Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Hum Gene Ther ; 30(10): 1263-1273, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31319709

RESUMEN

Glycogen storage diseases (GSDs) type I (GSDI) and type III (GSDIII), the most frequent hepatic GSDs, are due to defects in glycogen metabolism, mainly in the liver. In addition to hypoglycemia and liver pathology, renal, myeloid, or muscle complications affect GSDI and GSDIII patients. Currently, patient management is based on dietary treatment preventing severe hypoglycemia and increasing the lifespan of patients. However, most of the patients develop long-term pathologies. In the past years, gene therapy for GSDI has generated proof of concept for hepatic GSDs. This resulted in a recent clinical trial of adeno-associated virus (AAV)-based gene replacement for GSDIa. However, the current limitations of AAV-mediated gene transfer still represent a challenge for successful gene therapy in GSDI and GSDIII. Indeed, transgene loss over time was observed in GSDI liver, possibly due to the degeneration of hepatocytes underlying the physiopathology of both GSDI and GSDIII and leading to hepatic tumor development. Moreover, multitissue targeting requires high vector doses to target nonpermissive tissues such as muscle and kidney. Interestingly, recent pharmacological interventions or dietary regimen aiming at the amelioration of the hepatocyte abnormalities before the administration of gene therapy demonstrated improved efficacy in GSDs. In this review, we describe the advances in gene therapy and the limitations to be overcome to achieve efficient and safe gene transfer in GSDs.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/terapia , Enfermedad del Almacenamiento de Glucógeno Tipo I/terapia , Hipoglucemia/terapia , Animales , Ensayos Clínicos como Asunto , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Glucógeno/biosíntesis , Enfermedad del Almacenamiento de Glucógeno Tipo I/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/patología , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/patología , Hepatocitos/enzimología , Hepatocitos/patología , Humanos , Hipoglucemia/enzimología , Hipoglucemia/genética , Hipoglucemia/patología , Hígado/enzimología , Hígado/patología , Transgenes
2.
Nat Commun ; 7: 11229, 2016 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-27088557

RESUMEN

Glycogen is a branched glucose polymer and serves as an important energy store. Its debranching is a critical step in its mobilization. In animals and fungi, the 170 kDa glycogen debranching enzyme (GDE) catalyses this reaction. GDE deficiencies in humans are associated with severe diseases collectively termed glycogen storage disease type III (GSDIII). We report crystal structures of GDE and its complex with oligosaccharides, and structure-guided mutagenesis and biochemical studies to assess the structural observations. These studies reveal that distinct domains in GDE catalyse sequential reactions in glycogen debranching, the mechanism of their catalysis and highly specific substrate recognition. The unique tertiary structure of GDE provides additional contacts to glycogen besides its active sites, and our biochemical experiments indicate that they mediate its recruitment to glycogen and regulate its activity. Combining the understanding of the GDE catalysis and functional characterizations of its disease-causing mutations provides molecular insights into GSDIII.


Asunto(s)
Candida glabrata/enzimología , Proteínas Fúngicas/química , Sistema de la Enzima Desramificadora del Glucógeno/química , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Biocatálisis , Candida glabrata/genética , Dominio Catalítico , Cristalografía por Rayos X , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Glucógeno/química , Glucógeno/metabolismo , Sistema de la Enzima Desramificadora del Glucógeno/genética , Sistema de la Enzima Desramificadora del Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Oligosacáridos/química , Oligosacáridos/metabolismo , Unión Proteica , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
3.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 32(4): 502-5, 2015 Aug.
Artículo en Chino | MEDLINE | ID: mdl-26252094

RESUMEN

OBJECTIVE: To investigate the clinical features and AGL gene mutations in a family with glycogen storage disease type IIIa (GSD IIIa). METHODS: Clinical data for diagnosis, treatment and follow-up of a sick child with GSD III was collected and analyzed. Genomic DNA was extracted from the peripheral blood samples from the patient and his parents. Polymerase chain reaction and direct DNA sequencing were utilized to analyze all of the exons of the AGL gene. RESULTS: The genotype of the child was found to be c.3710_3711delTA/IVS14+1G>T. The former was a maternally-inherited mutation, which has not been reported previously. The latter was an abnormal splice-site mutation inherited from the father. CONCLUSION: Based on its clinical and molecular evidences, the patient was diagnosed as GSD IIIa in conjunction with retrobular optic neuritis.


Asunto(s)
Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Adulto , Pueblo Asiatico/genética , Secuencia de Bases , Preescolar , China , Femenino , Sistema de la Enzima Desramificadora del Glucógeno/metabolismo , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Mutación Puntual
4.
Mol Genet Metab ; 111(4): 467-76, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24613482

RESUMEN

Glycogen storage disease type IIIa (GSD IIIa) is caused by a deficiency of the glycogen debranching enzyme (GDE), which is encoded by the Agl gene. GDE deficiency leads to the pathogenic accumulation of phosphorylase limit dextrin (PLD), an abnormal glycogen, in the liver, heart, and skeletal muscle. To further investigate the pathological mechanisms behind this disease and develop novel therapies to treat this disease, we generated a GDE-deficient mouse model by removing exons after exon 5 in the Agl gene. GDE reduction was confirmed by western blot and enzymatic activity assay. Histology revealed massive glycogen accumulation in the liver, muscle, and heart of the homozygous affected mice. Interestingly, we did not find any differences in the general appearance, growth rate, and life span between the wild-type, heterozygous, and homozygous affected mice with ad libitum feeding, except reduced motor activity after 50 weeks of age, and muscle weakness in both the forelimb and hind legs of homozygous affected mice by using the grip strength test at 62 weeks of age. However, repeated fasting resulted in decreased survival of the knockout mice. Hepatomegaly and progressive liver fibrosis were also found in the homozygous affected mice. Blood chemistry revealed that alanine transaminase (ALT), aspartate transaminase (AST) and alkaline phosphatase (ALP) activities were significantly higher in the homozygous affected mice than in both wild-type and heterozygous mice and the activity of these enzymes further increased with fasting. Creatine phosphokinase (CPK) activity was normal in young and adult homozygous affected mice. However, the activity was significantly elevated after fasting. Hypoglycemia appeared only at a young age (3 weeks) and hyperlipidemia was not observed in our model. In conclusion, with the exception of normal lipidemia, these mice recapitulate human GSD IIIa; moreover, we found that repeated fasting was detrimental to these mice. This mouse model will be useful for future investigation regarding the pathophysiology and treatment strategy of human GSD III.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo III/patología , Animales , Ayuno/sangre , Femenino , Glucógeno/metabolismo , Sistema de la Enzima Desramificadora del Glucógeno/deficiencia , Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/sangre , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/fisiopatología , Humanos , Immunoblotting , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Fuerza Muscular , Especificidad de Órganos
5.
Ann Biol Clin (Paris) ; 70(6): 648-50, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23207808

RESUMEN

Glycogen storage disease type III (GSD III) is an autosomal recessive disorder caused by the deficiency of glycogen debranching enzyme (AGL). It is characterized by hepatomegaly, progressive myopathy, cardiomyopathy and fasting hypoglycemia. Several mutations in AGL gene have been described in different populations. The W1327X mutation was reported in one Tunisian patient resident in Italy. We looked in this report to determine the frequency of W1327X mutation among Tunisian patients. The W1327X mutation was screening in 26 GSD III patients originated from various geographic locations in Tunisia. The sequence analysis revealed that among nine patients carried the W1327X mutation. Eight of them were from six unrelated families and they were originated from Mahdia (centre of Tunisia) suggesting the existence of a founder effect in this region. Taking into account historical migratory waves, screening for this mutation should be performed in priority for molecular diagnosis confirmation of GSD III in North African populations.


Asunto(s)
Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Mutación , Cardiomiopatías/genética , Femenino , Efecto Fundador , Genotipo , Sistema de la Enzima Desramificadora del Glucógeno/deficiencia , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/epidemiología , Hepatomegalia/genética , Humanos , Masculino , Enfermedades Musculares/genética , Fenotipo , Triptófano , Túnez/epidemiología
7.
J Inherit Metab Dis ; 33(Suppl 2): S289-94, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20532819

RESUMEN

Repeated evaluation of biotinidase (BTD) activity was carried out for a long-term follow-up in patients with hepatic glycogen storage diseases (GSDs). The results indicated inter-intra variability among the GSD-Ia, GSD-III and GSD-IX patients. In addition, a c.1330G>C transversion in the BTD gene, resulting in a p.Asp444His substitution was detected in one allele of a GSD-Ia patient with sustained normal enzyme activity. Thus far, it is necessary to be cautious in the interpretation of the results of BTD activity as a presumptive GSD diagnostic element. It is not known why plasma BTD activity increases in GSDs patients, or the clinical importance of the increment. When viewed from a global perspective, there are some lines of biotin biology that could indicate a relationship between BTD´s behavior and GSDs.


Asunto(s)
Biotinidasa/sangre , Enfermedad del Almacenamiento de Glucógeno/enzimología , Hígado/enzimología , Argentina , Biomarcadores/sangre , Biotinidasa/genética , Estudios de Casos y Controles , Análisis Mutacional de ADN , Genotipo , Enfermedad del Almacenamiento de Glucógeno/sangre , Enfermedad del Almacenamiento de Glucógeno/diagnóstico , Enfermedad del Almacenamiento de Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Humanos , Mutación , Fenotipo , Regulación hacia Arriba
8.
J Inherit Metab Dis ; 33 Suppl 3: S215-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20490926

RESUMEN

Glycogen storage disease type III (GSD III) results from mutations of the AGL gene encoding the glycogen debrancher enzyme. The disease has clinical and biochemical heterogeneity reflecting the severity of the AGL mutations. We sought to characterise the molecular defects in our cohort of Irish patients with GSD III. Fifteen patients from eight unrelated Irish families were identified: six males and nine females. The age ranged from 2-39 years old, and all presented in the first 3 years of life. Four patients (of three families) had mild disease with hepatomegaly, mild hypoglycaemia and normal creatine kinase (CK) levels. Five families had more severe disease, with liver and skeletal muscle involvement and elevated CK. Eleven different mutations were identified amongst the eight families. Of the 11, six were novel: p.T512fs, p.S736fs, p.A1400fs, p.K1407fs, p.Y519X and p.D627Y. The family homozygous for p.A1400fs had the most severe phenotype (early-onset hypoglycaemia, massive hepatomegaly, myopathy and hypertrophic cardiomyopathy before age 2 years), which was not halted by aggressive carbohydrate and protein supplementation. Conversely, the only missense mutation identified in the cohort, p.D627Y, was associated with a mild phenotype. The phenotypic diversity in our GSD III cohort is mirrored by the allelic heterogeneity. We describe two novel null mutations in exon 32 in two families with severe GSD III resistant to current treatment modalities. Knowledge of the specific mutations segregating in this cohort may allow for the development of new therapeutic interventions.


Asunto(s)
Sistema de la Enzima Desramificadora del Glucógeno/deficiencia , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Progresión de la Enfermedad , Femenino , Efecto Fundador , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo III/epidemiología , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/terapia , Herencia , Heterocigoto , Homocigoto , Humanos , Lactante , Recién Nacido , Irlanda/epidemiología , Masculino , Mutación , Linaje , Fenotipo , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Adulto Joven
9.
Zhonghua Er Ke Za Zhi ; 47(8): 608-12, 2009 Aug.
Artículo en Chino | MEDLINE | ID: mdl-19951495

RESUMEN

OBJECTIVE: Glycogen storage disease type III (GSD III) is an autosomal recessive disease caused by glycogen debranching enzyme (GDE) gene (AGL gene) mutation resulting in hepatomegaly, hypoglycemia, short stature and hyperlipidemia. GSD IIIA, involves both liver and muscle, and accounts for up to 80% of GSD III. The definitive diagnosis depends on either mutation analysis or liver and muscle glycogen debranching enzyme activity tests. This study aimed to establish enzymologic diagnostic method for GSD IIIA firstly in China by detecting muscular GDE activity, glycogen content and structure and to determine the normal range of muscular GDE activity, glycogen content and structure in Chinese children. METHOD: Muscle samples were collected from normal controls (male 15, female 20; 12-78 years old), molecularly confirmed GSD III A patients (male 8, female 4, 2-27 years old) and other myopathy patients (male 9, 2-19 years old). Glycogen in the muscle homogenate was degraded into glucose by amyloglucosidase and phosphorylase respectively. The glycogen content and structure were identified by glucose yield determination. The debranching enzyme activity was determined using limit dextrin as substrate. Independent samples Kruskal-Wallis H test, Nemenyi-Wilcoxson-Wilcox test, and Chi-square test were used for statistical analyses by SPSS 11.5. RESULT: (1) GSD III A patients' glycogen content were higher, but G1P/G ratio and GDE activity were lower than those of the other two groups (P < 0.01). In all of the three parameters, there were no significant difference between normal controls and other myopathy patients. (2) The range of normal values: glycogen content 0.31%-0.43%, G1P/G ratio 22.37%- 26.43%, GDE activity 0.234-0.284 micromol/(g. min). (3) Enzymologic diagnostic method had a power similar to that of gene analysis in diagnosis of GSD-IIIA patients. The sensitivity and specificity of enzymologic diagnostic method and mutation detection were 91.7% and 100% respectively. CONCLUSION: Enzymologic diagnostic method of GSD IIIA was firstly established in China. The range of normal values was determined. This method could be used in diagnosing suspected GSD IIIA patients in the clinic.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo III/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Glucógeno/análisis , Músculos/química , Adolescente , Adulto , Anciano , Biopsia , Estudios de Casos y Controles , Niño , Preescolar , China , Femenino , Sistema de la Enzima Desramificadora del Glucógeno/análisis , Enfermedad del Almacenamiento de Glucógeno Tipo III/patología , Humanos , Masculino , Persona de Mediana Edad , Músculos/patología , Adulto Joven
10.
J Hum Genet ; 54(11): 681-6, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19834502

RESUMEN

Glycogen storage disease type III (GSD III) is an autosomal recessive disorder caused by deficiency in the glycogen debranching enzyme (gene symbol: AGL) with two enzyme activities: transferase and glucosidase. A missense mutation causing isolated glucosidase deficiency has never been reported. In this study, we examined 23 patients of Turkish ancestry and identified a novel missense mutation p.R1147G with isolated glucosidase deficiency, along with nine AGL mutations: six nonsense mutations (p.W373X, p.R595X, p.Q667X, p.Q1205X, p.W1327X and p.Q1376X), one deletion (c.1019delA) and two splicing mutation (c.293+2T>G and c.958+1G>A). As p.R1147G impaired glucosidase activity, but maintained transferase activity in vitro, a 12-year-old girl homozygous for p.R1147G was diagnosed with having isolated glucosidase deficiency. Of nine other mutations, p.W1327X and c.1019delA were recurrent, whereas seven mutations were novel. Six patients with p.W1327X were all from two nearby cities on the East Black Sea and shared the same AGL haplotype, indicating a founder effect in Turkish patients. Patients with the same mutations had identical haplotypes. Our results provide the first comprehensive overview of clinical and molecular features of Turkish GSD III patients and the first description of the missense mutation associated with isolated glucosidase deficiency.


Asunto(s)
Glucosidasas/genética , Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Mutación , Adolescente , Adulto , Secuencia de Aminoácidos , Niño , Preescolar , Codón sin Sentido , Análisis Mutacional de ADN , Femenino , Efecto Fundador , Geografía , Glucosidasas/deficiencia , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Haplotipos , Humanos , Lactante , Masculino , Mutación Missense , Polimorfismo de Nucleótido Simple , Sitios de Empalme de ARN/genética , Eliminación de Secuencia , Turquía , Adulto Joven
11.
Hum Mol Genet ; 18(11): 2045-52, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19299494

RESUMEN

Glycogen storage disease type III (GSDIII) is a metabolic disorder characterized by a deficiency in the glycogen debranching enzyme, amylo-1,6-glucosidase,4-alpha-glucanotransferase (AGL). Patients with GSDIII commonly exhibit hypoglycemia, along with variable organ dysfunction of the liver, muscle or heart tissues. The AGL protein binds to glycogen through its C-terminal region, and possesses two separate domains for the transferase and glucosidase activities. Most causative mutations are nonsense, and how they affect the enzyme is not well understood. Here we investigated four rare missense mutations to determine the molecular basis of how they affect AGL function leading to GSDIII. The L620P mutant primarily abolishes transferase activity while the R1147G variant impairs glucosidase function. Interestingly, mutations in the carbohydrate-binding domain (CBD; G1448R and Y1445ins) are more severe in nature, leading to significant loss of all enzymatic activities and carbohydrate binding ability, as well as enhancing targeting for proteasomal degradation. This region (Y1445-G1448R) displays virtual identity across human and bacterial species, suggesting an important role that has been conserved throughout evolution. Our results clearly indicate that inactivation of either enzymatic activity is sufficient to cause GSDIII disease and suggest that the CBD of AGL plays a major role to coordinate its functions and regulation by the ubiquitin-proteasome system.


Asunto(s)
Sistema de la Enzima Desramificadora del Glucógeno/genética , Sistema de la Enzima Desramificadora del Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Mutación Missense , Secuencia de Aminoácidos , Animales , Línea Celular , Chlorocebus aethiops , Sistema de la Enzima Desramificadora del Glucógeno/química , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Humanos , Datos de Secuencia Molecular , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia
12.
Am J Med Genet A ; 146A(22): 2911-5, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18924225

RESUMEN

We report on clinicopathological and whole body MRI analyses of the index patient of a large nonconsanguineous German-Ukraine family with homozygous and heterozygous AGL gene mutations at position p.W1327X (c.3980G > A). There are only limited reports on this phenotype with a homozygous genotype. The index patient, a 49-year-old woman presented with hepatomegaly, cardiomyopathy and moderate progressive proximal limb myopathy. Skeletal muscle showed severe vacuolar myopathy with storage of PAS-positive non-membrane-limited glycogen. An increase in glycogen content and completely decrease of debranching enzyme activity was measured in erythrocytes. Mutational analysis of the AGL gene showed a homozygous p.W1327X mutation. In the family, two brothers had been affected by severe infantile onset hepatomegaly and died within their first years of life by fatal liver cirrhosis. Furthermore, another sister severely affected by hepatomegaly, cardiomyopathy and proximal skeletal myopathy died at age 33. Three younger heterozygous sisters and a brother noticed exercise-induced myalgia and weakness since their teens. In sum, a homozygous p.W1327X mutation leads to a severe generalized glycogenosis types 3a and 3b within the same family. Even heterozygous p.W1327X mutation carriers may present with mild non-progressive neuromuscular symptoms, such as exercise-induced myalgia and fatigue.


Asunto(s)
Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Adulto , Anciano , Sustitución de Aminoácidos , Niño , Preescolar , Femenino , Sistema de la Enzima Desramificadora del Glucógeno/deficiencia , Enfermedad del Almacenamiento de Glucógeno Tipo III/patología , Homocigoto , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Mutación Puntual
13.
Clin Chem Lab Med ; 46(9): 1245-9, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18785866

RESUMEN

BACKGROUND: Glycogen storage disease type III (GSD-III) is an inborn error of glycogen metabolism caused by a deficiency of the glycogen debranching enzyme, amylo-1,6-glucosidase,4-alpha-glucanotransferase (AGL). Here, we describe two unrelated Korean patients with GSD-III and review their clinical and laboratory findings. METHODS: The patients were 18- and 11-month-old girls. They presented with hepatosplenomegaly, developmental delay and hypotonia. The routine laboratory findings showed an elevated serum aspartate aminotransferase, alanine aminotransferase, creatine kinase and triglyceride levels. The blood lactate and uric acid levels were within normal limits. PCR and direct sequencing were performed to determine genetic findings. RESULTS: Glycogen quantitation was markedly increased and AGL activity was undetectable in both patients. Sequence analysis of the AGL gene showed that both patients were compound heterozygotes for c.853C>T (p.R285X) and c.1735+1G>T in one patient, and c.2894_2896delGGAinsTG and c.4090G>C (p.D1364H) in the other patient. The c.2894_2896delGGAinsTG and c.4090G>C (p.D1364H) mutation was a novel finding. CONCLUSIONS: GSD-III should be ruled out when a patient presents with hepatic abnormalities, hypoglycemia, myopathy and hyperlipidemia. This is the first report of confirmation of GSD-III in Korean patients by biochemical and genetic findings.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo III/metabolismo , Glucógeno/metabolismo , Femenino , Sistema de la Enzima Desramificadora del Glucógeno/genética , Sistema de la Enzima Desramificadora del Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/patología , Humanos , Lactante , Corea (Geográfico) , Mutación
14.
Genes Dev ; 21(19): 2399-409, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17908927

RESUMEN

Cori's disease is a glycogen storage disorder characterized by a deficiency in the glycogen debranching enzyme, amylo-1,6-glucosidase,4-alpha-glucanotransferase (AGL). Here, we demonstrate that the G1448R genetic variant of AGL is unable to bind to glycogen and displays decreased stability that is rescued by proteasomal inhibition. AGL G1448R is more highly ubiquitinated than its wild-type counterpart and forms aggresomes upon proteasome impairment. Furthermore, the E3 ubiquitin ligase Malin interacts with and promotes the ubiquitination of AGL. Malin is known to be mutated in Lafora disease, an autosomal recessive disorder clinically characterized by the accumulation of polyglucosan bodies resembling poorly branched glycogen. Transfection studies in HepG2 cells demonstrate that AGL is cytoplasmic whereas Malin is predominately nuclear. However, after depletion of glycogen stores for 4 h, approximately 90% of transfected cells exhibit partial nuclear staining for AGL. Furthermore, stimulation of cells with agents that elevate cAMP increases Malin levels and Malin/AGL complex formation. Refeeding mice for 2 h after an overnight fast causes a reduction in hepatic AGL levels by 48%. Taken together, these results indicate that binding to glycogen crucially regulates the stability of AGL and, further, that its ubiquitination may play an important role in the pathophysiology of both Lafora and Cori's disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Sistema de la Enzima Desramificadora del Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo III/etiología , Enfermedad del Almacenamiento de Glucógeno/etiología , Glucógeno/metabolismo , Enfermedad de Lafora/etiología , Animales , Sitios de Unión , Proteínas Portadoras/genética , Línea Celular , Glucanos/metabolismo , Sistema de la Enzima Desramificadora del Glucógeno/análisis , Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Humanos , Enfermedad de Lafora/enzimología , Ratones , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas
15.
Acta Myol ; 26(1): 72-4, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17915576

RESUMEN

Deficiency of debrancher enzyme causes Glycogen Storage Disease (GSD) type III, an autosomal recessive disorder, characterized by tissue accumulation of abnormally structured glycogen. This report reviews current clinical and molecular knowledge about this disorder and describes the variability at phenotype and genotype levels of a large group of Italian GSDIII patients.


Asunto(s)
Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/fisiopatología , Dieta , Femenino , Genotipo , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/terapia , Humanos , Italia , Masculino , Fenotipo
16.
J Hum Genet ; 50(10): 538-542, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16189622

RESUMEN

Glycogen storage disease type IIIa (GSD IIIa) is an autosomal recessive disorder characterized by excessive accumulation of abnormal glycogen in the liver and muscles and caused by a deficiency in the glycogen debranching enzyme. The spectrum of AGL mutations in GSD IIIa patients depends on ethnic group-prevalent mutations have been reported in the North African Jewish population and in an isolate such as the Faroe islands, because of the founder effect, whereas heterogeneous mutations are responsible for the pathogenesis in Japanese patients. To shed light on molecular characteristics in Egypt, where high rate of consanguinity and large family size increase the frequency of recessive genetic diseases, we have examined three unrelated patients from the same area in Egypt. We identified three different individual AGL mutations; of these, two are novel deletions [4-bp deletion (750-753delAGAC) and 1-bp deletion (2673delT)] and one the nonsense mutation (W1327X) previously reported. All are predicted to lead to premature termination, which completely abolishes enzyme activity. Three consanguineous patients are homozygotes for their individual mutations. Haplotype analysis of mutant AGL alleles showed that each mutation was located on a different haplotype. Our results indicate the allelic heterogeneity of the AGL mutation in Egypt. This is the first report of AGL mutations in the Egyptian population.


Asunto(s)
Alelos , Codón sin Sentido , Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Eliminación de Secuencia , Análisis Mutacional de ADN , Egipto , Femenino , Frecuencia de los Genes , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Haplotipos , Humanos , Masculino
17.
Clin Chim Acta ; 335(1-2): 21-6, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12927680

RESUMEN

BACKGROUND: Type 3 glycogen storage disease is an inborn error of metabolism in young infants that often requires extensive workup. However, this disease manifests with few symptoms other than hepatosplenomegaly. At adolescence, this disease may cause myopathy and cardiomyopathy. Since a significant portion of referrals to pediatrics is for evaluation of a hepatosplenomegaly, the differential diagnosis of this disease assumes importance. METHODS: The clinical and biochemical findings in 26 patients with the type 3 glycogen storage disease were investigated. Biochemical parameters included ALT, AST, total CK and CK-MB. RESULTS: Changes in ALT, AST and total CK were observed to varying degrees. However, CK was found to be a diagnostic indicator for type 3 glycogen storage disease and appears to be a pathognomic marker. CONCLUSIONS: Use of CK may reduce the need for extensive diagnostic profiles and aid in the rapid identification and initiation of management for patients presenting with hepatosplenomegaly.


Asunto(s)
Creatina Quinasa/análisis , Enfermedad del Almacenamiento de Glucógeno Tipo III/diagnóstico , Adolescente , Adulto , Alanina Transaminasa/análisis , Aspartato Aminotransferasas/análisis , Cardiomiopatías/enzimología , Niño , Preescolar , Diagnóstico Diferencial , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/patología , Humanos , Hígado/enzimología , Masculino
18.
Hum Mutat ; 20(6): 480, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12442284

RESUMEN

Deficiency of amylo-1,6-glucosidase, 4-alpha-glucanotransferase enzyme (AGL or glycogen debranching enzyme) is causative of Glycogen Storage Disease type III, a rare autosomal recessive disorder of glycogen metabolism. The disease has been demonstrated to show clinical and biochemical heterogeneity, reflecting the genotype-phenotype heterogeneity among different subjects. The aim of this study was the molecular characterisation of eight unrelated patients from an ethnically heterogeneous population (six Italians, one from India and another one from Tunisia). We describe six novel mutations responsible for the disease (C234R, R675W, 2547delG, T38A, W1327X, IVS6 +3 A>G) and the presence in two Italian subjects of a splice variant (IVS21(+1) G>A) already described elsewhere. This last one is confirmed to be the most frequent mutation among the Italian patients come to our observation, accounting for 28% of 21 patients. One subject was found to be a compound heterozygous. Our data confirm the substantial genetic heterogeneity of this disease. Consequently, the strategy of mutation finding based on screening of recurrent common mutations is limited, as far as regards Italian GSD III patients, to check for the presence of IVS21(+1) G>A.


Asunto(s)
Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Adolescente , Adulto , Niño , Preescolar , ADN/química , ADN/genética , Análisis Mutacional de ADN , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Humanos , Masculino , Mutación
19.
Am J Med Genet ; 109(3): 183-90, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11977176

RESUMEN

Deficiency of amylo-1,6-glucosidase, 4-alpha-glucanotransferase enzyme (AGL or glycogen debrancher enzyme) is responsible for glycogen storage disease type III, a rare autosomal recessive disorder of glycogen metabolism. The AGL gene is located on chromosome 1p21, and contains 35 exons translated in a monomeric protein product. The disease has recognized clinical and biochemical heterogeneity, reflecting the genotype-phenotype heterogeneity among different subjects. The clinical manifestations of GSD III are represented by hepatomegaly, hypoglycemia, hyperlipidemia, short stature and, in a number of subjects, cardiomyopathy and myopathy. In this article, we discuss the genotypic-phenotypic heterogeneity of GSD III by the molecular characterization of mutations responsible for the disease on a collection of 18 independent alleles from the Mediterranean area. We identified by heteroduplex band shift, DNA direct sequencing, and restriction analysis, seven novel mutations (four nonsense point-mutations: R34X, S530X, R1218X, W1398X; two microinsertions: 1072insT and 4724insAA; and one bp deletion: 676DeltaG), together with two new cases carrying a IVS21 + 1 G --> A splicing site mutation previously described in Italian patients. Altogether, 15 alleles were characterized. The correlation between type of mutation and clinical severity was studied in six patients in whom both mutated alleles were detected. Our data confirm the extreme genetic heterogeneity of this disease, thus precluding a strategy of mutation finding based on screening of recurrent common mutations.


Asunto(s)
Sistema de la Enzima Desramificadora del Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/genética , Adolescente , Adulto , Anciano , Secuencia de Bases , Niño , Preescolar , ADN/química , ADN/genética , Análisis Mutacional de ADN , Femenino , Variación Genética , Enfermedad del Almacenamiento de Glucógeno Tipo III/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo III/patología , Humanos , Masculino , Región Mediterránea , Persona de Mediana Edad , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...