Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 24(17)2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37685964

ABSTRACT

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.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Brain Diseases, Metabolic , Glutaryl-CoA Dehydrogenase , Glutaryl-CoA Dehydrogenase/chemistry , Glutaryl-CoA Dehydrogenase/genetics , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/genetics , Protein Folding , Mutation, Missense , Protein Domains , Humans , Enzyme Stability , Solubility
2.
J Cell Biochem ; 120(3): 3367-3372, 2019 03.
Article in English | MEDLINE | ID: mdl-30203563

ABSTRACT

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.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/genetics , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Homozygote , Mutation, Missense , Amino Acid Metabolism, Inborn Errors/pathology , Brain Diseases, Metabolic/pathology , Female , Humans , Infant , Male , Pedigree
3.
Neuroradiology ; 58(7): 697-703, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26993811

ABSTRACT

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.


Subject(s)
Adenosine Kinase/deficiency , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/pathology , Brain/metabolism , Brain/pathology , Magnetic Resonance Imaging/methods , Proton Magnetic Resonance Spectroscopy/methods , Adenosine Kinase/metabolism , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Molecular Imaging/methods , Reproducibility of Results , Sensitivity and Specificity
4.
J Inherit Metab Dis ; 38(2): 265-72, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25214427

ABSTRACT

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.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/genetics , Brain/enzymology , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Lysine/metabolism , Pipecolic Acids/metabolism , Animals , Deamination , Disease Models, Animal , Genetic Predisposition to Disease , Liver/enzymology , Lysine/analogs & derivatives , Mice, Knockout , Oxidation-Reduction , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peroxisomes/enzymology , Phenotype
5.
Genet Mol Res ; 14(4): 14130-5, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26535729

ABSTRACT

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.


Subject(s)
Brain Diseases, Metabolic/genetics , Hypoxia-Ischemia, Brain/genetics , Pyridoxal Phosphate/therapeutic use , Pyridoxaminephosphate Oxidase/deficiency , Pyridoxaminephosphate Oxidase/genetics , Seizures/genetics , Brain Diseases, Metabolic/enzymology , Female , Humans , Hypoxia-Ischemia, Brain/enzymology , Infant, Newborn , Mutation , Pyridoxaminephosphate Oxidase/drug effects , Seizures/drug therapy , Seizures/enzymology , Thailand
6.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 32(2): 187-91, 2015 Apr.
Article in Zh | MEDLINE | ID: mdl-25863083

ABSTRACT

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.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Asian People/genetics , Brain Diseases, Metabolic/enzymology , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Point Mutation , Amino Acid Metabolism, Inborn Errors/diagnostic imaging , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Sequence , Base Sequence , Brain Diseases, Metabolic/diagnostic imaging , Brain Diseases, Metabolic/genetics , DNA Mutational Analysis , Exons , Female , Glutaryl-CoA Dehydrogenase/chemistry , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Infant , Magnetic Resonance Imaging , Male , Molecular Sequence Data , Radiography , Sequence Alignment
7.
Am J Hum Genet ; 89(6): 806-12, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152682

ABSTRACT

Thiamine pyrophosphate (TPP) is an essential cofactor of the cytosolic transketolase and of three mitochondrial enzymes involved in the oxidative decarboxylation of either pyruvate, α-ketoglutarate or branched chain amino acids. Thiamine is taken up by specific transporters into the cell and converted to the active TPP by thiamine pyrophosphokinase (TPK) in the cytosol from where it can be transported into mitochondria. Here, we report five individuals from three families presenting with variable degrees of ataxia, psychomotor retardation, progressive dystonia, and lactic acidosis. Investigation of the mitochondrial energy metabolism showed reduced oxidation of pyruvate but normal pyruvate dehydrogenase complex activity in the presence of excess TPP. A reduced concentration of TPP was found in the muscle and blood. Mutation analysis of TPK1 uncovered three missense, one splice-site, and one frameshift mutation resulting in decreased TPK protein levels.


Subject(s)
Abnormalities, Multiple/enzymology , Brain Diseases, Metabolic/enzymology , Metabolic Networks and Pathways/genetics , Pyruvic Acid/metabolism , Thiamin Pyrophosphokinase/deficiency , Abnormalities, Multiple/drug therapy , Abnormalities, Multiple/genetics , Acidosis, Lactic/enzymology , Acidosis, Lactic/genetics , Adolescent , Amino Acid Sequence , Base Sequence , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/genetics , Child , DNA Mutational Analysis , Enzyme Assays , Fatal Outcome , Female , Humans , Male , Molecular Sequence Data , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Mutation , Oxidation-Reduction , Pedigree , Thiamin Pyrophosphokinase/genetics , Thiamine/blood , Thiamine/metabolism , Thiamine/therapeutic use
8.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 31(5): 608-11, 2014 Oct.
Article in Zh | MEDLINE | ID: mdl-25297592

ABSTRACT

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.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/genetics , Genetic Predisposition to Disease/genetics , Glutaryl-CoA Dehydrogenase/genetics , Mutation , Adolescent , Amino Acid Metabolism, Inborn Errors/diagnostic imaging , Amino Acid Metabolism, Inborn Errors/enzymology , Base Sequence , Brain Diseases, Metabolic/diagnostic imaging , Brain Diseases, Metabolic/enzymology , China , DNA Mutational Analysis , Family Health , Female , Glutaryl-CoA Dehydrogenase/deficiency , Homozygote , Humans , Infant, Newborn , Magnetic Resonance Imaging , Male , Radiography
9.
ACS Chem Biol ; 19(7): 1544-1553, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38915184

ABSTRACT

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.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Brain Diseases, Metabolic , Humans , Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/enzymology , Glutarates/metabolism , Glutarates/chemistry , Losartan/pharmacology , Losartan/chemistry , Coenzyme A-Transferases/metabolism , Coenzyme A-Transferases/antagonists & inhibitors , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/chemistry , Valsartan , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Crystallography, X-Ray , Catalytic Domain , Acyl Coenzyme A/metabolism , Acyl Coenzyme A/chemistry , Models, Molecular , High-Throughput Screening Assays , Glutaryl-CoA Dehydrogenase/deficiency
10.
Mol Genet Metab ; 108(4): 206-11, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23433712

ABSTRACT

Mitochondrial aminoacyl-tRNA synthetases (mtARSs) are essential in the process of transferring genetic information from mitochondrial DNA to the complexes of the oxidative phosphorylation system. These synthetases perform an integral step in the initiation of mitochondrial protein synthesis by charging tRNAs with their cognate amino acids. All mtARSs are encoded by nuclear genes, nine of which have recently been described as disease genes for mitochondrial disorders. Unexpectedly, the clinical presentations of these diseases are highly specific to the affected synthetase. Encephalopathy is the most common manifestation but again with gene-specific outcomes. Other clinical presentations include myopathy with anemia, cardiomyopathy, tubulopathy and hearing loss with female ovarian dysgenesis. Here we review the described mutation types and the associated patient phenotypes. The identified mutation spectrum suggests that only mutation types that allow some residual tRNA-charging activity can result in the described mtARS diseases but the molecular mechanisms behind the selective tissue involvement are not currently understood.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Brain Diseases, Metabolic/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Mitochondria/genetics , Muscular Diseases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Brain Diseases, Metabolic/enzymology , DNA, Mitochondrial/genetics , Female , Hereditary Sensory and Motor Neuropathy/enzymology , Humans , Mitochondria/enzymology , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Muscular Diseases/enzymology , RNA, Transfer/genetics , RNA, Transfer/metabolism
11.
Mol Genet Metab ; 107(3): 375-82, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22999741

ABSTRACT

Mitochondrial dysfunction has been proposed to play an important role in the neuropathology of glutaric acidemia type I (GA I). However, the relevance of bioenergetics disruption and the exact mechanisms responsible for the cortical leukodystrophy and the striatum degeneration presented by GA I patients are not yet fully understood. Therefore, in the present work we measured the respiratory chain complexes activities I-IV, mitochondrial respiratory parameters state 3, state 4, the respiratory control ratio and dinitrophenol (DNP)-stimulated respiration (uncoupled state), as well as the activities of α-ketoglutarate dehydrogenase (α-KGDH), creatine kinase (CK) and Na+, K+-ATPase in cerebral cortex, striatum and hippocampus from 30-day-old Gcdh-/- and wild type (WT) mice fed with a normal or a high Lys (4.7%) diet. When a baseline (0.9% Lys) diet was given, we verified mild alterations of the activities of some respiratory chain complexes in cerebral cortex and hippocampus, but not in striatum from Gcdh-/- mice as compared to WT animals. Furthermore, the mitochondrial respiratory parameters and the activities of α-KGDH and CK were not modified in all brain structures from Gcdh-/- mice. In contrast, we found a significant reduction of Na(+), K(+)-ATPase activity associated with a lower degree of its expression in cerebral cortex from Gcdh-/- mice. Furthermore, a high Lys (4.7%) diet did not accentuate the biochemical alterations observed in Gcdh-/- mice fed with a normal diet. Since Na(+), K(+)-ATPase activity is required for cell volume regulation and to maintain the membrane potential necessary for a normal neurotransmission, it is presumed that reduction of this enzyme activity may represent a potential underlying mechanism involved in the brain swelling and cortical abnormalities (cortical atrophy with leukodystrophy) observed in patients affected by GA I.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/pathology , Cerebral Cortex/pathology , Corpus Striatum/pathology , Glutaryl-CoA Dehydrogenase/deficiency , Hippocampus/pathology , Sodium-Potassium-Exchanging ATPase/genetics , Amino Acid Metabolism, Inborn Errors/enzymology , Animals , Brain Diseases, Metabolic/enzymology , Cerebral Cortex/enzymology , Corpus Striatum/enzymology , Creatine Kinase/genetics , Creatine Kinase/metabolism , Down-Regulation , Electron Transport/genetics , Food, Formulated , Gene Expression , Glutaryl-CoA Dehydrogenase/genetics , Hippocampus/enzymology , Humans , Ketoglutarate Dehydrogenase Complex/genetics , Ketoglutarate Dehydrogenase Complex/metabolism , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation , Sodium-Potassium-Exchanging ATPase/metabolism
12.
Eur J Paediatr Neurol ; 39: 49-58, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35662016

ABSTRACT

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.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Brain Diseases, Metabolic , Glutaryl-CoA Dehydrogenase , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/genetics , Child , Cross-Sectional Studies , Glutaryl-CoA Dehydrogenase/chemistry , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Humans , Quality of Life
13.
Mol Genet Metab ; 103(1): 89-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21353613

ABSTRACT

Glutamine deficiency with hyperammonemia due to an inherited defect of glutamine synthetase (GS) was found in a 2 year old patient. He presented neonatal seizures and developed chronic encephalopathy. Thus, GS deficiency leads to severe neurological disease but is not always early lethal.


Subject(s)
Brain Diseases, Metabolic/enzymology , Glutamate-Ammonia Ligase/deficiency , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/pathology , Child, Preschool , Exanthema/pathology , Glutamate-Ammonia Ligase/genetics , Humans , Hyperammonemia/diagnosis , Hyperammonemia/enzymology , Hyperammonemia/pathology , Male , Mutation/genetics
14.
Mol Genet Metab ; 102(3): 343-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21176883

ABSTRACT

Glutaric acidemia type 1 (GA1) is a metabolic disease caused by a deficiency of glutaryl-CoA dehydrogenase (GCDH). Untreated patients mostly develop severe striatal degeneration. More than 200 mutations have been reported in the GCDH gene, and common R402W and IVS10-2A>C were found in Caucasian and Chinese/Taiwanese, respectively. However, in Japan, genetic mutations have only been reported in a few cases. Herein, we report the clinical and molecular basis of GA1 in 19 Japanese patients, including six previously reported patients. All cases showed high urinary glutaric acid excretion. Eleven patients were severely impaired (three patients died), three had mild impairment, and five showed normal development. Four of 5 patients that developed normally were detected in the presymptomatic stage by neonatal or sibling screening. Nineteen mutations in 26 alleles were identified, and eight of them (89 or 90delC, Y155C, IVS4+2T>C, G244S, Q352X, G354A, K361E, and 1144-1145delGC) were novel. S305L (12.1%, 4/34 alleles) was found in several cases, suggesting that this mutation is a common mutation. In contrast, R402W was not identified and IVS10-2A>C was only found in one allele, suggesting that Japanese patients with GA1 show allelic heterogeneity and have a different genetic background to patients from other countries. One of a pair of sisters with the same mutations (M339V/S305L) lacking residual activity was severely retarded, whereas the older girl remains asymptomatic at 22 years of age, indicating that genotype does not necessarily predict GA1 phenotype. We consistently found that there was no association between genotype and phenotype. However, children with mild impairment were diagnosed and treated earlier than severely impaired cases {4.7±2.5 months (range: 2-8 months) vs. 11.6±12.7 months (range: 4-51 months)}. Our results suggest that early detection and treatment but not genotype are associated with better patient outcome, reinforcing the importance of neonatal screening.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Brain Diseases, Metabolic , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/mortality , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/mortality , Child, Preschool , Female , Gene Order , Glutarates/urine , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Infant , Infant, Newborn , Japan , Male , Mutation , Pedigree
15.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 28(4): 374-8, 2011 Aug.
Article in Zh | MEDLINE | ID: mdl-21811973

ABSTRACT

OBJECTIVE: To investigate the mutations of glutaryl-CoA dehydrogenase (GCDH) gene in patients with glutaric aciduria type I(GA-1). METHODS: Genomic DNA was extracted from peripheral blood cells of the eight probands with GA-1 who were diagnosed by urine and blood analyses. By PCR and direct sequencing, all 11 exons and their flanking sequences of the GCDH gene were examined. Mutation search was also performed in some of their family members. RESULTS: Among the eight patients diagnosed by metabolic screening, seven patients belonged to classical infantile-onset. One patient, however, was adult-onset, who was admitted to the hospital because of suffering from ischemic cerebral stroke. The GCDH gene mutations were identified in all the eight probands with GA-1: five of them had compound heterozygous mutations, while the other three harbored only one heterozygous mutation. Totally, nine different mutations of the GCDH gene were identified in the eight probands, four of them were novel, i.e., c.148T>C, c.371G>A, 909delC and c.263G>A. CONCLUSION: GCDH gene mutations are identified in 8 patients with GA-1 in mainland China, including one adult patient with late onset. Four novel mutations of GCDH gene are found which expanded the mutational spectrum of the GCDH gene.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/genetics , DNA Mutational Analysis , Glutaryl-CoA Dehydrogenase/genetics , Adult , Amino Acid Sequence , Animals , Base Sequence , Exons/genetics , Female , Glutaryl-CoA Dehydrogenase/chemistry , Glutaryl-CoA Dehydrogenase/deficiency , Humans , Infant , Male , Molecular Sequence Data
16.
Protein Sci ; 30(10): 1995-2008, 2021 10.
Article in English | MEDLINE | ID: mdl-34288185

ABSTRACT

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.


Subject(s)
Aspartate Carbamoyltransferase , Brain Diseases, Metabolic/enzymology , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing) , Dihydroorotase , Animals , Aspartate Carbamoyltransferase/chemistry , Aspartate Carbamoyltransferase/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/chemistry , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Dihydroorotase/chemistry , Dihydroorotase/metabolism , Humans , Protein Domains
17.
BMC Neurosci ; 11: 51, 2010 Apr 23.
Article in English | MEDLINE | ID: mdl-20416081

ABSTRACT

BACKGROUND: The administration of the ketone bodies hydroxybutyrate and acetoacetate is known to exert a protective effect against metabolic disorders associated with cerebral pathologies. This suggests that the enhancement of their endogenous production might be a rational therapeutic approach. Ketone bodies are generated by fatty acid beta-oxidation, a process involving a mitochondrial oxido-reductase superfamily, with fatty acid-CoA thioesters as substrates. In this report, emphasis is on the penultimate step of the process, i.e. L-3-hydroxybutyryl-CoA dehydrogenase activity. We determined changes in enzyme activity and in circulating ketone body levels in the MPTP mouse model of Parkinson's disease. Since the active moiety of CoA is pantetheine, mice were treated with pantethine, its naturally-occurring form. Pantethine has the advantage of being known as an anti-inflammatory and hypolipidemic agent with very few side effects. RESULTS: We found that dehydrogenase activity and circulating ketone body levels were drastically reduced by the neurotoxin MPTP, whereas treatment with pantethine overcame these adverse effects. Pantethine prevented dopaminergic neuron loss and motility disorders. In vivo and in vitro experiments showed that the protection was associated with enhancement of glutathione (GSH) production as well as restoration of respiratory chain complex I activity and mitochondrial ATP levels. Remarkably, pantethine treatment boosted the circulating ketone body levels in MPTP-intoxicated mice, but not in normal animals. CONCLUSIONS: These finding demonstrate the feasibility of the enhancement of endogenous ketone body production and provide a promising therapeutic approach to Parkinson's disease as well as, conceivably, to other neurodegenerative disorders.


Subject(s)
Brain Diseases, Metabolic/drug therapy , Hydroxybutyrate Dehydrogenase/drug effects , Ketone Bodies/blood , Pantetheine/analogs & derivatives , Parkinsonian Disorders/drug therapy , Acyl Coenzyme A/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/physiopathology , Dopamine/metabolism , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Encephalitis/drug therapy , Encephalitis/enzymology , Encephalitis/physiopathology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glutathione/metabolism , Hydroxybutyrate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/drug therapy , Nerve Degeneration/enzymology , Nerve Degeneration/prevention & control , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pantetheine/metabolism , Pantetheine/pharmacology , Pantetheine/therapeutic use , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/physiopathology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Up-Regulation/drug effects , Up-Regulation/physiology
18.
Cell Mol Neurobiol ; 30(3): 445-51, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19838799

ABSTRACT

Diabetes mellitus (DM) is associated with increased risk of impaired cognitive function. Diabetic neuropathy is one of the most common and important complications of DM. Estrogens prevent neuronal loss in experimental models of neurodegeneration and accelerate nerve regeneration. Aromatase catalyzes the conversion of androgens to estrogens and expressed in a variety of tissues including neurons. Although insulin is known to regulate the activity of aromatase there is no study about the effects of diabetes on this enzyme. Present study was designed to investigate the effects of experimental diabetes on aromatase expression in nervous system. Gender-based differences were also investigated. Rats were injected with streptozotocin to induce diabetes. At the end of 4 and 12 weeks sciatic nerve and hippocampus homogenates were prepared and evaluated for aromatase proteins. Aromatase expressions in sciatic nerves of both genders were decreased in 4 weeks of diabetes, but in 12 weeks the enzyme levels were increased in females and reached to control levels in male animals. Aromatase levels were not altered in hippocampus at 4 weeks but increased at 12 weeks in female diabetic rats. No significant differences were observed at enzyme levels of hippocampus in male diabetic rats. Insulin therapy prevented all diabetes-induced changes. In conclusion, these results indicated for the first time that, DM altered the expression of aromatase both in central and peripheral nervous systems. Peripheral nervous system is more vulnerable to damage than central nervous system in diabetes. These effects of diabetes differ with gender and compensatory neuroprotective mechanisms are more efficient in female rats.


Subject(s)
Aromatase/metabolism , Brain Diseases, Metabolic/enzymology , Cognition Disorders/enzymology , Cytoprotection/physiology , Diabetes Complications/enzymology , Estrogens/biosynthesis , Animals , Aromatase/analysis , Brain Diseases, Metabolic/etiology , Brain Diseases, Metabolic/physiopathology , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Diabetes Complications/physiopathology , Diabetes Mellitus, Experimental , Diabetic Neuropathies/enzymology , Diabetic Neuropathies/physiopathology , Disease Models, Animal , Female , Hippocampus/enzymology , Hippocampus/physiopathology , Insulin/pharmacology , Male , Nerve Degeneration/enzymology , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Nerve Regeneration/physiology , Neuroprotective Agents/metabolism , Rats , Rats, Sprague-Dawley , Sciatic Nerve/enzymology , Sciatic Nerve/physiopathology , Sex Characteristics
19.
Pediatr Res ; 68(2): 159-64, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20453710

ABSTRACT

This patient presented on the first day of life with pronounced lactic acidosis with an elevated lactate/pyruvate ratio. Urine organic acids showed Krebs cycle metabolites and mildly elevated methylmalonate and methylcitrate. The acylcarnitine profile showed elevated propionylcarnitine and succinylcarnitine. Amino acids showed elevated glutamic acid, glutamine, proline, and alanine. From the age 2 of mo on, she had elevated transaminases and intermittent episodes of liver failure. Liver biopsy showed steatosis and a decrease of mitochondrial DNA to 50% of control. She had bilateral sensorineural hearing loss. Over the course of the first 2 y of life, she developed a progressively severe myopathy with pronounced muscle weakness eventually leading to respiratory failure, Leigh disease, and recurrent hepatic failure. The hepatic symptoms and the metabolic parameters temporarily improved on treatment with aspartate, but neither muscle symptoms nor brain lesions improved. Laboratory testing revealed a deficiency of succinyl-CoA ligase enzyme activity and protein in fibroblasts because of a novel homozygous mutation in the SUCLG1 gene: c.40A>T (p.M14L). Functional analysis suggests that this methionine is more likely to function as the translation initiator methionine, explaining the pathogenic nature of the mutation. Succinyl-CoA ligase deficiency due to an SUCLG1 mutation is a new cause for mitochondrial hepatoencephalomyopathy.


Subject(s)
Brain Diseases, Metabolic , Liver Diseases , Mitochondrial Diseases , Succinate-CoA Ligases/deficiency , Amino Acid Sequence , Base Sequence , Brain/metabolism , Brain/pathology , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/pathology , DNA Mutational Analysis , Fatal Outcome , Female , Humans , Infant , Infant, Newborn , Leigh Disease/enzymology , Leigh Disease/genetics , Leigh Disease/pathology , Liver Diseases/enzymology , Liver Diseases/genetics , Liver Diseases/pathology , Magnetic Resonance Imaging , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Molecular Sequence Data , Mutation , Succinate-CoA Ligases/genetics
20.
J Inherit Metab Dis ; 33 Suppl 3: S25-33, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20049532

ABSTRACT

We describe two neonates presenting with perinatal hypophosphatasia and severe epileptic encephalopathy resulting in death. Both had increased levels of urinary vanillactate, indicating functional deficiency of aromatic amino acid decarboxylase, a pyridoxal-5-phosphate (PLP)-dependent enzyme required for dopamine and serotonin biosynthesis. Clinical findings and results of subsequent metabolic investigations were consistent with secondary pyridoxine-deficient encephalopathy. These patients highlight the importance of tissue non-specific alkaline phosphatase in the neuronal PLP-dependent metabolism of neurotransmitters. In addition, the disturbance of PLP metabolism appears to underlie the predominant neurological presentation in our patients. We recommend the measurement of serum alkaline phosphatase (ALP) during the assessment of perinatal seizures.


Subject(s)
Alkaline Phosphatase/genetics , Biogenic Monoamines/metabolism , Brain Diseases, Metabolic/genetics , Hypophosphatasia/genetics , Hypoxia-Ischemia, Brain/genetics , Mutation , Pyridoxal Phosphate/deficiency , Pyridoxaminephosphate Oxidase/deficiency , Seizures/genetics , Vitamin B 6 Deficiency/genetics , Alkaline Phosphatase/metabolism , Anticonvulsants/therapeutic use , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/enzymology , Fatal Outcome , Female , Genetic Predisposition to Disease , Humans , Hypophosphatasia/complications , Hypophosphatasia/diagnosis , Hypophosphatasia/drug therapy , Hypophosphatasia/enzymology , Hypoxia-Ischemia, Brain/diagnosis , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/enzymology , Infant, Newborn , Male , Phenotype , Pyridoxaminephosphate Oxidase/genetics , Seizures/diagnosis , Seizures/drug therapy , Seizures/enzymology , Treatment Outcome , Vitamin B 6 Deficiency/diagnosis , Vitamin B 6 Deficiency/drug therapy , Vitamin B 6 Deficiency/enzymology , Vitamin B Complex/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL