Age-associated epigenetic drift: implications, and a case of epigenetic thrift?

It is now well established that the genomic landscape of DNA methylation (DNAm) gets altered as a function of age, a process we here call ‘epigenetic drift’. The biological, functional, clinical and evolutionary significance of this epigenetic drift, however, remains unclear. We here provide a brief review of epigenetic drift, focusing on the potential implications for ageing, stem cell biology and disease risk prediction. It has been demonstrated that epigenetic drift affects most of the genome, suggesting a global deregulation of DNAm patterns with age. A component of this drift is tissue-specific, allowing remarkably accurate age-predictive models to be constructed. Another component is tissue-independent, targeting stem cell differentiation pathways and affecting stem cells, which may explain the observed decline of stem cell function with age. Age-associated increases in DNAm target developmental genes, overlapping those associated with environmental disease risk factors and with disease itself, notably cancer. In particular, cancers and precursor cancer lesions exhibit aggravated age DNAm signatures. Epigenetic drift is also influenced by genetic factors. Thus, drift emerges as a promising biomarker for premature or biological ageing, and could potentially be used in geriatrics for disease risk prediction. Finally, we propose, in the context of human evolution, that epigenetic drift may represent a case of epigenetic thrift, or bet-hedging. In summary, this review demonstrates the growing importance of the ‘ageing epigenome’, with potentially far-reaching implications for understanding the effect of age on stem cell function and differentiation, as well as for disease prevention.

[1]  S. Ritz‐Timme,et al.  Molecular pathology and age estimation. , 2010, Forensic science international.

[2]  A. Feinberg,et al.  Intra-individual change over time in DNA methylation with familial clustering. , 2008, JAMA.

[3]  Martin Widschwendter,et al.  Distinctive topology of age-associated epigenetic drift in the human interactome , 2013, Proceedings of the National Academy of Sciences.

[4]  James A. Cuff,et al.  A Bivalent Chromatin Structure Marks Key Developmental Genes in Embryonic Stem Cells , 2006, Cell.

[5]  B. Lyn‐Cook,et al.  Age and gender affect DNMT3a and DNMT3b expression in human liver , 2008, Cell Biology and Toxicology.

[6]  T. Ideker,et al.  Genome-wide methylation profiles reveal quantitative views of human aging rates. , 2013, Molecular cell.

[7]  Finn Verner Jensen,et al.  Bayesian networks , 1998, Data Mining and Knowledge Discovery Handbook.

[8]  I. Weissman,et al.  Functionally distinct hematopoietic stem cells modulate hematopoietic lineage potential during aging by a mechanism of clonal expansion , 2010, Proceedings of the National Academy of Sciences.

[9]  Alfonso Valencia,et al.  Distinct DNA methylomes of newborns and centenarians , 2012, Proceedings of the National Academy of Sciences.

[10]  Jian-Bing Fan,et al.  Genome‐wide DNA methylation profiling , 2010, Wiley interdisciplinary reviews. Systems biology and medicine.

[11]  K. Maiese,et al.  The Wnt signaling pathway: aging gracefully as a protectionist? , 2008, Pharmacology & therapeutics.

[12]  D. Balding,et al.  Epigenome-wide association studies for common human diseases , 2011, Nature Reviews Genetics.

[13]  S. Baylin,et al.  Epigenetic gene silencing in cancer – a mechanism for early oncogenic pathway addiction? , 2006, Nature Reviews Cancer.

[14]  Andres Kriete,et al.  Robustness and aging - A systems-level perspective , 2013, Biosyst..

[15]  M. Kayser,et al.  Estimating human age from T-cell DNA rearrangements , 2010, Current Biology.

[16]  M. Pembrey,et al.  Time to take epigenetic inheritance seriously , 2002, European Journal of Human Genetics.

[17]  Andrew E. Teschendorff,et al.  Independent surrogate variable analysis to deconvolve confounding factors in large-scale microarray profiling studies , 2011, Bioinform..

[18]  DNA methylation studies using twins: what are they telling us? , 2012, Genome Biology.

[19]  T. C. Goldsmith Aging, evolvability, and the individual benefit requirement; medical implications of aging theory controversies. , 2008, Journal of theoretical biology.

[20]  Kirsten R. McEwen,et al.  An Unbiased Assessment of the Role of Imprinted Genes in an Intergenerational Model of Developmental Programming , 2012, PLoS genetics.

[21]  J. Mitteldorf Chaotic population dynamics and the evolution of ageing , 2006 .

[22]  Martin J. Aryee,et al.  Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in Rheumatoid Arthritis , 2013, Nature Biotechnology.

[23]  A. Aviv,et al.  Obesity, cigarette smoking, and telomere length in women , 2005, The Lancet.

[24]  Zhiping Weng,et al.  Paternally Induced Transgenerational Environmental Reprogramming of Metabolic Gene Expression in Mammals , 2010, Cell.

[25]  Wolfgang Wagner,et al.  Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. , 2010, Genome research.

[26]  H. Kitchener,et al.  Epigenetic variability in cells of normal cytology is associated with the risk of future morphological transformation , 2012, Genome Medicine.

[27]  Zohar Yakhini,et al.  Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer , 2007, Nature Genetics.

[28]  M. Esteller,et al.  Aberrant DNA methylation profiles in the premature aging disorders Hutchinson-Gilford Progeria and Werner syndrome , 2013, Epigenetics.

[29]  W. Wagner,et al.  Epigenetic-aging-signature to determine age in different tissues , 2011, Aging.

[30]  J. Mitteldorf Aging is not a process of wear and tear. , 2010, Rejuvenation research.

[31]  C. Hales,et al.  Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. 1992. , 2013, International journal of epidemiology.

[32]  N. Samani,et al.  Biological ageing and cardiovascular disease , 2008, Heart.

[33]  G. Satten,et al.  Age-associated DNA methylation in pediatric populations. , 2012, Genome research.

[34]  J. Mitteldorf Evolutionary Origins of Aging , 2010 .

[35]  F. Taddei,et al.  Bet-hedging and epigenetic inheritance in bacterial cell development , 2008, Proceedings of the National Academy of Sciences.

[36]  R. Yung,et al.  The Role of Epigenetics in Aging and Autoimmunity , 2010, Clinical reviews in allergy & immunology.

[37]  M. Pembrey,et al.  Sex-specific, male-line transgenerational responses in humans , 2006, European Journal of Human Genetics.

[38]  Zachary D. Smith,et al.  Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. , 2013, Cell stem cell.

[39]  David M. Simcha,et al.  Tackling the widespread and critical impact of batch effects in high-throughput data , 2010, Nature Reviews Genetics.

[40]  Stephan Beck,et al.  Taking the measure of the methylome , 2010, Nature Biotechnology.

[41]  A. Teschendorff,et al.  An Epigenetic Signature in Peripheral Blood Predicts Active Ovarian Cancer , 2009, PloS one.

[42]  T. Rando,et al.  Intrinsic Changes and Extrinsic Influences of Myogenic Stem Cell Function During Aging , 2007, Stem Cell Reviews.

[43]  O. Rando,et al.  Rewriting the Epigenome , 2012, Cell.

[44]  E. Nestler Epigenetics: Stress makes its molecular mark , 2012, Nature.

[45]  A. Feinberg,et al.  Comprehensive High‐Throughput Arrays for Relative Methylation (CHARM) , 2010, Current protocols in human genetics.

[46]  S. Baylin,et al.  Switch from monoallelic to biallelic human IGF2 promoter methylation during aging and carcinogenesis. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[47]  S. Baylin,et al.  Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon , 1994, Nature Genetics.

[48]  E. Cerasi,et al.  DIABETES MELLITUS , 1924, Nihon rinsho. Japanese journal of clinical medicine.

[49]  I. Ellis,et al.  A 1 Mb minimal amplicon at 8p11–12 in breast cancer identifies new candidate oncogenes , 2005, Oncogene.

[50]  I. Weissman,et al.  Cell intrinsic alterations underlie hematopoietic stem cell aging. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[51]  C. Rosano,et al.  Human sirtuins: an overview of an emerging drug target in age-related diseases and cancer. , 2013, Current drug targets.

[52]  E. Blackburn Cell biology: telomeres sans frontières. , 1990, Nature.

[53]  C. Keller,et al.  Increased Wnt Signaling During Aging Alters Muscle Stem Cell Fate and Increases Fibrosis , 2007, Science.

[54]  S. Horvath,et al.  Aging effects on DNA methylation modules in human brain and blood tissue , 2012, Genome Biology.

[55]  J. Issa,et al.  Epigenetic variation and cellular Darwinism , 2011, Nature Genetics.

[56]  S. Stevens,et al.  Blood leucocyte telomere DNA content predicts vascular telomere DNA content in humans with and without vascular disease. , 2008, European heart journal.

[57]  N. Ahuja,et al.  Accelerated age-related CpG island methylation in ulcerative colitis. , 2001, Cancer research.

[58]  L. Gordon,et al.  Longitudinal, genome-scale analysis of DNA methylation in twins from birth to 18 months of age reveals rapid epigenetic change in early life and pair-specific effects of discordance , 2013, Genome Biology.

[59]  Wells Jn,et al.  Thrift: a guide to thrifty genes, thrifty phenotypes and thrifty norms , 2009, International Journal of Obesity.

[60]  C. Hales,et al.  Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis , 1992, Diabetologia.

[61]  Quaid Morris,et al.  Computational purification of individual tumor gene expression profiles leads to significant improvements in prognostic prediction , 2013, Genome Medicine.

[62]  Ingo Ruczinski,et al.  Detectable clonal mosaicism from birth to old age and its relationship to cancer , 2012, Nature Genetics.

[63]  Robin M. Murray,et al.  Epigenome-Wide Scans Identify Differentially Methylated Regions for Age and Age-Related Phenotypes in a Healthy Ageing Population , 2012, PLoS genetics.

[64]  Scott M Langevin,et al.  Erratum to , 2012, Epigenetics.

[65]  E. Verdin,et al.  Sirtuins: critical regulators at the crossroads between cancer and aging , 2007, Oncogene.

[66]  Debashis Sahoo,et al.  Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age , 2011, Proceedings of the National Academy of Sciences.

[67]  E. Blackburn,et al.  Telomeres and telomerase: the path from maize, Tetrahymena and yeast to human cancer and aging , 2006, Nature Medicine.

[68]  T. Spector,et al.  Epigenetic differences arise during the lifetime of monozygotic twins. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[69]  P. Laird,et al.  Epigenetic stem cell signature in cancer , 2007, Nature Genetics.

[70]  Irving L. Weissman,et al.  A comprehensive methylome map of lineage commitment from hematopoietic progenitors , 2010, Nature.

[71]  Brian K. Kennedy,et al.  Sirtuins in Aging and Age-Related Disease , 2006, Cell.

[72]  Steve Horvath,et al.  Epigenetic Predictor of Age , 2011, PloS one.

[73]  A. Bird,et al.  CpG islands and the regulation of transcription. , 2011, Genes & development.

[74]  J. Hoeijmakers DNA damage, aging, and cancer. , 2009, The New England journal of medicine.

[75]  M. Toyota,et al.  Inflammation-related aberrant patterns of DNA methylation: detection and role in epigenetic deregulation of cancer cell transcriptome. , 2009, Methods in molecular biology.

[76]  A. Ferguson-Smith,et al.  CG dinucleotide periodicities recognized by the Dnmt3a–Dnmt3L complex are distinctive at retroelements and imprinted domains , 2009, Mammalian Genome.

[77]  M. Esteller,et al.  Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome , 2011, Epigenetics.

[78]  Giovanni Parmigiani,et al.  Half or more of the somatic mutations in cancers of self-renewing tissues originate prior to tumor initiation , 2013, Proceedings of the National Academy of Sciences.

[79]  M. Ross,et al.  Meta-analysis of IDH-mutant cancers identifies EBF1 as an interaction partner for TET2 , 2013, Nature Communications.

[80]  Paul Bertone,et al.  Widespread resetting of DNA methylation in glioblastoma-initiating cells suppresses malignant cellular behavior in a lineage-dependent manner. , 2013, Genes & development.

[81]  S. S. Ajay,et al.  Genome-wide DNA methylation profiles in hematopoietic stem and progenitor cells reveal overrepresentation of ETS transcription factor binding sites , 2012, Genome research.

[82]  T. Reed,et al.  Shared environmental factors associated with telomere length maintenance in elderly male twins , 2007, Aging cell.

[83]  J. Skurnick,et al.  Telomere Length as an Indicator of Biological Aging: The Gender Effect and Relation With Pulse Pressure and Pulse Wave Velocity , 2001, Hypertension.

[84]  T. Spector,et al.  Meta-analysis of telomere length in 19 713 subjects reveals high heritability, stronger maternal inheritance and a paternal age effect , 2013, European Journal of Human Genetics.

[85]  Julia Krushkal,et al.  Parental ages and levels of DNA methylation in the newborn are correlated , 2011, BMC Medical Genetics.

[86]  E. Epel,et al.  Telomeres and adversity: Too toxic to ignore , 2012, Nature.

[87]  João Pedro de Magalhães,et al.  Meta-analysis of age-related gene expression profiles identifies common signatures of aging , 2009, Bioinform..

[88]  Howard Y. Chang,et al.  Aging, Rejuvenation, and Epigenetic Reprogramming: Resetting the Aging Clock , 2012, Cell.

[89]  Megan F. Cole,et al.  Control of Developmental Regulators by Polycomb in Human Embryonic Stem Cells , 2006, Cell.

[90]  Tim Fenton,et al.  Identification and functional validation of HPV-mediated hypermethylation in head and neck squamous cell carcinoma , 2013, Genome Medicine.

[91]  G. Fahy,et al.  The future of aging : pathways to human life extension , 2010 .

[92]  J. Issa,et al.  Age-Related DNA Methylation Changes in Normal Human Prostate Tissues , 2007, Clinical Cancer Research.

[93]  J. Neel Diabetes mellitus: a "thrifty" genotype rendered detrimental by "progress"? , 1962, American journal of human genetics.

[94]  Arturas Petronis,et al.  Epigenetics as a unifying principle in the aetiology of complex traits and diseases , 2010, Nature.

[95]  Devin C. Koestler,et al.  DNA methylation arrays as surrogate measures of cell mixture distribution , 2012, BMC Bioinformatics.

[96]  Martin J Aryee,et al.  Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts , 2009, Nature Genetics.

[97]  E. Epel,et al.  Telomeres and lifestyle factors: roles in cellular aging. , 2012, Mutation research.

[98]  E. Blackburn Telomeres sans frontieres , 1990, Nature.

[99]  I. Komuro,et al.  UTF1 is a chromatin-associated protein involved in ES cell differentiation , 2007, The Journal of cell biology.

[100]  A. Feinberg,et al.  Increased methylation variation in epigenetic domains across cancer types , 2011, Nature Genetics.

[101]  P. O’Reilly,et al.  Identification of seven loci affecting mean telomere length and their association with disease , 2013, Nature Genetics.

[102]  Nianxiang Zhang,et al.  Widespread and Tissue Specific Age-related Dna Methylation Material Supplemental Related Content a Hallmark of Cancer Age-dependent Dna Methylation of Genes That Are Suppressed in Stem Cells Is , 2022 .

[103]  C. N. Hales,et al.  Non-insulin-dependent diabetes mellitus. , 1997, British medical bulletin.

[104]  Owen T McCann,et al.  Human aging-associated DNA hypermethylation occurs preferentially at bivalent chromatin domains. , 2010, Genome research.

[105]  B. Christensen,et al.  Aging and Environmental Exposures Alter Tissue-Specific DNA Methylation Dependent upon CpG Island Context , 2009, PLoS genetics.

[106]  N. Ahuja,et al.  Aging, methylation and cancer. , 2000, Histology and histopathology.

[107]  M. Blasco,et al.  Telomere length, stem cells and aging. , 2007, Nature chemical biology.

[108]  John D. Storey,et al.  Capturing Heterogeneity in Gene Expression Studies by Surrogate Variable Analysis , 2007, PLoS genetics.

[109]  Kelly M. McGarvey,et al.  A stem cell–like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing , 2007, Nature Genetics.

[110]  B. Korn,et al.  DNA methylation pattern changes upon long-term culture and aging of human mesenchymal stromal cells , 2010, Aging cell.

[111]  Scott M Langevin,et al.  The influence of aging, environmental exposures and local sequence features on the variation of DNA methylation in blood , 2011, Epigenetics.

[112]  T. Rohlf,et al.  Is adult stem cell aging driven by conflicting modes of chromatin remodeling? , 2012, BioEssays : news and reviews in molecular, cellular and developmental biology.

[113]  R. Stöger The thrifty epigenotype: An acquired and heritable predisposition for obesity and diabetes? , 2008, BioEssays : news and reviews in molecular, cellular and developmental biology.

[114]  Frank Lyko,et al.  Genome-wide promoter DNA methylation dynamics of human hematopoietic progenitor cells during differentiation and aging. , 2011, Blood.

[115]  B. Korn,et al.  Aging and Chronic Sun Exposure Cause Distinct Epigenetic Changes in Human Skin , 2010, PLoS genetics.

[116]  Andrew E. Teschendorff,et al.  An integrative network algorithm identifies age-associated differential methylation interactome hotspots targeting stem-cell differentiation pathways , 2013, Scientific Reports.

[117]  S. Baylin,et al.  Aging and DNA methylation in colorectal mucosa and cancer. , 1998, Cancer research.

[118]  Lee E. Edsall,et al.  Human DNA methylomes at base resolution show widespread epigenomic differences , 2009, Nature.

[119]  S. Lam,et al.  Genome-scale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression , 2012, Genome research.

[120]  Thomas A. Down,et al.  Identification of Type 1 Diabetes–Associated DNA Methylation Variable Positions That Precede Disease Diagnosis , 2010, PLoS genetics.

[121]  Gavin Giovannoni,et al.  Inactive or moderately active human promoters are enriched for inter-individual epialleles , 2013, Genome Biology.