Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
J Gastroenterol ; 56(9): 791-807, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34227026

RESUMO

BACKGROUND: Barrett's esophagus (BE) is a known precursor lesion and the strongest risk factor for esophageal adenocarcinoma (EAC), a common and lethal type of cancer. Prediction of risk, the basis for efficient intervention, is commonly solely based on histologic examination. This approach is challenged by problems such as inter-observer variability in the face of the high heterogeneity of dysplastic tissue. Molecular markers might offer an additional way to understand the carcinogenesis and improve the diagnosis-and eventually treatment. In this study, we probed significant proteomic changes during dysplastic progression from BE into EAC. METHODS: During endoscopic mucosa resection, epithelial and stromal tissue samples were collected by laser capture microdissection from 10 patients with normal BE and 13 patients with high-grade dysplastic/EAC. Samples were analyzed by mass spectrometry-based proteomic analysis. Expressed proteins were determined by label-free quantitation, and gene set enrichment was used to find differentially expressed pathways. The results were validated by immunohistochemistry for two selected key proteins (MSH6 and XPO5). RESULTS: Comparing dysplastic/EAC to non-dysplastic BE, we found in equal volumes of epithelial tissue an overall up-regulation in terms of protein abundance and diversity, and determined a set of 226 differentially expressed proteins. Significantly higher expressions of MSH6 and XPO5 were validated orthogonally and confirmed by immunohistochemistry. CONCLUSIONS: Our results demonstrate that disease-related proteomic alterations can be determined by analyzing minute amounts of cell-type-specific collected tissue. Further analysis indicated that alterations of certain pathways associated with carcinogenesis, such as micro-RNA trafficking, DNA damage repair, and spliceosome activity, exist in dysplastic/EAC.


Assuntos
Esôfago de Barrett/patologia , Expressão Gênica/fisiologia , Spliceossomos/genética , Esôfago de Barrett/genética , Progressão da Doença , Ressecção Endoscópica de Mucosa/métodos , Ressecção Endoscópica de Mucosa/estatística & dados numéricos , Humanos , MicroRNAs/metabolismo , Países Baixos , Spliceossomos/fisiologia
2.
PLoS One ; 15(7): e0233582, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32735620

RESUMO

The craniofacial developmental disorder Burn-McKeown Syndrome (BMKS) is caused by biallelic variants in the pre-messenger RNA splicing factor gene TXNL4A/DIB1. The majority of affected individuals with BMKS have a 34 base pair deletion in the promoter region of one allele of TXNL4A combined with a loss-of-function variant on the other allele, resulting in reduced TXNL4A expression. However, it is unclear how reduced expression of this ubiquitously expressed spliceosome protein results in craniofacial defects during development. Here we reprogrammed peripheral mononuclear blood cells from a BMKS patient and her unaffected mother into induced pluripotent stem cells (iPSCs) and differentiated the iPSCs into induced neural crest cells (iNCCs), the key cell type required for correct craniofacial development. BMKS patient-derived iPSCs proliferated more slowly than both mother- and unrelated control-derived iPSCs, and RNA-Seq analysis revealed significant differences in gene expression and alternative splicing. Patient iPSCs displayed defective differentiation into iNCCs compared to maternal and unrelated control iPSCs, in particular a delay in undergoing an epithelial-to-mesenchymal transition (EMT). RNA-Seq analysis of differentiated iNCCs revealed widespread gene expression changes and mis-splicing in genes relevant to craniofacial and embryonic development that highlight a dampened response to WNT signalling, the key pathway activated during iNCC differentiation. Furthermore, we identified the mis-splicing of TCF7L2 exon 4, a key gene in the WNT pathway, as a potential cause of the downregulated WNT response in patient cells. Additionally, mis-spliced genes shared common sequence properties such as length, branch point to 3' splice site (BPS-3'SS) distance and splice site strengths, suggesting that splicing of particular subsets of genes is particularly sensitive to changes in TXNL4A expression. Together, these data provide the first insight into how reduced TXNL4A expression in BMKS patients might compromise splicing and NCC function, resulting in defective craniofacial development in the embryo.


Assuntos
Processamento Alternativo , Atresia das Cóanas/patologia , Surdez/congênito , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Ribonucleoproteína Nuclear Pequena U5/deficiência , Spliceossomos/fisiologia , Apoptose , Diferenciação Celular , Técnicas de Reprogramação Celular , Atresia das Cóanas/genética , Células Clonais , Surdez/genética , Surdez/patologia , Transição Epitelial-Mesenquimal , Éxons/genética , Face/embriologia , Fácies , Feminino , Cabeça/embriologia , Cardiopatias Congênitas/genética , Humanos , Crista Neural/citologia , Regiões Promotoras Genéticas/genética , Sítios de Splice de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonucleoproteína Nuclear Pequena U5/genética , Deleção de Sequência , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Via de Sinalização Wnt
3.
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
4.
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
5.
Curr Biol ; 29(19): 3193-3199.e4, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31543449

RESUMO

An overwhelming majority of eukaryotic introns have GT/AG ends, whose identities play a critical role for their recognition and removal by the U2 spliceosome, a well-conserved complex of protein and RNAs. Introns with other splice sites exist at very low frequencies in various genomes, and some of them are processed by the U12 spliceosome. Here, we show that, in the chordate Fritillaria borealis, the majority of old introns have been lost and replaced by introns with highly divergent splice sites. The new introns of F. borealis are exceptionally diverse, though more frequently AG/AC or AG/AT, and features of thousands of them support an origin from transposons. They cannot be processed in human cells, but their splicing is rescued by mutating terminal dinucleotides to GT/AG. With lariat sequencing and splicing inhibitor assays, we show that F. borealis introns are spliced by the U2 spliceosome, which thus evolved to a different selectivity, with neither novel U1 small nuclear RNA (snRNA) types nor major remodeling of its protein and snRNA complements. This genome-wide recolonization by non-canonical introns emphasizes the importance of transposons as a resource of novel introns in a context of massive intron loss. An evolution of the spliceosome may also permit to neutralize harmful transposons through their conversion into introns.


Assuntos
Evolução Molecular , Íntrons/genética , Spliceossomos/fisiologia , Urocordados/genética , Animais
6.
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
7.
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
9.
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
10.
Biomed Res Int ; 2018: 3819719, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29789787

RESUMO

Serine/arginine-rich splicing factors (SRSFs) have one or two RNA recognition motifs in the N terminal and a serine/arginine-enriched domain in the C terminal. SRSFs are essential components of spliceosomes and are involved in alternative splicing, spliceosome assembly, mRNA export, and nonsense-mediated mRNA decay. The maintenance of cellular and tissue homeostasis relies on accurate alternative splicing, and various patterns of abnormal alternative splicing can cause different diseases. SRSF4 is associated with many physiological and pathological processes and has applications in the diagnosis and prognosis of specific diseases. In this review, we discuss knowledge of SRSF4 in physiological and pathological processes and highlight the applications of SRSF4 in the regulation of gene expression and associated diseases.


Assuntos
Fatores de Processamento de Serina-Arginina/genética , Fatores de Processamento de Serina-Arginina/fisiologia , Azoospermia/genética , Azoospermia/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica/genética , Humanos , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Polimorfismo de Nucleotídeo Único/genética , Spliceossomos/genética , Spliceossomos/fisiologia
12.
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
13.
Cancer Cell ; 30(3): 404-417, 2016 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-27622333

RESUMO

More than 80% of patients with the refractory anemia with ring sideroblasts subtype of myelodysplastic syndrome (MDS) have mutations in Splicing Factor 3B, Subunit 1 (SF3B1). We generated a conditional knockin mouse model of the most common SF3B1 mutation, Sf3b1(K700E). Sf3b1(K700E) mice develop macrocytic anemia due to a terminal erythroid maturation defect, erythroid dysplasia, and long-term hematopoietic stem cell (LT-HSC) expansion. Sf3b1(K700E) myeloid progenitors and SF3B1-mutant MDS patient samples demonstrate aberrant 3' splice-site selection associated with increased nonsense-mediated decay. Tet2 loss cooperates with Sf3b1(K700E) to cause a more severe erythroid and LT-HSC phenotype. Furthermore, the spliceosome modulator, E7017, selectively kills SF3B1(K700E)-expressing cells. Thus, SF3B1(K700E) expression reflects the phenotype of the mutation in MDS and may be a therapeutic target in MDS.


Assuntos
Eritropoese/fisiologia , Fosfoproteínas/genética , Fatores de Processamento de RNA/genética , Spliceossomos/fisiologia , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Eritropoese/genética , Células-Tronco Hematopoéticas/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Fosfoproteínas/deficiência , Fosfoproteínas/metabolismo , Mutação Puntual , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Splicing de RNA , Fatores de Processamento de RNA/deficiência , Fatores de Processamento de RNA/metabolismo
14.
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
15.
Nat Commun ; 7: 11168, 2016 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-27041075

RESUMO

Modified U1 snRNAs bound to intronic sequences downstream of the 5' splice site correct exon skipping caused by different types of mutations. Here we evaluate the therapeutic activity and structural requirements of these exon-specific U1 snRNA (ExSpeU1) particles. In a severe spinal muscular atrophy, mouse model, ExSpeU1, introduced by germline transgenesis, increases SMN2 exon 7 inclusion, SMN protein production and extends life span. In vitro, RNA mutant analysis and silencing experiments show that while U1A protein is dispensable, the 70K and stem loop IV elements mediate most of the splicing rescue activity through improvement of exon and intron definition. Our findings indicate that precise engineering of the U1 core spliceosomal RNA particle has therapeutic potential in pathologies associated with exon-skipping mutations.


Assuntos
Atrofia Muscular Espinal/genética , Splicing de RNA , RNA Nuclear Pequeno/fisiologia , Animais , Animais Geneticamente Modificados , Terapia Genética , Camundongos , Atrofia Muscular Espinal/patologia , Conformação de Ácido Nucleico , Fenótipo , Sítios de Splice de RNA , RNA Nuclear Pequeno/química , Spliceossomos/química , Spliceossomos/genética , Spliceossomos/fisiologia
16.
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
17.
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
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...