Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Genes (Basel) ; 12(5)2021 05 08.
Article in English | MEDLINE | ID: mdl-34066864

ABSTRACT

Glutaric aciduria type II (GA-II) is a rare autosomal recessive disease caused by defects in electron transfer flavoprotein (ETF), ultimately causing insufficiencies in multiple acyl-CoA dehydrogenase (MAD). 3-phosphoglycerate dehydrogenase (3-PHGDH) deficiency, is another rare autosomal disorder that appears due to a defect in the synthesis of L-serine amino acid. Several mutations of ETFDH and PHGDH genes have been associated with different forms of GA-II and serine deficiency, respectively. In this study, we report a unique case of GA-II with serine deficiency using biochemical, genetic, and in silico approaches. The proband of Syrian descent had positive newborn screening (NBS) for GA-II. At two years of age, the patient presented with developmental regression, ataxia, and intractable seizures. Results of amino acid profiling demonstrated extremely low levels of serine. Confirmatory tests for GA-II and whole exome sequencing (WES) were performed to determine the etiology of intractable seizure. Sequencing results indicated a previously reported homozygous missense mutation, c.679 C>A (p.Pro227Thr) in the ETFDH gene and a novel missense homozygous mutation c.1219 T>C (p.Ser407Pro) in the PHGDH gene. In silico tools predicted these mutations as deleterious. Here, the clinical and biochemical investigations indicate that ETFDH:p.Pro227Thr and PHGDH:p.Ser407Pro variants likely underlie the pathogenesis of GA-II and serine deficiency, respectively. This study indicates that two rare autosomal recessive disorders should be considered in consanguineous families, more specifically in those with atypical presentation.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/genetics , Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Microcephaly/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Phosphoglycerate Dehydrogenase/deficiency , Phosphoglycerate Dehydrogenase/genetics , Psychomotor Disorders/genetics , Seizures/genetics , Serine/deficiency , Carbohydrate Metabolism, Inborn Errors/blood , Carbohydrate Metabolism, Inborn Errors/pathology , Child, Preschool , Female , Humans , Microcephaly/blood , Microcephaly/pathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Mutation, Missense , Phosphoglycerate Dehydrogenase/blood , Psychomotor Disorders/blood , Psychomotor Disorders/pathology , Seizures/blood , Seizures/pathology , Serine/blood
2.
Am J Med Genet A ; 182(10): 2426-2431, 2020 10.
Article in English | MEDLINE | ID: mdl-32804429

ABSTRACT

Glutaric acidemia type 2 (GA2), also called multiple acyl-CoA dehydrogenase deficiency, is an autosomal recessive disorder of fatty acid, amino acid, and choline metabolism resulting in excretion of multiple organic acids and glycine conjugates as well as elevation of various plasma acylcarnitine species (C4-C18). It is caused by mutations in the ETFA, ETFB, or ETFDH genes which are involved in the transfer of electrons from 11 flavin-containing dehydrogenases to Coenzyme Q10 (CoQ10 ) of the mitochondrial electron transport chain (ETC). We report a patient who was originally reported as the first case with primary myopathic CoQ10 deficiency when he presented at 11.5 years with exercise intolerance and myopathy that improved after treatment with ubiquinone and carnitine. At age 23, his symptoms relapsed despite increasing doses of ubiquinone and he was shown to have biallelic mutations in the ETFDH gene. Treatment with riboflavin was started and ubiquinone was changed to ubiquinol. After 4 months, the patient recovered his muscle strength with normalization of laboratory exams and exercise tolerance. Functional studies on fibroblasts revealed decreased levels of ETFDH as well as of very long-chain acyl-CoA dehydrogenase and trifunctional protein α. In addition, the mitochondrial mass was decreased, with increased formation of reactive oxygen species and oxygen consumption rate, but with a decreased spared respiratory capacity, and decreased adenosine triphosphate level. These findings of widespread dysfunction of fatty acid oxidation and ETC enzymes support the impairment of a larger mitochondrial ETC supercomplex in our patient.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/genetics , Ataxia/genetics , Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Mitochondrial Diseases/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Muscle Weakness/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Ubiquinone/deficiency , Adult , Age of Onset , Ataxia/diagnosis , Ataxia/pathology , Child , Energy Metabolism/genetics , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/pathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Muscle Weakness/diagnosis , Muscle Weakness/pathology , Ubiquinone/analogs & derivatives , Ubiquinone/genetics , Young Adult
3.
Int J Mol Sci ; 21(15)2020 Jul 26.
Article in English | MEDLINE | ID: mdl-32722651

ABSTRACT

Inborn errors of Riboflavin (Rf) transport and metabolism have been recently related to severe human neuromuscular disorders, as resulting in profound alteration of human flavoproteome and, therefore, of cellular bioenergetics. This explains why the interest in studying the "flavin world", a topic which has not been intensively investigated before, has increased much over the last few years. This also prompts basic questions concerning how Rf transporters and FAD (flavin adenine dinucleotide) -forming enzymes work in humans, and how they can create a coordinated network ensuring the maintenance of intracellular flavoproteome. The concept of a coordinated cellular "flavin network", introduced long ago studying humans suffering for Multiple Acyl-CoA Dehydrogenase Deficiency (MADD), has been, later on, addressed in model organisms and more recently in cell models. In the frame of the underlying relevance of a correct supply of Rf in humans and of a better understanding of the molecular rationale of Rf therapy in patients, this review wants to deal with theories and existing experimental models in the aim to potentiate possible therapeutic interventions in Rf-related neuromuscular diseases.


Subject(s)
Flavoproteins/metabolism , Models, Biological , Multiple Acyl Coenzyme A Dehydrogenase Deficiency , Muscle Proteins/metabolism , Neuromuscular Diseases/metabolism , Riboflavin Deficiency/metabolism , Flavoproteins/genetics , Humans , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Muscle Proteins/genetics , Neuromuscular Diseases/genetics , Neuromuscular Diseases/pathology , Riboflavin/genetics , Riboflavin/metabolism , Riboflavin Deficiency/genetics
4.
Neuropathology ; 40(6): 531-539, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32608139

ABSTRACT

Multiple Acyl-CoA dehydrogenase deficiency (MADD), one of the most common lipid storage myopathies (LSMs), is a heterogeneous inherited muscular disorder that is pathologically characterized by numerous lipid droplets in muscle fibers due to lipid metabolism disturbance. MADD exhibits a wide range of clinical features, including skeletal muscle weakness and multisystem dysfunctions. However, MADD, as well as other types of LSM, associated with peripheral neuropathy has rarely been reported during the past four decades. Here, we present four Chinese patients affected by MADD with peripheral neuropathy in our neuromuscular center. Clinically, these four patients showed skeletal muscle weakness and prominent paresthesia. Muscle biopsy detected characteristic myopathological patterns of LSM, such as obvious lipid droplets in muscle fibers. Sural nerve biopsy revealed a severe reduction in number of myelinated nerve fibers, which is a typical neuropathological pattern of peripheral neuropathy. Causative ETFDH mutations were found in all four cases. The skeletal muscle weakness was rapidly improved after some treatments while paresthesia showed unsatisfactory improvement. The features of previously reported patients of this specific type are also summarized in this paper. We propose that MADD with peripheral neuropathy may be a new phenotypic subtype because the pathology and reaction to riboflavin treatment are different from those of traditional MADD, although further research on the precise pathogenesis and mechanisms is needed.


Subject(s)
Multiple Acyl Coenzyme A Dehydrogenase Deficiency/complications , Peripheral Nervous System Diseases/etiology , Adult , Electron-Transferring Flavoproteins/genetics , Female , Humans , Iron-Sulfur Proteins/genetics , Male , Middle Aged , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Mutation , Oxidoreductases Acting on CH-NH Group Donors/genetics , Riboflavin/therapeutic use
5.
Int J Neurosci ; 130(12): 1192-1198, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32064983

ABSTRACT

Background: Recessive mutations in ETFDH gene have been associated with Multiple Acyl-CoA dehydrogenase deficiency (MADD). The late-onset MADD is often muscle involved, presenting with lipid storage myopathy (LSM). The symptoms of LSM were heterogeneous and definite diagnosis of this disease depends on the pathology and gene test.Methods: Neurological examination, muscle biopsy, and MRI examinations were performed in a patient with a novel missense ETFDH mutation.Results: We describe a patient with lipid storage myopathy complicated with skin damage. In addition, the next generation revealed a novel missense mutation (c.970G > T, p.Val324Leu) in exon 8, which was predicted to be a disease-causing mutation by Mutation-taster, and destroy the function of the protein by Sift.Conclusion: These findings expand the known mutational spectrum of ETFDH and phenotype of MADD.


Subject(s)
Electron-Transferring Flavoproteins/genetics , Lipid Metabolism, Inborn Errors , Multiple Acyl Coenzyme A Dehydrogenase Deficiency , Muscular Dystrophies , Riboflavin/pharmacology , Skin/pathology , Vitamin B Complex/pharmacology , Humans , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/drug therapy , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnosis , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/drug therapy , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Muscular Dystrophies/diagnosis , Muscular Dystrophies/drug therapy , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Mutation, Missense , Riboflavin/administration & dosage , Vitamin B Complex/administration & dosage
6.
Curr Mol Med ; 19(7): 487-493, 2019.
Article in English | MEDLINE | ID: mdl-31418342

ABSTRACT

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.


Subject(s)
Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Acyl-CoA Dehydrogenase/deficiency , Acyl-CoA Dehydrogenase/genetics , Alleles , Female , Genetic Predisposition to Disease , Genotype , Humans , Infant, Newborn , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Mutation, Missense/genetics , Neonatal Screening , Portugal/epidemiology , Pregnancy , Prognosis , Riboflavin/genetics , Riboflavin/metabolism
7.
Chin Med J (Engl) ; 132(3): 275-284, 2019 Feb 05.
Article in English | MEDLINE | ID: mdl-30681493

ABSTRACT

BACKGROUND: Late-onset multiple acyl-coA dehydrogenase deficiency (MADD) is an autosomal recessive inherited metabolic disorder. It is still unclear about the muscle magnetic resonance image (MRI) pattern of the distal lower limb pre- and post-treatment in patients with late-onset MADD. This study described the clinical and genetic findings in a cohort of patients with late-onset MADD, and aimed to characterize the MRI pattern of the lower limbs. METHODS: Clinical data were retrospectively collected from clinic centers of Peking University People's Hospital between February 2014 and February 2018. Muscle biopsy, blood acylcarnitines, and urine organic acids profiles, and genetic analysis were conducted to establish the diagnosis of MADD in 25 patients. Muscle MRI of the thigh and leg were performed in all patients before treatment. Eight patients received MRI re-examinations after treatment. RESULTS: All patients presented with muscle weakness or exercise intolerance associated with variants in the electron transfer flavoprotein dehydrogenase gene. Muscle MRI showed a sign of both edema-like change and fat infiltration selectively involving in the soleus (SO) but sparing of the gastrocnemius (GA) in the leg. Similar sign of selective involvement of the biceps femoris longus (BFL) but sparing of the semitendinosus (ST) was observed in the thigh. The sensitivity and specificity of the combination of either "SO+/GA-" sign or "BFL+/ST-" sign for the diagnosis of late-onset MADD were 80.0% and 83.5%, respectively. Logistic regression model supported the findings. The edema-like change in the SO and BFL muscles were quickly recovered at 1 month after treatment, and the clinical symptom was also relieved. CONCLUSIONS: This study expands the clinical and genetic spectrums of late-onset MADD. Muscle MRI shows a distinct pattern in the lower limb of patients with late-onset MADD. The dynamic change of edema-like change in the affected muscles might be a potential biomarker of treatment response.


Subject(s)
Magnetic Resonance Imaging/methods , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnostic imaging , Muscle, Skeletal/diagnostic imaging , Adolescent , Adult , Biopsy/methods , Carnitine/analogs & derivatives , Carnitine/blood , Electron-Transferring Flavoproteins/genetics , Female , Hamstring Muscles/diagnostic imaging , Hamstring Muscles/metabolism , Hamstring Muscles/pathology , Humans , Iron-Sulfur Proteins/genetics , Male , Middle Aged , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Oxidoreductases Acting on CH-NH Group Donors/genetics , Retrospective Studies , Young Adult
9.
J Vet Intern Med ; 32(4): 1442-1446, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29957835

ABSTRACT

A 12-hours-old Paint filly was examined because of weakness and dull mentation after birth. Despite IV administered dextrose, the foal remained persistently hypoglycemic with increase in serum activity of muscle and liver enzymes. A postmortem diagnosis of lipid myopathy most similar to multiple acyl-CoA dehydrogenase deficiency (MADD) was confirmed by findings of myofiber lipid accumulation, elevated urine organic acids, and serum free acylcarnitines with respect to control foals. This report details a case of equine neonatal lipid storage myopathy with many biochemical characteristics of MADD. Lipid storage myopathies should be included as a differential diagnosis in foals with persistent weakness and hypoglycemia.


Subject(s)
Horse Diseases/diagnosis , Hypoglycemia/veterinary , Lipid Metabolism, Inborn Errors/veterinary , Animals , Carnitine/analogs & derivatives , Carnitine/blood , Diagnosis, Differential , Fatal Outcome , Female , Horse Diseases/pathology , Horses , Hypoglycemia/diagnosis , Hypoglycemia/pathology , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/pathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnosis , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/veterinary , Muscular Dystrophies/diagnosis , Muscular Dystrophies/pathology
10.
Chin Med J (Engl) ; 131(2): 144-150, 2018 Jan 20.
Article in English | MEDLINE | ID: mdl-29336361

ABSTRACT

BACKGROUND: Clinically, it is difficult to differentiate multiple acyl-CoA dehydrogenase deficiency (MADD) from immune-mediated necrotizing myopathy (IMNM) because they display similar symptoms. This study aimed to determine whether muscle magnetic resonance imaging (MRI) could be used for differential diagnosis between MADD and IMNM. METHODS: The study evaluated 25 MADD patients, confirmed by muscle biopsy and ETFDH gene testing, and 30 IMNM patients, confirmed by muscle biopsy. Muscles were assessed for edema and fatty replacement using thigh MRI (tMRI). Degrees and distribution patterns of fatty infiltration and edema in gluteus maximus and thigh muscles were compared. RESULTS: Total fatty infiltration and edema scores (median, [Q1, Q3]) were 4.00 (1.00, 15.00) and 0 (0, 4.00) in MADD and 14.50 (8.00, 20.75) and 22.00 (16.75, 32.00) in IMNM, respectively, which were significantly more severe in IMNM than that in MADD (P = 0.000 and P = 0.004, respectively). Edema scores for gluteus maximus, long head of biceps femoris, and semimembranosus were significantly higher in IMNM than in MADD (all P = 0.000). Fatty infiltration scores for anterior and medial compartments were significantly more severe in IMNM than that in MADD (all P = 0.000). CONCLUSION: Different patterns of muscle involvement on tMRI can contribute to differential diagnosis between MADD and IMNM when clinical suspicions alone are insufficient, thereby reducing the need for muscle biopsy.


Subject(s)
Magnetic Resonance Imaging/methods , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diagnostic imaging , Muscle, Skeletal/diagnostic imaging , Myositis/diagnostic imaging , Adult , Creatine Kinase/blood , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Muscle, Skeletal/pathology , Myositis/pathology
11.
Neuromuscul Disord ; 27(6): 581-584, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28433476

ABSTRACT

Multiple acyl-CoA dehydrogenation deficiency is genetically heterogenous metabolic disease with mutations in genes involved in electron transfer to the mitochondrial respiratory chain. Disease symptoms vary from severe neonatal form to late-onset presentation with metabolic acidosis, lethargy, vomiting, muscle pain and weakness. Riboflavin therapy has been shown to ameliorate diseases symptoms in some of these patients. Recently, mutations in FAD synthase have been described to cause multiple acyl-CoA dehydrogenation deficiency. We describe here the effect of riboflavin supplementation therapy in a previously reported adult patient with multiple acyl-CoA dehydrogenation deficiency having compound heterozygous gene variations in FLAD1 (MIM: 610595) encoding FAD synthase. We present thorough clinical history including laboratory investigations, muscle MRI, muscle biopsy and spiroergometric analyses comprising of a follow-up of 20 years. Our data suggest that patients with adult-onset multiple acyl-CoA dehydrogenation deficiency with FLAD1 gene mutations also benefit from long-term riboflavin therapy.


Subject(s)
Frameshift Mutation , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/diet therapy , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Mutation, Missense , Riboflavin/therapeutic use , Adult , Female , Heterozygote , Humans , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Muscle, Skeletal , Treatment Outcome
13.
Am J Hum Genet ; 98(6): 1130-1145, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27259049

ABSTRACT

Multiple acyl-CoA dehydrogenase deficiencies (MADDs) are a heterogeneous group of metabolic disorders with combined respiratory-chain deficiency and a neuromuscular phenotype. Despite recent advances in understanding the genetic basis of MADD, a number of cases remain unexplained. Here, we report clinically relevant variants in FLAD1, which encodes FAD synthase (FADS), as the cause of MADD and respiratory-chain dysfunction in nine individuals recruited from metabolic centers in six countries. In most individuals, we identified biallelic frameshift variants in the molybdopterin binding (MPTb) domain, located upstream of the FADS domain. Inasmuch as FADS is essential for cellular supply of FAD cofactors, the finding of biallelic frameshift variants was unexpected. Using RNA sequencing analysis combined with protein mass spectrometry, we discovered FLAD1 isoforms, which only encode the FADS domain. The existence of these isoforms might explain why affected individuals with biallelic FLAD1 frameshift variants still harbor substantial FADS activity. Another group of individuals with a milder phenotype responsive to riboflavin were shown to have single amino acid changes in the FADS domain. When produced in E. coli, these mutant FADS proteins resulted in impaired but detectable FADS activity; for one of the variant proteins, the addition of FAD significantly improved protein stability, arguing for a chaperone-like action similar to what has been reported in other riboflavin-responsive inborn errors of metabolism. In conclusion, our studies identify FLAD1 variants as a cause of potentially treatable inborn errors of metabolism manifesting with MADD and shed light on the mechanisms by which FADS ensures cellular FAD homeostasis.


Subject(s)
Frameshift Mutation/genetics , Mitochondrial Diseases/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Nucleotidyltransferases/genetics , Riboflavin/pharmacology , Vitamin B Complex/pharmacology , Adult , Blotting, Western , Case-Control Studies , Cells, Cultured , Electron Transport , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Flavin-Adenine Dinucleotide/metabolism , Gene Expression Profiling , Humans , Infant , Infant, Newborn , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/pathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/drug therapy , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mutagenesis, Site-Directed , Protein Binding , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin/drug effects , Skin/metabolism , Skin/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Young Adult
14.
Biochim Biophys Acta ; 1862(7): 1367-73, 2016 07.
Article in English | MEDLINE | ID: mdl-27085974

ABSTRACT

AIMS: Triglycerides droplets are massively stored in muscle in Lipid Storage Myopathies (LSM). We studied in muscle regulators of lipophagy, the expression of the transcription factor-EB (TFEB) (a master regulator of lysosomal biogenesis), and markers of autophagy which are induced by starvation and exert a transcriptional control on lipid catabolism. METHODS: We investigated the factors that regulate lipophagy in muscle biopsies from 6 patients with different types of LSM: 2 cases of riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency (MADD), 1 case of primary carnitine deficiency (CD), 2 cases of neutral lipid storage myopathy (NLSD-M), 1 case of carnitine-palmitoyl-transferase-II (CPT) deficiency. RESULTS: Conventional morphology and electron microscopy documented the lipid accumulation and its dramatic resolution after treatment. Muscle immunofluorescence showed that while in MADD and NLSD-M there was a co-localized expression of TFEB and p62-SQSTM1 (marker of protein aggregates) in some atrophic fibers, in CD and CPT-II deficiency the reaction was almost normal. In regenerating fibers, TFEB localized in the cytoplasm (inactive form), whereas in atrophic fibers it localized in the nuclei (active form). Lipid-accumulated/atrophic fibers did not display p62-positive protein aggregates, indicating, together with the LC3-II (marker of autophagosomes) and p62-SQSTM1 analysis, that the autophagic flux is often preserved and lipophagy occurs. CONCLUSION: In atrophic and regenerating fibers of patients with NLSD-M we observed TFEB over-expression; in other conditions autophagy markers are increased, suggesting lipophagy active role on human lipid metabolism.


Subject(s)
Cardiomyopathies/pathology , Carnitine O-Palmitoyltransferase/deficiency , Carnitine/deficiency , Hyperammonemia/pathology , Lipid Metabolism, Inborn Errors/pathology , Lipolysis , Metabolism, Inborn Errors/pathology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Muscles/pathology , Muscular Diseases/pathology , Muscular Dystrophies/pathology , Adolescent , Adult , Aged , Autophagy , Cardiomyopathies/metabolism , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Child , Female , Humans , Hyperammonemia/metabolism , Lipid Metabolism, Inborn Errors/metabolism , Male , Metabolism, Inborn Errors/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Muscles/metabolism , Muscular Diseases/metabolism , Muscular Dystrophies/metabolism
15.
Neuromuscul Disord ; 26(2): 170-5, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26821934

ABSTRACT

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.


Subject(s)
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
16.
Brain Dev ; 38(3): 293-301, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26403312

ABSTRACT

INTRODUCTION: An increasing number of adult patients have been diagnosed with fatty acid ß-oxidation disorders with the rising use of diagnostic technologies. In this study, clinical, biochemical, and molecular characteristics of 2 Japanese patients with adult-onset glutaric acidemia type II (GA2) were investigated and compared with those of pediatric cases. METHODS: The patients were a 58-year-old male and a 31-year-old male. In both cases, episodes of myopathic symptoms, including myalgia, muscle weakness, and liver dysfunction of unknown cause, had been noted for the past several years. Muscle biopsy, urinary organic acid analysis (OA), acylcarnitine (AC) analysis in dried blood spots (DBS) and serum, immunoblotting, genetic analysis, and an in vitro probe acylcarnitine (IVP) assay were used for diagnosis and investigation. RESULTS: In both cases, there was no obvious abnormality of AC in DBS or urinary OA, although there was a increase in medium- and long-chain ACs in serum; also, fat deposits were observed in the muscle biopsy. Immunoblotting and gene analysis revealed that both patients had GA2 due to a defect in electron transfer flavoprotein dehydrogenase (ETFDH). The IVP assay indicated no special abnormalities in either case. CONCLUSION: Late-onset GA2 is separated into the intermediate and myopathic forms. In the myopathic form, episodic muscular symptoms or liver dysfunction are primarily exhibited after later childhood. Muscle biopsy and serum (or plasma) AC analysis allow accurate diagnosis in contrast with other biochemical tests, such as analysis of AC in DBS, urinary OA, or the IVP assay, which show fewer abnormalities in the myopathic form compared to intermediate form.


Subject(s)
Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Adult , Age Factors , Carnitine/analogs & derivatives , Carnitine/blood , Humans , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/blood , Muscle Weakness/blood , Muscle Weakness/pathology , Muscular Diseases/blood , Muscular Diseases/pathology
17.
Orphanet J Rare Dis ; 9: 117, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25200064

ABSTRACT

BACKGROUND: Multiple acyl-CoA dehydrogenase deficiency (MADD) is an autosomal recessive disorder caused by deficiency of electron transfer flavoprotein or electron transfer flavoprotein dehydrogenase. The clinical picture of late-onset forms is highly variable with symptoms ranging from acute metabolic decompensations to chronic, mainly muscular problems or even asymptomatic cases. METHODS: All 350 cases of late-onset MADD reported in the literature to date have been analyzed and evaluated with respect to age at presentation, diagnostic delay, biochemical features and diagnostic parameters as well as response to treatment. RESULTS: Mean age at onset was 19.2 years. The mean delay between onset of symptoms and diagnosis was 3.9 years. Chronic muscular symptoms were more than twice as common as acute metabolic decompensations (85% versus 33% of patients, respectively). 20% had both acute and chronic symptoms. 5% of patients had died at a mean age of 5.8 years, while 3% of patients have remained asymptomatic until a maximum age of 14 years. Diagnosis may be difficult as a relevant number of patients do not display typical biochemical patterns of urine organic acids and blood acylcarnitines during times of wellbeing. The vast majority of patients carry mutations in the ETFDH gene (93%), while mutations in the ETFA (5%) and ETFB (2%) genes are the exceptions. Almost all patients with late-onset MADD (98%) are clearly responsive to riboflavin. CONCLUSIONS: Late-onset MADD is probably an underdiagnosed disease and should be considered in all patients with acute or chronic muscular symptoms or acute metabolic decompensation with hypoglycemia, acidosis, encephalopathy and hepatopathy. This may not only prevent patients from invasive diagnostic procedures such as muscle biopsies, but also help to avoid fatal metabolic decompensations.


Subject(s)
Genetic Heterogeneity , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Adult , Age of Onset , Female , Humans , Male , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Young Adult
19.
Mol Genet Metab ; 109(2): 154-60, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23628458

ABSTRACT

Multiple acyl-coenzyme A dehydrogenation deficiency (MADD) has a wide range of phenotypic variation ranging from a neonatal lethal form to a mild late-onset form. Our previous data showed that in a group of Chinese patients, a mild type of MADD characterized by myopathy with clinically no other systemic involvement was caused by mutations in electron transfer flavoprotein dehydrogenase (ETFDH) gene, which encodes electron transfer flavoprotein: ubiquinone oxidoreductase (ETF:QO). Coenzyme Q10 (CoQ10), a downstream electron receptor of ETF:QO was first reported deficient in muscle of MADD patients with ETFDH gene mutations. Nevertheless, this result was not confirmed in a recently published study. Therefore to elucidate muscle CoQ10 level in a large group of MADD patients may provide further insight into the pathomechanism and therapeutic strategies. In this study, we found that 34 riboflavin responsive patients with ETFDH gene mutations had an elevated CoQ10 pool in muscle by high performance liquid chromatography (HPLC). However, when CoQ10 levels were normalized to citrate synthase, a marker of mitochondrial mass, there was no significant difference between patients and normal controls. Meanwhile, the increased mitochondrial DNA copy number in muscle also supported that the elevated CoQ10 pool was mainly due to mitochondrial mass proliferation. The expression of CoQ10 biosynthesis genes showed no significant changes whereas genes involved in lipid metabolism, such as PPARα, were marked up regulated. Our results suggested that CoQ10 seems not to be a primary factor in riboflavin responsive MADD and the apparent increase in CoQ10 may be secondary to mitochondrial proliferation.


Subject(s)
Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Mitochondria, Muscle/physiology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Muscle, Skeletal/metabolism , Oxidoreductases Acting on CH-NH Group Donors/genetics , Ubiquinone/analogs & derivatives , Adolescent , Adult , Child , DNA, Mitochondrial/genetics , Female , Gene Expression , Humans , Lipid Metabolism , Male , Middle Aged , Mitochondria, Muscle/genetics , Mitochondrial Dynamics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/drug therapy , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Muscle, Skeletal/pathology , Mutation, Missense , Riboflavin/therapeutic use , Ubiquinone/genetics , Ubiquinone/metabolism , Young Adult
20.
J Proteomics ; 75(1): 221-8, 2011 Dec 10.
Article in English | MEDLINE | ID: mdl-21596162

ABSTRACT

Multiple acyl-CoA dehydrogenase deficiency (MADD) is a mitochondrial fatty acid oxidation disorder caused by mutations that affect electron transfer flavoprotein (ETF) or ETF:ubiquinone oxidoreductase (ETF-QO) or even due to unidentified disturbances of riboflavin metabolism. Besides all the available data on the molecular basis of FAO disorders, including MADD, the pathophysiological mechanisms underlying clinical phenotype development, namely at the mitochondrial level, are poorly understood. In order to contribute to the elucidation of these mechanisms, we isolated mitochondria from cultured fibroblasts, from a patient with a severe MADD presentation due to ETF-QO deficiency, characterize its mitochondrial proteome and compare it with normal controls. The used approach (2-DE-MS/MS) allowed the positive identification of 287 proteins in both patient and controls, presenting 35 of the significant differences in their relative abundance. Among the differentially expressed are proteins associated to binding/folding functions, mitochondrial antioxidant enzymes as well as proteins associated to apoptotic events. The overexpression of chaperones like Hsp60 or mitochondrial Grp75, antioxidant enzymes and apoptotic proteins reflects the mitochondrial response to a complete absence of ETF-QO. Our study provides a global perspective of the mitochondrial proteome plasticity in a severe case of MADD and highlights the main molecular pathways involved in its pathogenesis.


Subject(s)
Electron Transport Complex I/deficiency , Electron-Transferring Flavoproteins/deficiency , Mitochondria/metabolism , Mitochondrial Proteins/analysis , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/pathology , Proteome/analysis , Case-Control Studies , Electron Transport Complex I/metabolism , Electron-Transferring Flavoproteins/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Fibroblasts/metabolism , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Mutation , Oxidation-Reduction , Phenotype , Protein Binding , Protein Folding , Proteome/genetics , Proteome/metabolism , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...