Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Mol Cell ; 84(2): 359-374.e8, 2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38199006

RESUMO

Friedreich's ataxia (FA) is a debilitating, multisystemic disease caused by the depletion of frataxin (FXN), a mitochondrial iron-sulfur (Fe-S) cluster biogenesis factor. To understand the cellular pathogenesis of FA, we performed quantitative proteomics in FXN-deficient human cells. Nearly every annotated Fe-S cluster-containing protein was depleted, indicating that as a rule, cluster binding confers stability to Fe-S proteins. We also observed depletion of a small mitoribosomal assembly factor METTL17 and evidence of impaired mitochondrial translation. Using comparative sequence analysis, mutagenesis, biochemistry, and cryoelectron microscopy, we show that METTL17 binds to the mitoribosomal small subunit during late assembly and harbors a previously unrecognized [Fe4S4]2+ cluster required for its stability. METTL17 overexpression rescued the mitochondrial translation and bioenergetic defects, but not the cellular growth, of FXN-depleted cells. These findings suggest that METTL17 acts as an Fe-S cluster checkpoint, promoting translation of Fe-S cluster-rich oxidative phosphorylation (OXPHOS) proteins only when Fe-S cofactors are replete.


Assuntos
Ataxia de Friedreich , Proteínas Ferro-Enxofre , Humanos , Proteínas Ferro-Enxofre/genética , Proteínas Ferro-Enxofre/metabolismo , Microscopia Crioeletrônica , Frataxina , Biossíntese de Proteínas , Mitocôndrias/genética , Mitocôndrias/metabolismo , Ataxia de Friedreich/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo
2.
Nucleic Acids Res ; 49(D1): D1541-D1547, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33174596

RESUMO

The mammalian mitochondrial proteome is under dual genomic control, with 99% of proteins encoded by the nuclear genome and 13 originating from the mitochondrial DNA (mtDNA). We previously developed MitoCarta, a catalogue of over 1000 genes encoding the mammalian mitochondrial proteome. This catalogue was compiled using a Bayesian integration of multiple sequence features and experimental datasets, notably protein mass spectrometry of mitochondria isolated from fourteen murine tissues. Here, we introduce MitoCarta3.0. Beginning with the MitoCarta2.0 inventory, we performed manual review to remove 100 genes and introduce 78 additional genes, arriving at an updated inventory of 1136 human genes. We now include manually curated annotations of sub-mitochondrial localization (matrix, inner membrane, intermembrane space, outer membrane) as well as assignment to 149 hierarchical 'MitoPathways' spanning seven broad functional categories relevant to mitochondria. MitoCarta3.0, including sub-mitochondrial localization and MitoPathway annotations, is freely available at http://www.broadinstitute.org/mitocarta and should serve as a continued community resource for mitochondrial biology and medicine.


Assuntos
Bases de Dados de Proteínas , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Anotação de Sequência Molecular , Proteoma/metabolismo , Animais , Teorema de Bayes , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Conjuntos de Dados como Assunto , Humanos , Internet , Aprendizado de Máquina , Espectrometria de Massas , Camundongos , Mitocôndrias/genética , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/classificação , Proteínas Mitocondriais/genética , Proteoma/classificação , Proteoma/genética , Software
3.
Biochim Biophys Acta ; 1829(6-7): 624-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23500037

RESUMO

Nonsense-mediated mRNA decay (NMD) is an mRNA quality control mechanism that destabilizes aberrant mRNAs harboring premature termination (nonsense) codons (PTCs). Recent studies have shown that NMD also targets mRNAs transcribed from a large subset of wild-type genes. This raises the possibility that NMD itself is under regulatory control. Indeed, several recent studies have shown that NMD activity is modulated in specific cell types and that key components of the NMD pathway are regulated by several pathways, including microRNA circuits and NMD itself. Cellular stress also modulates the magnitude of NMD by mechanisms that are beginning to be understood. Here, we review the evidence that NMD is regulated and discuss the physiological role for this regulation. We propose that the efficiency of NMD is altered in some cellular contexts to regulate normal biological events. In disease states-such as in cancer-NMD is disturbed by intrinsic and extrinsic factors, resulting in altered levels of crucial NMD-targeted mRNAs that lead to downstream pathological consequences. This article is part of a Special Issue entitled: RNA Decay mechanisms.


Assuntos
MicroRNAs/genética , Degradação do RNAm Mediada por Códon sem Sentido/genética , RNA Mensageiro/genética , Estresse Fisiológico/genética , Códon sem Sentido , Humanos , Neoplasias/genética , Neoplasias/patologia , Splicing de RNA/genética
4.
Nature ; 446(7138): 926-9, 2007 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-17361132

RESUMO

The human and mouse genomes share a number of long, perfectly conserved nucleotide sequences, termed ultraconserved elements. Whereas these regions can act as transcriptional enhancers when upstream of genes, those within genes are less well understood. In particular, the function of ultraconserved elements that overlap alternatively spliced exons of genes encoding RNA-binding proteins is unknown. Here we report that in every member of the human SR family of splicing regulators, highly or ultraconserved elements are alternatively spliced, either as alternative 'poison cassette exons' containing early in-frame stop codons, or as alternative introns in the 3' untranslated region. These alternative splicing events target the resulting messenger RNAs for degradation by means of an RNA surveillance pathway called nonsense-mediated mRNA decay. Mouse orthologues of the human SR proteins exhibit the same unproductive splicing patterns. Three SR proteins have been previously shown to direct splicing of their own transcripts, and one of these is known to autoregulate its expression by coupling alternative splicing with decay; our results suggest that unproductive splicing is important for regulation of the entire SR family. We find that unproductive splicing associated with conserved regions has arisen independently in different SR genes, suggesting that splicing factors may readily acquire this form of regulation.


Assuntos
Processamento Alternativo/genética , Sequência Conservada/genética , DNA/genética , Proteínas de Ligação a RNA/genética , Animais , Éxons/genética , Humanos , Íntrons/genética , Camundongos , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/classificação
5.
J Biol Chem ; 286(46): 40038-43, 2011 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21969377

RESUMO

The Myc transcription factor plays a vital role in both normal cellular physiology and in many human cancers. We have recently demonstrated that nonsense-mediated RNA decay (NMD), a mechanism that rapidly degrades select mRNAs, is inhibited by the stress-induced phosphorylation of translation initiation factor eIF2α, and this inhibition stabilizes many transcripts necessary for tumorigenesis. Here, we demonstrate that NMD is inhibited by high Myc expression. We show that the phosphorylation of eIF2α, likely due to the ability of Myc to generate reactive oxygen species and augment endoplasmic reticulum stress, is necessary for the inhibition of NMD by Myc. The inhibition of NMD both stabilizes and up-regulates multiple Myc targets, suggesting that the inhibition of NMD may play an important role in the dynamic regulation of genes by Myc.


Assuntos
Linfócitos B/metabolismo , Regulação da Expressão Gênica/fisiologia , Degradação do RNAm Mediada por Códon sem Sentido/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Mensageiro/metabolismo , Linfócitos B/citologia , Linhagem Celular Tumoral , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico/fisiologia
6.
Blood Adv ; 3(14): 2199-2204, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31324640

RESUMO

Immune evasion is a hallmark of cancer and a central mechanism underlying acquired resistance to immune therapy. In allogeneic hematopoietic cell transplantation (alloHCT), late relapses can arise after prolonged alloreactive T-cell control, but the molecular mechanisms of immune escape remain unclear. To identify mechanisms of immune evasion, we performed a genetic analysis of serial samples from 25 patients with myeloid malignancies who relapsed ≥1 year after alloHCT. Using targeted sequencing and microarray analysis to determine HLA allele-specific copy number, we identified copy-neutral loss of heterozygosity events and focal deletions spanning class 1 HLA genes in 2 of 12 recipients of matched unrelated-donor HCT and in 1 of 4 recipients of mismatched unrelated-donor HCT. Relapsed clones, although highly related to their antecedent pretransplantation malignancies, frequently acquired additional mutations in transcription factors and mitogenic signaling genes. Previously, the study of relapse after haploidentical HCT established the paradigm of immune evasion via loss of mismatched HLA. Here, in the context of matched unrelated-donor HCT, HLA loss provides genetic evidence that allogeneic immune recognition may be mediated by minor histocompatibility antigens and suggests opportunities for novel immunologic approaches for relapse prevention.


Assuntos
Deleção de Genes , Antígenos HLA/genética , Transplante de Células-Tronco Hematopoéticas , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Alelos , Biomarcadores , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Leucemia Mieloide Aguda/terapia , Mutação , Polimorfismo de Nucleotídeo Único , Recidiva , Transplante Homólogo
7.
Cell Cycle ; 14(16): 2571-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26039820

RESUMO

The hostile tumor microenvironment results in the generation of intracellular stresses including hypoxia and nutrient deprivation. In order to adapt to such conditions, the cell utilizes several stress-response mechanisms, including the attenuation of protein synthesis, the inhibition of cellular proliferation, and induction of autophagy. Autophagy leads to the degradation of cellular contents, including damaged organelles and mutant proteins, which the cell can then use as an alternate energy source. Two integral changes to the signaling milieu to promote such a response include inhibition of the mammalian target of rapamycin complex 1 (mTORC1) and phosphorylation of eIF2α. This review will describe how conditions found in the tumor microenvironment regulate mTORC1 as well as eIF2α, the downstream impact of these modifications, and the implications in tumorigenesis. We will then discuss the remarkable similarities and overlapping function of these 2 signaling pathways, focusing on the response to amino acid deprivation, and present a new model involving crosstalk between them based on our recent work.


Assuntos
Fator de Iniciação 2 em Eucariotos/fisiologia , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Adaptação Fisiológica , Autofagia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Fosforilação , Processamento de Proteína Pós-Traducional , Receptor Cross-Talk , Transdução de Sinais , Resposta a Proteínas não Dobradas
8.
Sci Signal ; 8(367): ra27, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25759478

RESUMO

Amino acid deprivation promotes the inhibition of the kinase complex mTORC1 (mammalian target of rapamycin complex 1) and activation of the kinase GCN2 (general control nonrepressed 2). Signaling pathways downstream of both kinases have been thought to independently induce autophagy. We showed that these two amino acid-sensing systems are linked. We showed that pharmacological inhibition of mTORC1 led to activation of GCN2 and phosphorylation of the eukaryotic initiation factor 2α (eIF2α) in a mechanism dependent on the catalytic subunit of protein phosphatase 6 (PP6C). Autophagy induced by pharmacological inhibition of mTORC1 required PP6C, GCN2, and eIF2α phosphorylation. Although some of the PP6C mutants found in melanoma did not form a strong complex with PP6 regulatory subunits and were rapidly degraded, these mutants paradoxically stabilized PP6C encoded by the wild-type allele and increased eIF2α phosphorylation. Furthermore, these PP6C mutations were associated with increased autophagy in vitro and in human melanoma samples. Thus, these data showed that GCN2 activation and phosphorylation of eIF2α in response to mTORC1 inhibition are necessary for autophagy. Additionally, we described a role for PP6C in this process and provided a mechanism for PP6C mutations associated with melanoma.


Assuntos
Autofagia/fisiologia , Ativação Enzimática/fisiologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Melanoma/genética , Melanoma/fisiopatologia , Complexos Multiproteicos/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Aminoácidos/deficiência , Linhagem Celular Tumoral , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Ativação Enzimática/efeitos dos fármacos , Técnicas de Introdução de Genes , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Espectrometria de Massas , Alvo Mecanístico do Complexo 1 de Rapamicina , Microscopia de Fluorescência , Mutação/genética , Fosfoproteínas Fosfatases/genética , Fosforilação/efeitos dos fármacos , Sirolimo/farmacologia , Tunicamicina/farmacologia
9.
Mol Cancer Res ; 12(3): 433-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24336958

RESUMO

UNLABELLED: Recent whole genome melanoma sequencing studies have identified recurrent mutations in the gene encoding the catalytic subunit of serine/threonine phosphatase 6 (PPP6C/PP6C). However, the biochemical, functional, and clinical ramifications of these mutations are unknown. Sequencing PP6C from patients with melanoma (233 primary and 77 metastatic specimens) with extended prospective clinical outcome revealed a large number of hotspot mutations in patients with both primary and metastatic melanoma. Despite minimal association between stage and presence of PP6C mutations in patients with primary melanoma, a subpopulation of cells within each tumor did contain PP6C mutations, suggesting PP6C mutation is an early, but non-tumor-initiating event in melanoma. Among patients with primary melanoma with PP6C mutations, patients with stop mutations had significantly shorter recurrence-free survival compared with patients without stop mutations. In addition, PP6C mutations were independent of commonly observed BRAF and NRAS mutations. Biochemically, PP6C mutations could be classified as those that interact with PP6C regulatory subunits and those that do not. Mutations that did not bind to PP6C regulatory subunits were associated with increased phosphorylation of Aurora kinase, a PP6C substrate, and mitotic defects. However, both classes of PP6C mutations led to increased sensitivity to Aurora kinase inhibition. Together, these data support for the first time that PP6C mutations are molecularly, biochemically, and clinically heterogeneous. IMPLICATIONS: PP6C mutations have distinct functional and clinical consequences in melanoma, and confer sensitivity to Aurora A kinase inhibitors.


Assuntos
Aurora Quinases/antagonistas & inibidores , Melanoma/enzimologia , Melanoma/genética , Mutação , Fosfoproteínas Fosfatases/genética , Neoplasias Cutâneas/enzimologia , Neoplasias Cutâneas/genética , Aurora Quinases/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Melanoma/tratamento farmacológico , Melanoma/patologia , Estadiamento de Neoplasias , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/metabolismo , Prognóstico , Inibidores de Proteínas Quinases , Subunidades Proteicas , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia
10.
Mol Cell Biol ; 33(11): 2128-35, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23508110

RESUMO

Nonsense-mediated RNA decay (NMD) is an mRNA surveillance mechanism which rapidly degrades select cytoplasmic mRNAs. We and others have shown that NMD is a dynamically regulated process inhibited by amino acid deprivation, hypoxia, and other cellular stresses commonly generated by the tumor microenvironment. This inhibition of NMD can result in the accumulation of misfolded, mutated, and aggregated proteins, but how cells adapt to these aberrant proteins is unknown. Here we demonstrate that the inhibition of NMD activates autophagy, an established protein surveillance mechanism, both in vitro and in vivo. Conversely, the hyperactivation of NMD blunts the induction of autophagy in response to a variety of cellular stresses. The regulation of autophagy by NMD is due, in part, to stabilization of the documented NMD target ATF4. NMD inhibition increases intracellular amino acids, a hallmark of autophagy, and the concomitant inhibition of autophagy and NMD, either molecularly or pharmacologically, leads to synergistic cell death. Together these studies indicate that autophagy is an adaptive response to NMD inhibition and uncover a novel relationship between an mRNA surveillance system and a protein surveillance system, with important implications for the treatment of cancer.


Assuntos
Autofagia/genética , Degradação do RNAm Mediada por Códon sem Sentido , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Aminoácidos/metabolismo , Animais , Autofagia/efeitos dos fármacos , Morte Celular/genética , Cloroquina/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa/efeitos dos fármacos , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/imunologia , RNA Helicases , RNA Interferente Pequeno , Proteínas de Ligação a RNA , Transativadores/genética , Fatores de Transcrição/genética
11.
J Nucl Med ; 51(7): 1092-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20554721

RESUMO

UNLABELLED: Deoxycytidine kinase (dCK) is a rate-limiting enzyme in the deoxyribonucleoside salvage pathway and a critical determinant of therapeutic activity for several nucleoside analog prodrugs. We have previously reported the development of 1-(2'-deoxy-2'-(18)F-fluoro-beta-D-arabinofuranosyl)cytosine ((18)F-FAC), a new probe for PET of dCK activity in immune disorders and certain cancers. The objective of the current study was to develop PET probes with improved metabolic stability and specificity for dCK. Toward this goal, several candidate PET probes were synthesized and evaluated in vitro and in vivo. METHODS: High-pressure liquid chromatography was used to analyze the metabolic stability of (18)F-FAC and several newly synthesized analogs with the natural D-enantiomeric sugar configuration or the corresponding unnatural L-configuration. In vitro kinase and uptake assays were used to determine the affinity of the (18)F-FAC L-nucleoside analogs for dCK. The biodistribution of selected L-analogs in mice was determined by small-animal PET/CT. RESULTS: Candidate PET probes were selected using the following criteria: low susceptibility to deamination, high affinity for purified recombinant dCK, high uptake in dCK-expressing cell lines, and biodistribution in mice reflective of the tissue-expression pattern of dCK. Among the 10 newly developed candidate probes, 1-(2'-deoxy-2'-(18)F-fluoro-beta-L-arabinofuranosyl)cytosine (L-(18)F-FAC) and 1-(2'-deoxy-2'-(18)F-fluoro-beta-L-arabinofuranosyl)-5-methylcytosine (L-(18)F-FMAC) most closely matched the selection criteria. The selection of L-(18)F-FAC and L-(18)F-FMAC was validated by showing that these two PET probes could be used to image animal models of leukemia and autoimmunity. CONCLUSION: Promising in vitro and in vivo data warrant biodistribution and dosimetry studies of L-(18)F-FAC and L-(18)F-FMAC in humans.


Assuntos
Desoxicitidina Quinase/metabolismo , Compostos Radiofarmacêuticos , Nucleotídeos de Adenina , Animais , Arabinonucleosídeos , Ligação Competitiva , Biotransformação , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Clofarabina , Humanos , L-Lactato Desidrogenase/metabolismo , Transtornos Linfoproliferativos/diagnóstico por imagem , Camundongos , Fosforilação , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Proteínas Recombinantes , Especificidade por Substrato , Tomografia Computadorizada de Emissão , Imagem Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA