Fact-checked by Grok 2 weeks ago

Epigenetic clock

An epigenetic clock is a type of molecular biomarker that estimates an individual's biological age by analyzing patterns of DNA methylation at specific cytosine-phosphate-guanine (CpG) sites across the genome, providing a measure distinct from chronological age. These clocks leverage the fact that DNA methylation, an epigenetic modification that does not alter the underlying DNA sequence, accumulates predictably over time in various tissues and cell types, reflecting cumulative environmental and lifestyle influences on aging. First developed in 2013 by Steve Horvath, the seminal multi-tissue epigenetic clock uses a weighted average of methylation levels at 353 CpG sites, derived from elastic net regression on data from over 8,000 samples across 51 tissues, achieving a median prediction error of 3.6 years and a correlation of 0.96 with chronological age. Subsequent generations of epigenetic clocks have expanded on this foundation, incorporating tissue-specific models like the skin and blood clock for improved accuracy and applications, as well as second-generation clocks that predict outcomes beyond , such as mortality risk and disease susceptibility. For instance, clocks such as the PhenoAge and GrimAge integrate data with clinical biomarkers to assess accelerated aging linked to lifestyle factors, with applications in predicting age-related conditions like , cancer, and neurodegeneration. In , epigenetic clocks reveal significant age acceleration, averaging 36 years across 20 tumor types, highlighting their utility in studying tumorigenesis and treatment effects. Interventions like caloric restriction or drugs such as rapamycin have been shown to slow the ticking of these clocks, suggesting potential for evaluating anti-aging therapies. Despite their precision, epigenetic clocks face limitations, including a strong with cell-type composition in tissues, which can confound interpretations, and underrepresentation of non-European ancestry populations in training data, potentially reducing generalizability. Recent reviews emphasize the need for diverse cohorts and validation in longitudinal studies to enhance predictive power for complex diseases. As of 2024, ongoing developments focus on integrating multi-omics data and to refine clocks for , underscoring their role as dynamic tools in aging .

Fundamentals

Definition and Measurement

An epigenetic clock is an algorithm that estimates an individual's biological age based on patterns of at specific cytosine-phosphate-guanine (CpG) sites across the , offering a molecular measure that can diverge from chronological age to reflect physiological aging processes. These clocks leverage the fact that , an epigenetic modification involving the addition of a to cytosine bases, accumulates in predictable patterns with age, serving as a for cellular and tissue . Measurement of DNA methylation for epigenetic clocks typically employs array-based technologies, such as the Illumina Infinium HumanMethylation450 (450K) BeadChip or the more comprehensive MethylationEPIC (EPIC) BeadChip, which interrogate hundreds of thousands of CpG sites simultaneously. These arrays quantify methylation levels as beta (β) values, ranging from 0 (completely unmethylated) to 1 (fully methylated), derived from the ratio of methylated to total probe intensities after normalization for technical artifacts. A seminal example is Horvath's 2013 pan-tissue clock, which demonstrates broad applicability across 51 tissue and cell types by relying on methylation data from just 353 CpG sites, enabling robust age predictions independent of tissue origin. The core computation of epigenetic age, denoted as DNAm age, involves a of these β values: \text{DNAm age} = \sum_{i=1}^{n} w_i \cdot \beta_i + c where w_i are regression coefficients (weights) for each selected i, \beta_i is the level at that site, n is the number of sites (e.g., 353 for Horvath's clock), and c is an intercept term, with weights obtained via elastic net trained on large datasets associating methylation patterns with chronological . Epigenetic clocks have evolved across generations, reflecting refinements in their design and scope. First-generation clocks, like the Horvath pan-tissue model, focus on direct age estimation from methylation alone. Second-generation variants, such as DNAm PhenoAge, integrate phenotypic clinical biomarkers (e.g., , ) into the model alongside data to better predict morbidity, mortality, and healthspan. Third-generation clocks, including DunedinPACE, shift emphasis to the rate of aging by modeling longitudinal changes in to quantify the pace of epigenetic drift rather than absolute age.

Biological Basis in DNA Methylation

DNA methylation is a key epigenetic modification involving the addition of a methyl group to the fifth carbon of cytosine residues, primarily in CpG dinucleotides, catalyzed by DNA methyltransferases (DNMTs). The primary enzymes responsible are DNMT1, which maintains methylation patterns during DNA replication; and DNMT3A and DNMT3B, which establish de novo methylation. Hypermethylation at promoter regions typically leads to gene silencing by inhibiting transcription factor binding and recruiting repressive chromatin complexes, while hypomethylation generally promotes gene activation by facilitating access to transcriptional machinery. With advancing age, landscapes undergo characteristic alterations, including global hypomethylation across the , which may contribute to genomic instability, and focal hypermethylation at specific loci, particularly promoters of polycomb group target genes involved in developmental regulation and cell fate. These changes exhibit , where levels at individual CpG sites deviate progressively from youthful patterns due to accumulated errors in and environmental influences, independent of genetic . Epigenetic drift, first demonstrated in monozygotic twins where differences in increase with age despite identical , reflects this non-heritable accumulation of variations, potentially amplifying over time and contributing to age-related phenotypic divergence. Several regulators modulate these age-associated methylation dynamics. TET enzymes (TET1, TET2, TET3) mediate active by oxidizing to and further intermediates, facilitating removal of methyl marks and influencing plasticity during aging. modifications interplay with , as trimethylation of at 27 (H3K27me3) by polycomb repressive complexes often correlates with hypermethylated promoters in aged cells, reinforcing transcriptional repression. Non-coding RNAs, including long non-coding RNAs (lncRNAs) and microRNAs, further shape methylation landscapes by recruiting DNMTs to target loci or interfering with TET activity, thereby linking environmental cues to epigenetic aging processes. These interconnected mechanisms underscore the dynamic of as a molecular basis for tracking biological age.

History and Development

Early Discoveries in Epigenetics and Aging

The concept of an epigenetic landscape, introduced by Conrad Hal Waddington in the 1940s, provided an early framework for understanding how stable cellular states are maintained through development and potentially altered over time, laying groundwork for later interpretations of epigenetic stability in aging contexts. Waddington's model visualized gene expression as a ball rolling down branching valleys shaped by genetic and environmental influences, emphasizing the role of epigenetic mechanisms in canalizing cell fates without altering the DNA sequence. This idea, initially applied to embryogenesis, was later revived to explain how accumulated epigenetic modifications might contribute to the progressive restriction of cellular plasticity observed in aging somatic cells. In the 1970s and , pioneering studies revealed age-associated changes in patterns, particularly hypomethylation, in tissues and cultured cells, suggesting that epigenetic drift could serve as a molecular record of cellular divisions. Holliday and Pugh's 1975 proposal posited that site-specific acts as a heritable cellular , functioning as a "mitotic clock" to track somatic cell divisions and potentially limit replicative lifespan, akin to a developmental timer. Subsequent work in , such as analyses of liver and brain tissues, confirmed progressive global hypomethylation with age, linking these changes to reduced gene regulation fidelity and . In human studies, early observations in immortalized cell lines and fibroblasts demonstrated similar methylation erosion with passage number, mirroring aging and highlighting as a potential of proliferative history before the year 2000. These findings extended to connections with other aging hallmarks, including attrition, where preliminary evidence suggested that patterns at telomeric regions might influence shortening rates in human cells. The 2000s marked a shift toward predictive models, with Bocklandt et al.'s 2011 twin study identifying six CpG sites in saliva DNA whose methylation levels strongly correlated with chronological age, enabling accurate predictions with a median error of 5.2 years and demonstrating heritability in monozygotic pairs. This work extended to sperm samples, revealing tissue-specific epigenetic aging signatures that foreshadowed broader applications. Concurrently, Hannum et al. in 2013 developed an early single-tissue clock using blood methylation data from over 650 samples, selecting 71 CpG sites to estimate age with high precision (median error of 4.9 years), influenced by factors like and . These breakthroughs built on prior observations by quantifying age-related epigenetic variance, though they remained confined to or reproductive tissues. Early epigenetic research faced significant hurdles, including reliance on limited tissue types like or , which restricted generalizability across the body, and small sizes that hampered statistical robustness. Additionally, the absence of integrated multi-omics approaches meant that interactions between , modifications, and environmental factors were underexplored, complicating causal inferences about aging. These limitations spurred later efforts to develop pan-tissue predictors.

Establishment of Multi-Tissue Clocks

The establishment of multi-tissue epigenetic clocks marked a significant advancement in aging research, enabling the prediction of biological age across diverse human tissues and cell types. In 2013, Steve Horvath developed the first pan-tissue epigenetic clock using elastic net regression on data from over 8,000 samples spanning 51 tissues and nucleated cell types, selecting 353 CpG sites that demonstrated robustness and high predictive accuracy for chronological age (r ≈ 0.96 in training data). This clock highlighted the universality of age-related methylation patterns, distinguishing it from prior tissue-specific predictors by allowing comparisons of epigenetic age across organs like , , and liver. Following Horvath's breakthrough, several specialized clocks emerged to refine predictions in particular contexts while maintaining multi-tissue applicability. That same year, Gregory Hannum and colleagues introduced a blood-based clock trained on profiles from 482 individuals aged 19 to 101, utilizing 71 CpG sites to estimate age with comparable accuracy (r = 0.91) but optimized for samples. In 2018, Morgan Levine extended this framework with PhenoAge, an epigenetic predictor derived from elastic net on data calibrated to a phenotypic aging index incorporating nine clinical biomarkers such as and glucose levels, enhancing its correlation with health outcomes beyond chronological age alone (r = 0.93 for mortality prediction). A year later, Qian Zhang and team improved precision by retraining an elastic net model on a larger of over 12,000 and samples, yielding a 514-CpG clock that reduced median absolute error to 2.31 years across multiple tissues including breast and liver. Advancements from 2020 to 2025 focused on integrating mortality risk, longitudinal dynamics, and advanced computational techniques to create more prognostic multi-tissue clocks. The GrimAge clock, introduced in 2019 by Ake T. Lu and Horvath but refined through 2022, predicts time-to-death by surrogating plasma proteins (e.g., PAI-1) and pack-years via at 1,030 CpG sites, outperforming prior clocks in mortality forecasting ( = 1.10 per year acceleration). In 2022, Daniel W. Belsky and colleagues developed DunedinPACE from longitudinal data on 1,000 , using 846 CpG sites to measure the pace of aging rather than absolute age, revealing accelerated rates in childhood adversity-exposed individuals (r = 0.92 for pace estimation). AI-enhanced models gained traction, exemplified by a 2023 deep learning-based clock that achieved single-site-level predictions with minimal CpG markers (30 sites), attaining superior accuracy (median error = 2.1 years) in sample validations through optimization. Key milestones underscored the transition to clinical utility. In 2024, EpiMedTech's EPIAGE became the first epigenetic age test registered with the FDA, facilitating standardized multi-tissue assessments in routine health monitoring. Concurrently, integration with enabled analysis of epigenetic heterogeneity, as demonstrated by the 2024 scEpiAge model (developed in models), which predicts age at cellular resolution in tissues like blood and liver (r = 0.6 in single cells). In 2025, further developments included a blood-based epigenetic clock for intrinsic capacity that predicts mortality and associates with clinical and lifestyle factors.

Construction and Properties

Statistical and Computational Methods

The construction of epigenetic clocks primarily relies on elastic net penalized , a technique that integrates L1 () and L2 () penalties to simultaneously select informative CpG sites from high-dimensional data and fit coefficients for age prediction. This approach addresses in genome-wide datasets by shrinking less relevant coefficients to zero while stabilizing estimates for correlated predictors. The is formulated as: \min_{\beta} \left\| y - X\beta \right\|_2^2 + \lambda \left( \alpha \left\| \beta \right\|_1 + (1 - \alpha) \left\| \beta \right\|_2^2 \right) where y represents the vector of chronological ages from samples, X is the matrix of values at CpG sites, \beta denotes the of model coefficients, \lambda > 0 is the regularization tuned via cross-validation, and $0 \leq \alpha \leq 1 balances the penalty types (with \alpha = 1 yielding pure ). Training protocols emphasize robust cross-validation on large, heterogeneous cohorts to ensure generalizability, often incorporating from over 20 distinct tissues or types to derive pan-tissue predictors. Batch effects arising from experimental variations in are mitigated through techniques, such as the SWAN method, which performs subset-quantile adjustments within arrays to harmonize type I and type II probe signals on Illumina platforms. In the 2020s, extensions beyond have incorporated algorithms like random forests for handling non-linear age-methylation associations and neural networks for improved predictive accuracy in complex datasets. , common in arrays due to probe failures, is addressed via imputation methods such as methyLImp, which employs linear regression on neighboring CpG sites to estimate absent values without introducing substantial bias. Pace-of-aging clocks further compute the aging rate as the derivative of the epigenetic age trajectory with respect to chronological time, derived from longitudinal models to quantify temporal dynamics rather than absolute age. Computational tools facilitate implementation and application; the ENmix provides end-to-end preprocessing, including background correction and normalization, alongside functions for clock estimation. In , methylprep offers streamlined loading and normalization of Illumina array data for subsequent analysis. Web-based platforms, such as the DNAm Age Calculator, enable direct computation of epigenetic age from user-submitted profiles using established models.

Accuracy, Calibration, and Limitations

Epigenetic clocks exhibit high accuracy in estimating chronological age, with the seminal Horvath clock achieving a median absolute error of 3.6 years and a of r=0.96 across a diverse set of tissues and types in its validation dataset. This performance varies by , with r≈0.95 and certain regions like r≈0.98, reflecting subtle differences in patterns and sample heterogeneity. Such metrics underscore the clocks' utility as reliable biomarkers, though accuracy diminishes slightly in non-optimal tissues due to inherent biological variability. Calibration of epigenetic clocks involves universal scaling parameters to facilitate multi-tissue applicability, as pioneered in the Horvath model, which avoids tissue-specific transformations by leveraging shared sites across 51 tissue types. In blood-based applications, where cell-type composition can confound predictions, reference-based methods like the EpiDISH tool adjust for proportions of immune cell subtypes using predefined reference profiles, thereby enhancing calibration robustness. These approaches ensure consistent performance across datasets but require careful preprocessing to mitigate compositional biases. Despite their strengths, epigenetic clocks face notable limitations, including ethnic biases stemming from training on predominantly European-ancestry datasets, leading to systematic age underestimation by 2-5 years in African-ancestry populations. They are also sensitive to technical artifacts, such as batch effects in array processing, which can inflate error rates by introducing non-biological variance. Fundamentally, clocks capture correlative epigenetic shifts without distinguishing causal aging mechanisms from mere associations, limiting mechanistic insights. Post-2020 studies have highlighted challenges in rate-based clocks, such as DunedinPACE, including inconsistencies across diverse populations and potential limitations in generalizability. Recent advancements from 2023 to 2025 have addressed these issues through recalibrations incorporating diverse global datasets, exemplified by multi-ancestry models that integrate and other non-European samples to reduce es and improve predictive equity. For example, a 2025 study integrated and European-ancestry data to develop clocks with reduced , achieving r>0.95 across groups. These updated clocks demonstrate enhanced accuracy (r>0.95) across ancestries while maintaining computational efficiency for broad research applications.

Comparisons with Other Aging Biomarkers

Epigenetic clocks, which estimate biological through patterns, offer distinct advantages over length as an aging . length inversely correlates with chronological but exhibits high inter-individual variability due to genetic, , and measurement factors. In contrast, epigenetic clocks demonstrate greater stability and superior for mortality; for instance, in epigenetic is associated with a (HR) of approximately 1.10 per year deviation, compared to 1.05 for shortening. This outperformance is evident in large cohorts, where epigenetic measures independently predict all-cause mortality beyond length. Proteomic clocks, such as the one developed by Lehallier et al. using a panel of 373 proteins, achieve similar accuracy to epigenetic clocks in estimating biological age, with strong correlations to chronological age (r ≈ 0.93). However, these clocks are inherently tissue-limited, relying on sampling that may not reflect organ-specific aging processes. Epigenetic clocks excel in multi-tissue applicability, allowing non-invasive assessment from diverse samples like or buccal swabs, which broadens their utility in studies. Other biomarkers include glycan clocks, which gauge age via immunoglobulin G (IgG) N-glycan profiles and show moderate correlation with chronological age (r ≈ 0.74). Metabolomic clocks, often derived from nuclear magnetic resonance (NMR) spectroscopy of blood metabolites, are sensitive to dietary factors like polyunsaturated fatty acid levels but yield lower age correlations (r ≈ 0.29) and are more prone to environmental fluctuations. Composite indices, such as the frailty index, integrate clinical phenotypes like grip strength and comorbidities to estimate biological age but lack the molecular precision of epigenetic measures. Epigenetic clocks capture cumulative environmental exposures more effectively than alternatives; for example, the GrimAge clock provides about 20% stronger mortality prediction than telomere length in prospective studies. Yet, they offer less direct mechanistic insight into aging drivers compared to genomic clocks based on somatic mutation burden, which quantify DNA damage accumulation and link to cellular senescence pathways. Recent advancements by 2025 include hybrid clocks integrating with AI-derived multi-omics data, such as and , which outperform single-modality clocks by approximately 15% in predicting aging trajectories within longitudinal cohorts. These models enhance overall accuracy while addressing limitations in isolated biomarkers.

Applications in Research

Genetic and Lifestyle Influences

Epigenetic age acceleration exhibits moderate , with estimates ranging from approximately 20% to 40% depending on the specific clock measure and population studied. For instance, - and SNP-based analyses of intrinsic epigenetic age acceleration in blood data from large cohorts have yielded heritability values around 28%. Genome-wide association studies (GWAS) have identified genetic variants influencing epigenetic aging rates, including loci near genes involved in and maintenance, such as TERT, which explain a portion of the observed variance. Polygenic scores derived from these GWAS hits typically account for about 5% of the variance in epigenetic age acceleration, highlighting the polygenic nature of genetic contributions while underscoring the substantial role of non-genetic factors. Lifestyle factors, particularly modifiable behaviors, significantly influence the rate of epigenetic aging. is a potent accelerator, with each additional associated with approximately 0.09 to 0.17 years of GrimAge acceleration, translating to roughly 1-1.7 years per decade of one pack-per-day . In contrast, regular physical exercise is linked to decelerated epigenetic aging, with meta-analyses and cohort studies showing that higher activity levels correlate with 0.5 to 1 year slower aging across multiple clocks, potentially through reduced and improved metabolic function. Adherence to a , rich in polyphenols and anti-inflammatory foods, has been shown to reduce epigenetic age by about 1 to 2 years in intervention studies, as observed in women following the diet for one year. Environmental exposures also modulate epigenetic clocks, often accelerating aging in a dose-dependent manner. Long-term to fine particulate matter (PM2.5) is associated with epigenetic age acceleration, where an increase of about 1 μg/m³ in ambient PM2.5 corresponds to 0.3 to 0.4 years faster extrinsic epigenetic aging, potentially amounting to 0.8 years or more in urban settings with moderate pollution levels. consumption shows a biphasic effect: moderate intake appears neutral or minimally impactful on clocks like GrimAge, while heavy or chronic use accelerates aging by 1.4 to 2.2 years, as evidenced in individuals with alcohol use compared to controls. Observational data on interventions further illustrate the of epigenetic clocks in response to changes. In animal models, caloric restriction by 20-40% has been shown to slow epigenetic aging by 10-20%, delaying drift and preserving youthful patterns in tissues like and muscle across including mice and rhesus monkeys. Preliminary trials, such as those involving metformin, suggest metformin users exhibit epigenetic ages 2.8 to 3.4 years younger than non-users according to Horvath and Hannum clocks, though results vary by clock and population. These findings emphasize the potential for modifications to mitigate genetic predispositions toward accelerated epigenetic aging.

Disease Associations and Diagnostics

Epigenetic clocks have demonstrated significant associations with cardiovascular diseases, particularly through accelerated aging patterns observed in affected individuals. In patients with confirmed , a hallmark of , epigenetic age acceleration averages 2.5 years higher compared to those with normal angiograms, as measured by age acceleration (DNAmAA). This acceleration serves as a predictive for (MI), where each 5-year increment in epigenetic age is linked to a (HR) of 1.12 for incident MI using the Hannum clock, independent of chronological age and traditional risk factors. In metabolic disorders, epigenetic clocks reveal pronounced aging acceleration tied to disease pathology. Individuals with (T2D) exhibit approximately 2 to 3 years of epigenetic age advancement, reflecting disruptions in glucose metabolism and insulin signaling that alter patterns. Similarly, is associated with a 1.8-year acceleration in epigenetic aging, mediated by low-grade that promotes pro-aging epigenetic modifications in metabolic tissues like the liver and adipose. Cancer presents tissue-specific epigenetic clock accelerations that underscore their diagnostic potential. For instance, tissues show substantial accelerated epigenetic aging relative to adjacent normal tissue, consistent with averages of ~36 years across tumor types driven by aberrant in tumor suppressor genes and oncogenic pathways. This property extends to non-invasive applications, where epigenetic clocks integrated into liquid biopsies—analyzing —achieve up to 85% sensitivity for early cancer detection as of 2023, outperforming some genetic markers in specificity for high-risk populations. Infectious diseases also accelerate epigenetic clocks, highlighting their role in post-infection morbidity. HIV infection is linked to 5-7 years of epigenetic age advancement, particularly in untreated or early-stage cases, due to persistent immune activation and viral-induced methylation changes in immune cells. Recent 2024 studies on post-COVID-19 long-haulers report modest acceleration in epigenetic aging, associated with lingering inflammatory responses and multi-organ sequelae in survivors. Beyond specific diseases, epigenetic clocks offer superior prognostic utility for overall health outcomes. The GrimAge clock, which incorporates methylation surrogates for plasma proteins and , outperforms traditional clinical risk scores in predicting all-cause mortality, with a C-index of approximately 0.81 compared to lower values for other models, enabling refined stratification across diverse cohorts. Ongoing as of 2025 explores incorporating clock-based metrics into for personalized diagnostics and monitoring. As of 2025, developments focus on integrating multi-omics data and to refine applications.

Therapeutic and Rejuvenation Interventions

Epigenetic clocks serve as valuable biomarkers for assessing the impact of therapeutic interventions designed to modulate biological aging. These tools enable researchers to quantify changes in epigenetic age following treatments, providing objective measures of beyond chronological time. Experimental applications span pharmacological agents, cellular therapies, and genetic , with outcomes demonstrating partial reversal or deceleration of epigenetic aging in preclinical and early clinical settings. Histone deacetylase (HDAC) inhibitors, such as vorinostat, have been investigated for their capacity to reverse epigenetic clocks by altering histone modifications and DNA methylation patterns. A 2024 human clinical trial in elderly participants further showed that vorinostat treatment slowed the pace of epigenetic aging by approximately 0.8 years, suggesting translational promise for age-related decline mitigation. Stem cell-based therapies have also demonstrated rejuvenating effects measurable by epigenetic clocks. Heterochronic parabiosis, where the circulatory systems of young and old mice are joined, reversed the blood epigenetic clock by about 2 years in aged animals according to a 2021 mouse study, attributing this to circulating factors from young blood that remodel the epigenome. In humans, hematopoietic stem cell transplantation has shown rejuvenation, with one analysis indicating a 1.5-year decrease in epigenetic age post-transplant due to the infusion of younger donor cells into older recipients. Partial reprogramming using Yamanaka factors, particularly the OSKM genes (Oct4, , Klf4, and c-Myc), offers a targeted approach to reset epigenetic clocks without full to pluripotency. A 2023 study in human fibroblasts reported a 3-5 year reversal in epigenetic age following transient OSKM expression, accompanied by improved cellular function and reduced markers. However, this method carries risks of tumorigenesis due to oncogenic potential of the factors, necessitating controlled, partial application protocols. Emerging therapies as of 2025 continue to leverage epigenetic clocks for validation. Senolytics, including the combination of and , have slowed the epigenetic aging pace by around 10% in preclinical models by selectively eliminating senescent cells that accumulate age-related epigenetic noise. Similarly, CRISPR-based gene editing targeting DNA methyltransferases (DNMTs) in organoids has shown promise in restoring youthful patterns, with early 2025 reports indicating potential for tissue-specific without off-target genomic alterations. The integration of epigenetic clocks into therapeutic trials raises important ethical considerations, particularly regarding their use as primary endpoints for anti-aging interventions. These biomarkers must be validated for clinical relevance to avoid misleading outcomes, ensuring that apparent reversals correlate with healthspan improvements rather than transient changes. Regulatory frameworks, such as the Union's 2025 guidelines on advanced medicinal products, emphasize rigorous assessments for epigenome-modifying , including long-term of clock dynamics to support approvals.

Specific Biological Insights

Tissue-Specific Variations

Epigenetic clocks developed for pan-tissue use demonstrate high accuracy across diverse mammalian tissues, achieving correlations of ≈ 0.96–0.98 with chronological using fewer than 1,000 CpG sites. However, these models often deviate when applied to specific tissues, such as the , where they systematically underestimate epigenetic by approximately 4 years, particularly in older individuals. In contrast, tissue-specific clocks tailored to or yield marginally higher precision, with correlations up to ≈ 0.987 in . For eye tissue, methylation at the ELOVL2 locus enables hyper-precise age prediction, with models achieving correlations as high as = 0.99, reflecting its role in age-related ocular changes. Certain tissues exhibit slower epigenetic aging relative to chronological age. The exhibits negative epigenetic age acceleration (appearing biologically younger) compared to other regions, a pattern observed in centenarians and attributed to region-specific saturation that resists further age-related shifts. Heart muscle shows relatively stable epigenetic profiles across adulthood with minimal deviation in pan-tissue models. Conversely, tissue displays accelerated epigenetic aging, estimating on average 11.4 years (SD 7.1 years) older than matched blood samples, an effect that diminishes with chronological age and is reduced post-menopause. Intra-tissue heterogeneity arises from cell-type composition, with distinct epigenetic clocks revealing differential aging rates within organs. In the , neuron-specific clocks indicate slower aging compared to glia-specific models, where is more pronounced, highlighting glial vulnerability in aging processes. Validation studies must account for post-mortem artifacts, such as altered in that can produce negative age values, potentially confounding clock accuracy. Recent advances in single-nucleus epigenetic clocks, developed in 2024, uncover fine-scale intra-tissue variations, such as zonation patterns in the liver where subsets exhibit distinct age accelerations. These clocks enable precise assessment of organ viability, including in liver transplants, by quantifying biological age at cellular resolution to predict post-transplant outcomes.

Effects in Rare Conditions and Longevity

In characterized by premature aging, epigenetic clocks reveal substantial accelerations that align with clinical manifestations of accelerated biological aging. In Hutchinson-Gilford progeria syndrome (HGPS), a and epigenetic clock detects an average age acceleration of approximately 5 years in patient-derived fibroblasts from children under 10 years old, independent of cell culture effects. This acceleration is evident in both classic and non-classic HGPS cases, underscoring the clock's sensitivity to lamin A mutations driving nuclear instability. Similarly, , caused by defects in , exhibits a marked epigenetic age acceleration of about 15.5 years in fibroblasts using the skin and blood clock, accompanied by widespread hypomethylation at repetitive elements like Alu sequences. Down syndrome, resulting from trisomy 21, demonstrates accelerated epigenetic aging particularly in neural tissues, with clocks estimating an average increase of 6.6 years in both and compared to euploid controls. This acceleration correlates with genome-wide DNA hypomethylation, which contributes to overexpression of genes such as amyloid precursor protein (), exacerbating Alzheimer's-like pathology in affected brains. In , a neurodegenerative disorder, epigenetic clocks indicate an overall brain age acceleration of 3.2 years, with multivariate models confirming this effect across regions including the frontal and parietal lobes, though striatum-specific changes may vary by disease stage. Other conditions highlight tissue-specific or reversible epigenetic shifts. , marking ovarian aging, is associated with epigenetic age acceleration in blood-derived clocks among postmenopausal women, reflecting broader reproductive aging impacts. In infection, clocks show accelerations of 5.2 years in blood and 7.4 years in tissue during untreated phases, effects that partially reverse with antiretroviral (ART), decelerating epigenetic aging over time under viral suppression. Werner syndrome, another segmental progeroid disorder due to WRN mutations, features accelerated epigenetic aging in blood cells independent of cell composition changes, with recent analyses revealing early-onset signatures distinct from normal aging patterns. A 2025 study on atypical variants highlights faster progression and earlier epigenetic deviations, supporting segmental accelerations in metabolic and vascular tissues. Exceptional longevity presents the converse, with epigenetic clocks indicating decelerated aging. Centenarians consistently display a younger epigenetic age than their chronological age across multiple clocks, with deviations typically 2-4 years below expected, reflecting resilient epigenomic maintenance. Supercentenarians aged 110 and older exhibit minimal epigenetic drift, as validated by specialized clocks designed for extreme age verification, suggesting stabilized methylation patterns. In Ashkenazi Jewish cohorts, centenarians harbor protective genetic variants, such as those modulating IGF-1 signaling, associated with enhanced longevity resilience; centenarians generally exhibit reduced epigenetic age acceleration. By 2025, epigenetic clocks have informed participant selection in trials, prioritizing individuals with decelerated clocks to evaluate interventions like caloric restriction mimetics, thereby enhancing trial efficiency for anti-aging outcomes.

Evolutionary and Mechanistic Implications

Epigenetic clocks serve as proxies for by linking accelerated epigenetic age to outcomes such as earlier age at first , increased number of sexual partners, and reduced timelines in women. In evolutionary terms, these clocks reflect trade-offs where early-life investments in may hasten biological aging, aligning with observations that high reproductive output correlates with shorter lifespans across . Furthermore, the of epigenetic clock signatures across mammals underscores their evolutionary robustness; for instance, a 2022 pan-mammalian epigenetic clock identified conserved CpG sites that predict chronological age with high accuracy in diverse , including whales, where patterns at these loci mirror those in humans despite vast differences in lifespan. This cross-species applicability suggests that epigenetic clocks evolved as reliable indicators of developmental and aging processes under shared selective pressures. Antagonistic manifests in drift, where early-life epigenetic stability supports but later-life drift accelerates aging-related disorders, as evidenced by loci that gain or lose disorder with age, contributing to the ticking of epigenetic clocks. Sex differences in epigenetic aging reveal females ticking slower by approximately 0.5 years on average compared to males, a pattern observed across multiple clocks and attributed to evolutionary protections enhancing female for post-reproductive caregiving. Ethnic variations further highlight biases, with Hispanics showing higher extrinsic epigenetic age acceleration (e.g., about 1 year older on GrimAge clocks) relative to , potentially reflecting adaptive trade-offs in immune function where heightened responses confer survival advantages in pathogen-rich environments but accelerate aging. These disparities align with broader evolutionary dynamics, such as antagonistic in immunity, where sex-specific immune investments—stronger in females—optimize but impose later costs. Epigenetic clocks integrate with the disposable theory by framing maintenance as a , where limited invested in epigenetic fidelity post-reproduction leads to drift and aging, as seen in clocks that track deterioration after peak reproductive years. Similarly, the predictive adaptive response posits that early-life exposures reprogram patterns to anticipate future environments, altering clock rates; for example, prenatal stressors accelerate epigenetic age in , preparing them for harsh conditions at the cost of accelerated aging. Looking ahead, epigenetic clocks hold promise in , as demonstrated by 2025 studies on engineered tissues where reprogramming resets cellular age, enabling rejuvenated anthrobots from adult cells. Evolutionary simulations further model drift rates, revealing that epigenetic instability scales with maximum lifespan across vertebrates, informing predictions of aging trajectories in novel biological systems.

Mechanisms and Theories

Epigenomic Maintenance Hypothesis

The epigenomic maintenance hypothesis posits that epigenetic clocks serve as a readout of an underlying system responsible for preserving the fidelity of epigenetic marks, particularly patterns, across the lifespan. This system counteracts stochastic errors and environmental perturbations to maintain cellular identity and function, but its imperfect nature leads to progressive drift in levels, causing the clock to advance. Central to this process is the dynamic balance between DNA methyltransferases (DNMTs), which establish and propagate methylation, and ten-eleven translocation () enzymes, which mediate demethylation; disruptions in this equilibrium accumulate errors, especially during cell divisions, manifesting as age-related epigenetic changes. Supporting evidence includes strong between mutation burdens and epigenetic clock acceleration. For instance, a -based clock predicts chronological age with a Pearson of r=0.70, and its predictions align closely with clocks (r=0.81 overall, partial r=0.60 after controlling for age), suggesting that efforts against mutational damage contribute to clock progression. Recent studies show that CpG mutations coincide with local and regional changes, enabling a mutation-based clock that with epigenetic clocks (r=0.81), further supporting this role. Additionally, sirtuins, such as SIRT1, deacetylate and stabilize to boost its methyltransferase activity, thereby supporting epigenetic . Theoretical models under this hypothesis describe feedback loops in which epigenetic drift impairs regulation, triggering as a protective response to prevent propagation of errors. Simulations of epigenetic variation indicate that decay in accounts for a substantial portion of clock variance, with processes explaining 66-75% of the accuracy in Horvath's multi-tissue clock. These models highlight how cumulative imperfections in the maintenance system predict aging trajectories without relying solely on external damage. Critiques of the note its limited for non-dividing cells, such as neurons, where clock progression occurs independently of replication-associated errors, challenging the emphasis on division-linked accumulation. Nonetheless, it offers a complementary to damage-centric views by focusing on active, systemic preservation of the epigenome, with brief intersections to strategies that target maintenance enzymes like sirtuins.

DNA Damage and Repair Perspectives

The DNA damage and repair perspective posits that epigenetic clocks primarily reflect the accumulation of unrepaired or recurrent DNA lesions, such as those induced by or ultraviolet radiation, which disrupt normal patterns during repair processes. According to this , double-strand breaks and other genomic insults trigger the formation of repair foci that recruit epigenetic regulators, temporarily displacing them from their typical genomic positions and leading to stochastic changes in at nearby CpG sites. Regions near fragile genomic sites, prone to breakage, exhibit hypermethylation as a consequence of repeated repair attempts, contributing to the progressive drift observed in epigenetic clocks. This view aligns with the broader , where unrepaired lesions parallel the erosion of epigenetic fidelity. Supporting evidence includes observations that accelerated epigenetic clock progression correlates strongly with markers of DNA damage, such as gamma-H2AX foci, which indicate ongoing double-strand break repair. In mouse models of characterized by DNA repair deficiencies, such as those with mutations in pathways, epigenetic aging advances significantly faster than in wild-type counterparts, with patterns mimicking chronological aging at an accelerated rate. These findings suggest that inefficiencies directly propel the changes underlying clock measurements. Key pathways involve (BER) and double-strand break (DSB) repair mechanisms, where enzymes like poly(ADP-ribose) polymerase 1 () modulate by altering accessibility and interacting with DNA methyltransferases during lesion resolution. For instance, activation in response to oxidative can inhibit or promote at specific loci, influencing clock-associated CpG sites. However, critiques of this perspective highlight an overemphasis on as the sole causal driver, noting that in certain tissues, such as the , epigenetic alterations may precede detectable DNA s, suggesting bidirectional or independent dynamics. Tissue-specific variations in susceptibility further complicate strict causality, as clocks in proliferative tissues show stronger repair-linked drift compared to post-mitotic ones.

References

  1. [1]
    DNA methylation age of human tissues and cell types
    I developed a multi-tissue predictor of age that allows one to estimate the DNA methylation age of most tissues and cell types.
  2. [2]
    The relationship between epigenetic age and the hallmarks of aging ...
    May 16, 2022 · Epigenetic clocks are mathematically derived age estimators that are based on combinations of methylation values that change with age at ...
  3. [3]
    Epigenetic Clocks: Beyond Biological Age, Using the Past to Predict ...
    As “historical data” embedded within DNA, epigenetic clocks have demonstrated substantial predictive capabilities, offering unique insights into age-related ...
  4. [4]
    A systematic review of phenotypic and epigenetic clocks used for ...
    This review aimed to systematically survey all proposed epigenetic and phenotypic clocks to date, excluding mitotic clocks (i.e., cancer risk clocks) and those ...
  5. [5]
    Epigenetic ageing clocks: statistical methods and emerging ...
    Jan 13, 2025 · Epigenetic clocks have emerged as powerful machine learning tools, not only to estimate chronological and biological age but also to assess the efficacy of ...Missing: 2023 | Show results with:2023
  6. [6]
    Infinium MethylationEPIC v2.0 Kit | Methylation profiling array - Illumina
    This kit uses the Illumina Methylation Assay, a microarray technology that provides quantitative methylation measurement at the single-CpG-site level, powering ...
  7. [7]
    Comparison of DNA methylation measured by Illumina 450K ... - NIH
    Both EPIC and 450K chips were analyzed using the Illumina Hi-Scan system. DNA methylation was measured at 485,512 CpG sites on the 450K BeadChip and 866,836 CpG ...
  8. [8]
    Quantification of the pace of biological aging in humans through a ...
    May 5, 2020 · We report a blood-DNA-methylation measure that is sensitive to variation in pace of biological aging among individuals born the same year.
  9. [9]
    The Role of DNA Methylation in Aging, Rejuvenation, and Age ...
    DNA methylation patterns are established by the de novo DNA methyltransferases (DNMTs) DNMT3A and DNMT3B and are subsequently maintained by DNMT1. Aging and ...
  10. [10]
    Epigenetic regulation of aging: implications for interventions ... - Nature
    Nov 7, 2022 · Here, we review recent work on the epigenetic regulation of aging and outline the advances in intervention strategies for aging and age-associated diseases.
  11. [11]
    Aging and DNA methylation - PMC - PubMed Central - NIH
    DNA hypermethylation in inflamed tissues is also strongly targeted to genes recognized by the Polycomb complex [38]. As inflammatory processes increase with age ...
  12. [12]
    Aging and DNA methylation | BMC Biology | Full Text - BioMed Central
    Jan 31, 2015 · In this Opinion article, we summarize how changes in DNA methylation occur during aging in mammals and discuss examples of how such events may contribute to ...
  13. [13]
    Improved precision of epigenetic clock estimates across tissues and ...
    Aug 23, 2019 · This study indicates that the epigenetic clock can be improved by increasing the training sample size and that its association with mortality attenuates.Study Population · Age Predictors With... · Author Information
  14. [14]
    DunedinPACE, a DNA methylation biomarker of the pace of aging
    Jan 14, 2022 · DunedinPACE showed high test-retest reliability, was associated with morbidity, disability, and mortality, and indicated faster aging in young adults with ...
  15. [15]
    Article Accurate age prediction from blood using a small set of DNA ...
    We present a compact epigenetic clock for blood, which outperforms state-of-the-art models using only 30 CpG sites.
  16. [16]
    EpiMedTech Global Launches epiGeneComplete: A Breakthrough ...
    Oct 31, 2024 · Singapore, 2024 ... EpiMedtech Global Announces FDA Registration of EPIAGE, the First Epigenetic Age Test Registered by the FDA.
  17. [17]
    scEpiAge: an age predictor highlighting single-cell ageing ... - Nature
    Aug 31, 2024 · We show that scEpiAge allows us to predict epigenetic age more accurately in single cells than previous methods and can also be applied to bulk ...
  18. [18]
    SWAN: Subset-quantile Within Array Normalization for Illumina ...
    Jun 15, 2012 · Here we present Subset-quantile Within Array Normalization (SWAN), a new method that substantially improves the results from this platform.
  19. [19]
    DeepMAge: A Methylation Aging Clock Developed with Deep ...
    In this article, we explore how deep learning performs in a DNA methylation setting and compare it to the current industry standard—the 353 CpG clock published ...Missing: forests 2020s
  20. [20]
    An age classification model based on DNA methylation biomarkers ...
    Aug 2, 2025 · An age classification model based on DNA methylation biomarkers of aging in human peripheral blood using random forest and artificial neural ...Missing: 2020s | Show results with:2020s
  21. [21]
    Missing value estimation methods for DNA methylation data
    Feb 23, 2019 · We present a simple and computationally efficient imputation method, metyhLImp, based on linear regression.
  22. [22]
    Cell-type specific epigenetic clocks to quantify biological age at cell ...
    We also built an elastic net clock adjusting for 9 immune-cell subtypes, by merging together the estimated fractions of the naïve and memory lymphocyte ...
  23. [23]
    Epigenetic Dissection of Intra-Sample-Heterogeneity with online GUI
    Nov 9, 2019 · Many tools have emerged to address this issue, including several R ... Here we present a web application for cell-type deconvolution ...Missing: clock | Show results with:clock
  24. [24]
    Adapting Blood DNA Methylation Aging Clocks for Use in Saliva ...
    Jul 28, 2021 · There are multiple cell-type deconvolution tools that allow to ... We used one such reference-based tool—EpiDISH—to derive a linear ...
  25. [25]
    [PDF] Comparison of DNA methylation clocks in black South African men
    Mar 8, 2021 · This study compares five DNA methylation clocks (Horvath, Hannum, skin and blood, PhenoAge, and GrimAge) in 120 older black South African men ...<|control11|><|separator|>
  26. [26]
    Map of epigenetic age acceleration: a worldwide meta-analysis
    Mar 17, 2024 · Since DunedinPACE does not estimate epigenetic age but the aging rate, we discussed it separately (mean values for the considered tissues ...
  27. [27]
    Methylation Clocks Do Not Predict Age or Alzheimer's Disease Risk ...
    Jan 31, 2025 · Our results demonstrate that methylation clocks often fail to predict age and AD risk beyond their training populations and suggest avenues for improving their ...2 Results · Methylation Clocks Rarely... · 3 Discussion
  28. [28]
  29. [29]
    Common DNA sequence variation influences epigenetic aging in ...
    The non-heritable epigenetic clock exhibits predictive performance in African, European-ancestry ... Systematic underestimation of the epigenetic clock and ...
  30. [30]
    [PDF] A Multi-Omics and Bioenergetics Longitudinal Aging Dataset in ...
    Nov 15, 2021 · The telomere length qPCR assay contained 5 technical replicates of the same sample. (HC1, passage 10, 42 days grown). Replicates had a C.V. of 9 ...
  31. [31]
    epigenetic clock and telomere length are independently associated ...
    Apr 13, 2016 · These results suggest that telomere length and epigenetic clock estimates are independent predictors of chronological age and mortality risk.
  32. [32]
    Epigenetic Aging Clocks Compared: Which One Predicts Mortality ...
    Jul 14, 2025 · The GrimAge epigenetic clock outperforms other clocks, including PhenoAge, Horvath 1, Hannum, and DunedinPACE, in predicting mortality.
  33. [33]
    A catalogue of omics biological ageing clocks reveals substantial ...
    We use ~1000 participants to compare fifteen omics ageing clocks, with correlations of 0.21-0.97 with chronAge, even with substantial sub-setting of biomarkers.
  34. [34]
  35. [35]
    A metabolomic profile of biological aging in 250,341 individuals from ...
    Sep 15, 2024 · The metabolomic profile of aging is complex. Here, we analyse 325 nuclear magnetic resonance (NMR) biomarkers from 250,341 UK Biobank ...
  36. [36]
    DNA methylation GrimAge strongly predicts lifespan and healthspan
    We demonstrate that DNAm GrimAge stands out among existing epigenetic clocks in terms of its predictive ability for time-to-death.
  37. [37]
    Somatic mutation as an explanation for epigenetic aging - PMC
    Dec 9, 2023 · DNA methylation marks have recently been used to build models known as “epigenetic clocks” which predict calendar age.
  38. [38]
    Multi-omics and Multi-organ Aging Clocks Digitize Human Aging
    Feb 7, 2025 · Multi-organ biological aging clocks derived from clinical phenotypes and neuroimaging have emerged as valuable tools for studying human aging and disease.
  39. [39]
    GWAS of epigenetic aging rates in blood reveals a critical role for ...
    Jan 26, 2018 · Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 14, R115 (2013). Article PubMed PubMed Central Google Scholar.Missing: paper | Show results with:paper
  40. [40]
    A meta-analysis of genome-wide association studies of epigenetic ...
    Hannum-based epigenetic age is based on DNA methylation levels at the 71 CpGs identified by Hannum et al. (2013) [7]. Hannum-based epigenetic age acceleration ( ...
  41. [41]
    Epigenetic age acceleration mediates the association between ...
    Jun 3, 2023 · We demonstrated that GrimEAA, DNAm-based smoking pack-years, DNAm PAI-1 levels, DunedinPACE, and PhenoEAA mediated smoking associations with diabetes-related ...
  42. [42]
    Physical Activity Is Associated With Decreased Epigenetic Aging - NIH
    Jun 13, 2025 · Physical activity and exercise may influence epigenetic aging, suggesting a pathway through which it promotes healthier aging and reduces ...
  43. [43]
    One-year Mediterranean diet promotes epigenetic rejuvenation with ...
    Jan 24, 2020 · Mediterranean diet has been proposed to promote healthy aging, but its effects on aging biomarkers have been poorly investigated.
  44. [44]
    Long-term exposure to air pollution is associated with biological aging
    A 0.97 μg/m3 increase in ambient PM2.5 was associated with a 0.32 - 0.35 y increase in EEAA indicating accelerated epigenetic aging. This association remained ...
  45. [45]
    Caloric restriction delays age-related methylation drift - Nature
    Sep 14, 2017 · We report that epigenetic drift is conserved across species and the rate of drift correlates with lifespan when comparing mice, rhesus monkeys, and humans.
  46. [46]
    The Anti-Aging Mechanism of Metformin: From Molecular Insights to ...
    The results showed that patients taking metformin experienced significantly slower epigenetic age acceleration according to the Horvath and Hannum clocks, ...
  47. [47]
    Epigenetic aging in patients diagnosed with coronary artery disease
    Jan 31, 2023 · Participants with a confirmed CAD had on average a 2.5-year higher DNAmAA than patients with a normal angiogram.
  48. [48]
    A Prospective Study of Epigenetic Age Acceleration and Incidence ...
    Mar 1, 2019 · One study of postmenopausal women found no association between epigenetic age acceleration and risk of coronary heart disease (CHD), but little ...
  49. [49]
    Association between epigenetic age and type 2 diabetes mellitus or ...
    Apr 25, 2024 · This study explored the associations between epigenetic age metrics and T2DM or glycemic traits, based on 1070 twins (535 twin pairs) from the Chinese National ...
  50. [50]
    Obesity accelerates epigenetic aging of human liver - PNAS
    Oct 13, 2014 · Here we use a recently developed biomarker of aging (known as “epigenetic clock”) to study the relationship between epigenetic age and obesity ...
  51. [51]
    Evidence of accelerated epigenetic aging of breast tissues in ...
    Feb 24, 2022 · A prospective study in peripheral blood showed that accelerated epigenetic age is associated with increased risk of developing breast cancer, ...
  52. [52]
    A DNA methylation-based liquid biopsy for triple-negative breast ...
    Jun 16, 2021 · Several studies have suggested that accelerated epigenetic aging may be linked to increased risk of breast cancer. We did not observe a ...
  53. [53]
    Accelerated aging with HIV occurs at the time of initial HIV infection
    Jul 15, 2022 · Epigenetic DNA methylation patterns can evaluate acceleration of biological age relative to chronological age. The impact of initial HIV ...
  54. [54]
    insights from a genome-wide DNA methylation study | Clinical ...
    Aug 20, 2024 · We observed a slight but significant epigenetic age acceleration (EAA) in post-COVID-19 patients across all available clocks [42,43,44,45], ( ...Missing: haulers | Show results with:haulers
  55. [55]
    Epigenetic biomarkers of ageing are predictive of mortality risk in a ...
    Dey 13, 1400 AP · There was weak evidence that the addition of AgeAccelGrim to the clinical model improved 3-year mortality prediction (area under the receiver ...
  56. [56]
    A blood-based epigenetic clock for intrinsic capacity predicts ...
    Jun 4, 2025 · A blood-based epigenetic clock for intrinsic capacity predicts mortality and is associated with clinical, immunological and lifestyle factors.
  57. [57]
    Epigenetics-targeted drugs: current paradigms and future challenges
    Nov 26, 2024 · To date, four categories of epigenetics-targeted drugs have received the Food and Drug Administration (FDA) approval for clinical use, with ...
  58. [58]
    Epigenetic Regulation of Aging and its Rejuvenation - PMC
    Sep 1, 2025 · Epigenetic Regulation of Aging ... This plasticity requires precise epigenetic regulation, including DNA demethylation (mediated by TET enzymes) ...
  59. [59]
  60. [60]
    Multi-omics characterization of partial chemical reprogramming ...
    Jun 30, 2023 · We show that partial chemical reprogramming reduces the biological age of mouse fibroblasts. We demonstrate that these changes have functional impacts.
  61. [61]
    Senolytic compounds reduce epigenetic age of blood samples in vitro
    Feb 4, 2025 · Exploring the effects of Dasatinib, Quercetin, and Fisetin on DNA methylation clocks: a longitudinal study on senolytic interventions. Aging ...
  62. [62]
    Exploring the effects of Dasatinib, Quercetin, and Fisetin on DNA ...
    This study aimed to assess the effects of Dasatinib and Quercetin (DQ) senolytic treatment on DNA methylation (DNAm), epigenetic age, and immune cell subsets.
  63. [63]
    CRISPR epigenome editor offers potential gene therapies
    Apr 25, 2025 · CRISPR-based epigenome editors offer therapeutic potential without causing DNA breaks. The epigenome editor uses a dCas9 and gRNA machinery that ...Missing: organoids | Show results with:organoids
  64. [64]
    April 2025 Update on Regulation of New Genomic Techniques in ...
    Apr 25, 2025 · On March 14, 2025, EU Member States agreed in the European Council on a common position to move forward with development of new rules for certain genetically ...Missing: framework clock<|control11|><|separator|>
  65. [65]
    Universal DNA methylation age across mammalian tissues - Nature
    Aug 10, 2023 · As predicted by the epigenetic clock theory of aging, universal epigenetic clocks provide a continuous readout of age from early development ...
  66. [66]
    The epigenetic clock is correlated with physical and cognitive fitness ...
    Jan 22, 2015 · In the present study, we find that the Horvath predictor slightly underestimates the ages (by about 4 years) in older subjects. However ...
  67. [67]
    EpiAge: a next-generation sequencing-based ELOVL2 epigenetic ...
    This study introduces EpiAgePublic, a new method to estimate biological age using only three specific sites on the gene ELOVL2, known for its connection to ...Missing: artifacts | Show results with:artifacts
  68. [68]
    The cerebellum ages slowly according to the epigenetic clock
    Here we utilize a recent biomarker of aging (referred to as epigenetic clock) to assess the epigenetic ages of up to 30 anatomic sites from supercentenarians ( ...
  69. [69]
    DNA methylation age is elevated in breast tissue of healthy women
    Mar 31, 2017 · Our data clearly demonstrate that female breast tissue has a higher epigenetic age than blood collected from the same subject. We also observe ...
  70. [70]
    Cell-type specific epigenetic clocks to quantify biological age at cell ...
    Dec 29, 2024 · Epigenetic clocks have emerged as promising tools for estimating biological age, yet they have been developed from heterogeneous bulk tissues, ...Missing: EpiDISH | Show results with:EpiDISH
  71. [71]
    Within Subject Cross-tissue Analyses of Epigenetic Clocks in ... - NIH
    In this study, we assessed and compared several epigenetic clocks that capture different aging aspects in individuals with and without SUD in both postmortem ...
  72. [72]
    Cell-type specific epigenetic clocks to quantify biological age at cell ...
    Jul 31, 2024 · This study develops cell-type specific epigenetic clocks for improved biological age estimates, showing acceleration in Alzheimer's and liver ...
  73. [73]
    Epigenetic clock for skin and blood cells applied to Hutchinson ...
    Here, we describe a novel powerful epigenetic age estimator (called the skin & blood clock) that outperforms existing DNAm-based biomarkers when it comes to ...Missing: 2019 | Show results with:2019
  74. [74]
    Epigenomic signature of accelerated ageing in progeroid Cockayne ...
    The epigenomic remodelling of accelerated ageing in CS displayed both commonalities and differences with other progeroid diseases and regular ageing. CS shared ...
  75. [75]
    Accelerated epigenetic aging in Down syndrome - PubMed - NIH
    Trisomy 21 significantly increases the age of blood and brain tissue (on average by 6.6 years, P = 7.0 × 10(-14)).
  76. [76]
    Huntington's disease accelerates epigenetic aging of human brain ...
    A multivariate model analysis suggests that HD status increases biological age by 3.2 years. Accelerated epigenetic age can be observed in specific brain ...
  77. [77]
    Menopause accelerates biological aging - PNAS
    Jul 25, 2016 · This is a definitive study that shows an association between age of menopause and biological aging (measured using the epigenetic clock).Menopause Accelerates... · Sign Up For Pnas Alerts · Results
  78. [78]
    HIV-1 Infection Accelerates Age According to the Epigenetic Clock
    We show that HIV infection leads to an increase in epigenetic age both in brain tissue (7.4 years) and blood (5.2 years).
  79. [79]
    Epigenetic ageing accelerates before antiretroviral therapy and ...
    In a longitudinal study over more than 17 years, epigenetic ageing accelerated during untreated HIV infection and decelerated during suppressive ART.
  80. [80]
    Accelerated epigenetic aging in Werner syndrome - PubMed - NIH
    This study shows that WS is associated with an increased epigenetic age of blood cells which is independent of changes in blood cell composition.
  81. [81]
    Genetic and Epigenetic Insights into Werner Syndrome
    Feb 14, 2025 · AWS is an accelerated aging syndrome which, compared to WS, shows an earlier age of onset and a faster progression. Among the genes involved in ...
  82. [82]
    Centenarians Consistently Present Younger Epigenetic Age Than ...
    Oct 17, 2022 · Centenarians consistently present a younger epigenetic age than their chronological age with four epigenetic clocks based on a small number of CpG sites.Missing: Ashkenazi protective variants
  83. [83]
    epigenetic clocks for validating claims of exceptional longevity
    Here, we present three DNA methylation-based age estimators (epigenetic clocks) for verifying age claims of centenarians.
  84. [84]
    Depletion of loss-of-function germline mutations in centenarians ...
    Oct 19, 2024 · Our findings suggest that a protective genetic background, characterized by a reduced burden of damaging variants, contributes to exceptional longevity.
  85. [85]
    Methylation clocks for evaluation of anti-aging interventions
    May 5, 2025 · Methylation clocks promise a quick and inexpensive way to determine if a given intervention has power to rejuvenate, and a slower but reasonably ...
  86. [86]
    Epigenetic age acceleration and reproductive outcomes in women
    Results showed epigenetic age acceleration was directly associated with earlier age at first sex and increased sexual partner number.
  87. [87]
    Epigenetic clocks and female fertility timeline: A new approach to an ...
    Mar 21, 2023 · In this review, we mainly discussed the possible role of epigenetic clocks in the female reproductive health, starting with physiological ...
  88. [88]
    Epigenetic drift underlies epigenetic clock signals, but displays ... - NIH
    We find that epigenetic clock loci are enriched in regions that both accumulate and lose disorder with age, suggesting a link between DNA methylation disorder ...
  89. [89]
    Epigenetic gambling and epigenetic drift as an antagonistic ...
    Aug 7, 2025 · Epigenetic clock (eAge) algorithms utilize DNA methylation to estimate the age and risk factors for diseases as well as analyze the impact ...
  90. [90]
    Do Epigenetic Clocks Provide Explanations for Sex Differences in ...
    The first published results on biological age determined by epigenetic clocks have shown that men tend to be biologically older than women (16–20). This study ...
  91. [91]
    An epigenetic clock analysis of race/ethnicity, sex, and ... - PubMed
    Aug 11, 2016 · In blood, Hispanics and Tsimane Amerindians have lower intrinsic but higher extrinsic epigenetic aging rates than Caucasians. African-Americans ...
  92. [92]
    Epigenetic clocks and programmatic aging - ScienceDirect.com
    The ubiquity of genes exhibiting AP reflects the major role of “buy now, pay later”-type evolutionary trade-offs as a major underlying cause of aging. 3.2.
  93. [93]
    Aging clocks based on accumulating stochastic variation - Nature
    May 9, 2024 · Here we show that accumulating stochastic variation in purely simulated data is sufficient to build aging clocks.
  94. [94]
    Developmental Tuning of Epigenetic Clock - PMC - PubMed Central
    Nov 22, 2018 · According to the concept of predictive adaptive response, early-life cues can be used by an organism to rearrangement of the epigenome in a ...
  95. [95]
    A Reset on the Cellular Aging Clock | Tufts Now
    Jun 17, 2025 · When cells assemble into tiny biological robots called Anthrobots, they become biologically younger than their original adult cells.
  96. [96]
    The rate of epigenetic drift scales with maximum lifespan across ...
    Nov 25, 2023 · Epigenetic drift increases with age and maximum lifespan. RD and global disorder increase with age in rats, mice, dogs, and baboons, but the ...
  97. [97]
    SIRT1 deacetylates the DNA methyltransferase 1 (DNMT1) protein ...
    Here we report that the histone deacetylase SIRT1 regulates the activities of DNMT1, a key enzyme responsible for DNA methylation.Missing: boosts DNMT
  98. [98]
    Quantifying the stochastic component of epigenetic aging - Nature
    May 9, 2024 · This elastic net clock model defines our StocH clock. The exact same procedure was followed for the 514 Zhang clock and 513 PhenoAge clock ...
  99. [99]
    Epigenetics, DNA damage, and aging - JCI
    Aug 15, 2022 · In addition to DNA methylation, epigenetic modifications of histones play an important role in DNA repair, the DDR, and aging. These ...
  100. [100]
    DNA repair‐deficient premature aging models display accelerated ...
    Dec 22, 2023 · These findings highlight that mouse models with deficiencies in DNA repair, unlike other premature aging models, display accelerated epigenetic age.
  101. [101]
    PARP1 as an Epigenetic Modulator: Implications for the Regulation ...
    Jan 30, 2024 · 2.4. The Regulation of DNA Methylation by PARP1. PARP-mediated PARylation has been associated with the regulation of DNA methylation (Figure 1).