Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 146(5): 1804-1811, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36349561

RESUMEN

Corpus callosum defects are frequent congenital cerebral disorders caused by mutations in more than 300 genes. These include genes implicated in corpus callosum development or function, as well as genes essential for mitochondrial physiology. However, in utero corpus callosum anomalies rarely raise a suspicion of mitochondrial disease and are characterized by a very large clinical heterogeneity. Here, we report a detailed pathological and neuro-histopathological investigation of nine foetuses from four unrelated families with prenatal onset of corpus callosum anomalies, sometimes associated with other cerebral or extra-cerebral defects. Next generation sequencing allowed the identification of novel pathogenic variants in three different nuclear genes previously reported in mitochondrial diseases: TIMMDC1, encoding a Complex I assembly factor never involved before in corpus callosum defect; MRPS22, a protein of the small mitoribosomal subunit; and EARS2, the mitochondrial tRNA-glutamyl synthetase. The present report describes the antenatal histopathological findings in mitochondrial diseases and expands the genetic spectrum of antenatal corpus callosum anomalies establishing OXPHOS function as an important factor for corpus callosum biogenesis. We propose that, when observed, antenatal corpus callosum anomalies should raise suspicion of mitochondrial disease and prenatal genetic counselling should be considered.


Asunto(s)
Cuerpo Calloso , Enfermedades Mitocondriales , Humanos , Femenino , Embarazo , Cuerpo Calloso/patología , Agenesia del Cuerpo Calloso/genética , Agenesia del Cuerpo Calloso/patología , Enfermedades Mitocondriales/genética , Mitocondrias/patología , Mutación , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales
2.
PLoS Genet ; 15(6): e1008085, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31170154

RESUMEN

Mitochondrial dynamics is an essential physiological process controlling mitochondrial content mixing and mobility to ensure proper function and localization of mitochondria at intracellular sites of high-energy demand. Intriguingly, for yet unknown reasons, severe impairment of mitochondrial fusion drastically affects mtDNA copy number. To decipher the link between mitochondrial dynamics and mtDNA maintenance, we studied mouse embryonic fibroblasts (MEFs) and mouse cardiomyocytes with disruption of mitochondrial fusion. Super-resolution microscopy revealed that loss of outer mitochondrial membrane (OMM) fusion, but not inner mitochondrial membrane (IMM) fusion, leads to nucleoid clustering. Remarkably, fluorescence in situ hybridization (FISH), bromouridine labeling in MEFs and assessment of mitochondrial transcription in tissue homogenates revealed that abolished OMM fusion does not affect transcription. Furthermore, the profound mtDNA depletion in mouse hearts lacking OMM fusion is not caused by defective integrity or increased mutagenesis of mtDNA, but instead we show that mitochondrial fusion is necessary to maintain the stoichiometry of the protein components of the mtDNA replisome. OMM fusion is necessary for proliferating MEFs to recover from mtDNA depletion and for the marked increase of mtDNA copy number during postnatal heart development. Our findings thus link OMM fusion to replication and distribution of mtDNA.


Asunto(s)
ADN Mitocondrial/genética , Mitocondrias Cardíacas/genética , Dinámicas Mitocondriales/genética , Proteínas Mitocondriales/genética , Animales , Variaciones en el Número de Copia de ADN/genética , Replicación del ADN/genética , Fibroblastos , Humanos , Hibridación Fluorescente in Situ , Fusión de Membrana/genética , Ratones , Mitocondrias Cardíacas/metabolismo , Membranas Mitocondriales/metabolismo , Mutagénesis , Miocitos Cardíacos/metabolismo , Transcripción Genética
3.
Hum Mol Genet ; 28(9): 1445-1462, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30566640

RESUMEN

Mitochondria contain a dedicated translation system, which is responsible for the intramitochondrial synthesis of 13 mitochondrial DNA (mtDNA)-encoded polypeptides essential for the biogenesis of oxidative phosphorylation (OXPHOS) complexes I and III-V. Mutations in nuclear genes encoding factors involved in mitochondrial translation result in isolated or multiple OXPHOS deficiencies and mitochondrial disease. Here, we report the identification of disease-causing variants in the MRPS28 gene, encoding the small mitoribosomal subunit (mtSSU) protein bS1m in a patient with intrauterine growth retardation, craniofacial dysmorphism and developmental delay. Whole exome sequencing helped identify a seemingly homozygous missense variant NM_014018.2:c.356A>G, p.(Lys119Arg) which affected a highly conserved lysine residue. The variant was present in the mother in a heterozygous state, but not in the father who likely carried a large deletion spanning exon 2 and parts of introns 1 and 2 that could account for the apparent homozygosity of the patient. Polymerase chain reaction (PCR) amplification and Sanger sequencing of MRPS28 cDNA from patient fibroblasts revealed the presence of a truncated MRPS28 transcript, which lacked exon 2. Molecular and biochemical characterization of patient fibroblasts revealed a decrease in the abundance of the bS1m protein, decreased abundance of assembled mtSSU and inhibited mitochondrial translation. Consequently, OXPHOS biogenesis and cellular respiration were compromised in these cells. Expression of wild-type MRPS28 restored mitoribosomal assembly, mitochondrial translation and OXPHOS biogenesis, thereby demonstrating the deleterious nature of the identified MRPS28 variants. Thus, MRPS28 joins the increasing number of nuclear genes encoding mitoribosomal structural proteins linked to mitochondrial disease.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Retardo del Crecimiento Fetal/diagnóstico , Retardo del Crecimiento Fetal/genética , Proteínas Mitocondriales/genética , Mutación , Subunidades de Proteína/genética , Proteínas Ribosómicas/genética , Alelos , Secuencia de Aminoácidos , Respiración de la Célula/genética , Anomalías Craneofaciales/diagnóstico , Anomalías Craneofaciales/genética , Análisis Mutacional de ADN , Femenino , Fibroblastos/metabolismo , Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Imagen por Resonancia Magnética , Masculino , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/química , Modelos Moleculares , Fenotipo , Biosíntesis de Proteínas , Conformación Proteica , Proteínas Ribosómicas/química , Relación Estructura-Actividad , Secuenciación del Exoma
4.
Am J Hum Genet ; 102(4): 685-695, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29576219

RESUMEN

Biogenesis of the mitochondrial oxidative phosphorylation system, which produces the bulk of ATP for almost all eukaryotic cells, depends on the translation of 13 mtDNA-encoded polypeptides by mitochondria-specific ribosomes in the mitochondrial matrix. These mitoribosomes are dual-origin ribonucleoprotein complexes, which contain mtDNA-encoded rRNAs and tRNAs and ∼80 nucleus-encoded proteins. An increasing number of gene mutations that impair mitoribosomal function and result in multiple OXPHOS deficiencies are being linked to human mitochondrial diseases. Using exome sequencing in two unrelated subjects presenting with sensorineural hearing impairment, mild developmental delay, hypoglycemia, and a combined OXPHOS deficiency, we identified mutations in the gene encoding the mitochondrial ribosomal protein S2, which has not previously been implicated in disease. Characterization of subjects' fibroblasts revealed a decrease in the steady-state amounts of mutant MRPS2, and this decrease was shown by complexome profiling to prevent the assembly of the small mitoribosomal subunit. In turn, mitochondrial translation was inhibited, resulting in a combined OXPHOS deficiency detectable in subjects' muscle and liver biopsies as well as in cultured skin fibroblasts. Reintroduction of wild-type MRPS2 restored mitochondrial translation and OXPHOS assembly. The combination of lactic acidemia, hypoglycemia, and sensorineural hearing loss, especially in the presence of a combined OXPHOS deficiency, should raise suspicion for a ribosomal-subunit-related mitochondrial defect, and clinical recognition could allow for a targeted diagnostic approach. The identification of MRPS2 as an additional gene related to mitochondrial disease further expands the genetic and phenotypic spectra of OXPHOS deficiencies caused by impaired mitochondrial translation.


Asunto(s)
Alelos , Pérdida Auditiva Sensorineural/genética , Hipoglucemia/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Mutación/genética , Proteínas Ribosómicas/genética , Secuencia de Aminoácidos , Preescolar , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Femenino , Fibroblastos/metabolismo , Pérdida Auditiva Sensorineural/complicaciones , Humanos , Hipoglucemia/complicaciones , Lactante , Recién Nacido , Masculino , Enfermedades Mitocondriales/complicaciones , Proteínas Mitocondriales/química , Fosforilación Oxidativa , Subunidades de Proteína/genética , ARN Ribosómico/genética , Proteínas Ribosómicas/química
5.
Mol Genet Metab ; 134(3): 267-273, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34620555

RESUMEN

Most mitochondrial proteins are synthesized in the cytosol and targeted to mitochondria via N-terminal mitochondrial targeting signals (MTS) that are proteolytically removed upon import. Sometimes, MTS removal is followed by a cleavage of an octapeptide by the mitochondrial intermediate peptidase (MIP), encoded by the MIPEP gene. Previously, MIPEP variants were linked to four cases of multisystemic disorder presenting with cardiomyopathy, developmental delay, hypotonia and infantile lethality. We report here a patient carrying compound heterozygous MIPEP variants-one was not previously linked to mitochondrial disease-who did not have cardiomyopathy and who is alive at the age of 20 years. This patient had developmental delay, global hypotonia, mild optic neuropathy and mild ataxia. Functional characterization of patient fibroblasts and HEK293FT cells carrying MIPEP hypomorphic alleles demonstrated that deficient MIP activity was linked to impaired post-import processing of subunits from four of the five OXPHOS complexes and decreased abundance and activity of some of these complexes in human cells possibly underlying the development of mitochondrial disease. Thus, our work expands the genetic and clinical spectrum of MIPEP-linked disease and establishes MIP as an important regulator of OXPHOS biogenesis and function in human cells.


Asunto(s)
Cardiomiopatías/fisiopatología , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Enfermedades Mitocondriales/genética , Fenotipo , Alelos , Fibroblastos/metabolismo , Expresión Génica , Células HEK293 , Humanos , Masculino , Enfermedades Mitocondriales/complicaciones , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/fisiopatología , Mutación , Adulto Joven
7.
Dev Med Child Neurol ; 63(6): 705-711, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33511646

RESUMEN

AIM: To investigate cerebral blood flow (CBF) in acute episodes of Leigh syndrome compared with basal state in patients carrying pathogenic mitochondrial disease gene variants responsible for neurometabolic disorders. METHOD: Arterial spin labelling (ASL) magnetic resonance imaging (MRI) sequences were used to measure CBF in 27 patients with mitochondrial respiratory chain enzyme deficiencies, ascribed to pathogenic variants of reported disease genes who were undergoing either urgent neuroimaging for acute episodes of Leigh syndrome (Group I: 15 MRI, seven females, eight males; mean age 7y; range 7mo-14y) or routine brain MRI (Group II: 15 MRI, eight females, seven males; mean age 5y 2mo; range 2mo-12y). RESULTS: Patients displayed markedly increased CBF in the striatum (2.8-fold greater, p<0.001 [1.05-2.53]) during acute episodes of Leigh syndrome compared to basal conditions. Detection of elevated CBF preceded identification of structural MRI lesions in four out of 15 cases. INTERPRETATION: Our results suggest that increased CBF is an overt hallmark of Leigh syndrome episodes and ASL MRI sequences should facilitate early diagnosis of acute episodes of Leigh syndrome, especially during the first attack in young children, when structural MRI is insufficiently informative.


Asunto(s)
Encéfalo/diagnóstico por imagen , Circulación Cerebrovascular/fisiología , Enfermedad de Leigh/diagnóstico por imagen , Enfermedades Mitocondriales/diagnóstico por imagen , Adolescente , Encéfalo/fisiopatología , Niño , Preescolar , Femenino , Humanos , Lactante , Enfermedad de Leigh/fisiopatología , Imagen por Resonancia Magnética , Masculino , Enfermedades Mitocondriales/fisiopatología , Marcadores de Spin
8.
Hum Mutat ; 41(2): 397-402, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31680380

RESUMEN

Pathogenic GFM1 variants have been linked to neurological phenotypes with or without liver involvement, but only a few cases have been reported in the literature. Here, we report clinical, biochemical, and neuroimaging findings from nine unrelated children carrying GFM1 variants, 10 of which were not previously reported. All patients presented with neurological involvement-mainly axial hypotonia and dystonia during the neonatal period-with five diagnosed with West syndrome; two children had liver involvement with cytolysis episodes or hepatic failure. While two patients died in infancy, six exhibited a stable clinical course. Brain magnetic resonance imaging showed the involvement of basal ganglia, brainstem, and periventricular white matter. Mutant EFG1 and OXPHOS proteins were decreased in patient's fibroblasts consistent with impaired mitochondrial translation. Thus, we expand the genetic spectrum of GFM1-linked disease and provide detailed clinical profiles of the patients that will improve the diagnostic success for other patients carrying GFM1 mutations.


Asunto(s)
Fibroblastos/metabolismo , Regulación de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Proteínas Mitocondriales/genética , Mutación , Neuroimagen , Factor G de Elongación Peptídica/genética , Alelos , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Bases de Datos Genéticas , Femenino , Estudios de Asociación Genética/métodos , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Mitocondrias/genética , Neuroimagen/métodos , Linaje
9.
EMBO J ; 35(23): 2566-2583, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27797820

RESUMEN

Despite being one of the most studied proteases in bacteria, very little is known about the role of ClpXP in mitochondria. We now present evidence that mammalian CLPP has an essential role in determining the rate of mitochondrial protein synthesis by regulating the level of mitoribosome assembly. Through a proteomic approach and the use of a catalytically inactive CLPP, we produced the first comprehensive list of possible mammalian ClpXP substrates involved in the regulation of mitochondrial translation, oxidative phosphorylation, and a number of metabolic pathways. We further show that the defect in mitoribosomal assembly is a consequence of the accumulation of ERAL1, a putative 12S rRNA chaperone, and novel ClpXP substrate. The presented data suggest that the timely removal of ERAL1 from the small ribosomal subunit is essential for the efficient maturation of the mitoribosome and a normal rate of mitochondrial translation.


Asunto(s)
Endopeptidasa Clp/metabolismo , Proteínas de Unión al GTP/metabolismo , Mitocondrias/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribosomas/metabolismo , Animales , Células Cultivadas , Fibroblastos/fisiología , Ratones , Ratones Noqueados , Biosíntesis de Proteínas
10.
Am J Hum Genet ; 101(2): 239-254, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28777931

RESUMEN

The synthesis of all 13 mitochondrial DNA (mtDNA)-encoded protein subunits of the human oxidative phosphorylation (OXPHOS) system is carried out by mitochondrial ribosomes (mitoribosomes). Defects in the stability of mitoribosomal proteins or mitoribosome assembly impair mitochondrial protein translation, causing combined OXPHOS enzyme deficiency and clinical disease. Here we report four autosomal-recessive pathogenic mutations in the gene encoding the small mitoribosomal subunit protein, MRPS34, in six subjects from four unrelated families with Leigh syndrome and combined OXPHOS defects. Whole-exome sequencing was used to independently identify all variants. Two splice-site mutations were identified, including homozygous c.321+1G>T in a subject of Italian ancestry and homozygous c.322-10G>A in affected sibling pairs from two unrelated families of Puerto Rican descent. In addition, compound heterozygous MRPS34 mutations were identified in a proband of French ancestry; a missense (c.37G>A [p.Glu13Lys]) and a nonsense (c.94C>T [p.Gln32∗]) variant. We demonstrated that these mutations reduce MRPS34 protein levels and the synthesis of OXPHOS subunits encoded by mtDNA. Examination of the mitoribosome profile and quantitative proteomics showed that the mitochondrial translation defect was caused by destabilization of the small mitoribosomal subunit and impaired monosome assembly. Lentiviral-mediated expression of wild-type MRPS34 rescued the defect in mitochondrial translation observed in skin fibroblasts from affected subjects, confirming the pathogenicity of MRPS34 mutations. Our data establish that MRPS34 is required for normal function of the mitoribosome in humans and furthermore demonstrate the power of quantitative proteomic analysis to identify signatures of defects in specific cellular pathways in fibroblasts from subjects with inherited disease.


Asunto(s)
ADN Mitocondrial/genética , Enfermedad de Leigh/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Proteínas Ribosómicas/genética , Subunidades Ribosómicas Pequeñas de Eucariotas/genética , Adolescente , Secuencia de Bases , Niño , Preescolar , Exoma/genética , Femenino , Humanos , Lactante , Enfermedad de Leigh/enzimología , Masculino , Mitocondrias/genética , Fosforilación Oxidativa , Proteómica , Empalme del ARN/genética , Análisis de Secuencia de ADN
11.
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
12.
J Med Genet ; 55(6): 378-383, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29358270

RESUMEN

BACKGROUND: Because the mitochondrial respiratory chain (RC) is ubiquitous, its deficiency can theoretically give rise to any symptom in any organ or tissue at any age with any mode of inheritance, owing to the twofold genetic origin of respiratory enzyme machinery, that is, nuclear and mitochondrial. Not all respiratory enzyme deficiencies are primary and secondary or artefactual deficiency is frequently observed, leading to a number of misleading conclusions and inappropriate investigations in clinical practice. This study is aimed at investigating the potential role of brain MRI in distinguishing primary RC deficiency from phenocopies and other aetiologies. METHODS: Starting from a large series of 189 patients (median age: 3.5 years (8 days-56 years), 58% males) showing signs of RC enzyme deficiency, for whom both brain MRIs and disease-causing mutations were available, we retrospectively studied the positive predictive value (PPV) and the positive likelihood ratio (LR+) of brain MRI imaging and its ability to discriminate between two groups: primary deficiency of the mitochondrial RC machinery and phenocopies. RESULTS: Detection of (1) brainstem hyperintensity with basal ganglia involvement (P≤0.001) and (2) lactate peak with either brainstem or basal ganglia hyperintensity was highly suggestive of primary RC deficiency (P≤0.01). Fourteen items had a PPV>95% and LR+ was greater than 9 for seven signs. Biallelic SLC19A3 mutations represented the main differential diagnosis. Non-significant differences between the two groups were found for cortical/subcortical atrophy, leucoencephalopathy and involvement of caudate nuclei, spinothalamic tract and corpus callosum. CONCLUSION: Based on these results and owing to invasiveness of skeletal muscle biopsies and cost of high-throughput DNA sequencing, we suggest giving consideration to brain MRI imaging as a diagnostic marker and an informative investigation to be performed in patients showing signs of RC enzyme deficiency.


Asunto(s)
Atrofia/diagnóstico , Encéfalo/diagnóstico por imagen , Diagnóstico Diferencial , Enfermedades Mitocondriales/diagnóstico , Adolescente , Adulto , Atrofia/diagnóstico por imagen , Atrofia/fisiopatología , Encéfalo/patología , Niño , Preescolar , Femenino , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Enfermedades Mitocondriales/diagnóstico por imagen , Enfermedades Mitocondriales/patología , Valor Predictivo de las Pruebas , Adulto Joven
13.
Hum Mutat ; 39(12): 2047-2059, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30252186

RESUMEN

Aminoacyl-tRNA synthetases are ubiquitous enzymes, which universally charge tRNAs with their cognate amino acids for use in cytosolic or organellar translation. In humans, mutations in mitochondrial tRNA synthetases have been linked to different tissue-specific pathologies. Mutations in the KARS gene, which encodes both the cytosolic and mitochondrial isoform of lysyl-tRNA synthetase, cause predominantly neurological diseases that often involve deafness, but have also been linked to cardiomyopathy, developmental delay, and lactic acidosis. Using whole exome sequencing, we identified two compound heterozygous mutations, NM_001130089.1:c.683C>T p.(Pro228Leu) and NM_001130089.1:c.1438del p.(Leu480TrpfsX3), in a patient presenting with sensorineural deafness, developmental delay, hypotonia, and lactic acidosis. Nonsense-mediated mRNA decay eliminated the truncated mRNA transcript, rendering the patient hemizygous for the missense mutation. The c.683C>T mutation was previously described, but its pathogenicity remained unexamined. Molecular characterization of patient fibroblasts revealed a multiple oxidative phosphorylation deficiency due to impaired mitochondrial translation, but no evidence of inhibition of cytosolic translation. Reintroduction of wild-type mitochondrial KARS, but not the cytosolic isoform, rescued this phenotype confirming the disease-causing nature of p.(Pro228Leu) exchange and demonstrating the mitochondrial etiology of the disease. We propose that mitochondrial translation deficiency is the probable disease culprit in this and possibly other patients with mutations in KARS.


Asunto(s)
Acidosis Láctica/genética , Discapacidades del Desarrollo/genética , Fibroblastos/metabolismo , Pérdida Auditiva Sensorineural/genética , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Acidosis Láctica/metabolismo , Discapacidades del Desarrollo/metabolismo , Femenino , Fibroblastos/citología , Células HEK293 , Pérdida Auditiva Sensorineural/metabolismo , Humanos , Lactante , Mitocondrias/metabolismo , Fosforilación Oxidativa , Linaje , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Estabilidad del ARN , Secuenciación del Exoma/métodos
14.
Hum Mol Genet ; 25(R2): R115-R122, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27329762

RESUMEN

Mitochondrial diseases are heterogeneous and incurable conditions typically resulting from deficient ATP production in the cells. Mice, owing to their genetic and physiological similarity to humans as well as their relatively easy maintenance and propagation, are extremely valuable for studying mitochondrial diseases and are also indispensable for the preclinical evaluation of novel therapies for these devastating conditions. Here, we review the recent exciting developments in the field focusing on mouse models for mitochondrial disease genes although models for genes not involved in the pathogenesis of mitochondrial disease and therapeutic proof-of-concept studies using mouse models are also discussed.

15.
PLoS Genet ; 10(2): e1004110, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24516400

RESUMEN

Biogenesis of mammalian mitochondrial ribosomes requires a concerted maturation of both the small (SSU) and large subunit (LSU). We demonstrate here that the m(5)C methyltransferase NSUN4, which forms a complex with MTERF4, is essential in mitochondrial ribosomal biogenesis as mitochondrial translation is abolished in conditional Nsun4 mouse knockouts. Deep sequencing of bisulfite-treated RNA shows that NSUN4 methylates cytosine 911 in 12S rRNA (m5C911) of the SSU. Surprisingly, NSUN4 does not need MTERF4 to generate this modification. Instead, the NSUN4/MTERF4 complex is required to assemble the SSU and LSU to form a monosome. NSUN4 is thus a dual function protein, which on the one hand is needed for 12S rRNA methylation and, on the other hand interacts with MTERF4 to facilitate monosome assembly. The presented data suggest that NSUN4 has a key role in controlling a final step in ribosome biogenesis to ensure that only the mature SSU and LSU are assembled.


Asunto(s)
Proteínas Portadoras/genética , Metiltransferasas/genética , Mitocondrias/genética , ARN Ribosómico/genética , Ribosomas/genética , Animales , Proteínas Portadoras/metabolismo , Metilación de ADN/genética , Metiltransferasas/metabolismo , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Unión Proteica , ARN Ribosómico/biosíntesis , Ribosomas/ultraestructura , Factores de Transcripción/metabolismo
16.
Hum Mol Genet ; 23(10): 2580-92, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24399447

RESUMEN

Defects of the oxidative phosphorylation system, in particular of cytochrome-c oxidase (COX, respiratory chain complex IV), are common causes of Leigh syndrome (LS), which is a rare neurodegenerative disorder with severe progressive neurological symptoms that usually present during infancy or early childhood. The COX-deficient form of LS is commonly caused by mutations in genes encoding COX assembly factors, e.g. SURF1, SCO1, SCO2 or COX10. However, other mutations affecting genes that encode proteins not directly involved in COX assembly can also cause LS. The leucine-rich pentatricopeptide repeat containing protein (LRPPRC) regulates mRNA stability, polyadenylation and coordinates mitochondrial translation. In humans, mutations in Lrpprc cause the French Canadian type of LS. Despite the finding that LRPPRC deficiency affects the stability of most mitochondrial mRNAs, its pathophysiological effect has mainly been attributed to COX deficiency. Surprisingly, we show here that the impaired mitochondrial respiration and reduced ATP production observed in Lrpprc conditional knockout mouse hearts is caused by an ATP synthase deficiency. Furthermore, the appearance of inactive subassembled ATP synthase complexes causes hyperpolarization and increases mitochondrial reactive oxygen species production. Our findings shed important new light on the bioenergetic consequences of the loss of LRPPRC in cardiac mitochondria.


Asunto(s)
Mitocondrias Cardíacas/enzimología , ATPasas de Translocación de Protón Mitocondriales/deficiencia , Proteínas de Neoplasias/genética , Adenosina Trifosfato/biosíntesis , Animales , Complejo IV de Transporte de Electrones/metabolismo , Metabolismo Energético , Humanos , Enfermedad de Leigh/genética , Potencial de la Membrana Mitocondrial , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Cardíacas/patología , ATPasas de Translocación de Protón Mitocondriales/genética , Fosforilación Oxidativa , Consumo de Oxígeno , Multimerización de Proteína , Especies Reactivas de Oxígeno/metabolismo
17.
EMBO J ; 31(2): 443-56, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22045337

RESUMEN

Regulation of mtDNA expression is critical for maintaining cellular energy homeostasis and may, in principle, occur at many different levels. The leucine-rich pentatricopeptide repeat containing (LRPPRC) protein regulates mitochondrial mRNA stability and an amino-acid substitution of this protein causes the French-Canadian type of Leigh syndrome (LSFC), a neurodegenerative disorder characterized by complex IV deficiency. We have generated conditional Lrpprc knockout mice and show here that the gene is essential for embryonic development. Tissue-specific disruption of Lrpprc in heart causes mitochondrial cardiomyopathy with drastic reduction in steady-state levels of most mitochondrial mRNAs. LRPPRC forms an RNA-dependent protein complex that is necessary for maintaining a pool of non-translated mRNAs in mammalian mitochondria. Loss of LRPPRC does not only decrease mRNA stability, but also leads to loss of mRNA polyadenylation and the appearance of aberrant mitochondrial translation. The translation pattern without the presence of LRPPRC is misregulated with excessive translation of some transcripts and no translation of others. Our findings point to the existence of an elaborate machinery that regulates mammalian mtDNA expression at the post-transcriptional level.


Asunto(s)
Deficiencia de Citocromo-c Oxidasa/genética , Enfermedad de Leigh/genética , Mitocondrias Cardíacas/fisiología , Proteínas de Neoplasias/fisiología , Poliadenilación/fisiología , Biosíntesis de Proteínas/fisiología , Animales , ADN Mitocondrial/genética , Complejo IV de Transporte de Electrones/análisis , Células HeLa , Humanos , Sustancias Macromoleculares , Ratones , Ratones Noqueados , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Especificidad de Órganos , Polinucleotido Adenililtransferasa , Estabilidad del ARN , ARN Mensajero , Proteínas de Unión al ARN/metabolismo
19.
PLoS Genet ; 9(1): e1003178, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23300484

RESUMEN

Regulation of mitochondrial DNA (mtDNA) expression is critical for the control of oxidative phosphorylation in response to physiological demand, and this regulation is often impaired in disease and aging. We have previously shown that mitochondrial transcription termination factor 3 (MTERF3) is a key regulator that represses mtDNA transcription in the mouse, but its molecular mode of action has remained elusive. Based on the hypothesis that key regulatory mechanisms for mtDNA expression are conserved in metazoans, we analyzed Mterf3 knockout and knockdown flies. We demonstrate here that decreased expression of MTERF3 not only leads to activation of mtDNA transcription, but also impairs assembly of the large mitochondrial ribosomal subunit. This novel function of MTERF3 in mitochondrial ribosomal biogenesis is conserved in the mouse, thus we identify a novel and unexpected role for MTERF3 in coordinating the crosstalk between transcription and translation for the regulation of mammalian mtDNA gene expression.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster/genética , Mitocondrias , Proteínas Mitocondriales , Ribosomas , Animales , ADN Mitocondrial/genética , Proteínas de Drosophila/genética , Regulación de la Expresión Génica , Invertebrados/genética , Invertebrados/metabolismo , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Fosforilación Oxidativa , Ribosomas/genética , Ribosomas/metabolismo , Transcripción Genética
20.
Hum Mol Genet ; 22(10): 1983-93, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23393161

RESUMEN

Replication of the mammalian mitochondrial DNA (mtDNA) is dependent on the minimal replisome, consisting of the heterotrimeric mtDNA polymerase (POLG), the hexameric DNA helicase TWINKLE and the tetrameric single-stranded DNA-binding protein (mtSSB). TWINKLE has been shown to unwind DNA during the replication process and many disease-causing mutations have been mapped to its gene. Patients carrying Twinkle mutations develop multiple deletions of mtDNA, deficient respiratory chain function and neuromuscular symptoms. Despite its importance in human disease, it has been unclear whether TWINKLE is the only replicative DNA helicase in mammalian mitochondria. Furthermore, a substantial portion of mtDNA replication events is prematurely terminated at the end of mitochondrial control region (D-loop) and it is unknown whether TWINKLE also has a role in this abortive replication. Here, we present a conditional mouse knockout for Twinkle and demonstrate that TWINKLE is essential for mouse embryonic development and thus is the only replicative DNA helicase in mammalian mitochondria. Conditional knockout of Twinkle results in severe and rapid mtDNA depletion in heart and skeletal muscle. No replication intermediates or deleted mtDNA molecules are observed after Twinkle knockout, suggesting that TWINKLE once loaded is very processive. We also demonstrate that TWINKLE is essential for nascent H-strand synthesis in the D-loop, thus showing that there is no separate DNA helicase responsible for replication of this region. Our data thus suggest that the relative levels of abortive D-loop synthesis versus complete mtDNA replication are regulated and may provide a mechanism to control progression to complete mtDNA replication.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN/fisiología , ADN Mitocondrial/biosíntesis , Proteínas Mitocondriales/metabolismo , Animales , ADN Helicasas/genética , ADN Mitocondrial/genética , Enfermedades Genéticas Congénitas/enzimología , Enfermedades Genéticas Congénitas/genética , Humanos , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Mutación , Enfermedades Neuromusculares/enzimología , Enfermedades Neuromusculares/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA