DNA methylation
DNA methylation is an epigenetic modification in which a methyl group (CH₃) is covalently attached to the fifth carbon of the cytosine base (C5 position) in a DNA molecule, primarily occurring at cytosine residues followed by guanine (CpG dinucleotides) in vertebrates.[1] This process does not alter the underlying DNA sequence but influences gene expression by generally repressing transcription, either through direct inhibition of transcription factor binding to DNA or by recruiting methyl-binding proteins that facilitate chromatin compaction and gene silencing.[1] Catalyzed by a family of enzymes known as DNA methyltransferases (DNMTs), including DNMT1 for maintenance of methylation patterns during DNA replication and DNMT3A/DNMT3B for establishing new (de novo) methylation marks, DNA methylation plays essential roles in embryonic development, genomic imprinting, X-chromosome inactivation, and suppression of transposable elements.[2] 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.[3] In mammals, DNA methylation is highly dynamic, with waves of reprogramming occurring in early embryogenesis and germline cells to reset epigenetic marks, underscoring its importance in cellular differentiation and inheritance of epigenetic states across generations.[4]
Fundamentals
Definition and Chemical Basis
DNA methylation is an epigenetic modification involving the covalent addition of a methyl group (CH₃) to the 5-position of cytosine bases within DNA, primarily occurring at CpG dinucleotides in vertebrates and resulting in the formation of 5-methylcytosine (5mC).[5] 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 stability and heritability across cell divisions.00071-3) First identified in 1948 by Rollin Hotchkiss through paper chromatography analysis of calf thymus DNA, where he detected an unusual fraction of modified cytosine comprising about 1% of total pyrimidines, DNA methylation has since become central to understanding gene regulation and cellular identity.77315-0/fulltext)
Chemically, the methylation reaction entails the enzymatic transfer of a methyl group from the universal donor S-adenosylmethionine (SAM) to the carbon-5 position of cytosine, yielding 5mC and S-adenosylhomocysteine (SAH) as a byproduct; this process is catalyzed by DNA methyltransferases.[6] In structural terms, cytosine consists of a pyrimidine 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.[5] 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
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 pyrimidine ring of cytosine (left) transforming to 5-methylcytosine (right) via addition at C5, maintaining the overall ring structure and hydrogen bonding capability with guanine.[6]
In mammalian somatic cells, DNA methylation is highly prevalent, affecting approximately 70-80% of all CpG sites across the genome, which contributes to the establishment of tissue-specific epigenetic landscapes.[5] While CpG methylation dominates in vertebrates, non-CpG methylation—occurring at CHG, CHH, or CpA/CpT contexts—is more common in plants, where it plays roles in transposon silencing, and is also observed in mammalian embryonic stem cells at levels up to 25% of total cytosine modifications.[7]
Enzymatic Machinery
DNA methylation is primarily catalyzed by a family of enzymes known as DNA methyltransferases (DNMTs), which transfer a methyl group from the cofactor S-adenosylmethionine (SAM) to the 5-position of cytosine bases in DNA. In mammals, the core DNMTs include DNMT1, responsible for maintenance methylation, and DNMT3A and DNMT3B, which mediate de novo methylation.81656-6)[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 cytosine into an active site pocket where methylation occurs using SAM as the methyl donor.[8][9] This process is highly processive, allowing DNMT1 to methylate multiple sites on the same DNA molecule without dissociation.[10] DNMT1 activity is allosterically regulated; for instance, unmethylated CpG sites can inhibit its methylation of hemimethylated DNA, enhancing specificity.[11]
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.[12][13] 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.[14]
DNA demethylation counters methylation through both passive and active mechanisms. Passive demethylation occurs during DNA replication when maintenance methylation by DNMT1 is impaired, leading to dilution of 5-methylcytosine (5mC) over successive cell divisions.[15] Active demethylation is mediated by ten-eleven translocation (TET) enzymes (TET1, TET2, and TET3), which are α-ketoglutarate and Fe²⁺-dependent dioxygenases that iteratively oxidize 5mC to 5-hydroxymethylcytosine (5hmC), then to 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC). These oxidized derivatives are recognized by thymine DNA glycosylase (TDG), initiating base excision repair to replace them with unmodified cytosines.[16][15]
The SAM cycle provides the universal methyl donor for DNMT activity, where SAM is synthesized from methionine and ATP, donates its methyl group to become S-adenosylhomocysteine, and is regenerated via homocysteine remethylation in a folate-dependent pathway.[17] Disruptions in this cycle can alter global methylation levels. Overall, the interplay of these enzymes and cofactors tightly controls DNA methylation dynamics.[6]
Methylation Patterns
DNA methylation exhibits distinct genomic distribution patterns, primarily occurring at cytosine residues within CpG dinucleotides in vertebrates, 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 vertebrate 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 housekeeping genes. The standard criteria for identifying CGIs include a minimum length of 200 base pairs, a GC content greater than 50%, and an observed-to-expected CpG ratio exceeding 0.6.[18][19][18]
In addition to promoter-associated methylation, gene body methylation—defined as methylation within transcribed exons and introns—shows a positive correlation with transcriptional activity. Highly expressed genes often exhibit elevated methylation levels across their gene bodies, which may help maintain efficient elongation by RNA polymerase II. Conversely, gene body methylation is inversely related to the prevalence of alternative splicing, with lowly methylated gene bodies associated with higher splicing complexity and exon skipping 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 plants, where it contributes to transposon silencing and heterochromatin 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 plants.
Globally, DNA methylation is enriched at repetitive elements, such as transposable elements and satellite DNA, where hypermethylation prevents their mobilization and ensures genomic stability. This hypermethylation affects over 90% of CpG sites in repeats across vertebrate genomes. Tissue-specific variations further modulate these patterns; for instance, brain tissues exhibit relative hypomethylation compared to other organs, particularly in neuronal populations, reflecting adaptations for plasticity and higher non-CpG content.[20]
Conserved Biological Functions
Gene Expression Regulation
DNA methylation primarily regulates gene expression 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 chromatin modifications, ensuring stable gene silencing without altering the underlying DNA sequence.[6]
The core mechanism of repression involves MeCP2 binding to methylated DNA, which recruits histone deacetylases (HDACs) to remove acetyl groups from histones, leading to chromatin condensation and reduced accessibility for transcriptional machinery. Additionally, MeCP2 associates with histone methyltransferases that deposit repressive marks, such as trimethylation of histone H3 at lysine 9 (H3K9me), promoting heterochromatin formation and blocking the binding of activator transcription factors. This dual action—deacetylation via HDACs and methylation via H3K9me—compacts chromatin structure, effectively silencing target genes and preventing spurious activation. For instance, in vitro 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 gene silencing, while hypomethylation characterizes active transcription. Hypermethylated promoters recruit repressive complexes that inhibit RNA polymerase II initiation and elongation, as observed in genome-wide analyses where densely methylated CpG islands show near-complete transcriptional shutdown. Conversely, unmethylated promoters facilitate open chromatin and transcription factor access, enabling expression of housekeeping and lineage-specific genes. This inverse relationship is a hallmark of epigenetic control, with promoter methylation patterns dynamically shifting during cell differentiation to lock in committed states.[6]
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.[21]
In stem cells, bivalent promoters—marked by both active H3K4me3 and repressive H3K27me3—often remain unmethylated, maintaining a poised state for rapid activation or repression during differentiation. This bivalency poises developmental genes for timely expression, with DNA hypomethylation at these sites preventing premature silencing and allowing H3K4me3 to shield against de novo methylation. Upon lineage commitment, selective methylation gain at formerly bivalent promoters resolves this poise into stable repression or activation, as seen in embryonic stem cells where thousands of such domains regulate pluripotency networks. This interplay ensures developmental flexibility while safeguarding against ectopic expression.[22]
Transposable Element Silencing
DNA methylation plays a critical role in silencing transposable elements (TEs) by methylating their promoters, which prevents transcription and mobilization within the genome. This epigenetic modification establishes heritable repression, particularly in germline cells where TE activity poses a high risk of insertional mutagenesis. In mammals, Piwi-interacting RNAs (piRNAs) guide the de novo DNA methyltransferases DNMT3A and DNMT3L to TE loci during gametogenesis, ensuring targeted silencing before zygotic genome activation. This piRNA-directed mechanism is essential for maintaining fertility and preventing genomic instability, as disruptions in piRNA biogenesis lead to derepression and TE proliferation.[23][24]
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.[25][26]
The role of DNA methylation in TE silencing is evolutionarily conserved across eukaryotes, serving as a primary defense against insertional mutagenesis that could disrupt gene function or cause sterility. In vertebrates and plants, this mechanism has facilitated genome 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 evolution.[27]
DNA methylation interacts closely with histone modifications to reinforce TE silencing, particularly through trimethylation of histone H3 at lysine 9 (H3K9me3), which compacts chromatin 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 heterochromatin where both marks colocalize at ~80% of silenced TEs. Disruptions in either pathway compromise the other, highlighting their interdependence for genome defense.[28][29]
Genomic Imprinting and Stability
Genomic imprinting is an epigenetic phenomenon in mammals where certain genes are expressed in a parent-of-origin-specific manner, largely mediated by differential DNA methylation at imprinting control regions (ICRs). These ICRs are CpG-rich sequences that acquire methylation marks during gametogenesis, leading to monoallelic expression in the offspring. For instance, at the IGF2/H19 locus on mouse chromosome 7 (human 11p15.5), the paternal allele is methylated at the H19 ICR, which blocks binding of the insulator protein CTCF, 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.[30]
In female mammals, DNA methylation also plays a critical role in X-chromosome inactivation (XCI), a dosage compensation mechanism that silences one of the two X chromosomes to equalize gene expression with males. This process is initiated by the long non-coding RNA Xist, which coats the future inactive X chromosome (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 de novo methyltransferases like DNMT3A and DNMT3B targeting CpG islands to maintain lifelong silencing.[31][32][33]
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 heterochromatin inhibit illegitimate recombination and maintain proper kinetochore assembly during mitosis; loss of this methylation, as seen in DNMT1-deficient cells, increases centromeric recombination rates and chromosomal instability. Similarly, methylation in subtelomeric regions helps prevent recombination between telomeres, preserving end-to-end fusion avoidance and telomere length homeostasis, with hypomethylation linked to elevated telomere dysfunction and genomic rearrangements. For ribosomal DNA (rDNA) 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 cell divisions.[34][35][36]
During mammalian reproduction, DNA methylation patterns undergo erasure 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 5-methylcytosine to 5-hydroxymethylcytosine, while the maternal genome undergoes passive dilution through replication without maintenance methylation, resulting in global hypomethylation by the blastocyst stage. Imprinted loci are protected from this erasure by factors like Stella, preserving parent-specific marks. De novo 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.[37]
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 chordate evolution and is linked to increased gene copy numbers, enabling more precise control over methylation patterns essential for mammalian-specific processes like genomic imprinting.[38] The mammalian genome contains approximately 28 million CpG sites, with about 60% typically methylated, providing a vast substrate for these enzymes to establish tissue- and development-specific epigenomes.[39] 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.[40]
Tissue-specific DNA methylation profiles in mammals vary significantly, reflecting functional adaptations to diverse physiological demands. In the brain, 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 plasticity without cell division.[41] 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.[42] Such tissue contrasts underscore how methylation gradients—hypomethylated in dynamic neural tissues and hypermethylated in gametic cells—contribute to mammalian organ specialization and reproductive fidelity.[43]
During mammalian germline development, DNA methylation undergoes programmed waves that reset and reprogram the epigenome across generations. In spermatogenesis, methylation is erased in primordial germ cells, followed by de novo establishment in prospermatogonia around embryonic day 14.5 in mice, with piRNA-directed targeting of transposable elements via MIWI2 guiding DNMT3L/DNMT3A complexes to silence retrotransposons.[44] Oogenesis 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.[45] These biphasic waves ensure erasure of parental imprints and acquisition of gamete-specific marks, critical for totipotency and preventing transgenerational epigenetic defects.[46]
Non-CpG methylation, particularly at CA dinucleotides, represents a unique feature of the mammalian epigenome, predominantly occurring in embryonic stem cells (ESCs) where it constitutes up to 25% of total cytosine methylation. Mediated by DNMT3A and DNMT3B, this methylation targets gene bodies and intergenic regions in pluripotent cells, potentially stabilizing pluripotency networks by repressing developmental genes.[47] 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.[48] This transient non-CpG mark highlights a mammalian-specific layer of epigenetic flexibility during early development.[49]
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.[50][51] 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.[51]
The RNA-dependent DNA methylation (RdDM) pathway is a distinctive feature of plant epigenetics, guiding de novo methylation through 24-nucleotide small interfering RNAs (siRNAs) that target TEs and repetitive sequences.[52] In this process, plant-specific RNA polymerases IV and V produce precursor transcripts and scaffold RNAs, respectively, which facilitate siRNA loading onto Argonaute proteins and recruitment of methyltransferases to homologous DNA loci, primarily establishing CHH methylation.[53] RdDM operates iteratively to reinforce silencing, particularly at TE edges near genes, preventing deleterious insertions during stress or reproduction.[53]
Maintenance of methylation involves specialized enzymes: MET1, a CG-specific methyltransferase homologous to mammalian DNMT1, perpetuates symmetric CG methylation across cell divisions by recognizing hemimethylated DNA post-replication.[54] CMT3, a chromomethylase, maintains CHG methylation in a strand-symmetric manner, often coupled with histone H3 lysine 9 dimethylation (H3K9me2) at pericentromeric heterochromatin.[55] For asymmetric CHH contexts, DRM2 catalyzes de novo and maintenance methylation, relying on RdDM for targeting and exhibiting activity across all contexts but predominantly in CHH.[55][54] These enzymes collectively ensure context-specific fidelity, with disruptions leading to TE reactivation and genome instability.[54]
Beyond TE control, DNA methylation contributes to adaptive functions such as epiallele inheritance, where stable methylation variants at loci influence hybrid vigor (heterosis) by modulating gene expression differences between parental alleles.[56] For instance, in Arabidopsis hybrids, parental methylation states at epialleles correlate with enhanced growth and yield, transmitted meiotically without sequence changes.[57] In stress responses, drought induces targeted demethylation at promoter regions of tolerance genes, such as those involved in abscisic acid signaling, enabling rapid transcriptional activation and phenotypic plasticity in crops like rice and tomato.[58][59] This dynamic remodeling underscores methylation's role in environmental adaptation, with evolutionary pressures from TE expansion driving higher baseline levels compared to animals.[51]
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 genome by adding methyl groups to the same sequences. For instance, the Type II RM system EcoRI, derived from Escherichia coli, 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 EcoRI, are the most common and act independently without requiring additional host factors for restriction.[60]
Beyond defense, RM systems can influence bacterial gene expression and phase variation, where reversible switches in phenotype occur due to changes in methylation patterns. In some bacteria, site-specific methylation by RM methyltransferases regulates the expression of virulence genes; for example, in Yersinia enterocolitica, DNA adenine methyltransferase (Dam) methylation modulates the expression of the inv gene encoding invasin, a key adhesin that promotes bacterial invasion of host epithelial cells, with altered methylation levels affecting invasion efficiency.[61] Phase variation mediated by RM systems often involves stochastic expression of the methyltransferase, leading to heterogeneous methylation states across a population that enable adaptive responses to environmental pressures, such as host immune evasion.[62]
A prominent example of solitary methylation in bacteria is the Dam methylase in E. coli, which N6-methylates adenine in GATC sequences, occurring approximately every 256 base pairs in the genome. This methylation 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.[63] Dam methylation also regulates DNA replication timing by controlling the sequestration of the origin of replication (oriC), where fully methylated oriC allows initiation, while hemimethylation leads to binding of SeqA protein, delaying re-replication to ensure once-per-cell-cycle firing.[64]
Orphan methyltransferases, which lack paired restriction enzymes, perform regulatory functions independent of defense. These enzymes methylate specific motifs to influence processes like gene expression, DNA repair, and cell cycle progression; for instance, in various Gram-positive and Gram-negative bacteria, orphan methylases such as those targeting non-palindromic sequences modulate virulence factor production and adaptation to stress without endonuclease activity.[65] In pathogens, orphan methylation can create epigenetic mosaics that drive population-level heterogeneity, enhancing survival in diverse niches like the host gut.[66]
In Other Eukaryotes
In fungi such as Neurospora crassa, DNA methylation primarily mediates gene silencing through the DNA methyltransferase DIM-2, which is responsible for all known cytosine methylation in vegetative tissues. This process often follows repeat-induced point mutation (RIP), a pre-meiotic mechanism that introduces GC-to-AT transitions in duplicated sequences, leading to subsequent methylation and epigenetic silencing of repetitive elements.[67] DIM-2 interacts with heterochromatin protein 1 (HP1) to establish and maintain these silenced states, contributing to genomic stability.[68]
In insects, DNA methylation levels vary but are generally low compared to vertebrates. For instance, Drosophila melanogaster exhibits minimal genome-wide methylation (approximately 0.03–1% of cytosines), lacking a homolog of the maintenance methyltransferase DNMT1 and relying instead on the debated DNMT2 enzyme, which may primarily target RNA.[69] In the germline, piRNA pathways guide transposon silencing mainly through chromatin modifications rather than extensive DNA methylation, though low-level cytosine methylation occurs at specific loci.[70] In contrast, social insects like the honeybee (Apis mellifera) show higher methylation associated with caste differentiation, where differential methylation patterns in larvae influence gene expression, promoting worker or queen development through nutritional cues.[71]
Protists display highly variable DNA methylation patterns, often differing from the canonical 5-methylcytosine (5mC) seen in higher eukaryotes. In Tetrahymena thermophila, for example, N6-methyladenine (6mA) predominates at levels of 0.6–0.8% of adenines, particularly during conjugation and macronuclear development, where it influences nucleosome positioning and chromatin organization without detectable 5mC.[72] This adenine methylation supports programmed DNA elimination and genome restructuring essential for cellular differentiation.[73]
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 silencing repeats and adapting to developmental needs through diverse enzymatic and guiding mechanisms.[74]
Roles in Development and Physiology
Embryonic Development and Differentiation
During mammalian embryogenesis, DNA methylation undergoes profound reprogramming to reset epigenetic marks inherited from gametes and establish new patterns essential for development. Immediately following fertilization, the zygote experiences two waves of global demethylation: an active process mediated by TET3, which oxidizes 5-methylcytosine to 5-hydroxymethylcytosine primarily on the paternal genome, 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 genome for totipotency. By the blastocyst 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.[75]
In embryonic stem cells (ESCs) derived from the inner cell mass of the blastocyst, 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 differentiation, 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.[75]
As embryogenesis progresses to gastrulation and germ layer formation, DNA methylation dynamics shift to lock in cell identities during differentiation. 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 neurogenesis. This reciprocal regulation ensures stable cell fate transitions, with hypermethylation reinforcing barriers against alternative lineages and hypomethylation permitting tissue-specific gene expression.
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 methylation 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.[76] Such epimutations highlight the precision required in embryonic methylation waves to maintain genomic imprinting 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.[77][78] These changes contribute to epigenetic drift, a progressive divergence from youthful methylation profiles that correlates with chronological age across tissues.[79] A 2025 meta-analysis of over 15,000 human methylomes from 17 tissues revealed both conserved age-related methylation shifts and pronounced tissue-specific gradients, with the brain, liver, and lungs showing the most substantial changes, highlighting organ-level variations in epigenetic aging trajectories.[80]
Epigenetic clocks leverage these age-associated methylation patterns to estimate biological age from DNA methylation levels at specific CpG sites. The seminal Horvath clock, developed in 2013, uses 353 CpG sites to predict chronological age across diverse human tissues and cell types with high accuracy, serving as a pan-tissue biomarker of cumulative epigenetic effects.[81] Second-generation clocks, such as DNA methylation PhenoAge (DNAm PhenoAge), extend this by incorporating phenotypic measures like blood biomarkers to estimate phenotypic age, which better reflects healthspan and mortality risk beyond chronological age alone.[82]
These methylation alterations arise from mechanistic processes exacerbated by aging, including oxidative stress that promotes the oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) via TET enzymes, leading to passive demethylation as 5hmC is poorly recognized by maintenance methyltransferase DNMT1.[83] Additionally, replication errors during cell division contribute to the accumulation of hypomethylation, as imperfect fidelity in DNMT1-mediated maintenance methylation allows stochastic losses at repetitive regions over successive divisions.[84]
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.[85] Recent studies on senolytics, such as dasatinib plus quercetin or BET inhibitors like JQ1, demonstrate reversal of methylation age in vitro by clearing senescent cells that propagate aberrant methylation patterns, with senolytics like dasatinib plus quercetin showing effects in mouse models.[86][87]
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 physical activity, nutritional status, and stressors. These epigenetic modifications allow for rapid, reversible changes in gene expression without altering the underlying DNA sequence, facilitating phenotypic plasticity in response to acute or chronic exposures.[88]
In response to exercise, particularly aerobic training, skeletal muscle exhibits site-specific DNA hypomethylation at promoters of metabolic genes, enhancing their expression to support energy demands and mitochondrial biogenesis. For instance, endurance exercise induces hypomethylation of the PPARGC1A (PGC-1α) gene promoter in human skeletal muscle, 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 training onset, and contributes to the muscle's metabolic reprogramming. Similar patterns occur at other loci, such as those regulating glucose uptake and fatty acid oxidation, underscoring exercise's role in epigenetic tuning for physical performance.[89][90]
Dietary factors and environmental toxins profoundly influence the DNA methylation landscape by altering the availability of methyl donors. Folate deficiency impairs the synthesis of S-adenosylmethionine (SAM), the primary methyl group donor for DNA methyltransferases, leading to global and gene-specific hypomethylation that disrupts genomic stability. This reduction in SAM pools, often seen in malnutrition or malabsorption conditions, results in decreased methylation at CpG islands, potentially activating retrotransposons and altering gene regulation. Conversely, exposure to cigarette smoke promotes hypermethylation of tumor suppressor gene promoters, such as p16INK4A and MGMT, through upregulation of DNA methyltransferase 1 (DNMT1) and accumulation of tobacco-specific carcinogens like NNK, 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.[91][92][93]
Chronic stress triggers glucocorticoid-mediated alterations in DNA methylation, particularly within the hypothalamic-pituitary-adrenal (HPA) axis, to fine-tune the stress response. Exposure to elevated glucocorticoids, such as cortisol, induces hypermethylation at the NR3C1 (glucocorticoid receptor) promoter, reducing receptor expression and impairing negative feedback regulation of the HPA axis, which can perpetuate heightened stress reactivity. These methylation shifts occur in brain regions like the hippocampus and are reversible upon stress cessation, allowing for adaptive recalibration of neuroendocrine signaling. Glucocorticoids also promote dynamic demethylation at stress-responsive loci, facilitating rapid gene activation in immune and neuronal cells.[94][95]
Recent studies from 2024 and 2025 have highlighted how air pollution exposure alters brain DNA methylation patterns, contributing to neuroinflammation and increased risk of neurological disorders such as Alzheimer's disease. Fine particulate matter (PM2.5) induces differential methylation in hippocampal and cortical regions, affecting genes involved in synaptic plasticity and inflammation, as observed in rodent models and analyses of human biomarkers. For example, PM2.5 exposure has been associated with altered methylation at CpG sites linked to Alzheimer's neuropathology. These findings underscore the role of epigenetic changes in mediating the neurotoxic effects of chronic pollutant exposure.[96][97]
Implications in Disease
In Cancer
Aberrant DNA methylation patterns are a hallmark of cancer, characterized primarily by global hypomethylation of the genome and focal hypermethylation of gene 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.[98] For instance, acute hypomethylation in mouse models results in a more than six-fold increase in adenoma formation due to enhanced loss of heterozygosity at tumor suppressor loci.[98] In contrast, promoter hypermethylation silences tumor suppressor genes, contributing to oncogenesis; a prominent example is the hypermethylation of the O6-methylguanine-DNA methyltransferase (MGMT) promoter in gliomas, which impairs DNA repair and sensitizes tumors to alkylating agents like temozolomide, associating with improved patient survival in newly diagnosed glioblastoma.[99]
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 malignant transformation. These field cancerization effects, detected via genome-wide methylation profiling, highlight early epigenetic reprogramming in tissues like the esophagus and colon, correlating with increased cancer risk before histological changes occur.[100] In breast cancer, hypermethylation of the BRCA1 promoter exemplifies this by functionally mimicking germline mutations, leading to BRCAness phenotype with homologous recombination deficiency and heightened sensitivity to PARP inhibitors, independent of genetic alterations.[101]
Recent advances in 2025 have leveraged DNA methylation profiling for precise tumor classification, particularly in central nervous system (CNS) tumors. The Heidelberg 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.[102]
Therapeutic targeting of DNA methylation has advanced with DNA methyltransferase (DNMT) inhibitors, such as azacitidine, approved for myelodysplastic syndromes (MDS) and acute myeloid leukemia (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.[103] Combinations with immunotherapy enhance efficacy; low-dose azacitidine upregulates tumor-associated antigens and immune checkpoint expression, improving responses to PD-1 inhibitors in solid tumors and hematological malignancies by reprogramming the tumor microenvironment.[104]
In Neurological and Cardiovascular Disorders
DNA methylation dysregulation plays a significant role in various neurological disorders, particularly through alterations in gene expression that affect neuronal function and plasticity. In depression, hypermethylation of the brain-derived neurotrophic factor (BDNF) promoter has been consistently observed, leading to reduced BDNF expression and impaired neurotrophic support in affected individuals.[105] This epigenetic modification is associated with the pathophysiology of major depressive disorder (MDD), where peripheral blood mononuclear cells from MDD patients show significantly higher BDNF promoter methylation compared to healthy controls.[106] Similarly, recent investigations into substance use disorders highlight cocaine's impact on methylation patterns in reward pathways. A 2025 study revealed that cocaine exposure induces dynamic transcriptomic changes in dopamine D2 receptor (Drd2)-expressing medium spiny neurons within the nucleus accumbens, a key reward circuit region, potentially involving methylation-mediated regulation of gene expression during addiction development and withdrawal.[107] These findings build on earlier evidence that DNA methylation in the nucleus accumbens modulates incubation of cocaine craving, underscoring its role in long-term addictive behaviors.[108]
In neurodevelopmental disorders, mutations in the methyl-CpG-binding protein 2 (MECP2) gene exemplify how disruptions in methylation reading impair brain function. Rett syndrome, a severe neurodevelopmental condition, arises primarily from loss-of-function MECP2 mutations that compromise MeCP2's ability to bind methylated CpG sites and repress transcription, resulting in widespread gene dysregulation and neurological symptoms such as seizures and motor impairments.[109] These mutations, found in up to 80% of classic cases, alter MeCP2's interaction with methylated DNA, leading to aberrant expression of target genes critical for neuronal maturation.[110] Addiction mechanisms further intersect with these pathways, as opioid exposure induces aberrant DNA methylation at the mu-opioid receptor gene (OPRM1) promoter. Chronic opioid use correlates with increased OPRM1 promoter methylation in lymphocytes of addicts and medication users, potentially silencing receptor expression and contributing to tolerance and dependence.[111] This hypermethylation pattern is replicated in preclinical models, where it associates with heightened neonatal abstinence syndrome severity in opioid-exposed infants.[112]
Shifting to cardiovascular disorders, aberrant DNA methylation contributes to vascular pathology by modulating genes involved in inflammation and endothelial function. In atherosclerosis, hypermethylation of the estrogen receptor alpha (ESR1) promoter is linked to disease progression, with elevated homocysteine levels positively correlating with increased ESR1 methylation and lesion severity in arterial tissues.[113] This epigenetic silencing reduces ESR1 expression, impairing estrogen-mediated protection against plaque formation and promoting inflammatory responses in vascular smooth muscle cells.[114] Peripheral blood analyses confirm differential ESR1 promoter methylation in females with atherosclerosis, highlighting its potential as a biomarker for cardiovascular risk. In heart failure, 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.[115] This mechanism suggests that targeting methylation could mitigate TNF-α-driven remodeling in heart failure.
In Other Pathologies
DNA methylation alterations play a significant role in autoimmune diseases, particularly systemic lupus erythematosus (SLE), where hypomethylation of the interferon gamma gene (IFNG) in T cells contributes to dysregulated immune responses and hypersensitivity to interferon signaling.[116] This hypomethylation leads to overexpression of IFNG and other interferon-responsive genes, correlating with higher disease activity and autoantibody production in SLE patients.[117] Additionally, genetic variants in the DNA methyltransferase 3B (DNMT3B) gene, such as the rs2424913 polymorphism, are associated with increased SLE susceptibility by impairing de novo DNA methylation and exacerbating global hypomethylation in immune cells.[118]
In metabolic disorders, DNA methylation changes affect insulin production and adipose tissue function. Hypermethylation of the insulin (INS) gene promoter in pancreatic islets of individuals with type 2 diabetes (T2D) inversely correlates with gene expression, contributing to impaired beta-cell function and reduced insulin secretion.[119] In obesity, differential methylation at adipocyte enhancer regions alters the expression of genes involved in lipid metabolism and inflammation; for instance, loss of DNMT3A-mediated methylation at distal enhancers disrupts adipocyte differentiation and promotes morbid obesity phenotypes.[120][121]
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.[122] Host cells employ DNA methylation as a defense strategy by hypermethylating invading pathogen genomes, such as viral DNA, to silence foreign gene expression and limit replication; this is evident in the epigenetic repression of integrated viral elements by host DNA methyltransferases.[123]
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.[124] These methylation signatures, particularly at genes like ADM and RIN1, enable multi-omics integration for improved clinical management beyond traditional histopathology.[125]
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 sodium bisulfite, which deaminates unmethylated cytosines to uracils (read as thymines during sequencing), while 5-methylcytosine (5mC) remains unchanged, allowing for precise mapping via polymerase chain reaction (PCR) amplification and next-generation sequencing (NGS). This approach enables whole-genome bisulfite sequencing (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.[126][127]
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.[128]
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 bisulfite conversion, and amplifying via whole-genome amplification 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 multiplexing protocols and improved library preparation, enhance throughput to thousands of cells while reducing noise, enabling detailed studies of methylation dynamics in heterogeneous populations.[129][130]
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 machine learning 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, chromatin accessibility, and evolutionary conservation to predict methylation probabilities at specific sites. For instance, deep learning models like DeepMethyl employ stacked denoising autoencoders to classify CpG dinucleotide methylation states, achieving high accuracy by incorporating topological genome features alongside sequence motifs.[131] Such predictors are particularly useful for imputing missing methylation data in sparse datasets or simulating epigenetic landscapes in silico.[132]
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 linear regression models adjusted for confounders such as cell-type composition.[133] In single-cell contexts, recent AI 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.[134] These pipelines often integrate tools for quality control, normalization, and visualization, supporting downstream applications in developmental biology and pathology.
Epigenetic clocks, such as the Horvath clock, computationally estimate biological age by regressing chronological age against DNA methylation beta values at a panel of 353 CpG sites using elastic net regression, providing a robust metric for aging research across tissues.[81] This algorithm highlights hyper- and hypomethylated sites that correlate with age progression, with deviations (epigenetic age acceleration) linked to health 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 bisulfite sequencing data compared to non-shrunk alternatives.[135] These methods prioritize regions with biological relevance, such as promoter-associated CpGs, while controlling for multiple testing via false discovery rates.[136]
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.[137]
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.[138][137][139]
For aging and health assessment, epigenetic clocks derived from DNA methylation serve as biomarkers for biological age, aiding in risk 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 risk; 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, cardiovascular disease, and overall survival, integrating lifestyle and immunological factors for personalized health interventions. Emerging EpiScores extend this to forecast exposure to environmental risks and disease susceptibility, such as predicting liver or respiratory conditions with improved accuracy over first-generation clocks.[140][141][142]
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.[143][144][145]
Therapeutic monitoring via DNA methylation tracks responses to DNA methyltransferase inhibitors (DNMTi) like decitabine, which reverse aberrant hypermethylation in cancers. In breast cancer, methylation levels at promoter sites (e.g., in BRCA1 or MGMT) 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 drug resistance and optimizing combination therapies with immune checkpoint inhibitors. These approaches, validated in 2024-2025 clinical cohorts, enhance monitoring by detecting minimal residual disease through cfDNA changes as early as one cycle into treatment.