Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Acta Myol ; 39(1): 19-23, 2020 Mar.
Article En | MEDLINE | ID: mdl-32607475

Late-onset multiple acyl-CoA dehydrogenase deficiency (MADD) is a severe inborn error of fat metabolism. In late-onset MADD, hepatopathy in the form of steatosis is commonplace and considered a benign and stable condition that does not progress to more advanced stages of liver disease, however, progression to cirrhosis and acute liver failure (ALF) has been reported in two previous case reports. Here, we report a 22-year-old man, who suffered from late-onset MADD and died from cirrhosis and ALF. In the span of three months repeated clinical examinations, blood tests, and diagnostic imaging as well as liver biopsy revealed rapid progression of hepatopathy from steatosis to decompensated cirrhosis with portal hypertension. Routine studies for recognized etiologies found no evident cause besides MADD. This case report supports the findings of the two previous case reports and adds further evidence to the suggestion that late-onset MADD should be considered a rare cause of cirrhosis and ALF.


Fatty Liver , Hypertension, Portal , Late Onset Disorders , Liver Cirrhosis , Liver Failure, Acute , Multiple Acyl Coenzyme A Dehydrogenase Deficiency , Clinical Deterioration , Disease Progression , Electron-Transferring Flavoproteins/genetics , Fatal Outcome , Fatty Liver/diagnosis , Fatty Liver/genetics , Fatty Liver/physiopathology , Humans , Hypertension, Portal/diagnosis , Hypertension, Portal/etiology , Hypoglycemia/diagnosis , Hypoglycemia/etiology , Iron-Sulfur Proteins/genetics , Late Onset Disorders/diagnosis , Late Onset Disorders/mortality , Late Onset Disorders/physiopathology , Liver/diagnostic imaging , Liver/pathology , Liver Cirrhosis/diagnosis , Liver Cirrhosis/etiology , Liver Cirrhosis/physiopathology , Liver Failure, Acute/diagnosis , Liver Failure, Acute/etiology , Male , Medical History Taking , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/therapy , Mutation , Oxidoreductases Acting on CH-NH Group Donors/genetics , Patient Care/methods , Young Adult
2.
BMC Med Genet ; 21(1): 98, 2020 05 11.
Article En | MEDLINE | ID: mdl-32393189

BACKGROUND: Glutaric acidemia type II (GA II) or multiple acyl-CoA dehydrogenase deficiency (MADD, OMIM 231680) is an inherited autosomal recessive disease affecting fatty acid, amino acid and choline metabolism, due to mutations in one of three genes namely, electron transfer flavoprotein alpha-subunit, ETFA, electron transfer flavoprotein ß-subunit, ETFB and electron transfer flavoprotein dehydrogenase, ETFDH. Currently, few studies have reported genetic profiling of neonatal-onset GA II. This study aimed to identify the genetic mutations in a Chinese family with GA II. CASE PRESENTATION: We reported a case of GA II with purulent meningitis and septicemia and identified a novel ETFDH gene mutation in a female infant. The patient developed an episode of hypoglycemia and hypotonicity on the postnatal first day. Laboratory investigations revealed elevations of multiple acylcarnitines indicating glutaric acidemia type II in newborn screening analysis. Urinary organic acids were evaluated for the confirmation and revealed a high glutaric acid excretion. Genetic analysis revealed two mutations in the ETFDH gene (c.623_626 del / c. 1399G > C), which were considered to be the etiology for the disease. The novel mutation c.623_626 del was identified in the proband infant and her father, her mother was carriers of the mutation c.1399G > C. CONCLUSIONS: A novel variant (c.623_626 del) and a previously reported missense (c.1399G > C) in the ETFDH gene have been identified in the family. The two variants of ETFDH gene identified probably underlie the pathogenesis of Glutaric acidemia type II in this family, and also enlarge ETFDH genotype-phenotype correlations spectrum.


Electron-Transferring Flavoproteins/genetics , Genetic Predisposition to Disease , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Female , Genetic Association Studies , Genotype , Heterozygote , Humans , Infant , Infant, Newborn , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Mutation
3.
Am J Phys Med Rehabil ; 99(6): e71-e74, 2020 06.
Article En | MEDLINE | ID: mdl-31136308

Multiple acyl-CoA dehydrogenase deficiency is a rare autosomal recessive inborn error of metabolism. The late-onset multiple acyl-CoA dehydrogenase deficiency is frequently caused by mutations in ETFDH gene. Because of its clinical heterogeneity, diagnosis and treatment of late-onset multiple acyl-CoA dehydrogenase deficiency are often delayed. The authors described a previously healthy 40-yr-old Thai woman presenting with subacute severe weakness of bulbar-limb muscles and elevated serum creatine kinase. The authors emphasized the importance of needle EMG and prompt muscle histopathological evaluation, which rapidly led to the diagnosis and riboflavin therapy, resulting in a dramatic and rapid improvement before genetic study disclosed mutation in ETFDH gene.


Electromyography/methods , Lipid Metabolism, Inborn Errors/etiology , Lipid Metabolism, Inborn Errors/physiopathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/complications , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Muscular Dystrophies/etiology , Muscular Dystrophies/physiopathology , Adult , Female , Humans , Lipid Metabolism, Inborn Errors/drug therapy , Lipid Metabolism, Inborn Errors/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/drug therapy , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Muscular Dystrophies/drug therapy , Muscular Dystrophies/genetics , Riboflavin/therapeutic use
4.
J Inherit Metab Dis ; 42(5): 878-889, 2019 09.
Article En | MEDLINE | ID: mdl-31268564

Multiple acyl-CoA dehydrogenase deficiency (MADD) is an ultra-rare inborn error of mitochondrial fatty acid oxidation (FAO) and amino acid metabolism. Individual phenotypes and treatment response can vary markedly. We aimed to identify markers that predict MADD phenotypes. We performed a retrospective nationwide cohort study; then developed an MADD-disease severity scoring system (MADD-DS3) based on signs and symptoms with weighed expert opinions; and finally correlated phenotypes and MADD-DS3 scores to FAO flux (oleate and myristate oxidation rates) and acylcarnitine profiles after palmitate loading in fibroblasts. Eighteen patients, diagnosed between 1989 and 2014, were identified. The MADD-DS3 entails enumeration of eight domain scores, which are calculated by averaging the relevant symptom scores. Lifetime MADD-DS3 scores of patients in our cohort ranged from 0 to 29. FAO flux and [U-13 C]C2-, C5-, and [U-13 C]C16-acylcarnitines were identified as key variables that discriminated neonatal from later onset patients (all P < .05) and strongly correlated to MADD-DS3 scores (oleate: r = -.86; myristate: r = -.91; [U-13 C]C2-acylcarnitine: r = -.96; C5-acylcarnitine: r = .97; [U-13 C]C16-acylcarnitine: r = .98, all P < .01). Functional studies in fibroblasts were found to differentiate between neonatal and later onset MADD-patients and were correlated to MADD-DS3 scores. Our data may improve early prediction of disease severity in order to start (preventive) and follow-up treatment appropriately. This is especially relevant in view of the inclusion of MADD in population newborn screening programs.


Carnitine/analogs & derivatives , Fatty Acids/blood , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Severity of Illness Index , Carnitine/blood , Female , Humans , Infant, Newborn , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/blood , Retrospective Studies
7.
Acta Clin Belg ; 74(6): 451-455, 2019 Dec.
Article En | MEDLINE | ID: mdl-30451603

Muscle weakness is a nonspecific finding of myopathy of any etiology that include iatrogenic, toxic, endocrinological, infectious, immunologic, and metabolic disorders. Among the metabolic myopathies glutaric aciduria type II (GAII) is an autosomal recessively inherited rare disorder of fatty acid and amino acid metabolisms. The late onset form is heterogeneous in terms of symptomatology and severity and for the cases that chronic manifestations of lipid storage myopathy are the only clues for the disease, differential diagnosis can be challenging. Here we report two cases of GAII: the first one was 18-year old boy who presented with proximal muscle weakness and in another center, he was diagnosed as polymyositis and treated with immunosuppressive therapies. He admitted to our clinic with ongoing muscle weakness and symptoms that were related to the side effects of immunosuppressive therapies. The second case was also presented with muscle weakness. For both cases, muscle biopsies and urinary organic acid analyses were consistent with the diagnosis of GAII. To differentiate inflammatory myositis from non-inflammatory myopathies; rheumatic symptoms, accompanying complaints of the patient and autoantibody positivity can be helpful. To our knowledge this is the first report to underline the differential diagnosis of inflammatory myopathies from metabolic myopathies.


Acyl-CoA Dehydrogenase/deficiency , Late Onset Disorders , Lipid Metabolism, Inborn Errors , Multiple Acyl Coenzyme A Dehydrogenase Deficiency , Muscle, Skeletal/pathology , Muscular Dystrophies , Myositis/diagnosis , Adolescent , Biopsy/methods , Carnitine/administration & dosage , Diagnosis, Differential , Female , Humans , Late Onset Disorders/diagnosis , Late Onset Disorders/physiopathology , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/etiology , Lipid Metabolism, Inborn Errors/physiopathology , Male , Micronutrients/administration & dosage , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnosis , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Muscle Weakness/diagnosis , Muscle Weakness/etiology , Muscular Dystrophies/diagnosis , Muscular Dystrophies/etiology , Muscular Dystrophies/physiopathology , Riboflavin/administration & dosage , Severity of Illness Index , Urinalysis/methods , Young Adult
8.
Medicine (Baltimore) ; 97(48): e13153, 2018 Nov.
Article En | MEDLINE | ID: mdl-30508893

RATIONALE: Late-onset multiple acyl-coenzyme A dehydrogenase deficiency (MADD) mainly affects the neck extensor muscle group, which has been confirmed by novel mutations in electron-transferring-flavoprotein dehydrogenase (ETFDH). So far, a few cases have been reported with long-term follow-up. Here we report a case of late-onset MADD where the patient was followed up for 8 years during which time he underwent 2 muscle biopsies and 2 pathological examinations and his symptoms were significantly alleviated after appropriate treatments. PATIENT CONCERNS: In September 2009, a 16-year-old male patient was hospitalized due to gradually increasing difficulty in raising his head and weakness in limb muscles over a 6-month period. During the physical examination, the patient's neck extensor muscle strength was grade III-IV. His proximal limb muscle strength was grade IV, and his distal muscle strength was normal. His blood creatine kinase (CK) was 783 U/L. DIAGNOSIS: Muscle biopsy revealed a large number of vacuolar fibers, which were mainly type I fibers. These findings were consistent with the diagnosis of lipid storage myopathy (LSM). ETFDH gene test detected C.736G > A at exon 7 and C.920C > G at exon 8. INTERVENTIONS: Coenzyme Q10 treatment was administered. The first coenzyme Q10 40 mg tid was treated for three months, with the change of coenzyme Q10 20 mg tid for 6 months, followed by the change of coenzyme Q10 10 mg tid for long-term use. OUTCOMES: The patient's condition significantly improved after 3 months. At 7th year follow-up the patient's blood CK was normal, and a second muscle biopsy revealed no muscle vacuolar fibers and no increase in lipid droplets. Subsequently, the patient was withdrawn from the coenzyme Q10 treatment, and the condition of the patient remained normal. LESSONS: Muscle biopsy was the main method used to determine LSM. Treatment with riboflavin should be started when the diagnosis of LSM is definitive. Furthermore, ETFDH gene tests should be performed for further classification. Moreover, coenzyme Q10 may be another effective drug for MADD.


Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Oxidoreductases Acting on CH-NH Group Donors/genetics , Adolescent , Humans , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnosis , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/drug therapy , Muscle Strength/physiology , Muscle, Skeletal/physiopathology , Neck/physiopathology , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use
9.
Ann Neurol ; 84(5): 659-673, 2018 11.
Article En | MEDLINE | ID: mdl-30232818

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.


Electron-Transferring Flavoproteins/genetics , Flavin-Adenine Dinucleotide/metabolism , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Oxidoreductases Acting on CH-NH Group Donors/genetics , Animals , Disease Models, Animal , Female , Gene Knock-In Techniques , Homeostasis/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mutation
10.
J Neurol Sci ; 384: 121-125, 2018 Jan 15.
Article En | MEDLINE | ID: mdl-29249369

Neonatal-onset multiple acyl-CoA dehydrogenase deficiency (MADD type I) is an autosomal recessive disorder of the electron transfer flavoprotein function characterized by a severe clinical and biochemical phenotype, including congenital abnormalities with unresponsiveness to riboflavin treatment as distinguishing features. From a retrospective study, relying mainly on metabolic data, we have identified a novel mutation, c.1067G>A (p.Gly356Glu) in exon 8 of ETFDH, in three South African Caucasian MADD patients with the index patient presenting the hallmark features of type I MADD and two patients with compound heterozygous (c.1067G>A+c.1448C>T) mutations presenting with MADD type III. SDS-PAGE western blot confirmed the significant effect of this mutation on ETFDH structural instability. The identification of this novel mutation in three families originating from the South African Afrikaner population is significant to direct screening and strategies for this disease, which amongst the organic acidemias routinely screened for, is relatively frequently observed in this population group.


Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Mutation , Oxidoreductases Acting on CH-NH Group Donors/genetics , Child , Family , Fatal Outcome , Female , Humans , Infant, Newborn , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Phenotype , Retrospective Studies , South Africa , White People/genetics , Young Adult
11.
Mol Genet Metab ; 122(4): 182-188, 2017 12.
Article En | MEDLINE | ID: mdl-29122468

Vitamin B2, riboflavin is essential for cellular function, as it participates in a diversity of redox reactions central to human metabolism, through its role as precursor for the cofactors flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), which are electron carriers. The electron transfer flavoprotein (ETF) and its dehydrogenase (ETFDH), uses FAD as cofactor. The ETF and ETFDH are forming the electron transport pathway for many mitochondrial flavoprotein dehydrogenases involved in fatty acid, amino acid and choline metabolism. A variation in either ETF or ETFDH causes multiple acyl-CoA dehydrogenation deficiency (MADD), but genetic variations in the riboflavin metabolism or transportation of riboflavin can also cause MADD. The most common variations are located in the riboflavin transporter 2 (RFVT2) and 3 (RFVT3), that are highly expressed in brain and intestinal tissues, respectively. Deficiency of riboflavin transporter 1 (RFVT1), encoded by the SLC52A1 gene, highly expressed in the placenta, has only been reported once. We here report a case of transient MADD, caused by a heterozygous intronic variation, c.1134+11G>A, in the SLC52A1 gene encoding RFVT1. This variation creates a binding site for the splice inhibitory hnRNP A1 protein and causes exon 4 skipping. Riboflavin deficiency and maternal malnutrition during pregnancy might have been the determining factor in the outcome of this case.


Exons/genetics , Genetic Variation , Introns/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Receptors, G-Protein-Coupled/genetics , Riboflavin/metabolism , Case-Control Studies , DNA/blood , DNA/genetics , DNA/isolation & purification , DNA Mutational Analysis , Female , Fibroblasts/chemistry , HEK293 Cells , Heterozygote , Humans , Infant, Newborn , Membrane Transport Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/drug therapy , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Mutation , Oxidation-Reduction , Pregnancy , Riboflavin/genetics , Riboflavin/therapeutic use
12.
Lipids Health Dis ; 16(1): 185, 2017 Sep 26.
Article En | MEDLINE | ID: mdl-28950901

BACKGROUND: Glutaric aciduria type II (GA II) is an autosomal recessive disorder affecting fatty acid and amino acid metabolism. The late-onset form of GA II disorder is almost exclusively associated with mutations in the electron transfer flavoprotein dehydrogenase (ETFDH) gene. Till now, the clinical features of late-onset GA II vary widely and pose a great challenge for diagnosis. The aim of the current study is to characterize the clinical phenotypes and genetic basis of a late-onset GAII patient. METHODS: In this study, we described the clinical and biochemical manifestations of a 23-year-old female Chinese patient with late-onset GA II, and performed genomic DNA-based PCR amplifications and sequence analysis of ETFDH gene of the whole pedigree. We also used in-silicon tools to analyze the mutation and evaluated the pathogenicity of the mutation according to the criteria proposed by American College of Medical Genetics and Genomics (ACMG). RESULTS: The muscle biopsy of this patient revealed lipid storage myopathy. Blood biochemical test and urine organic acid analyses were consistent with GA II. Direct sequence analysis of the ETFDH gene (NM_004453) revealed compound heterozygous mutations: c.250G > A (p.A84T) on exon 3 and c.920C > G (p.S307C) on exon 8. Both mutations were classified as "pathogenic" according to ACMG criteria. CONCLUSIONS: In conclusion, our study described the phenotype and genotype of a late-onset GA II patient, reiterating the importance of ETFDH gene screening in these patients.


Electron-Transferring Flavoproteins/genetics , Genetic Association Studies , Heterozygote , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Mutation , Oxidoreductases Acting on CH-NH Group Donors/genetics , Age of Onset , Asian People , Base Sequence , Electron-Transferring Flavoproteins/chemistry , Exons , Female , Gene Expression , Genes, Recessive , Genotype , Humans , Iron-Sulfur Proteins/chemistry , Models, Molecular , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnosis , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/ethnology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Oxidoreductases Acting on CH-NH Group Donors/chemistry , Pedigree , Phenotype , Young Adult
13.
Neuropediatrics ; 48(3): 194-198, 2017 Jun.
Article En | MEDLINE | ID: mdl-28388738

Multiple acyl-CoA dehydrogenase deficiency (MADD) is a rare inborn error of metabolism affecting both fatty acid and amino acid oxidation. It can manifest at any age, but riboflavin-responsiveness has mainly been described in less severely affected patients. We describe an infant with severe MADD presenting with profound hypotonia and hepatomegaly. Treatment with riboflavin improved his muscle strength, liver size, and biochemical markers. A homozygous mutation of electron transfer flavoprotein dehydrogenase (ETFDH) was found. His motor skills continued to progress until a fatal infection-triggered deterioration at the age of 34 months. We show changes in brain magnetic resonance imaging over the course of the disease, with profound white matter abnormalities during the deterioration phase. Aggregates of mitochondria with abnormal cristae in muscle electron microscopy were noticed already in infancy. An unusual lactate dehydrogenase (LDH) isoenzyme pattern with LDH-1 predominance was additionally observed. This case demonstrates riboflavin-responsiveness in a severely affected infant with both muscular and extramuscular involvement and further underlines the variable nature of this disease.


Brain/diagnostic imaging , Hepatic Encephalopathy/complications , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/complications , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnostic imaging , Riboflavin/therapeutic use , Vitamin B Complex/therapeutic use , Brain/drug effects , Hepatic Encephalopathy/diagnostic imaging , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/pathology , Humans , Infant , Magnetic Resonance Imaging , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/drug therapy , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , White Matter/diagnostic imaging , White Matter/drug effects
15.
Neurol Sci ; 37(7): 1099-105, 2016 Jul.
Article En | MEDLINE | ID: mdl-27000805

Late-onset multiple acyl-CoA dehydrogenase deficiency (MADD) with electron transfer flavoprotein dehydrogenase (ETFDH) gene mutations is the most common lipid storage myopathy (LSM) in China. Its clinical features vary widely and pose a challenge for diagnosis. We presented the significant clinical heterogeneity among three Chinese late-onset MADD patients with similar ETFDH genotype by collecting clinical information, muscle histology, and genetic analysis. Three novel compound heterozygous variants of ETFDH gene were identified: c.892C > T (p.Pro298Ser), c.453delA (p.Glu152ArgfsTer15), and c.449_453delTAACA (p.Leu150Ter). Moreover, all patients carried a hotspot mutation c.250G > A (p.Ala84Thr). Western blot analysis of the patients' muscular tissue showed a significantly reduced ETFDH expression, and normal electron transfer flavoprotein A (ETFA) and electron transfer flavoprotein B (ETFB) expression. Two patients with similar genotypes (c.453delA and c.449_453delTAACA) presented a significant clinical heterogeneity. Among them, one exhibited muscle weakness and exercise intolerance as initial and major symptoms, and the other showed episodic recurrent gastrointestinal symptoms before a serious muscle weakness appeared in later life. The novel variants in ETFDH and the corresponding clinical features enrich the variant spectrum of late-onset MADD and provide a new insight into the genotype-phenotype relationship. Late-onset MADD should be included in differential diagnosis for adult myopathy along with chronic digestive disease.


Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Mutation/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Adolescent , Adult , Aged , Carnitine/analogs & derivatives , Carnitine/blood , Computational Biology , DNA Mutational Analysis , Electron-Transferring Flavoproteins/metabolism , Genotype , Humans , Iron-Sulfur Proteins/metabolism , Magnetic Resonance Imaging , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnostic imaging , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Muscle Weakness/complications , Muscle Weakness/genetics , Muscular Diseases/complications , Muscular Diseases/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism
16.
Neuromuscul Disord ; 26(2): 170-5, 2016 Feb.
Article En | MEDLINE | ID: mdl-26821934

Multiple Acyl-CoA dehydrogenase deficiency (MADD) is an autosomal recessive disorder of fatty acid oxidation. Most patients with late-onset MADD are clinically characterized by lipid storage myopathy with dramatic responsiveness to riboflavin treatment. Abnormalities of peripheral neuropathy have rarely been reported in patients with late-onset MADD. We describe six patients who presented with proximal limb weakness and loss of sensation in the distal limbs. Muscle biopsy revealed typical myopathological patterns of lipid storage myopathy and blood acylcarnitine profiles showed a combined elevation of multiple acylcarnitines supporting the diagnosis of MADD. However, nerve conduction investigations and sural nerve biopsies in these patients indicated severe axonal sensory neuropathy. Causative ETFDH gene mutations were found in all six cases. No other causative gene mutations were identified in mitochondrial DNA and genes associated with hereditary neuropathies through next-generation-sequencing panel. Late-onset patients with ETFDH mutations can present with proximal muscle weakness and distal sensory neuropathy, which might be a new phenotypic variation, but the precise underlying pathogenesis remains to be elucidated.


Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency , Oxidoreductases Acting on CH-NH Group Donors/genetics , Peripheral Nervous System Diseases , Adult , Age of Onset , Female , Humans , Male , Middle Aged , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/blood , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Mutation , Peripheral Nervous System Diseases/blood , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/physiopathology , Phenotype
17.
BMC Neurol ; 15: 114, 2015 Jul 24.
Article En | MEDLINE | ID: mdl-26205240

BACKGROUND: Late-onset multiple acyl-CoA dehydrogenase deficiency (MADD) is an autosomal recessive inherited disease of metabolic dysfunction clinically characterized by fluctuating proximal muscle weakness, excise intolerance, and dramatic riboflavin responsiveness. Dropped head syndrome can occasionally be observed in some severe patients with late-onset MADD; however, bent spine syndrome as an initial symptom had not been reported in patients with late-onset MADD. CASE PRESENTATION: A 46-year-old man lost the ability to hold his trunk upright, and had difficulty in raising his head, but he had no obvious symptoms of limb weakness. Meanwhile, he developed persistent numbness of limbs and lips around. Myopathological features and combined elevation of multiple acylcarnitines indicated that the axial myopathy might be caused by lipid storage myopathy. Cervical and lumbosacral MRI revealed a lot of abnormal signals diffusing along paravertebral muscles, while the abnormal signals almost disappeared after riboflavin treatment. Nerve conduction study indicated the patient suffering from predominantly sensory neuropathy and mildly motor neuropathy. Muscle pathology also demonstrated no typical neurogenic change, which was consistent with the electrophysiological findings. Causative mutations were found in the ETFDH gene. CONCLUSION: We report the first case of late-onset MADD with sensory neuropathy initially manifesting as bent spine syndrome and dropped head syndrome.


Lipid Metabolism, Inborn Errors/diagnosis , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnosis , Muscular Atrophy, Spinal/etiology , Muscular Dystrophies/diagnosis , Spinal Curvatures/etiology , Humans , Male , Middle Aged , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Muscle Weakness/etiology , Mutation , Riboflavin/therapeutic use
18.
J Child Neurol ; 30(1): 96-9, 2015 Jan.
Article En | MEDLINE | ID: mdl-24453145

Late-onset glutaric aciduria type II has been described recently as a rare but treatable cause of proximal myopathy in teenagers and adults. It is an autosomal recessive disease affecting fatty acid, amino acid, and choline metabolism. This is usually a result of 2 defective flavoproteins: either electron transfer flavoprotein (ETF) or electron transfer flavoprotein-ubiquinone oxidoreductase (ETF:QO). We present a 14-year-old boy with a background of autistic spectrum disorder who presented with severe muscle weakness and significant rhabdomyolysis. Before the onset of muscle weakness, he was very active but was completely bedridden at presentation. Diagnosis was established quickly by urine organic acid and plasma acylcarnitine analysis. He has shown significant improvement after starting oral riboflavin supplementation and is now fully mobile. This case highlights that late-onset glutaric aciduria type II is an important differential diagnosis to consider in teenagers presenting with proximal myopathy and rhabdomyolysis and it may not be associated with hypoglycemia.


Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnosis , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Muscular Diseases/physiopathology , Rhabdomyolysis/physiopathology , Adolescent , Carnitine/analogs & derivatives , Carnitine/blood , Humans , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/blood
19.
Muscle Nerve ; 49(3): 446-50, 2014 Mar.
Article En | MEDLINE | ID: mdl-23893693

INTRODUCTION: Glutaric aciduria type II (GAII) is a rare autosomal recessive disorder with variable clinical course. The disorder is caused by a defect in the mitochondrial electron transfer flavoprotein or the electron transfer flavoprotein dehydrogenase (ETFDH). METHODS: We performed clinical characterization, brain and whole body MRI, muscle histopathology, and genetic analysis of the ETFDH gene in a young woman. RESULTS: She presented with rhabdomyolysis and severe quadriparesis. We identified a novel homozygous missense mutation in ETFDH (c.1544G>T, p.Ser515Ile). Body fat MRI revealed a large amount of subcutaneous fat but no increase in visceral fat despite steatosis of liver and muscle. Diffusion tensor imaging (DTI) of cerebral MRI revealed reduced directionality of the white matter tracts. Histopathological findings showed lipid storage myopathy. CONCLUSIONS: In this study, we highlight diagnostic clues and body fat MRI in this rare metabolic disorder.


Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Mutation/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Adult , Anisotropy , Brain/pathology , Diffusion Tensor Imaging , Female , Humans , Magnetic Resonance Imaging , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnosis , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Whole Body Imaging
20.
PLoS Genet ; 9(6): e1003563, 2013 Jun.
Article En | MEDLINE | ID: mdl-23785301

Multiple Acyl-CoA Dehydrogenase Deficiency (MADD) is a severe mitochondrial disorder featuring multi-organ dysfunction. Mutations in either the ETFA, ETFB, and ETFDH genes can cause MADD but very little is known about disease specific mechanisms due to a paucity of animal models. We report a novel zebrafish mutant dark xavier (dxa(vu463) ) that has an inactivating mutation in the etfa gene. dxa(vu463) recapitulates numerous pathological and biochemical features seen in patients with MADD including brain, liver, and kidney disease. Similar to children with MADD, homozygote mutant dxa(vu463) zebrafish have a spectrum of phenotypes ranging from moderate to severe. Interestingly, excessive maternal feeding significantly exacerbated the phenotype. Homozygous mutant dxa(vu463) zebrafish have swollen and hyperplastic neural progenitor cells, hepatocytes and kidney tubule cells as well as elevations in triacylglycerol, cerebroside sulfate and cholesterol levels. Their mitochondria were also greatly enlarged, lacked normal cristae, and were dysfunctional. We also found increased signaling of the mechanistic target of rapamycin complex 1 (mTORC1) with enlarged cell size and proliferation. Treatment with rapamycin partially reversed these abnormalities. Our results indicate that etfa gene function is remarkably conserved in zebrafish as compared to humans with highly similar pathological, biochemical abnormalities to those reported in children with MADD. Altered mTORC1 signaling and maternal nutritional status may play critical roles in MADD disease progression and suggest novel treatment approaches that may ameliorate disease severity.


Electron-Transferring Flavoproteins/genetics , Mitochondrial Diseases/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Multiprotein Complexes/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Disease Models, Animal , Humans , Mechanistic Target of Rapamycin Complex 1 , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/physiopathology , Molecular Targeted Therapy , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/physiopathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/therapy , Multiprotein Complexes/antagonists & inhibitors , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , Zebrafish/genetics , Zebrafish/growth & development
...