Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
N Engl J Med ; 370(6): 533-42, 2014 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-24499211

RESUMO

BACKGROUND: Congenital disorders of glycosylation are genetic syndromes that result in impaired glycoprotein production. We evaluated patients who had a novel recessive disorder of glycosylation, with a range of clinical manifestations that included hepatopathy, bifid uvula, malignant hyperthermia, hypogonadotropic hypogonadism, growth retardation, hypoglycemia, myopathy, dilated cardiomyopathy, and cardiac arrest. METHODS: Homozygosity mapping followed by whole-exome sequencing was used to identify a mutation in the gene for phosphoglucomutase 1 (PGM1) in two siblings. Sequencing identified additional mutations in 15 other families. Phosphoglucomutase 1 enzyme activity was assayed on cell extracts. Analyses of glycosylation efficiency and quantitative studies of sugar metabolites were performed. Galactose supplementation in fibroblast cultures and dietary supplementation in the patients were studied to determine the effect on glycosylation. RESULTS: Phosphoglucomutase 1 enzyme activity was markedly diminished in all patients. Mass spectrometry of transferrin showed a loss of complete N-glycans and the presence of truncated glycans lacking galactose. Fibroblasts supplemented with galactose showed restoration of protein glycosylation and no evidence of glycogen accumulation. Dietary supplementation with galactose in six patients resulted in changes suggestive of clinical improvement. A new screening test showed good discrimination between patients and controls. CONCLUSIONS: Phosphoglucomutase 1 deficiency, previously identified as a glycogenosis, is also a congenital disorder of glycosylation. Supplementation with galactose leads to biochemical improvement in indexes of glycosylation in cells and patients, and supplementation with complex carbohydrates stabilizes blood glucose. A new screening test has been developed but has not yet been validated. (Funded by the Netherlands Organization for Scientific Research and others.).


Assuntos
Glucofosfatos/genética , Doença de Depósito de Glicogênio/genética , Fenótipo , Fosfoglucomutase/genética , Galactose/uso terapêutico , Genes Recessivos , Glucose/metabolismo , Glucofosfatos/metabolismo , Doença de Depósito de Glicogênio/dietoterapia , Doença de Depósito de Glicogênio/metabolismo , Glicoproteínas/biossíntese , Glicosilação , Humanos , Masculino , Mutação , Fosfoglucomutase/metabolismo , RNA Mensageiro/análise
2.
Neuropediatrics ; 45(6): 346-53, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25111564

RESUMO

Leigh syndrome (MIM 25600), also known as infantile subacute necrotizing encephalomyelopathy, is a neurodegenerative disorder with characteristic bilateral symmetric lesions in basal ganglia and subcortical brain regions. It is commonly associated with systemic cytochrome c oxidase (COX) deficiency and mutations in the SURF1 gene (MIM 185620), encoding a putative assembly or maintenance factor of COX. The clinical course is dominated by neurodevelopmental regression, brain stem, and basal ganglia involvement (e.g., dystonia, apnea) with death often occurring before the age of 10 years. Herein, we present three sisters carrying a previously reported homozygous SURF1 mutation (c.868_869insT) that is predicted to result in a truncated protein with loss of function. Our patients show heterogeneous clinical findings with different distribution patterns of metabolic lesions in brain magnetic resonance imaging (MRI) as well as a Chiari malformation with hydrocephalus in one patient. However, all three siblings show an unusual long survival (12 years and>16 years). COX activity was not detectable in one patient and strongly reduced in the other two. We discuss these findings with respect to a review of the literature. A total of 15 additional patients with survival>14 years have been reported so far. Overall, no clear genotype-phenotype correlations are detectable among these patients.


Assuntos
Doença de Leigh/genética , Doença de Leigh/mortalidade , Doença de Leigh/patologia , Encéfalo/patologia , Criança , Feminino , Humanos , Proteínas de Membrana/genética , Proteínas Mitocondriais/genética , Mutação , Linhagem , Irmãos
3.
J Hum Genet ; 54(11): 681-6, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19834502

RESUMO

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.


Assuntos
Glucosidases/genética , Sistema da Enzima Desramificadora do Glicogênio/genética , Doença de Depósito de Glicogênio Tipo III/genética , Mutação , Adolescente , Adulto , Sequência de Aminoácidos , Criança , Pré-Escolar , Códon sem Sentido , Análise Mutacional de DNA , Feminino , Efeito Fundador , Geografia , Glucosidases/deficiência , Doença de Depósito de Glicogênio Tipo III/enzimologia , Haplótipos , Humanos , Lactente , Masculino , Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Sítios de Splice de RNA/genética , Deleção de Sequência , Turquia , Adulto Jovem
4.
Clin Chem Lab Med ; 47(10): 1233-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19754354

RESUMO

BACKGROUND: Glycogen storage disease type III (GSD III) is caused by mutations in AGL which encodes for a single protein with two enzyme activities: oligo-1, 4-1, 4-glucantransferase (transferase) and amylo-1, 6-glucosidase. Activity of both enzymes is lost in most patients with GSD III, but in the very rare subtype IIId, transferase activity is deficient. Since the spectrum of AGL mutations is dependent on the ethnic group, we investigated the clinical and molecular characteristics in Egyptian patients with GSD III. METHODS: Clinical features were examined in five Egyptian patients. AGL was sequenced and AGL haplotypes were determined. RESULTS: Six novel AGL mutations were identified: a large deletion (c.3481-3588+1417del1525 bp), two insertions (c.1389insG and c.2368insA), two small deletions (c.2223-2224delGT and c.4041delT), and a missense mutation (p.L620P). p.L620P was found in a patient with IIId. Each mutation was located on a different AGL haplotype. CONCLUSIONS: Our results suggest that there is allelic and phenotypic heterogeneity of GSD III in Egypt. This is the second description of a large deletion in AGL. p.L620P is the second mutation found in GSD IIId.


Assuntos
População Negra/genética , Sistema da Enzima Desramificadora do Glicogênio/genética , Doença de Depósito de Glicogênio Tipo III/genética , Mutação de Sentido Incorreto , Deleção de Sequência , Sequência de Aminoácidos , Animais , Sequência de Bases , Estudos de Casos e Controles , Criança , Pré-Escolar , Sequência Consenso , Análise Mutacional de DNA , Egito , Sistema da Enzima Desramificadora do Glicogênio/química , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Humanos , Masculino , Dados de Sequência Molecular , Transferases/metabolismo
5.
Mol Genet Metab ; 95(1-2): 52-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18691923

RESUMO

We describe a 15-year-old boy and his 19-year-old sister with progressive dilated cardiomyopathy and mild non-progressive proximal lower limb myopathy, secondary to the accumulation of amylopectin-like fibrillar glycogen, (polyglucosan) bodies, in heart and skeletal muscle. Evidence of idiopathic amylopectinosis or polysaccharidosis was demonstrated in heart and skeletal muscle tissue by histology, electron microscopy, biochemical, and genetic analysis. In both siblings the heart muscle stored PAS-positive, proteinase-k resistant and partly diastase resistant granulo-filamentous material, simulating polyglucosan bodies. Glycogen branching enzyme activity, and phosphofructokinase enzyme activity, measured in skeletal muscle tissue and explanted heart tissue were all within the normal limits, however glycogen content was elevated. Furthermore, GBE1, PRKAG2, desmin, alphabeta-crystallin, ZASP, myotilin, and LAMP-2 gene sequencing revealed no mutation, excluding e.g. glycogen storage disease type 4 and desmin-related myofibrillar cardiomyopathies. In both patients the diagnosis of an idiopathic polysaccharidosis with progressive dilated cardiomyopathy was made, requiring heart transplantation at age 13 and 14, respectively. Both patients belong to an autosomal recessive group of biochemically and genetically unclassified severe vacuolar glycogen storage disease of the heart and skeletal muscle. Up to now unidentified glycogen synthesis or glycogen degradation pathways are supposed to contribute to this idiopathic glycogen storage disease.


Assuntos
Doença de Depósito de Glicogênio Tipo IV/diagnóstico , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Adolescente , Adulto , Feminino , Alemanha , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo IV/enzimologia , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Doença de Depósito de Glicogênio Tipo IV/patologia , Humanos , Masculino , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Miocárdio/enzimologia , Miocárdio/patologia , Linhagem , Irmãos
6.
Neuromuscul Disord ; 17(9-10): 698-706, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17643989

RESUMO

To describe the clinical and neurophysiological spectrum and prognosis in a large cohort of biochemically and genetically proven late onset Pompe patients. Thirty-eight diagnosed with late onset Pompe disease at our neuromuscular department during 1985 and 2006 are described in detail. The mean delay from onset of symptoms or first medical consultation until diagnosis was 10.4 and 7.1 years, respectively. A different diagnosis was suggested in 11 of 38 patients. Ten patients underwent repeated muscle biopsies before diagnosis of Pompe disease was established. Limb girdle weakness was the most frequent presenting sign. Six patients complained of myalgia. Wolf-Parkinson-White syndrome was found in 3 of 38 patients. Respiratory failure preceded the onset of overt limb muscle weakness in three patients. The course of the patients was progressive in all, but there was a wide variety of progression, which did not correlate with the age of disease onset. In 71% of the patients, neurophysiological investigations revealed a myopathic EMG pattern, half of the patients had spontaneous activity including complex repetitive discharges. A normal EMG was found in 9% of the patients. Nerve conduction studies were normal in all. Pompe disease should be taken into consideration in patients with unexplained limb girdle muscular weakness with respiratory failure. Cardiac manifestations may not be restricted to infantile Pompe disease.


Assuntos
Doença de Depósito de Glicogênio Tipo II/diagnóstico , Doença de Depósito de Glicogênio Tipo II/fisiopatologia , Adolescente , Adulto , Idade de Início , Criança , Pré-Escolar , Progressão da Doença , Eletromiografia , Feminino , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Debilidade Muscular , Distrofia Muscular do Cíngulo dos Membros , Transtornos Respiratórios/etiologia , Estudos Retrospectivos
8.
Clin Chim Acta ; 439: 162-7, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25451950

RESUMO

BACKGROUND: Glycogen storage disease type III (GSD III; MIM #232400) is an autosomal recessive inherited disorder characterized by fasting hypoglycemia, growth retardation, hepatomegaly, progressive myopathy, and cardiomyopathy. GSD III is caused by deficiency in the glycogen debranching enzyme (gene symbol: AGL). Molecular analyses of AGL have indicated heterogeneity depending on ethnic groups. In Turkey we reported 13 different AGL mutations from GSD III patients in the Eastern region; however, the full spectrum of AGL mutations in Turkish population remains unclear. Here we investigated 12 GSD III patients mostly from Western Turkey. METHODS: The full coding exons, their relevant exon-intron boundaries, and the 5'- and 3'-flanking regions of the patients' AGL were sequenced. AGL haplotypes were determined. Splicing mutations were characterized by RNA transcript analysis. RESULTS: Twelve different mutations were identified: 7 novel AGL mutations [69-base pair deletion (c.1056_1082+42del69), 21-base par deletion (c.3940_3949+11del21), two small duplications (c.364_365dupCT and c.1497_1500dupAGAG), and 3 splicing mutations (c.1736-11A>G, c.3259+1G>A and c.3588+2T>G)], along with 5 known mutations (c.1019delA, c.958+1G>A, c.4161+5G>A, p.R864X and p.R1218X). Transcripts of splicing mutations (c.1736-11A>G, c.3588+2T>G and c.4161+5G>A) were shown to cause aberrant splicing. AGL haplotype analyses suggested that c.1019delA and c.958+1G>A are founder mutations in Turkish patients, while p.R864X is a recurrent mutation. CONCLUSIONS: Our study broadens the spectrum of AGL mutations and demonstrates that mutations in Western Turkey are different from those in the Eastern region.


Assuntos
Doença de Depósito de Glicogênio Tipo III/genética , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Mutação , Análise de Sequência de DNA , Turquia , Adulto Jovem
9.
Cornea ; 22(2): 175-7, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12605057

RESUMO

BACKGROUND: Fabry disease is an X-linked genetic disorder involving sphingolipid catabolism, which is caused by lysosomal alpha-galactosidase A deficiency. Ophthalmological findings such as corneal and lens opacities and conjunctival and retinal vessel abnormalities can be the only and/or the first recognizable symptoms, especially in heterozygous females. METHODS: We report on a 34-year-old German woman with cornea verticillata. The alpha-galactosidase A activity was determined in leukocytes using a fluorescence substrate, and the sequence analysis of the alpha galactosidase A gene was performed with genomic DNA. RESULTS: The alpha-galactosidase A activity in leukocytes was significantly low (0.24 nmol/min/mg protein; normal range, 0.4-1.2), which is compatible with a heterozygote for Fabry disease. The following sequence analysis revealed a heterozygous transition in position IVS5 + 2 T > C. Transition of thymine (T) to cytosine (C) affects the donor splice motive of exon 5 and most probably leads to an aberrant splicing procedure of the alpha-galactosidase A gene. CONCLUSION: Our case emphasizes the importance of ophthalmological findings in Fabry disease. The subsequent biochemical and molecular analysis provides a secure diagnosis of female carriers of Fabry disease.


Assuntos
Opacidade da Córnea/enzimologia , Opacidade da Córnea/genética , Doença de Fabry/enzimologia , Doença de Fabry/genética , Mutação , alfa-Galactosidase/genética , Adulto , Análise Mutacional de DNA , Feminino , Heterozigoto , Humanos
11.
J Hum Genet ; 51(11): 958-963, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17047887

RESUMO

Glycogen storage disease type III (GSD III) is an autosomal recessive disorder characterized by excessive accumulation of abnormal glycogen in the liver and/or muscles and caused by deficiency in the glycogen debranching enzyme (AGL). Previous studies have revealed that the spectrum of AGL mutations in GSD III patients depends on ethnic grouping. We investigated nine GSD III patients from Germany, Canada, Afghanistan, Iran, and Turkey and identified six novel AGL mutations: one nonsense (W255X), three deletions (1019delA, 3202-3203delTA, and 1859-1869del11-bp), and two splicing mutations (IVS7 + 5G > A and IVS21 + 5insA), together with three previously reported ones (R864X, W1327X, and IVS21 + 1G > A). All mutations are predicted to lead to premature termination, which abolishes enzyme activity. Our molecular study on GSD III patients of different ethnic ancestry showed allelic heterogeneity of AGL mutations. This is the first AGL mutation report for German, Canadian, Afghan, Iranian and Turkish populations.


Assuntos
Sistema da Enzima Desramificadora do Glicogênio/genética , Doença de Depósito de Glicogênio Tipo III/etnologia , Doença de Depósito de Glicogênio Tipo III/genética , Mutação , Afeganistão , Canadá , Análise Mutacional de DNA , Genótipo , Alemanha , Haplótipos , Humanos , Irã (Geográfico) , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Turquia
12.
Acta Neuropathol ; 110(2): 151-7, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15986226

RESUMO

Three unrelated patients, one girl, one boy, and an adult female, aged 14, 11 and 41 years, respectively, at the time of biopsy, revealed lysosomal glycogen storage, autophagic vacuoles and peculiar globular inclusions of distinct ultrastructure, which were reducing but did not appear like true "reducing bodies" as described in the congenital myopathy "reducing body myopathy". All three patients had residual activity of acid alpha-glucosidase in their muscle biopsy samples. Leukocytes in the girl showed normal acid alpha-glucosidase activity, but in the boy activity was reduced. Molecular genetic analysis of the GAA gene revealed disease-causing mutations in each patient: H568L/R672W, IVS1-13T>G/G615F, and IVS1-13T>G/IVS1-13T>G. Although only one patient with such globular inclusions has been reported up to now, the three patients described here indicate that in the late-onset type of GSD II such inclusions may not be rare.


Assuntos
Doença de Depósito de Glicogênio Tipo II/patologia , Corpos de Inclusão/patologia , Músculo Esquelético/patologia , Adulto , Criança , Feminino , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/metabolismo , Humanos , Corpos de Inclusão/ultraestrutura , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestrutura , Mutação , Reação em Cadeia da Polimerase , alfa-Glucosidases/genética , alfa-Glucosidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA