Fact-checked by Grok 2 weeks ago

DNA damage theory of aging

The DNA damage theory of aging posits that the accumulation of unrepaired or imperfectly repaired DNA lesions over a lifetime is a fundamental driver of the aging process, leading to genomic instability, cellular dysfunction, , and eventual organismal decline. This theory emphasizes that DNA damage arises from both endogenous sources, such as and hydrolytic processes generating up to 10,000–100,000 lesions per per day, and exogenous factors like UV radiation and genotoxic agents. Imperfect repair mechanisms, including (BER) for oxidized bases and (NHEJ) for double-strand breaks, fail to fully mitigate this buildup, resulting in somatic mutations, chromosomal aberrations, and disrupted that underpin age-related phenotypes. Originally proposed in the late by physicists Giuseppe Failla and , who likened aging to the accumulation of akin to radiation-induced effects, the theory has evolved to encompass a broader spectrum of DNA damage beyond mere , including abasic sites, oxidized bases like , and interstrand crosslinks. Early formulations focused on target theory from radiation biology, suggesting that unrepaired hits to vital cellular components erode function over time. Subsequent refinements in the and beyond incorporated insights from pathways, highlighting how deficiencies in these systems accelerate aging. Compelling evidence supports the theory's core tenets, including meta-analyses of human studies demonstrating a significant age-related increase in DNA damage across tissues, with a correlation coefficient of r = 0.230 (95% CI: 0.111–0.342), influenced by factors like and assay techniques but independent of sex. In s, progeroid syndromes such as (due to WRN helicase mutations impairing ) and (NER and transcription-coupled repair defects) manifest accelerated aging features like neurodegeneration, skin atrophy, and shortened lifespan, directly linking repair failures to aging hallmarks. Animal models reinforce this: DNA repair-deficient mice, such as those lacking ERCC1 (involved in NER and interstrand crosslink repair), display premature frailty, , and renal dysfunction mirroring senescence. The consequences of persistent DNA damage extend beyond direct genomic instability, triggering protective cellular responses that paradoxically contribute to aging. For instance, double-strand breaks and oxidative lesions activate pathways leading to , where cells enter a permanent arrest state and secrete pro-inflammatory factors via the (SASP), promoting chronic inflammation and tissue remodeling dysfunction. Alternatively, severe damage induces , resulting in cell loss, particularly in post-mitotic tissues like neurons and cardiomyocytes, which exacerbates organ decline. These mechanisms intersect with other aging hallmarks, such as mitochondrial dysfunction (via mtDNA damage) and stem cell exhaustion, positioning DNA damage as a unifying causal factor. While the theory faces critiques—such as the observation that progeroid models exhibit segmental rather than comprehensive aging, and that DNA damage accumulation might sometimes result from rather than cause aging processes—diverse lines of evidence, including cancer survivor cohorts showing premature aging after genotoxic therapies, affirm its substantial role in modulating longevity. Interventions enhancing DNA repair, like caloric restriction which upregulates BER and NER, or pharmacological mimics of sirtuins that bolster genome stability, offer potential avenues for delaying age-related decline, though human translation remains challenging.

Fundamentals of the Theory

Historical Development

Building upon ideas by Giuseppe Failla and linking radiation-induced somatic mutations to aging, the DNA damage theory originated in the with early proposals linking radiation-induced DNA lesions to reduced lifespan. Peter Alexander first articulated this idea in 1967, positing that unrepaired DNA damage in somatic cells accumulates over time, leading to cellular dysfunction and organismal aging, distinct from heritable mutations. In the and , empirical studies advanced the theory by quantifying damage rates in animal models. Ronald W. Hart and Richard B. Setlow's work demonstrated a direct correlation between excision repair efficiency and maximum lifespan across mammalian species, including , suggesting that higher repair capacity limits damage accumulation and extends . Subsequent research by Hart in the measured age-related damage buildup in rodent organs, such as liver and , revealing species-specific patterns where shorter-lived exhibited faster accumulation rates, supporting models of progressive, repair-limited damage. The 1990s saw methodological breakthroughs enabling more direct assessment of DNA strand breaks in aging tissues. The , refined during this period, allowed single-cell quantification of DNA damage, with studies showing elevated strand breaks in lymphocytes and other tissues from older humans and animals compared to younger counterparts, confirming accumulation as a hallmark of aging. In the , syntheses of accumulating evidence refined the theory's focus. Harris Bernstein and Carol Bernstein emphasized that unrepaired DNA lesions, rather than fixed mutations, primarily drive aging through interference with transcription and replication, integrating prior findings into a cohesive prioritizing damage persistence.

Core Principles

The DNA damage theory of aging proposes that stochastic DNA damage, occurring at rates of approximately 10,000 to 100,000 lesions per per day, accumulates progressively over an organism's lifespan, eventually surpassing the capacity of systems and driving the decline in physiological function characteristic of aging. This accumulation arises from endogenous and environmental sources, leading to a gradual erosion of cellular integrity and contributing to age-related phenotypes such as reduced tissue regeneration and increased vulnerability to disease. A fundamental distinction of this theory is that aging stems not primarily from somatic mutations—alterations in the DNA sequence that may propagate during —but from unrepaired DNA lesions that persist and disrupt cellular through activation of DNA damage response (DDR) pathways. These lesions interfere with essential processes like transcription and replication without necessarily causing heritable sequence changes, thereby linking damage directly to functional impairment in both dividing and post-mitotic cells. Supporting evidence derives from model organisms, where DNA damage levels correlate inversely with lifespan independently of replication-associated errors; for example, in , mutants with enhanced exhibit extended , while repair-deficient strains show accelerated aging phenotypes. Similarly, in mice lacking key repair proteins like ERCC1, unrepaired damage accumulates in non-proliferative tissues, shortening lifespan without reliance on . Central to the theory is the concept of a "DNA damage threshold," at which persistent unrepaired lesions overwhelm repair mechanisms, triggering or to prevent propagation of compromised cells, thereby contributing to organismal aging. Conceptually, the rate of aging is proportional to the net rate of DNA damage accumulation minus repair efficiency, underscoring how imbalances in genotoxic stress and maintenance capacity dictate the pace of functional decline across species.

DNA Damage Mechanisms

Types and Sources of DNA Damage

DNA damage encompasses a variety of lesions that compromise the structural integrity and functional fidelity of the , arising from both internal cellular processes and external environmental factors. These damages are particularly relevant in the context of aging, as they challenge the cell's systems and contribute to progressive genomic instability. Endogenous sources, which originate within the cell, represent the primary drivers of routine DNA lesions, while exogenous sources introduce sporadic but potent insults. Among endogenous sources, (ROS) generated primarily by mitochondrial activity during aerobic metabolism are the chief culprits, producing approximately 10,000 to 100,000 oxidative lesions per human cell daily. These ROS oxidize DNA bases, notably converting guanine to 8-oxo-7,8-dihydroguanine (8-oxoG), a highly mutagenic that can lead to G-to-T transversions if unrepaired. Additionally, ROS attack the deoxyribose sugar backbone, generating single-strand breaks (SSBs) with fragmented ends that hinder . Replication errors during constitute another key endogenous source, where stalled or collapsed replication forks due to endogenous stresses like imbalances result in unrepaired lesions or mutations, exacerbating genomic instability over proliferative cycles in aging tissues. Exogenous sources introduce diverse lesions through environmental exposures. Ultraviolet (UV) radiation from sunlight primarily induces cyclobutane (CPDs) between adjacent or bases on the same DNA strand, distorting the helix and blocking replication and transcription. , such as from cosmic rays or , penetrates cells to produce densely ionizing tracks that cause clustered double-strand breaks (DSBs), the most lethal form of damage due to their propensity for chromosomal rearrangements. Specific types of DNA damage include base modifications, such as oxidative alterations like 8-oxoG and alkylations from endogenous methyl donors (e.g., S-adenosylmethionine) that add methyl groups to bases like or , impairing base pairing. Strand breaks encompass SSBs from ROS or replication fork collapse and DSBs from or oxidative clusters. Inter- and intra-strand crosslinks, often from endogenous aldehydes like or exogenous agents, covalently link DNA strands or bases, severely obstructing replication and repair. Additionally, telomere shortening due to the end-replication problem during cell divisions can lead to uncapped ends that activate DNA damage responses similar to DSBs. These lesions are mitigated by dedicated repair pathways, such as for oxidative and alkylated bases.

DNA Repair Pathways

Cells employ a suite of DNA repair pathways to counteract the accumulation of genomic damage, which is central to the DNA damage theory of aging, as unrepaired lesions contribute to cellular dysfunction and organismal decline. These pathways recognize specific types of damage and restore DNA integrity through coordinated enzymatic actions, with efficiency waning over time to exacerbate age-related pathologies. Key mechanisms include for small base modifications, for helix-distorting adducts, double-strand break repair via or , and mismatch repair for replication fidelity. Base excision repair (BER) primarily addresses oxidative damage and spontaneous base alterations, such as 8-oxoguanine from reactive oxygen species or deaminated bases. In this pathway, glycosylases like OGG1 excise the damaged base, creating an abasic site processed by APE1 endonuclease and subsequent polymerase and ligase activities to insert the correct nucleotide. BER capacity declines with age, leading to persistent single-strand breaks and increased mutagenesis, as observed in rodent models where BER enzyme levels and activity diminish in tissues like liver and brain. Poly(ADP-ribose) polymerase (PARP), a key sensor in BER, detects single-strand breaks and recruits repair factors, but its overactivation in aging cells can deplete cellular NAD+ and energy, promoting senescence. Nucleotide excision repair (NER) removes bulky, helix-distorting lesions, including cyclobutane induced by ultraviolet radiation. The process involves damage recognition by proteins like XPA and XPC, followed by unwinding via XPB and XPD helicases, excision of a 24-32 oligonucleotide by XPG and XPF-ERCC1 endonucleases, and gap-filling by and ligation. Deficiencies in NER, as exemplified by , are linked to premature aging phenotypes due to unchecked accumulation of photoproducts and transcriptional blockade. Age-associated reductions in NER efficiency further impair the removal of oxidative adducts in non-dividing cells, contributing to genomic instability. Double-strand breaks (DSBs), arising from or replication fork collapse, pose severe threats and are repaired by two main pathways: (HR) and (NHEJ). HR, active in S and G2 phases, uses a sister chromatid template for error-free repair, involving resection by MRN complex (MRE11-RAD50-NBS1), strand invasion mediated by RAD51, and synthesis by polymerases. In contrast, NHEJ predominates in G1 and ligates broken ends directly via Ku70/80, , and ligase IV, often introducing small insertions or deletions. Both pathways decline with age; for instance, NHEJ accuracy decreases in senescent cells, while HR efficiency drops due to reduced RAD51 loading, leading to persistent DSBs and chromosomal aberrations. () kinase senses DSBs, phosphorylating H2AX to form γ-H2AX foci that recruit repair machinery. Mismatch repair (MMR) corrects base mismatches and small insertion/deletion loops arising during , preventing somatic mutations. MutSα (MSH2-MSH6) or MutSβ (MSH2-MSH3) recognizes errors on the nascent strand, recruiting MutLα (MLH1-PMS2) for strand-specific incision, flap removal, and resynthesis. While primarily post-replicative, MMR also contributes to repairing certain oxidative lesions in cooperation with BER. Age-related MMR impairment, evidenced by increased in elderly tissues, heightens mutation rates and links to clonal expansion in aging organs. The DNA damage response integrates these pathways through central regulators like , which, upon or ATR activation, halts the to allow repair or triggers if damage is irreparable. As a , upregulates genes for repair proteins and inhibitors like p21. When primary repair fails, translesion synthesis (TLS) serves as an error-prone backup, enabling replication past lesions using specialized polymerases like POLη for UV dimers. TLS introduces mutations if not tightly regulated, contributing to the load in aging cells where high-damage burdens overwhelm accurate pathways. This tolerance mechanism preserves viability but accelerates genomic heterogeneity, a hallmark of aged tissues.

Accumulation of DNA Damage in Aging

General Patterns of Accumulation

DNA damage accumulates progressively over the lifespan, with unrepaired lesions exhibiting a marked increase post-maturity, often described as exponential in contexts such as populations where genomic instability accelerates. This buildup is quantified using sensitive assays, including the alkaline , which detects single- and double-strand breaks through DNA migration in an , and γ-H2AX foci counting, a marker of double-strand break induction that persists in aging cells. Steady-state levels of oxidative lesions, such as , rise 2- to 4-fold in various mammalian tissues from young to old, with some reports indicating up to 10-fold elevations in damage across species. attrition serves as a key cumulative marker, reflecting repeated oxidative insults and replication stress that shorten protective caps, leading to persistent DNA damage signals. The primary drivers of this accumulation include declining efficiency, particularly in pathways, which falter with age due to reduced activity in tissues like the and liver. Concurrently, mitochondrial dysfunction elevates (ROS) production, exacerbating oxidative lesions in both and mitochondrial genomes, as mtDNA damage levels can increase tenfold compared to nuclear DNA in aging humans. These factors create a feedback loop where unrepaired damage impairs mitochondrial function further, amplifying ROS and repair deficits. Cross-species comparisons reveal faster accumulation rates in than in humans, correlating with shorter lifespans; for instance, oxidative DNA damage in brain and muscle rises 2- to 3-fold over 18-24 months, while human tissues show more gradual increases over decades. Recent studies from 2024-2025 confirm this pattern in non-dividing cells, where mutagenic lesions and persistent double-strand breaks endure for months to years, generating mutations during occasional replications and contributing to systemic aging. Tissue-specific variations exist, but the overall systemic rise underscores DNA damage as a hallmark of organismal aging.

Impacts on Gene Expression

Accumulated DNA damage in aging disrupts normal by inducing transcriptional silencing at promoter regions, primarily through modifications and the formation of repair foci that hinder transcription machinery. DNA double-strand breaks and oxidative lesions trigger the recruitment of repair proteins, such as those in the pathway, which form persistent foci that physically block (Pol II) progression, leading to reduced transcription of nearby genes. Additionally, DNA damage activates modifications like of H2AX (γ-H2AX) and changes that compact , silencing promoters and preventing access by transcriptional activators. These mechanisms contribute to the progressive loss of gene expression fidelity observed in aging cells, distinct from the general buildup of damage lesions. In response to persistent DNA damage, cells upregulate stress response genes, including p21 (CDKN1A) and components of the (SASP), which promote and alter the extracellular environment. The (DDR) pathway, mediated by ATM/ATR kinases, activates , which transcriptionally induces p21 to enforce arrest and prevent propagation of damaged genomes. SASP factors, such as IL-6 and IL-8, are similarly upregulated via signaling triggered by chronic DDR, fostering a pro-inflammatory secretome that reinforces but can drive age-related pathologies through paracrine effects. This upregulation shifts cellular priorities toward survival and repair at the expense of normal proliferative and metabolic functions. Oxidative DNA damage, a prevalent form in aging, induces shifts in patterns, resulting in global hypomethylation alongside hypermethylation at specific CpG sites, which dysregulates . cause 8-oxoguanine lesions that interfere with activity, leading to widespread demethylation of repetitive elements and transposons, potentially activating retrotransposons and genomic instability. Conversely, promotes hypermethylation at promoter CpG islands of tumor suppressor genes and aging-related loci, silencing their expression through recruitment of repressive complexes like polycomb. These epigenetic alterations amplify the transcriptional impacts of damage, creating a feedback loop that accelerates aging hallmarks. Chromatin immunoprecipitation (ChIP) studies provide direct evidence that DNA damage correlates with reduced Pol II occupancy at gene promoters and bodies, indicating stalled transcription elongation. In aging models, analyses reveal decreased Pol II loading on actively transcribed genes due to DDR-induced pausing, with lesions causing genome-wide transcriptional stress. For instance, UV-induced damage or endogenous oxidative breaks diminish Pol II processivity, as quantified by reduced signal in ChIP assays, linking damage accumulation to the deterioration in senescent cells. Recent 2025 epigenetic studies further link DNA damage to the loss of stability, exacerbating dysregulation. Research demonstrates that double-strand breaks in heterochromatic regions trigger ubiquitin-mediated spreading of repressive marks, but unresolved damage leads to erosion of boundaries, destabilizing silent domains and permitting ectopic activation. Another study highlights how DDR-induced of variants relaxes heterochromatin for repair access, yet chronic damage causes persistent instability, correlating with age-related epigenetic drift. These findings underscore how damage compromises the structural integrity of compartments essential for balanced .

Brain

The brain exhibits particular vulnerability to DNA damage accumulation during aging due to the post-mitotic nature of its neurons, which cannot dilute damage through , and their high metabolic rate coupled with elevated oxygen consumption that generates (ROS). This oxidative environment promotes the formation of oxidized DNA lesions, such as (8-oxoG), which accumulate progressively over decades in neural tissues. Post-mitotic neurons rely heavily on robust mechanisms to maintain genomic integrity, but age-related declines in these pathways exacerbate damage persistence. Postmortem analyses of human brains reveal significant increases in oxidative DNA damage markers with aging, including elevated levels of 8-oxo-dG in both nuclear and mitochondrial DNA, as well as higher frequencies of single- and double-strand breaks, particularly in regions like the cortex and hippocampus. These lesions correlate with cognitive decline, as evidenced by reduced performance in memory and executive function tasks in older individuals with higher DNA oxidation burdens. Strand breaks, often resulting from unrepaired oxidative insults, further contribute to genomic instability and are more prevalent in aged neural tissue compared to other organs. Studies indicate declining DNA repair efficiency in the contributes to vulnerability in (AD) and (PD), with persistent double-strand breaks (DSBs) and oxidative lesions accelerating neuronal dysfunction. In AD, DNA repair deficits in hippocampal neurons correlate with , while in PD, declines in repair capacity in neurons of the are observed. A 2025 study suggests the brain's immune response to DNA damage may drive Alzheimer's progression through faulty repair attempts. Additionally, aging neurons exhibit selective repair prioritization, protecting essential genes involved in at the expense of others, potentially altering patterns associated with brain function. Unrepaired DSBs in neurons trigger as a protective mechanism against , leading to progressive neuronal loss in regions lacking significant renewal, such as the and most hippocampal areas. Unlike proliferative tissues, adult neurogenesis is limited to specific niches like the , leaving the majority of post-mitotic neurons irreplaceable and amplifying the impact of accumulated DNA damage on overall function. This selective contributes to age-related and functional decline without compensatory regeneration in non-neurogenic zones.

Muscle

In skeletal and cardiac muscle, the accumulation of DNA damage contributes significantly to age-related functional decline, particularly through oxidative insults that impair cellular energetics and structural integrity. Mitochondrial (ROS), generated during respiration, preferentially damage (mtDNA) due to its proximity to the and lack of protection, leading to point mutations and deletions that disrupt ATP synthesis. This mtDNA dysfunction reduces complex activity by 35-50%, resulting in ATP levels dropping to approximately 38% of normal in affected muscle fibers, which compromises contractile function and promotes fiber . In skeletal muscle, this process underlies , the progressive loss of mass and strength, while in cardiac muscle, it exacerbates age-related by impairing and increasing susceptibility to ischemia. Evidence from experimental models demonstrates elevated DNA strand breaks in aged muscle. Comet assays in rodents reveal higher levels of DNA single- and double-strand breaks in nuclei from aged mice compared to young controls, correlating with increased markers like . Human vastus lateralis biopsies from individuals aged 25-93 years confirm DNA damage accumulation, with 8-hydroxy-2'-deoxyguanosine (8-OHdG) levels rising approximately 5-fold from younger to older groups (from 3.28 to 21.10 per 10^5 dG bases), alongside a 25% decline in mtDNA abundance that parallels reduced ATP production. Telomere shortening in skeletal muscle myofibers further exacerbates aging pathology, with oxidative stress accelerating erosion in isolated fibers, leading to chromosomal end-to-end fusions and genomic instability that manifest as myofiber weakness and impaired regeneration. This shortening, observed in elderly human skeletal muscle compared to young controls, disrupts myonuclear organization and contributes to fusion events that compromise fiber integrity and contractile efficiency. Additionally, non-homologous end joining (NHEJ), a key double-strand break repair pathway, declines in efficacy with age; in aged skeletal muscle, NHEJ activation post-exercise is delayed, resulting in persistent breaks that heighten vulnerability to exercise-induced damage and reduce recovery. DNA damage accumulation in muscle accelerates under conditions of inactivity, forming a vicious cycle that promotes frailty. Disuse atrophy in aged rodents amplifies mtDNA mutations and ROS production, leading to rapid energetic decline and a 20-30% loss in muscle mass within weeks, mirroring accelerated sarcopenia and contributing to systemic frailty through diminished mobility and metabolic dysregulation.

Liver

The liver, serving as the principal organ for detoxification, experiences heightened exposure to reactive oxygen species (ROS) and environmental toxins during metabolic processing, resulting in substantial accumulation of DNA damage, particularly in mitochondrial DNA (mtDNA). This mtDNA damage disrupts oxidative phosphorylation and energy production, thereby impairing gluconeogenesis and contributing to metabolic dysregulation in aging hepatic tissue. In aged livers, specific lesions such as abasic (apurinic/apyrimidinic) sites and DNA crosslinks increase significantly, reflecting chronic oxidative stress and repair inefficiencies. These DNA damages are mechanistically linked to pathological outcomes, including the progression of liver fibrosis through activation of stellate cells and extracellular matrix remodeling, as well as reduced regenerative capacity due to impaired following injury. Studies in models demonstrate that aged livers exhibit heightened fibrotic responses to toxins like , correlating with elevated DNA lesion burdens that hinder timely repair and restoration. In models of , recent investigations highlight exhaustion of the (BER) pathway, which is critical for addressing oxidative base modifications and abasic sites; this impairment exacerbates lipid accumulation and by failing to mitigate ROS-induced genomic instability. For instance, BER-deficient hepatocytes show increased susceptibility to overload, linking repair deficits directly to metabolic dysfunction in non-alcoholic fatty liver disease (NAFLD). As a compensatory response to persistent DNA damage, hepatocytes undergo polyploidization, increasing their ploidy levels to buffer against oxidative insults and maintain functional genome copies, though this adaptation promotes and restricts proliferative renewal in aging livers. Polyploid hepatocytes thus act as a double-edged , enhancing short-term but limiting long-term regenerative potential and contributing to age-related hepatic decline.

Kidney

In the , DNA damage accumulates preferentially in glomerular and epithelial cells, contributing to the progressive decline in renal function observed during aging. (ROS) generated from hemodynamic stress and induce oxidative damage, leading to DNA strand breaks in these structures. This damage manifests as glomerular sclerosis and atrophy, with strand breaks in podocytes and cells promoting by disrupting the barrier and epithelial integrity. For instance, II-induced has been shown to elevate DNA double-strand breaks in renal tissues, exacerbating in experimental models. Histological and molecular evidence supports the accumulation of unrepaired DNA damage in aged kidneys. Levels of γ-H2AX, a marker of DNA double-strand breaks, are significantly elevated in renal biopsies from older individuals and animal models of aging, indicating persistent genomic instability in glomerular and tubular compartments. Human biopsy studies further reveal a decline in DNA repair capacity with age, as evidenced by reduced expression of repair proteins and increased persistence of damage foci in proximal tubular epithelial cells. This repair deficiency correlates with broader patterns of DNA damage buildup across aging tissues, but is particularly pronounced in the kidney due to its high metabolic demands and exposure to systemic toxins. Mitochondrial DNA (mtDNA) mutations play a specific role in driving progression amid aging-related damage. Somatic mtDNA mutations accumulate in renal cells, impairing mitochondrial function and amplifying ROS production, which further perpetuates nuclear DNA damage in the context of . Podocytes, the terminally differentiated cells of the glomerular filtration barrier, exhibit heightened sensitivity to this damage; and mtDNA alterations trigger and detachment, accelerating and . Studies in podocyte-specific models demonstrate that DNA damage response activation, such as via loss, leads to sustained double-strand breaks and functional impairment in these cells. The extent of DNA damage in the kidney correlates with clinical biomarkers of renal aging, notably the rise in serum levels, which reflects declining . In patients with early , increased oxidative DNA lesions and unrepaired breaks in tubular cells are associated with accelerated creatinine elevation and disease progression. This linkage underscores DNA damage as a mechanistic driver of age-related loss, independent of other comorbidities.

Stem Cells

Quiescent stem cells, such as hematopoietic stem cells (HSCs) and neural stem cells (NSCs), accumulate DNA damage over time primarily due to their low replication rates, which limit opportunities for repair during active cell cycling, coupled with exposure to high levels of reactive oxygen species (ROS) generated by mitochondrial activity in the niche environment. In HSCs, this damage includes persistent double-strand breaks and oxidative lesions that remain unrepaired in the G0 phase, leading to functional impairments upon eventual activation. Similarly, quiescent NSCs in the adult brain subventricular zone experience ROS-induced DNA lesions, contributing to their diminished proliferative capacity during aging. A key protective mechanism in these stem cells is asymmetric cell division, which segregates damaged DNA or cellular components preferentially to the differentiating daughter cell, thereby preserving the integrity of the self-renewing progenitor while directing compromised material toward terminal fates. This process helps mitigate stem cell exhaustion but ultimately dooms differentiated progeny to senescence or apoptosis, exacerbating tissue decline. Additionally, telomere shortening triggers crisis in aging stem cells, where critically short telomeres cause replicative stress and genomic instability upon division, further depleting the stem cell pool and impairing regenerative functions. Recent 2024 reviews highlight how persistent DNA breaks in stem cells drive through sustained activation of the DNA damage response, including p53-mediated pathways that enforce arrest and secretory phenotypes detrimental to . In HSCs, this accumulation correlates directly with reduced hematopoiesis in aging, manifesting as and immune dysregulation due to biased myeloid output and lowered lymphoid reconstitution. Embryonic stem cells, in contrast, exhibit enhanced DNA repair efficiency via robust and pathways to safeguard pluripotency, but this capacity declines in with age, progressively limiting their regenerative potential and contributing to organismal aging.

Relation to Mutations and Other Theories

DNA Damage vs. Somatic Mutations

The DNA damage theory of aging posits that the accumulation of various forms of DNA lesions, rather than solely heritable sequence alterations, drives age-related decline. Mutations represent only a minor subset of DNA damage outcomes, a small fraction of base modifications from endogenous damaging agents typically converting into permanent mutations during replication. The theory instead highlights non-mutagenic lesions, such as double-strand breaks or oxidative adducts, which can trigger cellular responses like , , or transcriptional dysregulation without altering the DNA sequence. For instance, unrepaired breaks may lead to chromosomal instability or , contributing to tissue atrophy independently of . Empirical evidence underscores that somatic mutation rates are too low to account for the widespread functional deterioration in aging. In humans, the somatic mutation rate averages around 47 single-base substitutions per cell per year, corresponding to approximately 1.5 × 10^{-8} mutations per per year, which accumulates to only thousands of changes over a lifetime—insufficient to disrupt the vast majority of the 3 billion base pairs or explain systemic aging phenotypes. In contrast, cells experience up to 70,000 DNA lesions daily from endogenous sources like , most of which are transiently repaired but can cause immediate or epigenetic perturbations if unresolved. Recent genomic sequencing studies reinforce this distinction, showing that age-related mutations do not correlate strongly with aging hallmarks, whereas persistent DNA damage does. A key illustration comes from , a premature aging disorder caused by defects in transcription-coupled . Patients exhibit accelerated aging features, including neurodegeneration and , due to unrepaired DNA lesions that impair transcription and induce , yet without an elevated somatic mutation burden compared to healthy individuals. This dissociation highlights how repair deficiencies amplify damage signaling and cellular dysfunction beyond mutagenesis. The DNA damage theory thus offers a broader than mutation-centric models, encompassing epigenetic alterations—such as modifications or changes at damage sites—that sequencing-based detection cannot capture. These epigenetic shifts, induced by persistent lesions, propagate age-related errors across cell generations. For example, oxidative DNA damage can lead to heritable that silences genes without sequence changes, further differentiating the theory's scope.

Integration with Broader Aging Theories

The DNA damage theory of aging intersects significantly with the free radical theory, originally proposed by Denham Harman in , which posits that (ROS) generated during metabolism cause cumulative cellular damage leading to aging. While the free radical theory emphasizes ROS as the primary agents of across biomolecules, the DNA damage perspective refines this by highlighting the specific consequences on and (mtDNA), where unrepaired lesions accumulate and impair cellular function over time. For instance, ROS-induced strand breaks and base modifications in mtDNA exacerbate bioenergetic decline, linking oxidative insults directly to genomic instability as a driver of . Telomere attrition represents another form of DNA damage that aligns the theory with telomere biology paradigms, where progressive shortening of chromosome ends during cell divisions eventually uncaps telomeres, eliciting DNA damage responses akin to double-strand breaks. This triggers pathways such as p53 activation and cellular senescence, amplifying age-related dysfunction across tissues. Furthermore, the senescence-associated secretory phenotype (SASP) induced by such damage promotes chronic low-grade inflammation (inflammaging), which in turn generates additional ROS and DNA lesions in neighboring cells, creating a deleterious feedback loop that accelerates tissue degeneration. Within the hallmarks of aging framework, established by López-Otín et al. in 2013 and updated in 2023, genomic instability—encompassing DNA damage accumulation—is positioned as a primary hallmark that causally contributes to secondary processes like epigenetic alterations and loss of proteostasis. This integration underscores DNA damage as a central nexus interconnecting multiple aging mechanisms. Recent 2025 studies further bridge this to metabolic theories, demonstrating that persistent DNA damage mediates declines in insulin signaling; for example, activation of the DNA damage response via ATM kinase suppresses insulin receptor expression and promotes neuronal insulin resistance, linking genomic insults to dysregulated nutrient sensing and age-related metabolic disorders.

Genetic Evidence from Defects and Enhancements

DNA Repair Defects and Premature Aging Syndromes

Premature aging syndromes arise from inherited defects in DNA repair pathways, providing compelling genetic evidence that unrepaired DNA damage accelerates aging-like phenotypes in humans. These disorders, such as , Hutchinson-Gilford progeria syndrome (HGPS), and , manifest symptoms including skin atrophy, hair graying, cardiovascular disease, and neurodegeneration, often with drastically shortened lifespans. Unlike typical aging, these syndromes exhibit segmental progeria, affecting specific tissues while sparing others, and they highlight how impaired repair leads to persistent DNA lesions that trigger without necessarily increasing cancer risk in all cases. Werner syndrome, caused by biallelic mutations in the WRN gene encoding a RecQ essential for (HR) and other repair processes, exemplifies direct deficiency driving premature aging. The WRN protein facilitates double-strand break repair by promoting canonical and suppressing error-prone alternatives, while also participating in ; its loss results in genomic instability, attrition, and accumulation of unrepaired damage that induces through disruption. Patients typically develop gray hair, cataracts, skin ulcers, , , and in their 20s–30s, with death often from or cardiovascular events in their 40s–50s, underscoring the role of unrepaired lesions in systemic decline. Hutchinson-Gilford progeria syndrome (HGPS) involves an indirect link to DNA repair through a dominant LMNA mutation (c.1824C>T) producing progerin, a farnesylated form of lamin A that disrupts nuclear architecture. Progerin causes chromatin perturbations, replication fork stalling, and sequestration of repair factors like RAD51 and 53BP1, leading to persistent double-strand breaks (DSBs) marked by elevated γH2AX foci and activated ATM/ATR checkpoints, which promote replicative arrest and premature senescence. Clinical features include alopecia with sparse gray hair, scleroderma-like skin changes, joint contractures, and severe atherosclerosis, with an average lifespan of about 14 years, primarily due to myocardial infarction; notably, HGPS lacks cancer predisposition despite genomic instability. Recent 2024 studies confirm progerin induces DNA damage independently of replication changes, elevating markers like DSBs in patient-derived cells and reinforcing its role in accelerated aging. Cockayne syndrome, resulting from mutations in ERCC6 (CSB) or ERCC8 () genes that impair transcription-coupled (TC-NER), demonstrates how unrepaired oxidative and UV-induced lesions in transcribed genes cause neurodegeneration and . Defective TC-NER leads to transcription-replication conflicts, in post-mitotic neurons, and systemic DNA damage accumulation, triggering without elevated or cancer risk—unlike , where global NER defects predominate. Symptoms encompass progressive , , , gray hair, retinal degeneration, and , with contributing to failure-to-thrive; lifespans vary by subtype but average 10–20 years, often ending in neurological decline. Case studies, including 2022 analyses of siblings with identical CSA mutations, reveal heterogeneous yet consistent neurodegeneration and elevated damage markers, affirming TC-NER's critical role in preventing premature aging phenotypes.

Enhanced DNA Repair and Increased Longevity

Genetic engineering to enhance mechanisms has provided compelling evidence supporting the role of efficient damage repair in extending lifespan. In mice, overexpression of the sirtuin 6 (SIRT6) gene, which promotes () and () of double-strand breaks, results in a significant increase in median lifespan—approximately 27% in males and 15% in females—accompanied by reduced genomic instability and improved metabolic . Similarly, transgenic mice overexpressing human MTH1 (hMTH1), a enzyme in the pathway that hydrolyzes oxidized triphosphates to prevent their incorporation into , exhibit prolonged lifespan with lower levels of oxidative DNA lesions, enhanced exploratory behavior in old age, and resistance to age-related cognitive decline. In lower organisms, upregulation of DNA repair genes similarly confers resistance to aging stressors and extends lifespan. In the nematode , long-lived mutants with enhanced (NER) capacity, such as those with increased expression of repair factors, show improved survival under oxidative and UV stress, correlating with delayed aging phenotypes. In budding yeast (), overexpression of regulators like Rpn4, which upregulates multiple DNA repair pathways including NER and BER, leads to elevated activity and replicative lifespan extension by mitigating accumulation of DNA damage during aging. In humans, genetic variations in DNA repair genes correlate with healthy aging outcomes. Polymorphisms in the OGG1 gene, encoding a key BER glycosylase for removing lesions from oxidative damage, have been associated with increased in elderly cohorts; specifically, favorable alleles in OGG1, often in combination with variants in other repair genes like XPD and XRCC1, are more frequent in long-lived individuals over 70, suggesting enhanced oxidative damage repair contributes to . These observations across species underscore the DNA damage theory by linking superior repair capacity to extended healthspan and lifespan.

Interventions Modulating DNA Damage

Dietary Restriction Effects

Dietary restriction (DR), often implemented as without , mitigates DNA damage accumulation in aging through multiple interconnected mechanisms. By reducing caloric intake, DR lowers the production of (ROS) from mitochondrial respiration, thereby decreasing oxidative insults to DNA. This reduction in ROS is linked to enhanced mitochondrial efficiency and biogenesis, which minimizes the generation of free radicals that cause base modifications like in DNA. Additionally, DR promotes , a process that selectively degrades damaged mitochondria (mitophagy), preventing the persistence of ROS-producing organelles and further protecting genomic integrity. Upregulation of (SIRT1), a NAD+-dependent deacetylase, plays a central role by deacetylating key DNA repair proteins such as Ku70, enhancing for double-strand break repair, and modulating to balance cell survival and in response to damage. In models, DR consistently demonstrates reduced DNA damage and extended lifespan, supporting its role in the DNA damage theory of aging. Long-term DR in rats, typically 30-40% below feeding, decreases mitochondrial production by approximately 45% and oxidative damage to by 30% in cardiac tissue, correlating with slower accumulation of age-related lesions across organs. These interventions significantly extend mean lifespan in mice and rats, with indicating 30-50% less overall DNA damage buildup in restricted animals compared to controls, as measured by markers like 8-oxo-dG in multiple tissues. Such effects are attributed to the combined ROS reduction and repair enhancement, as DR upregulates pathways that address oxidative lesions. Intermittent fasting, a variant of DR, similarly curbs DNA damage by mimicking caloric scarcity and activating repair pathways. In mice, alternate-day fasting reduces 8-oxo-dG levels in brain, liver, and kidney tissues, reflecting lower oxidative stress and improved mitochondrial turnover via autophagy. Human studies echo these findings; the CALERIE trial, involving 25% calorie restriction over two years, showed significant reductions in DNA damage markers in blood cells after six months, alongside decreased cellular senescence biomarkers in recent analyses. These outcomes suggest DR's potential to slow aging-related DNA damage in non-rodent species, though long-term human data remain limited.

Pharmacological and Reprogramming Approaches

Pharmacological interventions targeting DNA damage in aging primarily focus on boosting NAD+ levels to support repair pathways and eliminating senescent cells that accumulate due to unrepaired damage. NAD+ precursors such as nicotinamide mononucleotide (NMN) elevate intracellular NAD+ concentrations, which are essential for poly(ADP-ribose) polymerase 1 (PARP1)-mediated DNA repair and base excision repair (BER) mechanisms that counteract oxidative and other age-related lesions. With advancing age, NAD+ depletion impairs these processes, exacerbating genomic instability, but NMN supplementation in mouse models has been shown to mitigate DNA strand breaks and restore repair efficiency, thereby delaying age-associated decline. Similarly, senolytics like dasatinib combined with quercetin selectively induce apoptosis in DNA damage-induced senescent cells, which secrete pro-inflammatory factors that propagate further damage. In aged mice, senolytic treatment reduces senescent cell burden in tissues such as endothelium, lowering DNA damage markers and telomere dysfunction while improving vascular healthspan. These approaches parallel the DNA repair enhancements observed in dietary restriction but offer targeted, drug-based alternatives. Cellular reprogramming strategies represent a transformative intervention by reversing epigenetic alterations tied to accumulated DNA damage, without fully dedifferentiating cells into pluripotency. Partial reprogramming using subsets of Yamanaka factors, such as Oct4, Sox2, and Klf4 (OSK), transiently activates youthful gene expression patterns that bolster DNA repair capacity and reduce epigenetic aging clocks. In progeroid mouse models like Lmna-/- (LAKI), cyclic OSK induction extends median lifespan by approximately 40% and maximal lifespan by 32%, while decreasing DNA double-strand breaks and restoring nuclear architecture. These effects stem from OSK's ability to reactivate endogenous repair pathways, including non-homologous end joining, without tumorigenic risks associated with full reprogramming. Preclinical studies in Hutchinson-Gilford progeria syndrome (HGPS) models further demonstrate that short-term OSK expression ameliorates cellular hallmarks like nuclear blebbing and genomic instability, paving the way for potential human applications. Chemical-based reprogramming offers a non-genetic alternative, using small-molecule cocktails to mimic OSK effects and address DNA damage more safely and scalably. Optimized chemical mixtures, such as those combining valproic acid, CHIR99021, and RepSox, induce partial rejuvenation in human fibroblasts within days, restoring transcriptomic youthfulness and reducing markers of genomic instability. In aged mice, these cocktails extend lifespan by up to 30% and improve healthspan by enhancing mitochondrial function and DNA repair, with minimal off-target effects. Recent 2025 research highlights reprogramming's relevance to visible aging signs, such as gray hair, where DNA damage in melanocyte stem cells triggers a protective clearance mechanism that depletes pigment progenitors; partial reprogramming could potentially reverse this by mobilizing undamaged stem cells and restoring melanin production. Preclinical studies in progeria models show promise, with human trials for partial reprogramming in aging-related conditions entering early phases as of 2025, focusing on safety and epigenetic biomarkers. As of October 2025, the FDA has cleared the first human trial for partial reprogramming targeting neurodegeneration, highlighting progress toward clinical translation for DNA damage-related aging interventions.

Comparative Biology Across Species and Populations

DNA Repair Capacity in Mammals

Mammals exhibit significant variation in capacity, which correlates with maximum lifespan differences across species. Long-lived species generally demonstrate more efficient mechanisms compared to short-lived ones, supporting the DNA damage theory of aging by suggesting that superior repair delays the accumulation of genomic damage over time. For instance, liver tissue from long-lived mammals shows higher expression of genes compared to short-lived species, establishing a positive association between repair proficiency and . The (Heterocephalus glaber), with a maximum lifespan of approximately 30 years, exemplifies enhanced compared to the short-lived (Mus musculus), which lives only 2-3 years. cells display superior (NER) activity, as measured by more efficient removal of UV-induced DNA lesions, contributing to their exceptional and cancer resistance. Additionally, (HR), a key pathway for repairing double-strand breaks, is more efficient in due to evolutionary adaptations in proteins like SIRT6, which enhance repair fidelity. These mechanisms, along with adaptations in cGAS that potentiate and suppress , collectively reduce mutation rates and maintain genomic stability over decades. Recent comparative genomics studies have identified genetic underpinnings of these variations in exceptionally long-lived mammals. In bowhead whales (Balaena mysticetus), which can live over 200 years, genomic analyses reveal enhanced DNA double-strand break repair capacity and lower mutation rates, attributed to adaptations in repair pathways including potential gene expansions that bolster fidelity. Similarly, a 2024 analysis of bat genomes uncovered convergent signals in DNA repair genes, such as duplications in longevity-associated loci, in long-lived species like Myotis myotis (maximum lifespan ~37 years) compared to short-lived bats, linking these genomic features to extended lifespans. Overall, such higher repair capacities in long-lived mammals delay the progressive accumulation of irreparable DNA damage, providing a mechanistic explanation for species-specific longevity differences.

DNA Damage in Centenarians and Long-Lived Individuals

Studies on s and supercentenarians reveal significantly lower levels of DNA damage compared to average elderly individuals, supporting the role of resilient maintenance in exceptional . In lymphocytes and fibroblasts from centenarians, DNA damage markers such as γ-H2AX foci are barely detectable under basal conditions, contrasting with higher accumulation in cells from younger or typically aged adults. assays further confirm minimal DNA strand breaks in centenarian dermal fibroblasts, even after exposure to genotoxic stress like gamma , indicating efficient repair processes. These findings from cohorts highlight a between reduced DNA damage and extended healthspan, where centenarians maintain genomic akin to much younger individuals. Specific evidence points to enhanced (BER) pathways, including higher activity of enzymes like OGG1 in leukocytes from long-lived individuals, which effectively mitigates oxidative lesions such as 8-oxo-dG. Italian studies on cohorts demonstrate reduced 8-oxo-dG levels in peripheral blood mononuclear cells, associating this with superior defenses and lower accumulation over decades. Recent 2025 analyses of supercentenarians reinforce this, showing improved capacity and diminished chronic DNA damage signatures compared to non-centenarians, potentially decoupling extreme age from frailty. Notably, γ-H2AX foci are reduced by approximately 50% in cells carrying rare longevity-associated variants, underscoring targeted repair enhancements. Mechanisms underlying this resilience involve a combination of inherited genetic variants and lifestyle factors that bolster and protection. For instance, variants in genes like SIRT6, enriched in populations, enhance double-strand break repair and reduce basal DNA damage in multiple cell types. In , hematopoietic stem cells from long-lived individuals exhibit evident protection against DNA damage accumulation, preserving regenerative capacity through upregulated repair pathways and lower exposure. Lifestyle elements, such as Mediterranean diets in s, further amplify these genetic advantages by minimizing oxidative insults, contributing to overall genome integrity.

Menopause and Reproductive Aging

The DNA damage theory of aging posits that accumulated genomic insults, particularly in non-renewing oocytes, contribute significantly to reproductive senescence and menopause. Oocytes, arrested in prophase I of meiosis from fetal development, are highly susceptible to oxidative stress from reactive oxygen species (ROS), which induce DNA double-strand breaks (DSBs) and mitochondrial DNA (mtDNA) mutations over decades. These damages trigger apoptotic pathways, leading to follicular atresia and progressive depletion of the ovarian reserve. In parallel, telomere shortening in granulosa cells, driven by chronic oxidative damage and limited telomerase activity, impairs follicular support and accelerates oocyte loss. Evidence from human studies reveals elevated DNA damage markers, such as γ-H2AX foci indicating unrepaired DSBs, in perimenopausal ovaries compared to younger tissues, correlating with diminished follicle viability. Mouse models with defects in pathways, including BRCA1-mediated and polymerase β base-excision repair, exhibit accelerated oocyte atresia and premature ovarian failure, mimicking human onset. These models demonstrate that unrepaired DSBs accumulate in primordial follicles, hastening reserve exhaustion independent of chronological age. In humans, mtDNA mutations in granulosa cells and s rise with , correlating with decline observable by the early 40s, as mutated mitochondria impair energy production essential for oocyte maturation. A 2024 study further links gene variants to altered hormonal profiles during perimenopause, where unrepaired damage in ovarian cells disrupts synthesis and accelerates the transition to . Collectively, these processes drive the exhaustion of the finite , rendering DNA damage a key mechanistic driver of reproductive aging.

Atherosclerosis

In atherosclerosis, oxidized low-density lipoprotein (oxLDL) plays a central role in initiating endothelial damage by binding to the lectin-like oxidized (LOX-1) on endothelial cells, which triggers a surge in (ROS) production. This directly induces DNA strand breaks and base modifications, such as 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dG), compromising endothelial integrity and barrier function. The resulting DNA damage activates inflammatory pathways, including signaling, which upregulates adhesion molecules like and , promoting recruitment and infiltration into the subendothelial space. This inflammatory cascade further drives formation as recruited monocytes differentiate into macrophages that avidly uptake oxLDL via scavenger receptors, leading to lipid-laden cells that accumulate and contribute to early plaque development. Histological and biochemical analyses of human atherosclerotic lesions consistently reveal elevated 8-oxo-dG levels, particularly in plaque macrophages and vascular cells, indicating heightened oxidative DNA damage compared to healthy arterial . Deficiencies in mechanisms, such as (NER), amplify this damage and accelerate plaque progression; for example, NER-defective Ercc1 mutant mice display increased vascular cell , endothelial dysfunction, and heightened vascular stiffness as early as 8-16 weeks of age, predisposing to atherogenic changes. These findings underscore how impaired NER fails to excise bulky oxidative adducts from oxLDL-induced ROS, thereby sustaining genomic instability and promoting lesion instability. Telomere dysfunction in vascular smooth muscle cells (VSMCs) further links DNA damage to atherosclerotic calcification. Progressive telomere shortening in VSMCs elicits a DNA damage response, triggering replicative senescence and the senescence-associated secretory phenotype (SASP), which fosters a pro-calcific microenvironment through upregulation of osteogenic factors like BMP-2 and Runx2. This senescent shift promotes VSMC transdifferentiation into osteoblast-like cells, enhancing matrix mineralization and plaque calcification, as evidenced in ApoE−/− mouse models where telomere-targeted interventions reduce VSMC-derived calcified lesions.

Neurodegenerative Diseases

The DNA damage theory of aging posits that accumulated genomic instability contributes to neurodegenerative diseases, where elevated levels of DNA double-strand breaks (DSBs) and oxidative lesions are observed in patient brains. Recent 2025 reviews highlight increased DNA damage foci, such as γ-H2AX and 53BP1 markers, in s from individuals with (AD), (PD), and (HD), linking these hotspots to trinucleotide repeat expansions and impaired repair pathways. This accumulation exacerbates neuronal vulnerability, as unrepaired lesions trigger chronic and , hallmarks of these proteinopathies. In AD, amyloid-beta (Aβ) plaques exacerbate DSBs by inducing , which impairs (NHEJ) and repair mechanisms. Early intracellular Aβ accumulation precedes plaque formation and correlates with incipient DSBs in hippocampal neurons, leading to genomic instability. Tau hyperphosphorylation and tangle formation arise from aberrant DNA damage signaling, where persistent DSBs activate kinases like , promoting neurofibrillary pathology. In , mitochondrial DNA (mtDNA) damage from (ROS) drives loss, with defective (BER) failing to address oxidative lesions. α-Synuclein aggregates further block BER by sequestering repair proteins like OGG1, amplifying mtDNA mutations and spreading pathology to neighboring neurons via damaged mtDNA transfer. This creates a vicious cycle of ROS production and genomic stress in the . HD exemplifies direct causation, where unrepaired DNA lesions at CAG repeat regions cause somatic expansions, worsening polyglutamine toxicity in striatal neurons. Mutant huntingtin disrupts ATM-dependent oxidative repair by altering chromatin scaffolding, leading to persistent DSBs and repeat instability. Therapeutic targeting of ATM kinase shows promise; inhibition reduces genotoxic stress and neuroinflammation in PD models, while modulating ATM enhances repair in HD, potentially slowing progression across these diseases.

DNA Damage and the Epigenetic Clock

DNA damage contributes to the progression of epigenetic aging by altering patterns at specific cytosine-phosphate-guanine (CpG) sites that form the basis of Horvath's , a multi-tissue predictor developed from 353 CpG sites whose levels correlate strongly with chronological across various human tissues and cell types. This clock estimates biological by quantifying drifts—systematic gains or losses in over time—that deviate from expected patterns, and accumulating evidence indicates that DNA lesions, such as single-strand breaks or oxidative modifications, disrupt these patterns, causing the clock to "tick" faster and reflect accelerated biological aging. The underlying mechanism involves DNA damage triggering localized epigenetic reprogramming, where repair processes or damage-induced signaling pathways lead to aberrant methylation at clock-relevant CpG sites, often through the recruitment of methyltransferases or demethylases to damaged regions. For instance, oxidative DNA damage, a common form of genotoxic stress, preferentially affects CpG sites within the Horvath clock that are enriched in regulatory elements sensitive to reactive oxygen species, resulting in methylation drifts that mimic age-related changes and advance the epigenetic age estimate. Studies have identified subsets of these 353 CpG sites as particularly vulnerable to oxidative hits, with methylation alterations correlating directly with damage load and contributing to the clock's sensitivity as a biomarker of cumulative genomic insult. Empirical evidence supports this link through observations that genotoxin exposure accelerates the . For example, chronic exposure to like lead and , known genotoxins that induce DNA strand breaks and oxidative lesions, is associated with increased epigenetic age acceleration in cohorts, independent of chronological age. Similarly, in animal models with deficiencies, such as those mimicking disorders, epigenetic aging proceeds at a faster rate compared to wild-type controls, with elevated Δage (the difference between epigenetic and chronological age) reflecting heightened accumulation of unrepaired damage. Recent investigations, including 2024 analyses of environmental exposures, have further correlated markers of DNA strand breaks—such as γ-H2AX foci—with positive Δage values, underscoring the clock's responsiveness to genotoxic burdens. Beyond direct causation, the epigenetic clock integrates these damage signals as a for lifelong DNA insult accumulation, offering superior predictive power for outcomes. Meta-analyses demonstrate that deviations in clock-derived epigenetic forecast all-cause mortality with 8-15% increased risk per 5-year acceleration, outperforming chronological alone even after adjusting for traditional risk factors. This positions the clock not merely as a chronological tracker but as a functional readout of DNA damage's role in driving systemic aging processes.

Epigenetic Alterations from DNA Damage

DNA damage accumulates over time and induces specific epigenetic modifications that contribute to the aging process by altering structure and patterns. These changes include the erosion of repressive marks and shifts in , which can lead to genomic instability and loss of cellular identity. Unlike irreversible genetic mutations, many of these epigenetic alterations are potentially reversible, offering insights into aging reversal strategies. One prominent epigenetic alteration is the loss of , particularly through the erosion of lysine 9 trimethylation (), a key repressive mark that maintains genomic stability. DNA double-strand breaks trigger the relocalization of chromatin-modifying proteins, such as Suv39h1, away from regions to repair sites, resulting in progressive depletion and destabilization. This erosion promotes genomic instability by allowing inappropriate gene activation and transposon mobilization, which are hallmarks observed in aging cells and premature aging syndromes. Studies in mammalian models have shown that targeted restoration of can mitigate these age-related epigenetic drifts and delay aging phenotypes. DNA damage also drives global DNA hypomethylation, partly through intermediates in (BER) pathways, where oxidized bases like are processed, leading to transient demethylation events that accumulate with age. At damage sites, ATP-dependent remodelers facilitate eviction to allow repair machinery access, temporarily disrupting local epigenetic landscapes and contributing to broader hypomethylation patterns. This histone eviction and subsequent redeposition can perpetuate altered methylation states, exacerbating age-associated transcriptional noise and loss of epigenetic fidelity. In aging tissues, these changes correlate with increased accessibility at repetitive elements, further linking DNA damage to epigenetic dysregulation. The information theory of aging posits that DNA damage primarily erases youthful epigenetic information rather than causing permanent mutations, rendering these alterations reversible in principle. Proposed by Sinclair and colleagues, this framework emphasizes that epigenetic noise from damage disrupts the cell's ability to maintain its identity, but unlike sequence changes, the loss of marks like H3K9me3 or DNA methylation can be rewritten to restore function. Experimental induction of DNA breaks in mice accelerated epigenetic aging, while repair or reprogramming reversed these effects, supporting the theory's core tenet that epigenetic erasure is a causal and malleable driver of aging. Recent epigenetic rejuvenation studies in 2025 have demonstrated that transient expression of OSKM (Oct4, , , ) factors can reverse DNA damage-induced epigenetic alterations by restoring marks and patterns. In aged human fibroblasts and models, OSKM-mediated partial repaired H3K9me3 erosion at fragile sites and reduced global hypomethylation, alleviating genomic instability without full . These findings highlight OSKM's role in reactivating epigenetic maintenance pathways, such as those involving SIRT1 and DNMTs, to counteract damage accumulation and extend cellular lifespan. Advances in chemical reprogramming using small molecules have emerged in as a non-genetic approach to reset damage-induced epigenetic marks, targeting pathways like enzymes for demethylation control and HDAC inhibitors for mark recovery. Cocktails of compounds, such as those modulating Yamanaka factor mimics, have been shown to rejuvenate aged cells by restoring and alleviating BER-related hypomethylation, improving integrity . In models of accelerated aging, these small-molecule interventions have shown potential to reverse epigenetic signatures of damage.

References

  1. [1]
    A review and appraisal of the DNA damage theory of ageing
    The goal of this review is to provide an overview and analysis of the evidence suggesting DNA damage and its imperfect repair play a major role in human ageing.
  2. [2]
    DNA damage—how and why we age? - eLife
    Jan 29, 2021 · There is now sufficient and diverse evidence to support a cogent argument that DNA damage plays a causal role in aging.
  3. [3]
    DNA repair as a shared hallmark in cancer and ageing - FEBS Press
    Jul 14, 2022 · In this review, we revisit the processes involved in DNA damage repair and how these can contribute to diseases, including ageing and cancer ...Dna Repair As A Shared... · 2 Chromatin, Dna Damage, And... · 3 Dna Repair Pathways<|control11|><|separator|>
  4. [4]
    A review and appraisal of the DNA damage theory of ageing - PubMed
    May 10, 2011 · The data suggest a major role of DNA damage in the modulation of longevity, possibly through effects on cell dysfunction and loss.
  5. [5]
    DNA Damage as the Primary Cause of Aging
    Also reviewed is evidence that species longevity is directly related to tissue DNA-repair capacity and that aging may be accelerated by treatment with DNA- ...
  6. [6]
    Aging and DNA damage in humans: a meta-analysis study
    In conclusion, independent of the perspective theory of aging, our meta-analysis results show an age-related increase in DNA damage in humans. Furthermore, ...
  7. [7]
    The role of DNA lesions in the processes leading to aging in mice
    The role of DNA lesions in the processes leading to aging in mice. Symp Soc Exp Biol. 1967:21:29-50. Author. P Alexander. PMID: 4860956.Missing: cancer | Show results with:cancer
  8. [8]
    Correlation between deoxyribonucleic acid excision-repair and life ...
    Correlation between deoxyribonucleic acid excision-repair and life-span in a number of mammalian species ... Proc Natl Acad Sci U S A. 1974 Jun;71(6):2169-73. doi ...Missing: damage | Show results with:damage
  9. [9]
    Longevity-dependent organ-specific accumulation of DNA damage ...
    R W Hart. PMID: 6492898; DOI: 10.1016/0047-6374(84)90049-6. Abstract. To measure directly the accumulation of DNA damage with age, and to understand better the ...
  10. [10]
  11. [11]
    DNA Repair, Genome Stability, and Aging - ScienceDirect
    Here, we review evidence linking aging to nuclear DNA lesions: DNA damage accumulates with age, and DNA repair defects can cause phenotypes resembling premature ...Dna Damage, Reactive Oxygen... · Dna Repair And Aging · The Atm-P53 Axis: A Link...Missing: rodents | Show results with:rodents
  12. [12]
  13. [13]
    DNA damage repair: historical perspectives, mechanistic pathways ...
    Jul 9, 2021 · Therefore, in this review, we focus on the association between DDR and cancer. Due to the characteristic genomic instability of cancer cells, ...
  14. [14]
    Short-term calorie restriction enhances DNA repair by non ... - Nature
    Aug 14, 2020 · Several studies reported that CR prevents age-related decline in base excision and nucleotide excision repair pathways that repair ROS-induced ...
  15. [15]
    DNA damage response signaling pathways and targets for ... - Nature
    May 1, 2020 · Both proteins, one named mediator of MDC1 and another known as 53BP1, are also associated with the signaling of DNA DSBs. MDC1 is a major ...
  16. [16]
    New syndrome reconciles theories of ageing - Nature
    Mutations in components of the nucleotide excision repair (NER) pathway can cause either premature ageing or cancer susceptibility. ... xeroderma pigmentosum, a ...
  17. [17]
    Impaired DNA double-strand break repair contributes to the age ...
    Jul 8, 2016 · Failing to repair DNA double-strand breaks by either nonhomologous end joining (NHEJ) or homologous recombination (HR) poses a threat to genome integrity.
  18. [18]
    Somatic mutations in single human cardiomyocytes reveal age ...
    Aug 11, 2022 · The accumulation of somatic DNA mutations has been demonstrated to be a hallmark of aging in many human cell types, including postmitotic ...Missing: Hart | Show results with:Hart
  19. [19]
    FOXO3 signalling links ATM to the p53 apoptotic pathway ... - Nature
    Aug 14, 2012 · The protein ataxia-telangiectasia mutated (ATM) is activated by DNA double-strand breaks and regulates apoptosis via p53. Here we show that ...
  20. [20]
    Contributions of replicative and translesion DNA polymerases to ...
    May 4, 2023 · UV-induced lesions cause mutations when the NER pathway fails to correct the lesions, allowing them to persist into DNA replication2 where ...
  21. [21]
    DNA damage responses in ageing - PMC - PubMed Central - NIH
    Nov 20, 2019 · Various progeroid syndromes have been linked to genome instability, indicating an important causal role of DNA damage accumulation in the ageing process.
  22. [22]
    Does oxidative damage to DNA increase with age? - PNAS
    The levels of 8-oxo-2-deoxyguanosine (oxo8dG) in DNA isolated from tissues of rodents (male F344 rats, male B6D2F1 mice, male C57BL/6 mice, and female ...
  23. [23]
    Premature aging and metabolic diseases: the impact of telomere ...
    Moreover, significant telomere attrition results in the activation of DNA damage response and apoptotic signaling pathways, resulting in cell cycle arrest and ...
  24. [24]
    Mitochondrial DNA Oxidative Damage and Repair in Aging and ...
    A considerable amount of evidence points to oxidative damage to mitochondrial DNA (mtDNA) as a determinant event that occurs during aging.
  25. [25]
    Mitochondria in oxidative stress, inflammation and aging - Nature
    Jun 11, 2025 · It results in oxidative stress due to excessive reactive oxygen species (ROS) generation, which contributes to cell damage and death.
  26. [26]
    Prolonged persistence of mutagenic DNA lesions in somatic cells
    Jan 15, 2025 · This provides in vivo evidence for biochemical studies of translesion synthesis showing that a single lesion can result in indels, SNVs or ...
  27. [27]
    DNA damage, chromatin, and transcription: the trinity of aging - PMC
    DNA damage leads to local and global changes in gene expression and alterations in chromatin structure, aging-related transcriptional changes affect expression ...Missing: impacts | Show results with:impacts
  28. [28]
    Epigenetics, DNA damage, and aging - PMC - PubMed Central - NIH
    Aug 15, 2022 · At the histone level, histone PTMs that regulate chromatin compaction, gene expression, and the ability to repair DNA damage change with aging. ...
  29. [29]
    Senescence: A DNA damage response and its role in aging and ...
    Senescent cells exhibit permanent cell cycle arrest regulated by p16INK4A and p53-p21-Rb. Elevated expression of p53 upregulates the expression of p21, which ...Abstract · Introduction · DDR activation · Senescence in...
  30. [30]
    Hallmarks and mechanisms of cellular senescence in aging ... - Nature
    Aug 4, 2025 · Aging-induced DNA damage response activates p53, which in turn transcriptionally regulates key genes involved in cellular senescence [27]. One ...
  31. [31]
    Epigenetics, DNA damage, and aging - JCI
    Aug 15, 2022 · CpGs found within transposons tend to lose methylation with organism age, in contrast to promoter CpGs that gain methylation with age, resulting ...
  32. [32]
    Article Oxidative Damage Targets Complexes Containing DNA ...
    Nov 15, 2011 · We demonstrate that oxidative damage induces the formation of a large silencing complex(es) containing DNA methyltransferases and constituents of the polycomb ...
  33. [33]
    DNA damage-induced transcription stress triggers the genome-wide ...
    Jun 24, 2022 · Such Pol II obstructions by TBLs reduce or alter the pool of newly synthesized RNA molecules, causing severe cellular dysfunction. If not ...
  34. [34]
    Genome-wide RNA polymerase stalling shapes the transcriptome ...
    Jan 19, 2023 · Thus, accumulation of stochastic endogenous DNA damage during aging deteriorates basal transcription, which establishes the age-related ...
  35. [35]
    Fencing the genome: ubiquitin signaling restricts heterochromatin ...
    Aug 21, 2025 · The work uncovers a cell cycle-regulated chromatin-associated ubiquitin ligase circuit that maintains epigenetic homeostasis and reveals a novel ...
  36. [36]
    H1N76/77 deamidation facilitates chromatin remodelling and ... - NIH
    Aug 15, 2025 · Chromatin relaxation is a permissiven progress for DNA repair through enabling repair factors to access the damaged DNA.
  37. [37]
    The aging brain: Accumulation of DNA damage or neuron loss?
    The high oxygen consumption rate of postmitotic neurons leads to a progressive accumulation of oxidized DNA during decades of brain aging, which is especially ...
  38. [38]
    Mechanisms of DNA damage‐mediated neurotoxicity in ...
    May 2, 2022 · Increased oxidative DNA damage is observed in brain tissue from aged individuals (Mecocci et al, 1993) and patients with AD (Nunomura et al, ...
  39. [39]
    Role of Oxidative Damage in Alzheimer's Disease and ... - MDPI
    The increased amount of 8-OHdG in mtDNA and nuclear DNA is demonstrated in the aging human brain and in AD [50,51,64]. Hirai et al. have demonstrated that ...
  40. [40]
    Oxidative DNA damage in mild cognitive impairment and late-stage ...
    Increasing evidence supports a role for oxidative DNA damage in aging and several neurodegenerative diseases including Alzheimer's disease (AD).
  41. [41]
    DNA Strand Breaks, Neurodegeneration and Aging in the Brain - PMC
    Defective responses to DNA single- or double-strand breaks can result in neurological disease, underscoring the critical importance of DNA repair for neural ...Missing: renewal | Show results with:renewal
  42. [42]
    Unraveling the Mysteries of DNA Damage in the Brain - Berkeley Lab
    Jan 13, 2025 · Understanding how neurons repair genomic errors sheds light on aging and neurodegenerative disease progression.
  43. [43]
    Aging neurons prioritize essential genes when repairing DNA
    Jun 24, 2021 · Research has shown that a decline in DNA maintenance in neurons is linked to a decrease in their function, which may contribute to age-related ...
  44. [44]
    The Complex Mechanisms by Which Neurons Die Following DNA ...
    This review will discuss our current knowledge of the mechanisms of cell death induced by DNA damage in neurons and their potential importance in ...
  45. [45]
    The role of mitochondrial DNA mutations in aging and sarcopenia
    Aging is associated with a progressive loss of skeletal muscle mass and strength and the mechanisms mediating these effects likely involve mitochondrial DNA ( ...
  46. [46]
    Mitochondrial DNA Mutations Induce Mitochondrial Dysfunction ...
    Aging results in a progressive loss of skeletal muscle, a condition known as sarcopenia. Mitochondrial DNA (mtDNA) mutations accumulate with aging in skeletal ...
  47. [47]
    Mitochondrial ROS-Modulated mtDNA: A Potential Target for ...
    Mitochondrial DNA (mtDNA) damage is associated with the development of cardiovascular diseases. Cardiac aging plays a central role in cardiovascular diseases.Missing: sarcopenia | Show results with:sarcopenia
  48. [48]
    Skeletal muscle aging: influence of oxidative stress and physical ...
    Mar 21, 2017 · Extensive damage to mitochondrial DNA and impaired DNA repair mechanisms in skeletal muscle have been observed with advancing age [29, 78, 79].
  49. [49]
    Age-dependent increases in oxidative damage to DNA, lipids, and ...
    We examined three well-established markers of oxidative damage to DNA, lipids and protein in muscle biopsies obtained from 66 patients aged 25 to 93 years.
  50. [50]
    Decline in skeletal muscle mitochondrial function with aging in ... - NIH
    The current study demonstrates that older people have significantly higher oxidative damage to DNA and that mtDNA abundance decreases with age. This decreased ...
  51. [51]
    Telomeres Shorten in Response to Oxidative Stress in Mouse ... - NIH
    In both strains, oxidative stress resulted in significant telomere shortening in isolated skeletal muscle fibers, likely by different mechanisms.Missing: myofibers weakness
  52. [52]
    Genomic stability and telomere regulation in skeletal muscle tissue
    Studies showed telomere shortening in skeletal muscle in healthy elderly individuals when compared with young controls [69], as well as in experimental models ...Missing: weakness | Show results with:weakness
  53. [53]
    Decline of DNA damage response along with myogenic differentiation
    Nov 22, 2023 · We have studied the DNA repair capacity of muscle fiber nuclei and compared it with the one measured in proliferative myoblasts here.
  54. [54]
    Article DNA-PK Promotes the Mitochondrial, Metabolic, and Physical ...
    May 2, 2017 · Here we report that aging increases DNA breaks and activates DNA-dependent protein kinase (DNA-PK) in skeletal muscle, which suppresses ...Missing: post- | Show results with:post-
  55. [55]
    Mitochondrial pathways in sarcopenia of aging and disuse muscle ...
    In this review, we summarize the current knowledge on the mechanisms whereby mitochondrial dysfunction intervenes in the pathogenesis of sarcopenia and disuse ...
  56. [56]
    Inactivity and Skeletal Muscle Metabolism: A Vicious Cycle in Old Age
    Ager-related loss of muscle mass is linked to the reprogramming of skeletal muscle metabolism [39]. Ranging from DNA methylation/demethylation to histone ...
  57. [57]
    Role of Oxidative Stress in Liver Disorders - MDPI
    When free radical species elude cellular antioxidant defense mechanisms, they react with macromolecules generating toxic adducts that lead to the alteration of ...
  58. [58]
    Mitochondrial alterations in fatty liver diseases - Journal of Hepatology
    Oct 7, 2022 · Due to their role in fatty acid oxidation, lipogenesis and gluconeogenesis, mitochondria are involved in the pathogenesis of non-alcoholic fatty ...
  59. [59]
    Age-related Base Excision Repair Activity in Mouse Brain and Liver ...
    Most abasic sites are next processed by AP endonuclease (Ape), which nicks the phosphodiester backbone 5′ of the abasic site (25,26). A DNA polymerase fills in ...
  60. [60]
    Contributions of DNA interstrand cross-links to aging of cells and ...
    Although rare, DNA interstrand cross-links (ICLs) are among the most deadly types of damage. The cross-linking of the two complementary DNA strands prevents ...Psoralen/uva-Induced Icls... · Ercc1/xpf · Fa Pathway
  61. [61]
    Aging enhances liver fibrotic response in mice through hampering ...
    Clinical data identify age as a factor for severe liver fibrosis. We evaluate whether and how aging modulates the fibrotic response in a mouse model.
  62. [62]
    Liver regeneration in aged mice: new insights
    From these studies it emerges that the age-dependent increase of oxidative stress and inflammatory molecules plays a crucial role in liver regeneration.Missing: abasic crosslinks
  63. [63]
    Impaired base excision repair is related to the pathogenesis of non ...
    Dec 30, 2020 · The activity of BER is decreased in patients with NAFLD and in animal models of this disease. In order to better understand the underlying basis ...
  64. [64]
    The role of DNA repair deficiency in lipid accumulation
    BER-deficient cells are more prone to increased intracellular lipid accumulation after fatty acid exposure. •. NTHL1-KD cells show more pronounced alterations ...Missing: exhaustion | Show results with:exhaustion
  65. [65]
    Hepatocyte Polyploidy: Driver or Gatekeeper of Chronic Liver ...
    Oct 14, 2021 · This work strongly suggested that the DNA damage induced by oxidative stress is the main driving factor underlying polyploidization.
  66. [66]
    Hepatocyte polyploidization and its association with ... - Nature
    May 18, 2017 · Hepatocyte polyploidization is generally considered an indicator of terminal differentiation and cellular senescence, and related to the dysfunction of insulin ...
  67. [67]
    Oxidative DNA Damage in Kidneys and Heart of Hypertensive Mice ...
    AngII-treatment caused DNA double strand breaks in kidney and heart. This marker of DNA damage was more clearly increased than the oxidative stress, which ...
  68. [68]
    Association of glomerular DNA damage and DNA methylation with ...
    Jan 14, 2020 · Accumulation of DNA double-strand breaks (DSBs) is linked to aging and age ... accumulation of DNA damage, represented by increased γH2AX levels.
  69. [69]
    Persistent DNA damage underlies tubular cell polyploidization ... - NIH
    Nov 1, 2023 · Our data delineate a mechanistic pathway by which persistent DNA damage in the kidney tubular cells leads to kidney injury and development of CKD.
  70. [70]
    DNA damage in proximal tubules triggers systemic metabolic ...
    Apr 28, 2025 · The accumulation of DNA damage in renal proximal tubular epithelial cells (PTEC) is related to a decline in kidney function. Here, we report ...
  71. [71]
    Stressors damage kidneys by mutating mitochondrial DNA
    Oct 14, 2025 · Kidney damage that seemingly heals appears to mutate the DNA in the mitochondria of kidney cells, making the organ less resilient to future ...
  72. [72]
    Kidney-Specific Landscape of Aging Mitochondrial DNA Mutations ...
    Mutation spectrum analysis showed that mtDNA point mutations in the kidney increased significantly with age and were primarily G>A/C>T, indicative of polymerase ...Missing: chronic | Show results with:chronic
  73. [73]
    DNA-damaged podocyte-CD8 T cell crosstalk exacerbates kidney ...
    Apr 25, 2023 · Podocyte DNA damage alters the blood methylome, leading to changes in CD8 + T cells, which contribute to sustained renal injury in chronic kidney disease.
  74. [74]
    Podocyte senescence: from molecular mechanisms to therapeutics
    Loss of HDAC1/2 in podocytes results in DNA damage and senescence. Deletion of HDAC1/2 in podocytes results in sustained DNA double-strand breaks (DSBs) ...
  75. [75]
    DNA damage and repair in patients with early chronic kidney ...
    Sep 16, 2025 · A correlation analysis was performed to identify associations between levels of oxidative DNA damage, the repair enzyme, and CKD progression. An ...
  76. [76]
    DNA damage and repair in the hematopoietic system - PubMed
    Jan 25, 2022 · Generally, DNA damage in HSCs is endogenous, which is typically induced by reactive oxygen species (ROS), aldehydes, and replication stress.
  77. [77]
  78. [78]
    DNA damage and mutation. Types of DNA damage - BioDiscovery
    Feb 23, 2014 · ... DNA mutation is that DNA mutations are heritable, while DNA damage may be not. Generally, less than 0.1% (1 out of 1000) of the base ...
  79. [79]
    From DNA damage to mutations: all roads lead to aging - PMC
    DNA damage as a possible direct cause of aging was first postulated by Alexander (Alexander, 1967) and became generally accepted as a bona fide theory of aging ...
  80. [80]
    Somatic mutation rates scale with lifespan across mammals - Nature
    Apr 13, 2022 · If somatic mutations contribute to ageing, theory predicts that somatic mutation rates may inversely correlate with lifespan across species.
  81. [81]
    Endogenous DNA Damage as a Source of Genomic Instability ... - NIH
    Among the estimated 70,000 lesions per day that a normal cell is forced to cope with, each has the potential to be converted into a mutation. The most frequent ...Missing: percentage | Show results with:percentage
  82. [82]
    Somatic mutations in human ageing: New insights from DNA ...
    Recent DNA sequencing studies and inherited mutations do not support the idea that somatic mutations accumulating with age drive ageing phenotypes.Review Article · 2. Somatic Mutations... · 4. Impact Of Age-Related...
  83. [83]
    Interaction between the Cockayne syndrome B and p53 proteins
    The defect in DNA repair without any increase of the mutation load can be explained only by assuming that the lack of CSB forces damaged cells into the ...
  84. [84]
    Clinical implications of the basic defects in Cockayne Syndrome and ...
    The human repair deficient diseases xeroderma pigmentosum (XP) and Cockayne syndrome (CS) have specific phenotypes of cancer and neurodegeneration.
  85. [85]
    Genomic Instability and Epigenetic Changes during Aging - MDPI
    Increasing evidence indicates that age-related alterations in epigenetics are largely influenced by DNA damage. During DNA repair processes, DNA ...
  86. [86]
    The Free Radical Theory of Aging Is Dead. Long Live the Damage ...
    The free radical theory of aging posits that aging is caused by accumulation of damage inflicted by reactive oxygen species (ROS).
  87. [87]
    Updating the Free Radical Theory of Aging - Frontiers
    The free radical theory of aging hypothesizes that oxidative damage to the mtDNA induces random de novo mtDNA mutations which gradually accumulate over time, ...
  88. [88]
    Mitochondrial DNA and aging | Clinical Science - Portland Press
    The mitochondrial theory of aging can be considered as an extension and refinement of the free radical theory. Its major premise is that mtDNA mutations ...Mtdna Damage And The... · Mtdna Repair And Aging · Challenges To The...<|separator|>
  89. [89]
    Telomeres: history, health, and hallmarks of aging - ScienceDirect
    Jan 21, 2021 · This attrition ultimately triggers DNA damage responses such as cell cycle arrest, apoptosis, impaired differentiation, and/or senescence.
  90. [90]
    Telomere, aging and age-related diseases - PubMed
    Thus, the loss of telomeres may lead to DNA damage. The relationship between cellular senescence and telomere shortening is well established. Furthermore, ...
  91. [91]
    Cross-talk of inflammation and cellular senescence - Nature
    Dec 3, 2024 · Cellular aging is characterized by DNA damage, mitochondrial dysfunction, and permanent cell cycle arrest, ultimately leading to SASP. SASP ...
  92. [92]
    BIN1 deficiency leads to DNA damage and neuronal insulin ...
    Aug 21, 2025 · Persistently activated mTORC1 feedback inhibits the insulin signaling; meanwhile, ATM-dependent DNA Damage Response decreases the expression of ...
  93. [93]
    Premature aging in genetic diseases: what conclusions can be ...
    In this review we will focus on primary causes of well-investigated premature aging diseases Hutchinson-Gilford progeria syndrome (HGPS), WS, and Cockayne ...
  94. [94]
    Recent Advances in Understanding Werner Syndrome - PMC - NIH
    Sep 29, 2017 · Patients with WS have defects in DNA repair machinery and show genomic instability. WRN, in association with the telomere-protecting shelterin ...
  95. [95]
    Genomic instability and DNA damage responses in progeria arising ...
    Accumulation of mutant lamin A causes progressive changes in nuclear architecture in Hutchinson-Gilford progeria syndrome. ... DNA double strand breaks DNA repair ...
  96. [96]
    DNA-damage accumulation and replicative arrest in Hutchinson ...
    A common feature of progeria syndromes is a premature aging phenotype and an enhanced accumulation of DNA damage arising from a compromised repair system.
  97. [97]
  98. [98]
    Why Cockayne syndrome patients do not get cancer despite ... - PNAS
    The absence of cancer in CS patients results from the absence of UV-induced mutagenesis rather than from enhanced lethality.
  99. [99]
    Heterogeneous clinical features in Cockayne syndrome patients and ...
    Mar 5, 2022 · Background. Nucleotide Excision Repair (NER) is a complex DNA repair system capable of removing a variety of DNA lesions such as UV-induced ...
  100. [100]
    Restoration of energy homeostasis by SIRT6 extends healthy lifespan
    May 28, 2021 · Here, we show that SIRT6 overexpression leads to a reduction in frailty and lifespan extension in both male and female B6 mice.
  101. [101]
    Prolonged lifespan with enhanced exploratory behavior in mice ...
    May 4, 2013 · Longevity conferred by overexpression of a single nucleotide hydrolase in hMTH1-Tg animals is an example of lifespan extension associated with ...
  102. [102]
    Longevity and resistance to stress correlate with DNA repair ...
    The nematode Caenorhabditis elegans is a good model for studying longevity and DNA repair because of key advances in understanding the genetics of aging in ...
  103. [103]
    Elevated Proteasome Capacity Extends Replicative Lifespan in ...
    Expression of UPS genes in yeast and mammals is regulated ... upregulated DNA repair contributes to lifespan extension in cells with elevated Rpn4 levels.
  104. [104]
    PARP inhibitors elicit a cellular senescence mediated inflammatory ...
    May 2, 2025 · These results suggest that PARP inhibitor-induced DNA damage causes cellular senescence or senescence-like phenotypes, with elevated ...
  105. [105]
    The polymorphism of DNA repair genes XPD, XRCC1, OGG1, and ...
    Aug 23, 2014 · The polymorphism of DNA repair genes XPD, XRCC1, OGG1, and ERCC6, life expectancy, and the inclination to smoke. Human Genetics; Published: 23 ...
  106. [106]
    Caloric restriction decreases mitochondrial free radical generation at ...
    May 9, 2001 · Long-term caloric restriction decreased mitochondrial H2O2 generation by 45% and lowered mtDNA oxidative damage by 30% in rat hearts.
  107. [107]
    Calorie restriction induces mitochondrial biogenesis and ... - PNAS
    Calorie restriction (CR) is the most robust, nongenetic intervention that increases lifespan and reduces the rate of aging in a variety of species. Mechanisms ...
  108. [108]
    Caloric restriction induced epigenetic effects on aging - Frontiers
    Increasing data suggests that dietary changes can alter epigenetic marks associated with aging. Caloric restriction (CR)is considered an intervention to ...
  109. [109]
    Mechanisms and disease implications of sirtuin-mediated ... - Nature
    Sep 6, 2019 · Calorie restriction increases the level of SIRT1, which can deacetylate targets, including the DNA repair factor Ku70 and p53. The deacetylation ...
  110. [110]
    Caloric restriction and genomic stability - PMC - PubMed Central
    It has been suggested that CR extends longevity and reduces age-related pathologies by reducing the levels of DNA damage and mutations that accumulate with age.
  111. [111]
    Practicality of Intermittent Fasting in Humans and its Effect on ... - NIH
    An alternate-day fasting paradigm has been shown to reduce markers of oxidative stress and improve clinical findings.
  112. [112]
    Effect of 6-Month Calorie Restriction on Biomarkers of Longevity ...
    Apr 5, 2006 · DNA damage was significantly reduced from baseline in the calorie restriction, calorie restriction with exercise, and very low-calorie diet ...
  113. [113]
    Calorie restriction reduces biomarkers of cellular senescence in ...
    Nov 14, 2023 · ... DNA methylation data (Waziry et al., 2023). ... DNA methylation measures of biological aging in healthy adults from the CALERIE trial.
  114. [114]
    NAD+ in Aging: Molecular Mechanisms and Translational Implications
    NAD+ is critical for cellular energy and stress responses. Its depletion is linked to aging and may be a therapeutic target.
  115. [115]
    Unraveling the Mysteries of Aging | Harvard Medical School
    Mar 24, 2017 · Additionally, experiments conducted in mice show that treatment with the NAD precursor NMN mitigates age-related DNA damage and wards off DNA ...Missing: BER review
  116. [116]
    Senolytics: Eliminating Senescent Cells and Alleviating ... - Frontiers
    As a type of aging treatment strategy, senolytics can selectively clear senescent cells by inducing apoptosis and reduce the production of SASP, contributing ...
  117. [117]
    Senolytics reduce endothelial cell DNA damage and telomere ...
    May 23, 2023 · Senolytics are drugs that specifically induce cell death in senescent cells. Senolytic drugs have been shown to delay or reduce a multitude of ...
  118. [118]
    The therapeutic perspective of NAD + precursors in age-related ...
    This review sheds light on NAD + therapeutic efficiency in preclinical and clinical studies and the effect of the gut microbiota on NAD + metabolism.Missing: BER | Show results with:BER
  119. [119]
    Targeted partial reprogramming of age-associated cell states ...
    Sep 11, 2024 · Partial reprogramming via OSK expression in aged and stressed cells improved both median (about 40%) and maximal (about 32%) life spans in LAKI ...
  120. [120]
    In Vivo Amelioration of Age-Associated Hallmarks by Partial ...
    Here, we report that cyclic in vivo induction of OSKM in a mouse model of premature aging improves age-associated phenotypes and extends lifespan. In addition, ...
  121. [121]
    Chemically induced reprogramming to reverse cellular aging
    We have previously shown that the ectopic induction of the Yamanaka factors OCT4, SOX2, and KLF4 (OSK) in mammals can restore youthful DNA methylation patterns, ...
  122. [122]
    Chemical reprogramming ameliorates cellular hallmarks of aging ...
    Jun 30, 2025 · This study reveals that partial chemical reprogramming with defined small-molecule cocktails rejuvenates aged human cells and significantly extends lifespan ...
  123. [123]
    Scientists just found a surprising link between gray hair and cancer
    Oct 25, 2025 · Japanese researchers discovered that hair graying and melanoma share a surprising cellular origin. When DNA damage strikes melanocyte stem cells ...Missing: reversal reprogramming
  124. [124]
    Partial cellular reprogramming: A deep dive into an emerging ...
    Partial cellular reprogramming is able to improve or restore multiple age‐related phenotypes including lifespan, healthspan, epigenetic age, aging hallmarks, ...
  125. [125]
    DNA repair in species with extreme lifespan differences - PMC - NIH
    It is now well documented that even among phylogenetically related rodents, extreme differences in lifespan are common. Most notably, the naked mole rat (NMR), ...
  126. [126]
    DNA repair in species with extreme lifespan differences - Aging-US
    The accumulation of DNA damage and mutations in multicellular organisms increases the risk of cancer and is linked to aging [1–3]. Essential genome maintenance ...
  127. [127]
    Naked mole rat cells display more efficient excision repair than ...
    Our results suggest that the NMR has more efficient excision repair systems than the mouse, which may contribute to longevity and cancer resistance of this ...
  128. [128]
    SIRT6 Is Responsible for More Efficient DNA Double-Strand Break ...
    Apr 18, 2019 · Here we report that DNA DSB repair coevolves with longevity. Remarkably, both pathways of DSB repair, HR and NHEJ, are more efficient in long-lived species.
  129. [129]
    Evidence for improved DNA repair in long-lived bowhead whale
    Oct 29, 2025 · At more than 200 years, the maximum lifespan of the bowhead whale exceeds that of all other mammals ... efficiency of DNA repair pathways.
  130. [130]
    Comparative genomics reveals convergent signals associated with ...
    Aug 28, 2024 · Comparative genomics reveals convergent signals associated with the high metabolism and longevity in birds and bats.Missing: duplications | Show results with:duplications
  131. [131]
    Genomic stability, anti-inflammatory phenotype, and up-regulation of ...
    Jan 8, 2019 · Here we show a remarkable low level of DNA damage in cells from centenarians, i.e., almost absent comet tails and barely detectable levels of γ- ...
  132. [132]
    Super DNAging—New Insights Into DNA Integrity, Genome Stability ...
    Notably, cells of the oldest humans (centenarians) showed improved antioxidant defense and DNA repair activity compared to younger elderly and were even similar ...
  133. [133]
    Genetic signatures of exceptional longevity: a comprehensive ...
    Oct 7, 2025 · This research investigates common genetic variations (SNPs) shared among 3 centenarians and 18 supercentenarians, individuals aged 110 years or ...<|control11|><|separator|>
  134. [134]
    A rare human centenarian variant of SIRT6 enhances genome ...
    Basal γH2AX foci in cumate‐inducible SIRT6 fibroblasts. Foci were scored in at least 80 cells per condition. Representative images of the foci are shown in ...
  135. [135]
    Impairment of BRCA1-related DNA Double Strand Break Repair ...
    Increased DSBs in aging mouse and human oocytes. If DNA DSB repair efficiency decreases with age then DNA DSBs should accumulate in oocytes over time. As a ...
  136. [136]
    importance of DNA repair for maintaining oocyte quality in response ...
    Recent studies have shown that DNA DSBs can accumulate in oocytes in primordial follicles during reproductive ageing, and are readily induced by exogenous ...Dna Double-Strand Breaks · Dna Dsb Repair In Meiotic... · Dna Dsb Repair In Primordial...
  137. [137]
    The role of oxidative stress in ovarian aging: a review
    Sep 1, 2022 · Excessive levels of ROS not only induce mtDNA mutations to result in inefficient electron transport chain (ETC) expression, but also mediate ...
  138. [138]
    Estrogen deficiency reversibly induces telomere shortening in ...
    Estrogen deficiency or over-activity may cause ovarian tissue aging or tumorigenesis, respectively, through estrogen regulation of telomere remodeling.
  139. [139]
    Molecular Medicine Reports - Spandidos Publications
    Sep 18, 2023 · The progressive shortening of telomeres within granulosa cells (GCs) due to inadequate DNA repair and long-term oxidative stress has been ...
  140. [140]
    Spatiotemporal transcriptomic changes of human ovarian aging and ...
    Apr 9, 2024 · Overall, increased DNA damage and diminished DNA repair may contribute to aging in primordial or primary stage oocytes. Subsequently, ...
  141. [141]
    Increased DNA damage and repair deficiency in granulosa cells are ...
    Increased DSBs and reduced DNA repair in granulosa cells may contribute to ovarian aging. Discovery of therapeutics that targets these pathways might help ...Missing: perimenopausal | Show results with:perimenopausal
  142. [142]
    Oocytes can efficiently repair DNA double-strand breaks to ... - PNAS
    May 7, 2020 · Depletion of these oocytes commonly leads to infertility and early menopause ... DNA double-strand break repair leads to ovarian aging in mice and ...
  143. [143]
    Impairment of Pol β-related DNA base-excision repair leads to ...
    Nov 20, 2020 · Previous studies on germinal vesicle (GV) oocytes have focused on the role of DNA double-strand break (DSB) repair in ovarian aging [5].
  144. [144]
    The Molecular Regulation in the Pathophysiology in Ovarian Aging
    The mutation rate of mtDNA in granule cells (CCs) of oocytes in elderly ... BRCA Mutations, DNA Repair Deficiency, and Ovarian Aging. Biol Reprod, 93 ...
  145. [145]
    Mitochondria: the epigenetic regulators of ovarian aging and longevity
    Oxidative stress, mitochondrial DNA mutation, and impairment of antioxidant enzymes in aging. Exp Biol Med (Maywood N.J.). (2002) 227:671–82. doi: 10.1177 ...
  146. [146]
    Genetic links between ovarian ageing, cancer risk and de ... - Nature
    Sep 11, 2024 · For genes that inhibit DNA double-strand break repair, the hypothesis is that they cause premature depletion of the ovarian reserve owing to a ...
  147. [147]
  148. [148]
    DNA Damage and Repair in Atherosclerosis: Current Insights ... - NIH
    Small chemical alterations of bases, such as oxidation, methylation, deamination and hydroxylation producing single strand breaks (SSB), are targeted by BER.
  149. [149]
    Nucleotide Excision DNA Repair Is Associated With Age-Related ...
    Jun 15, 2012 · One of the DNA repair systems that is very important in this regard is nucleotide excision repair (NER), which removes a wide class of helix- ...
  150. [150]
    Ercc1 DNA repair deficiency results in vascular aging characterized ...
    Mar 7, 2024 · First, decreased efficiency of repair leads to presence of unrepaired DNA damage, which as a consequence upregulates p53 and p73 protein ...<|control11|><|separator|>
  151. [151]
    Atherosclerosis Is a Smooth Muscle Cell–Driven Tumor-Like Disease
    Apr 30, 2024 · Enhanced p53 pathway activity (Figure S1D; Figure 3A) suggests that the DNA damage response is induced in SDCs during atherosclerosis ...
  152. [152]
    Targeting Telomere Shortening in Vascular Aging and Atherosclerosis
    Shortened telomeres initiate DNA damage responses, increase oxidative stress, and activate inflammatory signaling, fostering a pro-inflammatory secretory ...
  153. [153]
    DNA damage and its links to neuronal aging and degeneration
    Jan 8, 2025 · DNA damage is a major risk factor for the decline of neuronal functions with age and in neurodegenerative diseases.Missing: seminal papers<|control11|><|separator|>
  154. [154]
    DNA Double Strand Breaks: A Common Theme in ... - ResearchGate
    Aug 9, 2025 · Accumulation of DNA damage and impairment of DNA repair systems are involved in the pathogenesis of different neurodegenerative diseases.
  155. [155]
    Early oxidative stress and DNA damage in Aβ-burdened ... - Nature
    Jul 14, 2024 · Our findings indicate that early accumulation of iAβ, prior to Aβ plaques, is accompanied by incipient oxidative stress and DSBs.
  156. [156]
    Genomic stress and impaired DNA repair in Alzheimer disease
    We review evidence that Aβ and Tau abnormalities induce excessive genomic stress and impair genome maintenance mechanisms, events that can promote DNA damage- ...Dna Damage And Genomic... · Impaired Genome Maintenance... · Aβ<|separator|>
  157. [157]
    Mitochondrial DNA damage triggers spread of Parkinson's disease ...
    Oct 2, 2023 · Injecting damaged mtDNA into mouse brain induced PDD-like behavioral symptoms, including neuropsychiatric, motor, and cognitive impairments.
  158. [158]
    DNA Damage and Parkinson's Disease - PMC - PubMed Central
    Apr 10, 2024 · One rarely considered hypothesis is that ROS produced by defective mitochondria will lead to the formation of oxidative DNA damage in nuclear DNA.
  159. [159]
    Base excision repair causes age-dependent accumulation of single ...
    Sep 7, 2021 · Here, we show that base excision repair generates genomic stress, promoting age-related neurodegeneration in a Caenorhabditis elegans PD model.
  160. [160]
    The central role of DNA damage and repair in CAG repeat diseases
    Recent human genetic evidence implicates DNA repair pathways, especially mismatch repair, in modifying the onset and progression of CAG repeat diseases.
  161. [161]
    Huntingtin is a scaffolding protein in the ATM oxidative DNA damage ...
    Nov 21, 2016 · We report that endogenous huntingtin protein directly participates in oxidative DNA damage repair. Using novel chromobodies to detect endogenous human ...Missing: unrepaired | Show results with:unrepaired
  162. [162]
    Mystery solved: New study reveals how DNA repair genes play a ...
    Feb 11, 2025 · Recent studies suggest the mutant Huntingtin CAG repeats are unstable in adult brain cells, especially in the vulnerable neurons such as the ...Missing: lesions ATM kinase
  163. [163]
    DNA Damage Response Regulation Alleviates Neuroinflammation ...
    Jun 20, 2025 · ATM inhibition may exert its protective effects in PD by mitigating genotoxic stress possibly arising from oxidative damage and other cellular ...
  164. [164]
    DNA Damage Repair in Huntington's Disease and Other ...
    CAG expansion in the androgen receptor, a transcription factor involved in DNA damage repair signaling, leads to spinal and bulbar muscle atrophy, SBMA, or ...Missing: unrepaired | Show results with:unrepaired
  165. [165]
    DNA methylation age of human tissues and cell types
    Finally, I characterize the 353 CpG sites that together form an aging clock in terms of chromatin states and tissue variance. Conclusions. I propose that DNA ...
  166. [166]
    Epigenetics and Oxidative Stress in Aging - PMC - NIH
    Jul 20, 2017 · We give a comprehensive overview on how the combination of different epigenetic alterations and oxidative stress affects the process of aging.
  167. [167]
    Age-dependent methylation in epigenetic clock CpGs is associated ...
    Sep 29, 2018 · ABSTRACT. Horvath's epigenetic clock consists of 353 CpGs whose methylation levels can accurately predict the age of individuals.
  168. [168]
    Lead and cadmium exposure was associated with faster epigenetic ...
    We found that exposure to lead and cadmium was associated with accelerated epigenetic age. These findings suggest the potential role of lead and cadmium in EAA.Missing: genotoxin | Show results with:genotoxin
  169. [169]
    DNA repair models: accelerated epigenetic age
    Dec 22, 2023 · These findings highlight that mouse models with deficiencies in DNA repair, unlike other premature aging models, display accelerated epigenetic age.
  170. [170]
    Epigenetic aging acceleration among World Trade Center-exposed ...
    Sep 30, 2025 · It's also possible that epigenetic clocks are capturing cumulative lifetime damage to DNA. These theories are not mutually exclusive, and ...
  171. [171]
    The epigenetic clock as a predictor of disease and mortality risk
    Apr 11, 2019 · Meta-analyses indicated that each 5-year increase in DNA methylation age was associated an 8 to 15% increased risk of mortality.
  172. [172]
    Loss of H3K9 trimethylation leads to premature aging - PMC
    Loss of H3K9me3 leads to age-associated epigenetic and transcriptional changes. To further confirm that the loss of H3K9me3 loss results in premature aging ...
  173. [173]
    DNA damage and Repair Modify DNA methylation and Chromatin ...
    Sep 15, 2016 · Our data argue that DNA methylation and chromatin remodelling induced by HR may be a source of permanent variation of gene expression in somatic cells.
  174. [174]
    (PDF) DNA damage and histone variants - ResearchGate
    When the DNA is damaged, repair enzymes need to gain access to the damage, and this requires epigenetic changes to the compact structure. Similar to mutations, ...
  175. [175]
    Unraveling Histone Loss in Aging and Senescence - MDPI
    In this review, we focus on histones, a central component of the epigenome, and consolidate the key findings of histone loss and genome-wide redistribution.Unraveling Histone Loss In... · 3. Histone Loss In Mammalian... · 8. Histone Complementation...
  176. [176]
    The Information Theory of Aging - PubMed - NIH
    Dec 15, 2023 · The Information Theory of Aging (ITOA) states that the aging process is driven by the progressive loss of youthful epigenetic information.
  177. [177]
    (PDF) The Information Theory of Aging - ResearchGate
    Dec 19, 2023 · The Information Theory of Aging (ITOA) states that the aging process is driven by the progressive loss of youthful epigenetic information.
  178. [178]
    Epigenetic Regulation of Aging and its Rejuvenation - An - 2025
    Sep 1, 2025 · This article provides an overview of the epigenetic mechanisms and key targets involved in aging, including DNA methylation, ...
  179. [179]
    Resetting the Biological Clock: OKSM and the Reversal of Aging
    Mar 19, 2025 · OKSM (Oct4, Sox2, Klf4, and c-Myc) transcription factors have demonstrated the ability to reset epigenetic markers, restoring youthful gene expression and ...
  180. [180]
    Exploring the Anti-Aging Potential of OSKM | Stemaid Institute
    Sep 25, 2025 · OSKM has been shown to reduce the number of senescent cells, characterized by the accumulation of DNA damage markers like histone γ-H2AX. By ...<|separator|>
  181. [181]
    Chemical reprogramming ameliorates cellular hallmarks of aging ...
    Here, we demonstrated that chemical-induced partial reprogramming can improve key drivers of aging including genomic instability and epigenetic alterations in ...