Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Trends Biochem Sci ; 47(12): 999-1008, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35961810

RESUMEN

Deep understanding of the pathophysiological role of the mitochondrial respiratory chain (MRC) relies on a well-grounded model explaining how its biogenesis is regulated. The lack of a consistent framework to clarify the modes and mechanisms governing the assembly of the MRC complexes and supercomplexes (SCs) works against progress in the field. The plasticity model was postulated as an attempt to explain the coexistence of mammalian MRC complexes as individual entities and associated in SC species. However, mounting data accumulated throughout the years question the universal validity of the plasticity model as originally proposed. Instead, as we argue here, a cooperative assembly model provides a much better explanation to the phenomena observed when studying MRC biogenesis in physiological and pathological settings.


Asunto(s)
Mitocondrias , Membranas Mitocondriales , Animales , Transporte de Electrón/fisiología , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Mamíferos
2.
Mol Cell ; 67(1): 96-105.e4, 2017 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-28673544

RESUMEN

Loss-of-function mutations in TTC19 (tetra-tricopeptide repeat domain 19) have been associated with severe neurological phenotypes and mitochondrial respiratory chain complex III deficiency. We previously demonstrated the mitochondrial localization of TTC19 and its link with complex III biogenesis. Here we provide detailed insight into the mechanistic role of TTC19, by investigating a Ttc19?/? mouse model that shows progressive neurological and metabolic decline, decreased complex III activity, and increased production of reactive oxygen species. By using both the Ttc19?/? mouse model and a range of human cell lines, we demonstrate that TTC19 binds to the fully assembled complex III dimer, i.e., after the incorporation of the iron-sulfur Rieske protein (UQCRFS1). The in situ maturation of UQCRFS1 produces N-terminal polypeptides, which remain bound to holocomplex III. We show that, in normal conditions, these UQCRFS1 fragments are rapidly removed, but when TTC19 is absent they accumulate within complex III, causing its structural and functional impairment.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Proteínas Hierro-Azufre/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Animales , Conducta Animal , Modelos Animales de Enfermedad , Complejo III de Transporte de Electrones/deficiencia , Complejo III de Transporte de Electrones/genética , Femenino , Genotipo , Células HeLa , Humanos , Proteínas Hierro-Azufre/genética , Cinética , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Mitocondriales , Proteínas Mitocondriales/genética , Actividad Motora , Degeneración Nerviosa , Sistema Nervioso/metabolismo , Sistema Nervioso/patología , Sistema Nervioso/fisiopatología , Fenotipo , Unión Proteica , Estabilidad Proteica , Proteolisis , Especies Reactivas de Oxígeno/metabolismo
3.
EMBO J ; 39(3): e102817, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31912925

RESUMEN

Mitochondrial respiratory chain (MRC) enzymes associate in supercomplexes (SCs) that are structurally interdependent. This may explain why defects in a single component often produce combined enzyme deficiencies in patients. A case in point is the alleged destabilization of complex I in the absence of complex III. To clarify the structural and functional relationships between complexes, we have used comprehensive proteomic, functional, and biogenetical approaches to analyze a MT-CYB-deficient human cell line. We show that the absence of complex III blocks complex I biogenesis by preventing the incorporation of the NADH module rather than decreasing its stability. In addition, complex IV subunits appeared sequestered within complex III subassemblies, leading to defective complex IV assembly as well. Therefore, we propose that complex III is central for MRC maturation and SC formation. Our results challenge the notion that SC biogenesis requires the pre-formation of fully assembled individual complexes. In contrast, they support a cooperative-assembly model in which the main role of complex III in SCs is to provide a structural and functional platform for the completion of overall MRC biogenesis.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/química , Complejo I de Transporte de Electrón/metabolismo , Proteómica/métodos , Línea Celular , Complejo I de Transporte de Electrón/genética , Complejo III de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Estabilidad de Enzimas , Humanos , Mitocondrias/metabolismo , Mutación , NAD/metabolismo
4.
Biochem Soc Trans ; 52(2): 873-885, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38526156

RESUMEN

In eukaryotic cells, mitochondria perform cellular respiration through a series of redox reactions ultimately reducing molecular oxygen to water. The system responsible for this process is the respiratory chain or electron transport system (ETS) composed of complexes I-IV. Due to its function, the ETS is the main source of reactive oxygen species (ROS), generating them on both sides of the mitochondrial inner membrane, i.e. the intermembrane space (IMS) and the matrix. A correct balance between ROS generation and scavenging is important for keeping the cellular redox homeostasis and other important aspects of cellular physiology. However, ROS generated in the mitochondria are important signaling molecules regulating mitochondrial biogenesis and function. The IMS contains a large number of redox sensing proteins, containing specific Cys-rich domains, that are involved in ETS complex biogenesis. The large majority of these proteins function as cytochrome c oxidase (COX) assembly factors, mainly for the handling of copper ions necessary for the formation of the redox reactive catalytic centers. A particular case of ROS-regulated COX assembly factor is COA8, whose intramitochondrial levels are increased by oxidative stress, promoting COX assembly and/or protecting the enzyme from oxidative damage. In this review, we will discuss the current knowledge concerning the role played by ROS in regulating mitochondrial activity and biogenesis, focusing on the COX enzyme and with a special emphasis on the functional role exerted by the redox sensitive Cys residues contained in the COX assembly factors.


Asunto(s)
Complejo IV de Transporte de Electrones , Mitocondrias , Oxidación-Reducción , Especies Reactivas de Oxígeno , Complejo IV de Transporte de Electrones/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Humanos , Animales , Estrés Oxidativo
5.
PLoS Biol ; 19(4): e3001166, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33826607

RESUMEN

Neural stem cell (NSC) transplantation induces recovery in animal models of central nervous system (CNS) diseases. Although the replacement of lost endogenous cells was originally proposed as the primary healing mechanism of NSC grafts, it is now clear that transplanted NSCs operate via multiple mechanisms, including the horizontal exchange of therapeutic cargoes to host cells via extracellular vesicles (EVs). EVs are membrane particles trafficking nucleic acids, proteins, metabolites and metabolic enzymes, lipids, and entire organelles. However, the function and the contribution of these cargoes to the broad therapeutic effects of NSCs are yet to be fully understood. Mitochondrial dysfunction is an established feature of several inflammatory and degenerative CNS disorders, most of which are potentially treatable with exogenous stem cell therapeutics. Herein, we investigated the hypothesis that NSCs release and traffic functional mitochondria via EVs to restore mitochondrial function in target cells. Untargeted proteomics revealed a significant enrichment of mitochondrial proteins spontaneously released by NSCs in EVs. Morphological and functional analyses confirmed the presence of ultrastructurally intact mitochondria within EVs with conserved membrane potential and respiration. We found that the transfer of these mitochondria from EVs to mtDNA-deficient L929 Rho0 cells rescued mitochondrial function and increased Rho0 cell survival. Furthermore, the incorporation of mitochondria from EVs into inflammatory mononuclear phagocytes restored normal mitochondrial dynamics and cellular metabolism and reduced the expression of pro-inflammatory markers in target cells. When transplanted in an animal model of multiple sclerosis, exogenous NSCs actively transferred mitochondria to mononuclear phagocytes and induced a significant amelioration of clinical deficits. Our data provide the first evidence that NSCs deliver functional mitochondria to target cells via EVs, paving the way for the development of novel (a)cellular approaches aimed at restoring mitochondrial dysfunction not only in multiple sclerosis, but also in degenerative neurological diseases.


Asunto(s)
Vesículas Extracelulares/metabolismo , Mitocondrias/metabolismo , Células-Madre Neurales/metabolismo , Animales , Transporte Biológico , Células Cultivadas , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/ultraestructura
6.
EMBO Rep ; 23(8): e54825, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35699132

RESUMEN

The mitochondrial respiratory chain (MRC) is composed of four multiheteromeric enzyme complexes. According to the endosymbiotic origin of mitochondria, eukaryotic MRC derives from ancestral proteobacterial respiratory structures consisting of a minimal set of complexes formed by a few subunits associated with redox prosthetic groups. These enzymes, which are the "core" redox centers of respiration, acquired additional subunits, and increased their complexity throughout evolution. Cytochrome c oxidase (COX), the terminal component of MRC, has a highly interspecific heterogeneous composition. Mammalian COX consists of 14 different polypeptides, of which COX7B is considered the evolutionarily youngest subunit. We applied proteomic, biochemical, and genetic approaches to investigate the COX composition in the invertebrate model Drosophila melanogaster. We identified and characterized a novel subunit which is widely different in amino acid sequence, but similar in secondary and tertiary structures to COX7B, and provided evidence that this object is in fact replacing the latter subunit in virtually all protostome invertebrates. These results demonstrate that although individual structures may differ the composition of COX is functionally conserved between vertebrate and invertebrate species.


Asunto(s)
Drosophila melanogaster , Complejo IV de Transporte de Electrones , Secuencia de Aminoácidos , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Mamíferos/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteómica
7.
J Med Genet ; 58(3): 155-167, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32439808

RESUMEN

BACKGROUND: Mitochondria provide ATP through the process of oxidative phosphorylation, physically located in the inner mitochondrial membrane (IMM). The mitochondrial contact site and organising system (MICOS) complex is known as the 'mitoskeleton' due to its role in maintaining IMM architecture. APOO encodes MIC26, a component of MICOS, whose exact function in its maintenance or assembly has still not been completely elucidated. METHODS: We have studied a family in which the most affected subject presented progressive developmental delay, lactic acidosis, muscle weakness, hypotonia, weight loss, gastrointestinal and body temperature dysautonomia, repetitive infections, cognitive impairment and autistic behaviour. Other family members showed variable phenotype presentation. Whole exome sequencing was used to screen for pathological variants. Patient-derived skin fibroblasts were used to confirm the pathogenicity of the variant found in APOO. Knockout models in Drosophila melanogaster and Saccharomyces cerevisiae were employed to validate MIC26 involvement in MICOS assembly and mitochondrial function. RESULTS: A likely pathogenic c.350T>C transition was found in APOO predicting an I117T substitution in MIC26. The mutation caused impaired processing of the protein during import and faulty insertion into the IMM. This was associated with altered MICOS assembly and cristae junction disruption. The corresponding mutation in MIC26 or complete loss was associated with mitochondrial structural and functional deficiencies in yeast and D. melanogaster models. CONCLUSION: This is the first case of pathogenic mutation in APOO, causing altered MICOS assembly and neuromuscular impairment. MIC26 is involved in the assembly or stability of MICOS in humans, yeast and flies.


Asunto(s)
Apolipoproteínas/genética , Trastorno Autístico/genética , Disfunción Cognitiva/genética , Proteínas de la Membrana/genética , Miopatías Mitocondriales/genética , Proteínas Mitocondriales/genética , Proteínas de Saccharomyces cerevisiae/genética , Acidosis Láctica/genética , Acidosis Láctica/patología , Animales , Trastorno Autístico/patología , Disfunción Cognitiva/patología , Drosophila melanogaster/genética , Fibroblastos/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Miopatías Mitocondriales/epidemiología , Miopatías Mitocondriales/patología , Unión Proteica , Saccharomyces cerevisiae/genética
8.
Neurobiol Dis ; 141: 104880, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32344152

RESUMEN

Mitochondrial ribosomal protein large 24 (MRPL24) is 1 of the 82 protein components of mitochondrial ribosomes, playing an essential role in the mitochondrial translation process. We report here on a baby girl with cerebellar atrophy, choreoathetosis of limbs and face, intellectual disability and a combined defect of complexes I and IV in muscle biopsy, caused by a homozygous missense mutation identified in MRPL24. The variant predicts a Leu91Pro substitution at an evolutionarily conserved site. Using human mutant cells and the zebrafish model, we demonstrated the pathological role of the identified variant. In fact, in fibroblasts we observed a significant reduction of MRPL24 protein and of mitochondrial respiratory chain complex I and IV subunits, as well a markedly reduced synthesis of the mtDNA-encoded peptides. In zebrafish we demonstrated that the orthologue gene is expressed in metabolically active tissues, and that gene knockdown induced locomotion impairment, structural defects and low ATP production. The motor phenotype was complemented by human WT but not mutant cRNA. Moreover, sucrose density gradient fractionation showed perturbed assembly of large subunit mitoribosomal proteins, suggesting that the mutation leads to a conformational change in MRPL24, which is expected to cause an aberrant interaction of the protein with other components of the 39S mitoribosomal subunit.


Asunto(s)
Proteínas Mitocondriales/genética , Trastornos del Movimiento/genética , Proteínas Ribosómicas/genética , Animales , Cerebelo/patología , Femenino , Humanos , Lactante , Leviviridae , Masculino , Trastornos del Movimiento/patología , Músculo Cuádriceps/patología , Pez Cebra
9.
Mol Genet Metab ; 131(3): 349-357, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33153867

RESUMEN

Isolated complex I (CI) deficiency is the most common cause of oxidative phosphorylation (OXPHOS) dysfunction. Whole-exome sequencing identified biallelic mutations in NDUFA8 (c.[293G > T]; [293G > T], encoding for an accessory subunit of CI, in two siblings with a favorable clinical evolution. The individuals reported here are practically asymptomatic, with the exception of slight failure to thrive and some language difficulties at the age of 6 and 9 years, respectively. These observations are remarkable since the vast majority of patients with CI deficiency, including the only NDUFA8 patient reported so far, showed an extremely poor clinical outcome. Western blot studies demonstrated that NDUFA8 protein was strongly reduced in the patients' fibroblasts and muscle extracts. In addition, there was a marked and specific decrease in the steady-state levels of CI subunits. BN-PAGE demonstrated an isolated defect in the assembly and the activity of CI with impaired supercomplexes formation and abnormal accumulation of CI subassemblies. Confocal microscopy analysis in fibroblasts showed rounder mitochondria and diminished branching degree of the mitochondrial network. Functional complementation studies demonstrated disease-causality for the identified mutation as lentiviral transduction with wild-type NDUFA8 cDNA restored the steady-state levels of CI subunits and completely recovered the deficient enzymatic activity in immortalized mutant fibroblasts. In summary, we provide additional evidence of the involvement of NDUFA8 as a mitochondrial disease-causing gene associated with altered mitochondrial morphology, CI deficiency, impaired supercomplexes formation, and very mild progression of the disease.


Asunto(s)
Predisposición Genética a la Enfermedad , Enfermedades Mitocondriales/genética , NADH Deshidrogenasa/genética , Fosforilación Oxidativa , Niño , Femenino , Fibroblastos/metabolismo , Humanos , Masculino , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/patología , Mitocondrias/genética , Mitocondrias/patología , Enfermedades Mitocondriales/patología , Hermanos , Secuenciación del Exoma
10.
Am J Hum Genet ; 96(4): 640-50, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25772934

RESUMEN

Microphthalmia with linear skin defects (MLS) syndrome is an X-linked male-lethal disorder also known as MIDAS (microphthalmia, dermal aplasia, and sclerocornea). Additional clinical features include neurological and cardiac abnormalities. MLS syndrome is genetically heterogeneous given that heterozygous mutations in HCCS or COX7B have been identified in MLS-affected females. Both genes encode proteins involved in the structure and function of complexes III and IV, which form the terminal segment of the mitochondrial respiratory chain (MRC). However, not all individuals with MLS syndrome carry a mutation in either HCCS or COX7B. The majority of MLS-affected females have severe skewing of X chromosome inactivation, suggesting that mutations in HCCS, COX7B, and other as-yet-unidentified X-linked gene(s) cause selective loss of cells in which the mutated X chromosome is active. By applying whole-exome sequencing and filtering for X-chromosomal variants, we identified a de novo nonsense mutation in NDUFB11 (Xp11.23) in one female individual and a heterozygous 1-bp deletion in a second individual, her asymptomatic mother, and an affected aborted fetus of the subject's mother. NDUFB11 encodes one of 30 poorly characterized supernumerary subunits of NADH:ubiquinone oxidoreductase, known as complex I (cI), the first and largest enzyme of the MRC. By shRNA-mediated NDUFB11 knockdown in HeLa cells, we demonstrate that NDUFB11 is essential for cI assembly and activity as well as cell growth and survival. These results demonstrate that X-linked genetic defects leading to the complete inactivation of complex I, III, or IV underlie MLS syndrome. Our data reveal an unexpected role of cI dysfunction in a developmental phenotype, further underscoring the existence of a group of mitochondrial diseases associated with neurocutaneous manifestations.


Asunto(s)
Complejo I de Transporte de Electrón/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Microftalmía/genética , Anomalías Cutáneas/genética , Inactivación del Cromosoma X/genética , Encéfalo/patología , Codón sin Sentido/genética , Exoma/genética , Femenino , Técnicas de Silenciamiento del Gen , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Células HeLa , Humanos , Inmunohistoquímica , Cariotipificación , Imagen por Resonancia Magnética , Microftalmía/patología , Linaje , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Anomalías Cutáneas/patología
11.
Acta Neuropathol ; 135(3): 409-425, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29270838

RESUMEN

Mitochondrial complex I deficiency occurs in the substantia nigra of individuals with Parkinson's disease. It is generally believed that this phenomenon is caused by accumulating mitochondrial DNA damage in neurons and that it contributes to the process of neurodegeneration. We hypothesized that if these theories are correct, complex I deficiency should extend beyond the substantia nigra to other affected brain regions in Parkinson's disease and correlate tightly with neuronal mitochondrial DNA damage. To test our hypothesis, we employed a combination of semiquantitative immunohistochemical analyses, Western blot and activity measurements, to assess complex I quantity and function in multiple brain regions from an extensively characterized population-based cohort of idiopathic Parkinson's disease (n = 18) and gender and age matched healthy controls (n = 11). Mitochondrial DNA was assessed in single neurons from the same areas by real-time PCR. Immunohistochemistry showed that neuronal complex I deficiency occurs throughout the Parkinson's disease brain, including areas spared by the neurodegenerative process such as the cerebellum. Activity measurements in brain homogenate confirmed a moderate decrease of complex I function, whereas Western blot was less sensitive, detecting only a mild reduction, which did not reach statistical significance at the group level. With the exception of the substantia nigra, neuronal complex I loss showed no correlation with the load of somatic mitochondrial DNA damage. Interestingly, α-synuclein aggregation was less common in complex I deficient neurons in the substantia nigra. We show that neuronal complex I deficiency is a widespread phenomenon in the Parkinson's disease brain which, contrary to mainstream theory, does not follow the anatomical distribution of neurodegeneration and is not associated with the neuronal load of mitochondrial DNA mutation. Our findings suggest that complex I deficiency in Parkinson's disease can occur independently of mitochondrial DNA damage and may not have a pathogenic role in the neurodegenerative process.


Asunto(s)
Encéfalo/metabolismo , Complejo I de Transporte de Electrón/deficiencia , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Anciano , Anciano de 80 o más Años , Encéfalo/patología , Daño del ADN , ADN Mitocondrial/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/patología , Enfermedades Mitocondriales/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/patología , Enfermedad de Parkinson/patología , Estudios Prospectivos , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , alfa-Sinucleína/metabolismo
12.
Am J Hum Genet ; 95(3): 315-25, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25175347

RESUMEN

Cytochrome c oxidase (COX) deficiency is a frequent biochemical abnormality in mitochondrial disorders, but a large fraction of cases remains genetically undetermined. Whole-exome sequencing led to the identification of APOPT1 mutations in two Italian sisters and in a third Turkish individual presenting severe COX deficiency. All three subjects presented a distinctive brain MRI pattern characterized by cavitating leukodystrophy, predominantly in the posterior region of the cerebral hemispheres. We then found APOPT1 mutations in three additional unrelated children, selected on the basis of these particular MRI features. All identified mutations predicted the synthesis of severely damaged protein variants. The clinical features of the six subjects varied widely from acute neurometabolic decompensation in late infancy to subtle neurological signs, which appeared in adolescence; all presented a chronic, long-surviving clinical course. We showed that APOPT1 is targeted to and localized within mitochondria by an N-terminal mitochondrial targeting sequence that is eventually cleaved off from the mature protein. We then showed that APOPT1 is virtually absent in fibroblasts cultured in standard conditions, but its levels increase by inhibiting the proteasome or after oxidative challenge. Mutant fibroblasts showed reduced amount of COX holocomplex and higher levels of reactive oxygen species, which both shifted toward control values by expressing a recombinant, wild-type APOPT1 cDNA. The shRNA-mediated knockdown of APOPT1 in myoblasts and fibroblasts caused dramatic decrease in cell viability. APOPT1 mutations are responsible for infantile or childhood-onset mitochondrial disease, hallmarked by the combination of profound COX deficiency with a distinctive neuroimaging presentation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Complejo IV de Transporte de Electrones/metabolismo , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Proteínas Mitocondriales/genética , Mutación/genética , Adolescente , Adulto , Células Cultivadas , Niño , Preescolar , Deficiencia de Citocromo-c Oxidasa , Complejo IV de Transporte de Electrones/genética , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Lactante , Leucoencefalopatías/enzimología , Imagen por Resonancia Magnética , Masculino , Mitocondrias/metabolismo , Mioblastos/metabolismo , Mioblastos/patología
13.
Biochim Biophys Acta Mol Basis Dis ; 1863(4): 961-967, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28132884

RESUMEN

The mitochondrial Elongation Factor Tu (EF-Tu), encoded by the TUFM gene, is a highly conserved GTPase, which is part of the mitochondrial protein translation machinery. In its activated form it delivers the aminoacyl-tRNAs to the A site of the mitochondrial ribosome. We report here on a baby girl with severe infantile macrocystic leukodystrophy with micropolygyria and a combined defect of complexes I and IV in muscle biopsy, caused by a novel mutation identified in TUFM. Using human mutant cells and the yeast model, we demonstrate the pathological role of the novel variant. Moreover, results of a molecular modeling study suggest that the mutant is inactive in mitochondrial polypeptide chain elongation, probably as a consequence of its reduced ability to bind mitochondrial aa-tRNAs. Four patients have so far been described with mutations in TUFM, and, following the first description of the disease in a single patient, we describe similar clinical and neuroradiological features in an additional patient.


Asunto(s)
Secuencia de Bases , ADN Mitocondrial/genética , Leucoencefalopatías/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Extensión de la Cadena Peptídica de Translación , Factor Tu de Elongación Peptídica/genética , Eliminación de Secuencia , ADN Mitocondrial/metabolismo , Femenino , Humanos , Leucoencefalopatías/metabolismo , Masculino , Mitocondrias/patología , Proteínas Mitocondriales/metabolismo , Factor Tu de Elongación Peptídica/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
14.
J Med Genet ; 53(12): 846-849, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27683825

RESUMEN

BACKGROUND: Assembly of cytochrome c oxidase (COX, complex IV, cIV), the terminal component of the mitochondrial respiratory chain, is assisted by several factors, most of which are conserved from yeast to humans. However, some of them, including COA7, are found in humans but not in yeast. COA7 is a 231aa-long mitochondrial protein present in animals, containing five Sel1-like tetratricopeptide repeat sequences, which are likely to interact with partner proteins. METHODS: Whole exome sequencing was carried out on a 19 year old woman, affected by early onset, progressive severe ataxia and peripheral neuropathy, mild cognitive impairment and a cavitating leukodystrophy of the brain with spinal cord hypotrophy. Biochemical analysis of the mitochondrial respiratory chain revealed the presence of isolated deficiency of cytochrome c oxidase (COX) activity in skin fibroblasts and skeletal muscle. Mitochondrial localization studies were carried out in isolated mitochondria and mitoplasts from immortalized control human fibroblasts. RESULTS: We found compound heterozygous mutations in COA7: a paternal c.410A>G, p.Y137C, and a maternal c.287+1G>T variants. Lentiviral-mediated expression of recombinant wild-type COA7 cDNA in the patient fibroblasts led to the recovery of the defect in COX activity and restoration of normal COX amount. In mitochondrial localization experiments, COA7 behaved as the soluble matrix protein Citrate Synthase. CONCLUSIONS: We report here the first patient carrying pathogenic mutations of COA7, causative of isolated COX deficiency and progressive neurological impairment. We also show that COA7 is a soluble protein localized to the matrix, rather than in the intermembrane space as previously suggested.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/metabolismo , Leucoencefalopatías/metabolismo , Proteínas Mitocondriales/genética , Mutación , Secuencia de Aminoácidos , Deficiencia de Citocromo-c Oxidasa/genética , Análisis Mutacional de ADN , Femenino , Humanos , Leucoencefalopatías/genética , Mitocondrias , Proteínas Mitocondriales/química , Alineación de Secuencia , Adulto Joven
15.
EMBO J ; 31(9): 2117-33, 2012 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22433842

RESUMEN

Mitochondria are dynamic subcellular organelles that convert nutrient intermediates into readily available energy equivalents. Optimal mitochondrial function is ensured by a highly evolved quality control system, coordinated by protein machinery that regulates a process of continual fusion and fission. In this work, we provide in vivo evidence that the ATP-independent metalloprotease OMA1 plays an essential role in the proteolytic inactivation of the dynamin-related GTPase OPA1 (optic atrophy 1). We also show that OMA1 deficiency causes a profound perturbation of the mitochondrial fusion-fission equilibrium that has important implications for metabolic homeostasis. Thus, ablation of OMA1 in mice results in marked transcriptional changes in genes of lipid and glucose metabolic pathways and substantial alterations in circulating blood parameters. Additionally, Oma1-mutant mice exhibit an increase in body weight due to increased adipose mass, hepatic steatosis, decreased energy expenditure and impaired thermogenenesis. These alterations are especially significant under metabolic stress conditions, indicating that an intact OMA1-OPA1 system is essential for developing the appropriate adaptive response to different metabolic stressors such as a high-fat diet or cold-shock. This study provides the first description of an unexpected role in energy metabolism for the metalloprotease OMA1 and reinforces the importance of mitochondrial quality control for normal metabolic function.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Metaloendopeptidasas/deficiencia , Metaloproteasas/deficiencia , Proteínas Mitocondriales/deficiencia , Obesidad/metabolismo , Termogénesis/fisiología , Adipocitos Marrones/metabolismo , Animales , Glucemia/análisis , Dieta Alta en Grasa , Embrión de Mamíferos , Fibroblastos/metabolismo , Metabolismo de los Lípidos , Metaloendopeptidasas/genética , Metaloproteasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/fisiología , Proteínas Mitocondriales/genética
16.
Mol Genet Metab ; 119(3): 214-222, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27623250

RESUMEN

We report here the case of a young male who started to show verbal fluency disturbance, clumsiness and gait anomalies at the age of 3.5years and presented bilateral striatal necrosis. Clinically, the diagnosis was compatible with Leigh syndrome but the underlying molecular defect remained elusive even after exome analysis using autosomal/X-linked recessive or de novo models. Dosage of respiratory chain activity on fibroblasts, but not in muscle, underlined a deficit in complex I. Re-analysis of heterozygous probably pathogenic variants, inherited from one healthy parent, identified the p.Ala178Pro in NDUFAF6, a complex I assembly factor. RNA analysis showed an almost mono-allelic expression of the mutated allele in blood and fibroblasts and puromycin treatment on cultured fibroblasts did not lead to the rescue of the maternal allele expression, not supporting the involvement of nonsense-mediated RNA decay mechanism. Complementation assay underlined a recovery of complex I activity after transduction of the wild-type gene. Since the second mutation was not detected and promoter methylation analysis resulted normal, we hypothesized a non-exonic event in the maternal allele affecting a regulatory element that, in conjunction with the paternal mutation, leads to the autosomal recessive disorder and the different allele expression in various tissues. This paper confirms NDUFAF6 as a genuine morbid gene and proposes the coupling of exome sequencing with mRNA analysis as a method useful for enhancing the exome sequencing detection rate when the simple application of classical inheritance models fails.


Asunto(s)
Exoma/genética , Enfermedad de Leigh/genética , Proteínas Mitocondriales/genética , Trastornos del Habla/genética , Alelos , Preescolar , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Enfermedad de Leigh/fisiopatología , Masculino , Mutación , Linaje , Fenotipo , ARN Mensajero/genética , Trastornos del Habla/fisiopatología , Degeneración Estriatonigral/congénito , Degeneración Estriatonigral/genética , Degeneración Estriatonigral/fisiopatología
17.
Nat Genet ; 38(5): 570-5, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16582910

RESUMEN

The mitochondrial (mt) DNA depletion syndromes (MDDS) are genetic disorders characterized by a severe, tissue-specific decrease of mtDNA copy number, leading to organ failure. There are two main clinical presentations: myopathic (OMIM 609560) and hepatocerebral (OMIM 251880). Known mutant genes, including TK2, SUCLA2, DGUOK and POLG, account for only a fraction of MDDS cases. We found a new locus for hepatocerebral MDDS on chromosome 2p21-23 and prioritized the genes on this locus using a new integrative genomics strategy. One of the top-scoring candidates was the human ortholog of the mouse kidney disease gene Mpv17. We found disease-segregating mutations in three families with hepatocerebral MDDS and demonstrated that, contrary to the alleged peroxisomal localization of the MPV17 gene product, MPV17 is a mitochondrial inner membrane protein, and its absence or malfunction causes oxidative phosphorylation (OXPHOS) failure and mtDNA depletion, not only in affected individuals but also in Mpv17-/- mice.


Asunto(s)
ADN Mitocondrial/genética , Membranas Intracelulares/metabolismo , Hepatopatías/genética , Proteínas de la Membrana/genética , Mitocondrias/metabolismo , Mutación , Secuencia de Aminoácidos , Animales , Células Cultivadas , Cromosomas Humanos Par 2 , Clonación Molecular , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Linaje , Síndrome
18.
Biochim Biophys Acta ; 1827(3): 285-93, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23168492

RESUMEN

The mammalian Complex III (CIII) assembly process is yet to be completely understood. There is still a lack in understanding of how the structural subunits are put together and which additional factors are involved. Here we describe the identification and characterization of LYRM7, a human protein displaying high sequence homology to the Saccharomyces cerevisiae protein Mzm1, which was recently shown as an assembly factor for Rieske Fe-S protein incorporation into the yeast cytochrome bc(1) complex. We conclude that human LYRM7, which we propose to be renamed MZM1L (MZM1-like), works as a human Rieske Fe-S protein (UQCRFS1) chaperone, binding to this subunit within the mitochondrial matrix and stabilizing it prior to its translocation and insertion into the late CIII dimeric intermediate within the mitochondrial inner membrane. Thus, LYRM7/MZM1L is a novel human CIII assembly factor involved in the UQCRFS1 insertion step, which enables formation of the mature and functional CIII enzyme.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Proteínas Hierro-Azufre/metabolismo , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Secuencia de Aminoácidos , Animales , Complejo III de Transporte de Electrones/fisiología , Células HEK293 , Células HeLa , Humanos , Ratones , Proteínas Mitocondriales/fisiología , Chaperonas Moleculares/fisiología , Datos de Secuencia Molecular
19.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167033, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38280294

RESUMEN

Mitochondrial disorders are hallmarked by the dysfunction of oxidative phosphorylation (OXPHOS) yet are highly heterogeneous at the clinical and genetic levels. Striking tissue-specific pathological manifestations are a poorly understood feature of these conditions, even if the disease-causing genes are ubiquitously expressed. To investigate the functional basis of this phenomenon, we analyzed several OXPHOS-related bioenergetic parameters, including oxygen consumption rates, electron transfer system (ETS)-related coenzyme Q (mtCoQ) redox state and production of reactive oxygen species (ROS) in mouse brain and liver mitochondria fueled by different substrates. In addition, we determined how these functional parameters are affected by ETS impairment in a tissue-specific manner using pathologically relevant mouse models lacking either Ndufs4 or Ttc19, leading to Complex I (CI) or Complex III (CIII) deficiency, respectively. Detailed OXPHOS analysis revealed striking differences between brain and liver mitochondria in the capacity of the different metabolic substrates to fuel the ETS, reduce the ETS-related mtCoQ, and to induce ROS production. In addition, ETS deficiency due to either CI or CIII dysfunction had a much greater impact on the intrinsic bioenergetic parameters of brain compared with liver mitochondria. These findings are discussed in terms of the still rather mysterious tissue-specific manifestations of mitochondrial disease.


Asunto(s)
Mitocondrias Hepáticas , Enfermedades Mitocondriales , Animales , Ratones , Mitocondrias Hepáticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Metabolismo Energético , Encéfalo/metabolismo , Enfermedades Mitocondriales/metabolismo , Complejo I de Transporte de Electrón/metabolismo
20.
Nat Commun ; 15(1): 3631, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684731

RESUMEN

Idiopathic Parkinson's disease (iPD) is believed to have a heterogeneous pathophysiology, but molecular disease subtypes have not been identified. Here, we show that iPD can be stratified according to the severity of neuronal respiratory complex I (CI) deficiency, and identify two emerging disease subtypes with distinct molecular and clinical profiles. The CI deficient (CI-PD) subtype accounts for approximately a fourth of all cases, and is characterized by anatomically widespread neuronal CI deficiency, a distinct cell type-specific gene expression profile, increased load of neuronal mtDNA deletions, and a predilection for non-tremor dominant motor phenotypes. In contrast, the non-CI deficient (nCI-PD) subtype exhibits no evidence of mitochondrial impairment outside the dopaminergic substantia nigra and has a predilection for a tremor dominant phenotype. These findings constitute a step towards resolving the biological heterogeneity of iPD with implications for both mechanistic understanding and treatment strategies.


Asunto(s)
ADN Mitocondrial , Complejo I de Transporte de Electrón , Complejo I de Transporte de Electrón/deficiencia , Mitocondrias , Enfermedades Mitocondriales , Enfermedad de Parkinson , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Humanos , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Masculino , ADN Mitocondrial/genética , Femenino , Mitocondrias/metabolismo , Mitocondrias/genética , Anciano , Sustancia Negra/metabolismo , Sustancia Negra/patología , Persona de Mediana Edad , Fenotipo , Neuronas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA