Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Inherit Metab Dis ; 42(5): 809-817, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31177572

RESUMO

The first step in branched-chain amino acid (BCAA) catabolism is catalyzed by the two BCAA transferase isoenzymes, cytoplasmic branched-chain amino acid transferase (BCAT) 1, and mitochondrial BCAT2. Defects in the second step of BCAA catabolism cause maple syrup urine disease (MSUD), a condition which has been far more extensively investigated. Here, we studied the consequences of BCAT2 deficiency, an ultra-rare condition in humans. We present genetic, clinical, and functional data in five individuals from four different families with homozygous or compound heterozygous BCAT2 mutations which were all detected following abnormal biochemical profile results or familial mutation segregation studies. We demonstrate that BCAT2 deficiency has a recognizable biochemical profile with raised plasma BCAAs and, in contrast with MSUD, low-normal branched-chain keto acids (BCKAs) with undetectable l-allo-isoleucine. Interestingly, unlike in MSUD, none of the individuals with BCAT2 deficiency developed acute encephalopathy even with exceptionally high BCAA levels. We observed wide-ranging clinical phenotypes in individuals with BCAT2 deficiency. While one adult was apparently asymptomatic, three individuals had presented with developmental delay and autistic features. We show that the biochemical characteristics of BCAT2 deficiency may be amenable to protein-restricted diet and that early treatment may improve outcome in affected individuals. BCAT2 deficiency is an inborn error of BCAA catabolism. At present, it is unclear whether developmental delay and autism are parts of the variable phenotypic spectrum of this condition or coincidental. Further studies will be required to explore this.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/genética , Aminoácidos de Cadeia Ramificada/sangue , Encéfalo/patologia , Mitocôndrias/patologia , Proteínas da Gravidez/deficiência , Transaminases/deficiência , Adolescente , Adulto , Encéfalo/diagnóstico por imagem , Criança , Pré-Escolar , Feminino , Homozigoto , Humanos , Imageamento por Ressonância Magnética , Masculino , Antígenos de Histocompatibilidade Menor/genética , Mutação , Fenótipo , Proteínas da Gravidez/genética , Transaminases/genética
2.
Hum Genet ; 134(8): 869-79, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26008905

RESUMO

Succinate dehydrogenase (SDH) is a crucial metabolic enzyme complex that is involved in ATP production, playing roles in both the tricarboxylic cycle and the mitochondrial respiratory chain (complex II). Isolated complex II deficiency is one of the rarest oxidative phosphorylation disorders with mutations described in three structural subunits and one of the assembly factors; just one case is attributed to recessively inherited SDHD mutations. We report the pathological, biochemical, histochemical and molecular genetic investigations of a male neonate who had left ventricular hypertrophy detected on antenatal scan and died on day one of life. Subsequent postmortem examination confirmed hypertrophic cardiomyopathy with left ventricular non-compaction. Biochemical analysis of his skeletal muscle biopsy revealed evidence of a severe isolated complex II deficiency and candidate gene sequencing revealed a novel homozygous c.275A>G, p.(Asp92Gly) SDHD mutation which was shown to be recessively inherited through segregation studies. The affected amino acid has been reported as a Dutch founder mutation p.(Asp92Tyr) in families with hereditary head and neck paraganglioma. By introducing both mutations into Saccharomyces cerevisiae, we were able to confirm that the p.(Asp92Gly) mutation causes a more severe oxidative growth phenotype than the p.(Asp92Tyr) mutant, and provides functional evidence to support the pathogenicity of the patient's SDHD mutation. This is only the second case of mitochondrial complex II deficiency due to inherited SDHD mutations and highlights the importance of sequencing all SDH genes in patients with biochemical and histochemical evidence of isolated mitochondrial complex II deficiency.


Assuntos
Cardiomiopatia Hipertrófica Familiar/genética , Genes Recessivos , Cardiopatias Congênitas/genética , Homozigoto , Proteínas Mitocondriais/genética , Mutação de Sentido Incorreto , Succinato Desidrogenase/genética , Substituição de Aminoácidos , Cardiomiopatia Hipertrófica Familiar/enzimologia , Ciclo do Ácido Cítrico/genética , Cardiopatias Congênitas/enzimologia , Humanos , Recém-Nascido , Masculino
3.
Hum Mutat ; 35(1): 86-95, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24123825

RESUMO

Multiple acyl-CoA dehydrogenation deficiency is a disorder of fatty acid and amino acid oxidation caused by defects of electron transfer flavoprotein (ETF) or its dehydrogenase (ETFDH). A clear relationship between genotype and phenotype makes genotyping of patients important not only diagnostically but also for prognosis and for assessment of treatment. In the present study, we show that a predicted benign ETFDH missense variation (c.158A>G/p.Lys53Arg) in exon 2 causes exon skipping and degradation of ETFDH protein in patient samples. Using splicing reporter minigenes and RNA pull-down of nuclear proteins, we show that the c.158A>G variation increases the strength of a preexisting exonic splicing silencer (ESS) motif UAGGGA. This ESS motif binds splice inhibitory hnRNP A1, hnRNP A2/B1, and hnRNP H proteins. Binding of these inhibitory proteins prevents binding of the positive splicing regulatory SRSF1 and SRSF5 proteins to nearby and overlapping exonic splicing enhancer elements and this causes exon skipping. We further suggest that binding of hnRNP proteins to UAGGGA is increased by triggering synergistic hnRNP H binding to GGG triplets located upstream and downsteam of the UAGGGA motif. A number of disease-causing exonic elements that induce exon skipping in other genes have a similar architecture as the one in ETFDH exon 2.


Assuntos
Adenosina/metabolismo , Flavoproteínas Transferidoras de Elétrons/genética , Flavoproteínas Transferidoras de Elétrons/metabolismo , Guanina/metabolismo , Proteínas Ferro-Enxofre/genética , Proteínas Ferro-Enxofre/metabolismo , Deficiência Múltipla de Acil Coenzima A Desidrogenase/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Splicing de RNA , Motivos de Aminoácidos , Cadáver , Elementos Facilitadores Genéticos , Éxons , Regulação da Expressão Gênica , Variação Genética , Células HEK293 , Células HeLa , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H/metabolismo , Humanos , Recém-Nascido , Deficiência Múltipla de Acil Coenzima A Desidrogenase/diagnóstico , Mutação de Sentido Incorreto , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas de Ligação a RNA/metabolismo , Análise de Sequência de DNA , Fatores de Processamento de Serina-Arginina , Elementos Silenciadores Transcricionais , Repetições de Trinucleotídeos
4.
J Inherit Metab Dis ; 36(4): 645-58, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23674167

RESUMO

Fatty acids are a major fuel for the body and fatty acid oxidation is particularly important during fasting, sustained aerobic exercise and stress. The myocardium and resting skeletal muscle utilise long-chain fatty acids as a major source of energy. Inherited disorders affecting fatty acid oxidation seriously compromise the function of muscle and other highly energy-dependent tissues such as brain, nerve, heart, kidney and liver. Such defects encompass a wide spectrum of clinical disease, presenting in the neonatal period or infancy with recurrent hypoketotic hypoglycaemic encephalopathy, liver dysfunction, hyperammonaemia and often cardiac dysfunction. In older children, adolescence or adults there is often exercise intolerance with episodic myalgia or rhabdomyolysis in association with prolonged aerobic exercise or other exacerbating factors. Some disorders are particularly associated with toxic metabolites that may contribute to encephalopathy, polyneuropathy, axonopathy and pigmentary retinopathy. The phenotypic diversity encountered in defects of fat oxidation is partly explained by genotype/phenotype correlation and certain identifiable environmental factors but there remain many unresolved questions regarding the complex interaction of genetic, epigenetic and environmental influences that dictate phenotypic expression. It is becoming increasingly clear that the view that most inherited disorders are purely monogenic diseases is a naive concept. In the future our approach to understanding the phenotypic diversity and management of patients will be more realistically achieved from a polygenic perspective.


Assuntos
Ácidos Graxos/metabolismo , Erros Inatos do Metabolismo Lipídico/metabolismo , Metabolismo dos Lipídeos/fisiologia , Humanos , Oxirredução , Fenótipo
5.
J Nutr ; 143(2): 142-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23236022

RESUMO

Plasma vitamin B-12 is the most commonly used biomarker of vitamin B-12 status, but the predictive value for low vitamin B-12 status is poor. The urinary methylmalonic acid (uMMA) concentration has potential as a functional biomarker of vitamin B-12 status, but the response to supplemental vitamin B-12 is uncertain. A study was conducted to investigate the responsiveness of uMMA to supplemental vitamin B-12 in comparison with other biomarkers of vitamin B-12 status [plasma vitamin B-12, serum holotranscobalamin (holoTC), plasma MMA] in elderly people with moderately poor vitamin B-12 status. A double-blind, placebo-controlled, randomized 8-wk intervention study was carried out using vitamin B-12 supplements (500 µg/d, 100 µg/d, and 10 µg/d cyanocobalamin) in 100 elderly people with a combined plasma vitamin B-12 <250 pmol/L and uMMA ratio (µmol MMA/mmol creatinine) >1.5. All biomarkers had a dose response to supplemental vitamin B-12. Improvements in plasma vitamin B-12 and serum holoTC were achieved at cobalamin supplements of 10 µg/d, but even 500 µg/d for 8 wk did not normalize plasma vitamin B-12 in 8% and serum holoTC in 12% of people. The response in uMMA was comparable with plasma MMA; 15-25% of people still showed evidence of metabolic deficiency after 500 µg/d cobalamin for 8 wk. There was a differential response in urinary and plasma MMA according to smoking behavior; the response was enhanced in ex-smokers compared with never-smokers. uMMA offers an alternative marker of metabolic vitamin-B12 status, obviating the need for blood sampling.


Assuntos
Envelhecimento , Suplementos Nutricionais , Ácido Metilmalônico/urina , Estado Nutricional , Deficiência de Vitamina B 12/dietoterapia , Vitamina B 12/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Apoproteínas/sangue , Biomarcadores/sangue , Biomarcadores/urina , Creatinina/urina , Estudos Transversais , Método Duplo-Cego , Feminino , Humanos , Masculino , Ácido Metilmalônico/sangue , Cooperação do Paciente , Fumar/efeitos adversos , Fatores de Tempo , Transcobalaminas/análise , Vitamina B 12/sangue , Vitamina B 12/uso terapêutico , Deficiência de Vitamina B 12/sangue , Deficiência de Vitamina B 12/fisiopatologia , Deficiência de Vitamina B 12/urina
6.
Am J Clin Nutr ; 95(3): 686-93, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22301932

RESUMO

BACKGROUND: An age-related deterioration of vitamin B-12 status has been well documented. The early detection of deficiency may prevent the development of serious clinical symptoms, but plasma vitamin B-12 concentration is known to be an imperfect measure of vitamin B-12 status. Urinary methylmalonic acid (MMA) may be a more informative biomarker of vitamin B-12 status; however, biochemical, dietary, and other lifestyle determinants are not known. OBJECTIVE: We identified determinants of urinary MMA concentrations in free-living men and women aged ≥65 y in the United Kingdom. DESIGN: A cross-sectional study in 591 men and women aged 65-85 y, with no clinical evidence of vitamin B-12 deficiency, was conducted to determine the demographic, clinical, and lifestyle determinants of urinary MMA concentration expressed as the ratio of micromoles of MMA to millimoles of creatinine (uMMA ratio). RESULTS: Twenty percent of subjects had plasma vitamin B-12 concentrations <200 pmol/L. Seventeen percent of the variation in the uMMA ratio could be explained by plasma holotranscobalamin and sex; total vitamin B-12 intake and measures of renal function and gastric function made only a small contribution to the model. The uMMA ratio was lower in people with moderately impaired renal function. CONCLUSIONS: Plasma holotranscobalamin and sex were the most important determinants of uMMA ratio in elderly people with no clinical diagnosis of renal impairment. This biomarker might underestimate vitamin B-12 deficiency in a population in which renal impairment is prevalent. This trial was registered at www.controlled-trials.com as ISRCJN83921062.


Assuntos
Ácido Metilmalônico/urina , Deficiência de Vitamina B 12/diagnóstico , Deficiência de Vitamina B 12/epidemiologia , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/urina , Estudos Transversais , Feminino , Humanos , Estilo de Vida , Masculino , Prevalência , Ensaios Clínicos Controlados Aleatórios como Assunto , Transcobalaminas/análise , Reino Unido/epidemiologia , Vitamina B 12/sangue
7.
Mol Genet Metab ; 101(2-3): 178-82, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20732827

RESUMO

Glutaric Aciduria type 1 (GA 1) is an inherited disorder of lysine and tryptophan catabolism that typically manifests in infants with acute cerebral injury associated with intercurrent illness. We investigated the clinical, biochemical and molecular features in 14 known GA 1 patients in South Africa, most of whom were recently confirmed following the implementation of sensitive urine organic acid screening at our laboratory. Age at diagnosis ranged from 3days to 5years and poor clinical outcome reflected the delay in diagnosis in all but one patient. Twelve patients were unrelated black South Africans of whom all those tested (n=11) were found homozygous for the same A293T mutation in the glutaryl-CoA dehydrogenase (GCDH) gene. Excretion of 3-hydroxyglutarate (3-OHGA) was >30.1µmol/mmol creatinine (reference range <2.5) in all cases but glutarate excretion varied with 5 patients considered low excretors (glutarate <50µmol/mmol creatinine). Fibroblast GCDH activity was very low or absent in all of five cases tested. Heterozygosity for the A293T mutation was found 1 in 36 (95% CI; 1/54 - 1/24) unrelated black South African newborns (n=750) giving a predicted prevalence rate for GA 1 of 1 in 5184 (95% CI; 1/11664 - 1/2304) in this population. GA 1 is a treatable but often missed inherited disorder with a previously unrecognised high carrier frequency of a single mutation in the South African black population.


Assuntos
Glutaril-CoA Desidrogenase/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/epidemiologia , Erros Inatos do Metabolismo dos Aminoácidos/genética , População Negra/genética , Encefalopatias Metabólicas/epidemiologia , Encefalopatias Metabólicas/genética , Pré-Escolar , Feminino , Glutaril-CoA Desidrogenase/genética , Humanos , Incidência , Lactente , Recém-Nascido , Masculino , África do Sul/epidemiologia
8.
Proc Natl Acad Sci U S A ; 106(35): 14820-4, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19706438

RESUMO

Mutations in the HSD17B10 gene were identified in two previously described mentally retarded males. A point mutation c.776G>C was found from a survivor (SV), whereas a potent mutation, c.419C>T, was identified in another deceased case (SF) with undetectable hydroxysteroid (17beta) dehydrogenase 10 (HSD10) activity. Protein levels of mutant HSD10(R130C) in patient SF and HSD10(E249Q) in patient SV were about half that of HSD10 in normal controls. The E249Q mutation appears to affect HSD10 subunit interactions, resulting in an allosteric regulatory enzyme. For catalyzing the oxidation of allopregnanolone by NAD+ the Hill coefficient of the mutant enzyme is approximately 1.3. HSD10(E249Q) was unable to catalyze the dehydrogenation of 2-methyl-3-hydroxybutyryl-CoA and the oxidation of allopregnanolone, a positive modulator of the gamma-aminobutyric acid type A receptor, at low substrate concentrations. Neurosteroid homeostasis is critical for normal cognitive development, and there is increasing evidence that a blockade of isoleucine catabolism alone does not commonly cause developmental disabilities. The results support the theory that an imbalance in neurosteroid metabolism could be a major cause of the neurological handicap associated with hydroxysteroid (17beta) dehydrogenase 10 deficiency.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/genética , Deficiência Intelectual/enzimologia , Deficiência Intelectual/genética , Isoleucina/metabolismo , Mutação Puntual , Esteroides/metabolismo , 3-Hidroxiacil-CoA Desidrogenases/química , 3-Hidroxiacil-CoA Desidrogenases/deficiência , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Biocatálise , Células Cultivadas , Fibroblastos/enzimologia , Humanos , Masculino , Modelos Moleculares , Estrutura Terciária de Proteína
9.
Mol Genet Metab ; 96(4): 201-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19217814

RESUMO

Investigation of seven patients from three families suspected of a fatty acid oxidation defect showed mean CPT-I enzyme activity of 5.9+/-4.9 percent of normal controls. The families, two Inuit, one First Nation, live in areas of Canada geographically very distant from each other. The CPT1 and CPT2 genes were fully sequenced in 5 of the patients. All were homozygous for the same P479L mutation in a highly conserved region of the CPT1 gene. Two patients from the first family were also homozygous for the CPT2 F352C polymorphism in the CPT2 gene. Genotyping the patients and their family members confirmed that all seven patients were homozygous for the P479L variant allele in the CPT1 gene, as were 27 of 32 family members. Three of the seven patients and two cousins had hypoketotic hypoglycemia attributable to CPT-Ia deficiency, but adults homozygous for the variant denied hypoglycemia. We screened 422 consecutive newborns from the region of one of the Inuit families for this variant; 294 were homozygous, 103 heterozygous, and only 25 homozygous normal; thus the frequency of this variant allele is 0.81. There was an infant death in one family and at least 10 more deaths in those infants (7 homozygous, 3 heterozygous) consecutively tested for the mutation at birth. Thus there is an astonishingly high frequency of CPT1 P479L variant and, judging from the enzyme analysis in the seven patients, also CPT-I deficiency in the areas of Canada inhabited by these families. Despite the deficiency of CPT-Ia which is the major rate-limiting enzyme for long chain fatty acid oxidation, clinical effects, with few exceptions, were slight or absent. One clue to explaining this paradox is that, judging from the fatty acid oxidation studies in whole blood and fibroblasts, the low residual activity of CPT-Ia is sufficient to allow a reasonable flux through the mitochondrial oxidation system. It is likely that the P479L variant is of ancient origin and presumably its preservation must have conveyed some advantage.


Assuntos
Indígena Americano ou Nativo do Alasca/genética , Substituição de Aminoácidos/genética , Carnitina O-Palmitoiltransferase/genética , Polimorfismo de Nucleotídeo Único/genética , Adulto , Canadá , Feminino , Fibroblastos/enzimologia , Humanos , Recém-Nascido , Masculino , Oxirredução , Reação em Cadeia da Polimerase , Gravidez
10.
Am J Med Genet A ; 146A(12): 1581-6, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18478588

RESUMO

We report on a 6-year-old girl who presented at 6 months of age with seizures, delayed psychomotor development and mild facial dysmorphism. A small muscular ventricular septal defect was documented on echocardiogram and brain MRI showed a frontal brain anomaly. Urine organic acid analysis revealed dicarboxylic aciduria, and plasma acylcarnitine analysis showed marked elevation of octanoyl (C8) and decanoyl (C10) carnitines with C8:C10 ratio of 9:1. These results were indicative of medium chain acyl-CoA dehydrogenase deficiency. ACADM gene sequencing showed an apparent homozygous c.166G > C (Ala31Pro) missense mutation in exon 3; however, only the mother was found to be a carrier of this novel missense mutation. This finding along with non-regressive developmental delay prompted further karyotype and genomic investigations. An interstitial deletion of chromosome 1 was detected by repeat G-banding: 46,XX,del(1)(p22.2p31.1). Parental karyotypes were normal. The deletion was characterized by array CGH analysis using a 1 Mb BAC/PAC array platform. Clones deleted extended from RP11-88B10 (1p31.1) to RP5-1007M22 (1p22.2), a 15.5 Mb deletion which includes the ACADM locus. Clinical review of 6/7 cases of interstitial deletions with breakpoints of 1p22 and 1p31/32, including the patient in this report, indicate a variable phenotype. Thus, although G-band breakpoints are similar, common breakpoints for these alterations are unlikely. This is the first report of a patient with fatty acid oxidation defect caused by a mutation in combination with an interstitial chromosomal deletion.


Assuntos
Acil-CoA Desidrogenase/metabolismo , Deleção Cromossômica , Cromossomos Humanos Par 1/genética , Transtornos Psicomotores/enzimologia , Transtornos Psicomotores/genética , Convulsões/enzimologia , Convulsões/genética , Acil-CoA Desidrogenase/genética , Carnitina/análogos & derivados , Carnitina/sangue , Criança , Análise Mutacional de DNA , Ácidos Dicarboxílicos/urina , Éxons , Fácies , Feminino , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Mutação de Sentido Incorreto , Análise de Sequência com Séries de Oligonucleotídeos , Transtornos Psicomotores/diagnóstico , Convulsões/diagnóstico
11.
Brain ; 130(Pt 8): 2045-54, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17584774

RESUMO

Multiple acyl-CoA dehydrogenation deficiency (MADD) is a disorder of fatty acid, amino acid and choline metabolism that can result from defects in two flavoproteins, electron transfer flavoprotein (ETF) or ETF: ubiquinone oxidoreductase (ETF:QO). Some patients respond to pharmacological doses of riboflavin. It is unknown whether these patients have defects in the flavoproteins themselves or defects in the formation of the cofactor, FAD, from riboflavin. We report 15 patients from 11 pedigrees. All the index cases presented with encephalopathy or muscle weakness or a combination of these symptoms; several had previously suffered cyclical vomiting. Urine organic acid and plasma acyl-carnitine profiles indicated MADD. Clinical and biochemical parameters were either totally or partly corrected after riboflavin treatment. All patients had mutations in the gene for ETF:QO. In one patient, we show that the ETF:QO mutations are associated with a riboflavin-sensitive impairment of ETF:QO activity. This patient also had partial deficiencies of flavin-dependent acyl-CoA dehydrogenases and respiratory chain complexes, most of which were restored to control levels after riboflavin treatment. Low activities of mitochondrial flavoproteins or respiratory chain complexes have been reported previously in two of our patients with ETF:QO mutations. We postulate that riboflavin-responsive MADD may result from defects of ETF:QO combined with general mitochondrial dysfunction. This is the largest collection of riboflavin-responsive MADD patients ever reported, and the first demonstration of the molecular genetic basis for the disorder.


Assuntos
Acil-CoA Desidrogenase/deficiência , Flavoproteínas Transferidoras de Elétrons/genética , Proteínas Ferro-Enxofre/genética , Miopatias Mitocondriais/genética , Mutação , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , Riboflavina/uso terapêutico , Adolescente , Adulto , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/genética , Carnitina/análogos & derivados , Carnitina/sangue , Criança , Pré-Escolar , Transporte de Elétrons/fisiologia , Ácidos Graxos/metabolismo , Feminino , Humanos , Masculino , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Erros Inatos do Metabolismo/patologia , Mitocôndrias Musculares/metabolismo , Miopatias Mitocondriais/tratamento farmacológico , Miopatias Mitocondriais/metabolismo , Miopatias Mitocondriais/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Oxirredução
12.
Am J Hum Genet ; 80(5): 931-7, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17436247

RESUMO

We present the first two identified cases of phosphoserine aminotransferase deficiency. This disorder of serine biosynthesis has been identified in two siblings who showed low concentrations of serine and glycine in plasma and cerebrospinal fluid. Clinically, the index patient presented with intractable seizures, acquired microcephaly, hypertonia, and psychomotor retardation and died at age 7 mo despite supplementation with serine (500 mg/kg/d) and glycine (200 mg/kg/d) from age 11 wk. The younger sibling received treatment from birth, which led to a normal outcome at age 3 years. Measurement of phosphoserine aminotransferase activity in cultured fibroblasts in the index patient was inconclusive, but mutational analysis revealed compound heterozygosity for two mutations in the PSAT1 gene--one frameshift mutation (c.delG107) and one missense mutation (c.299A-->C [p.Asp100Ala])--in both siblings. Expression studies of the p.Asp100Ala mutant protein revealed a V(max) of only 15% of that of the wild-type protein.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Serina/biossíntese , Transaminases/deficiência , Transaminases/genética , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Sequência de Aminoácidos , Sequência de Bases , Encéfalo/patologia , Pré-Escolar , DNA/genética , Feminino , Mutação da Fase de Leitura , Glicina/deficiência , Glicina/uso terapêutico , Heterozigoto , Humanos , Técnicas In Vitro , Lactente , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Serina/deficiência , Serina/uso terapêutico , Transaminases/química , Transaminases/metabolismo
13.
Clin Lab ; 51(5-6): 289-306, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15991803

RESUMO

Fatty acids are a major fuel for the body and fatty acid oxidation is particularly important during fasting, sustained aerobic exercise & stress. The myocardium and resting skeletal muscle utilise long-chain fatty acids as a major source of energy. Inherited disorders of fatty acid oxidation seriously compromise the function of muscle and other highly energy-dependent tissues such as brain, nerve, heart, kidney & liver. Defects of fatty acid oxidation lead to a range of neuromyopathic disease in both adults and children. Such defects encompass a wide spectrum of clinical disease, presenting in the neonatal period or infancy with recurrent hypoketotic hypoglycaemic encephalopathy, liver dysfunction and hyperammonaemia with neurosensory deficits secondary to the acute onset. In addition, there may be cardiac arrhythmias and/or progressive cardiomyopathy, which may give rise to secondary hypoxic-ischaemic encephalopathy. In older children, adolescence or adults there is often exercise intolerance with episodic myalgia or rhabdomyolysis in association with prolonged aerobic exercise or other exacerbating factors. Some disorders are particularly associated with toxic metabolites that may contribute to encephalopathy, polyneuropathy, axonopathy and pigmentary retinopathy. Diagnosis is through clinical suspicion with appropriate investigations in blood and urine taken during crisis. Definitive diagnosis is usually by fibroblast assay. Treatment is generally through avoidance of fasting, frequent carbohydrate rich feeds and in long-chain defects MCT supplementation. Novel treatments include the use of D,L-3-hydroxybutyrate and the potential use of fibrates to increase mutant protein levels in mild disorders.


Assuntos
Encefalopatias Metabólicas/etiologia , Ácidos Graxos/metabolismo , Acil-CoA Desidrogenase/deficiência , Encefalopatias Metabólicas/terapia , Carnitina/biossíntese , Carnitina/deficiência , Dietoterapia , Humanos , Proteínas Mitocondriais/deficiência , Oxirredução
14.
Eur J Biochem ; 271(20): 4053-63, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15479234

RESUMO

Medium-chain acyl-CoA dehydrogenase (MCAD) is a homotetrameric flavoprotein which catalyses the initial step of the beta-oxidation of medium-chain fatty acids. Mutations in MCAD may cause disease in humans. A Y42H mutation is frequently found in babies identified by newborn screening with MS/MS, yet there are no reports of patients presenting clinically with this mutation. As a basis for judging its potential consequences we have examined the protein phenotype of the Y42H mutation and the common disease-associated K304E mutation. Our studies of the intracellular biogenesis of the variant proteins at different temperatures in isolated mitochondria after in vitro translation, together with studies of cultured patient cells, indicated that steady-state levels of the Y42H variant in comparison to wild-type were decreased at higher temperature though to a lesser extent than for the K304E variant. To distinguish between effects of temperature on folding/assembly and the stability of the native enzyme, the thermal stability of the variant proteins was studied after expression and purification by dye affinity chromatography. This showed that, compared with the wild-type enzyme, the thermostability of the Y42H variant was decreased, but not to the same degree as that of the K304E variant. Substrate binding, interaction with the natural electron acceptor, and the binding of the prosthetic group, FAD, were only slightly affected by the Y42H mutation. Our study suggests that Y42H is a temperature sensitive mutation, which is mild at low temperatures, but may have deleterious effects at increased temperatures.


Assuntos
Acil-CoA Desidrogenase/genética , Acil-CoA Desidrogenase/metabolismo , Acil-CoA Desidrogenase/química , Acil-CoA Desidrogenase/deficiência , Substituição de Aminoácidos , Animais , Dicroísmo Circular/métodos , Estabilidade Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Fibroblastos/metabolismo , Ácido Glutâmico/genética , Histidina/genética , Humanos , Recém-Nascido , Linfócitos/citologia , Linfócitos/metabolismo , Lisina/genética , Mitocôndrias Hepáticas/metabolismo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ácido Mirístico/metabolismo , Triagem Neonatal , Oxirredução , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Tirosina/genética
15.
Ann Clin Biochem ; 41(Pt 4): 282-93, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15298740

RESUMO

Inherited metabolic disorders account for a small but significant number of sudden unexplained deaths in neonates, infants and occasionally older children. In particular, inherited disorders of fatty acid oxidation may closely mimic sudden infant death syndrome. Post-mortem investigations offer the final opportunity to establish a diagnosis. Such diagnoses are of great importance to the families concerned and provide the opportunity for genetic counselling and antenatal diagnosis. Current advances in technology, particularly in the case of electrospray ionization tandem mass spectrometry, have revolutionized the investigation of metabolic disease at post-mortem, facilitating the identification of a wide range of metabolic diseases in tiny samples of blood, plasma and bile. Such analyses may provide vital clues to diagnosis, usually in the form of acylcarnitine profiles. Accurate diagnosis relies on the timely collection of appropriate samples and the subsequent selection of informative testing. In order to maximize the chances of a diagnosis, a collaborative approach between the various disciplines is vital. A brief description of the more frequently encountered inherited disorders, collection and processing of appropriate samples and available investigations that may lead to accurate diagnosis are clearly described.


Assuntos
Erros Inatos do Metabolismo/complicações , Morte Súbita do Lactente/etiologia , Autopsia , Análise Química do Sangue , Células Cultivadas , Técnicas de Química Analítica , Fibroblastos/metabolismo , Humanos , Lactente , Recém-Nascido , Erros Inatos do Metabolismo/diagnóstico , Erros Inatos do Metabolismo/metabolismo , Espectrometria de Massas por Ionização por Electrospray , Morte Súbita do Lactente/diagnóstico
16.
Am J Med Genet ; 111(3): 271-84, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12210323

RESUMO

Primary systemic carnitine deficiency or carnitine uptake defect (OMIM 212140) is a potentially lethal, autosomal recessive disorder characterized by progressive infantile-onset cardiomyopathy, weakness, and recurrent hypoglycemic hypoketotic encephalopathy, which is highly responsive to L-carnitine therapy. Molecular analysis of the SLC22A5 (OCTN2) gene, encoding the high-affinity carnitine transporter, was done in 11 affected individuals by direct nucleotide sequencing of polymerase chain reaction products from all 10 exons. Carnitine uptake (at Km of 5 microM) in cultured skin fibroblasts ranged from 1% to 20% of normal controls. Eleven mutations (delF23, N32S, and one 11-bp duplication in exon 1; R169W in exon 3; a donor splice mutation [IVS3+1 G > A] in intron 3; frameshift mutations in exons 5 and 6; Y401X in exon 7; T440M, T468R and S470F in exon 8) are described. There was no correlation between residual uptake and severity of clinical presentation, suggesting that the wide phenotypic variability is likely related to exogenous stressors exacerbating carnitine deficiency. Most importantly, strict compliance with carnitine from birth appears to prevent the phenotype.


Assuntos
Cardiomiopatias/prevenção & controle , Carnitina/farmacologia , Proteínas de Transporte/genética , Coração/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Transporte de Cátions Orgânicos , Carnitina/deficiência , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Mutação , Linhagem , Membro 5 da Família 22 de Carreadores de Soluto , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...