Fact-checked by Grok 2 weeks ago

RNA-binding protein

RNA-binding proteins (RBPs) are a diverse class of proteins, with over 1,500 encoded in the , that specifically interact with molecules to form ribonucleoprotein complexes essential for regulating . These proteins bind to through specialized domains such as the RNA recognition motif (RRM), K-homology (KH) domain, and (ZnF) motifs, enabling interactions via hydrogen bonds, stacking, and hydrophobic contacts that influence structure and function. RBPs play pivotal roles across all stages of RNA metabolism, from transcription to degradation, and are found in all domains of life, underscoring their fundamental importance in cellular biology. The primary functions of RBPs include regulating pre-mRNA splicing, where they facilitate alternative inclusion or exclusion, as seen with proteins like RBFOX2 and SRSF1; controlling mRNA stability and decay, exemplified by HuR stabilizing transcripts and TTP promoting degradation; directing mRNA localization to specific cellular compartments via proteins such as CPEB1; and modulating translation efficiency through factors like . These activities often occur in dynamic, multi-protein complexes, such as the , which assembles to process nascent transcripts. Large-scale studies, including enhanced crosslinking immunoprecipitation (eCLIP) and RNA Bind-N-Seq, have mapped binding specificities for hundreds of human RBPs, revealing preferences for motifs like UGCA for many nuclear RBPs and enabling predictions of their regulatory impacts. Dysfunction in RBPs is implicated in numerous diseases, particularly neurodegeneration and cancer, where mutations or mislocalization disrupt RNA homeostasis—for instance, TDP-43 and FUS aggregates in () and (FTD), or SF3B1 mutations driving aberrant splicing in myelodysplastic syndromes. In cancer, RBPs like LIN28 promote oncogenesis by inhibiting let-7 maturation, while in viral infections, host RBPs can either restrict or facilitate replication. Ongoing research highlights RBPs as promising therapeutic targets, with approaches like small-molecule inhibitors and antisense aimed at restoring RNA in disease states.

Molecular Structure and Domains

Overall Protein Architecture

RNA-binding proteins (RBPs) are characterized by a modular architecture that integrates one or more RNA-binding domains (RBDs) with auxiliary elements to enable versatile interactions with targets. This modularity allows RBPs to achieve specificity and affinity through combinatorial arrangements of domains, such as tandem repeats of RRMs or domains, which are prevalent in proteins like heterogeneous nuclear ribonucleoproteins (hnRNPs). For instance, hnRNP A1 features two RRMs connected by a flexible linker, permitting simultaneous engagement with distant sequences and enhancing overall binding efficiency by up to 1000-fold compared to single-domain constructs. Auxiliary domains, including glycine-arginine (RG)-rich regions and low-complexity motifs, often flank these RBDs to regulate accessibility and promote multivalent interactions. A significant of RBP is the prevalence of intrinsically disordered regions (IDRs), which constitute over 80% of the sequence in approximately 20% of mammalian RBPs and contribute to structural flexibility. These IDRs, enriched in charged or polar residues like and , adopt transient conformations that facilitate rapid, adaptive to , often transitioning to partially ordered states upon complex formation. In hnRNPs and , IDRs such as or RG motifs enable the protein to scan extended RNA stretches or assemble into dynamic ribonucleoprotein complexes, underscoring their role in modulating interaction potential without rigid scaffolding. RBPs display a broad spectrum of sizes and complexities, reflecting their diverse roles across cellular contexts. Small, single-domain RBPs, such as certain coat proteins or isolated RRMs, typically range from 10 to 20 kDa, allowing compact and efficient RNA encapsulation in genomes. In contrast, eukaryotic RBPs like spliceosomal components (e.g., Prp8 at ~280 kDa) form large, multi-subunit assemblies exceeding 100 kDa per subunit, incorporating enzymatic and regulatory domains to coordinate complex processing machineries. This architectural variability—from minimalist effectors to elaborate cellular hubs—directly influences the scope and regulation of engagement. Insights into these architectural features have been derived primarily from and (NMR) spectroscopy, which highlight the dichotomy between folded RBDs and disordered regions. X-ray structures, such as that of the U1A RRM at 1.92 resolution, reveal compact β-α-β sandwiches ideal for sequence-specific contacts, while NMR studies capture the conformational ensembles of IDRs in hnRNPs, demonstrating their entropy-driven flexibility. These methods have collectively illuminated how modular designs balance stability and adaptability in RBP function.

Key RNA-Binding Domains

RNA-binding proteins (RBPs) employ a variety of modular domains to recognize and interact with RNA molecules, with the RNA recognition motif (RRM) being one of the most abundant and versatile. The RRM typically adopts a β-α-β-β-α-β fold spanning approximately 80-90 , featuring two conserved sequences known as RNP1 and RNP2 located on the central β-strands β3 and β1, respectively. These motifs contain aromatic residues, such as and , that facilitate binding to single-stranded RNA through base-stacking interactions and hydrogen bonding with the RNA backbone. A classic example is the U1 snRNP 70K protein, where the RRM binds to the 5' splice site of pre-mRNA via these conserved elements. Another key domain is the motif, a compact structure of about 70 characterized by a β-α-α-β-β-α fold with an invariant GXXG loop positioned between α-helices for gripping nucleic acids. This loop enables the KH domain to contact RNA phosphate groups, often targeting stems or loops in structured . The KH domain is exemplified in proteins like , which uses tandem KH motifs (KH3 and KH4) to recognize YCAY RNA clusters, and fragile X mental retardation protein (FMRP), where KH1 and KH2 domains bind G-quadruplexes or stem-loops in target mRNAs. Zinc finger domains, particularly the CCCH-type, coordinate zinc ions via and residues to stabilize flexible loops that directly engage . In tristetraprolin (TTP), the tandem CCCH zinc fingers form a compact structure that binds AU-rich elements (AREs) in the 3' untranslated regions of mRNAs, promoting their decay through recruitment of decay machinery. These domains typically feature three s and one per finger, enabling high-affinity, sequence-specific interactions. Additional domains expand the repertoire of RNA recognition; the double-stranded RNA-binding motif (dsRBM) consists of a 65-70 amino acid α-β-β-β-α fold that preferentially binds double-stranded RNA structures, inserting into the major and minor grooves via conserved lysine and arginine residues. This motif is crucial for proteins like ADAR enzymes that edit dsRNA. The Pumilio/FBF (PUF) domain, in contrast, features 8-12 tandem repeats of a 36-amino-acid helix-loop-helix structure that achieves sequence-specific binding to 3' UTRs through a modular code, where each repeat recognizes a single nucleotide via side-chain interactions. Pumilio proteins, for instance, bind UGUANAUA motifs to repress translation in developmental contexts. Many RBPs enhance binding affinity through combinations of domains, leveraging modular architectures for cooperative interactions. In fused in sarcoma (FUS), the RRM pairs with flanking arginine-glycine-glycine (RGG) boxes—intrinsically disordered regions rich in arginines that form electrostatic contacts with RNA phosphates—resulting in a 100-fold increase in affinity compared to the RRM alone (from >90 μM to 0.7 μM). Such synergies allow RBPs to achieve specificity and beyond single-domain capabilities, often integrating structured and disordered elements.

Diversity and Classification

Structural and Functional Diversity

RNA-binding proteins (RBPs) exhibit remarkable structural diversity, primarily classified by their domain composition, such as those containing the RNA recognition motif (RRM), the most prevalent RNA-binding domain in eukaryotes, versus non-RRM RBPs that rely on other motifs or intrinsically disordered regions for RNA interactions. This domain-based scheme highlights how RRMs, often found in multiple copies within a single protein, enable versatile RNA recognition, while non-RRM RBPs, including those with or domains, expand the repertoire of binding specificities. Functionally, RBPs are categorized by roles in processes, such as ribosomal proteins that facilitate core translation machinery, versus regulatory functions, exemplified by miRNA-binding proteins like that modulate post-transcriptional . In humans, studies have identified approximately 1,542 RBPs, accounting for approximately 7.5% of the protein-coding , with these proteins distributed across cellular compartments and essential for RNA metabolism in eukaryotes. This abundance underscores their ubiquity, extending to prokaryotes through orthologs like the bacterial Hfq protein, which binds small regulatory RNAs to influence in diverse lineages. RBPs are further delineated into core and accessory categories based on their involvement in fundamental versus specialized RNA processing; core RBPs form stable complexes in machineries like the , while accessory RBPs dynamically associate to fine-tune outcomes. Representative examples illustrate this functional spectrum: ubiquitously expressed heterogeneous nuclear ribonucleoproteins (hnRNPs) act as core regulators of pre-mRNA splicing and stability across tissues, whereas tissue-specific RBPs like muscleblind (MBNL) proteins predominantly function in striated muscle to control of developmental transcripts. MBNL's restricted expression highlights how RBPs adapt to cellular contexts, promoting muscle through targeted interactions. Beyond canonical RBPs, non-canonical examples include metabolic enzymes, such as glycolytic proteins like glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and , which bind mRNAs to regulate their localization and stability in addition to catalytic roles. These enzymes, lacking traditional RNA-binding domains, demonstrate how evolutionary repurposing enables RBPs to integrate with RNA control, often in stress-responsive pathways.

Evolutionary Aspects

RNA-binding proteins (RBPs) are believed to have ancient origins, tracing back to the hypothesized , where molecules served both informational and catalytic roles before the emergence of DNA and proteins. In this primordial scenario, early peptides likely acted as cofactors enhancing functionality, laying the groundwork for protein- interactions that evolved into modern RBPs. This evolutionary foundation is evidenced by the presence of conserved RNA-binding domains across all domains of life, including the RNA recognition motif (RRM), which is found in prokaryotes as small, single-domain proteins approximately 100 in length. For instance, the archaeal L7Ae protein, a component of the box C/D ribonucleoprotein complex involved in RNA modification, exhibits an RRM-like fold that binds kink-turn RNA structures, highlighting deep conservation from to eukaryotes. Similarly, bacterial RRMs, such as those in ribonuclease P, bind pre-tRNA substrates, underscoring the motif's role in fundamental RNA processing across prokaryotes. The transition to eukaryotic complexity involved significant expansions of RBP families through events, which enabled greater regulatory diversity in metabolism. In particular, the serine/arginine-rich ( family, key regulators of pre-mRNA splicing, proliferated via tandem duplications in metazoans, correlating with the increased density and complexity observed in multicellular eukaryotes. This expansion allowed to evolve specialized roles in coordinating assembly and enhancing splicing fidelity, adapting to the demands of more intricate networks. Such duplications not only diversified RBP functions but also facilitated tissue-specific , as seen in the broader repertoire of in vertebrates compared to simpler eukaryotes. Species-specific adaptations further illustrate RBP evolution, with certain proteins gaining specialized functions or being lost in response to ecological niches. Highly conserved RBPs like play prominent roles in neuronal RNA processing in vertebrates, including mRNA stabilization and splicing of genes critical for synaptic function and neuronal integrity, reflecting adaptations to complex nervous systems. Conversely, some parasites exhibit losses of specific RBPs, such as components of the pathway—including and proteins—due to genome streamlining and reduced selective pressure from transposons, as observed in protozoans like and some species (e.g., T. cruzi). These losses highlight how evolutionary pressures, including host-parasite interactions, can lead to divergent RBP repertoires. RBPs have in parallel with non-coding RNAs, particularly in protection mechanisms. The () pathway exemplifies this, where clade proteins bind piRNAs to silence transposons, preventing genomic instability during ; this system has undergone adaptive evolution across species to counter rapidly mutating transposable elements. Such is evident in the expansion and diversification of piRNA-binding proteins in metazoan germlines, ensuring reproductive fitness amid ongoing transposon invasions.

Functions in RNA Metabolism

Pre-mRNA Processing and Modification

RNA-binding proteins (RBPs) play essential roles in the co-transcriptional of pre-mRNA, ensuring the maturation of transcripts into functional mRNAs through mechanisms such as capping, splicing, , and . These nuclear events occur in coordination with transcription, where RBPs recognize specific sequence elements to recruit processing machinery and regulate efficiency. For instance, the RNA recognition motif (RRM) domains common in many RBPs facilitate initial binding to pre-mRNA targets. Dysregulation of these RBP-mediated processes can lead to aberrant isoform production and cellular dysfunction. In alternative splicing, RBPs like SR proteins and heterogeneous nuclear ribonucleoproteins (hnRNPs) antagonistically control exon inclusion or skipping, impacting the diversity of protein isoforms. SR proteins, such as SRSF1 (formerly SFRS1), bind to exonic splicing enhancers (ESEs) via their RNA-binding domains, recruiting the to promote exon inclusion and influencing the splicing of numerous transcripts. In contrast, hnRNPs, including hnRNP A1 and hnRNP L, bind to exonic splicing silencers (ESSs) to repress splicing by blocking splice site recognition or competing with for binding sites. This regulatory balance affects approximately 95% of human multi-exon genes, enabling tissue-specific and developmental isoform variation. RNA editing by RBPs introduces post-transcriptional modifications that alter coding potential without changing the genomic sequence. The family of RBPs, particularly ADAR1 and ADAR2, bind double-stranded structures in pre-mRNA and catalyze adenosine-to-inosine (A-to-I) , which is interpreted as during . A prominent example is ADAR2-mediated editing at the Q/R in the GluR2 subunit of receptors, converting a codon to and thereby modulating calcium permeability essential for synaptic function. Polyadenylation involves RBPs that define the 3' end of pre-mRNA, adding a poly(A) tail that enhances mRNA stability and export competence. The and specificity (CPSF) complex binds the polyadenylation signal (typically AAUAAA), while the (CstF) recognizes GU- or U-rich downstream elements to position the site accurately. These interactions stimulate endonucleolytic followed by poly(A) polymerase addition of the tail, with CPSF and CstF subunits like CPSF73 and CstF64 serving as key RNA-binding effectors. Capping at the 5' end links transcription initiation to via the cap-binding complex (CBC), composed of CBP80 and CBP20. Shortly after the nascent pre-mRNA emerges from , CBC binds the 7-methylguanosine cap structure, stabilizing the transcript and facilitating interactions with splicing and factors to coordinate overall maturation. This binding enhances processing efficiency and prevents premature .

mRNA Export, Localization, and Stability

RNA-binding proteins (RBPs) play essential roles in the nuclear export of mature mRNAs, ensuring their safe passage through nuclear pore complexes (NPCs) to the . The primary export receptor, the NXF1:NXT1 heterodimer (also known as TAP:p15), recognizes and binds to mRNA via interactions with adaptor proteins associated with the transcript. Specifically, NXF1:NXT1 docks onto the (EJC), a multiprotein assembly deposited ~20-24 upstream of exon-exon junctions during splicing, which marks the mRNA for export. The complex, recruited to the mRNA during transcription elongation, further facilitates this process by bridging the transcription machinery to the export factors; its components, such as ALYREF and THO, promote the handover of the mRNA from the EJC to NXF1:NXT1, exposing NXF1's RNA-binding domain for stable association. Once bound, NXF1:NXT1 interacts with nucleoporins like NUP153 and NUP98 in the NPC, enabling directional transport of the mRNP (messenger ribonucleoprotein) particle while preventing back-diffusion. This coordinated mechanism ensures efficient bulk export of most cellular mRNAs, with defects in NXF1 or TREX components leading to nuclear retention and cellular stress. In the cytoplasm, RBPs mediate the spatial localization of mRNAs to specific subcellular compartments, allowing localized and functional compartmentalization. A prominent example is zipcode-binding protein 1 (ZBP1, also known as IGF2BP1), which binds to "zipcode" motifs in the 3' untranslated region (UTR) of target mRNAs, such as β-actin mRNA. In neurons, ZBP1 facilitates the of these mRNAs along to dendrites via interactions with motor proteins like kinesin-1; this process is activity-dependent, with neuronal stimulation promoting ZBP1 release and local of β-actin for cytoskeletal remodeling at synapses. ZBP1's RNA-recognition motifs (RRMs) and K-homology () domains confer specificity to bipartite zipcode elements, ensuring precise dendritic localization essential for neuronal plasticity. Disruption of ZBP1 binding impairs mRNA trafficking, resulting in deficits in axonal and dendritic growth. RBPs also regulate mRNA stability by modulating decay rates, thereby controlling duration. The RBP HuR (human antigen R, ELAVL1) binds to AU-rich elements () in the 3' UTRs of proto-oncogene and mRNAs, such as c-fos and TNF-α, promoting their stabilization by competing with decay factors and enhancing nuclear export or cytoplasmic retention. HuR's three RRMs enable high-affinity binding to UUAUUUAUU motifs within , preventing deadenylation and exonucleolytic degradation; this stabilizes transcripts for hours to days, influencing and . In contrast, tristetraprolin (TTP, ZFP36) accelerates decay of ARE-containing mRNAs by recruiting the CCR4-NOT deadenylation complex via its zinc-finger domains, which recognize similar UAUUUAU sequences. TTP bridges the mRNA to the deadenylase subunit CNOT1, initiating poly(A) tail shortening followed by and 5'-3' degradation; phosphorylation by p38 MAPK inhibits this recruitment, transiently stabilizing targets during immune responses. A specialized stability control mechanism involves (NMD), where RBPs detect and degrade mRNAs with premature termination codons (PTCs) to prevent truncated protein production. The UPF proteins—UPF1 (an RNA helicase), UPF2, and UPF3—form the core NMD machinery; UPF3 associates with the EJC at junctions, while UPF2 bridges UPF3 to UPF1, which is recruited during pioneering round . Upon encountering a PTC >50 upstream of an junction, UPF1 phosphorylates via SMG1 , triggering mRNA ubiquitination, by DCP2, and degradation by XRN1 or the exosome. This process not only quality-controls transcripts but also regulates ~5-10% of the human , including tumor suppressors.

Translation and Post-Transcriptional Regulation

RNA-binding proteins (RBPs) play pivotal roles in regulating translation, the process by which mRNA is decoded to synthesize proteins, and in post-transcriptional mechanisms that fine-tune gene expression beyond initial RNA synthesis. These proteins interact with specific mRNA elements to modulate ribosome recruitment, initiation, elongation, and termination, ensuring translation occurs in a controlled manner responsive to cellular conditions. By binding to untranslated regions (UTRs) or structured RNA motifs, RBPs can enhance or repress protein production, integrating signals from metabolism, stress, and signaling pathways. A key aspect of cap-dependent translation initiation involves the eukaryotic initiation factor 4E (eIF4E), an RBP that specifically recognizes the 7-methylguanosine (m7G) cap at the 5' end of mRNA. eIF4E binding facilitates the recruitment of the 40S ribosomal subunit by bridging the mRNA cap to the eIF4F complex, which includes the scaffold protein eIF4G and the helicase eIF4A, thereby unwinding secondary structures to enable scanning for the start codon. Complementing this, the poly(A)-binding protein (PABP) binds the poly(A) tail at the mRNA 3' end, interacting with eIF4G to circularize the mRNA and promote efficient ribosome reinitiation and translation stimulation. This closed-loop configuration enhances translational efficiency by stabilizing the mRNA and coordinating 5' and 3' end interactions. RBPs also mediate translational repression, as exemplified by the iron-responsive element-binding protein (IRE-BP), also known as iron regulatory protein 1 (IRP1). Under low iron conditions, IRE-BP binds to iron-responsive elements (IREs), which are stem-loop structures in the 5' UTR of mRNAs encoding proteins like and aminolevulinic acid synthase. This binding sterically hinders scanning and initiation, thereby repressing to conserve iron for essential cellular processes. Iron availability modulates IRE-BP activity through conformational changes, relieving repression when iron levels rise. In , RBPs are central to (miRNA)-mediated silencing, where (AGO) proteins form the core of the (RISC). AGO proteins, such as AGO2 in mammals, bind miRNA-mRNA hybrids via base-pairing between the miRNA seed sequence and complementary sites typically in the mRNA 3' UTR, leading to translational repression or mRNA destabilization. This mechanism allows precise control of , with RISC recruitment inhibiting ribosome association and promoting deadenylation or . Alternative translation pathways, such as (IRES)-mediated initiation, rely on IRES-transacting factors (ITAFs) like polypyrimidine tract-binding protein (PTB) to facilitate cap-independent translation under stress conditions. PTB binds structured IRES elements in the 5' UTR of specific mRNAs, such as those for c-Myc or HIF-1α, recruiting ribosomes directly to internal start sites when cap-dependent mechanisms are impaired, such as during or viral infection. This enables selective protein synthesis to support cell survival and adaptation.

Mechanisms of Protein-RNA Interactions

Binding Motifs and Specificity

RNA-binding proteins (RBPs) achieve specificity in their interactions with RNA through diverse molecular motifs that recognize particular sequences or structural features. Sequence-specific binding often involves motifs rich in or , such as the UG repeats preferred by (TDP-43). TDP-43's two RNA recognition motifs (RRMs) cooperatively engage UG-rich sequences, with a minimum of six UG dinucleotides required for high-affinity binding, and affinity increasing with additional repeats due to inter-RRM stabilization upon RNA contact. Similarly, PUF family proteins utilize base-specific pockets within their Pumilio homology domains to recognize core motifs like UGCAC, where individual repeats contact specific through hydrogen bonding and stacking interactions, enabling modular sequence discrimination. Structural recognition motifs allow RBPs to target RNA secondary elements independent of primary sequence. For instance, the double-stranded RNA-binding motifs (dsRBMs) in protein kinase R (PKR) selectively bind stem-loop structures in viral RNAs, with the tandem dsRBMs clamping around A-form helical regions to detect double-stranded features longer than 10 base pairs. In ribosomal contexts, certain ribosomal proteins, such as S4 from Escherichia coli, recognize pseudoknot structures in their own mRNAs for autoregulation, where the protein's RNA-binding surface interacts with the pseudoknot's stacked helices and loop regions to inhibit translation. Combinatorial binding enhances specificity and affinity by integrating multiple motifs. TIAR, for example, employs three RRMs that cooperatively bind U-rich tracts, with the linker region between RRMs2 and RRMs3 contributing essential residues for nanomolar-affinity interactions with polyuridine sequences, allowing the protein to span extended RNA stretches. This multivalent engagement ensures selective targeting of U-rich elements in stress response mRNAs. Post-translational modifications further modulate binding motifs and specificity. can alter RBP affinity by changing motif accessibility or conformation; for instance, phosphorylation of the cytoplasmic polyadenylation element-binding protein (CPEB) by Aurora kinase (Eg2) enables recruitment of cleavage and specificity factor (CPSF) to stimulate poly(A) tail elongation of oocyte mRNAs, thereby regulating translational activation during development. Such modifications provide dynamic control over RBP-RNA interactions in response to cellular signals.

Dynamic Interaction Models

RNA-binding proteins (RBPs) exhibit transient associations with RNA, characterized by dynamic on-off kinetics that govern their regulatory roles. (SPR) has been instrumental in quantifying these kinetics, revealing association rates (k_on) typically in the range of 10^4 to 10^6 M^{-1} s^{-1} and rates (k_off) from 10^{-3} to 10^{-1} s^{-1} for various RBP-RNA complexes, such as those involving the iron-responsive element-binding protein (IRP1) with its RNA stem-loop. These measurements highlight how rapid binding and unbinding enable RBPs to scan and select specific RNA targets efficiently, with dissociation constants (K_D) often in the nanomolar to micromolar range, allowing for responsive regulation in cellular processes. A prominent example of dynamic interactions is the role of intrinsically disordered regions (IDRs) in driving liquid-liquid (LLPS), which concentrates RBPs and RNAs into membraneless compartments like . In G3BP1, an RBP central to assembly, the IDR acts as a tunable switch: under , elevated free concentrations trigger RNA-dependent LLPS by promoting multivalent interactions, leading to rapid condensate formation with phase separation occurring within seconds to minutes. This process is reversible, with disassembly upon stress relief, underscoring the transient nature of these assemblies that facilitate localized RNA regulation without fixed structures. Allosteric regulation further modulates RBP-RNA dynamics by linking binding at one site to conformational changes at another. In the spliceosomal U2AF heterodimer, comprising U2AF35 and U2AF65, binding of U2AF35 to U2AF65 via its UHM ligand motif allosterically enhances U2AF65's affinity for weak polypyrimidine tract RNAs, shifting the population toward an open conformation that promotes 3' splice site recognition and spliceosome assembly. This cooperative mechanism ensures precise timing in pre-mRNA splicing, where initial low-affinity interactions are stabilized through allosteric effects. Cellular environments profoundly influence these interactions through factors like , , , and inter-RBP competition. Elevated salt concentrations (e.g., 100-500 mM NaCl) screen electrostatic interactions, reducing RBP-RNA affinity as seen in the p19 protein's siRNA binding, where K_D increases from nanomolar at low salt to micromolar at high salt; similarly, shifts, such as acidification under , alter states and weaken binding in RBPs like TRBP2. , mimicking the cytosol's 300-400 mg/mL biomacromolecules, enhances association rates and stability by effects, boosting affinity up to 10-fold for RBPs like those in phase-separating condensates. Competition arises when multiple RBPs vie for overlapping RNA sites, as in the case of CELF1 and HuR contesting mRNA 3' UTR elements, where kinetic off-rates determine occupancy and downstream translational outcomes. Single-molecule (smFRET) provides insights into these conformational dynamics, revealing how binding induces rapid structural rearrangements in RBPs. For instance, smFRET studies of disordered RBPs like FUS show that engagement reduces inter-domain distances from ~6-8 nm in the apo state to ~4-5 nm upon binding, with transition times on the millisecond scale, reflecting coupled folding and binding mechanisms. These observations elucidate how environmental cues and kinetic competition integrate to fine-tune RBP function at the single-event level.

Roles in Development

Embryonic and Germline Development

RNA-binding proteins (RBPs) play pivotal roles in embryonic and development by regulating RNA localization, stability, and to ensure proper fate specification and . In the , Pumilio, an RBP, binds to the 3' untranslated regions (UTRs) of target mRNAs, including those involved in posterior patterning, to repress and maintain polarity. Specifically, Pumilio collaborates with Nanos to repress maternal hunchback mRNA in the posterior of the and early , preventing ectopic that would disrupt abdominal development; this interaction is essential for establishing the anterior-posterior axis. In mammals, Vasa, an ATP-dependent DEAD-box helicase RBP, is crucial for germline specification and transposon silencing via the pathway. Vasa facilitates the assembly of effector complexes in germ cells, promoting the ping-pong amplification cycle that cleaves transposon transcripts and safeguards genome integrity during . Mutations in Vasa lead to defects in biogenesis and increased transposon activity, resulting in sterility. Additionally, Deleted in Azoospermia-Like (DAZL), another RBP, promotes meiotic progression in human by binding and stabilizing mRNAs encoding meiosis-related proteins, such as those involved in formation; DAZL deficiency causes meiotic arrest and infertility. For totipotency maintenance in early embryos, RBPs associated with the pluripotency factor OCT4 regulate transcripts by modulating their post-transcriptional processing. OCT4 interacts with RBPs like heterogeneous nuclear ribonucleoprotein K (hnRNP K), which binds to and stabilizes mRNAs of pluripotency genes, ensuring their availability in embryonic s and preventing premature . In model organisms like , maternal RBPs contribute to anterior-posterior gradients by localizing and translating patterning mRNAs; for instance, RBPs such as Vasa and DAZL-like factors help establish polarity in oocytes, analogous to Bicoid-mediated gradients in . In C. elegans, P granules, structures, sequester germline-specific mRNAs via RBPs like MEG-3, an intrinsically disordered protein that forms condensates to protect and localize transcripts for fate, excluding RNAs during early cleavages.

Somatic and Neuronal Development

RNA-binding proteins (RBPs) play critical roles in post-embryonic by regulating events that drive tissue-specific maturation. In , Muscleblind-like (MBNL) proteins, particularly MBNL1 and MBNL2, orchestrate splicing switches essential for muscle development. These RBPs promote the inclusion of adult-specific exons in pre-mRNAs encoding contractile proteins, such as cardiac (cTNT) and α-tropomyosin, thereby facilitating the transition from fetal to mature muscle isoforms during myoblast . Loss of MBNL function disrupts this process, leading to aberrant splicing patterns observed in models, underscoring its necessity for proper formation. Similarly, in cardiac development, RBFOX1 (also known as Fox-1) regulates tissue-specific by binding to (U)GCAUG elements in pre-mRNAs, influencing the expression of genes involved in heart and function, such as those encoding channels and structural proteins. RBFOX1 in models results in cardiac dysfunction and increased susceptibility to , highlighting its indispensable role in cardiogenesis beyond initial embryonic stages. In neuronal development, RBPs contribute to synapse formation and circuit refinement through precise control of mRNA processing and localization. proteins ( and ), neuron-specific RBPs with domains, direct of pre-mRNAs encoding synaptic components, ensuring the proper assembly of neuronal s. For instance, regulates the splicing of SNAP-25, a t-SNARE protein critical for fusion, by promoting neuron-specific isoforms that support synaptic maturation in the developing . This splicing regulation extends to other synaptic proteins, such as neuroligins and channels, where binding to YCAY motifs silences cryptic exons, preventing and enabling functional protein diversity. Complementing this, Fragile X mental retardation protein (FMRP) acts as a translational in dendrites, binding to ~4% of mRNAs, including those for PSD-95 and MAP1B, to suppress local protein synthesis until synaptic stimulation. In FMRP-deficient models of , excessive dendritic translation disrupts spine morphology and circuit refinement, emphasizing FMRP's role in activity-dependent neuronal maturation. Further refinement of neural circuits involves RBPs in mRNA transport and neuronal maturation. Staufen2 facilitates the microtubule-dependent transport of mRNAs, such as β-actin and MAP1B, into axons, supporting dynamics and cues during circuit assembly. This transport mechanism allows localized translation in response to extracellular signals, guiding axonal and in post-mitotic neurons. Likewise, ELAV/Hu family proteins, including HuB (ELAVL2) and HuC (ELAVL3), promote neuronal maturation by stabilizing and enhancing translation of mRNAs involved in outgrowth and synapse stabilization, such as subunits. Overexpression of in neural progenitors accelerates , while depletion delays maturation, as seen in Hu knockout mice exhibiting impaired neuronal and . In humans, mutations in RBPs like FUS disrupt these processes, leading to developmental delays; for example, FUS variants alter mechanics and dynamics in iPSC-derived neurons, contributing to early synaptic deficits and motor delays observed in pediatric cases.

Roles in Disease

Involvement in Cancer

Dysregulation of RNA-binding proteins (RBPs) plays a pivotal role in oncogenesis by altering RNA , , and , thereby promoting tumor , progression, and resistance to . In cancer cells, aberrant expression or activity of RBPs disrupts post-transcriptional , leading to the accumulation of pro-oncogenic transcripts and suppression of tumor-suppressive pathways. This involvement spans multiple , including sustained proliferation, evasion of , and metastatic dissemination, making RBPs attractive therapeutic targets. Splicing aberrations driven by RBPs are a common feature in tumorigenesis, where altered generates isoforms that favor cancer cell survival and growth. For instance, overexpression of the serine/arginine-rich splicing factor 1 (SRSF1) promotes the inclusion of pro-survival exons in genes such as Bcl-x, resulting in the production of the anti-apoptotic isoform , which inhibits and enhances cell survival in . Similarly, SRSF1 upregulation in drives oncogenic splice switching of PTPMT1, contributing to tumor progression. Heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1), another key splicing regulator, stabilizes mRNA by binding to its 3' untranslated region, thereby increasing MYC protein levels and promoting proliferation in various cancers, including colorectal . These splicing changes, often linked to alternative splicing mechanisms, underscore how RBPs like SRSF1 and hnRNP A1 act as proto-oncogenes by favoring oncogenic isoforms. In proliferation control, RBPs modulate mRNA stability to enhance angiogenic and growth signaling pathways essential for tumor expansion. The RNA-binding protein HuR (ELAVL1) stabilizes vascular endothelial growth factor (VEGF) mRNA by binding to AU-rich elements in its 3' untranslated region, particularly under hypoxic conditions prevalent in tumors, thereby promoting angiogenesis and tumor vascularization in colorectal and breast cancers. Conversely, the STAR family protein Quaking (QKI) acts as a tumor suppressor by facilitating the biogenesis of circular RNAs (circRNAs) that sequester oncogenic factors, thereby inhibiting cell proliferation and inducing apoptosis. Loss of QKI expression correlates with reduced circRNA levels and increased tumor growth in multiple solid tumors. RBPs also influence metastasis by regulating epithelial-mesenchymal transition (EMT) and stress responses that enable invasion and dissemination. Z-DNA binding protein 1 (ZBP1) inhibits invasion by maintaining cell polarity and suppressing chemotaxis in breast cancer cells. Downregulation of ZBP1 enhances metastatic potential by disrupting this control. Likewise, loss of muscleblind-like 1 (MBNL1) promotes EMT by destabilizing Snail mRNA and altering splicing of EMT-related genes, leading to increased migration and colonization in breast and colorectal cancers; restoration of MBNL1 suppresses metastatic relapse. Therapeutic strategies targeting RBPs in cancer focus on small molecules that disrupt RNA-binding domains, such as the RNA recognition motif (RRM), to restore normal . For example, the inhibitor MS-444 targets the RRM of HuR, reducing mRNA stabilization of oncogenic transcripts in and solid tumors, with preclinical studies showing efficacy in suppressing proliferation. Emerging approaches include compounds mimicking TDP-43 loss-of-function effects in , where TDP-43 knockdown inhibits progression; therapeutic development for TDP-43 modulation is ongoing. These targeted interventions highlight the potential of RBP modulation to overcome therapy resistance in cancers driven by splicing and stability defects.

Implications in Neurodegenerative and Other Diseases

RNA-binding proteins (RBPs) play critical roles in neurodegenerative diseases, particularly through mechanisms involving and disruption of RNA processing. In (ALS) and (FTD), (TDP-43) undergoes liquid-liquid phase separation (LLPS) leading to cytoplasmic aggregation, which depletes nuclear TDP-43 and impairs , , and essential for neuronal function. This aggregation is a pathological hallmark observed in over 95% of ALS cases and approximately 50% of FTD cases, contributing to neuronal loss via toxic gain-of-function and loss-of-function effects on RNA metabolism. Similarly, fused in sarcoma (FUS) mutations in ALS cause nuclear depletion of FUS, resulting in widespread RNA processing defects including aberrant splicing and reduced recruitment of repair factors to DNA damage sites in motor neurons. These mutations disrupt FUS's normal nuclear localization, leading to cytoplasmic accumulation and impaired of genes critical for neuronal development and maintenance. In autoimmune diseases, RBPs contribute to immune dysregulation by influencing RNA-mediated tolerance and splicing fidelity. The Ro60 RBP, also known as SSA, binds non-coding RNAs such as Y RNAs to maintain , and its dysfunction in Sjögren's syndrome promotes through altered RNA processing and autoantibody production targeting Ro60 ribonucleoproteins. Autoantibodies against Ro60 are diagnostic markers in Sjögren's syndrome and correlate with glandular and interferon signaling dysregulation. Likewise, heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) serves as a major autoantigen in systemic lupus erythematosus (SLE), where immune targeting leads to mis-splicing of autoimmunity-related genes and enhanced T-cell reactivity, exacerbating systemic . This mis-splicing disrupts the regulation of inflammatory transcripts, contributing to the autoimmune phenotype observed in SLE patients. RBPs are also central to host defenses and viral evasion strategies during infections. Protein kinase R (PKR), an interferon-inducible RBP, detects double-stranded (dsRNA) produced by viruses, leading to its dimerization, autophosphorylation, and activation of an antiviral response that phosphorylates eIF2α to inhibit global while promoting formation. This mechanism restricts but can be modulated by pathogens to avoid detection. In infection, the viral non-structural protein 1 (NSP1) hijacks host RBPs such as the mRNA export factor NXF1 to block nuclear export of host mRNAs, thereby suppressing host and facilitating immune evasion through selective of viral RNAs. NSP1's interaction with the ribosomal mRNA channel further enhances this shutdown, allowing the virus to repurpose host RBPs for its lifecycle while evading innate antiviral responses. Beyond neurodegeneration and immunity, RBPs influence cardiovascular pathologies through splicing regulation. Mutations in RNA-binding motif protein 20 (RBM20) cause by inducing splicing defects in the titin (TTN) gene, leading to truncated titin isoforms that impair structure and cardiac contractility. These gain-of-function mutations in RBM20's RS domain result in hyperphosphorylation and mislocalization, exacerbating imbalances in cardiac-specific transcripts and promoting arrhythmogenic remodeling.

Current Research Directions

Experimental Techniques and Tools

The identification of RNA-binding proteins (RBPs) has been revolutionized by ribonomics approaches that leverage ultraviolet (UV) crosslinking to covalently link proteins to RNA in vivo, followed by purification and liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. A seminal method, known as interactome capture, involves UV crosslinking of cells, lysis, RNase treatment to fragment RNA, and oligo(dT)-based purification of polyadenylated RNA-protein complexes before LC-MS/MS identification of associated proteins. This technique, applied to human HeLa cells, identified 860 RBPs, expanding the known human RBP repertoire and highlighting previously uncharacterized proteins involved in RNA metabolism. Building on such efforts, a comprehensive 2014 census integrated interactome capture data with computational predictions and literature curation to catalog 1,542 human RBPs, providing a foundational resource for subsequent studies. More recent analyses, integrating additional data, estimate the human RBP repertoire at around 2,500 proteins. For mapping in vivo RBP binding sites, RNA immunoprecipitation followed by sequencing (RIP-seq) captures direct protein-RNA interactions under native conditions, enabling transcriptome-wide identification of bound RNAs. In RIP-seq, cells are lysed, RBP-RNA complexes are immunoprecipitated using specific antibodies, and associated RNAs are extracted, reverse-transcribed, and sequenced to reveal binding targets without crosslinking artifacts. This method has been instrumental in elucidating context-specific RBP functions, such as in stress responses where RBPs like TIA1 bind to AU-rich elements in mRNAs. Crosslinking and immunoprecipitation (CLIP) variants offer nucleotide-resolution mapping of RBP footprints by incorporating UV crosslinking to stabilize interactions and partial RNase digestion to generate RNA fragments centered on binding sites. High-throughput sequencing of RNA isolated by crosslinking immunoprecipitation (HITS-CLIP), introduced in 2008, sequences cDNA tags from immunoprecipitated complexes, clustering reads to identify enriched binding regions and infer motifs. Individual-nucleotide resolution CLIP (iCLIP), an advancement from 2014, improves efficiency through adaptive polymerase truncation and circularization, reducing biases and enabling precise footprint detection for RBPs like FUS/TLS in neuronal RNAs. Photoactivatable ribonucleoside-enhanced CLIP (PAR-CLIP) incorporates 4-thiouridine (4SU) labeling into nascent , inducing T-to-C transitions at crosslinking sites during reverse transcription for unambiguous mutation-based mapping of binding positions, as demonstrated for proteins in miRNA pathways. Functional validation of RBP roles often employs reporter minigene assays, particularly for splicing regulation, where hybrid constructs containing RBP-responsive exons are transfected into s to monitor inclusion/exclusion via RT-PCR or fluorescence. For instance, minigenes with binding sites recapitulated splicing patterns observed in vivo, confirming the protein's role in neuronal . CRISPR-based knockdown complements this by using guide RNAs to disrupt RBP genes, assessing phenotypic outcomes like altered processing or viability; early applications in 2013 validated essentiality of RBPs such as UPF1 in . Emerging tools extend RBP analysis to single-cell and computational . Single-cell RIP-seq variants, such as those combining nano-aptamer pulldowns with barcoded sequencing, isolate RBP-bound RNAs from individual cells, revealing heterogeneity in binding, as shown for HuR in immune cell populations. Post-2020 advancements integrate AlphaFold-predicted structures with to forecast RBP-RNA interfaces; recent developments, including AlphaFold3 (2024), further enable predictions of protein-RNA complexes, though accuracy for RNA structures remains under evaluation; for example, models trained on AlphaFold2 outputs predict binding affinities for motifs in proteins like TDP-43, aiding hypothesis generation for uncharacterized interactions.

Therapeutic and Diagnostic Applications

RNA-binding proteins (RBPs) have emerged as key targets in therapeutic strategies for diseases involving dysregulated RNA processing, particularly those linked to aberrant splicing and . One prominent example is the use of (ASOs) to correct splicing defects caused by SMN2 mutations in (), a neurodegenerative disorder resulting from insufficient SMN protein production. , an ASO that binds to an intronic splicing silencer in SMN2 pre-mRNA to promote 7 inclusion and increase full-length SMN expression, was approved by the FDA in December 2016 for treating pediatric and adult patients via . Clinical trials, such as the ENDEAR and CHERISH studies, demonstrated significant improvements in motor function and survival rates compared to , establishing as the first approved therapy for . Small-molecule inhibitors targeting RBP aggregation represent another avenue, especially for (ALS), where TDP-43 mislocalization and cytoplasmic aggregates disrupt RNA metabolism and contribute to death. Compounds like (SLS-005), which activates via TFEB to clear TDP-43 aggregates, completed a Phase 2/3 clinical trial for ALS (NCT05136885) in 2024, which did not meet its primary endpoint of slowing disease progression despite preclinical promise, but demonstrated good tolerability. Similarly, (MN-116), a that enhances lysosomal biogenesis and reduces TDP-43 pathology, is advancing in Phase 2/3 trials (NCT04057898), with evidence of in ALS patients. These approaches aim to restore TDP-43 function and mitigate neurodegeneration. In diagnostics, RBP-derived signatures in liquid biopsies offer non-invasive tools for early cancer detection by capturing tumor-released RNA regulators in . Circulating heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1), an RBP overexpressed in various malignancies and involved in and mRNA stabilization, has been identified as a biomarker for , with elevated levels correlating to tumor stage and aiding in alongside CEA. Proteomic analyses of from patients revealed hnRNP A1 as a potential indicator for early detection and monitoring, distinguishable from healthy controls. For neurodegeneration, components of the miRNA-induced silencing complex (miRISC), such as 2 (AGO2), are detectable in circulation and associated with disease-specific miRNA profiles in . Extracellular miRNAs bound to AGO2 in plasma reflect neuronal stress and serve as prognostic biomarkers, with dysregulated miRISC activity linked to TDP-43 pathology and motor neuron loss. Gene therapy approaches leverage viral vectors to deliver or edit RBPs, addressing genetic deficiencies in neurodevelopmental and cardiac disorders. Adeno-associated virus (AAV) vectors encoding the FMR1 gene have shown promise in fragile X syndrome models, where loss of FMRP—an RBP that regulates mRNA translation—leads to synaptic dysfunction and intellectual disability. Systemic AAV9-FMR1 administration in adult Fmr1 knockout mice crossed the blood-brain barrier, restoring FMRP expression in neurons and partially rescuing behavioral deficits like hyperactivity. In cardiomyopathy, CRISPR-based editing targets mutations in RBM20, an RBP that controls alternative splicing of cardiac genes; RBM20 variants cause dilated cardiomyopathy by disrupting titin isoform balance. Base editing of RBM20 mutations in human induced pluripotent stem cell-derived cardiomyocytes corrected splicing defects and improved contractility, paving the way for therapeutic applications. Despite these advances, translating RBP-targeted therapies faces significant hurdles, including off-target effects from ASOs and CRISPR tools that can alter unintended RNA transcripts or genomic sites, potentially causing toxicity. Delivery across the blood-brain barrier remains a barrier for CNS disorders like ALS and fragile X, necessitating specialized vectors or intrathecal routes, as seen with nusinersen. As of 2025, clinical pipelines continue to evolve, with ongoing Phase 2/3 trials for TDP-43 modulators in ALS (e.g., ibudilast) and preclinical AAV-FMR1 studies advancing toward IND-enabling stages, but long-term safety data and optimized delivery systems are critical for broader adoption.

References

  1. [1]
    A large-scale binding and functional map of human RNA ... - Nature
    Jul 29, 2020 · RBPs are a diverse class of proteins that are involved in regulating gene expression. They interact with RNA to form ribonucleoprotein complexes ...
  2. [2]
    How RNA binding proteins interact with RNA: molecules and ...
    SUMMARY. RNA binding proteins (RBPs) comprise a large class of over 2000 proteins that interact with transcripts in all manner of RNA-driven processes.
  3. [3]
    RNA-Binding Proteins Hold Key Roles in Function, Dysfunction, and ...
    RNA-binding proteins (RBPs) are multi-faceted proteins in the regulation of RNA or its RNA splicing, localisation, stability, and translation.
  4. [4]
  5. [5]
  6. [6]
  7. [7]
    The RNA recognition motif, a plastic RNA‐binding platform to ...
    Apr 21, 2005 · The RNP 1 and RNP 2 motifs are located in the central strands of the β-sheet, namely β3 and β1, respectively, and are highly conserved apart ...
  8. [8]
    U2AF homology motifs: protein recognition in the RRM world
    In most canonical RRM-RNA structures, conserved aromatic Phe/Tyr residues at the third RNP1 position or second RNP2 position stack with RNA bases or sugars, and ...
  9. [9]
    Dividing and Conquering the RNA Recognition Motif Family
    The RNA recognition motif (RRM) is a prevalent RNA-binding fold among proteins that are required for RNA biogenesis, processing, transport, and degradation.
  10. [10]
    Structure and function of KH domains - Valverde - 2008 - FEBS Press
    Apr 15, 2008 · The KH motif consists of approximately 70 amino acids, and is found in a diverse variety of proteins in archaea, bacteria and eukaryota [1,2].
  11. [11]
    The STAR RNA binding proteins GLD-1, QKI, SAM68 and SLM-2 ...
    May 20, 2009 · The feature of KH domains is an invariant GXXG loop located between α1/α2 that provides close contact with the phosphate groups such that the ...
  12. [12]
    Protein-RNA and Protein-Protein Recognition by Dual KH1/2 ...
    Nova harbors three K-homology (KH) motifs implicated in alternate splicing regulation of genes involved in inhibitory synaptic transmission. We report the ...
  13. [13]
    Structure of the KH1-KH2 Domains of Fragile X Mental Retardation ...
    We present an X-ray crystal structure of the tandem KH domains of human FMRP, which reveals the relative orientation of the KH1 and KH2 domains.
  14. [14]
    Regulation of mRNA stability by CCCH-type zinc-finger proteins in ...
    TTP directly binds to AREs in its target mRNAs to promote destabilization and decay by recruiting deadenylation and decapping complexes (10, 11, 25) (Fig. 2).
  15. [15]
    Interactions of CCCH Zinc Finger Proteins with mRNA
    Tristetraprolin (TTP), the prototype of a small family of CCCH tandem zinc finger (TZF) domain proteins, is a physiological stimulator of instability of the ...Missing: decay | Show results with:decay
  16. [16]
    The tandem CCCH zinc finger protein tristetraprolin and ... - PubMed
    The TTP pathway of TNF-alpha and GM-CSF mRNA degradation is a possible novel target for anti-TNF-alpha therapies for rheumatoid arthritis, and also for other ...Missing: type | Show results with:type<|separator|>
  17. [17]
    RNA recognition by double-stranded RNA binding domains
    The double-stranded RNA binding domain (dsRBD) is a small protein domain of 65–70 amino acids adopting an αβββα fold, whose central property is to bind to ...
  18. [18]
    Structure of the double‐stranded RNA‐binding domain of the protein ...
    The structure of dsRBD exhibits a dumb‐bell shape comprising two tandem linked dsRNA‐binding motifs (dsRBMs) both with an α‐β‐β‐β‐α fold. The structure, ...
  19. [19]
    Modular Recognition of RNA by a Human Pumilio-Homology Domain
    Introduction. Members of the Puf family of proteins regulate expression of mRNA expression by binding the 3′ untranslated regions (UTRs) of their mRNA targets.Results And Discussion · Rna Binding By Puf Family... · Rna Binding By Drosophila...
  20. [20]
    Understanding and engineering RNA sequence specificity of PUF ...
    PUF proteins are RNA-binding proteins named for founding members PUMILIO and FBF ... They regulate mRNA expression by binding to specific sequences in the 3′ UTR ...
  21. [21]
    “Boundary residues” between the folded RNA recognition motif and ...
    The binding affinity of wildtype FUS is 0.7 μM, whereas the two folded domains, RRM (>90 μM) and ZnF (>175 μM), show very weak affinity individually. Among the ...
  22. [22]
    Intrinsically disordered RGG/RG domains mediate degenerate ...
    May 27, 2017 · We report a detailed analysis of the RNA binding characteristics of intrinsically disordered RGG/RG domains from Fused in Sarcoma (FUS), FMRP and hnRNPU.
  23. [23]
    Deciphering the RRM-RNA recognition code - PubMed Central - NIH
    Jan 23, 2023 · RNA recognition motifs (RRM) are the most prevalent class of RNA binding domains in eucaryotes. Their RNA binding preferences have been ...
  24. [24]
    RNA recognition motifs of disease-linked RNA-binding proteins ...
    Apr 16, 2019 · RNA-binding proteins usually contain one or more RNA recognition motifs (RRMs), raising the intriguing question as to whether RRMs can mediate ...
  25. [25]
    RNA-binding proteins: modular design for efficient function - PMC
    Jul 12, 2017 · Many RNA-binding proteins have modular structures, being composed of multiple repeats of just a few basic domains that are arranged in a variety of ways.
  26. [26]
    Hfq and its constellation of RNA - PMC - PubMed Central
    Hfq is an RNA-binding protein that is common to diverse bacterial lineages and has key roles in the control of gene expression.
  27. [27]
    Blending and separating dynamics of RNA-binding proteins develop ...
    Aug 8, 2024 · Core and accessory RBPs in RNA splicing make two distinct meshworks adjacently but distinctly distributed throughout the nucleus.
  28. [28]
    The Muscleblind family of proteins: an emerging class of regulators ...
    In this review, we focus on the Muscleblind family of tissue-specific alternative splicing regulators. Muscleblind proteins bind pre-mRNA through an ...
  29. [29]
    Muscleblind-like proteins use modular domains to localize RNAs by ...
    Jun 9, 2023 · Muscleblind-like (MBNL) proteins, implicated in myotonic dystrophy and cancer, localize RNAs to myoblast membranes and neurites through unknown mechanisms.
  30. [30]
    The mutual interaction of glycolytic enzymes and RNA in post ...
    This review summarizes current knowledge about the RNA-binding activity of the ten core enzymes of glycolysis in plant, yeast, and animal cells.
  31. [31]
    The unconventional RNA-binding activity of metabolic enzymes
    Sep 3, 2025 · In this Review, we discuss the evidences that these enzymes, with a particular focus on enzymes participating in the glucose metabolism, can ...
  32. [32]
    Evolution of protein-RNA interactions - ScienceDirect.com
    Ancient RNA-binding themes​​ The “RNA world” hypothesis traditionally casts peptides as cofactors in an RNA-centric origin of life [12,13].
  33. [33]
    Structure of Pfu Pop5, an archaeal RNase P protein - PNAS
    The protein adopts an α–β sandwich fold highly homologous to the single-stranded RNA binding RRM domain. Furthermore, the three-dimensional arrangement of Pfu ...Sign Up For Pnas Alerts · Results · Discussion
  34. [34]
    Ribonuclease P: The Evolution of an Ancient RNA Enzyme - PMC
    The RNP domain is also known as RNA binding domain (RBD) or the RNA recognition motif (RRM). The protein from B. subtilis binds both the full length RNA and the ...
  35. [35]
    Regulation of Splicing Factors by Alternative Splicing and NMD Is ...
    The SR proteins comprise a family of highly similar proteins that expanded through a series of duplications (Busch and Hertel 2011; Richardson et al. 2011).<|separator|>
  36. [36]
    Evolution of SR protein and hnRNP splicing regulatory factors - Busch
    Sep 2, 2011 · The splicing of pre-mRNAs is an essential step of gene expression in eukaryotes. Introns are removed from split genes through the activities ...
  37. [37]
    RNA interference in protozoan parasites and its application
    We also discuss the evolutionary loss of the RNAi pathway in protozoan parasites such that RNA viruses may play a selective/driving force for the loss of RNAi.Rna Interference In... · 1. Introduction · 2. Discovery Rnai Pathways...
  38. [38]
    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...
  39. [39]
    The exon–exon junction complex provides a binding platform for ...
    Therefore, REF, TAP and p15 most likely dissociate from the EJC soon after mRNA export. Some EJC components do remain associated with spliced mRNA after export.Missing: NXF1 NXT1 TREX seminal
  40. [40]
    TREX exposes the RNA binding domain of Nxf1 to enable mRNA ...
    Our data indicate that TREX provides a license for mRNA export by driving Nxf1 into a conformation capable of binding mRNA.Missing: NXT1 exon junction EJC seminal
  41. [41]
    The principal mRNA nuclear export factor NXF1:NXT1 forms a ... - NIH
    Jan 27, 2015 · Abstract. The NXF1:NXT1 complex (also known as TAP:p15) is a general mRNA nuclear export factor that is conserved from yeast to humans.Missing: exon junction EJC seminal
  42. [42]
    Structure of the human core transcription-export complex reveals a ...
    Nov 16, 2020 · TREX selectively binds mRNA maturation marks and licenses mRNA for nuclear export by loading the export factor NXF1–NXT1. How TREX integrates ...Missing: seminal | Show results with:seminal
  43. [43]
    Zipcode Binding Protein 1 (ZBP1; IGF2BP1) - PubMed Central - NIH
    Jul 28, 2020 · In neurons, ZBP1 is important for localization of β-actin mRNA in dendrites. The localization of β-actin mRNA occurs in response to ...Missing: paper | Show results with:paper
  44. [44]
    Activity-Dependent Trafficking and Dynamic Localization of Zipcode ...
    Apr 15, 2003 · We have focused on the mRNA binding protein, zipcode binding protein 1 (ZBP1), which binds a 54 nucleotide (nt) localization sequence termed “ ...
  45. [45]
    Neurotrophin-Induced Transport of a β-Actin mRNP Complex ...
    Formation of an RNP complex between the β-actin mRNA zipcode sequence and Zipcode Binding Protein 1 (ZBP1) was required for its localization to growth cones.Missing: paper | Show results with:paper
  46. [46]
    Limited availability of ZBP1 restricts axonal mRNA localization and ...
    The 3′UTR of β-actin mRNA contains a conserved 'zipcode' that is essential for its localization in cultured cells through interaction with the zipcode binding ...Missing: paper | Show results with:paper
  47. [47]
    RNA stabilization by the AU-rich element binding protein, HuR ... - NIH
    Here we demonstrate a role for the embryonic lethal abnormal visual (ELAV) RNA-binding protein HuR in mRNA turnover in vivo.
  48. [48]
    RNA stabilization by the AU‐rich element binding protein, HuR, an ...
    Here we demonstrate a role for the embryonic lethal abnormal visual (ELAV) RNA‐binding protein HuR in mRNA turnover in vivo.
  49. [49]
    The roles of TTP and BRF proteins in regulated mRNA decay
    Dec 16, 2010 · The tandem CCCH zinc finger protein tristetraprolin and its relevance to cytokine mRNA turnover and arthritis. Arthritis Res Ther 2004, 6 ...
  50. [50]
    Nonsense-mediated mRNA decay in humans at a glance
    Feb 1, 2016 · Nonsense-mediated mRNA decay (NMD) is an mRNA quality-control mechanism that typifies all eukaryotes examined to date.<|control11|><|separator|>
  51. [51]
    The mammalian nonsense‐mediated mRNA decay pathway: To ...
    Jan 20, 2009 · The UPF proteins have been considered the conserved core of the NMD machinery, despite the organism or the mechanism by which PTCs are defined ...
  52. [52]
    Nonsense-Mediated mRNA Decay: Mechanisms and Recent ...
    Aug 19, 2025 · Additionally, an NMD mechanism involving only UPF1 and UPF2, bypassing UPF3 and other canonical components, has been reported in Trypanosoma ...
  53. [53]
  54. [54]
    binding protein is a eukaryotic translation initiation factor, which acts ...
    PABP can thus be considered a canonical translation initiation factor, integral to initiation complex formation at the 5′-end of mRNA.
  55. [55]
    Iron regulates the activity of the iron-responsive element binding ...
    The activity of the IRE-BP is itself regulated by iron such that when cells are treated with an iron source, the RNA binding activity is decreased.
  56. [56]
    Human Argonaute2 Mediates RNA Cleavage Targeted by miRNAs ...
    The effector complex targeting single-stranded complementary RNAs for degradation is known as the RNA-induced silencing complex (RISC) (Hammond et al., 2000).Article · Results · Experimental ProceduresMissing: paper | Show results with:paper
  57. [57]
    Cell stress modulates the function of splicing regulatory protein ...
    At present, how ITAFs modulate IRES-mediated translation remains at the periphery of our knowledge. Some ITAFs, such as La and PTB, may act as RNA ...
  58. [58]
    Characterization and functional implications of the RNA binding ...
    Sep 28, 2001 · Interestingly, TDP-43 can bind a minimum number of six UG (or TG) single-stranded dinucleotide stretches, and binding affinity increases with ...
  59. [59]
    Molecular basis of UG-rich RNA recognition by the human splicing ...
    Nov 17, 2013 · Our findings reveal not only how TDP-43 recognizes UG repeats but also how RNA binding-dependent inter-RRM interactions are crucial for TDP-43 function.
  60. [60]
    Recognition of viral RNA stem-loops by the tandem double-stranded ...
    Jan 17, 2013 · PKR consists of tandem dsRNA binding motifs (dsRBMs) connected via a flexible linker to a Ser/Thr kinase domain. Upon interaction with viral ...
  61. [61]
    in vitro activation by p34cdc2 and phosphorylation of a 58-kD CPE ...
    We propose that p34cdc2 kinase activity leads to the phosphorylation of the 58-kD CPE-binding protein and that this event is crucial for the cytoplasmic ...
  62. [62]
    Kinetic studies of RNA-protein interactions using surface plasmon ...
    This technology is a powerful tool with which to study the dynamics of RNA-protein interactions. We have used BIACORE extensively to obtain detailed insight ...
  63. [63]
    G3BP1 Is a Tunable Switch that Triggers Phase ... - PubMed - NIH
    Apr 16, 2020 · G3BP1, which functions as a molecular switch that triggers RNA-dependent LLPS in response to a rise in intracellular free RNA concentrations.
  64. [64]
    Recognition of the 3′ splice site RNA by the U2AF heterodimer ...
    The U2AF heterodimer promotes spliceosome assembly by a dynamic population shift toward the open conformation of U2AF65 to facilitate the recognition of weak ...
  65. [65]
    Effects of pH and salt concentration on the siRNA binding activity of ...
    We observe that the CIRV p19 protein binds to siRNA with nanomolar affinity and that this binding is sensitive to pH and salt concentration.
  66. [66]
    Role of pH in Modulating RNA-Protein Interactions in TRBP2-dsRBD2
    Dec 26, 2024 · Exposure to stress conditions, such as hypoxia, leads to a drop in intracellular pH, which, in turn, alters the ionization states of amino acid ...
  67. [67]
    Impact of crowded environments on binding between protein and ...
    Sep 3, 2021 · This suggests that the effects on the binding affinity found for adding PEG1 or PEG8 to the test tube are not primarily excluded volume effects.
  68. [68]
    Competition between RNA-binding proteins CELF1 and HuR ...
    Mar 25, 2015 · We show that CELF1 competes with the RBP HuR to modulate MYC translation and regulates intestinal epithelial homeostasis. Growth inhibition of ...Results · Myc 3′-Utr Is A Direct... · Celf1 Inhibits Myc...
  69. [69]
    Single molecule probing of disordered RNA binding proteins
    Feb 3, 2022 · Single-molecule FRET experiments report on RNA conformation changes during binding; Oligomerization of disordered proteins is determined by ...
  70. [70]
    Recruitment of Nanos to hunchback mRNA by Pumilio
    Taken together, these findings suggest models for how Nos and Pum collaboratively target hb mRNA. More generally, they suggest that Pum-like proteins from other ...
  71. [71]
    Divergent RNA‐binding Proteins, DAZL and VASA, Induce Meiotic ...
    Dec 12, 2011 · These results demonstrate that evolutionarily unrelated and divergent RNA-binding proteins can promote meiotic progression of human-derived germ ...
  72. [72]
    Dynamic RNA–protein interactions underlie the zebrafish maternal ...
    Using mRNA interactome capture, we identified 227 RBPs in zebrafish embryos before and during ZGA, hereby named the zebrafish MZT mRNA-bound proteome. This ...Missing: anterior- posterior
  73. [73]
    Recruitment of mRNAs to P granules by condensation with ... - NIH
    We find that P granules recruit mRNAs by condensation with the disordered protein MEG-3. MEG-3 traps mRNAs into non-dynamic condensates in vitro and binds to ~ ...
  74. [74]
    Muscleblind proteins regulate alternative splicing - PMC - NIH
    In this paper, we show that MBNL proteins function as potent target-specific regulators of pre-mRNA alternative splicing. This finding has particular relevance ...
  75. [75]
    An Overview of Alternative Splicing Defects Implicated in Myotonic ...
    Muscleblind-like proteins (MBNL) are a family of tissue-specific regulators of developmentally programmed alternative splicing. They regulate alternative ...
  76. [76]
    RNA binding proteins in the regulation of heart development - PMC
    RNA binding protein, fox-1 homolog (RBFOX) proteins are RRM-containing proteins (Figure 3) that specifically bind to (U)GCAUG elements and regulate alternative ...
  77. [77]
    RNA splicing regulated by RBFOX1 is essential for cardiac function ...
    Aug 15, 2015 · The rbfox1 gene encodes a 373-amino-acid protein containing an RNA-binding domain (RBD) motif that is highly conserved among RNA-binding ...<|separator|>
  78. [78]
    NOVA-dependent regulation of cryptic NMD exons controls synaptic ...
    Jan 22, 2013 · The neuronal RNA binding protein NOVA regulates splicing, shuttles to the cytoplasm, and co-localizes with target transcripts in dendrites.
  79. [79]
    Developmentally Regulated Switch in Alternatively Spliced SNAP ...
    Oct 6, 2004 · The t-SNARE SNAP-25 is regulated during brain growth and synaptogenesis at the level of expression and by alternative splicing between tandem ...Missing: RBP | Show results with:RBP
  80. [80]
    FMRP regulates mRNAs encoding distinct functions in the cell body ...
    Dec 23, 2021 · FMRP, the RNA-binding protein whose activity is lost in Fragile X syndrome, represses translation (Bassell and Warren, 2008; Costa-Mattioli et ...
  81. [81]
    Staufen 2 regulates mGluR long-term depression and Map1b mRNA ...
    Neurons use mRNA transport during development to regu- late axon determination (Morita and Sobue 2009), growth cone turning (Lin and Holt 2007), neurite ...
  82. [82]
    Molecular mechanisms behind mRNA localization in axons - Journals
    Sep 23, 2020 · mRNA localization is involved in regulating neuronal development and function by orchestrating rapid adaptive responses to extracellular cues.
  83. [83]
    Mammalian ELAV-like neuronal RNA-binding proteins HuB ... - PNAS
    Hu proteins are mammalian embryonic lethal abnormal visual system (ELAV)-like neuronal RNA-binding proteins that contain three RNA recognition motifs.Mammalian Elav-Like Neuronal... · Results · Roles Of Hu Proteins In...
  84. [84]
    The RNA-binding protein HuD regulates neuronal cell identity and ...
    Neural Hu proteins (HuB/C/D) are RNA-binding proteins that have been shown to induce neuronal differentiation activity when overexpressed in immature neural ...<|separator|>
  85. [85]
    Mutation of the ALS-/FTD-Associated RNA-Binding Protein FUS ...
    We found that mutant FUS reduced actin density in the growth cone, altering its mechanical properties. Therefore, FUS mutants may induce defects during early ...
  86. [86]
    RNA-binding proteins in ALS and FTD: from pathogenic ... - PubMed
    Jun 4, 2025 · A large body of evidence highlights the critical role of RNA-binding proteins (RBPs) - in particular TAR DNA-binding protein 43 (TDP-43) and ...
  87. [87]
    Molecular Mechanisms of Phase Separation and Amyloidosis of ...
    Oct 1, 2024 · FUS and TDP-43, two RNA-binding proteins from the heterogeneous nuclear ribonucleoprotein family, have gained significant attention in the ...
  88. [88]
    ALS-causative mutations in FUS/TLS confer gain and loss ... - Nature
    Jan 27, 2015 · These results indicate that there are probably extensive RNA processing defects in ALS patients with FUS/TLS mutations, both arising from ...
  89. [89]
    Mutant FUS causes DNA ligation defects to inhibit oxidative damage ...
    Sep 11, 2018 · We discover that loss of nuclear FUS caused DNA nick ligation defects in motor neurons due to reduced recruitment of XRCC1/LigIII to DNA strand breaks.
  90. [90]
    Ro60-Roles in RNA Processing, Inflammation, and Rheumatic ...
    Jul 14, 2024 · Ro60 autoantibodies serve as diagnostic markers for various autoimmune diseases, including Sjögren's disease (SjD) and systemic lupus erythematosus (SLE).Missing: syndrome immune
  91. [91]
    RBP–RNA interactions in the control of autoimmunity and ... - Nature
    Jan 5, 2023 · In this review, we summarize and discuss the functions of RBP–RNA interactions in controlling aberrant autoimmune inflammation and their potential as ...
  92. [92]
    The spliceosomal autoantigen heterogeneous nuclear ...
    Taken together, the pronounced peripheral T cell reactivity to hnRNP-A2 observed in the majority of SLE patients and the distinct phenotype of patient-derived ...
  93. [93]
    B cell epitopes of the heterogeneous nuclear ribonucleoprotein A2
    The spliceosomal autoantigen heterogeneous nuclear ribonucleoprotein A2 (hnRNP-A2) is a major T cell autoantigen in patients with systemic lupus erythematosus.
  94. [94]
    Double-stranded RNA-dependent protein kinase (PKR) in antiviral ...
    The interferon-inducible double-stranded RNA-dependent protein kinase (PKR) is one of the key antiviral arms of the innate immune system.
  95. [95]
    dsRNA-induced condensation of antiviral proteins modulates PKR ...
    One key response to dsRNA is the activation of PKR, an eIF2α kinase, which triggers translational arrest and the formation of stress granules. However, the ...
  96. [96]
    Nsp1 protein of SARS-CoV-2 disrupts the mRNA export machinery ...
    Feb 5, 2021 · We show that the virulence factor Nsp1 protein of SARS-CoV-2 interacts with the host messenger RNA (mRNA) export receptor heterodimer NXF1-NXT1.
  97. [97]
    SARS-CoV-2 Nsp1 binds the ribosomal mRNA channel to inhibit ...
    Sep 9, 2020 · Structural basis for translational shutdown and immune evasion by the Nsp1 protein of SARS-CoV-2. Science https://doi.org/10.1126/science ...
  98. [98]
    Alternative Splicing Regulator RBM20 and Cardiomyopathy - PubMed
    Nov 28, 2018 · RBM20 is a regulator of heart-specific alternative splicing and Rbm20 ΔRRM mice lacking the RRM domain are defective in the splicing regulation.
  99. [99]
    Gain-of-function cardiomyopathic mutations in RBM20 rewire ...
    Nov 3, 2021 · Mutations in the cardiac splicing factor RBM20 lead to malignant dilated cardiomyopathy (DCM). To understand the mechanism of RBM20-associated DCM, we ...
  100. [100]
    Insights into RNA biology from an atlas of mammalian ... - PubMed
    Jun 8, 2012 · Interactome capture is broadly applicable to study mRNA interactome composition and dynamics in varied biological settings. Copyright © 2012 ...
  101. [101]
    A census of human RNA-binding proteins - PubMed - NIH
    A census of human RNA-binding proteins. Nat Rev Genet. 2014 Dec;15(12):829-45. doi: 10.1038/nrg3813. Epub 2014 Nov 4. Authors. Stefanie Gerstberger , Markus ...
  102. [102]
    Robust single-cell discovery of RNA targets of RNA binding proteins ...
    While RIP-seq is useful for identifying gene targets of an RBP, CLIP-seq can resolve binding sites within different regions of a given target gene, which lends ...
  103. [103]
    Highly accurate protein structure prediction with AlphaFold - Nature
    Jul 15, 2021 · The AlphaFold network directly predicts the 3D coordinates of all heavy atoms for a given protein using the primary amino acid sequence and ...
  104. [104]
    Protein–RNA interaction prediction with deep learning: structure ...
    In this work, we give a thorough review of this field, surveying both the binding site and binding preference prediction problems and covering the commonly used ...
  105. [105]
    Nusinersen: First Global Approval - PubMed
    Nusinersen is approved in the USA for intrathecal use in paediatric and adult patients with SMA. Regulatory assessments for nusinersen as a treatment for SMA ...
  106. [106]
    U.S. FDA Approves Biogen's SPINRAZA™ (nusinersen), The First ...
    Dec 23, 2016 · SPINRAZA improved motor function in SMA patients; greater percentage of infantile-onset patients on SPINRAZA.
  107. [107]
    Emerging Therapies and Novel Targets for TDP-43 Proteinopathy in ...
    In this review, we will discuss the TDP-43 therapy development pipeline, beginning with therapies in current and upcoming clinical trials.
  108. [108]
  109. [109]
  110. [110]
    Heterogeneous nuclear ribonucleoprotein A1 is identified as a ...
    We proposed that hnRNP A1 could be considered as a novel serum tumor marker for CRC that may have significance in the detection and in the management of ...Missing: plasma | Show results with:plasma
  111. [111]
    Extracellular microRNAs in human circulation are associated with ...
    Our findings suggest that miRNAs in circulation are largely contained in functional miRISC/AGO2 complexes under normal physiological conditions.
  112. [112]
    Efficient Delivery of FMR1 across the Blood Brain Barrier ... - PubMed
    Feb 16, 2023 · Conclusion: These experiments demonstrate efficient, brain-specific delivery of Fmr1 via peripheral administration in adult mice. The gene ...
  113. [113]
    Efficient Delivery of FMR1 across the Blood Brain Barrier Using ...
    Feb 16, 2023 · The gene delivery led to partial alleviation of the Fmr1 ... FMR1 KO Mice Suggests the Feasibility of Gene Therapy for Fragile X Syndrome.
  114. [114]
    Precise genomic editing of pathogenic mutations in RBM20 rescues ...
    Nov 23, 2022 · Autosomal dominant mutations in the RNA binding motif protein 20 (RBM20) are a common cause of cardiomyopathy, accounting for 2 to 6% of ...
  115. [115]
    RNA-based therapeutics: an overview and prospectus - Nature
    Jul 23, 2022 · In this review, the mechanisms of action, challenges, solutions, and clinical application of RNA-based therapeutics have been comprehensively summarized.
  116. [116]
    Peptide-Functionalized Lipid Nanoparticles for Targeted Systemic ...
    Systemic delivery of large nucleic acids, such as mRNA, to the brain remains challenging in part due to the blood-brain barrier (BBB) and the tendency of ...<|control11|><|separator|>
  117. [117]
    Gene therapy using human FMRP isoforms driven by ... - Cell Press
    Oct 6, 2022 · Gene therapy using human FMRP isoforms driven by the human FMR1 promoter rescues fragile X syndrome mouse deficits.