Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Inherit Metab Dis ; 47(4): 731-745, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38356271

RESUMEN

Medium-chain acyl-CoA dehydrogenase deficiency (MCADD) is the most prevalent mitochondrial fatty acid ß-oxidation disorder. In this study, we assessed the variability of the lipid profile in MCADD by analysing plasma samples obtained from 25 children with metabolically controlled MCADD (following a normal diet with frequent feeding and under l-carnitine supplementation) and 21 paediatric control subjects (CT). Gas chromatography-mass spectrometry was employed for the analysis of esterified fatty acids, while high-resolution C18-liquid chromatography-mass spectrometry was used to analyse lipid species. We identified a total of 251 lipid species belonging to 15 distinct lipid classes. Principal component analysis revealed a clear distinction between the MCADD and CT groups. Univariate analysis demonstrated that 126 lipid species exhibited significant differences between the two groups. The lipid species that displayed the most pronounced variations included triacylglycerols and phosphatidylcholines containing saturated and monounsaturated fatty acids, specifically C14:0 and C16:0, which were found to be more abundant in MCADD. The observed changes in the plasma lipidome of children with non-decompensated MCADD suggest an underlying alteration in lipid metabolism. Therefore, longitudinal monitoring and further in-depth investigations are warranted to better understand whether such alterations are specific to MCADD children and their potential long-term impacts.


Asunto(s)
Acil-CoA Deshidrogenasa , Errores Innatos del Metabolismo Lipídico , Lipidómica , Fosfolípidos , Triglicéridos , Humanos , Errores Innatos del Metabolismo Lipídico/sangre , Lipidómica/métodos , Niño , Masculino , Femenino , Triglicéridos/sangre , Fosfolípidos/sangre , Preescolar , Acil-CoA Deshidrogenasa/deficiencia , Lactante , Adolescente , Metabolismo de los Lípidos , Estudios de Casos y Controles , Ácidos Grasos/sangre , Ácidos Grasos/metabolismo , Cromatografía de Gases y Espectrometría de Masas/métodos , Carnitina/sangre
2.
Mol Genet Metab ; 129(1): 13-19, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31836396

RESUMEN

BACKGROUND: Medium-Chain Acyl-CoA Dehydrogenase (MCAD) deficiency is a fatty acid oxidation disorder that can have variable clinical severity. There is still limited information on its clinical presentation and longitudinal history by genotype, and effectiveness of newborn screening (NBS). METHODS: Retrospective data were collected from 90 patients (44 female, 46 male) to compare biochemical data with clinical outcomes. The frequency of adverse events (number of hypoglycemia-related ER visits and admissions) was assessed by genotype (homozygosity or not for the common pathogenic variant, p.Lys329Glu, in the ACADM gene), and method of diagnosis (NBS vs. clinical). RESULTS: MCAD deficiency in Utah was more frequent compared to the United States average (1: 9266 versus 1:17,759 newborns). With age, C8-carnitine did not change significantly whereas C2-carnitine decreased (p < .001), possibly reflecting reduced carnitine supplementation typically seen with age. Children with MCAD deficiency had normal growth. p.Lys329Glu homozygotes had higher NBS C8-carnitine (23.4 ± 19.6 vs. 6.6 ± 3.0 µmol/L) and lifetime plasma C8-carnitine levels (6.2 ± 5 vs. 3.6 ± 1.9 µmol/L) compared to patients with at least one other pathogenic variant (p < .001 for both) and higher transaminases compared to compound heterozygotes (ALT 41.9 ± 6.2 vs. 31.5 ± 3.7 U/L, AST 63.9 ± 5.8 vs. 45.7 ± 1.8 U/L, p < .05 for both). On average, p.Lys329Glu homozygotes had more hypoglycemic events than compound heterozygotes (1.44 versus 0.49 events/patient) as did patients diagnosed clinically compared to those diagnosed by NBS (2.15 versus 0.62 events/patient), though these differences were not statistically significant. Neonatal death was observed before results of newborn screening were available in one patient homozygous for the common p.Lys329Glu pathogenic variant, but severe neonatal complications (hypoglycemia, cardiac arrhythmia) were also seen in patients with other mutations. No irreversible complications were observed after diagnosis in any patient with MCAD deficiency. DISCUSSION: Homozygosity for the common ACADM p.Lys329Glu pathogenic variant was associated with increased levels of C8-carnitine and transaminases. Newborn screening provides the opportunity to reduce morbidity and post-neonatal mortality in all patients with MCAD deficiency, regardless of genotype.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Genotipo , Homocigoto , Errores Innatos del Metabolismo Lipídico/diagnóstico , Tamizaje Neonatal , Acil-CoA Deshidrogenasa/genética , Adolescente , Adulto , Carnitina/sangre , Niño , Preescolar , Femenino , Variación Genética , Humanos , Recién Nacido , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/mortalidad , Masculino , Mutación , Fenotipo , Estudios Retrospectivos , Transaminasas/sangre , Estados Unidos , Utah , Adulto Joven
3.
Curr Mol Med ; 19(7): 487-493, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31418342

RESUMEN

BACKGROUND: Multiple Acyl-CoA Dehydrogenase Deficiency (MADD) is a congenital rare metabolic disease with broad clinical phenotypes and variable evolution. This inborn error of metabolism is caused by mutations in the ETFA, ETFB or ETFDH genes, which encode for the mitochondrial ETF and ETF:QO proteins. A considerable group of patients has been described to respond positively to riboflavin oral supplementation, which constitutes the prototypic treatment for the pathology. OBJECTIVES: To report mutations in ETFA, ETFB and ETFDH genes identified in Portuguese patients, correlating, whenever possible, biochemical and clinical outcomes with the effects of mutations on the structure and stability of the affected proteins, to better understand MADD pathogenesis at the molecular level. METHODS: MADD patients were identified based on the characteristic urinary profile of organic acids and/or acylcarnitine profiles in blood spots during newborn screening. Genotypic, clinical and biochemical data were collected for all patients. In silico structural analysis was employed using bioinformatic tools carried out in an ETF:QO molecular model for the identified missense mutations. RESULTS: A survey describing clinical and biochemical features of eight Portuguese MADD patients was made. Genotype analysis identified five ETFDH mutations, including one extension (p.X618QextX*14), two splice mutations (c.34+5G>C and c.405+3A>T) and two missense mutations (ETF:QO-p.Arg155Gly and ETF:QO-p.Pro534Leu), and one ETFB mutation (ETFß- p.Arg191Cys). Homozygous patients containing the ETFDH mutations p.X618QextX*14, c.34+5G>C and ETF:QO-p.Arg155Gly, all presented severe (lethal) MADD phenotypes. However, when any of these mutations are in heterozygosity with the known ETF:QO-p.Pro534Leu mild variant, the severe clinical effects are partly and temporarily attenuated. Indeed, the latter destabilizes an ETF-interacting loop, with no major functional consequences. However, the position 155 in ETF:QO is localized at the ubiquinone binding and membrane interacting domain, and is thus expected to perturb protein structure and membrane insertion, with severe functional effects. Structural analysis of molecular models is therefore demonstrated to be a valuable tool to rationalize the effects of mutations in the context of the clinical phenotype severity. CONCLUSION: Advanced molecular diagnosis, structural analysis and clinical correlations reveal that MADD patients harboring a severe prognosis mutation in one allele can actually revert to a milder phenotype by complementation with a milder mutation in the other allele. However, such patients are nevertheless in a precarious metabolic balance which can revert to severe fatal outcomes during catabolic stress or secondary pathology, thus requiring strict clinical follow-up.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/genética , Proteínas Hierro-Azufre/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/genética , Alelos , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Recién Nacido , Masculino , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/patología , Mutación Missense/genética , Tamizaje Neonatal , Portugal/epidemiología , Embarazo , Pronóstico , Riboflavina/genética , Riboflavina/metabolismo
4.
J Inherit Metab Dis ; 42(1): 159-168, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30740737

RESUMEN

BACKGROUND: Patients with very long chain acyl-CoA dehydrogenase deficiency (VLCADD), a long chain fatty acid oxidation disorder, are traditionally treated with a long chain triglyceride (LCT) restricted and medium chain triglyceride (MCT) supplemented diet. Introduction of VLCADD in newborn screening (NBS) programs has led to the identification of asymptomatic newborns with VLCADD, who may have a more attenuated phenotype and may not need dietary adjustments. OBJECTIVE: To define dietary strategies for individuals with VLCADD based on the predicted phenotype. METHOD: We evaluated long-term dietary histories of a cohort of individuals diagnosed with VLCADD identified before the introduction of VLCADD in NBS and their beta-oxidation (LC-FAO) flux score (rate of oleate oxidation) in cultured skin fibroblasts in relation to the clinical outcome. Based on these results a dietary strategy is proposed. RESULTS: Sixteen individuals with VLCADD were included. One had an LC-FAO flux score >90%, was not on a restricted diet and is asymptomatic to date. Four patients had an LC-FAO flux score <10%, and significant VLCADD related symptoms despite the use of strict diets including LCT restriction, MCT supplementation and nocturnal gastric drip feeding. Patients with an LC-FAO flux score between 10 and 90% (n = 11) showed a more heterogeneous phenotype. CONCLUSIONS: This study shows that a strict diet cannot prevent poor clinical outcome in severely affected patients and that the LC-FAO flux is a good predictor of clinical outcome in individuals with VLCADD identified before its introduction in NBS. Hereby, we propose an individualized dietary strategy based on the LC-FAO flux score.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Acil-CoA Deshidrogenasa/deficiencia , Síndromes Congénitos de Insuficiencia de la Médula Ósea/tratamiento farmacológico , Errores Innatos del Metabolismo Lipídico/tratamiento farmacológico , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Musculares/tratamiento farmacológico , Acil-CoA Deshidrogenasa de Cadena Larga/metabolismo , Síndromes Congénitos de Insuficiencia de la Médula Ósea/metabolismo , Dieta , Ácidos Grasos/administración & dosificación , Femenino , Humanos , Recién Nacido , Errores Innatos del Metabolismo Lipídico/metabolismo , Masculino , Enfermedades Mitocondriales/metabolismo , Enfermedades Musculares/metabolismo , Tamizaje Neonatal/métodos , Fenotipo , Triglicéridos/administración & dosificación
5.
Orphanet J Rare Dis ; 13(1): 120, 2018 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-30025539

RESUMEN

BACKGROUND: Mitochondrial acyl-CoA dehydrogenase family member 9 (ACAD9) is essential for the assembly of mitochondrial respiratory chain complex I. Disease causing biallelic variants in ACAD9 have been reported in individuals presenting with lactic acidosis and cardiomyopathy. RESULTS: We describe the genetic, clinical and biochemical findings in a cohort of 70 patients, of whom 29 previously unpublished. We found 34 known and 18 previously unreported variants in ACAD9. No patients harbored biallelic loss of function mutations, indicating that this combination is unlikely to be compatible with life. Causal pathogenic variants were distributed throughout the entire gene, and there was no obvious genotype-phenotype correlation. Most of the patients presented in the first year of life. For this subgroup the survival was poor (50% not surviving the first 2 years) comparing to patients with a later presentation (more than 90% surviving 10 years). The most common clinical findings were cardiomyopathy (85%), muscular weakness (75%) and exercise intolerance (72%). Interestingly, severe intellectual deficits were only reported in one patient and severe developmental delays in four patients. More than 70% of the patients were able to perform the same activities of daily living when compared to peers. CONCLUSIONS: Our data show that riboflavin treatment improves complex I activity in the majority of patient-derived fibroblasts tested. This effect was also reported for most of the treated patients and is mirrored in the survival data. In the patient group with disease-onset below 1 year of age, we observed a statistically-significant better survival for patients treated with riboflavin.


Asunto(s)
Acidosis/genética , Acidosis/metabolismo , Acil-CoA Deshidrogenasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Debilidad Muscular/genética , Debilidad Muscular/metabolismo , Riboflavina/uso terapéutico , Acidosis/patología , Actividades Cotidianas , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasa/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/patología , Cardiomiopatía Hipertrófica/patología , Complejo I de Transporte de Electrón/metabolismo , Femenino , Humanos , Masculino , Enfermedades Mitocondriales/patología , Debilidad Muscular/tratamiento farmacológico , Debilidad Muscular/patología , Pronóstico
6.
Zhonghua Er Ke Za Zhi ; 54(12): 927-930, 2016 Dec 02.
Artículo en Chino | MEDLINE | ID: mdl-27938594

RESUMEN

Objective: To investigate the incidence, clinical, biochemical and gene mutation characteristics of short chain acyl-coenzyme A dehydrogenase deficiency (SCADD). Method: From January, 2009 to October, 2015, a retrospective analysis of the urine organic acids and acyl-coenzyme A dehydrogenase (ACADS) gene mutation characteristics of patients diagnosed as SCADD by newborn screening using tandem mass spectrometry in Department of Genetics and Metabolism (Newborn screening Center of Zhejiang Province), Children's Hospital, Zhejiang University School of Medicine. Dietary guidance, life management and supplementation of L-carnitine were conducted, and growth and intelligence development were observed during follow-up among the SCADD patients. Result: A total of 1 430 024 neonates, seventeen cases were diagnosed with SCADD with an incidence of 1/84 117. All patients had no clinical symptoms, and intelligence and physical development were normal. Blood butylacyl-carnitine (C4) levels and the ratios increased, C4 0.713.14 µmol/L(reference value 0.03-0.48 µmol/L), C4/C2 0.07-0.23(reference value 0.01-0.04), C4/C3 0.65-2.04(reference value 0.05-0.39). Thirteen with increased urinary ethyl malonic acid (9.30-90.99 mg/g creatinine (reference value 0-6.20 mg/g creatinine )), one patient was accompanied by increased methyl succinic acid (12.33 mg/g creatinine(reference value 0-6.40 mg/g creatinine)), one subject with increased acetylglycine (3.52 mg/g creatinine(reference value 0-0.70 mg/g creatinine)). A total of 13 known mutations were detected in the ACADS gene, 1 homozygous mutation (c.1031A>G), the others are compound heterozygous mutations. One frameshift mutation (c.508_509delGC) and 12 missense mutations were detected. Common mutation were c. 1031A>G(35.3%), c. 164C>T(20.6%) and c. 991G>A(11.8%). SCADD in newborn screening program had no clinical symptoms and normal growth development after 8-42 months follow-up. Conclusion: Cases with SCADD had no clinical symptoms with an incidence of 1/84117. The c. 164C>T and c. 1031A>G may be the common mutations.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo Lipídico/genética , Tamizaje Neonatal/métodos , Acil-CoA Deshidrogenasa/sangre , Acil-CoA Deshidrogenasa/genética , Errores Innatos del Metabolismo de los Aminoácidos/sangre , Errores Innatos del Metabolismo de los Aminoácidos/etnología , Carnitina/sangre , Niño , China/epidemiología , Homocigoto , Humanos , Incidencia , Recién Nacido , Errores Innatos del Metabolismo Lipídico/sangre , Errores Innatos del Metabolismo Lipídico/etnología , Masculino , Mutación , Estudios Retrospectivos , Espectrometría de Masas en Tándem
7.
J Inherit Metab Dis ; 37(5): 783-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24623196

RESUMEN

The objective of this study was to test whether macromolecule oxidative damage and altered enzymatic antioxidative defenses occur in patients with medium-chain acyl coenzyme A dehydrogenase (MCAD) deficiency. We performed a cross-sectional observational study of in vivo parameters of lipid and protein oxidative damage and antioxidant defenses in asymptomatic, nonstressed, MCAD-deficient patients and healthy controls. Patients were subdivided into three groups based on therapy: patients without prescribed supplementation, patients with carnitine supplementation, and patients with carnitine plus riboflavin supplementation. Compared with healthy controls, nonsupplemented MCAD-deficient patients and patients receiving carnitine supplementation displayed decreased plasma sulfhydryl content (indicating protein oxidative damage). Increased erythrocyte superoxide dismutase (SOD) activity in patients receiving carnitine supplementation probably reflects a compensatory mechanism for scavenging reactive species formation. The combination of carnitine plus riboflavin was not associated with oxidative damage. These are the first indications that MCAD-deficient patients experience protein oxidative damage and that combined supplementation of carnitine and riboflavin may prevent these biochemical alterations. Results suggest involvement of free radicals in the pathophysiology of MCAD deficiency. The underlying mechanisms behind the increased SOD activity upon carnitine supplementation need to be determined. Further studies are necessary to determine the clinical relevance of oxidative stress, including the possibility of antioxidant therapy.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Antioxidantes/metabolismo , Errores Innatos del Metabolismo Lipídico/metabolismo , Estrés Oxidativo , Proteínas/metabolismo , Acil-CoA Deshidrogenasa/metabolismo , Adolescente , Adulto , Carnitina/uso terapéutico , Niño , Preescolar , Estudios Transversales , Eritrocitos/metabolismo , Femenino , Humanos , Lactante , Recién Nacido , Metabolismo de los Lípidos/genética , Masculino , Riboflavina/uso terapéutico , Vitaminas/uso terapéutico , Adulto Joven
9.
Orphanet J Rare Dis ; 8: 102, 2013 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-23842438

RESUMEN

BACKGROUND: Medium-chain acyl-CoA dehydrogenase deficiency (MCADD) is the most common inherited defect in the mitochondrial fatty acid oxidation pathway, resulting in significant morbidity and mortality in undiagnosed patients.Newborn screening (NBS) has considerably improved MCADD outcome, but the risk of complication remains in some patients. The aim of this study was to evaluate the relationship between genotype, biochemical parameters and clinical data at diagnosis and during follow-up, in order to optimize monitoring of these patients. METHODS: We carried out a multicenter study in southwest Europe, of MCADD patients detected by NBS. Evaluated NBS data included free carnitine (C0) and the acylcarnitines C8, C10, C10:1 together with C8/C2 and C8/C10 ratios, clinical presentation parameters and genotype, in 45 patients. Follow-up data included C0 levels, duration of carnitine supplementation and occurrence of metabolic crises. RESULTS: C8/C2 ratio and C8 were the most accurate biomarkers of MCADD in NBS. We found a high number of patients homozygous for the prevalent c.985A > G mutation (75%). Moreover, in these patients C8, C8/C10 and C8/C2 were higher than in patients with other genotypes, while median value of C0 was significantly lower (23 µmol/L vs 36 µmol/L).The average follow-up period was 43 months. To keep carnitine levels within the normal range, carnitine supplementation was required in 82% of patients, and for a longer period in patients homozygotes for the c.985A>G mutation than in patients with other genotypes (average 31 vs 18 months). Even with treatment, median C0 levels remained lower in homozygous patients than in those with other genotypes (14 µmol/L vs 22 µmol/L).Two patients died and another three suffered a metabolic crisis, all of whom were homozygous for the c.985 A>G mutation. CONCLUSIONS: Our data show a direct association between homozygosity for c.985A>G and lower carnitine values at diagnosis, and a higher dose of carnitine supplementation for maintenance within the normal range. This study contributes to a better understanding of the relationship between genotype and phenotype in newborn patients with MCADD detected through screening which could be useful in improving follow-up strategies and clinical outcome.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Cardiomiopatías/epidemiología , Carnitina/sangre , Carnitina/deficiencia , Hiperamonemia/epidemiología , Errores Innatos del Metabolismo Lipídico/diagnóstico , Enfermedades Musculares/epidemiología , Tamizaje Neonatal/métodos , Cardiomiopatías/diagnóstico , Cardiomiopatías/tratamiento farmacológico , Carnitina/administración & dosificación , Carnitina/análogos & derivados , Suplementos Dietéticos , Femenino , Estudios de Asociación Genética , Genotipo , Homocigoto , Humanos , Hiperamonemia/diagnóstico , Hiperamonemia/tratamiento farmacológico , Incidencia , Recién Nacido , Errores Innatos del Metabolismo Lipídico/tratamiento farmacológico , Errores Innatos del Metabolismo Lipídico/epidemiología , Masculino , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/tratamiento farmacológico , Fenotipo , Prevalencia , España/epidemiología , Complejo Vitamínico B/administración & dosificación , Complejo Vitamínico B/sangre
10.
J Clin Endocrinol Metab ; 98(4): 1667-75, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23426616

RESUMEN

BACKGROUND: It is not clear to what extent skeletal muscle is affected in patients with medium-chain acyl-coenzyme A dehydrogenase deficiency (MCADD). l-Carnitine is commonly used as a supplement in patients with MCADD, although its beneficial effect has not been verified. DESIGN: We investigated (1) fuel utilization during prolonged low-intensity exercise in patients with MCADD and (2) the influence of 4 weeks of oral l-carnitine supplementation on fuel utilization during exercise. METHODS: Four asymptomatic patients with MCADD and 11 untrained, healthy, age- and sex-matched control subjects were included. The subjects performed a 1-hour cycling test at a constant workload corresponding to 55% of Vo2max, while fat and carbohydrate metabolism was assessed, using the stable isotope technique and indirect calorimetry. The patients ingested 100 mg/kg/d of l-carnitine for 4 weeks, after which the cycling tests were repeated. RESULTS: At rest, palmitate oxidation and total fatty acid oxidation (FAO) rates were similar in patients and healthy control subjects. During constant workload cycling, palmitate oxidation and FAO rates increased in both groups, but increased 2 times as much in healthy control subjects as in patients (P = .007). Palmitate oxidation and FAO rates were unchanged by the l-carnitine supplementation. CONCLUSION: Our results indicate that patients with MCADD have an impaired ability to increase FAO during exercise but less so than that observed in patients with a number of other disorders of fat oxidation, which explains the milder skeletal muscle phenotype in MCADD. The use of carnitine supplementation in MCADD cannot be supported by the present findings.


Asunto(s)
Carnitina/farmacología , Ejercicio Físico/fisiología , Errores Innatos del Metabolismo Lipídico/metabolismo , Metabolismo de los Lípidos/fisiología , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/metabolismo , Adolescente , Adulto , Carnitina/administración & dosificación , Suplementos Dietéticos , Prueba de Esfuerzo , Tolerancia al Ejercicio/efectos de los fármacos , Tolerancia al Ejercicio/fisiología , Femenino , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Oxidación-Reducción/efectos de los fármacos , Proyectos de Investigación , Adulto Joven
11.
PLoS One ; 7(8): e41709, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22936979

RESUMEN

BACKGROUND: How signals from fatty acid metabolism are translated into changes in food intake remains unclear. Previously we reported that mice with a genetic inactivation of Acads (acyl-coenzyme A dehydrogenase, short-chain), the enzyme responsible for mitochondrial beta-oxidation of C4-C6 short-chain fatty acids (SCFAs), shift consumption away from fat and toward carbohydrate when offered a choice between diets. In the current study, we sought to indentify candidate genes and pathways underlying the effects of SCFA oxidation deficiency on food intake in Acads-/- mice. METHODOLOGY/PRINCIPAL FINDINGS: We performed a transcriptional analysis of gene expression in brain tissue of Acads-/- and Acads+/+ mice fed either a high-fat (HF) or low-fat (LF) diet for 2 d. Ingenuity Pathway Analysis revealed three top-scoring pathways significantly modified by genotype or diet: oxidative phosphorylation, mitochondrial dysfunction, and CREB signaling in neurons. A comparison of statistically significant responses in HF Acads-/- vs. HF Acads+/+ (3917) and Acads+/+ HF vs. LF Acads+/+ (3879) revealed 2551 genes or approximately 65% in common between the two experimental comparisons. All but one of these genes were expressed in opposite direction with similar magnitude, demonstrating that HF-fed Acads-deficient mice display transcriptional responses that strongly resemble those of Acads+/+ mice fed LF diet. Intriguingly, genes involved in both AMP-kinase regulation and the neural control of food intake followed this pattern. Quantitative RT-PCR in hypothalamus confirmed the dysregulation of genes in these pathways. Western blotting showed an increase in hypothalamic AMP-kinase in Acads-/- mice and HF diet increased, a key protein in an energy-sensing cascade that responds to depletion of ATP. CONCLUSIONS: Our results suggest that the decreased beta-oxidation of short-chain fatty acids in Acads-deficient mice fed HF diet produces a state of energy deficiency in the brain and that AMP-kinase may be the cellular energy-sensing mechanism linking fatty acid oxidation to feeding behavior in this model.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Dieta Alta en Grasa/efectos adversos , Transducción de Señal/efectos de los fármacos , Acil-CoA Deshidrogenasa/genética , Adenosina Trifosfato/metabolismo , Animales , Encéfalo , Ácidos Grasos/metabolismo , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
12.
J Inherit Metab Dis ; 35(1): 115-23, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21630065

RESUMEN

INTRODUCTION: There is little robust empirical evidence on which to base treatment recommendations for fatty acid oxidation disorders. While consensus guidelines are important, understanding areas where there is a lack of consensus is also critical to inform priorities for future evaluative research. METHODS: We surveyed Canadian metabolic physicians on the treatment of medium-chain acyl-CoA dehydrogenase (MCAD) deficiency, very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency, long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency, and mitochondrial trifunctional protein (MTP) deficiency. We ascertained physicians' opinions on the use of different interventions for the long-term management of patients as well as for the management of acute illness, focusing on identifying interventions characterized by high variability in opinions. We also investigated factors influencing treatment decisions. RESULTS: We received 18 responses (response rate 45%). Participants focused on avoidance of fasting and increased meal frequency as interventions for the management of MCAD deficiency. For the long-chain disorders, avoidance of fasting remained the most consistently endorsed intervention, with additional highly endorsed treatments differing for VLCAD versus LCHAD/MTP deficiency. L-carnitine supplementation and restriction of dietary fat were characterized by high variability in physicians' opinions, as were several interventions specific to long-chain disorders. Social factors and patient characteristics were important influences on treatment decisions. CONCLUSIONS: Based on our findings we suggest that high priority treatments for rigorous effectiveness studies could include L-carnitine supplementation (MCAD and LCHAD/MTP deficiencies), restriction of dietary fat, and, for the long-chain disorders, feeding practices for breastfed infants and the use of various supplements (essential fatty acids, carbohydrates, cornstarch, multivitamins).


Asunto(s)
Ácidos Grasos/metabolismo , Errores Innatos del Metabolismo Lipídico/diagnóstico , Errores Innatos del Metabolismo Lipídico/terapia , Oxígeno/química , 3-Hidroxiacil-CoA Deshidrogenasas/deficiencia , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Canadá , Carnitina/uso terapéutico , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Grasas de la Dieta , Conocimientos, Actitudes y Práctica en Salud , Humanos , 3-Hidroxiacil-CoA Deshidrogenasa de Cadena Larga , Enfermedades Mitocondriales/terapia , Proteína Trifuncional Mitocondrial , Complejos Multienzimáticos/deficiencia , Enfermedades Musculares/terapia , Encuestas y Cuestionarios
13.
Mol Cell Biochem ; 361(1-2): 329-35, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22015654

RESUMEN

The accumulation of octanoic (OA) and decanoic (DA) acids in tissue is the common finding in medium-chain acyl-coenzyme A dehydrogenase deficiency (MCADD), the most frequent defect of fatty acid oxidation. Affected patients present hypoketotic hypoglycemia, rhabdomyolysis, hepatomegaly, seizures and lethargy, which may progress to coma and death. At present, the pathophysiological mechanisms underlying hepatic and skeletal muscle alterations in affected patients are poorly known. Therefore, in the present work, we investigated the in vitro effects of OA and DA, the accumulating metabolites in MCADD, on various bioenergetics and oxidative stress parameters. It was verified that OA and DA decreased complexes I-III, II-III and IV activities in liver and also inhibit complex IV activity in skeletal muscle. In addition, DA decreased complexes II-III activity in skeletal muscle. We also verified that OA and DA increased TBA-RS levels and carbonyl content in both tissues. Finally, DA, but not OA, significantly decreased GSH levels in rat skeletal muscle. Our present data show that the medium-chain fatty acids that accumulate in MCADD impair electron transfer through respiratory chain and elicit oxidative damage in rat liver and skeletal muscle. It may be therefore presumed that these mechanisms are involved in the pathophysiology of the hepatopathy and rhabdomyolysis presented by MCADD-affected patients.


Asunto(s)
Caprilatos/metabolismo , Decanoatos/metabolismo , Hígado/metabolismo , Músculo Esquelético/metabolismo , Estrés Oxidativo , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/metabolismo , Animales , Caprilatos/farmacología , Creatina Quinasa/metabolismo , Decanoatos/farmacología , Transporte de Electrón , Complejo IV de Transporte de Electrones/metabolismo , Glutatión/metabolismo , Errores Innatos del Metabolismo Lipídico/metabolismo , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Mitocondrias/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/enzimología , Oxidación-Reducción , Carbonilación Proteica , Ratas , Ratas Wistar , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
14.
Curr Neurol Neurosci Rep ; 11(1): 97-103, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21046290

RESUMEN

Lipid storage myopathy (LSM) is pathologically characterized by prominent lipid accumulation in muscle fibers due to lipid dysmetabolism. Although extensive molecular studies have been performed, there are only four types of genetically diagnosable LSMs: primary carnitine deficiency (PCD), multiple acyl-coenzyme A dehydrogenase deficiency (MADD), neutral lipid storage disease with ichthyosis, and neutral lipid storage disease with myopathy. Making an accurate diagnosis, by specific laboratory tests including genetic analyses, is important for LSM as some of the patients are treatable: individuals with PCD show dramatic improvement with high-dose oral L-carnitine supplementation and increasing evidence indicates that MADD due to ETFDH mutations is riboflavin responsive.


Asunto(s)
Metabolismo de los Lípidos , Músculo Esquelético/patología , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/genética , Carnitina/deficiencia , Carnitina/genética , Diagnóstico Diferencial , Humanos , Eritrodermia Ictiosiforme Congénita/genética , Eritrodermia Ictiosiforme Congénita/patología , Eritrodermia Ictiosiforme Congénita/fisiopatología , Eritrodermia Ictiosiforme Congénita/terapia , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/patología , Errores Innatos del Metabolismo Lipídico/fisiopatología , Errores Innatos del Metabolismo Lipídico/terapia , Músculo Esquelético/fisiopatología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Enfermedades Musculares/terapia , Enfermedades de la Piel/enzimología , Enfermedades de la Piel/genética , Enfermedades de la Piel/patología , Enfermedades de la Piel/fisiopatología
15.
Life Sci ; 87(5-6): 139-46, 2010 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-20540954

RESUMEN

AIMS: In the present work we investigated the in vitro effect of cis-4-decenoic acid, the pathognomonic metabolite of medium-chain acyl-CoA dehydrogenase deficiency, on various parameters of bioenergetic homeostasis in rat brain mitochondria. MAIN METHODS: Respiratory parameters determined by oxygen consumption were evaluated, as well as membrane potential, NAD(P)H content, swelling and cytochrome c release in mitochondrial preparations from rat brain, using glutamate plus malate or succinate as substrates. The activities of citric acid cycle enzymes were also assessed. KEY FINDINGS: cis-4-decenoic acid markedly increased state 4 respiration, whereas state 3 respiration and the respiratory control ratio were decreased. The ADP/O ratio, the mitochondrial membrane potential, the matrix NAD(P)H levels and aconitase activity were also diminished by cis-4-decenoic acid. These data indicate that this fatty acid acts as an uncoupler of oxidative phosphorylation and as a metabolic inhibitor. cis-4-decenoic acid also provoked a marked mitochondrial swelling when either KCl or sucrose was used in the incubation medium and also induced cytochrome c release from mitochondria, suggesting a non-selective permeabilization of the inner mitochondrial membrane. SIGNIFICANCE: It is therefore presumed that impairment of mitochondrial homeostasis provoked by cis-4-decenoic acid may be involved in the brain dysfunction observed in medium-chain acyl-CoA dehydrogenase deficient patients.


Asunto(s)
Encéfalo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos Monoinsaturados/toxicidad , Mitocondrias/efectos de los fármacos , Acil-CoA Deshidrogenasa/deficiencia , Animales , Encéfalo/patología , Citocromos c/efectos de los fármacos , Citocromos c/metabolismo , Homeostasis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , NADP/efectos de los fármacos , NADP/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Wistar
17.
Tohoku J Exp Med ; 213(4): 351-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18075239

RESUMEN

Medium-chain acyl-CoA dehydrogenase deficiency (MCADD) is rare among Asian individuals, and the clinical course and biochemical findings remain unclear. We report herein a 3-year-old Japanese girl with MCADD. The diagnosis was suggested by acylcarnitine profiles and confirmed by enzyme activity and genetic analysis after clinical presentation. Our described method with high-performance liquid chromatography/tandem mass spectrometry allows quantification of levels of n-octanoylcarnitine (C8-N) and other isomers (e.g. valproylcarnitine). We examined the patient's acylcarnitine profiles in serum and urine samples during carnitine loading and 14-hr fasting tests with/without carnitine supplementation. Under hypocarnitinemia, serum level of C8-N was 0.16 micromol/l and C8-N/decanoylcarnitine (C10) ratio was 1.8, which did not correspond to the diagnostic criteria for MCADD. However, intravenous carnitine loading test (100 mg/kg/day for 3 days and 50 mg/kg/day for 1 day) led to increased serum C8-N levels and urinary excretion was obvious, strongly suggesting MCADD. In the fasting test with carnitine supplementation, marked production of acylcarnitines (C8-N > C2 >> C6 > C10) was found, compared to the fasting test without carnitine supplementation. These results indicate that carnitine supplementation may be useful for detoxification of accumulated acylcarnitines even in an asymptomatic state. Moreover, the one-point examination for serum C8-N level and/or C8-N/C10 ratio may make the diagnosis of MCADD difficult, particularly in the presence of significant hypocarnitinemia. To avoid this pitfall, attention should be given to serum levels of free carnitine, and carnitine loading may be demanded in hypocarnitinemia.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Pueblo Asiatico , Carnitina/análogos & derivados , Pruebas Diagnósticas de Rutina/métodos , Ayuno , Errores Innatos del Metabolismo Lipídico/enzimología , Glucemia , Carnitina/sangre , Carnitina/orina , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Humanos , Japón
18.
Mol Genet Metab ; 92(1-2): 109-14, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17689999

RESUMEN

A newborn female presented on the first day of life with clinical and biochemical findings consistent with multiple acyl-CoA dehydrogenase deficiency (MADD). Riboflavin supplementation corrected the biochemical abnormalities 24 h after commencing the vitamin. In vitro acylcarnitine profiling in intact fibroblasts both in normal and riboflavin depleted media showed normal oxidation of fatty acids excluding defects in electron transfer flavoprotein (ETF), or ETF ubiquinone oxidoreductase (ETF:QO), or a genetic abnormality in flavin metabolism. In addition, sequencing of the genes encoding ETF and ETF:QO in the proband did not reveal any pathogenic mutations. Determination of the maternal riboflavin status after delivery showed that the mother was riboflavin deficient. Repeat testing done two years after the infant's birth and while on a normal diet showed that the mother was persistently riboflavin deficient and showed a typical MADD profile on plasma acylcarnitine testing. A possible genetic defect in riboflavin transport of metabolism in the mother is postulated to be the cause of the transient MADD seen in the infant. Sequencing of the SLC16A12, RFK and FLAD1 genes encoding key enzymes in riboflavin transport of metabolism in the mother did not identify any pathogenic mutations. The underlying molecular basis of the mother's defect in riboflavin metabolism remains to be established.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Errores Innatos del Metabolismo Lipídico/diagnóstico , Deficiencia de Riboflavina/genética , Carnitina/análogos & derivados , Carnitina/sangre , Flavoproteínas Transportadoras de Electrones/genética , Femenino , Fibroblastos/citología , Fibroblastos/enzimología , Fibroblastos/patología , Humanos , Recién Nacido , Errores Innatos del Metabolismo Lipídico/genética , Desnutrición , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Madres , Oxidación-Reducción , Deficiencia de Riboflavina/metabolismo , Deficiencia de Riboflavina/patología , Piel/enzimología , Piel/patología , Simportadores , Vitaminas/administración & dosificación
19.
J Inherit Metab Dis ; 29(5): 631-6, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16972171

RESUMEN

Medium-chain acyl-CoA dehydrogenase deficiency (MCADD) is probably the most common inborn error of fatty acid oxidation (FAO). Routine L-carnitine supplementation in the treatment of MCADD is controversial. To establish the effects of L-carnitine supplementation during prolonged moderate-intensity exercise in MCADD, five patients and three control subjects were studied during 2 hours of moderate-intensity exercise after a 12-hour fast. Patients were studied twice, once with and once without L-carnitine supplementation (50 mg/kg per day). Blood samples were collected before, during and after exercise, and analysed for routine parameters, acylcarnitines and carnitine biosynthesis intermediates. Urine was collected before and after exercise, and analysed for acylcarnitines. All patients were able to complete the exercise test without any apparent clinical or biochemical adverse effects, even without L-carnitine supplementation. A significant rise in plasma free fatty acids and octanoylcarnitine levels during exercise was seen in all patients, indicating a substantial increase in FAO during exercise. Octanoylcarnitine levels in plasma were significantly higher in patients with L-carnitine supplementation, suggesting increased clearance of accumulating acylcarnitines. A statistically significant increase of plasma and urinary free carnitine levels, as well as of plasma gamma-butyrobetaine was seen in MCADD patients without L-carnitine supplementation. These data suggest an increase in carnitine biosynthesis. In conclusion, although L-carnitine supplementation may promote clearance of accumulating acylcarnitines during moderate-intensity exercise, no apparent beneficial effect of this supplementation on clinical and biochemical parameters was observed in MCADD patients. Our results suggest that MCADD patients are able to increase carnitine biosynthesis during exercise to compensate for carnitine losses.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/genética , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Carnitina/farmacología , Ejercicio Físico , Adolescente , Adulto , Carnitina/análogos & derivados , Carnitina/sangre , Carnitina/metabolismo , Niño , Suplementos Dietéticos , Ácidos Grasos/metabolismo , Femenino , Humanos , Masculino , Factores de Tiempo
20.
Clin Lab ; 51(5-6): 289-306, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15991803

RESUMEN

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


Asunto(s)
Encefalopatías Metabólicas/etiología , Ácidos Grasos/metabolismo , Acil-CoA Deshidrogenasa/deficiencia , Encefalopatías Metabólicas/terapia , Carnitina/biosíntesis , Carnitina/deficiencia , Dietoterapia , Humanos , Proteínas Mitocondriales/deficiencia , Oxidación-Reducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA