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










Base de dados
Intervalo de ano de publicação
1.
Nat Struct Mol Biol ; 30(10): 1536-1548, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37783853

RESUMO

Non-cleaving Cas9 (dCas9) is widely employed to manipulate specific gene loci, often with scant regard for unintended transcriptional effects. We demonstrate here that dCas9 mediates precise RNA polymerase II transcriptional pausing followed by transcription termination and potential alternative polyadenylation. By contrast, alternative splicing is unaffected, likely requiring more sustained alteration to elongation speed. The effect on transcription is orientation specific, with pausing only being induced when dCas9-associated guide RNA anneals to the non-template strand. Targeting the template strand induces minimal effects on transcription elongation and thus provides a neutral approach to recruit dCas9-linked effector domains to specific gene regions. In essence, we evaluate molecular effects of targeting dCas9 to mammalian transcription units. In so doing, we also provide new information on elongation by RNA polymerase II and coupled pre-mRNA processing.


Assuntos
RNA Polimerase II , Transcrição Gênica , Animais , Humanos , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Processamento Alternativo , Mamíferos/genética
2.
Cell ; 185(12): 2057-2070.e15, 2022 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-35688133

RESUMO

Spinal muscular atrophy (SMA) is a motor-neuron disease caused by mutations of the SMN1 gene. The human paralog SMN2, whose exon 7 (E7) is predominantly skipped, cannot compensate for the lack of SMN1. Nusinersen is an antisense oligonucleotide (ASO) that upregulates E7 inclusion and SMN protein levels by displacing the splicing repressors hnRNPA1/A2 from their target site in intron 7. We show that by promoting transcriptional elongation, the histone deacetylase inhibitor VPA cooperates with a nusinersen-like ASO to promote E7 inclusion. Surprisingly, the ASO promotes the deployment of the silencing histone mark H3K9me2 on the SMN2 gene, creating a roadblock to RNA polymerase II elongation that inhibits E7 inclusion. By removing the roadblock, VPA counteracts the chromatin effects of the ASO, resulting in higher E7 inclusion without large pleiotropic effects. Combined administration of the nusinersen-like ASO and VPA in SMA mice strongly synergizes SMN expression, growth, survival, and neuromuscular function.


Assuntos
Atrofia Muscular Espinal , Oligonucleotídeos Antissenso , Animais , Cromatina , Éxons , Camundongos , Atrofia Muscular Espinal/tratamento farmacológico , Atrofia Muscular Espinal/genética , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/uso terapêutico , Splicing de RNA
3.
Mol Cell ; 81(9): 1935-1950.e6, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33735606

RESUMO

Mammalian chromatin is the site of both RNA polymerase II (Pol II) transcription and coupled RNA processing. However, molecular details of such co-transcriptional mechanisms remain obscure, partly because of technical limitations in purifying authentic nascent transcripts. We present a new approach to characterize nascent RNA, called polymerase intact nascent transcript (POINT) technology. This three-pronged methodology maps nascent RNA 5' ends (POINT-5), establishes the kinetics of co-transcriptional splicing patterns (POINT-nano), and profiles whole transcription units (POINT-seq). In particular, we show by depletion of the nuclear exonuclease Xrn2 that this activity acts selectively on cleaved 5' P-RNA at polyadenylation sites. Furthermore, POINT-nano reveals that co-transcriptional splicing either occurs immediately after splice site transcription or is delayed until Pol II transcribes downstream sequences. Finally, we connect RNA cleavage and splicing with either premature or full-length transcript termination. We anticipate that POINT technology will afford full dissection of the complexity of co-transcriptional RNA processing.


Assuntos
Nanotecnologia , RNA Polimerase II/metabolismo , Precursores de RNA/biossíntese , Splicing de RNA , RNA Mensageiro/biossíntese , RNA-Seq , Transcrição Gênica , Exorribonucleases/genética , Exorribonucleases/metabolismo , Células HCT116 , Células HeLa , Humanos , Cinética , Poliadenilação , Capuzes de RNA , RNA Polimerase II/genética , Precursores de RNA/genética , RNA Mensageiro/genética
4.
Mol Biol Rep ; 47(4): 3031-3040, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32200451

RESUMO

We previously reported a 40-transcripts signature marking the normal mucosa to colorectal adenocarcinoma transition. Eight of these mRNAs also showed splicing alterations, including a specific intron 3 retention in tissue metalloprotease inhibitor I (TIMP1), which decreased during the early steps of colorectal cancer progression. To decipher the mechanism of intron 3 retention/splicing, we first searched for putative RNA binding protein binding sites onto the TIMP1 sequence. We identified potential serine arginine rich splicing factor 1 (SRSF1) and heterogeneous nuclear RiboNucleoProtein A1 (hnRNPA1) binding sites at the end of intron 3 and the beginning of exon 4, respectively. RNA immunoprecipitation showed that hnRNPA1, but not SRSF1 could bind to the corresponding region in TIMP1 pre-mRNA in live cells. Furthermore, using a TIMP1-based ex vivo minigene approach, together with a plasmon resonance in vitro RNA binding assay, we confirmed that hnRNPA1 could indeed bind to wild type TIMP1 exon 4 pre-mRNA and control TMP1 intron 3 splicing, the interaction being abolished in presence of a mutant sequence that disrupted this site. These results indicated that hnRNPA1, upon binding to TIMP1 exon 4, was a positive regulator of intron 3 splicing. We propose that this TIMP1-intron 3 + transcript belongs to the class of nuclear transcripts with "detained" introns, an abundant molecular class, including in cancer.


Assuntos
Neoplasias do Colo/genética , Ribonucleoproteína Nuclear Heterogênea A1/genética , Inibidor Tecidual de Metaloproteinase-1/genética , Processamento Alternativo , Sítios de Ligação/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias do Colo/metabolismo , Éxons , Células HCT116 , Ribonucleoproteína Nuclear Heterogênea A1/metabolismo , Humanos , Íntrons , Ligação Proteica/genética , Precursores de RNA/genética , Precursores de RNA/metabolismo , Splicing de RNA , Fatores de Processamento de Serina-Arginina/genética , Fatores de Processamento de Serina-Arginina/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo
5.
Mol Cell ; 72(6): 970-984.e7, 2018 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-30449723

RESUMO

Extensive tracts of the mammalian genome that lack protein-coding function are still transcribed into long noncoding RNA. While these lncRNAs are generally short lived, length restricted, and non-polyadenylated, how their expression is distinguished from protein-coding genes remains enigmatic. Surprisingly, depletion of the ubiquitous Pol-II-associated transcription elongation factor SPT6 promotes a redistribution of H3K36me3 histone marks from active protein coding to lncRNA genes, which correlates with increased lncRNA transcription. SPT6 knockdown also impairs the recruitment of the Integrator complex to chromatin, which results in a transcriptional termination defect for lncRNA genes. This leads to the formation of extended, polyadenylated lncRNAs that are both chromatin restricted and form increased levels of RNA:DNA hybrid (R-loops) that are associated with DNA damage. Additionally, these deregulated lncRNAs overlap with DNA replication origins leading to localized DNA replication stress and a cellular senescence phenotype. Overall, our results underline the importance of restricting lncRNA expression.


Assuntos
Proliferação de Células , Senescência Celular , Dano ao DNA , Replicação do DNA , DNA de Neoplasias/biossíntese , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias Uterinas/metabolismo , Animais , Montagem e Desmontagem da Cromatina , DNA Polimerase II/genética , DNA Polimerase II/metabolismo , DNA de Neoplasias/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Células HeLa , Histonas/metabolismo , Humanos , Metilação , Conformação de Ácido Nucleico , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo , Estabilidade de RNA , RNA Longo não Codificante/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , Fatores de Transcrição/genética , Transcrição Gênica , Neoplasias Uterinas/genética
6.
Cell Rep ; 18(12): 2868-2879, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28329680

RESUMO

We have previously found that UV irradiation promotes RNA polymerase II (RNAPII) hyperphosphorylation and subsequent changes in alternative splicing (AS). We show now that UV-induced DNA damage is not only necessary but sufficient to trigger the AS response and that photolyase-mediated removal of the most abundant class of pyrimidine dimers (PDs) abrogates the global response to UV. We demonstrate that, in keratinocytes, RNAPII is the target, but not a sensor, of the signaling cascade initiated by PDs. The UV effect is enhanced by inhibition of gap-filling DNA synthesis, the last step in the nucleotide excision repair pathway (NER), and reduced by the absence of XPE, the main NER sensor of PDs. The mechanism involves activation of the protein kinase ATR that mediates the UV-induced RNAPII hyperphosphorylation. Our results define the sequence UV-PDs-NER-ATR-RNAPII-AS as a pathway linking DNA damage repair to the control of both RNAPII phosphorylation and AS regulation.


Assuntos
Processamento Alternativo/genética , Reparo do DNA , Dímeros de Pirimidina/metabolismo , Raios Ultravioleta , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , DNA/metabolismo , Reparo do DNA/genética , Humanos , Queratinócitos/metabolismo , Queratinócitos/efeitos da radiação , Fosforilação/efeitos da radiação , RNA Polimerase II/metabolismo , Pele/citologia , Pele/efeitos da radiação , Transcrição Gênica/efeitos da radiação
7.
PLoS One ; 11(8): e0160428, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27479083

RESUMO

Electronic laboratory notebooks (ELNs) will probably replace paper laboratory notebooks (PLNs) in academic research due to their advantages in data recording, sharing and security. Despite several reports describing technical characteristics of ELNs and their advantages over PLNs, no study has directly tested ELN performance among researchers. In addition, the usage of tablet-based devices or wearable technology as ELN complements has never been explored in the field. To implement an ELN in our biomedical research institute, here we first present a technical comparison of six ELNs using 42 parameters. Based on this, we chose two ELNs, which were tested by 28 scientists for a 3-month period and by 80 students via hands-on practical exercises. Second, we provide two survey-based studies aimed to compare these two ELNs (PerkinElmer Elements and Microsoft OneNote) and to analyze the use of tablet-based devices. We finally explore the advantages of using wearable technology as ELNs tools. Among the ELNs tested, we found that OneNote presents almost all parameters evaluated (39/42) and both surveyed groups preferred OneNote as an ELN solution. In addition, 80% of the surveyed scientists reported that tablet-based devices improved the use of ELNs in different respects. We also describe the advantages of using OneNote application for Apple Watch as an ELN wearable complement. This work defines essential features of ELNs that could be used to improve ELN implementation and software development.


Assuntos
Registros , Software , Academias e Institutos , Pesquisa Biomédica
8.
J Mol Biol ; 428(12): 2636-2651, 2016 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-26979557

RESUMO

Multicellular organisms must ensure genome integrity to prevent accumulation of mutations, cell death, and cancer. The DNA damage response (DDR) is a complex network that senses, signals, and executes multiple programs including DNA repair, cell cycle arrest, senescence, and apoptosis. This entails regulation of a variety of cellular processes: DNA replication and transcription, RNA processing, mRNA translation and turnover, and post-translational modification, degradation, and relocalization of proteins. Accumulated evidence over the past decades has shown that RNAs and RNA metabolism are both regulators and regulated actors of the DDR. This review aims to present a comprehensive overview of the current knowledge on the many interactions between the DNA damage and RNA fields.


Assuntos
Dano ao DNA/genética , RNA/genética , Reparo do DNA/genética , Expressão Gênica/genética , Humanos
9.
Med Sci (Paris) ; 32(12): 1103-1110, 2016 Dec.
Artigo em Francês | MEDLINE | ID: mdl-28044974

RESUMO

Pre-mRNA splicing is an obligatory step required to assemble the vast majority of mRNAs in eukaryotes. In humans, each gene gives rise to at least two transcripts, with an average 6-8 spliced transcripts per gene. Pre-mRNA splicing is not unequivocal. Variations may occur, such that splicing can become alternative, thereby participating in increasing protein variability and restricting the gap that exists between the relatively low number of genes - between 20,000 and 25,000 in humans - and the much higher number of distinct proteins - at least 100,000. In addition, although alternative pre-mRNA splicing often fulfils cell-specific needs, many aberrant splicing events can happen and lead to either hereditary or acquired diseases such as neurodegenerative diseases or cancers. In those cases, alternative splicing events may serve as disease-associated markers, or even as targets for corrective approaches. In this review, we will summarize the main aspects of regulated alternative splicing. We will present the spliceosome, a large ribonucleoprotein complex that orchestrates the splicing reactions and that was recently identified as a preferential target for mutations in several pathologies. We shall discuss some spliceosome-associated defects linked to either cis (i.e on the DNA) or trans (e.g. in proteins) alterations of splicing machinery, like those that have been reported in genetic or acquired diseases.


Assuntos
Precursores de RNA/metabolismo , Splicing de RNA/fisiologia , Spliceossomos/fisiologia , Processamento Alternativo/genética , Animais , Progressão da Doença , Humanos , Mutação/fisiologia , Neoplasias/genética , Neoplasias/patologia , RNA Mensageiro/metabolismo , Spliceossomos/genética
10.
EMBO Rep ; 16(12): 1640-55, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26566663

RESUMO

Removal of introns from pre-mRNA precursors (pre-mRNA splicing) is a necessary step for the expression of most genes in multicellular organisms, and alternative patterns of intron removal diversify and regulate the output of genomic information. Mutation or natural variation in pre-mRNA sequences, as well as in spliceosomal components and regulatory factors, has been implicated in the etiology and progression of numerous pathologies. These range from monogenic to multifactorial genetic diseases, including metabolic syndromes, muscular dystrophies, neurodegenerative and cardiovascular diseases, and cancer. Understanding the molecular mechanisms associated with splicing-related pathologies can provide key insights into the normal function and physiological context of the complex splicing machinery and establish sound basis for novel therapeutic approaches.


Assuntos
Processamento Alternativo , Doença/genética , Splicing de RNA , RNA Mensageiro/genética , Animais , Éxons , Humanos , Íntrons , Distrofias Musculares/genética , Distrofias Musculares/terapia , Mutação , Neoplasias/etiologia , Neoplasias/genética , Neoplasias/terapia , Precursores de RNA/genética , Spliceossomos/genética , Spliceossomos/metabolismo
12.
Proc Natl Acad Sci U S A ; 111(44): 15622-9, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25313066

RESUMO

The roles of Argonaute proteins in cytoplasmic microRNA and RNAi pathways are well established. However, their implication in small RNA-mediated transcriptional gene silencing in the mammalian cell nucleus is less understood. We have recently shown that intronic siRNAs cause chromatin modifications that inhibit RNA polymerase II elongation and modulate alternative splicing in an Argonaute-1 (AGO1)-dependent manner. Here we used chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) to investigate the genome-wide distribution of AGO1 nuclear targets. Unexpectedly, we found that about 80% of AGO1 clusters are associated with cell-type-specific transcriptional enhancers, most of them (73%) overlapping active enhancers. This association seems to be mediated by long, rather than short, enhancer RNAs and to be more prominent in intragenic, rather than intergenic, enhancers. Paradoxically, crossing ChIP-seq with RNA-seq data upon AGO1 depletion revealed that enhancer-bound AGO1 is not linked to the global regulation of gene transcription but to the control of constitutive and alternative splicing, which was confirmed by an individual gene analysis explaining how AGO1 controls inclusion levels of the cassette exon 107 in the SYNE2 gene.


Assuntos
Processamento Alternativo/fisiologia , Proteínas Argonautas/metabolismo , Elementos Facilitadores Genéticos/fisiologia , Fatores de Iniciação em Eucariotos/metabolismo , Regulação da Expressão Gênica/fisiologia , RNA/metabolismo , Transcrição Gênica/fisiologia , Proteínas Argonautas/genética , Linhagem Celular , Fatores de Iniciação em Eucariotos/genética , Humanos , RNA/genética , Análise de Sequência de RNA
13.
J Biol Chem ; 289(46): 31792-31804, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25239623

RESUMO

Posttranscriptional gene regulation is governed by a network of RNA-binding proteins (RBPs) that interact with regulatory elements in the mRNA to modulate multiple molecular processes, including splicing, RNA transport, RNA stability, and translation. Mounting evidence indicates that there is a hierarchy within this network whereby certain RBPs cross-regulate other RBPs to coordinate gene expression. HuR, an RNA-binding protein we linked previously to aberrant VEGF mRNA metabolism in models of SOD1-associated amyotrophic lateral sclerosis, has been identified as being high up in this hierarchy, serving as a regulator of RNA regulators. Here we investigated the role of HuR in regulating two RBPs, TDP-43 and FUS/TLS, that have been linked genetically to amyotrophic lateral sclerosis. We found that HuR promotes the expression of both RBPs in primary astrocytes and U251 cells under normal and stressed (hypoxic) conditions. For TDP-43, we found that HuR binds to the 3' untranslated region (UTR) and regulates its expression through translational efficiency rather than RNA stability. With HuR knockdown, there was a shift of TDP-43 and FUS mRNAs away from polysomes, consistent with translational silencing. The TDP-43 splicing function was attenuated upon HuR knockdown and could be rescued by ectopic TDP-43 lacking the 3' UTR regulatory elements. Finally, conditioned medium from astrocytes in which HuR or TDP-43 was knocked down produced significant motor neuron and cortical neuron toxicity in vitro. These findings indicate that HuR regulates TDP-43 and FUS/TLS expression and that loss of HuR-mediated RNA processing in astrocytes can alter the molecular and cellular landscape to produce a toxic phenotype.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas ELAV/metabolismo , Regulação da Expressão Gênica , Proteína FUS de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas , Animais , Astrócitos/citologia , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Meios de Cultivo Condicionados/química , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Proteína Semelhante a ELAV 1 , Humanos , Hipóxia , Camundongos , Neurônios Motores/metabolismo , Fenótipo , RNA/química
14.
Mol Cell ; 54(4): 683-90, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24793692

RESUMO

Splicing is functionally coupled to transcription, linking the rate of RNA polymerase II (Pol II) elongation and the ability of splicing factors to recognize splice sites (ss) of various strengths. In most cases, slow Pol II elongation allows weak splice sites to be recognized, leading to higher inclusion of alternative exons. Using CFTR alternative exon 9 (E9) as a model, we show here that slowing down elongation can also cause exon skipping by promoting the recruitment of the negative factor ETR-3 onto the UG-repeat at E9 3' splice site, which displaces the constitutive splicing factor U2AF65 from the overlapping polypyrimidine tract. Weakening of E9 5' ss increases ETR-3 binding at the 3' ss and subsequent E9 skipping, whereas strengthening of the 5' ss usage has the opposite effect. This indicates that a delay in the cotranscriptional emergence of the 5' ss promotes ETR-3 recruitment and subsequent inhibition of E9 inclusion.


Assuntos
Processamento Alternativo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Éxons , Proteínas Nucleares/metabolismo , RNA Polimerase II/metabolismo , Sítios de Splice de RNA/fisiologia , Ribonucleoproteínas/metabolismo , Sítios de Ligação , Proteínas CELF , Células CACO-2 , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células HEK293 , Humanos , Modelos Genéticos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/genética , Fator de Processamento U2AF , Transcrição Gênica
15.
Nat Rev Mol Cell Biol ; 14(3): 153-65, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23385723

RESUMO

Alternative splicing was discovered simultaneously with splicing over three decades ago. Since then, an enormous body of evidence has demonstrated the prevalence of alternative splicing in multicellular eukaryotes, its key roles in determining tissue- and species-specific differentiation patterns, the multiple post- and co-transcriptional regulatory mechanisms that control it, and its causal role in hereditary disease and cancer. The emerging evidence places alternative splicing in a central position in the flow of eukaryotic genetic information, between transcription and translation, in that it can respond not only to various signalling pathways that target the splicing machinery but also to transcription factors and chromatin structure.


Assuntos
Processamento Alternativo , Biossíntese de Proteínas , Transdução de Sinais , Transcrição Gênica , Animais , Cromatina/genética , Cromatina/metabolismo , Eucariotos/genética , Humanos , Precursores de RNA/genética , Transdução de Sinais/genética , Spliceossomos/genética , Spliceossomos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Biochim Biophys Acta ; 1829(1): 134-40, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22975042

RESUMO

Alternative splicing has emerged as a key contributor to proteome diversity, highlighting the importance of understanding its regulation. In recent years it became apparent that splicing is predominantly cotranscriptional, allowing for crosstalk between these two nuclear processes. We discuss some of the links between transcription and splicing, with special emphasis on the role played by transcription elongation in the regulation of alternative splicing events and in particular the kinetic model of alternative splicing regulation. This article is part of a Special Issue entitled: RNA polymerase II Transcript Elongation.


Assuntos
Processamento Alternativo/fisiologia , Elongação da Transcrição Genética/fisiologia , Processamento Alternativo/genética , Animais , Cromatina/química , Cromatina/metabolismo , Cromatina/fisiologia , Humanos , Cinética , Modelos Biológicos , Ligação Proteica/fisiologia , RNA Polimerase II/metabolismo , RNA Polimerase II/fisiologia
17.
J Cyst Fibros ; 10(3): 212-6, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21317048

RESUMO

BACKGROUND: Cystic fibrosis is caused by mutations of the Cystic Fibrosis Transmembrane conductance Regulator gene (CFTR). Among the 1795 reported mutations, 221 (12.31%) are believed to affect pre-mRNA splicing. Nevertheless, not all splicing mutations have been demonstrated, by functional assays, to affect splicing in living cells. METHODS: We have used a minigene-based approach, coupled to site-specific mutagenesis, to analyze the effects of presumptive pre-mRNA splicing mutations. RESULTS: We show here that the intron 11 1811+1G>C and the intron 12 1898+3A>G mutations strongly affected CFTR pre-mRNA splicing. The encoded proteins are predicted to be defective, which would thus participate in the disease phenotype of carrier individuals. CONCLUSIONS: These results further validate the minigene strategy for the study of presumptive splice mutations, and report unanticipated defects in splicing. Such assays should improve the analysis of genotype-phenotype correlations.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Mutação , Precursores de RNA/genética , Splicing de RNA , Adenina , Processamento Alternativo , Citosina , Guanina , Heterozigoto , Humanos , Íntrons , Mutagênese Sítio-Dirigida , Fenótipo
18.
Nat Struct Mol Biol ; 17(11): 1358-66, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20972445

RESUMO

Pre-mRNA splicing is functionally coupled to transcription, and genotoxic stresses can enhance alternative exon inclusion by affecting elongating RNA polymerase II. We report here that various genotoxic stress inducers, including camptothecin (CPT), inhibit the interaction between Ewing's sarcoma proto-oncoprotein (EWS), an RNA polymerase II-associated factor, and YB-1, a spliceosome-associated factor. This results in the cotranscriptional skipping of several exons of the MDM2 gene, which encodes the main p53 ubiquitin ligase. This reversible exon skipping participates in the regulation of MDM2 expression that may contribute to the accumulation of p53 during stress exposure and its rapid shut-off when stress is removed. Finally, a splicing-sensitive microarray identified numerous exons that are skipped in response to CPT and EWS-YB-1 depletion. These data demonstrate genotoxic stress-induced alteration of the communication between the transcriptional and splicing machineries, which results in widespread exon skipping and plays a central role in the genotoxic stress response.


Assuntos
Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Éxons/genética , Modelos Genéticos , Proteínas Nucleares/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , Camptotecina/farmacologia , Linhagem Celular , DNA Polimerase II/metabolismo , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Fosforilação , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Splicing de RNA/fisiologia , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 de Ligação a Y-Box
19.
Nucleic Acids Res ; 38(20): 7273-85, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20631008

RESUMO

Cystic fibrosis is a prominent genetic disease caused by mutations of the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Among the many disease-causing alterations are pre-mRNA splicing defects that can hamper mandatory exon inclusion. CFTR exon 9 splicing depends in part on a polymorphic UG(m)U(n) sequence at the end of intron 8, which can be bound by TDP-43, leading to partial exon 9 skipping. CELF proteins, like CUG-BP1 and ETR-3, can also bind UG repeats and regulate splicing. We show here that ETR-3, but not CUG-BP1, strongly stimulates exon 9 skipping, although both proteins bind efficiently to the same RNA motif as TDP-43 and with higher affinity. We further show that the skipping of this exon may be due to the functional antagonism between U2AF65 and ETR-3 binding onto the polymorphic U or UG stretch, respectively. Importantly, we demonstrate that the divergent domain of ETR-3 is critical for CFTR exon 9 skipping, as shown by deletion and domain-swapping experiments. We propose a model whereby several RNA-binding events account for the complex regulation of CFTR exon 9 inclusion, with strikingly distinct activities of ETR-3 and CUG-BP1, related to the structure of their divergent domain.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Sítios de Ligação , Linhagem Celular , Éxons , Humanos , Íntrons , Proteínas Nucleares/metabolismo , Estrutura Terciária de Proteína , Precursores de RNA/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Sequências Repetitivas de Ácido Nucleico , Ribonucleoproteínas/metabolismo , Spliceossomos/metabolismo , Fator de Processamento U2AF
20.
Mol Genet Metab ; 99(3): 319-24, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19919903

RESUMO

We have recently found that a common synonymous single nucleotide polymorphism (SNP), c.1275A>G, in exon 9 of the glycoprotein 2 (GP2) gene was significantly underrepresented in French idiopathic chronic pancreatitis patients 20years old or younger at disease onset than in the control population. To further investigate to this preliminary genetic finding, we characterized the functionality of c.1275A>G in the context of a minigene system. Bioinformatics analysis predicted that c.1275A>G could lead to disruption/generation of exonic splicing enhancer hexamers within exon 9 of the GP2 gene. Minigene analysis revealed that both the wild-type and mutant sequences expressed a full-length transcript and a short transcript lacking exon 9. Quantitation of the relative amount of the two transcripts indicated that the fraction of the full-length transcript derived from c.1275A>G is much lower than that derived from the wild-type (51.9% vs 77.4%). Extinction of two splicing factors (SF2/ASF and SC35) by RNA interference also affected c.1275A>G more seriously than the wild-type in terms of exon 9 skipping. Exon 9 skipping was presumed to cause a loss of GP2 function. This study represents the first detailed analysis of any variation in the GP2 gene and gives some support to the putative association of c.1275A>G with disease protection.


Assuntos
Éxons/genética , Glicoproteínas de Membrana/genética , Pancreatite Crônica/genética , Polimorfismo Genético , Sequência de Aminoácidos , Sequência de Bases , Biologia Computacional/métodos , Elementos Facilitadores Genéticos , Proteínas Ligadas por GPI , Humanos , Dados de Sequência Molecular , Interferência de RNA , Precursores de RNA/genética , Precursores de RNA/metabolismo , Splicing de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...