Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Stem Cell Res ; 72: 103214, 2023 10.
Article in English | MEDLINE | ID: mdl-37769385

ABSTRACT

Glycogen storage disease type III (GSDIII) is an autosomal recessive disorder characterized by a deficiency of glycogen debranching enzyme (GDE) leading to cytosolic glycogen accumulation and inducing liver and muscle pathology. Skin fibroblasts from three GSDIII patients were reprogrammed into induced pluripotent stem cells (iPSCs) using non-integrated Sendai virus. All of the three lines exhibited normal morphology, expression of pluripotent markers, stable karyotype, potential of trilineage differentiation and absence of GDE expression, making them valuable tools for modeling GSDIII disease in vitro, studying pathological mechanisms and investigating potential treatments.


Subject(s)
Glycogen Debranching Enzyme System , Glycogen Storage Disease Type III , Induced Pluripotent Stem Cells , Humans , Glycogen Storage Disease Type III/metabolism , Glycogen Storage Disease Type III/pathology , Induced Pluripotent Stem Cells/metabolism , Liver/pathology , Muscles/metabolism , Muscles/pathology
2.
Orphanet J Rare Dis ; 17(1): 127, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35313948

ABSTRACT

BACKGROUND: Glycogen storage diseases (GSDs) are inherited glycogen metabolic disorders which have various subtypes. GSDs of type I, III, IV, VI, and IX show liver involvement and are considered as hepatic types of GSDs. Thus, liver transplantation (LT) has been proposed as a final therapy for these types of GSD. LT corrects the primary hepatic enzyme defect; however, the long-term outcomes of LT in these patients have not been extensively evaluated so far. There are few reports in the English literature about the outcome of GSD patients after LT. There has been no report from Iran. The present retrospective study aimed to evaluate the long-term outcomes of eight patients with GSD types I, III, and IV who underwent LT in the affiliated hospitals of Shiraz University of Medical Sciences, from March 2013 to June 2021. During this period, there were no patients with GSD VI and IX identified in this center. RESULTS: The median time of diagnosis of the GSDs and at transplant was 1 year and 11 years, respectively. All eight transplanted patients were alive at the time of follow-up in this study. None of them required a re-transplant. All of the patients showed normalized liver enzymes after LT with no sign of hypoglycemia. CONCLUSIONS: LT is an achievable treatment for end-stage hepatic involvement of GSDs with a cure for metabolic deficiency. Our experience in these eight patients shows a favorable outcome with no mortality and no major complication.


Subject(s)
Glycogen Storage Disease Type III , Glycogen Storage Disease Type I , Glycogen Storage Disease Type VI , Glycogen Storage Disease , Liver Transplantation , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/metabolism , Glycogen Storage Disease/surgery , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/metabolism , Glycogen Storage Disease Type I/surgery , Glycogen Storage Disease Type III/complications , Glycogen Storage Disease Type III/metabolism , Glycogen Storage Disease Type VI/complications , Glycogen Storage Disease Type VI/metabolism , Humans , Liver/metabolism , Retrospective Studies
3.
Mol Genet Genomic Med ; 9(10): e1779, 2021 10.
Article in English | MEDLINE | ID: mdl-34405590

ABSTRACT

BACKGROUND: The condition of uniparental disomy (UPD) occurs when an individual inherits two copies of a chromosome, or part of a chromosome, from one parent. Most cases of uniparental heterodisomy (UPhD) do not cause diseases, whereas cases of uniparental isodisomy (UPiD), while rare, may be pathogenic. Theoretically, UPiD may cause rare genetic diseases in a homozygous recessive manner. METHODS: A 4-year-old girl presented with congenital hearing loss, developmental delay, hepatomegaly, and other clinical features. She and her parents were genetically tested using trio whole exome sequencing (Trio-WES) and copy number variation sequencing (CNV-seq). In addition, we built a structural model to further examine the pathogenicity of the UPiD variants. RESULTS: Trio-WES identified a paternal UPiD in chromosome 1, and two homozygous pathogenic variants AGL c.4284T>G/p.Tyr1428* and USH2A c.6528T>A/p.Tyr2176* in the UPiD region. We further analyzed the pathogenicity of these two variations. The patient was diagnosed with Usher syndrome type 2A (USH2A) and glycogen storage disease type III (GSD3). CONCLUSIONS: Our study reports a rare case of a patient carrying two pathogenic variants of different genes caused by paternal UPiD, supporting the potential application of Trio-WES in detecting and facilitating the diagnosis of UPD.


Subject(s)
Chromosomes, Human, Pair 1 , Glycogen Storage Disease Type III/complications , Glycogen Storage Disease Type III/diagnosis , Paternal Inheritance , Uniparental Disomy , Usher Syndromes/complications , Usher Syndromes/diagnosis , Adult , Biomarkers , Child, Preschool , DNA Copy Number Variations , Female , Genetic Association Studies , Genetic Predisposition to Disease , Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease Type III/etiology , Glycogen Storage Disease Type III/metabolism , Humans , Male , Middle Aged , Models, Molecular , Pedigree , Sequence Analysis, DNA , Structure-Activity Relationship , Usher Syndromes/etiology , Usher Syndromes/metabolism , Exome Sequencing
4.
J Inherit Metab Dis ; 44(1): 226-239, 2021 01.
Article in English | MEDLINE | ID: mdl-33448466

ABSTRACT

Glycogen storage disease type IIIa (GSDIIIa) is an inborn error of carbohydrate metabolism caused by a debranching enzyme deficiency. A subgroup of GSDIIIa patients develops severe myopathy. The purpose of this study was to investigate whether acute nutritional ketosis (ANK) in response to ketone-ester (KE) ingestion is effective to deliver oxidative substrate to exercising muscle in GSDIIIa patients. This was an investigator-initiated, researcher-blinded, randomized, crossover study in six adult GSDIIIa patients. Prior to exercise subjects ingested a carbohydrate drink (~66 g, CHO) or a ketone-ester (395 mg/kg, KE) + carbohydrate drink (30 g, KE + CHO). Subjects performed 15-minute cycling exercise on an upright ergometer followed by 10-minute supine cycling in a magnetic resonance (MR) scanner at two submaximal workloads (30% and 60% of individual maximum, respectively). Blood metabolites, indirect calorimetry data, and in vivo 31 P-MR spectra from quadriceps muscle were collected during exercise. KE + CHO induced ANK in all six subjects with median peak ßHB concentration of 2.6 mmol/L (range: 1.6-3.1). Subjects remained normoglycemic in both study arms, but delta glucose concentration was 2-fold lower in the KE + CHO arm. The respiratory exchange ratio did not increase in the KE + CHO arm when workload was doubled in subjects with overt myopathy. In vivo 31 P MR spectra showed a favorable change in quadriceps energetic state during exercise in the KE + CHO arm compared to CHO in subjects with overt myopathy. Effects of ANK during exercise are phenotype-specific in adult GSDIIIa patients. ANK presents a promising therapy in GSDIIIa patients with a severe myopathic phenotype. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov identifier: NCT03011203.


Subject(s)
Beverages , Exercise , Glycogen Storage Disease Type III/diet therapy , Ketosis/chemically induced , Muscular Diseases/diet therapy , Adult , Blood Glucose/analysis , Carbohydrate Metabolism , Cross-Over Studies , Diet, Ketogenic , Dietary Carbohydrates , Esters/administration & dosage , Female , Glycogen Storage Disease Type III/metabolism , Humans , Ketones/administration & dosage , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Netherlands , Phenotype
5.
J Inherit Metab Dis ; 44(3): 521-533, 2021 05.
Article in English | MEDLINE | ID: mdl-33368379

ABSTRACT

Glycogen storage disorder type III (GSDIII) is a rare inborn error of metabolism due to loss of glycogen debranching enzyme activity, causing inability to fully mobilize glycogen stores and its consequent accumulation in various tissues, notably liver, cardiac and skeletal muscle. In the pediatric population, it classically presents as hepatomegaly with or without ketotic hypoglycemia and failure to thrive. In the adult population, it should also be considered in the differential diagnosis of left ventricular hypertrophy or hypertrophic cardiomyopathy, myopathy, exercise intolerance, as well as liver cirrhosis or fibrosis with subsequent liver failure. In this review article, we first present an overview of the biochemical and clinical aspects of GSDIII. We then focus on the recent findings regarding cardiac and neuromuscular impairment associated with the disease. We review new insights into the pathophysiology and clinical picture of this disorder, including symptomatology, imaging and electrophysiology. Finally, we discuss current and upcoming treatment strategies such as gene therapy aimed at the replacement of the malfunctioning enzyme to provide a stable and long-term therapeutic option for this debilitating disease.


Subject(s)
Genetic Therapy/methods , Glycogen Storage Disease Type III/therapy , Muscle, Skeletal/physiopathology , Adult , Animals , Child , Disease Models, Animal , Glycogen Storage Disease Type III/metabolism , Glycogen Storage Disease Type III/physiopathology , Hepatomegaly/metabolism , Humans , Hypoglycemia/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism
6.
Genet Med ; 21(12): 2686-2694, 2019 12.
Article in English | MEDLINE | ID: mdl-31263214

ABSTRACT

PURPOSE: In glycogen storage disease type III (GSD III), liver aminotransferases tend to normalize with age giving an impression that hepatic manifestations improve with age. However, despite dietary treatment, long-term liver complications emerge. We present a GSD III liver natural history study in children to better understand changes in hepatic parameters with age. METHODS: We reviewed clinical, biochemical, histological, and radiological data in pediatric patients with GSD III, and performed a literature review of GSD III hepatic findings. RESULTS: Twenty-six patients (median age 12.5 years, range 2-22) with GSD IIIa (n = 23) and IIIb (n = 3) were enrolled in the study. Six of seven pediatric patients showed severe fibrosis on liver biopsy (median [range] age: 1.25 [0.75-7] years). Markers of liver injury (aminotransferases), dysfunction (cholesterol, triglycerides), and glycogen storage (glucose tetrasaccharide, Glc4) were elevated at an early age, and decreased significantly thereafter (p < 0.001). Creatine phosphokinase was also elevated with no significant correlation with age (p = 0.4). CONCLUSION: Liver fibrosis can occur at an early age, and may explain the decrease in aminotransferases and Glc4 with age. Our data outlines the need for systematic follow-up and specific biochemical and radiological tools to monitor the silent course of the liver disease process.


Subject(s)
Glycogen Storage Disease Type III/pathology , Liver Cirrhosis/pathology , Adolescent , Biomarkers , Child , Child, Preschool , Cholesterol/analysis , Cholesterol/metabolism , Female , Glycogen , Glycogen Storage Disease/pathology , Glycogen Storage Disease Type I/pathology , Glycogen Storage Disease Type III/metabolism , Humans , Liver/pathology , Liver Cirrhosis/metabolism , Liver Diseases , Male , Oligosaccharides/analysis , Oligosaccharides/metabolism , Transaminases/analysis , Transaminases/metabolism , Triglycerides/analysis , Triglycerides/metabolism , Young Adult
7.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3407-3417, 2018 10.
Article in English | MEDLINE | ID: mdl-30076962

ABSTRACT

Glycogen disease type III (GSDIII), a rare incurable autosomal recessive disorder due to glycogen debranching enzyme deficiency, presents with liver, heart and skeletal muscle impairment, hepatomegaly and ketotic hypoglycemia. Muscle weakness usually worsens to fixed myopathy and cardiac involvement may present in about half of the patients during disease. Management relies on careful follow-up of symptoms and diet. No common agreement was reached on sugar restriction and treatment in adulthood. We administered two dietary regimens differing in their protein and carbohydrate content, high-protein (HPD) and high-protein/glucose-free (GFD), to our mouse model of GSDIII, starting at one month of age. Mice were monitored, either by histological, biochemical and molecular analysis and motor functional tests, until 10 months of age. GFD ameliorated muscle performance up to 10 months of age, while HPD showed little improvement only in young mice. In GFD mice, a decreased muscle glycogen content and fiber vacuolization was observed, even in aged animals indicating a protective role of proteins against skeletal muscle degeneration, at least in some districts. Hepatomegaly was reduced by about 20%. Moreover, the long-term administration of GFD did not worsen serum parameters even after eight months of high-protein diet. A decreased phosphofructokinase and pyruvate kinase activities and an increased expression of Krebs cycle and gluconeogenesis genes were seen in the liver of GFD fed mice. Our data show that the concurrent use of proteins and a strictly controlled glucose supply could reduce muscle wasting, and indicate a better metabolic control in mice with a glucose-free/high-protein diet.


Subject(s)
Diet, High-Protein/methods , Glycogen Storage Disease Type III/diet therapy , Hepatomegaly/diet therapy , Muscle, Skeletal/physiopathology , Animals , Citric Acid Cycle , Diet, High-Protein Low-Carbohydrate/methods , Disease Models, Animal , Female , Glycogen Storage Disease Type III/metabolism , Glycogen Storage Disease Type III/physiopathology , Hepatomegaly/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Phosphofructokinases/metabolism , Physical Conditioning, Animal , Pyruvate Kinase/metabolism , Treatment Outcome
8.
BMC Med Genet ; 19(1): 54, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29614965

ABSTRACT

BACKGROUND: Glycogen Storage Disease Type III (GSD III) is a rare autosomal recessive metabolic disorder caused by AGL gene mutation. There is significant heterogeneity between the clinical manifestations and the gene mutation of AGL among different ethnic groups. However, GSD III is rarely reported in Chinese population. CASE PRESENTATION: In this study, we aimed to study the genetic and clinical characteristics of four patients with GSD IIIa from China, especially the neurological manifestations. Meanwhile, we conducted a literature review of GSD IIIa cases reported in Chinese population to investigate the relationship between genotype and phenotype. CONCLUSIONS: Three different AGL gene mutations were identified in our patients: c.206dupA, c.1735 + 1G > T and c.2590 C>T. Moreover, progressive myopathy accompanied by elevated creatine kinase level was the main manifestation of our patients in adolescents. Our results showed that AGL c.206dupA was a novel mutation and caused severe clinical manifestations. AGL c.1735 + 1G > T might be a recurrent mutation in the Chinese population. Genetic analysis of AGL gene mutation combined with muscle magnetic resonance imaging (MRI) might provide greater benefit to the patient in diagnosing GSD IIIa, rather than an invasive diagnostic procedure of biopsy.


Subject(s)
Creatine Kinase/metabolism , Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease Type III/genetics , Muscular Diseases/genetics , Mutation , Child, Preschool , China , Female , Genetic Testing , Glycogen Storage Disease Type III/complications , Glycogen Storage Disease Type III/metabolism , Humans , Infant , Male , Muscular Diseases/etiology , Muscular Diseases/metabolism , Up-Regulation
9.
Mol Ther ; 26(3): 890-901, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29396266

ABSTRACT

Glycogen storage disease type III (GSDIII) is an autosomal recessive disorder caused by a deficiency of glycogen-debranching enzyme (GDE), which results in profound liver metabolism impairment and muscle weakness. To date, no cure is available for GSDIII and current treatments are mostly based on diet. Here we describe the development of a mouse model of GSDIII, which faithfully recapitulates the main features of the human condition. We used this model to develop and test novel therapies based on adeno-associated virus (AAV) vector-mediated gene transfer. First, we showed that overexpression of the lysosomal enzyme alpha-acid glucosidase (GAA) with an AAV vector led to a decrease in liver glycogen content but failed to reverse the disease phenotype. Using dual overlapping AAV vectors expressing the GDE transgene in muscle, we showed functional rescue with no impact on glucose metabolism. Liver expression of GDE, conversely, had a direct impact on blood glucose levels. These results provide proof of concept of correction of GSDIII with AAV vectors, and they indicate that restoration of the enzyme deficiency in muscle and liver is necessary to address both the metabolic and neuromuscular manifestations of the disease.


Subject(s)
Genetic Therapy , Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease Type III/genetics , Glycogen Storage Disease Type III/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Phenotype , Animals , Biomarkers , Blood Glucose , Dependovirus/genetics , Disease Models, Animal , Enzyme Activation , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glycogen/metabolism , Glycogen Debranching Enzyme System/metabolism , Glycogen Storage Disease Type III/diagnosis , Glycogen Storage Disease Type III/therapy , Hepatocytes/metabolism , Male , Mice , Mice, Knockout , Organ Specificity
11.
Ter Arkh ; 89(8): 88-94, 2017.
Article in Russian | MEDLINE | ID: mdl-28914857

ABSTRACT

Glycogen storage disease (GSD) is an inherited metabolic disorder characterized by early childhood lipid metabolic disturbances with potentially proatherogenic effects. The review outlines the characteristics of impaired lipid composition and other changes in the cardiovascular system in GSD types I and III. It analyzes the factors enabling and inhibiting the development of atherosclerosis in patients with GSD. The review describes the paradox of vascular resistance to the development of early atherosclerosis despite the proatherogenic composition of lipids in the patients of this group.


Subject(s)
Cardiovascular System , Glycogen Storage Disease Type III , Glycogen Storage Disease Type I , Lipid Metabolism , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/metabolism , Glycogen Storage Disease Type I/physiopathology , Glycogen Storage Disease Type III/complications , Glycogen Storage Disease Type III/metabolism , Glycogen Storage Disease Type III/physiopathology , Humans , Vascular Resistance
12.
Bone ; 86: 79-85, 2016 May.
Article in English | MEDLINE | ID: mdl-26924264

ABSTRACT

INTRODUCTION: Glycogen storage disease type III (GSDIII) is an inborn error of carbohydrate metabolism caused by deficient activity of glycogen debranching enzyme (GDE). It is characterized by liver, cardiac muscle and skeletal muscle involvement. The presence of systemic complications such as growth retardation, ovarian polycystosis, diabetes mellitus and osteopenia/osteoporosis has been reported. The pathogenesis of osteopenia/osteoporosis is still unclear. OBJECTIVES: The aim of the current study was to evaluate the bone mineral density (BMD) in GSDIII patients and the role of metabolic and endocrine factors and physical activity on bone status. METHODS: Nine GSDIII patients were enrolled (age 2-20years) and compared to eighteen age and sex matched controls. BMD was evaluated by Dual-emission-X-ray absorptiometry (DXA) and Quantitative ultrasound (QUS). Clinical and biochemical parameters of endocrine system function and bone metabolism were analyzed. Serum levels of the metabolic control markers were evaluated. Physical activity was evaluated by administering the International Physical Activity Questionnaire (IPAQ). RESULTS: GSDIII patients showed reduced BMD detected at both DXA and QUS, decreased serum levels of IGF-1, free IGF-1, insulin, calcitonin, osteocalcin (OC) and increased serum levels of C-terminal cross-linking telopeptide of type I collagen (CTX). IGF-1 serum levels inversely correlated with AST and ALT serum levels. DXA Z-score inversely correlated with cholesterol and triglycerides serum levels and directly correlated with IGF-1/IGFBP3 molar ratio. No difference in physical activity was observed between GSDIII patients and controls. DISCUSSION: Our data confirm the presence of reduced BMD in GSDIII. On the basis of the results, we hypothesized that metabolic imbalance could be the key factor leading to osteopenia, acting through different mechanisms: chronic hyperlipidemia, reduced IGF-1, Insulin and OC serum levels. Thus, the mechanism of osteopenia/osteoporosis in GSDIII is probably multifactorial and we speculate on the factors involved in its pathogenesis.


Subject(s)
Bone Density , Bone and Bones/metabolism , Glycogen Storage Disease Type III/metabolism , Glycogen Storage Disease Type III/physiopathology , Homeostasis , Absorptiometry, Photon , Adolescent , Biomarkers/blood , Calcitonin/blood , Case-Control Studies , Child , Child, Preschool , Cholesterol/blood , Exercise , Female , Glycogen Storage Disease Type III/blood , Hormones/blood , Humans , Male
13.
Comp Med ; 66(1): 41-51, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26884409

ABSTRACT

Glycogen storage disease type IIIa (GSD IIIa) is caused by a deficiency of glycogen debranching enzyme activity. Hepatomegaly, muscle degeneration, and hypoglycemia occur in human patients at an early age. Long-term complications include liver cirrhosis, hepatic adenomas, and generalized myopathy. A naturally occurring canine model of GSD IIIa that mimics the human disease has been described, with progressive liver disease and skeletal muscle damage likely due to excess glycogen deposition. In the current study, long-term follow-up of previously described GSD IIIa dogs until 32 mo of age (n = 4) and of family-owned GSD IIIa dogs until 11 to 12 y of age (n = 2) revealed that elevated concentrations of liver and muscle enzyme (AST, ALT, ALP, and creatine phosphokinase) decreased over time, consistent with hepatic cirrhosis and muscle fibrosis. Glycogen deposition in many skeletal muscles; the tongue, diaphragm, and heart; and the phrenic and sciatic nerves occurred also. Furthermore, the urinary biomarker Glc4, which has been described in many types of GSD, was first elevated and then decreased later in life. This urinary biomarker demonstrated a similar trend as AST and ALT in GSD IIIa dogs, indicating that Glc4 might be a less invasive biomarker of hepatocellular disease. Finally, the current study further demonstrates that the canine GSD IIIa model adheres to the clinical course in human patients with this disorder and is an appropriate model for developing novel therapies.


Subject(s)
Dog Diseases/metabolism , Glycogen Storage Disease Type III/veterinary , Age Factors , Animals , Biomarkers/blood , Biomarkers/urine , Disease Models, Animal , Disease Progression , Dog Diseases/pathology , Dogs , Female , Glycogen/metabolism , Glycogen Storage Disease Type III/metabolism , Glycogen Storage Disease Type III/pathology , Hepatomegaly/metabolism , Hepatomegaly/pathology , Hepatomegaly/veterinary , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/veterinary , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Muscular Diseases/veterinary , Species Specificity , Urolithiasis/metabolism , Urolithiasis/pathology , Urolithiasis/veterinary
14.
J Clin Neurosci ; 22(10): 1674-5, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26067541

ABSTRACT

We report a 25-year-old man with glycogenosis III who presented with a progressive 2 year history of fatigue, hand stiffness and cramping. The glycogenoses are a group of rare metabolic disorders which develop as a result of deficiencies in various enzymes involved in the metabolism of glycogen. Some, but not all, glycogenoses, may result in skeletal muscle pathology. Among those that result in vacuolar myopathic changes, glycogen storage disease III or debrancher enzyme deficiency, an autosomal recessive condition, is less commonly encountered than acid maltase (Type II) and myophosphorylase (Type V) deficiencies. Many patients with debrancher enzyme deficiency also have liver involvement. The neurological examination of our patient showed mild proximal limb weakness and decreased reflexes. He had elevated creatine kinase and aldolase levels. He also demonstrated some elevations in his liver function tests, suggesting possible liver involvement. A skeletal muscle biopsy demonstrated vacuolar myopathic changes (acid phosphatase negative) accompanied by focal endomysial fibrosis and chronic inflammation. An ultrastructural examination showed that his vacuoles were filled with glycogen material. An enzyme assay of skeletal muscle tissue showed a significant decrease in debrancher enzyme activity (11% of normal). We review the typical clinical presentation of patients with glycogenosis III and discuss the differential diagnoses of glycogenosis III versus the other glycogenoses resulting in vacuolar myopathy.


Subject(s)
Glycogen Storage Disease Type III/diagnosis , Muscle, Skeletal/pathology , Adult , Glycogen Storage Disease Type III/metabolism , Humans , Male , Muscle, Skeletal/metabolism
15.
Neurology ; 84(17): 1767-71, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25832663

ABSTRACT

OBJECTIVE: Glycogen storage disease type IIIa (GSDIIIa) is classically regarded as a glycogenosis with fixed weakness, but we hypothesized that exercise intolerance in GSDIIIa is related to muscle energy failure and that oral fructose ingestion could improve exercise tolerance in this metabolic myopathy. METHODS: We challenged metabolism with cycle-ergometer exercise and measured substrate turnover and oxidation rates using stable isotope methodology and indirect calorimetry in 3 patients and 6 age-matched controls on 1 day, and examined the effect of fructose ingestion on exercise tolerance in the patients on another day. RESULTS: Total fatty acid oxidation rates during exercise were higher in patients than controls, 32.1 (SE 1.2) vs 20.7 (SE 0.5; range 15.8-29.3) µmol/kg/min (p = 0.048), and oxidation of carbohydrates was lower in patients, 1.0 (SE 5.4) vs 38.4 (SE 8.0; range 23.0-77.1) µmol/kg/min (p = 0.024). Fructose ingestion improved exercise tolerance in the patients. CONCLUSION: Similar to patients with McArdle disease, in whom muscle glycogenolysis is also impaired, GSDIIIa is associated with a reduced skeletal muscle oxidation of carbohydrates and a compensatory increase in fatty acid oxidation, and fructose ingestion improves exercise tolerance. Our results indicate that GSDIIIa should not only be viewed as a glycogenosis with fixed skeletal muscle weakness, but should also be considered among the glycogenoses presenting with exercise-related dynamic symptoms caused by muscular energy deficiency. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that ingestion of fructose improves exercise tolerance in patients with GSDIIIa.


Subject(s)
Energy Metabolism/physiology , Exercise , Fructose/pharmacology , Glycogen Storage Disease Type III/metabolism , Muscle Weakness/metabolism , Muscle, Skeletal/metabolism , Adolescent , Adult , Fructose/administration & dosage , Glycogen Storage Disease Type III/diet therapy , Glycogen Storage Disease Type III/physiopathology , Humans , Muscle Weakness/diet therapy , Muscle, Skeletal/physiopathology , Young Adult
17.
Biochem Biophys Res Commun ; 455(1-2): 90-7, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25451272

ABSTRACT

We studied two patients from a nonconsanguineous family with life-long abnormal liver function, hepatomegaly and abnormal fatty acid profiles. Abnormal liver function, hypoglycemia and muscle weakness are observed in various genetic diseases, including medium-chain acyl-CoA dehydrogenase (MCAD) deficiency and glycogen storage diseases. The proband showed increased free fatty acids, mainly C8 and C10, resembling fatty acid oxidation disorder. However, no mutation was found in ACADM and ACADL gene. Sequencing of theamylo-alpha-1, 6-glucosidase, 4-alpha-glucanotransferase (AGL) gene showed that both patients were compound heterozygotes for c.118C > T (p.Gln40X) and c.753_756 del CAGA (p.Asp251Glufsx29), whereas their parents were each heterozygous for one of these mutations. The AGL protein was undetectable in EBV-B cells from the two patients. Transcriptome analysis demonstrated a significant different pattern of gene expression in both of patients' cells, including genes involving in the PPAR signaling pathway, fatty acid biosynthesis, lipid synthesis and visceral fat deposition and metabolic syndrome. This unique gene expression pattern is probably due to the absence of AGL, which potentially accounts for the observed clinical phenotypes of hyperlipidemia and hepatocyte steatosis in glycogen storage disease type IIIa.


Subject(s)
Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease Type III/genetics , Mutation , Acyl-CoA Dehydrogenase/deficiency , Adolescent , Cells, Cultured , Fatty Acids/biosynthesis , Fatty Acids, Unsaturated/biosynthesis , Gene Expression , Glycogen Storage Disease Type III/diagnosis , Glycogen Storage Disease Type III/metabolism , Humans , Lipid Metabolism, Inborn Errors/diagnosis , Male
18.
Vestn Ross Akad Med Nauk ; (7-8): 78-84, 2014.
Article in Russian | MEDLINE | ID: mdl-25563007

ABSTRACT

AIM: The purpose of the study was to assess mitochondrial dysfunction severity in patients with hepatic forms of glycogen storage disease (GSD). PATIENTS AND METHODS: We examined 53 children with GSD in the dynamics. Distribution of children by disease types was: 1st group--children with GSD type I, 2nd group--children with GSD type III, 3rd group--children with GSD type VI and IX; comparison group consisted of 34 healthy children. Intracellular dehydrogenases activity: succinate dehydrogenase (SDH), glycerol-3-phosphate-dehydrogenase (GPDH). nicotinamideadenin-H-dehydrogenase (NADH-D) and lactatdehydrogenase (LDH) was measured using the quantitative cytochemical method in the peripheral lymphocytes. RESULTS: It was revealed decrease of SDH- (p < 0.001) and GPDH-activities (p < 0.001), along with increase of the NADH-D activity (p < 0.05) in all patients with GSD, (SDH/ NADH-D) index was decreased (p < 0.001). LDH activity was increased in groups 1 (p < 0.05) and 3 (p < 0.01), compared with comparison group. The most pronounced intracellular enzymes activity deviations were observed in children with GSD type I, that correspond to more severe clinical form of GSD. It was found strong correlation between intracellular enzymes activity and both hepatomegaly level (R = 0.867) and metabolic acidosis severity (R = 0.987). CONCLUSION: Our investigation revealed features of mitochondrial dysfunction in children with GSD, depending on the GSD type. Activities of lymphocytes enzymes correlates with the main disease severity parameters and can be used as an additional diagnostic criteria in children with hepatic form of GSD.


Subject(s)
Glycogen Storage Disease Type III , Glycogen Storage Disease Type I , Glycogen Storage Disease Type VI , Liver , Lymphocytes/metabolism , Mitochondria/metabolism , Carbohydrate Metabolism , Child , Cytological Techniques/methods , Female , Glycogen Storage Disease Type I/diagnosis , Glycogen Storage Disease Type I/metabolism , Glycogen Storage Disease Type I/physiopathology , Glycogen Storage Disease Type III/diagnosis , Glycogen Storage Disease Type III/metabolism , Glycogen Storage Disease Type III/physiopathology , Glycogen Storage Disease Type VI/diagnosis , Glycogen Storage Disease Type VI/metabolism , Glycogen Storage Disease Type VI/physiopathology , Humans , Liver/metabolism , Liver/pathology , Liver/physiopathology , Male , Oxidoreductases/analysis , Oxidoreductases/classification , Oxidoreductases/metabolism , Severity of Illness Index , Statistics as Topic
19.
Mol Biol Rep ; 40(7): 4197-202, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23649758

ABSTRACT

Genetic deficiency of the glycogen debranching enzyme causes glycogen storage disease type III, an autosomal recessive inherited disorder. The gene encoding this enzyme is designated as AGL gene. The disease is characterized by fasting hypoglycemia, hepatomegaly, growth retardation, progressive myopathy and cardiomyopathy. In the present study, we present clinical features and molecular characterization of two consanguineous Tunisian siblings suffering from Glycogen storage disease type III. The full coding exons of the AGL gene and their corresponding exon-intron boundaries were amplified for the patients and their parents. Gene sequencing identified a novel single point mutation at the conserved polypyrimidine tract of intron 21 in a homozygous state (IVS21-8A>G). This variant cosegregated with the disease and was absent in 102 control chromosomes. In silico analysis using online resources showed a decreased score of the acceptor splice site of intron 21. RT-PCR analysis of the AGL splicing pattern revealed a 7 bp sequence insertion between exon 21 and exon 22 due to the creation of a new 3' splice site. The predicted mutant enzyme was truncated by the loss of 637 carboxyl-terminal amino acids as a result of premature termination. This novel mutation is the first mutation identified in the region of Bizerte and the tenth AGL mutation identified in Tunisia. Screening for this mutation can improve the genetic counseling and prenatal diagnosis of GSD III.


Subject(s)
Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease Type III/genetics , Introns , Point Mutation , Consanguinity , DNA Mutational Analysis , Female , Gene Order , Glycogen Storage Disease Type III/metabolism , Humans , Infant , Infant, Newborn , Male , RNA Splice Sites , Siblings , Tunisia
20.
Mol Genet Metab ; 109(1): 14-20, 2013 May.
Article in English | MEDLINE | ID: mdl-23507172

ABSTRACT

Myopathic symptoms in Glycogen Storage Disease Type IIIa (GSD IIIa) are generally ascribed to the muscle wasting that these patients suffer in adult life, but an inability to debranch glycogen likely also has an impact on muscle energy metabolism. We hypothesized that patients with GSD IIIa can experience exercise intolerance due to insufficient carbohydrate oxidation in skeletal muscle. Six patients aged 17-36-years were studied. We determined VO 2peak (peak oxygen consumption), the response to forearm exercise, and the metabolic and cardiovascular responses to cycle exercise at 70% of VO 2peak with either a saline or a glucose infusion. VO 2peak was below normal. Glucose improved the work capacity by lowering the heart rate, and increasing the peak work rate by 30% (108 W with glucose vs. 83 W with placebo, p=0.018). The block in muscle glycogenolytic capacity, combined with the liver involvement caused exercise intolerance with dynamic skeletal muscle symptoms (excessive fatigue and muscle pain), and hypoglycemia in 4 subjects. In this study we combined anaerobic and aerobic exercise to systematically study skeletal muscle metabolism and exercise tolerance in patients with GSD IIIa. Exercise capacity was significantly reduced, and our results indicate that this was due to a block in muscle glycogenolytic capacity. Our findings suggest that the general classification of GSD III as a glycogenosis characterized by fixed symptoms related to muscle wasting should be modified to include dynamic exercise-related symptoms of muscle fatigue. A proportion of the skeletal muscle symptoms in GSD IIIa, i.e. weakness and fatigue, may be related to insufficient energy production in muscle.


Subject(s)
Energy Metabolism , Fatigue/metabolism , Glycogen Storage Disease Type III/physiopathology , Muscle Weakness/metabolism , Adolescent , Adult , Body Mass Index , Carbohydrate Metabolism , Exercise , Fatigue/physiopathology , Female , Glycogen Storage Disease Type III/metabolism , Humans , Male , Muscle Weakness/physiopathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...