Abstract
Investigating aging has become a subject of intense biomedical focus. This has coincided with an unprecedented rise in epigenetic research. DNA methylation (DNAm) is the most comprehensively investigated epigenetic process. Epigenetic clocks are capable of statistically correlating DNAm changes with chronological age. DNAm changes are also proving to be a worthwhile biomarker of age-related disease, while emerging evidence suggests this epigenetic mechanism could be an effective diagnostic tool for disease detection. Such investigative progress has significant implications for health care. In this brief review we examine some recent findings in this area. The overarching aim and scope of the work is to address the relationship between aging, DNAm, and health. We commence by briefly introducing aging. Next, DNAm and age-related disease are discussed. Thirdly, we critically examine epigenetic clocks. We conclude by exploring recent advances in the use of biosensors for measuring DNAm and disease detection.
Keywords: Aging, DNA methylation, age-related disease, biomarker, epigenetics
Introduction - What is Aging and Why Do We Age?
Aging is a remarkably familiar yet incredibly difficult process to define. Attempts to define this captivating biological phenomenon have led to considerable debate within biogerontology [1]. Consequently, many definitions of aging exist. However, a simple definition of aging is that it is the changes which accumulate with age, that progressively render an organism increasingly vulnerable to mortality [2]. Despite the uncertainty in defining aging, the last few decades have witnessed considerable progress in unravelling both the proximate and ultimate causes of aging. The ultimate causes of aging center on why it evolved. Considerable debate surrounds this issue; however, subscribers to the classic evolutionary theories generally agree that aging is due to the diminishing capacity of natural selection with increasing age [3,4]. In other words, aging is mainly viewed as a non-adaptive by-product of evolution that escaped natural selection. This crucial evolutionary concept provides a template for understanding the proximate mechanisms of aging. Proximate mechanisms suggest how aging unfolds. Many processes have been empirically associated with aging in a wide variety of canonical organisms [5,6]. However, due to the complexity of aging, it is unlikely it can be reduced to a single process. Despite this, routine attempts are made at performing cartesian reduction on the aging process. Perhaps nowhere better exemplifies recent efforts to do this than the so called “hallmarks of aging” by López-Otín et al. (2023) [7]. In this work, aging is reduced to 12 biological processes. The hallmarks include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. Unquestionably each process is important, however as the authors would duly acknowledge other mechanisms, both discovered and yet to be elucidated, inevitably contribute to aging.
The recursive approach outlined above is understandable if it is acknowledged that biologists typically solve complex puzzles by breaking them down into more manageable problems; the solutions to the smaller problems are then assembled to provide an explanation of the larger problem [8,9]. Within this landscape it is apparent epigenetics has an increasing role to play in aging research. Epigenetics centers on the regulation of gene activity without altering the genetic sequence of DNA [10]. Several epigenetic processes exist; these include histone modification (acetylation and methylation), non-coding RNAs, chromatin remodeling, and DNA methylation (DNAm). DNAm is the most widely studied aspect of epigenetics. DNAm, is an epigenetic mark which occurs at cytosine phosphate-guanine (CpG) motifs; and is formed by the covalent bonding of methyl groups from S-adenosyl methionine to the fifth carbon of cytosines [11]. Persistent observational and empirical evidence over the last decade has revealed that age related changes to DNAm patterns correlate with the diseases of aging and chronological age in humans and other species [12-14]. There are numerous age-related diseases including dementia, cardiovascular disease (CVD), and cancer. The Surveillance, Epidemiology and End Results (SEER) Program (SEER 22 2017-2021) reported that the median age of a cancer diagnosis was 67 years of age. Notably, 30.2% of diagnoses occurred in people aged 65-74 years of age. This is much higher than in younger groups: 23.8% of cases were diagnosed in individuals aged 55-64 years, 11.0% in 45-54-year-olds, and 4.8% in 35-44 year-olds [15]. Similar trends are observed with heart disease and age. For example, the self-reported prevalence of heart disease in 2019 in adults between the ages of 18-44 years of age was 1.0%. This elevated to 9% in those aged 55-64 years, 14.3% in 65-74-year-olds and 24.2% in those over the age of 75 years [16]. Given that by 2050, the number of people over the age of 60 globally, will nearly double to 22%, (compared to 2015) [17], understanding the underpinning mechanisms of age-related disease is of vital importance. Thus, a continual drive exists to better understand how changes to DNAm are linked to both aging and age-related disease [18]. The aim and scope of this brief review is to examine the relationship between aging and DNAm.
DNA Methylation and Age-related Disease
As alluded to above, DNAm involves the covalent addition of a methyl group (-CH3) to a cytosine located within a CpG site. This process is tightly regulated by DNA methyltransferases (DNMT), which are responsible for maintenance methylation of hemimethylated DNA (DNMT1) during DNA replication, and de novo methylation (DNMT3a and DNMT3b) [19]. Demethylation is regulated by ten–eleven translocation (TET) enzymes [20]. The epigenetic mechanism of DNAm modulates gene expression; typically, transcriptional silencing is associated with hypermethylation of promotor sites, while gene expression is frequently upregulated when hypomethylation occurs in the gene body [21]. DNAm patterns change with age, in a process termed epigenetic drift. Specifically, regions exhibiting significant methylation in younger years often display reduced methylation levels, while hypomethylated sites gain methyl groups. This (accelerated) epigenetic drift has been associated with several age-related diseases, including diabetes [22], Alzheimer’s disease (AD) [23], dementia [24], CVD [25], and cancer [26].
Numerous modifiable risk factors are associated with aberrant DNAm patterns. One such risk factor is diet, which can encompass various aspects of nutrition including early postnatal overnutrition [27], vitamin intake [28,29] and alcohol intake [30,31]. Other risk factors include high BMI [32,33], physical inactivity [34,35], stress [36], exposure to pollutants [37,38], smoking [39,40], socioeconomic status [41,42], chemotherapy treatment [43] and virus exposure [44,45]. Significantly, these aberrant methylation patterns can be long lasting, persisting decades after exposure [46]. For example, research indicates that some CpG sites may remain differentially methylated up to 35 years after the cessation of smoking, whilst other revert to normal within decades [47]. Conversely, strategies to ameliorate aberrant methylation patterns have proved promising, with evidence suggesting that healthy diet and lifestyle treatment groups exhibit reduced epigenetic aging, which may have implications for age-related disease [48-50]. It is also important to recognize the significance of parental and grand-parental health on the epigenome of offspring [51]. For instance, there is evidence to indicate the parental exposure to pollution [52], prenatal stress [53], maternal undernutrition [54], maternal overnutrition [55], paternal under- [56] and over-nutrition [57] are also associated with perturbed offspring methylation in genes related to age-associated disease. Pivotal observations of the Dutch hunger winter birth cohort demonstrate the impact of famine on offspring in later life. Offspring were at a higher risk of developing several age-related diseases including type 2 diabetes, cancer, and CVD [58].
Typically affecting older people, CVD is the leading cause of death globally [59]. CVD is a general term for conditions affecting the heart or blood vessels, and typically affects older adults. CVD encompasses a range of conditions including coronary heart disease, cerebrovascular disease, and peripheral artery disease. A range of evidence exist which links perturbations in DNAm with CVD [60]. Recently we critically examined the role epigenetics has in vascular dementia (VaD), a condition related to CVD [61]. This work revealed the significant impact diet, aging, comorbidities, the gut microbiome, and pro-longevity molecules can have on vascular dementia pathobiology. More broadly, DNAm changes have been associated with the development and progression of CVD [60]. To this end, there have been several predictive models created in recent years which estimate CVD risk from DNAm [62].
In addition to vascular dementia, other forms of dementia such as AD [63], frontotemporal dementia [64], and Lewy Body dementia [65], have been associated with aberrant DNAm patterns. Over the past couple of decades, the mortality rate of dementia and AD has increased steadily. In 2016, these chronic and incurable neurodegenerative disorders were the leading cause of death in people over the age of 80 years in England [66]. In one epigenome-wide association study of the cortex, 334 differentially methylated positions (DMPs) were identified in AD [67]. Gene promoter DNAm has been observed to significantly differ with increased burden of amyloid beta (Aβ) plaques in neurons of the dentate gyrus. This includes the hypomethylation of the gene promoter for gamma-secretase which cleaves an amyloid precursor into Aβ peptides [68]. Other studies focused on extracranial tissues. One study found 64 DMPs associated with AD in white blood cells [69]. Significant alterations in DNAm patterns have also been observed in genes associated with lipid regulation and lysosomal transmembrane gene [63].
DNAm changes also play a significant role in cancer [21]. Importantly, the degree of methylation can be used as a prognostic marker, indicating the grade and sizing of a tumor [70,71]. Due to this association, the use of DNAm as a tool for detecting cancer in its early stages has emerged, particularly from liquid biopsy. This refers to a non-invasive collection of a sample such as blood, saliva, or urine to detect cancer from cfDNA, which has clear benefits when compared to the traditional collection of a biopsy [72]. As such techniques continue to progress, there is little doubt they will improve our understanding of the nexus between DNAm and aging.
DNA Methylation as a Measure of Aging
Many epigenetic clocks have been developed over the past decade to correlate biological age with chronological age [73,74]. Horvath’s clock is widely regarded as the first epigenetic clock [12]. It focuses on DNAm at 353 CpG sites and incorporates data from 51 healthy human tissues and cell types. This clock lacks specificity for disease. Moreover, it remains to be determined, how the 353 CpG’s which define the clock are linked to aging. In fact, they may have nothing to do with aging. Arguably the Hannum clock is simpler as it focuses on 71 CpG sites in blood [75]. However, this clock is limited to blood samples. The GrimAge clock, predicts mortality and age-related diseases using biomarkers including smoking and body mass index [76]. It is informed by clinical data from the Framingham study—this is a drawback as this is a very narrow demographic. PhenoAge combines DNAm data with a broad range of biomarkers to predict phenotypic age. It was also able to use DNAm biomarkers of aging to predict CVD and coronary heart disease. However, a limitation of PhenoAge is that its training data is built around a dataset that is not reflective of a diverse range of ethnic groups. This limitation of the clocks was brought into sharp focus recently when Yusipov and colleagues performed a comprehensive analysis using the largest compilation of publicly available DNAm data from healthy individuals—93 datasets encompassing 23 000 samples across 25 countries and 31 ethnicities [77]. The work investigated how geography, ethnicity, and health status influence epigenetic aging. The study evaluated PhenoAge and GrimAge. They identified significant inconsistencies in their outputs. PhenoAge often underestimated, while GrimAge tended to overestimate age in healthy individuals.
The criticism of the causality of epigenetic clocks outlined above is justified. However, emerging studies suggests that epigenetic clocks are improving. For instance, recent work was able to identify specific cytosines with methylation levels in genes implicated with longevity across different species [78]. Another clock was able to use DNAm levels to substantiate the age claims of centenarians [79]. Attempts to overcome clock limitations have also focused on using explainable AI (XAI) and deep learning (DL) tools. One study used a deep neural network-based epigenetic clock [80]. It was trained on a large dataset of 17 726 whole-blood samples across the human lifespan. Interestingly, it improved age prediction accuracy by integrating DNAm data and established CpG age markers. In another study DL and XAI were applied to large omic datasets [81]. A multi-view graph-level representation learning framework that incorporated biological networks was used to build molecular aging clocks at cell-type resolution. A ribosomal gene subnetwork linked to aging, independent of cell type was discovered. Using the same approach on DNAm data, they identified an age-related inflammatory pathway. This recent progress with richer AI methods suggests that the criticism of the original epigenetic clocks does not completely rule out the possibility that one day DNAm clocks will be embedded within a causal framework for aging. Thus, it is possible with further developments in AI and its applications to multi-omics data a more mechanistic understanding of aging could be achieved. This would constitute significant progress in how aging is measured.
Detecting DNA Methylation: A Diagnostic Tool for Future Healthcare?
Several methods exist which can quantify DNAm. Many analytical techniques require bisulphite treatment as a precursor to analysis. Bisulphite treatment results in the conversion of unmethylated cytosines to uracil, while methylated cytosine remain unchanged [21]. When followed with PCR, this results in methylated cytosines remaining as cytosines, while unmethylated cytosines appear as thymine. This modification can be detected at a single base level when followed by whole genome sequencing methods such as pyrosequencing [82]. In pyrosequencing, nucleotide incorporation is detected due to the release of pyrophosphate which transmits a light signal [82,83]. By performing a restriction digest prior to bisulphite treatment, targeted sequencing can be performed; reduced representation bisulphite treatment is a cost-effective alternative to genomic sequencing [84]. Microarray kits are a commonly used method of detecting DNAm due to the high throughput nature of the method and the ability to analyze up to almost one million CpG sites [85]. This method relies of the hybridization of test DNA to an immobilized oligonucleotide probe. Alternatively, nanopore technology enables the analysis of long sequence reads without the need for bisulphite treatment or PCR [86]. Other methods which don’t require bisulphite treatment include methylated DNA immunoprecipitation sequencing (MeDIP-seq), methylation-sensitive restriction enzyme sequencing (MRE-seq), and single-molecule real-time sequencing (SMRT-seq). In MEDIP-seq, 5mC antibodies bind to fragmented methylated DNA and undergo precipitation. Following DNA purification, sequencing can be conducted [87]. In MRE-seq, MREs can be exploited as an alternative way to study DNAm. These enzymes cleave only unmethylated sequences; the resulting fragments can be size-selected and sequenced (MRE-seq) [88]. SMRT-seq is a method which employs fluorescently labelled nucleotides and DNA polymerase. The addition of each nucleotide results in a fluorescence signal which indicates the nucleotide identity and/or modification such as 5mC [89].
Some electrochemical sensors rely on asymmetric PCR in place of traditional PCR methods to ascertain methylation. In this method, unequal concentrations of PCR primers result in ssDNA production. This complementary single stranded DNA can either be high in guanine (methylated, complementary to cytosine) or adenine (unmethylated, complementary to uracil). This outcome can be exploited by electrochemical techniques which utilize gold (working electrode). This is because adenines bind to gold with a higher affinity than guanines, resulting in greater resistance, which can be detected using techniques such as electrochemical impedance spectroscopy or differential pulse voltammetry [90]. DNA interactions with gold have also played an important role in the development of gold nanoparticle technology where DNAm can be detected via a color change [91,92].
Current research often centers on the exploration of DNAm data. Bioinformatics is a useful tool in epigenomic analysis, with numerous DNAm databases available [93-96] to aid in the understanding of the relationship between DNAm and disease [97], longevity [98], and environmental factors [99]. Following the advancement of machine learning, tools for diagnosis can be readily employed. For instance, one DL method achieved an overall accuracy of 95% for the diagnosis of CNS tumors, based on DNAm histopathology images and patient demographics [100]. Furthermore, in a clinical setting, it has been demonstrated that whole-genome bisulfite sequencing data for tumor samples could be utilized to predict long term survival in intrahepatic cholangiocarcinoma patients [101]. Moreover, cfDNA methylation data has been used to accurately predict patient response to neoadjuvant chemotherapy in 79% of bladder cancer cases [102].
Conclusions
Aging is an emergent process which is a by-product of the “simple” physical laws which have governed the evolution of life on this planet. In contrast to physical laws however, biological systems are far from simple. Natural selection has “generated” systems which are inherently complex. This complexity is all too evident when biological systems break down with age, and medical research strives to unravel the pathobiology of disease. This problem provokes the question, how can aging be better understood, so that diseases can be targeted earlier in life, and morbidity compressed. The challenge of elucidating aging becomes increasingly intractable if viewed solely from a physics perspective, and thus deemed due to increasing entropy with time. This is not scientific heresy. Solid arguments exist for interpreting aging entropically [103-105]. However, viewing aging through a thermodynamic lens does not mean the conundrum should be avoided. A significant body of evidence demonstrates that the trajectory of human aging is malleable [106]. Moreover, it has even been argued that aging is reversible. Indeed, DNAm is a strong candidate that could meditate the reversibility of aging because it has been shown that the partial reprogramming of induced pluripotent stem cells leads to rejuvenation in terms of epigenetic age [107].
The findings from this brief review also serve to emphasize the plasticity of aging and the contribution epigenetics makes to this. We revealed how epigenetics has presented significant therapeutic and diagnostic avenues for aging and health. It was illustrated how DNAm changes can be quantified and how these are associated with diseases including CVD, AD and cancer. An emphasis was also placed on how extrinsic factors such as diet and pollution can modulate the trajectory of epigenetic change. Measuring age using epigenetic clocks to determine health status was also critically evaluated. Importantly, we revealed DNAm clocks are imperfect measures of aging, which require further refinement if they are to be firmly embedded within a causality framework of aging. However, considerable effort is still required to understand the basic mechanisms of aging more broadly, and it is important not to lose sight of this, despite the remarkable progress which has been made in understanding aging and its relationship with health.
Acknowledgments
Some of this work was completed at a writing retreat at Missenden Abbey, Great Missenden, Buckinghamshire, UK, which was financially supported by the Food4Years network—a collaborative initiative funded by the Medical Research Council and the Biotechnology and Biological Sciences Research Council. In particular, the authors wish to thank Dr. Miriam Clegg from University College Cork and Dr. Chiara De Lucia of King’s College London, both members of Food4Years.
Glossary
- CpG
5’-C-phosphate-G-3’
- CVD
cardiovascular disease
- DNAm
DNA methylation
- DNMT
DNA methyltransferase
- PCR
polymerase chain reaction
- ssDNA
single stranded DNA
Author Contributions
MMcA: conceptualization, writing—original draft, writing—review and editing. AM (ORCID https://orcid.org/0000-0001-7557-6651): conceptualization, writing—original draft, writing—review and editing.
References
- Cohen AA, Kennedy BK, Anglas U, Bronikowski AM, Deelen J, Dufour F, et al. Lack of consensus on an aging biology paradigm? A global survey reveals an agreement to disagree, and the need for an interdisciplinary framework. Mech Ageing Dev. 2020. Oct;191:111316. 10.1016/j.mad.2020.111316 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo J, Huang X, Dou L, Yan M, Shen T, Tang W, et al. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther. 2022. Dec;7(1):391. 10.1038/s41392-022-01251-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mc Auley MT. Dietary restriction and ageing: recent evolutionary perspectives. Mech Ageing Dev. 2022. Dec;208:111741. 10.1016/j.mad.2022.111741 [DOI] [PubMed] [Google Scholar]
- Mc Auley MT. The evolution of ageing: classic theories and emerging ideas. Biogerontology. 2024. Oct;26(1):6. 10.1007/s10522-024-10143-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taormina G, Ferrante F, Vieni S, Grassi N, Russo A, Mirisola MG. Longevity: Lesson from Model Organisms. Genes (Basel). 2019. Jul;10(7):518. 10.3390/genes10070518 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brunet A. Old and new models for the study of human ageing. Nat Rev Mol Cell Biol. 2020. Sep;21(9):491–3. 10.1038/s41580-020-0266-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023. Jan;186(2):243–78. 10.1016/j.cell.2022.11.001 [DOI] [PubMed] [Google Scholar]
- Lazebnik Y. Can a biologist fix a radio?—Or, what I learned while studying apoptosis. Cancer Cell. 2002. Sep;2(3):179–82. 10.1016/S1535-6108(02)00133-2 [DOI] [PubMed] [Google Scholar]
- Sober E. Philosophy of biology. Routledge; 2018. 10.4324/9780429494871 [DOI] [Google Scholar]
- Wu Z, Zhang W, Qu J, Liu GH. Emerging epigenetic insights into aging mechanisms and interventions. Trends Pharmacol Sci. 2024. Feb;45(2):157–72. 10.1016/j.tips.2023.12.002 [DOI] [PubMed] [Google Scholar]
- Mc Auley MT, Mooney KM, Salcedo-Sora JE. Computational modelling folate metabolism and DNA methylation: implications for understanding health and ageing. Brief Bioinform. 2018. Mar;19(2):303–17. [DOI] [PubMed] [Google Scholar]
- Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14(10):R115. 10.1186/gb-2013-14-10-r115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell. 2013. Jan;49(2):359–67. 10.1016/j.molcel.2012.10.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horvath S, Haghani A, Macoretta N, Ablaeva J, Zoller JA, Li CZ, et al. DNA methylation clocks tick in naked mole rats but queens age more slowly than nonbreeders. Nat Aging. 2022. Jan;2(1):46–59. 10.1038/s43587-021-00152-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- SEER . Cancer Stat Facts: Cancer of Any Site [Internet]. 2024. [cited 2025 Sep 4]. Available from: https://seer.cancer.gov/statfacts/html/all.html
- National Center for Health Statistics . Respondent-reported prevalence of heart disease in adults aged 18 and over, by selected characteristics: United States, selected years 1997–2019 [Internet]. 2020. [cited 2025 Sep 4]. Available from: https://www.cdc.gov/nchs/data/hus/2020-2021/HDPrv.pdf
- WHO Ageing and health [Internet]. 2024. [cited 2025 Sep 4]. Available from: https://www.who.int/news-room/fact-sheets/detail/ageing-and-health
- Mc Auley MT. DNA methylation in genes associated with the evolution of ageing and disease: A critical review. Ageing Res Rev. 2021. Dec;72:101488. 10.1016/j.arr.2021.101488 [DOI] [PubMed] [Google Scholar]
- Lyko F. The DNA methyltransferase family: a versatile toolkit for epigenetic regulation. Nat Rev Genet. 2018. Feb;19(2):81–92. 10.1038/nrg.2017.80 [DOI] [PubMed] [Google Scholar]
- Zhang X, Zhang Y, Wang C, Wang X. TET (Ten-eleven translocation) family proteins: structure, biological functions and applications. Signal Transduct Target Ther. 2023. Aug;8(1):297. 10.1038/s41392-023-01537-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morgan AE, Davies TJ, Mc Auley MT. The role of DNA methylation in ageing and cancer. Proc Nutr Soc. 2018. Nov;77(4):412–22. 10.1017/S0029665118000150 [DOI] [PubMed] [Google Scholar]
- Nadiger N, Veed JK, Chinya Nataraj P, Mukhopadhyay A. DNA methylation and type 2 diabetes: a systematic review. Clin Epigenetics. 2024. May;16(1):67. 10.1186/s13148-024-01670-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang SC, Oelze B, Schumacher A. Age-specific epigenetic drift in late-onset Alzheimer’s disease. PLoS One. 2008. Jul;3(7):e2698. 10.1371/journal.pone.0002698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pérez RF, Alba-Linares JJ, Tejedor JR, Fernández AF, Calero M, Román-Domínguez A, et al. Blood DNA Methylation Patterns in Older Adults With Evolving Dementia. J Gerontol A Biol Sci Med Sci. 2022. Sep;77(9):1743–9. 10.1093/gerona/glac068 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carbonneau M, Li Y, Prescott B, Liu C, Huan T, Joehanes R, et al. Epigenetic Age Mediates the Association of Life’s Essential 8 With Cardiovascular Disease and Mortality. J Am Heart Assoc. 2024. Jun;13(11):e032743. 10.1161/JAHA.123.032743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu C, Wong EM, Joo JE, Hodge AM, Makalic E, Schmidt D, et al. Epigenetic Drift Association with Cancer Risk and Survival, and Modification by Sex. Cancers (Basel). 2021. Apr;13(8):1881. 10.3390/cancers13081881 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li G, Petkova TD, Laritsky E, Kessler N, Baker MS, Zhu S, et al. Early postnatal overnutrition accelerates aging-associated epigenetic drift in pancreatic islets. Environ Epigenet. 2019. Aug;5(3):dvz015. 10.1093/eep/dvz015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keshawarz A, Joehanes R, Ma J, Lee GY, Costeira R, Tsai PC, et al. Dietary and supplemental intake of vitamins C and E is associated with altered DNA methylation in an epigenome-wide association study meta-analysis. Epigenetics. 2023. Dec;18(1):2211361. 10.1080/15592294.2023.2211361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piyathilake CJ, Johanning GL, Macaluso M, Whiteside M, Oelschlager DK, Heimburger DC, et al. Localized folate and vitamin B-12 deficiency in squamous cell lung cancer is associated with global DNA hypomethylation. Nutr Cancer. 2000;37(1):99–107. 10.1207/S15327914NC3701_13 [DOI] [PubMed] [Google Scholar]
- Lai CQ, Parnell LD, Lee YC, Zeng H, Smith CE, McKeown NM, et al. The impact of alcoholic drinks and dietary factors on epigenetic markers associated with triglyceride levels. Front Genet. 2023. Feb;14:1117778. 10.3389/fgene.2023.1117778 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishida E, Nakamura M, Ikuta M, Shimada K, Matsuyoshi S, Kirita T, et al. Promotor hypermethylation of p14ARF is a key alteration for progression of oral squamous cell carcinoma. Oral Oncol. 2005. Jul;41(6):614–22. 10.1016/j.oraloncology.2005.02.003 [DOI] [PubMed] [Google Scholar]
- Lundgren S, Kuitunen S, Pietiläinen KH, Hurme M, Kähönen M, Männistö S, et al. BMI is positively associated with accelerated epigenetic aging in twin pairs discordant for body mass index. J Intern Med. 2022. Oct;292(4):627–40. 10.1111/joim.13528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langevin SM, Stone RA, Bunker CH, Grandis JR, Sobol RW, Taioli E. MicroRNA-137 promoter methylation in oral rinses from patients with squamous cell carcinoma of the head and neck is associated with gender and body mass index. Carcinogenesis. 2010. May;31(5):864–70. 10.1093/carcin/bgq051 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Światowy WJ, Drzewiecka H, Kliber M, Sąsiadek M, Karpiński P, Pławski A, et al. Physical Activity and DNA Methylation in Humans. Int J Mol Sci. 2021. Nov;22(23):12989. 10.3390/ijms222312989 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Slattery ML, Curtin K, Sweeney C, Levin TR, Potter J, Wolff RK, et al. Diet and lifestyle factor associations with CpG island methylator phenotype and BRAF mutations in colon cancer. Int J Cancer. 2007. Feb;120(3):656–63. 10.1002/ijc.22342 [DOI] [PubMed] [Google Scholar]
- Womersley JS, Nothling J, Toikumo S, Malan-Müller S, van den Heuvel LL, McGregor NW, et al. Childhood trauma, the stress response and metabolic syndrome: A focus on DNA methylation. Eur J Neurosci. 2022. May;55(9-10):2253–96. 10.1111/ejn.15370 [DOI] [PubMed] [Google Scholar]
- Blechter B, Cardenas A, Shi J, Wong JY, Hu W, Rahman ML, et al. Household air pollution and epigenetic aging in Xuanwei, China. Environ Int. 2023. Aug;178:108041. 10.1016/j.envint.2023.108041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tao L, Ge R, Xie M, Kramer PM, Pereira MA. Effect of trichloroethylene on DNA methylation and expression of early-intermediate protooncogenes in the liver of B6C3F1 mice. J Biochem Mol Toxicol. 1999;13(5):231–7. [DOI] [PubMed] [Google Scholar]
- Hoang TT, Lee Y, McCartney DL, Kersten ET, Page CM, Hulls PM, et al. BIOS Consortium . Comprehensive evaluation of smoking exposures and their interactions on DNA methylation. EBioMedicine. 2024. Feb;100:104956. 10.1016/j.ebiom.2023.104956 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Breitling LP, Yang R, Korn B, Burwinkel B, Brenner H. Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am J Hum Genet. 2011. Apr;88(4):450–7. 10.1016/j.ajhg.2011.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang YZ, Zhao W, Ammous F, Song Y, Du J, Shang L, et al. DNA Methylation Mediates the Association Between Individual and Neighborhood Social Disadvantage and Cardiovascular Risk Factors. Front Cardiovasc Med. 2022. May;9:848768. 10.3389/fcvm.2022.848768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McGuinness D, McGlynn LM, Johnson PC, MacIntyre A, Batty GD, Burns H, et al. Socio-economic status is associated with epigenetic differences in the pSoBid cohort. Int J Epidemiol. 2012. Feb;41(1):151–60. 10.1093/ije/dyr215 [DOI] [PubMed] [Google Scholar]
- Nannini DR, Cortese R, VonTungeln C, Hildebrandt GC. Chemotherapy-induced acceleration of DNA methylation-based biological age in breast cancer. Epigenetics. 2024. Dec;19(1):2360160. 10.1080/15592294.2024.2360160 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pietropaolo V, Prezioso C, Moens U. Role of Virus-Induced Host Cell Epigenetic Changes in Cancer. Int J Mol Sci. 2021. Aug;22(15):8346. 10.3390/ijms22158346 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toyooka S, Carbone M, Toyooka KO, Bocchetta M, Shivapurkar N, Minna JD, et al. Progressive aberrant methylation of the RASSF1A gene in simian virus 40 infected human mesothelial cells. Oncogene. 2002. Jun;21(27):4340–4. 10.1038/sj.onc.1205381 [DOI] [PubMed] [Google Scholar]
- Robinson N, Casement J, Gunter MJ, Huybrechts I, Agudo A, Barranco MR, et al. Anti-cancer therapy is associated with long-term epigenomic changes in childhood cancer survivors. Br J Cancer. 2022. Jul;127(2):288–300. 10.1038/s41416-022-01792-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guida F, Sandanger TM, Castagné R, Campanella G, Polidoro S, Palli D, et al. Dynamics of smoking-induced genome-wide methylation changes with time since smoking cessation. Hum Mol Genet. 2015. Apr;24(8):2349–59. 10.1093/hmg/ddu751 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fitzgerald KN, Hodges R, Hanes D, Stack E, Cheishvili D, Szyf M, et al. Potential reversal of epigenetic age using a diet and lifestyle intervention: a pilot randomized clinical trial. Aging (Albany NY). 2021. Apr;13(7):9419–32. 10.18632/aging.202913 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fiorito G, Caini S, Palli D, Bendinelli B, Saieva C, Ermini I, et al. DNA methylation-based biomarkers of aging were slowed down in a two-year diet and physical activity intervention trial: the DAMA study. Aging Cell. 2021. Oct;20(10):e13439. 10.1111/acel.13439 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maleknia M, Ahmadirad N, Golab F, Katebi Y, Haj Mohamad Ebrahim Ketabforoush A. DNA Methylation in Cancer: Epigenetic View of Dietary and Lifestyle Factors. Epigenet Insights. 2023. Sep;16:25168657231199893. 10.1177/25168657231199893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mc Auley MT. An evolutionary perspective of lifespan and epigenetic inheritance. Exp Gerontol. 2023. Aug;179:112256. 10.1016/j.exger.2023.112256 [DOI] [PubMed] [Google Scholar]
- Harney E, Paterson S, Collin H, Chan BH, Bennett D, Plaistow SJ. Pollution induces epigenetic effects that are stably transmitted across multiple generations. Evol Lett. 2022. Feb;6(2):118–35. 10.1002/evl3.273 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eberle C, Fasig T, Brüseke F, Stichling S. Impact of maternal prenatal stress by glucocorticoids on metabolic and cardiovascular outcomes in their offspring: A systematic scoping review. PLoS One. 2021. Jan;16(1):e0245386. 10.1371/journal.pone.0245386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zelko IN, Zhu J, Roman J. Maternal undernutrition during pregnancy alters the epigenetic landscape and the expression of endothelial function genes in male progeny. Nutr Res. 2019. Jan;61:53–63. 10.1016/j.nutres.2018.10.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Pollock CA, Saad S. Aberrant DNA Methylation Mediates the Transgenerational Risk of Metabolic and Chronic Disease Due to Maternal Obesity and Overnutrition. Genes (Basel). 2021. Oct;12(11):1653. 10.3390/genes12111653 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eberle C, Kirchner MF, Herden R, Stichling S. Paternal metabolic and cardiovascular programming of their offspring: A systematic scoping review. PLoS One. 2020. Dec;15(12):e0244826. 10.1371/journal.pone.0244826 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pastore A, Badolati N, Manfrevola F, Sagliocchi S, Laurenzi V, Musto G, et al. Pre-conceptional paternal diet impacts on offspring testosterone homoeostasis via epigenetic modulation of cyp19a1/aromatase activity. npj Metab Health Dis. 2024. Jun 17;2(1):8. [Google Scholar]
- De Rooij SR, Bleker LS, Painter RC, Ravelli AC, Roseboom TJ. Lessons learned from 25 Years of Research into Long term Consequences of Prenatal Exposure to the Dutch famine 1944-45: The Dutch famine Birth Cohort. Int J Environ Health Res. 2022. Jul;32(7):1432–46. 10.1080/09603123.2021.1888894 [DOI] [PubMed] [Google Scholar]
- Di Cesare M, Perel P, Taylor S, Kabudula C, Bixby H, Gaziano TA, et al. The Heart of the World. Glob Heart. 2024. Jan;19(1):11. 10.5334/gh.1288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krolevets M, Cate VT, Prochaska JH, Schulz A, Rapp S, Tenzer S, et al. DNA methylation and cardiovascular disease in humans: a systematic review and database of known CpG methylation sites. Clin Epigenetics. 2023. Mar;15(1):56. 10.1186/s13148-023-01468-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morgan AE, Mc Auley MT. Vascular dementia: from pathobiology to emerging perspectives. Ageing Res Rev. 2024. Apr;96:102278. 10.1016/j.arr.2024.102278 [DOI] [PubMed] [Google Scholar]
- Desiderio A, Pastorino M, Campitelli M, Longo M, Miele C, Napoli R, et al. DNA methylation in cardiovascular disease and heart failure: novel prediction models? Clin Epigenetics. 2024. Aug;16(1):115. 10.1186/s13148-024-01722-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruan T, Ling Y, Wu C, Niu Y, Liu G, Xu C, et al. Abnormal epigenetic modification of lysosome and lipid regulating genes in Alzheimer’s disease. J Alzheimers Dis. 2025. Apr;104(4):1185–200. 10.1177/13872877251322955 [DOI] [PubMed] [Google Scholar]
- Giannini LA, Boers RG, van der Ende EL, Poos JM, Jiskoot LC, Boers JB, et al. Distinctive cell-free DNA methylation characterizes presymptomatic genetic frontotemporal dementia. Ann Clin Transl Neurol. 2024. Mar;11(3):744–56. 10.1002/acn3.51997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harvey J, Imm J, Kouhsar M, Smith AR, Creese B, Smith RG, et al. Interrogating DNA methylation associated with Lewy body pathology in a cross brain-region and multi-cohort study. medRxiv. 2025. Mar 14;2025.03.13.25323837. 10.1101/2025.03.13.25323837 [DOI]
- Public Health England . Chapter 2: trends in mortality [Internet]. 2018. [cited 2025 Sep 4]. Available from: https://www.gov.uk/government/publications/health-profile-for-england-2018/chapter-2-trends-in-mortality#trends-in-age-specific-mortality-rates
- Shireby G, Dempster EL, Policicchio S, Smith RG, Pishva E, Chioza B, et al. DNA methylation signatures of Alzheimer’s disease neuropathology in the cortex are primarily driven by variation in non-neuronal cell-types. Nat Commun. 2022. Sep;13(1):5620. 10.1038/s41467-022-33394-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lang AL, Eulalio T, Fox E, Yakabi K, Bukhari SA, Kawas CH, et al. Methylation differences in Alzheimer’s disease neuropathologic change in the aged human brain. Acta Neuropathol Commun. 2022. Nov;10(1):174. 10.1186/s40478-022-01470-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin J, Yang S, Wang C, Yu E, Zhu Z, Shi J, et al. Prediction of Alzheimer’s Disease Using Patterns of Methylation Levels in Key Immunologic-Related Genes. J Alzheimers Dis. 2022;90(2):783–94. 10.3233/JAD-220701 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang AC, Buckingham L, Bishehsari F, Sutherland S, Ma K, Melson JE. Correlation of LINE-1 Hypomethylation With Size and Pathologic Extent of Dysplasia in Colorectal Tubular Adenomas. Clin Transl Gastroenterol. 2021. Jun;12(6):e00369. 10.14309/ctg.0000000000000369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhootra S, Jill N, Shanmugam G, Rakshit S, Sarkar K. DNA methylation and cancer: transcriptional regulation, prognostic, and therapeutic perspective. Med Oncol. 2023. Jan;40(2):71. 10.1007/s12032-022-01943-1 [DOI] [PubMed] [Google Scholar]
- Li P, Liu S, Du L, Mohseni G, Zhang Y, Wang C. Liquid biopsies based on DNA methylation as biomarkers for the detection and prognosis of lung cancer. Clin Epigenetics. 2022. Sep;14(1):118. 10.1186/s13148-022-01337-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duan R, Fu Q, Sun Y, Li Q. Epigenetic clock: A promising biomarker and practical tool in aging. Ageing Res Rev. 2022. Nov;81:101743. 10.1016/j.arr.2022.101743 [DOI] [PubMed] [Google Scholar]
- Hao Y, Han K, Wang T, Yu J, Ding H, Dao F. Exploring the potential of epigenetic clocks in aging research. Methods. 2024. Nov;231:37–44. 10.1016/j.ymeth.2024.09.001 [DOI] [PubMed] [Google Scholar]
- Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell. 2013. Jan;49(2):359–67. 10.1016/j.molcel.2012.10.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu AT, Quach A, Wilson JG, Reiner AP, Aviv A, Raj K, et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY). 2019. Jan;11(2):303–27. 10.18632/aging.101684 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yusipov I, Kalyakulina A, Trukhanov A, Franceschi C, Ivanchenko M. Map of epigenetic age acceleration: A worldwide analysis. Ageing Res Rev. 2024. Sep;100:102418. 10.1016/j.arr.2024.102418 [DOI] [PubMed] [Google Scholar]
- Lu AT, Fei Z, Haghani A, Robeck TR, Zoller JA, Li CZ, et al. Universal DNA methylation age across mammalian tissues. Nat Aging. 2023. Sep;3(9):1144–66. 10.1038/s43587-023-00462-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dec E, Clement J, Cheng K, Church GM, Fossel MB, Rehkopf DH, et al. Centenarian clocks: epigenetic clocks for validating claims of exceptional longevity. Geroscience. 2023. Jun;45(3):1817–35. 10.1007/s11357-023-00731-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martínez-Enguita D, Hillerton T, Åkesson J, Kling D, Lerm M, Gustafsson M. Precise and interpretable neural networks reveal epigenetic signatures of aging across youth in health and disease. Front Aging. 2025. Jan;5:1526146. 10.3389/fragi.2024.1526146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li ZP, Du Z, Huang DS, Teschendorff AE. Interpretable deep learning of single-cell and epigenetic data reveals novel molecular insights in aging. Sci Rep. 2025. Feb;15(1):5048. 10.1038/s41598-025-89646-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Higashimoto K, Hara S, Soejima H. DNA Methylation Analysis Using Bisulfite Pyrosequencing. Methods Mol Biol. 2023;2577:3–20. 10.1007/978-1-0716-2724-2_1 [DOI] [PubMed] [Google Scholar]
- Tost J, Gut IG. DNA methylation analysis by pyrosequencing. Nat Protoc. 2007;2(9):2265–75. 10.1038/nprot.2007.314 [DOI] [PubMed] [Google Scholar]
- Al Momani S, Rodger EJ, Stockwell PA, Eccles MR, Chatterjee A. Generating Sequencing-Based DNA Methylation Maps from Low DNA Input Samples. Methods Mol Biol. 2022;2458:3–21. 10.1007/978-1-0716-2140-0_1 [DOI] [PubMed] [Google Scholar]
- Noguera-Castells A, García-Prieto CA, Álvarez-Errico D, Esteller M. Validation of the new EPIC DNA methylation microarray (900K EPIC v2) for high-throughput profiling of the human DNA methylome. Epigenetics. 2023. Dec;18(1):2185742. 10.1080/15592294.2023.2185742 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flynn R, Washer S, Jeffries AR, Andrayas A, Shireby G, Kumari M, et al. Evaluation of nanopore sequencing for epigenetic epidemiology: a comparison with DNA methylation microarrays. Hum Mol Genet. 2022. Sep;31(18):3181–90. 10.1093/hmg/ddac112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taiwo O, Wilson GA, Morris T, Seisenberger S, Reik W, Pearce D, et al. Methylome analysis using MeDIP-seq with low DNA concentrations. Nat Protoc. 2012. Mar;7(4):617–36. 10.1038/nprot.2012.012 [DOI] [PubMed] [Google Scholar]
- Xing X, Zhang B, Li D, Wang T. Comprehensive Whole DNA Methylome Analysis by Integrating MeDIP-seq and MRE-seq. Methods Mol Biol. 2018;1708:209–46. 10.1007/978-1-4939-7481-8_12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gopalan-Nair R, Coissac A, Legrand L, Lopez-Roques C, Pécrix Y, Vandecasteele C, et al. Changes in DNA methylation contribute to rapid adaptation in bacterial plant pathogen evolution. PLoS Biol. 2024. Sep;22(9):e3002792. 10.1371/journal.pbio.3002792 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morgan AE, Acutt KD, Mc Auley MT. Electrochemically detecting DNA methylation in the EN1 gene promoter: implications for understanding ageing and disease. Biosci Rep. 2020. Nov;40(11):BSR20202571. 10.1042/BSR20202571 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sina AA, Carrascosa LG, Liang Z, Grewal YS, Wardiana A, Shiddiky MJ, et al. Epigenetically reprogrammed methylation landscape drives the DNA self-assembly and serves as a universal cancer biomarker. Nat Commun. 2018. Dec;9(1):4915. 10.1038/s41467-018-07214-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koowattanasuchat S, Ngernpimai S, Matulakul P, Thonghlueng J, Phanchai W, Chompoosor A, et al. Rapid detection of cancer DNA in human blood using cysteamine-capped AuNPs and a machine learning-enabled smartphone. RSC Adv. 2023. Jan;13(2):1301–11. 10.1039/D2RA05725E [DOI] [PMC free article] [PubMed] [Google Scholar]
- Battram T, Yousefi P, Crawford G, Prince C, Sheikhali Babaei M, Sharp G, et al. The EWAS Catalog: a database of epigenome-wide association studies. Wellcome Open Res. 2022. May;7:41. 10.12688/wellcomeopenres.17598.2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bao Y, Bai X, Bu C, Chen H, Chen H, Chen K, et al. CNCB-NGDC Members and Partners . Database Resources of the National Genomics Data Center, China National Center for Bioinformation in 2025. Nucleic Acids Res. 2025. Jan;53 D1:D30–44. 10.1093/nar/gkae978 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ragini S, Sakshi, Mani I, Singh V. Applications of bioinformatics in epigenetics. Prog Mol Biol Transl Sci. 2023;198:1–13. 10.1016/bs.pmbts.2023.03.023 [DOI] [PubMed] [Google Scholar]
- Haghani A, Li CZ, Robeck TR, Zhang J, Lu AT, Ablaeva J, et al. DNA methylation networks underlying mammalian traits. Science. 2023. Aug;381(6658):eabq5693. 10.1126/science.abq5693 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Navas-Acien A, Domingo-Relloso A, Subedi P, Riffo-Campos AL, Xia R, Gomez L, et al. Blood DNA Methylation and Incident Coronary Heart Disease: Evidence From the Strong Heart Study. JAMA Cardiol. 2021. Nov;6(11):1237–46. 10.1001/jamacardio.2021.2704 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crofts SJ, Latorre-Crespo E, Chandra T. DNA methylation rates scale with maximum lifespan across mammals. Nat Aging. 2024. Jan;4(1):27–32. 10.1038/s43587-023-00535-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W, Dong P, Li Y, Tang J, Liu S, Tu L, et al. Examining the potential causal relationships among smoking behaviors, blood DNA methylation profiles, and the development of coronary heart disease and myocardial infarction. Clin Epigenetics. 2024. Nov;16(1):173. 10.1186/s13148-024-01791-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoang DT, Shulman ED, Turakulov R, Abdullaev Z, Singh O, Campagnolo EM, et al. Prediction of DNA methylation-based tumor types from histopathology in central nervous system tumors with deep learning. Nat Med. 2024. Jul;30(7):1952–61. 10.1038/s41591-024-02995-8 [DOI] [PubMed] [Google Scholar]
- Chen X, Dong L, Chen L, Wang Y, Du J, Ma L, et al. Epigenome-wide development and validation of a prognostic methylation score in intrahepatic cholangiocarcinoma based on machine learning strategies. Hepatobiliary Surg Nutr. 2023. Aug;12(4):478–94. 10.21037/hbsn-21-424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu YT, Plets M, Morrison G, Cunha AT, Cen SY, Rhie SK, et al. Cell-free DNA Methylation as a Predictive Biomarker of Response to Neoadjuvant Chemotherapy for Patients with Muscle-invasive Bladder Cancer in SWOG S1314. Eur Urol Oncol. 2023. Oct;6(5):516–24. 10.1016/j.euo.2023.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hayflick L. Entropy explains aging, genetic determinism explains longevity, and undefined terminology explains misunderstanding both. PLoS Genet. 2007. Dec;3(12):e220. 10.1371/journal.pgen.0030220 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buescu J, Oliveira HM, Sousa M. Growth rate, evolutionary entropy and ageing across the tree of life. J Biol Dyn. 2023. Dec;17(1):2256766. 10.1080/17513758.2023.2256766 [DOI] [PubMed] [Google Scholar]
- Demetrius LA, Sahasranaman A, Ziehe M. Directionality theory and mortality patterns across the primate lineage. Biogerontology. 2024. Nov;25(6):1215–37. 10.1007/s10522-024-10134-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chmielewski PP. Human ageing as a dynamic, emergent and malleable process: from disease-oriented to health-oriented approaches. Biogerontology. 2020. Feb;21(1):125–30. 10.1007/s10522-019-09839-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olova N, Simpson DJ, Marioni RE, Chandra T. Partial reprogramming induces a steady decline in epigenetic age before loss of somatic identity. Aging Cell. 2019. Feb;18(1):e12877. 10.1111/acel.12877 [DOI] [PMC free article] [PubMed] [Google Scholar]
