Fact-checked by Grok 2 weeks ago

DNA methylation

DNA methylation is an epigenetic modification in which a (CH₃) is covalently attached to the fifth carbon of the base (C5 position) in a DNA molecule, primarily occurring at residues followed by guanine (CpG dinucleotides) in vertebrates. This process does not alter the underlying DNA but influences by generally repressing transcription, either through direct inhibition of to DNA or by recruiting methyl-binding proteins that facilitate compaction and . Catalyzed by a family of enzymes known as DNA methyltransferases (DNMTs), including for maintenance of methylation patterns during and DNMT3A/DNMT3B for establishing new () methylation marks, DNA methylation plays essential roles in embryonic development, , X-chromosome inactivation, and suppression of transposable elements. Aberrant DNA methylation patterns, such as global hypomethylation or hypermethylation of tumor suppressor genes, are hallmarks of various diseases, including cancer, and are influenced by environmental factors like diet and toxins. In mammals, DNA methylation is highly dynamic, with waves of reprogramming occurring in early embryogenesis and cells to reset epigenetic marks, underscoring its importance in and inheritance of epigenetic states across generations.

Fundamentals

Definition and Chemical Basis

DNA methylation is an epigenetic modification involving the covalent addition of a (CH₃) to the 5-position of bases within DNA, primarily occurring at CpG dinucleotides in vertebrates and resulting in the formation of (5mC). This modification alters the physical properties of DNA without changing its nucleotide sequence and is recognized as the most extensively studied epigenetic mark due to its and across cell divisions.00071-3) First identified in 1948 by Rollin Hotchkiss through analysis of calf DNA, where he detected an unusual of modified comprising about 1% of total pyrimidines, DNA methylation has since become central to understanding and cellular identity.77315-0/fulltext) Chemically, the methylation reaction entails the enzymatic transfer of a from the universal donor S-adenosylmethionine () to the carbon-5 position of , yielding 5mC and S-adenosylhomocysteine (SAH) as a byproduct; this process is catalyzed by DNA methyltransferases. In structural terms, consists of a ring with a keto group at position 2, an amino group at position 4, and a hydrogen at position 5; methylation introduces the CH₃ group at this C5 position, which does not disrupt base pairing but influences DNA-protein interactions. A simplified representation of this conversion can be depicted as:
   NH₂          NH₂
    |            |
   C=O    →     C=O
  /   \        /   \
H-C     N     H-C     N
 |     / \    |     / \
 N     C   C   N     C   C
 \    / \ /    \    / \ /  
  C--N   N      C--N   N-CH₃
   |             |
   H             H
This diagram illustrates the ring of (left) transforming to (right) via addition at , maintaining the overall ring structure and hydrogen bonding capability with . In mammalian cells, DNA methylation is highly prevalent, affecting approximately 70-80% of all CpG sites across the , which contributes to the establishment of tissue-specific epigenetic landscapes. While CpG methylation dominates in vertebrates, non-CpG methylation—occurring at CHG, CHH, or /CpT contexts—is more common in , where it plays roles in transposon silencing, and is also observed in mammalian embryonic stem cells at levels up to 25% of total modifications.

Enzymatic Machinery

DNA methylation is primarily catalyzed by a family of enzymes known as DNA methyltransferases (DNMTs), which transfer a from the cofactor to the 5-position of bases in DNA. In mammals, the core DNMTs include , responsible for maintenance methylation, and DNMT3A and DNMT3B, which mediate methylation.81656-6) These enzymes ensure the faithful propagation and establishment of methylation patterns across cell divisions. DNMT1 functions as the maintenance methyltransferase, preferentially targeting hemimethylated DNA that arises during replication to restore symmetric methylation on daughter strands. It recognizes hemimethylated CpG sites through its N-terminal domain, which interacts with replication foci, while the C-terminal catalytic domain flips the target into an pocket where methylation occurs using as the methyl donor. This process is highly processive, allowing DNMT1 to methylate multiple sites on the same DNA molecule without dissociation. DNMT1 activity is allosterically regulated; for instance, unmethylated CpG sites can inhibit its methylation of hemimethylated DNA, enhancing specificity. De novo methylation, which establishes new methylation patterns on previously unmethylated DNA, is carried out by DNMT3A and DNMT3B. These enzymes form heterotetrameric complexes that oligomerize along DNA, enabling cooperative methylation of adjacent CpG sites. DNMT3A and DNMT3B share structural similarities, including a catalytic domain that also flips cytosine for SAM-dependent methylation, but they exhibit tissue-specific expression and target preferences.81656-6) The catalytically inactive DNMT3L serves as a regulatory cofactor, stimulating DNMT3A and DNMT3B activity by stabilizing their complexes and modulating substrate specificity, particularly in germ cells. DNA demethylation counters methylation through both passive and active mechanisms. Passive demethylation occurs during when maintenance methylation by is impaired, leading to dilution of (5mC) over successive cell divisions. Active demethylation is mediated by ten-eleven translocation () enzymes (TET1, TET2, and TET3), which are α-ketoglutarate and Fe²⁺-dependent dioxygenases that iteratively oxidize 5mC to (5hmC), then to 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC). These oxidized derivatives are recognized by thymine DNA glycosylase (TDG), initiating to replace them with unmodified cytosines. The SAM cycle provides the universal methyl donor for DNMT activity, where is synthesized from and ATP, donates its methyl group to become S-adenosylhomocysteine, and is regenerated via remethylation in a folate-dependent pathway. Disruptions in this cycle can alter global methylation levels. Overall, the interplay of these enzymes and cofactors tightly controls DNA .

Methylation Patterns

DNA methylation exhibits distinct genomic distribution patterns, primarily occurring at residues within CpG dinucleotides in s, though non-canonical contexts exist in specific cell types and organisms. These patterns are characterized by sequence-specific preferences and regional enrichments that influence the epigenetic landscape. CpG islands (CGIs) represent a prominent feature of the methylome, defined as GC-rich regions spanning approximately 1-2 kb with a high density of CpG dinucleotides. These islands are typically unmethylated and are associated with about 70% of gene promoters, particularly those of genes. The standard criteria for identifying CGIs include a minimum of 200 base pairs, a greater than 50%, and an observed-to-expected CpG ratio exceeding 0.6. In addition to promoter-associated methylation, gene body methylation—defined as methylation within transcribed exons and introns—shows a positive with transcriptional activity. Highly expressed s often exhibit elevated methylation levels across their gene bodies, which may help maintain efficient elongation by . Conversely, gene body methylation is inversely related to the prevalence of , with lowly methylated gene bodies associated with higher splicing complexity and events. Non-CpG methylation, where cytosine methylation occurs in CHG or CHH contexts (H = A, C, or T), is less common in mammals but prevalent in , where it contributes to transposon silencing and maintenance. In mammalian systems, non-CpG methylation is prominent in embryonic stem cells (ESCs), comprising up to 25% of total cytosine methylation, primarily at CHG sites, and persists in post-mitotic neurons, where CH methylation levels can reach 10-20% of total methylation. These patterns are established by DNMT3A/DNMT3B enzymes in mammals and DRM2 in . Globally, DNA methylation is enriched at repetitive elements, such as transposable elements and , where hypermethylation prevents their mobilization and ensures genomic stability. This hypermethylation affects over 90% of CpG sites in repeats across genomes. Tissue-specific variations further modulate these patterns; for instance, tissues exhibit relative hypomethylation compared to other organs, particularly in neuronal populations, reflecting adaptations for and higher non-CpG content.

Conserved Biological Functions

Gene Expression Regulation

DNA methylation primarily regulates by repressing transcription, serving as a conserved epigenetic mechanism across eukaryotes to maintain cellular identity and developmental programs. In this context, methylation at cytosine residues within CpG dinucleotides recruits methyl-binding domain (MBD) proteins, such as MeCP2, which interpret the methylation signal and initiate downstream repressive cascades. These proteins bridge DNA methylation to modifications, ensuring stable without altering the underlying DNA sequence. The core mechanism of repression involves MeCP2 binding to methylated DNA, which recruits histone deacetylases (HDACs) to remove acetyl groups from s, leading to condensation and reduced accessibility for transcriptional machinery. Additionally, MeCP2 associates with histone methyltransferases that deposit repressive marks, such as trimethylation of at lysine 9 (H3K9me), promoting formation and blocking the binding of activator transcription factors. This dual action—deacetylation via HDACs and methylation via H3K9me—compacts structure, effectively silencing target genes and preventing spurious activation. For instance, studies demonstrate that MeCP2-mediated recruitment of these complexes directly represses transcription from methylated templates. At promoters, particularly those associated with CpG islands, DNA hypermethylation correlates strongly with , while hypomethylation characterizes active transcription. Hypermethylated promoters recruit repressive complexes that inhibit initiation and elongation, as observed in genome-wide analyses where densely methylated CpG islands show near-complete transcriptional shutdown. Conversely, unmethylated promoters facilitate open and access, enabling expression of housekeeping and lineage-specific genes. This inverse relationship is a hallmark of epigenetic , with promoter methylation patterns dynamically shifting during to lock in committed states. Enhancer methylation plays a more dynamic role, influencing cell-type specificity by modulating distal regulatory elements that loop to promoters. Unlike promoters, enhancers often exhibit intermediate methylation levels that correlate with tissue-specific activity; low methylation at active enhancers permits transcription factor binding and enhancer-promoter interactions, whereas higher methylation restricts these contacts. Genome-wide profiling reveals that cell-type-specific enhancers are hypomethylated in relevant lineages, such as liver-specific enhancers in hepatocytes, underscoring methylation's role in fine-tuning expression programs without global repression. This plasticity allows enhancers to respond to developmental cues, integrating signals for precise gene activation. In stem cells, bivalent promoters—marked by both active and repressive —often remain unmethylated, maintaining a poised state for rapid or repression during . This bivalency poises developmental genes for timely expression, with DNA hypomethylation at these sites preventing premature silencing and allowing to shield against de novo . Upon lineage commitment, selective methylation gain at formerly bivalent promoters resolves this poise into stable repression or , as seen in embryonic stem cells where thousands of such domains regulate pluripotency networks. This interplay ensures developmental flexibility while safeguarding against .

Transposable Element Silencing

DNA methylation plays a critical role in silencing (TEs) by methylating their promoters, which prevents transcription and mobilization within the genome. This epigenetic modification establishes heritable repression, particularly in cells where TE activity poses a high risk of . In mammals, Piwi-interacting RNAs () guide the de novo DNA methyltransferases DNMT3A and DNMT3L to TE loci during , ensuring targeted silencing before zygotic genome activation. This -directed mechanism is essential for maintaining fertility and preventing genomic instability, as disruptions in piRNA biogenesis lead to derepression and TE proliferation. Major classes of TEs affected by this silencing include long interspersed nuclear elements (LINEs), short interspersed nuclear elements (SINEs), and long terminal repeat (LTR) retrotransposons, which collectively comprise approximately 46% of the human genome. LINE-1 elements, the most abundant autonomous TEs, occupy about 17% of the genome and are particularly prone to retrotransposition if not methylated, while SINEs like Alu sequences and LTR elements from endogenous retroviruses add to the repetitive burden. Methylation at CpG islands within these TE promoters blocks RNA polymerase recruitment, reducing their transcriptional output by over 90% in somatic and germ cells. This widespread suppression not only curbs TE expansion but also prevents secondary effects like chromosomal rearrangements. The role of DNA methylation in TE silencing is evolutionarily conserved across eukaryotes, serving as a primary defense against insertional mutagenesis that could disrupt function or cause sterility. In vertebrates and , this mechanism has facilitated expansion by tolerating TE accumulation while keeping them inert, with losses in methyltransferase activity correlating to increased TE activity and reduced fitness. Defects in TE silencing pathways exemplify this conservation, underscoring the selective pressure for robust TE control throughout . DNA methylation interacts closely with histone modifications to reinforce TE silencing, particularly through trimethylation of histone H3 at lysine 9 (H3K9me3), which compacts and recruits methyl-binding proteins. In many species, H3K9me3 marks precede or coincide with DNA methylation at TE loci, creating a self-reinforcing loop where histone methyltransferases like SETDB1 deposit H3K9me3, facilitating DNMT recruitment and vice versa. This crosstalk ensures stable, multi-generational repression, as seen in mammalian pericentromeric where both marks colocalize at ~80% of silenced TEs. Disruptions in either pathway compromise the other, highlighting their interdependence for defense.

Genomic Imprinting and Stability

is an epigenetic phenomenon in mammals where certain genes are expressed in a parent-of-origin-specific manner, largely mediated by DNA methylation at imprinting control regions (ICRs). These ICRs are CpG-rich sequences that acquire methylation marks during , leading to monoallelic expression in the offspring. For instance, at the IGF2/H19 locus on mouse chromosome 7 ( 11p15.5), the paternal is methylated at the H19 ICR, which blocks binding of the insulator protein , allowing the shared enhancers to activate the upstream IGF2 gene while repressing H19; conversely, the unmethylated maternal ICR permits CTCF binding, insulating IGF2 from the enhancers and enabling H19 expression. In female mammals, DNA methylation also plays a critical role in X-chromosome inactivation (XCI), a dosage compensation that silences one of the two to equalize with males. This process is initiated by the long non-coding RNA , which coats the future inactive (Xi) in cis, recruiting silencing factors that lead to the spread of repressive histone modifications and subsequent DNA hypermethylation at gene promoters across the Xi. Methylation establishment occurs progressively after Xist coating, stabilizing the inactive state and preventing reactivation, with methyltransferases like DNMT3A and DNMT3B targeting CpG islands to maintain lifelong silencing. Beyond gene regulation, DNA methylation contributes to genomic stability by suppressing recombination at repetitive chromosomal regions. At centromeres, high levels of CpG methylation in pericentromeric inhibit illegitimate recombination and maintain proper assembly during ; loss of this methylation, as seen in DNMT1-deficient cells, increases centromeric recombination rates and chromosomal . Similarly, methylation in subtelomeric regions helps prevent recombination between s, preserving end-to-end avoidance and telomere homeostasis, with hypomethylation linked to elevated telomere dysfunction and genomic rearrangements. For ribosomal DNA () arrays, methylation at intergenic spacers and promoters regulates copy number by silencing redundant units, preventing unequal recombination that could amplify or contract the array; studies show a positive correlation between rDNA methylation levels and stable copy number maintenance across divisions. During , DNA methylation patterns undergo and re-establishment to reset imprints for the next generation. In early embryos, following fertilization, the paternal genome experiences active demethylation via TET3-mediated oxidation of to , while the maternal genome undergoes passive dilution through replication without maintenance methylation, resulting in global hypomethylation by the stage. Imprinted loci are protected from this by factors like , preserving parent-specific marks. methylation is then re-established post-implantation around embryonic day 6.5 in mice (equivalent to weeks 7-9 in humans), driven by DNMT3A/3L complexes, to reinstate gametic imprints and ensure stable transmission.

Occurrence Across Organisms

In Mammals

In mammals, DNA methylation exhibits distinct evolutionary adaptations that distinguish it from other vertebrates, particularly through the expansion of the DNMT3 subfamily of de novo methyltransferases. This expansion, which includes the emergence of DNMT3A, DNMT3B, and the regulatory cofactor DNMT3L, occurred during evolution and is linked to increased gene copy numbers, enabling more precise control over methylation patterns essential for mammalian-specific processes like . The mammalian genome contains approximately 28 million CpG sites, with about 60% typically methylated, providing a vast substrate for these enzymes to establish - and development-specific epigenomes. This evolutionary refinement supports the complexity of mammalian development, where DNMT3A2, a short isoform of DNMT3A, plays a conserved role across all mammalian groups in imprinting and de novo methylation. Tissue-specific DNA methylation profiles in mammals vary significantly, reflecting functional adaptations to diverse physiological demands. In the , particularly in neurons, the methylome shows notable hypomethylation, which is dynamically altered by neuronal activity; for instance, sensory stimuli can rapidly induce demethylation at activity-responsive genes, facilitating experience-dependent without . This contrasts with oocytes, where hypermethylation predominates in gene bodies of actively transcribed regions during growth, driven by transcription-coupled DNMT3A activity to establish oocyte-specific patterns that persist into early embryogenesis. Such tissue contrasts underscore how gradients—hypomethylated in dynamic neural tissues and hypermethylated in gametic cells—contribute to mammalian organ specialization and reproductive fidelity. During mammalian development, DNA methylation undergoes programmed waves that reset and reprogram the epigenome across generations. In , methylation is erased in primordial germ cells, followed by de novo establishment in prospermatogonia around embryonic day 14.5 in mice, with -directed targeting of transposable elements via MIWI2 guiding DNMT3L/DNMT3A complexes to silence retrotransposons. mirrors this with erasure in primordial germ cells and de novo waves during oocyte growth from the fetal stage through postnatal diplotene arrest, achieving near-complete methylation by maturity, though without prominent piRNA involvement. These biphasic waves ensure erasure of parental imprints and acquisition of gamete-specific marks, critical for totipotency and preventing transgenerational epigenetic defects. Non-CpG , particularly at dinucleotides, represents a unique feature of the mammalian epigenome, predominantly occurring in embryonic stem cells (ESCs) where it constitutes up to 25% of total methylation. Mediated by DNMT3A and DNMT3B, this methylation targets bodies and intergenic regions in pluripotent cells, potentially stabilizing pluripotency networks by repressing developmental genes. Levels decline sharply post-differentiation, dropping to less than 10% in lineages like neurons, as cells commit to specific fates and rely more on CpG methylation for stable repression. This transient non-CpG mark highlights a mammalian-specific layer of epigenetic flexibility during early development.

In Plants

In plants, DNA methylation occurs primarily in three sequence contexts: CG, CHG, and CHH (where H represents A, C, or T), with methylation levels covering approximately 20-30% of the genome, largely due to the proliferation of transposable elements (TEs) that constitute a significant portion of plant genomes. This extensive methylation serves as a key mechanism for TE silencing, adapting to the sessile lifestyle of plants by maintaining genome stability amid environmental pressures and developmental needs. The RNA-dependent DNA methylation (RdDM) pathway is a of plant epigenetics, guiding methylation through 24-nucleotide small interfering RNAs (siRNAs) that target TEs and repetitive sequences. In this process, plant-specific RNA polymerases IV and V produce precursor transcripts and scaffold RNAs, respectively, which facilitate siRNA loading onto proteins and recruitment of methyltransferases to homologous DNA loci, primarily establishing CHH methylation. RdDM operates iteratively to reinforce silencing, particularly at TE edges near genes, preventing deleterious insertions during stress or . Maintenance of methylation involves specialized enzymes: MET1, a CG-specific methyltransferase homologous to mammalian , perpetuates symmetric CG methylation across cell divisions by recognizing hemimethylated DNA post-replication. CMT3, a chromomethylase, maintains CHG methylation in a strand-symmetric manner, often coupled with lysine 9 dimethylation (H3K9me2) at pericentromeric . For asymmetric CHH contexts, DRM2 catalyzes de novo and maintenance , relying on RdDM for targeting and exhibiting activity across all contexts but predominantly in CHH. These enzymes collectively ensure context-specific fidelity, with disruptions leading to TE reactivation and . Beyond TE control, DNA contributes to adaptive functions such as epiallele , where stable methylation variants at loci influence hybrid vigor () by modulating differences between parental alleles. For instance, in hybrids, parental methylation states at epialleles correlate with enhanced growth and yield, transmitted meiotically without sequence changes. In stress responses, induces targeted demethylation at promoter regions of tolerance genes, such as those involved in signaling, enabling rapid transcriptional activation and in crops like and . This dynamic remodeling underscores methylation's role in environmental , with evolutionary pressures from TE expansion driving higher baseline levels compared to animals.

In Bacteria

In bacteria, DNA methylation primarily serves as a defense mechanism against invading foreign DNA, such as from bacteriophages, through restriction-modification (RM) systems. These systems consist of a restriction endonuclease that cleaves unmethylated DNA at specific recognition sequences and a cognate methyltransferase that protects the host by adding methyl groups to the same sequences. For instance, the Type II RM system , derived from , methylates the adenine in the sequence GAATTC, thereby shielding host DNA while allowing the endonuclease to degrade unmodified phage DNA. RM systems are classified into Types I, II, III, and IV based on their composition, sequence specificity, and mechanism; Type II systems, like , are the most common and act independently without requiring additional host factors for restriction. Beyond defense, RM systems can influence bacterial and phase variation, where reversible switches in occur due to changes in patterns. In some bacteria, site-specific by RM methyltransferases regulates the expression of virulence genes; for example, in , DNA methyltransferase () modulates the expression of the inv gene encoding invasin, a key adhesin that promotes bacterial of host epithelial cells, with altered levels affecting invasion efficiency. Phase variation mediated by RM systems often involves stochastic expression of the methyltransferase, leading to heterogeneous states across a population that enable adaptive responses to environmental pressures, such as host immune evasion. A prominent example of solitary methylation in is the methylase in E. coli, which N6-methylates in GATC sequences, occurring approximately every 256 base pairs in the . This is crucial for strand discrimination in post-replicative mismatch repair, where the transient hemimethylation of newly synthesized DNA directs the MutH endonuclease to nick the unmethylated strand, facilitating correction of replication errors by the MutS/MutL complex. methylation also regulates timing by controlling the sequestration of the (), where fully methylated allows initiation, while hemimethylation leads to binding of SeqA protein, delaying re-replication to ensure once-per-cell-cycle firing. Orphan methyltransferases, which lack paired restriction enzymes, perform regulatory functions independent of . These enzymes methylate specific motifs to influence processes like , , and progression; for instance, in various Gram-positive and , orphan methylases such as those targeting non-palindromic sequences modulate production and adaptation to stress without endonuclease activity. In pathogens, orphan can create epigenetic mosaics that drive population-level heterogeneity, enhancing survival in diverse niches like the host gut.

In Other Eukaryotes

In fungi such as , DNA methylation primarily mediates through the DIM-2, which is responsible for all known methylation in vegetative tissues. This process often follows repeat-induced (RIP), a pre-meiotic mechanism that introduces GC-to-AT transitions in duplicated sequences, leading to subsequent methylation and epigenetic silencing of repetitive elements. DIM-2 interacts with (HP1) to establish and maintain these silenced states, contributing to genomic stability. In insects, DNA methylation levels vary but are generally low compared to vertebrates. For instance, exhibits minimal genome-wide methylation (approximately 0.03–1% of cytosines), lacking a homolog of the maintenance methyltransferase and relying instead on the debated DNMT2 enzyme, which may primarily target . In the germline, piRNA pathways guide transposon silencing mainly through modifications rather than extensive DNA methylation, though low-level cytosine methylation occurs at specific loci. In contrast, social insects like the honeybee (Apis mellifera) show higher methylation associated with caste differentiation, where differential methylation patterns in larvae influence , promoting worker or queen development through nutritional cues. Protists display highly variable DNA methylation patterns, often differing from the canonical (5mC) seen in higher eukaryotes. In Tetrahymena thermophila, for example, N6-methyladenine (6mA) predominates at levels of 0.6–0.8% of s, particularly during conjugation and macronuclear development, where it influences positioning and organization without detectable 5mC. This adenine methylation supports programmed DNA elimination and genome restructuring essential for . Overall, DNA methylation in these non-mammalian, non-plant eukaryotes exhibits intermediate complexity, bridging bacterial restriction-modification systems and the more elaborate patterns in mammals, with roles in repeats and adapting to needs through diverse enzymatic and guiding mechanisms.

Roles in Development and Physiology

Embryonic Development and Differentiation

During mammalian embryogenesis, DNA methylation undergoes profound to reset epigenetic marks inherited from gametes and establish new patterns essential for development. Immediately following fertilization, the experiences two waves of global demethylation: an active process mediated by TET3, which oxidizes to primarily on the paternal , and a passive dilution through DNA replication-dependent mechanisms affecting both parental genomes before the first mitotic division. This demethylation erases most somatic methylation patterns, except at imprinting control regions, preparing the for totipotency. By the stage, de novo methylation is re-established by DNMT3A and DNMT3B enzymes, which, in cooperation with DNMT3L, target CpG islands and regulatory elements to reinstate methylation profiles critical for lineage specification. In embryonic stem cells (ESCs) derived from the of the , DNA methylation patterns support pluripotency by maintaining a bivalent epigenetic state. Enhancers associated with pluripotency genes, such as Oct4 and Nanog, are typically hypomethylated, allowing poised accessibility for transcription factors that drive self-renewal. Conversely, promoters and enhancers of lineage-specific genes, including those for mesodermal or ectodermal , are hypermethylated in ESCs, silencing them to prevent premature commitment and preserve multipotency. This selective methylation landscape ensures that ESCs remain undifferentiated until external signals trigger fate decisions. As embryogenesis progresses to and formation, DNA methylation dynamics shift to lock in cell identities during . Progressive de novo methylation by DNMT3A and DNMT3B stabilizes repressed states at non-lineage genes, while targeted demethylation—often via TET-mediated oxidation—activates lineage-appropriate genes; for instance, neuronal genes like Neurod1 undergo demethylation at their promoters and enhancers in neuroblasts, enabling . This reciprocal regulation ensures stable cell fate transitions, with hypermethylation reinforcing barriers against alternative lineages and hypomethylation permitting tissue-specific . Aberrant methylation during these reprogramming events can disrupt development, particularly affecting imprinted loci and leading to disorders like Beckwith-Wiedemann syndrome (BWS). In BWS, loss of at the KvDMR1 imprinting control region on chromosome 11p15 results in biallelic expression of IGF2 and silencing of CDKN1C, causing fetal overgrowth and increased tumor risk. Such epimutations highlight the precision required in embryonic methylation waves to maintain and normal differentiation.

Aging and Epigenetic Clocks

During aging, DNA methylation patterns undergo characteristic alterations, including global hypomethylation particularly at repetitive elements such as transposable elements and tandem repeats, alongside site-specific hypermethylation at promoter regions of certain genes. These changes contribute to epigenetic drift, a progressive divergence from youthful methylation profiles that correlates with chronological age across tissues. A 2025 of over 15,000 methylomes from 17 tissues revealed both conserved age-related methylation shifts and pronounced tissue-specific gradients, with the , liver, and lungs showing the most substantial changes, highlighting organ-level variations in epigenetic aging trajectories. Epigenetic clocks leverage these age-associated methylation patterns to estimate biological from DNA levels at specific CpG sites. The seminal Horvath clock, developed in 2013, uses 353 CpG sites to predict chronological across diverse tissues and types with high accuracy, serving as a pan-tissue of cumulative epigenetic effects. Second-generation clocks, such as DNA methylation PhenoAge (DNAm PhenoAge), extend this by incorporating phenotypic measures like biomarkers to estimate phenotypic , which better reflects healthspan and mortality beyond chronological alone. These methylation alterations arise from mechanistic processes exacerbated by aging, including that promotes the oxidation of (5mC) to (5hmC) via TET enzymes, leading to passive demethylation as 5hmC is poorly recognized by maintenance methyltransferase DNMT1. Additionally, replication errors during contribute to the accumulation of hypomethylation, as imperfect fidelity in DNMT1-mediated maintenance methylation allows stochastic losses at repetitive regions over successive divisions. Interventions targeting these processes can modulate epigenetic aging. Caloric restriction has been shown to slow the rate of epigenetic age acceleration, as evidenced by reduced methylation drift and slower progression of epigenetic clocks in long-term human trials. Recent studies on senolytics, such as plus or like , demonstrate reversal of methylation age by clearing senescent cells that propagate aberrant methylation patterns, with senolytics like plus showing effects in mouse models.

Response to Environmental Factors

DNA methylation plays a crucial role in mediating physiological adaptations to environmental stimuli, enabling cells to respond dynamically to external pressures such as , nutritional status, and stressors. These epigenetic modifications allow for rapid, reversible changes in without altering the underlying DNA sequence, facilitating in response to acute or chronic exposures. In response to exercise, particularly aerobic , skeletal muscle exhibits site-specific DNA hypomethylation at promoters of metabolic genes, enhancing their expression to support energy demands and . For instance, endurance exercise induces hypomethylation of the (PGC-1α) gene promoter in human , correlating with increased PGC-1α transcription and improved oxidative capacity. This hypomethylation is observed as an early adaptive response, occurring within hours to days of onset, and contributes to the muscle's metabolic . Similar patterns occur at other loci, such as those regulating and fatty acid oxidation, underscoring exercise's role in epigenetic tuning for physical performance. Dietary factors and environmental toxins profoundly influence the DNA methylation landscape by altering the availability of methyl donors. impairs the synthesis of S-adenosylmethionine (), the primary methyl group donor for s, leading to global and gene-specific hypomethylation that disrupts genomic stability. This reduction in SAM pools, often seen in or conditions, results in decreased at CpG islands, potentially activating retrotransposons and altering gene regulation. Conversely, exposure to cigarette smoke promotes hypermethylation of promoters, such as p16INK4A and , through upregulation of 1 () and accumulation of tobacco-specific carcinogens like , which enhance methylation machinery activity in lung epithelial cells. These changes reflect the body's attempt to silence potentially harmful genes in response to toxic insults. Chronic stress triggers glucocorticoid-mediated alterations in DNA methylation, particularly within the hypothalamic-pituitary-adrenal () axis, to fine-tune the response. Exposure to elevated glucocorticoids, such as , induces hypermethylation at the NR3C1 () promoter, reducing receptor expression and impairing regulation of the axis, which can perpetuate heightened reactivity. These methylation shifts occur in regions like the and are reversible upon cessation, allowing for adaptive recalibration of neuroendocrine signaling. Glucocorticoids also promote dynamic demethylation at stress-responsive loci, facilitating rapid activation in immune and neuronal cells. Recent studies from 2024 and 2025 have highlighted how exposure alters brain DNA patterns, contributing to and increased risk of neurological disorders such as . Fine (PM2.5) induces differential in hippocampal and cortical regions, affecting genes involved in and , as observed in models and analyses of biomarkers. For example, PM2.5 exposure has been associated with altered at CpG sites linked to Alzheimer's . These findings underscore the role of epigenetic changes in mediating the neurotoxic effects of chronic pollutant exposure.

Implications in Disease

In Cancer

Aberrant DNA methylation patterns are a hallmark of cancer, characterized primarily by global hypomethylation of the and focal hypermethylation of promoters. Global hypomethylation, often observed in repetitive elements like LINE-1 sequences, leads to chromosomal instability by promoting illegitimate recombination and activation of transposable elements, thereby accelerating tumor initiation and progression. For instance, acute hypomethylation in models results in a more than six-fold increase in formation due to enhanced at tumor suppressor loci. In contrast, promoter hypermethylation silences tumor suppressor , contributing to oncogenesis; a prominent example is the hypermethylation of the O6-methylguanine-DNA methyltransferase () promoter in gliomas, which impairs and sensitizes tumors to alkylating agents like , associating with improved patient survival in newly diagnosed . Epigenetic drift, involving stochastic changes in DNA methylation over time, manifests as field defects in precancerous tissues, where adjacent normal-appearing cells exhibit tumor-like methylation alterations, predisposing regions to . These effects, detected via genome-wide methylation profiling, highlight early epigenetic reprogramming in tissues like the and colon, correlating with increased cancer risk before histological changes occur. In , hypermethylation of the BRCA1 promoter exemplifies this by functionally mimicking mutations, leading to BRCAness phenotype with deficiency and heightened sensitivity to , independent of genetic alterations. Recent advances in 2025 have leveraged DNA methylation profiling for precise tumor classification, particularly in (CNS) tumors. The CNS Tumor Methylation Classifier version 12.8, trained on 7,495 profiles, expands to 184 subclasses with 95% accuracy, enabling hierarchical diagnosis that outperforms WHO classifications and supports personalized neuro-oncological strategies. Therapeutic targeting of DNA methylation has advanced with (DNMT) inhibitors, such as , approved for myelodysplastic syndromes (MDS) and (AML). In the AZA-001 trial, azacitidine extended median survival to 24.5 months versus 15 months with conventional care in high-risk MDS, achieving complete response in 20% of patients. Combinations with enhance efficacy; low-dose azacitidine upregulates tumor-associated antigens and expression, improving responses to PD-1 inhibitors in solid tumors and hematological malignancies by reprogramming the .

In Neurological and Cardiovascular Disorders

DNA methylation dysregulation plays a significant role in various neurological disorders, particularly through alterations in that affect neuronal function and plasticity. In , hypermethylation of the (BDNF) promoter has been consistently observed, leading to reduced BDNF expression and impaired neurotrophic support in affected individuals. This epigenetic modification is associated with the pathophysiology of (MDD), where peripheral blood mononuclear cells from MDD patients show significantly higher BDNF promoter compared to healthy controls. Similarly, recent investigations into substance use disorders highlight 's impact on patterns in reward pathways. A 2025 study revealed that exposure induces dynamic transcriptomic changes in dopamine D2 receptor (Drd2)-expressing medium spiny neurons within the , a key reward circuit region, potentially involving -mediated during development and withdrawal. These findings build on earlier evidence that DNA in the modulates incubation of craving, underscoring its role in long-term addictive behaviors. In neurodevelopmental disorders, in the methyl-CpG-binding protein 2 (MECP2) exemplify how disruptions in reading impair brain function. , a severe neurodevelopmental condition, arises primarily from loss-of-function MECP2 that compromise MeCP2's ability to bind methylated CpG sites and repress transcription, resulting in widespread dysregulation and neurological symptoms such as seizures and motor impairments. These , found in up to 80% of classic cases, alter MeCP2's interaction with methylated DNA, leading to aberrant expression of target critical for neuronal maturation. mechanisms further intersect with these pathways, as opioid exposure induces aberrant DNA at the mu-opioid receptor (OPRM1) promoter. Chronic opioid use correlates with increased OPRM1 promoter in lymphocytes of addicts and medication users, potentially silencing receptor expression and contributing to and dependence. This hypermethylation pattern is replicated in preclinical models, where it associates with heightened neonatal abstinence syndrome severity in -exposed infants. Shifting to cardiovascular disorders, aberrant DNA methylation contributes to vascular by modulating genes involved in and endothelial function. In , hypermethylation of the (ESR1) promoter is linked to disease progression, with elevated levels positively correlating with increased ESR1 methylation and lesion severity in arterial tissues. This epigenetic silencing reduces ESR1 expression, impairing estrogen-mediated protection against plaque formation and promoting inflammatory responses in vascular cells. Peripheral analyses confirm differential ESR1 promoter methylation in females with , highlighting its potential as a for cardiovascular risk. In , tumor necrosis factor-alpha (TNF-α) signaling exacerbates cardiac dysfunction by inducing promoter hypermethylation of key genes like sarcoplasmic reticulum Ca²⁺-ATPase 2a (SERCA2a). Elevated TNF-α levels, common in failing hearts, upregulate DNA methyltransferases, enhancing SERCA2a promoter methylation and reducing its expression, which impairs calcium handling and contractility. This mechanism suggests that targeting methylation could mitigate TNF-α-driven remodeling in .

In Other Pathologies

DNA methylation alterations play a significant role in autoimmune diseases, particularly systemic lupus erythematosus (SLE), where hypomethylation of the gene (IFNG) in T cells contributes to dysregulated immune responses and hypersensitivity to interferon signaling. This hypomethylation leads to overexpression of IFNG and other interferon-responsive genes, correlating with higher disease activity and autoantibody production in SLE patients. Additionally, genetic variants in the 3B (DNMT3B) gene, such as the rs2424913 polymorphism, are associated with increased SLE susceptibility by impairing DNA methylation and exacerbating global hypomethylation in immune cells. In metabolic disorders, DNA methylation changes affect insulin production and function. Hypermethylation of the insulin (INS) gene promoter in of individuals with (T2D) inversely correlates with , contributing to impaired beta-cell function and reduced insulin secretion. In obesity, differential methylation at enhancer regions alters the expression of genes involved in and ; for instance, loss of DNMT3A-mediated methylation at distal enhancers disrupts differentiation and promotes morbid phenotypes. DNA methylation also influences host-pathogen interactions in infectious diseases, particularly through mechanisms that maintain viral latency or mount antiviral defenses. In Epstein-Barr virus (EBV) infection, hypermethylation of the latent membrane protein 1 (LMP1) promoter region in type I latency programs restricts lytic reactivation, enabling persistent viral presence in B cells while evading immune detection. Host cells employ DNA methylation as a defense strategy by hypermethylating invading genomes, such as viral DNA, to silence foreign and limit replication; this is evident in the epigenetic repression of integrated viral elements by host DNA methyltransferases. Recent advances highlight the prognostic value of DNA methylation in thyroid cancer, where panels assessing progressive hypomethylation patterns during metastatic progression of papillary and follicular thyroid carcinomas serve as biomarkers for risk stratification and outcome prediction. These methylation signatures, particularly at genes like ADM and RIN1, enable multi-omics integration for improved clinical management beyond traditional histopathology.

Detection and Technological Advances

Experimental Detection Methods

Bisulfite sequencing serves as the gold standard for detecting DNA methylation at single-base resolution due to its ability to distinguish methylated from unmethylated cytosines. The method involves treating genomic DNA with , which deaminates unmethylated cytosines to uracils (read as thymines during sequencing), while (5mC) remains unchanged, allowing for precise mapping via (PCR) amplification and next-generation sequencing (NGS). This approach enables whole-genome (WGBS) for comprehensive coverage or targeted sequencing for specific loci. However, the harsh chemical conditions of bisulfite conversion can cause DNA fragmentation and degradation, reducing input DNA efficiency and complicating analysis of low-abundance samples. To address the high cost and complexity of WGBS, enrichment-based methods focus on CpG-dense regions, which represent the majority of regulatory methylation sites. Reduced representation bisulfite sequencing (RRBS) employs the methylation-insensitive restriction enzyme MspI to digest DNA at CCGG sites, enriching for CpG islands and promoters before bisulfite treatment and sequencing; this covers approximately 1-2% of the genome but captures over 85% of unmethylated CpG islands with base-pair resolution at a fraction of WGBS cost. Methylated DNA immunoprecipitation (MeDIP) uses an anti-5mC antibody to pull down methylated fragments from sonicated DNA, followed by NGS, providing genome-wide methylation profiles with moderate resolution suitable for comparative studies across samples. These techniques prioritize conceptual mapping of methylation patterns over exhaustive coverage, though MeDIP may introduce biases from antibody affinity variations. Single-cell bisulfite sequencing (scBS-seq) extends these methods to individual cells, revealing epigenetic heterogeneity in tissues like tumors or developing embryos by isolating nuclei, performing conversion, and amplifying via whole-genome before NGS. Despite challenges such as low genomic coverage (typically 10-50%) and amplification biases, scBS-seq has become feasible for rare cell types. Advancements in 2024-2025, including optimized protocols and improved library preparation, enhance throughput to thousands of cells while reducing noise, enabling detailed studies of dynamics in heterogeneous populations. For detecting oxidation derivatives like 5-hydroxymethylcytosine (5hmC), which bisulfite sequencing conflates with 5mC, oxidation-based methods provide specificity. Tet-assisted bisulfite sequencing (TAB-seq) first protects 5hmC with beta-glucosyltransferase to form glucosylated 5hmC, then uses TET enzymes to oxidize unprotected 5mC to 5-carboxylcytosine (5caC); subsequent bisulfite treatment converts 5caC and unmethylated cytosines to uracils, leaving protected 5hmC intact for direct readout at single-base resolution. This approach has been pivotal in mapping 5hmC in mammalian genomes, though it requires high-quality enzymes and is less suited for low-input samples compared to bisulfite variants.

Computational Prediction and Analysis

Computational prediction and analysis of DNA methylation involve bioinformatics tools and models that infer methylation patterns from genomic sequences, experimental data, or integrated multi-omics inputs, enabling large-scale studies without exhaustive wet-lab validation. These approaches leverage sequence features such as CpG island proximity, accessibility, and evolutionary conservation to predict methylation probabilities at specific sites. For instance, models like DeepMethyl employ stacked denoising autoencoders to classify CpG dinucleotide methylation states, achieving high accuracy by incorporating topological features alongside sequence motifs. Such predictors are particularly useful for imputing missing methylation data in sparse datasets or simulating epigenetic landscapes . Analysis pipelines facilitate the interpretation of methylation data through association studies and clustering techniques. Epigenome-wide association studies (EWAS) scan genome-wide methylation profiles to identify loci associated with phenotypes like disease risk or environmental exposures, typically using models adjusted for confounders such as cell-type composition. In single-cell contexts, recent advancements, including transformer-based models like scMeFormer, enable imputation and clustering of methylation states across thousands of cells, revealing cell-type-specific epigenetic heterogeneity with improved resolution over traditional methods. These pipelines often integrate tools for , , and , supporting downstream applications in and . Epigenetic clocks, such as the Horvath clock, computationally estimate biological by regressing chronological against DNA beta values at a panel of 353 CpG sites using elastic net regression, providing a robust metric for aging research across tissues. This highlights hyper- and hypomethylated sites that correlate with age progression, with deviations (epigenetic age acceleration) linked to outcomes. For identifying differentially methylated regions (DMRs), tools like DSS apply statistical models based on beta-binomial distributions with dispersion shrinkage to detect methylation differences between conditions, offering superior power for data compared to non-shrunk alternatives. These methods prioritize regions with biological relevance, such as promoter-associated CpGs, while controlling for multiple testing via false discovery rates.

Emerging Applications in Diagnostics

DNA methylation profiling has emerged as a powerful tool in clinical diagnostics, enabling non-invasive assessment of disease states through the analysis of methylation patterns in cell-free DNA (cfDNA) and other biological samples. By leveraging tissue-specific and disease-associated methylation signatures, these applications facilitate early detection, risk assessment, and personalized treatment strategies across various fields, including oncology, geriatrics, and forensics. Advances in high-throughput sequencing and computational integration have accelerated the translation of methylation biomarkers into practical diagnostic panels as of 2025. In cancer diagnostics, liquid biopsies based on cfDNA methylation have revolutionized multi-cancer early detection. The Galleri test, developed by GRAIL, analyzes methylation patterns in cfDNA to identify signals from over 50 cancer types with high specificity, detecting cancers at stages I-III in clinical trials involving thousands of participants. This approach outperforms traditional screening by covering multiple organ sites simultaneously, with reported sensitivities exceeding 50% for high-mortality cancers like pancreatic and esophageal. Similarly, the TAPS (tet-assisted pyridine borane sequencing) method enables whole-genome methylation analysis of cfDNA, achieving sensitive multi-cancer detection in asymptomatic individuals through multimodal integration of methylation and fragmentomics data. These tools support clinical decision-making by predicting tumor origin and reducing unnecessary biopsies. For aging and , epigenetic clocks derived from DNA methylation serve as biomarkers for biological , aiding in stratification for age-related diseases. These clocks, such as the Horvath and GrimAge models, quantify accelerated aging by measuring methylation at specific CpG sites, correlating with increased mortality ; for instance, a five-year deviation in epigenetic age is associated with an 8-15% higher all-cause mortality hazard. In 2025, blood-based epigenetic clocks for intrinsic capacity have been validated to predict frailty, , and overall survival, integrating and immunological factors for personalized health interventions. Emerging EpiScores extend this to forecast exposure to environmental and disease susceptibility, such as predicting liver or respiratory conditions with improved accuracy over first-generation clocks. In forensics, DNA methylation markers enable body fluid identification and age estimation from trace evidence, enhancing investigative capabilities. Tissue-specific CpG sites hypermethylated in blood allow differentiation of fluids like semen, saliva, and vaginal secretions with over 95% accuracy using targeted bisulfite sequencing. For age prediction, models based on methylation at clock CpGs achieve mean absolute errors of 3-5 years across diverse populations, even from degraded samples like burnt remains. Recent 2025 advancements using Oxford Nanopore sequencing have integrated adaptive sampling to simultaneously profile age and body fluid markers, providing a streamlined forensic workflow for crime scene analysis. Therapeutic monitoring via DNA methylation tracks responses to DNA methyltransferase inhibitors (DNMTi) like , which reverse aberrant hypermethylation in cancers. In , methylation levels at promoter sites (e.g., in or ) serve as dynamic biomarkers, with demethylation post-DNMTi treatment serving as a predictor of response in patients. AI-integrated panels analyze methylation profiles alongside multi-omics data to enable precision medicine, forecasting and optimizing combination therapies with inhibitors. These approaches, validated in 2024-2025 clinical cohorts, enhance monitoring by detecting through cfDNA changes as early as one cycle into treatment.

References

  1. [1]
    DNA methylation and its basic function - PubMed - NIH
    Jul 11, 2012 · In the mammalian genome, DNA methylation is an epigenetic mechanism involving the transfer of a methyl group onto the C5 position of the ...
  2. [2]
    DNA methyl transferase 1: regulatory mechanisms and implications ...
    Establishment and maintenance of methylation patterns are mediated by a family of enzymes called DNA methyltransferases (DNMTs). The human genome contains three ...
  3. [3]
    DNA methylation in development and disease - PubMed Central
    This review touches on DNA methylation changes occurring during aging, prostate cancer and BPH. We also summarize research on how environmental toxins affect ...
  4. [4]
    Methylation - National Human Genome Research Institute
    Methylation is a chemical modification of DNA and other molecules that may be retained as cells divide to make more cells. When found in DNA, methylation ...
  5. [5]
    DNA Methylation in Mammals - PMC - NIH
    DNA across the genome of mammalian somatic tissues is methylated at 70%–80% of all CpG sites. With the continual advances in technology there has been a ...
  6. [6]
    DNA Methylation and Its Basic Function | Neuropsychopharmacology
    Jul 11, 2012 · DNA methylation regulates gene expression by recruiting proteins involved in gene repression or by inhibiting the binding of transcription factor(s) to DNA.
  7. [7]
    Mammalian Non-CpG Methylation: Stem Cells and Beyond - PubMed
    Nov 11, 2014 · For some time it has been known that non-CpG methylation is abundant in plants and present in mammalian embryonic stem cells, but non-CpG ...
  8. [8]
    Structural insight into maintenance methylation by mouse DNA ...
    The DNA methyltransferase Dnmt1 is responsible for the propagation of methylation patterns to the next generation via its preferential methylation of ...
  9. [9]
    Structural basis for activation of DNMT1 | Nature Communications
    Nov 21, 2022 · DNMT1 is an essential enzyme that maintains genomic DNA methylation, and its function is regulated by mechanisms that are not yet fully ...
  10. [10]
    Processive Methylation of Hemimethylated CpG Sites by Mouse ...
    DNA methyltransferase Dnmt1 ensures clonal transmission of lineage-specific DNA methylation patterns in a mammalian genome during replication.Experimental Procedures · Substrates For Dna... · ResultsMissing: seminal paper<|separator|>
  11. [11]
    Allosteric control of mammalian DNA methyltransferases – a new ...
    DNMT1 contains multiple DNA binding sites, which contribute to the allosteric regulation of its activity and specificity. Several early studies showed that the ...
  12. [12]
    Structural basis for DNMT3A-mediated de novo DNA methylation
    Here we report a 2.65-Å crystal structure of the DNMT3A-DNMT3L-DNA complex where two DNMT3A monomers simultaneously attack two CpG dinucleotides.
  13. [13]
    Comprehensive structure-function characterization of DNMT3B and ...
    Jul 3, 2020 · Mammalian DNA methylation patterns are mainly generated by two de novo DNA methyltransferases, DNMT3A and DNMT3B. The catalytically inactive ...
  14. [14]
    The DNA methyltransferase-like protein DNMT3L stimulates de novo ...
    Coexpression of DNMT3L with Dnmt3a resulted in a striking stimulation of de novo methylation by Dnmt3a.
  15. [15]
    Roles of TET and TDG in DNA demethylation in proliferating and ...
    Jun 22, 2021 · TET enzymes mediate DNA demethylation by oxidizing 5-methylcytosine (5mC) in DNA to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), ...
  16. [16]
    TET (Ten-eleven translocation) family proteins: structure, biological ...
    Aug 11, 2023 · Although DNA methylation is generally stable, it can be removed by active demethylation associated with TET dioxygenases (DNA replication- ...
  17. [17]
    The Impact of One Carbon Metabolism on Histone Methylation
    Aug 27, 2019 · One Carbon Metabolism Regulates SAM Levels. Methylation is mediated by the metabolite S-adenosyl methionine (SAM) (Mentch et al., 2015), the ...
  18. [18]
    Comprehensive analysis of CpG islands in human chromosomes 21 ...
    CpG islands are useful markers for genes in organisms containing 5-methylcytosine in their genomes. In addition, CpG islands located in the promoter regions ...
  19. [19]
    CpG islands and the regulation of transcription
    2010). Approximately 70% of annotated gene promoters are associated with a CGI, making this the most common promoter type in the vertebrate genome (Saxonov et ...
  20. [20]
    Human body epigenome maps reveal noncanonical DNA ... - Nature
    Jun 1, 2015 · Here we report high coverage methylomes that catalogue cytosine methylation in all contexts for the major human organ systems, integrated with matched ...
  21. [21]
    DNA methylation of distal regulatory sites characterizes ...
    Mar 12, 2013 · We performed a systematic analysis of DNA methylation sites connected with gene expression profilesacross normal and cancerous human genomes.
  22. [22]
    Decoding the function of bivalent chromatin in development and ...
    H3K4me3 at bivalent promoters—a product of the underlying DNA sequence—persists in nearly all cell types irrespective of gene expression and confers protection ...
  23. [23]
    How mammalian piRNAs instruct de novo DNA methylation ... - Nature
    Sep 7, 2020 · This research provided the first mechanistic insights into how piRNA directs de novo DNA methylation in male mammals.
  24. [24]
    The piRNA Pathway Guards the Germline Genome Against ...
    In mouse, DNA methylation plays an essential role in silencing of transposable elements (TEs). In mouse male germ cells, this silencing is established in a ...
  25. [25]
    Regulatory transposable elements in the encyclopedia of DNA ...
    Aug 31, 2024 · In humans, at least 46% of the ~3.1 GB haploid genome is derived from TEs. Most TEs in the human genome can be classified into LINE, SINE, LTR ...
  26. [26]
    LINE dancing in the human genome: transposable elements and ...
    Oct 27, 2009 · Transposable elements (TEs) occupy almost half, 46%, of the human genome, making the TE content of our genome one of the highest among mammals, ...
  27. [27]
    Roles of transposable elements and DNA methylation in the ... - PNAS
    The origins of CpG islands (CGIs) are not known. They are relatively short GC-rich regions of DNA with a higher-than-expected occurrence of CpG dinucleotides ...Abstract · Sign Up For Pnas Alerts · Results
  28. [28]
    Taming transposable elements in vertebrates: from epigenetic ...
    Eukaryotes have evolved an array of complex, interconnected epigenetic mechanisms to silence and control TEs: small RNAs, Krüppel-associated box (KRAB) domain- ...
  29. [29]
    piRNAs, transposon silencing, and Drosophila germline development
    Nov 29, 2010 · Recent studies indicate that transposon silencing during germline development is imposed by Piwi-interacting RNAs (piRNAs), which guide a small ...
  30. [30]
    DNA methylation pathways and their crosstalk with histone methylation
    Methylation of DNA and of histone 3 at Lys 9 (H3K9) are highly correlated with gene silencing in eukaryotes from fungi to humans. Both of these epigenetic ...
  31. [31]
    The interplay between DNA and histone methylation - EMBO Press
    Apr 12, 2021 · A strong correlation exists between the genome‐wide distribution of DNA and histone methylation, suggesting an intimate relationship between these epigenetic ...
  32. [32]
    X chromosome-wide analyses of genomic DNA methylation ... - PNAS
    X chromosome inactivation is initiated by a cis-acting noncoding RNA, XIST, and final DNA methylation is established at the promoters of genes on the inactive X ...Missing: seminal | Show results with:seminal
  33. [33]
    Keeping the Centromere under Control: A Promising Role for DNA ...
    Aug 16, 2019 · Among the effects of DNA methylation on the maintenance of genome stability is its proposed ability to control crossover and recombination at ...
  34. [34]
    Centromere mitotic recombination in mammalian cells - PMC - NIH
    In addition, we demonstrate that loss of DNA methylation leads to increased centromere recombination, suggesting that the epigenetic status of centromeric ...
  35. [35]
    Making it or breaking it: DNA methylation and genome integrity - PMC
    This review focuses on our current understanding of DNA methylation in maintaining genome integrity in mammalian cells.
  36. [36]
    Evolutionary history of DNA methylation related genes in chordates
    Jan 22, 2020 · Increased copy numbers of DNA methylation related genes (dnmt, tet and tdg) have been found during chordate evolution due to successive whole genome ...
  37. [37]
    Chromatin and sequence features that define the fine and gross ...
    The human genome contains ∼28 million CpG sites, about 60% of which are methylated at the 5 position of the cytosine. Methylation of relatively CpG-rich ...
  38. [38]
    Evolution of the Short Form of DNMT3A, DNMT3A2, Occurred in the ...
    Our results show that DNMT3A2 is present in all mammalian groups and suggests that the basic DNMT3A/3A2-based DNA methylation mechanism is conserved at least in ...
  39. [39]
    Neuronal activity modifies DNA methylation landscape in the adult ...
    Our study provides the first global view of how the DNA methylation landscape of mature neurons is rapidly modified in response to external stimuli in vivo and ...
  40. [40]
    The role and mechanisms of DNA methylation in the oocyte
    Nov 29, 2019 · Here, we review the current state of knowledge of de novo DNA methylation mechanisms in the oocyte: how a distinctive gene-body methylation pattern is created.
  41. [41]
    Tissue specific DNA methylation of CpG islands in normal human ...
    These data demonstrate consistent tissue specific methylation for certain CpG islands, with clear differences between white and grey matter of the brain.
  42. [42]
    Small RNA guides for de novo DNA methylation in mammalian germ ...
    Proposed mechanism for piRNA-induced DNA methylation in male germ cells. Primary piRNAs produced from TE-rich piRNA clusters join Piwi (Mili and/or Miwi2) ...
  43. [43]
    Developmental alterations in DNA methylation during ...
    Feb 18, 2022 · Developmental alterations and stage-specific DNA methylation during gametogenesis from primordial germ cells (PGCs) to mature sperm are investigated.
  44. [44]
    Establishment and functions of DNA methylation in the germline - PMC
    These piRNAs are derived from repeat sequences and direct methylation and silencing of transposable elements in male germline, as well as of Rasgfr1, a ...
  45. [45]
    Mammalian Non-CpG Methylation: Stem Cells and Beyond - PMC
    Nov 11, 2014 · The percentage of cytosine methylation attributed to non-CpG methylation gradually decreased as differentiation increased and only <10% was ...Missing: declining | Show results with:declining
  46. [46]
    The evidence for functional non-CpG methylation in mammalian cells
    In this review, we will describe evidence for the existence of non-CpG methylation, specifically in mammalian genomes, and discuss the differences between CpG ...
  47. [47]
    (PDF) Differential landscape of non-CpG methylation in embryonic ...
    Methylated non-CpGs (mCpH; H means A, C, and T) have emerged as key epigenetic marks in mammalian embryonic stem cells (ESCs) and neurons, regulating cell type- ...Missing: declining | Show results with:declining
  48. [48]
    DNA methylation in plants: mechanisms and tools for targeted ...
    Mar 11, 2020 · In plants, DNA methylation is found in the CG, CHG and CHH sequence context (where H is A, C or T), and it is highly enriched over ...Summary · Introduction · II. Mechanisms of DNA... · III. Targeted manipulation of...
  49. [49]
    DNA Methylation and the Evolution of Developmental Complexity in ...
    Oct 3, 2018 · All land plants so far examined use DNA methylation to silence transposons (TEs). DNA methylation therefore appears to have been co-opted in ...
  50. [50]
    RNA-directed DNA methylation and demethylation in plants - PubMed
    Apr 21, 2009 · RNA-directed DNA methylation (RdDM) is a nuclear process in which small interfering RNAs (siRNAs) direct the cytosine methylation of DNA ...
  51. [51]
    RNA-directed DNA Methylation | PLOS Genetics - Research journals
    Oct 8, 2020 · RNA-directed DNA methylation (RdDM) is a biological process in which non-coding RNA molecules direct the addition of DNA methylation to specific DNA sequences.
  52. [52]
    DNA methylation-free Arabidopsis reveals crucial roles of ... - Nature
    Mar 14, 2022 · 2c), indicating that MET1, DRM1, DRM2, CMT3, and CMT2 are responsible for the maintenance of the entire DNA methylation in the Arabidopsis ...
  53. [53]
    Role of the DRM and CMT3 Methyltransferases in RNA-Directed ...
    In this report, we study the role of the DRM and CMT3 DNA methyltransferase genes in the initiation and maintenance of RdDM.
  54. [54]
    Stable inheritance of DNA methylation allows ... - Genome Biology
    Aug 16, 2017 · Differences in DNA methylation can arise as epialleles, which are loci that differ in chromatin state and are inherited over generations.
  55. [55]
    Parental DNA Methylation States Are Associated with Heterosis in ...
    DNA methylation differences between isogenic parental lines can directly or indirectly trigger heterosis in Arabidopsis hybrids.Missing: epiallele | Show results with:epiallele
  56. [56]
    Drought-induced site-specific DNA methylation and its association ...
    Dec 30, 2010 · Our results indicate that drought-induced genome-wide DNA methylation changes accounted for ∼12.1% of the total site-specific methylation differences in the ...
  57. [57]
    DNA Methylation Dynamics in Response to Drought Stress in Crops
    As a conserved epigenetic regulation, DNA methylation dynamically alters gene expression and chromosome interactions in plants' response to abiotic stresses.
  58. [58]
    Diverse Functions of Restriction-Modification Systems in Addition to ...
    Mar 7, 2013 · R-M systems recognize the methylation status of incoming foreign DNA, e.g., phage genomes. Methylated sequences are recognized as self, while ...
  59. [59]
    Overproduction of DNA Adenine Methyltransferase Alters Motility ...
    We showed previously that for the human pathogen Yersinia enterocolitica, overproduction of Dam results in increased invasion of epithelial cells. Since ...Missing: variation | Show results with:variation
  60. [60]
  61. [61]
    The Escherichia coli dam DNA methyltransferase modifies ... - PubMed
    The Escherichia coli dam adenine-N6 methyltransferase modifies DNA at GATC sequences. It is involved in post-replicative mismatch repair, control of DNA ...
  62. [62]
    Importance of state of methylation of oriC GATC sites in initiation of ...
    May 1, 1985 · In vivo and in vitro evidence is presented implicating a function of GATC methylation in the Escherichia coli replication origin, oriC, ...<|separator|>
  63. [63]
    regulatory functions of DNA methylation outside of host defense in ...
    Oct 15, 2020 · In bacteria, the study of DNA methylation has largely been in the context of restriction-modification systems, where DNA methylation serves to ...Missing: seminal papers
  64. [64]
    Bacterial DNA methyltransferase: A key to the epigenetic world with ...
    In bacteria, DNA methylation is the main form of epigenetic regulation and plays important roles in affecting clinically relevant phenotypes.
  65. [65]
    DNA methylation inhibits elongation but not initiation of transcription ...
    (1997) Cytosine methylation associated with repeat-induced point mutation (RIP) causes epigenetic gene silencing in Neurospora crassa. Genetics 146:509–523 ...
  66. [66]
    Direct Interaction between DNA Methyltransferase DIM-2 and HP1 Is ...
    DNA methylation is involved in gene silencing and genomic stability in mammals, plants, and fungi. Genetics studies of Neurospora crassa have revealed that ...Plasmid Constructs For The... · Neurospora Strains And... · Results
  67. [67]
    DNA methylation in Drosophila--a critical evaluation - PubMed
    It has been suggested that DNA methylation in Drosophila is simply a byproduct of Dnmt2, which is a DNA methyltransferase (Dnmt) according to structure and type ...
  68. [68]
    Off-target piRNA gene silencing in Drosophila melanogaster ... - NIH
    The cause of this variation is poorly understood. Studies in Drosophila, where DNA methylation is absent and piRNAs are the primary line of defense against TEs ...
  69. [69]
    DNA methylation is widespread and associated with differential ...
    Jul 7, 2009 · DNA methylation is widespread and associated with differential gene expression in castes of the honeybee, Apis mellifera.
  70. [70]
    N6-methyldeoxyadenosine directs nucleosome positioning in ...
    Nov 19, 2018 · 5-Methylcytosine (5mC) has ... 5a), indicating that this gene is at least partially responsible for DNA 6mA methylation in Tetrahymena.
  71. [71]
    DNA synthesis, methylation and degradation during conjugation in ...
    Jan 11, 1985 · We have investigated the timing of DNA synthesis, methylation and degradation during macronuclear development in the ciliate, Tetrahymena thermophila.
  72. [72]
    DNA methylation and the formation of heterochromatin in ... - Nature
    Apr 21, 2010 · Studies of the control and function of DNA methylation in Neurospora crassa have led to a greater understanding of heterochromatin formation.
  73. [73]
    Promoter DNA Methylation Patterns of Differentiated Cells Are ...
    Apr 21, 2010 · We find that most lineage-specification promoters are DNA hypomethylated and harbor a combination of trimethylated H3K4 and H3K27, whereas early ...
  74. [74]
    Diagnosis and Management of Beckwith-Wiedemann Syndrome
    About 80% of patients with BWS have a known molecular defect in the 11p15 region, most commonly due to aberrant DNA methylation (1). Normally, the paternal ...Abstract · Molecular Mechanisms of BWS · Molecular Genetic Testing for...
  75. [75]
    Aging and DNA methylation - PMC - PubMed Central
    During aging, DNA methylation changes include global hypomethylation and local hypermethylation, which is a hallmark of aging.
  76. [76]
    DNA methylation dynamics in aging: how far are we from ...
    In particular, DNA repeats undergo a generalised hypomethylation, while locus specific epigenetic changes are observed in gene-associated regulatory regions.
  77. [77]
    Epigenetic regulation of aging: implications for interventions ... - Nature
    Nov 7, 2022 · Aging is often marked by global DNA hypomethylation, but hypermethylation also occurs at selective CpG islands. ... Widespread and tissue specific ...
  78. [78]
    DNA Methylation Ageing Atlas Across 17 Human Tissues - bioRxiv
    Jul 24, 2025 · We conducted a meta-analysis of more than 15,000 human methylomes spanning 17 tissues, identifying both conserved and tissue-specific aging ...
  79. [79]
    DNA methylation age of human tissues and cell types
    ... 353 CpG sites that together form an aging clock in terms of chromatin states and tissue variance. I propose that DNA methylation age measures the cumulative ...
  80. [80]
    An epigenetic biomarker of aging for lifespan and healthspan - PMC
    As expected, between 1998 and 2007, mean change in DNAm PhenoAge was 8.51 years, whereas mean change in clinical phenotypic age was 8.88 years. Moreover, ...
  81. [81]
    Mechanism and Function of Oxidative Reversal of DNA and RNA ...
    TET proteins can oxidize 5-methylcytosine (5mC) to generate 5-hydroxymethylcytosine (5hmC), which is recognized poorly by DNA methyltransferase 1 (DNMT1) and ...
  82. [82]
    Epigenetics, DNA damage, and aging - JCI
    Aug 15, 2022 · In this Review, we focus on age-related epigenetic changes and their interactions with age-related genomic changes that instigate these events.
  83. [83]
    Effect of long-term caloric restriction on DNA methylation measures ...
    Feb 9, 2023 · Caloric restriction delays age-related methylation drift. ... Dietary intervention modifies DNA methylation age assessed by the epigenetic clock.
  84. [84]
    Senolytic compounds reduce epigenetic age of blood samples in vitro
    Feb 4, 2025 · Of eight tested compounds, JQ1, RG7112, nutlin-3a, and AMG232 reduced epigenetic age, indicating that this approach may be useful in drug screening for ...
  85. [85]
    Exploring the effects of Dasatinib, Quercetin, and Fisetin on DNA ...
    This study aimed to assess the effects of Dasatinib and Quercetin (DQ) senolytic treatment on DNA methylation (DNAm), epigenetic age, and immune cell subsets.
  86. [86]
    Impact of Physical Activity and Exercise on the Epigenome in ...
    Exercise generally results in DNA hypomethylation in key skeletal muscle genes, representing an early response that mediates skeletal muscle adaptations to ...
  87. [87]
    Impact of aerobic exercise and fatty acid supplementation on global ...
    Exercise training studies have demonstrated hypomethylation of the genome following exercise in both skeletal muscle ... exercise on PPARGC1A DNA methylation.
  88. [88]
    Skeletal Muscle PGC1a 21 Nucleosome Position and 2260 nt DNA ...
    Specifically, we hypothesize that acute exercise leads to hypomethylation of skeletal muscle PGC1a and repo- sitioning of the PGC1a 21 N away from the regula-.
  89. [89]
    Folate Deficiency Induces Genomic Uracil Misincorporation and ...
    Folate deficiency can therefore decrease global DNA methylation, which is associated with genetic instability and tumor formation.
  90. [90]
    Impact of Folate Deficiency on DNA Stability - ScienceDirect.com
    As a consequence of folate deficiency, cellular SAM is depleted, which in turn induces DNA hypomethylation and potentially induces proto-oncogene expression ...
  91. [91]
    The tobacco-specific carcinogen NNK induces DNA ... - PubMed
    The tobacco-specific carcinogen NNK induces DNA methyltransferase 1 accumulation and tumor suppressor gene hypermethylation in mice and lung cancer patients.
  92. [92]
    Glucocorticoid receptor gene methylation and HPA-axis regulation ...
    Higher NR3C1 methylation levels have been associated with a reduced NR3C1 expression, possibly leading to impaired negative feedback regulation of the HPA-axis.
  93. [93]
    Glucocorticoid exposure during hippocampal neurogenesis primes ...
    Aug 9, 2019 · Our data suggest that early exposure to GCs can change the set point of future transcriptional responses to stress by inducing lasting DNAm changes.
  94. [94]
    Air Pollution-Induced Neurotoxicity: The Relationship Between Air ...
    Studies suggest that air pollution induces epigenetic changes. These epigenetic alterations may affect the expression of genes involved in stress responses, ...
  95. [95]
    [PDF] Differential DNA methylation in blood as potential mediator of the ...
    Apr 16, 2025 · The study found that nine DNA methylation sites mediated the link between PM2.5 and Alzheimer's biomarkers, with some related to ...
  96. [96]
    DNA hypomethylation contributes to genomic instability and ... - NIH
    Aberrant DNA methylation promotes cancer progression by the inactivation of tumor suppressor genes via promoter methylation. Additionally, global DNA ...Missing: review paper
  97. [97]
    MGMT Gene Silencing and Benefit from Temozolomide in ...
    Mar 10, 2005 · We found that MGMT promoter methylation is associated with a favorable outcome after temozolomide chemotherapy in patients with newly diagnosed ...
  98. [98]
    DNA Methylation and Field Cancerization - Taylor & Francis Online
    It has now been established that precancerous cells that are adjacent to the tumor cells harbor tumor-specific genetic alterations in various organs, including ...
  99. [99]
    Methylation of Breast Cancer Predisposition Genes in Early-Onset ...
    DNA methylation can mimic the effects of both germline and somatic mutations for cancer predisposition genes such as BRCA1 and p16INK4a.<|control11|><|separator|>
  100. [100]
    Advancing CNS tumor diagnostics with expanded DNA methylation ...
    May 29, 2025 · We introduce the Heidelberg CNS Tumor Methylation Classifier version 12.8 (v12.8), trained using 7,495 methylation profiles, thereby expanding ...
  101. [101]
    New Ways to Use DNA Methyltransferase Inhibitors for the ... - NIH
    The AZA-001 study firmly established azacitidine as the treatment of choice for patients with high-risk myelodysplastic syndrome (MDS) not proceeding to ...
  102. [102]
    DNA Methyltransferase Inhibitors: Catalysts For Antitumour Immune ...
    Dec 12, 2019 · DNMTi can induce the expression of tumour-associated antigens and regulate the activity of immune cells to improve the antitumour immune response.
  103. [103]
    The associations between DNA methylation and depression - PubMed
    Apr 14, 2023 · It is noted that DNA hypermethylation, namely BDNF and NR3C1, is associated with increased risk of depression.
  104. [104]
    DNA methylation and expression of stress related genes in PBMC of ...
    Feb 16, 2017 · We found that BDNF, FKBP5, CRHBP, and NR3C1 gene promoters were significantly hypermethylated in MDD patients with and without suicidal ideation.
  105. [105]
    Cocaine-induced gene regulation in D1 and D2 neuronal ... - Nature
    Jun 12, 2025 · Our findings provide a high-resolution transcriptomic map of D1 and D2 MSNs, illustrating the dynamic changes induced by cocaine exposure and withdrawal.
  106. [106]
    Role of DNA Methylation in the Nucleus Accumbens in Incubation of ...
    May 27, 2015 · These results demonstrate a role for NAc DNA methylation, and downstream targets of DNA demethylation, in incubation of cocaine craving. cocaine ...
  107. [107]
    DNA methylation and Rett syndrome - PubMed
    Rett syndrome is a neurological disorder caused by mutations in the MeCP2 protein, which has been shown to bind methylated DNA and repress transcription.
  108. [108]
    Rett syndrome: methyl-CpG-binding protein 2 mutations ... - PubMed
    Molecular studies have identified MECP2 mutations in up to 80% of classic RTT patients; mutation type has some effect on the phenotypic manifestation of RTT, ...
  109. [109]
    Chronic opioid use is associated with increased DNA methylation ...
    This suggests that opioids may stimulate DNA methylation. The OPRM1 methylation had no immediate effect on μ-opioid receptor transcription and was not ...
  110. [110]
    Opioid Exposure is Associated with Aberrant DNA Methylation of ...
    The μ-opioid receptor (OPRM1) plays an important role in opiate addiction. The OPRM1 gene promoter showed hypermethylation in lymphocytes of opiate addicts ...
  111. [111]
    Aberrant DNA methylation in the pathogenesis of atherosclerosis
    May 1, 2016 · Data showed that Hcy levels were positively correlated with ESR1 promoter methylation and lesion severity. Severe HHcy may result in the ...
  112. [112]
    Differential ESR1 Promoter Methylation in the Peripheral Blood ...
    May 21, 2020 · ... Methylation of the estrogen receptor gene is associated with aging and atherosclerosis in the cardiovascular system. Cardiovasc. Res. 1999 ...
  113. [113]
    Tumor necrosis factor-alpha decreases sarcoplasmic reticulum Ca2 ...
    Conclusions: TNF-alpha increased DNA methyltransferase levels, thus enhancing the methylation in the SERCA2a promoter region with a result of reducing SERCA2a.
  114. [114]
    Genome-Wide DNA Methylation Analysis of Systemic Lupus ...
    This hypomethylation suggests that lupus patients are hypersensitive to interferon, as DNA methylation is typically an inhibitor of gene expression. We also ...
  115. [115]
    what is important to know about DNA methylation and its role in SLE ...
    Jul 25, 2018 · Overall, hypomethylation of interferon-responsive genes is associated with higher disease activity, renal disease, as well as autoantibodies.
  116. [116]
    DNMT3B (rs2424913) polymorphism is associated with systemic ...
    In particular, MTHFR or DNMT3B polymorphisms have been associated with inflammatory diseases. The MTHFR and DNMT genes participate in the DNA methylation ...
  117. [117]
    Insulin promoter DNA methylation correlates negatively with ... - NIH
    This study demonstrates that DNA methylation of the insulin promoter is increased in patients with type 2 diabetes and correlates negatively with insulin gene ...Missing: hypermethylation | Show results with:hypermethylation
  118. [118]
    Integrative genomic analyses in adipocytes implicate DNA ... - Nature
    May 15, 2023 · Recent experimental studies demonstrate that manipulation of DNA methylation enzymes in adipocytes can induce or prevent obesity and T2D, ...
  119. [119]
    Constitutive loss of DNMT3A causes morbid obesity through ... - eLife
    May 30, 2022 · DNMT3A-mediated DNA methylation is required for adipose tissue development. To examine how loss of DNMT3A contributes to functional changes in ...
  120. [120]
    EBV Latency Programs: Molecular and Epigenetic Regulation and ...
    Jul 21, 2025 · In the type I latency, Cp and LMP1 promoter regions show elevated DNA methylation levels compared to latency III, mirroring the enrichment ...
  121. [121]
    Geminiviruses employ host DNA glycosylases to subvert ... - Nature
    Jan 31, 2022 · DNA methylation can act as a defense mechanism against DNA viruses, including geminiviruses. Viral genomic DNA can be methylated, and therefore ...Results · βc1 Interacts With Nbros1l · βc1 Promotes Dme Activity...
  122. [122]
    DNA Methylation Dynamics and Prognostic Implications in ... - PubMed
    Mar 6, 2025 · These results show a progressive increase in DNA methylation alterations, mainly hypomethylation, during PTC and FTC metastatic progression, suggesting a ...
  123. [123]
    Multi-Omics-Based Characterization of DNA Methylation ...
    ADM has been recognized as a prognostic biomarker for thyroid cancer since 2022, while RIN1 exhibited the most significant methylation changes and the greatest ...
  124. [124]
    DNA methylation detection: Bisulfite genomic sequencing analysis
    Bisulfite genomic sequencing is regarded as a gold-standard technology for detection of DNA methylation because it provides a qualitative, quantitative and ...
  125. [125]
    Comprehensive comparison of enzymatic and bisulfite DNA ...
    Oct 3, 2025 · Bisulfite conversion is considered the gold standard for DNA methylation analysis, but it damages DNA and performs sub-optimally with clinical ...
  126. [126]
    Clinical and public health research using methylated DNA ... - NIH
    MeDIP is a fast and simple approach to determine DNA methylation on a genome-wide scale and compare DNA methylation patterns between two samples with diversely ...
  127. [127]
  128. [128]
    Advances in Epigenomic Sequencing and Their Applications in ...
    May 21, 2025 · scBS-seq is an accurate and reproducible technique to characterize the methylome of rare cell types and heterogeneous populations. In scBS-seq, ...
  129. [129]
    Predicting DNA Methylation State of CpG Dinucleotide Using ...
    Jan 22, 2016 · DNA methylation occurs more frequently at CpG sites, where a guanine nucleotide follows a cytosine nucleotide in the sequence of the genome. In ...
  130. [130]
    DeepMethylation: a deep learning based framework with GloVe and ...
    Sep 25, 2023 · We propose DeepMethylation, a novel methylation predictor with deep learning. Specifically, the DNA sequence is encoded with word embedding and GloVe in the ...
  131. [131]
    Epigenome-wide association studies: current knowledge, strategies ...
    Dec 4, 2021 · Epigenome-wide association studies (EWASes) investigate the association between a phenotype and epigenetic variants, most commonly DNA methylation.
  132. [132]
    Deep learning imputes DNA methylation states in single cells and ...
    Mar 12, 2025 · We introduce scMeFormer, a transformer-based deep learning model for imputing DNAm states at each CpG site in single cells.Missing: scMethyl | Show results with:scMethyl
  133. [133]
    DSS - Bioconductor
    It detectes differentially expressed genes (DEGs) from RNA-seq, and differentially methylated loci or regions (DML/DMRs) from bisulfite sequencing (BS-seq). The ...
  134. [134]
    Differential methylation analysis for bisulfite sequencing using DSS
    Dec 7, 2019 · Bisulfite sequencing (BS-seq) technology measures DNA methylation at single nucleotide resolution. A key task in BS-seq data analysis is to ...
  135. [135]
    Multimodal cell-free DNA whole-genome TAPS is sensitive ... - Nature
    Jan 8, 2025 · The analysis of circulating tumour DNA (ctDNA) through minimally invasive liquid biopsies is promising for early multi-cancer detection and ...
  136. [136]
    Putting early cancer detection to the test - Nature
    Feb 21, 2025 · Others, including Galleri, examine DNA methylation, which exerts a potent regulatory effect on gene expression. Grail oncologist Eric Klein ...Missing: liquid | Show results with:liquid
  137. [137]
    A large-scale, multi-centre validation study of an AI-empowered ...
    Oct 8, 2025 · The test could detect 14 common cancer types, accounting for 72% of global cancer deaths, with sensitivities ranging from 38.9 to 83.3%.
  138. [138]
    Epigenetic Clocks and EpiScore for Preventive Medicine - PubMed
    May 21, 2025 · This review explores the clinical potential of epigenetic clocks and EpiScores, composite biomarkers that predict health risks and physiological status.
  139. [139]
    A blood-based epigenetic clock for intrinsic capacity predicts ...
    Jun 4, 2025 · A blood-based epigenetic clock for intrinsic capacity predicts mortality and is associated with clinical, immunological and lifestyle factors.
  140. [140]
    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.
  141. [141]
  142. [142]
    Uncovering Forensic Evidence: A Path to Age Estimation through ...
    Another objective of DNA methylation in forensics is to identify the type of body fluid. In crime scenes, it is possible to find samples of unknown origin or ...
  143. [143]
    DNA methylation-based forensic framework for age prediction and ...
    Recent research by de Bruin et al. (2024) has validated and assessed the performance of the Oxford Nanopore MinION for analyzing DNA methylation for forensic ...
  144. [144]
    None
    Nothing is retrieved...<|separator|>
  145. [145]
    DNA methylation and machine learning - Clinical Epigenetics
    Oct 10, 2025 · More advanced sequencing techniques, such as whole-genome bisulfite sequencing (WGBS), and reduced representation bisulfite sequencing (RRBS), ...
  146. [146]
    Multi-omics based and AI-driven drug repositioning for epigenetic ...
    For example, the DNMTi decitabine, initially approved to reverse abnormal DNA methylation of silenced genes related to cell proliferation and differentiation in ...