Fact-checked by Grok 2 weeks ago

Non-coding RNA

Non-coding RNAs (ncRNAs) are a diverse class of RNA molecules transcribed from the genome that do not encode proteins, instead serving essential regulatory roles in gene expression, epigenetic modifications, and cellular architecture across all domains of life. These molecules constitute the vast majority of eukaryotic transcriptional output, with approximately 98% of human genome transcription producing ncRNAs rather than protein-coding messenger RNAs (mRNAs). Unlike mRNAs, ncRNAs function primarily through their structural and sequence-specific interactions with DNA, RNA, or proteins to modulate biological processes from development to disease. ncRNAs are broadly classified into two main categories based on length: small ncRNAs, typically under 200 , and long ncRNAs (lncRNAs), exceeding 200 . Small ncRNAs include microRNAs (miRNAs), which silence target mRNAs via degradation or translational repression; small interfering RNAs (siRNAs), involved in ; Piwi-interacting RNAs (piRNAs), crucial for transposon silencing in cells; and small nucleolar RNAs (snoRNAs), which guide modifications on other RNAs. lncRNAs, such as Xist (which coats the to initiate dosage compensation in females) and H19 (implicated in ), often exhibit tissue-specific expression and participate in complex regulatory networks by recruiting chromatin-modifying complexes or forming nuclear condensates. The functional versatility of ncRNAs has profoundly shaped eukaryotic complexity, enabling fine-tuned control over transcription, post-transcriptional processing, and genome organization. For instance, miRNAs regulate up to 60% of human genes by binding to 3' untranslated regions of mRNAs, while lncRNAs like MALAT1 influence and cancer progression through interactions with nuclear speckles. Dysregulation of ncRNAs is linked to numerous diseases, including cancers (e.g., overexpression of oncogenic lncRNAs), neurological disorders (e.g., lncRNA involvement in Alzheimer's), and genetic syndromes (e.g., miRNA mutations in congenital defects). Historically, ncRNAs were first recognized in the 1960s with the identification of transfer RNAs () and ribosomal RNAs () as non-coding components of the translation machinery, but their regulatory potential emerged prominently in the 1990s with the discovery of lin-4 miRNA in C. elegans. Advances in high-throughput sequencing have since revealed thousands of ncRNA loci, highlighting their evolutionary conservation and therapeutic promise, such as using antisense oligonucleotides to target disease-associated ncRNAs.

Definition and Classification

Definition and Characteristics

Non-coding RNAs (ncRNAs) are functional transcripts that do not encode proteins but instead perform diverse regulatory roles within cells. These molecules represent the majority of the eukaryotic , accounting for approximately 98% of all transcriptional output in humans. Unlike protein-coding messenger RNAs (mRNAs), ncRNAs lack substantial open reading frames (ORFs) and thus evade ribosomal translation. A defining feature of ncRNAs is their structural and functional diversity, spanning a broad size range from small ncRNAs under 200 to long ncRNAs over 200 in length. Many ncRNAs achieve enhanced through chemical modifications, such as 2'-O-methylation, which rigidifies the sugar conformation and protects against degradation. In contrast to mRNAs, which are typically equipped with a 5' 7-methylguanosine and a 3' poly-A tail to , , and , ncRNAs frequently omit these elements, enabling their direct participation in cellular rather than serving as protein templates. Evolutionary analyses reveal that numerous ncRNAs exhibit conservation of sequence and secondary structure across species, signaling their essential biological roles. In humans, this abundance is exemplified by over 100,000 long ncRNAs (with GENCODE annotating approximately 35,900 lncRNA genes and over 170,000 transcripts as of 2025) alongside approximately 2,654 mature microRNAs (as per miRBase v22, the latest release), highlighting the genomic scale of non-coding transcription.

Major Types of ncRNAs

Non-coding RNAs (ncRNAs) exhibit remarkable diversity, primarily categorized by their length, structure, and biogenesis pathways into small ncRNAs (generally under 200 ), long ncRNAs (over 200 ), circular RNAs, and other specialized classes. This classification highlights their varied roles in cellular processes, though detailed functions are explored elsewhere. Small ncRNAs, often involved in precise regulatory mechanisms, include several well-defined subclasses. MicroRNAs (miRNAs) are single-stranded RNAs approximately 21-25 in length that primarily mediate post-transcriptional by binding to target mRNAs. Small interfering RNAs (siRNAs), similar in size to miRNAs, arise from either endogenous double-stranded precursors or exogenous sources like RNAs and facilitate to degrade target transcripts. PIWI-interacting RNAs (piRNAs), typically 24-32 long, are predominantly expressed in cells where they associate with proteins to silence transposable elements and maintain stability. Small nucleolar RNAs (snoRNAs), ranging from 60 to 300 , guide chemical modifications such as and pseudouridylation on ribosomal RNAs (rRNAs) and other stable RNAs within the . Long non-coding RNAs (lncRNAs), exceeding 200 , represent a heterogeneous group transcribed mainly by and implicated in broad regulatory processes like and cellular signaling. Subclasses include enhancer RNAs (eRNAs), which are short, bidirectional transcripts from enhancer regions that promote , and promoter-associated RNAs, which influence transcriptional initiation at promoter sites. Circular RNAs (circRNAs) form covalently closed, single-stranded loops through back-splicing of pre-mRNA exons, rendering them highly stable due to resistance to degradation. These structures, often abundant in eukaryotic cells, can function as molecular sponges sequestering miRNAs and thereby modulating their availability. Among other ncRNAs, transfer RNAs (tRNAs) and ribosomal RNAs (rRNAs) serve essential housekeeping functions in protein synthesis, with tRNAs decoding mRNA codons and rRNAs forming the core of ribosomes. Ribozymes, catalytic ncRNAs capable of self-cleavage or , exemplify RNA's enzymatic potential and are found in various biological contexts, including viral genomes and cellular processing pathways. Biogenesis of ncRNAs varies by class; for instance, miRNAs and siRNAs typically require the endonuclease for processing double-stranded precursors into mature forms, while most lncRNAs and miRNA primary transcripts are generated via transcription. Recent discoveries have expanded the ncRNA repertoire to include emerging classes such as tRNA-derived small RNAs (tsRNAs), which are fragments cleaved from mature or precursor tRNAs and regulate independently of the . Similarly, small RNA fragments like ribosomal RNA-derived small RNAs (srRNAs) have been identified as functional regulators in stress responses and development.

History of Discovery

Early Observations

The earliest observations of non-coding RNAs emerged in the mid-20th century during investigations into protein synthesis, revealing RNA molecules that did not encode proteins but were indispensable for translation. In the 1950s, proposed the adaptor hypothesis, suggesting the existence of molecules that would serve as intermediaries to link to the nucleotide sequence of , thereby facilitating accurate polypeptide assembly without themselves being translated. Concurrently, (rRNA) was identified as a key structural and functional component of ribosomes, the cellular machinery for protein synthesis, with early characterizations in the 1930s and 1940s confirming its non-coding nature as it constituted the bulk of ribosomal mass without contributing to the amino acid sequence. These discoveries established tRNA and rRNA as essential non-translated RNAs, laying the groundwork for recognizing RNA's multifaceted roles beyond mere informational transfer. Key experiments in the 1960s further illuminated the non-coding properties of tRNAs. The first complete nucleotide sequence of yeast alanine tRNA, determined by Robert W. Holley and colleagues in 1965, demonstrated a cloverleaf secondary structure with no capable of encoding a protein, confirming its role solely as an adaptor in . This sequencing effort, which involved enzymatic and chromatographic analysis of fragments, provided direct evidence that tRNAs functioned through base-pairing with mRNA and attachment sites, rather than into polypeptides. Early studies on lambda also hinted at regulatory RNAs, with observations of transcription patterns suggesting non-coding transcripts that modulated during viral development, though their mechanisms remained unclear at the time. The 1970s and early 1980s brought pivotal insights into catalytic capabilities of non-coding RNAs, transforming perceptions of RNA functionality. Sidney Altman's group identified ribonuclease P (RNase P) in as an enzyme processing tRNA precursors, with a 1978 study revealing that its RNA subunit was essential for catalytic activity, marking the first demonstration of RNA acting as a biocatalyst independent of protein. Shortly thereafter, in 1982, Thomas Cech's laboratory reported self-splicing of the Tetrahymena ribosomal RNA intron, where the RNA itself excised the intervening sequence without protein assistance, establishing the concept of ribozymes and expanding RNA's enzymatic repertoire. By the 1980s, additional classes of non-coding RNAs were recognized for their regulatory roles in eukaryotic and prokaryotic systems. Small nuclear RNAs (snRNAs), first isolated in the 1960s but functionally characterized in the early 1980s, were shown to form ribonucleoprotein particles (snRNPs) integral to the , as evidenced by experiments linking U1 snRNP binding to splice sites in heterogeneous nuclear RNA. In bacteria, antisense RNAs emerged as regulators, with early 1980s discoveries in plasmids and phages demonstrating their ability to base-pair with target mRNAs to inhibit translation or promote degradation, as seen in systems like the ColIb . These findings prompted a conceptual shift in , moving away from viewing non-coding RNAs as inert "" byproducts—echoing earlier dismissals of non-genic transcripts—to acknowledging them as active, functional molecules that challenged the central dogma's protein-centric focus. The discoveries, in particular, underscored RNA's ancient catalytic potential, suggesting it predated proteins in evolutionary history and broadening the scope of genetic regulation.

Modern Advances and Milestones

The discovery of the first (miRNA), lin-4, in 1993 by Victor Ambros and Gary Ruvkun in the nematode marked a pivotal moment in ncRNA research, demonstrating its role in post-transcriptional regulation through antisense complementarity to the 3' untranslated region of the lin-14 mRNA. This finding was recognized with the 2024 Nobel Prize in Physiology or Medicine. This finding challenged the prevailing view that regulation primarily occurred at the transcriptional level and opened avenues for understanding small ncRNAs as temporal regulators of development. In the 2000s, the revealed that only about 1-2% of the encodes proteins, prompting intensive efforts to characterize the vast non-coding portions and their functional RNAs. This genomic insight spurred hunts for ncRNAs, highlighting their abundance and potential regulatory roles beyond protein-coding genes. Concurrently, the elucidation of (siRNA)-mediated (RNAi) by and in 1998, recognized with the , demonstrated how double-stranded RNAs could trigger sequence-specific , revolutionizing tools for ncRNA functional studies. The 2010s saw the ENCODE project's 2012 phase annotate over 9,000 long non-coding RNA (lncRNA) loci through integrative analysis of chromatin states, transcription, and RNA expression, establishing lncRNAs as a major class of functional ncRNAs with tissue-specific patterns. This effort, combined with GENCODE annotations, cataloged thousands of lncRNAs, revealing their evolutionary conservation and association with regulatory elements. Simultaneously, high-throughput RNA sequencing (RNA-seq) in the 2010s fueled a discovery boom for circular RNAs (circRNAs), identifying them as abundant, stable transcripts formed by back-splicing, often derived from exons and enriched in neural tissues. Seminal studies using RNA-seq detected tens of thousands of circRNAs across species, underscoring their prevalence and potential as miRNA sponges or regulatory elements. In the 2020s, single-cell has illuminated ncRNA dynamics during , capturing heterogeneous expression patterns of miRNAs, lncRNAs, and circRNAs at cellular resolution to reveal stage-specific regulatory networks in processes like embryogenesis and . For instance, analyses in have shown lncRNAs modulating cell fate transitions through precise spatiotemporal expression. CRISPR-based of ncRNAs advanced significantly, with 2023 studies developing targeted knockouts for lncRNAs that minimize off-target effects on nearby genes, enabling functional validation of their roles in cellular processes. Technological advances have further propelled ncRNA research, including deep sequencing for comprehensive profiling and sequencing (CLIP-seq) variants like iCLIP, which map protein-ncRNA binding sites at resolution to uncover networks. By 2024, integrations, such as 3 adaptations for , have enabled accurate predictions of ncRNA secondary and tertiary structures, facilitating modeling of their interactions with proteins and other molecules without relying solely on experimental data. Recent advances in epitranscriptomics from 2024 to 2025 have further illuminated the role of N6-methyladenosine (m6A) modifications on ncRNAs, influencing their , localization, and interactions, as evidenced by studies showing m6A readers like YTHDF2 promoting lncRNA to fine-tune . These modifications integrate ncRNAs into dynamic regulatory circuits, with recent mappings revealing m6A sites on circRNAs that affect their biogenesis and function in stress responses.

Biogenesis and Structural Features

Transcription and Processing Mechanisms

Non-coding RNAs (ncRNAs) are transcribed by the three major eukaryotic RNA polymerases, with specificity determined by promoter elements and the functional class of the ncRNA. Ribosomal RNAs (rRNAs) are primarily synthesized by (Pol I) in the , producing large precursor transcripts that undergo extensive processing to form mature 18S, 5.8S, and 28S rRNAs. Transfer RNAs (tRNAs) and small nuclear RNAs (snRNAs) are typically transcribed by (Pol III), which generates short transcripts with internal promoters and terminates at poly-T sequences, resulting in uncapped, non-polyadenylated RNAs. In contrast, microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are predominantly transcribed by (Pol II), incorporating a 5' 7-methylguanosine cap and often a 3' poly-A tail, similar to messenger RNAs (mRNAs), which enhances their stability and nuclear export. Processing of ncRNAs varies by length and class, with small ncRNAs generally requiring endonucleolytic cleavage for maturation, while long ncRNAs follow mRNA-like pathways but exhibit greater diversity. For miRNAs, the primary transcript (pri-miRNA) is cleaved in the by the Drosha-DGCR8 complex to form the precursor miRNA (pre-miRNA), a ~70-nucleotide , which is then exported to the and further processed by into a ~22-nucleotide mature miRNA duplex that loads into the protein of the . LncRNAs undergo splicing by the akin to mRNAs, involving recognition of 5' and 3' splice sites, formation, and ligation, though many retain introns or display patterns that contribute to isoform diversity and functional specificity. Small ncRNAs like tRNAs and snRNAs are processed through endonucleolytic trimming by RNase P and Z for tRNAs, or via the SMN complex for snRNA assembly into ribonucleoproteins, whereas lncRNAs are often polyadenylated by the canonical cleavage and specificity factor (CPSF) complex to promote stability and prevent degradation. Certain ncRNA classes guide post-transcriptional modifications on target RNAs, including rRNAs and snRNAs. C/D box small nucleolar RNAs (snoRNAs) direct 2'-O-methylation of ribose moieties via base-pairing with target RNAs, recruiting the fibrillarin (FBL) methyltransferase core protein; for example, the U85 snoRNA guides methylation at C45 of U5 snRNA. H/ACA box snoRNAs facilitate pseudouridylation by forming a four-way junction structure that positions the dyskerin (DKC1) complex to isomerize uridines, as seen in modifications of 18S rRNA at multiple sites. These modifications stabilize RNA structures and fine-tune ribosome function, with over 100 such sites identified in human rRNAs. Circular RNAs (circRNAs), a subclass of lncRNAs, form through back-splicing mechanisms, including lariat intermediates from spliceosomal intron excision or direct intron pairing via complementary Alu repeats, which juxtapose splice sites and yield covalently closed loops resistant to exonucleases.30509-4.pdf) Quality control mechanisms ensure the fidelity of ncRNA maturation and eliminate aberrant transcripts. The RNA exosome, a 3'-5' exoribonucleolytic complex comprising the core DIS3 and RRP6 subunits, degrades improperly processed ncRNAs, such as promoter upstream transcripts (PROMPTs) and cryptic unstable transcripts (CUTs), preventing their accumulation and potential interference with gene expression. Nuclear export of mature ncRNAs relies on karyopherin family exportins; for instance, Exportin-5 (XPO5) specifically recognizes the two-nucleotide 3' overhang of pre-miRNAs to facilitate Ran-GTP-dependent translocation through nuclear pores, while Exportin-4 (XPO4) mediates export of select circRNAs exceeding ~100 nucleotides. These pathways highlight length-specific adaptations: small ncRNAs (<200 nucleotides) depend on precise cleavage and exportin-mediated shuttling for rapid function, whereas long ncRNAs (>200 nucleotides) leverage polyadenylation and alternative splicing for enhanced stability and nuclear retention in some cases.00490-0)

Common Structural Motifs

Non-coding RNAs (ncRNAs) exhibit a variety of secondary and tertiary structural motifs that are essential for their stability, interactions, and functional specificity. These motifs often arise from base-pairing patterns that fold the RNA into compact architectures, enabling recognition by proteins or other RNAs. Common secondary structures include stem-loops and hairpins, while tertiary elements like pseudoknots and G-quadruplexes provide higher-order folding critical for or binding. In microRNAs (miRNAs), the precursor (pre-miRNA) adopts a characteristic stem-loop secondary structure, typically 60-80 long with a 2-nucleotide 3' overhang, which facilitates loading into the (RISC) by allowing recognition and processing by and proteins. Similarly, small nucleolar RNAs (snoRNAs) feature a conserved hairpin-hinge-hairpin-tail secondary structure, where the hairpins position antisense sequences to guide site-specific modifications such as 2'-O-methylation or pseudouridylation on ribosomal or spliceosomal RNAs. For small interfering RNAs (siRNAs), the double-stranded regions of their precursors are recognized by , which cleaves them into ~21-23 duplexes based on the helical and end structures, enabling subsequent RISC incorporation. Tertiary motifs further enhance ncRNA functionality through intricate folds. In ribozymes, pseudoknots—where a single-stranded region pairs with a discontinuous stem—stabilize active conformations for catalysis, as seen in the GlmS ribozyme's double pseudoknot architecture that positions the substrate for glucosamine-6-phosphate-mediated self-cleavage. Kink-turns (K-turns), sharp ~120-degree bends formed by tandem sheared G-A base pairs flanked by helices, contribute to catalytic competence in certain ribozymes by facilitating tertiary contacts, such as in variants of the hammerhead ribozyme where they aid in active site assembly. In long non-coding RNAs (lncRNAs), G-quadruplexes—stacks of G-tetrads stabilized by monovalent cations—promote structural stability and regulate localization, exemplified by conserved G-quadruplex motifs in NEAT1 that recruit the protein to paraspeckles. Class-specific features highlight adaptations in ncRNA architecture. Circular RNAs (circRNAs) form covalently closed loops via back-splicing, lacking free ends and thus resisting exonucleolytic degradation, which confers half-lives up to 10-fold longer than linear counterparts and supports sustained regulatory roles. PIWI-interacting RNAs (piRNAs) in clusters exhibit structural bases for ping-pong amplification, where antisense piRNAs with a 10A bias at the 5' end pair with sense transcripts, enabling Zucchini-mediated cleavage and secondary piRNA production from the 3' fragment to amplify transposon silencing signals.00660-6) Computational tools predict these motifs by minimizing or analyzing evolutionary signals. Algorithms like mfold and ViennaRNA employ dynamic programming to compute minimum secondary structures, incorporating thermodynamic parameters for base-pairing and stacking to model stem-loops and hairpins in ncRNAs. Evolutionary covariation—correlated substitutions maintaining base pairs across homologs—identifies conserved motifs by scoring compatibility, distinguishing functional structures from random folds with higher sensitivity at resolution. These structural elements create functional interfaces, such as binding pockets for protein partners. In miRISC, the miRNA guide strand anchors in Argonaute-2's domain via a nucleic acid-binding cleft, where the seed region (positions 2-8) positions for target mRNA recognition, enabling precise silencing. Overall, such motifs underscore how ncRNA architecture dictates molecular interactions without protein-coding potential.

Biological Roles

Gene Regulation

Non-coding RNAs (ncRNAs) play pivotal roles in regulating at the transcriptional and epigenetic levels, influencing architecture, promoter activity, and modifications to control when and where genes are activated or silenced. Long non-coding RNAs (lncRNAs) and other ncRNA classes, such as enhancer RNAs (eRNAs) and PIWI-interacting RNAs (piRNAs), mediate these processes by interacting with DNA, proteins, and -modifying complexes, often in a locus-specific manner. This regulation ensures precise spatiotemporal control of during , , and cellular responses. In transcriptional control, lncRNAs like exemplify cis-acting mechanisms by coating the to induce inactivation through looping and compaction. recruits silencing factors that organize the chromosome into a condensed , spreading repressive marks over large domains via repeat sequences that facilitate long-range interactions. Complementarily, eRNAs, short bidirectional transcripts from enhancer regions, enhance promoter activity by stabilizing enhancer-promoter loops and recruiting and complexes to boost (Pol II) recruitment and elongation. For instance, eRNAs can loop enhancers to target promoters, increasing transcription rates by up to several fold in active loci. Epigenetic modulation by ncRNAs involves directing histone and DNA modifications to enforce heritable gene silencing. The lncRNA HOTAIR acts in trans by recruiting the Polycomb Repressive Complex 2 (PRC2) to specific genomic sites, catalyzing trimethylation of histone H3 at lysine 27 (H3K27me3) to repress Hox gene clusters and other targets over distances exceeding 40 kb. Similarly, piRNAs guide PIWI proteins to transposon loci in germ cells, triggering de novo DNA methylation and H3K9me3 deposition to silence mobile elements and prevent genomic instability. This piRNA-directed pathway establishes methylation patterns during gametogenesis, with piRNA clusters amplifying signals via a ping-pong cycle for robust repression. ncRNAs operate as cis- or trans-acting regulators, with cis elements exerting local effects near their transcription site and trans elements influencing distant loci. Cis-acting examples include promoter-associated ncRNAs that block Pol II progression or stabilize local chromatin loops, such as those overlapping transcription start sites to fine-tune initiation. In contrast, trans-acting ncRNAs like miRNAs or HOTAIR diffuse to bind multiple targets genome-wide, with miRNAs exemplifying broad repression by base-pairing to mRNA sequences, though their indirect transcriptional effects arise via feedback on regulatory genes. Feedback loops further amplify control, where ncRNAs transcribed from enhancers contribute to super-enhancer formation, promoting phase separation into liquid-like condensates that concentrate transcription factors and coactivators for bursty, high-level gene activation. These condensates, driven by multivalent interactions, enhance target gene expression by orders of magnitude in cell-type-specific programs. Quantitative aspects of ncRNA regulation highlight dose-dependent effects, where repression efficiency scales nonlinearly with ncRNA abundance. For miRNA-mediated target repression, models incorporating reveal , often described by Hill coefficient functions with values around 2-4, indicating ultrasensitive responses where small changes in miRNA levels lead to sharp switches in target mRNA stability and translation. This ensures threshold-based control, preventing leaky expression in low-ncRNA states.

RNA Processing and Translation

Non-coding RNAs (ncRNAs) are integral to post-transcriptional RNA processing and the control of protein synthesis, influencing mRNA maturation, stability, and translational efficiency. In pre-mRNA splicing, small nuclear RNAs (snRNAs), including U1, , U4, U5, and U6, assemble into the to recognize splice sites and catalyze removal, ensuring accurate ligation for functional mRNA production. These snRNAs form dynamic ribonucleoprotein complexes (snRNPs) that undergo conformational changes during spliceosome activation, with U1 snRNA binding the 5' splice site and snRNA interacting with the to initiate the process. Small nucleolar RNAs (snoRNAs), such as U85, further enhance splicing fidelity by directing 2'-O-methylation and pseudouridylation modifications on spliceosomal snRNAs like U5, stabilizing their structure and optimizing catalytic activity. Beyond splicing, ncRNAs regulate mRNA stability through targeted degradation pathways. MicroRNAs (miRNAs) and small interfering RNAs (siRNAs), incorporated into RNA-induced silencing complexes (RISCs), bind to mRNA 3' untranslated regions (UTRs), recruiting deadenylation factors like the CCR4-NOT complex to shorten poly(A) tails, followed by via DCP1/DCP2 and 5'-to-3' exonucleolytic decay by XRN1. This deadenylation-dependent mechanism accounts for the majority (>66%) of miRNA-mediated repression in mammalian cells, with mRNA destabilization often dominating over direct translational inhibition. Circular RNAs (circRNAs), a class of covalently closed ncRNAs, function as competing endogenous RNAs (ceRNAs) by sequestering miRNAs through shared binding sites, thereby preventing miRNA-induced deadenylation and stabilizing target mRNAs. NcRNAs also directly modulate initiation and . Transfer RNAs (tRNAs) serve as adaptors that decode mRNA codons by delivering specific to the 's peptidyl (P) and aminoacyl (A) sites, while ribosomal RNAs (rRNAs) form the structural and catalytic core of the , facilitating formation during . In addition, ncRNAs influence mRNA export and localization by interacting with splicing factors. The lncRNA MALAT1, retained in nuclear speckles, binds serine/arginine-rich (SR) proteins such as SRSF1 and SRSF3, modulating their phosphorylation and distribution to regulate outcomes and promote nuclear retention of specific pre-mRNAs, thereby controlling their cytoplasmic export. A representative example is the let-7 miRNA, which represses initiation of target mRNAs by associating with proteins in miRNPs, interfering with the recruitment of the cap-binding protein and disrupting eIF4F complex assembly at the mRNA 5' cap.

Genome Defense and Maintenance

Non-coding RNAs (ncRNAs) play crucial roles in genome defense by countering viral infections through pathways. In and , small interfering RNAs (siRNAs) are generated from viral double-stranded RNA intermediates and guide the (RISC) to cleave complementary viral RNA, thereby restricting . This mechanism is conserved in animals, where siRNAs similarly mediate antiviral defense against a range of RNA viruses, including those in and mammals. In mammals, endogenous siRNAs (endo-siRNAs) derived from cellular transcripts have a limited role in antiviral defense, primarily in specific cell types like oocytes, where they contribute to transposon control and occasionally modulate viral responses, though pathways predominate. NcRNAs are essential for silencing transposable elements (TEs), mobile genetic invaders that threaten genome integrity. Piwi-interacting RNAs (piRNAs), a class of 24-32 ncRNAs, predominate in animal gonads, where they associate with proteins to form complexes that repress TE activity through both post-transcriptional cleavage and transcriptional . This piRNA pathway operates via nuage bodies, cytoplasmic granules in germ cells that facilitate piRNA biogenesis and amplify signals, ensuring TE suppression during . Additionally, long non-coding RNAs (lncRNAs) guide to heterochromatic regions, promoting TE immobilization by recruiting methyltransferases and establishing repressive marks. In and telomere maintenance, ncRNAs regulate processes to prevent genomic instability. , a lncRNA transcribed from subtelomeric promoters, accumulates at ends and stabilizes activity by interacting with the enzyme's RNA component (hTR), facilitating telomere elongation in human cells. also modulates RNA:DNA hybrids known as R-loops at telomeres, where these structures can either promote replication fork progression or, if unregulated, lead to replication stress; ncRNAs like help resolve such hybrids to maintain telomeric integrity. Dysregulated R-loops elsewhere in the are countered by ncRNAs that recruit helicases and ribonucleases, preventing persistent hybrids that could cause replication stalling and mutations. NcRNAs contribute to chromosome structure by organizing domains. The lncRNA coats the in female mammals, spreading from its locus to envelop the entire territory, which triggers modifications and compaction essential for X-chromosome inactivation and dosage compensation. This coating mechanism relies on Xist's repeat domains, which recruit silencing factors like PRC2 to enforce stable . Similarly, satellite II RNAs transcribed from pericentromeric regions interact with proteins such as HP1 to stabilize pericentromeric , promoting higher-order folding and function during . In DNA repair pathways, lncRNAs facilitate the recruitment of repair factors to sites of double-strand breaks (DSBs). For instance, lncRNAs such as and WRAP53 act as scaffolds, binding and localizing proteins like 53BP1 and to DSBs, thereby promoting or as appropriate. These ncRNAs enhance repair efficiency by modulating accessibility and coordinating protein assemblies at break sites, reducing the risk of genomic rearrangements.

Specialized Cellular Functions

Non-coding RNAs (ncRNAs) exhibit specialized functions in cellular signaling pathways, exemplified by the receptor RNA activator (), a (lncRNA) that modulates activity in a manner akin to signaling. enhances receptor-dependent by coordinating the functions of various transcription factors and promoting events. As a selective coactivator, interacts with the amino-terminal activation function of receptors, facilitating without encoding a protein itself. This RNA-mediated regulation allows to fine-tune responses to hormones, influencing processes such as and in hormone-responsive tissues. In prokaryotes, ncRNAs contribute to pathogen evasion strategies through environmental sensing, as seen with the small RNA (sRNA) RyhB in bacteria like Escherichia coli. RyhB senses iron availability and downregulates iron-storage and iron-using proteins during limitation, thereby optimizing resource allocation for survival in iron-scarce host environments. This iron-sparing response, regulated by the ferric uptake regulator (Fur), interlinks homeostasis with virulence, enabling pathogens to evade host immune mechanisms that exploit iron withholding. For instance, RyhB homologs in fish pathogens like Edwardsiella tarda similarly control iron-responsive genes, enhancing persistence during infection. Such mechanisms underscore the role of bacterial sRNAs in adaptive strategies against host defenses. Certain ncRNAs display bifunctionality, serving dual roles as regulators and structural scaffolds, with the 7SK small nuclear RNA (snRNA) providing a prominent example in eukaryotic transcription control. 7SK sequesters positive transcription elongation factor b (P-TEFb), comprising CDK9 and cyclin T, within the 7SK small nuclear ribonucleoprotein (snRNP) complex, thereby inhibiting its kinase activity and promoting promoter-proximal pausing of RNA polymerase II (Pol II). This inhibition, mediated by HEXIM1 binding to 7SK, prevents premature elongation and ensures precise gene expression timing. Simultaneously, 7SK facilitates Pol II pausing at gene promoters, acting as a scaffold for the pause-release machinery upon cellular signals, thus balancing transcriptional output. Dysregulation of this bifunctional role can disrupt elongation control, highlighting 7SK's integral position in transcriptional fidelity. Mitochondrial ncRNAs (mito-ncRNAs), including mitomiRs and lncRNAs, specialize in regulating energy metabolism within organelles. These RNAs modulate mitochondrial respiration and glycolysis by targeting nuclear-encoded mitochondrial genes, thereby influencing ATP production and cellular energy homeostasis. For example, mito-ncR-805 exerts protective effects by positively regulating mitochondrial energy metabolism during stress, enhancing oxidative phosphorylation efficiency. MitomiRs, such as those derived from mitochondrial DNA, further control mitochondrial function by altering the expression of metabolic enzymes, ensuring adaptive responses to bioenergetic demands. This specialized regulation is critical for maintaining mitochondrial integrity in high-energy tissues like the heart. Recent studies (as of 2025) have revealed additional roles for circular RNAs (circRNAs) as scaffolds for protein complexes in cellular signaling and biomolecular condensates, enhancing their regulatory diversity in development and stress responses. Beyond these, ncRNAs participate in and (SG) assembly, providing checkpoints for cellular fidelity under duress. LncRNAs such as LINC00152 regulate mitotic progression by influencing spindle assembly and chromosome segregation, enforcing G2/M checkpoint integrity in cells. Similarly, lncRNAs like MALAT1 modulate by altering expression of cyclins and CDK inhibitors, preventing unscheduled . In stress responses, lncRNAs including SNHG8 drive SG formation by interacting with RNA-binding proteins like G3BP1, sequestering non-translating mRNPs to protect transcripts during oxidative or . Non-coding RNAs also contribute to SG through RNA-RNA interactions and , enriching cytoplasmic granules with regulatory elements that facilitate mRNA and translational repression. These functions highlight ncRNAs' versatility in safeguarding cellular processes amid perturbations.

Roles in Disease

Cancer

Non-coding RNAs (ncRNAs) play pivotal roles in cancer by acting as oncogenes or tumor suppressors, influencing tumorigenesis through diverse regulatory mechanisms. Oncogenic ncRNAs, such as the (lncRNA) MALAT1, promote cancer progression and . MALAT1 is highly expressed in various cancers, including and , where it regulates by modulating serine/arginine splicing factors, thereby enhancing cell motility and invasive potential. For instance, in , MALAT1 depletion impairs without significantly altering global splicing patterns, underscoring its role in fine-tuning pro-metastatic gene expression. Similarly, the miR-21 functions as an by targeting tumor suppressor genes like PTEN and PDCD4, leading to increased and survival in cancers such as and colorectal. Overexpression of miR-21 is commonly observed in many solid tumors, correlating with advanced disease stages and poor . In contrast, tumor-suppressive ncRNAs counteract oncogenic processes. The miR-34 family, transcriptionally activated by , induces and arrest in multiple cancers, including and , by downregulating targets like and CDK4/6. Loss of miR-34 expression is linked to chemoresistance, as it diminishes p53-mediated apoptotic responses. Circular RNAs (circRNAs) can also exert suppressive effects by sponging oncogenic miRNAs; for example, certain circRNAs sequester miR-21 to alleviate its pro-proliferative actions, though ciRS-7 exemplifies a circRNA that sponges the tumor-suppressive miR-7, indirectly promoting oncogenesis in contexts like by derepressing miR-7 targets involved in proliferation. Mechanisms of ncRNA involvement in cancer include epigenetic silencing of tumor suppressors and modulation of key signaling pathways. LncRNAs and miRNAs often recruit epigenetic modifiers, such as DNMT3A, to hypermethylate promoters of genes like PTEN, thereby silencing tumor suppressors and facilitating tumor growth in cancers including ovarian and colorectal. Recent studies highlight ncRNA regulation of the PI3K/Akt pathway, a central oncogenic axis; for instance, lncRNAs like H19 activate PI3K/Akt to enhance and invasion in , while miRNAs such as miR-126 inhibit it to suppress in . Investigations from 2024-2025 emphasize bidirectional ncRNA-PI3K/Akt interactions, with circRNAs also implicated in pathway activation via miRNA sponging. NcRNAs hold promise as diagnostic biomarkers and therapeutic targets in cancer. The lncRNA PCA3 serves as a prostate cancer-specific biomarker, detectable in urine via the Progensa PCA3 assay, which improves detection specificity over PSA alone, aiding in biopsy decisions for patients with elevated PSA levels. Therapeutically, small interfering RNA (siRNA)-based approaches targeting ncRNAs are advancing; while no siRNA drugs are yet approved specifically for cancer as of 2025, investigational siRNAs against oncogenic ncRNAs like STAT3-modulating lncRNAs are in phase II/III trials for solid tumors, showing efficacy in reducing tumor burden when delivered via lipid nanoparticles. Recent advances in single-cell profiling have revealed ncRNA dynamics within tumor microenvironments, identifying miRNA gradients in immune cell interactions that drive immunosuppression in colorectal and breast cancers, as detailed in 2025 reviews. These insights enable precision targeting of ncRNA heterogeneity to overcome therapy resistance.

Neurological and Developmental Disorders

Non-coding RNAs (ncRNAs) play critical roles in the of neurological disorders, particularly through dysregulation that affects synaptic function and . In (AD), the (lncRNA) BACE1-AS stabilizes the mRNA of (BACE1), an enzyme essential for cleaving amyloid precursor protein to produce amyloid-beta (Aβ) peptides, thereby promoting Aβ accumulation and plaque formation. This stabilization occurs via RNA duplex formation between BACE1-AS and BACE1 mRNA, which enhances BACE1 expression under cellular stress conditions prevalent in AD brains. Additionally, deficits in microRNA-132 (miR-132) contribute to pathology by failing to suppress inositol 1,4,5-trisphosphate 3-kinase B (ITPKB), leading to exacerbated hyperphosphorylation and formation in AD mouse models and human patients. Restoring miR-132 expression has been shown to mitigate Aβ-induced and improve neuronal survival, highlighting its neuroprotective potential. In autism spectrum disorder (ASD), ncRNAs influence synaptic plasticity and neurodevelopmental wiring. LncRNA FMR1-AS1, associated with the fragile X mental retardation 1 (FMR1) gene, regulates synaptic protein translation by modulating the activity of fragile X mental retardation protein (FMRP), a key repressor of mRNA translation at synapses; its dysregulation disrupts dendritic spine morphology and long-term potentiation, core features of ASD. Copy number variations (CNVs) affecting miRNA clusters, such as those involving MIR590, MIR944, and MIR3618, are enriched in ASD genomes and alter the expression of genes critical for neuronal connectivity and excitatory-inhibitory balance. These CNV-associated miRNAs target pathways like synaptic scaffolding and chromatin remodeling, contributing to the heterogeneity of ASD phenotypes observed in genetic studies. Beyond specific disorders, ncRNAs are integral to developmental processes in the . MicroRNAs such as let-7, miR-124, and miR-9 act as master regulators of neuronal by promoting the transition from neural progenitors to neurons through post-transcriptional suppression of genes and enhancement of neuronal-specific transcripts. For instance, miR-124 facilitates morphogenesis by repressing non-neuronal targets, ensuring proper arborization during cortical development. In broader developmental contexts, lncRNAs like Upperhand demonstrate how ncRNA transcription can control tissue-specific ; originally identified in 2016 for regulating Hand2 in . Dysregulation of these ncRNAs during embryogenesis can lead to neurodevelopmental delays, underscoring their role in establishing neural circuits. Mechanistically, ncRNAs mediate key aspects of neuronal vulnerability in disorders. LncRNAs such as influence by localizing to dendritic compartments and modulating dynamics, which is disrupted in neurodevelopmental conditions like . In excitotoxicity, a process implicated in AD and stroke-related neurodegeneration, miR-107 regulates subunit expression to prevent excessive calcium influx and neuronal death following glutamate overload. This regulation helps maintain synaptic , and its impairment amplifies inflammatory cascades in affected brains. Recent studies as of 2025 have linked ncRNA dysregulation to exacerbated neurological disorders via COVID-19-induced . LncRNAs like those in the NEAT1 family promote microglial activation and release in response to , worsening aggregation and synaptic loss in patients with preexisting vulnerabilities. MiRNAs, including miR-155, are upregulated in during post-COVID , contributing to blood-brain barrier disruption and heightened risk for AD-like pathologies. These findings suggest ncRNAs as potential biomarkers and therapeutic targets for mitigating infection-triggered neurological sequelae.

Genetic and Metabolic Syndromes

Non-coding RNAs (ncRNAs) play critical roles in genetic and metabolic syndromes, particularly through dysregulation of imprinting and pathways. In imprinted disorders, ncRNAs mediate allele-specific , contributing to phenotypes such as growth abnormalities and metabolic imbalances. For instance, long non-coding RNAs (lncRNAs) like Airn in mice establish paternal imprinting of Igf2r by overlapping transcription with the Igf2r gene, preventing its expression on the paternal without requiring the mature lncRNA product itself. This mechanism highlights how ncRNA transcription, rather than the RNA molecule, can recruit repressive complexes to enforce imprinting. Prader-Willi syndrome (PWS), a characterized by , , and hyperphagia leading to , arises from loss of paternal expression in the 15q11.2-q13 region, including the SNORD116 cluster of small nucleolar RNAs (snoRNAs). Deletion or of the paternal SNORD116 locus disrupts of food intake and body weight, as evidenced by mouse models lacking Snord116 that exhibit increased energy expenditure, postnatal growth retardation, and hyperphagia mimicking PWS symptoms. These C/D box snoRNAs guide 2'-O-methylation of ribosomal RNAs and other targets, and their absence alters hypothalamic function, confirming SNORD116's essential role in the syndrome's metabolic . Cartilage-hair hypoplasia (CHH), an autosomal recessive metaphyseal , results from in the RMRP encoding the component of the RNase MRP endoribonuclease complex, a non-coding involved in multiple cellular processes including . These impair pre-rRNA cleavage at sites A0, A1, and A3, leading to defective 5.8S rRNA maturation and reduced 40S ribosomal subunit production, which manifests as , fine hair, and skeletal abnormalities due to disrupted chondrocyte proliferation in the growth plate. In CHH patient fibroblasts, RMRP variants cause inefficient promoter activity and RNA instability, further exacerbating assembly defects and immune dysfunction. Beyond direct genetic syndromes, ncRNAs influence metabolic conditions like non-alcoholic fatty liver disease (NAFLD), where lncRNAs regulate lipid homeostasis. For example, lncRNAs such as HULC and MALAT1 promote hepatic by modulating pathways like PPARγ and SREBP-1c, enhancing de novo lipogenesis and accumulation in hepatocytes. Recent reviews highlight over 50 NAFLD-associated lncRNAs that interact with miRNAs or transcription factors to dysregulate efflux and oxidation, positioning them as potential biomarkers and therapeutic targets. snoRNA-guided modifications are implicated in growth plate development disruptions within these syndromes, as seen in CHH where impaired RNase MRP activity indirectly affects rRNA pseudouridylation and , essential for function in proliferating chondrocytes. This leads to disorganized and metaphyseal dysplasia, underscoring ncRNAs' role in linking RNA to skeletal .

Other Pathological Conditions

Mutations in the miR-96 have been identified as a cause of non-syndromic autosomal dominant progressive , known as DFNA50, by disrupting the development and maintenance of sensory s in the . These mutations impair miR-96's regulatory role in and , leading to degeneration of auditory structures and sensorineural hearing that typically begins in adolescence and worsens over time. In models, miR-96 knockout or point mutations similarly result in cochlear dysfunction, underscoring its conserved function in maturation. Mitochondrial tRNA disorders, such as those caused by mutations in the MT-TL1 gene, contribute to mitochondrial encephalomyopathy, , and stroke-like episodes () syndrome by impairing . The most common variant, m.3243A>G in MT-TL1, which encodes the mitochondrial tRNA for (tRNALeu(UUR)), disrupts tRNA and aminoacylation, reducing mitochondrial protein and ATP in high-energy tissues like muscle and . This leads to systemic symptoms including , seizures, and , with levels influencing disease severity. In infectious diseases, non-coding RNAs play critical roles in host-pathogen interactions, where viral miRNAs often evade host immunity by modulating responses and pathways. For instance, viruses such as herpesviruses encode miRNAs that target host transcripts to suppress antiviral signaling, facilitating persistent . In the context of , SARS-CoV-2 indirectly exploits host miRNAs and lncRNAs to dysregulate immune evasion, with studies from 2024 linking altered miRNA profiles to exacerbated and immune suppression during . Long non-coding RNAs (lncRNAs) contribute to in (SLE) by promoting type I signaling, a hallmark of the disease. For example, the lncRNA linc00513 is overexpressed in SLE patients and acts as a positive regulator of the IFN pathway by enhancing and expression, thereby amplifying autoimmune responses. Similarly, NEAT1 upregulation in granulocytic myeloid-derived suppressor cells boosts IFN-I activation in B cells, contributing to production and disease pathogenesis. Recent 2025 studies have highlighted non-coding RNAs as promising therapeutic targets in inflammatory diseases, with exosomal miRNAs and lncRNAs modulating immune cell communication and storms in conditions like and . These investigations emphasize ncRNA-based interventions, such as targeting dysregulated miRNAs, to attenuate and restore immune .

References

  1. [1]
  2. [2]
    Non-coding RNAs: the architects of eukaryotic complexity - PMC
    Around 98% of all transcriptional output in humans is non-coding RNA. RNA-mediated gene regulation is widespread in higher eukaryotes.
  3. [3]
    Non-coding RNA - PubMed
    The term non-coding RNA (ncRNA) is commonly employed for RNA that does not encode a protein, but this does not mean that such RNAs do not contain information ...
  4. [4]
    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 ...
  5. [5]
    Beyond mRNA: The Role of Non-coding RNAs in Normal and ...
    Long non-coding RNAs are defined as a large class of RNAs with sequences longer than 200 nucleotides which differentiates them from classically defined small ...
  6. [6]
    Computational approaches towards understanding human long non ...
    Identification of lncRNAs broadly involves selection of non-protein coding transcripts longer than 200 nucleotides that lack an open reading frame (ORF) of ...
  7. [7]
    2′-O-methylation (Nm) in RNA: progress, challenges, and future ...
    However, the 2′-O-methyl group stabilizes the C3′-endo ribose conformation found in the A-form RNA helix or in stacked nucleotides, and as a result, each Nm ...
  8. [8]
    Long noncoding RNAs: Re-writing dogmas of RNA processing ... - NIH
    In place of a poly(A) tail, these transcripts are stabilized by highly conserved triple helical structures. Sno-lncRNAs likewise lack poly(A) tails and instead ...
  9. [9]
    Review Life without A tail: New formats of long noncoding RNAs
    While most long noncoding RNAs (lncRNAs) appear indistinguishable from mRNAs, having 5′ cap structures and 3′ poly(A) tails, recent work has revealed new ...
  10. [10]
    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.
  11. [11]
    estimate of the total number of true human miRNAs - Oxford Academic
    Mar 1, 2019 · In total, we extrapolated 2300 true human mature miRNAs, 1115 of which are currently annotated in miRBase V22. The experimentally validated ...INTRODUCTION · MATERIALS AND METHODS · RESULTS · DISCUSSION<|separator|>
  12. [12]
    Small Non-coding RNAs Mount a Silent Revolution in Gene ... - NIH
    The first described small ncRNAs, miRNAs, siRNAs and Piwi-interacting RNAs (piRNAs), are classified by distinct biogenesis pathways and regulatory ...Mirnas: Diverse Biogenesis... · Pirnas: Silencers Of The... · Endogenous Sirnas: A Natural...
  13. [13]
    The biogenesis, biology and characterization of circular RNAs - Nature
    Aug 8, 2019 · Circular RNAs (circRNAs) are covalently closed, endogenous biomolecules in eukaryotes with tissue-specific and cell-specific expression patterns.
  14. [14]
    The Noncoding RNA Revolution—Trashing Old Rules to Forge New ...
    Mar 27, 2014 · They regulate gene expression at the levels of transcription, RNA processing, and translation. They protect genomes from foreign nucleic acids.
  15. [15]
    Overview of MicroRNA Biogenesis, Mechanisms of Actions, and ...
    In general, the non-canonical miRNA biogenesis can be grouped into Drosha/DGCR8-independent and Dicer-independent pathways. Pre-miRNAs produced by the Drosha/ ...
  16. [16]
    an emerging small non-coding RNA with key roles in cancer - NIH
    Jun 3, 2022 · tsRNAs can exert functions such as regulating gene expression to influence cancer progression. Their dysregulation is closely associated with various cancers.Missing: srRNAs | Show results with:srRNAs
  17. [17]
    On Protein Synthesis - Francis Crick - Profiles in Science
    PDF · OCR (Text). Title: On Protein Synthesis; Creator: Crick, Francis, 1916-2004; Date: 1958; Periodical: The Symposia of the Society for Experimental Biology ...
  18. [18]
    rRNA—the evolution of that magic molecule - PMC - NIH
    Carl Woese (and George Fox) discovered the third form of life, the Archaebacteria (later renamed Archaea) and called rRNA the magic molecule.
  19. [19]
    STRUCTURE OF A RIBONUCLEIC ACID - PubMed
    1965 Mar 19;147(3664):1462-5. doi: 10.1126/science.147.3664.1462. Authors. R W HOLLEY, J APGAR, G A EVERETT, J T MADISON, M MARQUISEE, S H MERRILL, J R ...
  20. [20]
    Bacteriophage lambda: early pioneer and still relevant - PMC
    The first of these to be discovered was the P22 antirepressor (Ant), which binds to repressor and causes it to release repression (Susskind and Botstein, 1975).
  21. [21]
    Ribonuclease P: an enzyme with an essential RNA component.
    The presence of a discrete RNA component in RNase P appears to be essential for enzymatic function. A model is described for enzyme-substrate.Missing: paper | Show results with:paper
  22. [22]
    Self-splicing RNA: Autoexcision and autocyclization of the ribosomal ...
    Cech et al., 1982. Cech T.R., Zaug A.J., Grabowski P.J., Brehm S.L.. Transcription and splicing of the ribosomal RNA precursor of Tetrahymena. Busch H ...
  23. [23]
    Are snRNPs involved in splicing? - Nature
    Jan 10, 1980 · Here we present several lines of evidence that suggest a direct involvement of snRNPs in the splicing of hnRNA.
  24. [24]
    Evidence for a major role of antisense RNAs in cyanobacterial gene ...
    It was known early on that such natural antisense RNAs (asRNAs) control phage development and plasmid replication in bacteria (Wagner and Simons, 1994), yet ...
  25. [25]
    The Drosha-DGCR8 complex in primary microRNA processing
    RNase III proteins play key roles in microRNA (miRNA) biogenesis. The nuclear RNase III Drosha cleaves primary miRNAs (pri-miRNAs) to release hairpin-shaped pre ...
  26. [26]
    Splicing regulation by long noncoding RNAs - PMC - NIH
    Feb 7, 2018 · In this review, we focus on recent discoveries linking gene regulation by alternative splicing and its modulation by long and small ncRNAs.Missing: seminal | Show results with:seminal
  27. [27]
    A small nucleolar guide RNA functions both in 2′-O-ribose ... - NIH
    The U85 snoRNA directs 2′-O-methylation of the C45 and pseudouridylation of the U46 residues in the invariant loop 1 of the human U5 spliceosomal RNA.
  28. [28]
    snoRNAs: functions and mechanisms in biological processes, and ...
    May 12, 2022 · In this review, we summarize the reported functions of snoRNAs and the possible mechanisms underlying their role in tumorigenesis and cancer development.
  29. [29]
    Exportin-5 mediates the nuclear export of pre-microRNAs and short ...
    We demonstrate that human pre-miRNA nuclear export, and miRNA function, are dependent on Exportin-5. Exportin-5 can bind pre-miRNAs specifically in vitro.Missing: ncRNAs | Show results with:ncRNAs
  30. [30]
    Exportin 4 depletion leads to nuclear accumulation of a subset of ...
    Oct 1, 2022 · Here, we provide lines of evidence to demonstrate a link between the conserved Exportin 4 (XPO4) and nuclear export of a subset of circRNAs in metazoans.
  31. [31]
    Functional diversity of small nucleolar RNAs - Oxford Academic
    Small nucleolar RNAs (snoRNAs) are short non-protein-coding RNAs with a long-recognized role in tuning ribosomal and spliceosomal function by guiding ribose ...
  32. [32]
    MicroRNAs: regulators of gene expression and cell differentiation
    Dec 1, 2006 · Their immediate precursors, called pre-miRNAs, are characteristic stem-loop structures 60 to 80 bases in length with a 2-nucleotide 3′ overhang.
  33. [33]
    Elements essential for accumulation and function of small nucleolar ...
    The pseudouridylation guide snoRNAs share a common 'hairpin–hinge– hairpin–tail' secondary structure and two conserved sequence motifs, the H and ACA boxes, ...
  34. [34]
    Origins and Mechanisms of miRNAs and siRNAs - PubMed Central
    Double-stranded RNA precursors of various kinds are processed by a Dicer protein into short (∼20−30 nt) fragments. One strand of the processed duplex is loaded ...Sirnas And Mirnas: Themes In... · Micrornas · Microrna BiogenesisMissing: paper | Show results with:paper
  35. [35]
    Structural Investigation of the GlmS Ribozyme Bound to Its Catalytic ...
    The GlmS ribozyme adopts a compact double pseudoknot tertiary structure, with two closely packed helical stacks. Recognition of GlcN6P is achieved through ...
  36. [36]
    Biochemical characterization of the kink-turn RNA motif - PMC - NIH
    The kink-turn (K-turn or GA) motif, which causes a sharp bend in an RNA ... ribozyme folding and catalysis. RNA, 3, 838–849. [PMC free article] [PubMed] ...Missing: paper | Show results with:paper
  37. [37]
    G-quadruplexes offer a conserved structural motif for NONO ...
    Jun 4, 2020 · We find that G-quadruplex motifs are abundant and conserved features of NEAT1. Furthermore, we determine that NONO binds NEAT1 G-quadruplexes with structural ...
  38. [38]
    Circular RNA: metabolism, functions and interactions with proteins
    Dec 14, 2020 · Circular RNAs (CircRNAs) are single-stranded, covalently closed RNA molecules that are ubiquitous across species ranging from viruses to mammals.
  39. [39]
    Mfold web server for nucleic acid folding and hybridization prediction
    The objective of this web server is to provide easy access to RNA and DNA folding and hybridization software to the scientific community at large.Abstract · INTRODUCTION · SERVER CONTENT AND... · FUTURE DIRECTIONS
  40. [40]
    ViennaRNA Package 2.0 - PMC - PubMed Central
    The main secondary structure prediction tool is RNAfold , which computes the minimum free energy (MFE) and backtraces an optimal secondary structure. Using the ...
  41. [41]
    Evolutionary conservation of RNA sequence and structure - Rivas
    Mar 22, 2021 · Covariation together with covariation power allow to distinguish between three different conservation patterns in RNA alignments.
  42. [42]
    The Structure of Human Argonaute-2 in Complex with miR-20a - PMC
    The miRNA is bound at both ends—the 5′ end to the Mid domain with some contributions from the PIWI domain, and the 3′ end to the PAZ domain. The seed sequence ...Missing: seminal | Show results with:seminal
  43. [43]
    Gene regulation by long non-coding RNAs and its biological functions
    Dec 22, 2020 · In this Review, we discuss the mechanisms of lncRNA biogenesis, localization and functions in transcriptional, post-transcriptional and ...
  44. [44]
    Role of small nuclear RNAs in eukaryotic gene expression
    Apr 30, 2013 · Of the five major spliceosomal snRNAs (U1, U2, U4, U5 and U6), three of them (U2, U5 and U6) have clear structural and functional similarities ...
  45. [45]
    Mechanisms and regulation of spliceosome‐mediated pre‐mRNA ...
    Jul 7, 2024 · Pre-mRNA splicing, the removal of introns and ligation of flanking exons, is a crucial step in eukaryotic gene expression. The spliceosome ...
  46. [46]
    O‐ribose methylation and pseudouridylation of the U5 spliceosomal ...
    The U85 is the first example of a snoRNA that directs modification of an RNA polymerase II‐transcribed spliceosomal RNA and that functions both in RNA ...
  47. [47]
    Control of translation and mRNA degradation by miRNAs and siRNAs
    Moreover, in mammalian cells, translation repression by the Let-7 miRNA, or by tethered Argonaute proteins, can shift the mRNA target to lighter fractions in ...
  48. [48]
    Article mRNA Destabilization Is the Dominant Effect of Mammalian ...
    Oct 2, 2014 · Regardless of the miRNA, cell type, growth condition, or translational state, mRNA destabilization explains most (66%–>90%) miRNA-mediated repression.
  49. [49]
    From RNA to Protein - Molecular Biology of the Cell - NCBI Bookshelf
    These adaptors consist of a set of small RNA molecules known as transfer RNAs (tRNAs), each about 80 nucleotides in length.
  50. [50]
    Many lncRNAs, 5'UTRs, and pseudogenes are translated and ... - eLife
    Dec 19, 2015 · In addition, ~35% of mRNA coding genes are translated upstream of the primary protein-coding region (uORFs) and 4% are translated downstream ( ...
  51. [51]
    Antiviral RNAi in Insects and Mammals: Parallels and Differences
    Abstract. The RNA interference (RNAi) pathway is a potent antiviral defense mechanism in plants and invertebrates, in response to which viruses evolved ...
  52. [52]
    RNAi-mediated antiviral immunity in mammals - ScienceDirect.com
    Jul 14, 2018 · Accumulating evidence indicates that RNAi is an accessory antiviral mechanism in mammals. Humans have at least three antiviral defense mechanisms.
  53. [53]
    Endogenous small interfering RNAs in animals - PMC - NIH
    The necessity to preserve RNAi in mammals has been somewhat of an enigma as they seem to have mostly dispensed with siRNAs for antiviral defence, and some ...
  54. [54]
    The piRNA Pathway Guards the Germline Genome Against ...
    A specific RNA silencing pathway, termed the piRNA pathway operates in germ cells of animals to control TE activity.Missing: seminal | Show results with:seminal
  55. [55]
    piRNA pathway and the potential processing site, the nuage, in the ...
    In addition to silencing transposons, piRNAs also regulate other processes, such as chromosome segregation, mRNA degradation and germline differentiation.Missing: seminal paper
  56. [56]
    The emergence of piRNAs against transposon invasion to preserve ...
    Nov 10, 2017 · This review features our current understandings of mammalian PIWI-interacting RNAs (piRNAs) and their role in TE regulation in spermatogenesis.Primary Pirna Biogenesis · Pre-Pachytene Pirnas... · Pachytene Pirnas Regulate...
  57. [57]
    Telomeric Repeat-Containing RNA (TERRA): A Review of the ... - NIH
    Mar 18, 2022 · Recent evidences indicate that TERRA molecules play a crucial role in telomere length regulation, telomerase activity, and heterochromatin ...
  58. [58]
    TERRA transcripts localize at long telomeres to regulate telomerase ...
    Jun 12, 2024 · We report that TERRA transcripts relocate away from chromosome ends during telomere lengthening, leading to a reduced number of telomeric TERRA-hTR molecules.
  59. [59]
    A double-edged sword: R loops as threats to genome integrity and ...
    R loops are three-stranded nucleic acid structures that comprise nascent RNA hybridized with the DNA template, leaving the nontemplate DNA single-stranded.
  60. [60]
    X-Inactivation: Xist RNA Uses Chromosome Contacts to Coat the X
    Xist RNA spreads from the Xic to coat the X and contributes to the initial establishment of silencing and subsequent maintenance of XCI.
  61. [61]
    An Xist-activating antisense RNA required for X-chromosome ...
    Oct 19, 2015 · The Xist long non-coding (lnc) RNA is preferentially upregulated from the prospective inactive X and physically coats that X chromosome at the ...
  62. [62]
    Pericentromeric satellite RNAs as flexible protein partners in the ...
    Jul 7, 2024 · Pericentromeric heterochromatin is mainly composed of satellite DNA sequences. Although being historically associated with transcriptional ...
  63. [63]
    The Role of Long Non Coding RNAs in the Repair of DNA Double ...
    LncRNAs participate in several steps of DNA repair pathways and regulate the expression of key components of these pathways including p53 tumor suppressor gene.
  64. [64]
    The Role of Noncoding RNAs in Double-Strand Break Repair
    In this review, we will summarize studies reporting the different roles of ncRNAs in DSB repair and discuss how genome editing approaches, especially CRISPR/ ...
  65. [65]
    Steroid receptor RNA activator: Biologic function and role in disease
    Aug 1, 2016 · SRA is a type of long noncoding RNA (lncRNA) which coordinates the functions of various transcription factors, enhances steroid receptor-dependent gene ...
  66. [66]
    A Steroid Receptor Coactivator, SRA, Functions as an RNA and Is ...
    SRA is selective for steroid hormone receptors and mediates transactivation via their amino-terminal activation function. We provide functional and mechanistic ...
  67. [67]
    Steroid receptor RNA activator (SRA1): unusual bifaceted gene ...
    The steroid receptor RNA activator (SRA) is a unique modulator of steroid receptor transcriptional activity, as it is able to mediate its coregulatory effects ...
  68. [68]
    A small RNA regulates the expression of genes involved in iron ...
    The RyhB 90-nt RNA down-regulates a set of iron-storage and iron-using proteins when iron is limiting; it is itself negatively regulated by the ferric uptake ...Missing: evasion | Show results with:evasion
  69. [69]
    Fur and RyhB as major regulators of bacterial pathogenicity - PubMed
    Aug 31, 2015 · By sensing iron availability in the environment, Fur and RyhB are able to regulate, either directly or indirectly via other transcriptional ...Missing: sRNA | Show results with:sRNA
  70. [70]
    Battle for Metals: Regulatory RNAs at the Front Line - Frontiers
    Jul 4, 2022 · In pathogens, RyhB-like sRNAs interlink iron homeostasis and virulence. Porcheron et al. (2014) demonstrated that the deletion of ryhB gene in ...<|separator|>
  71. [71]
    Tiny but Mighty: Small RNAs—The Micromanagers of Bacterial ...
    RyhB. RyhB is a 95 nt sRNA that plays a key role in iron homeostasis and virulence in several pathogenic and non-pathogenic strains of bacteria including E.
  72. [72]
    Cracking the control of RNA polymerase II elongation by 7SK ...
    In this survey, we first review the fundamentals of gene transcription control by P-TEFb and its sequestration within the inhibitory 7SK snRNP complex.INTRODUCTION · KEEPING P-TEFb IN CHECK... · LIBERATING P-TEFb FROM...
  73. [73]
    Inhibition of P-TEFb (CDK9/Cyclin T) Kinase and RNA Polymerase II ...
    P-TEFb was inhibited by the HEXIM1 protein in a process that specifically required 7SK for mediating the HEXIM1:P-TEFb interaction.
  74. [74]
    Transcription elongation control by the 7SK snRNP complex
    The 7SK snRNP complex holds P-TEFb, which is released to become active and facilitate Pol II pause release. The complex is present at promoters.
  75. [75]
    Cracking the control of RNA polymerase II elongation by 7SK ...
    Sep 19, 2016 · Here, we provide an overview on stimulating Pol II pause release by P-TEFb and on sequestering P-TEFb into 7SK snRNP.<|separator|>
  76. [76]
    Mitochondrial noncoding RNA-regulatory network in cardiovascular ...
    Mar 5, 2020 · Mitochondrial ncRNAs participate in controlling cardiovascular pathogenesis by regulating glycolysis, mitochondrial energy status, and the expression of genes.Missing: mito- | Show results with:mito-
  77. [77]
    Retrograde signaling by a mtDNA-encoded non-coding RNA ...
    Oct 30, 2020 · The protective effects of mito-ncR-805 are associated with positive regulation of mitochondrial energy metabolism, and respiration.
  78. [78]
    Mitochondrial microRNAs: Key Drivers in Unraveling ... - PubMed
    Jan 13, 2025 · These mitochondrial miRNAs, also known as mitomiRs, are essential for regulating mitochondrial function and metabolism. MitomiRs can originate ...Missing: mito- | Show results with:mito-
  79. [79]
    Mitochondria-associated non-coding RNAs and their impact on drug ...
    Feb 26, 2025 · Non-coding RNAs (ncRNAs) enriched in mitochondria ... mitochondrial function, including mitochondrial substance exchange and energy metabolism.Missing: mito- | Show results with:mito-
  80. [80]
    The long non-coding RNA LINC00152 is essential for cell cycle ...
    May 23, 2017 · The long non-coding RNA LINC00152 is essential for cell cycle progression through mitosis in HeLa cells · Authors · Affiliations.
  81. [81]
    Cell cycle regulation by long non-coding RNAs - PubMed
    Many lncRNAs are involved in the regulation of critical cell cycle regulators such as the cyclins, CDKs, CDK inhibitors, pRB, and p53.
  82. [82]
    Long non-coding RNA SNHG8 drives stress granule formation in ...
    Sep 21, 2023 · The commonly differentially expressed lncRNAs interact with RNA-binding proteins that regulate stress granule formation. Among these lncRNAs, ...
  83. [83]
    RNA self-assembly contributes to stress granule formation and ...
    Feb 26, 2018 · Additional evidence that RNA–RNA interactions could contribute to stress granule assembly came from the partitioning of noncoding RNAs (ncRNAs) ...
  84. [84]
    The Integral Role of RNA in Stress Granule Formation and Function
    May 19, 2021 · Stress granules (SGs) are phase-separated, membraneless, cytoplasmic ribonucleoprotein (RNP) assemblies whose primary function is to promote cell survival.
  85. [85]
    The Emerging Roles of the β-Secretase BACE1 and the Long Non ...
    BACE1 or less BACE1-AS inhibition has shown therapeutic potentials particularly in decreasing manifestations of amyloid-linked neurodegenerative diseases. Here, ...
  86. [86]
    The effect of BACE1-AS on β-amyloid generation by regulating ...
    Oct 1, 2019 · BACE1-AS is involved in the mechanism regulating BACE1 expression and Aβ generation in APPsw transgenic cells.
  87. [87]
    miR-132/212 deficiency impairs tau metabolism and promotes ...
    These findings support a role for miR-132/212 in the regulation of tau pathology in mice and humans and provide new alternatives for therapeutic development.
  88. [88]
    Alzheimer's Disease and microRNA-132: A Widespread ... - Frontiers
    Accumulating pre-clinical results suggest that miR-132 may be involved in the progression of Aβ and tau pathology. Moreover, clinical studies have indicated ...
  89. [89]
    Regulatory role of LncRNA FMR1-AS1 in the pathogenesis ... - Nature
    Aug 17, 2025 · Previous studies have shown that FMR1 encodes the Fragile X Messenger Ribonucleoprotein 1 (FMRP), which regulates synaptic protein translation ...
  90. [90]
    Decoding microRNAs in autism spectrum disorder - PMC
    By analyzing CNV loci of miRNAs in the autism database, several CNV-miRNA were identified, including MIR590, MIR944, MIR3618, and MIR1306. (2) ...
  91. [91]
    Autism-associated miR-873 regulates ARID1B, SHANK3 ... - Nature
    Dec 1, 2020 · The relationship between gene expression and large copy number variations (CNVs) containing miRNAs has also been linked with ASD. A recent ...
  92. [92]
    microRNAs: key triggers of neuronal cell fate - Frontiers
    MicroRNAs let-7, microRNA-124, and microRNA-9 have been shown to promote the differentiation of neural stem cells and neural progenitors into specific neural ...
  93. [93]
    Regulation of LncRNAs and microRNAs in neuronal development ...
    Apr 5, 2023 · In this review, we summarize current knowledge on ncRNA taxonomy, biogenesis, and function, and discuss current research progress on ncRNAs.
  94. [94]
    A Novel Long Non-coding RNA, durga Modulates Dendrite Density ...
    Apr 9, 2017 · A Novel Long Non-coding RNA, durga Modulates Dendrite Density ... morphogenesis is linked to synaptic plasticity, learning and cognition.
  95. [95]
    Emerging Roles of Non-Coding RNA in Neuronal Function and ...
    miR-107 has been shown to play a key role in the regulation of excitotoxicity in ischaemic neuronal injury, associated with increased glutamate accumulation ...
  96. [96]
    unraveling the role of long non-coding RNAs in COVID-19 ...
    Aug 28, 2025 · The regulatory roles of lncRNAs in immune responses and inflammation suggest their potential utility in other infectious and inflammatory ...
  97. [97]
    MicroRNAs in long COVID: roles, diagnostic biomarker potential and ...
    Altered miRNA expression in Long COVID is linked to persistent inflammation, central nervous system dysfunction, and other chronic complications ...
  98. [98]
    Highly restricted deletion of the SNORD116 region is implicated in ...
    Jun 11, 2014 · The SNORD116 locus lies in the 15q11-13 region of paternally expressed genes implicated in Prader–Willi Syndrome (PWS), a complex disease ...
  99. [99]
    Snord116 is critical in the regulation of food intake and body weight
    Jan 4, 2016 · Here we show that mice lacking Snord116 globally have low birth weight, increased body weight gain, energy expenditure and hyperphagia.
  100. [100]
    A disease-linked lncRNA mutation in RNase MRP inhibits ribosome ...
    Feb 3, 2022 · Mutations associated with Cartilage Hair Hypoplasia impair pre-rRNA processing. Fibroblasts from CHH patients have reduced growth rates and cell ...
  101. [101]
    RNase MRP RNA and human genetic diseases | Cell Research
    Dec 26, 2006 · Mutations on RNase MRP RNA gene (RMRP) cause a recessively inherited developmental disorder, cartilage-hair hypoplasia (CHH).
  102. [102]
    A comprehensive review of long non-coding RNAs in the ...
    Feb 23, 2021 · There is a body of evidence that lncRNAs have a pivotal role in triglyceride, cholesterol, and lipoprotein metabolism. Moreover, lncRNAs by up- ...
  103. [103]
    Ribosome biogenesis in disease: new players and therapeutic targets
    Jan 9, 2023 · Cartilage Hair Hypoplasia (CHH) is an autosomal recessive disorder caused by mutations of the RNA component of the mitochondrial RNA ...
  104. [104]
    Hearing impairment due to Mir183/96/182 mutations suggests both ...
    The microRNA miR-96 is important for hearing, as point mutations in humans and mice result in dominant progressive hearing loss. Mir96 is expressed in sensory ...
  105. [105]
    A cell type–specific approach to elucidate the role of miR-96 in inner ...
    May 8, 2024 · Mutations in microRNA-96 (miR-96), a microRNA expressed within the hair cells (HCs) of the inner ear, result in progressive hearing loss in both mouse models ...Abstract · Background and summary · Results · Discussion
  106. [106]
    Targeted genome editing restores auditory function in adult mice ...
    Jul 10, 2024 · Mutations in microRNA-96 (MIR96) cause autosomal dominant deafness-50 (DFNA50), a form of delayed-onset hearing loss.
  107. [107]
    Melas Syndrome - StatPearls - NCBI Bookshelf - NIH
    Jan 25, 2024 · Numerous tRNA variations can contribute to the development of MELAS, with the most prevalent variant found in the MTTL1 mitochondrial gene. A ...
  108. [108]
    MT-TL1 gene: MedlinePlus Genetics
    May 1, 2014 · The MT-TL1 gene provides instructions for making a molecule called a transfer RNA (tRNA), which is a chemical cousin of DNA.
  109. [109]
    MELAS syndrome: Clinical manifestations, pathogenesis, and ...
    The most common mutation associated with MELAS syndrome is the m.3243A>G mutation in the MT-TL1 gene encoding the mitochondrial tRNALeu(UUR). The m.
  110. [110]
    Interferon and immunity: the role of microRNA in viral evasion ...
    It highlights how viruses can manipulate host cellular pathways, particularly miRNA expression, to evade the immune response.
  111. [111]
    Noncoding RNAs in the COVID-19 Saga: An Untold Story
    Many viruses have evolved mechanisms that exploit ncRNAs to evade the host immune system. Understanding the regulatory roles of ncRNAs in host immunity during ...
  112. [112]
    The relationship between microRNAs and COVID-19 complications
    Aug 22, 2024 · Sometimes, the viral proteins can impair host immunity with their own microRNAs and specific proteins [20]. The roles of TMPRSS2 and ACE2 in ...
  113. [113]
    Identification of LncRNA Linc00513 Containing Lupus-Associated ...
    In conclusion, our findings reveal the over expression of linc00513 plays a role in lupus pathogenesis by promoting IFN signaling pathway. SNP variants of ...
  114. [114]
    Identification of LncRNA Linc00513 Containing Lupus-Associated ...
    We demonstrated that linc00513 was a novel positive regulator of IFN signaling pathway and was responsible for the amplified IFN signaling in SLE patients. Our ...
  115. [115]
    LncRNA Expression Profiles in Systemic Lupus Erythematosus and ...
    (A) lncRNA NEAT1 is overexpressed in G-MDSCs and induces the promotion of G-MDSCs on IFN-I signaling activation of B cells, contributing to the pathogenesis of ...
  116. [116]
    Exosomal non-coding RNAs - Journal of Inflammation
    May 14, 2025 · This review synthesizes recent advances in how these ncRNAs orchestrate immune cell communication, modulate inflammatory mediators, and drive ...
  117. [117]
    Non-coding RNAs: emerging biomarkers and therapeutic targets in ...
    The current review aims to investigate the roles of ncRNAs as potential biomarkers and therapeutic targets in cancer and inflammatory diseases.