An epigenetic biomarker of aging for lifespan and healthspan

Morgan E Levine, Ake T Lu, Austin Quach, Brian H Chen, Themistocles L Assimes, Stefania Bandinelli, Lifang Hou, Andrea A Baccarelli, James D Stewart, Yun Li, Eric A Whitsel, James G Wilson, Alex P Reiner, Abraham Aviv, Kurt Lohman, Yongmei Liu, Luigi Ferrucci, Steve Horvath, Morgan E Levine, Ake T Lu, Austin Quach, Brian H Chen, Themistocles L Assimes, Stefania Bandinelli, Lifang Hou, Andrea A Baccarelli, James D Stewart, Yun Li, Eric A Whitsel, James G Wilson, Alex P Reiner, Abraham Aviv, Kurt Lohman, Yongmei Liu, Luigi Ferrucci, Steve Horvath

Abstract

Identifying reliable biomarkers of aging is a major goal in geroscience. While the first generation of epigenetic biomarkers of aging were developed using chronological age as a surrogate for biological age, we hypothesized that incorporation of composite clinical measures of phenotypic age that capture differences in lifespan and healthspan may identify novel CpGs and facilitate the development of a more powerful epigenetic biomarker of aging. Using an innovative two-step process, we develop a new epigenetic biomarker of aging, DNAm PhenoAge, that strongly outperforms previous measures in regards to predictions for a variety of aging outcomes, including all-cause mortality, cancers, healthspan, physical functioning, and Alzheimer's disease. While this biomarker was developed using data from whole blood, it correlates strongly with age in every tissue and cell tested. Based on an in-depth transcriptional analysis in sorted cells, we find that increased epigenetic, relative to chronological age, is associated with increased activation of pro-inflammatory and interferon pathways, and decreased activation of transcriptional/translational machinery, DNA damage response, and mitochondrial signatures. Overall, this single epigenetic biomarker of aging is able to capture risks for an array of diverse outcomes across multiple tissues and cells, and provide insight into important pathways in aging.

Keywords: epigenetic clock; DNA methylation; biomarker; healthspan.

Conflict of interest statement

CONFLICTS OF INTEREST: The Regents of the University of California is the sole owner of a provisional patent application directed at this invention for which MEL, SH are named inventors.

Figures

Figure 1
Figure 1
Roadmap for developing DNAm PhenoAge. The roadmap depicts our analytical procedures. In step 1, we developed an estimate of ‘Phenotypic Age’ based on clinical measure. Phenotypic age was developed using the NHANES III as training data, in which we employed a proportional hazard penalized regression model to narrow 42 biomarkers to 9 biomarkers and chronological age. This measure was then validated in NHANES IV and shown to be a strong predictor of both morbidity and mortality risk. In step 2, we developed an epigenetic biomarker of phenotypic age, which we call DNAm PhenoAge, by regressing phenotypic age (from step 1) on blood DNA methylation data, using the InCHIANTI data. This produced an estimate of DNAm PhenoAge based on 513 CpGs. We then validated our new epigenetic biomarker of aging, DNAm PhenoAge, using multiple cohorts, aging-related outcomes, and tissues/cells. In step 3, we examined the underlying biology of the 513 CpGs and the composite DNAm PhenoAge measure, using a variety of complementary data (gene expression, blood cell counts) and various genome annotation tools including chromatin state analysis and gene ontology enrichment.
Figure 2
Figure 2
Mortality Prediction by DNAm PhenoAge. (A) Using five samples from large epidemiological cohorts—two samples from the Women’s health Initiative, the Framingham Heart Study, the Normative Aging Study, and the Jackson Heart Study—we tested whether DNAm PhenoAge was predictive of all-cause mortality. The Fig. displays a forest plot for fixed-effect meta-analysis, based on Cox proportional hazard models, and adjusting for chronological age. Results suggest that DNAm PhenoAge is predictive of mortality in all samples, and that overall, a one-year increase in DNAm PhenoAge is associated with a 4.5% increase in the risk of death (p=9.9E-47). This is contrasted against the first generation of epigenetic biomarkers of aging by Hannum and Horvath, which exhibit less significant associations with lifespan (p=1.7E-21 and p=4.5E-5, respectively). (B and C) Using the WHI sample 1, we plotted Kaplan-Meier survival estimates using actual data from the fastest versus the slowest agers (panel B). We also applied the equation from the proportional hazard model to predict remaining life expectancy and plotted predicted survival assuming a chronological age of 50 and a DNAm PhenoAge of either 40 (slow ager), 50 (average ager), or 60 (fast ager) (panel C). Median life expectancy at age 50 was predicted to be approximately 81 years for the fastest agers, 83.5 years for average agers, and 86 years for the slowest agers.
Figure 3
Figure 3
Chronological age versus DNAm PhenoAge in a variety of tissues and cells. Although DNAm PhenoAge was developed using methylation data from whole blood, it also tracks chronological age in a wide variety of tissues and cells. (A) The correlation across all tissues/cells we examined is r=0.71. (B-ZJ) report results in different sources of DNA as indicated in panel headings. The numbers correspond to the data sets from (Horvath 2013). Overall, correlations range from r=0.35 (breast, panel O) to r=0.92 (temporal cortex in brain, panel L).
Figure 4
Figure 4
DNAm PhenoAge measured in dorsolateral prefrontal cortex relates to Alzheimer’s disease and related neuropathologies. Using postmortem data from the Religious Order Study (ROS) and the Memory and Aging Project (MAP), we find a moderate/high correlation between chronological age and DNAm PhenoAge (panel A). We also estimate the Pproportion of neurons via the CETS algorithm and show that it correlates with DNAm PhenoAge (B). Further furtherthe correlation between chronological agen and DNAm PhenoAge is increased after adjusting for the estimated proportion on neurons in each sample (panel C). We also find that DNAm PhenoAge is significantly higher (p=0.00046) among those with Alzheimer’s disease versus controls (panel D), and that it positively correlates with amyloid load (p=0.012, panel E), neuritic plaques (p=0.0032, panel F), diffuse plaques (p=0.036, panel G), and neurofibrillary tangles (p=0.0073, panel H).
Figure 5
Figure 5
Lifestyle factors versus DNAm PhenoAge acceleration in blood in the WHI. In this cross- sectional analysis, the correlation test analysis (bicor, biweight midcorrelation) between select variables and DNAm PhenoAgeAccel reveals that education, income, exercise, proxies of fruit/vegetable consumption, and HDL cholesterol are negatively associated (blue) with DNAm PhenoAge, i.e. younger epigenetic age. Conversely, CRP, insulin, glucose, triglycerides, BMI, waist-to-hip ratio, systolic blood pressure, and smoking have a positive association (red) with DNAm PhenoAge. All results have been adjusted for ethnicity and batch/data set. Similar results based on multivariate regression models can be found in Supplement 1: Fig. S6B.

References

    1. Kennedy BK, Berger SL, Brunet A, Campisi J, Cuervo AM, Epel ES, Franceschi C, Lithgow GJ, Morimoto RI, Pessin JE, Rando TA, Richardson A, Schadt EE, et al.. Geroscience: linking aging to chronic disease. Cell. 2014; 159:709–13. 10.1016/j.cell.2014.10.039
    1. Burch JB, Augustine AD, Frieden LA, Hadley E, Howcroft TK, Johnson R, Khalsa PS, Kohanski RA, Li XL, Macchiarini F, Niederehe G, Oh YS, Pawlyk AC, et al.. Advances in geroscience: impact on healthspan and chronic disease. J Gerontol A Biol Sci Med Sci. 2014. (Suppl 1); 69:S1–3. 10.1093/gerona/glu041
    1. Fraga MF, Esteller M. Epigenetics and aging: the targets and the marks. Trends Genet. 2007; 23:413–18. 10.1016/j.tig.2007.05.008
    1. Rakyan VK, Down TA, Maslau S, Andrew T, Yang TP, Beyan H, Whittaker P, McCann OT, Finer S, Valdes AM, Leslie RD, Deloukas P, Spector TD. Human aging-associated DNA hypermethylation occurs preferentially at bivalent chromatin domains. Genome Res. 2010; 20:434–39. 10.1101/gr.103101.109
    1. Teschendorff AE, Menon U, Gentry-Maharaj A, Ramus SJ, Weisenberger DJ, Shen H, Campan M, Noushmehr H, Bell CG, Maxwell AP, Savage DA, Mueller-Holzner E, Marth C, et al.. Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res. 2010; 20:440–46. 10.1101/gr.103606.109
    1. Jung M, Pfeifer GP. Aging and DNA methylation. BMC Biol. 2015; 13:7. 10.1186/s12915-015-0118-4
    1. Zheng SC, Widschwendter M, Teschendorff AE. Epigenetic drift, epigenetic clocks and cancer risk. Epigenomics. 2016; 8:705–19. 10.2217/epi-2015-0017
    1. Bocklandt S, Lin W, Sehl ME, Sánchez FJ, Sinsheimer JS, Horvath S, Vilain E. Epigenetic predictor of age. PLoS One. 2011; 6:e14821. 10.1371/journal.pone.0014821
    1. Garagnani P, Bacalini MG, Pirazzini C, Gori D, Giuliani C, Mari D, Di Blasio AM, Gentilini D, Vitale G, Collino S, Rezzi S, Castellani G, Capri M, et al.. Methylation of ELOVL2 gene as a new epigenetic marker of age. Aging Cell. 2012; 11:1132–34. 10.1111/acel.12005
    1. Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, Klotzle B, Bibikova M, Fan JB, Gao Y, Deconde R, Chen M, Rajapakse I, et al.. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell. 2013; 49:359–67. 10.1016/j.molcel.2012.10.016
    1. Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013; 14:R115. 10.1186/gb-2013-14-10-r115
    1. Weidner CI, Lin Q, Koch CM, Eisele L, Beier F, Ziegler P, Bauerschlag DO, Jöckel KH, Erbel R, Mühleisen TW, Zenke M, Brümmendorf TH, Wagner W. Aging of blood can be tracked by DNA methylation changes at just three CpG sites. Genome Biol. 2014; 15:R24. 10.1186/gb-2014-15-2-r24
    1. Lin Q, Weidner CI, Costa IG, Marioni RE, Ferreira MR, Deary IJ, Wagner W. DNA methylation levels at individual age-associated CpG sites can be indicative for life expectancy. Aging (Albany NY). 2016; 8:394–401. 10.18632/aging.100908
    1. Horvath S, Erhart W, Brosch M, Ammerpohl O, von Schönfels W, Ahrens M, Heits N, Bell JT, Tsai PC, Spector TD, Deloukas P, Siebert R, Sipos B, et al.. Obesity accelerates epigenetic aging of human liver. Proc Natl Acad Sci USA. 2014; 111:15538–43. 10.1073/pnas.1412759111
    1. Marioni RE, Shah S, McRae AF, Chen BH, Colicino E, Harris SE, Gibson J, Henders AK, Redmond P, Cox SR, Pattie A, Corley J, Murphy L, et al.. DNA methylation age of blood predicts all-cause mortality in later life. Genome Biol. 2015; 16:25. 10.1186/s13059-015-0584-6
    1. Marioni RE, Shah S, McRae AF, Ritchie SJ, Muniz-Terrera G, Harris SE, Gibson J, Redmond P, Cox SR, Pattie A, Corley J, Taylor A, Murphy L, et al.. The epigenetic clock is correlated with physical and cognitive fitness in the Lothian Birth Cohort 1936. Int J Epidemiol. 2015; 44:1388–96. 10.1093/ije/dyu277
    1. Horvath S, Garagnani P, Bacalini MG, Pirazzini C, Salvioli S, Gentilini D, Di Blasio AM, Giuliani C, Tung S, Vinters HV, Franceschi C. Accelerated epigenetic aging in Down syndrome. Aging Cell. 2015; 14:491–95. 10.1111/acel.12325
    1. Horvath S, Levine AJ. HIV-1 Infection Accelerates Age According to the Epigenetic Clock. J Infect Dis. 2015; 212:1563–73. 10.1093/infdis/jiv277
    1. Horvath S, Pirazzini C, Bacalini MG, Gentilini D, Di Blasio AM, Delledonne M, Mari D, Arosio B, Monti D, Passarino G, De Rango F, D’Aquila P, Giuliani C, et al.. Decreased epigenetic age of PBMCs from Italian semi-supercentenarians and their offspring. Aging (Albany NY). 2015; 7:1159–70. 10.18632/aging.100861
    1. Levine ME, Hosgood HD, Chen B, Absher D, Assimes T, Horvath S. DNA methylation age of blood predicts future onset of lung cancer in the women’s health initiative. Aging (Albany NY). 2015; 7:690–700. 10.18632/aging.100809
    1. Levine ME, Lu AT, Bennett DA, Horvath S. Epigenetic age of the pre-frontal cortex is associated with neuritic plaques, amyloid load, and Alzheimer’s disease related cognitive functioning. Aging (Albany NY). 2015; 7:1198–211. 10.18632/aging.100864
    1. Levine ME, Lu AT, Chen BH, Hernandez DG, Singleton AB, Ferrucci L, Bandinelli S, Salfati E, Manson JE, Quach A, Kusters CD, Kuh D, Wong A, et al.. Menopause accelerates biological aging. Proc Natl Acad Sci USA. 2016; 113:9327–32. 10.1073/pnas.1604558113
    1. Chen BH, Marioni RE, Colicino E, Peters MJ, Ward-Caviness CK, Tsai PC, Roetker NS, Just AC, Demerath EW, Guan W, Bressler J, Fornage M, Studenski S, et al.. DNA methylation-based measures of biological age: meta-analysis predicting time to death. Aging (Albany NY). 2016; 8:1844–65. 10.18632/aging.101020
    1. Quach A, Levine ME, Tanaka T, Lu AT, Chen BH, Ferrucci L, Ritz B, Bandinelli S, Neuhouser ML, Beasley JM, Snetselaar L, Wallace RB, Tsao PS, et al.. Epigenetic clock analysis of diet, exercise, education, and lifestyle factors. Aging (Albany NY). 2017; 9:419–46. 10.18632/aging.101168
    1. Dugue PA, Bassett JK, Joo JE, Jung CH, Ming Wong E, Moreno-Betancur M, Schmidt D, Makalic E, Li S, Severi G, Hodge AM, Buchanan DD, English DR, et al.. DNA methylation-based biological aging and cancer risk and survival: pooled analysis of seven prospective studies. Int J Cancer. 2018; 142:1611–19. 10.1002/ijc.31189
    1. Simpkin AJ, Howe LD, Tilling K, Gaunt TR, Lyttleton O, McArdle WL, Ring SM, Horvath S, Smith GD, Relton CL. The epigenetic clock and physical development during childhood and adolescence: longitudinal analysis from a UK birth cohort. Int J Epidemiol. 2017; 46:549–58.10.1093/ije/dyw307
    1. Maierhofer A, Flunkert J, Oshima J, Martin GM, Haaf T, Horvath S. Accelerated epigenetic aging in Werner syndrome. Aging (Albany NY). 2017; 9:1143–52. 10.18632/aging.101217
    1. Levine ME. Modeling the rate of senescence: can estimated biological age predict mortality more accurately than chronological age? J Gerontol A Biol Sci Med Sci. 2013; 68:667–74. 10.1093/gerona/gls233
    1. Belsky DW, Caspi A, Houts R, Cohen HJ, Corcoran DL, Danese A, Harrington H, Israel S, Levine ME, Schaefer JD, Sugden K, Williams B, Yashin AI, et al.. Quantification of biological aging in young adults. Proc Natl Acad Sci USA. 2015; 112:E4104–10. 10.1073/pnas.1506264112
    1. Li S, Wong EM, Joo JE, Jung CH, Chung J, Apicella C, Stone J, Dite GS, Giles GG, Southey MC, Hopper JL. Genetic and Environmental Causes of Variation in the Difference Between Biological Age Based on DNA Methylation and Chronological Age for Middle-Aged Women. Twin Res Hum Genet. 2015; 18:720–26. 10.1017/thg.2015.75
    1. Sebastiani P, Thyagarajan B, Sun F, Schupf N, Newman AB, Montano M, Perls TT. Biomarker signatures of aging. Aging Cell. 2017; 16:329–38. 10.1111/acel.12557
    1. Ferrucci L, Hesdorffer C, Bandinelli S, Simonsick EM. Frailty as a Nexus Between the Biology of Aging, Environmental Conditions and Clinical Geriatrics. Public Health Rev. 2010; 32:475–88. 10.1007/BF03391612
    1. Christiansen L, Lenart A, Tan Q, Vaupel JW, Aviv A, McGue M, Christensen K. DNA methylation age is associated with mortality in a longitudinal Danish twin study. Aging Cell. 2016; 15:149–54. 10.1111/acel.12421
    1. Perna L, Zhang Y, Mons U, Holleczek B, Saum KU, Brenner H. Epigenetic age acceleration predicts cancer, cardiovascular, and all-cause mortality in a German case cohort. Clin Epigenetics. 2016; 8:64. 10.1186/s13148-016-0228-z
    1. Breitling LP, Yang R, Korn B, Burwinkel B, Brenner H. Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am J Hum Genet. 2011; 88:450–57. 10.1016/j.ajhg.2011.03.003
    1. Joehanes R, Just AC, Marioni RE, Pilling LC, Reynolds LM, Mandaviya PR, Guan W, Xu T, Elks CE, Aslibekyan S, Moreno-Macias H, Smith JA, Brody JA, et al.. Epigenetic Signatures of Cigarette Smoking. Circ Cardiovasc Genet. 2016; 9:436–47. 10.1161/CIRCGENETICS.116.001506
    1. Lee KW, Pausova Z. Cigarette smoking and DNA methylation. Front Genet. 2013; 4:132. 10.3389/fgene.2013.00132
    1. Zhang Y, Wilson R, Heiss J, Breitling LP, Saum KU, Schöttker B, Holleczek B, Waldenberger M, Peters A, Brenner H. DNA methylation signatures in peripheral blood strongly predict all-cause mortality. Nat Commun. 2017; 8:14617. 10.1038/ncomms14617
    1. Bennett DA, Schneider JA, Arvanitakis Z, Wilson RS. Overview and findings from the religious orders study. Curr Alzheimer Res. 2012; 9:628–45. 10.2174/156720512801322573
    1. Bennett DA, Schneider JA, Buchman AS, Barnes LL, Boyle PA, Wilson RS. Overview and findings from the rush Memory and Aging Project. Curr Alzheimer Res. 2012; 9:646–63. 10.2174/156720512801322663
    1. Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000; 908:244–54. 10.1111/j.1749-6632.2000.tb06651.x
    1. Horvath S, Zhang Y, Langfelder P, Kahn RS, Boks MP, van Eijk K, van den Berg LH, Ophoff RA. Aging effects on DNA methylation modules in human brain and blood tissue. Genome Biol. 2012; 13:R97. 10.1186/gb-2012-13-10-r97
    1. Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: a tool for genome-wide complex trait analysis. Am J Hum Genet. 2011; 88:76–82. 10.1016/j.ajhg.2010.11.011
    1. Sierra F, Kohanski R. Geroscience and the trans-NIH Geroscience Interest Group, GSIG. Geroscience. 2017; 39:1–5. 10.1007/s11357-016-9954-6
    1. Belsky DW, Moffitt TE, Cohen AA, Corcoran DL, Levine ME, Prinz JA, Schaefer J, Sugden K, Williams B, Poulton R, Caspi A. Eleven Telomere, Epigenetic Clock, and Biomarker-Composite Quantifications of Biological Aging: Do They Measure the Same Thing? Am J Epidemiol. 2017. 10.1093/aje/kwx346
    1. Kubben N, Misteli T. Shared molecular and cellular mechanisms of premature ageing and ageing-associated diseases. Nat Rev Mol Cell Biol. 2017; 18:595–609. 10.1038/nrm.2017.68
    1. Merlo A, Herman JG, Mao L, Lee DJ, Gabrielson E, Burger PC, Baylin SB, Sidransky D. 5′ CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers. Nat Med. 1995; 1:686–92. 10.1038/nm0795-686
    1. Finch CE. (2007). CHAPTER 1 - Inflammation and Oxidation in Aging and Chronic Diseases. The Biology of Human Longevity. (Burlington: Academic Press), pp. 1-112.
    1. Finch CE. Evolution in health and medicine Sackler colloquium: Evolution of the human lifespan and diseases of aging: roles of infection, inflammation, and nutrition. Proc Natl Acad Sci USA. 2010. (Suppl 1); 107:1718–24. 10.1073/pnas.0909606106
    1. Moiseeva O, Mallette FA, Mukhopadhyay UK, Moores A, Ferbeyre G. DNA damage signaling and p53-dependent senescence after prolonged beta-interferon stimulation. Mol Biol Cell. 2006; 17:1583–92. 10.1091/mbc.E05-09-0858
    1. Gonskikh Y, Polacek N. Alterations of the translation apparatus during aging and stress response. Mech Ageing Dev. 2017. (Suppl C); 168:30–36. 10.1016/j.mad.2017.04.003
    1. Wolters S, Schumacher B. Genome maintenance and transcription integrity in aging and disease. Front Genet. 2013; 4:19. 10.3389/fgene.2013.00019
    1. Steffen KK, Dillin A. A Ribosomal Perspective on Proteostasis and Aging. Cell Metab. 2016; 23:1004–12. 10.1016/j.cmet.2016.05.013
    1. Garinis GA, Uittenboogaard LM, Stachelscheid H, Fousteri M, van Ijcken W, Breit TM, van Steeg H, Mullenders LH, van der Horst GT, Brüning JC, Niessen CM, Hoeijmakers JH, Schumacher B. Persistent transcription-blocking DNA lesions trigger somatic growth attenuation associated with longevity. Nat Cell Biol. 2009; 11:604–15. 10.1038/ncb1866
    1. Schumacher B, Garinis GA, Hoeijmakers JH. Age to survive: DNA damage and aging. Trends Genet. 2008; 24:77–85. 10.1016/j.tig.2007.11.004
    1. McMurray CT, Vijg J. Editorial overview: Molecular and genetic bases of disease: the double life of DNA. Curr Opin Genet Dev. 2014; 26:v–vii. 10.1016/j.gde.2014.09.002
    1. Levine ME, Crimmins EM. A Genetic Network Associated With Stress Resistance, Longevity, and Cancer in Humans. J Gerontol A Biol Sci Med Sci. 2016; 71:703–12. 10.1093/gerona/glv141
    1. Zhou KI, Pincus Z, Slack FJ. Longevity and stress in Caenorhabditis elegans. Aging (Albany NY). 2011; 3:733–53. 10.18632/aging.100367
    1. Lin YJ, Seroude L, Benzer S. Extended life-span and stress resistance in the Drosophila mutant methuselah. Science. 1998; 282:943–46. 10.1126/science.282.5390.943
    1. Yu BP, Chung HY. Stress resistance by caloric restriction for longevity. Ann N Y Acad Sci. 2001; 928:39–47. 10.1111/j.1749-6632.2001.tb05633.x
    1. Martin GM, Austad SN, Johnson TE. Genetic analysis of ageing: role of oxidative damage and environmental stresses. Nat Genet. 1996; 13:25–34. 10.1038/ng0596-25
    1. Houseman EA, Accomando WP, Koestler DC, Christensen BC, Marsit CJ, Nelson HH, Wiencke JK, Kelsey KT. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics. 2012; 13:86. 10.1186/1471-2105-13-86
    1. Horvath S, Gurven M, Levine ME, Trumble BC, Kaplan H, Allayee H, Ritz BR, Chen B, Lu AT, Rickabaugh TM, Jamieson BD, Sun D, Li S, et al.. An epigenetic clock analysis of race/ethnicity, sex, and coronary heart disease. Genome Biol. 2016; 17:171. 10.1186/s13059-016-1030-0

Source: PubMed

3
購読する