Skip to main content

MINI REVIEW article

Front. Bioinform., 02 June 2022
Sec. Genomic Analysis
Volume 2 - 2022 | https://doi.org/10.3389/fbinf.2022.847629

DNA Methylation, Aging, and Cancer Risk: A Mini-Review

www.frontiersin.orgLarry Chen1 www.frontiersin.orgPatricia A. Ganz2,3 www.frontiersin.orgMary E. Sehl2,4*
  • 1Computational and Systems Biology Program, University of California, Los Angeles, Los Angeles, CA, United States
  • 2Division of Hematology-Oncology, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, United States
  • 3Department of Health Policy and Management, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
  • 4Department of Computational Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, United States

Accumulation of somatic mutations and genomic instability are hallmarks of both aging and cancer. Epigenetic alterations occur across cell types and tissues with advancing age. DNA methylation-based estimates of biologic age can predict important age-related outcomes, including risk of frailty and mortality, and most recently have been shown to be associated with risk of developing cancer. In this mini-review, we examine pathways known to exhibit altered methylation in aging tissues, pre-malignant lesions, and tumors and review methodologies of epigenetic clocks that reliably predict cancer risk, including those derived from methylation studies of peripheral blood, as well as those methylation levels from within the tissues at high risk of cancer.

Introduction

Cancer incidence increases exponentially with advancing age, beginning at the midpoint of the lifespan in most mammalian species (Campisi and Yaswen, 20092009). Somatic mutations accumulate within cells with chronic cell cycling (Moskalev et al., 2013), leading to genomic instability, a hallmark of both aging and cancer (Hanahan and Weinberg, 2011; López-Otín et al., 2013). Over the past decade, epigenetic alterations that occur with advancing age across cell types and tissues have been identified (Teschendorff et al., 2010; Horvath, 2013), and methylation markers at select sites have been shown to reliably predict chronologic age (Teschendorff et al., 2010; Bocklandt et al., 2011; Hannum et al., 2013; Horvath, 2013). Epigenetic clocks have been further shown to predict age-related diseases and outcomes, including frailty and mortality, suggesting that they are reliable markers of biologic aging (Chen et al., 2016; Levine et al., 2018; Lu et al., 2019a). Importantly, methylation age has recently also been associated with cancer risk (Levine et al., 2015; Lu et al., 2019a; Yu et al., 2020). In this article, we will review studies of global methylation alterations that occur with advancing age and cancer risk, compare the development and features of first- and second-generation epigenetic clocks, as well as the epigenetic pacemaker clock and other methods, and illustrate their ability to predict risk of incident cancer.

Pathways With Aberrant Methylation in Malignant Tissues

DNA methylation is thought to play an important role in the etiology of complex traits, including cancer (Esteller, 2008; Petronis, 2010). The importance of DNA methylation in carcinogenesis was recognized with the discovery of numerous hypermethylated promoters of tumor suppressor genes in tumor samples, as well as findings confirming the role of DNA methylation in facilitating DNA damage, e.g., in the silencing of mismatch repair genes (Jones and Laird, 1999). Hundreds of genes, including key tumor suppressor genes, are hypermethylated at promoter CpG islands, and are either transcriptionally silenced or blocked from normal induction, in nearly every patient’s cancer compared with normal cell counterparts (Vaissière et al., 2009; Baylin and Jones, 2016; Xie et al., 2018). The similarity in epigenetic alterations that occur during tumorigenesis and senescence raises the question of whether programmatic changes that occur during senescence play a role in carcinogenesis. Though the promoter hypermethylation events in malignant transformation appear to arise independently of cellular senescence (Xie et al., 2018), further exploration is needed to identify a relationship between cancer risk and epigenetic events occurring in development and aging.

Age-Dependent Hypermethylation of Polycomb Group Target Proteins

Polycomb group proteins repress genes required for stem cell differentiation, and targets of polycomb group proteins (PCGTs) are repressed in human embryonic and adult stem cells through reversible chromatin modifications (Lee et al., 2006). PCGTs are 12-fold more likely to be methylated in cancer tissues than non-PCGTs, suggesting a mechanism of carcinogenesis where cells are locked in an un-differentiated state of self-renewal and predisposed to malignant transformation. In an analysis of whole blood samples from 261 postmenopausal women, Teschendorff demonstrated that PCGT CpGs are hypermethylated with advancing age, and this methylation signature was validated in seven independent data sets encompassing 900 samples, from multiple cell types and tissues including blood, ovarian cancer, cervix, and marrow mesenchymal stem and stromal cells (Teschendorff et al., 2010).

