Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 177(2): 339-351.e13, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30879786

RESUMO

Pre-mRNA splicing is executed by the spliceosome. Structural characterization of the catalytically activated complex (B∗) is pivotal for understanding the branching reaction. In this study, we assembled the B∗ complexes on two different pre-mRNAs from Saccharomyces cerevisiae and determined the cryo-EM structures of four distinct B∗ complexes at overall resolutions of 2.9-3.8 Å. The duplex between U2 small nuclear RNA (snRNA) and the branch point sequence (BPS) is discretely away from the 5'-splice site (5'SS) in the three B∗ complexes that are devoid of the step I splicing factors Yju2 and Cwc25. Recruitment of Yju2 into the active site brings the U2/BPS duplex into the vicinity of 5'SS, with the BPS nucleophile positioned 4 Å away from the catalytic metal M2. This analysis reveals the functional mechanism of Yju2 and Cwc25 in branching. These structures on different pre-mRNAs reveal substrate-specific conformations of the spliceosome in a major functional state.


Assuntos
Spliceossomos/fisiologia , Spliceossomos/ultraestrutura , Domínio Catalítico/fisiologia , Microscopia Crioeletrônica/métodos , Éxons , Íntrons , Proteínas Nucleares/metabolismo , Precursores de RNA/metabolismo , Sítios de Splice de RNA/genética , Splicing de RNA/fisiologia , Fatores de Processamento de RNA/metabolismo , RNA Nuclear Pequeno/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Spliceossomos/metabolismo
2.
Nat Rev Genet ; 17(1): 19-32, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26593421

RESUMO

The human transcriptome is composed of a vast RNA population that undergoes further diversification by splicing. Detecting specific splice sites in this large sequence pool is the responsibility of the major and minor spliceosomes in collaboration with numerous splicing factors. This complexity makes splicing susceptible to sequence polymorphisms and deleterious mutations. Indeed, RNA mis-splicing underlies a growing number of human diseases with substantial societal consequences. Here, we provide an overview of RNA splicing mechanisms followed by a discussion of disease-associated errors, with an emphasis on recently described mutations that have provided new insights into splicing regulation. We also discuss emerging strategies for splicing-modulating therapy.


Assuntos
Processamento Alternativo , Animais , Sequência de Bases , Terapia Genética , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Spliceossomos/fisiologia
3.
PLoS Comput Biol ; 16(3): e1007717, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32210422

RESUMO

Spatial organization is a characteristic of all cells, achieved in eukaryotic cells by utilizing both membrane-bound and membrane-less organelles. One of the key processes in eukaryotes is RNA splicing, which readies mRNA for translation. This complex and highly dynamical chemical process involves assembly and disassembly of many molecules in multiple cellular compartments and their transport among compartments. Our goal is to model the effect of spatial organization of membrane-less organelles (specifically nuclear speckles) and of organelle heterogeneity on splicing particle biogenesis in mammalian cells. Based on multiple sources of complementary experimental data, we constructed a spatial model of a HeLa cell to capture intracellular crowding effects. We then developed chemical reaction networks to describe the formation of RNA splicing machinery complexes and splicing processes within nuclear speckles (specific type of non-membrane-bound organelles). We incorporated these networks into our spatially-resolved human cell model and performed stochastic simulations for up to 15 minutes of biological time, the longest thus far for a eukaryotic cell. We find that an increase (decrease) in the number of nuclear pore complexes increases (decreases) the number of assembled splicing particles; and that compartmentalization is critical for the yield of correctly-assembled particles. We also show that a slight increase of splicing particle localization into nuclear speckles leads to a disproportionate enhancement of mRNA splicing and a reduction in the noise of generated mRNA. Our model also predicts that the distance between genes and speckles has a considerable effect on the mRNA production rate, with genes located closer to speckles producing mRNA at higher levels, emphasizing the importance of genome organization around speckles. The HeLa cell model, including organelles and sub-compartments, provides a flexible foundation to study other cellular processes that are strongly modulated by spatiotemporal heterogeneity.


Assuntos
Modelos Biológicos , Splicing de RNA/fisiologia , RNA Mensageiro/metabolismo , Spliceossomos , Biologia Computacional , Simulação por Computador , Células HeLa , Humanos , Espaço Intracelular/química , Espaço Intracelular/metabolismo , Espaço Intracelular/fisiologia , Cinética , RNA Mensageiro/química , Spliceossomos/química , Spliceossomos/metabolismo , Spliceossomos/fisiologia
4.
Nature ; 523(7558): 47-52, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-26106855

RESUMO

U4/U6.U5 tri-snRNP is a 1.5-megadalton pre-assembled spliceosomal complex comprising U5 small nuclear RNA (snRNA), extensively base-paired U4/U6 snRNAs and more than 30 proteins, including the key components Prp8, Brr2 and Snu114. The tri-snRNP combines with a precursor messenger RNA substrate bound to U1 and U2 small nuclear ribonucleoprotein particles (snRNPs), and transforms into a catalytically active spliceosome after extensive compositional and conformational changes triggered by unwinding of the U4 and U6 (U4/U6) snRNAs. Here we use cryo-electron microscopy single-particle reconstruction of Saccharomyces cerevisiae tri-snRNP at 5.9 Å resolution to reveal the essentially complete organization of its RNA and protein components. The single-stranded region of U4 snRNA between its 3' stem-loop and the U4/U6 snRNA stem I is loaded into the Brr2 helicase active site ready for unwinding. Snu114 and the amino-terminal domain of Prp8 position U5 snRNA to insert its loop I, which aligns the exons for splicing, into the Prp8 active site cavity. The structure provides crucial insights into the activation process and the active site of the spliceosome.


Assuntos
Modelos Moleculares , Ribonucleoproteína Nuclear Pequena U4-U6/química , Saccharomyces cerevisiae/química , Spliceossomos/fisiologia , Sítios de Ligação , Microscopia Crioeletrônica , Estrutura Quaternária de Proteína , RNA Helicases/química , RNA Helicases/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo , Ribonucleoproteína Nuclear Pequena U5/química , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Spliceossomos/química
5.
Proc Natl Acad Sci U S A ; 115(26): 6584-6589, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29891649

RESUMO

The spliceosome (SPL) is a majestic macromolecular machinery composed of five small nuclear RNAs and hundreds of proteins. SPL removes noncoding introns from precursor messenger RNAs (pre-mRNAs) and ligates coding exons, giving rise to functional mRNAs. Building on the first SPL structure solved at near-atomic-level resolution, here we elucidate the functional dynamics of the intron lariat spliceosome (ILS) complex through multi-microsecond-long molecular-dynamics simulations of ∼1,000,000 atoms models. The ILS essential dynamics unveils (i) the leading role of the Spp42 protein, which heads the gene maturation by tuning the motions of distinct SPL components, and (ii) the critical participation of the Cwf19 protein in displacing the intron lariat/U2 branch helix. These findings provide unprecedented details on the SPL functional dynamics, thus contributing to move a step forward toward a thorough understanding of eukaryotic pre-mRNA splicing.


Assuntos
Simulação por Computador , Íntrons/genética , Modelos Genéticos , Conformação de Ácido Nucleico , Precursores de RNA/metabolismo , Splicing de RNA/fisiologia , Proteínas Repressoras/fisiologia , Ribonucleoproteína Nuclear Pequena U5/fisiologia , Proteínas de Schizosaccharomyces pombe/fisiologia , Spliceossomos/fisiologia , Magnésio/fisiologia , Modelos Moleculares , Simulação de Dinâmica Molecular , Movimento (Física) , Análise de Componente Principal , Conformação Proteica , Precursores de RNA/genética , RNA Fúngico/genética , RNA Fúngico/metabolismo , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo , Proteínas Repressoras/química , Ribonucleoproteína Nuclear Pequena U5/química , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/química , Eletricidade Estática
6.
Dev Dyn ; 249(8): 924-945, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32315467

RESUMO

Mutations in core components of the spliceosome are responsible for a group of syndromes collectively known as spliceosomopathies. Patients exhibit microcephaly, micrognathia, malar hypoplasia, external ear anomalies, eye anomalies, psychomotor delay, intellectual disability, limb, and heart defects. Craniofacial malformations in these patients are predominantly found in neural crest cells-derived structures of the face and head. Mutations in eight genes SNRPB, RNU4ATAC, SF3B4, PUF60, EFTUD2, TXNL4, EIF4A3, and CWC27 are associated with craniofacial spliceosomopathies. In this review, we provide a brief description of the normal development of the head and the face and an overview of mutations identified in genes associated with craniofacial spliceosomopathies. We also describe a model to explain how and when these mutations are most likely to impact neural crest cells. We speculate that mutations in a subset of core splicing factors lead to disrupted splicing in neural crest cells because these cells have increased sensitivity to inefficient splicing. Hence, disruption in splicing likely activates a cellular stress response that includes increased skipping of regulatory exons in genes such as MDM2 and MDM4, key regulators of P53. This would result in P53-associated death of neural crest cells and consequently craniofacial malformations associated with spliceosomopathies.


Assuntos
Anormalidades Craniofaciais/genética , Deficiência Intelectual/genética , Transtornos Psicomotores/genética , Spliceossomos/fisiologia , Animais , Proteínas de Ciclo Celular/genética , Atresia das Cóanas/genética , Ciclofilinas/genética , RNA Helicases DEAD-box/genética , Surdez/congênito , Surdez/genética , Modelos Animais de Doenças , Fator de Iniciação 4A em Eucariotos/genética , Éxons , Fácies , Cardiopatias Congênitas/genética , Humanos , Camundongos , Microcefalia/genética , Micrognatismo/genética , Mutação , Crista Neural/citologia , Crista Neural/metabolismo , Células Neuroepiteliais/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Fatores de Processamento de RNA/genética , Ribonucleoproteína Nuclear Pequena U5/genética , Síndrome , Proteína Supressora de Tumor p53/genética
7.
Genes Chromosomes Cancer ; 58(12): 889-902, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31334570

RESUMO

Since the discovery of RNA splicing more than 40 years ago, our comprehension of the molecular events orchestrating constitutive and alternative splicing has greatly improved. Dysregulation of pre-mRNA splicing has been observed in many human diseases including neurodegenerative diseases and cancer. The recent identification of frequent somatic mutations in core components of the spliceosome in myeloid malignancies and functional analysis using model systems has advanced our knowledge of how splicing alterations contribute to disease pathogenesis. In this review, we summarize our current understanding on the mechanisms of how mutant splicing factors impact splicing and the resulting functional and pathophysiological consequences. We also review recent advances to develop novel therapeutic approaches targeting splicing catalysis and splicing regulatory proteins, and discuss emerging technologies using oligonucleotide-based therapies to modulate pathogenically spliced isoforms.


Assuntos
Leucemia Mieloide/genética , Síndromes Mielodisplásicas/genética , Spliceossomos/genética , Humanos , Leucemia Mieloide/terapia , Mutação , Síndromes Mielodisplásicas/terapia , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/terapia , Neoplasias/genética , Precursores de RNA/genética , Splicing de RNA , Fatores de Processamento de RNA/genética , Ribonucleoproteína Nuclear Pequena U2/genética , Ribonucleoproteínas Nucleares Pequenas/genética , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Spliceossomos/fisiologia , Fator de Processamento U2AF/genética
8.
Hum Mol Genet ; 26(13): 2472-2479, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28430982

RESUMO

Mitochondria are thought to have originated as free-living prokaryotes. Mitochondria organelles have small circular genomes with substantial structural and genetic similarity to bacteria. Contrary to the prevailing concept of intronless mitochondria, here we present evidence that mitochondrial RNA transcripts (mtRNA) are not limited to policystronic molecules, but also processed as nuclei-like transcripts that are differentially spliced and expressed in a cell-type specific manner. The presence of canonical splice sites in the mtRNA introns and of core components of the nuclei-encoded spliceosome machinery within the mitochondrial organelle suggest that nuclei-encoded spliceosome can mediate splicing of mtRNA.


Assuntos
Mitocôndrias/genética , RNA/genética , RNA/fisiologia , Núcleo Celular , Genoma , Humanos , Íntrons , Mitocôndrias/metabolismo , Splicing de RNA/fisiologia , RNA Mitocondrial , Spliceossomos/genética , Spliceossomos/fisiologia
9.
Mol Cell ; 43(6): 1033-9, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21925391

RESUMO

Poor understanding of the spliceosomal mechanisms to select intronic 3' ends (3'ss) is a major obstacle to deciphering eukaryotic genomes. Here, we discern the rules for global 3'ss selection in yeast. We show that, in contrast to the uniformity of yeast splicing, the spliceosome uses all available 3'ss within a distance window from the intronic branch site (BS), and that in ∼70% of all possible 3'ss this is likely to be mediated by pre-mRNA structures. Our results reveal that one of these RNA folds acts as an RNA thermosensor, modulating alternative splicing in response to heat shock by controlling alternate 3'ss availability. Thus, our data point to a deeper role for the pre-mRNA in the control of its own fate, and to a simple mechanism for some alternative splicing.


Assuntos
Processamento Alternativo , Precursores de RNA/fisiologia , RNA Fúngico/fisiologia , Saccharomyces cerevisiae/genética , Spliceossomos/fisiologia , Regiões 3' não Traduzidas , Biologia Computacional , Genoma Fúngico , Conformação de Ácido Nucleico , Precursores de RNA/química , Precursores de RNA/metabolismo , RNA Fúngico/química , RNA Fúngico/metabolismo , Análise de Sequência de RNA
10.
Genes Dev ; 25(4): 373-84, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21325135

RESUMO

Alternative splicing (AS) plays a major role in the generation of proteomic diversity and in gene regulation. However, the role of the basal splicing machinery in regulating AS remains poorly understood. Here we show that the core snRNP (small nuclear ribonucleoprotein) protein SmB/B' self-regulates its expression by promoting the inclusion of a highly conserved alternative exon in its own pre-mRNA that targets the spliced transcript for nonsense-mediated mRNA decay (NMD). Depletion of SmB/B' in human cells results in reduced levels of snRNPs and a striking reduction in the inclusion levels of hundreds of additional alternative exons, with comparatively few effects on constitutive exon splicing levels. The affected alternative exons are enriched in genes encoding RNA processing and other RNA-binding factors, and a subset of these exons also regulate gene expression by activating NMD. Our results thus demonstrate a role for the core spliceosomal machinery in controlling an exon network that appears to modulate the levels of many RNA processing factors.


Assuntos
Processamento Alternativo/genética , Processamento Alternativo/fisiologia , Precursores de RNA/metabolismo , Spliceossomos/metabolismo , Sequência de Bases , Sequência Conservada , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Mutação/fisiologia , Processamento Pós-Transcricional do RNA/genética , Processamento Pós-Transcricional do RNA/fisiologia , Elementos Reguladores de Transcrição/fisiologia , Ribonucleoproteínas Nucleares Pequenas/genética , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Ribonucleoproteínas Nucleares Pequenas/fisiologia , Spliceossomos/fisiologia , Transfecção , Proteínas Centrais de snRNP/genética , Proteínas Centrais de snRNP/metabolismo
11.
Proc Natl Acad Sci U S A ; 112(33): 10278-85, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-25883267

RESUMO

Eukaryogenesis is widely viewed as an improbable evolutionary transition uniquely affecting the evolution of life on this planet. However, scientific and popular rhetoric extolling this event as a singularity lacks rigorous evidential and statistical support. Here, we question several of the usual claims about the specialness of eukaryogenesis, focusing on both eukaryogenesis as a process and its outcome, the eukaryotic cell. We argue in favor of four ideas. First, the criteria by which we judge eukaryogenesis to have required a genuinely unlikely series of events 2 billion years in the making are being eroded by discoveries that fill in the gaps of the prokaryote:eukaryote "discontinuity." Second, eukaryogenesis confronts evolutionary theory in ways not different from other evolutionary transitions in individuality; parallel systems can be found at several hierarchical levels. Third, identifying which of several complex cellular features confer on eukaryotes a putative richer evolutionary potential remains an area of speculation: various keys to success have been proposed and rejected over the five-decade history of research in this area. Fourth, and perhaps most importantly, it is difficult and may be impossible to eliminate eukaryocentric bias from the measures by which eukaryotes as a whole are judged to have achieved greater success than prokaryotes as a whole. Overall, we question whether premises of existing theories about the uniqueness of eukaryogenesis and the greater evolutionary potential of eukaryotes have been objectively formulated and whether, despite widespread acceptance that eukaryogenesis was "special," any such notion has more than rhetorical value.


Assuntos
Evolução Biológica , Eucariotos/metabolismo , Origem da Vida , Animais , Metabolismo Energético , Escherichia coli/metabolismo , Células Eucarióticas/citologia , Evolução Molecular , Genoma Bacteriano , Humanos , Íntrons , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Fenótipo , Filogenia , Plantas , Células Procarióticas/citologia , Spliceossomos/fisiologia
12.
Proc Natl Acad Sci U S A ; 112(30): 9382-7, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26170331

RESUMO

The mechanisms by which poikilothermic organisms ensure that biological processes are robust to temperature changes are largely unknown. Temperature compensation, the ability of circadian rhythms to maintain a relatively constant period over the broad range of temperatures resulting from seasonal fluctuations in environmental conditions, is a defining property of circadian networks. Temperature affects the alternative splicing (AS) of several clock genes in fungi, plants, and flies, but the splicing factors that modulate these effects to ensure clock accuracy throughout the year remain to be identified. Here we show that GEMIN2, a spliceosomal small nuclear ribonucleoprotein assembly factor conserved from yeast to humans, modulates low temperature effects on a large subset of pre-mRNA splicing events. In particular, GEMIN2 controls the AS of several clock genes and attenuates the effects of temperature on the circadian period in Arabidopsis thaliana. We conclude that GEMIN2 is a key component of a posttranscriptional regulatory mechanism that ensures the appropriate acclimation of plants to daily and seasonal changes in temperature conditions.


Assuntos
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Regulação da Expressão Gênica de Plantas , Proteínas do Complexo SMN/fisiologia , Processamento Alternativo , Sequência de Aminoácidos , Arabidopsis/fisiologia , Proteínas de Arabidopsis/fisiologia , Ritmo Circadiano , Análise por Conglomerados , Evolução Molecular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Teste de Complementação Genética , Estudo de Associação Genômica Ampla , Humanos , Íntrons , Dados de Sequência Molecular , Mutação , Proteínas do Tecido Nervoso/genética , Folhas de Planta/fisiologia , RNA Nuclear Pequeno/genética , Proteínas de Ligação a RNA/genética , Proteínas do Complexo SMN/genética , Homologia de Sequência de Aminoácidos , Spliceossomos/fisiologia , Temperatura , Transcrição Gênica
13.
Genes Dev ; 24(8): 827-36, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20351053

RESUMO

Group II introns are self-splicing RNAs found in eubacteria, archaea, and eukaryotic organelles. They are mechanistically similar to the metazoan nuclear spliceosomal introns; therefore, group II introns have been invoked as the progenitors of the eukaryotic pre-mRNA introns. However, the ability of group II introns to function outside of the bacteria-derived organelles is debatable, since they are not found in the nuclear genomes of eukaryotes. Here, we show that the Lactococcus lactis Ll.LtrB group II intron splices accurately and efficiently from different pre-mRNAs in a eukaryote, Saccharomyces cerevisiae. However, a pre-mRNA harboring a group II intron is spliced predominantly in the cytoplasm and is subject to nonsense-mediated mRNA decay (NMD), and the mature mRNA from which the group II intron is spliced is poorly translated. In contrast, a pre-mRNA bearing the Tetrahymena group I intron or the yeast spliceosomal ACT1 intron at the same location is not subject to NMD, and the mature mRNA is translated efficiently. Thus, a group II intron can splice from a nuclear transcript, but RNA instability and translation defects would have favored intron loss or evolution into protein-dependent spliceosomal introns, consistent with the bacterial group II intron ancestry hypothesis.


Assuntos
Proteínas de Bactérias/genética , Núcleo Celular/metabolismo , Regulação Bacteriana da Expressão Gênica , Íntrons/genética , Lactococcus lactis/genética , Splicing de RNA/genética , Citoplasma/metabolismo , Éxons/genética , Estabilidade de RNA , Saccharomyces cerevisiae/genética , Spliceossomos/fisiologia
14.
Dev Biol ; 414(1): 58-71, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27046631

RESUMO

CACN-1/Cactin is a conserved protein identified in a genome-wide screen for genes that regulate distal tip cell migration in the nematode Caenorhabditis elegans. In addition to possessing distal tip cells that migrate past their correct stopping point, animals depleted of cacn-1 are sterile. In this study, we show that CACN-1 is needed in the soma for proper germ line development and maturation. When CACN-1 is depleted, sheath cells are absent and/or abnormal. When sheath cells are absent, hermaphrodites produce sperm, but do not switch appropriately to oocyte production. When sheath cells are abnormal, some oocytes develop but are not successfully ovulated and undergo endomitotic reduplication (Emo). Our previous proteomic studies show that CACN-1 interacts with a network of splicing factors. Here, these interactors were screened using RNAi. Depletion of many of these factors led to missing or abnormal sheath cells and germ line defects, particularly absent and/or Emo oocytes. These results suggest CACN-1 is part of a protein network that influences somatic gonad development and function through alternative splicing or post-transcriptional gene regulation.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Proteínas Nucleares/fisiologia , Oócitos/citologia , Oogênese/fisiologia , Animais , Movimento Celular , Feminino , Gônadas/citologia , Gônadas/fisiologia , Organismos Hermafroditas/fisiologia , Masculino , Microscopia de Fluorescência , Proteínas Nucleares/deficiência , Oogênese/genética , Mapeamento de Interação de Proteínas , Interferência de RNA , Fatores de Processamento de RNA/fisiologia , Processos de Determinação Sexual , Espermatogênese , Spliceossomos/fisiologia
15.
Biochim Biophys Acta ; 1847(9): 798-808, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25626174

RESUMO

During the evolution of eukaryotic genomes, many genes have been interrupted by intervening sequences (introns) that must be removed post-transcriptionally from RNA precursors to form mRNAs ready for translation. The origin of nuclear introns is still under debate, but one hypothesis is that the spliceosome and the intron-exon structure of genes have evolved from bacterial-type group II introns that invaded the eukaryotic genomes. The group II introns were most likely introduced into the eukaryotic genome from an α-proteobacterial predecessor of mitochondria early during the endosymbiosis event. These self-splicing and mobile introns spread through the eukaryotic genome and later degenerated. Pieces of introns became part of the general splicing machinery we know today as the spliceosome. In addition, group II introns likely brought intron maturases with them to the nucleus. Maturases are found in most bacterial introns, where they act as highly specific splicing factors for group II introns. In the spliceosome, the core protein Prp8 shows homology to group II intron-encoded maturases. While maturases are entirely intron specific, their descendant of the spliceosomal machinery, the Prp8 protein, is an extremely versatile splicing factor with multiple interacting proteins and RNAs. How could such a general player in spliceosomal splicing evolve from the monospecific bacterial maturases? Analysis of the organellar splicing machinery in plants may give clues on the evolution of nuclear splicing. Plants encode various proteins which are closely related to bacterial maturases. The organellar genomes contain one maturase each, named MatK in chloroplasts and MatR in mitochondria. In addition, several maturase genes have been found in the nucleus as well, which are acting on mitochondrial pre-RNAs. All plant maturases show sequence deviation from their progenitor bacterial maturases, and interestingly are all acting on multiple organellar group II intron targets. Moreover, they seem to function in the splicing of group II introns together with a number of additional nuclear-encoded splicing factors, possibly acting as an organellar proto-spliceosome. Together, this makes them interesting models for the early evolution of nuclear spliceosomal splicing. In this review, we summarize recent advances in our understanding of the role of plant maturases and their accessory factors in plants. This article is part of a Special Issue entitled: Chloroplast Biogenesis.


Assuntos
Endorribonucleases/fisiologia , Evolução Molecular , Nucleotidiltransferases/fisiologia , Spliceossomos/fisiologia , Núcleo Celular/enzimologia , Cloroplastos/enzimologia , Íntrons , Mitocôndrias/enzimologia , Splicing de RNA
16.
RNA ; 20(1): 46-60, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24231520

RESUMO

The stepwise assembly of the highly dynamic spliceosome is guided by RNA-dependent ATPases of the DEAD-box family, whose regulation is poorly understood. In the canonical assembly model, the U4/U6.U5 triple snRNP binds only after joining of the U1 and, subsequently, U2 snRNPs to the intron-containing pre-mRNA. Catalytic activation requires the exchange of U6 for U1 snRNA at the 5' splice site, which is promoted by the DEAD-box protein Prp28. Because Prp8, an integral U5 snRNP protein, is thought to be a central regulator of DEAD-box proteins, we conducted a targeted search in Prp8 for cold-insensitive suppressors of a cold-sensitive Prp28 mutant, prp28-1. We identified a cluster of suppressor mutations in an N-terminal bromodomain-like sequence of Prp8. To identify the precise defect in prp28-1 strains that is suppressed by the Prp8 alleles, we analyzed spliceosome assembly in vivo and in vitro. Surprisingly, in the prp28-1 strain, we observed a block not only to spliceosome activation but also to one of the earliest steps of assembly, formation of the ATP-independent commitment complex 2 (CC2). The Prp8 suppressor partially corrected both the early assembly and later activation defects of prp28-1, supporting a role for this U5 snRNP protein in both the ATP-independent and ATP-dependent functions of Prp28. We conclude that the U5 snRNP has a role in the earliest events of assembly, prior to its stable incorporation into the spliceosome.


Assuntos
Processamento Alternativo/fisiologia , RNA Helicases DEAD-box/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Trifosfato de Adenosina/farmacologia , Sequência de Aminoácidos , Substituição de Aminoácidos/fisiologia , RNA Helicases DEAD-box/genética , Modelos Moleculares , Dados de Sequência Molecular , Multimerização Proteica/efeitos dos fármacos , Multimerização Proteica/fisiologia , RNA Nuclear Pequeno/fisiologia , Ribonucleoproteína Nuclear Pequena U4-U6/genética , Ribonucleoproteína Nuclear Pequena U5/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Homologia de Sequência de Aminoácidos , Spliceossomos/efeitos dos fármacos , Spliceossomos/fisiologia
17.
RNA ; 20(7): 1078-89, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24865609

RESUMO

Biallelic mutations of the human RNU4ATAC gene, which codes for the minor spliceosomal U4atac snRNA, cause the developmental disorder, MOPD I/TALS. To date, nine separate mutations in RNU4ATAC have been identified in MOPD I patients. Evidence suggests that all of these mutations lead to abrogation of U4atac snRNA function and impaired minor intron splicing. However, the molecular basis of these effects is unknown. Here, we use a variety of in vitro and in vivo assays to address this question. We find that only one mutation, 124G>A, leads to significantly reduced expression of U4atac snRNA, whereas four mutations, 30G>A, 50G>A, 50G>C and 51G>A, show impaired binding of essential protein components of the U4atac/U6atac di-snRNP in vitro and in vivo. Analysis of MOPD I patient fibroblasts and iPS cells homozygous for the most common mutation, 51G>A, shows reduced levels of the U4atac/U6atac.U5 tri-snRNP complex as determined by glycerol gradient sedimentation and immunoprecipitation. In this report, we establish a mechanistic basis for MOPD I disease and show that the inefficient splicing of genes containing U12-dependent introns in patient cells is due to defects in minor tri-snRNP formation, and the MOPD I-associated RNU4ATAC mutations can affect multiple facets of minor snRNA function.


Assuntos
Nanismo/genética , Retardo do Crescimento Fetal/genética , Microcefalia/genética , Osteocondrodisplasias/genética , RNA Nuclear Pequeno/genética , Spliceossomos/genética , Animais , Sequência de Bases , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Nanismo/metabolismo , Nanismo/patologia , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Recém-Nascido , Microcefalia/metabolismo , Microcefalia/patologia , Dados de Sequência Molecular , Mutação , Conformação de Ácido Nucleico , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patologia , Ligação Proteica , RNA Nuclear Pequeno/química , RNA Nuclear Pequeno/metabolismo , Spliceossomos/química , Spliceossomos/fisiologia
18.
Adv Exp Med Biol ; 907: 189-213, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27256387

RESUMO

RNA editing is a cellular process used to expand and diversify the RNA transcripts produced from a generally immutable genome. In animals, the most prevalent type of RNA editing is adenosine (A) to inosine (I) deamination catalyzed by the ADAR family. Throughout development, A-to-I editing levels increase while ADAR expression is constant, suggesting cellular mechanisms to regulate A-to-I editing exist. Furthermore, in several disease states, ADAR expression levels are similar to the normal state, but A-to-I editing levels are altered. Therefore, understanding how these enzymes are regulated in normal tissues and misregulated in disease states is of profound importance. This chapter will both discuss how to identify A-to-I editing sites across the transcriptome and explore the mechanisms that regulate ADAR editing activity, with particular focus on the diverse types of RNA-binding proteins implicated in regulating A-to-I editing in vivo.


Assuntos
Adenosina Desaminase/fisiologia , Adenosina/metabolismo , Inosina/metabolismo , Edição de RNA , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/fisiologia , Anemia Macrocítica/enzimologia , Anemia Macrocítica/genética , Animais , Pareamento de Bases , Proteínas de Caenorhabditis elegans/fisiologia , Deleção Cromossômica , Cromossomos Humanos Par 5/enzimologia , Cromossomos Humanos Par 5/genética , Proteínas de Drosophila/fisiologia , Humanos , Camundongos , Doenças do Sistema Nervoso/enzimologia , Doenças do Sistema Nervoso/genética , Conformação de Ácido Nucleico , Splicing de RNA , Spliceossomos/fisiologia , Transcriptoma
19.
Proc Natl Acad Sci U S A ; 110(17): 6783-8, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23569281

RESUMO

Removal of introns from the precursors to messenger RNA (pre-mRNAs) requires close apposition of intron ends by the spliceosome, but when and how apposition occurs is unclear. We investigated the process by which intron ends are brought together using single-molecule fluorescence resonance energy transfer together with colocalization single-molecule spectroscopy, a combination of methods that can directly reveal how conformational transitions in macromolecular machines are coupled to specific assembly and disassembly events. The FRET measurements suggest that the 5' splice site and branch site remain physically separated throughout spliceosome assembly, and only approach one another after the spliceosome is activated for catalysis, at which time the pre-mRNA becomes highly dynamic. Separation of the sites of chemistry until very late in the splicing pathway may be crucial for preventing splicing at incorrect sites.


Assuntos
Conformação de Ácido Nucleico , Sítios de Splice de RNA/genética , Splicing de RNA/fisiologia , Spliceossomos/fisiologia , Sequência de Bases , Primers do DNA/genética , Processamento de Imagem Assistida por Computador , Microscopia de Fluorescência , Dados de Sequência Molecular , Oligonucleotídeos/genética , Saccharomyces cerevisiae , Análise Espectral
20.
J Hepatol ; 62(5): 1024-32, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25481564

RESUMO

BACKGROUND &/AIMS: The broadly used antiviral cytokine interferon-α (IFNα)'s mechanisms of action against HCV infection are not well understood. We previously identified SART1, a host protein involved in RNA splicing and pre-mRNA processing, as a regulator of IFN's antiviral effects. We hypothesized that SART1 regulates antiviral IFN effector genes (IEGs) through mRNA processing and splicing. METHODS: We performed siRNA knockdown in HuH7.5.1 cells and mRNA-sequencing with or without IFN treatment. Selected gene mRNA variants and their proteins, together with HCV replication, were monitored by qRT-PCR and Western blot in HCV OR6 replicon cells and the JFH1 HCV infectious model. RESULTS: We identified 419 genes with a greater than 2-fold expression difference between Neg siRNA and SART1 siRNA treated cells in the presence or absence of IFN. Bioinformatic analysis identified at least 10 functional pathways. SART1 knockdown reduced classical IFN stimulating genes (ISG) mRNA transcription including MX1 and OAS3. However, SART1 did not affect JAK-STAT pathway gene mRNA expression and IFN stimulated response element (ISRE) signaling. We identified alternative mRNA splicing events for several genes, including EIF4G3, GORASP2, ZFAND6, and RAB6A that contribute to their antiviral effects. EIF4G3 and GORASP2 were also confirmed to have anti-HCV effect. CONCLUSIONS: The spliceosome factor SART1 is not IFN-inducible but is an IEG. SART1 exerts its anti-HCV action through direct transcriptional regulation for some ISGs and alternative splicing for others, including EIF4G3, GORASP2. SART1 does not have an effect on IFN receptor or canonical signal transduction components. Thus, SART1 regulates ISGs using a novel, non-classical mechanism.


Assuntos
Antígenos de Neoplasias/genética , Hepacivirus/fisiologia , Hepatite C , Interferon-alfa , Splicing de RNA/genética , Ribonucleoproteínas Nucleares Pequenas/genética , Spliceossomos/fisiologia , Antivirais/metabolismo , Antivirais/farmacologia , Técnicas de Silenciamento de Genes , Hepatite C/genética , Hepatite C/virologia , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Replicação Viral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA