to exogenous ethylene. Food Chem. 2015;188:218–24. https://pubmed.ncbi.nlm.nih.gov/26041185/
Kalac¿ P, Krausová P. A review of dietary polyamines: formation, implications for growth and health and occurrence in foods. Food Chem. 2005;90(1–2):219–30. https://www.sciencedirect.com/science/article/abs/pii/S0308814604002961?via%3Dihub
Kozová M, Kalac P, Pelikánová T. Contents of biologically active polyamines in chicken meat, liver, heart and skin after slaughter and their changes during meat storage and cooking. Food Chem. 2009;116(2):419–25. https://www.sciencedirect.com/science/article/abs/pii/S0308814609002441?via%3Dihub
.
Binh PNT, Soda K, Kawakami M. Gross domestic product and dietary pattern among 49 western countries with a focus on polyamine intake. Health. 2010;02(11):1327–34. https://www.scirp.org/journal/paperinformation.aspx?paperid=3116
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Soda K, Binh P, Kawakami M. Mediterranean diet and polyamine intake: possible contribution of increased polyamine intake to inhibition of age-associated disease. NDS. Published online December 2010:1.; https://www.dovepress.com/mediterranean-diet-and-polyamine-intake-possible-contribution-of-incre-peer-reviewed-fulltext-article-NDS
Arulkumar A, Paramithiotis S, Paramasivam S. Biogenic amines in fresh fish and fishery products and emerging control. Aquac Fish. Published online March 16, 2021. https://www.sciencedirect.com/science/article/pii/S2468550X21000198. Accessed December 25, 2022.; https://www.sciencedirect.com/science/article/pii/S2468550X21000198
Cipolla BG, Havouis R, Moulinoux JP. Polyamine contents in current foods: a basis for polyamine reduced diet and a study of its long term observance and tolerance in prostate carcinoma patients. Amino Acids. 2007;33(2):203–12. https://pubmed.ncbi.nlm.nih.gov/17578651/
Kalac P. Health effects and occurrence of dietary polyamines: a review for the period 2005–mid 2013. Food Chem. 2014;161:27–39. https://pubmed.ncbi.nlm.nih.gov/24837918/
Soda K, Binh P, Kawakami M. Mediterranean diet and polyamine intake: possible contribution of increased polyamine intake to inhibition of age-associated disease. NDS. Published online December 2010:1.; https://www.dovepress.com/mediterranean-diet-and-polyamine-intake-possible-contribution-of-incre-peer-reviewed-fulltext-article-NDS
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Kalac P. Health effects and occurrence of dietary polyamines: a review for the period 2005–mid 2013. Food Chem. 2014;161:27–39. https://pubmed.ncbi.nlm.nih.gov/24837918/
Nishimura K, Shiina R, Kashiwagi K, Igarashi K. Decrease in polyamines with aging and their ingestion from food and drink. J Biochem. 2006;139(1):81–90. https://pubmed.ncbi.nlm.nih.gov/16428322/
Nishibori N, Fujihara S, Akatuki T. Amounts of polyamines in foods in Japan and intake by Japanese. Food Chem. 2007;100(2):491–7. https://www.sciencedirect.com/science/article/abs/pii/S0308814605008915?via%3Dihub
Cipolla BG, Havouis R, Moulinoux JP. Polyamine contents in current foods: a basis for polyamine reduced diet and a study of its long term observance and tolerance in prostate carcinoma patients. Amino Acids. 2007;33(2):203–12. https://pubmed.ncbi.nlm.nih.gov/17578651/
Nishibori N, Fujihara S, Akatuki T. Amounts of polyamines in foods in Japan and intake by Japanese. Food Chem. 2007;100(2):491–7. https://www.sciencedirect.com/science/article/abs/pii/S0308814605008915?via%3Dihub
Kalac P. Health effects and occurrence of dietary polyamines: a review for the period 2005–mid 2013. Food Chem. 2014;161:27–39. https://pubmed.ncbi.nlm.nih.gov/24837918/
Nishibori N, Fujihara S, Akatuki T. Amounts of polyamines in foods in Japan and intake by Japanese. Food Chem. 2007;100(2):491–7. https://www.sciencedirect.com/science/article/abs/pii/S0308814605008915?via%3Dihub
Atiya Ali M, Poortvliet E, Strömberg R, Yngve A. Polyamines in foods: development of a food database. Food Nutr Res. 2011;55(1):5572. https://pubmed.ncbi.nlm.nih.gov/21249159/
Kiechl S, Pechlaner R, Willeit P, et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am J Clin Nutr. 2018;108(2):371–80. https://pubmed.ncbi.nlm.nih.gov/29955838/
Pekar T, Bruckner K, Pauschenwein-Frantsich S, et al. The positive effect of spermidine in older adults suffering from dementia: first results of a 3-month trial. Wien Klin Wochenschr. 2021;133:484–91. https://pubmed.ncbi.nlm.nih.gov/33211152/
MacMillen H. Could consuming semen make you live longer? Cosmopolitan. https://www.cosmo.ph/relationships/could-semen-make-you-live-longer-src-intl-a1553–20161201?ref=feed_1. Published online November 17, 2016. Accessed May 19, 2021.; https://www.cosmo.ph/relationships/could-semen-make-you-live-longer-src-intl-a1553-20161201?ref=feed_1
Scott E. Drinking semen might help you live longer. Metro.co.uk. https://metro.co.uk/2016/11/18/drinking-semen-might-actually-help-you-live-longer-6266961/. Published November 18, 2016. Accessed April 29, 2021.; https://metro.co.uk/2016/11/18/drinking-semen-might-actually-help-you-live-longer-6266961/
Owen DH, Katz DF. A review of the physical and chemical properties of human semen and the formulation of a semen simulant. J Androl. 2005;26(4):459–69. https://pubmed.ncbi.nlm.nih.gov/15955884/
Fair WR, Clark RB, Wehner N. A correlation of seminal polyamine levels and semen analysis in the human. Fertil Steril. 1972;23(1):38–42. https://pubmed.ncbi.nlm.nih.gov/5008948/
Definition of testament. Merriam-Webster.com. https://www.merriam-webster.com/dictionary/testament. Accessed February 11, 2023.; https://www.merriam-webster.com/dictionary/testament
Agricultural Research Service, United States Department of Agriculture. Wheat germ, plain. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=wheat+germ&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1101819/nutrients. Published October 30, 2020. Accessed April 30, 2021.; https://fdc.nal.usda.gov/fdc-app.html?query=wheat+germ&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1101819/nutrients
Liaqat H, Jeong E, Kim KJ, Kim JY. Effect of wheat germ on metabolic markers: a systematic review and meta-analysis of randomized controlled trials. Food Sci Biotechnol. 2020;29(6):739–49. https://pubmed.ncbi.nlm.nih.gov/32523783/
McCarty MF, Lerner A. Perspective: low risk of Parkinson’s disease in quasi-vegan cultures may reflect GCN2-mediated upregulation of Parkin. Adv Nutr. 2021;12(2):355–62. https://pubmed.ncbi.nlm.nih.gov/32945884/
Cara L, Borel P, Armand M, et al. Plasma lipid lowering effects of wheat germ in hypercholesterolemic subjects. Plant Foods Hum Nutr. 1991;41(2):135–50. https://pubmed.ncbi.nlm.nih.gov/1649472/
Moreira-Rosário A, Pinheiro H, Marques C, Teixeira JA, Calhau C, Azevedo LF. Does intake of bread supplemented with wheat germ have a preventive role on cardiovascular disease risk markers in healthy volunteers? A randomised, controlled, crossover trial. BMJ Open. 2019;9(1):e023662. https://pubmed.ncbi.nlm.nih.gov/30659039/
Atallahi M, Amir Ali Akbari S, Mojab F, Alavi Majd H. Effects of wheat germ extract on the severity and systemic symptoms of primary dysmenorrhea: a randomized controlled clinical trial. Iran Red Crescent Med J. 2014;16(8). https://pubmed.ncbi.nlm.nih.gov/25389490/
Delzenne NM, Neyrinck AM, Cani PD. Gut microbiota and metabolic disorders: how prebiotic can work? Br J Nutr. 2013;109 Suppl 2:S81–5. https://pubmed.ncbi.nlm.nih.gov/23360884/
Milovic V. Polyamines in the gut lumen: bioavailability and biodistribution. Eur J Gastroenterol Hepatol. 2001;13(9):1021–5. https://pubmed.ncbi.nlm.nih.gov/11564949/
Matsumoto M, Kurihara S, Kibe R, Ashida H, Benno Y. Longevity in mice is promoted by probiotic-induced suppression of colonic senescence dependent on upregulation of gut bacterial polyamine production. PLoS One. 2011;6(8):e23652. https://pubmed.ncbi.nlm.nih.gov/21858192/
Noack J, Kleessen B, Proll J, Dongowski G, Blaut M. Dietary guar gum and pectin stimulate intestinal microbial polyamine synthesis in rats. J Nutr. 1998;128(8):1385–91. https://pubmed.ncbi.nlm.nih.gov/9687560/
Hunter DC, Burritt DJ. Polyamines of plant origin: an important dietary consideration for human health. In: Rao V, ed. Phytochemicals as Nutraceuticals: Global Approaches to Their Role in Nutrition and Health. InTech; 2012:225–44. https://www.intechopen.com/chapters/32904
Mäkivuokko H, Tiihonen K, Tynkkynen S, Paulin L, Rautonen N. The effect of age and non-steroidal anti-inflammatory drugs on human intestinal microbiota composition. Br J Nutr. 2010;103(2):227–34. https://pubmed.ncbi.nlm.nih.gov/19703328/
Hunter DC, Burritt DJ. Polyamines of plant origin: an important dietary consideration for human health. In: Rao V, ed. Phytochemicals as Nutraceuticals: Global Approaches to Their Role in Nutrition and Health. InTech; 2012:225–44. https://www.intechopen.com/chapters/32904
Matsumoto M, Aranami A, Ishige A, Watanabe K, Benno Y. LKM512 yogurt consumption improves the intestinal environment and induces the T-helper type 1 cytokine in adult patients with intractable atopic dermatitis. Clin Exp Allergy. 2007;37(3):358–70. https://pubmed.ncbi.nlm.nih.gov/17359386/
Matsumoto M, Kurihara S, Kibe R, Ashida H, Benno Y. Longevity in mice is promoted by probiotic-induced suppression of colonic senescence dependent on upregulation of gut bacterial polyamine production. PLoS One. 2011;6(8):e23652. https://pubmed.ncbi.nlm.nih.gov/21858192/
Kibe R, Kurihara S, Sakai Y, et al. Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Sci Rep. 2014;4(1):4548. https://pubmed.ncbi.nlm.nih.gov/24686447/
Matsumoto M, Kitada Y, Naito Y. Endothelial function is improved by inducing microbial polyamine production in the gut: a randomized placebo-controlled trial. Nutrients. 2019;11(5). https://pubmed.ncbi.nlm.nih.gov/31137855/
Matsumoto M. Prevention of atherosclerosis by the induction of microbial polyamine production in the intestinal lumen. Biol Pharm Bull. 2020;43(2):221–9. https://pubmed.ncbi.nlm.nih.gov/32009110/
Noack J, Kleessen B, Proll J, Dongowski G, Blaut M. Dietary guar gum and pectin stimulate intestinal microbial polyamine synthesis in rats. J Nutr. 1998;128(8):1385–91. https://pubmed.ncbi.nlm.nih.gov/9687560/
de Cabo R, Navas P. Spermidine to the rescue for an aging heart. Nat Med. 2016;22(12):1389–90. https://pubmed.ncbi.nlm.nih.gov/27923032/
Madeo F, Eisenberg T, Pietrocola F, Kroemer G. Spermidine in health and disease. Science. 2018;359(6374):eaan2788. https://pubmed.ncbi.nlm.nih.gov/29371440/
Pietrocola F, Castoldi F, Kepp O, Carmona-Gutierrez D, Madeo F, Kroemer G. Spermidine reduces cancer-related mortality in humans. Autophagy. 2019;15(2):362–5. https://pubmed.ncbi.nlm.nih.gov/30354939/
Chavez-Dominguez R, Perez-Medina M, Lopez-Gonzalez JS, Galicia-Velasco M, Aguilar-Cazares D. The double-edge sword of autophagy in cancer: from tumor suppression to pro-tumor activity. Front Oncol. 2020;10. https://pubmed.ncbi.nlm.nih.gov/33117715/
Madeo F, Eisenberg T, Pietrocola F, Kroemer G. Spermidine in health and disease. Science. 2018;359(6374):eaan2788. https://pubmed.ncbi.nlm.nih.gov/29371440/
Madeo F, Eisenberg T, Pietrocola F, Kroemer G. Spermidine in health and disease. Science. 2018;359(6374):eaan2788. https://pubmed.ncbi.nlm.nih.gov/29371440/
Barardo D, Thornton D, Thoppil H, et al. The DrugAge database of aging-related drugs. Aging Cell. 2017;16(3):594–7. https://pubmed.ncbi.nlm.nih.gov/28299908/
DrugAge: database of ageing-related drugs. https://genomics.senescence.info/drugs/stats.php. Updated February 7, 2023. Accessed February 11, 2023.; https://genomics.senescence.info/drugs/stats.php
Janssens GE, Houtkooper RH. Identification of longevity compounds with minimized probabilities of side effects. Biogerontology. 2020;21(6):709–19. https://pubmed.ncbi.nlm.nih.gov/32562114/
Hunter DC, Burritt DJ. Polyamines of plant origin: an important dietary consideration for human health. In: Rao V, ed. Phytochemicals as Nutraceuticals: Global Approaches to Their Role in Nutrition and Health. InTech; 2012:225–44. https://www.intechopen.com/chapters/32904
Larqué E, Sabater-Molina M, Zamora S. Biological significance of dietary polyamines. Nutrition. 2007;23(1):87–95. https://pubmed.ncbi.nlm.nih.gov/17113752/
Khandia R, Dadar M, Munjal A, et al. A comprehensive review of autophagy and its various roles in infectious, non-infectious, and lifestyle diseases: current knowledge and prospects for disease prevention, novel drug design, and therapy. Cells. 2019;8(7):674. https://pubmed.ncbi.nlm.nih.gov/31277291/
Hayflick L, Moorhead PS. 1961. The serial cultivation of human diploid cell strains. Exp. Cell Res. 25, 585–621.; https://pubmed.ncbi.nlm.nih.gov/13905658/
Zhang H, Simon AK. Polyamines reverse immune senescence via the translational control of autophagy. Autophagy. 2020;16(1):181–2. https://pubmed.ncbi.nlm.nih.gov/31679458/
Luo J, Si H, Jia Z, Liu D. Dietary anti-aging polyphenols and potential mechanisms. Antioxidants. 2021;10(2):283. https://pubmed.ncbi.nlm.nih.gov/33668479/
Schmitt R. Senotherapy: growing old and staying young? Pflugers Arch-Eur J Physiol. 2017;469(9):1051–9. https://pubmed.ncbi.nlm.nih.gov/28389776/
van Deursen JM. Senolytic therapies for healthy longevity. Science. 2019;364(6441):636–7. https://pubmed.ncbi.nlm.nih.gov/31097655/
Baker DJ, Petersen RC. Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives. J Clin Invest. 2018;128(4):1208–16. https://pubmed.ncbi.nlm.nih.gov/29457783/
Davan-Wetton CSA, Pessolano E, Perretti M, Montero-Melendez T. Senescence under appraisal: hopes and challenges revisited. Cell Mol Life Sci. 2021;78(7):3333–54. https://pubmed.ncbi.nlm.nih.gov/33439271/
Prašnikar E, Borišek J, Perdih A. Senescent cells as promising targets to tackle age-related diseases. Ageing Res Rev. 2021;66:101251. https://pubmed.ncbi.nlm.nih.gov/33385543/
Zhu Y, Tchkonia T, Pirtskhalava T, et al. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14(4):644–58. https://pubmed.ncbi.nlm.nih.gov/25754370/
van Deursen JM. Senolytic therapies for healthy longevity. Science. 2019;364(6441):636–7. https://pubmed.ncbi.nlm.nih.gov/31097655/
Mau T, Yung R. Adipose tissue inflammation in aging. Exp Gerontol. 2018;105:27–31. https://pubmed.ncbi.nlm.nih.gov/29054535/
Prašnikar E, Borišek J, Perdih A. Senescent cells as promising targets to tackle age-related diseases. Ageing Res Rev. 2021;66:101251. https://pubmed.ncbi.nlm.nih.gov/33385543/
de Keizer PLJ. The fountain of youth by targeting senescent cells? Trends Mol Med. 2017;23(1):6–17. https://pubmed.ncbi.nlm.nih.gov/28041565/
Prašnikar E, Borišek J, Perdih A. Senescent cells as promising targets to tackle age-related diseases. Ageing Res Rev. 2021;66:101251. https://pubmed.ncbi.nlm.nih.gov/33385543/
van Deursen JM. Senolytic therapies for healthy longevity. Science. 2019;364(6441):636–7. https://pubmed.ncbi.nlm.nih.gov/31097655/
Hofmann B. Young blood rejuvenates old bodies: a call for reflection when moving from mice to men. Transfus Med Hemother. 2018;45(1):67–71. https://pubmed.ncbi.nlm.nih.gov/29593463/
Ludwig FC, Elashoff RM. Mortality in syngeneic rat parabionts of different chronological age. Trans N Y Acad Sci. 1972;34(7):582–7. https://pubmed.ncbi.nlm.nih.gov/4507935/
Lavazza A, Garasic M. Vampires 2.0? The ethical quandaries of young blood infusion in the quest for eternal life. Med Health Care Philos. 2020;23(3):421–32. https://pubmed.ncbi.nlm.nih.gov/32447568/
Rebo J, Mehdipour M, Gathwala R, et al. A single heterochronic blood exchange reveals rapid inhibition of multiple tissues by old blood. Nat Commun. 2016;7(1):13363. https://pubmed.ncbi.nlm.nih.gov/27874859/
Mehdipour M, Skinner C, Wong N, et al. Rejuvenation of three germ layers tissues by exchanging old blood plasma with saline-albumin. Aging (Albany NY). 2020;12(10):8790–819. https://pubmed.ncbi.nlm.nih.gov/32474458/
Boada M, López OL, Olazarán J, et al. A randomized, controlled clinical trial of plasma exchange with albumin replacement for Alzheimer’s disease: primary results of the AMBAR Study. Alzheimers Dement. 2020;16(10):1412–25. https://pubmed.ncbi.nlm.nih.gov/32715623/
Biller-Andorno N. Young blood for old hands? A recent anti-ageing trial prompts ethical questions. Swiss Med Wkly. 2016;146(3940):w14359. https://pubmed.ncbi.nlm.nih.gov/27684581/
Xu M, Pirtskhalava T, Farr JN, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018;24(8):1246–56. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6082705/
Baker DJ, Childs BG, Durik M, et al. Naturally occurring p16INK4a-positive cells shorten healthy lifespan. Nature. 2016;530(7589):184–9. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4845101/
de Keizer PLJ. The fountain of youth by targeting senescent cells? Trends Mol Med. 2017;23(1):6–17. https://pubmed.ncbi.nlm.nih.gov/28041565/
Chen X, Yi Z, Wong GT, et al. Is exercise a senolytic medicine? A systematic review. Aging Cell. 2021;20(1). https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7811843/
Fontana L, Mitchell SE, Wang B, et al. The effects of graded caloric restriction: XII. Comparison of mouse to human impact on cellular senescence in the colon. Aging Cell. 2018;17(3):e12746. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5946078/
Rusznyák S, Szent-Györgyi A. Vitamin P: flavonols as vitamins. Nature. 1936;138(3479):27. https://www.nature.com/articles/138027a0
Belinha I, Amorim MA, Rodrigues P, et al. Quercetin increases oxidative stress resistance and longevity in Saccharomyces cerevisiae. J Agric Food Chem. 2007;55(6):2446–51. https://pubmed.ncbi.nlm.nih.gov/17323973/
Formica JV, Regelson W. Review of the biology of quercetin and related bioflavonoids. Food Chem Toxicol. 1995;33(12):1061–80. https://pubmed.ncbi.nlm.nih.gov/8847003/
Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med. 2020;288(5):518–36. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7405395/
Zhu Y, Tchkonia T, Pirtskhalava T, et al. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14(4):644–58. https://pubmed.ncbi.nlm.nih.gov/25754370/
Geng L, Liu Z, Wang S, et al. Low-dose quercetin positively regulates mouse healthspan. Protein Cell. 2019;10(10):770–5. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6776572/
Yang D, Wang T, Long M, Li P. Quercetin: its main pharmacological activity and potential application in clinical medicine. Oxid Med Cell Longev. 2020;2020:1–13. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7790550/
Murphy MM, Barraj LM, Herman D, Bi X, Cheatham R, Randolph RK. Phytonutrient intake by adults in the United States in relation to fruit and vegetable consumption. J Acad Nutr Diet. 2012;112(2):222–9. https://pubmed.ncbi.nlm.nih.gov/22741166/
Mai F, Glomb MA. Isolation of phenolic compounds from iceberg lettuce and impact on enzymatic browning. J Agric Food Chem. 2013;61(11):2868–74. https://pubmed.ncbi.nlm.nih.gov/23473017/
Murphy MM, Barraj LM, Herman D, Bi X, Cheatham R, Randolph RK. Phytonutrient intake by adults in the United States in relation to fruit and vegetable consumption. J Acad Nutr Diet. 2012;112(2):222–9. https://pubmed.ncbi.nlm.nih.gov/22741166/
Agricultural Research Service, United States Department of Agriculture. Onions, raw. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=onion&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/170000/nutrients. Published April 1, 2019. Accessed May 11, 2021.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/170000/nutrients
Agricultural Research Service, United States Department of Agriculture. Onions, red, raw. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=onion&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/790577/nutrients. Published April 1, 2020. Accessed May 11, 2021.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/170000/nutrients
Agricultural Research Service, United States Department of Agriculture. Apple, raw. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=apples&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1102644/nutrients. Published October 30, 2020. Accessed May 11, 2021.; https://fdc.nal.usda.gov/fdc-app.html?query=apples&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/1102644/nutrients
Formica JV, Regelson W. Review of the biology of quercetin and related bioflavonoids. Food Chem Toxicol. 1995;33(12):1061–80. https://pubmed.ncbi.nlm.nih.gov/8847003/
Amanzadeh E, Esmaeili A, Rahgozar S, Nourbakhshnia M. Application of quercetin in neurological disorders: from nutrition to nanomedicine. Rev Neurosci. 2019;30(5):555–72. https://pubmed.ncbi.nlm.nih.gov/30753166/
Vida RG, Fittler A, Somogyi-Végh A, Poór M. Dietary quercetin supplements: assessment of online product informations and quantitation of quercetin in the products by high-performance liquid chromatography. Phytother Res. 2019;33(7):1912–20. https://pubmed.ncbi.nlm.nih.gov/31155780/
Harwood M, Danielewska-Nikiel B, Borzelleca JF, Flamm GW, Williams GM, Lines TC. A critical review of the data related to the safety of quercetin and lack of evidence of in vivo toxicity, including lack of genotoxic/carcinogenic properties. Food Chem Toxicol. 2007;45(11):2179–205. https://pubmed.ncbi.nlm.nih.gov/17698276/
Hickson LJ, Langhi Prata LGP, Bobart SA, et al. Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine. 2019;47:446–56. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6796530/
Briggs ADM, Mizdrak A, Scarborough P. A statin a day keeps the doctor away: comparative proverb assessment modelling study. BMJ. 2013;347:f7267. https://www.bmj.com/content/347/bmj.f7267
Bondonno NP, Bondonno CP, Blekkenhorst LC, et al. Flavonoid-rich apple improves endothelial function in individuals at risk for cardiovascular disease: a randomized controlled clinical trial. Mol Nutr Food Res. 2018;62(3). https://pubmed.ncbi.nlm.nih.gov/29086478/
Huang H, Liao D, Dong Y, Pu R. Effect of quercetin supplementation on plasma lipid profiles, blood pressure, and glucose levels: a systematic review and meta-analysis. Nutr Rev. 2020;78(8):615–26. https://pubmed.ncbi.nlm.nih.gov/31940027/
Tabrizi R, Tamtaji OR, Mirhosseini N, et al. The effects of quercetin supplementation on lipid profiles and inflammatory markers among patients with metabolic syndrome and related disorders: a systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr. 2020;60(11):1855–68. https://pubmed.ncbi.nlm.nih.gov/31017459/
Mohammadi-Sartang M, Mazloom Z, Sherafatmanesh S, Ghorbani M, Firoozi D. Effects of supplementation with quercetin on plasma C-reactive protein concentrations: a systematic review and meta-analysis of randomized controlled trials. Eur J Clin Nutr. 2017;71(9):1033–9. https://pubmed.ncbi.nlm.nih.gov/28537580/
Nakagawa T, Itoh M, Ohta K, et al. Improvement of memory recall by quercetin in rodent contextual fear conditioning and human early-stage Alzheimer’s disease patients. Neuroreport. 2016;27(9):671–6. https://pubmed.ncbi.nlm.nih.gov/27145228/
Nishimura M, Ohkawara T, Nakagawa T, et al. A randomized, double-blind, placebo-controlled study evaluating the effects of quercetin-rich onion on cognitive function in elderly subjects. FFHD. 2017;7(6):353–74. https://ffhdj.com/index.php/ffhd/article/view/334
Kalus U, Pindur G, Jung F, et al. Influence of the onion as an essential ingredient of the Mediterranean diet on arterial blood pressure and blood fluidity. Arzneimittelforschung. 2000;50(9):795–801. https://pubmed.ncbi.nlm.nih.gov/11050695/
Hertog MG, Feskens EJ, Hollman PC, Katan MB, Kromhout D. Dietary antioxidant flavonoids and risk of coronary heart disease: the Zutphen Elderly Study. Lancet. 1993;342(8878):1007–11. https://pubmed.ncbi.nlm.nih.gov/8105262/
Briggs ADM, Mizdrak A, Scarborough P. A statin a day keeps the doctor away: comparative proverb assessment modelling study. BMJ. 2013;347:f7267. https://www.bmj.com/content/347/bmj.f7267
Hwang HV, Tran DT, Rebuffatti MN, Li CS, Knowlton AA. Investigation of quercetin and hyperoside as senolytics in adult human endothelial cells. PLoS ONE. 2018;13(1):e0190374. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5760026/
Khan S, Shukla S, Sinha S, Meeran SM. Epigenetic targets in cancer and aging: dietary and therapeutic interventions. Expert Opin Ther Targets. 2016;20(6):689–703. https://pubmed.ncbi.nlm.nih.gov/26667209/
Geng L, Liu Z, Zhang W, et al. Chemical screen identifies a geroprotective role of quercetin in premature aging. Protein Cell. 2019;10(6):417–35. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6538594/
Chondrogianni N, Kapeta S, Chinou I, Vassilatou K, Papassideri I, Gonos ES. Anti-ageing and rejuvenating effects of quercetin. Exp Gerontol. 2010;45(10):763–71. https://pubmed.ncbi.nlm.nih.gov/20619334/
Zhu Y, Doornebal EJ, Pirtskhalava T, et al. New agents that target senescent cells: the flavone, fisetin, and the BCL–XL inhibitors, A1331852 and A1155463. Aging (Albany NY). 2017;9(3):955–63. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5391241/
Wyld L, Bellantuono I, Tchkonia T, et al. Senescence and cancer: a review of clinical implications of senescence and senotherapies. Cancers (Basel). 2020;12(8):2134. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7464619/
Li W, Qin L, Feng R, et al. Emerging senolytic agents derived from natural products. Mech Ageing Dev. 2019;181:1–6. https://pubmed.ncbi.nlm.nih.gov/31077707/
Yousefzadeh MJ, Zhu Y, McGowan SJ, et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine. 2018;36:18–28. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6197652/
Maher P, Akaishi T, Abe K. Flavonoid fisetin promotes ERK-dependent long-term potentiation and enhances memory. PNAS. 2006;103(44):16568–73. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1637622/
Farsad-Naeimi A, Alizadeh M, Esfahani A, Darvish Aminabad E. Effect of fisetin supplementation on inflammatory factors and matrix metalloproteinase enzymes in colorectal cancer patients. Food Funct. 2018;9(4):2025–31. https://pubmed.ncbi.nlm.nih.gov/29541713/
Yousefzadeh MJ, Zhu Y, McGowan SJ, et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine. 2018;36:18–28. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6197652/
U.S. National Library of Medicine. Search results for fisetin. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/results?cond=&term=fisetin&cntry=&state=&city=&dist=. Accessed May 29, 2021.; https://clinicaltrials.gov/ct2/results?cond=&term=fisetin&cntry=&state=&city=&dist=
Grynkiewicz G, Demchuk OM. New perspectives for fisetin. Front Chem. 2019;7:697. https://pubmed.ncbi.nlm.nih.gov/31750288/
Rabin BM, Joseph JA, Shukitt-Hale B. Effects of age and diet on the heavy particle-induced disruption of operant responding produced by a ground-based model for exposure to cosmic rays. Brain Res. 2005;1036(1–2):122–9. https://pubmed.ncbi.nlm.nih.gov/15725409/
Miller MG, Thangthaeng N, Rutledge GA, Scott TM, Shukitt-Hale B. Dietary strawberry improves cognition in a randomised, double-blind, placebo-controlled trial in older adults. Br J Nutr. Published online January 20, 2021:1–11.; https://pubmed.ncbi.nlm.nih.gov/33468271/
Gao Q, Qin LQ, Arafa A, Eshak ES, Dong JY. Effects of strawberry intervention on cardiovascular risk factors: a meta-analysis of randomised controlled trials. Br J Nutr. 2020;124(3):241–6. https://pubmed.ncbi.nlm.nih.gov/32238201/
Schell J, Scofield RH, Barrett JR, et al. Strawberries improve pain and inflammation in obese adults with radiographic evidence of knee osteoarthritis. Nutrients. 2017;9(9):949. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5622709/
Ezzat-Zadeh Z, Henning SM, Yang J, et al. California strawberry consumption increased the abundance of gut microorganisms related to lean body weight, health and longevity in healthy subjects. Nutr Res. 2021;85:60–70. https://pubmed.ncbi.nlm.nih.gov/33450667/
Morotomi M, Nagai F, Watanabe Y. Description of Christensenella minuta gen. nov., sp. nov., isolated from human faeces, which forms a distinct branch in the order Clostridiales, and proposal of Christensenellaceae fam. nov. Int J Syst Evol. 2012;62(1):144–9. https://pubmed.ncbi.nlm.nih.gov/21357455/
Waters JL, Ley RE. The human gut bacteria Christensenellaceae are widespread, heritable, and associated with health. BMC Biol. 2019;17(1):83. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6819567/
Wang Y, Chang J, Liu X, et al. Discovery of piperlongumine as a potential novel lead for the development of senolytic agents. Aging (Albany NY). 2016;8(11):2915–26. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5191878/
Yadav V, Krishnan A, Vohora D. A systematic review on Piper longum L.: bridging traditional knowledge and pharmacological evidence for future translational research. J Ethnopharmacol. 2020;247:112255. https://pubmed.ncbi.nlm.nih.gov/31568819/
Kumar S, Kamboj J, Suman, Sharma S. Overview for various aspects of the health benefits of Piper Longum Linn. fruit. J Acupunct Meridian Stud. 2011;4(2):134–40. https://pubmed.ncbi.nlm.nih.gov/21704957/
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–217. https://pubmed.ncbi.nlm.nih.gov/23746838/
van Deursen JM. Senolytic therapies for healthy longevity. Science. 2019;364(6441):636–7. https://pubmed.ncbi.nlm.nih.gov/31097655/
López-León M, Goya RG. The emerging view of aging as a reversible epigenetic process. Gerontology. 2017;63(5):426–31. https://pubmed.ncbi.nlm.nih.gov/28538216/
Sallon S, Solowey E, Cohen Y, et al. Germination, genetics, and growth of an ancient date seed. Science. 2008;320(5882):1464. https://pubmed.ncbi.nlm.nih.gov/18556553/
Yashina S, Gubin S, Maksimovich S, Yashina A, Gakhova E, Gilichinsky D. Regeneration of whole fertile plants from 30,000-y-old fruit tissue buried in Siberian permafrost. Proc Natl Acad Sci U S A. 2012;109(10):4008–13. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3309767/
Rando TA, Chang HY. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell. 2012;148(1–2):46–57. https://pubmed.ncbi.nlm.nih.gov/22265401/
Rando TA, Chang HY. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell. 2012;148(1–2):46–57. https://pubmed.ncbi.nlm.nih.gov/22265401/
Американская кантри-певица и киноактриса. – Примеч. ред.
BBC News. 1997: Dolly the sheep is cloned. On this day: 1950–2005. BBC. http://news.bbc.co.uk/onthisday/hi/dates/stories/february/22/newsid_4245000/4245877.stm. Published February 22, 2005. Accessed May 26, 2021.; https://news.bbc.co.uk/onthisday/hi/dates/stories/february/22/newsid_4245000/4245877.stm
Gurdon JB. The cloning of a frog. Development. 2013;140(12):2446–8. https://pubmed.ncbi.nlm.nih.gov/23715536/
Burgstaller JP, Brem G. Aging of cloned animals: a mini-review. Gerontology. 2017;63(5):417–25. https://pubmed.ncbi.nlm.nih.gov/27820924/
López-León M, Goya RG. The emerging view of aging as a reversible epigenetic process. Gerontology. 2017;63(5):426–31. https://pubmed.ncbi.nlm.nih.gov/28538216/
Song S, Johnson FB. Epigenetic mechanisms impacting aging: a focus on histone levels and telomeres. Genes. 2018;9(4):201. https://pubmed.ncbi.nlm.nih.gov/29642537/
Rando TA, Chang HY. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell. 2012;148(1–2):46–57. https://pubmed.ncbi.nlm.nih.gov/22265401/
Burgstaller JP, Brem G. Aging of cloned animals: a mini-review. Gerontology. 2017;63(5):417–25. https://pubmed.ncbi.nlm.nih.gov/27820924/
Wakayama S, Kohda T, Obokata H, et al. Successful serial recloning in the mouse over multiple generations. Cell Stem Cell. 2013;12(3):293–7. https://pubmed.ncbi.nlm.nih.gov/23472871/
López-León M, Goya RG. The emerging view of aging as a reversible epigenetic process. Gerontology. 2017;63(5):426–31. https://pubmed.ncbi.nlm.nih.gov/28538216/
Waddington CH. The epigenotype. 1942. Int J Epidemiol. 2012;41(1):10–13. https://pubmed.ncbi.nlm.nih.gov/22186258/
Watson JD, Crick FHC. Molecular structure of nucleic acids: a structure for deoxyribose nucleic acid. Nature. 1953;171(4356):737–8. https://pubmed.ncbi.nlm.nih.gov/13054692/
Song S, Johnson FB. Epigenetic mechanisms impacting aging: a focus on histone levels and telomeres. Genes. 2018;9(4):201. https://pubmed.ncbi.nlm.nih.gov/29642537/
Salzberg SL. Open questions: how many genes do we have? BMC Biol. 2018;16(1):94. https://pubmed.ncbi.nlm.nih.gov/30124169/
Govindaraju D, Atzmon G, Barzilai N. Genetics, lifestyle and longevity: lessons from centenarians. Appl Transl Genom. 2015;4:23–32. https://pubmed.ncbi.nlm.nih.gov/26937346/
vel Szic KS, Declerck K, Vidakovic M, Vanden Berghe W. From inflammaging to healthy aging by dietary lifestyle choices: is epigenetics the key to personalized nutrition? Clin Epigenet. 2015;7(1):33. https://pubmed.ncbi.nlm.nih.gov/25861393/
Li X, Yi C. A novel epigenetic mark derived from vitamin C. Biochemistry. 2020;59(1):8–9. https://pubmed.ncbi.nlm.nih.gov/31538774/
Ciccarone F, Tagliatesta S, Caiafa P, Zampieri M. DNA methylation dynamics in aging: how far are we from understanding the mechanisms? Mech Ageing Dev. 2018;174:3–17. https://pubmed.ncbi.nlm.nih.gov/29268958/
Mitteldorf J. How does the body know how old it is? Introducing the epigenetic clock hypothesis. In: Yashin AI, Jazwinski SM, eds. Aging and Health – A Systems Biology Perspective. Interdisciplinary Topics in Gerontology, vol 40. Karger, Basel;2015:49–62. https://pubmed.ncbi.nlm.nih.gov/25341512/
Ashapkin VV, Kutueva LI, Vanyushin BF. Epigenetic clock: just a convenient marker or an active driver of aging? In: Guest PC, ed. Reviews on Biomarker Studies in Aging and Anti-Aging Research. Advances in Experimental Medicine and Biology, vol 1178. Springer Cham; 2019:175–206. https://pubmed.ncbi.nlm.nih.gov/31493228/
Vaiserman AM. Hormesis and epigenetics: is there a link? Ageing Res Rev. 2011;10(4):413–21. https://pubmed.ncbi.nlm.nih.gov/21292042/
Kawahata A, Sakamoto H. Some observations on sweating of the Aino. Jpn J Physiol. 1951;2(2):166–9. https://pubmed.ncbi.nlm.nih.gov/14897491/
Painter RC, Osmond C, Gluckman P, Hanson M, Phillips DI, Roseboom TJ. Transgenerational effects of prenatal exposure to the Dutch famine on neonatal adiposity and health in later life. BJOG. 2008;115(10):1243–9. https://pubmed.ncbi.nlm.nih.gov/18715409/
Ornish D, Magbanua MJ, Weidner G, et al. Changes in prostate gene expression in men undergoing an intensive nutrition and lifestyle intervention. Proc Natl Acad Sci USA. 2008;105(24):8369–74. https://pubmed.ncbi.nlm.nih.gov/18559852/
Corona M, Velarde RA, Remolina S, et al. Vitellogenin, juvenile hormone, insulin signaling, and queen honey bee longevity. Proc Natl Acad Sci USA. 2007;104(17):7128–33. https://pubmed.ncbi.nlm.nih.gov/17438290/
Bacalini MG, Friso S, Olivieri F, et al. Present and future of anti-ageing epigenetic diets. Mech Ageing Dev. 2014;136–137:101–15. https://pubmed.ncbi.nlm.nih.gov/24388875/
Kucharski R, Maleszka J, Foret S, Maleszka R. Nutritional control of reproductive status in honeybees via DNA methylation. Science. 2008;319(5871):1827–30. https://pubmed.ncbi.nlm.nih.gov/18339900/
Hadi A, Najafgholizadeh A, Aydenlu ES, et al. Royal jelly is an effective and relatively safe alternative approach to blood lipid modulation: a meta-analysis. J Funct Foods. 2018;41:202–9. https://www.sciencedirect.com/science/article/abs/pii/S1756464617307284?via%3Dihub
Ecker S, Beck S. The epigenetic clock: a molecular crystal ball for human aging? Aging (Albany NY). 2019;11(2):833–5. https://pubmed.ncbi.nlm.nih.gov/30669120/
Ecker S, Beck S. The epigenetic clock: a molecular crystal ball for human aging? Aging (Albany NY). 2019;11(2):833–5. https://pubmed.ncbi.nlm.nih.gov/30669120/
Fransquet PD, Wrigglesworth J, Woods RL, Ernst ME, Ryan J. The epigenetic clock as a predictor of disease and mortality risk: a systematic review and meta-analysis. Clin Epigenet. 2019;11(1):62. https://pubmed.ncbi.nlm.nih.gov/30975202/
Venter JC, Adams MD, Myers EW, et al. The sequence of the human genome. Science. 2001;291(5507):1304–51. https://pubmed.ncbi.nlm.nih.gov/11181995/
Unnikrishnan A, Freeman WM, Jackson J, Wren JD, Porter H, Richardson A. The role of DNA methylation in epigenetics of aging. Pharmacol Ther. 2019;195:172–85. https://pubmed.ncbi.nlm.nih.gov/30419258/
Устройство, выполняющее очень простое действие чрезвычайно сложным образом. Как правило, это происходит посредством длинной последовательности взаимодействий по «принципу домино». – Примеч. ред.
Mendelson MM. Epigenetic age acceleration: a biological doomsday clock for cardiovascular disease? Circ Genom Precis Med. 2018;11(3). https://pubmed.ncbi.nlm.nih.gov/29555673/
Unnikrishnan A, Freeman WM, Jackson J, Wren JD, Porter H, Richardson A. The role of DNA methylation in epigenetics of aging. Pharmacol Ther. 2019;195:172–85. https://pubmed.ncbi.nlm.nih.gov/30419258/
Mitteldorf J. A clinical trial using methylation age to evaluate current antiaging practices. Rejuvenation Res. 2019;22(3):201–9. https://pubmed.ncbi.nlm.nih.gov/30345885/
Mendelson MM. Epigenetic age acceleration: a biological doomsday clock for cardiovascular disease? Circ Genom Precis Med. 2018;11(3). https://pubmed.ncbi.nlm.nih.gov/29555673/
Social Security Administration. Actuarial life table. Period life table, 2017. Social Security Administration. https://www.ssa.gov/oact/STATS/table4c6.html. Accessed May 26, 2021.; https://www.ssa.gov/oact/STATS/table4c6.html
McCrory C, Fiorito G, Hernandez B, et al. GrimAge outperforms other epigenetic clocks in the prediction of age-related clinical phenotypes and all-cause mortality. J Gerontol A Biol Sci Med Sci. 2021;76(5):741–9. https://pubmed.ncbi.nlm.nih.gov/33211845/
Mitteldorf J. A clinical trial using methylation age to evaluate current antiaging practices. Rejuvenation Res. 2019;22(3):201–9. https://pubmed.ncbi.nlm.nih.gov/30345885/
Mendelson MM. Epigenetic age acceleration: a biological doomsday clock for cardiovascular disease? Circ Genom Precis Med. 2018;11(3). https://pubmed.ncbi.nlm.nih.gov/29555673/
Mitteldorf J. An incipient revolution in the testing of anti-aging strategies. Biochemistry (Mosc). 2018;83(12):1517–23. https://pubmed.ncbi.nlm.nih.gov/30878026/
Horvath S, Pirazzini C, Bacalini MG, et al. Decreased epigenetic age of PBMCs from Italian semi-supercentenarians and their offspring. Aging (Albany NY). 2015;7(12):1159–70. https://pubmed.ncbi.nlm.nih.gov/26678252/
Declerck K, Vanden Berghe W. Back to the future: epigenetic clock plasticity towards healthy aging. Mech Ageing Dev. 2018;174:18–29. https://pubmed.ncbi.nlm.nih.gov/29337038/
Austad SN, Bartke A. Sex differences in longevity and in responses to anti-aging interventions: a mini-review. Gerontology. 2015;62(1):40–6. https://pubmed.ncbi.nlm.nih.gov/25968226/
Robert L, Fulop T. Longevity and its regulation: centenarians and beyond. Interdiscip Top Gerontol. 2014;39:198–211. https://pubmed.ncbi.nlm.nih.gov/24862022/
Beach SRH, Dogan MV, Lei MK, et al. Methylomic aging as a window onto the influence of lifestyle: tobacco and alcohol use alter the rate of biological aging. J Am Geriatr Soc. 2015;63(12):2519–25. https://pubmed.ncbi.nlm.nih.gov/26566992/
Vyas CM, Hazra A, Chang SC, et al. Pilot study of DNA methylation, molecular aging markers and measures of health and well-being in aging. Transl Psychiatry. 2019;9(1):118. https://pubmed.ncbi.nlm.nih.gov/30886137/
Pavanello S, Campisi M, Tona F, Dal Lin C, Iliceto S. Exploring epigenetic age in response to intensive relaxing training: a pilot study to slow down biological age. Int J Environ Res Public Health. 2019;16(17):3074. https://pubmed.ncbi.nlm.nih.gov/31450859/
Chaix R, Alvarez-López MJ, Fagny M, et al. Epigenetic clock analysis in long-term meditators. Psychoneuroendocrinology. 2017;85:210–4. https://pubmed.ncbi.nlm.nih.gov/28889075/
Maegawa S, Lu Y, Tahara T, et al. Caloric restriction delays age-related methylation drift. Nat Commun. 2017;8(1):539. https://pubmed.ncbi.nlm.nih.gov/28912502/
Belsky DW, Huffman KM, Pieper CF, Shalev I, Kraus WE. Change in the rate of biological aging in response to caloric restriction: CALERIE Biobank analysis. J Gerontol A Biol Sci Med Sci. 2018;73(1):4–10. https://pubmed.ncbi.nlm.nih.gov/28531269/
Belsky DW, Huffman KM, Pieper CF, Shalev I, Kraus WE. Change in the rate of biological aging in response to caloric restriction: CALERIE Biobank analysis. J Gerontol A Biol Sci Med Sci. 2018;73(1):4–10. https://pubmed.ncbi.nlm.nih.gov/28531269/
Horvath S, Erhart W, Brosch M, et al. Obesity accelerates epigenetic aging of human liver. Proc Natl Acad Sci USA. 2014;111(43):15538–43. https://pubmed.ncbi.nlm.nih.gov/25313081/
de Toro-Martín J, Guénard F, Tchernof A, et al. Body mass index is associated with epigenetic age acceleration in the visceral adipose tissue of subjects with severe obesity. Clin Epigenetics. 2019;11(1):172. https://pubmed.ncbi.nlm.nih.gov/31791395/
Horvath S, Erhart W, Brosch M, et al. Obesity accelerates epigenetic aging of human liver. Proc Natl Acad Sci USA. 2014;111(43):15538–43. https://pubmed.ncbi.nlm.nih.gov/25313081/
Lu AT, Quach A, Wilson JG, et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY). 2019;11(2):303–27. https://pubmed.ncbi.nlm.nih.gov/30669119/
Quach A, Levine ME, Tanaka T, et al. Epigenetic clock analysis of diet, exercise, education, and lifestyle factors. Aging (Albany NY). 2017;9(2):419–37. https://pubmed.ncbi.nlm.nih.gov/28198702/
Hardy TM, Tollefsbol TO. Epigenetic diet: impact on the epigenome and cancer. Epigenomics. 2011;3(4):503–18. https://pubmed.ncbi.nlm.nih.gov/22022340/
Levine ME, Lu AT, Quach A, et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY). 2018;10(4):573–91. https://pubmed.ncbi.nlm.nih.gov/29676998/
Dugué PA, Bassett JK, Joo JE, et al. Association of DNA methylation-based biological age with health risk factors and overall and cause-specific mortality. Am J Epidemiol. 2018;187(3):529–38. https://pubmed.ncbi.nlm.nih.gov/29020168/
Lind PM, Salihovic S, Lind L. High plasma organochlorine pesticide levels are related to increased biological age as calculated by DNA methylation analysis. Environ Int. 2018;113:109–13. https://pubmed.ncbi.nlm.nih.gov/29421399/
Mariscal-Arcas M, Lopez-Martinez C, Granada A, Olea N, Lorenzo-Tovar ML, Olea-Serrano F. Organochlorine pesticides in umbilical cord blood serum of women from Southern Spain and adherence to the Mediterranean diet. Food Chem Toxicol. 2010;48(5):1311–5. https://pubmed.ncbi.nlm.nih.gov/20188779/
Ward-Caviness CK, Nwanaji-Enwerem JC, Wolf K, et al. Long-term exposure to air pollution is associated with biological aging. Oncotarget. 2016;7(46):74510–25. https://pubmed.ncbi.nlm.nih.gov/27793020/
Ryan J, Wrigglesworth J, Loong J, Fransquet PD, Woods RL. A systematic review and meta-analysis of environmental, lifestyle, and health factors associated with DNA methylation age. J Gerontol A Biol Sci Med Sci. 2020;75(3):481–94. https://pubmed.ncbi.nlm.nih.gov/31001624/
Mitteldorf J. A clinical trial using methylation age to evaluate current antiaging practices. Rejuvenation Res. 2019;22(3):201–9. https://pubmed.ncbi.nlm.nih.gov/30345885/
Fransquet PD, Wrigglesworth J, Woods RL, Ernst ME, Ryan J. The epigenetic clock as a predictor of disease and mortality risk: a systematic review and meta-analysis. Clin Epigenet. 2019;11(1):62. https://pubmed.ncbi.nlm.nih.gov/30975202/
Ashapkin VV, Kutueva LI, Vanyushin BF. Epigenetic clock: just a convenient marker or an active driver of aging? In: Guest PC, ed. Reviews on Biomarker Studies in Aging and Anti-Aging Research. Advances in Experimental Medicine and Biology, vol 1178. Springer Cham; 2019:175–206. https://pubmed.ncbi.nlm.nih.gov/31493228/
Nobel Media AB 2021. Shinya Yamanaka – Facts. NobelPrize.org. https://www.nobelprize.org/prizes/medicine/2012/yamanaka/facts/. Accessed June 5, 2021.; https://www.nobelprize.org/prizes/medicine/2012/yamanaka/facts/
Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76. https://pubmed.ncbi.nlm.nih.gov/16904174/
Shieh SJ, Cheng TC. Regeneration and repair of human digits and limbs: fact and fiction. Regeneration. 2015;2(4):149–68. https://pubmed.ncbi.nlm.nih.gov/27499873/
Lu Y, Brommer B, Tian X, et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature. 2020;588(7836):124–9. https://pubmed.ncbi.nlm.nih.gov/33268865/
Jacobsen SC, Brøns C, Bork-Jensen J, et al. Effects of short-term high-fat overfeeding on genome-wide DNA methylation in the skeletal muscle of healthy young men. Diabetologia. 2012;55(12):3341–9. https://pubmed.ncbi.nlm.nih.gov/22961225/
Perfilyev A, Dahlman I, Gillberg L, et al. Impact of polyunsaturated and saturated fat overfeeding on the DNA-methylation pattern in human adipose tissue: a randomized controlled trial. Am J Clin Nutr. 2017;105(4):991–1000. https://pubmed.ncbi.nlm.nih.gov/28275132/
Miles FL, Mashchak A, Filippov V, et al. DNA methylation profiles of vegans and non-vegetarians in the Adventist Health Study-2 cohort. Nutrients. 2020;12(12):3697. https://pubmed.ncbi.nlm.nih.gov/33266012/
Key TJ, Appleby PN, Crowe FL, Bradbury KE, Schmidt JA, Travis RC. Cancer in British vegetarians: updated analyses of 4998 incident cancers in a cohort of 32,491 meat eaters, 8612 fish eaters, 18,298 vegetarians, and 2246 vegans. Am J Clin Nutr. 2014;100 Suppl 1:378S-85S. https://pubmed.ncbi.nlm.nih.gov/24898235/
Tantamango-Bartley Y, Jaceldo-Siegl K, Fan J, Fraser G. Vegetarian diets and the incidence of cancer in a low-risk population. Cancer Epidemiol Biomarkers Prev. 2013;22(2):286–94. https://pubmed.ncbi.nlm.nih.gov/23169929/
McCord JM. Analysis of superoxide dismutase activity. Curr Protoc Toxicol. 2001;Chapter 7:Unit7.3. https://pubmed.ncbi.nlm.nih.gov/23045062/
Thaler R, Karlic H, Rust P, Haslberger AG. Epigenetic regulation of human buccal mucosa mitochondrial superoxide dismutase gene expression by diet. Br J Nutr. 2009;101(5):743–9. https://pubmed.ncbi.nlm.nih.gov/18684339/
Johnson AA, Akman K, Calimport SRG, Wuttke D, Stolzing A, de Magalhães JP. The role of DNA methylation in aging, rejuvenation, and age-related disease. Rejuvenation Res. 2012;15(5):483–94. https://pubmed.ncbi.nlm.nih.gov/23098078/
ElGendy K, Malcomson FC, Lara JG, Bradburn DM, Mathers JC. Effects of dietary interventions on DNA methylation in adult humans: systematic review and meta-analysis. Br J Nutr. 2018;120(9):961–76. https://pubmed.ncbi.nlm.nih.gov/30355391/
Miller JW. Factors associated with different forms of folate in human serum: the folate folio continues to grow. J Nutr. 2020;150(4):650–1. https://pubmed.ncbi.nlm.nih.gov/32119743/
Institute of Medicine (US) Standing Committee on the Scientific Evaluation of Dietary Reference Intakes and Its Panel on Folate, Other B Vitamins, and Choline. Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline. National Academies Press (US); 1998. https://pubmed.ncbi.nlm.nih.gov/23193625/
ter Borg S, Verlaan S, Hemsworth J, et al. Micronutrient intakes and potential inadequacies of community-dwelling older adults: a systematic review. Br J Nutr. 2015;113(8):1195–206. https://pubmed.ncbi.nlm.nih.gov/25822905/
Jacob RA, Gretz DM, Taylor PC, et al. Moderate folate depletion increases plasma homocysteine and decreases lymphocyte DNA methylation in postmenopausal women. J Nutr. 1998;128(7):1204–12. https://pubmed.ncbi.nlm.nih.gov/9649607/
Rampersaud GC, Kauwell GP, Hutson AD, Cerda JJ, Bailey LB. Genomic DNA methylation decreases in response to moderate folate depletion in elderly women. Am J Clin Nutr. 2000;72(4):998–1003. https://pubmed.ncbi.nlm.nih.gov/11010943/
Amenyah SD, Hughes CF, Ward M, et al. Influence of nutrients involved in one-carbon metabolism on DNA methylation in adults – a systematic review and meta-analysis. Nutr Rev. 2020;78(8):647–66. https://pubmed.ncbi.nlm.nih.gov/31977026/
Rampersaud GC, Kauwell GP, Hutson AD, Cerda JJ, Bailey LB. Genomic DNA methylation decreases in response to moderate folate depletion in elderly women. Am J Clin Nutr. 2000;72(4):998–1003. https://pubmed.ncbi.nlm.nih.gov/11010943/
Mathers JC, Strathdee G, Relton CL. Induction of epigenetic alterations by dietary and other environmental factors. Adv Genet. 2010;71:3–39. https://pubmed.ncbi.nlm.nih.gov/20933124/
Eaton SB, Eaton SB. Paleolithic vs. modern diets – selected pathophysiological implications. Eur J Nutr. 2000;39(2):67–70. https://pubmed.ncbi.nlm.nih.gov/10918987/
Метилентетрагидрофолатредуктаза, ключевой фермент фолатного цикла. – Примеч. ред.
Parkhurst E, Calonico E, Noh G. Medical decision support to reduce unwarranted methylene tetrahydrofolate reductase (MTHFR) genetic testing. J Med Syst. 2020;44(9):152. https://pubmed.ncbi.nlm.nih.gov/32737598/
Levin BL, Varga E. MTHFR: addressing genetic counseling dilemmas using evidence-based literature. J Genet Couns. 2016;25(5):901–11. https://pubmed.ncbi.nlm.nih.gov/27130656/
Porter K, Hoey L, Hughes CF, Ward M, McNulty H. Causes, consequences and public health implications of low B-vitamin status in ageing. Nutrients. 2016;8(11). https://pubmed.ncbi.nlm.nih.gov/27854316/
Friso S, Choi SW, Girelli D, et al. A common mutation in the 5,10-methylenetetrahydrofolate reductase gene affects genomic DNA methylation through an interaction with folate status. Proc Natl Acad Sci USA. 2002;99(8):5606–11. https://pubmed.ncbi.nlm.nih.gov/11929966/
Bailey LB. Folate, methyl-related nutrients, alcohol, and the MTHFR 677C®T polymorphism affect cancer risk: intake recommendations. J Nutr. 2003;133(11 Suppl 1):3748S-53S. https://pubmed.ncbi.nlm.nih.gov/14608109/
Levin BL, Varga E. MTHFR: addressing genetic counseling dilemmas using evidence-based literature. J Genet Couns. 2016;25(5):901–11. https://pubmed.ncbi.nlm.nih.gov/27130656/
Parkhurst E, Calonico E, Noh G. Medical decision support to reduce unwarranted methylene tetrahydrofolate reductase (MTHFR) genetic testing. J Med Syst. 2020;44(9):152. https://pubmed.ncbi.nlm.nih.gov/32737598/
Seitz HK, Matsuzaki S, Yokoyama A, Homann N, Väkeväinen S, Wang XD. Alcohol and cancer. Alcohol Clin Exp Res. 2001;25(5 Suppl ISBRA):137S-43S. https://pubmed.ncbi.nlm.nih.gov/15082451/
Bailey LB. Folate, methyl-related nutrients, alcohol, and the MTHFR 677C®T polymorphism affect cancer risk: intake recommendations. J Nutr. 2003;133(11 Suppl 1):3748S-53S. https://pubmed.ncbi.nlm.nih.gov/14608109/
Griswold MG, Fullman N, Hawley C, et al. Alcohol use and burden for 195 countries and territories, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2018;392(10152):1015–35. https://pubmed.ncbi.nlm.nih.gov/30146330/
Bo Y, Zhu Y, Tao Y, et al. Association between folate and health outcomes: an umbrella review of meta-analyses. Front Public Health. 2020;8:550753. https://pubmed.ncbi.nlm.nih.gov/33384976/
Bo Y, Zhu Y, Tao Y, et al. Association between folate and health outcomes: an umbrella review of meta-analyses. Front Public Health. 2020;8:550753. https://pubmed.ncbi.nlm.nih.gov/33384976/
Crider KS, Bailey LB, Berry RJ. Folic acid food fortification – its history, effect, concerns, and future directions. Nutrients. 2011;3(3):370–84. https://pubmed.ncbi.nlm.nih.gov/22254102/
Bailey SW, Ayling JE. The extremely slow and variable activity of dihydrofolate reductase in human liver and its implications for high folic acid intake. Proc Natl Acad Sci U S A. 2009;106(36):15424–9. https://pubmed.ncbi.nlm.nih.gov/19706381/
Selhub J, Rosenberg IH. Excessive folic acid intake and relation to adverse health outcome. Biochimie. 2016;126:71–8. https://pubmed.ncbi.nlm.nih.gov/27131640/
Troen AM, Mitchell B, Sorensen B, et al. Unmetabolized folic acid in plasma is associated with reduced natural killer cell cytotoxicity among postmenopausal women. J Nutr. 2006;136(1):189–94. https://pubmed.ncbi.nlm.nih.gov/16365081/
Bo Y, Zhu Y, Tao Y, et al. Association between folate and health outcomes: an umbrella review of meta-analyses. Front Public Health. 2020;8:550753. https://pubmed.ncbi.nlm.nih.gov/33384976/
U.S. Preventive Services Task Force. Final recommendation statement: folic acid for the prevention of neural tube defects: preventive medication. U.S. Preventive Services Task Force. https://www.uspreventiveservicestaskforce.org/uspstf/recommendation/folic-acid-for-the-prevention-of-neural-tube-defects-preventive-medication. Published January 10, 2017. Accessed May 26, 2021.; https://www.uspreventiveservicestaskforce.org/uspstf/recommendation/folic-acid-for-the-prevention-of-neural-tube-defects-preventive-medication
Dudeja PK, Torania SA, Said HM. Evidence for the existence of a carrier-mediated folate uptake mechanism in human colonic luminal membranes. Am J Physiol. 1997;272(6Pt1):G1408–15. https://pubmed.ncbi.nlm.nih.gov/9227476/
Strozzi GP, Mogna L. Quantification of folic acid in human feces after administration of Bifidobacterium probiotic strains. J Clin Gastroenterol. 2008;42 Suppl 3 Pt 2:S179–84. https://pubmed.ncbi.nlm.nih.gov/18685499/
Rando TA, Chang HY. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell. 2012;148(1–2):46–57. https://pubmed.ncbi.nlm.nih.gov/22265401/
Hellwig M, Henle T. Baking, ageing, diabetes: a short history of the Maillard reaction. Angew Chem Int Ed. 2014;53(39):10316–29. https://pubmed.ncbi.nlm.nih.gov/25044982/
Teodorowicz M, Hendriks WH, Wichers HJ, Savelkoul HFJ. Immunomodulation by processed animal feed: the role of Maillard reaction products and advanced glycation end-products (AGEs). Front Immunol. 2018;9:2088. https://pubmed.ncbi.nlm.nih.gov/30271411/
Sadowska-Bartosz I, Bartosz G. Effect of glycation inhibitors on aging and age-related diseases. Mech Ageing Dev. 2016;160:1–18. https://pubmed.ncbi.nlm.nih.gov/27671971/
Unnikrishnan R, Anjana RM, Mohan V. Drugs affecting HbA1c levels. Indian J Endocrinol Metab. 2012;16(4):528–31. https://pubmed.ncbi.nlm.nih.gov/22837911/
American Diabetes Association. Understanding A1C. American Diabetes Association website. https://www.diabetes.org/a1c. Accessed June 2, 2021.; https://www.diabetes.org/a1c
Sadowska-Bartosz I, Bartosz G. Effect of glycation inhibitors on aging and age-related diseases. Mech Ageing Dev. 2016;160:1–18. https://pubmed.ncbi.nlm.nih.gov/27671971/
Verzijl N, DeGroot J, Thorpe SR, et al. Effect of collagen turnover on the accumulation of advanced glycation end products. J Biol Chem. 2000;275(50):39027–31. https://pubmed.ncbi.nlm.nih.gov/10976109/
Fedintsev A, Moskalev A. Stochastic non-enzymatic modification of long-lived macromolecules – a missing hallmark of aging. Ageing Res Rev. 2020;62:101097. https://pubmed.ncbi.nlm.nih.gov/32540391/
Green AS. mTOR, glycotoxins and the parallel universe. Aging (Albany NY). 2018;10(12):3654–6. https://pubmed.ncbi.nlm.nih.gov/30540565/
Bettiga A, Fiorio F, Di Marco F, et al. The modern Western diet rich in advanced glycation end-products (AGES): an overview of its impact on obesity and early progression of renal pathology. Nutrients. 2019;11(8):1748. https://pubmed.ncbi.nlm.nih.gov/31366015/
Garay-Sevilla ME, Beeri MS, de la Maza MP, Rojas A, Salazar-Villanea S, Uribarri J. The potential role of dietary advanced glycation endproducts in the development of chronic non-infectious diseases: a narrative review. Nutr Res Rev. 2020;33(2):298–311. https://pubmed.ncbi.nlm.nih.gov/32238213/
Chen JH, Lin X, Bu C, Zhang X. Role of advanced glycation end products in mobility and considerations in possible dietary and nutritional intervention strategies. Nutr Metab (Lond). 2018;15(1):72. https://pubmed.ncbi.nlm.nih.gov/30337945/
Prasad C, Davis KE, Imrhan V, Juma S, Vijayagopal P. Advanced glycation end products and risks for chronic diseases: intervening through lifestyle modification. Am J Lifestyle Med. 2019;13(4):384–404. https://pubmed.ncbi.nlm.nih.gov/31285723/
Semba RD, Nicklett EJ, Ferrucci L. Does accumulation of advanced glycation end products contribute to the aging phenotype? J Gerontol A Biol Sci Med Sci. 2010;65A(9):963–75. https://pubmed.ncbi.nlm.nih.gov/20478906/
Green AS. mTOR, glycotoxins and the parallel universe. Aging (Albany NY). 2018;10(12):3654–6. https://pubmed.ncbi.nlm.nih.gov/30540565/
Sergi D, Boulestin H, Campbell FM, Williams LM. The role of dietary advanced glycation end products in metabolic dysfunction. Mol Nutr Food Res. 2021;65(1):1900934. https://pubmed.ncbi.nlm.nih.gov/32246887/
Sadowska-Bartosz I, Bartosz G. Effect of glycation inhibitors on aging and age-related diseases. Mech Ageing Dev. 2016;160:1–18. https://pubmed.ncbi.nlm.nih.gov/27671971/
. Šebeková K, Brouder Šebeková K. Glycated proteins in nutrition: friend or foe? Exp Gerontol. 2019;117:76–90. https://pubmed.ncbi.nlm.nih.gov/30458224/
Fedintsev A, Moskalev A. Stochastic non-enzymatic modification of long-lived macromolecules – a missing hallmark of aging. Ageing Res Rev. 2020;62:101097. https://pubmed.ncbi.nlm.nih.gov/32540391/
Azman KF, Zakaria R. D-galactose-induced accelerated aging model: an overview. Biogerontology. 2019;20(6):763–82. https://pubmed.ncbi.nlm.nih.gov/31538262/
Sadowska-Bartosz I, Bartosz G. Effect of glycation inhibitors on aging and age-related diseases. Mech Ageing Dev. 2016;160:1–18. https://pubmed.ncbi.nlm.nih.gov/27671971/
Fedintsev A, Moskalev A. Stochastic non-enzymatic modification of long-lived macromolecules – a missing hallmark of aging. Ageing Res Rev. 2020;62:101097. https://pubmed.ncbi.nlm.nih.gov/32540391/
Teissier T, Boulanger É. The receptor for advanced glycation end-products (RAGE) is an important pattern recognition receptor (PRR) for inflammaging. Biogerontology. 2019;20(3):279–301. https://pubmed.ncbi.nlm.nih.gov/30968282/
Green AS. mTOR, glycotoxins and the parallel universe. Aging (Albany NY). 2018;10(12):3654–6. https://pubmed.ncbi.nlm.nih.gov/30540565/
Teissier T, Boulanger É. The receptor for advanced glycation end-products (RAGE) is an important pattern recognition receptor (PRR) for inflammaging. Biogerontology. 2019;20(3):279–301. https://pubmed.ncbi.nlm.nih.gov/30968282/
Gill V, Kumar V, Singh K, Kumar A, Kim JJ. Advanced glycation end products (AGEs) may be a striking link between modern diet and health. Biomolecules. 2019;9(12):888. https://pubmed.ncbi.nlm.nih.gov/31861217/
Hellwig M, Henle T. Baking, ageing, diabetes: a short history of the Maillard reaction. Angew Chem Int Ed. 2014;53(39):10316–29. https://pubmed.ncbi.nlm.nih.gov/25044982/
Bettiga A, Fiorio F, Di Marco F, et al. The modern Western diet rich in advanced glycation end-products (AGES): an overview of its impact on obesity and early progression of renal pathology. Nutrients. 2019;11(8):1748. https://pubmed.ncbi.nlm.nih.gov/31366015/
Chen JH, Lin X, Bu C, Zhang X. Role of advanced glycation end products in mobility and considerations in possible dietary and nutritional intervention strategies. Nutr Metab (Lond). 2018;15(1):72. https://pubmed.ncbi.nlm.nih.gov/30337945/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Sgarbieri VC, Amaya J, Tanaka M, Chichester CO. Response of rats to amino acid supplementation of brown egg albumin. J Nutr. 1973;103(12):1731–8. https://pubmed.ncbi.nlm.nih.gov/4201784/
Koschinsky T, He CJ, Mitsuhashi T, et al. Orally absorbed reactive glycation products (glycotoxins): an environmental risk factor in diabetic nephropathy. Proc Natl Acad Sci USA. 1997;94(12):6474–9. https://pubmed.ncbi.nlm.nih.gov/9177242/
Gill V, Kumar V, Singh K, Kumar A, Kim JJ. Advanced glycation end products (AGEs) may be a striking link between modern diet and health. Biomolecules. 2019;9(12):888. https://pubmed.ncbi.nlm.nih.gov/31861217/
Zhang Q, Wang Y, Fu L. Dietary advanced glycation end-products: perspectives linking food processing with health implications. Compr Rev Food Sci Food Saf. 2020;19(5):2559–87. https://pubmed.ncbi.nlm.nih.gov/33336972/
Koschinsky T, He CJ, Mitsuhashi T, et al. Orally absorbed reactive glycation products (glycotoxins): an environmental risk factor in diabetic nephropathy. Proc Natl Acad Sci USA. 1997;94(12):6474–9. https://pubmed.ncbi.nlm.nih.gov/9177242/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Babtan AM, Ilea A, Bosca BA, et al. Advanced glycation end products as biomarkers in systemic diseases: premises and perspectives of salivary advanced glycation end products. Biomark Med. 2019;13(6):479–95. https://pubmed.ncbi.nlm.nih.gov/30968701/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Goldberg T, Cai W, Peppa M, et al. Advanced glycoxidation end products in commonly consumed foods. J Am Diet Assoc. 2004;104(8):1287–91. https://pubmed.ncbi.nlm.nih.gov/15281050/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Clarivate. Web of science. https://clarivate.com/webofsciencegroup/solutions/web-of-science/. Accessed June 5, 2021.; https://clarivate.com/webofsciencegroup/solutions/web-of-science/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Bettiga A, Fiorio F, Di Marco F, et al. The modern Western diet rich in advanced glycation end-products (AGES): an overview of its impact on obesity and early progression of renal pathology. Nutrients. 2019;11(8):1748. https://pubmed.ncbi.nlm.nih.gov/31366015/
Cai W, Uribarri J, Zhu L, et al. Oral glycotoxins are a modifiable cause of dementia and the metabolic syndrome in mice and humans. Proc Natl Acad Sci USA. 2014;111(13):4940–5. https://pubmed.ncbi.nlm.nih.gov/24567379/
Hellwig M, Gensberger-Reigl S, Henle T, Pischetsrieder M. Food-derived 1,2-dicarbonyl compounds and their role in diseases. Semin Cancer Biol. 2018;49:1–8. https://pubmed.ncbi.nlm.nih.gov/29174601/
Gómez-Ojeda A, Jaramillo-Ortíz S, Wrobel K, et al. Comparative evaluation of three different ELISA assays and HPLC-ESI–ITMS/MS for the analysis of Ne-carboxymethyl lysine in food samples. Food Chem. 2018;243:11–8. https://pubmed.ncbi.nlm.nih.gov/29146316/
Zhang Q, Wang Y, Fu L. Dietary advanced glycation end-products: perspectives linking food processing with health implications. Compr Rev Food Sci Food Saf. 2020;19(5):2559–87. https://pubmed.ncbi.nlm.nih.gov/33336972/
Kuzan A. Toxicity of advanced glycation end products (Review). Biomed Rep. 2021;14(5):46. https://pubmed.ncbi.nlm.nih.gov/33786175/
Morales FJ, Somoza V, Fogliano V. Physiological relevance of dietary melanoidins. Amino Acids. 2012;42(4):1097–109. https://pubmed.ncbi.nlm.nih.gov/20949365/
Ottum MS, Mistry AM. Advanced glycation end-products: modifiable environmental factors profoundly mediate insulin resistance. J Clin Biochem Nutr. 2015;57(1):1–12. https://pubmed.ncbi.nlm.nih.gov/26236094/
Cai W, Gao Q, Zhu L, Peppa M, He C, Vlassara H. Oxidative stress-inducing carbonyl compounds from common foods: novel mediators of cellular dysfunction. Mol Med. 2002;8(7):337–46. https://pubmed.ncbi.nlm.nih.gov/12393931/
Nicholl ID, Bucala R. Advanced glycation endproducts and cigarette smoking. Cell Mol Biol (Noisy-le-grand). 1998;44(7):1025–33. https://pubmed.ncbi.nlm.nih.gov/9846884/
Garay-Sevilla ME, Beeri MS, de la Maza MP, Rojas A, Salazar-Villanea S, Uribarri J. The potential role of dietary advanced glycation endproducts in the development of chronic non-infectious diseases: a narrative review. Nutr Res Rev. 2020;33(2):298–311. https://pubmed.ncbi.nlm.nih.gov/32238213/
Rungratanawanich W, Qu Y, Wang X, Essa MM, Song BJ. Advanced glycation end products (AGEs) and other adducts in aging-related diseases and alcohol-mediated tissue injury. Exp Mol Med. 2021;53(2):168–88. https://pubmed.ncbi.nlm.nih.gov/33568752/
Garay-Sevilla ME, Beeri MS, de la Maza MP, Rojas A, Salazar-Villanea S, Uribarri J. The potential role of dietary advanced glycation endproducts in the development of chronic non-infectious diseases: a narrative review. Nutr Res Rev. 2020;33(2):298–311. https://pubmed.ncbi.nlm.nih.gov/32238213/
Goldberg T, Cai W, Peppa M, et al. Advanced glycoxidation end products in commonly consumed foods. J Am Diet Assoc. 2004;104(8):1287–91. https://pubmed.ncbi.nlm.nih.gov/15281050/
del Castillo MD, Iriondo-DeHond A, Iriondo-DeHond M, et al. Healthy eating recommendations: good for reducing dietary contribution to the body’s advanced glycation/lipoxidation end products pool? Nutr Res Rev. 2021;34(1):48–63. https://pubmed.ncbi.nlm.nih.gov/32450931/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Rungratanawanich W, Qu Y, Wang X, Essa MM, Song BJ. Advanced glycation end products (AGEs) and other adducts in aging-related diseases and alcohol-mediated tissue injury. Exp Mol Med. 2021;53(2):168–88. https://pubmed.ncbi.nlm.nih.gov/33568752/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Davis KE, Prasad C, Vijayagopal P, Juma S, Adams-Huet B, Imrhan V. Contribution of dietary advanced glycation end products (AGE) to circulating AGE: role of dietary fat. Br J Nutr. 2015;114(11):1797–806. https://pubmed.ncbi.nlm.nih.gov/26392152/
Semba RD, Nicklett EJ, Ferrucci L. Does accumulation of advanced glycation end products contribute to the aging phenotype? J Gerontol A Biol Sci Med Sci. 2010;65A(9):963–75. https://pubmed.ncbi.nlm.nih.gov/20478906/
Senolt L, Braun M, Olejarova M, Forejtova S, Gatterova J, Pavelka K. Increased pentosidine, an advanced glycation end product, in serum and synovial fluid from patients with knee osteoarthritis and its relation with cartilage oligomeric matrix protein. Ann Rheum Dis. 2005;64(6):886–90. https://pubmed.ncbi.nlm.nih.gov/15897309/
Hein G, Wiegand R, Lehmann G, Stein G, Franke S. Advanced glycation end-products pentosidine and N epsilon-carboxymethyllysine are elevated in serum of patients with osteoporosis. Rheumatology (Oxford). 2003;42(10):1242–6. https://pubmed.ncbi.nlm.nih.gov/12777635/
Meerwaldt R, Graaff R, Oomen PHN, et al. Simple non-invasive assessment of advanced glycation endproduct accumulation. Diabetologia. 2004;47(7):1324–30. https://pubmed.ncbi.nlm.nih.gov/15243705/
Mahmoudi R, Jaisson S, Badr S, et al. Post-translational modification-derived products are associated with frailty status in elderly subjects. Clin Chem Lab Med. 2019;57(8):1153–61. https://pubmed.ncbi.nlm.nih.gov/30817296/
Cavero-Redondo I, Soriano-Cano A, Álvarez-Bueno C, et al. Skin autofluorescence – indicated advanced glycation end products as predictors of cardiovascular and all-cause mortality in high-risk subjects: a systematic review and meta-analysis. J Am Heart Assoc. 2018;7(18):e009833. https://pubmed.ncbi.nlm.nih.gov/30371199/
Igase M, Ohara M, Igase K, et al. Skin autofluorescence examination as a diagnostic tool for mild cognitive impairment in healthy people. J Alzheimers Dis. 2017;55(4):1481–7. https://pubmed.ncbi.nlm.nih.gov/27858716/
Cai W, Uribarri J, Zhu L, et al. Oral glycotoxins are a modifiable cause of dementia and the metabolic syndrome in mice and humans. Proc Natl Acad Sci U S A. 2014;111(13):4940–5. https://pubmed.ncbi.nlm.nih.gov/24567379/
Giem P, Beeson WL, Fraser GE. The incidence of dementia and intake of animal products: preliminary findings from the Adventist Health Study. Neuroepidemiology. 1993;12(1):28–36. https://pubmed.ncbi.nlm.nih.gov/8327020/
Cao GY, Li M, Han L, et al. Dietary fat intake and cognitive function among older populations: a systematic review and meta-analysis. J Prev Alzheimers Dis. 2019;6(3):204–11. https://pubmed.ncbi.nlm.nih.gov/31062836/
Holloway CJ, Cochlin LE, Emmanuel Y, et al. A high-fat diet impairs cardiac high-energy phosphate metabolism and cognitive function in healthy human subjects. Am J Clin Nutr. 2011;93(4):748–55. https://pubmed.ncbi.nlm.nih.gov/21270386/
Cai W, He JC, Zhu L, et al. Reduced oxidant stress and extended lifespan in mice exposed to a low glycotoxin diet: association with increased AGER1 expression. Am J Pathol. 2007;170(6):1893–902. https://pubmed.ncbi.nlm.nih.gov/17525257/
Akhter F, Chen D, Akhter A, et al. High dietary advanced glycation end products impair mitochondrial and cognitive function. J Alzheimers Dis. 2020;76(1):165–78. https://pubmed.ncbi.nlm.nih.gov/32444539/
Peppa M, He C, Hattori M, McEvoy R, Zheng F, Vlassara H. Fetal or neonatal low-glycotoxin environment prevents autoimmune diabetes in NOD mice. Diabetes. 2003;52(6):1441–8. https://pubmed.ncbi.nlm.nih.gov/12765955/
Tsakiri EN, Iliaki KK, Höhn A, et al. Diet-derived advanced glycation end products or lipofuscin disrupts proteostasis and reduces life span in Drosophila melanogaster. Free Radic Biol Med. 2013;65:1155–63. https://pubmed.ncbi.nlm.nih.gov/23999505/
Peppa M, He C, Hattori M, McEvoy R, Zheng F, Vlassara H. Fetal or neonatal low-glycotoxin environment prevents autoimmune diabetes in NOD mice. Diabetes. 2003;52(6):1441–8. https://pubmed.ncbi.nlm.nih.gov/12765955/
Cai W, He JC, Zhu L, et al. Oral glycotoxins determine the effects of calorie restriction on oxidant stress, age-related diseases, and lifespan. Am J Pathol. 2008;173(2):327–36. https://pubmed.ncbi.nlm.nih.gov/18599606/
Negrean M, Stirban A, Stratmann B, et al. Effects of low– and high-advanced glycation endproduct meals on macro-and microvascular endothelial function and oxidative stress in patients with type 2 diabetes mellitus. Am J Clin Nutr. 2007;85(5):1236–43. https://pubmed.ncbi.nlm.nih.gov/17490958/
. Šebeková K, Brouder Šebeková K. Glycated proteins in nutrition: friend or foe? Exp Gerontol. 2019;117:76–90. https://pubmed.ncbi.nlm.nih.gov/30458224/
. Šebeková K, Brouder Šebeková K. Glycated proteins in nutrition: friend or foe? Exp Gerontol. 2019;117:76–90. https://pubmed.ncbi.nlm.nih.gov/30458224/
Atkinson FS, Foster-Powell K, Brand-Miller JC. International tables of glycemic index and glycemic load values: 2008. Diabetes Care. 2008;31(12):2281–3. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2584181/
Gaesser GA, Rodriguez J, Patrie JT, Whisner CM, Angadi SS. Effects of glycemic index and cereal fiber on postprandial endothelial function, glycemia, and insulinemia in healthy adults. Nutrients. 2019;11(10):2387. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6835298/
Pereira MA, Swain J, Goldfine AB, Rifai N, Ludwig DS. Effects of a low-glycemic load diet on resting energy expenditure and heart disease risk factors during weight loss. JAMA. 2004;292(20):2482–90. https://pubmed.ncbi.nlm.nih.gov/15562127/
Jenkins DJ, Taylor RH, Goff DV, et al. Scope and specificity of acarbose in slowing carbohydrate absorption in man. Diabetes. 1981;30(11):951–4. https://pubmed.ncbi.nlm.nih.gov/7028548/
Augustin LSA, Kendall CWC, Jenkins DJA, et al. Glycemic index, glycemic load and glycemic response: an international scientific consensus summit from the International Carbohydrate Quality Consortium (ICQC). Nutr Metab Cardiovasc Dis. 2015;25(9):795–815. https://pubmed.ncbi.nlm.nih.gov/26160327/
Schnell O, Weng J, Sheu WH, et al. Acarbose reduces body weight irrespective of glycemic control in patients with diabetes: results of a worldwide, non-interventional, observational study data pool. J Diabetes Complicat. 2016;30(4):628–37. https://pubmed.ncbi.nlm.nih.gov/26935335/
Tsunosue M, Mashiko N, Ohta Y, et al. An a-glucosidase inhibitor, acarbose treatment decreases serum levels of glyceraldehyde-derived advanced glycation end products (AGEs) in patients with type 2 diabetes. Clin Exp Med. 2010;10(2):139–41. https://pubmed.ncbi.nlm.nih.gov/19834782/
Newman JC, Milman S, Hashmi SK, et al. Strategies and challenges in clinical trials targeting human aging. J Gerontol A Biol Sci Med Sci. 2016;71(11):1424–34. https://pubmed.ncbi.nlm.nih.gov/27535968/
Brewer RA, Gibbs VK, Smith DL. Targeting glucose metabolism for healthy aging. Nutr Healthy Aging. 2016;4(1):31–46. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5166514/
Jenkins D, Wolever T, Taylor R, Barker H, Fielden H. Exceptionally low blood glucose response to dried beans: comparison with other carbohydrate foods. BMJ. 1980;281(6240):578–80. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1713902/
Jenkins DJ, Wolever TM, Taylor RH, et al. Slow release dietary carbohydrate improves second meal tolerance. Am J Clin Nutr. 1982;35(6):1339–46. https://pubmed.ncbi.nlm.nih.gov/6282105/
Wolever TM, Jenkins DJ, Ocana AM, Rao VA, Collier GR. Second-meal effect: low-glycemic-index foods eaten at dinner improve subsequent breakfast glycemic response. Am J Clin Nutr. 1988;48(4):1041–7. https://pubmed.ncbi.nlm.nih.gov/2844076/
Mollard RC, Wong CL, Luhovyy BL, Anderson GH. First and second meal effects of pulses on blood glucose, appetite, and food intake at a later meal. Appl Physiol Nutr Metab. 2011;36(5):634–42. https://pubmed.ncbi.nlm.nih.gov/21957874/
Jenkins DJA, Kendall CWC, Augustin LSA, et al. Effect of legumes as part of a low glycemic index diet on glycemic control and cardiovascular risk factors in type 2 diabetes mellitus: a randomized controlled trial. Arch Intern Med. 2012;172(21):1653–60. https://pubmed.ncbi.nlm.nih.gov/23089999/
Sievenpiper JL, Chiavaroli L, de Souza RJ, et al. “Catalytic” doses of fructose may benefit glycaemic control without harming cardiometabolic risk factors: a small meta-analysis of randomised controlled feeding trials. Br J Nutr. 2012;108(3):418–23. https://pubmed.ncbi.nlm.nih.gov/22354959/
Christensen AS, Viggers L, Hasselström K, Gregersen S. Effect of fruit restriction on glycemic control in patients with type 2 diabetes – a randomized trial. Nutr J. 2013;12:29. https://pubmed.ncbi.nlm.nih.gov/23497350/
Choo VL, Viguiliouk E, Mejia SB, et al. Food sources of fructose-containing sugars and glycaemic control: systematic review and meta-analysis of controlled intervention studies. BMJ. 2018;363:k4644. https://pubmed.ncbi.nlm.nih.gov/30463844/
McSwiney FT, Doyle L. Low-carbohydrate ketogenic diets in male endurance athletes demonstrate different micronutrient contents and changes in corpuscular haemoglobin over 12 weeks. Sports (Basel). 2019;7(9):201. https://pubmed.ncbi.nlm.nih.gov/31480346/
Sweeney JS. Dietary factors that influence the dextrose tolerance test: a preliminary study. Arch Intern Med (Chic). 1927;40(6):818–30. https://jamanetwork.com/journals/jamainternalmedicine/article-abstract/535594
Manco M, Bertuzzi A, Salinari S, et al. The ingestion of saturated fatty acid triacylglycerols acutely affects insulin secretion and insulin sensitivity in human subjects. Br J Nutr. 2004;92(6):895–903. https://pubmed.ncbi.nlm.nih.gov/15613251/
Koska J, Ozias MK, Deer J, et al. A human model of dietary saturated fatty acid induced insulin resistance. Metabolism. 2016;65(11):1621–8. https://pubmed.ncbi.nlm.nih.gov/27733250/
Angeloni C, Zambonin L, Hrelia S. Role of methylglyoxal in Alzheimer’s disease. Biomed Res Int. 2014;2014:238485. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3966409/
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–16.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Beisswenger BG, Delucia EM, Lapoint N, Sanford RJ, Beisswenger PJ. Ketosis leads to increased methylglyoxal production on the Atkins diet. Ann N Y Acad Sci. 2005;1043:201–10. https://pubmed.ncbi.nlm.nih.gov/16037240/
Franz MJ. Protein and diabetes: much advice, little research. Curr Diab Rep. 2002;2(5):457–64. https://pubmed.ncbi.nlm.nih.gov/12643172/
Jones AW, Rössner S. False-positive breath-alcohol test after a ketogenic diet. Int J Obes (Lond). 2007;31(3):559–61. https://pubmed.ncbi.nlm.nih.gov/16894360/
Beisswenger BG, Delucia EM, Lapoint N, Sanford RJ, Beisswenger PJ. Ketosis leads to increased methylglyoxal production on the Atkins diet. Ann N Y Acad Sci. 2005;1043:201–10. https://pubmed.ncbi.nlm.nih.gov/16037240/
Tey SL, Salleh NB, Henry CJ, Forde CG. Effects of non-nutritive (artificial vs natural) sweeteners on 24-h glucose profiles. Eur J Clin Nutr. 2017;71(9):1129–32. https://pubmed.ncbi.nlm.nih.gov/28378852/
Coca-Cola. Nutrition facts – original 20 fl oz. https://us.coca-cola.com/products/coca-cola/original. Accessed December 26, 2022.; https://us.coca-cola.com/products/coca-cola/original
Tey SL, Salleh NB, Henry J, Forde CG. Effects of aspartame-, monk fruit-, stevia– and sucrose-sweetened beverages on postprandial glucose, insulin and energy intake. Int J Obes (Lond). 2017;41(3):450–7. https://pubmed.ncbi.nlm.nih.gov/27956737/
Pepino MY, Tiemann CD, Patterson BW, Wice BM, Klein S. Sucralose affects glycemic and hormonal responses to an oral glucose load. Diabetes Care. 2013;36(9):2530–5. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3747933/
Atkinson FS, Foster-Powell K, Brand-Miller JC. International tables of glycemic index and glycemic load values: 2008. Diabetes Care. 2008;31(12):2281–3. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2584181/
Brand JC, Nicholson PL, Thorburn AW, Truswell AS. Food processing and the glycemic index. Am J Clin Nutr. 1985;42(6):1192–6. https://pubmed.ncbi.nlm.nih.gov/4072954/
Atkinson FS, Foster-Powell K, Brand-Miller JC. International tables of glycemic index and glycemic load values: 2008. Diabetes Care. 2008;31(12):2281–3. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2584181/
Mofidi A, Ferraro ZM, Stewart KA, et al. The acute impact of ingestion of sourdough and whole-grain breads on blood glucose, insulin, and incretins in overweight and obese men. J Nutr Metab. 2012;2012:184710. https://pubmed.ncbi.nlm.nih.gov/22474577/
Scazzina F, Siebenhandl-Ehn S, Pellegrini N. The effect of dietary fibre on reducing the glycaemic index of bread. Br J Nutr. 2013;109(7):1163–74. https://pubmed.ncbi.nlm.nih.gov/23414580/
Jenkins DJ, Wesson V, Wolever TM, et al. Wholemeal versus wholegrain breads: proportion of whole or cracked grain and the glycaemic response. BMJ. 1988;297(6654):958–60. https://pubmed.ncbi.nlm.nih.gov/3142566/
Breen C, Ryan M, Gibney MJ, Corrigan M, O’Shea D. Glycemic, insulinemic, and appetite responses of patients with type 2 diabetes to commonly consumed breads. Diabetes Educ. 2013;39(3):376–86. https://pubmed.ncbi.nlm.nih.gov/23482513/
Reynolds AN, Mann J, Elbalshy M, et al. Wholegrain particle size influences postprandial glycemia in type 2 diabetes: a randomized crossover study comparing four wholegrain breads. Dia Care. 2020;43(2):476–9. https://pubmed.ncbi.nlm.nih.gov/31744812/
Burton P, Lightowler HJ. The impact of freezing and toasting on the glycaemic response of white bread. Eur J Clin Nutr. 2008;62(5):594–9. https://pubmed.ncbi.nlm.nih.gov/17426743/
Scazzina F, Siebenhandl-Ehn S, Pellegrini N. The effect of dietary fibre on reducing the glycaemic index of bread. Br J Nutr. 2013;109(7):1163–74. https://pubmed.ncbi.nlm.nih.gov/23414580/
Yadav BS, Sharma A, Yadav RB. Studies on effect of multiple heating/cooling cycles on the resistant starch formation in cereals, legumes and tubers. Int J Food Sci Nutr. 2009;60 Suppl 4:258–72. https://pubmed.ncbi.nlm.nih.gov/19562607/
de Morais Cardoso L, Pinheiro SS, Martino HSD, Pinheiro-Sant’Ana HM. Sorghum (Sorghum bicolor L.): nutrients, bioactive compounds, and potential impact on human health. Crit Rev Food Sci Nutr. 2017;57(2):372–90. https://pubmed.ncbi.nlm.nih.gov/25875451/
Narayanan J, Sanjeevi V, Rohini U, Trueman P, Viswanathan V. Postprandial glycaemic response of foxtail millet dosa in comparison to a rice dosa in patients with type 2 diabetes. Indian J Med Res. 2016;144(5):712–7. https://pubmed.ncbi.nlm.nih.gov/28361824/
Poquette NM, Gu X, Lee SO. Grain sorghum muffin reduces glucose and insulin responses in men. Food Funct. 2014;5(5):894–9. https://pubmed.ncbi.nlm.nih.gov/24608948/
Abdelgadir M, Abbas M, Järvi A, Elbagir M, Eltom M, Berne C. Glycaemic and insulin responses of six traditional Sudanese carbohydrate-rich meals in subjects with Type 2 diabetes mellitus. Diabet Med. 2005;22(2):213–7. https://pubmed.ncbi.nlm.nih.gov/15660741/
Chen Z, Glisic M, Song M, et al. Dietary protein intake and all-cause and cause-specific mortality: results from the Rotterdam Study and a meta-analysis of prospective cohort studies. Eur J Epidemiol. 2020;35(5):411–29. https://pubmed.ncbi.nlm.nih.gov/32076944/
Mazidi M, Katsiki N, Mikhailidis DP, Pella D, Banach M. Potato consumption is associated with total and cause-specific mortality: a population-based cohort study and pooling of prospective studies with 98,569 participants. Arch Med Sci. 2020;16(2):260–72. https://pubmed.ncbi.nlm.nih.gov/32190135/
Fernandes G, Velangi A, Wolever TMS. Glycemic index of potatoes commonly consumed in North America. J Am Diet Assoc. 2005;105(4):557–62. https://pubmed.ncbi.nlm.nih.gov/15800557/
Johnston CS, Steplewska I, Long CA, Harris LN, Ryals RH. Examination of the antiglycemic properties of vinegar in healthy adults. Ann Nutr Metab. 2010;56(1):74–9. https://pubmed.ncbi.nlm.nih.gov/20068289/
Leeman M, Östman E, Björck I. Vinegar dressing and cold storage of potatoes lowers postprandial glycaemic and insulinaemic responses in healthy subjects. Eur J Clin Nutr. 2005;59(11):1266–71. https://pubmed.ncbi.nlm.nih.gov/16034360/
Grussu D, Stewart D, McDougall GJ. Berry polyphenols inhibit a-amylase in vitro: identifying active components in rowanberry and raspberry. J Agric Food Chem. 2011;59(6):2324–31. https://pubmed.ncbi.nlm.nih.gov/21329358/
Sharma KK, Gupta RK, Gupta S, Samuel KC. Antihyperglycemic effect of onion: effect on fasting blood sugar and induced hyperglycemia in man. Indian J Med Res. 1977;65(3):422–9. https://pubmed.ncbi.nlm.nih.gov/336527/
Haldar S, Chia SC, Lee SH, et al. Polyphenol-rich curry made with mixed spices and vegetables benefits glucose homeostasis in Chinese males (Polyspice Study): a dose-response randomized controlled crossover trial. Eur J Nutr. 2019;58(1):301–13. https://pubmed.ncbi.nlm.nih.gov/29236165/
Azzeh FS. Synergistic effect of green tea, cinnamon and ginger combination on enhancing postprandial blood glucose. Pak J Biol Sci. 2013;16(2):74–9. https://pubmed.ncbi.nlm.nih.gov/24199490/
Hajizadeh-Sharafabad F, Varshosaz P, Jafari-Vayghan H, Alizadeh M, Maleki V. Chamomile (Matricaria recutita L.) and diabetes mellitus, current knowledge and the way forward: a systematic review. Complement Ther Med. 2020;48:102284. https://pubmed.ncbi.nlm.nih.gov/31987240/
Rafraf M, Zemestani M, Asghari-Jafarabadi M. Effectiveness of chamomile tea on glycemic control and serum lipid profile in patients with type 2 diabetes. J Endocrinol Invest. 2015;38(2):163–70. https://pubmed.ncbi.nlm.nih.gov/25194428/
Kermanian S, Mozaffari-Khosravi H, Dastgerdi G, Zavar-Reza J, Rahmanian M. The effect of chamomile tea versus black tea on glycemic control and blood lipid profiles in depressed patients with type 2 diabetes: a randomized clinical trial. JNFS, 2018;3(3):157–66. https://jnfs.ssu.ac.ir/article-1-197-en.pdf
Rafraf M, Zemestani M, Asghari-Jafarabadi M. Effectiveness of chamomile tea on glycemic control and serum lipid profile in patients with type 2 diabetes. J Endocrinol Invest. 2015;38(2):163–70. https://pubmed.ncbi.nlm.nih.gov/25194428/
Pirouzpanah S, Mahboob S, Sanayei M, Hajaliloo M, Safaeiyan A. The effect of chamomile tea consumption on inflammation among rheumatoid arthritis patients: randomized clinical trial. Prog Nutr. 2017;19(1-S)27–33. https://doi.org/10.23751/PN.V19I1-S.5171
Chang SM, Chen CH. Effects of an intervention with drinking chamomile tea on sleep quality and depression in sleep disturbed postnatal women: a randomized controlled trial. J Adv Nurs. 2016;72(2):306–15. https://pubmed.ncbi.nlm.nih.gov/26483209/
Zemestani M, Rafraf M, Asghari-Jafarabadi M. Chamomile tea improves glycemic indices and antioxidants status in patients with type 2 diabetes mellitus. Nutrition. 2016;32(1):66–72. https://pubmed.ncbi.nlm.nih.gov/26437613/
Villa-Rodriguez JA, Aydin E, Gauer JS, Pyner A, Williamson G, Kerimi A. Green and chamomile teas, but not acarbose, attenuate glucose and fructose transport via inhibition of GLUT2 and GLUT5. Mol Nutr Food Res. 2017;61(12):1700566. https://pubmed.ncbi.nlm.nih.gov/28868668/
Bowen AJ, Reeves RL. Diurnal variation in glucose tolerance. Arch Intern Med. 1967;119(3):261–4. https://pubmed.ncbi.nlm.nih.gov/6019944/
Van Cauter E, Polonsky KS, Scheen AJ. Roles of circadian rhythmicity and sleep in human glucose regulation. Endocr Rev. 1997;18(5):716–38. https://pubmed.ncbi.nlm.nih.gov/9331550/
Bandín C, Scheer FA, Luque AJ, et al. Meal timing affects glucose tolerance, substrate oxidation and circadian-related variables: a randomized, crossover trial. Int J Obes (Lond). 2015;39(5):828–33. https://pubmed.ncbi.nlm.nih.gov/25311083/
Gibbs M, Harrington D, Starkey S, Williams P, Hampton S. Diurnal postprandial responses to low and high glycaemic index mixed meals. Clin Nutr. 2014;33(5):889–94. https://pubmed.ncbi.nlm.nih.gov/24135087/
3,2 км/ч. – Примеч. ред.
Colberg SR, Zarrabi L, Bennington L, et al. Postprandial walking is better for lowering the glycemic effect of dinner than pre-dinner exercise in type 2 diabetic individuals. J Am Med Dir Assoc. 2009;10(6):394–7. https://pubmed.ncbi.nlm.nih.gov/19560716/
Haxhi J, Scotto di Palumbo A, Sacchetti M. Exercising for metabolic control: is timing important? Ann Nutr Metab. 2013;62(1):14–25. https://pubmed.ncbi.nlm.nih.gov/23208206/
Reynolds AN, Mann JI, Williams S, Venn BJ. Advice to walk after meals is more effective for lowering postprandial glycaemia in type 2 diabetes mellitus than advice that does not specify timing: a randomised crossover study. Diabetologia. 2016;59(12):2572–8. https://pubmed.ncbi.nlm.nih.gov/27747394/
Rahmadi A, Steiner N, Münch G. Advanced glycation endproducts as gerontotoxins and biomarkers for carbonyl-based degenerative processes in Alzheimer’s disease. Clin Chem Lab Med. 2011;49(3):385–91. https://pubmed.ncbi.nlm.nih.gov/21275816/
Green AS. mTOR, glycotoxins and the parallel universe. Aging (Albany NY). 2018;10(12):3654–6. https://pubmed.ncbi.nlm.nih.gov/30540565/
Uribarri J, He JC. The low AGE diet: a neglected aspect of clinical nephrology practice? Nephron. 2015;130(1):48–53. https://pubmed.ncbi.nlm.nih.gov/25871778/
Yamagishi S, Nakamura K, Matsui T, Inoue H, Takeuchi M. Oral administration of AST-120 (Kremezin) is a promising therapeutic strategy for advanced glycation end product (AGE)-related disorders. Med Hypotheses. 2007;69(3):666–8. https://pubmed.ncbi.nlm.nih.gov/17331665/
MIMS. Kremezin full prescribing information, dosage & side effects. https://www.mims.com/philippines/drug/info/kremezin?type=full. Accessed December 26, 2022.; https://www.mims.com/philippines/drug/info/kremezin?type=full
Uribarri J, Woodruff S, Goodman S, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010;110(6):911–6.e12. https://pubmed.ncbi.nlm.nih.gov/20497781/
Cerami C, Founds H, Nicholl I, et al. Tobacco smoke is a source of toxic reactive glycation products. Proc Natl Acad Sci USA. 1997;94(25):13915–20. https://pubmed.ncbi.nlm.nih.gov/9391127/
Green AS. mTOR, glycotoxins and the parallel universe. Aging (Albany NY). 2018;10(12):3654–6. https://pubmed.ncbi.nlm.nih.gov/30540565/
Green AS. mTOR, glycotoxins and the parallel universe. Aging (Albany NY). 2018;10(12):3654–6. https://pubmed.ncbi.nlm.nih.gov/30540565/
Kenyon C. The first long-lived mutants: discovery of the insulin/IGF-1 pathway for ageing. Philos Trans R Soc Lond B Biol Sci. 2011;366(1561):9–16. https://pubmed.ncbi.nlm.nih.gov/21115525/
Kenyon C, Chang J, Gensch E, Rudner A, Tabtiang R. A C. elegans mutant that lives twice as long as wild type. Nature. 1993;366(6454):461–4. https://pubmed.ncbi.nlm.nih.gov/8247153/
Kenyon C. The first long-lived mutants: discovery of the insulin/IGF-1 pathway for ageing. Philos Trans R Soc Lond B Biol Sci. 2011;366(1561):9–16. https://pubmed.ncbi.nlm.nih.gov/21115525/
Partridge L, Harvey PH. Gerontology. Methuselah among nematodes. Nature. 1993;366(6454):404–5. https://pubmed.ncbi.nlm.nih.gov/8247143/
Мрачный жнец – образ смерти. – Примеч. ред.
Coffer P. OutFOXing the grim reaper: novel mechanisms regulating longevity by Forkhead transcription factors. Sci STKE. 2003;2003(201):PE39. https://pubmed.ncbi.nlm.nih.gov/14506287/
Suh Y, Atzmon G, Cho MO, et al. Functionally significant insulin-like growth factor I receptor mutations in centenarians. Proc Natl Acad Sci U S A. 2008;105(9):3438–42. https://pubmed.ncbi.nlm.nih.gov/18316725/
Kenyon C. The first long-lived mutants: discovery of the insulin/IGF-1 pathway for ageing. Philos Trans R Soc Lond B Biol Sci. 2011;366(1561):9–16. https://pubmed.ncbi.nlm.nih.gov/21115525/
Laron Z, Kauli R, Lapkina L, Werner H. IGF-I deficiency, longevity and cancer protection of patients with Laron syndrome. Mutat Res Rev Mutat Res. 2017;772:123–33. https://pubmed.ncbi.nlm.nih.gov/28528685/
Vitale G, Pellegrino G, Vollery M, Hofland LJ. Role of IGF-1 system in the modulation of longevity: controversies and new insights from a centenarians’ perspective. Front Endocrinol. 2019;10:27. https://pubmed.ncbi.nlm.nih.gov/30774624/
Kenyon C. The plasticity of aging: insights from long-lived mutants. Cell. 2005;120(4):449–60. https://pubmed.ncbi.nlm.nih.gov/15734678/
Junnila RK, List EO, Berryman DE, Murrey JW, Kopchick JJ. The GH/IGF-1 axis in ageing and longevity. Nat Rev Endocrinol. 2013;9(6):366–76. https://pubmed.ncbi.nlm.nih.gov/23591370/
Vitale G, Barbieri M, Kamenetskaya M, Paolisso G. GH/IGF-I/insulin system in centenarians. Mech Ageing Dev. 2017;165(Pt B):107–14. https://pubmed.ncbi.nlm.nih.gov/27932301/
Vitale G, Brugts MP, Ogliari G, et al. Low circulating IGF-I bioactivity is associated with human longevity: findings in centenarians’ offspring. Aging (Albany NY). 2012;4(9):580–9. https://pubmed.ncbi.nlm.nih.gov/22983440/
Vitale G, Barbieri M, Kamenetskaya M, Paolisso G. GH/IGF-I/insulin system in centenarians. Mech Ageing Dev. 2017;165(Pt B):107–14. https://pubmed.ncbi.nlm.nih.gov/27932301/
Pawlikowska L, Hu D, Huntsman S, et al. Association of common genetic variation in the insulin/IGF1 signaling pathway with human longevity. Aging Cell. 2009;8(4):460–72. https://pubmed.ncbi.nlm.nih.gov/19489743/
Ben-Avraham D, Govindaraju DR, Budagov T, et al. The GH receptor exon 3 deletion is a marker of male-specific exceptional longevity associated with increased GH sensitivity and taller stature. Sci Adv. 2017;3(6):e1602025. https://pubmed.ncbi.nlm.nih.gov/28630896/
Teumer A, Qi Q, Nethander M, et al. Genomewide meta-analysis identifies loci associated with IGF-I and IGFBP-3 levels with impact on age-related traits. Aging Cell. 2016;15(5):811–24. https://pubmed.ncbi.nlm.nih.gov/27329260/
Milman S, Atzmon G, Huffman DM, et al. Low insulin-like growth factor-1 level predicts survival in humans with exceptional longevity. Aging Cell. 2014;13(4):769–71. https://pubmed.ncbi.nlm.nih.gov/24618355/
van der Spoel E, Rozing MP, Houwing-Duistermaat JJ, et al. Association analysis of insulin-like growth factor-1 axis parameters with survival and functional status in nonagenarians of the Leiden Longevity Study. Aging (Albany NY). 2015;7(11):956–63. https://pubmed.ncbi.nlm.nih.gov/26568155/
Suh Y, Atzmon G, Cho MO, et al. Functionally significant insulin-like growth factor I receptor mutations in centenarians. Proc Natl Acad Sci U S A. 2008;105(9):3438–42. https://pubmed.ncbi.nlm.nih.gov/18316725/
Tazearslan C, Huang J, Barzilai N, Suh Y. Impaired IGF1R signaling in cells expressing longevity-associated human IGF1R alleles. Aging Cell. 2011;10(3):551–4. https://pubmed.ncbi.nlm.nih.gov/21388493/
Bartke A. Healthy aging: is smaller better? – a mini-review. Gerontology. 2012;58(4):337–43. https://pubmed.ncbi.nlm.nih.gov/22261798/
Michell AR. Longevity of British breeds of dog and its relationships with sex, size, cardiovascular variables and disease. Vet Rec. 1999;145(22):625–9. https://pubmed.ncbi.nlm.nih.gov/10619607/
Sutter NB, Bustamante CD, Chase K, et al. A single IGF1 allele is a major determinant of small size in dogs. Science. 2007;316(5821):112–5. https://pubmed.ncbi.nlm.nih.gov/17412960/
Samaras TT. How height is related to our health and longevity: a review. Nutr Health. 2012;21(4):247–61. https://pubmed.ncbi.nlm.nih.gov/24620006/
Sohn K. Now, the taller die earlier: the curse of cancer. J Gerontol A Biol Sci Med Sci. 2016;71(6):713–9. https://pubmed.ncbi.nlm.nih.gov/25991828/
Samaras TT. How height is related to our health and longevity: a review. Nutr Health. 2012;21(4):247–61. https://pubmed.ncbi.nlm.nih.gov/24620006/
Samaras TT, Elrick H, Storms LH. Is height related to longevity? Life Sci. 2003;72(16):1781–802. https://pubmed.ncbi.nlm.nih.gov/12586217/
Samaras TT. How height is related to our health and longevity: a review. Nutr Health. 2012;21(4):247–61. https://pubmed.ncbi.nlm.nih.gov/24620006/
Один дюйм равен 2,54 см. – Примеч. ред.
Sohn K. Now, the taller die earlier: the curse of cancer. J Gerontol A Biol Sci Med Sci. 2016;71(6):713–9. https://pubmed.ncbi.nlm.nih.gov/25991828/
Walter RB, Brasky TM, Buckley SA, Potter JD, White E. Height as an explanatory factor for sex differences in human cancer. J Natl Cancer Inst. 2013;105(12):860–8. https://pubmed.ncbi.nlm.nih.gov/23708052/
Shors AR, Solomon C, McTiernan A, White E. Melanoma risk in relation to height, weight, and exercise (United States). Cancer Causes Control. 2001;12(7):599–606. https://pubmed.ncbi.nlm.nih.gov/11552707/
Walter RB, Brasky TM, Buckley SA, Potter JD, White E. Height as an explanatory factor for sex differences in human cancer. J Natl Cancer Inst. 2013;105(12):860–8. https://pubmed.ncbi.nlm.nih.gov/23708052/
Suh Y, Atzmon G, Cho MO, et al. Functionally significant insulin-like growth factor I receptor mutations in centenarians. Proc Natl Acad Sci U S A. 2008;105(9):3438–42. https://pubmed.ncbi.nlm.nih.gov/18316725/
Reed JC. Dysregulation of apoptosis in cancer. J Clin Oncol. 1999;17(9):2941–53. https://pubmed.ncbi.nlm.nih.gov/10561374/
Murphy N, Knuppel A, Papadimitriou N, et al. Insulin-like growth factor-1, insulin-like growth factor-binding protein-3, and breast cancer risk: observational and Mendelian randomization analyses with ~430 000 women. Ann Oncol. 2020;31(5):641–9. https://pubmed.ncbi.nlm.nih.gov/32169310/
Chi F, Wu R, Zeng Y, Xing R, Liu Y. Circulation insulin-like growth factor peptides and colorectal cancer risk: an updated systematic review and meta-analysis. Mol Biol Rep. 2013;40(5):3583–90. https://pubmed.ncbi.nlm.nih.gov/23269623/
Travis RC, Appleby PN, Martin RM, et al. A meta-analysis of individual participant data reveals an association between circulating levels of IGF-I and prostate cancer risk. Cancer Res. 2016;76(8):2288–300. https://pubmed.ncbi.nlm.nih.gov/26921328/
Cao H, Wang G, Meng L, et al. Association between circulating levels of IGF-1 and IGFBP-3 and lung cancer risk: a meta-analysis. PLoS One. 2012;7(11):e49884. https://pubmed.ncbi.nlm.nih.gov/23185474/
Li Y, Li Y, Zhang J, et al. Circulating insulin-like growth factor-1 level and ovarian cancer risk. Cell Physiol Biochem. 2016;38(2):589–97. https://pubmed.ncbi.nlm.nih.gov/26845340/
Gong Y, Zhang B, Liao Y, et al. Serum insulin-like growth factor axis and the risk of pancreatic cancer: systematic review and meta-analysis. Nutrients. 2017;9(4):394. https://pubmed.ncbi.nlm.nih.gov/28420208/
Hankinson SE, Willett WC, Colditz GA, et al. Circulating concentrations of insulin-like growth factor I and risk of breast cancer. Lancet. 1998;351(9113):1393–6. https://pubmed.ncbi.nlm.nih.gov/9593409/
Yee D. Insulin-like growth factor receptor inhibitors: baby or the bathwater? J Natl Cancer Inst. 2012;104(13):975–81. https://pubmed.ncbi.nlm.nih.gov/22761272/
Quan H, Tang H, Fang L, Bi J, Liu Y, Li H. IGF1(CA)19 and IGFBP-3–202A/C gene polymorphism and cancer risk: a meta-analysis. Cell Biochem Biophys. 2014;69(1):169–78. https://pubmed.ncbi.nlm.nih.gov/24310658/
Yokoyama NN, Denmon AP, Uchio EM, Jordan M, Mercola D, Zi X. When anti-aging studies meet cancer chemoprevention: can anti-aging agent kill two birds with one blow? Curr Pharmacol Rep. 2015;1(6):420–33. https://pubmed.ncbi.nlm.nih.gov/26756023/
Elia I, Doglioni G, Fendt SM. Metabolic hallmarks of metastasis formation. Trends Cell Biol. 2018;28(8):673–84. https://pubmed.ncbi.nlm.nih.gov/29747903/
Kleinberg DL, Wood TL, Furth PA, Lee AV. Growth hormone and insulin-like growth factor-I in the transition from normal mammary development to preneoplastic mammary lesions. Endocr Rev. 2009;30(1):51–74. https://pubmed.ncbi.nlm.nih.gov/19075184/
Yang SY, Miah A, Pabari A, Winslet M. Growth factors and their receptors in cancer metastases. Front Biosci (Landmark Ed). 2011;16:531–8. https://pubmed.ncbi.nlm.nih.gov/21196186/
Zhang Y, Ma B, Fan Q. Mechanisms of breast cancer bone metastasis. Cancer Lett. 2010;292(1):1–7. https://pubmed.ncbi.nlm.nih.gov/20006425/
Yang SY, Miah A, Pabari A, Winslet M. Growth factors and their receptors in cancer metastases. Front Biosci (Landmark Ed). 2011;16:531–8. https://pubmed.ncbi.nlm.nih.gov/21196186/
Sohn K. Now, the taller die earlier: the curse of cancer. J Gerontol A Biol Sci Med Sci. 2016;71(6):713–19. https://pubmed.ncbi.nlm.nih.gov/25991828/
Salvioli S, Capri M, Bucci L, et al. Why do centenarians escape or postpone cancer? The role of IGF-1, inflammation and p53. Cancer Immunol Immunother. 2009;58(12):1909–17. https://pubmed.ncbi.nlm.nih.gov/19139887/
Piantanelli L. Cancer and aging: from the kinetics of biological parameters to the kinetics of cancer incidence and mortality. Ann N Y Acad Sci. 1988;521:99–109. https://pubmed.ncbi.nlm.nih.gov/3377369/
Kenyon C. The plasticity of aging: insights from long-lived mutants. Cell. 2005;120(4):449–60. https://pubmed.ncbi.nlm.nih.gov/15734678/
Stanta G, Campagner L, Cavallieri F, Giarelli L. Cancer of the oldest old. What we have learned from autopsy studies. Clin Geriatr Med. 1997;13(1):55–68. https://pubmed.ncbi.nlm.nih.gov/8995100/
Salvioli S, Capri M, Bucci L, et al. Why do centenarians escape or postpone cancer? The role of IGF-1, inflammation and p53. Cancer Immunol Immunother. 2009;58(12):1909–17. https://pubmed.ncbi.nlm.nih.gov/19139887/
Laron Z, Pertzelan A, Mannheimer S. Genetic pituitary dwarfism with high serum concentration of growth hormone: a new inborn error of metabolism? Isr J Med Sci 1966;2:152–5. https://pubmed.ncbi.nlm.nih.gov/5916640/
Guevara-Aguirre J, Bautista C, Torres C, et al. Insights from the clinical phenotype of subjects with Laron syndrome in Ecuador. Rev Endocr Metab Disord. 2021;22(1):59–70. https://pubmed.ncbi.nlm.nih.gov/33047268/
Laron Z, Kauli R, Lapkina L, Werner H. IGF-I deficiency, longevity and cancer protection of patients with Laron syndrome. Mutat Res Rev Mutat Res. 2017;772:123–33. https://pubmed.ncbi.nlm.nih.gov/28528685/
Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, et al. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med. 2011;3(70):70ra13. https://pubmed.ncbi.nlm.nih.gov/21325617/
Boguszewski CL, Boguszewski MC da S. Growth hormone’s links to cancer. Endocr Rev. 2019;40(2):558–74. https://pubmed.ncbi.nlm.nih.gov/30500870/
Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, et al. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med. 2011;3(70):70ra13. https://pubmed.ncbi.nlm.nih.gov/21325617/
Laron Z, Kauli R, Lapkina L, Werner H. IGF-I deficiency, longevity and cancer protection of patients with Laron syndrome. Mutat Res Rev Mutat Res. 2017;772:123–33. https://pubmed.ncbi.nlm.nih.gov/28528685/
Ma H, Zhang T, Shen H, Cao H, Du J. The adverse events profile of anti-IGF-1R monoclonal antibodies in cancer therapy. Br J Clin Pharmacol. 2014;77(6):917–28. https://pubmed.ncbi.nlm.nih.gov/24033707/
Thissen JP, Ketelslegers JM, Underwood LE. Nutritional regulation of the insulin-like growth factors. Endocr Rev. 1994;15(1):80–101. https://pubmed.ncbi.nlm.nih.gov/8156941/
Lee C, Safdie FM, Raffaghello L, et al. Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res. 2010;70(4):1564–72. https://pubmed.ncbi.nlm.nih.gov/20145127/
Longo VD, Anderson RM. Nutrition, longevity and disease: from molecular mechanisms to interventions. Cell. 2022;185(9):1455–70. https://pubmed.ncbi.nlm.nih.gov/35487190/
Dunn SE, Kari FW, French J, et al. Dietary restriction reduces insulin-like growth factor I levels, which modulates apoptosis, cell proliferation, and tumor progression in p53-deficient mice. Cancer Res. 1997;57(21):4667–72. https://pubmed.ncbi.nlm.nih.gov/9354418/
Fontana L, Weiss EP, Villareal DT, Klein S, Holloszy JO. Long-term effects of calorie or protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell. 2008;7(5):681–7. https://pubmed.ncbi.nlm.nih.gov/18843793/
Schüler R, Markova M, Osterhoff MA, et al. Similar dietary regulation of IGF-1-and IGF-binding proteins by animal and plant protein in subjects with type 2 diabetes. Eur J Nutr. https://link.springer.com/article/10.1007/s00394–021–02518-y. Published online March 8, 2021. Accessed June 23, 2021.; https://pubmed.ncbi.nlm.nih.gov/33686453/
Allen NE, Appleby PN, Davey GK, Key TJ. Hormones and diet: low insulin-like growth factor-I but normal bioavailable androgens in vegan men. Br J Cancer. 2000;83(1):95–7. https://pubmed.ncbi.nlm.nih.gov/10883675/
Allen NE, Appleby PN, Davey GK, Kaaks R, Rinaldi S, Key TJ. The associations of diet with serum insulin-like growth factor I and its main binding proteins in 292 women meat-eaters, vegetarians, and vegans. Cancer Epidemiol Biomarkers Prev. 2002;11(11):1441–8. https://pubmed.ncbi.nlm.nih.gov/12433724/
Ngo TH, Barnard RJ, Tymchuk CN, Cohen P, Aronson WJ. Effect of diet and exercise on serum insulin, IGF-I, and IGFBP-1 levels and growth of LNCaP cells in vitro (United States). Cancer Causes Control. 2002;13(10):929–35. https://pubmed.ncbi.nlm.nih.gov/12588089/
Flood A, Mai V, Pfeiffer R, et al. The effects of a high-fruit and – vegetable, high-fiber, low-fat dietary intervention on serum concentrations of insulin, glucose, IGF-I and IGFBP-3. Eur J Clin Nutr. 2008;62(2):186–96. https://pubmed.ncbi.nlm.nih.gov/17487212/
Allen NE, Appleby PN, Davey GK, Key TJ. Hormones and diet: low insulin-like growth factor-I but normal bioavailable androgens in vegan men. Br J Cancer. 2000;83(1):95–7. https://pubmed.ncbi.nlm.nih.gov/10883675/
Allen NE, Appleby PN, Davey GK, Kaaks R, Rinaldi S, Key TJ. The associations of diet with serum insulin-like growth factor I and its main binding proteins in 292 women meat-eaters, vegetarians, and vegans. Cancer Epidemiol Biomarkers Prev. 2002;11(11):1441–8. https://pubmed.ncbi.nlm.nih.gov/12433724/
Berrino F, Bellati C, Secreto G, et al. Reducing bioavailable sex hormones through a comprehensive change in diet: the diet and androgens (DIANA) randomized trial. Cancer Epidemiol Biomarkers Prev. 2001;10(1):25–33. https://pubmed.ncbi.nlm.nih.gov/11205485/
Kaaks R, Bellati C, Venturelli E, et al. Effects of dietary intervention on IGF-I and IGF-binding proteins, and related alterations in sex steroid metabolism: the Diet and Androgens (DIANA) Randomised Trial. Eur J Clin Nutr. 2003;57(9):1079–88. https://pubmed.ncbi.nlm.nih.gov/12947426/
Pasanisi P, Bruno E, Venturelli E, et al. A dietary intervention to lower serum levels of IGF-I in BRCA mutation carriers. Cancers (Basel). 2018;10(9):309. https://pubmed.ncbi.nlm.nih.gov/30181513/
Gulick CN, Peddie MC, Cameron C, Bradbury K, Rehrer NJ. Physical activity, dietary protein and insulin-like growth factor 1: cross-sectional analysis utilising UK Biobank. Growth Horm IGF Res. 2020;55:101353. https://pubmed.ncbi.nlm.nih.gov/33002777/
Toden S, Belobrajdic DP, Bird AR, Topping DL, Conlon MA. Effects of dietary beef and chicken with and without high amylose maize starch on blood malondialdehyde, interleukins, IGF-I, insulin, leptin, MMP-2, and TIMP-2 concentrations in rats. Nutr Cancer. 2010;62(4):454–65. https://pubmed.ncbi.nlm.nih.gov/20432166/
Qin LQ, He K, Xu JY. Milk consumption and circulating insulin-like growth factor-I level: a systematic literature review. Int J Food Sci Nutr. 2009;60(S7):330–40. https://pubmed.ncbi.nlm.nih.gov/19746296/
Один галлон равен 4,55 л. – Примеч. ред.
Hoppe C, Kristensen M, Boiesen M, Kudsk J, Michaelsen KF, Mølgaard C. Short-term effects of replacing milk with cola beverages on insulin-like growth factor-I and insulin – glucose metabolism: a 10 d interventional study in young men. Br J Nutr. 2009;102(7):1047–51. https://pubmed.ncbi.nlm.nih.gov/15578035/
Harrison S, Lennon R, Holly J, et al. Does milk intake promote prostate cancer initiation or progression via effects on insulin-like growth factors (IGFs)? A systematic review and meta-analysis. Cancer Causes Control. 2017;28(6):497–528. https://pubmed.ncbi.nlm.nih.gov/28361446/
Adams AM, Smith AF. Risk perception and communication: recent developments and implications for anaesthesia. Anaesthesia. 2001;56(8):745–55. https://pubmed.ncbi.nlm.nih.gov/11493237/
Harrison S, Lennon R, Holly J, et al. Does milk intake promote prostate cancer initiation or progression via effects on insulin-like growth factors (IGFs)? A systematic review and meta-analysis. Cancer Causes Control. 2017;28(6):497–528. https://pubmed.ncbi.nlm.nih.gov/28361446/
Naghshi S, Sadeghi O, Larijani B, Esmaillzadeh A. High vs. low-fat dairy and milk differently affects the risk of all-cause, CVD, and cancer death: a systematic review and dose-response meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr. 2021;Jan 5:1–15. https://pubmed.ncbi.nlm.nih.gov/33397132/
Qin LQ, He K, Xu JY. Milk consumption and circulating insulin-like growth factor-I level: a systematic literature review. Int J Food Sci Nutr. 2009;60(7):330–40. https://pubmed.ncbi.nlm.nih.gov/19746296/
Jones CM, Heinrichs J. Growth charts for dairy heifers. Penn State Extension. https://extension.psu.edu/growth-charts-for-dairy-heifers. Updated July 28, 2017. Accessed June 9, 2021.; https://extension.psu.edu/growth-charts-for-dairy-heifers
Clatici VG, Voicu C, Voaides C, Roseanu A, Icriverzi M, Jurcoane S. Diseases of civilization – cancer, diabetes, obesity and acne – the implication of milk, IGF-1 and mTORC1. Maedica (Bucur). 2018;13(4):273–81. https://pubmed.ncbi.nlm.nih.gov/30774725/
Honegger A, Humbel RE. Insulin-like growth factors I and II in fetal and adult bovine serum. Purification, primary structures, and immunological cross-reactivities. J Biol Chem. 1986;261(2):569–75. https://pubmed.ncbi.nlm.nih.gov/3941093/
Collier RJ, Miller MA, Hildebrandt JR, et al. Factors affecting insulin-like growth factor-I concentration in bovine milk. J Dairy Sci. 1991;74(9):2905–11. https://pubmed.ncbi.nlm.nih.gov/1779049/
Kim WK, Ryu YH, Seo DS, Lee CY, Ko Y. Effects of oral administration of insulin-like growth factor-I on circulating concentration of insulin-like growth factor-I and growth of internal organs in weanling mice. Biol Neonate. 2006;89(3):199–204. https://pubmed.ncbi.nlm.nih.gov/16293962/
Clatici VG, Voicu C, Voaides C, Roseanu A, Icriverzi M, Jurcoane S. Diseases of civilization – cancer, diabetes, obesity and acne – the implication of milk, IGF-1 and mTORC1. Maedica (Bucur). 2018;13(4):273–81. https://pubmed.ncbi.nlm.nih.gov/30774725/
Allen NE, Key TJ. Re: plasma insulin-like growth factor-I, insulin-like growth factor-binding proteins, and prostate cancer risk: a prospective study. J Natl Cancer Inst. 2001;93(8):649–51. https://pubmed.ncbi.nlm.nih.gov/11309444/
Conover CA. Discrepancies in insulin-like growth factor signaling? No, not really. Growth Horm IGF Res. 2016;30–31:42–4. https://pubmed.ncbi.nlm.nih.gov/27792888/
Allen NE, Appleby PN, Davey GK, Kaaks R, Rinaldi S, Key TJ. The associations of diet with serum insulin-like growth factor I and its main binding proteins in 292 women meat-eaters, vegetarians, and vegans. Cancer Epidemiol Biomarkers Prev. 2002;11(11):1441–8. https://pubmed.ncbi.nlm.nih.gov/12433724/
Clemmons DR, Seek MM, Underwood LE. Supplemental essential amino acids augment the somatomedin-C/insulin-like growth factor I response to refeeding after fasting. Metabolism. 1985;34(4):391–5. https://pubmed.ncbi.nlm.nih.gov/3884968/
Mariotti F, Gardner CD. Dietary protein and amino acids in vegetarian diets – a review. Nutrients. 2019;11(11):2661. https://pubmed.ncbi.nlm.nih.gov/31690027/
Ten Have GAM, Engelen MPKJ, Soeters PB, Deutz NEP. Absence of post-prandial gut anabolism after intake of a low quality protein meal. Clin Nutr. 2012;31(2):273–82. https://pubmed.ncbi.nlm.nih.gov/22001026/
Katz DL, Doughty KN, Geagan K, Jenkins DA, Gardner CD. Perspective: the public health case for modernizing the definition of protein quality. Adv Nutr. 2019;10(5):755–64. https://pubmed.ncbi.nlm.nih.gov/31066877/
Freda PU, Shen W, Reyes-Vidal CM, et al. Skeletal muscle mass in acromegaly assessed by magnetic resonance imaging and dual-photon x-ray absorptiometry. J Clin Endocrinol Metab. 2009;94(8):2880–6. https://pubmed.ncbi.nlm.nih.gov/19491226/
Friedlander AL, Butterfield GE, Moynihan S, et al. One year of insulin-like growth factor I treatment does not affect bone density, body composition, or psychological measures in postmenopausal women. J Clin Endocrinol Metab. 2001;86(4):1496–503. https://pubmed.ncbi.nlm.nih.gov/11297574/
Levine ME, Suarez JA, Brandhorst S, et al. Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 2014;19(3):407–17. https://pubmed.ncbi.nlm.nih.gov/24606898/
Allen NE, Appleby PN, Davey GK, Kaaks R, Rinaldi S, Key TJ. The associations of diet with serum insulin-like growth factor I and its main binding proteins in 292 women meat-eaters, vegetarians, and vegans. Cancer Epidemiol Biomarkers Prev. 2002;11(11):1441–8. https://pubmed.ncbi.nlm.nih.gov/12433724/
Crimarco A, Springfield S, Petlura C, et al. A randomized crossover trial on the effect of plant-based compared with animal-based meat on trimethylamine-N-oxide and cardiovascular disease risk factors in generally healthy adults: Study With Appetizing Plantfood – Meat Eating Alternative Trial (SWAP-MEAT). Am J Clin Nutr. 2020;112(5):1188–99. https://pubmed.ncbi.nlm.nih.gov/32780794/
Arjmandi BH, Khalil DA, Smith BJ, et al. Soy protein has a greater effect on bone in postmenopausal women not on hormone replacement therapy, as evidenced by reducing bone resorption and urinary calcium excretion. J Clin Endocrinol Metab. 2003;88(3):1048–54. https://pubmed.ncbi.nlm.nih.gov/12629084/
Khalil DA, Lucas EA, Juma S, Smith BJ, Payton ME, Arjmandi BH. Soy protein supplementation increases serum insulin-like growth factor-I in young and old men but does not affect markers of bone metabolism. J Nutr. 2002;132(9):2605–8. https://pubmed.ncbi.nlm.nih.gov/12221217/
Maskarinec G, Takata Y, Murphy SP, Franke AA, Kaaks R. Insulin-like growth factor-1 and binding protein-3 in a 2-year soya intervention among premenopausal women. Br J Nutr. 2005;94(3):362–7. https://pubmed.ncbi.nlm.nih.gov/16176606/
Messina M, Magee P. Does soy protein affect circulating levels of unbound IGF-1? Eur J Nutr. 2018;57(2):423–32. https://pubmed.ncbi.nlm.nih.gov/28434035/
Nachvak SM, Moradi S, Anjom-Shoae J, et al. Soy, soy isoflavones, and protein intake in relation to mortality from all causes, cancers, and cardiovascular diseases: a systematic review and dose-response meta-analysis of prospective cohort studies. J Acad Nutr Diet. 2019;119(9):1483–1500.e17. https://pubmed.ncbi.nlm.nih.gov/31278047/
Applegate CC, Rowles JL III, Ranard KM, Jeon S, Erdman JW Jr. Soy consumption and the risk of prostate cancer: an updated systematic review and meta-analysis. Nutrients. 2018;10(1):40. https://pubmed.ncbi.nlm.nih.gov/29300347/
Willcox DC, Willcox BJ, Todoriki H, Suzuki M. The Okinawan diet: health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J Am Coll Nutr. 2009;28(sup4):500S-16S. https://pubmed.ncbi.nlm.nih.gov/20234038/
Lousuebsakul-Matthews V, Thorpe DL, Knutsen R, Beeson WL, Fraser GE, Knutsen SF. Legumes and meat analogues consumption are associated with hip fracture risk independently of meat intake among Caucasian men and women: the Adventist Health Study-2. Public Health Nutr. 2014;17(10):2333–43. https://pubmed.ncbi.nlm.nih.gov/24103482/
Mazidi M, Katsiki N, Mikhailidis DP, et al. Lower carbohydrate diets and all-cause and cause-specific mortality: a population-based cohort study and pooling of prospective studies. Eur Heart J. 2019;40(34):2870–9. https://pubmed.ncbi.nlm.nih.gov/31004146/
Fung TT, van Dam RM, Hankinson SE, Stampfer M, Willett WC, Hu FB. Low-carbohydrate diets and all-cause and cause-specific mortality: two cohort studies. Ann Intern Med. 2010;153(5):289–98. https://pubmed.ncbi.nlm.nih.gov/20820038/
Sun Y, Liu B, Snetselaar LG, et al. Association of major dietary protein sources with all-cause and cause-specific mortality: prospective cohort study. J Am Heart Assoc. 2021;10(5):e015553. https://pubmed.ncbi.nlm.nih.gov/33624505/
Huang J, Liao LM, Weinstein SJ, Sinha R, Graubard BI, Albanes D. Association between plant and animal protein intake and overall and cause-specific mortality. JAMA Intern Med. 2020;180(9):1173–84. https://pubmed.ncbi.nlm.nih.gov/32658243/
Levine ME, Suarez JA, Brandhorst S, et al. Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 2014;19(3):407–17. https://pubmed.ncbi.nlm.nih.gov/24606898/
Wu S. Meat and cheese may be as bad as smoking. USC News. https://news.usc.edu/59199/meat-and-cheese-may-be-as-bad-for-you-as-smoking/. Published March 4, 2014. Accessed June 11, 2021.; https://news.usc.edu/59199/meat-and-cheese-may-be-as-bad-for-you-as-smoking/
Wu S. Meat and cheese may be as bad as smoking. USC News. https://news.usc.edu/59199/meat-and-cheese-may-be-as-bad-for-you-as-smoking/. Published March 4, 2014. Accessed June 11, 2021.; https://news.usc.edu/59199/meat-and-cheese-may-be-as-bad-for-you-as-smoking/
Spiegelhalter D. Using speed of ageing and “microlives” to communicate the effects of lifetime habits and environment. BMJ. 2012;345:e8223. https://pubmed.ncbi.nlm.nih.gov/23247978/
Sample I. Diets high in meat, eggs and dairy could be as harmful to health as smoking. Guardian. https://www.theguardian.com/science/2014/mar/04/animal-protein-diets-smoking-meat-eggs-dairy. Published March 5, 2014. Accessed June 9, 2021.; https://www.theguardian.com/science/2014/mar/04/animal-protein-diets-smoking-meat-eggs-dairy
Philip Morris, Europe. Second-hand tobacco smoke in perspective. What risks do you take? Philip Morris Records; Master Settlement Agreement. UCSF Industry Documents Library. https://www.industrydocuments.ucsf.edu/docs/pkdl0113. Produced 1994. Accessed February 11 https://www.industrydocuments.ucsf.edu/docs/pkdl0113
Ngo TH, Barnard RJ, Tymchuk CN, Cohen P, Aronson WJ. Effect of diet and exercise on serum insulin, IGF-I, and IGFBP-1 levels and growth of LNCaP cells in vitro (United States). Cancer Causes Control. 2002;13(10):929–35. https://pubmed.ncbi.nlm.nih.gov/12588089/
Soliman S, Aronson WJ, Barnard RJ. Analyzing serum-stimulated prostate cancer cell lines after low-fat, high-fiber diet and exercise intervention. Evid Based Complement Alternat Med. 2011;2011:529053. https://pubmed.ncbi.nlm.nih.gov/19376839/
Barnard RJ, Ngo TH, Leung PS, Aronson WJ, Golding LA. A low-fat diet and/or strenuous exercise alters the IGF axis in vivo and reduces prostate tumor cell growth in vitro. Prostate. 2003;56(3):201–6. https://pubmed.ncbi.nlm.nih.gov/12772189/
Ornish D, Weidner G, Fair WR, et al. Intensive lifestyle changes may affect the progression of prostate cancer. J Urol. 2005;174(3):1065–9. https://pubmed.ncbi.nlm.nih.gov/16094059/
Ornish D, Magbanua MJM, Weidner G, et al. Changes in prostate gene expression in men undergoing an intensive nutrition and lifestyle intervention. Proc Natl Acad Sci U S A. 2008;105(24):8369–74. https://pubmed.ncbi.nlm.nih.gov/18559852/
Yang M, Kenfield SA, Van Blarigan EL, et al. Dairy intake after prostate cancer diagnosis in relation to disease-specific and total mortality. Int J Cancer. 2015;137(10):2462–9. https://pubmed.ncbi.nlm.nih.gov/25989745/
Tantamango-Bartley Y, Jaceldo-Siegl K, Fan J, Fraser G. Vegetarian diets and the incidence of cancer in a low-risk population. Cancer Epidemiol Biomarkers Prev. 2013;22(2):286–94. https://pubmed.ncbi.nlm.nih.gov/23169929/
Mucci LA, Tamimi R, Lagiou P, et al. Are dietary influences on the risk of prostate cancer mediated through the insulin-like growth factor system? BJU Int. 2001;87(9):814–20. https://pubmed.ncbi.nlm.nih.gov/11412218/
Gunnell D, Oliver SE, Peters TJ, et al. Are diet – prostate cancer associations mediated by the IGF axis? A cross-sectional analysis of diet, IGF-I and IGFBP-3 in healthy middle-aged men. Br J Cancer. 2003;88(11):1682–6. https://pubmed.ncbi.nlm.nih.gov/12771980/
Walfisch S, Walfisch Y, Kirilov E, et al. Tomato lycopene extract supplementation decreases insulin-like growth factor-I levels in colon cancer patients. Eur J Cancer Prev. 2007;16(4):298–303. https://pubmed.ncbi.nlm.nih.gov/17554202/
Xie Z, Yang F. The effects of lycopene supplementation on serum insulin-like growth factor 1 (IGF-1) levels and cardiovascular disease: a dose-response meta-analysis of clinical trials. Complement Ther Med. 2021;56:102632. https://pubmed.ncbi.nlm.nih.gov/33259908/
Rickard SE, Yuan YV, Thompson LU. Plasma insulin-like growth factor I levels in rats are reduced by dietary supplementation of flaxseed or its lignan secoisolariciresinol diglycoside. Cancer Lett. 2000;161(1):47–55. https://pubmed.ncbi.nlm.nih.gov/11078912/
Sturgeon SR, Volpe SL, Puleo E, et al. Dietary intervention of flaxseed: effect on serum levels of IGF-1, IGF-BP3, and C-peptide. Nutr Cancer. 2011;63(3):376–80. https://pubmed.ncbi.nlm.nih.gov/21462084/
Zhou JR, Yu L, Mai Z, Blackburn GL. Combined inhibition of estrogen-dependent human breast carcinoma by soy and tea bioactive components in mice. Int J Cancer. 2004;108(1):8–14. https://pubmed.ncbi.nlm.nih.gov/14618609/
Biernacka KM, Holly JMP, Martin RM, et al. Effect of green tea and lycopene on the insulin-like growth factor system: the ProDiet randomized controlled trial. Eur J Cancer Prev. 2019;28(6):569–75. https://pubmed.ncbi.nlm.nih.gov/30921005/
Samavat H, Wu AH, Ursin G, et al. Green tea catechin extract supplementation does not influence circulating sex hormones and insulin-like growth factor axis proteins in a randomized controlled trial of postmenopausal women at high risk of breast cancer. J Nutr. 2019;149(4):619–27. https://pubmed.ncbi.nlm.nih.gov/30926986/
Teas J, Irhimeh MR, Druker S, et al. Serum IGF-1 concentrations change with soy and seaweed supplements in healthy postmenopausal American women. Nutr Cancer. 2011;63(5):743–8. https://pubmed.ncbi.nlm.nih.gov/21711174/
Burgers AMG, Biermasz NR, Schoones JW, et al. Meta-analysis and dose-response metaregression: circulating insulin-like growth factor I (IGF-I) and mortality. J Clin Endocrinol Metab. 2011;96(9):2912–20. https://pubmed.ncbi.nlm.nih.gov/21795450/
LeRoith D. IGF-I: panacea or poison? J Clin Endocrinol Metab. 2010;95(10):4549–51. https://pubmed.ncbi.nlm.nih.gov/20926541/
Zhang WB, Aleksic S, Gao T, et al. Insulin-like growth factor-1 and IGF binding proteins predict all-cause mortality and morbidity in older adults. Cells. 2020;9(6):1368. https://pubmed.ncbi.nlm.nih.gov/32492897/
Larsson SC, Michaëlsson K, Burgess S. IGF-1 and cardiometabolic diseases: a Mendelian randomisation study. Diabetologia. 2020;63(9):1775–82. https://pubmed.ncbi.nlm.nih.gov/32548700/
Hartley A, Sanderson E, Paternoster L, et al. Mendelian randomization provides evidence for a causal effect of higher serum IGF-1 concentration on risk of hip and knee osteoarthritis. Rheumatology (Oxford). 2020;60(4):1676–86. https://pubmed.ncbi.nlm.nih.gov/33027520/
Larsson SC, Michaëlsson K, Burgess S. IGF-1 and cardiometabolic diseases: a Mendelian randomisation study. Diabetologia. 2020;63(9):1775–82. https://pubmed.ncbi.nlm.nih.gov/32548700/
Fan M, Li Y, Wang C, et al. Dietary protein consumption and the risk of type 2 diabetes: adose-response [sic] meta-analysis of prospective studies. Nutrients. 2019;11(11):2783. https://pubmed.ncbi.nlm.nih.gov/31731672/
Teumer A, Qi Q, Nethander M, et al. Genomewide meta-analysis identifies loci associated with IGF-I and IGFBP-3 levels with impact on age-related traits. Aging Cell. 2016;15(5):811–24. https://pubmed.ncbi.nlm.nih.gov/27329260/
Milman S, Atzmon G, Huffman DM, et al. Low insulin-like growth factor-1 level predicts survival in humans with exceptional longevity. Aging Cell. 2014;13(4):769–71. https://pubmed.ncbi.nlm.nih.gov/24618355/
Pawlikowska L, Hu D, Huntsman S, et al. Association of common genetic variation in the insulin/IGF1 signaling pathway with human longevity. Aging Cell. 2009;8(4):460–72. https://pubmed.ncbi.nlm.nih.gov/19489743/
Fontana L, Cummings NE, Arriola Apelo SI, et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 2016;16(2):520–30. https://pubmed.ncbi.nlm.nih.gov/27346343/
Chainani-Wu N, Weidner G, Purnell DM, et al. Changes in emerging cardiac biomarkers after an intensive lifestyle intervention. Am J Cardiol. 2011;108(4):498–507. https://pubmed.ncbi.nlm.nih.gov/21624543/
Levine ME, Suarez JA, Brandhorst S, et al. Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 2014;19(3):407–17. https://pubmed.ncbi.nlm.nih.gov/24606898/
Werner H, Laron Z. Role of the GH-IGF1 system in progression of cancer. Mol Cell Endocrinol. 2020;518:111003. https://pubmed.ncbi.nlm.nih.gov/32919021/
McCarty MF. A low-fat, whole-food vegan diet, as well as other strategies that down-regulate IGF-I activity, may slow the human aging process. Med Hypotheses. 2003;60(6):784–92. https://pubmed.ncbi.nlm.nih.gov/12699704/
Longo VD, Lieber MR, Vijg J. Turning anti-ageing genes against cancer. Nat Rev Mol Cell Biol. 2008;9(11):903–10. https://pubmed.ncbi.nlm.nih.gov/18946478/
McCarty MF. GCN2 and FGF21 are likely mediators of the protection from cancer, autoimmunity, obesity, and diabetes afforded by vegan diets. Med Hypotheses. 2014;83(3):365–71. https://pubmed.ncbi.nlm.nih.gov/25015767/
Piper MDW, Soultoukis GA, Blanc E, et al. Matching dietary amino acid balance to the in silico – translated exome optimizes growth and reproduction without cost to lifespan. Cell Metab. 2017;25(3):610–21. https://pubmed.ncbi.nlm.nih.gov/28273481/
Slavich GM. Understanding inflammation, its regulation, and relevance for health: a top scientific and public priority. Brain Behav Immun. 2015;45:13–4. https://pubmed.ncbi.nlm.nih.gov/25449576/
Egger G. In search of a germ theory equivalent for chronic disease. Prev Chronic Dis. 2012;9:E95. https://pubmed.ncbi.nlm.nih.gov/22575080/
Rubio-Ruiz ME, Peredo-Escárcega AE, Cano-Martínez A, Guarner-Lans V. An evolutionary perspective of nutrition and inflammation as mechanisms of cardiovascular disease. Int J Evol Biol. 2015:2015:179791.; https://pubmed.ncbi.nlm.nih.gov/26693381/
Rogers J. The inflammatory response in Alzheimer’s disease. J Periodontol. 2008;79(8 Suppl):1535–43. https://pubmed.ncbi.nlm.nih.gov/18673008/
Egger G. In search of a germ theory equivalent for chronic disease. Prev Chronic Dis. 2012;9:E95. https://pubmed.ncbi.nlm.nih.gov/22575080/
Ridker PM. C-reactive protein: a simple test to help predict risk of heart attack and stroke. Circulation. 2003;108(12):e81–5. https://pubmed.ncbi.nlm.nih.gov/14504253/
Bray C, Bell LN, Liang H, et al. Erythrocyte sedimentation rate and C-reactive protein measurements and their relevance in clinical medicine. WMJ. 2016;115(6):317–21. https://pubmed.ncbi.nlm.nih.gov/29094869/
Ridker PM. C-reactive protein: a simple test to help predict risk of heart attack and stroke. Circulation. 2003;108(12):e81–5. https://pubmed.ncbi.nlm.nih.gov/14504253/
Bottazzi B, Riboli E, Mantovani A. Aging, inflammation and cancer. Semin Immunol. 2018;40:74–82. https://pubmed.ncbi.nlm.nih.gov/30409538/
National Center for Injury Prevention and Control, CDC using WISQARSÔ.10 leading causes of death by age group, United States—2018. Centers for Disease Control and Prevention. https://www.cdc.gov/injury/images/lc-charts/leading_causes_of_death_by_age_group_2018_1100w850h.jpg. Accessed June 29, 2021.; https://www.cdc.gov/injury/images/lc-charts/leading_causes_of_death_by_age_group_2018_1100w850h.jpg
Weyh C, Krüger K, Strasser B. Physical activity and diet shape the immune system during aging. Nutrients. 2020;12(3):622. https://pubmed.ncbi.nlm.nih.gov/32121049/
Fagiolo U, Cossarizza A, Scala E, et al. Increased cytokine production in mononuclear cells of healthy elderly people. Eur J Immunol. 1993;23(9):2375–8. https://pubmed.ncbi.nlm.nih.gov/8370415/
Fulop T, Larbi A, Dupuis G, et al. Immunosenescence and inflamm-aging as two sides of the same coin: friends or foes? Front Immunol. 2018;8:1960. https://pubmed.ncbi.nlm.nih.gov/29375577/
Cevenini E, Monti D, Franceschi C. Inflamm-ageing. Curr Opin Clin Nutr Metab Care. 2013;16(1):14–20. https://pubmed.ncbi.nlm.nih.gov/23132168/
Franceschi C, Bonafè M, Valensin S, et al. Inflamm-aging: an evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908(1):244–54. https://pubmed.ncbi.nlm.nih.gov/10911963/
Tang Y, Fung E, Xu A, Lan HY. C-reactive protein and ageing. Clin Exp Pharmacol Physiol. 2017;44(S1):9–14. https://pubmed.ncbi.nlm.nih.gov/28378496/
Tait JL, Duckham RL, Milte CM, Main LC, Daly RM. Associations between inflammatory and neurological markers with quality of life and well-being in older adults. Exp Gerontol. 2019;125:110662. https://pubmed.ncbi.nlm.nih.gov/31323254/
Tang Y, Fung E, Xu A, Lan HY. C-reactive protein and ageing. Clin Exp Pharmacol Physiol. 2017;44(S1):9–14. https://pubmed.ncbi.nlm.nih.gov/28378496/
Rajasekaran S, Tangavel C, Anand SV KS, et al. Inflammaging determines health and disease in lumbar discs – evidence from differing proteomic signatures of healthy, aging, and degenerating discs. Spine J. 2020;20(1):48–59. https://pubmed.ncbi.nlm.nih.gov/31125691/
Pedersen BK. Anti-inflammation – just another word for anti-ageing? J Physiol. 2009;587(Pt 23):5515. https://pubmed.ncbi.nlm.nih.gov/19959548/
Barron E, Lara J, White M, Mathers JC. Blood-borne biomarkers of mortality risk: systematic review of cohort studies. PLoS ONE. 2015;10(6):e0127550. https://pubmed.ncbi.nlm.nih.gov/26039142/
Bottazzi B, Riboli E, Mantovani A. Aging, inflammation and cancer. Semin Immunol. 2018;40:74–82. https://pubmed.ncbi.nlm.nih.gov/30409538/
Franceschi C, Bonafè M, Valensin S, et al. Inflamm-aging: an evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908(1):244–54. https://pubmed.ncbi.nlm.nih.gov/10911963/
Puzianowska-Kuznicka M, Owczarz M, Wieczorowska-Tobis K, et al. Interleukin-6 and C-reactive protein, successful aging, and mortality: the PolSenior study. Immun Ageing. 2016;13:21. https://pubmed.ncbi.nlm.nih.gov/27274758/
Franceschi C, Ostan R, Santoro A. Nutrition and inflammation: are centenarians similar to individuals on calorie-restricted diets? Annu Rev Nutr. 2018;38:329–56. https://pubmed.ncbi.nlm.nih.gov/29852087/
Bonafè M, Olivieri F, Cavallone L, et al. A gender – dependent genetic predisposition to produce high levels of IL-6 is detrimental for longevity. Eur J Immunol. 2001;31(8):2357–61. https://pubmed.ncbi.nlm.nih.gov/11500818/
Man MQ, Elias PM. Could inflammaging and its sequelae be prevented or mitigated? Clin Interv Aging. 2019;14:2301–4. https://pubmed.ncbi.nlm.nih.gov/31920294/
Man MQ, Elias PM. Could inflammaging and its sequelae be prevented or mitigated? Clin Interv Aging. 2019;14:2301–4. https://pubmed.ncbi.nlm.nih.gov/31920294/
Hu L, Mauro TM, Dang E, et al. Epidermal dysfunction leads to an age-associated increase in levels of serum inflammatory cytokines. J Invest Dermatol. 2017;137(6):1277–85. https://pubmed.ncbi.nlm.nih.gov/28115059/
Ye L, Mauro TM, Dang E, et al. Topical applications of an emollient reduce circulating pro-inflammatory cytokine levels in chronically aged humans: a pilot clinical study. J Eur Acad Dermatol Venereol. 2019;33(11):2197–201. https://pubmed.ncbi.nlm.nih.gov/30835878/
Arai Y, Martin-Ruiz CM, Takayama M, et al. Inflammation, but not telomere length, predicts successful ageing at extreme old age: a longitudinal study of semi-supercentenarians. EBioMedicine. 2015;2(10):1549–58. https://pubmed.ncbi.nlm.nih.gov/26629551/
Furman D, Campisi J, Verdin E, et al. Chronic inflammation in the etiology of disease across the life span. Nat Med. 2019;25(12):1822–32. https://pubmed.ncbi.nlm.nih.gov/31806905/
Chambers ES, Akbar AN. Can blocking inflammation enhance immunity during aging? J Allergy Clin Immunol. 2020;145(5):1323–31. https://pubmed.ncbi.nlm.nih.gov/32386656/
Franceschi C, Garagnani P, Vitale G, Capri M, Salvioli S. Inflammaging and ‘garb-aging.’ Trends Endocrinol. Metab. 2017;28(3):199–212. https://pubmed.ncbi.nlm.nih.gov/27789101/
Monti D, Ostan R, Borelli V, Castellani G, Franceschi C. Inflammaging and human longevity in the omics era. Mech Ageing Dev. 2017;165(Pt B):129–38. https://pubmed.ncbi.nlm.nih.gov/28038993/
Meydani SN, Das SK, Pieper CF, et al. Long-term moderate calorie restriction inhibits inflammation without impairing cell-mediated immunity: a randomized controlled trial in non-obese humans. Aging (Albany NY). 2016;8(7):1416–31. https://pubmed.ncbi.nlm.nih.gov/27410480/
Choi J, Joseph L, Pilote L. Obesity and C-reactive protein in various populations: a systematic review and meta-analysis. Obes Rev. 2013;14(3):232–44. https://pubmed.ncbi.nlm.nih.gov/23171381/
Ellulu MS, Patimah I, Khaza’ai H, Rahmat A, Abed Y. Obesity and inflammation: the linking mechanism and the complications. Arch Med Sci. 2017;13(4):851–63. https://pubmed.ncbi.nlm.nih.gov/28721154/
Pasarica M, Sereda OR, Redman LM, et al. Reduced adipose tissue oxygenation in human obesity: evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes. 2009;58(3):718–25. https://pubmed.ncbi.nlm.nih.gov/19074987/
Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–808. https://pubmed.ncbi.nlm.nih.gov/14679176/
Cinti S, Mitchell G, Barbatelli G, et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J Lipid Res. 2005;46(11):2347–55. https://pubmed.ncbi.nlm.nih.gov/16150820/
Bays HE, González-Campoy JM, Bray GA, et al. Pathogenic potential of adipose tissue and metabolic consequences of adipocyte hypertrophy and increased visceral adiposity. Expert Rev Cardiovasc Ther. 2008;6(3):343–68. https://pubmed.ncbi.nlm.nih.gov/18327995/
Welsh P, Polisecki E, Robertson M, et al. Unraveling the directional link between adiposity and inflammation: a bidirectional Mendelian randomization approach. J Clin Endocrinol Metab. 2010;95(1):93–9. https://pubmed.ncbi.nlm.nih.gov/28199503/
Timpson NJ, Nordestgaard BG, Harbord RM, et al. C-reactive protein levels and body mass index: elucidating direction of causation through reciprocal Mendelian randomization. Int J Obes (Lond). 2011;35(2):300–8. https://pubmed.ncbi.nlm.nih.gov/20714329/
Chung S, Parks JS. Dietary cholesterol effects on adipose tissue inflammation. Curr Opin Lipidol. 2016;27(1):19–25. https://pubmed.ncbi.nlm.nih.gov/26655292/
Chung S, Cuffe H, Marshall SM, et al. Dietary cholesterol promotes adipocyte hypertrophy and adipose tissue inflammation in visceral, but not in subcutaneous, fat in monkeys. Arterioscler Thromb Vasc Biol. 2014;34(9):1880–7. https://pubmed.ncbi.nlm.nih.gov/24969772/
Chung S, Parks JS. Dietary cholesterol effects on adipose tissue inflammation. Curr Opin Lipidol. 2016;27(1):19–25. https://pubmed.ncbi.nlm.nih.gov/26655292/
Chung S, Cuffe H, Marshall SM, et al. Dietary cholesterol promotes adipocyte hypertrophy and adipose tissue inflammation in visceral, but not in subcutaneous, fat in monkeys. Arterioscler Thromb Vasc Biol. 2014;34(9):1880–7. https://pubmed.ncbi.nlm.nih.gov/24969772/
Xu Z, McClure ST, Appel LJ. Dietary cholesterol intake and sources among U.S. adults: results from National Health and Nutrition Examination Surveys (NHANES), 2001–2014. Nutrients. 2018;10(6):E771. https://pubmed.ncbi.nlm.nih.gov/29903993/
Morgan-Bathke ME, Jensen MD. Preliminary evidence for reduced adipose tissue inflammation in vegetarians compared with omnivores. Nutr J. 2019;18(1):45. https://pubmed.ncbi.nlm.nih.gov/31405384/
Hegsted DM. Dietary goals – a progressive view. Am J Clin Nutr. 1978;31(9):1504–9. https://pubmed.ncbi.nlm.nih.gov/28662/
Trumbo PR, Shimakawa T. Tolerable upper intake levels for trans fat, saturated fat, and cholesterol. Nutr Rev. 2011;69(5):270–8. https://pubmed.ncbi.nlm.nih.gov/21521229/
Chambers ES, Akbar AN. Can blocking inflammation enhance immunity during aging? J Allergy Clin Immunol. 2020;145(5):1323–31. https://pubmed.ncbi.nlm.nih.gov/32386656/
Zamboni M, Nori N, Brunelli A, Zoico E. How does adipose tissue contribute to inflammageing? Exp Gerontol. 2021;143:111162. https://pubmed.ncbi.nlm.nih.gov/33253807/
Buchwald H, Avidor Y, Braunwald E, et al. Bariatric surgery: a systematic review and meta-analysis. JAMA. 2004;292(14):1724–37. https://pubmed.ncbi.nlm.nih.gov/15479938/
Rao SR. Inflammatory markers and bariatric surgery: a meta-analysis. Inflamm Res. 2012;61(8):789–807. https://pubmed.ncbi.nlm.nih.gov/22588278/
Meydani SN, Das SK, Pieper CF, et al. Long-term moderate calorie restriction inhibits inflammation without impairing cell-mediated immunity: a randomized controlled trial in non-obese humans. Aging (Albany NY). 2016;8(7):1416–31. https://pubmed.ncbi.nlm.nih.gov/27410480/
Chambers ES, Akbar AN. Can blocking inflammation enhance immunity during aging? J Allergy Clin Immunol. 2020;145(5):1323–31. https://pubmed.ncbi.nlm.nih.gov/32386656/
Egger G. In search of a germ theory equivalent for chronic disease. Prev Chronic Dis. 2012;9:E95. https://pubmed.ncbi.nlm.nih.gov/22575080/
Egger G, Dixon J. Non-nutrient causes of low-grade, systemic inflammation: support for a ‘canary in the mineshaft’ view of obesity in chronic disease. Obes Rev. 2011;12(5):339–45. https://pubmed.ncbi.nlm.nih.gov/20701689/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Ryu S, Shivappa N, Veronese N, et al. Secular trends in Dietary Inflammatory Index among adults in the United States, 1999–2014. Eur J Clin Nutr. 2019;73(10):1343–51. https://pubmed.ncbi.nlm.nih.gov/30542148/
Xu H, Sjögren P, Ärnlöv J, et al. A proinflammatory diet is associated with systemic inflammation and reduced kidney function in elderly adults. J Nutr. 2015;145(4):729–35. https://pubmed.ncbi.nlm.nih.gov/25833776/
Han YY, Forno E, Shivappa N, Wirth MD, Hébert JR, Celedón JC. The Dietary Inflammatory Index and current wheeze among children and adults in the United States. J Allergy Clin Immunol Pract. 2018;6(3):834–41. https://pubmed.ncbi.nlm.nih.gov/29426751/
Cantero I, Abete I, Babio N, et al. Dietary Inflammatory Index and liver status in subjects with different adiposity levels within the PREDIMED trial. Clin Nutr. 2018;37(5):1736–43. https://pubmed.ncbi.nlm.nih.gov/28734553/
Shivappa N, Godos J, Hébert JR, et al. Dietary Inflammatory Index and cardiovascular risk and mortality – a meta-analysis. Nutrients. 2018;10(2):200. https://pubmed.ncbi.nlm.nih.gov/29439509/
Shivappa N, Wirth MD, Hurley TG, Hébert JR. Association between the dietary inflammatory index (DII) and telomere length and C-reactive protein from the National Health and Nutrition Examination Survey—1999–2002. Mol Nutr Food Res. 2017;61(4). https://pubmed.ncbi.nlm.nih.gov/29675557/
García-Calzón S, Zalba G, Ruiz-Canela M, et al. Dietary inflammatory index and telomere length in subjects with a high cardiovascular disease risk from the PREDIMED-NAVARRA study: cross-sectional and longitudinal analyses over 5 y. Am J Clin Nutr. 2015;102(4):897–904. https://pubmed.ncbi.nlm.nih.gov/26354530/
Shivappa N, Stubbs B, Hébert JR, et al. The relationship between the Dietary Inflammatory Index and incident frailty: a longitudinal cohort study. J Am Med Dir Assoc. 2018;19(1):77–82. https://pubmed.ncbi.nlm.nih.gov/28943182/
Cervo MMC, Scott D, Seibel MJ, et al. Proinflammatory diet increases circulating inflammatory biomarkers and falls risk in community-dwelling older men. J Nutr. 2020;150(2):373–81. https://pubmed.ncbi.nlm.nih.gov/31665502/
Kheirouri S, Alizadeh M. Dietary inflammatory potential and the risk of neurodegenerative diseases in adults. Epidemiol Rev. 2019;41(1):109–20. https://pubmed.ncbi.nlm.nih.gov/31565731/
Phillips CM, Shivappa N, Hébert JR, Perry IJ. Dietary inflammatory index and mental health: a cross-sectional analysis of the relationship with depressive symptoms, anxiety and well-being in adults. Clin Nutr. 2018;37(5):1485–91. https://pubmed.ncbi.nlm.nih.gov/28912008/
Godos J, Ferri R, Caraci F, et al. Dietary inflammatory index and sleep quality in southern Italian adults. Nutrients. 2019;11(6):1324. https://pubmed.ncbi.nlm.nih.gov/31200445/
Shivappa N, Jackson MD, Bennett F, Hébert JR. Increased dietary inflammatory index (DII) is associated with increased risk of prostate cancer in Jamaican men. Nutr Cancer. 2015;67(6):941–8. https://pubmed.ncbi.nlm.nih.gov/29439509/
Shivappa N, Hébert JR, Jalilpiran Y, Faghih S. Association between dietary inflammatory index and prostate cancer in Shiraz province of Iran. Asian Pac J Cancer Prev. 2018;19(2):415–20. https://pubmed.ncbi.nlm.nih.gov/29479991/
Shivappa N, Miao Q, Walker M, Hébert JR, Aronson KJ. Association between a dietary inflammatory index and prostate cancer risk in Ontario, Canada. Nutr Cancer. 2017;69(6):825–32. https://pubmed.ncbi.nlm.nih.gov/28718711/
Huang WQ, Mo XF, Ye YB, et al. A higher Dietary Inflammatory Index score is associated with a higher risk of breast cancer among Chinese women: a case-control study. Br J Nutr. 2017;117(10):1358–67. https://pubmed.ncbi.nlm.nih.gov/32104043/
Shivappa N, Sandin S, Löf M, Hébert JR, Adami HO, Weiderpass E. Prospective study of dietary inflammatory index and risk of breast cancer in Swedish women. Br J Cancer. 2015;113(7):1099–103. https://pubmed.ncbi.nlm.nih.gov/26335605/
Shivappa N, Hébert JR, Zucchetto A, et al. Dietary inflammatory index and endometrial cancer risk in an Italian case-control study. Br J Nutr. 2016;115(1):138–46. https://pubmed.ncbi.nlm.nih.gov/26507451/
Shivappa N, Hébert JR, Rosato V, et al. Dietary inflammatory index and ovarian cancer risk in a large Italian case-control study. Cancer Causes Control. 2016;27(7):897–906. https://pubmed.ncbi.nlm.nih.gov/27262447/
Shivappa N, Zucchetto A, Serraino D, Rossi M, La Vecchia C, Hébert JR. Dietary inflammatory index and risk of esophageal squamous cell cancer in a case-control study from Italy. Cancer Causes Control. 2015;26(10):1439–47. https://pubmed.ncbi.nlm.nih.gov/26208592/
Shivappa N, Hébert JR, Ferraroni M, La Vecchia C, Rossi M. Association between dietary inflammatory index and gastric cancer risk in an Italian case-control study. Nutr Cancer. 2016;68(8):1262–8. https://pubmed.ncbi.nlm.nih.gov/27636679/
Shivappa N, Hébert JR, Polesel J, et al. Inflammatory potential of diet and risk for hepatocellular cancer in a case-control study from Italy. Br J Nutr. 2016;115(2):324–31. https://pubmed.ncbi.nlm.nih.gov/26556602/
Shivappa N, Bosetti C, Zucchetto A, Serraino D, La Vecchia C, Hébert JR. Dietary inflammatory index and risk of pancreatic cancer in an Italian case-control study. Br J Nutr. 2015;113(2):292–8. https://pubmed.ncbi.nlm.nih.gov/25515552/
Shivappa N, Godos J, Hébert JR, et al. Dietary inflammatory index and colorectal cancer risk – a meta-analysis. Nutrients. 2017 Sep 20;9(9):1043. https://pubmed.ncbi.nlm.nih.gov/28930191/
Shivappa N, Hébert JR, Rosato V, et al. Dietary inflammatory index and renal cell carcinoma risk in an Italian case-control study. Nutr Cancer. 2017;69(6):833–9. https://pubmed.ncbi.nlm.nih.gov/28718670/
Shivappa N, Hébert JR, Rosato V, et al. Dietary inflammatory index and risk of bladder cancer in a large Italian case-control study. Urology. 2017;100:84–9. https://pubmed.ncbi.nlm.nih.gov/27693878/
Shivappa N, Hébert JR, Taborelli M, et al. Dietary inflammatory index and non-Hodgkin lymphoma risk in an Italian case-control study. Cancer Causes Control. 2017;28(7):791–9. https://pubmed.ncbi.nlm.nih.gov/28503716/
Fowler ME, Akinyemiju TF. Meta-analysis of the association between dietary inflammatory index (DII) and cancer outcomes. Int J Cancer. 2017;141(11):2215–27. https://pubmed.ncbi.nlm.nih.gov/28795402/
Shivappa N, Hebert JR, Kivimaki M, Akbaraly T. Alternate Healthy Eating Index 2010, Dietary Inflammatory Index and risk of mortality: results from the Whitehall II cohort study and meta-analysis of previous Dietary Inflammatory Index and mortality studies. Br J Nutr. 2017;118(3):210–21. https://pubmed.ncbi.nlm.nih.gov/28831955/
Edwards MK, Shivappa N, Mann JR, Hébert JR, Wirth MD, Loprinzi PD. The association between physical activity and dietary inflammatory index on mortality risk in U.S. adults. Phys Sportsmed. 2018;46(2):249–54. https://pubmed.ncbi.nlm.nih.gov/29463180/
Shivappa N, Harris H, Wolk A, Hebert JR. Association between inflammatory potential of diet and mortality among women in the Swedish Mammography Cohort. Eur J Nutr. 2016;55(5):1891–900. https://pubmed.ncbi.nlm.nih.gov/26227485/
Shivappa N, Blair CK, Prizment AE, Jacobs DR, Steck SE, Hébert JR. Association between inflammatory potential of diet and mortality in the Iowa Women’s Health study. Eur J Nutr. 2016;55(4):1491–502. https://pubmed.ncbi.nlm.nih.gov/26130324/
Tomata Y, Shivappa N, Zhang S, et al. Dietary inflammatory index and disability-free survival in community-dwelling older adults. Nutrients. 2018;10(12):1896. https://pubmed.ncbi.nlm.nih.gov/30513971/
Garcia-Arellano A, Martínez-González MA, Ramallal R, et al. Dietary inflammatory index and all-cause mortality in large cohorts: the SUN and PREDIMED studies. Clin Nutr. 2019;38(3):1221–31. https://pubmed.ncbi.nlm.nih.gov/30651193/
Nilsson MI, Bourgeois JM, Nederveen JP, et al. Lifelong aerobic exercise protects against inflammaging and cancer. PLoS One. 2019;14(1):e0210863. https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0210863
Bautmans I, Salimans L, Njemini R, Beyer I, Lieten S, Liberman K. The effects of exercise interventions on the inflammatory profile of older adults: a systematic review of the recent literature. Exp Gerontol. 2021;146:111236. https://pubmed.ncbi.nlm.nih.gov/33453323/
Ferrer MD, Capó X, Martorell M, et al. Regular practice of moderate physical activity by older adults ameliorates their anti-inflammatory status. Nutrients. 2018;10(11):1780. https://pubmed.ncbi.nlm.nih.gov/30453505/
Piercy KL, Troiano RP, Ballard RM, et al. The Physical Activity Guidelines for Americans. JAMA. 2018;320(19):2020–8. https://pubmed.ncbi.nlm.nih.gov/30418471/
Harvard T.H. Chan School of Public Health. Top food sources of saturated fat in the U.S. https://puntocritico.com/ausajpuntocritico/documentos/The_Nutrition_Source.pdf. Accessed November 23, 2021.; https://puntocritico.com/ausajpuntocritico/documentos/The_Nutrition_Source.pdf
Exler J, Lemar L, Smith J. Fat and fatty acid content of selected foods containing trans-fatty acids: special purpose table no. 1. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/arsuserfiles/80400525/data/classics/trans_fa.pdf. Published January 1996. Accessed June 20, 2021.; https://www.ars.usda.gov/arsuserfiles/80400525/data/classics/trans_fa.pdf
Vogel RA, Corretti MC, Plotnick GD. Effect of a single high-fat meal on endothelial function in healthy subjects. Am J Cardiol. 1997;79(3):350–4. https://pubmed.ncbi.nlm.nih.gov/9036757/
Deopurkar R, Ghanim H, Friedman J, et al. Differential effects of cream, glucose, and orange juice on inflammation, endotoxin, and the expression of Toll-like receptor-4 and suppressor of cytokine signaling-3. Diabetes Care. 2010;33(5):991–7. https://pubmed.ncbi.nlm.nih.gov/20067961/
Kesteloot HE, Sasaki S. Nutrition and the aging process: a population study. Am J Geriatr Cardiol. 1994;3(2):8–19. https://pubmed.ncbi.nlm.nih.gov/11416305/
Emerson SR, Kurti SP, Harms CA, et al. Magnitude and timing of the postprandial inflammatory response to a high-fat meal in healthy adults: a systematic review. Adv Nutr. 2017;8(2):213–25. https://pubmed.ncbi.nlm.nih.gov/28298267/
Harris TB, Ferrucci L, Tracy RP, et al. Associations of elevated interleukin-6 and C-reactive protein levels with mortality in the elderly. Am J Med. 1999;106(5):506–12. https://pubmed.ncbi.nlm.nih.gov/10335721/
Jonnalagadda SS, Egan SK, Heimbach JT, et al. Fatty acid consumption pattern of Americans: 1987–1988 USDA Nationwide Food Consumption Survey. Nutr Res. 1995;15(12):1767–81. https://agris.fao.org/agris-search/search.do?recordID=US19970167025
Carta G, Murru E, Banni S, Manca C. Palmitic acid: physiological role, metabolism and nutritional implications. Front Physiol. 2017;8:902. https://pubmed.ncbi.nlm.nih.gov/29167646/
Korbecki J, Bajdak-Rusinek K. The effect of palmitic acid on inflammatory response in macrophages: an overview of molecular mechanisms. Inflamm Res. 2019;68(11):915–32. https://pubmed.ncbi.nlm.nih.gov/31363792/
Deopurkar R, Ghanim H, Friedman J, et al. Differential effects of cream, glucose, and orange juice on inflammation, endotoxin, and the expression of Toll-like receptor-4 and suppressor of cytokine signaling-3. Diabetes Care. 2010;33(5):991–7. https://pubmed.ncbi.nlm.nih.gov/20067961/
Erridge C. Accumulation of stimulants of Toll-like receptor (TLR)-2 and TLR4 in meat products stored at 5 °C. J Food Sci. 2011;76(2):H72–9. https://pubmed.ncbi.nlm.nih.gov/21535770/
Erridge C. The capacity of foodstuffs to induce innate immune activation of human monocytes in vitro is dependent on food content of stimulants of Toll-like receptors 2 and 4. Br J Nutr. 2011;105(1):15–23. https://pubmed.ncbi.nlm.nih.gov/20849668/
Deopurkar R, Ghanim H, Friedman J, et al. Differential effects of cream, glucose, and orange juice on inflammation, endotoxin, and the expression of Toll-like receptor-4 and suppressor of cytokine signaling-3. Diabetes Care. 2010;33(5):991–7. https://pubmed.ncbi.nlm.nih.gov/20067961/
Herieka M, Faraj TA, Erridge C. Reduced dietary intake of pro-inflammatory Toll-like receptor stimulants favourably modifies markers of cardiometabolic risk in healthy men. Nutr Metab Cardiovasc Dis. 2016;26(3):194–200. https://pubmed.ncbi.nlm.nih.gov/26803597/
Одна американская унция = 28,3 грамма, но в данном случае это метафора, а не точное количество. – Примеч. ред.
Wassenaar TM, Zimmermann K. Lipopolysaccharides in food, food supplements, and probiotics: should we be worried? Eur J Microbiol Immunol (Bp). 2018;8(3):63–9. https://pubmed.ncbi.nlm.nih.gov/30345085/
Ghoshal S, Witta J, Zhong J, de Villiers W, Eckhardt E. Chylomicrons promote intestinal absorption of lipopolysaccharides. J Lipid Res. 2009;50(1):90–7. https://pubmed.ncbi.nlm.nih.gov/18815435/
Ghezzal S, Postal BG, Quevrain E, et al. Palmitic acid damages gut epithelium integrity and initiates inflammatory cytokine production. Biochim Biophys Acta Mol Cell Biol Lipids. 2020;1865(2):158530. https://pubmed.ncbi.nlm.nih.gov/31647994/
Harte AL, Varma MC, Tripathi G, et al. High fat intake leads to acute postprandial exposure to circulating endotoxin in type 2 diabetic subjects. Diabetes Care. 2012;35(2):375–82. https://pubmed.ncbi.nlm.nih.gov/22210577/
Erridge C. The capacity of foodstuffs to induce innate immune activation of human monocytes in vitro is dependent on food content of stimulants of Toll-like receptors 2 and 4. Br J Nutr. 2011;105(1):15–23. https://pubmed.ncbi.nlm.nih.gov/20849668/
Cho B, Kim MS, Chao K, Lawrence K, Park B, Kim K. Detection of fecal residue on poultry carcasses by laser-induced fluorescence imaging. J Food Sci. 2009;74(3):E154–9. https://pubmed.ncbi.nlm.nih.gov/19397721/
Giombelli A, Gloria MB. Prevalence of Salmonella and Campylobacter on broiler chickens from farm to slaughter and efficiency of methods to remove visible fecal contamination. J Food Prot. 2014;77(11):1851–9. https://pubmed.ncbi.nlm.nih.gov/25364917/
Erridge C. Accumulation of stimulants of Toll-like receptor (TLR)-2 and TLR4 in meat products stored at 5 °C. J Food Sci. 2011;76(2):H72–9. https://pubmed.ncbi.nlm.nih.gov/21535770/
Erridge C. Stimulants of Toll-like receptor (TLR)-2 and TLR-4 are abundant in certain minimally-processed vegetables. Food Chem Toxicol. 2011;49(6):1464–7. https://pubmed.ncbi.nlm.nih.gov/21376773/
Tournas VH. Spoilage of vegetable crops by bacteria and fungi and related health hazards. Crit Rev Microbiol. 2005;31(1):33–44. https://pubmed.ncbi.nlm.nih.gov/15839403/
Herieka M, Faraj TA, Erridge C. Reduced dietary intake of pro-inflammatory Toll-like receptor stimulants favourably modifies markers of cardiometabolic risk in healthy men. Nutr Metab Cardiovasc Dis. 2016;26(3):194–200. https://pubmed.ncbi.nlm.nih.gov/26803597/
Herieka M, Faraj TA, Erridge C. Reduced dietary intake of pro-inflammatory Toll-like receptor stimulants favourably modifies markers of cardiometabolic risk in healthy men. Nutr Metab Cardiovasc Dis. 2016;26(3):194–200. https://pubmed.ncbi.nlm.nih.gov/26803597/
Erridge C. Stimulants of Toll-like receptor (TLR)-2 and TLR-4 are abundant in certain minimally-processed vegetables. Food Chem Toxicol. 2011;49(6):1464–7. https://pubmed.ncbi.nlm.nih.gov/21376773/
Neale EP, Tapsell LC, Guan V, Batterham MJ. The effect of nut consumption on markers of inflammation and endothelial function: a systematic review and meta-analysis of randomised controlled trials. BMJ Open. 2017;7(11):e016863. https://pubmed.ncbi.nlm.nih.gov/29170286/
Chen CYO, Holbrook M, Duess MA, et al. Effect of almond consumption on vascular function in patients with coronary artery disease: a randomized, controlled, cross-over trial. Nutr J. 2015;14:61. https://pubmed.ncbi.nlm.nih.gov/26080804/
Li Z, Wong A, Henning SM, et al. Hass avocado modulates postprandial vascular reactivity and postprandial inflammatory responses to a hamburger meal in healthy volunteers. Food Funct. 2013;4(3):384–91. https://pubmed.ncbi.nlm.nih.gov/23196671/
Haskins CP, Henderson G, Champ CE. Meat, eggs, full-fat dairy, and nutritional boogeymen: does the way in which animals are raised affect health differently in humans? Crit Rev Food Sci Nutr. 2019;59(17):2709–19. https://pubmed.ncbi.nlm.nih.gov/29672133/
Eaton SB. Humans, lipids and evolution. Lipids. 1992;27(10):814–20. https://pubmed.ncbi.nlm.nih.gov/1435101/
Arya F, Egger S, Colquhoun D, Sullivan D, Pal S, Egger G. Differences in postprandial inflammatory responses to a ‘modern’ v. traditional meat meal: a preliminary study. Br J Nutr. 2010;104(5):724–8. https://pubmed.ncbi.nlm.nih.gov/20377925/
Wang Y, Lehane C, Ghebremeskel K, et al. Modern organic and broiler chickens sold for human consumption provide more energy from fat than protein. Public Health Nutr. 2010;13(3):400–8. https://pubmed.ncbi.nlm.nih.gov/19728900/
Kollander B, Widemo F, Ågren E, Larsen EH, Löschner K. Detection of lead nanoparticles in game meat by single particle ICP-MS following use of lead-containing bullets. Anal Bioanal Chem. 2017;409(7):1877–85. https://pubmed.ncbi.nlm.nih.gov/27966171/
Metryka E, Chibowska K, Gutowska I, et al. Lead (Pb) exposure enhances expression of factors associated with inflammation. Int J Mol Sci. 2018;19(6):1813. https://pubmed.ncbi.nlm.nih.gov/29925772/
Хронически повышенный уровень LPS, вызванный высококалорийной диетой. – Примеч. ред.
Harte AL, Varma MC, Tripathi G, et al. High fat intake leads to acute postprandial exposure to circulating endotoxin in type 2 diabetic subjects. Diabetes Care. 2012;35(2):375–82. https://pubmed.ncbi.nlm.nih.gov/22210577/
National Cancer Institute. Identification of top food sources of various dietary components. Epidemiology and Genomics Research Program website. https://epi.grants.cancer.gov/diet/foodsources. Updated November 30, 2019. Accessed June 20, 2021.; https://epi.grants.cancer.gov/diet/foodsources
Ghanim H, Batra M, Abuaysheh S, et al. Antiinflammatory and ROS suppressive effects of the addition of fiber to a high-fat high-calorie meal. J Clin Endocrinol Metab. 2017;102(3):858–69. https://pubmed.ncbi.nlm.nih.gov/27906549/
Simon AH, Lima PR, Almerinda M, Alves VF, Bottini PV, de Faria JB. Renal haemodynamic responses to a chicken or beef meal in normal individuals. Nephrol Dial Transplant. 1998;13(9):2261–4. https://pubmed.ncbi.nlm.nih.gov/9761506/
Kontessis P, Jones S, Dodds R, et al. Renal, metabolic and hormonal responses to ingestion of animal and vegetable proteins. Kidney Int. 1990 Jul;38(1):136–44. https://pubmed.ncbi.nlm.nih.gov/2166857/
Liu Z, Ho SC, Chen Y, Tang N, Woo J. Effect of whole soy and purified isoflavone daidzein on renal function – a 6-month randomized controlled trial in equol-producing postmenopausal women with prehypertension. Clin Biochem. 2014;47(13–14):1250–6. https://pubmed.ncbi.nlm.nih.gov/24877660/
Fioretto P, Trevisan R, Valerio A, et al. Impaired renal response to a meat meal in insulin-dependent diabetes: role of glucagon and prostaglandins. Am J Physiol. 1990;258(3 Pt 2):F675–83. https://pubmed.ncbi.nlm.nih.gov/2316671/
N-гликолилнейраминовая кислота. – Примеч. ред.
Varki A. Are humans prone to autoimmunity? Implications from evolutionary changes in hominin sialic acid biology. J Autoimmun. 2017;83:134–42. https://pubmed.ncbi.nlm.nih.gov/28755952/
Pham T, Gregg CJ, Karp F, et al. Evidence for a novel human-specific xeno-auto-antibody response against vascular endothelium. Blood. 2009;114(25):5225–35. https://pubmed.ncbi.nlm.nih.gov/19828701/
Alisson-Silva F, Kawanishi K, Varki A. Human risk of diseases associated with red meat intake: analysis of current theories and proposed role for metabolic incorporation of a non-human sialic acid. Mol Aspects Med. 2016;51:16–30. https://pubmed.ncbi.nlm.nih.gov/27421909/
Peri S, Kulkarni A, Feyertag F, Berninsone PM, Alvarez-Ponce D. Phylogenetic distribution of CMP-Neu5Ac hydroxylase (CMAH), the enzyme synthetizing the proinflammatory human xenoantigen Neu5Gc. Genome Biol Evol. 2018;10(1):207–19. https://pubmed.ncbi.nlm.nih.gov/29206915/
Samraj AN, Pearce OMT, Läubli H, et al. A red meat-derived glycan promotes inflammation and cancer progression. Proc Natl Acad Sci U S A. 2015;112(2):542–7. https://pubmed.ncbi.nlm.nih.gov/25548184/
Peri S, Kulkarni A, Feyertag F, Berninsone PM, Alvarez-Ponce D. Phylogenetic distribution of CMP-Neu5Ac hydroxylase (CMAH), the enzyme synthetizing the proinflammatory human xenoantigen Neu5Gc. Genome Biol Evol. 2018;10(1):207–19. https://pubmed.ncbi.nlm.nih.gov/29206915/
Jahan M, Thomson PC, Wynn PC, Wang B. The non-human glycan, N-glycolylneuraminic acid (Neu5Gc), is not expressed in all organs and skeletal muscles of nine animal species. Food Chem. 2021;343:128439. https://pubmed.ncbi.nlm.nih.gov/33127222/
Peri S, Kulkarni A, Feyertag F, Berninsone PM, Alvarez-Ponce D. Phylogenetic distribution of CMP-Neu5Ac hydroxylase (CMAH), the enzyme synthetizing the proinflammatory human xenoantigen Neu5Gc. Genome Biol Evol. 2018;10(1):207–19. https://pubmed.ncbi.nlm.nih.gov/29206915/
Jahan M, Thomson PC, Wynn PC, Wang B. The non-human glycan, N-glycolylneuraminic acid (Neu5Gc), is not expressed in all organs and skeletal muscles of nine animal species. Food Chem. 2021;343:128439. https://pubmed.ncbi.nlm.nih.gov/33127222/
Alisson-Silva F, Kawanishi K, Varki A. Human risk of diseases associated with red meat intake: analysis of current theories and proposed role for metabolic incorporation of a non-human sialic acid. Mol Aspects Med. 2016;51:16–30. https://pubmed.ncbi.nlm.nih.gov/27421909/
MacGregor GA, Markandu ND, Best FE, et al. Double-blind randomised crossover trial of moderate sodium restriction in essential hypertension. Lancet. 1982;1(8268):351–5. https://pubmed.ncbi.nlm.nih.gov/6120346/
Yi B, Titze J, Rykova M, et al. Effects of dietary salt levels on monocytic cells and immune responses in healthy human subjects: a longitudinal study. Transl Res. 2015;166(1):103–10. https://pubmed.ncbi.nlm.nih.gov/25497276/
Mickleborough TD, Lindley MR, Ray S. Dietary salt, airway inflammation, and diffusion capacity in exercise-induced asthma. Med Sci Sports Exerc. 2005;37(6):904–14. https://pubmed.ncbi.nlm.nih.gov/15947713/
Farez MF, Fiol MP, Gaitán MI, Quintana FJ, Correale J. Sodium intake is associated with increased disease activity in multiple sclerosis. J Neurol Neurosurg Psychiatry. 2015;86(1):26–31. https://pubmed.ncbi.nlm.nih.gov/28556498/
Krajina I, Stupin A, Šola M, Mihalj M. Oxidative stress induced by high salt diet – possible implications for development and clinical manifestation of cutaneous inflammation and endothelial dysfunction in Psoriasis vulgaris. Antioxidants (Basel). 2022;11(7):1269. https://pubmed.ncbi.nlm.nih.gov/35883760/
Carranza-León DA, Oeser A, Marton A, et al. Tissue sodium content in patients with systemic lupus erythematosus: association with disease activity and markers of inflammation. Lupus. 2020;29(5):455–62. https://pubmed.ncbi.nlm.nih.gov/32070186/
Jung SM, Kim Y, Kim J, et al. Sodium chloride aggravates arthritis via Th17 polarization. Yonsei Med J. 2019;60(1):88–97. https://pubmed.ncbi.nlm.nih.gov/30554495/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
United States Department of Health and Human Services, United States Department of Agriculture. Appendix 13. Food sources of dietary fiber. In: 2015–2020 Dietary Guidelines for Americans. 8th ed. DietaryGuidelines.gov. 2015:114–8.; https://health.gov/our-work/nutrition-physical-activity/dietary-guidelines/previous-dietary-guidelines/2015
Hostetler GL, Ralston RA, Schwartz SJ. Flavones: food sources, bioavailability, metabolism, and bioactivity. Adv Nutr. 2017;8(3):423–35. https://pubmed.ncbi.nlm.nih.gov/28507008/
Haytowitz DB, Bhagwat S, Harnly J, Holden JM, Gebhardt SE. Sources of flavonoids in the U.S. diet using USDA’s updated database on the flavonoid content of selected foods. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/ARSUserFiles/80400525/Articles/AICR06_flav.pdf. Published 2006. Accessed July 20, 2021.; https://www.ars.usda.gov/ARSUserFiles/80400525/Articles/AICR06_flav.pdf
Hostetler GL, Ralston RA, Schwartz SJ. Flavones: food sources, bioavailability, metabolism, and bioactivity. Adv Nutr. 2017;8(3):423–35. https://pubmed.ncbi.nlm.nih.gov/28507008/
Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L. The role of short-chain fatty acids in health and disease. In: Alt FW, ed. Advances in Immunology. Vol 121. Academic Press, Elsevier; 2014:91–119. https://pubmed.ncbi.nlm.nih.gov/24388214/
Pukatzki S, Provenzano D. Vibrio cholerae as a predator: lessons from evolutionary principles. Front Microbiol. 2013;4. https://pubmed.ncbi.nlm.nih.gov/24368907/
Chang PV, Hao L, Offermanns S, Medzhitov R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A. 2014;111(6):2247–52. https://pubmed.ncbi.nlm.nih.gov/24390544/
McRorie JW. Evidence-based approach to fiber supplements and clinically meaningful health benefits, part 1: what to look for and how to recommend an effective fiber therapy. Nutr Today. 2015;50(2):82–9. https://pubmed.ncbi.nlm.nih.gov/25972618/
Nilsson AC, Östman EM, Knudsen KEB, Holst JJ, Björck IME. A cereal-based evening meal rich in indigestible carbohydrates increases plasma butyrate the next morning. J Nutr. 2010;140(11):1932–6. https://pubmed.ncbi.nlm.nih.gov/20810606/
Meijer K, de Vos P, Priebe MG. Butyrate and other short-chain fatty acids as modulators of immunity: what relevance for health? Curr Opin Clin Nutr Metab Care. 2010;13(6):715–21. https://pubmed.ncbi.nlm.nih.gov/20823773/
Dai Z, Lu N, Niu J, Felson DT, Zhang Y. Dietary fiber intake in relation to knee pain trajectory. Arthritis Care Res (Hoboken). 2017;69(9):1331–9. https://pubmed.ncbi.nlm.nih.gov/27899003/
Dai Z, Niu J, Zhang Y, Jacques P, Felson DT. Dietary intake of fibre and risk of knee osteoarthritis in two US prospective cohorts [published correction appears in Ann Rheum Dis. 2017;76(12):2103]. Ann Rheum Dis. 2017;76(8):1411–9. https://pubmed.ncbi.nlm.nih.gov/28536116/
Vaughan A, Frazer ZA, Hansbro PM, Yang IA. COPD and the gut-lung axis: the therapeutic potential of fibre. J Thorac Dis. 2019;11(Suppl 17):S2173–80. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6831926/
Reynolds A, Mann J, Cummings J, Winter N, Mete E, Te Morenga L. Carbohydrate quality and human health: a series of systematic reviews and meta-analyses. Lancet. 2019;393(10170):434-45. https://pubmed.ncbi.nlm.nih.gov/30638909/
Brewer RA, Gibbs VK, Smith DL Jr. Targeting glucose metabolism for healthy aging. Nutr Healthy Aging. 2016;4(1):31–46. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5166514/
Su B, Liu H, Li J, et al. Acarbose treatment affects the serum levels of inflammatory cytokines and the gut content of bifidobacteria in Chinese patients with type 2 diabetes mellitus. J Diabetes. 2015;7(5):729–39. https://pubmed.ncbi.nlm.nih.gov/25327485/
Zhang X, Fang Z, Zhang C, et al. Effects of acarbose on the gut microbiota of prediabetic patients: a randomized, double-blind, controlled crossover trial. Diabetes Ther. 2017;8(2):293–307. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5380489/
Wolever TMS, Chiasson JL. Acarbose raises serum butyrate in human subjects with impaired glucose tolerance. Br J Nutr. 2000;84(1):57–61. https://pubmed.ncbi.nlm.nih.gov/10961161/
McCay CM, Ku CC, Woodward JC, Sehgal BS. Cellulose in the diet of rats and mice: two figures. J Nutr. 1934;8(4):435–47. https://academic.oup.com/jn/article-abstract/8/4/435/4727178
Smith BJ, Miller RA, Ericsson AC, Harrison DC, Strong R, Schmidt TM. Changes in the gut microbiome and fermentation products concurrent with enhanced longevity in acarbose-treated mice. BMC Microbiol. 2019;19(1):130. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6567620/
Hovey AL, Jones GP, Devereux HM, Walker KZ. Whole cereal and legume seeds increase faecal short chain fatty acids compared to ground seeds. Asia Pac J Clin Nutr. 2003;12(4):477–82. https://pubmed.ncbi.nlm.nih.gov/14672874/
Stephen AM, Cummings JH. The microbial contribution to human faecal mass. J Med Microbiol. 1980;13(1):45–56. https://pubmed.ncbi.nlm.nih.gov/7359576/
Singh RK, Chang HW, Yan D, et al. Influence of diet on the gut microbiome and implications for human health. J Transl Med. 2017;15(1):73. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5385025/
Franceschi C, Ostan R, Santoro A. Nutrition and inflammation: are centenarians similar to individuals on calorie-restricted diets? Annu Rev Nutr. 2018;38(1):329–56. https://pubmed.ncbi.nlm.nih.gov/29852087/
Minciullo PL, Catalano A, Mandraffino G, et al. Inflammaging and anti-inflammaging: the role of cytokines in extreme longevity. Arch Immunol Ther Exp (Warsz). 2016;64(2):111–26. https://pubmed.ncbi.nlm.nih.gov/26658771/
Minciullo PL, Catalano A, Mandraffino G, et al. Inflammaging and anti-inflammaging: the role of cytokines in extreme longevity. Arch Immunol Ther Exp (Warsz). 2016;64(2):111–26. https://pubmed.ncbi.nlm.nih.gov/26658771/
Säemann MD, Böhmig GA, Österreicher CH, et al. Anti-inflammatory effects of sodium butyrate on human monocytes: potent inhibition of IL-12 and up-regulation of IL-10 production. FASEB J. 2000;14(15):2380–2. https://pubmed.ncbi.nlm.nih.gov/11024006/
Vitaglione P, Mennella I, Ferracane R, et al. Whole-grain wheat consumption reduces inflammation in a randomized controlled trial on overweight and obese subjects with unhealthy dietary and lifestyle behaviors: role of polyphenols bound to cereal dietary fiber. Am J Clin Nutr. 2015;101(2):251–61. https://pubmed.ncbi.nlm.nih.gov/25646321/
Kohl A, Gögebakan Ö, Möhlig M, et al. Increased interleukin-10 but unchanged insulin sensitivity after 4 weeks of (1, 3)(1, 6)-ß-glycan consumption in overweight humans. Nutr Res. 2009;29(4):248–54. https://pubmed.ncbi.nlm.nih.gov/19410976/
Barclay GR, McKenzie H, Pennington J, Parratt D, Pennington CR. The effect of dietary yeast on the activity of stable chronic Crohn’s disease. Scand J Gastroenterol. 1992;27(3):196–200. https://pubmed.ncbi.nlm.nih.gov/1502481/
Cannistrà C, Finocchi V, Trivisonno A, Tambasco D. New perspectives in the treatment of hidradenitis suppurativa: surgery and brewer’s yeast-exclusion diet. Surgery. 2013;154(5):1126–30. https://pubmed.ncbi.nlm.nih.gov/23891479/
Franceschi C, Ostan R, Santoro A. Nutrition and inflammation: are centenarians similar to individuals on calorie-restricted diets? Annu Rev Nutr. 2018;38(1):329–56. https://pubmed.ncbi.nlm.nih.gov/29852087/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Barbaresko J, Koch M, Schulze MB, Nöthlings U. Dietary pattern analysis and biomarkers of low-grade inflammation: a systematic literature review. Nutr Rev. 2013;71(8):511–27. https://pubmed.ncbi.nlm.nih.gov/23865797/
Eichelmann F, Schwingshackl L, Fedirko V, Aleksandrova K. Effect of plant-based diets on obesity-related inflammatory profiles: a systematic review and meta-analysis of intervention trials. Obes Rev. 2016;17(11):1067–79. https://pubmed.ncbi.nlm.nih.gov/27405372/
Sutliffe JT, Wilson LD, de Heer HD, Foster RL, Carnot MJ. C-reactive protein response to a vegan lifestyle intervention. Complement Ther Med. 2015;23(1):32–7. https://pubmed.ncbi.nlm.nih.gov/25637150/
Macknin M, Kong T, Weier A, et al. Plant-based, no-added-fat or American Heart Association diets: impact on cardiovascular risk in obese children with hypercholesterolemia and their parents. J Pediatr. 2015;166(4):953–9.e1–3. https://pubmed.ncbi.nlm.nih.gov/25684089/
Hosseinpour-Niazi S, Mirmiran P, Fallah-Ghohroudi A, Azizi F. Non-soya legume-based therapeutic lifestyle change diet reduces inflammatory status in diabetic patients: a randomised cross-over clinical trial. Br J Nutr. 2015;114(2):213–9. https://pubmed.ncbi.nlm.nih.gov/26077375/
Watzl B, Kulling SE, Möseneder J, Barth SW, Bub A. A 4-wk intervention with high intake of carotenoid-rich vegetables and fruit reduces plasma C-reactive protein in healthy, nonsmoking men. Am J Clin Nutr. 2005;82(5):1052–8. https://pubmed.ncbi.nlm.nih.gov/16280438/
Lee-Kwan SH, Moore LV, Blanck HM, Harris DM, Galuska D. Disparities in state-specific adult fruit and vegetable consumption – United States, 2015. MMWR Morb Mortal Wkly Rep. 2017;66:1241–7. https://pubmed.ncbi.nlm.nih.gov/29145355/
Baden MY, Satija A, Hu FB, Huang T. Change in plant-based diet quality is associated with changes in plasma adiposity-associated biomarker concentrations in women. J Nutr. 2019;149(4):676–86. https://pubmed.ncbi.nlm.nih.gov/30927000/
Ricker MA, Haas WC. Anti-inflammatory diet in clinical practice: a review. Nutr Clin Pract. 2017;32(3):318–25. https://pubmed.ncbi.nlm.nih.gov/28350517/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Li K, Huang T, Zheng J, Wu K, Li D. Effect of marine-derived n-3 polyunsaturated fatty acids on C-reactive protein, interleukin 6 and tumor necrosis factor a: a meta-analysis. PLoS ONE. 2014;9(2):e88103. https://pubmed.ncbi.nlm.nih.gov/24505395/
Agricultural Research Service, United States Department of Agriculture. Search results: PUFA 22:6 n-3 (DHA) (g). FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/?component=1272. Published April 1, 2019. Accessed July 19, 2021.; https://fdc.nal.usda.gov/fdc-app.html#/?component=1272
Stella AB, Cappellari GG, Barazzoni R, Zanetti M. Update on the impact of omega 3 fatty acids on inflammation, insulin resistance and sarcopenia: a review. Int J Mol Sci. 2018;19(1):218. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5796167/
Alhassan A, Young J, Lean MEJ, Lara J. Consumption of fish and vascular risk factors: a systematic review and meta-analysis of intervention studies. Atherosclerosis. 2017;266:87–94. https://pubmed.ncbi.nlm.nih.gov/28992469/
Gopinath B, Buyken AE, Flood VM, Empson M, Rochtchina E, Mitchell P. Consumption of polyunsaturated fatty acids, fish, and nuts and risk of inflammatory disease mortality. Am J Clin Nutr. 2011;93(5):1073–9. https://pubmed.ncbi.nlm.nih.gov/21411616/
Raymond MR, Christensen KY, Thompson BA, Anderson HA. Associations between fish consumption and contaminant biomarkers with cardiovascular conditions among older male anglers in Wisconsin. J Occup Environ Med. 2016;58(7):676–82. https://pubmed.ncbi.nlm.nih.gov/27253229/
Tabung FK, Smith-Warner SA, Chavarro JE, et al. Development and validation of an empirical dietary inflammatory index. J Nutr. 2016;146(8):1560–70. https://pubmed.ncbi.nlm.nih.gov/27358416/
Hjartåker A, Knudsen MD, Tretli S, Weiderpass E. Consumption of berries, fruits and vegetables and mortality among 10,000 Norwegian men followed for four decades. Eur J Nutr. 2015;54(4):599–608. https://pubmed.ncbi.nlm.nih.gov/25087093/
Cassidy A, Rogers G, Peterson JJ, Dwyer JT, Lin H, Jacques PF. Higher dietary anthocyanin and flavonol intakes are associated with anti-inflammatory effects in a population of US adults. Am J Clin Nutr. 2015;102(1):172–81. https://pubmed.ncbi.nlm.nih.gov/26016863/
Nair AR, Mariappan N, Stull AJ, Francis J. Blueberry supplementation attenuates oxidative stress within monocytes and modulates immune cell levels in adults with metabolic syndrome: a randomized, double-blind, placebo-controlled trial. Food Funct. 2017;8(11):4118–28. https://pubmed.ncbi.nlm.nih.gov/29019365/
Moazen S, Amani R, Homayouni Rad A, Shahbazian H, Ahmadi K, Taha Jalali M. Effects of freeze-dried strawberry supplementation on metabolic biomarkers of atherosclerosis in subjects with type 2 diabetes: a randomized double-blind controlled trial. Ann Nutr Metab. 2013;63(3):256–64. https://pubmed.ncbi.nlm.nih.gov/24334868/
Moylan S, Berk M, Dean OM, et al. Oxidative & nitrosative stress in depression: why so much stress? Neurosci Biobehav Rev. 2014;45:46–62. https://pubmed.ncbi.nlm.nih.gov/24858007/
Franzini L, Ardigi D, Valtueña S, et al. Food selection based on high total antioxidant capacity improves endothelial function in a low cardiovascular risk population. Nutr Metab Cardiovasc Dis. 2012;22(1):50–7. https://pubmed.ncbi.nlm.nih.gov/20674303/
Sun CH, Li Y, Zhang YB, Wang F, Zhou XL, Wang F. The effect of vitamin – mineral supplementation on CRP and IL-6: a systemic review and meta-analysis of randomised controlled trials. Nutr Metab Cardiovasc Dis. 2011;21(8):576–83. https://pubmed.ncbi.nlm.nih.gov/20399082/
Fallah AA, Sarmast E, Fatehi P, Jafari T. Impact of dietary anthocyanins on systemic and vascular inflammation: systematic review and meta-analysis on randomised clinical trials. Food Chem Toxicol. 2020;135:110922. https://pubmed.ncbi.nlm.nih.gov/31669599/
do Rosario VA, Chang C, Spencer J, et al. Anthocyanins attenuate vascular and inflammatory responses to a high fat high energy meal challenge in overweight older adults: a cross-over, randomized, double-blind clinical trial. Clin Nutr. 2021;40(3):879–89. https://pubmed.ncbi.nlm.nih.gov/33071012/
O’Hara C, Ojo B, Emerson SR, et al. Acute freeze-dried mango consumption with a high-fat meal has minimal effects on postprandial metabolism, inflammation and antioxidant enzymes. Nutr Metab Insights. 2019;12:1178638819869946. https://pubmed.ncbi.nlm.nih.gov/31452602/
Wang P, Zhang Q, Hou H, et al. The effects of pomegranate supplementation on biomarkers of inflammation and endothelial dysfunction: a meta-analysis and systematic review. Complement Ther Med. 2020;49:102358. https://pubmed.ncbi.nlm.nih.gov/32147056/
Aptekmann NP, Cesar TB. Orange juice improved lipid profile and blood lactate of overweight middle-aged women subjected to aerobic training. Maturitas. 2010;67(4):343–7. https://pubmed.ncbi.nlm.nih.gov/20729016/
McAnulty LS, Nieman DC, Dumke CL, et al. Effect of blueberry ingestion on natural killer cell counts, oxidative stress, and inflammation prior to and after 2.5 h of running. Appl Physiol Nutr Metab. 2011;36(6):976–84. https://pubmed.ncbi.nlm.nih.gov/22111516/
Connolly DA, McHugh MP, Padilla-Zakour OI, Carlson L, Sayers SP. Efficacy of a tart cherry juice blend in preventing the symptoms of muscle damage. Br J Sports Med. 2006;40(8):679–83. https://pubmed.ncbi.nlm.nih.gov/16790484/
Peake JM, Suzuki K, Coombes JS. The influence of antioxidant supplementation on markers of inflammation and the relationship to oxidative stress after exercise. J Nutr Biochem. 2007;18(6):357–71. https://pubmed.ncbi.nlm.nih.gov/17156994/
Childs A, Jacobs C, Kaminski T, Halliwell B, Leeuwenburgh C. Supplementation with vitamin C and N-acetyl-cysteine increases oxidative stress in humans after an acute muscle injury induced by eccentric exercise. Free Radic Biol Med. 2001;31(6):745–53. https://pubmed.ncbi.nlm.nih.gov/11557312/
McHugh M. The health benefits of cherries and potential applications in sports. Scand J Med Sci Sports. 2011;21(5):615–6. https://pubmed.ncbi.nlm.nih.gov/21917014/
Blau LW. Cherry diet control for gout and arthritis. Tex Rep Biol Med. 1950;8(3):309–11. https://pubmed.ncbi.nlm.nih.gov/14776685/
Overman T. Pegloticase: a new treatment for gout. Pharmacotherapy Update. 2011;14(2):1–3. https://pubmed.ncbi.nlm.nih.gov/29204266/
Finkelstein Y, Aks SE, Hutson JR, et al. Colchicine poisoning: the dark side of an ancient drug. Clin Toxicol (Phila). 2010;48(5):407–14. https://pubmed.ncbi.nlm.nih.gov/20586571/
Fritsch PO, Sidoroff A. Drug-induced Stevens-Johnson syndrome/toxic epidermal necrolysis. Am J Clin Dermatol. 2000;1(6):349–60. https://pubmed.ncbi.nlm.nih.gov/11702611/
Wang M, Jiang X, Wu W, Zhang D. A meta-analysis of alcohol consumption and the risk of gout. Clin Rheumatol. 2013;32(11):1641–8. https://pubmed.ncbi.nlm.nih.gov/23881436/
Zhang Y, Chen C, Choi H, et al. Purine-rich foods intake and recurrent gout attacks. Ann Rheum Dis. 2012;71(9):1448–53. https://pubmed.ncbi.nlm.nih.gov/22648933/
Menzel J, Jabakhanji A, Biemann R, Mai K, Abraham K, Weikert C. Systematic review and meta-analysis of the associations of vegan and vegetarian diets with inflammatory biomarkers. Sci Rep. 2020;10:21736. https://pubmed.ncbi.nlm.nih.gov/33303765/
Eichelmann F, Schwingshackl L, Fedirko V, Aleksandrova K. Effect of plant-based diets on obesity-related inflammatory profiles: a systematic review and meta-analysis of intervention trials. Obes Rev. 2016;17(11):1067–79. https://pubmed.ncbi.nlm.nih.gov/27405372/
Tran E, Dale HF, Jensen C, Lied GA. Effects of plant-based diets on weight status: a systematic review. Diabetes Metab Syndr Obes. 2020;13:3433–48. https://pubmed.ncbi.nlm.nih.gov/33061504/
Shah B, Newman JD, Woolf K, et al. Anti-inflammatory effects of a vegan diet versus the American Heart Association – recommended diet in coronary artery disease trial. J Am Heart Assoc. 2018;7(23):e011367. https://pubmed.ncbi.nlm.nih.gov/30571591/
Margolis KL, Manson JE, Greenland P, et al. Leukocyte count as a predictor of cardiovascular events and mortality in postmenopausal women: the Women’s Health Initiative Observational Study. Arch Intern Med. 2005;165(5):500–8. https://pubmed.ncbi.nlm.nih.gov/15767524/
Leng SX, Xue QL, Huang Y, Ferrucci L, Fried LP, Walston JD. Baseline total and specific differential white blood cell counts and 5-year all-cause mortality in community-dwelling older women. Exp Gerontol. 2005;40(12):982–7. https://pubmed.ncbi.nlm.nih.gov/16183235/
Gkrania-Klotsas E, Ye Z, Cooper AJ, et al. Differential white blood cell count and type 2 diabetes: systematic review and meta-analysis of cross-sectional and prospective studies. PLoS One. 2010;5(10):e13405. https://pubmed.ncbi.nlm.nih.gov/20976133/
Leng SX, Xue QL, Huang Y, Ferrucci L, Fried LP, Walston JD. Baseline total and specific differential white blood cell counts and 5-year all-cause mortality in community-dwelling older women. Exp Gerontol. 2005;40(12):982–7. https://pubmed.ncbi.nlm.nih.gov/16183235/
de Labry LO, Campion EW, Glynn RJ, Vokonas PS. White blood cell count as a predictor of mortality: results over 18 years from the Normative Aging Study. J Clin Epidemiol. 1990;43(2):153–7. https://pubmed.ncbi.nlm.nih.gov/2303845/
Panagiotakos DB, Pitsavos C, Chrysohoou C, et al. Effect of exposure to secondhand smoke on markers of inflammation: the ATTICA study. Am J Med. 2004;116(3):145–50. https://pubmed.ncbi.nlm.nih.gov/14749157/
Swanson E. Prospective clinical study reveals significant reduction in triglyceride level and white blood cell count after liposuction and abdominoplasty and no change in cholesterol levels. Plast Reconstr Surg. 2011;128(3):182e-97e. https://pubmed.ncbi.nlm.nih.gov/21865992/
Domene PA, Moir HJ, Pummell E, Knox A, Easton C. The health-enhancing efficacy of Zumba® fitness: an 8-week randomised controlled study. J Sports Sci. 2016;34(15):1396–404. https://pubmed.ncbi.nlm.nih.gov/26571136/
Kjeldsen-Kragh J. Rheumatoid arthritis treated with vegetarian diets. Am J Clin Nutr. 1999;70(3 Suppl):594S-600S. https://pubmed.ncbi.nlm.nih.gov/10479237/
Schultz H, Ying GS, Dunaief JL, Dunaief DM. Rising plasma beta-carotene is associated with diminishing C-reactive protein in patients consuming a dark green leafy vegetable – rich, Low Inflammatory Foods Everyday (LIFE) diet. Am J Lifestyle Med. https://journals.sagepub.com/doi/10.1177/1559827619894954. Published December 21, 2019. Accessed June 26, 2021.; https://pubmed.ncbi.nlm.nih.gov/34916884/
Perzia B, Ying GS, Dunaief JL, Dunaief DM. Once-daily Low Inflammatory Foods Everyday (LIFE) smoothie or the full LIFE diet lowers C-reactive protein and raises plasma beta-carotene in 7 days. Am J Lifestyle Med. https://journals.sagepub.com/doi/10.1177/1559827620962458. Published October 5, 2020. Accessed June 26, 2021.; https://pubmed.ncbi.nlm.nih.gov/36389047/
Castenmiller JJM, West CE, Linssen JPH, van het Hof KH, Voragen AGJ. The food matrix of spinach is a limiting factor in determining the bioavailability of ß-carotene and to a lesser extent of lutein in humans. J Nutr. 1999;129(2):349–55. https://pubmed.ncbi.nlm.nih.gov/10024612/
Lin KH, Hsu CY, Huang YP, et al. Chlorophyll-related compounds inhibit cell adhesion and inflammation in human aortic cells. J Med Food. 2013;16(10):886–98. https://pubmed.ncbi.nlm.nih.gov/24066944/
Subramoniam A, Asha VV, Nair SA, et al. Chlorophyll revisited: anti-inflammatory activities of chlorophyll a and inhibition of expression of TNF-a gene by the same. Inflammation. 2012;35(3):959–66. https://pubmed.ncbi.nlm.nih.gov/22038065/
Jiang Y, Wu SH, Shu XO, et al. Cruciferous vegetable intake is inversely correlated with circulating levels of proinflammatory markers in women. J Acad Nutr Diet. 2014;114(5):700–8.e2. https://pubmed.ncbi.nlm.nih.gov/25165394/
Zhang X, Shu XO, Xiang YB, et al. Cruciferous vegetable consumption is associated with a reduced risk of total and cardiovascular disease mortality. Am J Clin Nutr. 2011;94(1):240–6. https://pubmed.ncbi.nlm.nih.gov/21593509/
Navarro SL, Schwarz Y, Song X, et al. Cruciferous vegetables have variable effects on biomarkers of systemic inflammation in a randomized controlled trial in healthy young adults. J Nutr. 2014;144(11):1850–7. https://pubmed.ncbi.nlm.nih.gov/25165394/
López-Chillón MT, Carazo-Díaz C, Prieto-Merino D, Zafrilla P, Moreno DA, Villaño D. Effects of long-term consumption of broccoli sprouts on inflammatory markers in overweight subjects. Clin Nutr. 2019;38(2):745–52. https://pubmed.ncbi.nlm.nih.gov/29573889/
Bentley J. Potatoes and tomatoes account for over half of U.S. vegetable availability. Economic Research Service, United States Department of Agriculture. https://www.ers.usda.gov/amber-waves/2015/september/potatoes-and-tomatoes-account-for-over-half-of-us-vegetable-availability. Published September 8, 2015. Accessed June 20, 2021.; https://www.ers.usda.gov/amber-waves/2015/september/potatoes-and-tomatoes-account-for-over-half-of-us-vegetable-availability/
Ghavipour M, Saedisomeolia A, Djalali M, et al. Tomato juice consumption reduces systemic inflammation in overweight and obese females. Br J Nutr. 2013;109(11):2031–5. https://pubmed.ncbi.nlm.nih.gov/23069270/
Burton-Freeman B, Talbot J, Park E, Krishnankutty S, Edirisinghe I. Protective activity of processed tomato products on postprandial oxidation and inflammation: a clinical trial in healthy weight men and women. Mol Nutr Food Res. 2012;56(4):622–31. https://pubmed.ncbi.nlm.nih.gov/22331646/
Markovits N, Ben Amotz A, Levy Y. The effect of tomato-derived lycopene on low carotenoids and enhanced systemic inflammation and oxidation in severe obesity. Isr Med Assoc J. 2009;11(10):598–601. https://pubmed.ncbi.nlm.nih.gov/20077945/
Dai X, Stanilka JM, Rowe CA, et al. Consuming Lentinula edodes (shiitake) mushrooms daily improves human immunity: a randomized dietary intervention in healthy young adults. J Am Coll Nutr. 2015;34(6):478–87. https://pubmed.ncbi.nlm.nih.gov/25866155/
World Cancer Research Fund / American Institute for Cancer Research. Food, Nutrition, Physical Activity, and the Prevention of Cancer: a Global Perspective. American Institute for Cancer Research; 2007. https://www.researchgate.net/publication/315725512_Food_Nutrition_Physical_Activity_and_the_Prevention_of_Cancer_A_Global_Perspective_Summary
American Heart Association. Types of whole grains. Heart.org. https://www.heart.org/en/healthy-living/healthy-eating/eat-smart/nutrition-basics/types-of-whole-grains. Published January 1, 2015. Accessed November 5, 2021.; https://www.heart.org/en/healthy-living/healthy-eating/eat-smart/nutrition-basics/types-of-whole-grains
Aune D, Keum N, Giovannucci E, et al. Whole grain consumption and risk of cardiovascular disease, cancer, and all cause and cause specific mortality: systematic review and dose-response meta-analysis of prospective studies. BMJ. 2016;353:i2716. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4908315/
Jacobs DR, Andersen LF, Blomhoff R. Whole-grain consumption is associated with a reduced risk of noncardiovascular, noncancer death attributed to inflammatory diseases in the Iowa Women’s Health Study. Am J Clin Nutr. 2007;85(6):1606–14. https://pubmed.ncbi.nlm.nih.gov/17556700/
Aune D, Keum N, Giovannucci E, et al. Whole grain consumption and risk of cardiovascular disease, cancer, and all cause and cause specific mortality: systematic review and dose-response meta-analysis of prospective studies. BMJ. 2016;353:i2716. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4908315/
Afshin A, Sun PJ, Fay KA, et al. Health effects of dietary risks in 195 countries, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2019;393(10184):1958–72. https://pubmed.ncbi.nlm.nih.gov/30954305/
Yu Z, Malik VS, Keum NN, et al. Associations between nut consumption and inflammatory biomarkers. Am J Clin Nutr. 2016;104(3):722–8. https://pubmed.ncbi.nlm.nih.gov/27465378/
Gopinath B, Buyken AE, Flood VM, Empson M, Rochtchina E, Mitchell P. Consumption of polyunsaturated fatty acids, fish, and nuts and risk of inflammatory disease mortality. Am J Clin Nutr. 2011;93(5):1073–9. https://pubmed.ncbi.nlm.nih.gov/21411616/
Chen GC, Zhang R, Martínez-González MA, et al. Nut consumption in relation to all-cause and cause-specific mortality: a meta-analysis 18 prospective studies. Food Funct. 2017;8(11):3893–905. https://pubmed.ncbi.nlm.nih.gov/28875220/
Xiao Y, Xia J, Ke Y, et al. Effects of nut consumption on selected inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. Nutrition. 2018;54:129–43. https://pubmed.ncbi.nlm.nih.gov/29852452/
Eftekhar Sadat B, Khadem Haghighian M, Alipoor B, Malek Mahdavi A, Asghari Jafarabadi M, Moghaddam A. Effects of sesame seed supplementation on clinical signs and symptoms in patients with knee osteoarthritis. Int J Rheum Dis. 2013;16(5):578–82. https://pubmed.ncbi.nlm.nih.gov/24164846/
Rodriguez-Leyva D, Weighell W, Edel AL, et al. Potent antihypertensive action of dietary flaxseed in hypertensive patients. Hypertension. 2013;62(6):1081–9. https://pubmed.ncbi.nlm.nih.gov/24126178/
Rahimlou M, Jahromi NB, Hasanyani N, Ahmadi AR. Effects of flaxseed interventions on circulating inflammatory biomarkers: a systematic review and meta-analysis of randomized controlled trials. Adv Nutr. 2019;10(6):1108–19. https://pubmed.ncbi.nlm.nih.gov/31115436/
Caligiuri SPB, Parikh M, Stamenkovic A, Pierce GN, Aukema HM. Dietary modulation of oxylipins in cardiovascular disease and aging. Am J Physiol Heart Circ Physiol. 2017;313(5):H903–18. https://pubmed.ncbi.nlm.nih.gov/28801523/
Caligiuri SPB, Aukema HM, Ravandi A, Pierce GN. Elevated levels of pro-inflammatory oxylipins in older subjects are normalized by flaxseed consumption. Exp Gerontol. 2014;59:51–7. https://pubmed.ncbi.nlm.nih.gov/24747581/
Srinivasan K. Anti-inflammatory influences of culinary spices and their bioactives. Food Rev Int. 2020;Nov:1–17. https://www.tandfonline.com/doi/abs/10.1080/87559129.2020.1839761?journalCode=lfri20
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Allijn IE, Vaessen SF, Quarles van Ufford LC, et al. Head-to-head comparison of anti-inflammatory performance of known natural products in vitro. PLoS ONE. 2016;11(5):e0155325. https://pubmed.ncbi.nlm.nih.gov/27163931/
Daily JW, Yang M, Park S. Efficacy of turmeric extracts and curcumin for alleviating the symptoms of joint arthritis: a systematic review and meta-analysis of randomized clinical trials. J Med Food. 2016;19(8):717–29. https://pubmed.ncbi.nlm.nih.gov/27533649/
Abidi A, Gupta S, Agarwal M, Bhalla HL, Saluja M. Evaluation of efficacy of curcumin as an add-on therapy in patients of bronchial asthma. J Clin Diagn Res. 2014;8(8):HC19–24. https://pubmed.ncbi.nlm.nih.gov/25302215/
Panahi Y, Sahebkar A, Parvin S, Saadat A. A randomized controlled trial on the anti-inflammatory effects of curcumin in patients with chronic sulphur mustard-induced cutaneous complications. Ann Clin Biochem. 2012;49(Pt 6):580–8. https://pubmed.ncbi.nlm.nih.gov/23038702/
Garg SK, Ahuja V, Sankar MJ, Kumar A, Moss AC. Curcumin for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev. 2012;10:CD008424. https://pubmed.ncbi.nlm.nih.gov/23076948/
Khajehdehi P, Zanjaninejad B, Aflaki E, et al. Oral supplementation of turmeric decreases proteinuria, hematuria, and systolic blood pressure in patients suffering from relapsing or refractory lupus nephritis: a randomized and placebo-controlled study. J Ren Nutr. 2012;22(1):50–7. https://pubmed.ncbi.nlm.nih.gov/21742514/
Vors C, Couillard C, Paradis ME, et al. Supplementation with resveratrol and curcumin does not affect the inflammatory response to a high-fat meal in older adults with abdominal obesity: a randomized, placebo-controlled crossover trial. J Nutr. 2018;148(3):379–88. https://pubmed.ncbi.nlm.nih.gov/29546309/
Derosa G, Maffioli P, Simental-Mendía LE, Bo S, Sahebkar A. Effect of curcumin on circulating interleukin-6 concentrations: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res. 2016;111:394–404. https://pubmed.ncbi.nlm.nih.gov/27392742/
Sahebkar A, Cicero AFG, Simental-Mendía LE, Aggarwal BB, Gupta SC. Curcumin downregulates human tumor necrosis factor-a levels: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res. 2016;107:234–42. https://pubmed.ncbi.nlm.nih.gov/27025786/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Morvaridzadeh M, Fazelian S, Agah S, et al. Effect of ginger (Zingiber officinale) on inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. Cytokine. 2020;135:155224. https://pubmed.ncbi.nlm.nih.gov/32763761/
Aryaeian N, Shahram F, Mahmoudi M, et al. The effect of ginger supplementation on some immunity and inflammation intermediate genes expression in patients with active Rheumatoid Arthritis. Gene. 2019;698:179–185. https://pubmed.ncbi.nlm.nih.gov/30844477/
Bartels EM, Folmer VN, Bliddal H, et al. Efficacy and safety of ginger in osteoarthritis patients: a meta-analysis of randomized placebo-controlled trials. Osteoar Cartil. 2015;23(1):13–21. https://pubmed.ncbi.nlm.nih.gov/25300574/
Haghighi M, Khalvat A, Toliat T, Jallaei SH. Comparing the effects of ginger (Zingiber officinale) extract and ibuprofen on patients with osteoarthritis. Arch Iran Med. 2005;8(4):267–71. https://www.researchgate.net/publication/235007127_Comparing_the_Effects_of_ginger_Zingiber_officinale_extract_and_ibuprofen_On_patients_with_osteoarthritis
Haniadka R, Saldanha E, Sunita V, Palatty PL, Fayad R, Baliga MS. A review of the gastroprotective effects of ginger (Zingiber officinale Roscoe). Food Funct. 2013;4(6):845–55. https://pubmed.ncbi.nlm.nih.gov/23612703/
Caunedo-Alvarez A, Gómez-Rodríguez BJ, Romero-Vázquez J, et al. Macroscopic small bowel mucosal injury caused by chronic nonsteroidal anti-inflammatory drugs (NSAID) use as assessed by capsule endoscopy. Rev Esp Enferm Dig. 2010;102(2):80–5. https://pubmed.ncbi.nlm.nih.gov/20361843/
Maghbooli M, Golipour F, Moghimi Esfandabadi A, Yousefi M. Comparison between the efficacy of ginger and sumatriptan in the ablative treatment of the common migraine. Phytother Res. 2014;28(3):412–5. https://pubmed.ncbi.nlm.nih.gov/23657930/
Kashefi F, Khajehei M, Alavinia M, Golmakani E, Asili J. Effect of ginger (Zingiber officinale) on heavy menstrual bleeding: a placebo-controlled, randomized clinical trial. Phytother Res. 2015;29(1):114–9. https://pubmed.ncbi.nlm.nih.gov/25298352/
Dugasani S, Pichika MR, Nadarajah VD, Balijepalli MK, Tandra S, Korlakunta JN. Comparative antioxidant and anti-inflammatory effects of [6]-gingerol, [8]-gingerol, [10]-gingerol and [6]-shogaol. J Ethnopharmacol. 2010;127(2):515–20. https://pubmed.ncbi.nlm.nih.gov/19833188/
Darooghegi Mofrad M, Milajerdi A, Koohdani F, Surkan PJ, Azadbakht L. Garlic supplementation reduces circulating C-reactive protein, tumor necrosis factor, and interleukin-6 in adults: a systematic review and meta-analysis of randomized controlled trials. J Nutr. 2019;149(4):605–18. https://pubmed.ncbi.nlm.nih.gov/30949665/
Moosavian SP, Paknahad Z, Habibagahi Z, Maracy M. The effects of garlic (Allium sativum) supplementation on inflammatory biomarkers, fatigue, and clinical symptoms in patients with active rheumatoid arthritis: a randomized, double-blind, placebo-controlled trial. Phytother Res. 2020;34(11):2953–62. https://pubmed.ncbi.nlm.nih.gov/32478922/
Taghizadeh M, Hamedifard Z, Jafarnejad S. Effect of garlic supplementation on serum C-reactive protein level: a systematic review and meta-analysis of randomized controlled trials. Phytother Res. 2019;33(2):243–52. https://pubmed.ncbi.nlm.nih.gov/30370629/
Percival SS, Vanden Heuvel JP, Nieves CJ, Montero C, Migliaccio AJ, Meadors J. Bioavailability of herbs and spices in humans as determined by ex vivo inflammatory suppression and DNA strand breaks. J Am Coll Nutr. 2012;31(4):288–94. https://pubmed.ncbi.nlm.nih.gov/23378457/
Payahoo L, Ostadrahimi A, Mobasseri M, et al. Anethum graveolens L. supplementation has anti-inflammatory effect in type 2 diabetic patients. Indian J Tradit Knowl. 2014:13(3):461–5.; https://www.researchgate.net/publication/267032371_Anethum_graveolens_L_supplementation_has_anti-inflammatory_effect_in_type_2_diabetic_patients
Vallianou N, Tsang C, Taghizadeh M, Davoodvandi A, Jafarnejad S. Effect of cinnamon (Cinnamomum zeylanicum) supplementation on serum C-reactive protein concentrations: a meta-analysis and systematic review. Complement Ther Med. 2019;42:271–8. https://pubmed.ncbi.nlm.nih.gov/30670254/
Vallianou N, Tsang C, Taghizadeh M, Davoodvandi A, Jafarnejad S. Effect of cinnamon (Cinnamomum Zeylanicum) supplementation on serum C-reactive protein concentrations: a meta-analysis and systematic review. Complement Ther Med. 2019;42:271–8. https://pubmed.ncbi.nlm.nih.gov/30670254/
Vázquez-Agell M, Urpi-Sarda M, Sacanella E, et al. Cocoa consumption reduces NF-¿B activation in peripheral blood mononuclear cells in humans. Nutr Metab Cardiovasc Dis. 2013;23(3):257–63. https://pubmed.ncbi.nlm.nih.gov/21824756/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Eshghpour M, Mortazavi H, Mohammadzadeh Rezaei NM, Nejat AH. Effectiveness of green tea mouthwash in postoperative pain control following surgical removal of impacted third molars: double blind randomized clinical trial. Daru. 2013;21(1):59. https://pubmed.ncbi.nlm.nih.gov/23866761/
Sridharan S, Archer N, Manning N. Premature constriction of the fetal ductus arteriosus following the maternal consumption of camomile herbal tea. Ultrasound Obstet Gynecol. 2009;34(3):358–9. https://pubmed.ncbi.nlm.nih.gov/19705407/
Burkewitz K, Weir HJM, Mair WB. AMPK as a pro-longevity target. In: Cordero MD, Viollet B, eds. AMP-Activated Protein Kinase. Experientia Supplementum. Vol 107. Springer; 2016:227–56. https://pubmed.ncbi.nlm.nih.gov/27812983/
Duthie GG, Wood AD. Natural salicylates: foods, functions and disease prevention. Food Funct. 2011;2(9):515–20. https://pubmed.ncbi.nlm.nih.gov/21879102/
Fuster V, Sweeny JM. Aspirin: a historical and contemporary therapeutic overview. Circulation. 2011;123(7):768–78. https://pubmed.ncbi.nlm.nih.gov/21343593/
Saad M, Abdelaziz HK, Mehta JL. Aspirin for primary prevention in the elderly. Aging (Albany NY). 2019;11(17):6618–9. https://pubmed.ncbi.nlm.nih.gov/31492828/
Patrono C, Baigent C. Role of aspirin in primary prevention of cardiovascular disease. Nat Rev Cardiol. 2019;16(11):675–86. https://pubmed.ncbi.nlm.nih.gov/31243390/
Duthie GG, Wood AD. Natural salicylates: foods, functions and disease prevention. Food Funct. 2011;2(9):515–20. https://pubmed.ncbi.nlm.nih.gov/21879102/
Duthie GG, Wood AD. Natural salicylates: foods, functions and disease prevention. Food Funct. 2011;2(9):515–20. https://pubmed.ncbi.nlm.nih.gov/21879102/
Blacklock CJ, Lawrence JR, Wiles D, et al. Salicylic acid in the serum of subjects not taking aspirin. Comparison of salicylic acid concentrations in the serum of vegetarians, non-vegetarians, and patients taking low dose aspirin. J Clin Pathol. 2001;54(7):553–5. https://pubmed.ncbi.nlm.nih.gov/11429429/
Knutsen SF. Lifestyle and the use of health services. Am J Clin Nutr. 1994;59(5 Suppl):1171S-5S. https://pubmed.ncbi.nlm.nih.gov/8172119/
McCarty MF. Dietary nitrate and reductive polyphenols may potentiate the vascular benefit and alleviate the ulcerative risk of low-dose aspirin. Med Hypotheses. 2013;80(2):186–90. https://pubmed.ncbi.nlm.nih.gov/23265354/
Scheier L. Salicylic acid: one more reason to eat your fruits and vegetables. J Am Diet Assoc. 2001;101(12):1406–8. https://pubmed.ncbi.nlm.nih.gov/11762733/
Baxter GJ, Graham AB, Lawrence JR, Wiles D, Paterson JR. Salicylic acid in soups prepared from organically and non-organically grown vegetables. Eur J Nutr. 2001;40(6):289–92. https://pubmed.ncbi.nlm.nih.gov/11876493/
Malakar S, Gibson PR, Barrett JS, Muir JG. Naturally occurring dietary salicylates: a closer look at common Australian foods. J Food Compos Anal. 2017;57:31–9. https://www.sciencedirect.com/science/article/abs/pii/S0889157516302241?via%3Dihub
Malakar S, Gibson PR, Barrett JS, Muir JG. Naturally occurring dietary salicylates: a closer look at common Australian foods. J Food Compos Anal. 2017;57:31–9. https://www.sciencedirect.com/science/article/abs/pii/S0889157516302241?via%3Dihub
Paterson JR, Srivastava R, Baxter GJ, Graham AB, Lawrence JR. Salicylic acid content of spices and its implications. J Agric Food Chem. 2006;54(8):2891–6. https://pubmed.ncbi.nlm.nih.gov/16608205/
Keszycka PK, Szkop M, Gajewska D. Overall content of salicylic acid and salicylates in food available on the European market. J Agric Food Chem. 2017;65(50):11085–91. https://pubmed.ncbi.nlm.nih.gov/29182277/
Gajewska D, Keszycka PK, Szkop M. Dietary salicylates in herbs and spices. Food Funct. 2019;10(11):7037–41. https://pubmed.ncbi.nlm.nih.gov/31625548/
Paterson JR, Srivastava R, Baxter GJ, Graham AB, Lawrence JR. Salicylic acid content of spices and its implications. J Agric Food Chem. 2006;54(8):2891–6. https://pubmed.ncbi.nlm.nih.gov/16608205/
Malakar S, Gibson PR, Barrett JS, Muir JG. Naturally occurring dietary salicylates: a closer look at common Australian foods. J Food Compos Anal. 2017;57:31–9. https://www.sciencedirect.com/science/article/abs/pii/S0889157516302241?via%3Dihub
Gajewska D, Keszycka PK, Szkop M. Dietary salicylates in herbs and spices. Food Funct. 2019;10(11):7037–41. https://pubmed.ncbi.nlm.nih.gov/31625548/
Blacklock CJ, Lawrence JR, Wiles D, et al. Salicylic acid in the serum of subjects not taking aspirin. Comparison of salicylic acid concentrations in the serum of vegetarians, non-vegetarians, and patients taking low dose aspirin. J Clin Pathol. 2001;54(7):553–5. https://pubmed.ncbi.nlm.nih.gov/11429429/
Популярное индийское блюдо, завезенное в Гоа португальскими моряками. – Примеч. ред.
Традиционные индийские блюда, приправленные куркумой, перцем чили, чесноком, кумином, кориандром, имбирем, тамариндом, лимонной кислотой, растительным маслом, уксусом и солью. – Примеч. ред.
Paterson JR, Srivastava R, Baxter GJ, Graham AB, Lawrence JR. Salicylic acid content of spices and its implications. J Agric Food Chem. 2006;54(8):2891–6. https://pubmed.ncbi.nlm.nih.gov/16608205/
Paterson JR, Srivastava R, Baxter GJ, Graham AB, Lawrence JR. Salicylic acid content of spices and its implications. J Agric Food Chem. 2006;54(8):2891–6. https://pubmed.ncbi.nlm.nih.gov/16608205/
Pasche B, Wang M, Pennison M, Jimenez H. Prevention and treatment of cancer with aspirin: where do we stand? Semin Oncol. 2014;41(3):397–401. https://pubmed.ncbi.nlm.nih.gov/25023355/
Baxter GJ, Graham AB, Lawrence JR, Wiles D, Paterson JR. Salicylic acid in soups prepared from organically and non-organically grown vegetables. Eur J Nutr. 2001;40(6):289–92. https://pubmed.ncbi.nlm.nih.gov/11876493/
Duthie GG, Wood AD. Natural salicylates: foods, functions and disease prevention. Food Funct. 2011;2(9):515–20. https://pubmed.ncbi.nlm.nih.gov/21879102/
Pawelec G. Aging as an inflammatory disease and possible reversal strategies. J Allergy Clin Immunol. 2020;145(5):1355–6. https://pubmed.ncbi.nlm.nih.gov/32142747/
Puzianowska-Kuznicka M, Owczarz M, Wieczorowska-Tobis K, et al. Interleukin-6 and C-reactive protein, successful aging, and mortality: the PolSenior study. Immun Ageing. 2016;13:21. https://pubmed.ncbi.nlm.nih.gov/27274758/
Assmann KE, Adjibade M, Shivappa N, et al. The inflammatory potential of the diet at midlife is associated with later healthy aging in French adults. J Nutr. 2018;148(3):437–44. https://pubmed.ncbi.nlm.nih.gov/29546305/
Pedersen BK. Anti-inflammation – just another word for anti-ageing? J Physiol. 2009;587(23):5515. https://pubmed.ncbi.nlm.nih.gov/19959548/
O’Keefe JH, Bell DSH. Postprandial hyperglycemia/hyperlipidemia (postprandial dysmetabolism) is a cardiovascular risk factor. Am J Cardiol. 2007;100(5):899–904. https://pubmed.ncbi.nlm.nih.gov/17719342/
Vézina C, Kudelski A, Sehgal SN. Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J Antibiot (Tokyo). 1975;28(10):721–6. https://pubmed.ncbi.nlm.nih.gov/1102508/
Garza-Lombó C, Gonsebatt ME. Mammalian target of rapamycin: its role in early neural development and in adult and aged brain function. Front Cell Neurosci. 2016;10:157. https://pubmed.ncbi.nlm.nih.gov/27378854/
Sabatini DM. Twenty-five years of mTOR: uncovering the link from nutrients to growth. PNAS. 2017;114(45):11818–25. https://pubmed.ncbi.nlm.nih.gov/29078414/
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21(4):183–203. https://pubmed.ncbi.nlm.nih.gov/31937935/
Blagosklonny MV. TOR-driven aging: speeding car without brakes. Cell Cycle. 2009;8(24):4055–9. https://pubmed.ncbi.nlm.nih.gov/19923900/
Schmeisser K, Parker JA. Pleiotropic effects of mTOR and autophagy during development and aging. Front Cell Dev Biol. 2019;7. https://pubmed.ncbi.nlm.nih.gov/31572724/
Vasunilashorn S, Finch CE, Crimmins EM, et al. Inflammatory gene variants in the Tsimane, an indigenous Bolivian population with a high infectious load. Biodemography Soc Biol. 2011;57(1):33–52. https://pubmed.ncbi.nlm.nih.gov/21845926/
Huebbe P, Schloesser A, Rimbach G. A nutritional perspective on cellular rejuvenation. Oncotarget. 2015;6(16):13846–7. https://pubmed.ncbi.nlm.nih.gov/26116836/
Sabatini DM. Twenty-five years of mTOR: uncovering the link from nutrients to growth. PNAS. 2017;114(45):11818–25. https://pubmed.ncbi.nlm.nih.gov/29078414/
Blagosklonny MV. Does rapamycin slow down time? Oncotarget. 2018;9(54):30210–2. https://pubmed.ncbi.nlm.nih.gov/30100983/
Wei Y, Zhang YJ, Cai Y. Growth or longevity: the TOR’s decision on lifespan regulation. Biogerontology. 2013;14(4):353–63. https://pubmed.ncbi.nlm.nih.gov/23740528/
Swindell WR. Meta-analysis of 29 experiments evaluating the effects of rapamycin on life span in the laboratory mouse. J Gerontol A Biol Sci Med Sci. 2017;72(8):1024–32. https://pubmed.ncbi.nlm.nih.gov/27519886/
Blagosklonny MV. Rapamycin for longevity: opinion article. Aging (Albany NY). 2019;11(19):8048–67. https://pubmed.ncbi.nlm.nih.gov/31586989/
Weichhart T. mTOR as regulator of lifespan, aging, and cellular senescence: a mini-review. Gerontology. 2018;64(2):127–34. https://pubmed.ncbi.nlm.nih.gov/29190625/
Sharp ZD, Strong R. The role of mTOR signaling in controlling mammalian life span: what a fungicide teaches us about longevity. J Gerontol A Biol Sci Med Sci. 2010;65A(6):580–9. https://pubmed.ncbi.nlm.nih.gov/20083554/
Kaeberlein M, Kennedy BK. A midlife longevity drug? Nature. 2009;460(7253):331–2. https://pubmed.ncbi.nlm.nih.gov/19606132/
Blagosklonny MV. Rapamycin for longevity: opinion article. Aging (Albany NY). 2019;11(19):8048–67. https://pubmed.ncbi.nlm.nih.gov/31586989/
Arriola Apelo SI, Lamming DW. Rapamycin: an inhibiTOR of aging emerges from the soil of Easter Island. J Gerontol A Biol Sci Med Sci. 2016;71(7):841–9. https://pubmed.ncbi.nlm.nih.gov/27208895/
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21(4):183–203. https://pubmed.ncbi.nlm.nih.gov/31937935/
Weichhart T. mTOR as regulator of lifespan, aging, and cellular senescence: a mini-review. Gerontology. 2018;64(2):127–34. https://pubmed.ncbi.nlm.nih.gov/29190625/
Stallone G, Schena A, Infante B, et al. Sirolimus for Kaposi’s sarcoma in renal-transplant recipients. N Engl J Med. 2005;352(13):1317–23. https://pubmed.ncbi.nlm.nih.gov/15800227/
Majumder S, Caccamo A, Medina DX, et al. Lifelong rapamycin administration ameliorates age-dependent cognitive deficits by reducing IL-1ß and enhancing NMDA signaling. Aging Cell. 2012;11(2):326–35. https://pubmed.ncbi.nlm.nih.gov/22212527/
Wilkinson JE, Burmeister L, Brooks SV, et al. Rapamycin slows aging in mice. Aging Cell. 2012;11(4):675–82. https://pubmed.ncbi.nlm.nih.gov/22587563/
An JY, Kerns KA, Ouellette A, et al. Rapamycin rejuvenates oral health in aging mice. Elife. 2020;9:e54318. https://pubmed.ncbi.nlm.nih.gov/32342860/
Altschuler RA, Kanicki A, Martin C, Kohrman DC, Miller RA. Rapamycin but not acarbose decreases age-related loss of outer hair cells in the mouse cochlea. Hear Res. 2018;370:11–5. https://pubmed.ncbi.nlm.nih.gov/30245283/
Lesniewski LA, Seals DR, Walker AE, et al. Dietary rapamycin supplementation reverses age-related vascular dysfunction and oxidative stress, while modulating nutrient-sensing, cell cycle, and senescence pathways. Aging Cell. 2017;16(1):17–26. https://pubmed.ncbi.nlm.nih.gov/27660040/
Zaseck LW, Miller RA, Brooks SV. Rapamycin attenuates age-associated changes in tibialis anterior tendon viscoelastic properties. J Gerontol A Biol Sci Med Sci. 2016;71(7):858–65. https://pubmed.ncbi.nlm.nih.gov/26809496/
Dai DF, Karunadharma PP, Chiao YA, et al. Altered proteome turnover and remodeling by short-term caloric restriction or rapamycin rejuvenate the aging heart. Aging Cell. 2014;13(3):529–39. https://pubmed.ncbi.nlm.nih.gov/24612461/
Arriola Apelo SI, Pumper CP, Baar EL, Cummings NE, Lamming DW. Intermittent administration of rapamycin extends the life span of female C57BL/6J mice. J Gerontol A Biol Sci Med Sci. 2016;71(7):876–81. https://pubmed.ncbi.nlm.nih.gov/27091134/
Bitto A, Ito TK, Pineda VV, et al. Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. Elife. 2016;5:e16351. https://pubmed.ncbi.nlm.nih.gov/27549339/
Urfer SR, Kaeberlein TL, Mailheau S, et al. A randomized controlled trial to establish effects of short-term rapamycin treatment in 24 middle-aged companion dogs. Geroscience. 2017;39(2):117–27. https://pubmed.ncbi.nlm.nih.gov/28374166/
González A, Hall MN, Lin SC, Hardie DG. AMPK and TOR: the Yin and Yang of cellular nutrient sensing and growth control. Cell Metab. 2020;31(3):472–92. https://pubmed.ncbi.nlm.nih.gov/32130880/
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21(4):183–203. https://pubmed.ncbi.nlm.nih.gov/31937935/
Michels KB, Ekbom A. Caloric restriction and incidence of breast cancer. JAMA. 2004;291(10):1226–30. https://pubmed.ncbi.nlm.nih.gov/15010444/
Wazir U, Newbold RF, Jiang WG, Sharma AK, Mokbel K. Prognostic and therapeutic implications of mTORC1 and Rictor expression in human breast cancer. Oncol Rep. 2013;29(5):1969–74. https://pubmed.ncbi.nlm.nih.gov/23503572/
Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients with anorexia nervosa and other eating disorders. A meta-analysis of 36 studies. Arch Gen Psychiatry. 2011;68(7):724–31. https://pubmed.ncbi.nlm.nih.gov/21727255/
Dar BA, Dar MA, Bashir S. Calorie restriction the fountain of youth. Food Nutr Sci. 2012;3(11):1522–6. https://www.scirp.org/journal/paperinformation.aspx?paperid=24485
Dirks AJ, Leeuwenburgh C. Caloric restriction in humans: potential pitfalls and health concerns. Mech Ageing Dev. 2006;127(1):1–7. https://pubmed.ncbi.nlm.nih.gov/16226298/
Bourzac K. Interventions: live long and prosper. Nature. 2012;492(7427):S18–20. https://pubmed.ncbi.nlm.nih.gov/23222670/
Nakagawa S, Lagisz M, Hector KL, Spencer HG. Comparative and meta-analytic insights into life extension via dietary restriction. Aging Cell. 2012;11(3):401–9. https://pubmed.ncbi.nlm.nih.gov/22268691/
Solon-Biet SM, McMahon AC, Ballard JWO, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19(3):418–30. https://pubmed.ncbi.nlm.nih.gov/24606899/
Ross MH. Length of life and nutrition in the rat. J Nutr. 1961;75:197–210. https://pubmed.ncbi.nlm.nih.gov/14494200/
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21(4):183–203. https://pubmed.ncbi.nlm.nih.gov/31937935/
Fontana L, Partridge L, Longo VD. Extending healthy life span – from yeast to humans. Science. 2010;328(5976):321–6. https://pubmed.ncbi.nlm.nih.gov/20395504/
Kitada M, Xu J, Ogura Y, Monno I, Koya D. Mechanism of activation of mechanistic target of rapamycin complex 1 by methionine. Front Cell Dev Biol. 2020;8:715. https://pubmed.ncbi.nlm.nih.gov/32850834/
Dumas SN, Lamming DW. Next generation strategies for geroprotection via mTORC1 inhibition. J Gerontol A Biol Sci Med Sci. 2020;75(1):14–23. https://pubmed.ncbi.nlm.nih.gov/30794726/
Norton LE, Layman DK, Bunpo P, Anthony TG, Brana DV, Garlick PJ. The leucine content of a complete meal directs peak activation but not duration of skeletal muscle protein synthesis and mammalian target of rapamycin signaling in rats. J Nutr. 2009;139(6):1103–9. https://pubmed.ncbi.nlm.nih.gov/19403715/
Schmidt JA, Rinaldi S, Scalbert A, et al. Plasma concentrations and intakes of amino acids in male meat-eaters, fish-eaters, vegetarians and vegans: a cross-sectional analysis in the EPIC-Oxford cohort. Eur J Clin Nutr. 2016;70(3):306–12. https://pubmed.ncbi.nlm.nih.gov/26395436/
Jafari S, Hezaveh E, Jalilpiran Y, et al. Plant-based diets and risk of disease mortality: a systematic review and meta-analysis of cohort studies. Crit Rev Food Sci Nutr. Published online May 6, 2021:1–13. Accessed June 23, 2021.; https://pubmed.ncbi.nlm.nih.gov/33951994/
Tantamango-Bartley Y, Jaceldo-Siegl K, Fan J, Fraser G. Vegetarian diets and the incidence of cancer in a low-risk population. Cancer Epidemiol Biomarkers Prev. 2013;22(2):286–94. https://pubmed.ncbi.nlm.nih.gov/23169929/
Green CL, Lamming DW. Regulation of metabolic health by essential dietary amino acids. Mech Ageing Dev. 2019;177:186–200. https://pubmed.ncbi.nlm.nih.gov/30044947/
Schmidt JA, Rinaldi S, Scalbert A, et al. Plasma concentrations and intakes of amino acids in male meat-eaters, fish-eaters, vegetarians and vegans: a cross-sectional analysis in the EPIC-Oxford cohort. Eur J Clin Nutr. 2016;70(3):306–12. https://pubmed.ncbi.nlm.nih.gov/26395436/
Willcox BJ, Willcox DC, Todoriki H, et al. Caloric restriction, the traditional Okinawan diet, and healthy aging: the diet of the world’s longest-lived people and its potential impact on morbidity and life span. Ann N Y Acad Sci. 2007;1114:434–55. https://pubmed.ncbi.nlm.nih.gov/17986602/
Davinelli S, Willcox DC, Scapagnini G. Extending healthy ageing: nutrient sensitive pathway and centenarian population. Immun Ageing. 2012;9:9. https://pubmed.ncbi.nlm.nih.gov/22524452/
Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161(13):1645–52. https://pubmed.ncbi.nlm.nih.gov/11434797/
Yasuda M, Tanaka Y, Kume S, et al. Fatty acids are novel nutrient factors to regulate mTORC1 lysosomal localization and apoptosis in podocytes. Biochim Biophys Acta. 2014;1842(7):1097–108. https://pubmed.ncbi.nlm.nih.gov/24726883/
Obersby D, Chappell DC, Dunnett A, Tsiami AA. Plasma total homocysteine status of vegetarians compared with omnivores: a systematic review and meta-analysis. Br J Nutr. 2013;109(5):785–94. https://pubmed.ncbi.nlm.nih.gov/23298782/
Khayati K, Antikainen H, Bonder EM, et al. The amino acid metabolite homocysteine activates mTORC1 to inhibit autophagy and form abnormal proteins in human neurons and mice. FASEB J. 2017;31(2):598–609. https://pubmed.ncbi.nlm.nih.gov/28148781/
Dumas SN, Lamming DW. Next generation strategies for geroprotection via mTORC1 inhibition. J Gerontol A Biol Sci Med Sci. 2020;75(1):14–23. https://pubmed.ncbi.nlm.nih.gov/30794726/
Melnik BC. Dietary intervention in acne: attenuation of increased mTORC1 signaling promoted by Western diet. Dermatoendocrinol. 2012;4(1):20–32. https://pubmed.ncbi.nlm.nih.gov/22870349/
Melnik BC. Linking diet to acne metabolomics, inflammation, and comedogenesis: an update. Clin Cosmet Investig Dermatol. 2015;8:371–88. https://pubmed.ncbi.nlm.nih.gov/26203267/
Moro T, Brightwell CR, Velarde B, et al. Whey protein hydrolysate increases amino acid uptake, mTORC1 signaling, and protein synthesis in skeletal muscle of healthy young men in a randomized crossover trial. J Nutr. 2019;149(7):1149–58. https://pubmed.ncbi.nlm.nih.gov/31095313/
Melnik BC. Milk – a nutrient system of mammalian evolution promoting mTORC1-dependent translation. Int J Mol Sci. 2015;16(8):17048–87. https://pubmed.ncbi.nlm.nih.gov/26225961/
Melnik BC, John SM, Carrera-Bastos P, Cordain L. The impact of cow’s milk-mediated mTORC1-signaling in the initiation and progression of prostate cancer. Nutr Metab (Lond). 2012;9(1):74. https://pubmed.ncbi.nlm.nih.gov/22891897/
Melnik BC. Milk – a nutrient system of mammalian evolution promoting mTORC1-dependent translation. Int J Mol Sci. 2015;16(8):17048–87. https://pubmed.ncbi.nlm.nih.gov/26225961/
Melnik BC. Lifetime impact of cow’s milk on overactivation of mTORC1: from fetal to childhood overgrowth, acne, diabetes, cancers, and neurodegeneration. Biomolecules. 2021;11(3):404. https://pubmed.ncbi.nlm.nih.gov/33803410/
Melnik BC, John SM, Schmitz G. Milk is not just food but most likely a genetic transfection system activating mTORC1 signaling for postnatal growth. Nutr J. 2013;12:103. https://pubmed.ncbi.nlm.nih.gov/23883112/
Cordain L, Lindeberg S, Hurtado M, Hill K, Eaton SB, Brand-Miller J. Acne vulgaris: a disease of Western civilization. Arch Dermatol. 2002;138(12):1584–90. https://pubmed.ncbi.nlm.nih.gov/12472346/
Danby FW. Acne and milk, the diet myth, and beyond. J Am Acad Dermatol. 2005;52(2):360–2. https://pubmed.ncbi.nlm.nih.gov/15692488/
Aghasi M, Golzarand M, Shab-Bidar S, Aminianfar A, Omidian M, Taheri F. Dairy intake and acne development: a meta-analysis of observational studies. Clin Nutr. 2019;38(3):1067–75. https://pubmed.ncbi.nlm.nih.gov/29778512/
Melnik BC. Linking diet to acne metabolomics, inflammation, and comedogenesis: an update. Clin Cosmet Investig Dermatol. 2015;8:371–88. https://pubmed.ncbi.nlm.nih.gov/26203267/
Melnik BC. Lifetime impact of cow’s milk on overactivation of mTORC1: from fetal to childhood overgrowth, acne, diabetes, cancers, and neurodegeneration. Biomolecules. 2021;11(3):404. https://pubmed.ncbi.nlm.nih.gov/33803410/
Melnik BC. Dietary intervention in acne: attenuation of increased mTORC1 signaling promoted by Western diet. Dermatoendocrinol. 2012;4(1):20–32. https://pubmed.ncbi.nlm.nih.gov/22870349/
Baron JA, Weiderpass E, Newcomb PA, et al. Metabolic disorders and breast cancer risk (United States). Cancer Causes Control. 2001;12(10):875–80. https://pubmed.ncbi.nlm.nih.gov/11808705/
Sutcliffe S, Giovannucci E, Isaacs WB, Willett WC, Platz EA. Acne and risk of prostate cancer. Int J Cancer. 2007;121(12):2688–92. https://pubmed.ncbi.nlm.nih.gov/17724724/
Melnik BC, John SM, Carrera-Bastos P, Cordain L. The impact of cow’s milk-mediated mTORC1-signaling in the initiation and progression of prostate cancer. Nutr Metab (Lond). 2012;9(1):74. https://pubmed.ncbi.nlm.nih.gov/22891897/
Sargsyan A, Dubasi HB. Milk consumption and prostate cancer: a systematic review. World J Mens Health. 2021;39(3):419–28. https://pubmed.ncbi.nlm.nih.gov/32777868/
Pettersson A, Kasperzyk JL, Kenfield SA, et al. Milk and dairy consumption among men with prostate cancer and risk of metastases and prostate cancer death. Cancer Epidemiol Biomarkers Prev. 2012;21(3):428–36. https://pubmed.ncbi.nlm.nih.gov/22315365/
Tognon G, Nilsson LM, Shungin D, et al. Nonfermented milk and other dairy products: associations with all-cause mortality. Am J Clin Nutr. 2017;105(6):1502–11. https://pubmed.ncbi.nlm.nih.gov/28490510/
Melnik BC, Schmitz G. Pasteurized non-fermented cow’s milk but not fermented milk is a promoter of mTORC1-driven aging and increased mortality. Ageing Res Rev. 2021;67:101270. https://pubmed.ncbi.nlm.nih.gov/33571703/
Gao X, Jia H, Chen G, Li C, Hao M. Yogurt intake reduces all-cause and cardiovascular disease mortality: a meta-analysis of eight prospective cohort studies. Chin J Integr Med. 2020;26(6):462–8. https://pubmed.ncbi.nlm.nih.gov/31970674/
Sahin K, Orhan C, Tuzcu M, et al. Tomato powder modulates NF-¿B, mTOR, and Nrf2 pathways during aging in healthy rats. J Aging Res. 2019;2019:1643243. https://pubmed.ncbi.nlm.nih.gov/30719353/
Takeshima M, Ono M, Higuchi T, Chen C, Hara T, Nakano S. Anti-proliferative and apoptosis-inducing activity of lycopene against three subtypes of human breast cancer cell lines. Cancer Sci. 2014;105(3):252–7. https://pubmed.ncbi.nlm.nih.gov/24397737/
Thomson CA, Ho E, Strom MB. Chemopreventive properties of 3,3’-diindolylmethane in breast cancer: evidence from experimental and human studies. Nutr Rev. 2016;74(7):432–43. https://pubmed.ncbi.nlm.nih.gov/27261275/
Du H, Zhang X, Zeng Y, et al. A novel phytochemical, DIM, inhibits proliferation, migration, invasion and TNF-a induced inflammatory cytokine production of synovial fibroblasts from rheumatoid arthritis patients by targeting MAPK and AKT/mTOR signal pathway. Front Immunol. 2019;10:1620. https://pubmed.ncbi.nlm.nih.gov/31396207/
Zhang Y, Gilmour A, Ahn YH, de la Vega L, Dinkova-Kostova AT. The isothiocyanate sulforaphane inhibits mTOR in an NRF2-independent manner. Phytomedicine. 2021;86:153062. https://pubmed.ncbi.nlm.nih.gov/31409554/
Li N, Wu X, Zhuang W, et al. Green leafy vegetable and lutein intake and multiple health outcomes. Food Chem. 2021;360:130145. https://pubmed.ncbi.nlm.nih.gov/34034049/
Sato A. mTOR, a potential target to treat autism spectrum disorder. CNS Neurol Disord Drug Targets. 2016;15(5):533–43. https://pubmed.ncbi.nlm.nih.gov/27071790/
Matusheski NV, Juvik JA, Jeffery EH. Heating decreases epithiospecifier protein activity and increases sulforaphane formation in broccoli. Phytochemistry. 2004;65(9):1273–81. https://pubmed.ncbi.nlm.nih.gov/15184012/
Singh K, Connors SL, Macklin EA, et al. Sulforaphane treatment of autism spectrum disorder (ASD). Proc Natl Acad Sci U S A. 2014;111(43):15550–5. https://pubmed.ncbi.nlm.nih.gov/25313065/
Wanke V, Cameroni E, Uotila A, et al. Caffeine extends yeast lifespan by targeting TORC1. Mol Microbiol. 2008;69(1):277–85. https://pubmed.ncbi.nlm.nih.gov/18513215/
Takahashi K, Yanai S, Shimokado K, Ishigami A. Coffee consumption in aged mice increases energy production and decreases hepatic mTOR levels. Nutrition. 2017;38:1–8. https://pubmed.ncbi.nlm.nih.gov/28526373/
Van Aller GS, Carson JD, Tang W, et al. Epigallocatechin gallate (EGCG), a major component of green tea, is a dual phosphoinositide-3-kinase/mTOR inhibitor. Biochem Biophys Res Commun. 2011;406(2):194–9. https://pubmed.ncbi.nlm.nih.gov/21300025/
Elsaie ML, Abdelhamid MF, Elsaaiee LT, Emam HM. The efficacy of topical 2 % green tea lotion in mild-to-moderate acne vulgaris. J Drugs Dermatol. 2009;8(4):358–64. https://pubmed.ncbi.nlm.nih.gov/19363854/
Cassidy A, Chung M, Zhao N, et al. Dose – response relation between tea consumption and risk of cardiovascular disease and all-cause mortality: a systematic review and meta-analysis of population-based studies. Adv Nutr. 2020;11(4):790–814. https://pubmed.ncbi.nlm.nih.gov/32073596/
Lamming DW. Inhibition of the mechanistic target of rapamycin (mTOR) – rapamycin and beyond. Cold Spring Harb Perspect Med. 2016;6(5). https://pubmed.ncbi.nlm.nih.gov/27048303/
Kennedy BK, Lamming DW. The mechanistic target of rapamycin: the grand conducTOR of metabolism and aging. Cell Metab. 2016;23(6):990–1003. https://pubmed.ncbi.nlm.nih.gov/27304501/
Morley JE. The mTOR conundrum: essential for muscle function, but dangerous for survival. J Am Med Dir Assoc. 2016;17(11):963–6. https://pubmed.ncbi.nlm.nih.gov/27780571/
Blagosklonny MV. Why men age faster but reproduce longer than women: mTOR and evolutionary perspectives. Aging (Albany NY). 2010;2(5):265–73. https://pubmed.ncbi.nlm.nih.gov/20519781/
Markofski MM, Dickinson JM, Drummond MJ, et al. Effect of age on basal muscle protein synthesis and mTORC1 signaling in a large cohort of young and older men and women. Exp Gerontol. 2015;65:1–7. https://pubmed.ncbi.nlm.nih.gov/25735236/
Leenders M, Verdijk LB, van der Hoeven L, et al. Prolonged leucine supplementation does not augment muscle mass or affect glycemic control in elderly type 2 diabetic men. J Nutr. 2011;141(6):1070–6. https://pubmed.ncbi.nlm.nih.gov/21525248/
Verhoeven S, Vanschoonbeek K, Verdijk LB, et al. Long-term leucine supplementation does not increase muscle mass or strength in healthy elderly men. Am J Clin Nutr. 2009;89(5):1468–75. https://pubmed.ncbi.nlm.nih.gov/19321567/
Tang H, Shrager JB, Goldman D. Rapamycin protects aging muscle. Aging (Albany NY). 2019;11(16):5868–70. https://pubmed.ncbi.nlm.nih.gov/31454792/
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21(4):183–203. https://pubmed.ncbi.nlm.nih.gov/31937935/
Kennedy BK, Lamming DW. The mechanistic target of rapamycin: the grand conducTOR of metabolism and aging. Cell Metab. 2016;23(6):990–1003. https://pubmed.ncbi.nlm.nih.gov/27304501/
Тор (Tor) – в германо-скандинавской мифологии бог грома и молний, защищающий богов и людей от великанов и чудовищ с помощью боевого молота (hammer). – Примеч. ред.
Lamming DW, Salmon AB. TORwards a victory over aging. J Gerontol A Biol Sci Med Sci. 2020;75(1):1–3. https://pubmed.ncbi.nlm.nih.gov/31544928/
Caldana C, Martins MCM, Mubeen U, Urrea-Castellanos R. The magic “hammer” of TOR: the multiple faces of a single pathway in the metabolic regulation of plant growth and development. J Exp Bot. 2019;70(8):2217–25. https://pubmed.ncbi.nlm.nih.gov/30722050/
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21(4):183–203. https://pubmed.ncbi.nlm.nih.gov/31937935/
Kaeberlein M, Galvan V. Rapamycin and Alzheimer’s disease: time for a clinical trial? Sci Transl Med. 2019;11(476):eaar4289. https://pubmed.ncbi.nlm.nih.gov/30674654/
Kapahi P, Chen D, Rogers AN, et al. With TOR, less is more: a key role for the conserved nutrient-sensing TOR pathway in aging. Cell Metab. 2010;11(6):453–65. https://pubmed.ncbi.nlm.nih.gov/20519118/
Sansevero TB. The Profit Machine. Cultiva Libros; 2009.
Harman D. The biologic clock: the mitochondria? J Am Geriatr Soc. 1972;20(4):145–7. https://pubmed.ncbi.nlm.nih.gov/5016631/
Talaulikar VS, Manyonda IT. Vitamin C as an antioxidant supplement in women’s health: a myth in need of urgent burial. Eur J Obstet Gynecol Reprod Biol. 2011;157(1):10–3. https://pubmed.ncbi.nlm.nih.gov/21507551/
Liebman SE, Le TH. Eat your broccoli: oxidative stress, NRF2, and sulforaphane in chronic kidney disease. Nutrients. 2021;13(1):266. https://pubmed.ncbi.nlm.nih.gov/33477669/
Peng C, Wang X, Chen J, et al. Biology of ageing and role of dietary antioxidants. Biomed Res Int. 2014;2014:831841. https://pubmed.ncbi.nlm.nih.gov/24804252/
Maes M, Galecki P, Chang YS, Berk M. A review on the oxidative and nitrosative stress (O&NS) pathways in major depression and their possible contribution to the (neuro)degenerative processes in that illness. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(3):676–92. https://pubmed.ncbi.nlm.nih.gov/20471444/
Peng C, Wang X, Chen J, et al. Biology of ageing and role of dietary antioxidants. Biomed Res Int. 2014;2014:831841. https://pubmed.ncbi.nlm.nih.gov/24804252/
Rinnerthaler M, Bischof J, Streubel MK, Trost A, Richter K. Oxidative stress in aging human skin. Biomolecules. 2015;5(2):545–89. https://pubmed.ncbi.nlm.nih.gov/25906193/
Logan S, Royce GH, Owen D, et al. Accelerated decline in cognition in a mouse model of increased oxidative stress. GeroScience. 2019;41(5):591–607. https://pubmed.ncbi.nlm.nih.gov/31641924/
Hensley K, Floyd RA. Reactive oxygen species and protein oxidation in aging: a look back, a look ahead. Arch Biochem Biophys. 2002;397(2):377–83. https://pubmed.ncbi.nlm.nih.gov/11795897/
Yeung AWK, Tzvetkov NT, El-Tawil OS, Bungau SG, Abdel-Daim MM, Atanasov AG. Antioxidants: scientific literature landscape analysis. Oxid Med Cell Longev. 2019;2019:8278454. https://pubmed.ncbi.nlm.nih.gov/30728893/
Bast A, Haenen GRMM. Ten misconceptions about antioxidants. Trends Pharmacol Sci. 2013;34(8):430–6. https://pubmed.ncbi.nlm.nih.gov/23806765/
Medvedev ZA. An attempt at a rational classification of theories of ageing. Biol Rev. 1990;65(3):375–98. https://pubmed.ncbi.nlm.nih.gov/2205304/
Fusco D, Colloca G, Lo Monaco MR, Cesari M. Effects of antioxidant supplementation on the aging process. Clin Interv Aging. 2007;2(3):377–87. https://pubmed.ncbi.nlm.nih.gov/18044188/
Barja G. Updating the mitochondrial free radical theory of aging: an integrated view, key aspects, and confounding concepts. Antioxid Redox Signal. 2013;19(12):1420–45. https://pubmed.ncbi.nlm.nih.gov/23642158/
Golubev A, Hanson AD, Gladyshev VN. A tale of two concepts: harmonizing the free radical and antagonistic pleiotropy theories of aging. Antioxid Redox Signal. 2018;29(10):1003–17. https://pubmed.ncbi.nlm.nih.gov/28874059/
Harman D. Aging: a theory based on free radical and radiation chemistry. J Gerontol. 1956;11(3):298–300. https://pubmed.ncbi.nlm.nih.gov/13332224/
Biesalski HK. Free radical theory of aging. Curr Opin Clin Nutr Metab Care. 2002;5(1):5–10. https://pubmed.ncbi.nlm.nih.gov/11790942/
Keane M, Semeiks J, Webb AE, et al. Insights into the evolution of longevity from the bowhead whale genome. Cell Rep. 2015;10(1):112–22. https://pubmed.ncbi.nlm.nih.gov/25565328/
.
Butler PG, Wanamaker AD Jr, Scourse JD, Richardson CA, Reynolds DJ. Variability of marine climate on the North Icelandic shelf in a 1357-year proxy archive based on growth increments in the bivalve Arctica islandica. Palaeogeogr, Palaeoclimatol, Palaeoecol. 2013;373:141–51. https://www.sciencedirect.com/science/article/abs/pii/S0031018212000302?via%3Dihub
Barja G. Updating the mitochondrial free radical theory of aging: an integrated view, key aspects, and confounding concepts. Antioxid Redox Signal. 2013;19(12):1420–45. https://pubmed.ncbi.nlm.nih.gov/23642158/
Barja G. Updating the mitochondrial free radical theory of aging: an integrated view, key aspects, and confounding concepts. Antioxid Redox Signal. 2013;19(12):1420–45. https://pubmed.ncbi.nlm.nih.gov/23642158/
Capt C, Passamonti M, Breton S. The human mitochondrial genome may code for more than 13 proteins. Mitochondrial DNA Part A. 2016;27(5):3098–101. https://pubmed.ncbi.nlm.nih.gov/25630734/
Willyard C. New human gene tally reignites debate. Nature. 2018;558(7710):354–5. https://pubmed.ncbi.nlm.nih.gov/29921859/
Venditti P, Masullo P, Di Meo S. Effect of training on H2O2 release by mitochondria from rat skeletal muscle. Arch Biochem Biophys. 1999;372(2):315–20. https://pubmed.ncbi.nlm.nih.gov/10600170/
Barja G. Updating the mitochondrial free radical theory of aging: an integrated view, key aspects, and confounding concepts. Antioxid Redox Signal. 2013;19(12):1420–45. https://pubmed.ncbi.nlm.nih.gov/23642158/
Ruiz MC, Ayala V, Portero-Otín M, Requena JR, Barja G, Pamplona R. Protein methionine content and MDA-lysine adducts are inversely related to maximum life span in the heart of mammals. Mech Ageing Dev. 2005;126(10):1106–14. https://pubmed.ncbi.nlm.nih.gov/15955547/
Gomez J, Sanchez-Roman I, Gomez A, et al. Methionine and homocysteine modulate the rate of ROS generation of isolated mitochondria in vitro. J Bioenerg Biomembr. 2011;43(4):377–86. https://pubmed.ncbi.nlm.nih.gov/21748404/
Barja G. Updating the mitochondrial free radical theory of aging: an integrated view, key aspects, and confounding concepts. Antioxid Redox Signal. 2013;19(12):1420–45. https://pubmed.ncbi.nlm.nih.gov/23642158/
Barja G. The mitochondrial free radical theory of aging. Prog Mol Biol Transl Sci. 2014;127:1–27. https://pubmed.ncbi.nlm.nih.gov/25149212/
Sanz A, Stefanatos RKA. The mitochondrial free radical theory of aging: a critical view. Curr Aging Sci. 2008;1(1):10–21. https://pubmed.ncbi.nlm.nih.gov/20021368/
Sanz A, Caro P, Ayala V, Portero-Otin M, Pamplona R, Barja G. Methionine restriction decreases mitochondrial oxygen radical generation and leak as well as oxidative damage to mitochondrial DNA and proteins. FASEB J. 2006;20(8):1064–73. https://pubmed.ncbi.nlm.nih.gov/16770005/
Barja G. Updating the mitochondrial free radical theory of aging: an integrated view, key aspects, and confounding concepts. Antioxid Redox Signal. 2013;19(12):1420–45. https://pubmed.ncbi.nlm.nih.gov/23642158/
Barja G. The mitochondrial free radical theory of aging. Prog Mol Biol Transl Sci. 2014;127:1–27. https://pubmed.ncbi.nlm.nih.gov/25149212/
López-Torres M, Barja G. Lowered methionine ingestion as responsible for the decrease in rodent mitochondrial oxidative stress in protein and dietary restriction possible implications for humans. Biochim Biophys Acta. 2008;1780(11):1337–47. https://pubmed.ncbi.nlm.nih.gov/18252204/
What we eat in America, NHANES 2017–2018. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/1718/tables_1–36%20and%2041–56_2017–2018.pdf. Published 2020. Accessed July 6, 2021.; https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/1718/wweia_2017_2018_data.pdf
López-Torres M, Barja G. Lowered methionine ingestion as responsible for the decrease in rodent mitochondrial oxidative stress in protein and dietary restriction possible implications for humans. Biochim Biophys Acta. 2008;1780(11):1337–47. https://pubmed.ncbi.nlm.nih.gov/18252204/
Fontana L, Cummings NE, Arriola Apelo SI, et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 2016;16(2):520–30. https://pubmed.ncbi.nlm.nih.gov/27346343/
Barja G. The mitochondrial free radical theory of aging. Prog Mol Biol Transl Sci. 2014;127:1–27. https://pubmed.ncbi.nlm.nih.gov/25149212/
López-Torres M, Barja G. Lowered methionine ingestion as responsible for the decrease in rodent mitochondrial oxidative stress in protein and dietary restriction possible implications for humans. Biochim Biophys Acta. 2008;1780(11):1337–47. https://pubmed.ncbi.nlm.nih.gov/18252204/
Darmadi-Blackberry I, Wahlqvist ML, Kouris-Blazos A, et al. Legumes: the most important dietary predictor of survival in older people of different ethnicities. Asia Pac J Clin Nutr. 2004;13(2):217–20. https://pubmed.ncbi.nlm.nih.gov/15228991/
Buettner D. The Blue Zones: 9 Lessons for Living Longer from the People Who’ve Lived the Longest. 2nd ed. National Geographic Books; 2012. https://www.worldcat.org/title/777659970
McCarty MF, Barroso-Aranda J, Contreras F. The low-methionine content of vegan diets may make methionine restriction feasible as a life extension strategy. Med Hypotheses. 2009;72(2):125–8. https://pubmed.ncbi.nlm.nih.gov/18789600/
Scudellari M. Myths that will not die. Nature. 2015;528(7582):322–5. https://pubmed.ncbi.nlm.nih.gov/26672537/
Stuart JA, Maddalena LA, Merilovich M, Robb EL. A midlife crisis for the mitochondrial free radical theory of aging. Longev Healthspan. 2014;3(1):4. https://pubmed.ncbi.nlm.nih.gov/24690218/
Golubev A, Hanson AD, Gladyshev VN. A tale of two concepts: harmonizing the free radical and antagonistic pleiotropy theories of aging. Antioxid Redox Signal. 2018;29(10):1003–17. https://pubmed.ncbi.nlm.nih.gov/28874059/
Bjelakovic G, Nikolova D, Gluud C. Antioxidant supplements and mortality. Curr Opin Clin Nutr Metab Care. 2014;17(1):40–4. https://pubmed.ncbi.nlm.nih.gov/24241129/
Bjelakovic G, Nikolova D, Simonetti RG, Gluud C. Antioxidant supplements for prevention of gastrointestinal cancers: a systematic review and meta-analysis. Lancet. 2004;364(9441):1219–28. https://pubmed.ncbi.nlm.nih.gov/15464182/
Serafini M, Jakszyn P, Luján-Barroso L, et al. Dietary total antioxidant capacity and gastric cancer risk in the European prospective investigation into cancer and nutrition study. Int J Cancer. 2012;131(4):E544–54. https://pubmed.ncbi.nlm.nih.gov/22072493/
Jacobs DR, Tapsell LC. Food synergy: the key to a healthy diet. Proc Nutr Soc. 2013;72(2):200–6. https://pubmed.ncbi.nlm.nih.gov/23312372/
Cömert ED, Gökmen V. Evolution of food antioxidants as a core topic of food science for a century. Food Res Int. 2018;105:76–93. https://pubmed.ncbi.nlm.nih.gov/29433271/
Barja G. Updating the mitochondrial free radical theory of aging: an integrated view, key aspects, and confounding concepts. Antioxid Redox Signal. 2013;19(12):1420–45. https://pubmed.ncbi.nlm.nih.gov/23642158/
Chial H, Craig J. mtDNA and mitochondrial diseases. Nature Education. 2008;1(1):217. https://www.nature.com/scitable/topicpage/mtdna-and-mitochondrial-diseases-903/
Tubbs A, Nussenzweig A. Endogenous DNA damage as a source of genomic instability in cancer. Cell. 2017;168(4):644–56. https://pubmed.ncbi.nlm.nih.gov/28187286/
Patel J, Baptiste BA, Kim E, Hussain M, Croteau DL, Bohr VA. DNA damage and mitochondria in cancer and aging. Carcinogenesis. 2020;41(12):1625–34. https://pubmed.ncbi.nlm.nih.gov/33146705/
Soares JP, Cortinhas A, Bento T, et al. Aging and DNA damage in humans: a meta-analysis study. Aging (Albany NY). 2014;6(6):432–9. https://pubmed.ncbi.nlm.nih.gov/25140379/
Belenguer-Varea Á, Tarazona-Santabalbina FJ, Avellana-Zaragoza JA, Martínez-Reig M, Mas-Bargues C, Inglés M. Oxidative stress and exceptional human longevity: systematic review. Free Radic Biol Med. 2020;149:51–63. https://pubmed.ncbi.nlm.nih.gov/31550529/
Patel J, Baptiste BA, Kim E, Hussain M, Croteau DL, Bohr VA. DNA damage and mitochondria in cancer and aging. Carcinogenesis. 2020;41(12):1625–34. https://pubmed.ncbi.nlm.nih.gov/33146705/
Yousefzadeh M, Henpita C, Vyas R, Soto-Palma C, Robbins P, Niedernhofer L. DNA damage – how and why we age? Elife. 2021;10:e62852. https://pubmed.ncbi.nlm.nih.gov/33512317/
Liochev SI. Reflections on the theories of aging, of oxidative stress, and of science in general. Is it time to abandon the free radical (oxidative stress) theory of aging? Antioxid Redox Signal. 2015;23(3):187–207. https://pubmed.ncbi.nlm.nih.gov/24949668/
Belenguer-Varea Á, Tarazona-Santabalbina FJ, Avellana-Zaragoza JA, Martínez-Reig M, Mas-Bargues C, Inglés M. Oxidative stress and exceptional human longevity: systematic review. Free Radic Biol Med. 2020;149:51–63. https://pubmed.ncbi.nlm.nih.gov/31550529/
Liguori I, Russo G, Curcio F, et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018;13:757–72. https://pubmed.ncbi.nlm.nih.gov/29731617/
Belenguer-Varea Á, Tarazona-Santabalbina FJ, Avellana-Zaragoza JA, Martínez-Reig M, Mas-Bargues C, Inglés M. Oxidative stress and exceptional human longevity: systematic review. Free Radic Biol Med. 2020;149:51–63. https://pubmed.ncbi.nlm.nih.gov/31550529/
Salmon AB, Richardson A, Pérez VI. Update on the oxidative stress theory of aging: does oxidative stress play a role in aging or healthy aging? Free Radic Biol Med. 2010;48(5):642–55. https://pubmed.ncbi.nlm.nih.gov/20036736/
Edrey YH, Salmon AB. Revisiting an age-old question regarding oxidative stress. Free Radic Biol Med. 2014;71:368–78. https://pubmed.ncbi.nlm.nih.gov/24704971/
Cannon G. Nutritional science for this century. Public Health Nutr. 2005;8(4):344–7. https://pubmed.ncbi.nlm.nih.gov/15975178/
Andrews P. Last common ancestor of apes and humans: morphology and environment. FPR. 2020;91(2):122–48. https://pubmed.ncbi.nlm.nih.gov/31533109/
Milton K. Nutritional characteristics of wild primate foods: do the diets of our closest living relatives have lessons for us? Nutrition. 1999;15(6):488–98. https://pubmed.ncbi.nlm.nih.gov/10378206/
Milton K. Back to basics: why foods of wild primates have relevance for modern human health. Nutrition. 2000;16(7–8):480–3. https://pubmed.ncbi.nlm.nih.gov/10906529/
Milton K. Hunter-gatherer diets: a different perspective. Am J Clin Nutr. 2000;71(3):665–7. https://pubmed.ncbi.nlm.nih.gov/10702155/
Milton K. Micronutrient intakes of wild primates: are humans different? Comp Biochem Physiol A Mol Integr Physiol. 2003;136(1):47–59. https://pubmed.ncbi.nlm.nih.gov/14527629/
Benzie IFF. Evolution of dietary antioxidants. Comp Biochem Physiol A Mol Integr Physiol. 2003;136(1):113–26. https://pubmed.ncbi.nlm.nih.gov/14527634/
Milton K. Nutritional characteristics of wild primate foods: do the diets of our closest living relatives have lessons for us? Nutrition. 1999;15(6):488–98. https://pubmed.ncbi.nlm.nih.gov/10378206/
Benzie IFF. Evolution of dietary antioxidants. Comp Biochem Physiol A Mol Integr Physiol. 2003;136(1):113–26. https://pubmed.ncbi.nlm.nih.gov/14527634/
Milton K. Nutritional characteristics of wild primate foods: do the diets of our closest living relatives have lessons for us? Nutrition. 1999;15(6):488–98. https://pubmed.ncbi.nlm.nih.gov/10378206/
Milton K. Micronutrient intakes of wild primates: are humans different? Comp Biochem Physiol A Mol Integr Physiol. 2003;136(1):47–59. https://pubmed.ncbi.nlm.nih.gov/14527629/
Benzie IFF. Evolution of dietary antioxidants. Comp Biochem Physiol A Mol Integr Physiol. 2003;136(1):113–26. https://pubmed.ncbi.nlm.nih.gov/14527634/
Schuch AP, Moreno NC, Schuch NJ, Menck CFM, Garcia CCM. Sunlight damage to cellular DNA: focus on oxidatively generated lesions. Free Radic Biol Med. 2017;107:110–24. https://pubmed.ncbi.nlm.nih.gov/28109890/
Benzie IFF. Evolution of dietary antioxidants. Comp Biochem Physiol Part A Mol Integr Physiol. 2003;136(1):113–26. https://pubmed.ncbi.nlm.nih.gov/14527634/
Benzie IFF. Evolution of dietary antioxidants. Comp Biochem Physiol Part A Mol Integr Physiol. 2003;136(1):113–26. https://pubmed.ncbi.nlm.nih.gov/14527634/
Coffey DS. Similarities of prostate and breast cancer: evolution, diet, and estrogens. Urology. 2001;57(4 Suppl 1):31–8. https://pubmed.ncbi.nlm.nih.gov/11295592/
Jallinoja P, Niva M, Helakorpi S, Kahma N. Food choices, perceptions of healthiness, and eating motives of self-identified followers of a low-carbohydrate diet. Food Nutr Res. 2014;58:23552. https://pubmed.ncbi.nlm.nih.gov/25490960/
Nestle M. Paleolithic diets: a sceptical view. Nutr Bull. 2000;25:43–7. https://nyuscholars.nyu.edu/en/publications/paleolithic-diets-a-sceptical-view
Vatner SF, Zhang J, Oydanich M, Berkman T, Naftalovich R, Vatner DE. Healthful aging mediated by inhibition of oxidative stress. Ageing Res Rev. 2020;64:101194. https://pubmed.ncbi.nlm.nih.gov/33091597/
Abbasalizad Farhangi M, Vajdi M. Dietary total antioxidant capacity (TAC) significantly reduces the risk of site-specific cancers: an updated systematic review and meta-analysis. Nutr Cancer. 2021;73(5):721–39. https://pubmed.ncbi.nlm.nih.gov/32462920/
Parohan M, Anjom-Shoae J, Nasiri M, Khodadost M, Khatibi SR, Sadeghi O. Dietary total antioxidant capacity and mortality from all causes, cardiovascular disease and cancer: a systematic review and dose-response meta-analysis of prospective cohort studies. Eur J Nutr. 2019;58(6):2175–89. https://pubmed.ncbi.nlm.nih.gov/30756144/
Jayedi A, Rashidy-Pour A, Parohan M, Zargar MS, Shab-Bidar S. Dietary antioxidants, circulating antioxidant concentrations, total antioxidant capacity, and risk of all-cause mortality: a systematic review and dose-response meta-analysis of prospective observational studies. Adv Nutr. 2018;9(6):701–16. https://pubmed.ncbi.nlm.nih.gov/30239557/
Carlsen MH, Halvorsen BL, Holte K, et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 2010;9:3. https://pubmed.ncbi.nlm.nih.gov/20096093/
Yang M, Chung SJ, Chung CE, et al. Estimation of total antioxidant capacity from diet and supplements in US adults. Br J Nutr. 2011;106(2):254–63. https://pubmed.ncbi.nlm.nih.gov/21320369/
Carlsen MH, Halvorsen BL, Holte K, et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 2010 Jan 22;9:3. https://pubmed.ncbi.nlm.nih.gov/20096093/
Bastin S, Henken K. Water content of fruits and vegetables. University of Kentucky College of Agriculture Cooperative Extension Service. https://www.academia.edu/5729963/Water_Content_of_Fruits_and_Vegetables. Published December 1997. Accessed November 11, 2021.; https://www.academia.edu/5729963/Water_Content_of_Fruits_and_Vegetables
Cao G, Prior RL. Comparison of different analytical methods for assessing total antioxidant capacity of human serum. Clin Chem. 1998;44(6 Pt 1):1309–15. https://pubmed.ncbi.nlm.nih.gov/9625058/
Halliwell B. The antioxidant paradox: less paradoxical now? Br J Clin Pharmacol. 2013;75(3):637–44. https://pubmed.ncbi.nlm.nih.gov/22420826/
van Poppel G, Poulsen H, Loft S, Verhagen H. No influence of beta carotene on oxidative DNA damage in male smokers. J Natl Cancer Inst. 1995;87(4):310–1. https://pubmed.ncbi.nlm.nih.gov/7707423/
Priemé H, Loft S, Nyyssönen K, Salonen JT, Poulsen HE. No effect of supplementation with vitamin E, ascorbic acid, or coenzyme Q10 on oxidative DNA damage estimated by 8-oxo-7,8-dihydro-2’-deoxyguanosine excretion in smokers. Am J Clin Nutr. 1997;65(2):503–7. https://pubmed.ncbi.nlm.nih.gov/9022536/
Cao G, Booth SL, Sadowski JA, Prior RL. Increases in human plasma antioxidant capacity after consumption of controlled diets high in fruit and vegetables. Am J Clin Nutr. 1998;68(5):1081–7. https://pubmed.ncbi.nlm.nih.gov/9808226/
Johnson SA, Feresin RG, Navaei N, et al. Effects of daily blueberry consumption on circulating biomarkers of oxidative stress, inflammation, and antioxidant defense in postmenopausal women with pre-and stage 1-hypertension: a randomized controlled trial. Food Funct. 2017;8(1):372–80. https://pubmed.ncbi.nlm.nih.gov/28059417/
Verhagen H, Poulsen HE, Loft S, van Poppel G, Willems MI, van Bladeren PJ. Reduction of oxidative DNA-damage in humans by brussels sprouts. Carcinogenesis. 1995;16(4):969–70. https://pubmed.ncbi.nlm.nih.gov/7728983/
Jayedi A, Rashidy-Pour A, Parohan M, Zargar MS, Shab-Bidar S. Dietary antioxidants, circulating antioxidant concentrations, total antioxidant capacity, and risk of all-cause mortality: a systematic review and dose-response meta-analysis of prospective observational studies. Adv Nutr. 2018;9(6):701–16. https://pubmed.ncbi.nlm.nih.gov/30239557/
Ha K, Kim K, Sakaki JR, Chun OK. Relative validity of dietary total antioxidant capacity for predicting all-cause mortality in comparison to diet quality indexes in US adults. Nutrients. 2020;12(5):1210. https://pubmed.ncbi.nlm.nih.gov/32344879/
Bastide N, Dartois L, Dyevre V, et al. Dietary antioxidant capacity and all-cause and cause-specific mortality in the E3N/EPIC cohort study. Eur J Nutr. 2017;56(3):1233–43. https://pubmed.ncbi.nlm.nih.gov/26887577/
Yang M, Chung SJ, Chung CE, et al. Estimation of total antioxidant capacity from diet and supplements in US adults. Br J Nutr. 2011;106(2):254–63. https://pubmed.ncbi.nlm.nih.gov/21320369/
Bastide N, Dartois L, Dyevre V, et al. Dietary antioxidant capacity and all-cause and cause-specific mortality in the E3N/EPIC cohort study. Eur J Nutr. 2017;56(3):1233–43. https://pubmed.ncbi.nlm.nih.gov/26887577/
Mohanty P, Hamouda W, Garg R, Aljada A, Ghanim H, Dandona P. Glucose challenge stimulates reactive oxygen species (ROS) generation by leucocytes. J Clin Endocrinol Metab. 2000;85(8):2970–3. https://pubmed.ncbi.nlm.nih.gov/10946914/
Prior RL, Gu L, Wu X, et al. Plasma antioxidant capacity changes following a meal as a measure of the ability of a food to alter in vivo antioxidant status. J Am Coll Nutr. 2007;26(2):170–81. https://pubmed.ncbi.nlm.nih.gov/17536129/
Darvin ME, Patzelt A, Knorr F, Blume-Peytavi U, Sterry W, Lademann J. One-year study on the variation of carotenoid antioxidant substances in living human skin: influence of dietary supplementation and stress factors. J Biomed Opt. 2008;13(4):044028. https://pubmed.ncbi.nlm.nih.gov/19021355/
Blacker BC, Snyder SM, Eggett DL, Parker TL. Consumption of blueberries with a high-carbohydrate, low-fat breakfast decreases postprandial serum markers of oxidation. Br J Nutr. 2013;109(9):1670–7. https://pubmed.ncbi.nlm.nih.gov/22935321/
Nair AR, Mariappan N, Stull AJ, Francis J. Blueberry supplementation attenuates oxidative stress within monocytes and modulates immune cell levels in adults with metabolic syndrome: a randomized, double-blind, placebo-controlled trial. Food Funct. 2017;8(11):4118–28. https://pubmed.ncbi.nlm.nih.gov/29019365/
Del Bó C, Riso P, Campolo J, et al. A single portion of blueberry (Vaccinium corymbosum L) improves protection against DNA damage but not vascular function in healthy male volunteers. Nutr Res. 2013;33(3):220–7. https://pubmed.ncbi.nlm.nih.gov/29019365/
Szeto YT, Chu WK, Benzie IFF. Antioxidants in fruits and vegetables: a study of cellular availability and direct effects on human DNA. Biosci Biotechnol Biochem. 2006;70(10):2551–5. https://pubmed.ncbi.nlm.nih.gov/17031063/
López-Uriarte P, Nogués R, Saez G, et al. Effect of nut consumption on oxidative stress and the endothelial function in metabolic syndrome. Clin Nutr. 2010;29(3):373–80. https://pubmed.ncbi.nlm.nih.gov/20064680/
Porrini M, Riso P. Lymphocyte lycopene concentration and DNA protection from oxidative damage is increased in women after a short period of tomato consumption. J Nutr. 2000;130(2):189–92. https://pubmed.ncbi.nlm.nih.gov/10720168/
Porrini M, Riso P, Oriani G. Spinach and tomato consumption increases lymphocyte DNA resistance to oxidative stress but this is not related to cell carotenoid concentrations. Eur J Nutr. 2002;41(3):95–100. https://pubmed.ncbi.nlm.nih.gov/12111045/
Frugé AD, Smith KS, Riviere AJ, et al. A dietary intervention high in green leafy vegetables reduces oxidative DNA damage in adults at increased risk of colorectal cancer: biological outcomes of the randomized controlled meat and three greens (M3G) feasibility trial. Nutrients. 2021;13(4):1220. https://pubmed.ncbi.nlm.nih.gov/33917165/
Pool-Zobel BL, Bub A, Müller H, Wollowski I, Rechkemmer G. Consumption of vegetables reduces genetic damage in humans: first results of a human intervention trial with carotenoid-rich foods. Carcinogenesis. 1997;18(9):1847–50. https://pubmed.ncbi.nlm.nih.gov/9328185/
Hoelzl C, Glatt H, Meinl W, et al. Consumption of Brussels sprouts protects peripheral human lymphocytes against 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) and oxidative DNA-damage: results of a controlled human intervention trial. Mol Nutr Food Res. 2008;52(3):330–41. https://pubmed.ncbi.nlm.nih.gov/18293303/
Fogarty MC, Hughes CM, Burke G, Brown JC, Davison GW. Acute and chronic watercress supplementation attenuates exercise-induced peripheral mononuclear cell DNA damage and lipid peroxidation. Br J Nutr. 2013;109(2):293–301. https://pubmed.ncbi.nlm.nih.gov/22475430/
Han KC, Wong WC, Benzie IFF. Genoprotective effects of green tea (Camellia sinensis) in human subjects: results of a controlled supplementation trial. Br J Nutr. 2011;105(2):171–9. https://pubmed.ncbi.nlm.nih.gov/20807462/
Pool-Zobel BL, Bub A, Müller H, Wollowski I, Rechkemmer G. Consumption of vegetables reduces genetic damage in humans: first results of a human intervention trial with carotenoid-rich foods. Carcinogenesis. 1997;18(9):1847–50. https://pubmed.ncbi.nlm.nih.gov/9328185/
Szeto YT, To TL, Pak SC, Kalle W. A study of DNA protective effect of orange juice supplementation. Appl Physiol Nutr Metab. 2013;38(5):533–6. https://pubmed.ncbi.nlm.nih.gov/23668761/
Guarnieri S, Riso P, Porrini M. Orange juice vs vitamin C: effect on hydrogen peroxide-induced DNA damage in mononuclear blood cells. Br J Nutr. 2007;97(4):639–43. https://pubmed.ncbi.nlm.nih.gov/17349075/
Pool-Zobel BL, Bub A, Müller H, Wollowski I, Rechkemmer G. Consumption of vegetables reduces genetic damage in humans: first results of a human intervention trial with carotenoid-rich foods. Carcinogenesis. 1997;18(9):1847–50. https://pubmed.ncbi.nlm.nih.gov/9328185/
Collins BH, Horská A, Hotten PM, Riddoch C, Collins AR. Kiwifruit protects against oxidative DNA damage in human cells and in vitro. Nutr Cancer. 2001;39(1):148–53. https://pubmed.ncbi.nlm.nih.gov/11588897/
Collins AR, Harrington V, Drew J, Melvin R. Nutritional modulation of DNA repair in a human intervention study. Carcinogenesis. 2003;24(3):511–5. https://pubmed.ncbi.nlm.nih.gov/12663512/
Collins AR, Harrington V, Drew J, Melvin R. Nutritional modulation of DNA repair in a human intervention study. Carcinogenesis. 2003;24(3):511–5. https://pubmed.ncbi.nlm.nih.gov/12663512/
Astley SB, Elliott RM, Archer DB, Southon S. Evidence that dietary supplementation with carotenoids and carotenoid-rich foods modulates the DNA damage: repair balance in human lymphocytes. Br J Nutr. 2004;91(1):63–72. https://pubmed.ncbi.nlm.nih.gov/14748939/
Ho CK, Choi SW, Siu PM, Benzie IFF. Effects of single dose and regular intake of green tea (Camellia sinensis) on DNA damage, DNA repair, and heme oxygenase-1 expression in a randomized controlled human supplementation study. Mol Nutr Food Res. 2014;58(6):1379–83. https://pubmed.ncbi.nlm.nih.gov/24585444/
Collins AR, Azqueta A, Langie SAS. Effects of micronutrients on DNA repair. Eur J Nutr. 2012;51(3):261–79. https://pubmed.ncbi.nlm.nih.gov/22362552/
Astley SB, Elliott RM, Archer DB, Southon S. Evidence that dietary supplementation with carotenoids and carotenoid-rich foods modulates the DNA damage: repair balance in human lymphocytes. Br J Nutr. 2004;91(1):63–72. https://pubmed.ncbi.nlm.nih.gov/14748939/
Vayndorf EM, Lee SS, Liu RH. Whole apple extracts increase lifespan, healthspan and resistance to stress in Caenorhabditis elegans. J Funct Foods. 2013;5(3):1236–43. https://pubmed.ncbi.nlm.nih.gov/23878618/
Wang J, Deng N, Wang H, et al. Effects of orange extracts on longevity, healthspan, and stress resistance in Caenorhabditis elegans. Molecules. 2020;25(2):351. https://pubmed.ncbi.nlm.nih.gov/31952185/
Wang E, Wink M. Chlorophyll enhances oxidative stress tolerance in Caenorhabditis elegans and extends its lifespan. PeerJ. 2016;4:e1879. https://pubmed.ncbi.nlm.nih.gov/27077003/
Salehi B, Azzini E, Zucca P, et al. Plant-derived bioactives and oxidative stress-related disorders: a key trend towards healthy aging and longevity promotion. Appl Sci. 2020;10(3):947. https://www.mdpi.com/2076-3417/10/3/947
Saul N, Pietsch K, Stürzenbaum SR, Menzel R, Steinberg CEW. Diversity of polyphenol action in Caenorhabditis elegans: between toxicity and longevity. J Nat Prod. 2011;74(8):1713–20. https://pubmed.ncbi.nlm.nih.gov/21805983/
Ferk F, Chakraborty A, Jäger W, et al. Potent protection of gallic acid against DNA oxidation: results of human and animal experiments. Mutat Res. 2011;715(1–2):61–71. https://pubmed.ncbi.nlm.nih.gov/21827773/
Ferk F, Kundi M, Brath H, et al. Gallic acid improves health-associated biochemical parameters and prevents oxidative damage of DNA in type 2 diabetes patients: results of a placebo-controlled pilot study. Mol Nutr Food Res. 2018;62(4). https://pubmed.ncbi.nlm.nih.gov/29193677/
Vayndorf EM, Lee SS, Liu RH. Whole apple extracts increase lifespan, healthspan and resistance to stress in Caenorhabditis elegans. J Funct Foods. 2013;5(3):1236–43. https://pubmed.ncbi.nlm.nih.gov/23878618/
Kampkötter A, Timpel C, Zurawski RF, et al. Increase of stress resistance and lifespan of Caenorhabditis elegans by quercetin. Comp Biochem Physiol B Biochem Mol Biol. 2008;149(2):314–23. https://pubmed.ncbi.nlm.nih.gov/18024103/
Shimizu C, Wakita Y, Inoue T, et al. Effects of lifelong intake of lemon polyphenols on aging and intestinal microbiome in the senescence-accelerated mouse prone 1 (SAMP1). Sci Rep. 2019;9(1):3671. https://pubmed.ncbi.nlm.nih.gov/30842523/
Rawal S, Singh P, Gupta A, Mohanty S. Dietary intake of Curcuma longa and Emblica officinalis increases life span in Drosophila melanogaster. Biomed Res Int. 2014;2014:910290. https://pubmed.ncbi.nlm.nih.gov/24967413/
Chattopadhyay D, Thirumurugan K. Longevity promoting efficacies of different plant extracts in lower model organisms. Mech Ageing Dev. 2018;171:47–57. https://pubmed.ncbi.nlm.nih.gov/29526449/
Bahadorani S, Hilliker AJ. Cocoa confers life span extension in Drosophila melanogaster. Nutr Res. 2008;28(6):377–82. https://pubmed.ncbi.nlm.nih.gov/19083435/
Rawal S, Singh P, Gupta A, Mohanty S. Dietary intake of Curcuma longa and Emblica officinalis increases life span in Drosophila melanogaster. Biomed Res Int. 2014;2014:910290. https://pubmed.ncbi.nlm.nih.gov/24967413/
Parohan M, Anjom-Shoae J, Nasiri M, Khodadost M, Khatibi SR, Sadeghi O. Dietary total antioxidant capacity and mortality from all causes, cardiovascular disease and cancer: a systematic review and dose-response meta-analysis of prospective cohort studies. Eur J Nutr. 2019;58(6):2175–89. https://pubmed.ncbi.nlm.nih.gov/30756144/
Percival SS, Vanden Heuvel JP, Nieves CJ, Montero C, Migliaccio AJ, Meadors J. Bioavailability of herbs and spices in humans as determined by ex vivo inflammatory suppression and DNA strand breaks. J Am Coll Nutr. 2012;31(4):288–94. https://pubmed.ncbi.nlm.nih.gov/23378457/
Kapoor MP, Suzuki K, Derek T, Ozeki M, Okubo T. Clinical evaluation of Emblica Officinalis Gatertn (Amla) in healthy human subjects: health benefits and safety results from a randomized, double-blind, crossover placebo-controlled study. Contemp Clin Trials Commun. 2020;17:100499. https://pubmed.ncbi.nlm.nih.gov/31890983/
Carlsen MH, Halvorsen BL, Holte K, et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 2010;9:3. https://pubmed.ncbi.nlm.nih.gov/20096093/
Carlsen MH, Halvorsen BL, Holte K, et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 2010;9:3. https://pubmed.ncbi.nlm.nih.gov/20096093/
Zhu C, Yan H, Zheng Y, Santos HO, Macit MS, Zhao K. Impact of cinnamon supplementation on cardiometabolic biomarkers of inflammation and oxidative stress: a systematic review and meta-analysis of randomized controlled trials. Complement Ther Med. 2020;53:102517. https://pubmed.ncbi.nlm.nih.gov/33066854/
Ninfali P, Mea G, Giorgini S, Rocchi M, Bacchiocca M. Antioxidant capacity of vegetables, spices and dressings relevant to nutrition. Br J Nutr. 2005;93(2):257–66. https://pubmed.ncbi.nlm.nih.gov/15788119/
Morvaridzadeh M, Sadeghi E, Agah S, et al. Effect of ginger (Zingiber officinale) supplementation on oxidative stress parameters: a systematic review and meta-analysis. J Food Biochem. 2021;45(2):e13612. https://pubmed.ncbi.nlm.nih.gov/33458848/
Askari M, Mozaffari H, Darooghegi Mofrad M, et al. Effects of garlic supplementation on oxidative stress and antioxidative capacity biomarkers: a systematic review and meta-analysis of randomized controlled trials. Phytother Res. 2021;35(6):3032–45. https://pubmed.ncbi.nlm.nih.gov/33484037/
Carlsen MH, Halvorsen BL, Holte K, et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 2010;9:3. https://pubmed.ncbi.nlm.nih.gov/20096093/
Mehrabani S, Arab A, Mohammadi H, Amani R. The effect of cocoa consumption on markers of oxidative stress: a systematic review and meta-analysis of interventional studies. Complement Ther Med. 2020;48:102240. https://pubmed.ncbi.nlm.nih.gov/31987247/
Grassi D, Desideri G, Necozione S, et al. Cocoa consumption dose-dependently improves flow-mediated dilation and arterial stiffness decreasing blood pressure in healthy individuals. J Hypertens. 2015;33(2):294–303. https://pubmed.ncbi.nlm.nih.gov/25380152/
Taubert D, Berkels R, Roesen R, Klaus W. Chocolate and blood pressure in elderly individuals with isolated systolic hypertension. JAMA. 2003;290(8):1029–30. https://pubmed.ncbi.nlm.nih.gov/12941673/
Carnevale R, Loffredo L, Pignatelli P, et al. Dark chocolate inhibits platelet isoprostanes via NOX2 down-regulation in smokers. J Thromb Haemost. 2012;10(1):125–32. https://pubmed.ncbi.nlm.nih.gov/22066819/
Parsaeyan N, Mozaffari-Khosravi H, Absalan A, Mozayan MR. Beneficial effects of cocoa on lipid peroxidation and inflammatory markers in type 2 diabetic patients and investigation of probable interactions of cocoa active ingredients with prostaglandin synthase-2 (PTGS-2/COX-2) using virtual analysis. J Diabetes Metab Disord. 2014;13(1):30. https://pubmed.ncbi.nlm.nih.gov/24495354/
Onuegbu AJ, Olisekodiaka JM, Irogue SE, et al. Consumption of soymilk reduces lipid peroxidation but may lower micronutrient status in apparently healthy individuals. J Med Food. 2018;21(5):506–10. https://pubmed.ncbi.nlm.nih.gov/29432056/
Ballard KD, Mah E, Guo Y, Pei R, Volek JS, Bruno RS. Low-fat milk ingestion prevents postprandial hyperglycemia-mediated impairments in vascular endothelial function in obese individuals with metabolic syndrome. J Nutr. 2013;143(10):1602–10. https://pubmed.ncbi.nlm.nih.gov/23966328/
Dickinson KM, Clifton PM, Keogh JB. Endothelial function is impaired after a high-salt meal in healthy subjects. Am J Clin Nutr. 2011;93(3):500–5. https://pubmed.ncbi.nlm.nih.gov/21228265/
Jablonski KL, Racine ML, Geolfos CJ, et al. Dietary sodium restriction reverses vascular endothelial dysfunction in middle-aged/older adults with moderately elevated systolic blood pressure. J Am Coll Cardiol. 2013;61(3):335–43. https://pubmed.ncbi.nlm.nih.gov/23141486/
McCord JM. Analysis of superoxide dismutase activity. Curr Protoc Toxicol. 2001;Chapter 7:Unit 7.3. https://pubmed.ncbi.nlm.nih.gov/23045062/
Chai SC, Davis K, Zhang Z, Zha L, Kirschner KF. Effects of tart cherry juice on biomarkers of inflammation and oxidative stress in older adults. Nutrients. 2019;11(2):228. https://pubmed.ncbi.nlm.nih.gov/30678193/
Dourado GKZS, Cesar TB. Investigation of cytokines, oxidative stress, metabolic, and inflammatory biomarkers after orange juice consumption by normal and overweight subjects. Food Nutr Res. 2015;59(1):28147. https://pubmed.ncbi.nlm.nih.gov/26490535/
Shema-Didi L, Sela S, Ore L, et al. One year of pomegranate juice intake decreases oxidative stress, inflammation, and incidence of infections in hemodialysis patients: a randomized placebo-controlled trial. Free Radic Biol Med. 2012;53(2):297–304. https://pubmed.ncbi.nlm.nih.gov/22609423/
Ghavipour M, Sotoudeh G, Ghorbani M. Tomato juice consumption improves blood antioxidative biomarkers in overweight and obese females. Clin Nutr. 2015;34(5):805–9. https://pubmed.ncbi.nlm.nih.gov/25466953/
Shyam R, Singh SN, Vats P, et al. Wheat grass supplementation decreases oxidative stress in healthy subjects: a comparative study with spirulina. J Altern Complement Med. 2007;13(8):789–91. https://pubmed.ncbi.nlm.nih.gov/17983333/
Basu A, Betts NM, Ortiz J, Simmons B, Wu M, Lyons TJ. Low-calorie cranberry juice decreases lipid oxidation and increases plasma antioxidant capacity in women with metabolic syndrome. Nutr Res. 2011;31(3):190–6. https://pubmed.ncbi.nlm.nih.gov/21481712/
de Lima Tavares Toscano L, Silva AS, de França ACL, et al. A single dose of purple grape juice improves physical performance and antioxidant activity in runners: a randomized, crossover, double-blind, placebo study. Eur J Nutr. 2020;59(7):2997–3007. https://pubmed.ncbi.nlm.nih.gov/31732851/
Cao G, Russell RM, Lischner N, Prior RL. Serum antioxidant capacity is increased by consumption of strawberries, spinach, red wine or vitamin C in elderly women. J Nutr. 1998;128(12):2383–90. https://pubmed.ncbi.nlm.nih.gov/9868185/
Ursini F, Zamburlini A, Cazzolato G, Maiorino M, Bon GB, Sevanian A. Postprandial plasma lipid hydroperoxides: a possible link between diet and atherosclerosis. Free Radic Biol Med. 1998;25(2):250–2. https://pubmed.ncbi.nlm.nih.gov/9667503/
Caccetta RAA, Burke V, Mori TA, Beilin LJ, Puddey IB, Croft KD. Red wine polyphenols, in the absence of alcohol, reduce lipid peroxidative stress in smoking subjects. Free Radic Biol Med. 2001;30(6):636–42. https://pubmed.ncbi.nlm.nih.gov/11295361/
Meagher EA, Barry OP, Burke A, et al. Alcohol-induced generation of lipid peroxidation products in humans. J Clin Invest. 1999;104(6):805–13. https://pubmed.ncbi.nlm.nih.gov/10491416/
Xue KX, Wang S, Ma GJ, et al. Micronucleus formation in peripheral-blood lymphocytes from smokers and the influence of alcohol– and tea-drinking habits. Int J Cancer. 1992;50(5):702–5. https://pubmed.ncbi.nlm.nih.gov/1544703/
Bloomer RJ, Trepanowski JF, Farney TM. Influence of acute coffee consumption on postprandial oxidative stress. Nutr Metab Insights. 2013;6:35–42. https://pubmed.ncbi.nlm.nih.gov/23935371/
Takahashi M, Miyashita M, Suzuki K, et al. Acute ingestion of catechin-rich green tea improves postprandial glucose status and increases serum thioredoxin concentrations in postmenopausal women. Br J Nutr. 2014;112(9):1542–50. https://pubmed.ncbi.nlm.nih.gov/25230741/
Leenen R, Roodenburg AJ, Tijburg LB, Wiseman SA. A single dose of tea with or without milk increases plasma antioxidant activity in humans. Eur J Clin Nutr. 2000;54(1):87–92. https://pubmed.ncbi.nlm.nih.gov/10694777/
Rashidinejad A, Birch EJ, Sun-Waterhouse D, Everett DW. Addition of milk to tea infusions: helpful or harmful? Evidence from in vitro and in vivo studies on antioxidant properties. Crit Rev Food Sci Nutr. 2017;57(15):3188–96. https://pubmed.ncbi.nlm.nih.gov/26517348/
Ho CK, Choi SW, Siu PM, Benzie IFF. Effects of single dose and regular intake of green tea (Camellia sinensis) on DNA damage, DNA repair, and heme oxygenase-1 expression in a randomized controlled human supplementation study. Mol Nutr Food Res. 2014;58(6):1379–83. https://pubmed.ncbi.nlm.nih.gov/24585444/
Han KC, Wong WC, Benzie IFF. Genoprotective effects of green tea (Camellia sinensis) in human subjects: results of a controlled supplementation trial. Br J Nutr. 2011;105(2):171–9. https://pubmed.ncbi.nlm.nih.gov/20807462/
Dias TR, Alves MG, Tomás GD, Socorro S, Silva BM, Oliveira PF. White tea as a promising antioxidant medium additive for sperm storage at room temperature: a comparative study with green tea. J Agric Food Chem. 2014;62(3):608–17. https://pubmed.ncbi.nlm.nih.gov/24372402/
Choi SW, Yeung VTF, Collins AR, Benzie IFF. Redox-linked effects of green tea on DNA damage and repair, and influence of microsatellite polymorphism in HMOX-1: results of a human intervention trial. Mutagenesis. 2015;30(1):129–37. https://pubmed.ncbi.nlm.nih.gov/25527735/
Leaf DA, Kleinman MT, Hamilton M, Deitrick RW. The exercise-induced oxidative stress paradox: the effects of physical exercise training. Am J Med Sci. 1999;317(5):295–300. https://pubmed.ncbi.nlm.nih.gov/10334116/
Mastaloudis A, Yu TW, O’Donnell RP, Frei B, Dashwood RH, Traber MG. Endurance exercise results in DNA damage as detected by the comet assay. Free Radic Biol Med. 2004;36(8):966–75. https://pubmed.ncbi.nlm.nih.gov/15059637/
Vollaard NBJ, Shearman JP, Cooper CE. Exercise-induced oxidative stress: myths, realities and physiological relevance. Sports Med. 2005;35(12):1045–62. https://pubmed.ncbi.nlm.nih.gov/16336008/
Mastaloudis A, Yu TW, O’Donnell RP, Frei B, Dashwood RH, Traber MG. Endurance exercise results in DNA damage as detected by the comet assay. Free Radic Biol Med. 2004;36(8):966–75. https://pubmed.ncbi.nlm.nih.gov/15059637/
Fisher-Wellman K, Bloomer RJ. Acute exercise and oxidative stress: a 30 year history. Dyn Med. 2009;8:1. https://pubmed.ncbi.nlm.nih.gov/19144121/
Ristow M, Zarse K, Oberbach A, et al. Antioxidants prevent health-promoting effects of physical exercise in humans. Proc Natl Acad Sci U S A. 2009;106(21):8665–70. https://pubmed.ncbi.nlm.nih.gov/19433800/
Braakhuis AJ. Effect of vitamin C supplements on physical performance. Curr Sports Med Rep. 2012;11(4):180–4. https://pubmed.ncbi.nlm.nih.gov/22777327/
Kashi DS, Shabir A, Da Boit M, Bailey SJ, Higgins MF. The efficacy of administering fruit-derived polyphenols to improve health biomarkers, exercise performance and related physiological responses. Nutrients. 2019;11(10):E2389. https://pubmed.ncbi.nlm.nih.gov/31591287/
Van der Avoort CMT, Van Loon LJC, Hopman MTE, Verdijk LB. Increasing vegetable intake to obtain the health promoting and ergogenic effects of dietary nitrate. Eur J Clin Nutr. 2018;72(11):1485–9. https://pubmed.ncbi.nlm.nih.gov/29559721/
Trapp D, Knez W, Sinclair W. Could a vegetarian diet reduce exercise-induced oxidative stress? A review of the literature. J Sports Sci. 2010;28(12):1261–8. https://pubmed.ncbi.nlm.nih.gov/20845212/
Lyall KA, Hurst SM, Cooney J, et al. Short-term blackcurrant extract consumption modulates exercise-induced oxidative stress and lipopolysaccharide-stimulated inflammatory responses. Am J Physiol Regul Integr Comp Physiol. 2009;297(1):R70–81. https://pubmed.ncbi.nlm.nih.gov/19403859/
Funes L, Carrera-Quintanar L, Cerdán-Calero M, et al. Effect of lemon verbena supplementation on muscular damage markers, proinflammatory cytokines release and neutrophils’ oxidative stress in chronic exercise. Eur J Appl Physiol. 2011;111(4):695–705. https://pubmed.ncbi.nlm.nih.gov/20967458/
Ghezzi P, Jaquet V, Marcucci F, Schmidt HHHW. The oxidative stress theory of disease: levels of evidence and epistemological aspects. Br J Pharmacol. 2017;174(12):1784–96. https://pubmed.ncbi.nlm.nih.gov/27425643/
Scudellari M. The science myths that will not die. Nature. 2015;528(7582):322–5. https://pubmed.ncbi.nlm.nih.gov/26672537/
Peng C, Wang X, Chen J, et al. Biology of ageing and role of dietary antioxidants. Biomed Res Int. 2014;2014:831841. https://pubmed.ncbi.nlm.nih.gov/24804252/
Milisav I, Ribaric S, Poljsak B. Antioxidant vitamins and ageing. Subcell Biochem. 2018;90:1–23. https://pubmed.ncbi.nlm.nih.gov/30779004/
Smejkal GB, Kakumanu S. Enzymes and their turnover numbers. Expert Rev Proteom. 2019;16(7):543–4. https://pubmed.ncbi.nlm.nih.gov/31220960/
Raghunath A, Sundarraj K, Nagarajan R, et al. Antioxidant response elements: discovery, classes, regulation and potential applications. Redox Biol. 2018;17:297–314. https://pubmed.ncbi.nlm.nih.gov/29775961/
Zang H, Mathew RO, Cui T. The dark side of Nrf2 in the heart. Front Physiol. 2020;11:722. https://pubmed.ncbi.nlm.nih.gov/32733266/
Brandes MS, Gray NE. NRF2 as a therapeutic target in neurodegenerative diseases. ASN Neuro. 2020;12:1759091419899782. https://pubmed.ncbi.nlm.nih.gov/31964153/
Sharma V, Kaur A, Singh TG. Counteracting role of nuclear factor erythroid 2-related factor 2 pathway in Alzheimer’s disease. Biomed Pharmacother. 2020;129:110373. https://pubmed.ncbi.nlm.nih.gov/32603894/
Yuan H, Xu Y, Luo Y, Wang NX, Xiao JH. Role of Nrf2 in cell senescence regulation. Mol Cell Biochem. 2021;476(1):247–59. https://pubmed.ncbi.nlm.nih.gov/32918185/
Raghunath A, Sundarraj K, Nagarajan R, et al. Antioxidant response elements: discovery, classes, regulation and potential applications. Redox Biol. 2018;17:297–314. https://pubmed.ncbi.nlm.nih.gov/29775961/
Raghunath A, Sundarraj K, Nagarajan R, et al. Antioxidant response elements: discovery, classes, regulation and potential applications. Redox Biol. 2018;17:297–314. https://pubmed.ncbi.nlm.nih.gov/29775961/
Ferguson LR, Schlothauer RC. The potential role of nutritional genomics tools in validating high health foods for cancer control: broccoli as example. Mol Nutr Food Res. 2012;56(1):126–46. https://pubmed.ncbi.nlm.nih.gov/22147677/
Sun Y, Yang T, Leak RK, Chen J, Zhang F. Preventive and protective roles of dietary Nrf2 activators against central nervous system diseases. CNS Neurol Disord Drug Targets. 2017;16(3):326–38. https://pubmed.ncbi.nlm.nih.gov/28042770/
Yang L, Palliyaguru DL, Kensler TW. Frugal chemoprevention: targeting Nrf2 with foods rich in sulforaphane. Semin Oncol. 2016;43(1):146–53. https://pubmed.ncbi.nlm.nih.gov/26970133/
Qu Z, Sun J, Zhang W, Yu J, Zhuang C. Transcription factor NRF2 as a promising therapeutic target for Alzheimer’s disease. Free Radic Biol Med. 2020;159:87–102. https://pubmed.ncbi.nlm.nih.gov/32730855/
Lewis KN, Mele J, Hayes JD, Buffenstein R. Nrf2, a guardian of healthspan and gatekeeper of species longevity. Integr Comp Biol. 2010;50(5):829–43. https://pubmed.ncbi.nlm.nih.gov/21031035/
Tullet JMA, Hertweck M, An JH, et al. Direct inhibition of the longevity-promoting factor SKN-1 by insulin-like signaling in C. elegans. Cell. 2008;132(6):1025–38. https://pubmed.ncbi.nlm.nih.gov/18358814/
Sykiotis GP, Bohmann D. Keap1/Nrf2 signaling regulates oxidative stress tolerance and lifespan in Drosophila. Dev Cell. 2008;14(1):76–85. https://pubmed.ncbi.nlm.nih.gov/18194654/
Lewis KN, Wason E, Edrey YH, Kristan DM, Nevo E, Buffenstein R. Regulation of Nrf2 signaling and longevity in naturally long-lived rodents. Proc Natl Acad Sci U S A. 2015;112(12):3722–7. https://pubmed.ncbi.nlm.nih.gov/25775529/
Yu C, Li Y, Holmes A, et al. RNA sequencing reveals differential expression of mitochondrial and oxidation reduction genes in the long-lived naked mole-rat when compared to mice. PLoS ONE. 2011;6(11):e26729. https://pubmed.ncbi.nlm.nih.gov/22073188/
Lewis KN, Wason E, Edrey YH, Kristan DM, Nevo E, Buffenstein R. Regulation of Nrf2 signaling and longevity in naturally long-lived rodents. Proc Natl Acad Sci U S A. 2015;112(12):3722–7. https://pubmed.ncbi.nlm.nih.gov/25775529/
Andziak B, O’Connor TP, Buffenstein R. Antioxidants do not explain the disparate longevity between mice and the longest-living rodent, the naked mole-rat. Mech Ageing Dev. 2005;126(11):1206–12. https://pubmed.ncbi.nlm.nih.gov/16087218/
Lewis KN, Wason E, Edrey YH, Kristan DM, Nevo E, Buffenstein R. Regulation of Nrf2 signaling and longevity in naturally long-lived rodents. Proc Natl Acad Sci U S A. 2015;112(12):3722–7. https://pubmed.ncbi.nlm.nih.gov/25775529/
Yuan H, Xu Y, Luo Y, Wang NX, Xiao JH. Role of Nrf2 in cell senescence regulation. Mol Cell Biochem. 2021;476(1):247–59. https://pubmed.ncbi.nlm.nih.gov/32918185/
Zhou L, Zhang H, Davies KJA, Forman HJ. Aging-related decline in the induction of Nrf2-regulated antioxidant genes in human bronchial epithelial cells. Redox Biol. 2018;14:35–40. https://pubmed.ncbi.nlm.nih.gov/28863281/
Mallard AR, Spathis JG, Coombes JS. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and exercise. Free Radic Biol Med. 2020;160:471–9. https://pubmed.ncbi.nlm.nih.gov/32871230/
Zhang DD, Chapman E. The role of natural products in revealing NRF2 function. Nat Prod Rep. 2020;37(6):797–826. https://pubmed.ncbi.nlm.nih.gov/32400766/
Su X, Jiang X, Meng L, Dong X, Shen Y, Xin Y. Anticancer activity of sulforaphane: the epigenetic mechanisms and the Nrf2 signaling pathway. Oxid Med Cell Longev. 2018;2018:5438179. https://pubmed.ncbi.nlm.nih.gov/29977456/
Bose C, Alves I, Singh P, et al. Sulforaphane prevents age-associated cardiac and muscular dysfunction through Nrf2 signaling. Aging Cell. 2020;19(11):e13261. https://pubmed.ncbi.nlm.nih.gov/33067900/
Kubo E, Chhunchha B, Singh P, Sasaki H, Singh DP. Sulforaphane reactivates cellular antioxidant defense by inducing Nrf2/ARE/Prdx6 activity during aging and oxidative stress. Sci Rep. 2017;7:14130. https://pubmed.ncbi.nlm.nih.gov/29074861/
Yuan H, Xu Y, Luo Y, Wang NX, Xiao JH. Role of Nrf2 in cell senescence regulation. Mol Cell Biochem. 2021;476(1):247–59. https://pubmed.ncbi.nlm.nih.gov/32918185/
Riso P, Martini D, Møller P, et al. DNA damage and repair activity after broccoli intake in young healthy smokers. Mutagenesis. 2010;25(6):595–602. https://pubmed.ncbi.nlm.nih.gov/20713433/
Hoelzl C, Glatt H, Meinl W, et al. Consumption of Brussels sprouts protects peripheral human lymphocytes against 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) and oxidative DNA-damage: results of a controlled human intervention trial. Mol Nutr Food Res. 2008;52(3):330–41. https://pubmed.ncbi.nlm.nih.gov/18293303/
Egner PA, Chen JG, Zarth AT, et al. Rapid and sustainable detoxication of airborne pollutants by broccoli sprout beverage: results of a randomized clinical trial in China. Cancer Prev Res. 2014;7(8):813–23. https://pubmed.ncbi.nlm.nih.gov/24913818/
Heber D, Li Z, Garcia-Lloret M, et al. Sulforaphane-rich broccoli sprout extract attenuates nasal allergic response to diesel exhaust particles. Food Funct. 2014;5(1):35–41. https://pubmed.ncbi.nlm.nih.gov/24287881/
Eagles SK, Gross AS, McLachlan AJ. The effects of cruciferous vegetable-enriched diets on drug metabolism: a systematic review and meta-analysis of dietary intervention trials in humans. Clin Pharmacol Ther. 2020;108(2):212–27. https://pubmed.ncbi.nlm.nih.gov/32086800/
Knatko EV, Ibbotson SH, Zhang Y, et al. Nrf2 activation protects against solar-simulated ultraviolet radiation in mice and humans. Cancer Prev Res (Phila). 2015;8(6):475–86. https://pubmed.ncbi.nlm.nih.gov/25804610/
Houghton CA, Fassett RG, Coombes JS. Sulforaphane and other nutrigenomic Nrf2 activators: can the clinician’s expectation be matched by the reality? Oxid Med Cell Longev. 2016;2016:7857186. https://pubmed.ncbi.nlm.nih.gov/26881038/
Aune D, Giovannucci E, Boffetta P, et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality – a systematic review and dose-response meta-analysis of prospective studies. Int J Epidemiol. 2017;46(3):1029–56. https://pubmed.ncbi.nlm.nih.gov/28338764/
Mori N, Shimazu T, Charvat H, et al. Cruciferous vegetable intake and mortality in middle-aged adults: a prospective cohort study. Clin Nutr. 2019;38(2):631–43. https://pubmed.ncbi.nlm.nih.gov/29739681/
Grünwald S, Stellzig J, Adam IV, et al. Longevity in the red flour beetle Tribolium castaneum is enhanced by broccoli and depends on nrf-2, jnk-1 and foxo-1 homologous genes. Genes Nutr. 2013;8(5):439–48. https://pubmed.ncbi.nlm.nih.gov/23321956/
Hanschen FS. Domestic boiling and salad preparation habits affect glucosinolate degradation in red cabbage (Brassica oleracea var. capitata f. rubra). Food Chem. 2020;321:126694. https://pubmed.ncbi.nlm.nih.gov/32244140/
Hernández-Ruiz Á, García-Villanova B, Guerra-Hernández E, Amiano P, Ruiz-Canela M, Molina-Montes E. A review of a priori defined oxidative balance scores relative to their components and impact on health outcomes. Nutrients. 2019;11(4):774. https://pubmed.ncbi.nlm.nih.gov/30987200/
Holland RD, Gehring T, Taylor J, Lake BG, Gooderham NJ, Turesky RJ. Formation of a mutagenic heterocyclic aromatic amine from creatinine in urine of meat eaters and vegetarians. Chem Res Toxicol. 2005;18(3):579–90. https://pubmed.ncbi.nlm.nih.gov/15777097/
Carvalho AM, Miranda AM, Santos FA, Loureiro APM, Fisberg RM, Marchioni DM. High intake of heterocyclic amines from meat is associated with oxidative stress. Br J Nutr. 2015;113(8):1301–7. https://pubmed.ncbi.nlm.nih.gov/25812604/
Macho-González A, Garcimartín A, López-Oliva ME, et al. Can meat and meat-products induce oxidative stress? Antioxidants (Basel). 2020;9(7):638. https://pubmed.ncbi.nlm.nih.gov/32698505/
Kanner J, Lapidot T. The stomach as a bioreactor: dietary lipid peroxidation in the gastric fluid and the effects of plant-derived antioxidants. Free Radic Biol Med. 2001;31(11):1388–95. https://pubmed.ncbi.nlm.nih.gov/11728810/
Mohamed B, Mohamed I. The effects of residual blood of carcasses on poultry technological quality. Food Nutri Sci. 2012;03(10):1382–6. https://www.scirp.org/journal/paperinformation.aspx?paperid=23386
Alvarado CZ, Richards MP, O’Keefe SF, Wang H. The effect of blood removal on oxidation and shelf life of broiler breast meat. Poult Sci. 2007;86(1):156–61. https://pubmed.ncbi.nlm.nih.gov/17179431/
Cohn JS. Oxidized fat in the diet, postprandial lipaemia and cardiovascular disease. Curr Opin Lipidol. 2002;13(1):19–24. https://pubmed.ncbi.nlm.nih.gov/11790959/
Gorelik S, Kanner J, Schurr D, Kohen R. A rational approach to prevent postprandial modification of LDL by dietary polyphenols. J Funct Foods. 2013;5(1):163–9. https://www.sciencedirect.com/science/article/pii/S1756464612001466?via%3Dihub
Jafari S, Hezaveh E, Jalilpiran Y, et al. Plant-based diets and risk of disease mortality: a systematic review and meta-analysis of cohort studies. Crit Rev Food Sci Nutr. https://www.tandfonline.com/doi/full/10.1080/10408398.2021.1918628. Published May 6, 2021. Accessed July 10, 2021.; https://www.tandfonline.com/doi/full/10.1080/10408398.2021.1918628
Cohn JS. Oxidized fat in the diet, postprandial lipaemia and cardiovascular disease. Curr Opin Lipidol. 2002;13(1):19–24. https://pubmed.ncbi.nlm.nih.gov/11790959/
Edalati S, Bagherzadeh F, Asghari Jafarabadi M, Ebrahimi-Mamaghani M. Higher ultra-processed food intake is associated with higher DNA damage in healthy adolescents. Br J Nutr. 2021;125(5):568–76. https://pubmed.ncbi.nlm.nih.gov/32513316/
Macho-González A, Garcimartín A, López-Oliva ME, et al. Can meat and meat-products induce oxidative stress? Antioxidants (Basel). 2020;9(7):638. https://pubmed.ncbi.nlm.nih.gov/32698505/
Aleksandrova K, Koelman L, Rodrigues CE. Dietary patterns and biomarkers of oxidative stress and inflammation: a systematic review of observational and intervention studies. Redox Biol. 2021;42:101869. https://pubmed.ncbi.nlm.nih.gov/33541846/
Benzie IFF, Wachtel-Galor S. Vegetarian diets and public health: biomarker and redox connections. Antioxid Redox Signal. 2010;13(10):1575–91. https://pubmed.ncbi.nlm.nih.gov/20222825/
Burri BJ. Antioxidant status in vegetarians versus omnivores: a mechanism for longer life? Nutrition. 2000;16(2):149–50. https://pubmed.ncbi.nlm.nih.gov/10755825/
Krajcovicová-Kudlácková M, Šimoncic R, Béderová A, Klvanová J, Brtková A, Grancicová E. Lipid and antioxidant blood levels in vegetarians. Nahrung. 1996;40(1):17–20. https://pubmed.ncbi.nlm.nih.gov/8975140/
Kováciková Z, Cerhata D, Kadrabová J, Madaric A, Ginter E. Antioxidant status in vegetarians and nonvegetarians in Bratislava region (Slovakia). Z Ernahrungswiss. 1998;37(2):178–82. https://pubmed.ncbi.nlm.nih.gov/9698645/
Nagyová A, Kudlácková M, Grancicová E, Magálová T. LDL oxidizability and antioxidative status of plasma in vegetarians. Ann Nutr Metab. 1998;42(6):328–32. https://pubmed.ncbi.nlm.nih.gov/9895420/
Boanca MM, Colosi HA, Craciun EC. The impact of the lacto-ovo vegetarian diet on the erythrocyte superoxide dismutase activity: a study in the Romanian population. Eur J Clin Nutr. 2014;68(2):184–8. https://pubmed.ncbi.nlm.nih.gov/24105324/
Krajcovicová-Kudlácková M, Valachovicová M, Pauková V, Dušinská M. Effects of diet and age on oxidative damage products in healthy subjects. Physiol Res. 2008;57(4):647–51. https://pubmed.ncbi.nlm.nih.gov/17705666/
Somannavar MS, Kodliwadmath MV. Correlation between oxidative stress and antioxidant defence in South Indian urban vegetarians and non-vegetarians. Eur Rev Med Pharmacol Sci. 2012;16(3):351–4. https://pubmed.ncbi.nlm.nih.gov/22530352/
Manjari V, Suresh Y, Sailaja Devi MM, Das UN. Oxidant stress, anti-oxidants and essential fatty acids in South Indian vegetarians and non-vegetarians. Prostaglandins Leukot Essent Fatty Acids. 2001;64(1):53–9. https://pubmed.ncbi.nlm.nih.gov/11161585/
Kim MK, Cho SW, Park YK. Long-term vegetarians have low oxidative stress, body fat, and cholesterol levels. Nutr Res Pract. 2012;6(2):155–61. https://pubmed.ncbi.nlm.nih.gov/22586505/
Szeto YT, Kwok TCY, Benzie IFF. Effects of a long-term vegetarian diet on biomarkers of antioxidant status and cardiovascular disease risk. Nutrition. 2004;20(10):863–6. https://pubmed.ncbi.nlm.nih.gov/15474873/
Gajski G, Geric M, Vucic Lovrencic M, et al. Analysis of health-related biomarkers between vegetarians and non-vegetarians: a multi-biomarker approach. J Funct Foods. 2018;48:643–53. https://www.sciencedirect.com/science/article/abs/pii/S1756464618304109?via%3Dihub
Poornima K, Cariappa M, Asha K, Kedilaya HP, Nandini M. Oxidant and antioxidant status in vegetarians and fish eaters. Indian J Clin Biochem. 2003;18(2):197–205. https://pubmed.ncbi.nlm.nih.gov/23105412/
Krajcovicová-Kudlácková M, Šimoncic R, Babinská K, Béderová A. Levels of lipid peroxidation and antioxidants in vegetarians. Eur J Epidemiol. 1995;11(2):207–11. https://pubmed.ncbi.nlm.nih.gov/7672077/
Nadimi H, Yousefinejad A, Djazayery A, Hosseini M, Hosseini S. Association of vegan diet with RMR, body composition and oxidative stress. Acta Sci Pol Technol Aliment. 2013;12(3):311–8. https://pubmed.ncbi.nlm.nih.gov/24584960/
Herrmann W, Schorr H, Purschwitz K, Rassoul F, Richter V. Total homocysteine, vitamin B12, and total antioxidant status in vegetarians. Clin Chem. 2001;47(6):1094–101. https://pubmed.ncbi.nlm.nih.gov/11375297/
van de Lagemaat EE, de Groot LCPGM, van den Heuvel EGHM. Vitamin B12 in relation to oxidative stress: a systematic review. Nutrients. 2019;11(2):E482. https://pubmed.ncbi.nlm.nih.gov/30823595/
Pawlak R, Lester SE, Babatunde T. The prevalence of cobalamin deficiency among vegetarians assessed by serum vitamin B12: a review of literature. Eur J Clin Nutr. 2014;68(5):541–8. https://pubmed.ncbi.nlm.nih.gov/24667752/
Poli G, Biasi F, Leonarduzzi G. Oxysterols in the pathogenesis of major chronic diseases. Redox Biol. 2013;1:125–30. https://pubmed.ncbi.nlm.nih.gov/24024145/
Wellington CL, Frikke-Schmidt R. Relation between plasma and brain lipids. Curr Opin Lipidol. 2016;27(3):225–32. https://pubmed.ncbi.nlm.nih.gov/27149391/
Poli G, Biasi F, Leonarduzzi G. Oxysterols in the pathogenesis of major chronic diseases. Redox Biol. 2013;1:125–30. https://pubmed.ncbi.nlm.nih.gov/24024145/
Gamba P, Testa G, Gargiulo S, Staurenghi E, Poli G, Leonarduzzi G. Oxidized cholesterol as the driving force behind the development of Alzheimer’s disease. Front Aging Neurosci. 2015;7. https://pubmed.ncbi.nlm.nih.gov/26150787/
Otaegui-Arrazola A, Menéndez-Carreño M, Ansorena D, Astiasarán I. Oxysterols: a world to explore. Food Chem Toxicol. 2010;48(12):3289–303. https://pubmed.ncbi.nlm.nih.gov/20870006/
Iuliano L, Micheletta F, Natoli S, et al. Measurement of oxysterols and a-tocopherol in plasma and tissue samples as indices of oxidant stress status. Anal Biochem. 2003;312(2):217–23. https://pubmed.ncbi.nlm.nih.gov/12531208/
Zarrouk A, Vejux A, Mackrill J, et al. Involvement of oxysterols in age-related diseases and ageing processes. Ageing Res Rev. 2014;18:148–62. https://pubmed.ncbi.nlm.nih.gov/25305550/
Otaegui-Arrazola A, Menéndez-Carreño M, Ansorena D, Astiasarán I. Oxysterols: a world to explore. Food Chem Toxicol. 2010;48(12):3289–303. https://pubmed.ncbi.nlm.nih.gov/20870006/
Zarrouk A, Vejux A, Mackrill J, et al. Involvement of oxysterols in age-related diseases and ageing processes. Ageing Res Rev. 2014;18:148–62. https://pubmed.ncbi.nlm.nih.gov/25305550/
Lordan S, Mackrill JJ, O’Brien NM. Oxysterols and mechanisms of apoptotic signaling: implications in the pathology of degenerative diseases. J Nutr Biochem. 2009;20(5):321–36. https://pubmed.ncbi.nlm.nih.gov/19345313/
Si R, Qu K, Jiang Z, Yang X, Gao P. Egg consumption and breast cancer risk: a meta-analysis. Breast Cancer. 2014;21(3):251–61. https://pubmed.ncbi.nlm.nih.gov/24504557/
Li C, Yang L, Zhang D, Jiang W. Systematic review and meta-analysis suggest that dietary cholesterol intake increases risk of breast cancer. Nutr Res. 2016;36(7):627–35. https://pubmed.ncbi.nlm.nih.gov/27333953/
Asghari A, Umetani M. Obesity and cancer: 27-hydroxycholesterol, the missing link. Int J Mol Sci. 2020;21(14):4822. https://pubmed.ncbi.nlm.nih.gov/32650428/
Nelson ER, Chang C, McDonnell DP. Cholesterol and breast cancer pathophysiology. Trends Endocrinol & Metab. 2014;25(12):649–55. https://pubmed.ncbi.nlm.nih.gov/25458418/
Kaiser J. Cholesterol forges link between obesity and breast cancer. Science. 2013;342(6162):1028. https://pubmed.ncbi.nlm.nih.gov/24288308/
Staprans I, Pan XM, Rapp JH, Feingold KR. Oxidized cholesterol in the diet is a source of oxidized lipoproteins in human serum. J Lipid Res. 2003;44(4):705–15. https://pubmed.ncbi.nlm.nih.gov/12562864/
Emanuel HA, Hassel CA, Addis PB, Bergmann SD, Zavoral JH. Plasma cholesterol oxidation products (oxysterols) in human subjects fed a meal rich in oxysterols. J Food Sci. 1991;56(3):843–7. https://ift.onlinelibrary.wiley.com/doi/10.1111/j.1365–2621.1991.tb05396.x
Natella F, Macone A, Ramberti A, et al. Red wine prevents the postprandial increase in plasma cholesterol oxidation products: a pilot study. Br J Nutr. 2011;105(12):1718–23. https://pubmed.ncbi.nlm.nih.gov/21294933/
Lordan S, Mackrill JJ, O’Brien NM. Oxysterols and mechanisms of apoptotic signaling: implications in the pathology of degenerative diseases. J Nutr Biochem. 2009;20(5):321–36. https://pubmed.ncbi.nlm.nih.gov/19345313/
Emanuel HA, Hassel CA, Addis PB, Bergmann SD, Zavoral JH. Plasma cholesterol oxidation products (oxysterols) in human subjects fed a meal rich in oxysterols. J Food Sci. 1991;56(3):843–7. https://ift.onlinelibrary.wiley.com/doi/10.1111/j.1365–2621.1991.tb05396.x
Khan MI, Min JS, Lee SO, et al. Cooking, storage, and reheating effect on the formation of cholesterol oxidation products in processed meat products. Lipids Health Dis. 2015;14:89. https://pubmed.ncbi.nlm.nih.gov/26260472/
Min JS, Lee SO, Khan MI, et al. Monitoring the formation of cholesterol oxidation products in model systems using response surface methodology. Lipids Health Dis. 2015;14:77. https://pubmed.ncbi.nlm.nih.gov/26201850/
Hur SJ, Park GB, Joo ST. Formation of cholesterol oxidation products (COPs) in animal products. Food Control. 2007;18(8):939–47. https://www.researchgate.net/publication/248511669_Formation_of_cholesterol_oxidation_products_COPS_in_animal_products
Echarte M, Ansorena D, Astiasarán I. Consequences of microwave heating and frying on the lipid fraction of chicken and beef patties. J Agric Food Chem. 2003;51(20):5941–5. https://pubmed.ncbi.nlm.nih.gov/13129298/
Hur SJ, Park GB, Joo ST. Formation of cholesterol oxidation products (COPs) in animal products. Food Control. 2007;18(8):939–47. https://www.researchgate.net/publication/248511669_Formation_of_cholesterol_oxidation_products_COPS_in_animal_products
Maldonado-Pereira L, Schweiss M, Barnaba C, Medina-Meza IG. The role of cholesterol oxidation products in food toxicity. Food Chem Toxicol. 2018;118:908–39. https://pubmed.ncbi.nlm.nih.gov/29940280/
Savage GP, Dutta PC, Rodriguez-Estrada MT. Cholesterol oxides: their occurrence and methods to prevent their generation in foods. Asia Pac J Clin Nutr. 2002;11(1):72–8. https://pubmed.ncbi.nlm.nih.gov/11890642/
Savage GP, Dutta PC, Rodriguez-Estrada MT. Cholesterol oxides: their occurrence and methods to prevent their generation in foods. Asia Pac J Clin Nutr. 2002;11(1):72–8. https://pubmed.ncbi.nlm.nih.gov/11890642/
Otaegui-Arrazola A, Menéndez-Carreño M, Ansorena D, Astiasarán I. Oxysterols: a world to explore. Food Chem Toxicol. 2010;48(12):3289–303. https://pubmed.ncbi.nlm.nih.gov/20870006/
Savage GP, Dutta PC, Rodriguez-Estrada MT. Cholesterol oxides: their occurrence and methods to prevent their generation in foods. Asia Pac J Clin Nutr. 2002;11(1):72–8. https://pubmed.ncbi.nlm.nih.gov/11890642/
Jacobson MS. Cholesterol oxides in Indian ghee: possible cause of unexplained high risk of atherosclerosis in Indian immigrant populations. Lancet. 1987;2(8560):656–8. https://pubmed.ncbi.nlm.nih.gov/2887943/
Raheja BS. Ghee, cholesterol, and heart disease. Lancet. 1987;2(8568):1144–5. https://pubmed.ncbi.nlm.nih.gov/2890036/
Connor JM. Global Price Fixing. 2nd ed. Springer-Verlag; 2008. https://worldcat.org/title/238586901
Bjelakovic G, Nikolova D, Gluud C. Antioxidant supplements to prevent mortality. JAMA. 2013;310(11):1178–9. https://pubmed.ncbi.nlm.nih.gov/24045742/
Sadowska-Bartosz I, Bartosz G. Effect of antioxidants supplementation on aging and longevity. Biomed Res Int. 2014;2014:404680. https://pubmed.ncbi.nlm.nih.gov/24783202/
Bast A, Haenen GRMM. Ten misconceptions about antioxidants. Trends Pharmacol Sci. 2013;34(8):430–6. https://pubmed.ncbi.nlm.nih.gov/23806765/
Vajdi M, Abbasalizad Farhangi M. Alpha-lipoic acid supplementation significantly reduces the risk of obesity in an updated systematic review and dose response meta-analysis of randomised placebo-controlled clinical trials. Int J Clin Pract. 2020;74(6):e13493. https://pubmed.ncbi.nlm.nih.gov/32091656/
de Barcelos IP, Haas RH. CoQ10 and aging. Biology (Basel). 2019;8(2):28. https://pubmed.ncbi.nlm.nih.gov/31083534/
Raizner AE, Quiñones MA. Coenzyme Q10 for patients with cardiovascular disease: JAAC Focus Seminar. J Am Coll Cardiol. 2021;77(5):609–19. https://pubmed.ncbi.nlm.nih.gov/33538259/
Arenas-Jal M, Suñé-Negre JM, García-Montoya E. Coenzyme Q10 supplementation: efficacy, safety, and formulation challenges. Compr Rev Food Sci Food Saf. 2020;19(2):574–94. https://pubmed.ncbi.nlm.nih.gov/33325173/
Nagase M, Yamamoto Y, Matsumoto N, Arai Y, Hirose N. Increased oxidative stress and coenzyme Q10 deficiency in centenarians. J Clin Biochem Nutr. 2018;63(2):129–36. https://pubmed.ncbi.nlm.nih.gov/30279624/
Varela-López A, Giampieri F, Battino M, Quiles JL. Coenzyme Q and its role in the dietary therapy against aging. Molecules. 2016;21(3):373. https://pubmed.ncbi.nlm.nih.gov/26999099/
Asencio C, Rodríguez-Aguilera JC, Ruiz-Ferrer M, Vela J, Navas P. Silencing of ubiquinone biosynthesis genes extends life span in Caenorhabditis elegans. FASEB J. 2003;17(9):1135–7. https://pubmed.ncbi.nlm.nih.gov/12709403/
Díaz-Casado ME, Quiles JL, Barriocanal-Casado E, et al. The paradox of coenzyme Q10 in aging. Nutrients. 2019;11(9):E2221. https://pubmed.ncbi.nlm.nih.gov/31540029/
Fan L, Feng Y, Chen GC, Qin LQ, Fu CL, Chen LH. Effects of coenzyme Q10 supplementation on inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res. 2017;119:128–36. https://pubmed.ncbi.nlm.nih.gov/28179205/
Akbari A, Mobini GR, Agah S, et al. Coenzyme Q10 supplementation and oxidative stress parameters: a systematic review and meta-analysis of clinical trials. Eur J Clin Pharmacol. 2020;76(11):1483–99. https://pubmed.ncbi.nlm.nih.gov/32583356/
Jafari M, Mousavi SM, Asgharzadeh A, Yazdani N. Coenzyme Q10 in the treatment of heart failure: a systematic review of systematic reviews. Indian Heart J. 2018;70(Suppl 1):S111–7. https://pubmed.ncbi.nlm.nih.gov/30122240/
Sazali S, Badrin S, Norhayati MN, Idris NS. Coenzyme Q10 supplementation for prophylaxis in adult patients with migraine – a meta-analysis. BMJ Open. 2021;11(1):e039358. https://pubmed.ncbi.nlm.nih.gov/33402403/
Arenas-Jal M, Suñé-Negre JM, García-Montoya E. Coenzyme Q10 supplementation: efficacy, safety, and formulation challenges. Compr Rev Food Sci Food Saf. 2020;19(2):574–94. https://pubmed.ncbi.nlm.nih.gov/33325173/
Qu J, Ma L, Zhang J, Jockusch S, Washington I. Dietary chlorophyll metabolites catalyze the photoreduction of plasma ubiquinone. Photochem Photobiol. 2013;89(2):310–3. https://pubmed.ncbi.nlm.nih.gov/22928808/
Littarru GP, Langsjoen P. Coenzyme Q10 and statins: biochemical and clinical implications. Mitochondrion. 2007;7S:S168–74. https://pubmed.ncbi.nlm.nih.gov/17482884/
Lee TK, Johnke RM, Allison RR, O’Brien KF, Dobbs LJ. Radioprotective potential of ginseng. Mutagenesis. 2005;20(4):237–43. https://pubmed.ncbi.nlm.nih.gov/15956041/
Fan S, Zhang Z, Su H, et al. Panax ginseng clinical trials: current status and future perspectives. Biomed Pharmacother. 2020;132:110832. https://pubmed.ncbi.nlm.nih.gov/33059260/
Shergis JL, Zhang AL, Zhou W, Xue CC. Panax ginseng in randomised controlled trials: a systematic review. Phytother Res. 2013;27(7):949–65. https://pubmed.ncbi.nlm.nih.gov/22969004/
Gui QF, Xu ZR, Xu KY, Yang YM. The efficacy of ginseng-related therapies in type 2 diabetes mellitus: an updated systematic review and meta-analysis. Medicine. 2016;95(6):e2584. https://pubmed.ncbi.nlm.nih.gov/26871778/
Szeto YT, Sin YSP, Pak SC, Kalle W. American ginseng tea protects cellular DNA within 2¿h from consumption: results of a pilot study in healthy human volunteers. Int J Food Sci Nutr. 2015;66(7):815–8. https://pubmed.ncbi.nlm.nih.gov/26393910/
Szeto YT, Lee LKY. Rapid but mild genoprotective effect on lymphocytic DNA with Panax notoginseng extract supplementation. J Intercult Ethnopharmacol. 2014;3(4):155–8. https://pubmed.ncbi.nlm.nih.gov/26401366/
Szeto YT, Ko AW. Acute genoprotective effects on lymphocytic DNA with ginseng extract supplementation. J Aging Res Clin Practice. 2013;2(2):174–7. https://www.researchgate.net/publication/244990213_Acute_genoprotective_effects_on_lymphocytic_DNA_with_ginseng_extract_supplementation
Kim HG, Yoo SR, Park HJ, et al. Antioxidant effects of Panax ginseng C.A. Meyer in healthy subjects: a randomized, placebo-controlled clinical trial. Food Chem Toxicol. 2011;49(9):2229–35. https://pubmed.ncbi.nlm.nih.gov/21699953/
Dickman JR, Koenig RT, Ji LL. American ginseng supplementation induces an oxidative stress in postmenopausal women. J Am Coll Nutr. 2009;28(2):219–28. https://pubmed.ncbi.nlm.nih.gov/19828907/
Flurkey K, Astle CM, Harrison DE. Life extension by diet restriction and N-acetyl-L-cysteine in genetically heterogeneous mice. J Gerontol A Biol Sci Med Sci. 2010;65(12):1275–84. https://pubmed.ncbi.nlm.nih.gov/20819793/
Oh SI, Park JK, Park SK. Lifespan extension and increased resistance to environmental stressors by N-Acetyl-L–Cysteine in Caenorhabditis elegans. Clinics. 2015;70(5):380–6. https://pubmed.ncbi.nlm.nih.gov/26039957/
Niraula P, Kim MS. N-Acetylcysteine extends lifespan of Drosophila via modulating ROS scavenger gene expression. Biogerontology. 2019;20(4):533–43. https://pubmed.ncbi.nlm.nih.gov/31115735/
Zoidis E, Seremelis I, Kontopoulos N, Danezis GP. Selenium-dependent antioxidant enzymes: actions and properties of selenoproteins. Antioxidants (Basel). 2018;7(5):66. https://pubmed.ncbi.nlm.nih.gov/29758013/
Schiavon M, Nardi S, dalla Vecchia F, Ertani A. Selenium biofortification in the 21st century: status and challenges for healthy human nutrition. Plant Soil. 2020;453(1–2):245–70. https://pubmed.ncbi.nlm.nih.gov/32836404/
Duarte GBS, Reis BZ, Rogero MM, et al. Consumption of Brazil nuts with high selenium levels increased inflammation biomarkers in obese women: a randomized controlled trial. Nutrition. 2019;63–64:162–8. https://pubmed.ncbi.nlm.nih.gov/31026738/
Xiang S, Dai Z, Man C, Fan Y. Circulating selenium and cardiovascular or all-cause mortality in the general population: a meta-analysis. Biol Trace Elem Res. 2020;195(1):55–62. https://pubmed.ncbi.nlm.nih.gov/31368032/
Bleys J, Navas-Acien A, Guallar E. Serum selenium levels and all-cause, cancer, and cardiovascular mortality among US adults. Arch Intern Med. 2008;168(4):404–10. https://pubmed.ncbi.nlm.nih.gov/18299496/
Rayman MP, Winther KH, Pastor-Barriuso R, et al. Effect of long-term selenium supplementation on mortality: results from a multiple-dose, randomised controlled trial. Free Radic Biol Med. 2018;127:46–54. https://pubmed.ncbi.nlm.nih.gov/29454039/
Faghihi T, Radfar M, Barmal M, et al. A randomized, placebo-controlled trial of selenium supplementation in patients with type 2 diabetes: effects on glucose homeostasis, oxidative stress, and lipid profile. Am J Ther. 2014;21(6):491–5. https://pubmed.ncbi.nlm.nih.gov/23633679/
Stranges S, Marshall JR, Natarajan R, et al. Effects of long-term selenium supplementation on the incidence of type 2 diabetes: a randomized trial. Ann Intern Med. 2007;147(4):217–23. https://pubmed.ncbi.nlm.nih.gov/17620655/
Talaulikar VS, Manyonda IT. Vitamin C as an antioxidant supplement in women’s health: a myth in need of urgent burial. Eur J Obstet Gynecol Reprod Biol. 2011;157(1):10–3. https://pubmed.ncbi.nlm.nih.gov/21507551/
Camarena V, Wang G. The epigenetic role of vitamin C in health and disease. Cell Mol Life Sci. 2016;73(8):1645–58. https://pubmed.ncbi.nlm.nih.gov/26846695/
Schaus R. The ascorbic acid content of human pituitary, cerebral cortex, heart, and skeletal muscle and its relation to age. Am J Clin Nutr. 1957;5(1):39–41. https://pubmed.ncbi.nlm.nih.gov/13394538/
Granger M, Eck P. Dietary vitamin C in human health. Adv Food Nutr Res. 2018;83:281–310. https://pubmed.ncbi.nlm.nih.gov/29477224/
Duarte TL, Lunec J. Review: When is an antioxidant not an antioxidant? A review of novel actions and reactions of vitamin C. Free Radic Res. 2005;39(7):671–86. https://pubmed.ncbi.nlm.nih.gov/16036346/
Childs A, Jacobs C, Kaminski T, Halliwell B, Leeuwenburgh C. Supplementation with vitamin C and N-acetyl-cysteine increases oxidative stress in humans after an acute muscle injury induced by eccentric exercise. Free Radic Biol Med. 2001;31(6):745–53. https://pubmed.ncbi.nlm.nih.gov/11557312/
Mendes-da-Silva RF, Lopes-de-Morais AAC, Bandim-da-Silva ME, et al. Prooxidant versus antioxidant brain action of ascorbic acid in well-nourished and malnourished rats as a function of dose: a cortical spreading depression and malondialdehyde analysis. Neuropharmacology. 2014;86:155–60. https://pubmed.ncbi.nlm.nih.gov/25008558/
Pallauf K, Bendall JK, Scheiermann C, et al. Vitamin C and lifespan in model organisms. Food Chem Toxicol. 2013;58:255–63. https://pubmed.ncbi.nlm.nih.gov/23643700/
Brauchla M, Dekker MJ, Rehm CD. Trends in vitamin C consumption in the United States: 1999–2018. Nutrients. 2021;13(2):420. https://pubmed.ncbi.nlm.nih.gov/33525516/
Thomas LDK, Elinder CG, Tiselius HG, Wolk A, Åkesson A. Ascorbic acid supplements and kidney stone incidence among men: a prospective study. JAMA Intern Med. 2013;173(5):386–8. https://pubmed.ncbi.nlm.nih.gov/23381591/
Fletcher RH. The risk of taking ascorbic acid. JAMA Intern Med. 2013;173(5):388. https://pubmed.ncbi.nlm.nih.gov/23381657/
Cavuoto P, Fenech MF. A review of methionine dependency and the role of methionine restriction in cancer growth control and life-span extension. Cancer Treat Rev. 2012;38(6):726–36. https://pubmed.ncbi.nlm.nih.gov/22342103/
Toledo C, Saltsman K. Genetics by the numbers. Inside Life Science. National Institute of General Medical Sciences. https://www.nigms.nih.gov/education/Inside-Life-Science/Pages/genetics-by-the-numbers.aspx. Published June 12, 2012. Accessed June 28, 2021.; https://nigms.nih.gov/education/Inside-Life-Science/Pages/Genetics-by-the-Numbers.aspx
Zhang F, Wang S, Gan L, et al. Protective effects and mechanisms of sirtuins in the nervous system. Prog Neurobiol. 2011;95(3):373–95. https://pubmed.ncbi.nlm.nih.gov/21930182/
Zhao L, Cao J, Hu K, et al. Sirtuins and their biological relevance in aging and age-related diseases. Aging Dis. 2020;11(4):927–45. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7390530/
Grabowska W, Sikora E, Bielak-Zmijewska A. Sirtuins, a promising target in slowing down the ageing process. Biogerontology. 2017;18(4):447–76. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5514220/
Kaeberlein M, McVey M, Guarente L. The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes Dev. 1999;13(19):2570–80. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC317077/
Zhao L, Cao J, Hu K, et al. Sirtuins and their biological relevance in aging and age-related diseases. Aging Dis. 2020;11(4):927–45. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7390530/
Satoh A, Brace CS, Rensing N, et al. Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 2013;18(3):416–30. https://pubmed.ncbi.nlm.nih.gov/24011076/
Kanfi Y, Naiman S, Amir G, et al. The sirtuin SIRT6 regulates lifespan in male mice. Nature. 2012;483(7388):218–21. https://pubmed.ncbi.nlm.nih.gov/22367546/
Brenner C. Sirtuins are not conserved longevity genes. Life Metabolism. Published online September 22, 2022. https://academic.oup.com/lifemeta/advance-article/doi/10.1093/lifemeta/loac025/6711379. Accessed December 27, 2022.; https://academic.oup.com/lifemeta/article/1/2/122/6711379
Giblin W, Skinner ME, Lombard DB. Sirtuins: guardians of mammalian healthspan. Trends Genet. 2014;30(7):271–86. https://pubmed.ncbi.nlm.nih.gov/24877878/
Wang RH, Sengupta K, Li C, et al. Impaired DNA damage response, genome instability, and tumorigenesis in SIRT1 mutant mice. Cancer Cell. 2008;14(4):312–23. https://pubmed.ncbi.nlm.nih.gov/18835033/
Lee SH, Lee JH, Lee HY, Min KJ. Sirtuin signaling in cellular senescence and aging. BMB Rep. 2019;52(1):24–34. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6386230/
Watroba M, Szukiewicz D. The role of sirtuins in aging and age-related diseases. Adv Med Sci. 2016;61(1):52–62. https://pubmed.ncbi.nlm.nih.gov/26521204/
Palacios JA, Herranz D, De Bonis ML, Velasco S, Serrano M, Blasco MA. SIRT1 contributes to telomere maintenance and augments global homologous recombination. J Cell Biol. 2010;191(7):1299–313. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3010065/
Morris BJ. Seven sirtuins for seven deadly diseases of aging. Free Radic Biol Med. 2013;56:133–71. https://pubmed.ncbi.nlm.nih.gov/23104101/
Giblin W, Skinner ME, Lombard DB. Sirtuins: guardians of mammalian healthspan. Trends Genet. 2014;30(7):271–86. https://pubmed.ncbi.nlm.nih.gov/24877878/
Flachsbart F, Croucher PJP, Nikolaus S, et al. Sirtuin 1 (SIRT1) sequence variation is not associated with exceptional human longevity. Exp Gerontol. 2006;41(1):98–102. https://pubmed.ncbi.nlm.nih.gov/16257164/
Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cell Biol. 2012;13(4):225–38. https://pubmed.ncbi.nlm.nih.gov/22395773/
Cantó C, Gerhart-Hines Z, Feige JN, et al. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature. 2009;458(7241):1056–60. https://pubmed.ncbi.nlm.nih.gov/19262508/
Xu W, Deng YY, Yang L, et al. Metformin ameliorates the proinflammatory state in patients with carotid artery atherosclerosis through sirtuin 1 induction. Transl Res. 2015;166(5):451–8. https://pubmed.ncbi.nlm.nih.gov/26141671/
Dang W. The controversial world of sirtuins. Drug Discov Today Technol. 2014;12:e9–17. https://pubmed.ncbi.nlm.nih.gov/25027380/
Guerra B, Guadalupe-Grau A, Fuentes T, et al. SIRT1, AMP-activated protein kinase phosphorylation and downstream kinases in response to a single bout of sprint exercise: influence of glucose ingestion. Eur J Appl Physiol. 2010;109(4):731–43. https://pubmed.ncbi.nlm.nih.gov/20217115/
Guerra B, Guadalupe-Grau A, Fuentes T, et al. SIRT1, AMP-activated protein kinase phosphorylation and downstream kinases in response to a single bout of sprint exercise: influence of glucose ingestion. Eur J Appl Physiol. 2010;109(4):731–43. https://pubmed.ncbi.nlm.nih.gov/20217115/
Asghari S, Asghari-Jafarabadi M, Somi MH, Ghavami SM, Rafraf M. Comparison of calorie-restricted diet and resveratrol supplementation on anthropometric indices, metabolic parameters, and serum sirtuin-1 levels in patients with nonalcoholic fatty liver disease: a randomized controlled clinical trial. J Am Coll Nutr. 2018;37(3):223–33. https://pubmed.ncbi.nlm.nih.gov/29313746/
Crujeiras AB, Parra D, Goyenechea E, Martínez JA. Sirtuin gene expression in human mononuclear cells is modulated by caloric restriction. Eur J Clin Invest. 2008;38(9):672–8. https://pubmed.ncbi.nlm.nih.gov/18837744/
Draznin B, Wang C, Adochio R, Leitner JW, Cornier MA. Effect of dietary macronutrient composition on AMPK and SIRT1 expression and activity in human skeletal muscle. Horm Metab Res. 2012;44(9):650–5. https://pubmed.ncbi.nlm.nih.gov/22674476/
Lilja S, Stoll C, Krammer U, et al. Five days periodic fasting elevates levels of longevity related Christensenella and sirtuin expression in humans. Int J Mol Sci. 2021;22(5):2331. https://pubmed.ncbi.nlm.nih.gov/33652686/
Heilbronn LK, Civitarese AE, Bogacka I, Smith SR, Hulver M, Ravussin E. Glucose tolerance and skeletal muscle gene expression in response to alternate day fasting. Obes Res. 2005;13(3):574–81. https://pubmed.ncbi.nlm.nih.gov/15833943/
Mansur AP, Roggerio A, Goes MFS, et al. Serum concentrations and gene expression of sirtuin 1 in healthy and slightly overweight subjects after caloric restriction or resveratrol supplementation: a randomized trial. Int J Cardiol. 2017;227:788–94. https://pubmed.ncbi.nlm.nih.gov/28029409/
Civitarese AE, Carling S, Heilbronn LK, et al. Calorie restriction increases muscle mitochondrial biogenesis in healthy humans. PLoS Med. 2007;4(3):e76. https://pubmed.ncbi.nlm.nih.gov/17341128/
Cantó C, Gerhart-Hines Z, Feige JN, et al. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature. 2009;458(7241):1056–60. https://pubmed.ncbi.nlm.nih.gov/19262508/
Giblin W, Skinner ME, Lombard DB. Sirtuins: guardians of mammalian healthspan. Trends Genet. 2014;30(7):271–86. https://pubmed.ncbi.nlm.nih.gov/24877878/
Watroba M, Szukiewicz D. The role of sirtuins in aging and age-related diseases. Adv Med Sci. 2016;61(1):52–62. https://pubmed.ncbi.nlm.nih.gov/26521204/
Giblin W, Skinner ME, Lombard DB. Sirtuins: guardians of mammalian healthspan. Trends Genet. 2014;30(7):271–86. https://pubmed.ncbi.nlm.nih.gov/24877878/
Smoliga JM, Blanchard O. Enhancing the delivery of resveratrol in humans: if low bioavailability is the problem, what is the solution? Molecules. 2014;19(11):17154–72. https://pubmed.ncbi.nlm.nih.gov/25347459/
Pezzuto JM. Resveratrol: twenty years of growth, development and controversy. Biomol Ther (Seoul). 2019;27(1):1–14. https://pubmed.ncbi.nlm.nih.gov/30332889/
Singh CK, Liu X, Ahmad N. Resveratrol, in its natural combination in whole grape, for health promotion and disease management. Ann N Y Acad Sci. 2015;1348(1):150–60. https://pubmed.ncbi.nlm.nih.gov/26099945/
Сравнительно низкий уровень сердечно-сосудистых и онкологических заболеваний у жителей Франции при высококалорийном рационе питания и обилии в нем жиров. – Примеч. ред.
Visioli F, Panaite SA, Tomé-Carneiro J. Wine’s phenolic compounds and health: a Pythagorean view. Molecules. 2020;25(18):4105. https://pubmed.ncbi.nlm.nih.gov/32911765/
Burr ML. Explaining the French paradox. J R Soc Health. 1995;115(4):217–9. https://pubmed.ncbi.nlm.nih.gov/7562866/
Vang O. What is new for resveratrol? Is a new set of recommendations necessary? Ann N Y Acad Sci. 2013;1290:1–11. https://pubmed.ncbi.nlm.nih.gov/23855460/
Resveratrol. National Library of Medicine. https://pubmed.ncbi.nlm.nih.gov/?term=resveratrol. Accessed January 18, 2023.; https://pubmed.ncbi.nlm.nih.gov/?term=resveratrol
Hector KL, Lagisz M, Nakagawa S. The effect of resveratrol on longevity across species: a meta-analysis. Biol Lett. 2012;8(5):790–3. https://pubmed.ncbi.nlm.nih.gov/22718956/
Rascón B, Hubbard BP, Sinclair DA, Amdam GV. The lifespan extension effects of resveratrol are conserved in the honey bee and may be driven by a mechanism related to caloric restriction. Aging (Albany NY). 2012;4(7):499–508. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3433935/
Hector KL, Lagisz M, Nakagawa S. The effect of resveratrol on longevity across species: a meta-analysis. Biol Lett. 2012;8(5):790–3. https://pubmed.ncbi.nlm.nih.gov/22718956/
Kim E, Ansell CM, Dudycha JL. Resveratrol and food effects on lifespan and reproduction in the model crustacean Daphnia. J Exp Zool A Ecol Genet Physiol. 2014;321(1):48–56. https://pubmed.ncbi.nlm.nih.gov/24133070/
Hector KL, Lagisz M, Nakagawa S. The effect of resveratrol on longevity across species: a meta-analysis. Biol Lett. 2012;8(5):790–3. https://pubmed.ncbi.nlm.nih.gov/22718956/
Pacholec M, Bleasdale JE, Chrunyk B, et al. SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. J Biol Chem. 2010;285(11):8340–51. https://pubmed.ncbi.nlm.nih.gov/20061378/
Cottart CH, Nivet-Antoine V, Beaudeux JL. Is resveratrol an imposter? Mol Nutr Food Res. 2015;59(1):7. https://pubmed.ncbi.nlm.nih.gov/25558005/
Tang PCT, Ng YF, Ho S, Gyda M, Chan SW. Resveratrol and cardiovascular health – promising therapeutic or hopeless illusion? Pharmacol Res. 2014;90:88–115. https://pubmed.ncbi.nlm.nih.gov/25151891/
Артефакт эксперимента (от лат. arte – «искусственно» + factus – «сделанный») – эффект в эксперименте, возникающий вследствие дефектов методики проведения опыта. – Примеч. ред.
Visioli F. The resveratrol fiasco. Pharmacol Res. 2014;90:87. https://pubmed.ncbi.nlm.nih.gov/25180457/
Roehr B. Cardiovascular researcher fabricated data in studies of red wine. BMJ. 2012;344:e406. https://pubmed.ncbi.nlm.nih.gov/22250221/
Visioli F. The resveratrol fiasco. Pharmacol Res. 2014;90:87. https://pubmed.ncbi.nlm.nih.gov/25180457/
Resveratrol clinical trial, humans from 2014/12/1–3000/12/12. National Library of Medicine. https://pubmed.ncbi.nlm.nih.gov/?term=resveratrol&filter=pubt.clinicaltrial&filter=dates.2014%2F12%2F1–3000%2F12%2F12&filter=hum_ani.humans. Accessed January 18, 2023.; https://pubmed.ncbi.nlm.nih.gov/?term=resveratrol&filter=pubt.clinicaltrial&filter=dates.2014%2F12%2F1-3000%2F12%2F12&filter=hum_ani.humans
Rabassa M, Zamora-Ros R, Urpi-Sarda M, et al. Association of habitual dietary resveratrol exposure with the development of frailty in older age: the Invecchiare in Chianti study. Am J Clin Nutr. 2015;102(6):1534–42. https://pubmed.ncbi.nlm.nih.gov/26490492/
Semba RD, Ferrucci L, Bartali B, et al. Resveratrol levels and all-cause mortality in older community-dwelling adults. JAMA Intern Med. 2014;174(7):1077–84. https://pubmed.ncbi.nlm.nih.gov/24819981/
Omidian M, Abdolahi M, Daneshzad E, et al. The effects of resveratrol on oxidative stress markers: a systematic review and meta-analysis of randomized clinical trials. Endocr Metab Immune Disord Drug Targets. 2020;20(5):718–27. https://pubmed.ncbi.nlm.nih.gov/31738139/
Koushki M, Lakzaei M, Khodabandehloo H, Hosseini H, Meshkani R, Panahi G. Therapeutic effect of resveratrol supplementation on oxidative stress: a systematic review and meta-analysis of randomised controlled trials. Postgrad Med J. 2020;96(1134):197–205. https://pubmed.ncbi.nlm.nih.gov/31628212/
Heger A, Ferk F, Nersesyan A, et al. Intake of a resveratrol-containing dietary supplement has no impact on DNA stability in healthy subjects. Mutat Res. 2012;749(1–2):82–6. https://pubmed.ncbi.nlm.nih.gov/22981768/
Zeraattalab-Motlagh S, Jayedi A, Shab-Bidar S. The effects of resveratrol supplementation in patients with type 2 diabetes, metabolic syndrome, and nonalcoholic fatty liver disease: an umbrella review of meta-analyses of randomized controlled trials. Am J Clin Nutr. 2021;114(5):1675–85. https://pubmed.ncbi.nlm.nih.gov/34320173/
Zhang T, He Q, Liu Y, Chen Z, Hu H. Efficacy and safety of resveratrol supplements on blood lipid and blood glucose control in patients with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Evid Based Complement Alternat Med. 2021;2021:5644171. https://pubmed.ncbi.nlm.nih.gov/34484395/
Zeraattalab-Motlagh S, Jayedi A, Shab-Bidar S. The effects of resveratrol supplementation in patients with type 2 diabetes, metabolic syndrome, and nonalcoholic fatty liver disease: an umbrella review of meta-analyses of randomized controlled trials. Am J Clin Nutr. 2021;114(5):1675–8. https://pubmed.ncbi.nlm.nih.gov/34320173/
Bashmakov YK, Assaad-Khalil SH, Abou Seif M, et al. Resveratrol promotes foot ulcer size reduction in type 2 diabetes patients. ISRN Endocrinol. 2014;2014:816307. https://pubmed.ncbi.nlm.nih.gov/24701359/
Moxey PW, Gogalniceanu P, Hinchliffe RJ, et al. Lower extremity amputations – a review of global variability in incidence. Diabet Med. 2011;28(10):1144–53. https://pubmed.ncbi.nlm.nih.gov/21388445/
Bhattarai G, Poudel SB, Kook SH, Lee JC. Resveratrol prevents alveolar bone loss in an experimental rat model of periodontitis. Acta Biomater. 2016;29:398–408. https://pubmed.ncbi.nlm.nih.gov/26497626/
Zhen L, Fan DS, Zhang Y, Cao XM, Wang LM. Resveratrol ameliorates experimental periodontitis in diabetic mice through negative regulation of TLR4 signaling. Acta Pharmacol Sin. 2015;36(2):221–8. https://pubmed.ncbi.nlm.nih.gov/25530164/
Javid AZ, Hormoznejad R, Yousefimanesh HA, Haghighi-Zadeh MH, Zakerkish M. Impact of resveratrol supplementation on inflammatory, antioxidant, and periodontal markers in type 2 diabetic patients with chronic periodontitis. Diabetes Metab Syndr. 2019;13(4):2769–74. https://pubmed.ncbi.nlm.nih.gov/31405706/
Samsamikor M, Daryani NE, Asl PR, Hekmatdoost A. Resveratrol supplementation and oxidative/anti-oxidative status in patients with ulcerative colitis: a randomized, double-blind, placebo-controlled pilot study. Arch Med Res. 2016;47(4):304–9. https://pubmed.ncbi.nlm.nih.gov/27664491/
Samsami-Kor M, Daryani NE, Asl PR, Hekmatdoost A. Anti-inflammatory effects of resveratrol in patients with ulcerative colitis: a randomized, double-blind, placebo-controlled pilot study. Arch Med Res. 2015;46(4):280–5. https://pubmed.ncbi.nlm.nih.gov/26002728/
Hussain SA, Marouf BH, Ali ZS, Ahmmad RS. Efficacy and safety of co-administration of resveratrol with meloxicam in patients with knee osteoarthritis: a pilot interventional study. Clin Interv Aging. 2018;13:1621–30. https://pubmed.ncbi.nlm.nih.gov/30233159/
Qasem RJ. The estrogenic activity of resveratrol: a comprehensive review of in vitro and in vivo evidence and the potential for endocrine disruption. Crit Rev Toxicol. 2020;50(5):439–62. https://pubmed.ncbi.nlm.nih.gov/32744480/
Dzator JSA, Howe PRC, Coupland KG, Wong RHX. A randomised, double-blind, placebo-controlled crossover trial of resveratrol supplementation for prophylaxis of hormonal migraine. Nutrients. 2022;14(9):1763. https://pubmed.ncbi.nlm.nih.gov/35565731/
Mansour A, Samadi M, Sanginabadi M, et al. Effect of resveratrol on menstrual cyclicity, hyperandrogenism and metabolic profile in women with PCOS. Clin Nutr. 2021;40(6):4106–12. https://pubmed.ncbi.nlm.nih.gov/33610422/
Zaw JJT, Howe PRC, Wong RHX. Long-term resveratrol supplementation improves pain perception, menopausal symptoms, and overall well-being in postmenopausal women: findings from a 24-month randomized, controlled, crossover trial. Menopause. 2020;28(1):40–9. https://pubmed.ncbi.nlm.nih.gov/32881835/
Li Q, Yang G, Xu H, Tang S, Lee WYW. Effects of resveratrol supplementation on bone quality: a systematic review and meta-analysis of randomized controlled trials. BMC Complement Med Ther. 2021;21(1):214. https://pubmed.ncbi.nlm.nih.gov/34420523/
Johnson JJ, Nihal M, Siddiqui IA, et al. Enhancing the bioavailability of resveratrol by combining it with piperine. Mol Nutr Food Res. 2011;55(8):1169–76. https://pubmed.ncbi.nlm.nih.gov/21714124/
Turner RS, Thomas RG, Craft S, et al. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology. 2015;85(16):1383-91 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4626244/
Gliemann L. What are the chances that resveratrol will be the drug of tomorrow? Pharmacol Res. 2018;129:139–40. https://pubmed.ncbi.nlm.nih.gov/29425727/
Semba RD, Ferrucci L, Bartali B, et al. Resveratrol levels and all-cause mortality in older community-dwelling adults. JAMA Intern Med. 2014;174(7):1077–84. https://pubmed.ncbi.nlm.nih.gov/24819981/
Wahab A, Gao K, Jia C, et al. Significance of resveratrol in clinical management of chronic diseases. Molecules. 2017;22(8):1329. https://pubmed.ncbi.nlm.nih.gov/28820474/
Scribbans TD, Ma JK, Edgett BA, et al. Resveratrol supplementation does not augment performance adaptations or fibre-type-specific responses to high-intensity interval training in humans. Appl Physiol Nutr Metab. 2014;39(11):1305–13. https://pubmed.ncbi.nlm.nih.gov/25211703/
Gliemann L, Schmidt JF, Olesen J, et al. Resveratrol blunts the positive effects of exercise training on cardiovascular health in aged men. J Physiol. 2013;591(Pt 20):5047–59. https://pubmed.ncbi.nlm.nih.gov/23878368/
Meng X, Zhou J, Zhao CN, Gan RY, Li HB. Health benefits and molecular mechanisms of resveratrol: a narrative review. Foods. 2020;9(3):340. https://pubmed.ncbi.nlm.nih.gov/32183376/
45,36 кг. – Примеч. ред.
Dybkowska E, Sadowska A, Swiderski F, Rakowska R, Wysocka K. The occurrence of resveratrol in foodstuffs and its potential for supporting cancer prevention and treatment. A review. Rocz Panstw Zakl Hig. 2018;69(1):5–14. https://pubmed.ncbi.nlm.nih.gov/29517181/
Morris BJ. Seven sirtuins for seven deadly diseases of aging. Free Radic Biol Med. 2013;56:133–71. https://pubmed.ncbi.nlm.nih.gov/23104101/
Lagouge M, Argmann C, Gerhart-Hines Z, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1a. Cell. 2006;127(6):1109–22. https://pubmed.ncbi.nlm.nih.gov/17112576/
Timmers S, Konings E, Bilet L, et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 2011;14(5):612–22. https://pubmed.ncbi.nlm.nih.gov/22055504/
Tang PCT, Ng YF, Ho S, Gyda M, Chan SW. Resveratrol and cardiovascular health – promising therapeutic or hopeless illusion? Pharmacol Res. 2014;90:88–115. https://pubmed.ncbi.nlm.nih.gov/25151891/
Gliemann L, Olesen J, Biensø RS, et al. Reply from Lasse Gliemann, Jesper Olesen, Rasmus Sjørup Biensø, Stefan Peter Mortensen, Michael Nyberg, Jens Bangsbo, Henriette Pilegaard and Ylva Hellsten. J Physiol. 2014;592(Pt 3):553. https://pubmed.ncbi.nlm.nih.gov/24488075/
Zhao L, Cao J, Hu K, et al. Sirtuins and their biological relevance in aging and age-related diseases. Aging Dis. 2020;11(4):927–45. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7390530/
Li D, Cui Y, Wang X, Liu F, Li X. Apple polyphenol extract alleviates lipid accumulation in free-fatty-acid-exposed HepG2 cells via activating autophagy mediated by SIRT1/AMPK signaling. Phytother Res. 2021;35(3):1416–31. https://pubmed.ncbi.nlm.nih.gov/33037751/
Gayer BA, Avendano EE, Edelson E, Nirmala N, Johnson EJ, Raman G. Effects of intake of apples, pears, or their products on cardiometabolic risk factors and clinical outcomes: a systematic review and meta-analysis. Curr Dev Nutr. 2019;3(10):nzz109. https://pubmed.ncbi.nlm.nih.gov/31667463/
Hodgson JM, Prince RL, Woodman RJ, et al. Apple intake is inversely associated with all-cause and disease-specific mortality in elderly women. Br J Nutr. 2016;115(5):860–7. https://pubmed.ncbi.nlm.nih.gov/26787402/
Spiegelhalter D. Using speed of ageing and “microlives” to communicate the effects of lifetime habits and environment. BMJ. 2012;345:e8223. https://pubmed.ncbi.nlm.nih.gov/23247978/
Xiang L, Sun K, Lu J, et al. Anti-aging effects of phloridzin, an apple polyphenol, on yeast via the SOD and Sir2 genes. Biosci Biotechnol Biochem. 2011;75(5):854–8. https://pubmed.ncbi.nlm.nih.gov/21597195/
Peng C, Chan HYE, Huang Y, Yu H, Chen ZY. Apple polyphenols extend the mean lifespan of Drosophila melanogaster. J Agric Food Chem. 2011;59(5):2097–106. https://pubmed.ncbi.nlm.nih.gov/21319854/
Shaposhnikov M, Latkin D, Plyusnina E, et al. The effects of pectins on life span and stress resistance in Drosophila melanogaster. Biogerontology. 2014;15(2):113–27. https://pubmed.ncbi.nlm.nih.gov/24305778/
Palermo V, Mattivi F, Silvestri R, La Regina G, Falcone C, Mazzoni C. Apple can act as anti-aging on yeast cells. Oxid Med Cell Longev. 2012;2012:491759. https://pubmed.ncbi.nlm.nih.gov/22970337/
Vayndorf EM, Lee SS, Liu RH. Whole apple extracts increase lifespan, healthspan and resistance to stress in Caenorhabditis elegans. J Funct Foods. 2013;5(3):1236–43. https://pubmed.ncbi.nlm.nih.gov/23878618/
Sunagawa T, Shimizu T, Kanda T, Tagashira M, Sami M, Shirasawa T. Procyanidins from apples (Malus pumila Mill.) extend the lifespan of Caenorhabditis elegans. Planta Med. 2011;77(2):122–7. https://pubmed.ncbi.nlm.nih.gov/20717869/
Song B, Wang H, Xia W, Zheng B, Li T, Liu RH. Combination of apple peel and blueberry extracts synergistically induced lifespan extension via DAF-16 in Caenorhabditis elegans. Food Funct. 2020;11(7):6170–85. https://pubs.rsc.org/en/content/articlelanding/2020/FO/D0FO00718H
Pallauf K, Giller K, Huebbe P, Rimbach G. Nutrition and healthy ageing: calorie restriction or polyphenol-rich “MediterrAsian” diet? Oxid Med Cell Longev. 2013;2013:707421. https://pubmed.ncbi.nlm.nih.gov/24069505/
Wu X, Cao N, Fenech M, Wang X. Role of sirtuins in maintenance of genomic stability: relevance to cancer and healthy aging. DNA Cell Biol. 2016;35(10):542–75. https://pubmed.ncbi.nlm.nih.gov/27380140/
Khazdouz M, Daryani NE, Alborzi F, et al. Effect of selenium supplementation on expression of SIRT1 and PGC-1a genes in ulcerative colitis patients: a double blind randomized clinical trial. Clin Nutr Res. 2020;9(4):284–95. https://pubmed.ncbi.nlm.nih.gov/33204668/
Stranges S, Marshall JR, Natarajan R, et al. Effects of long-term selenium supplementation on the incidence of type 2 diabetes: a randomized trial. Ann Intern Med. 2007;147(4):217–23. https://pubmed.ncbi.nlm.nih.gov/17620655/
Fusi J, Bianchi S, Daniele S, et al. An in vitro comparative study of the antioxidant activity and SIRT1 modulation of natural compounds. Biomed Pharmacother. 2018;101:805–19. https://pubmed.ncbi.nlm.nih.gov/29525677/
Yang Y, Duan W, Lin Y, et al. SIRT1 activation by curcumin pretreatment attenuates mitochondrial oxidative damage induced by myocardial ischemia reperfusion injury. Free Radic Biol Med. 2013;65:667–79. https://pubmed.ncbi.nlm.nih.gov/23880291/
Heshmati J, Golab F, Morvaridzadeh M, et al. The effects of curcumin supplementation on oxidative stress, Sirtuin-1 and peroxisome proliferator activated receptor ¿ coactivator 1a gene expression in polycystic ovarian syndrome (PCOS) patients: a randomized placebo-controlled clinical trial. Diabetes Metab Syndr. 2020;14(2):77–82. https://pubmed.ncbi.nlm.nih.gov/31991296/
Daneshi-Maskooni M, Keshavarz SA, Qorbani M, et al. Green cardamom supplementation improves serum irisin, glucose indices, and lipid profiles in overweight or obese non-alcoholic fatty liver disease patients: a double-blind randomized placebo-controlled clinical trial. BMC Complement Altern Med. 2019;19(1):59. https://pubmed.ncbi.nlm.nih.gov/30871514/
Daneshi-Maskooni M, Keshavarz SA, Qorbani M, et al. Green cardamom increases Sirtuin-1 and reduces inflammation in overweight or obese patients with non-alcoholic fatty liver disease: a double-blind randomized placebo-controlled clinical trial. Nutr Metab (Lond). 2018;15:63. https://pubmed.ncbi.nlm.nih.gov/30263038/
Zhong Y, Chen AF, Zhao J, Gu YJ, Fu GX. Serum levels of cathepsin D, sirtuin1, and endothelial nitric oxide synthase are correlatively reduced in elderly healthy people. Aging Clin Exp Res. 2016;28(4):641–5. https://pubmed.ncbi.nlm.nih.gov/26462844/
Kumar R, Mohan N, Upadhyay AD, et al. Identification of serum sirtuins as novel noninvasive protein markers for frailty. Aging Cell. 2014;13(6):975–80. https://pubmed.ncbi.nlm.nih.gov/25100619/
Kumar R, Chaterjee P, Sharma PK, et al. Sirtuin1: a promising serum protein marker for early detection of Alzheimer’s disease. PLoS One. 2013;8(4):e61560. https://pubmed.ncbi.nlm.nih.gov/23613875/
Yanagisawa S, Papaioannou AI, Papaporfyriou A, et al. Decreased serum sirtuin-1 in COPD. Chest. 2017;152(2):343–52. https://pubmed.ncbi.nlm.nih.gov/28506610/
Kazemi S, Yaghooblou F, Siassi F, et al. Cardamom supplementation improves inflammatory and oxidative stress biomarkers in hyperlipidemic, overweight, and obese pre-diabetic women: a randomized double-blind clinical trial. J Sci Food Agric. 2017;97(15):5296–301. https://pubmed.ncbi.nlm.nih.gov/28480505/
Shekarchizadeh-Esfahani P, Arab A, Ghaedi E, Hadi A, Jalili C. Effects of cardamom supplementation on lipid profile: a systematic review and meta-analysis of randomized controlled clinical trials. Phytother Res. 2020;34(3):475–85. https://pubmed.ncbi.nlm.nih.gov/31755188/
Daneshi-Maskooni M, Keshavarz SA, Qorbani M, et al. Green cardamom supplementation improves serum irisin, glucose indices, and lipid profiles in overweight or obese non-alcoholic fatty liver disease patients: a double-blind randomized placebo-controlled clinical trial. BMC Complement Altern Med. 2019;19(1):59. https://pubmed.ncbi.nlm.nih.gov/30871514/
Rajendrasozhan S, Yang SR, Kinnula VL, Rahman I. SIRT1, an antiinflammatory and antiaging protein, is decreased in lungs of patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008;177(8):861–70. https://pubmed.ncbi.nlm.nih.gov/18174544/
Caito S, Rajendrasozhan S, Cook S, et al. SIRT1 is a redox-sensitive deacetylase that is post-translationally modified by oxidants and carbonyl stress. FASEB J. 2010;24(9):3145–59. https://pubmed.ncbi.nlm.nih.gov/20385619/
Cai W, Uribarri J, Zhu L, et al. Oral glycotoxins are a modifiable cause of dementia and the metabolic syndrome in mice and humans. Proc Natl Acad Sci U S A. 2014;111(13):4940–5. https://pubmed.ncbi.nlm.nih.gov/24567379/
Rizzi L, Roriz-Cruz M. Sirtuin 1 and Alzheimer’s disease: an up-to-date review. Neuropeptides. 2018;71:54–60. https://pubmed.ncbi.nlm.nih.gov/30007474/
Cai W, Uribarri J, Zhu L, et al. Oral glycotoxins are a modifiable cause of dementia and the metabolic syndrome in mice and humans. Proc Natl Acad Sci U S A. 2014;111(13):4940–5. https://pubmed.ncbi.nlm.nih.gov/24567379/
Potthast AB, Nebl J, Wasserfurth P, et al. Impact of nutrition on short-term exercise-induced sirtuin regulation: vegans differ from omnivores and lacto-ovo vegetarians. Nutrients. 2020;12(4):1004. https://pubmed.ncbi.nlm.nih.gov/32260570/
Brenner C. Sirtuins are not conserved longevity genes. Life Metabolism. Published online September 22, 2022. https://academic.oup.com/lifemeta/advance-article/doi/10.1093/lifemeta/loac025/6711379. Accessed December 27, 2022.; https://academic.oup.com/lifemeta/article/1/2/122/6711379
Boccardi V, Mecocci P. Telomerase activation and human health-span: an open issue. Aging Clin Exp Res. 2018;30(2):221–3. https://pubmed.ncbi.nlm.nih.gov/28470632/
Shay JW, Wright WE. Telomeres and telomerase: three decades of progress. Nat Rev Genet. 2019;20(5):299–309. https://pubmed.ncbi.nlm.nih.gov/30760854/
Herrmann W, Herrmann M. The importance of telomere shortening for atherosclerosis and mortality. J Cardiovasc Dev Dis. 2020;7(3):29. https://pubmed.ncbi.nlm.nih.gov/32781553/
Serrano M, Blasco MA. Cancer and ageing: convergent and divergent mechanisms. Nat Rev Mol Cell Biol. 2007;8(9):715–22. https://pubmed.ncbi.nlm.nih.gov/17717516/
Bonafè M, Sabbatinelli J, Olivieri F. Exploiting the telomere machinery to put the brakes on inflamm-aging. Ageing Res Rev. 2020;59:101027. https://pubmed.ncbi.nlm.nih.gov/32068123/
Stone RC, Horvath K, Kark JD, Susser E, Tishkoff SA, Aviv A. Telomere length and the cancer – atherosclerosis trade-off. PLoS Genet. 2016;12(7):e1006144. https://pubmed.ncbi.nlm.nih.gov/27386863/
Shay JW, Wright WE. Telomeres and telomerase: three decades of progress. Nat Rev Genet. 2019;20(5):299–309. https://pubmed.ncbi.nlm.nih.gov/30760854/
Saretzki G. Telomeres, telomerase and ageing. Subcell Biochem. 2018;90:221–308. https://pubmed.ncbi.nlm.nih.gov/30779012/
Rizvi S, Raza ST, Mahdi F. Telomere length variations in aging and age-related diseases. Curr Aging Sci. 2014;7(3):161–7. https://pubmed.ncbi.nlm.nih.gov/25612739/
Wang J, Liu Y, Xia Q, et al. Potential roles of telomeres and telomerase in neurodegenerative diseases. Int J Biol Macromol. 2020;163:1060–78. https://pubmed.ncbi.nlm.nih.gov/32673712/
Leung CW, Laraia BA, Needham BL, et al. Soda and cell aging: associations between sugar-sweetened beverage consumption and leukocyte telomere length in healthy adults from the National Health and Nutrition Examination Surveys. Am J Public Health. 2014;104(12):2425–31. https://pubmed.ncbi.nlm.nih.gov/25322305/
Huang Z, Liu C, Ruan Y, et al. Dynamics of leukocyte telomere length in adults aged 50 and older: a longitudinal population-based cohort study. GeroScience. 2021;43(2):645–54. https://pubmed.ncbi.nlm.nih.gov/33469834/
Prieto-Oliveira P. Telomerase activation in the treatment of aging or degenerative diseases: a systematic review. Mol Cell Biochem. 2021;476(2):599–607. https://pubmed.ncbi.nlm.nih.gov/33001374/
Zhou J, Wang J, Shen Y, et al. The association between telomere length and frailty: a systematic review and meta-analysis. Exp Gerontol. 2018;106:16–20. https://pubmed.ncbi.nlm.nih.gov/29518479/
Cohen S, Janicki-Deverts D, Turner RB, et al. Association between telomere length and experimentally induced upper respiratory viral infection in healthy adults. JAMA. 2013;309(7):699–705. https://pubmed.ncbi.nlm.nih.gov/23423415/
Zhan Y, Clements MS, Roberts RO, et al. Association of telomere length with general cognitive trajectories: a meta-analysis of four prospective cohort studies. Neurobiol Aging. 2018;69:111–6. https://pubmed.ncbi.nlm.nih.gov/29870951/
Smith L, Luchini C, Demurtas J, et al. Telomere length and health outcomes: an umbrella review of systematic reviews and meta-analyses of observational studies. Ageing Res Rev. 2019;51:1–10. https://pubmed.ncbi.nlm.nih.gov/30776454/
Herrmann W, Herrmann M. The importance of telomere shortening for atherosclerosis and mortality. J Cardiovasc Dev Dis. 2020;7(3):29. https://pubmed.ncbi.nlm.nih.gov/32781553/
Zhan Y, Liu XR, Reynolds CA, Pedersen NL, Hägg S, Clements MS. Leukocyte telomere length and all-cause mortality: a between-within twin study with time-dependent effects using generalized survival models. Am J Epidemiol. 2018;187(10):2186–91. https://pubmed.ncbi.nlm.nih.gov/29961868/
Christensen K, Thinggaard M, McGue M, et al. Perceived age as clinically useful biomarker of ageing: cohort study. BMJ. 2009;339:b5262. https://pubmed.ncbi.nlm.nih.gov/20008378/
Christensen K, Thinggaard M, McGue M, et al. Perceived age as clinically useful biomarker of ageing: cohort study. BMJ. 2009;339:b5262. https://pubmed.ncbi.nlm.nih.gov/20008378/
Zhan Y, Hägg S. Association between genetically predicted telomere length and facial skin aging in the UK Biobank: a Mendelian randomization study. GeroScience. 2021;43(3):1519–25. https://pubmed.ncbi.nlm.nih.gov/33033864/
Astuti Y, Wardhana A, Watkins J, Wulaningsih W. Cigarette smoking and telomere length: a systematic review of 84 studies and meta-analysis. Environ Res. 2017;158:480–9. https://pubmed.ncbi.nlm.nih.gov/28704792/
Aviv A, Shay JW. Reflections on telomere dynamics and ageing-related diseases in humans. Philos Trans R Soc Lond B Biol Sci. 2018;373(1741):20160436. https://pubmed.ncbi.nlm.nih.gov/29335375/
Whittemore K, Vera E, Martínez-Nevado E, Sanpera C, Blasco MA. Telomere shortening rate predicts species life span. Proc Natl Acad Sci U S A. 2019;116(30):15122–7. https://pubmed.ncbi.nlm.nih.gov/31285335/
Fick LJ, Fick GH, Li Z, et al. Telomere length correlates with life span of dog breeds. Cell Rep. 2012;2(6):1530–6. https://pubmed.ncbi.nlm.nih.gov/23260664/
Muñoz-Lorente MA, Cano-Martin AC, Blasco MA. Mice with hyper-long telomeres show less metabolic aging and longer lifespans. Nat Commun. 2019;10(1):4723. https://pubmed.ncbi.nlm.nih.gov/31624261/
Blackburn EH, Epel ES, Lin J. Human telomere biology: a contributory and interactive factor in aging, disease risks, and protection. Science. 2015;350(6265):1193–8. https://pubmed.ncbi.nlm.nih.gov/26785477/
Zhu Y, Liu X, Ding X, Wang F, Geng X. Telomere and its role in the aging pathways: telomere shortening, cell senescence and mitochondria dysfunction. Biogerontology. 2019;20(1):1–16. https://pubmed.ncbi.nlm.nih.gov/30229407/
Blackburn EH, Epel ES, Lin J. Human telomere biology: a contributory and interactive factor in aging, disease risks, and protection. Science. 2015;350(6265):1193–8. https://pubmed.ncbi.nlm.nih.gov/26785477/
Tsuji A, Ishiko A, Takasaki T, Ikeda N. Estimating age of humans based on telomere shortening. Forensic Sci Int. 2002;126(3):197–9. https://pubmed.ncbi.nlm.nih.gov/12062940/
Huang Z, Liu C, Ruan Y, et al. Dynamics of leukocyte telomere length in adults aged 50 and older: a longitudinal population-based cohort study. GeroScience. 2021;43(2):645–54. https://pubmed.ncbi.nlm.nih.gov/33469834/
Blackburn EH. Telomeres and telomerase: the means to the end (Nobel lecture). Angew Chemie Int Ed Engl. 2010;49(41):7405–21. https://pubmed.ncbi.nlm.nih.gov/20821774/
Laberthonnière C, Magdinier F, Robin JD. Bring it to an end: does telomeres size matter? Cells. 2019;8(1):30. https://pubmed.ncbi.nlm.nih.gov/30626097/
Saretzki G. Telomeres, telomerase and ageing. Subcell Biochem. 2018;90:221–308. https://pubmed.ncbi.nlm.nih.gov/30779012/
Boccardi V, Mecocci P. Telomerase activation and human health-span: an open issue. Aging Clin Exp Res. 2018;30(2):221–3. https://pubmed.ncbi.nlm.nih.gov/28470632/
Flanary BE, Kletetschka G. Analysis of telomere length and telomerase activity in tree species of various life-spans, and with age in the bristlecone pine Pinus longaeva. Biogerontology. 2005;6(2):101–11. https://pubmed.ncbi.nlm.nih.gov/16034678/
Wright WE, Piatyszek MA, Rainey WE, Byrd W, Shay JW. Telomerase activity in human germline and embryonic tissues and cells. Dev Genet. 1996;18(2):173–9. https://pubmed.ncbi.nlm.nih.gov/8934879/
Shay JW, Bacchetti S. A survey of telomerase activity in human cancer. Eur J Cancer. 1997;33(5):787–91. https://pubmed.ncbi.nlm.nih.gov/9282118/
Lulkiewicz M, Bajsert J, Kopczynski P, Barczak W, Rubis B. Telomere length: how the length makes a difference. Mol Biol Rep. 2020;47(9):7181–8. https://pubmed.ncbi.nlm.nih.gov/32876842/
Huang Z, Liu C, Ruan Y, et al. Dynamics of leukocyte telomere length in adults aged 50 and older: a longitudinal population-based cohort study. GeroScience. 2021;43(2):645–54. https://pubmed.ncbi.nlm.nih.gov/33469834/
Chen W, Kimura M, Kim S, et al. Longitudinal versus cross-sectional evaluations of leukocyte telomere length dynamics: age-dependent telomere shortening is the rule. J Gerontol A Biol Sci Med Sci. 2011;66(3):312–9. https://pubmed.ncbi.nlm.nih.gov/21310811/
Epel ES, Merkin SS, Cawthon R, et al. The rate of leukocyte telomere shortening predicts mortality from cardiovascular disease in elderly men. Aging (Albany NY). 2008;1(1):81–8. https://pubmed.ncbi.nlm.nih.gov/20195384/
Tedone E, Arosio B, Gussago C, et al. Leukocyte telomere length and prevalence of age-related diseases in semisupercentenarians, centenarians and centenarians’ offspring. Exp Gerontol. 2014;58:90–5. https://pubmed.ncbi.nlm.nih.gov/24975295/
Tedone E, Huang E, O’Hara R, et al. Telomere length and telomerase activity in T cells are biomarkers of high-performing centenarians. Aging Cell. 2019;18(1):e12859. https://pubmed.ncbi.nlm.nih.gov/30488553/
Kamal S, Junaid M, Ejaz A, Bibi I, Akash MSH, Rehman K. The secrets of telomerase: retrospective analysis and future prospects. Life Sci. 2020;257:118115. https://pubmed.ncbi.nlm.nih.gov/32698073/
Boccardi V, Paolisso G. Telomerase activation: a potential key modulator for human healthspan and longevity. Ageing Res Rev. 2014;15:1–5. https://pubmed.ncbi.nlm.nih.gov/24561251/
Bär C, Blasco MA. Telomeres and telomerase as therapeutic targets to prevent and treat age-related diseases. F1000Res. 2016;5:89. https://pubmed.ncbi.nlm.nih.gov/27081482/
Tomás-Loba A, Flores I, Fernández-Marcos PJ, et al. Telomerase reverse transcriptase delays aging in cancer-resistant mice. Cell. 2008;135(4):609–22. https://pubmed.ncbi.nlm.nih.gov/19013273/
Bernardes de Jesus B, Vera E, Schneeberger K, et al. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol Med. 2012;4(8):691–704. https://pubmed.ncbi.nlm.nih.gov/22585399/
Bär C, Bernardes de Jesus B, Serrano R, et al. Telomerase expression confers cardioprotection in the adult mouse heart after acute myocardial infarction. Nat Commun. 2014;5:5863. https://pubmed.ncbi.nlm.nih.gov/25519492/
Rudolph KL, Chang S, Millard M, Schreiber-Agus N, DePinho RA. Inhibition of experimental liver cirrhosis in mice by telomerase gene delivery. Science. 2000;287(5456):1253–8. https://pubmed.ncbi.nlm.nih.gov/10678830/
Bär C, Bernardes de Jesus B, Serrano R, et al. Telomerase expression confers cardioprotection in the adult mouse heart after acute myocardial infarction. Nat Commun. 2014;5:5863. https://pubmed.ncbi.nlm.nih.gov/25519492/
Bernardes de Jesus B, Vera E, Schneeberger K, et al. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol Med. 2012;4(8):691–704. https://pubmed.ncbi.nlm.nih.gov/22585399/
Eitan E, Tichon A, Gazit A, Gitler D, Slavin S, Priel E. Novel telomerase-increasing compound in mouse brain delays the onset of amyotrophic lateral sclerosis. EMBO Mol Med. 2012;4(4):313–29. https://pubmed.ncbi.nlm.nih.gov/22351600/
Gilson E, Ségal-Bendirdjian E. The telomere story or the triumph of an open-minded research. Biochimie. 2010;92(4):321–6. https://pubmed.ncbi.nlm.nih.gov/20096746/
Suram A, Herbig U. The replicometer is broken: telomeres activate cellular senescence in response to genotoxic stresses. Aging Cell. 2014;13(5):780–6. https://pubmed.ncbi.nlm.nih.gov/25040628/
Shay JW, Wright WE. Telomeres and telomerase: three decades of progress. Nat Rev Genet. 2019;20(5):299–309. https://pubmed.ncbi.nlm.nih.gov/30760854/
Hornsby PJ. Telomerase and the aging process. Exp Gerontol. 2007;42(7):575–81. https://pubmed.ncbi.nlm.nih.gov/17482404/
Bodnar AG, Ouellette M, Frolkis M, et al. Extension of life-span by introduction of telomerase into normal human cells. Science. 1998;279(5349):349–52. https://pubmed.ncbi.nlm.nih.gov/9454332/
Bernardes de Jesus B, Vera E, Schneeberger K, et al. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol Med. 2012;4(8):691–704. https://pubmed.ncbi.nlm.nih.gov/22585399/
Huang Z, Liu C, Ruan Y, et al. Dynamics of leukocyte telomere length in adults aged 50 and older: a longitudinal population-based cohort study. GeroScience. 2021;43(2):645–54. https://pubmed.ncbi.nlm.nih.gov/33469834/
Broer L, Codd V, Nyholt DR, et al. Meta-analysis of telomere length in 19713 subjects reveals high heritability, stronger maternal inheritance and a paternal age effect. Eur J Hum Genet. 2013;21(10):1163–8. https://pubmed.ncbi.nlm.nih.gov/23321625/
Maugeri A, Barchitta M, Magnano San Lio R, et al. The effect of alcohol on telomere length: a systematic review of epidemiological evidence and a pilot study during pregnancy. Int J Environ Res Public Health. 2021;18(9):5038. https://pubmed.ncbi.nlm.nih.gov/34068820/
Ip P, Chung BHY, Ho FKW, et al. Prenatal tobacco exposure shortens telomere length in children. Nicotine Tob Res. 2017;19(1):111–8. https://pubmed.ncbi.nlm.nih.gov/27194546/
Zhao B, Vo HQ, Johnston FH, Negishi K. Air pollution and telomere length: a systematic review of 12,058 subjects. Cardiovasc Diagn Ther. 2018;8(4):480–92. https://pubmed.ncbi.nlm.nih.gov/30214863/
Aviv A, Shay JW. Reflections on telomere dynamics and ageing-related diseases in humans. Philos Trans R Soc Lond B Biol Sci. 2018;373(1741):20160436. https://pubmed.ncbi.nlm.nih.gov/29335375/
Galiè S, Canudas S, Muralidharan J, García-Gavilán J, Bulló M, Salas-Salvadó J. Impact of nutrition on telomere health: systematic review of observational cohort studies and randomized clinical trials. Adv Nutr. 2020;11(3):576–601. https://pubmed.ncbi.nlm.nih.gov/31688893/
Ornish D, Brown SE, Scherwitz LW, et al. Can lifestyle changes reverse coronary heart disease? The Lifestyle Heart Trial. Lancet. 1990;336(8708):129–33. https://pubmed.ncbi.nlm.nih.gov/1973470/
Ornish D, Weidner G, Fair WR, et al. Intensive lifestyle changes may affect the progression of prostate cancer. J Urol. 2005;174(3):1065–70. https://pubmed.ncbi.nlm.nih.gov/16094059/
U.S. National Library of Medicine. Can lifestyle changes reverse early-stage Alzheimer’s disease. ClincalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT04606420. Updated October 28, 2020. Accessed July 17, 2021.; https://clinicaltrials.gov/ct2/show/NCT04606420
Ornish D, Lin J, Daubenmier J, et al. Increased telomerase activity and comprehensive lifestyle changes: a pilot study. Lancet Oncol. 2008;9(11):1048–57. https://pubmed.ncbi.nlm.nih.gov/18799354/
Ornish D, Lin J, Daubenmier J, et al. Increased telomerase activity and comprehensive lifestyle changes: a pilot study. Lancet Oncol. 2008;9(11):1048–57. https://pubmed.ncbi.nlm.nih.gov/18799354/
Skordalakes E. Telomerase and the benefits of healthy living. Lancet Oncol. 2008;9(11):1023–4. https://pubmed.ncbi.nlm.nih.gov/19012852/
Ornish D, Lin J, Chan JM, et al. Effect of comprehensive lifestyle changes on telomerase activity and telomere length in men with biopsy-proven low-risk prostate cancer: 5-year follow-up of a descriptive pilot study. Lancet Oncol. 2013;14(11):1112–20. https://pubmed.ncbi.nlm.nih.gov/24051140/
В российском прокате – «Отпуск по обмену». – Примеч. ред.
Blackburn EH, Epel ES. Too toxic to ignore. Nature. 2012;490(7419):169–71. https://pubmed.ncbi.nlm.nih.gov/23060172/
Epel ES, Lin J, Dhabhar FS, et al. Dynamics of telomerase activity in response to acute psychological stress. Brain Behav Immun. 2010;24(4):531–9. https://pubmed.ncbi.nlm.nih.gov/20018236/
Damjanovic AK, Yang Y, Glaser R, et al. Accelerated telomere erosion is associated with a declining immune function of caregivers of Alzheimer’s disease patients. J Immunol. 2007;179(6):4249–54. https://pubmed.ncbi.nlm.nih.gov/17785865/
Schutte NS, Malouff JM, Keng SL. Meditation and telomere length: a meta-analysis. Psychol Health. 2020;35(8):901–15. https://pubmed.ncbi.nlm.nih.gov/31903785/
Cherkas LF, Hunkin JL, Kato BS, et al. The association between physical activity in leisure time and leukocyte telomere length. Arch Intern Med. 2008;168(2):154–8. https://pubmed.ncbi.nlm.nih.gov/18227361/
Tucker LA. Walking and biologic ageing: evidence based on NHANES telomere data. J Sports Sci. 2020;38(9):1026–35. https://pubmed.ncbi.nlm.nih.gov/32175820/
Lin X, Zhou J, Dong B. Effect of different levels of exercise on telomere length: A systematic review and meta-analysis. J Rehabil Med. 2019;51(7):473–8. https://pubmed.ncbi.nlm.nih.gov/31093683/
Mundstock E, Zatti H, Louzada FM, et al. Effects of physical activity in telomere length: Systematic review and meta-analysis. Ageing Res Rev. 2015;22:72–80. https://pubmed.ncbi.nlm.nih.gov/25956165/
Abrahin O, Cortinhas-Alves EA, Vieira RP, Guerreiro JF. Elite athletes have longer telomeres than sedentary subjects: a meta-analysis. Exp Gerontol. 2019;119:138–45. https://pubmed.ncbi.nlm.nih.gov/30735724/
Aguiar SS, Sousa CV, Santos PA, et al. Master athletes have longer telomeres than age-matched non-athletes. A systematic review, meta-analysis and discussion of possible mechanisms. Exp Gerontol. 2021;146:111212. https://pubmed.ncbi.nlm.nih.gov/33387607/
Denham J, Nelson CP, O’Brien BJ, et al. Longer leukocyte telomeres are associated with ultra-endurance exercise independent of cardiovascular risk factors. PLoS One. 2013;8(7):e69377. https://pubmed.ncbi.nlm.nih.gov/23936000/
Werner C, Fürster T, Widmann T, et al. Physical exercise prevents cellular senescence in circulating leukocytes and in the vessel wall. Circulation. 2009;120(24):2438–47. https://pubmed.ncbi.nlm.nih.gov/19948976/
Friedenreich CM, Wang Q, Ting NS, et al. Effect of a 12-month exercise intervention on leukocyte telomere length: results from the ALPHA Trial. Cancer Epidemiol. 2018;56:67–74. https://pubmed.ncbi.nlm.nih.gov/30075329/
Sjögren P, Fisher R, Kallings L, Svenson U, Roos G, Hellénius ML. Stand up for health – avoiding sedentary behaviour might lengthen your telomeres: secondary outcomes from a physical activity RCT in older people. Br J Sports Med. 2014;48(19):1407–9. https://pubmed.ncbi.nlm.nih.gov/25185586/
Mason C, Risques RA, Xiao L, et al. Independent and combined effects of dietary weight loss and exercise on leukocyte telomere length in postmenopausal women. Obesity (Silver Spring). 2013;21(12):E549–54. https://pubmed.ncbi.nlm.nih.gov/23640743/
Werner CM, Hecksteden A, Morsch A, et al. Differential effects of endurance, interval, and resistance training on telomerase activity and telomere length in a randomized, controlled study. Eur Heart J. 2019;40(1):34–46. https://pubmed.ncbi.nlm.nih.gov/30496493/
Werner CM, Hecksteden A, Morsch A, et al. Differential effects of endurance, interval, and resistance training on telomerase activity and telomere length in a randomized, controlled study. Eur Heart J. 2019;40(1):34–46. https://pubmed.ncbi.nlm.nih.gov/30496493/
To-Miles FYL, Backman CL. What telomeres say about activity and health: a rapid review. Can J Occup Ther. 2016;83(3):143–53. https://pubmed.ncbi.nlm.nih.gov/27053148/
Mason C, Risques RA, Xiao L, et al. Independent and combined effects of dietary weight loss and exercise on leukocyte telomere length in postmenopausal women. Obesity (Silver Spring). 2013;21(12):E549–54. https://pubmed.ncbi.nlm.nih.gov/23640743/
Himbert C, Thompson H, Ulrich CM. Effects of intentional weight loss on markers of oxidative stress, DNA repair and telomere length – a systematic review. Obes Facts. 2017;10(6):648–65. https://pubmed.ncbi.nlm.nih.gov/29237161/
Ornish D, Lin J, Daubenmier J, et al. Increased telomerase activity and comprehensive lifestyle changes: a pilot study. Lancet Oncol. 2008;9(11):1048–57. https://pubmed.ncbi.nlm.nih.gov/18799354/
Ornish D, Lin J, Chan JM, et al. Effect of comprehensive lifestyle changes on telomerase activity and telomere length in men with biopsy-proven low-risk prostate cancer: 5-year follow-up of a descriptive pilot study. Lancet Oncol. 2013;14(11):1112–20. https://pubmed.ncbi.nlm.nih.gov/24051140/
Lulkiewicz M, Bajsert J, Kopczynski P, Barczak W, Rubis B. Telomere length: how the length makes a difference. Mol Biol Rep. 2020;47(9):7181–8. https://pubmed.ncbi.nlm.nih.gov/32876842/
Prieto-Oliveira P. Telomerase activation in the treatment of aging or degenerative diseases: a systematic review. Mol Cell Biochem. 2021;476(2):599–607. https://pubmed.ncbi.nlm.nih.gov/33001374/
De Meyer T, Bekaert S, De Buyzere ML, et al. Leukocyte telomere length and diet in the apparently healthy, middle-aged Asklepios population. Sci Rep. 2018;8(1):6540. https://pubmed.ncbi.nlm.nih.gov/29695838/
Tucker LA. Milk fat intake and telomere length in U.S. women and men: the role of the milk fat fraction. Oxid Med Cell Longev. 2019;2019:1574021. https://pubmed.ncbi.nlm.nih.gov/31772698/
Marin C, Delgado-Lista J, Ramirez R, et al. Mediterranean diet reduces senescence-associated stress in endothelial cells. Age (Dordr). 2012;34(6):1309–16. https://pubmed.ncbi.nlm.nih.gov/21894446/
Alonso-Pedrero L, Ojeda-Rodríguez A, Martínez-González MA, Zalba G, Bes-Rastrollo M, Marti A. Ultra-processed food consumption and the risk of short telomeres in an elderly population of the Seguimiento Universidad de Navarra (SUN) Project. Am J Clin Nutr. 2020;111(6):1259–66. https://pubmed.ncbi.nlm.nih.gov/32330232/
Askari M, Heshmati J, Shahinfar H, Tripathi N, Daneshzad E. Ultra-processed food and the risk of overweight and obesity: a systematic review and meta-analysis of observational studies. Int J Obes (Lond). 2020;44(10):2080–91. https://pubmed.ncbi.nlm.nih.gov/32796919/
Pagliai G, Dinu M, Madarena MP, Bonaccio M, Iacoviello L, Sofi F. Consumption of ultra-processed foods and health status: a systematic review and meta-analysis. Br J Nutr. 2021;125(3):308–18. https://pubmed.ncbi.nlm.nih.gov/32792031/
Strandberg TE, Strandberg AY, Saijonmaa O, Tilvis RS, Pitkälä KH, Fyhrquist F. Association between alcohol consumption in healthy midlife and telomere length in older men. The Helsinki Businessmen Study. Eur J Epidemiol. 2012;27(10):815–22. https://pubmed.ncbi.nlm.nih.gov/22875407/
Maugeri A, Barchitta M, Magnano San Lio R, et al. The effect of alcohol on telomere length: a systematic review of epidemiological evidence and a pilot study during pregnancy. Int J Environ Res Public Health. 2021;18(9):5038. https://pubmed.ncbi.nlm.nih.gov/34068820/
Huang Y, Cao D, Chen Z, et al. Red and processed meat consumption and cancer outcomes: umbrella review. Food Chem. 2021;356:129697. https://pubmed.ncbi.nlm.nih.gov/33838606/
Fretts AM, Howard BV, Siscovick DS, et al. Processed meat, but not unprocessed red meat, is inversely associated with leukocyte telomere length in the Strong Heart Family Study. J Nutr. 2016;146(10):2013–8. https://pubmed.ncbi.nlm.nih.gov/22277554/
De Meyer T, Bekaert S, De Buyzere ML, et al. Leukocyte telomere length and diet in the apparently healthy, middle-aged Asklepios population. Sci Rep. 2018;8(1):6540. https://pubmed.ncbi.nlm.nih.gov/29695838/
Nettleton JA, Diez-Roux A, Jenny NS, Fitzpatrick AL, Jacobs DR Jr. Dietary patterns, food groups, and telomere length in the Multi-Ethnic Study of Atherosclerosis (MESA). Am J Clin Nutr. 2008;88(5):1405–12. https://pubmed.ncbi.nlm.nih.gov/18996878/
Galiè S, Canudas S, Muralidharan J, García-Gavilán J, Bulló M, Salas-Salvadó J. Impact of nutrition on telomere health: systematic review of observational cohort studies and randomized clinical trials. Adv Nutr. 2020;11(3):576–601. https://pubmed.ncbi.nlm.nih.gov/31688893/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Farzaneh-Far R, Lin J, Epel ES, Harris WS, Blackburn EH, Whooley MA. Association of marine omega-3 fatty acid levels with telomeric aging in patients with coronary heart disease. JAMA. 2010;303(3):250. https://pubmed.ncbi.nlm.nih.gov/20085953/
Pawelczyk T, Grancow-Grabka M, Trafalska E, Szemraj J, Zurner N, Pawelczyk A. Telomerase level increase is related to n-3 polyunsaturated fatty acid efficacy in first episode schizophrenia: secondary outcome analysis of the OFFER randomized clinical trial. Prog Neuropsychopharmacol Biol Psychiatry. 2018;83:142–8. https://pubmed.ncbi.nlm.nih.gov/31098654/
O’Callaghan N, Parletta N, Milte CM, Benassi-Evans B, Fenech M, Howe PRC. Telomere shortening in elderly individuals with mild cognitive impairment may be attenuated with ¿-3 fatty acid supplementation: a randomized controlled pilot study. Nutrition. 2014;30(4):489–91. https://pubmed.ncbi.nlm.nih.gov/24342530/
Holub A, Mousa S, Abdolahi A, et al. The effects of aspirin and N-3 fatty acids on telomerase activity in adults with diabetes mellitus. Nutr Metab Cardiovasc Dis. 2020;30(10):1795–9. https://pubmed.ncbi.nlm.nih.gov/32723580/
Kiecolt-Glaser JK, Epel ES, Belury MA, et al. Omega-3 fatty acids, oxidative stress, and leukocyte telomere length: a randomized controlled trial. Brain Behav Immun. 2013;28:16–24. https://pubmed.ncbi.nlm.nih.gov/23010452/
Barden A, O’Callaghan N, Burke V, et al. n–3 fatty acid supplementation and leukocyte telomere length in patients with chronic kidney disease. Nutrients. 2016;8(3):175. https://pubmed.ncbi.nlm.nih.gov/27007392/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Pitkänen N, Pahkala K, Rovio SP, et al. Effects of randomized controlled infancy-onset dietary intervention on leukocyte telomere length – the Special Turku Coronary Risk Factor Intervention Project (STRIP). Nutrients. 2021;13(2):318. https://pubmed.ncbi.nlm.nih.gov/33499376/
Marin C, Delgado-Lista J, Ramirez R, et al. Mediterranean diet reduces senescence-associated stress in endothelial cells. Age (Dordr). 2012;34(6):1309–16. https://pubmed.ncbi.nlm.nih.gov/21894446/
Canudas S, Becerra-Tomás N, Hernández-Alonso P, et al. Mediterranean diet and telomere length: a systematic review and meta-analysis. Adv Nutr. 2020;11(6):1544–54. https://pubmed.ncbi.nlm.nih.gov/32730558/
Tucker LA. Milk fat intake and telomere length in U.S. women and men: the role of the milk fat fraction. Oxid Med Cell Longev. 2019;2019:e1574021. https://pubmed.ncbi.nlm.nih.gov/31772698/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Tucker LA. Dietary fiber and telomere length in 5674 U.S. adults: an NHANES study of biological aging. Nutrients. 2018;10(4):400. https://pubmed.ncbi.nlm.nih.gov/29570620/
Fretts AM, Howard BV, Siscovick DS, et al. Processed meat, but not unprocessed red meat, is inversely associated with leukocyte telomere length in the Strong Heart Family Study. J Nutr. 2016;146(10):2013–8. https://pubmed.ncbi.nlm.nih.gov/22277554/
Leung CW, Laraia BA, Needham BL, et al. Soda and cell aging: associations between sugar-sweetened beverage consumption and leukocyte telomere length in healthy adults from the National Health and Nutrition Examination Surveys. Am J Public Health. 2014;104(12):2425–31. https://pubmed.ncbi.nlm.nih.gov/25322305/
Valdes AM, Andrew T, Gardner JP, et al. Obesity, cigarette smoking, and telomere length in women. Lancet. 2005;366(9486):662–4. https://pubmed.ncbi.nlm.nih.gov/16112303/
Institute of Medicine. Dietary Reference Intakes: Proposed Definition of Dietary Fiber. National Academies Press; 2001. https://pubmed.ncbi.nlm.nih.gov/25057569/
Xu Q, Parks CG, DeRoo LA, Cawthon RM, Sandler DP, Chen H. Multivitamin use and telomere length in women. Am J Clin Nutr. 2009;89(6):1857–63. https://pubmed.ncbi.nlm.nih.gov/19279081/
Min KB, Min JY. Association between leukocyte telomere length and serum carotenoid in US adults. Eur J Nutr. 2017;56(3):1045–52. https://pubmed.ncbi.nlm.nih.gov/26818530/
Liu JJ, Crous-Bou M, Giovannucci E, De Vivo I. Coffee consumption is positively associated with longer leukocyte telomere length in the Nurses’ Health Study. J Nutr. 2016;146(7):1373–8. https://pubmed.ncbi.nlm.nih.gov/27281805/
Tucker LA. Caffeine consumption and telomere length in men and women of the National Health and Nutrition Examination Survey (NHANES). Nutr Metab (Lond). 2017;14(1):10. https://pubmed.ncbi.nlm.nih.gov/28603543/
Freitas-Simoes TM, Ros E, Sala-Vila A. Telomere length as a biomarker of accelerated aging: is it influenced by dietary intake? Curr Opin Clin Nutr Metab Care. 2018;21(6):430–6. https://pubmed.ncbi.nlm.nih.gov/30148739/
Chan R, Woo J, Suen E, Leung J, Tang N. Chinese tea consumption is associated with longer telomere length in elderly Chinese men. Br J Nutr. 2010;103(1):107–13. https://pubmed.ncbi.nlm.nih.gov/19671205/
Sheng R, Gu ZL, Xie ML. Epigallocatechin gallate, the major component of polyphenols in green tea, inhibits telomere attrition mediated cardiomyocyte apoptosis in cardiac hypertrophy. Int J Cardiol. 2013;162(3):199–209. https://pubmed.ncbi.nlm.nih.gov/22000973/
Rusak G, Komes D, Likic S, Horžic D, Kovac M. Phenolic content and antioxidative capacity of green and white tea extracts depending on extraction conditions and the solvent used. Food Chem. 2008;110(4):852–8. https://pubmed.ncbi.nlm.nih.gov/26047270/
Hovanloo F, Fallah Huseini H, Hedayati M, Teimourian M. Effects of aerobic training combined with green tea extract on leukocyte telomere length, quality of life and body composition in elderly women. J Med Plants. 2016;15(59):47–57. https://www.researchgate.net/publication/309402738_Effects_of_Aerobic_Training_Combined_with_Green_Tea_Extract_on_Leukocyte_Telomere_Length_Quality_of_Life_and_Body_Composition_in_Elderly_Women
Tran HTT, Schreiner M, Schlotz N, Lamy E. Short-term dietary intervention with cooked but not raw Brassica leafy vegetables increases telomerase activity in CD8+ lymphocytes in a randomized human trial. Nutrients. 2019;11(4):786. https://pubmed.ncbi.nlm.nih.gov/30959753/
Sarma DN, Barrett ML, Chavez ML, et al. Safety of green tea extracts: a systematic review by the US Pharmacopeia. Drug Saf. 2008;31(6):469–84. https://pubmed.ncbi.nlm.nih.gov/18484782/
Yu Z, Samavat H, Dostal AM, et al. Effect of green tea supplements on liver enzyme elevation: results from a randomized intervention study in the United States. Cancer Prev Res (Phila). 2017;10(10):571–9. https://pubmed.ncbi.nlm.nih.gov/28765194/
Hu J, Webster D, Cao J, Shao A. The safety of green tea and green tea extract consumption in adults – results of a systematic review. Regul Toxicol Pharmacol. 2018;95:412–33. https://pubmed.ncbi.nlm.nih.gov/29580974/
O’Callaghan N, Parletta N, Milte CM, Benassi-Evans B, Fenech M, Howe PRC. Telomere shortening in elderly individuals with mild cognitive impairment may be attenuated with ¿-3 fatty acid supplementation: a randomized controlled pilot study. Nutrition. 2014;30(4):489–91. https://pubmed.ncbi.nlm.nih.gov/24342530/
Holub A, Mousa S, Abdolahi A, et al. The effects of aspirin and N-3 fatty acids on telomerase activity in adults with diabetes mellitus. Nutr Metab Cardiovasc Dis. 2020;30(10):1795–9. https://pubmed.ncbi.nlm.nih.gov/32723580/
Kiecolt-Glaser JK, Epel ES, Belury MA, et al. Omega-3 fatty acids, oxidative stress, and leukocyte telomere length: a randomized controlled trial. Brain Behav Immun. 2013;28:16–24. https://pubmed.ncbi.nlm.nih.gov/23010452/
Barden A, O’Callaghan N, Burke V, et al. n–3 fatty acid supplementation and leukocyte telomere length in patients with chronic kidney disease. Nutrients. 2016;8(3):175. https://pubmed.ncbi.nlm.nih.gov/27007392/
García-Calzón S, Martínez-González MA, Razquin C, et al. Mediterranean diet and telomere length in high cardiovascular risk subjects from the PREDIMED-NAVARRA study. Clin Nutr. 2016;35(6):1399–405. https://pubmed.ncbi.nlm.nih.gov/27083496/
Pusceddu I, Herrmann M, Kirsch SH, et al. Prospective study of telomere length and LINE-1 methylation in peripheral blood cells: the role of B vitamins supplementation. Eur J Nutr. 2016;55(5):1863–73. https://pubmed.ncbi.nlm.nih.gov/27083496/
Sharif R, Thomas P, Zalewski P, Fenech M. Zinc supplementation influences genomic stability biomarkers, antioxidant activity, and zinc transporter genes in an elderly Australian population with low zinc status. Mol Nutr Food Res. 2015;59(6):1200–12. https://pubmed.ncbi.nlm.nih.gov/25755079/
Zarei M, Zarezadeh M, Hamedi Kalajahi F, Javanbakht MH. The relationship between vitamin D and telomere/telomerase: a comprehensive review. J Frailty Aging. 2021;10(1):2–9. https://pubmed.ncbi.nlm.nih.gov/33331615/
Zhu H, Guo D, Li K, et al. Increased telomerase activity and vitamin D supplementation in overweight African Americans. Int J Obes (Lond). 2012;36(6):805–9. https://pubmed.ncbi.nlm.nih.gov/21986705/
Yang T, Wang H, Xiong Y, et al. Vitamin D supplementation improves cognitive function through reducing oxidative stress regulated by telomere length in older adults with mild cognitive impairment: a 12-month randomized controlled trial. J Alzheimers Dis. 2020;78(4):1509–18. https://pubmed.ncbi.nlm.nih.gov/33164936/
Guo Z, Lou Y, Kong M, Luo Q, Liu Z, Wu J. A systematic review of phytochemistry, pharmacology and pharmacokinetics on Astragali radix: implications for Astragali radix as a personalized medicine. Int J Mol Sci. 2019;20(6):1463. https://pubmed.ncbi.nlm.nih.gov/30909474/
Liu P, Zhao H, Luo Y. Anti-aging implications of Astragalus membranaceus (Huangqi): a well-known Chinese tonic. Aging Dis. 2017;8(6):868–86. https://pubmed.ncbi.nlm.nih.gov/29344421/
Fauce SR, Jamieson BD, Chin AC, et al. Telomerase-based pharmacologic enhancement of antiviral function of human CD8+ T lymphocytes. J Immunol. 2008;181(10):7400–6. https://pubmed.ncbi.nlm.nih.gov/18981163/
Dow CT, Harley CB. Evaluation of an oral telomerase activator for early age-related macular degeneration – a pilot study. Clin Ophthalmol. 2016;10:243–9. https://pubmed.ncbi.nlm.nih.gov/26869760/
United States of America before the Federal Trade Commission in the matter of Telomerase Activation Sciences, Inc., and Noel Thomas Patton. Docket No. C-4644. FTC.gov. https://www.ftc.gov/system/files/documents/cases/142_3103_-_telomerase_complaint_final.pdf. Updated April 19, 2018. Accessed December10, 2021.; https://www.ftc.gov/system/files/documents/cases/142_3103_-_telomerase_complaint_final.pdf
Tsoukalas D, Fragkiadaki P, Docea AO, et al. Discovery of potent telomerase activators: unfolding new therapeutic and anti-aging perspectives. Mol Med Rep. 2019;20(4):3701–8. https://pubmed.ncbi.nlm.nih.gov/31485647/
Tsoukalas D, Fragkiadaki P, Docea AO, et al. Discovery of potent telomerase activators: unfolding new therapeutic and anti-aging perspectives. Mol Med Rep. 2019;20(4):3701–8. https://pubmed.ncbi.nlm.nih.gov/31485647/
Chandrika UG, Kumara PAASP. Gotu kola (Centella asiatica): nutritional properties and plausible health benefits. Adv Food Nutr Res. 2015;76:125–57. https://pubmed.ncbi.nlm.nih.gov/26602573/
Tsoukalas D, Fragkiadaki P, Docea AO, et al. Discovery of potent telomerase activators: unfolding new therapeutic and anti-aging perspectives. Mol Med Rep. 2019;20(4):3701–8. https://pubmed.ncbi.nlm.nih.gov/31485647/
Puttarak P, Dilokthornsakul P, Saokaew S, et al. Effects of Centella asiatica (L.) Urb. on cognitive function and mood related outcomes: a systematic review and meta-analysis. Sci Rep. 2017;7(1):10646. https://pubmed.ncbi.nlm.nih.gov/28878245/
Larrick JW, Mendelsohn AR. Telomerase redux: ready for prime time? Rejuvenation Res. 2015;18(2):185–7. https://pubmed.ncbi.nlm.nih.gov/25790341/
Shammas MA. Telomeres, lifestyle, cancer, and aging. Curr Opin Clin Nutr Metab Care. 2011;14(1):28–34. https://pubmed.ncbi.nlm.nih.gov/21102320/
Prieto-Oliveira P. Telomerase activation in the treatment of aging or degenerative diseases: a systematic review. Mol Cell Biochem. 2021;476(2):599–607. https://pubmed.ncbi.nlm.nih.gov/33001374/
Artandi SE, Depinho RA. Telomeres and telomerase in cancer. Carcinogenesis. 2010;31(1):9–18. https://pubmed.ncbi.nlm.nih.gov/19887512/
Ornish D, Weidner G, Fair WR, et al. Intensive lifestyle changes may affect the progression of prostate cancer. J Urol. 2005;174(3):1065–70. https://pubmed.ncbi.nlm.nih.gov/16094059/
Skordalakes E. Telomerase and the benefits of healthy living. Lancet Oncol. 2008;9(11):1023–4. https://pubmed.ncbi.nlm.nih.gov/19012852/
Huzen J, Wong LS, van Veldhuisen DJ, et al. Telomere length loss due to smoking and metabolic traits. J Intern Med. 2014;275(2):155–63. https://pubmed.ncbi.nlm.nih.gov/24118582/
García-Calzón S, Moleres A, Martínez-González MA, et al. Dietary total antioxidant capacity is associated with leukocyte telomere length in a children and adolescent population. Clin Nutr. 2015;34(4):694–9. https://pubmed.ncbi.nlm.nih.gov/25131600/
Leung CW, Laraia BA, Needham BL, et al. Soda and cell aging: associations between sugar-sweetened beverage consumption and leukocyte telomere length in healthy adults from the National Health and Nutrition Examination Surveys. Am J Public Health. 2014;104(12):2425–31. https://pubmed.ncbi.nlm.nih.gov/25322305/
Nettleton JA, Diez-Roux A, Jenny NS, Fitzpatrick AL, Jacobs DR. Dietary patterns, food groups, and telomere length in the Multi-Ethnic Study of Atherosclerosis (MESA). Am J Clin Nutr. 2008;88(5):1405–12. https://pubmed.ncbi.nlm.nih.gov/18996878/
Gu Y, Honig LS, Schupf N, et al. Mediterranean diet and leukocyte telomere length in a multi-ethnic elderly population. Age (Dordr). 2015;37(2):9758. https://pubmed.ncbi.nlm.nih.gov/25750063/
Hou L, Savage SA, Blaser MJ, et al. Telomere length in peripheral leukocyte DNA and gastric cancer risk. Cancer Epidemiol Biomarkers Prev. 2009;18(11):3103–9. https://pubmed.ncbi.nlm.nih.gov/19861514/
Gu Y, Honig LS, Schupf N, et al. Mediterranean diet and leukocyte telomere length in a multi-ethnic elderly population. Age (Dordr). 2015;37(2):9758. https://pubmed.ncbi.nlm.nih.gov/25750063/
García-Calzón S, Moleres A, Martínez-González MA, et al. Dietary total antioxidant capacity is associated with leukocyte telomere length in a children and adolescent population. Clin Nutr. 2015;34(4):694–9. https://pubmed.ncbi.nlm.nih.gov/25131600/
Zainabadi K. A brief history of modern aging research. Exp Gerontol. 2018;104:35–42. https://pubmed.ncbi.nlm.nih.gov/29355705/
Zainabadi K. A brief history of modern aging research. Exp Gerontol. 2018;104:35–42. https://pubmed.ncbi.nlm.nih.gov/29355705/
Strong R, Miller RA, Antebi A, et al. Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an a-glucosidase inhibitor or a Nrf2-inducer. Aging Cell. 2016;15(5):872–84. https://pubmed.ncbi.nlm.nih.gov/27312235/
Gebreslassie M, Sampaio F, Nystrand C, Ssegonja R, Feldman I. Economic evaluations of public health interventions for physical activity and healthy diet: a systematic review. Prev Med. 2020;136:106100. https://pubmed.ncbi.nlm.nih.gov/32353572/
Lozano R, Naghavi M, Foreman K, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2095–128. https://pubmed.ncbi.nlm.nih.gov/23245604/
Mokdad AH, Ballestros K, Echko M, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Afshin A, Sur PJ, Fay KA, et al. Health effects of dietary risks in 195 countries, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2019;393(10184):1958–72. https://pubmed.ncbi.nlm.nih.gov/30954305/
Gebreslassie M, Sampaio F, Nystrand C, Ssegonja R, Feldman I. Economic evaluations of public health interventions for physical activity and healthy diet: a systematic review. Prev Med. 2020;136:106100. https://pubmed.ncbi.nlm.nih.gov/32353572/
Das P, Samarasekera U. The story of GBD 2010: a “super-human” effort. Lancet. 2012;380(9859):2067–70. https://pubmed.ncbi.nlm.nih.gov/23259158/
Mokdad AH, Ballestros K, Echko M, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Dato S, Bellizzi D, Rose G, Passarino G. The impact of nutrients on the aging rate: a complex interaction of demographic, environmental and genetic factors. Mech Ageing Dev. 2016;154:49–61. https://pubmed.ncbi.nlm.nih.gov/26876763/
Campisi J, Kapahi P, Lithgow GJ, Melov S, Newman JC, Verdin E. From discoveries in ageing research to therapeutics for healthy ageing. Nature. 2019;571(7764):183–92. https://pubmed.ncbi.nlm.nih.gov/31292558/
Govindaraju T, Sahle BW, McCaffrey TA, McNeil JJ, Owen AJ. Dietary patterns and quality of life in older adults: a systematic review. Nutrients. 2018;10(8):971. https://pubmed.ncbi.nlm.nih.gov/30050006/
Milte CM, McNaughton SA. Dietary patterns and successful ageing: a systematic review. Eur J Nutr. 2016;55(2):423–50. https://pubmed.ncbi.nlm.nih.gov/26695408/
Reedy J, Krebs-Smith SM, Miller PE, et al. Higher diet quality is associated with decreased risk of all-cause, cardiovascular disease, and cancer mortality among older adults. J Nutr. 2014;144(6):881–9. https://pubmed.ncbi.nlm.nih.gov/24572039/
McCullough ML. Diet patterns and mortality: common threads and consistent results. J Nutr. 2014;144(6):795–6. https://pubmed.ncbi.nlm.nih.gov/24717365/
Reedy J, Krebs-Smith SM, Miller PE, et al. Higher diet quality is associated with decreased risk of all-cause, cardiovascular disease, and cancer mortality among older adults. J Nutr. 2014;144(6):881–9. https://pubmed.ncbi.nlm.nih.gov/24572039/
Afshin A, Sur PJ, Fay KA, et al. Health effects of dietary risks in 195 countries, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2019;393(10184):1958–72. https://pubmed.ncbi.nlm.nih.gov/30954305/
Yip CSC, Chan W, Fielding R. The associations of fruit and vegetable intakes with burden of diseases: a systematic review of meta-analyses. J Acad Nutr Diet. 2019;119(3):464–81. https://pubmed.ncbi.nlm.nih.gov/30639206/
Fisher D. Study finds no link between secondhand smoke and cancer. Forbes. https://www.forbes.com/sites/danielfisher/2013/12/12/study-finds-no-link-between-secondhand-smoke-and-cancer/?sh=77c79a2565d4. Published December 12, 2013. Accessed December 12, 2021.; https://www.forbes.com/sites/danielfisher/2013/12/12/study-finds-no-link-between-secondhand-smoke-and-cancer/?sh=77c79a2565d4
Hackshaw AK, Law MR, Wald NJ. The accumulated evidence on lung cancer and environmental tobacco smoke. BMJ. 1997;315(7114):980–8. https://pubmed.ncbi.nlm.nih.gov/9365295/
Gori GB, Mantel N. Mainstream and environmental tobacco smoke. Regul Toxicol Pharmacol. 1991;14(1):88–105. https://pubmed.ncbi.nlm.nih.gov/1947248/
Barnes DE, Bero LA. Why review articles on the health effects of passive smoking reach different conclusions. JAMA. 1998;279(19):1566–70. https://pubmed.ncbi.nlm.nih.gov/9605902/
Drope J, Chapman S. Tobacco industry efforts at discrediting scientific knowledge of environmental tobacco smoke: a review of internal industry documents. J Epidemiol Community Health. 2001;55(8):588–94. https://pubmed.ncbi.nlm.nih.gov/11449018/
Barnes DE, Bero LA. Why review articles on the health effects of passive smoking reach different conclusions. JAMA. 1998;279(19):1566–70. https://pubmed.ncbi.nlm.nih.gov/9605902/
Fardet A, Boirie Y. Associations between food and beverage groups and major diet-related chronic diseases: an exhaustive review of pooled/meta-analyses and systematic reviews. Nutr Rev. 2014;72(12):741–62. https://pubmed.ncbi.nlm.nih.gov/25406801/
Fardet A, Boirie Y. Associations between food and beverage groups and major diet-related chronic diseases: an exhaustive review of pooled/meta-analyses and systematic reviews. Nutr Rev. 2014;72(12):741–62. https://pubmed.ncbi.nlm.nih.gov/25406801/
Abdelhamid AS, Brown TJ, Brainard JS, et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev. 2018;7:CD003177. https://pubmed.ncbi.nlm.nih.gov/30019766/
Gonzales JF, Barnard ND, Jenkins DJA, et al. Applying the precautionary principle to nutrition and cancer. J Am Coll Nutr. 2014;33(3):239–46. https://pubmed.ncbi.nlm.nih.gov/24870117/
Lane KE, Wilson M, Hellon TG, Davies IG. Bioavailability and conversion of plant based sources of omega-3 fatty acids – a scoping review to update supplementation options for vegetarians and vegans. Crit Rev Food Sci Nutr. 2022;62(18):4982–97. https://pubmed.ncbi.nlm.nih.gov/33576691/
Fardet A, Boirie Y. Associations between food and beverage groups and major diet-related chronic diseases: an exhaustive review of pooled/meta-analyses and systematic reviews. Nutr Rev. 2014;72(12):741–62. https://pubmed.ncbi.nlm.nih.gov/25406801/
Yip CSC, Lam W, Fielding R. A summary of meat intakes and health burdens. Eur J Clin Nutr. 2018;72(1):18–29. https://pubmed.ncbi.nlm.nih.gov/28792013/
Spiegelhalter D. Microlives. Understanding Uncertainty. http://understandinguncertainty.org/microlives. Published November 22, 2011. Accessed August 30, 2021.; https://understandinguncertainty.org/microlives
Spiegelhalter D. Using speed of ageing and “microlives” to communicate the effects of lifetime habits and environment. BMJ. 2012;345:e8223. https://pubmed.ncbi.nlm.nih.gov/23247978/
Spiegelhalter D. Using speed of ageing and “microlives” to communicate the effects of lifetime habits and environment. BMJ. 2012;345:e8223. https://pubmed.ncbi.nlm.nih.gov/23247978/
Zhuang P, Wu F, Mao L, et al. Egg and cholesterol consumption and mortality from cardiovascular and different causes in the United States: a population-based cohort study. PLoS Med. 2021;18(2):e1003508. https://pubmed.ncbi.nlm.nih.gov/33561122/
Zeraatkar D, Han MA, Guyatt GH, et al. Red and processed meat consumption and risk for all-cause mortality and cardiometabolic outcomes: a systematic review and meta-analysis of cohort studies. Ann Intern Med. 2019;171(10):703–10. https://pubmed.ncbi.nlm.nih.gov/31569213/
Heard CL, Rakow T, Spiegelhalter D. Comparing comprehension and perception for alternative speed-of-ageing and standard hazard ratio formats. Appl Cognit Psychol. 2018;32(1):81–93. https://onlinelibrary.wiley.com/doi/abs/10.1002/acp.3381
Heard CL, Rakow T, Spiegelhalter D. Comparing comprehension and perception for alternative speed-of-ageing and standard hazard ratio formats. Appl Cognit Psychol. 2018;32(1):81–93. https://onlinelibrary.wiley.com/doi/abs/10.1002/acp.3381
IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans; Volume 114: Red Meat and Processed Meat. IARC Press; 2018. https://monographs.iarc.who.int/wp-content/uploads/2018/06/mono114.pdf. Accessed December 19 https://monographs.iarc.who.int/wp-content/uploads/2018/06/mono114.pdf
Chaffetz J. Letter on behalf of the U.S. House of Representatives Committee on Oversight and Government Reform of the 114th Congress to Francis S. Collins, M.D., Ph.D., Director, National Institutes of Health. September 26, 2016.; https://oversight.house.gov/wp-content/uploads/2016/09/2016-09-26-JEC-to-Collins-NIH-IARC-Funding-due-10-10.pdf
Boobis AR, Cohen SM, Dellarco VL, et al. Classification schemes for carcinogenicity based on hazard-identification have become outmoded and serve neither science nor society. Regul Toxicol Pharmacol. 2016;82:158–66. https://pubmed.ncbi.nlm.nih.gov/27780763/
Wild CP. Letter to Dr. Francis S. Collins re: IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. October 5, 2016. https://monographs.iarc.who.int/ENG/News/LetterFromDrWild-to-DrCollins.pdf. Accessed December 19, 2021.; https://monographs.iarc.who.int/wp-content/uploads/2018/06/LetterFromDrWild-to-DrCollins.pdf
International Agency for Research on Cancer. World Health Organization. Q&A on the carcinogenicity of the consumption of red meat and processed meat. 2015. https://www.iarc.who.int/wp-content/uploads/2018/11/Monographs-QA_Vol114.pdf. Accessed December 28, 2022.; https://www.iarc.who.int/wp-content/uploads/2018/11/Monographs-QA_Vol114.pdf
IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans; Volume 114: Red Meat and Processed Meat. IARC Press; 2018. https://monographs.iarc.who.int/wp-content/uploads/2018/06/mono114.pdf. Accessed December 19 https://monographs.iarc.who.int/wp-content/uploads/2018/06/mono114.pdf
Office on Smoking and Health (US). The Health Consequences of Involuntary Exposure to Tobacco Smoke: A Report of the Surgeon General. Centers for Disease Control and Prevention (US); 2006. https://pubmed.ncbi.nlm.nih.gov/20669524/
Modica C, Lewis JH, Bay C. Colorectal cancer: applying the value transformation framework to increase the percent of patients receiving screening in federally qualified health centers. Prev Med Rep. 2019;15:100894. https://pubmed.ncbi.nlm.nih.gov/31198660/
Kim H, Caulfield LE, Rebholz CM. Healthy plant-based diets are associated with lower risk of all-cause mortality in US adults. J Nutr. 2018;148(4):624–31. https://pubmed.ncbi.nlm.nih.gov/29659968/
Bamia C, Trichopoulos D, Ferrari P, et al. Dietary patterns and survival of older Europeans: the EPIC – Elderly Study (European Prospective Investigation into Cancer and Nutrition). Public Health Nutr. 2007;10(6):590–8. https://pubmed.ncbi.nlm.nih.gov/17381929/
Kahleova H, Levin S, Barnard ND. Plant-based diets for healthy aging. J Am Coll Nutr. 2021;40(5):478–9. https://pubmed.ncbi.nlm.nih.gov/32643581/
Ekmekcioglu C. Nutrition and longevity – from mechanisms to uncertainties. Crit Rev Food Sci Nutr. 2020;60(18):3063–82. https://pubmed.ncbi.nlm.nih.gov/31631676/
Everitt AV, Hilmer SN, Brand-Miller JC, et al. Dietary approaches that delay age-related diseases. Clin Interv Aging. 2006;1(1):11–31. https://pubmed.ncbi.nlm.nih.gov/18047254/
Kahleova H, Levin S, Barnard ND. Plant-based diets for healthy aging. J Am Coll Nutr. 2021;40(5):478–9. https://pubmed.ncbi.nlm.nih.gov/32643581/
O’Hara JK. The $11 trillion reward: how simple dietary changes can save lives and money, and how we get there. UCSusa.org. https://www.ucsusa.org/sites/default/files/2019–09/11-trillion-reward.pdf. Published August 2013. Accessed December 15, 2021.; https://www.ucsusa.org/sites/default/files/2019-09/11-trillion-reward.pdf
Cross AJ, Pollock JRA, Bingham SA. Haem, not protein or inorganic iron, is responsible for endogenous intestinal N-nitrosation arising from red meat. Cancer Res. 2003;63(10):2358–60. https://pubmed.ncbi.nlm.nih.gov/12750250/
Tucker KL, Hallfrisch J, Qiao N, Muller D, Andres R, Fleg JL. The combination of high fruit and vegetable and low saturated fat intakes is more protective against mortality in aging men than is either alone: the Baltimore Longitudinal Study of Aging. J Nutr. 2005;135(3):556–61. https://pubmed.ncbi.nlm.nih.gov/15735093/
Jenkins DJ, Kendall CW. The Garden of Eden: plant-based diets, the genetic drive to store fat and conserve cholesterol, and implications for epidemiology in the 21st century. Epidemiology. 2006;17(2):128–30. https://pubmed.ncbi.nlm.nih.gov/16477249/
Eaton SB, Konner M. Paleolithic nutrition. A consideration of its nature and current implications. N Engl J Med. 1985;312(5):283–9. https://pubmed.ncbi.nlm.nih.gov/2981409/
Anderson JW, Konz EC, Jenkins DJ. Health advantages and disadvantages of weight-reducing diets: a computer analysis and critical review. J Am Coll Nutr. 2000;19(5):578–90. https://pubmed.ncbi.nlm.nih.gov/11022871/
Hladik CM, Pasquet P. The human adaptations to meat eating: a reappraisal. Hum Evol. 2002;17(3–4):199–206. https://link.springer.com/article/10.1007/BF02436371
Milton K. Micronutrient intakes of wild primates: are humans different? Comp Biochem Physiol A Mol Integr Physiol. 2003;136(1):47–59. https://pubmed.ncbi.nlm.nih.gov/14527629/
Jenkins DJA, Kendall CWC, Marchie A, et al. The Garden of Eden – plant based diets, the genetic drive to conserve cholesterol and its implications for heart disease in the 21st century. Comp Biochem Physiol A Mol Integr Physiol. 2003;136(1):141–51. https://pubmed.ncbi.nlm.nih.gov/14527636/
Larsen SC, Ängquist L, Sørensen TI, Heitmann BL. 24h urinary sodium excretion and subsequent change in weight, waist circumference and body composition. PLoS ONE. 2013;8(7):e69689. https://pubmed.ncbi.nlm.nih.gov/23936079/
Roberts WC. High salt intake, its origins, its economic impact, and its effect on blood pressure. Am J Cardiol. 2001;88(11):1338–46. https://pubmed.ncbi.nlm.nih.gov/11728372/
Yin X, Tian M, Neal B. Sodium reduction: how big might the risks and benefits be? Heart Lung Circ. 2021;30(2):180–5. https://pubmed.ncbi.nlm.nih.gov/32855069/
Afshin A, Sur PJ, Fay KA, et al. Health effects of dietary risks in 195 countries, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2019;393(10184):1958–72. https://pubmed.ncbi.nlm.nih.gov/30954305/
MacGregor GA, Markandu ND, Best FE, et al. Double-blind randomised crossover trial of moderate sodium restriction in essential hypertension. Lancet. 1982;1(8268):351–5. https://pubmed.ncbi.nlm.nih.gov/6120346/
Rudelt A, French S, Harnack L. Fourteen-year trends in sodium content of menu offerings at eight leading fast-food restaurants in the USA. Public Nutr. 2014;17(8):1682–8. https://pubmed.ncbi.nlm.nih.gov/24018166/
Suckling RJ, He FJ, Markandu ND, MacGregor GA. Dietary salt influences postprandial plasma sodium concentration and systolic blood pressure. Kidney Int. 2012;81(4):407–11. https://pubmed.ncbi.nlm.nih.gov/22048126/
Chobufo MD, Gayam V, Soluny J, et al. Prevalence and control rates of hypertension in the USA: 2017–2018. Int J Cardiol Hypertens. 2020;6:100044. https://pubmed.ncbi.nlm.nih.gov/33447770/
Celermajer DS, Neal B. Excessive sodium intake and cardiovascular disease: a-salting our vessels. J Am Coll Cardiol. 2013;61(3):344–5. https://pubmed.ncbi.nlm.nih.gov/23141488/
Mancilha-Carvalho J de J, de Souza e Silva NA. The Yanomami Indians in the INTERSALT Study. Arq Bras Cardiol. 2003;80(3):289–300. https://pubmed.ncbi.nlm.nih.gov/12856272/
Roberts WC. High salt intake, its origins, its economic impact, and its effect on blood pressure. Am J Cardiol. 2001;88(11):1338–46. https://pubmed.ncbi.nlm.nih.gov/11728372/
Cappuccio FP, Capewell S, Lincoln P, McPherson K. Policy options to reduce population salt intake. BMJ. 2011;343:d4995. https://pubmed.ncbi.nlm.nih.gov/21835876/
Toldrá F, Barat JM. Strategies for salt reduction in foods. Recent Pat Food Nutr Agric. 2012;4(1):19–25. https://pubmed.ncbi.nlm.nih.gov/22316270/
Appel LJ, Anderson CA. Compelling evidence for public health action to reduce salt intake. N Engl J Med. 2010;362(7):650–2. https://pubmed.ncbi.nlm.nih.gov/20089959/
Drewnowski A, Rehm CD. Sodium intakes of US children and adults from foods and beverages by location of origin and by specific food source. Nutrients. 2013;5(6):1840–55. https://pubmed.ncbi.nlm.nih.gov/23760055/
.;
Select Committee on Nutrition and Human Needs. Dietary Goals for the United States – Supplemental Views. U.S. Government Printing Office; 1977. https://naldc.nal.usda.gov/catalog/1759572
Foscolou A, Critselis E, Tyrovolas S, et al. The association of sodium intake with successful aging, in 3,349 middle-aged and older adults: results from the ATTICA and MEDIS cross-sectional epidemiological studies. Nutr Healthy Aging. 2020;5(4):287–96. https://content.iospress.com/articles/nutrition-and-healthy-aging/nha190080
Madiloggovit J, Chotechuang N, Trachootham D. Impact of self-tongue brushing on taste perception in Thai older adults: a pilot study. Geriatr Nurs. 2016;37(2):128–36. https://pubmed.ncbi.nlm.nih.gov/26747405/
Quirynen M, Avontroodt P, Soers C, Zhao H, Pauwels M, van Steenberghe D. Impact of tongue cleansers on microbial load and taste. J Clin Periodontol. 2004;31(7):506–10. https://pubmed.ncbi.nlm.nih.gov/15191584/
Madiloggovit J, Chotechuang N, Trachootham D. Impact of self-tongue brushing on taste perception in Thai older adults: a pilot study. Geriatr Nurs. 2016;37(2):128–36. https://pubmed.ncbi.nlm.nih.gov/26747405/
Sigurdsson EL. Salt: a taste of death? Scand J Prim Health Care. 2014;32(2):53–4. https://pubmed.ncbi.nlm.nih.gov/24939739/
Maleki A, Soltanian AR, Zeraati F, Sheikh V, Poorolajal J. The flavor and acceptability of six different potassium-enriched (sodium reduced) iodized salts: a single-blind, randomized, crossover design. Clin Hypertens. 2016;22(1):18. https://pubmed.ncbi.nlm.nih.gov/28031983/
Whelton PK, Appel LJ, Sacco RL, et al. Sodium, blood pressure, and cardiovascular disease: further evidence supporting the American Heart Association sodium reduction recommendations. Circulation. 2012;126(24):2880–9. https://pubmed.ncbi.nlm.nih.gov/23124030/
Cogswell ME, Zhang Z, Carriquiry AL, et al. Sodium and potassium intakes among US adults: NHANES 2003–2008. Am J Clin Nutr. 2012;96(3):647–57. https://pubmed.ncbi.nlm.nih.gov/22854410/
Sebastian A, Cordain L, Frassetto L, Banerjee T, Morris RC. Postulating the major environmental condition resulting in the expression of essential hypertension and its associated cardiovascular diseases: dietary imprudence in daily selection of foods in respect of their potassium and sodium content resulting in oxidative stress-induced dysfunction of the vascular endothelium, vascular smooth muscle, and perivascular tissues. Med Hypotheses. 2018;119:110–9. https://pubmed.ncbi.nlm.nih.gov/30122481/
Palmer BF, Clegg DJ. Achieving the benefits of a high-potassium, paleolithic diet, without the toxicity. Mayo Clin Proc. 2016;91(4):496–508. https://pubmed.ncbi.nlm.nih.gov/26948054/
Jew S, AbuMweis SS, Jones PJH. Evolution of the human diet: linking our ancestral diet to modern functional foods as a means of chronic disease prevention. J Med Food. 2009;12(5):925–34. https://pubmed.ncbi.nlm.nih.gov/19857053/
Drewnowski A, Maillot M, Rehm C. Reducing the sodium-potassium ratio in the US diet: a challenge for public health. Am J Clin Nutr. 2012;96(2):439–44. https://pubmed.ncbi.nlm.nih.gov/22760562/
van Buren L, Dötsch-Klerk M, Seewi G, Newson RS. Dietary impact of adding potassium chloride to foods as a sodium reduction technique. Nutrients. 2016;8(4):235. https://pubmed.ncbi.nlm.nih.gov/27110818/
Jafarnejad S, Mirzaei H, Clark CCT, Taghizadeh M, Ebrahimzadeh A. The hypotensive effect of salt substitutes in stage 2 hypertension: a systematic review and meta-analysis. BMC Cardiovasc Disord. 2020;20(1):98. https://pubmed.ncbi.nlm.nih.gov/32106813/
Chang HY, Hu YW, Yue CSJ, et al. Effect of potassium-enriched salt on cardiovascular mortality and medical expenses of elderly men. Am J Clin Nutr. 2006;83(6):1289–96. https://pubmed.ncbi.nlm.nih.gov/16762939/
Lambert K, Conley M, Dumont R, et al. Letter to the editor on “Potential use of salt substitutes to reduce blood pressure.” J Clin Hypertens. 2019;21(10):1609–10. https://pubmed.ncbi.nlm.nih.gov/31448881/
Farrand C, MacGregor G, Campbell NRC, Webster J. Potential use of salt substitutes to reduce blood pressure. J Clin Hypertens. 2019;21(3):350–4. https://pubmed.ncbi.nlm.nih.gov/30690859/
Greer RC, Marklund M, Anderson CAM, et al. Potassium-enriched salt substitutes as a means to lower blood pressure: benefits and risks. Hypertension. 2020;75(2):266–74. https://pubmed.ncbi.nlm.nih.gov/31838902/
Greer RC, Marklund M, Anderson CAM, et al. Potassium-enriched salt substitutes as a means to lower blood pressure: benefits and risks. Hypertension. 2020;75(2):266–74. https://pubmed.ncbi.nlm.nih.gov/31838902/
Greer RC, Marklund M, Anderson CAM, et al. Potassium-enriched salt substitutes as a means to lower blood pressure: benefits and risks. Hypertension. 2020;75(2):266–74. https://pubmed.ncbi.nlm.nih.gov/31838902/
Mokdad AH, Ballestros K, Echko M, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Mokdad AH, Ballestros K, Echko M, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Reedy J, Krebs-Smith SM, Miller PE, et al. Higher diet quality is associated with decreased risk of all-cause, cardiovascular disease, and cancer mortality among older adults. J Nutr. 2014;144(6):881–9. https://pubmed.ncbi.nlm.nih.gov/24572039/
Devries S, Willett W, Bonow RO. Nutrition education in medical school, residency training, and practice. JAMA. 2019;321(14):1351–2. https://pubmed.ncbi.nlm.nih.gov/30896728/
Freeman KJ, Grega ML, Friedman SM, et al. Lifestyle medicine reimbursement: a proposal for policy priorities informed by a cross-sectional survey of lifestyle medicine practitioners. Int J Environ Res Public Health. 2021;18(21):11632. https://pubmed.ncbi.nlm.nih.gov/34770148/
Brody H. Pharmaceutical industry financial support for medical education: benefit, or undue influence? J Law Med Ethics. 2009;37(3):451–60. https://pubmed.ncbi.nlm.nih.gov/19723256/
Proctor RN. The history of the discovery of the cigarette – lung cancer link: evidentiary traditions, corporate denial, global toll. Tob Control. 2012;21(2):87–91. https://pubmed.ncbi.nlm.nih.gov/22345227/
Office on Smoking and Health, National Center for Chronic Disease Prevention and Health Promotion, CDC. Tobacco use – United States, 1900–1999. JAMA. 1999;282(23):2202–4. https://pubmed.ncbi.nlm.nih.gov/10605963/
Editorial. The advertising of cigarettes. JAMA. 1948;138(9):652–3. https://jamanetwork.com/journals/jama/article-abstract/302011
Editorial. The advertising of cigarettes. JAMA. 1948;138(9):652–3. https://jamanetwork.com/journals/jama/article-abstract/302011
Proctor RN. The history of the discovery of the cigarette – lung cancer link: evidentiary traditions, corporate denial, global toll. Tob Control. 2012;21(2):87–91. https://pubmed.ncbi.nlm.nih.gov/22345227/
Gugiu PC, Gugiu MR. Levels of evidence: a reply to Berger and Knoll. Eval Health Prof. 2011;34(1):127–30. https://journals.sagepub.com/doi/10.1177/0163278710391467
Chopra M, Darnton-Hill I. Tobacco and obesity epidemics: not so different after all? BMJ. 2004;328(7455):1558–60. https://pubmed.ncbi.nlm.nih.gov/15217877/
Industries. OpenSecrets.org. https://www.opensecrets.org/federal-lobbying/industries. Published July 23, 2021. Accessed August 31, 2021.; https://www.opensecrets.org/federal-lobbying/industries
Maplight, Feed the Truth. Draining the Big Food swamp. FeedtheTruth.org. https://feedthetruth.org/wp-content/uploads/2021/08/FTT-DrainingTheSwamp-ExecSummary-FINAL.pdf. Published February 25, 2021. Accessed January 6, 2022.; https://www.readkong.com/page/draining-the-big-food-feed-the-truth-5969302
Ищи, кому выгодно (лат.). – Примеч. ред.
Sarna L, Bialous SA, Nandy K, Antonio ALM, Yang Q. Changes in smoking prevalences among health care professionals from 2003 to 2010–2011. JAMA. 2014;311(2):197–9. https://pubmed.ncbi.nlm.nih.gov/24399560/
Jindeel A. Health care providers who smoke. Am J Nurs. 2010;110(6):11. https://pubmed.ncbi.nlm.nih.gov/20505442/
Aggarwal M, Singh Ospina N, Kazory A, et al. The mismatch of nutrition and lifestyle beliefs and actions among physicians: a wake-up call. Am J Lifestyle Med. 2020;14(3):304–15. https://pubmed.ncbi.nlm.nih.gov/32477033/
Bertozzi B, Tosti V, Fontana L. Beyond calories: an integrated approach to promote health, longevity and well-being. Gerontology. 2017;63(1):13–9. https://pubmed.ncbi.nlm.nih.gov/27173125/
Fadnes LT, Økland JM, Haaland ØA, Johansson KA. Estimating impact of food choices on life expectancy: a modeling study. PLoS Med. 2022;19(2):e1003889. https://pubmed.ncbi.nlm.nih.gov/35134067/
Hooper L, Bunn D, Jimoh FO, Fairweather-Tait SJ. Water-loss dehydration and aging. Mech Ageing Dev. 2014;136–7:50–8. https://pubmed.ncbi.nlm.nih.gov/24333321/
Kenney WL, Chiu P. Influence of age on thirst and fluid intake. Med Sci Sports Exerc. 2001;33(9):1524–32. https://pubmed.ncbi.nlm.nih.gov/11528342/
Lorenzo I, Serra-Prat M, Yébenes JC. The role of water homeostasis in muscle function and frailty: a review. Nutrients. 2019;11(8):E1857. https://pubmed.ncbi.nlm.nih.gov/31405072/
Hooper L, Bunn D, Jimoh FO, Fairweather-Tait SJ. Water-loss dehydration and aging. Mech Ageing Dev. 2014;136–7:50–8. https://pubmed.ncbi.nlm.nih.gov/24333321/
Popkin BM, Armstrong LE, Bray GM, Caballero B, Frei B, Willett WC. A new proposed guidance system for beverage consumption in the United States. Am J Clin Nutr. 2006;83(3):529–42. https://pubmed.ncbi.nlm.nih.gov/16522898/
Walsh NP, Fortes MB, Purslow C, Esmaeelpour M. Author response: is whole body hydration an important consideration in dry eye? Invest Ophthalmol Vis Sci. 2013;54(3):1713–4. https://pubmed.ncbi.nlm.nih.gov/23471906/
Chan J, Knutsen SF, Blix GG, Lee JW, Fraser GE. Water, other fluids, and fatal coronary heart disease: the Adventist Health Study. Am J Epidemiol. 2002;155(9):827–33. https://pubmed.ncbi.nlm.nih.gov/11978586/
Cui R, Iso H, Eshak ES, Maruyama K, Tamakoshi A, JACC Study Group. Water intake from foods and beverages and risk of mortality from CVD: the Japan Collaborative Cohort (JACC) Study. Public Health Nutr. 2018;21(16):3011–7. https://pubmed.ncbi.nlm.nih.gov/30107863/
Stookey JD, Kavouras S¿, Suh H, Lang F. Underhydration is associated with obesity, chronic diseases, and death within 3 to 6 years in the U.S. population aged 51–70 years. Nutrients. 2020;12(4):E905. https://pubmed.ncbi.nlm.nih.gov/32224908/
Lim WH, Wong G, Lewis JR, et al. Total volume and composition of fluid intake and mortality in older women: a cohort study. BMJ Open. 2017;7(3):e011720. https://pubmed.ncbi.nlm.nih.gov/28341683/
Kant AK, Graubard BI. A prospective study of water intake and subsequent risk of all-cause mortality in a national cohort. Am J Clin Nutr. 2017;105(1):212–20. https://pubmed.ncbi.nlm.nih.gov/27903521/
Leurs LJ, Schouten LJ, Goldbohm RA, van den Brandt PA. Total fluid and specific beverage intake and mortality due to IHD and stroke in the Netherlands Cohort Study. Br J Nutr. 2010;104(8):1212–21. https://pubmed.ncbi.nlm.nih.gov/20456812/
Loomba RS, Aggarwal S, Arora RR. Raw water consumption does not affect all-cause or cardiovascular mortality: a secondary analysis. Am J Ther. 2016;23(6):e1287–92. https://pubmed.ncbi.nlm.nih.gov/25611360/
Hooper L, Bunn D, Jimoh FO, Fairweather-Tait SJ. Water-loss dehydration and aging. Mech Ageing Dev. 2014;136–7:50–8. https://pubmed.ncbi.nlm.nih.gov/24333321/
Masot O, Miranda J, Santamaría AL, Paraiso Pueyo E, Pascual A, Botigué T. Fluid intake recommendation considering the physiological adaptations of adults over 65 years: a critical review. Nutrients. 2020;12(11):E3383. https://pubmed.ncbi.nlm.nih.gov/33158071/
McKenzie AL, Muñoz CX, Armstrong LE. Accuracy of urine color to detect equal to or greater than 2 % body mass loss in men. J Athl Train. 2015;50(12):1306–9. https://pubmed.ncbi.nlm.nih.gov/26642041/
McKenzie AL, Armstrong LE. Monitoring body water balance in pregnant and nursing women: the validity of urine color. Ann Nutr Metab. 2017;70 Suppl 1:18–22. https://pubmed.ncbi.nlm.nih.gov/28614809/
Perrier ET, Johnson EC, McKenzie AL, Ellis LA, Armstrong LE. Urine colour change as an indicator of change in daily water intake: a quantitative analysis. Eur J Nutr. 2016;55(5):1943–9. https://pubmed.ncbi.nlm.nih.gov/26286348/
Kostelnik SB, Davy KP, Hedrick VE, Thomas DT, Davy BM. The validity of urine color as a hydration biomarker within the general adult population and athletes: a systematic review. J Am Coll Nutr. 2021;40(2):172–9. https://pubmed.ncbi.nlm.nih.gov/32330109/
Hooper L, Abdelhamid A, Attreed NJ, et al. Clinical symptoms, signs and tests for identification of impending and current water-loss dehydration in older people. Cochrane Database Syst Rev. 2015;(4):CD009647. https://pubmed.ncbi.nlm.nih.gov/25924806/
Benelam B, Wyness L. Hydration and health: a review. Nutr Bull. 2010;35:3–25. https://onlinelibrary.wiley.com/doi/full/10.1111/j.1467–3010.2009.01795.x
Vivanti AP. Origins for the estimations of water requirements in adults. Eur J Clin Nutr. 2012;66(12):1282–9. https://pubmed.ncbi.nlm.nih.gov/23093341/
Benelam B, Wyness L. Hydration and health: a review. Nutr Bull. 2010;35:3–25. https://onlinelibrary.wiley.com/doi/full/10.1111/j.1467–3010.2009.01795.x
Masot O, Miranda J, Santamaría AL, Paraiso Pueyo E, Pascual A, Botigué T. Fluid intake recommendation considering the physiological adaptations of adults over 65 years: a critical review. Nutrients. 2020;12(11):E3383. https://pubmed.ncbi.nlm.nih.gov/33158071/
Hoffman MD, Bross TL, Hamilton RT. Are we being drowned by overhydration advice on the Internet? Phys Sportsmed. 2016;44(4):343–8. https://pubmed.ncbi.nlm.nih.gov/27548748/
Onufrak SJ, Park S, Sharkey JR, Sherry B. The relationship of perceptions of tap water safety with intake of sugar-sweetened beverages and plain water among US adults. Public Health Nutr. 2014;17(1):179–85. https://pubmed.ncbi.nlm.nih.gov/23098620/
Saleh MA, Abdel-Rahman FH, Woodard BB, et al. Chemical, microbial and physical evaluation of commercial bottled waters in greater Houston area of Texas. J Environ Sci Health A Tox Hazard Subst Environ Eng. 2008;43(4):335–47. https://pubmed.ncbi.nlm.nih.gov/18273738/
Fardet A, Boirie Y. Associations between food and beverage groups and major diet-related chronic diseases: an exhaustive review of pooled/meta-analyses and systematic reviews. Nutr Rev. 2014;72(12):741–62. https://pubmed.ncbi.nlm.nih.gov/25406801/
. Øverby NC, Lillegaard ITL, Johansson L, Andersen LF. High intake of added sugar among Norwegian children and adolescents. Public Health Nutr. 2004;7(2):285–93. https://pubmed.ncbi.nlm.nih.gov/15003136/
Chikritzhs T, Stockwell T, Naimi T, Andreasson S, Dangardt F, Liang W. Has the leaning tower of presumed health benefits from ‘moderate’ alcohol use finally collapsed? Addiction. 2015;110(5):726–7. https://pubmed.ncbi.nlm.nih.gov/25613200/
Fillmore KM, Stockwell T, Chikritzhs T, Bostrom A, Kerr W. Moderate alcohol use and reduced mortality risk: systematic error in prospective studies and new hypotheses. Ann Epidemiol. 2007;17(5 Suppl):S16–23. https://pubmed.ncbi.nlm.nih.gov/17478320/
Johnson T, Gerson L, Hershcovici T, Stave C, Fass R. Systematic review: the effects of carbonated beverages on gastro-oesophageal reflux disease. Aliment Pharmacol Ther. 2010;31(6):607–14. https://pubmed.ncbi.nlm.nih.gov/20055784/
Lesser LI, Ebbeling CB, Goozner M, Wypij D, Ludwig DS. Relationship between funding source and conclusion among nutrition-related scientific articles. PLoS Med. 2007;4(1):e5. https://pubmed.ncbi.nlm.nih.gov/17214504/
Quik M. Smoking, nicotine and Parkinson’s disease. Trends Neurosci. 2004;27(9):561–8. https://pubmed.ncbi.nlm.nih.gov/15331239/
Searles Nielsen S, Gallagher LG, Lundin JI, et al. Environmental tobacco smoke and Parkinson’s disease. Mov Disord. 2012;27(2):293–6. https://pubmed.ncbi.nlm.nih.gov/22095755/
U.S. Department of Health and Human Services. The Health Consequences of Smoking—50 Years of Progress: A Report of the Surgeon General. Centers for Disease Control and Prevention; 2014. https://www.cdc.gov/tobacco/sgr/50th-anniversary/index.htm#complete-report
Nielsen SS, Franklin GM, Longstreth WT, Swanson PD, Checkoway H. Nicotine from edible Solanaceae and risk of Parkinson disease. Ann Neurol. 2013;74(3):472–7. https://pubmed.ncbi.nlm.nih.gov/23661325/
Aune D, Rosenblatt DAN, Chan DSM, et al. Dairy products, calcium, and prostate cancer risk: a systematic review and meta-analysis of cohort studies. Am J Clin Nutr. 2015;101(1):87–117. https://pubmed.ncbi.nlm.nih.gov/25527754/
Vasconcelos A, Santos T, Ravasco P, Neves PM. Dairy products: is there an impact on promotion of prostate cancer? A review of the literature. Front Nutr. 2019;6:62. https://pubmed.ncbi.nlm.nih.gov/31139629/
Aune D, Lau R, Chan DSM, et al. Dairy products and colorectal cancer risk: a systematic review and meta-analysis of cohort studies. Ann Oncol. 2012;23(1):37–45. https://pubmed.ncbi.nlm.nih.gov/21617020/
Veettil SK, Ching SM, Lim KG, Saokaew S, Phisalprapa P, Chaiyakunapruk N. Effects of calcium on the incidence of recurrent colorectal adenomas: a systematic review with meta-analysis and trial sequential analysis of randomized controlled trials. Medicine. 2017;96(32):e7661. https://pubmed.ncbi.nlm.nih.gov/28796047/
Gonzales JF, Barnard ND, Jenkins DJA, et al. Applying the precautionary principle to nutrition and cancer. J Am Coll Nutr. 2014;33(3):239–46. https://pubmed.ncbi.nlm.nih.gov/24870117/
Bridges, M. Moo-ove over, cow’s milk: the rise of plant-based dairy alternatives. Pract Gastroenterol. 2018;42(1):20–7. https://practicalgastro.com/2019/07/29/moo-ove-over-cows-milk-the-rise-of-plant-based-dairy-alternatives/
Boland, MA. Milk processors are going bankrupt as Americans ditch dairy. Bloomberg. https://www.bloomberg.com/news/articles/2020–01–10/distaste-for-dairy-sends-milk-processors-to-bankruptcy-court. Published January 10, 2020. Accessed January 6, 2022.; https://www.bloomberg.com/news/articles/2020-01-10/distaste-for-dairy-sends-milk-processors-to-bankruptcy-court?leadSource=uverify%20wall
Silva ARA, Silva MMN, Ribeiro BD. Health issues and technological aspects of plant-based alternative milk. Food Res Int. 2020;131:108972. https://pubmed.ncbi.nlm.nih.gov/32247441/
Jacobs ET, Foote JA, Kohler LN, Skiba MB, Thomson CA. Re-examination of dairy as a single commodity in US dietary guidance. Nutr Rev. 2020;78(3):225–34. https://pubmed.ncbi.nlm.nih.gov/31904838/
Vanga SK, Raghavan V. How well do plant based alternatives fare nutritionally compared to cow’s milk? J Food Sci Technol. 2018;55(1):10–20. https://pubmed.ncbi.nlm.nih.gov/29358791/
Непереносимость лактозы. – Примеч. ред.
Storhaug CL, Fosse SK, Fadnes LT. Country, regional, and global estimates for lactose malabsorption in adults: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 2017;2(10):738–46. https://pubmed.ncbi.nlm.nih.gov/28690131/
National Institute of Child Health and Human Development. Lactose intolerance: information for health care providers. U.S. Dept. of Health and Human Services, National Institutes of Health. http://purl.access.gpo.gov/GPO/LPS80173. Published January 2006. Accessed January 6, 2022.; https://purl.access.gpo.gov/GPO/LPS80173
Bertron P, Barnard ND, Mills M. Racial bias in federal nutrition policy, part I: the public health implications of variations in lactase persistence. J Natl Med Assoc. 1999;91(3):151–7. https://pubmed.ncbi.nlm.nih.gov/10203917/
Jacobs ET, Foote JA, Kohler LN, Skiba MB, Thomson CA. Re-examination of dairy as a single commodity in US dietary guidance. Nutr Rev. 2020;78(3):225–34. https://pubmed.ncbi.nlm.nih.gov/31904838/
Jacobs ET, Foote JA, Kohler LN, Skiba MB, Thomson CA. Re-examination of dairy as a single commodity in US dietary guidance. Nutr Rev. 2020;78(3):225–34. https://pubmed.ncbi.nlm.nih.gov/31904838/
Godlee F, Malone R, Timmis A, et al. Journal policy on research funded by the tobacco industry. Thorax. 2013;68(12):1090–1. https://pubmed.ncbi.nlm.nih.gov/24130154/
Yi M, Wu X, Zhuang W, et al. Tea consumption and health outcomes: umbrella review of meta-analyses of observational studies in humans. Mol Nutr Food Res. 2019;63(16):e1900389. https://pubmed.ncbi.nlm.nih.gov/31216091/
Zhang L, Jie G, Zhang J, Zhao B. Significant longevity-extending effects of EGCG on Caenorhabditis elegans under stress. Free Radic Biol Med. 2009;46(3):414–21. https://pubmed.ncbi.nlm.nih.gov/19061950/
Niu Y, Na L, Feng R, et al. The phytochemical, EGCG, extends lifespan by reducing liver and kidney function damage and improving age-associated inflammation and oxidative stress in healthy rats. Aging Cell. 2013;12(6):1041–9. https://pubmed.ncbi.nlm.nih.gov/23834676/
Yi M, Wu X, Zhuang W, et al. Tea consumption and health outcomes: umbrella review of meta-analyses of observational studies in humans. Mol Nutr Food Res. 2019;63(16):e1900389. https://pubmed.ncbi.nlm.nih.gov/31216091/
Spiegelhalter D. Using speed of ageing and “microlives” to communicate the effects of lifetime habits and environment. BMJ. 2012;345:e8223. https://pubmed.ncbi.nlm.nih.gov/23247978/
Yi M, Wu X, Zhuang W, et al. Tea consumption and health outcomes: umbrella review of meta-analyses of observational studies in humans. Mol Nutr Food Res. 2019;63(16):e1900389. https://pubmed.ncbi.nlm.nih.gov/31216091/
Jochmann N, Lorenz M, von Krosigk A, et al. The efficacy of black tea in ameliorating endothelial function is equivalent to that of green tea. Br J Nutr. 2008;99(4):863–8. https://pubmed.ncbi.nlm.nih.gov/17916273/
Lorenz M, Jochmann N, von Krosigk A, et al. Addition of milk prevents vascular protective effects of tea. Eur Heart J. 2007;28(2):219–23. https://pubmed.ncbi.nlm.nih.gov/17213230/
Ahmad AF, Rich L, Koch H, et al. Effect of adding milk to black tea on vascular function in healthy men and women: a randomised controlled crossover trial. Food Funct. 2018;9(12):6307–14. https://pubmed.ncbi.nlm.nih.gov/30411751/
Serafini M, Testa MF, Villaño D, et al. Antioxidant activity of blueberry fruit is impaired by association with milk. Free Radic Biol Med. 2009;46(6):769–74. https://pubmed.ncbi.nlm.nih.gov/19135520/
Serafini M, Bugianesi R, Maiani G, Valtuena S, De Santis S, Crozier A. Plasma antioxidants from chocolate. Nature. 2003;424(6952):1013. https://pubmed.ncbi.nlm.nih.gov/12944955/
Duarte GS, Farah A. Effect of simultaneous consumption of milk and coffee on chlorogenic acids’ bioavailability in humans. J Agric Food Chem. 2011;59(14):7925–31. https://pubmed.ncbi.nlm.nih.gov/21627318/
Получают из побегов аспалатуса линейного, кустарника из семейства бобовых. – Примеч. ред.
Chen W, Sudji IR, Wang E, Joubert E, van Wyk BE, Wink M. Ameliorative effect of aspalathin from rooibos (Aspalathus linearis) on acute oxidative stress in Caenorhabditis elegans. Phytomedicine. 2013;20(3–4):380–6. https://pubmed.ncbi.nlm.nih.gov/23218401/
Yoo KM, Hwang IK, Moon B. Comparative flavonoids contents of selected herbs and associations of their radical scavenging activity with antiproliferative actions in V79–4 cells. J Food Sci. 2009;74(6):C419–25. https://pubmed.ncbi.nlm.nih.gov/19723177/
Damiani E, Carloni P, Rocchetti G, et al. Impact of cold versus hot brewing on the phenolic profile and antioxidant capacity of rooibos (Aspalathus linearis) herbal tea. Antioxidants (Basel). 2019;8(10):499. https://pubmed.ncbi.nlm.nih.gov/31640245/
Cleverdon R, Elhalaby Y, McAlpine MD, Gittings W, Ward WE. Total polyphenol content and antioxidant capacity of tea bags: comparison of black, green, red rooibos, chamomile and peppermint over different steep times. Beverages. 2018;4(1):15. https://www.mdpi.com/2306-5710/4/1/15
Peterson J, Dwyer J, Jacques P, Rand W, Prior R, Chui K. Tea variety and brewing techniques influence flavonoid content of black tea. J Food Compost Anal. 2004;17(3–4):397–405. https://www.sciencedirect.com/science/article/abs/pii/S0889157504000614
Saklar S, Ertas E, Ozdemir IS, Karadeniz B. Effects of different brewing conditions on catechin content and sensory acceptance in Turkish green tea infusions. J Food Sci Technol. 2015;52(10):6639–46. https://pubmed.ncbi.nlm.nih.gov/26396411/
Pérez-Burillo S, Giménez R, Rufián-Henares JA, Pastoriza S. Effect of brewing time and temperature on antioxidant capacity and phenols of white tea: relationship with sensory properties. Food Chem. 2018;248:111–8. https://pubmed.ncbi.nlm.nih.gov/29329833/
Nikniaz Z, Mahdavi R, Ghaemmaghami SJ, Yagin NL, Nikniaz L. Effect of different brewing times on antioxidant activity and polyphenol content of loosely packed and bagged black teas (Camellia sinensis L.). Avicenna J Phytomed. 2016;6(3):313–21. https://pubmed.ncbi.nlm.nih.gov/27462554/
Malik VS, Li Y, Pan A, et al. Long-term consumption of sugar-sweetened and artificially sweetened beverages and risk of mortality in US adults. Circulation. 2019;139(18):2113–25. https://pubmed.ncbi.nlm.nih.gov/30882235/
Zhang YB, Jiang YW, Chen JX, Xia PF, Pan A. Association of consumption of sugar-sweetened beverages or artificially sweetened beverages with mortality: a systematic review and dose-response meta-analysis of prospective cohort studies. Adv Nutr. 2021;12(2):374–83. https://pubmed.ncbi.nlm.nih.gov/33786594/
Huang C, Huang J, Tian Y, Yang X, Gu D. Sugar sweetened beverages consumption and risk of coronary heart disease: a meta-analysis of prospective studies. Atherosclerosis. 2014;234(1):11–6. https://pubmed.ncbi.nlm.nih.gov/24583500/
Imamura F, O’Connor L, Ye Z, et al. Consumption of sugar sweetened beverages, artificially sweetened beverages, and fruit juice and incidence of type 2 diabetes: systematic review, meta-analysis, and estimation of population attributable fraction. BMJ. 2015;351:h3576. https://pubmed.ncbi.nlm.nih.gov/26199070/
Zhang YB, Jiang YW, Chen JX, Xia PF, Pan A. Association of consumption of sugar-sweetened beverages or artificially sweetened beverages with mortality: a systematic review and dose-response meta-analysis of prospective cohort studies. Adv Nutr. 2021;12(2):374–83. https://pubmed.ncbi.nlm.nih.gov/33786594/
Gardener H, Elkind MSV. Artificial sweeteners, real risks. Stroke. 2019;50(3):549–51. https://pubmed.ncbi.nlm.nih.gov/30760171/
Huang CW, Wang HD, Bai H, et al. Tequila regulates insulin-like signaling and extends life span in Drosophila melanogaster. J Gerontol A Biol Sci Med Sci. 2015;70(12):1461–9. https://pubmed.ncbi.nlm.nih.gov/26265729/
Didelot G, Molinari F, Tchénio P, et al. Tequila, a neurotrypsin ortholog, regulates long-term memory formation in Drosophila. Science. 2006;313(5788):851–3. https://pubmed.ncbi.nlm.nih.gov/16902143/
Griswold MG, Fullman N, Hawley C, et al. Alcohol use and burden for 195 countries and territories, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2018;392(10152):1015–35. https://pubmed.ncbi.nlm.nih.gov/30146330/
Degenhardt L, Charlson F, Ferrari A, et al. The global burden of disease attributable to alcohol and drug use in 195 countries and territories, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Psychiatry. 2018;5(12):987–1012. https://pubmed.ncbi.nlm.nih.gov/30392731/
CDC Morbidity and Mortality Weekly Report. Alcohol-attributable deaths and years of potential life lost – United States, 2001. http://www.cdc.gov/mmwr/preview/mmwrhtml/mm5337a2.htm. Published September 24, 2004. Accessed October 31. 2021.; https://www.cdc.gov/mmwr/preview/mmwrhtml/mm5337a2.htm
Martinez P, Kerr WC, Subbaraman MS, Roberts SCM. New estimates of the mean ethanol content of beer, wine, and spirits sold in the United States show a greater increase in per capita alcohol consumption than previous estimates. Alcohol Clin Exp Res. 2019;43(3):509–21. https://pubmed.ncbi.nlm.nih.gov/30742317/
Editorial. Alcohol and health: time for an overdue conversation. Lancet Gastroenterol Hepatol. 2020;5(3):229. https://pubmed.ncbi.nlm.nih.gov/32061324/
Seyedsadjadi N, Grant R. The potential benefit of monitoring oxidative stress and inflammation in the prevention of non-communicable diseases (NCDs). Antioxidants (Basel). 2020;10(1):15. https://pubmed.ncbi.nlm.nih.gov/33375428/
Guest J, Guillemin GJ, Heng B, Grant R. Lycopene pretreatment ameliorates acute ethanol induced NAD+ depletion in human astroglial cells. Oxid Med Cell Longev. 2015;2015:1–8. https://pubmed.ncbi.nlm.nih.gov/26075038/
Chen H, Chen T, Giudici P, Chen F. Vinegar functions on health: constituents, sources, and formation mechanisms. Compr Rev Food Sci Food Saf. 2016;15(6):1124–38. https://pubmed.ncbi.nlm.nih.gov/33401833/
Ali Z, Wang Z, Amir RM, et al. Potential uses of vinegar as a medicine and related in vivo mechanisms. Int J Vitam Nutr Res. 2018;86(3–4):1–12. https://pubmed.ncbi.nlm.nih.gov/29580192/
Bagnardi V, Rota M, Botteri E, et al. Alcohol consumption and site-specific cancer risk: a comprehensive dose-response meta-analysis. Br J Cancer. 2015;112(3):580–93. https://pubmed.ncbi.nlm.nih.gov/25422909/
Choi YJ, Myung SK, Lee JH. Light alcohol drinking and risk of cancer: a meta-analysis of cohort studies. Cancer Res Treat. 2018;50(2):474–87. https://pubmed.ncbi.nlm.nih.gov/28546524/
Testino G, Leone S, Sumberaz A, Borro P. Alcohol and cancer. Alcohol Clin Exp Res. 2015;39(11):2261. https://pubmed.ncbi.nlm.nih.gov/26332802/
Brien SE, Ronksley PE, Turner BJ, Mukamal KJ, Ghali WA. Effect of alcohol consumption on biological markers associated with risk of coronary heart disease: systematic review and meta-analysis of interventional studies. BMJ. 2011;342:d636. https://pubmed.ncbi.nlm.nih.gov/21343206/
Voight BF, Peloso GM, Orho-Melander M, et al. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet. 2012;380(9841):572–80. https://pubmed.ncbi.nlm.nih.gov/22607825/
Linsel-Nitschke P, Götz A, Erdmann J, et al. Lifelong reduction of LDL-cholesterol related to a common variant in the LDL-receptor gene decreases the risk of coronary artery disease – a Mendelian randomisation study. PLoS One. 2008;3(8):e2986. https://pubmed.ncbi.nlm.nih.gov/18714375/
Britton AR, Grobbee DE, den Ruijter HM, et al. Alcohol consumption and common carotid intima-media thickness: the USE-IMT Study. Alcohol Alcohol. 2017;52(4):483–6. https://pubmed.ncbi.nlm.nih.gov/28525540/
Отложение солей кальция на стенках артерий, питающих сердце. – Примеч. ред.
Pletcher MJ, Varosy P, Kiefe CI, Lewis CE, Sidney S, Hulley SB. Alcohol consumption, binge drinking, and early coronary calcification: findings from the Coronary Artery Risk Development in Young Adults (CARDIA) Study. Am J Epidemiol. 2005;161(5):423–33. https://pubmed.ncbi.nlm.nih.gov/15718478/
McFadden CB, Brensinger CM, Berlin JA, Townsend RR. Systematic review of the effect of daily alcohol intake on blood pressure. Am J Hypertens. 2005;18(2):276–86. https://pubmed.ncbi.nlm.nih.gov/15752957/
Xi B, Veeranki SP, Zhao M, Ma C, Yan Y, Mi J. Relationship of alcohol consumption to all-cause, cardiovascular, and cancer-related mortality in U.S. adults. J Am Coll Cardiol. 2017;70(8):913–22. https://pubmed.ncbi.nlm.nih.gov/28818200/
Xi B, Veeranki SP, Zhao M, Ma C, Yan Y, Mi J. Relationship of alcohol consumption to all-cause, cardiovascular, and cancer-related mortality in U.S. adults. J Am Coll Cardiol. 2017;70(8):913–22. https://pubmed.ncbi.nlm.nih.gov/28818200/
Stockwell T, Zhao J. Alcohol’s contribution to cancer is underestimated for exactly the same reason that its contribution to cardioprotection is overestimated. Addiction. 2017;112(2):230–2. https://pubmed.ncbi.nlm.nih.gov/27891690/
Doll R, Peto R, Boreham J, Sutherland I. Mortality from cancer in relation to smoking: 50 years observations on British doctors. Br J Cancer. 2005;92(3):426–9. https://pubmed.ncbi.nlm.nih.gov/15668706/
Stockwell T, Zhao J, Panwar S, Roemer A, Naimi T, Chikritzhs T. Do “moderate” drinkers have reduced mortality risk? A systematic review and meta-analysis of alcohol consumption and all-cause mortality. J Stud Alcohol Drugs. 2016;77(2):185–98. https://pubmed.ncbi.nlm.nih.gov/26997174/
Sattar N, Preiss D. Reverse causality in cardiovascular epidemiological research: more common than imagined? Circulation. 2017;135(24):2369–72. https://pubmed.ncbi.nlm.nih.gov/28606949/
Costantino G, Montano N, Casazza G. When should we change our clinical practice based on the results of a clinical study? The hierarchy of evidence. Intern Emerg Med. 2015;10(6):745–7. https://pubmed.ncbi.nlm.nih.gov/25860505/
Huynh K. Reducing alcohol intake improves heart health. Nat Rev Cardiol. 2014;11(9):495. https://pubmed.ncbi.nlm.nih.gov/25072907/
Stott DJ. Alcohol and mortality in older people: understanding the J-shaped curve. Age Ageing. 2020;49(3):332–3. https://pubmed.ncbi.nlm.nih.gov/32343789/
Costantino G, Montano N, Casazza G. When should we change our clinical practice based on the results of a clinical study? The hierarchy of evidence. Intern Emerg Med. 2015;10(6):745–7. https://pubmed.ncbi.nlm.nih.gov/25860505/
Mohammadi-Shemirani P, Chong M, Pigeyre M, Morton RW, Gerstein HC, Paré G. Effects of lifelong testosterone exposure on health and disease using Mendelian randomization. Elife. 2020;9:e58914. https://pubmed.ncbi.nlm.nih.gov/33063668/
Zuccolo L, Holmes MV. Commentary: Mendelian randomization-inspired causal inference in the absence of genetic data. Int J Epidemiol. 2017;46(3):962–5. https://pubmed.ncbi.nlm.nih.gov/28025256/
Zuccolo L, Holmes MV. Commentary: Mendelian randomization-inspired causal inference in the absence of genetic data. Int J Epidemiol. 2017;46(3):962–5. https://pubmed.ncbi.nlm.nih.gov/28025256/
Goulden R. Moderate alcohol consumption is not associated with reduced all-cause mortality. Am J Med. 2016;129(2):180–6.e4. https://pubmed.ncbi.nlm.nih.gov/26524703/
Zuccolo L, Holmes MV. Commentary: Mendelian randomization-inspired causal inference in the absence of genetic data. Int J Epidemiol. 2017;46(3):962–5. https://pubmed.ncbi.nlm.nih.gov/28025256/
Holmes MV, Dale CE, Zuccolo L, et al. Association between alcohol and cardiovascular disease: Mendelian randomisation analysis based on individual participant data. BMJ. 2014;349:g4164. https://pubmed.ncbi.nlm.nih.gov/25011450/
Xi B, Veeranki SP, Zhao M, Ma C, Yan Y, Mi J. Relationship of alcohol consumption to all-cause, cardiovascular, and cancer-related mortality in U.S. adults. J Am Coll Cardiol. 2017;70(8):913–22. https://pubmed.ncbi.nlm.nih.gov/28818200/
Costanzo S, de Gaetano G, Di Castelnuovo A, Djoussé L, Poli A, van Velden DP. Moderate alcohol consumption and lower total mortality risk: justified doubts or established facts? Nutr Metab Cardiovasc Dis. 2019;29(10):1003–8. https://pubmed.ncbi.nlm.nih.gov/31400826/
Oppenheimer GM, Bayer R. Is moderate drinking protective against heart disease? The science, politics and history of a public health conundrum. Milbank Q. 2020;98(1):39–56. https://pubmed.ncbi.nlm.nih.gov/31803980/
Skovenborg E, Grønbæk M, Ellison RC. Benefits and hazards of alcohol-the J-shaped curve and public health. DAT. 2021;21(1):54–69. https://portal.findresearcher.sdu.dk/en/publications/benefits-and-hazards-of-alcohol-the-j-shaped-curve-and-public-hea
Golder S, McCambridge J. Alcohol, cardiovascular disease and industry funding: a co-authorship network analysis of systematic reviews. Soc Sci Med. 2021;289:114450. https://pubmed.ncbi.nlm.nih.gov/34607052/
Costanzo S, de Gaetano G, Di Castelnuovo A, Djoussé L, Poli A, van Velden DP. Moderate alcohol consumption and lower total mortality risk: justified doubts or established facts? Nutr Metab Cardiovasc Dis. 2019;29(10):1003–8. https://pubmed.ncbi.nlm.nih.gov/31400826/
Connor J. Why do alcohol’s assumed benefits have any role in policymaking? J Stud Alcohol Drugs. 2016;77(2):201–2. https://pubmed.ncbi.nlm.nih.gov/26997176/
Rabin RC. Federal agency courted alcohol industry to fund study on benefits of moderate drinking. The New York Times. https://www.nytimes.com/2018/03/17/health/nih-alcohol-study-liquor-industry.html. Published March 17, 2018. Accessed October 21, 2021.; https://www.nytimes.com/2018/03/17/health/nih-alcohol-study-liquor-industry.html
Rabin RC. Federal agency courted alcohol industry to fund study on benefits of moderate drinking. The New York Times. https://www.nytimes.com/2018/03/17/health/nih-alcohol-study-liquor-industry.html. Published March 17, 2018. Accessed October 21, 2021.; https://www.nytimes.com/2018/03/17/health/nih-alcohol-study-liquor-industry.html
Rabin RC. Major study of drinking will be shut down. The New York Times. https://www.nytimes.com/2018/06/15/health/alcohol-nih-drinking.html. Published June 15, 2018. Accessed October 21, 2021.; https://www.nytimes.com/2018/06/15/health/alcohol-nih-drinking.html
Rabin RC. Federal agency courted alcohol industry to fund study on benefits of moderate drinking. The New York Times. https://www.nytimes.com/2018/03/17/health/nih-alcohol-study-liquor-industry.html. Published March 17, 2018. Accessed October 21, 2021.; https://www.nytimes.com/2018/03/17/health/nih-alcohol-study-liquor-industry.html
Braillon A, Wilson M. Does moderate alcohol consumption really have health benefits? BMJ. 2018;362:k3888. https://pubmed.ncbi.nlm.nih.gov/30224550/
Oppenheimer GM, Bayer R. Is moderate drinking protective against heart disease? The science, politics and history of a public health conundrum. Milbank Q. 2020;98(1):39–56. https://pubmed.ncbi.nlm.nih.gov/31803980/
Britton A. Moderate alcohol consumption and total mortality risk: do not advocate drinking for “health benefits.” Nutr Metab Cardiovasc Dis. 2019;29(10):1009–10. https://pubmed.ncbi.nlm.nih.gov/31362849/
Burton R, Sheron N. No level of alcohol consumption improves health. Lancet. 2018;392(10152):987–8. https://pubmed.ncbi.nlm.nih.gov/30146328/
Britton A. Moderate alcohol consumption and total mortality risk: do not advocate drinking for “health benefits.” Nutr Metab Cardiovasc Dis. 2019;29(10):1009–10. https://pubmed.ncbi.nlm.nih.gov/31362849/
Manolis TA, Manolis AA, Manolis AS. Cardiovascular effects of alcohol: a double-edged sword / how to remain at the nadir point of the J-curve? Alcohol. 2019;76:117–29. https://pubmed.ncbi.nlm.nih.gov/30735906/
Arora M, ElSayed A, Beger B, et al. The impact of alcohol consumption on cardiovascular health: myths and measures. Glob Heart. 2022;17(1):45. https://pubmed.ncbi.nlm.nih.gov/36051324/
Griswold MG, Fullman N, Hawley C, et al. Alcohol use and burden for 195 countries and territories, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2018;392(10152):1015–35. https://pubmed.ncbi.nlm.nih.gov/30146330/
Holahan CJ, Schutte KK, Brennan PL, et al. Wine consumption and 20-year mortality among late-life moderate drinkers. J Stud Alcohol Drugs. 2012;73(1):80–8. https://pubmed.ncbi.nlm.nih.gov/24588326/
Frankel EN, Kanner J, German JB, Parks E, Kinsella JE. Inhibition of oxidation of human low-density lipoprotein by phenolic substances in red wine. Lancet. 1993;341(8843):454–7. https://pubmed.ncbi.nlm.nih.gov/8094487/
Meagher EA, Barry OP, Burke A, et al. Alcohol-induced generation of lipid peroxidation products in humans. J Clin Invest. 1999;104(6):805–13. https://pubmed.ncbi.nlm.nih.gov/10491416/
Di Renzo L, Carraro A, Valente R, Iacopino L, Colica C, De Lorenzo A. Intake of red wine in different meals modulates oxidized LDL level, oxidative and inflammatory gene expression in healthy people: a randomized crossover trial. Oxid Med Cell Longev. 2014;2014:681318. https://pubmed.ncbi.nlm.nih.gov/24876915/
Caccetta RAA, Burke V, Mori TA, Beilin LJ, Puddey IB, Croft KD. Red wine polyphenols, in the absence of alcohol, reduce lipid peroxidative stress in smoking subjects. Free Radic Biol Med. 2001;30(6):636–42. https://pubmed.ncbi.nlm.nih.gov/11295361/
Schrieks IC, van den Berg R, Sierksma A, Beulens JWJ, Vaes WHJ, Hendriks HFJ. Effect of red wine consumption on biomarkers of oxidative stress. Alcohol Alcohol. 2013;48(2):153–9. https://pubmed.ncbi.nlm.nih.gov/22859618/
Chiva-Blanch G, Urpi-Sarda M, Ros E, et al. Dealcoholized red wine decreases systolic and diastolic blood pressure and increases plasma nitric oxide: short communication. Circ Res. 2012;111(8):1065–8. https://pubmed.ncbi.nlm.nih.gov/22955728/
Naissides M, Mamo JCL, James AP, Pal S. The effect of acute red wine polyphenol consumption on postprandial lipaemia in postmenopausal women. Atherosclerosis. 2004;177(2):401–8. https://pubmed.ncbi.nlm.nih.gov/15530916/
Williams MJA, Sutherland WHF, Whelan AP, McCormick MP, de Jong SA. Acute effect of drinking red and white wines on circulating levels of inflammation-sensitive molecules in men with coronary artery disease. Metabolism. 2004;53(3):318–23. https://pubmed.ncbi.nlm.nih.gov/15015143/
Agewall S, Wright S, Doughty RN, Whalley GA, Duxbury M, Sharpe N. Does a glass of red wine improve endothelial function? Eur Heart J. 2000;21(1):74–8. https://pubmed.ncbi.nlm.nih.gov/10610747/
Hashimoto M, Kim S, Eto M, et al. Effect of acute intake of red wine on flow-mediated vasodilatation of the brachial artery. Am J Cardiol. 2001;88(12):1457–60. https://pubmed.ncbi.nlm.nih.gov/11741577/
Boban M, Modun D, Music I, et al. Red wine induced modulation of vascular function: separating the role of polyphenols, ethanol, and urates. J Cardiovasc Pharmacol. 2006;47(5):695–701. https://pubmed.ncbi.nlm.nih.gov/16775510/
Whelan AP, Sutherland WHF, McCormick MP, Yeoman DJ, de Jong SA, Williams MJA. Effects of white and red wine on endothelial function in subjects with coronary artery disease. Intern Med J. 2004;34(5):224–8. https://pubmed.ncbi.nlm.nih.gov/15151666/
Karatzi K, Papamichael C, Aznaouridis K, et al. Constituents of red wine other than alcohol improve endothelial function in patients with coronary artery disease. Coron Artery Dis. 2004;15(8):485–90. https://pubmed.ncbi.nlm.nih.gov/15585989/
Shukitt-Hale B, Carey A, Simon L, Mark DA, Joseph JA. Effects of Concord grape juice on cognitive and motor deficits in aging. Nutrition. 2006;22(3):295–302. https://pubmed.ncbi.nlm.nih.gov/16412610/
Smith JM, Stouffer EM. Concord grape juice reverses the age-related impairment in latent learning in rats. Nutr Neurosci. 2014;17(2):81–7. https://pubmed.ncbi.nlm.nih.gov/23541291/
Американская компания, с 1956 года принадлежит Национальной виноградной кооперативной ассоциации, кооперативу производителей винограда. – Примеч. ред.
Krikorian R, Nash TA, Shidler MD, Shukitt-Hale B, Joseph JA. Concord grape juice supplementation improves memory function in older adults with mild cognitive impairment. Br J Nutr. 2010;103(5):730–4. https://pubmed.ncbi.nlm.nih.gov/20028599/
Wang DD, Li Y, Bhupathiraju SN, et al. Fruit and vegetable intake and mortality: results from 2 prospective cohort studies of US men and women and a meta-analysis of 26 cohort studies. Circulation. 2021;143(17):1642–54. https://pubmed.ncbi.nlm.nih.gov/33641343/
Dai Q, Borenstein AR, Wu Y, Jackson JC, Larson EB. Fruit and vegetable juices and Alzheimer’s disease: the Kame Project. Am J Med. 2006;119(9):751–9. https://pubmed.ncbi.nlm.nih.gov/16945610/
Dai Q, Borenstein AR, Wu Y, Jackson JC, Larson EB. Fruit and vegetable juices and Alzheimer’s disease: the Kame Project. Am J Med. 2006;119(9):751–9. https://pubmed.ncbi.nlm.nih.gov/16945610/
Mee KA, Gee DL. Apple fiber and gum arabic lowers total and low-density lipoprotein cholesterol levels in men with mild hypercholesterolemia. J Am Diet Assoc. 1997;97(4):422–4. https://pubmed.ncbi.nlm.nih.gov/9120199/
Buscemi S, Rosafio G, Arcoleo G, et al. Effects of red orange juice intake on endothelial function and inflammatory markers in adult subjects with increased cardiovascular risk. Am J Clin Nutr. 2012;95(5):1089–95. https://pubmed.ncbi.nlm.nih.gov/22492368/
Hägele FA, Büsing F, Nas A, et al. High orange juice consumption with or in-between three meals a day differently affects energy balance in healthy subjects. Nutr Diabetes. 2018;8(1):19. https://pubmed.ncbi.nlm.nih.gov/29695707/
Silaste ML, Alfthan G, Aro A, Kesäniemi YA, Hörkkö S. Tomato juice decreases LDL cholesterol levels and increases LDL resistance to oxidation. Br J Nutr. 2007;98(6):1251–8. https://pubmed.ncbi.nlm.nih.gov/17617941/
Samaras A, Tsarouhas K, Paschalidis E, et al. Effect of a special carbohydrate-protein bar and tomato juice supplementation on oxidative stress markers and vascular endothelial dynamics in ultra-marathon runners. Food Chem Toxicol. 2014;69:231–6. https://pubmed.ncbi.nlm.nih.gov/24705018/
Mazidi M, Katsiki N, George ES, Banach M. Tomato and lycopene consumption is inversely associated with total and cause-specific mortality: a population-based cohort study, on behalf of the International Lipid Expert Panel (ILEP). Br J Nutr. 2020;124(12):1303–10. https://pubmed.ncbi.nlm.nih.gov/31434581/
Pan B, Ge L, Lai H, et al. Association of soft drink and 100 % fruit juice consumption with all-cause mortality, cardiovascular diseases mortality, and cancer mortality: a systematic review and dose-response meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr. 2021;Jun 13:1–12. https://pubmed.ncbi.nlm.nih.gov/34121531/
Scheffers FR, Boer JMA. Sugar intake and all-cause mortality-differences between sugar-sweetened beverages, artificially sweetened beverages, and pure fruit juices. BMC Med. 2020;18(1):112. https://pubmed.ncbi.nlm.nih.gov/32316967/
Yip CSC, Chan W, Fielding R. The associations of fruit and vegetable intakes with burden of diseases: a systematic review of meta-analyses. J Acad Nutr Diet. 2019;119(3):464–81. https://pubmed.ncbi.nlm.nih.gov/30639206/
Leaf A. Long-lived populations: extreme old age. J Am Geriatr Soc. 1982;30(8):485–7. https://pubmed.ncbi.nlm.nih.gov/6212609/
Zak N. Evidence that Jeanne Calment died in 1934, not 1997. Rejuvenation Res. 2019;22(1):3–12. https://pubmed.ncbi.nlm.nih.gov/30696353/
Leaf A. Long-lived populations: extreme old age. J Am Geriatr Soc. 1982;30(8):485–7. https://pubmed.ncbi.nlm.nih.gov/6212609/
Mazess RB, Forman SH. Longevity and age exaggeration in Vilcabamba, Ecuador. J Gerontol. 1979;34(1):94–8. https://pubmed.ncbi.nlm.nih.gov/759498/
Poulain M, Herm A, Pes G. The Blue Zones: areas of exceptional longevity around the world. Vienna Yearb Popul Res. 2014;11:87–108. https://www.researchgate.net/publication/255508953_The_Blue_Zones_areas_of_exceptional_longevity_around_the_world
Willcox BJ, Willcox DC, Ferrucci L. Secrets of healthy aging and longevity from exceptional survivors around the globe: lessons from octogenarians to supercentenarians. J Gerontol A Biol Sci Med Sci. 2008;63(11):1181–5. https://pubmed.ncbi.nlm.nih.gov/19038832/
Willcox DC, Willcox BJ, Poon LW. Centenarian studies: important contributors to our understanding of the aging process and longevity. Curr Gerontol Geriatr Res. 2010;2010:484529. https://pubmed.ncbi.nlm.nih.gov/21804821/
Poulain M, Herm A, Pes G. The Blue Zones: areas of exceptional longevity around the world. Vienna Yearb Popul Res. 2014;11:87–108. https://www.researchgate.net/publication/255508953_The_Blue_Zones_areas_of_exceptional_longevity_around_the_world
Carter ED. Making the Blue Zones: neoliberalism and nudges in public health promotion. Soc Sci Med. 2015;133:374–82. https://pubmed.ncbi.nlm.nih.gov/25605430/
Madrigal-Leer F, Martìnez-Montandòn A, Solìs-Umaña M, et al. Clinical, functional, mental and social profile of the Nicoya Peninsula centenarians, Costa Rica, 2017. Aging Clin Exp Res. 2020;32(2):313–21. https://pubmed.ncbi.nlm.nih.gov/30919261/
Vatner SF, Zhang J, Oydanich M, Berkman T, Naftalovich R, Vatner DE. Healthful aging mediated by inhibition of oxidative stress. Ageing Res Rev. 2020;64:101194. https://pubmed.ncbi.nlm.nih.gov/33091597/
Marston HR, Niles-Yokum K, Silva PA. A commentary on Blue Zones®: a critical review of age-friendly environments in the 21st century and beyond. Int J Environ Res Public Health. 2021;18(2):837. https://pubmed.ncbi.nlm.nih.gov/33478140/
Panagiotakos DB, Chrysohoou C, Siasos G, et al. Sociodemographic and lifestyle statistics of oldest old people (80 years) living in Ikaria Island: the Ikaria Study. Cardiol Res Pract. 2011;2011:679187. https://pubmed.ncbi.nlm.nih.gov/21403883/
Food guidelines. BlueZones.com. https://www.bluezones.com/recipes/food-guidelines/. Accessed December 28, 2022.; https://www.bluezones.com/recipes/food-guidelines/
Meccariello R, D’Angelo S. Impact of polyphenolic-food on longevity: an elixir of life. An overview. Antioxidants (Basel). 2021;10(4):507. https://pubmed.ncbi.nlm.nih.gov/33805092/
Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161(13):1645–52. https://pubmed.ncbi.nlm.nih.gov/11434797/
Food guidelines. BlueZones.com. https://www.bluezones.com/recipes/food-guidelines/. Accessed December 28, 2022.; https://www.bluezones.com/recipes/food-guidelines/
Weber H. A lecture on means for the prolongation of life: delivered before the Royal College of Physicians of London. BMJ. 1903;2(2240):1445–51. https://pubmed.ncbi.nlm.nih.gov/20761218/
Stathakos D, Pratsinis H, Zachos I, et al. Greek centenarians: assessment of functional health status and life-style characteristics. Exp Gerontol. 2005;40(6):512–8. https://pubmed.ncbi.nlm.nih.gov/15935588/
Chen C. A survey of the dietary nutritional composition of centenarians. Chin Med J (Engl). 2001;114(10):1095–7. https://pubmed.ncbi.nlm.nih.gov/11677774/
Li Y, Bai Y, Tao QL, et al. Lifestyle of Chinese centenarians and their key beneficial factors in Chongqing, China. Asia Pac J Clin Nutr. 2014;23(2):309–14. https://pubmed.ncbi.nlm.nih.gov/24901102/
Ye JJ, Li JC, Peng L, et al. Nonagenarians and centenarians in a rural Han Chinese population: lifestyle and epidemics: letters to the editor. J Am Geriatr Soc. 2009;57(9):1723–4. https://pubmed.ncbi.nlm.nih.gov/19895443/
Vatner SF, Zhang J, Oydanich M, Berkman T, Naftalovich R, Vatner DE. Healthful aging mediated by inhibition of oxidative stress. Ageing Res Rev. 2020;64:101194. https://pubmed.ncbi.nlm.nih.gov/33091597/
Buettner D. The Blue Zones: 9 Lessons for Living Longer from the People Who’ve Lived the Longest. National Geographic; 2012. https://worldcat.org/title/777659970
Darmadi-Blackberry I, Wahlqvist ML, Kouris-Blazos A, et al. Legumes: the most important dietary predictor of survival in older people of different ethnicities. Asia Pac J Clin Nutr. 2004;13(2):217–20. https://pubmed.ncbi.nlm.nih.gov/15228991/
Agricultural Research Service, United States Department of Agriculture. Beans, NFS. FoodDataCentral. https://fdc.nal.usda.gov/fdc-app.html#/food-details/1100362/portions. Published October 30, 2020. Accessed February 16, 2022.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/1100362/portions
Fadnes LT, Økland JM, Haaland ØA, Johansson KA. Estimating impact of food choices on life expectancy: a modeling study. PLoS Med. 2022;19(2):e1003889. https://pubmed.ncbi.nlm.nih.gov/35134067/
U.S. Department of Agriculture. Beans, peas, and lentils. MyPlate.gov. https://www.myplate.gov/eat-healthy/protein-foods/beans-and-peas. Accessed February 16, 2022.; https://www.myplate.gov/eat-healthy/protein-foods/beans-and-peas
Drewnowski A, Rehm CD. Vegetable cost metrics show that potatoes and beans provide most nutrients per penny. PLoS One. 2013;8(5):e63277. https://pubmed.ncbi.nlm.nih.gov/23691007/
Kabagambe EK, Baylin A, Ruiz-Narvarez E, Siles X, Campos H. Decreased consumption of dried mature beans is positively associated with urbanization and nonfatal acute myocardial infarction. J Nutr. 2005;135(7):1770–5. https://pubmed.ncbi.nlm.nih.gov/15987863/
Luyken R, Pikaar NA, Polman H, Schippers FA. The influence of legumes on the serum cholesterol level. Voeding. 1962;23:447–53. https://pubmed.ncbi.nlm.nih.gov/14467529/
Ferreira H, Vasconcelos M, Gil AM, Pinto E. Benefits of pulse consumption on metabolism and health: a systematic review of randomized controlled trials. Crit Rev Food Sci Nutr. 2021;61(1):85–96. https://pubmed.ncbi.nlm.nih.gov/31983216/
Abeysekara S, Chilibeck PD, Vatanparast H, Zello GA. A pulse-based diet is effective for reducing total and LDL-cholesterol in older adults. Br J Nutr. 2012;108 Suppl 1:S103–10. https://pubmed.ncbi.nlm.nih.gov/22916805/
Tokede OA, Onabanjo TA, Yansane A, Gaziano JM, Djoussé L. Soya products and serum lipids: a meta-analysis of randomised controlled trials. Br J Nutr. 2015;114(6):831–43. https://pubmed.ncbi.nlm.nih.gov/21559039/
Kou T, Wang Q, Cai J, et al. Effect of soybean protein on blood pressure in postmenopausal women: a meta-analysis of randomized controlled trials. Food Funct. 2017;8(8):2663–71. https://pubmed.ncbi.nlm.nih.gov/28675204/
Bazzano LA, Thompson AM, Tees MT, Nguyen CH, Winham DM. Non-soy legume consumption lowers cholesterol levels: a meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis. 2011;21(2):94–103. https://pubmed.ncbi.nlm.nih.gov/19939654/
Sievenpiper JL, Kendall CW, Esfahani A, et al. Effect of non-oil-seed pulses on glycaemic control: a systematic review and meta-analysis of randomised controlled experimental trials in people with and without diabetes. Diabetologia. 2009;52(8):1479–95. https://pubmed.ncbi.nlm.nih.gov/19526214/
Palmer SM, Winham DM, Hradek C. Knowledge gaps of the health benefits of beans among low-income women. Am J Health Behav. 2018;42(1):27–38. https://pubmed.ncbi.nlm.nih.gov/29320336/
Hosseinpour-Niazi S, Mirmiran P, Fallah-Ghohroudi A, Azizi F. Non-soya legume-based therapeutic lifestyle change diet reduces inflammatory status in diabetic patients: a randomised cross-over clinical trial. Br J Nutr. 2015;114(2):213–9. https://pubmed.ncbi.nlm.nih.gov/26077375/
Mirmiran P, Hosseinpour-Niazi S, Azizi F. Therapeutic lifestyle change diet enriched in legumes reduces oxidative stress in overweight type 2 diabetic patients: a crossover randomised clinical trial. Eur J Clin Nutr. 2018;72(1):174–6. https://pubmed.ncbi.nlm.nih.gov/28722030/
Mullins AP, Arjmandi BH. Health benefits of plant-based nutrition: focus on beans in cardiometabolic diseases. Nutrients. 2021;13(2):519. https://pubmed.ncbi.nlm.nih.gov/33562498/
Mathur KS, Khan MA, Sharma RD. Hypocholesterolaemic effect of Bengal gram: a long-term study in man. Br Med J. 1968;1(5583):30–1. https://pubmed.ncbi.nlm.nih.gov/5636741/
Esselstyn CB. In cholesterol lowering, moderation kills. Cleve Clin J Med. 2000;67(8):560–4. https://pubmed.ncbi.nlm.nih.gov/10946449/
Геометрическая схема, которая используется для моделирования множеств и для схематичного изображения и отношений между ними. – Примеч. ред.
Tor-Roca A, Garcia-Aloy M, Mattivi F, Llorach R, Andres-Lacueva C, Urpi-Sarda M. Phytochemicals in legumes: a qualitative reviewed analysis. J Agric Food Chem. 2020;68(47):13486–96. https://pubmed.ncbi.nlm.nih.gov/33169614/
Bruno JA, Feldman CH, Konas DW, Kerrihard AL, Matthews EL. Incorporating sprouted chickpea flour in pasta increases brachial artery flow-mediated dilation. Physiol Int. 2019;106(3):207–12. https://pubmed.ncbi.nlm.nih.gov/31564118/
Zahradka P, Wright B, Weighell W, et al. Daily non-soy legume consumption reverses vascular impairment due to peripheral artery disease. Atherosclerosis. 2013;230(2):310–4. https://pubmed.ncbi.nlm.nih.gov/24075762/
West GB, Brown JH, Enquist BJ. A general model for the origin of allometric scaling laws in biology. Science. 1997;276(5309):122–6. https://pubmed.ncbi.nlm.nih.gov/9082983/
Levine HJ. Rest heart rate and life expectancy. J Am Coll Cardiol. 1997;30(4):1104–6. https://pubmed.ncbi.nlm.nih.gov/9316546/
Cook S, Hess OM. Resting heart rate and cardiovascular events: time for a new crusade? Eur Heart J. 2010;31(5):517–9. https://pubmed.ncbi.nlm.nih.gov/19933283/
Woodward M, Webster R, Murakami Y, et al. The association between resting heart rate, cardiovascular disease and mortality: evidence from 112,680 men and women in 12 cohorts. Eur J Prev Cardiol. 2014;21(6):719–26. https://pubmed.ncbi.nlm.nih.gov/22718796/
Woodward M, Webster R, Murakami Y, et al. The association between resting heart rate, cardiovascular disease and mortality: evidence from 112,680 men and women in 12 cohorts. Eur J Prev Cardiol. 2014;21(6):719–26. https://pubmed.ncbi.nlm.nih.gov/22718796/
Teodorescu C, Reinier K, Uy-Evanado A, Gunson K, Jui J, Chugh SS. Resting heart rate and risk of sudden cardiac death in the general population: influence of left ventricular systolic dysfunction and heart rate-modulating drugs. Heart Rhythm. 2013;10(8):1153–8. https://pubmed.ncbi.nlm.nih.gov/23680897/
Cooney MT, Vartiainen E, Laatikainen T, Juolevi A, Dudina A, Graham IM. Elevated resting heart rate is an independent risk factor for cardiovascular disease in healthy men and women. Am Heart J. 2010;159(4):612–9.e3. https://pubmed.ncbi.nlm.nih.gov/20362720/
Jenkins DJA, Kendall CWC, Augustin LSA, et al. Effect of legumes as part of a low glycemic index diet on glycemic control and cardiovascular risk factors in type 2 diabetes mellitus: a randomized controlled trial. Arch Intern Med. 2012;172(21):1653–60. https://pubmed.ncbi.nlm.nih.gov/23089999/
Sloan RP, Shapiro PA, DeMeersman RE, et al. The effect of aerobic training and cardiac autonomic regulation in young adults. Am J Public Health. 2009;99(5):921–8. https://pubmed.ncbi.nlm.nih.gov/19299682/
Viguiliouk E, Glenn AJ, Nishi SK, et al. Associations between dietary pulses alone or with other legumes and cardiometabolic disease outcomes: an umbrella review and updated systematic review and meta-analysis of prospective cohort studies. Adv Nutr. 2019;10(Suppl_4):S308–19. https://pubmed.ncbi.nlm.nih.gov/31728500/
Fadnes LT, Økland JM, Haaland ØA, Johansson KA. Estimating impact of food choices on life expectancy: a modeling study. PLoS Med. 2022;19(2):e1003889. https://pubmed.ncbi.nlm.nih.gov/35134067/
Schwingshackl L, Schwedhelm C, Hoffmann G, et al. Food groups and risk of all-cause mortality: a systematic review and meta-analysis of prospective studies. Am J Clin Nutr. 2017;105(6):1462–73. https://pubmed.ncbi.nlm.nih.gov/28446499/
Liu W, Hu B, Dehghan M, et al. Fruit, vegetable, and legume intake and the risk of all-cause, cardiovascular, and cancer mortality: a prospective study. Clin Nutr. 2021;40(6):4316–23. https://pubmed.ncbi.nlm.nih.gov/33581953/
Krebs-Smith SM, Guenther PM, Subar AF, Kirkpatrick SI, Dodd KW. Americans do not meet federal dietary recommendations. J Nutr. 2010;140(10):1832–8. https://pubmed.ncbi.nlm.nih.gov/20702750/
Desrochers N, Brauer PM. Legume promotion in counselling: an e-mail survey of dietitians. Can J Diet Pract Res. 2001;62(4):193–8. https://pubmed.ncbi.nlm.nih.gov/11742561/
Winham DM, Hutchins AM. Perceptions of flatulence from bean consumption among adults in 3 feeding studies. Nutr J. 2011;10(1):128. https://pubmed.ncbi.nlm.nih.gov/22104320/
Winham DM, Hutchins AM. Perceptions of flatulence from bean consumption among adults in 3 feeding studies. Nutr J. 2011;10(1):128. https://pubmed.ncbi.nlm.nih.gov/22104320/
Steggerda FR, Dimmick JF. Effects of bean diets on concentration of carbon dioxide in flatus. Am J Clin Nutr. 1966;19(2):120–4. https://pubmed.ncbi.nlm.nih.gov/5916034/
McEligot AJ, Gilpin EA, Rock CL, et al. High dietary fiber consumption is not associated with gastrointestinal discomfort in a diet intervention trial. J Am Diet Assoc. 2002;102(4):549–51. https://pubmed.ncbi.nlm.nih.gov/11985415/
How you can limit your gas production. 12 tips for dealing with flatulence. Harv Health Lett. 2007;32(12):3. https://pubmed.ncbi.nlm.nih.gov/18246621/
Zartl B, Silberbauer K, Loeppert R, Viernstein H, Praznik W, Mueller M. Fermentation of non-digestible raffinose family oligosaccharides and galactomannans by probiotics. Food Funct. 2018;9(3):1638–46. https://pubmed.ncbi.nlm.nih.gov/29465736/
Winham DM, Hutchins AM. Perceptions of flatulence from bean consumption among adults in 3 feeding studies. Nutr J. 2011;10:128. https://pubmed.ncbi.nlm.nih.gov/22104320/
Spiro HM. Fat, foreboding, and flatulence. Ann Intern Med. 1999;130(4 Pt 1):320–2. https://pubmed.ncbi.nlm.nih.gov/10068391/
Schneiderman N, Chirinos DA, Avilés-Santa ML, Heiss G. Challenges in preventing heart disease in hispanics: early lessons learned from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). Prog Cardiovasc Dis. 2014;57(3):253–61. https://pubmed.ncbi.nlm.nih.gov/25212986/
Kochanek KD, Murphy SL, Xu J, Arias E. Mortality in the United States, 2013. Centers for Disease Control and Prevention. NCHS Data Brief. No. 178. Published December 2014. Accessed December 26, 2021.; https://pubmed.ncbi.nlm.nih.gov/25549183/
The Hispanic paradox. Lancet. 2015;385(9981):1918. https://pubmed.ncbi.nlm.nih.gov/26090624/
Colón-Ramos U, Thompson FE, Yaroch AL, et al. Differences in fruit and vegetable intake among Hispanic subgroups in California: results from the 2005 California Health Interview Survey. J Am Diet Assoc. 2009;109(11):1878–85. https://pubmed.ncbi.nlm.nih.gov/19857629/
Reyes-Ortiz CA, Ju H, Eschbach K, Kuo YF, Goodwin JS. Neighbourhood ethnic composition and diet among Mexican-Americans. Public Health Nutr. 2009;12(12):2293–301. https://pubmed.ncbi.nlm.nih.gov/19254428/
Nieddu A, Vindas L, Errigo A, Vindas J, Pes GM, Dore MP. Dietary habits, anthropometric features and daily performance in two independent long-lived populations from Nicoya peninsula (Costa Rica) and Ogliastra (Sardinia). Nutrients. 2020;12(6):E1621. https://pubmed.ncbi.nlm.nih.gov/32492804/
Reyes-Ortiz CA, Ju H, Eschbach K, Kuo YF, Goodwin JS. Neighbourhood ethnic composition and diet among Mexican-Americans. Public Health Nutr. 2009;12(12):2293–301. https://pubmed.ncbi.nlm.nih.gov/19254428/
Shen J, Shan J, Zhu X, et al. Sex specific effects of capsaicin on longevity regulation. Exp Gerontol. 2020;130:110788. https://pubmed.ncbi.nlm.nih.gov/31790803/
Bonaccio M, Di Castelnuovo A, Costanzo S, et al. Chili pepper consumption and mortality in Italian adults. J Am Coll Cardiol. 2019;74(25):3139–49. https://pubmed.ncbi.nlm.nih.gov/31856971/
Chopan M, Littenberg B. The association of hot red chili pepper consumption and mortality: a large population-based cohort study. PLoS One. 2017;12(1):e0169876. https://pubmed.ncbi.nlm.nih.gov/28068423/
Lv J, Qi L, Yu C, et al. Consumption of spicy foods and total and cause specific mortality: population based cohort study. BMJ. 2015;351:h3942. https://pubmed.ncbi.nlm.nih.gov/26242395/
Hashemian M, Poustchi H, Murphy G, et al. Turmeric, pepper, cinnamon, and saffron consumption and mortality. J Am Heart Assoc. 2019;8(18):e012240. https://www.ahajournals.org/doi/10.1161/JAHA.119.012240
Janssens PLHR, Hursel R, Martens EAP, Westerterp-Plantenga MS. Acute effects of capsaicin on energy expenditure and fat oxidation in negative energy balance. PLoS One. 2013;8(7):e67786. https://pubmed.ncbi.nlm.nih.gov/23844093/
Bonaccio M, Di Castelnuovo A, Costanzo S, et al. Chili pepper consumption and mortality in Italian adults. J Am Coll Cardiol. 2019;74(25):3139–49. https://pubmed.ncbi.nlm.nih.gov/31856971/
American Heart Association News. Retired? Hardly – at 99, this pioneering heart doctor is still leading the way. American Heart Association. https://www.heart.org/en/news/2019/10/18/retired-hardly-at-99-this-pioneering-heart-doctor-is-still-leading-the-way. Published October 18, 2019. Accessed December 27, 2021.; https://www.heart.org/en/news/2019/10/18/retired-hardly-at-99-this-pioneering-heart-doctor-is-still-leading-the-way
Stamler J. Toward a modern Mediterranean diet for the 21st century. Nutr Metab Cardiovasc Dis. 2013;23(12):1159–62. https://pubmed.ncbi.nlm.nih.gov/24238655/
Nestle M. Mediterranean diets: historical and research overview. Am J Clin Nutr. 1995;61(6 Suppl):1313S-20S. https://pubmed.ncbi.nlm.nih.gov/7754981/
Keys A, Menotti A, Karvonen MJ, et al. The diet and 15-year death rate in the Seven Countries Study. Am J Epidemiol. 1986;124(6):903–15. https://pubmed.ncbi.nlm.nih.gov/3776973/
Davinelli S, Trichopoulou A, Corbi G, De Vivo I, Scapagnini G. The potential nutrigeroprotective role of Mediterranean diet and its functional components on telomere length dynamics. Ageing Res Rev. 2019;49:1–10. https://pubmed.ncbi.nlm.nih.gov/30448616/
Keys A. Mediterranean diet and public health: personal reflections. Am J Clin Nutr. 1995;61(6 Suppl):1321S-3S. https://pubmed.ncbi.nlm.nih.gov/7754982/
Russo GL, Siani A, Fogliano V, et al. The Mediterranean diet from past to future: key concepts from the second “Ancel Keys” International Seminar. Nutr Metab Cardiovasc Dis. 2021;31(3):717–32. https://pubmed.ncbi.nlm.nih.gov/33558092/
Voukiklaris GE, Kafatos A, Dontas AS. Changing prevalence of coronary heart disease risk factors and cardiovascular diseases in men of a rural area of Crete from 1960 to 1991. Angiology. 1996;47(1):43–9. https://pubmed.ncbi.nlm.nih.gov/8546344/
Altomare R, Cacciabaudo F, Damiano G, et al. The Mediterranean diet: a history of health. Iran J Public Health. 2013;42(5):449–57. https://pubmed.ncbi.nlm.nih.gov/23802101/
Keys A. Mediterranean diet and public health: personal reflections. Am J Clin Nutr. 1995;61(6 Suppl):1321S-3S. https://pubmed.ncbi.nlm.nih.gov/7754982/
Sofi F, Macchi C, Abbate R, Gensini GF, Casini A. Mediterranean diet and health status: an updated meta-analysis and a proposal for a literature-based adherence score. Public Health Nutr. 2014;17(12):2769–82. https://pubmed.ncbi.nlm.nih.gov/24476641/
Kastorini CM, Milionis HJ, Esposito K, Giugliano D, Goudevenos JA, Panagiotakos DB. The effect of Mediterranean diet on metabolic syndrome and its components: a meta-analysis of 50 studies and 534,906 individuals. J Am Coll Cardiol. 2011;57(11):1299–313. https://pubmed.ncbi.nlm.nih.gov/21392646/
Soltani S, Jayedi A, Shab-Bidar S, Becerra-Tomás N, Salas-Salvadó J. Adherence to the Mediterranean diet in relation to all-cause mortality: a systematic review and dose-response meta-analysis of prospective cohort studies. Adv Nutr. 2019;10(6):1029–39. https://pubmed.ncbi.nlm.nih.gov/31111871/
Bellavia A, Tektonidis TG, Orsini N, Wolk A, Larsson SC. Quantifying the benefits of Mediterranean diet in terms of survival. Eur J Epidemiol. 2016;31(5):527–30. https://pubmed.ncbi.nlm.nih.gov/26848763/
Critselis E, Panagiotakos D. Adherence to the Mediterranean diet and healthy ageing: current evidence, biological pathways, and future directions. Crit Rev Food Sci Nutr. 2020;60(13):2148–57. https://pubmed.ncbi.nlm.nih.gov/31272195/
Wang Y, Hao Q, Su L, Liu Y, Liu S, Dong B. Adherence to the Mediterranean diet and the risk of frailty in old people: a systematic review and meta-analysis. J Nutr Health Aging. 2018;22(5):613–8. https://pubmed.ncbi.nlm.nih.gov/29717762/
Eleftheriou D, Benetou V, Trichopoulou A, La Vecchia C, Bamia C. Mediterranean diet and its components in relation to all-cause mortality: meta-analysis. Br J Nutr. 2018;120(10):1081–97. https://pubmed.ncbi.nlm.nih.gov/30401007/
Pett KD, Willett WC, Vartiainen E, Katz DL. The Seven Countries Study. Eur Heart J. 2017;38(42):3119–21. https://pubmed.ncbi.nlm.nih.gov/29121230/
Montani JP. Ancel Keys: the legacy of a giant in physiology, nutrition, and public health. Obes Rev. 2021;22 Suppl 2:e13196. https://pubmed.ncbi.nlm.nih.gov/33496369/
Sparling PB. Legacy of nutritionist Ancel Keys. Mayo Clin Proc. 2020;95(3):615–7. https://pubmed.ncbi.nlm.nih.gov/32138891/
American Heart Association News. Retired? Hardly – at 99, this pioneering heart doctor is still leading the way. American Heart Association. https://www.heart.org/en/news/2019/10/18/retired-hardly-at-99-this-pioneering-heart-doctor-is-still-leading-the-way. Published October 18, 2019. Accessed December 27, 2021.; https://www.heart.org/en/news/2019/10/18/retired-hardly-at-99-this-pioneering-heart-doctor-is-still-leading-the-way
Paul M. As Jeremiah Stamler turns 100, ‘he continues to do brilliant science’. Northwestern Now. https://news.northwestern.edu/stories/2019/10/jeremiah-stamler/. Published October 29, 2019. Accessed December 27, 2021.; https://news.northwestern.edu/stories/2019/10/jeremiah-stamler/
Winter L. “Father of Preventive Cardiology” Jeremiah Stamler dies at 102. The Scientist. https://www.the-scientist.com/news-opinion/father-of-preventive-cardiology-jeremiah-stamler-dies-at-102–69718. Published February 18, 2022. Accessed April 4, 2022.; https://www.the-scientist.com/news-opinion/father-of-preventive-cardiology-jeremiah-stamler-dies-at-102-69718
Bes-Rastrollo M, Sánchez-Villegas A, de la Fuente C, de Irala J, Martínez JA, Martínez-González MA. Olive oil consumption and weight change: the SUN prospective cohort study. Lipids. 2006;41(3):249–56. https://pubmed.ncbi.nlm.nih.gov/16711599/
Guasch-Ferré M, Liu G, Li Y, et al. Olive oil consumption and cardiovascular risk in U.S. adults. J Am Coll Cardiol. 2020;75(15):1729–39. https://pubmed.ncbi.nlm.nih.gov/35027106/
Blankenhorn DH, Johnson RL, Mack WJ, El Zein HA, Vailas LI. The influence of diet on the appearance of new lesions in human coronary arteries. JAMA. 1990;263(12):1646–52. https://pubmed.ncbi.nlm.nih.gov/2407875/
Schwingshackl L, Bogensberger B, Bencic A, Knüppel S, Boeing H, Hoffmann G. Effects of oils and solid fats on blood lipids: a systematic review and network meta-analysis. J Lipid Res. 2018;59(9):1771–82. https://pubmed.ncbi.nlm.nih.gov/30006369/
Tentolouris N, Arapostathi C, Perrea D, Kyriaki D, Revenas C, Katsilambros N. Differential effects of two isoenergetic meals rich in saturated or monounsaturated fat on endothelial function in subjects with type 2 diabetes. Diabetes Care. 2008;31(12):2276–8. https://pubmed.ncbi.nlm.nih.gov/18835957/
Cortés B, Núñez I, Cofán M, et al. Acute effects of high-fat meals enriched with walnuts or olive oil on postprandial endothelial function. J Am Coll Cardiol. 2006;48(8):1666–71. https://pubmed.ncbi.nlm.nih.gov/17045905/
Vogel RA, Corretti MC, Plotnick GD. The postprandial effect of components of the Mediterranean diet on endothelial function. J Am Coll Cardiol. 2000;36(5):1455–60. https://pubmed.ncbi.nlm.nih.gov/11079642/
Vogel RA. Brachial artery ultrasound: a noninvasive tool in the assessment of triglyceride-rich lipoproteins. Clin Cardiol. 1999;22(Suppl II):II-34–9. https://pubmed.ncbi.nlm.nih.gov/10376195/
Rueda-Clausen CF, Silva FA, Lindarte MA, et al. Olive, soybean and palm oils intake have a similar acute detrimental effect over the endothelial function in healthy young subjects. Nutr Metab Cardiovasc Dis. 2007;17(1):50–7. https://pubmed.ncbi.nlm.nih.gov/17174226/
Ong PJ, Dean TS, Hayward CS, Della Monica PL, Sanders TAB, Collins P. Effect of fat and carbohydrate consumption on endothelial function. Lancet. 1999;354(9196):2134. https://pubmed.ncbi.nlm.nih.gov/10609824/
Casas-Agustench P, López-Uriarte P, Ros E, Bulló M, Salas-Salvadó J. Nuts, hypertension and endothelial function. Nutr Metab Cardiovasc Dis. 2011;21 Suppl 1:S21–33. https://pubmed.ncbi.nlm.nih.gov/20031380/
Park E, Edirisinghe I, Burton-Freeman B. Avocado fruit on postprandial markers of cardio-metabolic risk: a randomized controlled dose response trial in overweight and obese men and women. Nutrients. 2018;10(9):E1287. https://pubmed.ncbi.nlm.nih.gov/30213052/
Традиционный соус, в состав которого входят оливковое масло, бальзамический уксус, мед, горчица и чеснок. – Примеч. ред.
Agricultural Research Service, United States Department of Agriculture. Olives, ripe, canned (jumbo-super colossal). FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/food-details/169095/nutrients. Published April 1, 2019. Accessed December 28, 2022.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/169095/nutrients
Martínez-González MÁ, Corella D, Salas-Salvadó J, et al. Cohort profile: design and methods of the PREDIMED study. Int J Epidemiol. 2012;41(2):377–85. https://pubmed.ncbi.nlm.nih.gov/21172932/
Martínez-González MÁ, Corella D, Salas-Salvadó J, et al. Cohort profile: design and methods of the PREDIMED study. Int J Epidemiol. 2012;41(2):377–85. https://pubmed.ncbi.nlm.nih.gov/21172932/
Estruch R, Ros E, Salas-Salvadó J, et al. Primary prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med. 2013;368(14):1279–90. https://pubmed.ncbi.nlm.nih.gov/29897866/
Agarwal A, Ioannidis JPA. PREDIMED trial of Mediterranean diet: retracted, republished, still trusted? BMJ. 2019;364:l341. https://pubmed.ncbi.nlm.nih.gov/30733217/
Rees K, Takeda A, Martin N, et al. Mediterranean-style diet for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev. 2019;3:CD009825. https://pubmed.ncbi.nlm.nih.gov/30864165/
Martínez-González MA, Gea A, Ruiz-Canela M. The Mediterranean diet and cardiovascular health. Circ Res. 2019;124(5):779–98. https://pubmed.ncbi.nlm.nih.gov/30817261/
Estruch R, Ros E, Salas-Salvadó J, et al. Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. N Engl J Med. 2018;378(25):e34. https://pubmed.ncbi.nlm.nih.gov/29897866/
Sala-Vila A, Romero-Mamani ES, Gilabert R, et al. Changes in ultrasound-assessed carotid intima-media thickness and plaque with a Mediterranean diet: a substudy of the PREDIMED trial. Arterioscler Thromb Vasc Biol. 2014;34(2):439–45. https://pubmed.ncbi.nlm.nih.gov/24285581/
Estruch R, Ros E, Salas-Salvadó J, et al. Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. N Engl J Med. 2018;378(25):e34. https://pubmed.ncbi.nlm.nih.gov/29897866/
Tsao CW, Aday AW, Almarzooq ZI, et al. Heart disease and stroke statistics—2022 update: a report from the American Heart Association. Circulation. 2022;145(8):e153–639. https://pubmed.ncbi.nlm.nih.gov/35078371/
Estruch R, Ros E, Salas-Salvadó J, et al. Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. N Engl J Med. 2018;378(25):e34. https://pubmed.ncbi.nlm.nih.gov/29897866/
Guasch-Ferré M, Bulló M, Martínez-González MÁ, et al. Frequency of nut consumption and mortality risk in the PREDIMED nutrition intervention trial. BMC Med. 2013;11:164. https://pubmed.ncbi.nlm.nih.gov/23866098/
Guasch-Ferré M, Hu FB, Martínez-González MA, et al. Olive oil intake and risk of cardiovascular disease and mortality in the PREDIMED Study. BMC Med. 2014;12:78. https://pubmed.ncbi.nlm.nih.gov/24886626/
Keys A. Olive oil and coronary heart disease. Lancet. 1987;1(8539):983–4. https://pubmed.ncbi.nlm.nih.gov/2882379/
Valls-Pedret C, Sala-Vila A, Serra-Mir M, et al. Mediterranean diet and age-related cognitive decline: a randomized clinical trial. JAMA Intern Med. 2015;175(7):1094–103. https://pubmed.ncbi.nlm.nih.gov/25961184/
Martínez-González MÁ, Toledo E, Arós F, et al. Extra-virgin olive oil consumption reduces risk of atrial fibrillation: the PREDIMED trial. Circulation. 2014;130(1):18–26. https://pubmed.ncbi.nlm.nih.gov/24787471/
Ruiz-Canela M, Estruch R, Corella D, Salas-Salvadó J, Martínez-González MA. Association of Mediterranean diet with peripheral artery disease: the PREDIMED randomized trial. JAMA. 2014;311(4):415–7. https://pubmed.ncbi.nlm.nih.gov/24449321/
Salas-Salvadó J, Bulló M, Estruch R, et al. Prevention of diabetes with Mediterranean diets: a subgroup analysis of a randomized trial. Ann Intern Med. 2014;160(1):1–10. https://pubmed.ncbi.nlm.nih.gov/24573661/
Díaz-López A, Babio N, Martínez-González MA, et al. Erratum. Mediterranean diet, retinopathy, nephropathy, and microvascular diabetes complications: a post hoc analysis of a randomized trial. Diabetes Care 2015;38:2134–2141. Diabetes Care. 2018;41(10):2260–1. https://pubmed.ncbi.nlm.nih.gov/26370380/
Martínez-Lapiscina EH, Clavero P, Toledo E, et al. Virgin olive oil supplementation and long-term cognition: the PREDIMED-NAVARRA randomized, trial. J Nutr Health Aging. 2013;17(6):544–52. https://pubmed.ncbi.nlm.nih.gov/23732551/
Toledo E, Salas-Salvadó J, Donat-Vargas C, et al. Mediterranean diet and invasive breast cancer risk among women at high cardiovascular risk in the PREDIMED trial: a randomized clinical trial. JAMA Intern Med. 2015;175(11):1752–60. https://pubmed.ncbi.nlm.nih.gov/26365989/
Bogani P, Galli C, Villa M, Visioli F. Postprandial anti-inflammatory and antioxidant effects of extra virgin olive oil. Atherosclerosis. 2007;190(1):181–6. https://pubmed.ncbi.nlm.nih.gov/16488419/
Visioli F, Caruso D, Galli C, Viappiani S, Galli G, Sala A. Olive oils rich in natural catecholic phenols decrease isoprostane excretion in humans. Biochem Biophys Res Commun. 2000;278(3):797–9. https://pubmed.ncbi.nlm.nih.gov/11095986/
Bucciantini M, Leri M, Nardiello P, Casamenti F, Stefani M. Olive polyphenols: antioxidant and anti-inflammatory properties. Antioxidants (Basel). 2021;10(7):1044. https://pubmed.ncbi.nlm.nih.gov/34209636/
Tiong SH, Saparin N, Teh HF, et al. Natural organochlorines as precursors of 3-monochloropropanediol esters in vegetable oils. J Agric Food Chem. 2018;66(4):999–1007. https://pubmed.ncbi.nlm.nih.gov/29260544/
Gao B, Li Y, Huang G, Yu L. Fatty acid esters of 3-monochloropropanediol: a review. Annu Rev Food Sci Technol. 2019;10:259–84. https://pubmed.ncbi.nlm.nih.gov/30908955/
Yan J, Oey SB, van Leeuwen SPJ, van Ruth SM. Discrimination of processing grades of olive oil and other vegetable oils by monochloropropanediol esters and glycidyl esters. Food Chem. 2018;248:93–100. https://pubmed.ncbi.nlm.nih.gov/29329876/
Mossoba MM, Azizian H, Fardin-Kia AR, Karunathilaka SR, Kramer JKG. First application of newly developed FT-NIR spectroscopic methodology to predict authenticity of extra virgin olive oil retail products in the USA. Lipids. 2017;52(5):443–55. https://pubmed.ncbi.nlm.nih.gov/28401382/
Frankel EN, Mailed RJ, Wang SC, et al. Evaluation of extra-virgin olive oil sold in California. UC Davis Olive Center. https://olivecenter.ucdavis.edu/media/files/report2011three.pdf. Published April 2011. Accessed December 28, 2021.; https://issuu.com/oliveoiltimes/docs/report_041211_final_reduced
Martínez-González MA, Gea A, Ruiz-Canela M. The Mediterranean diet and cardiovascular health. Circ Res. 2019;124(5):779–98. https://pubmed.ncbi.nlm.nih.gov/30817261/
Huedo-Medina TB, Garcia M, Bihuniak JD, Kenny A, Kerstetter J. Methodologic quality of meta-analyses and systematic reviews on the Mediterranean diet and cardiovascular disease outcomes: a review. Am J Clin Nutr. 2016;103(3):841–50. https://pubmed.ncbi.nlm.nih.gov/26864357/
Galbete C, Schwingshackl L, Schwedhelm C, Boeing H, Schulze MB. Evaluating Mediterranean diet and risk of chronic disease in cohort studies: an umbrella review of meta-analyses. Eur J Epidemiol. 2018;33(10):909–31. https://pubmed.ncbi.nlm.nih.gov/30030684/
Martínez-González MA, Gea A, Ruiz-Canela M. The Mediterranean diet and cardiovascular health. Circ Res. 2019;124(5):779–98. https://pubmed.ncbi.nlm.nih.gov/30817261/
Galbete C, Schwingshackl L, Schwedhelm C, Boeing H, Schulze MB. Evaluating Mediterranean diet and risk of chronic disease in cohort studies: an umbrella review of meta-analyses. Eur J Epidemiol. 2018;33(10):909–31. https://pubmed.ncbi.nlm.nih.gov/30030684/
Martínez-González MA, Gea A, Ruiz-Canela M. The Mediterranean diet and cardiovascular health. Circ Res. 2019;124(5):779–98. https://pubmed.ncbi.nlm.nih.gov/30817261/
White C. Suspected research fraud: difficulties of getting at the truth. BMJ. 2005;331(7511):281–8. https://pubmed.ncbi.nlm.nih.gov/16052022/
Horton R. Expression of concern: Indo-Mediterranean diet heart study. Lancet. 2005;366(9483):354–6. https://pubmed.ncbi.nlm.nih.gov/16054927/
de Lorgeril M, Renaud S, Mamelle N, et al. Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet. 1994;343(8911):1454–9. https://pubmed.ncbi.nlm.nih.gov/7911176/
Simopoulos AP. Omega-3 fatty acids and antioxidants in edible wild plants. Biol Res. 2004;37(2):263–77. https://pubmed.ncbi.nlm.nih.gov/15455656/
Pourrajab B, Sharifi-Zahabi E, Soltani S, Shahinfar H, Shidfar F. Comparison of canola oil and olive oil consumption on the serum lipid profile in adults: a systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr. Published online July 22, 2022:1–15.; https://pubmed.ncbi.nlm.nih.gov/35866510/
Vogel RA, Corretti MC, Plotnick GD. The postprandial effect of components of the Mediterranean diet on endothelial function. J Am Coll Cardiol. 2000;36(5):1455–60. https://pubmed.ncbi.nlm.nih.gov/11079642/
de Lorgeril M, Renaud S, Mamelle N, et al. Mediterranean alpha-linolenic acid-rich diet in secondary prevention of coronary heart disease. Lancet. 1994;343(8911):1454–9. https://pubmed.ncbi.nlm.nih.gov/7911176/
de Lorgeril M, Salen P, Martin JL, Monjaud I, Delaye J, Mamelle N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study. Circulation. 1999;99(6):779–85. https://pubmed.ncbi.nlm.nih.gov/9989963/
Esselstyn CB, Gendy G, Doyle J, Golubic M, Roizen MF. A way to reverse CAD? J Fam Pract. 2014;63(7):356–64b. https://pubmed.ncbi.nlm.nih.gov/25198208/
Rimm EB, Stampfer MJ. Diet, lifestyle, and longevity – the next steps? JAMA. 2004;292(12):1490–2. https://pubmed.ncbi.nlm.nih.gov/15383521/
Drewnowski A, Hill JO, Wansink B, Murray R, Diekman C. Achieve better health with nutrient-rich foods. Nutr Today. 2012;47(1):23–9. https://journals.lww.com/nutritiontodayonline/Abstract/2012/01000/Achieve_Better_Health_With_Nutrient_Rich_Foods.5.aspx
Willcox DC, Willcox BJ, Todoriki H, Suzuki M. The Okinawan diet: health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J Am Coll Nutr. 2009;28 Suppl:500S-16S. https://pubmed.ncbi.nlm.nih.gov/20234038/
Willcox DC, Willcox BJ, He Q, Wang NC, Suzuki M. They really are that old: a validation study of centenarian prevalence in Okinawa. J Gerontol A Biol Sci Med Sci. 2008;63(4):338–49. https://pubmed.ncbi.nlm.nih.gov/18426957/
Shao A, Drewnowski A, Willcox DC, et al. Optimal nutrition and the ever-changing dietary landscape: a conference report. Eur J Nutr. 2017;56(Suppl 1):1–21. https://pubmed.ncbi.nlm.nih.gov/28474121/
Willcox DC, Scapagnini G, Willcox BJ. Healthy aging diets other than the Mediterranean: a focus on the Okinawan diet. Mech Ageing Dev. 2014;136–137:148–62. https://pubmed.ncbi.nlm.nih.gov/24462788/
Willcox BJ, Willcox DC, Todoriki H, et al. Caloric restriction, the traditional Okinawan diet, and healthy aging: the diet of the world’s longest-lived people and its potential impact on morbidity and life span. Ann N Y Acad Sci. 2007;1114:434–55. https://pubmed.ncbi.nlm.nih.gov/17986602/
Willcox BJ, Willcox DC, Todoriki H, et al. Caloric restriction, the traditional Okinawan diet, and healthy aging: the diet of the world’s longest-lived people and its potential impact on morbidity and life span. Ann N Y Acad Sci. 2007;1114:434–55. https://pubmed.ncbi.nlm.nih.gov/17986602/
Willcox DC, Willcox BJ, Todoriki H, Suzuki M. The Okinawan diet: health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J Am Coll Nutr. 2009;28 Suppl:500S-16S. https://pubmed.ncbi.nlm.nih.gov/20234038/
Suzuki M, Willcox DC, Rosenbaum MW, Willcox BJ. Oxidative stress and longevity in Okinawa: an investigation of blood lipid peroxidation and tocopherol in Okinawan centenarians. Curr Gerontol Geriatr Res. 2010;2010:380460. https://pubmed.ncbi.nlm.nih.gov/21490698/
Suzuki M, Wilcox BJ, Wilcox CD. Implications from and for food cultures for cardiovascular disease: longevity. Asia Pac J Clin Nutr. 2001;10(2):165–71. https://pubmed.ncbi.nlm.nih.gov/11710359/
Willcox DC, Willcox BJ, Todoriki H, Suzuki M. The Okinawan diet: health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J Am Coll Nutr. 2009;28 Suppl:500S-16S. https://pubmed.ncbi.nlm.nih.gov/20234038/
Willcox DC, Scapagnini G, Willcox BJ. Healthy aging diets other than the Mediterranean: a focus on the Okinawan diet. Mech Ageing Dev. 2014;136–7:148–62. https://pubmed.ncbi.nlm.nih.gov/24462788/
Willcox BJ, Willcox DC. Caloric restriction, caloric restriction mimetics, and healthy aging in Okinawa: controversies and clinical implications. Curr Opin Clin Nutr Metab Care. 2014;17(1):51–8. https://pubmed.ncbi.nlm.nih.gov/24316687/
Chen X, Jiao J, Zhuang P, et al. Current intake levels of potatoes and all-cause mortality in China: a population-based nationwide study. Nutrition. 2021;81:110902. https://pubmed.ncbi.nlm.nih.gov/32739659/
Center for Science in the Public Interest. 10 Best Foods. https://cspinet.org/eating-healthy/what-eat/10-best-foods. Accessed January 5, 2022.; https://cspinet.org/eating-healthy/what-eat/10-best-foods
Wilson CD, Pace RD, Bromfield E, Jones G, Lu JY. Consumer acceptance of vegetarian sweet potato products intended for space missions. Life Support Biosph Sci. 1998;5(3):339–46. https://pubmed.ncbi.nlm.nih.gov/11876201/
Drewnowski A. New metrics of affordable nutrition: which vegetables provide most nutrients for least cost? J Acad Nutr Diet. 2013;113(9):1182–7. https://pubmed.ncbi.nlm.nih.gov/23714199/
Sunthonkun P, Palajai R, Somboon P, Suan CL, Ungsurangsri M, Soontorngun N. Life-span extension by pigmented rice bran in the model yeast Saccharomyces cerevisiae. Sci Rep. 2019;9(1):18061. https://pubmed.ncbi.nlm.nih.gov/31792269/
Chen W, Müller D, Richling E, Wink M. Anthocyanin-rich purple wheat prolongs the life span of Caenorhabditis elegans probably by activating the DAF-16/FOXO transcription factor. J Agric Food Chem. 2013;61(12):3047–53. https://pubmed.ncbi.nlm.nih.gov/23470220/
Zuo Y, Peng C, Liang Y, et al. Black rice extract extends the lifespan of fruit flies. Food Funct. 2012;3(12):1271–9. https://pubmed.ncbi.nlm.nih.gov/22930061/
Lu X, Zhou Y, Wu T, Hao L. Ameliorative effect of black rice anthocyanin on senescent mice induced by D-galactose. Food Funct. 2014;5(11):2892–7. https://pubmed.ncbi.nlm.nih.gov/25190075/
Kano M, Takayanagi T, Harada K, Makino K, Ishikawa F. Antioxidative activity of anthocyanins from purple sweet potato, Ipomoera batatas cultivar Ayamurasaki. Biosci Biotechnol Biochem. 2005;69(5):979–88. https://pubmed.ncbi.nlm.nih.gov/15914919/
Majid M, Nasir B, Zahra SS, Khan MR, Mirza B, Haq I. Ipomoea batatas L. Lam. ameliorates acute and chronic inflammations by suppressing inflammatory mediators, a comprehensive exploration using in vitro and in vivo models. BMC Complement Altern Med. 2018;18(1):216. https://pubmed.ncbi.nlm.nih.gov/30005651/
Wang YJ, Zheng YL, Lu J, et al. Purple sweet potato color suppresses lipopolysaccharide-induced acute inflammatory response in mouse brain. Neurochem Int. 2010;56(3):424–30. https://pubmed.ncbi.nlm.nih.gov/19941923/
Wu DM, Lu J, Zheng YL, Zhou Z, Shan Q, Ma DF. Purple sweet potato color repairs D-galactose-induced spatial learning and memory impairment by regulating the expression of synaptic proteins. Neurobiol Learn Mem. 2008;90(1):19–27. https://pubmed.ncbi.nlm.nih.gov/18316211/
Sun C, Diao Q, Lu J, et al. Purple sweet potato color attenuated NLRP3 inflammasome by inducing autophagy to delay endothelial senescence. J Cell Physiol. 2019;234(5):5926–39. https://pubmed.ncbi.nlm.nih.gov/30585631/
Su W, Zhang C, Chen F, et al. Purple sweet potato color protects against hepatocyte apoptosis through Sirt1 activation in high-fat-diet-treated mice. Food Nutr Res. 2020;64:10.29219/fnr.v64.1509. https://pubmed.ncbi.nlm.nih.gov/32110174/
Han Y, Guo Y, Cui SW, Li H, Shan Y, Wang H. Purple Sweet Potato Extract extends lifespan by activating autophagy pathway in male Drosophila melanogaster. Exp Gerontol. 2021;144:111190. https://pubmed.ncbi.nlm.nih.gov/33301922/
Zhang X, Yang Y, Wu Z, Weng P. The modulatory effect of anthocyanins from purple sweet potato on human intestinal microbiota in vitro. J Agric Food Chem. 2016;64(12):2582–90. https://pubmed.ncbi.nlm.nih.gov/26975278/
Suda I, Ishikawa F, Hatakeyama M, et al. Intake of purple sweet potato beverage affects on serum hepatic biomarker levels of healthy adult men with borderline hepatitis. Eur J Clin Nutr. 2008;62(1):60–7. https://pubmed.ncbi.nlm.nih.gov/17299464/
Willcox DC, Willcox BJ, Todoriki H, Suzuki M. The Okinawan diet: health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J Am Coll Nutr. 2009;28(sup4):500S-16S. https://pubmed.ncbi.nlm.nih.gov/20234038/
Shi Z, Zhang T, Byles J, Martin S, Avery JC, Taylor AW. Food habits, lifestyle factors and mortality among oldest old Chinese: the Chinese Longitudinal Healthy Longevity Survey (CLHLS). Nutrients. 2015;7(9):7562–79. https://pubmed.ncbi.nlm.nih.gov/26371039/
Mejia SB, Messina M, Li SS, et al. A meta-analysis of 46 studies identified by the FDA demonstrates that soy protein decreases circulating LDL and total cholesterol concentrations in adults. J Nutr. 2019;149(6):968–81. https://pubmed.ncbi.nlm.nih.gov/31006811/
Mosallanezhad Z, Mahmoodi M, Ranjbar S, et al. Soy intake is associated with lowering blood pressure in adults: a systematic review and meta-analysis of randomized double-blind placebo-controlled trials. Complement Ther Med. 2021;59:102692. https://pubmed.ncbi.nlm.nih.gov/33636295/
Bazzano LA, Thompson AM, Tees MT, Nguyen CH, Winham DM. Non-soy legume consumption lowers cholesterol levels: a meta-analysis of randomized controlled trials. Nutr Metab Cardiovasc Dis. 2011;21(2):94–103. https://pubmed.ncbi.nlm.nih.gov/19939654/
Mejia SB, Messina M, Li SS, et al. A meta-analysis of 46 studies identified by the FDA demonstrates that soy protein decreases circulating LDL and total cholesterol concentrations in adults. J Nutr. 2019;149(6):968–81. https://pubmed.ncbi.nlm.nih.gov/31006811/
Tokede OA, Onabanjo TA, Yansane A, Gaziano JM, Djoussé L. Soya products and serum lipids: a meta-analysis of randomised controlled trials. Br J Nutr. 2015;114(6):831–43. https://pubmed.ncbi.nlm.nih.gov/21559039/
Yan Z, Zhang X, Li C, Jiao S, Dong W. Association between consumption of soy and risk of cardiovascular disease: a meta-analysis of observational studies. Eur J Prev Cardiol. 2017;24(7):735–47. https://pubmed.ncbi.nlm.nih.gov/28067550/
Nachvak SM, Moradi S, Anjom-Shoae J, et al. Soy, soy isoflavones, and protein intake in relation to mortality from all causes, cancers, and cardiovascular diseases: a systematic review and dose-response meta-analysis of prospective cohort studies. J Acad Nutr Diet. 2019;119(9):1483–1500.e17. https://pubmed.ncbi.nlm.nih.gov/31278047/
D’elia L, Rossi G, Ippolito R, Cappuccio FP, Strazzullo P. Habitual salt intake and risk of gastric cancer: a meta-analysis of prospective studies. Clin Nutr. 2012;31(4):489–98. https://pubmed.ncbi.nlm.nih.gov/22296873/
Kanda A, Hoshiyama Y, Kawaguchi T. Association of lifestyle parameters with the prevention of hypertension in elderly Japanese men and women: a four-year follow-up of normotensive subjects. Asia Pac J Public Health. 1999;11(2):77–81. https://pubmed.ncbi.nlm.nih.gov/11195162/
Ito K. Review of the health benefits of habitual consumption of miso soup: focus on the effects on sympathetic nerve activity, blood pressure, and heart rate. Environ Health Prev Med. 2020;25(1):45. https://pubmed.ncbi.nlm.nih.gov/32867671/
Kondo H, Tomari HS, Yamakawa S, et al. Long-term intake of miso soup decreases nighttime blood pressure in subjects with high-normal blood pressure or stage I hypertension. Hypertens Res. 2019;42(11):1757–67. https://pubmed.ncbi.nlm.nih.gov/31371810/
Du DD, Yoshinaga M, Sonoda M, Kawakubo K, Uehara Y. Blood pressure reduction by Japanese traditional Miso is associated with increased diuresis and natriuresis through dopamine system in Dahl salt-sensitive rats. Clin Exp Hypertens. 2014;36(5):359–66. https://pubmed.ncbi.nlm.nih.gov/24047246/
Willcox BJ, Willcox DC. Caloric restriction, caloric restriction mimetics, and healthy aging in Okinawa: controversies and clinical implications. Curr Opin Clin Nutr Metab Care. 2014;17(1):51–8. https://pubmed.ncbi.nlm.nih.gov/24316687/
Iso H, Kubota Y. Nutrition and disease in the Japan Collaborative Cohort Study for evaluation of cancer (JACC). Asian Pac J Cancer Prev. 2007;8 Suppl:35–80. https://pubmed.ncbi.nlm.nih.gov/18260705/
Lashmanova E, Proshkina E, Zhikrivetskaya S, et al. Fucoxanthin increases lifespan of Drosophila melanogaster and Caenorhabditis elegans. Pharmacol Res. 2015;100:228–41. https://pubmed.ncbi.nlm.nih.gov/26292053/
Zhao T, Zhang Q, Qi H, Liu X, Li Z. Extension of life span and improvement of vitality of Drosophila melanogaster by long-term supplementation with different molecular weight polysaccharides from Porphyra haitanensis. Pharmacol Res. 2008;57(1):67–72. https://pubmed.ncbi.nlm.nih.gov/18221885/
Wada K, Nakamura K, Tamai Y, et al. Seaweed intake and blood pressure levels in healthy pre-school Japanese children. Nutr J. 2011;10:83. https://pubmed.ncbi.nlm.nih.gov/21827710/
Ono A, Shibaoka M, Yano J, Asai Y, Fujita T. Eating habits and intensity of medication in elderly hypertensive outpatients. Hypertens Res. 2000;23(3):195–200. https://pubmed.ncbi.nlm.nih.gov/10821126/
Teas J, Baldeón ME, Chiriboga DE, Davis JR, Sarriés AJ, Braverman LE. Could dietary seaweed reverse the metabolic syndrome? Asia Pac J Clin Nutr. 2009;18(2):145–54. https://pubmed.ncbi.nlm.nih.gov/19713172/
Ma W, He X, Braverman L. Iodine content in milk alternatives. Thyroid. 2016;26(9):1308–10. https://pubmed.ncbi.nlm.nih.gov/27358189/
Flachowsky G, Franke K, Meyer U, Leiterer M, Schöne F. Influencing factors on iodine content of cow milk. Eur J Nutr. 2014;53(2):351–65. https://pubmed.ncbi.nlm.nih.gov/24185833/
Teas J, Pino S, Critchley A, Braverman LE. Variability of iodine content in common commercially available edible seaweeds. Thyroid. 2004;14(10):836–41. https://pubmed.ncbi.nlm.nih.gov/15588380/
Combet E. Iodine status, thyroid function, and vegetarianism. In: Vegetarian and Plant-Based Diets in Health and Disease Prevention. Elsevier; 2017:769–90. https://worldcat.org/title/988275855
Willcox DC, Willcox BJ, Todoriki H, Suzuki M. The Okinawan diet: health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J Am Coll Nutr. 2009;28 Suppl:500S-16S. https://pubmed.ncbi.nlm.nih.gov/20234038/
Sánchez JE, Jiménez-Pérez G, Liedo P. Can consumption of antioxidant rich mushrooms extend longevity?: antioxidant activity of Pleurotus spp. and its effects on Mexican fruit flies’ (Anastrepha ludens) longevity. Age (Dordr). 2015;37(6):107. https://pubmed.ncbi.nlm.nih.gov/26499817/
Beelman RB, Kalaras MD, Phillips AT, Richie JP. Is ergothioneine a ‘longevity vitamin’ limited in the American diet? J Nutr Sci. 2020;9:e52. https://pubmed.ncbi.nlm.nih.gov/33244403/
Beelman RB, Kalaras MD, Phillips AT, Richie JP. Is ergothioneine a ‘longevity vitamin’ limited in the American diet? J Nutr Sci. 2020;9:e52. https://pubmed.ncbi.nlm.nih.gov/33244403/
Ames BN. Prolonging healthy aging: longevity vitamins and proteins. Proc Natl Acad Sci U S A. 2018;115(43):10836–44. https://pubmed.ncbi.nlm.nih.gov/30322941/
Smith E, Ottosson F, Hellstrand S, et al. Ergothioneine is associated with reduced mortality and decreased risk of cardiovascular disease. Heart. 2020;106(9):691–7. https://pubmed.ncbi.nlm.nih.gov/31672783/
Paul BD, Snyder SH. The unusual amino acid L-ergothioneine is a physiologic cytoprotectant. Cell Death Differ. 2010;17(7):1134–40. https://pubmed.ncbi.nlm.nih.gov/19911007/
Beelman RB, Kalaras MD, Phillips AT, Richie JP. Is ergothioneine a ‘longevity vitamin’ limited in the American diet? J Nutr Sci. 2020;9:e52. https://pubmed.ncbi.nlm.nih.gov/33244403/
Beelman RB, Kalaras MD, Richie JP. Micronutrients and bioactive compounds in mushrooms: a recipe for healthy aging? Nutr Today. 2019;54(1):16–22. https://journals.lww.com/nutritiontodayonline/Abstract/2019/01000/Micronutrients_and_Bioactive_Compounds_in.5.aspx
Ba DM, Gao X, Al-Shaar L, et al. Prospective study of dietary mushroom intake and risk of mortality: results from continuous National Health and Nutrition Examination Survey (NHANES) 2003–2014 and a meta-analysis. Nutr J. 2021;20(1):80. https://pubmed.ncbi.nlm.nih.gov/34548082/
Cheah IK, Feng L, Tang RMY, Lim KHC, Halliwell B. Ergothioneine levels in an elderly population decrease with age and incidence of cognitive decline; a risk factor for neurodegeneration? Biochem Biophys Res Commun. 2016;478(1):162–7. https://pubmed.ncbi.nlm.nih.gov/27444382/
Kameda M, Teruya T, Yanagida M, Kondoh H. Frailty markers comprise blood metabolites involved in antioxidation, cognition, and mobility. Proc Natl Acad Sci U S A. 2020;117(17):9483–9. https://pubmed.ncbi.nlm.nih.gov/32295884/
Cheah IK, Feng L, Tang RMY, Lim KHC, Halliwell B. Ergothioneine levels in an elderly population decrease with age and incidence of cognitive decline; a risk factor for neurodegeneration? Biochem Biophys Res Commun. 2016;478(1):162–7. https://pubmed.ncbi.nlm.nih.gov/27444382/
Lagrange E, Vernoux JP. Warning on false or true morels and button mushrooms with potential toxicity linked to hydrazinic toxins: an update. Toxins (Basel). 2020;12(8):482. https://pubmed.ncbi.nlm.nih.gov/32751277/
Heer RS, Patel NB, Mandal AKJ, Lewis F, Missouris CG. Not a fungi to be with: shiitake mushroom flagellate dermatitis. Am J Emerg Med. 2020;38(2):412.e1–2. https://pubmed.ncbi.nlm.nih.gov/31864870/
Stijve T, Pittet A. Absence of agaritine in Pleurotus species and in other cultivated and wild-growing mushrooms not belonging to the genus Agaricus. Dtsch Lebensm-Rundsch. 2000;96(7):251–4. https://www.researchgate.net/publication/286669322_Absence_of_Agaritine_in_Pleurotus_species_and_in_other_cultivated_and_wild-growing_mushrooms_not_belonging_to_the_genus_Agaricus
Money NP. Are mushrooms medicinal? Fungal Biol. 2016;120(4):449–53. https://pubmed.ncbi.nlm.nih.gov/27020147/
Money NP. Are mushrooms medicinal? Fungal Biol. 2016;120(4):449–53. https://pubmed.ncbi.nlm.nih.gov/27020147/
Litten W. The most poisonous mushrooms. Sci Am. 1975;232(3):90–101. https://pubmed.ncbi.nlm.nih.gov/1114308/
Lim CS, Chhabra N, Leikin S, Fischbein C, Mueller GM, Nelson ME. Atlas of select poisonous plants and mushrooms. Dis Mon. 2016;62(3):41–66. https://pubmed.ncbi.nlm.nih.gov/26965743/
Грибы рода Amanita. В Европе это Amanita virosa, а в восточной и западной части Северной Америки – A. bisporigera и A. ocreata. В России известен как бледная поганка (Amanita phalloides). – Примеч. ред.
Culliton BJ. The destroying angel: a story of a search for an antidote. Science. 1974;185(4151):600–1. https://pubmed.ncbi.nlm.nih.gov/17791229/
Loyd AL, Richter BS, Jusino MA, et al. Identifying the “mushroom of immortality”: assessing the Ganoderma species composition in commercial reishi products. Front Microbiol. 2018;9:1557. https://pubmed.ncbi.nlm.nih.gov/30061872/
Wang J, Cao B, Zhao H, Feng J. Emerging roles of Ganoderma lucidum in anti-aging. Aging Dis. 2017;8(6):691–707. https://pubmed.ncbi.nlm.nih.gov/29344411/
Pan Y, Lin Z. Anti-aging effect of Ganoderma (Lingzhi) with health and fitness. Adv Exp Med Bio. 2019;1182:299–309. https://pubmed.ncbi.nlm.nih.gov/31777025/
Cuong VT, Chen W, Shi J, et al. The anti-oxidation and anti-aging effects of Ganoderma lucidum in Caenorhabditis elegans. Exp Gerontol. 2019;117:99–105. https://pubmed.ncbi.nlm.nih.gov/28750751/
Wang J, Cao B, Zhao H, Feng J. Emerging roles of Ganoderma lucidum in anti-aging. Aging Dis. 2017;8(6):691–707. https://pubmed.ncbi.nlm.nih.gov/29344411/
Hsu KD, Cheng KC. From nutraceutical to clinical trial: frontiers in Ganoderma development. App Microbiol Biotechnol. 2018;102(21). https://pubmed.ncbi.nlm.nih.gov/30182215/
Loyd AL, Richter BS, Jusino MA, et al. Identifying the “mushroom of immortality”: assessing the Ganoderma species composition in commercial reishi products. Front Microbiol. 2018;9:1557. https://pubmed.ncbi.nlm.nih.gov/30061872/
Loyd AL, Richter BS, Jusino MA, et al. Identifying the “mushroom of immortality”: assessing the Ganoderma species composition in commercial reishi products. Front Microbiol. 2018;9:1557. https://pubmed.ncbi.nlm.nih.gov/30061872/
Totelin L. When foods become remedies in ancient Greece: The curious case of garlic and other substances. J Ethnopharmacol. 2015;167:30–7. https://pubmed.ncbi.nlm.nih.gov/25173971/
Shi X, Lv Y, Mao C, et al. Garlic consumption and all-cause mortality among Chinese oldest-old individuals: a population-based cohort study. Nutrients. 2019;11(7):E1504. https://pubmed.ncbi.nlm.nih.gov/31262080/
Lau KK, Chan YH, Wong YK, et al. Garlic intake is an independent predictor of endothelial function in patients with ischemic stroke. J Nutr Health Aging. 2013;17(7):600–4. https://pubmed.ncbi.nlm.nih.gov/23933870/
Mahdavi-Roshan M, Mirmiran P, Arjmand M, Nasrollahzadeh J. Effects of garlic on brachial endothelial function and capacity of plasma to mediate cholesterol efflux in patients with coronary artery disease. Anatol J Cardiol. 2017;18(2):116–21. https://pubmed.ncbi.nlm.nih.gov/28554988/
Mahdavi-Roshan M, Zahedmehr A, Mohammad-Zadeh A, et al. Effect of garlic powder tablet on carotid intima-media thickness in patients with coronary artery disease: a preliminary randomized controlled trial. Nutr Health. 2013;22(2):143–55. https://pubmed.ncbi.nlm.nih.gov/25573347/
Shabani E, Sayemiri K, Mohammadpour M. The effect of garlic on lipid profile and glucose parameters in diabetic patients: a systematic review and meta-analysis. Prim Care Diabetes. 2019;13(1):28–42. https://pubmed.ncbi.nlm.nih.gov/30049636/
Xiong XJ, Wang PQ, Li SJ, Li XK, Zhang YQ, Wang J. Garlic for hypertension: a systematic review and meta-analysis of randomized controlled trials. Phytomedicine. 2015;22(3):352–61. https://pubmed.ncbi.nlm.nih.gov/25837272/
Atkin M, Laight D, Cummings MH. The effects of garlic extract upon endothelial function, vascular inflammation, oxidative stress and insulin resistance in adults with type 2 diabetes at high cardiovascular risk. A pilot double blind randomized placebo controlled trial. J Diabetes Complications. 2016;30(4):723–7. https://pubmed.ncbi.nlm.nih.gov/26954484/
Soleimani D, Paknahad Z, Askari G, Iraj B, Feizi A. Effect of garlic powder consumption on body composition in patients with nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled trial. Adv Biomed Res. 2016;5:2. https://pubmed.ncbi.nlm.nih.gov/26955623/
Shabani E, Sayemiri K, Mohammadpour M. The effect of garlic on lipid profile and glucose parameters in diabetic patients: a systematic review and meta-analysis. Prim Care Diabetes. 2019;13(1):28–42. https://pubmed.ncbi.nlm.nih.gov/30049636/
Rajan TV, Hein M, Porte P, Wikel S. A double-blinded, placebo-controlled trial of garlic as a mosquito repellant: a preliminary study. Med Vet Entomol. 2005;19(1):84–9. https://pubmed.ncbi.nlm.nih.gov/15752181/
Stjernberg L, Berglund J. Garlic as an insect repellent. JAMA. 2000;284(7):831. https://pubmed.ncbi.nlm.nih.gov/10938169/
Tunón H. Garlic as a tick repellent. JAMA. 2001;285(1):41–2. https://pubmed.ncbi.nlm.nih.gov/11150101/
Yusof YAM. Gingerol and its role in chronic diseases. Adv Exp Med Biol. 2016;929:177–207. https://pubmed.ncbi.nlm.nih.gov/27771925/
Liu J, Shi JZ, Yu LM, Goyer RA, Waalkes MP. Mercury in traditional medicines: is cinnabar toxicologically similar to common mercurials? Exp Biol Med (Maywood). 2008;233(7):810–7. https://pubmed.ncbi.nlm.nih.gov/18445765/
Anh NH, Kim SJ, Long NP, et al. Ginger on human health: a comprehensive systematic review of 109 randomized controlled trials. Nutrients. 2020;12(1):E157. https://pubmed.ncbi.nlm.nih.gov/31935866/
Bodagh MN, Maleki I, Hekmatdoost A. Ginger in gastrointestinal disorders: a systematic review of clinical trials. Food Sci Nutr. 2018;7(1):96–108. https://pubmed.ncbi.nlm.nih.gov/30680163/
Mowrey DB, Clayson DE. Motion sickness, ginger, and psychophysics. Lancet. 1982;1(8273):655–7. https://pubmed.ncbi.nlm.nih.gov/30680163/
Palatty PL, Haniadka R, Valder B, Arora R, Baliga MS. Ginger in the prevention of nausea and vomiting: a review. Crit Rev Food Sci Nutr. 2013;53(7):659–69. https://pubmed.ncbi.nlm.nih.gov/23638927/
Adib-Hajbaghery M, Hosseini FS. Investigating the effects of inhaling ginger essence on post-nephrectomy nausea and vomiting. Complement Ther Med. 2015;23(6):827–31. https://pubmed.ncbi.nlm.nih.gov/26645524/
Bartels EM, Folmer VN, Bliddal H, et al. Efficacy and safety of ginger in osteoarthritis patients: a meta-analysis of randomized placebo-controlled trials. Osteoarthritis Cartilage. 2015;23(1):13–21. https://pubmed.ncbi.nlm.nih.gov/25300574/
Khayat S, Kheirkhah M, Behboodi Moghadam Z, Fanaei H, Kasaeian A, Javadimehr M. Effect of treatment with ginger on the severity of premenstrual syndrome symptoms. ISRN Obstet Gynecol. 2014;2014:792708. https://pubmed.ncbi.nlm.nih.gov/24944825/
Ozgoli G, Goli M, Moattar F. Comparison of effects of ginger, mefenamic acid, and ibuprofen on pain in women with primary dysmenorrhea. J Altern Complement Med. 2009;15(2):129–32. https://pubmed.ncbi.nlm.nih.gov/19216660/
Martins LB, Rodrigues AMdS, Monteze NM, et al. Double-blind placebo-controlled randomized clinical trial of ginger (Zingiber officinale Rosc.) in the prophylactic treatment of migraine. Cephalalgia. 2020;40(1):88–95. https://pubmed.ncbi.nlm.nih.gov/29768938/
Chen L, Cai Z. The efficacy of ginger for the treatment of migraine: a meta-analysis of randomized controlled studies. Am J Emerg Med. 2021;46:567–71. https://pubmed.ncbi.nlm.nih.gov/33293189/
Pourmasoumi M, Hadi A, Rafie N, Najafgholizadeh A, Mohammadi H, Rouhani MH. The effect of ginger supplementation on lipid profile: a systematic review and meta-analysis of clinical trials. Phytomedicine. 2018;43:28–36. https://pubmed.ncbi.nlm.nih.gov/29747751/
Makhdoomi Arzati M, Mohammadzadeh Honarvar N, Saedisomeolia A, et al. The effects of ginger on fasting blood sugar, hemoglobin A1c, and lipid profiles in patients with type 2 diabetes. Int J Endocrinol Metab. 2017;15(4):e57927. https://pubmed.ncbi.nlm.nih.gov/29344037/
Hasani H, Arab A, Hadi A, Pourmasoumi M, Ghavami A, Miraghajani M. Does ginger supplementation lower blood pressure? A systematic review and meta-analysis of clinical trials. Phytother Res. 2019;33(6):1639–47. https://pubmed.ncbi.nlm.nih.gov/30972845/
Maharlouei N, Tabrizi R, Lankarani KB, et al. The effects of ginger intake on weight loss and metabolic profiles among overweight and obese subjects: a systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr. 2018:1–14.; https://pubmed.ncbi.nlm.nih.gov/29393665/
Morvaridzadeh M, Sadeghi E, Agah S, et al. Effect of ginger (Zingiber officinale) supplementation on oxidative stress parameters: a systematic review and meta-analysis. J Food Biochem. 2021;45(2):e13612. https://pubmed.ncbi.nlm.nih.gov/33458848/
Mazidi M, Gao HK, Rezaie P, Ferns GA. The effect of ginger supplementation on serum C-reactive protein, lipid profile and glycaemia: a systematic review and meta-analysis. Food Nutr Res. 2016;60:32613. https://pubmed.ncbi.nlm.nih.gov/27806832/
Choi JG, Kim SY, Jeong M, Oh MS. Pharmacotherapeutic potential of ginger and its compounds in age-related neurological disorders. Pharmacol Ther. 2018;182:56–69. https://pubmed.ncbi.nlm.nih.gov/28842272/
Bischoff-Kont I, Fürst R. Benefits of ginger and its constituent 6-shogaol in inhibiting inflammatory processes. Pharmaceuticals (Basel). 2021;14(6):571. https://pubmed.ncbi.nlm.nih.gov/34203813/
Teschke R, Xuan TD. Viewpoint: a contributory role of shell ginger (Alpinia zerumbet) for human longevity in Okinawa, Japan? Nutrients. 2018;10(2):166. https://pubmed.ncbi.nlm.nih.gov/29385084/
Upadhyay A, Chompoo J, Taira N, Fukuta M, Tawata S. Significant longevity-extending effects of Alpinia zerumbet leaf extract on the life span of Caenorhabditis elegans. Biosci Biotechnol Biochem. 2013;77(2):217–23. https://pubmed.ncbi.nlm.nih.gov/23391900/
Rasheed N. Ginger and its active constituents as therapeutic agents: recent perspectives with molecular evidences. Int J Health Sci (Qassim). 2020;14(6):1–3. https://pubmed.ncbi.nlm.nih.gov/33192225/
Lee EB, Kim JH, Kim YJ, et al. Lifespan-extending property of 6-shogaol from Zingiber officinale Roscoe in Caenorhabditis elegans. Arch Pharm Res. 2018;41(7):743–52. https://pubmed.ncbi.nlm.nih.gov/29978428/
Percival SS, Vanden Heuvel JP, Nieves CJ, Montero C, Migliaccio AJ, Meadors J. Bioavailability of herbs and spices in humans as determined by ex vivo inflammatory suppression and DNA strand breaks. J Am Coll Nutr. 2012;31(4):288–94. https://pubmed.ncbi.nlm.nih.gov/23378457/
Stepien K, Wojdyla D, Nowak K, Molon M. Impact of curcumin on replicative and chronological aging in the Saccharomyces cerevisiae yeast. Biogerontology. 2020;21(1):109–23. https://pubmed.ncbi.nlm.nih.gov/31659616/
Liao VHC, Yu CW, Chu YJ, Li WH, Hsieh YC, Wang TT. Curcumin-mediated lifespan extension in Caenorhabditis elegans. Mech Ageing Dev. 2011;132(10):480–7. https://pubmed.ncbi.nlm.nih.gov/21855561/
Suckow BK, Suckow MA. Lifespan extension by the antioxidant curcumin in Drosophila melanogaster. Int J Biomed Sci. 2006;2(4):402–5. https://pubmed.ncbi.nlm.nih.gov/23675008/
Kitani K, Osawa T, Yokozawa T. The effects of tetrahydrocurcumin and green tea polyphenol on the survival of male C57BL/6 mice. Biogerontology. 2007;8(5):567–73. https://pubmed.ncbi.nlm.nih.gov/17516143/
Lao CD, Ruffin MT IV, Normolle D, et al. Dose escalation of a curcuminoid formulation. BMC Complement Altern Med. 2006;6:10. https://pubmed.ncbi.nlm.nih.gov/16545122/
Bala K, Tripathy BC, Sharma D. Neuroprotective and anti-ageing effects of curcumin in aged rat brain regions. Biogerontology. 2006;7(2):81–9. https://pubmed.ncbi.nlm.nih.gov/16802111/
Percival SS, Vanden Heuvel JP, Nieves CJ, Montero C, Migliaccio AJ, Meadors J. Bioavailability of herbs and spices in humans as determined by ex vivo inflammatory suppression and DNA strand breaks. J Am Coll Nutr. 2012;31(4):288–94. https://pubmed.ncbi.nlm.nih.gov/23378457/
Percival SS, Vanden Heuvel JP, Nieves CJ, Montero C, Migliaccio AJ, Meadors J. Bioavailability of herbs and spices in humans as determined by ex vivo inflammatory suppression and DNA strand breaks. J Am Coll Nutr. 2012;31(4):288–94. https://pubmed.ncbi.nlm.nih.gov/23378457/
DiSilvestro RA, Joseph E, Zhao S, Bomser J. Diverse effects of a low dose supplement of lipidated curcumin in healthy middle aged people. Nutr J. 2012;11:79. https://pubmed.ncbi.nlm.nih.gov/23013352/
Rakha A, Rehman K, Babar Imran M, Shahid M, Jahan N. Mitigation of 131-I induced oxidative stress by supplementation of turmeric and green cardamom in thyroid patients. Int J Radiat Res. 2022;20(1):29–36. https://ijrr.com/article-1-4063-en.html
Thorogood M, Appleby PN, Key TJ, Mann J. Relation between body mass index and mortality in an unusually slim cohort. J Epidemiol Community Health. 2003;57(2):130–3. https://pubmed.ncbi.nlm.nih.gov/12540689/
Aune D, Sen A, Prasad M, et al. BMI and all cause mortality: systematic review and non-linear dose-response meta-analysis of 230 cohort studies with 3.74 million deaths among 30.3 million participants. BMJ. 2016;353:i2156. https://pubmed.ncbi.nlm.nih.gov/27146380/
Willcox DC, Willcox BJ, Todoriki H, Curb JD, Suzuki M. Caloric restriction and human longevity: what can we learn from the Okinawans? Biogerontology. 2006;7(3):173–7. https://pubmed.ncbi.nlm.nih.gov/16810568/
Willcox DC, Scapagnini G, Willcox BJ. Healthy aging diets other than the Mediterranean: a focus on the Okinawan diet. Mech Ageing Dev. 2014;136–7:148–62. https://pubmed.ncbi.nlm.nih.gov/24462788/
Willcox BJ, Willcox DC, Todoriki H, et al. Caloric restriction, the traditional Okinawan diet, and healthy aging: the diet of the world’s longest-lived people and its potential impact on morbidity and life span. Ann N Y Acad Sci. 2007;1114:434–55. https://pubmed.ncbi.nlm.nih.gov/17986602/
Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161(13):1645–52. https://pubmed.ncbi.nlm.nih.gov/11434797/
Willcox BJ, Willcox DC. Caloric restriction, caloric restriction mimetics, and healthy aging in Okinawa: controversies and clinical implications. Curr Opin Clin Nutr Metab Care. 2014;17(1):51–8. https://pubmed.ncbi.nlm.nih.gov/24316687/
Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161(13):1645–52. https://pubmed.ncbi.nlm.nih.gov/11434797/
Gavrilova NS, Gavrilov LA. Comments on dietary restriction, Okinawa diet and longevity. Gerontology. 2012;58(3):221–3. https://pubmed.ncbi.nlm.nih.gov/21893946/
Willcox DC, Willcox BJ, Todoriki H, Suzuki M. The Okinawan diet: health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J Am Coll Nutr. 2009;28 Suppl:500S-16S. https://pubmed.ncbi.nlm.nih.gov/20234038/
Willcox DC, Scapagnini G, Willcox BJ. Healthy aging diets other than the Mediterranean: a focus on the Okinawan diet. Mech Ageing Dev. 2014;136–7:148–62. https://pubmed.ncbi.nlm.nih.gov/24462788/
Martínez-González MA, Gea A, Ruiz-Canela M. The Mediterranean diet and cardiovascular health. Circ Res. 2019;124(5):779–98. https://pubmed.ncbi.nlm.nih.gov/30817261/
Marston HR, Niles-Yokum K, Silva PA. A commentary on Blue Zones®: a critical review of age-friendly environments in the 21st century and beyond. Int J Environ Res Public Health. 2021;18(2):837. https://pubmed.ncbi.nlm.nih.gov/33478140/
Willcox DC, Scapagnini G, Willcox BJ. Healthy aging diets other than the Mediterranean: a focus on the Okinawan diet. Mech Ageing Dev. 2014;136–7:148–62. https://pubmed.ncbi.nlm.nih.gov/24462788/
Cockerham WC, Yamori Y. Okinawa: an exception to the social gradient of life expectancy in Japan. Asia Pac J Clin Nutr. 2001;10(2):154–8. https://pubmed.ncbi.nlm.nih.gov/11710357/
Willcox DC, Willcox BJ, Todoriki H, Suzuki M. The Okinawan diet: health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load. J Am Coll Nutr. 2009;28(sup4):500S-16S. https://pubmed.ncbi.nlm.nih.gov/20234038/
Bajpai P. World’s 5 richest nations by GDP per capita. Nasdaq. https://www.nasdaq.com/articles/worlds-5-richest-nations-by-gdp-per-capita-2021–05–20. Published May 20, 2021. Accessed January 10, 2022.; https://www.nasdaq.com/articles/worlds-5-richest-nations-by-gdp-per-capita-2021-05-20
Robert L, Fulop T. Longevity and its regulation: centenarians and beyond. Interdiscip Top Gerontol. 2014;39:198–211. https://pubmed.ncbi.nlm.nih.gov/24862022/
Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161(13):1645–52. https://pubmed.ncbi.nlm.nih.gov/11434797/
Kent LM, Morton DP, Ward EJ, et al. The influence of religious affiliation on participant responsiveness to the Complete Health Improvement Program (CHIP) lifestyle intervention. J Relig Health. 2016;55(5):1561–73. https://pubmed.ncbi.nlm.nih.gov/26472654/
Fraser GE. Diet as primordial prevention in Seventh-Day Adventists. Prev Med. 1999;29(6):S18–23. https://pubmed.ncbi.nlm.nih.gov/10641813/
Orlich MJ, Chiu THT, Dhillon PK, et al. Vegetarian epidemiology: review and discussion of findings from geographically diverse cohorts. Adv Nutr. 2019;10(Suppl_4):S284–95. https://pubmed.ncbi.nlm.nih.gov/31728496/
Sloan RP, Bagiella E, Powell T. Religion, spirituality, and medicine. Lancet. 1999;353(9153):664–7. https://pubmed.ncbi.nlm.nih.gov/10030348/
Chida Y, Steptoe A, Powell LH. Religiosity/spirituality and mortality: a systematic quantitative review. Psychother Psychosom. 2009;78(2):81–90. https://pubmed.ncbi.nlm.nih.gov/19142047/
Sullivan AR. Mortality differentials and religion in the United States: religious affiliation and attendance. J Sci Study Relig. 2010;49(4):740–53. https://pubmed.ncbi.nlm.nih.gov/21318110/
Schnall E, Wassertheil-Smoller S, Swencionis C, et al. The relationship between religion and cardiovascular outcomes and all-cause mortality in the Women’s Health Initiative Observational Study. Psychol Health. 2010;25(2):249–63. https://pubmed.ncbi.nlm.nih.gov/20391218/
Hill TD, Ellison CG, Burdette AM, Taylor J, Friedman KL. Dimensions of religious involvement and leukocyte telomere length. Soc Sci Med. 2016;163:168–75. https://pubmed.ncbi.nlm.nih.gov/27174242/
Koenig HG, Nelson B, Shaw SF, Saxena S, Cohen HJ. Religious involvement and telomere length in women family caregivers. J Nerv Ment Dis. 2016;204(1):36–42. https://pubmed.ncbi.nlm.nih.gov/26669979/
Schnall E, Wassertheil-Smoller S, Swencionis C, et al. The relationship between religion and cardiovascular outcomes and all-cause mortality in the Women’s Health Initiative Observational Study. Psychol Health. 2010;25(2):249–63. https://pubmed.ncbi.nlm.nih.gov/20391218/
Sloan RP, Bagiella E, Powell T. Religion, spirituality, and medicine. Lancet. 1999;353(9153):664–7. https://pubmed.ncbi.nlm.nih.gov/10030348/
Morton D, Rankin P, Kent L, Dysinger W. The Complete Health Improvement Program (CHIP): history, evaluation, and outcomes. Am J Lifestyle Med. 2016;10(1):64–73. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6124862/
Kent LM, Morton DP, Ward EJ, et al. The influence of religious affiliation on participant responsiveness to the Complete Health Improvement Program (CHIP) lifestyle intervention. J Relig Health. 2016;55(5):1561–73. https://pubmed.ncbi.nlm.nih.gov/26472654/
World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen. 5th ed. WHO Press; 2010. https://apps.who.int/iris/handle/10665/44261
Orzylowska EM, Jacobson JD, Bareh GM, Ko EY, Corselli JU, Chan PJ. Food intake diet and sperm characteristics in a blue zone: a Loma Linda Study. Eur J Obstet Gynecol Reprod Biol. 2016;203:112–5. https://pubmed.ncbi.nlm.nih.gov/27280539/
Messina M, Watanabe S, Setchell KDR. Report on the 8th international symposium on the role of soy in health promotion and chronic disease prevention and treatment. J Nutr. 2009;139(4):796S-802S. https://pubmed.ncbi.nlm.nih.gov/19225130/
Zhang Y, Hood WR. Current versus future reproduction and longevity: a re-evaluation of predictions and mechanisms. J Exp Biol. 2016;219(Pt 20):3177–89. https://pubmed.ncbi.nlm.nih.gov/27802148/
Mukhopadhyay A, Tissenbaum HA. Reproduction and longevity: secrets revealed by C. elegans. Trends Cell Biol. 2007;17(2):65–71. https://pubmed.ncbi.nlm.nih.gov/17187981/
Hsin H, Kenyon C. Signals from the reproductive system regulate the lifespan of C. elegans. Nature. 1999;399(6734):362–6. https://pubmed.ncbi.nlm.nih.gov/10360574/
Flatt T, Min KJ, D’Alterio C, et al. Drosophila germ-line modulation of insulin signaling and lifespan. Proc Natl Acad Sci U S A. 2008;105(17):6368–73. https://pubmed.ncbi.nlm.nih.gov/18434551/
American Veterinary Medical Association. Banfield: spaying, neutering correlate with longer lives. JAVMA News. https://www.avma.org/javma-news/2013–07–01/banfield-spaying-neutering-correlate-longer-lives. Published June 19, 2013. Accessed January 10, 2022.; https://www.avma.org/javma-news/2013-07-01/banfield-spaying-neutering-correlate-longer-lives
Banfield Pet Hospital. State of Pet Health 2013 Report. Banfield.com. https://www.banfield.com/-/media/Project/Banfield/Main/en/general/SOPH-Infographic/PDFs/Banfield-State-of-Pet-Health-Report_2013.pdf?rev=a8612f3fa39141e3bf2876a5ed6760de&hash=D79B771D2C3539DF737353E65D310504. Accessed February 21, 2022.; https://www.banfield.com/en/pet-health/State-of-pet-health
Min KJ, Lee CK, Park HN. The lifespan of Korean eunuchs. Curr Biol. 2012;22(18):R792–3. https://pubmed.ncbi.nlm.nih.gov/23017989/
Reilly PR. Involuntary sterilization in the United States: a surgical solution. Q Rev Biol. 1987;62(2):153–70. https://pubmed.ncbi.nlm.nih.gov/3299450/
Buck v. Bell, 274 US 200 (1927).; https://supreme.justia.com/cases/federal/us/274/200/
Hamilton JB, Mestler GE. Mortality and survival: comparison of eunuchs with intact men and women in a mentally retarded population. J Gerontol. 1969;24(4):395–411. https://pubmed.ncbi.nlm.nih.gov/5362349/
Hsu CH, Posegga O, Fischbach K, Engelhardt H. Examining the trade-offs between human fertility and longevity over three centuries using crowdsourced genealogy data. PLoS One. 2021;16(8):e0255528. https://pubmed.ncbi.nlm.nih.gov/34351988/
Tabatabaie V, Atzmon G, Rajpathak SN, Freeman R, Barzilai N, Crandall J. Exceptional longevity is associated with decreased reproduction. Aging (Albany NY). 2011;3(12):1202–5. https://pubmed.ncbi.nlm.nih.gov/22199025/
Zwaan B, Bijlsma R, Hoekstra RF. Direct selection on life span in Drosophila melanogaster. Evolution. 1995;49(4):649–59. https://pubmed.ncbi.nlm.nih.gov/28565142/
Mukhopadhyay A, Tissenbaum HA. Reproduction and longevity: secrets revealed by C. elegans. Trends Cell Biol. 2007;17(2):65–71. https://pubmed.ncbi.nlm.nih.gov/17187981/
Franklin JC, Scheile BC, Brozek J, Keys A. Observations on human behavior in experimental semi-starvation and rehabilitation. J Clin Psychol. 1948;4(1):28–45. https://pubmed.ncbi.nlm.nih.gov/18903450/
Templeman NM, Murphy CT. Regulation of reproduction and longevity by nutrient-sensing pathways. J Cell Biol. 2018;217(1):93–106. https://pubmed.ncbi.nlm.nih.gov/29074705/
Chen X, Liu Y, Sun X, et al. Age at menarche and risk of all-cause and cardiovascular mortality: a systematic review and dose-response meta-analysis. Menopause. 2018;26(6):670–6. https://pubmed.ncbi.nlm.nih.gov/30562317/
Fuhrman BJ, Moore SC, Byrne C, et al. Association of the age at menarche with site-specific cancer risks in pooled data from nine cohorts. Cancer Res. 2021;81(8):2246–55. https://pubmed.ncbi.nlm.nih.gov/33820799/
Chen X, Liu Y, Sun X, et al. Age at menarche and risk of all-cause and cardiovascular mortality: a systematic review and dose-response meta-analysis. Menopause. 2018;26(6):670–6. https://pubmed.ncbi.nlm.nih.gov/30562317/
Goldberg M, D’Aloisio AA, O’Brien KM, Zhao S, Sandler DP. Pubertal timing and breast cancer risk in the Sister Study cohort. Breast Cancer Res. 2020;22(1):112. https://pubmed.ncbi.nlm.nih.gov/33109223/
Fuhrman BJ, Moore SC, Byrne C, et al. Association of the age at menarche with site-specific cancer risks in pooled data from nine cohorts. Cancer Res. 2021;81(8):2246–55. https://pubmed.ncbi.nlm.nih.gov/33820799/
Lee HS. Why should we be concerned about early menarche? Clin Exp Pediatr. 2020;64(1):26–7. https://pubmed.ncbi.nlm.nih.gov/32683812/
Martinez GM. Trends and patterns in menarche in the United States: 1995 through 2013–2017. Natl Health Stat Report. 2020;(146):1–12. https://pubmed.ncbi.nlm.nih.gov/33054923/
Eckert-Lind C, Busch AS, Petersen JH, et al. Worldwide secular trends in age at pubertal onset assessed by breast development among girls: a systematic review and meta-analysis. JAMA Pediatr. 2020;174(4):e195881. https://pubmed.ncbi.nlm.nih.gov/32040143/
Thankamony A, Ong KK, Ahmed ML, Ness AR, Holly JMP, Dunger DB. Higher levels of IGF-I and adrenal androgens at age 8 years are associated with earlier age at menarche in girls. J Clin Endocrinol Metab. 2012;97(5):E786–90. https://pubmed.ncbi.nlm.nih.gov/22419724/
Günther ALB, Karaolis-Danckert N, Kroke A, Remer T, Buyken AE. Dietary protein intake throughout childhood is associated with the timing of puberty. J Nutr. 2010;140(3):565–71. https://pubmed.ncbi.nlm.nih.gov/20042466/
Nguyen NTK, Fan HY, Tsai MC, et al. Nutrient intake through childhood and early menarche onset in girls: systematic review and meta-analysis. Nutrients. 2020;12(9):2544. https://pubmed.ncbi.nlm.nih.gov/32842616/
Rogers IS, Northstone K, Dunger DB, Cooper AR, Ness AR, Emmett PM. Diet throughout childhood and age at menarche in a contemporary cohort of British girls. Public Health Nutr. 2010;13(12):2052–63. https://pubmed.ncbi.nlm.nih.gov/20529402/
Jansen EC, Marín C, Mora-Plazas M, Villamor E. Higher childhood red meat intake frequency is associated with earlier age at menarche. J Nutr. 2015;146(4):792–8. https://pubmed.ncbi.nlm.nih.gov/26962195/
Schecter A, Cramer P, Boggess K, Stanley J, Olson JR. Levels of dioxins, dibenzofurans, PCB and DDE congeners in pooled food samples collected in 1995 at supermarkets across the United States. Chemosphere. 1997;34(5–7):1437–47. https://pubmed.ncbi.nlm.nih.gov/9134677/
Ouyang F, Perry MJ, Venners SA, et al. Serum DDT, age at menarche, and abnormal menstrual cycle length. Occup Environ Med. 2005;62(12):878–84. https://pubmed.ncbi.nlm.nih.gov/16299097/
Kahleova H, Levin S, Barnard ND. Plant-based diets for healthy aging. J Am Coll Nutr. 2021;40(5):478–9. https://pubmed.ncbi.nlm.nih.gov/32643581/
Fraser GE, Cosgrove CM, Mashchak AD, Orlich MJ, Altekruse SF. Lower rates of cancer and all-cause mortality in an Adventist cohort compared with a US Census population. Cancer. 2020;126(5):1102–11. https://pubmed.ncbi.nlm.nih.gov/31762009/
Dinu M, Abbate R, Gensini GF, Casini A, Sofi F. Vegetarian, vegan diets and multiple health outcomes: a systematic review with meta-analysis of observational studies. Crit Rev Food Sci Nutr. 2017;57(17):3640–9. https://pubmed.ncbi.nlm.nih.gov/26853923/
Singh PN, Arthur KN, Orlich MJ, et al. Global epidemiology of obesity, vegetarian dietary patterns, and noncommunicable disease in Asian Indians. Am J Clin Nutr. 2014;100 Suppl 1:359S-64S. https://pubmed.ncbi.nlm.nih.gov/24847857/
Singh PN, Sabaté J, Fraser GE. Does low meat consumption increase life expectancy in humans? Am J Clin Nutr. 2003;78(3 Suppl):526S-32S. https://pubmed.ncbi.nlm.nih.gov/12936945/
Giem P, Beeson WL, Fraser GE. The incidence of dementia and intake of animal products: preliminary findings from the Adventist Health Study. Neuroepidemiology. 1993;12(1):28–36. https://pubmed.ncbi.nlm.nih.gov/8327020/
Donner Y, Fortney K, Calimport SRG, Pfleger K, Shah M, Betts-LaCroix J. Great desire for extended life and health amongst the American public. Front Genet. 2015;6:353. https://pubmed.ncbi.nlm.nih.gov/26834780/
Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161(13):1645–52. https://pubmed.ncbi.nlm.nih.gov/11434797/
Lin CL, Wang JH, Chang CC, Chiu THT, Lin MN. Vegetarian diets and medical expenditure in Taiwan – a matched cohort study. Nutrients. 2019;11(11):E2688. https://pubmed.ncbi.nlm.nih.gov/32102976/
Kahleova H, Hrachovinova T, Hill M, et al. Vegetarian diet in type 2 diabetes – improvement in quality of life, mood and eating behaviour. Diabet Med. 2013;30(1):127–9. https://pubmed.ncbi.nlm.nih.gov/23050853/
Trapp C, Barnard N, Katcher H. A plant-based diet for type 2 diabetes. Diabetes Educ. 2010;36(1):33–48. https://pubmed.ncbi.nlm.nih.gov/20185610/
Barnard N, Scialli AR, Bertron P, Hurlick D, Edmondset K. Acceptability of a therapeutic low-fat, vegan diet in premenopausal women. J Nutr Educ. 2000;32(6):314–9. https://www.researchgate.net/publication/223584518_Acceptability_of_a_Therapeutic_Low-Fat_Vegan_Diet_in_Premenopausal_Women
Trapp C, Barnard N, Katcher H. A plant-based diet for type 2 diabetes. Diabetes Educ. 2010;36(1):33–48. https://pubmed.ncbi.nlm.nih.gov/20185610/
Hemler EC, Hu FB. Plant-based diets for cardiovascular disease prevention: all plant foods are not created equal. Curr Atheroscler Rep. 2019;21(5):18. https://pubmed.ncbi.nlm.nih.gov/30895476/
Kim H, Caulfield LE, Garcia-Larsen V, Steffen LM, Coresh J, Rebholz CM. Plant-based diets are associated with a lower risk of incident cardiovascular disease, cardiovascular disease mortality, and all-cause mortality in a general population of middle-aged adults. J Am Heart Assoc. 2019;8(16):e012865. https://pubmed.ncbi.nlm.nih.gov/31387433/
Huang J, Liao LM, Weinstein SJ, Sinha R, Graubard BI, Albanes D. Association between plant and animal protein intake and overall and cause-specific mortality. JAMA Intern Med. 2020;180(9):1173–84. https://pubmed.ncbi.nlm.nih.gov/32658243/
Huang J, Liao LM, Weinstein SJ, Sinha R, Graubard BI, Albanes D. Association between plant and animal protein intake and overall and cause-specific mortality. JAMA Intern Med. 2020;180(9):1173–84. https://pubmed.ncbi.nlm.nih.gov/32658243/
Sinha R, Cross AJ, Graubard BI, Leitzmann MF, Schatzkin A. Meat intake and mortality: a prospective study of over half a million people. Arch Intern Med. 2009;169(6):562–71. https://pubmed.ncbi.nlm.nih.gov/19307518/
Popkin BM. Reducing meat consumption has multiple benefits for the world’s health. Arch Intern Med. 2009;169(6):543. https://pubmed.ncbi.nlm.nih.gov/19307515/
Huang J, Liao LM, Weinstein SJ, Sinha R, Graubard BI, Albanes D. Association between plant and animal protein intake and overall and cause-specific mortality. JAMA Intern Med. 2020;180(9):1173–84. https://pubmed.ncbi.nlm.nih.gov/32658243/
Ortolá R, Struijk EA, García-Esquinas E, Rodríguez-Artalejo F, Lopez-Garcia E. Changes in dietary intake of animal and vegetable protein and unhealthy aging. Am J Med. 2020;133(2):231–9. https://pubmed.ncbi.nlm.nih.gov/33839765/
Ortolá R, Struijk EA, García-Esquinas E, Rodríguez-Artalejo F, Lopez-Garcia E. Changes in dietary intake of animal and vegetable protein and unhealthy aging. Am J Med. 2020;133(2):231–9. https://pubmed.ncbi.nlm.nih.gov/33839765/
Norman K, Klaus S. Veganism, aging and longevity: new insight into old concepts. Curr Opin Clin Nutr Metab Care. 2020;23(2):145–50. https://pubmed.ncbi.nlm.nih.gov/31895244/
Eleftheriou D, Benetou V, Trichopoulou A, La Vecchia C, Bamia C. Mediterranean diet and its components in relation to all-cause mortality: meta-analysis. Br J Nutr. 2018;120(10):1081–97. https://pubmed.ncbi.nlm.nih.gov/30401007/
Martínez-González MA, Sánchez-Tainta A, Corella D, et al. A provegetarian food pattern and reduction in total mortality in the Prevención con Dieta Mediterránea (PREDIMED) study. Am J Clin Nutr. 2014;100 Suppl 1:320S-8S. https://pubmed.ncbi.nlm.nih.gov/24871477/
Tuso PJ, Ismail MH, Ha BP, Bartolotto C. Nutritional update for physicians: plant-based diets. Perm J. 2013;17(2):61–6. https://pubmed.ncbi.nlm.nih.gov/23704846/
Tuso PJ, Ismail MH, Ha BP, Bartolotto C. Nutritional update for physicians: plant-based diets. Perm J. 2013;17(2):61–6. https://pubmed.ncbi.nlm.nih.gov/23704846/
Kaiser Permanente. Plant-based eating: using the healthy plate to eat well. Center for Healthy Living. https://thrive.kaiserpermanente.org/care-near-you/southern-california/center-for-healthy-living/wp-content/uploads/sites/30/2020/03/plant_based_diet_e.pdf. Updated 2019. Accessed January 17, 2022.; https://thrive.kaiserpermanente.org/care-near-you/southern-california/center-for-healthy-living/wp-content/uploads/sites/30/2020/03/plant_based_diet_e.pdf
Martínez-González MÁ, Hershey MS, Zazpe I, Trichopoulou A. Transferability of the Mediterranean diet to non-Mediterranean countries. What is and what is not the Mediterranean diet. Nutrients. 2017;9(11):E1226. https://pubmed.ncbi.nlm.nih.gov/29117146/
Avital K, Buch A, Hollander I, Brickner T, Goldbourt U. Adherence to a Mediterranean diet by vegetarians and vegans as compared to omnivores. Int J Food Sci Nutr. 2020;71(3):378–87. https://pubmed.ncbi.nlm.nih.gov/31558068/
M Nestle. Mediterranean diets: historical and research overview. Am J Clin Nutr. 1995;61(6):1313S–20S. https://pubmed.ncbi.nlm.nih.gov/7754981/
Sofi F, Dinu M, Pagliai G, et al. Low-calorie vegetarian versus Mediterranean diets for reducing body weight and improving cardiovascular risk profile: CARDIVEG study (cardiovascular prevention with vegetarian diet). Circulation. 2018;137(11):1103–13. https://pubmed.ncbi.nlm.nih.gov/29483085/
Barnard ND, Alwarith J, Rembert E, et al. A Mediterranean diet and low-fat vegan diet to improve body weight and cardiometabolic risk factors: a randomized, cross-over trial. J Am Nutr Assoc. 2022;41(2):127–39. https://pubmed.ncbi.nlm.nih.gov/33544066/
Tuso PJ, Ismail MH, Ha BP, Bartolotto C. Nutritional update for physicians: plant-based diets. Perm J. 2013;17(2):61–6. https://pubmed.ncbi.nlm.nih.gov/23704846/
Lane MM, Davis JA, Beattie S, et al. Ultraprocessed food and chronic noncommunicable diseases: a systematic review and meta-analysis of 43 observational studies. Obes Rev. 2021;22(3):e13146. https://pubmed.ncbi.nlm.nih.gov/33167080/
Katz DL. Plant-based diets for reversing disease and saving the planet: past, present, and future. Adv Nutr. 2019;10(Suppl_4):S304–7. https://pubmed.ncbi.nlm.nih.gov/31728489/
Gehring J, Touvier M, Baudry J, et al. Consumption of ultra-processed foods by pesco-vegetarians, vegetarians, and vegans: associations with duration and age at diet initiation. J Nutr. 2021;151(1):120–31. https://pubmed.ncbi.nlm.nih.gov/32692345/
Radnitz C, Ni J, Dennis D, Cerrito B. Health benefits of a vegan diet: current insights. Nutr Diet Suppl. 2020;12:57–85. https://www.dovepress.com/health-benefits-of-a-vegan-diet-current-insights-peer-reviewed-fulltext-article-NDS
Neff RA, Edwards D, Palmer A, Ramsing R, Righter A, Wolfson J. Reducing meat consumption in the USA: a nationally representative survey of attitudes and behaviours. Public Health Nutr. 2018;21(10):1835–44. https://pubmed.ncbi.nlm.nih.gov/29576031/
Radnitz C, Ni J, Dennis D, Cerrito B. Health benefits of a vegan diet: current insights. Nutr Diet Suppl. 2020;12:57–85. https://www.dovepress.com/health-benefits-of-a-vegan-diet-current-insights-peer-reviewed-fulltext-article-NDS
Almost half of UK vegans made the change in the last year, according to new data. Vegan Trade Journal. https://www.vegantradejournal.com/almost-half-of-uk-vegans-made-the-change-in-the-last-year-according-to-new-data/. November 19, 2018. Accessed December 28, 2022.; https://www.vegantradejournal.com/almost-half-of-uk-vegans-made-the-change-in-the-last-year-according-to-new-data/
Radnitz C, Beezhold B, DiMatteo J. Investigation of lifestyle choices of individuals following a vegan diet for health and ethical reasons. Appetite. 2015;90:31–6. https://pubmed.ncbi.nlm.nih.gov/25725486/
Orlich MJ, Chiu THT, Dhillon PK, et al. Vegetarian epidemiology: review and discussion of findings from geographically diverse cohorts. Adv Nutr. 2019;10(Suppl_4):S284–95. https://pubmed.ncbi.nlm.nih.gov/31728496/
Rocha JP, Laster J, Parag B, Shah NU. Multiple health benefits and minimal risks associated with vegetarian diets. Curr Nutr Rep. 2019;8(4):374–81. https://pubmed.ncbi.nlm.nih.gov/31705483/
Singh PN, Arthur KN, Orlich MJ, et al. Global epidemiology of obesity, vegetarian dietary patterns, and noncommunicable disease in Asian Indians. Am J Clin Nutr. 2014;100 Suppl 1:359S-64S. https://pubmed.ncbi.nlm.nih.gov/24847857/
Campbell EK, Fidahusain M, Campbell TM II. Evaluation of an eight-week whole-food plant-based lifestyle modification program. Nutrients. 2019;11(9):E2068. https://pubmed.ncbi.nlm.nih.gov/31484341/
Willcox DC, Scapagnini G, Willcox BJ. Healthy aging diets other than the Mediterranean: a focus on the Okinawan diet. Mech Ageing Dev. 2014;136–7:148–62. https://pubmed.ncbi.nlm.nih.gov/24462788/
Everitt AV, Hilmer SN, Brand-Miller JC, et al. Dietary approaches that delay age-related diseases. Clin Interv Aging. 2006;1(1):11–31. https://pubmed.ncbi.nlm.nih.gov/18047254/
Jacobs DR Jr, Orlich MJ. Diet pattern and longevity: do simple rules suffice? A commentary. Am J Clin Nutr. 2014;100(Suppl 1):313S-9S. https://pubmed.ncbi.nlm.nih.gov/24871470/
Kim H, Caulfield LE, Rebholz CM. Healthy plant-based diets are associated with lower risk of all-cause mortality in US adults. J Nutr. 2018;148(4):624–31. https://pubmed.ncbi.nlm.nih.gov/29659968/
Orlich MJ, Chiu THT, Dhillon PK, et al. Vegetarian epidemiology: review and discussion of findings from geographically diverse cohorts. Adv Nutr. 2019;10(Suppl_4):S284–95. https://pubmed.ncbi.nlm.nih.gov/31728496/
Hemler EC, Hu FB. Plant-based diets for cardiovascular disease prevention: all plant foods are not created equal. Curr Atheroscler Rep. 2019;21(5):18. https://pubmed.ncbi.nlm.nih.gov/30895476/
Kim H, Caulfield LE, Garcia-Larsen V, Steffen LM, Coresh J, Rebholz CM. Plant-based diets are associated with a lower risk of incident cardiovascular disease, cardiovascular disease mortality, and all-cause mortality in a general population of middle-aged adults. J Am Heart Assoc. 2019;8(16):e012865. https://pubmed.ncbi.nlm.nih.gov/31387433/
Martínez-González MA, Sánchez-Tainta A, Corella D, et al. A provegetarian food pattern and reduction in total mortality in the Prevención con Dieta Mediterránea (PREDIMED) study. Am J Clin Nutr. 2014;100 Suppl 1:320S-8S. https://pubmed.ncbi.nlm.nih.gov/24871477/
Li H, Zeng X, Wang Y, et al. A prospective study of healthful and unhealthful plant-based diet and risk of overall and cause-specific mortality. Eur J Nutr. Published online August 11, 2021.; https://pubmed.ncbi.nlm.nih.gov/34379193/
Keaver L, Ruan M, Chen F, et al. Plant– and animal-based diet quality and mortality among US adults: a cohort study. Br J Nutr. 2021;125(12):1405–15. https://pubmed.ncbi.nlm.nih.gov/32943123/
Kim H, Caulfield LE, Garcia-Larsen V, Steffen LM, Coresh J, Rebholz CM. Plant-based diets are associated with a lower risk of incident cardiovascular disease, cardiovascular disease mortality, and all-cause mortality in a general population of middle-aged adults. J Am Heart Assoc. 2019;8(16):e012865. https://pubmed.ncbi.nlm.nih.gov/31387433/
Li H, Zeng X, Wang Y, et al. A prospective study of healthful and unhealthful plant-based diet and risk of overall and cause-specific mortality. Eur J Nutr. Published online August 11, 2021.; https://pubmed.ncbi.nlm.nih.gov/34379193/
Baden MY, Liu G, Satija A, et al. Changes in plant-based diet quality and total and cause-specific mortality. Circulation. 2019;140(12):979–91. https://pubmed.ncbi.nlm.nih.gov/31401846/
Keaver L, Ruan M, Chen F, et al. Plant– and animal-based diet quality and mortality among US adults: a cohort study. Br J Nutr. 2021;125(12):1405–15. https://pubmed.ncbi.nlm.nih.gov/32943123/
McCarty MF. Proposal for a dietary “phytochemical index.” Med Hypotheses. 2004;63(5):813–7. https://pubmed.ncbi.nlm.nih.gov/15488652/
U.S. Department of Agriculture, Economic Research Service. Loss-adjusted food availability. https://www.ers.usda.gov/webdocs/DataFiles/50472/calories.xls?v=7455.7. Updated August 26, 2019. Accessed January 17, 2022.; https://www.ers.usda.gov/webdocs/DataFiles/50472/calories.xls?v=7455.7
U.S. Department of Agriculture, Economic Research Service. Loss-adjusted food availability. https://www.ers.usda.gov/webdocs/DataFiles/50472/calories.xls?v=7455.7. Updated August 26, 2019. Accessed January 17, 2022.; https://www.ers.usda.gov/webdocs/DataFiles/50472/calories.xls?v=7455.7
Mirmiran P, Bahadoran Z, Golzarand M, Shiva N, Azizi F. Association between dietary phytochemical index and 3-year changes in weight, waist circumference and body adiposity index in adults: Tehran Lipid and Glucose study. Nutr Metab (Lond). 2012;9(1):108. https://pubmed.ncbi.nlm.nih.gov/23206375/
Golzarand M, Bahadoran Z, Mirmiran P, Sadeghian-Sharif S, Azizi F. Dietary phytochemical index is inversely associated with the occurrence of hypertension in adults: a 3-year follow-up (the Tehran Lipid and Glucose Study). Eur J Clin Nutr. 2015;69(3):392–8. https://pubmed.ncbi.nlm.nih.gov/25387902/
Abshirini M, Mahaki B, Bagheri F, Siassi F, Koohdani F, Sotoudeh G. Higher intake of phytochemical-rich foods is inversely related to prediabetes: a case-control study. Int J Prev Med. 2018;9:64. https://pubmed.ncbi.nlm.nih.gov/30147853/
Kim M, Park K. Association between phytochemical index and metabolic syndrome. Nutr Res Pract. 2020;14(3):252–61. https://pubmed.ncbi.nlm.nih.gov/32528632/
Golzarand M, Mirmiran P, Bahadoran Z, Alamdari S, Azizi F. Dietary phytochemical index and subsequent changes of lipid profile: a 3-year follow-up in Tehran Lipid and Glucose Study in Iran. ARYA Atheroscler. 2014;10(4):203–10. https://pubmed.ncbi.nlm.nih.gov/25258636/
Darooghegi Mofrad M, Siassi F, Guilani B, Bellissimo N, Azadbakht L. Association of dietary phytochemical index and mental health in women: a cross-sectional study. Br J Nutr. 2019;121(9):1049–56. https://pubmed.ncbi.nlm.nih.gov/33298144/
Aghababayan S, Sheikhi Mobarakeh Z, Qorbani M, et al. Dietary phytochemical index and benign breast diseases: a case-control study. Nutr Cancer. 2020;72(6):1067–73. https://pubmed.ncbi.nlm.nih.gov/31475586/
Bahadoran Z, Karimi Z, Houshiar-Rad A, Mirzayi HR, Rashidkhani B. Dietary phytochemical index and the risk of breast cancer: a case control study in a population of Iranian women. Asian Pac J Cancer Prev. 2013;14(5):2747–51. https://pubmed.ncbi.nlm.nih.gov/23803026/
Bellavia A, Larsson SC, Bottai M, Wolk A, Orsini N. Fruit and vegetable consumption and all-cause mortality: a dose-response analysis. Am J Clin Nutr. 2013;98(2):454–9. https://pubmed.ncbi.nlm.nih.gov/23803880/
Juraske R, Mutel CL, Stoessel F, Hellweg S. Life cycle human toxicity assessment of pesticides: comparing fruit and vegetable diets in Switzerland and the United States. Chemosphere. 2009;77(7):939–45. https://pubmed.ncbi.nlm.nih.gov/19729188/
Bellavia A, Larsson SC, Bottai M, Wolk A, Orsini N. Fruit and vegetable consumption and all-cause mortality: a dose-response analysis. Am J Clin Nutr. 2013;98(2):454–9. https://pubmed.ncbi.nlm.nih.gov/23803880/
Nicklett EJ, Semba RD, Xue QL, et al. Fruit and vegetable intake, physical activity, and mortality in older community-dwelling women. J Am Geriatr Soc. 2012;60(5):862–8. https://pubmed.ncbi.nlm.nih.gov/22587851/
Lo YT, Chang YH, Wahlqvist ML, Huang HB, Lee MS. Spending on vegetable and fruit consumption could reduce all-cause mortality among older adults. Nutr J. 2012;11:113. https://pubmed.ncbi.nlm.nih.gov/23253183/
Dórea JG. Vegetarian diets and exposure to organochlorine pollutants, lead, and mercury. Am J Clin Nutr. 2004;80(1):237–8. https://pubmed.ncbi.nlm.nih.gov/15213054/
Hergenrather J, Hlady G, Wallace B, Savage E. Pollutants in breast milk of vegetarians. N Engl J Med. 1981;304(13):792. https://pubmed.ncbi.nlm.nih.gov/7464895/
Key TJ, Appleby PN, Spencer EA, et al. Cancer incidence in British vegetarians. Br J Cancer. 2009;101(1):192–7. https://pubmed.ncbi.nlm.nih.gov/19536095/
Dearfield KL, Edwards SR, O’Keefe MM, et al. Dietary estimates of dioxins consumed in U.S. Department of Agriculture – regulated meat and poultry products. J Food Prot. 2013;76(9):1597–607. https://pubmed.ncbi.nlm.nih.gov/23992505/
Hernández ÁR, Boada LD, Mendoza Z, et al. Consumption of organic meat does not diminish the carcinogenic potential associated with the intake of persistent organic pollutants (POPs). Environ Sci Pollut Res Int. 2017;24(5):4261–73. https://pubmed.ncbi.nlm.nih.gov/25893622/
U.S. Department of Agriculture, Economic Research Service. Per capita red meat and poultry consumption expected to decrease modestly in 2022. https://www.ers.usda.gov/data-products/chart-gallery/gallery/chart-detail/?chartId=103767. Last updated April 15, 2022. Accessed December 28, 2022.; https://www.ers.usda.gov/data-products/chart-gallery/gallery/chart-detail/?chartId=103767
Hernández ÁR, Boada LD, Mendoza Z, et al. Consumption of organic meat does not diminish the carcinogenic potential associated with the intake of persistent organic pollutants (POPs). Environ Sci Pollut Res Int. 2017;24(5):4261–73. https://pubmed.ncbi.nlm.nih.gov/25893622/
Ta CA, Zee JA, Desrosiers T, et al. Binding capacity of various fibre to pesticide residues under simulated gastrointestinal conditions. Food Chem Toxicol. 1999;37(12):1147–51. https://pubmed.ncbi.nlm.nih.gov/10654590/
Lee YM, Shin JY, Kim SA, Jacobs DR, Lee DH. Can habitual exercise help reduce serum concentrations of lipophilic chemical mixtures? Association between physical activity and persistent organic pollutants. Diabetes Metab J. 2020;44(5):764–74. https://pubmed.ncbi.nlm.nih.gov/32174058/
Genuis SJ, Lane K, Birkholz D. Human elimination of organochlorine pesticides: blood, urine, and sweat study. Biomed Res Int. 2016;2016:1624643. https://pubmed.ncbi.nlm.nih.gov/27800487/
Yiamouyiannis CA, Sanders RA, Watkins JB III, Martin BJ. Chronic physical activity: hepatic hypertrophy and increased total biotransformation enzyme activity. Biochem Pharmacol. 1992;44(1):121–7. https://pubmed.ncbi.nlm.nih.gov/8474015/
Watkins JB, Crawford ST, Sanders RA. Chronic voluntary exercise may alter hepatobiliary clearance of endogenous and exogenous chemicals in rats. Drug Metab Dispos. 1994;22(4):537–43. https://pubmed.ncbi.nlm.nih.gov/7956727/
Lupton SJ, O’Keefe M, Muñiz-Ortiz JG, Clinch N, Basu P. Survey of polychlorinated dibenzo-p-dioxins, polychlorinated dibenzofurans and non-ortho-polychlorinated biphenyls in US meat and poultry, 2012–13: toxic equivalency levels, patterns, temporal trends and implications. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 2017;34(11):1970–81. https://pubmed.ncbi.nlm.nih.gov/28632453/
González N, Marquès M, Nadal M, Domingo JL. Meat consumption: which are the current global risks? A review of recent (2010–2020) evidences. Food Res Int. 2020;137:109341. https://pubmed.ncbi.nlm.nih.gov/33233049/
Melina V, Craig W, Levin S. Position of the Academy of Nutrition and Dietetics: vegetarian diets. J Acad Nutr Diet. 2016;116(12):1970–80. https://pubmed.ncbi.nlm.nih.gov/27886704/
Johnston PK. Recognition: Mervyn G Hardinge. Am J Clin Nutr. 1999;70(3):431s-2s. https://pubmed.ncbi.nlm.nih.gov/10479213/
Ma Y, Pagoto SL, Griffith JA, et al. A dietary quality comparison of popular weight-loss plans. J Am Diet Assoc. 2007;107(10):1786–91. https://pubmed.ncbi.nlm.nih.gov/17904938/
Clarys P, Deliens T, Huybrechts I, et al. Comparison of nutritional quality of the vegan, vegetarian, semi-vegetarian, pesco-vegetarian and omnivorous diet. Nutrients. 2014;6(3):1318–32. https://pubmed.ncbi.nlm.nih.gov/24667136/
Farmer B, Larson BT, Fulgoni VL, Rainville AJ, Liepa GU. A vegetarian dietary pattern as a nutrient-dense approach to weight management: an analysis of the National Health and Nutrition Examination Survey 1999–2004. J Am Diet Assoc. 2011;111(6):819–27. https://pubmed.ncbi.nlm.nih.gov/21616194/
Van Horn L. Achieving nutrient density: a vegetarian approach. J Am Diet Assoc. 2011;111(6):799. https://pubmed.ncbi.nlm.nih.gov/21616188/
Keenan S, Mitts KG, Kurtz CA. Scurvy presenting as a medial head tear of the gastrocnemius. Orthopedics. 2002;25(6):689–91. https://pubmed.ncbi.nlm.nih.gov/12083582/
Mariotti F, ed. Vegetarian and Plant-Based Diets in Health and Disease Prevention. Academic Press; 2017. https://worldcat.org/title/988275855
Armstrong BK. Absorption of vitamin B12 from the human colon. Am J Clin Nutr. 1968;21(4):298–9. https://pubmed.ncbi.nlm.nih.gov/5647478/
Gupta ES, Sheth SP, Ganjiwale JD. Association of vitamin B12 deficiency and use of reverse osmosis processed water for drinking: a cross-sectional study from Western India. J Clin Diagn Res. 2016;10(5):OC37–40. https://pubmed.ncbi.nlm.nih.gov/27437269/
Pawlak R, Lester SE, Babatunde T. The prevalence of cobalamin deficiency among vegetarians assessed by serum vitamin B12: a review of literature. Eur J Clin Nutr. 2014;68(5):541–8. https://pubmed.ncbi.nlm.nih.gov/24667752/
Herrmann W, Geisel J. Vegetarian lifestyle and monitoring of vitamin B-12 status. Clin Chim Acta. 2002;326(1–2):47–59. https://pubmed.ncbi.nlm.nih.gov/12417096/
Mariotti F, ed. Vegetarian and Plant-Based Diets in Health and Disease Prevention. Academic Press; 2017. https://worldcat.org/title/988275855
Eitenmiller R, Ye L, Landen WO Jr. Vitamin Analysis for the Health and Food Sciences. 2nd ed. CRC Press; 2007:469.Book
Del Bo’ C, Riso P, Gardana C, Brusamolino A, Battezzati A, Ciappellano S. Effect of two different sublingual dosages of vitamin B12 on cobalamin nutritional status in vegans and vegetarians with a marginal deficiency: a randomized controlled trial. Clin Nutr. 2019;38(2):575–83. https://pubmed.ncbi.nlm.nih.gov/29499976/
MacFarlane AJ, Shi Y, Greene-Finestone LS. High-dose compared with low-dose vitamin B-12 supplement use is not associated with higher vitamin B-12 status in children, adolescents, and older adults. J Nutr. 2014;144(6):915–20. https://pubmed.ncbi.nlm.nih.gov/24699807/
Rajan S, Wallace JI, Brodkin KI, Beresford SA, Allen RH, Stabler SP. Response of elevated methylmalonic acid to three dose levels of oral cobalamin in older adults. J Am Geriatr Soc. 2002;50(11):1789–95. https://pubmed.ncbi.nlm.nih.gov/12410896/
Eussen S, de Groot L, Clarke R, et al. Oral cyanocobalamin supplementation in older people with vitamin B12 deficiency: a dose-finding trial. Arch Intern Med. 2005;165(10):1167–72. https://pubmed.ncbi.nlm.nih.gov/15911731/
Rizzo G, Laganà AS, Rapisarda AMC, et al. Vitamin B12 among vegetarians: status, assessment and supplementation. Nutrients. 2016;8(12):767. https://pubmed.ncbi.nlm.nih.gov/27916823/
Crane MG, Sample C, Patchett S, Register UD. Vitamin B12 studies in total vegetarians (vegans). J Nutr Med. 1994;4(4):419–30. https://www.tandfonline.com/doi/abs/10.3109/13590849409003591
Briani C, Dalla Torre C, Citton V, et al. Cobalamin deficiency: clinical picture and radiological findings. Nutrients. 2013;5(11):4521–39. https://pubmed.ncbi.nlm.nih.gov/24248213/
Crane MG, Sample C, Patchett S, Register UD. Vitamin B12 studies in total vegetarians (vegans). J Nutr Med. 1994;4(4):419–30. https://www.tandfonline.com/doi/abs/10.3109/13590849409003591
Mott A, Bradley T, Wright K, et al. Effect of vitamin K on bone mineral density and fractures in adults: an updated systematic review and meta-analysis of randomised controlled trials. Osteoporos Int. 2019;30(8):1543–59. https://pubmed.ncbi.nlm.nih.gov/31076817/
EFSA Panel on Dietetic Products, Nutrition and Allergies, Turck D, Bresson JL, et al. Dietary reference values for vitamin K. EFSA J. 2017;15(5):e04780. https://pubmed.ncbi.nlm.nih.gov/32625486/
Nakagawa K, Hirota Y, Sawada N, et al. Identification of UBIAD1 as a novel human menaquinone-4 biosynthetic enzyme. Nature. 2010;468(7320):117–21. https://pubmed.ncbi.nlm.nih.gov/20953171/
Kwok CS, Gulati M, Michos ED, et al. Dietary components and risk of cardiovascular disease and all-cause mortality: a review of evidence from meta-analyses. Eur J Prev Cardiol. 2019;26(13):1415–29. https://pubmed.ncbi.nlm.nih.gov/30971126/
Shea MK, Barger K, Booth SL, et al. Vitamin K status, cardiovascular disease, and all-cause mortality: a participant-level meta-analysis of 3 US cohorts. Am J Clin Nutr. 2020;111(6):1170–7. https://pubmed.ncbi.nlm.nih.gov/32359159/
Chase P, Mitchell K, Morley JE. In the steps of giants: the early geriatrics texts. J Am Geriatr Soc. 2000;48(1):89–94. https://pubmed.ncbi.nlm.nih.gov/10642028/
Stranges S, Takeda A, Martin N, Rees K. Cochrane corner: does the Mediterranean-style diet help in the prevention of cardiovascular disease? Heart. 2019;105(22):1691–4. https://pubmed.ncbi.nlm.nih.gov/31439660/
Wizard Edison says doctors of future will give no medicine. The Newark Advocate. https://archive.org/details/newark-advocate-1903–01–02/mode/1up?view=theater. Published January 2, 1903;46:47:1. Accessed February 21 https://archive.org/details/newark-advocate-1903-01-02/mode/1up?view=theater
American College of Lifestyle Medicine. About us. http://lifestylemedicine.org/about-us/. Accessed December 28, 2022.; https://lifestylemedicine.org/about-us/
Mokdad AH, Ballestros K, Echko M, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Vodovotz Y, Barnard N, Hu FB, et al. Prioritized research for the prevention, treatment, and reversal of chronic disease: recommendations from the Lifestyle Medicine Research Summit. Front Med (Lausanne). 2020;7:585744. https://pubmed.ncbi.nlm.nih.gov/33415115/
Zhang YB, Pan XF, Chen J, et al. Combined lifestyle factors, all-cause mortality and cardiovascular disease: a systematic review and meta-analysis of prospective cohort studies. J Epidemiol Community Health. 2021;75(1):92–9. https://pubmed.ncbi.nlm.nih.gov/32892156/
Willcox BJ, Willcox DC, Ferrucci L. Secrets of healthy aging and longevity from exceptional survivors around the globe: lessons from octogenarians to supercentenarians. J Gerontol A Biol Sci Med Sci. 2008;63(11):1181–5. https://pubmed.ncbi.nlm.nih.gov/19038832/
Ford ES, Bergmann MM, Kröger J, Schienkiewitz A, Weikert C, Boeing H. Healthy living is the best revenge: findings from the European Prospective Investigation Into Cancer and Nutrition – Potsdam study. Arch Intern Med. 2009;169(15):1355–62. https://pubmed.ncbi.nlm.nih.gov/19667296/
Platz EA, Willett WC, Colditz GA, Rimm EB, Spiegelman D, Giovannucci E. Proportion of colon cancer risk that might be preventable in a cohort of middle-aged US men. Cancer Causes Control. 2000;11(7):579–88. https://pubmed.ncbi.nlm.nih.gov/10977102/
Ford ES, Bergmann MM, Kröger J, Schienkiewitz A, Weikert C, Boeing H. Healthy living is the best revenge: findings from the European Prospective Investigation Into Cancer and Nutrition – Potsdam study. Arch Intern Med. 2009;169(15):1355–62. https://pubmed.ncbi.nlm.nih.gov/19667296/
Wahls TL. The seventy percent solution. J Gen Intern Med. 2011;26(10):1215–6. https://pubmed.ncbi.nlm.nih.gov/21253878/
Khaw KT, Wareham N, Bingham S, Welch A, Luben R, Day N. Combined impact of health behaviours and mortality in men and women: the EPIC-Norfolk prospective population study. PLoS Med. 2008;5(1):e12. https://pubmed.ncbi.nlm.nih.gov/18184033/
Машина из фильма «Назад в будущее». – Примеч. ред.
Wang K, Li Y, Liu G, et al. Healthy lifestyle for prevention of premature death among users and nonusers of common preventive medications: a prospective study in 2 US cohorts. JAHA. 2020;9(13):e016692. https://pubmed.ncbi.nlm.nih.gov/32578485/
King DE, Mainous AG III, Geesey ME. Turning back the clock: adopting a healthy lifestyle in middle age. Am J Med. 2007;120(7):598–603. https://pubmed.ncbi.nlm.nih.gov/17602933/
Nyberg ST, Singh-Manoux A, Pentti J, et al. Association of healthy lifestyle with years lived without major chronic diseases. JAMA Intern Med. 2020;180(5):760–8. https://pubmed.ncbi.nlm.nih.gov/32250383/
Hall WJ. Centenarians: metaphor becomes reality. Arch Intern Med. 2008;168(3):262–3. https://pubmed.ncbi.nlm.nih.gov/18268165/
Zhang S, Tomata Y, Discacciati A, et al. Combined healthy lifestyle behaviors and disability-free survival: the Ohsaki Cohort 2006 Study. J Gen Intern Med. 2019;34(9):1724–9. https://pubmed.ncbi.nlm.nih.gov/31144283/
Vallance JK, Gardiner PA, Lynch BM, et al. Evaluating the evidence on sitting, smoking, and health: is sitting really the new smoking? Am J Public Health. 2018;108(11):1478–82. https://pubmed.ncbi.nlm.nih.gov/30252516/
Rezende LFM, Sá TH, Mielke GI, Viscondi JYK, Rey-López JP, Garcia LMT. All-cause mortality attributable to sitting time: analysis of 54 countries worldwide. Am J Prev Med. 2016;51(2):253–63. https://pubmed.ncbi.nlm.nih.gov/27017420/
Vallance JK, Gardiner PA, Lynch BM, et al. Evaluating the evidence on sitting, smoking, and health: is sitting really the new smoking? Am J Public Health. 2018;108(11):1478–82. https://pubmed.ncbi.nlm.nih.gov/30252516/
Taylor DH Jr, Hasselblad V, Henley SJ, Thun MJ, Sloan FA. Benefits of smoking cessation for longevity. Am J Public Health. 2002;92(6):990–6. https://pubmed.ncbi.nlm.nih.gov/12036794/
Engelhard CL, Garson A, Dorn S. Reducing obesity: policy strategies from the tobacco wars. Methodist Debakey Cardiovasc J. 2009;5(4):46–50. https://pubmed.ncbi.nlm.nih.gov/20143597/
Cornelius ME, Wang TW, Jamal A, Loretan CG, Neff LJ. Tobacco product use among adults – United States, 2019. MMWR Morb Mortal Wkly Rep. 2020;69(46):1736–42. https://pubmed.ncbi.nlm.nih.gov/33211681/
Mokdad AH, Ballestros K, Echko M, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Barnard ND. The physician’s role in nutrition-related disorders: from bystander to leader. Virtual Mentor. 2013;15(4):367–72. https://pubmed.ncbi.nlm.nih.gov/23566788/
Ding D, Grunseit AC, Chau JY, Vo K, Byles J, Bauman AE. Retirement – a transition to a healthier lifestyle?: evidence from a large Australian study. Am J Prev Med. 2016;51(2):170–8. https://pubmed.ncbi.nlm.nih.gov/26972491/
Rebelo-Marques A, Lages ADS, Andrade R, et al. Aging hallmarks: the benefits of physical exercise. Front Endocrinol (Lausanne). 2018;9:258. https://pubmed.ncbi.nlm.nih.gov/29887832/
Wolf AM. Rodent diet aids and the fallacy of caloric restriction. Mech Ageing Dev. 2021;200:111584. https://pubmed.ncbi.nlm.nih.gov/34673082/
Seals DR, Justice JN, LaRocca TJ. Physiological geroscience: targeting function to increase healthspan and achieve optimal longevity. J Physiol. 2016;594(8):2001–24. https://pubmed.ncbi.nlm.nih.gov/25639909/
Lin YH, Chen Y-C, Tseng Y-C, Tsai S-T, Tseng Y-H. Physical activity and successful aging among middle-aged and older adults: a systematic review and meta-analysis of cohort studies. Aging (Albany NY). 2020;12(9):7704–16. https://pubmed.ncbi.nlm.nih.gov/32350152/
Troiano RP, Berrigan D, Dodd KW, Mâsse LC, Tilert T, McDowell M. Physical activity in the United States measured by accelerometer. Med Sci Sports Exerc. 2008;40(1):181–8. https://pubmed.ncbi.nlm.nih.gov/18091006/
Pedersen BK. Which type of exercise keeps you young? Curr Opin Clin Nutr Metab Care. 2019;22(2):167–73. https://pubmed.ncbi.nlm.nih.gov/30640736/
Di Lorito C, Long A, Byrne A, et al. Exercise interventions for older adults: a systematic review of meta-analyses. J Sport Health Sci. 2021;10(1):29–47. https://pubmed.ncbi.nlm.nih.gov/32525097/
Pahor M, Guralnik JM, Ambrosius WT, et al. Effect of structured physical activity on prevention of major mobility disability in older adults: the LIFE study randomized clinical trial. JAMA. 2014;311(23):2387–96. https://pubmed.ncbi.nlm.nih.gov/24866862/
Sherrington C, Fairhall N, Kwok W, et al. Evidence on physical activity and falls prevention for people aged 65+ years: systematic review to inform the WHO guidelines on physical activity and sedentary behaviour. Int J Behav Nutr Phys Act. 2020;17(1):144. https://pubmed.ncbi.nlm.nih.gov/33239019/
de Souto Barreto P, Rolland Y, Vellas B, Maltais M. Association of long-term exercise training with risk of falls, fractures, hospitalizations, and mortality in older adults: a systematic review and meta-analysis. JAMA Intern Med. 2019;179(3):394–405. https://pubmed.ncbi.nlm.nih.gov/30592475/
Soltani S, Hunter GR, Kazemi A, Shab-Bidar S. The effects of weight loss approaches on bone mineral density in adults: a systematic review and meta-analysis of randomized controlled trials. Osteoporos Int. 2016;27(9):2655–71. https://pubmed.ncbi.nlm.nih.gov/27154437/
García-Hermoso A, Ramirez-Vélez R, Sáez de Asteasu ML, et al. Safety and effectiveness of long-term exercise interventions in older adults: a systematic review and meta-analysis of randomized controlled trials. Sports Med. 2020;50(6):1095–106. https://pubmed.ncbi.nlm.nih.gov/32020543/
Di Lorito C, Long A, Byrne A, et al. Exercise interventions for older adults: a systematic review of meta-analyses. J Sport Health Sci. 2021;10(1):29–47. https://pubmed.ncbi.nlm.nih.gov/32525097/
Blumenthal JA, Babyak MA, Doraiswamy PM, et al. Exercise and pharmacotherapy in the treatment of major depressive disorder. Psychosom Med. 2007;69(7):587–96. https://pubmed.ncbi.nlm.nih.gov/17846259/
Gerbild H, Larsen CM, Graugaard C, Areskoug Josefsson K. Physical activity to improve erectile function: a systematic review of intervention studies. Sex Med. 2018;6(2):75–89. https://pubmed.ncbi.nlm.nih.gov/29661646/
Marquez DX, Aguiñaga S, Vásquez PM, et al. A systematic review of physical activity and quality of life and well-being. Transl Behav Med. 2020;10(5):1098–109. https://pubmed.ncbi.nlm.nih.gov/33044541/
Warburton DER, Bredin SSD. Health benefits of physical activity: a systematic review of current systematic reviews. Curr Opin Cardiol. 2017;32(5):541–56. https://pubmed.ncbi.nlm.nih.gov/28708630/
Fock KM, Khoo J. Diet and exercise in management of obesity and overweight. J Gastroenterol Hepatol. 2013;28 Suppl 4:59–63. https://pubmed.ncbi.nlm.nih.gov/24251706/
Archer E, Hand GA, Blair SN. Correction: Validity of U.S. nutritional surveillance: National Health and Nutrition Examination Survey caloric energy intake data, 1971–2010. PLoS One. 2013;8(10):10.1371/annotation/c313df3a-52bd-4cbe-af14–6676480d1a43. https://pubmed.ncbi.nlm.nih.gov/24130784/
Blair SN. Physical inactivity: the biggest public health problem of the 21st century. Br J Sports Med. 2009;43(1):1–2. https://pubmed.ncbi.nlm.nih.gov/19136507/
Mokdad AH, Ballestros K, Echko M, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Stanaway JD, Afshin A, Gakidou E, et al. Global, regional, and national comparative risk assessment of 84 behavioural, environmental and occupational, and metabolic risks or clusters of risks for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018;392(10159):1923–94. https://pubmed.ncbi.nlm.nih.gov/30496105/
Mokdad AH, Ballestros K, Echko M, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Campisi J, Kapahi P, Lithgow GJ, Melov S, Newman JC, Verdin E. From discoveries in ageing research to therapeutics for healthy ageing. Nature. 2019;571(7764):183–92. https://pubmed.ncbi.nlm.nih.gov/31292558/
World Health Organization. Global Health Risks: Mortality and Burden of Disease Attributable to Selected Major Risks. World Health Organization; 2009. https://apps.who.int/iris/handle/10665/44203
Lee IM, Shiroma EJ, Lobelo F, Puska P, Blair SN, Katzmarzyk PT. Impact of physical inactivity on the world’s major non-communicable diseases. Lancet. 2012;380(9838):219–29. https://pubmed.ncbi.nlm.nih.gov/22818936/
Strain T, Brage S, Sharp SJ, et al. Use of the prevented fraction for the population to determine deaths averted by existing prevalence of physical activity: a descriptive study. Lancet Glob Health. 2020;8(7):e920–30. https://pubmed.ncbi.nlm.nih.gov/32562648/
Wade KH, Richmond RC, Smith GD. Physical activity and longevity: how to move closer to causal inference. Br J Sports Med. 2018;52(14):890–1. https://pubmed.ncbi.nlm.nih.gov/29545236/
Richard A, Martin B, Wanner M, Eichholzer M, Rohrmann S. Effects of leisure-time and occupational physical activity on total mortality risk in NHANES III according to sex, ethnicity, central obesity, and age. J Phys Act Health. 2015;12(2):184–92. https://pubmed.ncbi.nlm.nih.gov/24770336/
Kujala UM. Is physical activity a cause of longevity? It is not as straightforward as some would believe. A critical analysis. Br J Sports Med. 2018;52(14):914–8. https://pubmed.ncbi.nlm.nih.gov/29545237/
Karvinen S, Waller K, Silvennoinen M, et al. Physical activity in adulthood: genes and mortality. Sci Rep. 2015;5:18259. https://pubmed.ncbi.nlm.nih.gov/26666586/
O’Keefe JH, Franklin B, Lavie CJ. Exercising for health and longevity vs peak performance: different regimens for different goals. Mayo Clin Proc. 2014;89(9):1171–5. https://pubmed.ncbi.nlm.nih.gov/25128073/
O’Keefe JH, Franklin B, Lavie CJ. Exercising for health and longevity vs peak performance: different regimens for different goals. Mayo Clin Proc. 2014;89(9):1171–5. https://pubmed.ncbi.nlm.nih.gov/25128073/
O’Keefe JH, Franklin B, Lavie CJ. Exercising for health and longevity vs peak performance: different regimens for different goals. Mayo Clin Proc. 2014;89(9):1171–5. https://pubmed.ncbi.nlm.nih.gov/25128073/
Lee D, Brellenthin AG, Thompson PD, Sui X, Lee IM, Lavie CJ. Running as a key lifestyle medicine for longevity. Prog Cardiovasc Dis. 2017;60(1):45–55. https://pubmed.ncbi.nlm.nih.gov/28365296/
Montgomery MJ, Kandi D. QuickStats: percentage of adults who met federal guidelines for aerobic physical activity through leisure-time activity, by race/ethnicity – National Health Interview Survey, 2008–2017. MMWR Morb Mortal Wkly Rep. 2019;68:292. https://pubmed.ncbi.nlm.nih.gov/30921300/
Lee D, Lavie CJ, Sui X, Blair SN. Running and mortality: is more actually worse? Mayo Clin Proc. 2016;91(4):534–6. https://pubmed.ncbi.nlm.nih.gov/27046526/
Schnohr P, Marott JL, O’Keefe JH. Reply: exercise and mortality reduction: recurring reverse J– or U-curves. J Am Coll Cardiol. 2015;65(24):2674–6. https://pubmed.ncbi.nlm.nih.gov/26088316/
Спортом на выносливость считаются спортивная ходьба; бег на средние и длинные дистанции; марафонский бег; велоспорт; плавание; гребля на академических лодках, байдарках, каноэ; лыжные гонки; конькобежный спорт; биатлон; спортивное ориентирование; триатлон. – Примеч. ред.
Barnard ND, Goldman DM, Loomis JF, et al. Plant-based diets for cardiovascular safety and performance in endurance sports. Nutrients. 2019;11(1):130. https://pubmed.ncbi.nlm.nih.gov/30634559/
Barnard ND, Goldman DM, Loomis JF, et al. Plant-based diets for cardiovascular safety and performance in endurance sports. Nutrients. 2019;11(1):130. https://pubmed.ncbi.nlm.nih.gov/30634559/
Sacks FM, Lichtenstein AH, Wu JHY, et al. Dietary fats and cardiovascular disease: a presidential advisory from the American Heart Association. Circulation. 2017;136(3):e1–23. https://pubmed.ncbi.nlm.nih.gov/28620111/
Smith MM, Trexler ET, Sommer AJ, et al. Unrestricted paleolithic diet is associated with unfavorable changes to blood lipids in healthy subjects. Int J Exerc Sci. 2014;7(2):128–39. Note this study has been retracted https://retractionwatch.com/2017/06/30/researcher-tangled-crossfit-loses-two-papers/
Barnard RJ, Ugianskis EJ, Martin DA, Inkeles SB. Role of diet and exercise in the management of hyperinsulinemia and associated atherosclerotic risk factors. Am J Cardiol. 1992;69(5):440–4. https://pubmed.ncbi.nlm.nih.gov/1736602/
Smith MM, Trexler ET, Sommer AJ, et al. Unrestricted paleolithic diet is associated with unfavorable changes to blood lipids in healthy subjects. Int J Exerc Sci. 2014;7(2):128–39. Note this study has been retracted https://retractionwatch.com/2017/06/30/researcher-tangled-crossfit-loses-two-papers/
Craddock JC, Neale EP, Peoples GE, Probst YC. Plant-based eating patterns and endurance performance: a focus on inflammation, oxidative stress and immune responses. Nutr Bull. 2020;45(2):123–32. https://onlinelibrary.wiley.com/doi/abs/10.1111/nbu.12427
В российском прокате – «Переломный момент». – Примеч. ред.
Barnard ND, Goldman DM, Loomis JF, et al. Plant-based diets for cardiovascular safety and performance in endurance sports. Nutrients. 2019;11(1):130. https://pubmed.ncbi.nlm.nih.gov/30634559/
Lynch HM, Wharton CM, Johnston CS. Cardiorespiratory fitness and peak torque differences between vegetarian and omnivore endurance athletes: a cross-sectional study. Nutrients. 2016;8(11):E726. https://pubmed.ncbi.nlm.nih.gov/27854281/
Boutros GH, Landry-Duval MA, Garzon M, Karelis AD. Is a vegan diet detrimental to endurance and muscle strength? Eur J Clin Nutr. 2020;74(11):1550–5. https://pubmed.ncbi.nlm.nih.gov/32332862/
Król W, Price S, Sliz D, et al. A vegan athlete’s heart – is it different? Morphology and function in echocardiography. Diagnostics (Basel). 2020;10(7):E477. https://pubmed.ncbi.nlm.nih.gov/32674452/
Veleba J, Matoulek M, Hill M, Pelikanova T, Kahleova H. “A vegetarian vs. conventional hypocaloric diet: the effect on physical fitness in response to aerobic exercise in patients with type 2 diabetes.” A parallel randomized study. Nutrients. 2016;8(11):671. https://pubmed.ncbi.nlm.nih.gov/27792174/
Veleba J, Matoulek M, Hill M, Pelikanova T, Kahleova H. “A vegetarian vs. conventional hypocaloric diet: the effect on physical fitness in response to aerobic exercise in patients with type 2 diabetes.” A parallel randomized study. Nutrients. 2016;8(11):671. https://pubmed.ncbi.nlm.nih.gov/27792174/
Kahleova H, Hrachovinova T, Hill M, Pelikanova T. Vegetarian diet in type 2 diabetes – improvement in quality of life, mood and eating behaviour. Diabet Med. 2013;30(1):127–9. https://pubmed.ncbi.nlm.nih.gov/23050853/
Kien CL, Bunn JY, Tompkins CL, et al. Substituting dietary monounsaturated fat for saturated fat is associated with increased daily physical activity and resting energy expenditure and with changes in mood. Am J Clin Nutr. 2013;97(4):689–97. https://pubmed.ncbi.nlm.nih.gov/23446891/
Dumas JA, Bunn JY, Nickerson J, et al. Dietary saturated fat and monounsaturated fat have reversible effects on brain function and the secretion of pro-inflammatory cytokines in young women. Metab Clin Exp. 2016;65(10):1582–8. https://pubmed.ncbi.nlm.nih.gov/27621193/
Kien CL, Bunn JY, Tompkins CL, et al. Substituting dietary monounsaturated fat for saturated fat is associated with increased daily physical activity and resting energy expenditure and with changes in mood. Am J Clin Nutr. 2013;97(4):689–97. https://pubmed.ncbi.nlm.nih.gov/23446891/
Kahleova H, Matoulek M, Malinska H, et al. Vegetarian diet improves insulin resistance and oxidative stress markers more than conventional diet in subjects with Type 2 diabetes. Diabet Med. 2011;28(5):549–59. https://pubmed.ncbi.nlm.nih.gov/21480966/
Roderka MN, Puri S, Batsis JA. Addressing obesity to promote healthy aging. Clin Geriatr Med. 2020;36(4):631–43. https://pubmed.ncbi.nlm.nih.gov/33010899/
Hales CM, Carroll MD, Fryar CD, Ogden CL. Prevalence of obesity and severe obesity among adults: United States, 2017–2018. NCHS Data Brief. 2020;(360):1–8. https://pubmed.ncbi.nlm.nih.gov/32487284/
Pontzer H, Yamada Y, Sagayama H, et al. Daily energy expenditure through the human life course. Science. 2021;373(6556):808–12. https://pubmed.ncbi.nlm.nih.gov/34385400/
Tam BT, Morais JA, Santosa S. Obesity and ageing: two sides of the same coin. Obes Rev. 2020;21(4):e12991. https://pubmed.ncbi.nlm.nih.gov/32020741/
Himbert C, Thompson H, Ulrich CM. Effects of intentional weight loss on markers of oxidative stress, DNA repair and telomere length – a systematic review. Obes Facts. 2017;10(6):648–65. https://pubmed.ncbi.nlm.nih.gov/29237161/
Bianchi VE. Weight loss is a critical factor to reduce inflammation. Clin Nutr ESPEN. 2018;28:21–35. https://pubmed.ncbi.nlm.nih.gov/30390883/
Tam BT, Morais JA, Santosa S. Obesity and ageing: two sides of the same coin. Obes Rev. 2020;21(4):e12991. https://pubmed.ncbi.nlm.nih.gov/32020741/
Ronan L, Alexander-Bloch AF, Wagstyl K, et al. Obesity associated with increased brain age from midlife. Neurobiol Aging. 2016;47:63–70. https://pubmed.ncbi.nlm.nih.gov/27562529/
Albanese E, Launer LJ, Egger M, et al. Body mass index in midlife and dementia: systematic review and meta-regression analysis of 589,649 men and women followed in longitudinal studies. Alzheimers Dement (Amst). 2017;8:165–78. https://pubmed.ncbi.nlm.nih.gov/28761927/
Chuang YF, An Y, Bilgel M, et al. Midlife adiposity predicts earlier onset of Alzheimer’s dementia, neuropathology and presymptomatic cerebral amyloid accumulation. Mol Psychiatry. 2016;21(7):910–5. https://pubmed.ncbi.nlm.nih.gov/26324099/
Olshansky SJ, Passaro DJ, Hershow RC, et al. A potential decline in life expectancy in the United States in the 21st century. N Engl J Med. 2005;352(11):1138–45. https://pubmed.ncbi.nlm.nih.gov/15784668/
Ludwig DS. Lifespan weighed down by diet. JAMA. 2016;315(21):2269–70. https://pubmed.ncbi.nlm.nih.gov/27043490/
Mann CC. Provocative study says obesity may reduce U.S. life expectancy. Science. 2005;307(5716):1716–7. https://pubmed.ncbi.nlm.nih.gov/15774742/
Sun Q, Townsend MK, Okereke OI, Franco OH, Hu FB, Grodstein F. Adiposity and weight change in mid-life in relation to healthy survival after age 70 in women: prospective cohort study. BMJ. 2009;339:b3796. https://pubmed.ncbi.nlm.nih.gov/20101015/
Santos-Lozano A, Pareja-Galeano H, Fuku N, et al. Implications of obesity in exceptional longevity. Ann Transl Med. 2016;4(20):416. https://pubmed.ncbi.nlm.nih.gov/27867968/
Tam BT, Morais JA, Santosa S. Obesity and ageing: two sides of the same coin. Obes Rev. 2020;21(4):e12991. https://pubmed.ncbi.nlm.nih.gov/32020741/
Pararasa C, Bailey CJ, Griffiths HR. Ageing, adipose tissue, fatty acids and inflammation. Biogerontology. 2015;16(2):235–48. https://pubmed.ncbi.nlm.nih.gov/25367746/
Rubin R. Postmenopausal women with a “normal” BMI might be overweight or even obese. JAMA. 2018;319(12):1185–7. https://pubmed.ncbi.nlm.nih.gov/29516084/
Jayedi A, Soltani S, Zargar MS, Khan TA, Shab-Bidar S. Central fatness and risk of all cause mortality: systematic review and dose-response meta-analysis of 72 prospective cohort studies. BMJ. 2020;370:m3324. https://pubmed.ncbi.nlm.nih.gov/32967840/
Klein S, Fontana L, Young VL, et al. Absence of an effect of liposuction on insulin action and risk factors for coronary heart disease. N Engl J Med. 2004;350(25):2549–57. https://pubmed.ncbi.nlm.nih.gov/15201411/
Blackburn G. Effect of degree of weight loss on health benefits. Obes Res. 1995;3 Suppl 2:211s-6s. https://pubmed.ncbi.nlm.nih.gov/8581779/
Klein S, Fontana L, Young VL, et al. Absence of an effect of liposuction on insulin action and risk factors for coronary heart disease. N Engl J Med. 2004;350(25):2549–57. https://pubmed.ncbi.nlm.nih.gov/15201411/
Chaston TB, Dixon JB. Factors associated with percent change in visceral versus subcutaneous abdominal fat during weight loss: findings from a systematic review. Int J Obes (Lond). 2008;32(4):619–28. https://pubmed.ncbi.nlm.nih.gov/18180786/
Haywood C, Sumithran P. Treatment of obesity in older persons – a systematic review. Obes Rev. 2019;20(4):588–98. https://pubmed.ncbi.nlm.nih.gov/30645010/
Muzumdar R, Allison DB, Huffman DM, et al. Visceral adipose tissue modulates mammalian longevity. Aging Cell. 2008;7(3):438–40. https://pubmed.ncbi.nlm.nih.gov/18363902/
Wiggins T, Guidozzi N, Welbourn R, Ahmed AR, Markar SR. Association of bariatric surgery with all-cause mortality and incidence of obesity-related disease at a population level: a systematic review and meta-analysis. PLoS Med. 2020;17(7):e1003206. https://pubmed.ncbi.nlm.nih.gov/32722673/
Kritchevsky SB, Beavers KM, Miller ME, et al. Intentional weight loss and all-cause mortality: a meta-analysis of randomized clinical trials. PLoS One. 2015;10(3):e0121993. https://pubmed.ncbi.nlm.nih.gov/25794148/
Wright N, Wilson L, Smith M, Duncan B, McHugh P. The BROAD study: a randomised controlled trial using a whole food plant-based diet in the community for obesity, ischaemic heart disease or diabetes. Nutr Diabetes. 2017;7(3):e256. https://pubmed.ncbi.nlm.nih.gov/28319109/
Hall KD, Guo J, Courville AB, et al. Effect of a plant-based, low-fat diet versus an animal-based, ketogenic diet on ad libitum energy intake. Nat Med. 2021;27(2):344–53. https://pubmed.ncbi.nlm.nih.gov/33479499/
Piers LS, Walker KZ, Stoney RM, Soares MJ, O’Dea K. Substitution of saturated with monounsaturated fat in a 4-week diet affects body weight and composition of overweight and obese men. Br J Nutr. 2003;90(3):717–27. https://pubmed.ncbi.nlm.nih.gov/13129479/
Rosqvist F, Iggman D, Kullberg J, et al. Overfeeding polyunsaturated and saturated fat causes distinct effects on liver and visceral fat accumulation in humans. Diabetes. 2014;63(7):2356–68. https://pubmed.ncbi.nlm.nih.gov/24550191/
Krishnan S, Cooper JA. Effect of dietary fatty acid composition on substrate utilization and body weight maintenance in humans. Eur J Nutr. 2014;53(3):691–710. https://pubmed.ncbi.nlm.nih.gov/24363161/
Jonnalagadda SS, Egan SK, Heimbach JT, et al. Fatty acid consumption pattern of Americans: 1987–1988 USDA Nationwide Food Consumption Survey. Nutr Res. 1995;15(12):1767–81. https://agris.fao.org/agris-search/search.do?recordID=US19970167025
Pimenta AS, Gaidhu MP, Habib S, et al. Prolonged exposure to palmitate impairs fatty acid oxidation despite activation of AMP-activated protein kinase in skeletal muscle cells. J Cell Physiol. 2008;217(2):478–85. https://pubmed.ncbi.nlm.nih.gov/18561258/
Chen YC, Cypess AM, Chen YC, et al. Measurement of human brown adipose tissue volume and activity using anatomic MR imaging and functional MR imaging. J Nucl Med. 2013;54(9):1584–7. https://pubmed.ncbi.nlm.nih.gov/23868958/
Darcy J, Tseng YH. ComBATing aging – does increased brown adipose tissue activity confer longevity? GeroScience. 2019;41(3):285–96. https://pubmed.ncbi.nlm.nih.gov/31230192/
Darcy J, Tseng YH. ComBATing aging – does increased brown adipose tissue activity confer longevity? GeroScience. 2019;41(3):285–96. https://pubmed.ncbi.nlm.nih.gov/31230192/
Ortega-Molina A, Efeyan A, Lopez-Guadamillas E, et al. Pten positively regulates brown adipose function, energy expenditure, and longevity. Cell Metab. 2012;15(3):382–94. https://pubmed.ncbi.nlm.nih.gov/22405073/
Vatner DE, Zhang J, Oydanich M, et al. Enhanced longevity and metabolism by brown adipose tissue with disruption of the regulator of G protein signaling 14. Aging Cell. 2018;17(4):e12751. https://pubmed.ncbi.nlm.nih.gov/29654651/
Hoffman JM, Valencak TG. Sex differences and aging: is there a role of brown adipose tissue? Mol Cell Endocrinol. 2021;531:111310. https://pubmed.ncbi.nlm.nih.gov/33989715/
Dong M, Lin J, Lim W, Jin W, Lee HJ. Role of brown adipose tissue in metabolic syndrome, aging, and cancer cachexia. Front Med. 2018;12(2):130–8. https://pubmed.ncbi.nlm.nih.gov/29119382/
Rogers NH. Brown adipose tissue during puberty and with aging. Ann Med. 2015;47(2):142–9. https://pubmed.ncbi.nlm.nih.gov/24888388/
Fuse S, Endo T, Tanaka R, et al. Effects of capsinoid intake on brown adipose tissue vascular density and resting energy expenditure in healthy, middle-aged adults: a randomized, double-blind, placebo-controlled study. Nutrients. 2020;12(9):E2676. https://pubmed.ncbi.nlm.nih.gov/32887379/
Smeets AJ, Janssens PL, Westerterp-Plantenga MS. Addition of capsaicin and exchange of carbohydrate with protein counteract energy intake restriction effects on fullness and energy expenditure. J Nutr. 2013;143(4):442–7. https://pubmed.ncbi.nlm.nih.gov/23406619/
Sugita J, Yoneshiro T, Hatano T, et al. Grains of paradise (Aframomum melegueta) extract activates brown adipose tissue and increases whole-body energy expenditure in men. Br J Nutr. 2013;110(4):733–8. https://pubmed.ncbi.nlm.nih.gov/23308394/
Maharlouei N, Tabrizi R, Lankarani KB, et al. The effects of ginger intake on weight loss and metabolic profiles among overweight and obese subjects: a systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr. 2018:1–14.; https://pubmed.ncbi.nlm.nih.gov/29393665/
Pellagra: secondary to antiobesity diet. Postgrad Med. 1955;17(3):37. https://pubmed.ncbi.nlm.nih.gov/14371224/
Afshin A, Forouzanfar MH, Reitsma BS, et al. Correspondence: health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377(1):13–27. https://pubmed.ncbi.nlm.nih.gov/28604169/
Berrigan D, Troiano RP, Graubard BI. BMI and mortality: the limits of epidemiological evidence. Lancet. 2016;388(10046):734–6. https://pubmed.ncbi.nlm.nih.gov/27423263/
Berrington de Gonzalez A, Hartge P, Cerhan JR, et al. Body-mass index and mortality among 1.46 million white adults. N Engl J Med. 2010;363(23):2211–9. https://pubmed.ncbi.nlm.nih.gov/21121834/
Afshin A, Forouzanfar MH, Reitsma BS, et al. Correspondence: health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377(1):13–27. https://pubmed.ncbi.nlm.nih.gov/28604169/
Aune D, Sen A, Prasad M, et al. BMI and all cause mortality: systematic review and non-linear dose-response meta-analysis of 230 cohort studies with 3.74 million deaths among 30.3 million participants. BMJ. 2016;353:i2156. https://pubmed.ncbi.nlm.nih.gov/27146380/
Greger M, Stone G. How Not to Die. Flatiron Books; 2015. https://www.worldcat.org/title/946602582
Rae DE, Ebrahim I, Roden LC. Sleep: a serious contender for the prevention of obesity and non-communicable diseases. JEMDSA. 2016;21(1):1–2. https://www.tandfonline.com/doi/full/10.1080/16089677.2016.1150574
Liu H, Chen A. Roles of sleep deprivation in cardiovascular dysfunctions. Life Sci. 2019;219:231–7. https://pubmed.ncbi.nlm.nih.gov/30630005/
Möller-Levet CS, Archer SN, Bucca G, et al. Effects of insufficient sleep on circadian rhythmicity and expression amplitude of the human blood transcriptome. Proc Natl Acad Sci U S A. 2013;110(12):E1132–41. https://pubmed.ncbi.nlm.nih.gov/23440187/
Calvin AD, Covassin N, Kremers WK, et al. Experimental sleep restriction causes endothelial dysfunction in healthy humans. J Am Heart Assoc. 2014;3(6):e001143. https://pubmed.ncbi.nlm.nih.gov/25424573/
Kohansieh M, Makaryus AN. Sleep deficiency and deprivation leading to cardiovascular disease. Int J Hypertens. 2015;2015:615681. https://pubmed.ncbi.nlm.nih.gov/26495139/
Calvin AD, Covassin N, Kremers WK, et al. Experimental sleep restriction causes endothelial dysfunction in healthy humans. J Am Heart Assoc. 2014;3(6):e001143. https://pubmed.ncbi.nlm.nih.gov/25424573/
Krueger PM, Friedman EM. Sleep duration in the United States: a cross-sectional population-based study. Am J Epidemiol. 2009;169(9):1052–63. https://pubmed.ncbi.nlm.nih.gov/19299406/
Golem DL, Martin-Biggers JT, Koenings MM, Davis KF, Byrd-Bredbenner C. An integrative review of sleep for nutrition professionals. Adv Nutr. 2014;5(6):742–59. https://pubmed.ncbi.nlm.nih.gov/25398735/
Shen J, Yang P, Luo X, et al. Green light extends Drosophila longevity. Exp Gerontol. 2021;147:111268. https://pubmed.ncbi.nlm.nih.gov/33539986/
Shen J, Yang P, Luo X, et al. Green light extends Drosophila longevity. Exp Gerontol. 2021;147:111268. https://pubmed.ncbi.nlm.nih.gov/33539986/
Bosman ES, Albert AY, Lui H, Dutz JP, Vallance BA. Skin exposure to narrow band ultraviolet (UVB) light modulates the human intestinal microbiome. Front Microbiol. 2019;10:2410. https://pubmed.ncbi.nlm.nih.gov/31708890/
Shen J, Yang P, Luo X, et al. Green light extends Drosophila longevity. Exp Gerontol. 2021;147:111268. https://pubmed.ncbi.nlm.nih.gov/33539986/
Li Q, Kobayashi M, Wakayama Y, et al. Effect of phytoncide from trees on human natural killer cell function. Int J Immunopathol Pharmacol. 2009;22(4):951–9. https://pubmed.ncbi.nlm.nih.gov/20074458/
Opperhuizen AL, Stenvers DJ, Jansen RD, Foppen E, Fliers E, Kalsbeek A. Light at night acutely impairs glucose tolerance in a time-, intensity– and wavelength-dependent manner in rats. Diabetologia. 2017;60(7):1333–43. https://pubmed.ncbi.nlm.nih.gov/28374068/
Kurina LM, McClintock MK, Chen JH, Waite LJ, Thisted RA, Lauderdale DS. Sleep duration and all-cause mortality: a critical review of measurement and associations. Ann Epidemiol. 2013;23(6):361–70. https://pubmed.ncbi.nlm.nih.gov/23622956/
He M, Deng X, Zhu Y, Huan L, Niu W. The relationship between sleep duration and all-cause mortality in the older people: an updated and dose-response meta-analysis. BMC Public Health. 2020;20(1):1179. https://pubmed.ncbi.nlm.nih.gov/32723316/
Yetish G, Kaplan H, Gurven M, et al. Natural sleep and its seasonal variations in three pre-industrial societies. Curr Biol. 2015;25(21):2862–8. https://pubmed.ncbi.nlm.nih.gov/26480842/
Shen X, Wu Y, Zhang D. Nighttime sleep duration, 24-hour sleep duration and risk of all-cause mortality among adults: a meta-analysis of prospective cohort studies. Sci Rep. 2016;6:21480. https://pubmed.ncbi.nlm.nih.gov/26900147/
García-Perdomo HA, Zapata-Copete J, Rojas-Cerón CA. Sleep duration and risk of all-cause mortality: a systematic review and meta-analysis. Epidemiol Psychiatr Sci. 2018;Jul 30:1–11. https://pubmed.ncbi.nlm.nih.gov/30058510/
Knutson KL, Turek FW. The U-shaped association between sleep and health: the 2 peaks do not mean the same thing. Sleep. 2006;29(7):878–9. https://pubmed.ncbi.nlm.nih.gov/16895253/
Grandner MA, Drummond SPA. Who are the long sleepers? Towards an understanding of the mortality relationship. Sleep Med Rev. 2007;11(5):341–60. https://pubmed.ncbi.nlm.nih.gov/17625932/
Kurina LM, McClintock MK, Chen JH, Waite LJ, Thisted RA, Lauderdale DS. Sleep duration and all-cause mortality: a critical review of measurement and associations. Ann Epidemiol. 2013;23(6):361–70. https://pubmed.ncbi.nlm.nih.gov/23622956/
Hirshkowitz M, Whiton K, Albert SM, et al. National Sleep Foundation’s sleep time duration recommendations: methodology and results summary. Sleep Health. 2015;1(1):40–3. https://pubmed.ncbi.nlm.nih.gov/29073412/
Pourmotabbed A, Boozari B, Babaei A, et al. Sleep and frailty risk: a systematic review and meta-analysis. Sleep Breath. 2020;24(3):1187–97. https://pubmed.ncbi.nlm.nih.gov/32215833/
Pourmotabbed A, Ghaedi E, Babaei A, et al. Sleep duration and sarcopenia risk: a systematic review and dose-response meta-analysis. Sleep Breath. 2020;24(4):1267–78. https://pubmed.ncbi.nlm.nih.gov/31832982/
Schwarz EI, Puhan MA, Schlatzer C, Stradling JR, Kohler M. Effect of CPAP therapy on endothelial function in obstructive sleep apnoea: a systematic review and meta-analysis. Respirology. 2015;20(6):889–95. https://pubmed.ncbi.nlm.nih.gov/26073295/
Bjorvatn B, Fiske E, Pallesen S. A self-help book is better than sleep hygiene advice for insomnia: a randomized controlled comparative study. Scand J Psychol. 2011;52(6):580–5. https://pubmed.ncbi.nlm.nih.gov/21790620/
Ancoli-Israel S. Sleep problems in older adults: putting myths to bed. Geriatrics. 1997;52(1):20–30. https://pubmed.ncbi.nlm.nih.gov/9003201/
Miner B, Kryger MH. Sleep in the aging population. Sleep Med Clin. 2017;12(1):31–8. https://pubmed.ncbi.nlm.nih.gov/28159095/
Lovato N, Lack L. Insomnia and mortality: a meta-analysis. Sleep Med Rev. 2019;43:71–83. https://pubmed.ncbi.nlm.nih.gov/30529432/
Barclay NL, Kocevska D, Bramer WM, Van Someren EJW, Gehrman P. The heritability of insomnia: a meta-analysis of twin studies. Genes Brain Behav. 2021;20(4):e12717. https://pubmed.ncbi.nlm.nih.gov/33222383/
Machado FV, Louzada LL, Cross NE, Camargos EF, Dang-Vu TT, Nóbrega OT. More than a quarter century of the most prescribed sleeping pill: systematic review of zolpidem use by older adults. Exp Gerontol. 2020;136:110962. https://pubmed.ncbi.nlm.nih.gov/32360985/
Kripke DF, Langer RD, Kline LE. Hypnotics’ association with mortality or cancer: a matched cohort study. BMJ Open. 2012;2(1):1–10. https://pubmed.ncbi.nlm.nih.gov/22371848/
Baber R. Climacteric commentaries. Better sleep but higher mortality risk. Climacteric. 2012;15(4):401. https://pubmed.ncbi.nlm.nih.gov/22950121/
Kripke DF, Langer RD, Kline LE. Hypnotics’ association with mortality or cancer: a matched cohort study. BMJ Open. 2012;2(1):1–10. https://pubmed.ncbi.nlm.nih.gov/22371848/
Rabin RC. New worries about sleeping pills. The New York Times: Well. https://well.blogs.nytimes.com/2012/03/12/new-worries-about-sleeping-pills. Published March 12, 2012. Accessed April 17, 2019.; https://well.blogs.nytimes.com/2012/03/12/new-worries-about-sleeping-pills
Kripke DF. Mortality risk of hypnotics: strengths and limits of evidence. Drug Saf. 2016;39(2):93–107. https://pubmed.ncbi.nlm.nih.gov/26563222/
Bianchi MT, Thomas RJ, Ellenbogen JM. Hypnotics and mortality risk. J Clin Sleep Med. 2012;8(4):351–2. https://pubmed.ncbi.nlm.nih.gov/22893762/
Kripke DF, Langer RD, Kline LE. Do no harm: not even to some degree. J Clin Sleep Med. 2012;8(4):353–4. https://pubmed.ncbi.nlm.nih.gov/22893763/
Huedo-Medina TB, Kirsch I, Middlemass J, Klonizakis M, Siriwardena AN. Effectiveness of non-benzodiazepine hypnotics in treatment of adult insomnia: meta-analysis of data submitted to the Food and Drug Administration. BMJ. 2012;345. https://pubmed.ncbi.nlm.nih.gov/23248080/
Buscemi N, Vandermeer B, Friesen C, et al. The efficacy and safety of drug treatments for chronic insomnia in adults: a meta-analysis of RCTs. J Gen Intern Med. 2007;22(9):1335–50. https://pubmed.ncbi.nlm.nih.gov/17619935/
Kripke DF, Langer RD, Kline LE. Do no harm: not even to some degree. J Clin Sleep Med. 2012;08(04):353–4. https://pubmed.ncbi.nlm.nih.gov/22893763/
Matheson E, Hainer BL. Insomnia: pharmacologic therapy. Am Fam Physician. 2017;96(1):29–35. https://pubmed.ncbi.nlm.nih.gov/28671376/
Brown RF, Thorsteinsson EB, Smithson M, Birmingham CL, Aljarallah H, Nolan C. Can body temperature dysregulation explain the co-occurrence between overweight/obesity, sleep impairment, late-night eating, and a sedentary lifestyle? Eat Weight Disord. 2017;22(4):599–608. https://pubmed.ncbi.nlm.nih.gov/28929462/
Brown RF, Thorsteinsson EB, Smithson M, Birmingham CL, Aljarallah H, Nolan C. Can body temperature dysregulation explain the co-occurrence between overweight/obesity, sleep impairment, late-night eating, and a sedentary lifestyle? Eat Weight Disord. 2017;22(4):599–608. https://pubmed.ncbi.nlm.nih.gov/28929462/
Sung EJ, Tochihara Y. Effects of bathing and hot footbath on sleep in winter. J Physiol Anthropol Appl Human Sci. 2000;19(1):21–7. https://pubmed.ncbi.nlm.nih.gov/10979246/
Aghamohammadi V, Salmani R, Ivanbagha R, Effati-Daryani F, Nasiri K. Footbath as a safe, simple, and non-pharmacological method to improve sleep quality of menopausal women. Res Nurs Health. 2020;43(6):621–8. https://pubmed.ncbi.nlm.nih.gov/33112004/
Haghayegh S, Khoshnevis S, Smolensky MH, Diller KR, Castriotta RJ. Before-bedtime passive body heating by warm shower or bath to improve sleep: a systematic review and meta-analysis. Sleep Med Rev. 2019;46:124–35. https://pubmed.ncbi.nlm.nih.gov/31102877/
Haghayegh S, Khoshnevis S, Smolensky MH, Diller KR, Castriotta RJ. Before-bedtime passive body heating by warm shower or bath to improve sleep: a systematic review and meta-analysis. Sleep Med Rev. 2019;46:124–35. https://pubmed.ncbi.nlm.nih.gov/31102877/
Liao WC, Wang L, Kuo CP, Lo C, Chiu MJ, Ting H. Effect of a warm footbath before bedtime on body temperature and sleep in older adults with good and poor sleep: an experimental crossover trial. Int J Nurs Stud. 2013;50(12):1607–16. https://pubmed.ncbi.nlm.nih.gov/23669188/
Kräuchi K, Cajochen C, Werth E, Wirz-Justice A. Warm feet promote the rapid onset of sleep. Nature. 1999;401(6748):36–7. https://pubmed.ncbi.nlm.nih.gov/10485703/
Ko Y, Lee JY. Effects of feet warming using bed socks on sleep quality and thermoregulatory responses in a cool environment. J Physiol Anthropol. 2018;37(1):13. https://pubmed.ncbi.nlm.nih.gov/29699592/
Matheson E, Hainer BL. Insomnia: pharmacologic therapy. Am Fam Physician. 2017;96(1):29–35. https://pubmed.ncbi.nlm.nih.gov/28671376/
Morin CM, Inoue Y, Kushida C, et al. Endorsement of European guideline for the diagnosis and treatment of insomnia by the World Sleep Society. Sleep Med. 2021;81:124–6. https://pubmed.ncbi.nlm.nih.gov/33667998/
Fatemeh G, Sajjad M, Niloufar R, Neda S, Leila S, Khadijeh M. Effect of melatonin supplementation on sleep quality: a systematic review and meta-analysis of randomized controlled trials. J Neurol. 2022;269(1):205–16. https://pubmed.ncbi.nlm.nih.gov/33417003/
Brzezinski A, Vangel MG, Wurtman RJ, et al. Effects of exogenous melatonin on sleep: a meta-analysis. Sleep Med Rev. 2005;9(1):41–50. https://pubmed.ncbi.nlm.nih.gov/15649737/
Williamson B, Tomlinson A, Naylor S, Gleich G. Contaminants in commercial preparations of melatonin. Mayo Clin Proc. 1997;72(11):1094–5. https://pubmed.ncbi.nlm.nih.gov/9374988/
Poeggeler B. Melatonin, aging, and age-related diseases: perspectives for prevention, intervention, and therapy. Endocrine. 2005;27(2):201–12. https://pubmed.ncbi.nlm.nih.gov/16217133/
Oaknin-Bendahan S, Anis Y, Nir I, Zisapel N. Effects of long-term administration of melatonin and a putative antagonist on the ageing rat. Neuroreport. 1995;6(5):785–8. https://pubmed.ncbi.nlm.nih.gov/7605949/
Kim J, Lee SL, Kang I, et al. Natural products from single plants as sleep aids: a systematic review. J Med Food. 2018;21(5):433–44. https://pubmed.ncbi.nlm.nih.gov/29356580/
Taibi DM, Landis CA, Petry H, Vitiello MV. A systematic review of valerian as a sleep aid: safe but not effective. Sleep Med Rev. 2007;11(3):209–30. https://pubmed.ncbi.nlm.nih.gov/17517355/
Afrasiabian F, Ardakani MM, Rahmani K, et al. Aloysia citriodora Palau (lemon verbena) for insomnia patients: a randomized, double-blind, placebo-controlled clinical trial of efficacy and safety. Phytother Res. 2019;33(2):350–9. https://pubmed.ncbi.nlm.nih.gov/30450627/
Zick SM, Wright BD, Sen A, Arnedt JT. Preliminary examination of the efficacy and safety of a standardized chamomile extract for chronic primary insomnia: a randomized placebo-controlled pilot study. BMC Complement Altern Med. 2011;11:78. https://pubmed.ncbi.nlm.nih.gov/21939549/
Hieu TH, Dibas M, Dila KAS, et al. Therapeutic efficacy and safety of chamomile for state anxiety, generalized anxiety disorder, insomnia, and sleep quality: a systematic review and meta-analysis of randomized trials and quasi-randomized trials. Phytother Res. 2019;33(6):1604–15. https://pubmed.ncbi.nlm.nih.gov/31006899/
St-Onge MP, Roberts A, Shechter A, Choudhury AR. Fiber and saturated fat are associated with sleep arousals and slow wave sleep. J Clin Sleep Med. 2016;12(1):19–24. https://pubmed.ncbi.nlm.nih.gov/26156950/
Grandner MA, Kripke DF, Naidoo N, Langer RD. Relationships among dietary nutrients and subjective sleep, objective sleep, and napping in women. Sleep Med. 2010;11(2):180. https://pubmed.ncbi.nlm.nih.gov/20005774/
McClernon FJ, Yancy WS, Eberstein JA, Atkins RC, Westman EC. The effects of a low-carbohydrate ketogenic diet and a low-fat diet on mood, hunger, and other self-reported symptoms. Obesity (Silver Spring). 2007;15(1):182–7. https://pubmed.ncbi.nlm.nih.gov/17228046/
Lana A, Struijk EA, Arias-Fernandez L, et al. Habitual meat consumption and changes in sleep duration and quality in older adults. Aging Dis. 2019;10(2):267–77. https://pubmed.ncbi.nlm.nih.gov/31011478/
Hansen AL, Dahl L, Olson G, et al. Fish consumption, sleep, daily functioning, and heart rate variability. J Clin Sleep Med. 2014;10(5):567–75. https://pubmed.ncbi.nlm.nih.gov/24812543/
Lana A, Struijk EA, Arias-Fernandez L, et al. Habitual meat consumption and changes in sleep duration and quality in older adults. Aging Dis. 2019;10(2):267–77. https://pubmed.ncbi.nlm.nih.gov/31011478/
Wurtman RJ, Wurtman JJ, Regan MM, McDermott JM, Tsay RH, Breu JJ. Effects of normal meals rich in carbohydrates or proteins on plasma tryptophan and tyrosine ratios. Am J Clin Nutr. 2003;77(1):128–32. https://pubmed.ncbi.nlm.nih.gov/12499331/
Beezhold BL, Johnston CS. Restriction of meat, fish, and poultry in omnivores improves mood: a pilot randomized controlled trial. Nutr J. 2012;11:9. https://pubmed.ncbi.nlm.nih.gov/22333737/
Merrill RM, Aldana SG, Greenlaw RL, Diehl HA, Salberg A. The effects of an intensive lifestyle modification program on sleep and stress disorders. J Nutr Health Aging. 2007;11(3):242–8. https://pubmed.ncbi.nlm.nih.gov/17508101/
St-Onge MP, Crawford A, Aggarwal B. Plant-based diets: reducing cardiovascular risk by improving sleep quality? Curr Sleep Med Rep. 2018;4(1):74–8. https://pubmed.ncbi.nlm.nih.gov/29910998/
Harsha SN, Anilakumar KR. Anxiolytic property of Lactuca sativa, effect on anxiety behaviour induced by novel food and height. Asian Pac J Trop Med. 2013;6(7):532–6. https://pubmed.ncbi.nlm.nih.gov/23768824/
Gonzálex-lima F, Valedón A, Stiehil WL. Depressant pharmacological effects of a component isolated from lettuce, Lactuca sativa. Int J Crude Drug Res. 1986;24(3):154–66. https://www.tandfonline.com/doi/abs/10.3109/13880208609060893
Kim H-W, Suh HJ, Choi H-S, Hong K-B, Jo K. Effectiveness of the sleep enhancement by green romaine lettuce (Lactuca sativa) in a rodent model. Biol Pharm Bull. 2019;42(10):1726–32. https://pubmed.ncbi.nlm.nih.gov/31582660/
Kim HD, Hong K-B, Noh DO, Suh HJ. Sleep-inducing effect of lettuce (Lactuca sativa) varieties on pentobarbital-induced sleep. Food Sci Biotechnol. 2017;26(3):807–14. https://pubmed.ncbi.nlm.nih.gov/30263607/
Pour ZS, Hosseinkhani A, Asadi N, et al. Double-blind randomized placebo-controlled trial on efficacy and safety of Lactuca sativa L. seeds on pregnancy-related insomnia. J Ethnopharmacol. 2018;227:176–80. https://pubmed.ncbi.nlm.nih.gov/30172900/
Thomas T. Perls, MD, MPH, FACP. Boston University School of Medicine website. https://www.bumc.bu.edu/busm/profile/thomas-perls/. Accessesd April 3, 2022.; https://www.bumc.bu.edu/busm/profile/thomas-perls/
Sebastiani P, Perls TT. The genetics of extreme longevity: lessons from the New England Centenarian Study. Front Gene. 2012;3:277. https://pubmed.ncbi.nlm.nih.gov/23226160/
Tomiyama AJ. Stress and obesity. Annu Rev Psychol. 2019;70:703–18. https://pubmed.ncbi.nlm.nih.gov/29927688/
Baxter AJ, Scott KM, Ferrari AJ, Norman RE, Vos T, Whiteford HA. Challenging the myth of an “epidemic” of common mental disorders: trends in the global prevalence of anxiety and depression between 1990 and 2010. Depress Anxiety. 2014;31(6):506–16. https://pubmed.ncbi.nlm.nih.gov/24448889/
Adam TC, Epel ES. Stress, eating and the reward system. Physiol Behav. 2007;91(4):449–58. https://pubmed.ncbi.nlm.nih.gov/17543357/
Tomiyama A. Stress and obesity. Annu Rev Psychol. 2019;70:703–18. https://pubmed.ncbi.nlm.nih.gov/29927688/
Zellner DA, Loaiza S, Gonzalez Z, et al. Food selection changes under stress. Physiol Behav. 2006;87(4):789–93. https://pubmed.ncbi.nlm.nih.gov/16519909/
Buchmann AF, Laucht M, Schmid B, Wiedemann K, Mann K, Zimmermann US. Cigarette craving increases after a psychosocial stress test and is related to cortisol stress response but not to dependence scores in daily smokers. J Psychopharmacol. 2010;24(2):247–55. https://pubmed.ncbi.nlm.nih.gov/18957475/
Magrys SA, Olmstead MC. Acute stress increases voluntary consumption of alcohol in undergraduates. Alcohol Alcohol. 2015;50(2):213–8. https://pubmed.ncbi.nlm.nih.gov/25557606/
Sinha R, Garcia M, Paliwal P, Kreek MJ, Rounsaville BJ. Stress-induced cocaine craving and hypothalamic-pituitary-adrenal responses are predictive of cocaine relapse outcomes. Arch Gen Psychiatry. 2006;63(3):324–31. https://pubmed.ncbi.nlm.nih.gov/16520439/
Rutters F, Pilz S, Koopman AD, et al. The association between psychosocial stress and mortality is mediated by lifestyle and chronic diseases: the Hoorn Study. Soc Sci Med. 2014;118:166–72. https://pubmed.ncbi.nlm.nih.gov/25137635/
Rodgers J, Cuevas AG, Williams DR, Kawachi I, Subramanian SV. The relative contributions of behavioral, biological, and psychological risk factors in the association between psychosocial stress and all-cause mortality among middle– and older-aged adults in the USA. Geroscience. 2021;43(2):655–72. https://pubmed.ncbi.nlm.nih.gov/33511488/
Strøm A, Jensen RA. Mortality from circulatory diseases in Norway 1940–1945. Lancet. 1951;1(6647):126–9. https://pubmed.ncbi.nlm.nih.gov/14795790/
Keys A. Coronary heart disease – the global picture. Atherosclerosis. 1975;22(2):149–92. https://pubmed.ncbi.nlm.nih.gov/1103902/
Malmros H. The relation of nutrition to health; a statistical study of the effect of the war-time on arteriosclerosis https://pubmed.ncbi.nlm.nih.gov/14789502/
Cronkite W. Poverty and want rip Netherlands; troops say Dutch suffered hunger https://www.nytimes.com/1944/09/29/archives/poverty-and-want-rip-netherlands-troops-say-dutch-suffered-hunger.html
Diet and stress in vascular disease. JAMA. 1961;176(9):806–7. https://pubmed.ncbi.nlm.nih.gov/14447689/
Hitchcott PK, Fastame MC, Penna MP. More to Blue Zones than long life: positive psychological characteristics. Health Risk Soc. 2018;20(3–4):163–81. https://www.tandfonline.com/doi/full/10.1080/13698575.2018.1496233
Manzoli L, Villari P, M Pirone G, Boccia A. Marital status and mortality in the elderly: a systematic review and meta-analysis. Soc Sci Med. 2007;64(1):77–94. https://pubmed.ncbi.nlm.nih.gov/17011690/
Ennis J, Majid U. “Death from a broken heart”: a systematic review of the relationship between spousal bereavement and physical and physiological health outcomes. Death Stud. 2021;45(7):538–51. https://pubmed.ncbi.nlm.nih.gov/31535594/
Friedmann E, Katcher AH, Lynch JJ, Thomas SA. Animal companions and one-year survival of patients after discharge from a coronary care unit. Public Health Rep. 1980;95(4):307–12. https://pubmed.ncbi.nlm.nih.gov/6999524/
Manzoli L, Villari P, M Pirone G, Boccia A. Marital status and mortality in the elderly: a systematic review and meta-analysis. Soc Sci Med. 2007;64(1):77–94. https://pubmed.ncbi.nlm.nih.gov/17011690/
Manzoli L, Villari P, M Pirone G, Boccia A. Marital status and mortality in the elderly: a systematic review and meta-analysis. Soc Sci Med. 2007;64(1):77–94. https://pubmed.ncbi.nlm.nih.gov/17011690/
Holt-Lunstad J, Smith TB, Baker M, Harris T, Stephenson D. Loneliness and social isolation as risk factors for mortality: a meta-analytic review. Perspect Psychol Sci. 2015;10(2):227–37. https://pubmed.ncbi.nlm.nih.gov/25910392/
Rico-Uribe LA, Caballero FF, Martín-María N, Cabello M, Ayuso-Mateos JL, Miret M. Association of loneliness with all-cause mortality: a meta-analysis. PLoS One. 2018;13(1):e0190033. https://pubmed.ncbi.nlm.nih.gov/29300743/
Shor E, Roelfs DJ. Social contact frequency and all-cause mortality: a meta-analysis and meta-regression. Soc Sci Med. 2015;128:76–86. https://pubmed.ncbi.nlm.nih.gov/25594955/
Stickley A, Koyanagi A, Roberts B, et al. Loneliness: its correlates and association with health behaviours and outcomes in nine countries of the former Soviet Union. PLoS One. 2013;8(7):e67978. https://pubmed.ncbi.nlm.nih.gov/23861843/
Holt-Lunstad J, Smith TB, Baker M, Harris T, Stephenson D. Loneliness and social isolation as risk factors for mortality: a meta-analytic review. Perspect Psychol Sci. 2015;10(2):227–37. https://pubmed.ncbi.nlm.nih.gov/25910392/
Kramer CK, Mehmood S, Suen RS. Dog ownership and survival: a systematic review and meta-analysis. Circ Cardiovasc Qual Outcomes. 2019;12(10):e005554. https://pubmed.ncbi.nlm.nih.gov/31592726/
Nagasawa M, Mitsui S, En S, et al. Oxytocin-gaze positive loop and the coevolution of human-dog bonds. Science. 2015;348(6232):333–6. https://pubmed.ncbi.nlm.nih.gov/25883356/
Friedmann E, Katcher AH, Lynch JJ, Thomas SA. Animal companions and one-year survival of patients after discharge from a coronary care unit. Public Health Rep. 1980;95(4):307–12. https://pubmed.ncbi.nlm.nih.gov/6999524/
Herzog H. The impact of pets on human health and psychological well-being: fact, fiction, or hypothesis? Curr Dir Psychol Sci. 2011;20(4):236–9. https://journals.sagepub.com/doi/10.1177/0963721411415220
Kazi DS. Who is rescuing whom? Dog ownership and cardiovascular health. Circ Cardiovasc Qual Outcomes. 2019;12(10):e005887. https://pubmed.ncbi.nlm.nih.gov/31592727/
Kazi DS. Who is rescuing whom? Dog ownership and cardiovascular health. Circ Cardiovasc Qual Outcomes. 2019;12(10):e005887. https://pubmed.ncbi.nlm.nih.gov/31592727/
Ko HJ, Youn CH, Kim SH, Kim SY. Effect of pet insects on the psychological health of community-dwelling elderly people: a single-blinded, randomized, controlled trial. Gerontology. 2016;62(2):200–9. https://pubmed.ncbi.nlm.nih.gov/26383099/
Puterbaugh JS. The emperor’s tailors: the failure of the medical weight loss paradigm and its causal role in the obesity of America. Diabetes Obes Metab. 2009;11(6):557–70. https://pubmed.ncbi.nlm.nih.gov/19383033/
Berg J, Seyedsadjadi N, Grant R. Increased consumption of plant foods is associated with increased bone mineral density. J Nutr Health Aging. 2020;24(4):388–97. https://pubmed.ncbi.nlm.nih.gov/32242206/
Gupta T, Das N, Imran S. The prevention and therapy of osteoporosis: a review on emerging trends from hormonal therapy to synthetic drugs to plant-based bioactives. J Diet Suppl. 2019;16(6):699–713. https://pubmed.ncbi.nlm.nih.gov/29985715/
Berg J, Seyedsadjadi N, Grant R. Increased consumption of plant foods is associated with increased bone mineral density. J Nutr Health Aging. 2020;24(4):388–97. https://pubmed.ncbi.nlm.nih.gov/32242206/
Berg J, Seyedsadjadi N, Grant R. Increased consumption of plant foods is associated with increased bone mineral density. J Nutr Health Aging. 2020;24(4):388–97. https://pubmed.ncbi.nlm.nih.gov/32242206/
Lorentzon M, Cummings SR. Osteoporosis: the evolution of a diagnosis. J Intern Med. 2015;277(6):650–61. https://pubmed.ncbi.nlm.nih.gov/25832448/
Gupta T, Das N, Imran S. The prevention and therapy of osteoporosis: a review on emerging trends from hormonal therapy to synthetic drugs to plant-based bioactives. J Diet Suppl. 2019;16(6):699–713. https://pubmed.ncbi.nlm.nih.gov/29985715/
Sahota O, Masud T. Osteoporosis: fact, fiction, fallacy and the future. Age Ageing. 1999;28(5):425–8. https://pubmed.ncbi.nlm.nih.gov/10529034/
Michaëlsson K, Melhus H, Ferm H, Ahlbom A, Pedersen NL. Genetic liability to fractures in the elderly. Arch Intern Med. 2005;165(16):1825–30. https://pubmed.ncbi.nlm.nih.gov/16157825/
Kanis JA, Odén A, McCloskey EV, et al. A systematic review of hip fracture incidence and probability of fracture worldwide. Osteoporos Int. 2012;23(9):2239–56. https://pubmed.ncbi.nlm.nih.gov/22419370/
Final recommendation statement: osteoporosis to prevent fractures: screening. U.S. Preventative Services Task Force website. https://www.uspreventiveservicestaskforce.org/uspstf/recommendation/osteoporosis-screening. Published June 26, 2018. Accessed March 6, 2022.; https://www.uspreventiveservicestaskforce.org/uspstf/recommendation/osteoporosis-screening
Luo H, Fan Q, Xiao S, Chen K. Changes in proton pump inhibitor prescribing trend over the past decade and pharmacists’ effect on prescribing practice at a tertiary hospital. BMC Health Serv Res. 2018;18(1):537. https://pubmed.ncbi.nlm.nih.gov/29996830/
Poly TN, Islam MM, Yang HC, Wu CC, Li YCJ. Proton pump inhibitors and risk of hip fracture: a meta-analysis of observational studies. Osteoporos Int. 2019;30(1):103–14. https://pubmed.ncbi.nlm.nih.gov/30539272/
Xun X, Yin Q, Fu Y, He X, Dong Z. Proton pump inhibitors and the risk of community-acquired pneumonia: an updated meta-analysis. Ann Pharmacother. 2022;56(5):524–32. https://pubmed.ncbi.nlm.nih.gov/34425689/
Moayyedi P, Lewis MA. Proton pump inhibitors and dementia: deciphering the data. Am J Gastroenterol. 2017;112(12):1809–11. https://pubmed.ncbi.nlm.nih.gov/29215629/
Vengrus CS, Delfino VD, Bignardi PR. Proton pump inhibitors use and risk of chronic kidney disease and end-stage renal disease. Minerva Urol Nephrol. 2021;73(4):462–70. https://pubmed.ncbi.nlm.nih.gov/33769018/
D’Silva KM, Mehta R, Mitchell M, et al. Proton pump inhibitor use and risk for recurrent Clostridioides difficile infection: a systematic review and meta-analysis. Clin Microbiol Infect. 2021;27(5):697–703. https://pubmed.ncbi.nlm.nih.gov/33465501/
Salvo EM, Ferko NC, Cash SB, Gonzalez A, Kahrilas PJ. Umbrella review of 42 systematic reviews with meta-analyses: the safety of proton pump inhibitors. Aliment Pharmacol Ther. 2021;54(2):129–43. https://pubmed.ncbi.nlm.nih.gov/34114655/
Sun S, Cui Z, Zhou M, et al. Proton pump inhibitor monotherapy and the risk of cardiovascular events in patients with gastro-esophageal reflux disease: a meta-analysis. Neurogastroenterol Motil. 2017;29(2):e12926. https://pubmed.ncbi.nlm.nih.gov/27577963/
Ben-Eltriki M, Green CJ, Maclure M, Musini V, Bassett KL, Wright JM. Do proton pump inhibitors increase mortality? A systematic review and in-depth analysis of the evidence. Pharmacol Res Perspect. 2020;8(5):e00651. https://pubmed.ncbi.nlm.nih.gov/32996701/
Safer DJ. Overprescribed medications for US adults: four major examples. J Clin Med Res. 2019;11(9):617–22. https://pubmed.ncbi.nlm.nih.gov/31523334/
Safer DJ. Overprescribed medications for US adults: four major examples. J Clin Med Res. 2019;11(9):617–22. https://pubmed.ncbi.nlm.nih.gov/31523334/
Ness-Jensen E, Hveem K, El-Serag H, Lagergren J. Lifestyle intervention in gastroesophageal reflux disease. Clin Gastroenterol Hepatol. 2016;14(2):175–82.e1–3. https://pubmed.ncbi.nlm.nih.gov/25956834/
Andrici J, Cox MR, Eslick GD. Cigarette smoking and the risk of Barrett’s esophagus: a systematic review and meta-analysis. J Gastroenterol Hepatol. 2013;28(8):1258–73. https://pubmed.ncbi.nlm.nih.gov/23611750/
Fan WJ, Hou YT, Sun XH, et al. Effect of high-fat, standard, and functional food meals on esophageal and gastric pH in patients with gastroesophageal reflux disease and healthy subjects. J Dig Dis. 2018;19(11):664–73. https://pubmed.ncbi.nlm.nih.gov/30270576/
Katz PO, Gerson LB, Vela MF. Guidelines for the diagnosis and management of gastroesophageal reflux disease. Am J Gastroenterol. 2013;108(3):308–28. https://pubmed.ncbi.nlm.nih.gov/23419381/
Newberry C, Lynch K. The role of diet in the development and management of gastroesophageal reflux disease: why we feel the burn. J Thorac Dis. 2019;11(Suppl 12):S1594–601. https://pubmed.ncbi.nlm.nih.gov/31489226/
Jung JG, Kang HW. Vegetarianism and the risk of gastroesophageal reflux disease. In: Vegetarian and Plant-Based Diets in Health and Disease Prevention. Elsevier; 2017:463–72. https://www.sciencedirect.com/science/article/abs/pii/B9780128039687000253?via%3Dihub
Law MR, Hackshaw AK. A meta-analysis of cigarette smoking, bone mineral density and risk of hip fracture: recognition of a major effect. BMJ. 1997;315(7112):841–6. https://pubmed.ncbi.nlm.nih.gov/9353503/
Patel RA, Wilson RF, Patel PA, Palmer RM. The effect of smoking on bone healing. Bone Joint Res. 2013;2(6):102–11. https://pubmed.ncbi.nlm.nih.gov/23836474/
Kim JH, Patel S. Is it worth discriminating against patients who smoke? A systematic literature review on the effects of tobacco use in foot and ankle surgery. J Foot Ankle Surg. 2017;56(3):594–9. https://pubmed.ncbi.nlm.nih.gov/28476393/
Bourne D, Plinke W, Hooker ER, Nielson CM. Cannabis use and bone mineral density: NHANES 2007–2010. Arch Osteoporos. 2017;12(1):29. https://pubmed.ncbi.nlm.nih.gov/28286929/
Sophocleous A, Robertson R, Ferreira NB, McKenzie J, Fraser WD, Ralston SH. Heavy cannabis use is associated with low bone mineral density and an increased risk of fractures. Am J Med. 2017;130(2):214–21. https://pubmed.ncbi.nlm.nih.gov/27593602/
Cosman F, de Beur SJ, LeBoff MS, et al. Clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int. 2014;25(10):2359–81. https://pubmed.ncbi.nlm.nih.gov/25182228/
Wright J. Marketing disease: is osteoporosis an example of “disease mongering”? Br J Nurs. 2009;18(17):1064–7. https://pubmed.ncbi.nlm.nih.gov/19798007/
Hudson B, Zarifeh A, Young L, Wells JE. Patients’ expectations of screening and preventive treatments. Ann Fam Med. 2012;10(6):495–502. https://pubmed.ncbi.nlm.nih.gov/23149525/
Black DM, Rosen CJ. Postmenopausal osteoporosis. N Engl J Med. 2016;374(21):2096–7. https://pubmed.ncbi.nlm.nih.gov/26789873/
Lems WF, Raterman HG. Critical issues and current challenges in osteoporosis and fracture prevention. An overview of unmet needs. Ther Adv Musculoskelet Dis. 2017;9(12):299–316. https://pubmed.ncbi.nlm.nih.gov/29201155/
Kolata G. Fearing drugs’ rare side effects, millions take their chances with osteoporosis. The New York Times. https://www.nytimes.com/2016/06/02/health/osteoporosis-drugs-bones.html. Published June 1, 2016. Accessed March 6, 2022.; https://www.nytimes.com/2016/06/02/health/osteoporosis-drugs-bones.html
Sales LP, Pinto AJ, Rodrigues SF, et al. Creatine supplementation (3 g/d) and bone health in older women: a 2-year, randomized, placebo-controlled trial. J Gerontol A Biol Sci Med Sci. 2020;75(5):931–8. https://pubmed.ncbi.nlm.nih.gov/31257405/
Candow DG, Forbes SC, Kirk B, Duque G. Current evidence and possible future applications of creatine supplementation for older adults. Nutrients. 2021;13(3):745. https://pubmed.ncbi.nlm.nih.gov/33652673/
NIH Consensus Development Panel on Osteoporosis Prevention, Diagnosis, and Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA. 2001;285(6):785–95. https://pubmed.ncbi.nlm.nih.gov/11440324/
Nestle M, Nesheim MC. To supplement or not to supplement: the U.S. Preventive Services Task Force recommendations on calcium and vitamin D. Ann Intern Med. 2013;158(9):701–2. https://pubmed.ncbi.nlm.nih.gov/23440174/
Grossman DC, Curry SJ, Owens DK, et al. Vitamin D, calcium, or combined supplementation for the primary prevention of fractures in community-dwelling adults: US Preventive Services Task Force recommendation statement. JAMA. 2018;319(15):1592–9. https://pubmed.ncbi.nlm.nih.gov/29677309/
Bolland MJ, Grey A, Reid IR. Calcium supplements and cardiovascular risk: 5 years on. Ther Adv Drug Saf. 2013;4(5):199–210. https://pubmed.ncbi.nlm.nih.gov/25114781/
Reid IR, Bristow SM, Bolland MJ. Calcium supplements: benefits and risks. J Intern Med. 2015;278(4):354–68. https://pubmed.ncbi.nlm.nih.gov/26174589/
Reid IR, Bolland MJ. Risk factors: calcium supplements and cardiovascular risk. Nat Rev Cardiol. 2012;9(9):497–8. https://pubmed.ncbi.nlm.nih.gov/22776986/
Bischoff-Ferrari HA, Dawson-Hughes B, Baron JA, et al. Calcium intake and hip fracture risk in men and women: a meta-analysis of prospective cohort studies and randomized controlled trials. Am J Clin Nutr. 2007;86(6):1780–90. https://pubmed.ncbi.nlm.nih.gov/18065599/
Bolland MJ, Grey A, Reid IR. Calcium supplements and cardiovascular risk: 5 years on. Ther Adv Drug Saf. 2013;4(5):199–210. https://pubmed.ncbi.nlm.nih.gov/25114781/
Willett WC, Ludwig DS. Milk and health. N Engl J Med. 2020;382(7):644–54. https://pubmed.ncbi.nlm.nih.gov/32053300/
Dawson-Hughes B, Jacques P, Shipp C. Dietary calcium intake and bone loss from the spine in healthy postmenopausal women. Am J Clin Nutr. 1987;46(4):685–7. https://pubmed.ncbi.nlm.nih.gov/3661483/
Sanders KM, Stuart AL, Williamson EJ, et al. Annual high-dose oral vitamin D and falls and fractures in older women: a randomized controlled trial. JAMA. 2010;303(18):1815–22. https://pubmed.ncbi.nlm.nih.gov/20460620/
Ginde AA, Blatchford P, Breese K, et al. High-dose monthly vitamin D for prevention of acute respiratory infection in older long-term care residents: a randomized clinical trial. J Am Geriatr Soc. 2017;65(3):496–503. https://pubmed.ncbi.nlm.nih.gov/27861708/
Bischoff-Ferrari HA, Dawson-Hughes B, Orav EJ, et al. Monthly high-dose vitamin D treatment for the prevention of functional decline: a randomized clinical trial. JAMA Intern Med. 2016;176(2):175–83. https://pubmed.ncbi.nlm.nih.gov/26747333/
Smith LM, Gallagher JC, Suiter C. Medium doses of vitamin D decrease falls and higher doses of daily vitamin D3 increase falls: a randomized clinical trial. J Steroid Biochem Mol Biol. 2017;173:317–22. https://pubmed.ncbi.nlm.nih.gov/28323044/
Burt LA, Billington EO, Rose MS, Raymond DA, Hanley DA, Boyd SK. Effect of high-dose vitamin D supplementation on volumetric bone density and bone strength: a randomized clinical trial. JAMA. 2019;322(8):736–45. https://pubmed.ncbi.nlm.nih.gov/31454046/
Burt LA, Billington EO, Rose MS, Kremer R, Hanley DA, Boyd SK. Adverse effects of high-dose vitamin D supplementation on volumetric bone density are greater in females than males. J Bone Miner Res. 2020;35(12):2404–14. https://pubmed.ncbi.nlm.nih.gov/31454046/
Iuliano S, Hill TR. Dairy foods and bone health throughout the lifespan: a critical appraisal of the evidence. Br J Nutr. 2019;121(7):763–72. https://pubmed.ncbi.nlm.nih.gov/30638442/
Byberg L, Warensjö-Lemming E. Milk consumption for the prevention of fragility fractures. Nutrients. 2020;12(9):E2720. https://pubmed.ncbi.nlm.nih.gov/32899514/
Willett WC, Ludwig DS. Milk and health. N Engl J Med. 2020;382(7):644–54. https://pubmed.ncbi.nlm.nih.gov/32053300/
Phillip A. Study: milk may not be very good for bones or the body. The Washington Post. https://www.washingtonpost.com/news/to-your-health/wp/2014/10/31/study-milk-may-not-be-very-good-for-bones-or-the-body/. Published October 31, 2014. Accessed March 23, 2022.; https://www.washingtonpost.com/news/to-your-health/wp/2014/10/31/study-milk-may-not-be-very-good-for-bones-or-the-body/
Michaëlsson K, Wolk A, Langenskiöld S, et al. Milk intake and risk of mortality and fractures in women and men: cohort studies. BMJ. 2014;349:g6015. https://pubmed.ncbi.nlm.nih.gov/25352269/
Cui X, Wang L, Zuo P, et al. D-galactose-caused life shortening in Drosophila melanogaster and Musca domestica is associated with oxidative stress. Biogerontology. 2004;5(5):317–25. https://pubmed.ncbi.nlm.nih.gov/15547319/
Cui X, Zuo P, Zhang Q, et al. Chronic systemic D-galactose exposure induces memory loss, neurodegeneration, and oxidative damage in mice: protective effects of R-alpha-lipoic acid. J Neurosci Res. 2006;84(3):647–54. https://pubmed.ncbi.nlm.nih.gov/16555301/
Simoons FJ. A geographic approach to senile cataracts: possible links with milk consumption, lactase activity, and galactose metabolism. Dig Dis Sci. 1982;27(3):257–64. https://pubmed.ncbi.nlm.nih.gov/6804198/
Sella R, Afshari NA. Nutritional effect on age-related cataract formation and progression. Curr Opin Ophthalmol. 2019;30(1):63–9. https://pubmed.ncbi.nlm.nih.gov/30320615/
Ding M, Li J, Qi L, et al. Associations of dairy intake with risk of mortality in women and men: three prospective cohort studies. BMJ. 2019;367:l6204. https://pubmed.ncbi.nlm.nih.gov/31776125/
Grey A, Bolland M. Web of industry, advocacy, and academia in the management of osteoporosis. BMJ. 2015;351:h3170. https://pubmed.ncbi.nlm.nih.gov/26198274/
Byberg L, Warensjö-Lemming E. Milk consumption for the prevention of fragility fractures. Nutrients. 2020;12(9):E2720. https://pubmed.ncbi.nlm.nih.gov/32899514/
Willett WC, Ludwig DS. Milk and health. N Engl J Med. 2020;382(7):644–54. https://pubmed.ncbi.nlm.nih.gov/32053300/
Dai Z, Kroeger CM, Lawrence M, Scrinis G, Bero L. Comparison of methodological quality between the 2007 and 2019 Canadian dietary guidelines. Public Health Nutr. 2020;23(16):2879–85. https://pubmed.ncbi.nlm.nih.gov/32552917/
Ausman LM, Oliver LM, Goldin BR, Woods MN, Gorbach SL, Dwyer JT. Estimated net acid excretion inversely correlates with urine pH in vegans, lacto-ovo vegetarians, and omnivores. J Ren Nutr. 2008;18(5):456–65. https://pubmed.ncbi.nlm.nih.gov/18721741/
Kerstetter JE, O’Brien KO, Caseria DM, Wall DE, Insogna KL. The impact of dietary protein on calcium absorption and kinetic measures of bone turnover in women. J Clin Endocrinol Metab. 2005;90(1):26–31. https://pubmed.ncbi.nlm.nih.gov/15546911/
Dawson-Hughes B, Harris SS, Ceglia L. Alkaline diets favor lean tissue mass in older adults. Am J Clin Nutr. 2008;87(3):662–5. https://pubmed.ncbi.nlm.nih.gov/18326605/
Groesbeck DK, Bluml RM, Kossoff EH. Long-term use of the ketogenic diet in the treatment of epilepsy. Dev Med Child Neurol. 2006;48(12):978–81. https://pubmed.ncbi.nlm.nih.gov/17109786/
Heikura IA, Burke LM, Hawley JA, et al. A short-term ketogenic diet impairs markers of bone health in response to exercise. Front Endocrinol (Lausanne). 2020;10:880. https://pubmed.ncbi.nlm.nih.gov/32038477/
Simm PJ, Bicknell-Royle J, Lawrie J, et al. The effect of the ketogenic diet on the developing skeleton. Epilepsy Res. 2017;136:62–6. https://pubmed.ncbi.nlm.nih.gov/28778055/
Bergqvist AG, Schall JI, Stallings VA, Zemel BS. Progressive bone mineral content loss in children with intractable epilepsy treated with the ketogenic diet. Am J Clin Nutr. 2008;88(6):1678–84. https://pubmed.ncbi.nlm.nih.gov/19064531/
Yancy WS, Olsen MK, Dudley T, Westman EC. Acid-base analysis of individuals following two weight loss diets. Eur J Clin Nutr. 2007;61(12):1416–22. https://pubmed.ncbi.nlm.nih.gov/17299473/
Gunaratnam K, Vidal C, Gimble JM, Duque G. Mechanisms of palmitate-induced lipotoxicity in human osteoblasts. Endocrinology. 2014;155(1):108–16. https://pubmed.ncbi.nlm.nih.gov/24169557/
Mozaffari H, Djafarian K, Mofrad MD, Shab-Bidar S. Dietary fat, saturated fatty acid, and monounsaturated fatty acid intakes and risk of bone fracture: a systematic review and meta-analysis of observational studies. Osteoporos Int. 2018;29(9):1949–61. https://pubmed.ncbi.nlm.nih.gov/29947872/
Frassetto L, Sebastian A. Age and systemic acid-base equilibrium: analysis of published data. J Gerontol A Biol Sci Med Sci. 1996;51(1):B91–9. https://pubmed.ncbi.nlm.nih.gov/8548506/
Frassetto L, Banerjee T, Powe N, Sebastian A. Acid balance, dietary acid load, and bone effects – a controversial subject. Nutrients. 2018;10(4):517. https://pubmed.ncbi.nlm.nih.gov/29690515/
Cao JJ, Whigham LD, Jahns L. Depletion and repletion of fruit and vegetable intake alters serum bone turnover markers: a 28-week single-arm experimental feeding intervention. Br J Nutr. 2018;120(5):500–7. https://pubmed.ncbi.nlm.nih.gov/30022739/
Hayhoe RPG, Abdelhamid A, Luben RN, Khaw KT, Welch AA. Dietary acid-base load and its association with risk of osteoporotic fractures and low estimated skeletal muscle mass. Eur J Clin Nutr. 2020;74(Suppl 1):33–42. https://pubmed.ncbi.nlm.nih.gov/32873955/
Macdonald R, Black A, Sandison R, Aucott L, et al. Two year double blind randomized controlled trial in postmenopausal women shows no gain in BMD with potassium citrate treatment. Paper presented at: 28th Annual Meeting of the American Society of Bone and Mineral Research; September 15–19 https://pubmed.ncbi.nlm.nih.gov/18689384/
Dawson-Hughes B. Acid-base balance of the diet-implications for bone and muscle. Eur J Clin Nutr. 2020;74(Suppl 1):7–13. https://pubmed.ncbi.nlm.nih.gov/32873951/
Jehle S, Hulter HN, Krapf R. Effect of potassium citrate on bone density, microarchitecture, and fracture risk in healthy older adults without osteoporosis: a randomized controlled trial. J Clin Endocrinol Metab. 2013;98(1):207–17. https://pubmed.ncbi.nlm.nih.gov/23162100/
Fang Y, Zhu J, Fan J, et al. Dietary Inflammatory Index in relation to bone mineral density, osteoporosis risk and fracture risk: a systematic review and meta-analysis. Osteoporos Int. 2021;32(4):633–43. https://pubmed.ncbi.nlm.nih.gov/32740669/
Mun H, Liu B, Pham THA, Wu Q. C-reactive protein and fracture risk: an updated systematic review and meta-analysis of cohort studies through the use of both frequentist and Bayesian approaches. Osteoporos Int. 2021;32(3):425–35. https://pubmed.ncbi.nlm.nih.gov/32935169/
Zhao F, Guo L, Wang X, Zhang Y. Correlation of oxidative stress-related biomarkers with postmenopausal osteoporosis: a systematic review and meta-analysis. Arch Osteoporos. 2021;16(1):4. https://pubmed.ncbi.nlm.nih.gov/33400044/
Brondani JE, Comim FV, Flores LM, Martini LA, Premaor MO. Fruit and vegetable intake and bones: a systematic review and meta-analysis. PLoS One. 2019;14(5):e0217223. https://pubmed.ncbi.nlm.nih.gov/31150426/
Zeng LF, Luo MH, Liang GH, et al. Can dietary intake of vitamin C – oriented foods reduce the risk of osteoporosis, fracture, and BMD loss? Systematic review with meta-analyses of recent studies. Front Endocrinol (Lausanne). 2019;10:844. https://pubmed.ncbi.nlm.nih.gov/32117042/
Sun Y, Liu C, Bo Y, et al. Dietary vitamin C intake and the risk of hip fracture: a dose-response meta-analysis. Osteoporos Int. 2018;29(1):79–87. https://pubmed.ncbi.nlm.nih.gov/29101410/
Mühlbauer RC, Lozano A, Reinli A, Wetli H. Various selected vegetables, fruits, mushrooms and red wine residue inhibit bone resorption in rats. J Nutr. 2003;133(11):3592–7. https://pubmed.ncbi.nlm.nih.gov/14608079/
Hooshmand S, Kern M, Metti D, et al. The effect of two doses of dried plum on bone density and bone biomarkers in osteopenic postmenopausal women: a randomized, controlled trial. Osteoporos Int. 2016;27(7):2271–9. https://pubmed.ncbi.nlm.nih.gov/26902092/
Law YY, Chiu HF, Lee HH, Shen YC, Venkatakrishnan K, Wang CK. Consumption of onion juice modulates oxidative stress and attenuates the risk of bone disorders in middle-aged and post-menopausal healthy subjects. Food Funct. 2016;7(2):902–12. https://pubmed.ncbi.nlm.nih.gov/26686359/
Mackinnon ES, Rao AV, Josse RG, Rao LG. Supplementation with the antioxidant lycopene significantly decreases oxidative stress parameters and the bone resorption marker N-telopeptide of type I collagen in postmenopausal women. Osteoporos Int. 2011;22(4):1091–101. https://pubmed.ncbi.nlm.nih.gov/20552330/
Russo C, Ferro Y, Maurotti S, et al. Lycopene and bone: an in vitro investigation and a pilot prospective clinical study. J Transl Med. 2020;18(1):43. https://pubmed.ncbi.nlm.nih.gov/31996227/
Gunn CA, Weber JL, McGill AT, Kruger MC. Increased intake of selected vegetables, herbs and fruit may reduce bone turnover in post-menopausal women. Nutrients. 2015;7(4):2499–517. https://pubmed.ncbi.nlm.nih.gov/25856221/
Cheraghi Z, Doosti-Irani A, Almasi-Hashiani A, et al. The effect of alcohol on osteoporosis: a systematic review and meta-analysis. Drug Alcohol Depend. 2019;197:197–202. https://pubmed.ncbi.nlm.nih.gov/30844616/
Godos J, Giampieri F, Chisari E, et al. Alcohol consumption, bone mineral density, and risk of osteoporotic fractures: a dose-response meta-analysis. Int J Environ Res Public Health. 2022;19(3):1515. https://pubmed.ncbi.nlm.nih.gov/35162537/
Lems WF, Raterman HG. Critical issues and current challenges in osteoporosis and fracture prevention. An overview of unmet needs. Ther Adv Musculoskelet Dis. 2017;9(12):299–316. https://pubmed.ncbi.nlm.nih.gov/29201155/
Ahn H, Park YK. Sugar-sweetened beverage consumption and bone health: a systematic review and meta-analysis. Nutr J. 2021;20(1):41. https://pubmed.ncbi.nlm.nih.gov/33952276/
Fatahi S, Namazi N, Larijani B, Azadbakht L. The association of dietary and urinary sodium with bone mineral density and risk of osteoporosis: a systematic review and meta-analysis. J Am Coll Nutr. 2018;37(6):522–32. https://pubmed.ncbi.nlm.nih.gov/29617220/
Mortensen SJ, Beeram I, Florance J, et al. Modifiable lifestyle factors associated with fragility hip fracture: a systematic review and meta-analysis. J Bone Miner Metab. 2021;39(5):893–902. https://pubmed.ncbi.nlm.nih.gov/33991260/
Shen CL, Chyu MC, Yeh JK, et al. Effect of green tea and Tai Chi on bone health in postmenopausal osteopenic women: a 6-month randomized placebo-controlled trial. Osteoporos Int. 2012;23(5):1541–52. https://pubmed.ncbi.nlm.nih.gov/21766228/
Shen CL, Wang P, Guerrieri J, Yeh JK, Wang JS. Protective effect of green tea polyphenols on bone loss in middle-aged female rats. Osteoporos Int. 2008;19(7):979–90. https://pubmed.ncbi.nlm.nih.gov/18084689/
Dostal AM, Arikawa A, Espejo L, Kurzer MS. Long-term supplementation of green tea extract does not modify adiposity or bone mineral density in a randomized trial of overweight and obese postmenopausal women. J Nutr. 2016;146(2):256–64. https://pubmed.ncbi.nlm.nih.gov/26701796/
Platt ID, Josse AR, Kendall CWC, Jenkins DJA, El-Sohemy A. Postprandial effects of almond consumption on human osteoclast precursors – an ex vivo study. Metabolism. 2011;60(7):923–9. https://pubmed.ncbi.nlm.nih.gov/20947104/
Fugh-Berman A, Pearson C. The overselling of hormone replacement therapy. Pharmacotherapy. 2002;22(9):1205–8. https://pubmed.ncbi.nlm.nih.gov/12222561/
Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321–33. https://pubmed.ncbi.nlm.nih.gov/12117397/
Oseni T, Patel R, Pyle J, Jordan VC. Selective estrogen receptor modulators and phytoestrogens. Planta Med. 2008;74(13):1656–65. https://pubmed.ncbi.nlm.nih.gov/18843590/
McCarty MF. Isoflavones made simple – genistein’s agonist activity for the beta-type estrogen receptor mediates their health benefits. Med Hypotheses. 2006;66(6):1093–114. https://pubmed.ncbi.nlm.nih.gov/16513288/
Chi F, Wu R, Zeng YC, Xing R, Liu Y, Xu ZG. Post-diagnosis soy food intake and breast cancer survival: a meta-analysis of cohort studies. Asian Pac J Cancer Prev. 2013;14(4):2407–12. https://pubmed.ncbi.nlm.nih.gov/23725149/
Sansai K, Na Takuathung M, Khatsri R, Teekachunhatean S, Hanprasertpong N, Koonrungsesomboon N. Effects of isoflavone interventions on bone mineral density in postmenopausal women: a systematic review and meta-analysis of randomized controlled trials. Osteoporos Int. 2020;31(10):1853–64. https://pubmed.ncbi.nlm.nih.gov/32524173/
Morabito N, Crisafulli A, Vergara C, et al. Effects of genistein and hormone-replacement therapy on bone loss in early postmenopausal women: a randomized double-blind placebo-controlled study. J Bone Miner Res. 2002;17(10):1904–12. https://pubmed.ncbi.nlm.nih.gov/12369794/
Lydeking-Olsen E, Beck-Jensen JE, Setchell KDR, Holm-Jensen T. Soymilk or progesterone for prevention of bone loss: a 2 year randomized, placebo-controlled trial. Eur J Nutr. 2004;43(4):246–57. https://pubmed.ncbi.nlm.nih.gov/15309425/
Koch L. Nutrition: High isoflavone intake delays puberty onset and may reduce breast cancer risk in girls. Nat Rev Endocrinol. 2010;6(11):595. https://pubmed.ncbi.nlm.nih.gov/21038502/
Jacobsen BK, Knutsen SF, Fraser GE. Does high soy milk intake reduce prostate cancer incidence? The Adventist Health Study (United States). Cancer Causes Control. 1998;9(6):553–7. https://pubmed.ncbi.nlm.nih.gov/10189040/
Fujisawa T, Ohashi Y, Shin R, Narai-Kanayama A, Nakagaki T. The effect of soymilk intake on the fecal microbiota, particularly Bifidobacterium species, and intestinal environment of healthy adults: a pilot study. Biosci Microbiota Food Health. 2017;36(1):33–7. https://pubmed.ncbi.nlm.nih.gov/28243549/
Eslami O, Shidfar F, Maleki Z, et al. Effect of soy milk on metabolic status of patients with nonalcoholic fatty liver disease: a randomized clinical trial. J Am Coll Nutr. 2019;38(1):51–8. https://pubmed.ncbi.nlm.nih.gov/30028245/
Mitchell JH, Collins AR. Effects of a soy milk supplement on plasma cholesterol levels and oxidative DNA damage in men – a pilot study. Eur J Nutr. 1999;38(3):143–8. https://pubmed.ncbi.nlm.nih.gov/10443336/
Maleki Z, Jazayeri S, Eslami O, et al. Effect of soy milk consumption on glycemic status, blood pressure, fibrinogen and malondialdehyde in patients with non-alcoholic fatty liver disease: a randomized controlled trial. Complement Ther Med. 2019;44:44–50. https://pubmed.ncbi.nlm.nih.gov/31126574/
Liao YH, Chen CN, Hu CY, Tsai SC, Kuo YC. Soymilk ingestion immediately after therapeutic exercise enhances rehabilitation outcomes in chronic stroke patients: a randomized controlled trial. NeuroRehabilitation. 2019;44(2):217–29. https://pubmed.ncbi.nlm.nih.gov/30856124/
Rivas M, Garay RP, Escanero JF, Cia P, Cia P, Alda JO. Soy milk lowers blood pressure in men and women with mild to moderate essential hypertension. J Nutr. 2002;132(7):1900–2. https://pubmed.ncbi.nlm.nih.gov/12097666/
Onuegbu AJ, Olisekodiaka JM, Onibon MO, Adesiyan AA, Igbeneghu CA. Consumption of soymilk lowers atherogenic lipid fraction in healthy individuals. J Med Food. 2011;14(3):257–60. https://pubmed.ncbi.nlm.nih.gov/21142946/
Vanga SK, Raghavan V. How well do plant based alternatives fare nutritionally compared to cow’s milk? J Food Sci Technol. 2018;55(1):10–20. https://pubmed.ncbi.nlm.nih.gov/29358791/
Shi Y, Zhan Y, Chen Y, Jiang Y. Effects of dairy products on bone mineral density in healthy postmenopausal women: a systematic review and meta-analysis of randomized controlled trials. Arch Osteoporos. 2020;15(1):48. https://pubmed.ncbi.nlm.nih.gov/32185512/
Byberg L, Warensjö-Lemming E. Milk consumption for the prevention of fragility fractures. Nutrients. 2020;12(9):E2720. https://pubmed.ncbi.nlm.nih.gov/32899514/
Akhavan Zanjani M, Rahmani S, Mehranfar S, et al. Soy foods and the risk of fracture: a systematic review of prospective cohort studies. Complement Med Res. 2022;29(2):172–81. https://pubmed.ncbi.nlm.nih.gov/34547749/
Zhang X, Shu XO, Li H, et al. Prospective cohort study of soy food consumption and risk of bone fracture among postmenopausal women. Arch Intern Med. 2005;165(16):1890–5. https://pubmed.ncbi.nlm.nih.gov/16157834/
Chen Z, Zheng W, Custer LJ, et al. Usual dietary consumption of soy foods and its correlation with the excretion rate of isoflavonoids in overnight urine samples among Chinese women in Shanghai. Nutr Cancer. 1999;33(1):82–7. https://pubmed.ncbi.nlm.nih.gov/10227048/
Prabhakaran MP. Isoflavone levels and the effect of processing on the content of isoflavones during the preparation of soymilk and tofu. Thesis submitted for the degree of doctor of philosophy to the National University of Singapore. 2005.; https://scholarbank.nus.edu.sg/handle/10635/15175
Petroski W, Minich DM. Is there such a thing as “anti-nutrients”? A narrative review of perceived problematic plant compounds. Nutrients. 2020;12(10):2929. https://pubmed.ncbi.nlm.nih.gov/32987890/
Berg J, Seyedsadjadi N, Grant R. Increased consumption of plant foods is associated with increased bone mineral density. J Nutr Health Aging. 2020;24(4):388–97. https://pubmed.ncbi.nlm.nih.gov/32242206/
Melaku YA, Gill TK, Appleton SL, Taylor AW, Adams R, Shi Z. Prospective associations of dietary and nutrient patterns with fracture risk: a 20-year follow-up study. Nutrients. 2017;9(11):1198. https://pubmed.ncbi.nlm.nih.gov/29088104/
Iguacel I, Miguel-Berges ML, Gómez-Bruton A, Moreno LA, Julián C. Veganism, vegetarianism, bone mineral density, and fracture risk: a systematic review and meta-analysis. Nutr Rev. 2019;77(1):1–18. https://pubmed.ncbi.nlm.nih.gov/30376075/
Karavasiloglou N, Selinger E, Gojda J, Rohrmann S, Kühn T. Differences in bone mineral density between adult vegetarians and nonvegetarians become marginal when accounting for differences in anthropometric factors. J Nutr. 2020;150(5):1266–71. https://pubmed.ncbi.nlm.nih.gov/32055831/
Iwaniec UT, Turner RT. Influence of body weight on bone mass, architecture and turnover. J Endocrinol. 2016;230(3):R115–30. https://pubmed.ncbi.nlm.nih.gov/27352896/
Tong TYN, Appleby PN, Armstrong MEG, et al. Vegetarian and vegan diets and risks of total and site-specific fractures: results from the prospective EPIC-Oxford study. BMC Med. 2020;18(1):353. https://bmcmedicine.biomedcentral.com/articles/10.1186/s12916-020-01815-3
Tong TYN, Appleby PN, Armstrong MEG, et al. Vegetarian and vegan diets and risks of total and site-specific fractures: results from the prospective EPIC-Oxford study. Table S6. Risks of hip fractures by age, sex, menopausal status, physical activity and BMI. BMC Med. 2020;18(1):353. https://bmcmedicine.biomedcentral.com/articles/10.1186/s12916-020-01815-3
Yao P, Bennett D, Mafham M, et al. Vitamin D and calcium for the prevention of fracture: a systematic review and meta-analysis. JAMA Netw Open. 2019;2(12):e1917789. https://pubmed.ncbi.nlm.nih.gov/31860103/
Heaney RP. The vitamin D requirement in health and disease. J Steroid Biochem Mol Biol. 2005;97(1–2):13–9. https://pubmed.ncbi.nlm.nih.gov/16026981/
Appleby P, Roddam A, Allen N, Key T. Comparative fracture risk in vegetarians and nonvegetarians in EPIC-Oxford. Eur J Clin Nutr. 2007;61(12):1400–6. https://pubmed.ncbi.nlm.nih.gov/17299475/
Lang T, LeBlanc A, Evans H, Lu Y, Genant H, Yu A. Cortical and trabecular bone mineral loss from the spine and hip in long-duration spaceflight. J Bone Miner Res. 2004;19(6):1006–12. https://pubmed.ncbi.nlm.nih.gov/15125798/
Troy KL, Mancuso ME, Butler TA, Johnson JE. Exercise early and often: effects of physical activity and exercise on women’s bone health. Int J Environ Res Public Health. 2018;15(5):E878. https://pubmed.ncbi.nlm.nih.gov/29710770/
Troy KL, Mancuso ME, Butler TA, Johnson JE. Exercise early and often: effects of physical activity and exercise on women’s bone health. Int J Environ Res Public Health. 2018;15(5):E878. https://pubmed.ncbi.nlm.nih.gov/29710770/
Lu YH, Rosner B, Chang G, Fishman LM. Twelve-minute daily yoga regimen reverses osteoporotic bone loss. Top Geriatr Rehabil. 2016;32(2):81–7. https://pubmed.ncbi.nlm.nih.gov/27226695/
Sfeir JG, Drake MT, Sonawane VJ, Sinaki M. Vertebral compression fractures associated with yoga: a case series. Eur J Phys Rehabil Med. 2018;54(6):947–51. https://pubmed.ncbi.nlm.nih.gov/29687967/
Cramer H, Ostermann T, Dobos G. Injuries and other adverse events associated with yoga practice: a systematic review of epidemiological studies. J Sci Med Sport. 2018;21(2):147–54. https://pubmed.ncbi.nlm.nih.gov/28958637/
Cramer H, Quinker D, Schumann D, Wardle J, Dobos G, Lauche R. Adverse effects of yoga: a national cross-sectional survey. BMC Complement Altern Med. 2019;19(1):190. https://pubmed.ncbi.nlm.nih.gov/31357980/
Zhu JK, Wu LD, Zheng RZ, Lan SH. Yoga is found hazardous to the meniscus for Chinese women. Chin J Traumatol. 2012;15(3):148–51. https://pubmed.ncbi.nlm.nih.gov/22663908/
Boddu P, Patel S, Shahrrava A. Sudden cardiac arrest from heat stroke: hidden dangers of hot yoga. Am J Med. 2016;129(8):e129–30. https://pubmed.ncbi.nlm.nih.gov/27107927/
insightSlice. Bone densitometer market global sales are expected to grow steadily to reach US$1.75 billion by 2031. Globe Newswire. https://www.globenewswire.com/news-release/2021/07/12/2261344/0/en/Bone-Densitometer-Market-Global-Sales-are-Expected-to-Grow-Steadily-to-Reach-US-1–75-billion-by-2031.html. Published July 12, 2021. Accessed March 18, 2022.; https://www.globenewswire.com/news-release/2021/07/12/2261344/0/en/Bone-Densitometer-Market-Global-Sales-are-Expected-to-Grow-Steadily-to-Reach-US-1-75-billion-by-2031.html
Stone KL, Seeley DG, Lui LY, et al. BMD at multiple sites and risk of fracture of multiple types: long-term results from the Study of Osteoporotic Fractures. J Bone Miner Res. 2003;18(11):1947–54. https://pubmed.ncbi.nlm.nih.gov/14606506/
De Laet CEDH, van Hout BA, Burger H, Hofman A, Pols HAP. Bone density and risk of hip fracture in men and women: cross sectional analysis. BMJ. 1997;315(7102):221–5. https://pubmed.ncbi.nlm.nih.gov/9253270/
Järvinen TLN, Sievänen H, Khan KM, Heinonen A, Kannus P. Shifting the focus in fracture prevention from osteoporosis to falls. BMJ. 2008;336(7636):124–6. https://pubmed.ncbi.nlm.nih.gov/18202065/
Nordström P, Eklund F, Björnstig U, et al. Do both areal BMD and injurious falls explain the higher incidence of fractures in women than in men? Calcif Tissue Int. 2011;89(3):203–10. https://pubmed.ncbi.nlm.nih.gov/21667164/
Wagner H, Melhus H, Gedeborg R, Pedersen NL, Michaëlsson K. Simply ask them about their balance – future fracture risk in a nationwide cohort study of twins. Am J Epidemiol. 2009;169(2):143–9. https://pubmed.ncbi.nlm.nih.gov/19064648/
Stone KL, Seeley DG, Lui LY, et al. BMD at multiple sites and risk of fracture of multiple types: long-term results from the Study of Osteoporotic Fractures. J Bone Miner Res. 2003;18(11):1947–54. https://pubmed.ncbi.nlm.nih.gov/14606506/
Järvinen TLN, Michaëlsson K, Aspenberg P, Sievänen H. Osteoporosis: the emperor has no clothes. J Intern Med. 2015;277(6):662–73. https://pubmed.ncbi.nlm.nih.gov/25809279/
Dequeker J, Nijs J, Verstraeten A, Geusens P, Gevers G. Genetic determinants of bone mineral content at the spine and radius: a twin study. Bone. 1987;8(4):207–9. https://pubmed.ncbi.nlm.nih.gov/3446256/
Michaëlsson K, Melhus H, Ferm H, Ahlbom A, Pedersen NL. Genetic liability to fractures in the elderly. Arch Intern Med. 2005;165(16):1825–30. https://pubmed.ncbi.nlm.nih.gov/16157825/
Wagner H, Melhus H, Pedersen NL, Michaëlsson K. Heritability of impaired balance: a nationwide cohort study in twins. Osteoporos Int. 2009;20(4):577–83. https://pubmed.ncbi.nlm.nih.gov/18802660/
Burger H, de Laet CEDH, Weel AEAM, Hofman A, Pols HAP. Added value of bone mineral density in hip fracture risk scores. Bone. 1999;25(3):369–74. https://pubmed.ncbi.nlm.nih.gov/10495142/
Järvinen TLN, Michaëlsson K, Aspenberg P, Sievänen H. Osteoporosis: the emperor has no clothes. J Intern Med. 2015;277(6):662–73. https://pubmed.ncbi.nlm.nih.gov/25809279/
Kannus P, Sievänen H, Palvanen M, Järvinen T, Parkkari J. Prevention of falls and consequent injuries in elderly people. Lancet. 2005;366(9500):1885–93. https://pubmed.ncbi.nlm.nih.gov/16310556/
Järvinen TLN, Michaëlsson K, Aspenberg P, Sievänen H. Osteoporosis: the emperor has no clothes. J Intern Med. 2015;277(6):662–73. https://pubmed.ncbi.nlm.nih.gov/25809279/
Tinetti ME. Preventing falls in elderly persons. N Engl J Med. 2003;348(1):42–9. https://pubmed.ncbi.nlm.nih.gov/12510042/
Sernbo I, Johnell O. Consequences of a hip fracture: a prospective study over 1 year. Osteoporos Int. 1993;3(3):148–53. https://pubmed.ncbi.nlm.nih.gov/8481591/
Tricco AC, Thomas SM, Veroniki AA, et al. Comparisons of interventions for preventing falls in older adults: a systematic review and meta-analysis. JAMA. 2017;318(17):1687–99. https://pubmed.ncbi.nlm.nih.gov/29114830/
Dautzenberg L, Beglinger S, Tsokani S, et al. Interventions for preventing falls and fall-related fractures in community-dwelling older adults: a systematic review and network meta-analysis. J Am Geriatr Soc. 2021;69(10):2973–84. https://pubmed.ncbi.nlm.nih.gov/34318929/
Sherrington C, Fairhall NJ, Wallbank GK, et al. Exercise for preventing falls in older people living in the community. Cochrane Database Syst Rev. 2019;1:CD012424. https://pubmed.ncbi.nlm.nih.gov/31792067/
Wong RMY, Chong KC, Law SW, et al. The effectiveness of exercises on fall and fracture prevention amongst community elderlies: a systematic review and meta-analysis. J Orthop Translat. 2020;24:58–65. https://pubmed.ncbi.nlm.nih.gov/32695605/
Karinkanta S, Heinonen A, Sievänen H, et al. A multi-component exercise regimen to prevent functional decline and bone fragility in home-dwelling elderly women: randomized, controlled trial. Osteoporos Int. 2007;18(4):453–62. https://pubmed.ncbi.nlm.nih.gov/17103296/
Karinkanta S, Kannus P, Uusi-Rasi K, Heinonen A, Sievänen H. Combined resistance and balance-jumping exercise reduces older women’s injurious falls and fractures: 5-year follow-up study. Age Ageing. 2015;44(5):784–9. https://pubmed.ncbi.nlm.nih.gov/25990940/
Korall AMB, Feldman F, Scott VJ, et al. Facilitators of and barriers to hip protector acceptance and adherence in long-term care facilities: a systematic review. J Am Med Dir Assoc. 2015;16(3):185–93. https://pubmed.ncbi.nlm.nih.gov/25704127/
Santesso N, Carrasco-Labra A, Brignardello-Petersen R. Hip protectors for preventing hip fractures in older people. Cochrane Database Syst Rev. 2014;(3):CD001255. https://pubmed.ncbi.nlm.nih.gov/24687239/
Dautzenberg L, Beglinger S, Tsokani S, et al. Interventions for preventing falls and fall-related fractures in community-dwelling older adults: a systematic review and network meta-analysis. J Am Geriatr Soc. 2021;69(10):2973–84. https://pubmed.ncbi.nlm.nih.gov/34318929/
STEADI. What you can do to prevent falls. Centers for Disease Control and Prevention. https://www.cdc.gov/steadi/pdf/STEADI-Brochure-WhatYouCanDo-508.pdf. Published 2017. Accessed March 11, 2022.; https://www.cdc.gov/steadi/patient.html
STEADI. Family caregivers: protect your loved ones from falling. Centers for Disease Control and Prevention. https://www.cdc.gov/steadi/pdf/patient/customizable/Caregiver-Brochure-Final-Customizable-508.pdf. Published 2018. Accessed March 11, 2022.; https://www.cdc.gov/steadi/patient.html
Pirruccio K, Ahn J. Fractures while walking leashed dogs – reply. JAMA Surg. 2019;154(11):1078. https://pubmed.ncbi.nlm.nih.gov/31389985/
Law MR, Wald NJ, Meade TW. Strategies for prevention of osteoporosis and hip fracture. BMJ. 1991;303(6800):453–9. https://pubmed.ncbi.nlm.nih.gov/1912840/
Järvinen TLN, Michaëlsson K, Aspenberg P, Sievänen H. Osteoporosis: the emperor has no clothes. J Intern Med. 2015;277(6):662–73. https://pubmed.ncbi.nlm.nih.gov/25809279/
Sullivan R. A brief journey into medical care and disease in ancient Egypt. J R Soc Med. 1995;88(3):141–5. https://pubmed.ncbi.nlm.nih.gov/7752157/
Chen TS, Chen PS. Gastroenterology in ancient Egypt. J Clin Gastroenterol. 1991;13(2):182–7. https://pubmed.ncbi.nlm.nih.gov/2033225/
Holl RM. Bowel movement: the sixth vital sign. Holist Nurs Pract. 2014;28(3):195–7. https://pubmed.ncbi.nlm.nih.gov/24722614/
Staller K, Cash BD. Myths and misconceptions about constipation: a new view for the 2020s. Am J Gastroenterol. 2020;115(11):1741–5. https://pubmed.ncbi.nlm.nih.gov/33156087/
Johanson JF, Kralstein J. Chronic constipation: a survey of the patient perspective. Aliment Pharmacol Ther. 2007;25(5):599–608. https://pubmed.ncbi.nlm.nih.gov/17305761/
Gokce AH, Gokce FS. Effects of bilateral transcutaneous tibial nerve stimulation on constipation severity in geriatric patients: a prospective clinical study. Geriatr Gerontol Int. 2020;20(2):101–5. https://pubmed.ncbi.nlm.nih.gov/31793185/
Sonnenberg A, Koch TR. Physician visits in the United States for constipation: 1958 to 1986. Dig Dis Sci. 1989;34(4):606–11. https://pubmed.ncbi.nlm.nih.gov/2784759/
Luthra P, Camilleri M, Burr NE, Quigley EMM, Black CJ, Ford AC. Efficacy of drugs in chronic idiopathic constipation: a systematic review and network meta-analysis. Lancet Gastroenterol Hepatol. 2019;4(11):831–44. https://pubmed.ncbi.nlm.nih.gov/31474542/
Chen J, Liu X, Bai T, Hou X. Impact of clinical outcome measures on placebo response rates in clinical trials for chronic constipation: a systematic review and meta-analysis. Clin Transl Gastroenterol. 2020;11(11):e00255. https://pubmed.ncbi.nlm.nih.gov/33259160/
Pont LG, Fisher M, Williams K. Appropriate use of laxatives in the older person. Drugs Aging. 2019;36(11):999–1005. https://pubmed.ncbi.nlm.nih.gov/31478168/
Lämås K, Karlsson S, Nolén A, Lövheim H, Sandman PO. Prevalence of constipation among persons living in institutional geriatric-care settings – a cross-sectional study. Scand J Caring Sci. 2017;31(1):157–63. https://pubmed.ncbi.nlm.nih.gov/27327073/
Tvistholm N, Munch L, Danielsen AK. Constipation is casting a shadow over everyday life – a systematic review on older people’s experience of living with constipation. J Clin Nurs. 2017;26(7–8):902–14. https://pubmed.ncbi.nlm.nih.gov/27271918/
Belsey J, Greenfield S, Candy D, Geraint M. Systematic review: impact of constipation on quality of life in adults and children. Aliment Pharmacol Ther. 2010;31(9):938–49. https://pubmed.ncbi.nlm.nih.gov/20180788/
Tvistholm N, Munch L, Danielsen AK. Constipation is casting a shadow over everyday life – a systematic review on older people’s experience of living with constipation. J Clin Nurs. 2017;26(7–8):902–14. https://pubmed.ncbi.nlm.nih.gov/27271918/
Emmanuel A, Mattace-Raso F, Neri MC, Petersen KU, Rey E, Rogers J. Constipation in older people: a consensus statement. Int J Clin Pract. 2017;71(1):e12920. https://discovery.ucl.ac.uk/id/eprint/1533175/
Pekmezaris R, Aversa L, Wolf-Klein G, Cedarbaum J, Reid-Durant M. The cost of chronic constipation. J Am Med Dir Assoc. 2002;3(4):224–8. https://pubmed.ncbi.nlm.nih.gov/12807642/
Modi RM, Hinton A, Pinkhas D, et al. Implementation of a defecation posture modification device. J Clin Gastroenterol. 2019;53(3):216–9. https://pubmed.ncbi.nlm.nih.gov/30346317/
Burkitt DP. A deficiency of dietary fiber may be one cause of certain colonic and venous disorders. Am J Dig Dis. 1976;21(2):104–8. https://pubmed.ncbi.nlm.nih.gov/1274909/
Burkitt DP. Hiatus hernia: is it preventable? Am J Clin Nutr. 1981;34(3):428–31. https://pubmed.ncbi.nlm.nih.gov/6259926/
Burkitt DP, James PA. Low-residue diets and hiatus hernia. Lancet. 1973;2(7821):128–30. https://pubmed.ncbi.nlm.nih.gov/4124047/
Burkitt DP. A deficiency of dietary fiber may be one cause of certain colonic and venous disorders. Am J Dig Dis. 1976;21(2):104–8. https://pubmed.ncbi.nlm.nih.gov/1274909/
Fox A, Tietze PH, Ramakrishnan K. Anorectal conditions: anal fissure and anorectal fistula. FP Essent. 2014;419:20–7. https://pubmed.ncbi.nlm.nih.gov/24742084/
Burkitt DP. Two blind spots in medical knowledge. Nurs Times. 1976;72(1):24–7. https://pubmed.ncbi.nlm.nih.gov/54904/
Burkitt DP. Hiatus hernia: is it preventable? Am J Clin Nutr. 1981;34(3):428–31. https://pubmed.ncbi.nlm.nih.gov/6259926/
Burkitt DP. Dietary fibre and “pressure diseases.” J R Coll Physicians Lond. 1975;9(2):138–46. https://pubmed.ncbi.nlm.nih.gov/1127617/
Kapoor WN, Peterson J, Karpf M. Defecation syncope: a symptom with multiple etiologies. Arch Intern Med. 1986;146(12):2377–9. https://pubmed.ncbi.nlm.nih.gov/3778072/
Greenfield JC, Rembert JC, Tindall GT. Transient changes in cerebral vascular resistance during the Valsalva maneuver in man. Stroke. 1984;15(1):76–9. https://pubmed.ncbi.nlm.nih.gov/6229907/
Benchimol A, Wang TF, Desser KB, Gartlan JL. The Valsalva maneuver and coronary arterial blood flow velocity. Studies in man. Ann Intern Med. 1972;77(3):357–60. https://pubmed.ncbi.nlm.nih.gov/5053728/
McGuire J, Green RS, Courter S, et al. Bed pan deaths. J Lab Clin Med. 1948;33(11):1457. https://pubmed.ncbi.nlm.nih.gov/18890042/
Emmanuel A, Mattace-Raso F, Neri MC, Petersen KU, Rey E, Rogers J. Constipation in older people: a consensus statement. Int J Clin Pract. 2017;71(1):e12920. https://discovery.ucl.ac.uk/id/eprint/1533175/
Annells M, Koch T. Faecal impaction: older people’s experiences and nursing practice. Br J Community Nurs. 2002;7(3):118–26. https://pubmed.ncbi.nlm.nih.gov/11904547/
Annells M, Koch T. Older people seeking solutions to constipation: the laxative mire. J Clin Nurs. 2002;11(5):603–12. https://pubmed.ncbi.nlm.nih.gov/12201887/
Sakakibara R, Tsunoyama K, Hosoi H, et al. Influence of body position on defecation in humans. Low Urin Tract Symptoms. 2010;2(1):16–21. https://pubmed.ncbi.nlm.nih.gov/26676214/
Isbit J. Is the porcelain throne to blame? Tech Coloproctol. 2015;19(3):193–4. https://pubmed.ncbi.nlm.nih.gov/25579878/
Davies GJ, Crowder M, Reid B, Dickerson JW. Bowel function measurements of individuals with different eating patterns. Gut. 1986;27(2):164–9. https://pubmed.ncbi.nlm.nih.gov/3005140/
Choi YI, Kim KO, Chung JW, et al. Effects of automatic abdominal massage device in treatment of chronic constipation patients: a prospective study. Dig Dis Sci. 2021;66(9):3105–12. https://pubmed.ncbi.nlm.nih.gov/33001346/
Rao SSC, Lembo A, Chey WD, Friedenberg K, Quigley EMM. Effects of the vibrating capsule on colonic circadian rhythm and bowel symptoms in chronic idiopathic constipation. Neurogastroenterol Motil. 2020;32(11):e13890. https://pubmed.ncbi.nlm.nih.gov/32449277/
Staller K, Cash BD. Myths and misconceptions about constipation: a new view for the 2020s. Am J Gastroenterol. 2020;115(11):1741–5. https://pubmed.ncbi.nlm.nih.gov/33156087/
Knowles CH, Grossi U, Chapman M, et al. Surgery for constipation: systematic review and practice recommendations: Results I: colonic resection. Colorectal Dis. 2017;19 Suppl 3:17–36. https://pubmed.ncbi.nlm.nih.gov/28960923/
Rao SSC, Brenner DM. Efficacy and safety of over-the-counter therapies for chronic constipation: an updated systematic review. Am J Gastroenterol. 2021;116(6):1156–81. https://pubmed.ncbi.nlm.nih.gov/33767108/
Rao SSC, Brenner DM. Efficacy and safety of over-the-counter therapies for chronic constipation: an updated systematic review. Am J Gastroenterol. 2021;116(6):1156–81. https://pubmed.ncbi.nlm.nih.gov/33767108/
Pont LG, Fisher M, Williams K. Appropriate use of laxatives in the older person. Drugs Aging. 2019;36(11):999–1005. https://pubmed.ncbi.nlm.nih.gov/31478168/
Rao SSC, Brenner DM. Efficacy and safety of over-the-counter therapies for chronic constipation: an updated systematic review. Am J Gastroenterol. 2021;116(6):1156–81. https://pubmed.ncbi.nlm.nih.gov/33767108/
Noergaard M, Traerup Andersen J, Jimenez-Solem E, Bring Christensen M. Long term treatment with stimulant laxatives – clinical evidence for effectiveness and safety? Scand J Gastroenterol. 2019;54(1):27–34. https://pubmed.ncbi.nlm.nih.gov/30700194/
Riemann JF, Schmidt H, Zimmermann W. The fine structure of colonic submucosal nerves in patients with chronic laxative abuse. Scand J Gastroenterol. 1980;15(6):761–8. https://pubmed.ncbi.nlm.nih.gov/7209384/
Serrano-Falcón B, Rey E. The safety of available treatments for chronic constipation. Expert Opin Drug Saf. 2017;16(11):1243–53. https://pubmed.ncbi.nlm.nih.gov/28756692/
Rao SSC, Brenner DM. Efficacy and safety of over-the-counter therapies for chronic constipation: an updated systematic review. Am J Gastroenterol. 2021;116(6):1156–81. https://pubmed.ncbi.nlm.nih.gov/33767108/
Leth PM, Gregersen M. Ethylene glycol poisoning. Forensic Sci Int. 2005;155(2–3):179–84. https://pubmed.ncbi.nlm.nih.gov/16226155/
Serrano-Falcón B, Rey E. The safety of available treatments for chronic constipation. Expert Opin Drug Saf. 2017;16(11):1243–53. https://pubmed.ncbi.nlm.nih.gov/28756692/
Lacy BE, Shea EP, Manuel M, Abel JL, Jiang H, Taylor DCA. Lessons learned: chronic idiopathic constipation patient experiences with over-the-counter medications. PLoS One. 2021;16(1):e0243318. https://pubmed.ncbi.nlm.nih.gov/33428631/
Lucak S, Lunsford TN, Harris LA. Evaluation and treatment of constipation in the geriatric population. Clin Geriatr Med. 2021;37(1):85–102. https://pubmed.ncbi.nlm.nih.gov/33213776/
Annells M, Koch T. Older people seeking solutions to constipation: the laxative mire. J Clin Nurs. 2002;11(5):603–12. https://pubmed.ncbi.nlm.nih.gov/12201887/
Wilson PB. Associations between physical activity and constipation in adult Americans: results from the National Health and Nutrition Examination Survey. Neurogastroenterol Motil. 2020;32(5):e13789. https://pubmed.ncbi.nlm.nih.gov/31905422/
Liu F, Kondo T, Toda Y. Brief physical inactivity prolongs colonic transit time in elderly active men. Int J Sports Med. 1993;14(8):465–7. https://pubmed.ncbi.nlm.nih.gov/8300274/
Asnicar F, Leeming ER, Dimidi E, et al. Blue poo: impact of gut transit time on the gut microbiome using a novel marker. Gut. 2021;70(9):1665–74. https://pubmed.ncbi.nlm.nih.gov/33722860/
Gao R, Tao Y, Zhou C, et al. Exercise therapy in patients with constipation: a systematic review and meta-analysis of randomized controlled trials. Scand J Gastroenterol. 2019;54(2):169–77. https://pubmed.ncbi.nlm.nih.gov/30843436/
Mari A, Mahamid M, Amara H, Baker FA, Yaccob A. Chronic constipation in the elderly patient: updates in evaluation and management. Korean J Fam Med. 2020;41(3):139–45. https://pubmed.ncbi.nlm.nih.gov/32062960/
Pont LG, Fisher M, Williams K. Appropriate use of laxatives in the older person. Drugs Aging. 2019;36(11):999–1005. https://pubmed.ncbi.nlm.nih.gov/31478168/
Burkitt DP. A deficiency of dietary fiber may be one cause of certain colonic and venous disorders. Am J Dig Dis. 1976;21(2):104–8. https://pubmed.ncbi.nlm.nih.gov/1274909/
Clemens R, Kranz S, Mobley AR, et al. Filling America’s fiber intake gap: summary of a roundtable to probe realistic solutions with a focus on grain-based foods. J Nutr. 2012;142(7):1390S-401S. https://pubmed.ncbi.nlm.nih.gov/22649260/
Sanjoaquin MA, Appleby PN, Spencer EA, Key TJ. Nutrition and lifestyle in relation to bowel movement frequency: a cross-sectional study of 20630 men and women in EPIC-Oxford. Public Health Nutr. 2004 Feb;7(1):77–83. https://pubmed.ncbi.nlm.nih.gov/14972075/
Schmier JK, Miller PE, Levine JA, et al. Cost savings of reduced constipation rates attributed to increased dietary fiber intakes: a decision-analytic model. BMC Public Health. 2014;14:374. https://pubmed.ncbi.nlm.nih.gov/24739472/
Oh SJ, Fuller G, Patel D, et al. Chronic constipation in the United States: results from a population-based survey assessing healthcare seeking and use of pharmacotherapy. Am J Gastroenterol. 2020;115(6):895–905. https://pubmed.ncbi.nlm.nih.gov/32324606/
Christodoulides S, Dimidi E, Fragkos KC, Farmer AD, Whelan K, Scott SM. Systematic review with meta-analysis: effect of fibre supplementation on chronic idiopathic constipation in adults. Aliment Pharmacol Ther. 2016;44(2):103–16. https://pubmed.ncbi.nlm.nih.gov/27170558/
Staller K, Cash BD. Myths and misconceptions about constipation: a new view for the 2020s. Am J Gastroenterol. 2020;115(11):1741–5. https://pubmed.ncbi.nlm.nih.gov/33156087/
Jalanka J, Major G, Murray K, et al. The effect of psyllium husk on intestinal microbiota in constipated patients and healthy controls. Int J Mol Sci. 2019;20(2):E433. https://pubmed.ncbi.nlm.nih.gov/30669509/
Hefny AF, Ayad AZ, Matev N, Bashir MO. Intestinal obstruction caused by a laxative drug (Psyllium): a case report and review of the literature. Int J Surg Case Rep. 2018;52:59–62. https://pubmed.ncbi.nlm.nih.gov/30321826/
Gill SK, Rossi M, Bajka B, Whelan K. Dietary fibre in gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol. 2021;18(2):101–16. https://pubmed.ncbi.nlm.nih.gov/33208922/
Threapleton DE, Greenwood DC, Evans CE, et al. Dietary fibre intake and risk of cardiovascular disease: systematic review and meta-analysis. BMJ. 2013;347:f6879. https://pubmed.ncbi.nlm.nih.gov/24355537/
Gill SK, Rossi M, Bajka B, Whelan K. Dietary fibre in gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol. 2021;18(2):101–16. https://pubmed.ncbi.nlm.nih.gov/33208922/
Maskarinec G, Takata Y, Pagano I, et al. Trends and dietary determinants of overweight and obesity in a multiethnic population. Obesity (Silver Spring). 2006;14(4):717–26. https://pubmed.ncbi.nlm.nih.gov/16741275/
Yao B, Fang H, Xu W, et al. Dietary fiber intake and risk of type 2 diabetes: a dose-response analysis of prospective studies. Eur J Epidemiol. 2014;29(2):79–88. https://pubmed.ncbi.nlm.nih.gov/24389767/
Fatahi S, Matin SS, Sohouli MH, et al. Association of dietary fiber and depression symptom: a systematic review and meta-analysis of observational studies. Complement Ther Med. 2021;56:102621. https://pubmed.ncbi.nlm.nih.gov/33220451/
Kim Y, Je Y. Dietary fiber intake and total mortality: a meta-analysis of prospective cohort studies. Am J Epidemiol. 2014;180(6):565–73. https://pubmed.ncbi.nlm.nih.gov/25143474/
Threapleton DE, Greenwood DC, Evans CEL, et al. Dietary fibre intake and risk of cardiovascular disease: systematic review and meta-analysis. BMJ. 2013;347:f6879. https://pubmed.ncbi.nlm.nih.gov/24355537/
Wolever TM, Jenkins DJ. What is a high fiber diet? Adv Exp Med Biol. 1997;427:35–42. https://pubmed.ncbi.nlm.nih.gov/9361828/
Shaper AG, Jones KW. Serum-cholesterol, diet, and coronary heart-disease in Africans and Asians in Uganda: 1959. Int J Epidemiol. 2012;41(5):1221–5. https://pubmed.ncbi.nlm.nih.gov/23045195/
Singh SA, Trowell HC. A case of coronary heart disease in an African. East Afr Med J. 1956;33(10):391–4. https://pubmed.ncbi.nlm.nih.gov/13375489/
Ikem I, Sumpio BE. Cardiovascular disease: the new epidemic in sub-Saharan Africa. Vascular. 2011;19(6):301–7. https://pubmed.ncbi.nlm.nih.gov/21940758/
Burkitt DP, Walker AR, Painter NS. Effect of dietary fibre on stools and the transit-times, and its role in the causation of disease. Lancet. 1972;2(7792):1408–12. https://pubmed.ncbi.nlm.nih.gov/4118696/
Dietary fiber market to reach $3.25 billion by 2017. Neutraceuticals World. https://nutraceuticalsworld.com/contents/view_breaking-news/2012–10–29/dietary-fiber-market-to-reach-325-billion-by-2017. Published October 29, 2012. Accessed March 29, 2022.; https://nutraceuticalsworld.com/contents/view_breaking-news/2012-10-29/dietary-fiber-market-to-reach-325-billion-by-2017
Eastwood M, Kritchevsky D. Dietary fiber: how did we get where we are? Annu Rev Nutr. 2005;25:1–8. https://pubmed.ncbi.nlm.nih.gov/16011456/
Threapleton DE, Greenwood DC, Evans CE, et al. Dietary fibre intake and risk of cardiovascular disease: systematic review and meta-analysis. BMJ. 2013;347:f6879. https://pubmed.ncbi.nlm.nih.gov/24355537/
Eastwood M, Kritchevsky D. Dietary fiber: how did we get where we are? Annu Rev Nutr. 2005;25:1–8. https://pubmed.ncbi.nlm.nih.gov/16011456/
McRorie JW. Evidence-based approach to fiber supplements and clinically meaningful health benefits, part 2: what to look for and how to recommend an effective fiber therapy. Nutr Today. 2015;50(2):90–7. https://pubmed.ncbi.nlm.nih.gov/25972618/
Agricultural Research Service, United States Department of Agriculture. Seeds, flaxseeds. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/food-details/169414/nutrients. Published April 1, 2019. Accessed February 22, 2023.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/169414/nutrients
Soltanian N, Janghorbani M. A randomized trial of the effects of flaxseed to manage constipation, weight, glycemia, and lipids in constipated patients with type 2 diabetes. Nutr Metab (Lond). 2018;15:36. https://pubmed.ncbi.nlm.nih.gov/29760761/
Soltanian N, Janghorbani M. Effect of flaxseed or psyllium vs. placebo on management of constipation, weight, glycemia, and lipids: a randomized trial in constipated patients with type 2 diabetes. Clin Nutr ESPEN. 2019;29:41–8. https://pubmed.ncbi.nlm.nih.gov/30661699/
Sun J, Bai H, Ma J, et al. Effects of flaxseed supplementation on functional constipation and quality of life in a Chinese population: a randomized trial. Asia Pac J Clin Nutr. 2020;29(1):61–7. https://pubmed.ncbi.nlm.nih.gov/32229443/
Hongisto SM, Paajanen L, Saxelin M, Korpela R. A combination of fibre-rich rye bread and yoghurt containing Lactobacillus GG improves bowel function in women with self-reported constipation. Eur J Clin Nutr. 2006;60(3):319–24. https://pubmed.ncbi.nlm.nih.gov/16251881/
Almario CV, Almario AA, Cunningham ME, Fouladian J, Spiegel BMR. Old farts – fact or fiction? Results from a population-based survey of 16,000 Americans examining the association between age and flatus. Clin Gastroenterol Hepatol. 2017;15(8):1308–10. https://pubmed.ncbi.nlm.nih.gov/28344066/
Behm RM. A special recipe to banish constipation. Geriatr Nurs. 1985;6(4):216–7. https://pubmed.ncbi.nlm.nih.gov/2989122/
Hull MA, McIntire DD, Atnip SD, et al. Randomized trial comparing 2 fiber regimens for the reduction of symptoms of constipation. Female Pelvic Med Reconstr Surg. 2011;17(3):128–33. https://pubmed.ncbi.nlm.nih.gov/22453784/
Lever E, Cole J, Scott SM, Emery PW, Whelan K. Systematic review: the effect of prunes on gastrointestinal function. Aliment Pharmacol Ther. 2014;40(7):750–8. https://pubmed.ncbi.nlm.nih.gov/25109788/
Sanjoaquin MA, Appleby PN, Spencer EA, Key TJ. Nutrition and lifestyle in relation to bowel movement frequency: a cross-sectional study of 20 630 men and women in EPIC – Oxford. Public Health Nutr. 2004;7(1):77–83. https://pubmed.ncbi.nlm.nih.gov/14972075/
Attaluri A, Donahoe R, Valestin J, Brown K, Rao SSC. Randomised clinical trial: dried plums (prunes) vs. psyllium for constipation. Aliment Pharmacol Ther. 2011;33(7):822–8. https://pubmed.ncbi.nlm.nih.gov/21323688/
Baek HI, Ha KC, Kim HM, et al. Randomized, double-blind, placebo-controlled trial of Ficus carica paste for the management of functional constipation. Asia Pac J Clin Nutr. 2016;25(3):487–96. https://pubmed.ncbi.nlm.nih.gov/27440682/
Venancio VP, Kim H, Sirven MA, et al. Polyphenol-rich mango (Mangifera indica L.) ameliorate functional constipation symptoms in humans beyond equivalent amount of fiber. Mol Nutr Food Res. 2018;62(12):e1701034. https://pubmed.ncbi.nlm.nih.gov/29733520/
Ojo B, El-Rassi GD, Payton ME, et al. Mango supplementation modulates gut microbial dysbiosis and short-chain fatty acid production independent of body weight reduction in C57BL/6 mice fed a high-fat diet. J Nutr. 2016;146(8):1483–91. https://pubmed.ncbi.nlm.nih.gov/27358411/
Kim H, Venancio VP, Fang C, Dupont AW, Talcott ST, Mertens-Talcott SU. Mango (Mangifera indica L.) polyphenols reduce IL-8, GRO, and GM-SCF plasma levels and increase Lactobacillus species in a pilot study in patients with inflammatory bowel disease. Nutr Res. 2020;75:85–94. https://pubmed.ncbi.nlm.nih.gov/32109839/
What are the key statistics about colorectal cancer? American Cancer Society website. http://www.cancer.org/cancer/colonandrectumcancer/detailedguide/colorectal-cancer-key-statistics. Updated January 12, 2022. Accessed March 29, 2022.; https://www.cancer.org/cancer/types/colon-rectal-cancer/about/key-statistics.html
American Cancer Society. Cancer Facts & Figures 2014. American Cancer Society; 2014. https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2014.html
U.S. Preventive Services Task Force. Screening for colorectal cancer. U.S. Preventive Services Task Force website. http://www.uspreventiveservicestaskforce.org/Home/GetFile/1/467/colcancsumm/pdf. Accessed March 29, 2022.; https://www.uspreventiveservicestaskforce.org/Home/GetFile/1/467/colcancsumm/pdf
Wender RC. Colorectal cancer screening should begin at 45. J Gastroenterol Hepatol. 2020;35(9):1461–3. https://pubmed.ncbi.nlm.nih.gov/32944996/
Anderson JC, Samadder JN. To screen or not to screen adults 45–49 years of age: that is the question. Am J Gastroenterol. 2018;113(12):1750–3. https://pubmed.ncbi.nlm.nih.gov/30385833/
Davidson KW, Barry MJ, Mangione CM, et al. Screening for colorectal cancer: US Preventive Services Task Force recommendation statement. JAMA. 2021;325(19):1965–77. https://pubmed.ncbi.nlm.nih.gov/34003218/
Mannucci A, Zuppardo RA, Rosati R, Leo MD, Perea J, Cavestro GM. Colorectal cancer screening from 45 years of age: thesis, antithesis and synthesis. World J Gastroenterol. 2019;25(21):2565–80. https://pubmed.ncbi.nlm.nih.gov/31210710/
Anderson JC, Samadder JN. To screen or not to screen adults 45–49 years of age: that is the question. Am J Gastroenterol. 2018;113(12):1750–3. https://pubmed.ncbi.nlm.nih.gov/30385833/
Hussan H, Patel A, Le Roux M, et al. Rising incidence of colorectal cancer in young adults corresponds with increasing surgical resections in obese patients. Clin Transl Gastroenterol. 2020;11(4):e00160. https://pubmed.ncbi.nlm.nih.gov/32352680/
Dairi O, Anderson JC, Butterly LF. Why is colorectal cancer increasing in younger age groups in the United States? Expert Rev Gastroenterol Hepatol. 2021;15(6):623–32. https://pubmed.ncbi.nlm.nih.gov/33480301/
U.S. Cancer Statistics Working Group. U.S. Cancer Statistics data visualizations tool, based on 2020 submission data (1999–2018). U.S. Department of Health and Human Services, Centers for Disease Control and Prevention and National Cancer Institute. www.cdc.gov/cancer/dataviz. Published June 2021. Accessed May 11, 2022.; https://gis.cdc.gov/Cancer/USCS/?CDC_AA_refVal=https%3A%2F%2Fwww.cdc.gov%2Fcancer%2Fdataviz%2Findex.htm#/AtAGlance/
Khan AM, Mucci LA. Concerning trends in colorectal cancer in the wake of Chadwick Boseman’s death. J Cancer Policy. 2020;26:100260. https://pubmed.ncbi.nlm.nih.gov/35656888/
Mueller NM, Hyams T, King-Marshall EC, Curbow BA. Colorectal cancer knowledge and perceptions among individuals below the age of 50. Psychooncology. 2022;31(3):436–41. https://pubmed.ncbi.nlm.nih.gov/34546622/
Imperiale TF, Kahi CJ, Rex DK. Lowering the starting age for colorectal cancer screening to 45 years: who will come… and should they? Clin Gastroenterol Hepatol. 2018;16(10):1541–4. https://pubmed.ncbi.nlm.nih.gov/30114484/
Davidson KW, Barry MJ, Mangione CM, et al. Screening for colorectal cancer: US Preventive Services Task Force recommendation statement. JAMA. 2021;325(19):1965–77. https://pubmed.ncbi.nlm.nih.gov/34003218/
Yabroff KR, Klabunde CN, Yuan G, et al. Are physicians’ recommendations for colorectal cancer screening guideline – consistent? J Gen Intern Med. 2011;26(2):177–84. https://pubmed.ncbi.nlm.nih.gov/20949328/
Swan H, Siddiqui AA, Myers RE. International colorectal cancer screening programs: population contact strategies, testing methods and screening rates. Pract Gastroenter. 2012;36(8):20–9. https://www.researchgate.net/publication/286884668_International_colorectal_cancer_screening_programs_Population_contact_strategies_testing_methods_and_screening_rates
Swan H, Siddiqui AA, Myers RE. International colorectal cancer screening programs: population contact strategies, testing methods and screening rates. Pract Gastroenter. 2012;36(8):20–9. https://www.researchgate.net/publication/286884668_International_colorectal_cancer_screening_programs_Population_contact_strategies_testing_methods_and_screening_rates
Butterfield S. Changes coming for colon cancer screening. ACP Internist. 2014;34(7):10–11. https://acpinternist.org/archives/2014/07/colonoscopy.htm
Hoffman RM, Levy BT, Allison JE. Rising use of multitarget stool DNA testing for colorectal cancer. JAMA Netw Open. 2021;4(9):e2122328. https://pubmed.ncbi.nlm.nih.gov/34473264/
Wang K, Ma W, Wu K, et al. Healthy lifestyle, endoscopic screening, and colorectal cancer incidence and mortality in the United States: a nationwide cohort study. PLoS Med. 2021;18(2):e1003522. https://pubmed.ncbi.nlm.nih.gov/33524029/
Larsen IK, Grotmol T, Almendingen K, Hoff G. Impact of colorectal cancer screening on future lifestyle choices: a three-year randomized controlled trial. Clin Gastroenterol Hepatol. 2007;5(4):477–83. https://pubmed.ncbi.nlm.nih.gov/17363335/
Hoff G, Thiis-Evensen E, Grotmol T, Sauar J, Vatn MH, Moen IE. Do undesirable effects of screening affect all-cause mortality in flexible sigmoidoscopy programmes? Experience from the Telemark Polyp Study 1983–1996. Eur J Cancer Prev. 2001;10(2):131–7. https://pubmed.ncbi.nlm.nih.gov/19483252/
Berstad P, Løberg M, Larsen IK, et al. Long-term lifestyle changes after colorectal cancer screening: randomised controlled trial. Gut. 2015;64(8):1268–76. https://pubmed.ncbi.nlm.nih.gov/25183203/
Wang K, Ma W, Wu K, et al. Healthy lifestyle, endoscopic screening, and colorectal cancer incidence and mortality in the United States: a nationwide cohort study. PLoS Med. 2021;18(2):e1003522. https://pubmed.ncbi.nlm.nih.gov/33524029/
Platz EA, Willett WC, Colditz GA, Rimm EB, Spiegelman D, Giovannucci E. Proportion of colon cancer risk that might be preventable in a cohort of middle-aged US men. Cancer Causes Control. 2000;11(7):579–88. https://pubmed.ncbi.nlm.nih.gov/10977102/
O’Keefe SJD, Li JV, Lahti L, et al. Fat, fiber and cancer risk in African Americans and rural Africans. Nat Commun. 2015;6:6342. https://pubmed.ncbi.nlm.nih.gov/25919227/
O’Keefe SJD, Li JV, Lahti L, et al. Fat, fiber and cancer risk in African Americans and rural Africans. Nat Commun. 2015;6:6342. https://pubmed.ncbi.nlm.nih.gov/25919227/
Weber C. Nutrition. Diet change alters microbiota and might affect cancer risk. Nat Rev Gastroenterol Hepatol. 2015;12(6):314. https://pubmed.ncbi.nlm.nih.gov/25963512/
Zhao Y, Zhan J, Wang Y, Wang D. The relationship between plant-based diet and risk of digestive system cancers: a meta-analysis based on 3,059,009 subjects. Front Public Health. 2022;10. https://pubmed.ncbi.nlm.nih.gov/35719615/
McCarty MF. Mortality from Western cancers rose dramatically among African-Americans during the 20th century: are dietary animal products to blame? Med Hypotheses. 2001;57(2):169–74. https://pubmed.ncbi.nlm.nih.gov/11461167/
Milsom I, Gyhagen M. The prevalence of urinary incontinence. Climacteric. 2019;22(3):217–22. https://pubmed.ncbi.nlm.nih.gov/30572737/
Wieland LS, Shrestha N, Lassi ZS, Panda S, Chiaramonte D, Skoetz N. Yoga for treating urinary incontinence in women. Cochrane Database Syst Rev. 2019;2:CD012668. https://pubmed.ncbi.nlm.nih.gov/30816997/
Pearlman A, Kreder K. Evaluation and treatment of urinary incontinence in the aging male. Postgrad Med. 2020;132(sup4):9–17. https://pubmed.ncbi.nlm.nih.gov/33017202/
Faleiro DJA, Menezes EC, Capeletto E, Fank F, Porto RM, Mazo GZ. Association of physical activity with urinary incontinence in older women: a systematic review. J Aging Phys Act. 2019;27(4):906–13. https://pubmed.ncbi.nlm.nih.gov/30859902/
Specht JKP. 9 myths of incontinence in older adults: both clinicians and the over-65 set need to know more. Am J Nurs. 2005;105(6):58–68. https://pubmed.ncbi.nlm.nih.gov/15930873/
Muller N. Myths about incontinence in aging adults. Ostomy Wound Manage. 2009;55(5):22. https://pubmed.ncbi.nlm.nih.gov/20560204/
Milsom I, Gyhagen M. The prevalence of urinary incontinence. Climacteric. 2019;22(3):217–22. https://pubmed.ncbi.nlm.nih.gov/30572737/
Specht JKP. 9 myths of incontinence in older adults: both clinicians and the over-65 set need to know more. Am J Nurs. 2005;105(6):58–68. https://pubmed.ncbi.nlm.nih.gov/15930873/
Pizzol D, Demurtas J, Celotto S, et al. Urinary incontinence and quality of life: a systematic review and meta-analysis. Aging Clin Exp Res. 2021;33(1):25–35. https://pubmed.ncbi.nlm.nih.gov/32964401/
Milsom I, Gyhagen M. The prevalence of urinary incontinence. Climacteric. 2019;22(3):217–22. https://pubmed.ncbi.nlm.nih.gov/30572737/
Ashton-Miller JA, DeLancey JOL. Functional anatomy of the female pelvic floor. Ann N Y Acad Sci. 2007;1101:266–96. https://pubmed.ncbi.nlm.nih.gov/17416924/
Kuh D, Cardozo L, Hardy R. Urinary incontinence in middle aged women: childhood enuresis and other lifetime risk factors in a British prospective cohort. J Epidemiol Community Health. 1999;53(8):453–8. https://pubmed.ncbi.nlm.nih.gov/10562862/
Danforth KN, Townsend MK, Lifford K, Curhan GC, Resnick NM, Grodstein F. Risk factors for urinary incontinence among middle-aged women. Am J Obstet Gynecol. 2006;194(2):339–45. https://pubmed.ncbi.nlm.nih.gov/16458626/
Robinson D, Giarenis I, Cardozo L. You are what you eat: the impact of diet on overactive bladder and lower urinary tract symptoms. Maturitas. 2014;79(1):8–13. https://pubmed.ncbi.nlm.nih.gov/25033724/
Imamura M, Williams K, Wells M, McGrother C. Lifestyle interventions for the treatment of urinary incontinence in adults. Cochrane Database Syst Rev. 2015;(12):CD003505. https://pubmed.ncbi.nlm.nih.gov/26630349/
Subak LL, Wing R, West DS, et al. Weight loss to treat urinary incontinence in overweight and obese women. N Engl J Med. 2009;360(5):481–90. https://pubmed.ncbi.nlm.nih.gov/19179316/
Stewart WF, Van Rooyen JB, Cundiff GW, et al. Prevalence and burden of overactive bladder in the United States. World J Urol. 2003;20(6):327–36. https://pubmed.ncbi.nlm.nih.gov/12811491/
Flore K, Fauber J, Wynn M. Drug firms helped create $3 billion overactive bladder market. Milwaukee Journal Sentinel. https://www.jsonline.com/story/news/investigations/2016/10/16/overactive-bladder-drug-companies-helped-create-3-billion-market/92030360/. Published October 15, 2016. Accessed August 24, 2022.; https://www.jsonline.com/story/news/investigations/2016/10/16/overactive-bladder-drug-companies-helped-create-3-billion-market/92030360/
Mitcheson HD, Samanta S, Muldowney K, et al. Vibegron (RVT-901/MK-4618/KRP-114V) administered once daily as monotherapy or concomitantly with tolterodine in patients with an overactive bladder: a multicenter, phase IIb, randomized, double-blind, controlled trial. Eur Urol. 2019;75(2):274–82. https://pubmed.ncbi.nlm.nih.gov/30661513/
Cho A, Eidelberg A, Butler DJ, et al. Efficacy of daily intake of dried cranberry 500 mg in women with overactive bladder: a randomized, double-blind, placebo controlled study. J Urol. 2021;205(2):507–13. https://pubmed.ncbi.nlm.nih.gov/32945735/
Ernst M, Gonka J, Povcher O, Kim J. Diet modification for overactive bladder: an evidence-based review. Curr Bladder Dysfunct Rep. 2015;10(1):25–30. https://link.springer.com/article/10.1007/s11884-014-0285-0
Dallosso H, Matthews R, McGrother C, Donaldson M. Diet as a risk factor for the development of stress urinary incontinence: a longitudinal study in women. Eur J Clin Nutr. 2004;58(6):920–6. https://pubmed.ncbi.nlm.nih.gov/15164113/
Robinson D, Giarenis I, Cardozo L. You are what you eat: the impact of diet on overactive bladder and lower urinary tract symptoms. Maturitas. 2014;79(1):8–13. https://pubmed.ncbi.nlm.nih.gov/25033724/
Urinary Incontinence and Pelvic Organ Prolapse in Women: Management. National Institute for Health and Care Excellence (NICE); 2019. https://pubmed.ncbi.nlm.nih.gov/31008559/
Burkhard FC, Bosch JLHR, Cruz F, et al. EAU guidelines on urinary incontinence. Vol 3. Eur Urol. 2011;59(3):387–400. https://pubmed.ncbi.nlm.nih.gov/21130559/
Le Berre M, Presse N, Morin M, et al. What do we really know about the role of caffeine on urinary tract symptoms? A scoping review on caffeine consumption and lower urinary tract symptoms in adults. Neurourol Urodyn. 2020;39(5):1217–33. https://pubmed.ncbi.nlm.nih.gov/32270903/
Sun S, Liu D, Jiao Z. Coffee and caffeine intake and risk of urinary incontinence: a meta-analysis of observational studies. BMC Urol. 2016;16(1):61. https://pubmed.ncbi.nlm.nih.gov/27716171/
Muller N. Myths about incontinence in aging adults. Ostomy Wound Manage. 2009;55(5):22. https://pubmed.ncbi.nlm.nih.gov/20560204/
Dasgupta J, Elliott RA, Doshani A, Tincello DG. Enhancement of rat bladder contraction by artificial sweeteners via increased extracellular Ca2+ influx. Toxicol Appl Pharmacol. 2006;217(2):216–24. https://pubmed.ncbi.nlm.nih.gov/17046038/
Russo E, Caretto M, Giannini A, et al. Management of urinary incontinence in postmenopausal women: an EMAS clinical guide. Maturitas. 2021;143:223–30. https://pubmed.ncbi.nlm.nih.gov/33008675/
Riemsma R, Hagen S, Kirschner-Hermanns R, et al. Can incontinence be cured? A systematic review of cure rates. BMC Med. 2017;15(1):63. https://pubmed.ncbi.nlm.nih.gov/28335792/
Wagg A, Compion G, Fahey A, Siddiqui E. Persistence with prescribed antimuscarinic therapy for overactive bladder: a UK experience. BJU Int. 2012;110(11):1767–74. https://pubmed.ncbi.nlm.nih.gov/22409769/
Hu JS, Pierre EF. Urinary incontinence in women: evaluation and management. Am Fam Physician. 2019;100(6):339–48. https://pubmed.ncbi.nlm.nih.gov/31524367/
Riemsma R, Hagen S, Kirschner-Hermanns R, et al. Can incontinence be cured? A systematic review of cure rates. BMC Med. 2017;15(1):63. https://pubmed.ncbi.nlm.nih.gov/28335792/
Cody JD, Jacobs ML, Richardson K, Moehrer B, Hextall A. Oestrogen therapy for urinary incontinence in post-menopausal women. Cochrane Database Syst Rev. 2012;2012(10):CD001405. https://pubmed.ncbi.nlm.nih.gov/23076892/
Russo E, Caretto M, Giannini A, et al. Management of urinary incontinence in postmenopausal women: an EMAS clinical guide. Maturitas. 2021;143:223–30. https://pubmed.ncbi.nlm.nih.gov/33008675/
Cody JD, Jacobs ML, Richardson K, Moehrer B, Hextall A. Oestrogen therapy for urinary incontinence in post-menopausal women. Cochrane Database Syst Rev. 2012;2012(10):CD001405. https://pubmed.ncbi.nlm.nih.gov/23076892/
Russo E, Caretto M, Giannini A, et al. Management of urinary incontinence in postmenopausal women: an EMAS clinical guide. Maturitas. 2021;143:223–30. https://pubmed.ncbi.nlm.nih.gov/33008675/
Kegel AH. Stress incontinence and genital relaxation; a nonsurgical method of increasing the tone of sphincters and their supporting structures. Ciba Clin Symp. 1952;4(2):35–51. https://pubmed.ncbi.nlm.nih.gov/14905555/
Kegel exercises: a how-to guide for women. Mayo Clinic. https://www.mayoclinic.org/healthy-lifestyle/womens-health/in-depth/kegel-exercises/art-20045283. Published September 15, 2020. Accessed August 24, 2022.; https://www.mayoclinic.org/healthy-lifestyle/womens-health/in-depth/kegel-exercises/art-20045283
Specht JKP. 9 myths of incontinence in older adults: both clinicians and the over-65 set need to know more. Am J Nurs. 2005;105(6):58–68. https://pubmed.ncbi.nlm.nih.gov/15930873/
Nazarpour S, Simbar M, Ramezani Tehrani F, Alavi Majd H. Effects of sex education and Kegel exercises on the sexual function of postmenopausal women: a randomized clinical trial. J Sex Med. 2017;14(7):959–67. https://pubmed.ncbi.nlm.nih.gov/28601506/
Vaughan CP, Markland AD. Urinary incontinence in women. Ann Intern Med. 2020;172(3):ITC17. https://pubmed.ncbi.nlm.nih.gov/32016335/
Kilpatrick KA, Paton P, Subbarayan S, et al. Non-pharmacological, non-surgical interventions for urinary incontinence in older persons: a systematic review of systematic reviews. The SENATOR project ONTOP series. Maturitas. 2020;133:42–8. https://pubmed.ncbi.nlm.nih.gov/32005422/
Dumoulin C, Cacciari LP, Hay-Smith EJC. Pelvic floor muscle training versus no treatment, or inactive control treatments, for urinary incontinence in women. Cochrane Database Syst Rev. 2018;(10). https://pubmed.ncbi.nlm.nih.gov/30288727/
Faleiro DJA, Menezes EC, Capeletto E, Fank F, Porto RM, Mazo GZ. Association of physical activity with urinary incontinence in older women: a systematic review. J Aging Phys Act. 2019;27(4):906–13. https://pubmed.ncbi.nlm.nih.gov/30859902/
Huang AJ, Chesney M, Lisha N, et al. A group-based yoga program for urinary incontinence in ambulatory women: feasibility, tolerability, and change in incontinence frequency over 3 months in a single-center randomized trial. Am J Obstet Gynecol. 2019;220(1):87.e1–13. https://pubmed.ncbi.nlm.nih.gov/30595143/
Wei JT, Calhoun E, Jacobsen SJ. Urologic Diseases in America Project: benign prostatic hyperplasia. J Urol. 2008;179(5 Suppl):S75–80. https://pubmed.ncbi.nlm.nih.gov/18405761/
Burnett AL, Wein AJ. Benign prostatic hyperplasia in primary care: what you need to know. J Urol. 2006;175(3 Pt 2):S19–24. https://pubmed.ncbi.nlm.nih.gov/16458735/
Trumble BC, Stieglitz J, Rodriguez DE, Linares EC, Kaplan HS, Gurven MD. Challenging the inevitability of prostate enlargement: low levels of benign prostatic hyperplasia among Tsimane forager-horticulturalists. J Gerontol A Biol Sci Med Sci. 2015;70(10):1262–8. https://pubmed.ncbi.nlm.nih.gov/25922348/
Taub DA, Wei JT. The economics of benign prostatic hyperplasia and lower urinary tract symptoms in the United States. Curr Urol Rep. 2006;7(4):272–81. https://pubmed.ncbi.nlm.nih.gov/16930498/
Zhang AY, Xu X. Prevalence, burden, and treatment of lower urinary tract symptoms in men aged 50 and older: a systematic review of the literature. SAGE Open Nurs. 2018;4:2377960818811773. https://pubmed.ncbi.nlm.nih.gov/33415211/
Traish AM, Mulgaonkar A, Giordano N. The dark side of 5a-reductase inhibitors’ therapy: sexual dysfunction, high Gleason grade prostate cancer and depression. Korean J Urol. 2014;55(6):367–79. https://pubmed.ncbi.nlm.nih.gov/24955220/
Cindolo L, Pirozzi L, Fanizza C, et al. Drug adherence and clinical outcomes for patients under pharmacological therapy for lower urinary tract symptoms related to benign prostatic hyperplasia: population-based cohort study. Eur Urol. 2015;68(3):418–25. https://pubmed.ncbi.nlm.nih.gov/25465970/
Roehrborn CG, Bruskewitz R, Nickel JC, et al. Sustained decrease in incidence of acute urinary retention and surgery with finasteride for 6 years in men with benign prostatic hyperplasia. J Urol. 2004;171(3):1194–8. https://pubmed.ncbi.nlm.nih.gov/14767299/
Irwig MS. How routine pharmacovigilance failed to identify finasteride’s persistent sexual side effects. Andrology. 2022;10(2):207–8. https://pubmed.ncbi.nlm.nih.gov/34713622/
Zhang AY, Xu X. Prevalence, burden, and treatment of lower urinary tract symptoms in men aged 50 and older: a systematic review of the literature. SAGE Open Nurs. 2018;4:2377960818811773. https://pubmed.ncbi.nlm.nih.gov/33415211/
Metcalfe C, Poon KS. Long-term results of surgical techniques and procedures in men with benign prostatic hyperplasia. Curr Urol Rep. 2011;12(4):265–73. https://pubmed.ncbi.nlm.nih.gov/21484456/
Burnett AL, Wein AJ. Benign prostatic hyperplasia in primary care: what you need to know. J Urol. 2006;175(3 Pt 2):S19–24. https://pubmed.ncbi.nlm.nih.gov/16458735/
Burnett AL, Wein AJ. Benign prostatic hyperplasia in primary care: what you need to know. J Urol. 2006;175(3 Pt 2):S19–24. https://pubmed.ncbi.nlm.nih.gov/16458735/
Gu F. Epidemiological survey of benign prostatic hyperplasia and prostatic cancer in China. Chin Med J. 2000;113(4):299–302. https://pubmed.ncbi.nlm.nih.gov/11775222/
Kraft TS, Stieglitz J, Trumble BC, Martin M, Kaplan H, Gurven M. Nutrition transition in 2 lowland Bolivian subsistence populations. Am J Clin Nutr. 2018;108(6):1183–95. https://pubmed.ncbi.nlm.nih.gov/30383188/
Trumble BC, Stieglitz J, Rodriguez DE, Linares EC, Kaplan HS, Gurven MD. Challenging the inevitability of prostate enlargement: low levels of benign prostatic hyperplasia among Tsimane forager-horticulturalists. J Gerontol A Biol Sci Med Sci. 2015;70(10):1262–8. https://pubmed.ncbi.nlm.nih.gov/25922348/
Cicero AFG, Allkanjari O, Busetto GM, et al. Nutraceutical treatment and prevention of benign prostatic hyperplasia and prostate cancer. Arch Ital Urol Androl. 2019;91(3). https://pubmed.ncbi.nlm.nih.gov/31577095/
Koskimäki J, Hakama M, Huhtala H, Tammela TL. Association of dietary elements and lower urinary tract symptoms. Scand J Urol Nephrol. 2000;34(1):46–50. https://pubmed.ncbi.nlm.nih.gov/10757270/
Bravi F, Bosetti C, Dal Maso L, et al. Food groups and risk of benign prostatic hyperplasia. Urology. 2006;67(1):73–9. https://pubmed.ncbi.nlm.nih.gov/16413336/
Galeone C, Pelucchi C, Talamini R, et al. Onion and garlic intake and the odds of benign prostatic hyperplasia. Urology. 2007;70(4):672–6. https://pubmed.ncbi.nlm.nih.gov/17991535/
Bravi F, Bosetti C, Dal Maso L, et al. Food groups and risk of benign prostatic hyperplasia. Urology. 2006;67(1):73–9. https://pubmed.ncbi.nlm.nih.gov/16413336/
Bhagwat S, Haytowitz DB, Holden JM. USDA database for the isoflavone content of selected foods: release 2.0. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/arsuserfiles/80400525/data/isoflav/isoflav_r2.pdf. Published September 2008. Accessed April 15, 2022.; https://www.ars.usda.gov/arsuserfiles/80400525/data/isoflav/isoflav_r2.pdf
Wong SYS, Lau WWY, Leung PC, Leung JCS, Woo J. The association between isoflavone and lower urinary tract symptoms in elderly men. Br J Nutr. 2007;98(6):1237–42. https://pubmed.ncbi.nlm.nih.gov/17640419/
Zhou Z, Wang Z, Chen C, et al. Transurethral prostate vaporization using an oval electrode in 82 cases of benign prostatic hyperplasia. Chin Med J. 1998;111(1):52–5. https://pubmed.ncbi.nlm.nih.gov/10322654/
Ornish D, Weidner G, Fair WR, et al. Intensive lifestyle changes may affect the progression of prostate cancer. J Urol. 2005;174(3):1065–9. https://pubmed.ncbi.nlm.nih.gov/16094059/
Barnard RJ, Gonzalez JH, Liva ME, Ngo TH. Effects of a low-fat, high-fiber diet and exercise program on breast cancer risk factors in vivo and tumor cell growth and apoptosis in vitro. Nutr Cancer. 2006;55(1):28–34. https://pubmed.ncbi.nlm.nih.gov/16965238/
Barnard RJ, Kobayashi N, Aronson WJ. Effect of diet and exercise intervention on the growth of prostate epithelial cells. Prostate Cancer Prostatic Dis. 2008;11(4):362–6. https://pubmed.ncbi.nlm.nih.gov/18283296/
Карликовая пальма, произрастающая на восточном побережье Северной Америки. – Примеч. ред.
Keehn A, Taylor J, Lowe FC. Phytotherapy for benign prostatic hyperplasia. Curr Urol Rep. 2016;17(7):53. https://pubmed.ncbi.nlm.nih.gov/27180172/
Trivisonno LF, Sgarbossa N, Alvez GA, et al. Serenoa repens for the treatment of lower urinary tract symptoms due to benign prostatic enlargement: a systematic review and meta-analysis. Investig Clin Urol. 2021;62(5):520–34. https://pubmed.ncbi.nlm.nih.gov/34488251/
Zendehdel A, Ansari M, Khatami F, Mansoursamaei S, Dialameh H. The effect of vitamin D supplementation on the progression of benign prostatic hyperplasia: a randomized controlled trial. Clin Nutr. 2021;40(5):3325–31. https://pubmed.ncbi.nlm.nih.gov/33213976/
Safwat AS, Hasanain A, Shahat A, et al. Cholecalciferol for the prophylaxis against recurrent urinary tract infection among patients with benign prostatic hyperplasia: a randomized, comparative study. World J Urol. 2019;37(7):1347–52. https://pubmed.ncbi.nlm.nih.gov/30361957/
Zhang W, Wang X, Liu Y, et al. Effects of dietary flaxseed lignan extract on symptoms of benign prostatic hyperplasia. J Med Food. 2008;11(2):207–14. https://pubmed.ncbi.nlm.nih.gov/18358071/
Vahlensieck W, Theurer C, Pfitzer E, Patz B, Banik N, Engelmann U. Effects of pumpkin seed in men with lower urinary tract symptoms due to benign prostatic hyperplasia in the one-year, randomized, placebo-controlled GRANU study. Urol Int. 2015;94(3):286–95. https://pubmed.ncbi.nlm.nih.gov/25196580/
Assessment report on Cucurbita pepo L., semen. European Medicines Agency. https://www.ema.europa.eu/en/documents/herbal-report/draft-assessment-report-cucurbita-pepo-l-semen_en.pdf. Published September 13, 2011. Accessed August 22, 2022.; https://www.ema.europa.eu/en/medicines/herbal/cucurbitae-semen
Matsuo T, Miyata Y, Sakai H. Effect of salt intake reduction on nocturia in patients with excessive salt intake. Neurourol Urodyn. 2019;38(3):927–33. https://pubmed.ncbi.nlm.nih.gov/30706965/
Bradley CS, Erickson BA, Messersmith EE, et al. Evidence for the impact of diet, fluid intake, caffeine, alcohol and tobacco on lower urinary tract symptoms: a systematic review. J Urol. 2017;198(5):1010–20. https://pubmed.ncbi.nlm.nih.gov/28479236/
Xue Z, Lin Y, Jiang Y, Wei N, Bi J. The evaluation of nocturia in patients with lower urinary tract symptoms suggestive of benign prostatic hyperplasia and the analysis of the curative effect after medical or placebo therapy for nocturia: a randomized placebo-controlled study. BMC Urol. 2018;18(1):115. https://pubmed.ncbi.nlm.nih.gov/30545338/
Johnson TM II, Sattin RW, Parmelee P, Fultz NH, Ouslander JG. Evaluating potentially modifiable risk factors for prevalent and incident nocturia in older adults. J Am Geriatr Soc. 2005;53(6):1011–6. https://pubmed.ncbi.nlm.nih.gov/15935026/
Tani M, Hirayama A, Torimoto K, Matsushita C, Yamada A, Fujimoto K. Guidance on water intake effectively improves urinary frequency in patients with nocturia. Int J Urol. 2014;21(6):595–600. https://pubmed.ncbi.nlm.nih.gov/24405404/
Soda T, Masui K, Okuno H, Terai A, Ogawa O, Yoshimura K. Efficacy of nondrug lifestyle measures for the treatment of nocturia. J Urol. 2010;184(3):1000–4. https://pubmed.ncbi.nlm.nih.gov/20643422/
Cho SY, Lee SL, Kim IS, Koo DH, Kim HK, Oh SJ. Short-term effects of systematized behavioral modification program for nocturia: a prospective study. Neurourol Urodyn. 2012;31(1):64–8. https://pubmed.ncbi.nlm.nih.gov/21826726/
Johnson TM. The chicken-or-egg dilemma with nocturia: which matters most, the water or the salt? J Clin Hypertens. 2020;22(4):639–41. https://pubmed.ncbi.nlm.nih.gov/32073711/
Matsuo T, Miyata Y, Sakai H. Daily salt intake is an independent risk factor for pollakiuria and nocturia. Int J Urol. 2017;24(5):384–9. https://pubmed.ncbi.nlm.nih.gov/28295650/
Alwis US, Monaghan TF, Haddad R, et al. Dietary considerations in the evaluation and management of nocturia. F1000Res. 2020;9(F1000 Faculty Rev):165. https://pubmed.ncbi.nlm.nih.gov/32185022/
Matsuo T, Miyata Y, Sakai H. Effect of salt intake reduction on nocturia in patients with excessive salt intake. Neurourol Urodyn. 2019;38(3):927–33. https://pubmed.ncbi.nlm.nih.gov/30706965/
Monaghan TF, Michelson KP, Wu ZD, et al. Sodium restriction improves nocturia in patients at a cardiology clinic. J Clin Hypertens (Greenwich). 2020;22(4):633–8. https://pubmed.ncbi.nlm.nih.gov/32049435/
Alwis US, Delanghe J, Dossche L, et al. Could evening dietary protein intake play a role in nocturnal polyuria? J Clin Med. 2020;9(8):E2532. https://pubmed.ncbi.nlm.nih.gov/32764521/
Vidlar A, Student V, Vostalova J, et al. Cranberry fruit powder (Flowens™) improves lower urinary tract symptoms in men: a double-blind, randomized, placebo-controlled study. World J Urol. 2016;34(3):419–24. https://pubmed.ncbi.nlm.nih.gov/26049866/
An YJ, Lee JY, Kim Y, Jun W, Lee YH. Cranberry powder attenuates benign prostatic hyperplasia in rats. J Med Food. 2020;23(12):1296–302. https://pubmed.ncbi.nlm.nih.gov/33136465/
Vidlar A, Vostalova J, Ulrichova J, et al. The effectiveness of dried cranberries (Vaccinium macrocarpon) in men with lower urinary tract symptoms. Br J Nutr. 2010;104(8):1181–9. https://pubmed.ncbi.nlm.nih.gov/20804630/
Vidlar A, Student V, Vostalova J, et al. Cranberry fruit powder (Flowens™) improves lower urinary tract symptoms in men: a double-blind, randomized, placebo-controlled study. World J Urol. 2016;34(3):419–24. https://pubmed.ncbi.nlm.nih.gov/26049866/
Ledda A, Belcaro G, Dugall M, et al. Supplementation with high titer cranberry extract (Anthocran®) for the prevention of recurrent urinary tract infections in elderly men suffering from moderate prostatic hyperplasia: a pilot study. Eur Rev Med Pharmacol Sci. 2016;20(24):5205–9. https://pubmed.ncbi.nlm.nih.gov/28051247/
Spettel S, Chughtai B, Feustel P, Kaufman A, Levin RM, De E. A prospective randomized double-blind trial of grape juice antioxidants in men with lower urinary tract symptoms. Neurourol Urodyn. 2013;32(3):261–5. https://pubmed.ncbi.nlm.nih.gov/22907790/
Edinger MS, Koff WJ. Effect of the consumption of tomato paste on plasma prostate – specific antigen levels in patients with benign prostate hyperplasia. Braz J Med Biol Res. 2006;39(8):1115–9. https://pubmed.ncbi.nlm.nih.gov/16906286/
Durak lker, Yilmaz E, Devrim E, Perk H, Kaçmaz M. Consumption of aqueous garlic extract leads to significant improvement in patients with benign prostate hyperplasia and prostate cancer. Nutr Res. 2003;23(2):199–204. https://www.sciencedirect.com/science/article/abs/pii/S0271531702004955?via%3Dihub
Jani B, Rajkumar C. Ageing and vascular ageing. Postgrad Med J. 2006;82(968):357–62. https://pubmed.ncbi.nlm.nih.gov/16754702/
Mosca L, Ferris A, Fabunmi R, Robertson RM, American Heart Association. Tracking women’s awareness of heart disease: an American Heart Association national study. Circulation. 2004;109(5):573–9. https://pubmed.ncbi.nlm.nih.gov/14761901/
Xu J. Mortality among centenarians in the United States, 2000–2014. NCHS Data Brief. 2016;(233):1–8. https://pubmed.ncbi.nlm.nih.gov/26828422/
Cushman M, Shay CM, Howard VJ, et al. Ten-year differences in women’s awareness related to coronary heart disease: results of the 2019 American Heart Association national survey: a special report from the American Heart Association. Circulation. 2021;143(7):e239–48. https://pubmed.ncbi.nlm.nih.gov/32954796/
Tao J, Qiu Y. All disease stems from vessels. Aging Med (Milton). 2020;3(4):224–5. https://pubmed.ncbi.nlm.nih.gov/33392426/
Jin K. A microcirculatory theory of aging. Aging Dis. 2019;10(3):676–83. https://pubmed.ncbi.nlm.nih.gov/31165010/
Möbius-Winkler S, Linke A, Adams V, Schuler G, Erbs S. How to improve endothelial repair mechanisms: the lifestyle approach. Expert Rev Cardiovasc Ther. 2010;8(4):573–80. https://pubmed.ncbi.nlm.nih.gov/20397830/
Sharma S, Pandey NN, Sinha M, et al. Randomized, double-blind, placebo-controlled trial to evaluate safety and therapeutic efficacy of angiogenesis induced by intraarterial autologous bone marrow-derived stem cells in patients with severe peripheral arterial disease. J Vasc Interv Radiol. 2021;32(2):157–63. https://pubmed.ncbi.nlm.nih.gov/33248918/
Altabas V, Altabas K, Kirigin L. Endothelial progenitor cells (EPCs) in ageing and age-related diseases: how currently available treatment modalities affect EPC biology, atherosclerosis, and cardiovascular outcomes. Mech Ageing Dev. 2016;159:49–62. https://pubmed.ncbi.nlm.nih.gov/26919825/
Hoetzer GL, Van Guilder GP, Irmiger HM, Keith RS, Stauffer BL, DeSouza CA. Aging, exercise, and endothelial progenitor cell clonogenic and migratory capacity in men. J Appl Physiol (1985). 2007;102(3):847–52. https://pubmed.ncbi.nlm.nih.gov/17158243/
Wang M, Monticone RE, McGraw KR. Proinflammation, profibrosis, and arterial aging. Aging Med (Milton). 2020;3(3):159–68. https://pubmed.ncbi.nlm.nih.gov/33103036/
Weech M, Altowaijri H, Mayneris-Perxachs J, et al. Replacement of dietary saturated fat with unsaturated fats increases numbers of circulating endothelial progenitor cells and decreases numbers of microparticles: findings from the randomized, controlled Dietary Intervention and VAScular function (DIVAS) study. Am J Clin Nutr. 2018;107(6):876–82. https://pubmed.ncbi.nlm.nih.gov/29741564/
Shi Q, Hubbard GB, Kushwaha RS, et al. Endothelial senescence after high-cholesterol, high-fat diet challenge in baboons. Am J Physiol Heart Circ Physiol. 2007;292(6):H2913–20. https://pubmed.ncbi.nlm.nih.gov/17277030/
Jeong HS, Kim S, Hong SJ, et al. Black raspberry extract increased circulating endothelial progenitor cells and improved arterial stiffness in patients with metabolic syndrome: a randomized controlled trial. J Med Food. 2016;19(4):346–52. https://pubmed.ncbi.nlm.nih.gov/26891216/
Choi EY, Lee H, Woo JS, et al. Effect of onion peel extract on endothelial function and endothelial progenitor cells in overweight and obese individuals. Nutrition. 2015;31(9):1131–5. https://pubmed.ncbi.nlm.nih.gov/26233871/
Kim W, Jeong MH, Cho SH, et al. Effect of green tea consumption on endothelial function and circulating endothelial progenitor cells in chronic smokers. Circ J. 2006;70(8):1052–7. https://pubmed.ncbi.nlm.nih.gov/16864941/
Keith M, Kuliszewski MA, Liao C, et al. A modified portfolio diet complements medical management to reduce cardiovascular risk factors in diabetic patients with coronary artery disease. Clin Nutr. 2015;34(3):541–8. https://pubmed.ncbi.nlm.nih.gov/25023926/
Steinberg D, Blumenthal S, Carleton RA, et al. Lowering blood cholesterol to prevent heart disease: NIH Consensus Development Conference statement. Nutr Rev. 1985;43(9):283–91. https://pubmed.ncbi.nlm.nih.gov/4058807/
Ference BA, Ginsberg HN, Graham I, et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur Heart J. 2017;38(32):2459–72. https://pubmed.ncbi.nlm.nih.gov/28444290/
Roberts WC. It’s the cholesterol, stupid! Am J Cardiol. 2010;106(9):1364–6. https://pubmed.ncbi.nlm.nih.gov/21029840/
Roberts WC. William Clifford Roberts, MD curriculum vitae. http://www.iscvdp.org/docs/WCRoberts-CV.pdf. Accessed May 13, 2022.;http://www.iscvdp.org/storage/app/media/william-clifford-roberts.pdf
Roberts WC. Quantitative extent of atherosclerotic plaque in the major epicardial coronary arteries in patients with fatal coronary heart disease, in coronary endarterectomy specimens, in aorta – coronary saphenous venous conduits, and means to prevent the plaques: a review after studying the coronary arteries for 50 years. Am J Cardiol. 2018;121(11):1413–35. https://pubmed.ncbi.nlm.nih.gov/29753395/
Ference BA, Ginsberg HN, Graham I, et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur Heart J. 2017;38(32):2459–72. https://pubmed.ncbi.nlm.nih.gov/28444290/
Fernández-Friera L, Fuster V, López-Melgar B, et al. Normal LDL – cholesterol levels are associated with subclinical atherosclerosis in the absence of risk factors. J Am Coll Cardiol. 2017;70(24):2979–91. https://pubmed.ncbi.nlm.nih.gov/29241485/
Nambi V, Bhatt DL. Primary prevention of atherosclerosis: time to take a selfie? J Am Coll Cardiol. 2017;70(24):2992–4. https://pubmed.ncbi.nlm.nih.gov/29241486/
Hochholzer W, Giugliano RP. Lipid lowering goals: back to nature? Ther Adv Cardiovasc Dis. 2010;4(3):185–91. https://pubmed.ncbi.nlm.nih.gov/20400493/
Fernández-Friera L, Fuster V, López-Melgar B, et al. Normal LDL – cholesterol levels are associated with subclinical atherosclerosis in the absence of risk factors. J Am Coll Cardiol. 2017;70(24):2979–91. https://pubmed.ncbi.nlm.nih.gov/29241485/
Gitin A, Pfeffer MA, Hennekens CH. Editorial commentary: the lower the LDL the better but how and how much? Trends Cardiovasc Med. 2018;28(5):355–6. https://pubmed.ncbi.nlm.nih.gov/29428160/
Law MR, Wald NJ. Risk factor thresholds: their existence under scrutiny. BMJ. 2002;324(7353):1570–6. https://pubmed.ncbi.nlm.nih.gov/12089098/
Hochholzer W, Giugliano RP. Lipid lowering goals: back to nature? Ther Adv Cardiovasc Dis. 2010;4(3):185–91. https://pubmed.ncbi.nlm.nih.gov/20400493/
O’Keefe JH, Cordain L, Harris WH, Moe RM, Vogel R. Optimal low-density lipoprotein is 50 to 70 mg/dL: lower is better and physiologically normal. J Am Coll Cardiol. 2004;43(11):2142–6. https://pubmed.ncbi.nlm.nih.gov/15172426/
Anderson JW, Konz EC, Jenkins DJ. Health advantages and disadvantages of weight-reducing diets: a computer analysis and critical review. J Am Coll Nutr. 2000;19(5):578–90. https://pubmed.ncbi.nlm.nih.gov/11022871/
Hochholzer W, Giugliano RP. Lipid lowering goals: back to nature? Ther Adv Cardiovasc Dis. 2010;4(3):185–91. https://pubmed.ncbi.nlm.nih.gov/20400493/
Law MR, Wald NJ. Risk factor thresholds: their existence under scrutiny. BMJ. 2002;324(7353):1570–6. https://pubmed.ncbi.nlm.nih.gov/12089098/
Roberts WC. Quantitative extent of atherosclerotic plaque in the major epicardial coronary arteries in patients with fatal coronary heart disease, in coronary endarterectomy specimens, in aorta – coronary saphenous venous conduits, and means to prevent the plaques: a review after studying the coronary arteries for 50 years. Am J Cardiol. 2018;121(11):1413–35. https://pubmed.ncbi.nlm.nih.gov/29753395/
Packard CJ. LDL cholesterol: How low to go? Trends Cardiovasc Med. 2018;28(5):348–54. https://pubmed.ncbi.nlm.nih.gov/29336946/
Packard CJ. LDL cholesterol: How low to go? Trends Cardiovasc Med. 2018;28(5):348–54. https://pubmed.ncbi.nlm.nih.gov/29336946/
Nambi V, Bhatt DL. Primary prevention of atherosclerosis: time to take a selfie? J Am Coll Cardiol. 2017;70(24):2992–4. https://pubmed.ncbi.nlm.nih.gov/29241486/
Hong KN, Fuster V, Rosenson RS, Rosendorff C, Bhatt DL. How low to go with glucose, cholesterol, and blood pressure in primary prevention of CVD. J Am Coll Cardiol. 2017;70(17):2171–85. https://pubmed.ncbi.nlm.nih.gov/29050566/
Baigent C, Blackwell L, Emberson J, et al. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet. 2010;376(9753):1670–81. https://pubmed.ncbi.nlm.nih.gov/21067804/
Guber K, Pemmasani G, Malik A, Aronow WS, Yandrapalli S, Frishman WH. Statins and higher diabetes mellitus risk: incidence, proposed mechanisms, and clinical implications. Cardiol Rev. 2021;29(6):314–22. https://pubmed.ncbi.nlm.nih.gov/32947479/
Hong KN, Fuster V, Rosenson RS, Rosendorff C, Bhatt DL. How low to go with glucose, cholesterol, and blood pressure in primary prevention of CVD. J Am Coll Cardiol. 2017;70(17):2171–85. https://pubmed.ncbi.nlm.nih.gov/29050566/
Glenn AJ, Li J, Lo K, et al. The Portfolio Diet and incident type 2 diabetes: findings from the Women’s Health Initiative prospective cohort study. Diabetes Care. 2023;46(1):28–37. https://pubmed.ncbi.nlm.nih.gov/36162007/
Sliding scale for LDL: how low should you go? The target for the safest amount of “bad” cholesterol continues to drift downward. Harv Heart Lett. 2011;21(12):5. https://pubmed.ncbi.nlm.nih.gov/21991609/
How low should your cholesterol go? Even lower may be better. For those at highest risk, very low cholesterol levels may help prevent a second heart attack or stroke. Health News. 2004;10(10):6. https://pubmed.ncbi.nlm.nih.gov/15584114/
De Biase SG, Fernandes SFC, Gianini RJ, Duarte JLG. Vegetarian diet and cholesterol and triglycerides levels. Arq Bras Cardiol. 2007;88(1):35–9. https://pubmed.ncbi.nlm.nih.gov/17364116/
Kahleova H, Levin S, Barnard ND. Vegetarian dietary patterns and cardiovascular disease. Prog Cardiovasc Dis. 2018;61(1):54–61. https://pubmed.ncbi.nlm.nih.gov/29800598/
The US Burden of Disease Collaborators, Mokdad AH, Ballestros K, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Huang Z, Xu A, Cheung BMY. The potential role of fibroblast growth factor 21 in lipid metabolism and hypertension. Curr Hypertens Rep. 2017;19(4):28. https://pubmed.ncbi.nlm.nih.gov/28337713/
Lewington S, Clarke R, Qizilbash N, et al. Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies [published correction appears in Lancet. 2003;361(9362):1060]. Lancet. 2002;360(9349):1903–13. https://pubmed.ncbi.nlm.nih.gov/12493255/
Kramer H, Cooper R. Pros and cons of intensive systolic blood pressure lowering. Curr Hypertens Rep. 2018;20(2):16. https://pubmed.ncbi.nlm.nih.gov/29511979/
Kjeldsen SE, Os I, Westheim A. Could adverse events offset the benefit of intensive blood pressure lowering treatment in the Systolic Blood Pressure Intervention Trial? J Hypertens. 2019;37(5):902–4. https://pubmed.ncbi.nlm.nih.gov/30920495/
Fuster V. No such thing as ideal blood pressure: a case for personalized medicine. J Am Coll Cardiol. 2016;67(25):3014–5. https://pubmed.ncbi.nlm.nih.gov/27339499/
Goldhamer A, Lisle D, Parpia B, Anderson SV, Campbell TC. Medically supervised water-only fasting in the treatment of hypertension. J Manipulative Physiol Ther. 2001;24(5):335–9. https://pubmed.ncbi.nlm.nih.gov/11416824/
McDougall J, Litzau K, Haver E, Saunders V, Spiller GA. Rapid reduction of serum cholesterol and blood pressure by a twelve-day, very low fat, strictly vegetarian diet. J Am Coll Nutr. 1995;14(5):491–6. https://pubmed.ncbi.nlm.nih.gov/8522729/
Brown MS, Goldstein JL. Biomedicine. Lowering LDL – not only how low, but how long? Science. 2006;311(5768):1721–3. https://pubmed.ncbi.nlm.nih.gov/16556829/
McGill HC, McMahan CA. Determinants of atherosclerosis in the young. Am J Cardiol. 1998;82(10B):30T-6T. https://pubmed.ncbi.nlm.nih.gov/9860371/
Strong JP, Malcom GT, McMahan CA, et al. Prevalence and extent of atherosclerosis in adolescents and young adults: implications for prevention from the Pathobiological Determinants of Atherosclerosis in Youth Study. JAMA. 1999;281(8):727–35. https://pubmed.ncbi.nlm.nih.gov/10052443/
Steinberg D, Glass CK, Witztum JL. Evidence mandating earlier and more aggressive treatment of hypercholesterolemia. Circulation. 2008;118(6):672–7. https://pubmed.ncbi.nlm.nih.gov/18678783/
Myerburg RJ, Junttila MJ. 2012. Sudden cardiac death caused by coronary heart disease. Circulation. 28;125(8):1043–52. https://pubmed.ncbi.nlm.nih.gov/22371442/
Brown MS, Goldstein JL. Biomedicine. Lowering LDL – not only how low, but how long? Science. 2006;311(5768):1721–3. https://pubmed.ncbi.nlm.nih.gov/16556829/
Steinberg D, Glass CK, Witztum JL. Evidence mandating earlier and more aggressive treatment of hypercholesterolemia. Circulation. 2008;118(6):672–7. https://pubmed.ncbi.nlm.nih.gov/18678783/
Cohen J, Pertsemlidis A, Kotowski IK, Graham R, Garcia CK, Hobbs HH. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9. Nat Genet. 2005;37(2):161–5. https://pubmed.ncbi.nlm.nih.gov/15654334/
Cohen JC, Boerwinkle E, Mosley TH, Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006;354(12):1264–72. https://pubmed.ncbi.nlm.nih.gov/16554528/
Robinson JG, Gidding SS. Curing atherosclerosis should be the next major cardiovascular prevention goal. J Am Coll Cardiol. 2014;63(25):2779–85. https://pubmed.ncbi.nlm.nih.gov/24814489/
Brown MS, Goldstein JL. Biomedicine. Lowering LDL – not only how low, but how long? Science. 2006;311(5768):1721–3. https://pubmed.ncbi.nlm.nih.gov/16556829/
Wang N, Fulcher J, Abeysuriya N, et al. Intensive LDL cholesterol – lowering treatment beyond current recommendations for the prevention of major vascular events: a systematic review and meta-analysis of randomised trials including 327¿037 participants. Lancet Diabetes Endocrinol. 2020;8(1):36–49. https://pubmed.ncbi.nlm.nih.gov/31862150/
Shapiro MD, Bhatt DL. “Cholesterol-years” for ASCVD risk prediction and treatment. J Am Coll Cardiol. 2020;76(13):1517–20. https://pubmed.ncbi.nlm.nih.gov/32972527/
Kaplan H, Thompson RC, Trumble BC, et al. Coronary atherosclerosis in indigenous South American Tsimane: a cross-sectional cohort study. Lancet. 2017;389(10080):1730–9. https://pubmed.ncbi.nlm.nih.gov/28320601/
Penson PE, Pirro M, Banach M. LDL–C: lower is better for longer – even at low risk. BMC Med. 2020;18(1):320. https://pubmed.ncbi.nlm.nih.gov/33032586/
Ference BA, Ginsberg HN, Graham I, et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur Heart J. 2017;38(32):2459–72. https://pubmed.ncbi.nlm.nih.gov/28444290/
Brown MS, Goldstein JL. Biomedicine. Lowering LDL – not only how low, but how long? Science. 2006;311(5768):1721–3. https://pubmed.ncbi.nlm.nih.gov/16556829/
Kahleova H, Levin S, Barnard ND. Vegetarian dietary patterns and cardiovascular disease. Prog Cardiovasc Dis. 2018;61(1):54–61. https://pubmed.ncbi.nlm.nih.gov/29800598/
O’Keefe JH, Cordain L, Harris WH, Moe RM, Vogel R. Optimal low-density lipoprotein is 50 to 70 mg/dL: lower is better and physiologically normal. J Am Coll Cardiol. 2004;43(11):2142–6. https://pubmed.ncbi.nlm.nih.gov/15172426/
Roberts WC. Cholesterol is the cause of atherosclerosis. Am J Cardiol. 2017;120(9):1696. https://pubmed.ncbi.nlm.nih.gov/28847597/
Kataoka Y, Hammadah M, Puri R, et al. Plaque microstructures in patients with coronary artery disease who achieved very low low-density lipoprotein cholesterol levels. Atherosclerosis. 2015;242(2):490–5. https://pubmed.ncbi.nlm.nih.gov/26298740/
Diamond DM, Ravnskov U. How statistical deception created the appearance that statins are safe and effective in primary and secondary prevention of cardiovascular disease. Expert Rev Clin Pharmacol. 2015;8(2):201–10. https://pubmed.ncbi.nlm.nih.gov/25672965/
Trewby PN, Reddy AV, Trewby CS, Ashton VJ, Brennan G, Inglis J. Are preventive drugs preventive enough? A study of patients’ expectation of benefit from preventive drugs. Clin Med (Lond). 2002;2(6):527–33. https://pubmed.ncbi.nlm.nih.gov/12528966/
Salami JA, Warraich H, Valero-Elizondo J, et al. National trends in statin use and expenditures in the US adult population from 2002 to 2013: insights from the Medical Expenditure Panel Survey. JAMA Cardiol. 2017;2(1):56–65. https://pubmed.ncbi.nlm.nih.gov/29358195/
Diprose W, Verster F. The preventive-pill paradox: how shared decision making could increase cardiovascular morbidity and mortality. Circulation. 2016;134(21):1599–600. https://pubmed.ncbi.nlm.nih.gov/27881503/
Ziaeian B, Fonarow GC. Statins and the prevention of heart disease. JAMA Cardiol. 2017;2(4):464. https://pubmed.ncbi.nlm.nih.gov/28122083/
ASCVD Risk Estimator Plus. American College of Cardiology. https://tools.acc.org/ASCVD-Risk-Estimator/. Accessed April 3, 2022.; https://tools.acc.org/ASCVD-Risk-Estimator/
Framingham Risk Score. Medscape. https://reference.medscape.com/calculator/framingham-cardiovascular-disease-risk. Accessed April 3, 2022.; https://reference.medscape.com/calculator/252/framingham-risk-score-2008
Reynolds Risk Score. https://www.reynoldsriskscore.org. Accessed April 3, 2022.; https://www.reynoldsriskscore.org/
Lloyd-Jones DM, Braun LT, Ndumele CE, et al. Use of risk assessment tools to guide decision-making in the primary prevention of atherosclerotic cardiovascular disease: a special report from the American Heart Association and American College of Cardiology. J Am Coll Cardiol. 2019;73(24):3153–67. https://pubmed.ncbi.nlm.nih.gov/30586766/
Lloyd-Jones DM, Braun LT, Ndumele CE, et al. Use of risk assessment tools to guide decision-making in the primary prevention of atherosclerotic cardiovascular disease: a special report from the American Heart Association and American College of Cardiology. J Am Coll Cardiol. 2019;73(24):3153–67. https://pubmed.ncbi.nlm.nih.gov/30586766/
Curry SJ, Krist AH, Owens DK, et al. Risk assessment for cardiovascular disease with nontraditional risk factors: US Preventive Services Task Force recommendation statement. JAMA. 2018;320(3):272–80. https://pubmed.ncbi.nlm.nih.gov/29998297/
Diamond DM, de Lorgeril M, Kendrick M, Ravnskov U, Rosch PJ. Formal comment on “Systematic review of the predictors of statin adherence for the primary prevention of cardiovascular disease.” PLoS One. 2019;14(1):e0205138. https://pubmed.ncbi.nlm.nih.gov/30653537/
Wei MY, Ito MK, Cohen JD, Brinton EA, Jacobson TA. Predictors of statin adherence, switching, and discontinuation in the USAGE survey: understanding the use of statins in America and gaps in patient education. J Clin Lipidol. 2013;7(5):472–83. https://pubmed.ncbi.nlm.nih.gov/24079289/
Ward NC, Watts GF, Eckel RH. Statin toxicity: mechanistic insights and clinical implications. Circ Res. 2019;124(2):328–50. https://pubmed.ncbi.nlm.nih.gov/30653440/
Zaleski AL, Taylor BA, Thompson PD. Coenzyme Q10 as treatment for statin-associated muscle symptoms – a good idea, but…. Adv Nutr. 2018;9(4):519S-23S. https://pubmed.ncbi.nlm.nih.gov/30032220/
Banach M, Serban C, Sahebkar A, et al. Effects of coenzyme Q10 on statin-induced myopathy: a meta-analysis of randomized controlled trials. Mayo Clin Proc. 2015;90(1):24–34. https://pubmed.ncbi.nlm.nih.gov/25440725/
Armour R, Zhou L. Outcomes of statin myopathy after statin withdrawal. J Clin Neuromuscul Dis. 2013;14(3):103–9. https://pubmed.ncbi.nlm.nih.gov/23492461/
Majeed A, Molokhia M. Urgent need to establish the true incidence of the side effects of statins. BMJ. 2014;348:g3650. https://pubmed.ncbi.nlm.nih.gov/24920685/
Finegold JA, Manisty CH, Goldacre B, Barron AJ, Francis DP. What proportion of symptomatic side effects in patients taking statins are genuinely caused by the drug? Systematic review of randomized placebo-controlled trials to aid individual patient choice. Eur J Prev Cardiol. 2014;21(4):464–74. https://pubmed.ncbi.nlm.nih.gov/24623264/
Climent E, Benaiges D, Pedro-Botet J. Statin treatment and increased diabetes risk. Possible mechanisms. Clínica e Investigación en Arteriosclerosis. 2019;31(5):228–32. https://pubmed.ncbi.nlm.nih.gov/30737072/
Mansi IA, English J, Zhang S, Mortensen EM, Halm EA. Long-term outcomes of short-term statin use in healthy adults: a retrospective cohort study. Drug Saf. 2016;39(6):543–59. https://pubmed.ncbi.nlm.nih.gov/26979831/
The Panel on Food Additives and Nutrient Sources, Aggett P, Aguilar F, et al. Scientific opinion on the safety of monacolins in red yeast rice. EFSA J. 2018;16(80):5368. https://pubmed.ncbi.nlm.nih.gov/32626016/
Gordon RY, Cooperman T, Obermeyer W, Becker DJ. Marked variability of monacolin levels in commercial red yeast rice products: buyer beware! Arch Intern Med. 2010;170(19):1722–7. https://pubmed.ncbi.nlm.nih.gov/20975018/
Righetti L, Dall’Asta C, Bruni R. Risk assessment of RYR food supplements: perception vs. reality. Front Nutr. 2021;8:792529. https://pubmed.ncbi.nlm.nih.gov/34950692/
Murphy SL, Kochanek KD, Xu J, Arias E. Mortality in the United States, 2020. NCHS Data Brief, No. 427. https://www.cdc.gov/nchs/products/databriefs/db427.htm. Published December 2021. Accessed January 3, 2023.; https://www.cdc.gov/nchs/products/databriefs/db427.htm
Jukema JW, Cannon CP, de Craen AJM, Westendorp RGJ, Trompet S. The controversies of statin therapy: weighing the evidence. J Am Coll Cardiol. 2012;60(10):875–81. https://pubmed.ncbi.nlm.nih.gov/22902202/
Newman CB, Preiss D, Tobert JA, et al. Statin safety and associated adverse events: a scientific statement from the American Heart Association. Arterioscler Thromb Vasc Biol. 2019;39(2):e38–81. https://pubmed.ncbi.nlm.nih.gov/30580575/
Redberg RF, Katz MH. Statins for primary prevention: the debate is intense, but the data are weak. JAMA. 2016;316(19):1979–81. https://pubmed.ncbi.nlm.nih.gov/27838702/
Ornish D, Scherwitz LW, Billings JH, et al. Intensive lifestyle changes for reversal of coronary heart disease. JAMA. 1998;280(23):2001–7. https://pubmed.ncbi.nlm.nih.gov/9863851/
Kelly J, Karlsen M, Steinke G. Type 2 diabetes remission and lifestyle medicine: a position statement from the American College of Lifestyle Medicine. Am J Lifestyle Med. 2020;14(4):406–19. https://pubmed.ncbi.nlm.nih.gov/33281521/
Esselstyn CB Jr, Gendy G, Doyle J, Golubic M, Roizen MF. A way to reverse CAD? J Fam Pract. 2014;63(7):356–64b. https://pubmed.ncbi.nlm.nih.gov/25198208/
Hochholzer W, Giugliano RP. Lipid lowering goals: back to nature? Ther Adv Cardiovasc Dis. 2010;4(3):185–91. https://pubmed.ncbi.nlm.nih.gov/20400493/
Steinberg D, Witztum JL. Inhibition of PCSK9: a powerful weapon for achieving ideal LDL cholesterol levels. Proc Natl Acad Sci U S A. 2009;106(24):9546–7. https://pubmed.ncbi.nlm.nih.gov/19506257/
Cohen J, Pertsemlidis A, Kotowski IK, Graham R, Garcia CK, Hobbs HH. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9. Nat Genet. 2005;37(2):161–5. https://pubmed.ncbi.nlm.nih.gov/15654334/
Jaworski K, Jankowski P, Kosior DA. PCSK9 inhibitors – from discovery of a single mutation to a groundbreaking therapy of lipid disorders in one decade. Arch Med Sci. 2017;13(4):914–29. https://pubmed.ncbi.nlm.nih.gov/28721159/
Qamar A, Bhatt DL. Effect of low cholesterol on steroid hormones and vitamin E levels: just a theory or real concern? Circ Res. 2015;117(8):662–4. https://pubmed.ncbi.nlm.nih.gov/26405182/
Blom DJ, Djedjos CS, Monsalvo ML, et al. Effects of evolocumab on vitamin E and steroid hormone levels: results from the 52-week, phase 3, double-blind, randomized, placebo-controlled DESCARTES study. Circ Res. 2015;117(8):731–41. https://pubmed.ncbi.nlm.nih.gov/26228031/
Qamar A, Libby P. Low-density lipoprotein cholesterol after an acute coronary syndrome: how low to go? Curr Cardiol Rep. 2019;21(8):77. https://pubmed.ncbi.nlm.nih.gov/31250329/
Hochholzer W, Giugliano RP. Lipid lowering goals: back to nature? Ther Adv Cardiovasc Dis. 2010;4(3):185–91. https://pubmed.ncbi.nlm.nih.gov/20400493/
Glueck CJ, Gartside P, Fallat RW, Sielski J, Steiner PM. Longevity syndromes: familial hypobeta and familial hyperalpha lipoproteinemia. J Lab Clin Med. 1976;88(6):941–57. https://pubmed.ncbi.nlm.nih.gov/186545/
Packard CJ. LDL cholesterol: How low to go? Trends Cardiovasc Med. 2018;28(5):348–54. https://pubmed.ncbi.nlm.nih.gov/29336946/
Gotto AM. Low-density lipoprotein cholesterol and cardiovascular risk reduction: how low is low enough without causing harm? JAMA Cardiol. 2018;3(9):802–3. https://pubmed.ncbi.nlm.nih.gov/30073330/
Packard CJ. LDL cholesterol: How low to go? Trends Cardiovasc Med. 2018;28(5):348–54. https://pubmed.ncbi.nlm.nih.gov/29336946/
Steinberg D. The cholesterol controversy is over. Why did it take so long? Circulation. 1989;80(4):1070–8. https://pubmed.ncbi.nlm.nih.gov/2676235/
Morgan DJ, Dhruva SS, Coon ER, Wright SM, Korenstein D. 2018 update on medical overuse. JAMA Intern Med. 2019;179(2):240–6. https://pubmed.ncbi.nlm.nih.gov/30508032/
Lyu H, Xu T, Brotman D, et al. Overtreatment in the United States. PLoS One. 2017;12(9):e0181970. https://pubmed.ncbi.nlm.nih.gov/28877170/
Rothberg MB, Scherer L, Kashef MA, et al. The effect of information presentation on beliefs about the benefits of elective percutaneous coronary intervention. JAMA Intern Med. 2014;174(10):1623–9. https://pubmed.ncbi.nlm.nih.gov/25156687/
Rothberg MB, Sivalingam SK, Ashraf J, et al. Summaries for patients: patients’ and cardiologists’ beliefs about a common heart procedure. Ann Intern Med. 2010;153(5):I46. https://pubmed.ncbi.nlm.nih.gov/20820040/
Laukkanen JA, Kunutsor SK, Lavie CJ. Percutaneous coronary intervention versus medical therapy in the treatment of stable coronary artery disease: an updated meta-analysis of contemporary randomized controlled trials. J Invasive Cardiol. 2021;33(8):E647–57. https://pubmed.ncbi.nlm.nih.gov/34338654/
Harvard Heart Letter. COURAGE to make choices. Harvard Health Publishing. https://www.health.harvard.edu/newsletter_article/courage-to-make-choices. Published June 1, 2007. Accessed April 5, 2022.; https://www.health.harvard.edu/newsletter_article/courage-to-make-choices
Al-Lamee R, Thompson D, Dehbi HM, et al. Percutaneous coronary intervention in stable angina (ORBITA): a double-blind, randomised controlled trial. Lancet. 2018;391(10115):31–40. https://pubmed.ncbi.nlm.nih.gov/29103656/
Kolata G. ‘Unbelievable’: heart stents fail to ease chest pain. The New York Times. https://www.nytimes.com/2017/11/02/health/heart-disease-stents.html. Published November 2, 2017. Accessed April 5, 2022.; https://www.nytimes.com/2017/11/02/health/heart-disease-stents.html
Al-Lamee R, Thompson D, Dehbi HM, et al. Percutaneous coronary intervention in stable angina (ORBITA): a double-blind, randomised controlled trial. Lancet. 2018;391(10115):31–40. https://pubmed.ncbi.nlm.nih.gov/29103656/
Doenst T, Haverich A, Serruys P, et al. PCI and CABG for treating stable coronary artery disease: JACC review topic of the week. J Am Coll Cardiol. 2019;73(8):964–76. https://pubmed.ncbi.nlm.nih.gov/30819365/
Rothberg MB. Coronary artery disease as clogged pipes: a misconceptual model. Circ Cardiovasc Qual Outcomes. 2013;6(1):129–32. https://pubmed.ncbi.nlm.nih.gov/23322809/
Trumbo PR, Shimakawa T. Tolerable upper intake levels for trans fat, saturated fat, and cholesterol. Nutr Rev. 2011;69(5):270–8. https://pubmed.ncbi.nlm.nih.gov/21521229/
World Health Organization. Countdown to 2023: WHO report on global trans-fat elimination 2021. Geneva: 2021.; https://www.who.int/publications/i/item/9789240031876
Wanders AJ, Zock PL, Brouwer IA. Trans fat intake and its dietary sources in general populations worldwide: a systematic review. Nutrients. 2017;9(8):E840. https://pubmed.ncbi.nlm.nih.gov/28783062/
Kahle L, Krebs-Smith SM, Reedy J, Rodgers AB, Signes C. Identification of top food sources of various food components. Epidemiology and Genomics Research Program. https://epi.grants.cancer.gov/diet/foodsources/top-food-sources-report-02212020.pdf. Updated November 30, 2019. Accessed April 5, 2022.; https://epi.grants.cancer.gov/diet/foodsources/top-food-sources-report-02212020.pdf
Xu Z, McClure ST, Appel LJ. Dietary cholesterol intake and sources among U.S. adults: results from National Health and Nutrition Examination Surveys (NHANES), 2001–2014. Nutrients. 2018;10(6):E771. https://pubmed.ncbi.nlm.nih.gov/29903993/
Kahle L, Krebs-Smith SM, Reedy J, Rodgers AB, Signes C. Identification of top food sources of various food components. Epidemiology and Genomics Research Program. https://epi.grants.cancer.gov/diet/foodsources/top-food-sources-report-02212020.pdf. Updated November 30, 2019. Accessed April 5, 2022.; https://epi.grants.cancer.gov/diet/foodsources/top-food-sources-report-02212020.pdf
Riccardi G, Giosuè A, Calabrese I, Vaccaro O. Dietary recommendations for prevention of atherosclerosis. Cardiovasc Res. 2022;118(5):1188–204. https://pubmed.ncbi.nlm.nih.gov/34229346/
Кокрейновская база данных систематических обзоров и метаанализов, которые обобщают и интерпретируют результаты медицинских исследований. – Примеч. ред.
Hooper L, Martin N, Abdelhamid A, Davey Smith G. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst Rev. 2015;(6):CD011737. https://pubmed.ncbi.nlm.nih.gov/26068959/
Sacks FM, Lichtenstein AH, Wu JHY, et al. Dietary fats and cardiovascular disease: a presidential advisory from the American Heart Association. Circulation. 2017;136(3):e1–30. https://pubmed.ncbi.nlm.nih.gov/28620111/
Hughes S. AHA issues ‘Presidential Advisory’ on harms of saturated fat. Medscape. https://www.medscape.com/viewarticle/881689. Published June 15, 2017. Accessed April 3, 2022.; https://www.medscape.com/viewarticle/881689
Sacks FM, Lichtenstein AH, Wu JHY, et al. Dietary fats and cardiovascular disease: a presidential advisory from the American Heart Association. Circulation. 2017;136(3):e1–30. https://pubmed.ncbi.nlm.nih.gov/28620111/
Bergeron N, Chiu S, Williams PT, M King S, Krauss RM. Effects of red meat, white meat, and nonmeat protein sources on atherogenic lipoprotein measures in the context of low compared with high saturated fat intake: a randomized controlled trial. Am J Clin Nutr. 2019;110(1):24–33. https://pubmed.ncbi.nlm.nih.gov/31161217/
Maki KC, Van Elswyk ME, Alexander DD, Rains TM, Sohn EL, McNeill S. A meta-analysis of randomized controlled trials that compare the lipid effects of beef versus poultry and/or fish consumption. J Clin Lipidol. 2012;6(4):352–61. https://pubmed.ncbi.nlm.nih.gov/22836072/
Connor WE, Connor SL. Dietary cholesterol and coronary heart disease. Curr Atheroscler Rep. 2002;4(6):425–32. https://pubmed.ncbi.nlm.nih.gov/12361489/
Khalighi Sikaroudi M, Soltani S, Kolahdouz-Mohammadi R, et al. The responses of different dosages of egg consumption on blood lipid profile: an updated systematic review and meta-analysis of randomized clinical trials. J Food Biochem. 2020;44(8):e13263. https://pubmed.ncbi.nlm.nih.gov/32524644/
Barnard ND, Long MB, Ferguson JM, Flores R, Kahleova H. Industry funding and cholesterol research: a systematic review. Am J Lifestyle Med. 2021;15(2):165–72. https://pubmed.ncbi.nlm.nih.gov/33786032/
Choi Y, Chang Y, Lee JE, et al. Egg consumption and coronary artery calcification in asymptomatic men and women. Atherosclerosis. 2015;241(2):305–12. https://pubmed.ncbi.nlm.nih.gov/26062990/
Zhong VW, Van Horn L, Cornelis MC, et al. Associations of dietary cholesterol or egg consumption with incident cardiovascular disease and mortality. JAMA. 2019;321(11):1081–95. https://pubmed.ncbi.nlm.nih.gov/30874756/
Abbasi J. Study puts eggs and dietary cholesterol back on the radar. JAMA. 2019;321(20):1959–61. https://pubmed.ncbi.nlm.nih.gov/31066864/
Physicians Comm for Responsible Med v. Vilsack, № 16-cv-00069-LB, 2016 US Dist LEXIS 141489, 2016 WL 5930585 (ND Cal 2016).; https://casetext.com/case/physicians-comm-for-responsible-med-v-vilsack-2
U.S. Department of Agriculture, U.S. Department of Health and Human Services. Dietary guidelines for Americans, 2015–2020. 8th ed. http://health.gov/dietaryguidelines/2015/guidelines/. Published December 2015. Accessed May 25, 2022; https://health.gov/dietaryguidelines/2015/guidelines/
U.S. Department of Agriculture, U.S. Department of Health and Human Services. Dietary guidelines for Americans, 2020–2025. 9th ed. https://www.dietaryguidelines.gov/sites/default/files/2020–12/Dietary_Guidelines_for_Americans_2020–2025.pdf. Published December 2020. Accessed April 5, 2022.; https://www.dietaryguidelines.gov/sites/default/files/2020-12/Dietary_Guidelines_for_Americans_2020-2025.pdf
Trumbo PR, Shimakawa T. Tolerable upper intake levels for trans fat, saturated fat, and cholesterol. Nutr Rev. 2011;69(5):270–8. https://pubmed.ncbi.nlm.nih.gov/21521229/
David Spence J. Dietary cholesterol and egg yolk should be avoided by patients at risk of vascular disease. J Transl Int Med. 2016;4(1):20–4. https://pubmed.ncbi.nlm.nih.gov/28191513/
Enas EA, Varkey B, Dharmarajan TS, Pare G, Bahl VK. Lipoprotein(a): an independent, genetic, and causal factor for cardiovascular disease and acute myocardial infarction. Indian Heart J. 2019;71(2):99–112. https://pubmed.ncbi.nlm.nih.gov/31280836/
Kotani K, Serban MC, Penson P, Lippi G, Banach M. Evidence-based assessment of lipoprotein(a) as a risk biomarker for cardiovascular diseases – some answers and still many questions. Crit Rev Clin Lab Sci. 2016;53(6):370–8. https://pubmed.ncbi.nlm.nih.gov/27173621/
Stulnig TM, Morozzi C, Reindl-Schwaighofer R, Stefanutti C. Looking at Lp(a) and related cardiovascular risk: from scientific evidence and clinical practice. Curr Atheroscler Rep. 2019;21(10):37. https://pubmed.ncbi.nlm.nih.gov/31350625/
Kostner KM, Kostner GM, Wierzbicki AS. Is Lp(a) ready for prime time use in the clinic? A pros-and-cons debate. Atherosclerosis. 2018;274:16–22. https://pubmed.ncbi.nlm.nih.gov/29747086/
Stender S. In equal amounts, the major ruminant trans fatty acid is as bad for LDL cholesterol as industrially produced trans fatty acids, but the latter are easier to remove from foods. Am J Clin Nutr. 2015;102(6):1301–2. https://pubmed.ncbi.nlm.nih.gov/26561633/
Gebauer SK, Destaillats F, Dionisi F, Krauss RM, Baer DJ. Vaccenic acid and trans fatty acid isomers from partially hydrogenated oil both adversely affect LDL cholesterol: a double-blind, randomized controlled trial. Am J Clin Nutr. 2015;102(6):1339–46. https://pubmed.ncbi.nlm.nih.gov/26561632/
Вариант вегетарианства, при котором разрешены не только продукты растительного происхождения, но также молоко и яйца. – Примеч. ред.
Masarei JR, Rouse IL, Lynch WJ, Robertson K, Vandongen R, Beilin LJ. Effects of a lacto-ovo vegetarian diet on serum concentrations of cholesterol, triglyceride, HDL–C, HDL2-C, HDL3-C, apoprotein-B, and Lp(a). Am J Clin Nutr. 1984;40(3):468–78. https://pubmed.ncbi.nlm.nih.gov/6089540/
Sahebkar A, Katsiki N, Ward N, Reiner Ž. Flaxseed supplementation reduces plasma lipoprotein(a) levels: a meta-analysis. Altern Ther Health Med. 2021;27(3):50–3. https://pubmed.ncbi.nlm.nih.gov/31634874/
Biswas TK, Chakrabarti S, Pandit S, Jana U, Dey SK. Pilot study evaluating the use of Emblica officinalis standardized fruit extract in cardio-respiratory improvement and antioxidant status of volunteers with smoking history. J Herb Med. 2014;4(4):188–94. https://www.sciencedirect.com/science/article/abs/pii/S2210803314000633
Najjar RS, Moore CE, Montgomery BD. Consumption of a defined, plant-based diet reduces lipoprotein(a), inflammation, and other atherogenic lipoproteins and particles within 4 weeks. Clin Cardiol. 2018;41(8):1062–8. https://pubmed.ncbi.nlm.nih.gov/30014498/
Berk KA, Yahya R, Verhoeven AJM, et al. Effect of diet-induced weight loss on lipoprotein(A) levels in obese individuals with and without type 2 diabetes. Diabetologia. 2017;60(6):989–97. https://pubmed.ncbi.nlm.nih.gov/28386638/
Najjar RS, Moore CE, Montgomery BD. A defined, plant-based diet utilized in an outpatient cardiovascular clinic effectively treats hypercholesterolemia and hypertension and reduces medications. Clin Cardiol. 2018;41(3):307–13. https://pubmed.ncbi.nlm.nih.gov/29575002/
Najjar RS, Moore CE, Montgomery BD. Consumption of a defined, plant-based diet reduces lipoprotein(a), inflammation, and other atherogenic lipoproteins and particles within 4 weeks. Clin Cardiol. 2018;41(8):1062–8. https://pubmed.ncbi.nlm.nih.gov/30014498/
Li H, Zeng X, Wang Y, et al. A prospective study of healthful and unhealthful plant-based diet and risk of overall and cause-specific mortality. Eur J Nutr. 2022;61(1):387–98. https://pubmed.ncbi.nlm.nih.gov/34379193/
Keaver L, Ruan M, Chen F, et al. Plant– and animal-based diet quality and mortality among US adults: a cohort study. Br J Nutr. 2021;125(12):1405–15. https://pubmed.ncbi.nlm.nih.gov/32943123/
Spiegelhalter D. Using speed of ageing and “microlives” to communicate the effects of lifetime habits and environment. BMJ. 2012;345:e8223. https://pubmed.ncbi.nlm.nih.gov/23247978/
Jafari S, Hezaveh E, Jalilpiran Y, et al. Plant-based diets and risk of disease mortality: a systematic review and meta-analysis of cohort studies. Crit Rev Food Sci Nutr. Published online May 6, 2021:1–13.; https://pubmed.ncbi.nlm.nih.gov/33951994/
Remde A, DeTurk SN, Almardini A, Steiner L, Wojda T. Plant-predominant eating patterns – how effective are they for treating obesity and related cardiometabolic health outcomes? – a systematic review. Nutr Rev. 2022;80(5):1094–104. https://pubmed.ncbi.nlm.nih.gov/34498070/
Benatar JR, Stewart RAH. Cardiometabolic risk factors in vegans; a meta-analysis of observational studies. PLoS One. 2018;13(12):e0209086. https://pubmed.ncbi.nlm.nih.gov/30571724/
Fontana L, Meyer TE, Klein S, Holloszy JO. Long-term low-calorie low-protein vegan diet and endurance exercise are associated with low cardiometabolic risk. Rejuvenation Res. 2007;10(2):225–34. https://pubmed.ncbi.nlm.nih.gov/17158430/
Chen GC, Chen PY, Su YC, et al. Vascular, cognitive, and psychomental survey on elderly recycling volunteers in Northern Taiwan. Front Neurol. 2018;9:1176. https://pubmed.ncbi.nlm.nih.gov/30687225/
McDougall J, Thomas LE, McDougall C, et al. Effects of 7 days on an ad libitum low-fat vegan diet: the McDougall Program cohort. Nutr J. 2014;13:99. https://pubmed.ncbi.nlm.nih.gov/25311617/
Bloomer RJ, Kabir MM, Canale RE, et al. Effect of a 21 day Daniel Fast on metabolic and cardiovascular disease risk factors in men and women. Lipids Health Dis. 2010;9:94. https://pubmed.ncbi.nlm.nih.gov/20815907/
Friedman SM, Barnett CH, Franki R, Pollock B, Garver B, Barnett TD. Jumpstarting health with a 15-day whole-food plant-based program. Am J Lifestyle Med. Published online April 8, 2021:155982762110063.; https://pubmed.ncbi.nlm.nih.gov/35706593/
Kapur NK, Musunuru K. Clinical efficacy and safety of statins in managing cardiovascular risk. Vasc Health Risk Manag. 2008;4(2):341–53. https://pubmed.ncbi.nlm.nih.gov/18561510/
Friedman SM, Barnett CH, Franki R, Pollock B, Garver B, Barnett TD. Jumpstarting health with a 15-day whole-food plant-based program. Am J Lifestyle Med. Published online April 8, 2021:155982762110063.; https://pubmed.ncbi.nlm.nih.gov/35706593/
Paz MA, de-La-Sierra A, Sáez M, et al. Treatment efficacy of anti-hypertensive drugs in monotherapy or combination: ATOM systematic review and meta-analysis of randomized clinical trials according to PRISMA statement. Medicine (Baltimore). 2016;95(30):e4071. https://pubmed.ncbi.nlm.nih.gov/27472680/
Lin CL, Fang TC, Gueng MK. Vascular dilatory functions of ovo-lactovegetarians compared with omnivores. Atherosclerosis. 2001;158(1):247–51. https://pubmed.ncbi.nlm.nih.gov/11500198/
Ernst E, Pietsch L, Matrai A, Eisenberg J. Blood rheology in vegetarians. Br J Nutr. 1986;56(3):555–60. https://pubmed.ncbi.nlm.nih.gov/3676231/
McCarty MF. Favorable impact of a vegan diet with exercise on hemorheology: implications for control of diabetic neuropathy. Med Hypotheses. 2002;58(6):476–86. https://pubmed.ncbi.nlm.nih.gov/12323113/
Dintenfass L. Effect of low-fat, low-protein diet on blood viscosity factors. Med J Aust. 1982;1(13):543. https://onlinelibrary.wiley.com/doi/abs/10.5694/j.1326–5377.1982.tb124177.x
Ernst E, Franz A. Blood fluidity score during vegetarian and hypocaloric diets – a pilot study. Complement Ther Med. 1995;3(2):70–1. https://www.sciencedirect.com/science/article/abs/pii/S0965229995800026?via%3Dihub
Tong TYN, Appleby PN, Bradbury KE, et al. Risks of ischaemic heart disease and stroke in meat eaters, fish eaters, and vegetarians over 18 years of follow-up: results from the prospective EPIC-Oxford study. BMJ. Published online September 4, 2019:l4897.; https://pubmed.ncbi.nlm.nih.gov/31484644/
Petermann-Rocha F, Parra-Soto S, Gray S, et al. Vegetarians, fish, poultry, and meat-eaters: who has higher risk of cardiovascular disease incidence and mortality? A prospective study from UK Biobank. Eur Heart J. 2021;42(12):1136–43. https://pubmed.ncbi.nlm.nih.gov/33313747/
Chiu THT, Chang HR, Wang LY, Chang CC, Lin MN, Lin CL. Vegetarian diet and incidence of total, ischemic, and hemorrhagic stroke in 2 cohorts in Taiwan. Neurology. 2020;94(11):e1112–21. https://pubmed.ncbi.nlm.nih.gov/32102976/
Baden MY, Shan Z, Wang F, et al. Quality of plant-based diet and risk of total, ischemic, and hemorrhagic stroke. Neurology. 2021;96(15):e1940–53. https://pubmed.ncbi.nlm.nih.gov/33692165/
Lu JW, Yu LH, Tu YK, et al. Risk of incident stroke among vegetarians compared to nonvegetarians: a systematic review and meta-analysis of prospective cohort studies. Nutrients. 2021;13(9):3019. https://pubmed.ncbi.nlm.nih.gov/34578897/
Jafari S, Hezaveh E, Jalilpiran Y, et al. Plant-based diets and risk of disease mortality: a systematic review and meta-analysis of cohort studies. Crit Rev Food Sci Nutr. Published online May 6, 2021:1–13.; https://pubmed.ncbi.nlm.nih.gov/33951994/
Mazidi M, Katsiki N, Mikhailidis DP, Sattar N, Banach M. Lower carbohydrate diets and all-cause and cause-specific mortality: a population-based cohort study and pooling of prospective studies. Eur Heart J. 2019;40(34):2870–9. https://pubmed.ncbi.nlm.nih.gov/31004146/
Schutz Y, Montani JP, Dulloo AG. Low-carbohydrate ketogenic diets in body weight control: a recurrent plaguing issue of fad diets? Obes Rev. 2021;22 Suppl 2:e13195. https://pubmed.ncbi.nlm.nih.gov/33471427/
Mazidi M, Katsiki N, Mikhailidis DP, Sattar N, Banach M. Lower carbohydrate diets and all-cause and cause-specific mortality: a population-based cohort study and pooling of prospective studies. Eur Heart J. 2019;40(34):2870–9. https://pubmed.ncbi.nlm.nih.gov/31004146/
Fleming RM. The effect of high-protein diets on coronary blood flow. Angiology. 2000;51(10):817–26. https://pubmed.ncbi.nlm.nih.gov/11108325/
Schwingshackl L, Hoffmann G. Low-carbohydrate diets impair flow-mediated dilatation: evidence from a systematic review and meta-analysis. Br J Nutr. 2013;110(5):969–70. https://pubmed.ncbi.nlm.nih.gov/23829973/
Nicholls SJ, Lundman P, Harmer JA, et al. Consumption of saturated fat impairs the anti-inflammatory properties of high-density lipoproteins and endothelial function. J Am Coll Cardiol. 2006;48(4):715–20. https://pubmed.ncbi.nlm.nih.gov/16904539/
Phillips SA, Jurva JW, Syed AQ, et al. Benefit of low-fat over low-carbohydrate diet on endothelial health in obesity. Hypertension. 2008;51(2):376–82. https://pubmed.ncbi.nlm.nih.gov/18195164/
Schwingshackl L, Hoffmann G. Low-carbohydrate diets impair flow-mediated dilatation: evidence from a systematic review and meta-analysis. Br J Nutr. 2013;110(5):969–70. https://pubmed.ncbi.nlm.nih.gov/23829973/
Mazidi M, Katsiki N, Mikhailidis DP, Sattar N, Banach M. Lower carbohydrate diets and all-cause and cause-specific mortality: a population-based cohort study and pooling of prospective studies. Eur Heart J. 2019;40(34):2870–9. https://pubmed.ncbi.nlm.nih.gov/31004146/
Young NJ, Metcalfe C, Gunnell D, et al. A cross-sectional analysis of the association between diet and insulin-like growth factor (IGF)-I, IGF-II, IGF-binding protein (IGFBP)-2, and IGFBP-3 in men in the United Kingdom. Cancer Causes Control. 2012;23(6):907–17. https://pubmed.ncbi.nlm.nih.gov/22527168/
Lee DH, Lee IK, Song K, et al. A strong dose-response relation between serum concentrations of persistent organic pollutants and diabetes: results from the National Health and Examination Survey 1999–2002. Diabetes Care. 2006;29(7):1638–44. https://pubmed.ncbi.nlm.nih.gov/16801591/
Ax E, Lampa E, Lind L, et al. Circulating levels of environmental contaminants are associated with dietary patterns in older adults. Environ Int. 2015;75:93–102. https://pubmed.ncbi.nlm.nih.gov/25461418/
Kris-Etherton PM, Harris WS, Appel LJ. Fish consumption, fish oil, omega-3 fatty acids, and cardiovascular disease. Arterioscler Thromb Vasc Biol. 2003;23(2):e20–30. https://pubmed.ncbi.nlm.nih.gov/12588785/
Shepherd CJ, Jackson AJ. Global fishmeal and fish-oil supply: inputs, outputs and markets. J Fish Biol. 2013;83(4):1046–66. https://pubmed.ncbi.nlm.nih.gov/24090562/
Abdelhamid AS, Brown TJ, Brainard JS, et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst Rev. 2018;7:CD003177. https://pubmed.ncbi.nlm.nih.gov/30019766/
de Magalhães JP, Müller M, Rainger GEd, Steegenga W. Fish oil supplements, longevity and aging. Aging (Albany NY). 2016;8(8):1578–82. https://pubmed.ncbi.nlm.nih.gov/27564420/
Bowman L, Mafham M, Wallendszus K, et al. Effects of n-3 fatty acid supplements in diabetes mellitus. N Engl J Med. 2018;379(16):1540–50. https://pubmed.ncbi.nlm.nih.gov/30146932/
Kalstad AA, Myhre PL, Laake K, et al. Effects of n-3 fatty acid supplements in elderly patients after myocardial infarction: a randomized, controlled trial. Circulation. 2021;143(6):528–39. https://pubmed.ncbi.nlm.nih.gov/33191772/
Nicholls SJ, Lincoff AM, Garcia M, et al. Effect of high-dose omega-3 fatty acids vs corn oil on major adverse cardiovascular events in patients at high cardiovascular risk: the STRENGTH randomized clinical trial. JAMA. 2020;324(22):2268–80. https://pubmed.ncbi.nlm.nih.gov/33190147/
Manson JE, Cook NR, Lee IM, et al. Marine n-3 fatty acids and prevention of cardiovascular disease and cancer. N Engl J Med. 2019;380(1):23–32. https://pubmed.ncbi.nlm.nih.gov/30415637/
Bhatt DL, Steg PG, Miller M, et al. Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N Engl J Med. 2019;380(1):11–22. https://pubmed.ncbi.nlm.nih.gov/30415628/
Park S, Lee S, Kim Y, et al. Causal effects of serum levels of n-3 or n-6 polyunsaturated fatty acids on coronary artery disease: Mendelian randomization study. Nutrients. 2021;13(5):1490. https://pubmed.ncbi.nlm.nih.gov/33924952/
Nicholls SJ, Nelson AJ. The fish-oil paradox. Curr Opin Lipidol. 2020;31(6):356–61. https://pubmed.ncbi.nlm.nih.gov/33027227/
Wennberg M, Tornevi A, Johansson I, Hörnell A, Norberg M, Bergdahl IA. Diet and lifestyle factors associated with fish consumption in men and women: a study of whether gender differences can result in gender-specific confounding. Nutr J. 2012;11:101. https://pubmed.ncbi.nlm.nih.gov/23210480/
Mariotti F. Animal and plant protein sources and cardiometabolic health. Adv Nutr. 2019;10(Suppl_4):S351–66. https://pubmed.ncbi.nlm.nih.gov/31728490/
Krittanawong C, Isath A, Hahn J, et al. Fish consumption and cardiovascular health: a systematic review. Am J Med. 2021;134(6):713–20. https://pubmed.ncbi.nlm.nih.gov/33444594/
Gardner CD, Mehta T, Bernstein A, Aronson D. Three factors that need to be addressed more consistently in nutrition studies: “Instead of what?”, “In what context?”, and “For what?.” Am J Health Promot. 2021;35(6):881–2. https://journals.sagepub.com/doi/full/10.1177/08901171211016191d
Song M, Fung TT, Hu FB, et al. Association of animal and plant protein intake with all-cause and cause-specific mortality. JAMA Intern Med. 2016;176(10):1453–63. https://pubmed.ncbi.nlm.nih.gov/27479196/
Rimm EB, Appel LJ, Chiuve SE, et al. Seafood long-chain n-3 polyunsaturated fatty acids and cardiovascular disease: a science advisory from the American Heart Association. Circulation. 2018;138(1):e35–47. https://pubmed.ncbi.nlm.nih.gov/29773586/
Guasch-Ferré M, Satija A, Blondin SA, et al. Meta-analysis of randomized controlled trials of red meat consumption in comparison with various comparison diets on cardiovascular risk factors. Circulation. 2019;139(15):1828–45. https://pubmed.ncbi.nlm.nih.gov/30958719/
Song M, Fung TT, Hu FB, et al. Association of animal and plant protein intake with all-cause and cause-specific mortality. JAMA Intern Med. 2016;176(10):1453–63. https://pubmed.ncbi.nlm.nih.gov/27479196/
Sun Y, Liu B, Snetselaar LG, et al. Association of major dietary protein sources with all-cause and cause-specific mortality: prospective cohort study. J Am Heart Assoc. 2021;10(5):e015553. https://pubmed.ncbi.nlm.nih.gov/33624505/
Song M, Fung TT, Hu FB, et al. Association of animal and plant protein intake with all-cause and cause-specific mortality. JAMA Intern Med. 2016;176(10):1453–63. https://pubmed.ncbi.nlm.nih.gov/27479196/
Esselstyn CB. Resolving the coronary artery disease epidemic through plant-based nutrition. Prev Cardiol. 2001;4(4):171–7. https://pubmed.ncbi.nlm.nih.gov/11832674/
Affairs of the Heart. Frontier profile: Bill Castelli. Scientific American Frontiers. http://www.pbs.org/saf/1104/features/castelli3.htm. Accessed February 24, 2023.; https://www.pbs.org/saf/1104/features/castelli3.htm
Keogh EV, Walsh RJ. Rate of greying of human hair. Nature. 1965;207(999):877–8. https://pubmed.ncbi.nlm.nih.gov/5885957/
Seiberg M. Age-induced hair greying – the multiple effects of oxidative stress. Int J Cosmet Sci. 2013;35(6):532–8. https://pubmed.ncbi.nlm.nih.gov/24033376/
Kumar AB, Shamim H, Nagaraju U. Premature graying of hair: review with updates. Int J Trichology. 2018;10(5):198–203. https://pubmed.ncbi.nlm.nih.gov/30607038/
Commo S, Gaillard O, Thibaut S, Thibaut S, Bernard BA. Absence of TRP-2 in melanogenic melanocytes of human hair. Pigment Cell Res. 2004;17(5):488–97. https://pubmed.ncbi.nlm.nih.gov/15357835/
Mastore M, Kohler L, Nappi AJ. Production and utilization of hydrogen peroxide associated with melanogenesis and tyrosinase-mediated oxidations of DOPA and dopamine. FEBS J. 2005;272(10):2407–15. https://pubmed.ncbi.nlm.nih.gov/15885091/
Wood JM, Decker H, Hartmann H, et al. Senile hair graying: H2O2-mediated oxidative stress affects human hair color by blunting methionine sulfoxide repair. FASEB J. 2009;23(7):2065–75. https://pubmed.ncbi.nlm.nih.gov/19237503/
Pandhi D, Khanna D. Premature graying of hair. Indian J Dermatol Venereol Leprol. 2013;79(5):641–53. https://pubmed.ncbi.nlm.nih.gov/23974581/
Tobin DJ, Paus R. Graying: gerontobiology of the hair follicle pigmentary unit. Exp Gerontol. 2001;36(1):29–54. https://pubmed.ncbi.nlm.nih.gov/11162910/
Fernandez-Flores A, Saeb-Lima M, Cassarino DS. Histopathology of aging of the hair follicle. J Cutan Pathol. 2019;46(7):508–19. https://pubmed.ncbi.nlm.nih.gov/30932205/
Mahendiratta S, Sarma P, Kaur H, et al. Premature graying of hair: risk factors, co-morbid conditions, pharmacotherapy and reversal – a systematic review and meta-analysis. Dermatol Ther. 2020;33(6):e13990. https://pubmed.ncbi.nlm.nih.gov/32654282/
Daulatabad D, Singal A, Grover C, Sharma SB, Chhillar N. Assessment of oxidative stress in patients with premature canities. Int J Trichology. 2015;7(3):91–4. https://pubmed.ncbi.nlm.nih.gov/26622150/
Babadjouni A, Foulad DP, Hedayati B, Evron E, Mesinkovska N. The effects of smoking on hair health: a systematic review. Skin Appendage Disord. 2021;7(4):251–64. https://pubmed.ncbi.nlm.nih.gov/34307472/
Kumar AB, Shamim H, Nagaraju U. Premature graying of hair: review with updates. Int J Trichology. 2018;10(5):198–203. https://pubmed.ncbi.nlm.nih.gov/30607038/
Acer E, Kaya Erdogan H, Igrek A, Parlak H, Saraçoglu ZN, Bilgin M. Relationship between diet, atopy, family history, and premature hair graying. J Cosmet Dermatol. 2019;18(2):665–70. https://pubmed.ncbi.nlm.nih.gov/30556257/
Addolorato G, Leggio L, Ojetti V, Capristo E, Gasbarrini G, Gasbarrini A. Effects of short-term moderate alcohol administration on oxidative stress and nutritional status in healthy males. Appetite. 2008;50(1):50–6. https://pubmed.ncbi.nlm.nih.gov/17602789/
Mahendiratta S, Sarma P, Kaur H, et al. Premature graying of hair: risk factors, co-morbid conditions, pharmacotherapy and reversal – a systematic review and meta-analysis. Dermatol Ther. 2020;33(6):e13990. https://pubmed.ncbi.nlm.nih.gov/32654282/
Acer E, Kaya Erdogan H, Igrek A, Parlak H, Saraçoglu ZN, Bilgin M. Relationship between diet, atopy, family history, and premature hair graying. J Cosmet Dermatol. 2019;18(2):665–70. https://pubmed.ncbi.nlm.nih.gov/30556257/
Kumar AB, Shamim H, Nagaraju U. Premature graying of hair: review with updates. Int J Trichology. 2018;10(5):198–203. https://pubmed.ncbi.nlm.nih.gov/30607038/
Noppakun N, Swasdikul D. Reversible hyperpigmentation of skin and nails with white hair due to vitamin B12 deficiency. Arch Dermatol. 1986;122(8):896–9. https://pubmed.ncbi.nlm.nih.gov/3740873/
Kumar AB, Shamim H, Nagaraju U. Premature graying of hair: review with updates. Int J Trichology. 2018;10(5):198–203. https://pubmed.ncbi.nlm.nih.gov/30607038/
Tobin DJ, Paus R. Graying: gerontobiology of the hair follicle pigmentary unit. Exp Gerontol. 2001;36(1):29–54. https://pubmed.ncbi.nlm.nih.gov/11162910/
Tai SY, Hsieh HM, Huang SP, Wu MT. Hair dye use, regular exercise, and the risk and prognosis of prostate cancer: multicenter case-control and case-only studies. BMC Cancer. 2016;16:242. https://pubmed.ncbi.nlm.nih.gov/26996776/
Towle KM, Grespin ME, Monnot AD. Personal use of hair dyes and risk of leukemia: a systematic literature review and meta-analysis. Cancer Med. 2017;6(10):2471–86. https://pubmed.ncbi.nlm.nih.gov/28925101/
Odutola MK, Nnakelu E, Giles GG, van Leeuwen MT, Vajdic CM. Lifestyle and risk of follicular lymphoma: a systematic review and meta-analysis of observational studies. Cancer Causes Control. 2020;31(11):979–1000. https://pubmed.ncbi.nlm.nih.gov/32851495/
Tai SY, Hsieh HM, Huang SP, Wu MT. Hair dye use, regular exercise, and the risk and prognosis of prostate cancer: multicenter case-control and case-only studies. BMC Cancer. 2016;16:242. https://pubmed.ncbi.nlm.nih.gov/26996776/
Takkouche B, Regueira-Méndez C, Montes-Martínez A. Risk of cancer among hairdressers and related workers: a meta-analysis. Int J Epidemiol. 2009;38(6):1512–31. https://pubmed.ncbi.nlm.nih.gov/19755396/
Qin L, Deng HY, Chen SJ, Wei W. A meta-analysis on the relationship between hair dye and the incidence of non-Hodgkin’s lymphoma. Med Princ Pract. 2019;28(3):222–30. https://pubmed.ncbi.nlm.nih.gov/30583293/
Park AM, Khan S, Rawnsley J. Hair biology: growth and pigmentation. Facial Plast Surg Clin North Am. 2018;26(4):415–24. https://pubmed.ncbi.nlm.nih.gov/30213423/
Williams R, Pawlus AD, Thornton MJ. Getting under the skin of hair aging: the impact of the hair follicle environment. Exp Dermatol. 2020;29(7):588–97. https://pubmed.ncbi.nlm.nih.gov/32358903/
Sadick NS, Callender VD, Kircik LH, Kogan S. New insight into the pathophysiology of hair loss trigger a paradigm shift in the treatment approach. J Drugs Dermatol. 2017;16(11):s135–40. https://pubmed.ncbi.nlm.nih.gov/29141068/
English RS Jr. A hypothetical pathogenesis model for androgenic alopecia: clarifying the dihydrotestosterone paradox and rate-limiting recovery factors. Med Hypotheses. 2018;111:73–81. https://pubmed.ncbi.nlm.nih.gov/29407002/
Carmina E, Azziz R, Bergfeld W, et al. Female pattern hair loss and androgen excess: a report from the multidisciplinary Androgen Excess and PCOS committee. J Clin Endocrinol Metab. 2019;104(7):2875–91. https://pubmed.ncbi.nlm.nih.gov/30785992/
Varothai S, Bergfeld WF. Androgenetic alopecia: an evidence-based treatment update. Am J Clin Dermatol. 2014;15(3):217–30. https://pubmed.ncbi.nlm.nih.gov/24848508/
Grymowicz M, Rudnicka E, Podfigurna A, et al. Hormonal effects on hair follicles. Int J Mol Sci. 2020;21(15):E5342. https://pubmed.ncbi.nlm.nih.gov/32731328/
Tai T, Kochhar A. Physiology and medical treatments for alopecia. Facial Plast Surg Clin North Am. 2020;28(2):149–59. https://pubmed.ncbi.nlm.nih.gov/32312501/
Hibberts NA, Howell AE, Randall VA. Balding hair follicle dermal papilla cells contain higher levels of androgen receptors than those from non-balding scalp. J Endocrinol. 1998;156(1):59–65. https://pubmed.ncbi.nlm.nih.gov/9496234/
Grymowicz M, Rudnicka E, Podfigurna A, et al. Hormonal effects on hair follicles. Int J Mol Sci. 2020;21(15):E5342. https://pubmed.ncbi.nlm.nih.gov/32731328/
Campo D, D’Acunzo V. Doctors and baldness: a five thousand year old challenge. G Ital Dermatol Venereol. 2016;151(1):93–101. https://pubmed.ncbi.nlm.nih.gov/25387848/
English RS Jr. A hypothetical pathogenesis model for androgenic alopecia: clarifying the dihydrotestosterone paradox and rate-limiting recovery factors. Med Hypotheses. 2018;111:73–81. https://pubmed.ncbi.nlm.nih.gov/29407002/
Campo D, D’Acunzo V. Doctors and baldness: a five thousand year old challenge. G Ital Dermatol Venereol. 2016;151(1):93–101. https://pubmed.ncbi.nlm.nih.gov/25387848/
Hamilton JB. Effect of castration in adolescent and young adult males upon further changes in the proportions of bare and hairy scalp. J Clin Endocrinol Metab. 1960;20:1309–18. https://pubmed.ncbi.nlm.nih.gov/13711016/
Collins DT. Children of sorrow: a history of the mentally retarded in Kansas. Bull Hist Med. 1965;39:53–78. https://pubmed.ncbi.nlm.nih.gov/14284409/
Kempton W, Kahn E. Sexuality and people with intellectual disabilities: a historical perspective. Sex Disabil. 1991;9(2):93–111. https://link.springer.com/article/10.1007/BF01101735
Flood E. Notes on the castration of idiot children. Am J Psychol. 1899;10(2):296–301. https://www.jstor.org/stable/1412485?origin=crossref
Lombardo PA. Preface & acknowledgments. In: Lombardo PA, ed. A Century of Eugenics in America: From the Indiana Experiment to the Human Genome Era. Indiana University Press; 2011:ix. https://worldcat.org/title/703156879
Scott ES. Sterilization of mentally retarded persons: reproductive rights and family privacy. Duke Law J. 1986;1986(5):806–65. https://pubmed.ncbi.nlm.nih.gov/11658848/
Wittmann E. To what extent were ideas and beliefs about eugenics held in Nazi Germany shared in Britain and the United States prior to the Second World War? Vesalius. 2004;10(1):16–9. https://pubmed.ncbi.nlm.nih.gov/15386878/
Bolland MJ, Ames RW, Grey AB, et al. Does degree of baldness influence vitamin D status? Med J Aust. 2008;189(11–12):674–5. https://pubmed.ncbi.nlm.nih.gov/19061473/
Trieu N, Eslick GD. Alopecia and its association with coronary heart disease and cardiovascular risk factors: a meta-analysis. Int J Cardiol. 2014;176(3):687–95. https://pubmed.ncbi.nlm.nih.gov/25150481/
Sinclair RD, English DR, Giles GG. Are bald men more virile than their well thatched contemporaries? Med J Aust. 2013;199(11):811–2. https://pubmed.ncbi.nlm.nih.gov/24329675/
Jin T, Wu T, Luo Z, Duan X, Deng S, Tang Y. Association between male pattern baldness and prostate disease: a meta-analysis. Urol Oncol. 2018;36(2):80.e7–15. https://pubmed.ncbi.nlm.nih.gov/29054497/
Mohammadi-Shemirani P, Chong M, Pigeyre M, Morton RW, Gerstein HC, Paré G. Effects of lifelong testosterone exposure on health and disease using Mendelian randomization. Elife. 2020;9:e58914. https://pubmed.ncbi.nlm.nih.gov/33063668/
Ata Korkmaz HA. Relationship between androgenic alopecia and white matter hyperintensities in apparently healthy subjects. Brain Imaging Behav. 2020;14(2):527–33. https://pubmed.ncbi.nlm.nih.gov/31250269/
Trieu N, Eslick GD. Alopecia and its association with coronary heart disease and cardiovascular risk factors: a meta-analysis. Int J Cardiol. 2014;176(3):687–95. https://pubmed.ncbi.nlm.nih.gov/25150481/
Bertoli MJ, Sadoughifar R, Schwartz RA, Lotti TM, Janniger CK. Female pattern hair loss: a comprehensive review. Dermatol Ther. 2020;33(6):e14055. https://pubmed.ncbi.nlm.nih.gov/32700775/
Bertoli MJ, Sadoughifar R, Schwartz RA, Lotti TM, Janniger CK. Female pattern hair loss: a comprehensive review. Dermatol Ther. 2020;33(6):e14055. https://pubmed.ncbi.nlm.nih.gov/32700775/
Lin RL, Garibyan L, Kimball AB, Drake LA. Systemic causes of hair loss. Ann Med. 2016;48(6):393–402. https://pubmed.ncbi.nlm.nih.gov/27145919/
van Zuuren EJ, Fedorowicz Z, Schoones J. Interventions for female pattern hair loss. Cochrane Database Syst Rev. 2016;(5):CD007628. https://pubmed.ncbi.nlm.nih.gov/27225981/
Lam SM. Hair loss and hair restoration in women. Facial Plast Surg Clin North Am. 2020;28(2):205–23. https://pubmed.ncbi.nlm.nih.gov/32312508/
Lam SM. Hair loss and hair restoration in women. Facial Plast Surg Clin North Am. 2020;28(2):205–23. https://pubmed.ncbi.nlm.nih.gov/32312508/
Lin RL, Garibyan L, Kimball AB, Drake LA. Systemic causes of hair loss. Ann Med. 2016;48(6):393–402. https://pubmed.ncbi.nlm.nih.gov/27145919/
Bauer M, Glenn T, Pilhatsch M, Pfennig A, Whybrow PC. Gender differences in thyroid system function: relevance to bipolar disorder and its treatment. Bipolar Disord. 2014;16(1):58–71. https://pubmed.ncbi.nlm.nih.gov/24245529/
Lin RL, Garibyan L, Kimball AB, Drake LA. Systemic causes of hair loss. Ann Med. 2016;48(6):393–402. https://pubmed.ncbi.nlm.nih.gov/27145919/
Williams R, Pawlus AD, Thornton MJ. Getting under the skin of hair aging: the impact of the hair follicle environment. Exp Dermatol. 2020;29(7):588–97. https://pubmed.ncbi.nlm.nih.gov/32358903/
Chien Yin GO, Siong-See JL, Wang ECE. Telogen Effluvium – a review of the science and current obstacles. J Dermatol Sci. 2021;101(3):156–63. https://pubmed.ncbi.nlm.nih.gov/33541773/
Sharquie KE, Jabbar RI. COVID-19 infection is a major cause of acute telogen effluvium. Ir J Med Sci. Published online August 31, 2021.; https://pubmed.ncbi.nlm.nih.gov/34467470/
Malkud S. Telogen effluvium: a review. J Clin Diagn Res. 2015;9(9):WE01–3. https://pubmed.ncbi.nlm.nih.gov/26500992/
Bertoli MJ, Sadoughifar R, Schwartz RA, Lotti TM, Janniger CK. Female pattern hair loss: a comprehensive review. Dermatol Ther. 2020;33(6):e14055. https://pubmed.ncbi.nlm.nih.gov/32700775/
Malkud S. Telogen effluvium: a review. J Clin Diagn Res. 2015;9(9):WE01–3. https://pubmed.ncbi.nlm.nih.gov/26500992/
Sharquie KE, Jabbar RI. COVID-19 infection is a major cause of acute telogen effluvium. Ir J Med Sci. Published online August 31, 2021.; https://pubmed.ncbi.nlm.nih.gov/34467470/
Trieu N, Eslick GD. Alopecia and its association with coronary heart disease and cardiovascular risk factors: a meta-analysis. Int J Cardiol. 2014;176(3):687–95. https://pubmed.ncbi.nlm.nih.gov/25150481/
Gatherwright J, Liu MT, Amirlak B, Gliniak C, Totonchi A, Guyuron B. The contribution of endogenous and exogenous factors to male alopecia: a study of identical twins. Plast Reconstr Surg. 2013;131(5):794e-801e. https://pubmed.ncbi.nlm.nih.gov/23629119/
Lolli F, Pallotti F, Rossi A, et al. Androgenetic alopecia: a review. Endocrine. 2017;57(1):9–17. https://pubmed.ncbi.nlm.nih.gov/28349362/
DiMarco G, McMichael A. Hair loss myths. J Drugs Dermatol. 2017;16(7):690–4. https://pubmed.ncbi.nlm.nih.gov/28697221/
Gatherwright J, Liu MT, Gliniak C, Totonchi A, Guyuron B. The contribution of endogenous and exogenous factors to female alopecia: a study of identical twins. Plast Reconstr Surg. 2012;130(6):1219–26. https://pubmed.ncbi.nlm.nih.gov/22878477/
Gatherwright J, Liu MT, Amirlak B, Gliniak C, Totonchi A, Guyuron B. The contribution of endogenous and exogenous factors to male alopecia: a study of identical twins. Plast Reconstr Surg. 2013;131(5):794e-801e. https://pubmed.ncbi.nlm.nih.gov/23629119/
Gatherwright J, Liu MT, Gliniak C, Totonchi A, Guyuron B. The contribution of endogenous and exogenous factors to female alopecia: a study of identical twins. Plast Reconstr Surg. 2012;130(6):1219–26. https://pubmed.ncbi.nlm.nih.gov/22878477/
D’Andrea S, Spaggiari G, Barbonetti A, Santi D. Endogenous transient doping: physical exercise acutely increases testosterone levels – results from a meta-analysis. J Endocrinol Invest. 2020;43(10):1349–71. https://pubmed.ncbi.nlm.nih.gov/32297287/
Wedick NM, Mantzoros CS, Ding EL, et al. The effects of caffeinated and decaffeinated coffee on sex hormone – binding globulin and endogenous sex hormone levels: a randomized controlled trial. Nutr J. 2012;11:86. https://pubmed.ncbi.nlm.nih.gov/23078574/
Gatherwright J, Liu MT, Amirlak B, Gliniak C, Totonchi A, Guyuron B. The contribution of endogenous and exogenous factors to male alopecia: a study of identical twins. Plast Reconstr Surg. 2013;131(5):794e–801e. https://pubmed.ncbi.nlm.nih.gov/23629119/
Gatherwright J, Liu MT, Gliniak C, Totonchi A, Guyuron B. The contribution of endogenous and exogenous factors to female alopecia: a study of identical twins. Plast Reconstr Surg. 2012;130(6):1219–26. https://pubmed.ncbi.nlm.nih.gov/22878477/
Babadjouni A, Foulad DP, Hedayati B, Evron E, Mesinkovska N. The effects of smoking on hair health: a systematic review. Skin Appendage Disord. 2021;7(4):251–64. https://pubmed.ncbi.nlm.nih.gov/34307472/
Lai CH, Chu NF, Chang CW, et al. Androgenic alopecia is associated with less dietary soy, lower [corrected] blood vanadium and rs1160312 1 polymorphism in Taiwanese communities. PLoS One. 2013;8(12):e79789. https://pubmed.ncbi.nlm.nih.gov/24386074/
Yu V, Juhász M, Chiang A, Atanaskova Mesinkovska N. Alopecia and associated toxic agents: a systematic review. Skin Appendage Disord. 2018;4(4):245–60. https://pubmed.ncbi.nlm.nih.gov/30410891/
Clarkson TW. The three modern faces of mercury. Environ Health Perspect. 2002;110(Suppl 1):11–23. https://pubmed.ncbi.nlm.nih.gov/11834460/
Ross JJ. Shakespeare’s chancre: did the bard have syphilis? Clin Infect Dis. 2005;40(3):399–404. https://pubmed.ncbi.nlm.nih.gov/15668863/
Centers for Disease Control and Prevention. Executive summary. Fourth National Report on Human Exposure to Environmental Chemicals. 2009.; https://cfpub.epa.gov/ncea/risk/hhra/recordisplay.cfm?deid=23995
Peters JB, Warren MP. Reversible alopecia associated with high blood mercury levels and early menopause: a report of two cases. Menopause. 2019;26(8):915–8. https://pubmed.ncbi.nlm.nih.gov/30939539/
Campo D, D’Acunzo V. Doctors and baldness: a five thousand year old challenge. G Ital Dermatol Venereol. 2016;151(1):93–101. https://pubmed.ncbi.nlm.nih.gov/25387848/
Nanda S, De Bedout V, Miteva M. Alopecia as a systemic disease. Clin Dermatol. 2019;37(6):618–28. https://pubmed.ncbi.nlm.nih.gov/31864440/
Park AM, Khan S, Rawnsley J. Hair biology: growth and pigmentation. Facial Plast Surg Clin North Am. 2018;26(4):415–24. https://pubmed.ncbi.nlm.nih.gov/30213423/
Sadick NS, Callender VD, Kircik LH, Kogan S. New insight into the pathophysiology of hair loss trigger a paradigm shift in the treatment approach. J Drugs Dermatol. 2017;16(11):s135–40. https://pubmed.ncbi.nlm.nih.gov/29141068/
Sand JP. Follicular unit transplantation. Facial Plast Surg Clin North Am. 2020;28(2):161–7. https://pubmed.ncbi.nlm.nih.gov/32312502/
Stoneburner J, Shauly O, Carey J, Patel KM, Stevens WG, Gould DJ. Contemporary management of alopecia: a systematic review and meta-analysis for surgeons. Aesthetic Plast Surg. 2020;44(1):97–113. https://pubmed.ncbi.nlm.nih.gov/31667549/
Vogel JE. Hair restoration complications: an approach to the unnatural-appearing hair transplant. Facial Plast Surg. 2008;24(4):453–61. https://pubmed.ncbi.nlm.nih.gov/19034821/
Rose PT. Advances in hair restoration. Dermatol Clin. 2018;36(1):57–62. https://pubmed.ncbi.nlm.nih.gov/29108547/
Umar S. Hair transplantation in patients with inadequate head donor supply using nonhead hair: report of 3 cases. Ann Plast Surg. 2011;67(4):332–5. https://pubmed.ncbi.nlm.nih.gov/21540728/
Stoneburner J, Shauly O, Carey J, Patel KM, Stevens WG, Gould DJ. Contemporary management of alopecia: a systematic review and meta-analysis for surgeons. Aesthetic Plast Surg. 2020;44(1):97–113. https://pubmed.ncbi.nlm.nih.gov/31667549/
Gatherwright J, Liu MT, Gliniak C, Totonchi A, Guyuron B. The contribution of endogenous and exogenous factors to female alopecia: a study of identical twins. Plast Reconstr Surg. 2012;130(6):1219–26. https://pubmed.ncbi.nlm.nih.gov/22878477/
Nadimi S. Complications with hair transplantation. Facial Plast Surg Clin North Am. 2020;28(2):225–35. https://pubmed.ncbi.nlm.nih.gov/32312509/
Gupta AK, Mays RR, Dotzert MS, Versteeg SG, Shear NH, Piguet V. Efficacy of non-surgical treatments for androgenetic alopecia: a systematic review and network meta-analysis. J Eur Acad Dermatol Venereol. 2018;32(12):2112–25. https://pubmed.ncbi.nlm.nih.gov/29797431/
Lotti T, Goren A, Verner I, D’Alessio PA, Franca K. Platelet rich plasma in androgenetic alopecia: a systematic review. Dermatol Ther. 2019;32(3):e12837. https://pubmed.ncbi.nlm.nih.gov/30667146/
Carloni R, Pechevy L, Postel F, Zielinski M, Gandolfi S. Is there a therapeutic effect of botulinum toxin on scalp alopecia? Physiopathology and reported cases: a systematic review of the literature. J Plast Reconstr Aesthet Surg. 2020;73(12):2210–6. https://pubmed.ncbi.nlm.nih.gov/32536461/
Wang Y, Zhang H, Zheng Q, Tang K, Fang R, Sun Q. Botulinum toxin as a double-edged sword in alopecia: a systematic review. J Cosmet Dermatol. 2020;19(10):2560–5. https://pubmed.ncbi.nlm.nih.gov/32745302/
Dodd EM, Winter MA, Hordinsky MK, Sadick NS, Farah RS. Photobiomodulation therapy for androgenetic alopecia: a clinician’s guide to home-use devices cleared by the Federal Drug Administration. J Cosmet Laser Ther. 2018;20(3):159–67. https://pubmed.ncbi.nlm.nih.gov/29020478/
Simunovic Z, Trobonjaca T, Trobonjaca Z. Treatment of medial and lateral epicondylitis – tennis and golfer’s elbow – with low level laser therapy: a multicenter double blind, placebo-controlled clinical study on 324 patients. J Clin Laser Med Surg. 1998;16(3):145–51. https://pubmed.ncbi.nlm.nih.gov/9743652/
Cohen PR. A case report of scrotal rejuvenation: laser treatment of angiokeratomas of the scrotum. Dermatol Ther (Heidelb). 2019;9(1):185–92. https://pubmed.ncbi.nlm.nih.gov/30478818/
Egger A, Resnik SR, Aickara D, et al. Examining the safety and efficacy of low-level laser therapy for male and female pattern hair loss: a review of the literature. Skin Appendage Disord. 2020;6(5):259–67. https://pubmed.ncbi.nlm.nih.gov/33088809/
Egger A, Resnik SR, Aickara D, et al. Examining the safety and efficacy of low-level laser therapy for male and female pattern hair loss: a review of the literature. Skin Appendage Disord. 2020;6(5):259–67. https://pubmed.ncbi.nlm.nih.gov/33088809/
Egger A, Resnik SR, Aickara D, et al. Examining the safety and efficacy of low-level laser therapy for male and female pattern hair loss: a review of the literature. Skin Appendage Disord. 2020;6(5):259–67. https://pubmed.ncbi.nlm.nih.gov/33088809/
Ledoux S, Flamant M, Calabrese D, Bogard C, Sami O, Coupaye M. What are the micronutrient deficiencies responsible for the most common nutritional symptoms after bariatric surgery? Obes Surg. 2020;30(5):1891–7. https://pubmed.ncbi.nlm.nih.gov/31960214/
DiMarco G, McMichael A. Hair loss myths. J Drugs Dermatol. 2017;16(7):690–4. https://pubmed.ncbi.nlm.nih.gov/28697221/
Thompson KG, Kim N. Dietary supplements in dermatology: a review of the evidence for zinc, biotin, vitamin D, nicotinamide, and Polypodium. J Am Acad Dermatol. 2021;84(4):1042–50. https://pubmed.ncbi.nlm.nih.gov/32360756/
Patel DP, Swink SM, Castelo-Soccio L. A review of the use of biotin for hair loss. Skin Appendage Disord. 2017;3(3):166–9. https://pubmed.ncbi.nlm.nih.gov/28879195/
FDA in brief: FDA reminds patients, health care professionals and laboratory personnel about the potential for biotin interference with certain test results, especially specific tests to aid in heart attack diagnoses. U.S. Food and Drug Administration. https://www.fda.gov/news-events/fda-brief/fda-brief-fda-reminds-patients-health-care-professionals-and-laboratory-personnel-about-potential#:~:text=Today%2C%20the%20U.S.%20Food%20and,and%20cause%20incorrect%20results%20that. Published November 5, 2019. Accessed July 2, 2022.; https://www.fda.gov/news-events/fda-brief/fda-brief-fda-reminds-patients-health-care-professionals-and-laboratory-personnel-about-potential
MacFarquhar JK, Broussard DL, Melstrom P, et al. Acute selenium toxicity associated with a dietary supplement. Arch Intern Med. 2010;170(3):256–61. https://pubmed.ncbi.nlm.nih.gov/20142570/
Almohanna HM, Ahmed AA, Tsatalis JP, Tosti A. The role of vitamins and minerals in hair loss: a review. Dermatol Ther (Heidelb). 2018;9(1):51–70. https://pubmed.ncbi.nlm.nih.gov/30547302/
Guo EL, Katta R. Diet and hair loss: effects of nutrient deficiency and supplement use. Dermatol Pract Concept. 2017;7(1):1–10. https://pubmed.ncbi.nlm.nih.gov/28243487/
DiMarco G, McMichael A. Hair loss myths. J Drugs Dermatol. 2017;16(7):690–4. https://pubmed.ncbi.nlm.nih.gov/28697221/
Bater K, Rieder E. Over-the-counter hair loss treatments: help or hype? J Drugs Dermatol. 2018;17(12):1317–21. https://pubmed.ncbi.nlm.nih.gov/30586264/
Yi Y, Qiu J, Jia J, et al. Severity of androgenetic alopecia associated with poor sleeping habits and carnivorous eating and junk food consumption – a web-based investigation of male pattern hair loss in China. Dermatol Ther. 2020;33(2):e13273. https://pubmed.ncbi.nlm.nih.gov/32061036/
Fortes C, Mastroeni S, Mannooranparampil T, Abeni D, Panebianco A. Mediterranean diet: fresh herbs and fresh vegetables decrease the risk of Androgenetic Alopecia in males. Arch Dermatol Res. 2018;310(1):71–6. https://pubmed.ncbi.nlm.nih.gov/29181579/
Lai CH, Chu NF, Chang CW, et al. Androgenic alopecia is associated with less dietary soy, lower [corrected] blood vanadium and rs1160312 1 polymorphism in Taiwanese communities. PLoS One. 2013;8(12):e79789. https://pubmed.ncbi.nlm.nih.gov/24386074/
Daniels G, Akram S, Westgate GE, Tamburic S. Can plant-derived phytochemicals provide symptom relief for hair loss? A critical review. Int J Cosmet Sci. 2019;41(4):332–45. https://pubmed.ncbi.nlm.nih.gov/31240739/
Hosking AM, Juhasz M, Atanaskova Mesinkovska N. Complementary and alternative treatments for alopecia: a comprehensive review. Skin Appendage Disord. 2019;5(2):72–89. https://pubmed.ncbi.nlm.nih.gov/30815439/
Herman A, Herman AP. Topically used herbal products for the treatment of hair loss: preclinical and clinical studies. Arch Dermatol Res. 2017;309(8):595–610. https://pubmed.ncbi.nlm.nih.gov/28695329/
Grothe T, Wandrey F, Schuerch C. Short communication: clinical evaluation of pea sprout extract in the treatment of hair loss. Phytother Res. 2020;34(2):428–31. https://pubmed.ncbi.nlm.nih.gov/31680356/
Harada N, Okajima K, Arai M, Kurihara H, Nakagata N. Administration of capsaicin and isoflavone promotes hair growth by increasing insulin-like growth factor-I production in mice and in humans with alopecia. Growth Horm IGF Res. 2007;17(5):408–15. https://pubmed.ncbi.nlm.nih.gov/17569567/
Troconis-Torres IG, Rojas-López M, Hernández-Rodríguez C, et al. Biochemical and molecular analysis of some commercial samples of chilli peppers from Mexico. J Biomed Biotech. 2012;2012:1–11. https://pubmed.ncbi.nlm.nih.gov/22665993/
Cho H, Kwon Y. Development of a database of capsaicinoid contents in foods commonly consumed in Korea. Food Sci Nutr. 2020;8(8):4611–24. https://pubmed.ncbi.nlm.nih.gov/32884741/
Harada N, Okajima K, Arai M, Kurihara H, Nakagata N. Administration of capsaicin and isoflavone promotes hair growth by increasing insulin-like growth factor-I production in mice and in humans with alopecia. Growth Horm IGF Res. 2007;17(5):408–15. https://pubmed.ncbi.nlm.nih.gov/17569567/
Bhagwat S, Haytowitz DB, Holden JM. USDA database for the isoflavone content of selected foods: release 2.0. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/arsuserfiles/80400525/data/isoflav/isoflav_r2.pdf. Published September 2008. Accessed April 15, 2022.; https://www.ars.usda.gov/arsuserfiles/80400525/data/isoflav/isoflav_r2.pdf
Cho YH, Lee SY, Jeong DW, et al. Effect of pumpkin seed oil on hair growth in men with androgenetic alopecia: a randomized, double-blind, placebo-controlled trial. Evid Based Complement Alternat Med. 2014;2014:549721. https://pubmed.ncbi.nlm.nih.gov/24864154/
Octa Sabal Plus. tradeKorea.com. https://www.tradekorea.com/product/detail/P291943/Octa-Sabal-Plus-.html. Accessed July 5, 2022.; https://www.tradekorea.com/product/detail/P291943/Octa-Sabal-Plus-.html
Hajhashemi V, Rajabi P, Mardani M. Beneficial effects of pumpkin seed oil as a topical hair growth promoting agent in a mice model. Avicenna J Phytomed. 2019;9(6):499–504. https://pubmed.ncbi.nlm.nih.gov/31763209/
Ibrahim IM, Hasan MS, Elsabaa KI, Elsaie ML. Pumpkin seed oil vs. minoxidil 5 % topical foam for the treatment of female pattern hair loss: a randomized comparative trial. J Cosmet Dermatol. 2021;20(9):2867–73. https://pubmed.ncbi.nlm.nih.gov/33544448/
Dhurat R, Chitallia J, May TW, et al. An open-label randomized multicenter study assessing the noninferiority of a caffeine-based topical liquid 0. 2 % versus minoxidil 5 % solution in male androgenetic alopecia. Skin Pharmacol Physiol. 2017;30(6):298–305. https://pubmed.ncbi.nlm.nih.gov/29055953/
Daniels G, Akram S, Westgate GE, Tamburic S. Can plant-derived phytochemicals provide symptom relief for hair loss? A critical review. Int J Cosmet Sci. 2019;41(4):332–45. https://pubmed.ncbi.nlm.nih.gov/31240739/
Randall VA, Ebling FJ. Seasonal changes in human hair growth. Br J Dermatol. 1991;124(2):146–51. https://pubmed.ncbi.nlm.nih.gov/2003996/
Fischer TW, Herczeg-Lisztes E, Funk W, Zillikens D, Bíró T, Paus R. Differential effects of caffeine on hair shaft elongation, matrix and outer root sheath keratinocyte proliferation, and transforming growth factor-ß2/insulin-like growth factor-1-mediated regulation of the hair cycle in male and female human hair follicles in vitro. Br J Dermatol. 2014;171(5):1031–43. https://pubmed.ncbi.nlm.nih.gov/24836650/
Dressler C, Blumeyer A, Rosumeck S, Arayesh A, Nast A. Efficacy of topical caffeine in male androgenetic alopecia. J Dtsch Dermatol Ges. 2017;15(7):734–41. https://pubmed.ncbi.nlm.nih.gov/28677188/
Bussoletti C, Tolaini MV, Celleno L. Efficacy of a cosmetic phyto-caffeine shampoo in female androgenetic alopecia. G Ital Dermatol Venereol. 2020;155(4):492–9. https://pubmed.ncbi.nlm.nih.gov/29512972/
Dressler C, Blumeyer A, Rosumeck S, Arayesh A, Nast A. Efficacy of topical caffeine in male androgenetic alopecia. J Dtsch Dermatol Ges. 2017;15(7):734–41. https://pubmed.ncbi.nlm.nih.gov/28677188/
Kwon OS, Han JH, Yoo HG, et al. Human hair growth enhancement in vitro by green tea epigallocatechin-3-gallate (EGCG). Phytomedicine. 2007;14(7–8):551–5. https://pubmed.ncbi.nlm.nih.gov/17092697/
Liao S, Hiipakka RA. Selective inhibition of steroid 5 a-reductase isozymes by tea epicatechin-3-gallate and epigallocatechin-3-gallate. Biochem Biophys Res Commun. 1995;214(3):833–8. https://pubmed.ncbi.nlm.nih.gov/7575552/
Kim YY, Up No S, Kim MH, et al. Effects of topical application of EGCG on testosterone-induced hair loss in a mouse model. Exp Dermatol. 2011;20(12):1015–7. https://pubmed.ncbi.nlm.nih.gov/21951062/
Berger RS, Fu JL, Smiles KA, et al. The effects of minoxidil, 1 % pyrithione zinc and a combination of both on hair density: a randomized controlled trial. Br J Dermatol. 2003;149(2):354–62. https://pubmed.ncbi.nlm.nih.gov/27552261/
Miao Y, Sun Y, Wang W, et al. 6-gingerol inhibits hair shaft growth in cultured human hair follicles and modulates hair growth in mice. PLoS One. 2013;8(2):e57226. https://pubmed.ncbi.nlm.nih.gov/23437345/
Li Y, Han M, Lin P, He Y, Yu J, Zhao R. Hair growth promotion activity and its mechanism of Polygonum multiflorum. Evid Based Complement Alternat Med. 2015;2015:517901. https://pubmed.ncbi.nlm.nih.gov/26294926/
Shin JY, Choi YH, Kim J, et al. Polygonum multiflorum extract support hair growth by elongating anagen phase and abrogating the effect of androgen in cultured human dermal papilla cells. BMC Complement Med Ther. 2020;20(1):144. https://pubmed.ncbi.nlm.nih.gov/32398000/
Teka T, Wang L, Gao J, et al. Polygonum multiflorum: recent updates on newly isolated compounds, potential hepatotoxic compounds and their mechanisms. J Ethnopharmacol. 2021;271:113864. https://pubmed.ncbi.nlm.nih.gov/33485980/
Hay IC, Jamieson M, Ormerod AD. Randomized trial of aromatherapy: successful treatment for alopecia areata. Arch Dermatol. 1998;134(11):1349–52. https://pubmed.ncbi.nlm.nih.gov/9828867/
Sharquie KE, Al-Obaidi HK. Onion juice (Allium cepa L.), a new topical treatment for alopecia areata. J Dermatol. 2002;29(6):343–6. https://pubmed.ncbi.nlm.nih.gov/12126069/
Hajheydari Z, Jamshidi M, Akbari J, Mohammadpour R. Combination of topical garlic gel and betamethasone valerate cream in the treatment of localized alopecia areata: a double-blind randomized controlled study. Indian J Dermatol Venereol Leprol. 2007;73(1):29–32. https://pubmed.ncbi.nlm.nih.gov/17314444/
Panahi Y, Taghizadeh M, Marzony ET, Sahebkar A. Rosemary oil vs minoxidil 2 % for the treatment of androgenetic alopecia: a randomized comparative trial. Skinmed. 2015;13(1):15–21. https://pubmed.ncbi.nlm.nih.gov/25842469/
Ajmani GS, Suh HH, Wroblewski KE, Pinto JM. Smoking and olfactory dysfunction: a systematic literature review and meta-analysis. Laryngoscope. 2017;127(8):1753–61. https://pubmed.ncbi.nlm.nih.gov/28561327/
Desiato VM, Levy DA, Byun YJ, Nguyen SA, Soler ZM, Schlosser RJ. The prevalence of olfactory dysfunction in the general population: a systematic review and meta-analysis. Am J Rhinol Allergy. 2021;35(2):195–205. https://pubmed.ncbi.nlm.nih.gov/32746612/
Stevens JC, Cain WS, Demarque A, Ruthruff AM. On the discrimination of missing ingredients: aging and salt flavor. Appetite. 1991;16(2):129–40. https://pubmed.ncbi.nlm.nih.gov/2064391/
Schäfer L, Schriever VA, Croy I. Human olfactory dysfunction: causes and consequences. Cell Tissue Res. 2021;383(1):569–79. https://pubmed.ncbi.nlm.nih.gov/33496882/
Nolan LS. Age-related hearing loss: why we need to think about sex as a biological variable. J Neurosci Res. 2020;98(9):1705–20. https://pubmed.ncbi.nlm.nih.gov/32557661/
Mao Z, Zhao L, Pu L, Wang M, Zhang Q, He DZZ. How well can centenarians hear? PLoS One. 2013;8(6):e65565. https://pubmed.ncbi.nlm.nih.gov/23755251/
Committee on Accessible and Affordable Hearing Health Care for Adults. Blazer DG, Domnitz S, Liverman CT, eds. Hearing Health Care for Adults: Priorities for Improving Access and Affordability. National Academies Press; 2016. https://pubmed.ncbi.nlm.nih.gov/27280276/
Goman AM, Lin FR. Prevalence of hearing loss by severity in the United States. Am J Public Health. 2016;106(10):1820–2. https://pubmed.ncbi.nlm.nih.gov/27552261/
Mao Z, Zhao L, Pu L, Wang M, Zhang Q, He DZZ. How well can centenarians hear? PLoS One. 2013;8(6):e65565. https://pubmed.ncbi.nlm.nih.gov/23755251/
Wattamwar K, Qian ZJ, Otter J, et al. Increases in the rate of age-related hearing loss in the older old. JAMA Otolaryngol Head Neck Surg. 2017;143(1):41–5. https://pubmed.ncbi.nlm.nih.gov/27632707/
Shukla A, Harper M, Pedersen E, et al. Hearing loss, loneliness, and social isolation: a systematic review. Otolaryngol Head Neck Surg. 2020;162(5):622–33. https://pubmed.ncbi.nlm.nih.gov/32151193/
Lawrence BJ, Jayakody DMP, Bennett RJ, Eikelboom RH, Gasson N, Friedland PL. Hearing loss and depression in older adults: a systematic review and meta-analysis. Gerontologist. 2020;60(3):e137–54. https://pubmed.ncbi.nlm.nih.gov/30835787/
Wattamwar K, Qian ZJ, Otter J, et al. Increases in the rate of age-related hearing loss in the older old. JAMA Otolaryngol Head Neck Surg. 2017;143(1):41–5. https://pubmed.ncbi.nlm.nih.gov/27632707/
Goman AM, Lin FR. Hearing loss in older adults – from epidemiological insights to national initiatives. Hear Res. 2018;369:29–32. https://pubmed.ncbi.nlm.nih.gov/29653842/
Brennan-Jones CG, Weeda E, Ferguson M. Cochrane corner: hearing aids for mild to moderate hearing loss in adults. Int J Audiol. 2018;57(7):479–82. https://pubmed.ncbi.nlm.nih.gov/29383941/
Mahmoudi E, Basu T, Langa K, et al. Can hearing aids delay time to diagnosis of dementia, depression, or falls in older adults? J Am Geriatr Soc. 2019;67(11):2362–9. https://pubmed.ncbi.nlm.nih.gov/31486068/
Goman AM, Lin FR. Hearing loss in older adults – from epidemiological insights to national initiatives. Hear Res. 2018;369:29–32. https://pubmed.ncbi.nlm.nih.gov/29653842/
Franck KH, Rathi VK. Regulation of over-the-counter hearing aids – deafening silence from the FDA. N Engl J Med. 2020;383(21):1997–2000. https://pubmed.ncbi.nlm.nih.gov/33207090/
Fact Sheet: cheaper hearing aids now in stores thanks to Biden-Harris administration competition agenda. WhiteHouse.gov. https://www.whitehouse.gov/briefing-room/statements-releases/2022/10/17/fact-sheet-cheaper-hearing-aids-now-in-stores-thanks-to-biden-harris-administration-competition-agenda/. Published October 17, 2022. Accessed January 3, 2023.; https://www.whitehouse.gov/briefing-room/statements-releases/2022/10/17/fact-sheet-cheaper-hearing-aids-now-in-stores-thanks-to-biden-harris-administration-competition-agenda
Michaud HN, Duchesne L. Aural rehabilitation for older adults with hearing loss: impacts on quality of life – a systematic review of randomized controlled trials. J Am Acad Audiol. 2017;28(7):596–609. https://pubmed.ncbi.nlm.nih.gov/28722643/
Ferguson MA, Kitterick PT, Chong LY, Edmondson-Jones M, Barker F, Hoare DJ. Hearing aids for mild to moderate hearing loss in adults. Cochrane Database Syst Rev. 2017;2017(9):CD012023. https://pubmed.ncbi.nlm.nih.gov/28944461/
Brennan-Jones CG, Weeda E, Ferguson M. Cochrane corner: hearing aids for mild to moderate hearing loss in adults. Int J Audiol. 2018;57(7):479–82. https://pubmed.ncbi.nlm.nih.gov/29383941/
Lerner S. Limitations of conventional hearing aids: examining common complaints and issues that can and cannot be remedied. Otolaryngol Clin North Am. 2019;52(2):211–20. https://pubmed.ncbi.nlm.nih.gov/30612754/
Davis A, McMahon CM, Pichora-Fuller KM, et al. Aging and hearing health: the life-course approach. Gerontologist. 2016;56 Suppl 2:S256–67. https://pubmed.ncbi.nlm.nih.gov/26994265/
McCormack A, Fortnum H. Why do people fitted with hearing aids not wear them? Int J Audiol. 2013;52(5):360–8. https://pubmed.ncbi.nlm.nih.gov/23473329/
Blustein J, Weinstein BE, Chodosh J. Marketing claims about using hearing aids to forestall or prevent dementia. JAMA Otolaryngol Head Neck Surg. 2020;146(8):765–6. https://pubmed.ncbi.nlm.nih.gov/32556250/
World Health Organization. Risk Reduction of Cognitive Decline and Dementia: WHO Guidelines. World Health Organization; 2019. https://www.who.int/publications/i/item/9789241550543
Schwartz SR, Magit AE, Rosenfeld RM, et al. Clinical practice guideline (update): earwax (cerumen impaction). Otolaryngol Head Neck Surg. 2017;156(1_suppl):S1–29. https://pubmed.ncbi.nlm.nih.gov/28045591/
Nagala S, Singh P, Tostevin P. Extent of cotton-bud use in ears. Br J Gen Pract. 2011;61(592):662–3. https://pubmed.ncbi.nlm.nih.gov/22054319/
Baxter P. Association between use of cotton tipped swabs and cerumen plugs. BMJ. 1983;287(6401):1260. https://pubmed.ncbi.nlm.nih.gov/6416358/
Barton RT. Q-tip otalgia. JAMA. 1972;220(12):1619. https://pubmed.ncbi.nlm.nih.gov/5067751/
Schwartz SR, Magit AE, Rosenfeld RM, et al. Clinical practice guideline (update): earwax (cerumen impaction). Otolaryngol Head Neck Surg. 2017;156(1_suppl):S1–29. https://pubmed.ncbi.nlm.nih.gov/28045591/
Oron Y, Zwecker-Lazar I, Levy D, Kreitler S, Roth Y. Cerumen removal: comparison of cerumenolytic agents and effect on cognition among the elderly. Arch Gerontol Geriatr. 2011;52(2):228–32. https://pubmed.ncbi.nlm.nih.gov/20417976/
Schwartz SR, Magit AE, Rosenfeld RM, et al. Clinical practice guideline (update): earwax (cerumen impaction). Otolaryngol Head Neck Surg. 2017;156(1_suppl):S1–29. https://pubmed.ncbi.nlm.nih.gov/28045591/
Lee LM, Govindaraju R, Hon SK. Cotton bud and ear cleaning – a loose tip cotton bud? Med J Malaysia. 2005;60(1):85–8. https://pubmed.ncbi.nlm.nih.gov/16250286/
Schwartz SR, Magit AE, Rosenfeld RM, et al. Clinical practice guideline (update): earwax (cerumen impaction). Otolaryngol Head Neck Surg. 2017;156(1_suppl):S1–29. https://pubmed.ncbi.nlm.nih.gov/28045591/
Goldman SA, Ankerstjerne JK, Welker KB, Chen DA. Fatal meningitis and brain abscess resulting from foreign body-induced otomastoiditis. Otolaryngol Head Neck Surg. 1998;118(1):6–8. https://pubmed.ncbi.nlm.nih.gov/9450821/
Aaron K, Cooper TE, Warner L, Burton MJ. Ear drops for the removal of ear wax. Cochrane ENT Group, ed. Cochrane Database of Systematic Reviews. 2018;7(CD012171). https://pubmed.ncbi.nlm.nih.gov/30043448/
Schwartz SR, Magit AE, Rosenfeld RM, et al. Clinical practice guideline (update): earwax (cerumen impaction). Otolaryngol Head Neck Surg. 2017;156(1_suppl):S1–29. https://pubmed.ncbi.nlm.nih.gov/28045591/
Schwartz SR, Magit AE, Rosenfeld RM, et al. Clinical practice guideline (update): earwax (cerumen impaction). Otolaryngol Head Neck Surg. 2017;156(1_suppl):S1–29. https://pubmed.ncbi.nlm.nih.gov/28045591/
Coppin R, Wicke D, Little P. Managing earwax in primary care: efficacy of self-treatment using a bulb syringe. Br J Gen Pract. 2008;58(546):44–9. https://pubmed.ncbi.nlm.nih.gov/18186996/
Coppin R, Wicke D, Little P. Randomized trial of bulb syringes for earwax: impact on health service utilization. Ann Fam Med. 2011;9(2):110–4. https://pubmed.ncbi.nlm.nih.gov/21403136/
Nieman CL, Oh ES. Hearing loss. Ann Intern Med. 2020;173(11):ITC81–96. https://pubmed.ncbi.nlm.nih.gov/33253610/
Dinsdale RC, Roland PS, Manning SC, Meyerhoff WL. Catastrophic otologic injury from oral jet irrigation of the external auditory canal. Laryngoscope. 1991;101(1 Pt 1):75–8. https://pubmed.ncbi.nlm.nih.gov/1984556/
Seely DR, Quigley SM, Langman AW. Ear candles – efficacy and safety. Laryngoscope. 1996;106(10):1226–9. https://pubmed.ncbi.nlm.nih.gov/8849790/
Seely DR, Quigley SM, Langman AW. Ear candles – efficacy and safety. Laryngoscope. 1996;106(10):1226–9. https://pubmed.ncbi.nlm.nih.gov/8849790/
Schwartz SR, Magit AE, Rosenfeld RM, et al. Clinical practice guideline (update): earwax (cerumen impaction). Otolaryngol Head Neck Surg. 2017;156(1_suppl):S1–29. https://pubmed.ncbi.nlm.nih.gov/28045591/
Mao Z, Zhao L, Pu L, Wang M, Zhang Q, He DZZ. How well can centenarians hear? PLoS One. 2013;8(6):e65565. https://pubmed.ncbi.nlm.nih.gov/23755251/
Donnison CP. Blood pressure in the African native. Lancet. 1929;213(5497):6–7. https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(00)49248-2/fulltext
Morse WR, McGill MD, Beh YT. Blood pressure amongst aboriginal ethnic groups of Szechwan Province, West China. Lancet. 1937;229(5929):966–8. https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(00)86708-2/fulltext
Mueller NT, Noya-Alarcon O, Contreras M, Appel LJ, Dominguez-Bello MG. Association of age with blood pressure across the lifespan in isolated Yanomami and Yekwana villages. JAMA Cardiol. 2018;3(12):1247–9. https://pubmed.ncbi.nlm.nih.gov/30427998/
Rosen S, Bergman M, Plester D, El-Mofty A, Satti MH. Presbycusis study of a relatively noise-free population in the Sudan. Ann Otol Rhinol Laryngol. 1962;71:727–43. https://pubmed.ncbi.nlm.nih.gov/13974856/
Goycoolea MV, Goycoolea HG, Farfan CR, Rodriguez LG, Martinez GC, Vidal R. Effect of life in industrialized societies on hearing in natives of Easter Island. Laryngoscope. 1986;96(12):1391–6. https://pubmed.ncbi.nlm.nih.gov/3784745/
Wu PZ, O’Malley JT, de Gruttola V, Liberman MC. Age-related hearing loss is dominated by damage to inner ear sensory cells, not the cellular battery that powers them. J Neurosci. 2020;40(33):6357–66. https://pubmed.ncbi.nlm.nih.gov/32690619/
Nieman CL, Oh ES. Hearing loss. Ann Intern Med. 2020;173(11):ITC81–96. https://pubmed.ncbi.nlm.nih.gov/33253610/
Momi SK, Wolber LE, Fabiane SM, MacGregor AJ, Williams FMK. Genetic and environmental factors in age-related hearing impairment. Twin Res Hum Genet. 2015;18(4):383–92. https://pubmed.ncbi.nlm.nih.gov/26081266/
Mao Z, Zhao L, Pu L, Wang M, Zhang Q, He DZZ. How well can centenarians hear? PLoS One. 2013;8(6):e65565. https://pubmed.ncbi.nlm.nih.gov/23755251/
Wang J, Puel JL. Presbycusis: an update on cochlear mechanisms and therapies. J Clin Med. 2020;9(1):E218. https://pubmed.ncbi.nlm.nih.gov/31947524/
Attarha M, Bigelow J, Merzenich MM. Unintended consequences of white noise therapy for tinnitus – otolaryngology’s cobra effect: a review. JAMA Otolaryngol Head Neck Surg. 2018;144(10):938–43. https://pubmed.ncbi.nlm.nih.gov/30178067/
Attarha M, Bigelow J, Merzenich MM. No evidence of broadband noise having any harmful effect on hearing. JAMA Otolaryngol Head Neck Surg. 2019;145(3):292–3. https://pubmed.ncbi.nlm.nih.gov/30676625/
Nieman CL, Oh ES. Hearing loss. Ann Intern Med. 2020;173(11):ITC81–96. https://pubmed.ncbi.nlm.nih.gov/33253610/
Joo Y, Cruickshanks KJ, Klein BEK, Klein R, Hong O, Wallhagen MI. The contribution of ototoxic medications to hearing loss among older adults. J Gerontol A Biol Sci Med Sci. 2020;75(3):561–6. https://pubmed.ncbi.nlm.nih.gov/31282945/
Panda NK, Modi R, Munjal S, Virk RS. Auditory changes in mobile users: is evidence forthcoming? Otolaryngol Head Neck Surg. 2011;144(4):581–5. https://pubmed.ncbi.nlm.nih.gov/21493239/
Alsanosi AA, Al-Momani MO, Hagr AA, Almomani FM, Shami IM, Al-Habeeb SF. The acute auditory effects of exposure for 60 minutes to mobile’s electromagnetic field. Saudi Med J. 2013;34(2):142–6. https://pubmed.ncbi.nlm.nih.gov/23396459/
Mandalà M, Colletti V, Sacchetto L, et al. Effect of Bluetooth headset and mobile phone electromagnetic fields on the human auditory nerve. Laryngoscope. 2014;124(1):255–9. https://pubmed.ncbi.nlm.nih.gov/23619813/
Rosen S, Olin P. Hearing loss and coronary heart disease. Arch Otolaryngol. 1965;82:236–43. https://pubmed.ncbi.nlm.nih.gov/14327021/
Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension. 2018;71(6):1269–324. https://pubmed.ncbi.nlm.nih.gov/29133354/
Rosen S, Olin P. Hearing loss and coronary heart disease. Arch Otolaryngol. 1965;82:236–43. https://pubmed.ncbi.nlm.nih.gov/14327021/
Rosen S, Bergman M, Plester D, El-Mofty A, Satti MH. Presbycusis study of a relatively noise-free population in the Sudan. Ann Otol Rhinol Laryngol. 1962;71:727–43. https://pubmed.ncbi.nlm.nih.gov/13974856/
Curhan SG, Halpin C, Wang M, Eavey RD, Curhan GC. Prospective study of dietary patterns and hearing threshold elevation. Am J Epidemiol. 2020;189(3):204–14. https://pubmed.ncbi.nlm.nih.gov/31608356/
Rosen S, Bergman M, Plester D, El-Mofty A, Satti MH. Presbycusis study of a relatively noise-free population in the Sudan. Ann Otol Rhinol Laryngol. 1962;71:727–43. https://pubmed.ncbi.nlm.nih.gov/13974856/
Gopinath B, Flood VM, McMahon CM, Burlutsky G, Brand-Miller J, Mitchell P. Dietary glycemic load is a predictor of age-related hearing loss in older adults. J Nutr. 2010;140(12):2207–12. https://pubmed.ncbi.nlm.nih.gov/20926604/
Samocha-Bonet D, Wu B, Ryugo DK. Diabetes mellitus and hearing loss: a review. Ageing Res Rev. 2021;71:101423. https://pubmed.ncbi.nlm.nih.gov/34384902/
Prasad MPR, Rao BD, Kalpana K, Rao MV, Patil JV. Glycaemic index and glycaemic load of sorghum products. J Sci Food Agric. 2015;95(8):1626–30. https://pubmed.ncbi.nlm.nih.gov/25092385/
Poquette NM, Gu X, Lee SO. Grain sorghum muffin reduces glucose and insulin responses in men. Food Funct. 2014;5(5):894–9. https://pubmed.ncbi.nlm.nih.gov/24608948/
Honkura Y, Matsuo H, Murakami S, et al. NRF2 is a key target for prevention of noise-induced hearing loss by reducing oxidative damage of cochlea. Sci Rep. 2016;6:19329. https://pubmed.ncbi.nlm.nih.gov/26776972/
Yang JR, Hidayat K, Chen CL, Li YH, Xu JY, Qin LQ. Body mass index, waist circumference, and risk of hearing loss: a meta-analysis and systematic review of observational study. Environ Health Prev Med. 2020;25(1):25. https://pubmed.ncbi.nlm.nih.gov/32590951/
Wang J, Puel JL. Presbycusis: an update on cochlear mechanisms and therapies. J Clin Med. 2020;9(1):E218. https://pubmed.ncbi.nlm.nih.gov/31947524/
de Rivera C, Shukitt-Hale B, Joseph JA, Mendelson JR. The effects of antioxidants in the senescent auditory cortex. Neurobiol Aging. 2006;27(7):1035–44. https://pubmed.ncbi.nlm.nih.gov/15950320/
Seidman MD, Khan MJ, Bai U, Shirwany N, Quirk WS. Biologic activity of mitochondrial metabolites on aging and age-related hearing loss. Am J Otol. 2000;21(2):161–7. https://pubmed.ncbi.nlm.nih.gov/10733178/
Sanz-Fernández R, Sánchez-Rodriguez C, Granizo JJ, Durio-Calero E, Martín-Sanz E. Accuracy of auditory steady state and auditory brainstem responses to detect the preventive effect of polyphenols on age-related hearing loss in Sprague-Dawley rats. Eur Arch Otorhinolaryngol. 2016;273(2):341–7. https://pubmed.ncbi.nlm.nih.gov/25673025/
Polanski JF, Cruz OL. Evaluation of antioxidant treatment in presbyacusis: prospective, placebo-controlled, double-blind, randomised trial. J Laryngol Otol. 2013;127(2):134–41. https://pubmed.ncbi.nlm.nih.gov/23318104/
Durga J, Verhoef P, Anteunis LJC, Schouten E, Kok FJ. Effects of folic acid supplementation on hearing in older adults: a randomized, controlled trial. Ann Intern Med. 2007;146(1):1–9. https://pubmed.ncbi.nlm.nih.gov/17200216/
Agricultural Research Service, United States Department of Agriculture. Lentils, mature seeds, cooked, boiled, without salt. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=kale&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/172421/nutrients. Published April 1, 2019. Accessed April 21, 2022.; https://fdc.nal.usda.gov/fdc-app.html?query=kale&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/172421/nutrients
Agricultural Research Service, United States Department of Agriculture. Edamame, frozen, prepared. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/food-details/168411/nutrients. Published April 1, 2019. Accessed February 21, 2023.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/169414/nutrients
Rodrigo L, Campos-Asensio C, Rodríguez MÁ, Crespo I, Olmedillas H. Role of nutrition in the development and prevention of age-related hearing loss: a scoping review. J Formos Med Assoc. 2021;120(1 Pt 1):107–20. https://pubmed.ncbi.nlm.nih.gov/32473863/
Pillsbury HC. Hypertension, hyperlipoproteinemia, chronic noise exposure: is there synergism in cochlear pathology? Laryngoscope. 1986;96(10):1112–38. https://pubmed.ncbi.nlm.nih.gov/3762287/
Sikora MA, Morizono T, Ward WD, Paparella MM, Leslie K. Diet-induced hyperlipidemia and auditory dysfunction. Acta Oto-Laryngologica. 1986;102(5–6):372–81. https://pubmed.ncbi.nlm.nih.gov/3788535/
Momi SK, Wolber LE, Fabiane SM, MacGregor AJ, Williams FMK. Genetic and environmental factors in age-related hearing impairment. Twin Res Hum Genet. 2015;18(4):383–92. https://pubmed.ncbi.nlm.nih.gov/26081266/
Gopinath B, Flood VM, Teber E, McMahon CM, Mitchell P. Dietary intake of cholesterol is positively associated and use of cholesterol-lowering medication is negatively associated with prevalent age-related hearing loss. J Nutr. 2011;141(7):1355–61. https://pubmed.ncbi.nlm.nih.gov/21613455/
Erkan AF, Beriat GK, Ekici B, Dogan C, Kocatürk S, Töre HF. Link between angiographic extent and severity of coronary artery disease and degree of sensorineural hearing loss. Herz. 2015;40(3):481–6. https://pubmed.ncbi.nlm.nih.gov/24049023/
Croll PH, Bos D, Vernooij MW, et al. Carotid atherosclerosis is associated with poorer hearing in older adults. J Am Med Dir Assoc. 2019;20(12):1617–22.e1. https://pubmed.ncbi.nlm.nih.gov/31399361/
Fischer ME, Schubert CR, Nondahl DM, et al. Subclinical atherosclerosis and increased risk of hearing impairment. Atherosclerosis. 2015;238(2):344–9. https://pubmed.ncbi.nlm.nih.gov/25555266/
Fischer ME, Schubert CR, Nondahl DM, et al. Subclinical atherosclerosis and increased risk of hearing impairment. Atherosclerosis. 2015;238(2):344–9. https://pubmed.ncbi.nlm.nih.gov/25555266/
Erkan AF, Beriat GK, Ekici B, Dogan C, Kocatürk S, Töre HF. Link between angiographic extent and severity of coronary artery disease and degree of sensorineural hearing loss. Herz. 2015;40(3):481–6. https://pubmed.ncbi.nlm.nih.gov/24049023/
Sikora MA, Morizono T, Ward WD, Paparella MM, Leslie K. Diet-induced hyperlipidemia and auditory dysfunction. Acta Oto-Laryngologica. 1986;102(5–6):372–81. https://pubmed.ncbi.nlm.nih.gov/3788535/
Saito T, Sato K, Saito H. An experimental study of auditory dysfunction associated with hyperlipoproteinemia. Arch Otorhinolaryngol. 1986;243(4):242–5. https://pubmed.ncbi.nlm.nih.gov/3778299/
Turpeinen O, Roine P, Pekkarinen M, et al. Effect on serum-cholesterol level of replacement of dietary milk fat by soybean oil. Lancet. January 23, 1960;196–9. https://pubmed.ncbi.nlm.nih.gov/13839984/
Hearing loss and coronary heart disease. JAMA. 1965;194(4):452 https://pubmed.ncbi.nlm.nih.gov/5897421/
Puga AM, Pajares MA, Varela-Moreiras G, Partearroyo T. Interplay between nutrition and hearing loss: state of art. Nutrients. 2018;11(1):E35. https://pubmed.ncbi.nlm.nih.gov/30586880/
Hearing loss and coronary heart disease. JAMA. 1965;194(4):452. https://pubmed.ncbi.nlm.nih.gov/5897421/
Rosen S, Olin P, Rosen HV. Dietary prevention of hearing loss. Acta Oto-Laryngologica. 1970;70(4):242–7. https://pubmed.ncbi.nlm.nih.gov/5491161/
Nobus D. The madness of Princess Alice: Sigmund Freud, Ernst Simmel and Alice of Battenberg at Kurhaus Schloß Tegel. Hist Psychiatry. 2020;31(2):147–62. https://pubmed.ncbi.nlm.nih.gov/31969019/
Brown-Séquard. Note on the effects produced on man by subcutaneous injections of a liquid obtained from the testicles of animals. Lancet. 1889;134(3438):105–7. https://www.sciencedirect.com/science/article/abs/pii/S0140673600641181
Kahn A. Regaining lost youth: the controversial and colorful beginnings of hormone replacement therapy in aging. J Gerontol A Biol Sci Med Sci. 2005;60(2):142–7. https://pubmed.ncbi.nlm.nih.gov/15814854/
Blue E. The strange career of Leo Stanley: remaking manhood and medicine at San Quentin State Penitentiary, 1913–1951. Pac Hist Rev. 2009;78(2):210–41. https://www.researchgate.net/publication/236335739_The_Strange_Career_of_Leo_Stanley_Remaking_Manhood_and_Medicine_at_San_Quentin_State_Penitentiary_1913-1951
Perls TT. Anti-aging quackery: human growth hormone and tricks of the trade – more dangerous than ever. J Gerontol A Biol Sci Med Sci. 2004;59(7):682–91. https://pubmed.ncbi.nlm.nih.gov/15304532/
Irwig MS, Fleseriu M, Jonklaas J, et al. Off-label use and misuse of testosterone, growth hormone, thyroid hormone, and adrenal supplements: risks and costs of a growing problem. Endocr Pract. 2020;26(3):340–53. https://pubmed.ncbi.nlm.nih.gov/32163313/
Regelson W. Growth hormone use. Science. 1987;235(4784):14c-5c. https://pubmed.ncbi.nlm.nih.gov/17769289/
Barkan AL. Growth hormone as an anti-aging therapy – do the benefits outweigh the risks? Nat Clin Pract Endocrinol Metab. 2007;3(7):508–9. https://pubmed.ncbi.nlm.nih.gov/17534272/
Irwig MS, Fleseriu M, Jonklaas J, et al. Off-label use and misuse of testosterone, growth hormone, thyroid hormone, and adrenal supplements: risks and costs of a growing problem. Endocr Pract. 2020;26(3):340–53. https://pubmed.ncbi.nlm.nih.gov/32163313/
Mullur RS. Making a difference in adrenal fatigue. Endocr Pract. 2018;24(12):1103–5. https://pubmed.ncbi.nlm.nih.gov/30289314/
Cadegiani FA, Kater CE. Adrenal fatigue does not exist: a systematic review. BMC Endocr Disord. 2016;16(1):48. https://pubmed.ncbi.nlm.nih.gov/27557747/
Nippoldt T. Mayo Clinic office visit. Adrenal fatigue: an interview with Todd Nippoldt, M.D. Mayo Clin Womens Healthsource. 2010;14(3):6. https://pubmed.ncbi.nlm.nih.gov/20110864/
Chimote BN, Chimote NM. Dehydroepiandrosterone (DHEA) and its sulfate (DHEA-S) in mammalian reproduction: known roles and novel paradigms. Vitam Horm. 2018;108:223–50. https://pubmed.ncbi.nlm.nih.gov/30029728/
Kim MJ, Morley JE. The hormonal fountains of youth: myth or reality? J Endocrinol Invest. 2005;28(11 Suppl Proceedings):5–14. https://pubmed.ncbi.nlm.nih.gov/16760618/
Peixoto C, Carrilho CG, Barros JA, et al. The effects of dehydroepiandrosterone on sexual function: a systematic review. Climacteric. 2017;20(2):129–37. https://pubmed.ncbi.nlm.nih.gov/28118059/
Rutkowski K, Sowa P, Rutkowska-Talipska J, Kuryliszyn-Moskal A, Rutkowski R. Dehydroepiandrosterone (DHEA): hypes and hopes. Drugs. 2014;74(11):1195–207. https://pubmed.ncbi.nlm.nih.gov/25022952/
ConsumerLab.com tests DHEA supplements, warns of differences in dose and price. ConsumerLab.com. https://www.consumerlab.com/news/dhea-dose-price/07–22–2015/. Published July 22, 2015. Accessed May 5, 2022.; https://www.consumerlab.com/news/dhea-dose-price/07-22-2015
Celec P, Stárka L. Dehydroepiandrosterone – is the fountain of youth drying out? Physiol Res. 2003;52(4):397–407. https://pubmed.ncbi.nlm.nih.gov/12899651/
Peixoto C, Carrilho CG, Barros JA, et al. The effects of dehydroepiandrosterone on sexual function: a systematic review. Climacteric. 2017;20(2):129–37. https://pubmed.ncbi.nlm.nih.gov/28118059/
Wiser A, Gonen O, Ghetler Y, Shavit T, Berkovitz A, Shulman A. Addition of dehydroepiandrosterone (DHEA) for poor-responder patients before and during IVF treatment improves the pregnancy rate: a randomized prospective study. Hum Reprod. 2010;25(10):2496–500. https://pubmed.ncbi.nlm.nih.gov/22456062/
Tartagni M, Cicinelli MV, Baldini D, et al. Dehydroepiandrosterone decreases the age-related decline of the in vitro fertilization outcome in women younger than 40 years old. Reprod Biol Endocrinol. 2015;13:18. https://ncbi.nlm.nih.gov/pmc/articles/PMC4355976/
Thompson RD, Carlson M. Liquid chromatographic determination of dehydroepiandrosterone (DHEA) in dietary supplement products. J AOAC Int. 2000;83(4):847–57. https://pubmed.ncbi.nlm.nih.gov/10995111/
Trichopoulou A, Bamia C, Kalapothaki V, Spanos E, Naska A, Trichopoulos D. Dehydroepiandrosterone relations to dietary and lifestyle variables in a general population sample. Ann Nutr Metab. 2003;47(3–4):158–64. https://pubmed.ncbi.nlm.nih.gov/12743468/
Remer T, Pietrzik K, Manz F. The short-term effect of dietary pectin on plasma levels and renal excretion of dehydroepiandrosterone sulfate. Z Ernahrungswiss. 1996;35(1):32–8. https://pubmed.ncbi.nlm.nih.gov/8776832/
Remer T, Pietrzik K, Manz F. Short-term impact of a lactovegetarian diet on adrenocortical activity and adrenal androgens. J Clin Endocrinol Metab. 1998;83(6):2132–7. https://pubmed.ncbi.nlm.nih.gov/9626151/
Hill P, Wynder EL, Garbaczewski L, Walker AR. Effect of diet on plasma and urinary hormones in South African black men with prostatic cancer. Cancer Res. 1982;42(9):3864–9. https://pubmed.ncbi.nlm.nih.gov/6179613/
Hill P, Garbaczewski L, Helman P, Huskisson J, Sporangisa E, Wynder EL. Diet, lifestyle, and menstrual activity. Am J Clin Nutr. 1980;33(6):1192–8. https://pubmed.ncbi.nlm.nih.gov/7386408/
Remer T, Pietrzik K, Manz F. Short-term impact of a lactovegetarian diet on adrenocortical activity and adrenal androgens. J Clin Endocrinol Metab. 1998;83(6):2132–7. https://pubmed.ncbi.nlm.nih.gov/9626151/
Grande M, Borobio V, Jimenez JM, et al. Antral follicle count as a marker of ovarian biological age to reflect the background risk of fetal aneuploidy. Hum Reprod. 2014;29(6):1337–43. https://pubmed.ncbi.nlm.nih.gov/24682614/
Bozdag G, Calis P, Zengin D, Tanacan A, Karahan S. Age related normogram for antral follicle count in general population and comparison with previous studies. Eur J Obstet Gynecol Reprod Biol. 2016;206:120–4. https://pubmed.ncbi.nlm.nih.gov/27689809/
Souter I, Chiu YH, Batsis M, et al. The association of protein intake (amount and type) with ovarian antral follicle counts among infertile women: results from the EARTH prospective study cohort. BJOG. 2017;124(10):1547–55. https://pubmed.ncbi.nlm.nih.gov/28278351/
Hartmann S, Lacorn M, Steinhart H. Natural occurrence of steroid hormones in food. Food Chem (Oxf). 1998;62(1):7–20. https://www.sciencedirect.com/science/article/abs/pii/S0308814697001507?via%3Dihub
Brinkman MT, Baglietto L, Krishnan K, et al. Consumption of animal products, their nutrient components and postmenopausal circulating steroid hormone concentrations. Eur J Clin Nutr. 2010;64(2):176–83. https://pubmed.ncbi.nlm.nih.gov/19904296/
Andersson AM, Skakkebaek NE. Exposure to exogenous estrogens in food: possible impact on human development and health. Eur J Endocrinol. 1999;140(6):477–85. https://pubmed.ncbi.nlm.nih.gov/10366402/
Souter I, Chiu YH, Batsis M, et al. The association of protein intake (amount and type) with ovarian antral follicle counts among infertile women: results from the EARTH prospective study cohort. BJOG. 2017;124(10):1547–55. https://pubmed.ncbi.nlm.nih.gov/28278351/
Lumsden MA, Sassarini J. The evolution of the human menopause. Climacteric. 2019;22(2):111–6. https://pubmed.ncbi.nlm.nih.gov/30712396/
National Center for Health Statistics. Health, United States, 2010: With Special Feature on Death and Dying. https://www.cdc.gov/nchs/data/hus/hus10.pdf. Published February 2011. Accessed May 5, 2022.; https://www.cdc.gov/nchs/data/hus/hus10.pdf
Llarena N, Hine C. Reproductive longevity and aging: geroscience approaches to maintain long-term ovarian fitness. J Gerontol A Biol Sci Med Sci. 2021;76(9):1551–60. https://pubmed.ncbi.nlm.nih.gov/32808646/
Gagnon A. Natural fertility and longevity. Fertil Steril. 2015;103(5):1109–16. https://pubmed.ncbi.nlm.nih.gov/25934597/
Giri R, Vincent AJ. Prevalence and risk factors of premature ovarian insufficiency/early menopause. Semin Reprod Med. 2020;38(4–05):237–46. https://pubmed.ncbi.nlm.nih.gov/33434933/
Stanford JL, Hartge P, Brinton LA, Hoover RN, Brookmeyer R. Factors influencing the age at natural menopause. J Chronic Dis. 1987;40(11):995–1002. https://pubmed.ncbi.nlm.nih.gov/3654908/
Boutot ME, Purdue-Smithe A, Whitcomb BW, et al. Dietary protein intake and early menopause in the Nurses’ Health Study II. Am J Epidemiol. 2018;187(2):270–7. https://pubmed.ncbi.nlm.nih.gov/28992246/
Conway F. Menopause matters: attending to the vitality of older women. J Women Aging. 2020;32(5):489–90. https://pubmed.ncbi.nlm.nih.gov/33225875/
Minkin MJ. Menopause: hormones, lifestyle, and optimizing aging. Obstet Gynecol Clin North Am. 2019;46(3):501–14. https://pubmed.ncbi.nlm.nih.gov/31378291/
Johnson A, Roberts L, Elkins G. Complementary and alternative medicine for menopause. J Evid Based Integr Med. 2019;24:2515690X19829380. https://pubmed.ncbi.nlm.nih.gov/30868921/
Minkin MJ. Menopause: hormones, lifestyle, and optimizing aging. Obstet Gynecol Clin North Am. 2019;46(3):501–14. https://pubmed.ncbi.nlm.nih.gov/31378291/
Pirhadi R, Sinai Talaulikar V, Onwude J, Manyonda I. It is all in the name: the importance of correct terminology in hormone replacement therapy. Post Reprod Health. 2020;26(3):142–6. https://pubmed.ncbi.nlm.nih.gov/32390508/
Hunter MM, Huang AJ, Wallhagen MI. “I’m going to stay young”: belief in anti-aging efficacy of menopausal hormone therapy drives prolonged use despite medical risks. PLoS One. 2020;15(5):e0233703. https://pubmed.ncbi.nlm.nih.gov/32469976/
Wilson RA, Wilson TA. The fate of the nontreated postmenopausal woman: a plea for the maintenance of adequate estrogen from puberty to the grave. J Am Geriatr Soc. 1963;11:347–62. https://pubmed.ncbi.nlm.nih.gov/14001078/
Wilson RA, Wilson TA. The basic philosophy of estrogen maintenance. J Am Geriatr Soc.1972;20(11):521–3. https://pubmed.ncbi.nlm.nih.gov/5082121/
Chew F, Wu X. Sources of information influencing the state-of-the-science gap in hormone replacement therapy usage. PLoS One. 2017;12(2):e0171189. https://pubmed.ncbi.nlm.nih.gov/28158240/
Fugh-Berman A. The science of marketing: how pharmaceutical companies manipulated medical discourse on menopause. Women’s Reprod Health. 2015;2(1):18–23. https://www.tandfonline.com/doi/full/10.1080/23293691.2015.1039448
Rubinstein H. Defining what is normal at menopause: how women’s and clinician’s different understandings may lead to a lack of provision for those in most need. Hum Fertil (Camb). 2014;17(3):218–22. https://pubmed.ncbi.nlm.nih.gov/24989874/
Tatsioni A, Siontis GCM, Ioannidis JPA. Partisan perspectives in the medical literature: a study of high frequency editorialists favoring hormone replacement therapy. J Gen Intern Med. 2010;25(9):914–9. https://pubmed.ncbi.nlm.nih.gov/20425148/
Rubinstein H. Defining what is normal at menopause: how women’s and clinician’s different understandings may lead to a lack of provision for those in most need. Hum Fertil (Camb). 2014;17(3):218–22. https://pubmed.ncbi.nlm.nih.gov/24989874/
Minkin MJ. Menopause: hormones, lifestyle, and optimizing aging. Obstet Gynecol Clin North Am. 2019;46(3):501–14. https://pubmed.ncbi.nlm.nih.gov/31378291/
Verkooijen HM, Bouchardy C, Vinh-Hung V, Rapiti E, Hartman M. The incidence of breast cancer and changes in the use of hormone replacement therapy: a review of the evidence. Maturitas. 2009;64(2):80–5. https://pubmed.ncbi.nlm.nih.gov/19709827/
Auchincloss H, Haagensen CD. Cancer of the breast possibly induced by estrogenic substance. JAMA. 1940;114(16):1517–23. https://jamanetwork.com/journals/jama/article-abstract/1160126
Anderson G, Cummings S, Freedman LS, et al. Design of the Women’s Health Initiative clinical trial and observational study. Control Clin Trials. 1998;19(1):61–109. https://pubmed.ncbi.nlm.nih.gov/9492970/
Cummings SR. Evaluating the benefits and risks of postmenopausal hormone therapy. Am J Med. 1991;91(5B):14S-8S. https://pubmed.ncbi.nlm.nih.gov/1750410/
Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321–33. https://pubmed.ncbi.nlm.nih.gov/12117397/
Marsden J. Hormone replacement therapy and female malignancy: what has the Million Women Study added to our knowledge? J Fam Plann Reprod Health Care. 2007;33(4):237–43. https://pubmed.ncbi.nlm.nih.gov/17925102/
Anderson GL, Limacher M, Assaf AR, et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women’s Health Initiative randomized controlled trial. JAMA. 2004;291(14):1701–12. https://pubmed.ncbi.nlm.nih.gov/15082697/
Fugh-Berman A, Pearson C. The overselling of hormone replacement therapy. Pharmacotherapy. 2002;22(9):1205–8. https://pubmed.ncbi.nlm.nih.gov/12222561/
Katz A. Observations and advertising: controversies in the prescribing of hormone replacement therapy. Health Care Women Int. 2003;24(10):927–39. https://pubmed.ncbi.nlm.nih.gov/14742130/
Majumdar SR, Almasi EA, Stafford RS. Promotion and prescribing of hormone therapy after report of harm by the Women’s Health Initiative. JAMA. 2004;292(16):1983–8. https://pubmed.ncbi.nlm.nih.gov/15507584/
Ravdin PM, Cronin KA, Howlader N, et al. The decrease in breast-cancer incidence in 2003 in the United States. N Engl J Med. 2007;356(16):1670–4. https://pubmed.ncbi.nlm.nih.gov/17442911/
Fugh-Berman A. The science of marketing: how pharmaceutical companies manipulated medical discourse on menopause. Women’s Reprod Health. 2015;2(1):18–23. https://www.tandfonline.com/doi/full/10.1080/23293691.2015.1039448
Roth JA, Etzioni R, Waters TM, et al. Economic return from the Women’s Health Initiative estrogen plus progestin clinical trial: a modeling study. Ann Intern Med. 2014;160(9):594–602. https://pubmed.ncbi.nlm.nih.gov/24798522/
Majumdar SR, Almasi EA, Stafford RS. Promotion and prescribing of hormone therapy after report of harm by the Women’s Health Initiative. JAMA. 2004;292(16):1983–8. https://pubmed.ncbi.nlm.nih.gov/15507584/
Carstens AJ. HRT prescriptions linked to 25 % of breast cancers in California. S Afr Med J. 2009;99(5):280. https://pubmed.ncbi.nlm.nih.gov/19588781/
Fugh-Berman AJ. The haunting of medical journals: how ghostwriting sold “HRT.” PLoS Med. 2010;7(9):e1000335. https://pubmed.ncbi.nlm.nih.gov/20838656/
Fugh-Berman A, Scialli AR. Gynecologists and estrogen: an affair of the heart. Perspect Biol Med. 2006;49(1):115–30. https://pubmed.ncbi.nlm.nih.gov/16489281/
Fugh-Berman AJ. The haunting of medical journals: how ghostwriting sold “HRT.” PLoS Med. 2010;7(9):e1000335. https://pubmed.ncbi.nlm.nih.gov/20838656/
Egilman AC, Kesselheim AS, Krumholz HM, Ross JS, Kim J, Kapczynski A. Confidentiality orders and public interest in drug and medical device litigation. JAMA Intern Med. 2020;180(2):292–9. https://pubmed.ncbi.nlm.nih.gov/31657836/
Fugh-Berman A, Scialli AR. Gynecologists and estrogen: an affair of the heart. Perspect Biol Med. 2006;49(1):115–30. https://pubmed.ncbi.nlm.nih.gov/16489281/
Tatsioni A, Siontis GCM, Ioannidis JPA. Partisan perspectives in the medical literature: a study of high frequency editorialists favoring hormone replacement therapy. J Gen Intern Med. 2010;25(9):914–9. https://pubmed.ncbi.nlm.nih.gov/20425148/
Fugh-Berman A, Pearson C. The overselling of hormone replacement therapy. Pharmacotherapy. 2002;22(9):1205–8. https://pubmed.ncbi.nlm.nih.gov/12222561/
Akesson A, Weismayer C, Newby PK, Wolk A. Combined effect of low-risk dietary and lifestyle behaviors in primary prevention of myocardial infarction in women. Arch Intern Med. 2007;167(19):2122–7. https://pubmed.ncbi.nlm.nih.gov/17954808/
American Academy of Family Physicians. Clinical preventative service recommendation: hormone replacement therapy. https://www.aafp.org/family-physician/patient-care/clinical-recommendations/all-clinical-recommendations/hrt.html. Accessed Aug 2, 2022.; https://www.aafp.org/family-physician/patient-care/clinical-recommendations/all-clinical-recommendations/hrt.html
Fick DM, Semla TP, Steinman M, et al. American Geriatrics Society 2019 updated AGS Beers criteria® for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2019;67(4):674–94. https://pubmed.ncbi.nlm.nih.gov/30693946/
Mosca L, Benjamin EJ, Berra K, et al. Effectiveness-based guidelines for the prevention of cardiovascular disease in women—2011 update. J Am Coll Cardiol. 2011;57(12):1404–23. https://pubmed.ncbi.nlm.nih.gov/21325087/
Grossman DC, Curry SJ, Owens DK, et al. Hormone therapy for the primary prevention of chronic conditions in postmenopausal women: US Preventive Services Task Force recommendation statement. JAMA. 2017;318(22):2224–33. https://pubmed.ncbi.nlm.nih.gov/29677309/
ACOG Committee Opinion No. 565: Hormone therapy and heart disease. Obstet Gynecol. 2013;121(6):1407–10. https://pubmed.ncbi.nlm.nih.gov/23812486/
Maclennan AH, Broadbent JL, Lester S, Moore V. Oral oestrogen and combined oestrogen/progestogen therapy versus placebo for hot flushes. Cochrane Database Syst Rev. 2004;(4):CD002978. https://pubmed.ncbi.nlm.nih.gov/15495039/
Santoro N, Allshouse A, Neal-Perry G, et al. Longitudinal changes in menopausal symptoms comparing women randomized to low-dose oral conjugated estrogens or transdermal estradiol plus micronized progesterone versus placebo: the Kronos Early Estrogen Prevention Study (KEEPS). Menopause. 2017;24(3):238–46. https://pubmed.ncbi.nlm.nih.gov/27779568/
Marjoribanks J, Farquhar CM, Roberts H, Lethaby A. Cochrane corner: long-term hormone therapy for perimenopausal and postmenopausal women. Heart. 2018;104(2):93–5. https://pubmed.ncbi.nlm.nih.gov/28739806/
Marjoribanks J, Farquhar C, Roberts H, Lethaby A, Lee J. Long-term hormone therapy for perimenopausal and postmenopausal women. Cochrane Database Syst Rev. 2017;1:CD004143. https://pubmed.ncbi.nlm.nih.gov/28093732/
Marjoribanks J, Farquhar CM, Roberts H, Lethaby A. Cochrane corner: long-term hormone therapy for perimenopausal and postmenopausal women. Heart. 2018;104(2):93–5. https://pubmed.ncbi.nlm.nih.gov/28739806/
Manson JE, Bassuk SS, Kaunitz AM, Pinkerton JV. The Women’s Health Initiative trials of menopausal hormone therapy: lessons learned. Menopause. 2020;27(8):918–28. https://pubmed.ncbi.nlm.nih.gov/32345788/
Kim JJ, Chapman-Davis E. Role of progesterone in endometrial cancer. Semin Reprod Med. 2010;28(1):81–90. https://pubmed.ncbi.nlm.nih.gov/20104432/
Maclennan AH, Broadbent JL, Lester S, Moore V. Oral oestrogen and combined oestrogen/progestogen therapy versus placebo for hot flushes. Cochrane Database Syst Rev. 2004;(4):CD002978. https://pubmed.ncbi.nlm.nih.gov/15495039/
Marjoribanks J, Farquhar CM, Roberts H, Lethaby A. Cochrane corner: long-term hormone therapy for perimenopausal and postmenopausal women. Heart. 2018;104(2):93–5. https://pubmed.ncbi.nlm.nih.gov/28739806/
Pinkerton JV. Hormone therapy for postmenopausal women. N Engl J Med. 2020;382(5):446–5. https://pubmed.ncbi.nlm.nih.gov/31995690/
Chew F, Wu X. Sources of information influencing the state-of-the-science gap in hormone replacement therapy usage. PLoS One. 2017;12(2):e0171189. https://pubmed.ncbi.nlm.nih.gov/28158240/
Bhavnani BR, Stanczyk FZ. Pharmacology of conjugated equine estrogens: efficacy, safety and mechanism of action. J Steroid Biochem Mol Biol. 2014;142:16–29. https://pubmed.ncbi.nlm.nih.gov/24176763/
ClinCalc DrugStats database. The top 300 of 2019. ClinCalc.com. https://clincalc.com/DrugStats/Top300Drugs.aspx. Updated September 12, 2021. Accessed May 5, 2022.; https://clincalc.com/DrugStats/Top300Drugs.aspx
Kling J. The strange case of Premarin. Mod Drug Discov. 2000;3(8):46–52. https://pubsapp.acs.org/subscribe/archive/mdd/v03/i08/html/kling.html
Pinkerton JV. Hormone therapy for postmenopausal women. N Engl J Med. 2020;382(5):446–5. https://pubmed.ncbi.nlm.nih.gov/31995690/
ACOG Practice Bulletin No. 141: management of menopausal symptoms. Obstet Gynecol. 2014;123(1):202–16. https://pubmed.ncbi.nlm.nih.gov/24463691/
Pinkerton JV. Hormone therapy for postmenopausal women. N Engl J Med. 2020;382(5):446–5. https://pubmed.ncbi.nlm.nih.gov/31995690/
Brawley OW, O’Regan RM. Breast cancer screening: time for rational discourse. Cancer. 2014;120(18):2800–2. https://pubmed.ncbi.nlm.nih.gov/24925095/
Biller-Andorno N, Jüni P. Abolishing mammography screening programs? A view from the Swiss Medical Board. N Engl J Med. 2014;370(21):1965–7. https://pubmed.ncbi.nlm.nih.gov/24738641/
Nelson AL. Controversies regarding mammography, breast self-examination, and clinical breast examination. Obstet Gynecol Clin North Am. 2013;40(3):413–27. https://pubmed.ncbi.nlm.nih.gov/24021250/
Loh KP, Stefan MS, Friderici J, et al. Healthcare professionals’ perceptions and knowledge of the USPSTF guidelines on breast self-examination. South Med J. 2015;108(8):459–62. https://pubmed.ncbi.nlm.nih.gov/26280768/
Welch HG. Screening mammography – a long run for a short slide? N Engl J Med. 2010;363(13):1276–8. https://pubmed.ncbi.nlm.nih.gov/20860510/
Gigerenzer G. Women’s perception of the benefit of breast cancer screening. Maturitas. 2010;67(1):5–6. https://pubmed.ncbi.nlm.nih.gov/20609537/
Atkins CD. Potential hazards of mammography. J Clin Oncol. 2007;25(5):604. https://pubmed.ncbi.nlm.nih.gov/17290073/
Barratt A. Overdiagnosis in mammography screening: a 45 year journey from shadowy idea to acknowledged reality. BMJ. 2015;350:h867. https://pubmed.ncbi.nlm.nih.gov/25736426/
Gøtzsche PC, Jørgensen KJ, Zahl PH, Mæhlen J. Why mammography screening has not lived up to expectations from the randomised trials. Cancer Causes Control. 2012;23(1):15–21. https://pubmed.ncbi.nlm.nih.gov/22072221/
Pace LE, Keating NL. A systematic assessment of benefits and risks to guide breast cancer screening decisions. JAMA. 2014;311(13):1327–35. https://pubmed.ncbi.nlm.nih.gov/24691608/
Gøtzsche PC, Jørgensen KJ, Zahl PH, Mæhlen J. Why mammography screening has not lived up to expectations from the randomised trials. Cancer Causes Control. 2012;23(1):15–21. https://pubmed.ncbi.nlm.nih.gov/22072221/
Sohn E. Screening: don’t look now. Nature. 2015;527(7578):S118–9. https://pubmed.ncbi.nlm.nih.gov/26580162/
Gotzsche P. Commentary: screening: a seductive paradigm that has generally failed us. Int J Epidemiol. 2015;44(1):278–80. https://pubmed.ncbi.nlm.nih.gov/25596213/
Derbyshire SWG. Second opinion: doctors, diseases and decisions in modern medicine. BMJ. 2003;327(7411):399. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1126826/
Jørgensen KJ, Gøtzsche PC. The background review for the USPSTF recommendation on screening for breast cancer. Ann Intern Med. 2010;152(8):538; author reply 538–9. https://pubmed.ncbi.nlm.nih.gov/20157097/
Domenighetti G, D’Avanzo B, Egger M, et al. Women’s perception of the benefits of mammography screening: population-based survey in four countries. Int J Epidemiol. 2003;32(5):816–21. https://pubmed.ncbi.nlm.nih.gov/14559757/
Virani SS, Alonso A, Aparicio HJ, et al. Heart disease and stroke statistics—2021 update: a report from the American Heart Association. Circulation. 2021;143(8):e254–743. https://pubmed.ncbi.nlm.nih.gov/33501848/
National Cancer Institute Surveillance, Epidemiology, and End Results Program. Cancer stat facts: female breast cancer. https://seer.cancer.gov/statfacts/html/breast.html. Accessed May 5, 2022.; https://seer.cancer.gov/statfacts/html/breast.html
Parise CA, Caggiano V. Breast cancer survival defined by the ER/PR/HER2 subtypes and a surrogate classification according to tumor grade and immunohistochemical biomarkers. J Cancer Epidemiol. 2014;2014:469251. https://pubmed.ncbi.nlm.nih.gov/24955090/
Romanos-Nanclares A, Willett WC, Rosner BA, et al. Healthful and unhealthful plant-based diets and risk of breast cancer in U.S. women: results from the Nurses’ Health Studies. Cancer Epidemiol Biomarkers Prev. 2021;30(10):1921–31. https://pubmed.ncbi.nlm.nih.gov/34289970/
Link LB, Canchola AJ, Bernstein L, et al. Dietary patterns and breast cancer risk in the California Teachers Study cohort. Am J Clin Nutr. 2013;98(6):1524–32. https://pubmed.ncbi.nlm.nih.gov/24108781/
Hankinson SE. Circulating levels of sex steroids and prolactin in premenopausal women and risk of breast cancer. In: Li JJ, Li SA, Mohla S, Rochefort H, Maudelonde T, eds. Hormonal Carcinogenesis V. Springer; 2008:161–9. https://pubmed.ncbi.nlm.nih.gov/18497040/
Key T, Appleby P, Barnes I, et al. Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. J Natl Cancer Inst. 2002;94(8):606–16. https://pubmed.ncbi.nlm.nih.gov/11959894/
Hankinson SE. Circulating levels of sex steroids and prolactin in premenopausal women and risk of breast cancer. In: Li JJ, Li SA, Mohla S, Rochefort H, Maudelonde T, eds. Hormonal Carcinogenesis V. Springer; 2008:161–9. https://pubmed.ncbi.nlm.nih.gov/18497040/
Cleary MP, Grossmann ME. Minireview: obesity and breast cancer: the estrogen connection. Endocrinology. 2009;150(6):2537–42. https://pubmed.ncbi.nlm.nih.gov/19372199/
Shultz TD, Leklem JE. Nutrient intake and hormonal status of premenopausal vegetarian Seventh-day Adventists and premenopausal nonvegetarians. Nutr Cancer. 1983;4(4):247–59. https://pubmed.ncbi.nlm.nih.gov/6224137/
Barbosa JC, Shultz TD, Filley SJ, Nieman DC. The relationship among adiposity, diet, and hormone concentrations in vegetarian and nonvegetarian postmenopausal women. Am J Clin Nutr. 1990;51(5):798–803. https://pubmed.ncbi.nlm.nih.gov/2159209/
Shultz TD, Howie BJ. In vitro binding of steroid hormones by natural and purified fibers. Nutr Cancer. 1986;8(2):141–7. https://pubmed.ncbi.nlm.nih.gov/3010251/
Goldin BR, Woods MN, Spiegelman DL, et al. The effect of dietary fat and fiber on serum estrogen concentrations in premenopausal women under controlled dietary conditions. Cancer. 1994;74(3 Suppl):1125–31. https://pubmed.ncbi.nlm.nih.gov/8039147/
Jew S, AbuMweis SS, Jones PJH. Evolution of the human diet: linking our ancestral diet to modern functional foods as a means of chronic disease prevention. J Med Food. 2009;12(5):925–34. https://pubmed.ncbi.nlm.nih.gov/19857053/
Goldin BR, Adlercreutz H, Dwyer JT, Swenson L, Warram JH, Gorbach SL. Effect of diet on excretion of estrogens in pre– and postmenopausal women. Cancer Res. 1981;41(9 Pt 2):3771–3. https://pubmed.ncbi.nlm.nih.gov/7260944/
Goldin BR, Adlercreutz H, Gorbach SL, et al. Estrogen excretion patterns and plasma levels in vegetarian and omnivorous women. N Engl J Med. 1982;307(25):1542–7. https://pubmed.ncbi.nlm.nih.gov/7144835/
Beezhold B, Radnitz C, McGrath RE, Feldman A. Vegans report less bothersome vasomotor and physical menopausal symptoms than omnivores. Maturitas. 2018;112:12–7. https://pubmed.ncbi.nlm.nih.gov/29704911/
Beezhold B, Radnitz C, McGrath RE, Feldman A. Vegans report less bothersome vasomotor and physical menopausal symptoms than omnivores. Maturitas. 2018;112:12–7. https://pubmed.ncbi.nlm.nih.gov/29704911/
Noll PRES, Campos CAS, Leone C, et al. Dietary intake and menopausal symptoms in postmenopausal women: a systematic review. Climacteric. 2021;24(2):128–38. https://pubmed.ncbi.nlm.nih.gov/33112163/
Cagnacci A, Cannoletta M, Palma F, Bellafronte M, Romani C, Palmieri B. Relation between oxidative stress and climacteric symptoms in early postmenopausal women. Climacteric. 2015;18(4):631–6. https://pubmed.ncbi.nlm.nih.gov/25536006/
Aslani Z, Abshirini M, Heidari-Beni M, et al. Dietary inflammatory index and dietary energy density are associated with menopausal symptoms in postmenopausal women: a cross-sectional study. Menopause. 2020;27(5):568–78. https://pubmed.ncbi.nlm.nih.gov/32068687/
Minkin MJ. Menopause: hormones, lifestyle, and optimizing aging. Obstet Gynecol Clin North Am. 2019;46(3):501–14. https://pubmed.ncbi.nlm.nih.gov/31378291/
Woyka J. Consensus statement for non-hormonal-based treatments for menopausal symptoms. Post Reprod Health. 2017;23(2):71–5. https://pubmed.ncbi.nlm.nih.gov/28643614/
Prentice RL, Howard BV, Van Horn L, et al. Nutritional epidemiology and the Women’s Health Initiative: a review. Am J Clin Nutr. 2021;113(5):1083–92. https://pubmed.ncbi.nlm.nih.gov/33876183/
Patterson RE, Kristal A, Rodabough R, et al. Changes in food sources of dietary fat in response to an intensive low-fat dietary intervention: early results from the Women’s Health Initiative. J Am Diet Assoc. 2003;103(4):454–60. https://pubmed.ncbi.nlm.nih.gov/12669007/
Patterson RE, Prentice RL, Beresford S, et al. Dietary adherence in the Women’s Health Initiative dietary modification trial. J Am Diet Assoc. 2004;104(4):654–8. https://pubmed.ncbi.nlm.nih.gov/15054353/
Kroenke CH, Caan BJ, Stefanick ML, et al. Effects of a dietary intervention and weight change on vasomotor symptoms in the Women’s Health Initiative. Menopause. 2012;19(9):980–8. https://pubmed.ncbi.nlm.nih.gov/22781782/
Rotolo O, Zinzi I, Veronese N, et al. Women in LOVe: lacto-ovo-vegetarian diet rich in omega-3 improves vasomotor symptoms in postmenopausal women. An exploratory randomized controlled trial. Endocr Metab Immune Disord Drug Targets. 2019;19(8):1232–9. https://pubmed.ncbi.nlm.nih.gov/31132980/
Cetisli NE, Saruhan A, Kivcak B. The effects of flaxseed on menopausal symptoms and quality of life. Holist Nurs Pract. 2015;29(3):151–7. https://pubmed.ncbi.nlm.nih.gov/25882265/
Messina M. Soy and health update: evaluation of the clinical and epidemiologic literature. Nutrients. 2016;8(12):E754. https://pubmed.ncbi.nlm.nih.gov/27886135/
Thomas AJ, Ismail R, Taylor-Swanson L, et al. Effects of isoflavones and amino acid therapies for hot flashes and co-occurring symptoms during the menopausal transition and early postmenopause: a systematic review. Maturitas. 2014;78(4):263–76. https://pubmed.ncbi.nlm.nih.gov/24951101/
Avis NE, Crawford SL, Greendale G, et al. Duration of menopausal vasomotor symptoms over the menopause transition. JAMA Intern Med. 2015;175(4):531–9. https://pubmed.ncbi.nlm.nih.gov/25686030/
Avis NE, Kaufert PA, Lock M, McKinlay SM, Vass K. The evolution of menopausal symptoms. Baillieres Clin Endocrinol Metab. 1993;7(1):17–32. https://pubmed.ncbi.nlm.nih.gov/8435051/
Thomas AJ, Ismail R, Taylor-Swanson L, et al. Effects of isoflavones and amino acid therapies for hot flashes and co-occurring symptoms during the menopausal transition and early postmenopause: a systematic review. Maturitas. 2014;78(4):263–76. https://pubmed.ncbi.nlm.nih.gov/24951101/
Lock M. Contested meanings of the menopause. Lancet. 1991;337(8752):1270–2. https://pubmed.ncbi.nlm.nih.gov/1674073/
Avis NE, Kaufert PA, Lock M, McKinlay SM, Vass K. The evolution of menopausal symptoms. Baillieres Clin Endocrinol Metab. 1993;7(1):17–32. https://pubmed.ncbi.nlm.nih.gov/8435051/
Lock M. Contested meanings of the menopause. Lancet. 1991;337(8752):1270–2. https://pubmed.ncbi.nlm.nih.gov/1674073/
Lock M. Ambiguities of aging: Japanese experience and perceptions of menopause. Cult Med Psychiatry. 1986;10(1):23–46. https://pubmed.ncbi.nlm.nih.gov/3486095/
Avis NE, Stellato R, Crawford S, et al. Is there a menopausal syndrome? Menopausal status and symptoms across racial/ethnic groups. Soc Sci Med. 2001;52(3):345–56. https://pubmed.ncbi.nlm.nih.gov/11330770/
Taku K, Melby MK, Kronenberg F, Kurzer MS, Messina M. Extracted or synthesized soybean isoflavones reduce menopausal hot flash frequency and severity: systematic review and meta-analysis of randomized controlled trials. Menopause. 2012;19(7):776–90. https://pubmed.ncbi.nlm.nih.gov/22433977/
Ghazanfarpour M, Sadeghi R, Roudsari RL. The application of soy isoflavones for subjective symptoms and objective signs of vaginal atrophy in menopause: a systematic review of randomised controlled trials. J Obstet Gynaecol. 2016;36(2):160–71. https://pubmed.ncbi.nlm.nih.gov/26440219/
Lambert MNT, Hu LM, Jeppesen PB. A systematic review and meta-analysis of the effects of isoflavone formulations against estrogen-deficient bone resorption in peri– and postmenopausal women. Am J Clin Nutr. 2017;106(3):801–11. https://pubmed.ncbi.nlm.nih.gov/28768649/
Su BYW, Tung TH, Chien WH. Effects of phytoestrogens on depressive symptoms in climacteric women: a meta-analysis of randomized controlled trials. J Altern Complement Med. 2018;24(8):850–1. https://pubmed.ncbi.nlm.nih.gov/29717895/
Cheng PF, Chen JJ, Zhou XY, et al. Do soy isoflavones improve cognitive function in postmenopausal women? A meta-analysis. Menopause. 2015;22(2):198–206. https://pubmed.ncbi.nlm.nih.gov/25003621/
Schmidt M, Arjomand-Wölkart K, Birkhäuser MH, et al. Consensus: soy isoflavones as a first-line approach to the treatment of menopausal vasomotor complaints. Gynecol Endocrinol. 2016;32(6):427–30. https://pubmed.ncbi.nlm.nih.gov/26943176/
Welty FK, Lee KS, Lew NS, Nasca M, Zhou JR. The association between soy nut consumption and decreased menopausal symptoms. J Womens Health (Larchmt). 2007;16(3):361–9. https://pubmed.ncbi.nlm.nih.gov/17439381/
Barnard ND, Kahleova H, Holtz DN, et al. A dietary intervention for vasomotor symptoms of menopause: a randomized, controlled trial. Menopause. 2023;30(1):80–7. https://pubmed.ncbi.nlm.nih.gov/36253903/
Barnard ND, Kahleova H, Holtz DN, et al. The Women’s Study for the Alleviation of Vasomotor Symptoms (WAVS): a randomized, controlled trial of a plant-based diet and whole soybeans for postmenopausal women. Menopause. 2021;28(10):1150–6. https://pubmed.ncbi.nlm.nih.gov/34260478/
Buja A, Pierbon M, Lago L, Grotto G, Baldo V. Breast cancer primary prevention and diet: an umbrella review. Int J Environ Res Public Health. 2020;17(13):E4731. https://pubmed.ncbi.nlm.nih.gov/32630215/
Messina M, Messina VL. Exploring the soyfood controversy. Nutr Today. 2013;48(2):68. https://www.researchgate.net/publication/271683198_Exploring_the_Soyfood_Controversy
Nachvak SM, Moradi S, Anjom-Shoae J, et al. Soy, soy isoflavones, and protein intake in relation to mortality from all causes, cancers, and cardiovascular diseases: a systematic review and dose-response meta-analysis of prospective cohort studies. J Acad Nutr Diet. 2019;119(9):1483–1500.e17. https://pubmed.ncbi.nlm.nih.gov/31278047/
Kelsey JL. A review of the epidemiology of human breast cancer. Epidemiol Rev. 1979;1:74–109. https://pubmed.ncbi.nlm.nih.gov/398270/
Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33. https://pubmed.ncbi.nlm.nih.gov/33433946/
Fraser GE, Jaceldo-Siegl K, Orlich M, Mashchak A, Sirirat R, Knutsen S. Dairy, soy, and risk of breast cancer: those confounded milks. Int J Epidemiol. 2020;49(5):1526–37. https://pubmed.ncbi.nlm.nih.gov/32095830/
Shu XO, Zheng Y, Cai H, et al. Soy food intake and breast cancer survival. JAMA. 2009;302(22):2437–43. https://pubmed.ncbi.nlm.nih.gov/19996398/
Guha N, Kwan ML, Quesenberry CP, Weltzien EK, Castillo AL, Caan BJ. Soy isoflavones and risk of cancer recurrence in a cohort of breast cancer survivors: the Life After Cancer Epidemiology study. Breast Cancer Res Treat. 2009;118(2):395–405. https://pubmed.ncbi.nlm.nih.gov/19221874/
Kang X, Zhang Q, Wang S, Huang X, Jin S. Effect of soy isoflavones on breast cancer recurrence and death for patients receiving adjuvant endocrine therapy. CMAJ. 2010;182(17):1857–62. https://pubmed.ncbi.nlm.nih.gov/20956506/
Rock CL, Doyle C, Demark-Wahnefried W, et al. Nutrition and physical activity guidelines for cancer survivors. CA Cancer J Clin. 2012;62(4):243–74. https://pubmed.ncbi.nlm.nih.gov/22539238/
Caan BJ, Natarajan L, Parker B, et al. Soy food consumption and breast cancer prognosis. Cancer Epidemiol Biomarkers Prev. 2011;20(5):854–8. https://pubmed.ncbi.nlm.nih.gov/21357380/
Zhang YF, Kang HB, Li BL, Zhang RM. Positive effects of soy isoflavone food on survival of breast cancer patients in China. Asian Pac J Cancer Prev. 2012;13(2):479–82. https://pubmed.ncbi.nlm.nih.gov/22524810/
Chi F, Wu R, Zeng YC, Xing R, Liu Y, Xu ZG. Post-diagnosis soy food intake and breast cancer survival: a meta-analysis of cohort studies. Asian Pac J Cancer Prev. 2013;14(4):2407–12. https://pubmed.ncbi.nlm.nih.gov/23725149/
Chi F, Wu R, Zeng YC, Xing R, Liu Y, Xu ZG. Post-diagnosis soy food intake and breast cancer survival: a meta-analysis of cohort studies. Asian Pac J Cancer Prev. 2013;14(4):2407–12. https://pubmed.ncbi.nlm.nih.gov/23725149/
Kang HB, Zhang YF, Yang JD, Lu KL. Study on soy isoflavone consumption and risk of breast cancer and survival. Asian Pac J Cancer Prev. 2012;13(3):995–8. https://pubmed.ncbi.nlm.nih.gov/22631686/
Buck K, Zaineddin AK, Vrieling A, Linseisen J, Chang-Claude J. Meta-analyses of lignans and enterolignans in relation to breast cancer risk. Am J Clin Nutr. 2010;92(1):141–53. https://pubmed.ncbi.nlm.nih.gov/20463043/
McCann SE, Thompson LU, Nie J, et al. Dietary lignan intakes in relation to survival among women with breast cancer: the Western New York Exposures and Breast Cancer (WEB) Study. Breast Cancer Res Treat. 2010;122(1):229–35. https://pubmed.ncbi.nlm.nih.gov/20033482/
Thompson LU, Chen JM, Li T, Strasser-Weippl K, Goss PE. Dietary flaxseed alters tumor biological markers in postmenopausal breast cancer. Clin Cancer Res. 2005;11(10):3828–35. https://pubmed.ncbi.nlm.nih.gov/15897583/
Calado A, Neves PM, Santos T, Ravasco P. The effect of flaxseed in breast cancer: a literature review. Front Nutr. 2018;5:4. https://pubmed.ncbi.nlm.nih.gov/29468163/
Hadi A, Askarpour M, Salamat S, Ghaedi E, Symonds ME, Miraghajani M. Effect of flaxseed supplementation on lipid profile: an updated systematic review and dose-response meta-analysis of sixty-two randomized controlled trials. Pharmacol Res. 2020;152:104622. https://pubmed.ncbi.nlm.nih.gov/31899314/
Khandouzi N, Zahedmehr A, Mohammadzadeh A, Sanati HR, Nasrollahzadeh J. Effect of flaxseed consumption on flow-mediated dilation and inflammatory biomarkers in patients with coronary artery disease: a randomized controlled trial. Eur J Clin Nutr. 2019;73(2):258–65. https://pubmed.ncbi.nlm.nih.gov/30127374/
Ursoniu S, Sahebkar A, Andrica F, et al. Effects of flaxseed supplements on blood pressure: a systematic review and meta-analysis of controlled clinical trial. Clin Nutr. 2016;35(3):615–25. https://pubmed.ncbi.nlm.nih.gov/26071633/
Khandouzi N, Zahedmehr A, Mohammadzadeh A, Sanati HR, Nasrollahzadeh J. Effect of flaxseed consumption on flow-mediated dilation and inflammatory biomarkers in patients with coronary artery disease: a randomized controlled trial. Eur J Clin Nutr. 2019;73(2):258–65. https://pubmed.ncbi.nlm.nih.gov/30127374/
Hadi A, Askarpour M, Ziaei R, Venkatakrishnan K, Ghaedi E, Ghavami A. Impact of flaxseed supplementation on plasma lipoprotein(A) concentrations: a systematic review and meta-analysis of randomized controlled trials. Phytother Res. 2020;34(7):1599–608. https://pubmed.ncbi.nlm.nih.gov/32073724/
Almehmadi A, Lightowler H, Chohan M, Clegg ME. The effect of a split portion of flaxseed on 24-h blood glucose response. Eur J Nutr. 2021;60(3):1363–73. https://pubmed.ncbi.nlm.nih.gov/32699911/
Ghazanfarpour M, Sadeghi R, Latifnejad Roudsari R, et al. Effects of flaxseed and Hypericum perforatum on hot flash, vaginal atrophy and estrogen-dependent cancers in menopausal women: a systematic review and meta-analysis. Avicenna J Phytomed. 2016;6(3):273–83. https://pubmed.ncbi.nlm.nih.gov/27462550/
Franco OH, Chowdhury R, Troup J, et al. Use of plant-based therapies and menopausal symptoms: a systematic review and meta-analysis. JAMA. 2016;315(23):2554–63. https://pubmed.ncbi.nlm.nih.gov/27327802/
Milligan SR, Kalita JC, Heyerick A, Rong H, De Cooman L, De Keukeleire D. Identification of a potent phytoestrogen in hops (Humulus lupulus L.) and beer. J Clin Endocrinol Metab. 1999;84(6):2249–52. https://pubmed.ncbi.nlm.nih.gov/10372741/
Milligan S, Kalita J, Pocock V, et al. Oestrogenic activity of the hop phyto-oestrogen, 8-prenylnaringenin. Reproduction. 2002;123(2):235–42. https://pubmed.ncbi.nlm.nih.gov/11866690/
Bradbury RB, White DE. 761. The chemistry of subterranean clover. Part I. Isolation of formononetin and genistein. J Chem Soc. 1951;(0):3447–9. https://pubs.rsc.org/en/content/articlelanding/1951/jr/jr9510003447
Gavaler JS, Rosenblum ER, Deal SR, Bowie BT. The phytoestrogen congeners of alcoholic beverages: current status. Proc Soc Exp Biol Med. 1995;208(1):98–102. https://pubmed.ncbi.nlm.nih.gov/7892304/
Pedrera-Zamorano JD, Lavado-Garcia JM, Roncero-Martin R, Calderon-Garcia JF, Rodriguez-Dominguez T, Canal-Macias ML. Effect of beer drinking on ultrasound bone mass in women. Nutrition. 2009;25(10):1057–63. https://pubmed.ncbi.nlm.nih.gov/19527924/
Aghamiri V, Mirghafourvand M, Mohammad-Alizadeh-Charandabi S, Nazemiyeh H. The effect of Hop (Humulus lupulus L.) on early menopausal symptoms and hot flashes: a randomized placebo-controlled trial. Complement Ther Clin Pract. 2016;23:130–5. https://pubmed.ncbi.nlm.nih.gov/25982391/
Schaefer O, Hümpel M, Fritzemeier KH, Bohlmann R, Schleuning WD. 8-Prenyl naringenin is a potent ERa selective phytoestrogen present in hops and beer. J Steroid Biochem Mol Biol. 2003;84(2–3):359–60. https://pubmed.ncbi.nlm.nih.gov/12711023/
Fugh-Berman A. “Bust enhancing” herbal products. Obstet Gynecol. 2003;101(6):1345–9. https://pubmed.ncbi.nlm.nih.gov/12798545/
Lê MG, Hill C, Kramar A, Flamanti R. Alcoholic beverage consumption and breast cancer in a French case-control study. Am J Epidemiol. 1984;120(3):350–7. https://pubmed.ncbi.nlm.nih.gov/6475912/
Salehi-Pourmehr H, Ostadrahimi A, Ebrahimpour-Mirzarezaei M, Farshbaf-Khalili A. Does aromatherapy with lavender affect physical and psychological symptoms of menopausal women? A systematic review and meta-analysis. Complement Ther Clin Pract. 2020;39:101150. https://pubmed.ncbi.nlm.nih.gov/32379682/
Kazemzadeh R, Nikjou R, Rostamnegad M, Norouzi H. Effect of lavender aromatherapy on menopause hot flushing: a crossover randomized clinical trial. J Chin Med Assoc. 2016;79(9):489–92. https://pubmed.ncbi.nlm.nih.gov/27388435/
Nikjou R, Kazemzadeh R, Asadzadeh F, Fathi R, Mostafazadeh F. The effect of lavender aromatherapy on the symptoms of menopause. J Natl Med Assoc. 2018;110(3):265–9. https://pubmed.ncbi.nlm.nih.gov/29778129/
Dos Reis Lucena L, Dos Santos-Junior JG, Tufik S, Hachul H. Lavender essential oil on postmenopausal women with insomnia: double-blind randomized trial. Complement Ther Med. 2021;59:102726. https://pubmed.ncbi.nlm.nih.gov/33905827/
Donelli D, Antonelli M, Bellinazzi C, Gensini GF, Firenzuoli F. Effects of lavender on anxiety: a systematic review and meta-analysis. Phytomedicine. 2019;65:153099. https://pubmed.ncbi.nlm.nih.gov/31655395/
Farshbaf-Khalili A, Kamalifard M, Namadian M. Comparison of the effect of lavender and bitter orange on anxiety in postmenopausal women: a triple-blind, randomized, controlled clinical trial. Complement Ther Clin Pract. 2018;31:132–8. https://pubmed.ncbi.nlm.nih.gov/29705445/
Kamalifard M, Farshbaf-Khalili A, Namadian M, Ranjbar Y, Herizchi S. Comparison of the effect of lavender and bitter orange on sleep quality in postmenopausal women: a triple-blind, randomized, controlled clinical trial. Women Health. 2018;58(8):851–65. https://pubmed.ncbi.nlm.nih.gov/28749734/
Latiff LA, Parhizkar S, Dollah MA, Hassan STS. Alternative supplement for enhancement of reproductive health and metabolic profile among perimenopausal women: a novel role of Nigella sativa. Iran J Basic Med Sci. 2014;17(12):980–5. https://pubmed.ncbi.nlm.nih.gov/25859301/
Rahimi Kian F, Bekhradi R, Rahimi R, Golzareh P, Mehran A. Evaluating the effect of fennel soft capsules on the quality of life and its different aspects in menopausal women: a randomized clinical trial. Nurs Pract Today. 2017;4(2):87–95. https://www.researchgate.net/publication/319183067_Evaluating_the_effect_of_fennel_soft_capsules_on_the_quality_of_life_and_its_different_aspects_in_menopausal_women_A_randomized_clinical_trial_ARTICLE_INFO_ABSTRACT
Khan TM, Wu DBC, Dolzhenko AV. Effectiveness of fenugreek as a galactagogue: a network meta-analysis. Phytother Res. 2018;32(3):402–12. https://pubmed.ncbi.nlm.nih.gov/29193352/
Hakimi S, Charandabi SMA, Shadbad MRS, et al. Effect of Fenugreek seed on early menopausal symptoms. Pharm Sci. 2005;(2):83–90. https://www.researchgate.net/publication/236619683_Effect_of_Fenugreek_seed_on_early_menopausal_symptoms
Tariq SH, Haren MT, Kim MJ, Morley JE. Andropause: is the emperor wearing any clothes? Rev Endocr Metab Disord. 2005;6(2):77–84. https://pubmed.ncbi.nlm.nih.gov/15843878/
Saad F, Gooren LJ. Late onset hypogonadism of men is not equivalent to the menopause. Maturitas. 2014;79(1):52–7. https://pubmed.ncbi.nlm.nih.gov/25042874/
Tariq SH, Haren MT, Kim MJ, Morley JE. Andropause: is the emperor wearing any clothes? Rev Endocr Metab Disord. 2005;6(2):77–84. https://pubmed.ncbi.nlm.nih.gov/15843878/
Swee DS, Gan EH. Late-onset hypogonadism as primary testicular failure. Front Endocrinol (Lausanne). 2019;10:372. https://pubmed.ncbi.nlm.nih.gov/31244778/
Vitry AI, Mintzes B. Disease mongering and low testosterone in men: the tale of two regulatory failures. Med J Aust. 2012;196(10):619–21. https://pubmed.ncbi.nlm.nih.gov/22676868/
Schwartz LM, Woloshin S. Low “T” as in “template”: how to sell disease. JAMA Intern Med. 2013;173(15):1460–2. https://pubmed.ncbi.nlm.nih.gov/23939516/
Perls T, Handelsman DJ. Disease mongering of age-associated declines in testosterone and growth hormone levels. J Am Geriatr Soc. 2015;63(4):809–11. https://pubmed.ncbi.nlm.nih.gov/25809947/
Eder S. Many players in the concerto of sex. Science. 2007;315(5817):1370. https://www.science.org/doi/abs/10.1126/science.1139058
Shomali ME. The use of anti-aging hormones. Melatonin, growth hormone, testosterone, and dehydroepiandrosterone: consumer enthusiasm for unproven therapies. Md Med J. 1997;46(4):181–6. https://pubmed.ncbi.nlm.nih.gov/9114695/
Rengachary SS, Colen C, Guthikonda M. Charles-Édouard Brown-Séquard: an eccentric genius. Neurosurgery. 2008;62(4):954–64. https://pubmed.ncbi.nlm.nih.gov/18496202/
Kim MJ, Morley JE. The hormonal fountains of youth: myth or reality? J Endocrinol Invest. 2005;28(11 Suppl Proceedings):5–14. https://pubmed.ncbi.nlm.nih.gov/16760618/
Swee DS, Gan EH. Late-onset hypogonadism as primary testicular failure. Front Endocrinol (Lausanne). 2019;10:372. https://pubmed.ncbi.nlm.nih.gov/31244778/
Sartorius G, Spasevska S, Idan A, et al. Serum testosterone, dihydrotestosterone and estradiol concentrations in older men self-reporting very good health: the healthy man study. Clin Endocrinol (Oxf). 2012;77(5):755–63. https://pubmed.ncbi.nlm.nih.gov/22563890/
Saad F, Gooren LJ. Late onset hypogonadism of men is not equivalent to the menopause. Maturitas. 2014;79(1):52–7. https://pubmed.ncbi.nlm.nih.gov/25042874/
Busnelli A, Somigliana E, Vercellini P. ‘Forever young’—testosterone replacement therapy: a blockbuster drug despite flabby evidence and broken promises. Human Reproduction. 2017;32(4):719–24. https://pubmed.ncbi.nlm.nih.gov/28333214/
Handelsman DJ. Irrational exuberance in testosterone prescribing: when will the bubble burst? Med Care. 2015;53(9):743–5. https://pubmed.ncbi.nlm.nih.gov/26270825/
Perls T, Handelsman DJ. Disease mongering of age-associated declines in testosterone and growth hormone levels. J Am Geriatr Soc. 2015;63(4):809–11. https://pubmed.ncbi.nlm.nih.gov/25809947/
Vitry AI, Mintzes B. Disease mongering and low testosterone in men: the tale of two regulatory failures. Med J Aust. 2012;196(10):619–21. https://pubmed.ncbi.nlm.nih.gov/22676868/
Mintzes B. The marketing of testosterone treatments for age-related low testosterone or “Low T.” Curr Opin Endocrinol Diabetes Obes. 2018;25(3):224–30. https://pubmed.ncbi.nlm.nih.gov/29570470/
Morley JE, Perry HM III, Kevorkian RT, Patrick P. Comparison of screening questionnaires for the diagnosis of hypogonadism. Maturitas. 2006;53(4):424–9. https://pubmed.ncbi.nlm.nih.gov/16140484/
Vitry AI, Mintzes B. Disease mongering and low testosterone in men: the tale of two regulatory failures. Med J Aust. 2012;196(10):619–21. https://pubmed.ncbi.nlm.nih.gov/22676868/
Dunn M, Mulrooney KJ, Forlini C, van de Ven K, Underwood M. The pharmaceuticalisation of “healthy” ageing: testosterone enhancement for longevity. Int J Drug Policy. 2021;95:103159. https://pubmed.ncbi.nlm.nih.gov/33583680/
Fugh-Berman A, Hogenmiller A. CME stands for commercial medical education: and ACCME still won’t address the issue. J Med Ethics. 2016;42(3):172–3. https://pubmed.ncbi.nlm.nih.gov/26676848/
Handelsman DJ. Irrational exuberance in testosterone prescribing: when will the bubble burst? Med Care. 2015;53(9):743–5. https://pubmed.ncbi.nlm.nih.gov/26270825/
Shores MM, Matsumoto AM. Testosterone, aging and survival: biomarker or deficiency. Curr Opin Endocrinol Diabetes Obes. 2014;21(3):209–16. https://pubmed.ncbi.nlm.nih.gov/24722173/
Perls T, Handelsman DJ. Disease mongering of age-associated declines in testosterone and growth hormone levels. J Am Geriatr Soc. 2015;63(4):809–11. https://pubmed.ncbi.nlm.nih.gov/25809947/
Al-Sharefi A, Quinton R. Current national and international guidelines for the management of male hypogonadism: helping clinicians to navigate variation in diagnostic criteria and treatment recommendations. Endocrinol Metab (Seoul). 2020;35(3):526–40. https://pubmed.ncbi.nlm.nih.gov/32981295/
Mok SF, Fennell C, Savkovic S, et al. Testosterone for androgen deficiency – like symptoms in men without pathologic hypogonadism: a randomized, placebo-controlled cross-over with masked choice extension clinical trial. J Gerontol A Biol Sci Med Sci. 2020;75(9):1723–31. https://pubmed.ncbi.nlm.nih.gov/31425577/
Bandari J, Ayyash OM, Emery SL, Wessel CB, Davies BJ. Marketing and testosterone treatment in the USA: a systematic review. Eur Urol Focus. 2017;3(4–5):395–402. https://pubmed.ncbi.nlm.nih.gov/29174614/
Huo S, Scialli AR, McGarvey S, et al. Treatment of men for “low testosterone”: a systematic review. PLoS One. 2016;11(9):e0162480. https://pubmed.ncbi.nlm.nih.gov/27655114/
Bandari J, Ayyash OM, Emery SL, Wessel CB, Davies BJ. Marketing and testosterone treatment in the USA: a systematic review. Eur Urol Focus. 2017;3(4–5):395–402. https://pubmed.ncbi.nlm.nih.gov/29174614/
Irwig MS, Fleseriu M, Jonklaas J, et al. Off-label use and misuse of testosterone, growth hormone, thyroid hormone, and adrenal supplements: risks and costs of a growing problem. Endocr Pract. 2020;26(3):340–53. https://pubmed.ncbi.nlm.nih.gov/32163313/
Wu FCW, Tajar A, Beynon JM, et al. Identification of late-onset hypogonadism in middle-aged and elderly men. N Engl J Med. 2010;363(2):123–35. https://pubmed.ncbi.nlm.nih.gov/20554979/
Miner M, Barkin J, Rosenberg MT. Testosterone deficiency: myth, facts, and controversy. Can J Urol. 2014;21(Suppl 2):39–54. https://pubmed.ncbi.nlm.nih.gov/24978631/
Salter CA, Mulhall JP. Guideline of guidelines: testosterone therapy for testosterone deficiency. BJU Int. 2019;124(5):722–9. https://pubmed.ncbi.nlm.nih.gov/31420972/
Grossmann M. Serum testosterone concentrations in older men: one size does not fit all. J Clin Endocrinol Metab. 2020;105(8):dgaa400. https://pubmed.ncbi.nlm.nih.gov/32584983/
Giagulli VA, Castellana M, Lisco G, Triggiani V. Critical evaluation of different available guidelines for late-onset hypogonadism. Andrology. 2020;8(6):1628–41. https://pubmed.ncbi.nlm.nih.gov/32593233/
Bandari J, Ayyash OM, Emery SL, Wessel CB, Davies BJ. Marketing and testosterone treatment in the USA: a systematic review. Eur Urol Focus. 2017;3(4–5):395–402. https://pubmed.ncbi.nlm.nih.gov/29174614/
Fugh-Berman A. Treating aging with testosterone. Am Fam Physician. 2017;96(7):428–30. https://pubmed.ncbi.nlm.nih.gov/29094915/
The truth about testosterone replacement therapy. Fountain of youth – or harbinger of health woes? Johns Hopkins Med Lett Health After 50. 2014;29(5):1–2. https://pubmed.ncbi.nlm.nih.gov/25097952/
Sansone A, Sansone M, Lenzi A, Romanelli F. Testosterone replacement therapy: the emperor’s new clothes. Rejuvenation Res. 2017;20(1):9–14. https://pubmed.ncbi.nlm.nih.gov/27124096/
Layton JB, Li D, Meier CR, et al. Testosterone lab testing and initiation in the United Kingdom and the United States, 2000 to 2011. J Clin Endocrinol Metab. 2014;99(3):835–42. https://pubmed.ncbi.nlm.nih.gov/24423353/
Bandari J, Ayyash OM, Emery SL, Wessel CB, Davies BJ. Marketing and testosterone treatment in the USA: a systematic review. Eur Urol Focus. 2017;3(4–5):395–402. https://pubmed.ncbi.nlm.nih.gov/29174614/
Busnelli A, Somigliana E, Vercellini P. ‘Forever young’—testosterone replacement therapy: a blockbuster drug despite flabby evidence and broken promises. Human Reproduction. 2017;32(4):719–24. https://pubmed.ncbi.nlm.nih.gov/28333214/
Tariq SH, Haren MT, Kim MJ, Morley JE. Andropause: is the emperor wearing any clothes? Rev Endocr Metab Disord. 2005;6(2):77–84. https://pubmed.ncbi.nlm.nih.gov/15843878/
Mok SF, Fennell C, Savkovic S, et al. Testosterone for androgen deficiency – like symptoms in men without pathologic hypogonadism: a randomized, placebo-controlled cross-over with masked choice extension clinical trial. J Gerontol A Biol Sci Med Sci. 2020;75(9):1723–31. https://pubmed.ncbi.nlm.nih.gov/31425577/
Fox CA, Ismail AA, Love DN, Kirkham KE, Loraine JA. Studies on the relationship between plasma testosterone levels and human sexual activity. J Endocrinol. 1972;52(1):51–8. https://pubmed.ncbi.nlm.nih.gov/5061159/
Effects of sexual activity on beard growth in man. Nature. 1970;226(5248):869–70. https://pubmed.ncbi.nlm.nih.gov/5444635/
Jannini EA, Screponi E, Carosa E, et al. Lack of sexual activity from erectile dysfunction is associated with a reversible reduction in serum testosterone. Int J Androl. 1999;22(6):385–92. https://pubmed.ncbi.nlm.nih.gov/10624607/
Dabbs JM, Mohammed S. Male and female salivary testosterone concentrations before and after sexual activity. Physiol Behav. 1992;52(1):195–7. https://pubmed.ncbi.nlm.nih.gov/1529008/
Mok SF, Fennell C, Savkovic S, et al. Testosterone for androgen deficiency – like symptoms in men without pathologic hypogonadism: a randomized, placebo-controlled cross-over with masked choice extension clinical trial. J Gerontol A Biol Sci Med Sci. 2020;75(9):1723–31. https://pubmed.ncbi.nlm.nih.gov/31425577/
Handelsman DJ. Testosterone and male aging: faltering hope for rejuvenation. JAMA. 2017;317(7):699–701. https://pubmed.ncbi.nlm.nih.gov/28241336/
Corona G, Guaraldi F, Rastrelli G, Sforza A, Maggi M. Testosterone deficiency and risk of cognitive disorders in aging males. World J Mens Health. 2021;39(1):9–18. https://pubmed.ncbi.nlm.nih.gov/32378366/
Zhang Z, Kang D, Li H. Testosterone and cognitive impairment or dementia in middle-aged or aging males: causation and intervention, a systematic review and meta-analysis. J Geriatr Psychiatry Neurol. 2021;34(5):405–17. https://pubmed.ncbi.nlm.nih.gov/32602403/
Sari Motlagh R, Quhal F, Mori K, et al. The risk of new onset dementia and/or Alzheimer disease among patients with prostate cancer treated with androgen deprivation therapy: a systematic review and meta-analysis. J Urol. 2021;205(1):60–7. https://pubmed.ncbi.nlm.nih.gov/32856962/
Resnick SM, Matsumoto AM, Stephens-Shields AJ, et al. Testosterone treatment and cognitive function in older men with low testosterone and age-associated memory impairment. JAMA. 2017;317(7):717–27. https://pubmed.ncbi.nlm.nih.gov/28241356/
Corona G, Guaraldi F, Rastrelli G, Sforza A, Maggi M. Testosterone deficiency and risk of cognitive disorders in aging males. World J Mens Health. 2021;39(1):9–18. https://pubmed.ncbi.nlm.nih.gov/32378366/
Handelsman DJ. Testosterone and male aging: faltering hope for rejuvenation. JAMA. 2017;317(7):699–701. https://pubmed.ncbi.nlm.nih.gov/28241336/
Snyder PJ, Bhasin S, Cunningham GR, et al. Effects of testosterone treatment in older men. N Engl J Med. 2016;374(7):611–24. https://pubmed.ncbi.nlm.nih.gov/26886521/
Diem SJ, Greer NL, MacDonald R, et al. Efficacy and safety of testosterone treatment in men: an evidence report for a clinical practice guideline by the American College of Physicians. Ann Intern Med. 2020;172(2):105–18. https://pubmed.ncbi.nlm.nih.gov/31905375/
Bandari J, Ayyash OM, Emery SL, Wessel CB, Davies BJ. Marketing and testosterone treatment in the USA: a systematic review. Eur Urol Focus. 2017;3(4–5):395–402. https://pubmed.ncbi.nlm.nih.gov/29174614/
Huo S, Scialli AR, McGarvey S, et al. Treatment of men for “low testosterone”: a systematic review. PLoS One. 2016;11(9):e0162480. https://pubmed.ncbi.nlm.nih.gov/27655114/
Snyder PJ, Kopperdahl DL, Stephens-Shields AJ, et al. Effect of testosterone treatment on volumetric bone density and strength in older men with low testosterone: a controlled clinical trial. JAMA Intern Med. 2017;177(4):471–9. https://pubmed.ncbi.nlm.nih.gov/28241231/
Zhang Z, Kang D, Li H. The effects of testosterone on bone health in males with testosterone deficiency: a systematic review and meta-analysis. BMC Endocr Disord. 2020;20(1):33. https://pubmed.ncbi.nlm.nih.gov/32145741/
Snyder PJ, Bhasin S, Cunningham GR, et al. Effects of testosterone treatment in older men. N Engl J Med. 2016;374(7):611–24. https://pubmed.ncbi.nlm.nih.gov/26886521/
Huo S, Scialli AR, McGarvey S, et al. Treatment of men for “low testosterone”: a systematic review. PLoS One. 2016;11(9):e0162480. https://pubmed.ncbi.nlm.nih.gov/27655114/
Maggi M, Filippi S, Vignozzi L, Rastrelli G. Controversial aspects of testosterone in the regulation of sexual function in late-onset hypogonadism. Andrology. 2020;8(6):1580–9. https://pubmed.ncbi.nlm.nih.gov/32248652/
Corona G, Rastrelli G, Morgentaler A, Sforza A, Mannucci E, Maggi M. Meta-analysis of results of testosterone therapy on sexual function based on international index of erectile function scores. Eur Urol. 2017;72(6):1000–11. https://pubmed.ncbi.nlm.nih.gov/28434676/
Maggi M, Filippi S, Vignozzi L, Rastrelli G. Controversial aspects of testosterone in the regulation of sexual function in late-onset hypogonadism. Andrology. 2020;8(6):1580–9. https://pubmed.ncbi.nlm.nih.gov/32248652/
Kwan M, Greenleaf WJ, Mann J, Crapo L, Davidson JM. The nature of androgen action on male sexuality: a combined laboratory-self-report study on hypogonadal men. J Clin Endocrinol Metab. 1983;57(3):557–62. https://pubmed.ncbi.nlm.nih.gov/6874890/
Maggi M, Filippi S, Vignozzi L, Rastrelli G. Controversial aspects of testosterone in the regulation of sexual function in late-onset hypogonadism. Andrology. 2020;8(6):1580–9. https://pubmed.ncbi.nlm.nih.gov/32248652/
Adlin EV. Age-related low testosterone. Ann Intern Med. 2020;172(2):151–2. https://pubmed.ncbi.nlm.nih.gov/31905380/
Lang PO, Samaras D, Samaras N. Testosterone replacement therapy in reversing “andropause”: what is the proof-of-principle? Rejuvenation Res. 2012;15(5):453–65. https://pubmed.ncbi.nlm.nih.gov/22656862/
Corona G, Isidori AM, Buvat J, et al. Testosterone supplementation and sexual function: a meta-analysis study. J Sex Med. 2014;11(6):1577–92. https://pubmed.ncbi.nlm.nih.gov/24697970/
Adlin EV. Age-related low testosterone. Ann Intern Med. 2020;172(2):151–2. https://pubmed.ncbi.nlm.nih.gov/31905380/
Lang PO, Samaras D, Samaras N. Testosterone replacement therapy in reversing “andropause”: what is the proof-of-principle? Rejuvenation Res. 2012;15(5):453–65. https://pubmed.ncbi.nlm.nih.gov/22656862/
Fisher AD, Corona G, Bandini E, et al. Psychobiological correlates of extramarital affairs and differences between stable and occasional infidelity among men with sexual dysfunctions. J Sex Med. 2009;6(3):866–75. https://pubmed.ncbi.nlm.nih.gov/19143911/
Wagels L, Votinov M, Kellermann T, Eisert A, Beyer C, Habel U. Exogenous testosterone enhances the reactivity to social provocation in males. Front Behav Neurosci. 2018;12:37. https://pubmed.ncbi.nlm.nih.gov/29551966/
Handelsman DJ. Pharmacoepidemiology of testosterone: impact of reimbursement policy on curbing off-label prescribing. Pharmacoepidemiol Drug Saf. 2020;29(9):1030–6. https://pubmed.ncbi.nlm.nih.gov/32743911/
Bhasin S, Brito JP, Cunningham GR, et al. Testosterone therapy in men with hypogonadism: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2018;103(5):1715–44. https://pubmed.ncbi.nlm.nih.gov/29562364/
Shin YS, Park JK. The optimal indication for testosterone replacement therapy in late onset hypogonadism. J Clin Med. 2019;8(2):209. https://pubmed.ncbi.nlm.nih.gov/30736442/
Roy CN, Snyder PJ, Stephens-Shields AJ, et al. Association of testosterone levels with anemia in older men: a controlled clinical trial. JAMA Intern Med. 2017;177(4):480–90. https://pubmed.ncbi.nlm.nih.gov/28241237/
Swee DS, Gan EH. Late-onset hypogonadism as primary testicular failure. Front Endocrinol (Lausanne). 2019;10:372. https://pubmed.ncbi.nlm.nih.gov/31244778/
Basaria S, Coviello AD, Travison TG, et al. Adverse events associated with testosterone administration. N Engl J Med. 2010;363(2):109–22. https://pubmed.ncbi.nlm.nih.gov/20592293/
Information update – possible cardiovascular problems associated with testosterone products. Government of Canada. https://recalls-rappels.canada.ca/en/alert-recall/information-update-possible-cardiovascular-problems-associated-testosterone-products. Published July 15, 2014. Accessed May 5, 2022.; https://recalls-rappels.canada.ca/en/alert-recall/information-update-possible-cardiovascular-problems-associated-testosterone-products
Warren CJ, Wisener J, Ward B, et al. YouTube as a patient education resource for male hypogonadism and testosterone therapy. Sex Med. 2021;9(2):100324. https://pubmed.ncbi.nlm.nih.gov/33752104/
Sansone A, Sansone M, Lenzi A, Romanelli F. Testosterone replacement therapy: the emperor’s new clothes. Rejuvenation Res. 2017;20(1):9–14. https://pubmed.ncbi.nlm.nih.gov/27124096/
Warren CJ, Wisener J, Ward B, et al. YouTube as a patient education resource for male hypogonadism and testosterone therapy. Sex Med. 2021;9(2):100324. https://pubmed.ncbi.nlm.nih.gov/33752104/
Huo S, Scialli AR, McGarvey S, et al. Treatment of men for “low testosterone”: a systematic review. PLoS One. 2016;11(9):e0162480. https://pubmed.ncbi.nlm.nih.gov/27655114/
Kohn TP, Mata DA, Ramasamy R, Lipshultz LI. Effects of testosterone replacement therapy on lower urinary tract symptoms: a systematic review and meta-analysis. Eur Urol. 2016;69(6):1083–90. https://pubmed.ncbi.nlm.nih.gov/26874809/
Huggins C, Hodges CV. Studies on prostatic cancer. I. The effect of castration, of estrogen and androgen injection on serum phosphatases in metastatic carcinoma of the prostate. Cancer Res. 1941;1(4):293–7. https://pubmed.ncbi.nlm.nih.gov/12050481/
Perlmutter MA, Lepor H. Androgen deprivation therapy in the treatment of advanced prostate cancer. Rev Urol. 2007;9(Suppl 1):S3–8. https://pubmed.ncbi.nlm.nih.gov/17387371/
Miner M, Barkin J, Rosenberg MT. Testosterone deficiency: myth, facts, and controversy. Can J Urol. 2014;21(Suppl 2):39–54. https://pubmed.ncbi.nlm.nih.gov/24978631/
Nelson WG, De Marzo AM, Isaacs WB. Prostate cancer. N Engl J Med. 2003;349(4):366–81. https://pubmed.ncbi.nlm.nih.gov/12878745/
Salter CA, Mulhall JP. Guideline of guidelines: testosterone therapy for testosterone deficiency. BJU Int. 2019;124(5):722–9. https://pubmed.ncbi.nlm.nih.gov/31420972/
Nelson WG, De Marzo AM, Isaacs WB. Prostate cancer. N Engl J Med. 2003;349(4):366–81. https://pubmed.ncbi.nlm.nih.gov/12878745/
Grossman DC, Curry SJ, Owens DK, et al. Screening for prostate cancer: US Preventive Services Task Force recommendation statement. JAMA. 2018;319(18):1901–13. https://pubmed.ncbi.nlm.nih.gov/29801017/
Draisma G, Etzioni R, Tsodikov A, et al. Lead time and overdiagnosis in prostate-specific antigen screening: importance of methods and context. J Natl Cancer Inst. 2009;101(6):374–83. https://pubmed.ncbi.nlm.nih.gov/19276453/
Division of Cancer Prevention and Control, Centers for Disease Control and Prevention. Prostate cancer statistics. CDC.gov. http://www.cdc.gov/cancer/prostate/statistics/index.htm. Updated September 2, 2014. Accessed May 15, 2022.; https://www.cdc.gov/cancer/prostate/statistics/index.htm
Grossman DC, Curry SJ, Owens DK, et al. Screening for prostate cancer: US Preventive Services Task Force recommendation statement. JAMA. 2018;319(18):1901–13. https://pubmed.ncbi.nlm.nih.gov/29801017/
Moyer VA, LeFevre ML, Sui AL, et al. Screening for prostate cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2012;157(2):120–34. https://pubmed.ncbi.nlm.nih.gov/22801674/
Livingston CJ, Freeman RJ, Mohammad A, et al. Choosing Wisely® in preventive medicine. Am J Prev Med. 2016;51(1):141–9. https://pubmed.ncbi.nlm.nih.gov/27155735/
Mulhem E, Fulbright N, Duncan N. Prostate cancer screening. Am Fam Physician. 2015;92(8):683–8. https://pubmed.ncbi.nlm.nih.gov/26554408/
Ivlev I, Jerabkova S, Mishra M, Cook LA, Eden KB. Prostate cancer screening patient decision aids: a systematic review and meta-analysis. Am J Prev Med. 2018;55(6):896–907. https://pubmed.ncbi.nlm.nih.gov/30337235/
Grossman DC, Curry SJ, Owens DK, et al. Screening for prostate cancer: US Preventive Services Task Force recommendation statement. JAMA. 2018;319(18):1901–13. https://pubmed.ncbi.nlm.nih.gov/29801017/
Carter HB. American Urological Association (AUA) guideline on prostate cancer detection: process and rationale. BJU Int. 2013;112(5):543–7. https://pubmed.ncbi.nlm.nih.gov/23924423/
Wilt TJ, Harris RP, Qaseem A, et al. Screening for cancer: advice for high-value care from the American College of Physicians. Ann Intern Med. 2015;162(10):718–25. https://pubmed.ncbi.nlm.nih.gov/25984846/
American Cancer Society recommendations for prostate cancer early detection. American Cancer Society. https://www.cancer.org/cancer/prostate-cancer/detection-diagnosis-staging/acs-recommendations.html. Updated April 23, 2021. Accessed May 5, 2022.; https://www.cancer.org/cancer/prostate-cancer/detection-diagnosis-staging/acs-recommendations.html
Grossman DC, Curry SJ, Owens DK, et al. Screening for prostate cancer: US Preventive Services Task Force recommendation statement. JAMA. 2018;319(18):1901–13. https://pubmed.ncbi.nlm.nih.gov/29801017/
Tikkinen KAO, Dahm P, Lytvyn L, et al. Prostate cancer screening with prostate-specific antigen (PSA) test: a clinical practice guideline. BMJ. 2018;362:k3581. https://pubmed.ncbi.nlm.nih.gov/30185545/
Gigerenzer G, Mata J, Frank R. Public knowledge of benefits of breast and prostate cancer screening in Europe. J Natl Cancer Inst. 2009;101(17):1216–20. https://pubmed.ncbi.nlm.nih.gov/19671770/
Grossman DC, Curry SJ, Owens DK, et al. Screening for prostate cancer: US Preventive Services Task Force recommendation statement. JAMA. 2018;319(18):1901–13. https://pubmed.ncbi.nlm.nih.gov/29801017/
Tikkinen KAO, Dahm P, Lytvyn L, et al. Prostate cancer screening with prostate-specific antigen (PSA) test: a clinical practice guideline. BMJ. 2018;362:k3581. https://pubmed.ncbi.nlm.nih.gov/30185545/
Grossman DC, Curry SJ, Owens DK, et al. Screening for prostate cancer: US Preventive Services Task Force recommendation statement. JAMA. 2018;319(18):1901–13. https://pubmed.ncbi.nlm.nih.gov/29801017/
Grossman DC, Curry SJ, Owens DK, et al. Screening for prostate cancer: US Preventive Services Task Force recommendation statement. JAMA. 2018;319(18):1901–13. https://pubmed.ncbi.nlm.nih.gov/29801017/
Early detection of prostate cancer. Harding Center for Risk Literacy. https://www.hardingcenter.de/sites/default/files/2021–11/fact%20%20box_PSA_EN_new_design_20201123_final.pdf. Updated November 2020. Accessed May 4, 2022.; https://www.hardingcenter.de/en/transfer-and-impact/fact-boxes/early-detection-of-cancer/early-detection-of-prostate-cancer-with-psa-testing
Early detection of prostate cancer. Harding Center for Risk Literacy. https://www.hardingcenter.de/sites/default/files/2021–11/fact%20%20box_PSA_EN_new_design_20201123_final.pdf. Updated November 2020. Accessed May 4, 2022.; https://www.hardingcenter.de/en/transfer-and-impact/fact-boxes/early-detection-of-cancer/early-detection-of-prostate-cancer-with-psa-testing
Our History. Endocrine Society. https://www.endocrine.org/our-community/advancing-endocrinology-and-public-health/history. Accessed May 5, 2022.; https://www.endocrine.org/our-community/advancing-endocrinology-and-public-health/history
Al-Sharefi A, Quinton R. Current national and international guidelines for the management of male hypogonadism: helping clinicians to navigate variation in diagnostic criteria and treatment recommendations. Endocrinol Metab (Seoul). 2020;35(3):526–40. https://pubmed.ncbi.nlm.nih.gov/32981295/
Swee DS, Gan EH. Late-onset hypogonadism as primary testicular failure. Front Endocrinol (Lausanne). 2019;10:372. https://pubmed.ncbi.nlm.nih.gov/31244778/
Rastrelli G, Carter EL, Ahern T, et al. Development of and recovery from secondary hypogonadism in aging men: prospective results from the EMAS. J Clin Endocrinol Metab. 2015;100(8):3172–82. https://pubmed.ncbi.nlm.nih.gov/26000545/
Davidson LM, Millar K, Jones C, Fatum M, Coward K. Deleterious effects of obesity upon the hormonal and molecular mechanisms controlling spermatogenesis and male fertility. Hum Fertil (Camb). 2015;18(3):184–93. https://pubmed.ncbi.nlm.nih.gov/26205254/
Camacho EM, Huhtaniemi IT, O’Neill TW, et al. Age-associated changes in hypothalamic-pituitary-testicular function in middle-aged and older men are modified by weight change and lifestyle factors: longitudinal results from the European Male Ageing Study. Eur J Endocrinol. 2013;168(3):445–55. https://pubmed.ncbi.nlm.nih.gov/23425925/
Corona G, Rastrelli G, Monami M, et al. Body weight loss reverts obesity-associated hypogonadotropic hypogonadism: a systematic review and meta-analysis. Eur J Endocrinol. 2013;168(6):829–43. https://pubmed.ncbi.nlm.nih.gov/23482592/
Armamento-Villareal R, Aguirre LE, Qualls C, Villareal DT. Effect of lifestyle intervention on the hormonal profile of frail, obese older men. J Nutr Health Aging. 2016;20(3):334–40. https://pubmed.ncbi.nlm.nih.gov/26892583/
Hayes LD, Elliott BT. Short-term exercise training inconsistently influences basal testosterone in older men: a systematic review and meta-analysis. Front Physiol. 2018;9:1878. https://pubmed.ncbi.nlm.nih.gov/30692929/
Fukui H, Yamashita M. The effects of music and visual stress on testosterone and cortisol in men and women. Neuro Endocrinol Lett. 2003;24(3–4):173–80. https://pubmed.ncbi.nlm.nih.gov/14523353/
Fukui H. Music and testosterone. A new hypothesis for the origin and function of music. Ann N Y Acad Sci. 2001;930:448–51. https://pubmed.ncbi.nlm.nih.gov/11458865/
Leproult R, Van Cauter E. Effect of 1 week of sleep restriction on testosterone levels in young healthy men. JAMA. 2011;305(21):2173–4. https://pubmed.ncbi.nlm.nih.gov/21632481/
Sarkola T, Eriksson CJP. Testosterone increases in men after a low dose of alcohol. Alcohol Clin Exp Res. 2003;27(4):682–5. https://pubmed.ncbi.nlm.nih.gov/12711931/
Sierksma A, Sarkola T, Eriksson CJP, van der Gaag MS, Grobbee DE, Hendriks HFJ. Effect of moderate alcohol consumption on plasma dehydroepiandrosterone sulfate, testosterone, and estradiol levels in middle-aged men and postmenopausal women: a diet-controlled intervention study. Alcohol Clin Exp Res. 2004;28(5):780–5. https://pubmed.ncbi.nlm.nih.gov/15166654/
Hang D, Kværner AS, Ma W, et al. Coffee consumption and plasma biomarkers of metabolic and inflammatory pathways in US health professionals. Am J Clin Nutr. 2019;109(3):635–47. https://pubmed.ncbi.nlm.nih.gov/30834441/
Wedick NM, Mantzoros CS, Ding EL, et al. The effects of caffeinated and decaffeinated coffee on sex hormone – binding globulin and endogenous sex hormone levels: a randomized controlled trial. Nutr J. 2012;11:86. https://pubmed.ncbi.nlm.nih.gov/23078574/
Balasubramanian A, Thirumavalavan N, Srivatsav A, Yu J, Lipshultz LI, Pastuszak AW. Testosterone imposters: an analysis of popular online testosterone boosting supplements. J Sex Med. 2019;16(2):203–12. https://pubmed.ncbi.nlm.nih.gov/32257853/
Clemesha CG, Thaker H, Samplaski MK. ‘Testosterone boosting’ supplements composition and claims are not supported by the academic literature. World J Mens Health. 2020;38(1):115–22. https://pubmed.ncbi.nlm.nih.gov/31385468/
Park HJ, Lee KS, Lee EK, Park NC. Efficacy and safety of a mixed extract of Trigonella foenum-graecum seed and Lespedeza cuneata in the treatment of testosterone deficiency syndrome: a randomized, double-blind, placebo-controlled clinical trial. World J Mens Health. 2018;36(3):230–8. https://pubmed.ncbi.nlm.nih.gov/29623697/
Idris S, Mishra A, Khushtar M. Recent therapeutic interventions of fenugreek seed: a mechanistic approach. Drug Res (Stuttg). 2021;71(4):180–92. https://pubmed.ncbi.nlm.nih.gov/33378775/
Khodamoradi K, Khosropanah MH, Ayati Z, et al. The effects of fenugreek on cardiometabolic risk factors in adults: a systematic review and meta-analysis. Complement Ther Med. 2020;52:102416. https://pubmed.ncbi.nlm.nih.gov/32951700/
Askarpour M, Alami F, Campbell MS, Venkatakrishnan K, Hadi A, Ghaedi E. Effect of fenugreek supplementation on blood lipids and body weight: a systematic review and meta-analysis of randomized controlled trials. J Ethnopharmacol. 2020;253:112538. https://pubmed.ncbi.nlm.nih.gov/32087319/
Mansoori A, Hosseini S, Zilaee M, Hormoznejad R, Fathi M. Effect of fenugreek extract supplement on testosterone levels in male: a meta-analysis of clinical trials. Phytother Res. 2020;34(7):1550–5. https://pubmed.ncbi.nlm.nih.gov/32048383/
Rao A, Steels E, Inder WJ, Abraham S, Vitetta L. Testofen, a specialised Trigonella foenum-graecum seed extract reduces age-related symptoms of androgen decrease, increases testosterone levels and improves sexual function in healthy aging males in a double-blind randomised clinical study. The Aging Male. 2016;19(2):134–42. https://pubmed.ncbi.nlm.nih.gov/26791805/
Askarpour M, Alami F, Campbell MS, Venkatakrishnan K, Hadi A, Ghaedi E. Effect of fenugreek supplementation on blood lipids and body weight: a systematic review and meta-analysis of randomized controlled trials. J Ethnopharmacol. 2020;253:112538. https://pubmed.ncbi.nlm.nih.gov/32087319/
Khodamoradi K, Khosropanah MH, Ayati Z, et al. The effects of fenugreek on cardiometabolic risk factors in adults: a systematic review and meta-analysis. Complement Ther Med. 2020;52:102416. https://pubmed.ncbi.nlm.nih.gov/32951700/
Mebazaa R, Rega B, Camel V. Analysis of human male armpit sweat after fenugreek ingestion: characterisation of odour active compounds by gas chromatography coupled to mass spectrometry and olfactometry. Food Chem. 2011;128(1):227–35. https://pubmed.ncbi.nlm.nih.gov/25214354/
Pearce KL, Tremellen K. The effect of macronutrients on reproductive hormones in overweight and obese men: a pilot study. Nutrients. 2019;11(12):3059. https://pubmed.ncbi.nlm.nih.gov/31847341/
Tremellen K, Hill A, Pearce K. Mechanistic insights into the aetiology of post-prandial decline in testosterone in reproductive-aged men. Andrologia. 2019;51(10):e13418. https://pubmed.ncbi.nlm.nih.gov/31475727/
Tremellen K, McPhee N, Pearce K, Benson S, Schedlowski M, Engler H. Endotoxin-initiated inflammation reduces testosterone production in men of reproductive age. Am J Physiol Endocrinol Metab. 2018;314(3):E206–13. https://pubmed.ncbi.nlm.nih.gov/29183872/
Lehtihet M, Arver S, Bartuseviciene I, Pousette A. S-testosterone decrease after a mixed meal in healthy men independent of SHBG and gonadotrophin levels. Andrologia. 2012;44(6):405–10. https://pubmed.ncbi.nlm.nih.gov/22524522/
Tremellen K, Hill A, Pearce K. Mechanistic insights into the aetiology of post-prandial decline in testosterone in reproductive-aged men. Andrologia. 2019;51(10):e13418. https://pubmed.ncbi.nlm.nih.gov/31475727/
Lindgren O, Carr RD, Deacon CF, et al. Incretin hormone and insulin responses to oral versus intravenous lipid administration in humans. J Clin Endocrinol Metab. 2011;96(8):2519–24. https://pubmed.ncbi.nlm.nih.gov/21593115/
Jeibmann A, Zahedi S, Simoni M, Nieschlag E, Byrne MM. Glucagon-like peptide-1 reduces the pulsatile component of testosterone secretion in healthy males. Eur J Clin Invest. 2005;35(9):565–72. https://pubmed.ncbi.nlm.nih.gov/16128863/
Tremellen K, Hill A, Pearce K. Mechanistic insights into the aetiology of post-prandial decline in testosterone in reproductive-aged men. Andrologia. 2019;51(10):e13418. https://pubmed.ncbi.nlm.nih.gov/31475727/
Anderson KE, Rosner W, Khan MS, et al. Diet-hormone interactions: protein/carbohydrate ratio alters reciprocally the plasma levels of testosterone and cortisol and their respective binding globulins in man. Life Sci. 1987;40(18):1761–8. https://pubmed.ncbi.nlm.nih.gov/3573976/
Cook TM, Russell JM, Barker ME. Dietary advice for muscularity, leanness and weight control in Men’s Health magazine: a content analysis. BMC Public Health. 2014;14:1062. https://pubmed.ncbi.nlm.nih.gov/25304148/
Schwartz A, Hunschede S, Lacombe RJS, et al. Acute decrease in plasma testosterone and appetite after either glucose or protein beverages in adolescent males. Clin Endocrinol (Oxf). 2019;91(2):295–303. https://pubmed.ncbi.nlm.nih.gov/31055857/
Whittaker J, Harris M. Low-carbohydrate diets and men’s cortisol and testosterone: systematic review and meta-analysis. Nutr Health. 2022;28(4):543–54. https://pubmed.ncbi.nlm.nih.gov/35254136/
Schwartz A, Hunschede S, Lacombe RJS, et al. Acute decrease in plasma testosterone and appetite after either glucose or protein beverages in adolescent males. Clin Endocrinol (Oxf). 2019;91(2):295–303. https://pubmed.ncbi.nlm.nih.gov/31055857/
Messina M. Soybean isoflavone exposure does not have feminizing effects on men: a critical examination of the clinical evidence. Fertil Steril. 2010;93(7):2095–104. https://pubmed.ncbi.nlm.nih.gov/20378106/
Hamilton-Reeves JM, Vazquez G, Duval SJ, Phipps WR, Kurzer MS, Messina MJ. Clinical studies show no effects of soy protein or isoflavones on reproductive hormones in men: results of a meta-analysis. Fertil Steril. 2010;94(3):997–1007. https://pubmed.ncbi.nlm.nih.gov/19524224/
Shultz TD, Bonorden WR, Seaman WR. Effect of short-term flaxseed consumption on lignan and sex hormone metabolism in men. Nutr Res. 1991;11(10):1089–100. https://www.sciencedirect.com/science/article/abs/pii/S0271531705806876
Takenaka T, Nagano M, Yamashita K, Kikuchi K. Flaxseed oil stimulates gynecomastia. BMJ Case Rep. 2020;13(12):e237948. https://pubmed.ncbi.nlm.nih.gov/33303502/
Schwartz LM, Woloshin S. Low “T” as in “template”: how to sell disease. JAMA Intern Med. 2013;173(15):1460–2. https://pubmed.ncbi.nlm.nih.gov/23939516/
Shores MM, Matsumoto AM. Testosterone, aging and survival: biomarker or deficiency. Curr Opin Endocrinol Diabetes Obes. 2014;21(3):209–16. https://pubmed.ncbi.nlm.nih.gov/24722173/
Fugh-Berman A. Treating aging with testosterone. Am Fam Physician. 2017;96(7):428–30. https://pubmed.ncbi.nlm.nih.gov/29094915/
Lee JH, Shah PH, Uma D, Salvi DJ, Rabbani R, Hamid P. Testosterone replacement therapy in hypogonadal men and myocardial infarction risk: systematic review & meta-analysis. Cureus. 2021;13(8):e17475. https://pubmed.ncbi.nlm.nih.gov/34513525/
Vigen R, O’Donnell CI, Barón AE, et al. Association of testosterone therapy with mortality, myocardial infarction, and stroke in men with low testosterone levels. JAMA. 2013;310(17):1829–36. https://pubmed.ncbi.nlm.nih.gov/24193080/
Regan JC, Partridge L. Gender and longevity: why men die earlier than women? Comparative and experimental evidence. Best Pract Res Clin Endocrinol Metab. 2013;27(4):467–79. https://pubmed.ncbi.nlm.nih.gov/24054925/
Tremellen K. Gut Endotoxin Leading to a Decline IN Gonadal function (GELDING) – a novel theory for the development of late onset hypogonadism in obese men. Basic Clin Androl. 2016;26:7. https://pubmed.ncbi.nlm.nih.gov/27340554/
Lang PO, Samaras D, Samaras N. Testosterone replacement therapy in reversing “andropause”: what is the proof-of-principle? Rejuvenation Res. 2012;15(5):453–65. https://pubmed.ncbi.nlm.nih.gov/22656862/
Regan JC, Partridge L. Gender and longevity: why men die earlier than women? Comparative and experimental evidence. Best Pract Res Clin Endocrinol Metab. 2013;27(4):467–79. https://pubmed.ncbi.nlm.nih.gov/24054925/
Tremellen K. Gut Endotoxin Leading to a Decline IN Gonadal function (GELDING) – a novel theory for the development of late onset hypogonadism in obese men. Basic Clin Androl. 2016;26:7. https://pubmed.ncbi.nlm.nih.gov/27340554/
Garratt M, Stout MB. Hormone actions controlling sex-specific life-extension. Aging (Albany NY). 2018;10(3):293–4. https://pubmed.ncbi.nlm.nih.gov/29514132/
Drori D, Folman Y. Interactive environmental and genetic effects on longevity in the male rat: litter size, exercise, electric shocks and castration. Exp Aging Res. 1986;12(2):59–64. https://pubmed.ncbi.nlm.nih.gov/3569385/
Min KJ, Lee CK, Park HN. The lifespan of Korean eunuchs. Curr Biol. 2012;22(18):R792–3. https://pubmed.ncbi.nlm.nih.gov/23017989/
Le Bourg É. No ground for advocating that Korean eunuchs lived longer than intact men. Gerontology. 2015;62(1):69–70. https://pubmed.ncbi.nlm.nih.gov/26138349/
Nieschlag E, Nieschlag S, Behre HM. Lifespan and testosterone. Nature. 1993;366(6452):215. https://pubmed.ncbi.nlm.nih.gov/8232579/
Spiegel AM. The Jeremiah Metzger lecture: a brief history of eugenics in America: implications for medicine in the 21st century. Trans Am Clin Climatol Assoc. 2019;130:216–34. https://pubmed.ncbi.nlm.nih.gov/31516187/
Buck v Bell, 274 US 200 (1927).; https://supreme.justia.com/cases/federal/us/274/200/
Hamilton JB, Mestler GE. Mortality and survival: comparison of eunuchs with intact men and women in a mentally retarded population. J Gerontol. 1969;24(4):395–411. https://pubmed.ncbi.nlm.nih.gov/5362349/
Handelsman DJ. Testosterone and male aging: faltering hope for rejuvenation. JAMA. 2017;317(7):699–701. https://pubmed.ncbi.nlm.nih.gov/28241336/
Montecino-Rodriguez E, Berent-Maoz B, Dorshkind K. Causes, consequences, and reversal of immune system aging. J Clin Invest. 2013;123(3):958–65. https://pubmed.ncbi.nlm.nih.gov/23454758/
Alam I, Almajwal AM, Alam W, et al. The immune-nutrition interplay in aging – facts and controversies. Nutr Healthy Aging. 2019;5(2):73–95. https://content.iospress.com/articles/nutrition-and-healthy-aging/nha170034
Xu W, Wong G, Hwang YY, Larbi A. The untwining of immunosenescence and aging. Semin Immunopathol. 2020;42(5):559–72. https://pubmed.ncbi.nlm.nih.gov/33165716/
Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. Immun Ageing. 2019;16:25. https://pubmed.ncbi.nlm.nih.gov/31528180/
Fagiolo U, Cossarizza A, Scala E, et al. Increased cytokine production in mononuclear cells of healthy elderly people. Eur J Immunol. 1993;23(9):2375–8. https://pubmed.ncbi.nlm.nih.gov/8370415/
Cevenini E, Monti D, Franceschi C. Inflamm-ageing. Curr Opin Clin Nutr Metab Care. 2013;16(1):14–20. https://pubmed.ncbi.nlm.nih.gov/23132168/
Crimmins EM. Age-related vulnerability to coronavirus disease 2019 (COVID-19): biological, contextual, and policy-related factors. Public Policy Aging Rep. 2020;30(4):142–6. https://pubmed.ncbi.nlm.nih.gov/33214754/
Painter SD, Ovsyannikova IG, Poland GA. The weight of obesity on the human immune response to vaccination. Vaccine. 2015;33(36):4422–9. https://pubmed.ncbi.nlm.nih.gov/26163925/
Neidich SD, Green WD, Rebeles J, et al. Increased risk of influenza among vaccinated adults who are obese. Int J Obes (Lond). 2017;41(9):1324–30. https://pubmed.ncbi.nlm.nih.gov/28584297/
Sjöström L. Review of the key results from the Swedish Obese Subjects (SOS) trial – a prospective controlled intervention study of bariatric surgery. J Intern Med. 2013;273(3):219–34. https://pubmed.ncbi.nlm.nih.gov/23163728/
Jahn J, Spielau M, Brandsch C, et al. Decreased NK cell functions in obesity can be reactivated by fat mass reduction. Obesity (Silver Spring). 2015;23(11):2233–41. https://pubmed.ncbi.nlm.nih.gov/26390898/
Walsh NP, Gleeson M, Shephard RJ, et al. Position statement. Part one: immune function and exercise. Exerc Immunol Rev. 2011;17:6–63. https://pubmed.ncbi.nlm.nih.gov/21446352/
Nieman DC. Moderate exercise improves immunity and decreases illness rates. Am J Lifestyle Med. 2011;5(4):338–45. https://www.researchgate.net/publication/254075198_Moderate_Exercise_Improves_Immunity_and_Decreases_Illness_Rates
Bigley AB, Rezvani K, Chew C, et al. Acute exercise preferentially redeploys NK-cells with a highly-differentiated phenotype and augments cytotoxicity against lymphoma and multiple myeloma target cells. Brain Behav Immun. 2014;39:160–71. https://pubmed.ncbi.nlm.nih.gov/24200514/
McTiernan A, Friedenreich CM, Katzmarzyk PT, et al. Physical activity in cancer prevention and survival: a systematic review. Med Sci Sports Exerc. 2019;51(6):1252–61. https://pubmed.ncbi.nlm.nih.gov/31095082/
Kohut ML, Arntson BA, Lee W, et al. Moderate exercise improves antibody response to influenza immunization in older adults. Vaccine. 2004;22(17–18):2298–306. https://pubmed.ncbi.nlm.nih.gov/15149789/
Ranadive SM, Cook M, Kappus RM, et al. Effect of acute aerobic exercise on vaccine efficacy in older adults. Med Sci Sports Exerc. 2014;46(3):455–61. https://pubmed.ncbi.nlm.nih.gov/23924918/
Long JE, Ring C, Drayson M, et al. Vaccination response following aerobic exercise: can a brisk walk enhance antibody response to pneumococcal and influenza vaccinations? Brain Behav Immun. 2012;26(4):680–7. https://pubmed.ncbi.nlm.nih.gov/22386744/
Antonelli M, Barbieri G, Donelli D. Effects of forest bathing (shinrin-yoku) on levels of cortisol as a stress biomarker: a systematic review and meta-analysis. Int J Biometeorol. 2019;63(8):1117–34. https://pubmed.ncbi.nlm.nih.gov/31001682/
Li Q, Kawada T. Effect of forest environments on human natural killer (NK) activity. Int J Immunopathol Pharmacol. 2011;24(1 Suppl):39S-44S. https://pubmed.ncbi.nlm.nih.gov/21329564/
Li Q, Morimoto K, Kobayashi M, et al. Visiting a forest, but not a city, increases human natural killer activity and expression of anti-cancer proteins. Int J Immunopathol Pharmacol. 2008;21(1):117–27. https://pubmed.ncbi.nlm.nih.gov/18336737/
Sumitomo K, Akutsu H, Fukuyama S, et al. Conifer-derived monoterpenes and forest walking. Mass Spectrom (Tokyo). 2015;4(1):A0042. https://pubmed.ncbi.nlm.nih.gov/26819913/
Li Q, Nakadai A, Matsushima H, et al. Phytoncides (wood essential oils) induce human natural killer cell activity. Immunopharmacol Immunotoxicol. 2006;28(2):319–33. https://pubmed.ncbi.nlm.nih.gov/16873099/
Shibata H, Fujiwara R, Iwamoto M, Matsuoka H, Yokoyama MM. Immunological and behavioral effects of fragrance in mice. Int J Neurosci. 1991;57(1–2):151–9. https://pubmed.ncbi.nlm.nih.gov/1938152/
Li Q, Kobayashi M, Wakayama Y, et al. Effect of phytoncide from trees on human natural killer cell function. Int J Immunopathol Pharmacol. 2009;22(4):951–9. https://pubmed.ncbi.nlm.nih.gov/20074458/
Fujimori H, Hisama M, Shibayama H, Iwaki M. Protecting effect of phytoncide solution, on normal human dermal fibroblasts against reactive oxygen species. J Oleo Sci. 2009;58(8):429–36. https://pubmed.ncbi.nlm.nih.gov/19584569/
Li Q, Kobayashi M, Kawada T. Relationships between percentage of forest coverage and standardized mortality ratios (SMR) of cancers in all prefectures in Japan. Open Public Health J. 2008;1(1):1–7. https://openpublichealthjournal.com/VOLUME/1/PAGE/1/ABSTRACT/
Ahmadi F, Ahmadi N. Nature as the most important coping strategy among cancer patients: a Swedish survey. J Relig Health. 2015;54(4):1177–90. https://pubmed.ncbi.nlm.nih.gov/24363200/
Brown R, Pang G, Husband AJ, King MG. Suppression of immunity to influenza virus infection in the respiratory tract following sleep disturbance. Reg Immunol. 1989;2(5):321–5. https://pubmed.ncbi.nlm.nih.gov/2562046/
Toth LA, Rehg JE. Effects of sleep deprivation and other stressors on the immune and inflammatory responses of influenza-infected mice. Life Sci. 1998;63(8):701–9. https://pubmed.ncbi.nlm.nih.gov/9718099/
Renegar KB, Floyd RA, Krueger JM. Effects of short-term sleep deprivation on murine immunity to influenza virus in young adult and senescent mice. Sleep. 1998;21(3):241–8. https://pubmed.ncbi.nlm.nih.gov/9595602/
Besedovsky L, Lange T, Born J. Sleep and immune function. Pflugers Arch. 2012;463(1):121–37. https://pubmed.ncbi.nlm.nih.gov/22071480/
Prather AA, Hall M, Fury JM, et al. Sleep and antibody response to hepatitis B vaccination. Sleep. 2012;35(8):1063–9. https://pubmed.ncbi.nlm.nih.gov/22851802/
Prather AA, Pressman SD, Miller GE, Cohen S. Temporal links between self-reported sleep and antibody responses to the influenza vaccine. Int J Behav Med. 2021;28(1):151–8. https://pubmed.ncbi.nlm.nih.gov/32236831/
Lange T, Perras B, Fehm HL, Born J. Sleep enhances the human antibody response to hepatitis A vaccination. Psychosom Med. 2003;65(5):831–5. https://pubmed.ncbi.nlm.nih.gov/14508028/
Lange T, Dimitrov S, Bollinger T, Diekelmann S, Born J. Sleep after vaccination boosts immunological memory. J Immunol. 2011;187(1):283–90. https://pubmed.ncbi.nlm.nih.gov/21632713/
Spiegel K, Sheridan JF, Van Cauter E. Effect of sleep deprivation on response to immunization. JAMA. 2002;288(12):1471–2. https://pubmed.ncbi.nlm.nih.gov/12243633/
Irwin M, McClintick J, Costlow C, Fortner M, White J, Gillin JC. Partial night sleep deprivation reduces natural killer and cellular immune responses in humans. FASEB J. 1996;10(5):643–53. https://pubmed.ncbi.nlm.nih.gov/8621064/
Irwin M, Mascovich A, Gillin JC, Willoughby R, Pike J, Smith TL. Partial sleep deprivation reduces natural killer cell activity in humans. Psychosom Med. 1994;56(6):493–8. https://pubmed.ncbi.nlm.nih.gov/7871104/
Patel SR, Malhotra A, Gao X, Hu FB, Neuman MI, Fawzi WW. A prospective study of sleep duration and pneumonia risk in women. Sleep. 2012;35(1):97–101. https://pubmed.ncbi.nlm.nih.gov/22215923/
Cohen S, Doyle WJ, Alper CM, Janicki-Deverts D, Turner RB. Sleep habits and susceptibility to the common cold. Arch Intern Med. 2009;169(1):62–7. https://pubmed.ncbi.nlm.nih.gov/19139325/
Prather AA, Janicki-Deverts D, Hall MH, Cohen S. Behaviorally assessed sleep and susceptibility to the common cold. Sleep. 2015;38(9):1353–9. https://pubmed.ncbi.nlm.nih.gov/26118561/
Besedovsky L, Born J. Sleep, don’t sneeze: longer sleep reduces the risk of catching a cold. Sleep. 2015;38(9):1341–2. https://pubmed.ncbi.nlm.nih.gov/26285007/
Fulop T, Larbi A, Dupuis G, et al. Immunosenescence and inflamm-aging as two sides of the same coin: friends or foes? Front Immunol. 2018;8:1960. https://pubmed.ncbi.nlm.nih.gov/29375577/
Lesourd B. Nutritional factors and immunological ageing. Proc Nutr Soc. 2006;65(3):319–25. https://pubmed.ncbi.nlm.nih.gov/16923315/
Xu W, Wong G, Hwang YY, Larbi A. The untwining of immunosenescence and aging. Semin Immunopathol. 2020;42(5):559–72. https://pubmed.ncbi.nlm.nih.gov/33165716/
Averill HM, Averill JE. The effect of daily apple consumption on dental caries experience, oral hygiene status and upper respiratory infections. N Y State Dent J. 1968;34(7):403–9. https://pubmed.ncbi.nlm.nih.gov/4385913/
Charland KM, Buckeridge DL, Hoen AG, et al. Relationship between community prevalence of obesity and associated behavioral factors and community rates of influenza-related hospitalizations in the United States. Influenza Other Respir Viruses. 2013;7(5):718–28. https://pubmed.ncbi.nlm.nih.gov/23136926/
U.S. Centers for Disease Control and Prevention. Prevention of pneumococcal disease: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR. 1997;46(RR-08):1–24. https://pubmed.ncbi.nlm.nih.gov/9132580/
Gibson A, Edgar J, Neville C et al. Effect of fruit and vegetable consumption on immune function in older people: a randomized controlled trial. Am J Clin Nutr. 2012;96(6):1429–36. https://pubmed.ncbi.nlm.nih.gov/23134881/
Skinner MA. Gold kiwifruit for immune support and reducing symptoms of cold and influenza. J Food Drug Anal. 2012;20:261–4. https://pubmed.ncbi.nlm.nih.gov/21349229/
Orhan F, Karakas T, Cakir M, Aksoy A, Baki A, Gedik Y. Prevalence of immunoglobulin E – mediated food allergy in 6–9-year-old urban schoolchildren in the eastern Black Sea region of Turkey. Clin Exp Allergy. 2009;39(7):1027–35. https://pubmed.ncbi.nlm.nih.gov/19400894/
Rancé F, Grandmottet X, Grandjean H. Prevalence and main characteristics of schoolchildren diagnosed with food allergies in France. Clin Exp Allergy. 2005;35(2):167–72. https://pubmed.ncbi.nlm.nih.gov/15725187/
Hunter DC, Skinner MA, Wolber FM, et al. Consumption of gold kiwifruit reduces severity and duration of selected upper respiratory tract infection symptoms and increases plasma vitamin C concentration in healthy older adults. Br J Nutr. 2012;108(7):1235–45. https://pubmed.ncbi.nlm.nih.gov/22172428/
Jefferson T, Jones M, Doshi P, Spencer EA, Onakpoya I, Heneghan CJ. Oseltamivir for influenza in adults and children: systematic review of clinical study reports and summary of regulatory comments. BMJ. 2014;348:g2545. https://pubmed.ncbi.nlm.nih.gov/24811411/
van der Gaag EJ, Hummel TZ. Food or medication? The therapeutic effects of food on the duration and incidence of upper respiratory tract infections: a review of the literature. Crit Rev Food Sci Nutr. 2021;61(16):2691–704. https://pubmed.ncbi.nlm.nih.gov/32648776/
Hawkins J, Baker C, Cherry L, Dunne E. Black elderberry (Sambucus nigra) supplementation effectively treats upper respiratory symptoms: a meta-analysis of randomized, controlled clinical trials. Complement Ther Med. 2019;42:361–5. https://pubmed.ncbi.nlm.nih.gov/30670267/
Macknin M, Wolski K, Negrey J, Mace S. Elderberry extract outpatient influenza treatment for emergency room patients ages 5 and above: a randomized, double-blind, placebo-controlled trial. J Gen Intern Med. 2020;35(11):3271–7. https://pubmed.ncbi.nlm.nih.gov/32929634/
Rothberg MB. Influenza, like COVID-19, needs randomized trials. J Gen Intern Med. 2021;36(6):1490–1. https://pubmed.ncbi.nlm.nih.gov/33483821/
David S, Cunningham R. Echinacea for the prevention and treatment of upper respiratory tract infections: a systematic review and meta-analysis. Complement Ther Med. 2019;44:18–26. https://pubmed.ncbi.nlm.nih.gov/31126553/
Weissman S, Lo A, Patel R, et al. An unusual culprit of drug-induced pancreatitis. Dig Dis Sci. 2020;65(5):1549–52. https://pubmed.ncbi.nlm.nih.gov/31571105/
Elderberry for influenza. Med Lett Drugs Ther. 2019;61(1566):32. https://pubmed.ncbi.nlm.nih.gov/30845105/
Pogorzelski E. Formation of cyanide as a product of decomposition of cyanogenic glucosides in the treatment of elderberry fruit (Sambucus nigra). J Sci Food Agric. 1982;33(5):496–8. https://onlinelibrary.wiley.com/doi/abs/10.1002/jsfa.2740330516
Centers for Disease Control. Poisoning from elderberry juice – California. MMWR Morb Mortal Wkly Rep. 1984;33(13):173–4. https://pubmed.ncbi.nlm.nih.gov/6422238/
McAnulty LS, Collier SR, Landram MJ, et al. Six weeks daily ingestion of whole blueberry powder increases natural killer cell counts and reduces arterial stiffness in sedentary males and females. Nutr Res. 2014;34(7):577–84. https://pubmed.ncbi.nlm.nih.gov/25150116/
Majdalawieh AF, Carr RI. In vitro investigation of the potential immunomodulatory and anti-cancer activities of black pepper (Piper nigrum) and cardamom (Elettaria cardamomum). J Med Food. 2010;13(2):371–81. https://pubmed.ncbi.nlm.nih.gov/20210607/
Pan P, Kang S, Wang Y, et al. Black raspberries enhance natural killer cell infiltration into the colon and suppress the progression of colorectal cancer. Front Immunol. 2017;8:997. https://pubmed.ncbi.nlm.nih.gov/28861089/
Huang YW, Lin CW, Pan P, et al. Black raspberries suppress colorectal cancer by enhancing Smad4 expression in colonic epithelium and natural killer cells. Front Immunol. 2020;11:570683. https://pubmed.ncbi.nlm.nih.gov/33424832/
Wang H, Gao T, Du Y, et al. Anticancer and immunostimulating activities of a novel homogalacturonan from Hippophae rhamnoides L. berry. Carbohydr Polym. 2015;131:288–96. https://pubmed.ncbi.nlm.nih.gov/26256187/
Larmo P, Alin J, Salminen E, Kallio H, Tahvonen R. Effects of sea buckthorn berries on infections and inflammation: a double-blind, randomized, placebo-controlled trial. Eur J Clin Nutr. 2008;62(9):123–30. https://pubmed.ncbi.nlm.nih.gov/17593932/
Agricultural Research Service, United States Department of Agriculture. Gogi berries, dried. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=goji&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/173032/nutrients. Published April 1, 2019. Accessed May 20, 2022.; https://fdc.nal.usda.gov/fdc-app.html?query=goji&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/173032/nutrients
Vidal K, Bucheli P, Gao Q, et al. Immunomodulatory effects of dietary supplementation with a milk-based wolfberry formulation in healthy elderly: a randomized, double-blind, placebo-controlled trial. Rejuvenation Res. 2012;15(1):89–97. https://pubmed.ncbi.nlm.nih.gov/22352435/
Mostafalou S, Abdollahi M. Pesticides: an update of human exposure and toxicity. Arch Toxicol. 2017;91(2):549–99. https://pubmed.ncbi.nlm.nih.gov/27722929/
Kapeleka JA, Sauli E, Ndakidemi PA. Pesticide exposure and genotoxic effects as measured by DNA damage and human monitoring biomarkers. Int J Environ Health Res. 2021;31(7):805–22. https://pubmed.ncbi.nlm.nih.gov/31736325/
Hemler EC, Chavarro JE, Hu FB. Organic foods for cancer prevention – worth the investment? JAMA Intern Med. 2018;178(12):1606–7. https://pubmed.ncbi.nlm.nih.gov/30422205/
Hemler EC, Chavarro JE, Hu FB. Organic foods for cancer prevention – worth the investment? JAMA Intern Med. 2018;178(12):1606–7. https://pubmed.ncbi.nlm.nih.gov/30422205/
Juraske R, Mutel CL, Stoessel F, Hellweg S. Life cycle human toxicity assessment of pesticides: comparing fruit and vegetable diets in Switzerland and the United States. Chemosphere. 2009;77(7):939–45. https://pubmed.ncbi.nlm.nih.gov/19729188/
Watzl B, Bub A, Brandstetter BR, Rechkemmer G. Modulation of human T-lymphocyte functions by the consumption of carotenoid-rich vegetables. Br J Nutr. 1999;82(5):383–9. https://pubmed.ncbi.nlm.nih.gov/10673911/
Watzl B, Bub A, Briviba K, Rechkemmer G. Supplementation of a low-carotenoid diet with tomato or carrot juice modulates immune functions in healthy men. Ann Nutr Metab. 2003;47(6):255–61. https://pubmed.ncbi.nlm.nih.gov/14520020/
Briviba K, Kulling SE, Möseneder J, Watzl B, Rechkemmer G, Bub A. Effects of supplementing a low-carotenoid diet with a tomato extract for 2 weeks on endogenous levels of DNA single strand breaks and immune functions in healthy non-smokers and smokers. Carcinogenesis. 2004;25(12):2373–8. https://pubmed.ncbi.nlm.nih.gov/15308586/
Watzl B, Bub A, Blockhaus M, et al. Prolonged tomato juice consumption has no effect on cell-mediated immunity of well-nourished elderly men and women. J Nutr. 2000;130(7):1719–23. https://pubmed.ncbi.nlm.nih.gov/10867042/
Müller L, Meyer M, Bauer RN, et al. Effect of broccoli sprouts and live attenuated influenza virus on peripheral blood natural killer cells: a randomized, double-blind study. PLoS One. 2016;11(1):e0147742. https://pubmed.ncbi.nlm.nih.gov/26820305/
Lord SJ, Rajotte RV, Korbutt GS, Bleackley RC. Granzyme B: a natural born killer. Immunol Rev. 2003;193:31–8. https://pubmed.ncbi.nlm.nih.gov/12752668/
Noah TL, Zhang H, Zhou H, et al. Effect of broccoli sprouts on nasal response to live attenuated influenza virus in smokers: a randomized, double-blind study. PLoS One. 2014;9(6):e98671. https://pubmed.ncbi.nlm.nih.gov/24910991/
Cho HY, Imani F, Miller-DeGraff L, et al. Antiviral activity of Nrf2 in a murine model of respiratory syncytial virus disease. Am J Respir Crit Care Med. 2009;179(2):138–50. https://pubmed.ncbi.nlm.nih.gov/18931336/
Wu CC, Chuang HY, Lin CY, et al. Inhibition of Epstein-Barr virus reactivation in nasopharyngeal carcinoma cells by dietary sulforaphane. Mol Carcinog. 2013;52(12):946–58. https://pubmed.ncbi.nlm.nih.gov/22641235/
Harvey CJ, Thimmulappa RK, Sethi S, et al. Targeting Nrf2 signaling improves bacterial clearance by alveolar macrophages in patients with COPD and in a mouse model. Sci Transl Med. 2011;3(78):78ra32. https://pubmed.ncbi.nlm.nih.gov/21490276/
Gasper AV, Al-janobi A, Smith JA, et al. Glutathione S-transferase M1 polymorphism and metabolism of sulforaphane from standard and high-glucosinolate broccoli. Am J Clin Nutr. 2005;82(6):1283–91. https://pubmed.ncbi.nlm.nih.gov/16332662/
Ritz T, Trueba AF, Vogel PD, Auchus RJ, Rosenfield D. Exhaled nitric oxide and vascular endothelial growth factor as predictors of cold symptoms after stress. Biol Psychol. 2018;132:116–24. https://pubmed.ncbi.nlm.nih.gov/29162553/
Sanders SP, Siekierski ES, Richards SM, Porter JD, Imani F, Proud D. Rhinovirus infection induces expression of type 2 nitric oxide synthase in human respiratory epithelial cells in vitro and in vivo. J Allergy Clin Immunol. 2001;107(2):235–43. https://pubmed.ncbi.nlm.nih.gov/9444985/
Domínguez R, Cuenca E, Maté-Muñoz JL, et al. Effects of beetroot juice supplementation on cardiorespiratory endurance in athletes. A systematic review. Nutrients. 2017;9(1):43. https://pubmed.ncbi.nlm.nih.gov/28067808/
Swathi Krishna S, Thennavan A, Kanthlal SK. Dietary foods containing nitric oxide donors can be early curators of SARS-CoV-2 infection: a possible role in the immune system. J Food Biochem. 2022;46(3):e13884. https://pubmed.ncbi.nlm.nih.gov/34374096/
Ritz T, Werchan CA, Kroll JL, Rosenfield D. Beetroot juice supplementation for the prevention of cold symptoms associated with stress: a proof-of-concept study. Physiol Behav. 2019;202:45–51. https://pubmed.ncbi.nlm.nih.gov/30682333/
Brown ES, Allsopp PJ, Magee PJ, et al. Seaweed and human health. Nutr Rev. 2014;72(3):205–16. https://pubmed.ncbi.nlm.nih.gov/24697280/
Tamama K. Potential benefits of dietary seaweeds as protection against COVID-19. Nutr Rev. 2021;79(7):814–23. https://pubmed.ncbi.nlm.nih.gov/32498269/
Miyake Y, Sasaki S, Ohya Y, et al. Dietary intake of seaweed and minerals and prevalence of allergic rhinitis in Japanese pregnant females: baseline data from the Osaka Maternal and Child Health Study. Ann Epidemiol. 2006;16(8):614–21. https://pubmed.ncbi.nlm.nih.gov/16406247/
Shan BE, Yoshida Y, Kuroda E, Yamashita U. Immunomodulating activity of seaweed extract on human lymphocytes in vitro. Int J Immunopharmacol. 1999;21(1):59–70. https://pubmed.ncbi.nlm.nih.gov/10411282/
Cooper R, Dragar C, Elliot K, Fitton JH, Godwin J, Thompson K. GFS, a preparation of Tasmanian Undaria pinnatifida is associated with healing and inhibition of reactivation of Herpes. BMC Complement Altern Med. 2002;2:11. https://pubmed.ncbi.nlm.nih.gov/16685055/
Ryan-Borchers TA, Park JS, Chew BP, McGuire MK, Fournier LR, Beerman KA. Soy isoflavones modulate immune function in healthy postmenopausal women. Am J Clin Nutr. 2006;83(5):1118–25. https://pubmed.ncbi.nlm.nih.gov/16965235/
Messina M, Nagata C, Wu AH. Estimated Asian adult soy protein and isoflavone intakes. Nutr Cancer. 2006;55(1):1–12. https://pubmed.ncbi.nlm.nih.gov/15050097/
Teas J, Hebert JR, Fitton JH, Zimba PV. Algae – a poor man’s HAART? Med Hypotheses. 2004;62(4):507–10. https://pubmed.ncbi.nlm.nih.gov/33341894/
Tamama K. Potential benefits of dietary seaweeds as protection against COVID-19. Nutr Rev. 2021;79(7):814–23. https://pubmed.ncbi.nlm.nih.gov/32498269/
Jung SJ, Jang HY, Jung ES, et al. Effects of Porphyra tenera supplementation on the immune system: a randomized, double-blind, and placebo-controlled clinical trial. Nutrients. 2020;12(6):E1642. https://fdc.nal.usda.gov/fdc-app.html?query=nori&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/168458/nutrients
Agricultural Research Service, United States Department of Agriculture. Seaweed, laver, raw. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=nori&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/168458/nutrients. Published April 1, 2019. Accessed May 20, 2022.; https://fdc.nal.usda.gov/fdc-app.html?query=onion&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/790577/nutrients
Jung SJ, Jang HY, Jung ES, et al. Effects of Porphyra tenera supplementation on the immune system: a randomized, double-blind, and placebo-controlled clinical trial. Nutrients. 2020;12(6):E1642. https://fdc.nal.usda.gov/fdc-app.html?query=nori&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/168458/nutrients
Ishihara K, Oyamada C, Sato Y, et al. Relationships between quality parameters and content of glycerol galactoside and porphyra-334 in dried laver nori Porphyra yezoensis. Fisheries Sci. 2008;74(1):167–73. https://pubmed.ncbi.nlm.nih.gov/18580401/
Neville V, Gleeson M, Folland JP. Salivary IgA as a risk factor for upper respiratory infections in elite professional athletes. Med Sci Sports Exerc. 2008 Jul;40(7):1228–36. https://pubmed.ncbi.nlm.nih.gov/18580401/
.;
Dietzen DJ. Amino acids, peptides, and proteins. In: Principles and Applications of Molecular Diagnostics. Elsevier; 2018:345–80. https://profiles.wustl.edu/en/publications/amino-acids-peptides-and-proteins
Otsuki T, Shimizu K, Iemitsu M, Kono I. Salivary secretory immunoglobulin A secretion increases after 4-weeks ingestion of chlorella-derived multicomponent supplement in humans: a randomized cross over study. Nutr J. 2011 Sep 9;10:91. https://pubmed.ncbi.nlm.nih.gov/21906314/
Nakano S, Takekoshi H, Nakano M. Chlorella (Chlorella pyrenoidosa) supplementation decreases dioxin and increases immunoglobulin a concentrations in breast milk. J Med Food. 2007;10(1):134–42. https://pubmed.ncbi.nlm.nih.gov/17472477/
Halperin SA, Smith B, Nolan C, Shay J, Kralovec J. Safety and immunoenhancing effect of a Chlorella-derived dietary supplement in healthy adults undergoing influenza vaccination: randomized, double-blind, placebo-controlled trial. CMAJ. 2003;169(2):111–7. https://pubmed.ncbi.nlm.nih.gov/12874157/
Otsuki T, Shimizu K, Iemitsu M, Kono I. Salivary secretory immunoglobulin A secretion increases after 4-weeks ingestion of chlorella-derived multicomponent supplement in humans: a randomized cross over study. Nutr J. 2011;10:91. https://pubmed.ncbi.nlm.nih.gov/21906314/
Kwak JH, Baek SH, Woo Y, et al. Beneficial immunostimulatory effect of short-term Chlorella supplementation: enhancement of Natural Killer cell activity and early inflammatory response (randomized, double-blinded, placebo-controlled trial). Nutr J. 2012;11:53. https://pubmed.ncbi.nlm.nih.gov/22849818/
Azocar J, Diaz A. Efficacy and safety of Chlorella supplementation in adults with chronic hepatitis C virus infection. World J Gastroenterol. 2013;19(7):1085–90. https://pubmed.ncbi.nlm.nih.gov/23467073/
Fallah AA, Sarmast E, Habibian Dehkordi S, et al. Effect of Chlorella supplementation on cardiovascular risk factors: a meta-analysis of randomized controlled trials. Clin Nutr. 2018;37(6 Pt A):1892–901. https://pubmed.ncbi.nlm.nih.gov/29037431/
Selvaraj V, Singh H, Ramaswamy S. Chlorella-induced psychosis. Psychosomatics. 2013;54(3):303–4. https://pubmed.ncbi.nlm.nih.gov/23680061/
Petrovska BB, Cekovska S. Extracts from the history and medical properties of garlic. Pharmacogn Rev. 2010;4(7):106–10. https://pubmed.ncbi.nlm.nih.gov/22228949/
Darooghegi Mofrad M, Milajerdi A, Koohdani F, Surkan PJ, Azadbakht L. Garlic supplementation reduces circulating C-reactive protein, tumor necrosis factor, and interleukin-6 in adults: a systematic review and meta-analysis of randomized controlled trials. J Nutr. 2019;149(4):605–18. https://pubmed.ncbi.nlm.nih.gov/30949665/
Louca P, Murray B, Klaser K, et al. Modest effects of dietary supplements during the COVID-19 pandemic: insights from 445 850 users of the COVID-19 Symptom Study app. BMJ Nutr Prev Health. 2021;4(1):149–57. https://pubmed.ncbi.nlm.nih.gov/34308122/
Lawson LD, Hunsaker SM. Allicin bioavailability and bioequivalence from garlic supplements and garlic foods. Nutrients. 2018;10(7):812. https://pubmed.ncbi.nlm.nih.gov/29937536/
Locatelli DA, Altamirano JC, González RE, Camargo AB. Home-cooked garlic remains a healthy food. J Funct Food. 2015;16:1–8. https://www.sciencedirect.com/science/article/abs/pii/S1756464615001851
Lawson LD, Hunsaker SM. Allicin bioavailability and bioequivalence from garlic supplements and garlic foods. Nutrients. 2018;10(7):812. https://pubmed.ncbi.nlm.nih.gov/29937536/
Wong A, Townley S. Herbal medicines and anaesthesia. Cont Educ Anaesth Crit Care Pain. 2011;11(1):14–7. https://academic.oup.com/bjaed/article/11/1/14/285726
Gadkari JV, Joshi VD. Effect of ingestion of raw garlic on serum cholesterol level, clotting time and fibrinolytic activity in normal subjects. J Postgrad Med. 1991;37(3):128–31. https://pubmed.ncbi.nlm.nih.gov/1784022/
Scharbert G, Kalb ML, Duris M, Marschalek C, Kozek-Langenecker SA. Garlic at dietary doses does not impair platelet function. Anesth Analg. 2007;105(5):1214–8. https://pubmed.ncbi.nlm.nih.gov/17959943/
Munch R, Barringer SA. Deodorization of garlic breath volatiles by food and food components. J Food Sci. 2014;79(4):C526–33. https://pubmed.ncbi.nlm.nih.gov/24592995/
Jeong SC, Koyyalamudi SR, Pang G. Dietary intake of Agaricus bisporus white button mushroom accelerates salivary immunoglobulin A secretion in healthy volunteers. Nutrition. 2012;28(5):527–31. https://pubmed.ncbi.nlm.nih.gov/22113068/
Jesenak M, Hrubisko M, Majtan J, Rennerova Z, Banovcin P. Anti-allergic effect of Pleuran (ß-glucan from Pleurotus ostreatus) in children with recurrent respiratory tract infections. Phytother Res. 2014;28(3):471–4. https://pubmed.ncbi.nlm.nih.gov/23744488/
Dai X, Stanilka JM, Rowe CA, et al. Consuming Lentinula edodes (shiitake) mushrooms daily improves human immunity: a randomized dietary intervention in healthy young adults. J Am Coll Nutr. 2015;34(6):478–87. https://pubmed.ncbi.nlm.nih.gov/25866155/
Xu Y, Na L, Ren Z, et al. Effect of dietary supplementation with white button mushrooms on host resistance to influenza infection and immune function in mice. Br J Nutr. 2013;109(6):1052–61. https://pubmed.ncbi.nlm.nih.gov/23200185/
Bobovcák M, Kuniaková R, Gabriž J, Majtán J. Effect of Pleuran (ß-glucan from Pleurotus ostreatus) supplementation on cellular immune response after intensive exercise in elite athletes. Appl Physiol Nutr Metab. 2010;35(6):755–62. https://pubmed.ncbi.nlm.nih.gov/21164546/
Cerletti C, Esposito S, Iacoviello L. Edible mushrooms and beta-glucans: impact on human health. Nutrients. 2021;13(7):2195. https://pubmed.ncbi.nlm.nih.gov/34202377/
Bergendiova K, Tibenska E, Majtan J. Pleuran (ß-glucan from Pleurotus ostreatus) supplementation, cellular immune response and respiratory tract infections in athletes. Eur J Appl Physiol. 2011;111(9):2033–40. https://pubmed.ncbi.nlm.nih.gov/21249381/
Lehne G, Haneberg B, Gaustad P, Johansen PW, Preus H, Abrahamsen TG. Oral administration of a new soluble branched ß-1,3-D-glucan is well tolerated and can lead to increased salivary concentrations of immunoglobulin A in healthy volunteers. Clin Exp Immunol. 2006;143(1):65–9. https://pubmed.ncbi.nlm.nih.gov/16367935/
Zhong K, Liu Z, Lu Y, Xu X. Effects of yeast ß-glucans for the prevention and treatment of upper respiratory tract infection in healthy subjects: a systematic review and meta-analysis. Eur J Nutr. 2021;60(8):4175–87. https://pubmed.ncbi.nlm.nih.gov/33900466/
Kohl A, Gögebakan O, Möhlig M, et al. Increased interleukin-10 but unchanged insulin sensitivity after 4 weeks of (1, 3)(1, 6)-ß-glycan consumption in overweight humans. Nutr Res. 2009;29(4):248–54. https://pubmed.ncbi.nlm.nih.gov/19410976/
Yenidogan E, Akgul GG, Gulcelik MA, Dinc S, Colakoglu MK, Kayaoglu HA. Effect of ß-glucan on drain fluid and amount of drainage following modified radical mastectomy. Adv Ther. 2014;31(1):130–9. https://pubmed.ncbi.nlm.nih.gov/24421054/
Koray M, Ak G, Kürklü E, et al. The effect of ß-glucan on recurrent aphthous stomatitis. J Altern Complement Med. 2009;15(2):111–2. https://pubmed.ncbi.nlm.nih.gov/19216661/
Mosikanon K, Arthan D, Kettawan A, Tungtrongchitr R, Prangthip P. Yeast ß-glucan modulates inflammation and waist circumference in overweight and obese subjects. J Diet Suppl. 2017;14(2):173–85. https://pubmed.ncbi.nlm.nih.gov/27715351/
Santas J, Lázaro E, Cuñé J. Effect of a polysaccharide-rich hydrolysate from Saccharomyces cerevisiae (LipiGo®) in body weight loss: randomised, double-blind, placebo-controlled clinical trial in overweight and obese adults. J Sci Food Agric. 2017;97(12):4250–7. https://pubmed.ncbi.nlm.nih.gov/28251654/
Vlassopoulou M, Yannakoulia M, Pletsa V, Zervakis GI, Kyriacou A. Effects of fungal beta-glucans on health – a systematic review of randomized controlled trials. Food Funct. 2021;12(8):3366–80. https://pubmed.ncbi.nlm.nih.gov/33876798/
Barclay GR, McKenzie H, Pennington J, Parratt D, Pennington CR. The effect of dietary yeast on the activity of stable chronic Crohn’s disease. Scand J Gastroenterol. 1992;27(3):196–200. https://pubmed.ncbi.nlm.nih.gov/1502481/
Cannistrà C, Finocchi V, Trivisonno A, Tambasco D. New perspectives in the treatment of hidradenitis suppurativa: surgery and brewer’s yeast – exclusion diet. Surgery. 2013;154(5):1126–30. https://pubmed.ncbi.nlm.nih.gov/23891479/
Stier H, Ebbeskotte V, Gruenwald J. Immune-modulatory effects of dietary Yeast Beta-1,3/1,6-D-glucan. Nutr J. 2014;13:38. https://pubmed.ncbi.nlm.nih.gov/24774968/
van Steenwijk HP, Bast A, de Boer A. Immunomodulating effects of fungal beta-glucans: from traditional use to medicine. Nutrients. 2021;13(4):1333. https://pubmed.ncbi.nlm.nih.gov/33920583/
Stier H, Ebbeskotte V, Gruenwald J. Immune-modulatory effects of dietary Yeast Beta-1,3/1,6-D-glucan. Nutr J. 2014;13:38. https://pubmed.ncbi.nlm.nih.gov/24774968/
Kamath AB, Wang L, Das H, Li L, Reinhold VN, Bukowski JF. Antigens in tea-beverage prime human V¿2Vd2 T cells in vitro and in vivo for memory and nonmemory antibacterial cytokine responses. Proc Natl Acad Sci U S A. 2003;100(10):6009–14. https://pubmed.ncbi.nlm.nih.gov/12719524/
Bukowski JF, Morita CT, Brenner MB. Human ¿d T cells recognize alkylamines derived from microbes, edible plants, and tea: implications for innate immunity. Immunity. 1999;11(1):57–65. https://pubmed.ncbi.nlm.nih.gov/10435579/
Hartmann T. Nachweis von n-butylamin in äpfeln. Experientia. 1967;23(8):680–1. https://pubmed.ncbi.nlm.nih.gov/6051708/
Ibe A, Saito K, Nakazato M, Kikuchi Y, Fujinuma K, Nishima T. Quantitative determination of amines in wine by liquid chromatography. J Assoc Off Anal Chem. 1991;74(4):695–8. https://pubmed.ncbi.nlm.nih.gov/1917817/
Jones BM, Al-Fattani M, Gooch H. The determination of amines in the vaginal secretions of women in health and disease. Int J STD AIDS. 1994;5(1):52–5. https://pubmed.ncbi.nlm.nih.gov/8142529/
Rowe CA, Nantz MP, Bukowski JF, Percival SS. Specific formulation of Camellia sinensis prevents cold and flu symptoms and enhances gamma,delta T cell function: a randomized, double-blind, placebo-controlled study. J Am Coll Nutr. 2007;26(5):445–52. https://pubmed.ncbi.nlm.nih.gov/17914132/
Kamath AB, Wang L, Das H, Li L, Reinhold VN, Bukowski JF. Antigens in tea-beverage prime human V¿2Vd2 T cells in vitro and in vivo for memory and nonmemory antibacterial cytokine responses. Proc Natl Acad Sci U S A. 2003;100(10):6009–14. https://pubmed.ncbi.nlm.nih.gov/12719524/
Park M, Yamada H, Matsushita K, et al. Green tea consumption is inversely associated with the incidence of influenza infection among schoolchildren in a tea plantation area of Japan. J Nutr. 2011;141(10):1862–70. https://pubmed.ncbi.nlm.nih.gov/21832025/
Watanabe I, Kuriyama S, Kakizaki M, et al. Green tea and death from pneumonia in Japan: the Ohsaki cohort study. Am J Clin Nutr. 2009;90(3):672–9. https://pubmed.ncbi.nlm.nih.gov/19625686/
Rowe CA, Nantz MP, Bukowski JF, Percival SS. Specific formulation of Camellia sinensis prevents cold and flu symptoms and enhances ¿d T cell function: a randomized, double-blind, placebo-controlled study. J Am Coll Nutr. 2007;26(5):445–52. https://pubmed.ncbi.nlm.nih.gov/17914132/
Rawangkan A, Kengkla K, Kanchanasurakit S, Duangjai A, Saokaew S. Anti-influenza with green tea catechins: a systematic review and meta-analysis. Molecules. 2021;26(13):4014. https://pubmed.ncbi.nlm.nih.gov/34209247/
Matsumoto K, Yamada H, Takuma N, Niino H, Sagesaka YM. Effects of green tea catechins and theanine on preventing influenza infection among healthcare workers: a randomized controlled trial. BMC Complement Altern Med. 2011;11:15. https://pubmed.ncbi.nlm.nih.gov/21338496/
Rawangkan A, Kengkla K, Kanchanasurakit S, Duangjai A, Saokaew S. Anti-influenza with green tea catechins: a systematic review and meta-analysis. Molecules. 2021;26(13):4014. https://pubmed.ncbi.nlm.nih.gov/34209247/
Furushima D, Nishimura T, Takuma N, et al. Prevention of acute upper respiratory infections by consumption of catechins in healthcare workers: a randomized, placebo-controlled trial. Nutrients. 2019;12(1):E4. https://pubmed.ncbi.nlm.nih.gov/31861349/
Song JM, Lee KH, Seong BL. Antiviral effect of catechins in green tea on influenza virus. Antiviral Res. 2005;68:66–74. https://pubmed.ncbi.nlm.nih.gov/16137775/
Furushima D, Otake Y, Koike N, et al. Investigation of the oral retention of tea catechins in humans: an exploratory interventional study. Nutrients. 2021;13(9):3024. https://pubmed.ncbi.nlm.nih.gov/34578903/
Satomura K, Kitamura T, Kawamura T, et al. Prevention of upper respiratory tract infections by gargling: a randomized trial. Am J Prev Med. 2005;29(4):302–7. https://pubmed.ncbi.nlm.nih.gov/16242593/
Rawangkan A, Kengkla K, Kanchanasurakit S, Duangjai A, Saokaew S. Anti-influenza with green tea catechins: a systematic review and meta-analysis. Molecules. 2021;26(13):4014. https://pubmed.ncbi.nlm.nih.gov/34209247/
Ide K, Yamada H, Kawasaki Y. Effect of gargling with tea and ingredients of tea on the prevention of influenza infection: a meta-analysis. BMC Public Health. 2016;16:396. https://pubmed.ncbi.nlm.nih.gov/27175786/
Goodall EC, Granados AC, Luinstra K, et al. Vitamin D3 and gargling for the prevention of upper respiratory tract infections: a randomized controlled trial. BMC Infect Dis. 2014;14:273. https://pubmed.ncbi.nlm.nih.gov/24885201/
Chow EP, Howden BP, Walker S, et al. Antiseptic mouthwash against pharyngeal Neisseria gonorrhoeae: a randomised controlled trial and an in vitro study. Sex Transm Infect. 2017;93(2):88–93. https://pubmed.ncbi.nlm.nih.gov/27998950/
Maddaford K, Fairley CK, Trumpour S, Chung M, Chow EPF. Sites in the oropharynx reached by different methods of using mouthwash: clinical implication for oropharyngeal gonorrhoea prevention. Sex Transm Infect. 2020;96(5):358–60. https://pubmed.ncbi.nlm.nih.gov/31628249/
Phillips TR, Fairley C, Maddaford K, et al. Duration of gargling and rinsing among frequent mouthwash users: a cross-sectional study. BMJ Open. 2020;10(9):e040754. https://pubmed.ncbi.nlm.nih.gov/32994261/
Kumari M, Kozyrskyj AL. Gut microbial metabolism defines host metabolism: an emerging perspective in obesity and allergic inflammation. Obes Rev. 2017;18(1):18–31. https://pubmed.ncbi.nlm.nih.gov/27862824/
Brown AJ, Goldsworthy SM, Barnes AA, et al. The orphan G protein – coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem. 2003;278(13):11312–9. https://pubmed.ncbi.nlm.nih.gov/12496283/
Ang Z, Ding JL. GPR41 and GPR43 in obesity and inflammation – protective or causative? Front Immunol. 2016;7:28. https://pubmed.ncbi.nlm.nih.gov/26870043/
Jiao J, Xu JY, Zhang W, Han S, Qin LQ. Effect of dietary fiber on circulating C-reactive protein in overweight and obese adults: a meta-analysis of randomized controlled trials. Int J Food Sci Nutr. 2015;66(1):114–9. https://pubmed.ncbi.nlm.nih.gov/25578759/
Halnes I, Baines KJ, Berthon BS, MacDonald-Wicks LK, Gibson PG, Wood LG. Soluble fibre meal challenge reduces airway inflammation and expression of GPR43 and GPR41 in asthma. Nutrients. 2017;9(1):57. https://pubmed.ncbi.nlm.nih.gov/28075383/
Van Landingham CB, Keast DR, Longnecker MP. Serum concentration of antibodies to mumps, but not measles, rubella, or varicella, is associated with intake of dietary fiber in the NHANES, 1999–2004. Nutrients. 2021;13(3):813. https://pubmed.ncbi.nlm.nih.gov/33801237/
Hagan T, Cortese M, Rouphael N, et al. Antibiotics-driven gut microbiome perturbation alters immunity to vaccines in humans. Cell. 2019;178(6):1313–28.e13. https://pubmed.ncbi.nlm.nih.gov/31491384/
Yeh TL, Shih PC, Liu SJ, et al. The influence of prebiotic or probiotic supplementation on antibody titers after influenza vaccination: a systematic review and meta-analysis of randomized controlled trials. Drug Des Devel Ther. 2018;12:217–30. https://pubmed.ncbi.nlm.nih.gov/29416317/
Haak BW, Littmann ER, Chaubard JL, et al. Impact of gut colonization with butyrate-producing microbiota on respiratory viral infection following allo-HCT. Blood. 2018;131(26):2978–86. https://pubmed.ncbi.nlm.nih.gov/29674425/
Williams LM, Stoodley IL, Berthon BS, Wood LG. The effects of prebiotics, synbiotics, and short-chain fatty acids on respiratory tract infections and immune function: a systematic review and meta-analysis. Adv Nutr. 2022;13(1):167–92. https://pubmed.ncbi.nlm.nih.gov/34543378/
Hao Q, Dong BR, Wu T. Probiotics for preventing acute upper respiratory tract infections. Cochrane Database Syst Rev. 2015;(2):CD006895. https://pubmed.ncbi.nlm.nih.gov/25927096/
Guillemard E, Tondu F, Lacoin F, Schrezenmeir J. Consumption of a fermented dairy product containing the probiotic Lactobacillus casei DN-114001 reduces the duration of respiratory infections in the elderly in a randomised controlled trial. Br J Nutr. 2010;103(1):58–68. https://pubmed.ncbi.nlm.nih.gov/19747410/
Fujita R, Iimuro S, Shinozaki T, et al. Decreased duration of acute upper respiratory tract infections with daily intake of fermented milk: a multicenter, double-blinded, randomized comparative study in users of day care facilities for the elderly population. Am J Infect Control. 2013;41(12):1231–5. https://pubmed.ncbi.nlm.nih.gov/23890374/
Turchet P, Laurenzano M, Auboiron S, Antoine JM. Effect of fermented milk containing the probiotic Lactobacillus casei DN-114001 on winter infections in free-living elderly subjects: a randomised, controlled pilot study. J Nutr Health Aging. 2003;7(2):75–7. https://pubmed.ncbi.nlm.nih.gov/12679825/
Xu W, Wong G, Hwang YY, Larbi A. The untwining of immunosenescence and aging. Semin Immunopathol. 2020;42(5):559–72. https://pubmed.ncbi.nlm.nih.gov/33165716/
Malter M, Schriever G, Eilber U. Natural killer cells, vitamins, and other blood components of vegetarian and omnivorous men. Nutr Cancer. 1989;12(3):271–8. https://pubmed.ncbi.nlm.nih.gov/2771803/
Franco EL. The sexually transmitted disease model for cervical cancer: incoherent epidemiologic findings and the role of misclassification of human papillomavirus infection. Epidemiology. 1991;2(2):98–106. https://pubmed.ncbi.nlm.nih.gov/1657209/
Skrabanek P. Cervical cancer in nuns and prostitutes: a plea for scientific continence. J Clin Epidemiol. 1988;41(6):577–82. https://pubmed.ncbi.nlm.nih.gov/3290397/
Snijders PJF, Steenbergen RDM, Heideman DAM, Meijer CJLM. HPV-mediated cervical carcinogenesis: concepts and clinical implications. J Pathol. 2006;208(2):152–64. https://pubmed.ncbi.nlm.nih.gov/16362994/
Goldstein MA, Goodman A, del Carmen MG, Wilbur DC. Case 10–2009: a 23-year-old woman with an abnormal Papanicolaou smear. Cabot RC, Harris NL, Shepard JAO, et al., eds. N Engl J Med. 2009;360(13):1337–44. https://pubmed.ncbi.nlm.nih.gov/19321871/
Srivastava S, Gupta S, Roy JK. High prevalence of oncogenic HPV-16 in cervical smears of asymptomatic women of eastern Uttar Pradesh, India: a population-based study. J Biosci. 2012;37(1):63–72. https://pubmed.ncbi.nlm.nih.gov/22357204/
Sedjo RL, Roe DJ, Abrahamsen M, et al. Vitamin A, carotenoids, and risk of persistent oncogenic human papillomavirus infection. Cancer Epidemiol Biomarkers Prev. 2002;11(9):876–84. https://pubmed.ncbi.nlm.nih.gov/12223432/
Tantamango-Bartley Y, Jaceldo-Siegl K, Fan J, Fraser G. Vegetarian diets and the incidence of cancer in a low-risk population. Cancer Epidemiol Biomarkers Prev. 2013;22(2):286–94. https://pubmed.ncbi.nlm.nih.gov/23169929/
Haddad EH, Berk LS, Kettering JD, Hubbard RW, Peters WR. Dietary intake and biochemical, hematologic, and immune status of vegans compared with nonvegetarians. Am J Clin Nutr. 1999;70(3 Suppl):586S-93S. https://pubmed.ncbi.nlm.nih.gov/10479236/
Merino J, Joshi AD, Nguyen LH, et al. Diet quality and risk and severity of COVID-19: a prospective cohort study. Gut. 2021;70(11):2096–104. https://pubmed.ncbi.nlm.nih.gov/34489306/
Hyun SH, Ahn HY, Kim HJ, et al. Immuno-enhancement effects of Korean Red Ginseng in healthy adults: a randomized, double-blind, placebo-controlled trial. J Ginseng Res. 2021;45(1):191–8. https://pubmed.ncbi.nlm.nih.gov/33437171/
Kim MS. Korean red ginseng tonic extends lifespan in D. melanogaster. Biomol Ther (Seoul). 2013;21(3):241–5. https://pubmed.ncbi.nlm.nih.gov/24265871/
Wang H, Zhang S, Zhai L, et al. Ginsenoside extract from ginseng extends lifespan and health span in Caenorhabditis elegans. Food Funct. 2021;12(15):6793–808. https://pubmed.ncbi.nlm.nih.gov/34109970/
Bittles AH, Fulder SJ, Grant EC, Nicholls MR. The effect of ginseng on lifespan and stress responses in mice. Gerontology. 1979;25(3):125–31. https://pubmed.ncbi.nlm.nih.gov/571386/
Carabin IG, Burdock GA, Chatzidakis C. Safety assessment of Panax ginseng. Int J Toxicol. 2000;19(4):293–301. https://www.researchgate.net/publication/247768227_Safety_Assessment_of_Panax_Ginseng
Hyun SH, Ahn HY, Kim HJ, et al. Immuno-enhancement effects of Korean Red Ginseng in healthy adults: a randomized, double-blind, placebo-controlled trial. J Ginseng Res. 2021;45(1):191–8. https://pubmed.ncbi.nlm.nih.gov/33437171/
Scaglione F, Ferrara F, Dugnani S, Falchi M, Santoro G, Fraschini F. Immunomodulatory effects of two extracts of Panax ginseng C.A. Meyer. Drugs Exp Clin Res. 1990;16(10):537–42. https://pubmed.ncbi.nlm.nih.gov/2100737/
Antonelli M, Donelli D, Firenzuoli F. Ginseng integrative supplementation for seasonal acute upper respiratory infections: a systematic review and meta-analysis. Complement Ther Med. 2020;52:102457. https://pubmed.ncbi.nlm.nih.gov/32951718/
Siegel RK. Ginseng abuse syndrome. Problems with the panacea. JAMA. 1979;241(15):1614–5. https://pubmed.ncbi.nlm.nih.gov/430716/
Norelli LJ, Xu C. Manic psychosis associated with ginseng: a report of two cases and discussion of the literature. J Diet Suppl. 2015;12(2):119–25. https://pubmed.ncbi.nlm.nih.gov/24689505/
Kakisaka Y, Ohara T, Tozawa H, et al. Panax ginseng: a newly identified cause of gynecomastia. Tohoku J Exp Med. 2012;228(2):143–5. https://pubmed.ncbi.nlm.nih.gov/23006978/
Viviano A, Steele D, Edsell M, Jahangiri M. Over-the-counter natural products in cardiac surgery: a case of ginseng-related massive perioperative bleeding. BMJ Case Rep. 2017;2017:bcr-2016–218068. https://pubmed.ncbi.nlm.nih.gov/28784871/
Antonelli M, Donelli D, Firenzuoli F. Ginseng integrative supplementation for seasonal acute upper respiratory infections: a systematic review and meta-analysis. Complement Ther Med. 2020;52:102457. https://pubmed.ncbi.nlm.nih.gov/32951718/
Alam I, Almajwal AM, Alam W, et al. The immune – nutrition interplay in aging – facts and controversies. Nutr Healthy Aging. 2019;5(2):73–95. https://content.iospress.com/articles/nutrition-and-healthy-aging/nha170034
Lesourd B. Nutritional factors and immunological ageing. Proc Nutr Soc. 2006;65(3):319–25. https://pubmed.ncbi.nlm.nih.gov/16923315/
Chandra RK. Effect of vitamin and trace-element supplementation on immune responses and infection in elderly subjects. Lancet. 1992;340(8828):1124–7. https://pubmed.ncbi.nlm.nih.gov/1359211/
Lesourd B. Nutritional factors and immunological ageing. Proc Nutr Soc. 2006;65(3):319–25. https://pubmed.ncbi.nlm.nih.gov/16923315/
Editors of The Lancet. Retraction – effect of vitamin and trace-element supplementation on immune responses and infection in elderly subject. Lancet. 2016;387(10017):417. https://pubmed.ncbi.nlm.nih.gov/26869554/
Retraction and closure – Nutr Res 2002;22: 5–11 and Nutr Res 2002;22: 85–87. Nutr Res. 2016;36(7):756. https://pubmed.ncbi.nlm.nih.gov/27333962/
Roberts S, Sternberg S. Do nutritional supplements improve cognitive function in the elderly? Nutrition. 2003;19(11–12):976–8. https://pubmed.ncbi.nlm.nih.gov/14624947/
Pryse-Phillips W. Inquiry into Dr. RK Chandra’s submitted paper to the BMJ #00/5797. Published October 23, 2009. Accessed Aug 10, 2021.; https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(16)00166-5/fulltext
Vlieg-Boerstra B, de Jong N, Meyer R, et al. Nutrient supplementation for prevention of viral respiratory tract infections in healthy subjects: a systematic review and meta-analysis. Allergy. 2022;77(5):1373–88. https://pubmed.ncbi.nlm.nih.gov/34626488/
Graat JM, Schouten EG, Kok FJ. Effect of daily vitamin E and multivitamin – mineral supplementation on acute respiratory tract infections in elderly persons: a randomized controlled trial. JAMA. 2002;288(6):715–21. https://pubmed.ncbi.nlm.nih.gov/12169075/
Liu BA, McGeer A, McArthur MA, et al. Effect of multivitamin and mineral supplementation on episodes of infection in nursing home residents: a randomized, placebo-controlled study. J Am Geriatr Soc. 2007;55(1):35–42. https://pubmed.ncbi.nlm.nih.gov/17233683/
Girodon F, Galan P, Monget AL, et al. Impact of trace elements and vitamin supplementation on immunity and infections in institutionalized elderly patients: a randomized controlled trial. Arch Intern Med. 1999;159(7):748–54. https://pubmed.ncbi.nlm.nih.gov/10218756/
Carpenter KJ. The discovery of vitamin C. Ann Nutr Metab. 2012;61(3):259–64. https://pubmed.ncbi.nlm.nih.gov/23183299/
Hemilä H, Chalker E. Vitamin C for preventing and treating the common cold. Cochrane Database Syst Rev. 2013;(1):CD000980. https://pubmed.ncbi.nlm.nih.gov/23440782/
Hemilä H, Chalker E. Vitamin C for preventing and treating the common cold. Cochrane Database Syst Rev. 2013;(1):CD000980. https://pubmed.ncbi.nlm.nih.gov/23440782/
Taylor EN, Stampfer MJ, Curhan GC. Dietary factors and the risk of incident kidney stones in men: new insights after 14 years of follow-up. J Am Soc Nephrol. 2004;15(12):3225–32. https://pubmed.ncbi.nlm.nih.gov/15579526/
Thomas LDK, Elinder CG, Tiselius HG, Wolk A, Åkesson A. Ascorbic acid supplements and kidney stone incidence among men: a prospective study. JAMA Intern Med. 2013;173(5):386. https://pubmed.ncbi.nlm.nih.gov/23381591/
Thomas LDK, Elinder CG, Tiselius HG, Wolk A, Åkesson A. Ascorbic acid supplements and kidney stone incidence among men: a prospective study. JAMA Intern Med. 2013;173(5):386. https://pubmed.ncbi.nlm.nih.gov/23381591/
Goncalves-Mendes N, Talvas J, Dualé C, et al. Impact of vitamin D supplementation on influenza vaccine response and immune functions in deficient elderly persons: a randomized placebo-controlled trial. Front Immunol. 2019;10:65. https://pubmed.ncbi.nlm.nih.gov/30800121/
Jolliffe DA, Camargo CA, Sluyter JD, et al. Vitamin D supplementation to prevent acute respiratory infections: a systematic review and meta-analysis of aggregate data from randomised controlled trials. Lancet Diabetes Endocrinol. 2021;9(5):276–92. https://pubmed.ncbi.nlm.nih.gov/33798465/
Meydani SN, Meydani M, Blumberg JB, et al. Vitamin E supplementation and in vivo immune response in healthy elderly subjects. A randomized controlled trial. JAMA. 1997;277(17):1380–6. https://pubmed.ncbi.nlm.nih.gov/9134944/
Graat JM, Schouten EG, Kok FJ. Effect of daily vitamin E and multivitamin – mineral supplementation on acute respiratory tract infections in elderly persons: a randomized controlled trial. JAMA. 2002;288(6):715–21. https://pubmed.ncbi.nlm.nih.gov/12169075/
Klein EA, Thompson IM, Tangen CM, et al. Vitamin E and the risk of prostate cancer: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA. 2011;306(14):1549–56. https://pubmed.ncbi.nlm.nih.gov/21990298/
Curtis AJ, Bullen M, Piccenna L, McNeil JJ. Vitamin E supplementation and mortality in healthy people: a meta-analysis of randomised controlled trials. Cardiovasc Drugs Ther. 2014;28(6):563–73. https://pubmed.ncbi.nlm.nih.gov/25398301/
Kasprak A. Did a noted pathologist write this viral coronavirus advice letter? Snopes. https://www.snopes.com/fact-check/zinc-lozenges-coronavirus/. Published March 2, 2020. Updated March 13, 2020. Accessed May 22, 2022.; https://www.snopes.com/fact-check/zinc-lozenges-coronavirus
Monto AS. Medical reviews. Coronaviruses. Yale J Biol Med. 1974;47(4):234–51. https://pubmed.ncbi.nlm.nih.gov/4617423/
Hemilä H. Zinc lozenges and the common cold: a meta-analysis comparing zinc acetate and zinc gluconate, and the role of zinc dosage. JRSM Open. 2017;8(5):2054270417694291. https://pubmed.ncbi.nlm.nih.gov/28515951/
Hemilä H, Petrus EJ, Fitzgerald JT, Prasad A. Zinc acetate lozenges for treating the common cold: an individual patient data meta-analysis. Br J Clin Pharmacol. 2016;82(5):1393–8. https://pubmed.ncbi.nlm.nih.gov/27378206/
Hemilä H, Chalker E. The effectiveness of high dose zinc acetate lozenges on various common cold symptoms: a meta-analysis. BMC Fam Pract. 2015;16:24. https://pubmed.ncbi.nlm.nih.gov/25888289/
Hemilä H. Zinc lozenges and the common cold: a meta-analysis comparing zinc acetate and zinc gluconate, and the role of zinc dosage. JRSM Open. 2017;8(5):2054270417694291. https://pubmed.ncbi.nlm.nih.gov/28515951/
Singh M, Das RR. Zinc for the common cold. Cochrane Database Syst Rev. 2013;(6):CD001364. https://pubmed.ncbi.nlm.nih.gov/23775705/
Sempértegui F, Estrella B, Rodríguez O, et al. Zinc as an adjunct to the treatment of severe pneumonia in Ecuadorian children: a randomized controlled trial. Am J Clin Nutr. 2014;99(3):497–505. https://pubmed.ncbi.nlm.nih.gov/24429536/
Prasad AS. Impact of the discovery of human zinc deficiency on health. J Am Coll Nutr. 2009;28(3):257–65. https://pubmed.ncbi.nlm.nih.gov/20150599/
Ervin RB, Kennedy-Stephenson J. Mineral intakes of elderly adult supplement and non-supplement users in the third National Health and Nutrition Examination Survey. J Nutr. 2002;132(11):3422–7. https://pubmed.ncbi.nlm.nih.gov/12421862/
King JC. Yet again, serum zinc concentrations are unrelated to zinc intakes. J Nutr. 2018;148(9):1399–401. https://pubmed.ncbi.nlm.nih.gov/30184229/
Agricultural Research Service, United States Department of Agriculture. Mollusks, oysters, eastern, canned. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=oysters&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/171981/nutrients. Published April 1, 2019. Accessed May 20, 2022.; https://fdc.nal.usda.gov/fdc-app.html?query=oysters&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/171981/nutrients
Agricultural Research Service, United States Department of Agriculture. Beans, baked, canned, plain or vegetarian. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=baked+beans&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/175182/nutrients. Published April 1, 2019. Accessed May 20, 2022.; https://fdc.nal.usda.gov/fdc-app.html?query=baked+beans&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/175182/nutrients
Abioye AI, Bromage S, Fawzi W. Effect of micronutrient supplements on influenza and other respiratory tract infections among adults: a systematic review and meta-analysis. BMJ Glob Health. 2021;6(1):e003176. https://pubmed.ncbi.nlm.nih.gov/33472840/
Bogden JD, Oleske JM, Lavenhar MA, et al. Effects of one year of supplementation with zinc and other micronutrients on cellular immunity in the elderly. J Am Coll Nutr. 1990;9(3):214–25. https://pubmed.ncbi.nlm.nih.gov/2358617/
Duchateau J, Delepesse G, Vrijens R, Collet H. Beneficial effects of oral zinc supplementation on the immune response of old people. Am J Med. 1981;70(5):1001–4. https://pubmed.ncbi.nlm.nih.gov/6972165/
Provinciali M, Montenovo A, Di Stefano G, et al. Effect of zinc or zinc plus arginine supplementation on antibody titre and lymphocyte subsets after influenza vaccination in elderly subjects: a randomized controlled trial. Age Ageing. 1998;27(6):715–22. https://pubmed.ncbi.nlm.nih.gov/10408666/
Singh M, Das RR. Zinc for the common cold. Cochrane Database Syst Rev. 2013;(6):CD001364. https://pubmed.ncbi.nlm.nih.gov/23775705/
Davidson TM, Smith WM. The Bradford Hill criteria and zinc-induced anosmia: a causality analysis. Arch Otolaryngol Head Neck Surg. 2010;136(7):673–6. https://pubmed.ncbi.nlm.nih.gov/20644061/
Eby GA. Treatment of acute lymphocytic leukemia using zinc adjuvant with chemotherapy and radiation – a case history and hypothesis. Med Hypotheses. 2005;64(6):1124–6. https://pubmed.ncbi.nlm.nih.gov/15823699/
Centers for Disease Control and Prevention. Ten great public health achievements – United States, 1900–1999. MMWR Morb Mortal Wkly Rep. 1999;48(12):241–3. https://pubmed.ncbi.nlm.nih.gov/10220250/
Vetter V, Denizer G, Friedland LR, Krishnan J, Shapiro M. Understanding modern-day vaccines: what you need to know. Ann Med. 2018;50(2):110–20. https://pubmed.ncbi.nlm.nih.gov/29172780/
Delany I, Rappuoli R, De Gregorio E. Vaccines for the 21st century. EMBO Mol Med. 2014;6(6):708–20. https://pubmed.ncbi.nlm.nih.gov/24803000/
Slifka AM, Park B, Gao L, Slifka MK. Incidence of tetanus and diphtheria in relation to adult vaccination schedules. Clin Infect Dis. 2021;72(2):285–92. https://pubmed.ncbi.nlm.nih.gov/32095828/
National Center for Immunization and Respiratory Diseases. Adult immunization schedule by vaccine and age group: recommendations for ages 19 years or older, United States, 2022. Centers for Disease Control and Prevention. https://www.cdc.gov/vaccines/schedules/hcp/imz/adult.html. Updated February 17, 2022. Accessed May 18, 2022.; https://www.cdc.gov/vaccines/schedules/hcp/imz/adult.html
Gidengil C, Goetz MB, Newberry S, et al. Safety of vaccines used for routine immunization in the United States: an updated systematic review and meta-analysis. Vaccine. 2021;39(28):3696–716. https://pubmed.ncbi.nlm.nih.gov/34049735/
Dudley MZ, Halsey NA, Omer SB, et al. The state of vaccine safety science: systematic reviews of the evidence. Lancet Infect Dis. 2020;20(5):e80–9. https://pubmed.ncbi.nlm.nih.gov/32278359/
Rolfes MA, Foppa IM, Garg S, et al. Annual estimates of the burden of seasonal influenza in the United States: a tool for strengthening influenza surveillance and preparedness. Influenza Other Respir Viruses. 2018;12(1):132–7. https://pubmed.ncbi.nlm.nih.gov/29446233/
Hunter P, Fryhofer SA, Szilagyi PG. Vaccination of adults in general medical practice. Mayo Clin Proc. 2020;95(1):169–83. https://pubmed.ncbi.nlm.nih.gov/31902413/
Resnick B, Gravenstein S, Schaffner W, Sobczyk E, Douglas RG. Beyond prevention of influenza: the value of flu vaccines. J Gerontol A Biol Sci Med Sci. 2018;73(12):1635–7. https://pubmed.ncbi.nlm.nih.gov/30418526/
Grohskopf LA, Alyanak E, Broder KR, Walter EB, Fry AM, Jernigan DB. Prevention and control of seasonal influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices – United States, 2019–20 influenza season. MMWR Recomm Rep. 2019;68(3):1–21. https://pubmed.ncbi.nlm.nih.gov/31441906/
Hunter P, Fryhofer SA, Szilagyi PG. Vaccination of adults in general medical practice. Mayo Clin Proc. 2020;95(1):169–83. https://pubmed.ncbi.nlm.nih.gov/31902413/
Goodwin K, Viboud C, Simonsen L. Antibody response to influenza vaccination in the elderly: a quantitative review. Vaccine. 2006;24(8):1159–69. https://pubmed.ncbi.nlm.nih.gov/16213065/
Effectiveness of seasonal flu vaccines from the 2005–2021 flu seasons. Centers for Disease Control and Prevention. https://www.cdc.gov/flu/images/vaccines-work/Vaccine-Effectiveness-Graphs-2021.pptx. Updated March 11, 2022. Accessed May 23, 2022.; https://www.cdc.gov/flu/images/vaccines-work/Vaccine-Effectiveness-Graphs-2021.pptx
Demicheli V, Rivetti D, Deeks JJ, Jefferson TO. Vaccines for preventing influenza in healthy adults. Cochrane Database Syst Rev. 2004;(3):CD001269. https://pubmed.ncbi.nlm.nih.gov/15266445/
Demicheli V, Jefferson T, Di Pietrantonj C, et al. Vaccines for preventing influenza in the elderly. Cochrane Database Syst Rev. 2018;2:CD004876.4 https://pubmed.ncbi.nlm.nih.gov/29388197/
Coll PP, Costello VW, Kuchel GA, Bartley J, McElhaney JE. The prevention of infections in older adults: vaccination. J Am Geriatr Soc. 2020;68(1):207–14. https://pubmed.ncbi.nlm.nih.gov/31613000/
Influenza vaccine for 2019–2020. Med Lett Drugs Ther. 2019;61(1583):161–6. https://pubmed.ncbi.nlm.nih.gov/31770355/
Coll PP, Costello VW, Kuchel GA, Bartley J, McElhaney JE. The prevention of infections in older adults: vaccination. J Am Geriatr Soc. 2020;68(1):207–14. https://pubmed.ncbi.nlm.nih.gov/31613000/
Dudley MZ, Halsey NA, Omer SB, et al. The state of vaccine safety science: systematic reviews of the evidence. Lancet Infect Dis. 2020;20(5):e80–9. https://pubmed.ncbi.nlm.nih.gov/32278359/
Influenza vaccine for 2019–2020. Med Lett Drugs Ther. 2019;61(1583):161–6. https://pubmed.ncbi.nlm.nih.gov/31770355/
Demicheli V, Jefferson T, Di Pietrantonj C, et al. Vaccines for preventing influenza in the elderly. Cochrane Database Syst Rev. 2018;2:CD004876.4 https://pubmed.ncbi.nlm.nih.gov/29388197/
Kwong JC, Schwartz KL, Campitelli MA, et al. Acute myocardial infarction after laboratory-confirmed influenza infection. N Engl J Med. 2018;378(4):345–53. https://pubmed.ncbi.nlm.nih.gov/29365305/
Corrales-Medina VF, Madjid M, Musher DM. Role of acute infection in triggering acute coronary syndromes. Lancet Infect Dis. 2010;10(2):83–92. https://pubmed.ncbi.nlm.nih.gov/20113977/
Cheng Y, Cao X, Cao Z, et al. Effects of influenza vaccination on the risk of cardiovascular and respiratory diseases and all-cause mortality. Ageing Res Rev. 2020;62:101124. https://pubmed.ncbi.nlm.nih.gov/32683040/
Okoli GN, Lam OLT, Racovitan F, et al. Seasonal influenza vaccination in older people: a systematic review and meta-analysis of the determining factors. PLoS One. 2020;15(6):e0234702. https://pubmed.ncbi.nlm.nih.gov/32555628/
Yedlapati SH, Khan SU, Talluri S, et al. Effects of influenza vaccine on mortality and cardiovascular outcomes in patients with cardiovascular disease: a systematic review and meta-analysis. J Am Heart Assoc. 2021;10(6):e019636. https://pubmed.ncbi.nlm.nih.gov/33719496/
Clar C, Oseni Z, Flowers N, Keshtkar-Jahromi M, Rees K. Influenza vaccines for preventing cardiovascular disease. Cochrane Database Syst Rev. 2015;(5):CD005050. https://pubmed.ncbi.nlm.nih.gov/25940444/
Nyhan B, Reifler J. Does correcting myths about the flu vaccine work? An experimental evaluation of the effects of corrective information. Vaccine. 2015;33(3):459–64. https://pubmed.ncbi.nlm.nih.gov/25499651/
Nyhan B, Reifler J, Richey S, Freed GL. Effective messages in vaccine promotion: a randomized trial. Pediatrics. 2014;133(4):e835–42. https://pubmed.ncbi.nlm.nih.gov/24590751/
Meszaros JR, Asch DA, Baron J, Hershey JC, Kunreuther H, Schwartz-Buzaglo J. Cognitive processes and the decisions of some parents to forego pertussis vaccination for their children. J Clin Epidemiol. 1996;49(6):697–703. https://pubmed.ncbi.nlm.nih.gov/8656233/
Nyhan B, Reifler J. Does correcting myths about the flu vaccine work? An experimental evaluation of the effects of corrective information. Vaccine. 2015;33(3):459–64. https://pubmed.ncbi.nlm.nih.gov/25499651/
. “Father of modern medicine”: the Johns Hopkins School of Medicine, 1889–1905. William Osler-Profiles in Science. NIH National Library of Medicine. https://profiles.nlm.nih.gov/spotlight/gf/feature/father-of-modern-medicine-the-johns-hopkins-school-of-medicine-1889-1905. Accessed May 20, 2022.; https://profiles.nlm.nih.gov/spotlight/gf/feature/father-of-modern-medicine-the-johns-hopkins-school-of-medicine-1889-1905
Brancati FL, Chow JW, Wagener MM, Vacarello SJ, Yu VL. Is pneumonia really the old man’s friend? Two-year prognosis after community-acquired pneumonia. Lancet. 1993;342(8862):30–3. https://pubmed.ncbi.nlm.nih.gov/8100295/
The top 10 causes of death. World Health Organization. https://www.who.int/en/news-room/fact-sheets/detail/the-top-10-causes-of-death. Published December 9, 2022. Accessed May 20, 2022.; https://www.who.int/en/news-room/fact-sheets/detail/the-top-10-causes-of-death
Heron M. Deaths: leading causes for 2019. Natl Vital Stat Rep. 2021;70(9):1–114. https://pubmed.ncbi.nlm.nih.gov/34520342/
van Werkhoven CH, Huijts SM. Vaccines to prevent pneumococcal community-acquired pneumonia. Clin Chest Med. 2018;39(4):733–52. https://pubmed.ncbi.nlm.nih.gov/30390745/
van Werkhoven CH, Huijts SM. Vaccines to prevent pneumococcal community-acquired pneumonia. Clin Chest Med. 2018;39(4):733–52. https://pubmed.ncbi.nlm.nih.gov/30390745/
US Department of Health and Human Services. Antibiotics resistance threats in the United States: 2019. https://www.cdc.gov/drugresistance/pdf/threats-report/2019-ar-threats-report-508.pdf. Centers for Disease Control and Prevention, US Dept of Health and Human Services. Updated December 2019. Accessed May 20, 2022.; https://www.cdc.gov/drugresistance/pdf/threats-report/2019-ar-threats-report-508.pdf
Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. Immun Ageing. 2019;16:25. https://pubmed.ncbi.nlm.nih.gov/31528180/
Thomas RE. Pneumococcal pneumonia and invasive pneumococcal disease in those 65 and older: rates of detection, risk factors, vaccine effectiveness, hospitalisation and mortality. Geriatrics (Basel). 2021;6(1):13. https://pubmed.ncbi.nlm.nih.gov/33557406/
Jaiswal V, Ang SP, Lnu K, et al. Effect of pneumococcal vaccine on mortality and cardiovascular outcomes: a systematic review and meta-analysis. J Clin Med. 2022;11(13):3799. https://pubmed.ncbi.nlm.nih.gov/35807082/
Ecarnot F, Bernabei R, Gabutti G, et al. Adult vaccination as the cornerstone of successful ageing: the case of herpes zoster vaccination. A European Interdisciplinary Council on Ageing (EICA) expert focus group. Aging Clin Exp Res. 2019;31(3):301–7. https://pubmed.ncbi.nlm.nih.gov/30805865/
McElhaney JE, Verschoor C, Pawelec G. Zoster vaccination in older adults: efficacy and public health implications. J Gerontol A Biol Sci Med Sci. 2019;74(8):1239–43. https://pubmed.ncbi.nlm.nih.gov/30945744/
Gagliardi AMZ, Andriolo BNG, Torloni MR, et al. Vaccines for preventing herpes zoster in older adults. Cochrane Database Syst Rev. 2019;2019(11):CD008858. https://pubmed.ncbi.nlm.nih.gov/26937872/
Schmader K. Herpes zoster. Ann Intern Med. 2018;169(3):ITC17. https://pubmed.ncbi.nlm.nih.gov/30083718/
Schmader K. Herpes zoster. Ann Intern Med. 2018;169(3):ITC17. https://pubmed.ncbi.nlm.nih.gov/30083718/
Zuin M, Rigatelli G, Adami A. Cerebrovascular events after herpes zoster infection: a risk that should be not underestimated. J Neurovirol. 2019;25(4):439–47. https://pubmed.ncbi.nlm.nih.gov/31069708/
Johnson RW, Levin MJ. Herpes zoster and its prevention by vaccination. Interdiscip Top Gerontol Geriatr. 2020;43:131–45. https://pubmed.ncbi.nlm.nih.gov/32305975/
Coll PP, Costello VW, Kuchel GA, Bartley J, McElhaney JE. The prevention of infections in older adults: vaccination. J Am Geriatr Soc. 2020;68(1):207–14. https://pubmed.ncbi.nlm.nih.gov/31613000/
Shafran SD. Prevention of shingles: better protection and better value with recombinant vaccine. Ann Intern Med. 2019;170(6):416–7. https://pubmed.ncbi.nlm.nih.gov/30776802/
Schmader K. Herpes zoster. Ann Intern Med. 2018;169(3):ITC17. https://pubmed.ncbi.nlm.nih.gov/30083718/
Shafran SD. Prevention of shingles: better protection and better value with recombinant vaccine. Ann Intern Med. 2019;170(6):416–7. https://pubmed.ncbi.nlm.nih.gov/30776802/
Johnson RW, Levin MJ. Herpes zoster and its prevention by vaccination. Interdiscip Top Gerontol Geriatr. 2020;43:131–45. https://pubmed.ncbi.nlm.nih.gov/32305975/
Le P. Which shingles vaccine for older adults? BMJ. 2018;363:k4203. https://pubmed.ncbi.nlm.nih.gov/30361204/
Thijssen E, van Caam A, van der Kraan PM. Obesity and osteoarthritis, more than just wear and tear: pivotal roles for inflamed adipose tissue and dyslipidaemia in obesity-induced osteoarthritis. Rheumatology (Oxford). 2015;54(4):588–600. https://pubmed.ncbi.nlm.nih.gov/25504962/
Kulkarni K, Karssiens T, Kumar V, Pandit H. Obesity and osteoarthritis. Maturitas. 2016;89:22–8. https://pubmed.ncbi.nlm.nih.gov/27180156/
Charlesworth J, Fitzpatrick J, Perera NKP, Orchard J. Osteoarthritis – a systematic review of long-term safety implications for osteoarthritis of the knee. BMC Musculoskelet Disord. 2019;20(1):151. https://pubmed.ncbi.nlm.nih.gov/30961569/
Berenbaum F, Walker C. Osteoarthritis and inflammation: a serious disease with overlapping phenotypic patterns. Postgrad Med. 2020;132(4):377–84. https://pubmed.ncbi.nlm.nih.gov/32100608/
Gress K, Charipova K, An D, et al. Treatment recommendations for chronic knee osteoarthritis. Best Pract Res Clin Anaesthesiol. 2020;34(3):369–82. https://pubmed.ncbi.nlm.nih.gov/33004154/
Leopoldino AO, Machado GC, Ferreira PH, et al. Paracetamol versus placebo for knee and hip osteoarthritis. Cochrane Database Syst Rev. 2019;2019(2):CD013273. https://pubmed.ncbi.nlm.nih.gov/30801133/
Negrini F, Negrini S. Is paracetamol better than placebo for knee and hip osteoarthritis? A Cochrane review summary with commentary. Int J Rheum Dis. 2020;23(4):595–6. https://pubmed.ncbi.nlm.nih.gov/32286015/
Leopoldino AO, Machado GC, Ferreira PH, et al. Paracetamol versus placebo for knee and hip osteoarthritis. Cochrane Database Syst Rev. 2019;2019(2):CD013273. https://pubmed.ncbi.nlm.nih.gov/30801133/
Negrini F, Negrini S. Is paracetamol better than placebo for knee and hip osteoarthritis? A Cochrane review summary with commentary. Int J Rheum Dis. 2020;23(4):595–6. https://pubmed.ncbi.nlm.nih.gov/32286015/
Bunchorntavakul C, Reddy KR. Acetaminophen (APAP or N-acetyl-p-aminophenol) and acute liver failure. Clin Liver Dis. 2018;22(2):325–46. https://pubmed.ncbi.nlm.nih.gov/29605069/
Conaghan PG, Arden N, Avouac B, Migliore A, Rizzoli R. Safety of paracetamol in osteoarthritis: what does the literature say? Drugs Aging. 2019;36(Suppl 1):7–14. https://pubmed.ncbi.nlm.nih.gov/31073920/
Machado GC, Maher CG, Ferreira PH, et al. Efficacy and safety of paracetamol for spinal pain and osteoarthritis: systematic review and meta-analysis of randomised placebo controlled trials. BMJ. 2015;350:h1225. https://pubmed.ncbi.nlm.nih.gov/25828856/
Berenbaum F, Walker C. Osteoarthritis and inflammation: a serious disease with overlapping phenotypic patterns. Postgrad Med. 2020;132(4):377–84. https://pubmed.ncbi.nlm.nih.gov/32100608/
Chow YY, Chin KY. The role of inflammation in the pathogenesis of osteoarthritis. Mediators Inflamm. 2020;2020:8293921. https://pubmed.ncbi.nlm.nih.gov/32189997/
Cooper C, Chapurlat R, Al-Daghri N, et al. Safety of oral non-selective non-steroidal anti-inflammatory drugs in osteoarthritis: what does the literature say? Drugs Aging. 2019;36(Suppl 1):15–24. https://pubmed.ncbi.nlm.nih.gov/31073921/
Ho KY, Cardosa MS, Chaiamnuay S, et al. Practice advisory on the appropriate use of NSAIDs in primary care. J Pain Res. 2020;13:1925–39. https://pubmed.ncbi.nlm.nih.gov/32821151/
Cooper C, Chapurlat R, Al-Daghri N, et al. Safety of oral non-selective non-steroidal anti-inflammatory drugs in osteoarthritis: what does the literature say? Drugs Aging. 2019;36(Suppl 1):15–24. https://pubmed.ncbi.nlm.nih.gov/31073921/
Ong CKS, Lirk P, Tan CH, Seymour RA. An evidence-based update on nonsteroidal anti-inflammatory drugs. Clin Med Res. 2007;5(1):19–34. https://pubmed.ncbi.nlm.nih.gov/17456832/
Bally M, Dendukuri N, Rich B, et al. Risk of acute myocardial infarction with NSAIDs in real world use: bayesian meta-analysis of individual patient data. BMJ. 2017;357:j1909. https://pubmed.ncbi.nlm.nih.gov/28487435/
Cooper C, Chapurlat R, Al-Daghri N, et al. Safety of oral non-selective non-steroidal anti-inflammatory drugs in osteoarthritis: what does the literature say? Drugs Aging. 2019;36(Suppl 1):15–24. https://pubmed.ncbi.nlm.nih.gov/31073921/
Zhang X, Donnan PT, Bell S, Guthrie B. Non-steroidal anti-inflammatory drug induced acute kidney injury in the community dwelling general population and people with chronic kidney disease: systematic review and meta-analysis. BMC Nephrol. 2017;18(1):256. https://pubmed.ncbi.nlm.nih.gov/28764659/
American Geriatrics Society recommends opioids as second-line therapy for chronic pain, instead of NSAIDs. Topics in Pain Management. 2009;25(1):9–10. https://journals.lww.com/topicsinpainmanagement/Citation/2009/08000/American_Geriatrics_Society_Recommends_Opioids_as.3.aspx
Cooper C, Chapurlat R, Al-Daghri N, et al. Safety of oral non-selective non-steroidal anti-inflammatory drugs in osteoarthritis: what does the literature say? Drugs Aging. 2019;36(Suppl 1):15–24. https://pubmed.ncbi.nlm.nih.gov/31073921/
Nissen SE. Cardiovascular safety of celecoxib, naproxen, or ibuprofen for arthritis. N Engl J Med. 2017;376(14):1390. https://pubmed.ncbi.nlm.nih.gov/28379793/
Ho KY, Cardosa MS, Chaiamnuay S, et al. Practice advisory on the appropriate use of NSAIDs in primary care. J Pain Res. 2020;13:1925–39. https://pubmed.ncbi.nlm.nih.gov/32821151/
Reginster JY, Bruyère O, Conaghan PG, McAlindon T, Cooper C. Importance of safety in the management of osteoarthritis and the need for updated meta-analyses and recommendations for reporting of harms. Drugs Aging. 2019;36(Suppl 1):3–6. https://pubmed.ncbi.nlm.nih.gov/31073919/
FDA approves three drugs for nonprescription use through Rx-to-OTC switch process. U.S. Food & Drug Administration. https://www.fda.gov/news-events/press-announcements/fda-approves-three-drugs-nonprescription-use-through-rx-otc-switch-process. Published February 14, 2020. Accessed June 1, 2022.; https://www.fda.gov/news-events/press-announcements/fda-approves-three-drugs-nonprescription-use-through-rx-otc-switch-process
Derry S, Conaghan P, Da Silva JAP, Wiffen PJ, Moore RA. Topical NSAIDs for chronic musculoskeletal pain in adults. Cochrane Database Syst Rev. 2016;4:CD007400. https://pubmed.ncbi.nlm.nih.gov/27103611/
Reginster JY, Bruyère O, Conaghan PG, McAlindon T, Cooper C. Importance of safety in the management of osteoarthritis and the need for updated meta-analyses and recommendations for reporting of harms. Drugs Aging. 2019;36(Suppl 1):3–6. https://pubmed.ncbi.nlm.nih.gov/31073919/
Honvo G, Leclercq V, Geerinck A, et al. Safety of topical non-steroidal anti-inflammatory drugs in osteoarthritis: outcomes of a systematic review and meta-analysis. Drugs Aging. 2019;36(Suppl 1):45–64. https://pubmed.ncbi.nlm.nih.gov/31073923/
Lim CC, Ang ATW, Kadir HBA, et al. Short-course systemic and topical non-steroidal anti-inflammatory drugs: impact on adverse renal events in older adults with co-morbid disease. Drugs Aging. 2021;38(2):147–56. https://pubmed.ncbi.nlm.nih.gov/33251568/
Lin T, Solomon DH, Tedeschi SK, Yoshida K, Kao Yang Y. Comparative risk of cardiovascular outcomes between topical and oral nonselective NSAIDs in Taiwanese patients with rheumatoid arthritis. JAHA. 2017;6(11):e006874. https://pubmed.ncbi.nlm.nih.gov/29079568/
Koenig KM, Ong KL, Lau EC, et al. The use of hyaluronic acid and corticosteroid injections among Medicare patients with knee osteoarthritis. J Arthroplasty. 2016;31(2):351–5. https://pubmed.ncbi.nlm.nih.gov/26421601/
Latourte A, Lellouche H. Update on corticosteroid, hyaluronic acid and platelet-rich plasma injections in the management of osteoarthritis. Joint Bone Spine. 2021;88(6):105204. https://pubmed.ncbi.nlm.nih.gov/33962034/
McAlindon TE, LaValley MP, Harvey WF, et al. Effect of intra-articular triamcinolone vs saline on knee cartilage volume and pain in patients with knee osteoarthritis. JAMA. 2017;317(19):1967–75. https://pubmed.ncbi.nlm.nih.gov/28510679/
Orchard JW. Is there a place for intra-articular corticosteroid injections in the treatment of knee osteoarthritis? BMJ. 2020;368:l6923. https://pubmed.ncbi.nlm.nih.gov/31941647/
Kompel AJ, Roemer FW, Murakami AM, Diaz LE, Crema MD, Guermazi A. Intra-articular corticosteroid injections in the hip and knee: perhaps not as safe as we thought? Radiology. 2019;293(3):656–63. https://pubmed.ncbi.nlm.nih.gov/31617798/
McAlindon TE, LaValley MP, Harvey WF, et al. Effect of intra-articular triamcinolone vs saline on knee cartilage volume and pain in patients with knee osteoarthritis. JAMA. 2017;317(19):1967–75. https://pubmed.ncbi.nlm.nih.gov/28510679/
Bliddal H, Henriksen M. Osteoarthritis: time to put steroid injections behind us? Nat Rev Rheumatol. 2017;13(9):519–20. https://pubmed.ncbi.nlm.nih.gov/28747801/
Jevsevar DS. Treatment of osteoarthritis of the knee: evidence-based guideline, 2nd edition. J Am Acad Orthop Surg. 2013;21(9):571–6. https://pubmed.ncbi.nlm.nih.gov/23996988/
Moseley JB, O’Malley K, Petersen NJ, et al. A controlled trial of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med. 2002;347(2):81–8. https://pubmed.ncbi.nlm.nih.gov/12110735/
Maffulli N. We are operating too much. J Orthop Traumatol. 2017;18(4):289–92. https://pubmed.ncbi.nlm.nih.gov/28879556/
Moseley JB, O’Malley K, Petersen NJ, et al. A controlled trial of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med. 2002;347(2):81–8. https://pubmed.ncbi.nlm.nih.gov/12110735/
McCormack RG, Hutchinson MR. Rocking the shoulder surgeon’s world. Br J Sports Med. 2017;51(24):1727. https://pubmed.ncbi.nlm.nih.gov/28646099/
Horton R. Surgical research or comic opera: questions, but few answers. Lancet. 1996;347(9007):984–5. https://pubmed.ncbi.nlm.nih.gov/8606606/
Lim HC, Adie S, Naylor JM, Harris IA. Randomised trial support for orthopaedic surgical procedures. PLoS One. 2014;9(6):e96745. https://pubmed.ncbi.nlm.nih.gov/24927114/
Wartolowska K, Judge A, Hopewell S, et al. Use of placebo controls in the evaluation of surgery: systematic review. BMJ. 2014;348:g3253. https://pubmed.ncbi.nlm.nih.gov/24850821/
Thorlund JB, Juhl CB, Roos EM, Lohmander LS. Arthroscopic surgery for degenerative knee: systematic review and meta-analysis of benefits and harms. BMJ. 2015;350:h2747. https://pubmed.ncbi.nlm.nih.gov/26080045/
Rongen JJ, Rovers MM, van Tienen TG, Buma P, Hannink G. Increased risk for knee replacement surgery after arthroscopic surgery for degenerative meniscal tears: a multi-center longitudinal observational study using data from the osteoarthritis initiative. Osteoarthritis Cartilage. 2017;25(1):23–9. https://pubmed.ncbi.nlm.nih.gov/27712957/
Moseley JB, O’Malley K, Petersen NJ, et al. A controlled trial of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med. 2002;347(2):81–8. https://pubmed.ncbi.nlm.nih.gov/12110735/
Thorlund JB. Deconstructing a popular myth: why knee arthroscopy is no better than placebo surgery for degenerative meniscal tears. Br J Sports Med. 2017;51(22):1630–1. https://pubmed.ncbi.nlm.nih.gov/28615215/
Zhang W. The powerful placebo effect in osteoarthritis. Clin Exp Rheumatol. 2019;37 Suppl 120(5):118–23. https://pubmed.ncbi.nlm.nih.gov/31621561/
Radiation dose. RadiologyInfo.org. https://www.radiologyinfo.org/en/info/safety-xray#81426d679d504b8f9a595b2b5d7d1eff. Published February 1, 2021. Accessed May 31, 2022.; https://www.radiologyinfo.org/en/info/safety-xray#81426d679d504b8f9a595b2b5d7d1eff
Mahler EAM, Minten MJM, Leseman-Hoogenboom MM, et al. Effectiveness of low-dose radiation therapy on symptoms in patients with knee osteoarthritis: a randomised, double-blinded, sham-controlled trial. Ann Rheum Dis. 2019;78(1):83–90. https://pubmed.ncbi.nlm.nih.gov/30366945/
Blackwell B, Bloomfield SS, Buncher CR. Demonstration to medical students of placebo responses and non-drug factors. Lancet. 1972;1(7763):1279–82. https://pubmed.ncbi.nlm.nih.gov/4113531/
de Craen AJM, Roos PJ, de Vries AL, Kleijnen J. Effect of colour of drugs: systematic review of perceived effect of drugs and of their effectiveness. BMJ. 1996;313(7072):1624–6. https://pubmed.ncbi.nlm.nih.gov/8991013/
Branthwaite A, Cooper P. Analgesic effects of branding in treatment of headaches. BMJ. 1981;282(6276):1576–8. https://pubmed.ncbi.nlm.nih.gov/6786566/
Waber RL, Shiv B, Carmon Z, Ariely D. Commercial features of placebo and therapeutic efficacy. JAMA. 2008;299(9):1016–7. https://pubmed.ncbi.nlm.nih.gov/18319411/
Zhang W, Zou K, Doherty M. Placebos for knee osteoarthritis: reaffirmation of “needle is better than pill.” Ann Intern Med. 2015;163(5):392–3. https://pubmed.ncbi.nlm.nih.gov/26215902/
Previtali D, Merli G, Di Laura Frattura G, Candrian C, Zaffagnini S, Filardo G. The long-lasting effects of “placebo injections” in knee osteoarthritis: a meta-analysis. Cartilage. 2021;13(1_suppl):185S-96S. https://pubmed.ncbi.nlm.nih.gov/32186401/
Zhang W. The powerful placebo effect in osteoarthritis. Clin Exp Rheumatol. 2019;37 Suppl 120(5):118–23. https://pubmed.ncbi.nlm.nih.gov/31621561/
Kaptchuk TJ, Stason WB, Davis RB, et al. Sham device v inert pill: randomised controlled trial of two placebo treatments. BMJ. 2006;332(7538):391–7. https://pubmed.ncbi.nlm.nih.gov/16452103/
Jevsevar DS. Treatment of osteoarthritis of the knee: evidence-based guideline, 2nd edition. J Am Acad Orthop Surg. 2013;21(9):571–6. https://pubmed.ncbi.nlm.nih.gov/23996988/
Starfield B. Is US health really the best in the world? JAMA. 2000;284(4):483–5. https://pubmed.ncbi.nlm.nih.gov/10904513/
Fässler M, Meissner K, Schneider A, Linde K. Frequency and circumstances of placebo use in clinical practice – a systematic review of empirical studies. BMC Med. 2010;8:15. https://pubmed.ncbi.nlm.nih.gov/20178561/
Zhang W. The powerful placebo effect in osteoarthritis. Clin Exp Rheumatol. 2019;37 Suppl 120(5):118–23. https://pubmed.ncbi.nlm.nih.gov/31621561/
Plato. The Republic. The Project Gutenberg. https://www.gutenberg.org/files/1497/1497-h/1497-h.htm. Published October 1998. Updated September 11, 2021. Accessed June 5, 2022.; https://www.gutenberg.org/files/1497/1497-h/1497-h.htm
Doherty M, Dieppe P. The “placebo” response in osteoarthritis and its implications for clinical practice. Osteoarthritis Cartilage. 2009;17(10):1255–62. https://pubmed.ncbi.nlm.nih.gov/19410027/
The humble humbug. Lancet. 1954;264(6833):321. https://sciencedirect.com/science/article/abs/pii/S0140673654902457
de Campos GC. Placebo effect in osteoarthritis: why not use it to our advantage? World J Orthop. 2015;6(5):416–20. https://pubmed.ncbi.nlm.nih.gov/26085983/
Leslie A. Ethics and practice of placebo therapy. Am J Med. 1954;16(6):854–62. https://pubmed.ncbi.nlm.nih.gov/13158374/
Bortoluzzi A, Furini F, Scirè CA. Osteoarthritis and its management – Epidemiology, nutritional aspects and environmental factors. Autoimmun Rev. 2018;17(11):1097–104. https://pubmed.ncbi.nlm.nih.gov/30213694/
Kulkarni K, Karssiens T, Kumar V, Pandit H. Obesity and osteoarthritis. Maturitas. 2016;89:22–8. https://pubmed.ncbi.nlm.nih.gov/27180156/
Berenbaum F, Wallace IJ, Lieberman DE, Felson DT. Modern-day environmental factors in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2018;14(11):674–81. https://pubmed.ncbi.nlm.nih.gov/30209413/
Bortoluzzi A, Furini F, Scirè CA. Osteoarthritis and its management – Epidemiology, nutritional aspects and environmental factors. Autoimmun Rev. 2018;17(11):1097–104. https://pubmed.ncbi.nlm.nih.gov/30213694/
Issa RI, Griffin TM. Pathobiology of obesity and osteoarthritis: integrating biomechanics and inflammation. Pathobiol Aging Age Relat Dis. 2012;2(2012):17470. https://pubmed.ncbi.nlm.nih.gov/22662293/
Felson DT, Zhang Y, Anthony JM, Naimark A, Anderson JJ. Weight loss reduces the risk for symptomatic knee osteoarthritis in women. Framingham Study. Ann Intern Med. 1992;116(7):535–9. https://pubmed.ncbi.nlm.nih.gov/1543306/
Gersing AS, Schwaiger BJ, Nevitt MC, et al. Is weight loss associated with less progression of changes in knee articular cartilage among obese and overweight patients as assessed with MR imaging over 48 months? Data from the Osteoarthritis Initiative. Radiology. 2017;284(2):508–20. https://pubmed.ncbi.nlm.nih.gov/28463057/
Christensen R, Astrup A, Bliddal H. Weight loss: the treatment of choice for knee osteoarthritis? A randomized trial. Osteoarthr Cartil. 2005;13(1):20–7. https://pubmed.ncbi.nlm.nih.gov/15639633/
Bernstein J. Not the last word: safety alert: one in 200 knee replacement patients die within 90 days of surgery. Clin Orthop Relat Res. 2017;475(2):318–23. https://pubmed.ncbi.nlm.nih.gov/27942969/
Albanes D, Jones DY, Micozzi MS, Mattson ME. Associations between smoking and body weight in the US population: analysis of NHANES II. Am J Public Health. 1987;77(4):439–44. https://pubmed.ncbi.nlm.nih.gov/3493709/
Hui M, Doherty M, Zhang W. Does smoking protect against osteoarthritis? Meta-analysis of observational studies. Ann Rheum Dis. 2011;70(7):1231–7. https://pubmed.ncbi.nlm.nih.gov/21474488/
Amin S, Niu J, Guermazi A, et al. Cigarette smoking and the risk for cartilage loss and knee pain in men with knee osteoarthritis. Ann Rheum Dis. 2007;66(1):18–22. https://pubmed.ncbi.nlm.nih.gov/17158140/
Järvholm B, Lewold S, Malchau H, Vingård E. Age, bodyweight, smoking habits and the risk of severe osteoarthritis in the hip and knee in men. Eur J Epidemiol. 2005;20(6):537–42. https://pubmed.ncbi.nlm.nih.gov/16121763/
Palmieri-Smith RM, Cameron KL, DiStefano LJ, et al. The role of athletic trainers in preventing and managing posttraumatic osteoarthritis in physically active populations: a consensus statement of the Athletic Trainers’ Osteoarthritis Consortium. J Athl Train. 2017;52(6):610–23. https://pubmed.ncbi.nlm.nih.gov/28653866/
Blackwell B, Bloomfield SS, Buncher CR. Demonstration to medical students of placebo responses and non-drug factors. Lancet. 1972;1(7763):1279–82. https://pubmed.ncbi.nlm.nih.gov/4113531/
Racunica TL, Teichtahl AJ, Wang Y, et al. Effect of physical activity on articular knee joint structures in community-based adults. Arthritis Rheum. 2007;57(7):1261–8. https://pubmed.ncbi.nlm.nih.gov/17907212/
Berenbaum F, Wallace IJ, Lieberman DE, Felson DT. Modern-day environmental factors in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2018;14(11):674–81. https://pubmed.ncbi.nlm.nih.gov/30209413/
Juhl C, Christensen R, Roos EM, Zhang W, Lund H. Impact of exercise type and dose on pain and disability in knee osteoarthritis: a systematic review and meta-regression analysis of randomized controlled trials. Arthritis Rheumatol. 2014;66(3):622–36. https://pubmed.ncbi.nlm.nih.gov/24574223/
Verhagen AP, Ferreira M, Reijneveld-van de Vendel EAE, et al. Do we need another trial on exercise in patients with knee osteoarthritis?: No new trials on exercise in knee OA. Osteoarthritis Cartilage. 2019;27(9):1266–9. https://pubmed.ncbi.nlm.nih.gov/31220609/
Juhl C, Christensen R, Roos EM, Zhang W, Lund H. Impact of exercise type and dose on pain and disability in knee osteoarthritis: a systematic review and meta-regression analysis of randomized controlled trials. Arthritis Rheumatol. 2014;66(3):622–36. https://pubmed.ncbi.nlm.nih.gov/24574223/
Verhagen AP, Ferreira M, Reijneveld-van de Vendel EAE, et al. Do we need another trial on exercise in patients with knee osteoarthritis?: No new trials on exercise in knee OA. Osteoarthritis Cartilage. 2019;27(9):1266–9. https://pubmed.ncbi.nlm.nih.gov/31220609/
Charlesworth J, Fitzpatrick J, Perera NKP, Orchard J. Osteoarthritis – a systematic review of long-term safety implications for osteoarthritis of the knee. BMC Musculoskelet Disord. 2019;20(1):151. https://pubmed.ncbi.nlm.nih.gov/30961569/
Dean E, Gormsen Hansen R. Prescribing optimal nutrition and physical activity as “first-line” interventions for best practice management of chronic low-grade inflammation associated with osteoarthritis: evidence synthesis. Arthritis. 2012;2012:1–28. https://pubmed.ncbi.nlm.nih.gov/23346399/
На русском языке вышла в 2020 году в издательстве «Манн, Иванов, Фербер».
Campbell TC. History of the term ‘whole food, plant based.’ T. Colin Campbell Center for Nutrition Studies. https://nutritionstudies.org/history-of-the-term-whole-food-plant-based/. Published November 29, 2016. Updated January 4, 2019. Accessed June 5, 2022.; https://nutritionstudies.org/history-of-the-term-whole-food-plant-based
Wang Y, Teichtahl AJ, Abram F, et al. Knee pain as a predictor of structural progression over 4 years: data from the Osteoarthritis Initiative, a prospective cohort study. Arthritis Res Ther. 2018;20(1):250. https://pubmed.ncbi.nlm.nih.gov/30400973/
Lu B, Driban JB, Xu C, Lapane KL, McAlindon TE, Eaton CB. Dietary fat intake and radiographic progression of knee osteoarthritis: data from the Osteoarthritis Initiative. Arthritis Care Res (Hoboken). 2017;69(3):368–75. https://pubmed.ncbi.nlm.nih.gov/27273934/
Sekar S, Shafie SR, Prasadam I, et al. Saturated fatty acids induce development of both metabolic syndrome and osteoarthritis in rats. Sci Rep. 2017;7:46457. https://pubmed.ncbi.nlm.nih.gov/28418007/
Shen P, Zhu Y, Zhu L, Weng F, Li X, Xu Y. Oxidized low density lipoprotein facilitates tumor necrosis factor-a mediated chondrocyte death via autophagy pathway. Mol Med Rep. 2017;16(6):9449–56. https://pubmed.ncbi.nlm.nih.gov/29039543/
Wu CL, Jain D, McNeill JN, et al. Dietary fatty acid content regulates wound repair and the pathogenesis of osteoarthritis following joint injury. Ann Rheum Dis. 2015;74(11):2076–83. https://pubmed.ncbi.nlm.nih.gov/25015373/
Beier F. Cholesterol and cartilage do not mix well. Nat Rev Rheumatol. 2019;15(5):253–4. https://pubmed.ncbi.nlm.nih.gov/30914770/
Sekar S, Shafie SR, Prasadam I, et al. Saturated fatty acids induce development of both metabolic syndrome and osteoarthritis in rats. Sci Rep. 2017;7:46457. https://pubmed.ncbi.nlm.nih.gov/28418007/
Ertürk C, Altay MA, Bilge A, Çelik H. Is there a relationship between serum ox-LDL, oxidative stress, and PON1 in knee osteoarthritis? Clin Rheumatol. 2017;36(12):2775–80. https://pubmed.ncbi.nlm.nih.gov/28631083/
Shen P, Zhu Y, Zhu L, Weng F, Li X, Xu Y. Oxidized low density lipoprotein facilitates tumor necrosis factor-a mediated chondrocyte death via autophagy pathway. Mol Med Rep. 2017;16(6):9449–56. https://pubmed.ncbi.nlm.nih.gov/29039543/
Cillero-Pastor B, Eijkel G, Kiss A, Blanco FJ, Heeren RMA. Time-of-flight secondary ion mass spectrometry-based molecular distribution distinguishing healthy and osteoarthritic human cartilage. Anal Chem. 2012;84(21):8909–16. https://pubmed.ncbi.nlm.nih.gov/22950553/
Shen P, Zhu Y, Zhu L, Weng F, Li X, Xu Y. Oxidized low density lipoprotein facilitates tumor necrosis factor-a mediated chondrocyte death via autophagy pathway. Mol Med Rep. 2017;16(6):9449–56. https://pubmed.ncbi.nlm.nih.gov/29039543/
Ertürk C, Altay MA, Bilge A, Çelik H. Is there a relationship between serum ox-LDL, oxidative stress, and PON1 in knee osteoarthritis? Clin Rheumatol. 2017;36(12):2775–80. https://pubmed.ncbi.nlm.nih.gov/28631083/
Wang J, Dong J, Yang J, Wang Y, Liu J. Association between statin use and incidence or progression of osteoarthritis: meta-analysis of observational studies. Osteoarthritis Cartilage. 2020;28(9):1170–9. https://pubmed.ncbi.nlm.nih.gov/32360737/
Haj-Mirzaian A, Mohajer B, Guermazi A, et al. Statin use and knee osteoarthritis outcome measures according to the presence of Heberden nodes: results from the Osteoarthritis Initiative. Radiology. 2019;293(2):396–404. https://pubmed.ncbi.nlm.nih.gov/31502936/
Clockaerts S, Van Osch GJVM, Bastiaansen-Jenniskens YM, et al. Statin use is associated with reduced incidence and progression of knee osteoarthritis in the Rotterdam study. Ann Rheum Dis. 2012;71(5):642–7. https://pubmed.ncbi.nlm.nih.gov/21989540/
Peeters G, Tett SE, Conaghan PG, Mishra GD, Dobson AJ. Is statin use associated with new joint-related symptoms, physical function, and quality of life? Results from two population-based cohorts of women. Arthritis Care Res (Hoboken). 2015;67(1):13–20. https://pubmed.ncbi.nlm.nih.gov/24964875/
Veronese N, Koyanagi A, Stubbs B, et al. Statin use and knee osteoarthritis outcomes: a longitudinal cohort study. Arthritis Care Res (Hoboken). 2019;71(8):1052–8. https://pubmed.ncbi.nlm.nih.gov/30144308/
Eymard F, Parsons C, Edwards MH, et al. Statin use and knee osteoarthritis progression: results from a post-hoc analysis of the SEKOIA trial. Joint Bone Spine. 2018;85(5):609–14. https://pubmed.ncbi.nlm.nih.gov/29037516/
Beattie MS, Lane NE, Hung YY, Nevitt MC. Association of statin use and development and progression of hip osteoarthritis in elderly women. J Rheumatol. 2005;32(1):106–10. https://pubmed.ncbi.nlm.nih.gov/15630734/
Wang J, Dong J, Yang J, Wang Y, Liu J. Association between statin use and incidence or progression of osteoarthritis: meta-analysis of observational studies. Osteoarthritis Cartilage. 2020;28(9):1170–9. https://pubmed.ncbi.nlm.nih.gov/32360737/
Kendall CWC, Jenkins DJA. A dietary portfolio: maximal reduction of low-density lipoprotein cholesterol with diet. Curr Atheroscler Rep. 2004;6(6):492–8. https://pubmed.ncbi.nlm.nih.gov/15485596/
Clinton CM, O’Brien S, Law J, Renier CM, Wendt MR. Whole-foods, plant-based diet alleviates the symptoms of osteoarthritis. Arthritis. 2015;2015:708152. https://pubmed.ncbi.nlm.nih.gov/25815212/
Clinton CM, O’Brien S, Law J, Renier CM, Wendt MR. Whole-foods, plant-based diet alleviates the symptoms of osteoarthritis. Arthritis. 2015;2015:708152. https://pubmed.ncbi.nlm.nih.gov/25815212/
Clinton CM, O’Brien S, Law J, Renier CM, Wendt MR. Whole-foods, plant-based diet alleviates the symptoms of osteoarthritis. Arthritis. 2015;2015:708152. https://pubmed.ncbi.nlm.nih.gov/25815212/
National Cancer Institute. Identification of Top Food Sources of Various Dietary Components. Epidemiology and Genomics Research Program. https://epi.grants.cancer.gov/diet/foodsources/top-food-sources-report-02212020.pdf. Updated November 30, 2019. Accessed June 2, 2022.; https://epi.grants.cancer.gov/diet/foodsources/top-food-sources-report-02212020.pdf
Vane JR. The mode of action of aspirin and similar compounds. J Allergy Clin Immunol. 1976;58(6):691–712. https://pubmed.ncbi.nlm.nih.gov/791988/
Clinton CM, O’Brien S, Law J, Renier CM, Wendt MR. Whole-foods, plant-based diet alleviates the symptoms of osteoarthritis. Arthritis. 2015;2015:708152. https://pubmed.ncbi.nlm.nih.gov/25815212/
Haug A, Olesen I, Christophersen OA. Individual variation and intraclass correlation in arachidonic acid and eicosapentaenoic acid in chicken muscle. Lipids Health Dis. 2010;9:37. https://pubmed.ncbi.nlm.nih.gov/20398309/
Dean E, Gormsen Hansen R. Prescribing optimal nutrition and physical activity as “first-line” interventions for best practice management of chronic low-grade inflammation associated with osteoarthritis: evidence synthesis. Arthritis. 2012;2012:1–28. https://pubmed.ncbi.nlm.nih.gov/23346399/
Toopchizadeh V, Dolatkhah N, Aghamohammadi D, Rasouli M, Hashemian M. Dietary inflammatory index is associated with pain intensity and some components of quality of life in patients with knee osteoarthritis. BMC Res Notes. 2020;13(1):448. https://pubmed.ncbi.nlm.nih.gov/32958008/
Liu Q, Hebert JR, Shivappa N, et al. Inflammatory potential of diet and risk of incident knee osteoarthritis: a prospective cohort study. Arthritis Res Ther. 2020;22(1):209. https://pubmed.ncbi.nlm.nih.gov/32912291/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Nilsson AC, Östman EM, Knudsen KEB, Holst JJ, Björck IME. A cereal-based evening meal rich in indigestible carbohydrates increases plasma butyrate the next morning. J Nutr. 2010;140(11):1932–6. https://pubmed.ncbi.nlm.nih.gov/20810606/
Meijer K, de Vos P, Priebe MG. Butyrate and other short-chain fatty acids as modulators of immunity: what relevance for health? Curr Opin Clin Nutr Metab Care. 2010;13(6):715–21. https://pubmed.ncbi.nlm.nih.gov/20823773/
Wang B, Zhou J, Wang K. Sodium butyrate abolishes the degradation of type II collagen in human chondrocytes. Biomed Pharmacother. 2018;102:1099–104. https://pubmed.ncbi.nlm.nih.gov/29710527/
Dai Z, Lu N, Niu J, Felson DT, Zhang Y. Dietary fiber intake in relation to knee pain trajectory. Arthritis Care Res (Hoboken). 2017;69(9):1331–9. https://pubmed.ncbi.nlm.nih.gov/27899003/
Dai Z, Niu J, Zhang Y, Jacques P, Felson DT. Dietary intake of fibre and risk of knee osteoarthritis in two US prospective cohorts [published correction appears in Ann Rheum Dis. 2017;76(12):2103]. Ann Rheum Dis. 2017;76(8):1411–9. https://pubmed.ncbi.nlm.nih.gov/28536116/
Schott EM, Farnsworth CW, Grier A, et al. Targeting the gut microbiome to treat the osteoarthritis of obesity. JCI Insight. 2018;3(8):95997. https://pubmed.ncbi.nlm.nih.gov/29669931/
Berenbaum F, Wallace IJ, Lieberman DE, Felson DT. Modern-day environmental factors in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2018;14(11):674–81. https://pubmed.ncbi.nlm.nih.gov/30209413/
Berenbaum F, Wallace IJ, Lieberman DE, Felson DT. Modern-day environmental factors in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2018;14(11):674–81. https://pubmed.ncbi.nlm.nih.gov/30209413/
Guan VX, Mobasheri A, Probst YC. A systematic review of osteoarthritis prevention and management with dietary phytochemicals from foods. Maturitas. 2019;122:35–43. https://pubmed.ncbi.nlm.nih.gov/30797528/
Canter PH, Wider B, Ernst E. The antioxidant vitamins A, C, E and selenium in the treatment of arthritis: a systematic review of randomized clinical trials. Rheumatology (Oxford). 2007;46(8):1223–33. https://pubmed.ncbi.nlm.nih.gov/17522095/
Kraus VB, Huebner JL, Stabler T, et al. Ascorbic acid increases the severity of spontaneous knee osteoarthritis in a guinea pig model. Arthritis Rheum. 2004;50(6):1822–31. https://pubmed.ncbi.nlm.nih.gov/15188359/
Haqqi TM, Anthony DD, Gupta S, et al. Prevention of collagen-induced arthritis in mice by a polyphenolic fraction from green tea. Proc Natl Acad Sci U S A. 1999;96(8):4524–9. https://pubmed.ncbi.nlm.nih.gov/10200295/
Leong DJ, Choudhury M, Hanstein R, et al. Green tea polyphenol treatment is chondroprotective, anti-inflammatory and palliative in a mouse posttraumatic osteoarthritis model. Arthritis Res Ther. 2014;16(6):508. https://pubmed.ncbi.nlm.nih.gov/25516005/
Bae JY, Han DW, Wakitani S, Nawata M, Hyon SH. Biological and biomechanical evaluations of osteochondral allografts preserved in cold storage solution containing epigallocatechin gallate. Cell Transplant. 2010;19(6):681–9. https://pubmed.ncbi.nlm.nih.gov/20525433/
Hu J, Webster D, Cao J, Shao A. The safety of green tea and green tea extract consumption in adults – results of a systematic review. Regul Toxicol Pharmacol. 2018;95:412–33. https://pubmed.ncbi.nlm.nih.gov/29580974/
Hashempur MH, Sadrneshin S, Mosavat SH, Ashraf A. Green tea (Camellia sinensis) for patients with knee osteoarthritis: a randomized open-label active-controlled clinical trial. Clin Nutr. 2018;37(1):85–90. https://pubmed.ncbi.nlm.nih.gov/28038881/
Connelly AE, Tucker AJ, Tulk H, et al. High-rosmarinic acid spearmint tea in the management of knee osteoarthritis symptoms. J Med Food. 2014;17(12):1361–7. https://pubmed.ncbi.nlm.nih.gov/25058311/
Lu B, Ahmad O, Zhang FF, et al. Soft drink intake and progression of radiographic knee osteoarthritis: data from the Osteoarthritis Initiative. BMJ Open. 2013;3(7):e002993. https://pubmed.ncbi.nlm.nih.gov/23872291/
Lu B, Driban JB, Duryea J, McAlindon T, Lapane KL, Eaton CB. Milk consumption and progression of medial tibiofemoral knee osteoarthritis: data from the Osteoarthritis Initiative. Arthritis Care Res (Hoboken). 2014;66(6):802–9. https://pubmed.ncbi.nlm.nih.gov/24706620/
Denissen KFM, Boonen A, Nielen JTH, et al. Consumption of dairy products in relation to the presence of clinical knee osteoarthritis: The Maastricht Study. Eur J Nutr. 2019;58(7):2693–704. https://pubmed.ncbi.nlm.nih.gov/30242468/
Arjmandi BH, Khalil DA, Lucas EA, et al. Soy protein may alleviate osteoarthritis symptoms. Phytomedicine. 2004;11(7–8):567–75. https://pubmed.ncbi.nlm.nih.gov/15636169/
Minaie M, Rostamian, A, Abbassian A, Ghayoumi A, Mashayekhi A. An investigation of the effect of dairy products on chronic knee osteoarthritis pain in patients referred to Tehran Rheumatology Clinic of Imam Khomeini Hospital. Int J Biol Pharm Allied Sci. 2017;6(2):218–26. https://www.researchgate.net/publication/316753084_AN_INVESTIGATION_OF_THE_EFFECT_OF_DAIRY_PRODUCTS_ON_CHRONIC_KNEE_OSTEOARTHRITIS_PAIN_IN_PATIENTS_REFERRED_TO_TEHRAN_RHEUMATOLOGY_CLINIC_OF_IMAM_KHOMEINI_HOSPITAL
Basu A, Kurien BT, Tran H, et al. Strawberries decrease circulating levels of tumor necrosis factor and lipid peroxides in knee osteoarthritis in obese adults. Food Funct. 2018;9(12):6218–26. https://pubmed.ncbi.nlm.nih.gov/30382270/
Schumacher HR, Pullman-Mooar S, Gupta SR, Dinnella JE, Kim R, McHugh MP. Randomized double-blind crossover study of the efficacy of a tart cherry juice blend in treatment of osteoarthritis (OA) of the knee. Osteoarthritis Cartilage. 2013;21(8):1035–41. https://pubmed.ncbi.nlm.nih.gov/23727631/
Collins MW, Saag KG, Singh JA. Is there a role for cherries in the management of gout? Ther Adv Musculoskelet Dis. 2019;11:1759720X19847018. https://pubmed.ncbi.nlm.nih.gov/31205513/
Ghavipour M, Sotoudeh G, Tavakoli E, Mowla K, Hasanzadeh J, Mazloom Z. Pomegranate extract alleviates disease activity and some blood biomarkers of inflammation and oxidative stress in Rheumatoid Arthritis patients. Eur J Clin Nutr. 2017;71(1):92–6. https://pubmed.ncbi.nlm.nih.gov/27577177/
Rasheed Z. Intake of pomegranate prevents the onset of osteoarthritis: molecular evidences. Int J Health Sci (Qassim). 2016;10(2):V–VIII. https://pubmed.ncbi.nlm.nih.gov/27103912/
Ahmed S, Wang N, Hafeez BB, Cheruvu VK, Haqqi TM. Punica granatum L. extract inhibits IL-1ß-induced expression of matrix metalloproteinases by inhibiting the activation of MAP kinases and NF-¿B in human chondrocytes in vitro. J Nutr. 2005;135(9):2096–102. https://pubmed.ncbi.nlm.nih.gov/16140882/
Lahart I, Darcy P, Gidlow C, Calogiuri G. The effects of green exercise on physical and mental wellbeing: a systematic review. Int J Environ Res Public Health. 2019;16(8):1352. https://pubmed.ncbi.nlm.nih.gov/30991724/
Moazen S, Amani R, Homayouni Rad A, Shahbazian H, Ahmadi K, Taha Jalali M. Effects of freeze-dried strawberry supplementation on metabolic biomarkers of atherosclerosis in subjects with type 2 diabetes: a randomized double-blind controlled trial. Ann Nutr Metab. 2013;63(3):256–64. https://pubmed.ncbi.nlm.nih.gov/24334868/
Chen T, Yan F, Qian J, et al. Randomized phase II trial of lyophilized strawberries in patients with dysplastic precancerous lesions of the esophagus. Cancer Prev Res (Phila). 2012;5(1):41–50. https://pubmed.ncbi.nlm.nih.gov/22135048/
Edirisinghe I, Banaszewski K, Cappozzo J, et al. Strawberry anthocyanin and its association with postprandial inflammation and insulin. Br J Nutr. 2011;106(6):913–22. https://pubmed.ncbi.nlm.nih.gov/21736853/
Schell J, Scofield RH, Barrett JR, et al. Strawberries improve pain and inflammation in obese adults with radiographic evidence of knee osteoarthritis. Nutrients. 2017;9(9):949. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5622709/
Basu A, Kurien BT, Tran H, et al. Strawberries decrease circulating levels of tumor necrosis factor and lipid peroxides in knee osteoarthritis in obese adults. Food Funct. 2018;9(12):6218–26. https://pubmed.ncbi.nlm.nih.gov/30382270/
Rutledge GA, Fisher DR, Miller MG, Kelly ME, Bielinski DF, Shukitt-Hale B. The effects of blueberry and strawberry serum metabolites on age-related oxidative and inflammatory signaling in vitro. Food Funct. 2019;10(12):7707–13. https://pubmed.ncbi.nlm.nih.gov/31746877/
Du C, Smith A, Avalos M, et al. Blueberries improve pain, gait performance, and inflammation in individuals with symptomatic knee osteoarthritis. Nutrients. 2019;11(2):E290. https://pubmed.ncbi.nlm.nih.gov/30699971/
Christensen R, Bartels EM, Altman RD, Astrup A, Bliddal H. Does the hip powder of Rosa canina (rosehip) reduce pain in osteoarthritis patients? – a meta-analysis of randomized controlled trials. Osteoarthritis Cartilage. 2008;16(9):965–72. https://pubmed.ncbi.nlm.nih.gov/18407528/
Bannuru RR, Schmid CH, Kent DM, Vaysbrot EE, Wong JB, McAlindon TE. Comparative effectiveness of pharmacologic interventions for knee osteoarthritis: a systematic review and network meta-analysis. Ann Intern Med. 2015;162(1):46–54. https://pubmed.ncbi.nlm.nih.gov/25560713/
Christensen R, Bartels EM, Altman RD, Astrup A, Bliddal H. Does the hip powder of Rosa canina (rosehip) reduce pain in osteoarthritis patients? – a meta-analysis of randomized controlled trials. Osteoarthritis Cartilage. 2008;16(9):965–72. https://pubmed.ncbi.nlm.nih.gov/18407528/
Davidson RK, Jupp O, de Ferrars R, et al. Sulforaphane represses matrix-degrading proteases and protects cartilage from destruction in vitro and in vivo. Arthritis Rheum. 2013;65(12):3130–40. https://pubmed.ncbi.nlm.nih.gov/23983046/
Davidson R, Gardner S, Jupp O, et al. Isothiocyanates are detected in human synovial fluid following broccoli consumption and can affect the tissues of the knee joint. Sci Rep. 2017;7(1):3398. https://pubmed.ncbi.nlm.nih.gov/28611391/
Davidson RK, Jupp O, de Ferrars R, et al. Sulforaphane represses matrix-degrading proteases and protects cartilage from destruction in vitro and in vivo. Arthritis Rheum. 2013;65(12):3130–40. https://pubmed.ncbi.nlm.nih.gov/23983046/
Broccoli in Osteoarthritis (BRIO). ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03878368. Published March 18, 2019. Updated May 17, 2022. Accessed June 5, 2022.; https://clinicaltrials.gov/ct2/show/NCT03878368
Riso P, Vendrame S, Del Bo’ C, et al. Effect of 10-day broccoli consumption on inflammatory status of young healthy smokers. Int J Food Sci Nutr. 2014;65(1):106–11. https://pubmed.ncbi.nlm.nih.gov/23992556/
Araya-Quintanilla F, Gutiérrez-Espinoza H, Muñoz-Yanez MJ, Sanchez-Montoya U, Lopez-Jeldes J. Effectiveness of ginger on pain and function in knee osteoarthritis: a PRISMA systematic review and meta-analysis. Pain Physician. 2020;23(2):E151–61. https://pubmed.ncbi.nlm.nih.gov/32214292/
Haghighi M, Khalvat A, Toliat T, Jallaei S. Comparing the effects of ginger (Zingiber officinale) extract and ibuprofen on patients with osteoarthritis. Arch Iran Med. 2005; 8(4):267–71. https://www.researchgate.net/publication/235007127_Comparing_the_Effects_of_ginger_Zingiber_officinale_extract_and_ibuprofen_On_patients_with_osteoarthritis
Drozdov VN, Kim VA, Tkachenko EV, Varvanina GG. Influence of a specific ginger combination on gastropathy conditions in patients with osteoarthritis of the knee or hip. J Altern Complement Med. 2012;18(6):583–8. https://pubmed.ncbi.nlm.nih.gov/22784345/
Caunedo-Alvarez A, Gómez-Rodríguez BJ, Romero-Vázquez J, et al. Macroscopic small bowel mucosal injury caused by chronic nonsteroidal anti-inflammatory drugs (NSAID) use as assessed by capsule endoscopy. Rev Esp Enferm Dig. 2010;102(2):80–5. https://pubmed.ncbi.nlm.nih.gov/20361843/
Therkleson T. Ginger compress therapy for adults with osteoarthritis. J Adv Nurs. 2010;66(10):2225–33. https://pubmed.ncbi.nlm.nih.gov/20626491/
Ding M, Leach MJ, Bradley H. A systematic review of the evidence for topical use of ginger. Explore (NY). 2013;9(6):361–4. https://pubmed.ncbi.nlm.nih.gov/24199775/
Percival SS, Vanden Heuvel JP, Nieves CJ, Montero C, Migliaccio AJ, Meadors J. Bioavailability of herbs and spices in humans as determined by ex vivo inflammatory suppression and DNA strand breaks. J Am Coll Nutr. 2012;31(4):288–94. https://pubmed.ncbi.nlm.nih.gov/23378457/
Wang Z, Singh A, Jones G, et al. Efficacy and safety of turmeric extracts for the treatment of knee osteoarthritis: a systematic review and meta-analysis of randomised controlled trials. Curr Rheumatol Rep. 2021;23(2):11. https://pubmed.ncbi.nlm.nih.gov/33511486/
Jamali N, Adib-Hajbaghery M, Soleimani A. The effect of curcumin ointment on knee pain in older adults with osteoarthritis: a randomized placebo trial. BMC Complement Med Ther. 2020;20(1):305. https://pubmed.ncbi.nlm.nih.gov/33032585/
Miyawaki T, Aono H, Toyoda-Ono Y, Maeda H, Kiso Y, Moriyama K. Antihypertensive effects of sesamin in humans. J Nutr Sci Vitaminol (Tokyo). 2009;55(1):87–91. https://pubmed.ncbi.nlm.nih.gov/19352068/
Wu WH, Kang YP, Wang NH, Jou HJ, Wang TA. Sesame ingestion affects sex hormones, antioxidant status, and blood lipids in postmenopausal women. J Nutr. 2006;136(5):1270–5. https://pubmed.ncbi.nlm.nih.gov/16614415/
Askari A, Ravansalar SA, Naghizadeh MM, et al. The efficacy of topical sesame oil in patients with knee osteoarthritis: a randomized double-blinded active-controlled non-inferiority clinical trial. Complement Ther Med. 2019;47:102183. https://pubmed.ncbi.nlm.nih.gov/31780006/
Savas BB, Alparslan GB, Korkmaz C. Effect of flaxseed poultice compress application on pain and hand functions of patients with hand osteoarthritis. Clin Rheumatol. 2019;38(7):1961–9. https://pubmed.ncbi.nlm.nih.gov/30806856/
Hashempur MH, Homayouni K, Ashraf A, Salehi A, Taghizadeh M, Heydari M. Effect of Linum usitatissimum L. (linseed) oil on mild and moderate carpal tunnel syndrome: a randomized, double-blind, placebo-controlled clinical trial. Daru. 2014;22:43. https://pubmed.ncbi.nlm.nih.gov/24887185/
Mosavat SH, Masoudi N, Hajimehdipoor H, et al. Efficacy of topical Linum usitatissimum L. (flaxseed) oil in knee osteoarthritis: a double-blind, randomized, placebo-controlled clinical trial. Complement Ther Clin Pract. 2018;31:302–7. https://pubmed.ncbi.nlm.nih.gov/29705472/
Nasser M, Tibi A, Savage-Smith E. Ibn Sina’s Canon of Medicine: 11th century rules for assessing the effects of drugs. J R Soc Med. 2009;102(2):78–80. https://pubmed.ncbi.nlm.nih.gov/19208873/
Hashmi MA, Khan A, Hanif M, Farooq U, Perveen S. Traditional uses, phytochemistry, and pharmacology of Olea europaea (olive). Evid Based Complement Alternat Med. 2015;2015:541591. https://pubmed.ncbi.nlm.nih.gov/25802541/
Fernandes J, Fialho M, Santos R, et al. Is olive oil good for you? A systematic review and meta-analysis on anti-inflammatory benefits from regular dietary intake. Nutrition. 2020;69:110559. https://pubmed.ncbi.nlm.nih.gov/31539817/
Khaw KT, Sharp SJ, Finikarides L, et al. Randomised trial of coconut oil, olive oil or butter on blood lipids and other cardiovascular risk factors in healthy men and women. BMJ Open. 2018;8(3):e020167. https://pubmed.ncbi.nlm.nih.gov/29511019/
Bohlooli S, Jastan M, Nakhostin-Roohi B, Mohammadi S, Baghaei Z. A pilot double-blinded, randomized, clinical trial of topical virgin olive oil versus piroxicam gel in osteoarthritis of the knee. J Clin Rheumatol. 2012;18(2):99–101. https://pubmed.ncbi.nlm.nih.gov/22334264/
Hekmatpou D, Mortaji S, Rezaei M, Shaikhi M. The effectiveness of olive oil in controlling morning inflammatory pain of phalanges and knees among women with rheumatoid arthritis: a randomized clinical trial. Rehabil Nurs. 2020;45(2):106–13. https://pubmed.ncbi.nlm.nih.gov/30192341/
Balagué F, Mannion AF, Pellisé F, Cedraschi C. Non-specific low back pain. Lancet. 2012;379(9814):482–91. https://pubmed.ncbi.nlm.nih.gov/21982256/
Zhang TT, Liu Z, Liu YL, Zhao JJ, Liu DW, Tian QB. Obesity as a risk factor for low back pain: a meta-analysis. Clin Spine Surg. 2018;31(1):22–7. https://pubmed.ncbi.nlm.nih.gov/27875413/
Shiri R, Lallukka T, Karppinen J, Viikari-Juntura E. Obesity as a risk factor for sciatica: a meta-analysis. Am J Epidemiol. 2014;179(8):929–37. https://pubmed.ncbi.nlm.nih.gov/24569641/
Xu X, Li X, Wu W. Association between overweight or obesity and lumbar disk diseases. J Spinal Disord Tech. 2015;28(10):370–6. https://pubmed.ncbi.nlm.nih.gov/25500506/
Shiri R, Lallukka T, Karppinen J, Viikari-Juntura E. Obesity as a risk factor for sciatica: a meta-analysis. Am J Epidemiol. 2014;179(8):929–37. https://pubmed.ncbi.nlm.nih.gov/24569641/
Xu X, Li X, Wu W. Association between overweight or obesity and lumbar disk diseases. J Spinal Disord Tech. 2015;28(10):370–6. https://pubmed.ncbi.nlm.nih.gov/25500506/
Kauppila LI. Atherosclerosis and disc degeneration/low-back pain – a systematic review. Eur J Vasc Endovasc Surg. 2009;37(6):661–70. https://pubmed.ncbi.nlm.nih.gov/19328027/
Kauppila LI, Mikkonen R, Mankinen P, Pelto-Vasenius K, Mäenpää I. MR aortography and serum cholesterol levels in patients with long-term nonspecific lower back pain. Spine (Phila Pa 1976). 2004;29(19):2147–52. https://pubmed.ncbi.nlm.nih.gov/15454707/
Ventegodt S, Merrick J. Dean Ornish should receive the Nobel prize in medicine. Int J Adolesc Med Health. 2012;24(2):97–8. https://pubmed.ncbi.nlm.nih.gov/22909930/
Blankenhorn DH, Hodis HN. George Lyman Duff memorial lecture. Arterial imaging and atherosclerosis reversal. Arterioscler Thromb. 1994;14(2):177–92. https://pubmed.ncbi.nlm.nih.gov/8305407/
Ostfeld RJ, Allen KE, Aspry K, et al. Vasculogenic erectile dysfunction: the impact of diet and lifestyle. Am J Med. 2021;134(3):310–6. https://pubmed.ncbi.nlm.nih.gov/33227246/
Renegar KB, Floyd RA, Krueger JM. Effects of short-term sleep deprivation on murine immunity to influenza virus in young adult and senescent mice. Sleep. 1998;21(3):241–8. https://pubmed.ncbi.nlm.nih.gov/9595602/
Vacaflor BE, Beauchet O, Jarvis GE, Schavietto A, Rej S. Mental health and cognition in older cannabis users: a review. Can Geriatr J. 2020;23(3):242–9. https://pubmed.ncbi.nlm.nih.gov/32904776/
Campeny E, López-Pelayo H, Nutt D, et al. The blind men and the elephant: systematic review of systematic reviews of cannabis use related health harms. Eur Neuropsychopharmacol. 2020;33:1–35. https://pubmed.ncbi.nlm.nih.gov/32165103/
Wang A, Lo A, Ubhi K, Cameron T. Small and transient effect of cannabis oil for osteoarthritis-related joint pain: a case report. Can J Hosp Pharm. 2021;74(2):156–8. https://pubmed.ncbi.nlm.nih.gov/33896956/
Vela J, Dreyer L, Petersen KK, Arendt-Nielsen L, Duch KS, Kristensen S. Cannabidiol treatment in hand osteoarthritis and psoriatic arthritis: a randomized, double-blind, placebo-controlled trial. Pain. 2022;163(6):1206–14. https://pubmed.ncbi.nlm.nih.gov/34510141/
Senftleber NK, Nielsen SM, Andersen JR, et al. Marine oil supplements for arthritis pain: a systematic review and meta-analysis of randomized trials. Nutrients. 2017;9(1):E42. https://pubmed.ncbi.nlm.nih.gov/28067815/
Gregory PJ, Sperry M, Wilson AF. Dietary supplements for osteoarthritis. Am Fam Physician. 2008;77(2):177–84. https://pubmed.ncbi.nlm.nih.gov/18246887/
McCarty MF, O’Keefe JH, DiNicolantonio JJ. Glucosamine for the treatment of osteoarthritis: the time has come for higher-dose trials. J Diet Suppl. 2019;16(2):179–92. https://pubmed.ncbi.nlm.nih.gov/29667462/
Kolasinski SL, Neogi T, Hochberg MC, et al. 2019 American College of Rheumatology/Arthritis Foundation guideline for the management of osteoarthritis of the hand, hip, and knee. Arthritis Care Res (Hoboken). 2020;72(2):149–62. https://pubmed.ncbi.nlm.nih.gov/31908149/
Kolasinski SL, Neogi T, Hochberg MC, et al. 2019 American College of Rheumatology/Arthritis Foundation guideline for the management of osteoarthritis of the hand, hip, and knee. Arthritis Rheumatol. 2020;72(2):220–33. https://pubmed.ncbi.nlm.nih.gov/31908149/
Reichenbach S, Sterchi R, Scherer M, et al. Meta-analysis: chondroitin for osteoarthritis of the knee or hip. Ann Intern Med. 2007;146(8):580–90. https://pubmed.ncbi.nlm.nih.gov/17438317/
Roman-Blas JA, Castañeda S, Sánchez-Pernaute O, et al. Combined treatment with chondroitin sulfate and glucosamine sulfate shows no superiority over placebo for reduction of joint pain and functional impairment in patients with knee osteoarthritis: a six-month multicenter, randomized, double-blind, placebo-controlled clinical trial. Arthritis Rheumatol. 2017;69(1):77–85. https://pubmed.ncbi.nlm.nih.gov/27477804/
Moskowitz RW. Role of collagen hydrolysate in bone and joint disease. Semin Arthritis Rheum. 2000;30(2):87–99. https://pubmed.ncbi.nlm.nih.gov/11071580/
Moskowitz RW. Role of collagen hydrolysate in bone and joint disease. Semin Arthritis Rheum. 2000;30(2):87–99. https://pubmed.ncbi.nlm.nih.gov/11071580/
Alahakoon AU, Oey I, Silcock P, Bremer P. Understanding the effect of pulsed electric fields on thermostability of connective tissue isolated from beef pectoralis muscle using a model system. Food Res Int. 2017;100(Pt 2):261–7. https://pubmed.ncbi.nlm.nih.gov/28888449/
García-Coronado JM, Martínez-Olvera L, Elizondo-Omaña RE, et al. Effect of collagen supplementation on osteoarthritis symptoms: a meta-analysis of randomized placebo-controlled trials. Int Orthop. 2019;43(3):531–8. https://pubmed.ncbi.nlm.nih.gov/30368550/
von Hippel PT. Do collagen supplements reduce symptoms of osteoarthritis? Meta-analytic results do not support strong conclusions. Int Orthop. 2021;45(12):3283–4. https://pubmed.ncbi.nlm.nih.gov/34636929/
Jabbari M, Barati M, Khodaei M, et al. Is collagen supplementation friend or foe in rheumatoid arthritis and osteoarthritis? A comprehensive systematic review. Int J Rheum Dis. 2022;25(9):973–81. https://pubmed.ncbi.nlm.nih.gov/35791039/
Sambeth A, Riedel WJ, Tillie DE, Blokland A, Postma A, Schmitt JAJ. Memory impairments in humans after acute tryptophan depletion using a novel gelatin-based protein drink. J Psychopharmacol. 2009;23(1):56–64. https://pubmed.ncbi.nlm.nih.gov/18515454/
Jabbari M, Barati M, Khodaei M, et al. Is collagen supplementation friend or foe in rheumatoid arthritis and osteoarthritis? A comprehensive systematic review. Int J Rheum Dis. 2022;25(9):973–81. https://pubmed.ncbi.nlm.nih.gov/35791039/
Bongers CCWG, Ten Haaf DSM, Catoire M, et al. Effectiveness of collagen supplementation on pain scores in healthy individuals with self-reported knee pain: a randomized controlled trial. Appl Physiol Nutr Metab. 2020;45(7):793–800. https://pubmed.ncbi.nlm.nih.gov/31990581/
Delgado-Saborit JM, Guercio V, Gowers AM, Shaddick G, Fox NC, Love S. A critical review of the epidemiological evidence of effects of air pollution on dementia, cognitive function and cognitive decline in adult population. Sci Total Environ. 2021;757:143734. https://pubmed.ncbi.nlm.nih.gov/33340865/
Gu YH, Bai JB, Chen XL, Wu WW, Liu XX, Tan XD. Healthy aging: a bibliometric analysis of the literature. Exp Gerontol. 2019;116:93–105. https://pubmed.ncbi.nlm.nih.gov/30590123/
Ritchie K, Kildea D. Is senile dementia “age-related” or “ageing-related”?—evidence from meta-analysis of dementia prevalence in the oldest old. Lancet. 1995;346(8980):931–4. https://pubmed.ncbi.nlm.nih.gov/7564727/
Wickelgren I. Is hippocampal cell death a myth? Science. 1996;271(5253):1229–30. https://pubmed.ncbi.nlm.nih.gov/8638100/
Sherzai D, Sherzai A. Preventing Alzheimer’s: our most urgent health care priority. Am J Lifestyle Med. 2019;13(5):451–61. https://pubmed.ncbi.nlm.nih.gov/31523210/
2022 Alzheimer’s disease facts and figures. Special report. More than normal aging: understanding mild cognitive impairment. Alzheimer’s Association. https://www.alz.org/media/Documents/alzheimers-facts-and-figures.pdf. 2022. Accessed January 8, 2023.; https://www.alz.org/media/Documents/alzheimers-facts-and-figures.pdf
Sherzai D, Sherzai A. Preventing Alzheimer’s: our most urgent health care priority. Am J Lifestyle Med. 2019;13(5):451–61. https://pubmed.ncbi.nlm.nih.gov/31523210/
de la Torre JC. A turning point for Alzheimer’s disease? Biofactors. 2012;38(2):78–83. https://pubmed.ncbi.nlm.nih.gov/22422426/
Lopez OL, Kuller LH. Epidemiology of aging and associated cognitive disorders: prevalence and incidence of Alzheimer’s disease and other dementias. Handb Clin Neurol. 2019;167:139–48. https://pubmed.ncbi.nlm.nih.gov/31753130/
Sengoku R. Aging and Alzheimer’s disease pathology. Neuropathol. 2020;40(1):22–9. https://pubmed.ncbi.nlm.nih.gov/31863504/
Kawas CH, Kim RC, Sonnen JA, Bullain SS, Trieu T, Corrada MM. Multiple pathologies are common and related to dementia in the oldest-old. Neurology. 2015;85(6):535–42. https://pubmed.ncbi.nlm.nih.gov/26180144/
Viña J, Sanz-Ros J. Alzheimer’s disease: only prevention makes sense. Eur J Clin Invest. 2018;48(10):e13005. https://pubmed.ncbi.nlm.nih.gov/30028503/
Román GC. Facts, myths, and controversies in vascular dementia. J Neurol Sci. 2004;226(1–2):49–52. https://pubmed.ncbi.nlm.nih.gov/15537519/
Grau-olivares M, Arboix A. Mild cognitive impairment in stroke patients with ischemic cerebral small-vessel disease: a forerunner of vascular dementia? Expert Rev Neurother. 2009;9(8):1201–17. https://pubmed.ncbi.nlm.nih.gov/19673608/
Schneider JA, Arvanitakis Z, Bang W, Bennett DA. Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology. 2007;69(24):2197–204. https://pubmed.ncbi.nlm.nih.gov/17568013/
Murphy SL, Kochanek KD, Xu J, Arias E. Mortality in the United States, 2020. NCHS Data Brief. 2021;(427):1–8. https://pubmed.ncbi.nlm.nih.gov/34978528/
Heron M. Deaths: leading causes for 2019. Natl Vital Stat Rep. 2021;70(9):1–114. https://pubmed.ncbi.nlm.nih.gov/34520342/
Haaksma ML, Eriksdotter M, Rizzuto D, et al. Survival time tool to guide care planning in people with dementia. Neurology. 2020;94(5):e538–48. https://pubmed.ncbi.nlm.nih.gov/31843808/
Lopez OL, Kuller LH. Epidemiology of aging and associated cognitive disorders: prevalence and incidence of Alzheimer’s disease and other dementias. Handb Clin Neurol. 2019;167:139–48. https://pubmed.ncbi.nlm.nih.gov/31753130/
Cahill S, Pierce M, Werner P, Darley A, Bobersky A. A systematic review of the public’s knowledge and understanding of Alzheimer’s disease and dementia. Alzheimer Dis Assoc Disord. 2015;29(3):255–75. https://pubmed.ncbi.nlm.nih.gov/26207322/
Goodwin JS. Geriatric ideology: the myth of the myth of senility. J Am Geriatr Soc. 1991;39(6):627–31. https://pubmed.ncbi.nlm.nih.gov/2037757/
Takao M, Hirose N, Arai Y, Mihara B, Mimura M. Neuropathology of supercentenarians – four autopsy case studies. Acta Neuropathol Commun. 2016;4(1):97. https://pubmed.ncbi.nlm.nih.gov/27590044/
den Dunnen WFA, Brouwer WH, Bijlard E, et al. No disease in the brain of a 115-year-old woman. Neurobiol Aging. 2008;29(8):1127–32. https://pubmed.ncbi.nlm.nih.gov/18534718/
Williams RW, Herrup K. The control of neuron number. Annu Rev Neurosci. 1988;11:423–53. https://pubmed.ncbi.nlm.nih.gov/3284447/
von Bartheld CS. Myths and truths about the cellular composition of the human brain: a review of influential concepts. J Chem Neuroanat. 2018;93:2–15. https://pubmed.ncbi.nlm.nih.gov/28873338/
Sherzai D, Sherzai A. Preventing Alzheimer’s: our most urgent health care priority. Am J Lifestyle Med. 2019;13(5):451–61. https://pubmed.ncbi.nlm.nih.gov/31523210/
Cahill S, Pierce M, Werner P, Darley A, Bobersky A. A systematic review of the public’s knowledge and understanding of Alzheimer’s disease and dementia. Alzheimer Dis Assoc Disord. 2015;29(3):255–75. https://pubmed.ncbi.nlm.nih.gov/26207322/
Birks JS, Harvey RJ. Donepezil for dementia due to Alzheimer’s disease. Cochrane Database Syst Rev. 2018;2018(6):CD001190. https://pubmed.ncbi.nlm.nih.gov/29923184/
McShane R, Westby MJ, Roberts E, et al. Memantine for dementia. Cochrane Database Syst Rev. 2019;3:CD003154. https://pubmed.ncbi.nlm.nih.gov/30891742/
Schmitt HP. On the paradox of ion channel blockade and its benefits in the treatment of Alzheimer disease. Med Hypotheses. 2005;65(2):259–65. https://pubmed.ncbi.nlm.nih.gov/15922097/
Blanco-Silvente L, Castells X, Garre-Olmo J, et al. Study of the strength of the evidence and the redundancy of the research on pharmacological treatment for Alzheimer’s disease: a cumulative meta-analysis and trial sequential analysis. Eur J Clin Pharmacol. 2019;75(12):1659–67. https://pubmed.ncbi.nlm.nih.gov/31435707/
Fink HA, Linskens EJ, MacDonald R, et al. Benefits and harms of prescription drugs and supplements for treatment of clinical Alzheimer-type dementia. Ann Intern Med. 2020;172(10):656–68. https://pubmed.ncbi.nlm.nih.gov/32340037/
Blanco-Silvente L, Castells X, Garre-Olmo J, et al. Study of the strength of the evidence and the redundancy of the research on pharmacological treatment for Alzheimer’s disease: a cumulative meta-analysis and trial sequential analysis. Eur J Clin Pharmacol. 2019;75(12):1659–67. https://pubmed.ncbi.nlm.nih.gov/31435707/
Rabinovici GD. Controversy and progress in Alzheimer’s disease – FDA approval of aducanumab. N Engl J Med. 2021;385(9):771–4. https://pubmed.ncbi.nlm.nih.gov/34320284/
Robinson JC. Why is aducanumab priced at $56,000 per patient? Lessons for drug-pricing reform. N Engl J Med. 2021;385(22):2017–9. https://pubmed.ncbi.nlm.nih.gov/34797614/
Moghavem N, Henderson VW, Greicius MD. Medicare should not cover aducanumab as a treatment for Alzheimer’s disease. Ann Neurol. 2021;90(3):331–3. https://pubmed.ncbi.nlm.nih.gov/34278596/
Rubin R. Recently approved Alzheimer drug raises questions that might never be answered. JAMA. 2021;326(6):469–72. https://pubmed.ncbi.nlm.nih.gov/34287610/
Crosson FJ, Covinsky K, Redberg RF. Medicare and the shocking US Food and Drug Administration approval of aducanumab: crisis or opportunity? JAMA Intern Med. 2021;181(10):1278–80. https://pubmed.ncbi.nlm.nih.gov/34254992/
Crosson FJ, Covinsky K, Redberg RF. Medicare and the shocking US Food and Drug Administration approval of aducanumab: crisis or opportunity? JAMA Intern Med. 2021;181(10):1278–80. https://pubmed.ncbi.nlm.nih.gov/34254992/
Rubin R. Recently approved Alzheimer drug raises questions that might never be answered. JAMA. 2021;326(6):469–72. https://pubmed.ncbi.nlm.nih.gov/34287610/
Lundebjerg NE. My head just exploded, now what? Aducanumab. J Am Geriatr Soc. 2021;69(9):2689–91. https://pubmed.ncbi.nlm.nih.gov/34227094/
Reardon S. FDA approves Alzheimer’s drug lecanemab amid safety concerns. Nature. 2023;613(7943):227–8. https://pubmed.ncbi.nlm.nih.gov/36627422/
Walsh S, Merrick R, Richard E, Nurock S, Brayne C. Lecanemab for Alzheimer’s disease. BMJ. 2022;379. https://pubmed.ncbi.nlm.nih.gov/36535691/
Reitz C. Alzheimer’s disease and the amyloid cascade hypothesis: a critical review. Int J Alzheimers Dis. 2012;2012:369808. https://pubmed.ncbi.nlm.nih.gov/22506132/
Price JL. What does it take to stay healthy past 100? Neurobiol Aging. 2008;29(8):1140–2. https://pubmed.ncbi.nlm.nih.gov/18524418/
Barber RC. The genetics of Alzheimer’s disease. Scientifica (Cairo). 2012;2012:246210. https://pubmed.ncbi.nlm.nih.gov/24278680/
Castello MA, Soriano S. On the origin of Alzheimer’s disease. Trials and tribulations of the amyloid hypothesis. Ageing Res Rev. 2014;13:10–2. https://pubmed.ncbi.nlm.nih.gov/24252390/
Ayton S, Bush AI. ß-amyloid: the known unknowns. Ageing Res Rev. 2021;65:101212. https://pubmed.ncbi.nlm.nih.gov/33188924/
SantaCruz KS, Sonnen JA, Pezhouh MK, Desrosiers MF, Nelson PT, Tyas SL. Alzheimer disease pathology in subjects without dementia in 2 studies of aging: the Nun Study and the Adult Changes in Thought Study. J Neuropathol Exp Neurol. 2011;70(10):832–40. https://pubmed.ncbi.nlm.nih.gov/21937909/
Ayton S, Bush AI. ß-amyloid: the known unknowns. Ageing Res Rev. 2021;65:101212. https://pubmed.ncbi.nlm.nih.gov/33188924/
Alzheimer A, Förstl H, Levy R. On certain peculiar diseases of old age. Hist Psychiatry. 1991;2(5):71–3. https://pubmed.ncbi.nlm.nih.gov/11622845/
Piller C. Blots on a field? Science. 2022;377(6604):358–63. https://pubmed.ncbi.nlm.nih.gov/35862524/
Ayton S, Bush AI. ß-amyloid: the known unknowns. Ageing Res Rev. 2021;65:101212. https://pubmed.ncbi.nlm.nih.gov/33188924/
Reitz C. Alzheimer’s disease and the amyloid cascade hypothesis: a critical review. Int J Alzheimers Dis. 2012;2012:369808. https://pubmed.ncbi.nlm.nih.gov/22506132/
Amtul Z. Why therapies for Alzheimer’s disease do not work: do we have consensus over the path to follow? Ageing Res Rev. 2016;25:70–84. https://pubmed.ncbi.nlm.nih.gov/26375861/
Ayton S, Bush AI. ß-amyloid: the known unknowns. Ageing Res Rev. 2021;65:101212. https://pubmed.ncbi.nlm.nih.gov/33188924/
Joseph J, Shukitt-Hale B, Denisova NA, Martin A, Perry G, Smith MA. Copernicus revisited: amyloid beta in Alzheimer’s disease. Neurobiol Aging. 2001;22(1):131–46. https://pubmed.ncbi.nlm.nih.gov/11164287/
Ayton S, Bush AI. ß-amyloid: the known unknowns. Ageing Res Rev. 2021;65:101212. https://pubmed.ncbi.nlm.nih.gov/33188924/
Braak H, Braak E. Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol Aging. 1997;18(4):351–7. https://pubmed.ncbi.nlm.nih.gov/9330961/
Grodstein F. How early can cognitive decline be detected? BMJ. 2011;344:d7652. https://pubmed.ncbi.nlm.nih.gov/22223829/
Torres-Acosta N, O’Keefe JH, O’Keefe EL, Isaacson R, Small G. Therapeutic potential of TNF-a inhibition for Alzheimer’s disease prevention. J Alzheimers Dis. 2020;78(2):619–26. https://pubmed.ncbi.nlm.nih.gov/33016914/
Viña J, Sanz-Ros J. Alzheimer’s disease: only prevention makes sense. Eur J Clin Invest. 2018;48(10):e13005. https://pubmed.ncbi.nlm.nih.gov/30028503/
de la Torre JC. Alzheimer’s disease is incurable but preventable. J Alzheimers Dis. 2010;20(3):861–70. https://pubmed.ncbi.nlm.nih.gov/20182017/
Barnes DE, Yaffe K. The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol. 2011;10(9):819–28. https://pubmed.ncbi.nlm.nih.gov/21775213/
Singh-Manoux A, Kivimaki M, Glymour MM, et al. Timing of onset of cognitive decline: results from Whitehall II prospective cohort study. BMJ. 2012;344:d7622. https://pubmed.ncbi.nlm.nih.gov/22223828/
Roher AE, Tyas SL, Maarouf CL, et al. Intracranial atherosclerosis as a contributing factor to Alzheimer’s disease dementia. Alzheimers Dement. 2011;7(4):436–44. https://pubmed.ncbi.nlm.nih.gov/21388893/
Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An English translation of Alzheimer’s 1907 paper, “Uber eine eigenartige Erkankung der Hirnrinde.” Clin Anat. 1995;8(6):429–31. https://pubmed.ncbi.nlm.nih.gov/8713166/
Sharma M. Preventing Alzheimer’s disease: some light in the darkness. J Am Coll Cardiol. 2019;74(15):1924–5. https://pubmed.ncbi.nlm.nih.gov/31601372/
Grande G, Qiu C, Fratiglioni L. Prevention of dementia in an ageing world: evidence and biological rationale. Ageing Res Rev. 2020;64:101045. https://pubmed.ncbi.nlm.nih.gov/32171784/
Roher AE, Tyas SL, Maarouf CL, et al. Intracranial atherosclerosis as a contributing factor to Alzheimer’s disease dementia. Alzheimers Dement. 2011;7(4):436–44. https://pubmed.ncbi.nlm.nih.gov/21388893/
Yarchoan M, Xie SX, Kling MA, et al. Cerebrovascular atherosclerosis correlates with Alzheimer pathology in neurodegenerative dementias. Brain. 2012;135(Pt 12):3749–56. https://pubmed.ncbi.nlm.nih.gov/23204143/
Honig LS, Kukull W, Mayeux R. Atherosclerosis and AD: analysis of data from the US National Alzheimer’s Coordinating Center. Neurology. 2005;64(3):494–500. https://pubmed.ncbi.nlm.nih.gov/15699381/
Roher AE, Tyas SL, Maarouf CL, et al. Intracranial atherosclerosis as a contributing factor to Alzheimer’s disease dementia. Alzheimers Dement. 2011;7(4):436–44. https://pubmed.ncbi.nlm.nih.gov/21388893/
de la Torre JC. Vascular basis of Alzheimer’s pathogenesis. Ann N Y Acad Sci. 2002;977:196–215. https://pubmed.ncbi.nlm.nih.gov/12480752/
Cahill S, Pierce M, Werner P, Darley A, Bobersky A. A systematic review of the public’s knowledge and understanding of Alzheimer’s disease and dementia. Alzheimer Dis Assoc Disord. 2015;29(3):255–75. https://pubmed.ncbi.nlm.nih.gov/26207322/
Corsinovi L, Biasi F, Poli G, Leonarduzzi G, Isaia G. Dietary lipids and their oxidized products in Alzheimer’s disease. Mol Nutr Food Res. 2011;55 Suppl 2:S161–72. https://pubmed.ncbi.nlm.nih.gov/21954186/
Mizuno T, Nakata M, Naiki H, et al. Cholesterol-dependent generation of a seeding amyloid beta-protein in cell culture. J Biol Chem. 1999;274(21):15110–4. https://pubmed.ncbi.nlm.nih.gov/10329717/
Harris JR, Milton NGN. Cholesterol in Alzheimer’s disease and other amyloidogenic disorders. Subcell Biochem. 2010;51:47–75. https://pubmed.ncbi.nlm.nih.gov/20213540/
US Food and Drug Administration. Important safety label changes to cholesterol-lowering statin drugs. http://www.fda.gov/Drugs/DrugSafety/ucm293101.htm. Published July 7, 2012. Accessed June 30, 2022.; https://www.fda.gov/Drugs/DrugSafety/ucm293101.htm
Rojas-Fernandez CH, Cameron JC. Is statin-associated cognitive impairment clinically relevant? A narrative review and clinical recommendations. Ann Pharmacother. 2012;46(4):549–57. https://pubmed.ncbi.nlm.nih.gov/22474137/
Sabbagh MN, Perez A, Holland TM, et al. Primary prevention recommendations to reduce the risk of cognitive decline. Alzheimers Dement. Published online January 13, 2022.; https://pubmed.ncbi.nlm.nih.gov/35026040/
Barnard ND, Bush AI, Ceccarelli A, et al. Dietary and lifestyle guidelines for the prevention of Alzheimer’s disease. Neurobiol Aging. 2014;35 Suppl 2:S74–8. https://pubmed.ncbi.nlm.nih.gov/24913896/
Wood WG, Li L, Müller WE, Eckert GP. Cholesterol as a causative factor in Alzheimer disease: a debatable hypothesis. J Neurochem. 2014;129(4):559–72. https://pubmed.ncbi.nlm.nih.gov/24329875/
Testa G, Staurenghi E, Zerbinati C, et al. Changes in brain oxysterols at different stages of Alzheimer’s disease: their involvement in neuroinflammation. Redox Biol. 2016;10:24–33. https://pubmed.ncbi.nlm.nih.gov/27687218/
Marwarha G, Ghribi O. Does the oxysterol 27-hydroxycholesterol underlie Alzheimer’s disease – Parkinson’s disease overlap? Exp Gerontol. 2015;68:13–8. https://pubmed.ncbi.nlm.nih.gov/25261765/
Wang HL, Wang YY, Liu XG, et al. Cholesterol, 24-hydroxycholesterol, and 27-hydroxycholesterol as surrogate biomarkers in cerebrospinal fluid in mild cognitive impairment and Alzheimer’s disease: a meta-analysis. J Alzheimers Dis. 2016;51(1):45–55. https://pubmed.ncbi.nlm.nih.gov/26836015/
Gamba P, Testa G, Gargiulo S, Staurenghi E, Poli G, Leonarduzzi G. Oxidized cholesterol as the driving force behind the development of Alzheimer’s disease. Front Aging Neurosci. 2015;7:119. https://pubmed.ncbi.nlm.nih.gov/26150787/
Deschaintre Y, Richard F, Leys D, Pasquier F. Treatment of vascular risk factors is associated with slower decline in Alzheimer disease. Neurology. 2009;73(9):674–80. https://pubmed.ncbi.nlm.nih.gov/19720973/
Wright JT Jr, Williamson JD, Whelton PK, et al. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med. 2015;373(22):2103–16. https://pubmed.ncbi.nlm.nih.gov/26551272/
Williamson JD, Pajewski NM, Auchus AP, et al. Effect of intensive vs standard blood pressure control on probable dementia. JAMA. 2019;321(6):553–61. https://pubmed.ncbi.nlm.nih.gov/30688979/
Wright JT Jr, Williamson JD, Whelton PK, et al. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med. 2015;373(22):2103–16. https://pubmed.ncbi.nlm.nih.gov/26551272/
Roher AE, Tyas SL, Maarouf CL, et al. Intracranial atherosclerosis as a contributing factor to Alzheimer’s disease dementia. Alzheimers Dement. 2011;7(4):436–44. https://pubmed.ncbi.nlm.nih.gov/21388893/
Pase MP, Herbert A, Grima NA, Pipingas A, O’Rourke MF. Arterial stiffness as a cause of cognitive decline and dementia: a systematic review and meta-analysis. Intern Med J. 2012;42(7):808–15. https://pubmed.ncbi.nlm.nih.gov/22151013/
Henskens LHG, van Oostenbrugge RJ, Kroon AA, de Leeuw PW, Lodder J. Brain microbleeds are associated with ambulatory blood pressure levels in a hypertensive population. Hypertension. 2008;51(1):62–8. https://pubmed.ncbi.nlm.nih.gov/18071062/
Kovacic JC, Fuster V. Atherosclerotic risk factors, vascular cognitive impairment, and Alzheimer disease. Mt Sinai J Med. 2012;79(6):664–73. https://pubmed.ncbi.nlm.nih.gov/23239205/
Longstreth WT, Bernick C, Manolio TA, Bryan N, Jungreis CA, Price TR. Lacunar infarcts defined by magnetic resonance imaging of 3660 elderly people: the Cardiovascular Health Study. Arch Neurol. 1998;55(9):1217–25. https://pubmed.ncbi.nlm.nih.gov/9740116/
Vermeer SE, Longstreth WT, Koudstaal PJ. Silent brain infarcts: a systematic review. Lancet Neurol. 2007;6(7):611–9. https://pubmed.ncbi.nlm.nih.gov/17582361/
Beauchet O, Celle S, Roche F, et al. Blood pressure levels and brain volume reduction: a systematic review and meta-analysis [published correction appears in J Hypertens. 2013;31(10):2106]. J Hypertens. 2013;31(8):1502–16. https://pubmed.ncbi.nlm.nih.gov/23811995/
Peila R, White LR, Petrovich H, et al. Joint effect of the APOE gene and midlife systolic blood pressure on late-life cognitive impairment: the Honolulu-Asia Aging Study. Stroke. 2001;32(12):2882–9. https://pubmed.ncbi.nlm.nih.gov/11739991/
Singer J, Trollor JN, Baune BT, Sachdev PS, Smith E. Arterial stiffness, the brain and cognition: a systematic review. Ageing Res Rev. 2014;15:16–27. https://pubmed.ncbi.nlm.nih.gov/24548924/
Salvi P, Giannattasio C, Parati G. High sodium intake and arterial stiffness. J Hypertens. 2018;36(4):754–8. https://pubmed.ncbi.nlm.nih.gov/29489612/
D’Elia L, Galletti F, La Fata E, Sabino P, Strazzullo P. Effect of dietary sodium restriction on arterial stiffness: systematic review and meta-analysis of the randomized controlled trials. J Hypertens. 2018;36(4):734–43. https://pubmed.ncbi.nlm.nih.gov/29084085/
Filippini T, Malavolti M, Whelton PK, Naska A, Orsini N, Vinceti M. Blood pressure effects of sodium reduction: dose-response meta-analysis of experimental studies. Circulation. 2021;143(16):1542–67. https://pubmed.ncbi.nlm.nih.gov/33586450/
Siriopol D, Covic A, Iliescu R, et al. Arterial stiffness mediates the effect of salt intake on systolic blood pressure. J Clin Hypertens (Greenwich). 2018;20(11):1587–94. https://pubmed.ncbi.nlm.nih.gov/30295011/
Santisteban MM, Iadecola C. Hypertension, dietary salt and cognitive impairment. J Cereb Blood Flow Metab. 2018;38(12):2112–28. https://pubmed.ncbi.nlm.nih.gov/30295560/
Faraco G, Hochrainer K, Segarra SG, et al. Dietary salt promotes cognitive impairment through tau phosphorylation. Nature. 2019;574(7780):686–90. https://pubmed.ncbi.nlm.nih.gov/31645758/
Fyfe I. High-salt diet promotes Alzheimer disease – like changes. Nat Rev Neurol. 2020;16(1):2–3. https://pubmed.ncbi.nlm.nih.gov/31712717/
Cahill S, Pierce M, Werner P, Darley A, Bobersky A. A systematic review of the public’s knowledge and understanding of Alzheimer’s disease and dementia. Alzheimer Dis Assoc Disord. 2015;29(3):255–75. https://pubmed.ncbi.nlm.nih.gov/26207322/
Hudson JM, Pollux PMJ, Mistry B, Hobson S. Beliefs about Alzheimer’s disease in Britain. Aging Ment Health. 2012;16(7):828–35. https://pubmed.ncbi.nlm.nih.gov/22416945/
Kivipelto M, Ngandu T, Laatikainen T, Winblad B, Soininen H, Tuomilehto J. Risk score for the prediction of dementia risk in 20 years among middle aged people: a longitudinal, population-based study. Lancet Neurol. 2006;5(9):735–41. https://pubmed.ncbi.nlm.nih.gov/16914401/
Chandra V, Ganguli M, Pandav R, et al. Prevalence of Alzheimer’s disease and other dementias in rural India: the Indo-US study. Neurology. 1998;51(4):1000–8. https://pubmed.ncbi.nlm.nih.gov/9781520/
Shetty PS. Nutrition transition in India. Public Health Nutr. 2002;5(1A):175–82. https://pubmed.ncbi.nlm.nih.gov/12027282/
Tsai JH, Huang CF, Lin MN, Chang CE, Chang CC, Lin CL. Taiwanese vegetarians are associated with lower dementia risk: a prospective cohort study. Nutrients. 2022;14(3):588. https://pubmed.ncbi.nlm.nih.gov/35276947/
Giem P, Beeson WL, Fraser GE. The incidence of dementia and intake of animal products: preliminary findings from the Adventist Health Study. Neuroepidemiology. 1993;12(1):28–36. https://pubmed.ncbi.nlm.nih.gov/8327020/
Kivipelto M, Helkala EL, Laakso MP, et al. Apolipoprotein E epsilon4 allele, elevated midlife total cholesterol level, and high midlife systolic blood pressure are independent risk factors for late-life Alzheimer disease. Ann Intern Med. 2002;137(3):149–55. https://pubmed.ncbi.nlm.nih.gov/12160362/
Deelen J, Evans DS, Arking DE, et al. A meta-analysis of genome-wide association studies identifies multiple longevity genes. Nat Commun. 2019;10(1):3669. https://pubmed.ncbi.nlm.nih.gov/31413261/
Rea IM, Dellet M, Mills KI, The ACUME2 Project. Living long and ageing well: is epigenomics the missing link between nature and nurture? Biogerontology. 2016;17(1):33–54. https://pubmed.ncbi.nlm.nih.gov/26133292/
Caruso C, Aiello A, Accardi G, Ciaglia E, Cattaneo M, Puca A. Genetic signatures of centenarians: implications for achieving successful aging. Curr Pharm Des. 2019;25(39):4133–8. https://pubmed.ncbi.nlm.nih.gov/31721694/
Abdullah MMH, Vazquez-Vidal I, Baer DJ, House JD, Jones PJH, Desmarchelier C. Common genetic variations involved in the inter-individual variability of circulating cholesterol concentrations in response to diets: a narrative review of recent evidence. Nutrients. 2021;13(2):695. https://pubmed.ncbi.nlm.nih.gov/33671529/
Sepehrnia B, Kamboh MI, Adams-Campbell LL, et al. Genetic studies of human apolipoproteins. X. The effect of the apolipoprotein E polymorphism on quantitative levels of lipoproteins in Nigerian blacks. Am J Hum Genet. 1989;45(4):586–91. https://pubmed.ncbi.nlm.nih.gov/2491016/
Laufs U, Dent R, Kostenuik PJ, Toth PP, Catapano AL, Chapman MJ. Why is hypercholesterolaemia so prevalent? A view from evolutionary medicine. Eur Heart J. 2019;40(33):2825–30. https://pubmed.ncbi.nlm.nih.gov/30169643/
World Health Organization. The top 10 causes of death. December 9, 2020. https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death. Accessed December 24, 2022.; https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death
Chiarini A, Armato U, Hu P, Dal Prà I. Danger-sensing/patten recognition receptors and neuroinflammation in Alzheimer’s disease. Int J Mol Sci. 2020;21(23):E9036. https://pubmed.ncbi.nlm.nih.gov/33261147/
Schmidt R, Schmidt H, Curb JD, Masaki K, White LR, Launer LJ. Early inflammation and dementia: a 25-year follow-up of the Honolulu-Asia Aging Study. Ann Neurol. 2002;52(2):168–74. https://pubmed.ncbi.nlm.nih.gov/12210786/
Jost BC, Grossberg GT. The natural history of Alzheimer’s disease: a brain bank study. J Am Geriatr Soc. 1995;43(11):1248–55. https://pubmed.ncbi.nlm.nih.gov/7594159/
Del Tredici K, Braak H. Neurofibrillary changes of the Alzheimer type in very elderly individuals: neither inevitable nor benign: Commentary on ‘No disease in the brain of a 115-year-old woman.’ Neurobiol Aging. 2008;29(8):1133–6. https://pubmed.ncbi.nlm.nih.gov/18584785/
Galasko DR, Peskind E, Clark CM, et al. Antioxidants for Alzheimer disease: a randomized clinical trial with cerebrospinal fluid biomarker measures. Arch Neurol. 2012;69(7):836–41. https://pubmed.ncbi.nlm.nih.gov/22431837/
Jensen MK, Cassidy A. Can dietary flavonoids play a role in Alzheimer’s disease risk prevention? Tantalizing population-based data out of Framingham. Am J Clin Nutr. 2020;112(2):241–2. https://pubmed.ncbi.nlm.nih.gov/32359140/
Shishtar E, Rogers GT, Blumberg JB, Au R, Jacques PF. Long-term dietary flavonoid intake and change in cognitive function in the Framingham Offspring cohort. Public Health Nutr. 2020;23(9):1576–88. https://pubmed.ncbi.nlm.nih.gov/32090722/
Tarozzi A, Morroni F, Merlicco A, et al. Neuroprotective effects of cyanidin 3-O-glucopyranoside on amyloid beta (25–35) oligomer-induced toxicity. Neurosci Lett. 2010;473(2):72–6. https://pubmed.ncbi.nlm.nih.gov/20152881/
Hattori M, Sugino E, Minoura K, et al. Different inhibitory response of cyanidin and methylene blue for filament formation of tau microtubule-binding domain. Biochem Biophys Res Commun. 2008;374(1):158–63. https://pubmed.ncbi.nlm.nih.gov/18619417/
Mandel SA, Weinreb O, Amit T, Youdim MB. Molecular mechanisms of the neuroprotective/neurorescue action of multi-target green tea polyphenols. Front Biosci (Schol Ed). 2012;4:581–98. https://pubmed.ncbi.nlm.nih.gov/22202078/
Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L. The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 2014;13(10):1045–60. https://pubmed.ncbi.nlm.nih.gov/25231526/
Crapper DR, Krishnan SS, Dalton AJ. Brain aluminum distribution in Alzheimer’s disease and experimental neurofibrillary degeneration. Science. 1973;180(4085):511–3. https://pubmed.ncbi.nlm.nih.gov/4735595/
Alfrey AC, LeGendre GR, Kaehny WD. The dialysis encephalopathy syndrome: possible aluminum intoxication. N Engl J Med. 1976;294(4):184–8. https://pubmed.ncbi.nlm.nih.gov/1244532/
Tomljenovic L. Aluminum and Alzheimer’s disease: after a century of controversy, is there a plausible link? J Alzheimers Dis. 2011;23(4):567–98. https://pubmed.ncbi.nlm.nih.gov/21157018/
Lidsky TI. Is the aluminum hypothesis dead? J Occup Environ Med. 2014;56(5 Suppl):S73–9. https://pubmed.ncbi.nlm.nih.gov/24806729/
Perl DP, Moalem S. Aluminum and Alzheimer’s disease, a personal perspective after 25 years. J Alzheimers Dis. 2006;9(3 Suppl):291–300. https://pubmed.ncbi.nlm.nih.gov/17004365/
Lidsky TI. Is the aluminum hypothesis dead? J Occup Environ Med. 2014;56(5 Suppl):S73–9. https://pubmed.ncbi.nlm.nih.gov/24806729/
Virk SA, Eslick GD. Brief report: meta-analysis of antacid use and Alzheimer’s disease: implications for the aluminum hypothesis. Epidemiology. 2015;26(5):769–73. https://pubmed.ncbi.nlm.nih.gov/26098935/
Reinke CM, Breitkreutz J, Leuenberger H. Aluminium in over-the-counter drugs: risks outweigh benefits? Drug Saf. 2003;26(14):1011–25. https://pubmed.ncbi.nlm.nih.gov/14583063/
Celik H, Celik N, Kocyigit A, Dikilitas M. The relationship between plasma aluminum content, lymphocyte DNA damage, and oxidative status in persons using aluminum containers and utensils daily. Clin Biochem. 2012;45(18):1629–33. https://pubmed.ncbi.nlm.nih.gov/22981396/
CRF – code of federal regulations Title 21. U.S Food & Drug Administration. https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?CFRPart=350&showFR=1. Updated March 29, 2022. Accessed July 4, 2022.; https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?CFRPart=350&showFR=1
Council of the European Communities. Council Directive 76/768/EEC of 27 July 1976 on the approximation of the laws of the Member States relating to cosmetic products. EUR-Lex. https://eur-lex.europa.eu/legal-content/EN/ALL/?uri=celex%3A31976L0768. Published July 27, 1976. Accessed February 24, 2023.; https://eur-lex.europa.eu/legal-content/EN/ALL/?uri=celex%3A31976L0768
Darbre PD, Mannello F, Exley C. Aluminium and breast cancer: Sources of exposure, tissue measurements and mechanisms of toxicological actions on breast biology. J Inorg Biochem. 2013;128:257–61. https://pubmed.ncbi.nlm.nih.gov/23899626/
Darbre PD. Metalloestrogens: an emerging class of inorganic xenoestrogens with potential to add to the oestrogenic burden of the human breast. J Appl Toxicol. 2006;26(3):191–7. https://pubmed.ncbi.nlm.nih.gov/16489580/
McGrath KG. An earlier age of breast cancer diagnosis related to more frequent use of antiperspirants/deodorants and underarm shaving. Eur J Cancer Prev. 2003;12(6):479–85. https://pubmed.ncbi.nlm.nih.gov/14639125/
Yokel RA, Hicks CL, Florence RL. Aluminum bioavailability from basic sodium aluminum phosphate, an approved food additive emulsifying agent, incorporated in cheese. Food Chem Toxicol. 2008;46(6):2261–6. https://pubmed.ncbi.nlm.nih.gov/18436363/
Al-Ashmawy MAM. Prevalence and public health significance of aluminum residues in milk and some dairy products. J Food Sci. 2011;76(3):T73–6. https://pubmed.ncbi.nlm.nih.gov/21535864/
Gleason A, Bush AI. Iron and ferroptosis as therapeutic targets in Alzheimer’s disease. Neurotherapeutics. 2021;18(1):252–64. https://pubmed.ncbi.nlm.nih.gov/33111259/
Nikseresht S, Bush AI, Ayton S. Treating Alzheimer’s disease by targeting iron. Br J Pharmacol. 2019;176(18):3622–35. https://pubmed.ncbi.nlm.nih.gov/30632143/
Gleason A, Bush AI. Iron and ferroptosis as therapeutic targets in Alzheimer’s disease. Neurotherapeutics. 2021;18(1):252–64. https://pubmed.ncbi.nlm.nih.gov/33111259/
Ayton S, James SA, Bush AI. Nanoscale imaging reveals big role for iron in Alzheimer’s disease. Cell Chem Biol. 2017;24(10):1192–4. https://pubmed.ncbi.nlm.nih.gov/29053948/
Ayton S, Diouf I, Bush AI, Alzheimer’s disease Neuroimaging Initiative. Evidence that iron accelerates Alzheimer’s pathology: a CSF biomarker study. J Neurol Neurosurg Psychiatry. 2018;89(5):456–60. https://pubmed.ncbi.nlm.nih.gov/28939683/
Miller LM, Wang Q, Telivala TP, Smith RJ, Lanzirotti A, Miklossy J. Synchrotron-based infrared and X-ray imaging shows focalized accumulation of Cu and Zn co-localized with beta-amyloid deposits in Alzheimer’s disease. J Struct Biol. 2006;155(1):30–7. https://pubmed.ncbi.nlm.nih.gov/16325427/
Morris MC, Evans DA, Tangney CC, et al. Dietary copper and high saturated and trans fat intakes associated with cognitive decline. Arch Neurol. 2006;63(8):1085–8. https://pubmed.ncbi.nlm.nih.gov/16908733/
Loef M, Walach H. Copper and iron in Alzheimer’s disease: a systematic review and its dietary implications. Br J Nutr. 2012;107(1):7–19. https://pubmed.ncbi.nlm.nih.gov/21767446/
Liyanage SI, Vilekar P, Weaver DF. Nutrients in Alzheimer’s disease: the interaction of diet, drugs and disease. Can J Neurol Sci. 2019;46(1):23–34. https://pubmed.ncbi.nlm.nih.gov/30688198/
Oleson S, Gonzales MM, Tarumi T, et al. Nutrient intake and cerebral metabolism in healthy middle-aged adults: implications for cognitive aging. Nutr Neurosci. 2017;20(8):489–96. https://pubmed.ncbi.nlm.nih.gov/27237189/
Okereke OI, Rosner BA, Kim DH, et al. Dietary fat types and 4-year cognitive change in community-dwelling older women. Ann Neurol. 2012;72(1):124–34. https://pubmed.ncbi.nlm.nih.gov/22605573/
Cao GY, Li M, Han L, et al. Dietary fat intake and cognitive function among older populations: a systematic review and meta-analysis. J Prev Alzheimers Dis. 2019;6(3):204–11. https://pubmed.ncbi.nlm.nih.gov/31062836/
Barbaresko J, Lellmann AW, Schmidt A, et al. Dietary factors and neurodegenerative disorders: an umbrella review of meta-analyses of prospective studies. Adv Nutr. 2020;11(5):1161–73. https://pubmed.ncbi.nlm.nih.gov/32427314/
Liyanage SI, Vilekar P, Weaver DF. Nutrients in Alzheimer’s disease: the interaction of diet, drugs and disease. Can J Neurol Sci. 2019;46(1):23–34. https://pubmed.ncbi.nlm.nih.gov/30688198/
Kahle L, Krebs-Smith SM, Reedy J, Rodgers AB, Signes C. Identification of Top Food Sources of Various Dietary Components. National Cancer Institute. https://epi.grants.cancer.gov/diet/foodsources. Updated June 8, 2022. Accessed June 30, 2022.; https://epi.grants.cancer.gov/diet/foodsources
Wahl D, Solon-Biet SM, Cogger VC, et al. Aging, lifestyle and dementia. Neurobiol Dis. 2019;130:104481. https://pubmed.ncbi.nlm.nih.gov/31136814/
Verheggen ICM, de Jong JJA, van Boxtel MPJ, et al. Increase in blood-brain barrier leakage in healthy, older adults. Geroscience. 2020;42(4):1183–93. https://pubmed.ncbi.nlm.nih.gov/32601792/
Farrall AJ, Wardlaw JM. Blood-brain barrier: ageing and microvascular disease – systematic review and meta-analysis. Neurobiol Aging. 2009;30(3):337–52. https://pubmed.ncbi.nlm.nih.gov/17869382/
Nation DA, Sweeney MD, Montagne A, et al. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med. 2019;25(2):270–6. https://pubmed.ncbi.nlm.nih.gov/30643288/
Verheggen ICM, de Jong JJA, van Boxtel MPJ, et al. Increase in blood-brain barrier leakage in healthy, older adults. Geroscience. 2020;42(4):1183–93. https://pubmed.ncbi.nlm.nih.gov/32601792/
Farrall AJ, Wardlaw JM. Blood-brain barrier: ageing and microvascular disease – systematic review and meta-analysis. Neurobiol Aging. 2009;30(3):337–52. https://pubmed.ncbi.nlm.nih.gov/17869382/
Verheggen ICM, de Jong JJA, van Boxtel MPJ, et al. Increase in blood-brain barrier leakage in healthy, older adults. Geroscience. 2020;42(4):1183–93. https://pubmed.ncbi.nlm.nih.gov/32601792/
Gustafson DR, Karlsson C, Skoog I, Rosengren L, Lissner L, Blennow K. Mid-life adiposity factors relate to blood-brain barrier integrity in late life. J Intern Med. 2007;262(6):643–50. https://pubmed.ncbi.nlm.nih.gov/17986201/
Freeman LR, Granholm ACE. Vascular changes in rat hippocampus following a high saturated fat and cholesterol diet. J Cereb Blood Flow Metab. 2012;32(4):643–53. https://pubmed.ncbi.nlm.nih.gov/22108721/
Ghribi O, Golovko MY, Larsen B, Schrag M, Murphy EJ. Deposition of iron and ß-amyloid plaques is associated with cortical cellular damage in rabbits fed with long-term cholesterol-enriched diets. J Neurochem. 2006;99(2):438–49. https://pubmed.ncbi.nlm.nih.gov/17029598/
Takechi R, Galloway S, Pallebage-Gamarallage MM, Lam V, Dhaliwal SS, Mamo JC. Probucol prevents blood – brain barrier dysfunction in wild-type mice induced by saturated fat or cholesterol feeding. Clin Exp Pharmacol Physiol. 2013;40(1):45–52. https://pubmed.ncbi.nlm.nih.gov/23167559/
Galloway S, Takechi R, Nesbit M, Pallebage-Gamarallage MM, Lam V, Mamo JCL. The differential effects of fatty acids on enterocytic abundance of amyloid-beta. Lipids Health Dis. 2019;18(1):209. https://pubmed.ncbi.nlm.nih.gov/31796080/
Boyt AA, Taddei K, Hallmayer J, et al. Relationship between lipid metabolism and plasma concentration of amyloid precursor protein and apolipoprotein E. Alzheimer’s Rep. 1999;2(6):339–46. https://researchers.mq.edu.au/en/publications/relationship-between-lipid-metabolism-and-plasma-concentration-of
Takechi R, Galloway S, Pallebage-Gamarallage MMS, Lam V, Mamo JCL. Dietary fats, cerebrovasculature integrity and Alzheimer’s disease risk. Prog Lipid Res. 2010;49(2):159–70. https://pubmed.ncbi.nlm.nih.gov/19896503/
Kauwe G, Tracy TE. Amyloid beta emerges from below the neck to disable the brain. PLoS Biol. 2021;19(9):e3001388. https://pubmed.ncbi.nlm.nih.gov/34525093/
Edwards LM, Murray AJ, Holloway CJ, et al. Short-term consumption of a high-fat diet impairs whole-body efficiency and cognitive function in sedentary men. FASEB J. 2011;25(3):1088–96. https://pubmed.ncbi.nlm.nih.gov/21106937/
Attuquayefio T, Stevenson RJ, Oaten MJ, Francis HM. A four-day Western-style dietary intervention causes reductions in hippocampal-dependent learning and memory and interoceptive sensitivity. PLoS ONE. 2017;12(2):e0172645. https://pubmed.ncbi.nlm.nih.gov/28231304/
Madison AA, Belury MA, Andridge R, et al. Afternoon distraction: a high-saturated-fat meal and endotoxemia impact postmeal attention in a randomized crossover trial. Am J Clin Nutr. 2020;111(6):1150–8. https://pubmed.ncbi.nlm.nih.gov/32393980/
Valdearcos M, Robblee MM, Benjamin DI, Nomura DK, Xu AW, Koliwad SK. Microglia dictate the impact of saturated fat consumption on hypothalamic inflammation and neuronal function. Cell Rep. 2014;9(6):2124–38. https://pubmed.ncbi.nlm.nih.gov/25497089/
Laposata M. Fatty acids: biochemistry to clinical significance. Am J Clin Pathol. 1995;104(2):172–9. https://pubmed.ncbi.nlm.nih.gov/7639192/
Sergi D, Kahn DE, Morris AC, Williams LM. Palmitic acid induces inflammation in hypothalamic neurons via ceramide synthesis. Proc Nutr Soc. 2016;75(OCE2):E46. https://www.cambridge.org/core/journals/proceedings-of-the-nutrition-society/article/palmitic-acid-induces-inflammation-in-hypothalamic-neurons-via-ceramide-synthesis/CA8587EB22475DA81A73C991C568DB3B
Berkseth KE, Guyenet SJ, Melhorn SJ, et al. Hypothalamic gliosis associated with high-fat diet feeding is reversible in mice: a combined immunohistochemical and magnetic resonance imaging study. Endocrinology. 2014;155(8):2858–67. https://pubmed.ncbi.nlm.nih.gov/24914942/
Ioannidis JP. Extrapolating from animals to humans. Sci Transl Med. 2012;4(151):151ps15. https://pubmed.ncbi.nlm.nih.gov/22972841/
Borg ML, Omran SF, Weir J, Meikle PJ, Watt MJ. Consumption of a high-fat diet, but not regular endurance exercise training, regulates hypothalamic lipid accumulation in mice. J Physiol (Lond). 2012;590(17):4377–89. https://pubmed.ncbi.nlm.nih.gov/22674717/
Agricultural Research Service, United States Department of Agriculture. Pork, cured, bacon, cooked, baked. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/food-details/167914/nutrients. Published April 1, 2019. Accessed June 30, 2022.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/167914/nutrients
Kien CL, Bunn JY, Tompkins CL, et al. Substituting dietary monounsaturated fat for saturated fat is associated with increased daily physical activity and resting energy expenditure and with changes in mood. Am J Clin Nutr. 2013;97(4):689–97. https://pubmed.ncbi.nlm.nih.gov/23446891/
Dumas JA, Bunn JY, Nickerson J, et al. Dietary saturated fat and monounsaturated fat have reversible effects on brain function and the secretion of pro-inflammatory cytokines in young women. Metab Clin Exp. 2016;65(10):1582–8. https://pubmed.ncbi.nlm.nih.gov/27621193/
Kien CL, Bunn JY, Tompkins CL, et al. Substituting dietary monounsaturated fat for saturated fat is associated with increased daily physical activity and resting energy expenditure and with changes in mood. Am J Clin Nutr. 2013;97(4):689–97. https://pubmed.ncbi.nlm.nih.gov/23446891/
Sherzai D, Sherzai A. Preventing Alzheimer’s: our most urgent health care priority. Am J Lifestyle Med. 2019;13(5):451–61. https://pubmed.ncbi.nlm.nih.gov/31523210/
West RK, Moshier E, Lubitz I, et al. Dietary advanced glycation end products are associated with decline in memory in young elderly. Mech Ageing Dev. 2014;140:10–2. https://pubmed.ncbi.nlm.nih.gov/25037023/
Srikanth V, Westcott B, Forbes J, et al. Methylglyoxal, cognitive function and cerebral atrophy in older people. J Gerontol A Biol Sci Med Sci. 2013;68(1):68–73. https://pubmed.ncbi.nlm.nih.gov/22496536/
Igase M, Ohara M, Igase K, et al. Skin autofluorescence examination as a diagnostic tool for mild cognitive impairment in healthy people. J Alzheimers Dis. 2017;55(4):1481–7. https://pubmed.ncbi.nlm.nih.gov/27858716/
Ko S, Ko H, Chu K, et al. The possible mechanism of advanced glycation end products (AGEs) for Alzheimer’s disease. PLoS One. 2015;10(11):e0143345. https://pubmed.ncbi.nlm.nih.gov/26587989/
Chou P, Wu M, Yang C, Shen C, Yang Y. Effect of advanced glycation end products on the progression of Alzheimer’s disease. J Alzheimers Dis. 2019;72(1):191–7. https://pubmed.ncbi.nlm.nih.gov/31561370/
Kim K-S, Lee Y-M, Lee H-W, Jacobs DR, Lee D-H. Associations between organochlorine pesticides and cognition in U.S. elders: National Health and Nutrition Examination Survey 1999–2002. Environ Int. 2015;75:87–92. https://pubmed.ncbi.nlm.nih.gov/25461417/
Bernard A. Elevated serum DDE and risk for Alzheimer disease. JAMA Neurol. 2014;71(8):1055–6. https://pubmed.ncbi.nlm.nih.gov/25111212/
Schecter A, Cramer P, Boggess K, Stanley J, Olson JR. Levels of dioxins, dibenzofurans, PCB and DDE congeners in pooled food samples collected in 1995 at supermarkets across the United States. Chemosphere. 1997;34(5–7):1437–47. https://pubmed.ncbi.nlm.nih.gov/9134677/
André P, Laugerette F, Féart C. Metabolic endotoxemia: a potential underlying mechanism of the relationship between dietary fat intake and risk for cognitive impairments in humans? Nutrients. 2019;11(8):1887. https://pubmed.ncbi.nlm.nih.gov/31412673/
Ghanim H, Batra M, Abuaysheh S, et al. Antiinflammatory and ROS suppressive effects of the addition of fiber to a high-fat high-calorie meal. J Clin Endocrinol Metab. 2017;102(3):858–69. https://pubmed.ncbi.nlm.nih.gov/27906549/
Risk Reduction of Cognitive Decline and Dementia: WHO Guidelines. World Health Organization; 2019. https://www.who.int/publications/i/item/9789241550543
Livingston G, Huntley J, Sommerlad A, et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet. 2020;396(10248):413–46. https://pubmed.ncbi.nlm.nih.gov/32738937/
Theadom A, Mahon S, Hume P, et al. Incidence of sports-related traumatic brain injury of all severities: a systematic review. Neuroepidemiology. 2020;54(2):192–9. https://pubmed.ncbi.nlm.nih.gov/31935738/
Mackay DF, Russell ER, Stewart K, MacLean JA, Pell JP, Stewart W. Neurodegenerative disease mortality in former professional soccer players. N Engl J Med. 2019;381(19):1801–8. https://pubmed.ncbi.nlm.nih.gov/31633894/
Asken BM, Rabinovici GD. Professional soccer and dementia risk – the ugly side of the beautiful game. JAMA Neurol. 2021;78(9):1049–51. https://pubmed.ncbi.nlm.nih.gov/34338741/
Walton SR, Brett BL, Chandran A, et al. Mild cognitive impairment and dementia reported by former professional football players over 50 yr of age: an NFL–LONG study. Med Sci Sports Exerc. 2022;54(3):424–31. https://pubmed.ncbi.nlm.nih.gov/34593716/
Castellani RJ, Perry G. Dementia pugilistica revisited. J Alzheimers Dis. 60(4):1209–21.; https://pubmed.ncbi.nlm.nih.gov/29036831/
GBD 2019 Dementia Collaborators. The burden of dementia due to Down syndrome, Parkinson’s disease, stroke, and traumatic brain injury: a systematic analysis for the Global Burden of Disease Study 2019. Neuroepidemiology. 2021;55(4):286–96. https://pubmed.ncbi.nlm.nih.gov/34182555/
Høye A. Bicycle helmets – to wear or not to wear? A meta-analyses of the effects of bicycle helmets on injuries. Accid Anal Prev. 2018;117:85–97. https://pubmed.ncbi.nlm.nih.gov/29677686/
Enniss TM, Basiouny K, Brewer B, et al. Primary prevention of contact sports – related concussions in amateur athletes: a systematic review from the Eastern Association for the Surgery of Trauma. Trauma Surg Acute Care Open. 2018;3(1):e000153. https://pubmed.ncbi.nlm.nih.gov/30023433/
Emery CA, Black AM, Kolstad A, et al. What strategies can be used to effectively reduce the risk of concussion in sport? A systematic review. Br J Sports Med. 2017;51(12):978–84. https://pubmed.ncbi.nlm.nih.gov/28254746/
Kivipelto M, Mangialasche F, Ngandu T. Lifestyle interventions to prevent cognitive impairment, dementia and Alzheimer disease. Nat Rev Neurol. 2018;14(11):653–66. https://pubmed.ncbi.nlm.nih.gov/30291317/
Wahl D, Solon-Biet SM, Cogger VC, et al. Aging, lifestyle and dementia. Neurobiol Dis. 2019;130:104481. https://pubmed.ncbi.nlm.nih.gov/31136814/
Zhong G, Wang Y, Zhang Y, Guo JJ, Zhao Y. Smoking is associated with an increased risk of dementia: a meta-analysis of prospective cohort studies with investigation of potential effect modifiers. PLoS One. 2015;10(3):e0118333. https://pubmed.ncbi.nlm.nih.gov/25763939/
Almeida OP, Garrido GJ, Alfonso H, et al. 24-month effect of smoking cessation on cognitive function and brain structure in later life. Neuroimage. 2011;55(4):1480–9. https://pubmed.ncbi.nlm.nih.gov/21281718/
Serrano-Pozo A, Growdon JH. Is Alzheimer’s disease risk modifiable? J Alzheimers Dis. 2019;67(3):795–819. https://pubmed.ncbi.nlm.nih.gov/30776012/
Delgado-Saborit JM, Guercio V, Gowers AM, Shaddick G, Fox NC, Love S. A critical review of the epidemiological evidence of effects of air pollution on dementia, cognitive function and cognitive decline in adult population. Sci Total Environ. 2021;757:143734. https://pubmed.ncbi.nlm.nih.gov/33340865/
Calderón-Garcidueñas L, Azzarelli B, Acuna H, et al. Air pollution and brain damage. Toxicol Pathol. 2002;30(3):373–89. https://pubmed.ncbi.nlm.nih.gov/12051555/
Maher BA, Ahmed IAM, Karloukovski V, et al. Magnetite pollution nanoparticles in the human brain. Proc Natl Acad Sci U S A. 2016;113(39):10797–801. https://pubmed.ncbi.nlm.nih.gov/27601646/
Delgado-Saborit JM, Guercio V, Gowers AM, Shaddick G, Fox NC, Love S. A critical review of the epidemiological evidence of effects of air pollution on dementia, cognitive function and cognitive decline in adult population. Sci Total Environ. 2021;757:143734. https://pubmed.ncbi.nlm.nih.gov/33340865/
Gupta S, Warner J. Alcohol-related dementia: a 21st-century silent epidemic? Br J Psychiatry. 2008;193(5):351–3. https://pubmed.ncbi.nlm.nih.gov/18978310/
Wahl D, Solon-Biet SM, Cogger VC, et al. Aging, lifestyle and dementia. Neurobiol Dis. 2019;130:104481. https://pubmed.ncbi.nlm.nih.gov/31136814/
Brennan SE, McDonald S, Page MJ, et al. Long-term effects of alcohol consumption on cognitive function: a systematic review and dose-response analysis of evidence published between 2007 and 2018. Syst Rev. 2020;9(1):33. https://pubmed.ncbi.nlm.nih.gov/32054517/
Andrews SJ, Goate A, Anstey KJ. Association between alcohol consumption and Alzheimer’s disease: a Mendelian randomization study. Alzheimers Dement. 2020;16(2):345–53. https://pubmed.ncbi.nlm.nih.gov/31786126/
van Eijk J, Demirakca T, Frischknecht U, Hermann D, Mann K, Ende G. Rapid partial regeneration of brain volume during the first 14 days of abstinence from alcohol. Alcohol Clin Exp Res. 2013;37(1):67–74. https://pubmed.ncbi.nlm.nih.gov/23072363/
Luna S, Cameron DJ, Ethell DW. Amyloid-ß and APP deficiencies cause severe cerebrovascular defects: important work for an old villain. PLoS One. 2013;8(9):e75052. https://pubmed.ncbi.nlm.nih.gov/24040383/
Soscia SJ, Kirby JE, Washicosky KJ, et al. The Alzheimer’s disease – associated amyloid beta-protein is an antimicrobial peptide. PLoS One. 2010;5(3):e9505. https://pubmed.ncbi.nlm.nih.gov/20209079/
Eimer WA, Vijaya Kumar DK, Navalpur Shanmugam NK, et al. Alzheimer’s disease – associated ß-amyloid is rapidly seeded by Herpesviridae to protect against brain infection. Neuron. 2018;99(1):56–63.e3. https://pubmed.ncbi.nlm.nih.gov/30001512/
Itzhaki RF, Lathe R, Balin BJ, et al. Microbes and Alzheimer’s disease. J Alzheimers Dis. 2016;51(4):979–84. https://pubmed.ncbi.nlm.nih.gov/26967229/
Tzeng NS, Chung CH, Lin FH, et al. Anti-herpetic medications and reduced risk of dementia in patients with herpes simplex virus infections – a nationwide, population-based cohort study in Taiwan. Neurotherapeutics. 2018;15(2):417–29. https://pubmed.ncbi.nlm.nih.gov/29488144/
Ogilvie RP, Patel SR. The epidemiology of sleep and obesity. Sleep Health. 2017;3(5):383–8. https://pubmed.ncbi.nlm.nih.gov/28923198/
Everson CA, Bergmann BM, Rechtschaffen A. Sleep deprivation in the rat: III. Total sleep deprivation. Sleep. 1989;12(1):13–21. https://pubmed.ncbi.nlm.nih.gov/2928622/
Xie L, Kang H, Xu Q, et al. Sleep drives metabolite clearance from the adult brain. Science. 2013;342(6156):373–7. https://pubmed.ncbi.nlm.nih.gov/24136970/
Absinta M, Ha SK, Nair G, et al. Human and nonhuman primate meninges harbor lymphatic vessels that can be visualized noninvasively by MRI. Elife. 2017;6:e29738. https://pubmed.ncbi.nlm.nih.gov/28971799/
Shokri-Kojori E, Wang G, Wiers C, et al. ß-Amyloid accumulation in the human brain after one night of sleep deprivation. PNAS. 2018;115(17):4483–8. https://pubmed.ncbi.nlm.nih.gov/29632177/
Kress BT, Iliff JJ, Xia M, et al. Impairment of paravascular clearance pathways in the aging brain. Ann Neurol. 2014;76(6):845–61. https://pubmed.ncbi.nlm.nih.gov/25204284/
Simka M, Czaja J, Kowalczyk D. Collapsibility of the internal jugular veins in the lateral decubitus body position: a potential protective role of the cerebral venous outflow against neurodegeneration. Med Hypotheses. 2019;133:109397. https://pubmed.ncbi.nlm.nih.gov/31526984/
Romanella SM, Roe D, Tatti E, et al. The sleep side of aging and Alzheimer’s disease. Sleep Med. 2021;77:209–25. https://pubmed.ncbi.nlm.nih.gov/32912799/
Brusco LI, Márquez M, Cardinali DP. Monozygotic twins with Alzheimer’s disease treated with melatonin: case report. J Pineal Res. 1998;25(4):260–3. https://pubmed.ncbi.nlm.nih.gov/9885996/
Vincent B. Protective roles of melatonin against the amyloid-dependent development of Alzheimer’s disease: a critical review. Pharmacol Res. 2018;134:223–37. https://pubmed.ncbi.nlm.nih.gov/29981776/
Wang YY, Zheng W, Ng CH, Ungvari GS, Wei W, Xiang YT. Meta-analysis of randomized, double-blind, placebo-controlled trials of melatonin in Alzheimer’s disease. Int J Geriatr Psychiatry. 2017;32(1):50–7. https://pubmed.ncbi.nlm.nih.gov/27645169/
Loef M, Walach H. Midlife obesity and dementia: meta-analysis and adjusted forecast of dementia prevalence in the United States and China. Obesity (Silver Spring). 2013;21(1):E51–5. https://pubmed.ncbi.nlm.nih.gov/23401370/
Yang Y, Shields GS, Guo C, Liu Y. Executive function performance in obesity and overweight individuals: a meta-analysis and review. Neurosci Biobehav Rev. 2018;84:225–44. https://pubmed.ncbi.nlm.nih.gov/29203421/
Walther K, Birdsill AC, Glisky EL, Ryan L. Structural brain differences and cognitive functioning related to body mass index in older females. Hum Brain Mapp. 2010;31(7):1052–64. https://pubmed.ncbi.nlm.nih.gov/19998366/
Willette AA, Kapogiannis D. Does the brain shrink as the waist expands? Ageing Res Rev. 2015;20:86–97. https://pubmed.ncbi.nlm.nih.gov/24768742/
Veronese N, Facchini S, Stubbs B, et al. Weight loss is associated with improvements in cognitive function among overweight and obese people: a systematic review and meta-analysis. Neurosci Biobehav Rev. 2017;72:87–94. https://pubmed.ncbi.nlm.nih.gov/27890688/
Veronese N, Facchini S, Stubbs B, et al. Weight loss is associated with improvements in cognitive function among overweight and obese people: a systematic review and meta-analysis. Neurosci Biobehav Rev. 2017;72:87–94. https://pubmed.ncbi.nlm.nih.gov/27890688/
Napoli N, Shah K, Waters DL, Sinacore DR, Qualls C, Villareal DT. Effect of weight loss, exercise, or both on cognition and quality of life in obese older adults. Am J Clin Nutr. 2014;100(1):189–98. https://pubmed.ncbi.nlm.nih.gov/24787497/
Erickson KI, Hillman C, Stillman CM, et al. Physical activity, cognition, and brain outcomes: a review of the 2018 physical activity guidelines. Med Sci Sports Exerc. 2019;51(6):1242–51. https://pubmed.ncbi.nlm.nih.gov/31095081/
Lee J. Effects of aerobic and resistance exercise interventions on cognitive and physiologic adaptations for older adults with mild cognitive impairment: a systematic review and meta-analysis of randomized control trials. Int J Environ Res Public Health. 2020;17(24):E9216. https://pubmed.ncbi.nlm.nih.gov/33317169/
Gomes-Osman J, Cabral DF, Morris TP, et al. Exercise for cognitive brain health in aging: a systematic review for an evaluation of dose. Neurol Clin Pract. 2018;8(3):257–65. https://pubmed.ncbi.nlm.nih.gov/30105166/
Sanders LMJ, Hortobágyi T, la Bastide-van Gemert S, van der Zee EA, van Heuvelen MJG. Dose-response relationship between exercise and cognitive function in older adults with and without cognitive impairment: a systematic review and meta-analysis. PLoS One. 2019;14(1):e0210036. https://pubmed.ncbi.nlm.nih.gov/30629631/
Gomes-Osman J, Cabral DF, Morris TP, et al. Exercise for cognitive brain health in aging: a systematic review for an evaluation of dose. Neurol Clin Pract. 2018;8(3):257–65. https://pubmed.ncbi.nlm.nih.gov/30105166/
Lamb SE, Sheehan B, Atherton N, et al. Dementia And Physical Activity (DAPA) trial of moderate to high intensity exercise training for people with dementia: randomised controlled trial. BMJ. 2018;361:k1675. https://pubmed.ncbi.nlm.nih.gov/29769247/
Ng TKS, Ho CSH, Tam WWS, Kua EH, Ho RCM. Decreased serum brain-derived neurotrophic factor (BDNF) levels in patients with Alzheimer’s disease (AD): a systematic review and meta-analysis. Int J Mol Sci. 2019;20(2):E257. https://pubmed.ncbi.nlm.nih.gov/30634650/
Qin XY, Cao C, Cawley NX, et al. Decreased peripheral brain-derived neurotrophic factor levels in Alzheimer’s disease: a meta-analysis study (N=7277). Mol Psychiatry. 2017;22(2):312–20. https://pubmed.ncbi.nlm.nih.gov/27113997/
Hsu TM, Kanoski SE. Blood-brain barrier disruption: mechanistic links between Western diet consumption and dementia. Front Aging Neurosci. 2014;6:88. https://pubmed.ncbi.nlm.nih.gov/24847262/
Du Y, Wu HT, Qin XY, et al. Postmortem brain, cerebrospinal fluid, and blood neurotrophic factor levels in Alzheimer’s disease: a systematic review and meta-analysis. J Mol Neurosci. 2018;65(3):289–300. https://pubmed.ncbi.nlm.nih.gov/29956088/
Qin XY, Cao C, Cawley NX, et al. Decreased peripheral brain-derived neurotrophic factor levels in Alzheimer’s disease: a meta-analysis study (N=7277). Mol Psychiatry. 2017;22(2):312–20. https://pubmed.ncbi.nlm.nih.gov/27113997/
Ng TKS, Ho CSH, Tam WWS, Kua EH, Ho RCM. Decreased serum brain-derived neurotrophic factor (BDNF) levels in patients with Alzheimer’s disease (AD): a systematic review and meta-analysis. Int J Mol Sci. 2019;20(2):E257. https://pubmed.ncbi.nlm.nih.gov/30634650/
Qin XY, Cao C, Cawley NX, et al. Decreased peripheral brain-derived neurotrophic factor levels in Alzheimer’s disease: a meta-analysis study (N=7277). Mol Psychiatry. 2017;22(2):312–20. https://pubmed.ncbi.nlm.nih.gov/27113997/
Lima Giacobbo B, Doorduin J, Klein HC, Dierckx RAJO, Bromberg E, de Vries EFJ. Brain-derived neurotrophic factor in brain disorders: focus on neuroinflammation. Mol Neurobiol. 2019;56(5):3295–312. https://pubmed.ncbi.nlm.nih.gov/30117106/
Weinstein G, Beiser AS, Choi SH, et al. Serum brain-derived neurotrophic factor and the risk for dementia: the Framingham Heart Study. JAMA Neurol. 2014;71(1):55–61. https://pubmed.ncbi.nlm.nih.gov/24276217/
Laske C, Stellos K, Hoffmann N, et al. Higher BDNF serum levels predict slower cognitive decline in Alzheimer’s disease patients. Int J Neuropsychopharmacol. 2011;14(3):399–404. https://pubmed.ncbi.nlm.nih.gov/20860877/
McPhee GM, Downey LA, Stough C. Neurotrophins as a reliable biomarker for brain function, structure and cognition: a systematic review and meta-analysis. Neurobiol Learn Mem. 2020;175:107298. https://pubmed.ncbi.nlm.nih.gov/32822863/
Lima Giacobbo B, Doorduin J, Klein HC, Dierckx RAJO, Bromberg E, de Vries EFJ. Brain-derived neurotrophic factor in brain disorders: focus on neuroinflammation. Mol Neurobiol. 2019;56(5):3295–312. https://pubmed.ncbi.nlm.nih.gov/30117106/
Szuhany KL, Bugatti M, Otto MW. A meta-analytic review of the effects of exercise on brain-derived neurotrophic factor. J Psychiatr Res. 2015;60:56–64. https://pubmed.ncbi.nlm.nih.gov/25455510/
Marquez CMS, Vanaudenaerde B, Troosters T, Wenderoth N. High-intensity interval training evokes larger serum BDNF levels compared with intense continuous exercise. J Appl Physiol (1985). 2015;119(12):1363–73. https://pubmed.ncbi.nlm.nih.gov/26472862/
Ferris LT, Williams JS, Shen CL. The effect of acute exercise on serum brain-derived neurotrophic factor levels and cognitive function. Med Sci Sports Exerc. 2007;39(4):728–34. https://pubmed.ncbi.nlm.nih.gov/17414812/
Coelho FM, Pereira DS, Lustosa LP, et al. Physical therapy intervention (PTI) increases plasma brain-derived neurotrophic factor (BDNF) levels in non-frail and pre-frail elderly women. Arch Gerontol Geriatr. 2012;54(3):415–20. https://pubmed.ncbi.nlm.nih.gov/21684022/
Loprinzi PD. Does brain-derived neurotrophic factor mediate the effects of exercise on memory? Phys Sportsmed. 2019;47(4):395–405. https://pubmed.ncbi.nlm.nih.gov/31002004/
Guelpa G. Starvation and purgation in the relief of disease. Br Med J. 1910;2(2597):1050–1. https://www.jstor.org/stable/25292424
Watkins E, Serpell L. The psychological effects of short-term fasting in healthy women. Front Nutr. 2016;3:27. https://pubmed.ncbi.nlm.nih.gov/27597946/
Fond G, Macgregor A, Leboyer M, Michalsen A. Fasting in mood disorders: neurobiology and effectiveness. A review of the literature. Psychiatry Res. 2013;209(3):253–8. https://pubmed.ncbi.nlm.nih.gov/23332541/
Araya AV, Orellana X, Espinoza J. Evaluation of the effect of caloric restriction on serum BDNF in overweight and obese subjects: preliminary evidences. Endocrine. 2008;33(3):300–4. https://pubmed.ncbi.nlm.nih.gov/19012000/
Witte AV, Fobker M, Gellner R, Knecht S, Flöel A. Caloric restriction improves memory in elderly humans. Proc Natl Acad Sci U S A. 2009;106(4):1255–60. https://pubmed.ncbi.nlm.nih.gov/19171901/
Araya AV, Orellana X, Espinoza J. Evaluation of the effect of caloric restriction on serum BDNF in overweight and obese subjects: preliminary evidences. Endocrine. 2008;33(3):300–4. https://pubmed.ncbi.nlm.nih.gov/19012000/
Guimarães LR, Jacka FN, Gama CS, et al. Serum levels of brain-derived neurotrophic factor in schizophrenia on a hypocaloric diet. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(6):1595–8. https://pubmed.ncbi.nlm.nih.gov/18582525/
Karczewska-Kupczewska M, Kowalska I, Nikolajuk A, et al. Circulating brain-derived neurotrophic factor concentration is downregulated by intralipid/heparin infusion or high-fat meal in young healthy male subjects. Diabetes Care. 2012;35(2):358–62. https://pubmed.ncbi.nlm.nih.gov/22210566/
Park HR, Park M, Choi J, Park KY, Chung HY, Lee J. A high-fat diet impairs neurogenesis: involvement of lipid peroxidation and brain-derived neurotrophic factor. Neurosci Lett. 2010;482(3):235–9. https://pubmed.ncbi.nlm.nih.gov/20670674/
Cott A. Controlled fasting treatment for schizophrenia. Orthomolecular Psychiatry. 1974;3(4):301–11. https://isom.ca/wp-content/uploads/2020/01/JOM_1974_03_4_12_Controlled_Fasting_Treatment_for_Schizophrenia.pdf
Beezhold BL, Johnston CS. Restriction of meat, fish, and poultry in omnivores improves mood: a pilot randomized controlled trial. Nutr J. 2012;11:9. https://pubmed.ncbi.nlm.nih.gov/22333737/
Neshatdoust S, Saunders C, Castle SM, et al. High-flavonoid intake induces cognitive improvements linked to changes in serum brain-derived neurotrophic factor: two randomised, controlled trials. Nutr Healthy Aging. 4(1):81–93.; https://pubmed.ncbi.nlm.nih.gov/28035345/
Sánchez-Villegas A, Galbete C, Martinez-González MA, et al. The effect of the Mediterranean diet on plasma brain-derived neurotrophic factor (BDNF) levels: the PREDIMED-NAVARRA randomized trial. Nutr Neurosci. 2011;14(5):195–201. https://pubmed.ncbi.nlm.nih.gov/22005283/
Geethanjali A, Lalitha P, Firdhouse JM. Analysis of curcumin content of turmeric samples from various states of India. Int J Pharma Chem Res. 2016;2(1):55–62. https://www.ijpacr.com/files/19-01-16/114619012016.pdf
Miller KB, Hurst WJ, Payne MJ, et al. Impact of alkalization on the antioxidant and flavanol content of commercial cocoa powders. J Agric Food Chem. 2008;56(18):8527–33. https://pubmed.ncbi.nlm.nih.gov/18710243/
Agricultural Research Service, United States Department of Agriculture. Cocoa, dry powder, unsweetened. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=cocoa&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/169593/nutrients. Published April 1, 2019. Accessed June 30, 2022.; https://fdc.nal.usda.gov/fdc-app.html?query=cocoa&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/169593/nutrients
Neshatdoust S, Saunders C, Castle SM, et al. High-flavonoid intake induces cognitive improvements linked to changes in serum brain-derived neurotrophic factor: two randomised, controlled trials. Nutr Healthy Aging. 4(1):81–93.; https://pubmed.ncbi.nlm.nih.gov/28035345/
Sandberg JC, Björck IME, Nilsson AC. Increased plasma brain-derived neurotrophic factor 10.5 h after intake of whole grain rye-based products in healthy subjects. Nutrients. 2018;10(8):E1097. https://pubmed.ncbi.nlm.nih.gov/30115826/
Intlekofer KA, Berchtold NC, Malvaez M, et al. Exercise and sodium butyrate transform a subthreshold learning event into long-term memory via a brain-derived neurotrophic factor-dependent mechanism. Neuropsychopharmacology. 2013;38(10):2027–34. https://pubmed.ncbi.nlm.nih.gov/23615664/
Gravesteijn E, Mensink RP, Plat J. Effects of nutritional interventions on BDNF concentrations in humans: a systematic review. Nutritional Neuroscience. Published online January 10, 2021:1–12.; https://pubmed.ncbi.nlm.nih.gov/33427118/
Marizzoni M, Cattaneo A, Mirabelli P, et al. Short-chain fatty acids and lipopolysaccharide as mediators between gut dysbiosis and amyloid pathology in Alzheimer’s disease. J Alzheimers Dis. 2020;78(2):683–97. https://pubmed.ncbi.nlm.nih.gov/33074224/
Vinarskaya AK, Balaban PM, Roshchin MV, Zuzina AB. Sodium butyrate as a selective cognitive enhancer for weak or impaired memory. Neurobiol Learn Mem. 2021;180:107414. https://pubmed.ncbi.nlm.nih.gov/33610771/
Fernando WMADB, Martins IJ, Morici M, et al. Sodium butyrate reduces brain amyloid-ß levels and improves cognitive memory performance in an Alzheimer’s disease transgenic mouse model at an early disease stage. J Alzheimers Dis. 2020;74(1):91–9. https://pubmed.ncbi.nlm.nih.gov/31958090/
Govindarajan N, Agis-Balboa RC, Walter J, Sananbenesi F, Fischer A. Sodium butyrate improves memory function in an Alzheimer’s disease mouse model when administered at an advanced stage of disease progression. J Alzheimers Dis. 2011;26(1):187–97. https://pubmed.ncbi.nlm.nih.gov/21593570/
Bourassa MW, Alim I, Bultman SJ, Ratan RR. Butyrate, neuroepigenetics and the gut microbiome: can a high fiber diet improve brain health? Neurosci Lett. 2016;625:56–63. https://pubmed.ncbi.nlm.nih.gov/26868600/
Braniste V, Al-Asmakh M, Kowal C, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 2014;6(263):263ra158. https://pubmed.ncbi.nlm.nih.gov/25411471/
Chen T, Kim CY, Kaur A, et al. Dietary fibre-based SCFA mixtures promote both protection and repair of intestinal epithelial barrier function in a Caco-2 cell model. Food Funct. 2017;8(3):1166–73. https://pubmed.ncbi.nlm.nih.gov/28174773/
Braniste V, Al-Asmakh M, Kowal C, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 2014;6(263):263ra158. https://pubmed.ncbi.nlm.nih.gov/25411471/
Bercik P, Park AJ, Sinclair D, et al. The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication. Neurogastroenterol Motil. 2011;23(12):1132–9. https://pubmed.ncbi.nlm.nih.gov/21988661/
Bravo JA, Forsythe P, Chew MV, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A. 2011;108(38):16050–5. https://pubmed.ncbi.nlm.nih.gov/21876150/
Clark KB, Naritoku DK, Smith DC, Browning RA, Jensen RA. Enhanced recognition memory following vagus nerve stimulation in human subjects. Nat Neurosci. 1999;2(1):94–8. https://pubmed.ncbi.nlm.nih.gov/10195186/
Liyanage SI, Vilekar P, Weaver DF. Nutrients in Alzheimer’s disease: the interaction of diet, drugs and disease. Can J Neurol Sci. 2019;46(1):23–34. https://pubmed.ncbi.nlm.nih.gov/30688198/
McGrattan AM, McGuinness B, McKinley MC, et al. Diet and inflammation in cognitive ageing and Alzheimer’s disease. Curr Nutr Rep. 2019;8(2):53–65. https://pubmed.ncbi.nlm.nih.gov/30949921/
David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–63. https://pubmed.ncbi.nlm.nih.gov/24336217/
van Soest APM, Hermes GDA, Berendsen AAM, et al. Associations between pro– and anti-inflammatory gastro-intestinal microbiota, diet, and cognitive functioning in Dutch healthy older adults: the NU-AGE Study. Nutrients. 2020;12(11):E3471. https://pubmed.ncbi.nlm.nih.gov/33198235/
Bruce-Keller AJ, Salbaum JM, Luo M, et al. Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biol Psychiatry. 2015;77(7):607–15. https://pubmed.ncbi.nlm.nih.gov/25173628/
Loeb MB, Molloy DW, Smieja M, et al. A randomized, controlled trial of doxycycline and rifampin for patients with Alzheimer’s disease. J Am Geriatr Soc. 2004;52(3):381–7. https://pubmed.ncbi.nlm.nih.gov/14962152/
Molloy DW, Standish TI, Zhou Q, Guyatt G, The DARAD Study Group. A multicenter, blinded, randomized, factorial controlled trial of doxycycline and rifampin for treatment of Alzheimer’s disease: the DARAD trial. Int J Geriatr Psychiatry. 2013;28(5):463–70. https://pubmed.ncbi.nlm.nih.gov/22718435/
Marx W, Scholey A, Firth J, et al. Prebiotics, probiotics, fermented foods and cognitive outcomes: a meta-analysis of randomized controlled trials. Neurosci Biobehav Rev. 2020;118:472–84. https://pubmed.ncbi.nlm.nih.gov/32860802/
Benton D, Williams C, Brown A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur J Clin Nutr. 2007;61(3):355–61. https://pubmed.ncbi.nlm.nih.gov/17151594/
Den H, Dong X, Chen M, Zou Z. Efficacy of probiotics on cognition, and biomarkers of inflammation and oxidative stress in adults with Alzheimer’s disease or mild cognitive impairment – a meta-analysis of randomized controlled trials. Aging (Albany NY). 2020;12(4):4010–39. https://pubmed.ncbi.nlm.nih.gov/32062613/
Ito K, Romero K. Placebo effect in subjects with cognitive impairment. Int Rev Neurobiol. 2020;153:213–30. https://pubmed.ncbi.nlm.nih.gov/32563289/
Block BR, Albanese SG, Hume AL. Online promotion of “brain health” supplements. Sr Care Pharm. 2021;36(10):489–92. https://pubmed.ncbi.nlm.nih.gov/34593090/
Stoehr GP, Jacobsen E, Jia Y, Snitz BE, Ganguli M. Trends in the use of medications and supplements to treat or prevent dementia: a population-based study. Alzheimer Dis Assoc Disord. 2020;34(2):148–55. https://pubmed.ncbi.nlm.nih.gov/31633558/
Block BR, Albanese SG, Hume AL. Online promotion of “brain health” supplements. Sr Care Pharm. 2021;36(10):489–92. https://pubmed.ncbi.nlm.nih.gov/34593090/
Case 1:17-cv-00124-LLS Document 72. Federal Trade Commission and People of the State of New York v Quincy Bioscience Holding Company, Inc. https://www.ftc.gov/system/files/documents/cases/quincy_bioscience_opinion_and_order.pdf. Published July 14, 2019. Accessed July 10, 2022.; https://www.ftc.gov/system/files/documents/cases/quincy_bioscience_opinion_and_order.pdf
Fair L. Prevagen complaint suggests mindfulness about memory claims. Federal Trade Commission. https://www.ftc.gov/business-guidance/blog/2017/01/prevagen-complaint-suggests-mindfulness-about-memory-claims. Published January 9, 2017. Accessed June 21, 2022.; https://www.ftc.gov/business-guidance/blog/2017/01/prevagen-complaint-suggests-mindfulness-about-memory-claims
Gabriel BA. AARP asks court to declare Prevagen ads misleading. AARP. https://www.aarp.org/politics-society/advocacy/info-2018/overturn-prevagen-decision-fd.html. Published March 21, 2018. Accessed June 21, 2022.; https://www.aarp.org/politics-society/advocacy/info-2018/overturn-prevagen-decision-fd.html
Scott GN. Does Prevagen® help memory loss? Medscape. https://www.medscape.com/viewarticle/860395. Published March 18, 2016. Accessed June 21, 2022.; https://www.medscape.com/viewarticle/860395
Eisner C. Americans took Prevagen for years – as the FDA questioned its safety. Wired. https://www.wired.com/story/prevagen-made-millions-fda-questioned-safety/. Published October 21, 2020. Accessed June 21, 2022.; https://www.wired.com/story/prevagen-made-millions-fda-questioned-safety/
Crawford C, Deuster PA. Be in the know: dietary supplements for cognitive performance. J Spec Oper Med. 2020;20(2):132–5. https://pubmed.ncbi.nlm.nih.gov/32573750/
Crawford C, Boyd C, Avula B, Wang YH, Khan IA, Deuster PA. A public health issue: dietary supplements promoted for brain health and cognitive performance. J Altern Complement Med. 2020;26(4):265–72. https://pubmed.ncbi.nlm.nih.gov/32119795/
Block BR, Albanese SG, Hume AL. Online promotion of “brain health” supplements. Sr Care Pharm. 2021;36(10):489–92. https://pubmed.ncbi.nlm.nih.gov/34593090/
Franke AG, Heinrich I, Lieb K, Fellgiebel A. The use of Ginkgo biloba in healthy elderly. Age (Dordr). 2014;36(1):435–44. https://pubmed.ncbi.nlm.nih.gov/23736956/
Yuan Q, Wang CW, Shi J, Lin ZX. Effects of Ginkgo biloba on dementia: an overview of systematic reviews. J Ethnopharmacol. 2017;195:1–9. https://pubmed.ncbi.nlm.nih.gov/27940086/
Birks J, Grimley Evans J. Ginkgo biloba for cognitive impairment and dementia. Cochrane Database Syst Rev. 2009;(1):CD003120. https://pubmed.ncbi.nlm.nih.gov/17443523/
Wang Y, Yang G, Gong J, et al. Ginseng for Alzheimer’s disease: a systematic review and meta-analysis of randomized controlled trials. Curr Top Med Chem. 2015;16(5):529–36. https://pubmed.ncbi.nlm.nih.gov/26268331/
Shakespeare W. The Tragedy of Hamlet, Prince of Denmark. Act IV, scene 5, line 3053. OpenSourceShakespeare. https://www.opensourceshakespeare.org/views/plays/play_view.php?WorkID=hamlet&Act=4&Scene=5&Scope=scene. Published 1786. Accessed July 1, 2022.; https://www.opensourceshakespeare.org/views/plays/play_view.php?WorkID=hamlet&Act=4&Scene=5&Scope=scene
Perry EK, Pickering AT, Wang WW, Houghton PJ, Perry NS. Medicinal plants and Alzheimer’s disease: from ethnobotany to phytotherapy. J Pharm Pharmacol. 1999;51(5):527–34. https://pubmed.ncbi.nlm.nih.gov/10411211/
Moss M, Cook J, Wesnes K, Duckett P. Aromas of rosemary and lavender essential oils differentially affect cognition and mood in healthy adults. Int J Neurosci. 2003;113(1):15–38. https://pubmed.ncbi.nlm.nih.gov/12690999/
Moss M, Oliver L. Plasma 1,8-cineole correlates with cognitive performance following exposure to rosemary essential oil aroma. Ther Adv Psychopharmacol. 2012;2(3):103–13. https://pubmed.ncbi.nlm.nih.gov/23983963/
Pengelly A, Snow J, Mills SY, Scholey A, Wesnes K, Butler LR. Short-term study on the effects of rosemary on cognitive function in an elderly population. J Med Food. 2012;15(1):10–7. https://pubmed.ncbi.nlm.nih.gov/21877951/
Shinjyo N, Green J. Are sage, rosemary and lemon balm effective interventions in dementia? A narrative review of the clinical evidence. Eur J Integr Med. 2017;15:83–96. https://www.sciencedirect.com/science/article/abs/pii/S187638201730149X
Kennedy DO, Wake G, Savelev S, et al. Modulation of mood and cognitive performance following acute administration of single doses of Melissa officinalis (lemon balm) with human CNS nicotinic and muscarinic receptor-binding properties. Neuropsychopharmacology. 2003;28(10):1871–81. https://pubmed.ncbi.nlm.nih.gov/12062586/
Perry NSL, Menzies R, Hodgson F, et al. A randomised double-blind placebo-controlled pilot trial of a combined extract of sage, rosemary and melissa, traditional herbal medicines, on the enhancement of memory in normal healthy subjects, including influence of age. Phytomedicine. 2018;39:42–8. https://pubmed.ncbi.nlm.nih.gov/29433682/
Moss M, Cook J, Wesnes K, Duckett P. Aromas of rosemary and lavender essential oils differentially affect cognition and mood in healthy adults. Int J Neurosci. 2003;113(1):15–38. https://pubmed.ncbi.nlm.nih.gov/12690999/
Jimbo D, Kimura Y, Taniguchi M, Inoue M, Urakami K. Effect of aromatherapy on patients with Alzheimer’s disease. Psychogeriatrics. 2009;9(4):173–9. https://pubmed.ncbi.nlm.nih.gov/20377818/
Eriksson PS. Neurogenesis and its implications for regeneration in the adult brain. J Rehabil Med. 2003;(41 Suppl):17–9. https://pubmed.ncbi.nlm.nih.gov/12817652/
Eriksson PS, Perfilieva E, Björk-Eriksson T, et al. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4(11):1313–7. https://pubmed.ncbi.nlm.nih.gov/9809557/
Take comfort in human neurogenesis. Nat Med. 1998;4(11):1207. https://pubmed.ncbi.nlm.nih.gov/9809518/
Jimbo D, Kimura Y, Taniguchi M, Inoue M, Urakami K. Effect of aromatherapy on patients with Alzheimer’s disease. Psychogeriatrics. 2009;9(4):173–9. https://pubmed.ncbi.nlm.nih.gov/20377818/
Aydin Yildirim T, Kitis Y. The effect of aromatherapy application on cognitive functions and daytime sleepiness in older adults living in a nursing home. Holist Nurs Pract. 2020;34(2):83–90. https://pubmed.ncbi.nlm.nih.gov/32049695/
Ayaz M, Sadiq A, Junaid M, Ullah F, Subhan F, Ahmed J. Neuroprotective and anti-aging potentials of essential oils from aromatic and medicinal plants. Front Aging Neurosci. 2017;9:168. https://pubmed.ncbi.nlm.nih.gov/28611658/
Zalomonson S, Freud T, Punchik B, Samson T, Lebedinsky S, Press Y. The results of a crossover placebo-controlled study of the effect of lavender oil on behavioral and psychological symptoms of dementia. Rejuvenation Res. 2019;22(3):246–53. https://pubmed.ncbi.nlm.nih.gov/30328781/
Ball EL, Owen-Booth B, Gray A, Shenkin SD, Hewitt J, McCleery J. Aromatherapy for dementia. Cochrane Database Syst Rev. 2020;2020(8):CD003150. https://pubmed.ncbi.nlm.nih.gov/32813272/
Burns A, Perry E, Holmes C, et al. A double-blind placebo-controlled randomized trial of Melissa officinalis oil and donepezil for the treatment of agitation in Alzheimer’s disease. Dement Geriatr Cogn Disord. 2011;31(2):158–64. https://pubmed.ncbi.nlm.nih.gov/21335973/
Watson K, Hatcher D, Good A. A randomised controlled trial of lavender (Lavandula angustifolia) and lemon balm (Melissa officinalis) essential oils for the treatment of agitated behaviour in older people with and without dementia. Complement Ther Med. 2019;42:366–73. https://pubmed.ncbi.nlm.nih.gov/30670268/
Hishikawa N, Takahashi Y, Amakusa Y, et al. Effects of turmeric on Alzheimer’s disease with behavioral and psychological symptoms of dementia. Ayu. 2012;33(4):499–504. https://pubmed.ncbi.nlm.nih.gov/23723666/
Zhu LN, Mei X, Zhang ZG, Xie YP, Lang F. Curcumin intervention for cognitive function in different types of people: a systematic review and meta-analysis. Phytother Res. 2019;33(3):524–33. https://pubmed.ncbi.nlm.nih.gov/30575152/
Baum L, Lam CWK, Cheung SKK, et al. Six-month randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin in patients with Alzheimer disease. J Clin Psychopharmacol. 2008;28(1):110–3. https://pubmed.ncbi.nlm.nih.gov/18204357/
Ringman JM, Frautschy SA, Teng E, et al. Oral curcumin for Alzheimer’s disease: tolerability and efficacy in a 24-week randomized, double blind, placebo-controlled study. Alzheimers Res Ther. 2012;4(5):43. https://pubmed.ncbi.nlm.nih.gov/23107780/
Gupta SC, Sung B, Kim JH, Prasad S, Li S, Aggarwal BB. Multitargeting by turmeric, the golden spice: from kitchen to clinic. Mol Nutr Food Res. 2013;57(9):1510–28. https://pubmed.ncbi.nlm.nih.gov/22887802/
Ahmed T, Gilani AH. Therapeutic potential of turmeric in Alzheimer’s disease: curcumin or curcuminoids? Phytother Res. 2014;28(4):517–25. https://pubmed.ncbi.nlm.nih.gov/23873854/
Hishikawa N, Takahashi Y, Amakusa Y, et al. Effects of turmeric on Alzheimer’s disease with behavioral and psychological symptoms of dementia. Ayu. 2012;33(4):499–504. https://pubmed.ncbi.nlm.nih.gov/23723666/
Moazen-Zadeh E, Abbasi SH, Safi-Aghdam H, et al. Effects of saffron on cognition, anxiety, and depression in patients undergoing coronary artery bypass grafting: a randomized double-blind placebo-controlled trial. J Altern Complement Med. 2018;24(4):361–8. https://pubmed.ncbi.nlm.nih.gov/29185780/
Farokhnia M, Shafiee Sabet M, Iranpour N, et al. Comparing the efficacy and safety of Crocus sativus L. with memantine in patients with moderate to severe Alzheimer’s disease: a double-blind randomized clinical trial. Hum Psychopharmacol. 2014;29(4):351–9. https://pubmed.ncbi.nlm.nih.gov/25163440/
Hausenblas HA, Heekin K, Mutchie HL, Anton S. A systematic review of randomized controlled trials examining the effectiveness of saffron (Crocus sativus L.) on psychological and behavioral outcomes. J Integr Med. 2015;13(4):231–40. https://pubmed.ncbi.nlm.nih.gov/26165367/
Ayati Z, Yang G, Ayati MH, Emami SA, Chang D. Saffron for mild cognitive impairment and dementia: a systematic review and meta-analysis of randomised clinical trials. BMC Complement Med Ther. 2020;20(1):333. https://pubmed.ncbi.nlm.nih.gov/33167948/
Tóth B, Hegyi P, Lantos T, et al. The efficacy of saffron in the treatment of mild to moderate depression: a meta-analysis. Planta Med. 2019;85(1):24–31. https://pubmed.ncbi.nlm.nih.gov/30036891/
Tabeshpour J, Sobhani F, Sadjadi SA, et al. A double-blind, randomized, placebo-controlled trial of saffron stigma (Crocus sativus L.) in mothers suffering from mild-to-moderate postpartum depression. Phytomedicine. 2017;36:145–52. https://pubmed.ncbi.nlm.nih.gov/29157808/
Finley JW, Gao S. A perspective on Crocus sativus L. (saffron) constituent crocin: a potent water-soluble antioxidant and potential therapy for Alzheimer’s disease. J Agric Food Chem. 2017;65(5):1005–20. https://pubmed.ncbi.nlm.nih.gov/28098452/
World Health Organization. WHO Monographs on Selected Medicinal Plants. Vol 3. World Health Organization; 2001. https://apps.who.int/iris/handle/10665/42052
Block BR, Albanese SG, Hume AL. Online promotion of “brain health” supplements. Sr Care Pharm. 2021;36(10):489–92. https://pubmed.ncbi.nlm.nih.gov/34593090/
Goodwill AM, Szoeke C. A systematic review and meta-analysis of the effect of low vitamin D on cognition. J Am Geriatr Soc. 2017;65(10):2161–8. https://pubmed.ncbi.nlm.nih.gov/28758188/
Kalra A, Teixeira AL, Diniz BS. Association of vitamin D levels with incident all-cause dementia in longitudinal observational studies: a systematic review and meta-analysis. J Prev Alzheimers Dis. 2020;7(1):14–20. https://pubmed.ncbi.nlm.nih.gov/32010921/
Orces C, Lorenzo C, Guarneros JE. The prevalence and determinants of vitamin D inadequacy among U.S. older adults: National Health and Nutrition Examination Survey 2007–2014. Cureus. 2019;11(8):e5300. https://pubmed.ncbi.nlm.nih.gov/31579639/
Riaz S, Malcangio M, Miller M, Tomlinson DR. A vitamin D3 derivative (CB1093) induces nerve growth factor and prevents neurotrophic deficits in streptozotocin-diabetic rats. Diabetologia. 1999;42(11):1308–13. https://pubmed.ncbi.nlm.nih.gov/10550414/
Durk MR, Han K, Chow ECY, et al. 1a,25-Dihydroxyvitamin D3 reduces cerebral amyloid-ß accumulation and improves cognition in mouse models of Alzheimer’s disease. J Neurosci. 2014;34(21):7091–101. https://pubmed.ncbi.nlm.nih.gov/24849345/
Hu J, Jia J, Zhang Y, Miao R, Huo X, Ma F. Effects of vitamin D3 supplementation on cognition and blood lipids: a 12-month randomised, double-blind, placebo-controlled trial. J Neurol Neurosurg Psychiatry. 2018;89(12):1341–7. https://pubmed.ncbi.nlm.nih.gov/30279212/
Jia J, Hu J, Huo X, Miao R, Zhang Y, Ma F. Effects of vitamin D supplementation on cognitive function and blood Aß-related biomarkers in older adults with Alzheimer’s disease: a randomised, double-blind, placebo-controlled trial. J Neurol Neurosurg Psychiatry. 2019;90(12):1347–52. https://pubmed.ncbi.nlm.nih.gov/31296588/
Castle M, Fiedler N, Pop LC, et al. Three doses of vitamin D and cognitive outcomes in older women: a double-blind randomized controlled trial. J Gerontol A Biol Sci Med Sci. 2020;75(5):835–42. https://pubmed.ncbi.nlm.nih.gov/30951148/
Schietzel S, Fischer K, Brugger P, et al. Effect of 2000 IU compared with 800 IU vitamin D on cognitive performance among adults age 60 years and older: a randomized controlled trial. Am J Clin Nutr. 2019;110(1):246–53. https://pubmed.ncbi.nlm.nih.gov/31152541/
Pettersen JA. Does high dose vitamin D supplementation enhance cognition?: a randomized trial in healthy adults. Exp Gerontol. 2017;90:90–7. https://pubmed.ncbi.nlm.nih.gov/28167237/
Dysken MW, Sano M, Asthana S, et al. Effect of vitamin E and memantine on functional decline in Alzheimer disease: the TEAM-AD VA Cooperative Randomized Trial. JAMA. 2014;311(1):33–44. https://pubmed.ncbi.nlm.nih.gov/24381967/
Sano M, Ernesto C, Thomas RG, et al. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer’s disease. N Engl J Med. 1997;336(17):1216–22. https://pubmed.ncbi.nlm.nih.gov/9110909/
Lloret A, Badía MC, Mora NJ, Pallardó FV, Alonso MD, Viña J. Vitamin E paradox in Alzheimer’s disease: it does not prevent loss of cognition and may even be detrimental. J Alzheimers Dis. 2009;17(1):143–9. https://pubmed.ncbi.nlm.nih.gov/19494439/
Yaffe K, Clemons TE, McBee WL, Lindblad AS. Impact of antioxidants, zinc, and copper on cognition in the elderly: a randomized, controlled trial. Neurology. 2004;63(9):1705–7. https://pubmed.ncbi.nlm.nih.gov/15534261/
Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of antioxidant vitamin supplementation in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002;360(9326):23–33. https://pubmed.ncbi.nlm.nih.gov/12114036/
Grodstein F, O’Brien J, Kang JH, et al. Long-term multivitamin supplementation and cognitive function in men: a randomized trial. Ann Intern Med. 2013;159(12):806–14. https://pubmed.ncbi.nlm.nih.gov/24490265/
Maylor EA, Simpson EEA, Secker DL, et al. Effects of zinc supplementation on cognitive function in healthy middle-aged and older adults: the ZENITH study. Br J Nutr. 2006;96(4):752–60. https://pubmed.ncbi.nlm.nih.gov/17010236/
Yaffe K, Clemons TE, McBee WL, Lindblad AS. Impact of antioxidants, zinc, and copper on cognition in the elderly: a randomized, controlled trial. Neurology. 2004;63(9):1705–7. https://pubmed.ncbi.nlm.nih.gov/15534261/
Kern J, Kern S, Blennow K, et al. Calcium supplementation and risk of dementia in women with cerebrovascular disease. Neurology. 2016;87(16):1674–80. https://pubmed.ncbi.nlm.nih.gov/27534711/
Obeid R, Herrmann W. Mechanisms of homocysteine neurotoxicity in neurodegenerative diseases with special reference to dementia. FEBS Lett. 2006;580(13):2994–3005. https://pubmed.ncbi.nlm.nih.gov/16697371/
Rutjes AWS, Denton DA, Di Nisio M, et al. Vitamin and mineral supplementation for maintaining cognitive function in cognitively healthy people in mid and late life. Cochrane Database Syst Rev. 2018;12:CD011906. https://pubmed.ncbi.nlm.nih.gov/30556597/
Zhang DM, Ye JX, Mu JS, Cui XP. Efficacy of vitamin B supplementation on cognition in elderly patients with cognitive-related diseases. J Geriatr Psychiatry Neurol. 2017;30(1):50–9. https://pubmed.ncbi.nlm.nih.gov/28248558/
Behrens A, Graessel E, Pendergrass A, Donath C. Vitamin B – Can it prevent cognitive decline? A systematic review and meta-analysis. Syst Rev. 2020;9(1):111. https://pubmed.ncbi.nlm.nih.gov/32414424/
de Jager CA, Oulhaj A, Jacoby R, Refsum H, Smith AD. Cognitive and clinical outcomes of homocysteine-lowering B-vitamin treatment in mild cognitive impairment: a randomized controlled trial. Int J Geriatr Psychiatry. 2012;27(6):592–600. https://pubmed.ncbi.nlm.nih.gov/21780182/
Wyss-Coray T. Ageing, neurodegeneration and brain rejuvenation. Nature. 2016;539(7628):180–6. https://pubmed.ncbi.nlm.nih.gov/27830812/
Smith AD, Smith SM, de Jager CA, et al. Homocysteine-lowering by B vitamins slows the rate of accelerated brain atrophy in mild cognitive impairment: a randomized controlled trial. PLoS One. 2010;5(9):e12244. https://pubmed.ncbi.nlm.nih.gov/20838622/
Douaud G, Refsum H, de Jager CA, et al. Preventing Alzheimer’s disease – related gray matter atrophy by B-vitamin treatment. Proc Natl Acad Sci U S A. 2013;110(23):9523–8. https://pubmed.ncbi.nlm.nih.gov/23690582/
Smith AD, Smith SM, de Jager CA, et al. Homocysteine-lowering by B vitamins slows the rate of accelerated brain atrophy in mild cognitive impairment: a randomized controlled trial. PLoS One. 2010;5(9):e12244. https://pubmed.ncbi.nlm.nih.gov/20838622/
McCaddon A, Miller JW. Assessing the association between homocysteine and cognition: reflections on Bradford Hill, meta-analyses, and causality. Nutr Rev. 2015;73(10):723–35. https://pubmed.ncbi.nlm.nih.gov/26293664/
Smith AD, Refsum H, Bottiglieri T, et al. Homocysteine and dementia: an international consensus statement. J Alzheimers Dis. 62(2):561–70.; https://pubmed.ncbi.nlm.nih.gov/29480200/
Clarke R, Bennett D, Parish S, et al. Effects of homocysteine lowering with B vitamins on cognitive aging: meta-analysis of 11 trials with cognitive data on 22,000 individuals. Am J Clin Nutr. 2014;100(2):657–66. https://pubmed.ncbi.nlm.nih.gov/20937919/
McCaddon A, Miller JW. Assessing the association between homocysteine and cognition: reflections on Bradford Hill, meta-analyses, and causality. Nutr Rev. 2015;73(10):723–35. https://pubmed.ncbi.nlm.nih.gov/26293664/
Aisen PS, Schneider LS, Sano M, et al. High-dose B vitamin supplementation and cognitive decline in Alzheimer disease: a randomized controlled trial. JAMA. 2008;300(15):1774–83. https://pubmed.ncbi.nlm.nih.gov/18854539/
Douaud G, Refsum H, de Jager CA, et al. Preventing Alzheimer’s disease – related gray matter atrophy by B-vitamin treatment. Proc Natl Acad Sci U S A. 2013;110(23):9523–8. https://pubmed.ncbi.nlm.nih.gov/23690582/
Zhang S, Tomata Y, Sugiyama K, Sugawara Y, Tsuji I. Mushroom consumption and incident dementia in elderly Japanese: the Ohsaki Cohort 2006 Study. J Am Geriatr Soc. 2017;65(7). https://pubmed.ncbi.nlm.nih.gov/28295137/
Durga J, van Boxtel MPJ, Schouten EG, et al. Effect of 3-year folic acid supplementation on cognitive function in older adults in the FACIT trial: a randomised, double blind, controlled trial. Lancet. 2007;369(9557):208–16. https://pubmed.ncbi.nlm.nih.gov/17240287/
DeRose DJ, Charles-Marcel ZL, Jamison JM, et al. Vegan diet-based lifestyle program rapidly lowers homocysteine levels. Prev Med. 2000;30(3):225–33. https://pubmed.ncbi.nlm.nih.gov/10684746/
Chandrasekhar C, Kiranmayi VS, Pasupuleti SK, Sarma KV, Sarma PV. Assessment of reference range of serum homocysteine from the post-therapy values of cobalamin and folate deficiency patients. J Assoc Physicians India. 2020;68(9):36–42. https://pubmed.ncbi.nlm.nih.gov/32798344/
Houghton LA, Green TJ, Donovan UM, Gibson RS, Stephen AM, O’Connor DL. Association between dietary fiber intake and the folate status of a group of female adolescents. Am J Clin Nutr. 1997;66(6):1414–21. https://pubmed.ncbi.nlm.nih.gov/9394694/
Guttormsen AB, Schneede J, Fiskerstrand T, Ueland PM, Refsum HM. Plasma concentrations of homocysteine and other aminothiol compounds are related to food intake in healthy human subjects. J Nutr. 1994;124(10):1934–41. https://pubmed.ncbi.nlm.nih.gov/7931702/
Obersby D, Chappell DC, Dunnett A, Tsiami AA. Plasma total homocysteine status of vegetarians compared with omnivores: a systematic review and meta-analysis. Br J Nutr. 2013;109(5):785–94. https://pubmed.ncbi.nlm.nih.gov/23298782/
. Öztürk S, Altieri M, Troisi P. Leonardo Da Vinci and stroke – vegetarian diet as a possible cause. Front Neurol Neurosci. 2010;27:1–10. https://pubmed.ncbi.nlm.nih.gov/20375518/
Crane MG, Register UD, Lukens RH, Gregory R. Cobalamin (CBL) studies on two total vegetarian (vegan) families. Vegetarian Nutrition (United Kingdom). 1998;2(3):87–92. https://agris.fao.org/agris-search/search.do?recordID=GB1997058132
Verghese J, Lipton RB, Katz MJ, et al. Leisure activities and the risk of dementia in the elderly. N Engl J Med. 2003;348(25):2508–16. https://pubmed.ncbi.nlm.nih.gov/12815136/
Cafferata RMT, Hicks B, von Bastian CC. Effectiveness of cognitive stimulation for dementia: a systematic review and meta-analysis. Psychol Bull. 2021;147(5):455–76. https://pubmed.ncbi.nlm.nih.gov/34292011/
Bian X, Wang Y, Zhao X, Zhang Z, Ding C. Does music therapy affect the global cognitive function of patients with dementia? A meta-analysis. NeuroRehabilitation. 2021;48(4):553–62. https://pubmed.ncbi.nlm.nih.gov/33967069/
Moreno-Morales C, Calero R, Moreno-Morales P, Pintado C. Music therapy in the treatment of dementia: a systematic review and meta-analysis. Front Med. 2020;7:160. https://pubmed.ncbi.nlm.nih.gov/32509790/
Lam HL, Li WTV, Laher I, Wong RY. Effects of music therapy on patients with dementia – a systematic review. Geriatrics (Basel). 2020;5(4):E62. https://pubmed.ncbi.nlm.nih.gov/32992767/
Wahl D, Solon-Biet SM, Cogger VC, et al. Aging, lifestyle and dementia. Neurobiol Dis. 2019;130:104481. https://pubmed.ncbi.nlm.nih.gov/31136814/
Liu YH, Gao X, Na M, Kris-Etherton PM, Mitchell DC, Jensen GL. Dietary pattern, diet quality, and dementia: a systematic review and meta-analysis of prospective cohort studies. J Alzheimers Dis. 2020;78(1):151–68. https://pubmed.ncbi.nlm.nih.gov/32955461/
Akbaraly T, Sabia S, Hagger-Johnson G, et al. Does overall diet in midlife predict future aging phenotypes? A cohort study. Am J Med. 2013;126(5):411–9.e3. https://pubmed.ncbi.nlm.nih.gov/23582933/
Risk Reduction of Cognitive Decline and Dementia: WHO Guidelines. World Health Organization; 2019. https://www.who.int/publications/i/item/9789241550543
Hughes TF, Andel R, Small BJ, et al. Midlife fruit and vegetable consumption and risk of dementia in later life in Swedish twins. Am J Geriatr Psychiatry. 2010;18(5):413–20. https://pubmed.ncbi.nlm.nih.gov/19910881/
Jiang X, Huang J, Song D, Deng R, Wei J, Zhang Z. Increased consumption of fruit and vegetables is related to a reduced risk of cognitive impairment and dementia: meta-analysis. Front Aging Neurosci. 2017;9:18. https://pubmed.ncbi.nlm.nih.gov/28223933/
Sherzai D, Sherzai A. Preventing Alzheimer’s: our most urgent health care priority. Am J Lifestyle Med. 2019;13(5):451–61. https://pubmed.ncbi.nlm.nih.gov/31523210/
Millin PM, Rickert GM. Effect of a strawberry and spinach dietary supplement on spatial learning in early and late middle-aged female rats. Antioxidants (Basel). 2018;8(1):E1. https://pubmed.ncbi.nlm.nih.gov/30577447/
Pandey KB, Rizvi SI. Plant polyphenols as dietary antioxidants in human health and disease. Oxid Med Cell Longev. 2009;2(5):270–8. https://pubmed.ncbi.nlm.nih.gov/20716914/
Cherniack EP. A plant-tastic treatment for cognitive disorders. Maturitas. 2012;72(4):265–6. https://pubmed.ncbi.nlm.nih.gov/22658645/
Khoo HE, Azlan A, Tang ST, Lim SM. Anthocyanidins and anthocyanins: colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr Res. 2017;61(1):1361779. https://pubmed.ncbi.nlm.nih.gov/28970777/
Shukitt-Hale B, Bielinski DF, Lau FC, Willis LM, Carey AN, Joseph JA. The beneficial effects of berries on cognition, motor behaviour and neuronal function in ageing. Br J Nutr. 2015;114(10):1542–9. https://pubmed.ncbi.nlm.nih.gov/26392037/
Krikorian R, Shidler MD, Nash TA, et al. Blueberry supplementation improves memory in older adults. J Agric Food Chem. 2010;58(7):3996–4000. https://pubmed.ncbi.nlm.nih.gov/20047325/
Miller MG, Hamilton DA, Joseph JA, Shukitt-Hale B. Dietary blueberry improves cognition among older adults in a randomized, double-blind, placebo-controlled trial. Eur J Nutr. 2018;57(3):1169–80. https://pubmed.ncbi.nlm.nih.gov/28283823/
Krikorian R, Kalt W, McDonald JE, Shidler MD, Summer SS, Stein AL. Cognitive performance in relation to urinary anthocyanins and their flavonoid-based products following blueberry supplementation in older adults at risk for dementia. J Funct Foods. 2020;64:103667. https://www.sciencedirect.com/science/article/pii/S1756464619305912
Barfoot KL, May G, Lamport DJ, Ricketts J, Riddell PM, Williams CM. The effects of acute wild blueberry supplementation on the cognition of 7–10-year-old schoolchildren. Eur J Nutr. 2019;58(7):2911–20. https://pubmed.ncbi.nlm.nih.gov/30327868/
Whyte AR, Schafer G, Williams CM. Cognitive effects following acute wild blueberry supplementation in 7– to 10-year-old children. Eur J Nutr. 2016;55(6):2151–62. https://pubmed.ncbi.nlm.nih.gov/26437830/
Whyte AR, Schafer G, Williams CM. The effect of cognitive demand on performance of an executive function task following wild blueberry supplementation in 7 to 10 years old children. Food Funct. 2017;8(11):4129–38. https://pubmed.ncbi.nlm.nih.gov/29026903/
Barfoot KL, Istas G, Feliciano RP, et al. Effects of daily consumption of wild blueberry on cognition and urinary metabolites in school-aged children: a pilot study. Eur J Nutr. 2021;60(8):4263–78. https://pubmed.ncbi.nlm.nih.gov/34023938/
Whyte AR, Rahman S, Bell L, et al. Improved metabolic function and cognitive performance in middle-aged adults following a single dose of wild blueberry. Eur J Nutr. 2021;60(3):1521–36. https://pubmed.ncbi.nlm.nih.gov/32747995/
Whyte AR, Williams CM. Effects of a single dose of a flavonoid-rich blueberry drink on memory in 8 to 10 y old children. Nutrition. 2015;31(3):531–4. https://pubmed.ncbi.nlm.nih.gov/25701345/
Lorenz M, Jochmann N, von Krosigk A, et al. Addition of milk prevents vascular protective effects of tea. Eur Heart J. 2007;28(2):219–23. https://pubmed.ncbi.nlm.nih.gov/17213230/
Effect of simultaneous consumption of soymilk and coffee on the urinary excretion of isoflavones, chlorogenic acids and metabolites in healthy adults. J Funct Foods. 2015;19:688–99. https://www.sciencedirect.com/science/article/pii/S1756464615004910?via%3Dihub
Serafini M, Bugianesi R, Maiani G, Valtuena S, De Santis S, Crozier A. Plasma antioxidants from chocolate. Nature. 2003;424(6952):1013. https://pubmed.ncbi.nlm.nih.gov/12944955/
Duarte GS, Farah A. Effect of simultaneous consumption of milk and coffee on chlorogenic acids’ bioavailability in humans. J Agric Food Chem. 2011;59(14):7925–31. https://pubmed.ncbi.nlm.nih.gov/21627318/
Serafini M, Testa MF, Villaño D, et al. Antioxidant activity of blueberry fruit is impaired by association with milk. Free Radic Biol Med. 2009;46(6):769–74. https://pubmed.ncbi.nlm.nih.gov/19135520/
Xiao D, Sandhu A, Huang Y, Park E, Edirisinghe I, Burton-Freeman BM. The effect of dietary factors on strawberry anthocyanins oral bioavailability. Food Funct. 2017;8(11):3970–9. https://pubmed.ncbi.nlm.nih.gov/28979957/
Serafini M, Testa MF, Villaño D, et al. Antioxidant activity of blueberry fruit is impaired by association with milk. Free Radic Biol Med. 2009;46(6):769–74. https://pubmed.ncbi.nlm.nih.gov/19135520/
Zhu Y, Sun J, Lu W, et al. Effects of blueberry supplementation on blood pressure: a systematic review and meta-analysis of randomized clinical trials. J Hum Hypertens. 2017;31(3):165–71. https://pubmed.ncbi.nlm.nih.gov/27654329/
Ahles S, Joris PJ, Plat J. Effects of berry anthocyanins on cognitive performance, vascular function and cardiometabolic risk markers: a systematic review of randomized placebo-controlled intervention studies in humans. Int J Mol Sci. 2021;22(12):6482. https://pubmed.ncbi.nlm.nih.gov/34204250/
Barfoot KL, Istas G, Feliciano RP, et al. Effects of daily consumption of wild blueberry on cognition and urinary metabolites in school-aged children: a pilot study. Eur J Nutr. 2021;60(8):4263–78. https://pubmed.ncbi.nlm.nih.gov/34023938/
Ahles S, Joris PJ, Plat J. Effects of berry anthocyanins on cognitive performance, vascular function and cardiometabolic risk markers: a systematic review of randomized placebo-controlled intervention studies in humans. Int J Mol Sci. 2021;22(12):6482. https://pubmed.ncbi.nlm.nih.gov/34204250/
Curtis PJ, Berends L, van der Velpen V, et al. Blueberry anthocyanin intake attenuates the postprandial cardiometabolic effect of an energy-dense food challenge: results from a double blind, randomized controlled trial in metabolic syndrome participants. Clin Nutr. 2022;41(1):165–76. https://pubmed.ncbi.nlm.nih.gov/34883305/
Boespflug EL, Eliassen JC, Dudley JA, et al. Enhanced neural activation with blueberry supplementation in mild cognitive impairment. Nutr Neurosci. 2018;21(4):297–305. https://pubmed.ncbi.nlm.nih.gov/28221821/
McNamara RK, Kalt W, Shidler MD, et al. Cognitive response to fish oil, blueberry, and combined supplementation in older adults with subjective cognitive impairment. Neurobiol Aging. 2018;64:147–56. https://pubmed.ncbi.nlm.nih.gov/29458842/
Newman S, Stygall J, Hirani S, Shaefi S, Maze M. Postoperative cognitive dysfunction after noncardiac surgery: a systematic review. Anesthesiology. 2007;106(3):572–90. https://pubmed.ncbi.nlm.nih.gov/17325517/
Traupe I, Giacalone M, Agrimi J, et al. Postoperative cognitive dysfunction and short-term neuroprotection from blueberries: a pilot study. Minerva Anestesiol. 2018;84(12):1352–60. https://pubmed.ncbi.nlm.nih.gov/29856175/
Brydges CR, Gaeta L. There is no meta-analytic evidence of blueberries improving cognitive performance or mood. Brain Behav Immun. 2020;85:192. https://pubmed.ncbi.nlm.nih.gov/31610217/
Giacalone M, Di Sacco F, Traupe I, Topini R, Forfori F, Giunta F. Antioxidant and neuroprotective properties of blueberry polyphenols: a critical review. Nutr Neurosci. 2011;14(3):119–25. https://pubmed.ncbi.nlm.nih.gov/21756533/
Carey AN, Pintea GI, Van Leuven S, et al. Red raspberry (Rubus ideaus) supplementation mitigates the effects of a high-fat diet on brain and behavior in mice. Nutr Neurosci. 2021;24(6):406–16. https://pubmed.ncbi.nlm.nih.gov/31328696/
Thangthaeng N, Poulose SM, Gomes SM, Miller MG, Bielinski DF, Shukitt-Hale B. Tart cherry supplementation improves working memory, hippocampal inflammation, and autophagy in aged rats. Age (Dordr). 2016;38(5–6):393–404. https://pubmed.ncbi.nlm.nih.gov/27578256/
Kent K, Charlton K, Roodenrys S, et al. Consumption of anthocyanin-rich cherry juice for 12 weeks improves memory and cognition in older adults with mild-to-moderate dementia. Eur J Nutr. 2017;56(1):333–41. https://pubmed.ncbi.nlm.nih.gov/26482148/
Chai SC, Jerusik J, Davis K, Wright RS, Zhang Z. Effect of Montmorency tart cherry juice on cognitive performance in older adults: a randomized controlled trial. Food Funct. 2019;10(7):4423–31. https://pubmed.ncbi.nlm.nih.gov/31287117/
Crews WD Jr, Harrison DW, Griffin ML, et al. A double-blinded, placebo-controlled, randomized trial of the neuropsychologic efficacy of cranberry juice in a sample of cognitively intact older adults: pilot study findings. J Altern Complement Med. 2005;11(2):305–9. https://pubmed.ncbi.nlm.nih.gov/18400709/
Whyte AR, Cheng N, Butler LT, Lamport DJ, Williams CM. Flavonoid-rich mixed berries maintain and improve cognitive function over a 6 h period in young healthy adults. Nutrients. 2019;11(11):2685. https://pubmed.ncbi.nlm.nih.gov/31698695/
Nilsson A, Salo I, Plaza M, Björck I. Effects of a mixed berry beverage on cognitive functions and cardiometabolic risk markers; a randomized cross-over study in healthy older adults. PLoS One. 2017;12(11):e0188173. https://pubmed.ncbi.nlm.nih.gov/29141041/
Ahles S, Joris PJ, Plat J. Effects of berry anthocyanins on cognitive performance, vascular function and cardiometabolic risk markers: a systematic review of randomized placebo-controlled intervention studies in humans. Int J Mol Sci. 2021;22(12):6482. https://pubmed.ncbi.nlm.nih.gov/34204250/
Jennings A, Steves CJ, Macgregor A, Spector T, Cassidy A. Increased habitual flavonoid intake predicts attenuation of cognitive ageing in twins. BMC Med. 2021;19(1):185. https://pubmed.ncbi.nlm.nih.gov/34420522/
Lee S, Kim EY, Shin C. Changes in brain volume associated with vegetable intake in a general population. J Am Coll Nutr. 2019;38(6):506–12. https://pubmed.ncbi.nlm.nih.gov/30897041/
Kang JH, Ascherio A, Grodstein F. Fruit and vegetable consumption and cognitive decline in aging women. Ann Neurol. 2005;57(5):713–20. https://pubmed.ncbi.nlm.nih.gov/15852398/
Morris MC, Evans DA, Tangney CC, Bienias JL, Wilson RS. Associations of vegetable and fruit consumption with age-related cognitive change. Neurology. 2006;67(8):1370–6. https://pubmed.ncbi.nlm.nih.gov/17060562/
Li W, Sun L, Yue L, Li G, Xiao S. The association between eating green vegetables every day and mild cognitive impairment: a community-based cross-sectional study in Shanghai. Neuropsychiatr Dis Treat. 2019;15:3213–8. https://pubmed.ncbi.nlm.nih.gov/31819449/
Morris MC, Wang Y, Barnes LL, Bennett DA, Dawson-Hughes B, Booth SL. Nutrients and bioactives in green leafy vegetables and cognitive decline: prospective study. Neurology. 2018;90(3):e214–22. https://pubmed.ncbi.nlm.nih.gov/29263222/
Kang JH, Ascherio A, Grodstein F. Fruit and vegetable consumption and cognitive decline in aging women. Ann Neurol. 2005;57(5):713–20. https://pubmed.ncbi.nlm.nih.gov/15852398/
Masci A, Mattioli R, Costantino P, et al. Neuroprotective effect of Brassica oleracea sprouts crude juice in a cellular model of Alzheimer’s disease. Oxid Med Cell Longev. 2015;2015:781938. https://pubmed.ncbi.nlm.nih.gov/26180595/
Klomparens EA, Ding Y. The neuroprotective mechanisms and effects of sulforaphane. Brain Circ. 2019;5(2):74–83. https://pubmed.ncbi.nlm.nih.gov/31334360/
Panjwani AA, Liu H, Fahey JW. Crucifers and related vegetables and supplements for neurologic disorders: what is the evidence? Curr Opin Clin Nutr Metab Care. 2018;21(6):451–7. https://pubmed.ncbi.nlm.nih.gov/30199394/
Song L, Thornalley PJ. Effect of storage, processing and cooking on glucosinolate content of Brassica vegetables. Food Chem Toxicol. 2007;45(2):216–24. https://pubmed.ncbi.nlm.nih.gov/17011103/
Nouchi R, Hu Q, Saito T, Kawata NYdS, Nouchi H, Kawashima R. Brain training and sulforaphane intake interventions separately improve cognitive performance in healthy older adults, whereas a combination of these interventions does not have more beneficial effects: evidence from a randomized controlled trial. Nutrients. 2021;13(2):352. https://pubmed.ncbi.nlm.nih.gov/33503851/
Jiraungkoorskul W. Review of neuro-nutrition used as anti-Alzheimer plant, spinach, Spinacia oleracea. Pharmacogn Rev. 2016;10(20):105–8. https://pubmed.ncbi.nlm.nih.gov/28082792/
Stanaway L, Rutherfurd-Markwick K, Page R, Ali A. Performance and health benefits of dietary nitrate supplementation in older adults: a systematic review. Nutrients. 2017;9(11):E1171. https://pubmed.ncbi.nlm.nih.gov/29077028/
Petrie M, Rejeski WJ, Basu S, et al. Beet root juice: an ergogenic aid for exercise and the aging brain. J Gerontol A Biol Sci Med Sci. 2017;72(9):1284–9. https://pubmed.ncbi.nlm.nih.gov/28329785/
Hammond BR, Renzi LM. Carotenoids. Adv Nutr. 2013;4(4):474–6. https://pubmed.ncbi.nlm.nih.gov/23858095/
Booth SL. Vitamin K: food composition and dietary intakes. Food Nutr Res. 2012;56:5505. https://pubmed.ncbi.nlm.nih.gov/22489217/
Tanprasertsuk J, Ferland G, Johnson MA, et al. Concentrations of circulating phylloquinone, but not cerebral menaquinone-4, are positively correlated with a wide range of cognitive measures: exploratory findings in centenarians. J Nutr. 2020;150(1):82–90. https://pubmed.ncbi.nlm.nih.gov/31504672/
Johnson EJ. Role of lutein and zeaxanthin in visual and cognitive function throughout the lifespan. Nutr Rev. 2014;72(9):605–12. https://pubmed.ncbi.nlm.nih.gov/25109868/
Erdman JW, Smith JW, Kuchan MJ, et al. Lutein and brain function. Foods. 2015;4(4):547–64. https://pubmed.ncbi.nlm.nih.gov/26566524/
Johnson EJ. Role of lutein and zeaxanthin in visual and cognitive function throughout the lifespan. Nutr Rev. 2014;72(9):605–12. https://pubmed.ncbi.nlm.nih.gov/25109868/
Kelly D, Coen RF, Akuffo KO, et al. Cognitive function and its relationship with macular pigment optical density and serum concentrations of its constituent carotenoids. J Alzheimers Dis. 2015;48(1):261–77. https://pubmed.ncbi.nlm.nih.gov/26401946/
Vishwanathan R, Schalch W, Johnson EJ. Macular pigment carotenoids in the retina and occipital cortex are related in humans. Nutr Neurosci. 2016;19(3):95–101. https://pubmed.ncbi.nlm.nih.gov/25752849/
Mewborn CM, Terry DP, Renzi-Hammond LM, Hammond BR, Miller LS. Relation of retinal and serum lutein and zeaxanthin to white matter integrity in older adults: a diffusion tensor imaging study. Arch Clin Neuropsychol. 2018;33(7):861–74. https://pubmed.ncbi.nlm.nih.gov/29161349/
Johnson EJ. Role of lutein and zeaxanthin in visual and cognitive function throughout the lifespan. Nutr Rev. 2014;72(9):605–12. https://pubmed.ncbi.nlm.nih.gov/25109868/
Edwards CG, Walk AM, Thompson SV, et al. Effects of 12-week avocado consumption on cognitive function among adults with overweight and obesity. Int J Psychophysiol. 2020;148:13–24. https://pubmed.ncbi.nlm.nih.gov/31846631/
Edwards CG, Walk AM, Thompson SV, et al. Effects of 12-week avocado consumption on cognitive function among adults with overweight and obesity. Int J Psychophysiol. 2020;148:13–24. https://pubmed.ncbi.nlm.nih.gov/31846631/
Hammond BR, Johnson EJ, Russell RM, et al. Dietary modification of human macular pigment density. Invest Ophthalmol Vis Sci. 1997;38(9):1795–801. https://pubmed.ncbi.nlm.nih.gov/9286268/
Bovier ER, Hammond BR. A randomized placebo-controlled study on the effects of lutein and zeaxanthin on visual processing speed in young healthy subjects. Arch Biochem Biophys. 2015;572:54–7. https://pubmed.ncbi.nlm.nih.gov/25483230/
Nouchi R, Suiko T, Kimura E, et al. Effects of lutein and astaxanthin intake on the improvement of cognitive functions among healthy adults: a systematic review of randomized controlled trials. Nutrients. 2020;12(3):E617. https://pubmed.ncbi.nlm.nih.gov/32120794/
Renzi LM, Dengler MJ, Puente A, Miller LS, Hammond BR. Relationships between macular pigment optical density and cognitive function in unimpaired and mildly cognitively impaired older adults. Neurobiol Aging. 2014;35(7):1695–9. https://pubmed.ncbi.nlm.nih.gov/24508218/
Nolan JM, Loskutova E, Howard A, et al. The impact of supplemental macular carotenoids in Alzheimer’s disease: a randomized clinical trial. J Alzheimers Dis. 2015;44(4):1157–69. https://pubmed.ncbi.nlm.nih.gov/25408222/
Saitsu Y, Nishide A, Kikushima K, Shimizu K, Ohnuki K. Improvement of cognitive functions by oral intake of Hericium erinaceus. Biomed Res. 2019;40(4):125–31. https://pubmed.ncbi.nlm.nih.gov/31413233/
Mori K, Inatomi S, Ouchi K, Azumi Y, Tuchida T. Improving effects of the mushroom Yamabushitake (Hericium erinaceus) on mild cognitive impairment: a double-blind placebo-controlled clinical trial. Phytother Res. 2009;23(3):367–72. https://pubmed.ncbi.nlm.nih.gov/18844328/
Rajaram S, Jones J, Lee GJ. Plant-based dietary patterns, plant foods, and age-related cognitive decline. Adv Nutr. 2019;10(Suppl_4):S422–36. https://pubmed.ncbi.nlm.nih.gov/31728502/
Huang Q, Braffett BH, Simmens SJ, Young HA, Ogden CL. Dietary polyphenol intake in us adults and 10-year trends: 2007–2016. J Acad Nutr Diet. 2020;120(11):1821–33. https://pubmed.ncbi.nlm.nih.gov/32807722/
Pham K, Mulugeta A, Zhou A, O’Brien JT, Llewellyn DJ, Hyppönen E. High coffee consumption, brain volume and risk of dementia and stroke. Nutr Neurosci. Published online June 24, 2021:1–12.; https://pubmed.ncbi.nlm.nih.gov/34165394/
Ran LS, Liu WH, Fang YY, et al. Alcohol, coffee and tea intake and the risk of cognitive deficits: a dose-response meta-analysis. Epidemiol Psychiatr Sci. 2021;30:e13. https://pubmed.ncbi.nlm.nih.gov/33568254/
Ran LS, Liu WH, Fang YY, et al. Alcohol, coffee and tea intake and the risk of cognitive deficits: a dose-response meta-analysis. Epidemiol Psychiatr Sci. 2021;30:e13. https://pubmed.ncbi.nlm.nih.gov/33568254/
Einöther SJ, Martens VE. Acute effects of tea consumption on attention and mood. Am J Clin Nutr. 2013;98(6 Suppl):1700S-8S. https://pubmed.ncbi.nlm.nih.gov/24172303/
Liu X, Du X, Han G, Gao W. Association between tea consumption and risk of cognitive disorders: a dose-response meta-analysis of observational studies. Oncotarget. 2017;8(26):43306–21. https://pubmed.ncbi.nlm.nih.gov/28496007/
Li XH, Li CY, Lu JM, Tian RB, Wei J. Allicin ameliorates cognitive deficits ageing-induced learning and memory deficits through enhancing of Nrf2 antioxidant signaling pathways. Neurosci Lett. 2012;514(1):46–50. https://pubmed.ncbi.nlm.nih.gov/22390900/
Luo JF, Dong Y, Chen JY, Lu JH. The effect and underlying mechanisms of garlic extract against cognitive impairment and Alzheimer’s disease: a systematic review and meta-analysis of experimental animal studies. J Ethnopharmacol. 2021;280:114423. https://pubmed.ncbi.nlm.nih.gov/34273446/
Tasnim S, Haque PS, Bari MS, et al. Allium sativum L. improves visual memory and attention in healthy human volunteers. Evid Based Complement Alternat Med. 2015;2015:103416. https://pubmed.ncbi.nlm.nih.gov/26351508/
Saenghong N, Wattanathorn J, Muchimapura S, et al. Zingiber officinale improves cognitive function of the middle-aged healthy women. Evid Based Complement Alternat Med. 2012;2012:383062. https://pubmed.ncbi.nlm.nih.gov/22235230/
Bin Sayeed MS, Shams T, Fahim Hossain S, et al. Nigella sativa L. seeds modulate mood, anxiety and cognition in healthy adolescent males. J Ethnopharmacol. 2014;152(1):156–62. https://pubmed.ncbi.nlm.nih.gov/24412554/
Bin Sayeed MS, Asaduzzaman M, Morshed H, Hossain MM, Kadir MF, Rahman MR. The effect of Nigella sativa Linn. seed on memory, attention and cognition in healthy human volunteers. J Ethnopharmacol. 2013;148(3):780–6. https://pubmed.ncbi.nlm.nih.gov/23707331/
Mazza E, Fava A, Ferro Y, et al. Impact of legumes and plant proteins consumption on cognitive performances in the elderly. J Transl Med. 2017;15(1):109. https://pubmed.ncbi.nlm.nih.gov/28532453/
An R, Liu G, Khan N, Yan H, Wang Y. Dietary habits and cognitive impairment risk among oldest-old Chinese. J Gerontol B Psychol Sci Soc Sci. 2019;74(3):474–83. https://pubmed.ncbi.nlm.nih.gov/28184889/
Cui C, Birru RL, Snitz BE, et al. Effects of soy isoflavones on cognitive function: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev. 2020;78(2):134–44. https://pubmed.ncbi.nlm.nih.gov/31504836/
File SE, Jarrett N, Fluck E, Duffy R, Casey K, Wiseman H. Eating soya improves human memory. Psychopharmacology (Berl). 2001;157(4):430–6. https://pubmed.ncbi.nlm.nih.gov/11605103/
Bhagwat S, Haytowitz DB, Holden JM. USDA database for the isoflavone content of selected foods: release 2.0. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/arsuserfiles/80400525/data/isoflav/isoflav_r2.pdf. Published September 2008. Accessed June 28, 2022.; https://www.ars.usda.gov/arsuserfiles/80400525/data/isoflav/isoflav_r2.pdf
Gleason CE, Fischer BL, Dowling NM, et al. Cognitive effects of soy isoflavones in patients with Alzheimer’s disease. J Alzheimers Dis. 2015;47(4):1009–19. https://pubmed.ncbi.nlm.nih.gov/26401779/
Sekikawa A, Higashiyama A, Lopresti BJ, et al. Associations of equol-producing status with white matter lesion and amyloid-ß deposition in cognitively normal elderly Japanese. Alzheimers Dement (N Y). 2020;6(1):e12089. https://pubmed.ncbi.nlm.nih.gov/33117881/
Yuan JP, Wang JH, Liu X. Metabolism of dietary soy isoflavones to equol by human intestinal microflora – implications for health. Mol Nutr Food Res. 2007;51(7):765–81. https://pubmed.ncbi.nlm.nih.gov/17579894/
Yuan JP, Wang JH, Liu X. Metabolism of dietary soy isoflavones to equol by human intestinal microflora – implications for health. Mol Nutr Food Res. 2007;51(7):765–81. https://pubmed.ncbi.nlm.nih.gov/17579894/
Sugiyama Y, Masumori N, Fukuta F, et al. Influence of isoflavone intake and equol-producing intestinal flora on prostate cancer risk. Asian Pac J Cancer Prev. 2013;14(1):1–4. https://pubmed.ncbi.nlm.nih.gov/23534704/
Setchell KDR, Cole SJ. Method of defining equol-producer status and its frequency among vegetarians. J Nutr. 2006;136(8):2188–93. https://pubmed.ncbi.nlm.nih.gov/16857839/
Rowland IR, Wiseman H, Sanders TA, Adlercreutz H, Bowey EA. Interindividual variation in metabolism of soy isoflavones and lignans: influence of habitual diet on equol production by the gut microflora. Nutr Cancer. 2000;36(1):27–32. https://pubmed.ncbi.nlm.nih.gov/10798213/
Setchell KDR, Brown NM, Summer S, et al. Dietary factors influence production of the soy isoflavone metabolite s-(-)equol in healthy adults. J Nutr. 2013;143(12):1950–8. https://pubmed.ncbi.nlm.nih.gov/24089421/
Setchell KDR, Cole SJ. Method of defining equol-producer status and its frequency among vegetarians. J Nutr. 2006;136(8):2188–93. https://pubmed.ncbi.nlm.nih.gov/16857839/
Foscolou A, D’Cunha NM, Naumovski N, et al. The association between whole grain products consumption and successful aging: a combined analysis of MEDIS and ATTICA epidemiological studies. Nutrients. 2019;11(6):E1221. https://pubmed.ncbi.nlm.nih.gov/31146435/
Shimizu C, Wakita Y, Kihara M, Kobayashi N, Tsuchiya Y, Nabeshima T. Association of lifelong intake of barley diet with healthy aging: changes in physical and cognitive functions and intestinal microbiome in senescence-accelerated mouse-prone 8(SAMP8). Nutrients. 2019;11(8):E1770. https://pubmed.ncbi.nlm.nih.gov/31374892/
Liu X, Guasch-Ferré M, Tobias DK, Li Y. Association of walnut consumption with total and cause-specific mortality and life expectancy in U.S. Adults. Nutrients. 2021;13(8):2699. https://pubmed.ncbi.nlm.nih.gov/34444859/
O’Brien J, Okereke O, Devore E, Rosner B, Breteler M, Grodstein F. Long-term intake of nuts in relation to cognitive function in older women. J Nutr Health Aging. 2014;18(5):496–502. https://pubmed.ncbi.nlm.nih.gov/24886736/
Valls-Pedret C, Sala-Vila A, Serra-Mir M, et al. Mediterranean diet and age-related cognitive decline: a randomized clinical trial. JAMA Intern Med. 2015;175(7):1094. https://pubmed.ncbi.nlm.nih.gov/25961184/
Greenwood CE, Parrott MD. Nutrition as a component of dementia risk reduction strategies. Healthc Manage Forum. 2017;30(1):40–5. https://pubmed.ncbi.nlm.nih.gov/28929899/
Jayedi A, Shab-Bidar S. Fish consumption and the risk of chronic disease: an umbrella review of meta-analyses of prospective cohort studies. Adv Nutr. 2020;11(5):1123–33. https://pubmed.ncbi.nlm.nih.gov/32207773/
Patan MJ, Kennedy DO, Husberg C, et al. Supplementation with oil rich in eicosapentaenoic acid, but not in docosahexaenoic acid, improves global cognitive function in healthy, young adults: results from randomized controlled trials. Am J Clin Nutr. 2021;114(3):914–24. https://pubmed.ncbi.nlm.nih.gov/34113957/
Macaron T, Giudici KV, Bowman GL, et al. Associations of Omega-3 fatty acids with brain morphology and volume in cognitively healthy older adults: a narrative review. Ageing Res Rev. 2021;67:101300. https://pubmed.ncbi.nlm.nih.gov/33607289/
Wennberg M, Tornevi A, Johansson I, Hörnell A, Norberg M, Bergdahl IA. Diet and lifestyle factors associated with fish consumption in men and women: a study of whether gender differences can result in gender-specific confounding. Nutr J. 2012;11:101. https://pubmed.ncbi.nlm.nih.gov/23210480/
Maciel E da S, Sonati JG, Galvão JA, Oetterer M. Fish consumption and lifestyle: a cross-sectional study. Food Sci Technol. 2019;39(suppl 1):141–5. https://www.scielo.br/j/cta/a/5nHP5vk9j8FbcYPZDfMQXvM/?lang=en
Li ZH, Zhong WF, Liu S, et al. Associations of habitual fish oil supplementation with cardiovascular outcomes and all cause mortality: evidence from a large population based cohort study. BMJ. 2020;368:m456. https://pubmed.ncbi.nlm.nih.gov/32131999/
Burckhardt M, Herke M, Wustmann T, Watzke S, Langer G, Fink A. Omega-3 fatty acids for the treatment of dementia. Cochrane Database Syst Rev. 2016;2016(4):CD009002. https://pubmed.ncbi.nlm.nih.gov/27063583/
Peters R, Breitner J, James S, et al. Dementia risk reduction: why haven’t the pharmacological risk reduction trials worked? An in-depth exploration of seven established risk factors. Alzheimers Dement (N Y). 2021;7(1):e12202. https://pubmed.ncbi.nlm.nih.gov/34934803/
Brainard JS, Jimoh OF, Deane KHO, et al. Omega-3, omega-6, and polyunsaturated fat for cognition: systematic review and meta-analysis of randomized trials. J Am Med Dir Assoc. 2020;21(10):1439–50.e21. https://pubmed.ncbi.nlm.nih.gov/32305302/
Piro A, Tagarelli G, Lagonia P, Tagarelli A, Quattrone A. Casimir Funk: his discovery of the vitamins and their deficiency disorders. Ann Nutr Metab. 2010;57(2):85–8. https://pubmed.ncbi.nlm.nih.gov/20805686/
Casmir F. The Journal of State Medicine. Volume XX: 341–368, 1912. The etiology of the deficiency diseases, Beri-beri, polyneuritis in birds, epidemic dropsy, scurvy, experimental scurvy in animals, infantile scurvy, ship beri-beri, pellagra. Nutr Rev. 1975;33(6):176–7. https://pubmed.ncbi.nlm.nih.gov/1095967/
Semba RD. The discovery of the vitamins. Int J Vitam Nutr Res. 2012;82(5):310–5. https://pubmed.ncbi.nlm.nih.gov/23798048/
Holman RT. The slow discovery of the importance of omega 3 essential fatty acids in human health. J Nutr. 1998;128(2 Suppl):427S-33S. https://pubmed.ncbi.nlm.nih.gov/9478042/
Davis BC, Kris-Etherton PM. Achieving optimal essential fatty acid status in vegetarians: current knowledge and practical implications. Am J Clin Nutr. 2003;78(3 Suppl):640S-6S. https://pubmed.ncbi.nlm.nih.gov/12936959/
Andrieu S, Guyonnet S, Coley N, et al. Effect of long-term omega 3 polyunsaturated fatty acid supplementation with or without multidomain intervention on cognitive function in elderly adults with memory complaints (MAPT): a randomised, placebo-controlled trial. Lancet Neurol. 2017;16(5):377–89. https://pubmed.ncbi.nlm.nih.gov/28359749/
Hooper C, Vellas B. Commentary: Fatty acids and Alzheimer’s disease: evidence on cognition and cortical ß-amyloid from secondary analyses of the Multidomain Alzheimer Preventive Trial. J Prev Alzheimers Dis. 2018;5(3):168–70. https://pubmed.ncbi.nlm.nih.gov/29972208/
Witte AV, Kerti L, Hermannstädter HM, et al. Long-chain omega-3 fatty acids improve brain function and structure in older adults. Cereb Cortex. 2014;24(11):3059–68. https://pubmed.ncbi.nlm.nih.gov/23796946/
Zhang YP, Miao R, Li Q, Wu T, Ma F. Effects of DHA supplementation on hippocampal volume and cognitive function in older adults with mild cognitive impairment: a 12-month randomized, double-blind, placebo-controlled trial. J Alzheimers Dis. 2017;55(2):497–507. https://pubmed.ncbi.nlm.nih.gov/27716665/
Sun GY, Simonyi A, Fritsche KL, et al. Docosahexaenoic acid (DHA): An essential nutrient and a nutraceutical for brain health and diseases. Prostaglandins Leukot Essent Fatty Acids. 2018;136:3–13. https://pubmed.ncbi.nlm.nih.gov/28314621/
Muskiet FAJ, Fokkema MR, Schaafsma A, Boersma ER, Crawford MA. Is docosahexaenoic acid (DHA) essential? Lessons from DHA status regulation, our ancient diet, epidemiology and randomized controlled trials. J Nutr. 2004;134(1):183–6. https://pubmed.ncbi.nlm.nih.gov/14704315/
Balachandar R, Soundararajan S, Bagepally BS. Docosahexaenoic acid supplementation in age-related cognitive decline: a systematic review and meta-analysis. Eur J Clin Pharmacol. 2020;76(5):639–48. https://pubmed.ncbi.nlm.nih.gov/32060571/
Lane KE, Wilson M, Hellon TG, Davies IG. Bioavailability and conversion of plant based sources of omega-3 fatty acids – a scoping review to update supplementation options for vegetarians and vegans. Crit Rev Food Sci Nutr. Published online February 12, 2021:1–16.; https://pubmed.ncbi.nlm.nih.gov/33576691/
Davis BC, Kris-Etherton PM. Achieving optimal essential fatty acid status in vegetarians: current knowledge and practical implications. Am J Clin Nutr. 2003;78(3 Suppl):640S-6S. https://pubmed.ncbi.nlm.nih.gov/12936959/
Brainard JS, Jimoh OF, Deane KHO, et al. Omega-3, omega-6, and polyunsaturated fat for cognition: systematic review and meta-analysis of randomized trials. J Am Med Dir Assoc. 2020;21(10):1439–50.e21. https://pubmed.ncbi.nlm.nih.gov/32305302/
Danthiir V, Hosking D, Burns NR, et al. Cognitive performance in older adults is inversely associated with fish consumption but not erythrocyte membrane n-3 fatty acids. J Nutr. 2014;144(3):311–20. https://pubmed.ncbi.nlm.nih.gov/24927114/
Laurin D, Verreault R, Lindsay J, Dewailly E, Holub BJ. Omega-3 fatty acids and risk of cognitive impairment and dementia. J Alzheimers Dis. 2003;5(4):315–22. https://pubmed.ncbi.nlm.nih.gov/14624027/
Foley MM, Seidel I, Sevier J, Wendt J, Kogan M. One man’s swordfish story: the link between Alzheimer’s disease and mercury exposure. Complement Ther Med. 2020;52:102499. https://pubmed.ncbi.nlm.nih.gov/32951747/
Xu L, Zhang W, Liu X, Zhang C, Wang P, Zhao X. Circulatory levels of toxic metals (aluminum, cadmium, mercury, lead) in patients with Alzheimer’s disease: a quantitative meta-analysis and systematic review. J Alzheimers Dis. 2018;62(1):361–72. https://pubmed.ncbi.nlm.nih.gov/32761898/
Srikumar TS, Johansson GK, Ockerman PA, Gustafsson JA, Akesson B. Trace element status in healthy subjects switching from a mixed to a lactovegetarian diet for 12 mo. Am J Clin Nutr. 1992;55(4):885–90. https://pubmed.ncbi.nlm.nih.gov/1550072/
Sherzai D, Sherzai A. Preventing Alzheimer’s: our most urgent health care priority. Am J Lifestyle Med. 2019;13(5):451–61. https://pubmed.ncbi.nlm.nih.gov/31523210/
Morris MC, Brockman J, Schneider JA, et al. Association of seafood consumption, brain mercury level, and APOE e4 status with brain neuropathology in older adults. JAMA. 2016;315(5):489–97. https://pubmed.ncbi.nlm.nih.gov/26836731/
Glynn A, Aune M, Darnerud PO, et al. Determinants of serum concentrations of organochlorine compounds in Swedish pregnant women: a cross-sectional study. Environ Health. 2007;6:2. https://pubmed.ncbi.nlm.nih.gov/17266775/
Bourdon JA, Bazinet TM, Arnason TT, Kimpe LE, Blais JM, White PA. Polychlorinated biphenyls (PCBs) contamination and aryl hydrocarbon receptor (AhR) agonist activity of Omega-3 polyunsaturated fatty acid supplements: implications for daily intake of dioxins and PCBs. Food Chem Toxicol. 2010;48(11):3093–7. https://pubmed.ncbi.nlm.nih.gov/20692313/
Arterburn LM, Oken HA, Bailey Hall E, Hamersley J, Kuratko CN, Hoffman JP. Algal-oil capsules and cooked salmon: nutritionally equivalent sources of docosahexaenoic acid. J Am Diet Assoc. 2008;108(7):1204–9. https://pubmed.ncbi.nlm.nih.gov/17713804/
Greene J, Ashburn SM, Razzouk L, Smith DA. Fish oils, coronary heart disease, and the environment. Am J Public Health. 2013;103(9):1568–76. https://pubmed.ncbi.nlm.nih.gov/23409906/
Cox PA, Sacks OW. Cycad neurotoxins, consumption of flying foxes, and ALS-PDC disease in Guam. Neurology. 2002;58(6):956–9. https://pubmed.ncbi.nlm.nih.gov/11914415/
Banack SA, Cox PA. Biomagnification of cycad neurotoxins in flying foxes: implications for ALS-PDC in Guam. Neurology. 2003;61(3):387–9. https://pubmed.ncbi.nlm.nih.gov/12913204/
Pablo J, Banack SA, Cox PA, et al. Cyanobacterial neurotoxin BMAA in ALS and Alzheimer’s disease. Acta Neurol Scand. 2009;120(4):216–25. https://pubmed.ncbi.nlm.nih.gov/19254284/
Brand LE, Pablo J, Compton A, Hammerschlag N, Mash DC. Cyanobacterial blooms and the occurrence of the neurotoxin beta-N-methylamino-L-alanine (BMAA) in South Florida aquatic food webs. Harmful Algae. 2010;9(6):620–35. https://pubmed.ncbi.nlm.nih.gov/21057660/
Bradley WG, Mash DC. Beyond Guam: the cyanobacteria/BMAA hypothesis of the cause of ALS and other neurodegenerative diseases. Amyotroph Lateral Scler. 2009;10 Suppl 2:7–20. https://pubmed.ncbi.nlm.nih.gov/19929726/
Cox PA, Davis DA, Mash DC, Metcalf JS, Banack SA. Dietary exposure to an environmental toxin triggers neurofibrillary tangles and amyloid deposits in the brain. Proc R Soc B. 2016;283(1823):20152397. https://pubmed.ncbi.nlm.nih.gov/26791617/
Meneely JP, Chevallier OP, Graham S, Greer B, Green BD, Elliott CT. ß-methylamino-L-alanine (BMAA) is not found in the brains of patients with confirmed Alzheimer’s disease. Sci Rep. 2016;6:36363. https://pubmed.ncbi.nlm.nih.gov/27821863/
Banack SA, Murch SJ. Methods for the chemical analysis of ß-N-Methylamino-L-alanine: what is known and what remains to be determined. Neurotox Res. 2018;33(1):184–91. https://pubmed.ncbi.nlm.nih.gov/28474174/
Mondo K, Hammerschlag N, Basile M, Pablo J, Banack SA, Mash DC. Cyanobacterial neurotoxin ß-N-methylamino-L-alanine (BMAA) in shark fins. Mar Drugs. 2012;10(2):509–20. https://pubmed.ncbi.nlm.nih.gov/22412816/
Torbick N, Ziniti B, Stommel E, et al. Assessing cyanobacterial harmful algal blooms as risk factors for amyotrophic lateral sclerosis. Neurotox Res. 2018;33(1):199–212. https://pubmed.ncbi.nlm.nih.gov/28470570/
Holtcamp W. Shark fin consumption may expose people to neurotoxic BMAA. Environ Health Perspect. 2012;120(5):a191. https://pubmed.ncbi.nlm.nih.gov/22548869/
Lance E, Arnich N, Maignien T, Biré R. Occurrence of ß-N-methylamino-L-alanine (BMAA) and isomers in aquatic environments and aquatic food sources for humans. Toxins (Basel). 2018;10(2):E83. https://pubmed.ncbi.nlm.nih.gov/29443939/
Mondo K, Broc Glover W, Murch SJ, et al. Environmental neurotoxins ß-N-methylamino-L-alanine (BMAA) and mercury in shark cartilage dietary supplements. Food Chem Toxicol. 2014;70:26–32. https://pubmed.ncbi.nlm.nih.gov/24755394/
Roy-Lachapelle A, Solliec M, Bouchard MF, Sauvé S. Detection of cyanotoxins in algae dietary supplements. Toxins (Basel). 2017;9(3):E76. https://pubmed.ncbi.nlm.nih.gov/28245621/
Glover WB, Baker TC, Murch SJ, Brown P. Determination of ß-N-methylamino-L-alanine, N-(2-aminoethyl)glycine, and 2,4-diaminobutyric acid in food products containing cyanobacteria by ultra-performance liquid chromatography and tandem mass spectrometry: single-laboratory validation. J AOAC Int. 2015;98(6):1559–65. https://pubmed.ncbi.nlm.nih.gov/26651568/
Lehtisalo J, Levälahti E, Lindström J, et al. Dietary changes and cognition over 2 years within a multidomain intervention trial – The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER). Alzheimers Dement. 2019;15(3):410–7. https://pubmed.ncbi.nlm.nih.gov/30527596/
Ngandu T, Lehtisalo J, Solomon A, et al. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): a randomised controlled trial. Lancet. 2015;385(9984):2255–63. https://pubmed.ncbi.nlm.nih.gov/25771249/
Montero-Odasso M, Ismail Z, Livingston G. One third of dementia cases can be prevented within the next 25¿years by tackling risk factors. The case “for” and “against.” Alzheimers Res Ther. 2020;12:81. https://pubmed.ncbi.nlm.nih.gov/32641088/
Lehtisalo J, Levälahti E, Lindström J, et al. Dietary changes and cognition over 2 years within a multidomain intervention trial – The Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER). Alzheimers Dement. 2019;15(3):410–7. https://pubmed.ncbi.nlm.nih.gov/30527596/
Knight A, Bryan J, Murphy K. The Mediterranean diet and age-related cognitive functioning: a systematic review of study findings and neuropsychological assessment methodology. Nutr Neurosci. 2017;20(8):449–68. https://pubmed.ncbi.nlm.nih.gov/27192034/
Coelho-Júnior HJ, Trichopoulou A, Panza F. Cross-sectional and longitudinal associations between adherence to Mediterranean diet with physical performance and cognitive function in older adults: a systematic review and meta-analysis. Ageing Res Rev. 2021;70:101395. https://pubmed.ncbi.nlm.nih.gov/34153553/
Radd-Vagenas S, Duffy SL, Naismith SL, Brew BJ, Flood VM, Fiatarone Singh MA. Effect of the Mediterranean diet on cognition and brain morphology and function: a systematic review of randomized controlled trials. Am J Clin Nutr. 2018;107(3):389–404. https://pubmed.ncbi.nlm.nih.gov/29566197/
Marseglia A, Xu W, Fratiglioni L, et al. Effect of the NU-AGE diet on cognitive functioning in older adults: a randomized controlled trial. Front Physiol. 2018;9:349. https://pubmed.ncbi.nlm.nih.gov/29670545/
Roberts RO, Geda YE, Cerhan JR, et al. Vegetables, unsaturated fats, moderate alcohol intake, and mild cognitive impairment. Dement Geriatr Cogn Disord. 2010;29(5):413–23. https://pubmed.ncbi.nlm.nih.gov/20502015/
Titova OE, Ax E, Brooks SJ, et al. Mediterranean diet habits in older individuals: associations with cognitive functioning and brain volumes. Exp Gerontol. 2013;48(12):1443–8. https://pubmed.ncbi.nlm.nih.gov/24126083/
Morris MC, Tangney CC, Wang Y, Sacks FM, Bennett DA, Aggarwal NT. MIND diet associated with reduced incidence of Alzheimer’s disease. Alzheimers Dement. 2015;11(9):1007–14. https://pubmed.ncbi.nlm.nih.gov/25681666/
Marcason W. What are the components to the MIND diet? J Acad Nutr Diet. 2015;115(10):1744. https://pubmed.ncbi.nlm.nih.gov/26407649/
Kheirouri S, Alizadeh M. MIND diet and cognitive performance in older adults: a systematic review. Crit Rev Food Sci Nutr. 2022;62(29):8059–77. https://pubmed.ncbi.nlm.nih.gov/33989093/
Morris MC, Tangney CC, Wang Y, et al. MIND diet slows cognitive decline with aging. Alzheimers Dement. 2015;11(9):1015–22. https://pubmed.ncbi.nlm.nih.gov/26086182/
Corley J. Adherence to the MIND diet is associated with 12-year all-cause mortality in older adults. Public Health Nutr. Published online September 3, 2020:1–10.; https://pubmed.ncbi.nlm.nih.gov/32878656/
Arjmand G, Abbas-Zadeh M, Eftekhari MH. Effect of MIND diet intervention on cognitive performance and brain structure in healthy obese women: a randomized controlled trial. Sci Rep. 2022;12(1):2871. https://pubmed.ncbi.nlm.nih.gov/35190536/
Berendsen AM, Kang JH, Feskens EJM, de Groot CPGM, Grodstein F, van de Rest O. Association of long-term adherence to the mind diet with cognitive function and cognitive decline in American women. J Nutr Health Aging. 2018;22(2):222–9. https://pubmed.ncbi.nlm.nih.gov/29380849/
Titova OE, Ax E, Brooks SJ, et al. Mediterranean diet habits in older individuals: associations with cognitive functioning and brain volumes. Exp Gerontol. 2013;48(12):1443–8. https://pubmed.ncbi.nlm.nih.gov/24126083/
Giem P, Beeson WL, Fraser GE. The incidence of dementia and intake of animal products: preliminary findings from the Adventist Health Study. Neuroepidemiology. 1993;12(1):28–36. https://pubmed.ncbi.nlm.nih.gov/8327020/
Morris MC, Tangney CC. Dietary fat composition and dementia risk. Neurobiol Aging. 2014;35 Suppl 2:S59–64. https://pubmed.ncbi.nlm.nih.gov/24970568/
Takechi R, Galloway S, Pallebage-Gamarallage MM, Lam V, Dhaliwal SS, Mamo JC. Probucol prevents blood – brain barrier dysfunction in wild-type mice induced by saturated fat or cholesterol feeding. Clin Exp Pharmacol Physiol. 2013;40(1):45–52. https://pubmed.ncbi.nlm.nih.gov/23167559/
Ortolá R, Struijk EA, García-Esquinas E, Rodríguez-Artalejo F, Lopez-Garcia E. Changes in dietary intake of animal and vegetable protein and unhealthy aging. Am J Med. 2020;133(2):231–9. https://pubmed.ncbi.nlm.nih.gov/33839765/
Szczechowiak K, Diniz BS, Leszek J. Diet and Alzheimer’s dementia – nutritional approach to modulate inflammation. Pharmacol Biochem Behav. 2019;184:172743. https://pubmed.ncbi.nlm.nih.gov/31356838/
Wu J, Song X, Chen GC, et al. Dietary pattern in midlife and cognitive impairment in late life: a prospective study in Chinese adults. Am J Clin Nutr. 2019;110(4):912–20. https://pubmed.ncbi.nlm.nih.gov/31374567/
Kheirouri S, Alizadeh M. MIND diet and cognitive performance in older adults: a systematic review. Crit Rev Food Sci Nutr. Published online May 14, 2021:1–19.; https://pubmed.ncbi.nlm.nih.gov/33989093/
Pistollato F, Battino M. Role of plant-based diets in the prevention and regression of metabolic syndrome and neurodegenerative diseases. Trends Food Sci Technol. 2014;40(1):62–81. https://www.sciencedirect.com/science/article/abs/pii/S0924224414001642
Sherchan P, Miles F, Orlich M, et al. Effects of lifestyle factors on cognitive resilience: commentary on “What this sunny, religious town in California teaches us about living longer.” Transl Stroke Res. 2020;11:161–4. https://pubmed.ncbi.nlm.nih.gov/32062815/
Sherzai D, Sherzai A. Preventing Alzheimer’s: our most urgent health care priority. Am J Lifestyle Med. 2019;13(5):451–61. https://pubmed.ncbi.nlm.nih.gov/31523210/
Plassman BL, Williams JW, Burke JR, Holsinger T, Benjamin S. Systematic review: factors associated with risk for and possible prevention of cognitive decline in later life. Ann Intern Med. 2010;153(3):182–93. https://pubmed.ncbi.nlm.nih.gov/20547887/
Daviglus ML, Plassman BL, Pirzada A, et al. Risk factors and preventive interventions for Alzheimer disease: state of the science. Arch Neurol. 2011;68(9):1185–90. https://pubmed.ncbi.nlm.nih.gov/21555601/
Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA. 1998;279(15):1200–5. https://pubmed.ncbi.nlm.nih.gov/9555760/
Friedland RP, Nandi S. A modest proposal for a longitudinal study of dementia prevention (with apologies to Jonathan Swift, 1729). J Alzheimers Dis. 2013;33(2):313–5. https://pubmed.ncbi.nlm.nih.gov/22986779/
Friedland RP, Nandi S. A modest proposal for a longitudinal study of dementia prevention (with apologies to Jonathan Swift, 1729). J Alzheimers Dis. 2013;33(2):313–5. https://pubmed.ncbi.nlm.nih.gov/22986779/
Preventive Medicine Research Institute. Lifestyle intervention for early Alzheimer’s disease. Identifier: NCT04606420. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT04606420. Published October 28, 2020. Accessed June 24, 2022.; https://clinicaltrials.gov/ct2/show/NCT04606420
Dean Ornish, Personal Communication.
Faulkner JA, Larkin LM, Claflin DR, Brooks SV. Age-related changes in the structure and function of skeletal muscles. Clin Exp Pharmacol Physiol. 2007;34(11):1091–6. https://pubmed.ncbi.nlm.nih.gov/17880359/
Garatachea N, Pareja-Galeano H, Sanchis-Gomar F, et al. Exercise attenuates the major hallmarks of aging. Rejuvenation Res. 2015;18(1):57–89. https://pubmed.ncbi.nlm.nih.gov/25431878/
Fougère B, van Kan GA, Vellas B, Cesari M. Redox systems, antioxidants and sarcopenia. Curr Protein Pept Sci. 2018;19(7):643–8. https://pubmed.ncbi.nlm.nih.gov/28317484/
Faulkner JA, Larkin LM, Claflin DR, Brooks SV. Age-related changes in the structure and function of skeletal muscles. Clin Exp Pharmacol Physiol. 2007;34(11):1091–6. https://pubmed.ncbi.nlm.nih.gov/17880359/
Scott D, Blizzard L, Fell J, Jones G. The epidemiology of sarcopenia in community living older adults: what role does lifestyle play? J Cachexia Sarcopenia Muscle. 2011;2(3):125–34. https://pubmed.ncbi.nlm.nih.gov/21966639/
Physical activity: PA-1 Reduce the proportion of adults who engage in no leisure-time physical activity. HealthyPeople.gov. https://www.healthypeople.gov/2020/data-search/Search-the-Data?nid=5052. Accessed August 12, 2022.; https://www.healthypeople.gov/2020/data-search/Search-the-Data?nid=5052
Faulkner JA, Larkin LM, Claflin DR, Brooks SV. Age-related changes in the structure and function of skeletal muscles. Clin Exp Pharmacol Physiol. 2007;34(11):1091–6. https://pubmed.ncbi.nlm.nih.gov/17880359/
Fougère B, van Kan GA, Vellas B, Cesari M. Redox systems, antioxidants and sarcopenia. Curr Protein Pept Sci. 2018;19(7):643–8. https://pubmed.ncbi.nlm.nih.gov/28317484/
Garatachea N, Pareja-Galeano H, Sanchis-Gomar F, et al. Exercise attenuates the major hallmarks of aging. Rejuvenation Res. 2015;18(1):57–89. https://pubmed.ncbi.nlm.nih.gov/25431878/
Xia L, Zhao R, Wan Q, et al. Sarcopenia and adverse health-related outcomes: an umbrella review of meta-analyses of observational studies. Cancer Med. 2020;9(21):7964–78. https://pubmed.ncbi.nlm.nih.gov/32924316/
Li R, Xia J, Zhang X, et al. Associations of muscle mass and strength with all-cause mortality among US older adults. Med Sci Sports Exerc. 2018;50(3):458–67. https://pubmed.ncbi.nlm.nih.gov/28991040/
Newman AB, Kupelian V, Visser M, et al. Strength, but not muscle mass, is associated with mortality in the health, aging and body composition study cohort. J Gerontol A Biol Sci Med Sci. 2006;61(1):72–7. https://pubmed.ncbi.nlm.nih.gov/16456196/
Talar K, Hernández-Belmonte A, Vetrovsky T, Steffl M, Kalamacka E, Courel-Ibáñez J. Benefits of resistance training in early and late stages of frailty and sarcopenia: a systematic review and meta-analysis of randomized controlled studies. J Clin Med. 2021;10(8):1630. https://pubmed.ncbi.nlm.nih.gov/33921356/
Rantanen T, Guralnik JM, Foley D, et al. Midlife hand grip strength as a predictor of old age disability. JAMA. 1999;281(6):558–60. https://pubmed.ncbi.nlm.nih.gov/10022113/
The Lancet. Bringing frailty into all realms of medicine. Lancet. 2019;394(10206):1298. https://pubmed.ncbi.nlm.nih.gov/31609212/
Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci. 2001;56(3):M146–57. https://pubmed.ncbi.nlm.nih.gov/11253156/
Pansarasa O, Pistono C, Davin A, et al. Altered immune system in frailty: genetics and diet may influence inflammation. Ageing Res Rev. 2019;54:100935. https://pubmed.ncbi.nlm.nih.gov/31326616/
Garatachea N, Pareja-Galeano H, Sanchis-Gomar F, et al. Exercise attenuates the major hallmarks of aging. Rejuvenation Res. 2015;18(1):57–89. https://pubmed.ncbi.nlm.nih.gov/25431878/
Rodríguez-Mañas L. Determinants of frailty and longevity: are they the same ones? Nestle Nutr Inst Workshop Ser. 2015;83:29–39. https://pubmed.ncbi.nlm.nih.gov/26485702/
Scott D, Blizzard L, Fell J, Jones G. The epidemiology of sarcopenia in community living older adults: what role does lifestyle play? J Cachexia Sarcopenia Muscle. 2011;2(3):125–34. https://pubmed.ncbi.nlm.nih.gov/21966639/
Frontera WR, Hughes VA, Fielding RA, Fiatarone MA, Evans WJ, Roubenoff R. Aging of skeletal muscle: a 12-yr longitudinal study. J Appl Physiol (1985). 2000;88(4):1321–6. https://pubmed.ncbi.nlm.nih.gov/10749826/
Shimokata H, Ando F, Yuki A, Otsuka R. Age-related changes in skeletal muscle mass among community-dwelling Japanese: a 12-year longitudinal study. Geriatr Gerontol Int. 2014;14 Suppl 1:85–92. https://pubmed.ncbi.nlm.nih.gov/24450565/
Moore SA, Hrisos N, Errington L, et al. Exercise as a treatment for sarcopenia: an umbrella review of systematic review evidence. Physiotherapy. 2020;107:189–201. https://pubmed.ncbi.nlm.nih.gov/32026819/
Paproski JJ, Finello GC, Murillo A, Mandel E. The importance of protein intake and strength exercises for older adults. JAAPA. 2019;32(11):32–6. https://pubmed.ncbi.nlm.nih.gov/31663893/
Moore SA, Hrisos N, Errington L, et al. Exercise as a treatment for sarcopenia: an umbrella review of systematic review evidence. Physiotherapy. 2020;107:189–201. https://pubmed.ncbi.nlm.nih.gov/32026819/
Talar K, Hernández-Belmonte A, Vetrovsky T, Steffl M, Kalamacka E, Courel-Ibáñez J. Benefits of resistance training in early and late stages of frailty and sarcopenia: a systematic review and meta-analysis of randomized controlled studies. J Clin Med. 2021;10(8):1630. https://pubmed.ncbi.nlm.nih.gov/33921356/
Leenders M, Verdijk LB, van der Hoeven L, van Kranenburg J, Nilwik R, van Loon LJC. Elderly men and women benefit equally from prolonged resistance-type exercise training. J Gerontol A Biol Sci Med Sci. 2013;68(7):769–79. https://pubmed.ncbi.nlm.nih.gov/23223011/
Buatois S, Miljkovic D, Manckoundia P, et al. Five times sit to stand test is a predictor of recurrent falls in healthy community-living subjects aged 65 and older. J Am Geriatr Soc. 2008;56(8):1575–7. https://pubmed.ncbi.nlm.nih.gov/18808608/
Kidd T, Mold F, Jones C, et al. What are the most effective interventions to improve physical performance in pre-frail and frail adults? A systematic review of randomised control trials. BMC Geriatr. 2019;19(1):184. https://pubmed.ncbi.nlm.nih.gov/31291884/
Tarazona-Santabalbina FJ, Gómez-Cabrera MC, Pérez-Ros P, et al. A multicomponent exercise intervention that reverses frailty and improves cognition, emotion, and social networking in the community-dwelling frail elderly: a randomized clinical trial. J Am Med Dir Assoc. 2016;17(5):426–33. https://pubmed.ncbi.nlm.nih.gov/26947059/
Paddon-Jones D, Leidy H. Dietary protein and muscle in older persons. Curr Opin Clin Nutr Metab Care. 2014;17(1):5–11. https://pubmed.ncbi.nlm.nih.gov/24310053/
Paddon-Jones D, Sheffield-Moore M, Urban RJ, et al. Essential amino acid and carbohydrate supplementation ameliorates muscle protein loss in humans during 28 days bedrest. J Clin Endocrinol Metab. 2004;89(9):4351–8. https://pubmed.ncbi.nlm.nih.gov/15356032/
Hvid LG, Suetta C, Nielsen JH, et al. Aging impairs the recovery in mechanical muscle function following 4 days of disuse. Exp Gerontol. 2014;52:1–8. https://pubmed.ncbi.nlm.nih.gov/24447828/
Lafont C, Gérard S, Voisin T, et al. Reducing “iatrogenic disability” in the hospitalized frail elderly. J Nutr Health Aging. 2011;15(8):645–60. https://pubmed.ncbi.nlm.nih.gov/21968859/
Breen L, Stokes KA, Churchward-Venne TA, et al. Two weeks of reduced activity decreases leg lean mass and induces “anabolic resistance” of myofibrillar protein synthesis in healthy elderly. J Clin Endocrinol Metab. 2013;98(6):2604–12. https://pubmed.ncbi.nlm.nih.gov/23589526/
Breen L, Stokes KA, Churchward-Venne TA, et al. Two weeks of reduced activity decreases leg lean mass and induces “anabolic resistance” of myofibrillar protein synthesis in healthy elderly. J Clin Endocrinol Metab. 2013;98(6):2604–12. https://pubmed.ncbi.nlm.nih.gov/23589526/
Berardi E, Madaro L, Lozanoska-Ochser B, et al. A pound of flesh: what cachexia is and what it is not. Diagnostics (Basel). 2021;11(1):116. https://pubmed.ncbi.nlm.nih.gov/33445790/
Bano G, Trevisan C, Carraro S, et al. Inflammation and sarcopenia: a systematic review and meta-analysis. Maturitas. 2017;96:10–5. https://pubmed.ncbi.nlm.nih.gov/28041587/
Marcos-Pérez D, Sánchez-Flores M, Proietti S, et al. Association of inflammatory mediators with frailty status in older adults: results from a systematic review and meta-analysis. Geroscience. 2020;42(6):1451–73. https://pubmed.ncbi.nlm.nih.gov/32803650/
Tuttle CSL, Thang LAN, Maier AB. Markers of inflammation and their association with muscle strength and mass: a systematic review and meta-analysis. Ageing Res Rev. 2020;64:101185. https://pubmed.ncbi.nlm.nih.gov/32992047/
Pansarasa O, Pistono C, Davin A, et al. Altered immune system in frailty: genetics and diet may influence inflammation. Ageing Res Rev. 2019;54:100935. https://pubmed.ncbi.nlm.nih.gov/31326616/
Bagheri A, Soltani S, Hashemi R, Heshmat R, Motlagh AD, Esmaillzadeh A. Inflammatory potential of the diet and risk of sarcopenia and its components. Nutr J. 2020;19(1):129. https://pubmed.ncbi.nlm.nih.gov/33248463/
Geng J, Deng L, Qiu S, et al. Dietary inflammatory potential and risk of sarcopenia: data from National Health and Nutrition Examination Surveys. Aging (Albany NY). 2020;13(2):1913–28. https://pubmed.ncbi.nlm.nih.gov/33318308/
Laclaustra M, Rodriguez-Artalejo F, Guallar-Castillon P, et al. The inflammatory potential of diet is related to incident frailty and slow walking in older adults. Clin Nutr. 2020;39(1):185–91. https://pubmed.ncbi.nlm.nih.gov/30737049/
Kim D, Park Y. Association between the dietary inflammatory index and risk of frailty in older individuals with poor nutritional status. Nutrients. 2018;10(10):E1363. https://pubmed.ncbi.nlm.nih.gov/30249038/
Laclaustra M, Rodriguez-Artalejo F, Guallar-Castillon P, et al. The inflammatory potential of diet is related to incident frailty and slow walking in older adults. Clin Nutr. 2020;39(1):185–91. https://pubmed.ncbi.nlm.nih.gov/30737049/
Shivappa N, Steck SE, Hurley TG, Hussey JR, Hébert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. https://pubmed.ncbi.nlm.nih.gov/23941862/
Montiel-Rojas D, Santoro A, Nilsson A, et al. Beneficial role of replacing dietary saturated fatty acids with polyunsaturated fatty acids in the prevention of sarcopenia: findings from the NU-AGE cohort. Nutrients. 2020;12(10):E3079. https://pubmed.ncbi.nlm.nih.gov/32295007/
Stoody EE, Obbagy J, Pannucci TR, et al. Dietary Guidelines for Americans 2020–2025: make every bite count with the dietary guidelines. Dietary Guidelines for Americans. https://www.dietaryguidelines.gov/sites/default/files/2020–12/Dietary_Guidelines_for_Americans_2020–2025.pdf. Published December 2020. Accessed August 4, 2022.; https://www.dietaryguidelines.gov/sites/default/files/2020-12/Dietary_Guidelines_for_Americans_2020-2025.pdf
Saturated fat. American Heart Association. https://www.heart.org/en/healthy-living/healthy-eating/eat-smart/fats/saturated-fats. Updated November 1, 2021. Accessed August 12, 2022.; https://www.heart.org/en/healthy-living/healthy-eating/eat-smart/fats/saturated-fats
Welch AA, MacGregor AJ, Minihane AM, et al. Dietary fat and fatty acid profile are associated with indices of skeletal muscle mass in women aged 18–79 years. J Nutr. 2014;144(3):327–34. https://pubmed.ncbi.nlm.nih.gov/24401817/
Kephart WC, Pledge CD, Roberson PA, et al. The three-month effects of a ketogenic diet on body composition, blood parameters, and performance metrics in CrossFit trainees: a pilot study. Sports (Basel). 2018;6(1):1. https://pubmed.ncbi.nlm.nih.gov/29910305/
Hall KD, Chen KY, Guo J, et al. Energy expenditure and body composition changes after an isocaloric ketogenic diet in overweight and obese men. Am J Clin Nutr. 2016;104(2):324–33. https://pubmed.ncbi.nlm.nih.gov/27385608/
Carpenter KJ. The history of enthusiasm for protein. J Nutr. 1986;116(7):1364–70. https://pubmed.ncbi.nlm.nih.gov/3528432/
McLaren DS. The great protein fiasco. Lancet. 1974;2(7872):93–6. https://pubmed.ncbi.nlm.nih.gov/4137270/
Carpenter KJ. The history of enthusiasm for protein. J Nutr. 1986;116(7):1364–70. https://pubmed.ncbi.nlm.nih.gov/3528432/
McLaren DS. The great protein fiasco revisited. Nutrition. 2000;16(6):464–5. https://pubmed.ncbi.nlm.nih.gov/10869908/
Carpenter KJ. Protein requirements of adults from an evolutionary perspective. Am J Clin Nutr. 1992;55(5):913–7. https://pubmed.ncbi.nlm.nih.gov/1570797/
Speth JD. The Paleoanthropology and Archaeology of Big-Game Hunting: Protein, Fat, or Politics? Springer; 2010. https://link.springer.com/chapter/10.1007/978-1-4419-6733-6_12
Davis TA, Nguyen HV, Garcia-Bravo R, et al. Amino acid composition of human milk is not unique. J Nutr. 1994;124(7):1126–32. https://pubmed.ncbi.nlm.nih.gov/8027865/
Ziegler EE. Adverse effects of cow’s milk in infants. Nestle Nutr Workshop Ser Pediatr Program. 2007;60:185–99. https://pubmed.ncbi.nlm.nih.gov/17664905/
Pedersen AN, Cederholm T. Health effects of protein intake in healthy elderly populations: a systematic literature review. Food Nutr Res. 2014;58:10.3402/fnr.v58.23364. https://pubmed.ncbi.nlm.nih.gov/24624051/
Berryman CE, Lieberman HR, Fulgoni VL, Pasiakos SM. Protein intake trends and conformity with the dietary reference intakes in the United States: analysis of the National Health and Nutrition Examination Survey, 2001–2014. Am J Clin Nutr. 2018;108(2):405–13. https://pubmed.ncbi.nlm.nih.gov/29931213/
Institute of Medicine of the National Academies. Dietary Reference Intakes for Energy, Carbohydrates, Fiber, Fat, Protein and Amino Acids (Macronutrients). The National Academies Press; 2002/2005. https://nap.nationalacademies.org/catalog/10490/dietary-reference-intakes-for-energy-carbohydrate-fiber-fat-fatty-acids-cholesterol-protein-and-amino-acids
Delimaris I. Adverse effects associated with protein intake above the recommended dietary allowance for adults. ISRN Nutr. 2013;2013:126929. https://pubmed.ncbi.nlm.nih.gov/24967251/
Volpi E, Campbell WW, Dwyer JT, et al. Is the optimal level of protein intake for older adults greater than the recommended dietary allowance? J Gerontol A Biol Sci Med Sci. 2013;68(6):677–81. https://pubmed.ncbi.nlm.nih.gov/23183903/
Shad BJ, Thompson JL, Breen L. Does the muscle protein synthetic response to exercise and amino acid – based nutrition diminish with advancing age? A systematic review. Am J Physiol Endocrinol Metab. 2016;311(5):E803–17. https://pubmed.ncbi.nlm.nih.gov/27555299/
Millward DJ. Protein requirements and aging. Am J Clin Nutr. 2014;100(4):1210–2. https://pubmed.ncbi.nlm.nih.gov/25240087/
Millward DJ, Fereday A, Gibson N, Pacy PJ. Aging, protein requirements, and protein turnover. Am J Clin Nutr. 1997;66(4):774–86. https://pubmed.ncbi.nlm.nih.gov/9322550/
Institute of Medicine of the National Academies. Dietary Reference Intakes for Energy, Carbohydrates, Fiber, Fat, Protein and Amino Acids (Macronutrients). The National Academies Press; 2002/2005. https://nap.nationalacademies.org/catalog/10490/dietary-reference-intakes-for-energy-carbohydrate-fiber-fat-fatty-acids-cholesterol-protein-and-amino-acids
European Food Safety Authority Panel on Dietetic Products, Nutrition and Allergies (NDA). Scientific opinion on dietary reference values for protein: dietary reference values for protein. EFSA J. 2012;10:2557–623. https://www.efsa.europa.eu/en/efsajournal/pub/2557
Joint Expert Consultation on Protein and Amino Acid Requirements in Human Nutrition, Weltgesundheitsorganisation, FAO, United Nations University, eds. Protein and Amino Acid Requirements in Human Nutrition: Report of a Joint WHO/FAO/UNU Expert Consultation [Geneva, 9–16 April 2002]. WHO; 2007. https://apps.who.int/iris/handle/10665/43411
Tieland M, Franssen R, Dullemeijer C, et al. The impact of dietary protein or amino acid supplementation on muscle mass and strength in elderly people: individual participant data and meta-analysis of RCT’s. J Nutr Health Aging. 2017;21(9):994–1001. https://pubmed.ncbi.nlm.nih.gov/29083440/
Reidy PT. Muscle or nothing! Where is the excess protein going in men with high protein intakes engaged in strength training? J Nutr. 2020;150(3):421–2. https://pubmed.ncbi.nlm.nih.gov/31897480/
Fluharty FL, McClure KE. Effects of dietary energy intake and protein concentration on performance and visceral organ mass in lambs. J Anim Sci. 1997;75(3):604–10. https://pubmed.ncbi.nlm.nih.gov/9078474/
Reidy PT. Muscle or nothing! Where is the excess protein going in men with high protein intakes engaged in strength training? J Nutr. 2020;150(3):421–2. https://pubmed.ncbi.nlm.nih.gov/31897480/
Reidy PT, Rasmussen BB. Role of ingested amino acids and protein in the promotion of resistance exercise – induced muscle protein anabolism. J Nutr. 2016;146(2):155–83. https://pubmed.ncbi.nlm.nih.gov/26764320/
Gielen E, Beckwée D, Delaere A, et al. Nutritional interventions to improve muscle mass, muscle strength, and physical performance in older people: an umbrella review of systematic reviews and meta-analyses. Nutr Rev. 2021;79(2):121–47. https://pubmed.ncbi.nlm.nih.gov/32483625/
ten Haaf DSM, Nuijten MAH, Maessen MFH, Horstman AMH, Eijsvogels TMH, Hopman MTE. Effects of protein supplementation on lean body mass, muscle strength, and physical performance in nonfrail community-dwelling older adults: a systematic review and meta-analysis. Am J Clin Nutr. 2018;108(5):1043–59. https://pubmed.ncbi.nlm.nih.gov/30475963/
Wouters OJ. Lobbying expenditures and campaign contributions by the pharmaceutical and health product industry in the United States, 1999–2018. JAMA Intern Med. 2020;180(5):688–97. https://pubmed.ncbi.nlm.nih.gov/32125357/
de Moraes MB, Avgerinou C, Fukushima FB, Vidal EIO. Nutritional interventions for the management of frailty in older adults: systematic review and meta-analysis of randomized clinical trials. Nutr Rev. 2021;79(8):889–913. https://pubmed.ncbi.nlm.nih.gov/33330911/
Tu DY, Kao FM, Tsai ST, Tung TH. Sarcopenia among the elderly population: a systematic review and meta-analysis of randomized controlled trials. Healthcare (Basel). 2021;9(6):650. https://pubmed.ncbi.nlm.nih.gov/34072617/
Roschel H, Hayashi AP, Fernandes AL, et al. Supplement-based nutritional strategies to tackle frailty: a multifactorial, double-blind, randomized placebo-controlled trial. Clin Nutr. 2021;40(8):4849–58. https://pubmed.ncbi.nlm.nih.gov/34358827/
Granic A, Hurst C, Dismore L, et al. Milk for skeletal muscle health and sarcopenia in older adults: a narrative review. Clin Interv Aging. 2020;15:695–714. https://pubmed.ncbi.nlm.nih.gov/32546988/
Huang LP, Condello G, Kuo CH. Effects of milk protein in resistance training – induced lean mass gains for older adults aged = 60 y: a systematic review and meta-analysis. Nutrients. 2021;13(8):2815. https://pubmed.ncbi.nlm.nih.gov/34444975/
Xu ZR, Tan ZJ, Zhang Q, Gui QF, Yang YM. The effectiveness of leucine on muscle protein synthesis, lean body mass and leg lean mass accretion in older people: a systematic review and meta-analysis. Br J Nutr. 2015;113(1):25–34. https://pubmed.ncbi.nlm.nih.gov/25234223/
Witkamp RF, van Norren K. Let thy food be thy medicine… when possible. Eur J Pharmacol. 2018;836:102–14. https://pubmed.ncbi.nlm.nih.gov/29936236/
Groen BBL, Horstman AM, Hamer HM, et al. Post-prandial protein handling: you are what you just ate. PLoS One. 2015;10(11):e0141582. https://pubmed.ncbi.nlm.nih.gov/26556791/
Figueiredo VC. Revisiting the roles of protein synthesis during skeletal muscle hypertrophy induced by exercise. Am J Physiol Regul Integr Comp Physiol. 2019;317(5):R709–18. https://pubmed.ncbi.nlm.nih.gov/31508978/
Mitchell CJ, Churchward-Venne TA, Parise G, et al. Acute post-exercise myofibrillar protein synthesis is not correlated with resistance training – induced muscle hypertrophy in young men. PLoS One. 2014;9(2):e89431. https://pubmed.ncbi.nlm.nih.gov/24586775/
Figueiredo VC. Revisiting the roles of protein synthesis during skeletal muscle hypertrophy induced by exercise. Am J Physiol Regul Integr Comp Physiol. 2019;317(5):R709–18. https://pubmed.ncbi.nlm.nih.gov/31508978/
Murphy CH, Oikawa SY, Phillips SM. Dietary protein to maintain muscle mass in aging: a case for per-meal protein recommendations. J Frailty Aging. 2016;5(1):49–58. https://pubmed.ncbi.nlm.nih.gov/26980369/
Bouillanne O, Neveux N, Nicolis I, Curis E, Cynober L, Aussel C. Long-lasting improved amino acid bioavailability associated with protein pulse feeding in hospitalized elderly patients: a randomized controlled trial. Nutrition. 2014;30(5):544–50. https://pubmed.ncbi.nlm.nih.gov/22992307/
Arnal MA, Mosoni L, Boirie Y, et al. Protein feeding pattern does not affect protein retention in young women. J Nutr. 2000;130(7):1700–4. https://pubmed.ncbi.nlm.nih.gov/10867039/
Arnal MA, Mosoni L, Boirie Y, et al. Protein pulse feeding improves protein retention in elderly women. Am J Clin Nutr. 1999;69(6):1202–8. https://pubmed.ncbi.nlm.nih.gov/10357740/
Bouillanne O, Curis E, Hamon-Vilcot B, et al. Impact of protein pulse feeding on lean mass in malnourished and at-risk hospitalized elderly patients: a randomized controlled trial. Clin Nutr. 2013;32(2):186–92. https://pubmed.ncbi.nlm.nih.gov/22992307/
Lim MT, Pan BJ, Toh DWK, Sutanto CN, Kim JE. Animal protein versus plant protein in supporting lean mass and muscle strength: a systematic review and meta-analysis of randomized controlled trials. Nutrients. 2021;13(2):661. https://pubmed.ncbi.nlm.nih.gov/33670701/
Tang JE, Moore DR, Kujbida GW, Tarnopolsky MA, Phillips SM. Ingestion of whey hydrolysate, casein, or soy protein isolate: effects on mixed muscle protein synthesis at rest and following resistance exercise in young men. J Appl Physiol (1985). 2009;107(3):987–92. https://pubmed.ncbi.nlm.nih.gov/19589961/
Haub MD, Wells AM, Tarnopolsky MA, Campbell WW. Effect of protein source on resistive-training-induced changes in body composition and muscle size in older men. Am J Clin Nutr. 2002;76(3):511–7. https://pubmed.ncbi.nlm.nih.gov/12197993/
Haub MD, Wells AM, Campbell WW. Beef and soy-based food supplements differentially affect serum lipoprotein – lipid profiles because of changes in carbohydrate intake and novel nutrient intake ratios in older men who resistive-train. Metabolism. 2005;54(6):769–74. https://pubmed.ncbi.nlm.nih.gov/15931612/
Wright CS, Zhou J, Sayer RD, Kim JE, Campbell WW. Effects of a high-protein diet including whole eggs on muscle composition and indices of cardiometabolic health and systemic inflammation in older adults with overweight or obesity: a randomized controlled trial. Nutrients. 2018;10(7):946. https://pubmed.ncbi.nlm.nih.gov/30041437/
Iglay HB, Apolzan JW, Gerrard DE, Eash JK, Anderson JC, Campbell WW. Moderately increased protein intake predominately from egg sources does not influence whole body, regional, or muscle composition responses to resistance training in older people. J Nutr Health Aging. 2009;13(2):108–14. https://pubmed.ncbi.nlm.nih.gov/19214338/
Bartholomae E, Incollingo A, Vizcaino M, Wharton C, Johnston CS. Mung bean protein supplement improves muscular strength in healthy, underactive vegetarian adults. Nutrients. 2019;11(10):E2423. https://pubmed.ncbi.nlm.nih.gov/31614532/
Vasconcelos QDJS, Bachur TPR, Aragão GF. Whey protein supplementation and its potentially adverse effects on health: a systematic review. Appl Physiol Nutr Metab. 2021;46(1):27–33. https://pubmed.ncbi.nlm.nih.gov/32702243/
Silverberg NB. Whey protein precipitating moderate to severe acne flares in 5 teenaged athletes. Cutis. 2012;90(2):70–2. https://pubmed.ncbi.nlm.nih.gov/22988649/
Simonart T. Acne and whey protein supplementation among bodybuilders. Dermatology. 2012;225(3):256–8. https://pubmed.ncbi.nlm.nih.gov/23257731/
Melnik BC, Zouboulis CC. Potential role of FoxO1 and mTORC1 in the pathogenesis of Western diet – induced acne. Exp Dermatol. 2013;22(5):311–5. https://pubmed.ncbi.nlm.nih.gov/23614736/
Liu KA, Lashinger LM, Rasmussen AJ, Hursting SD. Leucine supplementation differentially enhances pancreatic cancer growth in lean and overweight mice. Cancer Metab. 2014;2(1):6. https://pubmed.ncbi.nlm.nih.gov/24685128/
Cederroth CR, Vinciguerra M, Gjinovci A, et al. Dietary phytoestrogens activate AMP-activated protein kinase with improvement in lipid and glucose metabolism. Diabetes. 2008;57(5):1176–85. https://pubmed.ncbi.nlm.nih.gov/18420492/
Aubertin-Leheudre M, Lord C, Khalil A, Dionne IJ. Six months of isoflavone supplement increases fat-free mass in obese-sarcopenic postmenopausal women: a randomized double-blind controlled trial. Eur J Clin Nutr. 2007;61(12):1442–4. https://pubmed.ncbi.nlm.nih.gov/17311051/
Bhagwat S, Haytowitz DB, Holden JM. USDA database for the isoflavone content of selected foods: release 2.0. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/arsuserfiles/80400525/data/isoflav/isoflav_r2.pdf. Published September 2008. Accessed April 15, 2022.; https://www.ars.usda.gov/arsuserfiles/80400525/data/isoflav/isoflav_r2.pdf
Chan H, Ribeiro RV, Haden S, Hirani V. Plant-based dietary patterns, body composition, muscle strength and function in middle and older age: a systematic review. J Nutr Health Aging. 2021;25(8):1012–22. https://pubmed.ncbi.nlm.nih.gov/34545922/
Montiel-Rojas D, Nilsson A, Santoro A, et al. Fighting sarcopenia in ageing European adults: the importance of the amount and source of dietary proteins. Nutrients. 2020;12(12):3601. https://pubmed.ncbi.nlm.nih.gov/33255223/
Hengeveld LM, Wijnhoven HAH, Olthof MR, et al. Prospective associations of diet quality with incident frailty in older adults: the health, aging, and body composition study. J Am Geriatr Soc. 2019;67(9):1835–42. https://pubmed.ncbi.nlm.nih.gov/31267522/
Gazzani D, Zamboni F, Spelta F, et al. Vegetable but not animal protein intake is associated to a better physical performance: a study on a general population sample of adults. Food Nutr Res. 2019;63:3422. https://pubmed.ncbi.nlm.nih.gov/31565042/
Ortolá R, Struijk EA, García-Esquinas E, Rodríguez-Artalejo F, Lopez-Garcia E. Changes in dietary intake of animal and vegetable protein and unhealthy aging. Am J Med. 2020;133(2):231–9.e7. https://pubmed.ncbi.nlm.nih.gov/33839765/
Foscolou A, Critselis E, Tyrovolas S, et al. The association of animal and plant protein with successful ageing: a combined analysis of MEDIS and ATTICA epidemiological studies. Public Health Nutr. 2021;24(8):2215–24. https://pubmed.ncbi.nlm.nih.gov/32434609/
The nutrition source: protein. Harvard T.H. Chan School of Public Health. https://www.hsph.harvard.edu/nutritionsource/what-should-you-eat/protein/. Accessed August 3, 2022.; https://www.hsph.harvard.edu/nutritionsource/what-should-you-eat/protein/
Mariotti F, Gardner CD. Dietary protein and amino acids in vegetarian diets – a review. Nutrients. 2019;11(11):2661. https://pubmed.ncbi.nlm.nih.gov/31690027/
Ramarao PB, Norton HW, Johnson BC. The amino acids composition and nutritive value of proteins. v. amino acid requirements as a pattern for protein evaluation. J Nutr. 1964;82:88–92. https://pubmed.ncbi.nlm.nih.gov/14110945/
Mariotti F, Gardner CD. Dietary protein and amino acids in vegetarian diets – a review. Nutrients. 2019;11(11):2661. https://pubmed.ncbi.nlm.nih.gov/31690027/
Osborne TB, Mendel LB. Amino-acids in nutrition and growth. 1914. J Am Coll Nutr. 1993;12(5):484–5. https://pubmed.ncbi.nlm.nih.gov/8263262/
Davis TA, Nguyen HV, Garcia-Bravo R, et al. Amino acid composition of human milk is not unique. J Nutr. 1994;124(7):1126–32. https://pubmed.ncbi.nlm.nih.gov/8027865/
Sengupta P. The laboratory rat: relating its age with human’s. Int J Prev Med. 2013;4(6):624–30. https://pubmed.ncbi.nlm.nih.gov/23930179/
Young VR, Pellett PL. Plant proteins in relation to human protein and amino acid nutrition. Am J Clin Nutr. 1994;59(5 Suppl):1203S-12S. https://pubmed.ncbi.nlm.nih.gov/8172124/
Mariotti F, Gardner CD. Dietary protein and amino acids in vegetarian diets – a review. Nutrients. 2019;11(11):2661. https://pubmed.ncbi.nlm.nih.gov/31690027/
Hevia-Larraín V, Gualano B, Longobardi I, et al. High-protein plant-based diet versus a protein-matched omnivorous diet to support resistance training adaptations: a comparison between habitual vegans and omnivores. Sports Med. 2021;51(6):1317–30. https://pubmed.ncbi.nlm.nih.gov/33599941/
Damiano S, Muscariello E, La Rosa G, Di Maro M, Mondola P, Santillo M. Dual role of reactive oxygen species in muscle function: can antioxidant dietary supplements counteract age-related sarcopenia? Int J Mol Sci. 2019;20(15):E3815. https://pubmed.ncbi.nlm.nih.gov/31387214/
Muller FL, Song W, Liu Y, et al. Absence of CuZn superoxide dismutase leads to elevated oxidative stress and acceleration of age-dependent skeletal muscle atrophy. Free Radic Biol Med. 2006;40(11):1993–2004. https://pubmed.ncbi.nlm.nih.gov/16716900/
Sahni S, Dufour AB, Fielding RA, et al. Total carotenoid intake is associated with reduced loss of grip strength and gait speed over time in adults: The Framingham Offspring Study. Am J Clin Nutr. 2021;113(2):437–45. https://pubmed.ncbi.nlm.nih.gov/33181830/
Carr AC, Bozonet SM, Pullar JM, Simcock JW, Vissers MCM. Human skeletal muscle ascorbate is highly responsive to changes in vitamin C intake and plasma concentrations. Am J Clin Nutr. 2013;97(4):800–7. https://pubmed.ncbi.nlm.nih.gov/23446899/
Carr AC, Bozonet SM, Pullar JM, Simcock JW, Vissers MCM. Human skeletal muscle ascorbate is highly responsive to changes in vitamin C intake and plasma concentrations. Am J Clin Nutr. 2013;97(4):800–7. https://pubmed.ncbi.nlm.nih.gov/23446899/
Lewis LN, Hayhoe RPG, Mulligan AA, Luben RN, Khaw KT, Welch AA. Lower dietary and circulating vitamin C in middle– and older-aged men and women are associated with lower estimated skeletal muscle mass. J Nutr. 2020;150(10):2789–98. https://pubmed.ncbi.nlm.nih.gov/32851397/
Welch AA, Jennings A, Kelaiditi E, Skinner J, Steves CJ. Cross-sectional associations between dietary antioxidant vitamins C, E and carotenoid intakes and sarcopenic indices in women aged 18–79 years. Calcif Tissue Int. 2020;106(4):331–42. https://pubmed.ncbi.nlm.nih.gov/27417218/
Scott D, Blizzard L, Fell J, Giles G, Jones G. Associations between dietary nutrient intake and muscle mass and strength in community-dwelling older adults: the Tasmanian Older Adult Cohort Study. J Am Geriatr Soc. 2010;58(11):2129–34. https://pubmed.ncbi.nlm.nih.gov/21054294/
Saito K, Yokoyama T, Yoshida H, et al. A significant relationship between plasma vitamin C concentration and physical performance among Japanese elderly women. J Gerontol A Biol Sci Med Sci. 2012;67(3):295–301. https://pubmed.ncbi.nlm.nih.gov/21934124/
Takahashi F, Hashimoto Y, Kaji A, et al. Vitamin intake and loss of muscle mass in older people with type 2 diabetes: a prospective study of the KAMOGAWA-DM cohort. Nutrients. 2021;13(7):2335. https://pubmed.ncbi.nlm.nih.gov/34371843/
Saito K, Yokoyama T, Yoshida H, et al. A significant relationship between plasma vitamin C concentration and physical performance among Japanese elderly women. J Gerontol A Biol Sci Med Sci. 2012;67(3):295–301. https://pubmed.ncbi.nlm.nih.gov/21934124/
Tak YJ, Lee JG, Yi YH, et al. Association of handgrip strength with dietary intake in the Korean population: findings based on the Seventh Korea National Health and Nutrition Examination Survey (KNHANES VII-1), 2016. Nutrients. 2018;10(9):1180. https://pubmed.ncbi.nlm.nih.gov/30154371/
Gedmantaite A, Celis-Morales CA, Ho F, Pell JP, Ratkevicius A, Gray SR. Associations between diet and handgrip strength: a cross-sectional study from UK Biobank. Mech Ageing Dev. 2020;189:111269. https://pubmed.ncbi.nlm.nih.gov/32479757/
Fingeret M, Vollenweider P, Marques-Vidal P. No association between vitamin C and E supplementation and grip strength over 5 years: the Colaus study. Eur J Nutr. 2019;58(2):609–17. https://pubmed.ncbi.nlm.nih.gov/29484474/
Kenjale AA, Ham KL, Stabler T, et al. Dietary nitrate supplementation enhances exercise performance in peripheral arterial disease. J Appl Physiol (1985). 2011;110(6):1582–91. https://pubmed.ncbi.nlm.nih.gov/21454745/
Sobko T, Marcus C, Govoni M, Kamiya S. Dietary nitrate in Japanese traditional foods lowers diastolic blood pressure in healthy volunteers. Nitric Oxide. 2010;22(2):136–40. https://pubmed.ncbi.nlm.nih.gov/19887114/
What we eat in America, NHANES 2017–March 2020 prepandemic. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/1720/Table_1_NIN_GEN_1720.pdf. Published 2022. Accessed January 13, 2023.; https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/1720/Table_1_NIN_GEN_1720.pdf
Abete I, Konieczna J, Zulet MA, et al. Association of lifestyle factors and inflammation with sarcopenic obesity: data from the PREDIMED-Plus trial. J Cachexia Sarcopenia Muscle. 2019;10(5):974–84. https://pubmed.ncbi.nlm.nih.gov/31144432/
Chaput JP, Lord C, Cloutier M, et al. Relationship between antioxidant intakes and class I sarcopenia in elderly men and women. J Nutr Health Aging. 2007;11(4):363–9. https://pubmed.ncbi.nlm.nih.gov/17653501/
Verlaan S, Aspray TJ, Bauer JM, et al. Nutritional status, body composition, and quality of life in community-dwelling sarcopenic and non-sarcopenic older adults: a case-control study. Clin Nutr. 2017;36(1):267–74. https://pubmed.ncbi.nlm.nih.gov/26689868/
ter Borg S, de Groot LCPGM, Mijnarends DM, et al. Differences in nutrient intake and biochemical nutrient status between sarcopenic and nonsarcopenic older adults – results from the Maastricht Sarcopenia Study. J Am Med Dir Assoc. 2016;17(5):393–401. https://pubmed.ncbi.nlm.nih.gov/26825685/
Clifford T, Jeffries O, Stevenson EJ, Davies KAB. The effects of vitamin C and E on exercise-induced physiological adaptations: a systematic review and meta-analysis of randomised controlled trials. Crit Rev Food Sci Nutr. 2020;60(21):3669–79. https://pubmed.ncbi.nlm.nih.gov/31851538/
Jackson MA, Jeffery IB, Beaumont M, et al. Signatures of early frailty in the gut microbiota. Genome Med. 2016;8(1):8. https://pubmed.ncbi.nlm.nih.gov/26822992/
Ticinesi A, Nouvenne A, Cerundolo N, et al. Gut microbiota, muscle mass and function in aging: a focus on physical frailty and sarcopenia. Nutrients. 2019;11(7):E1633. https://pubmed.ncbi.nlm.nih.gov/31319564/
van Tongeren SP, Slaets JPJ, Harmsen HJM, Welling GW. Fecal microbiota composition and frailty. Appl Environ Microbiol. 2005;71(10):6438–42. https://pubmed.ncbi.nlm.nih.gov/16204576/
Montiel-Rojas D, Nilsson A, Santoro A, et al. Dietary fibre may mitigate sarcopenia risk: findings from the NU-AGE cohort of older European adults. Nutrients. 2020;12(4):E1075. https://pubmed.ncbi.nlm.nih.gov/32295007/
Berendsen AAM, van de Rest O, Feskens EJM, et al. Changes in dietary intake and adherence to the NU-AGE diet following a one-year dietary intervention among European older adults – results of the NU-AGE randomized trial. Nutrients. 2018;10(12):E1905. https://pubmed.ncbi.nlm.nih.gov/30518044/
Buigues C, Fernández-Garrido J, Pruimboom L, et al. Effect of a prebiotic formulation on frailty syndrome: a randomized, double-blind clinical trial. Int J Mol Sci. 2016;17(6):E932. https://pubmed.ncbi.nlm.nih.gov/27314331/
Lee MC, Tu YT, Lee CC, et al. Lactobacillus plantarum TWK10 improves muscle mass and functional performance in frail older adults: a randomized, double-blind clinical trial. Microorganisms. 2021;9(7):1466. https://pubmed.ncbi.nlm.nih.gov/34361902/
Yang HL, Feng P, Xu Y, Hou YY, Ojo O, Wang XH. The role of dietary fiber supplementation in regulating uremic toxins in patients with chronic kidney disease: a meta-analysis of randomized controlled trials. J Ren Nutr. 2021;31(5):438–47. https://pubmed.ncbi.nlm.nih.gov/33741249/
Strasser B, Wolters M, Weyh C, Krüger K, Ticinesi A. The effects of lifestyle and diet on gut microbiota composition, inflammation and muscle performance in our aging society. Nutrients. 2021;13(6):2045. https://pubmed.ncbi.nlm.nih.gov/34203776/
Enoki Y, Watanabe H, Arake R, et al. Indoxyl sulfate potentiates skeletal muscle atrophy by inducing the oxidative stress – mediated expression of myostatin and atrogin-1. Sci Rep. 2016;6:32084. https://pubmed.ncbi.nlm.nih.gov/27549031/
Dawson-Hughes B, Castaneda-Sceppa C, Harris SS, et al. Impact of supplementation with bicarbonate on lower-extremity muscle performance in older men and women. Osteoporos Int. 2010;21(7):1171–9. https://pubmed.ncbi.nlm.nih.gov/19727904/
Guder WG, Häussinger D, Gerok W. Renal and hepatic nitrogen metabolism in systemic acid base regulation. J Clin Chem Clin Biochem. 1987;25(8):457–66. https://pubmed.ncbi.nlm.nih.gov/3320262/
Hamm LL, Ambühl PM, Alpern RJ. Role of glucocorticoids in acidosis. Am J Kidney Dis. 1999;34(5):960–5. https://pubmed.ncbi.nlm.nih.gov/10561158/
Dawson-Hughes B, Harris SS, Ceglia L. Alkaline diets favor lean tissue mass in older adults. Am J Clin Nutr. 2008;87(3):662–5. https://pubmed.ncbi.nlm.nih.gov/18326605/
Buehlmeier J, Remer T, Frings-Meuthen P, Maser-Gluth C, Heer M. Glucocorticoid activity and metabolism with NaCl-induced low-grade metabolic acidosis and oral alkalization: results of two randomized controlled trials. Endocrine. 2016;52(1):139–47. https://pubmed.ncbi.nlm.nih.gov/26349936/
Dawson-Hughes B, Castaneda-Sceppa C, Harris SS, et al. Impact of supplementation with bicarbonate on lower-extremity muscle performance in older men and women. Osteoporos Int. 2010;21(7):1171–9. https://pubmed.ncbi.nlm.nih.gov/19727904/
Caciano SL, Inman CL, Gockel-Blessing EE, Weiss EP. Effects of dietary acid load on exercise metabolism and anaerobic exercise performance. J Sports Sci Med. 2015;14(2):364–71. https://pubmed.ncbi.nlm.nih.gov/25983586/
Trinchieri A. Development of a rapid food screener to assess the potential renal acid load of diet in renal stone formers (LAKE score). Arch Ital Urol Androl. 2012;84(1):36–8. https://pubmed.ncbi.nlm.nih.gov/22649959/
Cosgrove K, Johnston CS. Examining the impact of adherence to a vegan diet on acid-base balance in healthy adults. Plant Foods Hum Nutr. 2017;72(3):308–13. https://pubmed.ncbi.nlm.nih.gov/28677099/
Buehlmeier J, Remer T, Frings-Meuthen P, Maser-Gluth C, Heer M. Glucocorticoid activity and metabolism with NaCl-induced low-grade metabolic acidosis and oral alkalization: results of two randomized controlled trials. Endocrine. 2016;52(1):139–47. https://pubmed.ncbi.nlm.nih.gov/26349936/
Yoshida Y, Kosaki K, Sugasawa T, et al. High salt diet impacts the risk of sarcopenia associated with reduction of skeletal muscle performance in the Japanese population. Nutrients. 2020;12(11):3474. https://pubmed.ncbi.nlm.nih.gov/33198295/
Appel LJ, Anderson CAM. Compelling evidence for public health action to reduce salt intake. N Engl J Med. 2010;362(7):650–2. https://pubmed.ncbi.nlm.nih.gov/20089959/
Qian Q. Dietary influence on body fluid acid-base and volume balance: the deleterious “norm” furthers and cloaks subclinical pathophysiology. Nutrients. 2018;10(6):E778. https://pubmed.ncbi.nlm.nih.gov/29914153/
Milajerdi A, Hassanzadeh Keshteli A, Haghighatdoost F, Azadbakht L, Esmaillzadeh A, Adibi P. Dietary acid load in relation to depression and anxiety in adults. J Hum Nutr Diet. 2020;33(1):48–55. https://pubmed.ncbi.nlm.nih.gov/31173421/
Xue X, Liu Z, Li X, et al. The efficacy and safety of citrate mixture vs sodium bicarbonate on urine alkalization in Chinese primary gout patients with benzbromarone: a prospective, randomized controlled study. Rheumatology (Oxford). 2021;60(6):2661–71. https://pubmed.ncbi.nlm.nih.gov/33211886/
Sebastian A, Frassetto LA, Sellmeyer DE, Merriam RL, Morris RC. Estimation of the net acid load of the diet of ancestral preagricultural Homo sapiens and their hominid ancestors. Am J Clin Nutr. 2002;76(6):1308–16. https://pubmed.ncbi.nlm.nih.gov/12450898/
Logozzi M, Mizzoni D, Di Raimo R, et al. In vivo antiaging effects of alkaline water supplementation. J Enzyme Inhib Med Chem. 35(1):657–64.; https://pubmed.ncbi.nlm.nih.gov/32106720/
Magro M, Corain L, Ferro S, et al. Alkaline water and longevity: a murine study. Evid Based Complement Alternat Med. 2016;2016:3084126. https://pubmed.ncbi.nlm.nih.gov/27340414/
Wesson DE. Is NaHCO3 an antiaging elixir? Am J Physiol Renal Physiol. 2016;311(1):F182–3. https://pubmed.ncbi.nlm.nih.gov/27029429/
Kim J, Lee Y, Kye S, Chung YS, Kim KM. Association of vegetables and fruits consumption with sarcopenia in older adults: the Fourth Korea National Health and Nutrition Examination Survey. Age Ageing. 2015;44(1):96–102. https://pubmed.ncbi.nlm.nih.gov/24646604/
Koyanagi A, Veronese N, Solmi M, et al. Fruit and vegetable consumption and sarcopenia among older adults in low– and middle-income countries. Nutrients. 2020;12(3):E706. https://pubmed.ncbi.nlm.nih.gov/32155879/
García-Esquinas E, Rahi B, Peres K, et al. Consumption of fruit and vegetables and risk of frailty: a dose-response analysis of 3 prospective cohorts of community-dwelling older adults. Am J Clin Nutr. 2016;104(1):132–42. https://pubmed.ncbi.nlm.nih.gov/27194305/
Sim M, Blekkenhorst LC, Lewis JR, et al. Vegetable and fruit intake and injurious falls risk in older women: a prospective cohort study. Br J Nutr. 2018;120(8):925–34. https://pubmed.ncbi.nlm.nih.gov/30153877/
Schrager MA, Hilton J, Gould R, Kelly VE. Effects of blueberry supplementation on measures of functional mobility in older adults. Appl Physiol Nutr Metab. 2015;40(6):543–9. https://pubmed.ncbi.nlm.nih.gov/25909473/
Sangouni AA, Azar MRMH, Alizadeh M. Effects of garlic powder supplementation on insulin resistance, oxidative stress, and body composition in patients with non-alcoholic fatty liver disease: a randomized controlled clinical trial. Complement Ther Med. 2020;51:102428. https://pubmed.ncbi.nlm.nih.gov/32507439/
Pérez-Piñero S, Ávila-Gandía V, Rubio Arias JA, Muñoz-Carrillo JC, Losada-Zafrilla P, López-Román FJ. A 12-week randomized double-blind placebo-controlled clinical trial, evaluating the effect of supplementation with a spinach extract on skeletal muscle fitness in adults older than 50 years of age. Nutrients. 2021;13(12):4373. https://pubmed.ncbi.nlm.nih.gov/34959924/
Dirks-Naylor AJ. The benefits of coffee on skeletal muscle. Life Sci. 2015;143:182–6. https://pubmed.ncbi.nlm.nih.gov/26546720/
Sanchez AMJ, Bernardi H, Py G, Candau RB. Autophagy is essential to support skeletal muscle plasticity in response to endurance exercise. Am J Physiol Regul Integr Comp Physiol. 2014;307(8):R956–69. https://pubmed.ncbi.nlm.nih.gov/25121614/
Marzetti E, Calvani R, Cesari M, et al. Mitochondrial dysfunction and sarcopenia of aging: from signaling pathways to clinical trials. Int J Biochem Cell Biol. 2013;45(10):2288–301. https://pubmed.ncbi.nlm.nih.gov/23845738/
Guo Y, Niu K, Okazaki T, et al. Coffee treatment prevents the progression of sarcopenia in aged mice in vivo and in vitro. Exp Gerontol. 2014;50:1–8. https://pubmed.ncbi.nlm.nih.gov/24269808/
Jyväkorpi SK, Urtamo A, Kivimäki M, Strandberg TE. Associations of coffee drinking with physical performance in the oldest-old community-dwelling men The Helsinki Businessmen Study (HBS). Aging Clin Exp Res. 2021;33(5):1371–5. https://pubmed.ncbi.nlm.nih.gov/32638343/
Iwasaka C, Yamada Y, Nishida Y, et al. Association between habitual coffee consumption and skeletal muscle mass in middle-aged and older Japanese people. Geriatr Gerontol Int. 2021;21(10):950–8. https://pubmed.ncbi.nlm.nih.gov/34405954/
Wang T, Wu Y, Wang W, Zhang D. Association between coffee consumption and functional disability in older US adults. Br J Nutr. 2021;125(6):695–702. https://pubmed.ncbi.nlm.nih.gov/32778181/
Chung H, Moon JH, Kim JI, Kong MH, Huh JS, Kim HJ. Association of coffee consumption with sarcopenia in Korean elderly men: analysis using the Korea National Health and Nutrition Examination Survey, 2008–2011. Korean J Fam Med. 2017;38(3):141–7. https://pubmed.ncbi.nlm.nih.gov/28572890/
Wang T, Wu Y, Wang W, Zhang D. Association between coffee consumption and functional disability in older US adults. Br J Nutr. 2021;125(6):695–702. https://pubmed.ncbi.nlm.nih.gov/32778181/
Grgic J, Grgic I, Pickering C, Schoenfeld BJ, Bishop DJ, Pedisic Z. Wake up and smell the coffee: caffeine supplementation and exercise performance – an umbrella review of 21 published meta-analyses. Br J Sports Med. 2020;54(11):681–8. https://pubmed.ncbi.nlm.nih.gov/30926628/
Rivers WHR, Webber HN. The action of caffeine on the capacity for muscular work. J Physiol. 1907;36(1):33–47. https://pubmed.ncbi.nlm.nih.gov/16992882/
Grgic J, Grgic I, Pickering C, Schoenfeld BJ, Bishop DJ, Pedisic Z. Wake up and smell the coffee: caffeine supplementation and exercise performance – an umbrella review of 21 published meta-analyses. Br J Sports Med. 2020;54(11):681–8. https://pubmed.ncbi.nlm.nih.gov/30926628/
Duncan MJ, Clarke ND, Tallis J, Guimarães-Ferreira L, Leddington-Wright S. The effect of caffeine ingestion on functional performance in older adults. J Nutr Health Aging. 2014;18(10):883–7. https://pubmed.ncbi.nlm.nih.gov/25470803/
Norager CB, Jensen MB, Madsen MR, Laurberg S. Caffeine improves endurance in 75-yr-old citizens: a randomized, double-blind, placebo-controlled, crossover study. J Appl Physiol (1985). 2005;99(6):2302–6. https://pubmed.ncbi.nlm.nih.gov/16081625/
Bakuradze T, Parra GAM, Riedel A, et al. Four-week coffee consumption affects energy intake, satiety regulation, body fat, and protects DNA integrity. Food Res Int. 2014;63:420–7. https://www.sciencedirect.com/science/article/abs/pii/S0963996914003378
Jang YJ, Son HJ, Kim JS, et al. Coffee consumption promotes skeletal muscle hypertrophy and myoblast differentiation. Food Funct. 2018;9(2):1102–11. https://pubmed.ncbi.nlm.nih.gov/29359224/
McDermott MM, Criqui MH, Domanchuk K, et al. Cocoa to improve walking performance in older people with peripheral artery disease: the COCOA-PAD pilot randomized clinical trial. Circ Res. 2020;126(5):589–99. https://pubmed.ncbi.nlm.nih.gov/32078436/
Munguia L, Rubio-Gayosso I, Ramirez-Sanchez I, et al. High flavonoid cocoa supplement ameliorates plasma oxidative stress and inflammation levels while improving mobility and quality of life in older subjects: a double-blind randomized clinical trial. J Gerontol A Biol Sci Med Sci. 2019;74(10):1620–7. https://pubmed.ncbi.nlm.nih.gov/31056655/
Navrátil T, Kohlíková E, Petr M, Pelclová D, Heyrovský M, Pristoupilová K. Supplemented creatine induces changes in human metabolism of thiocompounds and one– and two-carbon units. Physiol Res. 2010;59(3):431–42. https://pubmed.ncbi.nlm.nih.gov/19249916/
Sumien N, Shetty RA, Gonzales EB. Creatine, creatine kinase, and aging. In: Harris JR, Korolchuk VI, eds. Biochemistry and Cell Biology of Ageing: Part I Biomedical Science. Vol 90. Springer; 2018:145–68. https://pubmed.ncbi.nlm.nih.gov/30779009/
Balestrino M, Adriano E. Beyond sports: efficacy and safety of creatine supplementation in pathological or paraphysiological conditions of brain and muscle. Med Res Rev. 2019;39(6):2427–59. https://pubmed.ncbi.nlm.nih.gov/31012130/
Kraemer WJ, Beeler MK, Post EM, et al. Physiological basis for creatine supplementation in skeletal muscle and the central nervous system. In: Sen CK, Nair S, Bagchi D, eds. Nutrition and Enhanced Sports Performance: Muscle Building, Endurance, and Strength. 2nd ed. Academic Press; 2019:581–94. https://www.sciencedirect.com/science/article/abs/pii/B9780128139226000497?via%3Dihub
Blancquaert L, Baguet A, Bex T, et al. Changing to a vegetarian diet reduces the body creatine pool in omnivorous women, but appears not to affect carnitine and carnosine homeostasis: a randomised trial. Br J Nutr. 2018;119(7):759–70. https://pubmed.ncbi.nlm.nih.gov/29569535/
Solis MY, Painelli V de S, Artioli GG, Roschel H, Otaduy MC, Gualano B. Brain creatine depletion in vegetarians? A cross-sectional ¹H-magnetic resonance spectroscopy (¹H-MRS) study. Br J Nutr. 2014;111(7):1272–4. https://pubmed.ncbi.nlm.nih.gov/24290771/
Blancquaert L, Baguet A, Bex T, et al. Changing to a vegetarian diet reduces the body creatine pool in omnivorous women, but appears not to affect carnitine and carnosine homeostasis: a randomised trial. Br J Nutr. 2018;119(7):759–70. https://pubmed.ncbi.nlm.nih.gov/29569535/
Shomrat A, Weinstein Y, Katz A. Effect of creatine feeding on maximal exercise performance in vegetarians. Eur J Appl Physiol. 2000;82(4):321–5. https://pubmed.ncbi.nlm.nih.gov/10958375/
Steenge GR, Verhoef P, Greenhaff PL. The effect of creatine and resistance training on plasma homocysteine concentration in healthy volunteers. Arch Intern Med. 2001;161(11):1455–6. https://pubmed.ncbi.nlm.nih.gov/11386896/
Chilibeck PD, Kaviani M, Candow DG, Zello GA. Effect of creatine supplementation during resistance training on lean tissue mass and muscular strength in older adults: a meta-analysis. Open Access J Sports Med. 2017;8:213–26. https://pubmed.ncbi.nlm.nih.gov/29138605/
Buford TW, Kreider RB, Stout JR, et al. International Society of Sports Nutrition position stand: creatine supplementation and exercise. J Int Soc Sports Nutr. 2007;4:6. https://pubmed.ncbi.nlm.nih.gov/17908288/
Riesberg LA, Weed SA, McDonald TL, Eckerson JM, Drescher KM. Beyond muscles: the untapped potential of creatine. Int Immunopharmacol. 2016;37:31–42. https://pubmed.ncbi.nlm.nih.gov/26778152/
Antonio J, Candow DG, Forbes SC, et al. Common questions and misconceptions about creatine supplementation: what does the scientific evidence really show? J Int Soc Sports Nutr. 2021;18(1):13. https://pubmed.ncbi.nlm.nih.gov/33557850/
Dolan E, Artioli GG, Pereira RMR, Gualano B. Muscular atrophy and sarcopenia in the elderly: is there a role for creatine supplementation? Biomolecules. 2019;9(11):E642. https://pubmed.ncbi.nlm.nih.gov/31652853/
Syrotuik DG, Bell GJ, Burnham R, Sim LL, Calvert RA, Maclean IM. Absolute and relative strength performance following creatine monohydrate supplementation combined with periodized resistance training: J Strength Cond Res. 2000;14(2):182–90. https://journals.lww.com/nsca-jscr/Abstract/2000/05000/Absolute_and_Relative_Strength_Performance.11.aspx
Beaudart C, Dawson A, Shaw SC, et al. Nutrition and physical activity in the prevention and treatment of sarcopenia: systematic review. Osteoporos Int. 2017;28(6):1817–33. https://pubmed.ncbi.nlm.nih.gov/28251287/
Antonio J, Candow DG, Forbes SC, et al. Common questions and misconceptions about creatine supplementation: what does the scientific evidence really show? J Int Soc Sports Nutr. 2021;18(1):13. https://pubmed.ncbi.nlm.nih.gov/33557850/
Chilibeck PD, Kaviani M, Candow DG, Zello GA. Effect of creatine supplementation during resistance training on lean tissue mass and muscular strength in older adults: a meta-analysis. Open Access J Sports Med. 2017;8:213–26. https://pubmed.ncbi.nlm.nih.gov/29138605/
Gualano B, Rawson ES, Candow DG, Chilibeck PD. Creatine supplementation in the aging population: effects on skeletal muscle, bone and brain. Amino Acids. 2016;48(8):1793–805. https://pubmed.ncbi.nlm.nih.gov/27108136/
Beaudart C, Dawson A, Shaw SC, et al. Nutrition and physical activity in the prevention and treatment of sarcopenia: systematic review. Osteoporos Int. 2017;28(6):1817–33. https://pubmed.ncbi.nlm.nih.gov/28251287/
Candow DG, Chilibeck PD, Chad KE, Chrusch MJ, Davison KS, Burke DG. Effect of ceasing creatine supplementation while maintaining resistance training in older men. J Aging Phys Act. 2004;12(3):219–31. https://pubmed.ncbi.nlm.nih.gov/15263100/
Beaudart C, Rabenda V, Simmons M, et al. Effects of protein, essential amino acids, ß-hydroxy ß-methylbutyrate, creatine, dehydroepiandrosterone and fatty acid supplementation on muscle mass, muscle strength and physical performance in older people aged 60 years and over. A systematic review on the literature. J Nutr Health Aging. 2018;22(1):117–30. https://pubmed.ncbi.nlm.nih.gov/29300431/
Candow DG, Forbes SC, Chilibeck PD, Cornish SM, Antonio J, Kreider RB. Effectiveness of creatine supplementation on aging muscle and bone: focus on falls prevention and inflammation. J Clin Med. 2019;8(4):E488. https://pubmed.ncbi.nlm.nih.gov/30978926/
MacRae PG, Lacourse M, Moldavon R. Physical performance measures that predict faller status in community-dwelling older adults. J Orthop Sports Phys Ther. 1992;16(3):123–8. https://pubmed.ncbi.nlm.nih.gov/18796765/
Dolan E, Artioli GG, Pereira RMR, Gualano B. Muscular atrophy and sarcopenia in the elderly: is there a role for creatine supplementation? Biomolecules. 2019;9(11):E642. https://pubmed.ncbi.nlm.nih.gov/31652853/
Morley JE, Argiles JM, Evans WJ, et al. Nutritional recommendations for the management of sarcopenia. J Am Med Dir Assoc. 2010;11(6):391–6. https://pubmed.ncbi.nlm.nih.gov/20627179/
Wu G. Important roles of dietary taurine, creatine, carnosine, anserine and 4-hydroxyproline in human nutrition and health. Amino Acids. 2020;52(3):329–60. https://pubmed.ncbi.nlm.nih.gov/32072297/
Hultman E, Söderlund K, Timmons JA, Cederblad G, Greenhaff PL. Muscle creatine loading in men. J Appl Physiol (1985). 1996;81(1):232–7. https://pubmed.ncbi.nlm.nih.gov/8828669/
Stares A, Bains M. The additive effects of creatine supplementation and exercise training in an aging population: a systematic review of randomized controlled trials. J Geriatr Phys Ther. 2020;43(2):99–112. https://pubmed.ncbi.nlm.nih.gov/30762623/
Ribeiro F, Longobardi I, Perim P, et al. Timing of creatine supplementation around exercise: a real concern? Nutrients. 2021;13(8):2844. https://pubmed.ncbi.nlm.nih.gov/34445003/
Bender A, Beckers J, Schneider I, et al. Creatine improves health and survival of mice. Neurobiol Aging. 2008;29(9):1404–11. https://pubmed.ncbi.nlm.nih.gov/17416441/
Korzun WJ. Oral creatine supplements lower plasma homocysteine concentrations in humans. Clin Lab Sci. 2004;17(2):102–6. https://pubmed.ncbi.nlm.nih.gov/15168891/
Moret S, Prevarin A, Tubaro F. Levels of creatine, organic contaminants and heavy metals in creatine dietary supplements. Food Chem. 2011;126(3):1232–8. https://www.sciencedirect.com/science/article/abs/pii/S0308814610016377
Cooperman T. Muscle & workout supplements review (creatine and branched-chain amino acids). ConsumerLab.com. https://www.consumerlab.com/reviews/review-creatine-bcaas/creatine/. Published January 23, 2017. Updated June 30, 2022. Accessed August 3, 2022.; https://www.consumerlab.com/reviews/review-creatine-bcaas/creatine/
Louis ED, Ferreira JJ. How common is the most common adult movement disorder? Update on the worldwide prevalence of essential tremor. Mov Disord. 2010;25(5):534–41. https://pubmed.ncbi.nlm.nih.gov/20175185/
Louis ED. Essential tremor then and now: how views of the most common tremor diathesis have changed over time. Parkinsonism Relat Disord. 2018;46(Suppl 1):S70–4. https://pubmed.ncbi.nlm.nih.gov/28747278/
Hopfner F, Helmich RC. The etiology of essential tremor: genes versus environment. Parkinsonism Relat Disord. 2018;46 Suppl 1:S92–6. https://pubmed.ncbi.nlm.nih.gov/28735798/
Pfau W, Skog K. Exposure to ß-carbolines norharman and harman. J Chromatogr B Analyt Technol Biomed Life Sci. 2004;802(1):115–26. https://pubmed.ncbi.nlm.nih.gov/15036003/
Gibis M, Weiss J. Inhibitory effect of marinades with hibiscus extract on formation of heterocyclic aromatic amines and sensory quality of fried beef patties. Meat Sci. 2010;85(4):735–42. https://pubmed.ncbi.nlm.nih.gov/20418021/
Busquets R, Puignou L, Galceran MT, Skog K. Effect of red wine marinades on the formation of heterocyclic amines in fried chicken breast. J Agric Food Chem. 2006;54(21):8376–84. https://pubmed.ncbi.nlm.nih.gov/17032054/
Smith JS, Ameri F, Gadgil P. Effect of marinades on the formation of heterocyclic amines in grilled beef steaks. J Food Sci. 2008;73(6):T100–5. https://pubmed.ncbi.nlm.nih.gov/19241593/
Khan MR, Busquets R, Azam M. Blueberry, raspberry, and strawberry extracts reduce the formation of carcinogenic heterocyclic amines in fried camel, beef and chicken meats. Food Control. 2021;123:107852. https://www.sciencedirect.com/science/article/abs/pii/S0956713520307684
Abdulrahman AA, Faisal K, Meshref AAA, Arshaduddin M. Low-dose acute vanillin is beneficial against harmaline-induced tremors in rats. Neurol Res. 2017;39(3):264–70. https://pubmed.ncbi.nlm.nih.gov/28095756/
Srinivasan S, Glover J, Tampi RR, Tampi DJ, Sewell DD. Sexuality and the older adult. Curr Psychiatry Rep. 2019;21(10):97. https://pubmed.ncbi.nlm.nih.gov/31522296/
Doll GM. Sexuality in nursing homes: practice and policy. J Gerontol Nurs. 2013;39(7):30–7. https://pubmed.ncbi.nlm.nih.gov/23614386/
Mishra BN. Secret of eternal youth; teaching from the centenarian hot spots (“blue zones”). Indian J Community Med. 2009;34(4):273–5. https://pubmed.ncbi.nlm.nih.gov/20165615/
Morton L. Sexuality in the older adult. Prim Care. 2017;44(3):429–38. https://pubmed.ncbi.nlm.nih.gov/28797370/
Gewirtz-Meydan A, Hafford-Letchfield T, Ayalon L, et al. How do older people discuss their own sexuality? A systematic review of qualitative research studies. Cult Health Sex. 2019;21(3):293–308. https://pubmed.ncbi.nlm.nih.gov/29863969/
Lindau ST, Schumm LP, Laumann EO, Levinson W, O’Muircheartaigh CA, Waite LJ. A study of sexuality and health among older adults in the United States. N Engl J Med. 2007;357(8):762–74. https://pubmed.ncbi.nlm.nih.gov/17715410/
Srinivasan S, Glover J, Tampi RR, Tampi DJ, Sewell DD. Sexuality and the older adult. Curr Psychiatry Rep. 2019;21(10):97. https://pubmed.ncbi.nlm.nih.gov/31522296/
Cao C, Yang L, Xu T, et al. Trends in sexual activity and associations with all-cause and cause-specific mortality among US adults. J Sex Med. 2020;17(10):1903–13. https://pubmed.ncbi.nlm.nih.gov/32665214/
Cao C, Yang L, Xu T, et al. Trends in sexual activity and associations with all-cause and cause-specific mortality among US adults. J Sex Med. 2020;17(10):1903–13. https://pubmed.ncbi.nlm.nih.gov/32665214/
Kay N, Allen J, Morley JE. Endorphins stimulate normal human peripheral blood lymphocyte natural killer activity. Life Sci. 1984;35(1):53–9. https://pubmed.ncbi.nlm.nih.gov/6204182/
Cao C, Yang L, Xu T, et al. Trends in sexual activity and associations with all-cause and cause-specific mortality among US adults. J Sex Med. 2020;17(10):1903–13. https://pubmed.ncbi.nlm.nih.gov/32665214/
Casazza K, Fontaine KR, Astrup A, et al. Myths, presumptions, and facts about obesity. N Engl J Med. 2013;368(5):446–54. https://pubmed.ncbi.nlm.nih.gov/23363498/
Bohlen JG, Held JP, Sanderson MO, Patterson RP. Heart rate, rate-pressure product, and oxygen uptake during four sexual activities. Arch Intern Med. 1984;144(9):1745–8. https://pubmed.ncbi.nlm.nih.gov/6476990/
Allen MS, Walter EE. Health-related lifestyle factors and sexual dysfunction: a meta-analysis of population-based research. J Sex Med. 2018;15(4):458–75. https://pubmed.ncbi.nlm.nih.gov/29523476/
Maiorino MI, Bellastella G, Caputo M, et al. Effects of Mediterranean diet on sexual function in people with newly diagnosed type 2 diabetes: the MÈDITA trial. J Diabetes Complications. 2016;30(8):1519–24. https://pubmed.ncbi.nlm.nih.gov/27614727/
Maiorino MI, Bellastella G, Chiodini P, et al. Primary prevention of sexual dysfunction with Mediterranean diet in type 2 diabetes: the MÈDITA randomized trial. Diabetes Care. 2016;39(9):e143–4. https://pubmed.ncbi.nlm.nih.gov/27352954/
Pazzaglia M. Body and odors: not just molecules, after all. Curr Dir Psychol Sci. 2015;24(4):329–33. https://journals.sagepub.com/doi/abs/10.1177/0963721415575329
Herz RS, Inzlicht M. Sex differences in response to physical and social factors involved in human mate selection: the importance of smell for women. Evol Hum Behav. 2002;23(5):359–64. https://www.sciencedirect.com/science/article/abs/pii/S1090513802000958
Habel U, Regenbogen C, Kammann C, Stickel S, Chechko N. Male brain processing of the body odor of ovulating women compared to that of pregnant women. Neuroimage. 2021;229:117733. https://pubmed.ncbi.nlm.nih.gov/33484852/
Nishihira J, Nishimura M, Tanaka A, Yamaguchi A, Taira T. Effects of 4-week continuous ingestion of champignon extract on halitosis and body and fecal odor. J Tradit Complement Med. 2015;7(1):110–6. https://pubmed.ncbi.nlm.nih.gov/28053896/
Nazzaro-Porro M, Passi S, Boniforti L, Belsito F. Effects of aging on fatty acids in skin surface lipids. J Invest Dermatol. 1979;73(1):112–7. https://pubmed.ncbi.nlm.nih.gov/448170/
Nishihira J, Nishimura M, Tanaka A, Yamaguchi A, Taira T. Effects of 4-week continuous ingestion of champignon extract on halitosis and body and fecal odor. J Tradit Complement Med. 2015;7(1):110–6. https://pubmed.ncbi.nlm.nih.gov/28053896/
Agricultural Research Service, United States Department of Agriculture. Mushrooms, white, raw. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html?query=mushrooms&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/169251/nutrients. Published April 1, 2019. Accessed August 24, 2022.; https://fdc.nal.usda.gov/fdc-app.html?query=mushrooms&utf8=%E2%9C%93&affiliate=usda&commit=Search#/food-details/169251/nutrients
Kephart JC. Chlorophyll derivatives – their chemistry, commercial preparation and uses. Econ Bot. 1955;9(1):3–38. https://link.springer.com/article/10.1007/BF02984956
Bohn T, Walczyk T, Leisibach S, Hurrell RF. Chlorophyll-bound magnesium in commonly consumed vegetables and fruits: relevance to magnesium nutrition. J Food Sci. 2006;69(9):S347–50. https://ift.onlinelibrary.wiley.com/doi/abs/10.1111/j.1365–2621.2004.tb09947.x
Olsson MJ, Lundström JN, Kimball BA, et al. The scent of disease: human body odor contains an early chemosensory cue of sickness. Psychol Sci. 2014;25(3):817–23. https://pubmed.ncbi.nlm.nih.gov/24452606/
Olsson MJ, Lundström JN, Kimball BA, et al. The scent of disease: human body odor contains an early chemosensory cue of sickness. Psychol Sci. 2014;25(3):817–23. https://pubmed.ncbi.nlm.nih.gov/24452606/
Havlicek J, Lenochova P. The effect of meat consumption on body odor attractiveness. Chem Senses. 2006;31(8):747–52. https://pubmed.ncbi.nlm.nih.gov/16891352/
Havlicek J, Lenochova P. The effect of meat consumption on body odor attractiveness. Chem Senses. 2006;31(8):747–52. https://pubmed.ncbi.nlm.nih.gov/16891352/
Erridge C. The capacity of foodstuffs to induce innate immune activation of human monocytes in vitro is dependent on food content of stimulants of Toll-like receptors 2 and 4. Br J Nutr. 2011;105(1):15–23. https://pubmed.ncbi.nlm.nih.gov/20849668/
Havlicek J, Lenochova P. The effect of meat consumption on body odor attractiveness. Chem Senses. 2006;31(8):747–52. https://pubmed.ncbi.nlm.nih.gov/16891352/
Scavello I, Maseroli E, Di Stasi V, Vignozzi L. Sexual health in menopause. Medicina (Kaunas). 2019;55(9):559. https://pubmed.ncbi.nlm.nih.gov/31480774/
Tiefer L. Female sexual dysfunction: a case study of disease mongering and activist resistance. PLoS Med. 2006;3(4):e178. https://pubmed.ncbi.nlm.nih.gov/16597176/
Angel K. The history of “Female Sexual Dysfunction” as a mental disorder in the 20th century. Curr Opin Psychiatry. 2010;23(6):536–41. https://pubmed.ncbi.nlm.nih.gov/20802336/
Meixel A, Yanchar E, Fugh-Berman A. Hypoactive sexual desire disorder: inventing a disease to sell low libido. J Med Ethics. 2015;41(10):859–62. https://pubmed.ncbi.nlm.nih.gov/26124287/
Jaspers L, Feys F, Bramer WM, Franco OH, Leusink P, Laan ETM. Efficacy and safety of flibanserin for the treatment of hypoactive sexual desire disorder in women: a systematic review and meta-analysis. JAMA Intern Med. 2016;176(4):453–62. https://pubmed.ncbi.nlm.nih.gov/26927498/
Woloshin S, Schwartz LM. US Food and Drug Administration approval of flibanserin: even the score does not add up. JAMA Intern Med. 2016;176(4):439–42. https://pubmed.ncbi.nlm.nih.gov/25423223/
Fugh-Berman A. Advise against flibanserin. Am J Nurs. 2016;116(3):13. https://pubmed.ncbi.nlm.nih.gov/26914033/
Moyad MA, Park K. What do most erectile dysfunction guidelines have in common? No evidence-based discussion or recommendation of heart-healthy lifestyle changes and/or Panax ginseng. Asian J Androl. 2012;14(6):830–41. https://pubmed.ncbi.nlm.nih.gov/23001440/
Maravilla KR, Heiman JR, Garland PA, et al. Dynamic MR imaging of the sexual arousal response in women. J Sex Marital Ther. 2003;29 Suppl 1:71–6. https://pubmed.ncbi.nlm.nih.gov/12735090/
Steinke EE. Sexual dysfunction in women with cardiovascular disease: what do we know? J Cardiovasc Nurs. 2010;25(2):151–8. https://pubmed.ncbi.nlm.nih.gov/20142751/
Towe M, La J, El-Khatib F, Roberts N, Yafi FA, Rubin R. Diet and female sexual health. Sex Med Rev. 2020;8(2):256–64. https://pubmed.ncbi.nlm.nih.gov/31669123/
Park K, Goldstein I, Andry C, Siroky MB, Krane RJ, Azadzoi KM. Vasculogenic female sexual dysfunction: the hemodynamic basis for vaginal engorgement insufficiency and clitoral erectile insufficiency. Int J Impot Res. 1997;9(1):27–37. https://pubmed.ncbi.nlm.nih.gov/9138056/
Fishbeck DW, Sebastiani AM. Comparative Anatomy: Manual of Vertebrate Dissection. 3rd ed. Morton Publishing; 2015. https://www.worldcat.org/title/910942083
Esposito K, Ciotola M, Maiorino MI, et al. Hyperlipidemia and sexual function in premenopausal women. J Sex Med. 2009;6(6):1696–703. https://pubmed.ncbi.nlm.nih.gov/19453904/
Duncan LE, Lewis C, Jenkins P, Pearson TA. Does hypertension and its pharmacotherapy affect the quality of sexual function in women? Am J Hypertens. 2000;13(6 Pt 1):640–7. https://pubmed.ncbi.nlm.nih.gov/10912747/
Baldassarre M, Alvisi S, Mancini I, et al. Impaired lipid profile is a risk factor for the development of sexual dysfunction in women. J Sex Med. 2016;13(1):46–54. https://pubmed.ncbi.nlm.nih.gov/26755086/
Esposito K, Ciotola M, Giugliano F, et al. Mediterranean diet improves sexual function in women with the metabolic syndrome. Int J Impot Res. 2007;19(5):486–91. https://pubmed.ncbi.nlm.nih.gov/17673936/
Levin RJ. The ins and outs of vaginal lubrication. Sex Relation Ther. 2003;18(4):509–13. https://www.tandfonline.com/doi/abs/10.1080/14681990310001609859
Fisher NDL, Hughes M, Gerhard-Herman M, Hollenberg NK. Flavanol-rich cocoa induces nitric-oxide-dependent vasodilation in healthy humans. J Hypertens. 2003;21(12):2281–6. https://pubmed.ncbi.nlm.nih.gov/14654748/
Salonia A, Fabbri F, Zanni G, et al. Chocolate and women’s sexual health: an intriguing correlation. J Sex Med. 2006;3(3):476–82. https://pubmed.ncbi.nlm.nih.gov/16681473/
Allouh MZ, Daradka HM, Al Barbarawi MM, Mustafa AG. Fresh onion juice enhanced copulatory behavior in male rats with and without paroxetine-induced sexual dysfunction. Exp Biol Med (Maywood). 2014;239(2):177–82. https://pubmed.ncbi.nlm.nih.gov/24302558/
Cai T, Gacci M, Mattivi F, et al. Apple consumption is related to better sexual quality of life in young women. Arch Gynecol Obstet. 2014;290(1):93–8. https://pubmed.ncbi.nlm.nih.gov/24518938/
Esposito K, Ciotola M, Giugliano F, et al. Mediterranean diet improves sexual function in women with the metabolic syndrome. Int J Impot Res. 2007;19(5):486–91. https://pubmed.ncbi.nlm.nih.gov/17673936/
Esposito K, Ciotola M, Giugliano F, et al. Mediterranean diet improves erectile function in subjects with the metabolic syndrome. Int J Impot Res. 2006;18(4):405–10. https://pubmed.ncbi.nlm.nih.gov/16395320/
Wang F, Dai S, Wang M, Morrison H. Erectile dysfunction and fruit/vegetable consumption among diabetic Canadian men. Urology. 2013;82(6):1330–5. https://pubmed.ncbi.nlm.nih.gov/24295250/
Maiorino MI, Bellastella G, Giugliano D, Esposito K. From inflammation to sexual dysfunctions: a journey through diabetes, obesity, and metabolic syndrome. J Endocrinol Invest. 2018;41(11):1249–58. https://pubmed.ncbi.nlm.nih.gov/29549630/
Wadden TA, West DS, Delahanty LM, et al. The Look AHEAD study: a description of the lifestyle intervention and the evidence supporting it. Obesity (Silver Spring). 2006;14(5):737–52. https://pubmed.ncbi.nlm.nih.gov/16855180/
Belalcazar LM, Haffner SM, Lang W, et al. Lifestyle intervention and/or statins for the reduction of C-reactive protein in type 2 diabetes: from the Look AHEAD study. Obesity (Silver Spring). 2013;21(5):944–50. https://pubmed.ncbi.nlm.nih.gov/23512860/
Wing RR, Bond DS, Gendrano IN, et al. Effect of intensive lifestyle intervention on sexual dysfunction in women with type 2 diabetes: results from an ancillary Look AHEAD study. Diabetes Care. 2013;36(10):2937–44. https://pubmed.ncbi.nlm.nih.gov/23757437/
Esposito K, Nappo F, Giugliano F, et al. Meal modulation of circulating interleukin 18 and adiponectin concentrations in healthy subjects and in patients with type 2 diabetes mellitus. Am J Clin Nutr. 2003;78(6):1135–40. https://pubmed.ncbi.nlm.nih.gov/14668275/
Blankenberg S, Tiret L, Bickel C, et al. Interleukin-18 is a strong predictor of cardiovascular death in stable and unstable angina. Circulation. 2002;106(1):24–30. https://pubmed.ncbi.nlm.nih.gov/12093765/
Levin RJ. The pharmacology of the human female orgasm – its biological and physiological backgrounds. Pharmacol Biochem Behav. 2014;121:62–70. https://pubmed.ncbi.nlm.nih.gov/24560912/
Heath RG. Pleasure and brain activity in man. Deep and surface electroencephalograms during orgasm. J Nerv Ment Dis. 1972;154(1):3–18. https://pubmed.ncbi.nlm.nih.gov/5007439/
Serrano SE, Braun J, Trasande L, Dills R, Sathyanarayana S. Phthalates and diet: a review of the food monitoring and epidemiology data. Environ Health. 2014;13(1):43. https://pubmed.ncbi.nlm.nih.gov/24894065/
Main KM, Mortensen GK, Kaleva MM, et al. Human breast milk contamination with phthalates and alterations of endogenous reproductive hormones in infants three months of age. Environ Health Perspect. 2006;114(2):270–6. https://pubmed.ncbi.nlm.nih.gov/16451866/
Swan SH, Liu F, Hines M, et al. Prenatal phthalate exposure and reduced masculine play in boys. Int J Androl. 2010;33(2):259–69. https://pubmed.ncbi.nlm.nih.gov/19919614/
Watkins DJ, Téllez-Rojo MM, Ferguson KK, et al. In utero and peripubertal exposure to phthalates and BPA in relation to female sexual maturation. Environ Res. 2014;134:233–41. https://pubmed.ncbi.nlm.nih.gov/28637469/
Chen FP, Chien MH. Lower concentrations of phthalates induce proliferation in human breast cancer cells. Climacteric. 2014;17(4):377–84. https://pubmed.ncbi.nlm.nih.gov/24228746/
Desdoits-Lethimonier C, Albert O, Le Bizec B, et al. Human testis steroidogenesis is inhibited by phthalates. Hum Reprod. 2012;27(5):1451–9. https://pubmed.ncbi.nlm.nih.gov/22402212/
Barrett ES, Parlett LE, Wang C, Drobnis EZ, Redmon JB, Swan SH. Environmental exposure to di-2-ethylhexyl phthalate is associated with low interest in sexual activity in premenopausal women. Horm Behav. 2014;66(5):787–92. https://pubmed.ncbi.nlm.nih.gov/25448532/
Koch HM, Lorber M, Christensen KLY, Pälmke C, Koslitz S, Brüning T. Identifying sources of phthalate exposure with human biomonitoring: results of a 48h fasting study with urine collection and personal activity patterns. Int J Hyg Environ Health. 2013;216(6):672–81. https://pubmed.ncbi.nlm.nih.gov/23333758/
Ji K, Lim Kho Y, Park Y, Choi K. Influence of a five-day vegetarian diet on urinary levels of antibiotics and phthalate metabolites: a pilot study with “Temple Stay” participants. Environ Res. 2010;110(4):375–82. https://pubmed.ncbi.nlm.nih.gov/20227070/
Serrano SE, Braun J, Trasande L, Dills R, Sathyanarayana S. Phthalates and diet: a review of the food monitoring and epidemiology data. Environ Health. 2014;13(1):43. https://pubmed.ncbi.nlm.nih.gov/24894065/
Schecter A, Lorber M, Guo Y, et al. Phthalate concentrations and dietary exposure from food purchased in New York State. Environ Health Perspect. 2013;121(4):473–9. https://pubmed.ncbi.nlm.nih.gov/23461894/
Serrano SE, Braun J, Trasande L, Dills R, Sathyanarayana S. Phthalates and diet: a review of the food monitoring and epidemiology data. Environ Health. 2014;13(1):43. https://pubmed.ncbi.nlm.nih.gov/24894065/
Koch HM, Lorber M, Christensen KLY, Pälmke C, Koslitz S, Brüning T. Identifying sources of phthalate exposure with human biomonitoring: results of a 48h fasting study with urine collection and personal activity patterns. Int J Hyg Environ Health. 2013;216(6):672–81. https://pubmed.ncbi.nlm.nih.gov/23333758/
Braun JM, Sathyanarayana S, Hauser R. Phthalate exposure and children’s health. Curr Opin Pediatr. 2013;25(2):247–54. https://pubmed.ncbi.nlm.nih.gov/23429708/
CPSC prohibits certain phthalates in children’s toys and child care products. United States Consumer Product Safety Commission. https://www.cpsc.gov/content/cpsc-prohibits-certain-phthalates-in-children%E2%80%99s-toys-and-child-care-products. Published October 20, 2017. Accessed August 24, 2022.; https://www.cpsc.gov/content/cpsc-prohibits-certain-phthalates-in-children%E2%80%99s-toys-and-child-care-products
Nilsson NH, Malmgren-Hansen B, Bernth N, Pedersen E, Pommer K. Survey and health assesment of chemicals substances in sex toys. Survey of Chemical Substances in Consumer Products. https://www2.mst.dk/udgiv/publications/2006/87–7052–227–8/pdf/87–7052–228–6.pdf. Published September 8, 2006. Accessed August 24, 2022.; https://www2.mst.dk/udgiv/publications/2006/87-7052-227-8/pdf/87-7052-228-6.pdf
Rao A, Steels E, Beccaria G, Inder WJ, Vitetta L. Influence of a specialized Trigonella foenum-graecum seed extract (libifem), on testosterone, estradiol and sexual function in healthy menstruating women, a randomised placebo controlled study. Phytother Res. 2015;29(8):1123–30. https://pubmed.ncbi.nlm.nih.gov/25914334/
Laughlin GA, Barrett-Connor E, Kritz-Silverstein D, von Mühlen D. Hysterectomy, oophorectomy, and endogenous sex hormone levels in older women: the Rancho Bernardo Study. J Clin Endocrinol Metab. 2000;85(2):645–51. https://pubmed.ncbi.nlm.nih.gov/10690870/
Simon JA, Kapner MD. The saga of testosterone for menopausal women at the Food and Drug Administration (FDA). J Sex Med. 2020;17(4):826–9. https://pubmed.ncbi.nlm.nih.gov/32253005/
Randolph JF, Zheng H, Avis NE, Greendale GA, Harlow SD. Masturbation frequency and sexual function domains are associated with serum reproductive hormone levels across the menopausal transition. J Clin Endocrinol Metab. 2015;100(1):258–66. https://pubmed.ncbi.nlm.nih.gov/25412335/
Smith T, Batur P. Prescribing testosterone and DHEA: the role of androgens in women. Cleve Clin J Med. 2021;88(1):35–43. https://pubmed.ncbi.nlm.nih.gov/33384313/
Davis SR, Baber R, Panay N, et al. Global consensus position statement on the use of testosterone therapy for women. Climacteric. 2019;22(5):429–34. https://pubmed.ncbi.nlm.nih.gov/31498871/
Pinkerton JV, Blackman I, Conner EA, Kaunitz AM. Risks of testosterone for postmenopausal women. Endocrinol Metab Clin North Am. 2021;50(1):139–50. https://pubmed.ncbi.nlm.nih.gov/33518182/
Davis SR, Baber R, Panay N, et al. Global consensus position statement on the use of testosterone therapy for women. Climacteric. 2019;22(5):429–34. https://pubmed.ncbi.nlm.nih.gov/31498871/
Fukui H, Yamashita M. The effects of music and visual stress on testosterone and cortisol in men and women. Neuro Endocrinol Lett. 2003;24(3–4):173–80. https://pubmed.ncbi.nlm.nih.gov/14523353/
Akdogan M, Tamer MN, Cüre E, Cüre MC, Köroglu BK, Delibas N. Effect of spearmint (Mentha spicata Labiatae) teas on androgen levels in women with hirsutism. Phytother Res. 2007;21(5):444-7. https://pubmed.ncbi.nlm.nih.gov/17310494/
Nikjou R, Kazemzadeh R, Asadzadeh F, Fathi R, Mostafazadeh F. The effect of lavender aromatherapy on the symptoms of menopause. J Natl Med Assoc. 2018;110(3):265–9. https://pubmed.ncbi.nlm.nih.gov/29778129/
Choi SY, Kang P, Lee HS, Seol GH. Effects of inhalation of essential oil of Citrus aurantium L. var. amara on menopausal symptoms, stress, and estrogen in postmenopausal women: a randomized controlled trial. Evid Based Complement Alternat Med. 2014;2014:796518. https://pubmed.ncbi.nlm.nih.gov/25024731/
Ghorbani Z, Mirghafourvand M. A meta-analysis of the efficacy of panax ginseng on menopausal women’s sexual function. Int J Womens Health Reprod Sci. 2018;7(1):124–33.https//ijwhr.net/text.php?id=399
Brooks NA, Wilcox G, Walker KZ, Ashton JF, Cox MB, Stojanovska L. Beneficial effects of Lepidium meyenii (Maca) on psychological symptoms and measures of sexual dysfunction in postmenopausal women are not related to estrogen or androgen content. Menopause. 2008;15(6):1157–62. https://pubmed.ncbi.nlm.nih.gov/18784609/
Paul S, Chakraborty S, Anand U, et al. Withania somnifera (L.) Dunal (Ashwagandha): a comprehensive review on ethnopharmacology, pharmacotherapeutics, biomedicinal and toxicological aspects. Biomed Pharmacother. 2021;143:112175. https://pubmed.ncbi.nlm.nih.gov/34649336/
Mandlik (Ingawale) DS, Namdeo AG. Pharmacological evaluation of Ashwagandha highlighting its healthcare claims, safety, and toxicity aspects. J Diet Suppl. 2021;18(2):183–226. https://pubmed.ncbi.nlm.nih.gov/32242751/
Dongre S, Langade D, Bhattacharyya S. Efficacy and safety of ashwagandha (Withania somnifera) root extract in improving sexual function in women: a pilot study. Biomed Res Int. 2015;2015:284154. https://pubmed.ncbi.nlm.nih.gov/26504795/
Ashwagandha. In: LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. National Institute of Diabetes and Digestive and Kidney Diseases; 2012. https://pubmed.ncbi.nlm.nih.gov/31643176/
Simon JA, Lukas VA. Distressing sexual function at midlife: unmet needs, practical diagnoses, and available treatments. Obstet Gynecol. 2017;130(4):889–905. https://pubmed.ncbi.nlm.nih.gov/28885410/
Portman DJ, Gass MLS, Kingsburg S, et al. Genitourinary syndrome of menopause: new terminology for vulvovaginal atrophy from the International Society for the Study of Women’s Sexual Health and the North American Menopause Society. Menopause. 2014;21(10):1063–8. https://pubmed.ncbi.nlm.nih.gov/25160739/
Scavello I, Maseroli E, Di Stasi V, Vignozzi L. Sexual health in menopause. Medicina (Kaunas). 2019;55(9):559. https://pubmed.ncbi.nlm.nih.gov/31480774/
Minkin MJ. Menopause: hormones, lifestyle, and optimizing aging. Obstet Gynecol Clin North Am. 2019;46(3):501–14. https://pubmed.ncbi.nlm.nih.gov/31378291/
Faubion SS, Sood R, Kapoor E. Genitourinary syndrome of menopause: management strategies for the clinician. Mayo Clin Proc. 2017;92(12):1842–9. https://pubmed.ncbi.nlm.nih.gov/29202940/
Scavello I, Maseroli E, Di Stasi V, Vignozzi L. Sexual health in menopause. Medicina (Kaunas). 2019;55(9):559. https://pubmed.ncbi.nlm.nih.gov/31480774/
Faubion SS, Sood R, Kapoor E. Genitourinary syndrome of menopause: management strategies for the clinician. Mayo Clin Proc. 2017;92(12):1842–9. https://pubmed.ncbi.nlm.nih.gov/29202940/
Herbenick D, Reece M, Hensel D, Sanders S, Jozkowski K, Fortenberry JD. Association of lubricant use with women’s sexual pleasure, sexual satisfaction, and genital symptoms: a prospective daily diary study. J Sex Med. 2011;8(1):202–12. https://pubmed.ncbi.nlm.nih.gov/21143591/
Mitchell CM, Reed SD, Diem S, et al. Efficacy of vaginal estradiol or vaginal moisturizer vs placebo for treating postmenopausal vulvovaginal symptoms: a randomized clinical trial. JAMA Intern Med. 2018;178(5):681–90. https://pubmed.ncbi.nlm.nih.gov/29554173/
Huang AJ, Grady D. Rethinking the approach to managing postmenopausal vulvovaginal symptoms. JAMA Intern Med. 2018;178(5):690–1. https://pubmed.ncbi.nlm.nih.gov/29554180/
Edwards D, Panay N. Treating vulvovaginal atrophy/genitourinary syndrome of menopause: how important is vaginal lubricant and moisturizer composition? Climacteric. 2016;19(2):151–61. https://pubmed.ncbi.nlm.nih.gov/26707589/
Adriaens E, Remon JP. Mucosal irritation potential of personal lubricants relates to product osmolality as detected by the slug mucosal irritation assay. Sex Transm Dis. 2008;35(5):512–6. https://pubmed.ncbi.nlm.nih.gov/18356773/
Edwards D, Panay N. Treating vulvovaginal atrophy/genitourinary syndrome of menopause: how important is vaginal lubricant and moisturizer composition? Climacteric. 2016;19(2):151–61. https://pubmed.ncbi.nlm.nih.gov/26707589/
Edwards D, Panay N. Treating vulvovaginal atrophy/genitourinary syndrome of menopause: how important is vaginal lubricant and moisturizer composition? Climacteric. 2016;19(2):151–61. https://pubmed.ncbi.nlm.nih.gov/26707589/
Scavello I, Maseroli E, Di Stasi V, Vignozzi L. Sexual health in menopause. Medicina (Kaunas). 2019;55(9):559. https://pubmed.ncbi.nlm.nih.gov/31480774/
Faubion SS, Sood R, Kapoor E. Genitourinary syndrome of menopause: management strategies for the clinician. Mayo Clin Proc. 2017;92(12):1842–9. https://pubmed.ncbi.nlm.nih.gov/29202940/
Cardozo L, Bachmann G, McClish D, Fonda D, Birgerson L. Meta-analysis of estrogen therapy in the management of urogenital atrophy in postmenopausal women: second report of the Hormones and Urogenital Therapy Committee. Obstet Gynecol. 1998;92(4 Pt 2):722–7. https://pubmed.ncbi.nlm.nih.gov/9764689/
Faubion SS, Sood R, Kapoor E. Genitourinary syndrome of menopause: management strategies for the clinician. Mayo Clin Proc. 2017;92(12):1842–9. https://pubmed.ncbi.nlm.nih.gov/29202940/
Lethaby A, Ayeleke RO, Roberts H. Local oestrogen for vaginal atrophy in postmenopausal women. Cochrane Database Syst Rev. 2016;(8):CD001500. https://pubmed.ncbi.nlm.nih.gov/27577677/
. Šimunic V, Banovic I, Ciglar S, Jeren L, Pavicic Baldani D, Šprem M. Local estrogen treatment in patients with urogenital symptoms. Int J Gynaecol Obstet. 2003;82(2):187–97. https://pubmed.ncbi.nlm.nih.gov/12873780/
Mitchell CM, Reed SD, Diem S, et al. Efficacy of vaginal estradiol or vaginal moisturizer vs placebo for treating postmenopausal vulvovaginal symptoms: a randomized clinical trial. JAMA Intern Med. 2018;178(5):681–90. https://pubmed.ncbi.nlm.nih.gov/29554173/
Pinkerton JV. Hormone therapy for postmenopausal women. N Engl J Med. 2020;382(5):446–5. https://pubmed.ncbi.nlm.nih.gov/31995690/
Premarin® Vaginal Cream Boxed Warning (conjugated estrogens). Pfizer. https://www.pfizermedicalinformation.com/en-us/premarin-vaginal-cream/boxed-warning. Updated September 2018. Accessed August 24, 2022.; https://www.pfizermedicalinformation.com/en-us/premarin-vaginal-cream/boxed-warning
Pinkerton JV, Kaunitz AM, Manson JE. Vaginal estrogen in the treatment of genitourinary syndrome of menopause and risk of endometrial cancer: an assessment of recent studies provides reassurance. Menopause. 2017;24(12):1329–32. https://pubmed.ncbi.nlm.nih.gov/29040220/
Bhupathiraju SN, Grodstein F, Stampfer MJ, et al. Vaginal estrogen use and chronic disease risk in the Nurses’ Health Study. Menopause. 2018;26(6):603–10. https://pubmed.ncbi.nlm.nih.gov/30562320/
Crandall CJ, Diamant A, Santoro N. Safety of vaginal estrogens: a systematic review. Menopause. 2020;27(3):339–60. https://pubmed.ncbi.nlm.nih.gov/31913230/
Kelsey JL, LiVolsi VA, Holford TR, et al. A case-control study of cancer of the endometrium. Am J Epidemiol. 1982;116(2):333–42. https://pubmed.ncbi.nlm.nih.gov/7114042/
Mørch LS, Kjaer SK, Keiding N, Løkkegaard E, Lidegaard Ø. The influence of hormone therapies on type I and II endometrial cancer: a nationwide cohort study. Int J Cancer. 2016;138(6):1506–15. https://pubmed.ncbi.nlm.nih.gov/26421912/
Pinkerton JV, Kaunitz AM, Manson JE. Vaginal estrogen in the treatment of genitourinary syndrome of menopause and risk of endometrial cancer: an assessment of recent studies provides reassurance. Menopause. 2017;24(12):1329–32. https://pubmed.ncbi.nlm.nih.gov/29040220/
Scavello I, Maseroli E, Di Stasi V, Vignozzi L. Sexual health in menopause. Medicina (Kaunas). 2019;55(9):559. https://pubmed.ncbi.nlm.nih.gov/31480774/
Eden JA. DHEA replacement for postmenopausal women: placebo or panacea? Climacteric. 2015;18(4):439–40. https://pubmed.ncbi.nlm.nih.gov/25731680/
Elraiyah T, Sonbol MB, Wang Z, et al. Clinical review: the benefits and harms of systemic dehydroepiandrosterone (DHEA) in postmenopausal women with normal adrenal function: a systematic review and meta-analysis. J Clin Endocrinol Metab. 2014;99(10):3536–42. https://pubmed.ncbi.nlm.nih.gov/25279571/
U.S. Food and Drug Administration. FDA approves Intrarosa for postmenopausal women experiencing pain during sex. FDA.gov. https://www.fda.gov/news-events/press-announcements/fda-approves-intrarosa-postmenopausal-women-experiencing-pain-during-sex. Published November 17, 2016. Accessed Sept 26, 2022.; https://www.fda.gov/news-events/press-announcements/fda-approves-intrarosa-postmenopausal-women-experiencing-pain-during-sex
Labrie F, Martel C, Bérubé R, et al. Intravaginal prasterone (DHEA) provides local action without clinically significant changes in serum concentrations of estrogens or androgens. J Steroid Biochem Mol Biol. 2013;138:359–67. https://pubmed.ncbi.nlm.nih.gov/23954500/
Faubion SS, Sood R, Kapoor E. Genitourinary syndrome of menopause: management strategies for the clinician. Mayo Clin Proc. 2017;92(12):1842–9. https://pubmed.ncbi.nlm.nih.gov/29202940/
Di Donato V, Schiavi MC, Iacobelli V, et al. Ospemifene for the treatment of vulvar and vaginal atrophy: a meta-analysis of randomized trials. Part II: evaluation of tolerability and safety. Maturitas. 2019;121:93–100. https://pubmed.ncbi.nlm.nih.gov/30509754/
Gold EB. Relation of demographic and lifestyle factors to symptoms in a multi-racial/ethnic population of women 40–55 years of age. Am Journal Epidemiol. 2000;152(5):463–73. https://pubmed.ncbi.nlm.nih.gov/10981461/
Ghazanfarpour M, Roudsari RL, Treglia G, Sadeghi R. Topical administration of isoflavones for treatment of vaginal symptoms in postmenopausal women: a systematic review of randomised controlled trials. J Obstet Gynaecol. 2015;35(8):783–7. https://pubmed.ncbi.nlm.nih.gov/25710207/
Lima SMRR, Bernardo BFA, Yamada SS, Reis BF, da Silva GMD, Galvão MAL. Effects of Glycine max (L.) Merr. soy isoflavone vaginal gel on epithelium morphology and estrogen receptor expression in postmenopausal women: a 12-week, randomized, double-blind, placebo-controlled trial. Maturitas. 2014;78(3):205–11. https://pubmed.ncbi.nlm.nih.gov/24856055/
Lima SMRR, Yamada SS, Reis BF, Postigo S, Galvão da Silva MAL, Aoki T. Effective treatment of vaginal atrophy with isoflavone vaginal gel. Maturitas. 2013;74(3):252–8. https://pubmed.ncbi.nlm.nih.gov/23312487/
Zhang J, Zhu Y, Pan L, Xia H, Ma J, Zhang A. Soy isoflavone improved female sexual dysfunction of mice via endothelial nitric oxide synthase pathway. Sex Med. 2019;7(3):345–51. https://pubmed.ncbi.nlm.nih.gov/31303464/
Nikander E, Rutanen EM, Nieminen P, Wahlström T, Ylikorkala O, Tiitinen A. Lack of effect of isoflavonoids on the vagina and endometrium in postmenopausal women. Fertil Steril. 2005;83(1):137–42. https://pubmed.ncbi.nlm.nih.gov/15652899/
Nourozi M, Haghollahi F, Ramezanzadeh F, Hanachi P. Effect of soy milk consumption on quality of life in Iranian postmenopausal women. J Family Reprod Health. 2015;9(2):93–100. https://pubmed.ncbi.nlm.nih.gov/26175764/
Hanachi P, Golkho S. The effect of soymilk on menopausal symptoms and total antioxidant levels in menopausal women. Malaysian J Med Health Sci. 2008;4(1):33–40. https://www.researchgate.net/publication/267037285_The_effect_of_soymilk_on_menopausal_symptoms_and_total_antioxidant_levels_in_menopausal_women
Padmaqriya S, Kumar SS. Quality of life of postmenopausal women receiving plant-based phytoestrogens. Int J Pharm Sci Res. 2020;11(10):4998–5003. https://ijpsr.com/bft-article/quality-of-life-of-postmenopausal-women-receiving-plant-based-phytoestrogens/
Amsterdam A, Abu-Rustum N, Carter J, Krychman M. Persistent sexual arousal syndrome associated with increased soy intake. J Sex Med. 2005;2(3):338–40. https://pubmed.ncbi.nlm.nih.gov/16422864/
Omidvar S, Esmailzadeh S, Baradaran M, Basirat Z. Effect of fennel on pain intensity in dysmenorrhoea: a placebo-controlled trial. Ayu. 2012;33(2):311–3. https://pubmed.ncbi.nlm.nih.gov/23559811/
Modaress Nejad V, Asadipour M. Comparison of the effectiveness of fennel and mefenamic acid on pain intensity in dysmenorrhoea. East Mediterr Health J. 2006;12(3–4):423–7. https://pubmed.ncbi.nlm.nih.gov/17037712/
Ghazanfarpour M, Shokrollahi P, Khadivzadeh T, et al. Effect of Foeniculum vulgare (fennel) on vaginal atrophy in postmenopausal women: a double-blind, randomized, placebo-controlled trial. Post Reprod Health. 2017;23(4):171–6. https://pubmed.ncbi.nlm.nih.gov/28990439/
Ghaffari P, Hosseininik M, Afrasiabifar A, et al. The effect of Fennel seed powder on estradiol levels, menopausal symptoms, and sexual desire in postmenopausal women. Menopause. 2020;27(11):1281–6. https://pubmed.ncbi.nlm.nih.gov/33110044/
Yaralizadeh M, Abedi P, Najar S, Namjoyan F, Saki A. Effect of Foeniculum vulgare (fennel) vaginal cream on vaginal atrophy in postmenopausal women: a double-blind randomized placebo-controlled trial. Maturitas. 2016;84:75–80. https://pubmed.ncbi.nlm.nih.gov/26617271/
Mazalzadeh F, Hekmat K, Namjouyan F, Saki A. Effect of Trigonella foenum (fenugreek) vaginal cream on vaginal atrophy in postmenopausal women. J Family Med Prim Care. 2020;9(6):2714–9. https://pubmed.ncbi.nlm.nih.gov/32984113/
Abedi P, Najafian M, Yaralizadeh M, Namjoyan F. Effect of fennel vaginal cream on sexual function in postmenopausal women: a double blind randomized controlled trial. J Med Life. 2018;11(1):24–8. https://pubmed.ncbi.nlm.nih.gov/29696061/
Poole C, Bushey B, Foster C, et al. The effects of a commercially available botanical supplement on strength, body composition, power output, and hormonal profiles in resistance-trained males. J Int Soc Sports Nutr. 2010;7:34. https://pubmed.ncbi.nlm.nih.gov/20979623/
Mansoori A, Hosseini S, Zilaee M, Hormoznejad R, Fathi M. Effect of fenugreek extract supplement on testosterone levels in male: a meta-analysis of clinical trials. Phytother Res. 2020;34(7):1550–5. https://pubmed.ncbi.nlm.nih.gov/32048383/
Rao A, Steels E, Inder WJ, Abraham S, Vitetta L. Testofen, a specialised Trigonella foenum-graecum seed extract reduces age-related symptoms of androgen decrease, increases testosterone levels and improves sexual function in healthy aging males in a double-blind randomised clinical study. Aging Male. 2016;19(2):134–42. https://pubmed.ncbi.nlm.nih.gov/26791805/
Bahmani M, Shirzad H, Mirhosseini M, Mesripour A, Rafieian-Kopaei M. A review on ethnobotanical and therapeutic uses of fenugreek (Trigonella foenum-graceum L). J Evid Based Complementary Altern Med. 2016;21(1):53–62. https://pubmed.ncbi.nlm.nih.gov/25922446/
Rao A, Steels E, Beccaria G, Inder WJ, Vitetta L. Influence of a specialized Trigonella foenum-graecum seed extract (libifem), on testosterone, estradiol and sexual function in healthy menstruating women, a randomised placebo controlled study. Phytother Res. 2015;29(8):1123–30. https://pubmed.ncbi.nlm.nih.gov/25914334/
Steels E, Steele ML, Harold M, Coulson S. Efficacy of a proprietary Trigonella foenum-graecum L. de-husked seed extract in reducing menopausal symptoms in otherwise healthy women: a double-blind, randomized, placebo-controlled study. Phytother Res. 2017;31(9):1316–22. https://pubmed.ncbi.nlm.nih.gov/28707431/
Safary M, Hakimi S, Mobaraki-Asl N, Amiri P, Tvassoli H, Delazar A. Comparison of the effects of fenugreek vaginal cream and ultra low-dose estrogen on atrophic vaginitis. Curr Drug Deliv. 2020;17(9):815–22. https://pubmed.ncbi.nlm.nih.gov/32640956/
Pill-free ways to improve your sex life. Exercise, smoking cessation, and alcohol moderation can help bring sexual activity back into the bedroom. Harv Health Lett. 2014;39(10):4. https://pubmed.ncbi.nlm.nih.gov/25230408/
Hall SA, Shackelton R, Rosen RC, Araujo AB. Sexual activity, erectile dysfunction, and incident cardiovascular events. Am J Cardiol. 2010;105(2):192–7. https://pubmed.ncbi.nlm.nih.gov/20102917/
Davey Smith G, Frankel S, Yarnell J. Sex and death: are they related? Findings from the Caerphilly Cohort Study. BMJ. 1997;315(7123):1641–4. https://pubmed.ncbi.nlm.nih.gov/9448525/
Fisher AD, Bandini E, Rastrelli G, et al. Sexual and cardiovascular correlates of male unfaithfulness. J Sex Med. 2012;9(6):1508–18. https://pubmed.ncbi.nlm.nih.gov/22510301/
Maggi M, Corona G. Love protects lover’s life. J Sex Med. 2011;8(4):931–5. https://pubmed.ncbi.nlm.nih.gov/21457464/
Davey Smith G, Frankel S, Yarnell J. Sex and death: are they related? Findings from the Caerphilly Cohort Study. BMJ. 1997;315(7123):1641–4. https://pubmed.ncbi.nlm.nih.gov/9448525/
Meldrum DR, Gambone JC, Morris MA, Meldrum DA, Esposito K, Ignarro LJ. The link between erectile and cardiovascular health: the canary in the coal mine. Am J Cardiol. 2011;108(4):599–606. https://pubmed.ncbi.nlm.nih.gov/21624550/
Esposito K, Giugliano D. Lifestyle/dietary recommendations for erectile dysfunction and female sexual dysfunction. Urol Clin North Am. 2011;38(3):293–301. https://pubmed.ncbi.nlm.nih.gov/21798391/
Dong JY, Zhang YH, Qin LQ. Erectile dysfunction and risk of cardiovascular disease: meta-analysis of prospective cohort studies. J Am Coll Cardiol. 2011;58(13):1378–85. https://pubmed.ncbi.nlm.nih.gov/21920268/
Ostfeld RJ, Allen KE, Aspry K, et al. Vasculogenic erectile dysfunction: the impact of diet and lifestyle. Am J Med. 2021;134(3):310–6. https://pubmed.ncbi.nlm.nih.gov/33227246/
Kent KC, Zwolak RM, Egorova NN, et al. Analysis of risk factors for abdominal aortic aneurysm in a cohort of more than 3 million individuals. J Vasc Surg. 2010;52(3):539–48. https://pubmed.ncbi.nlm.nih.gov/20630687/
Two-way street between erection problems and heart disease. Paying attention to heart health can be good for a man’s sex life. Harv Heart Lett. 2011;21(9):4. https://pubmed.ncbi.nlm.nih.gov/21649979/
Inman BA, Sauver JL, Jacobson DJ, et al. A population-based, longitudinal study of erectile dysfunction and future coronary artery disease. Mayo Clin Proc. 2009;84(2):108–13. https://pubmed.ncbi.nlm.nih.gov/19181643/
Geerkens MJM, Al-Itejawi HHM, Nieuwenhuijzen JA, et al. Sexual dysfunction and bother due to erectile dysfunction in the healthy elderly male population: prevalence from a systematic review. Eur Urol Focus. 2020;6(4):776–90. https://pubmed.ncbi.nlm.nih.gov/30878347/
Jackson G. Erectile dysfunction and coronary disease: evaluating the link. Maturitas. 2012;72(3):263–4. https://pubmed.ncbi.nlm.nih.gov/22503513/
Carto C, Pagalavan M, Nackeeran S, et al. Consumption of a healthy plant-based diet is associated with a decreased risk of erectile dysfunction: a cross-sectional study of the National Health and Nutrition Examination Survey. Urology. 2022;161:76–82. https://pubmed.ncbi.nlm.nih.gov/34979217/
Bauer SR, Breyer BN, Stampfer MJ, Rimm EB, Giovannucci EL, Kenfield SA. Association of diet with erectile dysfunction among men in the Health Professionals Follow-Up Study. JAMA Netw Open. 2020;3(11):e2021701. https://pubmed.ncbi.nlm.nih.gov/33185675/
Wang F, Dai S, Wang M, Morrison H. Erectile dysfunction and fruit/vegetable consumption among diabetic Canadian men. Urology. 2013;82(6):1330–5. https://pubmed.ncbi.nlm.nih.gov/24295250/
Mykoniatis I, Grammatikopoulou MG, Bouras E, et al. Sexual dysfunction among young men: overview of dietary components associated with erectile dysfunction. J Sex Med. 2018;15(2):176–82. https://pubmed.ncbi.nlm.nih.gov/29325831/
Gilbert SF, Zevit Z. Congenital human baculum deficiency: the generative bone of Genesis 2:21–23. Am J Med Genet. 2001;101(3):284–5. https://pubmed.ncbi.nlm.nih.gov/11424148/
Dawkins R. The Selfish Gene. 30th Anniversary ed. Oxford University Press; 2006. https://worldcat.org/title/62532503
Huynh LM, Liang K, Osman MM, et al. Organic diet and intermittent fasting are associated with improved erectile function. Urology. 2020;144:147–51. https://pubmed.ncbi.nlm.nih.gov/32717247/
Burnett AL. Environmental erectile dysfunction: can the environment really be hazardous to your erectile health? J Androl. 2008;29(3):229–36. https://pubmed.ncbi.nlm.nih.gov/18187396/
Espir ML, Hall JW, Shirreffs JG, Stevens DL. Impotence in farm workers using toxic chemicals. Br Med J. 1970;1(5693):423–5. https://pubmed.ncbi.nlm.nih.gov/5434665/
Oliva A, Giami A, Multigner L. Environmental agents and erectile dysfunction: a study in a consulting population. J Androl. 2002;23(4):546–50. https://pubmed.ncbi.nlm.nih.gov/12065462/
van de Vijver LPL, van Vliet MET. Health effects of an organic diet – consumer experiences in the Netherlands. J Sci Food Agric. 2012;92(14):2923–7. https://pubmed.ncbi.nlm.nih.gov/22331850/
Li DK, Zhou Z, Miao M, et al. Relationship between urine bisphenol-A level and declining male sexual function. J Androl. 2010;31(5):500–6. https://pubmed.ncbi.nlm.nih.gov/20467048/
Geens T, Aerts D, Berthot C, et al. A review of dietary and non-dietary exposure to bisphenol-A. Food Chem Toxicol. 2012;50(10):3725–40. https://pubmed.ncbi.nlm.nih.gov/22889897/
Roberts R. BPA exposure and health effects: educating physicians and patients. Am Fam Physician. 2012;85(11):1040–4. https://pubmed.ncbi.nlm.nih.gov/22962873/
Mirmira P, Evans-Molina C. Bisphenol A, obesity, and type 2 diabetes mellitus: genuine concern or unnecessary preoccupation? Transl Res. 2014;164(1):13–21. https://pubmed.ncbi.nlm.nih.gov/24686036/
Feldman HA, Johannes CB, Derby CA, et al. Erectile dysfunction and coronary risk factors: prospective results from the Massachusetts Male Aging Study. Prev Med. 2000;30(4):328–38. https://pubmed.ncbi.nlm.nih.gov/10731462/
Juenemann KP, Lue TF, Luo JA, Benowitz NL, Abozeid M, Tanagho EA. The effect of cigarette smoking on penile erection. J Urol. 1987;138(2):438–41. https://pubmed.ncbi.nlm.nih.gov/3599273/
Chan SSC, Leung DYP, Abdullah ASM, et al. Smoking-cessation and adherence intervention among Chinese patients with erectile dysfunction. Am J Prev Med. 2010;39(3):251–8. https://pubmed.ncbi.nlm.nih.gov/20709257/
Pizzol D, Demurtas J, Stubbs B, et al. Relationship between cannabis use and erectile dysfunction: a systematic review and meta-analysis. Am J Mens Health. 2019;13(6):1557988319892464. https://pubmed.ncbi.nlm.nih.gov/31795801/
Dallal RM, Chernoff A, O’Leary MP, Smith JA, Braverman JD, Quebbemann BB. Sexual dysfunction is common in the morbidly obese male and improves after gastric bypass surgery. J Am Coll Surg. 2008;207(6):859–64. https://pubmed.ncbi.nlm.nih.gov/19183532/
Khoo J, Piantadosi C, Duncan R, et al. Comparing effects of a low-energy diet and a high-protein low-fat diet on sexual and endothelial function, urinary tract symptoms, and inflammation in obese diabetic men. J Sex Med. 2011;8(10):2868–75. https://pubmed.ncbi.nlm.nih.gov/21819545/
Glina FPA, de Freitas Barboza JW, Nunes VM, Glina S, Bernardo WM. What is the impact of bariatric surgery on erectile function? A systematic review and meta-analysis. Sex Med Rev. 2017;5(3):393–402. https://pubmed.ncbi.nlm.nih.gov/28526630/
Allen MS, Walter EE. Health-related lifestyle factors and sexual dysfunction: a meta-analysis of population-based research. J Sex Med. 2018;15(4):458–75. https://pubmed.ncbi.nlm.nih.gov/29523476/
Silva AB, Sousa N, Azevedo LF, Martins C. Physical activity and exercise for erectile dysfunction: systematic review and meta-analysis. Br J Sports Med. 2017;51(19):1419–24. https://pubmed.ncbi.nlm.nih.gov/27707739/
Li J, Peng L, Cao D, He L, Li Y, Wei Q. Avanafil for the treatment of men with erectile dysfunction: a systematic review and meta-analysis of randomized controlled trials. Am J Mens Health. 2019;13(5):1557988319880764. https://pubmed.ncbi.nlm.nih.gov/31672076/
Gerbild H, Larsen CM, Graugaard C, Josefsson KA. Physical activity to improve erectile function: a systematic review of intervention studies. Sex Med. 2018;6(2):75–89. https://pubmed.ncbi.nlm.nih.gov/29661646/
Michiels M, Van der Aa F. Bicycle riding and the bedroom: can riding a bicycle cause erectile dysfunction? Urology. 2015;85(4):725–30. https://pubmed.ncbi.nlm.nih.gov/25681833/
Number of participants in bicycling in the United States from 2006 to 2020 (in millions). Statista. https://www.statista.com/statistics/191204/participants-in-bicycling-in-the-us-since-2006/. Published June 28, 2021. Accessed August 24, 2022.; https://www.statista.com/statistics/191204/participants-in-bicycling-in-the-us-since-2006/
Gan ZS, Ehlers ME, Lin FC, Wright ST, Figler BD, Coward RM. Systematic review and meta-analysis of cycling and erectile dysfunction. Sex Med Rev. 2021;9(2):304–11. https://pubmed.ncbi.nlm.nih.gov/32147498/
Michiels M, Van der Aa F. Bicycle riding and the bedroom: can riding a bicycle cause erectile dysfunction? Urology. 2015;85(4):725–30. https://pubmed.ncbi.nlm.nih.gov/25681833/
Dettori JR, Koepsell TD, Cummings P, Corman JM. Erectile dysfunction after a long-distance cycling event: associations with bicycle characteristics. J Urol. 2004;172(2):637–41. https://pubmed.ncbi.nlm.nih.gov/15247750/
Sommer F, Goldstein I, Korda JB. Bicycle riding and erectile dysfunction: a review. J Sex Med. 2010;7(7):2346–58. https://pubmed.ncbi.nlm.nih.gov/20102446/
Michiels M, Van der Aa F. Bicycle riding and the bedroom: can riding a bicycle cause erectile dysfunction? Urology. 2015;85(4):725–30. https://pubmed.ncbi.nlm.nih.gov/25681833/
Cai X, Tian Y, Wu T, Cao CX, Bu SY, Wang KJ. The role of statins in erectile dysfunction: a systematic review and meta-analysis. Asian J Androl. 2014;16(3):461–6. https://pubmed.ncbi.nlm.nih.gov/24556747/
Williams P, McBain H, Amirova A, Newman S, Mulligan K. Men’s beliefs about treatment for erectile dysfunction – what influences treatment use? A systematic review. Int J Impot Res. 2021;33(1):16–42. https://pubmed.ncbi.nlm.nih.gov/32231275/
Goldstein I. The hour lecture that changed sexual medicine – the Giles Brindley injection story. J Sex Med. 2012;9(2):337–42. https://pubmed.ncbi.nlm.nih.gov/22296605/
Barbas R, Llinas A, Prohens R. The solid state landscape of the sildenafil drug. J Pharm Sci. 2022;111(4):1104–9. https://pubmed.ncbi.nlm.nih.gov/34419482/
Klotz L. How (not) to communicate new scientific information: a memoir of the famous Brindley lecture. BJU Int. 2005;96(7):956–7. https://pubmed.ncbi.nlm.nih.gov/16225508/
Goldstein I. The hour lecture that changed sexual medicine – the Giles Brindley injection story. J Sex Med. 2012;9(2):337–42. https://pubmed.ncbi.nlm.nih.gov/22296605/
Ghofrani HA, Osterloh IH, Grimminger F. Sildenafil: from angina to erectile dysfunction to pulmonary hypertension and beyond. Nat Rev Drug Discov. 2006;5(8):689–702. https://pubmed.ncbi.nlm.nih.gov/16883306/
Williams P, McBain H, Amirova A, Newman S, Mulligan K. Men’s beliefs about treatment for erectile dysfunction – what influences treatment use? A systematic review. Int J Impot Res. 2021;33(1):16–42. https://pubmed.ncbi.nlm.nih.gov/32231275/
Souverein PC, Egberts ACG, Meuleman EJH, Urquhart J, Leufkens HGM. Incidence and determinants of sildenafil (dis)continuation: the Dutch cohort of sildenafil users. Int J Impot Res. 2002;14(4):259–65. https://pubmed.ncbi.nlm.nih.gov/12152115/
Salonia A, Rigatti P, Montorsi F. Sildenafil in erectile dysfunction: a critical review. Curr Med Res Opin. 2003;19(4):241–62. https://pubmed.ncbi.nlm.nih.gov/12841917/
Souverein PC, Egberts ACG, Meuleman EJH, Urquhart J, Leufkens HGM. Incidence and determinants of sildenafil (dis)continuation: the Dutch cohort of sildenafil users. Int J Impot Res. 2002;14(4):259–65. https://pubmed.ncbi.nlm.nih.gov/12152115/
Le B, Burnett AL. Evolution of penile prosthetic devices. Korean J Urol. 2015;56(3):179–86. https://pubmed.ncbi.nlm.nih.gov/25763121/
Schultheiss D, Gabouev AI, Jonas U. Nikolaj A. Bogoraz (1874–1952): pioneer of phalloplasty and penile implant surgery. J Sex Med. 2005;2(1):139–46. https://pubmed.ncbi.nlm.nih.gov/16422917/
Jain S, Bhojwani A, Terry TR. The role of penile prosthetic surgery in the modern management of erectile dysfunction. Postgrad Med J. 2000;76(891):22–5. https://pubmed.ncbi.nlm.nih.gov/10622775/
Ciftci H, Verit A, Savas M. Late complications of spontaneous urethral erosion of a malleable penile prosthesis in a young patient. Singapore Med J. 2012;53(6):e120–1. https://pubmed.ncbi.nlm.nih.gov/22711048/
Le B, Burnett AL. Evolution of penile prosthetic devices. Korean J Urol. 2015;56(3):179–86. https://pubmed.ncbi.nlm.nih.gov/25763121/
Chen L, Staubli SEL, Schneider MP, et al. Phosphodiesterase 5 inhibitors for the treatment of erectile dysfunction: a trade-off network meta-analysis. Eur Urol. 2015;68(4):674–80. https://pubmed.ncbi.nlm.nih.gov/25817916/
Matheeussen V, Maudens KE, Anseeuw K, Neels H. A non-fatal self-poisoning attempt with sildenafil. J Anal Toxicol. 2015;39(7):572–6. https://pubmed.ncbi.nlm.nih.gov/26139313/
Chen SP, Singh K, Lin SC. Use of phosphodiesterase inhibitors and prevalence of self-reported glaucoma in the United States. PLoS One. 2017;12(8):e0183388. https://pubmed.ncbi.nlm.nih.gov/28817686/
Kim SJ, Kim JH, Chang HK, Kim KH. Let’s rethinking about the safety of phosphodiesterase type 5 inhibitor in the patients with erectile dysfunction after radical prostatectomy. J Exerc Rehabil. 2016;12(3):143–7. https://pubmed.ncbi.nlm.nih.gov/27419107/
Mitra D, Robinson KC, Fisher DE. Melanoma and Viagra: an unexpected connection. Pigment Cell Melanoma Res. 2011;24(1):16–8. https://pubmed.ncbi.nlm.nih.gov/21290790/
Arozarena I, Sanchez-Laorden B, Packer L, et al. Oncogenic BRAF induces melanoma cell invasion by downregulating the cGMP-specific phosphodiesterase PDE5A. Cancer Cell. 2011;19(1):45–57. https://pubmed.ncbi.nlm.nih.gov/21215707/
Dhayade S, Kaesler S, Sinnberg T, et al. Sildenafil potentiates a cGMP-dependent pathway to promote melanoma growth. Cell Rep. 2016;14(11):2599–610. https://pubmed.ncbi.nlm.nih.gov/26971999/
Yafi FA, Sharlip ID, Becher EF. Update on the safety of phosphodiesterase type 5 inhibitors for the treatment of erectile dysfunction. Sex Med Rev. 2018;6(2):242–52. https://pubmed.ncbi.nlm.nih.gov/28923561/
Cohen PA, Venhuis BJ. Adulterated sexual enhancement supplements: more than mojo. JAMA Intern Med. 2013;173(13):1169–70. https://pubmed.ncbi.nlm.nih.gov/23699734/
Poon WT, Lam YH, Lee HHC, et al. Outbreak of hypoglycaemia: sexual enhancement products containing oral hypoglycaemic agent. Hong Kong Med J. 2009;15(3):196–200. https://pubmed.ncbi.nlm.nih.gov/19494375/
Jaksch F. Editorial: Are you concerned about the practice called “dry labbing” in the dietary supplement industry? Altern Med Rev. 2012;17(1):5. https://pubmed.ncbi.nlm.nih.gov/22502618/
Robbins R. A supplement maker tried to silence this Harvard doctor – and put academic freedom on trial. STAT. https://www.statnews.com/2017/01/10/supplement-harvard-pieter-cohen/. Published January 10, 2017. Accessed August 24, 2022.; https://www.statnews.com/2017/01/10/supplement-harvard-pieter-cohen/
Bagley N, Carroll AE, Cohen PA. Scientific trials – in the laboratories, not the courts. JAMA Intern Med. 2018;178(1):7–8. https://pubmed.ncbi.nlm.nih.gov/29114742/
Cohen PA, Bloszies C, Yee C, Gerona R. An amphetamine isomer whose efficacy and safety in humans has never been studied, ß-methylphenylethylamine (BMPEA), is found in multiple dietary supplements. Drug Test Anal. 2016;8(3–4):328–33. https://pubmed.ncbi.nlm.nih.gov/25847603/
Hi-Tech Pharms v Cohen, 16–10660-WGY (D Mass 2016).; https://www.govinfo.gov/app/details/USCOURTS-mad-1_16-cv-10660/
Bagley N, Carroll AE, Cohen PA. Scientific trials – in the laboratories, not the courts. JAMA Intern Med. 2018;178(1):7–8. https://pubmed.ncbi.nlm.nih.gov/29114742/
Bagley N, Carroll AE, Cohen PA. Scientific trials – in the laboratories, not the courts. JAMA Intern Med. 2018;178(1):7–8. https://pubmed.ncbi.nlm.nih.gov/29114742/
Hall-Lipsy E, Malanga S. Defamation lawsuits: academic sword or shield? EMBO Mol Med. 2017;9(12):1623–5. https://pubmed.ncbi.nlm.nih.gov/29038313/
Worner TM, Gordon GG, Leo MA, Lieber CS. Vitamin A treatment of sexual dysfunction in male alcoholics. Am J Clin Nutr. 1988;48(6):1431–5. https://pubmed.ncbi.nlm.nih.gov/3202091/
Ng CF, Lee CP, Ho AL, Lee VWY. Effect of niacin on erectile function in men suffering erectile dysfunction and dyslipidemia. J Sex Med. 2011;8(10):2883–93. https://pubmed.ncbi.nlm.nih.gov/21810191/
Biniaz V, Tayebi A, Ebadi A, Sadeghi S, Einollahi B. Effect of vitamin C supplementation on marital satisfaction in patients undergoing hemodialysis: a randomized, double-blind and placebo-controlled trial. Saudi J Kidney Dis Transpl. 2015;26(3):468–76. https://pubmed.ncbi.nlm.nih.gov/26022016/
Ghanbari-Homaie S, Ataei-Almanghadim K, Mirghafourvand M. Effect of vitamins on sexual function: a systematic review. Int J Vitam Nutr Res. Published online March 29, 2021:1–10.; https://pubmed.ncbi.nlm.nih.gov/33779240/
Elshahid ARM, Shahein IM, Mohammed YF, Ismail NF, Zakarria HBAER, GamalEl Din SF. Folic acid supplementation improves erectile function in patients with idiopathic vasculogenic erectile dysfunction by lowering peripheral and penile homocysteine plasma levels: a case-control study. Andrology. 2020;8(1):148–53. https://pubmed.ncbi.nlm.nih.gov/31237081/
Canguven O, Talib RA, El Ansari W, Yassin DJ, Al Naimi A. Vitamin D treatment improves levels of sexual hormones, metabolic parameters and erectile function in middle-aged vitamin D deficient men. Aging Male. 2017;20(1):9–16. https://pubmed.ncbi.nlm.nih.gov/28074679/
Balasubramanian A, Thirumavalavan N, Srivatsav A, et al. An analysis of popular online erectile dysfunction supplements. J Sex Med. 2019;16(6):843–52. https://pubmed.ncbi.nlm.nih.gov/31036522/
Srivatsav A, Balasubramanian A, Pathak UI, et al. Efficacy and safety of common ingredients in aphrodisiacs used for erectile dysfunction: a review. Sex Med Rev. 2020;8(3):431–42. https://pubmed.ncbi.nlm.nih.gov/32139335/
Jang DJ, Lee MS, Shin BC, Lee YC, Ernst E. Red ginseng for treating erectile dysfunction: a systematic review. Br J Clin Pharmacol. 2008;66(4):444–50. https://pubmed.ncbi.nlm.nih.gov/18754850/
Ichim MC, de Boer HJ. A review of authenticity and authentication of commercial ginseng herbal medicines and food supplements. Front Pharmacol. 2021;11:612071. https://pubmed.ncbi.nlm.nih.gov/33505315/
West E, Krychman M. Natural aphrodisiacs – a review of selected sexual enhancers. Sex Med Rev. 2015;3(4):279–88. https://pubmed.ncbi.nlm.nih.gov/27784600/
Zhu L, Han X, Zhu J, Du L, Liu L, Gong W. Severe acute intoxication with yohimbine: four simultaneous poisoning cases. Forensic Sci Int. 2021;320:110705. https://pubmed.ncbi.nlm.nih.gov/33529997/
Muncey W, Sellke N, Kim T, Mishra K, Thirumavalavan N, Loeb A. Alternative treatment for erectile dysfunction: a growing arsenal in men’s health. Curr Urol Rep. 2021;22(2):11. https://pubmed.ncbi.nlm.nih.gov/33420972/
Burnett AL, Nehra A, Breau RH, et al. Erectile dysfunction: AUA guideline. J Urol. 2018;200(3):633–41. https://pubmed.ncbi.nlm.nih.gov/29746858/
Hatzimouratidis K, Amar E, Eardley I, et al. Guidelines on male sexual dysfunction: erectile dysfunction and premature ejaculation. Eur Urol. 2010;57(5):804–14. https://pubmed.ncbi.nlm.nih.gov/20189712/
Maio G, Saraeb S, Marchiori A. Physical activity and PDE5 inhibitors in the treatment of erectile dysfunction: results of a randomized controlled study. J Sex Med. 2010;7(6):2201–8. https://pubmed.ncbi.nlm.nih.gov/20367777/
Esposito K, Ciotola M, Giugliano F, et al. Mediterranean diet improves erectile function in subjects with the metabolic syndrome. Int J Impot Res. 2006;18(4):405–10. https://pubmed.ncbi.nlm.nih.gov/16395320/
Meldrum DR, Gambone JC, Morris MA, Meldrum DAN, Esposito K, Ignarro LJ. The link between erectile and cardiovascular health: the canary in the coal mine. Am J Cardiol. 2011;108(4):599–606. https://pubmed.ncbi.nlm.nih.gov/21624550/
Gupta BP, Murad MH, Clifton MM, Prokop L, Nehra A, Kopecky SL. The effect of lifestyle modification and cardiovascular risk factor reduction on erectile dysfunction: a systematic review and meta-analysis. Arch Intern Med. 2011;171(20):1797–803. https://pubmed.ncbi.nlm.nih.gov/21911624/
Esselstyn CB. Resolving the coronary artery disease epidemic through plant-based nutrition. Prev Cardiol. 2001;4(4):171–7. https://pubmed.ncbi.nlm.nih.gov/11832674/
Barnett TD, Barnard ND, Radak TL. Development of symptomatic cardiovascular disease after self-reported adherence to the Atkins diet. J Am Diet Assoc. 2009;109(7):1263–5. https://pubmed.ncbi.nlm.nih.gov/19559147/
Ramírez R, Pedro-Botet J, García M, et al. Erectile dysfunction and cardiovascular risk factors in a Mediterranean diet cohort. Intern Med J. 2016;46(1):52–6. https://pubmed.ncbi.nlm.nih.gov/26482327/
Aldemir M, Okulu E, Neselioglu S, Erel O, Kayigil O. Pistachio diet improves erectile function parameters and serum lipid profiles in patients with erectile dysfunction. Int J Impot Res. 2011;23(1):32–8. https://pubmed.ncbi.nlm.nih.gov/21228801/
Salas-Huetos A, Moraleda R, Giardina S, et al. Effect of nut consumption on semen quality and functionality in healthy men consuming a Western-style diet: a randomized controlled trial. Am J Clin Nutr. 2018;108(5):953–62. https://pubmed.ncbi.nlm.nih.gov/30475967/
Salas-Huetos A, Muralidharan J, Galiè S, Salas-Salvadó J, Bulló M. Effect of nut consumption on erectile and sexual function in healthy males: a secondary outcome analysis of the FERTINUTS randomized controlled trial. Nutrients. 2019;11(6):E1372. https://pubmed.ncbi.nlm.nih.gov/31248067/
Aldemir M, Okulu E, Neselioglu S, Erel O, Kayigil O. Pistachio diet improves erectile function parameters and serum lipid profiles in patients with erectile dysfunction. Int J Impot Res. 2011;23(1):32–8. https://pubmed.ncbi.nlm.nih.gov/21228801/
Salas-Huetos A, Muralidharan J, Galiè S, Salas-Salvadó J, Bulló M. Effect of nut consumption on erectile and sexual function in healthy males: a secondary outcome analysis of the FERTINUTS randomized controlled trial. Nutrients. 2019;11(6):E1372. https://pubmed.ncbi.nlm.nih.gov/31248067/
Ohebshalom M, Mulhall JP. Transdermal and topical pharmacotherapy for male sexual dysfunction. Expert Opin Drug Deliv. 2005;2(1):115–20. https://pubmed.ncbi.nlm.nih.gov/16296739/
Bhupathiraju SN, Wedick NM, Pan A, et al. Quantity and variety in fruit and vegetable intake and risk of coronary heart disease. Am J Clin Nutr. 2013;98(6):1514–23. https://pubmed.ncbi.nlm.nih.gov/24088718/
Tamakoshi A, Tamakoshi K, Lin Y, Yagyu K, Kikuchi S. Healthy lifestyle and preventable death: findings from the Japan Collaborative Cohort (JACC) Study. Prev Med. 2009;48(5):486–92. https://pubmed.ncbi.nlm.nih.gov/19254743/
Wang F, Dai S, Wang M, Morrison H. Erectile dysfunction and fruit/vegetable consumption among diabetic Canadian men. Urology. 2013;82(6):1330–5. https://pubmed.ncbi.nlm.nih.gov/24295250/
Presley TD, Morgan AR, Bechtold E, et al. Acute effect of a high nitrate diet on brain perfusion in older adults. Nitric Oxide. 2011;24(1):34–42. https://pubmed.ncbi.nlm.nih.gov/20951824/
Rhim HC, Kim MS, Park YJ, et al. The potential role of arginine supplements on erectile dysfunction: a systemic review and meta-analysis. J Sex Med. 2019;16(2):223–34. https://pubmed.ncbi.nlm.nih.gov/30770070/
Grimble GK. Adverse gastrointestinal effects of arginine and related amino acids. J Nutr. 2007;137(6 Suppl 2):1693S-701S. https://pubmed.ncbi.nlm.nih.gov/17513449/
Rimando AM, Perkins-veazie PM. Determination of citrulline in watermelon rind. J Chromatogr A. 2005;1078(1–2):196–200. https://pubmed.ncbi.nlm.nih.gov/16007998/
Cormio L, De siati M, Lorusso F, et al. Oral L-citrulline supplementation improves erection hardness in men with mild erectile dysfunction. Urology. 2011;77(1):119–22. https://pubmed.ncbi.nlm.nih.gov/21195829/
Pfizer Annual Meeting of Shareholders 2014 Financial Report. http//www.pfizer.com/system/files/presentation/2014_Pfizer_Financial_Report.pdf. Accessed May 16, 2015.; https://www.pfizer.com/system/files/presentation/2014_Pfizer_Financial_Report.pdf
Johnson G. Watermelon board approves officers, budget, marketing plan. The Packer. http://www.thepacker.com/news/watermelon-board-approves-officers-budget-marketing-plan. February 24, 2015. Accessed May 16, 2015.; https://www.thepacker.com/news/watermelon-board-approves-officers-budget-marketing-plan
Lopresti AL, Drummond PD. Saffron (Crocus sativus) for depression: a systematic review of clinical studies and examination of underlying antidepressant mechanisms of action. Hum Psychopharmacol. 2014;29(6):517–27. https://pubmed.ncbi.nlm.nih.gov/25384672/
Talaei A, Hassanpour Moghadam M, Sajadi Tabassi SA, Mohajeri SA. Crocin, the main active saffron constituent, as an adjunctive treatment in major depressive disorder: a randomized, double-blind, placebo-controlled, pilot clinical trial. J Affect Disord. 2015;174:51–6. https://pubmed.ncbi.nlm.nih.gov/25484177/
Lopresti AL, Drummond PD. Saffron (Crocus sativus) for depression: a systematic review of clinical studies and examination of underlying antidepressant mechanisms of action. Hum Psychopharmacol. 2014;29(6):517–27. https://pubmed.ncbi.nlm.nih.gov/25384672/
Higgins A, Nash M, Lynch AM. Antidepressant-associated sexual dysfunction: impact, effects, and treatment. Drug Healthc Patient Saf. 2010;2:141–50. https://pubmed.ncbi.nlm.nih.gov/21701626/
Hu XH, Bull SA, Hunkeler EM, et al. Incidence and duration of side effects and those rated as bothersome with selective serotonin reuptake inhibitor treatment for depression: patient report versus physician estimate. J Clin Psychiatry. 2004;65(7):959–65. https://pubmed.ncbi.nlm.nih.gov/15291685/
Csoka AB, Shipko S. Persistent sexual side effects after SSRI discontinuation. Psychother Psychosom. 2006;75(3):187–8. https://pubmed.ncbi.nlm.nih.gov/16636635/
Modabbernia A, Sohrabi H, Nasehi AA, et al. Effect of saffron on fluoxetine-induced sexual impairment in men: randomized double-blind placebo-controlled trial. Psychopharmacology (Berl). 2012;223(4):381–8. https://pubmed.ncbi.nlm.nih.gov/22552758/
Kashani L, Raisi F, Saroukhani S, et al. Saffron for treatment of fluoxetine-induced sexual dysfunction in women: randomized double-blind placebo-controlled study. Hum Psychopharmacol. 2013;28(1):54–60. https://pubmed.ncbi.nlm.nih.gov/23280545/
Giesbers AAGM, Bruins JL, Kramer AEJL, Jonas U. New methods in the diagnosis of impotence: RigiScan® penile tumescence and rigidity monitoring and diagnostic papaverine hydrochloride injection. World J Urol. 1987;5(3):173–6. https://link.springer.com/article/10.1007/BF00326827
Mohammadzadeh-Moghadam H, Nazari SM, Shamsa A, et al. Effects of a topical saffron (Crocus sativus L) gel on erectile dysfunction in diabetics: a randomized, parallel-group, double-blind, placebo-controlled trial. J Evid Based Complementary Altern Med. 2015;20(4):283–6. https://pubmed.ncbi.nlm.nih.gov/25948674/
La J, Roberts NH, Yafi FA. Diet and men’s sexual health. Sex Med Rev. 2018;6(1):54–68. https://pubmed.ncbi.nlm.nih.gov/28778698/
Springmann M, Clark MA, Rayner M, Scarborough P, Webb P. The global and regional costs of healthy and sustainable dietary patterns: a modelling study. Lancet Planet Health. 2021;5(11):e797–807. https://pubmed.ncbi.nlm.nih.gov/34715058/
Flynn MM, Schiff AR. Economical healthy diets (2012): including lean animal protein costs more than using extra virgin olive oil. J Hunger Environ Nutr. 2015;10(4):467–82. https://www.tandfonline.com/doi/abs/10.1080/19320248.2015.1045675
Ahmed IA, Mikail MA, Zamakshshari N, Abdullah ASH. Natural anti-aging skincare: role and potential. Biogerontology. 2020;21(3):293–310. https://pubmed.ncbi.nlm.nih.gov/32162126/
1,86 м2. – Примеч. ред.
Gu Y, Han J, Jiang C, Zhang Y. Biomarkers, oxidative stress and autophagy in skin aging. Ageing Res Rev. 2020;59:101036. https://pubmed.ncbi.nlm.nih.gov/32105850/
Lowry WE. Its written all over your face: the molecular and physiological consequences of aging skin. Mech Ageing Dev. 2020;190:111315. https://pubmed.ncbi.nlm.nih.gov/32681843/
Ahmed IA, Mikail MA, Zamakshshari N, Abdullah ASH. Natural anti-aging skincare: role and potential. Biogerontology. 2020;21(3):293–310. https://pubmed.ncbi.nlm.nih.gov/32162126/
Makrantonaki E, Zouboulis CC. The skin as a mirror of the aging process in the human organism – state of the art and results of the aging research in the German National Genome Research Network 2 (NGFN-2). Exp Gerontol. 2007;42(9):879–86. https://pubmed.ncbi.nlm.nih.gov/17689905/
Ahmed IA, Mikail MA, Zamakshshari N, Abdullah ASH. Natural anti-aging skincare: role and potential. Biogerontology. 2020;21(3):293–310. https://pubmed.ncbi.nlm.nih.gov/32162126/
Makrantonaki E, Zouboulis CC. The skin as a mirror of the aging process in the human organism – state of the art and results of the aging research in the German National Genome Research Network 2 (NGFN-2). Exp Gerontol. 2007;42(9):879–86. https://pubmed.ncbi.nlm.nih.gov/17689905/
Chaudhary M, Khan A, Gupta M. Skin ageing: pathophysiology and current market treatment approaches. Curr Aging Sci. 2020;13(1):22–30. https://pubmed.ncbi.nlm.nih.gov/31530270/
Malik A, Hoenig LJ. Can aging be slowed down? Clin Dermatol. 2019;37(4):306–11. https://pubmed.ncbi.nlm.nih.gov/31345317/
Nikolakis G, Makrantonaki E, Zouboulis CC. Skin mirrors human aging. Horm Mol Biol Clin Investig. 2013;16(1):13–28. https://pubmed.ncbi.nlm.nih.gov/25436743/
Huang S, Haiminen N, Carrieri AP, et al. Human skin, oral, and gut microbiomes predict chronological age. mSystems. 2020;5(1):e00630–19. https://pubmed.ncbi.nlm.nih.gov/32047061/
Rinnerthaler M, Bischof J, Streubel MK, Trost A, Richter K. Oxidative stress in aging human skin. Biomolecules. 2015;5(2):545–89. https://pubmed.ncbi.nlm.nih.gov/25906193/
Ahmed IA, Mikail MA, Zamakshshari N, Abdullah ASH. Natural anti-aging skincare: role and potential. Biogerontology. 2020;21(3):293–310. https://pubmed.ncbi.nlm.nih.gov/32162126/
Nikolakis G, Makrantonaki E, Zouboulis CC. Skin mirrors human aging. Horm Mol Biol Clin Investig. 2013;16(1):13–28. https://pubmed.ncbi.nlm.nih.gov/25436743/
Singh G. Can we prevent skin aging? Indian J Dermatol Venereol Leprol. 2009;75(5):447–51. https://pubmed.ncbi.nlm.nih.gov/19736422/
Gu Y, Han J, Jiang C, Zhang Y. Biomarkers, oxidative stress and autophagy in skin aging. Ageing Res Rev. 2020;59:101036. https://pubmed.ncbi.nlm.nih.gov/32105850/
Silveira JEPS, Pedroso DMM. UV light and skin aging. Rev Environ Health. 2014;29(3):243–54. https://pubmed.ncbi.nlm.nih.gov/25241726/
Singh G. Can we prevent skin aging? Indian J Dermatol Venereol Leprol. 2009;75(5):447–51. https://pubmed.ncbi.nlm.nih.gov/19736422/
Ahmed IA, Mikail MA, Zamakshshari N, Abdullah ASH. Natural anti-aging skincare: role and potential. Biogerontology. 2020;21(3):293–310. https://pubmed.ncbi.nlm.nih.gov/32162126/
Gordon JRS, Brieva JC. Images in clinical medicine. Unilateral dermatoheliosis. N Engl J Med. 2012;366(16):e25. https://pubmed.ncbi.nlm.nih.gov/22512500/
Gunn DA, Dick JL, van Heemst D, et al. Lifestyle and youthful looks. Br J Dermatol. 2015;172(5):1338–45. https://pubmed.ncbi.nlm.nih.gov/25627783/
Malik A, Hoenig LJ. Can aging be slowed down? Clin Dermatol. 2019;37(4):306–11. https://pubmed.ncbi.nlm.nih.gov/31345317/
Singh G. Can we prevent skin aging? Indian J Dermatol Venereol Leprol. 2009;75(5):447–51. https://pubmed.ncbi.nlm.nih.gov/19736422/
Elsner P, Fluhr JW, Gehring W, et al. Anti-aging data and support claims – consensus statement. J Dtsch Dermatol Ges. 2011;9 Suppl 3:S1–32. https://pubmed.ncbi.nlm.nih.gov/22023288/
Gunn DA, Dick JL, van Heemst D, et al. Lifestyle and youthful looks. Br J Dermatol. 2015;172(5):1338–45. https://pubmed.ncbi.nlm.nih.gov/25627783/
Ahmed IA, Mikail MA, Zamakshshari N, Abdullah ASH. Natural anti-aging skincare: role and potential. Biogerontology. 2020;21(3):293–310. https://pubmed.ncbi.nlm.nih.gov/32162126/
Nikolakis G, Makrantonaki E, Zouboulis CC. Skin mirrors human aging. Horm Mol Biol Clin Investig. 2013;16(1):13–28. https://pubmed.ncbi.nlm.nih.gov/25436743/
Burford O, Jiwa M, Carter O, Parsons R, Hendrie D. Internet-based photoaging within Australian pharmacies to promote smoking cessation: randomized controlled trial. J Med Internet Res. 2013;15(3):e64. https://pubmed.ncbi.nlm.nih.gov/23531984/
Mahler HIM, Kulik JA, Gerrard M, Gibbons FX. Long-term effects of appearance-based interventions on sun protection behaviors. Health Psychol. 2007;26(3):350–60. https://pubmed.ncbi.nlm.nih.gov/17500622/
Misra BB. The chemical exposome of human aging. Front Genet. 2020;11:574936. https://pubmed.ncbi.nlm.nih.gov/33329714/
Wong QYA, Chew FT. Defining skin aging and its risk factors: a systematic review and meta-analysis. Sci Rep. 2021;11(1):22075. https://pubmed.ncbi.nlm.nih.gov/34764376/
Qiao Y, Li Q, Du HY, Wang QW, Huang Y, Liu W. Airborne polycyclic aromatic hydrocarbons trigger human skin cells aging through aryl hydrocarbon receptor. Biochem Biophys Res Commun. 2017;488(3):445–52. https://pubmed.ncbi.nlm.nih.gov/28526404/
Krutmann J, Liu W, Li L, et al. Pollution and skin: from epidemiological and mechanistic studies to clinical implications. J Dermatol Sci. 2014;76(3):163–8. https://pubmed.ncbi.nlm.nih.gov/25278222/
Van Rooij JG, Veeger MM, Bodelier-Bade MM, Scheepers PT, Jongeneelen FJ. Smoking and dietary intake of polycyclic aromatic hydrocarbons as sources of interindividual variability in the baseline excretion of 1-hydroxypyrene in urine. Int Arch Occup Environ Health. 1994;66(1):55–65. https://pubmed.ncbi.nlm.nih.gov/7927844/
Ramesh A, Walker SA, Hood DB, Guillén MD, Schneider K, Weyand EH. Bioavailability and risk assessment of orally ingested polycyclic aromatic hydrocarbons. Int J Toxicol. 2004;23(5):301–33. https://pubmed.ncbi.nlm.nih.gov/15513831/
Harris KL, Banks LD, Mantey JA, Huderson AC, Ramesh A. Bioaccessibility of polycyclic aromatic hydrocarbons: relevance to toxicity and carcinogenesis. Expert Opin Drug Metab Toxicol. 2013;9(11):1465–80. https://pubmed.ncbi.nlm.nih.gov/23898780/
Crinnion WJ. The role of persistent organic pollutants in the worldwide epidemic of type 2 diabetes mellitus and the possible connection to farmed Atlantic salmon (Salmo salar). Altern Med Rev. 2011;16(4):301–13. https://pubmed.ncbi.nlm.nih.gov/22214250/
Li Z, Romanoff L, Bartell S, et al. Excretion profiles and half-lives of ten urinary polycyclic aromatic hydrocarbon metabolites after dietary exposure. Chem Res Toxicol. 2012;25(7):1452–61. https://pubmed.ncbi.nlm.nih.gov/22663094/
Solway J, McBride M, Haq F, Abdul W, Miller R. Diet and dermatology: the role of a whole-food, plant-based diet in preventing and reversing skin aging – a review. J Clin Aesthet Dermatol. 2020 May;13(5):38–43. https://pubmed.ncbi.nlm.nih.gov/32802255/
Pontius AT, Smith PW. How to successfully incorporate antiaging and wellness into your practice: things you should know. Facial Plast Surg. 2010;26(1):12–5. https://pubmed.ncbi.nlm.nih.gov/20094964/
Smirnova MH. A will to youth: the woman’s anti-aging elixir. Soc Sci Med. 2012;75(7):1236–43. https://pubmed.ncbi.nlm.nih.gov/22742924/
Plastic Surgery Statistics Report: ASPS National Clearinghouse of Plastic Surgery Procedural Statistics 2020. American Society of Plastic Surgeons. https://www.plasticsurgery.org/documents/News/Statistics/2020/plastic-surgery-statistics-full-report-2020.pdf. Published 2020. Accessed August 31, 2022.; https://www.plasticsurgery.org/news/plastic-surgery-statistics
Barrett DM, Gerecci D, Wang TD. Facelift controversies. Facial Plast Surg Clin North Am. 2016;24(3):357–66. https://pubmed.ncbi.nlm.nih.gov/27400849/
Chopan M, Samant S, Mast BA. Contemporary analysis of rhytidectomy using the Tracking Operations and Outcomes for Plastic Surgeons database with 13,346 patients. Plast Reconstr Surg. 2020;145(6):1402–8. https://pubmed.ncbi.nlm.nih.gov/32459769/
Chopan M, Samant S, Mast BA. Contemporary analysis of rhytidectomy using the Tracking Operations and Outcomes for Plastic Surgeons database with 13,346 patients. Plast Reconstr Surg. 2020;145(6):1402–8. https://pubmed.ncbi.nlm.nih.gov/32459769/
Truswell WH. Approaches to reducing risk in rhytidectomy surgery. Facial Plast Surg Clin North Am. 2020;28(3):419–27. https://pubmed.ncbi.nlm.nih.gov/32503723/
Plastic Surgery Statistics Report: ASPS National Clearinghouse of Plastic Surgery Procedural Statistics 2020. American Society of Plastic Surgeons. https://www.plasticsurgery.org/documents/News/Statistics/2020/plastic-surgery-statistics-full-report-2020.pdf. Published 2020. Accessed August 31, 2022.; https://www.plasticsurgery.org/news/plastic-surgery-statistics
Lee KC, Pascal AB, Halepas S, Koch A. What are the most commonly reported complications with cosmetic botulinum toxin type A treatments? J Oral Maxillofac Surg. 2020;78(7):1190.e1–9. https://pubmed.ncbi.nlm.nih.gov/32192924/
Giordano CN, Matarasso SL, Ozog DM. Injectable and topical neurotoxins in dermatology: indications, adverse events, and controversies. J Am Acad Dermatol. 2017;76(6):1027–42. https://pubmed.ncbi.nlm.nih.gov/28522039/
Lee KC, Pascal AB, Halepas S, Koch A. What are the most commonly reported complications with cosmetic botulinum toxin type A treatments? J Oral Maxillofac Surg. 2020;78(7):1190.e1–9. https://pubmed.ncbi.nlm.nih.gov/32192924/
Plastic Surgery Statistics Report: ASPS National Clearinghouse of Plastic Surgery Procedural Statistics 2020. American Society of Plastic Surgeons. https://www.plasticsurgery.org/documents/News/Statistics/2020/plastic-surgery-statistics-full-report-2020.pdf. Published 2020. Accessed August 31, 2022.; https://www.plasticsurgery.org/news/plastic-surgery-statistics
DeVictor S, Ong AA, Sherris DA. Complications secondary to nonsurgical rhinoplasty: a systematic review and meta-analysis. Otolaryngol Head Neck Surg. 2021;165(5):611–6. https://pubmed.ncbi.nlm.nih.gov/33588622/
Vanaman M, Fabi SG, Carruthers J. Complications in the cosmetic dermatology patient: a review and our experience (Part 1). Dermatol Surg. 2016;42(1):1–11. https://pubmed.ncbi.nlm.nih.gov/26716709/
Tran AQ, Staropoli P, Rong AJ, Lee WW. Filler-associated vision loss. Facial Plast Surg Clin North Am. 2019;27(4):557–64. https://pubmed.ncbi.nlm.nih.gov/31587773/
Rayess HM, Svider PF, Hanba C, et al. A cross-sectional analysis of adverse events and litigation for injectable fillers. JAMA Facial Plast Surg. 2018;20(3):207–14. https://pubmed.ncbi.nlm.nih.gov/29270603/
Woodward J, Khan T, Martin J. Facial filler complications. Facial Plast Surg Clin North Am. 2015;23(4):447–58. https://pubmed.ncbi.nlm.nih.gov/26505541/
Plastic Surgery Statistics Report: ASPS National Clearinghouse of Plastic Surgery Procedural Statistics 2020. American Society of Plastic Surgeons. https://www.plasticsurgery.org/documents/News/Statistics/2020/plastic-surgery-statistics-full-report-2020.pdf. Published 2020. Accessed August 31, 2022.; https://www.plasticsurgery.org/news/plastic-surgery-statistics
Shah AR, Kennedy PM. The aging face. Med Clin North Am. 2018;102(6):1041–54. https://pubmed.ncbi.nlm.nih.gov/30342607/
Ganceviciene R, Liakou AI, Theodoridis A, Makrantonaki E, Zouboulis CC. Skin anti-aging strategies. Dermatoendocrinol. 2012;4(3):308–19. https://pubmed.ncbi.nlm.nih.gov/23467476/
Manríquez JJ, Cataldo K, Vera-Kellet C, Harz-Fresno I. Wrinkles. BMJ Clin Evid. 2014;2014:1711. https://pubmed.ncbi.nlm.nih.gov/25569867/
Neill US. Skin care in the aging female: myths and truths. J Clin Invest. 2012;122(2):473–7. https://pubmed.ncbi.nlm.nih.gov/22293186/
Vanaman M, Fabi SG, Carruthers J. Complications in the cosmetic dermatology patient: a review and our experience (Part 2). Dermatol Surg. 2016;42(1):12–20. https://pubmed.ncbi.nlm.nih.gov/26716709/
Ganceviciene R, Liakou AI, Theodoridis A, Makrantonaki E, Zouboulis CC. Skin anti-aging strategies. Dermatoendocrinol. 2012;4(3):308–19. https://pubmed.ncbi.nlm.nih.gov/23467476/
Sillanpää S, Salminen J-P, Eeva T. Breeding success and lutein availability in great tit (Parus major). Acta Oecologica. 2009;35(6):805–10. https://www.researchgate.net/publication/229107650_Breeding_success_and_lutein_availability_in_great_tit_Parus_major
Whitehead RD, Coetzee V, Ozakinci G, Perrett DI. Cross-cultural effects of fruit and vegetable consumption on skin color. Am J Public Health. 2012;102(2):212–3. https://pubmed.ncbi.nlm.nih.gov/22390434/
Stephen ID, Law Smith MJ, Stirrat MR, Perrett DI. Facial skin coloration affects perceived health of human faces. Int J Primatol. 2009;30(6):845–57. https://pubmed.ncbi.nlm.nih.gov/19946602/
Whitehead RD, Re D, Xiao D, Ozakinci G, Perrett DI. You are what you eat: within-subject increases in fruit and vegetable consumption confer beneficial skin-color changes. PLoS One. 2012;7(3):e32988. https://pubmed.ncbi.nlm.nih.gov/22412966/
Stahl W, Heinrich U, Jungmann H, et al. Increased dermal carotenoid levels assessed by noninvasive reflection spectrophotometry correlate with serum levels in women ingesting Betatene. J Nutr. 1998;128(5):903–7. https://pubmed.ncbi.nlm.nih.gov/9567001/
Stephen ID, Law Smith MJ, Stirrat MR, Perrett DI. Facial skin coloration affects perceived health of human faces. Int J Primatol. 2009;30(6):845–57. https://pubmed.ncbi.nlm.nih.gov/19946602/
Lefevre CE, Perrett DI. Fruit over sunbed: carotenoid skin colouration is found more attractive than melanin colouration. Q J Exp Psychol (Hove). 2015;68(2):284–93. https://pubmed.ncbi.nlm.nih.gov/25014019/
Pezdirc K, Hutchesson M, Whitehead R, Ozakinci G, Perrett D, Collins CE. Can dietary intake influence perception of and measured appearance? A systematic review. Nutr Res. 2015;35(3):175–97. https://pubmed.ncbi.nlm.nih.gov/25600848/
Greens to be gorgeous: why eating your five fruit and veg a day makes you sexy. Daily Mail. http://www.dailymail.co.uk/health/article-1228348/Eating-fruit-veg-makes-attractive-opposite-sex.html. November 17, 2009. Accessed September 7, 2022.; https://www.dailymail.co.uk/health/article-1228348/Eating-fruit-veg-makes-attractive-opposite-sex.html
Mitic V, Jovanovic VS, Dimitrijevic M, Cvetkovic J, Stojanovic G. Effect of food preparation technique on antioxidant activity and plant pigment content in some vegetables species. J Food Nutr Res. 2013;1(6):121–7. https://www.researchgate.net/publication/284422190_Effect_of_food_preparation_technique_on_antioxidant_activity_and_plant_pigment_content_in_some_vegetable_species
Meinke MC, Nowbary CK, Schanzer S, Vollert H, Lademann J, Darvin ME. Influences of orally taken carotenoid-rich curly kale extract on collagen I/elastin index of the skin. Nutrients. 2017;9(7):775. https://pubmed.ncbi.nlm.nih.gov/28753935/
Shoji T, Masumoto S, Moriichi N, Ohtake Y, Kanda T. Administration of apple polyphenol supplements for skin conditions in healthy women: a randomized, double-blind, placebo-controlled clinical trial. Nutrients. 2020;12(4):1071. https://pubmed.ncbi.nlm.nih.gov/32294883/
Nobile V, Michelotti A, Cestone E, et al. Skin photoprotective and antiageing effects of a combination of rosemary (Rosmarinus officinalis) and grapefruit (Citrus paradisi) polyphenols. Food Nutr Res. 2016;60:31871. https://pubmed.ncbi.nlm.nih.gov/27374032/
Stahl W, Heinrich U, Wiseman S, Eichler O, Sies H, Tronnier H. Dietary tomato paste protects against ultraviolet light-induced erythema in humans. J Nutr. 2001;131(5):1449–51. https://pubmed.ncbi.nlm.nih.gov/11340098/
Palombo P, Fabrizi G, Ruocco V, et al. Beneficial long-term effects of combined oral/topical antioxidant treatment with the carotenoids lutein and zeaxanthin on human skin: a double-blind, placebo-controlled study. Skin Pharmacol Physiol. 2007;20(4):199–210. https://pubmed.ncbi.nlm.nih.gov/17446716/
Köpcke W, Krutmann J. Protection from sunburn with ß-carotene – a meta-analysis. Photochem Photobiol. 2008;84(2):284–8. https://pubmed.ncbi.nlm.nih.gov/18086246/
Darvin M, Patzelt A, Gehse S, et al. Cutaneous concentration of lycopene correlates significantly with the roughness of the skin. Eur J Pharm Biopharm. 2008;69(3):943–7. https://pubmed.ncbi.nlm.nih.gov/18411044/
Hughes MCB, Williams GM, Pageon H, Fourtanier A, Green AC. Dietary antioxidant capacity and skin photoaging: a 15-year longitudinal study. J Invest Dermatol. 2021;141(4S):1111–8.e2. https://pubmed.ncbi.nlm.nih.gov/32682911/
Sundelin T, Lekander M, Kecklund G, Van Someren EJW, Olsson A, Axelsson J. Cues of fatigue: effects of sleep deprivation on facial appearance. Sleep. 2013;36(9):1355–60. https://pubmed.ncbi.nlm.nih.gov/23997369/
Axelsson J, Sundelin T, Ingre M, Van Someren EJW, Olsson A, Lekander M. Beauty sleep: experimental study on the perceived health and attractiveness of sleep deprived people. BMJ. 2010;341:c6614. https://pubmed.ncbi.nlm.nih.gov/21156746/
Atrooz F, Salim S. Sleep deprivation, oxidative stress and inflammation. Adv Protein Chem Struct Biol. 2020;119:309–36. https://pubmed.ncbi.nlm.nih.gov/31997771/
Lee CM, Watson REB, Kleyn CE. The impact of perceived stress on skin ageing. J Eur Acad Dermatol Venereol. 2020;34(1):54–8. https://pubmed.ncbi.nlm.nih.gov/31407395/
Noordam R, Gunn DA, Tomlin CC, et al. Cortisol serum levels in familial longevity and perceived age: the Leiden Longevity Study. Psychoneuroendocrinology. 2012;37(10):1669–75. https://pubmed.ncbi.nlm.nih.gov/22429748/
Smith S. The graying of the presidents. Boston.com. http://archive.boston.com/news/politics/2008/articles/2009/01/04/the_graying_of_the_presidents/. Published January 4, 2009. Accessed August 31, 2022.; https://archive.boston.com/news/politics/2008/articles/2009/01/04/the_graying_of_the_presidents/
Agrigoroaei S, Attardo AL, Lachman ME. Stress and subjective age: those with greater financial stress look older. Res Aging. 2017;39(10):1075–99. https://pubmed.ncbi.nlm.nih.gov/27422884/
Mukamal KJ. Alcohol consumption and self-reported sunburn: a cross-sectional, population-based survey. J Am Acad Dermatol. 2006;55(4):584–9. https://pubmed.ncbi.nlm.nih.gov/17010736/
Darvin ME, Sterry W, Lademann J, Patzelt A. Alcohol consumption decreases the protection efficiency of the antioxidant network and increases the risk of sunburn in human skin. Skin Pharmacol Physiol. 2013;26(1):45–51. https://pubmed.ncbi.nlm.nih.gov/23147451/
Darvin ME, Sterry W, Lademann J, Patzelt A. Alcohol consumption decreases the protection efficiency of the antioxidant network and increases the risk of sunburn in human skin. Skin Pharmacol Physiol. 2013;26(1):45–51. https://pubmed.ncbi.nlm.nih.gov/23147451/
Castelo-Branco C, Figueras F, Martínez de Osaba MJ, Vanrell JA. Facial wrinkling in postmenopausal women. Effects of smoking status and hormone replacement therapy. Maturitas. 1998;29(1):75–86. https://pubmed.ncbi.nlm.nih.gov/9643520/
Wong QYA, Chew FT. Defining skin aging and its risk factors: a systematic review and meta-analysis. Sci Rep. 2021;11(1):22075. https://pubmed.ncbi.nlm.nih.gov/34764376/
Walsh NP, Fortes MB, Raymond-Barker P, et al. Is whole-body hydration an important consideration in dry eye? Invest Ophthalmol Vis Sci. 2012;53(10):6622–7. https://pubmed.ncbi.nlm.nih.gov/22952120/
Sharma A, Hindman HB. Aging: a predisposition to dry eyes. J Ophthalmol. 2014;2014:781683. https://pubmed.ncbi.nlm.nih.gov/25197560/
Akdeniz M, Tomova-Simitchieva T, Dobos G, Blume-Peytavi U, Kottner J. Does dietary fluid intake affect skin hydration in healthy humans? A systematic literature review. Skin Res Technol. 2018;24(3):459–65. https://pubmed.ncbi.nlm.nih.gov/29392767/
Clarke KA, Dew TP, Watson REB, et al. Green tea catechins and their metabolites in human skin before and after exposure to ultraviolet radiation. J Nutr Biochem. 2016;27:203–10. https://pubmed.ncbi.nlm.nih.gov/26454512/
Fukushima Y, Takahashi Y, Hori Y, et al. Skin photoprotection and consumption of coffee and polyphenols in healthy middle-aged Japanese females. Int J Dermatol. 2015;54(4):410–8. https://pubmed.ncbi.nlm.nih.gov/25041334/
Fukushima Y, Takahashi Y, Kishimoto Y, et al. Consumption of polyphenols in coffee and green tea alleviates skin photoaging in healthy Japanese women. Clin Cosmet Investig Dermatol. 2020;13:165–72. https://pubmed.ncbi.nlm.nih.gov/32104042/
Chiu AE, Chan JL, Kern DG, Kohler S, Rehmus WE, Kimball AB. Double-blinded, placebo-controlled trial of green tea extracts in the clinical and histologic appearance of photoaging skin. Dermatol Surg. 2005;31(7 Pt 2):855–60. https://pubmed.ncbi.nlm.nih.gov/16029678/
Jeon HY, Kim JK, Kim WG, Lee SJ. Effects of oral epigallocatechin gallate supplementation on the minimal erythema dose and UV-induced skin damage. Skin Pharmacol Physiol. 2009;22(3):137–41. https://pubmed.ncbi.nlm.nih.gov/19212149/
Heinrich U, Moore CE, De Spirt S, Tronnier H, Stahl W. Green tea polyphenols provide photoprotection, increase microcirculation, and modulate skin properties of women. J Nutr. 2011;141(6):1202–8. https://pubmed.ncbi.nlm.nih.gov/21525260/
Farrar MD, Nicolaou A, Clarke KA, et al. A randomized controlled trial of green tea catechins in protection against ultraviolet radiation – induced cutaneous inflammation. Am J Clin Nutr. 2015;102(3):608–15. https://pubmed.ncbi.nlm.nih.gov/26178731/
Janjua R, Munoz C, Gorell E, et al. A two-year, double-blind, randomized placebo-controlled trial of oral green tea polyphenols on the long-term clinical and histologic appearance of photoaging skin. Dermatol Surg. 2009;35(7):1057–65. https://pubmed.ncbi.nlm.nih.gov/19469799/
Tjeerdsma F, Jonkman MF, Spoo JR. Temporary arrest of basal cell carcinoma formation in a patient with basal cell naevus syndrome (BCNS) since treatment with a gel containing various plant extracts. J Eur Acad Dermatol Venereol. 2011;25(2):244–5. https://pubmed.ncbi.nlm.nih.gov/20500542/
Camouse MM, Domingo DS, Swain FR, et al. Topical application of green and white tea extracts provides protection from solar-simulated ultraviolet light in human skin. Exp Dermatol. 2009;18(6):522–6. https://pubmed.ncbi.nlm.nih.gov/19492999/
Chiu AE, Chan JL, Kern DG, Kohler S, Rehmus WE, Kimball AB. Double-blinded, placebo-controlled trial of green tea extracts in the clinical and histologic appearance of photoaging skin. Dermatol Surg. 2005;31(7 Pt 2):855–60. https://pubmed.ncbi.nlm.nih.gov/16029678/
Kessels J, Voeten L, Nelemans P, et al. Topical sinecatechins, 10 %, ointment for superficial basal cell carcinoma: a randomized clinical trial. JAMA Dermatol. 2017;153(10):1061–3. https://pubmed.ncbi.nlm.nih.gov/28793140/
Petrova A, Davids LM, Rautenbach F, Marnewick JL. Photoprotection by honeybush extracts, hesperidin and mangiferin against UVB-induced skin damage in SKH-1 mice. J Photochem Photobiol B. 2011;103(2):126–39. https://pubmed.ncbi.nlm.nih.gov/21435898/
Choi SY, Hong JY, Ko EJ, et al. Protective effects of fermented honeybush (Cyclopia intermedia) extract (HU-018) against skin aging: a randomized, double-blinded, placebo-controlled study. J Cosmet Laser Ther. 2018;20(5):313–8. https://pubmed.ncbi.nlm.nih.gov/29388846/
Neukam K, Stahl W, Tronnier H, Sies H, Heinrich U. Consumption of flavanol-rich cocoa acutely increases microcirculation in human skin. Eur J Nutr. 2007;46(1):53–6. https://pubmed.ncbi.nlm.nih.gov/17164979/
Heinrich U, Neukam K, Tronnier H, Sies H, Stahl W. Long-term ingestion of high flavanol cocoa provides photoprotection against UV-induced erythema and improves skin condition in women. J Nutr. 2006;136(6):1565–9. https://pubmed.ncbi.nlm.nih.gov/16702322/
Yoon HS, Kim JR, Park GY, et al. Cocoa flavanol supplementation influences skin conditions of photo-aged women: a 24-week double-blind, randomized, controlled trial. J Nutr. 2016;146(1):46–50. https://pubmed.ncbi.nlm.nih.gov/26581682/
Anson G, Kane MAC, Lambros V. Sleep wrinkles: facial aging and facial distortion during sleep. Aesthet Surg J. 2016;36(8):931–40. https://pubmed.ncbi.nlm.nih.gov/27329660/
Kligman AM, Zheng P, Lavker RM. The anatomy and pathogenesis of wrinkles. Br J Dermatol. 1985;113(1):37–42. https://pubmed.ncbi.nlm.nih.gov/4015970/
Hillebrand GG, Liang Z, Yan X, Yoshii T. New wrinkles on wrinkling: an 8-year longitudinal study on the progression of expression lines into persistent wrinkles. Br J Dermatol. 2010;162(6):1233–41. https://pubmed.ncbi.nlm.nih.gov/20184587/
Vogeley C, Esser C, Tüting T, Krutmann J, Haarmann-Stemmann T. Role of the aryl hydrocarbon receptor in environmentally induced skin aging and skin carcinogenesis. Int J Mol Sci. 2019;20(23):6005. https://pubmed.ncbi.nlm.nih.gov/31795255/
Mekic S, Jacobs LC, Hamer MA, et al. A healthy diet in women is associated with less facial wrinkles in a large Dutch population-based cohort. J Am Acad Dermatol. 2019;80(5):1358–63.e2. https://pubmed.ncbi.nlm.nih.gov/29601935/
Cosgrove MC, Franco OH, Granger SP, Murray PG, Mayes AE. Dietary nutrient intakes and skin-aging appearance among middle-aged American women. Am J Clin Nutr. 2007;86(4):1225–31. https://pubmed.ncbi.nlm.nih.gov/17921406/
Foolad N, Vaughn AR, Rybak I, et al. Prospective randomized controlled pilot study on the effects of almond consumption on skin lipids and wrinkles. Phytother Res. 2019;33(12):3212–7. https://pubmed.ncbi.nlm.nih.gov/31576607/
De Spirt S, Stahl W, Tronnier H, et al. Intervention with flaxseed and borage oil supplements modulates skin condition in women. Br J Nutr. 2009;101(3):440–5. https://pubmed.ncbi.nlm.nih.gov/18761778/
Izumi T, Saito M, Obata A, Arii M, Yamaguchi H, Matsuyama A. Oral intake of soy isoflavone aglycone improves the aged skin of adult women. J Nutr Sci Vitaminol (Tokyo). 2007;53(1):57–62. https://pubmed.ncbi.nlm.nih.gov/17484381/
Oyama A, Ueno T, Uchiyama S, et al. The effects of natural S-equol supplementation on skin aging in postmenopausal women: a pilot randomized placebo-controlled trial. Menopause. 2012;19(2):202–10. https://pubmed.ncbi.nlm.nih.gov/21934634/
Fam VW, Holt RR, Keen CL, Sivamani RK, Hackman RM. Prospective evaluation of mango fruit intake on facial wrinkles and erythema in postmenopausal women: a randomized clinical pilot study. Nutrients. 2020;12(11):3381. https://pubmed.ncbi.nlm.nih.gov/33158079/
Katta R, Sanchez A, Tantry E. An anti-wrinkle diet: nutritional strategies to combat oxidation, inflammation and glycation. Skin Therapy Lett. 2020;25(2):3–7. https://pubmed.ncbi.nlm.nih.gov/32196147/
Solway J, McBride M, Haq F, Abdul W, Miller R. Diet and dermatology: the role of a whole-food, plant-based diet in preventing and reversing skin aging – a review. J Clin Aesthet Dermatol. 2020;13(5):38–43. https://pubmed.ncbi.nlm.nih.gov/32802255/
Pacifico A, Conic RRZ, Cristaudo A, et al. Diet-related phototoxic reactions in psoriatic patients undergoing phototherapy: results from a multicenter prospective study. Nutrients. 2021;13(9):2934. https://pubmed.ncbi.nlm.nih.gov/34578812/
Fusano M, Zane C, Calzavara-Pinton P, Bencini PL. Photodynamic therapy for actinic keratosis in vegan and omnivore patients: the role of diet on skin healing. J Dermatolog Treat. 2021;32(1):78–83. https://pubmed.ncbi.nlm.nih.gov/31076007/
Fusano M, Galimberti MG, Bencini PL. Laser removal of tattoos in vegan and omnivore patients. J Cosmet Dermatol. 2022;21(2):674–8. https://pubmed.ncbi.nlm.nih.gov/33813803/
Fusano M, Bencini PL, Fusano I, et al. Ultrapulsed CO2 resurfacing of photodamaged facial skin in vegan and omnivore patients: a multicentric study. Lasers Surg Med. 2021;53(10):1370–5. https://pubmed.ncbi.nlm.nih.gov/34015157/
Fusano M, Fusano I, Galimberti MG, Bencini M, Bencini PL. Comparison of postsurgical scars between vegan and omnivore patients. Dermatol Surg. 2020;46(12):1572–6. https://pubmed.ncbi.nlm.nih.gov/32769530/
Galimberti MG, Guida S, Pellacani G, Bencini PL. Hyaluronic acid filler for skin rejuvenation: the role of diet on outcomes. A pilot study. Dermatol Ther. 2018;31(4):e12646. https://pubmed.ncbi.nlm.nih.gov/30019474/
Fusano M, Galimberti MG, Bencini M, Fusano I, Bencini PL. Comparison of microfocused ultrasound with visualization for skin laxity among vegan and omnivore patients. J Cosmet Dermatol. 2021;20(9):2769–74. https://pubmed.ncbi.nlm.nih.gov/33533546/
Karlic H, Schuster D, Varga F, et al. Vegetarian diet affects genes of oxidative metabolism and collagen synthesis. Ann Nutr Metab. 2008;53(1):29–32. https://pubmed.ncbi.nlm.nih.gov/18772587/
Fusano M, Zane C, Calzavara-Pinton P, Bencini PL. Photodynamic therapy for actinic keratosis in vegan and omnivore patients: the role of diet on skin healing. J Dermatolog Treat. 2021;32(1):78–83. https://pubmed.ncbi.nlm.nih.gov/31076007/
Fusano M, Galimberti MG, Bencini PL. Laser removal of tattoos in vegan and omnivore patients. J Cosmet Dermatol. 2022;21(2):674–8. https://pubmed.ncbi.nlm.nih.gov/33813803/
Fusano M, Bencini PL, Fusano I, et al. Ultrapulsed CO2 resurfacing of photodamaged facial skin in vegan and omnivore patients: a multicentric study. Lasers Surg Med. 2021;53(10):1370–5. https://pubmed.ncbi.nlm.nih.gov/34015157/
Fusano M, Fusano I, Galimberti MG, Bencini M, Bencini PL. Comparison of postsurgical scars between vegan and omnivore patients. Dermatol Surg. 2020;46(12):1572–6. https://pubmed.ncbi.nlm.nih.gov/32769530/
Fusano M, Galimberti MG, Bencini M, Fusano I, Bencini PL. Comparison of microfocused ultrasound with visualization for skin laxity among vegan and omnivore patients. J Cosmet Dermatol. 2021;20(9):2769–74. https://pubmed.ncbi.nlm.nih.gov/33533546/
Kang AH, Trelstad RL. A collagen defect in homocystinuria. J Clin Invest. 1973;52(10):2571–8. https://pubmed.ncbi.nlm.nih.gov/4729050/
Rao VH, Bose SM. Effect of vitamin B12 on the formation of collagen and nucleic acids in the albino rat skins and granulomas. J Vitaminol (Kyoto). 1970;16(4):253–8. https://pubmed.ncbi.nlm.nih.gov/5497907/
Findlay CW. Effect of vitamin B12 on wound healing. Proc Soc Exp Biol Med. 1953;82(3):492–5. https://pubmed.ncbi.nlm.nih.gov/13047441/
Tuz MA, Mitchell A. The influence of anaemia on pressure ulcer healing in elderly patients. Br J Nurs. 2021;30(15):S32–8. https://pubmed.ncbi.nlm.nih.gov/34379458/
Kang AH, Trelstad RL. A collagen defect in homocystinuria. J Clin Invest. 1973;52(10):2571–8. https://pubmed.ncbi.nlm.nih.gov/4729050/
Makris EA, Responte DJ, Paschos NK, Hu JC, Athanasiou KA. Developing functional musculoskeletal tissues through hypoxia and lysyl oxidase-induced collagen cross-linking. Proc Natl Acad Sci U S A. 2014;111(45):E4832–41. https://pubmed.ncbi.nlm.nih.gov/25349395/
National Pressure Ulcer Advisory Panel, European Pressure Ulcer Advisory Panel and Pan Pacific Pressure Injury Alliance, Haesler E, ed. Prevention and Treatment of Pressure Ulcers: Quick Reference Guide. Cambridge Medi; 2014. https://www.epuap.org/wp-content/uploads/2016/10/quick-reference-guide-digital-npuap-epuap-pppia-jan2016.pdf
Smith MEB, Totten A, Hickam DH, et al. Pressure ulcer treatment strategies: a systematic comparative effectiveness review. Ann Intern Med. 2013;159(1):39–50. https://pubmed.ncbi.nlm.nih.gov/23817703/
Mariotti F, Gardner CD. Dietary protein and amino acids in vegetarian diets – a review. Nutrients. 2019;11(11):2661. https://pubmed.ncbi.nlm.nih.gov/31690027/
Berryman CE, Lieberman HR, Fulgoni VL, Pasiakos SM. Protein intake trends and conformity with the Dietary Reference Intakes in the United States: analysis of the National Health and Nutrition Examination Survey, 2001–2014. Am J Clin Nutr. 2018;108(2):405–13. https://pubmed.ncbi.nlm.nih.gov/29931213/
Jhawar N, Wang JV, Saedi N. Oral collagen supplementation for skin aging: a fad or the future? J Cosmet Dermatol. 2020;19(4):910–2. https://pubmed.ncbi.nlm.nih.gov/31411379/
de Lange C. Can a drink really make skin look younger? The Guardian. https://www.theguardian.com/science/2015/sep/27/nutricosmetics-drink-make-skin-look-younger-science. Published September 27, 2015. Accessed August 31, 2022.; https://www.theguardian.com/science/2015/sep/27/nutricosmetics-drink-make-skin-look-younger-science
Albornoz CA, Shah S, Murgia RD, Wang JV, Saedi N. Understanding aesthetic interest in oral collagen peptides: a 5-year national assessment. J Cosmet Dermatol. 2021;20(2):566–8. https://pubmed.ncbi.nlm.nih.gov/32559349/
de Miranda RB, Weimer P, Rossi RC. Effects of hydrolyzed collagen supplementation on skin aging: a systematic review and meta-analysis. Int J Dermatol. 2021;60(12):1449–61. https://pubmed.ncbi.nlm.nih.gov/33742704/
Albornoz CA, Shah S, Murgia RD, Wang JV, Saedi N. Understanding aesthetic interest in oral collagen peptides: a 5-year national assessment. J Cosmet Dermatol. 2021;20(2):566–8. https://pubmed.ncbi.nlm.nih.gov/32559349/
Jhawar N, Wang JV, Saedi N. Oral collagen supplementation for skin aging: a fad or the future? J Cosmet Dermatol. 2020;19(4):910–2. https://pubmed.ncbi.nlm.nih.gov/31411379/
Rustad AM, Nickles MA, McKenney JE, Bilimoria SN, Lio PA. Myths and media in oral collagen supplementation for the skin, nails, and hair: a review. J Cosmet Dermatol. 2022 Feb;21(2):438–43. https://pubmed.ncbi.nlm.nih.gov/34694676/
Spiro A, Lockyer S. Nutraceuticals and skin appearance: is there any evidence to support this growing trend? Nutr Bull. 2018;43(1):10–45. https://onlinelibrary.wiley.com/doi/10.1111/nbu.12304
Hujoel PP, Hujoel MLA. Vitamin C and scar strength: analysis of a historical trial and implications for collagen-related pathologies. Am J Clin Nutr. 2022;115(1):8–17. https://pubmed.ncbi.nlm.nih.gov/34396385/
Vitamin C: factsheet for health professionals. National Institutes of Health. https://ods.od.nih.gov/factsheets/VitaminC-HealthProfessional/. Updated March 26, 2021. Accessed September 7, 2022.; https://ods.od.nih.gov/factsheets/VitaminC-HealthProfessional/
Albornoz CA, Shah S, Murgia RD, Wang JV, Saedi N. Understanding aesthetic interest in oral collagen peptides: a 5-year national assessment. J Cosmet Dermatol. 2021;20(2):566–8. https://pubmed.ncbi.nlm.nih.gov/32559349/
Perez-Sanchez AC, Burns EK, Perez VM, Tantry EK, Prabhu S, Katta R. Safety concerns of skin, hair and nail supplements in retail stores. Cureus. 12(7):e9477.; https://pubmed.ncbi.nlm.nih.gov/32874806/
Avila-Rodríguez MI, Rodríguez Barroso LG, Sánchez ML. Collagen: a review on its sources and potential cosmetic applications. J Cosmet Dermatol. 2018;17(1):20–6. https://pubmed.ncbi.nlm.nih.gov/29144022/
Sionkowska A, Adamiak K, Musial K, Gadomska M. Collagen based materials in cosmetic applications: a review. Materials. 2020;13(19):4217. https://pubmed.ncbi.nlm.nih.gov/32977407/
Perez-Sanchez AC, Burns EK, Perez VM, Tantry EK, Prabhu S, Katta R. Safety concerns of skin, hair and nail supplements in retail stores. Cureus. 12(7):e9477.; https://pubmed.ncbi.nlm.nih.gov/32874806/
Rustad AM, Nickles MA, McKenney JE, Bilimoria SN, Lio PA. Myths and media in oral collagen supplementation for the skin, nails, and hair: a review. J Cosmet Dermatol. 2022;21(2):438–43. https://pubmed.ncbi.nlm.nih.gov/34694676/
Cao C, Xiao Z, Wu Y, Ge C. Diet and skin aging – from the perspective of food nutrition. Nutrients. 2020;12(3):870. https://pubmed.ncbi.nlm.nih.gov/32213934/
Cooperman T. Collagen supplements. ConsumerLab.com. https://www.consumerlab.com/reviews/collagen-supplements-review-peptides-hydrolysate/collagen/. Published September 28, 2019. Updated June 6, 2022. Accessed August 31, 2022.; https://www.consumerlab.com/reviews/collagen-supplements-review-peptides-hydrolysate/collagen/
Seror J, Stern M, Zarka R, Orr N. The potential use of novel plant-derived recombinant human collagen in aesthetic medicine. Plast Reconstr Surg. 2021;148(6S):32S-8S. https://pubmed.ncbi.nlm.nih.gov/34847096/
Huang CK, Miller TA. The truth about over-the-counter topical anti-aging products: a comprehensive review. Aesthet Surg J. 2007;27(4):402–12. https://pubmed.ncbi.nlm.nih.gov/19341668/
Neill US. Skin care in the aging female: myths and truths. J Clin Invest. 2012;122(2):473–7. https://pubmed.ncbi.nlm.nih.gov/22293186/
Elsner P, Fluhr JW, Gehring W, et al. Anti-aging data and support claims – consensus statement. J Dtsch Dermatol Ges. 2011;9 Suppl 3:S1–32. https://pubmed.ncbi.nlm.nih.gov/22023288/
Huang CK, Miller TA. The truth about over-the-counter topical anti-aging products: a comprehensive review. Aesthet Surg J. 2007;27(4):402–12. https://pubmed.ncbi.nlm.nih.gov/19341668/
Elsner P, Fluhr JW, Gehring W, et al. Anti-aging data and support claims – consensus statement. J Dtsch Dermatol Ges. 2011;9 Suppl 3:S1–32. https://pubmed.ncbi.nlm.nih.gov/22023288/
Mayes AE, Murray PG, Gunn DA, et al. Environmental and lifestyle factors associated with perceived facial age in Chinese women. PLoS One. 2010;5(12):e15270. https://pubmed.ncbi.nlm.nih.gov/21179450/
Gunn DA, Dick JL, van Heemst D, et al. Lifestyle and youthful looks. Br J Dermatol. 2015;172(5):1338–45. https://pubmed.ncbi.nlm.nih.gov/25627783/
Huang CK, Miller TA. The truth about over-the-counter topical anti-aging products: a comprehensive review. Aesthet Surg J. 2007;27(4):402–12. https://pubmed.ncbi.nlm.nih.gov/19341668/
Neill US. Skin care in the aging female: myths and truths. J Clin Invest. 2012;122(2):473–7. https://pubmed.ncbi.nlm.nih.gov/22293186/
Draelos ZD. Active agents in common skin care products. Plast Reconstr Surg. 2010;125(2):719–24. https://pubmed.ncbi.nlm.nih.gov/20124857/
Ramos-e-Silva M, Celem LR, Ramos-e-Silva S, Fucci-da-Costa AP. Anti-aging cosmetics: facts and controversies. Clin Dermatol. 2013;31(6):750–8. https://pubmed.ncbi.nlm.nih.gov/24160281/
Draelos ZD. Active agents in common skin care products. Plast Reconstr Surg. 2010;125(2):719–24. https://pubmed.ncbi.nlm.nih.gov/20124857/
Neill US. Skin care in the aging female: myths and truths. J Clin Invest. 2012;122(2):473–7. https://pubmed.ncbi.nlm.nih.gov/22293186/
Draelos ZD. Active agents in common skin care products. Plast Reconstr Surg. 2010;125(2):719–24. https://pubmed.ncbi.nlm.nih.gov/20124857/
Sunder S. Relevant topical skin care products for prevention and treatment of aging skin. Facial Plast Surg Clin North Am. 2019;27(3):413–8. https://pubmed.ncbi.nlm.nih.gov/31280856/
Bergstrom KG. Carrots before sticks: appealing to vanity promotes sun protection. J Drugs Dermatol. 2013;12(8):952–3. https://pubmed.ncbi.nlm.nih.gov/23986171/
Hughes MCB, Williams GM, Baker P, Green AC. Sunscreen and prevention of skin aging: a randomized trial. Ann Intern Med. 2013;158(11):781–90. https://pubmed.ncbi.nlm.nih.gov/23732711/
Draelos ZD. Active agents in common skin care products. Plast Reconstr Surg. 2010;125(2):719–24. https://pubmed.ncbi.nlm.nih.gov/20124857/
Neill US. Skin care in the aging female: myths and truths. J Clin Invest. 2012;122(2):473–7. https://pubmed.ncbi.nlm.nih.gov/22293186/
Lupo MP, Cole AL. Cosmeceutical peptides. Dermatol Ther. 2007;20(5):343–9. https://pubmed.ncbi.nlm.nih.gov/18045359/
Sunder S. Relevant topical skin care products for prevention and treatment of aging skin. Facial Plast Surg Clin North Am. 2019;27(3):413–8. https://pubmed.ncbi.nlm.nih.gov/31280856/
Ramos-e-Silva M, Celem LR, Ramos-e-Silva S, Fucci-da-Costa AP. Anti-aging cosmetics: facts and controversies. Clin Dermatol. 2013;31(6):750–8. https://pubmed.ncbi.nlm.nih.gov/24160281/
Zussman J, Ahdout J, Kim J. Vitamins and photoaging: do scientific data support their use? J Am Acad Dermatol. 2010;63(3):507–25. https://pubmed.ncbi.nlm.nih.gov/20189681/
Darlenski R, Surber C, Fluhr JW. Topical retinoids in the management of photodamaged skin: from theory to evidence-based practical approach. Br J Dermatol. 2010;163(6):1157–65. https://pubmed.ncbi.nlm.nih.gov/20633013/
Elmets CA. Long term topical tretinoin and excess mortality in older patients. NEJM Journal Watch. https://www.jwatch.org/jd200901300000001/2009/01/30/long-term-topical-tretinoin-and-excess-mortality. Published January 20, 2009. Accessed August 31, 2022.; https://www.jwatch.org/jd200901300000001/2009/01/30/long-term-topical-tretinoin-and-excess-mortality
Imhof L, Leuthard D. Topical over-the-counter antiaging agents: an update and systematic review. Dermatology. 2021;237(2):217–29. https://pubmed.ncbi.nlm.nih.gov/32882685/
Tran D, Townley JP, Barnes TM, Greive KA. An antiaging skin care system containing alpha hydroxy acids and vitamins improves the biomechanical parameters of facial skin. Clin Cosmet Investig Dermatol. 2015;8:9–17. https://pubmed.ncbi.nlm.nih.gov/25552908/
Bilal M, Iqbal HMN. An insight into toxicity and human-health-related adverse consequences of cosmeceuticals – a review. Sci Total Environ. 2019;670:555–68. https://pubmed.ncbi.nlm.nih.gov/30909033/
Neill US. Skin care in the aging female: myths and truths. J Clin Invest. 2012;122(2):473–7. https://pubmed.ncbi.nlm.nih.gov/22293186/
Bissett DL, Miyamoto K, Sun P, Li J, Berge CA. Topical niacinamide reduces yellowing, wrinkling, red blotchiness, and hyperpigmented spots in aging facial skin. Int J Cosmet Sci. 2004;26(5):231–8. https://pubmed.ncbi.nlm.nih.gov/18492135/
Levin J, Del Rosso JQ, Momin SB. How much do we really know about our favorite cosmeceutical ingredients? J Clin Aesthet Dermatol. 2010;3(2):22–41. https://pubmed.ncbi.nlm.nih.gov/20725560/
Imhof L, Leuthard D. Topical over-the-counter antiaging agents: an update and systematic review. Dermatology. 2021;237(2):217–29. https://pubmed.ncbi.nlm.nih.gov/32882685/
Sivamani RK, Jagdeo JR, Elsner P, Maibach HI, eds. Cosmeceuticals and Active Cosmetics. 3rd ed. CRC Press; 2016. https://worldcat.org/title/919719500
Bissett DL, Miyamoto K, Sun P, Li J, Berge CA. Topical niacinamide reduces yellowing, wrinkling, red blotchiness, and hyperpigmented spots in aging facial skin. Int J Cosmet Sci. 2004;26(5):231–8. https://pubmed.ncbi.nlm.nih.gov/18492135/
Hunt KJ, Hung SK, Ernst E. Botanical extracts as anti-aging preparations for the skin: a systematic review. Drugs Aging. 2010;27(12):973–85. https://pubmed.ncbi.nlm.nih.gov/21087067/
Bissett DL, Miyamoto K, Sun P, Li J, Berge CA. Topical niacinamide reduces yellowing, wrinkling, red blotchiness, and hyperpigmented spots in aging facial skin. Int J Cosmet Sci. 2004;26(5):231–8. https://pubmed.ncbi.nlm.nih.gov/18492135/
Bissett DL, Oblong JE, Berge CA. Niacinamide: A B vitamin that improves aging facial skin appearance. Dermatol Surg. 2005;31(7 Pt 2):860–5; discussion 865. https://pubmed.ncbi.nlm.nih.gov/16029679/
Bissett DL, Oblong JE, Berge CA. Niacinamide: A B vitamin that improves aging facial skin appearance. Dermatol Surg. 2005;31(7 Pt 2):860–5. https://pubmed.ncbi.nlm.nih.gov/16029679/
Zussman J, Ahdout J, Kim J. Vitamins and photoaging: do scientific data support their use? J Am Acad Dermatol. 2010;63(3):507–25. https://pubmed.ncbi.nlm.nih.gov/20189681/
Chiu P-C, Chan C–C, Lin H-M, Chiu H-C. The clinical anti-aging effects of topical kinetin and niacinamide in Asians: a randomized, double-blind, placebo-controlled, split-face comparative trial. J Cosmet Dermatol. 2007;6(4):243–9. https://pubmed.ncbi.nlm.nih.gov/18047609/
Kawada A, Konishi N, Oiso N, Kawara S, Date A. Evaluation of anti-wrinkle effects of a novel cosmetic containing niacinamide. J Dermatol. 2008;35(10):637–42. https://pubmed.ncbi.nlm.nih.gov/19017042/
Burke KE, Clive J, Combs GF, Commisso J, Keen CL, Nakamura RM. Effects of topical and oral vitamin E on pigmentation and skin cancer induced by ultraviolet irradiation in Skh:2 hairless mice. Nutr Cancer. 2000;38(1):87–97. https://pubmed.ncbi.nlm.nih.gov/11341050/
Sunder S. Relevant topical skin care products for prevention and treatment of aging skin. Facial Plast Surg Clin North Am. 2019;27(3):413–8. https://pubmed.ncbi.nlm.nih.gov/31280856/
Imhof L, Leuthard D. Topical over-the-counter antiaging agents: an update and systematic review. Dermatology. 2021;237(2):217–29. https://pubmed.ncbi.nlm.nih.gov/32882685/
Inui M, Ooe M, Fujii K, Matsunaka H, Yoshida M, Ichihashi M. Mechanisms of inhibitory effects of CoQ10 on UVB-induced wrinkle formation in vitro and in vivo. Biofactors. 2008;32(1–4):237–43. https://pubmed.ncbi.nlm.nih.gov/19096121/
Elsner P, Fluhr JW, Gehring W, et al. Anti-aging data and support claims – consensus statement. J Dtsch Dermatol Ges. 2011;9 Suppl 3:S1–32. https://pubmed.ncbi.nlm.nih.gov/22023288/
Nusgens BV, Humbert P, Rougier A, et al. Topically applied vitamin C enhances the mRNA level of collagens I and III, their processing enzymes and tissue inhibitor of matrix metalloproteinase 1 in the human dermis. J Invest Dermatol. 2001;116(6):853–9. https://pubmed.ncbi.nlm.nih.gov/11407971/
Traikovich SS. Use of topical ascorbic acid and its effects on photodamaged skin topography. Arch Otolaryngol Head Neck Surg. 1999;125(10):1091–8. https://pubmed.ncbi.nlm.nih.gov/10522500/
Sivamani RK, Jagdeo JR, Elsner P, Maibach HI, eds. Cosmeceuticals and Active Cosmetics. 3rd ed. CRC Press; 2016. https://worldcat.org/title/919719500
Traikovich SS. Use of topical ascorbic acid and its effects on photodamaged skin topography. Arch Otolaryngol Head Neck Surg. 1999;125(10):1091–8. https://pubmed.ncbi.nlm.nih.gov/10522500/
Raschke T, Koop U, Düsing HJ, et al. Topical activity of ascorbic acid: from in vitro optimization to in vivo efficacy. Skin Pharmacol Physiol. 2004;17(4):200–6. https://pubmed.ncbi.nlm.nih.gov/15258452/
Humbert PG, Haftek M, Creidi P, et al. Topical ascorbic acid on photoaged skin. Clinical, topographical and ultrastructural evaluation: double-blind study vs. placebo. Exp Dermatol. 2003;12(3):237–44. https://pubmed.ncbi.nlm.nih.gov/12823436/
High potency serum. Cellex-C. https://www.cellexcusa.com/products/high-potency-serum. Accessed September 7, 2022.; https://www.cellexcusa.com/products/high-potency-serum
Sunder S. Relevant topical skin care products for prevention and treatment of aging skin. Facial Plast Surg Clin North Am. 2019;27(3):413–8. https://pubmed.ncbi.nlm.nih.gov/31280856/
Huang CK, Miller TA. The truth about over-the-counter topical anti-aging products: a comprehensive review. Aesthet Surg J. 2007;27(4):402–12. https://pubmed.ncbi.nlm.nih.gov/19341668/
Alpha hydroxy acids. U.S. Food & Drug Administration. https://www.fda.gov/cosmetics/cosmetic-ingredients/alpha-hydroxy-acids#q6. Updated February 25, 2022. Accessed September 7, 2022.; https://www.fda.gov/cosmetics/cosmetic-ingredients/alpha-hydroxy-acids#q6
Gordon R. Skin cancer: an overview of epidemiology and risk factors. Semin Oncol Nurs. 2013;29(3):160–9. https://pubmed.ncbi.nlm.nih.gov/23958214/
Garcovich S, Colloca G, Sollena P, et al. Skin cancer epidemics in the elderly as an emerging issue in geriatric oncology. Aging Dis. 2017;8(5):643–61. https://pubmed.ncbi.nlm.nih.gov/28966807/
Strauss DG, Michele TM. Skin cancer prevention and sunscreen safety: commentary on American Society of Clinical Oncology policy statement on skin cancer prevention. JCO Oncol Pract. 2020;16(8):436–8. https://pubmed.ncbi.nlm.nih.gov/32603257/
Johnson MM, Leachman SA, Aspinwall LG, et al. Skin cancer screening: recommendations for data-driven screening guidelines and a review of the US Preventive Services Task Force controversy. Melanoma Manag. 2017;4(1):13–37. https://pubmed.ncbi.nlm.nih.gov/28758010/
Wernli KJ, Henrikson NB, Morrison CC, Nguyen M, Pocobelli G, Blasi PR. Screening for skin cancer in adults: updated evidence report and systematic review for the US Preventive Services Task Force. JAMA. 2016;316(4):436–47. https://pubmed.ncbi.nlm.nih.gov/27583318/
Katalinic A, Waldmann A, Weinstock MA, et al. Does skin cancer screening save lives?: an observational study comparing trends in melanoma mortality in regions with and without screening. Cancer. 2012;118(21):5395–402. https://pubmed.ncbi.nlm.nih.gov/22517033/
Boniol M, Autier P, Gandini S. Melanoma mortality following skin cancer screening in Germany. BMJ Open. 2015;5(9):e008158. https://pubmed.ncbi.nlm.nih.gov/26373399/
Brenner H. Mortality from malignant melanoma in an era of nationwide skin cancer screening. Dtsch Arztebl Int. 2015;112(38):627–8. https://pubmed.ncbi.nlm.nih.gov/26429633/
Boniol M, Autier P, Gandini S. Melanoma mortality following skin cancer screening in Germany. BMJ Open. 2015;5(9):e008158. https://pubmed.ncbi.nlm.nih.gov/26373399/
Wernli KJ, Henrikson NB, Morrison CC, Nguyen M, Pocobelli G, Blasi PR. Screening for skin cancer in adults: updated evidence report and systematic review for the US Preventive Services Task Force. JAMA. 2016;316(4):436–47. https://pubmed.ncbi.nlm.nih.gov/27583318/
Halpern AC, Marghoob AA, Reiter O. Melanoma warning signs: what you need to know about early signs of skin cancer. Skin Cancer Foundation. https://www.skincancer.org/skin-cancer-information/melanoma/melanoma-warning-signs-and-images/. Updated January 2021. Accessed September 7, 2022.; https://www.skincancer.org/skin-cancer-information/melanoma/melanoma-warning-signs-and-images/
Linos E, Katz KA, Colditz GA. Skin cancer – the importance of prevention. JAMA Intern Med. 2016;176(10):1435–6. https://pubmed.ncbi.nlm.nih.gov/27459394/
Burke KE, Clive J, Combs GF, Commisso J, Keen CL, Nakamura RM. Effects of topical and oral vitamin E on pigmentation and skin cancer induced by ultraviolet irradiation in Skh:2 hairless mice. Nutr Cancer. 2000;38(1):87–97. https://pubmed.ncbi.nlm.nih.gov/11341050/
Stern RS, Weinstein MC, Baker SG. Risk reduction for nonmelanoma skin cancer with childhood sunscreen use. Arch Dermatol. 1986;122(5):537–45. https://pubmed.ncbi.nlm.nih.gov/3707169/
Strauss DG, Michele TM. Skin cancer prevention and sunscreen safety: commentary on American Society of Clinical Oncology policy statement on skin cancer prevention. JCO Oncol Pract. 2020;16(8):436–8. https://pubmed.ncbi.nlm.nih.gov/32603257/
Li H, Colantonio S, Dawson A, Lin X, Beecker J. Sunscreen application, safety, and sun protection: the evidence. J Cutan Med Surg. 2019;23(4):357–69. https://pubmed.ncbi.nlm.nih.gov/31219707/
Strauss DG, Michele TM. Skin cancer prevention and sunscreen safety: commentary on American Society of Clinical Oncology policy statement on skin cancer prevention. JCO Oncol Pract. 2020;16(8):436–8. https://pubmed.ncbi.nlm.nih.gov/32603257/
Li H, Colantonio S, Dawson A, Lin X, Beecker J. Sunscreen application, safety, and sun protection: the evidence. J Cutan Med Surg. 2019;23(4):357–69. https://pubmed.ncbi.nlm.nih.gov/31219707/
Boo YC. Mechanistic basis and clinical evidence for the applications of nicotinamide (niacinamide) to control skin aging and pigmentation. Antioxidants (Basel). 2021;10(8):1315. https://pubmed.ncbi.nlm.nih.gov/34439563/
Surjana D, Halliday GM, Martin AJ, Moloney FJ, Damian DL. Oral nicotinamide reduces actinic keratoses in phase II double-blinded randomized controlled trials. J Invest Dermatol. 2012;132(5):1497–500. https://pubmed.ncbi.nlm.nih.gov/22297641/
Chen AC, Martin AJ, Choy B, et al. A phase 3 randomized trial of nicotinamide for skin-cancer chemoprevention. N Engl J Med. 2015;373(17):1618–26. https://pubmed.ncbi.nlm.nih.gov/26488693/
Byrne SN. How much sunlight is enough? Photochem Photobiol Sci. 2014;13(6):840–52. https://pubmed.ncbi.nlm.nih.gov/24770340/
Tsai J, Chien AL. Photoprotection for skin of color. Am J Clin Dermatol. 2022;23(2):195–205. https://pubmed.ncbi.nlm.nih.gov/35044638/
Strauss DG, Michele TM. Skin cancer prevention and sunscreen safety: commentary on American Society of Clinical Oncology policy statement on skin cancer prevention. JCO Oncol Pract. 2020;16(8):436–8. https://pubmed.ncbi.nlm.nih.gov/32603257/
Purdue MP, Beane Freeman LE, Anderson WF, Tucker MA. Recent trends in incidence of cutaneous melanoma among US Caucasian young adults. J Invest Dermatol. 2008;128(12):2905–8. https://pubmed.ncbi.nlm.nih.gov/18615112/
Boniol M, Autier P, Boyle P, Gandini S. Cutaneous melanoma attributable to sunbed use: systematic review and meta-analysis. BMJ. 2012;345:e4757. https://pubmed.ncbi.nlm.nih.gov/22833605/
Weinstock MA, Moses AM. Skin cancer meets vitamin D: the way forward for dermatology and public health. J Am Acad Dermatol. 2009;61(4):720–4. https://pubmed.ncbi.nlm.nih.gov/19751885/
Dahl MV. Sun exposure, vitamin D metabolism, and skin cancer. Mayo Clin Proc. 2004;79(5):699–700. https://pubmed.ncbi.nlm.nih.gov/15132420/
Grant WB, Garland CF, Holick MF. Comparisons of estimated economic burdens due to insufficient solar ultraviolet irradiance and vitamin D and excess solar UV irradiance for the United States. Photochem Photobiol. 2005;81(6):1276–86. https://pubmed.ncbi.nlm.nih.gov/16159309/
Grant WB. In defense of the sun: an estimate of changes in mortality rates in the United States if mean serum 25-hydroxyvitamin D levels were raised to 45 ng/mL by solar ultraviolet-B irradiance. Dermatoendocrinol. 2009;1(4):207–14. https://pubmed.ncbi.nlm.nih.gov/20592792/
Dahl MV. Sun exposure, vitamin D metabolism, and skin cancer. Mayo Clin Proc. 2004;79(5):699–700. https://pubmed.ncbi.nlm.nih.gov/15132420/
Gilchrest BA. Sun protection and vitamin D: three dimensions of obfuscation. J Steroid Biochem Mol Biol. 2007;103(3–5):655–63. https://pubmed.ncbi.nlm.nih.gov/17222550/
Citrin DL, Bloom DL, Grutsch JF, Mortensen SJ, Lis CG. Beliefs and perceptions of women with newly diagnosed breast cancer who refused conventional treatment in favor of alternative therapies. Oncologist. 2012;17(5):607–12. https://pubmed.ncbi.nlm.nih.gov/22531358/
Li H, Colantonio S, Dawson A, Lin X, Beecker J. Sunscreen application, safety, and sun protection: the evidence. J Cutan Med Surg. 2019;23(4):357–69. https://pubmed.ncbi.nlm.nih.gov/31219707/
Phillips TJ, Bhawan J, Yaar M, Bello Y, Lopiccolo D, Nash JF. Effect of daily versus intermittent sunscreen application on solar simulated UV radiation-induced skin response in humans. J Am Acad Dermatol. 2000;43(4):610–8. https://pubmed.ncbi.nlm.nih.gov/11004615/
Krutmann J, Berking C, Berneburg M, Diepgen TL, Dirschka T, Szeimies M. New strategies in the prevention of actinic keratosis: a critical review. Skin Pharmacol Physiol. 2015;28(6):281–9. https://pubmed.ncbi.nlm.nih.gov/11004615/
Krutmann J, Berking C, Berneburg M, Diepgen TL, Dirschka T, Szeimies M. New strategies in the prevention of actinic keratosis: a critical review. Skin Pharmacol Physiol. 2015;28(6):281–9. https://pubmed.ncbi.nlm.nih.gov/11004615/
Ramos-e-Silva M, Celem LR, Ramos-e-Silva S, Fucci-da-Costa AP. Anti-aging cosmetics: facts and controversies. Clin Dermatol. 2013;31(6):750–8. https://pubmed.ncbi.nlm.nih.gov/24160281/
Buller DB, Andersen PA, Walkosz BJ, et al. Compliance with sunscreen advice in a survey of adults engaged in outdoor winter recreation at high-elevation ski areas. J Am Acad Dermatol. 2012;66(1):63–70. https://pubmed.ncbi.nlm.nih.gov/21742410/
Isedeh P, Osterwalder U, Lim HW. Teaspoon rule revisited: proper amount of sunscreen application: Letter to the Editor. Photodermatol Photoimmunol Photomed. 2013;29(1):55–6. https://pubmed.ncbi.nlm.nih.gov/23281699/
Li H, Colantonio S, Dawson A, Lin X, Beecker J. Sunscreen application, safety, and sun protection: the evidence. J Cutan Med Surg. 2019;23(4):357–69. https://pubmed.ncbi.nlm.nih.gov/31219707/
Kligman LH, Akin FJ, Kligman AM. Sunscreens prevent ultraviolet photocarcinogenesis. J Am Acad Dermatol. 1980;3(1):30–5. https://pubmed.ncbi.nlm.nih.gov/6967495/
Calbó J, Pagès D, González JA. Empirical studies of cloud effects on UV radiation: a review. Rev Geophys. 2005;43(2). https://agupubs.onlinelibrary.wiley.com/doi/full/10.1029/2004RG000155
de Gálvez MV, Aguilera J, Buendía EA, Sánchez-Roldán C, Herrera-Ceballos E. Time required for a standard sunscreen to become effective following application: a UV photography study. J Eur Acad Dermatol Venereol. 2018;32(4):e123–4. https://pubmed.ncbi.nlm.nih.gov/29024154/
Stokes RP, Diffey BL. The water resistance of sunscreen and day-care products. Br J Dermatol. 1999;140(2):259–63. https://pubmed.ncbi.nlm.nih.gov/10233219/
Stokes RP, Diffey BL. A novel ex vivo technique to assess the sand/rub resistance of sunscreen products. Int J Cosmet Sci. 2000;22(5):329–34. https://pubmed.ncbi.nlm.nih.gov/18503420/
Tsai J, Chien AL. Photoprotection for skin of color. Am J Clin Dermatol. 2022;23(2):195–205. https://pubmed.ncbi.nlm.nih.gov/35044638/
Kaidbey KH, Agin PP, Sayre RM, Kligman AM. Photoprotection by melanin – a comparison of black and Caucasian skin. J Am Acad Dermatol. 1979;1(3):249–60. https://pubmed.ncbi.nlm.nih.gov/512075/
Sander M, Sander M, Burbidge T, Beecker J. The efficacy and safety of sunscreen use for the prevention of skin cancer. CMAJ. 2020;192(50):E1802–8. https://pubmed.ncbi.nlm.nih.gov/33318091/
National Cancer Institute. Sun-protective behavior. Cancer Trends Progress Report. https://progressreport.cancer.gov/prevention/sun_protection. Updated April 2022. Accessed Nov 19, 2022.; https://progressreport.cancer.gov/prevention/sun_protection
Tsai J, Chien AL. Photoprotection for skin of color. Am J Clin Dermatol. 2022;23(2):195–205. https://pubmed.ncbi.nlm.nih.gov/35044638/
Taylor SC, Alexis AF, Armstrong AW, Chiesa Fuxench ZC, Lim HW. Misconceptions of photoprotection in skin of color. J Am Acad Dermatol. 2022;86(3S):S9–17. https://pubmed.ncbi.nlm.nih.gov/34942293/
Tsai J, Chien AL. Photoprotection for skin of color. Am J Clin Dermatol. 2022;23(2):195–205. https://pubmed.ncbi.nlm.nih.gov/35044638/
Sander M, Sander M, Burbidge T, Beecker J. The efficacy and safety of sunscreen use for the prevention of skin cancer. CMAJ. 2020;192(50):E1802–8. https://pubmed.ncbi.nlm.nih.gov/33318091/
Barr J. Spray-on sunscreens need a good rub. J Am Acad Dermatol. 2005;52(1):180–1. https://pubmed.ncbi.nlm.nih.gov/15627116/
Sander M, Sander M, Burbidge T, Beecker J. The efficacy and safety of sunscreen use for the prevention of skin cancer. CMAJ. 2020;192(50):E1802–8. https://pubmed.ncbi.nlm.nih.gov/33318091/
Pearce K, Goldsmith WT, Greenwald R, Yang C, Mainelis G, Wright C. Characterization of an aerosol generation system to assess inhalation risks of aerosolized nano-enabled consumer products. Inhal Toxicol. 2019;31(9–10):357–67. https://pubmed.ncbi.nlm.nih.gov/31779509/
Food and Drug Administration, Department of Health and Human Services. 21 CFR parts 201, 310, 347, and 352. Sunscreen drug products for over-the-counter human use. Federal Register. 2019;84(38):6205–75. https://www.federalregister.gov/documents/2019/02/26/2019-03019/sunscreen-drug-products-for-over-the-counter-human-use
Black HS, Lenger WA, Gerguis J, Thornby JI. Relation of antioxidants and level of dietary lipid to epidermal lipid peroxidation and ultraviolet carcinogenesis. Cancer Res. 1985;45(12 Pt 1):6254–9. https://pubmed.ncbi.nlm.nih.gov/4063976/
Ibiebele TI, van der Pols JC, Hughes MC, Marks GC, Williams GM, Green AC. Dietary pattern in association with squamous cell carcinoma of the skin: a prospective study. Am J Clin Nutr. 2007;85(5):1401–8. https://pubmed.ncbi.nlm.nih.gov/17490979/
Black HS, Thornby JI, Wolf JE Jr, et al. Evidence that a low-fat diet reduces the occurrence of non-melanoma skin cancer. Int J Cancer. 1995;62(2):165–9. https://pubmed.ncbi.nlm.nih.gov/7622291/
Lumley E, Phillips P, Aber A, Buckley-Woods H, Jones GL, Michaels JA. Experiences of living with varicose veins: a systematic review of qualitative research. J Clin Nurs. 2019;28(7–8):1085–99. https://pubmed.ncbi.nlm.nih.gov/30461103/
Meissner MH. What is the medical rationale for the treatment of varicose veins? Phlebology. 2012;27(Suppl 1):27–33. https://pubmed.ncbi.nlm.nih.gov/22312064/
Raetz J, Wilson M, Collins K. Varicose veins: diagnosis and treatment. Am Fam Physician. 2019;99(11):682–8. https://pubmed.ncbi.nlm.nih.gov/31150188/
Atik D, Atik C, Karatepe C. The effect of external apple vinegar application on varicosity symptoms, pain, and social appearance anxiety: a randomized controlled trial. Evid Based Complement Alternat Med. 2016;2016:6473678. https://pubmed.ncbi.nlm.nih.gov/26881006/
Montgomery L, Seys J, Mees J. To pee, or not to pee: a review on envenomation and treatment in European jellyfish species. Mar Drugs. 2016;14(7):127. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4962017/
Wilcox CL, Headlam JL, Doyle TK, Yanagihara AA. Assessing the efficacy of first-aid measures in Physalia sp. Envenomation, using solution– and blood agarose-based models. Toxins (Basel). 2017;9(5):149. https://pubmed.ncbi.nlm.nih.gov/28445412/
Luu LA, Flowers RH, Kellams AL, et al. Apple cider vinegar soaks [0.5 %] as a treatment for atopic dermatitis do not improve skin barrier integrity. Pediatr Dermatol. 2019;36(5):634–9. https://pubmed.ncbi.nlm.nih.gov/31328306/
Feldstein S, Afshar M, Krakowski AC. Chemical burn from vinegar following an internet-based protocol for self-removal of nevi. J Clin Aesthet Dermatol. 2015;8(6):50. https://pubmed.ncbi.nlm.nih.gov/26155328/
Kroeger CM, Brown AW, Allison DB. Differences in Nominal Significance (DINS) Error leads to invalid conclusions: letter regarding, “Diet enriched with fresh coconut decreases blood glucose levels and body weight in normal adults.” J Complement Integr Med. 2019;16(2):/j/jcim.2019.16.issue-2/jcim-2018–0037/jcim-2018–0037.xml. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6957306/
Richardson JB, Dixon M. Varicose veins in tropical Africa. Lancet. 1977;1(8015):791–2. https://pubmed.ncbi.nlm.nih.gov/66583/
O’Keefe SJD, Chung D, Mahmoud N, et al. Why do African Americans get more colon cancer than Native Africans? J Nutr. 2007;137(1 Suppl):175S-82S. https://pubmed.ncbi.nlm.nih.gov/17182822/
Tuohy KM, Gougoulias C, Shen Q, Walton G, Fava F, Ramnani P. Studying the human gut microbiota in the trans-omics era – focus on metagenomics and metabonomics. Curr Pharm Des. 2009;15(13):1415–27. https://pubmed.ncbi.nlm.nih.gov/19442166/
Burkitt DP. Two blind spots in medical knowledge. Nurs Times. 1976;72(1):24–7. https://pubmed.ncbi.nlm.nih.gov/54904/
Crowe FL, Appleby PN, Allen NE, Key TJ. Diet and risk of diverticular disease in Oxford cohort of European Prospective Investigation into Cancer and Nutrition (EPIC): prospective study of British vegetarians and non-vegetarians. BMJ. 2011;343:d4131. https://pubmed.ncbi.nlm.nih.gov/21771850/
Knutsen SF. Lifestyle and the use of health services. Am J Clin Nutr. 1994;59(5 Suppl):1171S-5S. https://pubmed.ncbi.nlm.nih.gov/8172119/
Eddy TP, Taylor GF. Sublingual varicosities and vitamin C in elderly vegetarians. Age Ageing. 1977;6(1):6–13. https://pubmed.ncbi.nlm.nih.gov/842407/
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
Halteh P, Scher RK, Lipner SR. Over-the-counter and natural remedies for onychomycosis: do they really work? Cutis. 2016;98(5):E16–25. https://pubmed.ncbi.nlm.nih.gov/28040821/
Halteh P, Scher RK, Lipner SR. Over-the-counter and natural remedies for onychomycosis: do they really work? Cutis. 2016;98(5):E16–25. https://pubmed.ncbi.nlm.nih.gov/28040821/
Murdan S. Nail disorders in older people, and aspects of their pharmaceutical treatment. Int J Pharm. 2016;512(2):405–11. https://pubmed.ncbi.nlm.nih.gov/27180233/
Iorizzo M. Tips to treat the 5 most common nail disorders: brittle nails, onycholysis, paronychia, psoriasis, onychomycosis. Dermatol Clin. 2015;33(2):175–83. https://pubmed.ncbi.nlm.nih.gov/25828710/
Maddy AJ, Tosti A. Hair and nail diseases in the mature patient. Clin Dermatol. 2018;36(2):159–66. https://pubmed.ncbi.nlm.nih.gov/29566920/
Iorizzo M. Tips to treat the 5 most common nail disorders: brittle nails, onycholysis, paronychia, psoriasis, onychomycosis. Dermatol Clin. 2015;33(2):175–83. https://pubmed.ncbi.nlm.nih.gov/25828710/
Murdan S. Nail disorders in older people, and aspects of their pharmaceutical treatment. Int J Pharm. 2016;512(2):405–11. https://pubmed.ncbi.nlm.nih.gov/27180233/
Gupta AK, Konnikov N, Lynde CW. Single-blind, randomized, prospective study on terbinafine and itraconazole for treatment of dermatophyte toenail onychomycosis in the elderly. J Am Acad Dermatol. 2001;44(3):479–84. https://pubmed.ncbi.nlm.nih.gov/11209118/
Murdan S. Nail disorders in older people, and aspects of their pharmaceutical treatment. Int J Pharm. 2016;512(2):405–11. https://pubmed.ncbi.nlm.nih.gov/27180233/
Gupta AK, Daigle D, Foley KA. Network meta-analysis of onychomycosis treatments. Skin Appendage Disord. 2015;1(2):74–81. https://pubmed.ncbi.nlm.nih.gov/27170937/
Murdan S. Nail disorders in older people, and aspects of their pharmaceutical treatment. Int J Pharm. 2016;512(2):405–11. https://pubmed.ncbi.nlm.nih.gov/27180233/
Iorizzo M. Tips to treat the 5 most common nail disorders: brittle nails, onycholysis, paronychia, psoriasis, onychomycosis. Dermatol Clin. 2015;33(2):175–83. https://pubmed.ncbi.nlm.nih.gov/25828710/
Murdan S. Nail disorders in older people, and aspects of their pharmaceutical treatment. Int J Pharm. 2016;512(2):405–11. https://pubmed.ncbi.nlm.nih.gov/27180233/
Sleven R, Lanckacker E, Delputte P, Maes L, Cos P. Evaluation of topical antifungal products in an in vitro onychomycosis model. Mycoses. 2016;59(5):327–30. https://pubmed.ncbi.nlm.nih.gov/26857689/
Kelly S, Liu D, Wang T, Rajpara A, Franano C, Aires D. Vinegar sock soak for tinea pedis or onychomycosis. J Am Acad Dermatol. Published online September 22, 2017:S0190–9622(17)32448–9.; https://pubmed.ncbi.nlm.nih.gov/28947288/
Satchell AC, Saurajen A, Bell C, Barnetson RSC. Treatment of dandruff with 5 % tea tree oil shampoo. J Am Acad Dermatol. 2002;47(6):852–5. https://pubmed.ncbi.nlm.nih.gov/12451368/
Satchell AC, Saurajen A, Bell C, Barnetson RSC. Treatment of interdigital tinea pedis with 25 % and 50 % tea tree oil solution: a randomized, placebo-controlled, blinded study. Australas J Dermatol. 2002;43(3):175–8. https://pubmed.ncbi.nlm.nih.gov/12121393/
Buck DS, Nidorf DM, Addino JG. Comparison of two topical preparations for the treatment of onychomycosis: Melaleuca alternifolia (tea tree) oil and clotrimazole. J Fam Pract. 1994;38(6):601–5. https://pubmed.ncbi.nlm.nih.gov/8195735/
Murdan S. Nail disorders in older people, and aspects of their pharmaceutical treatment. Int J Pharm. 2016;512(2):405–11. https://pubmed.ncbi.nlm.nih.gov/27180233/
Maddy AJ, Tosti A. Hair and nail diseases in the mature patient. Clin Dermatol. 2018;36(2):159–66. https://pubmed.ncbi.nlm.nih.gov/29566920/
Ingrown toenails. American Academy of Family Physicians. https://www.aafp.org/dam/brand/aafp/pubs/afp/issues/2019/0801/p158-s1.pdf. Updated August 2019. Accessed September 1, 2022.; https://www.aafp.org/dam/brand/aafp/pubs/afp/issues/2019/0801/p158-s1.pdf
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
Ilfeld FW. Ingrown toenail treated with cotton collodion insert. Foot Ankle. 1991;11(5):312–3. https://pubmed.ncbi.nlm.nih.gov/2037270/
Ingrown toenails. American Academy of Family Physicians. https://www.aafp.org/dam/brand/aafp/pubs/afp/issues/2019/0801/p158-s1.pdf. Updated August 2019. Accessed September 1, 2022.; https://www.aafp.org/dam/brand/aafp/pubs/afp/issues/2019/0801/p158-s1.pdf
Ingrown toenails. American Academy of Family Physicians. https://www.aafp.org/dam/brand/aafp/pubs/afp/issues/2019/0801/p158-s1.pdf. Updated August 2019. Accessed September 1, 2022.; https://www.aafp.org/dam/brand/aafp/pubs/afp/issues/2019/0801/p158-s1.pdf
Ilfeld FW. Ingrown toenail treated with cotton collodion insert. Foot Ankle. 1991;11(5):312–3. https://pubmed.ncbi.nlm.nih.gov/2037270/
Gutiérrez-Mendoza D, De Anda-Juárez M, Ávalos VF, Martínez GR, Domínguez-Cherit J. “Cotton nail cast”: a simple solution for mild and painful lateral and distal nail embedding. Dermatol Surg. 2015;41(3):411–4. https://pubmed.ncbi.nlm.nih.gov/25738445/
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
Maddy AJ, Tosti A. Hair and nail diseases in the mature patient. Clin Dermatol. 2018;36(2):159–66. https://pubmed.ncbi.nlm.nih.gov/29566920/
Klafke GB, da Silva RA, de Pellegrin KT, Xavier MO. Analysis of the role of nail polish in the transmission of onychomycosis. An Bras Dermatol. 2018;93:930–1. https://pubmed.ncbi.nlm.nih.gov/30484548/
Shemer A, Trau H, Davidovici B, Grunwald MH, Amichai B. Onycomycosis due to artificial nails. J Eur Acad Dermatol Venereol. 2008;22(8):998–1000. https://pubmed.ncbi.nlm.nih.gov/18355194/
Kechijian P. Dangers of acrylic fingernails. JAMA.1990;263(3):458 https://jamanetwork.com/journals/jama/article-abstract/381522
Dimitris R, Ralph D. Management of simple brittle nails. Dermatol Ther. 2012;25(6):569–73. https://pubmed.ncbi.nlm.nih.gov/23210755/
Buck DS, Nidorf DM, Addino JG. Comparison of two topical preparations for the treatment of onychomycosis: Melaleuca alternifolia (tea tree) oil and clotrimazole. J Fam Pract. 1994;38(6):601–5. https://pubmed.ncbi.nlm.nih.gov/8195735/
Kuo PT, Whereat AF, Horwitz O. The effect of lipemia upon coronary and peripheral arterial circulation in patients with essential hyperlipemia. Am J Med. 1959;26(1):68–75. https://pubmed.ncbi.nlm.nih.gov/13606154/
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
Nail care products. U.S. Food & Drug Administration. https://www.fda.gov/cosmetics/cosmetic-products/nail-care-products#forma. Published February 25, 2022. Accessed September 7, 2022.; https://www.fda.gov/cosmetics/cosmetic-products/nail-care-products#forma
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
van de Kerkhof PCM, Pasch MC, Scher RK, et al. Brittle nail syndrome: a pathogenesis-based approach with a proposed grading system. J Am Acad Dermatol. 2005;53(4):644–51. https://pubmed.ncbi.nlm.nih.gov/16198786/
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
Cashman MW, Sloan SB. Nutrition and nail disease. Clin Dermatol. 2010;28(4):420–5. https://pubmed.ncbi.nlm.nih.gov/20620759/
Maddy AJ, Tosti A. Hair and nail diseases in the mature patient. Clin Dermatol. 2018;36(2):159–66. https://pubmed.ncbi.nlm.nih.gov/29566920/
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
Stern DK, Diamantis S, Smith E, et al. Water content and other aspects of brittle versus normal fingernails. J Am Acad Dermatol. 2007;57(1):31–6. https://pubmed.ncbi.nlm.nih.gov/17412454/
Stern DK, Diamantis S, Smith E, et al. Water content and other aspects of brittle versus normal fingernails. J Am Acad Dermatol. 2007;57(1):31–6. https://pubmed.ncbi.nlm.nih.gov/17412454/
Dimitris R, Ralph D. Management of simple brittle nails. Dermatol Ther. 2012;25(6):569–73. https://pubmed.ncbi.nlm.nih.gov/23210755/
Stern DK, Diamantis S, Smith E, et al. Water content and other aspects of brittle versus normal fingernails. J Am Acad Dermatol. 2007;57(1):31–6. https://pubmed.ncbi.nlm.nih.gov/17412454/
Dimitris R, Ralph D. Management of simple brittle nails. Dermatol Ther. 2012;25(6):569–73. https://pubmed.ncbi.nlm.nih.gov/23210755/
Yousif J, Farshchian M, Potts GA. Oral nail growth supplements: a comprehensive review. Int J Dermatol. 2022;61(8):916–22. https://pubmed.ncbi.nlm.nih.gov/34351622/
Lipner SR, Scher RK. Biotin for the treatment of nail disease: what is the evidence? J Dermatolog Treat. 2018;29(4):411–4. https://pubmed.ncbi.nlm.nih.gov/29057689/
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
Patel DP, Swink SM, Castelo-Soccio L. A review of the use of biotin for hair loss. Skin Appendage Disord. 2017;3(3):166–9. https://pubmed.ncbi.nlm.nih.gov/28879195/
Reilly JD, Cottrell DF, Martin RJ, Cuddeford DJ. Effect of supplementary dietary biotin on hoof growth and hoof growth rate in ponies: a controlled trial. Equine Vet J Suppl. 1998;(26):51–7. https://pubmed.ncbi.nlm.nih.gov/9932094/
Floersheim GL. [Treatment of brittle fingernails with biotin]. Z Hautkr. 1989;64(1):41–8. https://pubmed.ncbi.nlm.nih.gov/2648686/
Colombo VE, Gerber F, Bronhofer M, Floersheim GL. Treatment of brittle fingernails and onychoschizia with biotin: scanning electron microscopy. J Am Acad Dermatol. 1990;23(6 Pt 1):1127–32. https://pubmed.ncbi.nlm.nih.gov/2273113/
Chiavetta A, Mazzurco S, Secolo MP, Tomarchio G, Milani M. Treatment of brittle nail with a hydroxypropyl chitosan-based lacquer, alone or in combination with oral biotin: a randomized, assessor-blinded trial. Dermatol Ther. 2019;32(5):e13028. https://pubmed.ncbi.nlm.nih.gov/31344296/
Bowen R, Benavides R, Colón-Franco JM, et al. Best practices in mitigating the risk of biotin interference with laboratory testing. Clin Biochem. 2019;74:1–11. https://pubmed.ncbi.nlm.nih.gov/31473202/
Lipner SR. Update on biotin therapy in dermatology: time for a change. J Drugs Dermatol. 2020;19(12):1264–5. https://pubmed.ncbi.nlm.nih.gov/33346513/
John JJ, Cooley V, Lipner SR. Assessment of biotin supplementation among patients in an outpatient dermatology clinic. J Am Acad Dermatol. 2019;81(2):620–1. https://pubmed.ncbi.nlm.nih.gov/30630025/
Waqas B, Wu A, Yim E, Lipner SR. A survey-based study of physician practices regarding biotin supplementation. J Dermatolog Treat. 2022;33(1):573–4. https://pubmed.ncbi.nlm.nih.gov/32419559/
Reinecke JK, Hinshaw MA. Nail health in women. Int J Womens Dermatol. 2020;6(2):73–9. https://pubmed.ncbi.nlm.nih.gov/32258335/
Yousif J, Farshchian M, Potts GA. Oral nail growth supplements: a comprehensive review. Int J Dermatol. 2022;61(8):916–22. https://pubmed.ncbi.nlm.nih.gov/34351622/
Eke PI, Dye BA, Wei L, et al. Update on prevalence of periodontitis in adults in the United States: NHANES 2009 to 2012. J Periodontol. 2015;86(5):611–22. https://pubmed.ncbi.nlm.nih.gov/25688694/
Nascimento GG, Leite FRM, Conceição DA, Ferrúa CP, Singh A, Demarco FF. Is there a relationship between obesity and tooth loss and edentulism? A systematic review and meta-analysis. Obes Rev. 2016;17(7):587–98. https://pubmed.ncbi.nlm.nih.gov/27125768/
Kotronia E, Brown H, Papacosta AO, et al. Poor oral health and the association with diet quality and intake in older people in two studies in the UK and USA. Br J Nutr. 2021;126(1):118–30. https://pubmed.ncbi.nlm.nih.gov/33468264/
. ¿ntoniadou M, Varzakas T. Breaking the vicious circle of diet, malnutrition and oral health for the independent elderly. Crit Rev Food Sci Nutr. 2021;61(19):3233–55. https://pubmed.ncbi.nlm.nih.gov/32686465/
Gaewkhiew P, Sabbah W, Bernabé E. Does tooth loss affect dietary intake and nutritional status? A systematic review of longitudinal studies. J Dent. 2017;67:1–8. https://pubmed.ncbi.nlm.nih.gov/29097121/
Romandini M, Baima G, Antonoglou G, Bueno J, Figuero E, Sanz M. Periodontitis, edentulism, and risk of mortality: a systematic review with meta-analyses. J Dent Res. 2021;100(1):37–49. https://pubmed.ncbi.nlm.nih.gov/32866427/
Liljestrand JM, Havulinna AS, Paju S, Männistö S, Salomaa V, Pussinen PJ. Missing teeth predict incident cardiovascular events, diabetes, and death. J Dent Res. 2015;94(8):1055–62. https://pubmed.ncbi.nlm.nih.gov/25991651/
Friedman PK, Lamster IB. Tooth loss as a predictor of shortened longevity: exploring the hypothesis. Periodontol 2000. 2016;72(1):142–52. https://pubmed.ncbi.nlm.nih.gov/27501497/
Kaufman LB, Setiono TK, Doros G, et al. An oral health study of centenarians and children of centenarians. J Am Geriatr Soc. 2014;62(6):1168–73. https://pubmed.ncbi.nlm.nih.gov/24889721/
Liljestrand JM, Havulinna AS, Paju S, Männistö S, Salomaa V, Pussinen PJ. Missing teeth predict incident cardiovascular events, diabetes, and death. J Dent Res. 2015;94(8):1055–62. https://pubmed.ncbi.nlm.nih.gov/25991651/
Romandini M, Baima G, Antonoglou G, Bueno J, Figuero E, Sanz M. Periodontitis, edentulism, and risk of mortality: a systematic review with meta-analyses. J Dent Res. 2021;100(1):37–49. https://pubmed.ncbi.nlm.nih.gov/32866427/
Liljestrand JM, Havulinna AS, Paju S, Männistö S, Salomaa V, Pussinen PJ. Missing teeth predict incident cardiovascular events, diabetes, and death. J Dent Res. 2015;94(8):1055–62. https://pubmed.ncbi.nlm.nih.gov/25991651/
Kellesarian SV, Kellesarian TV, Ros Malignaggi V, et al. Association between periodontal disease and erectile dysfunction: a systematic review. Am J Mens Health. 2018;12(2):338–46. https://pubmed.ncbi.nlm.nih.gov/27030114/
Nascimento PC, Castro MML, Magno MB, et al. Association between periodontitis and cognitive impairment in adults: a systematic review. Front Neurol. 2019;10:323. https://pubmed.ncbi.nlm.nih.gov/31105630/
Fang WL, Jiang MJ, Gu BB, et al. Tooth loss as a risk factor for dementia: systematic review and meta-analysis of 21 observational studies. BMC Psychiatry. 2018;18(1):345. https://pubmed.ncbi.nlm.nih.gov/30342524/
Weijenberg RAF, Delwel S, Ho BV, van der Maarel-Wierink CD, Lobbezoo F. Mind your teeth – the relationship between mastication and cognition. Gerodontology. 2019;36(1):2–7. https://pubmed.ncbi.nlm.nih.gov/30480331/
Chen J, Ren CJ, Wu L, et al. Tooth loss is associated with increased risk of dementia and with a dose-response relationship. Front Aging Neurosci. 2018;10:415. https://pubmed.ncbi.nlm.nih.gov/30618721/
De Cicco V, Barresi M, Tramonti Fantozzi MP, Cataldo E, Parisi V, Manzoni D. Oral implant-prostheses: new teeth for a brighter brain. PLoS One. 2016;11(2):e0148715. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4771091/
Banu R F, Veeravalli PT, Kumar V A. Comparative evaluation of changes in brain activity and cognitive function of edentulous patients, with dentures and two-implant supported mandibular overdenture-pilot study. Clin Implant Dent Relat Res. 2016;18(3):580–7. https://pubmed.ncbi.nlm.nih.gov/25825258/
Awadalla HI, Ragab MH, Bassuoni MW, Fayed MT, Abbas MO. A pilot study of the role of green tea use on oral health. Int J Dent Hyg. 2011;9(2):110–6. https://pubmed.ncbi.nlm.nih.gov/21356006/
Balappanavar AY, Sardana V, Singh M. Comparison of the effectiveness of 0.5 % tea, 2 % neem and 0.2 % chlorhexidine mouthwashes on oral health: a randomized control trial. Indian J Dent Res. 2013;24(1):26–34. https://pubmed.ncbi.nlm.nih.gov/23852229/
Hasan S, Danishuddin M, Adil M, Singh K, Verma PK, Khan AU. Efficacy of E. officinalis on the cariogenic properties of Streptococcus mutans: a novel and alternative approach to suppress quorum-sensing mechanism. PLoS One. 2012;7(7):e40319. https://pubmed.ncbi.nlm.nih.gov/22792279/
Stoy PJ. Dental disease and civilization. Ulster Med J. 1951;20(2):144–58. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2479700/
Marcenes W, Kassebaum NJ, Bernabé E, et al. Global burden of oral conditions in 1990–2010: a systematic analysis. J Dent Res. 2013;92(7):592–7. https://pubmed.ncbi.nlm.nih.gov/23720570/
Sheiham A, James WPT. Diet and dental caries: the pivotal role of free sugars reemphasized. J Dent Res. 2015;94(10):1341–7. https://pubmed.ncbi.nlm.nih.gov/26261186/
Kearns CE, Apollonio D, Glantz SA. Sugar industry sponsorship of germ-free rodent studies linking sucrose to hyperlipidemia and cancer: an historical analysis of internal documents. PLoS Biol. 2017;15(11):e2003460. https://pubmed.ncbi.nlm.nih.gov/29161267/
Harris JL, Schwartz MB, Brownell KD, et al. Cereal FACTS 2012: limited progress in the nutrition quality and marketing of children’s cereals. Yale Rudd Center for Food Policy & Obesity; 2012. https://www.cerealfacts.org/
Policy statement on beverage vending machines in schools. American Academy of Pediatric Dentists. https://www.aapd.org/assets/news/upload/2002/118.pdf. Published May 2022. Accessed September 10, 2022.; https://www.aapd.org/assets/news/upload/2002/118.pdf
Jacobson MF. Lifting the veil of secrecy: corporate support for health and environmental professional associations, charities, and industry front groups. Center for Science in the Public Interest. https://www.cspinet.org/sites/default/files/attachment/lift_the_veil_intro.pdf. Published June 2003. Accessed September 10, 2022.; https://www.cspinet.org/sites/default/files/attachment/lift_the_veil_intro.pdf
AAPD leadership perspective on the AAPD Foundation’s collaboration with the Coca-Cola Foundation. American Academy of Pediatric Dentists. https://www.aapd.org/globalassets/assets/news/upload/2003/197.pdf. Accessed September 10, 2022.; https://www.aapd.org/globalassets/assets/news/upload/2003/197.pdf
Jacobson MF. Lifting the veil of secrecy: corporate support for health and environmental professional associations, charities, and industry front groups. Center for Science in the Public Interest. https://www.cspinet.org/sites/default/files/attachment/lift_the_veil_intro.pdf. Published June 2003. Accessed September 10, 2022.; https://www.cspinet.org/sites/default/files/attachment/lift_the_veil_intro.pdf
Sheiham A, James WPT. A reappraisal of the quantitative relationship between sugar intake and dental caries: the need for new criteria for developing goals for sugar intake. BMC Public Health. 2014;14(1):863. https://pubmed.ncbi.nlm.nih.gov/25228012/
Gibson SA. Breakfast cereal consumption in young children: associations with non-milk extrinsic sugars and caries experience: further analysis of data from the UK National Diet and Nutrition Survey of children aged 1.5–4.5 years. Public Health Nutr. 2000;3(2):227–32. https://pubmed.ncbi.nlm.nih.gov/10948390/
Curzon MEJ. Dietary carbohydrate and dental caries. In: Dobbing J, ed. A Balanced Diet?. Springer London, 1988: 57–75.; https://link.springer.com/chapter/10.1007/978-1-4471-1652-3_4
Cottrell RC. Letter to the editor, “Effect on caries of restricting sugars intake: systematic review to inform WHO guidelines.” J Dent Res. 2014;93(5):530. https://pubmed.ncbi.nlm.nih.gov/24595636/
Redberg RF. Cancer risks and radiation exposure from computed tomographic scans: how can we be sure that the benefits outweigh the risks? Arch Intern Med. 2009;169(22):2049–50. https://pubmed.ncbi.nlm.nih.gov/20008685/
Memon A, Rogers I, Paudyal P, Sundin J. Dental x-rays and the risk of thyroid cancer and meningioma: a systematic review and meta-analysis of current epidemiological evidence. Thyroid. 2019;29(11):1572–93. https://pubmed.ncbi.nlm.nih.gov/31502516/
Food and Drug Administration. Dental radiography: doses and film speed. https://www.fda.gov/radiation-emitting-products/nationwide-evaluation-x-ray-trendsnext/dental-radiography-doses-and-film-speed. Updated December 2, 2017. Accessed January 12, 2023.; https://www.fda.gov/radiation-emitting-products/nationwide-evaluation-x-ray-trendsnext/dental-radiography-doses-and-film-speed.
Memon A, Rogers I, Paudyal P, Sundin J. Dental x-rays and the risk of thyroid cancer and meningioma: a systematic review and meta-analysis of current epidemiological evidence. Thyroid. 2019;29(11):1572–93. https://pubmed.ncbi.nlm.nih.gov/31502516/
American Dental Association Council on Scientific Affairs. The use of dental radiographs: update and recommendations. J Am Dent Assoc. 2006;137(9):1304–12. https://pubmed.ncbi.nlm.nih.gov/16946440/
White SC, Mallya SM. Update on the biological effects of ionizing radiation, relative dose factors and radiation hygiene. Aust Dent J. 2012;57 Suppl 1:2–8. https://pubmed.ncbi.nlm.nih.gov/22376091/
Staufenbiel I, Weinspach K, Förster G, Geurtsen W, Günay H. Periodontal conditions in vegetarians: a clinical study. Eur J Clin Nutr. 2013;67(8):836–40. https://pubmed.ncbi.nlm.nih.gov/23714722/
Macri E, Lifshitz F, Ramos C, et al. Atherogenic cholesterol-rich diet and periodontal disease. Arch Oral Biol. 2014;59(7):679–86. https://pubmed.ncbi.nlm.nih.gov/24769219/
Kondo K, Ishikado A, Morino K, et al. A high-fiber, low-fat diet improves periodontal disease markers in high-risk subjects: a pilot study. Nutr Res. 2014;34(6):491–8. https://pubmed.ncbi.nlm.nih.gov/25026916/
Laine MA, Tolvanen M, Pienihäkkinen K, et al. The effect of dietary intervention on paraffin-stimulated saliva and dental health of children participating in a randomized controlled trial. Arch Oral Biol. 2014;59(2):217–25. https://pubmed.ncbi.nlm.nih.gov/24370194/
Wälti A, Lussi A, Seemann R. The effect of a chewing-intensive, high-fiber diet on oral halitosis: a clinical controlled study. Swiss Dent J. 2016;126(9):782–95. https://pubmed.ncbi.nlm.nih.gov/27655031/
Sambunjak D, Nickerson JW, Poklepovic T, et al. Flossing for the management of periodontal diseases and dental caries in adults. Cochrane Database Syst Rev. 2011;(12):CD008829. https://pubmed.ncbi.nlm.nih.gov/22161438/
Vernon LT, Da Silva APB, Seacat JD. In defense of flossing: part II – can we agree it’s premature to claim flossing is ineffective to help prevent periodontal diseases? J Evid Based Dent Pract. 2017;17(3):149–58. https://pubmed.ncbi.nlm.nih.gov/28865811/
Terézhalmy GT, Bartizek RD, Biesbrock AR. Plaque-removal efficacy of four types of dental floss. J Periodontol. 2008;79(2):245–51. https://pubmed.ncbi.nlm.nih.gov/18251638/
Mazhari F, Boskabady M, Moeintaghavi A, Habibi A. The effect of toothbrushing and flossing sequence on interdental plaque reduction and fluoride retention: a randomized controlled clinical trial. J Periodontol. 2018;89(7):824–32. https://pubmed.ncbi.nlm.nih.gov/29741239/
Baumgartner S, Imfeld T, Schicht O, Rath C, Persson RE, Persson GR. The impact of the Stone Age diet on gingival conditions in the absence of oral hygiene. J Periodontol. 2009;80(5):759–68. https://pubmed.ncbi.nlm.nih.gov/19405829/
Shi J, Maguer ML. Lycopene in tomatoes: chemical and physical properties affected by food processing. Crit Rev Food Sci Nutr. 2000;40(1):1–42. https://pubmed.ncbi.nlm.nih.gov/10674200/
Chandra RV, Prabhuji MLV, Roopa DA, Ravirajan S, Kishore HC. Efficacy of lycopene in the treatment of gingivitis: a randomised, placebo-controlled clinical trial. Oral Health Prev Dent. 2007;5(4):327–36. https://pubmed.ncbi.nlm.nih.gov/18173095/
Arora N, Avula H, Avula JK. The adjunctive use of systemic antioxidant therapy (lycopene) in nonsurgical treatment of chronic periodontitis: a short-term evaluation. Quintessence Int. 2013;44(6):395–405. https://pubmed.ncbi.nlm.nih.gov/23479592/
Belludi SA, Verma S, Banthia R, et al. Effect of lycopene in the treatment of periodontal disease: a clinical study. J Contemp Dent Pract. 2013;14(6):1054–9. https://pubmed.ncbi.nlm.nih.gov/24858750/
Benjamin N, Pattullo S, Weller R, Smith L, Ormerod A. Wound licking and nitric oxide. Lancet. 1997;349(9067):1776. https://pubmed.ncbi.nlm.nih.gov/9193412/
Jockel-Schneider Y, Goßner SK, Petersen N, et al. Stimulation of the nitrate-nitrite-NO-metabolism by repeated lettuce juice consumption decreases gingival inflammation in periodontal recall patients: a randomized, double-blinded, placebo-controlled clinical trial. J Clin Periodontol. 2016;43(7):603–8. https://pubmed.ncbi.nlm.nih.gov/26969836/
Burnett CL, Bergfeld WF, Belsito DV, et al. Final report on the safety assessment of Cocos nucifera (coconut) oil and related ingredients. Int J Toxicol. 2011;30(3 Suppl):5S-16S. https://pubmed.ncbi.nlm.nih.gov/21772024/
Sacks FM, Lichtenstein AH, Wu JHY, et al. Dietary fats and cardiovascular disease: a presidential advisory from the American Heart Association. Circulation. 2017;136(3):e1–23. https://pubmed.ncbi.nlm.nih.gov/28620111/
Freeman AM, Morris PB, Barnard N, et al. Trending cardiovascular nutrition controversies. J Am Coll Cardiol. 2017;69(9):1172–87. https://pubmed.ncbi.nlm.nih.gov/28254181/
Emissions of volatile aldehydes from heated cooking oils. Food Chem. 2010;120(1):59–65. https://www.sciencedirect.com/science/article/abs/pii/S0308814609011303?via%3Dihub
Bekeleski GM, McCombs G, Melvin W. Oil pulling: an ancient practice for a modern time. J Int Oral Health. 2012;4(3):1–10. https://api.semanticscholar.org/CorpusID:5170081?utm_source=wikipedia
Shanbhag VKL. Oil pulling for maintaining oral hygiene – a review. J Tradit Complement Med. 2017;7(1):106–9. https://pubmed.ncbi.nlm.nih.gov/28053895/
Cheema R, Sharma R, Choudhary E, Sharma S. A clinical investigation to test the efficacy of oil pulling, in reducing dentin hypersensitivity, as compared to a desensitizing tooth paste. Int J Sci Study. 2014;1(6):22–6. https://www.researchgate.net/publication/294726108_A_Clinical_Investigation_to_Test_the_Efficacy_of_Oil_Pulling_in_Reducing_Dentin_Hypersensitivity_as_Compared_to_a_Desensitizing_Tooth_Paste_Published_in_International_Journal_of_Scientific_Study_March
Hannig C, Wagenschwanz C, Pötschke S, et al. Effect of safflower oil on the protective properties of the in situ formed salivary pellicle. Caries Res. 2012;46(5):496–506. https://pubmed.ncbi.nlm.nih.gov/22813924/
Wheater M, Friedl Z. Effect of oil pulling on tooth whitening in vitro. J Adv Oral Res. 2016;7(1):20–3. https://journals.sagepub.com/doi/pdf/10.1177/2229411220160104
The practice of oil pulling. American Dental Association (ADA). https://web.archive.org/web/20140819060804/http:/www.ada.org/en/science-research/science-in-the-news/the-practice-of-oil-pulling. Published May 14, 2014. Accessed September 10, 2022.; https://web.archive.org/web/20140819060804/http:/www.ada.org/en/science-research/science-in-the-news/the-practice-of-oil-pulling
Wheater M, Friedl Z. Effect of oil pulling on tooth whitening in vitro. J Adv Oral Res. 2016;7(1):20–3. https://journals.sagepub.com/doi/pdf/10.1177/2229411220160104
Smits KPJ, Listl S, Jevdjevic M. Vegetarian diet and its possible influence on dental health: a systematic literature review. Community Dent Oral Epidemiol. 2020;48(1):7–13. https://pubmed.ncbi.nlm.nih.gov/31571246/
Ginter E. Vegetarian diets, chronic diseases and longevity. Bratisl Lek Listy. 2008;109(10):463–6. https://pubmed.ncbi.nlm.nih.gov/19166134/
Menzel J, Jabakhanji A, Biemann R, Mai K, Abraham K, Weikert C. Systematic review and meta-analysis of the associations of vegan and vegetarian diets with inflammatory biomarkers. Sci Rep. 2020;10(1):21736. https://pubmed.ncbi.nlm.nih.gov/33303765/
Hirayama T. An epidemiological study of oral and pharyngeal cancer in Central and South-East Asia. Bull World Health Organ. 1966;34(1):41–69. https://pubmed.ncbi.nlm.nih.gov/5295564/
Rao DN, Ganesh B, Rao RS, Desai PB. Risk assessment of tobacco, alcohol and diet in oral cancer – a case-control study. Int J Cancer. 1994;58(4):469–73. https://pubmed.ncbi.nlm.nih.gov/8056441/
Gangane N, Chawla S, Anshu, Subodh A, Gupta SS, Sharma SM. Reassessment of risk factors for oral cancer. Asian Pac J Cancer Prev. 2007;8(2):243–8. https://pubmed.ncbi.nlm.nih.gov/17696739/
Mishra A. Head and neck cancer in India – review of practices for prevention policy. Oral Dis. 2009;15(7):454–65. https://pubmed.ncbi.nlm.nih.gov/19413676/
Chainani-Wu N, Epstein J, Touger-Decker R. Diet and prevention of oral cancer: strategies for clinical practice. J Am Dent Assoc. 2011;142(2):166–9. https://pubmed.ncbi.nlm.nih.gov/21282682/
Smits KPJ, Listl S, Jevdjevic M. Vegetarian diet and its possible influence on dental health: a systematic literature review. Community Dent Oral Epidemiol. 2020;48(1):7–13. https://pubmed.ncbi.nlm.nih.gov/31571246/
Herman K, Czajczynska-Waszkiewicz A, Kowalczyk-Zajac M, Dobrzynski M. Assessment of the influence of vegetarian diet on the occurrence of erosive and abrasive cavities in hard tooth tissues. Postepy Hig Med Dosw. 2011;65:764–9. https://pubmed.ncbi.nlm.nih.gov/22173441/
Chu CH, Pang KKL, Lo ECM. Dietary behavior and knowledge of dental erosion among Chinese adults. BMC Oral Health. 2010;10:13. https://pubmed.ncbi.nlm.nih.gov/20525244/
Attin T, Siegel S, Buchalla W, Lennon AM, Hannig C, Becker K. Brushing abrasion of softened and remineralised dentin: an in situ study. Caries Res. 2004;38(1):62–6. https://pubmed.ncbi.nlm.nih.gov/14684979/
Staufenbiel I, Adam K, Deac A, Geurtsen W, Günay H. Influence of fruit consumption and fluoride application on the prevalence of caries and erosion in vegetarians – a controlled clinical trial. Eur J Clin Nutr. 2015;69(10):1156–60. https://pubmed.ncbi.nlm.nih.gov/25782429/
Walsh T, Worthington HV, Glenny AM, Marinho VC, Jeroncic A. Fluoride toothpastes of different concentrations for preventing dental caries. Cochrane Database Syst Rev. 2019;3:CD007868. https://pubmed.ncbi.nlm.nih.gov/30829399/
Centers for Disease Control and Prevention (CDC). Ten great public health achievements – United States, 1900–1999. MMWR Morb Mortal Wkly Rep. 1999;48(12):241–3. https://pubmed.ncbi.nlm.nih.gov/10220250/
Bellinger DC. Is fluoride potentially neurotoxic? JAMA Pediatr. 2019;173(10):915–7. https://pubmed.ncbi.nlm.nih.gov/31424483/
Valdez Jiménez L, López Guzmán OD, Cervantes Flores M, et al. In utero exposure to fluoride and cognitive development delay in infants. Neurotoxicology. 2017;59:65–70. https://pubmed.ncbi.nlm.nih.gov/28077305/
Needleman HL, Gatsonis CA. Low-level lead exposure and the IQ of children. A meta-analysis of modern studies. JAMA. 1990;263(5):673–8. https://pubmed.ncbi.nlm.nih.gov/2136923/
Unde MP, Patil RU, Dastoor PP. The untold story of fluoridation: revisiting the changing perspectives. Indian J Occup Environ Med. 2018;22(3):121–7. https://pubmed.ncbi.nlm.nih.gov/30647513/
Walchuk C, Suh M. Nutrition and the aging retina: a comprehensive review of the relationship between nutrients and their role in age-related macular degeneration and retina disease prevention. Adv Food Nutr Res. 2020;93:293–332. https://pubmed.ncbi.nlm.nih.gov/32711865/
Ruan Y, Jiang S, Gericke A. Age-related macular degeneration: role of oxidative stress and blood vessels. Int J Mol Sci. 2021;22(3):1296. https://pubmed.ncbi.nlm.nih.gov/33525498/
Glickman RD. Ultraviolet phototoxicity to the retina. Eye Contact Lens. 2011;37(4):196–205. https://pubmed.ncbi.nlm.nih.gov/21646980/
Abokyi S, To CH, Lam TT, Tse DY. Central role of oxidative stress in age-related macular degeneration: evidence from a review of the molecular mechanisms and animal models. Oxid Med Cell Longev. 2020;2020:7901270. https://pubmed.ncbi.nlm.nih.gov/32104539/
Abokyi S, To CH, Lam TT, Tse DY. Central role of oxidative stress in age-related macular degeneration: evidence from a review of the molecular mechanisms and animal models. Oxid Med Cell Longev. 2020;2020:7901270. https://pubmed.ncbi.nlm.nih.gov/32104539/
Blasiak J, Glowacki S, Kauppinen A, Kaarniranta K. Mitochondrial and nuclear DNA damage and repair in age-related macular degeneration. Int J Mol Sci. 2013;14(2):2996–3010. https://pubmed.ncbi.nlm.nih.gov/23434654/
Totan Y, Yagci R, Bardak Y, et al. Oxidative macromolecular damage in age-related macular degeneration. Curr Eye Res. 2009;34(12):1089–93. https://pubmed.ncbi.nlm.nih.gov/19958129/
Abokyi S, To CH, Lam TT, Tse DY. Central role of oxidative stress in age-related macular degeneration: evidence from a review of the molecular mechanisms and animal models. Oxid Med Cell Longev. 2020;2020:7901270. https://pubmed.ncbi.nlm.nih.gov/32104539/
Heesterbeek TJ, Lorés-Motta L, Hoyng CB, Lechanteur YTE, den Hollander AI. Risk factors for progression of age-related macular degeneration. Ophthalmic Physiol Opt. 2020;40(2):140–70. https://pubmed.ncbi.nlm.nih.gov/32100327/
Sin HPY, Liu DTL, Lam DSC. Lifestyle modification, nutritional and vitamins supplements for age-related macular degeneration. Acta Ophthalmol. 2013;91(1):6–11. https://pubmed.ncbi.nlm.nih.gov/22268800/
Jia YP, Sun L, Yu HS, et al. The pharmacological effects of lutein and zeaxanthin on visual disorders and cognition diseases. Molecules. 2017;22(4):E610. https://pubmed.ncbi.nlm.nih.gov/28425969/
Wegner A, Khoramnia R. Cataract is a self-defence reaction to protect the retina from oxidative damage. Med Hypotheses. 2011;76(5):741–4. https://pubmed.ncbi.nlm.nih.gov/21354712/
Kanter M. Lutein, zeaxanthin and eye health. Egg Nutrition Center. https://www.ee-staging.eggnutritioncenter.org/articles/lutein-zeaxanthin-and-eye-health/. Published May 16, 2022. Accessed September 15, 2022.; https://www.ee-staging.eggnutritioncenter.org/articles/lutein-zeaxanthin-and-eye-health/
Agricultural Research Service, United States Department of Agriculture. Kale, 113 g serving. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/food-details/1602525/nutrients. Published March 19, 2021. Accessed February 24, 2023.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/1602525/nutrients
Agricultural Research Service, United States Department of Agriculture. Spinach, frozen, chopped or leaf, cooked, boiled, drained, without salt. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/food-details/169288/nutrients. Published April 1, 2019. Accessed February 24, 2023.; https://fdc.nal.usda.gov/fdc-app.html#/food-details/169288/nutrients
Agricultural Research Service, United States Department of Agriculture. Component search: lutein + zeaxanthin (µg). FoodData Central https://fdc.nal.usda.gov/fdc-app.html#/?component=1123. Accessed January 25, 2023.; https://fdc.nal.usda.gov/fdc-app.html#/?component=1123
Wenzel AJ, Gerweck C, Barbato D, Nicolosi RJ, Handelman GJ, Curran-Celentano J. A 12-wk egg intervention increases serum zeaxanthin and macular pigment optical density in women. J Nutr. 2006;136(10):2568–73. https://pubmed.ncbi.nlm.nih.gov/16988128/
Hammond BR, Johnson EJ, Russell RM, et al. Dietary modification of human macular pigment density. Invest Ophthalmol Vis Sci. 1997;38(9):1795–801. https://pubmed.ncbi.nlm.nih.gov/9286268/
Abdel-Aal ESM, Akhtar H, Zaheer K, Ali R. Dietary sources of lutein and zeaxanthin carotenoids and their role in eye health. Nutrients. 2013;5(4):1169–85. https://pubmed.ncbi.nlm.nih.gov/23571649/
Eisenhauer B, Natoli S, Liew G, Flood VM. Lutein and zeaxanthin – food sources, bioavailability and dietary variety in age-related macular degeneration protection. Nutrients. 2017;9(2):E120. https://www.mdpi.com/2072-6643/9/2/120
Cheng CY, Chung WY, Szeto YT, Benzie IFF. Fasting plasma zeaxanthin response to Fructus barbarum L. (wolfberry; kei tze) in a food-based human supplementation trial. Br J Nutr. 2005;93(1):123–30. https://pubmed.ncbi.nlm.nih.gov/15705234/
Neelam K, Dey S, Sim R, Lee J, Au Eong KG. Fructus lycii: a natural dietary supplement for amelioration of retinal diseases. Nutrients. 2021;13(1):246. https://pubmed.ncbi.nlm.nih.gov/33467087/
Unlu NZ, Bohn T, Clinton SK, Schwartz SJ. Carotenoid absorption from salad and salsa by humans is enhanced by the addition of avocado or avocado oil. J Nutr. 2005;135(3):431–6. https://pubmed.ncbi.nlm.nih.gov/15735074/
Eriksen JN, Luu AY, Dragsted LO, Arrigoni E. In vitro liberation of carotenoids from spinach and Asia salads after different domestic kitchen procedures. Food Chem. 2016;203:23–7. https://pubmed.ncbi.nlm.nih.gov/26948584/
Eriksen JN, Luu AY, Dragsted LO, Arrigoni E. In vitro liberation of carotenoids from spinach and Asia salads after different domestic kitchen procedures. Food Chem. 2016;203:23–7. https://pubmed.ncbi.nlm.nih.gov/26948584/
Gorusupudi A, Nelson K, Bernstein PS. The Age-Related Eye Disease 2 Study: micronutrients in the treatment of macular degeneration. Adv Nutr. 2017;8(1):40–53. https://pubmed.ncbi.nlm.nih.gov/28096126/
Gorusupudi A, Nelson K, Bernstein PS. The Age-Related Eye Disease 2 Study: micronutrients in the treatment of macular degeneration. Adv Nutr. 2017;8(1):40–53. https://pubmed.ncbi.nlm.nih.gov/28096126/
Gorusupudi A, Nelson K, Bernstein PS. The Age-Related Eye Disease 2 Study: micronutrients in the treatment of macular degeneration. Adv Nutr. 2017;8(1):40–53. https://pubmed.ncbi.nlm.nih.gov/28096126/
Chew EY, Clemons TE, SanGiovanni JP, et al. Secondary analyses of the effects of lutein/zeaxanthin on age-related macular degeneration progression: AREDS2 report No. 3. JAMA Ophthalmol. 2014;132(2):142–9. https://pubmed.ncbi.nlm.nih.gov/24310343/
Lawrenson JG, Evans JR, Downie LE. A critical appraisal of national and international clinical practice guidelines reporting nutritional recommendations for age-related macular degeneration: are recommendations evidence-based? Nutrients. 2019;11(4):E823. https://pubmed.ncbi.nlm.nih.gov/30979051/
Evans JR, Lawrenson JG. Antioxidant vitamin and mineral supplements for preventing age-related macular degeneration. Cochrane Database Syst Rev. 2017;7:CD000253. https://pubmed.ncbi.nlm.nih.gov/28756617/
Broadhead GK, Grigg JR, Chang AA, McCluskey P. Dietary modification and supplementation for the treatment of age-related macular degeneration. Nutr Rev. 2015;73(7):448–62. https://pubmed.ncbi.nlm.nih.gov/26081455/
Rhone M, Basu A. Phytochemicals and age-related eye diseases. Nutr Rev. 2008;66(8):465–72. https://pubmed.ncbi.nlm.nih.gov/18667008/
Hammond BR, Fuld K, Snodderly DM. Iris color and macular pigment optical density. Exp Eye Res. 1996;62(3):293–7. https://pubmed.ncbi.nlm.nih.gov/8690039/
Stringham JM, Bovier ER, Wong JC, Hammond BR. The influence of dietary lutein and zeaxanthin on visual performance. J Food Sci. 2010;75(1):R24–9. https://pubmed.ncbi.nlm.nih.gov/20492192/
Hammond BR, Fletcher LM, Roos F, Wittwer J, Schalch W. A double-blind, placebo-controlled study on the effects of lutein and zeaxanthin on photostress recovery, glare disability, and chromatic contrast. Invest Ophthalmol Vis Sci. 2014;55(12):8583–9. https://pubmed.ncbi.nlm.nih.gov/25468896/
Rinninella E, Mele MC, Merendino N, et al. The role of diet, micronutrients and the gut microbiota in age-related macular degeneration: new perspectives from the gut – retina axis. Nutrients. 2018;10(11):E1677. https://pubmed.ncbi.nlm.nih.gov/30400586/
Hammond BR, Fletcher LM, Roos F, Wittwer J, Schalch W. A double-blind, placebo-controlled study on the effects of lutein and zeaxanthin on photostress recovery, glare disability, and chromatic contrast. Invest Ophthalmol Vis Sci. 2014;55(12):8583–9. https://pubmed.ncbi.nlm.nih.gov/25468896/
Sorond FA, Lipsitz LA, Hollenberg NK, Fisher NDL. Cerebral blood flow response to flavanol-rich cocoa in healthy elderly humans. Neuropsychiatr Dis Treat. 2008;4(2):433–40. https://pubmed.ncbi.nlm.nih.gov/18728792/
Rabin JC, Karunathilake N, Patrizi K. Effects of milk vs dark chocolate consumption on visual acuity and contrast sensitivity within 2 hours: a randomized clinical trial. JAMA Ophthalmol. 2018;136(6):678–81. https://pubmed.ncbi.nlm.nih.gov/29710322/
Westenskow PD. Nicotinamide: a novel treatment for age-related macular degeneration? Stem Cell Investig. 2017;4:86. https://pubmed.ncbi.nlm.nih.gov/29167807/
Dinu M, Pagliai G, Casini A, Sofi F. Food groups and risk of age-related macular degeneration: a systematic review with meta-analysis. Eur J Nutr. 2019;58(5):2123–43. https://pubmed.ncbi.nlm.nih.gov/29978377/
Mares-Perlman JA, Brady WE, Klein R, VandenLangenberg GM, Klein BE, Palta M. Dietary fat and age-related maculopathy. Arch Ophthalmol. 1995;113(6):743–8. https://pubmed.ncbi.nlm.nih.gov/7786215/
Ban N, Lee TJ, Sene A, et al. Impaired monocyte cholesterol clearance initiates age-related retinal degeneration and vision loss. JCI Insight. 2018;3(17):120824. https://pubmed.ncbi.nlm.nih.gov/30185655/
Rodriguez IR, Clark ME, Lee JW, Curcio CA. 7-ketocholesterol accumulates in ocular tissues as a consequence of aging and is present in high levels in drusen. Exp Eye Res. 2014;128:151–5. https://pubmed.ncbi.nlm.nih.gov/25261634/
Yin L, Shi Y, Liu X, et al. A rat model for studying the biological effects of circulating LDL in the choriocapillaris-BrM-RPE complex. Am J Pathol. 2012;180(2):541–9. https://pubmed.ncbi.nlm.nih.gov/22107828/
Gehlbach P, Li T, Hatef E. Statins for age-related macular degeneration. Cochrane Database Syst Rev. 2016;(8):CD006927. https://pubmed.ncbi.nlm.nih.gov/27490232/
Rhone M, Basu A. Phytochemicals and age-related eye diseases. Nutr Rev. 2008;66(8):465–72. https://pubmed.ncbi.nlm.nih.gov/18667008/
Park CY, Gu N, Lim CY, et al. The effect of Vaccinium uliginosum extract on tablet computer-induced asthenopia: randomized placebo-controlled study. BMC Complement Altern Med. 2016;16:296. https://pubmed.ncbi.nlm.nih.gov/27538497/
Kalt W, McDonald JE, Fillmore SAE, Tremblay F. Blueberry effects on dark vision and recovery after photobleaching: placebo-controlled crossover studies. J Agric Food Chem. 2014;62(46):11180–9. https://pubmed.ncbi.nlm.nih.gov/25335781/
Broadhead GK, Grigg JR, McCluskey P, Hong T, Schlub TE, Chang AA. Saffron therapy for the treatment of mild/moderate age-related macular degeneration: a randomised clinical trial. Graefes Arch Clin Exp Ophthalmol. 2019;257(1):31–40. https://pubmed.ncbi.nlm.nih.gov/30343354/
Tribble JR, Hui F, Jöe M, et al. Targeting diet and exercise for neuroprotection and neurorecovery in glaucoma. Cells. 2021;10(2):295. https://pubmed.ncbi.nlm.nih.gov/33535578/
Han B, Song M, Li L, Sun X, Lei Y. The application of nitric oxide for ocular hypertension treatment. Molecules. 2021;26(23):7306. https://pubmed.ncbi.nlm.nih.gov/34885889/
Han B, Song M, Li L, Sun X, Lei Y. The application of nitric oxide for ocular hypertension treatment. Molecules. 2021;26(23):7306. https://pubmed.ncbi.nlm.nih.gov/34885889/
Bastia E, Toris C, Bukowski JM, et al. NCX 1741, a novel nitric oxide-donating phosphodiesterase-5 inhibitor, exerts rapid and long-lasting intraocular pressure-lowering in cynomolgus monkeys. J Ocul Pharmacol Ther. 2021;37(4):215–22. https://pubmed.ncbi.nlm.nih.gov/33595367/
Coleman AL, Stone KL, Kodjebacheva G, et al. Glaucoma risk and the consumption of fruits and vegetables among older women in the study of osteoporotic fractures. Am J Ophthalmol. 2008;145(6):1081–9. https://pubmed.ncbi.nlm.nih.gov/18355790/
Giaconi JA, Yu F, Stone KL, et al. The association of consumption of fruits/vegetables with decreased risk of glaucoma among older African-American women in the study of osteoporotic fractures. Am J Ophthalmol. 2012;154(4):635–44. https://pubmed.ncbi.nlm.nih.gov/22818906/
Bao Y, Bertoia ML, Lenart EB, et al. Origin, methods, and evolution of the three Nurses’ Health Studies. Am J Public Health. 2016;106(9):1573–81. https://pubmed.ncbi.nlm.nih.gov/27459450/
Health Professionals Follow-up Study. Harvard T.H. Chan School of Public Health. https://sites.sph.harvard.edu/hpfs/questions-and-answers/. Accessed September 15, 2022.; https://www.hsph.harvard.edu/hpfs/questions-and-answers/
Kang JH, Willett WC, Rosner BA, Buys E, Wiggs JL, Pasquale LR. Association of dietary nitrate intake with primary open-angle glaucoma: a prospective analysis from the Nurses’ Health Study and Health Professionals Follow-up Study. JAMA Ophthalmol. 2016;134(3):294–303. https://pubmed.ncbi.nlm.nih.gov/26767881/
Zhu MM, Lai JSM, Choy BNK, et al. Physical exercise and glaucoma: a review on the roles of physical exercise on intraocular pressure control, ocular blood flow regulation, neuroprotection and glaucoma-related mental health. Acta Ophthalmol. 2018;96(6):e676–91. https://pubmed.ncbi.nlm.nih.gov/29338126/
Passo MS, Elliot DL, Goldberg L. Long-term effects of exercise conditioning on intraocular pressure in glaucoma suspects. J Glaucoma. 1992;1(1):39–41. https://pubmed.ncbi.nlm.nih.gov/19996871/
Chorich LJ III, Davidorf FH, Chambers RB, Weber PA. Bungee cord-associated ocular injuries. Am J Ophthalmol. 1998;125(2):270–2. https://pubmed.ncbi.nlm.nih.gov/9467466/
Kalthoff H, John S. [Intraocular pressure in snorkling and diving (author’s transl)]. Klin Monbl Augenheilkd. 1976;168(02):253–7. https://pubmed.ncbi.nlm.nih.gov/957553/
Jasien JV, Jonas JB, de Moraes CG, Ritch R. Intraocular pressure rise in subjects with and without glaucoma during four common yoga positions. PLoS One. 2015;10(12):e0144505. https://pubmed.ncbi.nlm.nih.gov/26698309/
Williams PT. Relationship of incident glaucoma versus physical activity and fitness in male runners. Med Sci Sports Exerc. 2009;41(8):1566–72. https://pubmed.ncbi.nlm.nih.gov/19568204/
Ramulu PY, Maul E, Hochberg C, Chan ES, Ferrucci L, Friedman DS. Real-world assessment of physical activity in glaucoma using an accelerometer. Ophthalmology. 2012;119(6):1159–66. https://pubmed.ncbi.nlm.nih.gov/22386950/
Ohguro H, Ohguro I, Katai M, Tanaka S. Two-year randomized, placebo-controlled study of black currant anthocyanins on visual field in glaucoma. Ophthalmologica. 2012;228(1):26–35. https://pubmed.ncbi.nlm.nih.gov/22377796/
McGlynnP. Welcome back black currants: forbidden fruit making a comeback in New York. Cornell Chronicle. https://news.cornell.edu/stories/2006/07/welcome-back-black-currants-forbidden-fruit-making-ny-comeback. Published July 26, 2006. Accessed February 25, 2006.; https://news.cornell.edu/stories/2006/07/welcome-back-black-currants-forbidden-fruit-making-ny-comeback
Sari MD. Ginkgo biloba extract effect on oxidative stress marker malonildialdehyde, redox enzyme gluthation peroxidase, visual field damage, and retinal nerve fiber layer thickness in primary open angle glaucoma. InnoPharm2 Second International Conference on Innovations in Pharmaceutical Medical and Bio Science. Innovare Academic Sciences. http://repository.usu.ac.id/handle/123456789/65446. Published February 11–12, 2017. Accessed September 15, 2022.; https://repository.usu.ac.id/handle/123456789/65446
Quaranta L, Bettelli S, Uva MG, Semeraro F, Turano R, Gandolfo E. Effect of Ginkgo biloba extract on preexisting visual field damage in normal tension glaucoma. Ophthalmology. 2003;110(2):359–62. https://pubmed.ncbi.nlm.nih.gov/12578781/
Guo X, Kong X, Huang R, et al. Effect of Ginkgo biloba on visual field and contrast sensitivity in Chinese patients with normal tension glaucoma: a randomized, crossover clinical trial. Invest Ophthalmol Vis Sci. 2014;55(1):110–6. https://pubmed.ncbi.nlm.nih.gov/24282229/
Bent S, Goldberg H, Padula A, Avins AL. Spontaneous bleeding associated with Ginkgo biloba: a case report and systematic review of the literature. J Gen Intern Med. 2005;20(7):657–61. https://pubmed.ncbi.nlm.nih.gov/16050865/
Hui F, Tang J, Williams PA, et al. Improvement in inner retinal function in glaucoma with nicotinamide (vitamin B3) supplementation: a crossover randomized clinical trial. Clin Exp Ophthalmol. 2020;48(7):903–14. https://pubmed.ncbi.nlm.nih.gov/32721104/
De Moraes CG, John SWM, Williams PA, Blumberg DM, Cioffi GA, Liebmann JM. Nicotinamide and pyruvate for neuroenhancement in open-angle glaucoma: a phase 2 randomized clinical trial. JAMA Ophthalmol. 2022;140(1):11–8. https://pubmed.ncbi.nlm.nih.gov/34792559/
Liu YC, Wilkins M, Kim T, Malyugin B, Mehta JS. Cataracts. Lancet. 2017;390(10094):600–12. https://pubmed.ncbi.nlm.nih.gov/28242111/
Liu YC, Wilkins M, Kim T, Malyugin B, Mehta JS. Cataracts. Lancet. 2017;390(10094):600–12. https://pubmed.ncbi.nlm.nih.gov/28242111/
Stephenson M. Dysphotopsia: not just black and white. Review of Ophthalmology. https://www.reviewofophthalmology.com/article/dysphotopsia-not-just-black-and-white. Published November 7, 2022. Accessed September 15, 2022.; https://www.reviewofophthalmology.com/article/dysphotopsia-not-just-black-and-white
Liu YC, Wilkins M, Kim T, Malyugin B, Mehta JS. Cataracts. Lancet. 2017;390(10094):600–12. https://pubmed.ncbi.nlm.nih.gov/28242111/
Liu YC, Wilkins M, Kim T, Malyugin B, Mehta JS. Cataracts. Lancet. 2017;390(10094):600–12. https://pubmed.ncbi.nlm.nih.gov/28242111/
Thorn DC, Grosas AB, Mabbitt PD, Ray NJ, Jackson CJ, Carver JA. The structure and stability of the disulfide-linked ¿s-crystallin dimer provide insight into oxidation products associated with lens cataract formation. J Mol Biol. 2019;431(3):483–97. https://pubmed.ncbi.nlm.nih.gov/30552875/
Weikel KA, Garber C, Baburins A, Taylor A. Nutritional modulation of cataract. Nutr Rev. 2014;72(1):30–47. https://pubmed.ncbi.nlm.nih.gov/24279748/
Westenskow PD. Nicotinamide: a novel treatment for age-related macular degeneration? Stem Cell Investig. 2017;4:86. https://pubmed.ncbi.nlm.nih.gov/29167807/
Weikel KA, Garber C, Baburins A, Taylor A. Nutritional modulation of cataract. Nutr Rev. 2014;72(1):30–47. https://pubmed.ncbi.nlm.nih.gov/24279748/
Hah YS, Chung HJ, Sontakke SB, et al. Ascorbic acid concentrations in aqueous humor after systemic vitamin C supplementation in patients with cataract: pilot study. BMC Ophthalmol. 2017;17(1):121. https://pubmed.ncbi.nlm.nih.gov/28693452/
Mares J. Food antioxidants to prevent cataract. JAMA. 2015;313(10):1048–9. https://pubmed.ncbi.nlm.nih.gov/25756441/
Sideri O, Tsaousis KT, Li HJ, Viskadouraki M, Tsinopoulos IT. The potential role of nutrition on lens pathology: a systematic review and meta-analysis. Surv Ophthalmol. 2019;64(5):668–78. https://pubmed.ncbi.nlm.nih.gov/30878580/
Wei L, Liang G, Cai C, Lv J. Association of vitamin C with the risk of age-related cataract: a meta-analysis. Acta Ophthalmol. 2016;94(3):e170–6. https://pubmed.ncbi.nlm.nih.gov/25735187/
Weikel KA, Garber C, Baburins A, Taylor A. Nutritional modulation of cataract. Nutr Rev. 2014;72(1):30–47. https://pubmed.ncbi.nlm.nih.gov/24279748/
Ma L, Hao Zx, Liu Rr, Yu RB, Shi Q, Pan JP. A dose-response meta-analysis of dietary lutein and zeaxanthin intake in relation to risk of age-related cataract. Graefes Arch Clin Exp Ophthalmol. 2014;252(1):63–70. https://pubmed.ncbi.nlm.nih.gov/24150707/
Christen WG, Glynn RJ, Sesso HD, et al. Age-related cataract in a randomized trial of vitamins E and C in men. Arch Ophthalmol. 2010;128(11):1397–405. https://pubmed.ncbi.nlm.nih.gov/21060040/
Kassoff A, Kassoff J, Buehler JA, et al. A randomized, placebo-controlled, clinical trial of high-dose supplementation with vitamins C and E and beta carotene for age-related cataract and vision loss: AREDS report no. 9. Arch Ophthalmol. 2001;119(10):1439–52. https://pubmed.ncbi.nlm.nih.gov/11594943/
Gritz DC, Srinivasan M, Smith SD, et al. The antioxidants in prevention of cataracts study: effects of antioxidant supplements on cataract progression in South India. Br J Ophthalmol. 2006;90(7):847–51. https://pubmed.ncbi.nlm.nih.gov/16556618/
Jiang H, Yin Y, Wu CR, et al. Dietary vitamin and carotenoid intake and risk of age-related cataract. Am J Clin Nutr. 2019;109(1):43–54. https://pubmed.ncbi.nlm.nih.gov/30624584/
Barker FM. Dietary supplementation: effects on visual performance and occurrence of AMD and cataracts. Curr Med Res Opin. 2010;26(8):2011–23. https://pubmed.ncbi.nlm.nih.gov/20590393/
Christen W, Glynn R, Sperduto R, Chew E, Buring J. Age-related cataract in a randomized trial of beta-carotene in women. Ophthalmic Epidemiol. 2004;11(5):401–12. https://pubmed.ncbi.nlm.nih.gov/15590586/
Chew EY, SanGiovanni JP, Ferris FL et al. Lutein/zeaxanthin for the treatment of age-related cataract: AREDS2 randomized trial report no. 4. JAMA Ophthalmol. 2013;131(7):843–50. https://pubmed.ncbi.nlm.nih.gov/23645227/
Maraini G, Williams SL, Camparini M, et al. A randomized, double-masked, placebo-controlled clinical trial of multivitamin supplementation for age-related lens opacities: Clinical Trial of Nutritional Supplements and Age-Related Cataract report no. 3. Ophthalmology. 2008;115(4):599–607.e1. https://pubmed.ncbi.nlm.nih.gov/18387406/
Mares J. Food antioxidants to prevent cataract. JAMA. 2015;313(10):1048–9. https://pubmed.ncbi.nlm.nih.gov/25756441/
Shivappa N, Hébert JR, Rashidkhani B, Ghanavati M. Inflammatory potential of diet is associated with increased odds of cataract in a case-control study from Iran. Int J Vitam Nutr Res. 2017;87(1–2):17–24. https://pubmed.ncbi.nlm.nih.gov/29327971/
Semba RD, Nicklett EJ, Ferrucci L. Does accumulation of advanced glycation end products contribute to the aging phenotype? J Gerontol A Biol Sci Med Sci. 2010;65(9):963–75. https://pubmed.ncbi.nlm.nih.gov/20478906/
Wu C, Han X, Yan X, et al. Impact of diet on the incidence of cataract surgery among diabetic patients: findings from the 45 and Up Study. Curr Eye Res. 2019;44(4):385–92. https://pubmed.ncbi.nlm.nih.gov/30433817/
Tan AG, Flood VM, Kifley A, et al. Wholegrain and legume consumption and the 5-year incidence of age-related cataract in the Blue Mountains Eye Study. Br J Nutr. 2020;124(3):306–15. https://pubmed.ncbi.nlm.nih.gov/32189601/
Люди, не употребляющие в пищу мяса теплокровных, но допускающие употребление рыбы, крабов и моллюсков. – Примеч. ред.
Appleby PN, Allen NE, Key TJ. Diet, vegetarianism, and cataract risk. Am J Clin Nutr. 2011;93(5):1128–35. https://pubmed.ncbi.nlm.nih.gov/21430115/
Appleby PN, Allen NE, Key TJ. Diet, vegetarianism, and cataract risk. Am J Clin Nutr. 2011;93(5):1128–35. https://pubmed.ncbi.nlm.nih.gov/21430115/
Fraser GE. Vegetarian diets: what do we know of their effects on common chronic diseases? Am J Clin Nutr. 2009;89(5):1607S-12S. https://pubmed.ncbi.nlm.nih.gov/19321569/
Chiu THT, Chang CC, Lin CL, Lin MN. A vegetarian diet is associated with a lower risk of cataract, particularly among individuals with overweight: a prospective study. J Acad Nutr Diet. 2021;121(4):669–77.e1. https://pubmed.ncbi.nlm.nih.gov/33309591/
Mitchell HS, Dodge WM. Cataract in rats fed on high lactose rations. J Nutr. 1935;9(1):37–49. https://pubmed.ncbi.nlm.nih.gov/394038/
Richter CP, Duke JR. Cataracts produced in rats by yogurt. Science. 1970;168(3937):1372–4. https://pubmed.ncbi.nlm.nih.gov/5444270/
Kinoshita JH. Cataracts in galactosemia. The Tonas S. Friedenwald Memorial Lecture. Invest Ophthalmol. 1965;4(5):786–99. https://pubmed.ncbi.nlm.nih.gov/5831988/
Couet C, Jan P, Debry G. Lactose and cataract in humans: a review. J Am Coll Nutr. 1991;10(1):79–86. https://pubmed.ncbi.nlm.nih.gov/1901325/
Wilson WA, Donnell GN. Cataracts in galactosemia. AMA Arch Ophthalmol. 1958;60(2):215–22. https://pubmed.ncbi.nlm.nih.gov/13558788/
Mustafa OM, Daoud YJ. Is dietary milk intake associated with cataract extraction history in older adults? An analysis from the US population. J Ophthalmol. 2020;2020:2562875. https://pubmed.ncbi.nlm.nih.gov/32148937/
Mustafa OM, Daoud YJ. Is dietary milk intake associated with cataract extraction history in older adults? An analysis from the US population. J Ophthalmol. 2020;2020:2562875. https://pubmed.ncbi.nlm.nih.gov/32148937/
Jacques PF, Phillips J, Hartz SC, Chylack LT. Lactose intake, galactose metabolism and senile cataract. Nutr Res. 1990;10(3):255–65. https://www.sciencedirect.com/science/article/abs/pii/S0271531705802672
Couet C, Jan P, Debry G. Lactose and cataract in humans: a review. J Am Coll Nutr. 1991;10(1):79–86. https://pubmed.ncbi.nlm.nih.gov/1901325/
Smith R. A good death. An important aim for health services and for us all. BMJ. 2000;320(7228):129–30. https://pubmed.ncbi.nlm.nih.gov/10634711/
Broad JB, Gott M, Kim H, Boyd M, Chen H, Connolly MJ. Where do people die? An international comparison of the percentage of deaths occurring in hospital and residential aged care settings in 45 populations, using published and available statistics. Int J Public Health. 2013;58(2):257–67. https://pubmed.ncbi.nlm.nih.gov/22892713/
Hetzler PT III, Dugdale LS. How do medicalization and rescue fantasy prevent healthy dying? AMA J Ethics. 2018;20(8):E766–73. https://pubmed.ncbi.nlm.nih.gov/30118427/
Quickstats: percentage distribution of deaths, by place of death – United States, 2000–2014. MMWR Morb Mortal Wkly Rep. 2016;65(13):357. https://www.cdc.gov/mmwr/volumes/65/wr/mm6513a6.htm
Wright AA, Keating NL, Balboni TA, Matulonis UA, Block SD, Prigerson HG. Place of death: correlations with quality of life of patients with cancer and predictors of bereaved caregivers’ mental health. J Clin Oncol. 2010;28(29):4457–64. https://pubmed.ncbi.nlm.nih.gov/20837950/
Mills M, Davies HT, Macrae WA. Care of dying patients in hospital. BMJ. 1994;309(6954):583–6. https://pubmed.ncbi.nlm.nih.gov/8086948/
Smith R. A good death. An important aim for health services and for us all. BMJ. 2000;320(7228):129–30. https://pubmed.ncbi.nlm.nih.gov/10634711/
Quill TE, Ganzini L, Truog RD, Pope TM. Voluntarily stopping eating and drinking among patients with serious advanced illness – clinical, ethical, and legal aspects. JAMA Intern Med. 2018;178(1):123–7. https://pubmed.ncbi.nlm.nih.gov/29114745/
2020 Edition: Hospice Facts and Figures. National Hospice and Palliative Care Organization. www.nhpco.org/factsfigures. Published August 20, 2020. Accessed September 20, 2022.; https://www.nhpco.org/hospice-facts-figures/
Connor SR, Pyenson B, Fitch K, Spence C, Iwasaki K. Comparing hospice and nonhospice patient survival among patients who die within a three-year window. J Pain Symptom Manage. 2007;33(3):238–46. https://pubmed.ncbi.nlm.nih.gov/17349493/
Temel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients with metastatic non-small-cell lung cancer. N Engl J Med. 2010;363(8):733–42. https://pubmed.ncbi.nlm.nih.gov/20818875/
Kelley AS, Meier DE. Palliative care – a shifting paradigm. N Engl J Med. 2010;363(8):781–2. https://pubmed.ncbi.nlm.nih.gov/20818881/
Irwin KE, Greer JA, Khatib J, Temel JS, Pirl WF. Early palliative care and metastatic non-small cell lung cancer: potential mechanisms of prolonged survival. Chron Respir Dis. 2013;10(1):35–47. https://pubmed.ncbi.nlm.nih.gov/23355404/
Winyard G, Macdonald L. The limits of palliative care. BMJ. 2014;349:g4285. https://pubmed.ncbi.nlm.nih.gov/24986853/
Ivanovic N, Büche D, Fringer A. Voluntary stopping of eating and drinking at the end of life – a ‘systematic search and review’ giving insight into an option of hastening death in capacitated adults at the end of life. BMC Palliat Care. 2014;13(1):1. https://pubmed.ncbi.nlm.nih.gov/24400678/
Symington BE. Ethics and the legalization of physician-assisted suicide. Ann Intern Med. 2018;168(11):833–4. https://pubmed.ncbi.nlm.nih.gov/29868808/
Blanke C, Ellis L, Meyskens F. Oregon’s death with dignity act – reply. JAMA Oncol. 2018;4(5):748. https://pubmed.ncbi.nlm.nih.gov/29450455/
Pope TM. Voluntarily stopping eating and drinking. Narrat Inq Bioeth. 2016;6(2):75–7. https://pubmed.ncbi.nlm.nih.gov/27763385/
Corbett M. VSED: death with dignity or without? Narrat Inq Bioeth. 2016;6(2):109–13. https://pubmed.ncbi.nlm.nih.gov/27763399/
Schwarz JK. Hospice care for patients who choose to hasten death by voluntarily stopping eating and drinking. J Hosp Palliat Nurs. 2014;16(3):126–31. https://journals.lww.com/jhpn/Fulltext/2014/05000/Hospice_Care_for_Patients_Who_Choose_to_Hasten.3.aspx
Stängle S, Schnepp W, Büche D, Häuptle C, Fringer A. Family physicians’ perspective on voluntary stopping of eating and drinking: a cross-sectional study. J Int Med Res. 2020;48(8):300060520936069. https://pubmed.ncbi.nlm.nih.gov/32787706/
Ivanovic N, Büche D, Fringer A. Voluntary stopping of eating and drinking at the end of life – a ‘systematic search and review’ giving insight into an option of hastening death in capacitated adults at the end of life. BMC Palliat Care. 2014;13(1):1. https://pubmed.ncbi.nlm.nih.gov/24400678/
Schwarz JK. Hospice care for patients who choose to hasten death by voluntarily stopping eating and drinking. J Hosp Palliat Nurs. 2014;16(3):126–31. https://journals.lww.com/jhpn/Fulltext/2014/05000/Hospice_Care_for_Patients_Who_Choose_to_Hasten.3.aspx
Lachman VD. Voluntary stopping of eating and drinking: an ethical alternative to physician-assisted suicide. Medsurg Nurs. 2015;24(1):56–9. https://pubmed.ncbi.nlm.nih.gov/26306358/
Ferrand E, Dreyfus JF, Chastrusse M, Ellien F, Lemaire F, Fischler M. Evolution of requests to hasten death among patients managed by palliative care teams in France: a multicentre cross-sectional survey (DemandE). Eur J Cancer. 2012;48(3):368–76. https://pubmed.ncbi.nlm.nih.gov/22036873/
Pope TM, Anderson LE. Voluntarily stopping eating and drinking: a legal treatment option at the end of life. Weidner Law Review. 2011;17:363–427. https://open.mitchellhamline.edu/facsch/278/
Bolt EE, Hagens M, Willems D, Onwuteaka-Philipsen BD. Primary care patients hastening death by voluntarily stopping eating and drinking. Ann Fam Med. 2015;13(5):421–8. https://pubmed.ncbi.nlm.nih.gov/26371262/
Gruenewald DA, Vandekieft G. Options of last resort: palliative sedation, physician aid in dying, and voluntary cessation of eating and drinking. Med Clin North Am. 2020;104(3):539–60. https://pubmed.ncbi.nlm.nih.gov/32312414/
Printz LA. Terminal dehydration, a compassionate treatment. Arch Intern Med. 1992;152(4):697–700. https://pubmed.ncbi.nlm.nih.gov/1373053/
Volicer L, Stets K. Acceptability of an advance directive that limits food and liquids in advanced dementia. Am J Hosp Palliat Care. 2016;33(1):55–63. https://pubmed.ncbi.nlm.nih.gov/25313239/
Ivanovic N, Büche D, Fringer A. Voluntary stopping of eating and drinking at the end of life – a ‘systematic search and review’ giving insight into an option of hastening death in capacitated adults at the end of life. BMC Palliat Care. 2014;13(1):1. https://pubmed.ncbi.nlm.nih.gov/24400678/
Eddy DM. A piece of my mind. A conversation with my mother. JAMA. 1994;272(3):179–81. https://pubmed.ncbi.nlm.nih.gov/8022025/
Jackonen S. Dehydration and hydration in the terminally ill: care considerations. Nurs Forum. 1997;32(3):5–13. https://pubmed.ncbi.nlm.nih.gov/9362876/
Rakatansky H. Complexities to consider when patients choose VSED (voluntarily stopping eating and drinking). R I Med (2013). 2017;100(2):12–3. https://pubmed.ncbi.nlm.nih.gov/28146593/
Menzel PT. Justifying a surrogate’s request to forego oral feeding. Am J Bioeth. 2019;19(1):92–4. https://pubmed.ncbi.nlm.nih.gov/30676907/
Perls TT. Anti-aging quackery: human growth hormone and tricks of the trade – more dangerous than ever. J Gerontol A Biol Sci Med Sci. 2004;59(7):682–91. https://pubmed.ncbi.nlm.nih.gov/15304532/
Pillitteri JL, Shiffman S, Rohay JM, Harkins AM, Burton SL, Wadden TA. Use of dietary supplements for weight loss in the United States: results of a national survey. Obesity (Silver Spring). 2008;16(4):790–6. https://pubmed.ncbi.nlm.nih.gov/18239570/
Sissung TM, Cordes LM, Figg WD. The Dietary Supplement Health and Education Act: are we healthier and better informed after 27 years? Lancet Oncol. 2021;22(7):915–6. https://pubmed.ncbi.nlm.nih.gov/34197743/
MacFarlane D, Hurlstone MJ, Ecker UKH. Protecting consumers from fraudulent health claims: a taxonomy of psychological drivers, interventions, barriers, and treatments. Soc Sci Med. 2020;259:112790. https://pubmed.ncbi.nlm.nih.gov/32067757/
Sissung TM, Cordes LM, Figg WD. The Dietary Supplement Health and Education Act: are we healthier and better informed after 27 years? Lancet Oncol. 2021;22(7):915–6. https://pubmed.ncbi.nlm.nih.gov/34197743/
Newmaster SG, Grguric M, Shanmughanandhan D, Ramalingam S, Ragupathy S. DNA barcoding detects contamination and substitution in North American herbal products. BMC Med. 2013;11:222. https://pubmed.ncbi.nlm.nih.gov/24120035/
Long J. FDA GMP inspectors cite 70 % of dietary supplement firms. Natural Products INSIDER. https://www.naturalproductsinsider.com/fda-gmp-inspectors-cite-70-dietary-supplement-firms. Published May 20, 2013. Accessed September 29, 2022.; https://www.naturalproductsinsider.com/regulatory/fda-gmp-inspectors-cite-70-dietary-supplement-firms
O’Connor A. New York attorney general targets supplements at major retailers. The New York Times. https://well.blogs.nytimes.com/2015/02/03/new-york-attorney-general-targets-supplements-at-major-retailers. Published February 3, 2015. Accessed September 29, 2022.; https://well.blogs.nytimes.com/2015/02/03/new-york-attorney-general-targets-supplements-at-major-retailers
Cohen PA, Maller G, Desouza R, Neal-Kababick J. Presence of banned drugs in dietary supplements following FDA recalls. JAMA. 2014;312(16):1691–3. https://pubmed.ncbi.nlm.nih.gov/25335153/
Marcus DM. Dietary supplements: what’s in a name? What’s in the bottle? Drug Test Anal. 2016;8(3–4):410–2. https://pubmed.ncbi.nlm.nih.gov/27072845/
Gahche JJ, Bailey RL, Potischman N, Dwyer JT. Dietary supplement use was very high among older adults in the United States in 2011–2014. J Nutr. 2017;147(10):1968–76. https://pubmed.ncbi.nlm.nih.gov/28855421/
Institute of Medicine. Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline. National Academy Press;1998. https://pubmed.ncbi.nlm.nih.gov/23193625/
O’Connor EA, Evans CV, Ivlev I, et al. Vitamin and mineral supplements for the primary prevention of cardiovascular disease and cancer: updated evidence report and systematic review for the US Preventive Services Task Force. JAMA. 2022;327(23):2334–47. https://pubmed.ncbi.nlm.nih.gov/35727272/
Guallar E, Stranges S, Mulrow C, Appel LJ, Miller ER. Enough is enough: stop wasting money on vitamin and mineral supplements. Ann Intern Med. 2013;159(12):850–1. https://pubmed.ncbi.nlm.nih.gov/24490268/
Jenkins DJA, Spence JD, Giovannucci EL, et al. Supplemental vitamins and minerals for cardiovascular disease prevention and treatment. J Am Coll Cardiol. 2021;77(4):423–36. https://pubmed.ncbi.nlm.nih.gov/33509399/
Biesalski HK, Tinz J. Multivitamin/mineral supplements: rationale and safety – a systematic review. Nutrition. 2017;33:76–82. https://pubmed.ncbi.nlm.nih.gov/27553772/
Mursu J, Robien K, Harnack LJ, Park K, Jacobs DR. Dietary supplements and mortality rate in older women: the Iowa Women’s Health Study. Arch Intern Med. 2011;171(18):1625–33. https://pubmed.ncbi.nlm.nih.gov/21987192/
Schwingshackl L, Boeing H, Stelmach-Mardas M, et al. Dietary supplements and risk of cause-specific death, cardiovascular disease, and cancer: a systematic review and meta-analysis of primary prevention trials. Adv Nutr. 2017;8(1):27–39. https://pubmed.ncbi.nlm.nih.gov/28096125/
Biesalski HK, Tinz J. Multivitamin/mineral supplements: rationale and safety – a systematic review. Nutrition. 2017;33:76–82. https://pubmed.ncbi.nlm.nih.gov/27553772/
Chang YY, Chiou WB. The liberating effect of weight loss supplements on dietary control: a field experiment. Nutrition. 2014;30(9):1007–10. https://pubmed.ncbi.nlm.nih.gov/24976417/
Chiou WB, Wan CS, Wu WH, Lee KT. A randomized experiment to examine unintended consequences of dietary supplement use among daily smokers: taking supplements reduces self-regulation of smoking. Addiction. 2011;106(12):2221–8. https://pubmed.ncbi.nlm.nih.gov/21806694/
Chang YY, Chiou WB. Taking weight-loss supplements may elicit liberation from dietary control. A laboratory experiment. Appetite. 2014;72:8–12. https://pubmed.ncbi.nlm.nih.gov/24096084/
Spindler SR, Mote PL, Flegal JM. Lifespan effects of simple and complex nutraceutical combinations fed isocalorically to mice. Age (Dordr). 2014;36(2):705–18. https://pubmed.ncbi.nlm.nih.gov/24370781/
Bjelakovic G, Gluud LL, Nikolova D, et al. Vitamin D supplementation for prevention of mortality in adults. Cochrane Database Syst Rev. 2014;(1):CD007470. https://pubmed.ncbi.nlm.nih.gov/24414552/
Pludowski P, Holick MF, Pilz S, et al. Vitamin D effects on musculoskeletal health, immunity, autoimmunity, cardiovascular disease, cancer, fertility, pregnancy, dementia and mortality – a review of recent evidence. Autoimmun Rev. 2013;12(10):976–89. https://pubmed.ncbi.nlm.nih.gov/23542507/
Autier P, Mullie P, Macacu A, et al. Effect of vitamin D supplementation on non-skeletal disorders: a systematic review of meta-analyses and randomised trials. Lancet Diabetes Endocrinol. 2017;5(12):986–1004. https://pubmed.ncbi.nlm.nih.gov/29102433/
Barbarawi M, Kheiri B, Zayed Y, et al. Vitamin D supplementation and cardiovascular disease risks in more than 83¿000 individuals in 21 randomized clinical trials: a meta-analysis. JAMA Cardiol. 2019;4(8):765–76. https://pubmed.ncbi.nlm.nih.gov/31215980/
Seida JC, Mitri J, Colmers IN, et al. Clinical review: effect of vitamin D3 supplementation on improving glucose homeostasis and preventing diabetes: a systematic review and meta-analysis [published correction appears in J Clin Endocrinol Metab. 2015;100(8):3219]. J Clin Endocrinol Metab. 2014;99(10):3551–60. https://pubmed.ncbi.nlm.nih.gov/25062463/
Jagannath VA, Filippini G, Di Pietrantonj C, et al. Vitamin D for the management of multiple sclerosis. Cochrane Database Syst Rev. 2018;9:CD008422. https://pubmed.ncbi.nlm.nih.gov/30246874/
Duan L, Han L, Liu Q, Zhao Y, Wang L, Wang Y. Effects of vitamin D supplementation on general and central obesity: results from 20 randomized controlled trials involving apparently healthy populations. Ann Nutr Metab. 2020;76(3):153–64. https://pubmed.ncbi.nlm.nih.gov/32645694/
Shahvazi S, Soltani S, Ahmadi SM, de Souza RJ, Salehi-Abargouei A. The effect of vitamin D supplementation on prostate cancer: a systematic review and meta-analysis of clinical trials. Horm Metab Res. 2019;51(1):11–21. https://pubmed.ncbi.nlm.nih.gov/30522147/
Beveridge LA, Struthers AD, Khan F, et al. Effect of vitamin D supplementation on blood pressure: a systematic review and meta-analysis incorporating individual patient data. JAMA Intern Med. 2015;175(5):745–54. https://pubmed.ncbi.nlm.nih.gov/25775274/
Giustina A, Adler RA, Binkley N, et al. Consensus statement from 2nd International Conference on Controversies in Vitamin D. Rev Endocr Metab Disord. 2020;21(1):89–116. https://pubmed.ncbi.nlm.nih.gov/32180081/
Jolliffe DA, Greenberg L, Hooper RL, et al. Vitamin D supplementation to prevent asthma exacerbations: a systematic review and meta-analysis of individual participant data. Lancet Respir Med. 2017;5(11):881–90. https://pubmed.ncbi.nlm.nih.gov/28986128/
Jolliffe DA, Greenberg L, Hooper RL, et al. Vitamin D to prevent exacerbations of COPD: systematic review and meta-analysis of individual participant data from randomised controlled trials. Thorax. 2019;74(4):337–45. https://pubmed.ncbi.nlm.nih.gov/30630893/
Okereke OI, Reynolds CF, Mischoulon D, et al. Effect of long-term vitamin D3 supplementation vs placebo on risk of depression or clinically relevant depressive symptoms and on change in mood scores: a randomized clinical trial. JAMA. 2020;324(5):471–80. https://pubmed.ncbi.nlm.nih.gov/32749491/
Cheng YC, Huang YC, Huang WL. The effect of vitamin D supplement on negative emotions: a systematic review and meta-analysis. Depress Anxiety. 2020;37(6):549–64. https://pubmed.ncbi.nlm.nih.gov/32365423/
Martineau AR, Jolliffe DA, Greenberg L, et al. Vitamin D supplementation to prevent acute respiratory infections: individual participant data meta-analysis. Health Technol Assess. 2019;23(2):1–44. https://pubmed.ncbi.nlm.nih.gov/30675873/
Maretzke F, Bechthold A, Egert S, et al. Role of vitamin D in preventing and treating selected extraskeletal diseases – an umbrella review. Nutrients. 2020;12(4):E969. https://pubmed.ncbi.nlm.nih.gov/32244496/
Heath AK, Kim IY, Hodge AM, English DR, Muller DC. Vitamin D status and mortality: a systematic review of observational studies. Int J Environ Res Public Health. 2019;16(3):E383. https://pubmed.ncbi.nlm.nih.gov/30700025/
Ferri E, Casati M, Cesari M, Vitale G, Arosio B. Vitamin D in physiological and pathological aging: lesson from centenarians. Rev Endocr Metab Disord. 2019;20(3):273–82. https://pubmed.ncbi.nlm.nih.gov/31654261/
Passeri G, Pini G, Troiano L, et al. Low vitamin D status, high bone turnover, and bone fractures in centenarians. J Clin Endocrinol Metab. 2003;88(11):5109–15. https://pubmed.ncbi.nlm.nih.gov/14602735/
Heath AK, Kim IY, Hodge AM, English DR, Muller DC. Vitamin D status and mortality: a systematic review of observational studies. Int J Environ Res Public Health. 2019;16(3):E383. https://pubmed.ncbi.nlm.nih.gov/30700025/
Rodríguez AJ, Scott D, Srikanth V, Ebeling P. Effect of vitamin D supplementation on measures of arterial stiffness: a systematic review and meta-analysis of randomized controlled trials. Clin Endocrinol (Oxf). 2016;84(5):645–57. https://pubmed.ncbi.nlm.nih.gov/26824510/
Hussin AM, Ashor AW, Schoenmakers I, Hill T, Mathers JC, Siervo M. Effects of vitamin D supplementation on endothelial function: a systematic review and meta-analysis of randomised clinical trials. Eur J Nutr. 2017;56(3):1095–104. https://pubmed.ncbi.nlm.nih.gov/26848580/
Biesalski HK, Tinz J. Multivitamin/mineral supplements: rationale and safety – a systematic review. Nutrition. 2017;33:76–82. https://pubmed.ncbi.nlm.nih.gov/27553772/
Zhang Y, Fang F, Tang J, et al. Association between vitamin D supplementation and mortality: systematic review and meta-analysis. BMJ. 2019;366:l4673. https://pubmed.ncbi.nlm.nih.gov/31405892/
Manson JE, Cook NR, Lee IM, et al. Vitamin D supplements and prevention of cancer and cardiovascular disease. N Engl J Med. 2019;380(1):33–44. https://pubmed.ncbi.nlm.nih.gov/30415629/
Manson JE, Cook NR, Lee IM, et al. Marine n-3 fatty acids and prevention of cardiovascular disease and cancer. N Engl J Med. 2019;380(1):23–32. https://pubmed.ncbi.nlm.nih.gov/30415637/
Giustina A, Bouillon R, Binkley N, et al. Controversies in vitamin D: a statement from the Third International Conference. JBMR Plus. 2020;4(12):e10417. https://pubmed.ncbi.nlm.nih.gov/33354643/
Frame LA, Fischer JP, Geller G, Cheskin LJ. Use of placebo in supplementation studies – vitamin D research illustrates an ethical quandary. Nutrients. 2018;10(3):347. https://pubmed.ncbi.nlm.nih.gov/29533982/
Manson JE, Cook NR, Lee IM, et al. Vitamin D supplements and prevention of cancer and cardiovascular disease. N Engl J Med. 2019;380(1):33–44. https://pubmed.ncbi.nlm.nih.gov/30415629/
Zhang Y, Fang F, Tang J, et al. Association between vitamin D supplementation and mortality: systematic review and meta-analysis. BMJ. 2019;366:l4673. https://pubmed.ncbi.nlm.nih.gov/31405892/
LeClair BM, Si C, Solomon J. Vitamin D supplementation and all-cause mortality. Am Fam Physician. 2020;102(1):online. https://pubmed.ncbi.nlm.nih.gov/32603077/
Autier P, Mullie P, Macacu A, et al. Effect of vitamin D supplementation on non-skeletal disorders: a systematic review of meta-analyses and randomised trials. Lancet Diabetes Endocrinol. 2017;5(12):986–1004. https://pubmed.ncbi.nlm.nih.gov/29102433/
Biesalski HK, Tinz J. Multivitamin/mineral supplements: rationale and safety – a systematic review. Nutrition. 2017;33:76–82. https://pubmed.ncbi.nlm.nih.gov/27553772/
Healthy diet. World Health Organization. https://cdn.who.int/media/docs/default-source/healthy-diet/healthy-diet-fact-sheet-394.pdf?sfvrsn=69f1f9a1_2&download=true. Updated August 2018. Accessed October 7, 2022.; https://www.who.int/publications/m/item/healthy-diet-factsheet394
Schwingshackl L, Schwedhelm C, Hoffmann G, et al. Food groups and risk of all-cause mortality: a systematic review and meta-analysis of prospective studies. Am J Clin Nutr. 2017;105(6):1462–73. https://pubmed.ncbi.nlm.nih.gov/28446499/
Pribis P, Shukitt-Hale B. Cognition: the new frontier for nuts and berries. Am J Clin Nutr. 2014;100 Suppl 1:347S-52S. https://pubmed.ncbi.nlm.nih.gov/24871475/
Aune D, Keum N, Giovannucci E, et al. Nut consumption and risk of cardiovascular disease, total cancer, all-cause and cause-specific mortality: a systematic review and dose-response meta-analysis of prospective studies. BMC Med. 2016;14(1):207. https://pubmed.ncbi.nlm.nih.gov/27916000/
Fadelu T, Zhang S, Niedzwiecki D, et al. Nut consumption and survival in patients with stage III colon cancer: results from CALGB 89803 (Alliance). J Clin Oncol. 2018;36(11):1112–20. https://pubmed.ncbi.nlm.nih.gov/29489429/
Fraser GE, Shavlik DJ. Risk factors for all-cause and coronary heart disease mortality in the oldest-old. The Adventist Health Study. Arch Intern Med. 1997;157(19):2249–58. https://pubmed.ncbi.nlm.nih.gov/9343002/
Ros E. Eat nuts, live longer. J Am Coll Cardiol. 2017;70(20):2533–5. https://pubmed.ncbi.nlm.nih.gov/29145953/
Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161(13):1645–52. https://pubmed.ncbi.nlm.nih.gov/11434797/
Chen GC, Zhang R, Martínez-González MA, et al. Nut consumption in relation to all-cause and cause-specific mortality: a meta-analysis 18 prospective studies. Food Funct. 2017;8(11):3893–905. https://pubmed.ncbi.nlm.nih.gov/28875220/
Aune D, Keum NN, Giovannucci E, et al. Nut consumption and risk of cardiovascular disease, total cancer, all-cause and cause-specific mortality: a systematic review and dose-response meta-analysis of prospective studies. BMC Med. 2016;14(1):207. https://pubmed.ncbi.nlm.nih.gov/27916000/
Fernández-Montero A, Bes-Rastrollo M, Barrio-López MT, et al. Nut consumption and 5-y all-cause mortality in a Mediterranean cohort: the SUN project. Nutrition. 2014;30(9):1022–7. https://pubmed.ncbi.nlm.nih.gov/24976427/
Baer HJ, Glynn RJ, Hu FB, et al. Risk factors for mortality in the Nurses’ Health Study: a competing risks analysis. Am J Epidemiol. 2011;173(3):319–29. https://pubmed.ncbi.nlm.nih.gov/21135028/
Fernández-Montero A, Martínez-González MA, Moreno-Galarraga L. Re. “Nut consumption and 5-y all-cause mortality in a Mediterranean cohort: the SUN project”: Authors’ response. Nutrition. 2015;31(10):1299–300. https://pubmed.ncbi.nlm.nih.gov/26333895/
Aune D, Keum N, Giovannucci E, et al. Nut consumption and risk of cardiovascular disease, total cancer, all-cause and cause-specific mortality: a systematic review and dose-response meta-analysis of prospective studies. BMC Med. 2016;14(1):207. https://pubmed.ncbi.nlm.nih.gov/27916000/
Kim Y, Keogh JB, Clifton PM. Does nut consumption reduce mortality and/or risk of cardiometabolic disease? An updated review based on meta-analyses. Int J Environ Res Public Health. 2019;16(24):4957. https://pubmed.ncbi.nlm.nih.gov/31817639/
Mohammadi-Sartang M, Bellissimo N, Totosy de Zepetnek JO, Bazyar H, Mahmoodi M, Mazloom Z. Effects of walnuts consumption on vascular endothelial function in humans: a systematic review and meta-analysis of randomized controlled trials. Clin Nutr ESPEN. 2018;28:52–8. https://pubmed.ncbi.nlm.nih.gov/30390893/
Schwingshackl L, Hoffmann G, Iqbal K, Schwedhelm C, Boeing H. Food groups and intermediate disease markers: a systematic review and network meta-analysis of randomized trials. Am J Clin Nutr. 2018;108(3):576–86. https://pubmed.ncbi.nlm.nih.gov/30535089/
Bitok E, Jaceldo-Siegl K, Rajaram S, et al. Favourable nutrient intake and displacement with long-term walnut supplementation among elderly: results of a randomised trial. Br J Nutr. 2017;118(3):201–9. https://pubmed.ncbi.nlm.nih.gov/28831957/
Sun Y, Liu B, Snetselaar LG, et al. Association of major dietary protein sources with all-cause and cause-specific mortality: prospective cohort study. J Am Heart Assoc. 2021;10(5):e015553. https://pubmed.ncbi.nlm.nih.gov/33624505/
Sabaté J. Nut consumption, vegetarian diets, ischemic heart disease risk, and all-cause mortality: evidence from epidemiologic studies. Am J Clin Nutr. 1999;70(3 Suppl):500S-3S. https://pubmed.ncbi.nlm.nih.gov/10479222/
Chen GC, Zhang R, Martínez-González MA, et al. Nut consumption in relation to all-cause and cause-specific mortality: a meta-analysis 18 prospective studies. Food Funct. 2017;8(11):3893–905. https://pubmed.ncbi.nlm.nih.gov/28875220/
Aune D, Keum NN, Giovannucci E, et al. Nut consumption and risk of cardiovascular disease, total cancer, all-cause and cause-specific mortality: a systematic review and dose-response meta-analysis of prospective studies. BMC Med. 2016;14(1):207. https://pubmed.ncbi.nlm.nih.gov/27916000/
Dreher ML, Maher CV, Kearney P. The traditional and emerging role of nuts in healthful diets. Nutr Rev. 2009;54(8):241–5. https://pubmed.ncbi.nlm.nih.gov/8961751/
Guarneiri LL, Cooper JA. Intake of nuts or nut products does not lead to weight gain, independent of dietary substitution instructions: a systematic review and meta-analysis of randomized trials. Adv Nutr. 2021;12(2):384–401. https://pubmed.ncbi.nlm.nih.gov/32945861/
Barman AK, Goel R, Sharma M, Mahanta PJ. Acute kidney injury associated with ingestion of star fruit: acute oxalate nephropathy. Indian J Nephrol. 2016;26(6):446–8. https://pubmed.ncbi.nlm.nih.gov/27942177/
Albersmeyer M, Hilge R, Schröttle A, Weiss M, Sitter T, Vielhauer V. Acute kidney injury after ingestion of rhubarb: secondary oxalate nephropathy in a patient with type 1 diabetes. BMC Nephrol. 2012;13:141. https://pubmed.ncbi.nlm.nih.gov/23110375/
Kikuchi Y, Seta K, Ogawa Y, et al. Chaga mushroom-induced oxalate nephropathy. Clin Nephrol. 2014;81(6):440–4. https://pubmed.ncbi.nlm.nih.gov/23149251/
Gandhi A, Nasser S, Kassis Akl N, Kotadia S. Quiz page June 2016: rapidly progressive kidney failure. Am J Kidney Dis. 2016;67(6):A15–7. https://pubmed.ncbi.nlm.nih.gov/27211373/
Syed F, Mena-Gutierrez A, Ghaffar U. A case of iced-tea nephropathy. N Engl J Med. 2015;372(14):1377–8. https://pubmed.ncbi.nlm.nih.gov/25830441/
Bernardino M, Parmar MS. Oxalate nephropathy from cashew nut intake. CMAJ. 2017;189(10):E405–8. https://pubmed.ncbi.nlm.nih.gov/27956392/
Haaskjold YL, Drotningsvik A, Leh S, Marti HP, Svarstad E. Renal failure due to excessive intake of almonds in the absence of Oxalobacter formigenes. Am J Med. 2015;128(12):e29–30. https://pubmed.ncbi.nlm.nih.gov/26235248/
Garland V, Herlitz L, Regunathan-Shenk R. Diet-induced oxalate nephropathy from excessive nut and seed consumption. BMJ Case Rep. 2020;13(11):e237212. https://pubmed.ncbi.nlm.nih.gov/33257378/
Carlsen MH, Halvorsen BL, Holte K, et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 2010;9(1):3. https://pubmed.ncbi.nlm.nih.gov/20096093/
Ros E, Mataix J. Fatty acid composition of nuts – implications for cardiovascular health. Br J Nutr. 2006;96 Suppl 2:S29–35. https://pubmed.ncbi.nlm.nih.gov/17125530/
Xiao Y, Huang W, Peng C, et al. Effect of nut consumption on vascular endothelial function: a systematic review and meta-analysis of randomized controlled trials. Clin Nutr. 2018;37(3):831–9. https://pubmed.ncbi.nlm.nih.gov/28457654/
Yang J, Liu RH, Halim L. Antioxidant and antiproliferative activities of common edible nut seeds. Food Sci Tech. 2009;42(1):1–8. https://www.sciencedirect.com/science/article/abs/pii/S0023643808001771
Arias-Fernández L, Machado-Fragua MD, Graciani A, et al. Prospective association between nut consumption and physical function in older men and women. J Gerontol A Biol Sci Med Sci. 2019;74(7):1091–7. https://pubmed.ncbi.nlm.nih.gov/30052782/
Freitas-Simoes TM, Wagner M, Samieri C, Sala-Vila A, Grodstein F. Consumption of nuts at midlife and healthy aging in women. J Aging Res. 2020;2020:5651737. https://pubmed.ncbi.nlm.nih.gov/32399296/
Guasch-Ferré M, Bulló M, Martínez-González MÁ, et al. Frequency of nut consumption and mortality risk in the PREDIMED nutrition intervention trial. BMC Med. 2013;11:164. https://pubmed.ncbi.nlm.nih.gov/23866098/
Toner CD. Communicating clinical research to reduce cancer risk through diet: walnuts as a case example. Nutr Res Pract. 2014;8(4):347–51. https://pubmed.ncbi.nlm.nih.gov/25110552/
Kwok CS, Gulati M, Michos ED, et al. Dietary components and risk of cardiovascular disease and all-cause mortality: a review of evidence from meta-analyses. Eur J Prev Cardiol. 2019;26(13):1415–29. https://pubmed.ncbi.nlm.nih.gov/30971126/
Li N, Wu X, Zhuang W, et al. Green leafy vegetable and lutein intake and multiple health outcomes. Food Chem. 2021;360:130145. https://pubmed.ncbi.nlm.nih.gov/34034049/
Helander HF, Fändriks L. Surface area of the digestive tract – revisited. Scand J Gastroenterol. 2014;49(6):681–9. https://pubmed.ncbi.nlm.nih.gov/24694282/
Sheridan BS, Lefranan BSL. Intraepithelial lymphocytes: to serve and protect. Curr Gastroenterol Rep. 2010;12(6):513–21. https://pubmed.ncbi.nlm.nih.gov/20890736/
Hooper LV. You AhR what you eat: linking diet and immunity. Cell 2011;147(3):489–91. https://pubmed.ncbi.nlm.nih.gov/22036556/
Serna E, Cespedes C, Vina J. Anti-aging physiological roles of aryl hydrocarbon receptor and its dietary regulators. Int J Mol Sci. 2020;22(1):E374. https://pubmed.ncbi.nlm.nih.gov/33396477/
Esser C. Biology and function of the aryl hydrocarbon receptor: report of an international and interdisciplinary conference. Arch Toxicol. 2012;86(8):1323–9. https://pubmed.ncbi.nlm.nih.gov/22371237/
Ashida H, Fukuda I, Yamashita T, Kanazawa K. Flavones and flavonols at dietary levels inhibit a transformation of aryl hydrocarbon receptor induced by dioxin. FEBS Lett. 2000;476(3):213–7. https://pubmed.ncbi.nlm.nih.gov/10913616/
Healthy eating saves lives. Institute for Health Metrics and Evaluation. https://www.healthdata.org/infographic/healthy-eating-saves-lives. Published April 3, 2019. Accessed October 10, 2022.; https://www.healthdata.org/infographic/healthy-eating-saves-lives
Cohen AJ, Brauer M, Burnett R, et al. Estimates and 25-year trends of the global burden of disease attributable to ambient air pollution: an analysis of data from the Global Burden of Diseases Study 2015. Lancet. 2017;389(10082):1907–18. https://pubmed.ncbi.nlm.nih.gov/28408086/
Lee BJ, Kim B, Lee K. Air pollution exposure and cardiovascular disease. Toxicol Res. 2014;30(2):71–5. https://pubmed.ncbi.nlm.nih.gov/25071915/
He G, Pan Y, Tanaka T. COVID-19, city lockdowns, and air pollution: evidence from China. https://www.medrxiv.org/content/10.1101/2020.03.29.20046649v2. Published April 21, 2020. Accessed October 9, 2022.; https://www.medrxiv.org/content/10.1101/2020.03.29.20046649v2
Air pollution data portal. World Health Organization. https://www.who.int/data/gho/data/themes/air-pollution. Accessed October 7. 2022.; https://www.who.int/data/gho/data/themes/air-pollution
Lee K, Greenstone M. Air Quality Life Index: annual update. https://aqli.epic.uchicago.edu/wp-content/uploads/2021/08/AQLI_2021-Report.EnglishGlobal.pdf. Published September 2021. Accessed October 8, 2022.; https://aqli.epic.uchicago.edu/wp-content/uploads/2021/08/AQLI_2021-Report.EnglishGlobal.pdf
Schikowski T, Hüls A. Air pollution and skin aging. Curr Environ Health Rep. 2020;7(1):58–64. https://pubmed.ncbi.nlm.nih.gov/31927691/
Peters R, Ee N, Peters J, Booth A, Mudway I, Anstey KJ. Air pollution and dementia: a systematic review. J Alzheimers Dis. 2019;70(s1):S145–63. https://pubmed.ncbi.nlm.nih.gov/30775976/
Lee K, Greenstone M. Air Quality Life Index: annual update. https://aqli.epic.uchicago.edu/wp-content/uploads/2021/08/AQLI_2021-Report.EnglishGlobal.pdf. Published September 2021. Accessed October 8, 2022.; https://aqli.epic.uchicago.edu/wp-content/uploads/2021/08/AQLI_2021-Report.EnglishGlobal.pdf
Olmo NRS, do Nascimento Saldiva PH, Braga ALF, Lin CA, de Paula Santos U, Pereira LAA. A review of low-level air pollution and adverse effects on human health: implications for epidemiological studies and public policy. Clinics (Sao Paulo). 2011;66(4):681–90. https://pubmed.ncbi.nlm.nih.gov/21655765/
Carlsten C, Salvi S, Wong GWK, Chung KF. Personal strategies to minimise effects of air pollution on respiratory health: advice for providers, patients and the public. Eur Respir J. 2020;55(6):1902056. https://pubmed.ncbi.nlm.nih.gov/32241830/
Sinharay R, Gong J, Barratt B, et al. Respiratory and cardiovascular responses to walking down a traffic-polluted road compared with walking in a traffic-free area in participants aged 60 years and older with chronic lung or heart disease and age-matched healthy controls: a randomised, crossover study. Lancet. 2018;391(10118):339–49. https://pubmed.ncbi.nlm.nih.gov/29221643/
Carlsten C, Salvi S, Wong GWK, Chung KF. Personal strategies to minimise effects of air pollution on respiratory health: advice for providers, patients and the public. Eur Respir J. 2020;55(6):1902056. https://pubmed.ncbi.nlm.nih.gov/32241830/
Allen RW, Barn P. Individual– and household-level interventions to reduce air pollution exposures and health risks: a review of the recent literature. Curr Environ Health Rep. 2020;7(4):424–40. https://pubmed.ncbi.nlm.nih.gov/33241434/
Barn PK, Elliott CT, Allen RW, Kosatsky T, Rideout K, Henderson SB. Portable air cleaners should be at the forefront of the public health response to landscape fire smoke. Environ Health. 2016;15(1):116. https://pubmed.ncbi.nlm.nih.gov/27887618/
Allen RW, Barn P. Individual– and household-level interventions to reduce air pollution exposures and health risks: a review of the recent literature. Curr Environ Health Rep. 2020;7(4):424–40. https://pubmed.ncbi.nlm.nih.gov/33241434/
Dong W, Liu S, Chu M, et al. Different cardiorespiratory effects of indoor air pollution intervention with ionization air purifier: findings from a randomized, double-blind crossover study among school children in Beijing. Environ Pollut. 2019;254(Pt B):113054. https://pubmed.ncbi.nlm.nih.gov/31473392/
Liu W, Huang J, Lin Y, et al. Negative ions offset cardiorespiratory benefits of PM2.5 reduction from residential use of negative ion air purifiers. Indoor Air. 2021;31(1):220–8. https://pubmed.ncbi.nlm.nih.gov/32757287/
Healthy eating saves lives. Institute for Health Metrics and Evaluation. https://www.healthdata.org/infographic/healthy-eating-saves-lives. Published April 3, 2019. Accessed October 10, 2022.; https://www.healthdata.org/infographic/healthy-eating-saves-lives
Eagles SK, Gross AS, McLachlan AJ. The effects of cruciferous vegetable-enriched diets on drug metabolism: a systematic review and meta-analysis of dietary intervention trials in humans. Clin Pharmacol Ther. 2020;108(2):212–27. https://pubmed.ncbi.nlm.nih.gov/32086800/
Gilliland FD, Li YF, Saxon A, Diaz-Sanchez D. Effect of glutathione-S-transferase M1 and P1 genotypes on xenobiotic enhancement of allergic responses: randomised, placebo-controlled crossover study. Lancet. 2004;363(9403):119–25. https://pubmed.ncbi.nlm.nih.gov/14726165/
Ritz SA, Wan J, Diaz-Sanchez D. Sulforaphane-stimulated phase II enzyme induction inhibits cytokine production by airway epithelial cells stimulated with diesel extract. Am J Physiol Lung Cell Mol Physiol. 2007;292(1):L33–9. https://pubmed.ncbi.nlm.nih.gov/16905640/
Heber D, Li Z, Garcia-Lloret M, et al. Sulforaphane-rich broccoli sprout extract attenuates nasal allergic response to diesel exhaust particles. Food Funct. 2014;5(1):35–41. https://pubmed.ncbi.nlm.nih.gov/24287881/
Müller L, Meyer M, Bauer RN, et al. Effect of broccoli sprouts and live attenuated influenza virus on peripheral blood natural killer cells: a randomized, double-blind study. PLoS One. 2016;11(1):e0147742. https://pubmed.ncbi.nlm.nih.gov/26820305/
Riso P, Vendrame S, Del Bo’ C, et al. Effect of 10-day broccoli consumption on inflammatory status of young healthy smokers. Int J Food Sci Nutr. 2014;65(1):106–11. https://pubmed.ncbi.nlm.nih.gov/23992556/
Egner PA, Chen JG, Zarth AT, et al. Rapid and sustainable detoxication of airborne pollutants by broccoli sprout beverage: results of a randomized clinical trial in China. Cancer Prev Res (Phila). 2014;7(8):813–23. https://pubmed.ncbi.nlm.nih.gov/24913818/
Clarke JD, Hsu A, Riedl K, et al. Bioavailability and inter-conversion of sulforaphane and erucin in human subjects consuming broccoli sprouts or broccoli supplement in a cross-over study design. Pharmacol Res. 2011;64(5):456–63. https://pubmed.ncbi.nlm.nih.gov/21816223/
Atwell LL, Hsu A, Wong CP, et al. Absorption and chemopreventive targets of sulforaphane in humans following consumption of broccoli sprouts or a myrosinase-treated broccoli sprout extract. Mol Nutr Food Res. 2015;59(3):424–33. https://pubmed.ncbi.nlm.nih.gov/25522265/
Larsen FJ, Schiffer TA, Ekblom B, et al. Dietary nitrate reduces resting metabolic rate: a randomized, crossover study in humans. Am J Clin Nutr. 2014;99(4):843–50. https://pubmed.ncbi.nlm.nih.gov/24500154/
Larsen FJ, Schiffer TA, Borniquel S, et al. Dietary inorganic nitrate improves mitochondrial efficiency in humans. Cell Metabolism. 2011;13(2):149–59. https://pubmed.ncbi.nlm.nih.gov/21284982/
Engan HK, Jones AM, Ehrenberg F, Schagatay E. Acute dietary nitrate supplementation improves dry static apnea performance. Respir Physiol Neurobiol. 2012;182(2–3):53–9. https://pubmed.ncbi.nlm.nih.gov/22588047/
Bailey SJ, Winyard P, Vanhatalo A, et al. Dietary nitrate supplementation reduces the O2 cost of low-intensity exercise and enhances tolerance to high-intensity exercise in humans. J Appl Physiol. 2009;107(4):1144–55. https://pubmed.ncbi.nlm.nih.gov/19661447/
European Food Safety Authority. Nitrate in vegetables: Scientific Opinion of the Panel on Contaminants in the Food chain. EFSA J. 2008;689:1–79. https://efsa.onlinelibrary.wiley.com/doi/10.2903/j.efsa.2008.689
Rocha BS, Laranjinha J. Nitrate from diet might fuel gut microbiota metabolism: minding the gap between redox signaling and inter-kingdom communication. Free Radic Biol Med. 2020;149:37–43. https://pubmed.ncbi.nlm.nih.gov/32045656/
Larsen FJ, Schiffer TA, Ekblom B, et al. Dietary nitrate reduces resting metabolic rate: a randomized, crossover study in humans. Am J Clin Nutr. 2014;99(4):843–50. https://pubmed.ncbi.nlm.nih.gov/24500154/
What we eat in America, NHANES 2017–March 2020 prepandemic. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/1720/Table_1_NIN_GEN_1720.pdf. Published 2022. Accessed January 13, 2023.; https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/1720/Table_1_NIN_GEN_1720.pdf
Larsen FJ, Schiffer TA, Ekblom B, et al. Dietary nitrate reduces resting metabolic rate: a randomized, crossover study in humans. Am J Clin Nutr. 2014;99(4):843–50. https://pubmed.ncbi.nlm.nih.gov/24500154/
Kwok CS, Gulati M, Michos ED, et al. Dietary components and risk of cardiovascular disease and all-cause mortality: a review of evidence from meta-analyses. Eur J Prev Cardiol. 2019;26(13):1415–29. https://pubmed.ncbi.nlm.nih.gov/30971126/
Van De Walle GP, Vukovich MD. The effect of nitrate supplementation on exercise tolerance and performance: a systematic review and meta-analysis. J Strength Cond Res. 2018;32(6):1796–808. https://pubmed.ncbi.nlm.nih.gov/29786633/
Campos HO, Drummond LR, Rodrigues QT, et al. Nitrate supplementation improves physical performance specifically in non-athletes during prolonged open-ended tests: a systematic review and meta-analysis. Br J Nutr. 2018;119(6):636–57. https://pubmed.ncbi.nlm.nih.gov/29553034/
Bailey SJ, Winyard P, Vanhatalo A, et al. Dietary nitrate supplementation reduces the O2 cost of low-intensity exercise and enhances tolerance to high-intensity exercise in humans. J Appl Physiol (1985). 2009;107(4):1144–55. https://pubmed.ncbi.nlm.nih.gov/19661447/
Lara J, Ashor AW, Oggioni C, Ahluwalia A, Mathers JC, Siervo M. Effects of inorganic nitrate and beetroot supplementation on endothelial function: a systematic review and meta-analysis. Eur J Nutr. 2016;55(2):451–9. https://pubmed.ncbi.nlm.nih.gov/25764393/
Carter SJ, Gruber AH, Raglin JS, Baranauskas MN, Coggan AR. Potential health effects of dietary nitrate supplementation in aging and chronic degenerative disease. Med Hypotheses. 2020;141:109732. https://pubmed.ncbi.nlm.nih.gov/32294579/
Sim M, Lewis JR, Blekkenhorst LC, et al. Dietary nitrate intake is associated with muscle function in older women. J Cachexia Sarcopenia Muscle. 2019;10(3):601–10. https://pubmed.ncbi.nlm.nih.gov/30907070/
Coggan AR, Hoffman RL, Gray DA, et al. A single dose of dietary nitrate increases maximal knee extensor angular velocity and power in healthy older men and women. J Gerontol A Biol Sci Med Sci. 2020;75(6):1154–60. https://pubmed.ncbi.nlm.nih.gov/31231758/
de Oliveira GV, Morgado M, Conte-Junior CA, Alvares TS. Acute effect of dietary nitrate on forearm muscle oxygenation, blood volume and strength in older adults: a randomized clinical trial. PLoS One. 2017;12(11):e0188893. https://pubmed.ncbi.nlm.nih.gov/29190751/
Borlaug BA. Cardiac aging and the fountain of youth. Eur J Heart Fail. 2016;18(6):611–2. https://pubmed.ncbi.nlm.nih.gov/27072490/
Rammos C, Hendgen-Cotta UB, Totzeck M, et al. Impact of dietary nitrate on age-related diastolic dysfunction. Eur J Heart Fail. 2016;18(6):599–610. https://pubmed.ncbi.nlm.nih.gov/27118445/
Lara J, Ashor AW, Oggioni C, Ahluwalia A, Mathers JC, Siervo M. Effects of inorganic nitrate and beetroot supplementation on endothelial function: a systematic review and meta-analysis. Eur J Nutr. 2016;55(2):451–9. https://pubmed.ncbi.nlm.nih.gov/25764393/
Walker MA, Bailey TG, McIlvenna L, Allen JD, Green DJ, Askew CD. Acute dietary nitrate supplementation improves flow mediated dilatation of the superficial femoral artery in healthy older males. Nutrients. 2019;11(5):E954. https://pubmed.ncbi.nlm.nih.gov/31035478/
Kenjale AA, Ham KL, Stabler T, et al. Dietary nitrate supplementation enhances exercise performance in peripheral arterial disease. J Appl Physiol (1985). 2011;110(6):1582–91. https://pubmed.ncbi.nlm.nih.gov/21454745/
McDonagh ST, Wylie LJ, Morgan PT, Vanhatalo A, Jones AM. A randomised controlled trial exploring the effects of different beverages consumed alongside a nitrate-rich meal on systemic blood pressure. Nutr Health. 2018;24(3):183–92. https://pubmed.ncbi.nlm.nih.gov/30099933/
Murphy M, Eliot K, Heuertz RM, Weiss E. Whole beetroot consumption acutely improves running performance. J Acad Nutr Diet. 2012;112(4):548–52. https://pubmed.ncbi.nlm.nih.gov/22709704/
Clements WT, Lee SR, Bloomer RJ. Nitrate ingestion: a review of the health and physical performance effects. Nutrients. 2014;6(11):5224–64. https://pubmed.ncbi.nlm.nih.gov/25412154/
Hord NG, Tang Y, Bryan NS. Food sources of nitrates and nitrites: the physiologic context for potential health benefits. Am J Clin Nutr. 2009;90(1):1–10. https://pubmed.ncbi.nlm.nih.gov/19439460/
Siervo M, Lara J, Ogbonmwan I, Mathers JC. Inorganic nitrate and beetroot juice supplementation reduces blood pressure in adults: a systematic review and meta-analysis. J Nutr. 2013;143(6):818–26. https://pubmed.ncbi.nlm.nih.gov/23596162/
Blekkenhorst LC, Lewis JR, Prince RL, et al. Nitrate-rich vegetables do not lower blood pressure in individuals with mildly elevated blood pressure: a 4-wk randomized controlled crossover trial. Am J Clin Nutr. 2018;107(6):894–908. https://pubmed.ncbi.nlm.nih.gov/29868911/
Rosier BT, Buetas E, Moya-Gonzalvez EM, Artacho A, Mira A. Nitrate as a potential prebiotic for the oral microbiome. Sci Rep. 2020;10(1):12895. https://pubmed.ncbi.nlm.nih.gov/32732931/
Bondonno CP, Liu AH, Croft KD, et al. Antibacterial mouthwash blunts oral nitrate reduction and increases blood pressure in treated hypertensive men and women. Am J Hypertens. 2015;28(5):572–5. https://pubmed.ncbi.nlm.nih.gov/25359409/
Tribble GD, Angelov N, Weltman R, et al. Frequency of tongue cleaning impacts the human tongue microbiome composition and enterosalivary circulation of nitrate. Front Cell Infect Microbiol. 2019;9:39. https://pubmed.ncbi.nlm.nih.gov/30881924/
Redmond AM, Meiklejohn C, Kidd TJ, Horvath R, Coulter C. Endocarditis after use of tongue scraper. Emerg Infect Dis. 2007;13(9):1440–1. https://pubmed.ncbi.nlm.nih.gov/18252139/
Capurso A, Capurso C. The Mediterranean way. Should elderly people eat leafy vegetables and beetroot to lower high blood pressure? Aging Clin Exp Res. 2021;33(9):2613–21. https://pubmed.ncbi.nlm.nih.gov/33389684/
Strazzullo P, Ferro-Luzzi A, Siani A, et al. Changing the Mediterranean diet: effects on blood pressure. J Hypertens. 1986;4(4):407–12. https://pubmed.ncbi.nlm.nih.gov/3534087/
Dellavalle CT, Daniel CR, Aschebrook-Kilfoy B, et al. Dietary intake of nitrate and nitrite and risk of renal cell carcinoma in the NIH-AARP Diet and Health Study. Br J Cancer. 2013;108(1):205–12. https://pubmed.ncbi.nlm.nih.gov/23169285/
Sebranek JG, Jackson-Davis AL, Myers KL, Lavieri NA. Beyond celery and starter culture: advances in natural/organic curing processes in the United States. Meat Sci. 2012 Nov;92(3):267–73. https://pubmed.ncbi.nlm.nih.gov/22445489/
Rohrmann S, Overvad K, Bueno-de-Mesquita HB, et al. Meat consumption and mortality – results from the European Prospective Investigation into Cancer and Nutrition. BMC Med. 2013;11:63. https://pubmed.ncbi.nlm.nih.gov/23497300/
Sinha R, Cross AJ, Graubard BI, Leitzmann MF, Schatzkin A. Meat intake and mortality: a prospective study of over half a million people. Arch Intern Med. 2009;169(6):562–71. https://pubmed.ncbi.nlm.nih.gov/19307518/
American Institute for Cancer Research. Recommendations for Cancer Prevention. http://www.aicr.org/reduce-your-cancer-risk/recommendations-for-cancer-prevention/recommendations_05_red_meat.html. April 17, 2011. Accessed October 7, 2022.; https://www.aicr.org/reduce-your-cancer-risk/recommendations-for-cancer-prevention/recommendations_05_red_meat.html
Vermeer ITM, Pachen DMFA, Dallinga JW, Kleinjans JCS, van Maanen JMS. Volatile N-nitrosamine formation after intake of nitrate at the ADI level in combination with an amine-rich diet. Environ Health Perspect. 1998;106(8):459–63. https://pubmed.ncbi.nlm.nih.gov/9681972/
van Breda SG, Mathijs K, Sági-Kiss V, et al. Impact of high drinking water nitrate levels on the endogenous formation of apparent N-nitroso compounds in combination with meat intake in healthy volunteers. Environ Health. 2019;18(1):87. https://pubmed.ncbi.nlm.nih.gov/31623611/
Berends JE, van den Berg LMM, Guggeis MA, et al. Consumption of nitrate-rich beetroot juice with or without vitamin C supplementation increases the excretion of urinary nitrate, nitrite, and N-nitroso compounds in humans. Int J Mol Sci. 2019;20(9):E2277. https://pubmed.ncbi.nlm.nih.gov/31072023/
Bartsch H, Ohshima H, Pignatelli B. Inhibitors of endogenous nitrosation. Mechanisms and implications in human cancer prevention. Mutat Res. 1988;202(2):307–24. https://pubmed.ncbi.nlm.nih.gov/3057363/
Dellavalle CT, Daniel CR, Aschebrook-kilfoy B, et al. Dietary intake of nitrate and nitrite and risk of renal cell carcinoma in the NIH-AARP Diet and Health Study. Br J Cancer. 2013;108(1):205–12. https://pubmed.ncbi.nlm.nih.gov/23169285/
Vermeer ITM, Pachen DMFA, Dallinga JW, Kleinjans JCS, van Maanen JMS. Volatile N-nitrosamine formation after intake of nitrate at the ADI level in combination with an amine-rich diet. Environ Health Perspect. 1998;106(8):459–63. https://pubmed.ncbi.nlm.nih.gov/9681972/
Grivetti LE, Corlett JL, Gordon BM, Lockett GT. Food in American history: Part 10. Greens: Part 1. Vegetable greens in a historical context. Nutr Today. 2008;42(2):88–94. https://journals.lww.com/nutritiontodayonline/Abstract/2007/03000/Food_in_American_History__Part_10__Greens__Part_1_.10.aspx
Krebs-Smith SM, Guenther PM, Subar AF, Kirkpatrick SI, Dodd KW. Americans do not meet federal dietary recommendations. J Nutr. 2010;140(10):1832–8. https://pubmed.ncbi.nlm.nih.gov/20702750/
Sobko T, Marcus C, Govoni M, Kamiya S. Dietary nitrate in Japanese traditional foods lowers diastolic blood pressure in healthy volunteers. Nitric Oxide. 2010;22(2):136–40. https://pubmed.ncbi.nlm.nih.gov/19887114/
Tamakoshi A, Tamakoshi K, Lin Y, Yagyu K, et al. Healthy lifestyle and preventable death: findings from the Japan Collaborative Cohort (JACC) Study. Prev Med. 2009;48(5):486–92. https://pubmed.ncbi.nlm.nih.gov/19254743/
Hung HC, Joshipura KJ, Jiang R, et al. Fruit and vegetable intake and risk of major chronic disease. J Natl Cancer Inst. 2004;96(21):1577–84. https://pubmed.ncbi.nlm.nih.gov/15523086/
Joshipura KJ, Hu FB, Manson JE, et al. The effect of fruit and vegetable intake on risk for coronary heart disease. Ann Intern Med. 2001;134(12):1106–14. https://pubmed.ncbi.nlm.nih.gov/11412050/
Joshipura KJ, Ascherio A, Manson JE, et al. Fruit and vegetable intake in relation to risk of ischemic stroke. JAMA. 1999;282(13):1233–9. https://pubmed.ncbi.nlm.nih.gov/10517425/
Kwok CS, Gulati M, Michos ED, et al. Dietary components and risk of cardiovascular disease and all-cause mortality: a review of evidence from meta-analyses. Eur J Prev Cardiol. 2019;26(13):1415–29. https://pubmed.ncbi.nlm.nih.gov/30971126/
Walker FB. Myocardial infarction after diet-induced warfarin resistance. Arch Intern Med. 1984;144(10):2089–90. https://pubmed.ncbi.nlm.nih.gov/6486994/
Herforth A, Arimond M, Álvarez-Sánchez C, Coates J, Christianson K, Muehlhoff E. A global review of food-based dietary guidelines. Adv Nutr. 2019;10(4):590–605. https://pubmed.ncbi.nlm.nih.gov/31041447/
Aune D, Giovannucci E, Boffetta P, et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality – a systematic review and dose-response meta-analysis of prospective studies. Int J Epidemiol. 2017;46(3):1029–56. https://pubmed.ncbi.nlm.nih.gov/28338764/
Hjartåker A, Knudsen MD, Tretli S, Weiderpass E. Consumption of berries, fruits and vegetables and mortality among 10,000 Norwegian men followed for four decades. Eur J Nutr. 2015;54(4):599–608. https://pubmed.ncbi.nlm.nih.gov/25087093/
Aune D, Giovannucci E, Boffetta P, et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality – a systematic review and dose-response meta-analysis of prospective studies. Int J Epidemiol. 2017;46(3):1029–56. https://pubmed.ncbi.nlm.nih.gov/28338764/
Aune D, Giovannucci E, Boffetta P, et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality – a systematic review and dose-response meta-analysis of prospective studies. Int J Epidemiol. 2017;46(3):1029–56. https://pubmed.ncbi.nlm.nih.gov/28338764/
Hernandez-Marin E, Galano A, Martínez A. Cis carotenoids: colorful molecules and free radical quenchers. J Phys Chem B. 2013;117(15):4050–61. https://pubmed.ncbi.nlm.nih.gov/23560647/
Carlsen MH, Halvorsen BL, Holte K, et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 2010;9:3. https://pubmed.ncbi.nlm.nih.gov/20096093/
Souza-Monteiro JR, Arrifano GPF, Queiroz AIDG, et al. Antidepressant and antiaging effects of açaí (Euterpe oleracea Mart.) in mice. Oxid Med Cell Longev. 2019;2019:3614960. https://pubmed.ncbi.nlm.nih.gov/31428223/
Peixoto HS, Roxo M, Krstin S, Röhrig T, Richling E, Wink M. An anthocyanin-rich extract of acai (Euterpe precatoria Mart.) increases stress resistance and retards aging-related markers in Caenorhabditis elegans. J Agric Food Chem. 2016;64(6):1283–90. https://pubmed.ncbi.nlm.nih.gov/26809379/
Sun X, Seeberger J, Alberico T, et al. Açai palm fruit (Euterpe oleracea Mart.) pulp improves survival of flies on a high fat diet. Exp Gerontol. 2010;45(3):243–51. https://pubmed.ncbi.nlm.nih.gov/20080168/
Mertens-Talcott SU, Rios J, Jilma-Stohlawetz P, et al. Pharmacokinetics of anthocyanins and antioxidant effects after the consumption of anthocyanin-rich acai juice and pulp (Euterpe oleracea Mart.) in human healthy volunteers. J Agric Food Chem. 2008;56(17):7796–802. https://pubmed.ncbi.nlm.nih.gov/18693743/
Kanner J, Lapidot T. The stomach as a bioreactor: dietary lipid peroxidation in the gastric fluid and the effects of plant-derived antioxidants. Free Radic Biol Med. 2001;31(11):1388–95. https://pubmed.ncbi.nlm.nih.gov/11728810/
Macho-González A, Garcimartín A, López-Oliva ME, et al. Can meat and meat-products induce oxidative stress? Antioxidants (Basel). 2020;9(7):638. https://pubmed.ncbi.nlm.nih.gov/32698505/
Gorelik S, Kanner J, Schurr D, Kohen R. A rational approach to prevent postprandial modification of LDL by dietary polyphenols. J Funct Foods. 2013;5(1):163–9. https://www.sciencedirect.com/science/article/pii/S1756464612001466?via%3Dihub
Ayala A, Muñoz MF, Argüelles S. Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014;2014:360438. https://pubmed.ncbi.nlm.nih.gov/24999379/
Haddad E, Jambazian P, Karunia M, Tanzman J, Sabaté J. A pecan-enriched diet increases ¿-tocopherol/cholesterol and decreases thiobarbituric acid reactive substances in plasma of adults. Nutr Res. 2006;26(8):397–402. https://www.researchgate.net/publication/237724081_A_pecan-enriched_diet_increases_g-tocopherolcholesterol_and_decreases_thiobarbituric_acid_reactive_substances_in_plasma_of_adults
Carlsen MH, Halvorsen BL, Holte K, et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 2010 Jan 22;9:3. https://pubmed.ncbi.nlm.nih.gov/20096093/
Li Z, Henning SM, Zhang Y, et al. Antioxidant-rich spice added to hamburger meat during cooking results in reduced meat, plasma, and urine malondialdehyde concentrations. Am J Clin Nutr. 2010;91(5):1180–4. https://pubmed.ncbi.nlm.nih.gov/20335545/
Gobert M, Rémond D, Loonis M, Buffière C, Santé-Lhoutellier V, Dufour C. Fruits, vegetables and their polyphenols protect dietary lipids from oxidation during gastric digestion. Food Funct. 2014;5(9):2166–74. https://pubmed.ncbi.nlm.nih.gov/25029433/
Di Renzo L, Carraro A, Valente R, Iacopino L, Colica C, De Lorenzo A. Intake of red wine in different meals modulates oxidized LDL level, oxidative and inflammatory gene expression in healthy people: a randomized crossover trial. Oxid Med Cell Longev. 2014;2014:681318. https://pubmed.ncbi.nlm.nih.gov/24876915/
Natella F, Macone A, Ramberti A, et al. Red wine prevents the postprandial increase in plasma cholesterol oxidation products: a pilot study. Br J Nutr. 2011;105(12):1718–23. https://pubmed.ncbi.nlm.nih.gov/21294933/
Mellor DD, Hamer H, Smyth S, Atkin SL, Courts FL. Antioxidant-rich spice added to hamburger meat during cooking results in reduced meat, plasma, and urine malondialdehyde concentrations. Am J Clin Nutr. 2010;92(4):996–7; author reply 997. https://pubmed.ncbi.nlm.nih.gov/20720254/
Li Z, Henning SM, Zhang Y, et al. Antioxidant-rich spice added to hamburger meat during cooking results in reduced meat, plasma, and urine malondialdehyde concentrations. Am J Clin Nutr. 2010;91(5):1180–4. https://pubmed.ncbi.nlm.nih.gov/20335545/
Zhang Y, Henning SM, Lee RP, et al. Turmeric and black pepper spices decrease lipid peroxidation in meat patties during cooking. Int J Food Sci Nutr. 2015;66(3):260–5. https://pubmed.ncbi.nlm.nih.gov/25582173/
Gorelik S, Kanner J, Schurr D, Kohen R. A rational approach to prevent postprandial modification of LDL by dietary polyphenols. J Funct Foods. 2013;5(1):163–9. https://www.sciencedirect.com/science/article/pii/S1756464612001466?via%3Dihub
Kanner J, Gorelik S, Roman S, Kohen R. Protection by polyphenols of postprandial human plasma and low-density lipoprotein modification: the stomach as a bioreactor. J Agric Food Chem. 2012;60(36):8790–6. https://pubmed.ncbi.nlm.nih.gov/22530973/
Urquiaga I, Ávila F, Echeverria G, Perez D, Trejo S, Leighton F. A Chilean berry concentrate protects against postprandial oxidative stress and increases plasma antioxidant activity in healthy humans. Oxid Med Cell Longev. 2017;2017:8361493. https://pubmed.ncbi.nlm.nih.gov/28243359/
Martini S, Conte A, Bottazzi S, Tagliazucchi D. Mediterranean diet vegetable foods protect meat lipids from oxidation during in vitro gastro-intestinal digestion. Int J Food Sci Nutr. 2020;71(4):424–39. https://pubmed.ncbi.nlm.nih.gov/31610682/
Tirosh O, Shpaizer A, Kanner J. Lipid peroxidation in a stomach medium is affected by dietary oils (olive/fish) and antioxidants: the Mediterranean versus Western diet. J Agric Food Chem. 2015;63(31):7016–23. https://pubmed.ncbi.nlm.nih.gov/26165509/
Martini S, Cavalchi M, Conte A, Tagliazucchi D. The paradoxical effect of extra-virgin olive oil on oxidative phenomena during in vitro co-digestion with meat. Food Res Int. 2018;109:82–90. https://pubmed.ncbi.nlm.nih.gov/29803495/
Sebastian RS, Enns CW, Goldman JD, Hoy MK, Moshfegh AJ. Salad consumption in the U.S.: what we eat in America, NHANES 2011–2014. Food Surveys Research Group, Dietary Data Brief No. 19. https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/DBrief/19_Salad_consumption_2011_2014.pdf. Published February 2018. Accessed October 12, 2022.; https://www.ars.usda.gov/research/publications/publication/?seq№ 115=350651
Eastman P. New research on antioxidants shows surprising role for coffee. Oncology Times. 2005;27(20):39–40. https://journals.lww.com/oncology-times/fulltext/2005/10250/new_research_on_antioxidants_shows_surprising_role.32.aspx
Kanner J, Selhub J, Shpaizer A, Rabkin B, Shacham I, Tirosh O. Redox homeostasis in stomach medium by foods: The Postprandial Oxidative Stress Index (POSI) for balancing nutrition and human health. Redox Biol. 2017;12:929–36. https://pubmed.ncbi.nlm.nih.gov/28478382/
Kanner J, Selhub J, Shpaizer A, Rabkin B, Shacham I, Tirosh O. Redox homeostasis in stomach medium by foods: The Postprandial Oxidative Stress Index (POSI) for balancing nutrition and human health. Redox Biol. 2017;12:929–36. https://pubmed.ncbi.nlm.nih.gov/28478382/
Kanner J, Selhub J, Shpaizer A, Rabkin B, Shacham I, Tirosh O. Redox homeostasis in stomach medium by foods: The Postprandial Oxidative Stress Index (POSI) for balancing nutrition and human health. Redox Biol. 2017;12:929–36. https://pubmed.ncbi.nlm.nih.gov/28478382/
Timoshnikov VA, Kobzeva TV, Polyakov NE, Kontoghiorghes GJ. Redox interactions of vitamin C and iron: inhibition of the pro-oxidant activity by deferiprone. Int J Mol Sci. 2020;21(11):3967. https://pubmed.ncbi.nlm.nih.gov/32486511/
Van Hecke T, Wouters A, Rombouts C, et al. Reducing compounds equivocally influence oxidation during digestion of a high-fat beef product, which promotes cytotoxicity in colorectal carcinoma cell lines. J Agric Food Chem. 2016;64(7):1600–9. https://pubmed.ncbi.nlm.nih.gov/26836477/
Baliga MS, Dsouza JJ. Amla (Emblica officinalis Gaertn), a wonder berry in the treatment and prevention of cancer. Eur J Cancer Prev. 2011;20(3):225–39. https://pubmed.ncbi.nlm.nih.gov/21317655/
Carlsen MH, Halvorsen BL, Holte K, et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr J. 2010;9:3. https://pubmed.ncbi.nlm.nih.gov/20096093/
Pathak P, Prasad BRG, Murthy NA, Hegde SN. The effect of Emblica officinalis diet on lifespan, sexual behavior, and fitness characters in Drosophila melanogaster. Ayu. 2011;32(2):279–84. https://pubmed.ncbi.nlm.nih.gov/22408317/
Variya BC, Bakrania AK, Patel SS. Emblica officinalis (amla): a review for its phytochemistry, ethnomedicinal uses and medicinal potentials with respect to molecular mechanisms. Pharmacol Res. 2016;111:180–200. https://pubmed.ncbi.nlm.nih.gov/27320046/
Akhtar MS, Ramzan A, Ali A, Ahmad M. Effect of Amla fruit (Emblica officinalis Gaertn.) on blood glucose and lipid profile of normal subjects and type 2 diabetic patients. Int J Food Sci Nutr. 2011;62(6):609–16. https://pubmed.ncbi.nlm.nih.gov/21495900/
Akhtar MS, Ramzan A, Ali A, Ahmad M. Effect of Amla fruit (Emblica officinalis Gaertn.) on blood glucose and lipid profile of normal subjects and type 2 diabetic patients. Int J Food Sci Nutr. 2011;62(6):609–16. https://pubmed.ncbi.nlm.nih.gov/21495900/
Kapoor MP, Suzuki K, Derek T, Ozeki M, Okubo T. Clinical evaluation of Emblica officinalis Gatertn (amla) in healthy human subjects: health benefits and safety results from a randomized, double-blind, crossover placebo-controlled study. Contemp Clin Trials Commun. 2020;17:100499. https://pubmed.ncbi.nlm.nih.gov/31890983/
Usharani P, Merugu PL, Nutalapati C. Evaluation of the effects of a standardized aqueous extract of Phyllanthus emblica fruits on endothelial dysfunction, oxidative stress, systemic inflammation and lipid profile in subjects with metabolic syndrome: a randomised, double blind, placebo controlled clinical study. BMC Complement Altern Med. 2019;19:97. https://pubmed.ncbi.nlm.nih.gov/31060549/
Akhtar MS, Ramzan A, Ali A, Ahmad M. Effect of Amla fruit (Emblica officinalis Gaertn.) on blood glucose and lipid profile of normal subjects and type 2 diabetic patients. Int J Food Sci Nutr. 2011;62(6):609–16. https://pubmed.ncbi.nlm.nih.gov/21495900/
Fatima N, Pingali U, Pilli R. Evaluation of Phyllanthus emblica extract on cold pressor induced cardiovascular changes in healthy human subjects. Pharmacognosy Res. 2014;6(1):29–35. https://pubmed.ncbi.nlm.nih.gov/24497739/
Karkon Varnosfaderani S, Hashem-Dabaghian F, Amin G, et al. Efficacy and safety of amla (Phyllanthus emblica L.) in non-erosive reflux disease: a double-blind, randomized, placebo-controlled clinical trial. J Integr Med. 2018;16(2):126–31. https://pubmed.ncbi.nlm.nih.gov/29526236/
Minich DM. A review of the science of colorful, plant-based food and practical strategies for “eating the rainbow.” J Nutr Metab. 2019;2019:2125070. https://pubmed.ncbi.nlm.nih.gov/33414957/
America’s phytonutrient report: quantifying the gap. Nutrilite Health Institute. https://www.pwrnewmedia.com/2009/nutrilite90921nmr/downloads/AmerciasPhytonutrientReport.pdf. Published September 11, 2009. Updated January 13, 2010. Accessed October 12, 2022.; https://www.pwrnewmedia.com/2009/nutrilite90921nmr/downloads/AmerciasPhytonutrientReport.pdf
Wood E, Hein S, Heiss C, Williams C, Rodriguez-Mateos A. Blueberries and cardiovascular disease prevention. Food Funct. 2019;10(12):7621–33. https://pubmed.ncbi.nlm.nih.gov/31776541/
Tena N, Asuero AG. Antioxidant capacity of anthocyanins and other vegetal pigments. Antioxidants (Basel). 2020;9(8):665. https://pubmed.ncbi.nlm.nih.gov/32722520/
Hair R, Sakaki JR, Chun OK. Anthocyanins, microbiome and health benefits in aging. Molecules. 2021;26(3):537. https://pubmed.ncbi.nlm.nih.gov/33494165/
Bonyadi N, Dolatkhah N, Salekzamani Y, Hashemian M. Effect of berry-based supplements and foods on cognitive function: a systematic review. Sci Rep. 2022;12(1):3239. https://pubmed.ncbi.nlm.nih.gov/35217779/
Lee J, Lee HK, Kim CY, et al. Purified high-dose anthocyanoside oligomer administration improves nocturnal vision and clinical symptoms in myopia subjects. Br J Nutr. 2005;93(6):895–9. https://pubmed.ncbi.nlm.nih.gov/16022759/
Camire ME. Bilberries and blueberries as functional foods and nutraceuticals. In: Mazza G, Oomah BD, eds. Herbs, Botanicals & Teas. Technomic Publishing Co Inc; 2000:289–319. https://worldcat.org/title/44440827
Nakaishi H, Matsumoto H, Tominaga S, Hirayama M. Effects of black current anthocyanoside intake on dark adaptation and VDT work-induced transient refractive alteration in healthy humans. Altern Med Rev. 2000;5(6):553–62. Erratum in: Altern Med Rev. 2001;6(1):60. https://pubmed.ncbi.nlm.nih.gov/11134978/
Camire ME. Bilberries and blueberries as functional foods and nutraceuticals. In: Mazza G, Oomah BD, eds. Herbs, Botanicals & Teas. Technomic Publishing Co Inc; 2000:289–319. https://worldcat.org/title/44440827
Xu L, Tian Z, Chen H, Zhao Y, Yang Y. Anthocyanins, anthocyanin-rich berries, and cardiovascular risks: systematic review and meta-analysis of 44 randomized controlled trials and 15 prospective cohort studies. Front Nutr. 2021;8:747884. https://pubmed.ncbi.nlm.nih.gov/34977111/
Biedermann L, Mwinyi J, Scharl M, et al. Bilberry ingestion improves disease activity in mild to moderate ulcerative colitis – an open pilot study. J Crohns Colitis. 2013;7(4):271–9. https://pubmed.ncbi.nlm.nih.gov/22883440/
Hair R, Sakaki JR, Chun OK. Anthocyanins, microbiome and health benefits in aging. Molecules. 2021;26(3):537. https://pubmed.ncbi.nlm.nih.gov/33494165/
Vendrame S, Guglielmetti S, Riso P, Arioli S, Klimis-Zacas D, Porrini M. Six-week consumption of a wild blueberry powder drink increases bifidobacteria in the human gut. J Agric Food Chem. 2011;59(24):12815–20. https://pubmed.ncbi.nlm.nih.gov/22060186/
Molan AL, Liu Z, Plimmer G. Evaluation of the effect of blackcurrant products on gut microbiota and on markers of risk for colon cancer in humans. Phytother Res. 2014;28(3):416–22. https://pubmed.ncbi.nlm.nih.gov/23674271/
Mayta-Apaza AC, Pottgen E, De Bodt J, et al. Impact of tart cherries polyphenols on the human gut microbiota and phenolic metabolites in vitro and in vivo. J Nutr Biochem. 2018;59:160–72. https://pubmed.ncbi.nlm.nih.gov/30055451/
Fallah AA, Sarmast E, Jafari T. Effect of dietary anthocyanins on biomarkers of glycemic control and glucose metabolism: a systematic review and meta-analysis of randomized clinical trials. Food Res Int. 2020;137:109379. https://pubmed.ncbi.nlm.nih.gov/33233081/
Stull AJ, Cash KC, Johnson WD, Champagne CM, Cefalu WT. Bioactives in blueberries improve insulin sensitivity in obese, insulin-resistant men and women. J Nutr. 2010;140(10):1764–8. https://pubmed.ncbi.nlm.nih.gov/20724487/
Kimble R, Keane KM, Lodge JK, Howatson G. Dietary intake of anthocyanins and risk of cardiovascular disease: a systematic review and meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr. 2019;59(18):3032–43. https://pubmed.ncbi.nlm.nih.gov/30277799/
Wedick NM, Pan A, Cassidy A, et al. Dietary flavonoid intakes and risk of type 2 diabetes in US men and women. Am J Clin Nutr. 2012;95(4):925–33. https://pubmed.ncbi.nlm.nih.gov/22357723/
Ahles S, Joris PJ, Plat J. Effects of berry anthocyanins on cognitive performance, vascular function and cardiometabolic risk markers: a systematic review of randomized placebo-controlled intervention studies in humans. Int J Mol Sci. 2021;22(12):6482. https://pubmed.ncbi.nlm.nih.gov/34204250/
Xu L, Tian Z, Chen H, Zhao Y, Yang Y. Anthocyanins, anthocyanin-rich berries, and cardiovascular risks: systematic review and meta-analysis of 44 randomized controlled trials and 15 prospective cohort studies. Front Nutr. 2021;8:747884. https://pubmed.ncbi.nlm.nih.gov/34977111/
Aune D, Giovannucci E, Boffetta P, et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality – a systematic review and dose-response meta-analysis of prospective studies. Int J Epidemiol. 2017;46(3):1029–56. https://pubmed.ncbi.nlm.nih.gov/28338764/
Del Bo’ C, Porrini M, Fracassetti D, Campolo J, Klimis-Zacas D, Riso P. A single serving of blueberry (V. corymbosum) modulates peripheral arterial dysfunction induced by acute cigarette smoking in young volunteers: a randomized-controlled trial. Food Funct. 2014;5(12):3107–16. https://pubmed.ncbi.nlm.nih.gov/25263326/
Zhu Y, Xia M, Yang Y, et al. Purified anthocyanin supplementation improves endothelial function via NO-cGMP activation in hypercholesterolemic individuals. Clin Chem. 2011;57(11):1524–33. https://pubmed.ncbi.nlm.nih.gov/21926181/
Rodriguez-Mateos A, Istas G, Boschek L, et al. Circulating anthocyanin metabolites mediate vascular benefits of blueberries: insights from randomized controlled trials, metabolomics, and nutrigenomics. J Gerontol A Biol Sci Med Sci. 2019;74(7):967–76. https://pubmed.ncbi.nlm.nih.gov/30772905/
Daneshzad E, Shab-Bidar S, Mohammadpour Z, Djafarian K. Effect of anthocyanin supplementation on cardio-metabolic biomarkers: a systematic review and meta-analysis of randomized controlled trials. Clin Nutr. 2019;38(3):1153–65. https://pubmed.ncbi.nlm.nih.gov/30007479/
Horbowicz M, Kosson R, Grzesiuk A, Debski H. Anthocyanins of fruits and vegetables – their occurrence, analysis and role in human nutrition. J Fruit Ornam Plant Res. 2008;68(1):5–22. https://www.researchgate.net/publication/284789289_Anthocyanins_of_fruits_and_vegetables-their_occurrence_analysis_and_role_in_human_nutrition
Ucar SK, Sözmen E, Yildirim HK, Coker M. Effect of blueberry tea on lipid and antioxidant status in children with heterozygous familial hypercholesterolemia: pilot study. Clin Lipidol. 2014;9(3):295–304. https://www.tandfonline.com/doi/pdf/10.2217/clp.14.26?needAccess=true
Pojer E, Mattivi F, Johnson D, Stockley CS. The case for anthocyanin consumption to promote human health: a review. Compr Rev Food Sci Food Saf. 2013;12(5):483–508. https://pubmed.ncbi.nlm.nih.gov/33412667/
Mattioli R, Francioso A, Mosca L, Silva P. Anthocyanins: a comprehensive review of their chemical properties and health effects on cardiovascular and neurodegenerative diseases. Molecules. 2020;25(17):E3809. https://pubmed.ncbi.nlm.nih.gov/32825684/
Lu X, Zhou Y, Wu T, Hao L. Ameliorative effect of black rice anthocyanin on senescent mice induced by D-galactose. Food Funct. 2014;5(11):2892–7. https://pubmed.ncbi.nlm.nih.gov/25190075/
Chen W, Müller D, Richling E, Wink M. Anthocyanin-rich purple wheat prolongs the life span of Caenorhabditis elegans probably by activating the DAF-16/FOXO transcription factor. J Agric Food Chem. 2013;61(12):3047–53. https://pubmed.ncbi.nlm.nih.gov/23470220/
Blau LW. Cherry diet control for gout and arthritis. Tex Rep Biol Med. 1950;8(3):309–11. https://pubmed.ncbi.nlm.nih.gov/14776685/
Kelley DS, Rasooly R, Jacob RA, Kader AA, Mackey BE. Consumption of Bing sweet cherries lowers circulating concentrations of inflammation markers in healthy men and women. J Nutr. 2006;136(4):981–6. https://pubmed.ncbi.nlm.nih.gov/16549461/
Kelley DS, Adkins Y, Laugero KD. A review of the health benefits of cherries. Nutrients. 2018;10(3):368. https://pubmed.ncbi.nlm.nih.gov/29562604/
Sun Y, Yolitz J, Alberico T, Sun X, Zou S. Lifespan extension by cranberry supplementation partially requires SOD2 and is life stage independent. Exp Gerontol. 2014;50:57–63. https://pubmed.ncbi.nlm.nih.gov/24316039/
Guha S, Cao M, Kane RM, Savino AM, Zou S, Dong Y. The longevity effect of cranberry extract in Caenorhabditis elegans is modulated by daf-16 and osr-1. Age (Dordr). 2013;35(5):1559–74. https://pubmed.ncbi.nlm.nih.gov/22864793/
Zhu M, Hu J, Perez E, et al. Effects of long-term cranberry supplementation on endocrine pancreas in aging rats. J Gerontol A Biol Sci Med Sci. 2011;66(11):1139–51. https://pubmed.ncbi.nlm.nih.gov/21768504/
Gao Y, Wei Y, Wang Y, Gao F, Chen Z. Lycium barbarum: a traditional Chinese herb and a promising anti-aging agent. Aging and Disease. 2017;8(6):778. https://pubmed.ncbi.nlm.nih.gov/29344416/
Neelam K, Dey S, Sim R, Lee J, Au Eong KG. Fructus lycii: a natural dietary supplement for amelioration of retinal diseases. Nutrients. 2021;13(1):246. https://pubmed.ncbi.nlm.nih.gov/33467087/
Jeszka-Skowron M, Zgola-Grzeskowiak A, Stanisz E, Waskiewicz A. Potential health benefits and quality of dried fruits: goji fruits, cranberries and raisins. Food Chem. 2017;221:228–36. https://pubmed.ncbi.nlm.nih.gov/27979197/
Wu WB, Hung DK, Chang FW, Ong ET, Chen BH. Anti-inflammatory and anti-angiogenic effects of flavonoids isolated from Lycium barbarum Linnaeus on human umbilical vein endothelial cells. Food Funct. 2012;3(10):1068–81. https://pubmed.ncbi.nlm.nih.gov/22751795/
Lee YJ, Ahn Y, Kwon O, et al. Dietary wolfberry extract modifies oxidative stress by controlling the expression of inflammatory mRNAs in overweight and hypercholesterolemic subjects: a randomized, double-blind, placebo-controlled trial. J Agric Food Chem. 2017;65(2):309–16. https://pubmed.ncbi.nlm.nih.gov/28027641/
Grassi F, Arroyo-Garcia R. Editorial: origins and domestication of the grape. Front Plant Sci. 2020;11:1176. https://pubmed.ncbi.nlm.nih.gov/32903797/
Yang J, Xiao YY. Grape phytochemicals and associated health benefits. Crit Rev Food Sci Nutr. 2013;53(11):1202–25. https://pubmed.ncbi.nlm.nih.gov/24007424/
Ghaedi E, Moradi S, Aslani Z, Kord-Varkaneh H, Miraghajani M, Mohammadi H. Effects of grape products on blood lipids: a systematic review and dose-response meta-analysis of randomized controlled trials. Food Funct. 2019;10(10):6399–416. https://pubmed.ncbi.nlm.nih.gov/31517353/
Rahbar AR, Mahmoudabadi MMS, Islam MS. Comparative effects of red and white grapes on oxidative markers and lipidemic parameters in adult hypercholesterolemic humans. Food Funct. 2015;6(6):1992–8. https://pubmed.ncbi.nlm.nih.gov/26007320/
Kanellos PT, Kaliora AC, Protogerou AD, Tentolouris N, Perrea DN, Karathanos VT. The effect of raisins on biomarkers of endothelial function and oxidant damage; an open-label and randomized controlled intervention. Food Res Int. 2017;102:674–80. https://pubmed.ncbi.nlm.nih.gov/29195999/
Chaves AA, Joshi MS, Coyle CM, et al. Vasoprotective endothelial effects of a standardized grape product in humans. Vascul Pharmacol. 2009;50(1–2):20-6. https://pubmed.ncbi.nlm.nih.gov/18805507/
Vaisman N, Niv E. Daily consumption of red grape cell powder in a dietary dose improves cardiovascular parameters: a double blind, placebo-controlled, randomized study. Int J Food Sci Nutr. 2015;66(3):342–9. https://pubmed.ncbi.nlm.nih.gov/25666417/
Yang J, Xiao YY. Grape phytochemicals and associated health benefits. Crit Rev Food Sci Nutr. 2013;53(11):1202–25. https://pubmed.ncbi.nlm.nih.gov/24007424/
Li X, Yang T, Sun Z. Hormesis in health and chronic diseases. Trends Endocrinol Metab. 2019;30(12):944–58. https://pubmed.ncbi.nlm.nih.gov/31521464/
Epel ES. The geroscience agenda: toxic stress, hormetic stress, and the rate of aging. Ageing ResRev. 2020;63:101167. https://pubmed.ncbi.nlm.nih.gov/32979553/
Collier R. Intermittent fasting: the science of going without. CMAJ. 2013;185(9):E363–4. https://pubmed.ncbi.nlm.nih.gov/23569168/
Bárcena C, Mayoral P, Quirós PM. Mitohormesis, an antiaging paradigm. Int Rev Cell Mol Biol. 2018;340:35–77. https://pubmed.ncbi.nlm.nih.gov/30072093/
Calabrese EJ, Dhawan G, Kapoor R, Iavicoli I, Calabrese V. What is hormesis and its relevance to healthy aging and longevity? Biogerontology. 2015;16(6):693–707. https://pubmed.ncbi.nlm.nih.gov/26349923/
Li X, Yang T, Sun Z. Hormesis in health and chronic diseases. Trends Endocrinol Metab. 2019;30(12):944–58. https://pubmed.ncbi.nlm.nih.gov/31521464/
Mao L, Franke J. Hormesis in aging and neurodegeneration – a prodigy awaiting dissection. Int J Mol Sci. 2013;14(7):13109–28. https://pubmed.ncbi.nlm.nih.gov/23799363/
Kaiser J. Hormesis. Sipping from a poisoned chalice. Science. 2003;302(5644):376–9. https://pubmed.ncbi.nlm.nih.gov/14563981
Calabrese EJ. Toxicology rewrites its history and rethinks its future: giving equal focus to both harmful and beneficial effects. Environ Toxicol Chem. 2011;30(12):2658–73. https://pubmed.ncbi.nlm.nih.gov/21932295/
Calabrese EJ, Dhawan G, Kapoor R, Iavicoli I, Calabrese V. What is hormesis and its relevance to healthy aging and longevity? Biogerontology. 2015;16(6):693–707. https://pubmed.ncbi.nlm.nih.gov/26349923/
Web of Science search results for ‘hormesis’ or ‘hormetic’. https://www.webofscience.com. Accessed December 20, 2022.; https://www.webofscience.com
Calabrese EJ, Dhawan G, Kapoor R, Iavicoli I, Calabrese V. What is hormesis and its relevance to healthy aging and longevity? Biogerontology. 2015;16(6):693–707. https://pubmed.ncbi.nlm.nih.gov/26349923/
Davey WP. Prolongation of life of Tribolium confusum apparently due to small doses of x-rays. J Exp Zool. 1919;28(3):447–58. https://pubmed.ncbi.nlm.nih.gov/21932295/
Calabrese EJ. Low doses of radiation can enhance insect lifespans. Biogerontology. 2013;14(4):365–81. https://pubmed.ncbi.nlm.nih.gov/23793937/
Sutou S. Low-dose radiation from A-bombs elongated lifespan and reduced cancer mortality relative to un-irradiated individuals. Genes Environ. 2018;40:26. https://pubmed.ncbi.nlm.nih.gov/30598710/
Thome C, Tharmalingam S, Pirkkanen J, Zarnke A, Laframboise T, Boreham DR. The REPAIR Project: examining the biological impacts of sub-background radiation exposure within SNOLAB, a deep underground laboratory. Radiat Res. 2017;188(4.2):470–4. https://pubmed.ncbi.nlm.nih.gov/28723273/
MacLeod RD, Van den Block L, eds. Textbook of Palliative Care. Springer International Publishing; 2019. https://www.scribd.com/document/544078970/Textbook-of-Palliative-Care
Marie Curie. NobelPrize.org. https://www.nobelprize.org/womenwhochangedscience/stories/marie-curie. Accessed December 26, 2022.; https://www.nobelprize.org/womenwhochangedscience/stories/marie-curie
Butler D. X-rays, not radium, may have killed Curie. Nature. 1995;377(6545):96. https://pubmed.ncbi.nlm.nih.gov/7675094/
Gradari S, Pallé A, McGreevy KR, Fontán-Lozano Á, Trejo JL. Can exercise make you smarter, happier, and have more neurons? A hormetic perspective. Front Neurosci. 2016;10:93. https://pubmed.ncbi.nlm.nih.gov/27013955/
Mastaloudis A, Yu TW, O’Donnell RP, Frei B, Dashwood RH, Traber MG. Endurance exercise results in DNA damage as detected by the comet assay. Free Radic Biol Med. 2004;36(8):966–75. https://pubmed.ncbi.nlm.nih.gov/15059637/
Mastaloudis A, Yu TW, O’Donnell RP, Frei B, Dashwood RH, Traber MG. Endurance exercise results in DNA damage as detected by the comet assay. Free Radic Biol Med. 2004;36(8):966–75. https://pubmed.ncbi.nlm.nih.gov/15059637/
Sharma A, Kaur T, Singh H, Kaur G. Intermittent fasting – dietary restriction as a biological hormetin for health benefits. In: The Science of Hormesis in Health and Longevity. Elsevier; 2019:99–104. https://www.sciencedirect.com/science/article/abs/pii/B9780128142530000085
Masoro EJ. The role of hormesis in life extension by dietary restriction. Interdiscip Top Gerontol. 2007;35:1–17. https://pubmed.ncbi.nlm.nih.gov/17063030/
Masoro EJ. The role of hormesis in life extension by dietary restriction. Interdiscip Top Gerontol. 2007;35:1–17. https://pubmed.ncbi.nlm.nih.gov/17063030/
Franco R, Navarro G, Martínez-Pinilla E. Hormetic and mitochondria-related mechanisms of antioxidant action of phytochemicals. Antioxidants (Basel). 2019;8(9):373. https://pubmed.ncbi.nlm.nih.gov/31487950/
Masoro EJ. The role of hormesis in life extension by dietary restriction. Interdiscip Top Gerontol. 2007;35:1–17. https://pubmed.ncbi.nlm.nih.gov/17063030/
Schultz JC. Shared signals and the potential for phylogenetic espionage between plants and animals. Integr Comp Biol. 2002;42(3):454–62. https://pubmed.ncbi.nlm.nih.gov/21708739/
Kennedy DO. Polyphenols and the human brain: plant “secondary metabolite” ecologic roles and endogenous signaling functions drive benefits. Adv Nutr. 2014;5(5):515–33. https://pubmed.ncbi.nlm.nih.gov/25469384/
Franco, Navarro, Martínez-Pinilla. Hormetic and mitochondria-related mechanisms of antioxidant action of phytochemicals. Antioxidants. 2019;8(9):373. https://pubmed.ncbi.nlm.nih.gov/31487950/
Glover N. The banana conjecture. Dessimoz Lab. https://lab.dessimoz.org/blog/2020/12/08/human-banana-orthologs. Published December 8, 2020. Accessed October 17, 2022.; https://lab.dessimoz.org/blog/2020/12/08/human-banana-orthologs
Wang DY, Kumar S, Hedges SB. Divergence time estimates for the early history of animal phyla and the origin of plants, animals and fungi. Proc Biol Sci. 1999;266(1415):163–71. https://pubmed.ncbi.nlm.nih.gov/10097391/
Kushiro T, Nambara E, McCourt P. Hormone evolution: the key to signalling. Nature. 2003;422(6928):122. https://pubmed.ncbi.nlm.nih.gov/12634761/
Kennedy DO. Polyphenols and the human brain: plant “secondary metabolite” ecologic roles and endogenous signaling functions drive benefits. Adv Nutr. 2014;5(5):515–33. https://pubmed.ncbi.nlm.nih.gov/25469384/
Schultz JC. Shared signals and the potential for phylogenetic espionage between plants and animals. Integr Comp Biol. 2002;42(3):454–62. https://pubmed.ncbi.nlm.nih.gov/21708739/
Mattson MP. Hormesis defined. Ageing Res Rev. 2008;7(1):1–7. https://pubmed.ncbi.nlm.nih.gov/18162444/
Langhans W. Food components in health promotion and disease prevention. J Agric Food Chem. 2018;66(10):2287–94. https://pubmed.ncbi.nlm.nih.gov/28603983/
Eid HM, Wright ML, Anil Kumar NV, et al. Significance of microbiota in obesity and metabolic diseases and the modulatory potential by medicinal plant and food ingredients. Front Pharmacol. 2017;8:387. https://pubmed.ncbi.nlm.nih.gov/28713266/
Hooper PL, Hooper PL, Tytell M, Vígh L. Xenohormesis: health benefits from an eon of plant stress response evolution. Cell Stress Chaperones. 2010;15(6):761–70. https://pubmed.ncbi.nlm.nih.gov/20524162/
Schultz JC. Shared signals and the potential for phylogenetic espionage between plants and animals. Integr Comp Biol. 2002;42(3):454–62. https://pubmed.ncbi.nlm.nih.gov/21708739/
Franco R, Navarro G, Martínez-Pinilla E. Hormetic and mitochondria-related mechanisms of antioxidant action of phytochemicals. Antioxidants (Basel). 2019;8(9):373. https://pubmed.ncbi.nlm.nih.gov/31487950/
Martel J, Ojcius DM, Ko YF, et al. Hormetic effects of phytochemicals on health and longevity. Trends Endocrinol Metab. 2019;30(6):335–46. https://pubmed.ncbi.nlm.nih.gov/31060881/
Dani C, Oliboni LS, Vanderlinde R, et al. Phenolic content and antioxidant activities of white and purple juices manufactured with organically– or conventionally-produced grapes. Food Chem Toxicol. 2007;45(12):2574-80. https://pubmed.ncbi.nlm.nih.gov/17683842/
Baxter GJ, Graham AB, Lawrence JR, et al. Salicylic acid in soups prepared from organically and non-organically grown vegetables. Eur J Nutr. 2001;40(6):289-92. https://pubmed.ncbi.nlm.nih.gov/11876493/
Choi SW, Yeung VTF, Collins AR, Benzie IFF. Redox-linked effects of green tea on DNA damage and repair, and influence of microsatellite polymorphism in HMOX-1: results of a human intervention trial. Mutagenesis. 2015;30(1):129–37. https://pubmed.ncbi.nlm.nih.gov/25527735/
Niu Y, Na L, Feng R, et al. The phytochemical, EGCG, extends lifespan by reducing liver and kidney function damage and improving age-associated inflammation and oxidative stress in healthy rats. Aging Cell. 2013;12(6):1041–9. https://pubmed.ncbi.nlm.nih.gov/23834676/
Yi M, Wu X, Zhuang W, et al. Tea consumption and health outcomes: umbrella review of meta-analyses of observational studies in humans. Mol Nutr Food Res. 2019;63(16):e1900389. https://pubmed.ncbi.nlm.nih.gov/31216091/
Spiegelhalter D. Using speed of ageing and “microlives” to communicate the effects of lifetime habits and environment. BMJ. 2012 Dec 14;345:e8223. https://pubmed.ncbi.nlm.nih.gov/23247978/
Zhang DD, Chapman E. The role of natural products in revealing NRF2 function. Nat Prod Rep. 2020;37(6):797–826. https://pubmed.ncbi.nlm.nih.gov/32400766/
Botonis PG, Miliotis PG, Kounalakis SN, Koskolou MD, Geladas ND. Effects of capsaicin application on the skin during resting exposure to temperate and warm conditions. Scand J Med Sci Sports. 2019;29(2):171–9. https://pubmed.ncbi.nlm.nih.gov/30294815/
Scharfenberg K, Wagner R, Wagner KG. The cytotoxic effect of ajoene, a natural product from garlic, investigated with different cell lines. Cancer Lett. 1990;53(2–3):103–8. https://pubmed.ncbi.nlm.nih.gov/2208068/
Friedman T, Shalom A, Westreich M. Self-inflicted garlic burns: our experience and literature review. Int J Dermatol. 2006;45(10):1161–3. https://pubmed.ncbi.nlm.nih.gov/17040429/
Mattson MP. Dietary factors, hormesis and health. Ageing Res Rev. 2008;7(1):43–8. https://pubmed.ncbi.nlm.nih.gov/17913594/
Murakami A. Modulation of protein quality control systems by food phytochemicals. J Clin Biochem Nutr. 2013;52(3):215–27. https://pubmed.ncbi.nlm.nih.gov/23704811/
Liu RH. Health benefits of fruit and vegetables are from additive and synergistic combinations of phytochemicals. Am J Clin Nutr. 2003;78(3 Suppl):517S-20S. https://pubmed.ncbi.nlm.nih.gov/12936943/
Holst B, Williamson G. Nutrients and phytochemicals: from bioavailability to bioefficacy beyond antioxidants. Curr Opin Biotechnol. 2008;19(2):73–82. https://pubmed.ncbi.nlm.nih.gov/18406129/
Aune D, Giovannucci E, Boffetta P, et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality – a systematic review and dose-response meta-analysis of prospective studies. Int J Epidemiol. 2017;46(3):1029–56. https://pubmed.ncbi.nlm.nih.gov/28338764/
Aune D, Giovannucci E, Boffetta P, et al. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality – a systematic review and dose-response meta-analysis of prospective studies. Int J Epidemiol. 2017;46(3):1029–56. https://pubmed.ncbi.nlm.nih.gov/28338764/
Long T, Zhang K, Chen Y, Wu C. Trends in diet quality among older US adults from 2001 to 2018. JAMA Netw Open. 2022;5(3):e221880. https://pubmed.ncbi.nlm.nih.gov/35275167/
Huang Q, Braffett BH, Simmens SJ, Young HA, Ogden CL. Dietary polyphenol intake in US adults and 10-year trends: 2007–2016. J Acad Nutr Diet. 2020;120(11):1821–33. https://pubmed.ncbi.nlm.nih.gov/32807722/
Long T, Zhang K, Chen Y, Wu C. Trends in diet quality among older US adults from 2001 to 2018. JAMA Netw Open. 2022;5(3):e221880. https://pubmed.ncbi.nlm.nih.gov/35275167/
Rathaur P, S R JK. Metabolism and pharmacokinetics of phytochemicals in the human body. Curr Drug Metab. 2019;20(14):1085–102. https://pubmed.ncbi.nlm.nih.gov/31902349/
Zhang F, Luo W, Shi Y, Fan Z, Ji G. Should we standardize the 1,700-year-old fecal microbiota transplantation? Am J Gastroenterol. 2012;107(11):1755. https://pubmed.ncbi.nlm.nih.gov/23160295/
Kong LY, Tan RX. Artemisinin, a miracle of traditional Chinese medicine. Nat Prod Rep. 2015;32(12):1617–21. https://pubmed.ncbi.nlm.nih.gov/26561737/
Sharma R, Padwad Y. Perspectives of the potential implications of polyphenols in influencing the interrelationship between oxi-inflammatory stress, cellular senescence and immunosenescence during aging. Trends Food Sci Technol. 2020;98:41–52. https://www.sciencedirect.com/science/article/abs/pii/S0924224419310969
Del Bo’ C, Bernardi S, Marino M, et al. Systematic review on polyphenol intake and health outcomes: is there sufficient evidence to define a health-promoting polyphenol-rich dietary pattern? Nutrients. 2019;11(6):E1355. https://pubmed.ncbi.nlm.nih.gov/31208133/
Williamson G, Holst B. Dietary reference intake (DRI) value for dietary polyphenols: are we heading in the right direction? Br J Nutr. 2008;99 Suppl 3:S55–8. https://pubmed.ncbi.nlm.nih.gov/18598589/
Ivey KL, Jensen MK, Hodgson JM, Eliassen AH, Cassidy A, Rimm EB. Association of flavonoid-rich foods and flavonoids with risk of all-cause mortality. Br J Nutr. 2017;117(10):1470–7. https://pubmed.ncbi.nlm.nih.gov/28606222/
Aliper A, Belikov AV, Garazha A, et al. In search for geroprotectors: in silico screening and in vitro validation of signalome-level mimetics of young healthy state. Aging (Albany NY). 2016;8(9):2127–52. https://pubmed.ncbi.nlm.nih.gov/27677171/
Lutchman V, Medkour Y, Samson E, et al. Discovery of plant extracts that greatly delay yeast chronological aging and have different effects on longevity-defining cellular processes. Oncotarget. 2016;7(13):16542–66. https://pubmed.ncbi.nlm.nih.gov/26918729/
Navrotskaya VV, Oxenkrug G, Vorobyova LI, Summergrad P. Berberine prolongs life span and stimulates locomotor activity of Drosophila melanogaster. Am J Plant Sci. 2012;3(7A):1037–40. https://pubmed.ncbi.nlm.nih.gov/26167392/
Chattopadhyay D, Thirumurugan K. Longevity promoting efficacies of different plant extracts in lower model organisms. Mech Ageing Dev. 2018;171:47–57. https://pubmed.ncbi.nlm.nih.gov/29526449/
Vayndorf EM, Lee SS, Liu RH. Whole apple extracts increase lifespan, healthspan and resistance to stress in Caenorhabditis elegans. J Funct Foods. 2013;5(3):1236–43. https://pubmed.ncbi.nlm.nih.gov/23878618/
Chattopadhyay D, Thirumurugan K. Longevity promoting efficacies of different plant extracts in lower model organisms. Mech Ageing Dev. 2018;171:47–57. https://pubmed.ncbi.nlm.nih.gov/29526449/
Dakik P, Rodriguez MEL, Junio JAB, et al. Discovery of fifteen new geroprotective plant extracts and identification of cellular processes they affect to prolong the chronological lifespan of budding yeast. Oncotarget. 2020;11(23):2182–203. https://pubmed.ncbi.nlm.nih.gov/32577164/
Chattopadhyay D, Thirumurugan K. Longevity promoting efficacies of different plant extracts in lower model organisms. Mech Ageing Dev. 2018;171:47–57. https://pubmed.ncbi.nlm.nih.gov/29526449/
Shimizu C, Wakita Y, Inoue T, et al. Effects of lifelong intake of lemon polyphenols on aging and intestinal microbiome in the senescence-accelerated mouse prone 1 (SAMP1). Sci Rep. 2019;9(1):3671. https://pubmed.ncbi.nlm.nih.gov/30842523/
Spindler SR, Mote PL, Flegal JM, Teter B. Influence on longevity of blueberry, cinnamon, green and black tea, pomegranate, sesame, curcumin, morin, pycnogenol, quercetin, and taxifolin fed iso-calorically to long-lived, F1 hybrid mice. Rejuvenation Res. 2013;16(2):143–51. https://pubmed.ncbi.nlm.nih.gov/23432089/
Kitani K, Osawa T, Yokozawa T. The effects of tetrahydrocurcumin and green tea polyphenol on the survival of male C57BL/6 mice. Biogerontology. 2007;8(5):567–73. https://pubmed.ncbi.nlm.nih.gov/17516143/
Spindler SR, Mote PL, Flegal JM, Teter B. Influence on longevity of blueberry, cinnamon, green and black tea, pomegranate, sesame, curcumin, morin, pycnogenol, quercetin, and taxifolin fed iso-calorically to long-lived, F1 hybrid mice. Rejuvenation Res. 2013;16(2):143–51. https://pubmed.ncbi.nlm.nih.gov/23432089/
Harris SB, Weindruch R, Smith GS, Mickey MR, and Walford RL. Dietary restriction alone and in combination with oral ethoxyquine/2-mercaptoethylamine in mice. J Gerontol. 1990;45(5):B141–7. https://pubmed.ncbi.nlm.nih.gov/2394907/
Aires DJ, Rockwell G, Wang T, et al. Potentiation of dietary restriction-induced lifespan extension by polyphenols. Biochim Biophys Acta. 2012;1822(4):522–6. https://pubmed.ncbi.nlm.nih.gov/22265987/
Brykman MC, Streusand Goldman V, Sarma N, Oketch-Rabah HA, Biswas D, Giancaspro GI. What should clinicians know about dietary supplement quality? AMA J Ethics. 2022;24(5):E382–9. https://pubmed.ncbi.nlm.nih.gov/35575569/
Margina D, Ilie M, Gradinaru D, Androutsopoulos VP, Kouretas D, Tsatsakis AM. Natural products – friends or foes? Toxicol Lett. 2015;236(3):154–67. https://pubmed.ncbi.nlm.nih.gov/25980574/
Ames BN. Prolonging healthy aging: longevity vitamins and proteins. Proc Natl Acad Sci U S A. 2018;115(43):10836–44. https://pubmed.ncbi.nlm.nih.gov/30322941/
Uysal U, Seremet S, Lamping JW, et al. Consumption of polyphenol plants may slow aging and associated diseases. Curr Pharm Des. 2013;19(34):6094–111. https://pubmed.ncbi.nlm.nih.gov/23448445/
Nakatani Y, Yaguchi Y, Komura T, et al. Sesamin extends lifespan through pathways related to dietary restriction in Caenorhabditis elegans. Eur J Nutr. 2018;57(3):1137–46. https://pubmed.ncbi.nlm.nih.gov/28239780/
Zuo Y, Peng C, Liang Y, et al. Sesamin extends the mean lifespan of fruit flies. Biogerontology. 2013;14(2):107–19. https://pubmed.ncbi.nlm.nih.gov/23291977/
Wichitsranoi J, Weerapreeyakul N, Boonsiri P, et al. Antihypertensive and antioxidant effects of dietary black sesame meal in pre-hypertensive humans. Nutr J. 2011;10:82. https://pubmed.ncbi.nlm.nih.gov/21827664/
Zhou L, Lin X, Abbasi AM, Zheng B. Phytochemical contents and antioxidant and antiproliferative activities of selected black and white sesame seeds. Biomed Res Int. 2016;2016:8495630. https://pubmed.ncbi.nlm.nih.gov/27597975/
Kim KS, Park SH. Anthrasesamone F from the seeds of black Sesamum indicum. Biosci Biotechnol Biochem. 2008;72(6):1626–7. https://pubmed.ncbi.nlm.nih.gov/18540089/
Lansky EP, Jiang W, Mo H, et al. Possible synergistic prostate cancer suppression by anatomically discrete pomegranate fractions. Invest New Drugs. 2005;23(1):11–20. https://pubmed.ncbi.nlm.nih.gov/15528976/
Seeram NP, Adams LS, Hardy ML, Heber D. Total cranberry extract versus its phytochemical constituents: antiproliferative and synergistic effects against human tumor cell lines. J Agric Food Chem. 2004;52(9):2512–7. https://pubmed.ncbi.nlm.nih.gov/15113149/
Brahmbhatt M, Gundala SR, Asif G, Shamsi SA, Aneja R. Ginger phytochemicals exhibit synergy to inhibit prostate cancer cell proliferation. Nutr Cancer. 2013;65(2):263–72. https://pubmed.ncbi.nlm.nih.gov/23441614/
Radhakrishnan S, Reddivari L, Sclafani R, Das UN, Vanamala J. Resveratrol potentiates grape seed extract induced human colon cancer cell apoptosis. Front Biosci (Elite Ed). 2011;3(4):1509–23. https://pubmed.ncbi.nlm.nih.gov/21622155/
Pérez-Sánchez A, Barrajón-Catalán E, Ruiz-Torres V, et al. Rosemary (Rosmarinus officinalis) extract causes ROS-induced necrotic cell death and inhibits tumor growth in vivo. Sci Rep. 2019;9(1):808. https://pubmed.ncbi.nlm.nih.gov/30692565/
Sporn MB, Liby KT. Is lycopene an effective agent for preventing prostate cancer? Cancer Prev Res (Phila). 2013;6(5):384–6. https://pubmed.ncbi.nlm.nih.gov/23483003/
Kumar NB, Besterman-Dahan K, Kang L, et al. Results of a randomized clinical trial of the action of several doses of lycopene in localized prostate cancer: administration prior to radical prostatectomy. Clin Med Urol. 2008;1:1–14. https://pubmed.ncbi.nlm.nih.gov/20354574/
Bowen P, Chen L, Stacewicz-Sapuntzakis M, et al. Tomato sauce supplementation and prostate cancer: lycopene accumulation and modulation of biomarkers of carcinogenesis. Exp Biol Med (Maywood). 2002;227(10):886–93. https://pubmed.ncbi.nlm.nih.gov/12424330/
Linnewiel-Hermoni K, Khanin M, Danilenko M, et al. The anti-cancer effects of carotenoids and other phytonutrients resides in their combined activity. Arch Biochem Biophys. 2015;572:28–35. https://pubmed.ncbi.nlm.nih.gov/25711533/
Talvas J, Caris-Veyrat C, Guy L, et al. Differential effects of lycopene consumed in tomato paste and lycopene in the form of a purified extract on target genes of cancer prostatic cells. Am J Clin Nutr. 2010;91(6):1716–24. https://pubmed.ncbi.nlm.nih.gov/20392890/
Warner M. Pandora’s Lunchbox: How Processed Food Took Over the American Meal. Simon & Schuster; 2014. https://worldcat.org/title/812258027
Liu RH. Potential synergy of phytochemicals in cancer prevention: mechanism of action. J Nutr. 2004;134(12 Suppl):3479S-85S. https://pubmed.ncbi.nlm.nih.gov/15570057/
Wang S, Meckling KA, Marcone MF, Kakuda Y, Tsao R. Synergistic, additive, and antagonistic effects of food mixtures on total antioxidant capacities. J Agric Food Chem. 2011;59(3):960–8. https://pubmed.ncbi.nlm.nih.gov/21222468/
Wang S, Meckling KA, Marcone MF, Kakuda Y, Tsao R. Synergistic, additive, and antagonistic effects of food mixtures on total antioxidant capacities. J Agric Food Chem. 2011;59(3):960–8. https://pubmed.ncbi.nlm.nih.gov/21222468/
Zhou JR, Yu L, Zhong Y, Blackburn GL. Soy phytochemicals and tea bioactive components synergistically inhibit androgen-sensitive human prostate tumors in mice. J Nutr. 2003;133(2):516–21. https://pubmed.ncbi.nlm.nih.gov/12566493/
Morré DJ, Morré DM. Synergistic Capsicum-tea mixtures with anticancer activity. J Pharm Pharmacol. 2003;55(7):987–94. https://pubmed.ncbi.nlm.nih.gov/12906756/
Ouhtit A, Gaur RL, Abdraboh M, et al. Simultaneous inhibition of cell-cycle, proliferation, survival, metastatic pathways and induction of apoptosis in breast cancer cells by a phytochemical super-cocktail: genes that underpin its mode of action. J Cancer. 2013;4(9):703–15. https://pubmed.ncbi.nlm.nih.gov/24312140/
Canene-Adams K, Lindshield BL, Wang S, Jeffery EH, Clinton SK, Erdman JW. Combinations of tomato and broccoli enhance antitumor activity in Dunning R3327-H prostate adenocarcinomas. Cancer Res. 2007;67(2):836–43. https://pubmed.ncbi.nlm.nih.gov/17213256/
.;
Boivin D, Lamy S, Lord-Dufour S, et al. Antiproliferative and antioxidant activities of common vegetables: a comparative study. Food Chem. 2009;112(2):374–80. https://www.sciencedirect.com/science/article/abs/pii/S0308814608006419
Yong LC, Petersen MR, Sigurdson AJ, Sampson LA, Ward EM. High dietary antioxidant intakes are associated with decreased chromosome translocation frequency in airline pilots. Am J Clin Nutr. 2009;90(5):1402–10. https://pubmed.ncbi.nlm.nih.gov/19793852/
Thompson HJ, Heimendinger J, Gillette C, et al. In vivo investigation of changes in biomarkers of oxidative stress induced by plant food rich diets. J Agric Food Chem. 2005;53(15):6126–32. https://pubmed.ncbi.nlm.nih.gov/16029006/
Thompson HJ, Heimendinger J, Diker A, et al. Dietary botanical diversity affects the reduction of oxidative biomarkers in women due to high vegetable and fruit intake. J Nutr. 2006;136(8):2207–12. https://pubmed.ncbi.nlm.nih.gov/16857842/
Bhupathiraju SN, Tucker KL. Greater variety in fruit and vegetable intake is associated with lower inflammation in Puerto Rican adults. Am J Clin Nutr. 2011;93(1):37–46. https://pubmed.ncbi.nlm.nih.gov/21068354/
Ye X, Bhupathiraju SN, Tucker KL. Variety in fruit and vegetable intake and cognitive function in middle-aged and older Puerto Rican adults. Br J Nutr. 2013;109(3):503–10. https://pubmed.ncbi.nlm.nih.gov/22717056/
Thomas R, Williams M, Sharma H, Chaudry A, Bellamy P. A double-blind, placebo-controlled randomised trial evaluating the effect of a polyphenol-rich whole food supplement on PSA progression in men with prostate cancer – the U.K. NCRN Pomi-T study. Prostate Cancer Prostatic Dis. 2014;17(2):180–6. https://pubmed.ncbi.nlm.nih.gov/24614693/
World Cancer Research Fund International. Diet, Nutrition, Physical Activity and Cancer: A Global Perspective: A Summary of the Third Expert Report. World Cancer Research Fund International; 2018. https://www.drugsandalcohol.ie/29052/
Ornish D, Weidner G, Fair WR, et al. Intensive lifestyle changes may affect the progression of prostate cancer. J Urol. 2005;174(3):1065–70. https://pubmed.ncbi.nlm.nih.gov/16094059/
Robinson VL. Rethinking the central dogma: noncoding RNAs are biologically relevant. Urol Oncol. 2009;27(3):304–6. https://pubmed.ncbi.nlm.nih.gov/19414118/
Robinson VL. Rethinking the central dogma: noncoding RNAs are biologically relevant. Urol Oncol. 2009;27(3):304–6. https://pubmed.ncbi.nlm.nih.gov/19414118/
McNeill EM, Hirschi KD. Roles of regulatory RNAs in nutritional control. Annu Rev Nutr. 2020;40:77–104. https://pubmed.ncbi.nlm.nih.gov/32966184/
Robinson VL. Rethinking the central dogma: noncoding RNAs are biologically relevant. Urol Oncol. 2009;27(3):304–6. https://pubmed.ncbi.nlm.nih.gov/19414118/
Ruvkun G. Glimpses of a tiny RNA world. Science. 2001;294(5543):797–9. https://pubmed.ncbi.nlm.nih.gov/11679654/
Roberts BM, Blewitt G, Dailey C, et al. Search for domain wall dark matter with atomic clocks on board global positioning system satellites. Nat Commun. 2017;8(1):1195. https://pubmed.ncbi.nlm.nih.gov/29084959/
Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of novel genes coding for small expressed RNAs. Science. 2001;294(5543):853–8. https://pubmed.ncbi.nlm.nih.gov/11679670/
Kalarikkal SP, Sundaram GM. Inter-kingdom regulation of human transcriptome by dietary microRNAs: emerging bioactives from edible plants to treat human diseases? Trends Food Sci Technol. 2021;118:723–34. https://www.sciencedirect.com/science/article/abs/pii/S0924224421005999
Robinson VL. Rethinking the central dogma: noncoding RNAs are biologically relevant. Urol Oncol. 2009;27(3):304–6. https://pubmed.ncbi.nlm.nih.gov/19414118/
Piovesan A, Caracausi M, Antonaros F, Pelleri MC, Vitale L. GeneBase 1.1: a tool to summarize data from NCBI gene datasets and its application to an update of human gene statistics. Database (Oxford). 2016;2016:baw153. https://pubmed.ncbi.nlm.nih.gov/28025344/
Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–54. https://pubmed.ncbi.nlm.nih.gov/8252621/
Díez-Sainz E, Lorente-Cebrián S, Aranaz P, Riezu-Boj JI, Martínez JA, Milagro FI. Potential mechanisms linking food-derived microRNAs, gut microbiota and intestinal barrier functions in the context of nutrition and human health. Front Nutr. 2021;8:586564. https://pubmed.ncbi.nlm.nih.gov/33768107/
Tarallo S, Pardini B, Mancuso G, et al. MicroRNA expression in relation to different dietary habits: a comparison in stool and plasma samples. Mutagenesis. 2014;29(5):385–91. https://pubmed.ncbi.nlm.nih.gov/25150024/
Majidinia M, Karimian A, Alemi F, Yousefi B, Safa A. Targeting miRNAs by polyphenols: novel therapeutic strategy for aging. Biochem Pharmacol. 2020;173:113688. https://pubmed.ncbi.nlm.nih.gov/31682793/
McNeill EM, Hirschi KD. Roles of regulatory RNAs in nutritional control. Annu Rev Nutr. 2020;40:77–104. https://pubmed.ncbi.nlm.nih.gov/32966184/
Cong L, Zhao Y, Pogue AI, Lukiw WJ. Role of microRNA (miRNA) and viroids in lethal diseases of plants and animals. Potential contribution to human neurodegenerative disorders. Biochemistry Moscow. 2018;83(9):1018–29. https://pubmed.ncbi.nlm.nih.gov/30472940/
McNeill EM, Hirschi KD. Roles of regulatory RNAs in nutritional control. Annu Rev Nutr. 2020;40:77–104. https://pubmed.ncbi.nlm.nih.gov/32966184/
Weber JA, Baxter DH, Zhang S, et al. The microRNA spectrum in 12 body fluids. Clin Chem. 2010;56(11):1733–41. https://pubmed.ncbi.nlm.nih.gov/20847327/
Alshehri B. Plant-derived xenomiRs and cancer: cross-kingdom gene regulation. Saudi J Biol Sci. 2021;28(4):2408–22. https://pubmed.ncbi.nlm.nih.gov/33911956/
Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–9. https://pubmed.ncbi.nlm.nih.gov/17486113/
Cammarata G, Duro G, Chiara TD, Curto AL, Taverna S, Candore G. Circulating miRNAs in successful and unsuccessful aging. A mini-review. Curr Pharm Des. 2019;25(39):4150–3. https://pubmed.ncbi.nlm.nih.gov/31742494/
Wang L, Sadri M, Giraud D, Zempleni J. RNase H2-dependent polymerase chain reaction and elimination of confounders in sample collection, storage, and analysis strengthen evidence that microRNAs in bovine milk are bioavailable in humans. J Nutr. 2018;148(1):153–9. https://pubmed.ncbi.nlm.nih.gov/29378054/
Bernstein E, Kim SY, Carmell MA, et al. Dicer is essential for mouse development. Nat Genet. 2003;35(3):215–7. https://pubmed.ncbi.nlm.nih.gov/14528307/
Kalarikkal SP, Sundaram GM. Inter-kingdom regulation of human transcriptome by dietary microRNAs: emerging bioactives from edible plants to treat human diseases? Trends Food Sci Technol. 2021;118:723–34. https://www.sciencedirect.com/science/article/abs/pii/S0924224421005999
Cammarata G, Duro G, Chiara TD, Curto AL, Taverna S, Candore G. Circulating miRNAs in successful and unsuccessful aging. A mini-review. Curr Pharm Des. 2019;25(39):4150–3. https://pubmed.ncbi.nlm.nih.gov/31742494/
Majidinia M, Mir SM, Mirza-Aghazadeh-Attari M, et al. MicroRNAs, DNA damage response and ageing. Biogerontology. 2020;21(3):275–91. https://pubmed.ncbi.nlm.nih.gov/32067137/
Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–54. https://pubmed.ncbi.nlm.nih.gov/8252621/
Boehm M, Slack F. A developmental timing microRNA and its target regulate life span in C. elegans. Science. 2005;310(5756):1954–7. https://pubmed.ncbi.nlm.nih.gov/16373574/
Morris BJ, Willcox DC, Donlon TA, Willcox BJ. BFOXO3: a major gene for human longevity – a mini-review. Gerontology. 2015;61(6):515–25. https://pubmed.ncbi.nlm.nih.gov/25832544/
Calissi G, Lam EWF, Link W. Therapeutic strategies targeting FOXO transcription factors. Nat Rev Drug Discov. 2021;20(1):21–38. https://pubmed.ncbi.nlm.nih.gov/33173189/
Pincus Z, Smith-Vikos T, Slack FJ. MicroRNA predictors of longevity in Caenorhabditis elegans. PLoS Genet. 2011;7(9):e1002306. https://pubmed.ncbi.nlm.nih.gov/21980307/
Green CD, Huang Y, Dou X, Yang L, Liu Y, Han JDJ. Impact of dietary interventions on noncoding RNA networks and mRNAs encoding chromatin-related factors. Cell Rep. 2017;18(12):2957–68. https://pubmed.ncbi.nlm.nih.gov/28329687/
Du WW, Yang W, Fang L, et al. miR-17 extends mouse lifespan by inhibiting senescence signaling mediated by MKP7. Cell Death Dis. 2014;5(7):e1355. https://pubmed.ncbi.nlm.nih.gov/25077541/
Cammarata G, Duro G, Chiara TD, Curto AL, Taverna S, Candore G. Circulating miRNAs in successful and unsuccessful aging. A mini-review. Curr Pharm Des. 2019;25(39):4150–3. https://pubmed.ncbi.nlm.nih.gov/31742494/
Smith-Vikos T, Liu Z, Parsons C, et al. A serum miRNA profile of human longevity: findings from the Baltimore Longitudinal Study of Aging (BLSA). Aging (Albany NY). 2016;8(11):2971–87. https://pubmed.ncbi.nlm.nih.gov/27824314/
Kumar P, Dezso Z, MacKenzie C, et al. Circulating miRNA biomarkers for Alzheimer’s disease. PLoS One. 2013;8(7):e69807. https://pubmed.ncbi.nlm.nih.gov/23922807/
Wu S, Kim TK, Wu X, et al. Circulating microRNAs and life expectancy among identical twins. Ann Hum Genet. 2016;80(5):247–56. https://pubmed.ncbi.nlm.nih.gov/27402348/
Kalarikkal SP, Sundaram GM. Inter-kingdom regulation of human transcriptome by dietary microRNAs: emerging bioactives from edible plants to treat human diseases? Trends Food Sci Technol. 2021;118:723–34. https://www.sciencedirect.com/science/article/abs/pii/S0924224421005999
Dávalos A, Pinilla L, López de Las Hazas MC, et al. Dietary microRNAs and cancer: a new therapeutic approach? Semin Cancer Biol. 2021;73:19–29. https://pubmed.ncbi.nlm.nih.gov/33086083/
Gkotzamanis V, Magriplis E, Panagiotakos D. The effect of physical activity interventions on cognitive function of older adults: a systematic review of clinical trials. Psychiatriki. Published online February 21, 2022.; https://pubmed.ncbi.nlm.nih.gov/35255465/
Pisani S, Mueller C, Huntley J, Aarsland D, Kempton MJ. A meta-analysis of randomised controlled trials of physical activity in people with Alzheimer’s disease and mild cognitive impairment with a comparison to donepezil. Int J Geriatr Psychiatry. 2021;36(10):1471–87. https://pubmed.ncbi.nlm.nih.gov/34490652/
Wang Y, Veremeyko T, Wong AHK, et al. Downregulation of miR-132/212 impairs S-nitrosylation balance and induces tau phosphorylation in Alzheimer’s disease. Neurobiol Aging. 2017;51:156–66. https://pubmed.ncbi.nlm.nih.gov/28089352/
Lugli G, Cohen AM, Bennett DA, et al. Plasma exosomal miRNAs in persons with and without Alzheimer disease: altered expression and prospects for biomarkers. PloS One. 2015;10(10):e0139233. https://pubmed.ncbi.nlm.nih.gov/26426747/
Radom-Aizik S, Zaldivar F, Leu S, Adams GR, Oliver S, Cooper DM. Effects of exercise on microRNA expression in young males peripheral blood mononuclear cells. Clin Transl Sci. 2012;5(1):32–8. https://pubmed.ncbi.nlm.nih.gov/22376254/
Nielsen S, Åkerström T, Rinnov A, et al. The miRNA plasma signature in response to acute aerobic exercise and endurance training. PLoS One. 2014;9(2):e87308. https://pubmed.ncbi.nlm.nih.gov/24586268/
Fernández-de Frutos M, Galán-Chilet I, Goedeke L, et al. MicroRNA 7 impairs insulin signaling and regulates aß levels through posttranscriptional regulation of the insulin receptor substrate 2, insulin receptor, insulin-degrading enzyme, and liver X receptor pathway. Mol Cell Biol. 2019;39(22):e00170–19. https://pubmed.ncbi.nlm.nih.gov/31501273/
Denk J, Boelmans K, Siegismund C, Lassner D, Arlt S, Jahn H. MicroRNA profiling of CSF reveals potential biomarkers to detect Alzheimer’s disease. PloS One. 2015;10(5):e0126423. https://pubmed.ncbi.nlm.nih.gov/25992776/
Nielsen S, Åkerström T, Rinnov A, et al. The miRNA plasma signature in response to acute aerobic exercise and endurance training. PloS One. 2014;9(2):e87308. https://pubmed.ncbi.nlm.nih.gov/24586268/
Barber JL, Zellars KN, Barringhaus KG, Bouchard C, Spinale FG, Sarzynski MA. The effects of regular exercise on circulating cardiovascular-related microRNAs. Sci Rep. 2019;9(1):7527. https://pubmed.ncbi.nlm.nih.gov/31101833/
Wu Y, Xu J, Xu J, et al. Lower serum levels of miR-29c-3p and miR-19b-3p as biomarkers for Alzheimer’s disease. Tohoku J Exp Med. 2017;242(2):129–36. https://pubmed.ncbi.nlm.nih.gov/28626163/
Arena A, Iyer AM, Milenkovic I, et al. Developmental expression and dysregulation of miR-146a and miR-155 in Down’s syndrome and mouse models of Down’s syndrome and Alzheimer’s disease. Curr Alzheimer Res. 2017;14(12):1305–17. https://pubmed.ncbi.nlm.nih.gov/28720071/
Alexandrov PN, Dua P, Hill JM, Bhattacharjee S, Zhao Y, Lukiw WJ. MicroRNA (miRNA) speciation in Alzheimer’s disease (AD) cerebrospinal fluid (CSF) and extracellular fluid (ECF). Int J Biochem Mol Biol. 2012;3(4):365–73. https://pubmed.ncbi.nlm.nih.gov/23301201/
Sawada S, Kon M, Wada S, Ushida T, Suzuki K, Akimoto T. Profiling of circulating microRNAs after a bout of acute resistance exercise in humans. PLoS One. 2013;8(7):e70823. https://pubmed.ncbi.nlm.nih.gov/23923026/
Li Y, Yao M, Zhou Q, et al. Dynamic regulation of circulating microRNAs during acute exercise and long-term exercise training in basketball athletes. Front Physiol. 2018;9:282. https://pubmed.ncbi.nlm.nih.gov/29662456/
Baggish AL, Hale A, Weiner RB, et al. Dynamic regulation of circulating microRNA during acute exhaustive exercise and sustained aerobic exercise training. J Physiol. 2011;589(Pt 16):3983–94. https://pubmed.ncbi.nlm.nih.gov/21690193/
Baggish AL, Park J, Min PK, et al. Rapid upregulation and clearance of distinct circulating microRNAs after prolonged aerobic exercise. J Appl Physiol (1985). 2014;116(5):522–31. https://pubmed.ncbi.nlm.nih.gov/24436293/
Improta-Caria AC, Nonaka CKV, Cavalcante BRR, De Sousa RAL, Aras Júnior R, Souza BS de F. Modulation of microRNAs as a potential molecular mechanism involved in the beneficial actions of physical exercise in Alzheimer disease. Int J Mol Sci. 2020;21(14):E4977. https://pubmed.ncbi.nlm.nih.gov/32674523/
Majidinia M, Karimian A, Alemi F, Yousefi B, Safa A. Targeting miRNAs by polyphenols: novel therapeutic strategy for aging. Biochem Pharmacol. 2020;173:113688. https://pubmed.ncbi.nlm.nih.gov/31682793/
García-Segura L, Pérez-Andrade M, Miranda-Ríos J. The emerging role of microRNAs in the regulation of gene expression by nutrients. J Nutrigenet Nutrigenomics. 2013;6(1):16–31. https://pubmed.ncbi.nlm.nih.gov/23445777/
Daimiel L, Micó V, Valls RM, et al. Impact of phenol-enriched virgin olive oils on the postprandial levels of circulating microRNAs related to cardiovascular disease. Mol Nutr Food Res. 2020;64(15):2000049. https://pubmed.ncbi.nlm.nih.gov/32562310/
López de Las Hazas MC, Gil-Zamorano J, Cofán M, et al. One-year dietary supplementation with walnuts modifies exosomal miRNA in elderly subjects. Eur J Nutr. 2021;60(4):1999–2011. https://pubmed.ncbi.nlm.nih.gov/32979076/
Ortega FJ, Cardona-Alvarado MI, Mercader JM, et al. Circulating profiling reveals the effect of a polyunsaturated fatty acid – enriched diet on common microRNAs. J Nutr Biochem. 2015;26(10):1095–101. https://pubmed.ncbi.nlm.nih.gov/26092372/
Shao D, Lian Z, Di Y, et al. Dietary compounds have potential in controlling atherosclerosis by modulating macrophage cholesterol metabolism and inflammation via miRNA. NPJ Sci Food. 2018;2:13. https://pubmed.ncbi.nlm.nih.gov/31304263/
Koolivand M, Ansari M, Piroozian F, Moein S, MalekZadeh K. Alleviating the progression of acute myeloid leukemia (AML) by sulforaphane through controlling miR-155 levels. Mol Biol Rep. 2018;45(6):2491–9. https://pubmed.ncbi.nlm.nih.gov/30350234/
Singh S, Raza W, Parveen S, Meena A, Luqman S. Flavonoid display ability to target microRNAs in cancer pathogenesis. Biochem Pharmacol. 2021;189:114409. https://pubmed.ncbi.nlm.nih.gov/33428895/
Guo X, Cai Q, Bao P, et al. Long-term soy consumption and tumor tissue microRNA and gene expression in triple negative breast cancer. Cancer. 2016;122(16):2544–51. https://pubmed.ncbi.nlm.nih.gov/27183356/
Boutas I, Kontogeorgi A, Dimitrakakis C, Kalantaridou SN. Soy isoflavones and breast cancer risk: a meta-analysis. In Vivo. 2022;36(2):556–62. https://pubmed.ncbi.nlm.nih.gov/35241506/
Qiu S, Jiang C. Soy and isoflavones consumption and breast cancer survival and recurrence: a systematic review and meta-analysis. Eur J Nutr. 2019;58(8):3079–90. https://pubmed.ncbi.nlm.nih.gov/30382332/
Tarallo S, Ferrero G, De Filippis F, et al. Stool microRNA profiles reflect different dietary and gut microbiome patterns in healthy individuals. Gut. 2022;71(7):1302–14. https://pubmed.ncbi.nlm.nih.gov/34315772/
Dinu M, Abbate R, Gensini GF, Casini A, Sofi F. Vegetarian, vegan diets and multiple health outcomes: a systematic review with meta-analysis of observational studies. Crit Rev Food Sci Nutr. 2017;57(17):3640–9. https://pubmed.ncbi.nlm.nih.gov/26853923/
Humphreys KJ, Conlon MA, Young GP, et al. Dietary manipulation of oncogenic microRNA expression in human rectal mucosa: a randomized trial. Cancer Prev Res (Phila). 2014;7(8):786–95. https://pubmed.ncbi.nlm.nih.gov/25092886/
Papaioannou MD, Koufaris C, Gooderham NJ. The cooked meat-derived mammary carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) elicits estrogenic-like microRNA responses in breast cancer cells. Toxicol Lett. 2014;229(1):9–16. https://pubmed.ncbi.nlm.nih.gov/24877718/
Yang WM, Jeong HJ, Park SY, Lee W. Induction of miR-29a by saturated fatty acids impairs insulin signaling and glucose uptake through translational repression of IRS-1 in myocytes. FEBS Lett. 2014;588(13):2170–6. https://pubmed.ncbi.nlm.nih.gov/24844433/
Dinu M, Abbate R, Gensini GF, Casini A, Sofi F. Vegetarian, vegan diets and multiple health outcomes: a systematic review with meta-analysis of observational studies. Crit Rev Food Sci Nutr. 2017;57(17):3640–9. https://pubmed.ncbi.nlm.nih.gov/26853923/
Sinha R, Rothman N, Brown ED, et al. High concentrations of the carcinogen 2-amino-1-methyl-6-phynylimidazo-[4,5-b]pyridine (PhIP) occur in chicken but are dependent on the cooking method. Cancer Res. 1995;55(20):4516–9. https://pubmed.ncbi.nlm.nih.gov/7553619/
Liu T, Gatto NM, Chen Z, et al. Vegetarian diets, circulating miRNA expression and healthspan in subjects living in the Blue Zone. Precis Clin Med. 2020;3(4):245–59. https://pubmed.ncbi.nlm.nih.gov/33391847/
Von Linné, C. Salvius L. Caroli Linnæi… systema naturæ per regna tria naturæ, secundum classes, ordines, genera, species, cum characteribus differentiis, synonymis, locis. Impensis Direct. Laurentii Salvii; 1758. https://www.biodiversitylibrary.org/bibliography/542
Haeckel E. Generelle morphologie der organismen. Allgemeine grundzüge der organischen formen-wissenschaft, mechanisch begründet durch die von Charles Darwin reformirte descendenztheorie. G. Reimer; 1866. https://www.biodiversitylibrary.org/bibliography/3953
Ruggiero MA, Gordon DP, Orrell TM, et al. A higher level classification of all living organisms. PLoS One. 2015;10(4):e0119248. https://pubmed.ncbi.nlm.nih.gov/25923521/
Dalmasso G, Nguyen HTT, Yan Y, et al. Microbiota modulate host gene expression via microRNAs. PLoS One. 2011;6(4):e19293. https://pubmed.ncbi.nlm.nih.gov/21559394/
Choi JW, Kim SC, Hong SH, Lee HJ. Secretable small RNAs via outer membrane vesicles in periodontal pathogens. J Dent Res. 2017;96(4):458–66. https://pubmed.ncbi.nlm.nih.gov/28068479/
Liu S, da Cunha AP, Rezende RM, et al. The host shapes the gut microbiota via fecal microRNA. Cell Host Microbe. 2016;19(1):32–43. https://pubmed.ncbi.nlm.nih.gov/26764595/
Munroe R. Family reunion. xkcd. https://xkcd.com/2608/. Accessed October 17, 2022.; https://xkcd.com/2608/
Wang DY, Kumar S, Hedges SB. Divergence time estimates for the early history of animal phyla and the origin of plants, animals and fungi. Proc Biol Sci. 1999;266(1415):163–71. https://pubmed.ncbi.nlm.nih.gov/10097391/
Zhao Y, Cong L, Lukiw WJ. Plant and animal microRNAs (miRNAs) and their potential for inter-kingdom communication. Cell Mol Neurobiol. 2018;38(1):133–40. https://pubmed.ncbi.nlm.nih.gov/28879580/
Zhang T, Jin Y, Zhao JH, et al. Host-induced gene silencing of the target gene in fungal cells confers effective resistance to the cotton wilt disease pathogen Verticillium dahliae. Mol Plant. 2016;9(6):939–42. https://pubmed.ncbi.nlm.nih.gov/26925819/
Cong L, Zhao Y, Pogue AI, Lukiw WJ. Role of microRNA (miRNA) and viroids in lethal diseases of plants and animals. Potential contribution to human neurodegenerative disorders. Biochemistry Moscow. 2018;83(9):1018–29. https://pubmed.ncbi.nlm.nih.gov/30472940/
McNeill EM, Hirschi KD. Roles of regulatory RNAs in nutritional control. Annu Rev Nutr. 2020;40:77–104. https://pubmed.ncbi.nlm.nih.gov/32966184/
Jia M, He J, Bai W, et al. Cross-kingdom regulation by dietary plant miRNAs: an evidence-based review with recent updates. Food Funct. 2021;12(20):9549–62. https://pubmed.ncbi.nlm.nih.gov/34664582/
Li Z, Xu R, Li N. MicroRNAs from plants to animals, do they define a new messenger for communication? Nutr Metab (Lond). 2018;15:68. https://pubmed.ncbi.nlm.nih.gov/30302122/
Cong L, Zhao Y, Pogue AI, Lukiw WJ. Role of microRNA (miRNA) and viroids in lethal diseases of plants and animals. Potential contribution to human neurodegenerative disorders. Biochemistry Moscow. 2018;83(9):1018–29. https://pubmed.ncbi.nlm.nih.gov/30472940/
Díez-Sainz E, Lorente-Cebrián S, Aranaz P, Riezu-Boj JI, Martínez JA, Milagro FI. Potential mechanisms linking food-derived microRNAs, gut microbiota and intestinal barrier functions in the context of nutrition and human health. Front Nutr. 2021;8:586564. https://pubmed.ncbi.nlm.nih.gov/33768107/
Zhao JH, Zhang T, Liu QY, Guo HS. Trans-kingdom RNAs and their fates in recipient cells: advances, utilization, and perspectives. Plant Commun. 2021;2(2):100167. https://pubmed.ncbi.nlm.nih.gov/33898979/
García-Segura L, Pérez-Andrade M, Miranda-Ríos J. The emerging role of microRNAs in the regulation of gene expression by nutrients. J Nutrigenet Nutrigenomics. 2013;6(1):16–31. https://pubmed.ncbi.nlm.nih.gov/23445777/
Campbell K. The doubts about dietary RNA. Nature. 2020;582:s10–1. https://www.nature.com/articles/d41586-020-01767-x
Zhu WJ, Liu Y, Cao YN, Peng LX, Yan ZY, Zhao G. Insights into health-promoting effects of plant microRNAs: a review. J Agric Food Chem. 2021;69(48):14372–86. https://pubmed.ncbi.nlm.nih.gov/34813309/
Li M, Chen T, He JJ, et al. Plant MIR167e-5p inhibits enterocyte proliferation by targeting ß-catenin. Cells. 2019;8(11):1385. https://pubmed.ncbi.nlm.nih.gov/31689969/
del Pozo-Acebo L, López de las Hazas M, Margollés A, Dávalos A, García-Ruiz A. Eating microRNAs: pharmacological opportunities for cross-kingdom regulation and implications in host gene and gut microbiota modulation. British J Pharmacology. 2021;178(11):2218–45. https://pubmed.ncbi.nlm.nih.gov/33644849/
Dávalos A, Pinilla L, López de Las Hazas MC, et al. Dietary microRNAs and cancer: a new therapeutic approach? Semin Cancer Biol. 2021;73:19–29. https://pubmed.ncbi.nlm.nih.gov/33086083/
Chin AR, Fong MY, Somlo G, et al. Cross-kingdom inhibition of breast cancer growth by plant miR159. Cell Res. 2016;26(2):217–28. https://pubmed.ncbi.nlm.nih.gov/26794868/
Cavallini A, Minervini F, Garbetta A, et al. High degradation and no bioavailability of artichoke miRNAs assessed using an in vitro digestion/Caco-2 cell model. Nutr Res. 2018;60:68–76. https://pubmed.ncbi.nlm.nih.gov/30527261/
Philip A, Ferro VA, Tate RJ. Determination of the potential bioavailability of plant microRNAs using a simulated human digestion process. Mol Nutr Food Res. 2015;59(10):1962–72. https://pubmed.ncbi.nlm.nih.gov/26147655/
Link J, Thon C, Schanze D, et al. Food-derived xeno-microRNAs: influence of diet and detectability in gastrointestinal tract – proof-of-principle study. Mol Nutr Food Res. 2019;63(2):e1800076. https://pubmed.ncbi.nlm.nih.gov/30378765/
Snow JW, Hale AE, Isaacs SK, Baggish AL, Chan SY. Ineffective delivery of diet-derived microRNAs to recipient animal organisms. RNA Biol. 2013;10(7):1107–16. https://pubmed.ncbi.nlm.nih.gov/23669076/
Link J, Thon C, Schanze D, et al. Food-derived xeno-microRNAs: influence of diet and detectability in gastrointestinal tract – proof-of-principle study. Mol Nutr Food Res. 2019;63(2):e1800076. https://pubmed.ncbi.nlm.nih.gov/30378765/
Kalarikkal SP, Sundaram GM. Inter-kingdom regulation of human transcriptome by dietary microRNAs: emerging bioactives from edible plants to treat human diseases? Trends Food Sci Technol. 2021;118:723–34. https://www.sciencedirect.com/science/article/abs/pii/S0924224421005999
Zhang L, Hou D, Chen X, et al. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: evidence of cross-kingdom regulation by microRNA. Cell Res. 2012;22(1):107–26. https://pubmed.ncbi.nlm.nih.gov/21931358/
Chen Q, Zhang F, Dong L, et al. SIDT1-dependent absorption in the stomach mediates host uptake of dietary and orally administered microRNAs. Cell Res. 2021;31(3):247–58. https://pubmed.ncbi.nlm.nih.gov/32801357/
Kalarikkal SP, Sundaram GM. Inter-kingdom regulation of human transcriptome by dietary microRNAs: emerging bioactives from edible plants to treat human diseases? Trends Food Sci Technol. 2021;118:723–34. https://www.sciencedirect.com/science/article/abs/pii/S0924224421005999
Wang Q, Zhuang X, Mu J, et al. Delivery of therapeutic agents by nanoparticles made of grapefruit-derived lipids. Nat Commun. 2013;4:1867. https://pubmed.ncbi.nlm.nih.gov/23695661/
Kalarikkal SP, Sundaram GM. Inter-kingdom regulation of human transcriptome by dietary microRNAs: emerging bioactives from edible plants to treat human diseases? Trends Food Sci Technol. 2021;118:723–34. https://www.sciencedirect.com/science/article/abs/pii/S0924224421005999
Zhu WJ, Liu Y, Cao YN, Peng LX, Yan ZY, Zhao G. Insights into health-promoting effects of plant microRNAs: a review. J Agric Food Chem. 2021;69(48):14372–86. https://pubmed.ncbi.nlm.nih.gov/34813309/
Yu B, Yang Z, Li J, et al. Methylation as a crucial step in plant microRNA biogenesis. Science. 2005;307(5711):932–5. https://pubmed.ncbi.nlm.nih.gov/15705854/
Link J, Thon C, Schanze D, et al. Food-derived xeno-microRNAs: influence of diet and detectability in gastrointestinal tract – proof-of-principle study. Mol Nutr Food Res. 2019;63(2):e1800076. https://pubmed.ncbi.nlm.nih.gov/30378765/
Liang G, Zhu Y, Sun B, et al. Assessing the survival of exogenous plant microRNA in mice. Food Sci Nutr. 2014;2(4):380–8. https://pubmed.ncbi.nlm.nih.gov/25473495/
Luo Y, Wang P, Wang X, et al. Detection of dietetically absorbed maize-derived microRNAs in pigs. Sci Rep. 2017;7(1):645. https://pubmed.ncbi.nlm.nih.gov/28381865/
Chen Q, Zhang F, Dong L, et al. SIDT1-dependent absorption in the stomach mediates host uptake of dietary and orally administered microRNAs. Cell Res. 2021;31(3):247–58. https://pubmed.ncbi.nlm.nih.gov/32801357/
Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJA. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–5. https://pubmed.ncbi.nlm.nih.gov/21423189/
Liang G, Zhu Y, Sun B, et al. Assessing the survival of exogenous plant microRNA in mice. Food Sci Nutr. 2014;2(4):380–8. https://pubmed.ncbi.nlm.nih.gov/25473495/
Zhang L, Hou D, Chen X, et al. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: evidence of cross-kingdom regulation by microRNA. Cell Res. 2012;22(1):107–26. https://pubmed.ncbi.nlm.nih.gov/21931358/
Chen X, Liu L, Chu Q, et al. Large-scale identification of extracellular plant miRNAs in mammals implicates their dietary intake. PLoS One. 2021;16(9):e0257878. https://pubmed.ncbi.nlm.nih.gov/34587184/
Li J, Zhang Y, Li D, et al. Small non-coding RNAs transfer through mammalian placenta and directly regulate fetal gene expression. Protein Cell. 2015;6(6):391–6. https://pubmed.ncbi.nlm.nih.gov/25963995/
Xiao J, Feng S, Wang X, et al. Identification of exosome-like nanoparticle-derived microRNAs from 11 edible fruits and vegetables. PeerJ. 2018;6:e5186. https://pubmed.ncbi.nlm.nih.gov/30083436/
Ju S, Mu J, Dokland T, et al. Grape exosome-like nanoparticles induce intestinal stem cells and protect mice from DSS-induced colitis. Mol Ther. 2013;21(7):1345–57. https://pubmed.ncbi.nlm.nih.gov/23752315/
Mu J, Zhuang X, Wang Q, et al. Interspecies communication between plant and mouse gut host cells through edible plant derived exosome-like nanoparticles. Mol Nutr Food Res. 2014;58(7):1561–73. https://pubmed.ncbi.nlm.nih.gov/24842810/
Cavalieri D, Rizzetto L, Tocci N, et al. Plant microRNAs as novel immunomodulatory agents. Sci Rep. 2016;6:25761. https://pubmed.ncbi.nlm.nih.gov/27167363/
Hou D, He F, Ma L, et al. The potential atheroprotective role of plant MIR156a as a repressor of monocyte recruitment on inflamed human endothelial cells. J Nutr Biochem. 2018;57:197–205. https://pubmed.ncbi.nlm.nih.gov/29751293/
Ojagbemi A, Okekunle AP, Owolabi M, et al. Dietary intakes of green leafy vegetables and incidence of cardiovascular diseases. Cardiovasc J Afr. 2021;32(4):215–23. https://pubmed.ncbi.nlm.nih.gov/34128951/
Kalarikkal SP, Sundaram GM. Inter-kingdom regulation of human transcriptome by dietary microRNAs: emerging bioactives from edible plants to treat human diseases? Trends Food Sci Technol. 2021;118:723–34. https://www.sciencedirect.com/science/article/abs/pii/S0924224421005999
Ngo SNT, Williams DB. Protective effect of isothiocyanates from cruciferous vegetables on breast cancer: epidemiological and preclinical perspectives. Anticancer Agents Med Chem. 2021;21(11):1413–30. https://pubmed.ncbi.nlm.nih.gov/32972351/
Li D, Yang J, Yang Y, et al. A timely review of cross-kingdom regulation of plant-derived microRNAs. Front Genet. 2021;12:613197. https://pubmed.ncbi.nlm.nih.gov/34012461/
Xiang J, Huang JC, Xu C, et al. [Effect of miRNA from Glycyrrhiza uralensis decoction on gene expression of human immune cells]. Zhongguo Zhong Yao Za Zhi. 2017;42(9):1752–6. https://pubmed.ncbi.nlm.nih.gov/29082701/
Qin Y, Zheng B, Yang G shan, et al. Salvia miltiorrhiza-derived Sal-miR-58 induces autophagy and attenuates inflammation in vascular smooth muscle cells. Mol Ther Nucleic Acids. 2020;21:492–511. https://pubmed.ncbi.nlm.nih.gov/32679544/
Yang GS, Zheng B, Qin Y, et al. Salvia miltiorrhiza-derived miRNAs suppress vascular remodeling through regulating OTUD7B/KLF4/NMHC IIA axis. Theranostics. 2020;10(17):7787–811. https://pubmed.ncbi.nlm.nih.gov/32685020/
Zhou LK, Zhou Z, Jiang XM, et al. Absorbed plant MIR2911 in honeysuckle decoction inhibits SARS-CoV-2 replication and accelerates the negative conversion of infected patients. Cell Discov. 2020;6(1):1–4. https://pubmed.ncbi.nlm.nih.gov/32802404/
Avsar B, Zhao Y, Li W, Lukiw WJ. Atropa belladonna expresses a microRNA (aba-miRNA-9497) highly homologous to Homo sapiens miRNA-378 (hsa-miRNA-378); both miRNAs target the 3’-untranslated region (3’-UTR) of the mRNA encoding the neurologically relevant https://pubmed.ncbi.nlm.nih.gov/31456135/
McNeill EM, Hirschi KD. Roles of regulatory RNAs in nutritional control. Annu Rev Nutr. 2020;40:77–104. https://pubmed.ncbi.nlm.nih.gov/32966184/
Wang L, Sadri M, Giraud D, Zempleni J. RNase H2-dependent polymerase chain reaction and elimination of confounders in sample collection, storage, and analysis strengthen evidence that microRNAs in bovine milk are bioavailable in humans. J Nutr. 2018;148(1):153–9. https://pubmed.ncbi.nlm.nih.gov/29378054/
Chen X, Liu L, Chu Q, et al. Large-scale identification of extracellular plant miRNAs in mammals implicates their dietary intake. PLoS One. 2021;16(9):e0257878. https://pubmed.ncbi.nlm.nih.gov/34587184/
Igaz I, Igaz P. Hypothetic interindividual and interspecies relevance of microRNAs released in body fluids. Exp Suppl. 2015;106:281–8. https://pubmed.ncbi.nlm.nih.gov/26608210/
Witwer KW, Zhang CY. Diet-derived microRNAs: unicorn or silver bullet? Genes Nutr. 2017;12:15. https://pubmed.ncbi.nlm.nih.gov/28694875/
Sundaram GM. Dietary non-coding RNAs from plants: fairy tale or treasure? Noncoding RNA Res. 2019;4(2):63–8. https://pubmed.ncbi.nlm.nih.gov/31193509/
McNeill EM, Hirschi KD. Roles of regulatory RNAs in nutritional control. Annu Rev Nutr. 2020;40:77–104. https://pubmed.ncbi.nlm.nih.gov/32966184/
Link J, Thon C, Schanze D, et al. Food-derived xeno-microRNAs: influence of diet and detectability in gastrointestinal tract – proof-of-principle study. Mol Nutr Food Res. 2019;63(2):e1800076. https://pubmed.ncbi.nlm.nih.gov/30378765/
Quintanilha B, Reis B, Duarte G, Cozzolino S, Rogero M. Nutrimiromics: role of microRNAs and nutrition in modulating inflammation and chronic diseases. Nutrients. 2017;9(11):1168. https://pubmed.ncbi.nlm.nih.gov/29077020/
Mar-Aguilar F, Arreola-Triana A, Mata-Cardona D, Gonzalez-Villasana V, Rodríguez-Padilla C, Reséndez-Pérez D. Evidence of transfer of miRNAs from the diet to the blood still inconclusive. PeerJ. 2020;8:e9567 https://pubmed.ncbi.nlm.nih.gov/32995073/
Wang W, Hang C, Zhang Y, et al. Dietary miR-451 protects erythroid cells from oxidative stress via increasing the activity of Foxo3 pathway. Oncotarget. 2017;8(63):107109–24. https://pubmed.ncbi.nlm.nih.gov/29291015/
Teodori L, Petrignani I, Giuliani A, et al. Inflamm-aging microRNAs may integrate signals from food and gut microbiota by modulating common signalling pathways. Mech Ageing Dev. 2019;182:111127. https://pubmed.ncbi.nlm.nih.gov/31401225/
Mar-Aguilar F, Arreola-Triana A, Mata-Cardona D, Gonzalez-Villasana V, Rodríguez-Padilla C, Reséndez-Pérez D. Evidence of transfer of miRNAs from the diet to the blood still inconclusive. PeerJ. 2020;8:e9567. https://pubmed.ncbi.nlm.nih.gov/32995073/
Humphreys KJ, Conlon MA, Young GP, et al. Dietary manipulation of oncogenic microRNA expression in human rectal mucosa: a randomized trial. Cancer Prev Res (Phila). 2014;7(8):786–95. https://pubmed.ncbi.nlm.nih.gov/25092886/
Baier S, Howard K, Cui J, Shu J, Zempleni J. MicroRNAs in chicken eggs are bioavailable in healthy adults and can modulate mRNA expression in peripheral blood mononuclear cells. FASEB J. 2015;29(S1):LB322. https://pubmed.ncbi.nlm.nih.gov/25122645/
Igaz I, Igaz P. Hypothetic interindividual and interspecies relevance of microRNAs released in body fluids. Exp Suppl. 2015;106:281–8. https://pubmed.ncbi.nlm.nih.gov/26608210/
Melnik BC, Schmitz G. MicroRNAs: milk’s epigenetic regulators. Best Pract Res Clin Endocrinol Metab. 2017;31(4):427–42. https://pubmed.ncbi.nlm.nih.gov/29221571/
Benmoussa A, Provost P. Milk microRNAs in health and disease. Compr Rev Food Sci Food Saf. 2019;18(3):703–22. https://pubmed.ncbi.nlm.nih.gov/33336926/
Tooley KL, El-Merhibi A, Cummins AG, et al. Maternal milk, but not formula, regulates the immune response to ß-lactoglobulin in allergy-prone rat pups. J Nutr. 2009;139(11):2145–51. https://pubmed.ncbi.nlm.nih.gov/19759244/
Melnik BC, Stremmel W, Weiskirchen R, John SM, Schmitz G. Exosome-derived microRNAs of human milk and their effects on infant health and development. Biomolecules. 2021;11(6):851. https://pubmed.ncbi.nlm.nih.gov/34200323/
Melnik BC, Stremmel W, Weiskirchen R, John SM, Schmitz G. Exosome-derived microRNAs of human milk and their effects on infant health and development. Biomolecules. 2021;11(6):851. https://pubmed.ncbi.nlm.nih.gov/34200323/
Melnik BC, Schmitz G. MicroRNAs: milk’s epigenetic regulators. Best Pract Res Clin Endocrinol Metab. 2017;31(4):427–42. https://pubmed.ncbi.nlm.nih.gov/29221571/
Melnik BC. Lifetime impact of cow’s milk on overactivation of mTORC1: from fetal to childhood overgrowth, acne, diabetes, cancers, and neurodegeneration. Biomolecules. 2021;11(3):404. https://pubmed.ncbi.nlm.nih.gov/33803410/
Melnik BC, Schmitz G. Exosomes of pasteurized milk: potential pathogens of Western diseases. J Transl Med. 2019;17(1):3. https://pubmed.ncbi.nlm.nih.gov/30602375/
Victora CG, Bahl R, Barros AJD, et al. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet. 2016;387(10017):475–90. https://pubmed.ncbi.nlm.nih.gov/26869575/
Chen Z, Xie Y, Luo J, et al. Milk exosome-derived miRNAs from water buffalo are implicated in immune response and metabolism process. BMC Vet Res. 2020;16(1):123. https://pubmed.ncbi.nlm.nih.gov/32349776/
Baier SR, Nguyen C, Xie F, Wood JR, Zempleni J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J Nutr. 2014;144(10):1495–500. https://pubmed.ncbi.nlm.nih.gov/25122645/
McNeill EM, Hirschi KD. Roles of regulatory RNAs in nutritional control. Annu Rev Nutr. 2020;40:77–104. https://pubmed.ncbi.nlm.nih.gov/32966184/
Melnik BC, Schmitz G. Exosomes of pasteurized milk: potential pathogens of Western diseases. J Transl Med. 2019;17(1):3. https://pubmed.ncbi.nlm.nih.gov/30602375/
Benmoussa A, Lee CHC, Laffont B, et al. Commercial dairy cow milk microRNAs resist digestion under simulated gastrointestinal tract conditions. J Nutr. 2016;146(11):2206–15. https://pubmed.ncbi.nlm.nih.gov/27708120/
López de Las Hazas MC, Del Pozo-Acebo L, Hansen MS, et al. Dietary bovine milk miRNAs transported in extracellular vesicles are partially stable during GI digestion, are bioavailable and reach target tissues but need a minimum dose to impact on gene expression. Eur J Nutr. 2022;61(2):1043–56. https://pubmed.ncbi.nlm.nih.gov/34716465/
Baier SR, Nguyen C, Xie F, Wood JR, Zempleni J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J Nutr. 2014;144(10):1495–500. https://pubmed.ncbi.nlm.nih.gov/25122645/
Wang L, Sadri M, Giraud D, Zempleni J. RNase H2-dependent polymerase chain reaction and elimination of confounders in sample collection, storage, and analysis strengthen evidence that microRNAs in bovine milk are bioavailable in humans. J Nutr. 2018;148(1):153–9. https://pubmed.ncbi.nlm.nih.gov/29378054/
Melnik BC, Schmitz G. Exosomes of pasteurized milk: potential pathogens of Western diseases. J Transl Med. 2019;17(1):3. https://pubmed.ncbi.nlm.nih.gov/30602375/
Melnik BC. Milk exosomal miRNAs: potential drivers of AMPK-to-mTORC1 switching in ß-cell de-differentiation of type 2 diabetes mellitus. Nutr Metab (Lond). 2019;16:85. https://pubmed.ncbi.nlm.nih.gov/31827573/
Melnik BC. Synergistic effects of milk-derived exosomes and galactose on a-synuclein pathology in Parkinson’s disease and type 2 diabetes mellitus. Int J Mol Sci. 2021;22(3):1059. https://pubmed.ncbi.nlm.nih.gov/33494388/
Melnik BC, Schmitz G. MicroRNAs: milk’s epigenetic regulators. Best Pract Res Clin Endocrinol Metab. 2017;31(4):427–42. https://pubmed.ncbi.nlm.nih.gov/29221571/
Melnik BC, Schmitz G. Exosomes of pasteurized milk: potential pathogens of Western diseases. J Transl Med. 2019;17(1):3. https://pubmed.ncbi.nlm.nih.gov/30602375/
Murata T, Takayama K, Katayama S, et al. miR-148a is an androgen-responsive microRNA that promotes LNCaP prostate cell growth by repressing its target CAND1 expression. Prostate Cancer Prostatic Dis. 2010;13(4):356–61. https://pubmed.ncbi.nlm.nih.gov/20820187/
Tate PL, Bibb R, Larcom LL. Milk stimulates growth of prostate cancer cells in culture. Nutr Cancer. 2011;63(8):1361–6. https://pubmed.ncbi.nlm.nih.gov/22043817/
Sargsyan A, Dubasi HB. Milk consumption and prostate cancer: a systematic review. World J Mens Health. 2021;39(3):419–28. https://pubmed.ncbi.nlm.nih.gov/32777868/
Melnik BC, Schmitz G. Milk’s role as an epigenetic regulator in health and disease. Diseases. 2017;5(1):12. https://pubmed.ncbi.nlm.nih.gov/28933365/
Melnik BC, Schmitz G. Exosomes of pasteurized milk: potential pathogens of Western diseases. J Transl Med. 2019;17(1):3. https://pubmed.ncbi.nlm.nih.gov/30602375/
Michaëlsson K, Wolk A, Langenskiöld S, et al. Milk intake and risk of mortality and fractures in women and men: cohort studies. BMJ. 2014;349:g6015. https://pubmed.ncbi.nlm.nih.gov/25352269/
Yu S, Zhao Z, Sun LM, Li P. Fermentation results in quantitative changes in milk-derived exosomes and different effects on cell growth and survival. J Agric Food Chem. 2017;65(6):1220–8. https://pubmed.ncbi.nlm.nih.gov/28085261/
Savaiano DA, Hutkins RW. Yogurt, cultured fermented milk, and health: a systematic review. Nutr Rev. 2021;79(5):599–614. https://pubmed.ncbi.nlm.nih.gov/32447398/
Wehbe Z, Kreydiyyeh S. Cow’s milk may be delivering potentially harmful undetected cargoes to humans. Is it time to reconsider dairy recommendations? Nutr Rev. 2022;80(4):874–88. https://pubmed.ncbi.nlm.nih.gov/34338770/
Melnik BC, Schmitz G. Exosomes of pasteurized milk: potential pathogens of Western diseases. J Transl Med. 2019;17(1):3. https://pubmed.ncbi.nlm.nih.gov/30602375/
Teodori L, Petrignani I, Giuliani A, et al. Inflamm-aging microRNAs may integrate signals from food and gut microbiota by modulating common signalling pathways. Mech Ageing Dev. 2019;182:111127. https://pubmed.ncbi.nlm.nih.gov/31401225/
Tsai F, Coyle WJ. The microbiome and obesity: is obesity linked to our gut flora? Curr Gastroenterol Rep. 2009;11(4):307–13. https://pubmed.ncbi.nlm.nih.gov/19615307/
Stephen AM, Cummings JH. The microbial contribution to human faecal mass. J Med Microbiol. 1980;13(1):45–56. https://pubmed.ncbi.nlm.nih.gov/7359576/
Singh RK, Chang HW, Yan D, et al. Influence of diet on the gut microbiome and implications for human health. J Transl Med. 2017;15(1):73. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5385025/
Kim A. Dysbiosis: a review highlighting obesity and inflammatory bowel disease. J Clin Gastroenterol. 2015;49 Suppl 1:S20–4. https://pubmed.ncbi.nlm.nih.gov/26447959/
Patterson E, Ryan PM, Cryan JF, et al. Gut microbiota, obesity and diabetes. Postgrad Med J. 2016;92(1087):286–300. https://pubmed.ncbi.nlm.nih.gov/26912499/
Tsai F, Coyle WJ. The microbiome and obesity: is obesity linked to our gut flora? Curr Gastroenterol Rep. 2009;11(4):307–13. https://pubmed.ncbi.nlm.nih.gov/19615307/
Wikoff W, Anfora A, Liu J, et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A. 2009;106(10):3698–703. https://pubmed.ncbi.nlm.nih.gov/19234110/
Sanmiguel C, Gupta A, Mayer EA. Gut microbiome and obesity: a plausible explanation for obesity. Curr Obes Rep. 2015;4(2):250–61. https://pubmed.ncbi.nlm.nih.gov/26029487/
Aydin S. Can peptides and gut microbiota be involved in the etiopathology of obesity? Obes Surg. 2017;27(1):202–4. https://pubmed.ncbi.nlm.nih.gov/27787759/
Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016;14(8):e1002533. https://pubmed.ncbi.nlm.nih.gov/27541692/
Goodacre R. Metabolomics of a superorganism. J Nutr. 2007;137(1 Suppl):259S-66S. https://pubmed.ncbi.nlm.nih.gov/17182837/
Simões CD, Maukonen J, Kaprio J, Rissanen A, Pietiläinen KH, Saarela M. Habitual dietary intake is associated with stool microbiota composition in monozygotic twins. J Nutr. 2013;143(4):417–23. https://pubmed.ncbi.nlm.nih.gov/23343669/
Biagi E, Nylund L, Candela M, et al. Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One. 2010;5(5):e10667. https://pubmed.ncbi.nlm.nih.gov/20498852/
Bana B, Cabreiro F. The microbiome and aging. Annu Rev Genet. 2019;53:239–61. https://pubmed.ncbi.nlm.nih.gov/31487470/
Bischoff SC. Microbiota and aging. Curr Opin Clin Nutr Metab Care. 2016;19(1):26–30. https://pubmed.ncbi.nlm.nih.gov/26560527/
Venkatakrishnan A, Holzknecht ZE, Holzknecht R, et al. Evolution of bacteria in the human gut in response to changing environments: an invisible player in the game of health. Comput Struct Biotechnol J. 2021;19:752–8. https://pubmed.ncbi.nlm.nih.gov/33552447/
Vermorken AJM, Cui Y, Kleerebezem R, Andrès E. Bowel movement frequency and cardiovascular mortality, a matter of fibers and oxidative stress? Atherosclerosis. 2016;253:278–80. https://pubmed.ncbi.nlm.nih.gov/27594542/
Lichtenstein GR. Letter from the editor. Gastroenterol Hepatol (N Y). 2013;9(9):552. https://pubmed.ncbi.nlm.nih.gov/24729764/
Allison DB, Roberts MS. On constructing the disorder of hysteria. J Med Philos. 1994;19(3):239–59. https://pubmed.ncbi.nlm.nih.gov/7964210/
Walter J, Armet AM, Finlay BB, Shanahan F. Establishing or exaggerating causality for the gut microbiome: lessons from human microbiota-associated rodents. Cell. 2020;180(2):221–32. https://pubmed.ncbi.nlm.nih.gov/31978342/
Copeland CE, Stahlfeld K. Two tall poppies and the discovery of Helicobacter pylori. J Am Coll Surg. 2012;214(2):237–41. https://pubmed.ncbi.nlm.nih.gov/22056357/
Walter J, Armet AM, Finlay BB, Shanahan F. Establishing or exaggerating causality for the gut microbiome: lessons from human microbiota-associated rodents. Cell. 2020;180(2):221–32. https://pubmed.ncbi.nlm.nih.gov/31978342/
Ragonnaud E, Biragyn A. Gut microbiota as the key controllers of “healthy” aging of elderly people. Immun Ageing. 2021;18(1):2. https://pubmed.ncbi.nlm.nih.gov/33397404/
Metchnikoff É. The Prolongation of Life: Optimistic Studies. G.P. Putnam’s Sons; 1908. https://www.gutenberg.org/files/51521/51521-h/51521-h.htm
Mackowiak PA. Recycling Metchnikoff: probiotics, the intestinal microbiome and the quest for long life. Front Public Health. 2013;1. https://pubmed.ncbi.nlm.nih.gov/24350221/
Vikhanski L. Elie Metchnikoff rediscovered: comeback of a founding father of gerontology. Gerontologist. 2016;56(Suppl_3):181. https://doi.org/10.1093/geront/gnw162.708
Gasbarrini G, Bonvicini F, Gramenzi A. Probiotics history. J Clin Gastroenterol. 2016;50(Supplement 2):S116–9. https://pubmed.ncbi.nlm.nih.gov/27741152/
Mackowiak PA. Recycling Metchnikoff: probiotics, the intestinal microbiome and the quest for long life. Front Public Health. 2013;1. https://pubmed.ncbi.nlm.nih.gov/24350221/
Mowat AM. Historical perspective: Metchnikoff and the intestinal microbiome. J Leukoc Biol. 2021;109(3):513–7. https://pubmed.ncbi.nlm.nih.gov/33630385/
Salazar N, Valdés-Varela L, González S, Gueimonde M, de Los Reyes-Gavilán CG. Nutrition and the gut microbiome in the elderly. Gut Microbes. 2017;8(2):82–97. https://pubmed.ncbi.nlm.nih.gov/27808595/
Narasimhan H, Ren CC, Deshpande S, Sylvia KE. Young at gut – turning back the clock with the gut microbiome. Microorganisms. 2021;9(3):555. https://pubmed.ncbi.nlm.nih.gov/33800340/
Madison AA, Kiecolt-Glaser JK. The gut microbiota and nervous system: age-defined and age-defying. Semin Cell Dev Biol. 2021;116:98–107. https://pubmed.ncbi.nlm.nih.gov/33422403/
Xu C, Zhu H, Qiu P. Aging progression of human gut microbiota. BMC Microbiol. 2019;19(1):236. https://pubmed.ncbi.nlm.nih.gov/31660868/
Galkin F, Mamoshina P, Aliper A, et al. Human gut microbiome aging clock based on taxonomic profiling and deep learning. iScience. 2020;23(6):101199. https://pubmed.ncbi.nlm.nih.gov/32534441/
Ling Z, Liu X, Cheng Y, Yan X, Wu S. Gut microbiota and aging. Crit Rev Food Sci Nutr. 2022;62(13):3509–34. https://pubmed.ncbi.nlm.nih.gov/33377391/
Vaiserman AM, Koliada AK, Marotta F. Gut microbiota: a player in aging and a target for anti-aging intervention. Ageing Res Rev. 2017;35:36–45. https://pubmed.ncbi.nlm.nih.gov/28109835/
An R, Wilms E, Masclee AAM, Smidt H, Zoetendal EG, Jonkers D. Age-dependent changes in GI physiology and microbiota: time to reconsider? Gut. 2018;67(12):2213–22. https://pubmed.ncbi.nlm.nih.gov/30194220/
Vemuri R, Gundamaraju R, Shastri MD, et al. Gut microbial changes, interactions, and their implications on human lifecycle: an ageing perspective. BioMed Res Int. 2018;2018:4178607. https://pubmed.ncbi.nlm.nih.gov/29682542/
Ragonnaud E, Biragyn A. Gut microbiota as the key controllers of “healthy” aging of elderly people. Immun Ageing. 2021;18(1):2. https://pubmed.ncbi.nlm.nih.gov/33397404/
Biagi E, Nylund L, Candela M, et al. Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One. 2010;5(5):e10667. https://pubmed.ncbi.nlm.nih.gov/20498852/
Badal VD, Vaccariello ED, Murray ER, et al. The gut microbiome, aging, and longevity: a systematic review. Nutrients. 2020;12(12):E3759. https://pubmed.ncbi.nlm.nih.gov/33297486/
Cai D, Zhao S, Li D, et al. Nutrient intake is associated with longevity characterization by metabolites and element profiles of healthy centenarians. Nutrients. 2016;8(9):E564. https://pubmed.ncbi.nlm.nih.gov/27657115/
Zhang S, Zeng B, Chen Y, et al. Gut microbiota in healthy and unhealthy long-living people. Gene. 2021;779:145510. https://pubmed.ncbi.nlm.nih.gov/33600956/
DeJong EN, Surette MG, Bowdish DME. The gut microbiota and unhealthy aging: disentangling cause from consequence. Cell Host Microbe. 2020;28(2):180–9. https://pubmed.ncbi.nlm.nih.gov/32791111/
Biagi E, Nylund L, Candela M, et al. Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One. 2010;5(5):e10667. https://pubmed.ncbi.nlm.nih.gov/20498852/
Biagi E, Franceschi C, Rampelli S, et al. Gut microbiota and extreme longevity. Curr Biol. 2016;26(11):1480–5. https://pubmed.ncbi.nlm.nih.gov/27185560/
Ling Z, Liu X, Cheng Y, Yan X, Wu S. Gut microbiota and aging. Crit Rev Food Sci Nutr. 2022;62(13):3509–34. https://pubmed.ncbi.nlm.nih.gov/33377391/
Yang HY, Liu SL, Ibrahim SA, et al. Oral administration of live Bifidobacterium substrains isolated from healthy centenarians enhanced immune function in BALB/c mice. Nutr Res. 2009;29(4):281–9. https://pubmed.ncbi.nlm.nih.gov/19410981/
Exopolysaccharides produced by lactic acid bacteria and Bifidobacteria: structures, physiochemical functions and applications in the food industry. Food Hydrocoll. 2019;94:475–99. https://www.sciencedirect.com/science/article/abs/pii/S0268005X18313298?via%3Dihub
Koo H, Falsetta ML, Klein MI. The exopolysaccharide matrix: a virulence determinant of cariogenic biofilm. J Dent Res. 2013;92(12):1065–73. https://pubmed.ncbi.nlm.nih.gov/24045647/
Xu R, Shang N, Li P. In vitro and in vivo antioxidant activity of exopolysaccharide fractions from Bifidobacterium animalis RH. Anaerobe. 2011;17(5):226–31. https://pubmed.ncbi.nlm.nih.gov/21132298/
O’Toole PW, Jeffery IB. Microbiome-health interactions in older people. Cell Mol Life Sci. 2018;75(1):119–28. https://pubmed.ncbi.nlm.nih.gov/28986601/
Everard A, Belzer C, Geurts L, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A. 2013;110(22):9066–71. https://pubmed.ncbi.nlm.nih.gov/23671105/
Shintouo CM, Mets T, Beckwee D, et al. Is inflammageing influenced by the microbiota in the aged gut? A systematic review. Exp Gerontol. 2020;141:111079. https://pubmed.ncbi.nlm.nih.gov/32882334/
Greathouse KL, Faucher MA, Hastings-Tolsma M. The gut microbiome, obesity, and weight control in women’s reproductive health. West J Nurs Res. 2017;39(8):1094–119. https://pubmed.ncbi.nlm.nih.gov/28303750/
Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16(1):35–56. https://pubmed.ncbi.nlm.nih.gov/30262901/
Desai MS, Seekatz AM, Koropatkin NM, et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell. 2016;167(5):1339–53.e21. https://pubmed.ncbi.nlm.nih.gov/27863247/
Roberts CL, Keita AV, Duncan SH, et al. Translocation of Crohn’s disease Escherichia coli across M-cells: contrasting effects of soluble plant fibres and emulsifiers. Gut. 2010;59(10):1331–9. https://pubmed.ncbi.nlm.nih.gov/20813719/
Biragyn A, Ferrucci L. Gut dysbiosis: a potential link between increased cancer risk in ageing and inflammaging. Lancet Oncol. 2018;19(6):e295–304. https://pubmed.ncbi.nlm.nih.gov/29893261/
Biagi E, Franceschi C, Rampelli S, et al. Gut microbiota and extreme longevity. Curr Biol. 2016;26(11):1480–5. https://pubmed.ncbi.nlm.nih.gov/27185560/
Ragonnaud E, Biragyn A. Gut microbiota as the key controllers of “healthy” aging of elderly people. Immun Ageing. 2021;18(1):2. https://pubmed.ncbi.nlm.nih.gov/33397404/
Biragyn A, Ferrucci L. Gut dysbiosis: a potential link between increased cancer risk in ageing and inflammaging. Lancet Oncol. 2018;19(6):e295–304. https://pubmed.ncbi.nlm.nih.gov/29893261/
Luan Z, Sun G, Huang Y, et al. Metagenomics study reveals changes in gut microbiota in centenarians: a cohort study of Hainan centenarians. Front Microbiol. 2020;11:1474. https://pubmed.ncbi.nlm.nih.gov/32714309/
Bárcena C, Valdés-Mas R, Mayoral P, et al. Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat Med. 2019;25(8):1234–42. https://pubmed.ncbi.nlm.nih.gov/31332389/
Vemuri R, Gundamaraju R, Shastri MD, et al. Gut microbial changes, interactions, and their implications on human lifecycle: an ageing perspective. BioMed Res Int. 2018;2018:4178607. https://pubmed.ncbi.nlm.nih.gov/29682542/
Wang F, Yu T, Huang G, et al. Gut microbiota community and its assembly associated with age and diet in Chinese centenarians. J Microbiol Biotechnol. 2015;25(8):1195–204. https://pubmed.ncbi.nlm.nih.gov/25839332/
C¿toi AF, Corina A, Katsiki N, et al. Gut microbiota and aging – a focus on centenarians. Biochim Biophys Acta Mol Basis Dis. 2020;1866(7):165765. https://pubmed.ncbi.nlm.nih.gov/32169505/
DeJong EN, Surette MG, Bowdish DME. The gut microbiota and unhealthy aging: disentangling cause from consequence. Cell Host Microbe. 2020;28(2):180–9. https://pubmed.ncbi.nlm.nih.gov/32791111/
Nagpal R, Mainali R, Ahmadi S, et al. Gut microbiome and aging: physiological and mechanistic insights. Nutr Healthy Aging. 2018;4(4):267–85. https://pubmed.ncbi.nlm.nih.gov/29951588/
O’Toole PW, Jeffery IB. Microbiome-health interactions in older people. Cell Mol Life Sci. 2018;75(1):119–28. https://pubmed.ncbi.nlm.nih.gov/28986601/
Biagi E, Rampelli S, Turroni S, Quercia S, Candela M, Brigidi P. The gut microbiota of centenarians: signatures of longevity in the gut microbiota profile. Mech Ageing Dev. 2017;165(Pt B):180–4. https://pubmed.ncbi.nlm.nih.gov/28049008/
O’Toole PW, Jeffery IB. Microbiome-health interactions in older people. Cell Mol Life Sci. 2018;75(1):119–28. https://pubmed.ncbi.nlm.nih.gov/28986601/
Buford TW. (Dis)Trust your gut: the gut microbiome in age-related inflammation, health, and disease. Microbiome. 2017;5(1):80. https://pubmed.ncbi.nlm.nih.gov/28709450/
Maier L, Pruteanu M, Kuhn M, et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555(7698):623–8. https://pubmed.ncbi.nlm.nih.gov/29555994/
Tiihonen K, Ouwehand AC, Rautonen N. Human intestinal microbiota and healthy ageing. Ageing Res Rev. 2010;9(2):107–16. https://pubmed.ncbi.nlm.nih.gov/19874918/
DeJong EN, Surette MG, Bowdish DME. The gut microbiota and unhealthy aging: disentangling cause from consequence. Cell Host Microbe. 2020;28(2):180–9. https://pubmed.ncbi.nlm.nih.gov/32791111/
Claesson MJ, Jeffery IB, Conde S, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488(7410):178–84. https://pubmed.ncbi.nlm.nih.gov/22797518/
Biagi E, Rampelli S, Turroni S, Quercia S, Candela M, Brigidi P. The gut microbiota of centenarians: signatures of longevity in the gut microbiota profile. Mech Ageing Dev. 2017;165(Pt B):180–4. https://pubmed.ncbi.nlm.nih.gov/28049008/
Rampelli S, Schnorr SL, Consolandi C, et al. Metagenome sequencing of the Hadza hunter-gatherer gut microbiota. Curr Biol. 2015;25(13):1682–93. https://pubmed.ncbi.nlm.nih.gov/25981789/
Rampelli S, Soverini M, D’Amico F, et al. Shotgun metagenomics of gut microbiota in humans with up to extreme longevity and the increasing role of xenobiotic degradation. mSystems. 2020;5(2):e00124–20. https://pubmed.ncbi.nlm.nih.gov/32209716/
Rampelli S, Soverini M, D’Amico F, et al. Shotgun metagenomics of gut microbiota in humans with up to extreme longevity and the increasing role of xenobiotic degradation. mSystems. 2020;5(2):e00124–20. https://pubmed.ncbi.nlm.nih.gov/32209716/
Cai D, Zhao S, Li D, et al. Nutrient intake is associated with longevity characterization by metabolites and element profiles of healthy centenarians. Nutrients. 2016;8(9):E564. https://pubmed.ncbi.nlm.nih.gov/27657115/
Smith P, Willemsen D, Popkes M, et al. Regulation of life span by the gut microbiota in the short-lived African turquoise killifish. eLife. 2017;6:e27014. https://pubmed.ncbi.nlm.nih.gov/28826469/
Chen Y, Zhang S, Zeng B, et al. Transplant of microbiota from long-living people to mice reduces aging-related indices and transfers beneficial bacteria. Aging (Albany NY). 2020;12(6):4778–93. https://pubmed.ncbi.nlm.nih.gov/32176868/
David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–63. https://pubmed.ncbi.nlm.nih.gov/24336217/
Chen Y, Zhang S, Zeng B, et al. Transplant of microbiota from long-living people to mice reduces aging-related indices and transfers beneficial bacteria. Aging (Albany NY). 2020;12(6):4778–93. https://pubmed.ncbi.nlm.nih.gov/32176868/
Kim M, Benayoun BA. The microbiome: an emerging key player in aging and longevity. Transl Med Aging. 2020;4:103–16. https://pubmed.ncbi.nlm.nih.gov/32832742/
Riccio P, Rossano R. Undigested food and gut microbiota may cooperate in the pathogenesis of neuroinflammatory diseases: a matter of barriers and a proposal on the origin of organ specificity. Nutrients. 2019;11(11):E2714. https://pubmed.ncbi.nlm.nih.gov/31717475/
Riccio P, Rossano R. Undigested food and gut microbiota may cooperate in the pathogenesis of neuroinflammatory diseases: a matter of barriers and a proposal on the origin of organ specificity. Nutrients. 2019;11(11):E2714. https://pubmed.ncbi.nlm.nih.gov/31717475/
Patel KP, Luo FJ, Plummer NS, Hostetter TH, Meyer TW. The production of p-cresol sulfate and indoxyl sulfate in vegetarians versus omnivores. Clin J Am Soc Nephrol. 2012;7(6):982–8. https://pubmed.ncbi.nlm.nih.gov/22490877/
Riccio P, Rossano R. Undigested food and gut microbiota may cooperate in the pathogenesis of neuroinflammatory diseases: a matter of barriers and a proposal on the origin of organ specificity. Nutrients. 2019;11(11):E2714. https://pubmed.ncbi.nlm.nih.gov/31717475/
Russo F, Linsalata M, Clemente C, et al. Inulin-enriched pasta improves intestinal permeability and modifies the circulating levels of zonulin and glucagon-like peptide 2 in healthy young volunteers. Nutr Res. 2012;32(12):940–6. https://pubmed.ncbi.nlm.nih.gov/23244539/
Donnadieu-Rigole H, Pansu N, Mura T, et al. Beneficial effect of alcohol withdrawal on gut permeability and microbial translocation in patients with alcohol use disorder. Alcohol Clin Exp Res. 2018;42(1):32–40. https://pubmed.ncbi.nlm.nih.gov/29030980/
Lambert GP, Schmidt A, Schwarzkopf K, Lanspa S. Effect of aspirin dose on gastrointestinal permeability. Int J Sports Med. 2012;33(6):421–5. https://pubmed.ncbi.nlm.nih.gov/22377941/
Ivey KJ, Baskin WN, Krause WJ, Terry B. Effect of aspirin and acid on human jejunal mucosa. An ultrastructural study. Gastroenterology. 1979;76(1):50–6. https://pubmed.ncbi.nlm.nih.gov/758147/
Tran CD, Hawkes J, Graham RD, et al. Zinc-fortified oral rehydration solution improved intestinal permeability and small intestinal mucosal recovery. Clin Pediatr (Phila). 2015;54(7):676–82. https://pubmed.ncbi.nlm.nih.gov/25520366/
Ling Z, Liu X, Cheng Y, Yan X, Wu S. Gut microbiota and aging. Crit Rev Food Sci Nutr. 2022;62(13):3509–34. https://pubmed.ncbi.nlm.nih.gov/33377391/
Giovannini S, Onder G, Liperoti R, et al. Interleukin-6, C-reactive protein, and tumor necrosis factor-alpha as predictors of mortality in frail, community-living elderly individuals. J Am Geriatr Soc. 2011;59(9):1679–85. https://pubmed.ncbi.nlm.nih.gov/21883115/
de Gonzalo-Calvo D, de Luxán-Delgado B, Martínez-Camblor P, et al. Chronic inflammation as predictor of 1-year hospitalization and mortality in elderly population. Eur J Clin Invest. 2012;42(10):1037–46. https://pubmed.ncbi.nlm.nih.gov/22624958/
Soysal P, Stubbs B, Lucato P, et al. Inflammation and frailty in the elderly: a systematic review and meta-analysis. Ageing Res Rev. 2016;31:1–8. https://pubmed.ncbi.nlm.nih.gov/27592340/
de Gonzalo-Calvo D, de Luxán-Delgado B, Rodríguez-González S, et al. Interleukin 6, soluble tumor necrosis factor receptor I and red blood cell distribution width as biological markers of functional dependence in an elderly population: a translational approach. Cytokine. 2012;58(2):193–8. https://pubmed.ncbi.nlm.nih.gov/22624958/
Thevaranjan N, Puchta A, Schulz C, et al. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe. 2017;21(4):455–66.e4. https://pubmed.ncbi.nlm.nih.gov/28407483/
Lustgarten MS. Classifying aging as a disease: the role of microbes. Front Genet. 2016;7:212. https://pubmed.ncbi.nlm.nih.gov/27990156/
Evans CJ, Ho Y, Daveson BA, et al. Place and cause of death in centenarians: a population-based observational study in England, 2001 to 2010. PLoS Med. 2014;11(6):e1001653. https://pubmed.ncbi.nlm.nih.gov/24892645/
Yende S, Tuomanen EI, Wunderink R, et al. Preinfection systemic inflammatory markers and risk of hospitalization due to pneumonia. Am J Respir Crit Care Med. 2005;172(11):1440–6. https://pubmed.ncbi.nlm.nih.gov/16166617/
Antunes G, Evans SA, Lordan JL, Frew AJ. Systemic cytokine levels in community-acquired pneumonia and their association with disease severity. Eur Respir J. 2002;20(4):990–5. https://pubmed.ncbi.nlm.nih.gov/16166617/
Reade MC, Yende S, D’Angelo G, et al. Differences in immune response may explain lower survival among older men with pneumonia. Crit Care Med. 2009;37(5):1655–62. https://pubmed.ncbi.nlm.nih.gov/19325487/
Hearps AC, Martin GE, Angelovich TA, et al. Aging is associated with chronic innate immune activation and dysregulation of monocyte phenotype and function. Aging Cell. 2012;11(5):867–75. https://pubmed.ncbi.nlm.nih.gov/22708967/
Thevaranjan N, Puchta A, Schulz C, et al. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe. 2017;21(4):455–66. https://pubmed.ncbi.nlm.nih.gov/28407483/
Biragyn A, Ferrucci L. Gut dysbiosis: a potential link between increased cancer risk in ageing and inflammaging. Lancet Oncol. 2018;19(6):e295–304. https://pubmed.ncbi.nlm.nih.gov/29893261/
Venkatakrishnan A, Holzknecht ZE, Holzknecht R, et al. Evolution of bacteria in the human gut in response to changing environments: an invisible player in the game of health. Comput Struct Biotechnol J. 2021;19:752–8. https://pubmed.ncbi.nlm.nih.gov/33552447/
Kilkkinen A, Rissanen H, Klaukka T, et al. Antibiotic use predicts an increased risk of cancer. Int J Cancer. 2008;123(9):2152–5. https://pubmed.ncbi.nlm.nih.gov/18704945/
Wise R, Hart T, Cars O, et al. Antimicrobial resistance. BMJ. 1998;317(7159):609–10. https://pubmed.ncbi.nlm.nih.gov/9727981/
Vemuri R, Gundamaraju R, Shastri MD, et al. Gut microbial changes, interactions, and their implications on human lifecycle: an ageing perspective. BioMed Res Int. 2018;2018:4178607. https://pubmed.ncbi.nlm.nih.gov/29682542/
Collignon PJ, Conly JM, Andremont A, et al. World Health Organization ranking of antimicrobials according to their importance in human medicine: a critical step for developing risk management strategies to control antimicrobial resistance from food animal production. Clin Infect Dis. 2016;63(8):1087–93. https://pubmed.ncbi.nlm.nih.gov/27439526/
Office of Technology Assessment. Drugs in Livestock Feed. Congress of the United States, Office of Technology Assessment; 1979. https://worldcat.org/title/1295046620
Galloway-Peña JR, Jenq RR. The only thing that stops a bad microbiome, is a good microbiome. Haematologica. 2019;104(8):1511–3. https://pubmed.ncbi.nlm.nih.gov/31366464/
Subirats J, Domingues A, Topp E. Does dietary consumption of antibiotics by humans promote antibiotic resistance in the gut microbiome? J Food Prot. 2019;82(10):1636–42. https://pubmed.ncbi.nlm.nih.gov/31512932/
Angulo FJ, Baker NL, Olsen SJ, Anderson A, Barrett TJ. Antimicrobial use in agriculture: controlling the transfer of antimicrobial resistance to humans. Semin Pediatr Infect Dis. 2004;15(2):78–85. https://pubmed.ncbi.nlm.nih.gov/15185190/
Milanovic V, Osimani A, Aquilanti L, et al. Occurrence of antibiotic resistance genes in the fecal DNA of healthy omnivores, ovo-lacto vegetarians and vegans. Mol Nutr Food Res. 2017;61(9). https://pubmed.ncbi.nlm.nih.gov/28464483/
Cabral DJ, Wurster JI, Korry BJ, Penumutchu S, Belenky P. Consumption of a Western-style diet modulates the response of the murine gut microbiome to ciprofloxacin. mSystems. 2020;5(4):e00317–20. https://pubmed.ncbi.nlm.nih.gov/32723789/
Schnizlein MK, Vendrov KC, Edwards SJ, Martens EC, Young VB. Dietary xanthan gum alters antibiotic efficacy against the murine gut microbiota and attenuates Clostridioides difficile colonization. mSphere. 2020;5(1):e00708–19. https://pubmed.ncbi.nlm.nih.gov/31915217/
Bassis CM. Live and diet by your gut microbiota. mBio. 2019;10(5):e02335–19. https://pubmed.ncbi.nlm.nih.gov/31594820/
Martínez Steele E, Baraldi LG, Louzada ML da C, Moubarac JC, Mozaffarian D, Monteiro CA. Ultra-processed foods and added sugars in the US diet: evidence from a nationally representative cross-sectional study. BMJ Open. 2016;6(3):e009892. https://pubmed.ncbi.nlm.nih.gov/26962035/
Wilck N, Matus MG, Kearney SM, et al. Salt-responsive gut commensal modulates TH17 axis and disease. Nature. 2017;551(7682):585–9. https://pubmed.ncbi.nlm.nih.gov/29143823/
Bisoendial R, Lubberts E. A mechanistic insight into the pathogenic role of interleukin 17A in systemic autoimmune diseases. Mediators Inflamm. 2022;2022:6600264. https://pubmed.ncbi.nlm.nih.gov/35620115/
Wilck N, Matus MG, Kearney SM, et al. Salt-responsive gut commensal modulates TH17 axis and disease. Nature. 2017;551(7682):585–9. https://pubmed.ncbi.nlm.nih.gov/29143823/
Suez J, Korem T, Zilberman-Schapira G, Segal E, Elinav E. Non-caloric artificial sweeteners and the microbiome: findings and challenges. Gut Microbes. 2015;6(2):149–55. https://pubmed.ncbi.nlm.nih.gov/25831243/
Laster J, Bonnes SL, Rocha J. Increased use of emulsifiers in processed foods and the links to obesity. Curr Gastroenterol Rep. 2019;21(11):61. https://pubmed.ncbi.nlm.nih.gov/31792622/
Naimi S, Viennois E, Gewirtz AT, Chassaing B. Direct impact of commonly used dietary emulsifiers on human gut microbiota. Microbiome. 2021;9(1):66. https://pubmed.ncbi.nlm.nih.gov/33752754/
Birkett A, Muir J, Phillips J, Jones G, O’Dea K. Resistant starch lowers fecal concentrations of ammonia and phenols in humans. Am J Clin Nutr. 1996;63(5):766–72. https://pubmed.ncbi.nlm.nih.gov/8615362/
Windey K, De Preter V, Verbeke K. Relevance of protein fermentation to gut health. Mol Nutr Food Res. 2012;56(1):184–96. https://pubmed.ncbi.nlm.nih.gov/22121108/
Magee E. A nutritional component to inflammatory bowel disease: the contribution of meat to fecal sulfide excretion. Nutrition. 1999;15(3):244–6. https://pubmed.ncbi.nlm.nih.gov/10198924/
Ge J, Han TJ, Liu J, et al. Meat intake and risk of inflammatory bowel disease: a meta-analysis. Turk J Gastroenterol. 2015;26(6):492–7. https://pubmed.ncbi.nlm.nih.gov/26575042/
Parra-Soto S, Ahumada D, Petermann-Rocha F, et al. Association of meat, vegetarian, pescatarian and fish-poultry diets with risk of 19 cancer sites and all cancer: findings from the UK Biobank prospective cohort study and meta-analysis. BMC Med. 2022;20(1):79. https://pubmed.ncbi.nlm.nih.gov/35655214/
Florin THJ, Neale G, Goretski S, Cummings JH. The sulfate content of foods and beverages. J Food Comp Anal. 1993;6(2):140–51. https://pubmed.ncbi.nlm.nih.gov/1855683/
Ananthakrishnan AN, Khalili H, Konijeti GG, et al. A prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitis. Gastroenterology. 2013;145(5):970–7. https://pubmed.ncbi.nlm.nih.gov/23912083/
Yao CK, Muir JG, Gibson PR. Review article: insights into colonic protein fermentation, its modulation and potential health implications. Aliment Pharmacol Ther. 2016;43(2):181–96. https://pubmed.ncbi.nlm.nih.gov/26527169/
Bosch S, Lemmen JPM, Menezes R, et al. The influence of lifestyle factors on fecal volatile organic compound composition as measured by an electronic nose. J Breath Res. 2019;13(4):046001. https://pubmed.ncbi.nlm.nih.gov/31170704/
How you can limit your gas production. 12 tips for dealing with flatulence. Harv Health Lett. 2007;32(12):3. https://pubmed.ncbi.nlm.nih.gov/18246621/
do Rosario VA, Fernandes R, Trindade EBS de M. Vegetarian diets and gut microbiota: important shifts in markers of metabolism and cardiovascular disease. Nutr Rev. 2016;74(7):444–54. https://pubmed.ncbi.nlm.nih.gov/27261272/
Magee EA, Richardson CJ, Hughes R, Cummings JH. Contribution of dietary protein to sulfide production in the large intestine: an in vitro and a controlled feeding study in humans. Am J Clin Nutr. 2000;72(6):1488–94. https://pubmed.ncbi.nlm.nih.gov/11101476/
Falony G, Vieira-Silva S, Raes J. Microbiology meets big data: the case of gut microbiota-derived trimethylamine. Annu Rev Microbiol. 2015;69:305–21. https://pubmed.ncbi.nlm.nih.gov/26274026/
Rak K, Rader DJ. Cardiovascular disease: the diet-microbe morbid union. Nature. 2011;472(7341):40–1. https://pubmed.ncbi.nlm.nih.gov/21475185/
Tang WHW, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368(17):1575–84. https://pubmed.ncbi.nlm.nih.gov/23614584/
Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–85. https://pubmed.ncbi.nlm.nih.gov/23563705/
Farhangi MA, Vajdi M, Asghari-Jafarabadi M. Gut microbiota-associated metabolite trimethylamine N-oxide and the risk of stroke: a systematic review and dose-response meta-analysis. Nutr J. 2020;19(1):76. https://pubmed.ncbi.nlm.nih.gov/32731904/
Zhai Q, Wang X, Chen C, et al. Prognostic value of plasma trimethylamine n-oxide levels in patients with acute ischemic stroke. Cell Mol Neurobiol. 2019;39(8):1201–6. https://pubmed.ncbi.nlm.nih.gov/31332666/
Zhu W, Wang Z, Tang WHW, Hazen SL. Gut microbe-generated trimethylamine N-oxide from dietary choline is prothrombotic in subjects. Circulation. 2017;135(17):1671–3. https://pubmed.ncbi.nlm.nih.gov/28438808/
Brunt VE, Gioscia-Ryan RA, Casso AG, et al. Trimethylamine-N-oxide promotes age-related vascular oxidative stress and endothelial dysfunction in mice and healthy humans. Hypertension. 2020;76(1):101–12. https://pubmed.ncbi.nlm.nih.gov/32520619/
He Z, Chen ZY. What are missing parts in the research story of trimethylamine-n-oxide (TMAO)? J Agric Food Chem. 2017;65(26):5227–8. https://pubmed.ncbi.nlm.nih.gov/28650144/
Coras R, Kavanaugh A, Boyd T, et al. Choline metabolite, trimethylamine N-oxide (TMAO), is associated with inflammation in psoriatic arthritis. Clin Exp Rheumatol. 2019;37(3):481–4. https://pubmed.ncbi.nlm.nih.gov/30620278/
Eyupoglu ND, Caliskan Guzelce E, Acikgoz A, et al. Circulating gut microbiota metabolite trimethylamine N-oxide and oral contraceptive use in polycystic ovary syndrome. Clin Endocrinol (Oxf). 2019;91(6):810–5. https://pubmed.ncbi.nlm.nih.gov/31556132/
Zeng ST, Guo L, Liu SK, et al. Egg consumption is associated with increased risk of ovarian cancer: evidence from a meta-analysis of observational studies. Clin Nutr. 2015;34(4):635–41. https://pubmed.ncbi.nlm.nih.gov/25108572/
Tse G, Eslick GD. Egg consumption and risk of GI neoplasms: dose-response meta-analysis and systematic review. Eur J Nutr. 2014;53(7):1581–90. https://pubmed.ncbi.nlm.nih.gov/24500371/
Chan CWH, Law BMH, Waye MMY, Chan JYW, So WKW, Chow KM. Trimethylamine-N-oxide as one hypothetical link for the relationship between intestinal microbiota and cancer – where we are and where shall we go? J Cancer. 2019;10(23):5874–82. https://pubmed.ncbi.nlm.nih.gov/31737123/
Heron M. Deaths: leading causes for 2017. Natl Vital Stat Rep. 2019;68(6):1–77. https://pubmed.ncbi.nlm.nih.gov/32501203/
Vogt NM, Romano KA, Darst BF, et al. The gut microbiota-derived metabolite trimethylamine N-oxide is elevated in Alzheimer’s disease. Alzheimers Res Ther. 2018;10(1):124. https://pubmed.ncbi.nlm.nih.gov/30579367/
Heron M. Deaths: leading causes for 2017. Natl Vital Stat Rep. 2019;68(6):1–77. https://pubmed.ncbi.nlm.nih.gov/32501203/
Ottiger M, Nickler M, Steuer C, et al. Trimethylamine-N-oxide (TMAO) predicts fatal outcomes in community-acquired pneumonia patients without evident coronary artery disease. Eur J Intern Med. 2016;36:67–73. https://pubmed.ncbi.nlm.nih.gov/27567042/
Rhee EP, Clish CB, Ghorbani A, et al. A combined epidemiologic and metabolomic approach improves CKD prediction. J Am Soc Nephrol. 2013;24(8):1330–8. https://pubmed.ncbi.nlm.nih.gov/23687356/
Farhangi MA. Gut microbiota – dependent trimethylamine N-oxide and all-cause mortality: findings from an updated systematic review and meta-analysis. Nutrition. 2020;78:110856. https://pubmed.ncbi.nlm.nih.gov/32592979/
Tang WH, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368(17):1575–84. https://pubmed.ncbi.nlm.nih.gov/23614584/
Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–85. https://pubmed.ncbi.nlm.nih.gov/23563705/
Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–85. https://pubmed.ncbi.nlm.nih.gov/23563705/
Demarquoy J, Georges, B, Rigault C, et al. Radioisotopic determination of L-carnitine content in foods commonly eaten in Western countries. Food Chem. 2004;86(1):137–42. https://doi.org/10.1016/j.foodchem.2003.09.023
Stefan M, Sharp M, Gheith R, et al. L-carnitine tartrate supplementation for 5 weeks improves exercise recovery in men and women: a randomized, double-blind, placebo-controlled trial. Nutrients. 2021;13(10):3432. https://pubmed.ncbi.nlm.nih.gov/34684429/
Koeth RA, Lam-Galvez BR, Kirsop J, et al. L–Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J Clin Invest. 2019;129(1):373–87. https://pubmed.ncbi.nlm.nih.gov/30530985/
Hernández-Alonso P, Cañueto D, Giardina S, et al. Effect of pistachio consumption on the modulation of urinary gut microbiota-related metabolites in prediabetic subjects. J Nutr Biochem. 2017;45:48–53. https://pubmed.ncbi.nlm.nih.gov/28432876/
Cashman JR, Xiong Y, Lin J, et al. In vitro and in vivo inhibition of human flavin-containing monooxygenase form 3 (FMO3) in the presence of dietary indoles. Biochem Pharmacol. 1999;58(6):1047–55. https://pubmed.ncbi.nlm.nih.gov/10509757/
Winther SA, Rossing P. TMAO: trimethylamine-N-oxide or time to minimize intake of animal products? J Clin Endocrinol Metab. 2020;105(12):e4958–60. https://pubmed.ncbi.nlm.nih.gov/32701146/
Galloway-Peña JR, Jenq RR. The only thing that stops a bad microbiome, is a good microbiome. Haematologica. 2019;104(8):1511–3. https://pubmed.ncbi.nlm.nih.gov/31366464/
Suez J, Zmora N, Segal E, Elinav E. The pros, cons, and many unknowns of probiotics. Nat Med. 2019;25(5):716–29. https://pubmed.ncbi.nlm.nih.gov/31061539/
Neunez M, Goldman M, Ghezzi P. Online information on probiotics: does it match scientific evidence? Front Med. 2020;6:296. https://pubmed.ncbi.nlm.nih.gov/32010699/
Hutchinson AN, Bergh C, Kruger K, et al. The effect of probiotics on health outcomes in the elderly: a systematic review of randomized, placebo-controlled studies. Microorganisms. 2021;9(6):1344. https://pubmed.ncbi.nlm.nih.gov/34205818/
Bafeta A, Koh M, Riveros C, Ravaud P. Harms reporting in randomized controlled trials of interventions aimed at modifying microbiota: a systematic review. Ann Intern Med. 2018;169(4):240–7. https://pubmed.ncbi.nlm.nih.gov/30014150/
Turjeman S, Koren O. ARGuing the case for (or against) probiotics. Trends Microbiol. 2021;29(11):959–60. https://pubmed.ncbi.nlm.nih.gov/34563432/
Montassier E, Valdés-Mas R, Batard E, et al. Probiotics impact the antibiotic resistance gene reservoir along the human GI tract in a person-specific and antibiotic-dependent manner. Nat Microbiol. 2021;6(8):1043–54. https://pubmed.ncbi.nlm.nih.gov/34226711/
Suez J, Zmora N, Zilberman-Schapira G, et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell. 2018;174(6):1406–23.e16. https://pubmed.ncbi.nlm.nih.gov/30193113/
Suez J, Zmora N, Segal E, Elinav E. The pros, cons, and many unknowns of probiotics. Nat Med. 2019;25(5):716–29. https://pubmed.ncbi.nlm.nih.gov/31061539/
Swain Ewald HA, Ewald PW. Natural selection, the microbiome, and public health. Yale J Biol Med. 2018;91(4):445–55. https://pubmed.ncbi.nlm.nih.gov/30588210/
Peng Z, Mao X, Zhang J, Du G, Chen J. Effective biodegradation of chicken feather waste by co-cultivation of keratinase producing strains. Microb Cell Fact. 2019;18(1):84. https://pubmed.ncbi.nlm.nih.gov/31103032/
Lewis ZT, Shani G, Masarweh CF, et al. Validating bifidobacterial species and subspecies identity in commercial probiotic products. Pediatr Res. 2016;79(3):445–52. https://pubmed.ncbi.nlm.nih.gov/26571226/
Drago L, Rodighiero V, Celeste T, Rovetto L, De Vecchi E. Microbiological evaluation of commercial probiotic products available in the USA in 2009. J Chemother. 2010;22(6):373–7. https://pubmed.ncbi.nlm.nih.gov/21303743/
Mazzantini D, Calvigioni M, Celandroni F, Lupetti A, Ghelardi E. Spotlight on the compositional quality of probiotic formulations marketed worldwide. Front Microbiol. 2021;12:693973. https://pubmed.ncbi.nlm.nih.gov/34354690/
Temmerman R, Pot B, Huys G, Swings J. Identification and antibiotic susceptibility of bacterial isolates from probiotic products. Int J Food Microbiol. 2003;81(1):1–10. https://pubmed.ncbi.nlm.nih.gov/12423913/
Mazzantini D, Calvigioni M, Celandroni F, Lupetti A, Ghelardi E. Spotlight on the compositional quality of probiotic formulations marketed worldwide. Front Microbiol. 2021;12:693973. https://pubmed.ncbi.nlm.nih.gov/34354690/
de Simone C. The unregulated probiotic market. Clin Gastroenterol Hepatol. 2019;17(5):809–17. https://pubmed.ncbi.nlm.nih.gov/29378309/
Suez J, Zmora N, Segal E, Elinav E. The pros, cons, and many unknowns of probiotics. Nat Med. 2019;25(5):716–29. https://pubmed.ncbi.nlm.nih.gov/31061539/
Kristensen NB, Bryrup T, Allin KH, Nielsen T, Hansen TH, Pedersen O. Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: a systematic review of randomized controlled trials. Genome Med. 2016;8(1):52. https://pubmed.ncbi.nlm.nih.gov/27159972/
Khalesi S, Bellissimo N, Vandelanotte C, Williams S, Stanley D, Irwin C. A review of probiotic supplementation in healthy adults: helpful or hype? Eur J Clin Nutr. 2019;73(1):24–37. https://pubmed.ncbi.nlm.nih.gov/29581563/
Tuohy KM, Conterno L, Gasperotti M, Viola R. Up-regulating the human intestinal microbiome using whole plant foods, polyphenols, and/or fiber. J Agric Food Chem. 2012;60(36):8776–82. https://pubmed.ncbi.nlm.nih.gov/22607578/
Scrimshaw NS, Murray EB. The acceptability of milk and milk products in populations with a high prevalence of lactose intolerance. Am J Clin Nutr. 1988;48(4 Suppl):1079–159. https://pubmed.ncbi.nlm.nih.gov/3140651/
Hertzler SR, Clancy SM. Kefir improves lactose digestion and tolerance in adults with lactose maldigestion. J Am Diet Assoc. 2003;103(5):582–7. https://pubmed.ncbi.nlm.nih.gov/12728216/
Merenstein DJ, Foster J, D’Amico F. A randomized clinical trial measuring the influence of kefir on antibiotic-associated diarrhea: the Measuring the Influence of Kefir (MILK) Study. Arch Pediatr Adolesc Med. 2009;163(8):750–4. https://pubmed.ncbi.nlm.nih.gov/19652108/
Nielsen ES, Garnås E, Jensen KJ, et al. Lacto-fermented sauerkraut improves symptoms in IBS patients independent of product pasteurisation – a pilot study. Food Funct. 2018;9(10):5323–35. https://pubmed.ncbi.nlm.nih.gov/30256365/
Kim HJ, Lim SY, Lee JS, et al. Fresh and pickled vegetable consumption and gastric cancer in Japanese and Korean populations: a meta-analysis of observational studies. Cancer Sci. 2010;101(2):508–16. https://pubmed.ncbi.nlm.nih.gov/19860848/
Morais S, Costa A, Albuquerque G, et al. Salt intake and gastric cancer: a pooled analysis within the Stomach cancer Pooling (StoP) Project. Cancer Causes Control. 2022;33(5):779–91. https://pubmed.ncbi.nlm.nih.gov/35304655/
Matsuyama S, Sawada N, Tomata Y, et al. Association between adherence to the Japanese diet and all-cause and cause-specific mortality: the Japan Public Health Center-based Prospective Study. Eur J Nutr. 2021;60(3):1327–36. https://pubmed.ncbi.nlm.nih.gov/32676701/
Ayala FR, Bauman C, Cogliati S, Lenini C, Bartolini M, Grau R. Microbial flora, probiotics, Bacillus subtilis and the search for a long and healthy human longevity. Microb Cell. 2017;4(4):133–6. https://pubmed.ncbi.nlm.nih.gov/28435840/
Katagiri R, Sawada N, Goto A, et al. Association of soy and fermented soy product intake with total and cause specific mortality: prospective cohort study. BMJ. 2020;368:m34. https://pubmed.ncbi.nlm.nih.gov/31996350/
Lo KKH, Wong AHC, Tam WWS, Ho SC. Citation classics in the nutrition and dietetics literature: 50 frequently cited articles: citation classics in nutrition and dietetics. Nutr Diet. 2016;73(4):356–68. https://doi.org/10.1111/1747-0080.12173
Shurney D, Pauly K. The gut microbiome and food as medicine: healthy microbiomes = healthy humans. Am J Health Promot. 2019;33(5):821–4. https://pubmed.ncbi.nlm.nih.gov/31120342/
Fechner A, Fenske K, Jahreis G. Effects of legume kernel fibres and citrus fibre on putative risk factors for colorectal cancer: a randomised, double-blind, crossover human intervention trial. Nutr J. 2013;12:101. https://pubmed.ncbi.nlm.nih.gov/24060277/
Wedlake L, Shaw C, McNair H, et al. Randomized controlled trial of dietary fiber for the prevention of radiation-induced gastrointestinal toxicity during pelvic radiotherapy. Am J Clin Nutr. 2017;106(3):849–57. https://pubmed.ncbi.nlm.nih.gov/28679552/
So D, Whelan K, Rossi M, et al. Dietary fiber intervention on gut microbiota composition in healthy adults: a systematic review and meta-analysis. Am J Clin Nutr. 2018;107(6):965–83. https://pubmed.ncbi.nlm.nih.gov/29757343/
Kumari M, Kozyrskyj AL. Gut microbial metabolism defines host metabolism: an emerging perspective in obesity and allergic inflammation. Obes Rev. 2017;18(1):18–31. https://pubmed.ncbi.nlm.nih.gov/27862824/
Halnes I, Baines KJ, Berthon BS, MacDonald-Wicks LK, Gibson PG, Wood LG. Soluble fibre meal challenge reduces airway inflammation and expression of GPR43 and GPR41 in asthma. Nutrients. 2017;9(1):57. https://pubmed.ncbi.nlm.nih.gov/28075383/
Kim YK, Shin C. The microbiota-gut-brain axis in neuropsychiatric disorders: pathophysiological mechanisms and novel treatments. Curr Neuropharmacol. 2018;16(5):559–73. https://pubmed.ncbi.nlm.nih.gov/28925886/
Dai Z, Niu J, Zhang Y, Jacques P, Felson DT. Dietary intake of fibre and risk of knee osteoarthritis in two US prospective cohorts [published correction appears in Ann Rheum Dis. 2017;76(12):2103]. Ann Rheum Dis. 2017;76(8):1411–9. https://pubmed.ncbi.nlm.nih.gov/28536116/
Dai Z, Lu N, Niu J, Felson DT, Zhang Y. Dietary fiber intake in relation to knee pain trajectory. Arthritis Care Res (Hoboken). 2017;69(9):1331–9. https://pubmed.ncbi.nlm.nih.gov/27899003/
Wedlake L, Shaw C, McNair H, et al. Randomized controlled trial of dietary fiber for the prevention of radiation-induced gastrointestinal toxicity during pelvic radiotherapy. Am J Clin Nutr. 2017;106(3):849–57. https://pubmed.ncbi.nlm.nih.gov/28679552/
Reynolds A, Mann J, Cummings J, Winter N, Mete E, Morenga LT. Carbohydrate quality and human health: a series of systematic reviews and meta-analyses. Lancet. 2019;393(10170):434–45. https://pubmed.ncbi.nlm.nih.gov/30638909/
Gopinath B, Flood VM, Kifley A, Louie JCY, Mitchell P. Association between carbohydrate nutrition and successful aging over 10 years. J Gerontol A Biol Sci Med Sci. 2016;71(10):1335–40. https://pubmed.ncbi.nlm.nih.gov/27252308/
Si Y, Liu X, Ye K, et al. Glucomannan hydrolysate promotes gut proliferative homeostasis and extends life span in Drosophila melanogaster. J Gerontol A Biol Sci Med Sci. 2019;74(10):1549–56. https://pubmed.ncbi.nlm.nih.gov/30252027/
McDonald P, Maizi BM, Arking R. Chemical regulation of mid– and late-life longevities in Drosophila. Exp Gerontol. 2013;48(2):240–9. https://pubmed.ncbi.nlm.nih.gov/23044027/
Reynolds A, Mann J, Cummings J, Winter N, Mete E, Morenga LT. Carbohydrate quality and human health: a series of systematic reviews and meta-analyses. Lancet. 2019;393(10170):434–45. https://pubmed.ncbi.nlm.nih.gov/30638909/
Buigues C, Fernández-Garrido J, Pruimboom L, et al. Effect of a prebiotic formulation on frailty syndrome: a randomized, double-blind clinical trial. Int J Mol Sci. 2016;17(6):E932. https://pubmed.ncbi.nlm.nih.gov/27314331/
Hippe B, Zwielehner J, Liszt K, Lassl C, Unger F, Haslberger AG. Quantification of butyryl CoA: acetate CoA-transferase genes reveals different butyrate production capacity in individuals according to diet and age. FEMS Microbiol Lett. 2011;316(2):130–5. https://pubmed.ncbi.nlm.nih.gov/21204931/
Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ, Lobley GE. Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Appl Environ Microbiol. 2007;73(4):1073–8. https://pubmed.ncbi.nlm.nih.gov/17189447/
Wu GD, Chen J, Hoffmann C, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334(6052):105–8. https://pubmed.ncbi.nlm.nih.gov/21885731/
Arumugam M, Raes J, Pelletier E, et al. Enterotypes of the human gut microbiome. Nature. 2011;473(7346):174–80. https://pubmed.ncbi.nlm.nih.gov/21508958/
Chen T, Long W, Zhang C, Liu S, Zhao L, Hamaker BR. Fiber-utilizing capacity varies in Prevotella– versus Bacteroides-dominated gut microbiota. Sci Rep. 2017;7(1):2594. https://pubmed.ncbi.nlm.nih.gov/28572676/
O’Keefe SJ, Chung D, Mahmoud N, et al. Why do African Americans get more colon cancer than Native Africans? J Nutr. 2007;137(1 Suppl):175S-82S. https://pubmed.ncbi.nlm.nih.gov/17182822/
David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–63. https://pubmed.ncbi.nlm.nih.gov/24336217/
Zhao F, Feng J, Li J, et al. Alterations of the gut microbiota in Hashimoto’s thyroiditis patients. Thyroid. 2018;28(2):175–86. https://pubmed.ncbi.nlm.nih.gov/29320965/
Tonstad S, Nathan E, Oda K, Fraser GE. Prevalence of hyperthyroidism according to type of vegetarian diet. Public Health Nutr. 2015;18(8):1482–7. https://pubmed.ncbi.nlm.nih.gov/25263477/
Precup G, Vodnar DC. Gut Prevotella as a possible biomarker of diet and its eubiotic versus dysbiotic roles: a comprehensive literature review. Br J Nutr. 2019;122(2):131–40. https://pubmed.ncbi.nlm.nih.gov/30924428/
David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–63. https://pubmed.ncbi.nlm.nih.gov/24336217/
Kumar M, Babaei P, Ji B, Nielsen J. Human gut microbiota and healthy aging: recent developments and future prospective. Nutr Healthy Aging. 4(1):3–16.; https://pubmed.ncbi.nlm.nih.gov/28035338/
Wu YT, Shen SJ, Liao KF, Huang CY. Dietary plant and animal protein sources oppositely modulate fecal Bilophila and Lachnoclostridium in vegetarians and omnivores. Microbiol Spectr. 2022;10(2):e0204721. https://pubmed.ncbi.nlm.nih.gov/35285706/
Singh RK, Chang HW, Yan D, et al. Influence of diet on the gut microbiome and implications for human health. J Transl Med. 2017;15(1):73. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5385025/
Franco-de-Moraes AC, de Almeida-Pititto B, da Rocha Fernandes G, Gomes EP, da Costa Pereira A, Ferreira SRG. Worse inflammatory profile in omnivores than in vegetarians associates with the gut microbiota composition. Diabetol Metab Syndr. 2017;9:62. https://pubmed.ncbi.nlm.nih.gov/28814977/
Kim MS, Hwang SS, Park EJ, Bae JW. Strict vegetarian diet improves the risk factors associated with metabolic diseases by modulating gut microbiota and reducing intestinal inflammation. Environ Microbiol Rep. 2013;5(5):765–75. https://pubmed.ncbi.nlm.nih.gov/24115628/
Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16(1):35–56. https://pubmed.ncbi.nlm.nih.gov/30262901/
Asnicar F, Berry SE, Valdes AM, et al. Microbiome connections with host metabolism and habitual diet from 1,098 deeply phenotyped individuals. Nat Med. 2021;27(2):321–32. https://pubmed.ncbi.nlm.nih.gov/33432175/
De Filippis F, Pellegrini N, Vannini L, et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 2016;65(11):1812–21. https://pubmed.ncbi.nlm.nih.gov/26416813/
Freeland KR, Wilson C, Wolever TM. Adaptation of colonic fermentation and glucagon-like peptide-1 secretion with increased wheat fibre intake for 1 year in hyperinsulinaemic human subjects. Br J Nutr. 2010;103(1):82–90. https://pubmed.ncbi.nlm.nih.gov/19664300/
Wu GD, Compher C, Chen EZ, et al. Comparative metabolomics in vegans and omnivores reveal constraints on diet-dependent gut microbiota metabolite production. Gut. 2016;65(1):63–72. https://pubmed.ncbi.nlm.nih.gov/25431456/
Institute of Medicine. Dietary Reference Intakes for Energy, Carbohydrate, Fiber, Fat, Fatty Acids, Cholesterol, Protein and Amino Acids. National Academy Press; 2005. https://worldcat.org/title/57373786
Jew S, Abumweis SS, Jones PJ. Evolution of the human diet: linking our ancestral diet to modern functional foods as a means of chronic disease prevention. J Med Food. 2009;12(5):925–34. https://pubmed.ncbi.nlm.nih.gov/19857053/
Leach JD, Sobolik KD. High dietary intake of prebiotic inulin-type fructans in the prehistoric Chihuahuan Desert. Br J Nutr. 2010;103(11):1558–61. https://pubmed.ncbi.nlm.nih.gov/20416127/
Institute of Medicine (U.S.). Dietary Reference Intakes: Proposed Definition of Dietary Fiber. National Academies Press; 2001. https://pubmed.ncbi.nlm.nih.gov/25057569/
Burkitt DP, Meisner P. How to manage constipation with high-fiber diet. Geriatrics. 1979;34(2):33–5 https://pubmed.ncbi.nlm.nih.gov/104901/
Requena T, Martínez-Cuesta MC, Peláez C. Diet and microbiota linked in health and disease. Food Funct. 2018;9(2):688–704. https://pubmed.ncbi.nlm.nih.gov/29410981/
Han M, Wang C, Liu P, Li D, Li Y, Ma X. Dietary fiber gap and host gut microbiota. Protein Pept Lett. 2017;24(5):388–96. https://pubmed.ncbi.nlm.nih.gov/28219317/
Venkatakrishnan A, Holzknecht ZE, Holzknecht R, et al. Evolution of bacteria in the human gut in response to changing environments: an invisible player in the game of health. Comput Struct Biotechnol J. 2021;19:752–8. https://pubmed.ncbi.nlm.nih.gov/33552447/
Hamaker BR, Cantu-Jungles TM. Discrete fiber structures dictate human gut bacteria outcomes. Trends Endocrinol Metab. 2020;31(11):803–5. https://pubmed.ncbi.nlm.nih.gov/32448722/
Tap J, Furet JP, Bensaada M, et al. Gut microbiota richness promotes its stability upon increased dietary fibre intake in healthy adults. Environ Microbiol. 2015;17(12):4954–64. https://pubmed.ncbi.nlm.nih.gov/26235304/
Walter J, Martínez I, Rose DJ. Holobiont nutrition: considering the role of the gastrointestinal microbiota in the health benefits of whole grains. Gut Microbes. 2013;4(4):340–6. https://pubmed.ncbi.nlm.nih.gov/23645316/
Toribio-Mateas M. Harnessing the power of microbiome assessment tools as part of neuroprotective nutrition and lifestyle medicine interventions. Microorganisms. 2018;6(2):35. https://pubmed.ncbi.nlm.nih.gov/29693607/
McRorie J. Clinical data support that psyllium is not fermented in the gut. Am J Gastroenterol. 2013;108(9):1541. https://pubmed.ncbi.nlm.nih.gov/24005363/
Almeida A, Mitchell AL, Boland M, et al. A new genomic blueprint of the human gut microbiota. Nature. 2019;568(7753):499–504. https://pubmed.ncbi.nlm.nih.gov/30745586/
Pereira FC, Berry D. Microbial nutrient niches in the gut. Environ Microbiol. 2017;19(4):1366–78. https://pubmed.ncbi.nlm.nih.gov/28035742/
O’Keefe SJD. The need to reassess dietary fiber requirements in healthy and critically ill patients. Gastroenterol Clin North Am. 2018;47(1):219–29. https://pubmed.ncbi.nlm.nih.gov/29413014/
Swain Ewald HA, Ewald PW. Natural selection, the microbiome, and public health. Yale J Biol Med. 2018;91(4):445–55. https://pubmed.ncbi.nlm.nih.gov/30588210/
Dewulf EM, Cani PD, Claus SP, et al. Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut. 2013;62(8):1112–21. https://pubmed.ncbi.nlm.nih.gov/23135760/
Hill P, Muir JG, Gibson PR. Controversies and recent developments of the low-FODMAP diet. Gastroenterol Hepatol (N Y). 2017;13(1):36–45. https://pubmed.ncbi.nlm.nih.gov/28420945/
Yadav BS, Sharma A, Yadav RB. Studies on effect of multiple heating/cooling cycles on the resistant starch formation in cereals, legumes and tubers. Int J Food Sci Nutr. 2009;60 Suppl 4:258–72. https://pubmed.ncbi.nlm.nih.gov/19562607/
Fernando WMU, Hill JE, Zello GA, Tyler RT, Dahl WJ, Van Kessel AG. Diets supplemented with chickpea or its main oligosaccharide component raffinose modify faecal microbial composition in healthy adults. Benef Microbes. 2010;1(2):197–207. https://pubmed.ncbi.nlm.nih.gov/21831757/
Jin S, Je Y. Nuts and legumes consumption and risk of colorectal cancer: a systematic review and meta-analysis. Eur J Epidemiol. 2022;37(6):569–85. https://pubmed.ncbi.nlm.nih.gov/35622305/
Hangen L, Bennink MR. Consumption of black beans and navy beans (Phaseolus vulgaris) reduced azoxymethane-induced colon cancer in rats. Nutr Cancer. 2002;44(1):60–5. https://pubmed.ncbi.nlm.nih.gov/12672642/
Holscher HD. Diet affects the gastrointestinal microbiota and health. J Acad Nutr Diet. 2020;120(4):495–9. https://pubmed.ncbi.nlm.nih.gov/32199522/
Venkataraman A, Sieber JR, Schmidt AW, Waldron C, Theis KR, Schmidt TM. Variable responses of human microbiomes to dietary supplementation with resistant starch. Microbiome. 2016;4(1):33. https://pubmed.ncbi.nlm.nih.gov/27357127/
Liu S, Ren F, Zhao L, et al. Starch and starch hydrolysates are favorable carbon sources for bifidobacteria in the human gut. BMC Microbiol. 2015;15:54. https://pubmed.ncbi.nlm.nih.gov/25887661/
Hellström PM, Grybäck P, Jacobsson H. The physiology of gastric emptying. Best Pract Res Clin Anaesthesiol. 2006;20(3):397–407. https://pubmed.ncbi.nlm.nih.gov/17080692/
Grundy MM, Edwards CH, Mackie AR, Gidley MJ, Butterworth PJ, Ellis PR. Re-evaluation of the mechanisms of dietary fibre and implications for macronutrient bioaccessibility, digestion and postprandial metabolism. Br J Nutr. 2016;116(5):816–33. https://pubmed.ncbi.nlm.nih.gov/27385119/
Edwards CH, Grundy MM, Grassby T, et al. Manipulation of starch bioaccessibility in wheat endosperm to regulate starch digestion, postprandial glycemia, insulinemia, and gut hormone responses: a randomized controlled trial in healthy ileostomy participants. Am J Clin Nutr. 2015;102(4):791–800. https://pubmed.ncbi.nlm.nih.gov/26333512/
Hareland G. Evaluation of flour particle size distribution by laser diffraction, sieve analysis and near-infrared reflectance spectroscopy. J Cereal Sci. 1994;20(2):183–90. https://doi.org/10.1006/jcrs.1994.1058
Holscher HD, Taylor AM, Swanson KS, Novotny JA, Baer DJ. Almond consumption and processing affects the composition of the gastrointestinal microbiota of healthy adult men and women: a randomized controlled trial. Nutrients. 2018;10(2):126. https://pubmed.ncbi.nlm.nih.gov/29373513/
McCarty MF, DiNicolantonio JJ. Acarbose, lente carbohydrate, and prebiotics promote metabolic health and longevity by stimulating intestinal production of GLP-1. Open Heart. 2015;2(1):e000205. https://pubmed.ncbi.nlm.nih.gov/25685364/
Helander HF, Fändriks L. The enteroendocrine “letter cells”—time for a new nomenclature? Scand J Gastroenterol. 2012;47(1):3–12. https://pubmed.ncbi.nlm.nih.gov/22126593/
FDA approves new drug treatment for chronic weight management, first since 2014. U.S. Food and Drug Administration. https://www.fda.gov/news-events/press-announcements/fda-approves-new-drug-treatment-chronic-weight-management-first-2014. Published June 4, 2021. Accessed February 25, 2023.; https://www.fda.gov/news-events/press-announcements/fda-approves-new-drug-treatment-chronic-weight-management-first-2014
Kumari M, Kozyrskyj AL. Gut microbial metabolism defines host metabolism: an emerging perspective in obesity and allergic inflammation. Obes Rev. 2017;18(1):18–31. https://pubmed.ncbi.nlm.nih.gov/27862824/
Freeland KR, Wolever TM. Acute effects of intravenous and rectal acetate on glucagon-like peptide-1, peptide YY, ghrelin, adiponectin and tumour necrosis factor-alpha. Br J Nutr. 2010;103(3):460–6. https://pubmed.ncbi.nlm.nih.gov/19818198/
Greenway F, O’Neil CE, Stewart L, Rood J, Keenan M, Martin R. Fourteen weeks of treatment with Viscofiber increased fasting levels of glucagon-like peptide-1 and peptide-YY. J Med Food. 2007;10(4):720–4. https://pubmed.ncbi.nlm.nih.gov/18158848/
Sandberg JC, Björck IM, Nilsson AC. Rye-based evening meals favorably affected glucose regulation and appetite variables at the following breakfast; a randomized controlled study in healthy subjects. PLoS ONE. 2016;11(3):e0151985. https://pubmed.ncbi.nlm.nih.gov/26990559/
Han Y, Xiao H. Whole food – based approaches to modulating gut microbiota and associated diseases. Annu Rev Food Sci Technol. 2020;11:119–43. https://pubmed.ncbi.nlm.nih.gov/31951487/
Kahle K, Kraus M, Scheppach W, Ackermann M, Ridder F, Richling E. Studies on apple and blueberry fruit constituents: do the polyphenols reach the colon after ingestion? Mol Nutr Food Res. 2006;50(4–5):418–23. https://pubmed.ncbi.nlm.nih.gov/16548015/
Martel J, Ojcius DM, Ko YF, Young JD. Phytochemicals as prebiotics and biological stress inducers. Trends Biochem Sci. 2020;45(6):462–71. https://pubmed.ncbi.nlm.nih.gov/32413323/
Routray W, Orsat V. Blueberries and their anthocyanins: factors affecting biosynthesis and properties. Comp Rev Food Sci and Food Saf. 2011;10(6):303–20. https://doi.org/10.1111/j.1541–4337.2011.00164.x
Hidalgo M, Oruna-Concha MJ, Kolida S, et al. Metabolism of anthocyanins by human gut microflora and their influence on gut bacterial growth. J Agric Food Chem. 2012;60:3882–90. https://pubmed.ncbi.nlm.nih.gov/22439618/
Stevenson D, Scalzo J. Anthocyanin composition and content of blueberries. J Berry Res. 2012;2(4):179–89. https://dx.doi.org/10.3233/JBR-2012-038
Vendrame S, Guglielmetti S, Riso P, Arioli S, Klimis-Zacas D, Porrini M. Six-week consumption of a wild blueberry powder drink increases bifidobacteria in the human gut. J Agric Food Chem. 2011;59(24):12815–20. https://pubmed.ncbi.nlm.nih.gov/22060186/
Shinohara K, Ohashi Y, Kawasumi K, Terada A, Fujisawa T. Effect of apple intake on fecal microbiota and metabolites in humans. Anaerobe. 2010;16(5):510–5. https://pubmed.ncbi.nlm.nih.gov/20304079/
Drasar B, Jenkins D. Bacteria, diet, and large bowel cancer. Am J Clin Nutr. 1976;29:1410–6. https://pubmed.ncbi.nlm.nih.gov/998551/
Mitsou EK, Kougia E, Nomikos T, Yannakoulia M, Mountzouris KC, Kyriacou A. Effect of banana consumption on faecal microbiota: a randomised, controlled trial. Anaerobe. 2011;17(6):384–7. https://pubmed.ncbi.nlm.nih.gov/21524710/
Del Bo’ C, Bernardi S, Cherubini A, et al. A polyphenol-rich dietary pattern improves intestinal permeability, evaluated as serum zonulin levels, in older subjects: the MaPLE randomised controlled trial. Clin Nutr. 2021;40(5):3006–18. https://pubmed.ncbi.nlm.nih.gov/33388204/
Jin JS, Touyama M, Hisada T, Benno Y. Effects of green tea consumption on human fecal microbiota with special reference to Bifidobacterium species. Microbiol Immunol. 2012;56(11):729–39. https://pubmed.ncbi.nlm.nih.gov/22924537/
Jaquet M, Rochat I, Moulin J, Cavin C, Bibiloni R. Impact of coffee consumption on the gut microbiota: a human volunteer study. Int J Food Microbiol. 2009;130(2):117–21. https://pubmed.ncbi.nlm.nih.gov/19217682/
Jaquet M, Rochat I, Moulin J, Cavin C, Bibiloni R. Impact of coffee consumption on the gut microbiota: a human volunteer study. Int J Food Microbiol. 2009;130(2):117–21. https://pubmed.ncbi.nlm.nih.gov/19217682/
Sarin S, Marya C, Nagpal R, Oberoi SS, Rekhi A. Preliminary clinical evidence of the antiplaque, antigingivitis efficacy of a mouthwash containing 2 % green tea – a randomised clinical trial. Oral Health Prev Dent. 2015;13(3):197–203. https://pubmed.ncbi.nlm.nih.gov/25610918/
Elsaie ML, Abdelhamid MF, Elsaaiee LT, Emam HM. The efficacy of topical 2 % green tea lotion in mild-to-moderate acne vulgaris. J Drugs Dermatol. 2009;8(4):358–64. https://pubmed.ncbi.nlm.nih.gov/19363854/
Ikeda S, Kanoya Y, Nagata S. Effects of a foot bath containing green tea polyphenols on interdigital tinea pedis. Foot (Edinb). 2013;23(2–3):58–62. https://pubmed.ncbi.nlm.nih.gov/23499394/
Jin JS, Touyama M, Hisada T, Benno Y. Effects of green tea consumption on human fecal microbiota with special reference to Bifidobacterium species. Microbiol Immunol. 2012;56(11):729–39. https://pubmed.ncbi.nlm.nih.gov/22924537/
Sun H, Chen Y, Cheng M, Zhang X, Zheng X, Zhang Z. The modulatory effect of polyphenols from green tea, oolong tea and black tea on human intestinal microbiota in vitro. J Food Sci Technol. 2018;55(1):399–407. https://pubmed.ncbi.nlm.nih.gov/29358833/
Ma ZJ, Wang HJ, Ma XJ, et al. Modulation of gut microbiota and intestinal barrier function during alleviation of antibiotic-associated diarrhea with Rhizoma Zingiber officinale (Ginger) extract. Food Funct. 2020;11(12):10839–51. https://pubmed.ncbi.nlm.nih.gov/33241234/
Hazan S. Rapid improvement in Alzheimer’s disease symptoms following fecal microbiota transplantation: a case report. J Int Med Res. 2020;48(6):300060520925930. https://pubmed.ncbi.nlm.nih.gov/32600151/
Marx W, Scholey A, Firth J, et al. Prebiotics, probiotics, fermented foods and cognitive outcomes: a meta-analysis of randomized controlled trials. Neurosci Biobehav Rev. 2020;118:472–84. https://pubmed.ncbi.nlm.nih.gov/32860802/
Frontiers Editorial Office. Expression of concern: effect of probiotic supplementation on cognitive function and metabolic status in Alzheimer’s disease: a randomized, double-blind and controlled trial. Front Aging Neurosci. 2020;12:602204. https://pubmed.ncbi.nlm.nih.gov/33192496/
. [Information on the investigation and handling of suspected fraudulent thesis]. Ministry of Science and Technology of the People’s Republic of China. https://www.most.gov.cn/tztg/202101/t20210121_172330.html. Published January 21, 2021. Accessed November 7, 2021.; https://www.most.gov.cn/tztg/202101/t20210121_172330.html.
Syed YY. Sodium oligomannate: first approval. Drugs. 2020;80(4):441–4. https://pubmed.ncbi.nlm.nih.gov/32020555/
Czank C, Cassidy A, Zhang Q, et al. Human metabolism and elimination of the anthocyanin, cyanidin-3-glucoside: a 13C-tracer study. Am J Clin Nutr. 2013;97(5):995–1003. https://pubmed.ncbi.nlm.nih.gov/23604435/
Djedjibegovic J, Marjanovic A, Panieri E, Saso L. Ellagic acid-derived urolithins as modulators of oxidative stress. Oxid Med Cell Longev. 2020;2020:1–15. https://pubmed.ncbi.nlm.nih.gov/32774676/
Bakkalbasi E, Mentes O, Artik N. Food ellagitannins – occurrence, effects of processing and storage. Crit Rev Food Sci Nutr. 2009;49(3):283–98. https://pubmed.ncbi.nlm.nih.gov/19093271/
Ryu D, Mouchiroud L, Andreux PA, et al. Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat Med. 2016;22(8):879–88. https://pubmed.ncbi.nlm.nih.gov/27400265/
Djedjibegovic J, Marjanovic A, Panieri E, Saso L. Ellagic acid-derived urolithins as modulators of oxidative stress. Oxid Med Cell Longev. 2020;2020:1–15. https://pubmed.ncbi.nlm.nih.gov/32774676/
Ryu D, Mouchiroud L, Andreux PA, et al. Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat Med. 2016;22(8):879–88. https://pubmed.ncbi.nlm.nih.gov/27400265/
Drummond MJ, Addison O, Brunker L, et al. Downregulation of E3 ubiquitin ligases and mitophagy-related genes in skeletal muscle of physically inactive, frail older women: a cross-sectional comparison. J Gerontol A Biol Sci Med Sci. 2014;69(8):1040–8. https://pubmed.ncbi.nlm.nih.gov/24526667/
Ryu D, Mouchiroud L, Andreux PA, et al. Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat Med. 2016;22(8):879–88. https://pubmed.ncbi.nlm.nih.gov/27400265/
Andreux PA, Blanco-Bose W, Ryu D, et al. The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans. Nat Metab. 2019;1(6):595–603. https://pubmed.ncbi.nlm.nih.gov/32694802/
Liu S, D’Amico D, Shankland E, et al. Effect of urolithin A supplementation on muscle endurance and mitochondrial health in older adults. JAMA Netw Open. 2022;5(1):e2144279. https://pubmed.ncbi.nlm.nih.gov/35050355/
Singh A, D’Amico D, Andreux PA, et al. Direct supplementation with Urolithin A overcomes limitations of dietary exposure and gut microbiome variability in healthy adults to achieve consistent levels across the population. Eur J Clin Nutr. 2022;76(2):297–308. https://pubmed.ncbi.nlm.nih.gov/34117375/
González-Sarrías A, García-Villalba R, Romo-Vaquero M, et al. Clustering according to urolithin metabotype explains the interindividual variability in the improvement of cardiovascular risk biomarkers in overweight-obese individuals consuming pomegranate: a randomized clinical trial. Mol Nutr Food Res. 2017;61(5):1600830. https://pubmed.ncbi.nlm.nih.gov/27879044/
Wu YT, Shen SJ, Liao KF, Huang CY. Dietary plant and animal protein sources oppositely modulate fecal Bilophila and Lachnoclostridium in vegetarians and omnivores. Microbiol Spectr. 2022;10(2):e0204721. https://pubmed.ncbi.nlm.nih.gov/35285706/
Alfei S, Marengo B, Zuccari G. Oxidative stress, antioxidant capabilities, and bioavailability: ellagic acid or urolithins? Antioxidants (Basel). 2020;9(8):E707. https://pubmed.ncbi.nlm.nih.gov/32759749/
Prentice AM. Starvation in humans: evolutionary background and contemporary implications. Mech Ageing Dev. 2005;126(9):976–81. https://pubmed.ncbi.nlm.nih.gov/15907972/
Wilhelmi de Toledo F, Buchinger A, Burggrabe H, et al. Fasting therapy – an expert panel update of the 2002 consensus guidelines. Forsch Komplementmed. 2013;20(6):434–43. https://pubmed.ncbi.nlm.nih.gov/24434758/
Longo VD, Mattson MP. Fasting: molecular mechanisms and clinical applications. Cell Metab. 2014;19(2):181–92. https://pubmed.ncbi.nlm.nih.gov/24440038/
Michalsen A, Li C. Fasting therapy for treating and preventing disease – current state of evidence. Forsch Komplementmed. 2013;20(6):444–53. https://pubmed.ncbi.nlm.nih.gov/24434759/
Kozubík A, Pospísil M. Protective effect of intermittent fasting on the mortality of gamma-irradiated mice. Strahlentherapie. 1982;158(12):734–8. https://pubmed.ncbi.nlm.nih.gov/6761903/
Dossey L. Longevity. Altern Ther Health Med. 2002;8(3):12–6 https://pubmed.ncbi.nlm.nih.gov/12017488/
Ziegler CC, Sidani MA. Diets for successful aging. Clin Geriatr Med. 2011;27(4):577–89. https://pubmed.ncbi.nlm.nih.gov/22062442/
Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: an update. Ageing Res Rev. 2017;39:36–45. https://pubmed.ncbi.nlm.nih.gov/27544442/
Hindhede M. The effect of food restriction during war on mortality in Copenhagen. JAMA. 1920;74(6):381–2. https://jamanetwork.com/journals/jama/article-abstract/223580
Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: an update. Ageing Res Rev. 2017;39:36–45. https://pubmed.ncbi.nlm.nih.gov/27544442/
Austad SN, Hoffman JM. Beyond calorie restriction: aging as a biological target for nutrient therapies. Curr Opin Biotechnol. 2021;70:56–60. https://pubmed.ncbi.nlm.nih.gov/33360494/
Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: an update. Ageing Res Rev. 2017;39:36–45. https://pubmed.ncbi.nlm.nih.gov/27544442/
Austad SN. Life extension by dietary restriction in the bowl and doily spider, Frontinella pyramitela. Exp Gerontol. 1989;24(1):83–92. https://pubmed.ncbi.nlm.nih.gov/2707314/
Burkewitz K, Zhang Y, Mair WB. AMPK at the nexus of energetics and aging. Cell Metab. 2014;20(1):10–25. https://pubmed.ncbi.nlm.nih.gov/24726383/
Speakman JR. Why does caloric restriction increase life and healthspan? The “clean cupboards” hypothesis. Natl Sci Rev. 2020;7(7):1153–6. https://pubmed.ncbi.nlm.nih.gov/34692140/
Mani K, Javaheri A, Diwan A. Lysosomes mediate benefits of intermittent fasting in cardiometabolic disease: the janitor is the undercover boss. Compr Physiol. 2018;8(4):1639–67. https://pubmed.ncbi.nlm.nih.gov/30215867/
Dulloo AG. Explaining the failures of obesity therapy: willpower attenuation, target miscalculation or metabolic compensation? Int J Obes (Lond). 2012;36(11):1418–20. https://pubmed.ncbi.nlm.nih.gov/23147189/
Redman LM, Heilbronn LK, Martin CK, et al. Metabolic and behavioral compensations in response to caloric restriction: implications for the maintenance of weight loss. PLoS ONE. 2009;4(2):e4377. https://pubmed.ncbi.nlm.nih.gov/19198647/
Hall KD. Metabolic adaptations to weight loss. Obesity (Silver Spring). 2018;26(5):790–1. https://pubmed.ncbi.nlm.nih.gov/29637734/
Schwartz A, Doucet E. Relative changes in resting energy expenditure during weight loss: a systematic review. Obes Rev. 2010;11(7):531–47. https://pubmed.ncbi.nlm.nih.gov/19761507/
Osborne TB, Mendel LB, Ferry EL. The effect of retardation of growth upon the breeding period and duration of life of rats. Science. 1917;45(1160):294–5. https://pubmed.ncbi.nlm.nih.gov/17760202/
Redman LM, Smith SR, Burton JH, Martin CK, Il’yasova D, Ravussin E. Metabolic slowing and reduced oxidative damage with sustained caloric restriction support the rate of living and oxidative damage theories of aging. Cell Metab. 2018;27(4):805–15.e4. https://pubmed.ncbi.nlm.nih.gov/29576535/
European hare (Lepus europaeus) longevity, ageing, and life history. Human Ageing Genomic Resources. https://genomics.senescence.info/species/entry.php?species=Lepus_europaeus. Published October 14, 2017. December 9, 2022.; https://genomics.senescence.info/species/entry.php?species=Lepus_europaeus
Galapagos tortoise (Chelonoidis nigra) longevity, ageing, and life history. Human Ageing Genomic Resources. https://genomics.senescence.info/species/entry.php?species=Chelonoidis_nigra. Published 2018. Accessed December 9, 2022.; https://genomics.senescence.info/species/entry.php?species=Chelonoidis_nigra
Civitarese AE, Carling S, Heilbronn LK, et al. Calorie restriction increases muscle mitochondrial biogenesis in healthy humans. PLoS MED. 2007;4(3):e76. https://pubmed.ncbi.nlm.nih.gov/17341128/
Redman LM, Smith SR, Burton JH, Martin CK, Il’yasova D, Ravussin E. Metabolic slowing and reduced oxidative damage with sustained caloric restriction support the rate of living and oxidative damage theories of aging. Cell Metab. 2018;27(4):805–15.e4. https://pubmed.ncbi.nlm.nih.gov/29576535/
Tamakoshi A, Tamakoshi K, Lin Y, Yagyu K, Kikuchi S, JACC Study Group. Healthy lifestyle and preventable death: findings from the Japan Collaborative Cohort (JACC) Study. Prev Med. 2009;48(5):486–92. https://pubmed.ncbi.nlm.nih.gov/19254743/
Bourzac K. Interventions: live long and prosper. Nature. 2012;492(7427):S18–20. https://pubmed.ncbi.nlm.nih.gov/23222670/
Rebrin I, Forster MJ, Sohal RS. Association between life-span extension by caloric restriction and thiol redox state in two different strains of mice. Free Radic Biol Med. 2011;51(1):225–33. https://pubmed.ncbi.nlm.nih.gov/21530646/
Life expectancy. Centers for Disease Control and Prevention. https://www.cdc.gov/nchs/fastats/life-expectancy.htm. Updated September 6, 2022. Accessed December 9, 2022.; https://www.cdc.gov/nchs/fastats/life-expectancy.htm
Speakman JR, Hambly C. Starving for life: what animal studies can and cannot tell us about the use of caloric restriction to prolong human lifespan. J Nutr. 2007;137(4):1078–86. https://pubmed.ncbi.nlm.nih.gov/17374682/
Speakman JR, Hambly C. Starving for life: what animal studies can and cannot tell us about the use of caloric restriction to prolong human lifespan. J Nutr. 2007;137(4):1078–86. https://pubmed.ncbi.nlm.nih.gov/17374682/
Liao CY, Rikke BA, Johnson TE, Diaz V, Nelson JF. Genetic variation in the murine lifespan response to dietary restriction: from life extension to life shortening. Aging Cell. 2010;9(1):92–5. https://pubmed.ncbi.nlm.nih.gov/21388497/
Wolf AM. Rodent diet aids and the fallacy of caloric restriction. Mech Ageing Dev. 2021;200:111584. https://pubmed.ncbi.nlm.nih.gov/34673082/
Weichhart T. mTOR as regulator of lifespan, aging, and cellular senescence: a mini-review. Gerontology. 2018;64(2):127–34. https://pubmed.ncbi.nlm.nih.gov/29190625/
Ingram DK, de Cabo R. Calorie restriction in rodents: caveats to consider. Ageing Res Rev. 2017;39:15–28. https://pubmed.ncbi.nlm.nih.gov/28610949/
Lawler DF, Larson BT, Ballam JM, et al. Diet restriction and ageing in the dog: major observations over two decades. Br J Nutr. 2008;99(4):793–805. https://pubmed.ncbi.nlm.nih.gov/18062831/
Fryar CD, Carroll MD, Afful J. Prevalence of overweight, obesity, and extreme obesity among adults aged 20 and over: United States, 1960–1962 through 2017–2018. Centers for Disease Control and Prevention. https://www.cdc.gov/nchs/data/hestat/obesity-adult-17–18/overweight-obesity-adults-H.pdf. Published December 2020. Accessed November 22, 2022.; https://www.cdc.gov/nchs/data/hestat/obesity-adult-17-18/obesity-adult.htm
Grover SA, Kaouache M, Rempel P, et al. Years of life lost and healthy life-years lost from diabetes and cardiovascular disease in overweight and obese people: a modelling study. Lancet Diabetes Endocrinol. 2015;3(2):114–22. https://pubmed.ncbi.nlm.nih.gov/25483220/
Bodkin NL, Alexander TM, Ortmeyer HK, Johnson E, Hansen BC. Mortality and morbidity in laboratory-maintained rhesus monkeys and effects of long-term dietary restriction. J Gerontol A Biol Sci Med Sci. 2003;58(3):212–9. https://pubmed.ncbi.nlm.nih.gov/12634286/
Colman RJ, Beasley TM, Kemnitz JW, Johnson SC, Weindruch R, Anderson RM. Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys. Nat Commun. 2014;5(1):3557. https://pubmed.ncbi.nlm.nih.gov/24691430/
Mattison JA, Roth GS, Beasley TM, et al. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature. 2012;489(7415):318–21. https://pubmed.ncbi.nlm.nih.gov/22932268/
Pifferi F, Terrien J, Marchal J, et al. Caloric restriction increases lifespan but affects brain integrity in grey mouse lemur primates. Commun Biol. 2018;1:30. https://pubmed.ncbi.nlm.nih.gov/30271916/
Le Bourg E. Dietary restriction studies in humans: focusing on obesity, forgetting longevity. Gerontology. 2012;58(2):126–8. https://pubmed.ncbi.nlm.nih.gov/21701153/
Harbottle EJ, Birmingham CL, Sayani F. Anorexia nervosa: a survival analysis. Eat Weight Disord. 2008;13(2):e32–4. https://pubmed.ncbi.nlm.nih.gov/18612251/
Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: an update. Ageing Res Rev. 2017;39:36–45. https://pubmed.ncbi.nlm.nih.gov/27544442/
Harbottle EJ, Birmingham CL, Sayani F. Anorexia nervosa: a survival analysis. Eat Weight Disord. 2008;13(2):e32–4. https://pubmed.ncbi.nlm.nih.gov/18612251/
Machado PPP, Grilo CM, Crosby RD. Evaluation of the DSM-5 severity indicator for anorexia nervosa. Eur Eat Disord Rev. 2017;25(3):221–3. https://pubmed.ncbi.nlm.nih.gov/28402070/
Fryar CD, Kruszon-Moran D, Gu Q, Ogden CL. Mean body weight, height, waist circumference, and body mass index among adults: United States, 1999–2000 through 2015–2016. Natl Health Stat Report. 2018;(122):1–16. https://pubmed.ncbi.nlm.nih.gov/30707668/
Mehler PS, Koutsavlis A. Anorexia nervosa, albumin, and inflammation. Am J Med. 2021;134(6):e401. https://pubmed.ncbi.nlm.nih.gov/34049636/
Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients with anorexia nervosa and other eating disorders: a meta-analysis of 36 studies. Arch Gen Psychiatry. 2011;68(7):724–31. https://pubmed.ncbi.nlm.nih.gov/21727255/
Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: an update. Ageing Res Rev. 2017;39:36–45. https://pubmed.ncbi.nlm.nih.gov/27544442/
Lee MB, Hill CM, Bitto A, Kaeberlein M. Antiaging diets: separating fact from fiction. Science. 2021;374(6570):eabe7365. https://pubmed.ncbi.nlm.nih.gov/34793210/
Most J, Gilmore LA, Smith SR, Han H, Ravussin E, Redman LM. Significant improvement in cardiometabolic health in healthy nonobese individuals during caloric restriction-induced weight loss and weight loss maintenance. Am J Physiol Endocrinol Metab. 2018;314(4):E396–405. https://pubmed.ncbi.nlm.nih.gov/29351490/
Anderson RM, Le Couteur DG, de Cabo R. Caloric restriction research: new perspectives on the biology of aging. J Gerontol A Biol Sci Med Sci. 2017;73(1):1–3. https://pubmed.ncbi.nlm.nih.gov/29240911/
Richardson A, Austad SN, Ikeno Y, Unnikrishnan A, McCarter RJ. Significant life extension by ten percent dietary restriction. Ann N Y Acad Sci. 2016;1363(1):11–7. https://pubmed.ncbi.nlm.nih.gov/26695614/
Dirks AJ, Leeuwenburgh C. Caloric restriction in humans: potential pitfalls and health concerns. Mech Ageing Dev. 2006;127(1):1–7. https://pubmed.ncbi.nlm.nih.gov/16226298/
Martin CK, Bhapkar M, Pittas AG, et al. Effect of calorie restriction on mood, quality of life, sleep, and sexual function in healthy nonobese adults: the CALERIE 2 randomized clinical trial. JAMA Intern Med. 2016;176(6):743–52. https://pubmed.ncbi.nlm.nih.gov/27136347/
Most J, Gilmore LA, Smith SR, Han H, Ravussin E, Redman LM. Significant improvement in cardiometabolic health in healthy nonobese individuals during caloric restriction-induced weight loss and weight loss maintenance. Am J Physiol Endocrinol Metab. 2018;314(4):E396–405. https://pubmed.ncbi.nlm.nih.gov/29351490/
Ravussin E, Redman LM, Rochon J, et al. A 2-year randomized controlled trial of human caloric restriction: feasibility and effects on predictors of health span and longevity. J Gerontol A Biol Sci Med Sci. 2015;70(9):1097–104. https://pubmed.ncbi.nlm.nih.gov/26187233/
Das SK, Roberts SB, Bhapkar MV, et al. Body-composition changes in the Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy (CALERIE)-2 study: a 2-y randomized controlled trial of calorie restriction in nonobese humans. Am J Clin Nutr. 2017;105(4):913–27. https://pubmed.ncbi.nlm.nih.gov/28228420/
Franklin JC, Scheile BC. Observations on human behavior in experimental semi-starvation and rehabilitation. J Clin Psychol. 1948;4(1):28–45. https://pubmed.ncbi.nlm.nih.gov/18903450/
Dulloo AG, Jacquet J, Montani JP. How dieting makes some fatter: from a perspective of human body composition autoregulation. Proc Nutr Soc. 2012;71(3):379–89. https://pubmed.ncbi.nlm.nih.gov/22475574/
Redman LM, Smith SR, Burton JH, Martin CK, Il’yasova D, Ravussin E. Metabolic slowing and reduced oxidative damage with sustained caloric restriction support the rate of living and oxidative damage theories of aging. Cell Metab. 2018;27(4):805–15.e4. https://pubmed.ncbi.nlm.nih.gov/29576535/
Marlatt KL, Redman LM, Burton JH, Martin CK, Ravussin E. Persistence of weight loss and acquired behaviors 2 y after stopping a 2-y calorie restriction intervention. Am J Clin Nutr. 2017;105(4):928–35. https://pubmed.ncbi.nlm.nih.gov/28275127/
Dorling JL, van Vliet S, Huffman KM, et al. Effects of caloric restriction on human physiological, psychological, and behavioral outcomes: highlights from CALERIE phase 2. Nutr Rev. 2020;79(1):98–113. https://pubmed.ncbi.nlm.nih.gov/32940695/
Kahathuduwa CN, Binks M, Martin CK, Dawson JA. Extended calorie restriction suppresses overall and specific food cravings: a systematic review and a meta-analysis. Obes Rev. 2017;18(10):1122–35. https://pubmed.ncbi.nlm.nih.gov/28557246/
Redman LM, Smith SR, Burton JH, Martin CK, Il’yasova D, Ravussin E. Metabolic slowing and reduced oxidative damage with sustained caloric restriction support the rate of living and oxidative damage theories of aging. Cell Metab. 2018;27(4):805–15.e4. https://pubmed.ncbi.nlm.nih.gov/29576535/
Il’yasova D, Fontana L, Bhapkar M, et al. Effects of 2 years of caloric restriction on oxidative status assessed by urinary F2-isoprostanes: the CALERIE 2 randomized clinical trial. Aging Cell. 2018;17(2):e12719. https://pubmed.ncbi.nlm.nih.gov/29424490/
Omodei D, Licastro D, Salvatore F, Crosby SD, Fontana L. Serum from humans on long-term calorie restriction enhances stress resistance in cell culture. Aging (Albany NY). 2013;5(8):599–606. https://pubmed.ncbi.nlm.nih.gov/23912304/
Dorling JL, van Vliet S, Huffman KM, et al. Effects of caloric restriction on human physiological, psychological, and behavioral outcomes: highlights from CALERIE phase 2. Nutr Rev. 2020;79(1):98–113. https://pubmed.ncbi.nlm.nih.gov/32940695/
Dorling JL, van Vliet S, Huffman KM, et al. Effects of caloric restriction on human physiological, psychological, and behavioral outcomes: highlights from CALERIE phase 2. Nutr Rev. 2020;79(1):98–113. https://pubmed.ncbi.nlm.nih.gov/32940695/
Block G, Dresser CM, Hartman AM, Carroll MD. Nutrient sources in the American diet: quantitative data from the NHANES II survey. II. Macronutrients and fats. Am J Epidemiol. 1985;122(1):13–26. https://pubmed.ncbi.nlm.nih.gov/4014199/
Dorling JL, van Vliet S, Huffman KM, et al. Effects of caloric restriction on human physiological, psychological, and behavioral outcomes: highlights from CALERIE phase 2. Nutr Rev. 2020;79(1):98–113. https://pubmed.ncbi.nlm.nih.gov/32940695/
Tang X, Zhao W, Lu M, et al. Relationship between central obesity and the incidence of cognitive impairment and dementia from cohort studies involving 5,060,687 participants. Neurosci Biobehav Rev. 2021;130:301–13. https://pubmed.ncbi.nlm.nih.gov/34464646/
Yu Q, Zou L, Kong Z, Yang L. Cognitive impact of calorie restriction: a narrative review. J Am Med Dir Assoc. 2020;21(10):1394–401. https://pubmed.ncbi.nlm.nih.gov/32693996/
Phillips MCL. Fasting as a therapy in neurological disease. Nutrients. 2019;11(10):2501. https://pubmed.ncbi.nlm.nih.gov/31627405/
Lü W, Yu T, Kuang W. Effects of dietary restriction on cognitive function: a systematic review and meta-analysis. Nutr Neurosci. Published online April 25, 2022:1–11.; https://pubmed.ncbi.nlm.nih.gov/35469542/
Lü W, Yu T, Kuang W. Effects of dietary restriction on cognitive function: a systematic review and meta-analysis. Nutr Neurosci. Published online April 25, 2022:1–11.; https://pubmed.ncbi.nlm.nih.gov/35469542/
Witte AV, Fobker M, Gellner R, Knecht S, Flöel A. Caloric restriction improves memory in elderly humans. Proc Natl Acad Sci U S A. 2009;106(4):1255–60. https://pubmed.ncbi.nlm.nih.gov/19171901/
Leclerc E, Trevizol AP, Grigolon RB, et al. The effect of caloric restriction on working memory in healthy non-obese adults. CNS Spectr. 2020;25(1):2–8. https://pubmed.ncbi.nlm.nih.gov/30968820/
Lindseth GN, Lindseth PD, Jensen WC, Petros TV, Helland BD, Fossum DL. Dietary effects on cognition and pilots’ flight performance. Int J Aviat Psychol. 2011;21(3):269–82. https://pubmed.ncbi.nlm.nih.gov/29353985/
Benau EM, Orloff NC, Janke EA, Serpell L, Timko CA. A systematic review of the effects of experimental fasting on cognition. Appetite. 2014;77:52–61. https://pubmed.ncbi.nlm.nih.gov/24583414/
Zajac I, Herreen D, Hunkin H, et al. Modified fasting compared to true fasting improves blood glucose levels and subjective experiences of hunger, food cravings and mental fatigue, but not cognitive function: results of an acute randomised cross-over trial. Nutrients. 2020;13(1):E65. https://pubmed.ncbi.nlm.nih.gov/33379191/
Lieberman HR, Caruso CM, Niro PJ, et al. A double-blind, placebo-controlled test of 2 d of calorie deprivation: effects on cognition, activity, sleep, and interstitial glucose concentrations. Am J Clin Nutr. 2008;88(3):667–76. https://pubmed.ncbi.nlm.nih.gov/18779282/
Dar BA, Dar MA, Bashir S. Calorie restriction the fountain of youth. Food Nutr Sci. 2012;3(11):1522–6. https://www.scirp.org/journal/paperinformation.aspx?paperid=24485
Dirks AJ, Leeuwenburgh C. Caloric restriction in humans: potential pitfalls and health concerns. Mech Ageing Dev. 2006;127(1):1–7. https://pubmed.ncbi.nlm.nih.gov/16226298/
Hunt ND, Li GD, Zhu M, et al. Effect of calorie restriction and refeeding on skin wound healing in the rat. Age (Dordr). 2012;34(6):1453–8. https://pubmed.ncbi.nlm.nih.gov/22037865/
Reed MJ, Penn PE, Li Y, et al. Enhanced cell proliferation and biosynthesis mediate improved wound repair in refed, caloric-restricted mice. Mech Ageing Dev. 1996;89(1):21–43. https://pubmed.ncbi.nlm.nih.gov/8819104/
Bergendahl M, Vance ML, Iranmanesh A, Thorner MO, Veldhuis JD. Fasting as a metabolic stress paradigm selectively amplifies cortisol secretory burst mass and delays the time of maximal nyctohemeral cortisol concentrations in healthy men. J Clin Endocrinol Metab. 1996;81(2):692–9. https://pubmed.ncbi.nlm.nih.gov/8636290/
Nakamura Y, Walker BR, Ikuta T. Systematic review and meta-analysis reveals acutely elevated plasma cortisol following fasting but not less severe calorie restriction. Stress. 2016;19(2):151–7. https://pubmed.ncbi.nlm.nih.gov/26586092/
Speakman JR, Hambly C. Starving for life: what animal studies can and cannot tell us about the use of caloric restriction to prolong human lifespan. J Nutr. 2007;137(4):1078–86. https://pubmed.ncbi.nlm.nih.gov/17374682/
Mattison JA, Roth GS, Beasley TM, et al. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature. 2012;489(7415):318–21. https://pubmed.ncbi.nlm.nih.gov/22932268/
Sun D, Muthukumar AR, Lawrence RA, Fernandes G. Effects of calorie restriction on polymicrobial peritonitis induced by cecum ligation and puncture in young C57BL/6 mice. Clin Diagn Lab Immunol. 2001;8(5):1003–11. https://pubmed.ncbi.nlm.nih.gov/11527818/
Gardner EM, Beli E, Clinthorne JF, Duriancik DM. Energy intake and response to infection with influenza. Annu Rev Nutr. 2011;31:353–67. https://pubmed.ncbi.nlm.nih.gov/21548773/
Barbosa ASAA, Diório SM, Pedrini SCB, et al. Nutritional status and immune response in murine experimental Jorge Lobo’s disease. Mycoses. 2015;58(9):522–30. https://pubmed.ncbi.nlm.nih.gov/26156007/
Kristan DM. Chronic calorie restriction increases susceptibility of laboratory mice (Mus musculus) to a primary intestinal parasite infection. Aging Cell. 2007;6(6):817–25. https://pubmed.ncbi.nlm.nih.gov/17973970/
Clinthorne JF, Adams DJ, Fenton JI, Ritz BW, Gardner EM. Short-term re-feeding of previously energy-restricted C57BL/6 male mice restores body weight and body fat and attenuates the decline in natural killer cell function after primary influenza infection. J Nutr. 2010;140(8):1495–501. https://pubmed.ncbi.nlm.nih.gov/20534876/
Clinthorne JF, Adams DJ, Fenton JI, Ritz BW, Gardner EM. Short-term re-feeding of previously energy-restricted C57BL/6 male mice restores body weight and body fat and attenuates the decline in natural killer cell function after primary influenza infection. J Nutr. 2010;140(8):1495–501. https://pubmed.ncbi.nlm.nih.gov/20534876/
Nicoll R, Henein MY. Caloric restriction and its effect on blood pressure, heart rate variability and arterial stiffness and dilatation: a review of the evidence. Int J Mol Sci. 2018;19(3):751. https://pubmed.ncbi.nlm.nih.gov/29518898/
Florian JP, Baisch FJ, Heer M, Pawelczyk JA. Caloric restriction decreases orthostatic tolerance independently from 6° head-down bedrest. PLoS ONE. 2015;10(4):e0118812. https://pubmed.ncbi.nlm.nih.gov/25915488/
Lu CC, Diedrich A, Tung CS, et al. Water ingestion as prophylaxis against syncope. Circulation. 2003;108(21):2660–5. https://pubmed.ncbi.nlm.nih.gov/14623807/
Racette SB, Rochon J, Uhrich ML, et al. Effects of two years of calorie restriction on aerobic capacity and muscle strength. Med Sci Sports Exerc. 2017;49(11):2240–9. https://pubmed.ncbi.nlm.nih.gov/29045325/
Cava E, Yeat NC, Mittendorfer B. Preserving healthy muscle during weight loss. Adv Nutr. 2017;8(3):511–9. https://pubmed.ncbi.nlm.nih.gov/28507015/
Smith GI, Yoshino J, Kelly SC, et al. High-protein intake during weight loss therapy eliminates the weight-loss-induced improvement in insulin action in obese postmenopausal women. Cell Rep. 2016;17(3):849–61. https://pubmed.ncbi.nlm.nih.gov/27732859/
Sardeli AV, Komatsu TR, Mori MA, Gáspari AF, Chacon-Mikahil MPT. Resistance training prevents muscle loss induced by caloric restriction in obese elderly individuals: a systematic review and meta-analysis. Nutrients. 2018;10(4):423. https://pubmed.ncbi.nlm.nih.gov/29596307/
Villareal DT, Fontana L, Weiss EP, et al. Bone mineral density response to caloric restriction-induced weight loss or exercise-induced weight loss: a randomized controlled trial. Arch Intern Med. 2006;166(22):2502–10. https://pubmed.ncbi.nlm.nih.gov/17159017/
Romashkan SV, Das SK, Villareal DT, et al. Safety of two-year caloric restriction in non-obese healthy individuals. Oncotarget. 2016;7(15):19124–33. https://pubmed.ncbi.nlm.nih.gov/26992237/
Villareal DT, Kotyk JJ, Armamento-Villareal RC, et al. Reduced bone mineral density is not associated with significantly reduced bone quality in men and women practicing long-term calorie restriction with adequate nutrition. Aging Cell. 2011;10(1):96–102. https://pubmed.ncbi.nlm.nih.gov/20969721/
Lee YM, Kim KS, Jacobs DR, Lee DH. Persistent organic pollutants in adipose tissue should be considered in obesity research. Obes Rev. 2017;18(2):129–39. https://pubmed.ncbi.nlm.nih.gov/27911986/
Vaz R, Slorach SA, Hofvander Y. Organochlorine contaminants in Swedish human milk: studies conducted at the National Food Administration 1981–1990. Food Addit Contam. 1993;10(4):407–18. https://pubmed.ncbi.nlm.nih.gov/8405580/
Jandacek RJ, Heubi JE, Buckley DD, et al. Reduction of the body burden of PCBs and DDE by dietary intervention in a randomized trial. J Nutr Biochem. 2014;25(4):483–8. https://pubmed.ncbi.nlm.nih.gov/24629911/
Sera N, Morita K, Nagasoe M, Tokieda H, Kitaura T, Tokiwa H. Binding effect of polychlorinated compounds and environmental carcinogens on rice bran fiber. J Nutr Biochem. 2005;16(1):50–8. https://pubmed.ncbi.nlm.nih.gov/15629241/
Tufan F, Soyluk O, Karan MA. Healthy behaviors potentially due to calorie restriction. JAMA Intern Med. 2016;176(11):1724. https://pubmed.ncbi.nlm.nih.gov/27820642/
Villareal DT, Fontana L, Das SK, et al. Effect of two-year caloric restriction on bone metabolism and bone mineral density in non-obese younger adults: a randomized clinical trial. J Bone Miner Res. 2016;31(1):40–51. https://pubmed.ncbi.nlm.nih.gov/26332798/
Khong TK, Kimpton J. Moderate calorie restriction improves cardiometabolic risk factors in healthy individuals. Drug Ther Bull. 2020;58(9):135–6. https://pubmed.ncbi.nlm.nih.gov/32843424/
Key T, Davey G. Prevalence of obesity is low in people who do not eat meat. BMJ. 1996;313(7060):816–7. https://pubmed.ncbi.nlm.nih.gov/8842088/
Kennedy ET, Bowman SA, Spence JT, Freedman M, King J. Popular diets: correlation to health, nutrition, and obesity. J Am Diet Assoc. 2001;101(4):411–20. https://pubmed.ncbi.nlm.nih.gov/11320946/
Rizzo NS, Jaceldo-Siegl K, Sabate J, Fraser GE. Nutrient profiles of vegetarian and nonvegetarian dietary patterns. J Acad Nutr Diet. 2013;113(12):1610–9. https://pubmed.ncbi.nlm.nih.gov/23988511/
Sinclair DA, LaPlante MD. Lifespan: The Revolutionary Science of Why We Age – and Why We Don’t Have To. Atria Books; 2019. https://worldcat.org/title/1121458577
Willcox DC, Willcox BJ, Todoriki H, Curb JD, Suzuki M. Caloric restriction and human longevity: what can we learn from the Okinawans? Biogerontology. 2006;7(3):173–7. https://pubmed.ncbi.nlm.nih.gov/16810568/
Kouda K, Iki M. Beneficial effects of mild stress (hormetic effects): dietary restriction and health. J Physiol Anthropol. 2010;29(4):127–32. https://pubmed.ncbi.nlm.nih.gov/20686325/
Twain M. My Debut As A Literary Person: With Other Essays and Stories. American Pub Co; 1903. https://worldcat.org/title/54203287
Twain M. A Connecticut Yankee in King Arthur’s Court. Charles L. Webster and Co; 1889. https://worldcat.org/title/1110204204
Harris L, McGarty A, Hutchison L, Ells L, Hankey C. Short-term intermittent energy restriction interventions for weight management: a systematic review and meta-analysis. Obes Rev. 2018;19(1):1–13. https://pubmed.ncbi.nlm.nih.gov/28975722/
Trepanowski JF, Kroeger CM, Barnosky A, et al. Effect of alternate-day fasting on weight loss, weight maintenance, and cardioprotection among metabolically healthy obese adults: a randomized clinical trial. JAMA Intern Med. 2017;177(7):930–8. https://pubmed.ncbi.nlm.nih.gov/28459931/
Arguin H, Dionne IJ, Sénéchal M, et al. Short– and long-term effects of continuous versus intermittent restrictive diet approaches on body composition and the metabolic profile in overweight and obese postmenopausal women: a pilot study. Menopause. 2012;19(8):870–6. https://pubmed.ncbi.nlm.nih.gov/22735163/
Corley BT, Carroll RW, Hall RM, Weatherall M, Parry-Strong A, Krebs JD. Intermittent fasting in type 2 diabetes mellitus and the risk of hypoglycaemia: a randomized controlled trial. Diabet Med. 2018;35(5):588–94. https://pubmed.ncbi.nlm.nih.gov/29405359/
Lammers LA, Achterbergh R, de Vries EM, et al. Short-term fasting alters cytochrome P450-mediated drug metabolism in humans. Drug Metab Dispos. 2015;43(6):819–28. https://pubmed.ncbi.nlm.nih.gov/25795462/
de Cabo R, Mattson MP. Effects of intermittent fasting on health, aging, and disease. N Engl J Med. 2019;381(26):2541–51. https://pubmed.ncbi.nlm.nih.gov/31881139/
da Luz FQ, Hay P, Gibson AA, et al. Does severe dietary energy restriction increase binge eating in overweight or obese individuals? A systematic review. Obes Rev. 2015;16(8):652–65. https://pubmed.ncbi.nlm.nih.gov/26094791/
Horne BD, May HT, Anderson JL, et al. Usefulness of routine periodic fasting to lower risk of coronary artery disease among patients undergoing coronary angiography. Am J Cardiol. 2008;102(7):814–9. https://pubmed.ncbi.nlm.nih.gov/18805103/
Enstrom JE. Cancer and total mortality among active Mormons. Cancer. 1978;42(4):1943–51. https://pubmed.ncbi.nlm.nih.gov/709540/
Horne BD, Muhlestein JB, May HT, et al. Relation of routine, periodic fasting to risk of diabetes mellitus, and coronary artery disease in patients undergoing coronary angiography. Am J Cardiol. 2012;109(11):1558–62. https://pubmed.ncbi.nlm.nih.gov/22425331/
Bartholomew CL, Muhlestein JB, Anderson JL, et al. Association of periodic fasting lifestyles with survival and incident major adverse cardiovascular events in patients undergoing cardiac catheterization. Eur J Prev Cardiol. 2022;28(16):1774–81. https://pubmed.ncbi.nlm.nih.gov/33624026/
Tsaban G. Routine periodic fasting reduces all-cause mortality and heart failure incidence: new insights on old habits. Eur J Prev Cardiol. 2022;28(16):1782–3. https://pubmed.ncbi.nlm.nih.gov/33624012/
Enstrom JE. Cancer and total mortality among active Mormons. Cancer. 1978;42(4):1943–51. https://pubmed.ncbi.nlm.nih.gov/709540/
The Doctrine and Covenants of The Church of Jesus Christ of Latter-day Saints. Section 89:12. The Church of Jesus Christ of Latter-day Saints; 2013. https://www.churchofjesuschrist.org/study/scriptures/dc-testament/dc/89?lang=eng. Accessed December 9 https://www.churchofjesuschrist.org/study/scriptures/dc-testament/dc/89?lang=eng
Vallejo EA. La dieta de hambre a dias alternos en la alimentacion de los viejos. Rev Clin Esp. 1956;63:25–7. https://pubmed.ncbi.nlm.nih.gov/13420784/
Stunkard AJ. Nutrition, aging and obesity: a critical review of a complex relationship. Int J Obes. 1983;7(3):201–20. https://pubmed.ncbi.nlm.nih.gov/6885229/
Brandhorst S, Choi IY, Wei M, et al. A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metab. 2015;22(1):86–99. https://pubmed.ncbi.nlm.nih.gov/26094889/
Abbasi J. Can a diet that mimics fasting turn back the clock? JAMA. 2017;318(3):227–9. https://pubmed.ncbi.nlm.nih.gov/28658487/
Wei M, Brandhorst S, Shelehchi M, et al. Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease. Sci Transl Med. 2017;9(377):8700. https://pubmed.ncbi.nlm.nih.gov/28202779/
Gill S, Panda S. A smartphone app reveals erratic diurnal eating patterns in humans that can be modulated for health benefits. Cell Metab. 2015;22(5):789–98. https://pubmed.ncbi.nlm.nih.gov/26411343/
Anton SD, Moehl K, Donahoo WT, et al. Flipping the metabolic switch: understanding and applying the health benefits of fasting. Obesity (Silver Spring). 2018;26(2):254–68. https://pubmed.ncbi.nlm.nih.gov/29086496/
Sutton EF, Beyl R, Early KS, Cefalu WT, Ravussin E, Peterson CM. Early time-restricted feeding improves insulin sensitivity, blood pressure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab. 2018;27(6):1212–21.e3. https://pubmed.ncbi.nlm.nih.gov/29754952/
Schloss J, Steel A. Medical synopsis: nightly fasting may assist breast cancer patients and other people with cancer. Adv Integr Med. 2016;3(2):66–7. https://www.sciencedirect.com/science/article/pii/S2212958816300738?via%3Dihub
Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161(13):1645–52. https://pubmed.ncbi.nlm.nih.gov/11434797/
Currenti W, Godos J, Castellano S, et al. Association between time restricted feeding and cognitive status in older Italian adults. Nutrients. 2021;13(1):E191. https://pubmed.ncbi.nlm.nih.gov/33435416/
Mayer J. Should you starve yourself thin? Family Health/Today’s Health. February, 1977.; https://agris.fao.org/agris-search/search.do?recordID=US201302986920
Keys A. Caloric undernutrition and starvation, with notes on protein deficiency. J Am Med Assoc. 1948;138(7):500–11. https://pubmed.ncbi.nlm.nih.gov/18884888/
Michalsen A, Li C. Fasting therapy for treating and preventing disease – current state of evidence. Forsch Komplementmed. 2013;20(6):444–53. https://pubmed.ncbi.nlm.nih.gov/24434759/
Schnitker MA, Mattman PE, Bliss TL. A clinical study of malnutrition in Japanese prisoners of war. Ann Intern Med. 1951;35(1):69–96. https://pubmed.ncbi.nlm.nih.gov/14847450/
Boateng AA, Sriram K, Meguid MM, Crook M. Refeeding syndrome: treatment considerations based on collective analysis of literature case reports. Nutrition. 2010;26(2):156–67. https://pubmed.ncbi.nlm.nih.gov/20122539/
Finnell JS, Saul BC, Goldhamer AC, Myers TR. Is fasting safe? A chart review of adverse events during medically supervised, water-only fasting. BMC Complement Altern Med. 2018;18:67. https://pubmed.ncbi.nlm.nih.gov/29458369/
Michalsen A, Li C. Fasting therapy for treating and preventing disease – current state of evidence. Forsch Komplementmed. 2013;20(6):444–53. https://pubmed.ncbi.nlm.nih.gov/24434759/
Runcie J, Thomson TJ. Prolonged starvation – a dangerous procedure? Br Med J. 1970;3(5720):432–5. https://pubmed.ncbi.nlm.nih.gov/5454322/
Sailaja BS, He XC, Li L. Stem cells matter in response to fasting. Cell Rep. 2015;13(11):2325–6. https://pubmed.ncbi.nlm.nih.gov/26705824/
Raffaghello L, Lee C, Safdie FM, et al. Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy. Proc Natl Acad Sci U S A. 2008;105(24):8215–20. https://pubmed.ncbi.nlm.nih.gov/18378900/
Dorff TB, Groshen S, Garcia A, et al. Safety and feasibility of fasting in combination with platinum-based chemotherapy. BMC Cancer. 2016;16:360. https://pubmed.ncbi.nlm.nih.gov/27282289/
Nencioni A, Caffa I, Cortellino S, Longo VD. Fasting and cancer: molecular mechanisms and clinical application. Nat Rev Cancer. 2018;18(11):707–19. https://pubmed.ncbi.nlm.nih.gov/30327499/
Lugtenberg RT, de Groot S, Kaptein AA, et al. Quality of life and illness perceptions in patients with breast cancer using a fasting mimicking diet as an adjunct to neoadjuvant chemotherapy in the phase 2 DIRECT (BOOG 2013–14) trial. Breast Cancer Res Treat. 2021;185(3):741–58. https://pubmed.ncbi.nlm.nih.gov/33179154/
Caffa I, Spagnolo V, Vernieri C, et al. Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature. 2020;583(7817):620–4. https://pubmed.ncbi.nlm.nih.gov/32669709/
de Groot S, Lugtenberg RT, Cohen D, et al. Fasting mimicking diet as an adjunct to¿neoadjuvant chemotherapy for breast cancer in the multicentre randomized phase 2 DIRECT trial. Nat Commun. 2020;11(1):3083. https://pubmed.ncbi.nlm.nih.gov/32576828/
Vernieri C, Ligorio F, Zattarin E, Rivoltini L, de Braud F. Fasting-mimicking diet plus chemotherapy in breast cancer treatment. Nat Commun. 2020;11(1):4274. https://pubmed.ncbi.nlm.nih.gov/32848145/
de Groot S, Lugtenberg RT, Cohen D, et al. Fasting mimicking diet as an adjunct to¿neoadjuvant chemotherapy for breast cancer in the multicentre randomized phase 2 DIRECT trial. Nat Commun. 2020;11(1):3083. https://pubmed.ncbi.nlm.nih.gov/32576828/
Vernieri C, Ligorio F, Zattarin E, Rivoltini L, de Braud F. Fasting-mimicking diet plus chemotherapy in breast cancer treatment. Nat Commun. 2020;11(1):4274. https://pubmed.ncbi.nlm.nih.gov/32848145/
Lee C, Longo VD. Fasting vs dietary restriction in cellular protection and cancer treatment: from model organisms to patients. Oncogene. 2011;30(30):3305–16. https://pubmed.ncbi.nlm.nih.gov/21516129/
Lee C, Safdie FM, Raffaghello L, et al. Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res. 2010;70(4):1564–72. https://pubmed.ncbi.nlm.nih.gov/20145127/
Lee C, Raffaghello L, Brandhorst S, et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med. 2012;4(124):124ra27. https://pubmed.ncbi.nlm.nih.gov/22323820/
Longo VD, Lieber MR, Vijg J. Turning anti-ageing genes against cancer. Nat Rev Mol Cell Biol. 2008;9(11):903–10. https://pubmed.ncbi.nlm.nih.gov/18946478/
Clemmons DR, Klibanski A, Underwood LE, et al. Reduction of plasma immunoreactive somatomedin C during fasting in humans. J Clin Endocrinol Metab. 1981;53(6):1247–50. https://pubmed.ncbi.nlm.nih.gov/7197688/
Fontana L, Weiss EP, Villareal DT, Klein S, Holloszy JO. Long-term effects of calorie or protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell. 2008;7(5):681–7. https://pubmed.ncbi.nlm.nih.gov/18843793/
Kazemi A, Speakman JR, Soltani S, Djafarian K. Effect of calorie restriction or protein intake on circulating levels of insulin like growth factor I in humans: a systematic review and meta-analysis. Clin Nutr. 2020;39(6):1705–16. https://pubmed.ncbi.nlm.nih.gov/31431306/
Fontana L, Villareal DT, Das SK, et al. Effects of 2-year calorie restriction on circulating levels of IGF-1, IGF-binding proteins and cortisol in nonobese men and women: a randomized clinical trial. Aging Cell. 2016;15(1):22–7. https://pubmed.ncbi.nlm.nih.gov/26443692/
Most J, Tosti V, Redman LM, Fontana L. Calorie restriction in humans: an update. Ageing Res Rev. 2017;39:36–45. https://pubmed.ncbi.nlm.nih.gov/27544442/
Kazemi A, Speakman JR, Soltani S, Djafarian K. Effect of calorie restriction or protein intake on circulating levels of insulin like growth factor I in humans: a systematic review and meta-analysis. Clin Nutr. 2020;39(6):1705–16. https://pubmed.ncbi.nlm.nih.gov/31431306/
Fontana L, Weiss EP, Villareal DT, Klein S, Holloszy JO. Long-term effects of calorie or protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell. 2008;7(5):681–7. https://pubmed.ncbi.nlm.nih.gov/18843793/
Solon-Biet SM, Mitchell SJ, de Cabo R, Raubenheimer D, Le Couteur DG, Simpson SJ. Macronutrients and caloric intake in health and longevity. J Endocrinol. 2015;226(1):R17–28. https://pubmed.ncbi.nlm.nih.gov/26021555/
Piper MDW, Partridge L, Raubenheimer D, Simpson SJ. Dietary restriction and ageing: a unifying perspective. Cell Metab. 2011;14(2):154–60. https://pubmed.ncbi.nlm.nih.gov/21803286/
Pamplona R, Barja G. Mitochondrial oxidative stress, aging and caloric restriction: the protein and methionine connection. Biochim Biophys Acta. 2006;1757(5–6):496–508. https://pubmed.ncbi.nlm.nih.gov/16574059/
Speakman JR, Mitchell SE, Mazidi M. Calories or protein? The effect of dietary restriction on lifespan in rodents is explained by calories alone. Exp Gerontol. 2016;86:28–38. https://pubmed.ncbi.nlm.nih.gov/27006163/
Solon-Biet SM, McMahon AC, Ballard JWO, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19(3):418–30 https://pubmed.ncbi.nlm.nih.gov/24606899/
Nishimura T, Nakatake Y, Konishi M, Itoh N. Identification of a novel FGF, FGF-21, preferentially expressed in the liver. Biochim Biophys Acta. 2000;1492(1):203–6. https://pubmed.ncbi.nlm.nih.gov/10858549/
McCarty MF. Practical prospects for boosting hepatic production of the “pro-longevity” hormone FGF21. Horm Mol Biol Clin Investig. 2017;30(2). https://pubmed.ncbi.nlm.nih.gov/26741352/
Andersen B, Straarup EM, Heppner KM, et al. FGF21 decreases body weight without reducing food intake or bone mineral density in high-fat fed obese rhesus macaque monkeys. Int J Obes (Lond). 2018;42(6):1151–60. https://pubmed.ncbi.nlm.nih.gov/29892039/
Zhang Y, Xie Y, Berglund ED, et al. The starvation hormone, fibroblast growth factor-21, extends lifespan in mice. eLife. 2012;1:e00065. https://pubmed.ncbi.nlm.nih.gov/23066506/
Solon-Biet SM, Mitchell SJ, de Cabo R, Raubenheimer D, Le Couteur DG, Simpson SJ. Macronutrients and caloric intake in health and longevity. J Endocrinol. 2015;226(1):R17–28. https://pubmed.ncbi.nlm.nih.gov/26021555/
Lee MB, Hill CM, Bitto A, Kaeberlein M. Antiaging diets: Separating fact from fiction. Science. 2021;374(6570):eabe7365. https://pubmed.ncbi.nlm.nih.gov/34793210/
Riera CE, Dillin A. Can aging be ‘drugged’?. Nat Med. 2015;21(12):1400–5. https://pubmed.ncbi.nlm.nih.gov/26646496/
Huang Z, Xu A, Cheung BMY. The potential role of fibroblast growth factor 21 in lipid metabolism and hypertension. Curr Hypertens Rep. 2017;19(4):28. https://pubmed.ncbi.nlm.nih.gov/28337713/
Pérez-Martí A, Sandoval V, Marrero PF, Haro D, Relat J. Nutritional regulation of fibroblast growth factor 21: from macronutrients to bioactive dietary compounds. Horm Mol Biol Clin Investig. 2016;30(1). https://pubmed.ncbi.nlm.nih.gov/27583468/
Sonoda J, Chen MZ, Baruch A. FGF21-receptor agonists: an emerging therapeutic class for obesity-related diseases. Horm Mol Biol Clin Investig. 2017;30(2). https://pubmed.ncbi.nlm.nih.gov/28525362/
Talukdar S, Zhou Y, Li D, et al. A long-acting FGF21 molecule, PF-05231023, decreases body weight and improves lipid profile in non-human primates and type 2 diabetic subjects. Cell Metab. 2016;23(3):427–40. https://pubmed.ncbi.nlm.nih.gov/26959184/
Harrison SA, Ruane PJ, Freilich BL, et al. Efruxifermin in non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled, phase 2a trial. Nat Med. 2021;27(7):1262–71. https://pubmed.ncbi.nlm.nih.gov/34239138/
Jimenez V, Jambrina C, Casana E, et al. FGF21 gene therapy as treatment for obesity and insulin resistance. EMBO Mol Med. 2018;10(8):e8791. https://pubmed.ncbi.nlm.nih.gov/29987000/
Cuevas-Ramos D, Almeda-Valdés P, Meza-Arana CE, et al. Exercise increases serum fibroblast growth factor 21 (FGF21) levels. PLoS One. 2012;7(5):e38022. https://pubmed.ncbi.nlm.nih.gov/22701542/
Cuevas-Ramos D, Almeda-Valdés P, Meza-Arana CE, et al. Exercise increases serum fibroblast growth factor 21 (FGF21) levels. PLoS One. 2012;7(5):e38022. https://pubmed.ncbi.nlm.nih.gov/22701542/
Khalafi M, Alamdari KA, Symonds ME, Nobari H, Carlos-Vivas J. Impact of acute exercise on immediate and following early post-exercise FGF-21 concentration in adults: systematic review and meta-analysis. Hormones (Athens). 2021;20(1):23–33. https://pubmed.ncbi.nlm.nih.gov/33151509/
Keihanian A, Arazi H, Kargarfard M. Effects of aerobic versus resistance training on serum fetuin-A, fetuin-B, and fibroblast growth factor-21 levels in male diabetic patients. Physiol Int. 2019;106(1):70–80. https://pubmed.ncbi.nlm.nih.gov/30888221/
Erickson A, Moreau R. The regulation of FGF21 gene expression by metabolic factors and nutrients. Horm Mol Biol Clin Investig. 2016;30(1). https://pubmed.ncbi.nlm.nih.gov/27285327/
Zhang Y, Xie Y, Berglund ED, et al. The starvation hormone, fibroblast growth factor-21, extends lifespan in mice. eLife. 2012;1:e00065. https://pubmed.ncbi.nlm.nih.gov/23066506/
Salminen A, Kauppinen A, Kaarniranta K. FGF21 activates AMPK signaling: impact on metabolic regulation and the aging process. J Mol Med (Berl). 2017;95(2):123–31. https://pubmed.ncbi.nlm.nih.gov/27678528/
Holmes D. Fasting induces FGF21 in humans. Nat Rev Endocrinol. 2016;12(1):3. https://pubmed.ncbi.nlm.nih.gov/26585659/
Fazeli PK, Lun M, Kim SM, et al. FGF21 and the late adaptive response to starvation in humans. J Clin Invest. 2015;125(12):4601–11. https://pubmed.ncbi.nlm.nih.gov/26529252/
Gälman C, Lundåsen T, Kharitonenkov A, et al. The circulating metabolic regulator FGF21 is induced by prolonged fasting and PPARa activation in man. Cell Metab. 2008;8(2):169–74. https://pubmed.ncbi.nlm.nih.gov/18680716/
Murata Y, Nishio K, Mochiyama T, et al. Fgf21 impairs adipocyte insulin sensitivity in mice fed a low-carbohydrate, high-fat ketogenic diet. PLoS One. 2013;8(7):e69330. https://pubmed.ncbi.nlm.nih.gov/23874946/
Gälman C, Lundåsen T, Kharitonenkov A, et al. The circulating metabolic regulator FGF21 is induced by prolonged fasting and PPARa activation in man. Cell Metab. 2008;8(2):169–74. https://pubmed.ncbi.nlm.nih.gov/18680716/
Crujeiras AB, Gomez-Arbelaez D, Zulet MA, et al. Plasma FGF21 levels in obese patients undergoing energy-restricted diets or bariatric surgery: a marker of metabolic stress? Int J Obes. 2017;41(10):1570–8. https://pubmed.ncbi.nlm.nih.gov/28588304/
Christodoulides C, Dyson P, Sprecher D, Tsintzas K, Karpe F. Circulating fibroblast growth factor 21 is induced by peroxisome proliferator-activated receptor agonists but not ketosis in man. J Clin Endocrinol Metab. 2009;94(9):3594–601. https://pubmed.ncbi.nlm.nih.gov/19531592/
Asle Mohammadi Zadeh M, Kargarfard M, Marandi SM, Habibi A. Diets along with interval training regimes improves inflammatory & anti-inflammatory condition in obesity with type 2 diabetes subjects. J Diabetes Metab Disord. 2018;17(2):253–67. https://pubmed.ncbi.nlm.nih.gov/30918861/
Salminen A, Kaarniranta K, Kauppinen A. Integrated stress response stimulates FGF21 expression: systemic enhancer of longevity. Cell Signal. 2017;40:10–21. https://pubmed.ncbi.nlm.nih.gov/28844867/
Fazeli PK, Lun M, Kim SM, et al. FGF21 and the late adaptive response to starvation in humans. J Clin Invest. 2015;125(12):4601–11. https://pubmed.ncbi.nlm.nih.gov/26529252/
Lundsgaard AM, Fritzen AM, Sjøberg KA, et al. Circulating FGF21 in humans is potently induced by short term overfeeding of carbohydrates. Mol Metab. 2017;6(1):22–9. https://pubmed.ncbi.nlm.nih.gov/28123934/
Li H, Gao Z, Zhang J, et al. Sodium butyrate stimulates expression of fibroblast growth factor 21 in liver by inhibition of histone deacetylase 3. Diabetes. 2012;61(4):797–806. https://pubmed.ncbi.nlm.nih.gov/22338096/
Erickson A, Moreau R. The regulation of FGF21 gene expression by metabolic factors and nutrients. Horm Mol Biol Clin Investig. 2016;30(1). https://pubmed.ncbi.nlm.nih.gov/27285327/
Harrison DE, Strong R, Allison DB, et al. Acarbose, 17-a-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell. 2014;13(2):273–82. https://pubmed.ncbi.nlm.nih.gov/24245565/
McCarty MF. Practical prospects for boosting hepatic production of the “pro-longevity” hormone FGF21. Horm Mol Biol Clin Invest. 2015;30(2). https://pubmed.ncbi.nlm.nih.gov/26741352/
Laeger T, Henagan TM, Albarado DC, et al. FGF21 is an endocrine signal of protein restriction. J Clin Invest. 2014;124(9):3913–22. https://pubmed.ncbi.nlm.nih.gov/25133427/
Trumbo P, Schlicker S, Yates AA, Poos M. Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty acids, cholesterol, protein and amino acids. J Am Diet Assoc. 2002;102(11):1621–30. https://pubmed.ncbi.nlm.nih.gov/12449285/
Nutrient intakes from food: mean amounts and percentages of calories from protein, carbohydrate, fat, and alcohol, one day, 2005–2006. Agricultural Research Service, United States Department of Agriculture. https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/0506/table_2_nif_05.pdf. Published 2008. Accessed January 19, 2023.; https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/0506/table_2_nif_05.pdf
Fontana L, Cummings NE, Arriola Apelo SI, et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 2016;16(2):520–30. https://pubmed.ncbi.nlm.nih.gov/27346343/
Fontana L, Cummings NE, Arriola SI, et al. Supplemental information: decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. https://www.cell.com/cms/10.1016/j.celrep.2016.05.092/attachment/1cc73bb8-d48a-497a-8cb6–16828f45777b/mmc1.pdf. Published July 12, 2016. Accessed January 1, 2023.; https://www.cell.com/cms/10.1016/j.celrep.2016.05.092/attachment/1cc73bb8-d48a-497a-8cb6-16828f45777b/mmc1.pdf
Fontana L, Cummings NE, Arriola Apelo SI, et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 2016;16(2):520–30. https://pubmed.ncbi.nlm.nih.gov/27346343/
Müller TD, Tschöp MH. Play down protein to play up metabolism? J Clin Invest. 2014;124(9):3691–3. https://pubmed.ncbi.nlm.nih.gov/25133420/
Fontana L, Cummings NE, Arriola Apelo SI, et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 2016;16(2):520–30. https://pubmed.ncbi.nlm.nih.gov/27346343/
Maida A, Zota A, Sjøberg KA, et al. A liver stress-endocrine nexus promotes metabolic integrity during dietary protein dilution. J Clin Invest. 2016;126(9):3263–78. https://pubmed.ncbi.nlm.nih.gov/27548521/
Gosby AK, Conigrave AD, Lau NS, et al. Testing protein leverage in lean humans: a randomised controlled experimental study. PLoS One. 2011;6(10):e25929. https://pubmed.ncbi.nlm.nih.gov/22022472/
Gosby AK, Lau NS, Tam CS, et al. Raised FGF-21 and triglycerides accompany increased energy intake driven by protein leverage in lean, healthy individuals: a randomised trial. PLoS One. 2016;11(8):e0161003. https://pubmed.ncbi.nlm.nih.gov/27536869/
Fontana L, Cummings NE, Arriola Apelo SI, et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 2016;16(2):520–30. https://pubmed.ncbi.nlm.nih.gov/27346343/
Kitada M, Ogura Y, Monno I, Koya D. The impact of dietary protein intake on longevity and metabolic health. EBioMedicine. 2019;43:632–40. https://pubmed.ncbi.nlm.nih.gov/30975545/
Mladenovic D, Radosavljevic T, Hrncic D, Rasic-Markovic A, Stanojlovic O. The effects of dietary methionine restriction on the function and metabolic reprogramming in the liver and brain – implications for longevity. Rev Neurosci. 2019;30(6):581–93. https://pubmed.ncbi.nlm.nih.gov/30817309/
Cole JT. Metabolism of BCAAs. In: Rajendram R, et al, eds. Branched Chain Amino Acids in Clinical Nutrition. Vol 1. Springer Science+Business Media; 2015:13–24. https://link.springer.com/chapter/10.1007/978-1-4939-1923-9_2
Kitada M, Ogura Y, Monno I, Koya D. The impact of dietary protein intake on longevity and metabolic health. EBioMedicine. 2019;43:632–40. https://pubmed.ncbi.nlm.nih.gov/30975545/
Ables GP, Johnson JE. Pleiotropic responses to methionine restriction. Exp Gerontol. 2017;94:83–8. https://pubmed.ncbi.nlm.nih.gov/28108330/
McCarty MF. GCN2 and FGF21 are likely mediators of the protection from cancer, autoimmunity, obesity, and diabetes afforded by vegan diets. Med Hypotheses. 2014;83(3):365–71. https://pubmed.ncbi.nlm.nih.gov/25015767/
McCarty MF. The moderate essential amino acid restriction entailed by low-protein vegan diets may promote vascular health by stimulating FGF21 secretion. Horm Mol Biol Clin Investig. 2016;30(1). https://pubmed.ncbi.nlm.nih.gov/26872317/
Lonnie M, Johnstone AM. The public health rationale for promoting plant protein as an important part of a sustainable and healthy diet. Nutr Bull. 2020;45(3):281–93. https://onlinelibrary.wiley.com/doi/10.1111/nbu.12453
McCarty MF. Practical prospects for boosting hepatic production of the “pro-longevity” hormone FGF21. Horm Mol Biol Clin Investig. 2015;30(2). https://pubmed.ncbi.nlm.nih.gov/26741352/
Castaño-Martinez T, Schumacher F, Schumacher S, et al. Methionine restriction prevents onset of type 2 diabetes in NZO mice. FASEB J. 2019;33(6):7092–102. https://pubmed.ncbi.nlm.nih.gov/30841758/
Castaño-Martinez T, Schumacher F, Schumacher S, et al. Methionine restriction prevents onset of type 2 diabetes in NZO mice. FASEB J. 2019;33(6):7092–102. https://pubmed.ncbi.nlm.nih.gov/30841758/
Longo VD, Mattson MP. Fasting: molecular mechanisms and clinical applications. Cell Metab. 2014;19(2):181–92. https://pubmed.ncbi.nlm.nih.gov/24440038/
Hill CM, Albarado DC, Coco LG, et al. FGF21 is required for protein restriction to extend lifespan and improve metabolic health in male mice. Nat Commun. 2022;13(1):1897. https://pubmed.ncbi.nlm.nih.gov/35393401/
Estévez M, Xiong Y. Intake of oxidized proteins and amino acids and causative oxidative stress and disease: recent scientific evidences and hypotheses. J Food Sci. 2019;84(3):387–96. https://pubmed.ncbi.nlm.nih.gov/30714623/
Seyedsadjadi N, Berg J, Bilgin AA, Braidy N, Salonikas C, Grant R. High protein intake is associated with low plasma NAD+ levels in a healthy human cohort. PLoS One. 2018;13(8):e0201968. https://pubmed.ncbi.nlm.nih.gov/30114226/
Solon-Biet SM, Mitchell SJ, de Cabo R, Raubenheimer D, Le Couteur DG, Simpson SJ. Macronutrients and caloric intake in health and longevity. J Endocrinol. 2015;226(1):R17–28. https://pubmed.ncbi.nlm.nih.gov/26021555/
Brandhorst S, Longo VD. Protein quantity and source, fasting-mimicking diets, and longevity. Adv Nutr. 2019;10(Suppl_4):S340–50. https://pubmed.ncbi.nlm.nih.gov/31728501/
Lee C, Longo VD. Fasting vs dietary restriction in cellular protection and cancer treatment: from model organisms to patients. Oncogene. 2011;30(30):3305–16. https://pubmed.ncbi.nlm.nih.gov/21516129/
Lee C, Safdie FM, Raffaghello L, et al. Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res. 2010;70(4):1564–72. https://pubmed.ncbi.nlm.nih.gov/20145127/
Lee C, Raffaghello L, Brandhorst S, et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med. 2012;4(124):124ra27. https://pubmed.ncbi.nlm.nih.gov/22323820/
Thissen JP, Ketelslegers JM, Underwood LE. Nutritional regulation of the insulin-like growth factors. Endocr Rev. 1994;15(1):80–101. https://pubmed.ncbi.nlm.nih.gov/8156941/
Fontana L, Weiss EP, Villareal DT, Klein S, Holloszy JO. Long-term effects of calorie or protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell. 2008;7(5):681–7. https://pubmed.ncbi.nlm.nih.gov/18843793/
Allen NE, Appleby PN, Davey GK, Kaaks R, Rinaldi S, Key TJ. The associations of diet with serum insulin-like growth factor I and its main binding proteins in 292 women meat-eaters, vegetarians, and vegans. Cancer Epidemiol Biomarkers Prev. 2002;11(11):1441–8. https://pubmed.ncbi.nlm.nih.gov/12433724/
Allen NE, Appleby PN, Davey GK, Key TJ. Hormones and diet: low insulin-like growth factor-I but normal bioavailable androgens in vegan men. Br J Cancer. 2000;83(1):95–7. https://pubmed.ncbi.nlm.nih.gov/10883675/
Fontana L, Klein S, Holloszy JO. Long-term low-protein, low-calorie diet and endurance exercise modulate metabolic factors associated with cancer risk. Am J Clin Nutr. 2006;84(6):1456–62. https://pubmed.ncbi.nlm.nih.gov/17158430/
Dunn SE, Kari FW, French J, et al. Dietary restriction reduces insulin-like growth factor I levels, which modulates apoptosis, cell proliferation, and tumor progression in p53-deficient mice. Cancer Res. 1997;57(21):4667–72. https://pubmed.ncbi.nlm.nih.gov/9354418/
Fontana L, Weiss EP, Villareal DT, Klein S, Holloszy JO. Long-term effects of calorie or protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell. 2008;7(5):681–7. https://pubmed.ncbi.nlm.nih.gov/18843793/
McCarty MF. A low-fat, whole-food vegan diet, as well as other strategies that down-regulate IGF-I activity, may slow the human aging process. Med Hypotheses. 2003;60(6):784–92. https://pubmed.ncbi.nlm.nih.gov/12699704/
Murphy N, Carreras-Torres R, Song M, et al. Circulating levels of insulin-like growth factor 1 and insulin-like growth factor binding protein 3 associate with risk of colorectal cancer based on serologic and Mendelian randomization analyses. Gastroenterology. 2020;158(5):1300–12.e20. https://pubmed.ncbi.nlm.nih.gov/31884074/
Larsson SC, Michaëlsson K, Burgess S. IGF-1 and cardiometabolic diseases: a Mendelian randomisation study. Diabetologia. 2020;63(9):1775–82. https://pubmed.ncbi.nlm.nih.gov/32548700/
Hartley A, Sanderson E, Paternoster L, et al. Mendelian randomization provides evidence for a causal effect of higher serum IGF-1 concentration on risk of hip and knee osteoarthritis. Rheumatology (Oxford). 2021;60(4):1676–86. https://pubmed.ncbi.nlm.nih.gov/33027520/
Nashiro K, Guevara-Aguirre J, Braskie MN, et al. Brain structure and function associated with younger adults in growth hormone receptor-deficient humans. J Neurosci. 2017;37(7):1696–707. https://pubmed.ncbi.nlm.nih.gov/28073935/
Schut AFC, Janssen JAMJL, Deinum J, et al. Polymorphism in the promoter region of the insulin-like growth factor I gene is related to carotid intima-media thickness and aortic pulse wave velocity in subjects with hypertension. Stroke. 2003;34(7):1623–7. https://pubmed.ncbi.nlm.nih.gov/12791939/
Potassium-rich foods linked to lower stroke risk. Harvard Health Publishing. https://www.health.harvard.edu/heart-health/potassium-rich-foods-linked-to-lower-stroke-risk-. Published November 14, 2014. Accessed January 2, 2023.; https://www.health.harvard.edu/heart-health/potassium-rich-foods-linked-to-lower-stroke-risk-
McCay CM, Dilley WE, Crowell MF. Growth rates of brook trout reared upon purified rations, upon dry skim milk diets, and upon feed combinations of cereal grains. J Nutr. 1929;1(3):233–46. https://doi.org/10.1093/jn/1.3.233
McDonald RB, Ramsey JJ. Honoring Clive McCay and 75 years of calorie restriction research. J Nutr. 2010;140(7):1205–10. https://pubmed.ncbi.nlm.nih.gov/20484554/
Speakman JR, Mitchell SE, Mazidi M. Calories or protein? The effect of dietary restriction on lifespan in rodents is explained by calories alone. Exp Gerontol. 2016;86:28–38. https://pubmed.ncbi.nlm.nih.gov/27006163/
Nakagawa S, Lagisz M, Hector KL, Spencer HG. Comparative and meta-analytic insights into life extension via dietary restriction. Aging Cell. 2012;11(3):401–9. https://pubmed.ncbi.nlm.nih.gov/22268691/
Solon-Biet SM, McMahon AC, Ballard JWO, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19(3):418–30. https://pubmed.ncbi.nlm.nih.gov/24606899/
Solon-Biet SM, McMahon AC, Ballard JWO, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19(3):418–30. https://pubmed.ncbi.nlm.nih.gov/24606899/
Simpson SJ, Le Couteur DG, Raubenheimer D. Putting the balance back in diet. Cell. 2015;161(1):18–23. https://pubmed.ncbi.nlm.nih.gov/25815981/
Le Couteur DG, Tay SS, Solon-Biet S, et al. The influence of macronutrients on splanchnic and hepatic lymphocytes in aging mice. J Gerontol A Biol Sci Med Sci. 2015;70(12):1499–507. https://pubmed.ncbi.nlm.nih.gov/25335766/
Solon-Biet SM, Mitchell SJ, Coogan SCP, et al. Dietary protein to carbohydrate ratio and caloric restriction: comparing metabolic outcomes in mice. Cell Rep. 2015;11(10):1529–34. https://pubmed.ncbi.nlm.nih.gov/26027933/
Le Couteur DG, Tay SS, Solon-Biet S, et al. The influence of macronutrients on splanchnic and hepatic lymphocytes in aging mice. J Gerontol A Biol Sci Med Sci. 2015;70(12):1499–507. https://pubmed.ncbi.nlm.nih.gov/25335766/
Solon-Biet SM, McMahon AC, Ballard JWO, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19(3):418–30. https://pubmed.ncbi.nlm.nih.gov/24606899/
Ingram DK, de Cabo R. Calorie restriction in rodents: caveats to consider. Ageing Res Rev. 2017;39:15–28. https://pubmed.ncbi.nlm.nih.gov/28610949/
Speakman JR, Mitchell SE, Mazidi M. Calories or protein? The effect of dietary restriction on lifespan in rodents is explained by calories alone. Exp Gerontol. 2016;86:28–38. https://pubmed.ncbi.nlm.nih.gov/27006163/
Swindell WR. Dietary restriction in rats and mice: a meta-analysis and review of the evidence for genotype-dependent effects on lifespan. Ageing Res Rev. 2012;11(2):254–70. https://pubmed.ncbi.nlm.nih.gov/22210149/
Solon-Biet SM, McMahon AC, Ballard JWO, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19(3):418–30. https://pubmed.ncbi.nlm.nih.gov/24606899/
Speakman JR, Mitchell SE, Mazidi M. Calories or protein? The effect of dietary restriction on lifespan in rodents is explained by calories alone. Exp Gerontol. 2016;86:28–38. https://pubmed.ncbi.nlm.nih.gov/27006163/
Simpson SJ, Le Couteur DG, Raubenheimer D. Putting the balance back in diet. Cell. 2015;161(1):18–23. https://pubmed.ncbi.nlm.nih.gov/25815981/
Speakman JR, Mitchell SE, Mazidi M. Calories or protein? The effect of dietary restriction on lifespan in rodents is explained by calories alone. Exp Gerontol. 2016;86:28–38. https://pubmed.ncbi.nlm.nih.gov/27006163/
Solon-Biet SM, McMahon AC, Ballard JWO, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19(3):418–30. https://pubmed.ncbi.nlm.nih.gov/24606899/
Solon-Biet SM, Mitchell SJ, de Cabo R, Raubenheimer D, Le Couteur DG, Simpson SJ. Macronutrients and caloric intake in health and longevity. J Endocrinol. 2015;226(1):R17–28. https://pubmed.ncbi.nlm.nih.gov/26021555/
Wali JA, Raubenheimer D, Senior AM, Le Couteur DG, Simpson SJ. Cardio-metabolic consequences of dietary carbohydrates: reconciling contradictions using nutritional geometry. Cardiovasc Res. 2021;117(2):386–401. https://pubmed.ncbi.nlm.nih.gov/32386289/
Wali JA, Milner AJ, Luk AWS, et al. Impact of dietary carbohydrate type and protein-carbohydrate interaction on metabolic health. Nat Metab. 2021;3(6):810–28. https://pubmed.ncbi.nlm.nih.gov/34099926/
Vogtschmidt YD, Raben A, Faber I, et al. Is protein the forgotten ingredient: effects of higher compared to lower protein diets on cardiometabolic risk factors. A systematic review and meta-analysis of randomised controlled trials. Atherosclerosis. 2021;328:124–35. https://pubmed.ncbi.nlm.nih.gov/34120735/
Wali JA, Raubenheimer D, Senior AM, Le Couteur DG, Simpson SJ. Cardio-metabolic consequences of dietary carbohydrates: reconciling contradictions using nutritional geometry. Cardiovasc Res. 2021;117(2):386–401. https://pubmed.ncbi.nlm.nih.gov/32386289/
Simpson SJ, Le Couteur DG, Raubenheimer D, et al. Dietary protein, aging and nutritional geometry. Ageing Res Rev. 2017;39:78–86. https://pubmed.ncbi.nlm.nih.gov/28274839/
Le Couteur DG, Solon-Biet S, Wahl D, et al. New horizons: dietary protein, ageing and the Okinawan ratio. Age Ageing. 2016;45(4):443–7. https://pubmed.ncbi.nlm.nih.gov/27130207/
Willcox BJ, Willcox DC, Todoriki H, et al. Caloric restriction, the traditional Okinawan diet, and healthy aging: the diet of the world’s longest-lived people and its potential impact on morbidity and life span. Ann N Y Acad Sci. 2007;1114:434–55. https://pubmed.ncbi.nlm.nih.gov/17986602/
Rizza W, Veronese N, Fontana L. What are the roles of calorie restriction and diet quality in promoting healthy longevity? Ageing Res Rev. 2014;13:38–45. https://pubmed.ncbi.nlm.nih.gov/24291541/
Willcox BJ, Willcox DC, Todoriki H, et al. Caloric restriction, the traditional Okinawan diet, and healthy aging: the diet of the world’s longest-lived people and its potential impact on morbidity and life span. Ann N Y Acad Sci. 2007;1114:434–55. https://pubmed.ncbi.nlm.nih.gov/17986602/
Willcox BJ, Willcox DC, Todoriki H, et al. Caloric restriction, the traditional Okinawan diet, and healthy aging: the diet of the world’s longest-lived people and its potential impact on morbidity and life span. Ann N Y Acad Sci. 2007;1114:434–55. https://pubmed.ncbi.nlm.nih.gov/17986602/
Abraham S, Lowenstein FW, Johnson CL. Preliminary Findings of the First Health and Nutrition Examination Survey, United States, 1971–1972. U.S. Department of Health, Education, and Welfare, Public Health Service, Health Resources Administration, National Center for Health Statistics; 1974. https://stacks.cdc.gov/view/cdc/82306
Le Couteur DG, Solon-Biet S, Wahl D, et al. New horizons: dietary protein, ageing and the Okinawan ratio. Age Ageing. 2016;45(4):443–7. https://pubmed.ncbi.nlm.nih.gov/27130207/
Chen Z, Glisic M, Song M, et al. Dietary protein intake and all-cause and cause-specific mortality: results from the Rotterdam Study and a meta-analysis of prospective cohort studies. Eur J Epidemiol. 2020;35(5):411–29. https://pubmed.ncbi.nlm.nih.gov/32076944/
Green CL, Lamming DW, Fontana L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat Rev Mol Cell Biol. 2022;23(1):56–73. https://pubmed.ncbi.nlm.nih.gov/34518687/
Chen Z, Glisic M, Song M, et al. Dietary protein intake and all-cause and cause-specific mortality: results from the Rotterdam Study and a meta-analysis of prospective cohort studies. Eur J Epidemiol. 2020;35(5):411–29. https://pubmed.ncbi.nlm.nih.gov/32076944/
Levine ME, Suarez JA, Brandhorst S, et al. Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 2014;19(3):407–17. https://pubmed.ncbi.nlm.nih.gov/24606898/
Mirzaei H, Suarez JA, Longo VD. Protein and amino acid restriction, aging and disease: from yeast to humans. Trends Endocrinol Metab. 2014;25(11):558–66. https://pubmed.ncbi.nlm.nih.gov/25153840/
Hoy MK, Clemens JC, Moshfegh A. Protein intake of adults: what we eat in America, NHANES 2015–2016. Food Surveys Research Group, Dietary Data Brief No. 29. https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/DBrief/29_Protein_Intake_of_Adults_1516.pdf. Published January 2021. Accessed December 26, 2022.; https://www.ars.usda.gov/ARSUserFiles/80400530/pdf/DBrief/29_Protein_Intake_of_Adults_1516.pdf
Mirzaei H, Suarez JA, Longo VD. Protein and amino acid restriction, aging and disease: from yeast to humans. Trends Endocrinol Metab. 2014;25(11):558–66. https://pubmed.ncbi.nlm.nih.gov/25153840/
Huang J, Liao LM, Weinstein SJ, Sinha R, Graubard BI, Albanes D. Association between plant and animal protein intake and overall and cause-specific mortality. JAMA Intern Med. 2020;180(9):1173–84. https://pubmed.ncbi.nlm.nih.gov/32658243/
Meroño T, Zamora-Ros R, Hidalgo-Liberona N, et al. Animal protein intake is inversely associated with mortality in older adults: the InCHIANTI study. J Gerontol A Biol Sci Med Sci. 2022;77(9):1866–72. https://pubmed.ncbi.nlm.nih.gov/34849845/
Naghshi S, Sadeghi O, Willett WC, Esmaillzadeh A. Dietary intake of total, animal, and plant proteins and risk of all cause, cardiovascular, and cancer mortality: systematic review and dose-response meta-analysis of prospective cohort studies. BMJ. 2020;370:m2412. https://pubmed.ncbi.nlm.nih.gov/32699048/
Ortolá R, Struijk EA, García-Esquinas E, Rodríguez-Artalejo F, Lopez-Garcia E. Changes in dietary intake of animal and vegetable protein and unhealthy aging. Am J Med. 2020;133(2):231–9.e7. https://pubmed.ncbi.nlm.nih.gov/33839765/
Sun Y, Liu B, Snetselaar LG, et al. Association of major dietary protein sources with all-cause and cause-specific mortality: prospective cohort study. J Am Heart Assoc. 2021;10(5):e015553. https://pubmed.ncbi.nlm.nih.gov/33624505/
Zhong VW, Allen NB, Greenland P, et al. Protein foods from animal sources, incident cardiovascular disease and all-cause mortality: a substitution analysis. Int J Epidemiol. 2021;50(1):223–33. https://pubmed.ncbi.nlm.nih.gov/33411911/
Zheng J, Zhu T, Yang G, et al. The isocaloric substitution of plant-based and animal-based protein in relation to aging-related health outcomes: a systematic review. Nutrients. 2022;14(2):272. https://pubmed.ncbi.nlm.nih.gov/35057453/
Willcox BJ, Willcox DC, Todoriki H, et al. Caloric restriction, the traditional Okinawan diet, and healthy aging: the diet of the world’s longest-lived people and its potential impact on morbidity and life span. Ann N Y Acad Sci. 2007;1114:434–55. https://pubmed.ncbi.nlm.nih.gov/17986602/
Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161(13):1645–52. https://pubmed.ncbi.nlm.nih.gov/11434797/
Rizzo NS, Jaceldo-Siegl K, Sabate J, Fraser GE. Nutrient profiles of vegetarian and nonvegetarian dietary patterns. J Acad Nutr Diet. 2013;113(12):1610–9. https://pubmed.ncbi.nlm.nih.gov/23988511/
Kitada M, Ogura Y, Monno I, Koya D. The impact of dietary protein intake on longevity and metabolic health. EBioMedicine. 2019;43:632–40. https://pubmed.ncbi.nlm.nih.gov/30975545/
Schüler R, Markova M, Osterhoff MA, et al. Similar dietary regulation of IGF-1-and IGF-binding proteins by animal and plant protein in subjects with type 2 diabetes. Eur J Nutr. 2021;60(6):3499–504. https://pubmed.ncbi.nlm.nih.gov/33686453/
Kahleova H, Fleeman R, Hlozkova A, Holubkov R, Barnard ND. A plant-based diet in overweight individuals in a 16-week randomized clinical trial: metabolic benefits of plant protein. Nutr Diabetes. 2018;8(1):58. https://pubmed.ncbi.nlm.nih.gov/30405108/
Dorling JL, Martin CK, Redman LM. Calorie restriction for enhanced longevity: the role of novel dietary strategies in the present obesogenic environment. Ageing Res Rev. 2020;64:101038. https://pubmed.ncbi.nlm.nih.gov/32109603/
Appleton BS, Campbell TC. Inhibition of aflatoxin-initiated preneoplastic liver lesions by low dietary protein. Nutr Cancer. 1982;3(4):200–6. https://pubmed.ncbi.nlm.nih.gov/6128727/
Solon-Biet SM, McMahon AC, Ballard JWO, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19(3):418–30. https://pubmed.ncbi.nlm.nih.gov/24606899/
Solon-Biet SM, Mitchell SJ, de Cabo R, Raubenheimer D, Le Couteur DG, Simpson SJ. Macronutrients and caloric intake in health and longevity. J Endocrinol. 2015;226(1):R17–28. https://pubmed.ncbi.nlm.nih.gov/26021555/
Fontana L, Adelaiye RM, Rastelli AL, et al. Dietary protein restriction inhibits tumor growth in human xenograft models. Oncotarget. 2013;4(12):2451–61. https://pubmed.ncbi.nlm.nih.gov/24353195/
Fontana L, Adelaiye RM, Rastelli AL, et al. Dietary protein restriction inhibits tumor growth in human xenograft models. Oncotarget. 2013;4(12):2451–61. https://pubmed.ncbi.nlm.nih.gov/24353195/
Rubio-Patiño C, Bossowski JP, De Donatis GM, et al. Low-protein diet induces IRE1a-dependent anticancer immunosurveillance. Cell Metab. 2018;27(4):828–42.e7. https://pubmed.ncbi.nlm.nih.gov/29551590/
Orillion A, Damayanti NP, Shen L, et al. Dietary protein restriction reprograms tumor-associated macrophages and enhances immunotherapy. Clin Cancer Res. 2018;24(24):6383–95. https://pubmed.ncbi.nlm.nih.gov/30190370/
Pili R, Fontana L. Low-protein diet in cancer: ready for prime time? Nat Rev Endocrinol. 2018;14(7):384–6. https://pubmed.ncbi.nlm.nih.gov/29765134/
Gao X, Sanderson SM, Dai Z, et al. Dietary methionine influences therapy in mouse cancer models and alters human metabolism. Nature. 2019;572(7769):397–401. https://pubmed.ncbi.nlm.nih.gov/31367041/
Solon-Biet SM, McMahon AC, Ballard JWO, et al. The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice. Cell Metab. 2014;19(3):418–30. https://pubmed.ncbi.nlm.nih.gov/24606899/
Trepanowski JF, Canale RE, Marshall KE, Kabir MM, Bloomer RJ. Impact of caloric and dietary restriction regimens on markers of health and longevity in humans and animals: a summary of available findings. Nutr J. 2011;10:107. https://pubmed.ncbi.nlm.nih.gov/21981968/
Pamplona R, Barja G. Mitochondrial oxidative stress, aging and caloric restriction: the protein and methionine connection. Biochim Biophys Acta. 2006;1757(5–6):496–508. https://pubmed.ncbi.nlm.nih.gov/16574059/
McIsaac RS, Lewis KN, Gibney PA, Buffenstein R. From yeast to human: exploring the comparative biology of methionine restriction in extending eukaryotic life span. Ann N Y Acad Sci. 2016;1363:155–70. https://pubmed.ncbi.nlm.nih.gov/26995762/
Gorbunova V, Bozzella MJ, Seluanov A. Rodents for comparative aging studies: from mice to beavers. Age (Dordr). 2008;30(2–3):111–9. https://pubmed.ncbi.nlm.nih.gov/19424861/
Zimmerman JA, Malloy V, Krajcik R, Orentreich N. Nutritional control of aging. Exp Gerontol. 2003;38(1–2):47–52. https://pubmed.ncbi.nlm.nih.gov/12543260/
Swindell WR. Dietary restriction in rats and mice: a meta-analysis and review of the evidence for genotype-dependent effects on lifespan. Ageing Res Rev. 2012;11(2):254–70. https://pubmed.ncbi.nlm.nih.gov/22210149/
Miller RA, Buehner G, Chang Y, Harper JM, Sigler R, Smith-Wheelock M. Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell. 2005;4(3):119–25. https://pubmed.ncbi.nlm.nih.gov/15924568/
Yu D, Yang SE, Miller BR, et al. Short-term methionine deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms. FASEB J. 2018;32(6):3471–82. https://pubmed.ncbi.nlm.nih.gov/29401631/
Miller RA, Buehner G, Chang Y, Harper JM, Sigler R, Smith-Wheelock M. Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell. 2005;4(3):119–25. https://pubmed.ncbi.nlm.nih.gov/15924568/
Miller RA, Buehner G, Chang Y, Harper JM, Sigler R, Smith-Wheelock M. Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell. 2005;4(3):119–25. https://pubmed.ncbi.nlm.nih.gov/15924568/
Yu D, Yang SE, Miller BR, et al. Short-term methionine deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms. FASEB J. 2018;32(6):3471–82. https://pubmed.ncbi.nlm.nih.gov/29401631/
Ruckenstuhl C, Netzberger C, Entfellner I, et al. Lifespan extension by methionine restriction requires autophagy-dependent vacuolar acidification. PLoS Genet. 2014;10(5):e1004347. https://pubmed.ncbi.nlm.nih.gov/24785424/
Sharma S, Dixon T, Jung S, et al. Dietary methionine restriction reduces inflammation independent of FGF21 action. Obesity (Silver Spring). 2019;27(8):1305–13. https://pubmed.ncbi.nlm.nih.gov/31207147/
Miller RA, Buehner G, Chang Y, Harper JM, Sigler R, Smith-Wheelock M. Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell. 2005;4(3):119–25. https://pubmed.ncbi.nlm.nih.gov/15924568/
Brown-Borg HM, Rakoczy SG, Wonderlich JA, et al. Growth hormone signaling is necessary for lifespan extension by dietary methionine. Aging Cell. 2014;13(6):1019–27. https://pubmed.ncbi.nlm.nih.gov/25234161/
Harper AE, Benevenga NJ, Wohlhueter RM. Effects of ingestion of disproportionate amounts of amino acids. Physiol Rev. 1970;50(3):428–558. https://pubmed.ncbi.nlm.nih.gov/4912906/
López-Torres M, Barja G. Lowered methionine ingestion as responsible for the decrease in rodent mitochondrial oxidative stress in protein and dietary restriction. Possible implications for humans. Biochim Biophys Acta. 2008;1780(11):1337–47. https://pubmed.ncbi.nlm.nih.gov/18252204/
Mori N, Hirayama K. Long-term consumption of a methionine-supplemented diet increases iron and lipid peroxide levels in rat liver. J Nutr. 2000;130(9):2349–55. https://pubmed.ncbi.nlm.nih.gov/10958834/
Hidiroglou N, Gilani GS, Long L, et al. The influence of dietary vitamin E, fat, and methionine on blood cholesterol profile, homocysteine levels, and oxidizability of low density lipoprotein in the gerbil. J Nutr Biochem. 2004;15(12):730–40. https://pubmed.ncbi.nlm.nih.gov/15607646/
Sanz A, Caro P, Ayala V, Portero-Otin M, Pamplona R, Barja G. Methionine restriction decreases mitochondrial oxygen radical generation and leak as well as oxidative damage to mitochondrial DNA and proteins. FASEB J. 2006;20(8):1064–73. https://pubmed.ncbi.nlm.nih.gov/16770005/
Caro P, Gomez J, Sanchez I, et al. Effect of 40 % restriction of dietary amino acids (except methionine) on mitochondrial oxidative stress and biogenesis, AIF and SIRT1 in rat liver. Biogerontology. 2009;10(5):579–92. https://pubmed.ncbi.nlm.nih.gov/19039676/
Moskovitz J, Bar-Noy S, Williams WM, Requena J, Berlett BS, Stadtman ER. Methionine sulfoxide reductase (MsrA) is a regulator of antioxidant defense and lifespan in mammals. Proc Natl Acad Sci U S A. 2001;98(23):12920–5. https://pubmed.ncbi.nlm.nih.gov/11606777/
Pamplona R, Barja G. Mitochondrial oxidative stress, aging and caloric restriction: the protein and methionine connection. Biochim Biophys Acta. 2006;1757(5–6):496–508. https://pubmed.ncbi.nlm.nih.gov/16574059/
Lushchak O, Strilbytska OM, Yurkevych I, Vaiserman AM, Storey KB. Implications of amino acid sensing and dietary protein to the aging process. Exp Gerontol. 2019;115:69–78. https://pubmed.ncbi.nlm.nih.gov/30502540/
Ruan H, Tang XD, Chen ML, et al. High-quality life extension by the enzyme peptide methionine sulfoxide reductase. Proc Natl Acad Sci U S A. 2002;99(5):2748–53. https://pubmed.ncbi.nlm.nih.gov/11867705/
Takauji Y, Wada T, Takeda A, et al. Restriction of protein synthesis abolishes senescence features at cellular and organismal levels. Sci Rep. 2016;6:18722. https://pubmed.ncbi.nlm.nih.gov/26729469/
Green CL, Lamming DW, Fontana L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat Rev Mol Cell Biol. 2022;23(1):56–73. https://pubmed.ncbi.nlm.nih.gov/34518687/
Koziel R, Ruckenstuhl C, Albertini E, et al. Methionine restriction slows down senescence in human diploid fibroblasts. Aging Cell. 2014;13(6):1038–48. https://pubmed.ncbi.nlm.nih.gov/25273919/
Brown-Borg HM, Buffenstein R. Cutting back on the essentials: can manipulating intake of specific amino acids modulate health and lifespan? Ageing Res Rev. 2017;39:87–95. https://pubmed.ncbi.nlm.nih.gov/27570078/
Johnson JE, Johnson FB. Methionine restriction activates the retrograde response and confers both stress tolerance and lifespan extension to yeast, mouse and human cells. PLoS One. 2014;9(5):e97729. https://pubmed.ncbi.nlm.nih.gov/24830393/
Agrawal V, Alpini SEJ, Stone EM, Frenkel EP, Frankel AE. Targeting methionine auxotrophy in cancer: discovery & exploration. Expert Opin Biol Ther. 2012;12(1):53–61. https://pubmed.ncbi.nlm.nih.gov/22171665/
Han Q, Tan Y, Hoffman RM. Oral dosing of recombinant methioninase is associated with a 70 % drop in PSA in a patient with bone-metastatic prostate cancer and 50 % reduction in circulating methionine in a high-stage ovarian cancer patient. Anticancer Res. 2020;40(5):2813–9. https://pubmed.ncbi.nlm.nih.gov/32366428/
Cavuoto P, Fenech MF. A review of methionine dependency and the role of methionine restriction in cancer growth control and life-span extension. Cancer Treat Rev. 2012;38(6):726–36. https://pubmed.ncbi.nlm.nih.gov/22342103/
Heilbronn LK, Panda S. Alternate-day fasting gets a safe bill of health. Cell Metab. 2019;30(3):411–3. https://pubmed.ncbi.nlm.nih.gov/31484053/
Stekovic S, Hofer SJ, Tripolt N, et al. Alternate day fasting improves physiological and molecular markers of aging in healthy, non-obese humans. Cell Metab. 2019;30(3):462–76. https://pubmed.ncbi.nlm.nih.gov/31471173/
McCarty MF, Barroso-Aranda J, Contreras F. The low-methionine content of vegan diets may make methionine restriction feasible as a life extension strategy. Med Hypotheses. 2009;72(2):125–8. https://pubmed.ncbi.nlm.nih.gov/18789600/
Kitada M, Ogura Y, Monno I, Koya D. The impact of dietary protein intake on longevity and metabolic health. EBioMedicine. 2019;43:632–40. https://pubmed.ncbi.nlm.nih.gov/30975545/
Yamamoto J, Han Q, Simon M, Thomas D, Hoffman RM. Methionine restriction: ready for prime time in the cancer clinic? Anticancer Res. 2022;42(2):641–4. https://pubmed.ncbi.nlm.nih.gov/35093861/
Product information: Hominex®-2. Abbott Laboratories Inc. Updated May 3, 2022.; https://www.abbottnutrition.com/our-products/hominex-2
Yamamoto J, Han Q, Simon M, Thomas D, Hoffman RM. Methionine restriction: ready for prime time in the cancer clinic? Anticancer Res. 2022;42(2):641–4. https://pubmed.ncbi.nlm.nih.gov/35093861/
McCarty MF, Barroso-Aranda J, Contreras F. The low-methionine content of vegan diets may make methionine restriction feasible as a life extension strategy. Med Hypotheses. 2009;72(2):125–8. https://pubmed.ncbi.nlm.nih.gov/18789600/
Agricultural Research Service, United States Department of Agriculture. Component search: methionine. FoodData Central. https://fdc.nal.usda.gov/fdc-app.html#/food-search?component=1215. Accessed January 25, 2023.; https://fdc.nal.usda.gov/fdc-app.html#/food-search?component=1215
Gaudichon C, Calvez J. Determinants of amino acid bioavailability from ingested protein in relation to gut health. Curr Opin Clin Nutr Metab Care. 2021;24(1):55–61. https://pubmed.ncbi.nlm.nih.gov/33093304/
McCarty MF, Barroso-Aranda J, Contreras F. The low-methionine content of vegan diets may make methionine restriction feasible as a life extension strategy. Med Hypotheses. 2009;72(2):125–8. https://pubmed.ncbi.nlm.nih.gov/18789600/
Cavuoto P, Fenech MF. A review of methionine dependency and the role of methionine restriction in cancer growth control and life-span extension. Cancer Treat Rev. 2012;38(6):726–36. https://pubmed.ncbi.nlm.nih.gov/22342103/
Cavuoto P, Fenech MF. A review of methionine dependency and the role of methionine restriction in cancer growth control and life-span extension. Cancer Treat Rev. 2012;38(6):726–36. https://pubmed.ncbi.nlm.nih.gov/22342103/
Krajcovicova-Kudlackova M, Babinska K, Valachovicova M. Health benefits and risks of plant proteins. Bratisl Lek Listy. 2005;106(6–7):231–4. https://pubmed.ncbi.nlm.nih.gov/16201743/
Schmidt JA, Rinaldi S, Scalbert A, et al. Plasma concentrations and intakes of amino acids in male meat-eaters, fish-eaters, vegetarians and vegans: a cross-sectional analysis in the EPIC-Oxford cohort. Eur J Clin Nutr. 2016;70(3):306–12. https://pubmed.ncbi.nlm.nih.gov/26395436/
Wu G, Han L, Shi Y, et al. Effect of different levels of dietary methionine restriction on relieving oxidative stress and behavioral deficits in middle-aged mice fed low-, medium-, or high-fat diet. J Funct Foods. 2020;65:103782. https://doi.org/10.1016/j.jff.2020.103782
Trepanowski JF, Canale RE, Marshall KE, Kabir MM, Bloomer RJ. Impact of caloric and dietary restriction regimens on markers of health and longevity in humans and animals: a summary of available findings. Nutr J. 2011;10:107. https://pubmed.ncbi.nlm.nih.gov/21981968/
Schmidt JA, Rinaldi S, Scalbert A, et al. Plasma concentrations and intakes of amino acids in male meat-eaters, fish-eaters, vegetarians and vegans: a cross-sectional analysis in the EPIC-Oxford cohort. Eur J Clin Nutr. 2016;70(3):306–12. https://pubmed.ncbi.nlm.nih.gov/26395436/
Green CL, Lamming DW, Fontana L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat Rev Mol Cell Biol. 2022;23(1):56–73. https://pubmed.ncbi.nlm.nih.gov/34518687/
Yu D, Yang SE, Miller BR, et al. Short-term methionine deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms. FASEB J. 2018;32(6):3471–82. https://pubmed.ncbi.nlm.nih.gov/29401631/
Elshorbagy AK, Valdivia-Garcia M, Mattocks DAL, et al. Cysteine supplementation reverses methionine restriction effects on rat adiposity: significance of stearoyl-coenzyme A desaturase. J Lipid Res. 2011;52(1):104–12. https://pubmed.ncbi.nlm.nih.gov/20871132/
Le LT, Sabaté J. Beyond meatless, the health effects of vegan diets: findings from the Adventist cohorts. Nutrients. 2014;6(6):2131–47. https://pubmed.ncbi.nlm.nih.gov/24871675/
Tonstad S, Stewart K, Oda K, Batech M, Herring RP, Fraser GE. Vegetarian diets and incidence of diabetes in the Adventist Health Study-2. Nutr Metab Cardiovasc Dis. 2013;23(4):292–9. https://pubmed.ncbi.nlm.nih.gov/21983060/
Dong Z, Gao X, Chinchilli VM, et al. Association of dietary sulfur amino acid intake with mortality from diabetes and other causes. Eur J Nutr. 2022;61(1):289–98. https://pubmed.ncbi.nlm.nih.gov/34327571/
Dong Z, Gao X, Chinchilli VM, et al. Association of dietary sulfur amino acid intake with mortality from diabetes and other causes. Eur J Nutr. 2022;61(1):289–98. https://pubmed.ncbi.nlm.nih.gov/34327571/
Dong Z, Gao X, Chinchilli VM, et al. Association of sulfur amino acid consumption with cardiometabolic risk factors: cross-sectional findings from NHANES III. EClinicalMedicine. 2020;19:100248. https://pubmed.ncbi.nlm.nih.gov/32140669/
López-Torres M, Barja G. Lowered methionine ingestion as responsible for the decrease in rodent mitochondrial oxidative stress in protein and dietary restriction possible implications for humans. Biochim Biophys Acta. 2008;1780(11):1337–47. https://pubmed.ncbi.nlm.nih.gov/18252204/
Di Buono M, Wykes LJ, Ball RO, Pencharz PB. Dietary cysteine reduces the methionine requirement in men. Am J Clin Nutr. 2001;74(6):761–6. https://pubmed.ncbi.nlm.nih.gov/11722957/
Elshorbagy AK, Valdivia-Garcia M, Mattocks DAL, et al. Cysteine supplementation reverses methionine restriction effects on rat adiposity: significance of stearoyl-coenzyme A desaturase. J Lipid Res. 2011;52(1):104–12. https://pubmed.ncbi.nlm.nih.gov/20871132/
Duran-Ortiz S, List EO, Basu R, Kopchick JJ. Extending lifespan by modulating the growth hormone/insulin-like growth factor-1 axis: coming of age. Pituitary. 2021;24(3):438–56. https://pubmed.ncbi.nlm.nih.gov/33459974/
Schmidt JA, Rinaldi S, Scalbert A, et al. Plasma concentrations and intakes of amino acids in male meat-eaters, fish-eaters, vegetarians and vegans: a cross-sectional analysis in the EPIC-Oxford cohort. Eur J Clin Nutr. 2016;70(3):306–12. https://pubmed.ncbi.nlm.nih.gov/26395436/
Lederer AK, Maul-Pavicic A, Hannibal L, et al. Vegan diet reduces neutrophils, monocytes and platelets related to branched-chain amino acids – a randomized, controlled trial. Clin Nutr. 2020;39(11):3241–50. https://pubmed.ncbi.nlm.nih.gov/32147197/
Tanrikulu-Kucuk S, Ademoglu E. Dietary restriction of amino acids other than methionine prevents oxidative damage during aging: involvement of telomerase activity and telomere length. Life Sci. 2012;90(23–24):924–8. https://pubmed.ncbi.nlm.nih.gov/22564407/
Solon-Biet SM, Mitchell SJ, de Cabo R, Raubenheimer D, Le Couteur DG, Simpson SJ. Macronutrients and caloric intake in health and longevity. J Endocrinol. 2015;226(1):R17–28. https://pubmed.ncbi.nlm.nih.gov/26021555/
Solon-Biet SM, Cogger VC, Pulpitel T, et al. Branched chain amino acids impact health and lifespan indirectly via amino acid balance and appetite control. Nat Metab. 2019;1(5):532–45. https://pubmed.ncbi.nlm.nih.gov/31656947/
Lu J, Temp U, Müller-Hartmann A, Esser J, Grönke S, Partridge L. Sestrin is a key regulator of stem cell function and lifespan in response to dietary amino acids. Nat Aging. 2021;1(1):60–72. https://pubmed.ncbi.nlm.nih.gov/37117991/
Richardson NE, Konon EN, Schuster HS, et al. Lifelong restriction of dietary branched-chain amino acids has sex-specific benefits for frailty and lifespan in mice. Nat Aging. 2021;1(1):73–86. https://pubmed.ncbi.nlm.nih.gov/33796866/
Richardson NE, Konon EN, Schuster HS, et al. Lifelong restriction of dietary branched-chain amino acids has sex-specific benefits for frailty and lifespan in mice. Nat Aging. 2021;1(1):73–86. https://pubmed.ncbi.nlm.nih.gov/33796866/
Green CL, Lamming DW, Fontana L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat Rev Mol Cell Biol. 2022;23(1):56–73. https://pubmed.ncbi.nlm.nih.gov/34518687/
Solon-Biet SM, Mitchell SJ, de Cabo R, Raubenheimer D, Le Couteur DG, Simpson SJ. Macronutrients and caloric intake in health and longevity. J Endocrinol. 2015;226(1):R17–28. https://pubmed.ncbi.nlm.nih.gov/26021555/
Lee MB, Hill CM, Bitto A, Kaeberlein M. Antiaging diets: Separating fact from fiction. Science. 2021;374(6570):eabe7365. https://pubmed.ncbi.nlm.nih.gov/34793210/
Cordain L, Lindeberg S, Hurtado M, Hill K, Eaton SB, Brand-Miller J. Acne vulgaris: a disease of Western civilization. Arch Dermatol. 2002;138(12):1584–90. https://pubmed.ncbi.nlm.nih.gov/12472346/
Melnik BC, John SM, Plewig G. Acne: risk indicator for increased body mass index and insulin resistance. Acta Derm Venereol. 2013;93(6):644–9. https://pubmed.ncbi.nlm.nih.gov/23975508/
Blair MC, Neinast MD, Arany Z. Whole-body metabolic fate of branched-chain amino acids. Biochem J. 2021;478(4):765–76. https://pubmed.ncbi.nlm.nih.gov/33626142/
Neinast M, Murashige D, Arany Z. Branched chain amino acids. Annu Rev Physiol. 2019;81:139–64. https://pubmed.ncbi.nlm.nih.gov/30485760/
Tournissac M, Vandal M, Tremblay C, et al. Dietary intake of branched-chain amino acids in a mouse model of Alzheimer’s disease: effects on survival, behavior, and neuropathology. Alzheimers Dement (N Y). 2018;4:677–87. https://pubmed.ncbi.nlm.nih.gov/30560200/
Larsson SC, Markus HS. Branched-chain amino acids and Alzheimer’s disease: a Mendelian randomization analysis. Sci Rep. 2017;7(1):13604. https://pubmed.ncbi.nlm.nih.gov/29051501/
Tynkkynen J, Chouraki V, van der Lee SJ, et al. Association of branched-chain amino acids and other circulating metabolites with risk of incident dementia and Alzheimer’s disease: a prospective study in eight cohorts. Alzheimers Dement. 2018;14(6):723–33. https://pubmed.ncbi.nlm.nih.gov/29519576/
Le Couteur DG, Solon-Biet SM, Cogger VC, et al. Branched chain amino acids, aging and age-related health. Ageing Res Rev. 2020;64:101198. https://pubmed.ncbi.nlm.nih.gov/33132154/
Xu B, Wang M, Pu L, Shu C, Li L, Han L. Association of dietary intake of branched-chain amino acids with long-term risks of CVD, cancer and all-cause mortality. Public Health Nutr. 2022;25(12):3390–400. https://pubmed.ncbi.nlm.nih.gov/34930509/
Le Couteur DG, Solon-Biet SM, Cogger VC, et al. Branched chain amino acids, aging and age-related health. Ageing Res Rev. 2020;64:101198. https://pubmed.ncbi.nlm.nih.gov/33132154/
Insulin resistance. Cleveland Clinic. https://my.clevelandclinic.org/health/diseases/22206-insulin-resistance. Updated December 16, 2021. Accessed December 26, 2022.; https://my.clevelandclinic.org/health/diseases/22206-insulin-resistance
Zhang X, Li J, Zheng S, Luo Q, Zhou C, Wang C. Fasting insulin, insulin resistance, and risk of cardiovascular or all-cause mortality in non-diabetic adults: a meta-analysis. Biosci Rep. 2017;37(5):BSR20170947. https://pubmed.ncbi.nlm.nih.gov/28811358/
Ju SY, Lee JY, Kim DH. Association of metabolic syndrome and its components with all-cause and cardiovascular mortality in the elderly: a meta-analysis of prospective cohort studies. Medicine (Baltimore). 2017;96(45):e8491. https://pubmed.ncbi.nlm.nih.gov/29137039/
Bishop CA, Machate T, Henning T, et al. Detrimental effects of branched-chain amino acids in glucose tolerance can be attributed to valine induced glucotoxicity in skeletal muscle. Nutr Diabetes. 2022;12(1):1–9. https://pubmed.ncbi.nlm.nih.gov/35418570/
Jang C, Oh SF, Wada S, et al. A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance. Nat Med. 2016;22(4):421–6. https://pubmed.ncbi.nlm.nih.gov/26950361/
Williams KJ, Wu X. Imbalanced insulin action in chronic over nutrition: clinical harm, molecular mechanisms, and a way forward. Atherosclerosis. 2016;247:225–82. https://pubmed.ncbi.nlm.nih.gov/26967715/
Cummings NE, Williams EM, Kasza I, et al. Restoration of metabolic health by decreased consumption of branched-chain amino acids. J Physiol. 2018;596(4):623–45. https://pubmed.ncbi.nlm.nih.gov/29266268/
Solon-Biet SM, Cogger VC, Pulpitel T, et al. Branched chain amino acids impact health and lifespan indirectly via amino acid balance and appetite control. Nat Metab. 2019;1(5):532–45. https://pubmed.ncbi.nlm.nih.gov/31656947/
Nie C, He T, Zhang W, Zhang G, Ma X. Branched chain amino acids: beyond nutrition metabolism. Int J Mol Sci. 2018;19(4):954. https://pubmed.ncbi.nlm.nih.gov/29570613/
Bishop CA, Machate T, Henning T, et al. Detrimental effects of branched-chain amino acids in glucose tolerance can be attributed to valine induced glucotoxicity in skeletal muscle. Nutr Diabetes. 2022;12(1):1–9. https://pubmed.ncbi.nlm.nih.gov/35418570/
Rhee EP, Ho JE, Chen MH, et al. A genome-wide association study of the human metabolome in a community-based cohort. Cell Metab. 2013;18(1):130–43. https://pubmed.ncbi.nlm.nih.gov/23823483/
Lotta LA, Scott RA, Sharp SJ, et al. Genetic predisposition to an impaired metabolism of the branched-chain amino acids and risk of type 2 diabetes: a Mendelian randomisation analysis. PLoS Med. 2016;13(11):e1002179. https://pubmed.ncbi.nlm.nih.gov/27898682/
Mahendran Y, Jonsson A, Have CT, et al. Genetic evidence of a causal effect of insulin resistance on branched-chain amino acid levels. Diabetologia. 2017;60(5):873–8. https://pubmed.ncbi.nlm.nih.gov/28184960/
White PJ, Newgard CB. Branched-chain amino acids in disease. Science. 2019;363(6427):582–3. https://pubmed.ncbi.nlm.nih.gov/30733403/
Okekunle AP, Zhang M, Wang Z, et al. Dietary branched-chain amino acids intake exhibited a different relationship with type 2 diabetes and obesity risk: a meta-analysis. Acta Diabetol. 2019;56(2):187–95. https://pubmed.ncbi.nlm.nih.gov/30413881/
Ridaura VK, Faith JJ, Rey FE, et al. Cultured gut microbiota from twins discordant for obesity modulate adiposity and metabolic phenotypes in mice. Science. 2013;341(6150):1241214. https://pubmed.ncbi.nlm.nih.gov/24009397/
Bachmann OP, Dahl DB, Brechtel K, et al. Effects of intravenous and dietary lipid challenge on intramyocellular lipid content and the relation with insulin sensitivity in humans. Diabetes. 2001;50(11):2579–84. https://pubmed.ncbi.nlm.nih.gov/11679437/
Arany Z, Neinast M. Branched chain amino acids in metabolic disease. Curr Diab Rep. 2018;18(10):76. https://pubmed.ncbi.nlm.nih.gov/30112615/
Smith GI, Yoshino J, Stromsdorfer KL, et al. Protein ingestion induces muscle insulin resistance independent of leucine-mediated mTOR activation. Diabetes. 2015;64(5):1555–63. https://pubmed.ncbi.nlm.nih.gov/25475435/
Manco M, Bertuzzi A, Salinari S, et al. The ingestion of saturated fatty acid triacylglycerols acutely affects insulin secretion and insulin sensitivity in human subjects. Br J Nutr. 2004;92(6):895–903. https://pubmed.ncbi.nlm.nih.gov/15613251/
Fontana L, Cummings NE, Arriola Apelo SI, et al. Decreased consumption of branched-chain amino acids improves metabolic health. Cell Rep. 2016;16(2):520–30. https://pubmed.ncbi.nlm.nih.gov/27346343/
Cummings NE, Williams EM, Kasza I, et al. Restoration of metabolic health by decreased consumption of branched-chain amino acids. J Physiol. 2018;596(4):623–45. https://pubmed.ncbi.nlm.nih.gov/29266268/
Wolfe RR. Branched-chain amino acids and muscle protein synthesis in humans: myth or reality? J Int Soc Sports Nutr. 2017;14(1):30. https://pubmed.ncbi.nlm.nih.gov/28852372/
Buse MG. In vivo effects of branched chain amino acids on muscle protein synthesis in fasted rats. Horm Metab Res. 1981;13(9):502–5. https://pubmed.ncbi.nlm.nih.gov/7298019/
Louard RJ, Barrett EJ, Gelfand RA. Effect of infused branched-chain amino acids on muscle and whole-body amino acid metabolism in man. Clin Sci (Lond). 1990;79(5):457–66. https://pubmed.ncbi.nlm.nih.gov/2174312/
Louard RJ, Barrett EJ, Gelfand RA. Overnight branched-chain amino acid infusion causes sustained suppression of muscle proteolysis. Metabolism. 1995;44(4):424–9. https://pubmed.ncbi.nlm.nih.gov/7723664/
Plotkin DL, Delcastillo K, Van Every DW, Tipton KD, Aragon AA, Schoenfeld BJ. Isolated leucine and branched-chain amino acid supplementation for enhancing muscular strength and hypertrophy: a narrative review. Int J Sport Nutr Exerc Metab. 2021;31(3):292–301. https://pubmed.ncbi.nlm.nih.gov/33741748/
Isanejad M, LaCroix AZ, Thomson CA, et al. Branched-chain amino acid, meat intake and risk of type 2 diabetes in the Women’s Health Initiative. Br J Nutr. 2017;117(11):1523–30. https://pubmed.ncbi.nlm.nih.gov/28721839/
Adeva-Andany MM, González-Lucán M, Fernández-Fernández C, Carneiro-Freire N, Seco-Filgueira M, Pedre-Piñeiro AM. Effect of diet composition on insulin sensitivity in humans. Clin Nutr ESPEN. 2019;33:29–38. https://pubmed.ncbi.nlm.nih.gov/31451269/
Isanejad M, Lacroix AZ, Thomson CA, et al. Branched-chain amino acid, meat intake and risk of type 2 diabetes in the Women’s Health Initiative. Br J Nutr. 2017;117(11):1523–30. https://pubmed.ncbi.nlm.nih.gov/28721839/
Malik VS, Li Y, Tobias DK, Pan A, Hu FB. Dietary protein intake and risk of type 2 diabetes in US men and women. Am J Epidemiol. 2016;183(8):715–28. https://pubmed.ncbi.nlm.nih.gov/27022032/
Le Couteur DG, Solon-Biet SM, Cogger VC, et al. Branched chain amino acids, aging and age-related health. Ageing Res Rev. 2020;64:101198. https://pubmed.ncbi.nlm.nih.gov/33132154/
Hagve M, Simbo SY, Ruebush LE, et al. Postprandial concentration of circulating branched chain amino acids are able to predict the carbohydrate content of the ingested mixed meal. Clin Nutr. 2021;40(8):5020–9. https://pubmed.ncbi.nlm.nih.gov/34365036/
Hosseinpour-Niazi S, Mirmiran P, Hedayati M, Azizi F. Substitution of red meat with legumes in the therapeutic lifestyle change diet based on dietary advice improves cardiometabolic risk factors in overweight type 2 diabetes patients: a cross-over randomized clinical trial. Eur J Clin Nutr. 2015;69(5):592–7. https://pubmed.ncbi.nlm.nih.gov/25351652/
Viguiliouk E, Stewart SE, Jayalath VH, et al. Effect of replacing animal protein with plant protein on glycemic control in diabetes: a systematic review and meta-analysis of randomized controlled trials. Nutrients. 2015;7(12):9804–24. https://pubmed.ncbi.nlm.nih.gov/26633472/
Schmidt JA, Rinaldi S, Scalbert A, et al. Plasma concentrations and intakes of amino acids in male meat-eaters, fish-eaters, vegetarians and vegans: a cross-sectional analysis in the EPIC-Oxford cohort. Eur J Clin Nutr. 2016;70(3):306–12. https://pubmed.ncbi.nlm.nih.gov/26395436/
Lederer AK, Maul-Pavicic A, Hannibal L, et al. Vegan diet reduces neutrophils, monocytes and platelets related to branched-chain amino acids – a randomized, controlled trial. Clin Nutr. 2020;39(11):3241–50. https://pubmed.ncbi.nlm.nih.gov/32147197/
Kahleova H, Klementova M, Herynek V, et al. The effect of a vegetarian vs conventional hypocaloric diabetic diet on thigh adipose tissue distribution in subjects with type 2 diabetes: a randomized study. J Am Coll Nutr. 2017;36(5):364–9. https://pubmed.ncbi.nlm.nih.gov/28604251/
Lee Y, Park K. Adherence to a vegetarian diet and diabetes risk: a systematic review and meta-analysis of observational studies. Nutrients. 2017;9(6):603. https://pubmed.ncbi.nlm.nih.gov/28613258/
Kahleova H, Klementova M, Herynek V, et al. The effect of a vegetarian vs conventional hypocaloric diabetic diet on thigh adipose tissue distribution in subjects with type 2 diabetes: a randomized study. J Am Coll Nutr. 2017;36(5):364–9. https://pubmed.ncbi.nlm.nih.gov/28604251/
Goff LM, Bell JD, So PW, Dornhorst A, Frost GS. Veganism and its relationship with insulin resistance and intramyocellular lipid. Eur J Clin Nutr. 2005;59(2):291–8. https://pubmed.ncbi.nlm.nih.gov/15523486/
Valachovicová M, Krajcovicová-Kudlácková M, Blazícek P, Babinská K. No evidence of insulin resistance in normal weight vegetarians. A case control study. Eur J Nutr. 2006;45(1):52–4. https://pubmed.ncbi.nlm.nih.gov/15940383/
Kuo CS, Lai NS, Ho LT, Lin CL. Insulin sensitivity in Chinese ovo-lactovegetarians compared with omnivores. Eur J Clin Nutr. 2004;58(2):312–6. https://pubmed.ncbi.nlm.nih.gov/14749752/
Toth MJ, Poehlman ET. Sympathetic nervous system activity and resting metabolic rate in vegetarians. Metab Clin Exp. 1994;43(5):621–5. https://pubmed.ncbi.nlm.nih.gov/8177051/
Hung CJ, Huang PC, Li YH, Lu SC, Ho LT, Chou HF. Taiwanese vegetarians have higher insulin sensitivity than omnivores. Br J Nutr. 2006;95(1):129–35. https://pubmed.ncbi.nlm.nih.gov/16441925/
Kahleova H, Petersen KF, Shulman GI, et al. Effect of a low-fat vegan diet on body weight, insulin sensitivity, postprandial metabolism, and intramyocellular and hepatocellular lipid levels in overweight adults: a randomized clinical trial. JAMA Netw Open. 2020;3(11):e2025454. https://pubmed.ncbi.nlm.nih.gov/33252690/
McCarty MF. The origins of western obesity: a role for animal protein? Med Hypotheses. 2000;54(3):488–94. https://pubmed.ncbi.nlm.nih.gov/10783494/
Remer T, Pietrzik K, Manz F. A moderate increase in daily protein intake causing an enhanced endogenous insulin secretion does not alter circulating levels or urinary excretion of dehydroepiandrosterone sulfate. Metab Clin Exp. 1996;45(12):1483–6. https://pubmed.ncbi.nlm.nih.gov/8969280/
Gulliford MC, Bicknell EJ, Scarpello JH. Differential effect of protein and fat ingestion on blood glucose responses to high– and low-glycemic-index carbohydrates in noninsulin-dependent diabetic subjects. Am J Clin Nutr. 1989;50(4):773–7. https://pubmed.ncbi.nlm.nih.gov/2679037/
Ballance S, Knutsen SH, Fosvold ØW, Wickham M, Trenado CD, Monro J. Glyceamic and insulinaemic response to mashed potato alone, or with broccoli, broccoli fibre or cellulose in healthy adults. Eur J Nutr. 2018;57(1):199–207. https://pubmed.ncbi.nlm.nih.gov/27655525/
Gannon MC, Nuttall FQ, Neil BJ, Westphal SA. The insulin and glucose responses to meals of glucose plus various proteins in type II diabetic subjects. Metab Clin Exp. 1988;37(11):1081–8. https://pubmed.ncbi.nlm.nih.gov/3054432/
Gojda J, Rossmeislová L, Straková R, et al. Chronic dietary exposure to branched chain amino acids impairs glucose disposal in vegans but not in omnivores. Eur J Clin Nutr. 2017;71(5):594–601. https://pubmed.ncbi.nlm.nih.gov/28145418/
Draper CF, Vassallo I, Di Cara A, et al. A 48-hour vegan diet challenge in healthy women and men induces a branch-chain amino acid related, health associated, metabolic signature. Mol Nutr Food Res. 2018;62(3):1700703. https://pubmed.ncbi.nlm.nih.gov/29087622/
Le Couteur DG, Solon-Biet S, Cogger VC, et al. The impact of low-protein high-carbohydrate diets on aging and lifespan. Cell Mol Life Sci. 2016;73(6):1237–52. https://pubmed.ncbi.nlm.nih.gov/26718486/
Draper CF, Vassallo I, Di Cara A, et al. A 48-hour vegan diet challenge in healthy women and men induces a BRANCH-chain amino acid related, health associated, metabolic signature. Mol Nutr Food Res. 2018;62(3):1700703. https://pubmed.ncbi.nlm.nih.gov/29087622/
Kalantar-Zadeh K, Kramer HM, Fouque D. High-protein diet is bad for kidney health: unleashing the taboo. Nephrol Dial Transplant. 2020;35(1):1–4. https://pubmed.ncbi.nlm.nih.gov/31697325/
Mittendorfer B, Klein S, Fontana L. A word of caution against excessive protein intake. Nat Rev Endocrinol. 2020;16(1):59–66. https://pubmed.ncbi.nlm.nih.gov/31728051/
Larsen TM, Dalskov SM, van Baak M, et al. Diets with high or low protein content and glycemic index for weight-loss maintenance. N Engl J Med. 2010;363(22):2102–13. https://pubmed.ncbi.nlm.nih.gov/21105792/
Brandhorst S, Longo VD. Protein quantity and source, fasting-mimicking diets, and longevity. Adv Nutr. 2019;10(Suppl_4):S340–50. https://pubmed.ncbi.nlm.nih.gov/31728501/
Sifferlin A. What diet helps people live the longest? Time. 2015;185(6–7):93. https://pubmed.ncbi.nlm.nih.gov/25928954/
Fontana L. The Path to Longevity: How to Reach 100 with the Health and Stamina of a 40-Year-Old. Hardie Grant Books; 2020. https://worldcat.org/title/1129687546
Harden A, Young WJ. The alcoholic ferment of yeast-juice. Part II. – The coferment of yeast-juice. Proc R Soc Lond B. 1906;78(526):369–75. https://archive.org/details/philtrans05349481
Reiten OK, Wilvang MA, Mitchell SJ, Hu Z, Fang EF. Preclinical and clinical evidence of NAD+ precursors in health, disease, and ageing. Mech Ageing Dev. 2021;199:111567. https://pubmed.ncbi.nlm.nih.gov/34517020/
Strømland Ø, Diab J, Ferrario E, Sverkeli LJ, Ziegler M. The balance between NAD+ biosynthesis and consumption in ageing. Mech Ageing Dev. 2021;199:111569. https://pubmed.ncbi.nlm.nih.gov/34509469/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
Katsyuba E, Romani M, Hofer D, Auwerx J. NAD+ homeostasis in health and disease. Nat Metab. 2020;2(1):9–31. https://pubmed.ncbi.nlm.nih.gov/32694684/
Zapata-Pérez R, Wanders RJA, van Karnebeek CDM, Houtkooper RH. NAD+ homeostasis in human health and disease. EMBO Mol Med. 2021;13(7):e13943. https://pubmed.ncbi.nlm.nih.gov/34041853/
Giblin W, Skinner ME, Lombard DB. Sirtuins: guardians of mammalian healthspan. Trends Genet. 2014;30(7):271–86. https://pubmed.ncbi.nlm.nih.gov/24877878/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
Liu L, Su X, Quinn WJ, et al. Quantitative analysis of NAD synthesis-breakdown fluxes. Cell Metab. 2018;27(5):1067–80.e5. https://pubmed.ncbi.nlm.nih.gov/29685734/
Ziegler M, Nikiforov AA. NAD on the rise again. Nat Metab. 2020;2(4):291–2. https://pubmed.ncbi.nlm.nih.gov/32694607/
Zapata-Pérez R, Wanders RJA, van Karnebeek CDM, Houtkooper RH. NAD+ homeostasis in human health and disease. EMBO Mol Med. 2021;13(7):e13943. https://pubmed.ncbi.nlm.nih.gov/34041853/
Jacobson MK, Jacobson EL. Vitamin B3 in health and disease: toward the second century of discovery. Methods Mol Biol. 2018;1813:3–8. https://pubmed.ncbi.nlm.nih.gov/30097857/
Chini CCS, Tarragó MG, Chini EN. NAD and the aging process: role in life, death and everything in between. Mol Cell Endocrinol. 2017;455:62–74. https://pubmed.ncbi.nlm.nih.gov/27825999/
Bogan KL, Brenner C. Nicotinic acid, nicotinamide, and nicotinamide riboside: a molecular evaluation of NAD+ precursor vitamins in human nutrition. Annu Rev Nutr. 2008;28:115–30. https://pubmed.ncbi.nlm.nih.gov/18429699/
Kirkland JB, Meyer-Ficca ML. Niacin. In: Advances in Food and Nutrition Research. Elsevier;2018;83:83–149. https://pubmed.ncbi.nlm.nih.gov/29477227/
Yang Y, Sauve AA. NAD+ metabolism: bioenergetics, signaling and manipulation for therapy. Biochim Biophys Acta. 2016;1864(12):1787–800. https://pubmed.ncbi.nlm.nih.gov/27374990/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
Soma M, Lalam SK. The role of nicotinamide mononucleotide (NMN) in anti-aging, longevity, and its potential for treating chronic conditions. Mol Biol Rep. 2022;49(10):9737–48. https://pubmed.ncbi.nlm.nih.gov/35441939/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
She J, Sheng R, Qin ZH. Pharmacology and potential implications of nicotinamide adenine dinucleotide precursors. Aging Dis. 2021;12(8):1879–97. https://pubmed.ncbi.nlm.nih.gov/34881075/
Pflanzer LR. A startup that’s developed an anti-aging supplement just raised $20 million. Business Insider. https://www.businessinsider.com/elysium-health-raises-20-million-and-presents-clinical-data-2016–12. Published December 7, 2016. Accessed January 10, 2023.; https://www.businessinsider.com/elysium-health-raises-20-million-and-presents-clinical-data-2016-12
Goldstein J. Harvard researcher tied to Shaklee “anti-aging tonic” Vivix. Wall Street Journal. https://www.wsj.com/articles/BL-HEB-3860. Published December 26, 2008. Accessed January 10, 2023.; https://www.wsj.com/articles/BL-HEB-3860
Peluso A, Damgaard MV, Mori MAS, Treebak JT. Age-dependent decline of NAD+—universal truth or confounded consensus? Nutrients. 2021;14(1):101. https://pubmed.ncbi.nlm.nih.gov/35010977/
McReynolds MR, Chellappa K, Chiles E, et al. NAD+ flux is maintained in aged mice despite lower tissue concentrations. Cell Syst. 2021;12(12):1160–72.e4. https://pubmed.ncbi.nlm.nih.gov/34559996/
Peluso A, Damgaard MV, Mori MAS, Treebak JT. Age-dependent decline of NAD+—universal truth or confounded consensus? Nutrients. 2021;14(1):101. https://pubmed.ncbi.nlm.nih.gov/35010977/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
Mills KF, Yoshida S, Stein LR, et al. Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab. 2016;24(6):795–806. https://pubmed.ncbi.nlm.nih.gov/28068222/
Cerutti R, Pirinen E, Lamperti C, et al. NAD+-dependent activation of Sirt1 corrects the phenotype in a mouse model of mitochondrial disease. Cell Metab. 2014;19(6):1042–9. https://pubmed.ncbi.nlm.nih.gov/24814483/
Fang EF, Lautrup S, Hou Y, et al. NAD+ in aging: molecular mechanisms and translational implications. Trends Mol Med. 2017;23(10):899–916. https://pubmed.ncbi.nlm.nih.gov/28899755/
Okur MN, Mao B, Kimura R, et al. Short-term NAD+ supplementation prevents hearing loss in mouse models of Cockayne syndrome. NPJ Aging Mech Dis. 2020;6:1. https://pubmed.ncbi.nlm.nih.gov/31934345/
Yang Q, Cong L, Wang Y, et al. Increasing ovarian NAD+ levels improve mitochondrial functions and reverse ovarian aging. Free Radic Biol Med. 2020;156:1–10. https://pubmed.ncbi.nlm.nih.gov/32492457/
Gong B, Pan Y, Vempati P, et al. Nicotinamide riboside restores cognition through an upregulation of proliferator-activated receptor-¿ coactivator 1a regulated ß-secretase 1 degradation and mitochondrial gene expression in Alzheimer’s mouse models. Neurobiol Aging. 2013;34(6):1581–8. https://pubmed.ncbi.nlm.nih.gov/23312803/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
de Picciotto NE, Gano LB, Johnson LC, et al. Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell. 2016;15(3):522–30. https://pubmed.ncbi.nlm.nih.gov/26970090/
Yao Z, Yang W, Gao Z, Jia P. Nicotinamide mononucleotide inhibits JNK activation to reverse Alzheimer disease. Neurosci Lett. 2017;647:133–40. https://pubmed.ncbi.nlm.nih.gov/28330719/
Ryu D, Zhang H, Ropelle ER, et al. NAD+ repletion improves muscle function in muscular dystrophy and counters global PARylation. Sci Transl Med. 2016;8(361):361ra139. https://pubmed.ncbi.nlm.nih.gov/27798264/
Takeda K, Okumura K. Nicotinamide mononucleotide augments the cytotoxic activity of natural killer cells in young and elderly mice. Biomed Res. 2021;42(5):173–9. https://pubmed.ncbi.nlm.nih.gov/34544993/
Tran MT, Zsengeller ZK, Berg AH, et al. PGC1a drives NAD biosynthesis linking oxidative metabolism to renal protection. Nature. 2016;531(7595):528–32. https://pubmed.ncbi.nlm.nih.gov/26982719/
Mukherjee S, Chellappa K, Moffitt A, et al. Nicotinamide adenine dinucleotide biosynthesis promotes liver regeneration. Hepatology. 2017;65(2):616–30. https://pubmed.ncbi.nlm.nih.gov/27809334/
Gomes AP, Price NL, Ling AJY, et al. Declining NAD+ induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell. 2013;155(7):1624–38. https://pubmed.ncbi.nlm.nih.gov/24360282/
Dutta S, Sengupta P. Men and mice: relating their ages. Life Sci. 2016;152:244–8. https://pubmed.ncbi.nlm.nih.gov/26596563/
Giblin W, Skinner ME, Lombard DB. Sirtuins: guardians of mammalian healthspan. Trends Genet. 2014;30(7):271–86. https://pubmed.ncbi.nlm.nih.gov/24877878/
Anderson RM, Bitterman KJ, Wood JG, et al. Manipulation of a nuclear NAD+ salvage pathway delays aging without altering steady-state NAD+ levels. J Biol Chem. 2002;277(21):18881–90. https://pubmed.ncbi.nlm.nih.gov/11884393/
Mouchiroud L, Houtkooper RH, Moullan N, et al. The NAD+/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell. 2013;154(2):430–41. https://pubmed.ncbi.nlm.nih.gov/23870130/
Zhang H, Ryu D, Wu Y, et al. NAD¿ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science. 2016;352(6292):1436–43. https://pubmed.ncbi.nlm.nih.gov/27127236/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol. 2022;198:114946. https://pubmed.ncbi.nlm.nih.gov/35134387/
Bogan KL, Brenner C. Nicotinic acid, nicotinamide, and nicotinamide riboside: a molecular evaluation of NAD+ precursor vitamins in human nutrition. Annu Rev Nutr. 2008;28:115–30. https://pubmed.ncbi.nlm.nih.gov/18429699/
Liu L, Su X, Quinn WJ, et al. Quantitative analysis of NAD synthesis-breakdown fluxes. Cell Metab. 2018;27(5):1067–80.e5. https://pubmed.ncbi.nlm.nih.gov/29685734/
Shats I, Williams JG, Liu J, et al. Bacteria boost mammalian host NAD metabolism by engaging the deamidated biosynthesis pathway. Cell Metab. 2020;31(3):564–79.e7. https://pubmed.ncbi.nlm.nih.gov/32130883/
Romani M, Hofer DC, Katsyuba E, Auwerx J. Niacin: an old lipid drug in a new NAD+ dress. J Lipid Res. 2019;60(4):741–6. https://pubmed.ncbi.nlm.nih.gov/30782960/
Gasperi V, Sibilano M, Savini I, Catani MV. Niacin in the central nervous system: an update of biological aspects and clinical applications. Int J Mol Sci. 2019;20(4):974. https://pubmed.ncbi.nlm.nih.gov/30813414/
Altschul R, Hoffer A. Effects of salts of nicotinic acid on serum cholesterol. Br Med J. 1958;2(5098):713–4. https://pubmed.ncbi.nlm.nih.gov/13572876/
Schandelmaier S, Briel M, Saccilotto R, et al. Niacin for primary and secondary prevention of cardiovascular events. Cochrane Database Syst Rev. 2017;6(6):CD009744. https://pubmed.ncbi.nlm.nih.gov/28616955/
Canner PL, Berge KG, Wenger NK, et al. Fifteen year mortality in Coronary Drug Project patients: long-term benefit with niacin. J Am Coll Cardiol. 1986;8(6):1245–55. https://pubmed.ncbi.nlm.nih.gov/3782631/
Boden WE, Probstfield JL, Anderson T, et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med. 2011;365(24):2255–67. https://pubmed.ncbi.nlm.nih.gov/22085343/
Landray MJ, Haynes R, Hopewell JC, et al. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med. 2014;371(3):203–12. https://pubmed.ncbi.nlm.nih.gov/25014686/
Schandelmaier S, Briel M, Saccilotto R, et al. Niacin for primary and secondary prevention of cardiovascular events. Cochrane Database Syst Rev. 2017;6(6):CD009744. https://pubmed.ncbi.nlm.nih.gov/28616955/
Superko HR, Zhao XQ, Hodis HN, Guyton JR. Niacin and heart disease prevention: engraving its tombstone is a mistake. J Clin Lipidol. 2017;11(6):1309–17. https://pubmed.ncbi.nlm.nih.gov/28927896/
Krumholz HM. Niacin: time to believe outcomes over surrogate outcomes: if not now, when? Circ Cardiovasc Qual Outcomes. 2016;9(4):343–4. https://pubmed.ncbi.nlm.nih.gov/27407051/
Knopp RH, Ginsberg J, Albers JJ, et al. Contrasting effects of unmodified and time-release forms of niacin on lipoproteins in hyperlipidemic subjects: clues to mechanism of action of niacin. Metabolism. 1985;34(7):642–50. https://pubmed.ncbi.nlm.nih.gov/3925290/
Goldie C, Taylor AJ, Nguyen P, McCoy C, Zhao XQ, Preiss D. Niacin therapy and the risk of new-onset diabetes: a meta-analysis of randomised controlled trials. Heart. 2016;102(3):198–203. https://pubmed.ncbi.nlm.nih.gov/26370223/
Lloyd-Jones DM, Morris PB, Ballantyne CM, et al. 2016 ACC expert consensus decision pathway on the role of non-statin therapies for LDL-cholesterol lowering in the management of atherosclerotic cardiovascular disease risk: a report of the American College of Cardiology Task Force on Clinical Expert Consensus Documents. J Am Coll Cardiol. 2016;68(1):92–125. https://pubmed.ncbi.nlm.nih.gov/27046161/
Kent S, Haynes R, Hopewell JC, et al. Effects of vascular and nonvascular adverse events and of extended-release niacin with laropiprant on health and healthcare costs. Circ Cardiovasc Qual Outcomes. 2016;9(4):348–54. https://pubmed.ncbi.nlm.nih.gov/27407053/
Pirinen E, Auranen M, Khan NA, et al. Niacin cures systemic NAD+ deficiency and improves muscle performance in adult-onset mitochondrial myopathy. Cell Metab. 2020;31(6):1078–90.e5. https://pubmed.ncbi.nlm.nih.gov/32386566/
Zapata-Pérez R, Wanders RJA, van Karnebeek CDM, Houtkooper RH. NAD+ homeostasis in human health and disease. EMBO Mol Med. 2021;13(7):e13943. https://pubmed.ncbi.nlm.nih.gov/34041853/
Pirinen E, Auranen M, Khan NA, et al. Niacin cures systemic NAD+ deficiency and improves muscle performance in adult-onset mitochondrial myopathy. Cell Metab. 2020;31(6):1078–90.e5. https://pubmed.ncbi.nlm.nih.gov/32386566/
Pirinen E, Auranen M, Khan NA, et al. Niacin cures systemic NAD+ deficiency and improves muscle performance in adult-onset mitochondrial myopathy. Cell Metab. 2020;31(6):1078–90.e5. https://pubmed.ncbi.nlm.nih.gov/32386566/
Morris BJ. Seven sirtuins for seven deadly diseases of aging. Free Radic Biol Med. 2013;56:133–71. https://pubmed.ncbi.nlm.nih.gov/23104101/
Zhong O, Wang J, Tan Y, Lei X, Tang Z. Effects of NAD+ precursor supplementation on glucose and lipid metabolism in humans: a meta-analysis. Nutr Metab (Lond). 2022;19(1):20. https://pubmed.ncbi.nlm.nih.gov/35303905/
Goldie C, Taylor AJ, Nguyen P, McCoy C, Zhao XQ, Preiss D. Niacin therapy and the risk of new-onset diabetes: a meta-analysis of randomised controlled trials. Heart. 2016;102(3):198–203. https://pubmed.ncbi.nlm.nih.gov/26370223/
Meyer-Ficca M, Kirkland JB. Niacin. Adv Nutr. 2016;7(3):556–8. https://pubmed.ncbi.nlm.nih.gov/27184282/
Williamson G, Holst B. Dietary reference intake (DRI) value for dietary polyphenols: are we heading in the right direction? Br J Nutr. 2008;99 Suppl 3:S55–8. https://pubmed.ncbi.nlm.nih.gov/18598589/
Pitkin RM, Allen LH, Bailey LB, et al. Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline: a report of the Standing Committee of the Scientific Evaluation of Dietary Reference Intakes and its Panel on Folate, Other B Vitamins and Choline and Subcommittee on Upper Reference Levels of Nutrients. National Academies Press (US); 1998. https://pubmed.ncbi.nlm.nih.gov/23193625/
Benyó Z, Gille A, Kero J, et al. GPR109A (PUMA-g/HM74A) mediates nicotinic acid-induced flushing. J Clin Invest. 2005;115(12):3634–40. https://pubmed.ncbi.nlm.nih.gov/16322797/
DiPalma JR, Thayer WS. Use of niacin as a drug. Annu Rev Nutr. 1991;11:169–87. https://pubmed.ncbi.nlm.nih.gov/1832551/
Fukushima T. Niacin metabolism and Parkinson’s disease. Environ Health Prev Med. 2005;10(1):3–8. https://pubmed.ncbi.nlm.nih.gov/21432157/
Abdellatif M, Sedej S, Kroemer G. NAD+ metabolism in cardiac health, aging, and disease. Circulation. 2021;144(22):1795–817. https://pubmed.ncbi.nlm.nih.gov/34843394/
Elvehjem CA, Madden RJ, Strong FM, Woolley DW. Relation of nicotinic acid and nicotinic acid amide to canine black tongue. J Am Chem Soc. 1937;59(9):1767–8. https://pubs.acs.org/doi/abs/10.1021/ja01288a509
Yoshino J, Baur JA, Imai SI. NAD+ intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab. 2018;27(3):513–28. https://pubmed.ncbi.nlm.nih.gov/29249689/
Giacalone S, Spigariolo CB, Bortoluzzi P, Nazzaro G. Oral nicotinamide: the role in skin cancer chemoprevention. Dermatol Ther. 2021;34(3):e14892. https://pubmed.ncbi.nlm.nih.gov/33595161/
Kelly G. A review of the sirtuin system, its clinical implications, and the potential role of dietary activators like resveratrol: part 1. Altern Med Rev. 2010;15(3):245–63. https://pubmed.ncbi.nlm.nih.gov/21155626/
Morris BJ. Seven sirtuins for seven deadly diseases of aging. Free Radic Biol Med. 2013;56:133–71. https://pubmed.ncbi.nlm.nih.gov/23104101/
Schmeisser K, Mansfeld J, Kuhlow D, et al. Role of sirtuins in lifespan regulation is linked to methylation of nicotinamide. Nat Chem Biol. 2013;9(11):693–700. https://pubmed.ncbi.nlm.nih.gov/24077178/
Mitchell SJ, Bernier M, Aon MA, et al. Nicotinamide improves aspects of healthspan, but not lifespan, in mice. Cell Metab. 2018;27(3):667–76.e4. https://pubmed.ncbi.nlm.nih.gov/29514072/
Elliott RB, Pilcher CC, Stewart A, Fergusson D, McGregor MA. The use of nicotinamide in the prevention of type 1 diabetes. Ann N Y Acad Sci. 1993;696:333–41. https://pubmed.ncbi.nlm.nih.gov/8109840/
Pozzilli P, Browne PD, Kolb H, et al. Meta-analysis of nicotinamide treatment in patients with recent-onset IDDM. Diabetes Care. 1996;19(12):1357–63. https://pubmed.ncbi.nlm.nih.gov/8941464/
Connell NJ, Grevendonk L, Fealy CE, et al. NAD+-precursor supplementation with L-tryptophan, nicotinic acid, and nicotinamide does not affect mitochondrial function or skeletal muscle function in physically compromised older adults. J Nutr. 2021;151(10):2917–31. https://pubmed.ncbi.nlm.nih.gov/34191033/
Gasperi V, Sibilano M, Savini I, Catani MV. Niacin in the central nervous system: an update of biological aspects and clinical applications. Int J Mol Sci. 2019;20(4):974. https://pubmed.ncbi.nlm.nih.gov/30813414/
Winter SL, Boyer JL. Hepatic toxicity from large doses of vitamin B3 (nicotinamide). N Engl J Med. 1973;289(22):1180–2. https://pubmed.ncbi.nlm.nih.gov/4271091/
Reddi KK, Kodicek E. Metabolism of nicotinic acid and related compounds in man and rat. Biochem J. 1953;53(2):286–94. https://pubmed.ncbi.nlm.nih.gov/13032067/
Braidy N, Liu Y. NAD+ therapy in age-related degenerative disorders: a benefit/risk analysis. Exp Gerontol. 2020;132:110831. https://pubmed.ncbi.nlm.nih.gov/31917996/
Willets JM, Lunec J, Williams AC, Griffiths HR. Neurotoxicity of nicotinamide derivatives: their role in the aetiology of Parkinson’s disease. Biochem Soc Trans. 1993;21 (Pt 3)(3):299S. https://pubmed.ncbi.nlm.nih.gov/8224447/
Harrison IF, Powell NM, Dexter DT. The histone deacetylase inhibitor nicotinamide exacerbates neurodegeneration in the lactacystin rat model of Parkinson’s disease. J Neurochem. 2019;148(1):136–56. https://pubmed.ncbi.nlm.nih.gov/30269333/
Parsons RB, Smith ML, Williams AC, Waring RH, Ramsden DB. Expression of nicotinamide N-methyltransferase (E.C. 2.1.1.1) in the Parkinsonian brain. J Neuropathol Exp Neurol. 2002;61(2):111–24. https://pubmed.ncbi.nlm.nih.gov/11853016/
Li D, Tian YJ, Guo J, et al. Nicotinamide supplementation induces detrimental metabolic and epigenetic changes in developing rats. Br J Nutr. 2013;110(12):2156–64. https://pubmed.ncbi.nlm.nih.gov/23768418/
Kang-Lee YA, McKee RW, Wright SM, Swendseid ME, Jenden DJ, Jope RS. Metabolic effects of nicotinamide administration in rats. J Nutr. 1983;113(2):215–21. https://pubmed.ncbi.nlm.nih.gov/6218261/
Hwang ES, Song SB. Possible adverse effects of high-dose nicotinamide: mechanisms and safety assessment. Biomolecules. 2020;10(5):687. https://pubmed.ncbi.nlm.nih.gov/32365524/
Tian YJ, Li D, Ma Q, et al. Excess nicotinamide increases plasma serotonin and histamine levels. Sheng Li Xue Bao. 2013;65(1):33–8. https://pubmed.ncbi.nlm.nih.gov/23426511/
Sun WP, Li D, Lun YZ, et al. Excess nicotinamide inhibits methylation-mediated degradation of catecholamines in normotensives and hypertensives. Hypertens Res. 2012;35(2):180–5. https://pubmed.ncbi.nlm.nih.gov/21918528/
Tinelli C, Di Pino A, Ficulle E, Marcelli S, Feligioni M. Hyperhomocysteinemia as a risk factor and potential nutraceutical target for certain pathologies. Front Nutr. 2019;6:49. https://pubmed.ncbi.nlm.nih.gov/31069230/
Avalos JL, Bever KM, Wolberger C. Mechanism of sirtuin inhibition by nicotinamide: altering the NAD+ cosubstrate specificity of a Sir2 enzyme. Mol Cell. 2005;17(6):855–68. https://pubmed.ncbi.nlm.nih.gov/15780941/
Mitchell SJ, Bernier M, Aon MA, et al. Nicotinamide improves aspects of healthspan, but not lifespan, in mice. Cell Metab. 2018;27(3):667–76.e4. https://pubmed.ncbi.nlm.nih.gov/29514072/
Bitterman KJ, Anderson RM, Cohen HY, Latorre-Esteves M, Sinclair DA. Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast Sir2 and human SIRT1. J Biol Chem. 2002;277(47):45099–107. https://pubmed.ncbi.nlm.nih.gov/12297502/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
Cantó C, Houtkooper RH, Pirinen E, et al. The NAD+ precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab. 2012;15(6):838–47. https://pubmed.ncbi.nlm.nih.gov/22682224/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol. 2022;198:114946. https://pubmed.ncbi.nlm.nih.gov/35134387/
Conze D, Brenner C, Kruger CL. Safety and metabolism of long-term administration of NIAGEN (Nicotinamide riboside chloride) in a randomized, double-blind, placebo-controlled clinical trial of healthy overweight adults. Sci Rep. 2019;9(1):9772. https://pubmed.ncbi.nlm.nih.gov/31278280/
Elhassan YS, Kluckova K, Fletcher RS, et al. Nicotinamide riboside augments the aged human skeletal muscle NAD+ metabolome and induces transcriptomic and anti-inflammatory signatures. Cell Rep. 2019;28(7):1717–28.e6. https://pubmed.ncbi.nlm.nih.gov/31412242/
Dollerup OL, Chubanava S, Agerholm M, et al. Nicotinamide riboside does not alter mitochondrial respiration, content or morphology in skeletal muscle from obese and insulin-resistant men. J Physiol. 2020;598(4):731–54. https://pubmed.ncbi.nlm.nih.gov/31710095/
Remie CME, Roumans KHM, Moonen MPB, et al. Nicotinamide riboside supplementation alters body composition and skeletal muscle acetylcarnitine concentrations in healthy obese humans. Am J Clin Nutr. 2020;112(2):413–26. https://pubmed.ncbi.nlm.nih.gov/32320006/
Stocks B, Ashcroft SP, Joanisse S, et al. Nicotinamide riboside supplementation does not alter whole-body or skeletal muscle metabolic responses to a single bout of endurance exercise. J Physiol. 2021;599(5):1513–31. https://pubmed.ncbi.nlm.nih.gov/33492681/
Mehmel M, Jovanovic N, Spitz U. Nicotinamide riboside – the current state of research and therapeutic uses. Nutrients. 2020;12(6):1616. https://pubmed.ncbi.nlm.nih.gov/32486488/
Katsyuba E, Romani M, Hofer D, Auwerx J. NAD+ homeostasis in health and disease. Nat Metab. 2020;2(1):9–31. https://pubmed.ncbi.nlm.nih.gov/32694684/
Martens CR, Denman BA, Mazzo MR, et al. Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat Commun. 2018;9(1):1286. https://pubmed.ncbi.nlm.nih.gov/29599478/
Dolopikou CF, Kourtzidis IA, Margaritelis NV, et al. Acute nicotinamide riboside supplementation improves redox homeostasis and exercise performance in old individuals: a double-blind cross-over study. Eur J Nutr. 2020;59(2):505–15. https://pubmed.ncbi.nlm.nih.gov/30725213/
Martens CR, Denman BA, Mazzo MR, et al. Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat Commun. 2018;9(1):1286. https://pubmed.ncbi.nlm.nih.gov/29599478/
Dolopikou CF, Kourtzidis IA, Margaritelis NV, et al. Acute nicotinamide riboside supplementation improves redox homeostasis and exercise performance in old individuals: a double-blind cross-over study. Eur J Nutr. 2020;59(2):505–15. https://pubmed.ncbi.nlm.nih.gov/30725213/
Elhassan YS, Kluckova K, Fletcher RS, et al. Nicotinamide riboside augments the aged human skeletal muscle NAD+ metabolome and induces transcriptomic and anti-inflammatory signatures. Cell Rep. 2019;28(7):1717–28.e6. https://pubmed.ncbi.nlm.nih.gov/31412242/
Remie CME, Roumans KHM, Moonen MPB, et al. Nicotinamide riboside supplementation alters body composition and skeletal muscle acetylcarnitine concentrations in healthy obese humans. Am J Clin Nutr. 2020;112(2):413–26. https://pubmed.ncbi.nlm.nih.gov/32320006/
Martens CR, Denman BA, Mazzo MR, et al. Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat Commun. 2018;9(1):1286. https://pubmed.ncbi.nlm.nih.gov/29599478/
Dolopikou CF, Kourtzidis IA, Margaritelis NV, et al. Acute nicotinamide riboside supplementation improves redox homeostasis and exercise performance in old individuals: a double-blind cross-over study. Eur J Nutr. 2020;59(2):505–15. https://pubmed.ncbi.nlm.nih.gov/30725213/
Remie CME, Roumans KHM, Moonen MPB, et al. Nicotinamide riboside supplementation alters body composition and skeletal muscle acetylcarnitine concentrations in healthy obese humans. Am J Clin Nutr. 2020;112(2):413–26. https://pubmed.ncbi.nlm.nih.gov/32320006/
Martens CR, Denman BA, Mazzo MR, et al. Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat Commun. 2018;9(1):1286. https://pubmed.ncbi.nlm.nih.gov/29599478/
Dolopikou CF, Kourtzidis IA, Margaritelis NV, et al. Acute nicotinamide riboside supplementation improves redox homeostasis and exercise performance in old individuals: a double-blind cross-over study. Eur J Nutr. 2020;59(2):505–15. https://pubmed.ncbi.nlm.nih.gov/30725213/
Elhassan YS, Kluckova K, Fletcher RS, et al. Nicotinamide riboside augments the aged human skeletal muscle NAD+ metabolome and induces transcriptomic and anti-inflammatory signatures. Cell Rep. 2019;28(7):1717–28.e6. https://pubmed.ncbi.nlm.nih.gov/31412242/
Remie CME, Roumans KHM, Moonen MPB, et al. Nicotinamide riboside supplementation alters body composition and skeletal muscle acetylcarnitine concentrations in healthy obese humans. Am J Clin Nutr. 2020;112(2):413–26. https://pubmed.ncbi.nlm.nih.gov/32320006/
Dollerup OL, Christensen B, Svart M, et al. A randomized placebo-controlled clinical trial of nicotinamide riboside in obese men: safety, insulin-sensitivity, and lipid-mobilizing effects. Am J Clin Nutr. 2018;108(2):343–53. https://pubmed.ncbi.nlm.nih.gov/29992272/
Stocks B, Ashcroft SP, Joanisse S, et al. Nicotinamide riboside supplementation does not alter whole-body or skeletal muscle metabolic responses to a single bout of endurance exercise. J Physiol. 2021;599(5):1513–31. https://pubmed.ncbi.nlm.nih.gov/33492681/
Dollerup OL, Chubanava S, Agerholm M, et al. Nicotinamide riboside does not alter mitochondrial respiration, content or morphology in skeletal muscle from obese and insulin-resistant men. J Physiol. 2020;598(4):731–54. https://pubmed.ncbi.nlm.nih.gov/31710095/
Elhassan YS, Kluckova K, Fletcher RS, et al. Nicotinamide riboside augments the aged human skeletal muscle NAD+ metabolome and induces transcriptomic and anti-inflammatory signatures. Cell Rep. 2019;28(7):1717–28.e6. https://pubmed.ncbi.nlm.nih.gov/31412242/
Martens CR, Denman BA, Mazzo MR, et al. Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat Commun. 2018;9(1):1286. https://pubmed.ncbi.nlm.nih.gov/29599478/
Dolopikou CF, Kourtzidis IA, Margaritelis NV, et al. Acute nicotinamide riboside supplementation improves redox homeostasis and exercise performance in old individuals: a double-blind cross-over study. Eur J Nutr. 2020;59(2):505–15. https://pubmed.ncbi.nlm.nih.gov/30725213/
Dollerup OL, Trammell SAJ, Hartmann B, et al. Effects of nicotinamide riboside on endocrine pancreatic function and incretin hormones in nondiabetic men with obesity. J Clin Endocrinol Metab. 2019;104(11):5703–14. https://pubmed.ncbi.nlm.nih.gov/31390002/
Brakedal B, Dölle C, Riemer F, et al. The NADPARK study: a randomized phase I trial of nicotinamide riboside supplementation in Parkinson’s disease. Cell Metab. 2022;34(3):396–407.e6. https://pubmed.ncbi.nlm.nih.gov/35235774/
Martens CR, Denman BA, Mazzo MR, et al. Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat Commun. 2018;9(1):1286. https://pubmed.ncbi.nlm.nih.gov/29599478/
Dolopikou CF, Kourtzidis IA, Margaritelis NV, et al. Acute nicotinamide riboside supplementation improves redox homeostasis and exercise performance in old individuals: a double-blind cross-over study. Eur J Nutr. 2020;59(2):505–15. https://pubmed.ncbi.nlm.nih.gov/30725213/
Brenner C. Anti-inflammatory plus this observation among the men. Twitter. https://twitter.com/charlesmbrenner/status/1161314766176083968. Published August 14, 2019. Accessed January 13, 2023.; https://twitter.com/charlesmbrenner/status/1161314766176083968
Elhassan YS, Kluckova K, Fletcher RS, et al. Nicotinamide riboside augments the aged human skeletal muscle NAD+ metabolome and induces transcriptomic and anti-inflammatory signatures. Cell Rep. 2019;28(7):1717–28.e6. https://pubmed.ncbi.nlm.nih.gov/31412242/
Remie CME, Roumans KHM, Moonen MPB, et al. Nicotinamide riboside supplementation alters body composition and skeletal muscle acetylcarnitine concentrations in healthy obese humans. Am J Clin Nutr. 2020;112(2):413–26. https://pubmed.ncbi.nlm.nih.gov/32320006/
Chi Y, Sauve AA. Nicotinamide riboside, a trace nutrient in foods, is a Vitamin B3 with effects on energy metabolism and neuroprotection. Curr Opin Clin Nutr Metab Care. 2013;16(6):657–61. https://pubmed.ncbi.nlm.nih.gov/24071780/
Katsyuba E, Romani M, Hofer D, Auwerx J. NAD+ homeostasis in health and disease. Nat Metab. 2020;2(1):9–31. https://pubmed.ncbi.nlm.nih.gov/32694684/
Stocks B, Ashcroft SP, Joanisse S, et al. Nicotinamide riboside supplementation does not alter whole-body or skeletal muscle metabolic responses to a single bout of endurance exercise. J Physiol. 2021;599(5):1513–31. https://pubmed.ncbi.nlm.nih.gov/33492681/
Campbell MTD, Jones DS, Andrews GP, Li S. Understanding the physicochemical properties and degradation kinetics of nicotinamide riboside, a promising vitamin B3 nutritional supplement. Food Nutr Res. 2019;63. https://pubmed.ncbi.nlm.nih.gov/31807125/
Shats I, Williams JG, Liu J, et al. Bacteria boost mammalian host NAD metabolism by engaging the deamidated biosynthesis pathway. Cell Metab. 2020;31(3):564–79.e7. https://pubmed.ncbi.nlm.nih.gov/32130883/
Stocks B, Ashcroft SP, Joanisse S, et al. Nicotinamide riboside supplementation does not alter whole-body or skeletal muscle metabolic responses to a single bout of endurance exercise. J Physiol. 2021;599(5):1513–31. https://pubmed.ncbi.nlm.nih.gov/33492681/
Dollerup OL, Chubanava S, Agerholm M, et al. Nicotinamide riboside does not alter mitochondrial respiration, content or morphology in skeletal muscle from obese and insulin-resistant men. J Physiol. 2020;598(4):731–54. https://pubmed.ncbi.nlm.nih.gov/31710095/
Sauve AA. Metabolic disease, NAD metabolism, nicotinamide riboside, and the gut microbiome: connecting the dots from the gut to physiology. mSystems. 2022;7(1):e01223–21. https://pubmed.ncbi.nlm.nih.gov/35076274/
Conze D, Brenner C, Kruger CL. Safety and metabolism of long-term administration of NIAGEN (Nicotinamide riboside chloride) in a randomized, double-blind, placebo-controlled clinical trial of healthy overweight adults. Sci Rep. 2019;9(1):9772. https://pubmed.ncbi.nlm.nih.gov/31278280/
Dellinger RW, Santos SR, Morris M, et al. Repeat dose NRPT (nicotinamide riboside and pterostilbene) increases NAD+ levels in humans safely and sustainably: a randomized, double-blind, placebo-controlled study. NPJ Aging Mech Dis. 2017;3:17. https://pubmed.ncbi.nlm.nih.gov/29184669/
Wolf AM. Rodent diet aids and the fallacy of caloric restriction. Mech Ageing Dev. 2021;200:111584. https://pubmed.ncbi.nlm.nih.gov/34673082/
Brenner C, Boileau AC. Pterostilbene raises low density lipoprotein cholesterol in people. Clin Nutr. 2019;38(1):480–1. https://pubmed.ncbi.nlm.nih.gov/30482564/
Martens CR, Denman BA, Mazzo MR, et al. Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat Commun. 2018;9(1):1286. https://pubmed.ncbi.nlm.nih.gov/29599478/
Dollerup OL, Christensen B, Svart M, et al. A randomized placebo-controlled clinical trial of nicotinamide riboside in obese men: safety, insulin-sensitivity, and lipid-mobilizing effects. Am J Clin Nutr. 2018;108(2):343–53. https://pubmed.ncbi.nlm.nih.gov/29992272/
Riche DM, Riche KD, Blackshear CT, et al. Pterostilbene on metabolic parameters: a randomized, double-blind, and placebo-controlled trial. Evid Based Complement Alternat Med. 2014;2014:459165. https://pubmed.ncbi.nlm.nih.gov/25057276/
Airhart SE, Shireman LM, Risler LJ, et al. An open-label, non-randomized study of the pharmacokinetics of the nutritional supplement nicotinamide riboside (NR) and its effects on blood NAD+ levels in healthy volunteers. PLoS One. 2017;12(12):e0186459. https://pubmed.ncbi.nlm.nih.gov/29211728/
Palmer RD, Elnashar MM, Vaccarezza M. Precursor comparisons for the upregulation of nicotinamide adenine dinucleotide. Novel approaches for better aging. Aging Med (Milton). 2021;4(3):214–20. https://pubmed.ncbi.nlm.nih.gov/34553119/
Kourtzidis IA, Stoupas AT, Gioris IS, et al. The NAD+ precursor nicotinamide riboside decreases exercise performance in rats. J Int Soc Sports Nutr. 2016;13:32. https://pubmed.ncbi.nlm.nih.gov/27489522/
Kourtzidis IA, Dolopikou CF, Tsiftsis AN, et al. Nicotinamide riboside supplementation dysregulates redox and energy metabolism in rats: implications for exercise performance. Exp Physiol. 2018;103(10):1357–66. https://pubmed.ncbi.nlm.nih.gov/30007015/
Shi W, Hegeman MA, Doncheva A, Bekkenkamp-Grovenstein M, de Boer VCJ, Keijer J. High dose of dietary nicotinamide riboside induces glucose intolerance and white adipose tissue dysfunction in mice fed a mildly obesogenic diet. Nutrients. 2019;11(10):2439. https://pubmed.ncbi.nlm.nih.gov/31614949/
Sun P, Qie S, Pan B. Nicotinamide riboside will play an important role in anti-aging therapy in humans, especially in the face skin anti-aging treatment. Aesthetic Plast Surg. 2022;46(Suppl 1):192–4. https://pubmed.ncbi.nlm.nih.gov/33977340/
Turck D, Castenmiller J, de Henauw S, et al. Safety of nicotinamide riboside chloride as a novel food pursuant to Regulation (EU) 2015/2283 and bioavailability of nicotinamide from this source, in the context of Directive 2002/46/EC. EFSA J. 2019;17(8):5775. https://pubmed.ncbi.nlm.nih.gov/32626405/
Leduc-Gaudet JP, Dulac M, Reynaud O, Ayoub MB, Gouspillou G. Nicotinamide riboside supplementation to improve skeletal muscle mitochondrial health and whole-body glucose homeostasis: does it actually work in humans? J Physiol. 2020;598(4):619–20. https://pubmed.ncbi.nlm.nih.gov/31879956/
Yoshino J, Baur JA, Imai SI. NAD+ intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab. 2018;27(3):513–28. https://pubmed.ncbi.nlm.nih.gov/29249689/
Okabe K, Yaku K, Uchida Y, et al. Oral administration of nicotinamide mononucleotide is safe and efficiently increases blood nicotinamide adenine dinucleotide levels in healthy subjects. Front Nutr. 2022;9:868640. https://pubmed.ncbi.nlm.nih.gov/35479740/
Airhart SE, Shireman LM, Risler LJ, et al. An open-label, non-randomized study of the pharmacokinetics of the nutritional supplement nicotinamide riboside (NR) and its effects on blood NAD+ levels in healthy volunteers. PLoS One. 2017;12(12):e0186459. https://pubmed.ncbi.nlm.nih.gov/29211728/
Soma M, Lalam SK. The role of nicotinamide mononucleotide (NMN) in anti-aging, longevity, and its potential for treating chronic conditions. Mol Biol Rep. 2022;49(10):9737–48. https://pubmed.ncbi.nlm.nih.gov/35441939/
Poddar SK, Sifat AE, Haque S, Nahid NA, Chowdhury S, Mehedi I. Nicotinamide mononucleotide: exploration of diverse therapeutic applications of a potential molecule. Biomolecules. 2019;9(1):34. https://pubmed.ncbi.nlm.nih.gov/30669679/
Schmidt MS, Brenner C. Absence of evidence that Slc12a8 encodes a nicotinamide mononucleotide transporter. Nat Metab. 2019;1(7):660–1. https://pubmed.ncbi.nlm.nih.gov/32694648/
Grozio A, Mills KF, Yoshino J, et al. Slc12a8 is a nicotinamide mononucleotide transporter. Nat Metab. 2019;1(1):47–57. https://pubmed.ncbi.nlm.nih.gov/31131364/
Mills KF, Yoshida S, Stein LR, et al. Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab. 2016;24(6):795–806. https://pubmed.ncbi.nlm.nih.gov/28068222/
Zhang H, Ryu D, Wu Y, et al. NAD¿ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science. 2016;352(6292):1436–43. https://pubmed.ncbi.nlm.nih.gov/27127236/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
Irie J, Inagaki E, Fujita M, et al. Effect of oral administration of nicotinamide mononucleotide on clinical parameters and nicotinamide metabolite levels in healthy Japanese men. Endocr J. 2020;67(2):153–60. https://pubmed.ncbi.nlm.nih.gov/31685720/
Okabe K, Yaku K, Uchida Y, et al. Oral administration of nicotinamide mononucleotide is safe and efficiently increases blood nicotinamide adenine dinucleotide levels in healthy subjects. Front Nutr. 2022;9:868640. https://pubmed.ncbi.nlm.nih.gov/35479740/
Yoshino M, Yoshino J, Kayser BD, et al. Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science. 2021;372(6547):1224–9. https://pubmed.ncbi.nlm.nih.gov/33888596/
Liao B, Zhao Y, Wang D, Zhang X, Hao X, Hu M. Nicotinamide mononucleotide supplementation enhances aerobic capacity in amateur runners: a randomized, double-blind study. J Int Soc Sports Nutr. 2021;18(1):54. https://pubmed.ncbi.nlm.nih.gov/34238308/
Kim M, Seol J, Sato T, Fukamizu Y, Sakurai T, Okura T. Effect of 12-week intake of nicotinamide mononucleotide on sleep quality, fatigue, and physical performance in older Japanese adults: a randomized, double-blind placebo-controlled study. Nutrients. 2022;14(4):755. https://pubmed.ncbi.nlm.nih.gov/35215405/
Yoshino M, Yoshino J, Kayser BD, et al. Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science. 2021;372(6547):1224–9. https://pubmed.ncbi.nlm.nih.gov/33888596/
Abdellatif M, Baur JA. NAD+ metabolism and cardiometabolic health: the human evidence. Cardiovasc Res. 2021;117(9):e106–9. https://pubmed.ncbi.nlm.nih.gov/34320167/
Yoshino M, Yoshino J, Kayser BD, et al. Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science. 2021;372(6547):1224–9. https://pubmed.ncbi.nlm.nih.gov/33888596/
Benson D. Christopher W. Shade, PhD: nicotinamide mononucleotide. Integr Med (Encinitas). 2019;18(6):42–4. https://pubmed.ncbi.nlm.nih.gov/32549856/
Shade C. The science behind NMN – a stable, reliable NAD+ activator and anti-aging molecule. Integr Med (Encinitas). 2020;19(1):12–4. https://pubmed.ncbi.nlm.nih.gov/32549859/
Mills KF, Yoshida S, Stein LR, et al. Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab. 2016;24(6):795–806. https://pubmed.ncbi.nlm.nih.gov/28068222/
Ummarino S, Mozzon M, Zamporlini F, et al. Simultaneous quantitation of nicotinamide riboside, nicotinamide mononucleotide and nicotinamide adenine dinucleotide in milk by a novel enzyme-coupled assay. Food Chem. 2017;221:161–8. https://pubmed.ncbi.nlm.nih.gov/27979136/
Turner J, Licollari A, Mihalcea E, Tan A. Safety evaluation for Restorin® NMN, a NAD+ precursor. Front Pharmacol. 2021;12:749727. https://pubmed.ncbi.nlm.nih.gov/34867355/
You Y, Gao Y, Wang H, et al. Subacute toxicity study of nicotinamide mononucleotide via oral administration. Front Pharmacol. 2020;11:604404. https://pubmed.ncbi.nlm.nih.gov/33384603/
Poddar SK, Sifat AE, Haque S, Nahid NA, Chowdhury S, Mehedi I. Nicotinamide mononucleotide: exploration of diverse therapeutic applications of a potential molecule. Biomolecules. 2019;9(1):34. https://pubmed.ncbi.nlm.nih.gov/30669679/
NDI 1259-B-Nicotinamide Mononucleotide (NMN) from Inner Mongolia Kingdomway Pharmaceutical Limited. U.S. Food and Drug Administration. https://www.regulations.gov/document/FDA-2022-S-0023–0051. Published November 8, 2022. Accessed February 25, 2023.; https://www.regulations.gov/document/FDA-2022-S-0023-0051
Ramsey KM, Mills KF, Satoh A, Imai SI. Age-associated loss of Sirt1-mediated enhancement of glucose-stimulated insulin secretion in beta cell-specific Sirt1-overexpressing (BESTO) mice. Aging Cell. 2008;7(1):78–88. https://pubmed.ncbi.nlm.nih.gov/18005249/
Li C, Wu LE. Risks and rewards of targeting NAD+ homeostasis in the brain. Mech Ageing Dev. 2021;198:111545. https://pubmed.ncbi.nlm.nih.gov/34302821/
Braidy N, Liu Y. NAD+ therapy in age-related degenerative disorders: a benefit/risk analysis. Exp Gerontol. 2020;132:110831. https://pubmed.ncbi.nlm.nih.gov/31917996/
Cohen MS. Axon degeneration: too much NMN is actually bad? Curr Biol. 2017;27(8):R310–2. https://pubmed.ncbi.nlm.nih.gov/28441566/
Williams PA, Harder JM, John SWM. Glaucoma as a metabolic optic neuropathy: making the case for nicotinamide treatment in glaucoma. J Glaucoma. 2017;26(12):1161–8. https://pubmed.ncbi.nlm.nih.gov/28858158/
Di Stefano M, Nascimento-Ferreira I, Orsomando G, et al. A rise in NAD precursor nicotinamide mononucleotide (NMN) after injury promotes axon degeneration. Cell Death Differ. 2015;22(5):731–42. https://pubmed.ncbi.nlm.nih.gov/25323584/
Di Stefano M, Loreto A, Orsomando G, et al. NMN deamidase delays Wallerian degeneration and rescues axonal defects caused by NMNAT2 deficiency in vivo. Curr Biol. 2017;27(6):784–94. https://pubmed.ncbi.nlm.nih.gov/28262487/
Cohen MS. Axon degeneration: too much NMN is actually bad? Curr Biol. 2017;27(8):R310–2. https://pubmed.ncbi.nlm.nih.gov/28441566/
Quantitative analysis of twenty-two NMN consumer products. ChromaDex. https://s23.q4cdn.com/937095816/files/doc_downloads/2021/Quantitative-Analysis-of-22-NMN-Consumer-Products-Oct-2021.pdf. Published October 20, 2021. Accessed January 10, 2023.; https://investors.chromadex.com/investor-resources/Market-Surveillance/default.aspx
Cooperman T. NAD booster supplements review (NAD+/NADH, nicotinamide riboside, and NMN). ConsumerLab.com. https://www.consumerlab.com/reviews/nmn-nadh-nicotinamide-riboside/nmn-nadh-nicotinamide-riboside/#related-clinical-updates. Published November 2, 2021. Updated November 14, 2022. Accessed January 11, 2023.; https://www.consumerlab.com/reviews/nmn-nadh-nicotinamide-riboside/nmn-nadh-nicotinamide-riboside/#related-clinical-updates
Correll WA, Viswanathan S. Warning letter: ChromaDex MARCS-CMS 607692–11/17/2020. U.S. Food and Drug Administration. https://www.fda.gov/inspections-compliance-enforcement-and-criminal-investigations/warning-letters/chromadex-607692–11172020. Updated December 1, 2020. Accessed January 10, 2023.; https://www.fda.gov/inspections-compliance-enforcement-and-criminal-investigations/warning-letters/chromadex-607692-11172020
BBB National Programs. ChromaDex, Inc. Discontinues advertising claims for Tru Niagen dietary supplement following national advertising division challenge. Cision PR Newswire. https://www.prnewswire.com/news-releases/chromadex-inc-discontinues-advertising-claims-for-tru-niagen-dietary-supplement-following-national-advertising-division-challenge-301392733.html. Published October 5, 2021. Accessed January 10, 2023.; https://www.prnewswire.com/news-releases/chromadex-inc-discontinues-advertising-claims-for-tru-niagen-dietary-supplement-following-national-advertising-division-challenge-301392733.html
Segall PE, Timiras PS. Patho-physiologic findings after chronic tryptophan deficiency in rats: a model for delayed growth and aging. Mech Ageing Dev. 1976;5(2):109–24. https://pubmed.ncbi.nlm.nih.gov/933560/
De Marte ML, Enesco HE. Influence of low tryptophan diet on survival and organ growth in mice. Mech Ageing Dev. 1986;36(2):161–71. https://pubmed.ncbi.nlm.nih.gov/3784629/
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol. 2022;198:114946. https://pubmed.ncbi.nlm.nih.gov/35134387/
Kimura N, Fukuwatari T, Sasaki R, Shibata K. Comparison of metabolic fates of nicotinamide, NAD+ and NADH administered orally and intraperitoneally; characterization of oral NADH. J Nutr Sci Vitaminol. 2006;52(2):142–8. https://pubmed.ncbi.nlm.nih.gov/16802695/
Zapata-Pérez R, Tammaro A, Schomakers BV, et al. Reduced nicotinamide mononucleotide is a new and potent NAD+ precursor in mammalian cells and mice. FASEB J. 2021;35(4):e21456. https://pubmed.ncbi.nlm.nih.gov/33724555/
Giroud-Gerbetant J, Joffraud M, Giner MP, et al. A reduced form of nicotinamide riboside defines a new path for NAD+ biosynthesis and acts as an orally bioavailable NAD+ precursor. Mol Metab. 2019;30:192–202. https://pubmed.ncbi.nlm.nih.gov/31767171/
Chini CCS, Peclat TR, Gomez LS, et al. Dihydronicotinamide riboside is a potent NAD+ precursor promoting a pro-inflammatory phenotype in macrophages. Front Immunol. 2022;13:840246. https://pubmed.ncbi.nlm.nih.gov/35281060/
Sonavane M, Hayat F, Makarov M, Migaud ME, Gassman NR. Dihydronicotinamide riboside promotes cell-specific cytotoxicity by tipping the balance between metabolic regulation and oxidative stress. PLoS One. 2020;15(11):e0242174. https://pubmed.ncbi.nlm.nih.gov/33166357/
Chini CCS, Peclat TR, Gomez LS, et al. Dihydronicotinamide riboside is a potent NAD+ precursor promoting a pro-inflammatory phenotype in macrophages. Front Immunol. 2022;13:840246. https://pubmed.ncbi.nlm.nih.gov/35281060/
Poljsak B, Kovac V, Milisav I. Healthy lifestyle recommendations: do the beneficial effects originate from NAD+ amount at the cellular level? Oxid Med Cell Longev. 2020;2020:8819627. https://pubmed.ncbi.nlm.nih.gov/33414897/
Oakey LA, Fletcher RS, Elhassan YS, et al. Metabolic tracing reveals novel adaptations to skeletal muscle cell energy production pathways in response to NAD+ depletion. Wellcome Open Res. 2018;3:147. https://pubmed.ncbi.nlm.nih.gov/30607371/
Braidy N, Liu Y. NAD+ therapy in age-related degenerative disorders: a benefit/risk analysis. Exp Gerontol. 2020;132:110831. https://pubmed.ncbi.nlm.nih.gov/31917996/
Palmer RD, Vaccarezza M. Nicotinamide adenine dinucleotide and the sirtuins caution: pro-cancer functions. Aging Med (Milton). 2021;4(4):337–44. https://pubmed.ncbi.nlm.nih.gov/34964015/
Poljsak B, Kovac V, Milisav I. Healthy lifestyle recommendations: do the beneficial effects originate from NAD+ amount at the cellular level? Oxid Med Cell Longev. 2020;2020:8819627. https://pubmed.ncbi.nlm.nih.gov/33414897/
Liu Y, Clement J, Grant R, Sachdev P, Braidy N. Quantitation of NAD+: why do we need to measure it? Biochim Biophys Acta Gen Subj. 2018;1862(12):2527–32. https://pubmed.ncbi.nlm.nih.gov/30048742/
Chini EN. Of mice and men: NAD+ boosting with niacin provides hope for mitochondrial myopathy patients. Cell Metab. 2020;31(6):1041–3. https://pubmed.ncbi.nlm.nih.gov/32492387/
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol. 2022;198:114946. https://pubmed.ncbi.nlm.nih.gov/35134387/
McReynolds MR, Chellappa K, Baur JA. Age-related NAD+ decline. Exp Gerontol. 2020;134:110888. https://pubmed.ncbi.nlm.nih.gov/32097708/
Katsyuba E, Romani M, Hofer D, Auwerx J. NAD+ homeostasis in health and disease. Nat Metab. 2020;2(1):9–31. https://pubmed.ncbi.nlm.nih.gov/32694684/
Katsyuba E, Romani M, Hofer D, Auwerx J. NAD+ homeostasis in health and disease. Nat Metab. 2020;2(1):9–31. https://pubmed.ncbi.nlm.nih.gov/32694684/
Elysium Health. U. S. District Court invalidates Dartmouth patents asserted by ChromaDex. Cision PR Newswire. https://www.prnewswire.com/news-releases/us-district-court-invalidates-dartmouth-patents-asserted-by-chromadex-301381257.html. Published September 21, 2021. Accessed January 28, 2023.; https://www.prnewswire.com/news-releases/us-district-court-invalidates-dartmouth-patents-asserted-by-chromadex-301381257.html
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol. 2022;198:114946. https://pubmed.ncbi.nlm.nih.gov/35134387/
Rajman L, Chwalek K, Sinclair DA. Therapeutic potential of NAD-boosting molecules: the in vivo evidence. Cell Metab. 2018;27(3):529–47. https://pubmed.ncbi.nlm.nih.gov/29514064/
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol. 2022;198:114946. https://pubmed.ncbi.nlm.nih.gov/35134387/
de Guia RM, Agerholm M, Nielsen TS, et al. Aerobic and resistance exercise training reverses age-dependent decline in NAD+ salvage capacity in human skeletal muscle. Physiol Rep. 2019;7(12):e14139. https://pubmed.ncbi.nlm.nih.gov/31207144/
Zhou CC, Yang X, Hua X, et al. Hepatic NAD+ deficiency as a therapeutic target for non-alcoholic fatty liver disease in ageing. Br J Pharmacol. 2016;173(15):2352–68. https://pubmed.ncbi.nlm.nih.gov/27174364/
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol. 2022;198:114946. https://pubmed.ncbi.nlm.nih.gov/35134387/
Koltai E, Szabo Z, Atalay M, et al. Exercise alters SIRT1, SIRT6, NAD and NAMPT levels in skeletal muscle of aged rats. Mech Ageing Dev. 2010;131(1):21–8. https://pubmed.ncbi.nlm.nih.gov/19913571/
Liu LY, Wang F, Zhang XY, et al. Nicotinamide phosphoribosyltransferase may be involved in age-related brain diseases. PLoS One. 2012;7(10):e44933. https://pubmed.ncbi.nlm.nih.gov/23071504/
Anderson RM, Bitterman KJ, Wood JG, Medvedik O, Sinclair DA. Nicotinamide and PNC1 govern lifespan extension by calorie restriction in Saccharomyces cerevisiae. Nature. 2003;423(6936):181–5. https://pubmed.ncbi.nlm.nih.gov/12736687/
Balan V, Miller GS, Kaplun L, et al. Life span extension and neuronal cell protection by Drosophila nicotinamidase. J Biol Chem. 2008;283(41):27810–9. https://pubmed.ncbi.nlm.nih.gov/18678867/
Yoshida M, Satoh A, Lin JB, et al. Extracellular vesicle-contained eNAMPT delays aging and extends lifespan in mice. Cell Metab. 2019;30(2):329–42.e5. https://pubmed.ncbi.nlm.nih.gov/31204283/
Brouwers B, Stephens NA, Costford SR, et al. Elevated nicotinamide phosphoribosyl transferase in skeletal muscle augments exercise performance and mitochondrial respiratory capacity following exercise training. Front Physiol. 2018;9:704. https://pubmed.ncbi.nlm.nih.gov/29942262/
Costford SR, Brouwers B, Hopf ME, et al. Skeletal muscle overexpression of nicotinamide phosphoribosyl transferase in mice coupled with voluntary exercise augments exercise endurance. Mol Metab. 2018;7:1–11. https://pubmed.ncbi.nlm.nih.gov/29146412/
Frederick DW, Davis JG, Dávila A Jr, et al. Increasing NAD synthesis in muscle via nicotinamide phosphoribosyltransferase is not sufficient to promote oxidative metabolism. J Biol Chem. 2015;290(3):1546–58. https://pubmed.ncbi.nlm.nih.gov/25411251/
Dollerup OL, Chubanava S, Agerholm M, et al. Nicotinamide riboside does not alter mitochondrial respiration, content or morphology in skeletal muscle from obese and insulin-resistant men. J Physiol. 2020;598(4):731–54. https://pubmed.ncbi.nlm.nih.gov/31710095/
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol. 2022;198:114946. https://pubmed.ncbi.nlm.nih.gov/35134387/
Costford SR, Bajpeyi S, Pasarica M, et al. Skeletal muscle NAMPT is induced by exercise in humans. Am J Physiol Endocrinol Metab. 2010;298(1):E117–26. https://pubmed.ncbi.nlm.nih.gov/19887595/
Lamb DA, Moore JH, Mesquita PHC, et al. Resistance training increases muscle NAD+ and NADH concentrations as well as NAMPT protein levels and global sirtuin activity in middle-aged, overweight, untrained individuals. Aging (Albany NY). 2020;12(10):9447–60. https://pubmed.ncbi.nlm.nih.gov/32369778/
Ruan Q, Ruan J, Zhang W, Qian F, Yu Z. Targeting NAD+ degradation: the therapeutic potential of flavonoids for Alzheimer’s disease and cognitive frailty. Pharmacol Res. 2018;128:345–58. https://pubmed.ncbi.nlm.nih.gov/28847709/
Soma M, Lalam SK. The role of nicotinamide mononucleotide (NMN) in anti-aging, longevity, and its potential for treating chronic conditions. Mol Biol Rep. 2022;49(10):9737–48. https://pubmed.ncbi.nlm.nih.gov/35441939/
Skidmore CJ, Davies MI, Goodwin PM, et al. The involvement of poly(ADP-ribose) polymerase in the degradation of NAD caused by ¿-radiation and N-methyl-N-nitrosourea. Eur J Biochem. 1979;101(1):135–42. https://pubmed.ncbi.nlm.nih.gov/228934/
Pacher P, Szabó C. Role of poly(ADP-ribose) polymerase 1 (PARP-1) in cardiovascular diseases: the therapeutic potential of PARP inhibitors. Cardiovasc Drug Rev. 2007;25(3):235–60. https://pubmed.ncbi.nlm.nih.gov/17919258/
Palmer RD, Vaccarezza M. Nicotinamide adenine dinucleotide and the sirtuins caution: pro-cancer functions. Aging Med (Milton). 2021;4(4):337–44. https://pubmed.ncbi.nlm.nih.gov/34964015/
Amici SA, Young NA, Narvaez-Miranda J, et al. CD38 is robustly induced in human macrophages and monocytes in inflammatory conditions. Front Immunol. 2018;9:1593. https://pubmed.ncbi.nlm.nih.gov/30042766/
Polzonetti V, Carpi FM, Micozzi D, Pucciarelli S, Vincenzetti S, Napolioni V. Population variability in CD38 activity: correlation with age and significant effect of TNF-a-308GA and CD38 184CG SNPs. Mol Genet Metab. 2012;105(3):502–7. https://pubmed.ncbi.nlm.nih.gov/22236458/
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol. 2022;198:114946. https://pubmed.ncbi.nlm.nih.gov/35134387/
Wu S, Zhang R. CD38-expressing macrophages drive age-related NAD+ decline. Nat Metab. 2020;2(11):1186–7. https://pubmed.ncbi.nlm.nih.gov/33199923/
Chini CCS, Peclat TR, Warner GM, et al. CD38 ecto-enzyme in immune cells is induced during aging and regulates NAD+ and NMN levels. Nat Metab. 2020;2(11):1284–304. https://pubmed.ncbi.nlm.nih.gov/33199925/
Larrick JW, Mendelsohn AR. Finally, a regimen to extend human life expectancy. Rejuvenation Res. 2018;21(3):278–82. https://pubmed.ncbi.nlm.nih.gov/29781380/
Li Y, Pan A, Wang DD, et al. Impact of healthy lifestyle factors on life expectancies in the US population. Circulation. 2018;138(4):345–55. https://pubmed.ncbi.nlm.nih.gov/29712712/
Ruby JG, Wright KM, Rand KA, et al. Estimates of the heritability of human longevity are substantially inflated due to assortative mating. Genetics. 2018;210(3):1109–24. https://pubmed.ncbi.nlm.nih.gov/30401766/
Kaeberlein M, Creevy KE, Promislow DEL. The dog aging project: translational geroscience in companion animals. Mamm Genome. 2016;27(7–8):279–88. https://pubmed.ncbi.nlm.nih.gov/27143112/
Scott CT, DeFrancesco L. Selling long life. Nat Biotechnol. 2015;33(1):31–40. https://pubmed.ncbi.nlm.nih.gov/25574633/
Chase P, Mitchell K, Morley JE. In the steps of giants: the early geriatrics texts. J Am Geriatr Soc. 2000;48(1):89–94. https://pubmed.ncbi.nlm.nih.gov/10642028/
King DE, Mainous AG III, Geesey ME. Turning back the clock: adopting a healthy lifestyle in middle age. Am J Med. 2007;120(7):598–603. https://pubmed.ncbi.nlm.nih.gov/17602933/
Ma H, Xue Q, Wang X, et al. Adding salt to foods and hazard of premature mortality. Eur Heart J. 2022;43(30):2878–88. https://pubmed.ncbi.nlm.nih.gov/35808995/
Cevenini E, Monti D, Franceschi C. Inflamm-ageing. Curr Opin Clin Nutr Metab Care. 2013;16(1):14–20. https://pubmed.ncbi.nlm.nih.gov/23132168/
Ioannidis JPA. The challenge of reforming nutritional epidemiologic research. JAMA. 2018;320(10):969–70. https://pubmed.ncbi.nlm.nih.gov/30422271/
The US Burden of Disease Collaborators, Mokdad AH, Ballestros K, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444. https://pubmed.ncbi.nlm.nih.gov/29634829/
King ML Jr. Stride Toward Freedom: The Montgomery Story. Harper; 1958. https://worldcat.org/title/610192372
Mathers JC. Obesity and mortality: is childhood obesity shortening life expectancy? Maturitas. 2015;81(1):1–2. https://pubmed.ncbi.nlm.nih.gov/25708225/
Ludwig DS. Lifespan weighed down by diet. JAMA. 2016;315(21):2269–70. https://pubmed.ncbi.nlm.nih.gov/27043490/
Mathers JC. Obesity and mortality: is childhood obesity shortening life expectancy? Maturitas. 2015;81(1):1–2. https://pubmed.ncbi.nlm.nih.gov/25708225/
Xu J, Murphy SL, Kochanek KD, Arias E. Mortality in the United States, 2015. NCHS Data Brief. 2016;(267):1–8. https://pubmed.ncbi.nlm.nih.gov/27930283/
Olshansky SJ, Passaro DJ, Hershow RC, et al. A potential decline in life expectancy in the United States in the 21st century. N Engl J Med. 2005;352(11):1138–45. https://pubmed.ncbi.nlm.nih.gov/15784668/
Masters RK, Aron LY, Woolf SH. Changes in Life Expectancy between 2019 and 2021 in the United States and 21 Peer Countries. Public and Global Health; 2022. https://pubmed.ncbi.nlm.nih.gov/35416991/
Walker BR, Colledge NR. Davidson’s Principles and Practices of Medicine E-Book. Elsevier Health Sciences; 2013:169 https://worldcat.org/title/1334359473
Shay CM, Ning H, Allen NB, et al. Status of cardiovascular health in US adults: prevalence estimates from the National Health and Nutrition Examination Surveys (NHANES) 2003–2008. Circulation. 2012;125(1):45–56. https://pubmed.ncbi.nlm.nih.gov/22095826/
Roger VL, Go AS, Lloyd-Jones DM, et al. Heart disease and stroke statistics—2012 update: a report from the American Heart Association. Circulation. 2012;125(1):e2–220. https://pubmed.ncbi.nlm.nih.gov/22179539/
Murray CJ, Atkinson C, Bhalla K, et al. The state of US health, 1990–2010: burden of diseases, injuries, and risk factors. JAMA. 2013;310(6):591–608. https://pubmed.ncbi.nlm.nih.gov/23842577/
Villines TC. Risks of the American lifestyle: insights from a trans-Pacific comparison of coronary artery calcium progression. Circ: Cardiovasc Imaging. 2019;12(2):e008810. https://www.ahajournals.org/doi/10.1161/CIRCIMAGING.119.008810
Mokdad AH, Ballestros K, Echko J, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Mayo Clinic News Network. Nearly 7 in 10 Americans take prescription drugs, Mayo Clinic, Olmsted Medical Center find. MayoClinic.org. http://newsnetwork.mayoclinic.org/discussion/nearly-7-in-10-americans-take-prescription-drugs-mayo-clinic-olmsted-medical-center-find/. Published June 19, 2013. Accessed January 29, 2023.; https://newsnetwork.mayoclinic.org/discussion/nearly-7-in-10-americans-take-prescription-drugs-mayo-clinic-olmsted-medical-center-find/
Mokdad AH, Ballestros K, Echko J, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72. https://pubmed.ncbi.nlm.nih.gov/29634829/
Wansink B, Kniffin KM, Shimizu M. Death row nutrition. Curious conclusions of last meals. Appetite. 2012;59(3):837–43. https://pubmed.ncbi.nlm.nih.gov/22925848/
Ezzati M, Riboli E. Can noncommunicable diseases be prevented? Lessons from studies of populations and individuals. Science. 2012;337(6101):1482–7. https://pubmed.ncbi.nlm.nih.gov/22997325/
Katz DL, Frates EP, Bonnet JP, Gupta SK, Vartiainen E, Carmona RH. Lifestyle as medicine: the case for a True Health Initiative. Am J Health Promot. 2018;32(6):1452–8. https://pubmed.ncbi.nlm.nih.gov/28523941/
Goldberg JP, Hellwig JP. Nutrition research in the media: the challenge facing scientists. J Am Coll Nutr. 1997;16(6):544–50. https://pubmed.ncbi.nlm.nih.gov/9430082/
Keys A. Nutrition and capacity for work. Occup Med. 1946;2(6):536–45. https://pubmed.ncbi.nlm.nih.gov/20288136/
Bodai BI, Nakata TE, Wong WT, et al. Lifestyle medicine: a brief review of its dramatic impact on health and survival. Perm J. 2018;22:17–025. https://pubmed.ncbi.nlm.nih.gov/29035175/
Katz DL, Frates EP, Bonnet JP, Gupta SK, Vartiainen E, Carmona RH. Lifestyle as medicine: the case for a True Health Initiative. Am J Health Promot. 2018;32(6):1452–8. https://pubmed.ncbi.nlm.nih.gov/28523941/
THI_About Us. True Health Initiative. https://www.truehealthinitiative.org/about_us/. Accessed January 29, 2023.; https://www.truehealthinitiative.org/about_us/
Pledge of support for core principles. True Health Initiative. https://www.truehealthinitiative.org/wp-content/uploads/2021/02/THI_Pledge_2021–02–23.pdf. Published February 23, 2021. Accessed January 29, 2023.; https://www.truehealthinitiative.org/
Milton K. Hunter-gatherer diets – a different perspective. Am J Clin Nutr. 2000;71(3):665–7. https://pubmed.ncbi.nlm.nih.gov/10702155/
Tutin CEG, Fernandez M. Composition of the diet of chimpanzees and comparisons with that of sympatric lowland gorillas in the Lopé Reserve, Gabon. Am J Primatol. 1993;30(3):195–211. https://pubmed.ncbi.nlm.nih.gov/31937009/
Beitz DC, Bauer JE, Behnke KC, et al. Nutrient Requirements of Dogs and Cats. National Academies Press; 2006. https://worldcat.org/title/62741464
Roberts WC. We think we are one, we act as if we are one, but we are not one. Am J Cardiol. 1990;66(10):896. https://pubmed.ncbi.nlm.nih.gov/2220599/
Nestle M. Paleolithic diets: a sceptical view. Nutr Bull. 2000;25(1):43–7. https://nyuscholars.nyu.edu/en/publications/paleolithic-diets-a-sceptical-view
Walker AR. Are health and ill-health lessons from hunter-gatherers currently relevant? Am J Clin Nutr. 2001;73(2):353–6. https://pubmed.ncbi.nlm.nih.gov/11157335/
Kahleova H, Levin S, Barnard ND. Vegetarian dietary patterns and cardiovascular disease. Prog Cardiovasc Dis. 2018;61(1):54–61. https://pubmed.ncbi.nlm.nih.gov/29800598/