Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Hum Mutat ; 28(6): 599-612, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17311297

RESUMO

We describe 94 pathogenic NF1 gene alterations in a cohort of 97 Austrian neurofibromatosis type 1 patients meeting the NIH criteria. All mutations were fully characterized at the genomic and mRNA levels. Over half of the patients carried novel mutations, and only a quarter carried recurrent minor-lesion mutations at 16 mutational warm spots. The remaining patients carried NF1 microdeletions (7%) and rare recurring mutations. Thirty-six of the mutations (38%) altered pre-mRNA splicing, and fall into five groups: exon skipping resulting from mutations at authentic splice sites (type I), cryptic exon inclusion caused by deep intronic mutations (type II), creation of de novo splice sites causing loss of exonic sequences (type III), activation of cryptic splice sites upon authentic splice-site disruption (type IV), and exonic sequence alterations causing exon skipping (type V). Extensive in silico analyses of 37 NF1 exons and surrounding intronic sequences suggested that the availability of a cryptic splice site combined with a strong natural upstream 3' splice site (3'ss)is the main determinant of cryptic splice-site activation upon 5' splice-site disruption. Furthermore, the exonic sequences downstream of exonic cryptic 5' splice sites (5'ss) resemble intronic more than exonic sequences with respect to exonic splicing enhancer and silencer density, helping to distinguish between exonic cryptic and pseudo 5'ss. This study provides valuable predictors for the splicing pathway used upon 5'ss mutation, and underscores the importance of using RNA-based techniques, together with methods to identify microdeletions and intragenic copy-number changes, for effective and reliable NF1 mutation detection.


Assuntos
Processamento Alternativo/genética , Genes da Neurofibromatose 1 , Mutação , Neurofibromatose 1/genética , Sítios de Splice de RNA/genética , Adulto , Áustria , Criança , Estudos de Coortes , Simulação por Computador , Análise Mutacional de DNA , Éxons , Humanos , Neurofibromatose 1/diagnóstico , Sensibilidade e Especificidade , Deleção de Sequência
2.
J Virol ; 75(18): 8487-97, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11507194

RESUMO

The synthesis of human immunodeficiency virus type 1 (HIV-1) mRNAs is a complex process by which more than 30 different mRNA species are produced by alternative splicing of a single primary RNA transcript. HIV-1 splice sites are used with significantly different efficiencies, resulting in different levels of mRNA species in infected cells. Splicing of Tat mRNA, which is present at relatively low levels in infected cells, is repressed by the presence of exonic splicing silencers (ESS) within the two tat coding exons (ESS2 and ESS3). These ESS elements contain the consensus sequence PyUAG. Here we show that the efficiency of splicing at 3' splice site A2, which is used to generate Vpr mRNA, is also regulated by the presence of an ESS (ESSV), which has sequence homology to ESS2 and ESS3. Mutagenesis of the three PyUAG motifs within ESSV increases splicing at splice site A2, resulting in increased Vpr mRNA levels and reduced skipping of the noncoding exon flanked by A2 and D3. The increase in Vpr mRNA levels and the reduced skipping also occur when splice site D3 is mutated toward the consensus sequence. By in vitro splicing assays, we show that ESSV represses splicing when placed downstream of a heterologous splice site. A1, A1(B), A2, and B1 hnRNPs preferentially bind to ESSV RNA compared to ESSV mutant RNA. Each of these proteins, when added back to HeLa cell nuclear extracts depleted of ESSV-binding factors, is able to restore splicing repression. The results suggest that coordinate repression of HIV-1 RNA splicing is mediated by members of the hnRNP A/B protein family.


Assuntos
Regiões 3' não Traduzidas , Éxons , Inativação Gênica , HIV-1/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B , Splicing de RNA , RNA Viral , Ribonucleoproteínas/metabolismo , Regiões 5' não Traduzidas , Sítios de Ligação , Sequência Consenso , Células HeLa , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas , Humanos , Mutagênese
3.
RNA ; 7(3): 471-82, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11333026

RESUMO

SR proteins play critical roles in the major pre-mRNA splicing pathway. A second pathway processes U12-dependent AT-AC introns. We demonstrate, by biochemical complementation, the requirement for SR proteins in splicing of AT-AC introns. Whereas SR proteins were sufficient to activate splicing of a P120 AT-AC intron, splicing of a sodium channel AT-AC intron required an additional nuclear fraction. Individual recombinant SR proteins promoted splicing of both substrates, but displayed marked preferences. SR proteins supported basal AT-AC splicing, and also splicing stimulation via a downstream enhancer or conventional 5' splice site. Analysis of chimeric transcripts revealed that information dispersed throughout exons and introns dictates SR protein specificity and the requirement for the additional nuclear fraction. Thus, SR proteins function in both major and minor splicing pathways, and in coordinating the activities of both spliceosomes via exon definition. These results suggest that despite the substantial differences in intron consensus sequences and in four of the five snRNPs in each spliceosome, at least some of the interactions involving SR proteins are conserved between the two pathways.


Assuntos
Éxons , Íntrons , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Precursores de RNA/metabolismo , Splicing de RNA , RNA Mensageiro/metabolismo , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Sistema Livre de Células , Células HeLa , Humanos , Canal de Sódio Disparado por Voltagem NAV1.4 , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina , Canais de Sódio/genética
4.
Curr Opin Cell Biol ; 13(3): 302-9, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11343900

RESUMO

The past year has witnessed refinements in models of spliceosome assembly pathways and in the understanding of how splicing factors of the serine/arginine-rich (SR) protein family function. The role of splicing in human genetic diseases has also received a lot of attention recently as exonic splicing enhancers become better understood.


Assuntos
Proteínas Nucleares/metabolismo , Precursores de RNA/metabolismo , Sítios de Splice de RNA/fisiologia , Splicing de RNA/fisiologia , Spliceossomos/metabolismo , Animais , Terapia Biológica/métodos , Variação Genética/genética , Humanos , Modelos Moleculares , Proteínas Nucleares/genética , Precursores de RNA/genética , Sítios de Splice de RNA/genética , Splicing de RNA/genética , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina , Spliceossomos/genética
5.
EMBO J ; 20(4): 864-71, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11179230

RESUMO

SR proteins purified from uninfected HeLa cells inhibit adenovirus IIIa pre-mRNA splicing by binding to the intronic IIIa repressor element (3RE). In contrast, SR proteins purified from late adenovirus-infected cells are functionally inactivated as splicing repressor proteins by a virus-induced dephosphorylation. We have shown that the adenovirus E4-ORF4 protein, which binds the cellular protein phos phatase 2A (PP2A) and activates IIIa splicing in vitro and in vivo, induces SR protein dephosphorylation. Here we show that E4-ORF4 interacts with only a subset of SR proteins present in HeLa cells. Thus, E4-ORF4 interacts efficiently with SF2/ASF and SRp30c, but not with other SR proteins. Interestingly, E4-ORF4 interacts with SF2/ASF through the latter's RNA recognition motifs. Furthermore, E4-ORF4 interacts preferentially with the hyperphosphorylated form of SR proteins found in uninfected HeLa cells. E4-ORF4 mutant proteins that fail to bind strongly to PP2A or SF2/ASF do not relieve the repressive effect of HeLa SR proteins on IIIa pre-mRNA splicing in transient transfection experiments, suggesting that an interaction between all three proteins is required for E4-ORF4-induced SR protein dephosphorylation.


Assuntos
Proteínas E4 de Adenovirus/metabolismo , Proteínas de Neoplasias/metabolismo , Splicing de RNA , Células HeLa , Humanos , Fases de Leitura Aberta , Fosforilação , Ligação Proteica
6.
Nat Genet ; 27(1): 55-8, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11137998

RESUMO

Point mutations can generate defective and sometimes harmful proteins. The nonsense-mediated mRNA decay (NMD) pathway minimizes the potential damage caused by nonsense mutations. In-frame nonsense codons located at a minimum distance upstream of the last exon-exon junction are recognized as premature termination codons (PTCs), targeting the mRNA for degradation. Some nonsense mutations cause skipping of one or more exons, presumably during pre-mRNA splicing in the nucleus; this phenomenon is termed nonsense-mediated altered splicing (NAS), and its underlying mechanism is unclear. By analyzing NAS in BRCA1, we show here that inappropriate exon skipping can be reproduced in vitro, and results from disruption of a splicing enhancer in the coding sequence. Enhancers can be disrupted by single nonsense, missense and translationally silent point mutations, without recognition of an open reading frame as such. These results argue against a nuclear reading-frame scanning mechanism for NAS. Coding-region single-nucleotide polymorphisms (cSNPs) within exonic splicing enhancers or silencers may affect the patterns or efficiency of mRNA splicing, which may in turn cause phenotypic variability and variable penetrance of mutations elsewhere in a gene.


Assuntos
Códon sem Sentido/genética , Éxons/genética , Genes BRCA1/genética , Mutação de Sentido Incorreto/genética , Splicing de RNA/genética , Motivos de Aminoácidos , Substituição de Aminoácidos/genética , Proteína BRCA1/química , Proteína BRCA1/genética , Sequência de Bases , Elementos Facilitadores Genéticos/genética , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/química , Fases de Leitura Aberta/genética , Fenótipo , Fosfoproteínas/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina
7.
Mol Cell ; 8(6): 1351-61, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11779509

RESUMO

SR proteins recognize exonic splicing enhancer (ESE) elements and promote exon use, whereas certain hnRNP proteins bind to exonic splicing silencer (ESS) elements and block exon recognition. We investigated how ESS3 in HIV-1 tat exon 3 blocks splicing promoted by one SR protein (SC35) but not another (SF2/ASF). hnRNP A1 mediates silencing by binding initially to a required high-affinity site in ESS3, which then promotes further hnRNP A1 association with the upstream region of the exon. Both SC35 and SF2/ASF recognize upstream ESE motifs, but only SF2/ASF prevents secondary hnRNP A1 binding, presumably by blocking its cooperative propagation along the exon. The differential antagonism between a negative and two positive regulators exemplifies how inclusion of an alternative exon can be modulated.


Assuntos
Éxons/genética , Inativação Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Splicing de RNA/genética , Sequências Reguladoras de Ácido Nucleico/genética , Ribonucleoproteínas/antagonistas & inibidores , Sequência de Bases , Sítios de Ligação , Extratos Celulares , Produtos do Gene tat/genética , Teste de Complementação Genética , Glicina/metabolismo , HIV-1/genética , Células HeLa , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas , Humanos , Cinética , Modelos Genéticos , Mutação/genética , Ligação Proteica , Estrutura Terciária de Proteína , Precursores de RNA/química , Precursores de RNA/genética , Precursores de RNA/metabolismo , Proteínas de Ligação a RNA , Ribonucleoproteínas/química , Ribonucleoproteínas/deficiência , Ribonucleoproteínas/metabolismo , Fatores de Processamento de Serina-Arginina , Especificidade por Substrato , Produtos do Gene tat do Vírus da Imunodeficiência Humana
8.
Genes Dev ; 14(24): 3166-78, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11124808

RESUMO

SR proteins are essential pre-mRNA splicing factors that act at the earliest stages of splice-site recognition and spliceosome assembly, as well as later in the splicing pathway. SR proteins consist of one or two RNA-recognition motifs and a characteristic arginine/serine-rich C-terminal RS domain. The RS domain, which is extensively phosphorylated, mediates the subcellular localization of individual SR proteins and also functions as a splicing activation module, apparently by engaging in protein-protein interactions. The RS domain of SF2/ASF is dispensable for the concentration-dependent effects of this SR protein on alternative splice-site selection. However, this RS domain is highly conserved phylogenetically, and was shown to be required for constitutive splicing in vitro and for cell viability. Here, we demonstrate that the RS domain of SF2/ASF is, in fact, dispensable for splicing of several substrates, including constitutive and enhancer-dependent pre-mRNAs. The requirement for this RS domain is substrate specific, and correlates with the strength of the splicing signals. When the 3' splice site is weak, both the SF2/ASF RS domain and U2AF(35) are required for splicing. These results show the existence of an RS domain-independent function of SR proteins in constitutive and enhancer-dependent splicing, and suggest mechanisms for their role in enhancer function besides U2AF recruitment.


Assuntos
Proteínas Nucleares/metabolismo , Precursores de RNA/genética , Splicing de RNA , RNA Mensageiro/genética , Sequência de Bases , Elementos Facilitadores Genéticos , Éxons , Humanos , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Sítios de Splice de RNA , Proteínas de Ligação a RNA , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Fatores de Processamento de Serina-Arginina , Fator de Processamento U2AF
9.
Virology ; 275(1): 145-57, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11017796

RESUMO

The Epstein-Barr virus (EBV) nuclear antigen-1 (EBNA-1) is required for the maintenance of the viral chromosome in latently infected, proliferating cells and plays a role in latent cycle DNA replication. EBNA-1 also functions as a positive and negative regulator of EBV gene expression. We have investigated the interaction of EBNA-1 with p32, a host mitochondrial protein that associates with EBNA-1 in EBV-positive Burkitt's lymphoma cells. Using a chromatin immunoprecipitation assay, we found that a fraction of p32 localizes to the viral latent cycle origin of DNA replication oriP in vivo. p32 binds EBNA-1 independently of other proteins or DNA. EBNA-1 variants lacking one of two p32 binding elements did not interact stably with p32 in cultured cells and were defective for both transcriptional activation of a reporter gene linked to oriP FR and replication and/or maintenance of a plasmid bearing oriP. These results support a role for p32 in transcriptional activation by EBNA-1 and suggest that p32 plays a role in EBV latent cycle DNA replication.


Assuntos
Replicação do DNA/genética , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Receptores de Hialuronatos , Proteínas/metabolismo , Ativação Transcricional , Latência Viral/genética , Replicação Viral/genética , Animais , Sítios de Ligação , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patologia , Proteínas de Transporte , Linhagem Celular , Núcleo Celular/química , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , DNA/genética , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/genética , Regulação Viral da Expressão Gênica , Genes Reporter/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiologia , Humanos , Proteínas Mitocondriais , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Plasmídeos/genética , Testes de Precipitina , Ligação Proteica , Proteínas/genética , Origem de Replicação/genética , Deleção de Sequência/genética , Especificidade por Substrato , Transfecção , Células Tumorais Cultivadas
10.
Mol Cell Biol ; 20(22): 8303-18, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11046128

RESUMO

The first component known to recognize and discriminate among potential 5' splice sites (5'SSs) in pre-mRNA is the U1 snRNP. However, the relative levels of U1 snRNP binding to alternative 5'SSs do not necessarily determine the splicing outcome. Strikingly, SF2/ASF, one of the essential SR protein-splicing factors, causes a dose-dependent shift in splicing to a downstream (intron-proximal) site, and yet it increases U1 snRNP binding at upstream and downstream sites simultaneously. We show here that hnRNP A1, which shifts splicing towards an upstream 5'SS, causes reduced U1 snRNP binding at both sites. Nonetheless, the importance of U1 snRNP binding is shown by proportionality between the level of U1 snRNP binding to the downstream site and its use in splicing. With purified components, hnRNP A1 reduces U1 snRNP binding to 5'SSs by binding cooperatively and indiscriminately to the pre-mRNA. Mutations in hnRNP A1 and SF2/ASF show that the opposite effects of the proteins on 5'SS choice are correlated with their effects on U1 snRNP binding. Cross-linking experiments show that SF2/ASF and hnRNP A1 compete to bind pre-mRNA, and we conclude that this competition is the basis of their functional antagonism; SF2/ASF enhances U1 snRNP binding at all 5'SSs, the rise in simultaneous occupancy causing a shift in splicing towards the downstream site, whereas hnRNP A1 interferes with U1 snRNP binding such that 5'SS occupancy is lower and the affinities of U1 snRNP for the individual sites determine the site of splicing.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B , Proteínas Nucleares/metabolismo , Sítios de Splice de RNA , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Ribonucleoproteínas/metabolismo , Processamento Alternativo , Sítios de Ligação , Ligação Competitiva , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas , Modelos Biológicos , Proteínas Nucleares/genética , Precursores de RNA/genética , Precursores de RNA/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U1/genética , Ribonucleoproteínas/genética , Fatores de Processamento de Serina-Arginina
11.
J Biol Chem ; 275(37): 29170-7, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10880506

RESUMO

Splicing of the last intron (intron D) of the bovine growth hormone pre-mRNA requires the presence of a downstream exonic splicing enhancer (ESE). This enhancer is contained within a 115-nucleotide FspI-PvuII (FP) fragment located in the middle of the last exon (exon 5). Previous work showed that the splicing factor SF2/ASF binds to this FP region and stimulates splicing of intron D in vitro. However, the precise sequences recognized by SF2/ASF within the FP region had not been determined. Here we used multiple strategies to map the SF2/ASF binding sites and determine their importance for ESE function. Taking advantage of the fact that SF2/ASF ultraviolet (UV) cross-links specifically to RNA containing the FP sequence, we first mapped a major SF2/ASF binding site by UV cross-linking and reverse transcription. This strategy identified a 29-nucleotide SF2/ASF binding region in the middle of the FP sequence containing the 7-nucleotide purine-rich motif described previously. Interestingly, this binding region is neither sufficient, nor absolutely required for SF2/ASF-mediated splicing, suggesting that additional SF2/ASF binding sites are present. The location of these additional sites was determined by electrophoretic mobility shift analysis of various subfragments of the FP sequence. Antisense 2'-O-methyl oligoribonucleotides complementary to selected SF2/ASF binding sites block bovine growth hormone intron D splicing. Thus, multiple SF2/ASF binding sites within the exonic splicing enhancer contribute to maximal enhancer activity.


Assuntos
Elementos Facilitadores Genéticos , Éxons , Hormônio do Crescimento/genética , Proteínas Nucleares/metabolismo , Splicing de RNA , Animais , Sítios de Ligação , Bovinos , Células HeLa , Ribonucleoproteínas Nucleares Heterogêneas , Humanos , Íntrons , Precursores de RNA/metabolismo , Proteínas de Ligação a RNA , Ribonucleoproteínas/metabolismo , Fatores de Processamento de Serina-Arginina
12.
J Clin Endocrinol Metab ; 85(5): 1928-36, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10843177

RESUMO

Many of the human myometrial proteins associated with uterine quiescence and the switch to coordinated contractions at the onset of labor exist as alternatively spliced isoforms. There is now extensive evidence to indicate that the nuclear concentrations of the trans-acting splicing regulators SF2/ASF and hnRNP A1/A1B are fundamental in regulating the expression of specific protein isoforms derived from alternative splicing of single precursor messenger ribonucleic acid transcripts. The question thus arose as to whether these factors were also involved in regulating the expression of specific myometrial protein species within different uterine regions during human gestation and parturition. SF2/ASF and hnRNP A1/A1B expression was therefore determined in paired upper (corpus) and lower segment myometrial samples taken from individual women at term/during spontaneous labor and compared with nonpregnant control samples using specific monoclonal antibodies. We report that SF2/ASF levels were substantially increased in the lower uterine region, and this was associated with a parallel decrease in levels of hnRNP A1/A1B during gestation. Conversely, the opposite pattern was observed within the upper uterine region during pregnancy, where hnRNP A1/A1B was significantly up-regulated and SF2/ASF levels were much less than those found in the lower uterine segment. The differential expression of hnRNP A1/A1B and SF2/ASF in the upper and lower uterine segments may have a primary role in defining the formation of specific myometrial protein species associated with the known contractile and relaxatory properties of these regions before and during parturition.


Assuntos
Processamento Alternativo , Regulação da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B , Trabalho de Parto/metabolismo , Miométrio/metabolismo , Proteínas Nucleares/genética , Gravidez/metabolismo , Ribonucleoproteínas/genética , Feminino , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas , Humanos , Isoformas de Proteínas/genética , Proteínas de Ligação a RNA/genética , Fatores de Processamento de Serina-Arginina , Spliceossomos/genética
13.
J Biol Chem ; 275(33): 25411-7, 2000 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-10827081

RESUMO

NIPP1 is a regulatory subunit of a species of protein phosphatase-1 (PP1) that co-localizes with splicing factors in nuclear speckles. We report that the N-terminal third of NIPP1 largely consists of a Forkhead-associated (FHA) protein interaction domain, a known phosphopeptide interaction module. A yeast two-hybrid screening revealed an interaction between this domain and a human homolog (CDC5L) of the fission yeast protein cdc5, which is required for G(2)/M progression and pre-mRNA splicing. CDC5L and NIPP1 co-localized in nuclear speckles in COS-1 cells. Furthermore, an interaction between CDC5L, NIPP1, and PP1 in rat liver nuclear extracts could be demonstrated by co-immunoprecipitation and/or co-purification experiments. The binding of the FHA domain of NIPP1 to CDC5L was dependent on the phosphorylation of CDC5L, e.g. by cyclin E-Cdk2. When expressed in COS-1 or HeLa cells, the FHA domain of NIPP1 did not affect the number of cells in the G(2)/M transition. However, the FHA domain blocked beta-globin pre-mRNA splicing in nuclear extracts. A mutation in the FHA domain that abolished its interaction with CDC5L also canceled its anti-splicing effects. We suggest that NIPP1 either targets CDC5L or an associated protein for dephosphorylation by PP1 or serves as an anchor for both PP1 and CDC5L.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular/metabolismo , Endorribonucleases , Peptídeos e Proteínas de Sinalização Intracelular , Mitose , Fosfoproteínas Fosfatases/metabolismo , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Bovinos , Proteínas de Ciclo Celular/química , Núcleo Celular/metabolismo , Separação Celular , Citometria de Fluxo , Imunofluorescência , Glutationa Transferase/metabolismo , Células HeLa , Humanos , Fígado/metabolismo , Dados de Sequência Molecular , Músculo Esquelético/química , Mutação , Fosfoproteínas Fosfatases/química , Fosforilação , Testes de Precipitina , Proteína Fosfatase 1 , Estrutura Terciária de Proteína , Coelhos , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Schizosaccharomyces/química , Proteínas de Schizosaccharomyces pombe , Homologia de Sequência de Aminoácidos , Técnicas do Sistema de Duplo-Híbrido
14.
J Cell Biol ; 149(2): 307-16, 2000 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-10769024

RESUMO

Individual members of the serine-arginine (SR) and heterogeneous nuclear ribonucleoprotein (hnRNP) A/B families of proteins have antagonistic effects in regulating alternative splicing. Although hnRNP A1 accumulates predominantly in the nucleus, it shuttles continuously between the nucleus and the cytoplasm. Some but not all SR proteins also undergo nucleo-cytoplasmic shuttling, which is affected by phosphorylation of their serine/arginine (RS)-rich domain. The signaling mechanisms that control the subcellular localization of these proteins are unknown. We show that exposure of NIH-3T3 and SV-40 transformed green monkey kidney (COS) cells to stress stimuli such as osmotic shock or UVC irradiation, but not to mitogenic activators such as PDGF or EGF, results in a marked cytoplasmic accumulation of hnRNP A1, concomitant with an increase in its phosphorylation. These effects are mediated by the MKK(3/6)-p38 pathway, and moreover, p38 activation is necessary and sufficient for the induction of hnRNP A1 cytoplasmic accumulation. The stress-induced increase in the cytoplasmic levels of hnRNP A/B proteins and the concomitant decrease in their nuclear abundance are paralleled by changes in the alternative splicing pattern of an adenovirus E1A pre-mRNA splicing reporter. These results suggest the intriguing possibility that signaling mechanisms regulate pre-mRNA splicing in vivo by influencing the subcellular distribution of splicing factors.


Assuntos
Processamento Alternativo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Regulação da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Quinases/metabolismo , Ribonucleoproteínas/metabolismo , Células 3T3 , Animais , Células COS , Linhagem Celular Transformada , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas , MAP Quinase Quinase 3 , MAP Quinase Quinase 6 , Camundongos , Concentração Osmolar , Fosforilação , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes/biossíntese , Transdução de Sinais , Vírus 40 dos Símios , Transfecção , Raios Ultravioleta
15.
Mol Cell Biol ; 20(3): 1063-71, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10629063

RESUMO

Exonic splicing enhancers (ESEs) are important cis elements required for exon inclusion. Using an in vitro functional selection and amplification procedure, we have identified a novel ESE motif recognized by the human SR protein SC35 under splicing conditions. The selected sequences are functional and specific: they promote splicing in nuclear extract or in S100 extract complemented by SC35 but not by SF2/ASF. They can also function in a different exonic context from the one used for the selection procedure. The selected sequences share one or two close matches to a short and highly degenerate octamer consensus, GRYYcSYR. A score matrix was generated from the selected sequences according to the nucleotide frequency at each position of their best match to the consensus motif. The SC35 score matrix, along with our previously reported SF2/ASF score matrix, was used to search the sequences of two well-characterized splicing substrates derived from the mouse immunoglobulin M (IgM) and human immunodeficiency virus tat genes. Multiple SC35 high-score motifs, but only two widely separated SF2/ASF motifs, were found in the IgM C4 exon, which can be spliced in S100 extract complemented by SC35. In contrast, multiple high-score motifs for both SF2/ASF and SC35 were found in a variant of the Tat T3 exon (lacking an SC35-specific silencer) whose splicing can be complemented by either SF2/ASF or SC35. The motif score matrix can help locate SC35-specific enhancers in natural exon sequences.


Assuntos
Elementos Facilitadores Genéticos , Éxons , Proteínas Nucleares/metabolismo , Splicing de RNA , Ribonucleoproteínas , Animais , Sequência de Bases , Sítios de Ligação , Núcleo Celular/metabolismo , Sistema Livre de Células , Sequência Consenso , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina
16.
Proc Natl Acad Sci U S A ; 96(24): 13789-94, 1999 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-10570151

RESUMO

The conserved CDC5 family of Myb-related proteins performs an essential function in cell cycle control at G(2)/M. Although c-Myb and many Myb-related proteins act as transcription factors, herein, we implicate CDC5 proteins in pre-mRNA splicing. Mammalian CDC5 colocalizes with pre-mRNA splicing factors in the nuclei of mammalian cells, associates with core components of the splicing machinery in nuclear extracts, and interacts with the spliceosome throughout the splicing reaction in vitro. Furthermore, genetic depletion of the homolog of CDC5 in Saccharomyces cerevisiae, CEF1, blocks the first step of pre-mRNA processing in vivo. These data provide evidence that eukaryotic cells require CDC5 proteins for pre-mRNA splicing.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-myb , Precursores de RNA , Splicing de RNA , Células 3T3 , Animais , Proteínas de Ciclo Celular/genética , Núcleo Celular/metabolismo , Proteínas Fúngicas/genética , Humanos , Camundongos , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas , Proteínas de Ligação a RNA , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae , Schizosaccharomyces , Proteínas de Schizosaccharomyces pombe , Spliceossomos , Frações Subcelulares , Quinase 1 Polo-Like
19.
Proc Natl Acad Sci U S A ; 96(19): 10655-60, 1999 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-10485881

RESUMO

Exonic splicing enhancers (ESEs) activate pre-mRNA splicing by promoting the use of the flanking splice sites. They are recognized by members of the serine/arginine-rich (SR) family of proteins, such as splicing factor 2/alternative splicing factor (SF2/ASF), which recruit basal splicing factors to form the initial complexes during spliceosome assembly. The in vitro splicing kinetics of an ESE-dependent IgM pre-mRNA suggested that an SF2/ASF-specific ESE has additional functions later in the splicing reaction, after the completion of the first catalytic step. A bimolecular exon ligation assay, which physically uncouples the first and second catalytic steps of splicing in a trans-splicing reaction, was adapted to test the function of the ESE after the first step. A 3' exon containing the SF2/ASF-specific ESE underwent bimolecular exon ligation, whereas 3' exons without the ESE or with control sequences did not. The ESE-dependent trans-splicing reaction occurred after inactivation of U1 or U2 small nuclear ribonucleoprotein particles, compatible with a functional assay for events after the first step of splicing. The ESE-dependent step appears to take place before the ATP-independent part of the second catalytic step. Bimolecular exon ligation also occurred in an S100 cytosolic extract, requiring both the SF2/ASF-dependent ESE and complementation with SF2/ASF. These data suggest that some ESEs can act late in the splicing reaction, together with appropriate SR proteins, to enhance the second catalytic step of splicing.


Assuntos
Catálise , Elementos Facilitadores Genéticos/fisiologia , Precursores de RNA/metabolismo , Splicing de RNA/fisiologia , Trifosfato de Adenosina/farmacologia , Adenoviridae/genética , Escherichia coli/genética , Éxons , Imunoglobulina M/metabolismo , Cinética , Modelos Genéticos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oligonucleotídeos/metabolismo , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina , Fatores de Tempo , Trans-Splicing/fisiologia
20.
Mol Biochem Parasitol ; 102(1): 103-15, 1999 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-10477180

RESUMO

The protozoan parasite Trypanosoma brucei relies on trans-splicing to process its mRNAs. A novel nuclear serine/arginine (SR)-rich trypanosomal protein (TSR1) was characterized which contains two RNA recognition motifs. The TSR1 protein appears to be homologous to RNA-binding SR proteins of the cis-splicing machinery from higher eukaryotes. Moreover, in the yeast two-hybrid system, TSR1 is able to interact with the human splicing factors involved in the recognition of the 3' splicing site (U2AF35/U2AF65). In both procyclic and bloodstream forms of T. brucei, TSR1 was found to localize in the nucleus. In the bloodstream stage TSR1 showed the speckles pattern characteristic of SR proteins involved in cis-splicing. Moreover, TSR1 was able to specifically bind the spliced leader (SL) RNA involved in trans-splicing in trypanosomes by the yeast three-hybrid system. These and other observations suggest that TSR1 may be involved in trans-splicing in T. brucei.


Assuntos
Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Trans-Splicing/genética , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA de Protozoário/análise , Imunofluorescência , Proteínas Fúngicas/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA de Protozoário/metabolismo , RNA Líder para Processamento/genética , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...