Variability in Age-Related Methylation Patterns in Premalignant Lesions

Methylation markers drift differentially with age between normal and premalignant tissues. In pre-malignant dysplastic tissues, age-PCGT CpGs were more highly methylated than in normal samples, suggesting that age may contribute to carcinogenesis by irreversibly silencing genes that are suppressed in stem cells (Teschendorff et al., 2010). Importantly, in dysplastic tissues, differential variability in methylation identifies cancer risk markers more reliably than differences in mean methylation (Teschendorff and Widschwendter, 2012). Differentially variable features identified in precursor non-invasive lesions exhibit significantly increased enrichment for developmental genes compared with differentially methylated sites (Teschendorff and Widschwendter, 2012). In studies of normal and pre-malignant esophageal tissues, differential methylomic drift occurs in Barrett’s esophagus (BE) relative to normal squamous tissue (Curtius et al., 2016). Using a Bayesian model incorporating longitudinal methylomic drift rates, patient age, and methylation data from BE and normal squamous tissue, Curtius et al. have developed a molecular clock to reliably estimate patient-specific BE onset times, providing information about how long an individual has lived with the precursor lesion (Curtius et al., 2016).

DNA Methylation-Based Estimates of Age

Table 1 summarizes 16 epigenetic clocks that have been developed over the past decade. These clocks reliably estimate chronologic age based on methylation levels at select CpGs (Di Lena et al., 2021). In an early work analyzing methylation patterns in saliva associated with advancing age, lasso penalized regression was used to screen for the top predictors of age, and a leave-one-out regression analysis was used to form an accurate epigenetic predictor of age (Bocklandt et al., 2011). Subsequently, Horvath developed a multi-tissue epigenetic clock across 51 healthy tissues and cell types, that reliably estimated methylation age across cell types and tissues (Horvath, 2013). This epigenetic clock was found to be close to zero for embryonic and induced pluripotent stem cells, applied across species to chimpanzees (Horvath, 2013), and importantly was later found to be accelerated in disease states (Horvath et al., 2014; Rickabaugh et al., 2015) and predictive of frailty and mortality (Chen et al., 2016). This clock utilizes elastic net regression on a transformed continuous, monotonically increasing function of age to select a set of CpGs whose weighted average reliably predict age across a wide spectrum of tissues and cell types. Elastic net regression linearly combines the l1 (lasso) and l2 (ridge) penalty terms. While the lasso tends to select only one variable from a group of highly correlated variables, the quadratic expression elevates the loss function toward being convex, allowing a larger number of variables to be included when there is a higher correlation of variables and higher grouping effect. To avoid low efficiency in predictability and high bias from subjecting coefficients to two types of shrinkages, coefficients are rescaled by multiplying them by (1 + l2). The Hannum clock similarly uses elastic net regression to estimate chronologic age from methylation levels at 71 CpGs in peripheral blood (Hannum et al., 2013). Blood-based epigenetic age measures Intrinsic and Extrinsic epigenetic age incorporate information on cell composition (imputed from methylation data at additional sites) to estimate age. While Intrinsic epigenetic age is independent of changes in cell distribution that occur with advancing age, Extrinsic epigenetic age is positively and negatively correlated with estimated proportions of naïve and senescent cytotoxic T lymphocytes. More recently, second generation clocks have been developed, including Phenotypic age (Levine et al., 2018) and Grim age (Lu et al., 2019a) which are both more closely associated with lifespan, and utilize a two-step process to estimate biologic age. In the development of the Phenotypic age clock, first 1) Cox penalized regression is used to identify a set of biomarkers that best predict aging-related mortality, and next 2) a mortality score based on the regression coefficients from step 1 is converted into units of years and the resultant phenotypic age estimate is regressed on DNA methylation using an elastic net regression analysis. Grim age clock also involves a two-step process in which 1) methylation data is used to estimate smoking pack-years and levels of plasma proteins known to be associated with morbidity or mortality, and 2) time-to-death is regressed on these DNA methylation-based surrogate biomarkers, resulting in a mortality risk estimate that is transformed into units of years. The Skin and Blood clock is a robust estimator of methylation age in fibroblasts, keratinocytes, buccal cells, endothelial cells, lymphoblastoid cells, skin, blood, and saliva, and was developed when it was found that other clocks performed poorly in human fibroblasts and other skin cells (Horvath et al., 2018). A DNA methylation-based estimate of telomere length, DNAmTL, is a measure of cell replicative history, and outperforms measured leukocyte telomere length in predicting time to death and age-related pathologies (Lu et al., 2019b). Stubbs et al. developed an accurate multi-tissue age estimator in mice, with CpGs from pathways involved in morphogenesis and development (Stubbs et al., 2017). Additional clocks have been developed to more accurately predict age in human cortical tissue (Shireby et al., 2020), skeletal muscle (Voisin et al., 2020), and pediatric buccal epithelial (McEwen et al., 2020) tissues. Finally, epigenetic clocks have been developed to predict gestation age using methylation levels of cells from umbilical cord and placental tissues (Bohlin et al., 2016; Knight et al., 2016; Mayne et al., 2017; Lee et al., 2019).

TABLE 1
www.frontiersin.org

TABLE 1. Features of epigenetic clocks.

