Fact-checked by Grok 2 weeks ago

Epigenetics

Epigenetics is the study of heritable changes in gene expression that occur without alterations to the underlying DNA sequence, allowing cells to control which genes are active or silenced in response to developmental cues, environmental influences, and other factors. These modifications, collectively known as the epigenome, regulate gene activity by adding or removing chemical tags to DNA or associated proteins, ensuring that different cell types—such as neurons and liver cells—express distinct sets of genes from the same genome despite identical DNA. Coined in the 1940s by Conrad Waddington to describe the interaction between genes and their products during development, the term has evolved to encompass mechanisms that are mitotically heritable during cell division and potentially meiotically heritable across generations. The primary mechanisms of epigenetics include DNA methylation and histone modifications, which together form a dynamic layer of regulation. DNA methylation involves the addition of methyl groups to bases, particularly at CpG dinucleotides, typically repressing gene transcription by inhibiting the binding of transcription factors or recruiting repressive proteins; this process is maintained by enzymes like during . Histone modifications, on the other hand, entail the , methylation, phosphorylation, or other chemical alterations to proteins around which DNA is wrapped into ; for instance, histone acetylation generally loosens chromatin structure to promote gene activation, while certain methylations can either activate or repress genes depending on the specific residue modified. Additional mechanisms, such as non-coding RNAs, can also influence epigenetic states by guiding chromatin-modifying complexes to target genes. Epigenetic changes play a crucial role in normal development, , and adaptation to environmental stimuli, such as , , or toxins, which can alter the epigenome and affect outcomes across an individual's lifetime. These modifications are reversible and tissue-specific, enabling precise control over —for example, ensuring that insulin-producing genes are active only in pancreatic beta cells. Dysregulation of epigenetic processes contributes to various diseases, including cancers, where aberrant silences tumor suppressor genes, and metabolic disorders influenced by environmental exposures during critical developmental windows. Research into epigenetics holds promise for therapeutic interventions, such as drugs targeting deacetylases, and underscores the interplay between , , and in shaping phenotypes.

Definitions and History

Core Definition

Epigenetics was coined by British developmental biologist in to describe the branch of that studies the causal interactions between genes and their products, which bring the into being. Waddington introduced the term to bridge the gap between and , emphasizing the dynamic developmental processes that mediate how genetic information is realized in an organism's traits. In contemporary usage, epigenetics refers to stable, heritable changes in that do not involve alterations to the underlying DNA sequence. These changes encompass mechanisms such as , histone modifications, and non-coding RNA-associated , which regulate structure and accessibility to influence transcriptional activity. Unlike genetic mutations, which permanently alter the DNA sequence and are typically irreversible, epigenetic modifications are potentially reversible and can respond to environmental cues, such as , , or toxins, allowing for adaptive plasticity in gene regulation. Prominent examples include X-chromosome inactivation in female mammals, where one X chromosome is transcriptionally silenced to achieve dosage compensation, and , where parental origin determines allele-specific expression through differential epigenetic marks.

Historical Foundations

The concept of , denoting the progressive unfolding of form during embryonic development from undifferentiated material, traces back to in the 4th century BCE, who described it as a teleological process where potentialities are realized sequentially rather than pre-existing fully formed. This view contrasted sharply with , a dominant theory from the 17th to 18th centuries, which posited that organisms develop from miniature, preformed versions of themselves encapsulated within gametes, as advocated by figures like and based on early microscopic observations of spermatozoa and eggs. The preformationist doctrine, often illustrated by models suggesting in nested embryos, aimed to resolve debates on generation but faced challenges from observations of developmental anomalies, gradually yielding to revived epigenetic ideas by the late 18th century through naturalists like Caspar Friedrich Wolff and . In the 19th and early 20th centuries, the rediscovery of Gregor Mendel's 1865 laws of inheritance in 1900 by Hugo de Vries, Carl Correns, and Erich von Tschermak established genetics as a field focused on discrete heritable units, yet it highlighted gaps in explaining continuous phenotypic variability and environmental influences on traits beyond strict genotypic determinism. The formulation of the central dogma of molecular biology by Francis Crick in 1958, stating that genetic information flows unidirectionally from DNA to RNA to proteins, further emphasized genes as the primary drivers of heredity, but it underscored unresolved questions about how identical genotypes could yield diverse phenotypes in development and adaptation. These limitations prompted embryologists to seek mechanisms bridging genetics and environmental interactions, setting the stage for epigenetics as a complementary framework. Conrad Hal Waddington, a developmental , formalized the term "epigenetics" in 1942 to describe the causal mechanisms operating between and during development, coining it as "the interaction of with their products in the production of ." In the , he introduced the concept of canalization, referring to the developmental buffering that stabilizes phenotypes against genetic or environmental perturbations, allowing robust pathways to form despite variability, as demonstrated in his experiments with where heat shock induced phenotypic changes that became genetically assimilated over generations. Waddington's iconic epigenetic landscape model, depicted in 1957 as a hilly terrain with branching valleys representing developmental trajectories constrained by genetic and epigenetic factors, illustrated how cells "roll" toward stable fates influenced by both internal networks and external cues, emphasizing the dynamic interplay over rigid genetic predetermination. The 1953 discovery of DNA's double-helix structure by and shifted biological focus toward , temporarily sidelining epigenetic inquiries, yet by the 1960s and 1970s, interest resurged in molecular bases for cellular memory and . A pivotal advancement came in 1975 when Robin Holliday and John Pugh, independently proposed by Arthur D. Riggs who linked it to X-chromosome inactivation, proposed that site-specific DNA modifications, particularly , serve as heritable signals maintaining gene expression states across cell divisions without altering the DNA sequence, providing a mechanism for stable in mammals. During the 1980s and 1990s, research solidified DNA methylation's role in mammals, with studies identifying its prevalence at CpG dinucleotides in genomes and its association with , as mapped in detail by Adrian Bird's group showing tissue-specific patterns that regulate imprinting and X-chromosome inactivation. Concurrently, the functional significance of acetylation emerged, building on Vincent Allfrey's 1964 detection of dynamic modifications but gaining epigenetic prominence in the 1990s through discoveries of acetyltransferases (HATs) like those identified by and David Allis, which were shown to promote open and transcriptional activation, contrasting with deacetylation's repressive effects. These findings established covalent modifications as key epigenetic regulators, integrating them into models of -mediated control.

Epigenetic Mechanisms

DNA Methylation

is a key epigenetic mechanism involving the covalent addition of a to the fifth carbon of bases in DNA, primarily at CpG dinucleotides, which represses by inhibiting binding and promoting compaction. This modification, known as (5mC), accounts for approximately 1% of total DNA bases in cells. In the , there are about 28 million CpG sites, the majority of which are methylated in a tissue-specific manner to maintain cellular . The biochemical process is catalyzed by DNA methyltransferases (DNMTs), a family of enzymes that transfer a methyl group from S-adenosylmethionine to cytosine. DNMT1 primarily functions in maintenance methylation, recognizing hemimethylated DNA during replication to copy the parental strand's methylation pattern to the daughter strand, ensuring heritability across cell divisions. In contrast, DNMT3A and DNMT3B mediate de novo methylation, establishing new patterns on previously unmethylated DNA during development and differentiation. DNA methylation patterns exhibit dynamic alterations in disease states, such as cancer, where global hypomethylation of repetitive elements and gene bodies leads to genomic instability and activation, while hypermethylation of promoter CpG islands silences tumor suppressor genes like TP53 and BRCA1. These opposing changes contribute to tumorigenesis by disrupting normal gene regulation. Demethylation occurs through two main pathways: passive dilution, which happens during when maintenance methylation by is impaired, leading to progressive loss of 5mC over cell divisions; and active demethylation, initiated by ten-eleven translocation () enzymes that oxidize 5mC to (5hmC) and further to 5-formylcytosine and 5-carboxylcytosine, which are then excised by . TET-mediated oxidation is crucial for erasing marks in processes like embryonic . This mechanism is evolutionarily conserved across prokaryotes and eukaryotes, where it serves roles in gene regulation and defense against foreign DNA, though the specific targets differ—cytosine in eukaryotes versus adenine or cytosine in bacteria. DNA methylation often interacts with histone modifications to reinforce gene silencing, such as through recruitment of proteins that deposit repressive marks like H3K9me3.

Histone Modifications

Histone modifications are post-translational alterations to the amino acid residues of histone proteins, primarily occurring on the N-terminal tails of the core histones H2A, H2B, H3, and H4, which together form the octameric nucleosome core around which DNA is wrapped to create chromatin. These modifications, including acetylation, methylation, phosphorylation, and ubiquitination, are catalyzed by specific enzymes such as histone acetyltransferases (HATs) and deacetylases (HDACs) for acetylation, and histone methyltransferases (HMTs) for methylation, thereby influencing chromatin structure and accessibility. For instance, acetylation neutralizes the positive charge on lysine residues, reducing the affinity between histones and negatively charged DNA to promote an open chromatin conformation, while methylation can either activate or repress gene expression depending on the specific residue and degree of methylation. The code hypothesis posits that combinations of these modifications on tails serve as a "code" that is recognized by effector proteins, which in turn recruit -remodeling complexes or transcriptional machinery to regulate . This hypothesis, proposed by Jenuwein and Allis, suggests that distinct modification patterns provide binding sites for reader domains, such as bromodomains that preferentially bind acetylated lysines to facilitate transcriptional . Over 100 distinct types of modifications have been identified, each contributing to the nuanced control of dynamics and genome function. Certain modifications act as activating marks, such as trimethylation of at 4 (), which is enriched at active gene promoters and correlates with ongoing transcription by recruiting factors like TFIID. In contrast, repressive marks include trimethylation of H3 at 27 (), mediated by the Polycomb repressive complex 2 (PRC2), which compacts to silence developmental genes, and trimethylation at 9 (H3K9me), associated with formation and long-term gene repression. These marks often work in opposition; for example, and can coexist at bivalent promoters in embryonic stem cells to poise genes for activation or repression during . Histone modifications are highly dynamic, with enzymes like , the catalytic subunit of PRC2, depositing in a context-specific manner during embryonic to maintain cellular and prevent premature . This reversibility allows rapid responses to cellular signals, ensuring precise spatiotemporal control of . In some cases, histone modifications compound with to enhance transcriptional silencing, though their primary roles remain distinct in regulation.

Non-Coding RNAs

Non-coding RNAs (ncRNAs) constitute the vast majority of the eukaryotic transcriptome, accounting for approximately 98% of transcriptional output in humans, with the remainder encoding . These RNA molecules, which lack protein-coding capacity, exert profound influence on epigenetic by guiding chromatin-modifying complexes to specific genomic loci, thereby silencing or activating in a heritable manner without changing the underlying DNA sequence. In eukaryotes, ncRNAs have evolved as versatile regulators, conserved across species from to mammals, enabling precise control over developmental processes and genomic stability. The primary classes of ncRNAs involved in epigenetic mechanisms include microRNAs (miRNAs), small interfering RNAs (siRNAs), PIWI-interacting RNAs (piRNAs), and long non-coding RNAs (lncRNAs). miRNAs are small RNAs approximately 22 nucleotides in length that typically function post-transcriptionally by binding to target mRNAs, leading to their degradation or translational repression; however, they also contribute to epigenetic silencing by targeting the 3' untranslated regions of mRNAs encoding key epigenetic enzymes, such as DNA methyltransferases. For instance, the miR-29 family directly targets DNMT3A and DNMT3B, reducing their expression and thereby decreasing at aberrant sites. siRNAs, typically 20-25 nucleotides long, mediate (RNAi) pathways that trigger epigenetic modifications, particularly formation and , in organisms like fission yeast and plants. In these systems, siRNAs are generated from double-stranded precursors and recruit proteins to homologous genomic sequences, promoting lysine 9 (H3K9me) and subsequent . This mechanism exemplifies how siRNAs bridge RNA-based recognition with chromatin-level repression to maintain pericentromeric . piRNAs, ranging from 24 to 31 nucleotides, are specialized for protection, where they silence transposable elements (transposons) to preserve integrity during . Produced in a ping-pong amplification cycle involving clade proteins, piRNAs guide these effectors to transposon transcripts, inducing DNA methylation and modifications that prevent transposon mobilization. This silencing is essential in species from flies to mammals, ensuring and transgenerational epigenetic stability. lncRNAs, defined as transcripts longer than 200 nucleotides, represent the most diverse class, often acting as scaffolds or guides for chromatin-modifying complexes to regulate large-scale epigenetic domains. In humans, the genome encodes around 20,000 lncRNA genes, many of which are primate-specific yet functionally conserved in their regulatory roles across eukaryotes. A paradigmatic example is Xist, a lncRNA expressed exclusively from the inactive X chromosome in female mammals, which coats the entire X chromosome in cis, recruiting Polycomb repressive complex 2 (PRC2) and other factors to establish silencing through H3K27 trimethylation (H3K27me3) and DNA methylation. Another well-studied lncRNA, HOTAIR, originates from the HOXC locus and interacts with PRC2 to propagate H3K27me3 marks across distant genomic regions, such as the HOXD cluster, thereby coordinating Hox gene repression during development. lncRNAs like these frequently integrate with histone modification pathways, enhancing the specificity of epigenetic control.

Other Mechanisms

Prions represent a class of epigenetic mechanisms involving heritable changes in protein conformation without alterations to the underlying DNA or RNA sequences. In yeast, the Sup35 protein can adopt a self-propagating amyloid form known as [PSI+], which induces phenotypic switches such as reduced nonsense suppression, allowing for rapid adaptation to environmental stresses through non-genetic inheritance. This prion state is maintained cytoplasmically and transmitted to daughter cells, exemplifying protein-based epigenetic memory that persists across generations. Prion-like domains in eukaryotic proteins, characterized by low-complexity sequences prone to aggregation, extend this mechanism to higher organisms; for instance, the RNA-binding protein FUS contains such a domain that facilitates phase-separated condensates but can drive pathological aggregation in neurodegenerative diseases like amyotrophic lateral sclerosis (ALS). Chromatin remodeling complexes provide another layer of epigenetic regulation by dynamically repositioning in an ATP-dependent manner, thereby influencing accessibility without covalent modifications. The family, conserved from to humans, acts as a multi-subunit motor that slides, ejects, or restructures nucleosomes to facilitate binding and enhancer activation during and . Mutations in SWI/SNF components are prevalent in cancers and neurodevelopmental disorders, underscoring their role in maintaining epigenetic landscapes for proper cellular identity. These complexes often cooperate with variants to fine-tune nucleosome stability, bridging structural changes with functional outcomes in regulation. RNA methylation, particularly N6-methyladenosine (m6A) on , emerges as an epitranscriptomic mechanism that epigenetically modulates post-transcriptional processes. The m6A modification, installed by the METTL3-METTL14 writer complex, primarily affects mRNA and efficiency by recruiting reader proteins like YTHDF2, which promotes , or YTHDF1, which enhances cap-independent . Dysregulated m6A levels have been linked to diseases including cancer and neurological disorders, where it influences RNA localization and splicing as well. This dynamic mark integrates with other epigenetic signals to control plasticity. Nuclear architecture contributes to epigenetic inheritance through , where organizes into distinct liquid-like compartments that enforce spatial segregation of active and repressive domains. , marked by open, transcriptionally permissive structures, tends to localize centrally in the , while forms dense, peripheral condensates stabilized by interactions with lamina-associated proteins and HP1. Liquid-liquid driven by intrinsically disordered regions in proteins enables rapid reconfiguration of these compartments in response to signals, preserving epigenetic states across divisions. This organization not only compartmentalizes the but also influences long-range interactions essential for stable .

Molecular and Cellular Basis

Nucleosome Positioning and Histone Variants

Nucleosomes serve as the fundamental units of chromatin, consisting of approximately 147 base pairs of DNA wrapped around a histone octamer composed of two copies each of the core histones H2A, H2B, H3, and H4. This wrapping occurs in about 1.65 left-handed superhelical turns, compacting the DNA and influencing its accessibility to regulatory factors. Nucleosome positioning, which refers to the precise placement of these particles along the DNA, is not random but modulated by intrinsic DNA sequence features, such as poly(dA:dT) tracts that act as barriers to nucleosome occupancy due to their rigidity and propensity for straight conformations. These sequences promote nucleosome exclusion or repositioning, thereby creating regions of higher chromatin accessibility and facilitating processes like transcription initiation. Histone variants further diversify nucleosome function by replacing canonical histones in specific genomic contexts, altering stability and regulatory potential. For instance, the variant H2A.Z is enriched at gene promoters, where it destabilizes to enhance accessibility and promote transcriptional activation, often in combination with other marks. In contrast, the centromeric variant CENP-A substitutes for in at centromeres, forming specialized domains essential for assembly and accurate segregation during . These variants contribute to epigenetic memory by maintaining distinct states that guide cellular processes without altering the underlying DNA sequence. To prevent the inappropriate propagation of epigenetic states, domains are delineated by elements, which function as barriers and boundaries. These DNA sequences, such as those bound by proteins like in vertebrates or Su(Hw) in , block the spreading of repressive or active modifications between adjacent genomic regions, thereby preserving domain-specific patterns. Insulators achieve this by facilitating higher-order looping or directly impeding the diffusion of chromatin-modifying complexes. Nucleosome positioning is inherited through semi-conservative , where parental are randomly distributed to daughter strands and complemented by newly synthesized deposited via dedicated chaperones. The assembly factor 1 (CAF-1) plays a central role in this process, preferentially depositing H3-H4 tetramers onto newly replicated DNA in a replication-coupled manner, thereby reestablishing occupancy and positioning to maintain epigenetic information across cell divisions. This mechanism ensures the propagation of positioned , particularly at regulatory elements. A subset of in active genes exhibit precise positioning, underscoring the dynamic of in transcribed regions while highlighting the importance of sequence-directed barriers for functional organization.

DNA Damage, Repair, and Epigenetic Changes

DNA damage and repair processes are intricately linked to epigenetic modifications, as the resolution of lesions often requires that can propagate or alter heritable marks such as and modifications. Double-strand breaks (DSBs) and oxidative lesions trigger signaling cascades that recruit epigenetic regulators, ensuring repair fidelity while potentially inducing long-term changes in patterns. These intersections highlight how repair pathways not only restore genomic but also influence epigenetic landscapes, with implications for cellular memory and . Oxidative damage, particularly the formation of 8-oxoguanine (8-oxoG), is primarily repaired through (BER), initiated by 8-oxoguanine DNA glycosylase 1 (OGG1). This process can alter nearby by inhibiting the methylation of adjacent cytosines in CpG dinucleotides and facilitating the recruitment of enzymes, which oxidize 5-methylcytosine to , promoting active demethylation via subsequent BER steps. Such changes convert signals into epigenetic responses, enabling gene activation at promoter regions without permanent sequence alterations. For instance, 8-oxoG accumulation in gene promoters acts as a transient epigenetic mark that coordinates repair with transcriptional reprogramming. In (), a high-fidelity DSB repair pathway, eviction and reassembly are critical for accessing damaged DNA and restoring structure. During , parental marked with —an active transcription-associated modification—are disassembled at break sites and preferentially reassembled onto repaired DNA, thereby propagating these epigenetic marks across generations of nucleosomes. This mechanism ensures the inheritance of active states post-repair, influencing local . also modifies the pattern of the repaired segment, integrating sequence restoration with epigenetic fidelity. Non-homologous end joining (NHEJ), the predominant DSB repair pathway in non-dividing cells, often results in small insertions or deletions at repair junctions, which can lead to of at these sites. The error-prone nature of NHEJ disrupts CpG continuity during end ligation, potentially erasing local epigenetic silencing and contributing to genomic instability or altered gene regulation. This at junctions provides a mechanism for epigenetic heterogeneity arising from imperfect repair. DSBs activate and signaling, which recruit epigenetic modifiers to facilitate repair. poly(ADP-ribosyl)ates histones and factors at break sites, enabling the swift recruitment of histone demethylases like KDM4D to remove repressive marks and promote relaxation for NHEJ or access. Similarly, kinase phosphorylates and recruits epigenetic readers such as BRD7, which in turn assembles polycomb repressive complexes to condense and suppress transcription near breaks, ensuring repair prioritization. These pathways thus couple damage sensing with dynamic epigenetic reprogramming. During , repair plays a pivotal role in altering genomic imprints, where double-strand breaks facilitate crossover formation that can disrupt or reconfigure patterns at imprinted loci. This process contributes to transgenerational epigenetic effects by erasing parental imprints in primordial germ cells and establishing new ones, with recombination hotspots in imprinted regions amplifying the potential for heritable changes. Such alterations ensure proper differentiation while allowing environmental influences to propagate epigenetically across generations.

Techniques for Studying Epigenetics

Chromatin immunoprecipitation (ChIP) is a foundational technique for studying protein-DNA interactions in epigenetic regulation, involving the use of antibodies to selectively pull down chromatin fragments containing specific modified histones or transcription factors bound to DNA, followed by analysis via quantitative PCR (qPCR) or high-throughput sequencing (ChIP-seq) to map binding sites genome-wide. Developed in the late 1980s and refined for sequencing applications in the 2000s, ChIP-seq enables precise identification of histone modification patterns, such as H3K27ac for active enhancers, across cell types. CUT&RUN (Cleavage Under Targets and Release Using Nuclease) is an advanced antibody-based method that uses targeted micrococcal nucase to cleave DNA near protein binding sites, offering higher resolution and reduced background compared to ChIP for mapping histone modifications and transcription factors. Introduced in 2017, it is particularly useful for low-input samples and has become a standard for precise epigenetic profiling. Bisulfite sequencing remains the gold standard for detecting DNA methylation at cytosine residues, treating genomic DNA with sodium bisulfite to convert unmethylated cytosines to uracils while leaving methylated cytosines unchanged, allowing subsequent PCR amplification and sequencing to map methylation at single-base resolution. Introduced in 1992, this method has been pivotal for quantifying 5-methylcytosine (5mC) levels in CpG islands and repetitive elements, though it requires careful optimization to minimize DNA degradation during bisulfite conversion. Methylation-sensitive restriction enzymes provide a targeted approach for differential analysis of DNA methylation by selectively digesting unmethylated DNA at recognition sites, such as HpaII (which cleaves CCGG only when unmethylated), enabling comparison of digested versus undigested fragments via PCR or Southern blotting to assess methylation status in specific loci. This enzymatic method, dating back to the 1970s but widely adopted in epigenetics by the 1990s, is cost-effective for validating methylation changes in promoter regions without full genome sequencing. Fluorescent in situ hybridization () visualizes epigenetic chromatin domains in three dimensions by hybridizing fluorescently labeled probes to specific DNA sequences in fixed cells, revealing spatial organization of modified structures like foci or topologically associating domains (TADs). Enhanced multiplexed variants, such as oligoFISH, allow simultaneous detection of multiple epigenetic marks alongside genomic loci, providing insights into nuclear architecture at the single-cell level. Nanopore sequencing offers a direct, label-free method to detect epigenetic base modifications, such as 5mC and 6mA, by measuring changes in ionic current as DNA translocates through a protein , bypassing the need for conversion and preserving long-range information. Since its application to epigenetics around , this third-generation technology has improved accuracy for native modification calling, particularly in complex genomes, with tools like Nanopolish enabling methylation detection at high accuracy (typically >85% in validated datasets). scATAC-seq (single-cell Assay for Transposase-Accessible using sequencing), developed in 2015, assays accessibility by transposase-mediated insertion of sequencing adapters into open regions, enabling epigenetic profiling of regulatory elements at individual cell resolution to uncover cell-type-specific accessibility landscapes. scATAC-seq has been integrated with multi-omics approaches (e.g., SHARE-seq in 2019) to dissect dynamic epigenetic states in heterogeneous tissues. Complementing this, long-read sequencing facilitates phasing of epigenetic marks by resolving allele-specific modifications over kilobase-scale distances, as demonstrated by tools like MethPhaser that leverage signals alongside variants. The project has extensively mapped reference epigenomes across hundreds of human types using these techniques, including ChIP-seq for histone marks, for , and ATAC-seq for accessibility, generating comprehensive datasets that reveal conserved epigenetic regulatory principles.

Biological Functions

Development and Cell Differentiation

Epigenetics plays a pivotal role in embryonic by orchestrating cell fate decisions and ensuring proper tissue specification through dynamic modifications that the from totipotency to differentiated states. During early embryogenesis, the undergoes extensive epigenetic reprogramming, including waves of that erase parental imprints and activate the zygotic . This process begins shortly after fertilization, with the paternal undergoing rapid active demethylation mediated by TET3 enzymes, followed by passive demethylation in the maternal during subsequent divisions, facilitating zygotic activation (ZGA) around the 8-cell stage in humans. These demethylation events are essential for resetting the epigenome and enabling the expression of developmental genes, preventing the inheritance of gamete-specific marks that could disrupt lineage progression. A key example of epigenetic reprogramming is seen in the generation of induced pluripotent stem cells (iPSCs), where somatic cells are reverted to a pluripotent state using the Yamanaka factors—Oct4, , , and c-Myc. These transcription factors drive the erasure of somatic epigenetic marks, including DNA hypermethylation and repressive histone modifications, to restore an embryonic-like epigenome capable of self-renewal and . This process mimics natural developmental reprogramming but highlights the plasticity of epigenetic states, as incomplete erasure can lead to residual somatic memory that biases potential. In pluripotent cells like embryonic stem cells, bivalent chromatin domains—characterized by the coexistence of activating and repressive histone marks—poise developmental genes for timely activation during lineage commitment. These domains silence lineage-specific genes while keeping them accessible for rapid expression upon differentiation signals, ensuring coordinated cell fate transitions. As development proceeds to , DNA methylation progressively locks in differentiated states; for instance, in myoblasts, hypermethylation of non-muscle gene promoters silences alternative lineages, stabilizing myogenic identity and preventing . This epigenetic locking is crucial for maintaining tissue-specific gene expression patterns established during early embryogenesis. Environmental factors can also imprint lasting epigenetic changes during development, influencing offspring outcomes across generations. The Dutch Hunger Winter famine of 1944–1945 demonstrated this, as periconceptional exposure led to persistent hypomethylation at the in adult survivors, correlating with altered growth regulation. Such influences underscore how extrinsic cues can modulate epigenetic landscapes at critical developmental windows, affecting cell differentiation and long-term physiology without altering the DNA sequence.

Gene Expression Regulation

Epigenetic mechanisms play a crucial role in fine-tuning in mature cells, enabling precise control over transcriptional activity to maintain cellular identity and respond to internal signals without altering the DNA sequence. These processes involve modifications such as histone acetylation and that alter accessibility, allowing transcription factors to bind or be repelled from regulatory regions. In steady-state conditions, these marks ensure stable yet dynamic gene regulation, distinguishing active from repressed states across the . Enhancers and silencers are key cis-regulatory elements modulated by epigenetic marks to orchestrate activity. Histone , particularly at residues like H3K27, promotes an open conformation that facilitates the formation of chromatin loops, bringing enhancers into proximity with promoters and enabling . For instance, by histone acetyltransferases (HATs) such as p300/CBP neutralizes positive charges on s, reducing DNA- and exposing binding sites for activators. Conversely, silencers often feature repressive marks like or , which recruit Polycomb repressive complexes to compact and inhibit access, thereby silencing target . Active enhancers marked by H3K27 cover approximately 1-2% of the mammalian but are estimated to regulate up to 80% of protein-coding through long-range interactions. Feedback loops involving non-coding RNAs further refine epigenetic . MicroRNAs (miRNAs), such as miR-29, can form double-negative feedback circuits by targeting epigenetic regulators like DNA methyltransferases (DNMTs), which in turn influence at the miRNA's own genomic locus, leading to self-silencing or amplification of repressive states. This autoregulatory mechanism helps stabilize expression levels in mature cells, preventing aberrant activation. Metabolic sensing integrates cellular energy status into epigenetic control, with serving as a central metabolite that directly fuels histone . Fluctuations in levels, derived from glucose, fatty acids, or , modulate HAT activity and thus openness at regulatory elements. For example, high availability during nutrient abundance promotes of histones at active loci, enhancing transcription of metabolic , while scarcity restricts it to prioritize essential functions. Epigenetic plasticity allows environmental cues to dynamically alter marks in mature cells. Dietary components, such as like butyrate produced by , act as natural HDAC inhibitors, increasing and activating genes involved in and . This responsiveness ensures to nutritional changes while preserving cellular .

Transgenerational Effects

Transgenerational epigenetic inheritance refers to the transmission of epigenetic modifications across multiple generations without alterations to the underlying DNA sequence, potentially influencing phenotypes in offspring. This phenomenon challenges traditional views of inheritance by suggesting that environmental exposures can leave lasting marks on the germline, affecting descendants. In mammals, such inheritance is constrained by extensive epigenetic reprogramming during gametogenesis and early embryogenesis, yet certain marks can persist or be re-established, enabling limited transgenerational effects. During development in mammals, epigenetic reprogramming involves partial erasure of patterns, particularly in germ cells (PGCs), where global demethylation occurs to reset the epigenome for totipotency. However, this process is not complete; genomic imprints—differentially methylated regions essential for parental-specific —resist erasure and are maintained through protective mechanisms involving specific DNA-binding proteins and modifications. This selective retention allows imprinted marks to be transmitted across generations, while most other epigenetic signals are wiped out, limiting the scope of transgenerational inheritance to specific loci. Mechanisms facilitating transgenerational effects include (ncRNA) transfer in and retention in oocytes. In , small RNAs such as piwi-interacting RNAs (piRNAs) and microRNAs can mediate paramutation-like silencing, where they guide or modifications in the offspring's , propagating environmental responses. For instance, exposure-induced changes in ncRNAs have been shown to integrate with , altering in subsequent generations. In oocytes, certain variants and their post-translational modifications are retained post-fertilization, escaping the paternal pronuclear demethylation wave and potentially influencing embryonic . These RNA- and -based carriers provide a molecular basis for bypassing partial barriers. Evidence from model organisms demonstrates these effects clearly. In rats, gestational exposure to the fungicide vinclozolin induces DNA hypomethylation in sperm, leading to transgenerational transmission of reduced fertility and increased disease susceptibility across at least three generations (F1 to F3), with altered methylation at over 200 loci persisting in the germline. Similarly, in Caenorhabditis elegans, 2024 studies have confirmed that small RNAs mediate heritable avoidance behaviors against pathogens, sustaining epigenetic silencing for multiple generations through ncRNA amplification in the germline. These findings highlight RNA-directed mechanisms as robust in invertebrates, contrasting with the more restricted inheritance in vertebrates, though reproducibility of such effects remains debated. In humans, transgenerational effects remain largely correlative, with historical studies providing suggestive evidence. The Överkalix study in northern linked grandpaternal during periods of food scarcity, such as the 1911–1912 events, to increased metabolic disease risk in grandchildren, inherited through the paternal line. While direct causation is challenging to establish due to ethical constraints on studies, these observations align with animal models and underscore the role of early-life exposures in shaping intergenerational health outcomes via epigenetic marks.

Epigenetics in Organisms and Systems

Bacterial Epigenetics

Bacterial epigenetics refers to heritable changes in gene expression that do not involve alterations to the underlying DNA sequence, primarily mediated through DNA methylation and nucleoid structuring in prokaryotes. Unlike eukaryotic systems, bacteria lack histones and instead rely on nucleoid-associated proteins (NAPs) such as HU and integration host factor (IHF) to compact and organize the chromosome, influencing gene accessibility and expression. HU, a highly abundant heterodimeric protein, bends and loops DNA to facilitate nucleoid compaction and regulate transcription, while IHF specifically binds to curved DNA sequences to promote architectural changes that affect promoter activity and replication initiation. These proteins enable dynamic chromatin-like organization essential for epigenetic regulation in the absence of nucleosomes. A key mechanism in bacterial epigenetics is DNA methylation, exemplified by the DNA methyltransferase (Dam) enzyme in , which specifically methylates the residue in GATC sequences shortly after replication. This methylation regulates the expression of numerous genes, including those involved in , by altering promoter activity and protein-DNA interactions; for instance, in Salmonella typhimurium, Dam methylation regulates at least 20 genes induced during , including -associated genes, enabling to host environments. In contrast to eukaryotic methylation, bacterial methylation like Dam provides rapid, sequence-specific epigenetic marks that propagate through cell divisions via hemi-methylated intermediates. Restriction-modification (RM) systems represent another cornerstone of bacterial epigenetics, functioning as primitive immune barriers that distinguish self from foreign DNA while exerting heritable epigenetic control. These systems consist of a methyltransferase that epigenetically modifies specific DNA motifs on the host and a restriction endonuclease that cleaves unmethylated DNA, such as from invading phages; the methylation pattern is inherited through semi-conservative replication, where hemi-methylated daughter strands are fully methylated before becoming susceptible to restriction. Approximately 40% of bacterial genomes harbor Type II RM systems, which not only defend against phages but also modulate by selectively permitting or blocking the integration of exogenous DNA, thereby shaping bacterial evolution and diversity. Phase variation, a epigenetic switch for reversible changes, is often mediated by methylation-dependent mechanisms in , allowing population-level bet-hedging against environmental stresses. In , flagellar phase variation between phase 1 (FliC) and phase 2 (FljB) antigens is regulated by at the hin locus, where methylation blocks recombinase access to GATC sites, stabilizing the current phase until demethylation permits switching; this heritable enhances immune evasion by generating antigenic diversity within a clonal . Such -blocked recombination exemplifies how bacterial epigenetics facilitates adaptive phenotypic heterogeneity without genetic .

Epigenetics in the Brain

Epigenetic mechanisms play a crucial role in the by modulating neural , which enables adaptive changes in neuronal structure and function in response to experience. These processes involve dynamic alterations in , modifications, and non-coding RNAs that influence without altering the underlying DNA sequence. In neural contexts, epigenetics facilitates the fine-tuning of synaptic connections and circuit remodeling, essential for learning, , and behavioral . Disruptions in these mechanisms can impair cognitive functions and contribute to neuropsychiatric conditions. During neuronal , the repressor element-1 silencing (REST) mediates repression through recruitment of Polycomb repressive complex 2 (PRC2), which deposits the mark at neuronal promoters. This epigenetic silencing prevents premature expression of neuron-specific genes in non-neuronal cells, ensuring proper timing of during development. In embryonic stem cells and neural progenitors, REST-associated complexes maintain poised states, characterized by bivalent and marks, which resolve into active or repressed configurations as cells commit to neuronal lineages. Epigenetic regulation is integral to memory formation, particularly through modulation of synaptic plasticity like (LTP) in the . (HDAC) inhibitors, such as and , enhance LTP by increasing histone acetylation at promoters of plasticity-related genes, thereby promoting their transcription and strengthening synaptic efficacy. This effect is mediated via activation of the CREB:CBP transcriptional complex, which drives expression of genes involved in formation and . Additionally, learning experiences induce demethylation at the BDNF promoter, particularly exon IV, facilitating increased expression of (BDNF), a key regulator of synaptic growth and LTP maintenance during contextual . Stress responses in the are epigenetically shaped, with exposure altering patterns in the to prime future reactivity. Elevated during periods of hippocampal , such as in early development, induce lasting hypermethylation at stress-responsive gene loci, shifting the transcriptional set point toward heightened sensitivity in adulthood. This programming effect persists, influencing expression and hypothalamic-pituitary-adrenal axis feedback, thereby modulating vulnerability to . Recent advances in single-cell have illuminated cell-type-specific epigenetic landscapes in the , revealing distinct DNA modification profiles between neurons and . For instance, a 2023 study using whole-genome oxidative demonstrated that neurons exhibit higher levels of hydroxymethylation (hmC) at sites compared to . Aberrant patterns in the , such as hypermethylation of neuronal genes, have been implicated in , contributing to dysregulated and synaptic dysfunction.

Epigenetics in Aging

Epigenetic changes accumulate with age, contributing to and reduced through mechanisms such as epigenetic drift, a characterized by the progressive loss of patterns, particularly at regions. This drift leads to global hypomethylation and site-specific hypermethylation, resulting in the erosion of structure and increased genomic instability. In aging cells, this loss of epigenetic fidelity disrupts gene regulation, promoting the expression of transposable elements and inflammation-associated genes, which exacerbate . A key tool for quantifying these age-related epigenetic alterations is the , exemplified by Horvath's 2013 model, which utilizes levels at 353 specific CpG sites across various human tissues to predict chronological age with a error of 3.6 years. This clock reveals accelerated epigenetic aging in conditions of cellular stress and correlates with attrition, where progressive shortening is associated with the erosion of the repressive mark at telomeric regions, further linking epigenetic instability to chromosomal end protection failure. Such correlations highlight how epigenetic drift intersects with dynamics to drive organismal aging phenotypes. Interventions targeting epigenetic mechanisms, such as caloric restriction, mitigate these changes by activating deacetylases like SIRT1, which enhance deacetylation and maintain integrity, thereby extending lifespan in model organisms including , , and mice. By increasing NAD+ levels, caloric restriction boosts activity, counteracting age-related hyperacetylation of and promoting cellular resilience against . Recent advancements in 2025 have integrated multi-omics data into epigenetic clocks, such as ensemble models combining with other layers like transcriptomics, achieving improved prediction accuracy with a median absolute error approaching ±2.5 years and higher correlations (R=0.98) to chronological age. These updates enable more precise tracking of interventions' effects on biological age, underscoring epigenetics' role in longevity research.

Medical and Clinical Applications

Genomic Imprinting and Twin Studies

represents a key epigenetic mechanism that silences specific parental , ensuring monoallelic expression of approximately 150 genes in the . These imprinted genes are crucial for , development, and metabolism, with the IGF2/H19 locus on chromosome 11p15.5 exemplifying parent-of-origin effects: the paternal IGF2 allele is expressed to promote fetal , while the maternal H19 allele produces a that represses IGF2 on the same chromosome. This differential expression arises from epigenetic marks established during , where the paternal and maternal alleles acquire distinct modifications that are stably maintained through cell divisions. The primary mechanism of genomic imprinting involves imprinting control regions (ICRs) characterized by germline-derived differentially methylated regions (). These , such as the intergenic between IGF2 and H19, undergo allele-specific in (paternal marks) or oocytes (maternal marks), often involving binding sites that regulate looping and insulator function. For instance, hypomethylation of the paternal H19 allows -mediated insulation, preventing IGF2 activation from the maternal , while hypermethylation on the paternal permits IGF2 expression. These gamete-specific imprints resist the global epigenetic in early embryos, ensuring parent-of-origin fidelity throughout development. Disruptions in establishment or maintenance can lead to imprinting disorders, as seen in Beckwith-Wiedemann syndrome, where loss of maternal at the H19/IGF2 ICR results in biallelic IGF2 expression, causing overgrowth, , and elevated cancer risk. Twin studies, especially in monozygotic () twins who share identical genomes, highlight how environmental factors drive epigenetic variation independent of . Although MZ twins start with highly concordant epigenomes at birth, differences in accumulate over time due to divergent lifestyles, such as , exercise, and stress exposure. By adulthood, older MZ twins (aged 50-70 years) exhibit substantial intra-pair discordance in methylation profiles, with studies reporting differences across hundreds of CpG sites and global hypomethylation patterns reflecting environmental divergence. For example, in the Finnish Twin Cohort, BMI discordance between MZ twin pairs is associated with accelerated epigenetic aging, including altered at obesity-related loci and repetitive elements like LINE-1, where higher correlates with hypomethylation and increased epigenetic age by approximately 1 month per BMI unit. These findings underscore how non-shared environments modulate epigenetic drift, providing a model for dissecting gene-environment interactions in imprinting stability and disease susceptibility.

Role in Diseases

Epigenetic dysregulation plays a central in the of various diseases by altering without changing the underlying DNA sequence. In cancer, aberrant patterns are particularly prominent, with promoter hypermethylation leading to of tumor suppressor genes and global hypomethylation contributing to genomic instability and activation. These changes are observed across multiple tumor types and represent early events in tumorigenesis. In cancers, promoter hypermethylation of the p16^INK4a gene silences this key tumor suppressor, which normally inhibits progression, thereby promoting uncontrolled proliferation; this mechanism is documented in various carcinomas, including those of the head, neck, and . Complementing this, global DNA hypomethylation affects repetitive elements and gene regions, leading to the activation of oncogenes and chromosomal instability, a phenomenon noted in the majority of human tumors as an early oncogenic driver. For instance, hypomethylation of LINE-1 retrotransposons, a marker of global demethylation, is reduced to 55-60% in tumor tissues compared to 70-90% in normal cells, correlating with aggressive disease features. Neurological disorders also exhibit profound epigenetic alterations, such as in Fragile X syndrome, where hypermethylation of the FMR1 promoter silences the gene encoding the fragile X mental retardation protein, essential for synaptic function, resulting in intellectual disability and behavioral deficits; this methylation occurs post-expansion of CGG repeats and is a hallmark of full-mutation cases. In addiction, particularly cocaine dependence, acute and chronic exposure induces histone H3 acetylation in the nucleus accumbens, a brain reward region, enhancing the expression of immediate-early genes like c-fos and reinforcing drug-seeking behavior through altered transcriptional plasticity. This cocaine-mediated increase in histone acetylation can reach approximately 50% above baseline levels following repeated administration. Metabolic diseases like type 2 diabetes involve epigenetic changes in pancreatic beta cells, where altered DNA methylation patterns impair insulin secretion and beta-cell identity maintenance. Specifically, hypermethylation or hypomethylation of genes such as PDX1 and INS disrupts glucose-stimulated insulin secretion, contributing to hyperglycemia and disease progression in susceptible individuals exposed to environmental factors like obesity. These methylation shifts are dynamic and can be influenced by aging and lifestyle, underscoring epigenetics as a bridge between environmental triggers and beta-cell dysfunction. Recent clinical data from 2025 highlight the prognostic value of epigenetics in neurodegenerative diseases, with -based epigenetic clocks derived from peripheral blood predicting progression from cognitively normal states to or , offering insights up to several years before symptom onset. Such signatures capture accelerated biological aging and correlate with and accumulation. Additionally, single nucleotide polymorphisms (SNPs) at CpG sites, including those near UBASH3B and NFKBIE loci, modulate local levels, thereby influencing disease risk by altering gene-epigenome interactions in immune and inflammatory pathways. These meSNPs (methylation quantitative loci) explain a significant portion of variation in and link genetic variants to phenotypic outcomes in complex diseases.

Epigenetic Therapies and Drugs

Epigenetic therapies target enzymes involved in , modification, and to reverse aberrant epigenetic alterations associated with diseases, particularly cancer. These drugs modulate without altering the underlying DNA sequence, offering potential for treating conditions where epigenetic dysregulation plays a key role. As of 2025, several classes of epigenetic inhibitors have received FDA approval, primarily for hematologic malignancies, while investigational agents continue to advance in clinical development. Histone deacetylase (HDAC) inhibitors represent one of the earliest approved classes of epigenetic drugs, functioning by blocking HDAC enzymes that remove acetyl groups from histones, leading to a more open chromatin structure and reactivation of silenced tumor suppressor genes. Vorinostat (Zolinza), approved by the FDA in 2006, is a pan-HDAC inhibitor indicated for the treatment of cutaneous manifestations in patients with cutaneous T-cell lymphoma (CTCL) who have progressive, persistent, or recurrent disease on or following two systemic therapies. By inhibiting HDACs, vorinostat promotes histone acetylation, which reactivates epigenetically silenced genes and induces cell cycle arrest, differentiation, or apoptosis in cancer cells. Other HDAC inhibitors, such as romidepsin and belinostat, have also gained approval for specific lymphomas, expanding the therapeutic arsenal for epigenetic modulation in hematologic cancers. DNA methyltransferase (DNMT) inhibitors work by incorporating into DNA and covalently trapping enzymes, resulting in DNA hypomethylation that restores expression of genes silenced by hypermethylation. (Vidaza), approved by the FDA in 2004, and (Dacogen), approved in 2006, are nucleoside analogs used for the treatment of patients with myelodysplastic syndromes (MDS), including those who have failed prior or are ineligible for transplantation. These agents induce hypomethylation of DNA, leading to re-expression of tumor suppressor genes and improved hematologic responses in a significant proportion of MDS patients. Low-dose regimens of these drugs prioritize their demethylating effects over , highlighting their role as targeted epigenetic therapies. An oral formulation of combined with cedazuridine was approved in 2020 to enhance patient convenience while maintaining efficacy. Among emerging epigenetic drugs from 2020 to 2025, inhibitors of , a , have shown promise in solid tumors. Tazemetostat (Tazverik), the first EZH2 inhibitor, received accelerated FDA approval in 2020 for adults and pediatric patients aged 16 years and older with locally advanced or metastatic not eligible for complete resection. By selectively inhibiting EZH2, tazemetostat reduces marks, alleviating repression and promoting antitumor activity in EZH2-mutated or overexpressing cancers. and extraterminal (BET) inhibitors, which disrupt the reading of acetylated histones by BET proteins, are under investigation for inflammatory conditions and cancers. For instance, selective BET inhibitors like VYN202 are in clinical trials for , demonstrating effects by attenuating pro-inflammatory transcription. Combination therapies integrating epigenetic drugs with have emerged as a strategy to enhance antitumor immune responses in cancer trials. HDAC and DNMT inhibitors can increase presentation and reprogram the , synergizing with inhibitors like PD-1/ blockers to improve response rates in solid and hematologic malignancies. Ongoing phase III trials, such as those combining inhibitors with or , underscore this approach's potential. As of 2025, more than 20 epigenetic drugs are in phase III trials, with many emphasizing precision medicine guided by epigenomic to identify responsive patients.

Research and Emerging Advances

Epigenome Editing

Epigenome editing represents a targeted approach to modify epigenetic marks on DNA and histones without altering the underlying genetic sequence, primarily leveraging catalytically inactive Cas9 (dCas9) fused to epigenetic effector domains. This technology enables precise control over by recruiting modifiers such as TET1 for to activate silenced genes or for transcriptional repression through formation. For instance, dCas9-TET1 fusions catalyze the oxidation of to at specific loci, facilitating demethylation and gene activation, while dCas9- recruits repressive complexes like TRIM28 and SETDB1 to induce marks for long-term silencing. These fusions are guided by single-guide RNAs (sgRNAs) to user-defined genomic sites, allowing multiplexed editing for complex regulatory landscapes. In therapeutic applications, epigenome editing has shown promise in silencing latent viral reservoirs and correcting hemoglobinopathies. For HIV, dCas9-KRAB fusions targeted to the HIV-1 long terminal repeat (LTR) promoter have achieved stable repression of proviral transcription in cell models by imposing repressive chromatin states, reducing viral reactivation without excising the DNA. Similarly, in sickle cell disease, dCas9 fused to activators like VP64 or p300 has been used to demethylate and acetylate enhancers at the γ-globin locus, boosting fetal hemoglobin (HbF) production to counteract sickling; recent epigenome editing of repressor binding sites in human hematopoietic stem cells restored HbF synthesis and ameliorated disease phenotypes in preclinical models. A notable 2025 preclinical study demonstrated the potential of epigenome editing for metabolic disorders, where a multiplex dCas9-p300 system reprogrammed human fibroblasts into insulin-producing β-like cells by activating key genes such as PDX1, NKX6.1, and MAFA through targeted histone acetylation. Key advantages of epigenome editing include its reversibility, as epigenetic marks can be dynamically altered or reset, and the avoidance of off-target mutations since dCas9 lacks nuclease activity, minimizing risks associated with double-strand breaks. However, challenges persist, particularly in efficient delivery of large dCas9 fusion proteins to target tissues, often requiring viral vectors or nanoparticles that face immunogenicity and transduction barriers, and in ensuring the persistence of edits against cellular turnover and incomplete epigenetic memory.00721-X) Ongoing efforts focus on optimizing effector domains and delivery systems to enhance durability in vivo.00721-X)

Advanced Methodologies

Single-cell epigenomics has advanced significantly post-2020, enabling high-resolution profiling of epigenetic states in individual cells. Single-cell bisulfite sequencing (scBS-seq) measures DNA methylation at base-pair resolution by converting unmethylated cytosines to uracils, allowing genome-wide analysis when paired with single-cell RNA sequencing. Recent tools like MethSCAn, introduced in 2024, process datasets up to 100,350 cells, incorporating read-position-aware quantitation and variably methylated regions detection to improve signal-to-noise ratios and cell-type separation, identifying over 63,000 such regions in benchmarks. Complementing this, single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) maps chromatin accessibility, with 2024 innovations like CellSpace providing scalable, sequence-informed embeddings that enhance resolution by learning joint representations of DNA k-mers and cells, mitigating batch effects across thousands of cells from multiple donors and inferring transcription factor activities without predefined motifs. Long-read sequencing technologies, such as those from PacBio and Oxford Nanopore, have revolutionized the detection of phased epigenetic modifications over kilobase-scale regions. PacBio's highly accurate long-read (HiFi) sequencing achieves over 99.9% accuracy and directly detects (5mC) modifications without chemical conversion, enabling haplotype-level phasing of patterns. Oxford Nanopore's R10.4 flowcells further improve calling accuracy with oxidative bisulfite sequencing benchmarks, reducing strand bias and supporting precise characterization across 7,000+ samples at 20x coverage. These methods surpass short-read approaches by resolving complex, repetitive genomic contexts where phasing is critical for understanding allele-specific epigenetics. Multi-omics integration combines epigenomic data with transcriptomic profiles to cell-type heterogeneity in bulk tissues. EpiDISH, an established framework updated through 2020, estimates cell-type fractions from arrays using reference-based algorithms like robust partial correlations and constrained projection, supporting for blood, epithelial, and breast tissues. When integrated with transcriptome data, it facilitates of cell-type-specific differentially methylated cytosines, enhancing inference in epigenome-wide studies by virtually microdissecting mixed samples. Artificial intelligence applications, particularly , now predict epigenetic marks directly from DNA sequences with high fidelity. Recent 2025 models, such as Methyl-GP, achieve over 95% accuracy in forecasting three types of (5mC, 4mC, 6mA) across species by leveraging Gaussian processes and sequence features, outperforming convolutional neural networks in cross-validation tests. These predictive tools, built on architectures like transformers, enable annotation of uncharacterized genomes and reveal sequence determinants of epigenetic patterning without experimental assays.

Future Challenges and Directions

One major challenge in epigenetics research lies in distinguishing meaningful epigenetic signals from inherent , where variations in epigenetic marks can obscure true biological signals, complicating the of in cellular identity and processes. This issue is particularly pronounced in single-cell analyses, where technical from low-input samples further hampers accurate profiling of patterns. Additionally, off-target effects remain a critical hurdle in epigenome editing technologies, such as CRISPR-based tools, where unintended modifications to non-target sites can lead to aberrant or long-term cellular dysfunction, despite efforts to minimize them through modular designs. Validating in humans presents further difficulties, as ethical constraints limit direct experimentation, and observational studies often struggle to disentangle epigenetic effects from genetic or environmental confounders across generations. Looking ahead, personalized epigenomedicine holds promise through the integration of artificial intelligence to analyze complex epigenomic datasets, enabling tailored interventions that account for individual epigenetic profiles alongside genetic data. In environmental epigenetics, emerging directions focus on how climate change induces heritable epigenetic marks, such as altered DNA methylation in response to temperature shifts and pollution, potentially affecting population-level adaptation and disease susceptibility. A 2025 update on environmental epigenetics emphasizes the need for global initiatives to map pollution-induced changes, akin to exposome projects that track cumulative exposures and their epigenetic footprints to inform public health strategies. Integrating epigenetics with genomics, particularly by combining epigenetic risk scores with polygenic risk scores, enhances predictive accuracy for complex traits and diseases, offering a more comprehensive framework for risk stratification. Ethical concerns in epigenetics are amplified by the risks of heritable , where even transient modifications could inadvertently produce transgenerational effects, raising questions about for and equitable to such technologies. Furthermore, faces misuse through pseudoscientific claims, such as unproven "epigenetic detox" regimens that falsely promise to reverse environmental damage via lifestyle interventions, diverting attention from evidence-based research and potentially harming public trust.

References

  1. [1]
    What is epigenetics?: MedlinePlus Genetics
    Jun 11, 2021 · Epigenetics is the study of how cells control gene activity without changing the DNA sequence. "Epi-"means on or above in Greek.
  2. [2]
    Epigenetics - National Human Genome Research Institute
    Epigenetics (also sometimes called epigenomics) is a field of study focused on changes in DNA that do not involve alterations to the underlying sequence.
  3. [3]
    Epigenetics: Definition, Mechanisms and Clinical Perspective - NIH
    Epigenetics has been defined and today is generally accepted as ''the study of changes in gene function that are mitotically and/or meiotically heritable.
  4. [4]
    The epigenotype. 1942 - PubMed
    Dr Waddington proposes the name 'epigenotype.' He here describes some of the general characteristics of an epigenotype, with special reference to the fruit-fly ...
  5. [5]
    [PDF] epigenotype.pdf - Ehud Lamm
    Waddington makes it very clear that the 'organizing relations' are a dynamic network of interactions occurring during embryogenesis, and that the study of this ...
  6. [6]
    Epigenetics: Principles and Practice - PMC - NIH
    Epigenetics is defined as heritable changes in gene expression that are, unlike mutations, not attributable to alterations in the sequence of DNA.
  7. [7]
    An operational definition of epigenetics - Genes & Development
    Definition: “An epigenetic trait is a stably heritable phenotype resulting from changes in a chromosome without alterations in the DNA sequence.” The ...Missing: modern | Show results with:modern
  8. [8]
    Genetics, Epigenetic Mechanism - StatPearls - NCBI Bookshelf - NIH
    Epigenetics is the study of heritable and stable changes in gene expression that occur through alterations in the chromosome rather than in the DNA sequence.Missing: unlike | Show results with:unlike
  9. [9]
    X-chromosome inactivation: a historic topic that's still hot
    Nov 23, 2023 · XCI consists of the transcriptional silencing of one X chromosome in XX females to compensate for the excess of X-linked gene products compared with XY males.
  10. [10]
    Epigenetic Mechanisms of Genomic Imprinting: Common Themes in ...
    This review examines some examples of the epigenetic mechanisms responsible for genomic imprinting in mammals, plants, and insects.
  11. [11]
    Epigenetics: The origins and evolution of a fashionable topic
    Aug 1, 2016 · Waddington defined “epigenetics” as the “whole complex of developmental processes” that lie between “genotype and phenotype”. In his ...
  12. [12]
    Introduction: sketches of a conceptual history of epigenesis
    Oct 23, 2018 · Epigenesis was understood as the idea that there was no pre-established organization whereas with preformationism, the parts were already ...
  13. [13]
    From Mendel to epigenetics: History of genetics - ScienceDirect.com
    The origins of genetics are to be found in Gregor Mendel's memoir on plant hybridization (1865). However, the word 'genetics' was only coined in 1906.Missing: variability | Show results with:variability
  14. [14]
    A Brief History of Epigenetics - PMC - PubMed Central - NIH
    The term “epigenetics” was originally used to denote the poorly understood processes by which a fertilized zygote developed into a mature, complex organism.2. Clues From Genetics And... · 5. The Role Of Chromatin · Figure 1Missing: rediscovery dogma
  15. [15]
    Conrad Waddington and the origin of epigenetics
    Mar 15, 2015 · Waddington's diagram to show how the developmental landscape relates to individual genes (bottom pegs) through networks of interactions in the ...Missing: canalization | Show results with:canalization
  16. [16]
    Epigenetic Landscape | Embryo Project Encyclopedia
    Oct 30, 2007 · Waddington envisioned the epigenetic landscape as a series of ridges and valleys a cell can traverse on its way to a final tissue type. He ...
  17. [17]
    DNA modification mechanisms and gene activity during development
    DNA modification mechanisms and gene activity during development. Science. 1975 Jan 24;187(4173):226-32. Authors. R Holliday, J E Pugh. PMID: 1111098. No ...
  18. [18]
    DNA methylation: a historical perspective - ScienceDirect
    Holliday and Pugh proposed a model for switching genes on and off that utilizes two different specialized enzymes for de novo methylation and maintenance ...
  19. [19]
    From 1957 to Nowadays: A Brief History of Epigenetics - PMC
    Oct 14, 2020 · Indeed, DNA methylation was first described in 1965, while histone methylation, acetylation, phosphorylation, ubiquitylation, sumoylation and ...2. Dna Methylation · 6. Histone Variants · 9. Epidrugs: Towards...
  20. [20]
    Two pioneers of epigenetics: their different paths to chromatin ...
    Stuart Schreiber discovered an enzyme in mammalian cells that removed histone acetylation marks and was related to a yeast gene known to regulate transcription.
  21. [21]
    DNA Methylation and Its Basic Function | Neuropsychopharmacology
    Jul 11, 2012 · Historically, DNA methylation was discovered in mammals as early as DNA was identified as the genetic material (Avery et al, 1944; McCarty and ...
  22. [22]
    DNA methylation patterns and epigenetic memory
    Mammalian DNA methylation patterns vary in time and space. In human somatic cells, m5C accounts for ∼1% of total DNA bases and therefore affects 70%–80% of all ...
  23. [23]
    Methylation across the central dogma in health and diseases - Nature
    Aug 25, 2023 · DNMT3A and DNMT3B are mainly responsible for de novo DNA methylation and DNMT1 for the maintenance of the established DNA methylation pattern ...
  24. [24]
    DNA methylation, imprinting and cancer - Nature
    Feb 28, 2002 · While global hypomethylation is detectable in the cancer genome, regions of hypermethylation are commonly found. ... Global DNA hypomethylation ...Establishment Of Methylation... · Methylation And... · Cpg Island Hypermethylation
  25. [25]
    DNA Methyltransferases: From Evolution to Clinical Applications
    The MTases family catalyzes DNA methylation. This process is conserved from archaea to eukaryotes, from fertilization to every stage of development, and from ...2.2. Dna Cytosine 5′mtases... · 2.3. Regulating Dna... · 2.4. Dnmts In Embryonic...<|control11|><|separator|>
  26. [26]
    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 ...
  27. [27]
    Histone post-translational modifications — cause and consequence ...
    Mar 25, 2022 · This Review explores the contribution of histone PTMs to the regulation of genome function by discussing when these modifications play a causative (or ...
  28. [28]
    Regulation of chromatin by histone modifications | Cell Research
    Feb 15, 2011 · Here, we describe the known histone modifications, define where they are found genomically and discuss some of their functional consequences.
  29. [29]
    Translating the Histone Code - Science
    Translating the Histone Code · The histone code hypothesis predicts that the modification marks on the histone tails should provide binding sites for effector ...
  30. [30]
    Mapping epigenetic modifications by sequencing technologies
    Sep 1, 2023 · Over 100 distinct modifications have been found in histone, including acetylation (Ac), methylation (Me), phosphorylation (P), ubiquitylation ( ...
  31. [31]
    Ezh2, the histone methyltransferase of PRC2, regulates the balance ...
    Loss of function of Ezh2 removes the repressive mark of trimethylated histone H3 at lysine 27 (H3K27me3) in cortical progenitor cells and also prevents its ...
  32. [32]
    MicroRNA-29 family reverts aberrant methylation in lung cancer by ...
    Oct 2, 2007 · Here we show that expression of miR-29s is inversely correlated to DNMT3A and -3B in lung cancer tissues, and that miR-29s directly target both DNMT3A and -3B.Missing: DNMT3 | Show results with:DNMT3
  33. [33]
    Epigenetics: heterochromatin meets RNAi | Cell Research - Nature
    Feb 3, 2009 · Exogenous siRNAs are thus capable of stable and specific epigenetic regulation of target genes, raising the possibility that similar mechanism ...
  34. [34]
    piRNAs against transposons preserve genome integrity
    Nov 10, 2017 · This review features our current understandings of mammalian PIWI-interacting RNAs (piRNAs) and their role in TE regulation in spermatogenesis.
  35. [35]
    Long non-coding RNAs: definitions, functions, challenges ... - Nature
    Jan 3, 2023 · We address lncRNA definition, nomenclature, conservation, expression, phenotypic visibility, functional assays and molecular mechanisms ...
  36. [36]
    Changes in the middle region of Sup35 profoundly alter the ... - PNAS
    Dec 10, 2002 · The yeast prion [PSI+] provides an epigenetic mechanism for the inheritance of new phenotypes through self-perpetuating changes in protein ...
  37. [37]
    Prions are a common mechanism for phenotypic inheritance in wild ...
    The self-templating conformations of yeast prion proteins act as epigenetic elements of inheritance. Yeast prions might provide a mechanism for generating ...Prions Are A Common... · Introduction · Sup35 Amyloid Inheritance In...
  38. [38]
    Prion-like domains as epigenetic regulators, scaffolds for subcellular ...
    Sep 15, 2016 · In this review, we highlight recent advances in our understanding of the role of prions in enabling rapid adaptation to environmental stress in yeast.
  39. [39]
    Epigenetic pioneering by SWI/SNF family remodelers - ScienceDirect
    Jan 18, 2024 · Here, we review evidence for the centrality of the conserved SWI/SNF family of nucleosome remodeling complexes, both in pioneering and in mediating enhancer- ...
  40. [40]
    The SWI/SNF chromatin remodeling complex: a critical regulator of ...
    Apr 26, 2024 · The SWI/SNF chromatin remodeling complex functions by displacing nucleosomes near important regulatory sites which can facilitate transcription ...Introduction · History of SWI/SNF discovery... · Glucose metabolism · Perspectives
  41. [41]
    m6A RNA methylation: from mechanisms to therapeutic potential
    m6A methylation regulates gene expression by influencing numerous aspects of mRNA metabolism, including pre‐mRNA processing, nuclear export, decay, and ...
  42. [42]
    The role of m6A modification in the biological functions and diseases
    Feb 21, 2021 · Furthermore, RNA m6A modification has been documented to play important roles for regulating RNA splicing, translation, stability, translocation ...
  43. [43]
    Heterochromatin organization and phase separation - PMC - NIH
    Jan 29, 2023 · Protein-based liquid–liquid phase separation (LLPS) plays an essential role in the formation and dynamic regulation of heterochromatin compartmentalization.
  44. [44]
    Epigenetic regulatory layers in the 3D nucleus - ScienceDirect.com
    Feb 1, 2024 · Euchromatin represents the transcriptionally active, decompacted form of chromatin and tends to physically localize toward the nuclear interior, ...
  45. [45]
    DNA topology in chromatin is defined by nucleosome spacing
    Oct 27, 2017 · The core of a typical nucleosome contains 145 to 147 base pairs (bp) of DNA making 1.7 left-handed superhelical turns around a histone octamer ( ...
  46. [46]
    Nucleosome Positioning in Saccharomyces cerevisiae - ASM Journals
    Jun 6, 2011 · Thus, antinucleosomal sequences such as poly(dA:dT) tracts create a “barrier” that favors the formation of highly positioned nucleosomes ...
  47. [47]
    Determinants of nucleosome positioning - PMC - NIH
    The DNA sequence is critical for rotational positioning along the DNA helix, and it also is an important determinant for nucleosome occupancy. In particular, ...
  48. [48]
    Chromatin insulators: regulatory mechanisms and epigenetic ...
    Insulators are regulatory DNA elements that create boundaries in chromatin, delineating the ranges over which other regulatory influences take effect.Missing: authoritative source
  49. [49]
    Chromatin Domains, Insulators, and the Regulation of Gene ... - NIH
    The DNA sequence elements called insulators have two basic kinds of properties. Barrier elements block the propagation of heterochromatic structures into ...Missing: authoritative source
  50. [50]
    Structural insights into histone binding and nucleosome assembly ...
    Aug 25, 2023 · CAF-1 is a heterotrimeric protein complex responsible for the deposition of newly synthesized histones H3 and H4 onto DNA in replication-coupled nucleosome ...Missing: semi- conservative
  51. [51]
    Nucleosome positioning: How is it established, and why does it ...
    Mar 15, 2010 · Here, we review the literature on mapping nucleosome positions in various organisms, and discuss how nucleosome positions are established.
  52. [52]
    DNA damage and Repair Modify DNA methylation and Chromatin ...
    Sep 15, 2016 · We find that HR modifies the DNA methylation pattern of the repaired segment. HR also alters local histone H3 methylation as well chromatin ...
  53. [53]
    Base Excision Repair of Tandem Modifications in a Methylated CpG ...
    Likewise, 8-oxoG formed at CpG dinucleotides inhibits the methylation of the adjacent cytosine (34–36), and this could result in an altered methylation pattern.
  54. [54]
    Oxidative DNA damage is epigenetic by regulating gene ... - PNAS
    Jan 31, 2017 · We demonstrate that ROS-mediated oxidation of DNA to yield 8-oxo-7,8-dihydroguanine (OG) in gene promoters is a signaling agent for gene activation.
  55. [55]
    Non-homologous end joining induced alterations in DNA methylation
    The mechanism described here provides evidence for alterations in methylation profiles as a result of NHEJ repair, a process which could explain the epigenetic ...Missing: junctions | Show results with:junctions
  56. [56]
    PARP1-dependent recruitment of KDM4D histone demethylase to ...
    Feb 3, 2014 · Our findings show that KDM4D lysine demethylase is swiftly recruited to DNA breakage sites via its C-terminal region in a PARP1-dependent manner.Parp1-Dependent Recruitment... · Results · Parp1 Adp Ribosylates Kdm4d...
  57. [57]
    ATM‐Dependent Recruitment of BRD7 is required for ...
    Sep 3, 2020 · Furthermore, ATM-mediated BRD7 phosphorylation is required for recruitment of the PRC2 complex, NuRD complex, DSB sensor complex MRE11-RAD50- ...
  58. [58]
    Human Imprinted Chromosomal Regions Are Historical Hot-Spots of ...
    Imprinted chromosomal regions have two unusual characteristics in terms of meiotic recombination: They have unusually high recombination rates compared with ...
  59. [59]
    Chromatin immunoprecipitation sequencing (ChIP-Seq) on ... - Nature
    Chromatin immunoprecipitation (ChIP) is a technique for identifying and characterizing elements in protein-DNA interactions involved in gene regulation or ...Missing: review | Show results with:review
  60. [60]
    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 ...
  61. [61]
    DNA Methylation: A Timeline of Methods and Applications - Frontiers
    Oct 24, 2011 · Methylation-Sensitive Restriction Enzymes. Restriction enzymes cleave DNA through recognition of specific nucleotide motifs. Amongst the ...
  62. [62]
    FISH Going Meso-Scale: A Microscopic Search for Chromatin ...
    Here we are reviewing recent developments in FISH-based approaches for detection, quantitative measurements, and validation of contact chromatin domains.
  63. [63]
    Single-cell sequencing technology applied to epigenetics for the ...
    Oct 11, 2023 · This paper provides a comprehensive review of the emerging analytical and experimental approaches of single-cell sequencing in various omics, focusing ...
  64. [64]
    MethPhaser: methylation-based long-read haplotype phasing of ...
    Jun 22, 2024 · We designed MethPhaser, a method that utilizes methylation signals from Oxford Nanopore Technologies to extend Single Nucleotide Variation (SNV)-based phasing.
  65. [65]
    Mapping Human Epigenomes - PMC - PubMed Central - NIH
    In this review, we discuss the current epigenome mapping toolkit and utilities of epigenome maps. We focus particularly on mapping of DNA methylation.
  66. [66]
  67. [67]
    DNA methylation analysis of human myoblasts during in vitro ... - NIH
    DNA methylation modulates the functions of key transcriptional factors to coordinately regulate muscle-related genes during myogenic differentiation.
  68. [68]
    DNA methylation and differentiation: silencing, upregulation and ...
    We review new insights into the functionality of DNA methylation during differentiation, with emphasis on the methylomes of myoblasts, myotubes and skeletal ...
  69. [69]
    Histone H3K27ac separates active from poised enhancers ... - PNAS
    We find that histone H3K27ac distinguishes active enhancers from inactive/poised enhancer elements containing H3K4me1 alone.
  70. [70]
    Transcriptional silencers: driving gene expression with the brakes on
    Silencers are regulatory DNA elements that reduce transcription from their target promoters; they are the repressive counterparts of enhancers.
  71. [71]
    MicroRNAs and epigenetics - Sato - 2011 - FEBS Press - Wiley
    Mar 12, 2011 · These findings suggest that epigenetic silencing of miR-9 loci constitutes an early event in breast carcinogenesis. Furthermore, the miR-9 ...
  72. [72]
    Modulation of Epigenetic Regulators and Cell Fate Decisions by ...
    The miRNA modulates the activity of SWI/SNF, which in turn modulates the expression of the miRNA through a double-negative feedback loop. Namely, miR-199a ...
  73. [73]
    Histone acylation marks respond to metabolic perturbations ... - Nature
    Dec 11, 2020 · Histone acetylation has been intimately linked to cellular metabolism because of its sensitivity to the abundance of acetyl-coenzyme A (Ac-CoA).
  74. [74]
    The many metabolic sources of acetyl-CoA to support histone ...
    The extent of histone acetylation is thus regulated by changes in the expression and activity of HAT and HDAC enzymes. In the field of cancer, it is often ...
  75. [75]
    Diet and the epigenome | Nature Communications
    Aug 28, 2018 · This diet promotes formation of β-hydroxybutyrate, an HDAC inhibitor, and leads to changes in H3ac and H3K4me3 in the hippocampus and rescue of ...
  76. [76]
    Food Bioactive HDAC Inhibitors in the Epigenetic Regulation of ...
    Model demonstrating that food bioactives (phytochemicals) inhibit histone deacetylase (HDAC) activity as a cardio-protective mechanism.
  77. [77]
    Reprogramming DNA methylation in the mammalian life cycle
    Jan 5, 2013 · In this review, we discuss the dynamics of DNA methylation reprogramming in PGCs and the zygote, the mechanisms involved and the biological significance of ...
  78. [78]
    Epigenetic reprogramming in the germline: towards the ground state ...
    Epigenetic reprogramming in the germline provides a developmental model to study the erasure of epigenetic memory as it occurs naturally in vivo.
  79. [79]
    DNA methylation dynamics during epigenetic reprogramming in the ...
    DNA methylation poses a fundamental epigenetic barrier that guides and restricts differentiation and prevents regression into an undifferentiated state.Dna Methylation · Dna Demethylation · Imprinted Genes<|separator|>
  80. [80]
    Integration of sperm ncRNA-directed DNA methylation and DNA ...
    Jan 12, 2021 · The current study investigates the potential integration of DNA methylation, ncRNA, and histone alterations in the epigenetic transgenerational ...
  81. [81]
    Epigenetic Transgenerational Inheritance of Altered Sperm Histone ...
    Mar 28, 2018 · The current study was designed to investigate the potential role of histones in sperm to help mediate the epigenetic transgenerational inheritance.
  82. [82]
    Endocrine Disruptor Vinclozolin Induced Epigenetic ...
    Vinclozolin has been shown to promote an epigenetic alteration in the germ line that appears to transmit a transgenerational disease state (8). The previous ...
  83. [83]
    Selfish conflict underlies RNA-mediated parent-of-origin effects
    Mar 6, 2024 · Our findings show that parent-of-origin effects can evolve by co-option of the piRNA pathway and hinder the spread of selfish genes that require sex for their ...
  84. [84]
    Human transgenerational responses to early-life experience - NIH
    Jul 25, 2014 · Compelling historical findings of paternal transgenerational effects in humans come from the Överkalix population in northern Sweden using ...
  85. [85]
    Paternal grandfather's access to food predicts all-cause and cancer ...
    Dec 11, 2018 · The findings from the Överkalix cohorts imply that grandparental access to food during their slow growth period can modify diabetes and all- ...
  86. [86]
    Coordination of genomic structure and transcription by the main ...
    HU is a highly abundant DNA architectural protein that is involved in compacting the DNA of the bacterial nucleoid and in regulating the main DNA transactions.
  87. [87]
    HU multimerization shift controls nucleoid compaction - Science
    Jul 29, 2016 · Similar to histones in eukaryotes, nucleoid-associated proteins (NAPs) are essential for bacterial nucleoid condensation and resulting ...Missing: epigenetics | Show results with:epigenetics<|separator|>
  88. [88]
    Nucleoid-associated proteins shape chromatin structure and ...
    A range of nucleoid-associated proteins (NAPs) in bacteria, which function to compact, structure, and regulate large portions of bacterial chromosomes.
  89. [89]
    An Essential Role for DNA Adenine Methylation in Bacterial Virulence
    Dam regulated the expression of at least 20 genes ... DNA methylation affects gene expression but not global chromatin structure in Escherichia coli , ...
  90. [90]
    The Mechanism by which DNA Adenine Methylase and PapI ...
    Roles of DNA adenine methylation in regulating bacterial gene expression and virulence ... regulates Pap phase variation in Escherichia coli. EMBO J. 1995 ...
  91. [91]
    Restriction-Modification Systems as Mobile Epigenetic Elements
    Restriction-modification systems consist of a modification enzyme that epigenetically methylates a specific DNA sequence, and a restriction endonuclease ( ...Missing: heritable | Show results with:heritable
  92. [92]
    Restriction-modification systems have shaped the evolution and ...
    Within R-M systems, Type II are the most abundant, present in 39.2% of bacterial genomes (3) with a mean of ∼0.5 systems per genome (5). Type II R-M systems ...
  93. [93]
    A Type I Restriction Modification System Influences Genomic ...
    Aug 2, 2021 · A Type I Restriction Modification System Influences Genomic Evolution Driven by Horizontal Gene Transfer in Paenibacillus polymyxa. Considered ...
  94. [94]
    Phase variation controls expression of Salmonella ... - NIH
    OxyR binding prevents GATC methylation in vitro. In epigenetic phase variation, DNA methylation confers heritability of the expression phase (van der Woude ...
  95. [95]
    Phase and Antigenic Variation in Bacteria - ASM Journals
    Flagellar phase variation in Salmonella ... Genetic analysis of the mechanism of the Salmonella phase variation site specific recombination system.
  96. [96]
    Molecular switches — the ON and OFF of bacterial phase variation
    Mar 1, 2002 · Inversions mediated by both FimB and FimE require the presence of Lrp, previously known as methylation blocking factor (Mbf). Type 1 ...
  97. [97]
    REST–Mediated Recruitment of Polycomb Repressor Complexes in ...
    Altogether we found that REST was required for the recruitment of RNF2 and the maintenance of H3K27Me3 on a selected number of neuronal genes and that REST and ...
  98. [98]
    Polycomb- and REST-associated histone deacetylases are ... - eLife
    Sep 24, 2014 · Poised neuronal genes are repressed by REST and Polycomb in embryonic stem cells independently and via different chromatin modifications.
  99. [99]
    Histone Deacetylase Inhibitors Enhance Memory and Synaptic ...
    Jun 6, 2007 · Our results suggest that HDAC inhibitors enhance memory processes by the activation of key genes regulated by the CREB:CBP transcriptional complex.
  100. [100]
    Epigenetic Regulation of bdnf Gene Transcription in the ...
    These results suggest epigenetic modification of the bdnf gene as a mechanism for isoform-specific gene readout during memory consolidation.
  101. [101]
    Glucocorticoid exposure during hippocampal neurogenesis primes ...
    Sep 22, 2020 · Our data suggest that early exposure to GCs can change the set point of future transcriptional responses to stress by inducing lasting DNAm changes.
  102. [102]
    Differential usage of DNA modifications in neurons, astrocytes, and ...
    Nov 13, 2023 · Neurons, astrocytes, and microglia demonstrate different genome-wide levels of mCG, hmCG, and mCH that are reproducible across analytical methods.<|separator|>
  103. [103]
    DNA Methylation and Schizophrenia: Current Literature and Future ...
    Oct 26, 2021 · Epigenetic modifications like DNA methylation are a risk factor for schizophrenia. Targeted gene approach studies attempted to find candidate gene methylation, ...<|control11|><|separator|>
  104. [104]
    Age-related epigenetic drift and phenotypic plasticity loss - NIH
    Nov 24, 2016 · Aging results in global hypomethylation in the DNA genome but induces hypermethylation in the regulatory regions of certain key genes (orange ...
  105. [105]
    Epigenetics and aging | Science Advances
    Jul 29, 2016 · A growing number of studies have revealed that progressive changes to epigenetic information accompany aging in both dividing and nondividing cells.
  106. [106]
    DNA methylation age of human tissues and cell types
    I refer to the 353 CpGs as (epigenetic) clock CpGs since their weighted average (formed by the regression coefficients) amounts to an epigenetic clock.
  107. [107]
    Telomere Dynamics in Cardiovascular Aging: From Molecular... - LWW
    Aug 1, 2025 · Shorter telomeres and epigenetic dysregulation (subtelomeric hypo-/hypermethylation, H3K9me3 loss) mark accelerated vascular aging, while ...<|control11|><|separator|>
  108. [108]
    Sirtuins and calorie restriction | Nature Reviews Molecular Cell Biology
    Feb 29, 2012 · Calorie restriction (CR) is by far the most robust intervention, genetic or environmental, for slowing down ageing in mammals.Original Research Papers · About This Article · Cite This ArticleMissing: models | Show results with:models
  109. [109]
    enhancing epigenetic age assessment with a multi-clock framework
    Aug 6, 2025 · Epigenetic clocks have transformed aging research by providing molecular biomarkers that estimate biological age based on DNA methylation (DNAm) ...
  110. [110]
    Genomic Imprinting: Insights into Diverse Epigenetic Regulatory ...
    Jul 22, 2025 · This review examines these different mechanisms throughout early embryonic development and offers insights into the interactions among key ...
  111. [111]
    Genomic imprinting in mammals: its life cycle, molecular ... - Nature
    Feb 1, 2011 · The epigenetic modifications including DNA methylation at the germline DMRs undergo dynamic reprogramming during germ cell development but, on ...
  112. [112]
    Mechanisms of Genomic Imprinting - PMC - PubMed Central - NIH
    Differential methylation of this region is observed in sperm and is maintained during the global methylation changes observed during early embryogenesis.
  113. [113]
    Genomic Imprinting: CTCF Protects the Boundaries - ScienceDirect
    The DNA-binding protein CTCF, which acts as a chromatin 'insulator', regulates imprinting of the mammalian Igf2 and H19 genes in a methylation-sensitive manner.
  114. [114]
    Epigenetic and genetic alterations of the imprinting disorder ... - Nature
    May 30, 2013 · Beckwith–Wiedemann syndrome (BWS) is a representative imprinting disorder characterized by macrosomia, macroglossia and abdominal wall defects, and exhibits a ...
  115. [115]
    Epigenetic differences arise during the lifetime of monozygotic twins
    Jul 18, 2005 · We found that, although twins are epigenetically indistinguishable during the early years of life, older monozygous twins exhibited remarkable differences.Missing: adulthood | Show results with:adulthood
  116. [116]
    Epigenetic variation during the adult lifespan: cross-sectional and ...
    In this study on 230 monozygotic twin pairs (MZ pairs), aged 18–89 years, we investigated the occurrence of epigenetic changes over the adult lifespan.
  117. [117]
    BMI is positively associated with accelerated epigenetic aging in ...
    We aimed to assess the cross-sectional association of body mass index (BMI) with age acceleration in twins to limit confounding by genetics and shared ...
  118. [118]
    The Relationship between Body Mass Index, Obesity, and LINE-1 ...
    The study found a negative association between BMI and LINE-1 methylation; obese women had lower LINE-1 methylation than normal weight women.
  119. [119]
    DNA hypomethylation in cancer cells - PMC - NIH
    DNA hypomethylation is the almost constant companion to hypermethylation of the genome in cancer, just usually (but not always) in different sequences.
  120. [120]
    Promoter Hypermethylation of Tumor-Suppressor Genes p16 INK4a ...
    Apr 12, 2019 · Silencing of tumor-suppressor genes (TSGs) by DNA promoter hypermethylation is an early event in carcinogenesis; hence, TSGs may serve as ...
  121. [121]
    Tumor DNA hypomethylation of LINE-1 is associated with low ... - NIH
    Previous studies have shown that DNA methylation levels of LINE-1 were from 70 to 90% in normal tissues (47,48), and were from 55 to 60% in tumor tissues. These ...
  122. [122]
    DNA Methylation, Mechanisms of FMR1 Inactivation and ... - NIH
    Feb 16, 2021 · In some of the FXS hESCs lines, the FMR1 promoter is partially methylated, ranging from 24% to 65%. These methylation levels remain stable in ...
  123. [123]
    CBP in the nucleus accumbens regulates cocaine-induced histone ...
    Nov 23, 2011 · A complete loss of CBP in NAc neurons results in decreased histone acetylation and significantly altered c-fos expression in response to cocaine.
  124. [124]
    Chronic Cocaine-Induced H3 Acetylation and Transcriptional ...
    Our present data show that chronic cocaine use induced an increase in histone acetylation of ∼50%, and augmenting this increase in histone acetylation with HDAC ...
  125. [125]
    DNA methylation in the pathogenesis of type 2 diabetes in humans
    This strengthened the notion that the age-related methylation changes improve β-cell function and provided proof of principle that methylation markers can be ...
  126. [126]
    Epigenetics in β-cell adaptation and type 2 diabetes - PMC - NIH
    Among genes that were differentially methylated and expressed, FICD and TPX2 were shown to repress GSIS function in the human β-cell line, EndoC-βH1 cells.
  127. [127]
    DNA methylation age from peripheral blood predicts progression to ...
    May 27, 2025 · We found that two second-generation epigenetic clocks, DNAmPhenoAge and DNAmGrimAge, predicted progression from cognitively normal aging to mild cognitive ...
  128. [128]
    SNPs located at CpG sites modulate genome-epigenome interaction
    Our data suggest a potential biological mechanism underlying associations between common genetic variants, epigenetic processes, and disease phenotypes. We ...
  129. [129]
    Epigenetic drugs in cancer therapy - PMC - PubMed Central
    Feb 26, 2025 · Several epi-drugs are FDA approved for the treatment of cancer and include DNMTi, HDACi and HMTi. At present, there are two DNMTi, four HDACi ...
  130. [130]
    [PDF] Approval Letter(s) - accessdata.fda.gov
    Oct 6, 2006 · This new drug application provides for the use of Zolinza (vorinostat) Capsules for the treatment of cutaneous manifestations in patients with ...
  131. [131]
    Clinical efficacy and mechanistic insights of FDA-approved HDAC ...
    May 1, 2025 · Several HDAC inhibitors (HDACis) have demonstrated significant anticancer effects, with four FDA-approved molecules—vorinostat, romidepsin, ...
  132. [132]
    Epigenetic reprogramming reverses the relapse-specific gene ... - NIH
    In the present study, we demonstrate that the histone deacetylase inhibitor vorinostat not only reprograms the aberrant gene expression profile of relapsed ...
  133. [133]
    Concise Drug Review: Azacitidine and Decitabine - PMC - NIH
    Originally, they were intended as cytotoxic drugs. However, it was discovered that a low dose of these drugs could cause DNA demethylation by inactivation of ...Introduction · Table 2 · Mechanism Of Action
  134. [134]
    FDA approves oral combination of decitabine and cedazuridine for ...
    Jul 7, 2020 · FDA approves oral combination of decitabine and cedazuridine for myelodysplastic syndromes. On July 7, 2020, the Food and Drug Administration ...
  135. [135]
    FDA approves tazemetostat for advanced epithelioid sarcoma
    Jan 24, 2020 · On January 23, 2020, the Food and Drug Administration granted accelerated approval to tazemetostat (TAZVERIK, Epizyme, Inc.) for adults and ...Missing: EZH2 inhibitor
  136. [136]
    Epigenetics-targeted drugs: current paradigms and future challenges
    Nov 26, 2024 · The roles of histone methylation and acetylation in tumor progression have been extensively explored, with numerous reviews summarizing ...
  137. [137]
    Combining Epigenetic and Immunotherapy to Combat Cancer
    Current clinical trials are testing this combination therapy as a potent new cancer management strategy. Cancer Res; 76(7); 1683–9. ©2016 AACR. Introduction.
  138. [138]
    A New Trend in Cancer Treatment: The Combination of Epigenetics ...
    In this review, we summarize the current understanding of the key mechanisms of how epigenetic mechanisms affect cancer immune responses.
  139. [139]
    Engineered nanotechnology for epigenetic therapy in cancer treatment
    ### Summary of Epigenetic Drugs in Phase 3 Clinical Trials
  140. [140]
    CRISPR/Cas9-based engineering of the epigenome - PMC - NIH
    This review discusses the unprecedented opportunity that CRISPR/(d)Cas9 technology offers for investigating and manipulating the epigenome.
  141. [141]
    Programmable epigenome editing by transient delivery of CRISPR ...
    Aug 26, 2025 · We next applied RENDER to deliver the TET1-dCas9 (v4) epigenome editor that can enzymatically remove repressive DNA methylation from target gene ...
  142. [142]
    Dissecting the epigenetic regulation of the fetal hemoglobin genes ...
    Jul 10, 2025 · Editing a γ-globin repressor binding site restores fetal hemoglobin synthesis and corrects the sickle cell disease phenotype . Sci Adv.
  143. [143]
    Epigenetic Regulation of β-Globin Genes and the Potential to ... - MDPI
    In β-thalassemia, elevated fetal γ-globin levels lower the imbalance between α- and non-α globin chains, reduce the formation of α-globin precipitates and thus ...2. Epigenetic Regulation Of... · 2.2. Histone Modifications · 3. Epigenome Editing
  144. [144]
    Direct Epigenetic Reprogramming of Human Somatic Cells into ...
    Oct 2, 2025 · Restoring insulin production through replacement of pancreatic β-cells presents a promising strategy for treating individuals with type 1 ...
  145. [145]
    Toward the Development of Epigenome Editing-Based Therapeutics
    Several challenges in epigenome editing have been discussed: improving target specificity, selecting optimal cell types for epigenome editing, avoiding ...
  146. [146]
    Epigenetic noise: Unappreciated process helps cells change identity
    Aug 20, 2025 · “The moral of the story is that sometimes the random background noise can be just as important as the signal.” The study, “Thymic epithelial ...
  147. [147]
    DNA methylation and machine learning - Clinical Epigenetics
    Oct 10, 2025 · Despite these breakthroughs, single-cell DNA methylation profiling faces significant challenges, including high technical noise due to low input ...
  148. [148]
    Epigenome editing based treatment: Progresses and challenges
    Sep 1, 2025 · The ongoing transition of epigenome editing techniques from foundational research to clinical applications highlights several key strategies.
  149. [149]
    A critical view on transgenerational epigenetic inheritance in humans
    Jul 30, 2018 · The factors include endocrine disruptors, high fat diet, obesity, diabetes, undernourishment as well as trauma. These studies investigated DNA ...
  150. [150]
    Transgenerational epigenetic inheritance: a critical perspective
    Aug 29, 2024 · Transgenerational epigenetic inheritance in humans and other vertebrates has been controversial for over 150 years and remains so.
  151. [151]
    Artificial intelligence for comprehensive DNA methylation analysis
    Sep 15, 2025 · The PRMxAI model demonstrates remarkable performance, achieving 87.17% accuracy on mono-methylarginine datasets and 90.40% accuracy on di- ...
  152. [152]
    To live or let die? Epigenetic adaptations to climate change—a review
    Another key feature of epigenetic mechanisms is their reversibility, which can lead to both short- or long-term phenotypic plasticity through modulation of gene ...
  153. [153]
    (PDF) Environmental Epigenetics 2025 update - ResearchGate
    Aug 20, 2025 · PDF | On Feb 18, 2025, Michael K Skinner published Environmental Epigenetics 2025 update | Find, read and cite all the research you need on ...
  154. [154]
    Epigenetics and the exposome: A new era of disease prevention
    Those fingerprints act as a record of past exposures, from air pollution and lead to diet and exercise, providing vital clues about disease risk and even ...Missing: consensus projects
  155. [155]
    Integration of genetic and epigenetic markers for risk stratification - NIH
    As combining polygenic risk with environmental risk factors improves risk stratification, then integration of epigenetic markers and polygenic risk into a risk ...
  156. [156]
    Methylation risk scores are associated with a collection of ... - Nature
    Aug 25, 2022 · Tangentially, while polygenic risk scores provide a lifetime risk for a patient, methylation risk scores may provide the current risk of the ...
  157. [157]
    Is epigenome editing non-inheritable? Implications for ethics and the ...
    Nov 2, 2023 · Epigenome editing offers ethical advantages with non-inheritable gene expression control. However, concerns arise regarding potential transgenerational effects ...
  158. [158]
    Separating Fact from Fiction: The “Magic” of Epigenetics?
    May 8, 2015 · The current favorite in the world of irregular medicine appears to be epigenetics, which almost always amounts to magic or “The Secret” when they use the word.<|separator|>
  159. [159]
    The Latest In Epigenetics: How Your Environment And Genes Interact
    Currently, there are around 200 active clinical trials. Unfortunately, people have used epigenetics and the research results to promote pseudoscientific agendas ...