Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Hum Mutat ; 37(3): 280-91, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26670336

ABSTRACT

The Hutchinson Gilford Progeria Syndrome (HGPS) is a rare genetic disease leading to accelerated aging. Three mutations of the LMNA gene leading to HGPS were identified. The more frequent ones, c.1824C>T and c.1822G>A, enhance the use of the intron 11 progerin 5'splice site (5'SS) instead of the LMNA 5'SS, leading to the production of the truncated dominant negative progerin. The less frequent c.1868C>G mutation creates a novel 5'SS (LAΔ35 5'SS), inducing the production of another truncated LMNA protein (LAΔ35). Our data show that the progerin 5'SS is used at low yield in the absence of HGPS mutation, whereas utilization of the LAΔ35 5'SS is dependent upon the presence of the c.1868C>G mutation. In the perspective to correct HGPS splicing defects, we investigated whether SR proteins can modify the relative yields of utilization of intron 11 5'SSs. By in cellulo and in vitro assays, we identified SRSF5 as a direct key regulator increasing the utilization of the LMNA 5'SS in the presence of the HGPS mutations. Enhanced SRSF5 expression in dermal fibroblasts of HGPS patients as well as PDGF-BB stimulation of these cells decreased the utilization of the progerin 5'SS, and improves nuclear morphology, opening new therapeutic perspectives for premature aging.


Subject(s)
Fibroblasts/metabolism , Lamin Type A/genetics , Progeria/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , HeLa Cells , Humans , Progeria/genetics , RNA-Binding Proteins/genetics
2.
EMBO J ; 31(1): 162-74, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22002536

ABSTRACT

SRSF2 (SC35) is a key player in the regulation of alternative splicing events and binds degenerated RNA sequences with similar affinity in nanomolar range. We have determined the solution structure of the SRSF2 RRM bound to the 5'-UCCAGU-3' and 5'-UGGAGU-3' RNA, both identified as SRSF2 binding sites in the HIV-1 tat exon 2. RNA recognition is achieved through a novel sandwich-like structure with both termini forming a positively charged cavity to accommodate the four central nucleotides. To bind both RNA sequences equally well, SRSF2 forms a nearly identical network of intermolecular interactions by simply flipping the bases of the two consecutive C or G nucleotides into either anti or syn conformation. We validate this unusual mode of RNA recognition functionally by in-vitro and in-vivo splicing assays and propose a 5'-SSNG-3' (S=C/G) high-affinity binding consensus sequence for SRSF2. In conclusion, in addition to describe for the first time the RNA recognition mode of SRSF2, we provide the precise consensus sequence to identify new putative binding sites for this splicing factor.


Subject(s)
Cytosine/chemistry , Guanine/chemistry , Ribonucleoproteins/chemistry , Amino Acid Sequence , Base Sequence , Binding Sites , Crystallography, X-Ray , Cytosine/metabolism , Exons , Guanine/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA Splicing , Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
3.
Traffic ; 13(2): 257-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22023725

ABSTRACT

The reorganization of nuclear structures is an important early feature of apoptosis and involves the activity of specific proteases and nucleases. Well-known is the condensation and fragmentation of chromatin; however, much less is understood about the mechanisms involved in the reorganization of structures from the interchromatin space, such as interchromatin granule clusters (IGCs). In this study, we show that the initial enlargement and rounding-up of IGCs correlate with a decrease in mRNA transcription and are caspase-independent, but involve protein phosphatases PP1/PP2A. Subsequently, multiple enlarged IGCs dissociate from chromatin and fuse into a single structure. The dissociation requires caspase activity and involves caspase-activated DNase (CAD). Apoptotic IMR-5 cells, lacking a proper processing of CAD, show multiple enlarged IGCs that remain linked with chromatin. Overexpression of CAD in IMR-5 cells results in the dissociation of IGCs from chromatin, but the fusion into a single structure remains disturbed. Nuclear matrix protein NuMA is reorganized in a caspase-dependent way around fused IGCs. In conclusion, we show here that the apoptotic rearrangement of IGCs, the nuclear matrix and chromatin are closely associated, occur in defined stages and depend on the activity of protein phosphatases, caspases and CAD.


Subject(s)
Antigens, Nuclear/metabolism , Apoptosis/physiology , Caspases/metabolism , Deoxyribonucleases/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Protein Phosphatase 2/metabolism , Ribonucleoproteins/metabolism , Spliceosomes/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Caspase Inhibitors , Cell Cycle Proteins , Cell Line, Tumor , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Deoxyribonucleases/genetics , Humans , Intranuclear Space/drug effects , Intranuclear Space/metabolism , Intranuclear Space/ultrastructure , Mice , Mice, Inbred BALB C , Models, Biological , Nuclear Proteins/metabolism , Nucleophosmin , Phosphorylation/drug effects , Poly-ADP-Ribose Binding Proteins , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Ribonucleoprotein, U1 Small Nuclear/metabolism , Serine-Arginine Splicing Factors , Staurosporine/pharmacology , Transfection , snRNP Core Proteins/metabolism
4.
PLoS Genet ; 7(12): e1002390, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22194695

ABSTRACT

Alternative splicing amplifies the information content of the genome, creating multiple mRNA isoforms from single genes. The evolutionarily conserved splicing activator Tra2ß (Sfrs10) is essential for mouse embryogenesis and implicated in spermatogenesis. Here we find that Tra2ß is up-regulated as the mitotic stem cell containing population of male germ cells differentiate into meiotic and post-meiotic cells. Using CLIP coupled to deep sequencing, we found that Tra2ß binds a high frequency of exons and identified specific G/A rich motifs as frequent targets. Significantly, for the first time we have analysed the splicing effect of Sfrs10 depletion in vivo by generating a conditional neuronal-specific Sfrs10 knock-out mouse (Sfrs10(fl/fl); Nestin-Cre(tg/+)). This mouse has defects in brain development and allowed correlation of genuine physiologically Tra2ß regulated exons. These belonged to a novel class which were longer than average size and importantly needed multiple cooperative Tra2ß binding sites for efficient splicing activation, thus explaining the observed splicing defects in the knockout mice. Regulated exons included a cassette exon which produces a meiotic isoform of the Nasp histone chaperone that helps monitor DNA double-strand breaks. We also found a previously uncharacterised poison exon identifying a new pathway of feedback control between vertebrate Tra2 proteins. Both Nasp-T and the Tra2a poison exon are evolutionarily conserved, suggesting they might control fundamental developmental processes. Tra2ß protein isoforms lacking the RRM were able to activate specific target exons indicating an additional functional role as a splicing co-activator. Significantly the N-terminal RS1 domain conserved between flies and humans was essential for the splicing activator function of Tra2ß. Versions of Tra2ß lacking this N-terminal RS1 domain potently repressed the same target exons activated by full-length Tra2ß protein.


Subject(s)
Embryonic Development/genetics , Exons/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Alternative Splicing/genetics , Animals , Autoantigens/genetics , Autoantigens/metabolism , Base Sequence , Binding Sites , Brain/abnormalities , Cell Cycle Proteins , Cell Differentiation , DNA Breaks, Double-Stranded , Evolution, Molecular , Germ Cells/cytology , Male , Meiosis/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Serine-Arginine Splicing Factors , Spermatogenesis/genetics
5.
Hum Mol Genet ; 20(23): 4540-55, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21875900

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder phenotypically characterized by many features of premature aging. Most cases of HGPS are due to a heterozygous silent mutation (c.1824C>T; p.Gly608Gly) that enhances the use of an internal 5' splice site (5'SS) in exon 11 of the LMNA pre-mRNA and leads to the production of a truncated protein (progerin) with a dominant negative effect. Here we show that HGPS mutation changes the accessibility of the 5'SS of LMNA exon 11 which is sequestered in a conserved RNA structure. Our results also reveal a regulatory role of a subset of serine-arginine (SR)-rich proteins, including serine-arginine rich splicing factor 1 (SRSF1) and SRSF6, on utilization of the 5'SS leading to lamin A or progerin production and a modulation of this regulation in the presence of the c.1824C>T mutation is shown directly on HGPS patient cells. Mutant mice carrying the equivalent mutation in the LMNA gene (c.1827C>T) also accumulate progerin and phenocopy the main cellular alterations and clinical defects of HGPS patients. RNAi-induced depletion of SRSF1 in the HGPS-like mouse embryonic fibroblasts (MEFs) allowed progerin reduction and dysmorphic nuclei phenotype correction, whereas SRSF6 depletion aggravated the HGPS-like MEF's phenotype. We demonstrate that changes in the splicing ratio between lamin A and progerin are key factors for lifespan since heterozygous mice harboring the mutation lived longer than homozygous littermates but less than the wild-type. Genetic and biochemical data together favor the view that physiological progerin production is under tight control of a conserved splicing mechanism to avoid precocious aging.


Subject(s)
Aging, Premature/genetics , Evolution, Molecular , Lamin Type A/genetics , RNA Splicing/genetics , Animals , Base Sequence , Cells, Cultured , Conserved Sequence/genetics , Exons/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Progeria/genetics , Progeria/pathology , Protein Isoforms/genetics , Protein Precursors/genetics , RNA/chemistry , RNA/genetics , RNA Splice Sites/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Serine-Arginine Splicing Factors , Transfection
6.
Nucleic Acids Res ; 39(18): 8092-104, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21724598

ABSTRACT

Tra2ß regulates a number of splicing switches including activation of the human testis-specific exon HIPK3-T in the Homeodomain Interacting Protein Kinase 3 gene. By testing HIPK3-T exons of different intrinsic strengths, we found Tra2ß most efficiently activated splicing inclusion of intrinsically weak exons, although these were spliced at a lower overall level. Both the RRM and N-terminal RS-rich region of Tra2ß were required for splicing activation. Bioinformatic searches for splicing enhancers and repressors mapped four physically distinct exonic splicing enhancers (ESEs) within HIPK3-T, each containing the known Tra2ß AGAA-rich binding site. Surprisingly disruption of each single ESE prevented Tra2ß-mediated activation, although single mutated exons could still bind Tra2ß protein by gel shifts and functional splicing analyses. Titration experiments indicate an additive model of HIPK3-T splicing activation, requiring availability of an array of four distinct ESEs to enable splicing activation. To enable this efficient Tra2ß-mediated splicing switch to operate, a closely adjacent downstream and potentially competitive stronger 5'-splice site is actively repressed. Our data indicate that a novel arrangement of multiple mono-specific AGAA-rich ESEs coupled to a weak 5'-splice site functions as a responsive gauge. This gauge monitors changes in the specific nuclear concentration of the RNA binding protein Tra2ß, and co-ordinately regulates HIPK3-T exon splicing inclusion.


Subject(s)
Alternative Splicing , Intracellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid , Exons , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , RNA Splice Sites , RNA-Binding Proteins/chemistry , Serine-Arginine Splicing Factors
7.
Nucleic Acids Res ; 39(8): 3388-403, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21183462

ABSTRACT

Retroviruses require both spliced and unspliced RNAs for replication. Accumulation of Rous Sarcoma virus (RSV) unspliced RNA depends upon the negative regulator of splicing (NRS). Its 5'-part is considered as an ESE binding SR proteins. Its 3'-part contains a decoy 5'-splice site (ss), which inhibits splicing at the bona fide 5'-ss. Only the 3D structure of a small NRS fragment had been experimentally studied. Here, by chemical and enzymatic probing, we determine the 2D structure of the entire RSV NRS. Structural analysis of other avian NRSs and comparison with all sequenced avian NRSs is in favour of a phylogenetic conservation of the NRS 2D structure. By combination of approaches: (i) in vitro and in cellulo splicing assays, (ii) footprinting assays and (iii) purification and analysis of reconstituted RNP complex, we define a small NRS element retaining splicing inhibitory property. We also demonstrate the capability of the SR protein 9G8 to increase NRS activity in vitro and in cellulo. Altogether these data bring new insights on how NRS fine tune splicing activity.


Subject(s)
Alternative Splicing , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Viral/chemistry , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid , Rous sarcoma virus/genetics , Base Sequence , Binding Sites , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Proteins , Nucleic Acid Conformation , RNA, Viral/metabolism , Serine-Arginine Splicing Factors
8.
Nucleic Acids Res ; 38(4): 1353-66, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19965769

ABSTRACT

Alternative splicing is regulated in part by variations in the relative concentrations of a variety of factors, including serine/arginine-rich (SR) proteins. The SR protein SC35 self-regulates its expression by stimulating unproductive splicing events in the 3' untranslated region of its own pre-mRNA. Using various minigene constructs containing the terminal retained intron and flanking exons, we identified in the highly conserved last exon a number of exonic splicing enhancer elements responding specifically to SC35, and showed an inverse correlation between affinity of SC35 and enhancer strength. The enhancer region, which is included in a long stem loop, also contains repressor elements, and is recognized by other RNA-binding proteins, notably hnRNP H protein and TAR DNA binding protein (TDP-43). Finally, in vitro and in cellulo experiments indicated that hnRNP H and TDP-43 antagonize the binding of SC35 to the terminal exon and specifically repress the use of SC35 terminal 3' splice site. Our study provides new information about the molecular mechanisms of SC35-mediated splicing activation. It also highlights the existence of a complex network of self- and cross-regulatory mechanisms between splicing regulators, which controls their homeostasis and offers many ways of modulating their concentration in response to the cellular environment.


Subject(s)
Alternative Splicing , Introns , Nuclear Proteins/genetics , RNA-Binding Proteins/genetics , Base Sequence , Binding Sites , Binding, Competitive , DNA-Binding Proteins/metabolism , Exons , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Molecular Sequence Data , Nuclear Proteins/metabolism , RNA Splice Sites , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid , Serine-Arginine Splicing Factors
9.
J Cell Sci ; 123(Pt 1): 40-50, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20016065

ABSTRACT

RBMY is a male germline RNA binding protein and potential alternative splicing regulator, but the lack of a convenient biological system has made its cellular functions elusive. We found that human RBMY fused to green fluorescent protein was strictly nuclear in transfected cells, but spatially enriched in areas around nuclear speckles with some components of the exon junction complex (EJC). Human RBMY (hRBMY) and the EJC components Magoh and Y14 also physically interacted but, unlike these two proteins, hRBMY protein did not shuttle to the cytoplasm. In addition, it relocalised into nucleolar caps after inhibition of RNA polymerase II transcription. Protein interactions were also detected between RBMY and splicing factors 9G8 and transformer-2 protein homolog beta (Tra2-beta), mediated by multiple regions of the RBMY protein that contain serine/arginine-rich dipeptides, but not by the single region lacking such dipeptides. These interactions modulated the splicing of several pre-mRNAs regulated by 9G8 and Tra2-beta. Importantly, ectopic expression of hRBMY stimulated the inclusion of a testis-enriched exon from the Acinus gene, whereas 9G8 and Tra2-beta repressed this exon. We propose that hRBMY associates with regions of the nucleus enriched in nascent RNA and participates in the regulation of specific splicing events in the germline by modulating the activity of constitutively expressed splicing factors.


Subject(s)
Alternative Splicing , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA-Binding Proteins/metabolism , Testis/metabolism , Animals , Arginine , HeLa Cells , Humans , Male , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/chemistry , Nucleocytoplasmic Transport Proteins/chemistry , Protein Binding , Protein Engineering , Protein Transport , RNA Polymerase II/metabolism , RNA-Binding Proteins/chemistry , Serine , Serine-Arginine Splicing Factors , Testis/cytology , Transcriptional Activation
10.
PLoS Genet ; 5(11): e1000707, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19893608

ABSTRACT

The human testis has almost as high a frequency of alternative splicing events as brain. While not as extensively studied as brain, a few candidate testis-specific splicing regulator proteins have been identified, including the nuclear RNA binding proteins RBMY and hnRNP G-T, which are germ cell-specific versions of the somatically expressed hnRNP G protein and are highly conserved in mammals. The splicing activator protein Tra2beta is also highly expressed in the testis and physically interacts with these hnRNP G family proteins. In this study, we identified a novel testis-specific cassette exon TLE4-T within intron 6 of the human transducing-like enhancer of split 4 (TLE4) gene which makes a more transcriptionally repressive TLE4 protein isoform. TLE4-T splicing is normally repressed in somatic cells because of a weak 5' splice site and surrounding splicing-repressive intronic regions. TLE4-T RNA pulls down Tra2beta and hnRNP G proteins which activate its inclusion. The germ cell-specific RBMY and hnRNP G-T proteins were more efficient in stimulating TLE4-T incorporation than somatically expressed hnRNP G protein. Tra2b bound moderately to TLE4-T RNA, but more strongly to upstream sites to potently activate an alternative 3' splice site normally weakly selected in the testis. Co-expression of Tra2beta with either hnRNP G-T or RBMY re-established the normal testis physiological splicing pattern of this exon. Although they can directly bind pre-mRNA sequences around the TLE4-T exon, RBMY and hnRNP G-T function as efficient germ cell-specific splicing co-activators of TLE4-T. Our study indicates a delicate balance between the activity of positive and negative splicing regulators combinatorially controls physiological splicing inclusion of exon TLE4-T and leads to modulation of signalling pathways in the testis. In addition, we identified a high-affinity binding site for hnRNP G-T protein, showing it is also a sequence-specific RNA binding protein.


Subject(s)
Alternative Splicing , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Nuclear Proteins/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Spermatozoa/metabolism , Testis/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Exons , Humans , Introns , Male , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
11.
BMC Cell Biol ; 10: 82, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19912651

ABSTRACT

BACKGROUND: Active pre-mRNA splicing occurs co-transcriptionally, and takes place throughout the nucleoplasm of eukaryotic cells. Splicing decisions are controlled by networks of nuclear RNA-binding proteins and their target sequences, sometimes in response to signalling pathways. Sam68 (Src-associated in mitosis 68 kDa) is the prototypic member of the STAR (Signal Transduction and Activation of RNA) family of RNA-binding proteins, which regulate splicing in response to signalling cascades. Nuclear Sam68 protein is concentrated within subnuclear organelles called SLM/Sam68 Nuclear Bodies (SNBs), which also contain some other splicing regulators, signalling components and nucleic acids. RESULTS: We used proteomics to search for the major interacting protein partners of nuclear Sam68. In addition to Sam68 itself and known Sam68-associated proteins (heterogeneous nuclear ribonucleoproteins hnRNP A1, A2/B1 and G), we identified hnRNP L as a novel Sam68-interacting protein partner. hnRNP L protein was predominantly present within small nuclear protein complexes approximating to the expected size of monomers and dimers, and was quantitatively associated with nucleic acids. hnRNP L spatially co-localised with Sam68 as a novel component of SNBs and was also observed within the general nucleoplasm. Localisation within SNBs was highly specific to hnRNP L and was not shared by the closely-related hnRNP LL protein, nor any of the other Sam68-interacting proteins we identified by proteomics. The interaction between Sam68 and hnRNP L proteins was observed in a cell line which exhibits low frequency of SNBs suggesting that this association also takes place outside SNBs. Although ectopic expression of hnRNP L and Sam68 proteins independently affected splicing of CD44 variable exon v5 and TJP1 exon 20 minigenes, these proteins did not, however, co-operate with each other in splicing regulation of these target exons. CONCLUSION: Here we identify hnRNP L as a novel SNB component. We show that, compared with other identified Sam68-associated hnRNP proteins and hnRNP LL, this co-localisation within SNBs is specific to hnRNP L. Our data suggest that the novel Sam68-hnRNP L protein interaction may have a distinct role within SNBs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/analysis , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Heterogeneous-Nuclear Ribonucleoprotein L/chemistry , Humans , Mice , Molecular Sequence Data , Nucleic Acids/metabolism , Proteomics
12.
Front Biosci (Landmark Ed) ; 14(7): 2714-29, 2009 01 01.
Article in English | MEDLINE | ID: mdl-19273230

ABSTRACT

Alternative splicing plays a key role in the production of numerous proteins by complex lentiviruses such as HIV-1. The study of HIV-1 RNA splicing has provided useful information not only about the physiology of the virus, but also about the general mechanisms that regulate mammalian pre-mRNA alternative splicing. Like all retroviruses, a fraction of HIV-1 transcripts remains intact to serve as genomic RNA and to code for Gag and Gag-Pol protein precursors. In addition, splicing is important for controlling the production of some viral proteins, which could otherwise have a negative effect on the infected cell. Here, we summarize how the utilization of HIV-1 splicing sites is limited by the binding of nuclear factors to cis-acting silencer elements, taking into account the role of RNA secondary structure in these mechanisms. We also describe how the poorly efficient HIV-1 acceptor sites are nevertheless activated by serine/arginine-rich proteins. Finally, we discuss how nuclear factors that interact with both the transcription and splicing machineries also participate in the control of HIV-1 RNA splicing.


Subject(s)
Alternative Splicing , HIV-1/genetics , Nucleic Acid Conformation , RNA, Viral/physiology , Viral Proteins/physiology , Exons , RNA, Viral/chemistry , Regulatory Sequences, Nucleic Acid
13.
EMBO Rep ; 8(4): 372-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17318228

ABSTRACT

The RBMY (RNA-binding motif gene on Y chromosome) protein encoded by the human Y chromosome is important for normal sperm development. Although its precise molecular RNA targets are unknown at present, it is suggested that human RBMY (hRBMY) participates in splicing in the testis. Using systematic evolution of ligands by exponential enrichment, we found that RNA stem-loops capped by a C(A)/(U)CAA pentaloop are high-affinity binding targets for hRBMY. Subsequent nuclear magnetic resonance structural determination of the hRBMY RNA recognition motif (RRM) in complex with a high-affinity target showed two distinct modes of RNA recognition. First, the RRM beta-sheet surface binds to the RNA loop in a sequence-specific fashion. Second, the beta2-beta3 loop of the hRBMY inserts into the major groove of the RNA stem. The first binding mode might be conserved in the paralogous protein heterogeneous nuclear RNP G, whereas the second mode of binding is found only in hRBMY. This structural difference could be at the origin of the function of RBMY in spermatogenesis.


Subject(s)
Nuclear Proteins/chemistry , RNA-Binding Proteins/chemistry , RNA/chemistry , Testis/metabolism , Amino Acid Motifs , Amino Acid Sequence , Directed Molecular Evolution , Electrophoretic Mobility Shift Assay , Humans , Male , Molecular Sequence Data , Mutagenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Structure, Secondary , RNA/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , SELEX Aptamer Technique , Sequence Analysis, Protein
14.
J Cell Sci ; 120(Pt 2): 309-19, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17200140

ABSTRACT

The scaffold attachment factor SAFB1 and its recently discovered homologue SAFB2 might provide an important link between pre-mRNA splicing, intracellular signalling and transcription. Using novel mono-specific antisera, we found endogenous SAFB2 protein has a different spatial distribution from SAFB1 within the nucleus where it is found in much larger nuclear complexes (up to 670 kDa in size), and a distinct pattern of expression in adult human testis. By contrast, SAFB1 protein predominantly exists either as smaller complexes or as a monomeric protein. Our results suggest stable core complexes containing components comprised of SAFB1, SAFB2 and the RNA binding proteins Sam68 and hnRNPG exist in parallel with free SAFB1 protein. We found that SAFB2 protein, like SAFB1, acts as a negative regulator of a tra2beta variable exon. Despite showing an involvement in splicing, we detected no stable interaction between SAFB proteins and SR or SR-related splicing regulators, although these were also found in stable higher molecular mass complexes. Each of the detected alternative splicing regulator complexes exists independently of intact nucleic acids, suggesting they might be pre-assembled and recruited to nascent transcripts as modules to facilitate alternative splicing, and/or they represent nuclear storage compartments from which active proteins are recruited.


Subject(s)
Alternative Splicing , Matrix Attachment Region Binding Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , RNA Splicing , Receptors, Estrogen/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Line , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Male , Matrix Attachment Region Binding Proteins/genetics , Models, Biological , Nuclear Matrix-Associated Proteins/genetics , Protein Structure, Tertiary , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , RNA-Binding Proteins/metabolism , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Two-Hybrid System Techniques
15.
EMBO J ; 25(21): 5126-37, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17036044

ABSTRACT

The sequence-specific RNA-binding proteins SRp20 and 9G8 are the smallest members of the serine- and arginine-rich (SR) protein family, well known for their role in splicing. They also play a role in mRNA export, in particular of histone mRNAs. We present the solution structures of the free 9G8 and SRp20 RNA recognition motifs (RRMs) and of SRp20 RRM in complex with the RNA sequence 5'CAUC3'. The SRp20-RNA structure reveals that although all 4 nt are contacted by the RRM, only the 5' cytosine is primarily recognized in a specific way. This might explain the numerous consensus sequences found by SELEX (systematic evolution of ligands by exponential enrichment) for the RRM of 9G8 and SRp20. Furthermore, we identify a short arginine-rich peptide adjacent to the SRp20 and 9G8 RRMs, which does not contact RNA but is necessary and sufficient for interaction with the export factor Tip-associated protein (TAP). Together, these results provide a molecular description for mRNA and TAP recognition by SRp20 and 9G8.


Subject(s)
Cell Nucleus/metabolism , Models, Molecular , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Motifs , Humans , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins , Nucleocytoplasmic Transport Proteins/chemistry , Nucleocytoplasmic Transport Proteins/genetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Binding/genetics , Protein Structure, Tertiary , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors
16.
Hum Mol Genet ; 14(16): 2289-303, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16000324

ABSTRACT

We have discovered a new exon of the homeodomain-interacting kinase HipK3 that incorporates a premature stop codon and is included only in the human testis. To investigate this, we tested the effects of transfecting cells with green fluorescent protein fusions of RNA-binding proteins implicated in spermatogenesis using a novel assay based on multi-fraction fluorescence-activated cell sorting (MF-FACS). This allows the effect of a controlled titration of any splicing factor on the splicing of endogenous genes to be studied in vivo. We found that Tra2beta recapitulates testis-specific splicing of endogenous HipK3 in a concentration-dependent manner and binds specifically to a long purine-rich sequence in the novel exon. This sequence was also specifically bound by hnRNP A1, hnRNP H, ASF/SF2 and SRp40, but not by 9G8. Consistent with these observations, in vitro studies showed that this sequence shifts splicing to a downstream 5' splice site within a heterologous pre-mRNA substrate in the presence of Tra2beta, ASF/SF2 and SRp40, whereas hnRNP A1 specifically inhibits this choice. By mutating the purine-rich sequence in the context of the HipK3 gene, we also show that it is the major determinant of Tra2beta- and hnRNP A1-mediated regulation. Tra2 is essential for sex determination and spermatogenesis in flies, and Tra2beta protein was most highly expressed in testis out of six mouse tissues, whereas hnRNP A1 is down-regulated during germ cell development. Therefore, our data imply an evolutionarily conserved role for Tra2 proteins in spermatogenesis and suggest that an elevated concentration of Tra2beta may convert it into a tissue-specific splicing factor.


Subject(s)
Alternative Splicing , Exons , RNA-Binding Proteins/metabolism , Testis/metabolism , Animals , Base Sequence , Drug Resistance, Multiple , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Molecular Sequence Data , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/genetics , Spermatogenesis , Up-Regulation
17.
Article in English | MEDLINE | ID: mdl-15210328

ABSTRACT

Alternative splicing of pre-messenger RNA (pre-mRNA) is a highly regulated process that allows expansion of the potential of expression of the genome in higher eukaryotes and involves many factors. Among them, the family of the serine- and arginine-rich proteins (SR proteins) plays a pivotal role: it has essential functions during spliceosome assembly and also interacts with RNA regulatory sequences on the pre-mRNA as well as with multiple cofactors. Collectively, SR proteins, because of their capacity to recognize multiple RNA sequences with a broad specificity, are at the heart of the regulation pathways that lead to the choice of alternative splice sites. Moreover, a growing body of evidence shows that the mechanisms of splicing regulation are not limited to the basic involvement of cis- and trans-acting factors at the pre-mRNA level, but result from intricate pathways, initiated sometimes by stimuli that are external to the cell and integrate SR proteins (and other factors) within an extremely sophisticated network of molecular machines associated with one another. This review focuses on the molecular aspects of the functions of SR proteins. In particular, we discuss the different ways in which SR proteins manage to achieve a high level of specificity in splicing regulation, even though they are also involved in the constitutive reaction.


Subject(s)
Alternative Splicing/physiology , RNA-Binding Proteins/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Enhancer Elements, Genetic , Humans , Models, Biological , Molecular Sequence Data , Protein Structure, Tertiary , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Sequence Homology, Amino Acid , Signal Transduction
18.
J Biol Chem ; 279(29): 29963-73, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15123677

ABSTRACT

Splicing is a crucial step for human immunodeficiency virus, type 1 (HIV-1) multiplication; eight acceptor sites are used in competition to produce the vif, vpu, vpr, nef, env, tat, and rev mRNAs. The effects of SR proteins have only been investigated on a limited number of HIV-1 splicing sites by using small HIV-1 RNA pieces. To understand how SR proteins influence the use of HIV-1 splicing sites, we tested the effects of overproduction of individual SR proteins in HeLa cells on the splicing pattern of an HIV-1 RNA that contained all the splicing sites. The steady state levels of the HIV-1 mRNAs produced were quantified by reverse transcriptase-PCR. For interpretation of the data, transcripts containing one or several of the HIV-1 acceptor sites were spliced in vitro in the presence or the absence of one of the tested SR proteins. Both in vivo and in vitro, acceptor sites A2 and A3 were found to be strongly and specifically regulated by SR proteins. ASF/SF2 strongly activates site A2 and to a lesser extent site A1. As a result, upon ASF/SF2 overexpression, the vpr mRNA steady state level is specifically increased. SC35 and SRp40, but not 9G8, strongly activate site A3, and their overexpression ex vivo induces a dramatic accumulation of the tat mRNA, to the detriment of most of the other viral mRNAs. Here we showed by Western blot analysis that the Nef protein synthesis is strongly decreased by overexpression of SC35, SRp40, and ASF/SF2. Finally, activation by ASF/SF2 and 9G8 was found to be independent of the RS domain. This is the first investigation of the effects of variations of individual SR protein concentrations that is performed ex vivo on an RNA containing a complex set of splicing sites.


Subject(s)
HIV-1/metabolism , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/metabolism , RNA, Viral , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Sarcoplasmic Reticulum/metabolism , Alternative Splicing , Binding Sites , Blotting, Western , HeLa Cells , Humans , Models, Genetic , Plasmids/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA, Small Nuclear/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serine-Arginine Splicing Factors , Transcription, Genetic
19.
Nucleic Acids Res ; 32(3): 1214-23, 2004.
Article in English | MEDLINE | ID: mdl-14973203

ABSTRACT

Coffin-Lowry syndrome (CLS) is caused by mutations in the RSK2 gene encoding a protein kinase of the Ras signalling pathway. We have studied two point mutations which cause aberrant splicing but do not concern the invariant GT or AG nucleotides of splice sites. The first, an A-->G transition at position +3 of the 5' splice site of exon 6, results in vivo and in vitro in exon skipping and premature translation termination. The natural 5' splice site, although intrinsically weak, is not transactivated under normal conditions. Consequently, replacement of an A/U by a G/U base pairing with U1 snRNA reduces its strength below a critical threshold. The second mutation, an A-->G transition 11 nt upstream of exon 5, creates a new AG near the natural 3' splice site. In vitro this synthetic 3' AG is used exclusively by the splicing machinery. In vivo this splicing event is also observed, but is underestimated because the resulting RSK2 mRNA contains premature stop codons which trigger the nonsense-mediated decay process. We show that a particular mechanism is involved in the aberrant splicing of exon 5, implying involvement of the natural 3' AG during the first catalytic step and the new 3' AG during the second step. Thus, our results explain how these mutations cause severe forms of CLS.


Subject(s)
Coffin-Lowry Syndrome/genetics , Introns , Point Mutation , RNA Splicing , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Base Sequence , Cell Line , Coffin-Lowry Syndrome/enzymology , HeLa Cells , Humans , RNA Splice Sites , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
20.
Cancer Res ; 63(23): 8203-11, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14678976

ABSTRACT

DNA topoisomerase I (Topo I) specifically phosphorylates arginine-serine-rich (SR proteins) splicing factors and is potentially involved in pre-mRNA-splicing regulation. Using a Topo I-deficient murine B lymphoma-derived subclone (P388-45/C) selected for its resistance to high dosage of the antitumor drug camptothecin, we show that Topo I depletion results in the hypophosphorylation of SR proteins and impairs exonic splicing enhancer (ESE)-dependent but not constitutive splicing. The Affymetrix GeneChip system analysis revealed that several alternatively spliced genes, characterized by small exons and large introns, are down-regulated in Topo I-deficient cells. Given that ectopic expression of green fluorescent protein-Topo I fusion in Topo I-deficient cells restores both wild-type phosphorylation of SR proteins and ESE-dependent splicing, we conclude that Topo I-mediated phosphorylation plays a specific role in ESE-regulated splicing.


Subject(s)
DNA Topoisomerases, Type I/deficiency , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , RNA Splicing/physiology , Alternative Splicing , Animals , Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , DNA Topoisomerases, Type I/genetics , Down-Regulation , Drug Resistance, Neoplasm , Exons/physiology , Gene Expression Regulation, Leukemic , Green Fluorescent Proteins , Leukemia P388/enzymology , Leukemia P388/genetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Phosphorylation , RNA-Binding Proteins , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine-Arginine Splicing Factors , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...