Despite high accuracy, epigenetic clocks do not permit characterization of the non-linear epigenetic aging patterns that occur across the entire lifespan. Recently, a new method was developed to model epigenetic changes with age while accounting for the nonlinearities of this relationship that occur with advancing age (Snir et al., 2019). This integrated framework, based on evolutionary models, addresses the acceleration and deceleration of epigenetic changes that occur over time, and has been applied to methylation data from broad age ranges and multiple tissue types. The Epigenetic Pacemaker (EPM) employs a fast conditional expectation maximization algorithm to model epigenetic states associated with a phenotype of interest, such as aging and type 2 diabetes mellitus. In this algorithm, each methylation site is assigned an independent rate of change and starting methylation value, while each individual is assigned an epigenetic state. Given i methylation sites and j individuals, a single methylation site can be described as:

mij^ = mi0+ risj+ij

Where mij^ is the observed methylation value, mi0 is the initial methylation value, ri is the rate of change, sj is the epigenetic state, and ij is a normally distributed error term. The goal of the EPM is to find the optimal values of the initial methylation value, rate of change, and epigenetic state to minimize the error between the predicted and observed methylation values across a system of methylation sites. The epigenetic state is then updated through each iteration of the EPM to minimize the error across the observed epigenetic landscape. Because the epigenetic state is updated while fitting the EPM, the assumption of linearity between the methylation values and the phenotypic trait of interest is relaxed. In addition to examining age as an outcome of interest, these models can be employed to study additional phenotypes of interest, including risk of cancer.

Epigenetic Clocks Predicting Risk of Cancer

Older tissues are at greater risk of malignant transformation because of acquired mutations that occur in the setting of prolonged epithelial proliferation. Several recent studies have demonstrated that accelerated aging in peripheral blood predicts subsequent development of cancer (Levine et al., 2015; Kresovich et al., 2019a; Zheng et al., 2016; Perna et al., 2016; Durso et al., 2017; Kresovich et al., 2019b). Table 2 summarizes epigenetic clocks that have demonstrated association with cancer risk. Pan-tissue clock acceleration in peripheral blood is associated with later development of lung cancer (Levine et al., 2015), breast cancer (Durso et al., 2017), and male colon cancer (Durso et al., 2017). Grim age, a strong predictor of mortality, is associated with time to any cancer (Lu et al., 2019a). Intrinsic epigenetic age acceleration in peripheral blood is associated with risk of post-menopausal breast cancer, with epigenetic acceleration detected up to 10 years prior to cancer diagnosis (Ambatipudi et al., 2017). In a large study examining methylation in 2,764 cancer free women in the Sister Study, 1,566 of whom subsequently developed breast cancer after an average of 6 years, acceleration of Pan-tissue age, Hannum age, and Phenotypic age each predicted risk of subsequent breast cancer (Kresovich et al., 2019b). In this study, Grim age was associated with invasive breast cancer in post-menopausal women (Kresovich et al., 2019a). Using data from seven nested case-control studies of peripheral blood DNA methylation and colorectal, gastric, kidney, lung, prostate, and urothelial cancer, and B cell lymphoma from the Melbourne Collaborative Cohort Study, epigenetic aging was associated with both risk of cancer and increased risk of death after cancer diagnosis (Dugué et al., 2018). A five-year increase in age acceleration was associated with a 4–9% increase in risk of cancer, and a 2–5% increased risk of death following cancer diagnosis (Dugué et al., 2018).

TABLE 2
www.frontiersin.org

TABLE 2. Epigenetic clocks predictive of cancer risk.

In addition to the associations found between cancer risk and epigenetic aging in peripheral blood, several studies have examined age-related epigenetic changes in tissues that subsequently develop cancer. Epigenetic aging is associated with cancer risk (in at-risk tissues) and prognosis (in cancerous tissues). For example, Pan-tissue age acceleration in colon cancer samples has been linked with colon cancer molecular subtypes and improved prognosis prediction because it is linked with overall survival (Zheng et al., 2019). In breast tumor samples, methylation studies within the breast of very young women with more aggressive breast cancer exhibit accelerated DNA methylation age compared with breast cancer in older counterparts, suggesting a role of accelerated epigenetic aging in breast cancer risk (Oltra et al., 2019). In addition, methylation-based markers of cell replication have been associated with cancer risk, including the epigenetic mitotic clock (EpiTOC) which approximates the rate of stem cell division in normal tissues by focusing on promoter CpG sites that localize to PCGT genes, and has been shown to be accelerated in precancerous lesions and cancer (Yang et al., 2016). Furthermore, Youn et al. demonstrated that quantitative estimates of mitotic age (total number of cell divisions) of a tissue, derived using the stochastic replication errors accumulated during cell divisions predict shorter disease associated survival in thirteen cancer types studied (Youn and Wang, 2018). In healthy breast tissue, methylation of tumor suppressor genes APC (Lewis et al., 2005; Euhus et al., 2008) and RASSF1 (Lewis et al., 2005) is associated with breast cancer risk as measured by the Gail model risk score. In a recent study comparing disease-free breast tissue cores from women at high versus average risk for breast cancer using the Tyrer-Cuzick model, 1698 DNA methylation aberrations were identified in high-risk breast tissues, from pathways involving cell adhesion, ErbB, and protein kinase A signaling (Marino et al., 2021). A global study of age-related DNA methylation changes in healthy breast demonstrated that increased methylation primarily occurs at enhancer regions of binding sites for chromatin remodeling genes (Johnson et al., 2017). Epigenetic age of healthy breast is elevated above chronologic age and appears older with other tissues in the body (Horvath, 2013; Sehl et al., 2017), including matched peripheral blood samples from healthy breast tissue donors (Sehl et al., 2017). Estrogen stimulation and chronic cell cycling are thought to drive accelerated aging in breast tissue (Pike et al., 1983; Pike et al., 1993). Risk factors for breast cancer that relate to lifetime estrogen exposure, including earlier menarche and elevated body mass index, are associated with accelerated Grim age in healthy breast tissue (Sehl et al., 2021). Likewise Pan-tissue age, Hannum age, and Phenotypic age in peripheral blood are associated with risk factors for breast cancer, including BMI and alcohol use (Chen et al., 2019). Furthermore, in peripheral blood, an epigenome wide analysis of estimated lifetime estrogen exposure (ELEE) in 216 women in the EPIC-Italy study identified a methylation index of ELEE based on 694 CpGs, and developed a methylation index based on 31 of these most varying CpGs that predicted subsequent breast cancer risk in 440 women with incident breast cancer and 440 controls from the Generations Study (Johansson et al., 2019). An increase of DNA methylationbased ELEE of 1 year was associated with a 5% increase in breast cancer risk (Johansson et al., 2019).

DNA methylation studies comparing normal adjacent breast tissue from women with breast cancer and healthy tissues from cancer-free women revealed epigenetic field effects, with aberrant methylation in specific pathways related to stem cell differentiation, including WNT signaling, known to be epigenetically deregulated in cancer (Teschendorff et al., 2016). Furthermore, epigenetic age in normal adjacent breast tissue from luminal breast cancer patients is increased compared with healthy breast tissue from donors with no history of breast cancer (Hofstatter et al., 2018). In a recent study of 107 breast tumor samples compared with 45 paired adjacent-normal breast tissue samples and 459 normal breast samples, DNA methylation age was estimated using 286 CpGs out of over two million candidate CpGs. Breast tumor samples exhibited age acceleration, appeared 12 and 13 years older than adjacent normal and normal breast tissue with identified pathways involving cellular development and morphology, epidermal growth factor and estrogen receptor signaling (Castle et al., 2020).

Finally, a recent study of epigenetic age-related methylation changes in healthy mammary epithelial tissues demonstrated accelerated epigenetic aging in 12 women with germline mutations in cancer susceptibility genes (Miyano et al., 2021). This study used a breast-specific molecular clock based on methylation of ELF5, a marker critical for mammary development. This finding suggests a link between inherited alterations in DNA repair capacity and accelerated epigenetic aging in tissues at highest risk of developing malignancy.

Conclusion and Future Directions

DNA methylation-based estimates of biologic age are associated with both cancer risk factors and risk of incident cancer, suggesting a potential mechanistic link between genomic instability, epigenetic age acceleration, and carcinogenesis. Further work is needed to investigate alterations in transcriptomic and proteomic pathways that accompany epigenetic age acceleration prior to the development of cancer. Identification of these changes could lead to targets for chemoprevention in individuals at high risk for cancer. In addition, future studies should identify nonlinear trends in epigenetic age that are associated with cancer risk and modeling epigenetic states that are associated with risk of cancer. Integrative analyses of methylation age along with genomic, transcriptomic, and proteomic data within an individual prior to the development of cancer may ultimately be used to develop predictive tools that could be used to guide risk reduction strategies.

Author Contributions

All authors contributed to the conceptualization, review of the literature, and writing of the manuscript. All authors have approved the completed version of the manuscript and are accountable for all aspects of the work.

Funding

This study was funded by a Susan G. Komen Career Catalyst Award for Basic and Translational Science CCR16380478. PAG was supported by the Breast Cancer Research Foundation (BCRF). PAG severs on the scientific advisory board for the BCRF.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Ambatipudi, S., Horvath, S., Perrier, F., Cuenin, C., Hernandez-Vargas, H., Le Calvez-Kelm, F., et al. (2017). DNA Methylome Analysis Identifies Accelerated Epigenetic Ageing Associated with Postmenopausal Breast Cancer Susceptibility. Eur. J. Cancer 75, 299–307. doi:10.1016/j.ejca.2017.01.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Baylin, S. B., and Jones, P. A. (2016). Epigenetic Determinants of Cancer. Cold Spring Harb Perspect. Biol. 8 (9). doi:10.1101/cshperspect.a019505

PubMed Abstract | CrossRef Full Text | Google Scholar

Bocklandt, S., Lin, W., Sehl, M. E., Sánchez, F. J., Sinsheimer, J. S., Horvath, S., et al. (2011). Epigenetic Predictor of Age. PLoS One 6 (6), e14821. doi:10.1371/journal.pone.0014821

PubMed Abstract | CrossRef Full Text | Google Scholar

Bohlin, J., Håberg, S. E., Magnus, P., Reese, S. E., Gjessing, H. K., Magnus, M. C., et al. (2016). Prediction of Gestational Age Based on Genome-wide Differentially Methylated Regions. Genome Biol. 17 (1), 207. doi:10.1186/s13059-016-1063-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Campisi, J., and Yaswen, P. (20092009). Aging and Cancer Cell Biology, 2009. Aging Cell 8 (3), 221–225. doi:10.1111/j.1474-9726.2009.00475.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Castle, J. R., Lin, N., Liu, J., Storniolo, A. M. V., Shendre, A., Hou, L., et al. (2020). Estimating Breast Tissue-specific DNA Methylation Age Using Next-Generation Sequencing Data. Clin. Epigenetics 12 (1), 45. doi:10.1186/s13148-020-00834-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, B. H., Marioni, R. E., Colicino, E., Peters, M. J., Ward-Caviness, C. K., Tsai, P. C., et al. (2016). DNA Methylation-Based Measures of Biological Age: Meta-Analysis Predicting Time to Death. Aging (Albany NY) 8 (9), 1844–1865. doi:10.18632/aging.101020

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, M., Wong, E. M., Nguyen, T. L., Dite, G. S., Stone, J., Dugué, P. A., et al. (2019). DNA Methylation-Based Biological Age, Genome-wide Average DNA Methylation, and Conventional Breast Cancer Risk Factors. Sci. Rep. 9 (1), 15055. doi:10.1038/s41598-019-51475-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Curtius, K., Wong, C. J., Hazelton, W. D., Kaz, A. M., Chak, A., Willis, J. E., et al. (2016). A Molecular Clock Infers Heterogeneous Tissue Age Among Patients with Barrett's Esophagus. Plos Comput. Biol. 12 (5), e1004919. doi:10.1371/journal.pcbi.1004919

PubMed Abstract | CrossRef Full Text | Google Scholar

Di Lena, P., Sala, C., and Nardini, C. (2021). Estimage: a Webserver Hub for the Computation of Methylation Age. Nucleic Acids Res. 49 (W1), W199–W206. doi:10.1093/nar/gkab426

PubMed Abstract | CrossRef Full Text | Google Scholar

Dugué, P. A., Bassett, J. K., Joo, J. E., Jung, C. H., Ming Wong, E., Moreno-Betancur, M., et al. (2018). DNA Methylation-Based Biological Aging and Cancer Risk and Survival: Pooled Analysis of Seven Prospective Studies. Int. J. Cancer 142 (8), 1611–1619. doi:10.1002/ijc.31189

PubMed Abstract | CrossRef Full Text | Google Scholar

Durso, D. F., Bacalini, M. G., Sala, C., Pirazzini, C., Marasco, E., Bonafé, M., et al. (2017). Acceleration of Leukocytes' Epigenetic Age as an Early Tumor and Sex-specific Marker of Breast and Colorectal Cancer. Oncotarget 8 (14), 23237–23245. doi:10.18632/oncotarget.15573

PubMed Abstract | CrossRef Full Text | Google Scholar

Esteller, M. (2008). Epigenetics in Cancer. N. Engl. J. Med. 358 (11), 1148–1159. doi:10.1056/NEJMra072067

PubMed Abstract | CrossRef Full Text | Google Scholar

Euhus, D. M., Bu, D., Milchgrub, S., Xie, X. J., Bian, A., Leitch, A. M., et al. (2008). DNA Methylation in Benign Breast Epithelium in Relation to Age and Breast Cancer Risk. Cancer Epidemiol. Biomarkers Prev. 17 (5), 1051–1059. doi:10.1158/1055-9965.EPI-07-2582

PubMed Abstract | CrossRef Full Text | Google Scholar

Hanahan, D., and Weinberg, R. A. (2011). Hallmarks of Cancer: the Next Generation. Cell 144 (5), 646–674. doi:10.1016/j.cell.2011.02.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Hannum, G., Guinney, J., Zhao, L., Zhang, L., Hughes, G., Sadda, S., et al. (2013). Genome-wide Methylation Profiles Reveal Quantitative Views of Human Aging Rates. Mol. Cel 49 (2), 359–367. doi:10.1016/j.molcel.2012.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Hofstatter, E. W., Horvath, S., Dalela, D., Gupta, P., Chagpar, A. B., Wali, V. B., et al. (2018). Increased Epigenetic Age in normal Breast Tissue from Luminal Breast Cancer Patients. Clin. Epigenetics 10 (1), 112. doi:10.1186/s13148-018-0534-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Horvath, S. (2013). DNA Methylation Age of Human Tissues and Cell Types. Genome Biol. 14 (10), R115. doi:10.1186/gb-2013-14-10-r115

PubMed Abstract | CrossRef Full Text | Google Scholar

Horvath, S., Erhart, W., Brosch, M., Ammerpohl, O., von Schönfels, W., Ahrens, M., et al. (2014). Obesity Accelerates Epigenetic Aging of Human Liver. Proc. Natl. Acad. Sci. U S A. 111 (43), 15538–15543. doi:10.1073/pnas.1412759111

PubMed Abstract | CrossRef Full Text | Google Scholar

Horvath, S., Oshima, J., Martin, G. M., Lu, A. T., Quach, A., Cohen, H., et al. (2018). Epigenetic Clock for Skin and Blood Cells Applied to Hutchinson Gilford Progeria Syndrome and Ex Vivo Studies. Aging (Albany NY) 10 (7), 1758–1775. doi:10.18632/aging.101508

PubMed Abstract | CrossRef Full Text | Google Scholar

Johansson, A., Palli, D., Masala, G., Grioni, S., Agnoli, C., Tumino, R., et al. (2019). Epigenome-wide Association Study for Lifetime Estrogen Exposure Identifies an Epigenetic Signature Associated with Breast Cancer Risk. Clin. Epigenetics 11 (1), 66. doi:10.1186/s13148-019-0664-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnson, K. C., Houseman, E. A., King, J. E., and Christensen, B. C. (2017). Normal Breast Tissue DNA Methylation Differences at Regulatory Elements Are Associated with the Cancer Risk Factor Age. Breast Cancer Res. 19 (1), 81. doi:10.1186/s13058-017-0873-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Jones, P. A., and Laird, P. W. (1999). Cancer Epigenetics Comes of Age. Nat. Genet. 21 (2), 163–167. doi:10.1038/5947

PubMed Abstract | CrossRef Full Text | Google Scholar

Knight, A. K., Craig, J. M., Theda, C., Baekvad-Hansen, M., Bybjerg-Grauholm, J., Hansen, C. S., et al. (2016). An Epigenetic Clock for Gestational Age at Birth Based on Blood Methylation Data. Genome Biol. 17 (1), 206. doi:10.1186/s13059-016-1068-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Kresovich, J. K., Xu, Z., O'Brien, K. M., Weinberg, C. R., Sandler, D. P., and Taylor, J. A. (2019). Epigenetic Mortality Predictors and Incidence of Breast Cancer. Aging (Albany NY) 11 (24), 11975–11987. doi:10.18632/aging.102523

PubMed Abstract | CrossRef Full Text | Google Scholar

Kresovich, J. K., Xu, Z., O'Brien, K. M., Weinberg, C. R., Sandler, D. P., and Taylor, J. A. (2019). Methylation-Based Biological Age and Breast Cancer Risk. J. Natl. Cancer Inst. 111 (10), 1051–1058. doi:10.1093/jnci/djz020

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, T. I., Jenner, R. G., Boyer, L. A., Guenther, M. G., Levine, S. S., Kumar, R. M., et al. (2006). Control of Developmental Regulators by Polycomb in Human Embryonic Stem Cells. Cell 125 (2), 301–313. doi:10.1016/j.cell.2006.02.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, Y., Choufani, S., Weksberg, R., Wilson, S. L., Yuan, V., Burt, A., et al. (2019). Placental Epigenetic Clocks: Estimating Gestational Age Using Placental DNA Methylation Levels. Aging (Albany NY) 11 (12), 4238–4253. doi:10.18632/aging.102049

PubMed Abstract | CrossRef Full Text | Google Scholar

Levine, M. E., Hosgood, H. D., Chen, B., Absher, D., Assimes, T., and Horvath, S. (2015). DNA Methylation Age of Blood Predicts Future Onset of Lung Cancer in the Women's Health Initiative. Aging (Albany NY) 7 (9), 690–700. doi:10.18632/aging.100809

PubMed Abstract | CrossRef Full Text | Google Scholar

Levine, M. E., Lu, A. T., Quach, A., Chen, B. H., Assimes, T. L., Bandinelli, S., et al. (2018). An Epigenetic Biomarker of Aging for Lifespan and Healthspan. Aging (Albany NY) 10 (4), 573–591. doi:10.18632/aging.101414

PubMed Abstract | CrossRef Full Text | Google Scholar

Lewis, C. M., Cler, L. R., Bu, D. W., Zöchbauer-Müller, S., Milchgrub, S., Naftalis, E. Z., et al. (2005). Promoter Hypermethylation in Benign Breast Epithelium in Relation to Predicted Breast Cancer Risk. Clin. Cancer Res. 11 (1), 166–172.

PubMed Abstract | Google Scholar

López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M., and Kroemer, G. (2013). The Hallmarks of Aging. Cell 153 (6), 1194–1217. doi:10.1016/j.cell.2013.05.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, A. T., Quach, A., Wilson, J. G., Reiner, A. P., Aviv, A., Raj, K., et al. (2019). DNA Methylation GrimAge Strongly Predicts Lifespan and Healthspan. Aging (Albany NY) 11 (2), 303–327. doi:10.18632/aging.101684

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, A. T., Seeboth, A., Tsai, P. C., Sun, D., Quach, A., Reiner, A. P., et al. (2019). DNA Methylation-Based Estimator of Telomere Length. Aging (Albany NY) 11 (16), 5895–5923. doi:10.18632/aging.102173

PubMed Abstract | CrossRef Full Text | Google Scholar

Marino, N., German, R., Podicheti, R., Rush, D. B., Rockey, P., Huang, J., et al. (2021). Aberrant Epigenetic and Transcriptional Events Associated with Breast Cancer Risk. bioRxiv 1, 1. doi:10.1101/2021.09.14.460320

CrossRef Full Text | Google Scholar

Mayne, B. T., Leemaqz, S. Y., Smith, A. K., Breen, J., Roberts, C. T., and Bianco-Miotto, T. (2017). Accelerated Placental Aging in Early Onset Preeclampsia Pregnancies Identified by DNA Methylation. Epigenomics 9 (3), 279–289. doi:10.2217/epi-2016-0103

PubMed Abstract | CrossRef Full Text | Google Scholar

McEwen, L. M., O'Donnell, K. J., McGill, M. G., Edgar, R. D., Jones, M. J., MacIsaac, J. L., et al. (2020). The PedBE Clock Accurately Estimates DNA Methylation Age in Pediatric Buccal Cells. Proc. Natl. Acad. Sci. U S A. 117 (38), 23329–23335. doi:10.1073/pnas.1820843116

PubMed Abstract | CrossRef Full Text | Google Scholar

Miyano, M., Sayaman, R. W., Shalabi, S. F., Senapati, P., Lopez, J. C., Angarola, B. L., et al. (2021). Breast-Specific Molecular Clocks Comprised of ELF5 Expression and Promoter Methylation Identify Individuals Susceptible to Cancer Initiation. Cancer Prev. Res. (Phila) 14 (8), 779–794. doi:10.1158/1940-6207.CAPR-20-0635

PubMed Abstract | CrossRef Full Text | Google Scholar

Moskalev, A. A., Shaposhnikov, M. V., Plyusnina, E. N., Zhavoronkov, A., Budovsky, A., Yanai, H., et al. (2013). The Role of DNA Damage and Repair in Aging through the Prism of Koch-like Criteria. Ageing Res. Rev. 12 (2), 661–684. doi:10.1016/j.arr.2012.02.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Oltra, S. S., Peña-Chilet, M., Flower, K., Martinez, M. T., Alonso, E., Burgues, O., et al. (2019). Acceleration in the DNA Methylation Age in Breast Cancer Tumours from Very Young Women. Sci. Rep. 9 (1), 14991. doi:10.1038/s41598-019-51457-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Perna, L., Zhang, Y., Mons, U., Holleczek, B., Saum, K. U., and Brenner, H. (2016). Epigenetic Age Acceleration Predicts Cancer, Cardiovascular, and All-Cause Mortality in a German Case Cohort. Clin. Epigenetics 8, 64. doi:10.1186/s13148-016-0228-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Petronis, A. (2010). Epigenetics as a Unifying Principle in the Aetiology of Complex Traits and Diseases. Nature 465 (7299), 721–727. doi:10.1038/nature09230

PubMed Abstract | CrossRef Full Text | Google Scholar

Pike, M. C., Krailo, M. D., Henderson, B. E., and Casagrande, J. T., (1983). 'Hormonal' Risk Factors, 'breast Tissue Age' and the Age-Incidence of Breast Cancer. Nature 303 (5920), 767–770. doi:10.1038/303767a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Pike, M. C., Spicer, D. V., Dahmoush, L., and Press, M. F. (1993). Estrogens, Progestogens, normal Breast Cell Proliferation, and Breast Cancer Risk. Epidemiol. Rev. 15 (1), 17–35. doi:10.1093/oxfordjournals.epirev.a036102

PubMed Abstract | CrossRef Full Text | Google Scholar

Rickabaugh, T. M., Baxter, R. M., Sehl, M., Sinsheimer, J. S., Hultin, P. M., Hultin, L. E., et al. (2015). Acceleration of Age-Associated Methylation Patterns in HIV-1-Infected Adults. PLoS One 10 (3), e0119201. doi:10.1371/journal.pone.0119201

PubMed Abstract | CrossRef Full Text | Google Scholar

Sehl, M. E., Henry, J. E., Storniolo, A. M., Ganz, P. A., and Horvath, S. (2017). DNA Methylation Age Is Elevated in Breast Tissue of Healthy Women. Breast Cancer Res. Treat. 164 (1), 209–219. doi:10.1007/s10549-017-4218-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Sehl, M. E., Henry, J. E., Storniolo, A. M., Horvath, S., and Ganz, P. A. (2021). The Effects of Lifetime Estrogen Exposure on Breast Epigenetic Age. Cancer Epidemiol. Biomarkers Prev. 30, 1241–1249. doi:10.1158/1055-9965.EPI-20-1297

PubMed Abstract | CrossRef Full Text | Google Scholar

Shireby, G. L., Davies, J. P., Francis, P. T., Burrage, J., Walker, E. M., Neilson, G. W. A., et al. (2020). Recalibrating the Epigenetic Clock: Implications for Assessing Biological Age in the Human Cortex. Brain 143 (12), 3763–3775. doi:10.1093/brain/awaa334

PubMed Abstract | CrossRef Full Text | Google Scholar

Snir, S., Farrell, C., and Pellegrini, M. (2019). Human Epigenetic Ageing Is Logarithmic with Time across the Entire Lifespan. Epigenetics 14 (9), 912–926. doi:10.1080/15592294.2019.1623634

PubMed Abstract | CrossRef Full Text | Google Scholar

Stubbs, T. M., Bonder, M. J., Stark, A. K., Krueger, F., Team, B. I. A. C., von Meyenn, F., et al. (2017). Multi-tissue DNA Methylation Age Predictor in Mouse. Genome Biol. 18 (1), 68. doi:10.1186/s13059-017-1203-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Teschendorff, A. E., Gao, Y., Jones, A., Ruebner, M., Beckmann, M. W., Wachter, D. L., et al. (2016). DNA Methylation Outliers in normal Breast Tissue Identify Field Defects that Are Enriched in Cancer. Nat. Commun. 7, 10478. doi:10.1038/ncomms10478

PubMed Abstract | CrossRef Full Text | Google Scholar

Teschendorff, A. E., Menon, U., Gentry-Maharaj, A., Ramus, S. J., Weisenberger, D. J., Shen, H., et al. (2010). Age-dependent DNA Methylation of Genes that Are Suppressed in Stem Cells Is a Hallmark of Cancer. Genome Res. 20 (4), 440–446. doi:10.1101/gr.103606.109

PubMed Abstract | CrossRef Full Text | Google Scholar

Teschendorff, A. E., and Widschwendter, M. (2012). Differential Variability Improves the Identification of Cancer Risk Markers in DNA Methylation Studies Profiling Precursor Cancer Lesions. Bioinformatics 28 (11), 1487–1494. doi:10.1093/bioinformatics/bts170

PubMed Abstract | CrossRef Full Text | Google Scholar

Vaissière, T., Hung, R. J., Zaridze, D., Moukeria, A., Cuenin, C., Fasolo, V., et al. (2009). Quantitative Analysis of DNA Methylation Profiles in Lung Cancer Identifies Aberrant DNA Methylation of Specific Genes and its Association with Gender and Cancer Risk Factors. Cancer Res. 69 (1), 243–252. doi:10.1158/0008-5472.CAN-08-2489

PubMed Abstract | CrossRef Full Text | Google Scholar

Voisin, S., Harvey, N. R., Haupt, L. M., Griffiths, L. R., Ashton, K. J., Coffey, V. G., et al. (2020). An Epigenetic Clock for Human Skeletal Muscle. J. Cachexia Sarcopenia Muscle 11 (4), 887–898. doi:10.1002/jcsm.12556

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie, W., Kagiampakis, I., Pan, L., Zhang, Y. W., Murphy, L., Tao, Y., et al. (2018). DNA Methylation Patterns Separate Senescence from Transformation Potential and Indicate Cancer Risk. Cancer Cell 33 (2), 309–e5. doi:10.1016/j.ccell.2018.01.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Z., Wong, A., Kuh, D., Paul, D. S., Rakyan, V. K., Leslie, R. D., et al. (2016). Correlation of an Epigenetic Mitotic Clock with Cancer Risk. Genome Biol. 17 (1), 205. doi:10.1186/s13059-016-1064-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Youn, A., and Wang, S. (2018). The MiAge Calculator: a DNA Methylation-Based Mitotic Age Calculator of Human Tissue Types. Epigenetics 13 (2), 192–206. doi:10.1080/15592294.2017.1389361

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, M., Hazelton, W. D., Luebeck, G. E., and Grady, W. M. (2020). Epigenetic Aging: More Than Just a Clock when it Comes to Cancer. Cancer Res. 80 (3), 367–374. doi:10.1158/0008-5472.CAN19-0924

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, C., Li, L., and Xu, R. (2019). Association of Epigenetic Clock with Consensus Molecular Subtypes and Overall Survival of Colorectal Cancer. Cancer Epidemiol. Biomarkers Prev. 28 (10), 1720–1724. doi:10.1158/1055-9965.EPI-19-0208

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, Y., Joyce, B. T., Colicino, E., Liu, L., Zhang, W., Dai, Q., et al. (2016). Blood Epigenetic Age May Predict Cancer Incidence and Mortality. EBioMedicine 5, 68–73. doi:10.1016/j.ebiom.2016.02.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: aging, epigenetic clocks, DNA methylation, carcinogenesis, cancer risk

Citation: Chen L, Ganz PA and Sehl ME (2022) DNA Methylation, Aging, and Cancer Risk: A Mini-Review. Front. Bioinform. 2:847629. doi: 10.3389/fbinf.2022.847629

Received: 03 January 2022; Accepted: 21 March 2022;
Published: 02 June 2022.

Edited by:

Christine Nardini, National Research Council (CNR), Italy

Reviewed by:

Claudia Sala, University of Bologna, Italy

Copyright © 2022 Chen, Ganz and Sehl. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Mary E. Sehl, msehl@mednet.ucla.edu

Download