Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 400
Filtrar
1.
Magn Reson Med ; 91(3): 1115-1121, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38009988

RESUMO

PURPOSE: Glycogen storage disease type III (GSD III) is a rare inherited metabolic disease characterized by excessive accumulation of glycogen in liver, skeletal muscle, and heart. Currently, there are no widely available noninvasive methods to assess tissue glycogen levels and disease load. Here, we use glycogen nuclear Overhauser effect (glycoNOE) MRI to quantify hepatic glycogen levels in a mouse model of GSD III. METHODS: Agl knockout mice (n = 13) and wild-type controls (n = 10) were scanned for liver glycogen content using glycoNOE MRI. All mice were fasted for 12 to 16 h before MRI scans. GlycoNOE signal was quantified by fitting the Z-spectrum using a four-pool Voigt lineshape model. Next, the fitted direct water saturation pool was removed and glycoNOE signal was estimated from the integral of the residual Z spectrum within -0.6 to -1.4 ppm. Glycogen concentration was also measured ex vivo using a biochemical assay. RESULTS: GlycoNOE MRI clearly distinguished Agl knockout mice from wild-type controls, showing a statistically significant difference in glycoNOE signals in the livers across genotypes. There was a linear correlation between glycoNOE signal and glycogen concentration determined by the biochemical assay. The obtained glycoNOE maps of mouse livers also showed higher glycogen levels in Agl knockout mice compared to wild-type mice. CONCLUSION: GlycoNOE MRI was used successfully as a noninvasive method to detect liver glycogen levels in mice, suggesting the potential of this method to be applied to assess glycogen storage diseases.


Assuntos
Doença de Depósito de Glicogênio Tipo III , Animais , Camundongos , Doença de Depósito de Glicogênio Tipo III/diagnóstico por imagem , Doença de Depósito de Glicogênio Tipo III/genética , Glicogênio/metabolismo , Glicogênio Hepático , Modelos Animais de Doenças , Imageamento por Ressonância Magnética , Camundongos Knockout
2.
J Clin Invest ; 134(2)2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38015640

RESUMO

Glycogen storage disease type III (GSDIII) is a rare inborn error of metabolism affecting liver, skeletal muscle, and heart due to mutations of the AGL gene encoding for the glycogen debranching enzyme (GDE). No curative treatment exists for GSDIII. The 4.6 kb GDE cDNA represents the major technical challenge toward the development of a single recombinant adeno-associated virus-derived (rAAV-derived) vector gene therapy strategy. Using information on GDE structure and molecular modeling, we generated multiple truncated GDEs. Among them, an N-terminal-truncated mutant, ΔNter2-GDE, had a similar efficacy in vivo compared with the full-size enzyme. A rAAV vector expressing ΔNter2-GDE allowed significant glycogen reduction in heart and muscle of Agl-/- mice 3 months after i.v. injection, as well as normalization of histology features and restoration of muscle strength. Similarly, glycogen accumulation and histological features were corrected in a recently generated Agl-/- rat model. Finally, transduction with rAAV vectors encoding ΔNter2-GDE corrected glycogen accumulation in an in vitro human skeletal muscle cellular model of GSDIII. In conclusion, our results demonstrated the ability of a single rAAV vector expressing a functional mini-GDE transgene to correct the muscle and heart phenotype in multiple models of GSDIII, supporting its clinical translation to patients with GSDIII.


Assuntos
Sistema da Enzima Desramificadora do Glicogênio , Doença de Depósito de Glicogênio Tipo III , Humanos , Camundongos , Ratos , Animais , Doença de Depósito de Glicogênio Tipo III/genética , Doença de Depósito de Glicogênio Tipo III/terapia , Sistema da Enzima Desramificadora do Glicogênio/genética , Músculo Esquelético/metabolismo , Glicogênio/metabolismo , Transgenes
3.
Stem Cell Res ; 72: 103214, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37769385

RESUMO

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.


Assuntos
Sistema da Enzima Desramificadora do Glicogênio , Doença de Depósito de Glicogênio Tipo III , Células-Tronco Pluripotentes Induzidas , Humanos , Doença de Depósito de Glicogênio Tipo III/metabolismo , Doença de Depósito de Glicogênio Tipo III/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fígado/patologia , Músculos/metabolismo , Músculos/patologia
4.
Eur J Med Res ; 28(1): 253, 2023 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-37488624

RESUMO

The aim of the Protocole National De Diagnostic et de Soins/French National Protocol for Diagnosis and Healthcare (PNDS) is to provide advice for health professionals on the optimum care provision and pathway for patients with glycogen storage disease type III (GSD III).The protocol aims at providing tools that make the diagnosis, defining the severity and different damages of the disease by detailing tests and explorations required for monitoring and diagnosis, better understanding the different aspects of the treatment, defining the modalities and organisation of the monitoring. This is a practical tool, to which health care professionals can refer. PNDS cannot, however, predict all specific cases, comorbidities, therapeutic particularities or hospital care protocols, and does not seek to serve as a substitute for the individual responsibility of the physician in front of his/her patient.


Assuntos
Doença de Depósito de Glicogênio Tipo III , Médicos , Humanos , Feminino , Masculino , Pessoal de Saúde , Hospitais
5.
JCI Insight ; 7(23)2022 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-36264632

RESUMO

Glycogen debranching enzyme deficiency in glycogen storage disease type III (GSD III) results in excessive glycogen accumulation in multiple tissues, primarily the liver, heart, and skeletal muscle. We recently reported that an adeno-associated virus vector expressing a bacterial debranching enzyme (pullulanase) driven by the ubiquitous CMV enhancer/chicken ß-actin (CB) promoter cleared glycogen in major affected tissues of infant GSD IIIa mice. In this study, we developed a potentially novel dual promoter consisting of a liver-specific promoter (LSP) and the CB promoter for gene therapy in adult GSD IIIa mice. Ten-week treatment with an adeno-associated virus vector containing the LSP-CB dual promoter in adult GSD IIIa mice significantly increased pullulanase expression and reduced glycogen contents in the liver, heart, and skeletal muscle, accompanied by the reversal of liver fibrosis, improved muscle function, and a significant decrease in plasma biomarkers alanine aminotransferase, aspartate aminotransferase, and creatine kinase. Compared with the CB promoter, the dual promoter effectively decreased pullulanase-induced cytotoxic T lymphocyte responses and enabled persistent therapeutic gene expression in adult GSD IIIa mice. Future studies are needed to determine the long-term durability of dual promoter-mediated expression of pullulanase in adult GSD IIIa mice and in large animal models.


Assuntos
Doença de Depósito de Glicogênio Tipo III , Camundongos , Animais , Linfócitos T Citotóxicos , Terapia Genética , Glicogênio
6.
BMC Pediatr ; 22(1): 284, 2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35578201

RESUMO

BACKGROUND: Glycogen storage disease type III (GSD III) is a rare autosomal recessive glycogenolysis disorder due to AGL gene variants, characterized by hepatomegaly, fasting hypoglycemia, hyperlipidemia, elevated hepatic transaminases, growth retardation, progressive myopathy, and cardiomyopathy. However, it is not easy to make a definite diagnosis in early stage of disease only based on the clinical phenotype and imageology due to its clinical heterogeneity. CASE PRESENTATION: We report a two-year-old girl with GSD III from a nonconsanguineous Chinese family, who presented with hepatomegaly, fasting hypoglycemia, hyperlipidemia, elevated levels of transaminases. Accordingly, Sanger sequencing, whole­exome sequencing of family trios, and qRT-PCR was performed, which revealed that the patient carried the compound heterogeneous variants, a novel frameshift mutation c.597delG (p. Q199Hfs*2) and a novel large gene fragment deletion of the entire exon 13 in AGL gene. The deletion of AGL was inherited from the proband's father and the c.597delG variant was from the mother. CONCLUSIONS: In this study, we identified two novel variants c.597delG (p. Q199Hfs*2) and deletion of the entire exon 13 in AGL in a Chinese GSD III patient. We extend the mutation spectrum of AGL. We suggest that high-throughput sequencing technology can detect and screen pathogenic variant, which is a scientific basis about genetic counseling and clinical diagnosis.


Assuntos
Doença de Depósito de Glicogênio Tipo III , Hipoglicemia , China , Doença de Depósito de Glicogênio Tipo III/diagnóstico , Doença de Depósito de Glicogênio Tipo III/genética , Hepatomegalia , Humanos , Mutação , Transaminases
7.
Orphanet J Rare Dis ; 17(1): 127, 2022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35313948

RESUMO

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.


Assuntos
Doença de Depósito de Glicogênio Tipo III , Doença de Depósito de Glicogênio Tipo I , Doença de Depósito de Glicogênio Tipo VI , Doença de Depósito de Glicogênio , Transplante de Fígado , Doença de Depósito de Glicogênio/diagnóstico , Doença de Depósito de Glicogênio/metabolismo , Doença de Depósito de Glicogênio/cirurgia , Doença de Depósito de Glicogênio Tipo I/complicações , Doença de Depósito de Glicogênio Tipo I/metabolismo , Doença de Depósito de Glicogênio Tipo I/cirurgia , Doença de Depósito de Glicogênio Tipo III/complicações , Doença de Depósito de Glicogênio Tipo III/metabolismo , Doença de Depósito de Glicogênio Tipo VI/complicações , Doença de Depósito de Glicogênio Tipo VI/metabolismo , Humanos , Fígado/metabolismo , Estudos Retrospectivos
8.
Orphanet J Rare Dis ; 17(1): 28, 2022 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-35101075

RESUMO

BACKGROUND: Individuals with glycogen storage disease IIIa (GSD IIIa) (OMIM #232400) experience muscle weakness and exercise limitation that worsen through adulthood. However, normative data for markers of physical capacity, such as strength and cardiovascular fitness, are limited. Furthermore, the impact of the disease on muscle size and quality is unstudied in weight bearing skeletal muscle, a key predictor of physical function. We aim to produce normative reference values of aerobic capacity and strength in individuals with GSD IIIa, and to investigate the role of muscle size and quality on exercise impairment. RESULTS: Peak oxygen uptake (V̇O2peak) was lower in the individuals with GSD IIIa than predicted based on demographic data (17.0 (9.0) ml/kg/min, 53 (24)% of predicted, p = 0.001). Knee extension maximum voluntary contraction (MVC) was also substantially lower than age matched predicted values (MVC: 146 (116) Nm, 57% predicted, p = 0.045), though no difference was found in MVC relative to body mass (1.88 (2.74) Nm/kg, 61% of predicted, p = 0.263). There was a strong association between aerobic capacity and maximal leg strength (r = 0.920; p = 0.003). Substantial inter-individual variation was present, with a high physical capacity group that had normal leg strength (MVC), and relatively high V̇O2peak, and a low physical capacity that display impaired strength and substantially lower V̇O2peak. The higher physical capacity sub-group were younger, had larger Vastus Lateralis (VL) muscles, greater muscle quality, undertook more physical activity (PA), and reported higher health-related quality of life. CONCLUSIONS: V̇O2peak and knee extension strength are lower in individuals with GSD IIIa than predicted based on their demographic data. Patients with higher physical capacity have superior muscle size and structure characteristics and higher health-related quality of life, than those with lower physical capacity. This study provides normative values of these important markers of physical capacity.


Assuntos
Doença de Depósito de Glicogênio Tipo III , Qualidade de Vida , Adulto , Exercício Físico/fisiologia , Humanos , Força Muscular/fisiologia , Músculo Esquelético
9.
J Pediatr Endocrinol Metab ; 35(4): 451-462, 2022 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-35038814

RESUMO

OBJECTIVES: To reveal the different clinical presentations of liver glycogen storage disease type IX (GSD IX), which is a clinically and genetically heterogeneous type of glycogenosis. METHODS: The data from the electronic hospital records of 25 patients diagnosed with liver GSD IX was reviewed. Symptoms, clinical findings, and laboratory and molecular analysis were assessed. RESULTS: Of the patients, 10 had complaints of short stature in the initial presentation additionally other clinical findings. Elevated serum transaminases were found in 20 patients, and hepatomegaly was found in 22 patients. Interestingly, three patients were referred due to neurodevelopmental delay and hypotonia, while one was referred for only autism. One patient who presented with neurodevelopmental delay developed hepatomegaly and elevated transaminases during the disease later on. Three of the patients had low hemoglobin A1C and fructosamine values that were near the lowest reference range. Two patients had left ventricular hypertrophy. Three patients developed osteopenia during follow-up, and one patient had osteoporosis after puberty. The most common gene variant, PHKA2, was observed in 16 patients, 10 variants were novel and six variants were defined before. Six patients had variants in PHKG2, two variants were not defined before and four variants were defined before. PHKB variants were found in three patients. One patient had two novel splice site mutations in trans position. It was revealed that one novel homozygous variant and one defined homozygous variant were found in PHKB. CONCLUSIONS: This study revealed that GSD IX may present with only hypotonia and neurodevelopmental delay without liver involvement in the early infantile period. It should be emphasized that although liver GSDIX is thought of as a benign disease, it might present with multisystemic involvement and patients should be screened with echocardiography, bone mineral densitometry, and psychometric evaluation.


Assuntos
Doença de Depósito de Glicogênio Tipo III , Doença de Depósito de Glicogênio , Doença de Depósito de Glicogênio/diagnóstico , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio Tipo III/diagnóstico , Doença de Depósito de Glicogênio Tipo III/genética , Hepatomegalia , Humanos , Mutação , Fosforilase Quinase/genética
11.
Rev. Asoc. Colomb. Cien. Biol. (En línea) ; 1(34): 10-17, 2022. tab, ilus
Artigo em Espanhol | LILACS, COLNAL | ID: biblio-1372379

RESUMO

Introducción: La enfermedad por almacenamiento del glucógeno tipo III (GSDIII, Glycogen storage disease type III) o Enfermedad de Cori Forbes es un trastorno del proceso de glucogenólisis ocasionado por variantes del gen AGL que codifica la enzima desramificante del glucógeno; se encuentra ubicado en el cromosoma 1p21.2 y su alteración genera una degradación incompleta del glucógeno, llevando a una acumulación de dextrina límite en órganos blanco, ocasionando organomegalia y disfunción. Objetivo: Caracterizar molecularmente un paciente lactante mayor con diagnóstico clínico y bioquímico sospechoso de GSDIII. Materiales y Métodos: Paciente lactante mayor masculino con antecedente de displasia broncopulmonar, infección respiratoria aguda, reflujo gastroesofágico, hepatomegalia e intolerancia a la lactosa. Se realizó estudio molecular mediante secuenciación de exoma completo; las variantes reportadas fueron evaluadas por Software de predicción como: Mutation Tas-ter, PROVEAN, UMD-Predictor, POLYPHEN, SIFT, Human Splicing Finder. Finalmente, se realizó una red de interacción génica mediante el programa GeneMania para determinar asociaciones génicas cercanas. Resultados: Se identifi caron 3 variantes heterocigotas ubicadas en el gen AGL: p.Arg910* que ocasiona pérdida del dominio amilo-1,6 glucosidasa y el dominio de unión al glucógeno, y las variantes p.Trp373Cys, p.Asn565Ser que generan cambios missense en la proteína. El análisis de significancia clínica por medio de métodos in-sílico determinó una clasificación patogénica para todas las variantes. La red de interacción permitió observar asociaciones entre el gen AGL y los genes FOXA2, PPP1R3B, NHLRC1 y GCK, que tienen relación con procesos metabólicos. Conclusión: una sospecha clínica inicial, a través de una buena historia clínica y la pertinencia de estudios bioquímicos-metabólicos-genómicos dirigidos, permite brindar un correcto diagnóstico, tratamiento y seguimiento, acercándonos a la medicina de precisión.


Introduction: Glycogen storage disease type III (GSDIII) or Cori Forbes disease is a disorder of the glycogeno-lysis process caused by variants of the AGL gene that encodes the glycogen debranching enzyme; It is located on chromosome 1p21.2 and its alteration generate an incomplete degradation of glycogen, leading to an accumu-lation of borderline dextrin in target organs, causing organomegaly and dysfunction. Objective: To characterize at the molecular level an elderly male lactating patient from southwestern Colombia with a clinical, biochemical diagnosis suspected of GSDIII. Materials and methods: An elderly male infant with a history of bronchopul-monary dysplasia, acute respiratory infection, gastroesophageal refl ux, hepatomegaly, and lactose intolerance. A molecular study was performed by whole exome sequencing; the reported variants were evaluated by prediction software such as Mutation Taster, PROVEAN, UMD-Predictor, POLYPHEN, SIFT, Human Splicing Finder. Fi-nally, a gene interaction network was performed using the GeneMania program to determine close gene associa-tions. Results: 3 heterozygous variants located in the AGL gene were identifi ed: p.Arg910 * that causes loss of the amyl-1,6 glucosidase domain and the glycogen-binding domain, and the variants p.Trp373Cys, p.Asn565 in the protein. The analysis of clinical signifi cance by means of in-silico methods determined a pathogenic classifi cation for all the variants. The interaction network will observe associations between the AGL gene and the FOXA2, PPP1R3B, NHLRC1 and GCK genes, which are related to metabolic processes. Conclusion: an initial clinical suspicion, through a good clinical history and the relevance of directed biochemical-metabolic-genomic studies, allows us to provide a correct diagnosis, treatment, and follow-up, bringing us closer to precision medicine


Assuntos
Humanos , Masculino , Lactente , Biologia Computacional , Doença de Depósito de Glicogênio Tipo III , Colômbia
12.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 38(11): 1073-1076, 2021 Nov 10.
Artigo em Chinês | MEDLINE | ID: mdl-34729746

RESUMO

OBJECTIVE: To explore the clinical features and genetic basis of two children with glycogen storage disease type III (GSD III). METHODS: The probands and their parents were subjected to genetic testing, and the pathogenity of candidate variants was analyzed by using bioinformatic tools. RESULTS: Sequencing has identified compound heterozygous variants of the AGL gene in both children, namely c.1423+1G>A and c.3701-2A>G in case 1, and c.4213_c.4214insA (p.Glu1405Glufs*17) and c.3589-3C>G in case 2. Both children were diagnosed with GSD III. Literature review suggested that the main type variant among Chinese patients with GSD III involve splice sites of the AGL gene, with c.1735+1G>T being the most common. Based on the American College of Medical Genetics and Genomics standards and guidelines,c.1423+1G>A, c.3701-2A>G and c.4213_c.4214insA variants of AGL gene were predicted to be of pathogenic (PVS1+PM2+PM3, PVS1+PM2+PM3, PVS1+PM2+PP5), and c.3589-3C>G variant was predicted to be of uncertain significance (PM2+PM3+PP3). CONCLUSION: The compound heterozygous variants of the AGL gene probably underlay the GSD III in both children. Above findings have enriched the spectrum of genetic variants underlying this disease.


Assuntos
Doença de Depósito de Glicogênio Tipo III , Criança , Testes Genéticos , Genômica , Doença de Depósito de Glicogênio Tipo III/genética , Humanos , Mutação
13.
Mol Genet Metab ; 134(3): 223-234, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34649782

RESUMO

INTRODUCTION: Early recognized manifestations of GSD III include hypoglycemia, hepatomegaly, and elevated liver enzymes. Motor symptoms such as fatigue, muscle weakness, functional impairments, and muscle wasting are typically reported in the 3rd to 4th decade of life. OBJECTIVE: In this study, we investigated the early musculoskeletal findings in children with GSD IIIa, compared to a cohort of adults with GSD IIIa. METHODS: We utilized a comprehensive number of physical therapy outcome measures to cross-sectionally assess strength and gross motor function including the modified Medical Research Council (mMRC) scale, grip and lateral/key pinch, Gross Motor Function Measure (GMFM), Gait, Stairs, Gowers, Chair (GSGC) test, 6 Minute Walk Test (6MWT), and Bruininks-Oseretsky Test of Motor Proficiency Ed. 2 (BOT-2). We also assessed laboratory biomarkers (AST, ALT, CK and urine Glc4) and conducted whole-body magnetic resonance imaging (WBMRI) to evaluate for proton density fat fraction (PDFF) in children with GSD IIIa. Nerve Conduction Studies and Electromyography results were analyzed where available and a thorough literature review was conducted. RESULTS: There were a total of 22 individuals with GSD IIIa evaluated in our study, 17 pediatric patients and 5 adult patients. These pediatric patients demonstrated weakness on manual muscle testing, decreased grip and lateral/key pinch strength, and decreased functional ability compared to non-disease peers on the GMFM, 6MWT, BOT-2, and GSGC. Additionally, all laboratory biomarkers analyzed and PDFF obtained from WBMRI were increased in comparison to non-diseased peers. In comparison to the pediatric cohort, adults demonstrated worse overall performance on functional assessments demonstrating the expected progression of disease phenotype with age. CONCLUSION: These results demonstrate the presence of early musculoskeletal involvement in children with GSD IIIa, most evident on physical therapy assessments, in addition to the more commonly reported hepatic symptoms. Muscular weakness in both children and adults was most significant in proximal and trunk musculature, and intrinsic musculature of the hands. These findings indicate the importance of early assessment of patients with GSD IIIa for detection of muscular weakness and development of treatment approaches that target both the liver and muscle.


Assuntos
Doença de Depósito de Glicogênio Tipo III/diagnóstico por imagem , Imageamento por Ressonância Magnética/estatística & dados numéricos , Modalidades de Fisioterapia/normas , Imagem Corporal Total/estatística & dados numéricos , Adolescente , Adulto , Criança , Pré-Escolar , Estudos de Coortes , Estudos Transversais , Feminino , Humanos , Lactente , Recém-Nascido , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Debilidade Muscular/patologia , Músculo Esquelético/patologia , Imagem Corporal Total/normas , Adulto Jovem
14.
Mol Genet Genomic Med ; 9(10): e1779, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34405590

RESUMO

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.


Assuntos
Cromossomos Humanos Par 1 , Doença de Depósito de Glicogênio Tipo III/complicações , Doença de Depósito de Glicogênio Tipo III/diagnóstico , Herança Paterna , Dissomia Uniparental , Síndromes de Usher/complicações , Síndromes de Usher/diagnóstico , Adulto , Biomarcadores , Pré-Escolar , Variações do Número de Cópias de DNA , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Sistema da Enzima Desramificadora do Glicogênio/química , Sistema da Enzima Desramificadora do Glicogênio/genética , Doença de Depósito de Glicogênio Tipo III/etiologia , Doença de Depósito de Glicogênio Tipo III/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Linhagem , Análise de Sequência de DNA , Relação Estrutura-Atividade , Síndromes de Usher/etiologia , Síndromes de Usher/metabolismo , Sequenciamento do Exoma
16.
J Clin Neuromuscul Dis ; 22(4): 224-227, 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-34019008

RESUMO

ABSTRACT: Glycogen storage disease type III is a rare inherited disease caused by decreased activity of glycogen debranching enzyme. It affects primarily the liver, cardiac muscle, and skeletal muscle. Pure involvement of the skeletal muscle with adult onset is extremely rare. We report on a patient with myopathy due to glycogen storage disease III, and describe the clinical features, and pathologic and genetic findings.


Assuntos
Doença de Depósito de Glicogênio Tipo III/diagnóstico , Doenças Musculares/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético
17.
J Inherit Metab Dis ; 44(1): 226-239, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33448466

RESUMO

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.


Assuntos
Bebidas , Exercício Físico , Doença de Depósito de Glicogênio Tipo III/dietoterapia , Cetose/induzido quimicamente , Doenças Musculares/dietoterapia , Adulto , Glicemia/análise , Metabolismo dos Carboidratos , Estudos Cross-Over , Dieta Cetogênica , Carboidratos da Dieta , Ésteres/administração & dosagem , Feminino , Doença de Depósito de Glicogênio Tipo III/metabolismo , Humanos , Cetonas/administração & dosagem , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Doenças Musculares/metabolismo , Países Baixos , Fenótipo
19.
J Inherit Metab Dis ; 44(3): 521-533, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33368379

RESUMO

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.


Assuntos
Terapia Genética/métodos , Doença de Depósito de Glicogênio Tipo III/terapia , Músculo Esquelético/fisiopatologia , Adulto , Animais , Criança , Modelos Animais de Doenças , Doença de Depósito de Glicogênio Tipo III/metabolismo , Doença de Depósito de Glicogênio Tipo III/fisiopatologia , Hepatomegalia/metabolismo , Humanos , Hipoglicemia/metabolismo , Fígado/metabolismo , Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo
20.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-922000

RESUMO

OBJECTIVE@#To explore the clinical features and genetic basis of two children with glycogen storage disease type III (GSD III).@*METHODS@#The probands and their parents were subjected to genetic testing, and the pathogenity of candidate variants was analyzed by using bioinformatic tools.@*RESULTS@#Sequencing has identified compound heterozygous variants of the AGL gene in both children, namely c.1423+1G>A and c.3701-2A>G in case 1, and c.4213_c.4214insA (p.Glu1405Glufs*17) and c.3589-3C>G in case 2. Both children were diagnosed with GSD III. Literature review suggested that the main type variant among Chinese patients with GSD III involve splice sites of the AGL gene, with c.1735+1G>T being the most common. Based on the American College of Medical Genetics and Genomics standards and guidelines,c.1423+1G>A, c.3701-2A>G and c.4213_c.4214insA variants of AGL gene were predicted to be of pathogenic (PVS1+PM2+PM3, PVS1+PM2+PM3, PVS1+PM2+PP5), and c.3589-3C>G variant was predicted to be of uncertain significance (PM2+PM3+PP3).@*CONCLUSION@#The compound heterozygous variants of the AGL gene probably underlay the GSD III in both children. Above findings have enriched the spectrum of genetic variants underlying this disease.


Assuntos
Criança , Humanos , Testes Genéticos , Genômica , Doença de Depósito de Glicogênio Tipo III/genética , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...