Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 84(2): 345-358.e5, 2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38199007

RESUMO

Cellular proteostasis requires transport of polypeptides across membranes. Although defective transport processes trigger cytosolic rescue and quality control mechanisms that clear translocases and membranes from unproductive cargo, proteins that are synthesized within mitochondria are not accessible to these mechanisms. Mitochondrial-encoded proteins are inserted cotranslationally into the inner membrane by the conserved insertase OXA1L. Here, we identify TMEM126A as a OXA1L-interacting protein. TMEM126A associates with mitochondrial ribosomes and translation products. Loss of TMEM126A leads to the destabilization of mitochondrial translation products, triggering an inner membrane quality control process, in which newly synthesized proteins are degraded by the mitochondrial iAAA protease. Our data reveal that TMEM126A cooperates with OXA1L in protein insertion into the membrane. Upon loss of TMEM126A, the cargo-blocked OXA1L insertase complexes undergo proteolytic clearance by the iAAA protease machinery together with its cargo.


Assuntos
Mitocôndrias , Membranas Mitocondriais , Mitocôndrias/genética , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Biossíntese de Proteínas , Ribossomos/metabolismo , Peptídeo Hidrolases/metabolismo
2.
Hum Mol Genet ; 33(R1): R47-R52, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38779773

RESUMO

The mitochondrial oxidative phosphorylation (OXPHOS) system produces the majority of energy required by cells. Given the mitochondrion's endosymbiotic origin, the OXPHOS machinery is still under dual genetic control where most OXPHOS subunits are encoded by the nuclear DNA and imported into mitochondria, while a small subset is encoded on the mitochondrion's own genome, the mitochondrial DNA (mtDNA). The nuclear and mtDNA encoded subunits must be expressed and assembled in a highly orchestrated fashion to form a functional OXPHOS system and meanwhile prevent the generation of any harmful assembly intermediates. While several mechanisms have evolved in eukaryotes to achieve such a coordinated expression, this review will focus on how the translation of mtDNA encoded OXPHOS subunits is tailored to OXPHOS assembly.


Assuntos
DNA Mitocondrial , Mitocôndrias , Fosforilação Oxidativa , Biossíntese de Proteínas , Mitocôndrias/metabolismo , Mitocôndrias/genética , Humanos , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Animais
3.
PLoS Genet ; 19(1): e1010573, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36608143

RESUMO

Mammalian mitochondrial DNA (mtDNA) is inherited uniparentally through the female germline without undergoing recombination. This poses a major problem as deleterious mtDNA mutations must be eliminated to avoid a mutational meltdown over generations. At least two mechanisms that can decrease the mutation load during maternal transmission are operational: a stochastic bottleneck for mtDNA transmission from mother to child, and a directed purifying selection against transmission of deleterious mtDNA mutations. However, the molecular mechanisms controlling these processes remain unknown. In this study, we systematically tested whether decreased autophagy contributes to purifying selection by crossing the C5024T mouse model harbouring a single pathogenic heteroplasmic mutation in the tRNAAla gene of the mtDNA with different autophagy-deficient mouse models, including knockouts of Parkin, Bcl2l13, Ulk1, and Ulk2. Our study reveals a statistically robust effect of knockout of Bcl2l13 on the selection process, and weaker evidence for the effect of Ulk1 and potentially Ulk2, while no statistically significant impact is seen for knockout of Parkin. This points at distinctive roles of these players in germline purifying selection. Overall, our approach provides a framework for investigating the roles of other important factors involved in the enigmatic process of purifying selection and guides further investigations for the role of BCL2L13 in the elimination of non-synonymous mutations in protein-coding genes.


Assuntos
DNA Mitocondrial , Transmissão Vertical de Doenças Infecciosas , Animais , Camundongos , Feminino , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Mitocôndrias/genética , Células Germinativas/metabolismo , Mutação , Autofagia/genética , Mamíferos/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
4.
Mol Cell ; 63(4): 621-632, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27499296

RESUMO

Mitochondria are essential for numerous cellular processes, yet hundreds of their proteins lack robust functional annotation. To reveal functions for these proteins (termed MXPs), we assessed condition-specific protein-protein interactions for 50 select MXPs using affinity enrichment mass spectrometry. Our data connect MXPs to diverse mitochondrial processes, including multiple aspects of respiratory chain function. Building upon these observations, we validated C17orf89 as a complex I (CI) assembly factor. Disruption of C17orf89 markedly reduced CI activity, and its depletion is found in an unresolved case of CI deficiency. We likewise discovered that LYRM5 interacts with and deflavinates the electron-transferring flavoprotein that shuttles electrons to coenzyme Q (CoQ). Finally, we identified a dynamic human CoQ biosynthetic complex involving multiple MXPs whose topology we map using purified components. Collectively, our data lend mechanistic insight into respiratory chain-related activities and prioritize hundreds of additional interactions for further exploration of mitochondrial protein function.


Assuntos
Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Mapeamento de Interação de Proteínas/métodos , Mapas de Interação de Proteínas , Proteômica/métodos , Bases de Dados de Proteínas , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Proteínas Mitocondriais/genética , Interferência de RNA , Transdução de Sinais , Transfecção , Ubiquinona/metabolismo
5.
Am J Hum Genet ; 103(4): 592-601, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30245030

RESUMO

Isolated complex I deficiency is a common biochemical phenotype observed in pediatric mitochondrial disease and often arises as a consequence of pathogenic variants affecting one of the ∼65 genes encoding the complex I structural subunits or assembly factors. Such genetic heterogeneity means that application of next-generation sequencing technologies to undiagnosed cohorts has been a catalyst for genetic diagnosis and gene-disease associations. We describe the clinical and molecular genetic investigations of four unrelated children who presented with neuroradiological findings and/or elevated lactate levels, highly suggestive of an underlying mitochondrial diagnosis. Next-generation sequencing identified bi-allelic variants in NDUFA6, encoding a 15 kDa LYR-motif-containing complex I subunit that forms part of the Q-module. Functional investigations using subjects' fibroblast cell lines demonstrated complex I assembly defects, which were characterized in detail by mass-spectrometry-based complexome profiling. This confirmed a marked reduction in incorporated NDUFA6 and a concomitant reduction in other Q-module subunits, including NDUFAB1, NDUFA7, and NDUFA12. Lentiviral transduction of subjects' fibroblasts showed normalization of complex I. These data also support supercomplex formation, whereby the ∼830 kDa complex I intermediate (consisting of the P- and Q-modules) is in complex with assembled complex III and IV holoenzymes despite lacking the N-module. Interestingly, RNA-sequencing data provided evidence that the consensus RefSeq accession number does not correspond to the predominant transcript in clinically relevant tissues, prompting revision of the NDUFA6 RefSeq transcript and highlighting not only the importance of thorough variant interpretation but also the assessment of appropriate transcripts for analysis.


Assuntos
Complexo I de Transporte de Elétrons/deficiência , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Mutação/genética , Alelos , Sequência de Aminoácidos , Complexo I de Transporte de Elétrons/genética , Feminino , Fibroblastos/patologia , Heterogeneidade Genética , Humanos , Lactente , Masculino , Mitocôndrias/genética , Fenótipo , Alinhamento de Sequência
6.
Am J Hum Genet ; 101(4): 525-538, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28942965

RESUMO

Complement component 1 Q subcomponent-binding protein (C1QBP; also known as p32) is a multi-compartmental protein whose precise function remains unknown. It is an evolutionary conserved multifunctional protein localized primarily in the mitochondrial matrix and has roles in inflammation and infection processes, mitochondrial ribosome biogenesis, and regulation of apoptosis and nuclear transcription. It has an N-terminal mitochondrial targeting peptide that is proteolytically processed after import into the mitochondrial matrix, where it forms a homotrimeric complex organized in a doughnut-shaped structure. Although C1QBP has been reported to exert pleiotropic effects on many cellular processes, we report here four individuals from unrelated families where biallelic mutations in C1QBP cause a defect in mitochondrial energy metabolism. Infants presented with cardiomyopathy accompanied by multisystemic involvement (liver, kidney, and brain), and children and adults presented with myopathy and progressive external ophthalmoplegia. Multiple mitochondrial respiratory-chain defects, associated with the accumulation of multiple deletions of mitochondrial DNA in the later-onset myopathic cases, were identified in all affected individuals. Steady-state C1QBP levels were decreased in all individuals' samples, leading to combined respiratory-chain enzyme deficiency of complexes I, III, and IV. C1qbp-/- mouse embryonic fibroblasts (MEFs) resembled the human disease phenotype by showing multiple defects in oxidative phosphorylation (OXPHOS). Complementation with wild-type, but not mutagenized, C1qbp restored OXPHOS protein levels and mitochondrial enzyme activities in C1qbp-/- MEFs. C1QBP deficiency represents an important mitochondrial disorder associated with a clinical spectrum ranging from infantile lactic acidosis to childhood (cardio)myopathy and late-onset progressive external ophthalmoplegia.


Assuntos
Cardiomiopatias/genética , Proteínas de Transporte/genética , Transporte de Elétrons/fisiologia , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Mutação , Adulto , Idade de Início , Idoso , Alelos , Sequência de Aminoácidos , Animais , Cardiomiopatias/complicações , Cardiomiopatias/patologia , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Células Cultivadas , Pré-Escolar , Estudos de Coortes , DNA Mitocondrial , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Recém-Nascido , Masculino , Camundongos , Pessoa de Meia-Idade , Doenças Mitocondriais/complicações , Doenças Mitocondriais/patologia , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Fosforilação Oxidativa , Linhagem , Conformação Proteica , Homologia de Sequência , Índice de Gravidade de Doença , Adulto Jovem
7.
J Inherit Metab Dis ; 43(1): 25-35, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31119744

RESUMO

Given the rapidly decreasing cost and increasing speed and accessibility of massively parallel technologies, the integration of comprehensive genomic, transcriptomic, and proteomic data into a "multi-omics" diagnostic pipeline is within reach. Even though genomic analysis has the capability to reveal all possible perturbations in our genetic code, analysis typically reaches a diagnosis in just 35% of cases, with a diagnostic gap arising due to limitations in prioritization and interpretation of detected variants. Here we review the utility of complementing genetic data with transcriptomic data and give a perspective for the introduction of proteomics into the diagnostic pipeline. Together these methodologies enable comprehensive capture of the functional consequence of variants, unobtainable by the analysis of each methodology in isolation. This facilitates functional annotation and reprioritization of candidate genes and variants-a promising approach to shed light on the underlying molecular cause of a patient's disease, increasing diagnostic rate, and allowing actionability in clinical practice.


Assuntos
Erros Inatos do Metabolismo/diagnóstico , Biologia de Sistemas/métodos , Epigenômica/métodos , Perfilação da Expressão Gênica/métodos , Genômica/métodos , Humanos , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Metabolômica/métodos , Proteômica/métodos , Biologia de Sistemas/tendências , Transcriptoma/genética
8.
Brain ; 142(1): 50-58, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30576410

RESUMO

Physical stress, including high temperatures, may damage the central metabolic nicotinamide nucleotide cofactors [NAD(P)H], generating toxic derivatives [NAD(P)HX]. The highly conserved enzyme NAD(P)HX dehydratase (NAXD) is essential for intracellular repair of NAD(P)HX. Here we present a series of infants and children who suffered episodes of febrile illness-induced neurodegeneration or cardiac failure and early death. Whole-exome or whole-genome sequencing identified recessive NAXD variants in each case. Variants were predicted to be potentially deleterious through in silico analysis. Reverse-transcription PCR confirmed altered splicing in one case. Subject fibroblasts showed highly elevated concentrations of the damaged cofactors S-NADHX, R-NADHX and cyclic NADHX. NADHX accumulation was abrogated by lentiviral transduction of subject cells with wild-type NAXD. Subject fibroblasts and muscle biopsies showed impaired mitochondrial function, higher sensitivity to metabolic stress in media containing galactose and azide, but not glucose, and decreased mitochondrial reactive oxygen species production. Recombinant NAXD protein harbouring two missense variants leading to the amino acid changes p.(Gly63Ser) and p.(Arg608Cys) were thermolabile and showed a decrease in Vmax and increase in KM for the ATP-dependent NADHX dehydratase activity. This is the first study to identify pathogenic variants in NAXD and to link deficient NADHX repair with mitochondrial dysfunction. The results show that NAXD deficiency can be classified as a metabolite repair disorder in which accumulation of damaged metabolites likely triggers devastating effects in tissues such as the brain and the heart, eventually leading to early childhood death.


Assuntos
Hidroliases/deficiência , Doenças Neurodegenerativas/genética , Pré-Escolar , Simulação por Computador , Feminino , Febre/complicações , Febre/metabolismo , Fibroblastos/metabolismo , Vetores Genéticos , Humanos , Hidroliases/genética , Lactente , Cinética , Lentivirus , Masculino , Mitocôndrias/metabolismo , Mutação , NAD/análogos & derivados , NAD/metabolismo , Doenças Neurodegenerativas/complicações , Doenças Neurodegenerativas/metabolismo , Cultura Primária de Células , Sequenciamento Completo do Genoma
9.
Am J Hum Genet ; 98(2): 358-62, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26805782

RESUMO

Molecular diagnosis of mitochondrial disorders is challenging because of extreme clinical and genetic heterogeneity. By exome sequencing, we identified three different bi-allelic truncating mutations in TANGO2 in three unrelated individuals with infancy-onset episodic metabolic crises characterized by encephalopathy, hypoglycemia, rhabdomyolysis, arrhythmias, and laboratory findings suggestive of a defect in mitochondrial fatty acid oxidation. Over the course of the disease, all individuals developed global brain atrophy with cognitive impairment and pyramidal signs. TANGO2 (transport and Golgi organization 2) encodes a protein with a putative function in redistribution of Golgi membranes into the endoplasmic reticulum in Drosophila and a mitochondrial localization has been confirmed in mice. Investigation of palmitate-dependent respiration in mutant fibroblasts showed evidence of a functional defect in mitochondrial ß-oxidation. Our results establish TANGO2 deficiency as a clinically recognizable cause of pediatric disease with multi-organ involvement.


Assuntos
Alelos , Arritmias Cardíacas/genética , Cardiomiopatias/genética , Mutação , Arritmias Cardíacas/diagnóstico , Cardiomiopatias/diagnóstico , Pré-Escolar , Exoma , Feminino , Humanos , Lactente , Masculino , Mitocôndrias/genética , Mitocôndrias/metabolismo , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Linhagem
10.
Am J Hum Genet ; 99(4): 894-902, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27616477

RESUMO

To safeguard the cell from the accumulation of potentially harmful metabolic intermediates, specific repair mechanisms have evolved. APOA1BP, now renamed NAXE, encodes an epimerase essential in the cellular metabolite repair for NADHX and NADPHX. The enzyme catalyzes the epimerization of NAD(P)HX, thereby avoiding the accumulation of toxic metabolites. The clinical importance of the NAD(P)HX repair system has been unknown. Exome sequencing revealed pathogenic biallelic mutations in NAXE in children from four families with (sub-) acute-onset ataxia, cerebellar edema, spinal myelopathy, and skin lesions. Lactate was elevated in cerebrospinal fluid of all affected individuals. Disease onset was during the second year of life and clinical signs as well as episodes of deterioration were triggered by febrile infections. Disease course was rapidly progressive, leading to coma, global brain atrophy, and finally to death in all affected individuals. NAXE levels were undetectable in fibroblasts from affected individuals of two families. In these fibroblasts we measured highly elevated concentrations of the toxic metabolite cyclic-NADHX, confirming a deficiency of the mitochondrial NAD(P)HX repair system. Finally, NAD or nicotinic acid (vitamin B3) supplementation might have therapeutic implications for this fatal disorder.


Assuntos
Proteínas de Transporte/genética , Doenças Metabólicas/genética , Mutação , NAD/análogos & derivados , Doenças do Sistema Nervoso/genética , Racemases e Epimerases/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Pré-Escolar , Evolução Fatal , Feminino , Fibroblastos , Humanos , Lactente , Masculino , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , NAD/metabolismo , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Neuroimagem , Anormalidades da Pele/genética , Anormalidades da Pele/patologia
11.
Am J Hum Genet ; 99(3): 674-682, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27523597

RESUMO

We have used whole-exome sequencing in ten individuals from four unrelated pedigrees to identify biallelic missense mutations in the nuclear-encoded mitochondrial inorganic pyrophosphatase (PPA2) that are associated with mitochondrial disease. These individuals show a range of severity, indicating that PPA2 mutations may cause a spectrum of mitochondrial disease phenotypes. Severe symptoms include seizures, lactic acidosis, cardiac arrhythmia, and death within days of birth. In the index family, presentation was milder and manifested as cardiac fibrosis and an exquisite sensitivity to alcohol, leading to sudden arrhythmic cardiac death in the second decade of life. Comparison of normal and mutant PPA2-containing mitochondria from fibroblasts showed that the activity of inorganic pyrophosphatase was significantly reduced in affected individuals. Recombinant PPA2 enzymes modeling hypomorphic missense mutations had decreased activity that correlated with disease severity. These findings confirm the pathogenicity of PPA2 mutations and suggest that PPA2 is a cardiomyopathy-associated protein, which has a greater physiological importance in mitochondrial function than previously recognized.


Assuntos
Morte Súbita Cardíaca/etiologia , Pirofosfatase Inorgânica/deficiência , Pirofosfatase Inorgânica/genética , Doenças Mitocondriais/genética , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Mutação de Sentido Incorreto/genética , Acidose Láctica/genética , Adolescente , Adulto , Sequência de Aminoácidos , Animais , Arritmias Cardíacas/genética , Cardiomiopatias/enzimologia , Cardiomiopatias/genética , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Criança , Pré-Escolar , Morte Súbita Cardíaca/patologia , Etanol/efeitos adversos , Exoma/genética , Feminino , Fibroblastos/citologia , Fibroblastos/patologia , Fibrose/enzimologia , Fibrose/genética , Fibrose/patologia , Humanos , Lactente , Recém-Nascido , Pirofosfatase Inorgânica/química , Pirofosfatase Inorgânica/metabolismo , Masculino , Mitocôndrias/enzimologia , Mitocôndrias/genética , Mitocôndrias/patologia , Doenças Mitocondriais/enzimologia , Doenças Mitocondriais/patologia , Doenças Mitocondriais/fisiopatologia , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Modelos Moleculares , Linhagem , Fenótipo , Convulsões , Adulto Jovem
12.
Am J Hum Genet ; 99(3): 735-743, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27545679

RESUMO

SQSTM1 (sequestosome 1; also known as p62) encodes a multidomain scaffolding protein involved in various key cellular processes, including the removal of damaged mitochondria by its function as a selective autophagy receptor. Heterozygous variants in SQSTM1 have been associated with Paget disease of the bone and might contribute to neurodegeneration in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Using exome sequencing, we identified three different biallelic loss-of-function variants in SQSTM1 in nine affected individuals from four families with a childhood- or adolescence-onset neurodegenerative disorder characterized by gait abnormalities, ataxia, dysarthria, dystonia, vertical gaze palsy, and cognitive decline. We confirmed absence of the SQSTM1/p62 protein in affected individuals' fibroblasts and found evidence of a defect in the early response to mitochondrial depolarization and autophagosome formation. Our findings expand the SQSTM1-associated phenotypic spectrum and lend further support to the concept of disturbed selective autophagy pathways in neurodegenerative diseases.


Assuntos
Ataxia/genética , Autofagia/genética , Distonia/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/fisiopatologia , Proteína Sequestossoma-1/deficiência , Paralisia Supranuclear Progressiva/genética , Adolescente , Adulto , Idade de Início , Ataxia/complicações , Autofagossomos/metabolismo , Autofagossomos/patologia , Criança , Transtornos Cognitivos/genética , Disartria/complicações , Disartria/genética , Distonia/complicações , Feminino , Fibroblastos/metabolismo , Marcha/genética , Humanos , Masculino , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Transtornos dos Movimentos/complicações , Transtornos dos Movimentos/genética , Doenças Neurodegenerativas/complicações , Linhagem , Fenótipo , RNA Mensageiro/análise , Proteína Sequestossoma-1/genética , Paralisia Supranuclear Progressiva/complicações , Adulto Jovem
13.
Biochemistry ; 57(5): 861-871, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29283245

RESUMO

Fluorescent protein-based pH sensors are useful tools for measuring protein trafficking through pH changes associated with endo- and exocytosis. However, commonly used pH-sensing probes are ubiquitously expressed with their protein of interest throughout the cell, hindering our ability to focus on specific trafficking pools of proteins. We developed a family of excitation ratiometric, activatable pH responsive tandem dyes, consisting of a pH sensitive Cy3 donor linked to a fluorogenic malachite green acceptor. These cell-excluded dyes are targeted and activated upon binding to a genetically expressed fluorogen-activating protein and are suitable for selective labeling of surface proteins for analysis of endocytosis and recycling in live cells using both confocal and superresolution microscopy. Quantitative profiling of the endocytosis and recycling of tagged ß2-adrenergic receptor (B2AR) at a single-vesicle level revealed differences among B2AR agonists, consistent with more detailed pharmacological profiling.


Assuntos
Carbocianinas/análise , Corantes/análise , Endocitose/fisiologia , Exocitose/fisiologia , Corantes Fluorescentes/análise , Transporte Proteico/fisiologia , Corantes de Rosanilina/análise , Anticorpos de Cadeia Única/análise , Endossomos/metabolismo , Endossomos/ultraestrutura , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Indicadores e Reagentes/análise , Microscopia Confocal , Receptores Adrenérgicos beta 2/metabolismo
14.
Am J Hum Genet ; 97(2): 319-28, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26189817

RESUMO

Deficiencies in respiratory-chain complexes lead to a variety of clinical phenotypes resulting from inadequate energy production by the mitochondrial oxidative phosphorylation system. Defective expression of mtDNA-encoded genes, caused by mutations in either the mitochondrial or nuclear genome, represents a rapidly growing group of human disorders. By whole-exome sequencing, we identified two unrelated individuals carrying compound heterozygous variants in TRMT5 (tRNA methyltransferase 5). TRMT5 encodes a mitochondrial protein with strong homology to members of the class I-like methyltransferase superfamily. Both affected individuals presented with lactic acidosis and evidence of multiple mitochondrial respiratory-chain-complex deficiencies in skeletal muscle, although the clinical presentation of the two affected subjects was remarkably different; one presented in childhood with failure to thrive and hypertrophic cardiomyopathy, and the other was an adult with a life-long history of exercise intolerance. Mutations in TRMT5 were associated with the hypomodification of a guanosine residue at position 37 (G37) of mitochondrial tRNA; this hypomodification was particularly prominent in skeletal muscle. Deficiency of the G37 modification was also detected in human cells subjected to TRMT5 RNAi. The pathogenicity of the detected variants was further confirmed in a heterologous yeast model and by the rescue of the molecular phenotype after re-expression of wild-type TRMT5 cDNA in cells derived from the affected individuals. Our study highlights the importance of post-transcriptional modification of mitochondrial tRNAs for faithful mitochondrial function.


Assuntos
Doenças Mitocondriais/genética , Modelos Moleculares , Processamento Pós-Transcricional do RNA/genética , RNA de Transferência/genética , tRNA Metiltransferases/genética , Sequência de Aminoácidos , Pareamento de Bases , Sequência de Bases , Exoma/genética , Mutação da Fase de Leitura/genética , Humanos , Doenças Mitocondriais/patologia , Dados de Sequência Molecular , Linhagem , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , tRNA Metiltransferases/química
15.
Am J Hum Genet ; 97(1): 163-9, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-26073778

RESUMO

Acute liver failure (ALF) in infancy and childhood is a life-threatening emergency. Few conditions are known to cause recurrent acute liver failure (RALF), and in about 50% of cases, the underlying molecular cause remains unresolved. Exome sequencing in five unrelated individuals with fever-dependent RALF revealed biallelic mutations in NBAS. Subsequent Sanger sequencing of NBAS in 15 additional unrelated individuals with RALF or ALF identified compound heterozygous mutations in an additional six individuals from five families. Immunoblot analysis of mutant fibroblasts showed reduced protein levels of NBAS and its proposed interaction partner p31, both involved in retrograde transport between endoplasmic reticulum and Golgi. We recommend NBAS analysis in individuals with acute infantile liver failure, especially if triggered by fever.


Assuntos
Falência Hepática Aguda/genética , Proteínas de Neoplasias/genética , Sequência de Bases , Transporte Biológico/genética , Exoma/genética , Fibroblastos/metabolismo , Frequência do Gene , Alemanha , Humanos , Immunoblotting , Lactente , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Linhagem , Recidiva , Análise de Sequência de DNA
16.
Am J Hum Genet ; 96(2): 309-17, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25658047

RESUMO

Primary coenzyme Q10 (CoQ10) deficiencies are rare, clinically heterogeneous disorders caused by mutations in several genes encoding proteins involved in CoQ10 biosynthesis. CoQ10 is an essential component of the electron transport chain (ETC), where it shuttles electrons from complex I or II to complex III. By whole-exome sequencing, we identified five individuals carrying biallelic mutations in COQ4. The precise function of human COQ4 is not known, but it seems to play a structural role in stabilizing a multiheteromeric complex that contains most of the CoQ10 biosynthetic enzymes. The clinical phenotypes of the five subjects varied widely, but four had a prenatal or perinatal onset with early fatal outcome. Two unrelated individuals presented with severe hypotonia, bradycardia, respiratory insufficiency, and heart failure; two sisters showed antenatal cerebellar hypoplasia, neonatal respiratory-distress syndrome, and epileptic encephalopathy. The fifth subject had an early-onset but slowly progressive clinical course dominated by neurological deterioration with hardly any involvement of other organs. All available specimens from affected subjects showed reduced amounts of CoQ10 and often displayed a decrease in CoQ10-dependent ETC complex activities. The pathogenic role of all identified mutations was experimentally validated in a recombinant yeast model; oxidative growth, strongly impaired in strains lacking COQ4, was corrected by expression of human wild-type COQ4 cDNA but failed to be corrected by expression of COQ4 cDNAs with any of the mutations identified in affected subjects. COQ4 mutations are responsible for early-onset mitochondrial diseases with heterogeneous clinical presentations and associated with CoQ10 deficiency.


Assuntos
Ataxia/genética , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Debilidade Muscular/genética , Fenótipo , Ubiquinona/deficiência , Sequência de Aminoácidos , Ataxia/patologia , Sequência de Bases , Exoma/genética , Evolução Fatal , Feminino , Componentes do Gene , Humanos , Masculino , Doenças Mitocondriais/patologia , Dados de Sequência Molecular , Debilidade Muscular/patologia , Mutação/genética , Linhagem , Saccharomyces cerevisiae , Análise de Sequência de DNA , Ubiquinona/genética
17.
J Inherit Metab Dis ; 41(3): 525-532, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29372369

RESUMO

Exome wide sequencing techniques have revolutionized molecular diagnostics in patients with suspected inborn errors of metabolism or neuromuscular disorders. However, the diagnostic yield of 25-60% still leaves a large fraction of individuals without a diagnosis. This indicates a causative role for non-exonic regulatory variants not covered by whole exome sequencing. Here we review how systematic RNA-sequencing analysis (RNA-seq, "transcriptomics") lead to a molecular diagnosis in 10-35% of patients in whom whole exome sequencing failed to do so. Importantly, RNA-sequencing based discoveries cannot only guide molecular diagnosis but might also unravel therapeutic intervention points such as antisense oligonucleotide treatment for splicing defects as recently reported for spinal muscular atrophy.


Assuntos
Genômica/métodos , Erros Inatos do Metabolismo/diagnóstico , Técnicas de Diagnóstico Molecular/métodos , Análise de Sequência de RNA/métodos , Transcriptoma , Testes Genéticos , Humanos , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Sequenciamento do Exoma
18.
Hum Mol Genet ; 24(8): 2247-66, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25552653

RESUMO

Mitochondrial diseases often exhibit tissue-specific pathologies, but this phenomenon is poorly understood. Here we present regulation of mitochondrial translation by the Mitochondrial Translation Optimization Factor 1, MTO1, as a novel player in this scenario. We demonstrate that MTO1 mediates tRNA modification and controls mitochondrial translation rate in a highly tissue-specific manner associated with tissue-specific OXPHOS defects. Activation of mitochondrial proteases, aberrant translation products, as well as defects in OXPHOS complex assembly observed in MTO1 deficient mice further imply that MTO1 impacts translation fidelity. In our mouse model, MTO1-related OXPHOS deficiency can be bypassed by feeding a ketogenic diet. This therapeutic intervention is independent of the MTO1-mediated tRNA modification and involves balancing of mitochondrial and cellular secondary stress responses. Our results thereby establish mammalian MTO1 as a novel factor in the tissue-specific regulation of OXPHOS and fine tuning of mitochondrial translation accuracy.


Assuntos
Proteínas de Transporte/metabolismo , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/metabolismo , Fosforilação Oxidativa , Biossíntese de Proteínas , RNA de Transferência/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Dieta Cetogênica , Fibroblastos/metabolismo , Humanos , Camundongos , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Proteínas Mitocondriais , Dados de Sequência Molecular , Especificidade de Órgãos , RNA de Transferência/genética , Proteínas de Ligação a RNA , Alinhamento de Sequência
19.
Brain ; 139(Pt 2): 346-54, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26626369

RESUMO

Thioredoxin 2 (TXN2; also known as Trx2) is a small mitochondrial redox protein essential for the control of mitochondrial reactive oxygen species homeostasis, apoptosis regulation and cell viability. Exome sequencing in a 16-year-old adolescent suffering from an infantile-onset neurodegenerative disorder with severe cerebellar atrophy, epilepsy, dystonia, optic atrophy, and peripheral neuropathy, uncovered a homozygous stop mutation in TXN2. Analysis of patient-derived fibroblasts demonstrated absence of TXN2 protein, increased reactive oxygen species levels, impaired oxidative stress defence and oxidative phosphorylation dysfunction. Reconstitution of TXN2 expression restored all these parameters, indicating the causal role of TXN2 mutation in disease development. Supplementation with antioxidants effectively suppressed cellular reactive oxygen species production, improved cell viability and mitigated clinical symptoms during short-term follow-up. In conclusion, our report on a patient with TXN2 deficiency suggests an important role of reactive oxygen species homeostasis for human neuronal maintenance and energy metabolism.


Assuntos
Homeostase/fisiologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/deficiência , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/metabolismo , Criança , Humanos , Masculino , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Doenças Neurodegenerativas/genética , Oxirredução , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Tiorredoxinas/genética
20.
J Hepatol ; 65(2): 377-85, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27151179

RESUMO

BACKGROUND & AIMS: Next generation sequencing approaches have tremendously improved the diagnosis of rare genetic diseases. It may however be faced with difficult clinical interpretation of variants. Inherited enzymatic diseases provide an invaluable possibility to evaluate the function of the defective enzyme in human cell biology. This is the case for respiratory complex III, which has 11 structural subunits and requires several assembly factors. An important role of complex III in liver function is suggested by its frequent impairment in human cases of genetic complex III defects. METHODS: We report the case of a child with complex III defect and acute liver dysfunction with lactic acidosis, hypoglycemia, and hyperammonemia. Mitochondrial activities were assessed in liver and fibroblasts using spectrophotometric assays. Genetic analysis was done by exome followed by Sanger sequencing. Functional complementation of defective fibroblasts was performed using lentiviral transduction followed by enzymatic analyses and expression assays. RESULTS: Homozygous, truncating, mutations in LYRM7 and MTO1, two genes encoding essential mitochondrial proteins were found. Functional complementation of the complex III defect in fibroblasts demonstrated the causal role of LYRM7 mutations. Comparison of the patient's clinical history to previously reported patients with complex III defect due to nuclear DNA mutations, some actually followed by us, showed striking similarities allowing us to propose common pathophysiology. CONCLUSIONS: Profound complex III defect in liver does not induce actual liver failure but impedes liver adaptation to prolonged fasting leading to severe lactic acidosis, hypoglycemia, and hyperammonemia, potentially leading to irreversible brain damage. LAY SUMMARY: The diagnosis of rare genetic disease has been tremendously accelerated by the development of high throughput sequencing technology. In this paper we report the investigations that have led to identify LYRM7 mutations causing severe hepatic defect of respiratory complex III. Based on the comparison of the patient's phenotype with other cases of complex III defect, we propose that profound complex III defect in liver does not induce actual liver failure but impedes liver adaptation to prolonged fasting.


Assuntos
Jejum , Adaptação Fisiológica , Sequenciamento de Nucleotídeos em Larga Escala , Homozigoto , Humanos , Fígado , Proteínas Mitocondriais , Respiração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA