Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Orphanet J Rare Dis ; 11(1): 65, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27189384

RESUMO

BACKGROUND: As little information is available on children with non-classic presentations of Pompe disease, we wished to gain knowledge of specific clinical characteristics and genotypes. We included all patients younger than 18 years, who had been evaluated at the Pompe Center in Rotterdam, the Netherlands, between 1975 and 2012, excluding those with the classic-infantile form. None were treated with enzyme replacement therapy at the time of evaluation. We collected information on first symptoms, diagnosis, use of a wheelchair and/or respirator, and enzyme and mutation analysis and assessed muscle strength, pulmonary function, and cardiac parameters. RESULTS: Thirty-one patients participated. Median age at symptom onset was 2.6 years (range 0.5-13y) and at diagnosis 4.0 years. Most first problems were delayed motor development and problems related to limb-girdle weakness. Fatigue, persistent diarrhea and problems in raising the head in supine position were other first complaints. Ten patients were asymptomatic at time of diagnosis. Five of them developed symptoms before inclusion in this study. Over 50 % of all patients had low or absent reflexes, a myopathic face, and scoliosis; 29 % were underweight. Muscle strength of the neck flexors, hip extensors, hip flexors, and shoulder abductors were most frequently reduced. Pulmonary function was decreased in over 48 % of the patients; 2 patients had cardiac hypertrophy. Patients with mutations other than the c.-32-13T > G were overall more severely affected, while 18 out of the 21 patients (86 %) with the c.-32-13T > G/'null' genotype were male. CONCLUSIONS: Our study shows that Pompe disease can present with severe mobility and respiratory problems during childhood. Pompe disease should be considered in the differential diagnosis of children with less familiar signs such as disproportional weakness of the neck flexors, unexplained fatigue, persistent diarrhea and unexplained high CK/ASAT/ALAT. Disease presentation appears to be different from adult patients. The majority of affected children with GAA genotype c.-32-13T > G/'null' appeared to be male.


Assuntos
Genótipo , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/patologia , Adolescente , Criança , Pré-Escolar , Estudos Transversais , Diagnóstico Diferencial , Feminino , Humanos , Lactente , Masculino , Atividade Motora , Mutação , alfa-Glucosidases/genética , alfa-Glucosidases/metabolismo
2.
Gene ; 537(1): 41-5, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24384324

RESUMO

Pompe disease is a clinically and genetically heterogeneous autosomal recessive disorder caused by lysosomal acid α-glucosidase (GAA) deficiency. We report on two affected members of a non-consanguineous Caucasian family, including a classical infantile-onset patient with severe cardiomyopathy (IO) and his paternal grandmother with the adult-onset (AO) form. Two compound heterozygous sequence variants of the GAA gene were identified in each patient by mutation analyses (IO=c.1211A>G and c.1798C>T; AO=c.1211A>G and c.692+5G>T). For this study, the biochemical phenotype resulting from the missense mutation c.1211A>G in exon 8, which converts a highly conserved aspartate to glycine (p.Asp404Gly), was of specific interest because it had not been reported previously. Western blotting revealed a robust expression of all GAA isoforms in quadriceps muscle of both patients (fully CRIM positive), while enzymatic activity was 3.6% (IO) and 6.6% (AO) of normal controls. To further validate these findings, the c.1211A>G sequence variant was introduced in wild type GAA cDNA and over-expressed in HEK293T cells. Site-directed mutagenesis analyses confirmed that the mutation does not affect processing or expression of GAA protein, but rather impairs enzyme function. Similar results were reported for c.1798C>T (p.Arg600Cys), which further supports the biochemical phenotype observed in IO. The third mutation (c.692+5G>T, in intron 3) was predicted to affect normal splicing of the GAA mRNA, and qPCR indeed verified a 4-fold lower mRNA expression in AO. It is concluded that the novel sequence variant c.1211A>G results in full CRIM but significantly lower GAA activity, which in combination with c.1798C>T leads to infantile-onset Pompe disease. We surmise that the difference in disease severity between the two family members in this study is due to a milder effect of the intronic mutation c.692+5G>T (vs. c.1798C>T) on phenotype, partially preserving GAA activity and delaying onset in the proband (paternal grandmother).


Assuntos
Glucana 1,4-alfa-Glucosidase/genética , Glucana 1,4-alfa-Glucosidase/metabolismo , Doença de Depósito de Glicogênio Tipo II/genética , Mutação de Sentido Incorreto , Idade de Início , Estudos de Casos e Controles , Feminino , Doença de Depósito de Glicogênio Tipo II/enzimologia , Doença de Depósito de Glicogênio Tipo II/epidemiologia , Células HEK293 , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Fenótipo , Gravidez , Músculo Quadríceps/enzimologia , Splicing de RNA , Valores de Referência
3.
J Inherit Metab Dis ; 32(3): 416-23, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19387865

RESUMO

Enzyme analysis for Pompe disease in leukocytes has been greatly improved by the introduction of acarbose, a powerful inhibitor of interfering alpha-glucosidases, which are present in granulocytes but not in lymphocytes. Here we show that the application of acarbose in the enzymatic assay employing the artificial substrate 4-methylumbelliferyl-alpha-D: -glucoside (MU-alphaGlc) is insufficient to clearly distinguish patients from healthy individuals in all cases. Also, the ratios of the activities without/with acarbose only marginally discriminated Pompe patients and healthy individuals. By contrast, when the natural substrate glycogen is used, the activity in leukocytes from patients (n = 82) with Pompe disease is at most 17% of the lowest control value. The use of artificial substrate in an assay with isolated lymphocytes instead of total leukocytes is a poor alternative as blood samples older than one day invariably yield lymphocyte preparations that are contaminated with granulocytes. To diagnose Pompe disease in leukocytes we recommend the use of glycogen as substrate in the presence of acarbose. This assay unequivocally excludes Pompe disease. To also exclude pseudo-deficiency of acid alpha-glucosidase caused by the sequence change c.271G>A (p.D91N or GAA2; homozygosity in approximately 1:1000 caucasians), a second assay employing MU-alphaGlc substrate plus acarbose or DNA analysis is required.


Assuntos
Acarbose/metabolismo , Glucana 1,4-alfa-Glucosidase/análise , Doença de Depósito de Glicogênio Tipo II/diagnóstico , Doença de Depósito de Glicogênio Tipo II/enzimologia , Glicogênio/metabolismo , Leucócitos/enzimologia , Acarbose/farmacologia , Técnicas de Diagnóstico Neurológico , Glucana 1,4-alfa-Glucosidase/antagonistas & inibidores , Glucana 1,4-alfa-Glucosidase/metabolismo , Glicogênio/farmacologia , Doença de Depósito de Glicogênio Tipo II/sangue , Doença de Depósito de Glicogênio Tipo II/patologia , Humanos , Recém-Nascido , Leucócitos/patologia , Especificidade por Substrato
4.
J Neurol Sci ; 275(1-2): 46-50, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18757064

RESUMO

BACKGROUND AND OBJECTIVE: Pompe disease is an inherited metabolic disorder caused by deficiency of acid alpha-glucosidase. All affected neonates have a severe hypertrophic cardiomyopathy, leading to cardiac failure and death within the first year of life. We investigated the presence and extent of cardiac involvement in children and adults with Pompe disease with the common c.-32-13T>G genotype to determine the usefulness of cardiac screening in these patients with relatively 'milder' phenotypes. METHODS: Cardiac dimensions and function were evaluated through echocardiography, electrocardiography and Holter monitoring. The total group comprised 68 patients with Pompe disease, of whom 22 patients had disease onset before the age of 18. RESULTS: Two patients (3%) had cardiac abnormalities possibly related to Pompe disease: Electrocardiography showed a Wolff-Parkinson-White pattern in an 8-year-old girl, and one severely affected adult patient had a mild hypertrophic cardiomyopathy. This hypertrophy did not change during treatment with recombinant human alpha-glucosidase. In addition, four adult patients showed minor cardiac abnormalities which did not exceed the prevalence in the general population and were attributed to advanced age, hypertension or pre-existing cardiac pathology unrelated to Pompe disease. CONCLUSIONS: Cardiac involvement is rare in Pompe patients with the common c.-32-13T>G genotype. The younger patients were not more frequently affected than the adults. Electrocardiographic evaluation appears to be appropriate as initial screening tool. Extensive cardiac screening seems indicated only if the electrocardiogram is abnormal or the patient has a history of cardiac disease.


Assuntos
Glucana 1,4-alfa-Glucosidase/genética , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/fisiopatologia , Cardiopatias/etiologia , Mutação/genética , Adulto , Fatores Etários , Idoso , Criança , Eletrocardiografia/métodos , Saúde da Família , Feminino , Genótipo , Cardiopatias/genética , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Ultrassonografia/métodos
5.
Neurology ; 68(2): 110-5, 2007 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-17210890

RESUMO

BACKGROUND: Pompe disease (acid maltase deficiency, glycogen storage disease type II; OMIM 232300) is an autosomal recessive lysosomal storage disorder characterized by acid alpha-glucosidase deficiency due to mutations in the GAA gene. Progressive skeletal muscle weakness affects motor and respiratory functions and is typical for all forms of Pompe disease. Cardiac hypertrophy is an additional fatal symptom in the classic infantile subtype. c.-32-13T-->G is the most common mutation in adults. OBJECTIVE: To delineate the disease variation among patients with this mutation and to define the c.-32-13T-->G haplotypes in search for genotype-phenotype correlations. METHODS: We studied 98 compound heterozygotes with a fully deleterious mutation (11 novel mutations are described) and the common c.-32-13T-->G mutation. RESULTS: All patients were Caucasian. None had the classic infantile form of Pompe disease. The clinical course varied far more than anticipated (age at diagnosis <1 to 78 years; age at onset: <1 to 52 years). The acid alpha-glucosidase activities in a subset of patients ranged from 4 to 19.9 nmol/mg/h. Twelve different c.-32-13T-->G haplotypes were identified based on 17 single-nucleotide polymorphisms located in the GAA gene. In 76% of the cases, c.-32-13T-->G was encountered in the second most common GAA core haplotype (DHRGEVVT). In only one case was c.-32-13T-->G encountered in the major GAA core haplotype (DRHGEIVT). CONCLUSION: Patients with the same c.-32-13T-->G haplotype (c.q. GAA genotype) may manifest first symptoms at different ages, indicating that secondary factors may substantially influence the clinical course of patients with this mutation.


Assuntos
Predisposição Genética para Doença/genética , Doença de Depósito de Glicogênio Tipo II/epidemiologia , Doença de Depósito de Glicogênio Tipo II/genética , Haplótipos/genética , Medição de Risco/métodos , alfa-Glucosidases/genética , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Estudos de Coortes , Análise Mutacional de DNA , Feminino , Doença de Depósito de Glicogênio Tipo II/enzimologia , Humanos , Lactente , Recém-Nascido , Internacionalidade , Masculino , Pessoa de Meia-Idade , Mutação , Prevalência
7.
Hum Mol Genet ; 8(12): 2145-53, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10545593

RESUMO

Pompe's disease or glycogen storage disease type II (GSDII) belongs to the family of inherited lysosomal storage diseases. The underlying deficiency of acid alpha-glucosidase leads in different degrees of severity to glycogen storage in heart, skeletal and smooth muscle. There is currently no treatment for this fatal disease, but the applicability of enzyme replacement therapy is under investigation. For this purpose, recombinant human acid alpha-glucosidase has been produced on an industrial scale in the milk of transgenic rabbits. In this paper we demonstrate the therapeutic effect of this enzyme in our knockout mouse model of GSDII. Full correction of acid alpha-glucosidase deficiency was obtained in all tissues except brain after a single dose of i.v. enzyme administration. Weekly enzyme infusions over a period of 6 months resulted in degradation of lysosomal glycogen in heart, skeletal and smooth muscle. The tissue morphology improved substantially despite the advanced state of disease at the start of treatment. The results have led to the start of a Phase II clinical trial of enzyme replacement therapy in patients.


Assuntos
Glucana 1,4-alfa-Glucosidase/metabolismo , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Leite/enzimologia , Animais , Animais Geneticamente Modificados , Glucana 1,4-alfa-Glucosidase/genética , Glucana 1,4-alfa-Glucosidase/uso terapêutico , Doença de Depósito de Glicogênio Tipo II/enzimologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Miocárdio/ultraestrutura , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , alfa-Glucosidases
8.
Eur J Hum Genet ; 7(6): 713-6, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10482961

RESUMO

Glycogen storage disease type II (GSD H) is an autosomal recessive myopathy. Early and late-onset phenotypes are distinguished - infantile, juvenile and adult. Three mutations in the acid alpha-glucosidase gene are common in the Dutch patient population: IVS1(-13T-->G), 525delT and delexon18. 63% of Dutch GSD II patients carry one or two of these mutations, and the genotype-phenotype correlation is known. To determine the frequency of GSD II, we have screened an unselected sample of neonates for the occurrence of these three mutations. Based on the calculated carrier frequencies, the predicted frequency of the disease is 1 in 40000 divided by 1 in 138 000 for infantile GSD II and 1 in 57 000 for adult GSD II. This is about two to four times higher than previously suggested, which is a reason to become more familiar with the presentation of GSD II in its different clinical forms and to adjust the risk assessment for genetic counselling.


Assuntos
Doença de Depósito de Glicogênio Tipo II/diagnóstico , Doença de Depósito de Glicogênio Tipo II/epidemiologia , Doença de Depósito de Glicogênio Tipo II/genética , Alelos , Análise Mutacional de DNA , Éxons , Deleção de Genes , Aconselhamento Genético , Testes Genéticos , Genótipo , Heterozigoto , Humanos , Recém-Nascido , Países Baixos/epidemiologia , Penetrância , Fenótipo , Mutação Puntual , Prevalência , Fatores de Risco
9.
Community Genet ; 2(2-3): 91-6, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-15181339

RESUMO

OBJECTIVES: To compare the overall birth prevalence of diagnosed glycogen storage disease type II (GSD II) with the predicted frequency based on mutation screening, in order to determine whether GSD II is an underdiagnosed condition, and to analyze which medical disciplines recognize GSD II. METHODS: Retrospective data on all enzymatic diagnoses of GSD II were collected from diagnostic labs throughout the Netherlands, covering the period from January 1, 1972 to December 31, 1996. Age-specific diagnostic incidence rates were calculated for the entire study period. By adding together the diagnostic incidences for all age groups, we calculated the birth prevalence of diagnosed GSD II and compared these figures with the predicted frequency based on mutation screening in a random sample from the general population. The medical specialization of the referring clinicians was also recorded. RESULTS: GSD II was diagnosed in 154 individuals, including 11 prenatal diagnoses. The birth prevalences of the various phenotypes were 1/101,000 (infantile form), 1/720,000 (juvenile form) and 1/53,000 (adult form). The birth prevalence of the adult and infantile phenotype together was 1/35,000. Eighty-two percent of the patients were diagnosed in university hospitals. Of the patients with infantile GSD II, 71% were diagnosed by a pediatrician, whereas most patients with adult GSD II were diagnosed by a neurologist (80%). CONCLUSIONS: There is no evidence for the underdiagnosis of GSD II in the Netherlands, as the calculated birth prevalences of the disease are consistent with previous predictions based on mutation screening in a random sample of newborns. The worldwide birth prevalence of the disease may well be higher than 1 in 100,000. GSD II is mainly diagnosed in university hospitals.

10.
Hum Mol Genet ; 7(11): 1815-24, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9736785

RESUMO

Glycogen storage disease type II (GSDII) is caused by lysosomal acid alpha-glucosidase deficiency. Patients have a rapidly fatal or slowly progressive impairment of muscle function. Enzyme replacement therapy is under investigation. For large-scale, cost-effective production of recombinant human acid alpha-glucosidase in the milk of transgenic animals, we have fused the human acid alpha-glucosidase gene to 6.3 kb of the bovine alphaS1-casein gene promoter and have tested the performance of this transgene in mice. The highest production level reached was 2 mg/ml. The major fraction of the purified recombinant enzyme has a molecular mass of 110 kDa and resembles the natural acid alpha-glucosidase precursor from human urine and the recombinant precursor secreted by CHO cells, with respect to pH optimum, Km, Vmax, N-terminal amino acid sequence and glycosylation pattern. The therapeutic potential of the recombinant enzyme produced in milk is demonstrated in vitro and in vivo. The precursor is taken up in a mannose 6-phosphate receptor-dependent manner by cultured fibroblasts, is converted to mature enzyme of 76 kDa and depletes the glycogen deposit in fibroblasts of patients. When injected intravenously, the milk enzyme corrects the acid alpha-glucosidase deficiency in heart and skeletal muscle of GSDII knockout mice.


Assuntos
Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Leite/enzimologia , Proteínas Recombinantes/genética , alfa-Glucosidases/genética , alfa-Glucosidases/metabolismo , Animais , Células CHO , Bovinos , Cricetinae , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transgenes , alfa-Glucosidases/deficiência
11.
Clin Genet ; 53(5): 379-82, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9660056

RESUMO

In nine Dutch patients with the infantile form of glycogen storage disease type II (GSDII), who were compound heterozygous for either 525delT or exon18del (1), sequence analysis was performed to search for the mutations in the second lysosomal alpha-glucosidase allele. One patient had a novel TG deletion at cDNA position 379 + 380. Surprisingly five of the nine patients had the same two base pair changes: A921 --> T and G925 --> A. The first change is a well-known polymorphism but the second one is a novel mutation and results in the substitution of Gly309 by Arg. By screening 43 other GSDII patients the same mutation was found in two other cases, one from The Netherlands and one from France. To verify its deleterious effect, the mutation was introduced in the wild type lysosomal alpha-glucosidase cDNA and expressed in COS cells.


Assuntos
Doença de Depósito de Glicogênio Tipo II/genética , Alelos , Substituição de Aminoácidos , Animais , Células COS/citologia , Células COS/enzimologia , Células COS/metabolismo , Células Cultivadas , DNA/análise , DNA/genética , Análise Mutacional de DNA , Frequência do Gene , Genes/genética , Genótipo , Doença de Depósito de Glicogênio Tipo II/enzimologia , Heterozigoto , Homozigoto , Humanos , Lisossomos/enzimologia , Mutagênese Sítio-Dirigida , Fenótipo , Mutação Puntual/genética , Deleção de Sequência , alfa-Glucosidases/genética
12.
Hum Mutat ; 11(3): 209-15, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9521422

RESUMO

Glycogen Storage Disease type II (GSDII) is caused by the deficiency of lysosomal alpha-glucosidase (acid maltase). This paper reports on the characterization of the molecular defects in 6 infantile patients from Turkish ancestry. Five of the 6 patients had reduced levels of the lysosomal alpha-glucosidase precursor. Conversion to mature enzyme was impaired in all cases, and the lysosomal alpha-glucosidase activity in all patients fibroblasts was less than 0.5% of control. DNA sequence analysis revealed 3 new mutations. One mutation, found in 3 patients in homozygous form, was a double insertion in exon 19 (2471AG-->CAGG) leading to a frameshift after Pro 913. It is the first insertion mutation described in the lysosomal alpha-glucosidase gene. Two patients were homozygous for missense mutations leading to the substitution of Ser to Pro at amino acid 566 (S566P) in one case and of Pro to Arg at amino acid 768 (P768R) in the other. One patient was found to have a Gly to Arg missense mutation at amino acid 643 (G643R), previously identified in an adult patient (Hermans et al., 1993), combined with a silent second allele. The latter 3 mutations were introduced in the wild type lysosomal alpha-glucosidase cDNA and expressed in COS cells to analyze their effect. Precursor species of 110 kD were formed but the maturation was impaired. As a result there was an overall deficiency of catalytic activity, which is in accordance with the findings in the patients fibroblasts and with the clinical phenotype.


Assuntos
Glucana 1,4-alfa-Glucosidase/genética , Doença de Depósito de Glicogênio Tipo II/enzimologia , Doença de Depósito de Glicogênio Tipo II/genética , Mutação/genética , Animais , Células COS , Células Cultivadas , Consanguinidade , Análise Mutacional de DNA , Fibroblastos/enzimologia , Glucana 1,4-alfa-Glucosidase/química , Humanos , Lactente , Recém-Nascido , Mutagênese Sítio-Dirigida , Países Baixos/epidemiologia , Polimorfismo Conformacional de Fita Simples , Pele/citologia , Turquia/etnologia , alfa-Glucosidases
13.
Hum Mol Genet ; 7(1): 53-62, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9384603

RESUMO

Glycogen storage disease type II (GSDII; Pompe disease), caused by inherited deficiency of acid alpha-glucosidase, is a lysosomal disorder affecting heart and skeletal muscles. A mouse model of this disease was obtained by targeted disruption of the murine acid alpha-glucosidase gene (Gaa) in embryonic stem cells. Homozygous knockout mice (Gaa -/-) lack Gaa mRNA and have a virtually complete acid alpha-glucosidase deficiency. Glycogen-containing lysosomes are detected soon after birth in liver, heart and skeletal muscle cells. By 13 weeks of age, large focal deposits of glycogen have formed. Vacuolar spaces stain positive for acid phosphatase as a sign of lysosomal pathology. Both male and female knockout mice are fertile and can be intercrossed to produce progeny. The first born knockout mice are at present 9 months old. Overt clinical symptoms are still absent, but the heart is typically enlarged and the electrocardiogram is abnormal. The mouse model will help greatly to understand the pathogenic mechanism of GSDII and is a valuable instrument to explore the efficacy of different therapeutic interventions.


Assuntos
Cardiomegalia/metabolismo , Doença de Depósito de Glicogênio Tipo II/metabolismo , Doença de Depósito de Glicogênio Tipo II/fisiopatologia , Glicogênio/metabolismo , alfa-Glucosidases/deficiência , Animais , Cardiomegalia/genética , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Feminino , Doença de Depósito de Glicogênio Tipo II/genética , Masculino , Camundongos , Camundongos Knockout , alfa-Glucosidases/genética
14.
J Inherit Metab Dis ; 20(4): 556-8, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9266392

RESUMO

A novel mutation, C118T, in exon 2 of the acid alpha-glucosidase gene has been found in an infant with glycogen storage disease type II. This mutation is predicted to result in protein truncation. The phenotype was that of the severe infantile form of the disorder with lack of motor development, but with eye regard, social smile and vocalization. The parents were heterozygous for C118T and belong to an Islamic community opposed to termination of pregnancy. As the C118T mutation results in the loss of one of two AvaI sites present in an informative PCR product, reliable premarriage carrier detection became possible and was acceptable to the members of this extended family.


Assuntos
Doença de Depósito de Glicogênio Tipo II/genética , alfa-Glucosidases/genética , Broncopneumonia/etiologia , DNA/análise , Análise Mutacional de DNA , Primers do DNA , Ecocardiografia , Evolução Fatal , Feminino , Doença de Depósito de Glicogênio Tipo II/psicologia , Heterozigoto , Humanos , Lactente , Masculino , Dados de Sequência Molecular , Mutação , Paquistão , Gravidez
15.
J Biol Chem ; 272(18): 11816-23, 1997 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-9115239

RESUMO

In mammalian tissues two types of cGMP-dependent protein kinase (cGK) have been identified. In contrast to the dimeric cGK I, cGK II purified from pig intestine was shown previously to behave as a monomer. However, recombinant rat cGK II was found to have hydrodynamic parameters indicative of a homodimer. Chemical cross-linking studies showed that pig cGK II in intestinal membranes has a dimeric structure as well. However, after purification, cGK II was found to be partly proteolyzed into C-terminal monomeric fragments. Phosphorylation studies in rat intestinal brush borders revealed that the potency of cGMP analogs to stimulate or inhibit native cGK II in vitro (i.e. 8-(4-chlorophenylthio)-cGMP > cGMP > beta-phenyl-1,N2-etheno-8-bromo-cGMP > beta-phenyl-1,N2-etheno-cGMP and Rp-8-(4-chlorophenylthio)-cGMPs > Rp-beta-phenyl-1, N2-etheno-8-bromo-cGMPs, respectively) correlated well with their potency to stimulate or inhibit cGK II-mediated Cl- secretion across intestinal epithelium but differed strikingly from their potency to affect cGK I activity. These data show that the N terminus of cGK II is involved in dimerization and that endogenous cGK II displays a distinct activation/inhibition profile with respect to cGMP analogs, which permits a pharmacological dissection between cGK II- and cGK I-mediated physiological processes.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/química , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Isoenzimas/química , Animais , Centrifugação com Gradiente de Concentração , Cromatografia em Gel , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/isolamento & purificação , Dimerização , Inibidores Enzimáticos/farmacologia , Mucosa Intestinal/enzimologia , Isoenzimas/isolamento & purificação , Isoenzimas/metabolismo , Jejuno , Cinética , Masculino , Mamíferos , Microvilosidades/enzimologia , Peso Molecular , Ratos , Ratos Wistar , Suínos
16.
Hum Mutat ; 9(1): 17-22, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-8990003

RESUMO

Mutation analysis was performed in a nonconsanguineous Dutch caucasian family with a grandfather presenting the first symptoms of glycogen storage disease type II (acid alpha-glucosidase deficiency) in the sixth decade of life and a grandchild with onset of symptoms shortly after birth. The grandfather was identified as compound heterozygote having the IVS1(-13T-->G)/delta T525 combination of mutant acid alpha-glucosidase alleles, the affected third generation offspring as homozygote delta T525/delta T525. The disease phenotypes in this family are in accordance with the genotypes since the IVS1(-13T-->G) mutation reduces acid alpha-glucosidase synthesis by 60 to 80%, whereas the delta T525 mutation completely prohibits the formation of catalytically active enzyme. Four additional families were identified with patients homozygote for delta T525 and five others with an equally deleterious delta T525/delta exon 18 genotype. The nine latter patients had typically the infantile form of glycogen storage disease type II. The genotype-phenotype correlation is irrefutable.


Assuntos
Doença de Depósito de Glicogênio Tipo II/patologia , Feminino , Genótipo , Doença de Depósito de Glicogênio Tipo II/etnologia , Doença de Depósito de Glicogênio Tipo II/genética , Heterozigoto , Homozigoto , Humanos , Masculino , Países Baixos , Linhagem
17.
Biochem Biophys Res Commun ; 241(2): 414-8, 1997 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-9425285

RESUMO

A new method is described for detection of mutations in the lysosomal alpha-glucosidase gene (GAA) leading to Glycogen Storage Disease type II (GSDII). A key feature of the method is isolation and reverse transcription of mRNA followed by PCR amplification of lysosomal alpha-glucosidase cDNA with M13-extended primers. Dye labeled primers are used for cycle sequencing and an ABI PRISM 377 DNA sequencing system for analysis. The method is rapid and complementary to the automated sequencing of all the 19, PCR amplified, coding exons of the GAA gene. The advantages and pitfalls of this new method are discussed in the light of the results obtained with an infantile GSDII patient. A new splice site mutation in the GAA gene of this patient was identified, IVS16(+2T-->C), resulting in the deletion of 16 base pairs of exon 16.


Assuntos
Doença de Depósito de Glicogênio Tipo II/genética , Lisossomos/enzimologia , Mutação , alfa-Glucosidases/genética , DNA Complementar/genética , Éxons , Testes Genéticos/métodos , Humanos , Lactente , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Splicing de RNA , RNA Mensageiro/genética , Análise de Sequência de DNA/métodos
18.
Biochim Biophys Acta ; 1308(2): 93-6, 1996 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-8764823

RESUMO

Enzyme replacement therapy is at present the option of choice for treatment of lysosomal storage diseases. To explore the feasibility of lysosomal enzyme production in milk of transgenic animals, the human acid alpha-glucosidase cDNA was placed under control of the alpha S1-casein promoter and expressed in mice. The milk contained recombinant enzyme at a concentration up to 1.5 micrograms/ml. Enzyme purified from milk of transgenic mice was internalized via the mannose 6-phosphate receptor and corrected enzyme deficiency in fibroblasts from patients. We conclude that transgenically produced human acid alpha-glucosidase meets the criteria for therapeutic application.


Assuntos
Glucana 1,4-alfa-Glucosidase/biossíntese , Doença de Depósito de Glicogênio Tipo II/metabolismo , Leite/enzimologia , Proteínas Recombinantes/biossíntese , Animais , Células Cultivadas , DNA Complementar/genética , Fibroblastos/citologia , Glucana 1,4-alfa-Glucosidase/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , alfa-Glucosidases
19.
J Med Genet ; 33(6): 458-64, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8782044

RESUMO

Up to now eight patients with alpha-NAGA deficiency have been described. This includes the newly identified patient reported here who died unexpectedly aged 1 1/2 years of hypoxia during convulsions; necropsy was not performed. Three patients have been genotyped previously and here we report the mutations in the other five patients, including two new mutations (S160C and E193X). The newly identified patient is consanguineous with the first patients reported with alpha-NAGA deficiency and neuroaxonal dystrophy and they all had the alpha-NAGA genotype E325K/E325K. Clinical heterogeneity among patients with alpha-NAGA deficiency is extreme. Two affected sibs, homozygotes for E325K, are severely affected and have the signs and symptoms of infantile neuroaxonal dystrophy, but prominent vacuolisation is lacking. The mildly affected patients (two families, three patients) at the opposite end of the clinical spectrum have clear vacuolisation and angiokeratoma but no overt neurological manifestations. Two of them are homozygous for the stop mutation E193X, leading to complete loss of alpha-NAGA protein. These observations are difficult to reconcile with a simple genotype-phenotype correlation and we suggest that factors or genes other than alpha-NAGA contribute to the clinical heterogeneity of the eight patients with alpha-NAGA deficiency. At the metabolic level, the patients with alpha-NAGA deficiency are similar. The major abnormal urinary oligosaccharides are sialylglycopeptides of the O linked type. Our enzymatic studies indicated that these compounds are not the primary lysosomal storage products.


Assuntos
Hexosaminidases/deficiência , Hexosaminidases/genética , Pele/enzimologia , Animais , Células Cultivadas , Criança , Pré-Escolar , Feminino , Genótipo , Humanos , Lactente , Masculino , Mutação , Linhagem , Fenótipo , Coelhos , Pele/citologia , alfa-N-Acetilgalactosaminidase
20.
Clin Genet ; 49(6): 325-8, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8884087

RESUMO

We describe two unrelated Dutch patients with typical symptoms of infantile glycogen storage disease type II (GSD II) and virtual absence of acid alpha-glucosidase activity in leukocytes and cultured skin fibroblasts. The patients were identified as homozygotes for a deletion of exon 18 of the acid alpha-glucosidase gene (GAA). The in-frame deletion manifests at the protein level in a characteristic way: the enzyme precursor is smaller than normal and degraded in the endoplasmic reticulum or Golgi complex. These case present an evident example of a genotype-phenotype correlation in glycogen storage disease type II.


Assuntos
Deleção Cromossômica , Precursores Enzimáticos/metabolismo , Éxons , Doença de Depósito de Glicogênio Tipo II/genética , Homozigoto , Lisossomos/enzimologia , alfa-Glucosidases/deficiência , Humanos , Recém-Nascido , Masculino , Linhagem , alfa-Glucosidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...