Skip to main content
RNA Biology logoLink to RNA Biology
. 2020 Jan 28;17(4):417–424. doi: 10.1080/15476286.2020.1717059

Molecular entrapment by RNA: an emerging tool for disrupting protein–RNA interactions in vivo

Tarjani N Shukla 1, Jane Song 1, Zachary T Campbell 1,
PMCID: PMC7237136  PMID: 31957541

ABSTRACT

mRNA function is controlled by RNA-binding proteins. The specificity of RNA-binding factors for their targets is critical in that it enables all subsequent regulation. Despite widespread recognition of the pervasive role RNA-binding proteins play in development and disease, they remain challenging to target with small molecules. A renaissance in RNA therapeutics has led to the identification of modifications that substantially increase RNA stability. When combined with information regarding specificity, a new class of oligonucleotide mimics has emerged as a means to competitively disrupt the regulation of endogenous substrates. These decoys have been used to inhibit RNA-binding proteins in living animals. Decoys will likely provide new insights into the expansive roles of RNA-binding proteins in biology and disease. Here, we describe examples where they have been used and discuss how they could be applied to new targets.

KEYWORDS: RNA decoys, RNA protein interactions, Specificity, RNA-binding proteins

Introduction

RNA-binding proteins (RBPs) dictate virtually every aspect of mRNA function. The life of an mRNA begins with transcription in the nucleus followed by processing, export, localization, storage, translation, and ultimately decay. Each of these steps hinges on recognition of regulatory features situated in a given transcript by RBPs [1]. The interaction between cis-acting elements and their protein partners is fundamental to RNA control. Three parallel advances enable new tools for probing RBP function in vivo. These are systematic identification of RNA-binding proteins, tools to profile specificity, and chemical modifications that increase RNA stability in vivo. The use of rationally designed competitive inhibitors may enable major advances in our ability to achieve the coveted goal of understanding biological functions of liaisons between RBPs and their targets in vivo [2,3].

Genome-wide approaches to identify RNA-binding proteins in human cells have revealed over a thousand RBPs [4,5]. Several themes have emerged from unbiased assessments of the mRNA associated proteome [6]. First, many interactions between RBPs and RNA occur without the use of canonical domains (e.g. RRM, KH, etc.) [7]. Unstructured regions appear to serve prominent roles in RNA-recognition by proteins. Second, sites of RNA-binding are subject to post-translational modifications. This suggests that RNA binding can be fine-tuned within the cell. Finally, RBPs are deeply conserved between basal eukaryotes and metazoan [7]. This implies that RBPs contribute to organismal fitness and are maintained after millions of years of evolutionary divergence as a result. Beyond general principles elucidated from analyses of the growing compendium of RBPs, an equally challenging problem is to understand the specificity of these factors for targeted RNAs.

Numerous methods have emerged to characterize the specificity of RNA–protein interactions. Several enable the identification of RNA targets in cells (e.g. HITS-CLIP, iCLIP, eCLIP, sCLIP, irCLIP, PAPERCLIP, PAR-CLIP, CRAC, etc.) [817]. These approaches are tremendously powerful and yield transcriptome-wide views into RNA-binding preferences. Yet, extracting quantitative information is challenging as is discriminating functional sites from non-productive interactions. Additional complexity arises from combinatorial control of an RNA by protein complexes [18]. Protein partners can induce substantial changes inspecificity [1922]. Thus, in vitro methods (e.g. SEQRS, RNA-bind and seq, RNA-compete, TGA, etc.) to assess the specificity of individual components and defined protein complexes are an invaluable source of information [2327]. Despite tremendous advances in the area, specificity and targeting are generally insufficient to infer precise biological functions for the vast majority of RBPs.

Understanding the regulatory roles of RBPs requires strategies to perturb their interaction with targets – ideally in animal models that recapitulate key features of diseases (e.g. mice and rats). Genetic approaches remain the dominant approach and can be achieved in numerous ways. For example, conditional deletion enables cell-type specific knockout in adult animals [28]. This strategy is highly desirable for genes that are essential for development and thus cannot be examined in a whole-body knockout. Unfortunately, mouse models can require substantial investments of time and resources. Comparatively rapid approaches (e.g. antisense oligonucleotides, RNA interference, etc.) hinge on RNA-destabilization and/or translational repression to reduce protein abundance [29,30]. These methods are less robust than genetic deletion as they transiently diminish protein abundance. Nevertheless, several features of synthetic oligonucleotides for RBP depletion make the approach broadly appealing. First, the translational potential of ASOs is increasingly apparent given that more than a half dozen are FDA approved [31]. Because of the proven safety and efficacy of therapeutic oligonucleotides, this area of pharmacology is ripe for repurposing. Second, substantial improvements in the persistence and targeting of oligos can be achieved through covalent modifications to nucleotides and phosphodiester linkages. Third, the ability to mimic consensus binding elements is revolutionary given our lack of understanding of the biological importance for most RBPs. Because of the diversity of mechanisms employed by RNA-binding proteins, decoys enable manipulation of a broad range of mechanistic functions of RBPs that require direct binding to RNA and their physiological properties. Key regulatory events that could be accessed with this approach include stability, translation, processing and phase separation (Fig. 1). To date, translation and processing have been targeted with this strategy in vivo [2,3]. Yet, the diverse roles that RBPs play in disease biology suggest that potential applications are widespread.

Figure 1.

Figure 1.

Four classes of RNA-binding protein (RBP) decoys and their potential implications.

(A) RNA decoys promote mRNA destabilization. In this example, the RBP protects the mRNA from 3ʹ end degradation through inhibition of deadenylase activity. In the presence of the decoy, the RBP is unable to bind to the mRNA transcript due to its preferred binding affinity to the decoy. The anticipated outcome is reduced mRNA stability, as deadeynlation is more efficient resulting in reduced stability. (B) RNA decoys promote translation repression. The RBP in this example promotes recruitment of the eIF4F complex. When the decoy is introduced, translation is no longer enhanced resulting in diminished rates of initiation. (C) RNA decoys that influence exon skipping. An RBP binds to an intronic splice enhancer that promotes the inclusion of an alternative exon (exon 2). In the presence of an mRNA decoy, the RBP is no longer bound to the mRNA and exon 2 is skipped. (D) RNA decoys that antagonize phase separation events. RBPs that undergo phase transitions within a cell exist in equilibrium between phase-separated granules and soluble RBP-mRNA complexes. When a decoy is introduced in the cell, the solubility of an RBP could be increased resulting in a reduction in the proportion of the RBP in a phase-separated granule.

RNA–protein interactions are prominent in disease states [6,32,33]. Pain is exemplary. Translational control of mRNA in the peripheral nervous system has emerged as a critical mechanism that governs the transition from acute to chronic pain [3437]. Phosphorylation of the mRNA cap-binding protein eIF4E is required for persistent pain in mice [38]. Similarly, multiple RBPs have been linked to pain (e.g. FMRP, CPEB, PABP, etc.) [2,3943]. In the central nervous system, RBPs play equally critical roles. Mutations in multiple RBPs (e.g. TARDBP, FUS, hnRNAp A1, A2, B1, etc.) have been implicated in neurodegeneration [44]. Thus, neurons rely heavily on RNA control to accomplish a range of biological functions spanning plasticity and viability. General translation factors also play prominent roles in the growth of cancer cells [45,46]. Compromised cap-dependent translation impairs tumorigenesis in vivo [47,48]. Many sequence-specific RNA-binding proteins appear to contribute to oncogenesis. One hundred and thirty-nine RBPs are consistently mutated in cancer and 76 may contain driver mutations [49]. It is unclear how many are essential in vivo. Yet, for the vast majority of RBPs implicated in cancer, their biological functions are not fully understood. As the technology for studying RBPs in living animals improves, our appreciation of RBPs in human health is likely to expand substantially. Stabilized RNA decoys are one such tool that could be applied to study RBP function in pre-clinical models.

Rise of the decoys in vivo

The initial proof of concept for the use of RBP decoys suitable for use in vivo targeted the Poly(A)-binding protein (PABP) [2]. To test the notion that PABP is specific for poly(A), PABP was first subjected to an unbiased selection and high-throughput sequencing analysis. Based on these data, a compact 12-base RNA termed a specificity-derived competitive inhibitor oligonucleotide or SPOT-ON was devised. A variety of modifications can increase RNA stability and have differing effects on the immune response. To enhance the stability of the SPOT-ON, 2ʹO-methyl linkages were introduced as well as terminal 5ʹ and 3ʹ phosphorothioates. The SPOT-ON RNA displayed a half-life on the order of 10 days as compared to 18 h for an unmodified poly(A) sequence. Importantly, the modifications did not significantly impair binding to the target. Introduction of the SPOT-ON to cells resulted in attenuation of nascent translation specifically at the initiation phase. In neurons, the SPOT-ON reduced translation both in the soma and at sites of local translation in axons. To demonstrate efficacy in vivo, the SPOT-ON was examined using standard mouse models of pain sensitization. In these animals, a specific behavioural response called paw withdrawal, is scored after application of a von Frey filament. Under baseline conditions, animals have a withdrawal threshold of ~1g of force. Injection of inflammatory mediators or noxious chemicals cause nociceptor activity to increase and promote sensitivity to mechanical stimulation. A decrease in withdrawal threshold to ~0.3g is interpreted as an enhanced pain response. In this model, pain sensitization can be blocked with a wide array of pharmacological agents that reduce cap-dependent or poly(A)-dependent translation[50]. Similarly, injection of modest amounts of the poly(A) SPOT-ON ca. 1µg blocks sensitization[2]. Given the complexity of the system, a logical question becomes if the underlying mechanism is neuronally mediated. To address this point, a second series of experiments was conducted with a stimulus that is highly specific for neurons, capsaicin. The SPOT-ON blocked sensitization following injection of capsaicin at a high dose (10 µg). This suggests that the relevant site of action of the compound is in the axons that innervate the paw. It is unclear how the oligo is taken up by neurons and should be noted that carriers were not used in this study (e.g. polymers).

A second example of RNA-decoys targeted RNA-processing factors implicated in cancer – specifically RBFOX, SRSF1, and PTBP1[3]. The oligos, termed splicing factor decoys, were designed based on consensus sequences identified from the literature and confirmed by pulldowns and splicing assays. As with SPOT-ONs, 2ʹOmethyl substitutions were added to all of the ribose groups. In all but one experiment, the backbone was unmodified. To improve the potency of the oligos, tandem copies of binding elements were introduced ranging in number from 1 to 4. Intriguingly, increasing the number of binding sites reduced equilibrium dissociation constants implying that cooperative effects promote synergistic binding to the decoys. Injection of the RBFOX oligo (8 pg) with a fully modified phosphorothioate backbone into zebrafish caused severe developmental defects of musculature in vivo. In a subsequent series of experiments, PTBP1 and SRSF1 decoys impaired growth of breast and glioblastoma cancer cells in vitro, respectively [3,51]. Intriguingly, glioblastoma tumours seeded into the brain were noticeably smaller 3 weeks after injection of a single dose of an SRSF1 decoy. These data suggest that RBP decoys targeted to RNA processing may provide a promising new type of therapeutic for cancer. As the oligonucleotides were injected without carriers, it is similarly unclear how they were taken up in vivo.

Decoys in vitro

A third example implies that RBP decoys could be used to attenuate viral transcription. Efficient transcription of HIV-1 and replication can be reduced by mimics of the TAR RNA[52]. The TAR RNA element promotes transcription by RNA-pol II through increasing phosphorylation of the C-terminal domain (CTD) via a mechanism that requires cyclin T1 and P-TEFb CTD kinase[53]. To generate a stable decoy of the TAR RNA, a circular RNA was generated[52]. The circular RNA was shown to persist for more than 12 h in HeLa nuclear extracts[52]. The RNA decoy blocks HIV-transcription in vitro and has yet to be demonstrated in vivo. Yet, the approach provides a valuable demonstration for how to decoy oligos can be used to modulate transcription. The use of circular RNAs provides an intriguing alternative to stabilize RNA-decoys. Circular RNAs are also less immunogenic than other types of RNA (e.g. mRNA) and can be tolerated at higher dosages in vivo[54].

A fourth example employs the use of RBP decoys as a means to prevent TDP-43 cytoplasmic inclusions[55]. TDP-43 is implicated in amyotrophic lateral sclerosis (ALS) and is characterized by aberrant phase transitions that lead to neurotoxic effects [5557]. A modified oligonucleotide decoy of the one of the TDP-43 RNA-recognition motifs was made to determine its effects on optogenetically induced TDP-43 phase transitions[55]. HEK293 cells were treated with the TDP-43 decoy 4 h before optogenetic induction of TDP-43 assemblies. The bait oligonucleotide reduced TDP-43 dependent phase transition in a dose-dependent manner and thus caused a decrease in neurotoxicity[55]. The TDP-43 decoy prevents exaggerated phase transition and neurotoxicity in vitro. Although the effect of the decoy has not been characterized in vivo, these data indicate that RBP decoys can also be used to modify aberrant phase transitions and suppress accumulation of RBPs to prevent cytotoxicity.

Limitations

Despite the efficacy of RBP decoys in a limited number of in vivo studies, there are several areas where they can be substantially improved. The specificity of the decoy oligo for the target RBP is crucial. There are at least four general strategies that could be employed to characterize the specificity of existing decoys and potentially improve targeting. First, numerous modifications to ASOs improve their targeting to mRNA (e.g. LNAs)[58]. It is unclear how similar modifications would impact binding to proteins (and is likely idiosyncratic depending on the RBP) but nevertheless these could improve specificity. Second, many RBPs bind to short compact sequences [59,60]. As a result, a decoy could be recognized by multiple factors. Small molecule inhibitors provide a useful parallel, perhaps modified RNA bases could be used to design RBP decoys that are rejected by some proteins due to steric clashes but that retain binding to their preferred target. The disadvantage of this approach is the need for structural information and the requirement for sophisticated medicinal chemistry. Third, validation of RBP targeting would ideally be conducted in an unbiased way. For example, CLIP with modified oligos followed by mass spectrometry would provide a means to identify off-target RBPs that bind to a decoy. Fourth and finally, validation of on-target specificity is critical. Ideally, the combined use of genetic models lacking an RBP of interest can be used to confirm the biological consequences of a given decoy. A priori, the oligo should be inert in a system devoid of the targeted factor.

Precise delivery of oligonucleotides to target cells is a coveted goal that extends to decoy oligos. Conjugation of ASOs to cell-penetrating peptides has been demonstrated to increase delivery of oligos relative to naked oligos[61]. Similar additions could improve cellular uptake of decoys. A related problem is targeting of oligos to specific cell types. Conjugation of small molecules to the RNA could be used to impart cellular specificity. For example, the addition of folate to miRNAs substantially improves targeting to breast cancer cells and tumours[62]. Finally, not all tissue types are equally accessible via injection. Tremendous progress in polymer and lipid-based carriers has been shown to aid in the delivery of antisense oligonucleotides[63]. Their use for decoys could enable delivery to difficult tissues to access such as the lung[64].

Opportunities

The advent of decoy oligonucleotides that compete with endogenous RNA substrates enables tremendous tools for discerning the biological functions of RBPs. Given the widespread involvement of RNA-binding proteins in disease, specificity is well established for many potential targets (Table 1). We describe potential targets that have been validated in vivo in three biological contexts, neurodegeneration, cancer, and pain. While the general approach should be applicable to many disease states, these models are particularly well suited given that multiple RBPs are integral to each process.

Table 1.

Potential RNA-binding proteins as targets for decoys implicated in disease.

Potential target RBP Biological function Disorder
RBPs in neurodegeneration and developmental disorders
Poly(A) binding protein nuclear 1
(PABPN1)
PABPN1 binds to the poly(A) tail of the mRNA, promoted stability and export[65,67]. Oculopharyngeal muscular dystrophy
(OPMD)
Fused in sarcoma RNA-binding protein
(FUS)
FUS facilitates regulates alternative splicing of mRNA and RNA export from the nucleus[68,69]. Amyotrophic lateral sclerosis (ALS)
Matrin 3 (MATR3) MATR3 is involved in RNA decay and splicing[7780]. Amyotrophic lateral sclerosis (ALS)
RhoA-specific guanine nucleotide exchange factor
(p190RhoGEF)
p190RhoGEF binds to the light neurofilament (NF-L) mRNA[81,82]. Motor neuron degeneration
DEAD-box helicase 3 (DDX3X) DDX3X is a member of the DEAD-box helicase family and binds to the Asp-Glu-Ala-Asp (DEAD) sequence to stimulate ATP-dependent RNA helicase activity[83]. Fragile X Syndrome (FXS)
RBPs in cancer
Epithelial splicing regulatory protein 1
(ESRP1)
ESRP1 is involved in mRNA splicing of specific epithelial cell proteins by binding to GU-rich motifs on the mRNA[85]. Colorectal cancer
HuR or ELAV-like RNA-binding protein 1 (Elavl1) HuR binds AU-rich motifs on the mRNA, regulates mRNA stability and decay[86]. Meningioma
Heterogeneous nucear ribonucleoproteins (hnRNPA1 and hnRNPM) hNRNP proteins are a class of nuclear RNA-binding proteins that are known to be involved in stability, alternative splicing, transcription, and translation[88]. Colon and gastric cancers
KH-type splicing regulatory protein (KHSRP) KHSRP is an RNA-binding protein implicated in splicing[92]. Lung cancer
Musashi RNA-binding proteins (MSIs) MSI1 controls mRNA on a post-transcriptional basis[93]. Osteosarcoma
LIN28 LIN28 binds promotes translation of targets. Also implicated in miRNA control[95,104]. Tumorigenesis of many cancer
RBPs in pain
HuD or ELAV-like RNA-binding protein 4 (Elavl4) HuD is neuron specific, binds cis-acting AU-rich elements to coordinate mRNA stability[100]. Anti-retroviral induced neuropathy
Eukaryotic translation initiation factor 4E (eIF4E) eIF4E is a key component of the eIF4F complex to promote translation after binding to the 5ʹ cap on the target mRNA[105]. Chronic pain
Cytoplasmic polyadenylation element binding protein (CPEB) CPEB facilitates polyadenylation and elongation of the poly(A) tail on the mRNA[106]. Chronic pain

RBP targets in neurodegeneration and developmental dissorders

Genetic lesions in the PABPN1 gene that cause protein misfolding are linked to the onset of oculopharyngeal muscular dystrophy (OPMD)[65]. PABPN1 is one of 6-PABP proteins but is restricted to the nucleus[66]. In this compartment, it regulates the length of the Poly(A) tail and promotes export and stability. Knockdown of PABN1 with viral vectors in murine models reduces muscle fibrosis and restores muscle strength in mice with OPMD[67]. Decoys could be used to target PABPN1 as a means to resolve muscular dystrophy onset without the use of virus-based therapies.

Gain-of-function mutations in the RNA-binding protein FUS cause amyotrophic lateral sclerosis (ALS) [68,69]. FUS plays a role in regulating RNA polymerase II and has been implicated in regulating alternative splicing [7073]. FUS is primarily located in the nucleus, but C-terminus mutations can induce phase separation of FUS resulting in cytoplasmic inclusions [7476]. These, in turn, disrupt RNA metabolism. Decoys that bind to FUS could increase FUS solubility and decrease its propensity for aggregation. The prior example of decoys for TDP-43 establishes a valuable proof of concept for this approach[55].

Similarly, RNA- and DNA-binding protein Matrin 3 (MATR3) has been implicated in ALS[77]. MATR3 is involved in the regulation of alternative splicing and regulation of mRNA nuclear export [7880]. Deletion mutants of an RRM promote aggregation of MATR3 in the nucleus[77]. MATR3 is neurotoxic when RNA-binding activity is removed [77,78]. Given that pathogenic mutations in MATR3 reduce its solubility, one way to modulate MATR3 function would be through the use of RNA decoys.

We propose a similar mechanism of action for an RNA decoy against p190RhoGEF, a protein involved in motor neuron degeneration. p190RhoGEF binds the NF-L mRNA and plays a role in NF-L protein aggregation[81]. NF-L aggregation promotes neuron degeneration [81,82]. siRNA knockdown of p190RhoGEF causes reversal of NF-L protein aggregation in this context[82]. RNA decoys tailored to p190RhoGEF could prevent its association with the NF-L mRNA and might attenuate motor neuron degeneration.

DDX3X is a DEAD-box helicase that has recently been implicated as a modifier of RAN (non AUG) translation, specifically in the context of Fragile X syndrome (FXS) [83,84]. Knockdown of DDX3X in vivo and in cell lines reduces FMR1-associated RAN translation and decreased neurotoxicity in those models[83]. Decoys against DDX3X could prevent stimulation of FMR1-associated RAN translation and resolve core FXS behavioural deficits.

RBP targets in cancer

ESRP1 promotes colorectal cancer[85]. It regulates splicing in epithelial cells[85]. ESRP1 overexpression promotes AKT activation and upregulation of fibroblast growth factor receptor[85]. The use of RNA decoys could clarify the role of ESRP1 as a tumour promoting RBP.

HuR has been identified as a potential target in human meningiomas[86]. HuR regulates many aspects of mRNA function including splicing and stability [86,87]. Overexpression of HuR has been linked to increased tumour expression and poor prognosis[86]. siRNA knockdown of HuR in vivo has been shown to reduce tumour progression[87]. Decoys targeted to HuR have the potential to attenuate meningioma growth.

The hnRNP family of RNA-binding proteins function as key regulators in mRNA stability[88]. Overexpression and aberrant activity of hnRNPA1 and hnRNPM has been linked to gastric and colon cancers, respectively, [89,90]. Specific reduction of these proteins via siRNA and viral vector knockdown indicate significant decreases in tumour formation and cancer progression [89,90]. Decoys could reduce aberrant hnRNPA1 and hnRNPM activity and cancer progression.

KHSRP has been identified as a key regulatory protein involved in non-small cell lung cancer[91]. KHSRP functions as a regulatory protein in the context of alternative splicing, cell proliferation, and cell differentiation[92]. Viral knockdown of KHSRP in vivo results in a decrease in cancer cell proliferation and migration[91]. The use of RNA decoys against KHSRP in this context might reduce the proliferation of lung cancer cells.

MSI1 is a key player in the formation and progression of many cancers[93]. This family of proteins function redundantly and MSI1 plays a functional role in post-transcriptional regulation [93,94]. In the context of osteosarcoma, knockdown of MSI1 in vivo via shRNA has been shown to reduce cancer cell proliferation[94]. A decoy against MSI1 has the potential to suppress aberrant cell proliferation.

LIN28 has been implicated in tumour formation and cancer cell proliferation [95,96]. LIN28 has been characterized as an oncogene and is overexpressed in most cancers [95,97]. It regulates the let-7 miRNA [95,97,98]. Knockdown of LIN28 with siRNAs in murine models decreases tumour growth and formation significantly[97]. A LIN28 decoy can sponge the RBP away from the let-7 miRNA to allow for functional let-7 activity and resolve aberrant cancer cell proliferation.

RBP targets in pain

Protein translation and pain are intimately linked [34,37]. This relationship coupled with evidence that RBPs are highly expressed in neurons, makes RBPs a compelling target for decoys[36]. HuD is an RNA-binding protein that is involved in regulating mRNA stability and expression in neurons[99]. HuD has emerged as a possible driver of nucleoside reverse transcriptase inhibitor (NRTI) induced neuropathy [100,101]. Protein kinase C (PKC) inhibitors prevent overexpression of HuD after NRTI treatment and block neuropathy in vivo[100]. RNA decoys against HuD in the context of NRTI-induced neuropathy are desirable because of the broad targeting of PKC.

Similarly, CPEB regulates cytoplasmic polyadenylation and translation[102]. It is implicated in chronic pain and promotes the persistence of a ‘pain memory’[42]. Activation of CPEB in neurons coincides with the generation of a pain memory[42]. In murine models, ASOs injected intrathecally against CPEB prevent hyperalgesia and formation of a pain memory, but do not reverse priming[42]. Decoys against CPEB have the potential to serve as a therapeutic to prevent chronic pain. Small molecule inhibitors of translation are potentially useful as analgesics [38,103]. RNA decoys specifically directed against eIF4E and possibly other components of the general translation apparatus could prevent pain-associated behaviours. These decoys have the potential to reduce eIF4E and CPEB expression and resolve the downstream consequences of their overexpression in the context of chronic pain.

Future directions

We have described examples where decoys have been used in vivo and highlighted potential opportunities to improve the design, specificity formulation, delivery, and validation. Given the increasing acceptance of ASOs in the clinic, repurposing stabilized oligos directed against RBPs provides a new pharmacological paradigm with incredible translational potential. The targets we describe represent a very small subset of RBPs. As our understanding of their specificity grows, decoys could be used to leverage specificity as a means to perturb biological function in an in vivo context. This approach should be broadly applicable to a range of pre-clinical models.

Funding Statement

This work was supported by NIH R01NS100788 to ZTC.

Disclosure statement

No potential conflict of interest was reported by the authors.

References

  • [1].Glisovic T, Bachorik JL, Yong J, et al. RNA-binding proteins and post-transcriptional gene regulation. FEBS Lett. 2008;582:1977–1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Barragán-Iglesias P, Lou T-F, Bhat VD, et al. Inhibition of Poly(A)-binding protein with a synthetic RNA mimic reduces pain sensitization in mice. Nat Commun. 2018;9:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Denichenko P, Mogilevsky M, Cléry A, et al. Specific inhibition of splicing factor activity by decoy RNA oligonucleotides. Nat Commun. 2019;10:1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Castello A, Fischer B, Eichelbaum K, et al. Insights into RNA biology from an atlas of mammalian mRNA-binding proteins. Cell. 2012;149:1393–1406. [DOI] [PubMed] [Google Scholar]
  • [5].Baltz AG, Munschauer M, Schwanhäusser B, et al. The mRNA-bound proteome and its global occupancy profile on protein-coding transcripts. Mol Cell. 2012;46:674–690. [DOI] [PubMed] [Google Scholar]
  • [6].Castello A, Fischer B, Hentze MW, et al. RNA-binding proteins in Mendelian disease. Trends Genet. 2013;29:318–327. [DOI] [PubMed] [Google Scholar]
  • [7].Beckmann BM, Horos R, Fischer B, et al. The RNA-binding proteomes from yeast to man harbour conserved enigmRBPs. Nat Commun. 2015;6:10127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Licatalosi DD, Mele A, Fak JJ, et al. HITS-CLIP yields genome-wide insights into brain alternative RNA processing. Nature. 2008;456:464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Konig J, Zarnack K, Rot G, et al. iCLIP reveals the function of hnRNP particles in splicing at individual nucleotide resolution. Nat Struct Mol Biol. 2010;17:909–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Van Nostrand EL, Pratt GA, Shishkin AA, et al. Robust transcriptome-wide discovery of RNA-binding protein binding sites with enhanced CLIP (eCLIP). Nat Methods. 2016;13:508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Kargapolova Y, Levin M, Lackner K, et al. sCLIP—an integrated platform to study RNA–protein interactomes in biomedical research: identification of CSTF2tau in alternative processing of small nuclear RNAs. Nucleic Acids Res. 2017;45:6074–6086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Zarnegar BJ, Flynn RA, Shen Y, et al. irCLIP platform for efficient characterization of protein–RNA interactions. Nat Methods. 2016;13:489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Hwang HW, Park C, Goodarzi H, et al. PAPERCLIP identifies microRNA targets and a role of CstF64/64tau in promoting non-canonical poly(A) site usage. Cell Rep. 2016;15:423–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Hafner M, Landthaler M, Burger L, et al. PAR-CliP–a method to identify transcriptome-wide the binding sites of RNA binding proteins. J Vis Exp. 2010. DOI: 10.3791/2034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Kishore S, Jaskiewicz L, Burger L, et al. A quantitative analysis of CLIP methods for identifying binding sites of RNA-binding proteins. Nat Methods. 2011;8:559–564. [DOI] [PubMed] [Google Scholar]
  • [16].Philippe N, Salson M, Commes T, et al. CRAC: an integrated approach to the analysis of RNA-seq reads. Genome Biol. 2013;14:R30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Lin C, Miles WO, Beyond CLIP.. advances and opportunities to measure RBP–RNA and RNA–RNA interactions. Nucleic Acids Res. 2019;47:5490–5501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Pique M, Lopez JM, Foissac S, et al. A combinatorial code for CPE-mediated translational control. Cell. 2008;132:434–448. [DOI] [PubMed] [Google Scholar]
  • [19].Campbell ZT, Bhimsaria D, Valley C, et al. Cooperativity in RNA-protein interactions: global analysis of RNA binding specificity. Cell Rep. 2012;1:570–581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Menichelli E, Wu J, Campbell ZT, et al. Biochemical characterization of the Caenorhabditis elegans FBF.CPB-1 translational regulation complex identifies conserved protein interaction hotspots. J Mol Biol. 2013;425:725–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Campbell ZT, Menichelli E, Friend K, et al. Identification of a conserved interface between PUF and CPEB proteins. J Biol Chem. 2012;287:18854–18862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Weidmann CA, Qiu C, Arvola RM, et al. Drosophila Nanos acts as a molecular clamp that modulates the RNA-binding and repression activities of Pumilio. Elife. 2016;5:e17096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Lou TF, Weidmann CA, Killingsworth J, et al. Integrated analysis of RNA-binding protein complexes using in vitro selection and high-throughput sequencing and sequence specificity landscapes (SEQRS). Methods. 2017;118–119:171–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Lambert N, Robertson A, Jangi M, et al. RNA Bind-n-Seq: quantitative assessment of the sequence and structural binding specificity of RNA binding proteins. Mol Cell. 2014;54:887–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Ray D, Kazan H, Cook KB, et al. A compendium of RNA-binding motifs for decoding gene regulation. Nature. 2013;499:172–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Zhou Q, Kunder N, De la Paz JA, et al. Global pairwise RNA interaction landscapes reveal core features of protein recognition. Nat Commun. 2018;9:2511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Campbell ZT, Wickens M.. Probing RNA–protein networks: biochemistry meets genomics. Trends Biochem Sci. 2015;40:157–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Bouabe H, Okkenhaug K. Gene targeting in mice: a review. Methods in molecular biology. ;2013;1064:315–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Kole R, Krainer AR, Altman S. RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov. 2012;11:125–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Dias N, Stein CA. Antisense oligonucleotides: basic concepts and mechanisms. Mol Cancer Ther. 2002;1:347–355. [PubMed] [Google Scholar]
  • [31].Stein CA, Castanotto D. FDA-approved oligonucleotide therapies in 2017. Mol Ther. 2017;25:1069–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Cooper TA, Wan L, Dreyfuss G. RNA and disease. Cell. 2009;136:777–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Lukong KE, Chang K, Khandjian EW, et al. RNA-binding proteins in human genetic disease. Trends Genet. 2008;24:416–425. [DOI] [PubMed] [Google Scholar]
  • [34].de la Peña JBI, Song JJ, Campbell ZT. RNA control in pain: blame it on the messenger. Wiley Interdiscip Rev RNA. 2019;10:e1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Khoutorsky A, Price TJ. Translational control mechanisms in persistent pain. Trends Neurosci. 2018;41:100–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].de la Peña JB, Campbell ZT. RNA-binding proteins as targets for pain therapeutics. Neurobiol Pain. 2018;4:2–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Loerch S, de la Peña JB, Song J, et al. Translational controls in pain. In: The Oxford handbook of neuronal protein synthesis. Oxford University Press; 2019. DOI: 10.1093/oxfordhb/9780190686307.013.22. [DOI] [Google Scholar]
  • [38].Moy JK, Khoutorsky A, Asiedu MN, et al. The MNK–eIF4E signaling axis contributes to injury-induced nociceptive plasticity and the development of chronic pain. J Neurosci. 2017;37:7481–7499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Price TJ, Melemedjian OK. Fragile X Mental Retardation Protein (FMRP) and the spinal sensory system. In: Modeling fragile X syndrome. Berlin, Heidelberg: Springer; 2012. p. 41–59. DOI: 10.1007/978-3-642-21649-7_4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Price TJ, Rashid MH, Millecamps M, et al. Decreased nociceptive sensitization in mice lacking the Fragile X Mental retardation protein: role of mGluR1/5 and mTOR. J Neurosci. 2007;27:13958–13967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Ferrari LF, Bogen O, Levine JD. Role of nociceptor CaMKII in transition from acute to chronic pain (hyperalgesic priming) in male and female rats. J Neurosci. 2013;33:11002–11011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Bogen O, Alessandri-Haber N, Chu C, et al. Generation of a pain memory in the primary afferent nociceptor triggered by PKCϵ activation of CPEB. J Neurosci. 2012;32:2018–2026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Ferrari LF, Bogen O, Alessandri-Haber N, et al. Transient decrease in nociceptor GRK2 expression produces long-term enhancement in inflammatory pain. Neuroscience. 2012;222:392–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Conlon EG, Manley JL. RNA-binding proteins in neurodegeneration: mechanisms in aggregate. Genes Dev. 2017;31:1509–1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Hinnebusch AG, Ivanov IP, Sonenberg N. Translational control by 5ʹ-untranslated regions of eukaryotic mRNAs. Science. 2016;352:1413–1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Pelletier J, Graff J, Ruggero D, et al. Targeting the eIF4F translation initiation complex: a critical nexus for cancer development. Cancer Res. 2015;75:250–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Truitt ML, Conn C, Shi Z, et al. Differential requirements for eIF4E dose in normal development and cancer. Cell. 2015;162:59–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Konicek BW, Stephens JR, McNulty AM, et al. Therapeutic inhibition of MAP kinase interacting kinase blocks eukaryotic initiation factor 4E phosphorylation and suppresses outgrowth of experimental lung metastases. Cancer Res. 2011;71:1849–1857. [DOI] [PubMed] [Google Scholar]
  • [49].Wang Z-L, Li B, Luo Y-X, et al. Comprehensive genomic characterization of RNA-binding proteins across human cancers. Cell Rep. 2018;22:286–298. [DOI] [PubMed] [Google Scholar]
  • [50].Melemedjian OK, Asiedu MN, Tillu DV, et al. IL-6- and NGF-induced rapid control of protein synthesis and nociceptive plasticity via convergent signaling to the eIF4F complex. J. Neurosci. 2010;30:15113–15123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Fregoso OI, Das S, Akerman M, et al. Splicing-factor oncoprotein SRSF1 stabilizes p53 via RPL5 and induces cellular senescence. Mol Cell. 2013;50:56–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Bohjanen PR, Colvin RA, Puttaraju M, et al. A small circular TAR RNA decoy specifically inhibits Tat-activated HIV-1 transcription. Nucleic Acids Res. 1996;24:3733–3738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Garber ME, Wei P, Jones KA. HIV-1 Tat interacts with cyclin T1 to direct the P-TEFb CTD kinase complex to TAR RNA. Cold Spring Harb Symp Quant Biol. 1998;63:371–380. [DOI] [PubMed] [Google Scholar]
  • [54].Wesselhoeft RA, Kowalski PS, Parker-Hale FC, et al. RNA circularization diminishes immunogenicity and can extend translation duration In Vivo. Mol Cell. 2019;74:508–520.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Mann JR, Gleixner AM, Mauna JC, et al. RNA binding antagonizes neurotoxic phase transitions of TDP-43. Neuron. 2019;102:321–338.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Scotter EL, Chen H-J, Shaw CE. TDP-43 proteinopathy and ALS: insights into disease mechanisms and therapeutic targets. Neurotherapeutics. 2015;12:352–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].François-Moutal L, Felemban R, Scott DD, et al. Small molecule targeting TDP-43’s RNA recognition motifs reduces locomotor defects in a drosophila model of Amyotrophic Lateral Sclerosis (ALS). ACS Chem Biol. 2019;14:2006–2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Swayze EE, Siwkowski AM, Wancewicz EV, et al. Antisense oligonucleotides containing locked nucleic acid improve potency but cause significant hepatotoxicity in animals. Nucleic Acids Res. 2007;35:687–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Ray D, Kazan H, Chan ET, et al. Rapid and systematic analysis of the RNA recognition specificities of RNA-binding proteins. Nat Biotechnol. 2009;27:667–670. [DOI] [PubMed] [Google Scholar]
  • [60].Dominguez D, Freese P, Alexis MS, et al. Sequence, structure, and context preferences of human RNA binding proteins. Mol Cell. 2018;70:854–867.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].McClorey G, Banerjee S. Cell-penetrating peptides to enhance delivery of oligonucleotide-based therapeutics. Biomedicines. 2018;6:51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Orellana EA, Tenneti S, Rangasamy L, et al. FolamiRs: ligand-targeted, vehicle-free delivery of microRNAs for the treatment of cancer. Sci Transl Med. 2017;9:eaam9327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Juliano RL. The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 2016;44:6518–6548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Patel AK, Kaczmarek JC, Bose S, et al. Inhaled nanoformulated mRNA polyplexes for protein production in lung epithelium. Adv Mater. 2019;31:1805116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Abu-Baker A, Rouleau GA. Oculopharyngeal muscular dystrophy: recent advances in the understanding of the molecular pathogenic mechanisms and treatment strategies. Biochim Biophys Acta - Mol Basis Dis. 2007;1772:173–185. [DOI] [PubMed] [Google Scholar]
  • [66].Wigington CP, Williams KR, Meers MP, et al. Poly(A) RNA-binding proteins and polyadenosine RNA: new members and novel functions. Wiley Interdiscip Rev RNA. 2014;5:601–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Malerba A, Klein P, Bachtarzi H, et al. PABPN1 gene therapy for oculopharyngeal muscular dystrophy. Nat Commun. 2017;8:14848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Sharma A, Lyashchenko AK, Lu L, et al. ALS-associated mutant FUS induces selective motor neuron degeneration through toxic gain of function. Nat Commun. 2016;7:10465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].López-Erauskin J, Tadokoro T, Baughn MW, et al. ALS/FTD-linked mutation in FUS suppresses intra-axonal protein synthesis and drives disease without nuclear loss-of-function of FUS. Neuron. 2018;100:816–830.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Nakaya T, Alexiou P, Maragkakis M, et al. FUS regulates genes coding for RNA-binding proteins in neurons by binding to their highly conserved introns. RNA. 2013;19:498–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Tan AY, Riley TR, Coady T, et al. TLS/FUS (translocated in liposarcoma/fused in sarcoma) regulates target gene transcription via single-stranded DNA response elements. Proc Natl Acad Sci U S A. 2012;109:6030–6035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Tan AY, Manley JL. TLS inhibits RNA polymerase III transcription. Mol Cell Biol. 2010;30:186–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Lerga A, Hallier M, Delva L, et al. Identification of an RNA binding specificity for the potential splicing factor TLS. J Biol Chem. 2001;276:6807–6816. [DOI] [PubMed] [Google Scholar]
  • [74].Kovar H. Dr. Jekyll and Mr. Hyde: the two faces of the FUS/EWS/TAF15 protein family. Sarcoma. 2011;2011:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Dormann D, Rodde R, Edbauer D, et al. ALS-associated fused in sarcoma (FUS) mutations disrupt transportin-mediated nuclear import. Embo J. 2010;29:2841–2857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Kamelgarn M, Chen J, Kuang L, et al. ALS mutations of FUS suppress protein translation and disrupt the regulation of nonsense-mediated decay. Proc Natl Acad Sci U S A. 2018;115:E11904–E11913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Malik AM, Miguez RA, Li X, et al. Matrin 3-dependent neurotoxicity is modified by nucleic acid binding and nucleocytoplasmic localization. Elife. 2018;7:e35977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Boehringer A, Garcia-Mansfield K, Singh G, et al. ALS Associated Mutations in Matrin 3 Alter Protein-Protein Interactions and Impede mRNA Nuclear Export. Sci Rep. 2017;7:1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Coelho MB, Attig J, Bellora N, et al. Nuclear matrix protein Matrin3 regulates alternative splicing and forms overlapping regulatory networks with PTB. Embo J. 2015;34:653–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Uemura Y, Oshima T, Yamamoto M, et al. Matrin3 binds directly to intronic pyrimidine-rich sequences and controls alternative splicing. Genes Cells. 2017;22:785–798. [DOI] [PubMed] [Google Scholar]
  • [81].Lin H, Schlaepfer WW. Role of neurofilament aggregation in motor neuron disease. Ann Neurol. 2006;60:399–406. [DOI] [PubMed] [Google Scholar]
  • [82].Lin H, Zhai J, Schlaepfer WW. RNA-binding protein is involved in aggregation of light neurofilament protein and is implicated in the pathogenesis of motor neuron degeneration. Hum Mol Genet. 2005;14:3643–3659. [DOI] [PubMed] [Google Scholar]
  • [83].Linsalata AE, He F, Malik AM, et al. DDX3X and specific initiation factors modulate FMR1 repeat-associated non-AUG-initiated translation. EMBO Rep. 2019;20:e47498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Cheng W, Wang S, Zhang Z, et al. CRISPR-Cas9 screens identify the RNA helicase DDX3X as a repressor of C9ORF72 (GGGGCC)n repeat-associated non-AUG translation. Neuron. 2019;104:885–898.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Fagoonee S, Picco G, Orso F, et al. The RNA-binding protein ESRP1 promotes human colorectal cancer progression. Oncotarget. 2017;8:10007–10024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Gauchotte G, Hergalant S, Vigouroux C, et al. Cytoplasmic overexpression of RNA-binding protein HuR is a marker of poor prognosis in meningioma, and HuR knockdown decreases meningioma cell growth and resistance to hypoxia. J Pathol. 2017;242:421–434. [DOI] [PubMed] [Google Scholar]
  • [87].Muralidharan R, Babu A, Amreddy N, et al. Tumor-targeted nanoparticle delivery of HuR siRNA inhibits lung tumor growth In Vitro and In Vivo by disrupting the oncogenic activity of the RNA-binding protein HuR. Mol Cancer Ther. 2017;16:1470–1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Geuens T, Bouhy D, Timmerman V. The hnRNP family: insights into their role in health and disease. Hum Genet. 2016;135:851–867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Chen Y, Liu J, Wang W, et al. High expression of hnRNPA1 promotes cell invasion by inducing EMT in gastric cancer. Oncol Rep. 2018;39:1693–1701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Chen T-M, Lai M-C, Li Y-H, et al. hnRNPM induces translation switch under hypoxia to promote colon cancer development. EBioMedicine. 2019;41:299–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Yan M, Sun L, Li J, et al. RNA-binding protein KHSRP promotes tumor growth and metastasis in non-small cell lung cancer. J Exp Clin Cancer Res. 2019;38:478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Briata P, Bordo D, Puppo M, et al. Diverse roles of the nucleic acid-binding protein KHSRP in cell differentiation and disease. Wiley Interdiscip Rev RNA. 2016;7:227–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Li N, Yousefi M, Nakauka-Ddamba A, et al. The Msi family of RNA-binding proteins function redundantly as intestinal oncoproteins. Cell Rep. 2015;13:2440–2455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Niu J, Zhao X, Liu Q, et al. Knockdown of MSI1 inhibited the cell proliferation of human osteosarcoma cells by targeting p21 and p27. Oncol Lett. 2017;14:5271–5278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Balzeau J, Menezes MR, Cao S, et al. The LIN28/let-7 pathway in cancer. Front Genet. 2017;8:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].O’Day E, Le MTN, Imai S, et al. An RNA-binding protein, Lin28, recognizes and remodels G-quartets in the MicroRNAs (miRNAs) and mRNAs it regulates. J Biol Chem. 2015;290:17909–17922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Piskounova E, Polytarchou C, Thornton J, et al. Lin28A and Lin28B inhibit let-7 MicroRNA biogenesis by distinct mechanisms. Cell. 2011;147:1066–1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Hagan JP, Piskounova E, Gregory RI. Lin28 recruits the TUTase Zcchc11 to inhibit let-7 maturation in mouse embryonic stem cells. Nat Struct Mol Biol. 2009;16:1021–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Deschênes-Furry J, Perrone-Bizzozero N, Jasmin BJ. The RNA-binding protein HuD: A regulator of neuronal differentiation, maintenance and plasticity. BioEssays. 2006;28:822–833. [DOI] [PubMed] [Google Scholar]
  • [100].Sanna MD, Quattrone A, Mello T, et al. The RNA-binding protein HuD promotes spinal GAP43 overexpression in antiretroviral-induced neuropathy. Exp Neurol. 2014;261:343–353. [DOI] [PubMed] [Google Scholar]
  • [101].Sanna MD, Quattrone A, Ghelardini C, et al. PKC-mediated HuD-GAP43 pathway activation in a mouse model of antiretroviral painful neuropathy. Pharmacol Res. 2014;81:44–53. [DOI] [PubMed] [Google Scholar]
  • [102].Richter JD. CPEB: a life in translation. Trends Biochem Sci. 2007;32:279–285. [DOI] [PubMed] [Google Scholar]
  • [103].Ferrari LF, Bogen O, Chu C, et al. Peripheral administration of translation inhibitors reverses increased hyperalgesia in a model of chronic pain in the rat. J Pain. 2013;14:731–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Wilbert ML, Huelga S, Kapeli K, et al. LIN28 binds messenger RNAs at GGAGA motifs and regulates splicing factor abundance. Mol Cell. 2012;48:195–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Gingras A-C, Raught B, Sonenberg N. eIF4 initiation factors: effectors of mRNA recruitment to ribosomes and regulators of translation. Annu Rev Biochem. 1999;68:913–963. [DOI] [PubMed] [Google Scholar]
  • [106].Hodgman R, Tay J, Mendez R, et al. CPEB phosphorylation and cytoplasmic polyadenylation are catalyzed by the kinase IAK1/Eg2 in maturing mouse oocytes. Development. 2001;128:2815–2822. [DOI] [PubMed] [Google Scholar]

Articles from RNA Biology are provided here courtesy of Taylor & Francis

RESOURCES