Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(39): e2305756120, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37722062

RESUMO

Mutations in RNA/DNA-binding proteins cause amyotrophic lateral sclerosis (ALS), but the underlying disease mechanisms remain unclear. Here, we report that a set of ALS-associated proteins, namely FUS, EWSR1, TAF15, and MATR3, impact the expression of genes encoding the major histocompatibility complex II (MHC II) antigen presentation pathway. Both subunits of the MHC II heterodimer, HLA-DR, are down-regulated in ALS gene knockouts/knockdown in HeLa and human microglial cells, due to loss of the MHC II transcription factor CIITA. Importantly, hematopoietic progenitor cells (HPCs) derived from human embryonic stem cells bearing the FUSR495X mutation and HPCs derived from C9ORF72 ALS patient induced pluripotent stem cells also exhibit disrupted MHC II expression. Given that HPCs give rise to numerous immune cells, our data raise the possibility that loss of the MHC II pathway results in global failure of the immune system to protect motor neurons from damage that leads to ALS.


Assuntos
Esclerose Lateral Amiotrófica , Humanos , Esclerose Lateral Amiotrófica/genética , Apresentação de Antígeno/genética , Genes MHC da Classe II , Complexo Principal de Histocompatibilidade , Neurônios Motores , Proteínas de Ligação a RNA/genética , Proteínas Associadas à Matriz Nuclear
2.
Cancer Cell ; 35(2): 283-296.e5, 2019 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-30712845

RESUMO

SF3B1 is recurrently mutated in chronic lymphocytic leukemia (CLL), but its role in the pathogenesis of CLL remains elusive. Here, we show that conditional expression of Sf3b1-K700E mutation in mouse B cells disrupts pre-mRNA splicing, alters cell development, and induces a state of cellular senescence. Combination with Atm deletion leads to the overcoming of cellular senescence and the development of CLL-like disease in elderly mice. These CLL-like cells show genome instability and dysregulation of multiple CLL-associated cellular processes, including deregulated B cell receptor signaling, which we also identified in human CLL cases. Notably, human CLLs harboring SF3B1 mutations exhibit altered response to BTK inhibition. Our murine model of CLL thus provides insights into human CLL disease mechanisms and treatment.


Assuntos
Linfócitos B/imunologia , Senescência Celular , Deleção de Genes , Leucemia Linfocítica Crônica de Células B/genética , Mutação , Neoplasias Experimentais/genética , Fosfoproteínas/genética , Fatores de Processamento de RNA/genética , Receptores de Antígenos de Linfócitos B/imunologia , Adenina/análogos & derivados , Tirosina Quinase da Agamaglobulinemia/antagonistas & inibidores , Tirosina Quinase da Agamaglobulinemia/metabolismo , Processamento Alternativo , Animais , Antineoplásicos/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia/deficiência , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Senescência Celular/efeitos dos fármacos , Dano ao DNA , Predisposição Genética para Doença , Instabilidade Genômica , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Fenótipo , Fosfoproteínas/metabolismo , Piperidinas , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Fatores de Processamento de RNA/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
3.
Nucleic Acids Res ; 46(22): 11939-11951, 2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30398641

RESUMO

Understanding the molecular pathways disrupted in motor neuron diseases is urgently needed. Here, we employed CRISPR knockout (KO) to investigate the functions of four ALS-causative RNA/DNA binding proteins (FUS, EWSR1, TAF15 and MATR3) within the RNAP II/U1 snRNP machinery. We found that each of these structurally related proteins has distinct roles with FUS KO resulting in loss of U1 snRNP and the SMN complex, EWSR1 KO causing dissociation of the tRNA ligase complex, and TAF15 KO resulting in loss of transcription factors P-TEFb and TFIIF. However, all four ALS-causative proteins are required for association of the ASC-1 transcriptional co-activator complex with the RNAP II/U1 snRNP machinery. Remarkably, mutations in the ASC-1 complex are known to cause a severe form of Spinal Muscular Atrophy (SMA), and we show that an SMA-causative mutation in an ASC-1 component or an ALS-causative mutation in FUS disrupts association between the ASC-1 complex and the RNAP II/U1 snRNP machinery. We conclude that ALS and SMA are more intimately tied to one another than previously thought, being linked via the ASC-1 complex.


Assuntos
Esclerose Lateral Amiotrófica/genética , Atrofia Muscular Espinal/genética , Proteínas Associadas à Matriz Nuclear/genética , Proteína EWS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/genética , Proteínas de Ligação a RNA/genética , Fatores Associados à Proteína de Ligação a TATA/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Sistemas CRISPR-Cas , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Edição de Genes , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patologia , Proteínas Associadas à Matriz Nuclear/deficiência , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fator B de Elongação Transcricional Positiva/genética , Fator B de Elongação Transcricional Positiva/metabolismo , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo , Proteína EWS de Ligação a RNA/deficiência , Proteína FUS de Ligação a RNA/deficiência , Ribonucleoproteína Nuclear Pequena U1/genética , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Spliceossomos/química , Spliceossomos/metabolismo , Fatores Associados à Proteína de Ligação a TATA/deficiência , Fatores de Transcrição TFII/genética , Fatores de Transcrição TFII/metabolismo
4.
JCI Insight ; 3(19)2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30282833

RESUMO

The identification of targetable vulnerabilities in the context of therapeutic resistance is a key challenge in cancer treatment. We detected pervasive aberrant splicing as a characteristic feature of chronic lymphocytic leukemia (CLL), irrespective of splicing factor mutation status, which was associated with sensitivity to the spliceosome modulator, E7107. Splicing modulation affected CLL survival pathways, including members of the B cell lymphoma-2 (BCL2) family of proteins, remodeling antiapoptotic dependencies of human and murine CLL cells. E7107 treatment decreased myeloid cell leukemia-1 (MCL1) dependence and increased BCL2 dependence, sensitizing primary human CLL cells and venetoclax-resistant CLL-like cells from an Eµ-TCL1-based adoptive transfer murine model to treatment with the BCL2 inhibitor venetoclax. Our data provide preclinical rationale to support the combination of venetoclax with splicing modulators to reprogram apoptotic dependencies in CLL for treating venetoclax-resistant CLL cases.


Assuntos
Processamento Alternativo/efeitos dos fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Compostos de Epóxi/farmacologia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Macrolídeos/farmacologia , Sulfonamidas/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Ensaios de Seleção de Medicamentos Antitumorais , Compostos de Epóxi/uso terapêutico , Feminino , Humanos , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Macrolídeos/uso terapêutico , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Mutação , Proteína de Sequência 1 de Leucemia de Células Mieloides/antagonistas & inibidores , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Fosfoproteínas/genética , Cultura Primária de Células , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Fatores de Processamento de RNA/genética , Spliceossomos/efeitos dos fármacos , Spliceossomos/metabolismo , Sulfonamidas/uso terapêutico , Tiofenos/farmacologia , Tiofenos/uso terapêutico
5.
Sci Rep ; 8(1): 8755, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29884807

RESUMO

Mutations in multiple RNA/DNA binding proteins cause Amyotrophic Lateral Sclerosis (ALS). Included among these are the three members of the FET family (FUS, EWSR1 and TAF15) and the structurally similar MATR3. Here, we characterized the interactomes of these four proteins, revealing that they largely have unique interactors, but share in common an association with U1 snRNP. The latter observation led us to analyze the interactome of the U1 snRNP machinery. Surprisingly, this analysis revealed the interactome contains ~220 components, and of these, >200 are shared with the RNA polymerase II (RNAP II) machinery. Among the shared components are multiple ALS and Spinal muscular Atrophy (SMA)-causative proteins and numerous discrete complexes, including the SMN complex, transcription factor complexes, and RNA processing complexes. Together, our data indicate that the RNAP II/U1 snRNP machinery functions in a wide variety of molecular pathways, and these pathways are candidates for playing roles in ALS/SMA pathogenesis.


Assuntos
Proteínas Associadas à Matriz Nuclear/metabolismo , Mapas de Interação de Proteínas , RNA Polimerase II/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Células HeLa , Humanos
6.
Elife ; 62017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28177281

RESUMO

Genomic instability is a hallmark of human cancer, and results in widespread somatic copy number alterations. We used a genome-scale shRNA viability screen in human cancer cell lines to systematically identify genes that are essential in the context of particular copy-number alterations (copy-number associated gene dependencies). The most enriched class of copy-number associated gene dependencies was CYCLOPS (Copy-number alterations Yielding Cancer Liabilities Owing to Partial losS) genes, and spliceosome components were the most prevalent. One of these, the pre-mRNA splicing factor SF3B1, is also frequently mutated in cancer. We validated SF3B1 as a CYCLOPS gene and found that human cancer cells harboring partial SF3B1 copy-loss lack a reservoir of SF3b complex that protects cells with normal SF3B1 copy number from cell death upon partial SF3B1 suppression. These data provide a catalog of copy-number associated gene dependencies and identify partial copy-loss of wild-type SF3B1 as a novel, non-driver cancer gene dependency.


Assuntos
Dosagem de Genes , Neoplasias/genética , Neoplasias/patologia , Fosfoproteínas/genética , Fatores de Processamento de RNA/genética , Linhagem Celular Tumoral , Humanos
7.
Cancer Cell ; 30(5): 750-763, 2016 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-27818134

RESUMO

Mutations in SF3B1, which encodes a spliceosome component, are associated with poor outcome in chronic lymphocytic leukemia (CLL), but how these contribute to CLL progression remains poorly understood. We undertook a transcriptomic characterization of primary human CLL cells to identify transcripts and pathways affected by SF3B1 mutation. Splicing alterations, identified in the analysis of bulk cells, were confirmed in single SF3B1-mutated CLL cells and also found in cell lines ectopically expressing mutant SF3B1. SF3B1 mutation was found to dysregulate multiple cellular functions including DNA damage response, telomere maintenance, and Notch signaling (mediated through KLF8 upregulation, increased TERC and TERT expression, or altered splicing of DVL2 transcript, respectively). SF3B1 mutation leads to diverse changes in CLL-related pathways.


Assuntos
Processamento Alternativo , Perfilação da Expressão Gênica/métodos , Leucemia Linfocítica Crônica de Células B/genética , Mutação , Fosfoproteínas/genética , Fatores de Processamento de RNA/genética , Linhagem Celular Tumoral , Proteínas Desgrenhadas/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores Notch/genética , Transdução de Sinais
8.
Proc Natl Acad Sci U S A ; 112(28): 8608-13, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26124092

RESUMO

Pre-mRNA splicing is coupled to transcription by RNA polymerase II (RNAP II). We previously showed that U1 small nuclear ribonucleoprotein (snRNP) associates with RNAP II, and both RNAP II and U1 snRNP are also the most abundant factors associated with the protein fused-in-sarcoma (FUS), which is mutated to cause the neurodegenerative disease amyotrophic lateral sclerosis. Here, we show that an antisense morpholino that base-pairs to the 5' end of U1 snRNA blocks splicing in the coupled system and completely disrupts the association between U1 snRNP and both FUS and RNAP II, but has no effect on the association between FUS and RNAP II. Conversely, we found that U1 snRNP does not interact with RNAP II in FUS knockdown extracts. Moreover, using these extracts, we found that FUS must be present during the transcription reaction in order for splicing to occur. Together, our data lead to a model that FUS functions in coupling transcription to splicing via mediating an interaction between RNAP II and U1 snRNP.


Assuntos
RNA Polimerase II/metabolismo , Splicing de RNA/fisiologia , Proteína FUS de Ligação a RNA/fisiologia , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Transcrição Gênica/fisiologia , Sequência de Bases , Humanos , RNA Nuclear Pequeno/química , RNA Nuclear Pequeno/metabolismo , Proteína FUS de Ligação a RNA/metabolismo
9.
Nucleic Acids Res ; 43(6): 3208-18, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25735748

RESUMO

Mutations in FUS cause amyotrophic lateral sclerosis (ALS), but the molecular pathways leading to neurodegeneration remain obscure. We previously found that U1 snRNP is the most abundant FUS interactor. Here, we report that components of the U1 snRNP core particle (Sm proteins and U1 snRNA), but not the mature U1 snRNP-specific proteins (U1-70K, U1A and U1C), co-mislocalize with FUS to the cytoplasm in ALS patient fibroblasts harboring mutations in the FUS nuclear localization signal (NLS). Similar results were obtained in HeLa cells expressing the ALS-causing FUS R495X NLS mutation, and mislocalization of Sm proteins is RRM-dependent. Moreover, as observed with FUS, knockdown of any of the U1 snRNP-specific proteins results in a dramatic loss of SMN-containing Gems. Significantly, knockdown of U1 snRNP in zebrafish results in motor axon truncations, a phenotype also observed with FUS, SMN and TDP-43 knockdowns. Our observations linking U1 snRNP to ALS patient cells with FUS mutations, SMN-containing Gems, and motor neurons indicate that U1 snRNP is a component of a molecular pathway associated with motor neuron disease. Linking an essential canonical splicing factor (U1 snRNP) to this pathway provides strong new evidence that splicing defects may be involved in pathogenesis and that this pathway is a potential therapeutic target.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Sinais de Localização Nuclear/genética , Proteína FUS de Ligação a RNA/genética , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Animais Geneticamente Modificados , Citoplasma/metabolismo , Gêmeos de Corpos Enovelados/metabolismo , Gêmeos de Corpos Enovelados/patologia , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Mutação , Domínios e Motivos de Interação entre Proteínas , Proteína FUS de Ligação a RNA/química , Proteína FUS de Ligação a RNA/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ribonucleoproteína Nuclear Pequena U1/antagonistas & inibidores , Ribonucleoproteína Nuclear Pequena U1/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas Centrais de snRNP/genética , Proteínas Centrais de snRNP/metabolismo
10.
Nat Neurosci ; 16(7): 851-5, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23708140

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease whose causes are still poorly understood. To identify additional genetic risk factors, we assessed the role of de novo mutations in ALS by sequencing the exomes of 47 ALS patients and both of their unaffected parents (n = 141 exomes). We found that amino acid-altering de novo mutations were enriched in genes encoding chromatin regulators, including the neuronal chromatin remodeling complex (nBAF) component SS18L1 (also known as CREST). CREST mutations inhibited activity-dependent neurite outgrowth in primary neurons, and CREST associated with the ALS protein FUS. These findings expand our understanding of the ALS genetic landscape and provide a resource for future studies into the pathogenic mechanisms contributing to sporadic ALS.


Assuntos
Esclerose Lateral Amiotrófica/genética , Proteínas de Ligação a DNA/genética , Exoma/genética , Predisposição Genética para Doença/genética , Mutação/genética , Proteínas Nucleares/genética , Transativadores/genética , Adulto , Animais , Células Cultivadas , Córtex Cerebral/citologia , Dendritos/genética , Dendritos/metabolismo , Embrião de Mamíferos , Saúde da Família , Feminino , Genótipo , Humanos , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade , Neurônios Motores/citologia , Neurônios Motores/fisiologia , Proteína FUS de Ligação a RNA/genética , Adulto Jovem
11.
Cell Rep ; 2(4): 799-806, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23022481

RESUMO

Mutations in the RNA binding protein FUS cause amyotrophic lateral sclerosis (ALS), a fatal adult motor neuron disease. Decreased expression of SMN causes the fatal childhood motor neuron disorder spinal muscular atrophy (SMA). The SMN complex localizes in both the cytoplasm and nuclear Gems, and loss of Gems is a cellular hallmark of fibroblasts in patients with SMA. Here, we report that FUS associates with the SMN complex, mediated by U1 snRNP and by direct interactions between FUS and SMN. Functionally, we show that FUS is required for Gem formation in HeLa cells, and expression of FUS containing a severe ALS-causing mutation (R495X) also results in Gem loss. Strikingly, a reduction in Gems is observed in ALS patient fibroblasts expressing either mutant FUS or TDP-43, another ALS-causing protein that interacts with FUS. The physical and functional interactions among SMN, FUS, TDP-43, and Gems indicate that ALS and SMA share a biochemical pathway, providing strong support for the view that these motor neuron diseases are related.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Atrofia Muscular Espinal/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Proteínas do Complexo SMN/metabolismo , Esclerose Lateral Amiotrófica/patologia , Proteína DEAD-box 20/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Atrofia Muscular Espinal/patologia , Mutação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteína FUS de Ligação a RNA/antagonistas & inibidores , Proteína FUS de Ligação a RNA/genética , Ribonucleoproteínas Nucleares Pequenas/metabolismo , Proteínas do Complexo SMN/genética
12.
PLoS One ; 7(8): e43804, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22928037

RESUMO

The conserved TREX complex, which contains UAP56, Aly, CIP29, and the multi-subunit THO complex, functions in mRNA export. Recently, several putative new components of the human TREX complex were identified by mass spectrometry. Here, we investigated the function of two of these, PDIP3 and ZC11A. Our data indicate that both of these proteins are components of a common TREX complex and function in mRNA export. Recently, we found that both CIP29 and Aly associate with the DEAD box helicase UAP56 and with the TREX complex in an ATP-dependent manner. We now show that this is also the case for PDIP3 and ZC11A. Thus, together with previous work, our data indicate that the TREX complex participates in multiple ATP-dependent interactions.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Nucleares/metabolismo , Transporte de RNA , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/química , Células HeLa , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/química , Ligação Proteica , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Dedos de Zinco
13.
N Engl J Med ; 365(26): 2497-506, 2011 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-22150006

RESUMO

BACKGROUND: The somatic genetic basis of chronic lymphocytic leukemia, a common and clinically heterogeneous leukemia occurring in adults, remains poorly understood. METHODS: We obtained DNA samples from leukemia cells in 91 patients with chronic lymphocytic leukemia and performed massively parallel sequencing of 88 whole exomes and whole genomes, together with sequencing of matched germline DNA, to characterize the spectrum of somatic mutations in this disease. RESULTS: Nine genes that are mutated at significant frequencies were identified, including four with established roles in chronic lymphocytic leukemia (TP53 in 15% of patients, ATM in 9%, MYD88 in 10%, and NOTCH1 in 4%) and five with unestablished roles (SF3B1, ZMYM3, MAPK1, FBXW7, and DDX3X). SF3B1, which functions at the catalytic core of the spliceosome, was the second most frequently mutated gene (with mutations occurring in 15% of patients). SF3B1 mutations occurred primarily in tumors with deletions in chromosome 11q, which are associated with a poor prognosis in patients with chronic lymphocytic leukemia. We further discovered that tumor samples with mutations in SF3B1 had alterations in pre-messenger RNA (mRNA) splicing. CONCLUSIONS: Our study defines the landscape of somatic mutations in chronic lymphocytic leukemia and highlights pre-mRNA splicing as a critical cellular process contributing to chronic lymphocytic leukemia.


Assuntos
DNA de Neoplasias/análise , Leucemia Linfocítica Crônica de Células B/genética , Mutação , Spliceossomos/genética , Adulto , Deleção Cromossômica , Cromossomos Humanos Par 11/genética , Exoma/genética , Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mutação de Sentido Incorreto , Splicing de RNA
14.
Genes Dev ; 25(5): 440-4, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21363962

RESUMO

Duplex formation between the branch point-binding region (BBR) of U2 snRNA and the branch point sequence (BPS) in the intron is essential for splicing. Both the BBR and BPS interact with the U2 small nuclear ribonucleoprotein (snRNP)-associated SF3b complex, which is the target of the anti-tumor drug E7107. We show that E7107 blocks spliceosome assembly by preventing tight binding of U2 snRNP to pre-mRNA. E7107 has no apparent effect on U2 snRNP integrity. Instead, E7107 abolishes an ATP-dependent conformational change in U2 snRNP that exposes the BBR. We conclude that SF3b is required for this remodeling, which exposes the BBR for tight U2 snRNP binding to pre-mRNA.


Assuntos
Antineoplásicos/farmacologia , Compostos de Epóxi/farmacologia , Macrolídeos/farmacologia , Fosfoproteínas/metabolismo , Ribonucleoproteína Nuclear Pequena U2/metabolismo , Spliceossomos/efeitos dos fármacos , Sítios de Ligação , Células HeLa , Humanos , Ligação Proteica/efeitos dos fármacos , Precursores de RNA/metabolismo , Fatores de Processamento de RNA
15.
Genes Dev ; 24(18): 2043-53, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20844015

RESUMO

The conserved TREX mRNA export complex is known to contain UAP56, Aly, Tex1, and the THO complex. Here, we carried out proteomic analysis of immunopurified human TREX complex and identified the protein CIP29 as the only new component with a clear yeast relative (known as Tho1). Tho1 is known to function in mRNA export, and we provide evidence that CIP29 likewise functions in this process. Like the known TREX components, a portion of CIP29 localizes in nuclear speckle domains, and its efficient recruitment to mRNA is both splicing- and cap-dependent. We show that UAP56 mediates an ATP-dependent interaction between the THO complex and both CIP29 and Aly, indicating that TREX assembly is ATP-dependent. Using recombinant proteins expressed in Escherichia coli, we show that UAP56, Aly, and CIP29 form an ATP-dependent trimeric complex, and UAP56 bridges the interaction between CIP29 and Aly. We conclude that the interaction of two conserved export proteins, CIP29 and Aly, with UAP56 is strictly regulated by ATP during assembly of the TREX complex.


Assuntos
Trifosfato de Adenosina/metabolismo , RNA Helicases DEAD-box/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células COS , Proteínas de Ciclo Celular/metabolismo , Chlorocebus aethiops , RNA Helicases DEAD-box/genética , Exodesoxirribonucleases/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/genética , Processamento Pós-Transcricional do RNA/fisiologia , Transporte de RNA
16.
Nucleic Acids Res ; 36(14): 4708-18, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18628297

RESUMO

The conserved RNA helicase DDX3 is of major medical importance due to its involvement in numerous cancers, human hepatitis C virus (HCV) and HIV. Although DDX3 has been reported to have a wide variety of cellular functions, its precise role remains obscure. Here, we raised a new antibody to DDX3 and used it to show that DDX3 is evenly distributed throughout the cytoplasm at steady state. Consistent with this observation, HA-tagged DDX3 also localizes to the cytoplasm. RNAi of DDX3 in both human and Drosophila cells shows that DDX3 is required for cell viability. Moreover, using RNAi, we show that DDX3 is required for expression of protein from reporter constructs. In contrast, we did not detect a role for DDX3 in nuclear steps in gene expression. Further insight into the function of DDX3 came from the observation that its major interaction partner is the multi-component translation initiation factor eIF3. We conclude that a primary function for DDX3 is in protein translation, via an interaction with eIF3.


Assuntos
RNA Helicases DEAD-box/fisiologia , Fator de Iniciação 3 em Eucariotos/metabolismo , Biossíntese de Proteínas , Animais , Anticorpos , Citoplasma/enzimologia , RNA Helicases DEAD-box/antagonistas & inibidores , RNA Helicases DEAD-box/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Células HeLa , Humanos , RNA Helicases/antagonistas & inibidores , RNA Helicases/genética , RNA Helicases/fisiologia , Interferência de RNA
17.
Blood ; 110(5): 1458-65, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17475909

RESUMO

Shwachman-Diamond syndrome (SDS) is an autosomal recessive disorder characterized by bone marrow failure, exocrine pancreatic dysfunction, and leukemia predisposition. Mutations in the SBDS gene are identified in most patients with SDS. SBDS encodes a highly conserved protein of unknown function. Data from SBDS orthologs suggest that SBDS may play a role in ribosome biogenesis or RNA processing. Human SBDS is enriched in the nucleolus, the major cellular site of ribosome biogenesis. Here we report that SBDS nucleolar localization is dependent on active rRNA transcription. Cells from patients with SDS or Diamond-Blackfan anemia are hypersensitive to low doses of actinomycin D, an inhibitor of rRNA transcription. The addition of wild-type SBDS complements the actinomycin D hypersensitivity of SDS patient cells. SBDS migrates together with the 60S large ribosomal subunit in sucrose gradients and coprecipitates with 28S ribosomal RNA (rRNA). Loss of SBDS is not associated with a discrete block in rRNA maturation or with decreased levels of the 60S ribosomal subunit. SBDS forms a protein complex with nucleophosmin, a multifunctional protein implicated in ribosome biogenesis and leukemogenesis. Our studies support the addition of SDS to the growing list of human bone marrow failure syndromes involving the ribosome.


Assuntos
Doenças da Medula Óssea/metabolismo , Doenças Genéticas Inatas/metabolismo , Pancreatopatias/metabolismo , Proteínas/metabolismo , RNA Ribossômico/metabolismo , Ribossomos/metabolismo , Doenças da Medula Óssea/genética , Doenças da Medula Óssea/patologia , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Células Cultivadas , Dactinomicina/farmacologia , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/patologia , Predisposição Genética para Doença , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Inibidores da Síntese de Ácido Nucleico/farmacologia , Nucleofosmina , Pancreatopatias/genética , Pancreatopatias/patologia , Ligação Proteica/genética , Proteínas/genética , RNA Ribossômico/genética , Ribossomos/genética , Síndrome , Transcrição Gênica/efeitos dos fármacos
18.
J Mol Med (Berl) ; 85(2): 149-61, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17206408

RESUMO

Mutations that affect the splicing of pre-mRNA are a major cause of human disease. Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a T to C transition at base pair 6 of IKBKAP intron 20. This mutation results in variable tissue-specific skipping of exon 20. Previously, we reported that the plant cytokinin kinetin dramatically increases exon 20 inclusion in RNA isolated from cultured FD cells. The goal of the current study was to investigate the nature of the FD splicing defect and the mechanism by which kinetin improves exon inclusion, as such knowledge will facilitate the development of future therapeutics aimed at regulating mRNA splicing. In this study, we demonstrate that treatment of FD lymphoblast cell lines with kinetin increases IKBKAP mRNA and IKAP protein to normal levels. Using a series of minigene constructs, we show that deletion of a region at the end of IKBKAP exon 20 disrupts the ability of kinetin to improve exon inclusion, pinpointing a kinetin responsive sequence element. We next performed a screen of endogenously expressed genes with multiple isoforms resulting from exon skipping events and show that kinetin's ability to improve exon inclusion is not limited to IKBKAP. Lastly, we highlight the potential of kinetin for the treatment of other human splicing disorders by showing correction of a splicing defect in neurofibromatosis.


Assuntos
Proteínas de Transporte/genética , Disautonomia Familiar/tratamento farmacológico , Cinetina/uso terapêutico , Splicing de RNA/efeitos dos fármacos , Proteínas de Transporte/análise , Proteínas de Transporte/efeitos dos fármacos , Linhagem Celular Tumoral , Éxons/efeitos dos fármacos , Humanos , Cinetina/farmacologia , Neurofibromatoses/tratamento farmacológico , Neurofibromatoses/genética , RNA Mensageiro/análise , RNA Mensageiro/efeitos dos fármacos , Fatores de Elongação da Transcrição
19.
Proc Natl Acad Sci U S A ; 101(7): 1858-62, 2004 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-14769921

RESUMO

The TREX (transcription/export) complex couples transcription elongation to the nuclear export of mRNAs. In this article, we show that the poly(A)(+) RNA-binding proteins Gbp2 and Hrb1, which resemble the serine-arginine-rich (SR) family of splicing factors found in higher eukaryotes, are specifically associated with the yeast TREX complex. We also show that Gbp2 and Hrb1 interact with Ctk1, a kinase that phosphorylates the C-terminal domain of RNA polymerase II during transcription elongation. Consistent with these findings, Gbp2 and Hrb1 associate with actively transcribed genes throughout their entire lengths. By using an RNA immunoprecipitation assay, we show that Gbp2 and Hrb1 also are bound to transcripts that are derived from these genes. We conclude that recruitment of the SR-like proteins Gbp2 and Hrb1 to mRNA occurs cotranscriptionally by means of association with the TREX complex and/or Ctk1.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Proteínas Quinases , Transporte de RNA , RNA Fúngico/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transcrição Gênica , Proteínas de Ligação a DNA/metabolismo , Regulação Fúngica da Expressão Gênica , Genes Fúngicos/genética , Substâncias Macromoleculares , Proteínas de Transporte Nucleocitoplasmático , Peptídeos/metabolismo , Proteínas de Ligação a Poli(A) , Testes de Precipitina , Ligação Proteica , RNA Fúngico/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
20.
Mol Pharmacol ; 63(2): 359-67, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12527807

RESUMO

Expression of the human DNA topoisomerase IIalpha (topo IIalpha) gene is positively regulated by the binding of the nuclear factor Y (NF-Y) transcription factor to four of five inverted CCAAT boxes (ICBs) located in its promoter. We have demonstrated previously that expression of the p53 tumor suppressor inhibits human topo IIalpha promoter activity in murine (10)1 cells. In this report, we demonstrate that the inhibition of topo IIalpha gene expression by wild-type p53 correlates with the decreased binding of the transcription factor NF-Y to the first four ICBs of the topo IIalpha promoter. The expression of mutant p53 does not affect the binding of NF-Y. In NIH3T3 cells, we show that topo II-targeted drugs inhibit the binding of NF-Y to ICB sites in the topo IIalpha promoter. This effect is seen not only with drugs that result in DNA strand breaks but also with drugs that inhibit the catalytic activity of topo II, and even with the mitotic spindle inhibitor, vinblastine. Further experiments with p53-null (10)1 cells treated with these same drugs also demonstrate decreased NF-Y binding to the topo IIalpha ICBs. The data presented points to the existence of both p53-dependent and -independent mechanisms for regulating NF-Y binding to ICBs in the topo IIalpha promoter and thus the modulation of topo IIalpha gene expression.


Assuntos
Antineoplásicos/farmacologia , Fator de Ligação a CCAAT/metabolismo , DNA Topoisomerases Tipo II/genética , Expressão Gênica/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Células 3T3 , Animais , Antígenos de Neoplasias , Sítios de Ligação , Fator de Ligação a CCAAT/efeitos dos fármacos , Proteínas de Ligação a DNA , Genes Supressores de Tumor , Humanos , Camundongos , Fatores de Transcrição/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA