Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Am J Med Genet A ; 173(6): 1601-1606, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28544736

RESUMO

We describe the clinical presentation and 17 years follow up of a boy, born to consanguineous parents and presenting with intellectual disability (ID), autism, "marfanoid" dysmorphic features, and moderate abnormalities of sulfite metabolism compatible with molybdenum cofactor deficiency, but normal sulfite oxidase activity in cultured skin fibroblasts. Genomic exome analysis revealed a homozygous MOCS3 missense mutation, leading to a p.Ala257Thr substitution in the highly conserved ubiquitin-like-domain of the protein. MOCS3 is the third protein, besides MOCS1 and MOCS2, involved in the biosynthesis of the molybdenum cofactor and has a dual ubiquitin-like function in tRNA thiolation. It is plausible that the phenotype results from deficiency of this dual function, not only from defective synthesis of molybdenum cofactor, which would explain similarities and differences from the MOCS1 and MOCS2-related disorders. This observation should encourage testing of additional ID patients with mild abnormalities of sulfite metabolism for MOCS3 mutations.


Assuntos
Transtorno Autístico/genética , Deficiência Intelectual/genética , Erros Inatos do Metabolismo dos Metais/genética , Nucleotidiltransferases/genética , Sulfurtransferases/genética , Adolescente , Transtorno Autístico/complicações , Transtorno Autístico/fisiopatologia , Expressão Gênica , Homozigoto , Humanos , Deficiência Intelectual/complicações , Deficiência Intelectual/fisiopatologia , Masculino , Erros Inatos do Metabolismo dos Metais/complicações , Erros Inatos do Metabolismo dos Metais/fisiopatologia , Mutação de Sentido Incorreto , Fenótipo
2.
Mol Genet Metab ; 119(4): 307-310, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27771289

RESUMO

BACKGROUND: N-acetylglutamate synthase (NAGS) plays a key role in the removal of ammonia via the urea cycle by catalyzing the synthesis of N-acetylglutamate (NAG), the obligatory cofactor in the carbamyl phosphate synthetase 1 reaction. Enzymatic analysis of NAGS in liver homogenates has remained insensitive and inaccurate, which prompted the development of a novel method. METHODS: UPLC-MS/MS was used in conjunction with stable isotope (N-acetylglutamic-2,3,3,4,4-d5 acid) dilution for the quantitative detection of NAG produced by the NAGS enzyme. The assay conditions were optimized using purified human NAGS and the optimized enzyme conditions were used to measure the activity in mouse liver homogenates. RESULTS: A low signal-to-noise ratio in liver tissue samples was observed due to non-enzymatic formation of N-acetylglutamate and low specific activity, which interfered with quantitative analysis. Quenching of acetyl-CoA immediately after the incubation circumvented this analytical difficulty and allowed accurate and sensitive determination of mammalian NAGS activity. The specificity of the assay was validated by demonstrating a complete deficiency of NAGS in liver homogenates from Nags -/- mice. CONCLUSION: The novel NAGS enzyme assay reported herein can be used for the diagnosis of inherited NAGS deficiency and may also be of value in the study of secondary hyperammonemia present in various inborn errors of metabolism as well as drug treatment.


Assuntos
Aminoácido N-Acetiltransferase/genética , Carbamoil-Fosfato Sintase (Amônia)/genética , Hiperamonemia/diagnóstico , Distúrbios Congênitos do Ciclo da Ureia/diagnóstico , Acetilcoenzima A/metabolismo , Aminoácido N-Acetiltransferase/metabolismo , Animais , Carbamoil-Fosfato Sintase (Amônia)/deficiência , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatologia , Fígado/enzimologia , Camundongos , Camundongos Knockout , Espectrometria de Massas em Tandem , Distúrbios Congênitos do Ciclo da Ureia/genética , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/fisiopatologia
3.
J Inherit Metab Dis ; 38(5): 873-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25860818

RESUMO

Alpha-aminoadipic and alpha-ketoadipic aciduria is an autosomal recessive inborn error of lysine, hydroxylysine, and tryptophan degradation. To date, DHTKD1 mutations have been reported in two alpha-aminoadipic and alpha-ketoadipic aciduria patients. We have now sequenced DHTKD1 in nine patients diagnosed with alpha-aminoadipic and alpha-ketoadipic aciduria as well as one patient with isolated alpha-aminoadipic aciduria, and identified causal mutations in eight. We report nine novel mutations, including three missense mutations, two nonsense mutations, two splice donor mutations, one duplication, and one deletion and insertion. Two missense mutations, one of which was reported before, were observed in the majority of cases. The clinical presentation of this group of patients was inhomogeneous. Our results confirm that alpha-aminoadipic and alpha-ketoadipic aciduria is caused by mutations in DHTKD1, and further establish that DHTKD1 encodes the E1 subunit of the alpha-ketoadipic acid dehydrogenase complex.


Assuntos
Ácido 2-Aminoadípico/metabolismo , Adipatos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/genética , Cetona Oxirredutases/genética , Ácido 2-Aminoadípico/urina , Adipatos/urina , Adolescente , Adulto , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Pré-Escolar , Feminino , Humanos , Recém-Nascido , Complexo Cetoglutarato Desidrogenase , Cetona Oxirredutases/deficiência , Cetona Oxirredutases/metabolismo , Masculino , Adulto Jovem
4.
Biochim Biophys Acta ; 1832(6): 773-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23485643

RESUMO

Carnitine acyltransferases catalyze the reversible conversion of acyl-CoAs into acylcarnitine esters. This family includes the mitochondrial enzymes carnitine palmitoyltransferase 2 (CPT2) and carnitine acetyltransferase (CrAT). CPT2 is part of the carnitine shuttle that is necessary to import fatty acids into mitochondria and catalyzes the conversion of acylcarnitines into acyl-CoAs. In addition, when mitochondrial fatty acid ß-oxidation is impaired, CPT2 is able to catalyze the reverse reaction and converts accumulating long- and medium-chain acyl-CoAs into acylcarnitines for export from the matrix to the cytosol. However, CPT2 is inactive with short-chain acyl-CoAs and intermediates of the branched-chain amino acid oxidation pathway (BCAAO). In order to explore the origin of short-chain and branched-chain acylcarnitines that may accumulate in various organic acidemias, we performed substrate specificity studies using purified recombinant human CrAT. Various saturated, unsaturated and branched-chain acyl-CoA esters were tested and the synthesized acylcarnitines were quantified by ESI-MS/MS. We show that CrAT converts short- and medium-chain acyl-CoAs (C2 to C10-CoA), whereas no activity was observed with long-chain species. Trans-2-enoyl-CoA intermediates were found to be poor substrates for this enzyme. Furthermore, CrAT turned out to be active towards some but not all the BCAAO intermediates tested and no activity was found with dicarboxylic acyl-CoA esters. This suggests the existence of another enzyme able to handle the acyl-CoAs that are not substrates for CrAT and CPT2, but for which the corresponding acylcarnitines are well recognized as diagnostic markers in inborn errors of metabolism.


Assuntos
Aminoácidos de Cadeia Ramificada/química , Aminoácidos de Cadeia Ramificada/metabolismo , Carnitina O-Acetiltransferase/química , Carnitina O-Acetiltransferase/metabolismo , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Acil Coenzima A/química , Acil Coenzima A/genética , Acil Coenzima A/metabolismo , Aminoácidos de Cadeia Ramificada/genética , Carnitina O-Acetiltransferase/genética , Carnitina O-Palmitoiltransferase/química , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Ácidos Graxos/genética , Humanos , Especificidade por Substrato/fisiologia
5.
Mol Genet Metab ; 111(3): 404-407, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24100244

RESUMO

An infant carrying a heterozygous c.43_46delACTA and a heterozygous c.668 G>A mutation in the ALPL gene with hypophosphatasia in the absence of bone deformities presented with therapy-resistant seizures. Pyridoxal phosphate was extremely high in CSF and plasma. Pyridoxine treatment had only a transient effect and the severe encephalopathy was fatal. Repeated brain MRIs showed progressive cerebral damage. The precise metabolic cause of the seizures remains unknown and pyridoxine treatment apparently does not cure the epilepsy.


Assuntos
Epilepsia/patologia , Hipofosfatasia/genética , Hipofosfatasia/patologia , Piridoxina/administração & dosagem , Fosfatase Alcalina/genética , Resistência a Medicamentos , Epilepsia/complicações , Epilepsia/mortalidade , Humanos , Hipofosfatasia/sangue , Hipofosfatasia/líquido cefalorraquidiano , Hipofosfatasia/mortalidade , Lactente , Masculino , Fosfato de Piridoxal/sangue , Fosfato de Piridoxal/líquido cefalorraquidiano , Convulsões/genética , Convulsões/patologia
6.
J Inherit Metab Dis ; 37(3): 353-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24154984

RESUMO

BACKGROUND: Valproic acid (VPA) is an effective antiepileptic drug that may induce progressive microvesicular steatosis. The impairment of mitochondrial function may be an important metabolic effect of VPA treatment with potential adverse consequences. OBJECTIVE: To investigate the influence of VPA on the activity of GTP- and ATP-specific succinate:CoA ligases (G-SUCL and A-SUCL). METHODS: The GTP- and ATP-specific SUCL activities were measured in human fibroblasts in the reverse direction, i.e. the formation of succinyl-CoA. These were assessed at different concentrations of succinate in the presence of VPA, valproyl-CoA and zinc chloride, an established inhibitor of the enzymes. Activities were measured using an optimized HPLC procedure. RESULTS: Valproyl-CoA (1 mM) inhibited the activity of A-SUCL and G-SUCL by 45-55% and 25-50%, respectively. VPA (1 mM) had no influence on the activity of the two enzymes. DISCUSSION: Valproyl-CoA appears to affect the activity of SUCL, especially with the ATP-specific enzyme. Considering the key role of SUCL in the Krebs cycle, interference with its activity might impair the cellular energy status. Moreover, A-SUCL is bound to the nucleoside diphosphate kinase (NDPK), which is responsible for the mitochondrial (deoxy)nucleotide synthesis. An inhibition of A-SUCL might influence the activity of NDPK inducing an imbalance of nucleotides in the mitochondria and eventually mitochondrial DNA depletion. This may account for the potential liver failure associated with valproate therapy, reported in patients with deficiencies within the mitochondrial DNA replicase system such as polymerase gamma 1.


Assuntos
Acil Coenzima A/farmacologia , Trifosfato de Adenosina/fisiologia , Guanosina Trifosfato/fisiologia , Succinato-CoA Ligases/antagonistas & inibidores , DNA Mitocondrial/metabolismo , Humanos , Falência Hepática/induzido quimicamente , Núcleosídeo-Difosfato Quinase/fisiologia , Ácido Valproico/efeitos adversos , Ácido Valproico/farmacologia
7.
J Inherit Metab Dis ; 36(6): 967-72, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23392989

RESUMO

Rett syndrome is a neurodevelopmental disorder characterized by cognitive and locomotor regression and stereotypic hand movements. The disorder is caused by mutations in the X chromosomal MECP2 a gene encoding methyl CpG-binding protein. It has been associated with disturbances of cerebral folate homeostasis, as well as with speculations on a compromised DNA-methylation. Folinic acid is the stable form of folate. Its derived intermediate 5-MTHF supports the conversion of homocysteine to methionine, the precursor of S-adenosylmethionine (SAM). This in turn donates its methyl group to various acceptors, including DNA, thereby being converted to S-adenosylhomocysteine (SAH). The SAM/SAH ratio reflects the methylation potential. The goal of our study was to influence DNA methylation processes and ameliorate the clinical symptoms in Rett syndrome. Therefore we examined the hypothesis that folinic acid supplementation, besides increasing cerebrospinal fluid (CSF) 5-MTHF (p = 0.003), influences SAM and SAH and their ratio. In our randomized, double-blind crossover study on folinic acid supplementation, ten female Rett patients received both folinic acid and placebo for 1 year each. It was shown that both SAM and SAH levels in the CSF remained unchanged following folinic acid administration (p = 0.202 and p = 0.097, respectively) in spite of a rise of plasma SAM and SAH (p = 0.007; p = 0.009). There was no significant change in the SAM/SAH ratio either in plasma or CSF. The apparent inability of Rett patients to upregulate SAM and SAH levels in the CSF may contribute to the biochemical anomalies of the Rett syndrome. Our studies warrant further attempts to promote DNA methylation in the true region of interest, i.e. the brain.


Assuntos
Ácido Fólico/uso terapêutico , Síndrome de Rett/tratamento farmacológico , S-Adenosil-Homocisteína/sangue , S-Adenosil-Homocisteína/líquido cefalorraquidiano , S-Adenosilmetionina/sangue , S-Adenosilmetionina/líquido cefalorraquidiano , Adolescente , Adulto , Criança , Pré-Escolar , Suplementos Nutricionais , Feminino , Ácido Fólico/análogos & derivados , Ácido Fólico/líquido cefalorraquidiano , Ácido Fólico/farmacologia , Humanos , Lactente , Síndrome de Rett/sangue , Síndrome de Rett/líquido cefalorraquidiano , Adulto Jovem
8.
J Inherit Metab Dis ; 36(6): 923-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23296368

RESUMO

Increased urinary 3-methylglutaconic acid excretion is a relatively common finding in metabolic disorders, especially in mitochondrial disorders. In most cases 3-methylglutaconic acid is only slightly elevated and accompanied by other (disease specific) metabolites. There is, however, a group of disorders with significantly and consistently increased 3-methylglutaconic acid excretion, where the 3-methylglutaconic aciduria is a hallmark of the phenotype and the key to diagnosis. Until now these disorders were labelled by roman numbers (I-V) in the order of discovery regardless of pathomechanism. Especially, the so called "unspecified" 3-methylglutaconic aciduria type IV has been ever growing, leading to biochemical and clinical diagnostic confusion. Therefore, we propose the following pathomechanism based classification and a simplified diagnostic flow chart for these "inborn errors of metabolism with 3-methylglutaconic aciduria as discriminative feature". One should distinguish between "primary 3-methylglutaconic aciduria" formerly known as type I (3-methylglutaconyl-CoA hydratase deficiency, AUH defect) due to defective leucine catabolism and the--currently known--three groups of "secondary 3-methylglutaconic aciduria". The latter should be further classified and named by their defective protein or the historical name as follows: i) defective phospholipid remodelling (TAZ defect or Barth syndrome, SERAC1 defect or MEGDEL syndrome) and ii) mitochondrial membrane associated disorders (OPA3 defect or Costeff syndrome, DNAJC19 defect or DCMA syndrome, TMEM70 defect). The remaining patients with significant and consistent 3-methylglutaconic aciduria in whom the above mentioned syndromes have been excluded, should be referred to as "not otherwise specified (NOS) 3-MGA-uria" until elucidation of the underlying pathomechanism enables proper (possibly extended) classification.


Assuntos
Glutaratos/urina , Erros Inatos do Metabolismo/classificação , Erros Inatos do Metabolismo/diagnóstico , Terminologia como Assunto , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/urina , Síndrome de Barth/diagnóstico , Síndrome de Barth/genética , Síndrome de Barth/urina , Cardiomiopatia Dilatada/diagnóstico , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/urina , Ataxia Cerebelar/diagnóstico , Ataxia Cerebelar/genética , Ataxia Cerebelar/urina , Coreia/diagnóstico , Coreia/genética , Coreia/urina , Diagnóstico Diferencial , Humanos , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/urina , Atrofia Óptica/diagnóstico , Atrofia Óptica/genética , Atrofia Óptica/urina , Paraplegia Espástica Hereditária/diagnóstico , Paraplegia Espástica Hereditária/genética , Paraplegia Espástica Hereditária/urina
9.
J Inherit Metab Dis ; 36(6): 961-6, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23242558

RESUMO

Phosphohydroxylysinuria has been described in two patients with neurological symptoms, but the deficient enzyme or mutated gene has never been identified. In the present work, we tested the hypothesis that this condition is due to mutations in the AGXT2L2 gene, recently shown to encode phosphohydroxylysine phospholyase. DNA analysis from a third patient, without neurological symptoms, but with an extreme hyperlaxicity of the joints, shows the existence of two mutations, p. Gly240Arg and p.Glu437Val, both in the heterozygous state. Sequencing of cDNA clones derived from fibroblasts mRNA indicated that the two mutations were allelic. Both mutations replace conserved residues. The mutated proteins were produced as recombinant proteins in Escherichia coli and HEK293T cells and shown to be very largely insoluble, whereas the wild-type one was produced as a soluble and active protein. We conclude that phosphohydroxylysinuria is due to mutations in the AGXT2L2 gene and the resulting lack of activity of phosphohydroxylysine phospholyase in vivo. The finding that the nul alleles of p.Gly240Arg and p.Glu437Val are present at low frequencies in the European and/or North American population suggests that this condition is more common than previously thought. The diversity of the clinical symptoms described in three patients with phosphohydroxylysinuria indicates that this is most likely not a neurometabolic disease.


Assuntos
Alanina Transaminase/genética , Epilepsias Mioclônicas/genética , Deficiência Intelectual/genética , Mutação de Sentido Incorreto , Fósforo-Oxigênio Liases/genética , Alanina Transaminase/química , Sequência de Aminoácidos , Células Cultivadas , Pré-Escolar , Feminino , Células HEK293 , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único/fisiologia , Dobramento de Proteína , Homologia de Sequência
11.
J Inherit Metab Dis ; 35(3): 443-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22189597

RESUMO

BACKGROUND: Valproic acid (VPA) is a widely used anticonvulsant drug which affects mitochondrial metabolism including the catabolism of fatty acids and branched-chain amino acids. AIMS: To elucidate the effect of valproate on the leucine pathway through a targeted metabolomics approach and the evaluation of the effects of valproate on the activity of biotinidase and 3-methylcrotonyl-CoA carboxylase (3MCC). METHODS: Urine organic acid analysis was performed in patients under VPA therapy and healthy controls using gas-chromatography/mass spectrometry (GC-MS). Biotinidase activity was determined in plasma samples of both groups using an optimized spectrophotometric assay. After immunoprecipitation of short-chain enoyl-CoA hydratase (crotonase, ECHS1), 3MCC activity was measured in human liver homogenate using high-performance liquid chromatography (HPLC), in the absence and presence of valproyl-CoA. RESULTS: The levels of 3-hydroxyisovaleric acid (3OH-IVA), one secondary metabolite of the leucine pathway, were significantly elevated in human urine after VPA treatment. Biotinidase activity in plasma samples ranged from very low to normal levels in treated patients as compared with controls. Enzyme activity measurements revealed inhibition of 3-methylcrotonyl-CoA carboxylase by valproyl-CoA (IC(50) = 1.36 mM). Furthermore, we show that after complete immunoprecipitation of crotonase in a human liver homogenate, 3-hydroxyisovaleryl-CoA is not formed. DISCUSSION: Our results suggest the interference of VPA with the activity of 3MCC through a potential cumulative effect: direct inhibition of the enzyme activity by the drug metabolite valproyl-CoA and the inhibition of biotinidase by valproate and/or its metabolites. These interactions may be associated with the skin rash and hair loss which are side effects often reported in VPA-treated patients.


Assuntos
Carbono-Carbono Ligases/antagonistas & inibidores , Carbono-Carbono Ligases/química , Inibidores Enzimáticos/farmacologia , Valeratos/metabolismo , Biotinidase/metabolismo , Estudos de Casos e Controles , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia Gasosa-Espectrometria de Massas/métodos , Humanos , Concentração Inibidora 50 , Cinética , Leucina/metabolismo , Fígado/enzimologia , Metabolômica/métodos , Modelos Químicos , Ácido Valproico/farmacologia
12.
Biochim Biophys Acta ; 1802(7-8): 639-48, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20362666

RESUMO

Dihydropyrimidinase (DHP) is the second enzyme of the pyrimidine degradation pathway and catalyses the ring opening of 5,6-dihydrouracil and 5,6-dihydrothymine. To date, only 11 individuals have been reported suffering from a complete DHP deficiency. Here, we report on the clinical, biochemical and molecular findings of 17 newly identified DHP deficient patients as well as the analysis of the mutations in a three-dimensional framework. Patients presented mainly with neurological and gastrointestinal abnormalities and markedly elevated levels of 5,6-dihydrouracil and 5,6-dihydrothymine in plasma, cerebrospinal fluid and urine. Analysis of DPYS, encoding DHP, showed nine missense mutations, two nonsense mutations, two deletions and one splice-site mutation. Seventy-one percent of the mutations were located at exons 5-8, representing 41% of the coding sequence. Heterologous expression of 11 mutant enzymes in Escherichia coli showed that all but two missense mutations yielded mutant DHP proteins without significant activity. Only DHP enzymes containing the mutations p.R302Q and p.T343A possessed a residual activity of 3.9% and 49%, respectively. The crystal structure of human DHP indicated that the point mutations p.R490C, p.R302Q and p.V364M affect the oligomerization of the enzyme. In contrast, p.M70T, p.D81G, p.L337P and p.T343A affect regions near the di-zinc centre and the substrate binding site. The p.S379R and p.L7V mutations were likely to cause structural destabilization and protein misfolding. Four mutations were identified in multiple unrelated DHP patients, indicating that DHP deficiency may be more common than anticipated.


Assuntos
Amidoidrolases/química , Amidoidrolases/genética , Doenças Metabólicas/genética , Adolescente , Adulto , Amidoidrolases/deficiência , Amidoidrolases/metabolismo , Criança , Pré-Escolar , Feminino , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Doenças Metabólicas/enzimologia , Modelos Biológicos , Modelos Moleculares , Fenótipo , Estabilidade Proteica , Estrutura Secundária de Proteína , Relação Estrutura-Atividade , Adulto Jovem
13.
J Hepatol ; 55(2): 426-34, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21147182

RESUMO

BACKGROUND & AIMS: Hyperammonemia is a frequent side-effect of valproic acid (VPA) therapy, which points to an imbalance between ammoniagenesis and ammonia disposal via the urea cycle. The impairment of this liver-specific metabolic pathway induced either by primary genetic defects or by secondary causes, namely associated with drugs administration, may result in accumulation of ammonia. To elucidate the mechanisms which underlie VPA-induced hyperammonemia, the aim of this study was to evaluate the effect of both VPA and its reactive intermediate, valproyl-CoA (VP-CoA), on the synthesis of N-acetylglutamate (NAG), a prime metabolite activator of the urea cycle. METHODS: The amount of NAG in livers of rats treated with VPA was quantified by HPLC-MS/MS. The NAG synthase (NAGS) activity was evaluated in vitro in rat liver mitochondria, and the effect of both VPA and VP-CoA was characterized. RESULTS: The present results clearly show that VP-CoA is a stronger inhibitor of NAGS activity in vitro than the parent drug VPA. The hepatic levels of NAG were significantly reduced in VPA-treated rats as compared with control tissues. CONCLUSIONS: These data strongly suggest that the hyperammonemia observed in patients under VPA treatment may result from a direct inhibition of the NAGS activity by VP-CoA. The subsequent reduced availability of NAG will impair the flux through the urea cycle and compromise the major role of this pathway in ammonia detoxification.


Assuntos
Acil Coenzima A/metabolismo , Aminoácido N-Acetiltransferase/antagonistas & inibidores , Hiperamonemia/induzido quimicamente , Hiperamonemia/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Ácido Valproico/toxicidade , Acil Coenzima A/toxicidade , Amônia/sangue , Animais , Anticonvulsivantes/toxicidade , Citrulina/biossíntese , Modelos Animais de Doenças , Glutamatos/metabolismo , Glutamina/sangue , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Ratos , Ratos Wistar , Ureia/sangue , Ureia/metabolismo
14.
Drug Metab Dispos ; 39(7): 1155-60, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21430231

RESUMO

Many biological systems including the oxidative catabolic pathway for branched-chain amino acids (BCAAs) are affected in vivo by valproate therapy. In this study, we investigated the potential effect of valproic acid (VPA) and some of its metabolites on the metabolism of BCAAs. In vitro studies were performed using isovaleryl-CoA dehydrogenase (IVD), isobutyryl-CoA dehydrogenase (IBD), and short branched-chain acyl-CoA dehydrogenase (SBCAD), enzymes involved in the degradation pathway of leucine, valine, and isoleucine. The enzymatic activities of the three purified human enzymes were measured using optimized high-performance liquid chromatography procedures, and the respective kinetic parameters were determined in the absence and presence of VPA and the corresponding CoA and dephosphoCoA conjugates. Valproyl-CoA and valproyl-dephosphoCoA inhibited IVD activity significantly by a purely competitive mechanism with K(i) values of 74 ± 4 and 170 ± 12 µM, respectively. IBD activity was not affected by any of the tested VPA esters. However, valproyl-CoA did inhibit SBCAD activity by a purely competitive mechanism with a K(i) of 249 ± 29 µM. In addition, valproyl-dephosphoCoA inhibited SBCAD activity via a distinct mechanism (K(i) = 511 ± 96 µM) that appeared to be of the mixed type. Furthermore, we show that both SBCAD and IVD are active, using valproyl-CoA as a substrate. The catalytic efficiency of SBCAD turned out to be much higher than that of IVD, demonstrating that SBCAD is the most probable candidate for the first dehydrogenation step of VPA ß-oxidation. Our data explain some of the effects of valproate on the branched-chain amino acid metabolism and shed new light on the biotransformation pathway of valproate.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Butiril-CoA Desidrogenase/metabolismo , Isovaleril-CoA Desidrogenase/metabolismo , Ácido Valproico/metabolismo , Cromatografia Líquida de Alta Pressão , Oxirredução , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
15.
J Inherit Metab Dis ; 34(1): 159-64, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21110228

RESUMO

We report on three patients (two siblings and one unrelated) presenting in infancy with progressive muscle weakness and paralysis of the diaphragm. Metabolic studies revealed a profile of plasma acylcarnitines and urine organic acids suggestive of a mild form of the multiple acyl-CoA dehydrogenation defect (MADD, ethylmalonic/adipic acid syndrome). Subsequently, a profound flavin deficiency in spite of a normal dietary riboflavin intake was established in the plasma of all three children, suggesting a riboflavin transporter defect. Genetic analysis of these patients demonstrated mutations in the C20orf54 gene which encodes the human homolog of a rat riboflavin transporter. This gene was recently implicated in the Brown-Vialetto-Van Laere syndrome, a rare neurological disorder which may either present in infancy with neurological deterioration with hypotonia, respiratory insufficiency and early death, or later in life with deafness and progressive ponto-bulbar palsy. Supplementation of riboflavin rapidly improved the clinical symptoms as well as the biochemical abnormalities in our patients, demonstrating that high dose riboflavin is a potential treatment for the Brown-Vialetto-Van Laere syndrome as well as for the Fazio Londe syndrome which is considered to be the same disease entity without the deafness.


Assuntos
Paralisia Bulbar Progressiva/genética , Proteínas de Membrana Transportadoras/genética , Erros Inatos do Metabolismo/diagnóstico , Erros Inatos do Metabolismo/terapia , Deficiência Múltipla de Acil Coenzima A Desidrogenase/diagnóstico , Riboflavina/metabolismo , Paralisia Bulbar Progressiva/complicações , Paralisia Bulbar Progressiva/diagnóstico , Paralisia Bulbar Progressiva/terapia , Criança , Diagnóstico Diferencial , Feminino , Perda Auditiva Neurossensorial/complicações , Perda Auditiva Neurossensorial/diagnóstico , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/terapia , Humanos , Lactente , Masculino , Erros Inatos do Metabolismo/genética , Deficiência Múltipla de Acil Coenzima A Desidrogenase/genética , Irmãos
16.
J Inherit Metab Dis ; 34(3): 677-94, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21431622

RESUMO

Glutaric aciduria type I (synonym, glutaric acidemia type I) is a rare organic aciduria. Untreated patients characteristically develop dystonia during infancy resulting in a high morbidity and mortality. The neuropathological correlate is striatal injury which results from encephalopathic crises precipitated by infectious diseases, immunizations and surgery during a finite period of brain development, or develops insidiously without clinically apparent crises. Glutaric aciduria type I is caused by inherited deficiency of glutaryl-CoA dehydrogenase which is involved in the catabolic pathways of L-lysine, L-hydroxylysine and L-tryptophan. This defect gives rise to elevated glutaric acid, 3-hydroxyglutaric acid, glutaconic acid, and glutarylcarnitine which can be detected by gas chromatography/mass spectrometry (organic acids) or tandem mass spectrometry (acylcarnitines). Glutaric aciduria type I is included in the panel of diseases that are identified by expanded newborn screening in some countries. It has been shown that in the majority of neonatally diagnosed patients striatal injury can be prevented by combined metabolic treatment. Metabolic treatment that includes a low lysine diet, carnitine supplementation and intensified emergency treatment during acute episodes of intercurrent illness should be introduced and monitored by an experienced interdisciplinary team. However, initiation of treatment after the onset of symptoms is generally not effective in preventing permanent damage. Secondary dystonia is often difficult to treat, and the efficacy of available drugs cannot be predicted precisely in individual patients. The major aim of this revision is to re-evaluate the previous diagnostic and therapeutic recommendations for patients with this disease and incorporate new research findings into the guideline.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/terapia , Encefalopatias Metabólicas/diagnóstico , Encefalopatias Metabólicas/terapia , Guias de Prática Clínica como Assunto , Algoritmos , Erros Inatos do Metabolismo dos Aminoácidos/complicações , Encefalopatias Metabólicas/complicações , Serviços Médicos de Emergência/métodos , Glutaril-CoA Desidrogenase/deficiência , Humanos , Recém-Nascido , Programas de Rastreamento/métodos , Monitorização Fisiológica/métodos , Triagem Neonatal/métodos , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/terapia
17.
J Pediatr ; 156(1): 121-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19800078

RESUMO

OBJECTIVE: To gain insight into the pathophysiological and clinical consequences of short-chain acyl-coenzyme A dehydrogenase deficiency (SCADD). STUDY DESIGN: A retrospective study of 15 fasting and 6 fat-loading tests in 15 Dutch patients with SCADD, divided into 3 genotype groups. Metabolic and endocrinologic measurements and the biochemical characteristics of SCADD, ethylmalonic acid (EMA), and C4-carnitine were studied. RESULTS: Three patients had development of hypoglycemia during fasting; all of these had originally presented with hypoglycemia. Metabolic and endocrinologic measurements remained normal during all tests. The EMA excretion increased in response to fasting and fat loading, and plasma C4-carnitine remained stable. Test results did not differ between the 3 genotype groups. CONCLUSIONS: The metabolic profiles of the 3 patients with development of hypoglycemia resemble idiopathic ketotic hypoglycemia. Because hypoglycemia generally requires a metabolic work-up and because SCADD is relatively prevalent, SCADD may well be diagnosed coincidently, thus being causally unrelated to the hypoglycemia. If SCADD has any other pathologic consequences, the accumulation of potentially toxic metabolites such as EMA is most likely involved. However, the results of our study indicate that there is no clear pathophysiological significance, irrespective of genotype, supporting the claim that SCADD is not suited for inclusion in newborn screening programs.


Assuntos
Butiril-CoA Desidrogenase/deficiência , Erros Inatos do Metabolismo/fisiopatologia , Adolescente , Criança , Pré-Escolar , Gorduras Insaturadas na Dieta/administração & dosagem , Jejum , Feminino , Humanos , Hipoglicemia/enzimologia , Lactente , Recém-Nascido , Masculino , Triagem Neonatal , Óleos de Plantas/administração & dosagem , Estudos Retrospectivos , Óleo de Girassol
18.
J Neurol Neurosurg Psychiatry ; 81(3): 310-2, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20185470

RESUMO

Peroxisomal acyl-coenzyme A oxidase deficiency (formerly also called pseudoneonatal adrenoleucodystrophy) is a disorder of peroxisomal fatty acid oxidation with a severe presentation. Most patients present at birth or in early infancy, and the mean age of death was 5 years in a recently published cohort of 22 patients. Brain imaging shows a progressive leucodystrophy. The authors report here the first adult patients (two siblings, 52 and 55 years old) with peroxisomal acyl-coenzyme A oxidase deficiency with a remarkably mild clinical presentation. Magnetic resonance brain imaging revealed profound atrophy of the brainstem and cerebellum.


Assuntos
Tronco Encefálico/patologia , Cerebelo/patologia , Oxirredutases/deficiência , Transtornos Peroxissômicos/diagnóstico , Acil-CoA Oxidase , Atrofia , Progressão da Doença , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Exame Neurológico , Oxirredutases/genética , Transtornos Peroxissômicos/genética , Fenótipo
19.
Pediatr Res ; 67(3): 304-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19952864

RESUMO

Short-chain acyl-CoA dehydrogenase deficiency (SCADD) is an inborn error, biochemically characterized by increased plasma butyrylcarnitine (C4-C) concentration and increased ethylmalonic acid (EMA) excretion and caused by rare mutations and/or common gene variants in the SCAD encoding gene. Although its clinical relevance is not clear, SCADD is included in most US newborn screening programs. Riboflavin, the precursor of flavin adenine dinucleotide (FAD, cofactor), might be effective for treating SCADD. We assessed the FAD status and evaluated the effects of riboflavin treatment in a prospective open-label cohort study involving 16 patients with SCADD, subdivided into mutation/mutation (mut/mut), mutation/variant (mut/var), and variant/variant (var/var) genotype groups. Blood FAD levels were normal in all patients before therapy, but significantly lower in the mut/var and var/var groups compared with the mut/mut group. Riboflavin treatment resulted in a decrease in EMA excretion in the mut/var group and in a subjective clinical improvement in four patients from this group. However, this improvement persisted after stopping treatment. These results indicate that high-dose riboflavin treatment may improve the biochemical features of SCADD, at least in patients with a mut/var genotype and low FAD levels. As our study could not demonstrate a clinically relevant effect of riboflavin, general use of riboflavin cannot be recommended.


Assuntos
Butiril-CoA Desidrogenase/deficiência , Flavina-Adenina Dinucleotídeo/sangue , Erros Inatos do Metabolismo Lipídico/tratamento farmacológico , Riboflavina/administração & dosagem , Complexo Vitamínico B/administração & dosagem , Adolescente , Biomarcadores/sangue , Biomarcadores/urina , Butiril-CoA Desidrogenase/genética , Carnitina/análogos & derivados , Carnitina/sangue , Criança , Pré-Escolar , Feminino , Flavina-Adenina Dinucleotídeo/urina , Predisposição Genética para Doença , Humanos , Lactente , Erros Inatos do Metabolismo Lipídico/sangue , Erros Inatos do Metabolismo Lipídico/enzimologia , Erros Inatos do Metabolismo Lipídico/genética , Erros Inatos do Metabolismo Lipídico/urina , Masculino , Malonatos/urina , Mutação , Fenótipo , Estudos Prospectivos , Resultado do Tratamento
20.
J Inherit Metab Dis ; 33 Suppl 3: S443-53, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20978941

RESUMO

Ethylmalonic encephalopathy (EE) is a rare metabolic disorder caused by dysfunction of ETHE1, a mitochondrial dioxygenase involved in hydrogen sulfide (H2S) detoxification. Patients present in infancy with psychomotor retardation, chronic diarrhea, orthostatic acrocyanosis and relapsing petechiae. High levels of lactic acid, ethymalonic acid (EMA) and methylsuccinic acid (MSA) are detected in body fluids. Several pathways may contribute to the pathophysiology, including isoleucine, methionine and fatty acid metabolism. We report on a 15-month-old male presenting with typical EE associated with a homozygous ETHE1 mutation. We investigated oral isoleucine (150 mg/kg), methionine (100 mg/kg), fatty acid loading tests and isoleucine-restricted diet (200 mg/day) for any effects on several metabolic parameters. Before loading tests or specific dietary interventions, EMA, C4-C5 acylcarnitines and most acylglycines were elevated, indicating functional deficiency of short chain acyl-CoA (SCAD) as well as all branched acyl-CoA dehydrogenases. Excretion of EMA and n-butyrylglycine increased following each of the loads, and isoleucine led to increased levels of derivative metabolites. An isoleucine-restricted diet for 8 days corrected some of the abnormalities but led to no obvious clinical improvement and only partial effects on EMA. A principal component analysis supports the inference that these dietary conditions have consistent effects on the global metabolic profile. Our results suggest that multiple pathways modulate EMA levels in EE. They might all interact with H2S toxicity. Prolonged dietary interventions involving the restriction for branched aminoacids, fatty acids and methionine could be discussed as auxiliary therapeutical strategies in EE.


Assuntos
Encefalopatias Metabólicas Congênitas/enzimologia , Proteínas Mitocondriais/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Púrpura/enzimologia , Aminoácidos/uso terapêutico , Biomarcadores/sangue , Biomarcadores/urina , Encefalopatias Metabólicas Congênitas/diagnóstico , Encefalopatias Metabólicas Congênitas/dietoterapia , Encefalopatias Metabólicas Congênitas/genética , Dieta com Restrição de Proteínas , Suplementos Nutricionais , Predisposição Genética para Doença , Homozigoto , Humanos , Lactente , Masculino , Malonatos/sangue , Malonatos/urina , Proteínas Mitocondriais/genética , Mutação , Proteínas de Transporte Nucleocitoplasmático/genética , Fenótipo , Análise de Componente Principal , Púrpura/diagnóstico , Púrpura/dietoterapia , Púrpura/genética , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA