Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 305
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 24(11): 3238-47, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25721401

RESUMO

Acyl-CoA dehydrogenase 9 (ACAD9) is an assembly factor for mitochondrial respiratory chain Complex I (CI), and ACAD9 mutations are recognized as a frequent cause of CI deficiency. ACAD9 also retains enzyme ACAD activity for long-chain fatty acids in vitro, but the biological relevance of this function remains controversial partly because of the tissue specificity of ACAD9 expression: high in liver and neurons and minimal in skin fibroblasts. In this study, we hypothesized that this enzymatic ACAD activity is required for full fatty acid oxidation capacity in cells expressing high levels of ACAD9 and that loss of this function is important in determining phenotype in ACAD9-deficient patients. First, we confirmed that HEK293 cells express ACAD9 abundantly. Then, we showed that ACAD9 knockout in HEK293 cells affected long-chain fatty acid oxidation along with Cl, both of which were rescued by wild type ACAD9. Further, we evaluated whether the loss of ACAD9 enzymatic fatty acid oxidation affects clinical severity in patients with ACAD9 mutations. The effects on ACAD activity of 16 ACAD9 mutations identified in 24 patients were evaluated using a prokaryotic expression system. We showed that there was a significant inverse correlation between residual enzyme ACAD activity and phenotypic severity of ACAD9-deficient patients. These results provide evidence that in cells where it is strongly expressed, ACAD9 plays a physiological role in fatty acid oxidation, which contributes to the severity of the phenotype in ACAD9-deficient patients. Accordingly, treatment of ACAD9 patients should aim at counteracting both CI and fatty acid oxidation dysfunctions.


Assuntos
Acil-CoA Desidrogenases/genética , Complexo I de Transporte de Elétrons/metabolismo , Ácidos Graxos/metabolismo , Doenças Mitocondriais/enzimologia , Acil-CoA Desidrogenases/deficiência , Animais , Estudos de Associação Genética , Células HEK293 , Humanos , Camundongos , Doenças Mitocondriais/patologia , Mutação de Sentido Incorreto , Oxirredução , Multimerização Proteica , Índice de Gravidade de Doença
2.
Hum Mol Genet ; 23(5): 1311-9, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24158852

RESUMO

Oxidative phosphorylation and fatty acid oxidation are two major metabolic pathways in mitochondria. Acyl-CoA dehydrogenase 9 (ACAD9), an enzyme assumed to play a role in fatty acid oxidation, was recently identified as a factor involved in complex I biogenesis. Here we further investigated the role of ACAD9's enzymatic activity in fatty acid oxidation and complex I biogenesis. We provide evidence indicating that ACAD9 displays enzyme activity in vivo. Knockdown experiments in very-long-chain acyl-CoA dehydrogenase (VLCAD)-deficient fibroblasts revealed that ACAD9 is responsible for the production of C14:1-carnitine from oleate and C12-carnitine from palmitate. These results explain the origin of these obscure acylcarnitines that are used to diagnose VLCAD deficiency in humans. Knockdown of ACAD9 in control fibroblasts did not reveal changes in the acylcarnitine profiles upon fatty acid loading. Next, we investigated whether catalytic activity of ACAD9 was necessary for complex I biogenesis. Catalytically inactive ACAD9 gave partial-to-complete rescue of complex I biogenesis in ACAD9-deficient cells and was incorporated in high-molecular-weight assembly intermediates. Our results underscore the importance of the ACAD9 protein in complex I assembly and suggest that the enzymatic activity is a rudiment of the duplication event.


Assuntos
Acil-CoA Desidrogenases/metabolismo , Ácidos Graxos/metabolismo , Acil-CoA Desidrogenase de Cadeia Longa/deficiência , Acil-CoA Desidrogenase de Cadeia Longa/metabolismo , Acil-CoA Desidrogenases/química , Acil-CoA Desidrogenases/deficiência , Acil-CoA Desidrogenases/genética , Carnitina/biossíntese , Catálise , Linhagem Celular , Síndrome Congênita de Insuficiência da Medula Óssea , Complexo I de Transporte de Elétrons/deficiência , Ativação Enzimática , Humanos , Erros Inatos do Metabolismo Lipídico/metabolismo , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Modelos Moleculares , Peso Molecular , Doenças Musculares/metabolismo , Mutação , Oxirredução , Fosforilação Oxidativa , Conformação Proteica
3.
Mol Genet Metab ; 99(3): 263-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20036593

RESUMO

INTRODUCTION: Medium chain acyl-CoA dehydrogenase (MCAD) deficiency is one of the most common inborn errors of metabolism. Affected patients have impaired ability to break down medium chain fatty acids during fasting, and typically present in the early years of life with hypoketotic hypoglycemia, Reye syndrome-like symptoms, brain damage or death. The development of newborn screening (NBS) for MCAD deficiency has greatly improved outcome, but some patients still appear at risk for severe complications. We reviewed the outcome of patients identified with MCAD deficiency by the New York State NBS process to identify biochemical or genotypic markers which might predict outcome. METHOD: All eight NBS follow-up centers in New York State contributed the cases of MCAD deficiency diagnosed by newborn screen, who received diagnostic and follow-up care in their clinic. Data reviewed included gender, age, birthweight, initial NBS octanoylcarnitine level (C8) and C8/C2 ratio, follow-up C8 and hexanoylglycine, race/ethnicity, and presence of neonatal or later symptoms. RESULTS: We identified 53 cases of MCAD deficiency. More than one quarter of patients had a post-neonatal symptomatic admission (predominantly lethargy associated with an intercurrent illness). No genotype or C8 level was protective for neonatal or later symptoms. There was a relationship between initial C8 level or C8/C2 ratio and occurrence of later symptoms (7.3 micromol/L in the asymptomatic vs. 19.1 micromol/L in the symptomatic, p<0.0002 for C8, and 0.26 vs. 0.6, respectively, for C8/C2 ratio, p<0.012). Four infants had initial C8 level >30 micromol/L; these infants had a high rate of symptomatic or multiple symptomatic episodes or a history of sibling death from "SIDS", and typically had deletion, nonsense or splice sites mutations. Infants having a history of a symptomatic episode were more likely to have higher initial C8 on NBS and a genotype predicted to strongly affect protein function. In our ethnically diverse group of patients, the c.985A>G mutation was rarely found in non-Caucasians. DISCUSSION: No genotype or metabolite profile is protective from symptoms. The strong relationship between initial C8 level and outcome suggests that in at least some cases neonates having high initial C8 levels may be demonstrating an increased susceptibility to catabolic stress, and may merit additional precautions. Our data also suggest that these infants are more likely to carry severe mutations including homozygosity for the common mutation, deletions, nonsense or splice site mutations. The reports of significant lethargy or hypoglycemia during intercurrent illness in over one quarter of cases even when early medical intervention is recommended (and even when initial C8 is not profoundly elevated) underscores the importance of continued vigilance to prevent stressful fasting in this disorder.


Assuntos
Acil-CoA Desidrogenases/deficiência , Acil-CoA Desidrogenases/genética , Erros Inatos do Metabolismo/diagnóstico , Erros Inatos do Metabolismo/fisiopatologia , Triagem Neonatal/métodos , Carnitina/análogos & derivados , Carnitina/sangue , Ácidos Graxos/metabolismo , Feminino , Genótipo , Humanos , Recém-Nascido , Masculino , Erros Inatos do Metabolismo/genética , Mutação , New York , Fenótipo , Prognóstico
4.
Rev Neurol (Paris) ; 166(3): 289-94, 2010 Mar.
Artigo em Francês | MEDLINE | ID: mdl-19592060

RESUMO

INTRODUCTION: Multiple acyl-CoA dehydrogenase deficiency (MADD) is a rare genetic disease involving fatty acid oxidation. It is due to the deficiency of one of the two electron transporters: electron transfer flavoprotein (ETF) or electron transfer flavoprotein ubiquinone oxydoreductase (ETF-QO). Symptoms begin more often in childhood or in young adulthood with a multisystemic disease with encephalopathy or muscular weakness. CASE REPORTS: We report here two adult cases with ETF-QO deficiency, confirmed by mutation analysis (ETFDH gene), revealed by a muscular weakness associated with muscle lipidosis. One of our patients presented an acute encephalopathy with vomiting ten years before the onset of muscular symptoms. The second patient exhibited a slowly progressive pelvic girdle muscle weakness. Diagnosis was established by characteristic abnormalities of acylcarnitine profile by tandem mass spectrometry. For both patients, a dramatic clinical improvement was observed under treatment with riboflavine and L-carnitine. CONCLUSION: Since it is a treatable disorder, this diagnosis must be considered by performing an acylcarnitine profile in all patients presenting with an unexplained muscular weakness.


Assuntos
Acil-CoA Desidrogenases/deficiência , Erros Inatos do Metabolismo Lipídico/enzimologia , Erros Inatos do Metabolismo Lipídico/genética , Lipidoses/tratamento farmacológico , Lipidoses/genética , Adulto , Biópsia , Encefalopatias Metabólicas/genética , Carnitina/análogos & derivados , Carnitina/análise , Carnitina/metabolismo , Corantes , Análise Mutacional de DNA , Transporte de Elétrons/genética , Flavoproteínas Transferidoras de Elétrons/genética , Flavoproteínas Transferidoras de Elétrons/metabolismo , Feminino , Humanos , Erros Inatos do Metabolismo Lipídico/patologia , Lipidoses/patologia , Masculino , Pessoa de Meia-Idade , Debilidade Muscular/etiologia , Debilidade Muscular/genética , Músculo Esquelético/patologia , Riboflavina/metabolismo , Espectrometria de Massas em Tandem , Adulto Jovem
5.
J Pediatr Endocrinol Metab ; 32(10): 1181-1185, 2019 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-31473688

RESUMO

Mitochondrial acyl-CoA dehydrogenase 9 (ACAD9) deficiency is one of the common causes of respiratory chain complex I deficiency, which is characterized by cardiomyopathy, lactic acidemia, and muscle weakness. Infantile cardiomyopathy is the most common phenotype and is usually lethal by the age of 5 years. Riboflavin treatment is known to be effective in ~65% of the patients; however, the remaining are unresponsive to riboflavin and are in need of additional treatment measures. In this report, we describe a patient with ACAD9 deficiency who developed progressive cardiomyopathy at 8 months of age. As the patient's left ventricular ejection fraction (LVEF) kept decreasing to 45.4% at 1 year 8 months, sodium pyruvate treatment was introduced together with a beta-blocker and coenzyme Q10. This resulted in a steady improvement, with full and sustained normalization of cardiac function without riboflavin. The therapy, therefore, might be a useful addition for the treatment of ACAD9 deficiency.


Assuntos
Acidose/tratamento farmacológico , Acil-CoA Desidrogenase/deficiência , Acil-CoA Desidrogenases/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Cardiomiopatias/tratamento farmacológico , Cardiomiopatia Hipertrófica/tratamento farmacológico , Carvedilol/administração & dosagem , Doenças Mitocondriais/tratamento farmacológico , Debilidade Muscular/tratamento farmacológico , Piruvatos/administração & dosagem , Ubiquinona/análogos & derivados , Acidose/complicações , Acidose/patologia , Antagonistas Adrenérgicos beta/administração & dosagem , Erros Inatos do Metabolismo dos Aminoácidos/complicações , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Cardiomiopatias/complicações , Cardiomiopatias/patologia , Cardiomiopatia Hipertrófica/complicações , Cardiomiopatia Hipertrófica/patologia , Quimioterapia Combinada , Feminino , Humanos , Recém-Nascido , Doenças Mitocondriais/complicações , Doenças Mitocondriais/patologia , Debilidade Muscular/complicações , Debilidade Muscular/patologia , Prognóstico , Ubiquinona/administração & dosagem , Vitaminas/administração & dosagem
6.
Neuromuscul Disord ; 18(5): 355-64, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18406615

RESUMO

The aim of the current study was to assess lipid metabolism in horses with atypical myopathy. Urine samples from 10 cases were subjected to analysis of organic acids, glycine conjugates, and acylcarnitines revealing increased mean excretion of lactic acid, ethylmalonic acid, 2-methylsuccinic acid, butyrylglycine, (iso)valerylglycine, hexanoylglycine, free carnitine, C2-, C3-, C4-, C5-, C6-, C8-, C8:1-, C10:1-, and C10:2-carnitine as compared with 15 control horses (12 healthy and three with acute myopathy due to other causes). Analysis of plasma revealed similar results for these predominantly short-chain acylcarnitines. Furthermore, measurement of dehydrogenase activities in lateral vastus muscle from one horse with atypical myopathy indeed showed deficiencies of short-chain acyl-CoA dehydrogenase (0.66 as compared with 2.27 and 2.48 in two controls), medium-chain acyl-CoA dehydrogenase (0.36 as compared with 4.31 and 4.82 in two controls) and isovaleryl-CoA dehydrogenase (0.74 as compared with 1.43 and 1.61 nmol min(-1) mg(-1) in two controls). A deficiency of several mitochondrial dehydrogenases that utilize flavin adenine dinucleotide as cofactor including the acyl-CoA dehydrogenases of fatty acid beta-oxidation, and enzymes that degrade the CoA-esters of glutaric acid, isovaleric acid, 2-methylbutyric acid, isobutyric acid, and sarcosine was suspected in 10 out of 10 cases as the possible etiology for a highly fatal and prevalent toxic equine muscle disease similar to the combined metabolic derangements seen in human multiple acyl-CoA dehydrogenase deficiency also known as glutaric acidemia type II.


Assuntos
Acil-CoA Desidrogenases/deficiência , Doenças dos Cavalos/metabolismo , Doenças Musculares/metabolismo , Acil-CoA Desidrogenase/deficiência , Acil-CoA Desidrogenase/metabolismo , Acil-CoA Desidrogenases/metabolismo , Animais , Ácido Butírico/sangue , Ácido Butírico/urina , Butiril-CoA Desidrogenase/deficiência , Butiril-CoA Desidrogenase/metabolismo , Carnitina/análogos & derivados , Carnitina/sangue , Carnitina/urina , Cromatografia Líquida de Alta Pressão , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Glutaratos/sangue , Glutaratos/urina , Doenças dos Cavalos/enzimologia , Doenças dos Cavalos/patologia , Cavalos , Isovaleril-CoA Desidrogenase/deficiência , Isovaleril-CoA Desidrogenase/metabolismo , Ácido Láctico/sangue , Ácido Láctico/urina , Masculino , Microscopia Eletrônica , Microscopia de Fluorescência , Músculos/patologia , Músculos/ultraestrutura , Doenças Musculares/enzimologia , Doenças Musculares/patologia , Riboflavina/sangue
7.
Mol Genet Metab ; 95(4): 195-200, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18977676

RESUMO

Short-chain acyl-CoA dehydrogenase deficiency (SCADD) is a disorder of mitochondrial fatty acid oxidation that leads to the accumulation of butyrylcarnitine and ethylmalonic acid in blood and urine. Originally described with a relatively severe phenotype, most patients are now diagnosed through newborn screening by tandem mass spectrometry and remain asymptomatic. Molecular analysis of affected individuals has identified a preponderance of private inactivating point mutations and one common one present in high frequency in individuals of Ashkenazi Jewish ancestry. In addition, two polymorphic variants have been identified that have little affect on enzyme kinetics but impair folding and stability. Individuals homozygous for one of these variants or compound heterozygous for one of each often show an increased level of ethylmalonic acid excretion that appears not to be clinically significant. The combination of asymptomatic affected newborns and the frequent variants can cause much confusion in evaluating and treating individuals with SCADD. The long-term consequences and the need for chronic therapy remain current topics of contention and investigation.


Assuntos
Acil-CoA Desidrogenases/deficiência , Acil-CoA Desidrogenases/genética , Erros Inatos do Metabolismo Lipídico/enzimologia , Acil-CoA Desidrogenases/química , Animais , Feminino , Testes Genéticos , Humanos , Recém-Nascido , Erros Inatos do Metabolismo Lipídico/diagnóstico , Erros Inatos do Metabolismo Lipídico/genética , Erros Inatos do Metabolismo Lipídico/fisiopatologia , Masculino , Mutação , Polimorfismo Genético
8.
ACS Synth Biol ; 7(1): 86-97, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29216425

RESUMO

The ethylmalonyl-CoA pathway (EMCP) is an anaplerotic reaction sequence in the central carbon metabolism of numerous Proteo- and Actinobacteria. The pathway features several CoA-bound mono- and dicarboxylic acids that are of interest as platform chemicals for the chemical industry. The EMCP, however, is essential for growth on C1 and C2 carbon substrates and therefore cannot be simply interrupted to drain these intermediates. In this study, we aimed at reengineering central carbon metabolism of the Alphaproteobacterium Methylobacterium extorquens AM1 for the specific production of EMCP derivatives in the supernatant. Establishing a heterologous glyoxylate shunt in M. extorquens AM1 restored wild type-like growth in several EMCP knockout strains on defined minimal medium with acetate as carbon source. We further engineered one of these strains that carried a deletion of the gene encoding crotonyl-CoA carboxylase/reductase to demonstrate in a proof-of-concept the specific production of crotonic acid in the supernatant on a defined minimal medium. Our experiments demonstrate that it is in principle possible to further exploit the EMCP by establishing an alternative central carbon metabolic pathway in M. extorquens AM1, opening many possibilities for the biotechnological production of EMCP-derived compounds in future.


Assuntos
Acil Coenzima A/genética , Proteínas de Bactérias/genética , Carbono/metabolismo , Glioxilatos/metabolismo , Engenharia Metabólica , Methylobacterium extorquens/metabolismo , Ácido Acético/metabolismo , Acil Coenzima A/deficiência , Acil-CoA Desidrogenases/deficiência , Acil-CoA Desidrogenases/genética , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Proteínas de Bactérias/metabolismo , Crotonatos/metabolismo , Formiato Desidrogenases/genética , Formiato Desidrogenases/metabolismo , Isocitrato Liase/genética , Isocitrato Liase/metabolismo , Malato Sintase/genética , Malato Sintase/metabolismo , Metanol/química , Metanol/metabolismo , Methylobacterium extorquens/genética , Methylobacterium extorquens/crescimento & desenvolvimento , Oxirredução , Espectrofotometria
9.
Sci Rep ; 8(1): 1165, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29348607

RESUMO

Mitochondrial complex I (CI) deficiency is the most frequent cause of oxidative phosphorylation (OXPHOS) disorders in humans. In order to benchmark the effects of CI deficiency on mitochondrial bioenergetics and dynamics, respiratory chain (RC) and endoplasmic reticulum (ER)-mitochondria communication, and superoxide production, fibroblasts from patients with mutations in the ND6, NDUFV1 or ACAD9 genes were analyzed. Fatty acid metabolism, basal and maximal respiration, mitochondrial membrane potential, and ATP levels were decreased. Changes in proteins involved in mitochondrial dynamics were detected in various combinations in each cell line, while variable changes in RC components were observed. ACAD9 deficient cells exhibited an increase in RC complex subunits and DDIT3, an ER stress marker. The level of proteins involved in ER-mitochondria communication was decreased in ND6 and ACAD9 deficient cells. |ΔΨ| and cell viability were further decreased in all cell lines. These findings suggest that disruption of mitochondrial bioenergetics and dynamics, ER-mitochondria crosstalk, and increased superoxide contribute to the pathophysiology in patients with ACAD9 deficiency. Furthermore, treatment of ACAD9 deficient cells with JP4-039, a novel mitochondria-targeted reactive oxygen species, electron and radical scavenger, decreased superoxide level and increased basal and maximal respiratory rate, identifying a potential therapeutic intervention opportunity in CI deficiency.


Assuntos
Acil-CoA Desidrogenases/genética , Complexo I de Transporte de Elétrons/deficiência , Fibroblastos/enzimologia , Doenças Mitocondriais/genética , NADH Desidrogenase/genética , Espécies Reativas de Oxigênio/metabolismo , Acil-CoA Desidrogenases/deficiência , Trifosfato de Adenosina/agonistas , Trifosfato de Adenosina/biossíntese , Transporte de Elétrons/efeitos dos fármacos , Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/genética , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Sequestradores de Radicais Livres/farmacologia , Expressão Gênica , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Doenças Mitocondriais/enzimologia , Doenças Mitocondriais/patologia , NADH Desidrogenase/deficiência , Óxidos de Nitrogênio/farmacologia , Fosforilação Oxidativa/efeitos dos fármacos , Cultura Primária de Células , Espécies Reativas de Oxigênio/antagonistas & inibidores
10.
J Clin Invest ; 86(3): 1000-3, 1990 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2394825

RESUMO

We sequenced polymerase chain reaction (PCR)-amplified variant medium chain acyl-CoA dehydrogenase (MCAD) cDNAs in cultured fibroblasts from three MCAD-deficient patients. In all three patients, an A to G transition was identified at position 985 of the coding region. Since no appropriate restriction sites for detecting this point mutation were found, we devised a PCR method that amplifies an 87-bp fragment from position 955. In the 5' primer encompassing positions 955 to 984, A-981 was artificially substituted with C. With the presence of C-981 and G-985, an Nco I restriction site is introduced in the mutant copies. When cDNA or genomic DNA from fibroblasts of nine MCAD-deficient patients were tested with this method, the copies from all of them completely cleaved into two shorter fragments by Nco I, indicating their homozygosity for the A----G-985 transition. In contrast, the copies from all eight controls remained intact. Thus, this A----G-985 transition is the single prevalent mutation causing MCAD deficiency, a highly unusual feature for any genetic disorder. The PCR/Nco I digestion method is suitable for the diagnosis of MCAD deficiency.


Assuntos
Acil-CoA Desidrogenases/deficiência , Acil-CoA Desidrogenase , Acil-CoA Desidrogenases/genética , Sequência de Bases , Desoxirribonucleases de Sítio Específico do Tipo II , Humanos , Dados de Sequência Molecular , Mutação , Reação em Cadeia da Polimerase , RNA Mensageiro/genética
11.
J Clin Invest ; 79(5): 1303-9, 1987 May.
Artigo em Inglês | MEDLINE | ID: mdl-3571488

RESUMO

We describe two patients with short-chain acyl-coenzyme A (CoA) dehydrogenase (SCADH) deficiency. Neonate I excreted large amounts of ethylmalonate and methylsuccinate; ethylmalonate excretion increased after a medium-chain triglyceride load. Neonate II died postnatally and excreted ethylmalonate, butyrate, 3-hydroxybutyrate, adipate, and lactate. Both neonates' fibroblasts catabolized [1-14C]butyrate poorly (29-64% of control). Neonate I had moderately decreased [1-14C]octanoate catabolism (43-60% of control), while neonate II oxidized this substrate normally; both catabolized radiolabeled palmitate, succinate, and/or leucine normally. Cell sonicates from neonates I and II dehydrogenated [2,3-3H]butyryl-CoA poorly (41 and 53% of control) and [2,3-3H]octanoyl-CoA more effectively (59 and 95% of control). Mitochondrial acyl-CoA dehydrogenase (ADH) activities with butyryl- and octanoyl-CoAs were 37 and 56% of control in neonate I, and 47 and 81% of control in neonate II, respectively. Monospecific medium-chain ADH (MCADH) antisera inhibited MCADH activity towards both butyryl- and octanoyl-CoAs, revealing SCADH activities to be 1 and 11% of control for neonates I and II, respectively. Fibroblast SCADH and MCADH activities were normal in an adult female with muscular SCADH deficiency.


Assuntos
Acil-CoA Desidrogenases/deficiência , Erros Inatos do Metabolismo Lipídico/enzimologia , Ácido 3-Hidroxibutírico , Adipatos/urina , Adulto , Butiratos/urina , Ácido Butírico , Feminino , Fibroblastos/enzimologia , Humanos , Hidroxibutiratos/urina , Lactatos/urina , Ácido Láctico , Erros Inatos do Metabolismo Lipídico/urina , Malonatos/urina , Succinatos/urina
12.
J Clin Invest ; 83(5): 1605-13, 1989 May.
Artigo em Inglês | MEDLINE | ID: mdl-2565344

RESUMO

Complementary DNAs encoding the precursor of human placental short chain acyl-coenzyme A (CoA) dehydrogenase (SCAD) (EC 1.3.99.2) were cloned and sequenced. The cDNA inserts in these clones were 1,852 bases in length combined, and encoded the entire 412-amino acid precursor SCAD (mol wt 44,303). This sequence included the 24-amino acid leader peptide moiety (mol wt 2,576) and 388 amino acids corresponding to the mature protein (mol wt 41,727). The comparison of SCAD and medium chain acyl-CoA dehydrogenase sequences revealed a high degree of homology, suggesting that these enzymes evolved from a common ancestral gene and belong to a gene family. We also studied mutant human SCAD in cultured skin fibroblasts from three patients with hereditary SCAD deficiency. Labeling fibroblast cultures with [35S]-methionine followed by immunoprecipitation with anti-SCAD antibody revealed that a normal size variant SCAD protein was synthesized. In all of the three SCAD-deficient cell lines, the size of variant SCAD mRNA as determined by Northern blotting using one of the normal SCAD cDNA as a probe was also normal, and no difference was observed on Southern blots in the restriction patterns of mutant genomic DNA using EcoRI, TaqI, HincII, and BamHI. These results suggest that the defects in SCAD in these cell lines are caused by a point mutation.


Assuntos
Acil-CoA Desidrogenases/genética , Clonagem Molecular , DNA/isolamento & purificação , Variação Genética , Acil-CoA Desidrogenase , Acil-CoA Desidrogenases/biossíntese , Acil-CoA Desidrogenases/deficiência , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Fibroblastos/metabolismo , Humanos , Fígado/enzimologia , Dados de Sequência Molecular , Placenta/enzimologia , Polimorfismo de Fragmento de Restrição , Biossíntese de Proteínas , Ratos , Homologia de Sequência do Ácido Nucleico
13.
J Clin Invest ; 81(1): 171-5, 1988 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3335634

RESUMO

Genetic deficiency of short-chain acyl-coenzyme A (CoA) dehydrogenase activity was demonstrated in cultured fibroblasts from a 2-yr-old female whose early postnatal life was complicated by poor feeding, emesis, and failure to thrive. She demonstrated progressive skeletal muscle weakness and developmental delay. Her plasma total carnitine level (35 nmol/ml) was low-normal, but was esterified to an abnormal degree (55% vs. control of less than 10%). Her skeletal muscle total carnitine level was low (7.6 nmol/mg protein vs. control of 14 +/- 2 nmol/mg protein) and was 75% esterified. Mild lipid deposition was noted in type I muscle fibers. Fibroblasts from this patient had 50% of control levels of acyl-CoA dehydrogenase activity towards butyryl-CoA as substrate at a concentration of 50 muM in a fluorometric assay based on the reduction of electron transfer flavoprotein. All of this residual activity was inhibited by an antibody against medium-chain acyl-CoA dehydrogenase. These data demonstrated that medium-chain acyl-CoA dehydrogenase accounted for 50% of the activity towards the short-chain substrate, butyryl-CoA, under these conditions, but that antibody against that enzyme could be used to unmask the specific and virtually complete deficiency of short-chain acyl-CoA dehydrogenase in this patient. Fibroblasts from her parents had intermediate levels of activity towards butyryl-CoA, consistent with the autosomal recessive inheritance of this metabolic defect.


Assuntos
Acil-CoA Desidrogenases/deficiência , Fibroblastos/enzimologia , Músculos/enzimologia , Acil-CoA Desidrogenase , Acil-CoA Desidrogenases/antagonistas & inibidores , Acil-CoA Desidrogenases/genética , Acil-CoA Desidrogenases/imunologia , Adolescente , Adulto , Anticorpos/fisiologia , Carnitina/deficiência , Células Cultivadas , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Músculos/fisiopatologia , Especificidade por Substrato
14.
J Clin Invest ; 76(3): 963-9, 1985 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3840178

RESUMO

Medium-chain acyl-coenzyme A (CoA) dehydrogenase (MCADH; EC 1.3.99.3) deficiency (MCD) is an inborn error of beta-oxidation. We measured 3H2O formed by the dehydrogenation of [2,3-3H]acyl-CoAs in a 3H-release assay. Short-chain acyl-CoA dehydrogenase (SCADH; EC 1.3.99.2), MCADH, and isovaleryl-CoA dehydrogenase (IVDH; EC 1.3.99.10) activities were assayed with 100 microM [2,3-3H]butyryl-, -octanoyl-, and -isovaleryl-CoAs, respectively, in fibroblasts cultured from normal controls and MCD patients. Without the artificial electron acceptor phenazine methosulfate (PMS), MCADH activity in fibroblast mitochondrial sonic supernatants (MS) was 54% of control in two MCD cell lines (P less than 0.05). Addition of 10 mM PMS raised control acyl-CoA dehydrogenase activities 16-fold and revealed MCADH and SCADH activities to be 5 (P less than 0.01) and 73% (P greater than 0.1) of control, respectively. Thus, the catalytic defect in MCD involves substrate binding and/or dehydrogenation by MCADH and not the subsequent reoxidation of reduced MCADH by electron acceptors. 20 microM flavin adenine dinucleotide (FAD) did not stimulate MCD MCADH activity in either the 3H-release or electron-transfer(ring) flavoprotein-linked dye-reduction assays. Mixing experiments revealed no MCADH inhibitor in MCD MS; IVDH activities were identical in both control and MCD MS. In postmortem liver MS from another MCD patient, 3H2O formation from [2,3-3H]octanoyl-CoA was 15% of control. When 3H2O formation was assayed with 200 microM [2,3-3H]acyl-CoAs, 15 mM PMS, and 20 microM FAD in fibroblast sonic supernatants from seven MCD cell lines, SCADH, MCADH, and IVDH activities were 72-112% (P greater than 0.1), 4-9% (P less than 0.01), and 86-135% (P greater than 0.1) of control, respectively, revealing no significant biochemical heterogeneity among these patients.


Assuntos
Acil-CoA Desidrogenases/deficiência , Mitocôndrias Hepáticas/enzimologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Acil Coenzima A/metabolismo , Acil-CoA Desidrogenases/metabolismo , Adulto , Fibroblastos/enzimologia , Flavina-Adenina Dinucleotídeo/farmacologia , Hemiterpenos , Humanos , Lactente , Isovaleril-CoA Desidrogenase , Erros Inatos do Metabolismo Lipídico/enzimologia , Masculino , Metilfenazônio Metossulfato/farmacologia , Oxirredutases/metabolismo , Ácidos Pentanoicos/sangue , Frações Subcelulares/enzimologia
15.
J Clin Invest ; 85(5): 1575-82, 1990 May.
Artigo em Inglês | MEDLINE | ID: mdl-1692038

RESUMO

Two distinct mutant alleles of the precursor (p) short chain acyl-CoA dehydrogenase (SCAD) gene were identified in a SCAD-deficient patient (YH2065) using the polymerase chain reaction to amplify cDNA synthesized from total RNA from her fibroblasts. Cells from this patient had previously been shown to synthesize a labile variant SCAD in contrast to the normal stability of variant SCADs in two other SCAD-deficient cell lines (Naito, E., Y. Indo, and K. Tanaka. 1989. J. Clin. Invest. 84:1671-1674). In the present study, both mutant alleles of YH2065 were found to contain a C----T transition, one at position 136 and the other at position 319 of the coding region of pSCAD cDNA. Clones of cDNA amplified from this region showed only one of the C----T transitions, indicating that each mutation was derived from different pSCAD alleles. Each of these mutations altered a known restriction endonuclease site, and restriction analysis of additional cDNA clones from amplified mutant cDNA and Southern blotting of mutant genomic DNA confirmed the presence of two unique mutant alleles in YH2065, indicating YH2065 is a compound heterozygote. These C----T transitions result in the substitution of Arg-22 and Arg-83 of the mature SCAD with Trp and Cys, respectively.


Assuntos
Acil-CoA Desidrogenases/genética , Variação Genética , Erros Inatos do Metabolismo Lipídico/genética , Mutação , Acil-CoA Desidrogenases/deficiência , Alelos , Southern Blotting , Butiril-CoA Desidrogenase , Linhagem Celular , Células Cultivadas , Clonagem Molecular , Feminino , Biblioteca Genômica , Humanos , Erros Inatos do Metabolismo Lipídico/enzimologia , Reação em Cadeia da Polimerase , RNA/genética , RNA/isolamento & purificação , RNA Mensageiro/genética , Valores de Referência , Mapeamento por Restrição
16.
J Clin Invest ; 94(4): 1477-83, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7929823

RESUMO

Medium chain acyl-CoA dehydrogenase (MCAD) deficiency is the most common known genetic disorder of fatty acid oxidation. Most (approximately 80%) cases are homozygous for a single mutation: A to G replacement at nucleotide 985 (A985G). MCAD deficiency typically presents in the second year of life as hypoketotic hypoglycemia associated with fasting and may progress to liver failure, coma, and death. Prompt diagnosis and management may prevent long-term sequelae. MCAD deficiency was verified by analysis of urinary acylglycine and serum acylcarnitine species from two neonates referred for diagnosis. Full-length cDNA and MCAD exon 7 and 11 genomic clones were prepared for sequence analysis. Normal and mutant cDNAs were expressed in bacteria, and enzymatic activity was assayed by the ferricenium hexaflurophosphate method. Four compound heterozygote individuals from two unrelated families with A985G on one allele and a novel G to A mutation at nucleotide 583 (G583A) as the second mutant allele presented with MCAD deficiency in the first week of life. The expressed G583A mutant protein lacks enzymatic activity. This novel mutation, G583A, is associated with severe MCAD deficiency causing hypoglycemia or sudden, unexpected neonatal death. This previously unrecognized phenotype of MCAD deficiency may contribute significantly to preventable infant deaths.


Assuntos
Acil-CoA Desidrogenases/deficiência , Acil-CoA Desidrogenases/genética , Erros Inatos do Metabolismo Lipídico/genética , Mutação Puntual/fisiologia , Morte Súbita do Lactente/etiologia , Acil-CoA Desidrogenase , Acil-CoA Desidrogenases/metabolismo , Sequência de Bases , Clonagem Molecular , Análise Mutacional de DNA , DNA Complementar , Escherichia coli/genética , Éxons/genética , Feminino , Expressão Gênica , Humanos , Hipoglicemia/genética , Recém-Nascido , Erros Inatos do Metabolismo Lipídico/enzimologia , Masculino , Dados de Sequência Molecular , Hibridização de Ácido Nucleico , Linhagem , RNA Mensageiro/análise
17.
Clin Biochem ; 40(3-4): 235-41, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17222812

RESUMO

OBJECTIVE: To determine the clinical efficacy and cost-effectiveness of newborn screening for MCADD using tandem mass spectrometry (MS/MS) compared with clinical diagnosis within the Canadian context. DESIGN AND METHODS: A systematic review of the clinical and economic literature was performed. For primary economic analysis, a decision-tree model was built based on the available information, the impact of newborn screening on the health care and the relevant Canadian data. RESULTS: Twenty-one clinical and two economic studies met the selection criteria. Mean incidence of MCADD was approximately 1:16,000. Clinical sensitivity and specificity were 100% and 99.99%, respectively. Screening significantly lowered morbidity and mortality. Both economic studies showed that screening for MCADD using MS/MS was cost-effective if willingness-to-pay was US 50,000 dollars. Our primary economic analysis showed that screening was cost-effective based on the cost-effective threshold of C 20,000 dollars per QALY. CONCLUSION: Screening consumes more resources than no screening but attains better health outcomes.


Assuntos
Acil-CoA Desidrogenases/deficiência , Erros Inatos do Metabolismo Lipídico/diagnóstico , Triagem Neonatal/economia , Triagem Neonatal/métodos , Espectrometria de Massas em Tandem/economia , Espectrometria de Massas em Tandem/métodos , Canadá , Análise Custo-Benefício , Humanos , Recém-Nascido
18.
Artigo em Inglês | MEDLINE | ID: mdl-9308370

RESUMO

Rapid progress in DNA technology has entailed the possibility of readily detecting mutations in disease genes. In contrast to this, techniques to characterize the effects of mutations are still very time consuming. It has turned out that many of the mutations detected in disease genes are missense mutations. Characterization of the effect of these mutations is particularly important in order to establish that they are disease causing and to estimate their severity. We use the experiences with investigation of medium-chain acyl-CoA dehydrogenase deficiency as an example to illustrate that (i) impaired folding is a common effect of missense mutations occurring in genetic diseases, (ii) increasing the level of available chaperones may augment the level of functional mutant protein in vivo, and (iii) one mutation may have multiple effects. The interplay between the chaperones assisting folding and proteases that attack folding intermediates is decisive for how large a proportion of a mutant polypeptide impaired in folding acquires the functional structure. This constitutes a protein quality control system, and the handling of a given mutant protein by this system may vary due to environmental conditions or genetic variability in its components. The possibility that intraindividual differences in the handling of mutant proteins may be a mechanism accounting for phenotypic variability is discussed.


Assuntos
Acil-CoA Desidrogenases/química , Acil-CoA Desidrogenases/deficiência , Erros Inatos do Metabolismo/enzimologia , Acil-CoA Desidrogenase , Acil-CoA Desidrogenases/genética , Animais , Ácidos Graxos/metabolismo , Humanos , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Mitocôndrias/metabolismo , Modelos Biológicos , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Mutação , Conformação Proteica , Dobramento de Proteína
20.
Hum Pathol ; 49: 27-32, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26826406

RESUMO

Complex I deficiency causes Leigh syndrome, fatal infant lactic acidosis, and neonatal cardiomyopathy. Mutations in more than 100 nuclear DNA and mitochondrial DNA genes miscode for complex I subunits or assembly factors. ACAD9 is an acyl-CoA dehydrogenase with a novel function in assembly of complex I; biallelic mutations cause progressive encephalomyopathy, recurrent Reye syndrome, and fatal cardiomyopathy. We describe the first autopsy in fatal neonatal lethal lactic acidosis due to mutations in ACAD9 that reduced complex I activity. We identified mitochondrial hyperplasia in cardiac myocytes, diaphragm muscle, and liver and renal tubules in formalin-fixed, paraffin-embedded tissue using immunohistochemistry for mitochondrial antigens. Whole-exome sequencing revealed compound heterozygous variants in the ACAD9 gene: c.187G>T (p.E63*) and c.941T>C (p.L314P). The nonsense mutation causes late infantile lethality; the missense variant is novel. Autopsy-derived fibroblasts had reduced complex I activity (53% of control) with normal activity in complexes II to IV, similar to reported cases of ACAD9 deficiency.


Assuntos
Acidose Láctica/diagnóstico , Acidose/diagnóstico , Acil-CoA Desidrogenase/deficiência , Acil-CoA Desidrogenases/genética , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Cardiomiopatia Hipertrófica/diagnóstico , Códon sem Sentido , Diafragma/patologia , Complexo I de Transporte de Elétrons/deficiência , Túbulos Renais/patologia , Doença de Leigh/diagnóstico , Mitocôndrias Cardíacas/patologia , Mitocôndrias Hepáticas/patologia , Mitocôndrias Musculares/patologia , Doenças Mitocondriais/diagnóstico , Insuficiência de Múltiplos Órgãos/diagnóstico , Debilidade Muscular/diagnóstico , Acidose/enzimologia , Acidose/genética , Acidose/patologia , Acidose Láctica/enzimologia , Acidose Láctica/genética , Acidose Láctica/patologia , Acil-CoA Desidrogenase/genética , Acil-CoA Desidrogenases/deficiência , Erros Inatos do Metabolismo dos Aminoácidos/enzimologia , Erros Inatos do Metabolismo dos Aminoácidos/genética , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Autopsia , Cardiomiopatia Hipertrófica/enzimologia , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/patologia , Causas de Morte , Células Cultivadas , Análise Mutacional de DNA , DNA Mitocondrial/genética , Diafragma/enzimologia , Complexo I de Transporte de Elétrons/genética , Evolução Fatal , Fibroblastos/enzimologia , Fibroblastos/patologia , Predisposição Genética para Doença , Humanos , Hiperplasia , Imuno-Histoquímica , Recém-Nascido , Túbulos Renais/enzimologia , Doença de Leigh/enzimologia , Doença de Leigh/genética , Doença de Leigh/patologia , Masculino , Mitocôndrias Cardíacas/enzimologia , Mitocôndrias Hepáticas/enzimologia , Mitocôndrias Musculares/enzimologia , Doenças Mitocondriais/enzimologia , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia , Insuficiência de Múltiplos Órgãos/enzimologia , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/patologia , Debilidade Muscular/enzimologia , Debilidade Muscular/genética , Debilidade Muscular/patologia , Fenótipo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA