Fact-checked by Grok 2 weeks ago

Enhancer RNA

Enhancer RNAs (eRNAs) are a class of non-coding RNAs transcribed from enhancer regions, which are distal cis-regulatory DNA elements that activate transcription of target genes in a spatiotemporal manner. While traditionally considered non-coding, recent evidence suggests that some eRNAs may have coding potential and produce micropeptides. These RNAs, typically short and unstable, serve as markers of active enhancers and play crucial roles in facilitating enhancer-promoter looping, modulating chromatin structure, and enhancing transcriptional output. The discovery of eRNAs emerged in from independent genome-wide studies using techniques like followed by sequencing (ChIP-seq) and (RNA-seq), which revealed widespread transcription by at extragenic enhancer sites in macrophages and neurons. In humans, eRNAs number approximately 40,000 to 65,000, with the majority being bidirectional (termed 2d-eRNAs), ranging from 0.5 to 2 kilobases in length, non-polyadenylated, and exhibiting low stability—about 90- to 100-fold less than messenger RNAs or long non-coding RNAs. Roughly 10% are unidirectional (1d-eRNAs), longer than 4 kilobases, and polyadenylated, often overlapping with enhancer-associated long non-coding RNAs (elncRNAs). These molecules are predominantly nuclear, present in low copy numbers (0.5 to 20 per ), and can undergo modifications such as N6-methyladenosine (m6A) and (m5C) that influence their stability and function. eRNAs contribute to gene regulation through multiple mechanisms, including stabilizing loops by interacting with and complexes to approximate enhancers and promoters. They also promote the release of paused from negative elongation factor (NELF) at promoters, thereby facilitating productive transcription elongation of target . Additionally, eRNAs can trap transcription factors like YY1 and co-activators such as to enhance their recruitment, alter accessibility by recruiting acetyltransferases like CBP/p300, and participate in phase-separated transcriptional condensates to concentrate regulatory machinery. Their expression levels correlate positively with nearby activity, underscoring their role as dynamic regulators in , cellular responses, and diseases including cancer.

Overview and Discovery

Definition and Properties

Enhancer RNAs (eRNAs) are a class of non-coding RNAs (ncRNAs) transcribed from enhancer regions in the , which are distal cis-regulatory DNA elements that modulate . These transcripts typically range from 500 to 2000 in length (0.5 to 2 kb) and serve as reliable markers of active enhancers, distinguishing them from silent or poised regulatory elements. Key biochemical and biophysical properties of eRNAs include their predominantly bidirectional transcription from both strands of the enhancer DNA, resulting in divergent transcripts that often overlap at the enhancer . eRNAs are generally expressed at low abundance, with copy numbers per ranging from 0.5 to 20, and exhibit short half-lives, typically less than 2 hours, due to rapid degradation by the nuclear RNA exosome. Most eRNAs, particularly the bidirectional subtype (2d-eRNAs), lack and are unspliced, contributing to their instability; in contrast, a smaller subset of unidirectional eRNAs (1d-eRNAs) may be polyadenylated and longer. These molecules are primarily localized in the , where they remain associated with marked by active modifications such as H3K27 acetylation (H3K27ac) and H3K4 monomethylation (H3K4me1). In distinction from other ncRNAs, eRNAs are uniquely enhancer-derived and transient, differing from messenger RNAs (mRNAs), which are protein-coding, longer, polyadenylated, and stable with half-lives often exceeding 10 hours. Similarly, long non-coding RNAs (lncRNAs) are typically over 200 , more stable, frequently spliced, and capable of diverse structural or distant regulatory roles, whereas eRNAs are shorter, enhancer-specific, and primarily involved in local cis-regulation without coding potential. The evolutionary conservation of eRNAs is variable, with enhancer sequences and their transcripts showing modest preservation across metazoans, though functional activity is more consistently maintained; promoter-proximal enhancers tend to exhibit higher sequence conservation compared to distal ones, reflecting their critical regulatory roles.

Historical Milestones

The concept of enhancer transcription emerged from early observations of pervasive transcription in mammalian genomes. In , Efroni et al. reported widespread hypertranscription in embryonic cells using tiling microarray , revealing bidirectional transcripts emanating from intergenic regions, including those later identified as enhancers. This laid the groundwork for genome-wide studies that directly linked enhancer activity to RNA production. In 2010, two landmark studies confirmed the prevalence of enhancer-derived transcripts. Kim et al. utilized GRO-seq in mouse cortical neurons to detect bidirectional transcription at activity-regulated enhancers, demonstrating their association with stimulus-induced gene expression. Concurrently, Orom et al. identified thousands of long noncoding enhancer RNAs (eRNAs) in human cell lines through integrated analysis of chromatin marks and RNA sequencing, showing their enhancer-like function in promoting target gene activation. These findings established eRNAs as a class of noncoding RNAs transcribed from active enhancers. Subsequent research in 2011–2013 further validated and expanded this discovery. Andersson et al. (2014) analyzed cap analysis of gene expression () data across mammalian tissues, highlighting the broad prevalence of short, bidirectional transcripts from enhancer loci. In 2013, Lam et al. demonstrated that eRNA production correlates with enhancer strength in mouse macrophages, using GRO-seq to show that nuclear receptor Rev-erbα represses by inhibiting eRNA synthesis at target enhancers. Initially, enhancer transcription faced skepticism, often dismissed as transcriptional noise or a non-functional byproduct of open chromatin. This view shifted around 2013 with functional studies providing evidence of eRNAs' regulatory roles; for instance, Li et al. showed that eRNAs facilitate estrogen-dependent gene activation in human breast cancer cells by promoting enhancer-promoter looping and mediator recruitment. By the mid-2010s, large-scale efforts scaled eRNA mapping to genome-wide atlases. The FANTOM5 project in 2014 used deep CAGE sequencing across human and mouse cell types and tissues to catalog tens of thousands of enhancer transcripts, confirming their cell-type specificity and correlation with active chromatin states.

Biogenesis and Classification

Transcriptional Origin

Enhancer activation is initiated by the of transcription factors (TFs) and coactivators, such as p300/CBP, to specific motifs within enhancer DNA sequences. This promotes , notably H3K27ac ( of 27 on ), which opens structure and facilitates access for the transcriptional apparatus. The of (Pol II) to these sites follows, often in a serine 5-phosphorylated form indicative of , enabling the start of transcription at distal regulatory elements. Despite their distal positions relative to promoters, enhancers possess core promoter-like elements, including and , that support Pol II pre-initiation complex assembly and transcription start site selection. This architectural similarity allows enhancers to function as bidirectional promoters, where most arise from divergent transcription producing paired sense and antisense strands from a central core region. Such bidirectionality is a widespread feature of active enhancers, distinguishing them from unidirectional promoter transcription. Transcription initiation at enhancers is tightly regulated by accessibility, as evidenced by DNase I hypersensitive sites that denote nucleosome-depleted regions permissive to TF binding and Pol II engagement. In super-enhancers—clusters of tightly spaced enhancers marked by dense TF and occupancy—Pol II recruitment and transcriptional output are amplified, driving higher eRNA production compared to typical enhancers and supporting robust activation of cell-identity genes. eRNAs are predominantly short transcripts, usually spanning less than 2–3 , owing to inefficient termination signals and the common absence of sites, which results in non-polyadenylated products subject to rapid turnover. Termination frequently relies on the Integrator complex, which cleaves nascent Pol II transcripts at weak sites to prematurely halt elongation and release the polymerase.

Types of eRNAs

Enhancer RNAs (eRNAs) are primarily classified into two categories based on their transcriptional directionality, length, polyadenylation status, and processing: unidirectional (1D) eRNAs and bidirectional (2D) eRNAs. This classification reflects differences in their biogenesis and stability, which influence their roles in gene regulation. The majority of eRNAs are 2D eRNAs, which are produced from typical enhancers through bidirectional transcription by . These transcripts are typically short, ranging from 0.5 to 2 kb in length, and lack at their 3' ends, rendering them unstable and transient with half-lives often under 10 minutes. This instability arises despite 5' capping, due to the lack of and reliance on rapid degradation pathways, primarily mediated by the RNA exosome , a multiprotein assembly that performs 3'-to-5' exonucleolytic degradation. In contrast, 1D eRNAs are transcribed unidirectionally from enhancers, often those associated with super-enhancers—clusters of enhancers that drive robust, cell-type-specific . These eRNAs are longer than 4 kb, and undergo 3' end processing similar to messenger RNAs (mRNAs), including cleavage and by the cleavage and polyadenylation specificity factor (CPSF) complex. This processing confers greater stability compared to 2D eRNAs, allowing them to persist longer in the and potentially exert prolonged regulatory effects, as observed in contexts like neuronal differentiation where super-enhancer-derived 1D eRNAs, such as those near NR2F1, support lineage commitment. Stability of eRNAs is further modulated by post-transcriptional modifications and protein interactions. For instance, N6-methyladenosine (m6A) modifications, deposited by the METTL3-METTL14 writer complex, can enhance the stability of select eRNAs by recruiting reader proteins that protect against degradation, a mechanism elucidated in studies since 2021. Additionally, binding of heterogeneous ribonucleoproteins (hnRNPs), such as hnRNPL, promotes retention of eRNAs by anchoring them to or other nuclear components, preventing export and facilitating local regulatory functions. Rare variants of eRNAs deviate from these canonical forms. Some bidirectional eRNAs acquire , potentially through alternative 3' end processing signals, leading to increased and distinct regulatory potential, though such cases are uncommon. Emerging from 2025 also indicates that approximately 12% of intergenic eRNAs harbor long open reading frames (ORFs >300 ), some of which are translated into small peptides (smORFs), suggesting dual non-coding and coding functionalities in a of these transcripts.

Expression Dynamics

Genomic Prevalence

Enhancer RNAs (eRNAs) are transcribed from enhancer regions throughout the , with genome-wide analyses indicating that approximately 10,000 to 50,000 enhancers actively produce eRNAs in a given , depending on the state of cellular activity. These transcribed enhancers collectively span about 1-2% of the genome, as determined by chromatin accessibility and modification profiles in projects like and the Roadmap Epigenomics Consortium (collectively mapping regulatory elements across 127 human cell types and tissues). In highly active states, such as human embryonic stem cells, around 20,000 enhancers are transcriptionally engaged, reflecting a baseline for pluripotent cells. Cross-species comparisons reveal substantial conservation among mammals but reduced prevalence in invertebrates. In mice, the FANTOM5 atlas identifies roughly 33,000 active enhancers producing eRNAs, with about 60% of human enhancers showing orthologous activity in murine tissues, underscoring evolutionary stability in mammalian regulatory landscapes. By contrast, invertebrate genomes exhibit far fewer eRNA-associated enhancers; for instance, the fruit fly (Drosophila melanogaster) and nematode (Caenorhabditis elegans) collectively harbor only about 2,300 eRNAs across sampled tissues, compared to over 135,000 in mammals, highlighting a divergence in enhancer transcription complexity. eRNA production displays strong cell-type specificity, with approximately 80% of enhancers active in only one or a few types, leading to 10-20% overlap between distinct lineages. This specificity is particularly pronounced in tissues exhibiting high , such as neurons and immune s, where dynamic enhancer transcription supports adaptive . Notably, 5-10% of eRNAs originate from super-enhancers, which are clustered regulatory domains numbering 1,000-3,000 per and disproportionately drive expression of s despite comprising a minority of total enhancers. These prevalence patterns are primarily derived from large-scale consortia including ENCODE, Roadmap Epigenomics (up to 127 epigenomes), and FANTOM5 (across 800+ human samples), which integrate CAGE-seq, ChIP-seq, and DNase-seq to annotate transcribed enhancers genome-wide.

Temporal and Spatial Patterns

Enhancer RNAs (eRNAs) exhibit dynamic temporal expression patterns that align closely with cellular transitions and external cues. During developmental differentiation, eRNAs are among the earliest transcribed elements, often induced rapidly upon transcription factor (TF) activation. For instance, in myoblast differentiation triggered by serum stimulation, eRNAs peak as early as 15 minutes post-induction, preceding the activation of TF promoters and non-TF genes, thereby initiating waves of coordinated target gene expression. This rapid onset facilitates the timely establishment of cell fate programs in transitioning mammalian cells, such as stem cells shifting toward lineage commitment. In response to stimuli, eRNA expression shows swift upregulation, typically within minutes to hours, mirroring the kinetics of associated signaling pathways. Hormone stimulation, such as treatment in cells, induces eRNA transcription at binding sites within 10 to 40 minutes, with maximum levels often reached by 40 minutes and preceding or coinciding with target mRNA induction. Similarly, inflammatory signals activate eRNAs in immune cells, where their synthesis precedes target gene transcription in lipopolysaccharide-stimulated macrophages. These patterns highlight eRNAs' role in amplifying acute regulatory responses to environmental or hormonal inputs. Cell-state dynamics further modulate eRNA expression, with elevated levels observed in proliferating or transitioning cells and rhythmic fluctuations in specific tissues. In the liver, approximately 30% of eRNAs display circadian oscillations, peaking in phased clusters (e.g., 71% between ZT18 and ZT3) that correlate strongly with nearby rhythms and binding sites like Rev-erbα (r=0.9 within 200 kb). This diurnal rhythmicity supports metabolic adaptations in hepatocytes, which undergo daily cycles of and quiescence influenced by feeding-fasting cues. Spatially, eRNAs are predominantly confined to the , where they localize near active enhancers without accumulating at enhancer-promoter loops. Single-molecule in cells confirms eRNAs as exclusively nuclear transcripts, with nascent forms detected at enhancer sites but rare co-localization (<27%) with target mRNAs even at peak induction. Tissue-specific profiles underscore their role in organ development; in the , eRNAs from transcribed enhancers are enriched in cerebellar cells during postnatal , regulating genes like Nfib and Atoh1 in the external and internal granule layers. These brain-enriched eRNAs exhibit cerebellum-specific expression (z-score 2.62 vs. other tissues, p=3.02E-111), supporting spatial patterning in neuronal differentiation. Regulatory inputs from signaling pathways provide feedback that shapes eRNA dynamics, often through recruitment. In immune responses, activation induces eRNAs at immune-related enhancers, such as those near IFNG, where eRNA synthesis enhances binding and histone acetylation via p300 in macrophages and contexts. At the single-cell level, eRNA expression reveals significant variability, as seen in estrogen-stimulated cells where only 62.5% of cells express specific eRNAs like FOXC1 antisense at peak times, independent of ERα or MLL1 activity. Single-cell nascent sequencing further uncovers heterogeneous eRNA profiles across states, linking variability to pathway-specific in diverse populations. Recent atlases, such as those in helper T cell differentiation (as of 2024), highlight cell type-resolved eRNA dynamics underlying immune responses.

Functional Mechanisms

Enhancer-Promoter Looping

Enhancer-promoter looping represents a key mechanism by which enhancer RNAs (eRNAs) enable long-range gene regulation, typically spanning distances of 10-100 kb between enhancers and their target promoters. eRNAs stabilize these loops by interacting with components of the complex, including subunits such as SMC3 and RAD21, and the Mediator complex, which collectively facilitate the physical proximity required for transcriptional activation. This interaction promotes the extrusion and anchoring of loops, allowing transcription factors and at enhancers to contact promoter regions effectively. Prior to the identification of eRNAs' roles around 2013, models of enhancer-promoter looping focused primarily on protein-mediated interactions, such as those involving and for domain insulation or for bridging, without invoking noncoding RNAs. These early proposals, dating back to the following enhancer discovery, emphasized DNA architectural proteins in facilitating loops but lacked evidence for RNA contributions. Subsequent studies updated these models by demonstrating that eRNAs actively participate in loop stabilization, integrating RNA-based regulation into the 3D architecture. Experimental evidence for eRNAs' involvement in looping comes from knockdown studies showing direct functional impacts. For instance, in estrogen receptor-alpha (ERα)-driven systems, depletion of eRNAs from enhancers regulating genes like GREB1 and reduced cohesin and occupancy, disrupted enhancer-promoter interactions as measured by chromatin interaction assays (e.g., ), and consequently lowered target by up to 50-70%. Similarly, in androgen receptor contexts, knockdown of prostate-specific antigen () enhancer eRNA impaired AR-dependent looping and transcription. These findings highlight eRNAs' necessity for maintaining loop integrity during hormone-induced activation. In the broader 3D context, eRNAs often mark loop anchors and correlate with topological associating domains (TADs) identified via sequencing, where enhancer-promoter contacts are enriched within TAD boundaries to ensure regulatory specificity. Integration of eRNA expression profiles with data reveals that active eRNA loci are associated with enhanced enhancer-promoter interactions, particularly in cell-type-specific TADs. Such patterns underscore eRNAs' role in modulating topology for precise control.

Protein Recruitment and Interactions

Enhancer RNAs (eRNAs) play a critical role in transcription factor (TF) trapping at regulatory elements, thereby enhancing occupancy and local concentration. Specifically, eRNAs bind to the ubiquitously expressed YY1, stabilizing its association with enhancers and promoters across the , which increases YY1 signal in assays and promotes efficient gene activation. eRNAs also facilitate the recruitment of coactivators such as and MED1 to assemble transcriptional complexes at super-enhancers. These interactions enhance 's residence time on acetylated , boosting transcriptional output, and contribute to that concentrate the transcriptional machinery. The intrinsically disordered regions of and MED1, modulated by eRNA binding, drive liquid-like at these loci, as extended from foundational studies on coactivator hubs. In regulating (Pol II), eRNAs promote the release of paused Pol II by interacting with the negative elongation factor complex (NELF), displacing it from promoters of immediate early genes and facilitating productive elongation. This process involves eRNA-mediated recruitment of positive transcription elongation factor b (P-TEFb), which phosphorylates Pol II's C-terminal domain to overcome pausing and enhance transcriptional output. The specificity of eRNA-protein interactions is mediated by distinct RNA structural motifs, such as unpaired stretches that enable multivalent binding to NELF subunits, ensuring targeted displacement without relying solely on sequence complementarity. Genome-wide mapping of these interactions has been advanced by techniques like RNA conformation sequencing (RIC-seq), which captures proximal RNA-protein associations at enhancers to reveal regulatory networks. Although most eRNAs function locally in the , emerging evidence suggests rare instances of their to the , where they may exert trans-acting effects by influencing distal protein activities, such as modulating or of target mRNAs through interactions with RNA-binding proteins.

Epigenetic and Chromatin Effects

Enhancer RNAs (eRNAs) play a pivotal role in modulating by recruiting and activating acetyltransferases such as (CBP) and p300. These non-coding transcripts bind directly to the HAT domain of CBP, stimulating its enzymatic activity and promoting the deposition of 27 (H3K27ac), a hallmark modification of active enhancers. This interaction enhances enhancer activity by facilitating an open conformation conducive to binding and recruitment. In addition to , eRNAs influence dynamics through interactions with Polycomb repressive complex 2 (PRC2), which deposits repressive marks. By binding to the subunit of PRC2, eRNAs inhibit its methyltransferase activity, thereby repressing deposition at enhancers and preventing compaction. Furthermore, N6-methyladenosine (m6A) modifications on eRNAs enhance their stability and functional interactions, such as participation in phase-separated transcriptional condensates to concentrate regulatory machinery. eRNAs also contribute to chromatin remodeling by stabilizing interactions with SWI/SNF complexes, which possess activity to reposition and increase chromatin accessibility. Specifically, eRNAs associate with the BRG1 subunit via its AT-hook domain, facilitating the recruitment of the esBAF variant of to enhancer regions and promoting eviction for sustained enhancer openness. The production of eRNAs establishes loops that reinforce enhancer , as increased H3K27ac deposition enhances eRNA transcription from the same locus, amplifying the epigenetic marks in a self-sustaining manner. This regulatory circuit ensures robust and persistent enhancer activation during developmental and stimulus-responsive processes. Quantitative analyses using ChIP-seq data demonstrate strong positive correlations between eRNA expression levels and H3K27ac peak intensities across mammalian genomes, underscoring eRNAs as reliable predictors of active enhancer .

Alternative Roles

Early hypotheses proposed that enhancer RNAs (eRNAs) represent transcriptional noise, arising as non-functional byproducts during the scanning or activation of enhancer regions by , without contributing to regulatory processes. This view, articulated in a analysis of yeast transcription, suggested that much of the pervasive Pol II initiation at enhancers lacks specificity and fidelity, akin to random transcriptional events rather than purposeful . Subsequent investigations in the explored whether eRNA functions could be attributed solely to the act of transcription at enhancers, independent of the sequence or product itself, potentially by mechanically opening structures to facilitate factor access. For instance, enhancer transcription has been shown to promote local and maintain open configurations, as evidenced by experiments where inhibiting transcription with drugs like 5,6-dichloro-1-β-D-ribofuranosylbenzimidazole altered enhancer accessibility without directly targeting the eRNA molecule. This transcription-per-se model posits that the passage of Pol II through enhancers disrupts nucleosomes and recruits remodeling complexes, decoupling eRNA biogenesis from any sequence-specific role. While eRNAs predominantly exert cis-regulatory effects on nearby genes through local interactions, rare instances of trans activity have been documented, where eRNAs are exported from the to influence distant genomic loci or even other chromosomes. Early evidence from comprehensive transcriptomic mapping in cells indicated that some eRNAs can traffic to the and potentially modulate in trans, though such cases remain exceptional compared to the typical cis-limited scope. Emerging research has uncovered potential non-canonical roles for eRNAs, including the of open reading frames (ORFs) within select transcripts. A 2025 analysis revealed that approximately 12% of intergenic human eRNAs harbor long ORFs exceeding 300 , with many exhibiting active into peptides, suggesting a subset of eRNAs may function as or dual-purpose RNAs beyond pure non-coding regulation. Additionally, eRNAs contribute to RNA-mediated processes, where N6-methyladenosine (m6A)-modified eRNAs recruit readers like YTHDC1 to form liquid-like condensates at enhancers, stabilizing transcriptional hubs independent of traditional super-enhancer dynamics. Post-2013 studies have largely refuted the notion of eRNAs as mere transcriptional noise by demonstrating sequence-specific functions essential for enhancer activity. For example, targeted depletion of specific eRNA sequences, but not scrambled controls, disrupted enhancer-promoter looping and target gene activation, highlighting the necessity of eRNA integrity for regulatory outcomes. These findings, supported by functional assays across diverse cell types, established eRNAs as active participants in transcription, shifting the paradigm from byproducts to integral regulators.

Detection Methods

Sequencing-Based Techniques

Sequencing-based techniques have revolutionized the detection and quantification of enhancer RNAs (eRNAs), which are typically low-abundance, non-polyadenylated transcripts produced from enhancer regions. These methods leverage high-throughput sequencing to capture nascent or steady-state RNAs, enabling genome-wide mapping of eRNA expression and distinguishing them from other non-coding RNAs based on their bidirectional and enhancer-associated characteristics. By integrating sequencing data with epigenetic marks, such as lysine 27 acetylation (H3K27ac) via ChIP-seq, researchers can validate active enhancers and quantify eRNA production with high precision. Global run-on sequencing (GRO-seq) and its derivative, precision nuclear run-on sequencing (PRO-seq), are pivotal for capturing nascent transcripts directly from engaged RNA polymerase II, providing a snapshot of active transcription at enhancers. GRO-seq involves isolating nuclei, allowing polymerases to extend nascent RNAs with labeled nucleotides, and sequencing the resulting short fragments to map transcriptionally active regions, including eRNAs from bidirectional enhancers. Introduced in 2008, GRO-seq revealed widespread enhancer transcription in human cells, identifying thousands of eRNA-producing loci. PRO-seq refines this by achieving base-pair resolution through biotinylated nucleotides and directional sequencing, enabling precise mapping of polymerase positions and eRNA start sites. Developed in 2013, PRO-seq has been widely adopted to detect low-level eRNA transcription in diverse cell types, such as during differentiation or stress responses. A variant, PRO-cap, further enhances start-site identification by enriching for capped nascent RNAs, facilitating the distinction of eRNA transcription initiation from promoter activity. Standard RNA sequencing (RNA-seq) variants, adapted for non-polyadenylated transcripts like eRNAs, rely on rRNA depletion or total RNA capture to detect low-abundance species. Total RNA-seq with rRNA depletion using probes or enzymatic methods allows sequencing of the entire transcriptome, including unstable eRNAs, and has identified enhancer-associated transcripts in bulk tissues by aligning reads to annotated enhancer regions. This approach is particularly useful for quantifying eRNA levels in steady-state conditions, though it may underestimate nascent transcription compared to run-on methods. Enhancer-specific protocols, such as cap analysis of gene expression (CAGE), target the 5' caps of transcripts to map precise transcription start sites (TSSs), enabling the identification of eRNA promoters within H3K27ac-enriched enhancers. The FANTOM5 consortium's CAGE dataset, generated from over 1,000 human samples in 2014, cataloged tens of thousands of eRNA TSSs, distinguishing them from other non-coding RNAs through bidirectional expression patterns and enhancer overlap. eRNA-seq pipelines often integrate CAGE with ChIP-seq data to filter for enhancer-specific signals, improving annotation accuracy. Advancements in single-cell sequencing have extended eRNA detection to cellular resolution, revealing cell-type-specific enhancer activity. Single-cell RNA sequencing (scRNA-seq) variants, such as those using 3' or 5' end capture, can profile eRNAs but often require modifications for non-polyadenylated transcripts; for instance, total RNA capture in scRNA-seq detects eRNA expression in immune cell subsets, correlating with enhancer accessibility. Post-2020 developments, including single-cell GRO-seq (scGRO-seq), combine nascent RNA labeling with droplet-based partitioning to map eRNA transcription in individual cells, uncovering coordinated enhancer activation during development. Recent advancements include scGRO-seq (as of 2024), which provides genome-wide, single-cell resolution of transcription to analyze eRNA burst kinetics and enhancer-promoter interactions. Random displacement amplification sequencing (RamDA-seq), introduced in 2018, provides full-length total RNA profiling at single-cell resolution without poly(A) selection, enabling sensitive detection of eRNAs and their dynamics in embryonic stem cell differentiation. RamDA-seq has profiled hundreds of cell-type-specific eRNAs, highlighting their role in recursive splicing and enhancer regulation. Computational pipelines are essential for annotating and analyzing sequencing data to identify eRNAs from raw reads. Tools like facilitate eRNA annotation by quantifying reads in genomic features, performing motif enrichment on enhancer regions, and integrating with ChIP-seq for H3K27ac overlap to prioritize active eRNA loci. 's analyzeRNA.pl module processes nascent RNA data from GRO-seq or PRO-seq, assigning reads to bidirectional enhancer transcripts and filtering artifacts. Specialized tools, such as eRNA-IDO, offer identification of eRNAs from assembled transcriptomes, incorporating to predict enhancer origins and annotate functional elements. Databases like provide curated eRNA profiles from cancer transcriptomes, aggregating data from (TCGA) to enable querying of 9,108 detectable eRNAs across 33 cancer types, including their target genes and clinical associations. These resources support reproducible eRNA discovery, with highlighting prognostic eRNAs in over 60% of cases.

Interaction and Structural Assays

Interaction and structural assays provide critical insights into the molecular interactions and conformational dynamics of enhancer RNAs (eRNAs), enabling researchers to dissect their roles in gene regulation beyond mere detection. These methods focus on mapping eRNA associations with proteins and chromatin, as well as probing their secondary structures, which influence binding affinity and functional outcomes. RNA immunoprecipitation techniques, such as crosslinking and immunoprecipitation followed by sequencing (CLIP-seq) variants, have been instrumental in identifying direct protein-binding sites on eRNAs. For instance, photoactivatable ribonucleoside-enhanced CLIP (PAR-CLIP) has revealed that estrogen receptor alpha (ERα) physically associates with specific eRNAs to mediate enhancer decommissioning in breast cancer cells. Similarly, RNA immunoprecipitation (RIP) assays have shown that bromodomain-containing protein 4 (BRD4) binds to eRNAs, such as the SNAI1 enhancer RNA, to enforce local enhancer activity and stimulate target gene transcription. These assays preserve in vivo binding patterns through UV crosslinking or immunoprecipitation, allowing nucleotide-resolution mapping of interaction sites that highlight eRNA's role in recruiting transcriptional coactivators. To investigate eRNA-chromatin contacts, proximity ligation-based methods like in situ conformation sequencing (RIC-seq) capture spatial interactions between eRNAs and promoter-associated nascent RNAs, thereby mapping enhancer-promoter looping at single-nucleotide . RIC-seq has been applied to generate enhancer-promoter interaction maps, revealing how Alu-derived complementary sequences in eRNAs facilitate looping and activation in cells. Complementary techniques, such as capture hybridization of targets sequencing (CHART-seq), validate these loops by enriching eRNA-chromatin hybrids and sequencing interaction junctions, providing evidence for eRNA-mediated stabilization of three-dimensional architecture. These assays underscore eRNA's involvement in bridging enhancers and promoters, with RIC-seq particularly effective for global profiling of intra- and intermolecular contacts . Structural probing assays elucidate eRNA folding patterns that modulate protein binding and functional modifications. Selective 2'-hydroxyl acylation analyzed by and mutational profiling (SHAPE-MaP) chemically modifies flexible RNA to infer secondary structures, revealing how eRNA folds influence of regulatory factors like complex components. Similarly, dimethyl sulfate sequencing (DMS-seq) identifies and reactivities to map unpaired regions. For instance, studies have shown that m6A-modified sites on eRNAs adopt specific structures that enhance YTHDC1 binding and phase-separated formation for transcriptional (as of 2021). These methods have shown that eRNA secondary structures, particularly around m6A sites, facilitate interactions with readers like YTHDC1, thereby linking chemical modifications to dynamics. In vivo functional assays further test eRNA contributions by manipulating their localization and activity. CRISPR-dCas9 tethering systems fuse eRNA sequences to single guide RNAs, directing catalytically inactive to target enhancers and assessing impacts on transcription; for example, tethering estrogen-regulated eRNAs to their origins in cells stimulates ERα recruitment and H3K27 acetylation, confirming their sufficiency for enhancer activation. Live-cell imaging with MS2 stem-loop tags, co-expressed with MS2 coat protein fused to fluorescent markers, tracks eRNA nuclear localization and dynamics, revealing their retention at enhancer loci during active transcription. Emerging assays employ (FRAP) to evaluate eRNA modulation of liquid-like condensates; FRAP analysis has demonstrated that eRNAs buffer the phase behavior of enhancer-associated proteins like , promoting droplet fluidity and super-enhancer recruitment in hormone-stimulated cells. These techniques collectively validate eRNA's mechanistic roles in real-time cellular contexts.

Biological Implications

Developmental Roles

Enhancer RNAs (eRNAs) transcribed from pluripotency-associated super-enhancers are critical for maintaining () identity by sustaining the expression of core pluripotency factors. For instance, eRNAs derived from enhancers at the Nanog locus facilitate accessibility and epigenetic activation of Nanog, ensuring self-renewal and preventing premature in mouse ESCs; depletion of TET1/2 enzymes reduces these eRNAs by 30-60%, leading to decreased Nanog expression and compromised pluripotency. During , lineage-specific eRNAs emerge to drive targeted gene activation and . In neuronal , the eRNA from the Bdnf Enh170 intergenic enhancer, located approximately 170 kb upstream of the Bdnf gene, is induced upon neuronal activity and promotes expression of Bdnf transcript variants, supporting dendritic branching, growth, and overall neuronal maturation in cortical neurons. Similarly, in myogenic , the DRReRNA (also known as MUNC) from a distal regulatory region recruits to enhance looping and activate Myogenin, facilitating myotube formation. In organogenesis, eRNAs contribute to tissue-specific gene regulation during heart and limb development. For heart development, the CARMN eRNA, transcribed from a super-enhancer associated with the cardiac mesoderm enhancer locus, interacts with PRC2 to repress non-cardiac genes and promote cardiomyocyte differentiation and cardiac commitment, underscoring its role in cardiogenesis. In limb patterning, tissue-specific eRNAs mark distant-acting enhancers, including those regulating Sonic hedgehog (Shh), by correlating with enhancer activity in limb buds and facilitating Shh-mediated anterior-posterior patterning through chromatin interactions. Evolutionarily, eRNAs associated with developmental enhancers exhibit functional conservation across vertebrates, with orthologous enhancers producing similar eRNA profiles despite sequence divergence, preserving regulatory roles in embryogenesis from to mammals.

Disease Associations and Therapeutics

Dysregulated enhancer RNAs (eRNAs) have been implicated in various , where they contribute to tumorigenesis and progression. In , the TMZR1-eRNA, transcribed from the super-enhancer region, regulates cell sensitivity to by controlling expression; its knockdown sensitizes cells to chemotherapy-induced and is overexpressed in resistant tumors. In lung (LUAD), eRNA signatures drive growth pathways and serve as prognostic models; a 7-eRNA predicts survival and reveals eRNA-mediated regulation of tumor invasiveness and clinical outcomes. Similarly, in , the LTFe eRNA promotes ferroptosis resistance by activating the LTFe-LTF axis, enhancing epigenetic regulation of lipid metabolism and tumor progression, positioning it as a potential therapeutic target. Beyond cancer, eRNAs influence non-oncologic diseases through genetic variants and regulatory disruptions. In , single polymorphisms (SNPs) within eRNA-transcribing regions, such as rs258760, associate with disease risk by modulating immune-relevant cellular processes; these eRNA-linked SNPs provide novel functional insights into . In neurodegeneration, particularly , eRNAs regulating BDNF expression are disrupted, leading to reduced neurotrophic support and ; abnormal enhancer activity at the BDNF locus exacerbates neuronal loss, linking eRNA dysregulation to cognitive decline. Pathological mechanisms involving eRNAs often promote and serve as . eRNAs facilitate resistance via transcriptional condensates that stabilize oncogenic pathways, as seen in progression where dynamic eRNA expression sustains tumor adaptation under therapy. The IRS2e eRNA acts as a prognostic in head and neck , where its overexpression correlates with poor survival by enhancing IRS2-mediated oncogenic signaling. Therapeutic strategies targeting eRNAs show promise in overcoming resistance and improving outcomes. Antisense oligonucleotides (ASOs) or CRISPR-based knockdown of NET1e in inhibits and reverses to agents like BEZ235 by disrupting NET1 expression. eRNA signatures also predict immunotherapy response; an antitumor eRNA profile in immune cells enhances accuracy in forecasting efficacy across cancers. Recent advances from 2021-2025 highlight eRNA signatures for chemoresistance prediction, such as in metastases where eRNA profiles identify therapy vulnerabilities. While most eRNAs are non-coding, select translated eRNAs emerge as novel targets by producing functional peptides that drive tumor growth. Databases like CancereRNAQTL facilitate clinical translation by mapping eRNA quantitative trait loci across cancers, enabling personalized prognostic and therapeutic applications.

References

  1. [1]
    Enhancer RNAs: mechanisms in transcriptional regulation and ... - NIH
    Aug 3, 2023 · In this review, we discussed the biological characteristics of eRNAs, their functions in transcriptional regulation, the regulation factors ...
  2. [2]
    Enhancer RNAs in transcriptional regulation: recent insights - Frontiers
    May 16, 2023 · Enhancers are a class of cis-regulatory elements in the genome that instruct the spatiotemporal transcriptional program.
  3. [3]
    Enhancer RNAs: mechanisms in transcriptional regulation and ...
    Aug 3, 2023 · eRNAs are non-coding RNA molecules produced by the transcription of DNA cis-acting elements, enhancer fragments.
  4. [4]
    Enhancer RNAs are an important regulatory layer of the epigenome
    Jun 8, 2020 · Leading the charge in the RNA revolution is a class of ncRNAs that are synthesized at active enhancers, called enhancer RNAs (eRNAs).
  5. [5]
    Enhancer RNA: What we know and what we can achieve - PMC
    Feb 16, 2022 · In this review, we discuss eRNA production, characteristics, functions and mechanics. eRNAs can determine chromatin accessibility, histone ...Missing: papers | Show results with:papers
  6. [6]
    Functional annotation of structural ncRNAs within enhancer RNAs in ...
    Nov 14, 2017 · Relative to the weakly-transcribed enhancers, eRNA regions were longer, closer to the nearest TSS, less evolutionarily conserved, and had looser ...
  7. [7]
    Global transcription in pluripotent embryonic stem cells - PMC
    Here we show that the ES cell genome is transcriptionally globally hyperactive and undergoes global silencing as cells differentiate.
  8. [8]
  9. [9]
  10. [10]
  11. [11]
    Enhancer RNAs - an overview | ScienceDirect Topics
    Enhancer RNAs are most commonly bidirectional (2D-eRNAs) and nonpolyadenylated; however, it was observed that eRNAs can also be unidirectional (1D-eRNAs) and ...
  12. [12]
    The Unforeseen Non-Coding RNAs in Head and Neck Cancer - MDPI
    Generally, 1D-eRNAs are long (over 4 kb) and polyadenylated, while 2D-eRNAs are relatively short (<2 kb) and are not polyadenylated [84,85,86,87]. eRNAs play ...<|separator|>
  13. [13]
    RNA-Degrading Exosome Complexes: Molecular Mechanisms and ...
    Apr 17, 2025 · The RNA exosome is a conserved multiprotein complex essential for 3′-to-5′ RNA degradation in eukaryotic cells.Missing: eRNA | Show results with:eRNA<|separator|>
  14. [14]
    Current progress and future perspective of super-enhancers
    Jul 2, 2025 · Unidirectional transcription generates long (over 4 kb) and polyadenylated eRNA, which is called 1D eRNA (Koch et al., 2011). Bidirectional ...
  15. [15]
    The novel lncRNA lnc-NR2F1 is pro-neurogenic and mutated ... - eLife
    Jan 10, 2019 · lnc-Nr2f1, a long non-coding RNA focally deleted in autism patients, directly occupies and regulates neuronal genes.Missing: eRNA | Show results with:eRNA
  16. [16]
    Biological roles of enhancer RNA m6A modification and its ...
    May 30, 2025 · YTHDF2 recognises the m6A modification in LAST1 mRNA, resulting in reduced stability. By increasing LATS1 mRNA expression and activating YAP ...
  17. [17]
    MyoD induced enhancer RNA interacts with hnRNPL to activate ...
    Dec 19, 2019 · CLIP-seq identifies that hnRNPL binds to eRNAs transcriptome-wide in several cell types, suggesting hnRNPL/eRNA binding could be a general ...
  18. [18]
    Thematic Minireview Series on Results from the ENCODE Project
    Sep 5, 2012 · Based on current estimates of up to 50,000 enhancers in any given cell type and the fact that enhancers tend to be cell type-specific, it has ...
  19. [19]
    Integrative analysis of 111 reference human epigenomes - Nature
    Feb 18, 2015 · Approximately 5% of each reference epigenome shows enhancer and promoter signatures, which are twofold enriched for evolutionarily conserved ...
  20. [20]
    Accurate Promoter and Enhancer Identification in 127 ENCODE and ...
    We map promoters and enhancers for 127 cell types and tissues from the ENCODE and Roadmap Epigenomics projects, today's largest compendium of chromatin assays.Missing: prevalence | Show results with:prevalence
  21. [21]
    An atlas of active enhancers across human cell types and tissues
    We use the FANTOM5 panel of samples covering the majority of human tissues and cell types to produce an atlas of active, in vivo transcribed enhancers. We show ...
  22. [22]
    Animal-eRNAdb: a comprehensive animal enhancer RNA database
    Sep 22, 2021 · We systematically characterized 185 177 eRNAs from 5085 samples across 10 species by mapping the RNA sequencing data to the regions of known enhancers.
  23. [23]
    An integrated encyclopedia of DNA elements in the human genome
    Sep 5, 2012 · Accounting for all these elements, a surprisingly large amount of the human genome, 80.4%, is covered by at least one ENCODE-identified element ...
  24. [24]
    Transcribed enhancers lead waves of coordinated transcription in ...
    Arner et al. used cap analysis of gene expression (CAGE) to analyze gene enhancer and promoter activities in a number of human and mouse cell types. The RNA of ...
  25. [25]
    Enhancer Transcripts Mark Active Estrogen Receptor Binding Sites
    Apr 16, 2013 · In the studies described herein, we used Global Run-On Sequencing (GRO-seq), a method that assays the location and orientation of all active RNA ...
  26. [26]
    From Super-Enhancer Non-coding RNA to Immune Checkpoint
    Only ~2% of the transcribed human genome is accounted for by protein coding exons, thus non-coding RNAs constitute the majority of transcripts (1). Long non- ...
  27. [27]
    Circadian Enhancers Coordinate Multiple Phases of Rhythmic Gene ...
    Furthermore, we show that eRNA oscillations are highly predictive of the rhythmicity and phase of transcription at nearby genes, demonstrating a large-scale and ...
  28. [28]
    Single-cell profiling reveals that eRNA accumulation at enhancer ...
    Dec 8, 2016 · To examine the role of eRNAs in modulating transcription at the single-cell level, we examined their spatial expression patterns in MCF-7 breast ...
  29. [29]
    Identification and characterization of transcribed enhancers during ...
    Jun 26, 2023 · In this study, eRNA transcription was analyzed to identify TEs active during cerebellar development, as a proxy for the developing brain.
  30. [30]
    Single-cell nascent RNA sequencing unveils coordinated global ...
    Jun 5, 2024 · Our analyses of genes and enhancers across 2,635 individual mouse embryonic stem (ES) cells provide a comprehensive view of the dynamic nature ...<|control11|><|separator|>
  31. [31]
    Functional roles of enhancer RNAs for oestrogen-dependent ...
    Jun 2, 2013 · Our data indicate that eRNAs are likely to have important functions in many regulated programs of gene transcription.
  32. [32]
    Enhancer RNAs participate in androgen receptor-driven looping that ...
    Apr 28, 2014 · We report that enhancer RNAs (eRNAs), a class of long noncoding RNAs, participate in the androgen receptor (AR)-dependent looping complex.
  33. [33]
    eRNA profiling uncovers the enhancer landscape of oesophageal ...
    Feb 20, 2023 · Multiple eRNA regions identified in patient samples are therefore associated with super enhancers as exemplified by the ELF3 super enhancer ...
  34. [34]
    Transcription factor trapping by RNA in gene regulatory elements
    Oct 29, 2015 · We show that the ubiquitously expressed TF Yin-Yang 1 (YY1) binds to both gene regulatory elements and their associated RNA species across the entire genome.
  35. [35]
    Coactivator condensation at super-enhancers links phase ... - Science
    These hubs have the potential to phase-separate at higher concentrations. Indeed, Sabari et al. showed that at super-enhancers, BRD4 and Mediator form liquid- ...
  36. [36]
    Enhancer RNAs stimulate Pol II pause release by harnessing ...
    May 4, 2022 · In addition to orchestrating transcription factor binding, enhancers themselves are transcribed bidirectionally by RNA polymerase II (Pol II), ...
  37. [37]
    Diversity and Emerging Roles of Enhancer RNA in Regulation of ...
    A surprising discovery regarding enhancers was made in 2010, when it was shown that enhancer regions are actively transcribed (De Santa et al., 2010; Kim et al.
  38. [38]
    Predicting enhancer transcription and activity from chromatin ...
    Sep 12, 2013 · We provided a powerful tool to identify active enhancers and reveal the relationship between chromatin modifications, eRNA production and enhancer activity.
  39. [39]
    Full article: Transcriptional control by enhancers and enhancer RNAs
    eRNAs are a byproduct of necessary transcription at enhancers. Instead of the eRNA transcript itself, the act of transcription could promote chromatin ...Emerging Paradigms For Erna... · Erna Transcripts Are... · Conclusions And Future...
  40. [40]
    Transcriptional landscape and clinical utility of enhancer RNAs for ...
    Oct 8, 2019 · Enhancer is a distal regulatory DNA that enhances the transcription of a target gene by interacting with target gene promoter. Traditionally ...Missing: origin | Show results with:origin
  41. [41]
    An atlas of active enhancers across human cell types and tissues
    Mar 26, 2014 · Uniquely, FANTOM5 CAGE allows for direct comparison between transcriptional activity of the enhancer and of putative target gene TSSs across a ...Missing: CAGE eRNA
  42. [42]
    Single-cell full-length total RNA sequencing uncovers dynamics of ...
    Feb 12, 2018 · Here we describe random displacement amplification sequencing (RamDA-seq), the first full-length total RNA-sequencing method for single cells.
  43. [43]
    eRNA-IDO: A One-stop Platform for Identification, Interactome ...
    Aug 23, 2024 · This study presents eRNA-IDO, the first integrative computational platform for the identification, interactome discovery, and functional annotation of human ...
  44. [44]
    Identification of a SNAI1 enhancer RNA that drives cancer cell ...
    Mar 25, 2025 · Mechanistically, SNAI1e binds bromodomain containing protein 4 (BRD4) to enforce the local enhancer activity and stimulate SNAI1 transcription.
  45. [45]
  46. [46]
  47. [47]
    Enhancer RNA from STAT3 locus affects temozolomide ...
    Apr 10, 2025 · In this work, we found eRNA from the STAT3 locus (TMZR1-eRNA) that controls the sensitivity of glioblastoma cells to temozolomide.<|control11|><|separator|>
  48. [48]
    Unveiling Enhancer RNAs in Gliomas: A Systematic Review ... - NIH
    Oct 15, 2025 · (2025) identified a novel eRNA transcribed from the STAT3 super-enhancer region, termed TMZR1-eRNA, which regulates glioblastoma cell ...
  49. [49]
    Development and Validation of a 7-eRNA Prognostic Signature for ...
    Collectively, this study constructed a 7-eRNA prognostic model for LUAD, providing a powerful tool for clinical risk assessment and uncovering ...
  50. [50]
    Deciphering functional landscape and clinical implications of ...
    Aug 12, 2025 · Clinical correlation studies evaluating eRNA levels in patients undergoing therapy could further clarify their prognostic or predictive utility.
  51. [51]
  52. [52]
    Enhancer RNA transcription pinpoints functional genetic variants ...
    Mar 31, 2025 · Bidirectional enhancer RNA (eRNA) transcription is a widespread response to environmental signals and glucocorticoids.
  53. [53]
    Enhancer RNA (eRNA) in Human Diseases - MDPI
    Sep 30, 2022 · The onset of neurodegenerative diseases has been linked to abnormal BDNF expression regulated by Bdnf ... The role of BDNF in Alzheimer's disease.
  54. [54]
    Enhancer RNA–mediated transcriptional regulatory programs reveal ...
    Jun 6, 2025 · Recent studies have demonstrated that enhancers can maintain a drug-resistant state via their targeted transcriptional programs (13). Therefore, ...<|control11|><|separator|>
  55. [55]
    Characterization of an Enhancer RNA Signature Reveals Treatment ...
    An eRNA immunotherapy signature developed using antitumor eRNAs in tumor-infiltrating immune cells improves the prediction of response to immunotherapy.
  56. [56]
    Enhancer RNAs profiling in colorectal cancer liver metastases ...
    Jun 11, 2025 · Moreover, eRNAs are involved in cancer drug resistance. Overexpression of neuroepithelial cell transforming gene 1 eRNA (NET1e) induces ...
  57. [57]
    Enhancer RNA in cancer: identification, expression, resources ...
    Apr 26, 2025 · Enhancer RNA (eRNA), a type of non-coding RNA transcribed from enhancer regions, serves as a class of critical regulatory elements in gene ...
  58. [58]
    eRNAQTL
    The CancereRNAQTL Data Portal is a robust data-driven platform that allows scientific researchers to search and download data for analysis.