Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Making sense of the ageing methylome

Abstract

Over time, the human DNA methylation landscape accrues substantial damage, which has been associated with a broad range of age-related diseases, including cardiovascular disease and cancer. Various age-related DNA methylation changes have been described, including at the level of individual CpGs, such as differential and variable methylation, and at the level of the whole methylome, including entropy and correlation networks. Here, we review these changes in the ageing methylome as well as the statistical tools that can be used to quantify them. We detail the evidence linking DNA methylation to ageing phenotypes and the longevity strategies aimed at altering both DNA methylation patterns and machinery to extend healthspan and lifespan. Lastly, we discuss theories on the mechanistic causes of epigenetic ageing.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Linear models classify age-associated changes in DNAm.
Fig. 2: Entropy measures chaos in the ageing methylome.
Fig. 3: Correlation networks reveal connectivity in the ageing methylome.
Fig. 4: DMPs and VMPs reflect primary and secondary ageing processes.
Fig. 5: Proposed mechanisms of epigenetic ageing.

Similar content being viewed by others

References

  1. Seals, D. R., Justice, J. N. & Larocca, T. J. Physiological geroscience: targeting function to increase healthspan and achieve optimal longevity. J. Physiol. 594, 2001–2024 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Petsko, G. A. A seat at the table. Genome Biol. 9, 113 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Partridge, L., Deelen, J. & Slagboom, P. E. Facing up to the global challenges of ageing. Nature 561, 45–56 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Crimmins, E. M. Lifespan and healthspan: past, present, and promise. Gerontologist 55, 901–911 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Harper, S. Economic and social implications of aging societies. Science 346, 587–591 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Jones, M. J., Goodman, S. J. & Kobor, M. S. DNA methylation and healthy human aging. Aging Cell 14, 924–932 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. The hallmarks of aging. Cell 153, 1194–1217 (2013). This review proposes nine hallmarks of ageing and categorizes these hallmarks into three groups: primary hallmarks, antagonistic hallmarks and integrative hallmarks. Epigenetic alterations are described here as a primary hallmark of ageing.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Gladyshev, V. N. Aging: progressive decline in fitness due to the rising deleteriome adjusted by genetic, environmental, and stochastic processes. Aging Cell 15, 594–602 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cartee, G. D., Hepple, R. T., Bamman, M. M. & Zierath, J. R. Exercise promotes healthy aging of skeletal muscle. Cell Metab. 23, 1034–1047 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kolovou, G. D., Kolovou, V. & Mavrogeni, S. We are ageing. Biomed. Res. Int. 2014, 808307 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Holloszy, J. O. The biology of aging. Mayo Clin. Proc. 75, S3–S9 (2000).

    Article  PubMed  Google Scholar 

  12. Hägg, S. & Jylhävä, J. Sex differences in biological aging with a focus on human studies. eLife 10, e63425 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Zhang, W., Qu, J., Liu, G.-H. & Belmonte, J. C. I. The ageing epigenome and its rejuvenation. Nat. Rev. Mol. Cell Biol. 21, 137–150 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Kane, A. E. & Sinclair, D. A. Epigenetic changes during aging and their reprogramming potential. Crit. Rev. Biochem. Mol. Biol. 54, 61–83 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Greenberg, M. V. C. & Bourc’his, D. The diverse roles of DNA methylation in mammalian development and disease. Nat. Rev. Mol. Cell Biol. 20, 590–607 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Bauer, M. Cell-type-specific disturbance of DNA methylation pattern: a chance to get more benefit from and to minimize cohorts for epigenome-wide association studies. Int. J. Epidemiol. 47, 917–927 (2018).

    Article  PubMed  Google Scholar 

  17. Teschendorff, A. E. & Relton, C. L. Statistical and integrative system-level analysis of DNA methylation data. Nat. Rev. Genet. 19, 129–147 (2018). This review describes the statistical techniques, and challenges, associated with analysing DNAm data. Notably, this paper describes methods to handle cell-type heterogeneity in DNAm, various feature selection tools and how to approach integrated systems-level data analysis.

    Article  CAS  PubMed  Google Scholar 

  18. Feil, R. & Fraga, M. F. Epigenetics and the environment: emerging patterns and implications. Nat. Rev. Genet. 13, 97–109 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Turner, D. et al. DNA methylation across the genome in aged human skeletal muscle tissue and muscle stem cells: the role of HOX genes and physical activity. Sci. Rep. 10, 15360 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Barrès, R. et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell Metab. 15, 405–411 (2012).

    Article  PubMed  CAS  Google Scholar 

  21. Urdinguio, R. G. et al. Physical exercise shapes the mouse brain epigenome. Mol. Metab. 54, 101398 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Voisin, S. et al. Dietary fat quality impacts genome-wide DNA methylation patterns in a cross-sectional study of Greek preadolescents. Eur. J. Hum. Genet. 23, 654–662 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Pauwels, S. et al. Maternal intake of methyl-group donors affects DNA methylation of metabolic genes in infants. Clin. Epigenetics 9, 16 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Joubert, B. R. et al. DNA methylation in newborns and maternal smoking in pregnancy: genome-wide consortium meta-analysis. Am. J. Hum. Genet. 98, 680–696 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Elliott, H. R. et al. Differences in smoking associated DNA methylation patterns in South Asians and Europeans. Clin. Epigenetics 6, 4 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Tsaprouni, L. G. et al. Cigarette smoking reduces DNA methylation levels at multiple genomic loci but the effect is partially reversible upon cessation. Epigenetics 9, 1382–1396 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Christensen, B. C. et al. Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet. 5, e1000602 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Plusquin, M. et al. DNA methylation and exposure to ambient air pollution in two prospective cohorts. Environ. Int. 108, 127–136 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang, L. et al. Epigenome-wide meta-analysis of DNA methylation differences in prefrontal cortex implicates the immune processes in Alzheimer’s disease. Nat. Commun. 11, 6114 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Li, P. et al. Epigenetic dysregulation of enhancers in neurons is associated with Alzheimer’s disease pathology and cognitive symptoms. Nat. Commun. 10, 2246 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Altuna, M. et al. DNA methylation signature of human hippocampus in Alzheimer’s disease is linked to neurogenesis. Clin. Epigenetics 11, 91 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Pellegrini, C. et al. A meta-analysis of brain DNA methylation across sex, age, and Alzheimer’s disease points for accelerated epigenetic aging in neurodegeneration. Front. Aging Neurosci. 13, 639428 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Huo, Z. et al. DNA methylation variability in Alzheimer’s disease. Neurobiol. Aging 76, 35–44 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Palou-Márquez, G., Subirana, I., Nonell, L., Fernández-Sanlés, A. & Elosua, R. DNA methylation and gene expression integration in cardiovascular disease. Clin. Epigenetics 13, 75 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Movassagh, M. et al. Differential DNA methylation correlates with differential expression of angiogenic factors in human heart failure. PLoS ONE 5, e8564 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Fernández-Sanlés, A. et al. DNA methylation biomarkers of myocardial infarction and cardiovascular disease. Clin. Epigenetics 13, 86 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Klutstein, M., Moss, J., Kaplan, T. & Cedar, H. Contribution of epigenetic mechanisms to variation in cancer risk among tissues. Proc. Natl Acad. Sci. USA 114, 2230–2234 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Issa, J. P. Aging and epigenetic drift: a vicious cycle. J. Clin. Invest. 124, 24–29 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Klutstein, M., Nejman, D., Greenfield, R. & Cedar, H. DNA methylation in cancer and aging. Cancer Res. 76, 3446–3450 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Teschendorff, A. E. et al. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 20, 440–446 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hannum, G. et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol. Cell 49, 359–367 (2013). This paper demonstrates how DNAm is used to build an epigenetic age predictor in blood, widely known as Hannum’s blood clock. It also introduces concepts such as ‘entropy’ and a formula for calculating Shannon entropy using DNAm data.

    Article  CAS  PubMed  Google Scholar 

  42. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008). This paper outlines WGCNA, a powerful framework for correlation network analysis using DNAm or gene expression data.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Horvath, S. et al. Aging effects on DNA methylation modules in human brain and blood tissue. Genome Biol. 13, R97 (2012). This work demonstrates the usefulness of WGCNA for identifying correlation networks or ‘modules’ in blood and brain tissue that are associated with ageing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wilson, V. L., Smith, R. A., Ma, S. & Cutler, R. G. Genomic 5-methyldeoxycytidine decreases with age. J. Biol. Chem. 262, 9948–9951 (1987).

    Article  CAS  PubMed  Google Scholar 

  45. Fuke, C. et al. Age related changes in 5-methylcytosine content in human peripheral leukocytes and placentas: an HPLC-based study. Ann. Hum. Genet. 68, 196–204 (2004).

    Article  CAS  PubMed  Google Scholar 

  46. Vanyushin, B., Nemirovsky, L., Klimenko, V., Vasiliev, V. & Belozersky, A. The 5-methylcytosine in DNA of rats. Gerotologia 19, 138–152 (1973).

    Article  CAS  Google Scholar 

  47. Unnikrishnan, A. et al. Revisiting the genomic hypomethylation hypothesis of aging. Ann. NY. Acad. Sci. 1418, 69–79 (2018).

    Article  PubMed  Google Scholar 

  48. Heyn, H. et al. Distinct DNA methylomes of newborns and centenarians. Proc. Natl Acad. Sci. USA 109, 10522–10527 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Unnikrishnan, A. et al. The role of DNA methylation in epigenetics of aging. Pharmacol. Ther. 195, 172–185 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Lister, R. et al. Global epigenomic reconfiguration during mammalian brain development. Science 341, 1237905 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Raddatz, G. et al. Aging is associated with highly defined epigenetic changes in the human epidermis. Epigenetics Chromatin 6, 36 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hadad, N. et al. Absence of genomic hypomethylation or regulation of cytosine-modifying enzymes with aging in male and female mice. Epigenetics Chromatin 9, 30 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Cole, J. J. et al. Diverse interventions that extend mouse lifespan suppress shared age-associated epigenetic changes at critical gene regulatory regions. Genome Biol. 18, 58 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Eckhardt, F. et al. DNA methylation profiling of human chromosomes 6, 20 and 22. Nat. Genet. 38, 1378–1385 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Day, K. et al. Differential DNA methylation with age displays both common and dynamic features across human tissues that are influenced by CpG landscape. Genome Biol. 14, R102 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Mammalian Methylation Consortium et al. Universal DNA methylation age across mammalian tissues. Preprint at bioRxiv https://doi.org/10.1101/2021.01.18.426733v1 (2021). This paper performs a multi-tissue, multispecies EWAS of differential methylation and age from more than 59 tissue types and 128 mammalian species. The identification of age-associated DMPs that are shared between tissues and mammalian species is suggestive of a universal, evolutionarily conserved ageing mechanism that is tightly linked to development.

  57. Rakyan, V. K. et al. Human aging-associated DNA hypermethylation occurs preferentially at bivalent chromatin domains. Genome Res. 20, 434–439 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Bell, J. T. et al. DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines. Genome Biol. 12, R10 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Slieker, R. C. et al. Age-related accrual of methylomic variability is linked to fundamental ageing mechanisms. Genome Biol. 17, 191 (2016). This paper characterizes VMPs as a distinct class of age-associated DNAm changes. It also links VMPs to age-associated changes in gene expression and describes changes in Shannon entropy with age.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 14, R115 (2013). This paper describes Horvath’s pan-tissue epigenetic clock, an epigenetic age predictor for multiple human tissues and cell types.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Peters, T. J. et al. De novo identification of differentially methylated regions in the human genome. Epigenetics Chromatin 8, 6 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Jaffe, A. E. et al. Bump hunting to identify differentially methylated regions in epigenetic epidemiology studies. Int. J. Epidemiol. 41, 200–209 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Pedersen, B. S., Schwartz, D. A., Yang, I. V. & Kechris, K. J. comb-p: software for combining, analyzing, grouping and correcting spatially correlated P-values. Bioinformatics 28, 2986–2988 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Aryee, M. J. et al. Minfi: a flexible and comprehensive Bioconductor package for the analysis of Infinium DNA methylation microarrays. Bioinformatics 30, 1363–1369 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Butcher, L. M. & Beck, S. Probe Lasso: a novel method to rope in differentially methylated regions with 450K DNA methylation data. Methods 72, 21–28 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Field, A. E. et al. DNA methylation clocks in aging: categories, causes, and consequences. Mol. Cell 71, 882–895 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Levine, M. E. et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging 10, 573–591 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Horvath, S. et al. An epigenetic clock analysis of race/ethnicity, sex, and coronary heart disease. Genome Biol. 17, 171 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Voisin, S. et al. An epigenetic clock for human skeletal muscle. J. Cachexia. Sarcopenia Muscle 11, 887–898 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Bocklandt, S. et al. Epigenetic predictor of age. PLoS ONE 6, e14821 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Bell, C. G. et al. DNA methylation aging clocks: challenges and recommendations. Genome Biol. 20, 249 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Tibshirani, R. Regression shrinkage and selection via the Lasso. J. R. Stat. Soc. 58, 267–288 (1996).

    Google Scholar 

  74. Zou, H. & Hastie, T. Regularization and variable selection via the elastic net. J. R. Stat. Soc. Ser. B Stat. Methodol. 67, 301–320 (2005).

    Article  Google Scholar 

  75. Horvath, S. & Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 19, 371–384 (2018).

    Article  CAS  PubMed  Google Scholar 

  76. Zhu, T., Zheng, S. C., Paul, D. S., Horvath, S. & Teschendorff, A. E. Cell and tissue type independent age-associated DNA methylation changes are not rare but common. Aging 10, 3541–3557 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Shireby, G. L. et al. Recalibrating the epigenetic clock: implications for assessing biological age in the human cortex. Brain 143, 3763–3775 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Voisin, S. et al. Meta-analysis of genome-wide DNA methylation and integrative OMICs in human skeletal muscle. J. Cachexia. Sarcopenia Muscle 12, 1064–1078 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Thompson, M. J., Horvath, S. & Pellegrini, M. An epigenetic aging clock for dogs and wolves. Aging 9, 1055–1068 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Horvath, S. et al. DNA methylation aging and transcriptomic studies in horses. Nat. Commun. 13, 40 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Schachtschneider, K. M. et al. Epigenetic clock and DNA methylation analysis of porcine models of aging and obesity. Geroscience 43, 2467–2483 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Sugrue, V. J. et al. Castration delays epigenetic aging and feminizes dna methylation at androgen-regulated loci. eLife 10, e64932 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Wilkinson, G. S. et al. DNA methylation predicts age and provides insight into exceptional longevity of bats. Nat. Commun. 12, 1615 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Wang, T. et al. Epigenetic aging signatures in mice livers are slowed by dwarfism, calorie restriction and rapamycin treatment. Genome Biol. 18, 57 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Horvath, S. et al. DNA methylation clocks tick in naked mole rats but queens age more slowly than nonbreeders. Nat. Aging 2, 46–59 (2022).

    Article  PubMed  Google Scholar 

  86. Robeck, T. R. et al. Multi-species and multi-tissue methylation clocks for age estimation in toothed whales and dolphins. Commun. Biol. 4, 642 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Raj, K. et al. Epigenetic clock and methylation studies in cats. Geroscience 43, 2363–2378 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Prado, N. A. et al. Epigenetic clock and methylation studies in elephants. Aging Cell 20, e13414 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Horvath, S. et al. DNA methylation age analysis of rapamycin in common marmosets. Geroscience 43, 2413–2425 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Horvath, S. et al. Epigenetic clock and methylation studies in the rhesus macaque. Geroscience 43, 2441–2453 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Yuan, T. et al. An integrative multi-scale analysis of the dynamic DNA methylation landscape in aging. PLoS Genet. 11, e1004996 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Chen, B. H. et al. DNA methylation-based measures of biological age: meta-analysis predicting time to death. Aging 8, 1844–1865 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Teschendorff, A. E., West, J. & Beck, S. Age-associated epigenetic drift: implications, and a case of epigenetic thrift? Hum. Mol. Genet. 22, 7–15 (2013).

    Article  CAS  Google Scholar 

  94. Trapp, A., Kerepesi, C. & Gladyshev, V. N. Profiling epigenetic age in single cells. Nat. Aging 1, 1189–1201 (2021). This paper presents a novel computational framework for estimating the epigenetic age of single cells and demonstrates that individual cell types do not age at the same rate at the epigenetic level.

    Article  Google Scholar 

  95. Tejedor, J. R. & Fraga, M. F. Interindividual epigenetic variability: sound or noise? BioEssays 39, 1700055 (2017).

    Article  Google Scholar 

  96. Wang, Y., Pedersen, N. L. & Hägg, S. Implementing a method for studying longitudinal DNA methylation variability in association with age. Epigenetics 13, 866–874 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Fernández, A. F. et al. H3K4me1 marks DNA regions hypomethylated during aging in human stem and differentiated cells. Genome Res. 29, 27–40 (2015).

    Article  CAS  Google Scholar 

  98. Oh, G. et al. Epigenetic assimilation in the aging human brain. Genome Biol. 17, 76 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Phipson, B. & Oshlack, A. DiffVar: a new method for detecting differential variability with application to methylation in cancer and aging. Genome Biol. 15, 465 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Yusipov, I. et al. Age-related DNA methylation changes are sex-specific: a comprehensive assessment. Aging 12, 24057–24080 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Vershinina, O., Bacalini, M. G., Zaikin, A., Franceschi, C. & Ivanchenko, M. Disentangling age-dependent DNA methylation: deterministic, stochastic, and nonlinear. Sci. Rep. 11, 9201 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Planterose Jiménez, B. et al. Equivalent DNA methylation variation between monozygotic co-twins and unrelated individuals reveals universal epigenetic inter-individual dissimilarity. Genome Biol. 22, 18 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Fraga, M. F. et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl Acad. Sci. USA 102, 10604–10609 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Talens, R. P. et al. Epigenetic variation during the adult lifespan: cross-sectional and longitudinal data on monozygotic twin pairs. Aging Cell 11, 694–703 (2012).

    Article  CAS  PubMed  Google Scholar 

  105. Van Dongen, J. et al. Genetic and environmental influences interact with age and sex in shaping the human methylome. Nat. Commun. 7, 11115 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Breusch, T. S. & Pagan, A. R. A simple test for heteroscedasticity and random coefficient variation. Econometrica 47, 1287–1294 (1979).

    Article  Google Scholar 

  107. Mur, J. et al. DNA methylation in APOE: the relationship with Alzheimer’s and with cardiovascular health. Alzheimers Dement. Transl. Res. Clin. Interv. 6, e12026 (2020).

    Google Scholar 

  108. Hayflick, L. Entropy explains aging, genetic determinism explains longevity, and undefined terminology explains misunderstanding both. PLoS Genet. 3, 2351–2354 (2007).

    Article  CAS  Google Scholar 

  109. Martin-Herranz, D. et al. Screening for genes that accelerate the epigenetic ageing clock in humans reveals a role for the H3K36 methyltransferase NSD1. Genome Biol. 20, 146 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Hayano, M. et al. DNA break-induced epigenetic drift as a cause of mammalian aging. Preprint at bioRxiv https://doi.org/10.1101/808659 (2019).

  111. Jenkinson, G., Pujadas, E., Goutsias, J. & Feinberg, A. P. Potential energy landscapes identify the information-theoretic nature of the epigenome. Nat. Genet. 49, 719–729 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Yan, Q. et al. Epigenetic mutation load is weakly correlated with epigenetic age acceleration. Aging 12, 17863–17894 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Shannon, C. E. A mathematical theory of communication. Bell Syst. Tech. J. 27, 623–656 (1948).

    Article  Google Scholar 

  114. Sziráki, A., Tyshkovskiy, A. & Gladyshev, V. N. Global remodeling of the mouse DNA methylome during aging and in response to calorie restriction. Aging Cell 17, e12738 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Mendelsohn, A. R. & Larrick, J. W. The DNA methylome as a biomarker for epigenetic instability and human aging. Rejuvenation Res. 16, 74–77 (2013).

    Article  CAS  PubMed  Google Scholar 

  116. Rando, T. A. & Wyss-Coray, T. Asynchronous, contagious and digital aging. Nat. Aging 1, 29–35 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Rudolph, K. L. DNA-methylation aging at single-cell level. Nat. Aging 1, 1086–1087 (2021).

    Article  Google Scholar 

  118. Zhang, L. et al. DNA methylation landscape reflects the spatial organization of chromatin in different cells. Biophys. J. 113, 1395–1404 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Haerter, J. O., Lövkvist, C., Dodd, I. B. & Sneppen, K. Collaboration between CpG sites is needed for stable somatic inheritance of DNA methylation states. Nucleic Acids Res. 42, 2235–2244 (2014).

    Article  CAS  PubMed  Google Scholar 

  120. Li, G. et al. Joint profiling of DNA methylation and chromatin architecture in single cells. Nat. Methods 16, 991–993 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Lövkvist, C., Dodd, I. B., Sneppen, K. & Haerter, J. O. DNA methylation in human epigenomes depends on local topology of CpG sites. Nucleic Acids Res. 44, 5123–5132 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. Mallona, I., Ausso, S., Diez-Villanueva, A., Moreno, V. & Peinado, M. A. DNA co-methylation networks outline the structure and remodeling dynamics of colorectal cancer epigenome. Preprint at bioRxiv https://doi.org/10.1101/428730v2 (2020).

  123. Fortin, J. P. & Hansen, K. D. Reconstructing A/B compartments as revealed by Hi-C using long-range correlations in epigenetic data. Genome Biol. 16, 180 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. West, J., Widschwendter, M. & Teschendorff, A. E. Distinctive topology of age-associated epigenetic drift in the human interactome. Proc. Natl Acad. Sci. USA 110, 14138–14143 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Willis, C. R. G. et al. Network analysis of human muscle adaptation to aging and contraction. Aging 12, 740–755 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Zhang, B. & Horvath, S. A general framework for weighted gene co-expression network analysis. Stat. Appl. Genet. Mol. Biol. 4, 17 (2005).

    Article  Google Scholar 

  127. Yip, A. M. & Horvath, S. Gene network interconnectedness and the generalized topological overlap measure. BMC Bioinformatics 8, 22 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Bonder, M. J. et al. Disease variants alter transcription factor levels and methylation of their binding sites. Nat. Genet. 49, 131–138 (2017).

    Article  CAS  PubMed  Google Scholar 

  129. Belsky, D. W. et al. Quantification of biological aging in young adults. Proc. Natl Acad. Sci. USA 112, E4104–E4110 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Jylhävä, J., Pedersen, N. L. & Hägg, S. Biological age predictors. EBioMedicine 21, 29–36 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  131. Lacey, M., Baribault, C., Ehrlich, K. C. & Ehrlich, M. Atherosclerosis-associated differentially methylated regions can reflect the disease phenotype and are often at enhancers. Atherosclerosis 280, 183–191 (2019).

    Article  CAS  PubMed  Google Scholar 

  132. Xue, Y. et al. Aberrantly methylated-differentially expressed genes identify novel atherosclerosis risk subtypes. Front. Genet. 11, 569572 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Bakshi, C., Vijayvergiya, R. & Dhawan, V. Aberrant DNA methylation of M1-macrophage genes in coronary artery disease. Sci. Rep. 9, 1429 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Kim, J. Y. et al. Promoter methylation changes in ALOX12 and AIRE1: novel epigenetic markers for atherosclerosis. Clin. Epigenetics 12, 66 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Kazmi, N. et al. Associations between high blood pressure and DNA methylation. PLoS ONE 15, e0227728 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Richard, M. A. et al. DNA methylation analysis identifies loci for blood pressure regulation. Am. J. Hum. Genet. 101, 888–902 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Bacos, K. et al. Blood-based biomarkers of age-associated epigenetic changes in human islets associate with insulin secretion and diabetes. Nat. Commun. 7, 11089 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Dayeh, T. et al. Genome-wide DNA methylation analysis of human pancreatic islets from type 2 diabetic and non-diabetic donors identifies candidate genes that influence insulin secretion. PLoS Genet. 10, e1004160 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  139. Volkov, P. et al. Whole-genome bisulfite sequencing of human pancreatic islets reveals novel differentially methylated regions in type 2 diabetes pathogenesis. Diabetes 66, 1074–1085 (2017).

    Article  CAS  PubMed  Google Scholar 

  140. Barajas-Olmos, F. et al. Altered DNA methylation in liver and adipose tissues derived from individuals with obesity and type 2 diabetes. BMC Med. Genet. 19, 28 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Fernández-Tajes, J. et al. Genome-wide DNA methylation analysis of articular chondrocytes reveals a cluster of osteoarthritic patients. Ann. Rheum. Dis. 73, 668–677 (2014).

    Article  PubMed  CAS  Google Scholar 

  142. Aref-Eshghi, E. et al. Genome-wide DNA methylation study of hip and knee cartilage reveals embryonic organ and skeletal system morphogenesis as major pathways involved in osteoarthritis. BMC Musculoskelet. Disord. 16, 287 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Delgado-Calle, J. et al. Genome-wide profiling of bone reveals differentially methylated regions in osteoporosis and osteoarthritis. Arthritis Rheum. 65, 197–205 (2013).

    Article  CAS  PubMed  Google Scholar 

  144. Wang, Y., Li, F., Zhang, G., Kang, L. & Guan, H. Ultraviolet-B induces ERCC6 repression in lens epithelium cells of age-related nuclear cataract through coordinated DNA hypermethylation and histone deacetylation. Clin. Epigenetics 8, 62 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Teschendorff, A. E. et al. Correlation of smoking-associated DNA methylation changes in buccal cells with DNA methylation changes in epithelial cancer. JAMA Oncol. 1, 476–485 (2015).

    Article  PubMed  Google Scholar 

  146. Teschendorff, A. E. et al. DNA methylation outliers in normal breast tissue identify field defects that are enriched in cancer. Nat. Commun. 7, 10478 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Hansen, K. D. et al. Increased methylation variation in epigenetic domains across cancer types. Nat. Genet. 43, 768–775 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Pérez, R. F., Tejedor, J. R., Bayón, G. F., Fernández, A. F. & Fraga, M. F. Distinct chromatin signatures of DNA hypomethylation in aging and cancer. Aging Cell 17, e12744 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Chatsirisupachai, K., Lesluyes, T., Paraoan, L., Van Loo, P. & de Magalhães, J. P. An integrative analysis of the age-associated multi-omic landscape across cancers. Nat. Commun. 12, 2345 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Levine, M. E. et al. DNA methylation age of blood predicts future onset of lung cancer in the Women’s Health Initiative. Aging 7, 690–700 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Jones, A. et al. Role of DNA methylation and epigenetic silencing of HAND2 in endometrial cancer development. PLoS Med. 10, e1001551 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Lu, A. T. et al. Genetic architecture of epigenetic and neuronal ageing rates in human brain regions. Nat. Commun. 8, 15353 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Levine, M. E., Lu, A. T., Bennett, D. A. & Horvath, S. Epigenetic age of the pre-frontal cortex is associated with neuritic plaques, amyloid load, and Alzheimer’s disease related cognitive functioning. Aging 7, 1198–1211 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Quach, A. et al. Epigenetic clock analysis of diet, exercise, education, and lifestyle factors. Aging 9, 419–437 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Yang, Z. et al. Correlation of an epigenetic mitotic clock with cancer risk. Genome Biol. 17, 205 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Teschendorff, A. E. A comparison of epigenetic mitotic-like clocks for cancer risk prediction. Genome Med. 12, 56 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Breitling, L. P. et al. Frailty is associated with the epigenetic clock but not with telomere length in a German cohort. Clin. Epigenetics 8, 21 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  158. Vidal, L. et al. Specific increase of methylation age in osteoarthritis cartilage. Osteoarthr. Cartil. 24, S63–S534 (2016).

    Article  Google Scholar 

  159. Vidal-Bralo, L. et al. Specific premature epigenetic aging of cartilage in osteoarthritis. Aging 8, 2222–2231 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Horvath, S. & Ritz, B. R. Increased epigenetic age and granulocyte counts in the blood of Parkinson’s disease patients. Aging 7, 1130–1142 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Teschendorff, A. E. et al. Epigenetic variability in cells of normal cytology is associated with the risk of future morphological transformation. Genome Med. 4, 24 (2012). This paper demonstrates that VMPs that undergo age-associated changes overlap with VMPs in healthy tissue that will go on to develop cancer. This highlights the importance of investigating VMPs in the context of biological ageing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Landau, D. A. et al. Locally disordered methylation forms the basis of intratumor methylome variation in chronic lymphocytic leukemia. Cancer Cell 26, 813–825 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Li, E. & Zhang, Y. DNA methylation in mammals. Cold Spring Harb. Perspect. Biol. 6, a019133 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. Salameh, Y., Bejaoui, Y. & El Hajj, N. DNA methylation biomarkers in aging and age-related diseases. Front. Genet. 11, 171 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Deelen, J., Beekman, M., Capri, M., Franceschi, C. & Slagboom, P. E. Identifying the genomic determinants of aging and longevity in human population studies: progress and challenges. BioEssays 35, 386–396 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Capp, J. P. & Thomas, F. Tissue-disruption-induced cellular stochasticity and epigenetic drift: common origins of aging and cancer? BioEssays 43, e2000140 (2021).

    Article  PubMed  CAS  Google Scholar 

  167. Rakyan, V. K. et al. Identification of type 1 diabetes-associated DNA methylation variable positions that precede disease diagnosis. PLoS Genet. 7, e1002300 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Houseman, E. A. et al. Reference-free deconvolution of DNA methylation data and mediation by cell composition effects. BMC Bioinformatics 17, 259 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Birney, E., Smith, G. D. & Greally, J. M. Epigenome-wide association studies and the interpretation of disease -omics. PLoS Genet. 12, e1006105 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  170. Jones, P. A. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 13, 484–492 (2012).

    Article  CAS  PubMed  Google Scholar 

  171. Longo, V. D., Di Tano, M., Mattson, M. P. & Guidi, N. Intermittent and periodic fasting, longevity and disease. Nat. Aging 1, 47–59 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  172. Flanagan, E. W., Most, J., Mey, J. T. & Redman, L. M. Calorie restriction and aging in humans. Annu. Rev. Nutr. 40, 105–133 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Gensous, N. et al. The impact of caloric restriction on the epigenetic signatures of aging. Int. J. Mol. Sci. 20, 2022 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  174. Maegawa, S. et al. Caloric restriction delays age-related methylation drift. Nat. Commun. 8, 539 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  175. Hahn, O. et al. Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism. Genome Biol. 18, 56 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Kim, C. H. et al. Short-term calorie restriction ameliorates genomewide, age-related alterations in DNA methylation. Aging Cell 15, 1074–1081 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Hadad, N. et al. Caloric restriction mitigates age-associated hippocampal differential CG and non-CG methylation. Neurobiol. Aging 67, 53–66 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Haghani, A. et al. DNA methylation networks underlying mammalian traits. Preprint at bioRxiv https://doi.org/10.1101/2021.03.16.435708v1 (2021).

  179. Partridge, L., Fuentealba, M. & Kennedy, B. K. The quest to slow ageing through drug discovery. Nat. Rev. Drug. Discov. 19, 513–532 (2020).

    Article  CAS  PubMed  Google Scholar 

  180. Harrison, D. E. et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392–395 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Wilkinson, J. E. et al. Rapamycin slows aging in mice. Aging Cell 11, 675–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  182. Spilman, P. et al. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-β levels in a mouse model of Alzheimer’s disease. PLoS ONE 5, e9979 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Popovich, I. G. et al. Lifespan extension and cancer prevention in HER-2/neu transgenic mice treated with low intermittent doses of rapamycin. Cancer Biol. Ther. 15, 586–592 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Reifsnyder, P. C., Flurkey, K., Te, A. & Harrison, D. E. Rapamycin treatment benefits glucose metabolism in mouse models of type 2 diabetes. Aging 8, 3120–3130 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Neff, F. et al. Rapamycin extends murine lifespan but has limited effects on aging. J. Clin. Invest. 123, 3272–3291 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Urfer, S. R. et al. A randomized controlled trial to establish effects of short-term rapamycin treatment in 24 middle-aged companion dogs. Geroscience 39, 117–127 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Ross, C. et al. Metabolic consequences of long-term rapamycin exposure on common marmoset monkeys (Callithrix jacchus). Aging 7, 964–973 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Mannick, J. B. et al. mTOR inhibition improves immune function in the elderly. Sci. Transl Med. 6, 268ra179 (2014).

    Article  PubMed  CAS  Google Scholar 

  189. Horvath, S., Lu, A. T., Cohen, H. & Raj, K. Rapamycin retards epigenetic ageing of keratinocytes independently of its effects on replicative senescence, proliferation and differentiation. Aging 11, 3238–3249 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Novelle, M. G., Ali, A., Diéguez, C., Bernier, M. & de Cabo, R. Metformin: a hopeful promise in aging research. Cold Spring Harb. Perspect. Med. 6, a025932 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  191. Fahy, G. M. et al. Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell 18, e13028 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Li, M. et al. Effect of metformin on the epigenetic age of peripheral blood in patients with diabetes mellitus (DM). Preprint at Research Square https://doi.org/10.21203/rs.3.rs-131293/v1 (2021).

  193. Covarrubias, A. J., Perrone, R., Grozio, A. & Verdin, E. NAD+ metabolism and its roles in cellular processes during ageing. Nat. Rev. Mol. Cell Biol. 22, 119–141 (2021).

    Article  CAS  PubMed  Google Scholar 

  194. Massudi, H. et al. Age-associated changes in oxidative stress and NAD+ metabolism in human tissue. PLoS ONE 7, e42357 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Zhu, X. H., Lu, M., Lee, B. Y., Ugurbil, K. & Chen, W. In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences. Proc. Natl Acad. Sci. USA 112, 2876–2881 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Zhou, C. C. et al. Hepatic NAD+ deficiency as a therapeutic target for non-alcoholic fatty liver disease in ageing. Br. J. Pharmacol. 173, 2352–2368 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Clement, J., Wong, M., Poljak, A., Sachdev, P. & Braidy, N. The plasma NAD+ metabolome is dysregulated in ‘normal’ aging. Rejuvenation Res. 22, 121–130 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Katsyuba, E., Romani, M., Hofer, D. & Auwerx, J. NAD+ homeostasis in health and disease. Nat. Metab. 2, 9–31 (2020).

    Article  CAS  PubMed  Google Scholar 

  199. Zhang, T. & Kraus, W. L. SIRT1-dependent regulation of chromatin and transcription: linking NAD+ metabolism and signaling to the control of cellular functions. Biochim. Biophys. Acta 1804, 1666–1675 (2010).

    Article  CAS  PubMed  Google Scholar 

  200. Wakeling, L. A. et al. SIRT1 affects DNA methylation of polycomb group protein target genes, a hotspot of the epigenetic shift observed in ageing. Hum. Genomics 9, 14 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Salminen, A., Kaarniranta, K., Hiltunen, M. & Kauppinen, A. Krebs cycle dysfunction shapes epigenetic landscape of chromatin: novel insights into mitochondrial regulation of aging process. Cell. Signal. 26, 1598–1603 (2014).

    Article  CAS  PubMed  Google Scholar 

  202. Wang, Y. et al. α-Ketoglutarate ameliorates age-related osteoporosis via regulating histone methylations. Nat. Commun. 11, 5596 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Zhang, Z. et al. α-Ketoglutarate delays age-related fertility decline in mammals. Aging Cell 20, e13291 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Asadi Shahmirzadi, A. et al. α-Ketoglutarate, an endogenous metabolite, extends lifespan and compresses morbidity in aging mice. Cell Metab. 32, 447–456 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Amenyah, S. D. et al. Nutritional epigenomics and age-related disease. Curr. Dev. Nutr. 4, nzaa097 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Demidenko, O. et al. Rejuvant®, a potential life-extending compound formulation with α-ketoglutarate and vitamins, conferred an average 8 year reduction in biological aging, after an average of 7 months of use, in the TruAge DNA methylation test. Aging 13, 24485–24499 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Madeo, F., Eisenberg, T., Pietrocola, F. & Kroemer, G. Spermidine in health and disease. Science 359, eaan2788 (2018).

    Article  PubMed  CAS  Google Scholar 

  208. Soda, K. Spermine and gene methylation: a mechanism of lifespan extension induced by polyamine-rich diet. Amino Acids 52, 213–224 (2020).

    Article  CAS  PubMed  Google Scholar 

  209. Eisenberg, T. et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat. Med. 22, 1428–1438 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Yue, F. et al. Spermidine prolongs lifespan and prevents liver fibrosis and hepatocellular carcinoma by activating MAP1S-mediated autophagy. Cancer Res. 77, 2938–2951 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Kiechl, S. et al. Higher spermidine intake is linked to lower mortality: a prospective population-based study. Am. J. Clin. Nutr. 108, 371–380 (2018).

    Article  PubMed  Google Scholar 

  212. Soda, K., Kano, Y., Chiba, F., Koizumi, K. & Miyaki, Y. Increased polyamine intake inhibits age-associated alteration in global DNA methylation and 1,2-dimethylhydrazine-induced tumorigenesis. PLoS ONE 8, e64357 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Fukui, T., Soda, K., Takao, K. & Rikiyama, T. Extracellular spermine activates DNA methyltransferase 3A and 3B. Int. J. Mol. Sci. 20, 1254 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  214. Soda, K. Polyamine metabolism and gene methylation in conjunction with one-carbon metabolism. Int. J. Mol. Sci. 19, 3106 (2018).

    Article  PubMed Central  CAS  Google Scholar 

  215. Simpson, D. J., Olova, N. N. & Chandra, T. Cellular reprogramming and epigenetic rejuvenation. Clin. Epigenetics 13, 170 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  216. Lu, Y. et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature 588, 124–129 (2020). This work demonstrates that the delivery of three Yamanaka factors (OSK) to mouse retinal ganglion cells reprogrammes the epigenome and transcriptome, promotes axon regeneration after injury and improves vision loss in old mice. The paper also demonstrates that TET demethylation enzymes are necessary for this process to occur, suggesting DNAm is intrinsic to the ageing process and its functional reversal.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Gill, D. et al. Multi-omic rejuvenation of human cells by maturation phase transient reprogramming. eLife 11, e71624 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Taylor, D. Physical activity is medicine for older adults. Postgrad. Med. J. 90, 26–32 (2014).

    Google Scholar 

  219. Yan, X. et al. The gene SMART study: method, study design, and preliminary findings. BMC Genomics 18, 15–28 (2017).

    Article  CAS  Google Scholar 

  220. Smith, C. et al. Uncovering the bone–muscle interaction and its implications for the health and function of older adults (the Wellderly Project): protocol for a randomized controlled crossover trial. JMIR Res. Protoc. 10, e18777 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  221. Bell, J. T. et al. Epigenome-wide scans identify differentially methylated regions for age and age-related phenotypes in a healthy ageing population. PLoS Genet. 8, e1002629 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Thompson, R. F. et al. Tissue-specific dysregulation of DNA methylation in aging. Aging Cell 9, 506–518 (2010).

    Article  CAS  PubMed  Google Scholar 

  223. Lee, T. I. et al. Control of developmental regulators by Polycomb in human embryonic stem cells. Cell 125, 301–313 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem. Cells Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  225. Yu, M., Hazelton, W. D., Luebeck, G. E. & Grady, W. M. Epigenetic aging: more than just a clock when it comes to cancer. Cancer Res. 80, 367–374 (2020).

    Article  CAS  PubMed  Google Scholar 

  226. Luebeck, G. E. et al. Implications of epigenetic drift in colorectal neoplasia. Cancer Res. 79, 495–504 (2019).

    Article  CAS  PubMed  Google Scholar 

  227. Knight, A. K. et al. An epigenetic clock for gestational age at birth based on blood methylation data. Genome Biol. 17, 206 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  228. Kerepesi, C., Zhang, B., Lee, S.-G., Trapp, A. & Gladyshev, V. N. Epigenetic clocks reveal a rejuvenation event during embryogenesis followed by aging. Sci. Adv. 7, 4–11 (2021). This paper applies epigenetic clocks to track the precise changes in biological age during the early stages of prenatal development in mice and humans. The work demonstrates that there is a rejuvenation event during embryogenesis where biological age is reset to zero, which marks the beginning of organismal ageing.

    Article  CAS  Google Scholar 

  229. Raj, K. in Epigenetics of Aging and Longevity 95–118 (Elsevier, 2018).

  230. Chiavellini, P. et al. Aging and rejuvenation — a modular epigenome model. Aging 13, 4734–4746 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Raj, K. & Horvath, S. Current perspectives on the cellular and molecular features of epigenetic ageing. Exp. Biol. Med. 245, 1532–1542 (2020). This perspective piece proposes a mechanistic understanding of the biological phenomenon underpinning the epigenetic clock.

    Article  CAS  Google Scholar 

  232. Ermolaeva, M., Neri, F., Ori, A. & Rudolph, K. L. Cellular and epigenetic drivers of stem cell ageing. Nat. Rev. Mol. Cell Biol. 19, 594–610 (2018).

    Article  CAS  PubMed  Google Scholar 

  233. Jonkman, T. H. et al. Functional genomics analysis identifies T and NK cell activation as a driver of epigenetic clock progression. Genome Biol. 23, 24 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Dabin, J., Fortuny, A. & Polo, S. E. Epigenome maintenance in response to DNA damage. Mol. Cell 62, 712–727 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Jaiswal, S. & Ebert, B. L. Clonal hematopoiesis in human aging and disease. Science 366, eaan4673 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Klein, C. J. et al. Mutations in DNMT1 cause hereditary sensory neuropathy with dementia and hearing loss. Nat. Genet. 43, 595–600 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Baets, J. et al. Defects of mutant DNMT1 are linked to a spectrum of neurological disorders. Brain 138, 845–861 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  238. Bell-Pedersen, D. et al. Circadian rhythms from multiple oscillators: lessons from diverse organisms. Nat. Rev. Drug Discov. 6, 544–556 (2005).

    CAS  Google Scholar 

  239. Acosta-Rodríguez, V. A., Rijo-Ferreira, F., Green, C. B. & Takahashi, J. S. Importance of circadian timing for aging and longevity. Nat. Commun. 12, 2862 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  240. Etchegaray, J. P. & Mostoslavsky, R. Interplay between metabolism and epigenetics: a nuclear adaptation to environmental changes. Mol. Cell 62, 695–711 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Oh, E. S. & Petronis, A. Origins of human disease: the chrono-epigenetic perspective. Nat. Rev. Genet. 22, 533–546 (2021). This review describes the circadian dynamics of the epigenome and offers a new perspective into the proximal causes of ageing and disease.

    Article  CAS  PubMed  Google Scholar 

  242. Azzi, A. et al. Circadian behavior is light-reprogrammed by plastic DNA methylation. Nat. Neurosci. 17, 377–382 (2014).

    Article  CAS  PubMed  Google Scholar 

  243. Oh, G. et al. Circadian oscillations of cytosine modification in humans contribute to epigenetic variability, aging, and complex disease. Genome Biol. 20, 2 (2019). This paper reveals that oscillating CpGs are present in human neutrophils and demonstrates how CpG oscillations may contribute to epigenetic variability in humans, as well as ageing and risk for disease.

    Article  PubMed  PubMed Central  Google Scholar 

  244. Oh, G. et al. Cytosine modifications exhibit circadian oscillations that are involved in epigenetic diversity and aging. Nat. Commun. 9, 644 (2018). This paper demonstrates that CpGs in liver and lung tissues in mice exhibit circadian oscillations, which precede age-related changes in DNAm. The work also shows that the amplitudes of oscillating CpGs are highly correlated with linear age-dependent DNAm changes.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  245. Reinke, H. & Asher, G. Crosstalk between metabolism and circadian clocks. Nat. Rev. Mol. Cell Biol. 20, 227–241 (2019).

    Article  CAS  PubMed  Google Scholar 

  246. Takahashi, J. S. Transcriptional architecture of the mammalian circadian clock. Nat. Rev. Genet. 18, 164–179 (2017).

    Article  CAS  PubMed  Google Scholar 

  247. Yeung, J. & Naef, F. Rhythms of the genome: circadian dynamics from chromatin topology, tissue-specific gene expression, to behavior. Trends Genet. 34, 915–926 (2018).

    Article  CAS  PubMed  Google Scholar 

  248. Mure, L. S. et al. Diurnal transcriptome atlas of a primate across major neural and peripheral tissues. Science 359, eaao0318 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  249. Ruben, M. D. et al. A database of tissue-specific rhythmically expressed human genes has potential applications in circadian medicine. Sci. Transl Med. 10, eaat8806 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  250. López-Otín, C., Galluzzi, L., Freije, J. M. P., Madeo, F. & Kroemer, G. Metabolic control of longevity. Cell 166, 802–821 (2016).

    Article  PubMed  CAS  Google Scholar 

  251. Zwighaft, Z. et al. Circadian clock control by polyamine levels through a mechanism that declines with age. Cell Metab. 22, 874–885 (2015).

    Article  CAS  PubMed  Google Scholar 

  252. Levine, D. C. et al. NAD+ controls circadian reprogramming through PER2 nuclear translocation to counter aging. Mol. Cell 78, 835–849.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Asher, G. et al. Poly(ADP-ribose) polymerase 1 participates in the phase entrainment of circadian clocks to feeding. Cell 142, 943–953 (2010).

    Article  CAS  PubMed  Google Scholar 

  254. Kumar, V. & Takahashi, J. S. PARP around the clock. Cell 142, 841–843 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Imai, S. & Guarente, L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 24, 464–471 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Yang, J.-H. et al. Erosion of the epigenetic landscape and loss of cellular identity as a cause of aging in mammals. Preprint at bioRxiv https://doi.org/10.1101/808642v1 (2019).

  257. Schumacher, B., Pothof, J., Vijg, J. & Hoeijmakers, J. H. J. The central role of DNA damage in the ageing process. Nature 592, 695–703 (2021). This paper positions DNA damage as a unifying causal mechanism that leads to ageing.

    Article  CAS  PubMed  Google Scholar 

  258. Ciccarone, F., Zampieri, M. & Caiafa, P. PARP1 orchestrates epigenetic events setting up chromatin domains. Semin. Cell Dev. Biol. 63, 123–134 (2017).

    Article  CAS  PubMed  Google Scholar 

  259. Oberdoerffer, P. et al. SIRT1 redistribution on chromatin promotes genomic stability but alters gene expression during aging. Cell 135, 907–918 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Shimizu, I., Yoshida, Y., Suda, M. & Minamino, T. DNA damage response and metabolic disease. Cell Metab. 20, 967–977 (2014).

    Article  CAS  PubMed  Google Scholar 

  261. Jing, H. & Lin, H. Sirtuins in epigenetic regulation. Chem. Rev. 115, 2350–2375 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. O’Hagan, H. M., Mohammad, H. P. & Baylin, S. B. Double strand breaks can initiate gene silencing and SIRT1-dependent onset of DNA methylation in an exogenous promoter CpG island. PLoS Genet. 4, e1000155 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  263. O’Hagan, H. M. et al. Oxidative damage targets complexes containing DNA methyltransferases, SIRT1, and Polycomb members to promoter CpG islands. Cancer Cell 20, 606–619 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  264. O’Hagan, H. M. Chromatin modifications during repair of environmental exposure-induced DNA damage: a potential mechanism for stable epigenetic alterations. Environ. Mol. Mutagen. 55, 278–291 (2014).

    Article  PubMed  CAS  Google Scholar 

  265. Ding, N. et al. Mismatch repair proteins recruit DNA methyltransferase 1 to sites of oxidative DNA damage. J. Mol. Cell Biol. 8, 244–254 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Jung, M. & Pfeifer, G. P. Aging and DNA methylation. BMC Biol. 13, 7 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  267. Booth, L. & Brunet, A. The ageing epigenome. Mol. Cell 62, 728–744 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  268. Khokhlova, E., Fesenko, Z. S., Sopova, J. V. & Leonova, E. I. Features of DNA repair in the early stages of mammalian embryonic development. Genes 11, 1138 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  269. Vilenchik, M. M. & Knudson, A. G. Endogenous DNA double-strand breaks: production, fidelity of repair, and induction of cancer. Proc. Natl Acad. Sci. USA 100, 12871–12876 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Liu, Z. et al. Underlying features of epigenetic aging clocks in vivo and in vitro. Aging Cell 19, e13229 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Christiansen, C. et al. Novel DNA methylation signatures of tobacco smoking with trans-ethnic effects. Clin. Epigenetics 13, 36 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Cohen, A. A. Aging across the tree of life: the importance of a comparative perspective for the use of animal models in aging. Biochim. Biophys. Acta 1864, 2680–2689 (2018).

    Article  CAS  Google Scholar 

  273. Arneson, A. et al. A mammalian methylation array for profiling methylation levels at conserved sequences. Nat. Commun. 13, 783 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Horvath, S. et al. Reversing age: dual species measurement of epigenetic age with a single clock. Preprint at bioRxiv https://doi.org/10.1101/2020.05.07.082917 (2020).

  275. Lenart, P., Kuruczova, D., Joshi, P. K. & Bienertová-Vašků, J. Male mortality rates mirror mortality rates of older females. Sci. Rep. 9, 10589 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  276. Link, C., Jaffe, A. E. & Irizarry, R. A. Accounting for cellular heterogeneity is critical in epigenome-wide association studies. Genome Biol. 15, R31 (2014).

    Article  Google Scholar 

  277. Huang, Y. et al. The behaviour of 5-hydroxymethylcytosine in bisulfite sequencing. PLoS ONE 5, e8888 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  278. Jin, S. G., Kadam, S. & Pfeifer, G. P. Examination of the specificity of DNA methylation profiling techniques towards 5-methylcytosine and 5-hydroxymethylcytosine. Nucleic Acids Res. 38, e125 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  279. Spiers, H., Hannon, E., Schalkwyk, L. C., Bray, N. J. & Mill, J. 5-Hydroxymethylcytosine is highly dynamic across human fetal brain development. BMC Genomics 18, 738 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  280. Willer, C. J., Li, Y. & Abecasis, G. R. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–2191 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  281. Gladyshev, V. N. The ground zero of organismal life and aging. Trends Mol. Med. 27, 11–19 (2021).

    Article  CAS  PubMed  Google Scholar 

  282. Smyth, G. limma: Linear Models for Microarray Data (Springer, 2005).

  283. Guo, S. et al. Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat. Genet. 49, 635–642 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  284. Vanderkraats, N. D., Hiken, J. F., Decker, K. F. & Edwards, J. R. Discovering high-resolution patterns of differential DNA methylation that correlate with gene expression changes. Nucleic Acids Res. 41, 6816–6827 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  285. Schlosberg, C. E., VanderKraats, N. D. & Edwards, J. R. Modeling complex patterns of differential DNA methylation that associate with gene expression changes. Nucleic Acids Res. 45, 5100–5111 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  286. Friedman, J., Hastie, T. & Tibshirani, R. Regularization paths for generalized linear models via coordinate descent. J. Stat. Softw. 33, 1–22 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

K.S. and S.V. researched the literature and wrote the manuscript. All authors provided substantial contributions to discussions of the content and reviewed and/or edited the manuscript before submission.

Corresponding authors

Correspondence to Nir Eynon or Sarah Voisin.

Ethics declarations

Competing interests

S.H. is a founder of the non-profit Epigenetic Clock Development Foundation, which plans to license several patents from his employer UC Regents. These patents list S.H. as inventor. All other authors declare no competing interests.

Peer review

Peer Review Information

Nature Reviews Genetics thanks Mario F. Fraga who co-reviewed with Raúl F. Pérez, Sara Hägg, and David Sinclair who co-reviewed with Patrick T. Griffin for their contribution to the peer review of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

blockFinder: https://bioconductor.org/packages/release/bioc/html/minfi.html

bumphunter: https://bioconductor.org/packages/release/bioc/html/bumphunter.html

comb-p: https://github.com/brentp/combined-pvalues

DiffVar: https://bioconductor.org/packages/release/bioc/html/missMethyl.html

DMRcate: https://bioconductor.org/packages/release/bioc/html/DMRcate.html

glmnet: https://CRAN.R-project.org/package=glmnet

limma: https://bioconductor.org/packages/release/bioc/html/limma.html

Probe Lasso: https://www.bioconductor.org/packages/release/bioc/html/ChAMP.html

TAME trial: https://www.afar.org/tame-trial

WGCNA: https://horvath.genetics.ucla.edu/html/CoexpressionNetwork/Rpackages/WGCNA/

Glossary

CpGs

(Cytosine–guanine dinucleotides). Regions of DNA whereby a cytosine resides alongside a guanine to form a CpG.

Whole-genome bisulfite sequencing

A sequencing technology to survey and quantify DNA methylation (DNAm) at the single-base resolution on a genome-wide scale.

Reduced representation bisulfite sequencing

A sequencing technology to quantify DNA methylation (DNAm) at the single-base resolution at specific regions of interest, such as promoters and repeat regions.

Epigenome-wide association studies

(EWAS). The genome-wide investigations of the association of epigenetic marks, such as DNA methylation (DNAm), and a disease or trait of interest.

Methylation fraction

For a given DNA methylation (DNAm) locus in the human genome, the proportion of DNA strands that are methylated relative to the total number of DNA strands (0–100%), in a given population of cells.

Heteroscedastic

The variance of the residuals of a variable is non-constant or unequal across a range of values.

Breusch–Pagan test

A statistical test for heteroscedasticity of the errors in a linear regression model. It works by determining whether the errors of the response variable (such as DNA methylation (DNAm)) are dependent on the independent variable with continuous values (such as age).

White test

A statistical test for heteroscedasticity of the errors in a regression model. Unlike the Breusch–Pagan test, the White test can be used to identify both linear and non-linear forms of heteroscedasticity.

Bartlett’s test

A statistical test for heteroscedasticity between two discrete groups. This test assumes normality for each group and is thus sensitive to departures from normality.

Levene’s test

A statistical test for heteroscedasticity that compares deviations from the mean between groups.

Brown–Forsythe test

A statistical test for heteroscedasticity that compares deviations from the median between groups.

R package

A collection of functions, code, documentation and data bundled into a standardized format that can be downloaded and installed by R users.

Singular value decomposition

(SVD). A technique used to reduce the dimensionality of a data matrix. This is useful for identifying sources of variation.

Homoscedastic

The variance of the residuals of a variable is constant across a range of values.

GrimAge clock

An epigenetic clock that is a predictor of both lifespan and healthspan. DNA methylation (DNAm) GrimAge is the output of an epigenetic clock, which utilizes a machine learning algorithm trained against a linear combination of chronological age, sex, DNAm-based surrogate biomarkers for seven plasma proteins and smoking pack-years.

TruAge clock

A direct-to-consumer epigenetic age test built using a machine learning algorithm trained to predict chronological age of a saliva sample from a limited number of cytosine–guanine dinucleotides (CpGs) in CpG islands (CGIs) and promoter regions.

Polycomb repressive complexes

(PRCs). Multiprotein complexes of Polycomb group proteins. PRCs modify epigenetic marks to control gene repression and are involved in regulating developmental genes in a multitude of cell types, including embryonic and adult stem cells.

CpG islands

(CGIs). Regions of the genome containing a high frequency of cytosine–guanine dinucleotide (CpG) repeats.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Seale, K., Horvath, S., Teschendorff, A. et al. Making sense of the ageing methylome. Nat Rev Genet 23, 585–605 (2022). https://doi.org/10.1038/s41576-022-00477-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-022-00477-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing