Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 49(21): 12502-12516, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34850109

ABSTRACT

Circular RNAs (circRNAs) are noncoding RNAs that exist in all eukaryotes investigated and are derived from back-splicing of certain pre-mRNA exons. Here, we report the application of artificial circRNAs designed to act as antisense-RNAs. We systematically tested a series of antisense-circRNAs targeted to the SARS-CoV-2 genome RNA, in particular its structurally conserved 5'-untranslated region. Functional assays with both reporter transfections as well as with SARS-CoV-2 infections revealed that specific segments of the SARS-CoV-2 5'-untranslated region can be efficiently accessed by specific antisense-circRNAs, resulting in up to 90% reduction of virus proliferation in cell culture, and with a durability of at least 48 h. Presenting the antisense sequence within a circRNA clearly proved more efficient than in the corresponding linear configuration and is superior to modified antisense oligonucleotides. The activity of the antisense-circRNA is surprisingly robust towards point mutations in the target sequence. This strategy opens up novel applications for designer circRNAs and promising therapeutic strategies in molecular medicine.


Subject(s)
Genome, Viral/genetics , RNA, Antisense/genetics , RNA, Circular/genetics , RNA, Viral/genetics , SARS-CoV-2/genetics , Virus Replication/genetics , 5' Untranslated Regions/genetics , Animals , Antiviral Agents/metabolism , Base Sequence , COVID-19/prevention & control , COVID-19/virology , Cell Proliferation/genetics , Chlorocebus aethiops , Drug Design , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA-Seq/methods , SARS-CoV-2/physiology , Vero Cells
2.
Nucleic Acids Res ; 48(21): 12326-12335, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33231682

ABSTRACT

Circular RNAs (circRNAs) are a class of noncoding RNAs, generated from pre-mRNAs by circular splicing of exons and functionally largely uncharacterized. Here we report on the design, expression, and characterization of artificial circRNAs that act as protein sponges, specifically binding and functionally inactivating hnRNP (heterogeneous nuclear ribonucleoprotein) L. HnRNP L regulates alternative splicing, depending on short CA-rich RNA elements. We demonstrate that designer hnRNP L-sponge circRNAs with CA-repeat or CA-rich sequence clusters can efficiently and specifically modulate splicing-regulatory networks in mammalian cells, including alternative splicing patterns and the cellular distribution of a splicing factor. This new strategy can in principle be applied to any RNA-binding protein, opening up new therapeutic strategies in molecular medicine.


Subject(s)
Alternative Splicing , Exons , Genetic Engineering/methods , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , RNA Precursors/genetics , RNA, Circular/genetics , Base Pairing , Gene Silencing , HEK293 Cells , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein L/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Humans , Immunoprecipitation/methods , Introns , Nucleic Acid Conformation , Nucleotide Motifs , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , RNA Precursors/metabolism , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , RNA, Circular/biosynthesis , RNA, Circular/chemistry
3.
Nat Commun ; 10(1): 2266, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31118463

ABSTRACT

How multidomain RNA-binding proteins recognize their specific target sequences, based on a combinatorial code, represents a fundamental unsolved question and has not been studied systematically so far. Here we focus on a prototypical multidomain RNA-binding protein, IMP3 (also called IGF2BP3), which contains six RNA-binding domains (RBDs): four KH and two RRM domains. We establish an integrative systematic strategy, combining single-domain-resolved SELEX-seq, motif-spacing analyses, in vivo iCLIP, functional validation assays, and structural biology. This approach identifies the RNA-binding specificity and RNP topology of IMP3, involving all six RBDs and a cluster of up to five distinct and appropriately spaced CA-rich and GGC-core RNA elements, covering a >100 nucleotide-long target RNA region. Our generally applicable approach explains both specificity and flexibility of IMP3-RNA recognition, allows the prediction of IMP3 targets, and provides a paradigm for the function of multivalent interactions with multidomain RNA-binding proteins in gene regulation.


Subject(s)
Models, Molecular , RNA, Messenger/metabolism , RNA-Binding Motifs/physiology , RNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , High-Throughput Nucleotide Sequencing/methods , Protein Binding/physiology , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , SELEX Aptamer Technique , Sequence Analysis, DNA/methods
4.
J Extracell Vesicles ; 7(1): 1424473, 2018.
Article in English | MEDLINE | ID: mdl-29359036

ABSTRACT

Circular RNAs (circRNAs) are a novel class of noncoding RNAs present in all eukaryotic cells investigated so far and generated by a special mode of alternative splicing of pre-mRNAs. Thereby, single exons, or multiple adjacent and spliced exons, are released in a circular form. CircRNAs are cell-type specifically expressed, are unusually stable, and can be found in various body fluids such as blood and saliva. Here we analysed circRNAs and the corresponding linear splice isoforms from human platelets, where circRNAs are particularly abundant, compared with other hematopoietic cell types. In addition, we isolated extracellular vesicles from purified and in vitro activated human platelets, using density-gradient centrifugation, followed by RNA-seq analysis for circRNA detection. We could demonstrate that circRNAs are packaged and released within both types of vesicles (microvesicles and exosomes) derived from platelets. Interestingly, we observed a selective release of circRNAs into the vesicles, suggesting a specific sorting mechanism. In sum, circRNAs represent yet another class of extracellular RNAs that circulate in the body and may be involved in signalling pathways. Since platelets are essential for central physiological processes such as haemostasis, wound healing, inflammation and cancer metastasis, these findings should greatly extend the potential of circRNAs as prognostic and diagnostic biomarkers.

5.
Sci Rep ; 6: 31313, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27510448

ABSTRACT

Circular RNAs (circRNAs) constitute a new class of noncoding RNAs in higher eukaryotes generated from pre-mRNAs by alternative splicing. Here we investigated in mammalian cells the association of circRNAs with proteins. Using glycerol gradient centrifugation, we characterized in cell lysates circRNA-protein complexes (circRNPs) of distinct sizes. By polysome-gradient fractionation we found no evidence for efficient translation of a set of abundant circRNAs in HeLa cells. To identify circRNPs with a specific protein component, we focused on IMP3 (IGF2BP3, insulin-like growth factor 2 binding protein 3), a known tumor marker and RNA-binding protein. Combining RNA-seq analysis of IMP3-co-immunoprecipitated RNA and filtering for circular-junction reads identified a set of IMP3-associated circRNAs, which were validated and characterized. In sum, our data suggest that specific circRNP families exist defined by a common protein component. In addition, this provides a general approach to identify circRNPs with a given protein component.


Subject(s)
RNA-Binding Proteins/metabolism , RNA/metabolism , Sequence Analysis, RNA/methods , Computational Biology , HeLa Cells , Humans , Immunoprecipitation , Protein Binding , RNA/genetics , RNA, Circular
6.
Cell Rep ; 10(1): 103-11, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25543144

ABSTRACT

Circular RNAs (circRNAs), an abundant class of noncoding RNAs in higher eukaryotes, are generated from pre-mRNAs by circularization of adjacent exons. Using a set of 15 circRNAs, we demonstrated their cell-type-specific expression and circular versus linear processing in mammalian cells. Northern blot analysis combined with RNase H cleavage conclusively proved a circular configuration for two examples, LPAR1 and HIPK3. To address the circularization mechanism, we analyzed the sequence requirements using minigenes derived from natural circRNAs. Both canonical splice sites are required for circularization, although they vary in flexibility and potential use of cryptic sites. Surprisingly, we found that no specific circRNA exon sequence is necessary and that potential flanking intron structures can modulate circularization efficiency. In combination with splice inhibitor assays, our results argue that the canonical spliceosomal machinery functions in circRNA biogenesis, constituting an alternative splicing mode.


Subject(s)
Alternative Splicing/genetics , Exons/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , RNA Splice Sites/genetics , RNA, Untranslated/genetics , HEK293 Cells , Humans , Introns/genetics , Nucleic Acid Conformation , RNA Precursors/genetics , Spliceosomes/genetics
7.
Dev Cell ; 31(1): 87-99, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25313962

ABSTRACT

Cell-type-specific splicing generates numerous alternatively spliced transcripts playing important roles for organ development and homeostasis, but only a few tissue-specific splicing factors have been identified. We found that RBM24 governs a large number of muscle-specific splicing events that are critically involved in cardiac and skeletal muscle development and disease. Targeted inactivation of RBM24 in mice disrupted cardiac development and impaired sarcomerogenesis in striated muscles. In vitro splicing assays revealed that recombinant RBM24 is sufficient to promote muscle-specific exon inclusion in nuclear extracts of nonmuscle cells. Furthermore, we demonstrate that binding of RBM24 to an intronic splicing enhancer (ISE) is essential and sufficient to overcome repression of exon inclusion by an exonic splicing silencer (ESS) containing PTB and hnRNP A1/A2 binding sites. Introduction of ESS and ISE converted a constitutive exon into an RMB24-dependent alternative exon. We reason that RBM24 is a major regulator of alternative splicing in striated muscles.


Subject(s)
Alternative Splicing , RNA-Binding Proteins/metabolism , Sarcomeres/metabolism , Animals , Exons , HeLa Cells , Heart/embryology , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Introns , Mice , Myocardium/metabolism , Organ Specificity , RNA-Binding Proteins/genetics
8.
Nucleic Acids Res ; 42(10): 6603-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24748659

ABSTRACT

Trans-splicing in trypanosomes adds a 39-nucleotide mini-exon from the spliced leader (SL) RNA to the 5' end of each protein-coding sequence. On the other hand, cis-splicing of the few intron-containing genes requires the U1 small nuclear ribonucleoprotein (snRNP) particle. To search for potential new functions of the U1 snRNP in Trypanosoma brucei, we applied genome-wide individual-nucleotide resolution crosslinking-immunoprecipitation (iCLIP), focusing on the U1 snRNP-specific proteins U1C and U1-70K. Surprisingly, U1C and U1-70K interact not only with the U1, but also with U6 and SL RNAs. In addition, mapping of crosslinks to the cis-spliced PAP [poly(A) polymerase] pre-mRNA indicate an active role of these proteins in 5' splice site recognition. In sum, our results demonstrate that the iCLIP approach provides insight into stable and transient RNA-protein contacts within the spliceosomal network. We propose that the U1 snRNP may represent an evolutionary link between the cis- and trans-splicing machineries, playing a dual role in 5' splice site recognition on the trans-spliceosomal SL RNP as well as on pre-mRNA cis-introns.


Subject(s)
Protozoan Proteins/metabolism , RNA Splicing , Ribonucleoprotein, U1 Small Nuclear/metabolism , Spliceosomes/metabolism , Trans-Splicing , Trypanosoma brucei brucei/genetics , Cell Nucleus/chemistry , Genome, Protozoan , Protozoan Proteins/analysis , RNA Precursors/metabolism , RNA Splice Sites , RNA, Messenger/metabolism , RNA, Small Nuclear/metabolism , Ribonucleoprotein, U1 Small Nuclear/analysis , Trypanosoma brucei brucei/metabolism
9.
RNA Biol ; 11(2): 146-55, 2014.
Article in English | MEDLINE | ID: mdl-24526010

ABSTRACT

Heterogeneous nuclear ribonucleoprotein L (hnRNP L) is a multifunctional RNA-binding protein that is involved in many different processes, such as regulation of transcription, translation, and RNA stability. We have previously characterized hnRNP L as a global regulator of alternative splicing, binding to CA-repeat, and CA-rich RNA elements. Interestingly, hnRNP L can both activate and repress splicing of alternative exons, but the precise mechanism of hnRNP L-mediated splicing regulation remained unclear. To analyze activities of hnRNP L on a genome-wide level, we performed individual-nucleotide resolution crosslinking-immunoprecipitation in combination with deep-sequencing (iCLIP-Seq). Sequence analysis of the iCLIP crosslink sites showed significant enrichment of C/A motifs, which perfectly agrees with the in vitro binding consensus obtained earlier by a SELEX approach, indicating that in vivo hnRNP L binding targets are mainly determined by the RNA-binding activity of the protein. Genome-wide mapping of hnRNP L binding revealed that the protein preferably binds to introns and 3' UTR. Additionally, position-dependent splicing regulation by hnRNP L was demonstrated: The protein represses splicing when bound to intronic regions upstream of alternative exons, and in contrast, activates splicing when bound to the downstream intron. These findings shed light on the longstanding question of differential hnRNP L-mediated splicing regulation. Finally, regarding 3' UTR binding, hnRNP L binding preferentially overlaps with predicted microRNA target sites, indicating global competition between hnRNP L and microRNA binding. Translational regulation by hnRNP L was validated for a subset of predicted target 3'UTRs.


Subject(s)
3' Untranslated Regions , Alternative Splicing , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Introns , MicroRNAs/metabolism , RNA, Messenger/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Gene Regulatory Networks , Genome, Human , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , High-Throughput Nucleotide Sequencing , Humans , Immunoprecipitation
10.
PLoS Genet ; 9(10): e1003856, 2013.
Article in English | MEDLINE | ID: mdl-24146627

ABSTRACT

The U1 small nuclear ribonucleoprotein (snRNP)-specific U1C protein participates in 5' splice site recognition and regulation of pre-mRNA splicing. Based on an RNA-Seq analysis in HeLa cells after U1C knockdown, we found a conserved, intra-U1 snRNP cross-regulation that links U1C and U1-70K expression through alternative splicing and U1 snRNP assembly. To investigate the underlying regulatory mechanism, we combined mutational minigene analysis, in vivo splice-site blocking by antisense morpholinos, and in vitro binding experiments. Alternative splicing of U1-70K pre-mRNA creates the normal (exons 7-8) and a non-productive mRNA isoform, whose balance is determined by U1C protein levels. The non-productive isoform is generated through a U1C-dependent alternative 3' splice site, which requires an adjacent cluster of regulatory 5' splice sites and binding of intact U1 snRNPs. As a result of nonsense-mediated decay (NMD) of the non-productive isoform, U1-70K mRNA and protein levels are down-regulated, and U1C incorporation into the U1 snRNP is impaired. U1-70K/U1C-deficient particles are assembled, shifting the alternative splicing balance back towards productive U1-70K splicing, and restoring assembly of intact U1 snRNPs. Taken together, we established a novel feedback regulation that controls U1-70K/U1C homeostasis and ensures correct U1 snRNP assembly and function.


Subject(s)
Alternative Splicing/genetics , RNA Precursors/genetics , RNA Splicing/genetics , Ribonucleoprotein, U1 Small Nuclear/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , DNA Mutational Analysis , Embryo, Nonmammalian , Gene Knockdown Techniques , HeLa Cells , Humans , RNA Splice Sites/genetics , Spliceosomes/genetics , Zebrafish/genetics , Zebrafish/growth & development
11.
Nucleic Acids Res ; 40(12): 5666-78, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22402488

ABSTRACT

CD45 encodes a trans-membrane protein-tyrosine phosphatase expressed in diverse cells of the immune system. By combinatorial use of three variable exons 4-6, isoforms are generated that differ in their extracellular domain, thereby modulating phosphatase activity and immune response. Alternative splicing of these CD45 exons involves two heterogeneous ribonucleoproteins, hnRNP L and its cell-type specific paralog hnRNP L-like (LL). To address the complex combinatorial splicing of exons 4-6, we investigated hnRNP L/LL protein expression in human B-cells in relation to CD45 splicing patterns, applying RNA-Seq. In addition, mutational and RNA-binding analyses were carried out in HeLa cells. We conclude that hnRNP LL functions as the major CD45 splicing repressor, with two CA elements in exon 6 as its primary target. In exon 4, one element is targeted by both hnRNP L and LL. In contrast, exon 5 was never repressed on its own and only co-regulated with exons 4 and 6. Stable L/LL interaction requires CD45 RNA, specifically exons 4 and 6. We propose a novel model of combinatorial alternative splicing: HnRNP L and LL cooperate on the CD45 pre-mRNA, bridging exons 4 and 6 and looping out exon 5, thereby achieving full repression of the three variable exons.


Subject(s)
Alternative Splicing , Exons , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Leukocyte Common Antigens/genetics , B-Lymphocytes/metabolism , Cell Line , HeLa Cells , Humans , Mutation , Regulatory Sequences, Ribonucleic Acid
12.
EMBO J ; 30(10): 1965-76, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21468032

ABSTRACT

Precise 5' splice-site recognition is essential for both constitutive and regulated pre-mRNA splicing. The U1 small nuclear ribonucleoprotein particle (snRNP)-specific protein U1C is involved in this first step of spliceosome assembly and important for stabilizing early splicing complexes. We used an embryonically lethal U1C mutant zebrafish, hi1371, to investigate the potential genomewide role of U1C for splicing regulation. U1C mutant embryos contain overall stable, but U1C-deficient U1 snRNPs. Surprisingly, genomewide RNA-Seq analysis of mutant versus wild-type embryos revealed a large set of specific target genes that changed their alternative splicing patterns in the absence of U1C. Injection of ZfU1C cRNA into mutant embryos and in vivo splicing experiments in HeLa cells after siRNA-mediated U1C knockdown confirmed the U1C dependency and specificity, as well as the functional conservation of the effects observed. In addition, sequence motif analysis of the U1C-dependent 5' splice sites uncovered an association with downstream intronic U-rich elements. In sum, our findings provide evidence for a new role of a general snRNP protein, U1C, as a mediator of alternative splicing regulation.


Subject(s)
Alternative Splicing , Gene Expression Regulation , Ribonucleoproteins, Small Nuclear/metabolism , Zebrafish Proteins/metabolism , Zebrafish/physiology , Animals , Embryo, Nonmammalian/physiology , Genetic Complementation Test , HeLa Cells , Humans , Molecular Sequence Data , Mutation , RNA Precursors/chemistry , RNA Precursors/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Sequence Analysis, DNA , Zebrafish/genetics , Zebrafish Proteins/genetics
13.
RNA Biol ; 7(1): 56-64, 2010.
Article in English | MEDLINE | ID: mdl-19946215

ABSTRACT

Heterogeneous nuclear ribonucleoprotein (hnRNP) L can regulate alternative mRNA splicing in diverse ways, binding to exonic or intronic sites and acting as either an activator or repressor. To investigate the mechanistic basis of hnRNP L-regulated alternative splicing, we focus here on two specific cases of hnRNP L-dependent splice site recognition. First, in the case of TJP1 our microarray data had suggested that exon 20 inclusion is regulated by hnRNP L as a repressor. Here we demonstrate by mutational analysis that exon skipping is mediated by a short silencer sequence consisting of three hnRNP L high-score binding motifs located upstream of the 3' splice site of the regulated exon. UV crosslinking and immunoprecipitation experiments showed that hnRNP L binding interferes with 3' splice site recognition by U2AF65. Second, SLC2A2 contains a CA-repeat sequence close to the 5' splice site of the regulated exon 4. Using psoralen crosslinking, we demonstrate that hnRNP L represses splicing by preventing 5' splice site recognition of the U1 snRNP. In sum, our data provide new insights into the mechanisms of how hnRNP L-bound to intronic sites-regulates exon recognition.


Subject(s)
Alternative Splicing/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Mammals/genetics , RNA Splice Sites/genetics , Animals , Base Sequence , Exons/genetics , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , HeLa Cells , Humans , Introns/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Sequence Data , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , RNA Precursors/metabolism , Ribonucleoproteins/metabolism , Silencer Elements, Transcriptional/genetics , Splicing Factor U2AF , Zonula Occludens-1 Protein
14.
Mol Cell Biol ; 29(6): 1442-51, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19124611

ABSTRACT

We recently characterized human hnRNP L as a global regulator of alternative splicing, binding to CA-repeat and CA-rich elements. Here we report that hnRNP L autoregulates its own expression on the level of alternative splicing. Intron 6 of the human hnRNP L gene contains a short exon that, if used, introduces a premature termination codon, resulting in nonsense-mediated decay (NMD). This "poison exon" is preceded by a highly conserved CA-rich cluster extending over 800 nucleotides that binds hnRNP L and functions as an unusually extended, intronic enhancer, promoting inclusion of the poison exon. As a result, excess hnRNP L activates NMD of its own mRNA, thereby creating a negative autoregulatory feedback loop and contributing to homeostasis of hnRNP L levels. We present experimental evidence for this mechanism, based on NMD inactivation, hnRNP L binding assays, and hnRNP L-dependent alternative splicing of heterologous constructs. In addition, we demonstrate that hnRNP L cross-regulates inclusion of an analogous poison exon in the hnRNP L-like pre-mRNA, which explains the reciprocal expression of the two closely related hnRNP L proteins.


Subject(s)
Alternative Splicing , Heterogeneous-Nuclear Ribonucleoprotein L/biosynthesis , Enhancer Elements, Genetic , Exons , Gene Expression Regulation , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Humans , Introns , Nucleic Acid Conformation , RNA Precursors/biosynthesis , RNA Precursors/genetics
15.
RNA ; 14(2): 284-96, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18073345

ABSTRACT

Alternative mRNA splicing patterns are determined by the combinatorial control of regulator proteins and their target RNA sequences. We have recently characterized human hnRNP L as a global regulator of alternative splicing, binding to diverse C/A-rich elements. To systematically identify hnRNP L target genes on a genome-wide level, we have combined splice-sensitive microarray analysis and an RNAi-knockdown approach. As a result, we describe 11 target genes of hnRNP L that were validated by RT-PCR and that represent several new modes of hnRNP L-dependent splicing regulation, involving both activator and repressor functions: first, intron retention; second, inclusion or skipping of cassette-type exons; third, suppression of multiple exons; and fourth, alternative poly(A) site selection. In sum, this approach revealed a surprising diversity of splicing-regulatory processes as well as poly(A) site selection in which hnRNP L is involved.


Subject(s)
Alternative Splicing/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/physiology , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/metabolism , Amino Acid Motifs , Exons , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein L/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Humans , Introns , Polyadenylation , RNA Interference
16.
Proc Natl Acad Sci U S A ; 104(16): 6608-13, 2007 Apr 17.
Article in English | MEDLINE | ID: mdl-17416673

ABSTRACT

The spliceosome cycle consists of assembly, catalysis, and recycling phases. Recycling of postspliceosomal U4 and U6 small nuclear ribonucleoproteins (snRNPs) requires p110/SART3, a general splicing factor. In this article, we report that the zebrafish earl grey (egy) mutation maps in the p110 gene and results in a phenotype characterized by thymus hypoplasia, other organ-specific defects, and death by 7 to 8 days postfertilization. U4/U6 snRNPs were disrupted in egy mutant embryos, demonstrating the importance of p110 for U4/U6 snRNP recycling in vivo. Surprisingly, expression profiling of the egy mutant revealed an extensive network of coordinately up-regulated components of the spliceosome cycle, providing a mechanism compensating for the recycling defect. Together, our data demonstrate that a mutation in a general splicing factor can lead to distinct defects in organ development and cause disease.


Subject(s)
DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Spliceosomes/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish , Animals , Genes, Lethal , Mutagenesis , Organ Specificity/genetics , Phenotype , RNA Splicing Factors , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Thymus Gland/abnormalities , Zebrafish/genetics , Zebrafish/metabolism
18.
EMBO J ; 24(11): 1988-98, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15889141

ABSTRACT

We have recently identified an intronic polymorphic CA-repeat region in the human endothelial nitric oxide synthase (eNOS) gene as an important determinant of the splicing efficiency, requiring specific binding of hnRNP L. Here, we analyzed the position requirements of this CA-repeat element, which revealed its potential role in alternative splicing. In addition, we defined the RNA binding specificity of hnRNP L by SELEX: not only regular CA repeats are recognized with high affinity but also certain CA-rich clusters. Therefore, we have systematically searched the human genome databases for CA-repeat and CA-rich elements associated with alternative 5' splice sites (5'ss), followed by minigene transfection assays. Surprisingly, in several specific human genes that we tested, intronic CA RNA elements could function either as splicing enhancers or silencers, depending on their proximity to the alternative 5'ss. HnRNP L was detected specifically bound to these diverse CA elements. These data demonstrated that intronic CA sequences constitute novel and widespread regulatory elements of alternative splicing.


Subject(s)
Alternative Splicing/physiology , Dinucleotide Repeats , Introns/genetics , RNA Splice Sites , Alternative Splicing/genetics , Base Sequence , Cross-Linking Reagents , DNA, Recombinant/genetics , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Humans , Molecular Sequence Data , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , RNA/radiation effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...