Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Acta Crystallogr E Crystallogr Commun ; 71(Pt 7): 816-20, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26279875

RESUMO

The title compound, C16H10, crystallizes with four unique mol-ecules, designated 1-4, in the asymmetric unit of the monoclinic unit cell. None of the mol-ecules is planar, with the benzene rings of mol-ecules 1-4 inclined to one another at angles of 42.41 (4), 24.07 (6), 42.59 (4) and 46.88 (4)°, respectively. In the crystal, weak C-H⋯π(ring) interactions, augmented by even weaker C C-H⋯π(alkyne) contacts, generate a three-dimensional network structure with inter-linked columns of mol-ecules formed along the c-axis direction.

2.
Eur J Paediatr Neurol ; 19(5): 525-32, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26008862

RESUMO

BACKGROUND: Two siblings with hypertrophic cardiomyopathy and brain atrophy were diagnosed with Complex I deficiency based on low enzyme activity in muscle and high lactate/pyruvate ratio in fibroblasts. METHODS: Whole exome sequencing results of fibroblast gDNA from one sibling was narrowed down to 190 SNPs or In/Dels in 185 candidate genes by selecting non-synonymous coding sequence base pair changes that were not present in the SNP database. RESULTS: Two compound heterozygous mutations were identified in both siblings in NDUFV2, encoding the 24 kDa subunit of Complex I. The intronic mutation (c.IVS2 + 1delGTAA) is disease causing and has been reported before. The other mutation is novel (c.669_670insG, p.Ser224Valfs*3) and predicted to cause a pathogenic frameshift in the protein. Subsequent investigation of 10 probands with complex I deficiency from different families revealed homozygosity for the intronic c.IVS2 + 1delGTAA mutation in a second, consanguineous family. In this family three of five siblings were affected. Interestingly, they presented with Leigh syndrome but no cardiac involvement. The same genotype had been reported previously in a two families but presenting with hypertrophic cardiomyopathy, trunk hypotonia and encephalopathy. CONCLUSION: We have identified NDUFV2 mutations in two families with Complex I deficiency, including a novel mutation. The diagnosis of Leigh syndrome expands the clinical phenotypes associated with the c.IVS2 + 1delGTAA mutation in this gene.


Assuntos
Exoma , Doença de Leigh/genética , Mutação , NADH Desidrogenase/genética , Exoma/genética , Feminino , Humanos , Linhagem , Fenótipo , Irmãos
4.
Acta Crystallogr Sect E Struct Rep Online ; 70(Pt 7): 9-13, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25161495

RESUMO

The title compounds, [Co2(C8H11BrO3)(CO)6], (1), and [Co2(C12H16Br2O4)(CO)6], (2), result from the replacement of two carbonyl ligands from dicobalt octa-carbonyl by the alkynes 4-hy-droxy-but-2-ynyl 2-bromo-2-methyl-propano-ate and but-2-yne-1,4-diyl bis-(2-bromo-2-methyl-propano-ate), respectively. Both mol-ecules have classic tetra-hedral C2Co2 cluster cores with the Co(II) atoms in a highly distorted octa-hedral coordination geometry. The alkyne ligands both adopt a cis-bent conformation on coordination. In the crystal structure of (1), classical O-H⋯O and non-classical C-H⋯O contacts form inversion dimers. These combine with weak O⋯O and Br⋯O contacts to stack the mol-ecules into inter-connected columns along the b-axis direction. C-H⋯O and C-H⋯Br contacts stabilize the packing for (2), and a weak Br⋯O contact is also observed. Inter-connected columns of mol-ecules again form along the b-axis direction.

5.
J Mater Sci Mater Med ; 25(12): 2743-56, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25085242

RESUMO

An amine-functionalized succinyl chitosan and an oxidized dextran were synthesized and mixed in aqueous solution to form an in situ chitosan/dextran injectable, surgical hydrogel for adhesion prevention. Rheological characterization showed that the rate of gelation and moduli were tunable based on amine and aldehyde levels, as well as polymer concentrations. The CD hydrogels have been shown to be effective post-operative aids in prevention of adhesions in ear, nose, and throat surgeries and abdominal surgeries in vivo. In vitro biocompatibility testing was performed on CD hydrogels containing one of two oxidized dextrans, an 80 % oxidized (CD-100) or 25 % (CD-25) oxidized dextran. However, the CD-100 hydrogel showed moderate cytotoxicity in vitro to Vero cells. SC component of the CD hydrogel, however, showed no cytotoxic effect. In order to increase the biocompatibility of the hydrogel, a lower aldehyde level hydrogel was developed. CD-25 was found to be non-cytotoxic to L929 fibroblasts. The in vivo pro-inflammatory response of the CD-25 hydrogel, after intraperitoneal injection in BALB/c mice, was also determined by measuring serum TNF-α levels and by histological analysis of tissues. TNF-α levels were similar in mice injected with CD-25 hydrogel as compared to the negative saline injected control; and were significantly different (P < 0.05) as compared to the positive, lipopolysaccharide, injected control. Histological examination revealed no inflammation seen in CD hydrogel injected mice. The results of these in vitro and in vivo studies demonstrate the biocompatibility of the CD hydrogel as a post-operative aid for adhesion prevention.


Assuntos
Curativos Hidrocoloides , Materiais Biocompatíveis/síntese química , Sobrevivência Celular/fisiologia , Quitosana/química , Dextranos/química , Hidrogéis/química , Aderências Teciduais/prevenção & controle , Animais , Materiais Biocompatíveis/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Quitosana/farmacologia , Chlorocebus aethiops , Dextranos/farmacologia , Módulo de Elasticidade , Desenho de Equipamento , Análise de Falha de Equipamento , Dureza , Humanos , Hidrogéis/farmacologia , Teste de Materiais , Células Vero
6.
Brain ; 137(Pt 2): 366-79, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24334290

RESUMO

Patients with nonketotic hyperglycinemia and deficient glycine cleavage enzyme activity, but without mutations in AMT, GLDC or GCSH, the genes encoding its constituent proteins, constitute a clinical group which we call 'variant nonketotic hyperglycinemia'. We hypothesize that in some patients the aetiology involves genetic mutations that result in a deficiency of the cofactor lipoate, and sequenced genes involved in lipoate synthesis and iron-sulphur cluster biogenesis. Of 11 individuals identified with variant nonketotic hyperglycinemia, we were able to determine the genetic aetiology in eight patients and delineate the clinical and biochemical phenotypes. Mutations were identified in the genes for lipoate synthase (LIAS), BolA type 3 (BOLA3), and a novel gene glutaredoxin 5 (GLRX5). Patients with GLRX5-associated variant nonketotic hyperglycinemia had normal development with childhood-onset spastic paraplegia, spinal lesion, and optic atrophy. Clinical features of BOLA3-associated variant nonketotic hyperglycinemia include severe neurodegeneration after a period of normal development. Additional features include leukodystrophy, cardiomyopathy and optic atrophy. Patients with lipoate synthase-deficient variant nonketotic hyperglycinemia varied in severity from mild static encephalopathy to Leigh disease and cortical involvement. All patients had high serum and borderline elevated cerebrospinal fluid glycine and cerebrospinal fluid:plasma glycine ratio, and deficient glycine cleavage enzyme activity. They had low pyruvate dehydrogenase enzyme activity but most did not have lactic acidosis. Patients were deficient in lipoylation of mitochondrial proteins. There were minimal and inconsistent changes in cellular iron handling, and respiratory chain activity was unaffected. Identified mutations were phylogenetically conserved, and transfection with native genes corrected the biochemical deficiency proving pathogenicity. Treatments of cells with lipoate and with mitochondrially-targeted lipoate were unsuccessful at correcting the deficiency. The recognition of variant nonketotic hyperglycinemia is important for physicians evaluating patients with abnormalities in glycine as this will affect the genetic causation and genetic counselling, and provide prognostic information on the expected phenotypic course.


Assuntos
Variação Genética/genética , Glutarredoxinas/genética , Hiperglicinemia não Cetótica/genética , Mutação/genética , Proteínas/genética , Sulfurtransferases/genética , Atrofia , Criança , Pré-Escolar , Evolução Fatal , Feminino , Glutarredoxinas/química , Humanos , Hiperglicinemia não Cetótica/diagnóstico , Hiperglicinemia não Cetótica/patologia , Lactente , Masculino , Proteínas Mitocondriais , Proteínas/química , Índice de Gravidade de Doença , Sulfurtransferases/química
7.
PLoS One ; 8(3): e57195, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469184

RESUMO

Alaskan Husky Encephalopathy (AHE) has been previously proposed as a mitochondrial encephalopathy based on neuropathological similarities with human Leigh Syndrome (LS). We studied 11 Alaskan Husky dogs with AHE, but found no abnormalities in respiratory chain enzyme activities in muscle and liver, or mutations in mitochondrial or nuclear genes that cause LS in people. A genome wide association study was performed using eight of the affected dogs and 20 related but unaffected control AHs using the Illumina canine HD array. SLC19A3 was identified as a positional candidate gene. This gene controls the uptake of thiamine in the CNS via expression of the thiamine transporter protein THTR2. Dogs have two copies of this gene located within the candidate interval (SLC19A3.2 - 43.36-43.38 Mb and SLC19A3.1 - 43.411-43.419 Mb) on chromosome 25. Expression analysis in a normal dog revealed that one of the paralogs, SLC19A3.1, was expressed in the brain and spinal cord while the other was not. Subsequent exon sequencing of SLC19A3.1 revealed a 4bp insertion and SNP in the second exon that is predicted to result in a functional protein truncation of 279 amino acids (c.624 insTTGC, c.625 C>A). All dogs with AHE were homozygous for this mutation, 15/41 healthy AH control dogs were heterozygous carriers while 26/41 normal healthy AH dogs were wild type. Furthermore, this mutation was not detected in another 187 dogs of different breeds. These results suggest that this mutation in SLC19A3.1, encoding a thiamine transporter protein, plays a critical role in the pathogenesis of AHE.


Assuntos
Encefalopatias Metabólicas/genética , Encefalopatias Metabólicas/veterinária , Doenças do Cão/genética , Proteínas de Membrana Transportadoras/genética , Mutação , Animais , Sequência de Bases , Transporte Biológico/genética , Encefalopatias Metabólicas/metabolismo , Encefalopatias Metabólicas/patologia , Doenças do Cão/metabolismo , Doenças do Cão/patologia , Cães , Éxons , Feminino , Loci Gênicos , Estudo de Associação Genômica Ampla , Heterozigoto , Homozigoto , Humanos , Doença de Leigh/genética , Doença de Leigh/metabolismo , Doença de Leigh/patologia , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Tiamina/metabolismo
8.
Mol Genet Metab ; 108(1): 40-50, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23266196

RESUMO

We report on two families with Sengers syndrome and mutations in the acylglycerol kinase gene (AGK). In the first family, two brothers presented with vascular strokes, lactic acidosis, cardiomyopathy and cataracts, abnormal muscle cell histopathology and mitochondrial function. One proband had very abnormal mitochondria with citrate synthase crystals visible in electron micrographs, associated with markedly high citrate synthase activity. Exome sequencing was used to identify mutations in the AGK gene in the index patient. Targeted sequencing confirmed the same homozygous mutation (c.3G>A, p.M1I) in the brother. The second family had four affected members, of which we examined two. They also presented with similar clinical symptoms, but no strokes. Postmortem heart and skeletal muscle tissues showed low complex I, III and IV activities in the heart, but normal in the muscle. Skin fibroblasts showed elevated lactate/pyruvate ratios and low complex I+III activity. Targeted sequencing led to identification of a homozygous c.979A>T, p.K327* mutation. AGK is located in the mitochondria and phosphorylates monoacylglycerol and diacylglycerol to lysophosphatidic acid and phosphatidic acid. Disruption of these signaling molecules affects the mitochondria's response to superoxide radicals, resulting in oxidative damage to mitochondrial DNA, lipids and proteins, and stimulation of cellular detoxification pathways. High levels of manganese superoxide dismutase protein were detected in all four affected individuals, consistent with increased free radical damage. Phosphatidic acid is also involved in the synthesis of phospholipids and its loss will result in changes to the lipid composition of the inner mitochondrial membrane. These effects manifest as cataract formation in the eye, respiratory chain dysfunction and cardiac hypertrophy in heart tissue. These two pedigrees confirm that mutation of AGK is responsible for the severe neonatal presentation of Sengers syndrome. The identification of citrate synthase precipitates by electron microscopy and the presence of vascular strokes in two siblings may expand the cellular and clinical phenotype of this disease.


Assuntos
Cardiomiopatias/enzimologia , Catarata/enzimologia , Citrato (si)-Sintase/química , Mitocôndrias/enzimologia , Mutação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Sequência de Bases , Pré-Escolar , Cristalização , Primers do DNA , Feminino , Humanos , Lactente , Masculino , Linhagem
9.
Mitochondrion ; 12(6): 623-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23022402

RESUMO

We report the design and implementation of the first phase 3 trial of CoenzymeQ10 (CoQ10) in children with genetic mitochondrial diseases. A novel, rigorous set of eligibility criteria was established. The trial, which remains open to recruitment, continues to address multiple challenges to the recruitment of patients, including widely condoned empiric use of CoQ10 by individuals with proven or suspected mitochondrial disease and skepticism among professional and lay mitochondrial disease communities about participating in placebo-controlled trials. These attitudes represent significant barriers to the ethical and scientific evaluation--and ultimate approval--of nutritional and pharmacological therapies for patients with life-threatening inborn errors of energy metabolism.


Assuntos
Doenças Mitocondriais/congênito , Doenças Mitocondriais/tratamento farmacológico , Ubiquinona/análogos & derivados , Pesquisa Biomédica , Humanos , Projetos de Pesquisa , Ubiquinona/uso terapêutico
10.
Biochem J ; 441(1): 275-83, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21880015

RESUMO

LRPPRC (leucine-rich pentatricopeptide repeat-containing) has been shown to be essential for the maturation of COX (cytochrome c oxidase), possibly by stabilizing RNA transcripts of COXI, COXII and COXIII genes encoded in mtDNA (mitochondrial DNA). We established a mouse 'gene-trap' model using ES cells (embryonic stem cells) in which the C-terminus of LRPPRC has been replaced with a ß-geo construct. Mice homozygous for this modification were found to be subject to embryonic lethality, with death before 12.5 dpc (days post-coitum). Biochemical analysis of MEFs (mouse embryonic fibroblasts) isolated from homozygous mutants showed a major decrease in COX activity, with slight reductions in other respiratory chain complexes with mtDNA encoded components. Constructs of LRPPRC containing different numbers of PPRs (pentatricopeptide repeats) were expressed as recombinant proteins and tested for their ability to bind to the COXI mRNA transcript. Full binding required the first 19 PPR motifs. A specific segment of COXI mRNA was identified as the binding target for LRPPRC, encoded by mouse mtDNA nucleotides 5961-6020. These data strongly suggest that LRPPRC is involved in the maturation of COX, and is involved in stabilizing of mitochondrial mRNAs encoding COX transcripts.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Mitocôndrias/metabolismo , Proteínas de Neoplasias/metabolismo , RNA/metabolismo , Animais , Linhagem Celular , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/classificação , Complexo IV da Cadeia de Transporte de Elétrons/genética , Embrião de Mamíferos , Células-Tronco Embrionárias , Fibroblastos/citologia , Fibroblastos/metabolismo , Genótipo , Camundongos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Mutação , Proteínas de Neoplasias/genética , Receptores Ativados por Proliferador de Peroxissomo/genética , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Ligação Proteica , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , beta-Galactosidase/metabolismo
11.
J Pharm Pharm Sci ; 14(3): 438-49, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22202226

RESUMO

Leigh syndrome French Canadian (LSFC) is a recessive disease caused by mutations in the LRPPRC gene (leucine-rich pentatricopeptide repeat containing protein). These mutations induce a cytochrome c oxidase (COX) deficiency resulting in episodes of acute acidotic crisis that will often lead to death. There is no effective treatment. Methylene blue (MB) is a redox dye that increases COX content and activity in vitro and in vivo suggesting that MB could prevent and treat LSFC. In this study, the protective effect of low-concentration MB was tested on two LSFC cell lines, including LSFC-F1, homozygous for the mutation A354V, and LSFC-F2 a compound heterozygous for the mutations A354V and C12775STOP. MB effect on metabolic activity was assessed on both LSFC cells in stable and acidotic conditions. For LSFC-F1, results showed that metabolic activity drastically decline after 96 hours in both conditions but not LSFC-F2 and normal cells. MB completely prevents the decrease of metabolic activity in LSFC-F1. Intracellular ATP content was also measured in both culture media. After 96 hours in acidotic medium, ATP content was almost completely depleted for both LSFC cells. Interestingly, MB completely restores ATP content in LSFC-F1 and LSFC-F2 cells. Finally, MB strongly improves the survival of both LSFC cells.


Assuntos
Deficiência de Citocromo-c Oxidase/metabolismo , Metabolismo Energético/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Doença de Leigh/metabolismo , Azul de Metileno/farmacologia , Pele/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Deficiência de Citocromo-c Oxidase/genética , Deficiência de Citocromo-c Oxidase/patologia , Relação Dose-Resposta a Droga , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Heterozigoto , Homozigoto , Humanos , Concentração de Íons de Hidrogênio , Doença de Leigh/genética , Doença de Leigh/patologia , Mutação , Proteínas de Neoplasias/genética , Pele/metabolismo , Pele/patologia , Fatores de Tempo
12.
Cancer Cell ; 20(5): 674-88, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22094260

RESUMO

To identify FDA-approved agents targeting leukemic cells, we performed a chemical screen on two human leukemic cell lines and identified the antimicrobial tigecycline. A genome-wide screen in yeast identified mitochondrial translation inhibition as the mechanism of tigecycline-mediated lethality. Tigecycline selectively killed leukemia stem and progenitor cells compared to their normal counterparts and also showed antileukemic activity in mouse models of human leukemia. ShRNA-mediated knockdown of EF-Tu mitochondrial translation factor in leukemic cells reproduced the antileukemia activity of tigecycline. These effects were derivative of mitochondrial biogenesis that, together with an increased basal oxygen consumption, proved to be enhanced in AML versus normal hematopoietic cells and were also important for their difference in tigecycline sensitivity.


Assuntos
Antineoplásicos/farmacologia , Genes Mitocondriais , Leucemia/tratamento farmacológico , Minociclina/análogos & derivados , Mitocôndrias/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Minociclina/farmacologia , Proteínas Mitocondriais/genética , Fator Tu de Elongação de Peptídeos/genética , RNA Interferente Pequeno , Saccharomyces cerevisiae/efeitos dos fármacos , Tigeciclina
13.
Am J Hum Genet ; 89(4): 486-95, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-21944046

RESUMO

Severe combined deficiency of the 2-oxoacid dehydrogenases, associated with a defect in lipoate synthesis and accompanied by defects in complexes I, II, and III of the mitochondrial respiratory chain, is a rare autosomal recessive syndrome with no obvious causative gene defect. A candidate locus for this syndrome was mapped to chromosomal region 2p14 by microcell-mediated chromosome transfer in two unrelated families. Unexpectedly, analysis of genes in this area identified mutations in two different genes, both of which are involved in [Fe-S] cluster biogenesis. A homozygous missense mutation, c.545G>A, near the splice donor of exon 6 in NFU1 predicting a p.Arg182Gln substitution was found in one of the families. The mutation results in abnormal mRNA splicing of exon 6, and no mature protein could be detected in fibroblast mitochondria. A single base-pair duplication c.123dupA was identified in BOLA3 in the second family, causing a frame shift that produces a premature stop codon (p.Glu42Argfs(∗)13). Transduction of fibroblast lines with retroviral vectors expressing the mitochondrial, but not the cytosolic isoform of NFU1 and with isoform 1, but not isoform 2 of BOLA3 restored both respiratory chain function and oxoacid dehydrogenase complexes. NFU1 was previously proposed to be an alternative scaffold to ISCU for the biogenesis of [Fe-S] centers in mitochondria, and the function of BOLA3 was previously unknown. Our results demonstrate that both play essential roles in the production of [Fe-S] centers for the normal maturation of lipoate-containing 2-oxoacid dehydrogenases, and for the assembly of the respiratory chain complexes.


Assuntos
Proteínas de Transporte/genética , Mutação , Oxirredutases/metabolismo , Proteínas/genética , Citosol/metabolismo , Transporte de Elétrons , Éxons , Saúde da Família , Feminino , Fibroblastos/metabolismo , Homozigoto , Humanos , Proteínas Ferro-Enxofre/metabolismo , Masculino , Mitocôndrias/metabolismo , Proteínas Mitocondriais , Mutação de Sentido Incorreto
14.
J Med Genet ; 48(3): 183-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21266382

RESUMO

BACKGROUND: The natural history of all known patients with French-Canadian Leigh disease (Saguenay-Lac-St-Jean cytochrome c oxidase deficiency, MIM220111, SLSJ-COX), the largest known cohort of patients with a genetically homogeneous, nuclear encoded congenital lactic acidosis, was studied. RESULTS: 55 of 56 patients were homozygous for the A354V mutation in LRPPRC. One was a genetic compound (A354V/C1277Xdel8). Clinical features included developmental delay, failure to thrive, characteristic facial appearance and, in 90% of patients, acute crises that have not previously been detailed, either metabolic (fulminant lactic acidosis) and/or neurological (Leigh syndrome and/or stroke-like episodes). Survival ranged from 5 days to >30 years. 46/56 patients (82%) died, at a median age of 1.6 years. Of 73 crises, 38 (52%) were fatal. The immediate causes of death were multiple organ failure and/or Leigh disease. Major predictors of mortality during crises (p<0.005) were hyperglycaemia, hepatic cytolysis, and altered consciousness at admission. Compared to a group of SURF1-deficient Leigh syndrome patients assembled from the literature, SLSJ-COX is distinct by the occurrence of metabolic crises, leading to earlier and higher mortality (p=0.001). CONCLUSION: SLSJ-COX is clinically distinct, with acute fatal acidotic crises on a backdrop of chronic moderate developmental delay and hyperlactataemia. Leigh syndrome is common. Stroke-like episodes can occur. The Leigh syndrome of SLSJ-COX differs from that of SURF1-related COX deficiency. SLSJ-COX has a different spectrum of associated abnormalities, acidotic crises being particularly suggestive of LRPPRC related Leigh syndrome. Even among A354V homozygotes, pronounced differences in survival and severity occur, showing that other genetic and/or environmental factors can influence outcome.


Assuntos
Deficiência de Citocromo-c Oxidase/genética , Doença de Leigh/genética , Mutação , Proteínas de Neoplasias/genética , Acidose Láctica/genética , Adolescente , Criança , Pré-Escolar , Feminino , Seguimentos , Homozigoto , Humanos , Lactente , Doença de Leigh/metabolismo , Modelos Logísticos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas de Neoplasias/metabolismo , Fenótipo , Estudos Retrospectivos , Adulto Jovem
15.
Mitochondrion ; 11(1): 191-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20920610

RESUMO

Mutations in the TMEM70 gene are responsible for a familial form of complex V deficiency presenting with 3-methylglutaconic aciduria, lactic acidosis, cardiomyopathy and mitochondrial myopathy. Here we present a case of TMEM70 deficiency due to compound heterozygous mutations, who displayed abnormal mitochondria with whorled cristae in muscle. Immunogold electron microscopy and tomography shows for the first time that nucleoid clusters of mtDNA are disrupted in the abnormal mitochondria, with both nucleoids and mitochondrial respiratory chain complexes confined to the outer rings of the whorls. This could explain the differential effects on the expression and assembly of complex V in different tissues.


Assuntos
DNA Mitocondrial/genética , Heterozigoto , Proteínas de Membrana/deficiência , Mitocôndrias Musculares/ultraestrutura , Mitocôndrias/ultraestrutura , Doenças Mitocondriais/genética , Proteínas Mitocondriais/deficiência , Mutação , Partículas Submitocôndricas/ultraestrutura , Acidose Láctica/genética , Acidose Láctica/metabolismo , Acidose Láctica/patologia , Adulto , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Feminino , Fibroblastos/metabolismo , Humanos , Recém-Nascido , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias Musculares/genética , Mitocôndrias Musculares/metabolismo , Doenças Mitocondriais/patologia , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Partículas Submitocôndricas/genética , Tomografia
16.
Mitochondrion ; 11(2): 264-72, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21050896

RESUMO

Cytochrome c oxidase (COX) activity reflects the expressed level of respiratory chain complexes, mtDNA levels, titer and mass of mitochondria. Activity is also indicative of the overall fitness of mt-transcription factors and the import, transcription and translation of mt-proteins. We have developed a high-throughput assay to measure COX activity using live cells to screen chemical libraries for compounds capable of increasing COX activity. These libraries have revealed four examples which elevated the activities of COX in NIH-3T3 fibroblasts and in fibroblasts from patients with COX defects independent of the peroxisome proliferator activated receptor family.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/efeitos dos fármacos , Animais , Western Blotting , Linhagem Celular Transformada , Corantes , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Ativação Enzimática , Fibroblastos/enzimologia , Humanos , Camundongos , Células NIH 3T3
17.
Biochem Biophys Res Commun ; 402(2): 443-7, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20965148

RESUMO

The A to G transition mutation at position 3260 of the mitochondrial genome is usually associated with cardiomyopathy and myopathy. One Japanese kindred reported the phenotype of mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes (MELAS syndrome) in association with the A3260G mtDNA mutation. We describe the first Caucasian cases of MELAS syndrome associated with the A3260G mutation. Furthermore, this mutation was associated with exercise-induced rhabdomyolysis, hearing loss, seizures, cardiomyopathy, and autism in the large kindred. We conclude that the A3260G mtDNA mutation is associated with wide phenotypic heterogeneity with MELAS and other "classical" mitochondrial phenotypes being manifestations.


Assuntos
Transtorno Autístico/genética , Cardiomiopatias/genética , DNA Mitocondrial/genética , Genes Mitocondriais , Síndrome MELAS/genética , Rabdomiólise/genética , Adulto , Criança , Pré-Escolar , Feminino , Predisposição Genética para Doença , Humanos , Síndrome MELAS/patologia , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/patologia , Linhagem , Mutação Puntual , Adulto Jovem
18.
Biochem Biophys Res Commun ; 399(3): 331-5, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20637729

RESUMO

Mitochondrial dysfunction is involved in the underlying pathology of Parkinson's Disease (PD). PINK1 deficiency, which gives rise to familial early-onset PD, is associated with this dysfunction as well as increased oxidative stress. We have established primary fibroblast cell lines from two patients with PD who carry mutations in the PINK1 gene. The phosphorylation of Akt is abrogated in the presence of oxidative stressors in the complete absence of PINK1 suggesting enhanced apoptotic signalling. We have found an imbalance between the production of reactive oxygen species where the capacity of the cell to remove these toxins by anti-oxidative enzymes is greatly reduced. The expression levels of the anti-oxidant enzymes glutathione peroxidase-1, MnSOD, peroxiredoxin-3 and thioredoxin-2 were diminished. The p66(Shc) adaptor protein has recently been identified to become activated by oxidative stress by phosphorylation at residue Ser36 which then translocates to the mitochondrial inner membrane space. The phosphorylation of p66(Shc) at Ser36 is significantly increased in PINK1 deficient cell lines under normal tissue culture conditions, further still in the presence of compounds which elicit oxidative stress. The stable transfection of PINK1 in the fibroblasts which display the null phenotype ameliorates the hyper-phosphorylation of p66(Shc).


Assuntos
Estresse Oxidativo , Doença de Parkinson/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Linhagem Celular , Fibroblastos/metabolismo , Glutationa Peroxidase/metabolismo , Humanos , Peroxirredoxinas/metabolismo , Fosforilação , Proteínas Quinases/genética , Serina/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Superóxido Dismutase/metabolismo , Tiorredoxinas/metabolismo , Glutationa Peroxidase GPX1
19.
Mol Genet Metab ; 98(4): 378-82, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19699667

RESUMO

We report here the identification of a patient with muscle-specific glycogen synthase deficiency. The 8-year-old patient showed no prior signs of distress before collapsing during a bout of exercise, resulting in death. Initial post-mortem analysis of tissues suggested death was due to metabolic complications of mitochondrial myopathy, but upon further examination it was found that the anomalies were indicative of mitochondrial proliferation and oxidative compensation. A homozygous two base pair deletion was identified in exon 2 of GYS1, and the parents and sibling were confirmed as heterozygous carriers of the deletion. This case highlights the importance of differentiating between mitochondrial compensatory phenomena and true mitochondrial disease, and suggests that GYS1 deficiency could be a common cause of sudden cardiac death in children. Children with abnormal cardiac responses to increased workloads as well as those with defined myocardial disease should therefore be tested for GYS1 deficiency.


Assuntos
Morte Súbita Cardíaca/etiologia , Fibroblastos/enzimologia , Fibroblastos/patologia , Glicogênio Fosforilase Muscular/genética , Mutação/genética , Pele/patologia , Sequência de Bases , Extratos Celulares , Criança , Análise Mutacional de DNA , Evolução Fatal , Feminino , Humanos , Lactatos/metabolismo , Masculino , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , Dados de Sequência Molecular , Linhagem , Sonicação , Coloração e Rotulagem
20.
Mol Genet Metab ; 98(4): 349-55, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19640754

RESUMO

Fibroblast cell lines are frequently used to diagnose genetic mitochondrial defects in children. The effect of enzyme deficiency on overall flux rate through metabolic pathways is, however, not generally considered. We have transposed an experimental paradigm that was developed for isolated perfused organs using (13)C-labeled substrates and (13)C-isotopomer analysis to probe pyruvate mitochondrial metabolism in cultured human fibroblast cell lines with normal or genetically mutant pyruvate decarboxylation (PDC) or carboxylation (PC) activity. Cells were incubated with 1mM [U-(13)C]pyruvate, and the (13)C-molar percent enrichment (MPE) of intracellular pyruvate, citrate, malate (as a surrogate of oxaloacetate) and aspartate was assessed by mass spectrometry. We estimated various flux ratios relevant to metabolic pathways involved in energy production, namely pyruvate formation, PDC, PC, and citrate recycling in the citric acid cycle (CAC). In all cell lines, exogenous pyruvate was predominately decarboxylated (PC/PDC ratios 0.01-0.3). PC-deficient cell lines displayed an expected negligible contribution of PC flux to oxaloacetate formation for citrate synthesis (PC/CS), which was associated with a greater contribution of PDC to acetyl-CoA formation (PDC/CS), and greater recycling of (13)C-labeled citrate into the CAC. In PDH-deficient cell lines, metabolic flux alterations were most apparent in cells with more than 50% reduction in enzyme activity. This led to an unexpected lower PC/CS flux ratio, while the PDC/CS flux ratio was unchanged. These data illustrate the usefulness of this approach in identifying unexpected metabolic consequences of genetic defects related to pyruvate metabolism.


Assuntos
Fibroblastos/metabolismo , Espectrometria de Massas/métodos , Mutação/genética , Ácido Pirúvico/metabolismo , Ácido Aspártico/metabolismo , Isótopos de Carbono , Linhagem Celular , Cromatografia Líquida , Ácido Cítrico/metabolismo , Ciclo do Ácido Cítrico/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Marcação por Isótopo , Malatos/metabolismo , Masculino , Ácido Pirúvico/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA