Fact-checked by Grok 2 weeks ago

Epigenomics

Epigenomics is the genome-wide study of epigenetic modifications—heritable changes in gene expression that do not alter the underlying DNA sequence—which regulate cellular function through mechanisms such as DNA methylation, histone modifications, and non-coding RNA activity. These modifications, including the addition of methyl groups to cytosine bases in CpG dinucleotides (primarily silencing gene promoters) and post-translational alterations to histone proteins (such as acetylation to promote transcription or methylation to repress it), enable dynamic control of gene activity in response to environmental cues like diet, stress, or toxins. Emerging as a field in the early 2000s with advances in high-throughput sequencing, epigenomics builds on classical epigenetics—coined by Conrad Waddington in 1942—to map these processes across entire genomes, revealing cell-type-specific patterns essential for development, differentiation, and homeostasis. The significance of epigenomics lies in its role bridging and environmental influences, with applications spanning normal and . In development, epigenetic marks establish identity; for instance, differential patterns distinguish embryonic cells from differentiated lineages. In disease, aberrations such as hypermethylation of tumor suppressor genes contribute to cancers like and colorectal , while hypomethylation at retrotransposons drives genomic instability. Epigenomic profiling has also illuminated aging-related disorders through "epigenetic clocks" like Horvath's, which predict biological age via methylation at ~353 CpG sites, and cardiovascular conditions via associations at Alu repeat elements. Recent advances, fueled by technologies like single-cell for accessibility and (e.g., Nanopore for base-resolution ), have enabled large-scale projects such as the Epigenomics , which has mapped reference epigenomes for over 100 types and tissues. Therapeutically, FDA-approved demethylating agents like treat myelodysplastic syndromes by reversing aberrant silencing, while CRISPR-based epigenome editors offer precise, heritable modulation without DNA cuts—paving the way for targeted interventions in complex diseases. These developments underscore epigenomics' potential to transform diagnostics, risk prediction, and , particularly in and .

Overview

Definition and Scope

Epigenomics is the study of the epigenome, defined as the complete set of chemical modifications to DNA and associated proteins that regulate gene expression without altering the underlying DNA sequence. These modifications, collectively known as epigenetic marks, include processes such as DNA methylation and histone modifications, which form a dynamic layer atop the fixed genome to control which genes are active in specific cells or conditions. Unlike the genome, which represents the static DNA blueprint inherited from parents, the epigenome is modifiable and responsive, enabling cells with identical genetic material to exhibit diverse functions and phenotypes. The scope of epigenomics encompasses the genome-wide analysis of these heritable yet reversible changes that influence cellular identity, , and to environmental cues. For instance, epigenetic marks help establish and maintain tissue-specific patterns, allowing a to differ functionally from a despite sharing the same . This field also examines how external factors, such as , , or toxins, can induce epigenomic alterations that persist across divisions or even generations, contributing to . Epigenomics holds profound biological significance by elucidating how variations in epigenetic landscapes drive diversity in gene regulation across organisms, tissues, and physiological states. These variations are crucial for processes like embryonic development, where epigenomic ensures proper , and for responses to aging or environmental stress, where dysregulated marks can lead to altered activity. By bridging and environmental influences, epigenomics provides insights into the mechanisms underlying cellular adaptability and the origins of and diseases.

Historical Development

The concept of epigenetics was first introduced by British embryologist Conrad Hal Waddington in 1942, who coined the term to describe the interplay between genetic factors and environmental influences in embryonic development, envisioning it as a bridge between genotype and phenotype. Waddington's framework emphasized dynamic processes that canalize developmental pathways, laying the groundwork for understanding heritable changes beyond DNA sequence alterations. In the and , research shifted toward molecular mechanisms, with a particular focus on as a key epigenetic modifier. Robin Holliday and Jeffrey E. Pugh proposed in 1975 that DNA modification, specifically methylation, could serve as a stable mechanism for regulating gene activity during development and . Concurrently, Arthur D. Riggs advanced this idea by linking DNA methylation to X-chromosome inactivation and the maintenance of differentiated states, suggesting that methylation patterns could propagate through cell divisions to enforce epigenetic memory. The 1990s marked the resurgence of in , driven by discoveries of and the role of in parent-of-origin-specific . This period solidified as a field integrating with environmental responsiveness. By the , the discipline expanded to genome-wide scales, exemplified by the launch of the Human Epigenome Project in 2003, an international effort to map profiles across human cell types and tissues. The project, initiated the same year, further propelled epigenomics by generating comprehensive maps of states, modifications, and regulatory elements, revealing the functional architecture of the human epigenome. Post-2010 advancements in next-generation sequencing (NGS) technologies revolutionized epigenomics, enabling high-throughput profiling of epigenetic marks across entire genomes at unprecedented resolution and scale. In the , integration with single-cell techniques has allowed for dissecting epigenetic heterogeneity within tissues, uncovering cell-type-specific modifications and dynamic responses in development and disease. Influential contributions include Andrew P. Feinberg's 1983 demonstration of global DNA hypomethylation in human cancers, which highlighted ' role in tumorigenesis and spurred oncology-focused research. Additionally, the 2006 in Physiology or Medicine awarded to Andrew Z. Fire and Craig C. Mello for discovering underscored non-coding RNAs' epigenetic regulatory functions, bridging posttranscriptional silencing with chromatin-level control.

Epigenetic Mechanisms

DNA Methylation

is a fundamental epigenetic modification involving the covalent addition of a to the fifth carbon of bases, primarily forming (5mC) at CpG dinucleotides in mammalian genomes. This process is catalyzed by a family of DNA methyltransferases (DNMTs), including , which maintains methylation patterns during by recognizing hemimethylated CpG sites, and de novo methyltransferases DNMT3A and DNMT3B, which establish new methylation marks on previously unmethylated DNA. DNMT3L acts as a regulatory subunit that enhances the activity of DNMT3A and DNMT3B without catalytic function itself. These enzymes transfer a from S-adenosylmethionine (SAM) to , resulting in stable epigenetic marks that can be inherited through cell divisions. Genome-wide, DNA methylation exhibits distinct patterns that correlate with regulation and structure. In mammalian somatic cells, approximately 70-80% of CpG sites are methylated, with hypermethylation typically occurring at promoters and repetitive to repress transcription, while hypomethylation is enriched in active bodies and intergenic regions. CpG islands—unmethylated, CpG-rich regions often located at promoters—remain largely protected from methylation to facilitate , whereas tissue-specific methylation profiles emerge during development, influencing cellular identity. These patterns are not uniform; for instance, global hypomethylation occurs in early embryos and primordial germ cells, followed by waves of methylation to establish lineage-specific epigenomes. Biologically, DNA methylation plays critical roles in genomic stability and developmental processes. It is essential for , where parent-of-origin-specific silences one of imprinted genes, ensuring monoallelic expression in offspring. In females, facilitates X-chromosome inactivation by contributing to the stable silencing of genes on the inactive through promoter hypermethylation, while the promoter is hypomethylated to enable its expression. Additionally, suppresses transposable elements, preventing their mobilization and maintaining genome integrity, particularly in germ cells and early embryos. During development, dynamic changes orchestrate cell differentiation; for example, global demethylation in zygotes erases parental imprints, enabling totipotency, while subsequent remethylation establishes somatic patterns. A key variant of 5mC is 5-hydroxymethylcytosine (5hmC), generated by ten-eleven translocation (TET) enzymes through oxidation of 5mC, serving as an intermediate in active demethylation pathways. Unlike 5mC, 5hmC is enriched in gene bodies of actively transcribed genes and is particularly abundant in postmitotic neurons, where it promotes terminal differentiation and modulates neuronal gene expression without leading to full demethylation. This modification adds a layer of regulatory complexity, potentially influencing DNA methylation's interplay with histone modifications in chromatin organization.

Histone Modifications

Histones are small, basic proteins that package DNA into nucleosomes, the fundamental units of chromatin, consisting of an octamer formed by two copies each of the core histones H2A, H2B, H3, and H4 around which approximately 147 base pairs of DNA are wrapped in 1.65 left-handed superhelical turns. These nucleosomes further compact into higher-order chromatin structures, influencing DNA accessibility and gene expression. The amino-terminal tails of histones protrude from the nucleosome core and are subject to diverse post-translational modifications (PTMs), which dynamically regulate chromatin architecture and transcriptional states. Common histone PTMs include acetylation, methylation, phosphorylation, and ubiquitination, primarily occurring on , , serine, and other residues within the histone tails. neutralizes the positive charge of residues, reducing the affinity between histones and negatively charged DNA, thereby promoting an open conformation conducive to transcription; this process is catalyzed by histone acetyltransferases (HATs) such as Gcn5 and reversed by histone deacetylases (HDACs). , which can occur on or residues in mono-, di-, or tri-methylated forms, has context-dependent effects: for instance, H3K4 trimethylation () marks active transcription start sites, while H3K9 or H3K27 trimethylation ( or ) is associated with repression. Histone methyltransferases (HMTs), such as SUV39H1 for and EZH2 (the catalytic subunit of Polycomb repressive complex 2, or PRC2) for , add methyl groups using S-adenosylmethionine as a cofactor. adds a negatively charged group, often on serine or , altering interactions; ubiquitination, a small ubiquitin-like modifier addition, typically influences other PTMs or protein recruitment. These modifications are reversible and balanced by opposing enzymes, ensuring precise control over gene activity. The code hypothesis posits that the combinatorial patterns of these PTMs on tails encode specific signals recognized by effector proteins, extending the information potential of the to regulate function. For example, at promoters recruits readers containing or domains, facilitating transcription initiation, while recruits Polycomb group proteins via chromodomains to maintain repression. , found in proteins like , specifically bind acetylated lysines, stabilizing open and promoting elongation by . This "code" allows for nuanced, heritable without altering the DNA sequence. Genome-wide, histone modifications exhibit distinct distribution patterns that correlate with functional genomic elements. and are enriched at active promoters and enhancers, marking regions of high transcriptional output, whereas predominates at poised or repressed loci, often in developmental genes. is broadly distributed in constitutive , contributing to centromeric . These marks are dynamic, responding to cellular contexts; for instance, phosphorylation of H3 at serine 10 (H3S10ph) surges during , coinciding with chromosome condensation and spreading from pericentromeric regions, which temporarily displaces repressive readers like HP1 to allow mitotic progression. Enzyme regulation fine-tunes these modifications for context-specific effects. , for example, is recruited by Polycomb group proteins to target genes, catalyzing to enforce repression in stem cells and during development, with its activity modulated by cofactors like SUZ12 and EED in the PRC2 complex. Similarly, HMTs like SET1/MLL complexes deposit at active promoters in a transcription-coupled manner. Histone modifications often cooperate with in , where recruits DNA methyltransferases to perpetuate heterochromatic states.

Non-coding RNAs

Non-coding RNAs (ncRNAs) play a pivotal role in epigenomic regulation by guiding chromatin-modifying complexes to specific genomic loci, thereby influencing gene silencing without altering the DNA sequence. These molecules encompass diverse classes, including long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and PIWI-interacting RNAs (piRNAs), each contributing uniquely to the establishment and maintenance of epigenetic landscapes. LncRNAs, typically longer than 200 nucleotides, often act as scaffolds or recruiters for epigenetic effectors, while miRNAs and piRNAs primarily modulate post-transcriptional control and transposon repression, respectively. LncRNAs exemplify RNA-guided epigenetic programming through their interactions with chromatin modifiers, such as in X-chromosome inactivation where the lncRNA coats the inactive to recruit silencing complexes, leading to widespread gene repression. Another prominent example is HOTAIR, a lncRNA that scaffolds the Polycomb Repressive Complex 2 (PRC2) to facilitate trimethylation of at lysine 27 (), thereby promoting formation at target loci. These mechanisms highlight how lncRNAs bridge sequences with protein effectors to enforce stable epigenetic states, often in coordination with modifications. MicroRNAs (miRNAs), small ncRNAs approximately 22 in length, regulate epigenetic enzymes post-transcriptionally by binding to the 3' untranslated regions of target mRNAs, leading to their degradation or translational repression. For instance, miR-29 targets DNA methyltransferases (DNMTs), reducing activity and altering global epigenomic patterns. Similarly, miR-148 regulates DNMT3b, influencing methylation during development. This feedback loop between miRNAs and epigenetic machinery underscores their role in fine-tuning accessibility across cell types. PIWI-interacting RNAs (piRNAs), 24-31 nucleotides long, are essential for transposon silencing in the germline, where they direct de novo DNA methylation to suppress retrotransposon activity and maintain genomic stability. In male mammals, piRNAs bound to MIWI2 guide the DNMT3A/DNMT3L complex to transposon sequences during prospermatogonia, establishing methylation patterns that persist through spermatogenesis. This piRNA-directed methylation prevents transposon mobilization, which could otherwise disrupt epigenetic integrity in offspring. At the genome-wide level, ncRNAs profoundly impact epigenomic states, as evidenced by lncRNA expression patterns that correlate with altered landscapes in cancer, where dysregulated lncRNAs like HOTAIR drive aberrant deposition across tumor suppressor loci. PiRNAs similarly enforce methylation hotspots in the , safeguarding against heritable mutations. These broad effects illustrate ncRNAs' capacity to orchestrate large-scale epigenetic reprogramming. Emerging research in the 2020s has revealed additional layers, such as circular RNAs (circRNAs)—covalently closed ncRNA forms—that influence acetylation by acting as decoys for acetyltransferases or recruiting modifiers to enhancers, thereby modulating gene activation in dynamic cellular contexts. Furthermore, ncRNAs contribute to , with sperm-borne piRNAs and lncRNAs transmitting patterns across generations in response to environmental cues, as observed in mouse models of stress exposure. These insights expand the scope of ncRNA functions beyond immediate regulation to heritable epigenomic memory.

Integration with Other Omics

Relation to Genomics

Epigenomics and genomics both pertain to the study of the genome but differ fundamentally in their focus and scope. Genomics primarily investigates the static DNA sequence, including variations such as single nucleotide polymorphisms (SNPs) and copy number variations (CNVs), which form the genetic blueprint inherited across generations. In contrast, epigenomics examines dynamic, heritable modifications to the DNA or associated proteins—such as DNA methylation, histone modifications, and chromatin remodeling—that regulate gene expression without altering the underlying nucleotide sequence.00236-1) These epigenetic marks provide a layer of functional information atop the genomic sequence, enabling cells to respond to environmental cues and developmental signals. The fields are complementary, as epigenetic mechanisms help address gaps in genomic studies, particularly the "missing " observed in genome-wide studies (GWAS). GWAS often for only a fraction of due to their emphasis on sequence variants, leaving substantial unexplained variance that epigenetic factors may bridge through interactions with the . For instance, quantitative loci (mQTLs) represent genomic variants that influence levels, linking sequence differences to epigenetic states and thereby modulating in a tissue-specific manner. This integration reveals how epigenomic profiles can explain phenotypic variation beyond what alone predicts, such as in complex diseases. Overlaps between the epigenome and highlight bidirectional influences on genomic and function. Epigenetic modifications, particularly at CpG dinucleotides, can elevate rates by promoting spontaneous of to , resulting in C-to-T transitions that contribute to genetic in conditions like cancer. Conversely, shapes epigenetic mark distribution; for example, CpG islands—regions of high CpG density often located at gene promoters—tend to remain unmethylated, facilitating active transcription and protecting against aberrant silencing. These interactions underscore how the epigenome both responds to and modulates the genomic landscape. The integration of epigenomics with gained momentum following the project's 2012 findings, which mapped functional elements across the and demonstrated that much of the harbors regulatory activity through epigenetic signatures. revealed over 80% of the genome's involvement in biochemical processes, including enhancer and promoter regions identified via marks, expanding the understanding of regulatory elements far beyond protein-coding sequences. This work established epigenomics as essential for interpreting genomic data, particularly in identifying disease-associated variants in non-coding regions. Epigenomics interfaces with transcriptomics by linking stable modifications to dynamic outputs, where specific epigenetic marks serve as predictors of abundance and transcriptional activity. For instance, trimethylation of at 4 (H3K4me3) is prominently enriched at transcription start sites (TSSs) of actively transcribed genes, correlating positively with occupancy and nascent production across diverse cell types. This association enables epigenomic profiling, such as ChIP-seq for , to forecast levels. Integrating epigenomic data with further uncovers regulatory feedback loops, such as how transcription factors bind modified histones to reinforce or attenuate synthesis in response to cellular signals. In relation to proteomics, epigenomic alterations influence protein recruitment and overall proteome composition by modulating chromatin accessibility and post-transcriptional processes. Histone post-translational modifications (PTMs), including and , recruit specific "reader" proteins—such as bromodomain-containing complexes for acetylated lysines—that facilitate the assembly of transcriptional machinery or alter positioning to expose DNA elements. These changes extend to proteome diversity through chromatin's role in regulating , where epigenetic marks like H3K36me3 near exon-intron boundaries promote recruitment, leading to isoform-specific protein variants that enhance functional complexity in eukaryotic cells. For example, patterns at splice sites can suppress non-canonical splicing events, thereby constraining proteome variability in differentiated tissues. Multi-omics integrations highlight epigenomics as a foundational layer bridging to epitranscriptomics and beyond, with RNA modifications emerging as a parallel regulatory mechanism akin to chromatin-based control. Epitranscriptomic marks, such as N6-methyladenosine (m6A) on mRNAs, often co-occur with epigenetic signatures to fine-tune translation efficiency, forming an extended network that influences both transcript stability and protein output. In the 2020s, spatial multi-omics technologies have advanced this connectivity by mapping epigenome-transcriptome correlations within intact tissues; for instance, methods like spatial ATAC–RNA-seq enable simultaneous profiling of chromatin accessibility and polyA+ RNA in mouse brain sections, revealing cell-type-specific regulatory hubs that correlate H3K27ac peaks with localized gene expression gradients. Similarly, recent assays combining DNA methylation with spatial transcriptomics in mouse tissues have quantified how epigenomic heterogeneity drives tissue-specific transcriptome landscapes. Challenges in these integrations arise from the disparate temporal dynamics of molecular layers, where epigenetic marks persist over divisions while and protein half-lives range from minutes to days, complicating causal inferences in multi-omics datasets. This variability necessitates advanced computational , such as batch-effect correction in joint epigenomic-proteomic analyses, to align stable states with profiles without introducing biases. Despite these hurdles, such approaches have illuminated how epigenomic perturbations propagate through transcriptomic and proteomic networks to maintain cellular .

Experimental Methods

DNA Methylation Assays

DNA methylation assays are essential tools for profiling the addition of methyl groups to bases, primarily at CpG dinucleotides, across the . These techniques enable researchers to map methylation patterns at single-base resolution or targeted sites, providing insights into and epigenetic states. The most widely adopted methods rely on chemical conversion, enzymatic digestion, or hybridization arrays, each balancing coverage, cost, and resolution. Bisulfite sequencing stands as the gold standard for DNA methylation analysis due to its ability to achieve base-specific detection. The process involves treating genomic DNA with , which deaminates unmethylated cytosines to uracils (read as thymines during sequencing), while leaving methylated cytosines (, 5mC) unchanged, allowing differentiation through subsequent sequencing. This method was first described in 1992 and has since become foundational for epigenomic studies. A key variant, whole-genome (WGBS), extends this approach to provide comprehensive, unbiased coverage of the entire methylome by sequencing the bisulfite-converted genome at high depth, as demonstrated in early applications to model organisms like . To address the high cost and data volume of WGBS, restriction enzyme-based methods enrich for methylation-relevant regions prior to bisulfite conversion. Reduced representation (RRBS) employs the methylation-insensitive enzyme MspI, which cleaves at CCGG sites regardless of status, followed by size selection of short fragments that disproportionately represent CpG-rich areas such as promoters and enhancers. Complementary approaches use methylation-sensitive restriction enzymes like HpaII, which cuts only unmethylated CCGG sites, in contrast to MspI, enabling differential assessment of levels in these motifs before or alongside treatment. These strategies reduce sequencing requirements while focusing on biologically significant loci. Array-based assays offer a cost-effective for high-throughput studies in large cohorts, interrogating predefined CpG sites via hybridization. The Infinium MethylationEPIC v2.0 BeadChip from Illumina (as of 2024) targets over 935,000 CpG sites, including enhancers and other regulatory elements, using two-color to quantify levels at each probe. This has facilitated extensive epigenome-wide studies (EWAS) due to its reproducibility and scalability. Despite their utility, DNA methylation assays face inherent limitations that can influence data interpretation. Bisulfite-based methods, including WGBS and RRBS, exhibit biases toward CpG-rich regions in the latter due to enzymatic enrichment, potentially underrepresenting sparsely methylated areas like gene bodies or intergenic spaces. Additionally, standard bisulfite conversion cannot distinguish 5mC from (5hmC), an oxidized derivative associated with active demethylation; specialized protocols like Tet-assisted bisulfite sequencing (TAB-seq), which uses TET enzymes to protect 5hmC before conversion, are required for specific detection. Bisulfite treatment also causes significant DNA fragmentation and incomplete conversion, reducing input efficiency and introducing potential artifacts.

Histone Modification Profiling

Histone modification profiling encompasses a suite of techniques designed to map the genomic distribution of post-translational modifications on proteins, which play crucial roles in regulating structure and . These methods predominantly employ immunoprecipitation-based strategies, leveraging antibodies to selectively enrich chromatin fragments bearing specific modifications, such as or methylation on histone tails. By identifying enrichment patterns, researchers can delineate functional chromatin states, from active promoters marked by to repressive domains via H3K27me3. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) represents the cornerstone of modern histone modification profiling, offering genome-wide insights with high sensitivity and resolution. The process begins with crosslinking cells to preserve protein-DNA interactions, followed by chromatin fragmentation via ; specific antibodies, such as those targeting H3K27ac for enhancer , are then used to immunoprecipitate modified nucleosomes. Enriched DNA fragments are sequenced, and bioinformatics tools perform peak calling to quantify enrichment over input controls, revealing localized hotspots or broad domains. This approach was pioneered in a 2007 study that generated high-resolution maps of 20 histone methylations in CD4+ T cells, demonstrating their association with transcriptional activity and silencing. ChIP-seq has since become indispensable for dissecting dynamic epigenetic landscapes in diverse biological contexts. An earlier iteration, chromatin immunoprecipitation followed by microarray analysis (ChIP-chip), laid the groundwork for genome-scale histone studies before the advent of next-generation sequencing. In ChIP-chip, immunoprecipitated DNA is hybridized to oligonucleotide microarrays tiling promoters or entire genomes, enabling detection of modification-enriched regions through comparative signal intensities. This method excelled in targeted analyses of specific loci but suffered from lower resolution and array design limitations, prompting its replacement by ChIP-seq for comprehensive profiling. Seminal applications included mapping histone acetylation and methylation across the yeast genome, which uncovered periodic patterns tied to transcription units. To overcome ChIP-seq's drawbacks, including the need for millions of cells and sonication-induced biases, advanced variants like CUT&RUN and CUT&TAG have emerged as efficient alternatives for low-input epigenomic mapping. CUT&RUN, introduced in 2017, immobilizes intact nuclei on magnetic beads and tethers protein A- (pA-MNase) to the target , enabling precise DNA cleavage near histone modifications and direct release of fragments for sequencing; this yields high signal-to-noise ratios from as few as 100 cells, minimizing artifacts from chromatin shearing. Building on this, CUT&TAG (2019) replaces MNase with protein A-Tn5 , which performs targeted tagmentation—simultaneous cleavage and adapter insertion—in , further reducing workflow steps and background while supporting single-cell applications for marks. Both techniques enhance accessibility for rare samples, such as primary tissues, by preserving native context. These profiling methods deliver base-pair , facilitating the characterization of both sharp peaks (e.g., at active enhancers) and expansive domains, such as H3K9me3-marked stretches that span kilobases to megabases and enforce stable gene repression. In applications, ChIP-seq and its variants have illuminated H3K9me3's role in maintaining pericentromeric silencing and developmental barriers, with broad domains often validated across cell types to link modifications to phenotypic outcomes. Nonetheless, specificity remains a key challenge, as polyclonal reagents may cross-react with structurally similar epitopes (e.g., mono- vs. tri-methylation), inflating false positives and requiring orthogonal validation like or engineered controls to ensure accuracy. Ongoing efforts focus on monoclonal and epitope-tagging to mitigate these issues.

Chromatin Accessibility Assays

Chromatin accessibility assays probe the openness of structure across the genome, identifying regions susceptible to enzymatic digestion or , which often correspond to active regulatory elements such as promoters and enhancers. These methods reveal the landscape of accessible DNA without targeting specific epigenetic modifications, providing insights into potential regulatory activity in various cell types and conditions. By mapping hypersensitive sites or nucleosome-depleted regions, they highlight areas where transcription factors and other proteins can bind more readily, influencing patterns in epigenomic studies. DNase-seq, one of the earliest genome-wide chromatin accessibility assays, employs DNase I nuclease to selectively cleave open chromatin regions, followed by high-throughput sequencing of the resulting DNA fragments to identify DNase hypersensitive sites (DHSs). This technique was first applied genome-wide in 2008 using primary human CD4+ T cells, enabling the detection of regulatory elements with high resolution. A landmark application in 2012 generated the first comprehensive map of DHSs across 125 diverse human cell and tissue types, demonstrating that accessible chromatin regions are highly cell-type specific and enriched for transcription factor binding motifs. DNase-seq has been instrumental in annotating millions of potential regulatory elements, though it requires substantial input material (typically millions of cells) and can introduce biases from DNase I sequence preferences. ATAC-seq (Assay for Transposase-Accessible with sequencing) offers a faster and more sensitive alternative, utilizing hyperactive Tn5 transposase to insert sequencing adapters directly into accessible regions in native nuclei, followed by amplification and sequencing. Introduced in 2013, this method requires only 500–50,000 cells, making it suitable for limited samples, and provides nucleotide-resolution mapping of open , positioning, and footprints in a single . has revolutionized epigenomic profiling by reducing preparation time to under three hours and enabling the identification of regulatory elements with high concordance to DNase-seq results, while also capturing additional information on compaction. MNase-seq (Micrococcal Nuclease sequencing) focuses on positioning by digesting with micrococcal , which preferentially cleaves between , followed by sequencing of protected mononucleosomal DNA fragments to map the +1 at transcription start sites and linker regions. Pioneered in a genome-wide study of human CD4+ T cells in , this assay reveals periodic arrays and positions where is more or less accessible based on protection levels. MNase-seq provides precise boundaries for cores (approximately 147 ) and has been used to uncover dynamic changes in occupancy during cellular activation, though it can exhibit digestion biases at certain sequence motifs. Advancements in single-cell resolution, particularly single-cell (scATAC-seq), extend these assays to heterogeneous populations, allowing profiling of in individual cells since its in 2015. scATAC-seq adapts the Tn5-based approach to isolate and sequence accessible regions from thousands of single nuclei, revealing cell-type-specific regulatory landscapes and epigenetic heterogeneity in processes like and . Post-2015 improvements, including droplet-based and combinatorial indexing strategies, have scaled scATAC-seq to profile over 100,000 cells, enhancing its utility in epigenomic atlases. Recent extensions include spatial ATAC-seq variants for mapping in tissue contexts (as of 2024). To incorporate three-dimensional chromatin organization, chromatin accessibility assays are often integrated with , a technique that maps long-range interactions, thereby linking open regions to distal looping events. For instance, combining with Hi-C identifies enhancer-promoter contacts within accessible domains, providing a more complete view of regulatory networks. Chromatin accessibility often correlates positively with histone acetylation marks, such as H3K27ac, indicating active enhancers, though accessibility assays capture broader structural openness independent of specific modifications.
AssayKey Enzyme/MechanismInput RequirementResolutionSeminal Reference
DNase-seqDNase I cleavage of open Millions of cellsDHSs at ~1-10 kbBoyle et al. (2008)
Tn5 insertion500–50,000 cellsNucleotide-levelBuenrostro et al. (2013)
Micrococcal digestion of linkersMillions of cells~147 bp nucleosomesSchones et al. (2008)
scATAC-seqSingle-cell Tn5 Single cells (thousands profiled)Cell-type specificBuenrostro et al. (2015)

Direct Detection Techniques

Direct detection techniques in epigenomics enable the identification of base modifications in native molecules without chemical conversion or amplification, providing unbiased and high-resolution insights into epigenetic states. These methods leverage single-molecule technologies to capture kinetic or electrical signals associated with modifications such as (5mC) and (5hmC), offering advantages over traditional bisulfite-based assays by preserving sequence context and distinguishing subtle variants. Single-molecule real-time (SMRT) sequencing, developed by (PacBio), detects DNA modifications through alterations in the kinetics of during incorporation. In this approach, modified bases like 5mC induce changes in interpulse duration and pulse width, reflecting slowed incorporation rates that span several bases in a context-dependent manner. The seminal demonstration of this applied it to bacterial genomes, achieving single-molecule for N6-methyladenosine and 5mC with up to 85% at low false positive rates using circular consensus sequencing. Oxford Nanopore Technologies' nanopore sequencing measures ionic current disruptions as DNA or translocates through a protein , allowing direct readout of native modifications without pretreatment. Modifications such as 5mC cause distinct signal deviations, enabling base-level calling with tools like Nanopolish, which analyzes raw fast5 signal data for probabilistic modification scoring. Post-2020 advancements have improved accuracy to over 99% for base calling on updated flow cells (e.g., R10.4), and expanded detection to more than 12 endogenous marks, including m6A in , through integrations like DeepSignal. Optical mapping utilizes nanochannel arrays to linearize and image high-molecular-weight DNA molecules, facilitating long-range epigenomic profiling without sequencing. In one approach, 5hmC is selectively labeled with fluorophores (e.g., via enzymatic protection and ) alongside genetic motifs, allowing to generate maps spanning 100 kbp to 1 Mbp with 82% labeling efficiency and high signal-to-noise ratios. This method has been applied to cells, detecting 5hmC densities as low as 0.0029% in regions like the HLA locus, where it outperforms enrichment-based techniques for variable genomic contexts. These techniques collectively address limitations of conversion-based methods by avoiding DNA degradation and bias, enabling native distinction of 5mC from 5hmC, and supporting long-read applications that resolve structural variants in repetitive epigenomic landscapes. Recent progress includes enhancements for SMRT achieving 99% concordance with orthogonal validation and tools for quantitative , broadening their use in complex eukaryotic epigenomes.

Computational and Modeling Approaches

Bioinformatics for Epigenomic Data

Bioinformatics plays a crucial role in epigenomics by enabling the processing, analysis, and interpretation of high-throughput data from assays like , ChIP-seq, and , which generate vast amounts of sequence reads requiring alignment, quality control, and statistical modeling to identify meaningful epigenetic patterns. These pipelines address the complexity of epigenomic datasets, which often involve sparse signals and technical variability, ensuring reproducible results through standardized computational workflows. Data processing begins with read alignment, particularly challenging for bisulfite-treated DNA due to symmetric cytosine-to-thymine conversions; the BWA-meth tool, an extension of the Burrows-Wheeler Aligner (BWA), efficiently maps these reads by supporting insertions, deletions, and clipping while maintaining high accuracy for long sequences. For ChIP-seq data profiling modifications or binding, peak calling identifies enriched genomic regions using Model-based Analysis of ChIP-Seq (MACS2), which models background noise and fragment sizes to detect peaks with improved sensitivity over earlier versions, especially for narrow or broad signals. Differential analysis, such as comparing methylation levels across conditions, employs tools like edgeR, which adapts negative binomial models originally for to handle count-based methylation data from reduced representation , enabling robust detection of differentially methylated regions while accounting for biological variability. Integration of epigenomic data with other omics layers is facilitated by frameworks like Multi-Omics Factor Analysis (MOFA), an method that decomposes multi-omic datasets into latent factors to uncover shared variation, such as joint epigenome-transcriptome patterns in disease contexts. tools enhance interpretation; the Integrative Genomics Viewer (IGV) supports real-time browsing of aligned reads and tracks across scales, while the WashU Epigenome Browser offers interactive multi-track views tailored for epigenomic comparisons, including long-range interactions. Machine learning approaches, particularly models from the 2020s, have advanced epigenomic analysis by predicting epigenetic marks directly from DNA sequence; for instance, ChromBPNet uses convolutional neural networks to model base-resolution accessibility, deconvolving assay biases to reveal footprints and regulatory variants. Imputation of missing data, common in sparse epigenomic profiles, benefits from methods like eDICE, which employs attention mechanisms to infer unobserved tracks from available ones, improving accuracy in cross-cell type predictions as demonstrated in benchmarks. Key challenges include mitigating batch effects, which introduce technical artifacts across experiments and can confound biological signals in large-scale studies, addressed through normalization strategies in tools like MOFA. Single-cell epigenomic data exacerbates sparsity, where low coverage limits per-cell resolution, requiring imputation or aggregation methods while preserving heterogeneity. Standards from the project guide these processes, recommending uniform pipelines for alignment, quality metrics (e.g., FRiP scores for ChIP-seq), and data deposition to ensure interoperability and reproducibility.

Theoretical Modeling

Theoretical modeling in epigenomics employs mathematical frameworks and computational simulations to elucidate the dynamic through chromatin structure and modifications, without relying on empirical data processing tools. These approaches predict probabilistic outcomes of epigenetic processes, such as chromatin looping and modification propagation, by integrating principles from , dynamical systems, and processes. Such models facilitate hypothesis generation regarding the stability and variability of epigenetic states across cell cycles and populations. Polymer models represent as a semi-flexible , capturing its conformational and mechanical constraints to predict spatial interactions like enhancer-promoter looping. In the Gaussian approximation, suitable for flexible segments, the of end-to-end distance r between segments is given by P(r) \sim \exp\left(-\frac{3r^2}{2Nl^2}\right), where N denotes the number of segments and l the persistence length, reflecting 's semi-rigidity due to packaging. This model, extended to wormlike s for stiffer regions, quantifies looping probabilities under equilibrium conditions, showing how epigenetic marks like histone acetylation increase flexibility and enhance contact frequencies. For chromosomes, finite thickness further modulates these probabilities, reducing long-range interactions in compacted states. Epigenetic switching models describe the bistable propagation of modifications, such as , through maintenance mechanisms during replication. A basic for methylation level M (fraction of methylated sites) illustrates this as \frac{dM}{dt} = k_1 (1 - M) - k_2 M, where k_1 governs maintenance methylation by enzymes like and k_2 represents demethylation rates, leading to stable methylated or unmethylated states depending on parameter balances. arises in extended models incorporating cooperative enzyme binding, enabling epigenetic memory where silenced states persist despite dilution. These frameworks reveal thresholds for switching, influenced by histone interactions, and predict front propagation of silencing domains along . Network models formalize code interactions as regulatory networks, using logic for discrete states or ordinary equations (ODEs) for continuous dynamics, augmented by simulations to account for single- noise. networks toggle marks (e.g., for repression) based on rules like "if acetylated, then activate transcription," simulating combinatorial code readout. ODE-based variants, such as \frac{dH_i}{dt} = f(\sum w_{ij} H_j), where H_i is modification level i and w_{ij} interaction weights, capture feedback loops in Polycomb-Trithorax systems. extensions, via Gillespie algorithms, quantify variability in modification patterns, explaining heterogeneous epigenetic landscapes in populations. Recent advances in the 2020s include theoretical models exploring confinement mechanisms for epigenetic modifications of nucleosomes, which predict effects on modification spreading in nuclear microenvironments. As of 2025, further developments incorporate trans-dimensional approaches using hidden Markov models to uncover alterations in cancer via patterns. Integration with enhances predictive epigenomics by training models on simulated trajectories to forecast modification outcomes from initial states, improving accuracy in complex scenarios like cancer-associated rewiring.

Applications and Future Directions

Role in Development and Evolution

Epigenomic reprogramming occurs in waves during early mammalian embryonic , beginning with widespread in the shortly after fertilization, which erases most parental epigenetic marks to establish totipotency.00331-4) This initial demethylation phase, driven by active and passive mechanisms involving TET3-mediated oxidation and dilution during divisions, persists until the stage, allowing for the activation of the embryonic . Subsequently, de novo is re-established by enzymes such as DNMT3A and DNMT3B around implantation, progressively building lineage-specific epigenomic landscapes that support fate decisions. These reprogramming events ensure the transition from gametic to embryonic epigenomes, with incomplete erasure of certain imprints maintaining parent-of-origin-specific expression.00331-4) In embryonic stem cells (ESCs), bivalent chromatin domains—characterized by the coexistence of activating and repressive histone modifications—poise key developmental genes for activation during while keeping them silenced in the pluripotent state.00380-1) First identified in mouse ESCs, these domains are enriched at promoters of Hox and other lineage-specifying genes, enabling rapid transcriptional responses upon differentiation signals by resolving bivalency into active or repressed states. This epigenetic architecture supports the maintenance of pluripotency factors like Oct4 and Nanog, which counteract Polycomb repressive complex 2 (PRC2)-mediated to prevent premature lineage commitment. During , epigenomic memory mechanisms stabilize lineage commitments through persistent modifications, such as stable deposition by EZH2-containing PRC2 complexes, which represses alternative fate genes in committed cells like neurons. For instance, in neural progenitors differentiating into neurons, accumulates at and developmental regulators, enforcing stable repression and preventing . Single-cell epigenomics has further elucidated these processes by mapping dynamic trajectories of accessibility and marks, revealing branching paths where cells progressively lose pluripotency-associated open while gaining lineage-specific modifications. These trajectories highlight how epigenomic landscapes guide probabilistic fate choices, with tools like scATAC-seq and scChIP-seq uncovering intermediate states during mesodermal or neuroectodermal commitment. In evolutionary contexts, transgenerational epigenetics enables adaptive responses to environmental stressors, as evidenced by altered patterns persisting across generations in human cohorts exposed to . The Dutch Hunger Winter of 1944–1945 induced hypomethylation at the IGF2 differentially methylated region in periconceptionally exposed offspring, with effects detectable decades later and potentially transmitted to subsequent generations via gametic epigenomes. This methylation change correlates with altered growth regulation, illustrating how nutritional stress can imprint heritable epigenetic variants that influence metabolic traits without genetic mutations. Epigenetic mechanisms also facilitate adaptive plasticity in plants and animals facing environmental challenges, allowing rapid phenotypic adjustments that contribute to evolutionary fitness. In plants, stress-induced DNA methylation and histone variants enable transgenerational priming for drought or pathogen resistance, as seen in Arabidopsis where RdDM pathway-mediated silencing of transposable elements confers heritable tolerance. Similarly, in animals like Daphnia, predation cues trigger heritable chromatin remodeling that enhances defensive traits across generations. Post-2020 studies have linked these processes to climate adaptation, showing that ocean acidification in corals induces DNA methylation changes that improve calcification resilience in offspring, while in forest trees, temperature shifts alter CG methylation to modulate phenology and survival. Such epigenetic plasticity expands the evolvability of populations by buffering genetic constraints and accelerating adaptation to anthropogenic climate pressures.

Implications in Disease and Medicine

Epigenomic alterations play a central role in pathogenesis, particularly through aberrant and modifications that disrupt . In cancer, promoter hypermethylation frequently silences tumor suppressor genes, such as the O6-methylguanine-DNA methyltransferase (), leading to increased genomic instability and resistance in gliomas and other tumors. This epigenetic silencing of is observed across various human cancers and serves as a prognostic marker, associating with poorer outcomes while predicting responsiveness to alkylating agents like . In neurodegenerative disorders, mutations in variants like H3.3 contribute to dysregulation and neuronal dysfunction; for instance, mutations in H3F3A and H3F3B genes cause a rare pediatric neurodegenerative syndrome characterized by developmental delay, seizures, and progressive brain atrophy in affected individuals. Epigenomic profiling has advanced disease diagnostics by enabling non-invasive detection and risk assessment. The Horvath epigenetic clock, based on methylation levels at 353 CpG sites across diverse tissues, accurately predicts chronological age and serves as a biomarker for age-related diseases, with accelerated clock progression linked to increased cancer and cardiovascular risks. In oncology, cell-free DNA (cfDNA) methylation patterns from liquid biopsies offer sensitive biomarkers for early cancer detection; for example, specific methylation signatures in plasma cfDNA distinguish lung cancer patients from healthy controls with high accuracy, facilitating monitoring without invasive procedures. These approaches extend to other malignancies, such as breast and colorectal cancers, where cfDNA hypermethylation at tumor-specific loci enhances diagnostic specificity over traditional methods. Therapeutic strategies targeting the epigenome have gained clinical traction, focusing on reversing pathological modifications. Histone deacetylase (HDAC) inhibitors like , approved by the FDA in 2006 for (CTCL), promote histone acetylation to reactivate silenced genes and induce in cancer cells, demonstrating objective response rates of 24-30% in relapsed patients. DNA methyltransferase (DNMT) inhibitors, such as , are standard treatments for myelodysplastic syndromes and , where they hypomethylate DNA to restore tumor suppressor expression and improve survival in high-risk patients. Emerging CRISPR-based epigenome editing uses catalytically inactive Cas9 (dCas9) fused to epigenetic effectors, such as TET1 for demethylation or DNMT3A for methylation, to precisely modulate without DNA cleavage; post-2016 advancements have enabled targeted reversal of disease-associated marks in cellular models of cancer and neurological disorders. Looking toward 2025, personalized epigenome therapies hold promise for tailoring interventions based on individual profiles, potentially improving outcomes in precision and . As of 2025, advances in epigenome editing, including improved dCas9 specificity and applications in models, are progressing toward clinical trials. However, challenges persist, including off-target editing effects from dCas9 systems that could alter unintended genomic regions, necessitating enhanced specificity through improved guide RNAs and delivery vectors. Ongoing clinical trials aim to integrate epigenomic clocks with editing for patient stratification, though long-term safety and reversibility remain key hurdles for widespread adoption.

References

  1. [1]
    Epigenomic insights into common human disease pathology - PMC
    Apr 11, 2024 · This review will illustrate the current successes that have so far arisen though integrative analysis of the epigenome but also the immense ...
  2. [2]
    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.
  3. [3]
  4. [4]
    Epigenomics Fact Sheet
    Aug 16, 2020 · Epigenomics is a field in which researchers chart the locations and understand the functions of all the chemical tags that mark the genome.Missing: biological | Show results with:biological
  5. [5]
    Epigenome Mapping in Normal and Disease States - PubMed Central
    An epigenome can be defined as the combination of all genome-wide chromatin modifications in any given cell type that directs its unique gene expression pattern ...Missing: definition | Show results with:definition
  6. [6]
    Science Highlights | NIH Common Fund
    In 2008, the NIH launched an Epigenomics program, and one of its goals was mapping modifications across the entire genome of many different human cells and ...
  7. [7]
    Epigenomics | Learn Science at Scitable
    ### Summary of Epigenomics: The New Tool in Studying Complex Diseases
  8. [8]
    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 ...Missing: source | Show results with:source
  9. [9]
    Epigenotype | International Journal of Epidemiology | Oxford Academic
    Dec 20, 2011 · * Waddington CH. The Epigenotpye. Endeavour 1942; 18–20 Reprinted with permission. Published by Oxford University Press on behalf of the ...Missing: Conrad source
  10. [10]
    DNA modification mechanisms and gene activity during development
    This paper discusses DNA modification mechanisms and gene activity during development, including topics like DNA metabolism, genes, and methylation.
  11. [11]
    X inactivation, differentiation, and DNA methylation - PubMed
    A model based on DNA methylation is proposed to explain the initiation and maintenance of mammalian X inactivation and certain aspects of other permanent ...<|separator|>
  12. [12]
    Epigenetics: The origins and evolution of a fashionable topic
    Aug 1, 2016 · Following the discovery of imprinted genes in mice and men in the 1990s, epigenetics became closely associated with DNA methylation (Haig, 2011) ...
  13. [13]
    Human epigenome project--up and running - PubMed
    The Wellcome Trust Sanger Institute and Epigenomics AG recently announced the launch of the Human Epigenome Project - a 5-year project to map the sites of ...
  14. [14]
    ENCODE: A Sourcebook of Epigenomes and Chromatin Language
    The project gives us new insights into the organization and regulation of the human genome and epigenome. Here, we sought to summarize particular aspects of the ...Missing: epigenomics contributions
  15. [15]
    Next generation sequencing based approaches to epigenomics - PMC
    Next generation sequencing has brought epigenomic studies to the forefront of current research. The power of massively parallel sequencing coupled to ...Library Preparation · Histone Modification... · Bisulfite Conversion Based...
  16. [16]
    Advances and challenges in epigenomic single-cell sequencing ...
    Understanding multicellular physiology and pathobiology requires analysis of the relationship between genotype, chromatin organisation and phenotype.Missing: 2020s | Show results with:2020s
  17. [17]
    Hypomethylation distinguishes genes of some human cancers from ...
    Jan 6, 1983 · Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Andrew P. Feinberg &; Bert Vogelstein. Nature volume ...
  18. [18]
    DNA Methylation and Its Basic Function | Neuropsychopharmacology
    Jul 11, 2012 · DNA methylation regulates gene expression by recruiting proteins involved in gene repression or by inhibiting the binding of transcription factor(s) to DNA.Missing: epigenomics | Show results with:epigenomics<|control11|><|separator|>
  19. [19]
    Regulation of mammalian DNA methyltransferases: a route to new ...
    DNA methyltransferases (DNMTs) establish and maintain DNA methylation patterns at specific regions of the genome, thereby contributing to gene regulation.Missing: seminal | Show results with:seminal
  20. [20]
    DNA Methylation in Mammals - PMC - NIH
    CpG Islands and Patterns of DNA Methylation across the Genome. DNA across the genome of mammalian somatic tissues is methylated at 70%–80% of all CpG sites.
  21. [21]
    DNA methylation patterns and epigenetic memory
    ... CpG, which is the major target for methylation in animals. Most other invertebrate genomes have moderately high levels of methyl-CpG concentrated in large ...<|control11|><|separator|>
  22. [22]
    DNA Methylation: Superior or Subordinate in the Epigenetic ... - PMC
    In this review, we will comprehensively examine the current understanding of the connections between DNA methylation and other epigenetic marks and discuss ...Missing: seminal | Show results with:seminal
  23. [23]
    Mechanisms of human DNA methylation, alteration of methylation ...
    Jul 30, 2023 · The main functions of this modification are genomic imprinting, X chromosome inactivation, suppression of transposons, regulation of gene ...
  24. [24]
    Mammalian DNA methylome dynamics: mechanisms, functions and ...
    This Review examines the mechanisms and functions of regulated DNA methylation and demethylation, highlighting how these dynamics contribute to mammalian ...
  25. [25]
    5-Hydroxymethylcytosine-mediated active demethylation is required ...
    Dec 17, 2021 · 5-Hydroxymethylcytosine-mediated active DNA demethylation occurs in postmitotic neurons, and is required for their terminal differentiation ...
  26. [26]
    5-hydroxymethylcytosine accumulation in postmitotic neurons ...
    Aug 28, 2017 · The main insight from this study is that the role of 5-hydroxymethylcytosine (5hmC) in postmitotic neurons is to sculpt the genome occupancy ...
  27. [27]
    Mechanisms of DNA methylation regulatory function and crosstalk ...
    In this review, we discuss the dynamic interplay between DNA methylation and histone lysine methylation as epigenetic regulators of genome function.Missing: seminal | Show results with:seminal
  28. [28]
    Crystal structure of the nucleosome core particle at 2.8 Å resolution
    Sep 18, 1997 · The X-ray crystal structure of the nucleosome core particle of chromatin shows in atomic detail how the histone protein octamer is assembled.
  29. [29]
    Translating the histone code - PubMed - NIH
    A diverse array of posttranslational modifications that largely impinge on histone amino termini, thereby regulating access to the underlying DNA.
  30. [30]
    linking histone acetylation to chromatin assembly and gene activation
    The recent identification and cloning of histone acetyltransferase ... 1996 Apr;6(2):176-84. doi: 10.1016/s0959-437x(96)80048-7. Authors. J E Brownell , C D Allis ...
  31. [31]
    A Mammalian Histone Deacetylase Related to the Yeast ... - Science
    Trapoxin is a microbially derived cyclotetrapeptide that inhibits histone deacetylation in vivo and causes mammalian cells to arrest in the cell cycle.
  32. [32]
    Regulation of chromatin structure by site-specific histone H3 ...
    Here we show that human SUV39H1 and murine Suv39h1--mammalian homologues of Drosophila Su(var)3-9 and of Schizosaccharomyces pombe clr4--encode histone H3- ...
  33. [33]
    Epigenetic frontiers: miRNAs, long non-coding RNAs and ...
    Oct 16, 2024 · This review highlights the important roles of DNA methylation, histone modifications and non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs) and long non ...
  34. [34]
    MicroRNAs and epigenetics - Sato - 2011 - FEBS Press - Wiley
    Mar 12, 2011 · At the same time, a variety of miRNAs regulate epigenetic pathway-related molecules, most notably polycomb group proteins, HDACs and DNA ...Micrornas And Epigenetics · Epigenetically-Regulated... · Mirnas Regulating Epigenetic...<|separator|>
  35. [35]
    How mammalian piRNAs instruct de novo DNA methylation ... - Nature
    Sep 7, 2020 · This research provided the first mechanistic insights into how piRNA directs de novo DNA methylation in male mammals.Missing: review | Show results with:review
  36. [36]
    Roles of piRNAs in transposon and pseudogene regulation of ...
    Jan 8, 2021 · In this review, we summarize how PIWI proteins and piRNAs regulate the expression of many disparate RNAs, describing a highly complex global genomic regulatory ...
  37. [37]
    Long non-coding RNAs and their implications in cancer epigenetics
    In this review, we highlight how some of the lncRNAs implicated in cancer are involved in the epigenetic control of gene expression. While very few lncRNAs have ...
  38. [38]
    Circular RNA: metabolism, functions and interactions with proteins
    Dec 14, 2020 · In regard to the epigenome, covalent modifying enzymes are recruited to DNA (methylation) or histones (methylation and acetylation) to alter ...
  39. [39]
    Epigenetic inheritance of diet-induced and sperm-borne ... - Nature
    Jun 5, 2024 · Exercise alters mouse sperm small noncoding RNAs and induces a transgenerational modification of male offspring conditioned fear and anxiety.
  40. [40]
    From genomics to epigenomics: a loftier view of life - Nature
    Epigenetic information is amenable to much faster changes than the DNA sequence itself. Therefore, such studies might allow the isolation of the epigenomic ...
  41. [41]
    GWAS: heritability missing in action? | European Journal of Human ...
    Mar 17, 2010 · Within the field of gene × environment interactions, we must not omit to consider epigenetic processes that may contribute substantially to a ...
  42. [42]
    Identification of 55,000 Replicated DNA Methylation QTL - Nature
    Dec 4, 2018 · Genetic control of DNA methylation is a potential candidate for explaining the many identified SNP associations with disease that are not found ...Results · Identification Of Mqtl · Materials And Methods<|control11|><|separator|>
  43. [43]
    Missing heritability: is the gap closing? An analysis of 32 complex ...
    Apr 12, 2017 · Epigenetic variation including methylation profiles have been suggested as another source of missing heritability ... GWAS. The new method ...
  44. [44]
    Methylation across the central dogma in health and diseases - Nature
    Aug 25, 2023 · DNA cytosine methylation can directly promote genetic mutations by spontaneous hydrolytic deamination of 5mC, which leads to C→T transition ...
  45. [45]
    The correlation between CpG methylation and gene expression is ...
    Jul 24, 2024 · The absence of methylated CpGs in these sequences typically correlates with increased gene expression, indicating a regulatory role for methylation.
  46. [46]
    An integrated encyclopedia of DNA elements in the human genome
    Sep 5, 2012 · The Encyclopedia of DNA Elements (ENCODE) project aims to delineate all functional elements encoded in the human genome. Operationally, we ...
  47. [47]
    H3K4me3 Breadth Is Linked to Cell Identity and Transcriptional ...
    Jul 31, 2014 · Trimethylation of histone H3 at lysine 4 (H3K4me3) is a chromatin modification known to mark the transcription start sites of active genes.
  48. [48]
    Modeling the relationship of epigenetic modifications to transcription ...
    Mar 27, 2015 · Enrichments of histone modifications, H3K4me2, H3K4me3 and H3ac, at transcription start sites (TSSs) are positively related to the extents of ...Missing: seminal | Show results with:seminal
  49. [49]
    Predicting gene expression from histone marks using chromatin ...
    Mar 29, 2024 · To understand the complex relationship between histone mark activity and gene expression, recent advances have used in silico predictions ...
  50. [50]
    Quantitative Proteomic Analysis of Histone Modifications - PMC
    Additionally, histone PTMs can modify chromatin structure and function either by recruiting PTM-specific binding proteins (also called “readers”) and their ...
  51. [51]
    The Crosstalk Between Epigenetic Mechanisms and Alternative ...
    In this review, we discuss recent studies and summarize the current knowledge on the epigenetic regulation of alternative RNA processing.Chromatin-Based Regulation... · Epigenetic Regulation and... · Conclusion
  52. [52]
    Epigenetics in Alternative Pre-mRNA Splicing - ScienceDirect.com
    Jan 7, 2011 · A major recent discovery is that chromatin structure and epigenetic histone modifications act as key regulators of alternative splicing.
  53. [53]
    Epitranscriptomics and epigenetics: two sides of the same coin?
    Sep 3, 2024 · Here we will discuss the relationship between canonical and non-canonical epigenetic mechanisms that govern gene expression.Missing: extension omics
  54. [54]
    Spatial epigenome–transcriptome co-profiling of mammalian tissues
    Mar 15, 2023 · We present two technologies for spatially resolved, genome-wide, joint profiling of the epigenome and transcriptome by cosequencing chromatin accessibility and ...Missing: 2020s studies
  55. [55]
    Spatial joint profiling of DNA methylome and transcriptome in tissues
    Sep 3, 2025 · Emerging spatial multi-omics technologies now enable profiling of molecular features in native tissue microenvironments, significantly enhancing ...Missing: 2020s | Show results with:2020s
  56. [56]
    Making Sense of the Epigenome Using Data Integration Approaches
    In this review we provide an overview of recent data analysis approaches to integrate various omics layers to understand epigenetic mechanisms of complex ...<|control11|><|separator|>
  57. [57]
    Uniting the Omics: Integrating Proteomics and Transcriptomics Data
    Mar 8, 2024 · Integrating proteomics and transcriptomics data poses several challenges due to the inherent complexities and heterogeneity of biological datasets.
  58. [58]
    Mapping epigenetic modifications by sequencing technologies
    Sep 1, 2023 · Traditional, bisulfite sequencing is the gold standard in DNA methylation sequencing but it severely damages DNA [4]. Recently, EM-Seq and TAPS ...
  59. [59]
    High-resolution profiling of histone methylations in the human genome
    May 18, 2007 · Histone modifications are implicated in influencing gene expression. We have generated high-resolution maps for the genome-wide distribution ...
  60. [60]
    Genome-wide analysis of histone modifications by ChIP-on-chip
    Here, we provide validated ChIP-on-chip methods for analyzing histone modification patterns at genome-scale in mammalian cells.Missing: review | Show results with:review
  61. [61]
    An efficient targeted nuclease strategy for high-resolution mapping ...
    Jan 12, 2017 · Skene and Henikoff have now developed a new method, called CUT&RUN (which is short for “Cleavage Under Targets & Release Using Nuclease”) to map ...
  62. [62]
    CUT&Tag for efficient epigenomic profiling of small samples ... - Nature
    Apr 29, 2019 · Here we describe Cleavage Under Targets and Tagmentation (CUT&Tag), an enzyme-tethering strategy that provides efficient high-resolution sequencing libraries.
  63. [63]
    A coarse-graining approach for identifying broad domains from ChIP ...
    Some histone modifications, such as H3K27me3 and H3K9me3, usually mark broad domains in the genome ranging from kilobases (kb) to megabases (Mb) long, ...
  64. [64]
    Quality of histone modification antibodies undermines chromatin ...
    Oct 28, 2015 · Raising a specific histone modification antibody is not a trivial task; this is mostly due to the hypermodified state of the histone tail, ...
  65. [65]
    High-Resolution Mapping and Characterization of Open Chromatin ...
    Jan 25, 2008 · Comprehensive Genome-Wide Identification of DNase I HS Sites. To perform DNase-seq, we generated a single DNase I library from primary human CD4 ...
  66. [66]
    Transposition of native chromatin for fast and sensitive epigenomic ...
    Oct 6, 2013 · ATAC-seq queries the location of open chromatin, the binding of DNA-associated proteins and chromatin compaction at nucleotide resolution.
  67. [67]
    Dynamic Regulation of Nucleosome Positioning in the Human ...
    Mar 7, 2008 · We have generated genome-wide maps of nucleosome positions in both resting and activated human CD4 + T cells by direct sequencing of nucleosome ends.
  68. [68]
    Single-cell chromatin accessibility reveals principles of regulatory ...
    We have developed a single-cell Assay for Transposase-Accessible Chromatin (scATAC-seq), improving on the state-of-the-art sensitivity by >500-fold. ATAC-seq ...
  69. [69]
  70. [70]
  71. [71]
  72. [72]
  73. [73]
    Data standards - ENCODE
    ENCODE uses standards for epigenomic assays, including DNA binding, accessibility, methylation, 3D structure, transcription, RNA, and single-cell assays.
  74. [74]
    Eleven grand challenges in single-cell data science | Genome Biology
    Feb 7, 2020 · Sparsity in scRNA-seq data can hinder downstream analyses and is still challenging to model or handle appropriately, calling for further method ...
  75. [75]
    Model-based Analysis of ChIP-Seq (MACS) - Genome Biology
    Sep 17, 2008 · Here, we present Model-based Analysis of ChIP-Seq data, MACS, which addresses these issues and gives robust and high resolution ChIP-Seq peak ...
  76. [76]
    Integrative genomics viewer | Nature Biotechnology
    Jan 1, 2011 · The IGV makes use of efficient, multi-resolution file formats to enable real-time exploration of arbitrarily large data sets over all resolution ...
  77. [77]
    WashU Epigenome Browser update 2022 | Nucleic Acids Research
    Apr 12, 2022 · Abstract. WashU Epigenome Browser (https://epigenomegateway.wustl.edu/browser/) is a web-based genomic data exploration tool that provides ...Abstract · INTRODUCTION · NEW FEATURES · OUTLOOK
  78. [78]
    ChromBPNet: bias factorized, base-resolution deep learning models ...
    Dec 25, 2024 · We introduce ChromBPNet, a deep learning DNA sequence model of base-resolution accessibility profiles that detects, learns and deconvolves assay-specific ...Missing: profiling | Show results with:profiling
  79. [79]
    Getting personal with epigenetics: towards individual-specific ...
    Aug 7, 2023 · We developed eDICE, an attention-based deep learning model that is trained to impute missing epigenomic tracks by conditioning on observed tracks.
  80. [80]
    The ENCODE Imputation Challenge: a critical assessment of ...
    Apr 18, 2023 · The ENCODE Imputation Challenge: a critical assessment of methods for cross-cell type imputation of epigenomic profiles. Jacob Matthew Schreiber ...
  81. [81]
    Assessing and mitigating batch effects in large-scale omics studies
    Oct 3, 2024 · In this review, we highlight the profound negative impact of batch effects and the urgent need to address this challenging problem in large-scale omics studies.
  82. [82]
    [PDF] Polymer physics of nuclear organization and function - bioRxiv
    The second part (section 3 and 4) describes polymer models, starting with Rouse and other associated polymer models used to described chromatin. Asymptotic.
  83. [83]
    Looping Probabilities in Model Interphase Chromosomes - PMC
    Here we present a reliable expression for looping probabilities in equilibrated wormlike chains with finite diameters or capture radii, which strongly modifies ...Model And Methods · Results · Chromosome Territories And...
  84. [84]
    Epigenetic chromatin silencing: bistability and front propagation - PMC
    We formulate a coarse-grained model of chromatin silencing in yeast and study the conditions under which the system becomes bistable, allowing for different ...
  85. [85]
    Model‐based robustness and bistability analysis for methylation ...
    Mar 27, 2021 · We developed a mathematical model describing methylation-based epigenetic memory systems that operate in a bistable range and capture ...Missing: propagation | Show results with:propagation
  86. [86]
    A Stochastic Model of Epigenetic Dynamics in Somatic Cell ...
    This probabilistic Boolean network describes the interplay between gene expression, chromatin modifications, and DNA methylation. The model incorporates recent ...Missing: epigenomics | Show results with:epigenomics
  87. [87]
    Mechanistic stochastic model of histone modification pattern formation
    Oct 27, 2014 · In this study, we present a stochastic mathematical model that describes the molecular mechanisms of histone modification pattern formation ...Missing: Boolean | Show results with:Boolean
  88. [88]
    Confinement Mechanisms for Epigenetic Modifications of ...
    Sep 5, 2023 · For all simulations that are not purely local as described above, the system is simulated as an agent-based model on a one-dimensional ...
  89. [89]
    Artificial Intelligence in cancer epigenomics: a review on advances ...
    Jun 14, 2025 · The integration of AI and cancer epigenomics has significantly advanced precision oncology, with DNA methylation biomarkers becoming a key ...Missing: 2020s | Show results with:2020s
  90. [90]
    Rebooting the Epigenomes during Mammalian Early Embryogenesis
    Here, we review the recent progress in the understanding of epigenetic reprogramming in early embryos and how it contributes to the mammalian early development.
  91. [91]
    Review Epigenetic regulation of early human embryo development
    Dec 7, 2023 · The epigenomes of highly specialized sperm and oocytes are reprogrammed following fertilization to establish a totipotent state suitable for ...
  92. [92]
    A bivalent chromatin structure marks key developmental genes in ...
    We propose that bivalent domains silence developmental genes in ES cells while keeping them poised for activation.
  93. [93]
    H3K27me3-mediated epigenetic regulation in pluripotency ...
    We overview and discuss how H3K27me3 and its modifiers regulate pluripotency maintenance and early lineage differentiation.
  94. [94]
    EZH2-Mediated H3K27me3 Targets Transcriptional Circuits of ...
    May 11, 2022 · Here, we investigated the role of the EZH2-mediated H3K27me3 apposition in neuronal differentiation. We made use of a transgenic mouse model ...
  95. [95]
    Epigenome profiling identifies H3K27me3 regulation of extracellular ...
    Sep 17, 2025 · H3K27me3 is enriched on neuronal differentiation and ECM genes. Identification of PRC2 as a regulator of human NPC cell fate decisions led us ...
  96. [96]
    Single-cell epigenomic reconstruction of developmental trajectories ...
    Jun 24, 2024 · In this study, we use human brain and retina organoid models and present single-cell profiling of H3K27ac, H3K27me3 and H3K4me3 histone ...
  97. [97]
    Persistent epigenetic differences associated with prenatal exposure ...
    Nov 4, 2008 · Here we report that periconceptional exposure to famine during the Dutch Hunger Winter is associated with lower methylation of the IGF2 DMR 6 ...
  98. [98]
    DNA methylation as a mediator of the association between prenatal ...
    Jan 31, 2018 · We focus on our previous studies of the Dutch Hunger Winter, where we documented that prenatal adversity is associated with increases in adverse ...<|separator|>
  99. [99]
    Epigenetic variation: A major player in facilitating plant fitness under ...
    Recent research in plant epigenetics has increased our understanding of how epigenetic variability can contribute to adaptive phenotypic plasticity in natural ...Missing: animals | Show results with:animals
  100. [100]
    How does epigenetics influence the course of evolution? - Journals
    Apr 19, 2021 · Second, epigenetic variation enhances phenotypic plasticity and phenotypic variance and thus can modulate the effect of natural selection on ...How Does Epigenetics... · Abstract · (a) Epigenetic Inheritance...
  101. [101]
    To live or let die? Epigenetic adaptations to climate change—a review
    In this review, we compile existing evidence of epigenetic involvement in acclimatization and adaptation to climate change and discuss derived perspectives.
  102. [102]
    MGMT hypermethylation: a prognostic foe, a predictive friend
    Aug 1, 2007 · The promoter CpG island hypermethylation-associated gene silencing of MGMT is associated with a wide spectrum of human tumors. This epigenetic ...Missing: epigenomics | Show results with:epigenomics
  103. [103]
    the role of O6-methylguanine DNA methyltransferase in human cancer
    Jan 8, 2004 · The methylation-mediated silencing of MGMT has two consequences for cancer. First, tumors with MGMT methylation have a new mutator phenotype ...Missing: epigenomics | Show results with:epigenomics
  104. [104]
    Germline mutations in Histone 3 Family 3A and 3B cause ... - PubMed
    Dec 2, 2020 · 3 beyond cancer: Germline mutations in Histone 3 Family 3A and 3B cause a previously unidentified neurodegenerative disorder in 46 patients.
  105. [105]
    DNA methylation age of human tissues and cell types - PubMed - NIH
    Finally, I characterize the 353 CpG sites that together form an aging clock in terms of chromatin states and tissue variance. Conclusions: I propose that ...
  106. [106]
    Liquid biopsies based on DNA methylation as biomarkers ... - PubMed
    Sep 24, 2022 · Liquid biopsies based on DNA methylation as biomarkers for the detection and prognosis of lung cancer. Clin Epigenetics. 2022 Sep 24;14(1): ...
  107. [107]
    Liquid Biopsy of Methylation Biomarkers in Cell-Free DNA - PubMed
    Jan 23, 2021 · Liquid biopsies, in particular, analysis of cell-free DNA (cfDNA), have emerged as a promising noninvasive diagnostic approach in oncology.
  108. [108]
    vorinostat for treatment of advanced primary cutaneous T ... - PubMed
    On October 6, 2006, the U.S. Food and Drug Administration granted regular approval to vorinostat (Zolinza(R); Merck & Co., Inc., Whitehouse Station, NJ), ...
  109. [109]
    Targeting DNA methylation - PubMed - NIH
    Jun 15, 2009 · Two nucleoside inhibitors of DNA methylation, azacitidine and decitabine, are now standard of care for the treatment of the myelodysplastic syndrome.Missing: DNMT | Show results with:DNMT
  110. [110]
    CRISPR-based epigenome editing: mechanisms and applications
    Nov 22, 2023 · CRISPR/Cas9 technology has been remodeled for epigenome editing by generating a 'dead' Cas9 protein (dCas9) that lacks nuclease activity and juxtaposing it ...
  111. [111]
    Epigenome editing for targeted DNA (de)methylation - PubMed
    Mar 5, 2024 · This review focuses on the recent developments in epigenome editing tools/techniques, technological limitations, and future perspectives
  112. [112]
    CRISPR/Cas9-based epigenome editing: An overview of ... - PubMed
    Jul 15, 2019 · Here, we review strategies for recruitment of effector domains, used in gene regulation and epigenome editing, to the dCas9 DNA-targeting protein.