Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 595(7865): 120-124, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34079125

RESUMEN

Compartmentalization is a defining characteristic of eukaryotic cells, and partitions distinct biochemical processes into discrete subcellular locations. Microscopy1 and biochemical fractionation coupled with mass spectrometry2-4 have defined the proteomes of a variety of different organelles, but many intracellular compartments have remained refractory to such approaches. Proximity-dependent biotinylation techniques such as BioID provide an alternative approach to define the composition of cellular compartments in living cells5-7. Here we present a BioID-based map of a human cell on the basis of 192 subcellular markers, and define the intracellular locations of 4,145 unique proteins in HEK293 cells. Our localization predictions exceed the specificity of previous approaches, and enabled the discovery of proteins at the interface between the mitochondrial outer membrane and the endoplasmic reticulum that are crucial for mitochondrial homeostasis. On the basis of this dataset, we created humancellmap.org as a community resource that provides online tools for localization analysis of user BioID data, and demonstrate how this resource can be used to understand BioID results better.


Asunto(s)
Biotinilación , Compartimento Celular , Transporte de Proteínas , Proteoma/análisis , Proteoma/química , Células Cultivadas , Conjuntos de Datos como Asunto , Retículo Endoplásmico/química , Retículo Endoplásmico/metabolismo , Células HEK293 , Células HeLa , Homeostasis , Humanos , Espectrometría de Masas , Mitocondrias/química , Mitocondrias/metabolismo , Orgánulos/química , Orgánulos/metabolismo , Proteoma/metabolismo , Reproducibilidad de los Resultados
2.
Nucleic Acids Res ; 51(21): 11797-11812, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-37823603

RESUMEN

The human mitochondrial ribosome contains three [2Fe-2S] clusters whose assembly pathway, role, and implications for mitochondrial and metabolic diseases are unknown. Here, structure-function correlation studies show that the clusters play a structural role during mitoribosome assembly. To uncover the assembly pathway, we have examined the effect of silencing the expression of Fe-S cluster biosynthetic and delivery factors on mitoribosome stability. We find that the mitoribosome receives its [2Fe-2S] clusters from the GLRX5-BOLA3 node. Additionally, the assembly of the small subunit depends on the mitoribosome biogenesis factor METTL17, recently reported containing a [4Fe-4S] cluster, which we propose is inserted via the ISCA1-NFU1 node. Consistently, fibroblasts from subjects suffering from 'multiple mitochondrial dysfunction' syndrome due to mutations in BOLA3 or NFU1 display previously unrecognized attenuation of mitochondrial protein synthesis that contributes to their cellular and pathophysiological phenotypes. Finally, we report that, in addition to their structural role, one of the mitoribosomal [2Fe-2S] clusters and the [4Fe-4S] cluster in mitoribosome assembly factor METTL17 sense changes in the redox environment, thus providing a way to regulate organellar protein synthesis accordingly.


Asunto(s)
Proteínas Hierro-Azufre , Enfermedades Mitocondriales , Ribosomas Mitocondriales , Humanos , Proteínas Portadoras/metabolismo , Hierro/metabolismo , Proteínas Hierro-Azufre/química , Metiltransferasas/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Ribosomas Mitocondriales/metabolismo , Azufre/metabolismo , Enfermedades Mitocondriales/metabolismo
4.
Nucleic Acids Res ; 48(14): 7924-7943, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32652011

RESUMEN

Biogenesis of mammalian mitochondrial ribosomes (mitoribosomes) involves several conserved small GTPases. Here, we report that the Obg family protein GTPBP5 or MTG2 is a mitochondrial protein whose absence in a TALEN-induced HEK293T knockout (KO) cell line leads to severely decreased levels of the 55S monosome and attenuated mitochondrial protein synthesis. We show that a fraction of GTPBP5 co-sediments with the large mitoribosome subunit (mtLSU), and crosslinks specifically with the 16S rRNA, and several mtLSU proteins and assembly factors. Notably, the latter group includes MTERF4, involved in monosome assembly, and MRM2, the methyltransferase that catalyzes the modification of the 16S mt-rRNA A-loop U1369 residue. The GTPBP5 interaction with MRM2 was also detected using the proximity-dependent biotinylation (BioID) assay. In GTPBP5-KO mitochondria, the mtLSU lacks bL36m, accumulates an excess of the assembly factors MTG1, GTPBP10, MALSU1 and MTERF4, and contains hypomethylated 16S rRNA. We propose that GTPBP5 primarily fuels proper mtLSU maturation by securing efficient methylation of two 16S rRNA residues, and ultimately serves to coordinate subunit joining through the release of late-stage mtLSU assembly factors. In this way, GTPBP5 provides an ultimate quality control checkpoint function during mtLSU assembly that minimizes premature subunit joining to ensure the assembly of the mature 55S monosome.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Ribosomas Mitocondriales/enzimología , Proteínas de Unión al GTP Monoméricas/metabolismo , ARN Ribosómico 16S/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/enzimología , Línea Celular , GTP Fosfohidrolasas/metabolismo , Células HEK293 , Humanos , Metilación , Metiltransferasas/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/fisiología , Ribosomas Mitocondriales/metabolismo , Proteínas de Unión al GTP Monoméricas/fisiología , Fosforilación Oxidativa , Biosíntesis de Proteínas , ARN Ribosómico 16S/química , Subunidades Ribosómicas Grandes de Eucariotas/química , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Factores de Transcripción/metabolismo
5.
EMBO Rep ; 18(1): 28-38, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27974379

RESUMEN

Pseudouridylation is a common post-transcriptional modification in RNA, but its functional consequences at the cellular level remain largely unknown. Using a proximity-biotinylation assay, we identified a protein module in mitochondrial RNA granules, platforms for post-transcriptional RNA modification and ribosome assembly, containing several proteins of unknown function including three uncharacterized pseudouridine synthases, TRUB2, RPUSD3, and RPUSD4. TRUB2 and RPUSD4 were previously identified as core essential genes in CRISPR/Cas9 screens. Depletion of the individual enzymes produced specific mitochondrial protein synthesis and oxidative phosphorylation assembly defects without affecting mitochondrial mRNA levels. Investigation of the molecular targets in mitochondrial RNA by pseudouridine-Seq showed that RPUSD4 plays a role in the pseudouridylation of a single residue in the 16S rRNA, a modification that is essential for its stability and assembly into the mitochondrial ribosome, while TRUB2/RPUSD3 were similarly involved in pseudouridylating specific residues in mitochondrial mRNAs. These results establish essential roles for epitranscriptomic modification of mitochondrial RNA in mitochondrial protein synthesis, oxidative phosphorylation, and cell survival.


Asunto(s)
Supervivencia Celular , Transferasas Intramoleculares/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/biosíntesis , Proteínas Portadoras , Línea Celular , Humanos , Unión Proteica , Transporte de Proteínas , ARN/genética , ARN/metabolismo , Transporte de ARN , ARN Mitocondrial , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Ribosomas/metabolismo
6.
Hum Mol Genet ; 24(2): 480-91, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25214534

RESUMEN

French Canadian Leigh Syndrome (LSFC) is an early-onset, progressive neurodegenerative disorder with a distinct pattern of tissue involvement. Most cases are caused by a founder missense mutation in LRPPRC. LRPPRC forms a ribonucleoprotein complex with SLIRP, another RNA-binding protein, and this stabilizes polyadenylated mitochondrial mRNAs. LSFC fibroblasts have reduced levels of LRPPRC and a specific complex IV assembly defect; however, further depletion of mutant LRPPRC results in a complete failure to assemble a functional oxidative phosphorylation system, suggesting that LRPPRC levels determine the nature of the biochemical phenotype. We tested this hypothesis in cultured muscle cells and tissues from LSFC patients. LRPPRC levels were reduced in LSFC muscle cells, resulting in combined complex I and IV deficiencies. A similar combined deficiency was observed in skeletal muscle. Complex IV was only moderately reduced in LSFC heart, but was almost undetectable in liver. Both of these tissues showed elevated levels of complexes I and III. Despite the marked biochemical differences, the steady-state levels of LRPPRC and mitochondrial mRNAs were extremely low, LRPPRC was largely detergent-insoluble, and SLIRP was undetectable in all LSFC tissues. The level of the LRPPRC/SLIRP complex appeared much reduced in control tissues by the first dimension blue-native polyacrylamide gel electrophoresis (BN-PAGE) analysis compared with fibroblasts, and even by second dimension analysis it was virtually undetectable in control heart. These results point to tissue-specific pathways for the post-transcriptional handling of mitochondrial mRNAs and suggest that the biochemical defects in LSFC reflect the differential ability of tissues to adapt to the mutation.


Asunto(s)
Enfermedad de Leigh/metabolismo , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células Cultivadas , Humanos , Enfermedad de Leigh/genética , Hígado/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Especificidad de Órganos , Fosforilación Oxidativa , Unión Proteica , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo
7.
Hum Mol Genet ; 24(14): 4103-13, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25911677

RESUMEN

Protein synthesis in mitochondria is initiated by formylmethionyl-tRNA(Met) (fMet-tRNA(Met)), which requires the activity of the enzyme MTFMT to formylate the methionyl group. We investigated the molecular consequences of mutations in MTFMT in patients with Leigh syndrome or cardiomyopathy. All patients studied were compound heterozygotes. Levels of MTFMT in patient fibroblasts were almost undetectable by immunoblot analysis, and BN-PAGE analysis showed a combined oxidative phosphorylation (OXPHOS) assembly defect involving complexes I, IV and V. The synthesis of only a subset of mitochondrial polypeptides (ND5, ND4, ND1, COXII) was decreased, whereas all others were translated at normal or even increased rates. Expression of the wild-type cDNA rescued the biochemical phenotype when MTFMT was expressed near control levels, but overexpression produced a dominant-negative phenotype, completely abrogating assembly of the OXPHOS complexes, suggesting that MTFMT activity must be tightly regulated. fMet-tRNA(Met) was almost undetectable in control cells and absent in patient cells by high-resolution northern blot analysis, but accumulated in cells overexpressing MTFMT. Newly synthesized COXI was under-represented in complex IV immunoprecipitates from patient fibroblasts, and two-dimensional BN-PAGE analysis of newly synthesized mitochondrial translation products showed an accumulation of free COXI. Quantitative mass spectrophotometry of an N-terminal COXI peptide showed that the ratio of formylated to unmodified N-termini in the assembled complex IV was ∼350:1 in controls and 4:1 in patient cells. These results show that mitochondrial protein synthesis can occur with inefficient formylation of methionyl-tRNA(Met), but that assembly of complex IV is impaired if the COXI N-terminus is not formylated.


Asunto(s)
Ciclooxigenasa 1/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Metionina/química , Células Cultivadas , Cromatografía Liquida , Ciclooxigenasa 1/genética , Complejo IV de Transporte de Electrones/genética , Exoma , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Silenciador del Gen , Heterocigoto , Humanos , Enfermedad de Leigh/genética , Mitocondrias/metabolismo , Mutación , Fosforilación Oxidativa , Biosíntesis de Proteínas , ARN de Transferencia de Metionina/genética , ARN de Transferencia de Metionina/metabolismo , Análisis de Secuencia de ADN , Espectrometría de Masas en Tándem
8.
Hum Mol Genet ; 24(10): 2841-7, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25652405

RESUMEN

Addition of the trinucleotide cytosine/cytosine/adenine (CCA) to the 3' end of transfer RNAs (tRNAs) is essential for translation and is catalyzed by the enzyme TRNT1 (tRNA nucleotidyl transferase), which functions in both the cytoplasm and mitochondria. Exome sequencing revealed TRNT1 mutations in two unrelated subjects with different clinical features. The first presented with acute lactic acidosis at 3 weeks of age and developed severe developmental delay, hypotonia, microcephaly, seizures, progressive cortical atrophy, neurosensorial deafness, sideroblastic anemia and renal Fanconi syndrome, dying at 21 months. The second presented at 3.5 years with gait ataxia, dysarthria, gross motor regression, hypotonia, ptosis and ophthalmoplegia and had abnormal signals in brainstem and dentate nucleus. In subject 1, muscle biopsy showed combined oxidative phosphorylation (OXPHOS) defects, but there was no OXPHOS deficiency in fibroblasts from either subject, despite a 10-fold-reduction in TRNT1 protein levels in fibroblasts of the first subject. Furthermore, in normal controls, TRNT1 protein levels are 10-fold lower in muscle than in fibroblasts. High resolution northern blots of subject fibroblast RNA suggested incomplete CCA addition to the non-canonical mitochondrial tRNA(Ser(AGY)), but no obvious qualitative differences in other mitochondrial or cytoplasmic tRNAs. Complete knockdown of TRNT1 in patient fibroblasts rendered mitochondrial tRNA(Ser(AGY)) undetectable, and markedly reduced mitochondrial translation, except polypeptides lacking Ser(AGY) codons. These data suggest that the clinical phenotypes associated with TRNT1 mutations are largely due to impaired mitochondrial translation, resulting from defective CCA addition to mitochondrial tRNA(Ser(AGY)), and that the severity of this biochemical phenotype determines the severity and tissue distribution of clinical features.


Asunto(s)
Mitocondrias/genética , Enfermedades Mitocondriales/genética , Mutación , Biosíntesis de Proteínas/genética , ARN Nucleotidiltransferasas/genética , ARN de Transferencia de Serina/metabolismo , Niño , Preescolar , Exoma , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Mitocondrias/metabolismo , ARN Nucleotidiltransferasas/metabolismo , Análisis de Secuencia de ADN , Síndrome
9.
Hum Genet ; 134(9): 981-91, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26099313

RESUMEN

Leigh syndrome (LS) is a rare heterogeneous progressive neurodegenerative disorder usually presenting in infancy or early childhood. Clinical presentation is variable and includes psychomotor delay or regression, acute neurological or acidotic episodes, hypotonia, ataxia, spasticity, movement disorders, and corresponding anomalies of the basal ganglia and brain stem on magnetic resonance imaging. To date, 35 genes have been associated with LS, mostly involved in mitochondrial respiratory chain function and encoded in either nuclear or mitochondrial DNA. We used whole-exome sequencing to identify disease-causing variants in four patients with basal ganglia abnormalities and clinical presentations consistent with LS. Compound heterozygote variants in ECHS1, encoding the enzyme enoyl-CoA hydratase were identified. One missense variant (p.Thr180Ala) was common to all four patients and the haplotype surrounding this variant was also shared, suggesting a common ancestor of French-Canadian origin. Rare mutations in ECHS1 as well as in HIBCH, the enzyme downstream in the valine degradation pathway, have been associated with LS or LS-like disorders. A clear clinical overlap is observed between our patients and the reported cases with ECHS1 or HIBCH deficiency. The main clinical features observed in our cohort are T2-hyperintense signal in the globus pallidus and putamen, failure to thrive, developmental delay or regression, and nystagmus. Respiratory chain studies are not strikingly abnormal in our patients: one patient had a mild reduction of complex I and III and another of complex IV. The identification of four additional patients with mutations in ECHS1 highlights the emerging importance of this pathway in LS.


Asunto(s)
Enoil-CoA Hidratasa/genética , Exoma , Enfermedad de Leigh/genética , Anomalías Múltiples/sangre , Errores Innatos del Metabolismo de los Aminoácidos/sangre , Canadá , Preescolar , Análisis Mutacional de ADN , Femenino , Haplotipos , Heterocigoto , Humanos , Lactante , Enfermedad de Leigh/diagnóstico , Imagen por Resonancia Magnética , Masculino , Mutación , Linaje , Tioléster Hidrolasas/sangre , Tioléster Hidrolasas/deficiencia
10.
Am J Hum Genet ; 90(1): 142-51, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22243966

RESUMEN

We investigated a family in which the index subject presented with severe congenital lactic acidosis and dysmorphic features associated with a cytochrome c oxidase (COX)-assembly defect and a specific decrease in the synthesis of COX I, the subunit that nucleates COX assembly. Using a combination of microcell-mediated chromosome transfer, homozygosity mapping, and transcript profiling, we mapped the gene defect to chromosome 12 and identified a homozygous missense mutation (c.88G>A) in C12orf62. C12orf62 was not detectable by immunoblot analysis in subject fibroblasts, and retroviral expression of the wild-type C12orf62 cDNA rescued the biochemical phenotype. Furthermore, siRNA-mediated knockdown of C12orf 62 recapitulated the biochemical defect in control cells and exacerbated it in subject cells. C12orf62 is apparently restricted to the vertebrate lineage. It codes for a very small (6 kDa), uncharacterized, single-transmembrane protein that localizes to mitochondria and elutes in a complex of ∼110 kDa by gel filtration. COX I, II, and IV coimmunoprecipated with an epitope-tagged version of C12orf62, and 2D blue-native-polyacrylamide-gel-electrophoresis analysis of newly synthesized mitochondrial COX subunits in subject fibroblasts showed that COX assembly was impaired and that the nascent enzyme complex was unstable. We conclude that C12orf62 is required for coordination of the early steps of COX assembly with the synthesis of COX I.


Asunto(s)
Acidosis Láctica/genética , Ciclooxigenasa 1/biosíntesis , Complejo IV de Transporte de Electrones/biosíntesis , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Mutación Missense , Resultado Fatal , Femenino , Fibroblastos/enzimología , Homocigoto , Humanos , Recién Nacido , Mitocondrias/enzimología , Mitocondrias/genética
11.
Am J Hum Genet ; 91(4): 737-43, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-23022098

RESUMEN

Mutations in the genes composing the mitochondrial translation apparatus are an important cause of a heterogeneous group of oxidative phosphorylation (OXPHOS) disorders. We studied the index case in a consanguineous family in which two children presented with severe encephalopathy, lactic acidosis, and intractable seizures leading to an early fatal outcome. Blue native polyacrylamide gel electrophoretic (BN-PAGE) analysis showed assembly defects in all of the OXPHOS complexes with mtDNA-encoded structural subunits, and these defects were associated with a severe deficiency in mitochondrial translation. Immunoblot analysis showed reductions in the steady-state levels of several structural subunits of the mitochondrial ribosome. Whole-exome sequencing identified a homozygous missense mutation (c.1250G>A) in an uncharacterized gene, RMND1 (required for meiotic nuclear division 1). RMND1 localizes to mitochondria and behaves as an integral membrane protein. Retroviral expression of the wild-type RMND1 cDNA rescued the biochemical phenotype in subject cells, and siRNA-mediated knockdown of the protein recapitulated the defect. BN-PAGE, gel filtration, and mass spectrometry analyses showed that RMND1 forms a high-molecular-weight and most likely homopolymeric complex (∼240 kDa) that does not assemble in subject fibroblasts but that is rescued by expression of RMND1 cDNA. The p.Arg417Gln substitution, predicted to be in a coiled-coil domain, which is juxtaposed to a transmembrane domain at the extreme C terminus of the protein, does not alter the steady-state level of RMND1 but might prevent protein-protein interactions in this complex. Our results demonstrate that the RMND1 complex is necessary for mitochondrial translation, possibly by coordinating the assembly or maintenance of the mitochondrial ribosome.


Asunto(s)
Proteínas de Ciclo Celular/genética , Discapacidad Intelectual/genética , Mitocondrias/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Mutación Missense , Biosíntesis de Proteínas , Espasmos Infantiles/genética , Consanguinidad , ADN Mitocondrial/genética , Exoma , Femenino , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Recién Nacido , Síndrome de Lennox-Gastaut , Proteínas de la Membrana/genética , Fenotipo , Dominios y Motivos de Interacción de Proteínas/genética , Ribosomas/genética
12.
Nat Genet ; 38(1): 93-100, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16311595

RESUMEN

Methylmalonic aciduria and homocystinuria, cblC type (OMIM 277400), is the most common inborn error of vitamin B(12) (cobalamin) metabolism, with about 250 known cases. Affected individuals have developmental, hematological, neurological, metabolic, ophthalmologic and dermatologic clinical findings. Although considered a disease of infancy or childhood, some individuals develop symptoms in adulthood. The cblC locus was mapped to chromosome region 1p by linkage analysis. We refined the chromosomal interval using homozygosity mapping and haplotype analyses and identified the MMACHC gene. In 204 individuals, 42 different mutations were identified, many consistent with a loss of function of the protein product. One mutation, 271dupA, accounted for 40% of all disease alleles. Transduction of wild-type MMACHC into immortalized cblC fibroblast cell lines corrected the cellular phenotype. Molecular modeling predicts that the C-terminal region of the gene product folds similarly to TonB, a bacterial protein involved in energy transduction for cobalamin uptake.


Asunto(s)
Proteínas Portadoras/genética , Homocistinuria/genética , Errores Innatos del Metabolismo/genética , Ácido Metilmalónico/orina , Mutación , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Línea Celular , Mapeo Cromosómico , Secuencia Conservada , Fibroblastos/metabolismo , Haplotipos/genética , Humanos , Proteínas de la Membrana/química , Datos de Secuencia Molecular , Oxidorreductasas , Pliegue de Proteína , Homología Estructural de Proteína , Vitamina B 12/metabolismo
13.
Life Sci Alliance ; 7(1)2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37931956

RESUMEN

Mitochondria interact with the ER at structurally and functionally specialized membrane contact sites known as mitochondria-ER contact sites (MERCs). Combining proximity labelling (BioID), co-immunoprecipitation, confocal microscopy and subcellular fractionation, we found that the ER resident SMP-domain protein ESYT1 was enriched at MERCs, where it forms a complex with the outer mitochondrial membrane protein SYNJ2BP. BioID analyses using ER-targeted, outer mitochondrial membrane-targeted, and MERC-targeted baits, confirmed the presence of this complex at MERCs and the specificity of the interaction. Deletion of ESYT1 or SYNJ2BP reduced the number and length of MERCs. Loss of the ESYT1-SYNJ2BP complex impaired ER to mitochondria calcium flux and provoked a significant alteration of the mitochondrial lipidome, most prominently a reduction of cardiolipins and phosphatidylethanolamines. Both phenotypes were rescued by reexpression of WT ESYT1 and an artificial mitochondria-ER tether. Together, these results reveal a novel function for ESYT1 in mitochondrial and cellular homeostasis through its role in the regulation of MERCs.


Asunto(s)
Calcio , Retículo Endoplásmico , Mitocondrias , Sinaptotagminas , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Homeostasis , Lípidos , Mitocondrias/metabolismo , Sinaptotagminas/metabolismo
14.
Hum Mutat ; 34(10): 1366-70, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23878101

RESUMEN

Isolated cytochrome c oxidase (COX) deficiency is a common cause of mitochondrial disease, yet its genetic basis remains unresolved in many patients. Here, we identified novel compound heterozygous mutations in SCO1 (p.M294V, p.Val93*) in one such patient with fatal encephalopathy. The patient lacked the severe hepatopathy (p.P174L) or hypertrophic cardiomyopathy (p.G132S) observed in previously reported SCO1 cases, so we investigated whether allele-specific defects in SCO1 function might underlie the genotype-phenotype relationships. Fibroblasts expressing p.M294V had a relatively modest decrease in COX activity compared with those expressing p.P174L, whereas both SCO1 lines had marked copper deficiencies. Overexpression of known pathogenic variants in SCO1 fibroblasts showed that p.G132S exacerbated the COX deficiency, whereas COX activity was partially or fully restored by p.P174L and p.M294V, respectively. These data suggest that the clinical phenotypes in SCO1 patients might reflect the residual capacity of the pathogenic alleles to perform one or both functions of SCO1.


Asunto(s)
Acidosis Láctica/genética , Proteínas de la Membrana/genética , Mutación , Atrofias Olivopontocerebelosas/genética , Acidosis Láctica/metabolismo , Alelos , Secuencia de Aminoácidos , Análisis Mutacional de ADN , Resultado Fatal , Orden Génico , Humanos , Lactante , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares , Datos de Secuencia Molecular , Atrofias Olivopontocerebelosas/metabolismo , Alineación de Secuencia
15.
Hum Mol Genet ; 20(23): 4634-43, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21890497

RESUMEN

MTU1 (TRMU) is a mitochondrial enzyme responsible for the 2-thiolation of the wobble U in tRNA(Lys), tRNA(Glu) and tRNA(Gln), a post-transcriptional modification believed to be important for accurate and efficient synthesis of the 13 respiratory chain subunits encoded by mtDNA. Mutations in MTU1 are associated with acute infantile liver failure, and this has been ascribed to a transient lack of cysteine, the sulfur donor for the thiouridylation reaction, resulting in a mitochondrial translation defect during early development. A mutation in tRNA(Lys) that causes myoclonic epilepsy with ragged-red fibers (MERRF) is also reported to prevent modification of the wobble U. Here we show that mitochondrial translation is unaffected in fibroblasts from an MTU1 patient, in which MTU1 is undetectable by immunoblotting, despite the severe reduction in the 2-thiolation of mitochondrial tRNA(Lys), tRNA(Glu) and tRNA(Gln). The only respiratory chain abnormality that we could observe in these cells was an accumulation of a Complex II assembly intermediate, which, however, did not affect the level of the fully assembled enzyme. The identical phenotype was observed by siRNA-mediated knockdown of MTU1 in HEK 293 cells. Further, the mitochondrial translation deficiencies present in myoblasts from mitochondrial encephalomyopathy, lactic acidosis and stroke-like episode and MERRF patients, which are associated with defects in post-transcriptional modification of mitochondrial tRNAs, did not worsen following knockdown of MTU1 in these cells. This study demonstrates that MTU1 is not required for mitochondrial translation at normal steady-state levels of tRNAs, and that it may possess an as yet uncharacterized function in another sulfur-trafficking pathway.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Biosíntesis de Proteínas , ARNt Metiltransferasas/metabolismo , Electroforesis en Gel de Poliacrilamida , Fibroblastos/enzimología , Fibroblastos/patología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Síndrome MELAS/enzimología , Síndrome MELAS/patología , Síndrome MERRF/enzimología , Síndrome MERRF/patología , Proteínas Mitocondriales/deficiencia , Mutación/genética , Mioblastos/enzimología , Mioblastos/patología , Fosforilación Oxidativa , ARN de Transferencia/metabolismo , Tiouridina/metabolismo , ARNt Metiltransferasas/deficiencia
16.
Am J Hum Genet ; 87(1): 115-22, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20598281

RESUMEN

We investigated the genetic basis for a global and uniform decrease in mitochondrial translation in fibroblasts from patients in two unrelated pedigrees who developed Leigh syndrome, optic atrophy, and ophthalmoplegia. Analysis of the assembly of the oxidative phosphorylation complexes showed severe decreases of complexes I, IV, and V and a smaller decrease in complex III. The steady-state levels of mitochondrial mRNAs, tRNAs, and rRNAs were not reduced, nor were those of the mitochondrial translation elongation factors or the protein components of the mitochondrial ribosome. Using homozygosity mapping, we identified a 1 bp deletion in C12orf65 in one patient, and DNA sequence analysis showed a different 1 bp deletion in the second patient. Both mutations predict the same premature stop codon. C12orf65 belongs to a family of four mitochondrial class I peptide release factors, which also includes mtRF1a, mtRF1, and Ict1, all characterized by the presence of a GGQ motif at the active site. However, C12orf65 does not exhibit peptidyl-tRNA hydrolase activity in an in vitro assay with bacterial ribosomes. We suggest that it might play a role in recycling abortive peptidyl-tRNA species, released from the ribosome during the elongation phase of translation.


Asunto(s)
Enfermedad de Leigh/genética , Mitocondrias/metabolismo , Oftalmoplejía/genética , Atrofia Óptica/genética , Factores de Terminación de Péptidos/genética , Células Cultivadas , Niño , Preescolar , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Enfermedad de Leigh/metabolismo , Masculino , Mitocondrias/genética , Proteínas Mitocondriales , Mutación , Oftalmoplejía/metabolismo , Atrofia Óptica/metabolismo , Fosforilación Oxidativa , Linaje , Factores de Elongación de Péptidos/metabolismo , Factores de Terminación de Péptidos/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , ARN Ribosómico/metabolismo , ARN de Transferencia/metabolismo , Ribosomas/metabolismo , Adulto Joven
17.
Methods Mol Biol ; 2615: 153-172, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36807791

RESUMEN

Mitochondrial nucleoids are compact nucleoprotein complexes, in which mtDNA is located, replicated, and transcribed. Several proteomic approaches have been previously employed to identify nucleoid proteins; however, a consensus list of nucleoid-associated proteins has not been generated. Here we describe a proximity-biotinylation assay, BioID, which allows identification of proximity interactors of mitochondrial nucleoid proteins. It uses a promiscuous biotin ligase fused to a protein of interest which covalently attaches biotin to lysine residues of its proximal neighbors. Biotinylated proteins can be further enriched by a biotin-affinity purification and identified by mass-spectrometry. BioID can identify transient and weak interactions and can be used to identify changes in the interactions upon different cellular treatments, for different protein isoforms or for pathogenic variants.


Asunto(s)
Biotina , Proteómica , Animales , Biotina/química , Proteínas , Espectrometría de Masas , Biotinilación , Mamíferos
18.
Life Sci Alliance ; 6(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36977595

RESUMEN

Mutations in SLC25A46 underlie a wide spectrum of neurodegenerative diseases associated with alterations in mitochondrial morphology. We established an SLC25A46 knock-out cell line in human fibroblasts and studied the pathogenicity of three variants (p.T142I, p.R257Q, and p.E335D). Mitochondria were fragmented in the knock-out cell line and hyperfused in all pathogenic variants. The loss of SLC25A46 led to abnormalities in the mitochondrial cristae ultrastructure that were not rescued by the expression of the variants. SLC25A46 was present in discrete puncta at mitochondrial branch points and tips of mitochondrial tubules, co-localizing with DRP1 and OPA1. Virtually, all fission/fusion events were demarcated by a SLC25A46 focus. SLC25A46 co-immunoprecipitated with the fusion machinery, and loss of function altered the oligomerization state of OPA1 and MFN2. Proximity interaction mapping identified components of the ER membrane, lipid transfer proteins, and mitochondrial outer membrane proteins, indicating that it is present at interorganellar contact sites. SLC25A46 loss of function led to altered mitochondrial lipid composition, suggesting that it may facilitate interorganellar lipid flux or play a role in membrane remodeling associated with mitochondrial fusion and fission.


Asunto(s)
Dinámicas Mitocondriales , Proteínas Mitocondriales , Humanos , Dinámicas Mitocondriales/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Lípidos , Proteínas de Transporte de Fosfato/genética , Proteínas de Transporte de Fosfato/metabolismo
19.
Cell Rep ; 37(12): 110139, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34936866

RESUMEN

The ATPase Family AAA Domain Containing 3A (ATAD3A), is a mitochondrial inner membrane protein conserved in metazoans. ATAD3A has been associated with several mitochondrial functions, including nucleoid organization, cholesterol metabolism, and mitochondrial translation. To address its primary role, we generated a neuronal-specific conditional knockout (Atad3 nKO) mouse model, which developed a severe encephalopathy by 5 months of age. Pre-symptomatic mice showed aberrant mitochondrial cristae morphogenesis in the cortex as early as 2 months. Using a multi-omics approach in the CNS of 2-to-3-month-old mice, we found early alterations in the organelle membrane structure. We also show that human ATAD3A associates with different components of the inner membrane, including OXPHOS complex I, Letm1, and prohibitin complexes. Stochastic Optical Reconstruction Microscopy (STORM) shows that ATAD3A is regularly distributed along the inner mitochondrial membrane, suggesting a critical structural role in inner mitochondrial membrane and its organization, most likely in an ATPase-dependent manner.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Fosforilación Oxidativa , ATPasas Asociadas con Actividades Celulares Diversas/genética , Animales , Encefalopatías/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Eliminación de Secuencia , Transcriptoma
20.
Hum Mol Genet ; 17(23): 3697-707, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18753147

RESUMEN

The majority of patients with MELAS (mitochondrial encephalomyophathy, lactic acidosis, stroke-like episodes) carry a heteroplasmic A3243G mutation in the mitochondrial tRNA(Leu(UUR)). The mutation prevents modification of the wobble U base, impairing translation at UUA and UUG codons; however, whether this results in amino acid misincorporation in the mitochondrial translation products remains controversial. We tested this hypothesis in homoplasmic mutant myoblasts isolated from a MELAS patient and investigated whether overexpression of the mitochondrial translation elongation factors could suppress the translation defect. Blue-Native gel electrophoretic analysis demonstrated an almost complete lack of assembly of respiratory chain complexes I, IV and V in MELAS myoblasts. This phenotype could be partially suppressed by overexpression of EFTu or EFG2 but not EFTs or EFG1. Despite the severity of the assembly defect, overall mitochondrial protein synthesis was only moderately affected, but some anomalously migrating translation products were present. Pulse-chase labeling showed reduced stability of all mitochondrial translation products consistent with the assembly defect. Labeling patterns of the translation products were similar with [(3)H]-leucine or [(3)H]-phenylalanine, showing that loss of the wobble U modification did not permit decoding of UUY codons; however, endoproteinase fingerprint analysis showed clear evidence of amino acid misincorporation in three polypeptides: CO III, CO II and ATP6. Taken together, these data demonstrate that the A3243G mutation produces both loss- and gain-of-function phenotypes, explaining the apparent discrepancy between the severity of the translation and respiratory chain assembly defects, and suggest a function for EFG2 in quality control of translation elongation.


Asunto(s)
Aminoácidos/metabolismo , Antígenos de Neoplasias/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Expresión Génica , Síndrome MELAS/genética , Proteínas Mitocondriales/metabolismo , Mutación Missense , Factor G de Elongación Peptídica/metabolismo , Factor Tu de Elongación Peptídica/metabolismo , ARN de Transferencia de Leucina/genética , Aminoácidos/genética , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Células Cultivadas , Niño , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Femenino , Humanos , Síndrome MELAS/metabolismo , Mitocondrias/química , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Mioblastos/química , Mioblastos/metabolismo , Factor G de Elongación Peptídica/química , Factor G de Elongación Peptídica/genética , Factor Tu de Elongación Peptídica/química , Factor Tu de Elongación Peptídica/genética , Biosíntesis de Proteínas , Estabilidad Proteica , ARN de Transferencia de Leucina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA