Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
J Med Genet ; 60(10): 1006-1015, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37055166

RESUMO

BACKGROUND: Enoyl-CoA hydratase short-chain 1 (ECHS1) is an enzyme involved in the metabolism of branched chain amino acids and fatty acids. Mutations in the ECHS1 gene lead to mitochondrial short-chain enoyl-CoA hydratase 1 deficiency, resulting in the accumulation of intermediates of valine. This is one of the most common causative genes in mitochondrial diseases. While genetic analysis studies have diagnosed numerous cases with ECHS1 variants, the increasing number of variants of uncertain significance (VUS) in genetic diagnosis is a major problem. METHODS: Here, we constructed an assay system to verify VUS function for ECHS1 gene. A high-throughput assay using ECHS1 knockout cells was performed to index these phenotypes by expressing cDNAs containing VUS. In parallel with the VUS validation system, a genetic analysis of samples from patients with mitochondrial disease was performed. The effect on gene expression in cases was verified by RNA-seq and proteome analysis. RESULTS: The functional validation of VUS identified novel variants causing loss of ECHS1 function. The VUS validation system also revealed the effect of the VUS in the compound heterozygous state and provided a new methodology for variant interpretation. Moreover, we performed multiomics analysis and identified a synonymous substitution p.P163= that results in splicing abnormality. The multiomics analysis complemented the diagnosis of some cases that could not be diagnosed by the VUS validation system. CONCLUSIONS: In summary, this study uncovered new ECHS1 cases based on VUS validation and omics analysis; these analyses are applicable to the functional evaluation of other genes associated with mitochondrial disease.


Assuntos
Doenças Mitocondriais , Humanos , Fenótipo , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Mutação/genética , Enoil-CoA Hidratase/genética , Enoil-CoA Hidratase/metabolismo , Testes Genéticos
2.
J Hum Genet ; 68(9): 649-652, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37246162

RESUMO

Approximately 80% of rare diseases have a genetic cause, and an accurate genetic diagnosis is necessary for disease management, prognosis prediction, and genetic counseling. Whole-exome sequencing (WES) is a cost-effective approach for exploring the genetic cause, but several cases often remain undiagnosed. We combined whole genome sequencing (WGS) and RNA sequencing (RNA-seq) to identify the pathogenic variants in an unsolved case using WES. RNA-seq revealed aberrant exon 4 and exon 6 splicing of ITPA. WGS showed a previously unreported splicing donor variant, c.263+1G>A, and a novel heterozygous deletion, including exon 6. Detailed examination of the breakpoint indicated the deletion caused by recombination between Alu elements in different introns. The proband was found to have developmental and epileptic encephalopathies caused by variants in the ITPA gene. The combination of WGS and RNA-seq may be effective in diagnosing conditions in proband who could not be diagnosed using WES.


Assuntos
Família , Pirofosfatases , Humanos , Sequenciamento do Exoma , Sequenciamento Completo do Genoma , Éxons , Análise de Sequência de RNA
4.
Int Heart J ; 63(5): 970-977, 2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36104228

RESUMO

Hypertrophic cardiomyopathy is a common cardiac complication in mitochondrial disorders, and the morbidity rate in neonatal cases is up to 40%. The mortality rate within 3 months for neonatal-onset mitochondrial cardiomyopathy is known to be high because there is currently no established treatment.We report the case of a male infant with neonatal-onset mitochondrial disorder presenting lactic acidosis and hypertrophic cardiomyopathy. Genetic analysis of the patient revealed recurrent m.13513G>A, p.Asp393Asn in mitochondrially encoded NADH dehydrogenase 5 gene (MT-ND5). Low-dose propranolol was initially administered for cardiomyopathy; however, he developed hypertrophic obstructive cardiomyopathy (HOCM) at 3 months of age. To reduce the risk of hypoglycemia associated with high-dose propranolol, cibenzoline, a class Ia antiarrhythmic drug, was added at a dose of 2.5 mg/kg/day and increased weekly to 7.5 mg/kg/day with monitoring of the blood concentration of cibenzoline. Left ventricular outflow tract stenosis (LVOTS) dramatically improved from 5.4 to 1.3 m/second in LVOTS peak velocity after 6 weeks, without notable adverse effects. The plasma N-terminal pro-brain natriuretic peptide level decreased from 65,854 to 10,044 pg/mL. Furthermore, myocardial hypertrophy also improved, as the left ventricular mass index decreased from 173.1 to 108.9 g/m2 after 3 months of the treatment.The administration of cibenzoline, in conjunction with low-dose propranolol, may serve an effective treatment for HOCM in infantile patients with mitochondrial disorders.


Assuntos
Antiarrítmicos , Cardiomiopatia Hipertrófica , Antiarrítmicos/uso terapêutico , Cardiomiopatia Hipertrófica/complicações , Cardiomiopatia Hipertrófica/diagnóstico , Cardiomiopatia Hipertrófica/tratamento farmacológico , Constrição Patológica , Humanos , Imidazóis , Recém-Nascido , Masculino , NADH Desidrogenase/farmacologia , NADH Desidrogenase/uso terapêutico , Propranolol/farmacologia , Propranolol/uso terapêutico , Função Ventricular Esquerda
5.
Hum Mutat ; 42(11): 1422-1428, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34405929

RESUMO

Isolated complex I deficiency is the most common cause of pediatric mitochondrial disease. Exome sequencing (ES) has revealed many complex I causative genes. However, there are limitations associated with identifying causative genes by ES analysis. In this study, we performed multiomics analysis to reveal the causal variants. We here report two cases with mitochondrial complex I deficiency. In both cases, ES identified a novel c.580G>A (p.Glu194Lys) variant in NDUFV2. One case additionally harbored c.427C>T (p.Arg143*), but no other variants were observed in the other case. RNA sequencing showed aberrant exon splicing of NDUFV2 in the unsolved case. Genome sequencing revealed a novel heterozygous deletion in NDUFV2, which included one exon and resulted in exon skipping. Detailed examination of the breakpoint revealed that an Alu insertion-mediated rearrangement caused the deletion. Our report reveals that combined use of transcriptome sequencing and GS was effective for diagnosing cases that were unresolved by ES.


Assuntos
Elementos Alu , Complexo I de Transporte de Elétrons/deficiência , Deleção de Genes , Genoma Humano , Mutação INDEL , Doenças Mitocondriais/genética , NADH Desidrogenase/genética , Análise de Sequência de RNA/métodos , Complexo I de Transporte de Elétrons/genética , Feminino , Humanos , Lactente , Masculino , Doenças Mitocondriais/diagnóstico , Linhagem
6.
Am J Hum Genet ; 103(2): 221-231, 2018 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-30057030

RESUMO

Bloom syndrome, caused by biallelic mutations in BLM, is characterized by prenatal-onset growth deficiency, short stature, an erythematous photosensitive malar rash, and increased cancer predisposition. Diagnostically, a hallmark feature is the presence of increased sister chromatid exchanges (SCEs) on cytogenetic testing. Here, we describe biallelic mutations in TOP3A in ten individuals with prenatal-onset growth restriction and microcephaly. TOP3A encodes topoisomerase III alpha (TopIIIα), which binds to BLM as part of the BTRR complex, and promotes dissolution of double Holliday junctions arising during homologous recombination. We also identify a homozygous truncating variant in RMI1, which encodes another component of the BTRR complex, in two individuals with microcephalic dwarfism. The TOP3A mutations substantially reduce cellular levels of TopIIIα, and consequently subjects' cells demonstrate elevated rates of SCE. Unresolved DNA recombination and/or replication intermediates persist into mitosis, leading to chromosome segregation defects and genome instability that most likely explain the growth restriction seen in these subjects and in Bloom syndrome. Clinical features of mitochondrial dysfunction are evident in several individuals with biallelic TOP3A mutations, consistent with the recently reported additional function of TopIIIα in mitochondrial DNA decatenation. In summary, our findings establish TOP3A mutations as an additional cause of prenatal-onset short stature with increased cytogenetic SCEs and implicate the decatenation activity of the BTRR complex in their pathogenesis.

7.
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
8.
Clin Genet ; 98(2): 155-165, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32385911

RESUMO

Mitochondrial complex I deficiency is caused by pathogenic variants in mitochondrial and nuclear genes associated with complex I structure and assembly. We report the case of a patient with NDUFA8-related mitochondrial disease. The patient presented with developmental delay, microcephaly, and epilepsy. His fibroblasts showed apparent biochemical defects in mitochondrial complex I. Whole-exome sequencing revealed that the patient carried a homozygous variant in NDUFA8. His fibroblasts showed a reduction in the protein expression level of not only NDUFA8, but also the other complex I subunits, consistent with assembly defects. The enzyme activity of complex I and oxygen consumption rate were restored by reintroducing wild-typeNDUFA8 cDNA into patient fibroblasts. The functional properties of the variant in NDUFA8 were also investigated using NDUFA8 knockout cells expressing wild-type or mutated NDUFA8 cDNA. These experiments further supported the pathogenicity of the variant in complex I assembly. This is the first report describing that the loss of NDUFA8, which has not previously been associated with mitochondrial disease, causes severe defect in the assembly of mitochondrial complex I, leading to progressive neurological and developmental abnormalities.


Assuntos
Deficiências do Desenvolvimento/genética , Complexo I de Transporte de Elétrons/deficiência , Microcefalia/genética , Doenças Mitocondriais/genética , NADH Desidrogenase/genética , Adolescente , Adulto , Criança , Pré-Escolar , Deficiências do Desenvolvimento/diagnóstico por imagem , Deficiências do Desenvolvimento/fisiopatologia , Complexo I de Transporte de Elétrons/genética , Epilepsia/diagnóstico por imagem , Epilepsia/genética , Epilepsia/fisiopatologia , Técnicas de Inativação de Genes , Predisposição Genética para Doença , Homozigoto , Humanos , Lactente , Masculino , Microcefalia/diagnóstico por imagem , Microcefalia/fisiopatologia , Doenças Mitocondriais/diagnóstico por imagem , Doenças Mitocondriais/fisiopatologia , Adulto Jovem
9.
J Inherit Metab Dis ; 43(4): 819-826, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31967322

RESUMO

Leigh syndrome is a major phenotype of mitochondrial diseases in children. With new therapeutic options being proposed, assessing the mortality and clinical condition of Leigh syndrome patients is crucial for evaluating therapeutics. As data are scarce in Japan, we analysed the mortality rate and clinical condition of Japanese Leigh syndrome patients that we diagnosed since 2007. Data from 166 Japanese patients diagnosed with Leigh syndrome from 2007 to 2017 were reviewed. Patients' present status, method of ventilation and feeding, and degree of disability as of April 2018 was analysed. Overall, 124 (74.7%) were living, 40 (24.1%) were deceased, and 2 (1.2%) were lost to follow-up. Median age of living patients was 8 years (1-39 years). Median length of disease course was 91 months for living patients and 23.5 months for deceased patients. Nearly 90% of deaths occurred by age 6. Mortality rate of patients with onset before 6 months of age was significantly higher than that of onset after 6 months. All patients with neonatal onset were either deceased or bedridden. MT-ATP6 deficiency caused by m.8993T>G mutation and MT-ND5 deficiency induced a severe form of Leigh syndrome. Patients with NDUFAF6, ECHS1, and SURF1 deficiency had relatively mild symptoms and better survival. The impact of onset age on prognosis varied across the genetic diagnoses. The clinical condition of many patients was poor; however, few did not require mechanical ventilation or tube-feeding and were not physically dependent. Early disease onset and genetic diagnosis may have prognostic value.


Assuntos
Doença de Leigh/genética , Doença de Leigh/mortalidade , Adolescente , Adulto , Idade de Início , Criança , Pré-Escolar , DNA/genética , DNA Mitocondrial/genética , Feminino , Humanos , Lactente , Japão/epidemiologia , Estimativa de Kaplan-Meier , Doença de Leigh/diagnóstico , Imageamento por Ressonância Magnética , Masculino , Mutação/genética , Fenótipo , Taxa de Sobrevida , Adulto Jovem
10.
BMC Nephrol ; 21(1): 376, 2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32859164

RESUMO

BACKGROUND: Primary coenzyme Q10 (CoQ10) deficiency of genetic origin is one of a few treatable focal segmental glomerulosclerosis (FSGS). Renal morphologic evidence for COQ8B mutation and CoQ10 deficiencies of other gene mutations is assessed using electron microscopy with marked increase of abnormal-shaped mitochondria in podocytes. However, light microscopic morphologic features of deficiencies other than FSGS have not been reported. CASE PRESENTATION: A 30-year-old woman was admitted to our hospital because proteinuria was found during four consecutive medical checkups. She had no medical history or family history of proteinuria and severe renal dysfunction. The swollen podocytes were stained to the same extent as mitochondria-rich proximal tubular cells under both Masson's trichrome and hematoxylin-eosin staining, whereas no mitochondrial abnormalities were detected under the first electron microscopic views. As proteinuria and estimated glomerular filtration rate (eGFR) deteriorated after pregnancy, we reevaluated the additional electron microscopic views and detected mitochondrial abnormalities. Genetic testing revealed COQ8B mutation (c.532C > T, p.R178W); therefore, we diagnosed COQ8B nephropathy. CoQ10 supplementation improved proteinuria and stopped eGFR reduction. CONCLUSIONS: This is the first report of granular swollen podocytes due to mitochondrial diseases detected under light microscopy. We propose that this finding can be the clue for the diagnosis of both COQ8B nephropathy and the other CoQ10 deficiencies.


Assuntos
Glomerulosclerose Segmentar e Focal/patologia , Mitocôndrias/ultraestrutura , Doenças Mitocondriais/patologia , Podócitos/ultraestrutura , Proteínas Quinases/genética , Adulto , Feminino , Glomerulosclerose Segmentar e Focal/genética , Humanos , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/genética , Mutação de Sentido Incorreto
11.
Nucleic Acids Res ; 46(4): 1565-1583, 2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29390138

RESUMO

Modified uridine containing taurine, 5-taurinomethyluridine (τm5U), is found at the anticodon first position of mitochondrial (mt-)transfer RNAs (tRNAs). Previously, we reported that τm5U is absent in mt-tRNAs with pathogenic mutations associated with mitochondrial diseases. However, biogenesis and physiological role of τm5U remained elusive. Here, we elucidated τm5U biogenesis by confirming that 5,10-methylene-tetrahydrofolate and taurine are metabolic substrates for τm5U formation catalyzed by MTO1 and GTPBP3. GTPBP3-knockout cells exhibited respiratory defects and reduced mitochondrial translation. Very little τm5U34 was detected in patient's cells with the GTPBP3 mutation, demonstrating that lack of τm5U results in pathological consequences. Taurine starvation resulted in downregulation of τm5U frequency in cultured cells and animal tissues (cat liver and flatfish). Strikingly, 5-carboxymethylaminomethyluridine (cmnm5U), in which the taurine moiety of τm5U is replaced with glycine, was detected in mt-tRNAs from taurine-depleted cells. These results indicate that tRNA modifications are dynamically regulated via sensing of intracellular metabolites under physiological condition.


Assuntos
RNA de Transferência/metabolismo , Taurina/deficiência , Uridina/análogos & derivados , Animais , Proteínas de Transporte/fisiologia , Gatos , Pré-Escolar , Feminino , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/fisiologia , Células HEK293 , Células HeLa , Humanos , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , RNA de Transferência/química , Proteínas de Ligação a RNA , Uridina/biossíntese
12.
Neurogenetics ; 20(1): 9-25, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30607703

RESUMO

Pentatricopeptide repeat domain proteins are a large family of RNA-binding proteins involved in mitochondrial RNA editing, stability, and translation. Mitochondrial translation machinery defects are an expanding group of genetic diseases in humans. We describe a patient who presented with low birth weight, mental retardation, and optic atrophy. Brain MRI showed abnormal bilateral signals at the basal ganglia and brainstem, and the patient was diagnosed as Leigh syndrome. Exome sequencing revealed two potentially loss-of-function variants [c.415-2A>G, and c.1747_1748insCT (p.Phe583Serfs*3)] in PTCD3 (also known as MRPS39). PTCD3, a member of the pentatricopeptide repeat domain protein family, is a component of the small mitoribosomal subunit. The patient had marked decreases in mitochondrial complex I and IV levels and activities, oxygen consumption and ATP biosynthesis, and generalized mitochondrial translation defects in fibroblasts. Quantitative proteomic analysis revealed decreased levels of the small mitoribosomal subunits. Complementation experiments rescued oxidative phosphorylation complex I and IV levels and activities, ATP biosynthesis, and MT-RNR1 rRNA transcript level, providing functional validation of the pathogenicity of identified variants. This is the first report of an association of PTCD3 mutations with Leigh syndrome along with combined oxidative phosphorylation deficiencies caused by defects in the mitochondrial translation machinery.


Assuntos
Proteínas de Arabidopsis/genética , Doença de Leigh/genética , Mutação/genética , Fosforilação Oxidativa , Proteínas de Ligação a RNA/genética , Feminino , Humanos , Mitocôndrias/genética , Linhagem
13.
PLoS Genet ; 12(1): e1005679, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26741492

RESUMO

Mitochondrial disorders have the highest incidence among congenital metabolic disorders characterized by biochemical respiratory chain complex deficiencies. It occurs at a rate of 1 in 5,000 births, and has phenotypic and genetic heterogeneity. Mutations in about 1,500 nuclear encoded mitochondrial proteins may cause mitochondrial dysfunction of energy production and mitochondrial disorders. More than 250 genes that cause mitochondrial disorders have been reported to date. However exact genetic diagnosis for patients still remained largely unknown. To reveal this heterogeneity, we performed comprehensive genomic analyses for 142 patients with childhood-onset mitochondrial respiratory chain complex deficiencies. The approach includes whole mtDNA and exome analyses using high-throughput sequencing, and chromosomal aberration analyses using high-density oligonucleotide arrays. We identified 37 novel mutations in known mitochondrial disease genes and 3 mitochondria-related genes (MRPS23, QRSL1, and PNPLA4) as novel causative genes. We also identified 2 genes known to cause monogenic diseases (MECP2 and TNNI3) and 3 chromosomal aberrations (6q24.3-q25.1, 17p12, and 22q11.21) as causes in this cohort. Our approaches enhance the ability to identify pathogenic gene mutations in patients with biochemically defined mitochondrial respiratory chain complex deficiencies in clinical settings. They also underscore clinical and genetic heterogeneity and will improve patient care of this complex disorder.


Assuntos
Exoma/genética , Heterogeneidade Genética , Mitocôndrias/genética , Doenças Mitocondriais/genética , Adolescente , Criança , Pré-Escolar , Aberrações Cromossômicas , DNA Mitocondrial/genética , Feminino , Fibroblastos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mutação INDEL/genética , Lactente , Recém-Nascido , Masculino , Mitocôndrias/patologia , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/patologia , Polimorfismo de Nucleotídeo Único/genética
14.
Plant J ; 91(3): 455-465, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28407383

RESUMO

Chloroplast and mitochondrial DNA encodes genes that are essential for photosynthesis and respiration, respectively. Thus, loss of integrity of the genomic DNA of organelles leads to a decline in organelle function and alteration of organelle genetic information. RECA (RECA1 and RECA2) and RECG, which are homologs of bacterial homologous recombination repair (HRR) factors RecA and RecG, respectively, play an important role in the maintenance of integrity of the organelle genome by suppressing aberrant recombination between short dispersed repeats (SDRs) in the moss Physcomitrella patens. On the other hand, MutS homolog 1 (MSH1), a plant-specific MSH with a C-terminal GIY-YIG endonuclease domain, is involved in the maintenance of integrity of the organelle genome in the angiosperm Arabidopsis thaliana. Here, we address the role of the duplicated MSH1 genes, MSH1A and MSH1B, in P. patens, in which MSH1A lacks the C-terminal endonuclease domain. MSH1A and MSH1B localized to both chloroplast and mitochondrial nucleoids in protoplast cells. Single and double knockout (KO) mutants of MSH1A and MSH1B showed no obvious morphological defects; however, MSH1B KO and double KO mutants, as well as MSH1B GIY-YIG deletion mutants, exhibited genomic instability due to recombination between SDRs in chloroplasts and mitochondria. Creating double KO mutations of each combination of MSH1B, RECA2 and RECG synergistically increased recombination between SDRs in chloroplasts and mitochondria. These results show the role of MSH1 in the maintenance of integrity of the organelle genome and the genetic interaction between MSH1 and homologs of HRR factors in the basal land plant P. patens.


Assuntos
Bryopsida/genética , Bryopsida/metabolismo , Instabilidade Genômica/genética , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Cloroplastos/metabolismo , DNA de Plantas/genética , Mitocôndrias/metabolismo , Plantas Geneticamente Modificadas/genética , Plantas Geneticamente Modificadas/metabolismo
15.
Am J Hum Genet ; 97(5): 761-8, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26522469

RESUMO

S-adenosylmethionine (SAM) is the predominant methyl group donor and has a large spectrum of target substrates. As such, it is essential for nearly all biological methylation reactions. SAM is synthesized by methionine adenosyltransferase from methionine and ATP in the cytoplasm and subsequently distributed throughout the different cellular compartments, including mitochondria, where methylation is mostly required for nucleic-acid modifications and respiratory-chain function. We report a syndrome in three families affected by reduced intra-mitochondrial methylation caused by recessive mutations in the gene encoding the only known mitochondrial SAM transporter, SLC25A26. Clinical findings ranged from neonatal mortality resulting from respiratory insufficiency and hydrops to childhood acute episodes of cardiopulmonary failure and slowly progressive muscle weakness. We show that SLC25A26 mutations cause various mitochondrial defects, including those affecting RNA stability, protein modification, mitochondrial translation, and the biosynthesis of CoQ10 and lipoic acid.


Assuntos
Sistemas de Transporte de Aminoácidos/genética , Proteínas de Ligação ao Cálcio/genética , Metilação de DNA , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia , Debilidade Muscular/genética , Mutação/genética , S-Adenosilmetionina/metabolismo , Sequência de Aminoácidos , Pré-Escolar , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Debilidade Muscular/patologia , Linhagem , Prognóstico , Estabilidade de RNA , Homologia de Sequência de Aminoácidos , Ácido Tióctico/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
16.
J Pediatr ; 193: 256-260, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29249525

RESUMO

The diagnosis of Barth syndrome is challenging owing to the wide phenotypic spectrum with allelic heterogeneity. Here we report 3 cases of Barth syndrome with phenotypic and allelic heterogeneity that were diagnosed by different approaches, including whole exome sequencing and final confirmation by reverse-transcription polymease chain reaction.


Assuntos
Síndrome de Barth/diagnóstico , Fatores de Transcrição/genética , Aciltransferases , Síndrome de Barth/genética , Humanos , Lactente , Recém-Nascido , Masculino , Mutação , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Sequenciamento do Exoma/métodos
17.
Am J Hum Genet ; 95(6): 708-20, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25434004

RESUMO

Respiratory chain deficiencies exhibit a wide variety of clinical phenotypes resulting from defective mitochondrial energy production through oxidative phosphorylation. These defects can be caused by either mutations in the mtDNA or mutations in nuclear genes coding for mitochondrial proteins. The underlying pathomechanisms can affect numerous pathways involved in mitochondrial physiology. By whole-exome and candidate gene sequencing, we identified 11 individuals from 9 families carrying compound heterozygous or homozygous mutations in GTPBP3, encoding the mitochondrial GTP-binding protein 3. Affected individuals from eight out of nine families presented with combined respiratory chain complex deficiencies in skeletal muscle. Mutations in GTPBP3 are associated with a severe mitochondrial translation defect, consistent with the predicted function of the protein in catalyzing the formation of 5-taurinomethyluridine (τm(5)U) in the anticodon wobble position of five mitochondrial tRNAs. All case subjects presented with lactic acidosis and nine developed hypertrophic cardiomyopathy. In contrast to individuals with mutations in MTO1, the protein product of which is predicted to participate in the generation of the same modification, most individuals with GTPBP3 mutations developed neurological symptoms and MRI involvement of thalamus, putamen, and brainstem resembling Leigh syndrome. Our study of a mitochondrial translation disorder points toward the importance of posttranscriptional modification of mitochondrial tRNAs for proper mitochondrial function.


Assuntos
Acidose Láctica/genética , Encefalopatias/genética , Cardiomiopatia Hipertrófica/genética , Proteínas de Ligação ao GTP/genética , Processamento de Proteína Pós-Traducional , Acidose Láctica/fisiopatologia , Sequência de Aminoácidos , Encéfalo/patologia , Encefalopatias/fisiopatologia , Cardiomiopatia Hipertrófica/fisiopatologia , Linhagem Celular , Criança , Pré-Escolar , Consanguinidade , Feminino , Fibroblastos , Proteínas de Ligação ao GTP/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Dados de Sequência Molecular , Mutação , Linhagem , Biossíntese de Proteínas , Interferência de RNA , RNA de Transferência/genética , RNA de Transferência/metabolismo , Alinhamento de Sequência
18.
J Hum Genet ; 62(5): 539-547, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28123175

RESUMO

Tafazzin, encoded by the TAZ gene, is a mitochondrial membrane-associated protein that remodels cardiolipin (CL), an important mitochondrial phospholipid. TAZ mutations are associated with Barth syndrome (BTHS). BTHS is an X-linked multisystemic disorder affecting usually male patients. Through sequence analysis of TAZ, we found one novel mutation c.39_60del p.(Pro14Alafs*19) by whole-exome sequencing and a reported missense mutation c.280C>T p.(Arg94Cys) by Sanger sequencing in two male patients (Pt1 and Pt2). Patient with c.280C>T mutation had dilated cardiomyopathy, while another patient with c.39_60del mutation had no feature of cardiomyopathy. A reported m.1555A>G homoplasmic variant was also identified in the patient having mutation c.39_60del by whole mitochondrial DNA sequencing method. This variant was not considered to be the main cause of mitochondrial dysfunction based on a cytoplasmic hybrid (cybrid) assay. Tafazzin expression was absent in both patient-derived fibroblast cells. Complementation of TAZ expression in fibroblasts from the patient with the novel mutation c.39_60del restored mitochondrial respiratory complex assembly. High-performance liquid chromatography-tandem mass spectrometry-based metabolic analysis revealed the decline of CL and the accumulation of monolysocardiolipin, indicating the loss of tafazzin activity. Owing to phenotypic variability, it is difficult to diagnose BTHS based on clinical features only. We conclude that genetic analysis should be performed to avoid underdiagnosis of this potentially life-threatening inborn error of metabolism.


Assuntos
Cardiomiopatias/genética , Mitocôndrias/genética , Doenças Mitocondriais/genética , Mutação/genética , Fatores de Transcrição/genética , Aciltransferases , Sequência de Bases , Criança , Pré-Escolar , Transporte de Elétrons/genética , Feminino , Genótipo , Humanos , Recém-Nascido , Masculino , Fenótipo , Gravidez , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
19.
J Inherit Metab Dis ; 40(5): 685-693, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28429146

RESUMO

Leigh syndrome (LS) is a progressive neurodegenerative disorder of infancy and early childhood. It is clinically diagnosed by typical manifestations and characteristic computed tomography (CT) or magnetic resonance imaging (MRI) studies. Unravelling mitochondrial respiratory chain (MRC) dysfunction behind LS is essential for deeper understanding of the disease, which may lead to the development of new therapies and cure. The aim of this study was to evaluate the clinical validity of various diagnostic tools in confirming MRC disorder in LS and Leigh-like syndrome (LL). The results of enzyme assays, molecular analysis, and cellular oxygen consumption rate (OCR) measurements were examined. Of 106 patients, 41 were biochemically and genetically verified, and 34 had reduced MRC activity but no causative mutations. Seven patients with normal MRC complex activities had mutations in the MT-ATP6 gene. Five further patients with normal activity in MRC were identified with causative mutations. Conversely, 12 out of 60 enzyme assays performed for genetically verified patients returned normal results. No biochemical or genetic background was confirmed for 19 patients. OCR was reduced in ten out of 19 patients with negative enzyme assay results. Inconsistent enzyme assay results between fibroblast and skeletal muscle biopsy samples were observed in 33% of 37 simultaneously analyzed cases. These data suggest that highest diagnostic rate is reached using a combined enzymatic and genetic approach, analyzing more than one type of biological materials where suitable. Microscale oxygraphy detected MRC impairment in 50% cases with no defect in MRC complex activities.


Assuntos
Doença de Leigh/diagnóstico , Adolescente , Adulto , Povo Asiático , Criança , Transporte de Elétrons/genética , Feminino , Fibroblastos/fisiologia , Humanos , Doença de Leigh/genética , Masculino , Mitocôndrias/genética , ATPases Mitocondriais Próton-Translocadoras/genética , Músculo Esquelético/fisiologia , Mutação/genética , Consumo de Oxigênio/genética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA