Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
RNA ; 28(3): 303-319, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34893560

RESUMO

Spinal muscular atrophy (SMA) is a devastating neurodegenerative disease caused by reduced amounts of the ubiquitously expressed Survival of Motor Neuron (SMN) protein. In agreement with its crucial role in the biogenesis of spliceosomal snRNPs, SMN-deficiency is correlated to numerous splicing alterations in patient cells and various tissues of SMA mouse models. Among the snRNPs whose assembly is impacted by SMN-deficiency, those involved in the minor spliceosome are particularly affected. Importantly, splicing of several, but not all U12-dependent introns has been shown to be affected in different SMA models. Here, we have investigated the molecular determinants of this differential splicing in spinal cords from SMA mice. We show that the branchpoint sequence (BPS) is a key element controlling splicing efficiency of minor introns. Unexpectedly, splicing of several minor introns with suboptimal BPS is not affected in SMA mice. Using in vitro splicing experiments and oligonucleotides targeting minor or major snRNAs, we show for the first time that splicing of these introns involves both the minor and major machineries. Our results strongly suggest that splicing of a subset of minor introns is not affected in SMA mice because components of the major spliceosome compensate for the loss of minor splicing activity.


Assuntos
Atrofia Muscular Espinal/genética , Splicing de RNA , Spliceossomos/metabolismo , Animais , Células HeLa , Humanos , Íntrons , Camundongos , Atrofia Muscular Espinal/metabolismo , Sítios de Splice de RNA , Ribonucleoproteínas Nucleares Pequenas/metabolismo
2.
iScience ; 23(1): 100809, 2020 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-31927482

RESUMO

Spinal muscular atrophy (SMA) is a devastating motor neuron disorder caused by mutations in the survival motor neuron (SMN) gene. It remains unclear how SMN deficiency leads to the loss of motor neurons. By screening Schizosaccharomyces pombe, we found that the growth defect of an SMN mutant can be alleviated by deletion of the actin-capping protein subunit gene acp1+. We show that SMN mutated cells have splicing defects in the profilin gene, which thus directly hinder actin cytoskeleton homeostasis including endocytosis and cytokinesis. We conclude that deletion of acp1+ in an SMN mutant background compensates for actin cytoskeleton alterations by restoring redistribution of actin monomers between different types of cellular actin networks. Our data reveal a direct correlation between an impaired function of SMN in snRNP assembly and defects in actin dynamics. They also point to important common features in the pathogenic mechanism of SMA and ALS.

3.
RNA ; 23(6): 899-909, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28258160

RESUMO

Spinal muscular atrophy (SMA) is caused by mutations and/or deletions of the survival motor neuron gene (SMN1). Besides its function in the biogenesis of spliceosomal snRNPs, SMN might possess a motor neuron specific role and could function in the transport of axonal mRNAs and in the modulation of local protein translation. Accordingly, SMN colocalizes with axonal mRNAs of differentiated NSC-34 motor neuron-like cells. We recently showed that SMN depletion gives rise to a decrease in the axonal transport of the mRNAs encoding Annexin A2 (Anxa2). In this work, we have characterized the structural features of the Anxa2 mRNA required for its axonal targeting by SMN. We found that a G-rich motif located near the 3'UTR is essential for axonal localization of the Anxa2 transcript. We also show that mutations in the motif sequence abolish targeting of Anxa2 reporter mRNAs in axon-like structures of differentiated NSC-34 cells. Finally, localization of both wild-type and mutated Anxa2 reporters is restricted to the cell body in SMN-depleted cells. Altogether, our studies show that this G-motif represents a novel and essential determinant for axonal localization of the Anxa2 mRNA mediated by the SMN complex.


Assuntos
Anexina A2/genética , Anexina A2/metabolismo , Axônios/metabolismo , Motivos de Nucleotídeos , RNA Mensageiro , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Regiões 3' não Traduzidas , Animais , Anexina A2/química , Sequência de Bases , Linhagem Celular , Quadruplex G , Expressão Gênica , Genes Reporter , Humanos , Camundongos , Neurônios Motores/metabolismo , Ligação Proteica , Transporte Proteico , Transporte de RNA
4.
Mol Cell Biol ; 34(4): 595-605, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24298023

RESUMO

During the early steps of snRNP biogenesis, the survival motor neuron (SMN) complex acts together with the methylosome, an entity formed by the pICln protein, WD45, and the PRMT5 methyltransferase. To expand our understanding of the functional relationship between pICln and SMN in vivo, we performed a genetic analysis of an uncharacterized Schizosaccharomyces pombe pICln homolog. Although not essential, the S. pombe ICln (SpICln) protein is important for optimal yeast cell growth. The human ICLN gene complements the Δicln slow-growth phenotype, demonstrating that the identified SpICln sequence is the bona fide human homolog. Consistent with the role of human pICln inferred from in vitro experiments, we found that the SpICln protein is required for optimal production of the spliceosomal snRNPs and for efficient splicing in vivo. Genetic interaction approaches further demonstrate that modulation of ICln activity is unable to compensate for growth defects of SMN-deficient cells. Using a genome-wide approach and reverse transcription (RT)-PCR validation tests, we also show that splicing is differentially altered in Δicln cells. Our data are consistent with the notion that splice site selection and spliceosome kinetics are highly dependent on the concentration of core spliceosomal components.


Assuntos
Canais Iônicos/genética , Neurônios Motores/metabolismo , Splicing de RNA/genética , Schizosaccharomyces/genética , Spliceossomos/genética , Citoplasma/genética , Citoplasma/metabolismo , Humanos , Canais Iônicos/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Ligação Proteica/genética , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Schizosaccharomyces/metabolismo , Spliceossomos/metabolismo
5.
RNA ; 19(12): 1755-66, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24152552

RESUMO

Spinal muscular atrophy is a neuromuscular disease resulting from mutations in the SMN1 gene, which encodes the survival motor neuron (SMN) protein. SMN is part of a large complex that is essential for the biogenesis of spliceosomal small nuclear RNPs. SMN also colocalizes with mRNAs in granules that are actively transported in neuronal processes, supporting the hypothesis that SMN is involved in axonal trafficking of mRNPs. Here, we have performed a genome-wide analysis of RNAs present in complexes containing the SMN protein and identified more than 200 mRNAs associated with SMN in differentiated NSC-34 motor neuron-like cells. Remarkably, ~30% are described to localize in axons of different neuron types. In situ hybridization and immuno-fluorescence experiments performed on several candidates indicate that these mRNAs colocalize with the SMN protein in neurites and axons of differentiated NSC-34 cells. Moreover, they localize in cell processes in an SMN-dependent manner. Thus, low SMN levels might result in localization deficiencies of mRNAs required for axonogenesis.


Assuntos
Neuritos/metabolismo , RNA Mensageiro/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Animais , Anexina A2/genética , Anexina A2/metabolismo , Linhagem Celular , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Técnicas de Silenciamento de Genes , Genoma , Camundongos , Neurônios Motores/metabolismo , Junção Neuromuscular/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Transporte de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Selenoproteína W/genética , Selenoproteína W/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/genética
6.
Nucleic Acids Res ; 41(2): 1255-72, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23221635

RESUMO

Spinal muscular atrophy is a severe motor neuron disease caused by reduced levels of the ubiquitous Survival of MotoNeurons (SMN) protein. SMN is part of a complex that is essential for spliceosomal UsnRNP biogenesis. Signal recognition particle (SRP) is a ribonucleoprotein particle crucial for co-translational targeting of secretory and membrane proteins to the endoplasmic reticulum. SRP biogenesis is a nucleo-cytoplasmic multistep process in which the protein components, except SRP54, assemble with 7S RNA in the nucleolus. Then, SRP54 is incorporated after export of the pre-particle into the cytoplasm. The assembly factors necessary for SRP biogenesis remain to be identified. Here, we show that 7S RNA binds to purified SMN complexes in vitro and that SMN complexes associate with SRP in cellular extracts. We identified the RNA determinants required. Moreover, we report a specific reduction of 7S RNA levels in the spinal cord of SMN-deficient mice, and in a Schizosaccharomyces pombe strain carrying a temperature-degron allele of SMN. Additionally, microinjected antibodies directed against SMN or Gemin2 interfere with the association of SRP54 with 7S RNA in Xenopus laevis oocytes. Our data show that reduced levels of the SMN protein lead to defect in SRP steady-state level and describe the SMN complex as the first identified cellular factor required for SRP biogenesis.


Assuntos
RNA Citoplasmático Pequeno/metabolismo , Proteínas do Complexo SMN/metabolismo , Partícula de Reconhecimento de Sinal/metabolismo , Alelos , Animais , Anticorpos/farmacologia , Sequência de Bases , Citoplasma/metabolismo , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Atrofia Muscular Espinal/metabolismo , Mutação , RNA Citoplasmático Pequeno/química , RNA Nuclear Pequeno/metabolismo , Proteínas do Complexo SMN/antagonistas & inibidores , Proteínas do Complexo SMN/imunologia , Schizosaccharomyces/genética , Partícula de Reconhecimento de Sinal/química , Medula Espinal/metabolismo , Xenopus laevis
7.
Hum Mol Genet ; 20(4): 641-8, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21098506

RESUMO

The survival of motor neuron (SMN) protein is essential for cytoplasmic assembly of spliceosomal snRNPs. Although the normal proportion of endogenous snRNAs is unevenly altered in spinal muscular atrophy (SMA) tissues, the biogenesis of individual snRNPs is not dramatically affected in SMN-deficient cells. The SMN protein is also required for normal Cajal body (CB) formation, but the functional consequences of CB disruption upon SMN deficiency have not yet been analyzed at the level of macromolecular snRNPs assembly. Here, we show that the SMN protein is required for tri-snRNPs formation and that the level of the minor U4atac/U6atac/U5 tri-snRNPs is dramatically decreased in lymphoblasts derived from a patient suffering from a severe form of SMA. We found also that splicing of some, but not all, minor introns is inhibited in these cells, demonstrating links between SMN deficiency and differential alterations of splicing events mediated by the minor spliceosome. Our results suggest that SMA might result from the inefficient splicing of one or only a few pre-mRNAs carrying minor introns and coding for proteins required for motor neurons function and/or organization.


Assuntos
Íntrons/genética , Linfócitos/patologia , Splicing de RNA/genética , RNA Nuclear Pequeno/genética , Ribonucleoproteínas Nucleares Pequenas , Atrofias Musculares Espinais da Infância/patologia , Spliceossomos/patologia , Sobrevivência Celular/genética , Corpos Enovelados/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Ribonucleoproteínas Nucleares Pequenas/genética , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Atrofias Musculares Espinais da Infância/genética
8.
EMBO J ; 29(11): 1817-29, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20400941

RESUMO

Spinal muscular atrophy results from deletions or mutations in the survival of motor neuron (SMN1) gene. The SMN protein has an essential role in the biogenesis of spliceosomal snRNPs, but the link between a defect in this process and specific splicing inhibition of pre-mRNAs has not been established. In this study, we report the construction of a temperature-degron (td) allele of the Schizosaccharomyces pombe SMN protein and show that its depletion at 37 degrees C affects splicing and formation of U1, U2, U4 and U5 snRNPs, but not of U6 and U3 ribonucleoproteins. The function of the tdSMN allele in snRNP assembly is already perturbed at 25 degrees C, suggesting a deleterious effect of the tag at this temperature. Using a genome-wide approach, we report that introns react unequally to lower levels of snRNPs in tdSMN cells and that increasing the length of the polypyrimidine tract can improve the splicing efficiency of some, but not all, affected introns. Altogether, our results suggest that the defects observed in tdSMN fission yeast cells mimic splicing deficits observed in SMN-deficient metazoan cells.


Assuntos
Genes Fúngicos , Precursores de RNA/metabolismo , Splicing de RNA , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/genética , Spliceossomos/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Alelos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Humanos , Íntrons , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Mutação , Precursores de RNA/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas Nucleares Pequenas/genética , Spliceossomos/genética , Proteínas Centrais de snRNP
9.
Hum Mutat ; 29(12): 1412-24, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18561205

RESUMO

Numerous unclassified variants (UVs) have been found in the mismatch repair genes MLH1 and MSH2 involved in hereditary nonpolyposis colorectal cancer (HNPCC or Lynch syndrome). Some of these variants may have an effect on pre-mRNA splicing, either by altering degenerate positions of splice site sequences or by affecting intronic or exonic splicing regulatory sequences such as exonic splicing enhancers (ESEs). In order to determine the consequences of UVs on splicing, we used a functional assay of exon inclusion. For each variant, mutant and wild-type exons to be tested were PCR-amplified from patient genomic DNA together with approximately 150 bp of flanking sequences and were inserted into a splicing reporter minigene. After transfection into HeLa cells, the effects on splicing were evaluated by RT-PCR analysis and systematic sequencing. A total of 22 UVs out of 85 different variant alleles examined in 82 families affected splicing, including four exonic variants that affected putative splicing regulatory elements. We analyzed short stretches spanning the latter variants by cloning them into the ESE-dependent central exon of a three-exon splicing minigene and we showed in cell transfection experiments that the wild-type sequences indeed contain functional ESEs. We then used this construct to query for ESE elements in the MLH1 or MSH2 regions affected by 14 previously reported exonic splicing mutations and showed that they also contain functional ESEs. These splicing assays represent a valuable tool for the interpretation of UVs and should contribute to the optimization of the molecular diagnosis of the Lynch syndrome and of other genetic diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Proteína 2 Homóloga a MutS/genética , Mutação , Proteínas Nucleares/genética , Splicing de RNA , Família , Humanos , Proteína 1 Homóloga a MutL
10.
J Biol Chem ; 283(4): 2060-9, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18039666

RESUMO

Tgs1 is the hypermethylase responsible for m(3)G cap formation of U small nuclear RNAs (U snRNAs) and small nucleolar RNAs (snoRNAs). In vertebrates, hypermethylation of snRNAs occurs in the cytoplasm, whereas this process takes place in the nucleus for snoRNAs. Accordingly, the hypermethylase is found in both compartments with a diffuse localization in the cytoplasm and a concentration in Cajal bodies in the nucleoplasm. In this study, we report that the Tgs1 hypermethylase exists as two species, a full-length cytoplasmic isoform and a shorter nuclear isoform of 65-70 kDa. The short isoform exhibits methyltransferase activity and associates with components of box C/D and H/ACA snoRNPs, pointing to a role of this isoform in hypermethylation of snoRNAs. We also show that production of the short Tgs1 isoform is inhibited by MG132, suggesting that it results from proteasomal limited processing of the full-length Tgs1 protein. Together, our results suggest that proteasome maturation constitutes a mechanism regulating Tgs1 function by generating Tgs1 species with different substrate specificities, subcellular localizations, and functions.


Assuntos
Núcleo Celular/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Capuzes de RNA/metabolismo , Processamento Pós-Transcricional do RNA/fisiologia , RNA Nuclear Pequeno/metabolismo , tRNA Metiltransferases/metabolismo , Antineoplásicos/farmacologia , Núcleo Celular/genética , Citoplasma/enzimologia , Citoplasma/genética , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Leupeptinas/farmacologia , Metilação/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/genética , Capuzes de RNA/genética , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Nuclear Pequeno/genética , tRNA Metiltransferases/genética
11.
PLoS One ; 2(2): e253, 2007 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-17327915

RESUMO

The genetic programs specifying eye development are highly conserved during evolution and involve the vertebrate Pax-6 gene and its Drosophila melanogaster homolog eyeless (ey). Here we report that the SR protein B52/SRp55 controls a novel developmentally regulated splicing event of eyeless that is crucial for eye growth and specification in Drosophila. B52/SRp55 generates two isoforms of eyeless differing by an alternative exon encoding a 60-amino-acid insert at the beginning of the paired domain. The long isoform has impaired ability to trigger formation of ectopic eyes and to bind efficiently Eyeless target DNA sequences in vitro. When over-produced in the eye imaginal disc, this isoform induces a small eye phenotype, whereas the isoform lacking the alternative exon triggers eye over-growth and strong disorganization. Our results suggest that B52/SRp55 splicing activity is used during normal eye development to control eye organogenesis and size through regulation of eyeless alternative splicing.


Assuntos
Processamento Alternativo , Olho Composto de Artrópodes/crescimento & desenvolvimento , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Nucleares/fisiologia , Fosfoproteínas/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Olho Composto de Artrópodes/anormalidades , Sequência Consenso , Proteínas de Ligação a DNA/fisiologia , Drosophila/genética , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/genética , Éxons/genética , Dados de Sequência Molecular , Organogênese/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Estrutura Terciária de Proteína , Precursores de RNA/genética , Precursores de RNA/metabolismo , Fatores de Processamento de RNA , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
12.
Mol Cell Biol ; 27(8): 3087-97, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17283056

RESUMO

Deciphering the role of alternative splicing in developmental processes relies on the identification of key genes whose expression is controlled by splicing regulators throughout the growth of a whole organism. Modulating the expression levels of five SR proteins in the developing eye of Drosophila melanogaster revealed that these splicing factors induce various phenotypic alterations in eye organogenesis and also affect viability. Although the SR proteins dASF/SF2 and B52 caused defects in ommatidia structure, only B52 impaired normal axonal projections of photoreceptors and neurogenesis in visual ganglia. Microarray analyses revealed that many transcripts involved in brain organogenesis have altered splicing profiles upon both loss and gain of B52 function. Conversely, a large proportion of transcripts regulated by dASF/SF2 are involved in eye development. These differential and specific effects of SR proteins indicate that they function to confer accuracy to developmental gene expression programs by facilitating the cell lineage decisions that underline the generation of tissue identities.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Olho/crescimento & desenvolvimento , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Animais Geneticamente Modificados , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Sobrevivência Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Olho/citologia , Olho/metabolismo , Feminino , Genes Controladores do Desenvolvimento , Genes de Insetos , Imunoprecipitação , Larva/citologia , Masculino , Morfogênese , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Células Fotorreceptoras de Invertebrados/citologia , Células Fotorreceptoras de Invertebrados/metabolismo , Splicing de RNA/genética , Fatores de Processamento de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Prog Mol Subcell Biol ; 44: 65-87, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17076265

RESUMO

Serine- and arginine-rich (SR) proteins constitute a highly conserved family of pre-mRNA splicing factors that play key roles in the regulation of splice site selection, and thereby in the control of alternative splicing processes. In addition to conserved sequences at the splice junctions, splice site selection also depends upon different sets of auxiliary cis regulatory elements known as exonic and intronic splicing enhancers (ESEs and ISEs) or exonic and intronic silencers (ESSs and ISSs). Specific binding of SR proteins to their cognate splicing enhancers as well as binding of splicing repressor to silencer sequences serve to enhance or inhibit recognition of weak splice sites by the splicing machinery. Given that the vast majority of human genes contain introns and that most pre-mRNAs containing multiple exons undergo alternative splicing, mutations disrupting or creating such auxiliary elements can result in aberrant splicing events at the origin of various human diseases. In the past few years, numerous studies have reported several approaches allowing correction of such aberrant splicing events by targeting either the mutated sequences or the splicing regulators whose binding is affected by the mutation. The aim of the present review is to highlight the different means by which it is possible to modulate the activity of SR splicing factors and to bring out those holding the greatest promises for the development of therapeutic treatments.


Assuntos
Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Terapêutica/métodos , Aclarubicina/farmacologia , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Inibidores Enzimáticos/farmacologia , Humanos , Proteínas Nucleares/genética , Fosforilação/efeitos dos fármacos , Proteínas de Ligação a RNA/genética , Fatores de Processamento de Serina-Arginina
14.
Proc Natl Acad Sci U S A ; 102(24): 8764-9, 2005 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-15939885

RESUMO

The prevalence of alternative splicing as a target for alterations leading to human genetic disorders makes it highly relevant for therapy. Here we have used in vitro splicing reactions with different splicing reporter constructs to screen 4,000 chemical compounds for their ability to selectively inhibit spliceosome assembly and splicing. We discovered indole derivatives as potent inhibitors of the splicing reaction. Importantly, compounds of this family specifically inhibit exonic splicing enhancer (ESE)-dependent splicing, because they interact directly and selectively with members of the serine-arginine-rich protein family. Treatment of cells expressing reporter constructs with ESE sequences demonstrated that selected indole derivatives mediate inhibition of ESE usage in vivo and prevent early splicing events required for HIV replication. This discovery opens the exciting possibility of a causal pharmacological treatment of aberrant splicing in human genetic disorders and development of new antiviral therapeutic approaches.


Assuntos
Processamento Alternativo/genética , HIV-1/metabolismo , Indóis/metabolismo , Proteínas Nucleares/metabolismo , Splicing de RNA/fisiologia , Ribonucleoproteínas/metabolismo , Spliceossomos/metabolismo , Processamento Alternativo/efeitos dos fármacos , Primers do DNA , Vetores Genéticos/genética , HIV-1/efeitos dos fármacos , Células HeLa , Humanos , Indóis/farmacologia , Splicing de RNA/efeitos dos fármacos , Fatores de Processamento de Serina-Arginina , Espectrometria de Fluorescência , Spliceossomos/genética
15.
Mol Cell Biol ; 25(8): 3286-94, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15798212

RESUMO

Pyruvate dehydrogenase (PDH) complex deficiency is a major cause of lactic acidosis and Leigh's encephalomyelopathies in infancy and childhood, resulting in early death in the majority of patients. Most of the molecular defects have been localized in the coding regions of the E1alpha PDH gene. Recently, we identified a novel mutation of the E1alpha PDH gene in a patient with an encephalopathy and lactic acidosis. This mutation, located downstream of exon 7, activates a cryptic splice donor and leads to the retention of intronic sequences. Here, we demonstrate that the mutation results in an increased binding of the SR protein SC35. Consistently, ectopic overexpression of this splicing factor enhanced the use of the cryptic splice site, whereas small interfering RNA-mediated reduction of the SC35 protein levels in primary fibroblasts from the patient resulted in the almost complete disappearance of the aberrantly spliced E1alpha PDH mRNA. Our findings open the exciting prospect for a novel therapy of an inherited disease by altering the level of a specific splicing factor.


Assuntos
Acidose Láctica/genética , Processamento Alternativo/fisiologia , Doença de Leigh/genética , Proteínas Nucleares/metabolismo , Piruvato Desidrogenase (Lipoamida)/genética , Sítios de Splice de RNA/fisiologia , Ribonucleoproteínas/metabolismo , Acidose Láctica/metabolismo , Processamento Alternativo/genética , Células Cultivadas , Éxons/genética , Fibroblastos/metabolismo , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Íntrons/genética , Doença de Leigh/metabolismo , Mutação/genética , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Complexo Piruvato Desidrogenase/genética , Doença da Deficiência do Complexo de Piruvato Desidrogenase/genética , Doença da Deficiência do Complexo de Piruvato Desidrogenase/metabolismo , Interferência de RNA , Sítios de Splice de RNA/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/antagonistas & inibidores , Ribonucleoproteínas/genética , Fatores de Processamento de Serina-Arginina , Síndrome
16.
Mol Pharmacol ; 67(4): 1186-94, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15625279

RESUMO

Pre-mRNA splicing is an essential step of the expression of most metazoan protein-coding genes, which is often regulated in a cell type-specific or developmental manner. We have demonstrated previously that human DNA topoisomerase I, an extensively studied target for anticancer drugs, also has an intrinsic protein kinase activity that specifically phosphorylates proteins involved in splice site selection. Therefore, DNA topoisomerase I was recently shown to play a critical role in alternative splicing. Here, we have exploited these novel properties of DNA topoisomerase I to develop entirely novel diospyrin derivatives targeting its protein kinase activity and thereby modulating pre-mRNA splicing. Although some derivatives indeed inhibit kinase activity of topoisomerase I, they did not block reactions of topoisomerase I on DNA. However, these drugs interfere with camptothecin-dependent topoisomerase I-mediated DNA cleavage, implying that diospyrin derivatives mediate a conformational change of topoisomerase I. It is note-worthy that in vitro splicing reactions revealed that diospyrin derivatives alter various steps of splicing. Some diospyrin derivatives inhibit either the first or the second catalytic step of splicing but not spliceosome assembly, whereas diospyrin itself prevents the formation of full spliceosome. Our data revealed for the first time that diospyrin derivatives are able to stall the dynamic assembly of the spliceosome and open the exciting possibility of using these derivatives to correct aberrant splicing in human genetic diseases.


Assuntos
Inibidores Enzimáticos/farmacologia , Naftoquinonas/farmacologia , Spliceossomos/efeitos dos fármacos , Inibidores da Topoisomerase I , DNA/metabolismo , Fosforilação , Splicing de RNA/efeitos dos fármacos , Relação Estrutura-Atividade
17.
J Cell Sci ; 117(Pt 19): 4551-8, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15331664

RESUMO

Exposure of cells to stressful conditions results in the rapid synthesis of a subset of specialized proteins termed heat shock proteins (HSPs) which function in protecting the cell against damage. The stress-induced activation of hsp genes is controlled by the heat shock transcription factor 1 (HSF1). At the cellular level, one of the most striking effects of stress is the rapid and reversible redistribution of HSF1 into a few nuclear structures termed nuclear stress granules which form primarily on the 9q12 locus in humans. Within these structures, HSF1 binds to satellite III repeated elements and drives the RNA polymerase II-dependent transcription of these sequences into stable RNAs which remain associated with the 9q12 locus for a certain time after synthesis. Other proteins, in particular splicing factors, were also shown to relocalize to the granules upon stress. Here, we investigated the role of stress-induced satellite III transcripts in the relocalization of splicing factors to the granules. We show that the recruitment of the two serine/arginine-rich (SR) proteins SF2/ASF and SRp30c requires the presence of stress-induced satellite III transcripts. In agreement with these findings, we identified the second RNA-recognition motif (RRM2) of hSF2/ASF as the motif required for the targeting to the granules, and we showed by immunoprecipitation that the endogenous hSF2/ASF protein is present in a complex with satellite III transcripts in stressed cells in vivo. Interestingly, satellite III transcripts also immunoprecipitate together with small nuclear ribonucleoproteins (snRNPs) in vivo whereas the intronless hsp70 transcripts do not, supporting the proposal that these transcripts are subject to splicing. Altogether, these data highlight the central role for satellite III transcripts in the targeting and/or retention of splicing factors into the granules upon stress.


Assuntos
Estruturas do Núcleo Celular/metabolismo , DNA Satélite/metabolismo , Proteínas de Ligação a DNA/metabolismo , Splicing de RNA/fisiologia , Estruturas do Núcleo Celular/genética , DNA Satélite/genética , Proteínas de Ligação a DNA/genética , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Células HeLa , Fatores de Transcrição de Choque Térmico , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Splicing de RNA/genética , Proteínas de Ligação a RNA , Ribonucleoproteínas Nucleares Pequenas/genética , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Fatores de Processamento de Serina-Arginina , Fatores de Transcrição
18.
Mol Cancer Res ; 2(1): 53-61, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14757846

RESUMO

We have recently shown that the topoisomerase II inhibitor, etoposide (VP16), could trigger caspase-2 pre-mRNA splicing in human leukemic cell lines. This leads to increased inclusion of exon 9, which is specifically inserted into the short caspase-2S isoform mRNA and absent from the long caspase-2L isoform mRNA. One of the consequences of this alternative splicing is a decrease in the total amount of the mature form of caspase-2L mRNA and protein. In this study, we analyzed the effects of several representative molecules of various classes of cytotoxic agents on caspase-2 pre-mRNA splicing in both U937 leukemic cells and in HeLa cervix carcinoma cells. Very strikingly, both topoisomerase I (camptothecin and homocamptothecin derivatives) and II (VP16, amsacrine, doxorubicin, mitoxantrone) inhibitors induced exon 9 inclusion. DNA intercalating glycosyl indolocarbazole derivatives as well as DNA alkylating agents, such as cisplatin and melphalan, antimetabolites like 5-fluorouracil, and mitotic spindle poisons like vinblastine had no effect. Therefore, both classes of DNA topoisomerases can control pre-mRNA splicing of the caspase-2 transcript. In addition, the splicing reaction brought about by camptothecin was hampered in human CEM/C2 and in murine P388-45R leukemic deficient in topoisomerase I activity. Conversely, VP16 did not trigger caspase-2 alternative splicing in human HL60/MX2 leukemic cells harboring a mutant topoisomerase II. Minigene transfection analysis revealed that topoisomerase inhibitors did not change the splicing profile when cis-acting elements in intron-9, reported to control exon 9 inclusion independently of drug treatment, were removed. Rather, our experiments suggest that exon 9 inclusion induced by topoisomerase inhibitors reflects the activity exerted by topoisomerase I or II on proteins that control splicing reactions, or their direct involvement in pre-mRNA splicing.


Assuntos
Processamento Alternativo/efeitos dos fármacos , Cisteína Endopeptidases/genética , Inibidores Enzimáticos/farmacologia , Splicing de RNA/efeitos dos fármacos , RNA Mensageiro/genética , Inibidores da Topoisomerase I , Inibidores da Topoisomerase II , Antibióticos Antineoplásicos/farmacologia , Apoptose/fisiologia , Western Blotting , Caspase 2 , Linhagem Celular Tumoral/metabolismo , Reagentes de Ligações Cruzadas , Vetores Genéticos , Células HL-60 , Humanos , Inibidores da Síntese de Ácido Nucleico , Plasmídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células U937
19.
Cancer Res ; 63(23): 8203-11, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14678976

RESUMO

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.


Assuntos
DNA Topoisomerases Tipo I/deficiência , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Splicing de RNA/fisiologia , Processamento Alternativo , Animais , Antineoplásicos/farmacologia , Camptotecina/farmacologia , DNA Topoisomerases Tipo I/genética , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Éxons/fisiologia , Regulação Leucêmica da Expressão Gênica , Proteínas de Fluorescência Verde , Leucemia P388/enzimologia , Leucemia P388/genética , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Fosforilação , Proteínas de Ligação a RNA , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Processamento de Serina-Arginina , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...