Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Genes Dev ; 34(15-16): 1075-1088, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32616520

RESUMO

Nonsense-mediated decay (NMD) is a translation-dependent RNA quality control mechanism that occurs in the cytoplasm. However, it is unknown how NMD regulates the stability of RNAs translated at the endoplasmic reticulum (ER). Here, we identify a localized NMD pathway dedicated to ER-translated mRNAs. We previously identified NBAS, a component of the Syntaxin 18 complex involved in Golgi-to-ER trafficking, as a novel NMD factor. Furthermore, we show that NBAS fulfills an independent function in NMD. This ER-NMD pathway requires the interaction of NBAS with the core NMD factor UPF1, which is partially localized at the ER in the proximity of the translocon. NBAS and UPF1 coregulate the stability of ER-associated transcripts, in particular those associated with the cellular stress response. We propose a model where NBAS recruits UPF1 to the membrane of the ER and activates an ER-dedicated NMD pathway, thus providing an ER-protective function by ensuring quality control of ER-translated mRNAs.


Assuntos
Retículo Endoplasmático/metabolismo , Degradação do RNAm Mediada por Códon sem Sentido , Retículo Endoplasmático/enzimologia , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Biossíntese de Proteínas , RNA Helicases/metabolismo
2.
RNA ; 28(9): 1224-1238, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35768279

RESUMO

The DExD/H-box RNA helicase DHX34 is a nonsense-mediated decay (NMD) factor that together with core NMD factors coregulates NMD targets in nematodes and in vertebrates. Here, we show that DHX34 is also associated with the human spliceosomal catalytic C complex. Mapping of DHX34 endogenous binding sites using cross-linking immunoprecipitation (CLIP) revealed that DHX34 is preferentially associated with pre-mRNAs and locates at exon-intron boundaries. Accordingly, we observed that DHX34 regulates a large number of alternative splicing (AS) events in mammalian cells in culture, establishing a dual role for DHX34 in both NMD and pre-mRNA splicing. We previously showed that germline DHX34 mutations associated to familial myelodysplasia (MDS)/acute myeloid leukemia (AML) predisposition abrogate its activity in NMD. Interestingly, we observe now that DHX34 regulates the splicing of pre-mRNAs that have been linked to AML/MDS predisposition. This is consistent with silencing experiments in hematopoietic stem/progenitor cells (HSPCs) showing that loss of DHX34 results in differentiation blockade of both erythroid and myeloid lineages, which is a hallmark of AML development. Altogether, these data unveil new cellular functions of DHX34 and suggest that alterations in the levels and/or activity of DHX34 could contribute to human disease.


Assuntos
Leucemia Mieloide Aguda , Síndromes Mielodisplásicas , Processamento Alternativo , Animais , Humanos , Leucemia Mieloide Aguda/genética , Mamíferos/genética , Síndromes Mielodisplásicas/genética , Degradação do RNAm Mediada por Códon sem Sentido , RNA Helicases/genética , RNA Helicases/metabolismo , Precursores de RNA/genética , Precursores de RNA/metabolismo , Splicing de RNA , RNA Mensageiro/genética
3.
Nucleic Acids Res ; 44(4): 1483-95, 2016 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-26773057

RESUMO

The Nonsense-mediated mRNA decay (NMD) pathway selectively degrades mRNAs harboring premature termination codons (PTCs) but also regulates the abundance of a large number of cellular RNAs. The central role of NMD in the control of gene expression requires the existence of buffering mechanisms that tightly regulate the magnitude of this pathway. Here, we will focus on the mechanism of NMD with an emphasis on the role of RNA helicases in the transition from NMD complexes that recognize a PTC to those that promote mRNA decay. We will also review recent strategies aimed at uncovering novel trans-acting factors and their functional role in the NMD pathway. Finally, we will describe recent progress in the study of the physiological role of the NMD response.


Assuntos
Códon sem Sentido/genética , Regulação da Expressão Gênica/genética , Degradação do RNAm Mediada por Códon sem Sentido/genética , Estabilidade de RNA/genética , Humanos , Redes e Vias Metabólicas/genética , RNA Helicases/genética , RNA Mensageiro/genética
4.
EMBO Rep ; 16(1): 71-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25452588

RESUMO

Nonsense-mediated mRNA decay (NMD) is a surveillance mechanism that degrades mRNAs harboring premature termination codons (PTCs). We have conducted a genome-wide RNAi screen in Caenorhabditis elegans that resulted in the identification of five novel NMD genes that are conserved throughout evolution. Two of their human homologs, GNL2 (ngp-1) and SEC13 (npp-20), are also required for NMD in human cells. We also show that the C. elegans gene noah-2, which is present in Drosophila melanogaster but absent in humans, is an NMD factor in fruit flies. Altogether, these data identify novel NMD factors that are conserved throughout evolution, highlighting the complexity of the NMD pathway and suggesting that yet uncovered novel factors may act to regulate this process.


Assuntos
Caenorhabditis elegans/genética , Proteínas de Transporte/metabolismo , Drosophila melanogaster/genética , Proteínas de Ligação ao GTP/metabolismo , Degradação do RNAm Mediada por Códon sem Sentido/fisiologia , Proteínas Nucleares/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Transporte/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Proteínas do Ovo/genética , Proteínas do Ovo/metabolismo , Embrião não Mamífero , Evolução Molecular , Proteínas de Ligação ao GTP/genética , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Nucleares/genética , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
5.
Nucleic Acids Res ; 41(17): 8319-31, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23828042

RESUMO

The nonsense-mediated mRNA decay (NMD) pathway selectively degrades mRNAs harboring premature termination codons but also regulates the abundance of cellular RNAs. We sought to identify transcripts that are regulated by two novel NMD factors, DHX34 and neuroblastoma amplified sequence (NBAS), which were identified in a genome-wide RNA interference screen in Caenorhabditis elegans and later shown to mediate NMD in vertebrates. We performed microarray expression profile analysis in human cells, zebrafish embryos and C. elegans that were individually depleted of these factors. Our analysis revealed that a significant proportion of genes are co-regulated by DHX34, NBAS and core NMD factors in these three organisms. Further analysis indicates that NMD modulates cellular stress response pathways and membrane trafficking across species. Interestingly, transcripts encoding different NMD factors were sensitive to DHX34 and NBAS depletion, suggesting that these factors participate in a conserved NMD negative feedback regulatory loop, as was recently described for core NMD factors. In summary, we find that DHX34 and NBAS act in concert with core NMD factors to co-regulate a large number of endogenous RNA targets. Furthermore, the conservation of a mechanism to tightly control NMD homeostasis across different species highlights the importance of the NMD response in the control of gene expression.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Degradação do RNAm Mediada por Códon sem Sentido , RNA Helicases/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Evolução Molecular , Perfilação da Expressão Gênica , Células HeLa , Homeostase , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/fisiologia , RNA Helicases/antagonistas & inibidores , RNA Mensageiro/metabolismo , Transativadores/fisiologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/antagonistas & inibidores
6.
Nat Struct Mol Biol ; 14(2): 147-54, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17220898

RESUMO

Telomeres can fold into t-loops that may result from the invasion of the 3' overhang into duplex DNA. Their formation is facilitated in vitro by the telomeric protein TRF2, but very little is known regarding the mechanisms involved. Here we reveal that TRF2 generates positive supercoiling and condenses DNA. Using a variety of TRF2 mutants, we demonstrate a strong correlation between this topological activity and the ability to stimulate strand invasion. We also report that these properties require the combination of the TRF-homology (TRFH) domain of TRF2 with either its N- or C-terminal DNA-binding domains. We propose that TRF2 complexes, by constraining DNA around themselves in a right-handed conformation, can induce untwisting of the neighboring DNA, thereby favoring strand invasion. Implications of this topological model in t-loop formation and telomere homeostasis are discussed.


Assuntos
DNA/química , Telômero/química , Proteína 2 de Ligação a Repetições Teloméricas/química , DNA Super-Helicoidal/química , Humanos , Microscopia de Força Atômica , Conformação de Ácido Nucleico , Proteína 1 de Ligação a Repetições Teloméricas/química
7.
Elife ; 92020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33205750

RESUMO

Nonsense-mediated mRNA decay (NMD) is a surveillance pathway that degrades aberrant mRNAs and also regulates the expression of a wide range of physiological transcripts. RUVBL1 and RUVBL2 AAA-ATPases form an hetero-hexameric ring that is part of several macromolecular complexes such as INO80, SWR1, and R2TP. Interestingly, RUVBL1-RUVBL2 ATPase activity is required for NMD activation by an unknown mechanism. Here, we show that DHX34, an RNA helicase regulating NMD initiation, directly interacts with RUVBL1-RUVBL2 in vitro and in cells. Cryo-EM reveals that DHX34 induces extensive changes in the N-termini of every RUVBL2 subunit in the complex, stabilizing a conformation that does not bind nucleotide and thereby down-regulates ATP hydrolysis of the complex. Using ATPase-deficient mutants, we find that DHX34 acts exclusively on the RUVBL2 subunits. We propose a model, where DHX34 acts to couple RUVBL1-RUVBL2 ATPase activity to the assembly of factors required to initiate the NMD response.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas de Transporte/metabolismo , Microscopia Crioeletrônica , DNA Helicases/metabolismo , RNA Helicases/metabolismo , ATPases Associadas a Diversas Atividades Celulares/genética , Proteínas de Transporte/genética , Clonagem Molecular , DNA Helicases/genética , Regulação Enzimológica da Expressão Gênica , Células HEK293 , Humanos , RNA Helicases/genética
8.
Nat Commun ; 11(1): 1044, 2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-32098966

RESUMO

The inclusion of familial myeloid malignancies as a separate disease entity in the revised WHO classification has renewed efforts to improve the recognition and management of this group of at risk individuals. Here we report a cohort of 86 acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) families with 49 harboring germline variants in 16 previously defined loci (57%). Whole exome sequencing in a further 37 uncharacterized families (43%) allowed us to rationalize 65 new candidate loci, including genes mutated in rare hematological syndromes (ADA, GP6, IL17RA, PRF1 and SEC23B), reported in prior MDS/AML or inherited bone marrow failure series (DNAH9, NAPRT1 and SH2B3) or variants at novel loci (DHX34) that appear specific to inherited forms of myeloid malignancies. Altogether, our series of MDS/AML families offer novel insights into the etiology of myeloid malignancies and provide a framework to prioritize variants for inclusion into routine diagnostics and patient management.


Assuntos
Mutação em Linhagem Germinativa , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenosina Desaminase/genética , Dineínas do Axonema/genética , Estudos de Coortes , Humanos , Degradação do RNAm Mediada por Códon sem Sentido , Linhagem , Perforina/genética , Glicoproteínas da Membrana de Plaquetas/genética , RNA Helicases/genética , Receptores de Interleucina-17/genética , Proteínas de Transporte Vesicular/genética , Sequenciamento do Exoma
9.
Nucleic Acids Res ; 35(22): 7688-97, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17984081

RESUMO

The Smg proteins Smg5, Smg6 and Smg7 are involved in nonsense-mediated RNA decay (NMD) in metazoans, but no orthologs have been found in the budding yeast Saccharomyces cerevisiae. Sequence alignments reveal that yeast Ebs1p is similar in structure to the human Smg5-7, with highest homology to Smg7. We demonstrate here that Ebs1p is involved in NMD and behaves similarly to human Smg proteins. Indeed, both loss and overexpression of Ebs1p results in stabilization of NMD targets. However, Ebs1-loss in yeast or Smg7-depletion in human cells only partially disrupts NMD and in the latter, Smg7-depletion is partially compensated for by Smg6. Ebs1p physically interacts with the NMD helicase Upf1p and overexpressed Ebs1p leads to recruitment of Upf1p into cytoplasmic P-bodies. Furthermore, Ebs1p localizes to P-bodies upon glucose starvation along with Upf1p. Overall our findings suggest that NMD is more conserved in evolution than previously thought, and that at least one of the Smg5-7 proteins is conserved in budding yeast.


Assuntos
Códon sem Sentido , RNA Mensageiro/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Sequência de Aminoácidos , Proteínas de Transporte/química , Deleção de Genes , Regulação Fúngica da Expressão Gênica , Células HeLa , Humanos , Dados de Sequência Molecular , Mutação , Biossíntese de Proteínas , RNA Helicases/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Alinhamento de Sequência , Sirolimo/farmacologia , Telomerase/genética
10.
Nat Commun ; 8: 15114, 2017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28466845

RESUMO

MiRNA biogenesis is highly regulated at the post-transcriptional level; however, the role of sequence and secondary RNA structure in this process has not been extensively studied. A single G to A substitution present in the terminal loop of pri-mir-30c-1 in breast and gastric cancer patients had been previously described to result in increased levels of mature miRNA. Here, we report that this genetic variant directly affects Drosha-mediated processing of pri-mir-30c-1 in vitro and in cultured cells. Structural analysis of this variant revealed an altered RNA structure that facilitates the interaction with SRSF3, an SR protein family member that promotes pri-miRNA processing. Our results are compatible with a model whereby a genetic variant in pri-mir-30c-1 leads to a secondary RNA structure rearrangement that facilitates binding of SRSF3 resulting in increased levels of miR-30c. These data highlight that primary sequence determinants and RNA structure are key regulators of miRNA biogenesis.


Assuntos
Neoplasias da Mama/genética , MicroRNAs/genética , Processamento Pós-Transcricional do RNA/genética , Neoplasias da Mama/metabolismo , Feminino , Variação Genética , Células HEK293 , Humanos , Técnicas In Vitro , Células MCF-7 , MicroRNAs/metabolismo , Modelos Genéticos , Conformação de Ácido Nucleico , RNA , Ribonuclease III/metabolismo , Fatores de Processamento de Serina-Arginina/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
11.
Nat Commun ; 7: 10585, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26841701

RESUMO

Nonsense-mediated decay (NMD) is a messenger RNA quality-control pathway triggered by SMG1-mediated phosphorylation of the NMD factor UPF1. In recent times, the RNA helicase DHX34 was found to promote mRNP remodelling, leading to activation of NMD. Here we demonstrate the mechanism by which DHX34 functions in concert with SMG1. DHX34 comprises two distinct structural units, a core that binds UPF1 and a protruding carboxy-terminal domain (CTD) that binds the SMG1 kinase, as shown using truncated forms of DHX34 and electron microscopy of the SMG1-DHX34 complex. Truncation of the DHX34 CTD does not affect binding to UPF1; however, it compromises DHX34 binding to SMG1 to affect UPF1 phosphorylation and hence abrogate NMD. Altogether, these data suggest the existence of a complex comprising SMG1, UPF1 and DHX34, with DHX34 functioning as a scaffold for UPF1 and SMG1. This complex promotes UPF1 phosphorylation leading to functional NMD.


Assuntos
Degradação do RNAm Mediada por Códon sem Sentido , Fosfatidilinositol 3-Quinases/metabolismo , RNA Helicases/metabolismo , Transativadores/metabolismo , Células HEK293 , Células HeLa , Humanos , Imunoprecipitação , Técnicas In Vitro , Microscopia Eletrônica , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases
12.
Cell Rep ; 8(6): 1845-1856, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25220460

RESUMO

Nonsense-mediated decay (NMD) is a surveillance mechanism that degrades aberrant mRNAs. A complex comprising SMG1, UPF1, and the translation termination factors eRF1 and eRF3 (SURF) is assembled in the vicinity of a premature termination codon. Subsequently, an interaction with UPF2, UPF3b, and the exon junction complex induces the formation of the decay-inducing complex (DECID) and triggers NMD. We previously identified the RNA helicase DHX34 as an NMD factor in C. elegans and in vertebrates. Here, we investigate the mechanism by which DHX34 activates NMD in human cells. We show that DHX34 is recruited to the SURF complex via its preferential interaction with hypophosphorylated UPF1. A series of molecular transitions induced by DHX34 include enhanced recruitment of UPF2, increased UPF1 phosphorylation, and dissociation of eRF3 from UPF1. Thus, DHX34 promotes mRNP remodeling and triggers the conversion from the SURF complex to the DECID complex resulting in NMD activation.


Assuntos
Degradação do RNAm Mediada por Códon sem Sentido , Fatores de Terminação de Peptídeos/metabolismo , RNA Helicases/metabolismo , Células HEK293 , Humanos , Fatores de Terminação de Peptídeos/química , Fosforilação , Ligação Proteica , RNA Helicases/antagonistas & inibidores , RNA Helicases/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA , Ribonucleoproteínas/metabolismo , Transativadores/química , Transativadores/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
13.
Chromosoma ; 115(6): 413-25, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16741708

RESUMO

The physical ends of chromosomes, known as telomeres, protect chromosome ends from nucleolytic degradation and DNA repair activities. Conventional DNA replication enzymes lack the ability to fully replicate telomere ends. In addition, nucleolytic activities contribute to telomere erosion. Short telomeres trigger DNA damage checkpoints, which mediate cellular senescence. Telomere length homeostasis requires telomerase, a cellular reverse transcriptase, which uses an internal RNA moiety as a template for the synthesis of telomere repeats. Telomerase elongates the 3' ends of chromosomes, whereas the complementary strand is filled in by conventional DNA polymerases. In humans, telomerase is ubiquitously expressed only during the first weeks of embryogenesis, and is subsequently downregulated in most cell types. Correct telomere length setting is crucial for long-term survival. The telomere length reserve must be sufficient to avoid premature cellular senescence and the acceleration of age-related disease. On the other side, telomere shortening suppresses tumor formation through limiting the replicative potential of cells. In recent years, novel insight into the regulation of telomerase at chromosome ends has increased our understanding on how telomere length homeostasis in telomerase-positive cells is achieved. Factors that recruit telomerase to telomeres in a cell cycle-dependent manner have been identified in Saccharomyces cerevisiae. In humans, telomerase assembles with telomeres during S phase of the cell cycle. Presumably through mediating formation of alternative telomere structures, telomere-binding proteins regulate telomerase activity in cis to favor preferential elongation of the shortest telomeres. Phosphoinositide 3-kinase related kinases are also required for telomerase activation at chromosome ends, at least in budding and fission yeast. In vivo analysis of telomere elongation kinetics shows that telomerase does not act on every telomere in each cell cycle but that it exhibits an increasing preference for telomeres as their lengths decline. This suggests a model in which telomeres switch between extendible and nonextendible states in a length-dependent manner. In this review we expand this model to incorporate the finding that telomerase levels also limit telomere length and we propose a second switch between a non-telomerase-associated "extendible" and a telomerase-associated "extending" state.


Assuntos
Telomerase/metabolismo , Telômero/metabolismo , Animais , Ciclo Celular/fisiologia , Homeostase/genética , Humanos , Modelos Biológicos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Proteínas de Ligação a Telômeros/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa