1932

Abstract

The trajectory of human aging varies widely from one individual to the next due to complex interactions between the genome and the environment that influence the aging process. Such differences in age-specific mortality and disease risk among same-aged individuals reflect variation in the pace of biological aging. Certain mechanisms involved in the progression of biological aging originate in the epigenome, where chemical modifications to the genome are able to alter gene expression without modifying the underlying DNA sequence. The epigenome serves as an interface for environmental signals, which are able to “get under the skin” to influence health and aging. A number of the molecular mechanisms involved in the aging process have been identified, although few aging phenotypes have been definitively traced to their underlying molecular causes thus far. In this review, we discuss variation in human biological aging and the epigenome's role in promoting heterogeneity in human longevity and healthspan.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-anthro-052721-090516
2023-10-23
2024-05-06
Loading full text...

Full text loading...

/deliver/fulltext/anthro/52/1/annurev-anthro-052721-090516.html?itemId=/content/journals/10.1146/annurev-anthro-052721-090516&mimeType=html&fmt=ahah

Literature Cited

  1. Anderson JA, Johnston RA, Lea AJ, Campos FA, Voyles TN et al. 2021. High social status males experience accelerated epigenetic aging in wild baboons. eLife 10:e66128
    [Google Scholar]
  2. Arneson A, Haghani A, Thompson MJ, Pellegrini M, Kwon SB et al. 2022. A mammalian methylation array for profiling methylation levels at conserved sequences. Nat. Commun 13:1783
    [Google Scholar]
  3. Austad SN, Hoffman JM. 2018. Is antagonistic pleiotropy ubiquitous in aging biology?. Evol. Med. Public Health 2018:1287–94
    [Google Scholar]
  4. Baker GT 3rd, Sprott RL 1988. Biomarkers of aging. Exp. Gerontol 23:4–5223–39
    [Google Scholar]
  5. Barzilai N, Huffman DM, Muzumdar RH, Bartke A. 2012. The critical role of metabolic pathways in aging. Diabetes 61:61315–22
    [Google Scholar]
  6. Bell CG, Lowe R, Adams PD, Baccarelli AA, Beck S et al. 2019. DNA methylation aging clocks: challenges and recommendations. Genome Biol 20:1249
    [Google Scholar]
  7. Benayoun BA, Pollina EA, Brunet A. 2015. Epigenetic regulation of ageing: linking environmental inputs to genomic stability. Nat. Rev. Mol. Cell Biol. 16:10593–610
    [Google Scholar]
  8. Bhatia-Dey N, Kanherkar RR, Stair SE, Makarev EO, Csoka AB. 2016. Cellular senescence as the causal nexus of aging. Front. Genet. 7:13
    [Google Scholar]
  9. Bocklandt S, Lin W, Sehl ME, Sánchez FJ, Sinsheimer JS et al. 2011. Epigenetic predictor of age. PLOS ONE 6:6e14821
    [Google Scholar]
  10. Booth LN, Brunet A. 2016. The aging epigenome. Mol. Cell 62:5728–44
    [Google Scholar]
  11. Bribiescas RG. 2020. Aging, life history, and human evolution. Annu. Rev. Anthropol 49:101–21
    [Google Scholar]
  12. Briller S, Carrillo E. 2020. Applying anthropological insight in an aging world. Oxford Research Encyclopedia of Anthropology Oxford, UK: Oxford Univ. Press https://doi.org/10.1093/acrefore/9780190854584.013.2
    [Crossref] [Google Scholar]
  13. Brunet A, Berger SL. 2014. Epigenetics of aging and aging-related disease. J. Gerontol. A Biol. Sci. Med. Sci 69:Suppl. 1S17–20
    [Google Scholar]
  14. Brunet A, Rando TA. 2017. Interaction between epigenetic and metabolism in aging stem cells. Curr. Opin. Cell Biol. 45:1–7
    [Google Scholar]
  15. Campbell RR, Wood MA. 2019. How the epigenome integrates information and reshapes the synapse. Nat. Rev. Neurosci 20:3133–47
    [Google Scholar]
  16. Caspari R, Lee S-H. 2006. Is human longevity a consequence of cultural change or modern biology?. Am. J. Phys. Anthropol. 129:4512–17
    [Google Scholar]
  17. Chen BH, Marioni RE, Colicino E, Peters MJ, Ward-Caviness CK et al. 2016. DNA methylation-based measures of biological age: meta-analysis predicting time to death. Aging 8:91844–65
    [Google Scholar]
  18. Cheong A, Nagel ZD. 2022. Human variation in DNA repair, immune function, and cancer risk. Front. Immunol 13:899574
    [Google Scholar]
  19. Chera S, Buzgariu W, Ghila L, Galliot B. 2009. Autophagy in Hydra: a response to starvation and stress in early animal evolution. Biochim. Biophys. Acta 1793:91432–43
    [Google Scholar]
  20. Chiou KL, Montague MJ, Goldman EA, Watowich MM, Sams SN et al. 2020. Rhesus macaques as a tractable physiological model of human ageing. Philos. Trans. R. Soc. B 375:181120190612
    [Google Scholar]
  21. Choi H, Joe S, Nam H. 2019. Development of tissue-specific age predictors using DNA methylation data. Genes 10:11888
    [Google Scholar]
  22. Christensen BC, Houseman EA, Marsit CJ, Zheng S, Wrensch MR et al. 2009. Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context. PLOS Genet 5:8e1000602
    [Google Scholar]
  23. Christensen K, Doblhammer G, Rau R, Vaupel JW. 2009. Ageing populations: the challenges ahead. Lancet 374:96961196–208
    [Google Scholar]
  24. Cohen AA, Ferrucci L, Fülöp T, Gravel D, Hao N et al. 2022. A complex systems approach to aging biology. Nat. Aging 2:7580–91
    [Google Scholar]
  25. Cole JJ, Robertson NA, Rather MI, Thomson JP, McBryan T et al. 2017. Diverse interventions that extend mouse lifespan suppress shared age-associated epigenetic changes at critical gene regulatory regions. Genome Biol 18:158
    [Google Scholar]
  26. Crews DE. 1993. Biological anthropology and human aging: some current directions in aging research. Annu. Rev. Anthropol 22:395–423
    [Google Scholar]
  27. Crews DE. 2022. Aging, frailty, and design of built environments. J. Physiol. Anthropol 41:12
    [Google Scholar]
  28. Crimmins EM. 2015. Lifespan and healthspan: past, present, and promise. Gerontologist 55:6901–11
    [Google Scholar]
  29. De Paoli-Iseppi R, Deagle BE, McMahon CR, Hindell MA, Dickinson JL, Jarman SN. 2017. Measuring animal age with DNA methylation: from humans to wild animals. Front. Genet. 8:106
    [Google Scholar]
  30. Didier ES, MacLean AG, Mohan M, Didier PJ, Lackner AA, Kuroda MJ 2016. Contributions of nonhuman primates to research on aging. Vet. Pathol 53:2277–90
    [Google Scholar]
  31. Dirks RAM, Stunnenberg HG, Marks H. 2016. Genome-wide epigenomic profiling for biomarker discovery. Clin. Epigenet 8:122
    [Google Scholar]
  32. Elliott ML, Caspi A, Houts RM, Ambler A, Broadbent JM et al. 2021. Disparities in the pace of biological aging among midlife adults of the same chronological age have implications for future frailty risk and policy. Nat. Aging 1:3295–308
    [Google Scholar]
  33. Engelfriet PM, Jansen EHJM, Picavet HSJ, Dolle MET. 2013. Biochemical markers of aging for longitudinal studies in humans. Epidemiol. Rev. 35:1132–51
    [Google Scholar]
  34. Field AE, Robertson NA, Wang T, Havas A, Ideker T, Adams PD. 2018. DNA methylation clocks in aging: categories, causes, and consequences. Mol. Cell 71:6882–95
    [Google Scholar]
  35. Flatt T, Partridge L. 2018. Horizons in the evolution of aging. BMC Biol 16:193
    [Google Scholar]
  36. Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML et al. 2005. Epigenetic differences arise during the lifetime of monozygotic twins. PNAS 102:3010604–9
    [Google Scholar]
  37. Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A. 2018. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol 14:10576–90
    [Google Scholar]
  38. Fulop T, Larbi A, Pawelec G, Khalil A, Cohen AA et al. 2023. Immunology of aging: the birth of inflammaging. Clin. Rev. Allergy Immunol. 64:2109–22
    [Google Scholar]
  39. Galkin F, Mamoshina P, Aliper A, de Magalhães JP, Gladyshev VN, Zhavoronkov A. 2020. Biohorology and biomarkers of aging: current state-of-the-art, challenges and opportunities. Ageing Res. Rev. 60:101050
    [Google Scholar]
  40. Gassen NC, Chrousos GP, Binder EB, Zannas AS. 2017. Life stress, glucocorticoid signaling, and the aging epigenome: implications for aging-related diseases. Neurosci. Biobehav. Rev 74:356–65
    [Google Scholar]
  41. Gluckman PD, Hanson MA, Low FM. 2019. Evolutionary and developmental mismatches are consequences of adaptive developmental plasticity in humans and have implications for later disease risk. Philos. Trans. R. Soc. B 374:177020180109
    [Google Scholar]
  42. Goldman EA, Chiou KL, Watowich MM, Mercer A, Sams SN et al. 2022. A generalizable epigenetic clock captures aging in two nonhuman primates. bioRxiv. https://doi.org/10.1101/2022.11.01.514719
  43. Greer EL, Dowlatshahi D, Banko MR, Villen J, Hoang K et al. 2007. An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Curr. Biol. 17:191646–56
    [Google Scholar]
  44. Hahn O, Grönke S, Stubbs TM, Ficz G, Hendrich O et al. 2017. Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism. Genome Biol. 18:56
    [Google Scholar]
  45. Hahn O, Stubbs TM, Reik W, Grönke S, Beyer A, Partridge L. 2018. Hepatic gene body hypermethylation is a shared epigenetic signature of murine longevity. PLOS Genet 14:11e1007766
    [Google Scholar]
  46. Haldane J. 1941. New Paths in Genetics London: George Allen & Unwin
  47. Hannum G, Guinney J, Zhao L, Zhang L, Hughes G et al. 2013. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol. Cell 49:2359–67
    [Google Scholar]
  48. Horvath S. 2013. DNA methylation age of human tissues and cell types. Genome Biol 14:10R115
    [Google Scholar]
  49. Horvath S, Erhart W, Brosch M, Ammerpohl O, von Schonfels W et al. 2014. Obesity accelerates epigenetic aging of human liver. PNAS 111:4315538–43
    [Google Scholar]
  50. Horvath S, Raj K. 2018. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 19:6371–84
    [Google Scholar]
  51. Horvath S, Zoller JA, Haghani A, Jasinska AJ, Raj K et al. 2021. Epigenetic clock and methylation studies in the rhesus macaque. GeroScience 43:2441–53
    [Google Scholar]
  52. Issa J-P. 2014. Aging and epigenetic drift: a vicious cycle. J. Clin. Investig. 124:124–29
    [Google Scholar]
  53. Jasinska AJ, Haghani A, Zoller JA, Li CZ, Arneson A et al. 2022. Epigenetic clock and methylation studies in vervet monkeys. GeroScience 44:2699–717
    [Google Scholar]
  54. Jones JH. 2011. Primates and the evolution of long, slow life histories. Curr. Biol. 21:18R708–17
    [Google Scholar]
  55. Jylhävä J, Pedersen NL, Hägg S. 2017. Biological age predictors. EBioMedicine 21:29–36
    [Google Scholar]
  56. Kananen L, Marttila S, Nevalainen T, Kummola L, Junttila I et al. 2016. The trajectory of the blood DNA methylome ageing rate is largely set before adulthood: evidence from two longitudinal studies. Age 38:365
    [Google Scholar]
  57. Kane AE, Sinclair DA. 2019. Epigenetic changes during aging and their reprogramming potential. Crit. Rev. Biochem. Mol. Biol. 54:161–83
    [Google Scholar]
  58. Kaplan H, Hill K, Lancaster J, Hurtado AM. 2000. A theory of human life history evolution: diet, intelligence, and longevity. Evol. Anthropol 9:4156–85
    [Google Scholar]
  59. Klass MR. 1983. A method for the isolation of longevity mutants in the nematode Caenorhabditiselegans and initial results. Mech. Ageing Dev. 22:3–4279–86
    [Google Scholar]
  60. Klemm SL, Shipony Z, Greenleaf WJ. 2019. Chromatin accessibility and the regulatory epigenome. Nat. Rev. Genet. 20:4207–20
    [Google Scholar]
  61. Klimovich A, Rehm A, Wittlieb J, Herbst E-M, Benavente R, Bosch TCG. 2018. Non-senescent Hydra tolerates severe disturbances in the nuclear lamina. Aging 10:5951–72
    [Google Scholar]
  62. Kuzawa CW, Thayer ZM. 2011. Timescales of human adaptation: the role of epigenetic processes. Epigenomics 3:2221–34
    [Google Scholar]
  63. Lee KWK, Pausova Z. 2013. Cigarette smoking and DNA methylation. Front. Genet. 4:132
    [Google Scholar]
  64. Lemoine M. 2021. The evolution of the hallmarks of aging. Front. Genet. 12:693071
    [Google Scholar]
  65. Levine ME, Lu AT, Bennett DA, Horvath S. 2015. Epigenetic age of the pre-frontal cortex is associated with neuritic plaques, amyloid load, and Alzheimer's disease related cognitive functioning. Aging 7:121198–211
    [Google Scholar]
  66. Levine ME, Lu AT, Quach A, Chen BH, Assimes TL et al. 2018. An epigenetic biomarker of aging for lifespan and healthspan. Aging 10:4573–91
    [Google Scholar]
  67. Levine ME, McDevitt RA, Meer M, Perdue K, Di Francesco A et al. 2020. A rat epigenetic clock recapitulates phenotypic aging and co-localizes with heterochromatin. eLife 9:e59201
    [Google Scholar]
  68. Li A, Koch Z, Ideker T. 2022. Epigenetic aging: biological age prediction and informing a mechanistic theory of aging. J. Intern. Med. 292:5733–44
    [Google Scholar]
  69. Lombard DB, Chua KF, Mostoslavsky R, Franco S, Gostissa M, Alt FW. 2005. DNA repair, genome stability, and aging. Cell 120:4497–512
    [Google Scholar]
  70. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. 2013. The hallmarks of aging. Cell 153:61194–217
    [Google Scholar]
  71. López-Otín C, Galluzzi L, Freije JMP, Madeo F, Kroemer G. 2016. Metabolic control of longevity. Cell 166:4802–21
    [Google Scholar]
  72. Lowe R, Barton C, Jenkins CA, Ernst C, Forman O et al. 2018. Ageing-associated DNA methylation dynamics are a molecular readout of lifespan variation among mammalian species. Genome Biol 19:22
    [Google Scholar]
  73. Lu AT, Quach A, Wilson JG, Reiner AP, Aviv A et al. 2019. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 11:2303–27
    [Google Scholar]
  74. Maegawa S, Lu Y, Tahara T, Lee JT, Madzo J et al. 2017. Caloric restriction delays age-related methylation drift. Nat. Commun. 8:539
    [Google Scholar]
  75. Mamoshina P, Kochetov K, Putin E, Cortese F, Aliper A et al. 2018. Population specific biomarkers of human aging: a big data study using South Korean, Canadian, and Eastern European patient populations. J. Gerontol. A Biol. Sci. Med. Sci 73:111482–90
    [Google Scholar]
  76. Martin CL, Ghastine L, Lodge EK, Dhingra R, Ward-Caviness CK. 2022. Understanding health inequalities through the lens of social epigenetics. Annu. Rev. Public Health 43:235–54
    [Google Scholar]
  77. McCrory C, Fiorito G, Hernandez B, Polidoro S, O'Halloran AM et al. 2021. GrimAge outperforms other epigenetic clocks in the prediction of age-related clinical phenotypes and all-cause mortality. J. Gerontol. A 76:5741–49
    [Google Scholar]
  78. McDade TW, Ryan CP, Jones MJ, Hoke MK, Borja J et al. 2019. Genome-wide analysis of DNA methylation in relation to socioeconomic status during development and early adulthood. Am. J. Phys. Anthropol 169:3–11
    [Google Scholar]
  79. Medawar PB. 1957. 1952. An unsolved problem of biology. Uniqueness of the Individual44–70. London: Methuen
    [Google Scholar]
  80. Mendenhall AR, Martin GM, Kaeberlein M, Anderson RM. 2021. Cell-to-cell variation in gene expression and the aging process. GeroScience 43:1181–96
    [Google Scholar]
  81. Mortimer RK, Johnston JR. 1959. Life span of individual yeast cells. Nature 183:46771751–52
    [Google Scholar]
  82. Moskalev A, Aliper A, Smit-McBride Z, Buzdin A, Zhavoronkov A. 2014. Genetics and epigenetics of aging and longevity. Cell Cycle 13:71063–77
    [Google Scholar]
  83. Mulligan CJ. 2016. Early environments, stress, and the epigenetics of human health. Annu. Rev. Anthropol 45:233–49
    [Google Scholar]
  84. Nwanaji-Enwerem JC, Weisskopf MG, Baccarelli AA. 2018. Multi-tissue DNA methylation age: molecular relationships and perspectives for advancing biomarker utility. Ageing Res. Rev. 45:15–23
    [Google Scholar]
  85. Pan Y. 2011. Mitochondria, reactive oxygen species, and chronological aging: a message from yeast. Exp. Gerontol. 46:11847–52
    [Google Scholar]
  86. Perna L, Zhang Y, Mons U, Holleczek B, Saum K-U, Brenner H. 2016. Epigenetic age acceleration predicts cancer, cardiovascular, and all-cause mortality in a German case cohort. Clin. Epigenet. 8:64
    [Google Scholar]
  87. Petkovich DA, Podolskiy DI, Lobanov AV, Lee S-G, Miller RA, Gladyshev VN. 2017. Using DNA methylation profiling to evaluate biological age and longevity interventions. Cell Metab 25:4954–960.e6
    [Google Scholar]
  88. Pickering AM, Lehr M, Gendron CM, Pletcher SD, Miller RA. 2017. Mitochondrial thioredoxin reductase 2 is elevated in long-lived primate as well as rodent species and extends fly mean lifespan. Aging Cell 16:4683–92
    [Google Scholar]
  89. Porter HL, Brown CA, Roopnarinesingh X, Giles CB, Georgescu C et al. 2021. Many chronological aging clocks can be found throughout the epigenome: implications for quantifying biological aging. Aging Cell 20:11e13492
    [Google Scholar]
  90. Prado NA, Brown JL, Zoller JA, Haghani A, Yao M et al. 2021. Epigenetic clock and methylation studies in elephants. Aging Cell 20:7e13414
    [Google Scholar]
  91. Promislow D, Anderson RM, Scheffer M, Crespi B, DeGregori J et al. 2022. Resilience integrates concepts in aging research. iScience 25:5104199
    [Google Scholar]
  92. Quach A, Levine ME, Tanaka T, Lu AT, Chen BH et al. 2017. Epigenetic clock analysis of diet, exercise, education, and lifestyle factors. Aging 9:2419–46
    [Google Scholar]
  93. Rakyan VK, Down TA, Maslau S, Andrew T, Yang T-P et al. 2010. Human aging-associated DNA hypermethylation occurs preferentially at bivalent chromatin domains. Genome Res 20:4434–39
    [Google Scholar]
  94. Ryan CP. 2021.. “ Epigenetic clocks”: theory and applications in human biology. Am. J. Hum. Biol. 33:3e23488
    [Google Scholar]
  95. Salminen A. 2021. Increased immunosuppression impairs tissue homeostasis with aging and age-related diseases. J. Mol. Med. 99:11–20
    [Google Scholar]
  96. Salminen A, Ojala J, Kaarniranta K, Kauppinen A. 2012. Mitochondrial dysfunction and oxidative stress activate inflammasomes: impact on the aging process and age-related diseases. Cell Mol. Life Sci. 69:182999–3013
    [Google Scholar]
  97. Seale K, Horvath S, Teschendorff A, Eynon N, Voisin S. 2022. Making sense of the ageing methylome. Nat. Rev. Genet. 23:10585–605
    [Google Scholar]
  98. Sebastiani P, Solovieff N, DeWan AT, Walsh KM, Puca A et al. 2012. Genetic signatures of exceptional longevity in humans. PLOS ONE 7:1e29848
    [Google Scholar]
  99. Sen P, Shah PP, Nativio R, Berger SL. 2016. Epigenetic mechanisms of longevity and aging. Cell 166:4822–39
    [Google Scholar]
  100. Teschendorff AE. 2020. A comparison of epigenetic mitotic-like clocks for cancer risk prediction. Genome Med 12:156
    [Google Scholar]
  101. Thompson RF, Atzmon G, Gheorghe C, Liang HQ, Lowes C et al. 2010. Tissue-specific dysregulation of DNA methylation in aging. Aging Cell 9:4506–18
    [Google Scholar]
  102. Trevathan WR. 2007. Evolutionary medicine. Annu. Rev. Anthropol. 36:139–54
    [Google Scholar]
  103. Turan ZG, Parvizi P, Dönertaş HM, Tung J, Khaitovich P, Somel M. 2019. Molecular footprint of Medawar's mutation accumulation process in mammalian aging. Aging Cell 18:4e12965
    [Google Scholar]
  104. Vaiserman A, Krasnienkov D. 2021. Telomere length as a marker of biological age: state-of-the-art, open issues, and future perspectives. Front. Genet. 11:630186
    [Google Scholar]
  105. van Leeuwen KM, van Loon MS, van Nes FA, Bosmans JE, de Vet HCW et al. 2019. What does quality of life mean to older adults? A thematic synthesis. PLOS ONE 14:3e0213263
    [Google Scholar]
  106. Vaupel JW, Villavicencio F, Bergeron-Boucher M-P. 2021. Demographic perspectives on the rise of longevity. PNAS 118:9e2019536118
    [Google Scholar]
  107. von Zglinicki T, Martin-Ruiz C. 2005. Telomeres as biomarkers for ageing and age-related diseases. Curr. Mol. Med. 5:2197–203
    [Google Scholar]
  108. Wagner W. 2017. Epigenetic aging clocks in mice and men. Genome Biol 18:1107
    [Google Scholar]
  109. Wahl S, Drong A, Lehne B, Loh M, Scott WR et al. 2017. Epigenome-wide association study of body mass index, and the adverse outcomes of adiposity. Nature 541:763581–86
    [Google Scholar]
  110. Wang K, Liu H, Hu Q, Wang L, Liu J et al. 2022. Epigenetic regulation of aging: implications for interventions of aging and diseases. Signal Transduct. Target. Ther 7:1374
    [Google Scholar]
  111. Wang T, Tsui B, Kreisberg JF, Robertson NA, Gross AM et al. 2017. Epigenetic aging signatures in mice livers are slowed by dwarfism, calorie restriction and rapamycin treatment. Genome Biol 18:157
    [Google Scholar]
  112. Warner DA, Miller DAW, Bronikowski AM, Janzen FJ. 2016. Decades of field data reveal that turtles senesce in the wild. PNAS 113:236502–7
    [Google Scholar]
  113. Weissman A. 1892. The Germ Plasm: A Theory of Heredity Jena, Ger: G. Fischer
  114. West J, Widschwendter M, Teschendorff AE. 2013. Distinctive topology of age-associated epigenetic drift in the human interactome. PNAS 110:3514138–43
    [Google Scholar]
  115. White AJ, Kresovich JK, Keller JP, Xu Z, Kaufman JD et al. 2019. Air pollution, particulate matter composition and methylation-based biologic age. Environ. Int. 132:105071
    [Google Scholar]
  116. WHO (World Health Organ.) 2021. WHO Guideline on Self-Care Interventions for Health and Well-Being Geneva: WHO https://www.who.int/publications/i/item/9789240052192
  117. Williams GC. 1957. Pleiotropy, natural selection, and the evolution of senescence. Evolution 11:4398–411
    [Google Scholar]
  118. Xia X, Chen W, McDermott J, Han J-DJ. 2017. Molecular and phenotypic biomarkers of aging. F1000Res 6:860
    [Google Scholar]
/content/journals/10.1146/annurev-anthro-052721-090516
Loading
/content/journals/10.1146/annurev-anthro-052721-090516
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error