Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 99(1): 208-16, 2016 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-27374773

RESUMEN

Mitochondrial complex I deficiency results in a plethora of often severe clinical phenotypes manifesting in early childhood. Here, we report on three complex-I-deficient adult subjects with relatively mild clinical symptoms, including isolated, progressive exercise-induced myalgia and exercise intolerance but with normal later development. Exome sequencing and targeted exome sequencing revealed compound-heterozygous mutations in TMEM126B, encoding a complex I assembly factor. Further biochemical analysis of subject fibroblasts revealed a severe complex I deficiency caused by defective assembly. Lentiviral complementation with the wild-type cDNA restored the complex I deficiency, demonstrating the pathogenic nature of these mutations. Further complexome analysis of one subject indicated that the complex I assembly defect occurred during assembly of its membrane module. Our results show that TMEM126B defects can lead to complex I deficiencies and, interestingly, that symptoms can occur only after exercise.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Proteínas de la Membrana/genética , Enfermedades Mitocondriales/genética , Debilidad Muscular/genética , Mutación , Adolescente , Adulto , Niño , Complejo I de Transporte de Electrón/genética , Ejercicio Físico , Exoma/genética , Prueba de Complementación Genética , Heterocigoto , Humanos , Lactante , Masculino , Adulto Joven
2.
J Pediatr ; 196: 309-313.e3, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29395179

RESUMEN

We demonstrate that a heterozygous nuclear variant in the gene encoding mitochondrial complex I subunit NDUFV1 aggravates the cellular phenotype in the presence of a mitochondrial DNA variant in complex I subunit ND1. Our findings suggest that heterozygous variants could be more significant in inherited mitochondrial diseases than hitherto assumed.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Enfermedades Mitocondriales/genética , NADH Deshidrogenasa/genética , Niño , ADN Mitocondrial/genética , Complejo I de Transporte de Electrón/genética , Femenino , Pruebas Genéticas/métodos , Heterocigoto , Humanos , Recién Nacido , Masculino , Enfermedades Mitocondriales/diagnóstico , Mutación , Fenotipo
3.
Hum Mol Genet ; 23(5): 1311-9, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24158852

RESUMEN

Oxidative phosphorylation and fatty acid oxidation are two major metabolic pathways in mitochondria. Acyl-CoA dehydrogenase 9 (ACAD9), an enzyme assumed to play a role in fatty acid oxidation, was recently identified as a factor involved in complex I biogenesis. Here we further investigated the role of ACAD9's enzymatic activity in fatty acid oxidation and complex I biogenesis. We provide evidence indicating that ACAD9 displays enzyme activity in vivo. Knockdown experiments in very-long-chain acyl-CoA dehydrogenase (VLCAD)-deficient fibroblasts revealed that ACAD9 is responsible for the production of C14:1-carnitine from oleate and C12-carnitine from palmitate. These results explain the origin of these obscure acylcarnitines that are used to diagnose VLCAD deficiency in humans. Knockdown of ACAD9 in control fibroblasts did not reveal changes in the acylcarnitine profiles upon fatty acid loading. Next, we investigated whether catalytic activity of ACAD9 was necessary for complex I biogenesis. Catalytically inactive ACAD9 gave partial-to-complete rescue of complex I biogenesis in ACAD9-deficient cells and was incorporated in high-molecular-weight assembly intermediates. Our results underscore the importance of the ACAD9 protein in complex I assembly and suggest that the enzymatic activity is a rudiment of the duplication event.


Asunto(s)
Acil-CoA Deshidrogenasas/metabolismo , Ácidos Grasos/metabolismo , Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Acil-CoA Deshidrogenasa de Cadena Larga/metabolismo , Acil-CoA Deshidrogenasas/química , Acil-CoA Deshidrogenasas/deficiencia , Acil-CoA Deshidrogenasas/genética , Carnitina/biosíntesis , Catálisis , Línea Celular , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Complejo I de Transporte de Electrón/deficiencia , Activación Enzimática , Humanos , Errores Innatos del Metabolismo Lipídico/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Modelos Moleculares , Peso Molecular , Enfermedades Musculares/metabolismo , Mutación , Oxidación-Reducción , Fosforilación Oxidativa , Conformación Proteica
4.
Hum Mol Genet ; 23(23): 6356-65, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25008109

RESUMEN

Complex III (cytochrome bc1) is a protein complex of the mitochondrial inner membrane that transfers electrons from ubiquinol to cytochrome c. Its assembly requires the coordinated expression of mitochondrial-encoded cytochrome b and nuclear-encoded subunits and assembly factors. Complex III deficiency is a severe multisystem disorder caused by mutations in subunit genes or assembly factors. Sequence-profile-based orthology predicts C11orf83, hereafter named UQCC3, to be the ortholog of the fungal complex III assembly factor CBP4. We describe a homozygous c.59T>A missense mutation in UQCC3 from a consanguineous patient diagnosed with isolated complex III deficiency, displaying lactic acidosis, hypoglycemia, hypotonia and delayed development without dysmorphic features. Patient fibroblasts have reduced complex III activity and lower levels of the holocomplex and its subunits than controls. They have no detectable UQCC3 protein and have lower levels of cytochrome b protein. Furthermore, in patient cells, cytochrome b is absent from a high-molecular-weight complex III. UQCC3 is reduced in cells depleted for the complex III assembly factors UQCC1 and UQCC2. Conversely, absence of UQCC3 in patient cells does not affect UQCC1 and UQCC2. This suggests that UQCC3 functions in the complex III assembly pathway downstream of UQCC1 and UQCC2 and is consistent with what is known about the function of Cbp4 and of the fungal orthologs of UQCC1 and UQCC2, Cbp3 and Cbp6. We conclude that UQCC3 functions in complex III assembly and that the c.59T>A mutation has a causal role in complex III deficiency.


Asunto(s)
Proteínas Portadoras/genética , Citocromos b/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Proteínas de Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Consanguinidad , Complejo III de Transporte de Electrones/deficiencia , Complejo III de Transporte de Electrones/genética , Estabilidad de Enzimas , Femenino , Fibroblastos/metabolismo , Humanos , Recién Nacido , Proteínas de la Membrana/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Mutación Missense
5.
J Inherit Metab Dis ; 39(1): 59-65, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26310962

RESUMEN

We reported before that the minimal alveolar concentration (MAC) of isoflurane is decreased in complex I-deficient mice lacking the NDUFS4 subunit of the respiratory chain (RC) (1.55 and 0.81% at postnatal (PN) 22-25 days and 1.68 and 0.65% at PN 31-34 days for wildtype (WT) and CI-deficient KO, respectively). A more severe respiratory depression was caused by 1.0 MAC isoflurane in KO mice (respiratory rate values of 86 and 45 at PN 22-25 days and 69 and 29 at PN 31-34 days for anesthetized WT and KO, respectively). Here, we address the idea that isoflurane anesthesia causes a much larger decrease in brain mitochondrial ATP production in KO mice thus explaining their increased sensitivity to this anesthetic. Brains from WT and KO mice of the above study were removed immediately after MAC determination at PN 31-34 days and a mitochondria-enriched fraction was prepared. Aliquots were used for measurement of maximal ATP production in the presence of pyruvate, malate, ADP and creatine and, after freeze-thawing, the maximal activity of the individual RC complexes in the presence of complex-specific substrates. CI activity was dramatically decreased in KO, whereas ATP production was decreased by only 26% (p < 0.05). The activities of CII, CIII, and CIV were the same for WT and KO. Isoflurane anesthesia decreased the activity of CI by 30% (p < 0.001) in WT. In sharp contrast, it increased the activity of CII by 37% (p < 0.001) and 50% (p < 0.001) and that of CIII by 37% (p < 0.001) and 40% (p < 0.001) in WT and KO, respectively, whereas it tended to increase that of CIV in both WT and KO. Isoflurane anesthesia increased ATP production by 52 and 69% in WT (p < 0.05) and KO (p < 0.01), respectively. Together these findings indicate that isoflurane anesthesia interferes positively rather than negatively with the ability of CI-deficient mice brain mitochondria to convert their main substrate pyruvate into ATP.


Asunto(s)
Adenosina Trifosfato/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/metabolismo , Isoflurano/administración & dosificación , Mitocondrias/efectos de los fármacos , Anestesia/métodos , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Ácido Pirúvico/metabolismo
6.
PLoS Genet ; 9(12): e1004034, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24385928

RESUMEN

Mitochondrial oxidative phosphorylation (OXPHOS) is responsible for generating the majority of cellular ATP. Complex III (ubiquinol-cytochrome c oxidoreductase) is the third of five OXPHOS complexes. Complex III assembly relies on the coordinated expression of the mitochondrial and nuclear genomes, with 10 subunits encoded by nuclear DNA and one by mitochondrial DNA (mtDNA). Complex III deficiency is a debilitating and often fatal disorder that can arise from mutations in complex III subunit genes or one of three known complex III assembly factors. The molecular cause for complex III deficiency in about half of cases, however, is unknown and there are likely many complex III assembly factors yet to be identified. Here, we used Massively Parallel Sequencing to identify a homozygous splicing mutation in the gene encoding Ubiquinol-Cytochrome c Reductase Complex Assembly Factor 2 (UQCC2) in a consanguineous Lebanese patient displaying complex III deficiency, severe intrauterine growth retardation, neonatal lactic acidosis and renal tubular dysfunction. We prove causality of the mutation via lentiviral correction studies in patient fibroblasts. Sequence-profile based orthology prediction shows UQCC2 is an ortholog of the Saccharomyces cerevisiae complex III assembly factor, Cbp6p, although its sequence has diverged substantially. Co-purification studies show that UQCC2 interacts with UQCC1, the predicted ortholog of the Cbp6p binding partner, Cbp3p. Fibroblasts from the patient with UQCC2 mutations have deficiency of UQCC1, while UQCC1-depleted cells have reduced levels of UQCC2 and complex III. We show that UQCC1 binds the newly synthesized mtDNA-encoded cytochrome b subunit of complex III and that UQCC2 patient fibroblasts have specific defects in the synthesis or stability of cytochrome b. This work reveals a new cause for complex III deficiency that can assist future patient diagnosis, and provides insight into human complex III assembly by establishing that UQCC1 and UQCC2 are complex III assembly factors participating in cytochrome b biogenesis.


Asunto(s)
Citocromos b/biosíntesis , Complejo III de Transporte de Electrones/genética , Proteínas de la Membrana/genética , Enfermedades Mitocondriales/genética , Consanguinidad , Citocromos b/genética , Complejo III de Transporte de Electrones/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Homocigoto , Humanos , Proteínas de la Membrana/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales/patología , Enfermedades Mitocondriales/terapia , Proteínas Mitocondriales/genética , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutación , Fosforilación Oxidativa , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
7.
Hum Mutat ; 36(1): 34-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25339201

RESUMEN

COA6/C1ORF31 is involved in cytochrome c oxidase (complex IV) biogenesis. We present a new pathogenic COA6 variant detected in a patient with neonatal hypertrophic cardiomyopathy and isolated complex IV deficiency. For the first time, clinical details about a COA6-deficient patient are given and patient fibroblasts are functionally characterized: COA6 protein is undetectable and steady-state levels of complex IV and several of its subunits are reduced. The monomeric COX1 assembly intermediate accumulates. Using pulse-chase experiments, we demonstrate an increased turnover of mitochondrial encoded complex IV subunits. Although monomeric complex IV is decreased in patient fibroblasts, the CI/CIII2 /CIVn -supercomplexes remain unaffected. Copper supplementation shows a partial rescue of complex IV deficiency in patient fibroblasts. We conclude that COA6 is required for complex IV subunit stability. Furthermore, the proposed role in the copper delivery pathway to complex IV subunits is substantiated and a therapeutic lead for COA6-deficient patients is provided.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Deficiencia de Citocromo-c Oxidasa/genética , Complejo IV de Transporte de Electrones/genética , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/patología , Cobre/administración & dosificación , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Células HEK293 , Humanos , Recién Nacido , Mitocondrias/metabolismo
8.
Hum Mol Genet ; 22(4): 656-67, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23125284

RESUMEN

The mitochondrial respiratory chain complex IV (cytochrome c oxidase) is a multi-subunit enzyme that transfers electrons from cytochrome c to molecular oxygen, yielding water. Its biogenesis requires concerted expression of mitochondria- and nuclear-encoded subunits and assembly factors. In this report, we describe a homozygous missense mutation in FAM36A from a patient who displays ataxia and muscle hypotonia. The FAM36A gene is a remote, putative ortholog of the fungal complex IV assembly factor COX20. Messenger RNA (mRNA) and protein co-expression analyses support the involvement of FAM36A in complex IV function in mammals. The c.154A>C mutation in the FAM36A gene, a mutation that is absent in sequenced exomes, leads to a reduced activity and lower levels of complex IV and its protein subunits. The FAM36A protein is nearly absent in patient's fibroblasts. Cells affected by the mutation accumulate subassemblies of complex IV that contain COX1 but are almost devoid of COX2 protein. We observe co-purification of FAM36A and COX2 proteins, supporting that the FAM36A defect hampers the early step of complex IV assembly at the incorporation of the COX2 subunit. Lentiviral complementation of patient's fibroblasts with wild-type FAM36A increases the complex IV activity as well as the amount of holocomplex IV and of individual subunits. These results establish the function of the human gene FAM36A/COX20 in complex IV assembly and support a causal role of the gene in complex IV deficiency.


Asunto(s)
Anomalías Múltiples/genética , Ataxia/genética , Deficiencia de Citocromo-c Oxidasa/genética , Canales Iónicos/genética , Hipotonía Muscular/genética , Multimerización de Proteína , Anomalías Múltiples/metabolismo , Secuencia de Aminoácidos , Animales , Ataxia/metabolismo , Secuencia de Bases , Células Cultivadas , Niño , Consanguinidad , Deficiencia de Citocromo-c Oxidasa/metabolismo , Análisis Mutacional de ADN , Complejo IV de Transporte de Electrones/metabolismo , Expresión Génica , Humanos , Canales Iónicos/metabolismo , Ácido Láctico/sangre , Ácido Láctico/líquido cefalorraquídeo , Masculino , Proteínas de la Membrana/genética , Ratones , Mitocondrias/enzimología , Proteínas Mitocondriales/genética , Datos de Secuencia Molecular , Hipotonía Muscular/metabolismo , Mutación Missense , Proteínas de Saccharomyces cerevisiae/genética
9.
Hum Mol Genet ; 22(15): 3138-51, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23575228

RESUMEN

It is estimated that the human mitochondrial proteome consists of 1000-1500 distinct proteins. The majority of these support the various biochemical pathways that are active in these organelles. Individuals with an oxidative phosphorylation disorder of unknown cause provide a unique opportunity to identify novel genes implicated in mitochondrial biology. We identified a homozygous deletion of CEP89 in a patient with isolated complex IV deficiency, intellectual disability and multisystemic problems. CEP89 is a ubiquitously expressed and highly conserved gene of unknown function. Immunocytochemistry and cellular fractionation experiments showed that CEP89 is present both in the cytosol and in the mitochondrial intermembrane space. Furthermore, we ascertained in vitro that downregulation of CEP89 resulted in a severe decrease in complex IV in-gel activity and altered mobility, suggesting that the complex is aberrantly formed. Two-dimensional BN-SDS gel analysis revealed that CEP89 associates with a high-molecular weight complex. Together, these data confirm a role for CEP89 in mitochondrial metabolism. In addition, we modeled CEP89 loss of function in Drosophila. Ubiquitous knockdown of fly Cep89 decreased complex IV activity and resulted in complete lethality. Furthermore, Cep89 is required for mitochondrial integrity, membrane depolarization and synaptic transmission of photoreceptor neurons, and for (sub)synaptic organization of the larval neuromuscular junction. Finally, we tested neuronal Cep89 knockdown flies in the light-off jump reflex habituation assay, which revealed its role in learning. We conclude that CEP89 proteins play an important role in mitochondrial metabolism, especially complex IV activity, and are required for neuronal and cognitive function across evolution.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Niño , Cromosomas Humanos Par 19 , Deficiencia de Citocromo-c Oxidasa/genética , Deficiencia de Citocromo-c Oxidasa/metabolismo , Citosol , Modelos Animales de Enfermedad , Drosophila/genética , Proteínas de Drosophila/genética , Femenino , Eliminación de Gen , Expresión Génica , Técnicas de Silenciamiento del Gen , Homocigoto , Humanos , Aprendizaje , Proteínas Asociadas a Microtúbulos , Mitocondrias/genética , Mutación , Especificidad de Órganos/genética , Polimorfismo de Nucleótido Simple , Transporte de Proteínas , Sinapsis/genética , Sinapsis/metabolismo
10.
Am J Hum Genet ; 88(4): 488-93, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21457908

RESUMEN

The assembly of mitochondrial respiratory chain complex IV (cytochrome c oxidase) involves the coordinated action of several assembly chaperones. In Saccharomyces cerevisiae, at least 30 different assembly chaperones have been identified. To date, pathogenic mutations leading to a mitochondrial disorder have been identified in only seven of the corresponding human genes. One of the genes for which the relevance to human pathology is unknown is C2orf64, an ortholog of the S. cerevisiae gene PET191. This gene has previously been shown to be a complex IV assembly factor in yeast, although its exact role is still unknown. Previous research in a large cohort of complex IV deficient patients did not support an etiological role of C2orf64 in complex IV deficiency. In this report, a homozygous mutation in C2orf64 is described in two siblings affected by fatal neonatal cardiomyopathy. Pathogenicity of the mutation is supported by the results of a complementation experiment, showing that complex IV activity can be fully restored by retroviral transduction of wild-type C2orf64 in patient-derived fibroblasts. Detailed analysis of complex IV assembly intermediates in patient fibroblasts by 2D-BN PAGE revealed the accumulation of a small assembly intermediate containing subunit COX1 but not the COX2, COX4, or COX5b subunits, indicating that C2orf64 is involved in an early step of the complex IV assembly process. The results of this study demonstrate that C2orf64 is essential for human complex IV assembly and that C2orf64 mutational analysis should be considered for complex IV deficient patients, in particular those with hypertrophic cardiomyopathy.


Asunto(s)
Cardiomiopatías/enzimología , Cardiomiopatías/genética , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Enfermedades Mitocondriales/enzimología , Enfermedades Mitocondriales/genética , Mutación Missense , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , Consanguinidad , Análisis Mutacional de ADN , Electroforesis en Gel Bidimensional , Resultado Fatal , Femenino , Fibroblastos/enzimología , Prueba de Complementación Genética , Homocigoto , Humanos , Recién Nacido , Masculino , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Linaje , Multimerización de Proteína , Homología de Secuencia de Aminoácido
11.
Nucleic Acids Res ; 40(9): 4040-51, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22238375

RESUMEN

In a comparative genomics study for mitochondrial ribosome-associated proteins, we identified C7orf30, the human homolog of the plant protein iojap. Gene order conservation among bacteria and the observation that iojap orthologs cannot be transferred between bacterial species predict this protein to be associated with the mitochondrial ribosome. Here, we show colocalization of C7orf30 with the large subunit of the mitochondrial ribosome using isokinetic sucrose gradient and 2D Blue Native polyacrylamide gel electrophoresis (BN-PAGE) analysis. We co-purified C7orf30 with proteins of the large subunit, and not with proteins of the small subunit, supporting interaction that is specific to the large mitoribosomal complex. Consistent with this physical association, a mitochondrial translation assay reveals negative effects of C7orf30 siRNA knock-down on mitochondrial gene expression. Based on our data we propose that C7orf30 is involved in ribosomal large subunit function. Sequencing the gene in 35 patients with impaired mitochondrial translation did not reveal disease-causing mutations in C7orf30.


Asunto(s)
Proteínas Mitocondriales/fisiología , Biosíntesis de Proteínas , Proteínas Ribosómicas/fisiología , Subunidades Ribosómicas Grandes de Eucariotas/química , Secuencia de Aminoácidos , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Genes Bacterianos , Células HEK293 , Humanos , Mitocondrias/genética , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Datos de Secuencia Molecular , Nucleótidos/metabolismo , Operón , Filogenia , Estructura Terciaria de Proteína , Proteínas Ribosómicas/química , Proteínas Ribosómicas/genética , Análisis de Secuencia de ADN
12.
Proc Natl Acad Sci U S A ; 108(21): 8657-62, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21555543

RESUMEN

Intracellular chemical reactions generally constitute reaction-diffusion systems located inside nanostructured compartments like the cytosol, nucleus, endoplasmic reticulum, Golgi, and mitochondrion. Understanding the properties of such systems requires quantitative information about solute diffusion. Here we present a novel approach that allows determination of the solvent-dependent solute diffusion constant (D(solvent)) inside cell compartments with an experimentally quantifiable nanostructure. In essence, our method consists of the matching of synthetic fluorescence recovery after photobleaching (FRAP) curves, generated by a mathematical model with a realistic nanostructure, and experimental FRAP data. As a proof of principle, we assessed D(solvent) of a monomeric fluorescent protein (AcGFP1) and its tandem fusion (AcGFP1(2)) in the mitochondrial matrix of HEK293 cells. Our results demonstrate that diffusion of both proteins is substantially slowed by barriers in the mitochondrial matrix (cristae), suggesting that cells can control the dynamics of biochemical reactions in this compartment by modifying its nanostructure.


Asunto(s)
Mitocondrias/ultraestructura , Proteínas/metabolismo , Compartimento Celular , Difusión , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Cinética , Mitocondrias/metabolismo , Nanoestructuras/ultraestructura , Soluciones
13.
J Biol Chem ; 287(50): 41851-60, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23038253

RESUMEN

Studies employing native PAGE suggest that most nDNA-encoded CI subunits form subassemblies before assembling into holo-CI. In addition, in vitro evidence suggests that some subunits can directly exchange in holo-CI. Presently, data on the kinetics of these two incorporation modes for individual CI subunits during CI maintenance are sparse. Here, we used inducible HEK293 cell lines stably expressing AcGFP1-tagged CI subunits and quantified the amount of tagged subunit in mitoplasts and holo-CI by non-native and native PAGE, respectively, to determine their CI incorporation efficiency. Analysis of time courses of induction revealed three subunit-specific patterns. A first pattern, represented by NDUFS1, showed overlapping time courses, indicating that imported subunits predominantly incorporate into holo-CI. A second pattern, represented by NDUFV1, consisted of parallel time courses, which were, however, not quantitatively overlapping, suggesting that imported subunits incorporate at similar rates into holo-CI and CI assembly intermediates. The third pattern, represented by NDUFS3 and NDUFA2, revealed a delayed incorporation into holo-CI, suggesting their prior appearance in CI assembly intermediates and/or as free monomers. Our analysis showed the same maximum incorporation into holo-CI for NDUFV1, NDUFV2, NDUFS1, NDUFS3, NDUFS4, NDUFA2, and NDUFA12 with nearly complete loss of endogenous subunit at 24 h of induction, indicative of an equimolar stoichiometry and unexpectedly rapid turnover. In conclusion, the results presented demonstrate that newly formed nDNA-encoded CI subunits rapidly incorporate into holo-CI in a subunit-specific manner.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Homeostasis/fisiología , Proteínas Mitocondriales/metabolismo , Subunidades de Proteína/metabolismo , Animales , Cricetinae , Cricetulus , Complejo I de Transporte de Electrón/genética , Células HEK293 , Humanos , Cinética , Proteínas Mitocondriales/genética , Subunidades de Proteína/genética
14.
Biochim Biophys Acta ; 1822(2): 168-75, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22036843

RESUMEN

In this study, we investigated the pathogenicity of a homozygous Asp446Asn mutation in the NDUFS2 gene of a patient with a mitochondrial respiratory chain complex I deficiency. The clinical, biochemical, and genetic features of the NDUFS2 patient were compared with those of 4 patients with previously identified NDUFS2 mutations. All 5 patients presented with Leigh syndrome. In addition, 3 out of 5 showed hypertrophic cardiomyopathy. Complex I amounts in the patient carrying the Asp446Asn mutation were normal, while the complex I activity was strongly reduced, showing that the NDUFS2 mutation affects complex I enzymatic function. By contrast, the 4 other NDUFS2 patients showed both a reduced amount and activity of complex I. The enzymatic defect in fibroblasts of the patient carrying the Asp446Asn mutation was rescued by transduction of wild type NDUFS2. A 3-D model of the catalytic core of complex I showed that the mutated amino acid residue resides near the coenzyme Q binding pocket. However, the K(M) of complex I for coenzyme Q analogs of the Asp446Asn mutated complex I was similar to the K(M) observed in other complex I defects and in controls. We propose that the mutation interferes with the reduction of coenzyme Q or with the coupling of coenzyme Q reduction with the conformational changes involved in proton pumping of complex I.


Asunto(s)
Complejo I de Transporte de Electrón/genética , Enfermedad de Leigh/genética , Mitocondrias/enzimología , Mutación , NADH Deshidrogenasa/genética , Secuencia de Aminoácidos , Animales , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Catálisis , Complejo I de Transporte de Electrón/metabolismo , Femenino , Fibroblastos/metabolismo , Homocigoto , Humanos , Lactante , Recién Nacido , Enfermedad de Leigh/enzimología , Enfermedad de Leigh/metabolismo , Potencial de la Membrana Mitocondrial/genética , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/metabolismo , Membranas Mitocondriales/enzimología , Membranas Mitocondriales/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , NADH Deshidrogenasa/metabolismo , Conformación Proteica , Transducción Genética/métodos , Ubiquinona/metabolismo
15.
Biochim Biophys Acta ; 1817(10): 1925-36, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22430089

RESUMEN

Human mitochondrial complex I (CI) deficiency is associated with progressive neurological disorders. To better understand the CI pathomechanism, we here studied how deletion of the CI gene NDUFS4 affects cell metabolism. To this end we compared immortalized mouse embryonic fibroblasts (MEFs) derived from wildtype (wt) and whole-body NDUFS4 knockout (KO) mice. Mitochondria from KO cells lacked the NDUFS4 protein and mitoplasts displayed virtually no CI activity, moderately reduced CII, CIII and CIV activities and normal citrate synthase and CV (F(o)F(1)-ATPase) activity. Native electrophoresis of KO cell mitochondrial fractions revealed two distinct CI subcomplexes of ~830kDa (enzymatically inactive) and ~200kDa (active). The level of fully-assembled CII-CV was not affected by NDUFS4 gene deletion. KO cells exhibited a moderately reduced maximal and routine O(2) consumption, which was fully inhibited by acute application of the CI inhibitor rotenone. The aberrant CI assembly and reduced O(2) consumption in KO cells were fully normalized by NDUFS4 gene complementation. Cellular [NAD(+)]/[NADH] ratio, lactate production and mitochondrial tetramethyl rhodamine methyl ester (TMRM) accumulation were slightly increased in KO cells. In contrast, NDUFS4 gene deletion did not detectably alter [NADP(+)]/[NADPH] ratio, cellular glucose consumption, the protein levels of hexokinases (I and II) and phosphorylated pyruvate dehydrogenase (P-PDH), total cellular adenosine triphosphate (ATP) level, free cytosolic [ATP], cell growth rate, and reactive oxygen species (ROS) levels. We conclude that the NDUFS4 subunit is of key importance in CI stabilization and that, due to the metabolic properties of the immortalized MEFs, NDUFS4 gene deletion has only modest effects at the live cell level. This article is part of a special issue entitled: 17th European Bioenergetics Conference (EBEC 2012).


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Embrión de Mamíferos/enzimología , Fibroblastos/enzimología , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Animales , Línea Celular Transformada , Complejo I de Transporte de Electrón/genética , Embrión de Mamíferos/citología , Estabilidad de Enzimas/fisiología , Fibroblastos/citología , Eliminación de Gen , Humanos , Ácido Láctico/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , NAD/genética , NAD/metabolismo , NADP/genética , NADP/metabolismo , Fosforilación/fisiología , ATPasas de Translocación de Protón/genética , ATPasas de Translocación de Protón/metabolismo , Complejo Piruvato Deshidrogenasa/genética , Complejo Piruvato Deshidrogenasa/metabolismo
16.
Brain ; 135(Pt 1): 12-22, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22036961

RESUMEN

Complex I deficiency is the most frequent cause of oxidative phosphorylation disorders. The disease features a large diversity of clinical symptoms often leading to progressive encephalomyopathies with a fatal outcome. There is currently no cure, and although disease-causing mutations have been found in the genes encoding complex I subunits, half of the cases remain unexplained. However, in the past 5 years a new class of complex I disease genes has emerged with the finding of specific assembly factors. So far nine such genes have been described and it is believed that in the near future more will be found. In this review, we will address whether the functions of these chaperones point towards a general molecular mechanism of disease and whether this enables us to design a treatment for complex I deficiency.


Asunto(s)
ADN Mitocondrial/genética , Enfermedades Mitocondriales/genética , Chaperonas Moleculares/genética , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/genética , Humanos , Enfermedades Mitocondriales/terapia , Fosforilación Oxidativa
17.
Nucleic Acids Res ; 39(10): 4284-99, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21278163

RESUMEN

Here we show that c17orf42, hereafter TEFM (transcription elongation factor of mitochondria), makes a critical contribution to mitochondrial transcription. Inactivation of TEFM in cells by RNA interference results in respiratory incompetence owing to decreased levels of H- and L-strand promoter-distal mitochondrial transcripts. Affinity purification of TEFM from human mitochondria yielded a complex comprising mitochondrial transcripts, mitochondrial RNA polymerase (POLRMT), pentatricopeptide repeat domain 3 protein (PTCD3), and a putative DEAD-box RNA helicase, DHX30. After RNase treatment only POLRMT remained associated with TEFM, and in human cultured cells TEFM formed foci coincident with newly synthesized mitochondrial RNA. Based on deletion mutants, TEFM interacts with the catalytic region of POLRMT, and in vitro TEFM enhanced POLRMT processivity on ss- and dsDNA templates. TEFM contains two HhH motifs and a Ribonuclease H fold, similar to the nuclear transcription elongation regulator Spt6. These findings lead us to propose that TEFM is a mitochondrial transcription elongation factor.


Asunto(s)
Mitocondrias/genética , Proteínas Mitocondriales/fisiología , ARN/biosíntesis , Factores de Transcripción/fisiología , Factores de Elongación Transcripcional/fisiología , Dominio Catalítico , Línea Celular , ADN Mitocondrial/análisis , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/metabolismo , Silenciador del Gen , Humanos , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/química , Fosforilación Oxidativa , Estructura Terciaria de Proteína , ARN/metabolismo , ARN Mitocondrial , Factores de Elongación Transcripcional/antagonistas & inhibidores , Factores de Elongación Transcripcional/química
18.
Proteomics ; 12(9): 1349-62, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22589185

RESUMEN

Most eukaryotic cells depend on mitochondrial OXidative PHOSphorylation (OXPHOS) in their ATP supply. The cellular consequences of OXPHOS defects and the pathophysiological mechanisms in related disorders are incompletely understood. Using a quantitative proteomics approach we provide evidence that a genetic defect of complex-I of the OXPHOS system may associate with transcriptional derangements of mitochondrial biogenesis through stabilization of the master transcriptional regulator PPARγ co-activator 1α (PGC-1α) protein. Chronic oxidative stress suppresses the gene expression of PGC-1α but concomitant inhibition of the ubiquitin-proteasome system (UPS) can stabilize this co-activator protein, thereby inducing its downstream metabolic gene expression programs. Thus, mitochondrial biogenesis, which lays at the heart of the homeostatic control of energy metabolism, can be deregulated by secondary impairments of the protein turnover machinery.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Células Cultivadas , Complejo I de Transporte de Electrón , Fibroblastos , Expresión Génica , Proteínas de Choque Térmico/genética , Humanos , Peróxido de Hidrógeno/metabolismo , Proteínas Mitocondriales/análisis , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Fosforilación Oxidativa , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Proteoma , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/genética
19.
Biochim Biophys Acta ; 1807(12): 1624-33, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21978538

RESUMEN

Complex I (CI) of the oxidative phosphorylation system is assembled from 45 subunits encoded by both the mitochondrial and nuclear DNA. Defective mitochondrial translation is a major cause of mitochondrial disorders and proper understanding of its mechanisms and consequences is fundamental to rational treatment design. Here, we used a live cell approach to assess its consequences on CI assembly. The approach consisted of fluorescence recovery after photobleaching (FRAP) imaging of the effect of mitochondrial translation inhibition by chloramphenicol (CAP) on the dynamics of AcGFP1-tagged CI subunits NDUFV1, NDUFS3, NDUFA2 and NDUFB6 and assembly factor NDUFAF4. CAP increased the mobile fraction of the subunits, but not NDUFAF4, and decreased the amount of CI, demonstrating that CI is relatively immobile and does not associate with NDUFAF4. CAP increased the recovery kinetics of NDUFV1-AcGFP1 to the same value as obtained with AcGFP1 alone, indicative of the removal of unbound NDUFV1 from the mitochondrial matrix. Conversely, CAP decreased the mobility of NDUFS3-AcGFP1 and, to a lesser extent, NDUFB6-AcGFP1, suggestive of their enrichment in less mobile subassemblies. Little, if any, change in mobility of NDUFA2-AcGFP1 could be detected, suggesting that the dynamics of this accessory subunit of the matrix arm remains unaltered. Finally, CAP increased the mobility of NDUFAF4-AcGFP1, indicative of interaction with a more mobile membrane-bound subassembly. Our results show that the protein interactions of CI subunits and assembly factors are differently altered when mitochondrial translation is defective.


Asunto(s)
Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/fisiología , Biosíntesis de Proteínas , Subunidades de Proteína/metabolismo , Línea Celular , Complejo I de Transporte de Electrón/genética , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Mitocondrias/genética , Subunidades de Proteína/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
20.
Am J Hum Genet ; 84(6): 718-27, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19463981

RESUMEN

Mitochondrial complex I deficiency is the most prevalent and least understood disorder of the oxidative phosphorylation system. The genetic cause of many cases of isolated complex I deficiency is unknown because of insufficient understanding of the complex I assembly process and the factors involved. We performed homozygosity mapping and gene sequencing to identify the genetic defect in five complex I-deficient patients from three different families. All patients harbored mutations in the NDUFAF3 (C3ORF60) gene, of which the pathogenic nature was assessed by NDUFAF3-GFP baculovirus complementation in fibroblasts. We found that NDUFAF3 is a genuine mitochondrial complex I assembly protein that interacts with complex I subunits. Furthermore, we show that NDUFAF3 tightly interacts with NDUFAF4 (C6ORF66), a protein previously implicated in complex I deficiency. Additional gene conservation analysis links NDUFAF3 to bacterial-membrane-insertion gene cluster SecF/SecD/YajC and to C8ORF38, also implicated in complex I deficiency. These data not only show that NDUFAF3 mutations cause complex I deficiency but also relate different complex I disease genes by the close cooperation of their encoded proteins during the assembly process.


Asunto(s)
Proteínas de Unión a Calmodulina/genética , Complejo I de Transporte de Electrón/genética , Proteínas de la Membrana/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Consanguinidad , Análisis Mutacional de ADN , Electroforesis en Gel de Poliacrilamida , Resultado Fatal , Femenino , Prueba de Complementación Genética , Humanos , Lactante , Recién Nacido , Masculino , Modelos Genéticos , Datos de Secuencia Molecular , Linaje , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA