Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Hum Genet ; 111(4): 714-728, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38579669

RESUMO

Argininosuccinate lyase deficiency (ASLD) is a recessive metabolic disorder caused by variants in ASL. In an essential step in urea synthesis, ASL breaks down argininosuccinate (ASA), a pathognomonic ASLD biomarker. The severe disease forms lead to hyperammonemia, neurological injury, and even early death. The current treatments are unsatisfactory, involving a strict low-protein diet, arginine supplementation, nitrogen scavenging, and in some cases, liver transplantation. An unmet need exists for improved, efficient therapies. Here, we show the potential of a lipid nanoparticle-mediated CRISPR approach using adenine base editors (ABEs) for ASLD treatment. To model ASLD, we first generated human-induced pluripotent stem cells (hiPSCs) from biopsies of individuals homozygous for the Finnish founder variant (c.1153C>T [p.Arg385Cys]) and edited this variant using the ABE. We then differentiated the hiPSCs into hepatocyte-like cells that showed a 1,000-fold decrease in ASA levels compared to those of isogenic non-edited cells. Lastly, we tested three different FDA-approved lipid nanoparticle formulations to deliver the ABE-encoding RNA and the sgRNA targeting the ASL variant. This approach efficiently edited the ASL variant in fibroblasts with no apparent cell toxicity and minimal off-target effects. Further, the treatment resulted in a significant decrease in ASA, to levels of healthy donors, indicating restoration of the urea cycle. Our work describes a highly efficient approach to editing the disease-causing ASL variant and restoring the function of the urea cycle. This method relies on RNA delivered by lipid nanoparticles, which is compatible with clinical applications, improves its safety profile, and allows for scalable production.


Assuntos
Argininossuccinato Liase , Acidúria Argininossuccínica , Humanos , Argininossuccinato Liase/genética , Acidúria Argininossuccínica/genética , Acidúria Argininossuccínica/terapia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , RNA Guia de Sistemas CRISPR-Cas , Ureia , Edição de Genes/métodos
2.
Hum Mol Genet ; 28(2): 258-268, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30285085

RESUMO

Recessively inherited variants in AARS2 (NM_020745.2) encoding mitochondrial alanyl-tRNA synthetase (mt-AlaRS) were first described in patients presenting with fatal infantile cardiomyopathy and multiple oxidative phosphorylation defects. To date, all described patients with AARS2-related fatal infantile cardiomyopathy are united by either a homozygous or compound heterozygous c.1774C>T (p.Arg592Trp) missense founder mutation that is absent in patients with other AARS2-related phenotypes. We describe the clinical, biochemical and molecular investigations of two unrelated boys presenting with fatal infantile cardiomyopathy, lactic acidosis and respiratory failure. Oxidative histochemistry showed cytochrome c oxidase-deficient fibres in skeletal and cardiac muscle. Biochemical studies showed markedly decreased activities of mitochondrial respiratory chain complexes I and IV with a mild decrease of complex III activity in skeletal and cardiac muscle. Using next-generation sequencing, we identified a c.1738C>T (p.Arg580Trp) AARS2 variant shared by both patients that was in trans with a loss-of-function heterozygous AARS2 variant; a c.1008dupT (p.Asp337*) nonsense variant or an intragenic deletion encompassing AARS2 exons 5-7. Interestingly, our patients did not harbour the p.Arg592Trp AARS2 founder mutation. In silico modelling of the p.Arg580Trp substitution suggested a deleterious impact on protein stability and folding. We confirmed markedly decreased mt-AlaRS protein levels in patient fibroblasts, skeletal and cardiac muscle, although mitochondrial protein synthesis defects were confined to skeletal and cardiac muscle. In vitro data showed that the p.Arg580Trp variant had a minimal effect on activation, aminoacylation or misaminoacylation activities relative to wild-type mt-AlaRS, demonstrating that instability of mt-AlaRS is the biological mechanism underlying the fatal cardiomyopathy phenotype in our patients.


Assuntos
Alanina-tRNA Ligase/metabolismo , Cardiomiopatias/enzimologia , Alanina-tRNA Ligase/genética , Cardiomiopatias/genética , Doenças em Gêmeos/genética , Estabilidade Enzimática , Fibroblastos/metabolismo , Genes Recessivos , Humanos , Lactente , Ácido Láctico , Masculino , Mitocôndrias/metabolismo , Proteínas Mitocondriais/biossíntese , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Linhagem , Insuficiência Respiratória/enzimologia
3.
Nucleic Acids Res ; 46(2): 849-860, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29228266

RESUMO

Accuracy of protein synthesis is enabled by the selection of amino acids for tRNA charging by aminoacyl-tRNA synthetases (ARSs), and further enhanced by the proofreading functions of some of these enzymes for eliminating tRNAs mischarged with noncognate amino acids. Mouse models of editing-defective cytoplasmic alanyl-tRNA synthetase (AlaRS) have previously demonstrated the importance of proofreading for cytoplasmic protein synthesis, with embryonic lethal and progressive neurodegeneration phenotypes. Mammalian mitochondria import their own set of nuclear-encoded ARSs for translating critical polypeptides of the oxidative phosphorylation system, but the importance of editing by the mitochondrial ARSs for mitochondrial proteostasis has not been known. We demonstrate here that the human mitochondrial AlaRS is capable of editing mischarged tRNAs in vitro, and that loss of the proofreading activity causes embryonic lethality in mice. These results indicate that tRNA proofreading is essential in mammalian mitochondria, and cannot be overcome by other quality control mechanisms.


Assuntos
Alanina-tRNA Ligase/genética , Mitocôndrias/genética , Edição de RNA , RNA de Transferência/genética , Aminoacilação de RNA de Transferência/genética , Alanina-tRNA Ligase/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Mamíferos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mutação , Biossíntese de Proteínas/genética , RNA de Transferência/metabolismo , Homologia de Sequência de Aminoácidos
4.
Hum Mol Genet ; 26(17): 3352-3361, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28645153

RESUMO

Leigh syndrome is a severe infantile encephalopathy with an exceptionally variable genetic background. We studied the exome of a child manifesting with Leigh syndrome at one month of age and progressing to death by the age of 2.4 years, and identified novel compound heterozygous variants in PNPT1, encoding the polynucleotide phosphorylase (PNPase). Expression of the wild type PNPT1 in the subject's myoblasts functionally complemented the defects, and the pathogenicity was further supported by structural predictions and protein and RNA analyses. PNPase is a key enzyme in mitochondrial RNA metabolism, with suggested roles in mitochondrial RNA import and degradation. The variants were predicted to locate in the PNPase active site and disturb the RNA processing activity of the enzyme. The PNPase trimer formation was not affected, but specific RNA processing intermediates derived from mitochondrial transcripts of the ND6 subunit of Complex I, as well as small mRNA fragments, accumulated in the subject's myoblasts. Mitochondrial RNA processing mediated by the degradosome consisting of hSUV3 and PNPase is poorly characterized, and controversy on the role and location of PNPase within human mitochondria exists. Our evidence indicates that PNPase activity is essential for the correct maturation of the ND6 transcripts, and likely for the efficient removal of degradation intermediates. Loss of its activity will result in combined respiratory chain deficiency, and a classic respiratory chain-deficiency-associated disease, Leigh syndrome, indicating an essential role for the enzyme for normal function of the mitochondrial respiratory chain.


Assuntos
Exorribonucleases/genética , Exorribonucleases/metabolismo , Doença de Leigh/genética , Pré-Escolar , Exoma , Exorribonucleases/química , Feminino , Expressão Gênica , Humanos , Lactente , Recém-Nascido , Doença de Leigh/metabolismo , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Polirribonucleotídeo Nucleotidiltransferase , RNA/metabolismo , RNA Mensageiro/metabolismo , RNA Mitocondrial
5.
Hum Mol Genet ; 26(8): 1432-1443, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28158749

RESUMO

De novo mutations in ATAD3A (ATPase family AAA-domain containing protein 3A) were recently found to cause a neurological syndrome with developmental delay, hypotonia, spasticity, optic atrophy, axonal neuropathy, and hypertrophic cardiomyopathy. Using whole-exome sequencing, we identified a dominantly inherited heterozygous variant c.1064G > A (p.G355D) in ATAD3A in a mother presenting with hereditary spastic paraplegia (HSP) and axonal neuropathy and her son with dyskinetic cerebral palsy, both with disease onset in childhood. HSP is a clinically and genetically heterogeneous disorder of the upper motor neurons. Symptoms beginning in early childhood may resemble spastic cerebral palsy. The function of ATAD3A, a mitochondrial inner membrane AAA ATPase, is yet undefined. AAA ATPases form hexameric rings, which are catalytically dependent on the co-operation of the subunits. The dominant-negative patient mutation affects the Walker A motif, which is responsible for ATP binding in the AAA module of ATAD3A, and we show that the recombinant mutant ATAD3A protein has a markedly reduced ATPase activity. We further show that overexpression of the mutant ATAD3A fragments the mitochondrial network and induces lysosome mass. Similarly, we observed altered dynamics of the mitochondrial network and increased lysosomes in patient fibroblasts and neurons derived through differentiation of patient-specific induced pluripotent stem cells. These alterations were verified in patient fibroblasts to associate with upregulated basal autophagy through mTOR inactivation, resembling starvation. Mutations in ATAD3A can thus be dominantly inherited and underlie variable neurological phenotypes, including HSP, with intrafamiliar variability. This finding extends the group of mitochondrial inner membrane AAA proteins associated with spasticity.


Assuntos
Adenosina Trifosfatases/genética , Paralisia Cerebral/genética , Proteínas de Membrana/genética , Proteínas Mitocondriais/genética , Paraplegia Espástica Hereditária/genética , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/biossíntese , Adolescente , Adulto , Axônios/metabolismo , Axônios/patologia , Paralisia Cerebral/patologia , Pré-Escolar , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Proteínas de Membrana/biossíntese , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Dinâmica Mitocondrial/genética , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/patologia , Proteínas Mitocondriais/biossíntese , Mutação , Paraplegia Espástica Hereditária/patologia , Serina-Treonina Quinases TOR/genética
6.
Biochemistry ; 56(9): 1227-1238, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28206745

RESUMO

DNA polymerase γ (Pol γ) is a key component of the mitochondrial DNA replisome and an important cause of neurological diseases. Despite the availability of its crystal structures, the molecular mechanism of DNA replication, the switch between polymerase and exonuclease activities, the site of replisomal interactions, and functional effects of patient mutations that do not affect direct catalysis have remained elusive. Here we report the first atomistic classical molecular dynamics simulations of the human Pol γ replicative complex. Our simulation data show that DNA binding triggers remarkable changes in the enzyme structure, including (1) completion of the DNA-binding channel via a dynamic subdomain, which in the apo form blocks the catalytic site, (2) stabilization of the structure through the distal accessory ß-subunit, and (3) formation of a putative transient replisome-binding platform in the "intrinsic processivity" subdomain of the enzyme. Our data indicate that noncatalytic mutations may disrupt replisomal interactions, thereby causing Pol γ-associated neurodegenerative disorders.


Assuntos
DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/metabolismo , Mitocôndrias/enzimologia , Simulação de Dinâmica Molecular , Domínio Catalítico , DNA/metabolismo , DNA Polimerase gama , Humanos , Mutação , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/genética , Estrutura Secundária de Proteína , Rotação
7.
Am J Med Genet A ; 173(1): 225-230, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27683074

RESUMO

Isolated defects of the mitochondrial respiratory complex II (succinate dehydrogenase, SDH) are rare, accounting for approximately 2% of all respiratory chain deficiency diagnoses. Here, we report clinical and molecular investigations of three family members with a heterozygous mutation in the large flavoprotein subunit SDHA previously described to cause complex II deficiency. The index patient presented with bilateral optic atrophy and ocular movement disorder, a progressive polyneuropathy, psychiatric involvement, and cardiomyopathy. Two of his children presented with cardiomyopathy and methylglutaconic aciduria in early childhood. The daughter deceased at the age of 7 months due to cardiac insufficiency. The 30-year old son presents with cardiomyopathy and developed bilateral optic atrophy in adulthood. Of the four nuclear encoded proteins composing complex II (SDHA, SDHB, SDHC, SDHD) and currently known assembly factors SDHAF1 and SDHAF2 mainly recessively inherited mutations have been described in SDHA, SDHB, SDHD, and SDHAF1 to be causative for mitochondrial disease phenotypes. This is the second report presenting autosomal dominant inheritance of a SDHA mutation.© 2016 Wiley Periodicals, Inc.


Assuntos
Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Complexo II de Transporte de Elétrons/deficiência , Mutação , Fenótipo , Succinato Desidrogenase/genética , Adolescente , Alelos , Substituição de Aminoácidos , Biomarcadores , Códon , Análise Mutacional de DNA , Evolução Fatal , Feminino , Genes Mitocondriais , Genótipo , Humanos , Masculino , Modelos Moleculares , Linhagem , Conformação Proteica , Succinato Desidrogenase/química
8.
Am J Med Genet A ; 170(6): 1433-8, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26888048

RESUMO

We report a 10-year-old girl presenting with severe neonatal hypertrophic cardiomyopathy (HCM), feeding difficulties, mildly abnormal facial features, and progressive skeletal muscle symptoms but with normal cognitive development. Targeted oligonucleotide-selective sequencing of 101 cardiomyopathy genes revealed the genetic diagnosis, and the mutation was verified by Sanger sequencing in the patient and her parents. To offer insights into the potential mechanism of patient mutation, protein structural analysis was performed using the resolved structure of human activated HRAS protein with bound GTP analogue (PDB id 5P21) in Discovery Studio 4.5 (Dassault Systèmes Biovia, San Diego, CA). The patient with hypertrophic cardiomyopathy and normal cognitive development was diagnosed with an HRAS mutation c.173C>T (p.T58I), a milder variant of Costello syndrome affecting a highly conserved amino acid, threonine 58. Our analysis suggests that the p.G12 mutations slow GTP hydrolysis rendering HRAS unresponsive to GTPase activating proteins, and resulting in permanently active state. The p.T58I mutation likely affects binding of guanidine-nucleotide-exchange factors, thereby promoting the active state but also allowing for slow inactivation. Patients with the HRAS mutation c.173C>T (p.T58I) might go undiagnosed because of the milder phenotype compared with other mutations causing Costello syndrome. We expand the clinical and molecular picture of the rare HRAS mutation by reporting the first case in Europe and the fourth case in the literature. Our protein structure analysis offers insights into the mechanism of the mildly activating p.T58I mutation. © 2016 Wiley Periodicals, Inc.


Assuntos
Cardiomiopatia Hipertrófica/diagnóstico , Cardiomiopatia Hipertrófica/genética , Síndrome de Costello/diagnóstico , Síndrome de Costello/genética , Mutação , Fenótipo , Proteínas Proto-Oncogênicas p21(ras)/genética , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Alelos , Biomarcadores , Análise Mutacional de DNA , Ecocardiografia , Estudos de Associação Genética , Testes Genéticos , Genótipo , Humanos , Recém-Nascido , Masculino , Radiografia Torácica , Índice de Gravidade de Doença
9.
Hum Mol Genet ; 21(20): 4521-9, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22833457

RESUMO

Next-generation sequencing has turned out to be a powerful tool to uncover genetic basis of childhood mitochondrial disorders. We utilized whole-exome analysis and discovered novel compound heterozygous mutations in FARS2 (mitochondrial phenylalanyl transfer RNA synthetase), encoding the mitochondrial phenylalanyl transfer RNA (tRNA) synthetase (mtPheRS) in two patients with fatal epileptic mitochondrial encephalopathy. The mutations affected highly conserved amino acids, p.I329T and p.D391V. Recently, a homozygous FARS2 variant p.Y144C was reported in a Saudi girl with mitochondrial encephalopathy, but the pathogenic role of the variant remained open. Clinical features, including postnatal onset, catastrophic epilepsy, lactic acidemia, early lethality and neuroimaging findings of the patients with FARS2 variants, resembled each other closely, and neuropathology was consistent with Alpers syndrome. Our structural analysis of mtPheRS predicted that p.I329T weakened ATP binding in the aminoacylation domain, and in vitro studies with recombinant mutant protein showed decreased affinity of this variant to ATP. Furthermore, p.D391V and p.Y144C were predicted to disrupt synthetase function by interrupting the rotation of the tRNA anticodon stem-binding domain from a closed to an open form. In vitro characterization indicated reduced affinity of p.D391V mutant protein to phenylalanine, whereas p.Y144C disrupted tRNA binding. The stability of p.I329T and p.D391V mutants in a refolding assay was impaired. Our results imply that the three FARS2 mutations directly impair aminoacylation function and stability of mtPheRS, leading to a decrease in overall tRNA charging capacity. This study establishes a new genetic cause of infantile mitochondrial Alpers encephalopathy and reports a new mitochondrial aminoacyl-tRNA synthetase as a cause of mitochondrial disease.


Assuntos
Esclerose Cerebral Difusa de Schilder/genética , Mitocôndrias/enzimologia , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Fenilalanina-tRNA Ligase/genética , Sequência de Aminoácidos , Anticódon/metabolismo , Sequência de Bases , Esclerose Cerebral Difusa de Schilder/enzimologia , Esclerose Cerebral Difusa de Schilder/metabolismo , Exoma , Feminino , Humanos , Lactente , Mitocôndrias/metabolismo , Doenças Mitocondriais/enzimologia , Doenças Mitocondriais/metabolismo , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Dados de Sequência Molecular , Mutação , Fenilalanina-tRNA Ligase/química , Fenilalanina-tRNA Ligase/metabolismo , Dobramento de Proteína , RNA de Transferência/genética , RNA de Transferência/metabolismo
10.
Am J Hum Genet ; 88(5): 635-42, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21549344

RESUMO

Infantile cardiomyopathies are devastating fatal disorders of the neonatal period or the first year of life. Mitochondrial dysfunction is a common cause of this group of diseases, but the underlying gene defects have been characterized in only a minority of cases, because tissue specificity of the manifestation hampers functional cloning and the heterogeneity of causative factors hinders collection of informative family materials. We sequenced the exome of a patient who died at the age of 10 months of hypertrophic mitochondrial cardiomyopathy with combined cardiac respiratory chain complex I and IV deficiency. Rigorous data analysis allowed us to identify a homozygous missense mutation in AARS2, which we showed to encode the mitochondrial alanyl-tRNA synthetase (mtAlaRS). Two siblings from another family, both of whom died perinatally of hypertrophic cardiomyopathy, had the same mutation, compound heterozygous with another missense mutation. Protein structure modeling of mtAlaRS suggested that one of the mutations affected a unique tRNA recognition site in the editing domain, leading to incorrect tRNA aminoacylation, whereas the second mutation severely disturbed the catalytic function, preventing tRNA aminoacylation. We show here that mutations in AARS2 cause perinatal or infantile cardiomyopathy with near-total combined mitochondrial respiratory chain deficiency in the heart. Our results indicate that exome sequencing is a powerful tool for identifying mutations in single patients and allows recognition of the genetic background in single-gene disorders of variable clinical manifestation and tissue-specific disease. Furthermore, we show that mitochondrial disorders extend to prenatal life and are an important cause of early infantile cardiac failure.


Assuntos
Alanina-tRNA Ligase/genética , Cardiomiopatia Hipertrófica/genética , Mitocôndrias/genética , Doenças Mitocondriais/genética , Mutação de Sentido Incorreto , Pareamento de Bases , Cardiomiopatia Hipertrófica/patologia , Análise Mutacional de DNA , DNA Mitocondrial/genética , Feminino , Humanos , Lactente , Recém-Nascido , Doenças Mitocondriais/patologia , Linhagem , Estrutura Terciária de Proteína
11.
J Med Genet ; 50(3): 151-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23315540

RESUMO

BACKGROUND: The genetic complexity of infantile cardiomyopathies is remarkable, and the importance of mitochondrial translation defects as a causative factor is only starting to be recognised. We investigated the genetic basis for infantile onset recessive hypertrophic cardiomyopathy in two siblings. METHODS AND RESULTS: Analysis of respiratory chain enzymes revealed a combined deficiency of complexes I and IV in the heart and skeletal muscle. Exome sequencing uncovered a homozygous mutation (L156R) in MRPL44 of both siblings. MRPL44 encodes a protein in the large subunit of the mitochondrial ribosome and is suggested to locate in close proximity to the tunnel exit of the yeast mitochondrial ribosome. We found severely reduced MRPL44 levels in the patient's heart, skeletal muscle and fibroblasts suggesting that the missense mutation affected the protein stability. In patient fibroblasts, decreased MRPL44 affected assembly of the large ribosomal subunit and stability of 16S rRNA leading to complex IV deficiency. Despite this assembly defect, de novo mitochondrial translation was only mildly affected in fibroblasts suggesting that MRPL44 may have a function in the assembly/stability of nascent mitochondrial polypeptides exiting the ribosome. Retroviral expression of wild-type MRPL44 in patient fibroblasts rescued the large ribosome assembly defect and COX deficiency. CONCLUSIONS: These findings indicate that mitochondrial ribosomal subunit defects can generate tissue-specific manifestations, such as cardiomyopathy.


Assuntos
Cardiomiopatia Hipertrófica/genética , Exoma , Doenças Mitocondriais/genética , Proteínas Mitocondriais/genética , Mutação , Proteínas Ribossômicas/genética , Adolescente , Sequência de Aminoácidos , Cardiomiopatia Hipertrófica/congênito , Ciclo-Oxigenase 1 , Complexo I de Transporte de Elétrons , Complexo IV da Cadeia de Transporte de Elétrons , Exoma/genética , Evolução Fatal , Feminino , Fibroblastos/metabolismo , Humanos , Lactente , Doenças Mitocondriais/congênito , Dados de Sequência Molecular , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Miocárdio/química , Miocárdio/metabolismo , Linhagem , Alinhamento de Sequência , Análise de Sequência de DNA
12.
Nucleic Acids Res ; 39(21): 9072-84, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21824913

RESUMO

Mutations in Pol γ represent a major cause of human mitochondrial diseases, especially those affecting the nervous system in adults and in children. Recessive mutations in Pol γ represent nearly half of those reported to date, and they are nearly uniformly distributed along the length of the POLG1 gene (Human DNA Polymerase gamma Mutation Database); the majority of them are linked to the most severe form of POLG syndrome, Alpers-Huttenlocher syndrome. In this report, we assess the structure-function relationships for recessive disease mutations by reviewing existing biochemical data on site-directed mutagenesis of the human, Drosophila and yeast Pol γs, and their homologs from the family A DNA polymerase group. We do so in the context of a molecular model of Pol γ in complex with primer-template DNA, which we have developed based upon the recently solved crystal structure of the apoenzyme form. We present evidence that recessive mutations cluster within five distinct functional modules in the catalytic core of Pol γ. Our results suggest that cluster prediction can be used as a diagnosis-supporting tool to evaluate the pathogenic role of new Pol γ variants.


Assuntos
DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/genética , Esclerose Cerebral Difusa de Schilder/genética , Mutação , Biocatálise , Domínio Catalítico/genética , DNA Polimerase gama , DNA Polimerase Dirigida por DNA/metabolismo , Humanos , Mitocôndrias/enzimologia
13.
J Pediatr ; 160(3): 473-479.e1, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21962599

RESUMO

OBJECTIVE: To assess the long-term clinical course of carnitine palmitoyltransferase 1A (CPT1A) deficiency, caused by the c.1364A>C (p.K455T) mutation, and the carrier frequency of this mutation in Finland. STUDY DESIGN: This was a long-term follow-up of patients in whom the common mutation was detected. RESULTS: Between 1999 and 2010, 6 cases of CPT1A deficiency were diagnosed and treated with a high-carbohydrate, low-fat diet. The patients experienced their first symptoms during the first years of life, provoked by viral illness and/or fasting. The clinical features included hypoketotic hypoglycemia, hepatopathy, and loss of consciousness, ranging from transient unconsciousness to prolonged hyperlipidemic coma. Five cases carried a homozygous c.1364A>C (p.K455T) mutation, whereas 1 case had a compound c.1364A>C/c.1493A>C (p.Y498S) mutation. During dietary therapy, the patients had few transient decompensations. No carriers of mutation c.1364A>C were detected by minisequencing of 150 control samples. CONCLUSION: Even though CPT1A deficiency may be life-threatening and lead to prolonged coma, the long-term prognosis is good. A genotype-phenotype correlation implies that the mutations detected are disease-causing. Despite Finland's location close to the Arctic polar region, the carrier frequency of the c.1364A>C mutation in Finland is far lower than that of the variants found in Alaskan, Canadian, and Greenland native populations.


Assuntos
Carnitina O-Palmitoiltransferase/genética , Heterozigoto , Mutação de Sentido Incorreto , Adolescente , Adulto , Western Blotting , Carnitina O-Palmitoiltransferase/deficiência , Carnitina O-Palmitoiltransferase/metabolismo , Criança , Desenvolvimento Infantil , Pré-Escolar , Dietoterapia , Feminino , Finlândia/epidemiologia , Seguimentos , Estudos de Associação Genética , Humanos , Masculino , Adulto Jovem
14.
Biochim Biophys Acta ; 1802(6): 545-51, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20153822

RESUMO

Mitochondrial DNA polymerase, POLG, is the sole DNA polymerase found in animal mitochondria. In humans, POLGalpha W748S in cis with an E1143G mutation has been linked to a new type of recessive ataxia, MIRAS, which is the most common inherited ataxia in Finland. We investigated the biochemical phenotypes of the W748S amino acid change, using recombinant human POLG. We measured processive and non-processive DNA polymerase activity, DNA binding affinity, enzyme processivity, and subunit interaction with recombinant POLGbeta. In addition, we studied the effects of the W748S and E1143G mutations in primary human cell cultures using retroviral transduction. Here, we examined cell viability, mitochondrial DNA copy number, and products of mitochondrial translation. Our results indicate that the W748S mutant POLGalpha does not exhibit a clear biochemical phenotype, making it indistinguishable from wild type POLGalpha and as such, fail to replicate previously published results. Furthermore, results from the cell models were concurrent with the findings from patients, and support our biochemical findings.


Assuntos
DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Variação Genética , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Domínio Catalítico/genética , Células Cultivadas , DNA Polimerase gama , Primers do DNA/genética , DNA Intergênico/genética , DNA Mitocondrial/genética , DNA Polimerase Dirigida por DNA/química , Genes Recessivos , Humanos , Técnicas In Vitro , Modelos Moleculares , Mutagênese Sítio-Dirigida , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Degenerações Espinocerebelares/enzimologia , Degenerações Espinocerebelares/genética , Síndrome
15.
Proc Natl Acad Sci U S A ; 105(10): 3763-7, 2008 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-18316732

RESUMO

Electron transfer in complex I from Escherichia coli was investigated by an ultrafast freeze-quench approach. The reaction of complex I with NADH was stopped in the time domain from 90 mus to 8 ms and analyzed by electron paramagnetic resonance (EPR) spectroscopy at low temperatures. The data show that after binding of the first molecule of NADH, two electrons move via the FMN cofactor to the iron-sulfur (Fe/S) centers N1a and N2 with an apparent time constant of approximately 90 mus, implying that these two centers should have the highest redox potential in the enzyme. The rate of reduction of center N2 (the last center in the electron transfer sequence) is close to that predicted by electron transfer theory, which argues for the absence of coupled proton transfer or conformational changes during electron transfer from FMN to N2. After fast reduction of N1a and N2, we observe a slow, approximately 1-ms component of reduction of other Fe/S clusters. Because all elementary electron transfer rates between clusters are several orders of magnitude higher than this observed rate, we conclude that the millisecond component is limited by a single process corresponding to dissociation of the oxidized NAD(+) molecule from its binding site, where it prevents entry of the next NADH molecule. Despite the presence of approximately one ubiquinone per enzyme molecule, no transient semiquinone formation was observed, which has mechanistic implications, suggesting a high thermodynamic barrier for ubiquinone reduction to the semiquinone radical. Possible consequences of these findings for the proton translocation mechanism are discussed.


Assuntos
Complexo I de Transporte de Elétrons/química , Escherichia coli/química , Espectroscopia de Ressonância de Spin Eletrônica , Transporte de Elétrons , Cinética , NAD , Oxirredução , Thermus thermophilus
16.
Biochim Biophys Acta ; 1787(8): 1024-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19261245

RESUMO

Studies on the activity of Complex I from Escherichia coli in the presence of different metal cations revealed at least two high affinity metal-binding sites. Membrane-bound or isolated Complex I was activated by K(+) (apparent binding constant approximately 125 microM) and inhibited by La(3+) (IC(50)= 1 microM). K(+) and La(3+) do not occupy the same site. Possible localization of these metal-binding sites and their implication in catalysis are discussed.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Sítios de Ligação , Cátions/metabolismo , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/isolamento & purificação , Escherichia coli/genética , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Cinética , Lantânio/metabolismo , Lantânio/farmacologia , Mutação , Potássio/metabolismo , Potássio/farmacologia , Ligação Proteica , Quinona Redutases/antagonistas & inibidores , Quinona Redutases/genética , Quinona Redutases/metabolismo
17.
Biochim Biophys Acta ; 1787(1): 68-73, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19061856

RESUMO

Replacement of glutamate 95 for glutamine in the NADH- and FMN-binding NuoF subunit of E. coli Complex I decreased NADH oxidation activity 2.5-4.8 times depending on the used electron acceptor. The apparent K(m) for NADH was 5.2 and 10.4 microM for the mutant and wild type, respectively. Analysis of the inhibitory effect of NAD(+) on activity showed that the E95Q mutation caused a 2.4-fold decrease of K(i)(NAD+) in comparison to the wild type enzyme. ADP-ribose, which differs from NAD(+) by the absence of the positively charged nicotinamide moiety, is also a competitive inhibitor of NADH binding. The mutation caused a 7.5-fold decrease of K(i)(ADP-ribose) relative to wild type enzyme. Based on these findings we propose that the negative charge of Glu95 accelerates turnover of Complex I by electrostatic interaction with the negatively charged phosphate groups of the substrate nucleotide during operation, which facilitates release of the product NAD(+). The E95Q mutation was also found to cause a positive shift of the midpoint redox potential of the FMN, from -350 mV to -310 mV, which suggests that the negative charge of Glu95 is also involved in decreasing the midpoint potential of the primary electron acceptor of Complex I.


Assuntos
Complexo I de Transporte de Elétrons/química , Proteínas de Escherichia coli/química , Glutamina/genética , Domínio Catalítico , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Cinética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Mutação , NAD/metabolismo , Oxirredução , Ligação Proteica , Quinona Redutases/química , Quinona Redutases/genética , Quinona Redutases/metabolismo
18.
Nat Commun ; 11(1): 5927, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33230181

RESUMO

Mitochondrial acyl-coenzyme A species are emerging as important sources of protein modification and damage. Succinyl-CoA ligase (SCL) deficiency causes a mitochondrial encephalomyopathy of unknown pathomechanism. Here, we show that succinyl-CoA accumulates in cells derived from patients with recessive mutations in the tricarboxylic acid cycle (TCA) gene succinyl-CoA ligase subunit-ß (SUCLA2), causing global protein hyper-succinylation. Using mass spectrometry, we quantify nearly 1,000 protein succinylation sites on 366 proteins from patient-derived fibroblasts and myotubes. Interestingly, hyper-succinylated proteins are distributed across cellular compartments, and many are known targets of the (NAD+)-dependent desuccinylase SIRT5. To test the contribution of hyper-succinylation to disease progression, we develop a zebrafish model of the SCL deficiency and find that SIRT5 gain-of-function reduces global protein succinylation and improves survival. Thus, increased succinyl-CoA levels contribute to the pathology of SCL deficiency through post-translational modifications.


Assuntos
Acil Coenzima A/metabolismo , Doenças Mitocondriais/patologia , Succinato-CoA Ligases/genética , Animais , Células Cultivadas , Feminino , Humanos , Lactente , Lisina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Doenças Mitocondriais/metabolismo , Mutação , Proteômica , Sirtuínas/deficiência , Sirtuínas/genética , Sirtuínas/metabolismo , Succinato-CoA Ligases/deficiência , Succinato-CoA Ligases/metabolismo , Análise de Sobrevida , Peixe-Zebra
19.
Biochim Biophys Acta ; 1777(9): 1166-72, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18590697

RESUMO

Analysis of the amino acid sequences of subunits NuoM and NuoN in the membrane domain of Complex I revealed a clear common pattern, including two lysines that are predicted to be located within the membrane, and which are important for quinone reductase activity. Site-directed mutations of the amino acid residues E144, K234, K265 and W243 in this pattern were introduced into the chromosomal gene nuoM of Escherichia coli Complex I. The activity of mutated Complex I was studied in both membranes and in purified Complex I. The quinone reductase activity was practically lost in K234A, K234R and E144A, decreased in W243A and K265A but unchanged in E144D. Complex I from all these mutants contained 1 mol tightly bound ubiquinone per mol FMN like wild type enzyme. The mutant enzymes E144D, W243A and K265A had wild type sensitivity to rolliniastatin and complete proton-pumping efficiency of Complex I. Remarkably, the subunits NuoL and NuoH in the membrane domain also appear to contain conserved lysine residues in transmembrane helices, which may give a clue of the mechanism of proton translocation. A tentative principle of proton translocation by Complex I is suggested based on electrostatic interactions of lysines in the membrane subunits.


Assuntos
Membrana Celular/metabolismo , Sequência Conservada , Complexo I de Transporte de Elétrons/metabolismo , Transferência de Energia , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Lisina/metabolismo , NADH Desidrogenase/química , NADH Desidrogenase/metabolismo , Sequência de Aminoácidos , Complexo I de Transporte de Elétrons/química , Escherichia coli/enzimologia , Escherichia coli/crescimento & desenvolvimento , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Mutantes/metabolismo , Mutação/genética , Fenótipo , Estrutura Secundária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Análise de Sequência de Proteína
20.
Cell Metab ; 30(6): 1040-1054.e7, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31523008

RESUMO

Mitochondrial dysfunction elicits stress responses that safeguard cellular homeostasis against metabolic insults. Mitochondrial integrated stress response (ISRmt) is a major response to mitochondrial (mt)DNA expression stress (mtDNA maintenance, translation defects), but the knowledge of dynamics or interdependence of components is lacking. We report that in mitochondrial myopathy, ISRmt progresses in temporal stages and development from early to chronic and is regulated by autocrine and endocrine effects of FGF21, a metabolic hormone with pleiotropic effects. Initial disease signs induce transcriptional ISRmt (ATF5, mitochondrial one-carbon cycle, FGF21, and GDF15). The local progression to 2nd metabolic ISRmt stage (ATF3, ATF4, glucose uptake, serine biosynthesis, and transsulfuration) is FGF21 dependent. Mitochondrial unfolded protein response marks the 3rd ISRmt stage of failing tissue. Systemically, FGF21 drives weight loss and glucose preference, and modifies metabolism and respiratory chain deficiency in a specific hippocampal brain region. Our evidence indicates that FGF21 is a local and systemic messenger of mtDNA stress in mice and humans with mitochondrial disease.


Assuntos
DNA Mitocondrial/metabolismo , Fatores de Crescimento de Fibroblastos/fisiologia , Mitocôndrias/metabolismo , Miopatias Mitocondriais/metabolismo , Estresse Fisiológico/fisiologia , Fatores Ativadores da Transcrição/metabolismo , Animais , Linhagem Celular , DNA Mitocondrial/genética , Escherichia coli , Feminino , Fatores de Crescimento de Fibroblastos/genética , Fator 15 de Diferenciação de Crescimento/metabolismo , Humanos , Masculino , Camundongos , Mitocôndrias/genética , Miopatias Mitocondriais/genética , Deleção de Sequência , Estresse Fisiológico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA