Fact-checked by Grok 2 weeks ago

Long non-coding RNA

Long non-coding RNAs (lncRNAs) are a diverse class of non-coding RNA transcripts longer than 200 nucleotides that lack significant protein-coding potential and are primarily transcribed by RNA polymerase II. These molecules, often spanning from hundreds to over 100,000 nucleotides, exhibit low sequence conservation across species but harbor conserved functional domains, such as repetitive elements in cases like the XIST lncRNA involved in X-chromosome inactivation. Unlike protein-coding messenger RNAs (mRNAs), lncRNAs typically undergo processing similar to mRNAs, including capping, splicing, and polyadenylation, though some lack poly(A) tails and localize to specific subcellular compartments like the nucleus or cytoplasm. Discovered in the 1990s through studies of imprinted genes like H19 and dosage compensation mechanisms like , lncRNAs have since been cataloged extensively via projects such as GENCODE, revealing thousands of such transcripts in the . They function predominantly as regulators of through diverse mechanisms, including modification, transcriptional interference, and post-transcriptional modulation of mRNA stability and . For instance, lncRNAs can act as scaffolds to recruit -modifying complexes, form RNA-DNA hybrids (R-loops) to influence replication or repair, or sequester proteins to alter signaling pathways. Biologically, lncRNAs are essential for processes such as embryonic development, cell differentiation, and maintenance of genomic stability, with examples like NEAT1 forming paraspeckles for nuclear organization and preventing chromosomal instability. Dysregulation of lncRNAs is implicated in numerous diseases, including cancers—where promotes —and neurodegenerative disorders, positioning them as potential biomarkers and therapeutic targets due to their tissue-specific expression patterns. Despite advances, challenges persist in elucidating their precise mechanisms, given their low phenotypic visibility in studies and context-dependent functions, underscoring the need for standardized and enhanced functional assays.

Fundamentals

Definition and Discovery

Long non-coding RNAs (lncRNAs) are defined as transcripts longer than 200 that lack significant protein-coding potential. This arbitrary length threshold distinguishes them from shorter non-coding RNAs, such as microRNAs or small nucleolar RNAs, while their non-coding nature is assessed through computational tools evaluating features like short open reading frames (typically fewer than 100 codons) and poor evolutionary conservation of potential coding sequences. Unlike protein-coding messenger (mRNAs), lncRNAs show minimal association with ribosomes and are primarily functional as RNA molecules rather than templates for translation. The discovery of lncRNAs began with isolated examples in the late 1980s and early 1990s, predating genome-wide studies. One of the first identified was H19, a maternally imprinted transcript reported in 1991, which encodes an abundant RNA during mouse embryonic development without an open reading frame. Similarly, the Xist RNA, essential for X-chromosome inactivation, was characterized in 1991 as a 15 kb nuclear transcript expressed exclusively from the inactive X chromosome in female mammals. These early findings highlighted lncRNAs' roles in imprinting and dosage compensation but were viewed as exceptions in a predominantly protein-coding transcriptome paradigm. A major shift occurred in the mid-2000s with high-throughput technologies that unveiled the non-coding transcriptome's scale. Tiling array experiments, such as those from the FANTOM consortium in 2005, detected thousands of unannotated transcripts across the mouse genome. further accelerated discoveries from 2008 onward, identifying pervasive transcription beyond protein-coding genes and confirming lncRNAs' ubiquity in eukaryotic cells. Nomenclature evolved alongside these advances, initially focusing on genomic context; the term "large intergenic non-coding RNAs" (lincRNAs) was coined in for intergenic transcripts longer than 200 nucleotides. By the early 2010s, the broader "lncRNA" designation encompassed diverse classes, including antisense and intronic types, as recommended by bodies like the . The project's 2012 GENCODE v7 annotation established rigorous criteria for lncRNA identification, requiring evidence of transcription, , and absence of protein-coding capacity via tools like and . This framework emphasized functional annotation over mere length, prioritizing transcripts with biochemical signatures akin to mRNAs but without translational output.

Abundance and Diversity

Long non-coding RNAs (lncRNAs) represent a substantial fraction of the eukaryotic , with estimates indicating that they comprise over 68% of annotated genes in deep-sequencing surveys of the polyadenylated (polyA+) RNA fraction. In s, conservative annotations from GENCODE release 49 (2025) identify 35,899 lncRNA genes, producing 191,079 transcripts, while broader catalogs such as NONCODE v6.0 (updated 2021) document around 173,112 lncRNA transcripts, highlighting the variability in detection and annotation approaches. These numbers underscore that lncRNAs may outnumber protein-coding genes, potentially exceeding 100,000 loci when including unannotated or lowly expressed variants, though many remain uncharacterized due to challenges in distinguishing them from transcriptional noise or degraded mRNAs. LncRNAs exhibit considerable diversity in their and expression profiles. Based on their positional relationship to protein-coding genes, they are broadly categorized as intergenic (also known as lincRNAs), antisense, intronic, and enhancer-associated, with intergenic lncRNAs often residing in regions between genes and antisense types overlapping the opposite strand of coding loci. Expression-wise, lncRNAs display higher tissue specificity than mRNAs, with studies reporting that up to 70% of lncRNAs are restricted to specific s or types compared to about 40% for protein-coding transcripts, reflecting their roles in fine-tuned regulatory networks during development and . This variability is further evident in their sequence features, where lncRNAs generally have lower sequence and fewer exons than mRNAs, contributing to their functional . Across eukaryotes, lncRNA abundance scales with organismal complexity, with humans harboring over 100,000 predicted lncRNAs compared to far fewer in simpler models like yeast (), where only hundreds of stable lncRNAs have been identified despite pervasive transcription covering 75-85% of the genome. Recent advances in long-read and direct RNA sequencing (2023-2024) have doubled estimates of lncRNA diversity in non-model organisms, such as , revealing thousands more transcripts in species like and crops, often linked to stress responses and development. Despite these insights, annotation remains incomplete, as databases like NONCODE continue to expand with experimentally validated entries, emphasizing the need for improved computational tools to resolve the full spectrum of lncRNA repertoires.

Biogenesis and Properties

Transcription and Processing

Long non-coding RNAs (lncRNAs) are primarily transcribed by (Pol II), similar to protein-coding messenger RNAs (mRNAs), with the vast majority of annotated lncRNA loci showing Pol II occupancy and associated marks such as lysine 4 trimethylation () at promoters and H3K36me3 in bodies. This Pol II-dependent transcription enables lncRNAs to undergo co-transcriptional processing akin to mRNAs, although a small subset of lncRNAs is transcribed by RNA polymerases I or III, such as certain intergenic transcripts derived from rDNA repeats or Pol III-driven elements like Alu-derived lncRNAs. LncRNA promoters often exhibit bidirectional activity, co-transcribing with nearby protein-coding in convergent or divergent orientations, which can influence local architecture and . Additionally, many lncRNAs arise from enhancer regions, where divergent transcription from bidirectional enhancers produces unstable enhancer RNAs (eRNAs) that can mature into longer lncRNA forms, contributing to regulatory feedback loops. Following transcription initiation, lncRNA precursors undergo canonical processing steps that mirror those of mRNAs, including 5' capping with a 7-methylguanosine (m7G) cap shortly after Pol II initiation, which protects the transcript from exonucleases and facilitates subsequent maturation. Splicing removes via the , with most lncRNAs being multi-exonic and featuring an average of ~2 exons (thus ~1 ) per transcript, fewer than typical mRNAs, though lengths are typically longer than in mRNAs, leading to inefficient or retained splicing in some cases. At the 3' end, cleavage and occur at sites (), resulting in many lncRNAs being polyadenylated (polyA+), similar to mRNAs, while others are polyA- and often more nuclear-retained, due to variability in signals. Mature polyA+ lncRNAs are exported to the via the TAP/NXF1 (also known as Mex67-Mtr2 in ) heterodimer, which binds the polyA tail through adaptor proteins like the complex, facilitating translocation through complexes. LncRNA maturation exhibits notable variations that distinguish it from mRNA processing, including instances of unspliced transcripts that retain introns and function in the , such as certain stress-responsive lncRNAs that evade full splicing due to weak sites. Although circular RNAs (circRNAs) form via backsplicing and are considered a distinct class, some lncRNAs may form stable secondary structures that mimic looped conformations, enhancing stability in specific contexts. Studies have highlighted Pol II pausing and termination as key factors in lncRNA biogenesis; for example, promoter-proximal pausing mediated by the NELF complex allows co-transcriptional regulation, while termination dynamics can generate isoform diversity beyond standard mRNA pathways. Aberrant lncRNA transcripts, such as those with improper capping, splicing defects, or failed , are subject to rigorous mechanisms to prevent accumulation of dysfunctional RNAs. The 5'-3' exonuclease XRN2 plays a central role in degrading these aberrant species co-transcriptionally or shortly after release from Pol II, often in coordination with surveillance factors like the exosome complex, thereby maintaining genomic stability and preventing interference with normal . LncRNAs exhibit half-lives comparable to mRNAs, averaging around 10-15 hours depending on subcellular location, though some studies report medians of ~3-5 hours for specific subsets, reflecting variable turnover via pathways like XRN2-mediated decay. This variability, measured via genome-wide metabolic labeling, contrasts with the more stable profiles of mRNAs and highlights lncRNAs' adaptation for dynamic cellular responses.

Structure and Stability

Long non-coding RNAs (lncRNAs) often adopt complex secondary structures characterized by stem-loops, hairpins, and modular domains that contribute to their functional architectures. These elements are predicted computationally using tools like RNAfold, which employs thermodynamic models to forecast stable folding patterns based on base-pairing probabilities. For instance, the lncRNA HOTAIR exhibits an intricate secondary structure comprising multiple hairpins and repeat motifs, including a 300-nucleotide domain with tandem stem-loops that facilitate modular interactions. Such structural motifs enhance the specificity of lncRNA recognition by binding partners, as demonstrated by studies altering stem lengths and loop sizes in HOTAIR. Beyond secondary folding, lncRNAs engage in tertiary interactions that stabilize higher-order conformations, particularly through associations with RNA-binding proteins (RBPs). Heterogeneous nuclear ribonucleoproteins (hnRNPs), such as hnRNP A2/B1, bind to specific motifs in lncRNAs like HOTAIR, promoting compact tertiary structures via multivalent contacts that bridge distant RNA domains. Additionally, lncRNAs such as NEAT1 drive into membraneless organelles like paraspeckles by forming dynamic RNA-protein networks; NEAT1's modular domains, including repeat-containing regions, nucleate aggregation with RBPs like and SFPQ, resulting in liquid-like condensates that sequester cellular components. These interactions underscore the role of lncRNA tertiary architecture in compartmentalizing nuclear processes. The stability of lncRNAs is modulated by sequence elements and protein interactions that influence their degradation and subcellular persistence. AU-rich elements (AREs) in lncRNA transcripts promote rapid decay by recruiting decay factors, similar to their role in mRNAs, though lncRNAs generally exhibit variable half-lives due to processing differences. Stabilizing RBPs like HuR counteract this by binding AU-rich or cytosine-rich stretches; for example, the lncRNA recruits HuR to its target sites, delaying exonucleolytic degradation and extending transcript lifespan. Furthermore, many lncRNAs display prolonged retention compared to cytoplasmic mRNAs, attributed to weak splice site motifs and interactions with RBPs that hinder export, thereby maintaining higher steady-state levels in the . Structural studies, including imaging techniques, have revealed conformational flexibility in lncRNA-RBP complexes like NEAT1 in paraspeckles. As of 2025, genome-wide studies continue to refine lncRNA annotations, highlighting roles of additional modifications like in modulating structure and function. Concurrently, investigations into epitranscriptomic modifications demonstrate that N6-methyladenosine (m6A) marks subtly alter lncRNA folding by disrupting base stacking in stems, leading to more open conformations that influence RBP affinity and stability, as observed in thermodynamic analyses of m6A-modified lncRNAs like MALAT1.

Genomic Organization

Location and Architecture

Long non-coding RNAs (lncRNAs) are transcribed from diverse genomic contexts, including intergenic regions, where they are classified as long intergenic non-coding RNAs (lincRNAs), as well as genic regions involving intronic and exonic overlaps with protein-coding genes. Antisense lncRNAs, transcribed from the opposite strand of protein-coding genes, represent another major category, often overlapping with sense transcripts. A substantial proportion of lncRNA loci, estimated at around 20-40% in various annotations, are associated with regulatory elements such as enhancers and promoters, reflecting their potential integration into gene regulatory networks. LncRNA loci exhibit distinctive architectural features that mirror those of protein-coding genes, including the presence of bidirectional promoters that drive divergent transcription from shared regulatory elements. These loci are frequently enriched within topologically associating domains (TADs), self-interacting regions that compartmentalize the , with lincRNA promoters often positioned relative to orthologous genes in syntenic blocks across . Clustering of lncRNA genes in syntenic regions underscores their positional conservation, facilitating coordinated regulation within loops. From an evolutionary perspective, lncRNA architectures arise through mechanisms such as tandem duplications, which contribute to the rapid birth and diversification of lncRNA repertoires, often leading to volatile turnover across lineages. Recent chromatin conformation capture studies, including analyses from 2023-2025, have revealed that many lncRNA loci localize at or near TAD boundaries and loops, influencing higher-order . In terms of annotation, approximately 19-20% of annotated lincRNAs overlap with or are in proximity to pseudogenes, complicating their classification and highlighting evolutionary relationships between non-coding and defunct coding elements. Tools like lncRNASNP provide resources for assessing how single polymorphisms (SNPs) and mutations impact lncRNA secondary structures and regulatory architectures, aiding in the functional of these loci.

Translation Potential

Long non-coding RNAs (lncRNAs) exhibit limited coding potential primarily due to the presence of short open reading frames (sORFs) typically shorter than 100 amino acids, which often fail to produce stable, functional proteins. Additionally, upstream ORFs (uORFs) in the 5' regions of many lncRNAs can inhibit downstream translation by stalling ribosomes or promoting premature termination. Ribosome profiling studies indicate that fewer than 5% of lncRNAs show ribosome association patterns consistent with productive translation, in contrast to protein-coding mRNAs, underscoring their predominantly non-coding nature. Despite these limitations, emerging evidence from ribosome sequencing (Ribo-seq) demonstrates that a subset of lncRNAs undergoes to yield micropeptides. Studies, including a 2022 analysis in embryogenesis revealing that approximately 30% of lncRNAs harbor sORFs actively engaged by s, resulting in the regulated production of 100 to 300 micropeptides, and human studies identifying approximately 299 lncRNA-encoded small encoded peptides (SEPs) across cell lines and tissues, support this. For instance, the lncRNA ASncmtRNA, transcribed from the mitochondrial genome, encodes the micropeptide SHLP2, which localizes to mitochondria and modulates cellular processes such as energy metabolism and in tumor cells. These micropeptides often exert biologically significant roles, including regulation of ; for example, certain lncRNA-derived peptides enhance mitochondrial complex IV activity, thereby promoting G1/S transition. Translation of lncRNAs is further constrained by regulatory mechanisms that disfavor efficient initiation. (IRES) elements, which enable cap-independent , are rare in lncRNAs compared to or select cellular mRNAs. Moreover, cap-dependent scanning is often inefficient due to stable secondary structures at the 5' ends of lncRNAs, which impede progression. The translational potential of lncRNAs remains controversial, with debates centering on whether observed ribosome associations represent true protein synthesis or experimental artifacts such as non-specific binding. Recent mass spectrometry-based validations have addressed these concerns, identifying approximately 85 to 100 unique lncRNA-derived peptides in human cells, confirming their existence and tissue-specific expression. As of 2025, recent reviews highlight advances in validating lncRNA-derived micropeptides using mass spectrometry and their roles in cellular metabolism. These findings suggest that while translation is not a dominant feature of lncRNAs, the resulting micropeptides may contribute to fine-tuned regulatory functions in cellular physiology.

Regulatory Functions

Transcriptional Regulation

Long non-coding RNAs (lncRNAs) play critical roles in by modulating the initiation, pausing, and elongation phases of (Pol II)-mediated transcription through both - and mechanisms. These RNAs often interact with , transcription factors, or components of the basal transcriptional machinery to fine-tune in a context-specific manner. In cis-acting mechanisms, lncRNAs primarily influence nearby genes on the same chromosome by facilitating architecture changes or recruiting regulatory es. For instance, enhancer-associated lncRNAs (eRNAs) can tether promoters to distal enhancers via interactions with the Mediator , promoting long-range DNA looping that enhances Pol II recruitment and transcriptional activation. A seminal example is the lncRNA ncRNA-a, which binds the Mediator subunit MED12 to stabilize enhancer-promoter loops, thereby activating the expression of target genes such as via looping. Similarly, the lncRNA FENDRR regulates mesodermal genes during embryonic by associating with Polycomb repressive 2 (PRC2) and Trithorax group (TrxG)/mixed- leukemia (MLL) es, leading to balanced H3K27me3 and H3K4me3 modifications that either repress or activate transcription in a gene-specific fashion; in limb models, FENDRR depletion disrupts these states, impairing patterning and . These cis effects ensure precise spatial control over transcription without requiring the lncRNA to diffuse far from its site of synthesis. Trans-acting mechanisms allow lncRNAs to regulate distal or multiple genes by sequestering transcription factors or modulating the core transcriptional apparatus. One prominent mode involves lncRNAs acting as decoys to inhibit transcription factor activity; for example, the lncRNA NKILA binds the /IκB complex in the , masking IκB phosphorylation sites to prevent IKK-mediated IκB and subsequent activation, thereby suppressing -dependent transcriptional activation of pro-inflammatory genes. Additionally, some lncRNAs target the basal machinery, such as the C-terminal domain (CTD) of Pol II, to influence pausing and ; certain nuclear lncRNAs bind the unphosphorylated Pol II CTD to sterically hinder progression or recruit kinases like P-TEFb for CTD at Ser2, facilitating pause release. Pol III-transcribed lncRNAs, such as 7SK RNA, further exemplify this by scaffolding the 7SK small nuclear ribonucleoprotein () complex, which sequesters P-TEFb in an inhibitory state; upon signaling, 7SK dissociates, releasing active P-TEFb to phosphorylate the Pol II CTD and promote transcriptional at paused genes. Recent studies highlight lncRNAs' involvement in Pol II pausing release, a key checkpoint for productive transcription. For instance, a 2023 review of indicated that these lncRNAs coordinate with NELF and DSIF complexes at promoter-proximal regions to facilitate pause release of Pol II; their depletion stabilizes paused Pol II, impairing elongation at developmental genes; this mechanism underscores lncRNAs' role in balancing transcriptional poising for rapid responses. Overall, these regulatory strategies enable lncRNAs to integrate cellular signals into precise transcriptional outputs.

Post-Transcriptional Regulation

Long non-coding RNAs (lncRNAs) play a pivotal role in modulating mRNA splicing by interacting with components and influencing patterns. A prominent example is MALAT1, which localizes to nuclear speckles and interacts with serine/arginine-rich () splicing factors, thereby regulating their and distribution to promote efficient assembly.00621-0) MALAT1 also stabilizes small nuclear ribonucleoproteins (snRNPs) within these subnuclear structures, facilitating the of splicing machinery to nascent pre-mRNAs.01303-2) Beyond direct interactions, lncRNAs such as NEAT1 and can induce switches in by altering the localization of splicing factors like PTBP1, leading to inclusion or skipping in target transcripts. These mechanisms ensure precise post-transcriptional processing, with dysregulation often linked to cellular phenotypes like . In mRNA stability regulation, lncRNAs frequently act as competing endogenous RNAs (ceRNAs) by sequestering microRNAs (miRNAs), thereby preventing miRNA-mediated degradation of target mRNAs. The PTENP1 lncRNA exemplifies this, functioning as a ceRNA that sponges miR-21 and other miRNAs in the PTEN network, stabilizing PTEN mRNA and suppressing tumor progression in cancers like . Similarly, lncRNAs interfere with pathways, such as Staufen1-mediated mRNA (SMD), where they form RNA duplexes with 3' untranslated regions (UTRs) of target mRNAs to recruit Staufen1 and Upf1, triggering and 5'-to-3' exonucleolytic degradation. This SMD pathway, activated by lncRNAs like BACE1-AS, fine-tunes the stability of specific transcripts during stress or development. Quantitative models of ceRNA competition highlight that effective sponging requires substantial overlap in miRNA response elements (MREs), with derepression thresholds exceeding endogenous MRE abundance by at least 1.5-fold to impact miRNA repression significantly. LncRNAs also exert control over mRNA translation through mechanisms involving upstream open reading frames (uORFs) and dynamics. Some lncRNAs, such as those with translated uORFs, regulate their own expression by modulating reinitiation or stalling at uORF stop codons, indirectly influencing associated protein networks. In trans, lncRNAs like bind and prevent stalling on nascent peptides during cardiac , thereby maintaining translational fidelity and attenuating pathological remodeling. Additionally, certain lncRNAs serve as precursors for small interfering RNAs (siRNAs) in specific contexts, such as in mammalian germ cells, where they are processed into pri-siRNAs to guide and aid siRNA biogenesis pathways. Recent advances, including transcriptome-scale CRISPR-Cas13 screens in , have identified over 700 context-specific essential lncRNAs across human cell lines, with subsets implicated in splicing networks through perturbation of events and integrity. These screens revealed approximately 50 lncRNAs that, when disrupted, alter splicing factor localization and pre-mRNA processing efficiency, underscoring their non-redundant roles in post-transcriptional networks. Such findings, combined with computational models assessing MRE overlap in ceRNA interactions, provide a for predicting lncRNA-mediated regulatory impacts without exhaustive derivations.

Epigenetic Regulation

Long non-coding RNAs (lncRNAs) play crucial roles in epigenetic regulation by modulating states and facilitating heritable through interactions with histone-modifying complexes. These RNAs often act in or to recruit enzymatic machinery that deposits repressive marks, such as lysine 9 (H3K9) and lysine 27 (H3K27) trimethylation, thereby establishing and maintaining domains essential for developmental processes like and dosage compensation. By serving as scaffolds or guides, lncRNAs enable precise targeting of these complexes to specific genomic loci, influencing without altering the underlying DNA sequence.01206-6) A prominent example of lncRNA-mediated imprinting control is the Airn lncRNA, which silences the imprinted Igf2r gene in cis during mouse embryonic development. Airn recruits the G9a to the Igf2r promoter, leading to H3K9 dimethylation and subsequent transcriptional repression in extraembryonic tissues. This mechanism ensures parent-of-origin-specific , with Airn transcription initiating as early as the stage and persisting to maintain silencing. Disruption of Airn or G9a abolishes this repression, highlighting the lncRNA's direct role in epigenetic imprinting.00205-5) In X-chromosome inactivation (XCI), the Xist lncRNA coats the future inactive X chromosome (Xi) in female mammals, recruiting Polycomb repressive complex 2 (PRC2) to catalyze H3K27me3 deposition across large chromatin domains. This coating spreads bidirectionally from the Xist locus, initiating chromosome-wide silencing and forming the Barr body. The antisense Tsix lncRNA antagonizes Xist by competing for shared regulatory elements and preventing Xist upregulation on the active X chromosome, thus ensuring monoallelic Xist expression and proper XCI choice. PRC2 recruitment by Xist involves specific RNA domains, such as the A-repeat, which directly bind EZH2, the catalytic subunit of PRC2, to propagate repressive marks.01415-3)81659-2)00093-5) Beyond imprinting and XCI, lncRNAs like HOTAIR mediate epigenetic modifications at loci by bridging PRC2 and lysine-specific demethylase 1 (LSD1). HOTAIR tethers PRC2 to induce and LSD1 to remove the active H3K4me mark, coordinately repressing in trans across chromosomes. This dual action establishes bivalent states that poise developmental genes for activation or silencing. Similarly, telomeric repeat-containing RNA () promotes formation at chromosome ends by recruiting PRC1 and PRC2, facilitating , , and H4K20me3 marks that protect s from DNA damage and recombination. TERRA's localization to telomeric repeats stabilizes these repressive domains, preventing telomere dysfunction.00501-9) Recent studies have revealed advanced mechanisms, including lncRNAs' involvement in phase-separated condensates that sustain epigenetic memory. For instance, certain lncRNAs nucleate liquid-liquid of PRC2 and other factors, concentrating them at target loci to reinforce propagation during . Additionally, epitranscriptomic modifications like N6-methyladenosine (m6A) on lncRNAs influence PRC2 binding; m6A-marked chromatin-associated lncRNAs recruit PRC2 via readers like RBFOX2, enhancing focal deposition and in hematopoiesis. In , m6A on HOTAIR modulates its stability and interaction with epigenetic complexes, underscoring the interplay between modifications and regulation.01206-6)

Genome Maintenance

Long non-coding RNAs (lncRNAs) play critical roles in regulating timing by influencing states at replication origins. For instance, ASAR lncRNAs, such as ASAR6 and ASAR15, control chromosome-wide replication timing in by promoting the spreading of , which delays replication initiation in specific genomic regions. These lncRNAs interact with chromatin-modifying complexes during the , ensuring coordinated replication with phases like progression and preventing untimely origin firing that could lead to genomic instability. In maintaining chromosome stability, lncRNAs contribute to telomere protection and centromere function. The telomeric repeat-containing RNA (TERRA), a lncRNA transcribed from subtelomeric regions, regulates telomere length by inhibiting activity through direct binding to the enzyme's RNA component, thus preventing excessive . Additionally, TERRA promotes at telomeres by forming R-loops that facilitate pathways, supporting alternative lengthening of telomeres (ALT) in telomerase-deficient cells. At centromeres, lncRNAs like CCTT mediate assembly by facilitating RNA-DNA and RNA-protein interactions that recruit the centromere protein CENP-C to centromeric DNA, ensuring proper attachment during . LncRNAs are integral to the by recruiting repair factors and modulating damage signaling. The lncRNA maintains genomic stability by sequestering PUMILIO proteins, which otherwise destabilize mRNAs involved in control and ; depletion leads to increased DNA damage and . In resolution, recent studies highlight lncRNAs' roles in preventing persistent RNA-DNA hybrids that trigger DNA breaks; for example, TERRA-associated R-loops at telomeres are resolved to support recombination-based repair, with dysregulation linked to activation in cancer cells. Alu-derived lncRNAs, such as those in lincRNA-p21, aid in by forming double-stranded RNA structures via inverted Alu repeats, which activate protein kinase R (PKR) to enhance p53-mediated repair pathways following double-strand breaks. Quantitative impacts of lncRNAs on replication dynamics underscore their protective functions. Depletion of the chromatin-associated lncREST results in sustained replication fork progression, increasing fork speed by approximately 20-30% under stress conditions and leading to unrepaired DNA damage due to bypassed stalling signals. Similarly, knockdown of the ATR/Chk1-interacting lncRNA (ACIL) suppresses replication fork speed by up to 40% during hydroxyurea-induced stress, highlighting lncRNAs' role in stabilizing fork velocity for error-free duplication.

Evolutionary Aspects

Sequence Conservation

Long non-coding RNAs (lncRNAs) generally exhibit low sequence conservation at the nucleotide level compared to protein-coding mRNAs, with most showing less than 10% sequence beyond mammalian due to rapid evolutionary turnover and relaxed selective constraints. In comparisons between and , exonic sequences of lncRNAs typically display low , often below 30%, such as 22% in cases like the Rmst lncRNA, reflecting weaker purifying selection on primary sequences. This limited conservation is evident in large-scale genomic alignments, where only a small of lncRNAs—estimated at around 5%—maintain sequence similarity across broader clades. Promoters of lncRNAs demonstrate higher conservation than their exonic regions, with PhastCons conservation scores around 0.2-0.3 in human-mouse comparisons, comparable to those of protein-coding genes, suggesting stronger selective pressure on transcriptional initiation sites. Functional domains within lncRNAs, such as RNA-binding motifs or repeat elements, often show elevated conservation; for instance, the GAS5 lncRNA retains approximately 70% nucleotide homology in its exons between human and mouse. Additionally, syntenic preservation occurs at lncRNA loci across mammals, with approximately 25% of human lncRNAs having orthologs in mice based on genomic synteny and expression patterns, indicating that genomic context contributes to evolutionary stability even when sequences diverge. Comparative genomics tools reveal that lncRNAs have lower PhastCons scores than mRNAs, with mean placental scores for lncRNA exons typically below those of exons, underscoring reduced nucleotide-level preservation. Alignments via the facilitate these analyses by mapping multi-species sequences and identifying conserved patches. A 2023 pan-eukaryote study highlighted evolutionary bursts of lncRNA conservation particularly in vertebrates, where multidimensional analyses (including PhastCons thresholds >0.58) identified subsets with enhanced sequence retention linked to regulatory roles. Notable exceptions include ultra-conserved elements (UCEs) within lncRNAs, such as transcribed ultra-conserved RNAs (T-ucRNAs or ucRNAs), which exhibit 100% identity over >200 across , , and genomes, comprising 481 such regions with high functional potential. As of 2024, computational tools have identified additional conserved lncRNA functions across vertebrates using multi-omics data.

Functional Conservation

Functional conservation of long non-coding RNAs (lncRNAs) refers to the preservation of their biological roles across species despite limited sequence similarity, often through maintained interactions with conserved cellular machinery or regulatory networks. A key example of functional synteny is observed in X-chromosome dosage compensation, where the lncRNA in placental mammals orchestrates , while a functional ortholog, Rsx (RNA-on-the-silent X), performs a similar cis-acting role in marsupials by silencing the paternal during imprinted X-chromosome inactivation. This highlights how lncRNA-mediated mechanisms can evolve to achieve equivalent dosage balance in diverse mammalian lineages. Similarly, approximately 25% of annotated lncRNAs exhibit functional orthologs in mice, as evidenced by shared expression patterns, genomic synteny, and phenotypic effects upon perturbation, underscoring the selective pressure on lncRNA loci for retained regulatory functions despite sequence divergence. Adaptive evolution of lncRNAs is particularly evident in , where rapid turnover of brain-specific lncRNAs has contributed to lineage-specific neurodevelopmental traits. For instance, many lncRNAs display during early development, with structural but high sequence variability, suggesting that their roles in modulating neuronal and are preserved through secondary structures or binding motifs rather than primary sequences. This turnover is exemplified by lncRNA-derived genes unique to s that encode proteins influencing brain-specific functions, indicating accelerated evolution in cognitive-related pathways. Furthermore, lncRNA interactions with conserved RNA-binding proteins (RBPs), such as TLS/FUS, demonstrate functional preservation; the lncRNA hsrω, which regulates FUS dimethylation, rescues ALS-like phenotypes in human cellular models, illustrating cross-species of RBP-lncRNA complexes in formation and neurodegeneration. Cross-species evidence further supports lncRNA functional conservation through comparable knockout phenotypes. The lncRNA NEAT1, essential for paraspeckle assembly in mammals, is conserved in marsupials where it supports paraspeckle formation during late , indicating preserved architectural roles in across mammals. In evolutionary developmental (evo-devo) contexts, recent studies reveal lncRNAs in plants that mirror animal immunity by fine-tuning stress responses; for example, conserved lncRNA motifs in regulate pathogen defense and tolerance via epigenetic silencing, paralleling immune modulation in animals through shared mechanisms like . These findings highlight lncRNAs' adaptability in stress adaptation across kingdoms. Metrics of functional conservation often surpass sequence-level analyses, as demonstrated by phenotypic rescue experiments and network-level preservation. In cross-species assays, human lncRNA orthologs rescue knockout phenotypes in zebrafish models, such as restoring cellular proliferation defects, confirming functional equivalence despite sequence divergence. Network-level conservation is notably higher, with ceRNA (competing endogenous RNA) hubs—where lncRNAs act as miRNA sponges—preserved across tumors and species, as hub lncRNAs dominate regulatory interactions in cancer hallmarks, indicating robust evolutionary retention of interaction networks over individual sequences. These approaches reveal that lncRNA functions are under stronger selective constraint at the systems level, facilitating orthologous roles in development and disease.

Biological and Pathological Roles

Roles in Development and Physiology

Long non-coding RNAs (lncRNAs) play pivotal roles in developmental processes by facilitating essential mechanisms such as X-chromosome dosage compensation and . The lncRNA is crucial for X-chromosome inactivation in female mammals, where it coats the inactive to silence gene expression and equalize dosage between sexes. Similarly, Kcnq1ot1, a paternally expressed lncRNA within the Kcnq1 imprinting domain on chromosome 11p15.5, mediates epigenetic silencing of neighboring genes, ensuring proper fetal growth and development through monoallelic expression. In limb development, lncRNAs contribute to patterning by regulating key signaling pathways; for instance, the lncRNA Maenli acts as a limb-specific regulator of the En1, which in turn influences Fgf8 expression to guide anterior-posterior limb patterning. In physiological contexts, lncRNAs modulate immune responses and to maintain organismal . NEAT1 lncRNA organizes paraspeckles that sequester antiviral factors, enhancing the host's against viral infections such as hantavirus by promoting expression. Recent studies have also implicated NEAT1 in natural killer () cell function, where it can inhibit cytotoxicity in pathological contexts like by acting as a competing endogenous RNA (ceRNA) for miR-125 to upregulate MCEMP1, as highlighted in 2025 reviews on lncRNA roles in NK . In , lncRNAs aid and growth under abiotic challenges; for example, various lncRNAs regulate signaling and uptake to enhance tolerance to and , as summarized in 2023-2024 reviews. LncRNAs further contribute to tissue through metabolic and rhythmic regulation. The imprinted lncRNA H19 promotes insulin sensitivity and by activating the AMPK pathway, thereby fine-tuning systemic glucose . In circadian physiology, lncRNAs influence dynamics to sustain rhythmic . 2024 findings reveal oscillations in accessibility at core clock gene loci, establishing " clocks" for temporal . These functions often integrate within multi-lncRNA networks during . In cardiac development, the lncRNA coordinates a network of cardiovascular genes by associating with complexes, promoting the transition from multipotent to committed cardiac progenitors. Such networks underscore the broader physiological orchestration by lncRNAs, linking regulatory mechanisms to developmental and homeostatic outcomes.

Associations with Diseases

Long non-coding RNAs (lncRNAs) have been implicated in the of various human diseases through their dysregulation, which disrupts normal regulatory functions and contributes to disease progression. In cancer, lncRNAs can act as oncogenes or tumor suppressors, influencing tumor growth, , and therapy resistance. For instance, the lncRNA HOTAIR is upregulated in primary tumors and promotes by reprogramming through interactions with the Polycomb Repressive Complex 2 (PRC2), leading to epigenetic silencing of tumor suppressor genes. HOTAIR expression correlates with poorer metastatic-free and overall survival in patients. Similarly, HOTAIR drives progression by modulating multiple signaling pathways, including those involved in and invasion. Recent reviews highlight the role of lncRNAs like HOTAIR in shaping the , where they facilitate immune evasion and stromal interactions to support . In contrast, the lncRNA GAS5 functions as a tumor suppressor by inhibiting , migration, and invasion while promoting across multiple cancers, including , gastric, and carcinomas; its downregulation is associated with advanced disease stages and reduced patient survival. In cardiovascular diseases, lncRNAs contribute to vascular dysfunction and cardiac remodeling. The lncRNA ANRIL, located at the 9p21 locus, promotes by acting as a scaffold for PRC1 and PRC2 complexes, repressing genes involved in and , which leads to plaque formation and instability. Genetic variants in the ANRIL locus are strongly associated with risk. In , the lncRNA MIAT is upregulated and regulates myocardial and , making it a promising therapeutic target; recent studies from 2025 emphasize MIAT's role in pathological cardiac remodeling post-myocardial and its potential for siRNA-based interventions to improve cell viability. Neurological and immune-related diseases also feature lncRNA dysregulation with causal implications. In , the lncRNA BACE1-AS stabilizes BACE1 mRNA by forming RNA duplexes that prevent its degradation, thereby increasing β-secretase activity and amyloid-β production, which exacerbates neuronal damage. Elevated BACE1-AS levels have been observed in Alzheimer's patients and amyloid precursor protein transgenic models. For immune disorders, lncRNAs modulate natural killer () cell function and contribute to diseases like cancers and autoimmune conditions; a 2025 review details how lncRNAs such as MALAT1 and RMRP regulate NK cell , , and immune escape in tumor microenvironments, with dysregulation linked to impaired in NK cell-associated pathologies. LncRNAs serve as valuable biomarkers due to their stability and disease-specific expression patterns. Circulating lncRNAs, detectable in or , offer non-invasive diagnostic potential; for example, PCA3 is highly specific to and is FDA-approved for detecting high-grade tumors via urine assays, where its overexpression correlates with disease progression and signaling. Genome-wide association studies (GWAS) have identified disease-associated variants in lncRNA loci, with recent analyses indicating that a substantial proportion—estimated up to around 30%—of such variants fall within lncRNA regions due to expanded annotations, influencing susceptibility to cancers, cardiovascular diseases, and immune disorders through altered expression or structure.

Therapeutic Potential

Targeting Approaches

Antisense oligonucleotides (ASOs) represent a primary strategy for targeting long non-coding RNAs (lncRNAs) by inducing their degradation through recruitment of RNase H enzymes. Gapmer ASOs, which feature a central DNA segment flanked by modified nucleotides, hybridize to target lncRNA sequences and trigger cleavage by endogenous RNase H, effectively reducing lncRNA levels. For instance, LNA-gapmer ASOs targeting the oncogenic lncRNA MALAT1 have demonstrated inhibition of gene expression in proteasome subunits, leading to anti-proliferative effects in multiple myeloma models and advancing to preclinical evaluations for broader cancer applications. Locked nucleic acids (LNAs), incorporated into ASOs, enhance binding affinity and nuclease resistance, thereby improving stability and potency in vivo; LNA-modified gapmers have shown superior downregulation of nuclear lncRNAs compared to unmodified counterparts, with applications in silencing disease-associated lncRNAs like Xist. RNA interference (RNAi) approaches utilize small interfering RNAs (siRNAs) or short hairpin RNAs (shRNAs) to achieve sequence-specific degradation of lncRNA transcripts via the (RISC). These tools have been widely applied to knock down lncRNAs in cellular studies, revealing functional roles in gene regulation; for example, siRNAs targeting lncRNAs like NEAT1 effectively suppress paraspeckle formation and associated pathological processes. Recent advances in CRISPR-Cas13 systems further refine RNAi-like targeting by enabling precise and cleavage with high specificity, avoiding off-target DNA effects; 2024 developments in Cas13-based screens have identified essential lncRNAs through transcriptome-wide perturbations, demonstrating strand- and transcript-specific silencing in cancer cells. Small molecules offer an alternative for modulating lncRNA function by binding to specific structural motifs, such as triple helices or G-quadruplexes, thereby disrupting protein interactions or stability. Compounds targeting the triple helix in lncRNAs like MALAT1 have been shown to reduce RNA levels and inhibit downstream oncogenic signaling, providing a scaffold for structure-based drug design. For NEAT1, small molecules that bind G-quadruplex motifs interfere with NONO protein recruitment, destabilizing paraspeckle assembly and attenuating immune responses in disease models. A 2025 review has discussed the potential of small-molecule inhibitors targeting lncRNAs like CHRF to block fibrotic pathways by preventing lncRNA-mediated epigenetic changes in cardiac tissue. Gene therapy strategies employ viral vectors, such as (AAV), to deliver suppressors that overexpress RNAi components or decoy elements targeting lncRNAs. These approaches can be combined with miRNA mimics to enhance .

Challenges and Recent Advances

One major challenge in developing therapeutics targeting long non-coding RNAs (lncRNAs) is efficient , particularly due to their frequent localization, which complicates access for cytoplasmic-acting agents like small interfering RNAs (siRNAs). Antisense (ASOs) have shown greater in silencing lncRNAs compared to approaches, which perform better on cytoplasmic ones, highlighting the need for localization-specific strategies. Off-target effects further hinder progress, as non-specific binding can lead to unintended and toxicity. To address these, nanoparticles (LNPs) have emerged as a promising platform, with formulations like those incorporating ionizable lipids enabling liver-specific uptake via E-mediated , as demonstrated in mRNA therapeutics and adaptable to lncRNA modulators. Specificity remains a critical barrier, as lncRNAs often adopt complex secondary structures that impede binding of therapeutic agents, while isoform variability across tissues can result in incomplete or heterogeneous targeting. Additionally, synthetic RNA analogs risk activating innate immune responses through Toll-like receptors, leading to release and potential , which necessitates chemical modifications such as 2'-O-methylation to enhance stability and reduce . These issues underscore the importance of high-fidelity design tools to minimize off-target interactions and isoform-specific effects. Clinical translation remains primarily preclinical, with studies such as a 2024 investigation demonstrating that LNP-delivered targeting HOTAIR reduced tumorigenic properties in solid tumor models, suggesting potential for future evaluations in advanced cancers. In cardiovascular applications, 2025 preclinical successes include lncRNA MIR181A1HG silencing, which significantly attenuated atherosclerotic lesion burden in aortic models by modulating vascular inflammation, offering promise for plaque stabilization therapies. Emerging technologies are accelerating lncRNA therapeutic , including AI-driven models like graph neural networks that predict lncRNA-disease associations with high accuracy; for example, the 2025 LDA-GMCB integrates multi-head self-attention for scalable inference of novel links. Base editing strategies, particularly RNA-specific deaminases fused to guide RNAs, enable precise correction of lncRNA variants influencing stability and function, as evidenced by tools predicting editing impacts in databases like LNCediting. Insights from plant-derived lncRNAs are informing agrobiotech, where 2025 reviews highlight their roles in stress tolerance and , inspiring engineered crops with enhanced through lncRNA .

References

  1. [1]
    Long non-coding RNAs: definitions, functions, challenges ... - Nature
    Jan 3, 2023 · We address lncRNA definition, nomenclature, conservation, expression, phenotypic visibility, functional assays and molecular mechanisms ...
  2. [2]
  3. [3]
    Gene regulation by long non-coding RNAs and its biological functions
    Dec 22, 2020 · In this Review, we discuss the mechanisms of lncRNA ... Long non-coding RNAs: definitions, functions, challenges and recommendations.
  4. [4]
    Molecular Mechanisms and Function Prediction of Long Noncoding ...
    However, some lncRNAs are known to have ORFs longer than 100 codons, while some protein coding genes have fewer than 100 amino acids, such as RCI2A gene in ...Missing: minimal | Show results with:minimal
  5. [5]
    Long noncoding RNAs in biology and hematopoiesis | Blood
    By arbitrary definition, lncRNAs are greater than 200 nucleotides and represent a diverse group with many functions. ... Similar to protein-coding genes, many ...
  6. [6]
    Parental imprinting of the mouse H19 gene - Nature
    May 9, 1991 · Bartolomei,; Sharon Zemel &; Shirley M. Tilghman. Nature volume 351, pages 153–155 (1991)Cite this article. 3812 Accesses. 1083 Citations.
  7. [7]
    Conservation of position and exclusive expression of mouse Xist ...
    May 23, 1991 · We report here that the mouse Xist gene maps to the Xic region of the mouse X chromosome and, using an interspecific Mus spretus/Mus musculus domesticus F 1 ...
  8. [8]
    A short guide to long non-coding RNA gene nomenclature - PMC
    Long intergenic lncRNAs (lincRNAs) that lie between pc gene loci are named with a common root symbol (LINC, 'long intergenic non-coding RNA') and an iterated, ...
  9. [9]
    An integrated encyclopedia of DNA elements in the human genome
    Sep 5, 2012 · Comparing lncRNAs to other ENCODE data indicates that lncRNAs are generated through a pathway similar to that for protein-coding genes. The ...Missing: criteria | Show results with:criteria
  10. [10]
    The Landscape of Long Noncoding RNAs in the Human ...
    Over 68% (58,648) of genes were classified as lncRNAs, of which 79% (48,952) were previously unannotated. About 1% (597) of the lncRNAs harbored ultraconserved ...
  11. [11]
    Human Release 44 Statistics - GENCODE
    General stats ; Long non-coding RNA genes, 19922 ; Small non-coding RNA genes, 7566 ; Pseudogenes, 14735 ; - processed pseudogenes, 10660 ; - unprocessed pseudogenes ...Missing: abundance 2023
  12. [12]
    NONCODEV6: an updated database dedicated to long non-coding ...
    The number of human lncRNAs has increased from 172 216 to 173 112. The number of mouse lncRNAs increased from 131 697 to 131 974. The number of plant lncRNAs is ...
  13. [13]
    LncExpDB: an expression database of human long non-coding RNAs
    Oct 12, 2020 · Compared with mRNAs, particularly, lncRNAs feature higher tissue-specificity (12) and wider subcellular localizations (13), indicating their ...<|control11|><|separator|>
  14. [14]
    ATP-dependent chromatin remodeling shapes the long noncoding ...
    Similarly, 75%–85% of the genome of the budding yeast Saccharomyces cerevisiae is estimated to be transcribed (Nagalakshmi et al. 2008). Although lncRNAs are ...
  15. [15]
    Advances of Long Non-Coding RNA in Perennial Plants - MDPI
    This review systematically summarizes the classification and regulatory mechanisms of plant lncRNAs, with a focus on recent advances in research on their ...
  16. [16]
    Direct RNA sequencing in plants: Practical applications and future ...
    Nov 11, 2024 · Direct RNA sequencing (DRS) has emerged as a new and powerful sequencing platform for precise detection of native and full-length transcripts.
  17. [17]
    Post-transcriptional gene regulation by long noncoding RNA - NIH
    After transcription, most lncRNAs are processed like protein-coding RNAs, including 5'-end capping, 3'-end polyadenylation, splicing of introns, and ...
  18. [18]
    Distinct regulatory functions and biological roles of lncRNA splice ...
    Introns and exons of lncRNA genes are usually longer than those of PCGs. However, the average number of exons per lncRNA gene is lower than that of PCGs (3.02 ...
  19. [19]
    RNA polyadenylation patterns in the human transcriptome
    Mar 30, 2022 · While the main target of polyadenylation is mRNA, up to 70% of lncRNAs contain multiple PASs (Hu et al., 2017). LncRNAs are defined as non- ...
  20. [20]
    Sequence-dependent recruitment of SRSF1 and SRSF7 to ...
    Jun 7, 2021 · Sequence-dependent recruitment of SRSF1 and SRSF7 to intronless lncRNA NKILA promotes nuclear export via the TREX/TAP pathway Open Access.
  21. [21]
    Inefficient splicing of long non-coding RNAs is associated with ... - NIH
    Aug 6, 2023 · Recent reports show that long non-coding RNAs (lncRNAs) have inefficient splicing and fewer alternative splice variants than mRNAs.
  22. [22]
    Regulation of long non-coding RNAs and genome dynamics by the ...
    Feb 4, 2020 · In this Review, we describe the RNA surveillance factors and discuss the vital role of lncRNA surveillance in orchestrating various biological processes.
  23. [23]
    Genome-wide analysis of lncRNA stability in human - PMC
    Apr 16, 2021 · The average half-llife of lncRNAs and mRNAs was 3.96 h and 6.35 h, and the median was 2.76 h and 4.18 h, respectively. The Kolmogorov-Smirnov ...
  24. [24]
    Genome-wide analysis of long noncoding RNA stability - PMC
    LncRNA half-lives vary over a wide range, comparable to, although on average less than, that of mRNAs, suggestive of complex metabolism and widespread ...
  25. [25]
    HOTAIR forms an intricate and modular secondary structure - PMC
    Apr 9, 2015 · The HOTAIR structure reveals a degree of structural organization comparable to well-folded RNAs, like the group II intron, rRNA or lncRNA ...
  26. [26]
    Long Non-coding RNA Structure and Function: Is There a Link? - PMC
    Aug 24, 2018 · Using multiple mutagenesis assays, the secondary structures such as stem length, hairpin pairing, bulge size and position, and apical loop ...
  27. [27]
    Global profiling of hnRNP A2/B1-RNA binding on ... - PubMed - NIH
    We recently identified heterogeneous ribonucleoprotein (hnRNP) A2/B1 as an adaptor of the human HOTAIR lncRNA. hnRNP A2 and B1 are splice isoforms of the same ...
  28. [28]
    Molecular anatomy of the architectural NEAT1 noncoding RNA
    May 1, 2019 · NEAT1 possesses three distinct RNA domains (A, B, and C), which function in stabilization (A), isoform switching (B), and paraspeckle assembly (C).Missing: tertiary | Show results with:tertiary
  29. [29]
    Long noncoding RNA turnover - PMC - PubMed Central - NIH
    In this review, we summarize our current knowledge of the factors and mechanisms that modulate mammalian lncRNA stability.
  30. [30]
    The lncRNA HMS recruits RNA-binding protein HuR to stabilize the 3
    We show that HuR physically interacts with the cytosine-rich stretch of HMS and HOXC10 3′-UTR to stabilize HOXC10 mRNA.
  31. [31]
    Mechanisms of Long Noncoding RNA Nuclear Retention - PubMed
    Recent studies have revealed the complex mechanisms that govern lncRNA nuclear retention. We review current understanding of how the transcription and ...
  32. [32]
    LncRNA–Protein Interactions: A Key to Deciphering LncRNA ... - NIH
    Jun 17, 2025 · We delve into the interactions between lncRNAs and RNA-binding proteins (RBPs), explore their implications in cellular processes, and examine bioinformatic and ...
  33. [33]
    Interconnections between m6A RNA modification, RNA structure ...
    Nov 7, 2023 · Overall, the m6A modification leads to subtle changes in the RNA folding kinetics that result in small changes in the local RNA structure.
  34. [34]
    Thermodynamic insights into N6-methyladenosine-modified ...
    Using naturally occurring m6A-modified RNAs, including lncRNA MALAT1, we examined the effects of m6A on RNA folding and protein binding. Biophysical experiments ...
  35. [35]
    On the classification of long non-coding RNAs - PMC
    Here, we summarize classification methods of lncRNAs according to their four major features, namely, genomic location and context, effect exerted on DNA ...
  36. [36]
    Long non-coding RNAs in humans: Classification, genomic ...
    Dec 6, 2024 · LncRNAs are categorized into sense, antisense, bidirectional, intronic and intergenic lncRNAs based on their genomic location [2]. The ...
  37. [37]
    Long noncoding RNAs in B-cell development and activation | Blood
    LncRNA genomic locations have the potential to provide insight into function. A substantial proportion of multiexon (48%) and single-exon (48%) loci were ...<|separator|>
  38. [38]
    A Bidirectional Non-Coding RNA Promoter Mediates Long-Range ...
    This work studies the long-range gene expression regulatory role of a long non-coding RNA gene promoter using chromosome conformation capture methods.
  39. [39]
    Topologically associating domain underlies tissue specific ... - NIH
    Apr 26, 2023 · In this study, we demonstrated that lincRNA loci are significantly enriched within TAD regions and their localization is associated with tissue ...Missing: syntenic | Show results with:syntenic
  40. [40]
    Genomic positional conservation identifies topological anchor point ...
    Mar 15, 2018 · Syntenic lncRNAs were defined as positionally conserved if their promoters were genomically associated with orthologous genes and produced ...
  41. [41]
    Volatile evolution of long noncoding RNA repertoires
    We explore the molecular mechanisms promoting the birth and rapid evolution of lncRNA genes with an emphasis on the influence of bidirectional transcription ...
  42. [42]
    The lncRNA DUBR is regulated by CTCF and coordinates chromatin ...
    Feb 24, 2025 · Additionally, analysis of Hi-C data revealed that the CTCF-BS was located close to a topologically associating domain (TAD) boundary ...
  43. [43]
    Comparative genomic analyses highlight the contribution of ... - NIH
    Oct 16, 2017 · Moreover, 19% of these lincRNAs have overlaps with or are close to pseudogenes in the human genome. Conclusions. We suggest that a notable ...
  44. [44]
    an updated database for functional variants in long non-coding RNAs
    Nov 9, 2022 · lncRNASNP is dedicated to providing a comprehensive repository of single nucleotide polymorphisms (SNPs) and somatic mutations in lncRNAs and their impacts on ...Missing: architecture | Show results with:architecture
  45. [45]
    Long non-coding RNA-encoded micropeptides - Nature
    Oct 23, 2024 · This review aims to elucidate the functions of lncRNA-encoded micropeptides and explore their potential applications as therapeutic targets in cancer.
  46. [46]
    Alternative ORFs and small ORFs: shedding light on the dark ...
    Aug 29, 2019 · The uORF-encoded protein can stall the ribosome due to interactions between the nascent peptides and the ribosome exit channel, resulting in the ...
  47. [47]
    Translation and natural selection of micropeptides from long non ...
    Oct 31, 2022 · We show that around 30% of lncRNAs contain small ORFs engaged by ribosomes, leading to regulated translation of 100 to 300 micropeptides.
  48. [48]
    A lncRNA-encoded mitochondrial micropeptide exacerbates ...
    Feb 15, 2023 · The functions of microglia include immune monitoring, regulating immune response, phagocytosis, synaptic pruning, maintaining synaptic structure ...
  49. [49]
    LncPepAtlas: a comprehensive resource for exploring the ...
    Oct 22, 2024 · Mass spectrometry validated lncRNA-encoded peptides. We have downloaded all available MS datasets from the PRIDE database, a comprehensive ...
  50. [50]
    The tissue-specific lncRNA Fendrr is an essential regulator of heart ...
    We illustrate that Fendrr acts by modifying the chromatin signatures of genes involved in the formation and differentiation of the lateral mesoderm lineage ...Missing: looping | Show results with:looping
  51. [51]
    Transcriptional regulatory functions of nuclear long noncoding RNAs
    More recently, lncRNAs have been demonstrated to interact with chromatin at several thousand different locations across multiple chromosomes and to modulate ...
  52. [52]
    RNA in unexpected places: long non-coding RNA functions in ... - NIH
    Oct 9, 2013 · lncRNAs can bind Pol II directly to inhibit transcription (part d). Formation of lncRNA-DNA triplex structures can also inhibit the assembly of ...Lncrnas As Regulators Of... · Figure 2. Lncrnas Regulate... · Regulators Of Mrna...Missing: percentage | Show results with:percentage
  53. [53]
    7SK RNA, a non-coding RNA regulating P-TEFb, a ... - PubMed
    7SK RNA is an abundant 331 nt nuclear transcript generated by RNA polymerase III. Binding of 7SK RNA to HEXIM1/2 turns these proteins into inhibitors of P-TEFb.Missing: scaffolding | Show results with:scaffolding
  54. [54]
    Enhancer RNAs: mechanisms in transcriptional regulation and ...
    Aug 3, 2023 · Facilitating RNA Pol II pause-release to enhance transcription extension. RNA Pol II pause is a genome-wide regulatory mechanism of higher ...Functions Of Ernas · Facilitating Rna Pol Ii... · Regulation Of Ernas...
  55. [55]
    Noncoding RNAs regulate alternative splicing in Cancer
    Jan 6, 2021 · In this review, we summarize the current mechanisms by which ncRNAs regulate AS in cancers and discuss their potential clinical applications as biomarkers and ...
  56. [56]
    Full article: The nexus of long noncoding RNAs, splicing factors ...
    Nov 28, 2023 · This review focuses on the regulation of AS by lncRNAs through their interaction with splicing factors.
  57. [57]
    PTEN, PTENP1, microRNAs, and ceRNA Networks - MDPI
    One of the most contemporary findings in PTEN regulation is the post-transcriptional regulation by its pseudogene long non-coding RNA (lncRNA) and microRNA ( ...
  58. [58]
    Pseudogene PTENP1 Functions as a Competing Endogenous RNA ...
    Here, we report that pseudogene PTENP1 serves as a ceRNA to modulate PTEN expression regulation by miR21. PTENP1 suppresses tumor growth, invasion, and ...
  59. [59]
    lncRNAs transactivate Staufen1-mediated mRNA decay by ... - NIH
    Aug 10, 2011 · Staufen1 (STAU1)-mediated mRNA decay (SMD) degrades translationally active mRNAs that bind the double-stranded (ds)RNA binding protein STAU1 ...
  60. [60]
    Assessing the ceRNA Hypothesis with Quantitative Measurements ...
    Jun 5, 2014 · Recent studies have reported that competitive endogenous RNAs (ceRNAs) can act as sponges for a microRNA (miRNA) through their binding sites ...Missing: overlap | Show results with:overlap
  61. [61]
    Many lncRNAs, 5'UTRs, and pseudogenes are translated and ... - eLife
    Dec 19, 2015 · In this regard, lncRNAs are transcribed by RNA polymerase II, capped, and polyadenylated, and hence are largely indistinguishable from mRNAs ...
  62. [62]
    The long noncoding RNA CARDINAL attenuates cardiac ... - JCI
    May 14, 2024 · These data suggested that CARDINAL might regulate ribosome stalling in cardiac hypertrophy. Previously, multiple conserved amino acid motifs ...
  63. [63]
    TERRA recruitment of polycomb to telomeres is essential for histone ...
    Apr 18, 2018 · We find that 20q-TERRAs are essential for the establishment of H3K9me3, H4K20me3, and H3K27me3 heterochromatin marks at telomeres.
  64. [64]
    Epitranscriptomic advances in normal and malignant hematopoiesis
    Sep 19, 2025 · RBFOX2 binds m⁶A-marked chromatin-associated RNAs (caRNAs), recruiting PRC2 and promoting focal H3K27me3 deposition. TET2 loss increases m⁵C ...
  65. [65]
    HOTAIR requires epitranscriptomic modification to exert its pivotal ...
    Oct 24, 2025 · While m6A epitranscriptomic modification has been shown to impact several mRNAs maturation, stability/degradation, nuclear/cytoplasm export ...
  66. [66]
    ASAR lncRNAs control DNA replication timing through interactions ...
    Jun 18, 2024 · ASARs are a family of very-long noncoding RNAs that control replication timing on individual human autosomes, and are essential for chromosome stability.
  67. [67]
    Epigenetic control of chromosome-associated lncRNA genes ...
    Oct 22, 2022 · ASARs are long noncoding RNA genes that control replication timing of entire human chromosomes in cis. The three known ASAR genes are ...
  68. [68]
    TERRA long noncoding RNA: At the interphase of telomere damage ...
    TERRA long noncoding RNAs play key roles in telomere function and maintenance. They can orchestrate telomeric chromatin remodeling, regulate telomere ...
  69. [69]
    Telomeric repeat-containing RNA TERRA: a noncoding ... - Frontiers
    Emerging evidence indicate that TERRA transcripts form DNA-RNA hybrids at chromosome ends which can promote homologous recombination among telomeres, delaying ...
  70. [70]
    LncRNA CCTT-mediated RNA-DNA and RNA-protein interactions ...
    Nov 3, 2022 · LncRNA CCTT-mediated RNA-DNA and RNA-protein interactions facilitate the recruitment of CENP-C to centromeric DNA during kinetochore assembly.
  71. [71]
    Noncoding RNA NORAD regulates genomic stability by ... - NIH
    NORAD maintains genomic stability by sequestering PUMILIO proteins, which repress the stability and translation of messenger RNAs to which they bind.
  72. [72]
    Inverted repeat Alu elements in the human lincRNA-p21 adopt a ...
    Jul 4, 2016 · We sequenced the human lincRNA-p21 (hLincRNA-p21) and found that it has a single exon that includes inverted repeat Alu elements (IRAlus).
  73. [73]
    The chromatin-associated lncREST ensures effective replication ...
    Feb 1, 2024 · LncREST-depleted cells experience sustained replication fork progression and accumulate un-signaled DNA damage.
  74. [74]
    ATR/Chk1 interacting lncRNA modulates DNA damage response to ...
    Moreover, the speed of replication fork prolongation was significantly suppressed in ACIL knockdown cells upon HU exposure (Fig. 6B). Under replication stress, ...
  75. [75]
    The evolution of lncRNA repertoires and expression patterns in ...
    Jan 19, 2014 · Here we present a large-scale evolutionary study of lncRNA repertoires and expression patterns, in 11 tetrapod species.
  76. [76]
    Long noncoding RNA genes: conservation of sequence and brain ...
    Conservation of lncRNA sequence​​ Nevertheless, and in contrast to coding sequence, exonic conservation was observed to be <30% and was as low as 4% (for Rmst) ...
  77. [77]
    A subset of conserved mammalian long non-coding RNAs are ...
    Aug 30, 2017 · We estimate that ~ 55 annotated conserved human lncRNAs are derived from parts of ancestral protein-coding genes, and loss of coding potential ...Missing: minimal | Show results with:minimal
  78. [78]
    Evolutionary conservation of long noncoding RNAs - PubMed Central
    Here, we review the extent of interspecies conservation among different lncRNAs, with a focus on a subset of lncRNAs that have been functionally investigated.
  79. [79]
    Transcriptomic analyses reveal groups of co-expressed, syntenic ...
    We classified 20% of the lncRNAs (1,532 out of 7,635) into syntenic families, which is a much lower rate than for protein-coding genes (50.3%). This result ...
  80. [80]
    Multidimensional conservation analysis decodes the expression of ...
    Apr 5, 2023 · The conserved and non-conserved lncRNAs exhibit a significant difference in their abundance, transcript number, tissue specificity, etc.
  81. [81]
    Ultra-conserved RNA: a novel biological tool with diagnostic and ...
    Apr 10, 2023 · Ultra-conserved RNA (ucRNA) is a subset of long non-coding RNA, that is highly conserved among mice, rats and humans.
  82. [82]
    Dosage compensation: A new player in X chromosome upregulation
    Oct 24, 2022 · In marsupials, imprinted XCI (iXCI) of the paternal X is triggered by lncRNA Rsx, which collaborates with XCU regulated to maintain an X:A ratio ...
  83. [83]
    Structurally Conserved Primate LncRNAs Are Transiently Expressed ...
    Jan 10, 2019 · In this article, Salama and colleagues identified transiently expressed (TrEx) lncRNAs from human, chimpanzee, orangutan, and rhesus pluripotent ...
  84. [84]
    De novo genes with an lncRNA origin encode unique human brain ...
    Jan 2, 2023 · Human de novo genes can originate from neutral long non-coding RNA (lncRNA) loci and are evolutionarily significant in general.
  85. [85]
    The lncRNA hsrω regulates arginine dimethylation of human FUS to ...
    Oct 23, 2019 · The lncRNA hsrω is known to interact with and organize several RBPs like the Drosophila FUS (Cabeza), TDP-43 (TBPH), hnRNPAB (Squid) and ...Results · The Lncrna Hsrω Is A Potent... · The Lncrna Hsrω Regulates...
  86. [86]
    Functional conservation of the lncRNA NEAT1 in the ancestrally ...
    We show that (i) NEAT1 is paraspeckle-associated in opossum (ii) NEAT1 expression is strongly induced in late gestation in uterine/placental extracts.
  87. [87]
    From environmental responses to adaptation: the roles of plant ...
    Apr 30, 2024 · LncRNAs can positively or negatively modulate stress responses, affecting processes such as hormone signaling, temperature tolerance, and nutrient deficiency ...Missing: animal immunity
  88. [88]
    Computational prediction and experimental validation identify ...
    Jan 9, 2024 · Here we present lncRNA Homology Explorer (lncHOME), a computational pipeline that identifies a unique class of long noncoding RNAs (lncRNAs) with conserved ...
  89. [89]
    Comprehensive characterization of lncRNA-mRNA related ceRNA ...
    Network hub analysis suggest that conserved hub lncRNAs may dominate different cancer hallmarks across various tumors. Network dynamic analysis found that a ...Missing: conservation | Show results with:conservation
  90. [90]
    Long noncoding RNA XIST: Mechanisms for X chromosome ... - NIH
    A growing body of evidence has revealed that the lncRNA XIST, an important regulator in X chromosome dosage compensation in placental mammals, can play pivotal ...
  91. [91]
    The long noncoding RNA Kcnq1ot1 organises a lineage-specific ...
    The paternally expressed long noncoding RNA (ncRNA) Kcnq1ot1 regulates epigenetic gene silencing in an imprinted gene cluster in cis over a distance of 400 kb ...
  92. [92]
    Combinatorial effects on gene expression at the Lbx1/Fgf8 locus ...
    Feb 10, 2022 · Fgf8 is required for outgrowth and patterning of the limbs. Nat ... Non-coding deletions identify Maenli lncRNA as a limb-specific En1 regulator.
  93. [93]
    The Long Noncoding RNA NEAT1 Exerts Antihantaviral Effects by ...
    Apr 13, 2017 · In this study, we identified the lncRNA NEAT1 as a vital antiviral modulator. NEAT1 was dramatically upregulated after Hantaan virus (HTNV) infection.
  94. [94]
    The role of long non-coding RNAs in NK cell biology and diseases
    Aug 25, 2025 · Long non-coding RNAs (lncRNAs) are pivotal regulators of gene expression, increasingly recognized for their roles in immune responses and ...
  95. [95]
    Current perspectives of lncRNAs in abiotic and biotic stress ...
    Jan 8, 2024 · We highlight the unique role of lncRNAs in integrating plant response to adverse environmental conditions with different aspects of plant growth and ...
  96. [96]
    H19 lncRNA Promotes Skeletal Muscle Insulin Sensitivity in Part by ...
    Sep 10, 2018 · Skeletal muscle plays a pivotal role in regulating systemic glucose homeostasis in part through the conserved cellular energy sensor AMPK.
  97. [97]
    Circadian-driven tissue specificity is constrained under caloric ...
    Jun 20, 2024 · Long noncoding RNAs (lncRNAs) are a particularly noteworthy class, but their regulatory role in circadian rhythms remains incompletely ...
  98. [98]
    Long non-coding RNAs: roles in cellular stress responses and ... - NIH
    lncRNAs can modulate gene expression and participate in chromatin remodeling, thereby influencing the cellular response to DNA damage. This modulation is ...Lncrnas In Cellular Stress... · Lncrnas And Genotoxic Stress · Lncrna Questions Outstanding...
  99. [99]
    Drugging the lncRNA MALAT1 via LNA gapmeR ASO inhibits gene ...
    Drugging the lncRNA MALAT1 via LNA gapmeR ASO inhibits gene expression of proteasome subunits and triggers anti-multiple myeloma activity.
  100. [100]
    Therapeutic Targeting of Long Non-Coding RNAs in Cancer - NIH
    One of the major advances in ASO chemistry is the development of locked nucleic acids (LNA) and S-constrained ethyl (cEt) modifications, which have ...
  101. [101]
    Locked nucleic acids (LNAs) reveal sequence requirements ... - PNAS
    We target LNAs at Xist RNA and show displacement from the X chromosome with fast kinetics. Xist transcript stability is not affected.
  102. [102]
    Long Non-Coding RNA Targeting and Transcriptional De-Repression
    RNAi is best known for the work demonstrating that ∼22 nucleotide small interfering RNA (siRNAs) or microRNAs (miRNAs) could be used to functionally target and ...
  103. [103]
    Discovering the crucial function of long noncoding RNAs - Nature
    Dec 11, 2024 · They use the RNA-targeting Cas13 nuclease to develop a transcriptome-wide CRISPR perturbation screen that targets lncRNA, and identify 778 ...
  104. [104]
    Article Transcriptome-scale RNA-targeting CRISPR screens reveal ...
    Dec 26, 2024 · We have developed RNA-targeting CRISPR screens to systematically perturb lncRNAs on a transcriptome scale with transcript- and strand-specificity.
  105. [105]
    Selective Small-Molecule Targeting of a Triple Helix Encoded ... - NIH
    First, the RNA structure itself can be disrupted (e.g., compound 16) to reduce MALAT1 levels. Second, small-molecule binding may serve to inhibit other ...
  106. [106]
    G-quadruplexes offer a conserved structural motif for NONO ...
    Jun 4, 2020 · The long non-coding RNA NEAT1 serves as a scaffold for the assembly of paraspeckles, membraneless nuclear organelles involved in gene regulation ...
  107. [107]
    LncRNA CHRF: molecular mechanisms and therapeutic potentials ...
    Jun 19, 2025 · This mechanistic insight establishes a solid foundation for designing CHRF-directed small-molecule inhibitors. 5 Conclusion and discussion.2.1 Lncrna Chrf In... · 2.2 Lncrna Chrf In Cancers · 2.3 Lncrna Chrf In Fibrotic...
  108. [108]
    Long Non-coding RNA Based Therapy for Cardiovascular Disease
    Sep 3, 2025 · One of the most relevant and widely studied lncRNAs in CVDs is MIAT, an intergenic lncRNA located on chromosome 22. It has been reported that ...
  109. [109]
    Therapeutic potential of synthetic microRNA mimics based on the ...
    Mar 22, 2025 · These results suggest that mimics based on the consensus sequence of the miR-15/107 group have therapeutic potential in a range of cancer types.
  110. [110]
    A combinatorial microRNA therapeutics approach to suppressing ...
    MicroRNA mimics are excellent therapeutic candidates because of their ability to repress multiple oncogenic pathways at once. Here we treated the aggressive ...