Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
ACS Chem Biol ; 19(7): 1544-1553, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38915184

RESUMO

Glutaric Aciduria Type 1 (GA1) is a serious inborn error of metabolism with no pharmacological treatments. A novel strategy to treat this disease is to divert the toxic biochemical intermediates to less toxic or nontoxic metabolites. Here, we report a putative novel target, succinyl-CoA:glutarate-CoA transferase (SUGCT), which we hypothesize suppresses the GA1 metabolic phenotype through decreasing glutaryl-CoA and the derived 3-hydroxyglutaric acid. SUGCT is a type III CoA transferase that uses succinyl-CoA and glutaric acid as substrates. We report the structure of SUGCT, develop enzyme- and cell-based assays, and identify valsartan and losartan carboxylic acid as inhibitors of the enzyme in a high-throughput screen of FDA-approved compounds. The cocrystal structure of SUGCT with losartan carboxylic acid revealed a novel pocket in the active site and further validated the high-throughput screening approach. These results may form the basis for the future development of new pharmacological intervention to treat GA1.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos , Encefalopatias Metabólicas , Humanos , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Encefalopatias Metabólicas/tratamento farmacológico , Encefalopatias Metabólicas/metabolismo , Encefalopatias Metabólicas/enzimologia , Glutaratos/metabolismo , Glutaratos/química , Losartan/farmacologia , Losartan/química , Coenzima A-Transferases/metabolismo , Coenzima A-Transferases/antagonistas & inibidores , Coenzima A-Transferases/genética , Coenzima A-Transferases/química , Valsartana , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/química , Cristalografia por Raios X , Domínio Catalítico , Acil Coenzima A/metabolismo , Acil Coenzima A/química , Modelos Moleculares , Ensaios de Triagem em Larga Escala , Glutaril-CoA Desidrogenase/deficiência
2.
Int J Mol Sci ; 24(17)2023 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-37685964

RESUMO

Glutaric acidemia type 1 (GA1) is a neurotoxic metabolic disorder due to glutaryl-CoA dehydrogenase (GCDH) deficiency. The high number of missense variants associated with the disease and their impact on GCDH activity suggest that disturbed protein conformation can affect the biochemical phenotype. We aimed to elucidate the molecular basis of protein loss of function in GA1 by performing a parallel analysis in a large panel of GCDH missense variants using different biochemical and biophysical methodologies. Thirteen GCDH variants were investigated in regard to protein stability, hydrophobicity, oligomerization, aggregation, and activity. An altered oligomerization, loss of protein stability and solubility, as well as an augmented susceptibility to aggregation were observed. GA1 variants led to a loss of enzymatic activity, particularly when present at the N-terminal domain. The reduced cellular activity was associated with loss of tetramerization. Our results also suggest a correlation between variant sequence location and cellular protein stability (p < 0.05), with a more pronounced loss of protein observed with variant proximity to the N-terminus. The broad panel of variant-mediated conformational changes of the GCDH protein supports the classification of GA1 as a protein-misfolding disorder. This work supports research toward new therapeutic strategies that target this molecular disease phenotype.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos , Encefalopatias Metabólicas , Glutaril-CoA Desidrogenase , Glutaril-CoA Desidrogenase/química , Glutaril-CoA Desidrogenase/genética , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/genética , Dobramento de Proteína , Mutação de Sentido Incorreto , Domínios Proteicos , Humanos , Estabilidade Enzimática , Solubilidade
3.
Eur J Paediatr Neurol ; 39: 49-58, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35662016

RESUMO

AIM: To evaluate the pathogenic variants in GCDH gene and to assess the neurodevelopmental outcomes in children with Glutaric aciduria type 1 (GA-1). METHOD: Cross-sectional observational study between January 2019 and June 2020 in consecutive North Indian children with a clinical and biochemical suspicion of GA-1. Variants in the coding regions of GCDH gene were identified through Sanger sequencing. Neurodevelopmental and quality of life assessment was done using standardized scales. RESULTS: 24 children with GA-1 were identified. The median age at diagnosis was 12 months and the median delay in diagnosis was 3 months. Genetic analysis was done in 14 cases. It revealed 12 variants (11 missense and one nonsense) from 13 patients. Most of the pathogenic variants were in exon 9 and exon 5. Three novel variants were identified in three patients: two missense variants c.169G > A (p.Glu57Lys), c.1048T > C (p.Cys350Arg) and one nonsense variant c.331C > T (p.Lys111Ter). On neurodevelopmental assessment, majority of children with GA-1 were non ambulatory (62.5%), had limited hand skills (58.3%) and impaired communication (58.3%). Overall, poor global development was noted in 43.7%. A pre-existing developmental delay was significantly associated with impaired communication skills (p = 0.03), and the number of episodes of encephalopathy were significantly associated with impaired gross motor skill (p = 0.02). Presence of encephalopathy was significantly associated with poor performance in social emotional (p = 0.01) and cognitive (p = 0.03) domains of Developmental Profile-III scale and development of severe dystonia (p = 0.01). CONCLUSION: Our findings highlight the clinical, biochemical, radiological and genetic spectrum of GA-1 in children in North India and report the presence of novel pathogenic variations.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos , Encefalopatias Metabólicas , Glutaril-CoA Desidrogenase , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/genética , Criança , Estudos Transversais , Glutaril-CoA Desidrogenase/química , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/genética , Humanos , Qualidade de Vida
4.
Protein Sci ; 30(10): 1995-2008, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34288185

RESUMO

CAD is a 1.5 MDa particle formed by hexameric association of a 250 kDa protein divided into different enzymatic domains, each catalyzing one of the initial reactions for de novo biosynthesis of pyrimidine nucleotides: glutaminase-dependent Carbamoyl phosphate synthetase, Aspartate transcarbamoylase, and Dihydroorotase. The pathway for de novo pyrimidine synthesis is essential for cell proliferation and is conserved in all living organisms, but the covalent linkage of the first enzymatic activities into a multienzymatic CAD particle is unique to animals. In other organisms, these enzymatic activities are encoded as monofunctional proteins for which there is abundant structural and biochemical information. However, the knowledge about CAD is scarce and fragmented. Understanding CAD requires not only to determine the three-dimensional structures and define the catalytic and regulatory mechanisms of the different enzymatic domains, but also to comprehend how these domains entangle and work in a coordinated and regulated manner. This review summarizes significant progress over the past 10 years toward the characterization of CAD's architecture, function, regulatory mechanisms, and cellular compartmentalization, as well as the recent finding of a new and rare neurometabolic disorder caused by defects in CAD activities.


Assuntos
Aspartato Carbamoiltransferase , Encefalopatias Metabólicas/enzimologia , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante) , Di-Hidro-Orotase , Animais , Aspartato Carbamoiltransferase/química , Aspartato Carbamoiltransferase/metabolismo , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/química , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/metabolismo , Di-Hidro-Orotase/química , Di-Hidro-Orotase/metabolismo , Humanos , Domínios Proteicos
5.
J Cell Biochem ; 120(3): 3367-3372, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30203563

RESUMO

Glutaric acidemia type I (GA-1) is an inborn error of metabolism due to deficiency of glutaryl-CoA dehydrogenase (GCDH), which catalyzes the conversion of glutaryl-CoA to crotonyl-CoA. GA-1 occurs in about 1 in 100 000 infants worldwide. The GCDH gene is on human chromosome 19p13.2, spans about 7 kb and comprises 11 exons and 10 introns. Tandem mass spectrometry (MS/MS) was used for clinical diagnosis in a proband from Iran with GA-1. Sanger sequencing was performed using primers specific for coding exons and exon-intron flanking regions of the GCDH gene in the proband. Cosegregation analysis and in silico assessment were performed to confirm the pathogenicity of the candidate variant. A novel homozygous missense variant c.1147C > A (p.Arg383Ser) in exon 11 of GCDH was identified. Examination of variant through in silico software tools determines its deleterious effect on protein in terms of function and stability. The variant cosegregates with the disease in family. In this study, the clinical and molecular aspects of GA-1 were investigated, which showed one novel mutation in the GCDH gene in an Iranian patient. The variant is categorized as pathogenic according to the the guideline of the American College of Medical Genetics and Genomics (ACMG) for variant interpretation. This mutation c.1147C > A (p.Arg383Ser) may also be prevalent among Iranian populations.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/genética , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/genética , Homozigoto , Mutação de Sentido Incorreto , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Encefalopatias Metabólicas/patologia , Feminino , Humanos , Lactente , Masculino , Linhagem
6.
J Diabetes Res ; 2018: 8431249, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29744369

RESUMO

Diabetic encephalopathy is a complication of diabetes mellitus characterized by impaired cognitive functions. Protein kinase C (PKC) isoforms are rarely reported on diabetic encephalopathy, although they have been believed to play crucial roles in other diabetic complications. In this study, streptozotocin- (STZ-) induced diabetic mice were found to exhibit learning and memory deficits in the Morris water maze test. Meanwhile, the expression of cPKCßII, nPKCε, and cPKCγ did not change in the hippocampus, cortex, and striatum at 2 and 8 weeks after STZ injection. The nPKCε translocation to the membrane, where it is activated, was not altered in the above brain regions at 2 and 8 weeks after STZ injection. Nevertheless, cPKCßII translocation to the membrane was significantly decreased in the cortex and hippocampus at 8 weeks after STZ injection. The translocation of cPKCγ from the cytosol to the membrane was remarkably decreased in the hippocampus at 2 and 8 weeks and in the cortex and striatum at 8 weeks after STZ injection. In addition, deletion of cPKCγ aggravated the impairment of spatial learning and memory. In conclusion, our results suggest that the decrease in the activity of cPKCßII and cPKCγ, especially cPKCγ, may play key roles in the pathogenesis of diabetic encephalopathy.


Assuntos
Encefalopatias Metabólicas/enzimologia , Complicações do Diabetes/enzimologia , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Tipo 1/enzimologia , Isoformas de Proteínas/metabolismo , Proteína Quinase C/metabolismo , Animais , Córtex Cerebral/enzimologia , Hipocampo/enzimologia , Masculino , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Camundongos
7.
Mol Neurobiol ; 54(6): 4795-4805, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27510504

RESUMO

Patients affected by glutaric aciduria type I (GA-I) show progressive cortical leukoencephalopathy whose pathogenesis is poorly known. In the present work, we exposed cortical astrocytes of wild-type (Gcdh +/+ ) and glutaryl-CoA dehydrogenase knockout (Gcdh -/- ) mice to the oxidative stress inducer menadione and measured mitochondrial bioenergetics, redox homeostasis, and cell viability. Mitochondrial function (MTT and JC1-mitochondrial membrane potential assays), redox homeostasis (DCFH oxidation, nitrate and nitrite production, GSH concentrations and activities of the antioxidant enzymes SOD and GPx), and cell death (propidium iodide incorporation) were evaluated in primary cortical astrocyte cultures of Gcdh +/+ and Gcdh -/- mice unstimulated and stimulated by menadione. We also measured the pro-inflammatory response (TNFα levels, IL1-ß and NF-ƙB) in unstimulated astrocytes obtained from these mice. Gcdh -/- mice astrocytes were more vulnerable to menadione-induced oxidative stress (decreased GSH concentrations and altered activities of the antioxidant enzymes), mitochondrial dysfunction (decrease of MTT reduction and JC1 values), and cell death as compared with Gcdh +/+ astrocytes. A higher inflammatory response (TNFα, IL1-ß and NF-ƙB) was also observed in Gcdh -/- mice astrocytes. These data indicate a higher susceptibility of Gcdh -/- cortical astrocytes to oxidative stress and mitochondrial dysfunction, probably leading to cell death. It is presumed that these pathomechanisms may contribute to the cortical leukodystrophy observed in GA-I patients.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/patologia , Astrócitos/patologia , Encefalopatias Metabólicas/patologia , Córtex Cerebral/patologia , Glutaril-CoA Desidrogenase/deficiência , Mitocôndrias/metabolismo , Degeneração Neural/patologia , Estresse Oxidativo/efeitos dos fármacos , Vitamina K 3/toxicidade , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Animais , Antioxidantes/metabolismo , Astrócitos/efeitos dos fármacos , Encefalopatias Metabólicas/enzimologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fluoresceínas/metabolismo , Glutationa Peroxidase/metabolismo , Mediadores da Inflamação/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Degeneração Neural/enzimologia , Óxido Nítrico/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
9.
Neuroradiology ; 58(7): 697-703, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26993811

RESUMO

INTRODUCTION: Adenosine kinase deficiency (ADK deficiency) is a recently described disorder of methionine and adenosine metabolism resulting in a neurological phenotype with developmental delay, muscular hypotonia, and epilepsy as well as variable systemic manifestations. The underlying neuropathology is poorly understood. We have investigated MRI and (1)H-MRS changes in ADK deficiency in order to better understand the in vivo neuropathologic changes of ADK deficiency. METHODS: Systematic evaluation of 21 MRIs from eight patients (age range 9 days-14.6 years, mean 3.9 years, median 2.7 years) including diffusion-weighted imaging in six and (1)H-MRS in five patients. RESULTS: Brain maturation was delayed in the neonatal period and in infancy (6/6), but ultimately complete. White matter changes occurring in five of eight patients were discrete, periventricular, and unspecific (4/5), or diffuse with sparing of optic radiation, corona radiata, and pyramidal tracts (1/5). Choline was low in white matter spectra (3/3), while there was no indication of low creatine in white matter or basal ganglia (5/5), and diffusion was variably decreased or increased. Central tegmental tract hyperintensity was a common finding (6/8), as was supratentorial atrophy (6/8). CONCLUSIONS: MRI changes in ADK deficiency consist of delayed but ultimately completed brain maturation with later onset of mostly unspecific white matter changes and potentially transient central tegmental tract hyperintensity. Immaturity on neonatal MRI is consistent with prenatal onset of disease and reduced choline with lower membrane turnover resulting in delayed myelination and deficient myelin maintenance.


Assuntos
Adenosina Quinase/deficiência , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Imageamento por Ressonância Magnética/métodos , Espectroscopia de Prótons por Ressonância Magnética/métodos , Adenosina Quinase/metabolismo , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Imagem Molecular/métodos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
10.
Clin Chim Acta ; 453: 75-9, 2016 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-26656312

RESUMO

Glutaric aciduria type I (GA-I) is a rare autosomal recessive metabolic disorder caused by deficiency of glutaryl-CoA dehydrogenase (GCDH), leading to an abnormal metabolism of lysine, hydroxylysine and tryptophan. It results in accumulations of glutaric acid, 3-hydroxyglutaric acid and glutaconic acid. Clinical features include the sudden onset of encephalopathy, hypotonia and macrocephaly usually before age 18months. Here we report five cases of GA-I confirmed with mutation analysis. GCDH gene mutations were identified in all five probands with GA-I. Three of them had compound heterozygous mutations and two had homozygous mutations. Mutations of two alleles (c.334G>T and IVS11-11A>G) were novel and both of them were confirmed to be splice site mutations by reverse transcription PCR.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/genética , Encefalopatias Metabólicas/diagnóstico , Encefalopatias Metabólicas/genética , Glutaril-CoA Desidrogenase/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Sequência de Bases , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/metabolismo , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Glutaratos/metabolismo , Glutaril-CoA Desidrogenase/química , Glutaril-CoA Desidrogenase/genética , Glutaril-CoA Desidrogenase/metabolismo , Heterozigoto , Humanos , Masculino , Modelos Moleculares , Mutação , Conformação Proteica
11.
Mol Neurobiol ; 53(9): 6459-6475, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26607633

RESUMO

We investigated the effects of an acute intrastriatal QUIN administration on cellular redox and bioenergetics homeostasis, as well as on important signaling pathways in the striatum of wild-type (Gcdh +/+ , WT) and knockout mice for glutaryl-CoA dehydrogenase (Gcdh -/- ) fed a high lysine (Lys, 4.7 %) chow. QUIN increased lactate release in both Gcdh +/+ and Gcdh -/- mice and reduced the activities of complex IV and creatine kinase only in the striatum of Gcdh -/- mice. QUIN also induced lipid and protein oxidative damage and increased the generation of reactive nitrogen species, as well as the activities of the antioxidant enzymes glutathione peroxidase, superoxide dismutase 2, and glutathione-S-transferase in WT and Gcdh -/- animals. Furthermore, QUIN induced DCFH oxidation (reactive oxygen species production) and reduced GSH concentrations (antioxidant defenses) in Gcdh -/- . An early increase of Akt and phospho-Erk 1/2 in the cytosol and Nrf2 in the nucleus was also observed, as well as a decrease of cytosolic Keap1caused by QUIN, indicating activation of the Nrf2 pathway mediated by Akt and phospho-Erk 1/2, possibly as a compensatory protective mechanism against the ongoing QUIN-induced toxicity. Finally, QUIN increased NF-κB and diminished IκBα expression, evidencing a pro-inflammatory response. Our data show a disruption of energy and redox homeostasis associated to inflammation induced by QUIN in the striatum of Gcdh -/- mice submitted to a high Lys diet. Therefore, it is presumed that QUIN may possibly contribute to the pathophysiology of striatal degeneration in children with glutaric aciduria type I during inflammatory processes triggered by infections or vaccinations.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/patologia , Metabolismo Energético/efeitos dos fármacos , Glutaril-CoA Desidrogenase/deficiência , Estresse Oxidativo/efeitos dos fármacos , Ácido Quinolínico/toxicidade , Transdução de Sinais/efeitos dos fármacos , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Animais , Antioxidantes/metabolismo , Encefalopatias Metabólicas/metabolismo , Dieta , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fluoresceínas/metabolismo , Glutaril-CoA Desidrogenase/metabolismo , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Lactatos/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Malondialdeído/metabolismo , Camundongos Knockout , Modelos Biológicos , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Neostriado/patologia , Nitratos/metabolismo , Nitritos/metabolismo , Oxirredução , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ácido Quinolínico/administração & dosagem , Compostos de Sulfidrila/metabolismo , Superóxido Dismutase/metabolismo
12.
Genet Mol Res ; 14(4): 14130-5, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26535729

RESUMO

Pyridoxal 5'-phosphate (PLP)-responsive epilepsy is a rare autosomal recessive epileptic disorder caused by deficiency of pyridox(am)-ne 5'-phosphate oxidase (PNPO). Neonatal onset seizures in PLP responsive epilepsy are usually resistant to common anticonvulsants and pyridoxine, but respond to PLP. Various PNPO mutations are associated with this disorder. In this report, we have described a case of a female baby with neonatal onset seizures responding to PLP. Exome sequencing revealed that the patient was compound heterozygous for pathogenic mutations [c.546+1G>A (IVS5+1 G>A) and c.620delG (p.G207VfsX215)] in the PNPO gene. The c.546+1G>A was inherited from the mother while the c.620delG was inherited from the father. Both mutations were absent in 122 unrelated Thai controls. The results of this study indicated the presence of two newly identified mutations in this Thai patient with PLP-responsive epilepsy for the first time, expanding the mutational spectrum of PNPO.


Assuntos
Encefalopatias Metabólicas/genética , Hipóxia-Isquemia Encefálica/genética , Fosfato de Piridoxal/uso terapêutico , Piridoxaminafosfato Oxidase/deficiência , Piridoxaminafosfato Oxidase/genética , Convulsões/genética , Encefalopatias Metabólicas/enzimologia , Feminino , Humanos , Hipóxia-Isquemia Encefálica/enzimologia , Recém-Nascido , Mutação , Piridoxaminafosfato Oxidase/efeitos dos fármacos , Convulsões/tratamento farmacológico , Convulsões/enzimologia , Tailândia
13.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 32(2): 187-91, 2015 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-25863083

RESUMO

OBJECTIVE: To report on clinical features of four patients with glutaric academia type Ⅰ (GA-1) and mutations identified in the glutaryl-CoA dehydrogenase (GCDH) gene. METHODS: All of the patients underwent magnetic resonance imaging (MRI) analysis. Blood acylcarnitine and urine organic acid were analyzed with tandem mass spectrometry and gas chromatographic mass spectrometry. Genomic DNA was extracted from peripheral blood samples. The 11 exons and flanking sequences of the GCDH gene were amplified with PCR and subjected to direct DNA sequencing. RESULTS: Mutations of the GCDH gene were identified in all of the patients. Three had homozygous mutations. A recurrent mutation, IVS10-2A>C, was found in the four unrelated families, while the mutation of c.245G>C (p.Arg82Pro) was novel. CONCLUSION: IVS10-2A>C is likely a founder mutation for Chinese population in Wenzhou.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Povo Asiático/genética , Encefalopatias Metabólicas/enzimologia , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/genética , Mutação Puntual , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico por imagem , Erros Inatos do Metabolismo dos Aminoácidos/genética , Sequência de Aminoácidos , Sequência de Bases , Encefalopatias Metabólicas/diagnóstico por imagem , Encefalopatias Metabólicas/genética , Análise Mutacional de DNA , Éxons , Feminino , Glutaril-CoA Desidrogenase/química , Glutaril-CoA Desidrogenase/metabolismo , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Dados de Sequência Molecular , Radiografia , Alinhamento de Sequência
14.
J Inherit Metab Dis ; 38(2): 265-72, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25214427

RESUMO

Inherited deficiencies of the L-lysine catabolic pathway cause glutaric aciduria type I and pyridoxine-dependent epilepsy. Dietary modulation of cerebral L-lysine metabolism is thought to be an important therapeutic intervention for these diseases. To better understand cerebral L-lysine degradation, we studied in mice the two known catabolic routes -- pipecolate and saccharopine pathways -- using labeled stable L-lysine and brain peroxisomes purified according to a newly established protocol. Experiments with labeled stable L-lysine show that cerebral L-pipecolate is generated along two pathways: i) a minor proportion retrograde after ε-deamination of L-lysine along the saccharopine pathway, and ii) a major proportion anterograde after α-deamination of L-lysine along the pipecolate pathway. In line with these findings, we observed only little production of saccharopine in the murine brain. L-pipecolate oxidation was only detectable in brain peroxisomes, but L-pipecolate oxidase activity was low (7 ± 2µU/mg protein). In conclusion, L-pipecolate is a major degradation product from L-lysine in murine brain generated by α-deamination of this amino acid.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/genética , Encéfalo/enzimologia , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/genética , Lisina/metabolismo , Ácidos Pipecólicos/metabolismo , Animais , Desaminação , Modelos Animais de Doenças , Predisposição Genética para Doença , Fígado/enzimologia , Lisina/análogos & derivados , Camundongos Knockout , Oxirredução , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Peroxissomos/enzimologia , Fenótipo
15.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 31(5): 608-11, 2014 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-25297592

RESUMO

OBJECTIVE: To review the clinical features of a families affected with glutaric acidemia type I (GA-1) and screen potential mutations in glutaryl-CoA dehydrogenase (GCDH) gene. METHODS: Clinical data of the patients and their family members was analyzed. Genomic DNA was extracted from peripheral blood samples. The 11 exons and flanking sequences of the GCDH gene were amplified with PCR and subjected to direct DNA sequencing. RESULTS: Two patients have manifested macrocephaly. Imaging analysis revealed arachnoid cyst and subdural effusion. The elder sister had encephalopathy crisis. The younger sister had significantly raised glutaric acid, whilst the elder sister was normal during the non-acute phase. Genetic analysis has revealed a homozygous c.1244-2A> C mutation of the GCDH gene in both patients. CONCLUSION: The clinical features and mutation of the GCDH gene have been delineated in a Chinese family affected with GA-1. The c.1244-2A> C mutation may be particularly common in the Chinese population.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/genética , Encefalopatias Metabólicas/genética , Predisposição Genética para Doença/genética , Glutaril-CoA Desidrogenase/genética , Mutação , Adolescente , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico por imagem , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Sequência de Bases , Encefalopatias Metabólicas/diagnóstico por imagem , Encefalopatias Metabólicas/enzimologia , China , Análise Mutacional de DNA , Saúde da Família , Feminino , Glutaril-CoA Desidrogenase/deficiência , Homozigoto , Humanos , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Radiografia
16.
Ann Clin Lab Sci ; 44(2): 213-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24795062

RESUMO

Mutations in the glutaryl-CoA dehydrogenase gene can result in Glutaric aciduria type 1(GA 1) by accumulation of glutaric acid, 3-hydroxyglutaric acid (3-OH-GA), and glutarylcarnitine (C5DC). GA 1 is characterized by macrocephaly, subdural hemorrhage (SDH), and dystonic movement disorder after acute encephalopathic crisis. We report a Korean patient with GA1 and a novel mutation. A 16-month-old boy presented with SDH, macrocephaly, and developmental delay. In the neurologic examination, the patient had mild axial hypotonia, but otherwise normal neurologic functions. The brain MRI showed large amounts of bilateral SDH and high signal intensity in both basal ganglia and thalamus. Metabolic screening tests detected highly elevated urinary GA levels but 3-OH-glutaric acid was normal. C5DC was 0.94 µM/L (reference range < 0.3 µM/L). The patient had compound heterozygous mutations of the GCDH gene: p.Arg257Gln (c.770G>A) and p.Cys308Arg (c.922T>C). p.Cys308Arg is a novel mutation; reports of p.Arg257Gln were also rare both in Caucasians and Asian populations. In summary, we hereby report one Korean patient with GA1 with clinical, biochemical, and radiologic characteristics confirmed by genetic analysis.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Povo Asiático/genética , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/genética , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/genética , Mutação de Sentido Incorreto/genética , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico por imagem , Sequência de Bases , Encéfalo/enzimologia , Encéfalo/patologia , Encefalopatias Metabólicas/diagnóstico por imagem , Pré-Escolar , Análise Mutacional de DNA , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Dados de Sequência Molecular , República da Coreia , Tomografia Computadorizada por Raios X
17.
PLoS One ; 9(3): e90477, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24594605

RESUMO

We determined mRNA expression of the ionotropic glutamate receptors NMDA (NR1, NR2A and NR2B subunits), AMPA (GluR2 subunit) and kainate (GluR6 subunit), as well as of the glutamate transporters GLAST and GLT1 in cerebral cortex and striatum of wild type (WT) and glutaryl-CoA dehydrogenase deficient (Gchh-/-) mice aged 7, 30 and 60 days. The protein expression levels of some of these membrane proteins were also measured. Overexpression of NR2A and NR2B in striatum and of GluR2 and GluR6 in cerebral cortex was observed in 7-day-old Gcdh-/-. There was also an increase of mRNA expression of all NMDA subunits in cerebral cortex and of NR2A and NR2B in striatum of 30-day-old Gcdh-/- mice. At 60 days of life, all ionotropic receptors were overexpressed in cerebral cortex and striatum of Gcdh-/- mice. Higher expression of GLAST and GLT1 transporters was also verified in cerebral cortex and striatum of Gcdh-/- mice aged 30 and 60 days, whereas at 7 days of life GLAST was overexpressed only in striatum from this mutant mice. Furthermore, high lysine intake induced mRNA overexpression of NR2A, NR2B and GLAST transcripts in striatum, as well as of GluR2 and GluR6 in both striatum and cerebral cortex of Gcdh-/- mice. Finally, we found that the protein expression of NR2A, NR2B, GLT1 and GLAST were significantly greater in cerebral cortex of Gcdh-/- mice, whereas NR2B and GLT1 was similarly enhanced in striatum, implying that these transcripts were translated into their products. These results provide evidence that glutamate receptor and transporter expression is higher in Gcdh-/- mice and that these alterations may be involved in the pathophysiology of GA I and possibly explain, at least in part, the vulnerability of striatum and cerebral cortex to injury in patients affected by GA I.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/patologia , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Encefalopatias Metabólicas/patologia , Córtex Cerebral/metabolismo , Glutaril-CoA Desidrogenase/deficiência , Neostriado/metabolismo , Receptores de Glutamato/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Sistema X-AG de Transporte de Aminoácidos/genética , Animais , Encefalopatias Metabólicas/enzimologia , Córtex Cerebral/patologia , Dieta , Feminino , Regulação da Expressão Gênica , Glutaril-CoA Desidrogenase/metabolismo , Lisina/metabolismo , Masculino , Camundongos , Neostriado/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Glutamato/genética
18.
PLoS One ; 9(2): e87715, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498361

RESUMO

Glutaric aciduria type 1 (GA1) is an inherited neurometabolic disorder caused by mutations in the GCDH gene encoding glutaryl-CoA dehydrogenase (GCDH), which forms homo- and heteromeric complexes in the mitochondrial matrix. GA1 patients are prone to the development of encephalopathic crises which lead to an irreversible disabling dystonic movement disorder. The clinical and biochemical manifestations of GA1 vary considerably and lack correlations to the genotype. Using an affinity chromatography approach we report here for the first time on the identification of mitochondrial proteins interacting directly with GCDH. Among others, dihydrolipoamide S-succinyltransferase (DLST) involved in the formation of glutaryl-CoA, and the ß-subunit of the electron transfer flavoprotein (ETFB) serving as electron acceptor, were identified as GCDH binding partners. We have adapted the yellow fluorescent protein-based fragment complementation assay and visualized the oligomerization of GCDH as well as its direct interaction with DLST and ETFB in mitochondria of living cells. These data suggest that GCDH is a constituent of multimeric mitochondrial dehydrogenase complexes, and the characterization of their interrelated functions may provide new insights into the regulation of lysine oxidation and the pathophysiology of GA1.


Assuntos
Aciltransferases/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Encefalopatias Metabólicas/enzimologia , Flavoproteínas Transferidoras de Elétrons/metabolismo , Glutaratos/metabolismo , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/metabolismo , Proteínas Mitocondriais/metabolismo , Western Blotting , Cromatografia de Afinidade , Humanos , Imunoprecipitação , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
19.
Neurology ; 81(9): 849-50, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23884036

RESUMO

Glutaric aciduria type I (GA-I) is an autosomal recessive disease caused by a deficiency of the mitochondrial enzyme glutaryl CoA dehydrogenase (GCDH). This metabolic block causes increased urinary concentrations of glutaric and 3-hydroxyglutaric acids. The accumulation and excretion of glutarylcarnitine esters leads to secondary carnitine deficiency. GA-I has an incidence of 1:30,000. The clinical hallmark of GA-I is an acute encephalopathic crisis, with bilateral striatal necrosis presented by severe dystonic dyskinetic disorder. Most patients have their first symptoms during infancy, but some have a less severe form of the disease and some may even remain asymptomatic.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/patologia , Glutaril-CoA Desidrogenase/metabolismo , Idade de Início , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Transporte Biológico , Encefalopatias Metabólicas/diagnóstico , Carnitina/análogos & derivados , Carnitina/urina , Glutaratos/urina , Glutaril-CoA Desidrogenase/deficiência , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
20.
PLoS One ; 8(5): e63084, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23658800

RESUMO

In glutaric aciduria type 1 (GA1), glutaryl-CoA dehydrogenase (GCDH) deficiency has been shown to be responsible for the accumulation of glutaric acid and striatal degeneration. However, the mechanisms by which GA1 induces striatal degeneration remain unclear. In this study, we aimed to establish a novel neuronal model of GA1 and to investigate the effects of GCDH deficiency and lysine-related metabolites on the viability of rat striatal neurons. Thus we constructed a lentiviral vector containing short hairpin RNA targeted against the GCDH gene expression (lentivirus-shRNA) in neurons. A virus containing a scrambled short hairpin RNA construct served as a control. Addition of lysine (5 mmol/L) was used to mimic hypermetabolism. Cell viability was measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. Apoptosis was assessed using Hoechst33342 staining and Annexin V-PE/7-AAD staining. The mitochondrial membrane potential (MPP) was monitored using tetramethylrhodamine methyl ester. The expression levels of caspases 3, 8, and 9 were determined by Western blotting. We found that lentivirus-shRNA induced apoptosis and decreased MMP levels in neurons, and addition of 5 mmol/L lysine enhanced this effect markedly. Lentivirus-shRNA upregulated the protein levels of caspases 3 and 9 regardless of the presence of 5 mmol/L lysine. The expression level of caspase 8 was higher in neurons co-treated with lentivirus-shRNA and 5 mmol/L lysine than in control. Benzyloxy-carbonyl-Val-Ala-Asp(OMe)-fluoromethylketone, a pan-caspase inhibitor, blocked the apoptosis induced by lentivirus-shRNA and 5 mmol/L lysine to a great extent. These results indicate that the targeted suppression of GCDH by lentivirus-mediated shRNA and excessive intake of lysine may be a useful cell model of GA1. These also suggest that GA1-induced striatal degeneration is partially caspase-dependent.


Assuntos
Apoptose/genética , Técnicas de Silenciamento de Genes , Glutaril-CoA Desidrogenase/genética , Lentivirus/genética , Lisina/farmacologia , Neurônios/citologia , RNA Interferente Pequeno , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Transporte Biológico/genética , Encefalopatias Metabólicas/enzimologia , Encefalopatias Metabólicas/metabolismo , Encefalopatias Metabólicas/patologia , Inibidores de Caspase/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Glutaril-CoA Desidrogenase/deficiência , Glutaril-CoA Desidrogenase/metabolismo , Lisina/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/genética , Neostriado/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA