Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Eur J Hum Genet ; 30(8): 976-979, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34764427

RESUMEN

Mitochondrial flavin adenine dinucleotide (FAD) transporter deficiencies are new entities recently reported to cause a neuro-myopathic phenotype. We report three patients from two unrelated families who presented primarily with hypoketotic hypoglycemia. They all had acylcarnitine profiles suggestive of multiple acyl-CoA dehydrogenase deficiency (MADD) with negative next-generation sequencing of electron-transfer flavoprotein genes (ETFA, ETFB, and ETFDH). Whole exome sequencing revealed a homozygous c.272 G > T (p.Gly91Val) variant in exon 2 of the SLC25A32 gene. The three patients shared the same variant, and they all demonstrated similar clinical and biochemical improvement with riboflavin supplementation. To date, these are the first patients to be reported with hypoketotic hypoglycemia without the neuromuscular phenotype previously reported in patients with SLC25A32 deficiency.


Asunto(s)
Hipoglucemia , Proteínas Hierro-Azufre , Proteínas de Transporte de Membrana , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Humanos , Hipoglucemia/genética , Proteínas Hierro-Azufre/genética , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Riboflavina/metabolismo
2.
Hum Mol Genet ; 31(7): 1115-1129, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-34718578

RESUMEN

To observe a long-term prognosis in late-onset multiple acyl-coenzyme-A dehydrogenation deficiency (MADD) patients and to determine whether riboflavin should be administrated in the long-term and high-dosage manner, we studied the clinical, pathological and genetic features of 110 patients with late-onset MADD in a single neuromuscular center. The plasma riboflavin levels and a long-term follow-up study were performed. We showed that fluctuating proximal muscle weakness, exercise intolerance and dramatic responsiveness to riboflavin treatment were essential clinical features for all 110 MADD patients. Among them, we identified 106 cases with ETFDH variants, 1 case with FLAD1 variants and 3 cases without causal variants. On muscle pathology, fibers with cracks, atypical ragged red fibers (aRRFs) and diffuse decrease of SDH activity were the distinctive features of these MADD patients. The plasma riboflavin levels before treatment were significantly decreased in these patients as compared to healthy controls. Among 48 MADD patients with a follow-up of 6.1 years on average, 31 patients were free of muscle weakness recurrence, while 17 patients had episodes of slight muscle weakness upon riboflavin withdrawal, but recovered after retaking a small-dose of riboflavin for a short-term. Multivariate Cox regression analysis showed vegetarian diet and masseter weakness were independent risk factors for muscle weakness recurrence. In conclusion, fibers with cracks, aRRFs and diffuse decreased SDH activity could distinguish MADD from other genotypes of lipid storage myopathy. For late-onset MADD, increased fatty acid oxidation and reduced riboflavin levels can induce episodes of muscle symptoms, which can be treated by short-term and small-dose of riboflavin therapy.


Asunto(s)
Proteínas Hierro-Azufre , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , Acilcoenzima A/genética , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Estudios de Seguimiento , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Proteínas Hierro-Azufre/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/diagnóstico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Debilidad Muscular/patología , Músculo Esquelético/metabolismo , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Estudios Retrospectivos , Riboflavina/genética , Riboflavina/uso terapéutico
3.
Genes (Basel) ; 12(11)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34828403

RESUMEN

Apple exhibits typical gametophytic self-incompatibility, in which self-S-RNase can arrest pollen tube growth, leading to failure of fertilization. To date, there have been few studies on how to resist the toxicity of self-S-RNase. In this study, pollen tube polyamines were found to respond to self-S-RNase and help pollen tubes defend against self-S-RNase. In particular, the contents of putrescine, spermidine, and spermine in the pollen tube treated with self-S-RNase were substantially lower than those treated with non-self-S-RNase. Further analysis of gene expression of key enzymes in the synthesis and degradation pathways of polyamines found that the expression of DIAMINE OXIDASE 4 (MdDAO4) as well as several polyamine oxidases such as POLYAMINE OXIDASES 3 (MdPAO3), POLYAMINE OXIDASES 4 (MdPAO4), and POLYAMINE OXIDASES 6 (MdPAO6) were significantly up-regulated under self-S-RNase treatment, resulting in the reduction of polyamines. Silencing MdPAO6 in pollen tubes alleviates the inhibitory effect of self-S-RNase on pollen tube growth. In addition, exogenous polyamines also enhance pollen tube resistance to self-S-RNase. Transcriptome sequencing data found that polyamines may communicate with S-RNase through the calcium signal pathway, thereby regulating the growth of the pollen tubes. To summarize, our results suggested that polyamines responded to the self-incompatibility reaction and could enhance pollen tube tolerance to S-RNase, thus providing a potential way to break self-incompatibility in apple.


Asunto(s)
Malus/metabolismo , Poliaminas/metabolismo , Autoincompatibilidad en las Plantas con Flores , Malus/genética , Malus/fisiología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Polen/genética , Polen/metabolismo , Polen/fisiología , Poliamino Oxidasa
4.
Crit Rev Biochem Mol Biol ; 56(4): 360-372, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33823724

RESUMEN

Electron transfer flavoprotein dehydrogenase, also called ETF-ubiquinone oxidoreductase (ETF-QO), is a protein localized in the inner membrane of mitochondria, playing a central role in the electron-transfer system. Indeed, ETF-QO mediates electron transport from flavoprotein dehydrogenases to the ubiquinone pool. ETF-QO mutations are often associated with riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency (RR-MADD, OMIM#231680), a multisystem genetic disease characterized by various clinical manifestations with different degrees of severity. In this review, we outline the clinical features correlated with ETF-QO deficiency and the benefits obtained from different treatments, such as riboflavin, L-carnitine and/or coenzyme Q10 supplementation, and a diet poor in fat and protein. Moreover, we provide a detailed summary of molecular and bioinformatic investigations, describing the mutations identified in ETFDH gene and highlighting their predicted impact on enzymatic structure and activity. In addition, we report biochemical and functional analysis, performed in HEK293 cells and patient fibroblasts and muscle cells, to show the relationship between the nature of ETFDH mutations, the variable impairment of enzyme function, and the different degrees of RR-MADD severity. Finally, we describe in detail 5 RR-MADD patients carrying different ETFDH mutations and presenting variable degrees of clinical symptom severity.


Asunto(s)
Flavoproteínas Transportadoras de Electrones , Proteínas Hierro-Azufre , Mitocondrias , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , Animales , Carnitina/genética , Carnitina/metabolismo , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Humanos , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Mitocondrias/enzimología , Mitocondrias/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/enzimología , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/genética , Ubiquinona/metabolismo
5.
Mol Med Rep ; 22(5): 4396-4402, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33000234

RESUMEN

Multiple acyl­CoA dehydrogenase deficiency (MADD) is a rare autosomal recessive disorder of fatty acid metabolism caused by defects in electron transfer flavoprotein (ETF) or electron transfer flavoprotein dehydrogenase (ETFDH). These defects are mainly classified into the neonatal and late­onset types, based on their clinical manifestations. ETFDH gene mutations are generally considered to be associated with the late­onset type. The present study reported an adult woman with late­onset MADD accompanied with biochemical and muscle biopsy findings indicating metabolic disorders. Gene sequencing analysis showed that the c.1514T>C homozygous mutation in the region of the 12th exon of the ETFDH gene, which led to the amino acid substitution p.I505T (isoleucine > threonine), resulting in defective ETFDH protein function. The results of family verification revealed that the homozygous mutation originated from her parents. The female patient was treated with a large dose of vitamin B2, L­carnitine and coenzyme Q10, and the symptoms were significantly relieved. The c.1514T>C mutation in the ETFDH gene, was considered as a novel pathogenic mutation that had not been previously reported. Therefore, it was hypothesized that this mutation was responsible for the clinical characteristics of the adult female patient. Overall, this novel mutation could expand the spectrum of the ETFDH gene mutation and provide the basis for the etiological and prenatal diagnosis of MADD.


Asunto(s)
Sustitución de Aminoácidos , 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 , Análisis de Secuencia de ADN/métodos , Adulto , Edad de Inicio , Exones , Femenino , Homocigoto , Humanos , Linaje , Polimorfismo de Nucleótido Simple
6.
PLoS Negl Trop Dis ; 14(10): e0008762, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33044977

RESUMEN

Deoxyhypusine synthase (DHS) catalyzes the first step of the post-translational modification of eukaryotic translation factor 5A (eIF5A), which is the only known protein containing the amino acid hypusine. Both proteins are essential for eukaryotic cell viability, and DHS has been suggested as a good candidate target for small molecule-based therapies against eukaryotic pathogens. In this work, we focused on the DHS enzymes from Brugia malayi and Leishmania major, the causative agents of lymphatic filariasis and cutaneous leishmaniasis, respectively. To enable B. malayi (Bm)DHS for future target-based drug discovery programs, we determined its crystal structure bound to cofactor NAD+. We also reported an in vitro biochemical assay for this enzyme that is amenable to a high-throughput screening format. The L. major genome encodes two DHS paralogs, and attempts to produce them recombinantly in bacterial cells were not successful. Nevertheless, we showed that ectopic expression of both LmDHS paralogs can rescue yeast cells lacking the endogenous DHS-encoding gene (dys1). Thus, functionally complemented dys1Δ yeast mutants can be used to screen for new inhibitors of the L. major enzyme. We used the known human DHS inhibitor GC7 to validate both in vitro and yeast-based DHS assays. Our results show that BmDHS is a homotetrameric enzyme that shares many features with its human homologue, whereas LmDHS paralogs are likely to form a heterotetrameric complex and have a distinct regulatory mechanism. We expect our work to facilitate the identification and development of new DHS inhibitors that can be used to validate these enzymes as vulnerable targets for therapeutic interventions against B. malayi and L. major infections.


Asunto(s)
Antihelmínticos/farmacología , Antiprotozoarios/farmacología , Brugia Malayi/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas del Helminto/antagonistas & inhibidores , Leishmania major/efectos de los fármacos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Antihelmínticos/química , Antiprotozoarios/química , Brugia Malayi/enzimología , Brugia Malayi/genética , Brugia Malayi/crecimiento & desarrollo , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Proteínas del Helminto/química , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Ensayos Analíticos de Alto Rendimiento , Leishmania major/enzimología , Leishmania major/genética , Leishmania major/crecimiento & desarrollo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/química , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Alineación de Secuencia
7.
Medicine (Baltimore) ; 99(37): e21944, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32925727

RESUMEN

RATIONALE: Multiple acyl-CoA dehydrogenase deficiency (MADD) is a rare inborn error of metabolism affecting fatty acid, amino acid, and choline metabolism. The clinical manifestation of MADD is heterogeneous, from severe neonatal forms to mild late-onset forms. PATIENT CONCERNS: Here, we report a patient who presented with severe hypoglycemia and exercise intolerance suggestive of MADD. Serum tandem mass spectrometry analysis indicated elevated levels of various acyl carnitines at 25 days of age. Exome sequencing of the proband revealed compound heterozygous mutations, c. 413T>G (p.Leu138Arg) and c.1667C > G (p.Pro556Arg), in the ETFDH gene as the probable causative mutations. DIAGNOSES: Based on the patient's clinical presentation and test results, the patient was diagnosed with MADD. INTERVENTIONS: A high-calorie and reduced-fat diet was given together with oral supplements of L-carnitine (150 mg/day). OUTCOMES: He passed away at the age of 4 months because of severe respiratory distress accompanied by muscle weakness. LESSONS: He passed away at the age of 4 months because of severe respiratory distress accompanied by muscle weakness. Clinicians should consider MADD in the differential diagnosis when patients present with muscle weakness and biochemical abnormalities. Gene testing plays a critical role in confirming the diagnosis of MADD and may not only prevent the need for invasive testing but also allow for timely initiation of treatment.


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 , Humanos , Recién Nacido , Masculino , Mutación
8.
Med Sci Monit ; 25: 9103-9111, 2019 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-31785094

RESUMEN

BACKGROUND In this study, we investigated the clinical and pathological features of patients with lipid storage myopathy (LSM) complicated with hyperuricemia, to improve clinicians' understanding of metabolic multi-muscular disorder with metabolic disorders, and to reduce the risk of missed diagnosis of LSM. MATERIAL AND METHODS From January 2005 to December 2017, 8 patients underwent muscle biopsy and diagnosed by muscle pathology and genetic testing in our hospital. All 8 patients were in compliance with LSM diagnosis. We collected data on the patient's clinical performance, adjuvant examination, treatment, and outcomes to provide a comprehensive report and description of LSM patients with hyperuricemia. RESULTS All patients were diagnosed as having ETFDH gene mutations. The main clinical manifestations of patients were chronic limb and trunk weakness, limb numbness, and muscle pain. The serum creatine kinase (CK) values in all patients were higher than normal values. Electromyography showed 3 cases of simple myogenic damage and 3 cases of neurogenic injury. Hematuria metabolic screening showed that 2 patients had elevated glutaric aciduria, and 1 patient had elevated fatty acyl carnitine in the blood. All patients were given riboflavin treatment, and the clinical symptoms were significantly improved, and 3 patients returned to normal uric acid levels after treatment. Pathological staining showed an abnormal deposition of lipid droplets in muscle fibers. CONCLUSIONS If an adolescent hyperuricemia patient has abnormal limb weakness, exercise intolerance, and elevated serum CK values, clinicians need to be highly alert to the possibility of LSM. Early diagnosis and treatment of LSM should improve the clinical symptoms and quality of life and reduce complications.


Asunto(s)
Hiperuricemia/fisiopatología , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/fisiopatología , Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Adolescente , Adulto , Carnitina/análogos & derivados , Carnitina/metabolismo , Niño , China , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Femenino , Humanos , Hiperuricemia/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Errores Innatos del Metabolismo Lipídico/metabolismo , Masculino , Debilidad Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/genética , Distrofias Musculares/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Calidad de Vida , Riboflavina/metabolismo , Adulto Joven
9.
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
10.
J Sci Food Agric ; 99(8): 4082-4093, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30761554

RESUMEN

BACKGROUND: Selenium (Se)-induced phytotoxicity has been linked to oxidative injury triggered by the accumulation of reactive oxygen species (ROS) due to the disturbance of anti-oxidative systems. However, the way Se stress induces hydrogen peroxide (H2 O2 ) production in plants is a long-standing question. Here we identified the role of polyamine oxidase (PAO) in H2 O2 production in the root of Brassica rapa upon Se stress. RESULTS: Studying Se-induced growth inhibition, H2 O2 accumulation, and oxidative injury in the root of Brassica rapa, we found that excessive Se exposure resulted in a remarkable increase in PAO activity. Inhibition of PAO activity led to decreased H2 O2 content and alleviated oxidative injury in the Se-treated root. These results indicated that Se stress induced PAO-dependent H2 O2 production. A total of six BrPAO family members were discovered in the genome of B. rapa by in silico analysis. Se stress pronouncedly upregulated the expression of most BrPAOs and further transient expression analysis proved that it could lead to H2 O2 production. CONCLUSION: These results suggest that Se stress upregulates the expression of a set of BrPAOs which further enhances PAO activity, contributing to H2 O2 generation in roots. © 2019 Society of Chemical Industry.


Asunto(s)
Brassica rapa/genética , Peróxido de Hidrógeno/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Proteínas de Plantas/metabolismo , Selenio/metabolismo , Brassica rapa/enzimología , Brassica rapa/crecimiento & desarrollo , Brassica rapa/metabolismo , Regulación de la Expresión Génica de las Plantas , Estrés Oxidativo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Proteínas de Plantas/genética , Especies Reactivas de Oxígeno/metabolismo , Poliamino Oxidasa
11.
BMC Neurol ; 18(1): 219, 2018 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-30587156

RESUMEN

BACKGROUND: Multiple acyl-CoA dehydrogenase deficiency (MADD) showed great clinical heterogeneity and poses a challenge to diagnosis. Guillain-Barré syndrome (GBS) is an acute-onset autoimmune-mediated peripheral neuropathy. However, no patients of acute-onset MADD mimicking the GBS phenotype are reported previously. CASE PRESENTATION: Two patients displayed acute-onset limb weakness, areflexia, and length-dependent sensory disturbances, which clinically indicate the diagnosis of GBS, but electrophysiological and cerebrospinal fluid results threw doubtful points to the initial diagnosis. The muscle biopsy showed lipid storage disorder; and compound heterozygous mutations in the electron transfer flavoprotein dehydrogenase (ETFDH) gene were found in the two patients through targeted next generation sequencing, which provided the definite diagnostic evidences of late-onset MADD. Muscle weakness was quickly improved by riboflavin supplementation, but sensory disturbances required a long-term treatment. DISCUSSION: The present two cases have demonstrated that MADD can mimic GBS. Taking into consideration the significant differences of therapeutic regimen and prognosis, MADD should be included in the differential diagnosis of GBS.


Asunto(s)
Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/diagnóstico , Biopsia , Diagnóstico Diferencial , Flavoproteínas Transportadoras de Electrones/genética , Síndrome de Guillain-Barré/diagnóstico , Humanos , Proteínas Hierro-Azufre/genética , Masculino , Persona de Mediana Edad , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Debilidad Muscular/etiología , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Fenotipo , Adulto Joven
12.
Lipids Health Dis ; 17(1): 254, 2018 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-30424791

RESUMEN

BACKGROUND: Deficiency of electron transfer flavoprotein dehydrogenase (ETFDH) is associated with multiple acyl-CoA dehydrogenase deficiency (MADD). This disorder is an autosomal recessive lipid storage myopathy (LSM) that exhibits a wide range of clinical features, including myopathy, weakness and multisystem dysfunctions. Many patients with late onset of MADD improve when treated with riboflavin and are also referred to as RR-MADD (riboflavin-responsive multiple Acyl-CoA dehydrogenase disorder). METHODS: In this study, we report the clinical and genetic characterization of a novel RR-MADD patient. Biochemical data were obtained from analysis of muscle and plasma samples. DNA and RNA were extracted from peripheral blood, and sequence analysis and expression study of ETFDH gene were performed. Finally, the impact of mutations on ETFDH folding was evaluated using bioinformatic tools. RESULTS: Patient initially presented with vomiting, muscle weakness, and acidosis. Muscle biopsy revealed typical myopathological patterns of lipid storage myopathy and blood acylcarnitine profiles showed a combined elevation of long and medium chain acylcarnitines, supporting the diagnosis of RR-MADD. Molecular analysis of ETFDH gene revealed two heterozygous mutations, a novel splice variation in intron 10, c.1285 + 1G > A, and the previously reported c.560C > T missense mutation. RT-PCR analysis showed an alteration of ETFDH RNA splicing which in turn should lead to the production of a truncated protein. The in silico prediction analysis of ETFDH tridimensional structure demonstrated that the missense mutation resulted in instability and loss of protein activation, while the splice site variation induced a dramatic conformational change of the truncated protein. After MCT diet supplemented with carnitine and riboflavin, the patient showed significant biochemical and clinical improvement, in spite of severe molecular defect. CONCLUSION: This case report extends the spectrum of ETFDH mutations in MADD, providing further evidence that patients presenting at least one missense mutation in the FAD-binding domain may respond to either carnitine or riboflavin treatment, due to the recovery of some enzymatic activity.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/genética , Proteínas Hierro-Azufre/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Carnitina/uso terapéutico , Simulación por Computador , Análisis Mutacional de ADN , Quimioterapia Combinada , Flavoproteínas Transportadoras de Electrones/metabolismo , Femenino , Humanos , Proteínas Hierro-Azufre/metabolismo , Persona de Mediana Edad , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/enzimología , Músculo Esquelético/enzimología , Mutación Missense , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Conformación Proteica , Riboflavina/uso terapéutico
13.
Ann Neurol ; 84(5): 659-673, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30232818

RESUMEN

OBJECTIVE: Riboflavin-responsive multiple acyl-coenzyme A dehydrogenation deficiency (RR-MADD) is an inherited fatty acid metabolism disorder mainly caused by genetic defects in electron transfer flavoprotein-ubiquinone oxidoreductase (ETF:QO). The variant ETF:QO protein folding deficiency, which can be corrected by therapeutic dosage of riboflavin supplement, has been identified in HEK-293 cells and is believed to be the molecular mechanism of this disease. To verify this hypothesis in vivo, we generated Etfdh (h)A84T knockin (KI) mice. METHODS: Tissues from these mice as well as muscle biopsies and fibroblasts from 7 RR-MADD patients were used to examine the flavin adenine dinucleotide (FAD) concentration and ETF:QO protein amount. RESULTS: All of the homozygous KI mice (Etfdh (h)A84T/(h)A84T , KI/KI) were initially normal. After being given a high-fat and vitamin B2 -deficient (HF-B2 D) diet for 5 weeks, they developed weight loss, movement ability defects, lipid storage in muscle and liver, and elevated serum acyl-carnitine levels, which are clinically and biochemically similar to RR-MADD patients. Both ETF:QO protein and FAD concentrations were significantly decreased in tissues of HF-B2 D-KI/KI mice and in cultured fibroblasts from RR-MADD patients. After riboflavin treatment, ETF:QO protein increased in proportion to elevated FAD concentrations, but not related to mRNA levels. These results were further confirmed in cultured fibroblasts from RR-MADD patients. INTERPRETATION: For the first time, we successfully developed a RR-MADD mice model and confirmed that FAD homeostasis disturbances played a crucial role on the pathomechanism of RR-MADD in this mouse model and culture cells from patients. Supplementation of riboflavin may stabilize variant ETF:QO protein by rebuilding FAD homeostasis. Ann Neurol 2018;84:667-681.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/genética , Flavina-Adenina Dinucleótido/metabolismo , Proteínas Hierro-Azufre/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/metabolismo , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/fisiopatología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Homeostasis/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación
14.
PLoS One ; 13(8): e0202677, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30138353

RESUMEN

Curcumin, a natural polyphenol that contributes to the flavor and yellow pigment of the spice turmeric, is known for its antioxidant, anti-inflammatory, and anticarcinogenic properties. Capable of affecting the initiation, promotion, and progression of carcinogenesis through multiple mechanisms, curcumin has potential utility for both chemoprevention and chemotherapy. Previous studies demonstrated that curcumin can inhibit ornithine decarboxylase (ODC) activity in human leukemia and breast cancer cells, and pretreatment with dietary curcumin blocks carcinogen-induced ODC activity in rodent models of skin, colon, and renal cancer. The current study investigated the regulation of polyamine metabolism in human gastric and colon carcinoma cell lines in response to curcumin. Curcumin treatment significantly induced spermine oxidase (SMOX) mRNA and activity, which results in the generation of hydrogen peroxide, a source of ROS. Simultaneously, curcumin down regulated spermidine/spermine N1-acetyltransferase (SSAT) activity and the biosynthetic enzymes ODC and S-adenosylmethionine decarboxylase (SAMDC), thereby diminishing intracellular polyamine pools. Combination treatments using curcumin with the ODC inhibitor 2-difluoromethylornithine (DFMO), an agent currently in clinical chemoprevention trials, significantly enhanced inhibition of ODC activity and decreased growth of GI cancer cell lines beyond that observed with either agent alone. Similarly, combining curcumin with the polyamine analogue bis(ethyl)norspermine enhanced growth inhibition that was accompanied by enhanced accumulation of the analogue and decreased intracellular polyamine levels beyond those observed with either agent alone. Importantly, cotreatment with curcumin permitted the lowering of the effective dose of ODC inhibitor or polyamine analogue. These studies provide insight into the polyamine-related mechanisms involved in the cancer cell response to curcumin and its potential as a chemopreventive or chemotherapeutic agent in the GI tract.


Asunto(s)
Antineoplásicos/farmacología , Vías Biosintéticas/efectos de los fármacos , Curcumina/farmacología , Neoplasias Gastrointestinales/metabolismo , Poliaminas/metabolismo , Espermina/análogos & derivados , Acetiltransferasas/metabolismo , Adenosilmetionina Descarboxilasa/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Eflornitina/farmacología , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ornitina Descarboxilasa/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Espermina/farmacología , Poliamino Oxidasa
15.
Free Radic Res ; 52(11-12): 1445-1455, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30003820

RESUMEN

Multiple acyl-CoA dehydrogenase deficiency (MADD), an autosomal recessive metabolic disorder of fatty acid metabolism, is mostly caused by mutations in the ETFA, ETFB or ETFDH genes that result in dysfunctions in electron transfer flavoprotein (ETF) or electron transfer flavoprotein-ubiquinone dehydrogenase (ETFDH). In ß-oxidation, fatty acids are processed to generate acyl-CoA, which is oxidised by flavin adenine dinucleotide and transfers an electron to ETF and, through ETFDH, to mitochondrial respiratory complex III to trigger ATP synthesis. Coenzyme Q10 (CoQ10) is believed to be a potential treatment that produces symptom relief in some MADD patients. CoQ10 acts as a key regulator linking ETFDH and mitochondrial respiratory complex III. Our aim is to investigate the effectiveness of CoQ10 in serving in the ETF/ETFDH system to improve mitochondrial function and to reduce lipotoxicity. In this study, we used lymphoblastoid cells with an ETFDH mutation from MADD patients. ETFDH dysfunction caused insufficient ß-oxidation, leading to increasing lipid droplet and lipid peroxide accumulation. In contrast, supplementation with CoQ10 significantly recovered mitochondrial function and concurrently decreased the generation of reactive oxygen species and lipid peroxides, inhibited the accumulation of lipid droplets and the formation of the NOD-like receptor family pyrin domain-containing three (NLRP3) inflammasome, and reduced interleukin-1ß release and cell death. These results clarify the causal role of CoQ10 in coupling the electron transport chain with ß-oxidation, which may promote the development of CoQ10-directed therapies for MADD patients.


Asunto(s)
Ácidos Grasos/metabolismo , Inflamasomas/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Fosforilación Oxidativa/efectos de los fármacos , Ubiquinona/análogos & derivados , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Flavoproteínas Transportadoras de Electrones/deficiencia , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Humanos , Inflamasomas/metabolismo , Proteínas Hierro-Azufre/deficiencia , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Mitocondrias/metabolismo , Mutación , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Oxidación-Reducción/efectos de los fármacos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/deficiencia , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Ubiquinona/administración & dosificación , Ubiquinona/metabolismo , Ubiquinona/farmacología
17.
Zhongguo Dang Dai Er Ke Za Zhi ; 19(9): 975-978, 2017 Sep.
Artículo en Chino | MEDLINE | ID: mdl-28899466

RESUMEN

OBJECTIVE: To investigate the clinical and genetic features of two families with late-onset glutaric aciduria type II caused by ETFDH mutations. METHODS: Target gene sequence capture and next generation sequencing were used for sequencing of suspected patients and their family members. The patients' clinical features were retrospectively analyzed and literature review was performed. RESULTS: The probands of the two families had a clinical onset at the ages of 10 years and 5.5 years respectively, with the clinical manifestations of muscle weakness and muscle pain. Laboratory examinations revealed significant increases in the serum levels of creatine kinase, creatine kinase-MB, and lactate dehydrogenase. Tandem mass spectrometry showed increases in various types of acylcarnitines. The analysis of urine organic acids showed an increase in glutaric acid. Electromyography showed myogenic damage in both patients. Gene detection showed two novel mutations in the ETFDH gene (c.1331T>C from the mother and c.824C>T from the father) in patient 1, and the patient's younger brother carried the c.1331T>C mutation but had a normal phenotype. In patient 2, there was a novel mutation (c.177insT from the father) and a known mutation (c.1474T>C from the mother) in the ETFDH gene. Several family members carried such mutations. Both patients were diagnosed with glutaric aciduria type II. Their symptoms were improved after high-dose vitamin B2 treatment. CONCLUSIONS: For patients with unexplained muscle weakness and pain, serum creatine kinase, acylcarnitines, and urinary organic acids should be measured, and the possibility of glutaric aciduria type II should be considered. Genetic detection is helpful to make a confirmed diagnosis.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/genética , Proteínas Hierro-Azufre/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Niño , Biología Computacional , Femenino , Humanos , Masculino , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico
18.
Muscle Nerve ; 56(3): 479-485, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27935074

RESUMEN

INTRODUCTION: c.250G>A (p.Ala84Thr) in ETFDH is the most common mutation that causes later-onset multiple acyl-coenzyme A dehydrogenase deficiency (MADD) in the southern Chinese population. No functional study has targeted this mutation. METHODS: Using cells expressing ETFDH-wild-type (WT) or ETFDH-mutant (p.Ala84Thr), reactive oxygen species (ROS) production and neurite length were analyzed, followed by pathomechanism exploration and drug screening. RESULTS: Increased ROS production and marked neurite shortening were observed in the cells expressing the ETFDH-mutant, compared with WT. Further studies demonstrated that suberic acid, an accumulated intermediate metabolite in MADD, could significantly impair neurite outgrowth of NSC34 cells, but neurite shortening could be restored by supplementation with carnitine, riboflavin, or Coenzyme Q10. CONCLUSIONS: Neurite shortening caused by the c.250G>A mutation in ETFDH suggests that neural defects could be underdiagnosed in human patients with MADD. This impairment might be treatable with mitochondrial cofactor supplementation. Muscle Nerve 56: 479-485, 2017.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/biosíntesis , Flavoproteínas Transportadoras de Electrones/genética , Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación/fisiología , Proyección Neuronal/fisiología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/biosíntesis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Línea Celular , Humanos , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/metabolismo , Neuritas/metabolismo , Proyección Neuronal/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/farmacología
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 33(2): 191-4, 2016 Apr.
Artículo en Chino | MEDLINE | ID: mdl-27060313

RESUMEN

OBJECTIVE: To analyze the clinical features and gene mutations in an adolescent patient affected with late-onset multiple aeyl-CoA dehydrogenase deficiency (MADD) with severe fatty liver. METHODS: Potential mutations of the ETFDH gene were detected with polymerase chain reaction (PCR) and DNA sequencing. RESULTS: The 13-year-and-10-month girl has presented with weakness without any other special manifestation. Laboratory tests demonstrated an elevation of myocardial enzyme levels, total cholesterol, lactic acid and abnormal serum free fatty acids. H magnetic resonance spectroscopy revealed severe fatty liver. An increase in multiple plasma acyl-carnitines was detected by gas chromatography/mass spectrometry and isobutyrylglycine in urine by screening with tandem mass spectrometry. Genetic analysis demonstrated 2 heterozygous missense mutations c.250G>A (p.Ala84Thr) and c.353G>T (p.Cys118Phe) in the ETFDH gene. The diagnosis of MADD was confirmed. The patient was given large dose of vitamin B2, which resulted in rapid clinical and biochemical improvement. CONCLUSION: A common mutation c.250G>A and a novel mutation c.353G>T in the ETFDH gene were identified in the patient. The pathogenic role of c.353G>T (p.Cys118Phe) deserves further study. Early diagnosis of MADD and appropriate therapy is crucial for the prognosis.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/genética , Hígado Graso/genética , Proteínas Hierro-Azufre/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Adolescente , Adulto , Secuencia de Bases , Ácidos Grasos no Esterificados/sangre , Hígado Graso/sangre , Hígado Graso/enzimología , Femenino , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/sangre , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/enzimología , Mutación , Linaje
20.
Neurol Sci ; 36(6): 853-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25827849

RESUMEN

We report a case with late onset riboflavin-responsive multiple acyl-CoA dehydrogenation deficiency (MADD) characterized by decreased acyl-carnitine profile in serum which is consistent with primary systemic carnitine deficiency (CDSP) while just the contrary to a typical MADD. This patient complained with muscle weakness, muscle pain and intermittent vomiting, and was diagnosed as polymyositis, received prednisone therapy before consulted with us. Muscle biopsy revealed mild lipid storage. The findings of serum acyl-carnitines were consistent with CDSP manifesting as decreased free and total carnitines in serum. But oral L-carnitine supplementation was not very effective to this patient and mutation analysis of the SLC22A5 gene for CDSP was normal. Later, another acyl-carnitine analysis revealed a typical MADD profile in serum, which was characterized by increased multiple acyl-carnitines. Compound heterozygous mutations were identified in electron transferring-flavoprotein dehydrogenase (ETFDH) gene which confirmed the diagnosis of MADD. After administration of riboflavin, he improved dramatically, both clinically and biochemically. Thus, late onset riboflavin-responsive MADD should be included in the differential diagnosis for adult carnitine deficiency.


Asunto(s)
Acilcoenzima A/sangre , Carnitina/análogos & derivados , Flavoproteínas Transportadoras de Electrones/genética , Proteínas Hierro-Azufre/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Mutación/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Carnitina/uso terapéutico , Análisis Mutacional de ADN/métodos , Humanos , Masculino , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/diagnóstico , Músculo Esquelético/patología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA