Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Pathol ; 263(1): 8-21, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38332735

RESUMO

Pompe disease is a lysosomal storage disorder that preferentially affects muscles, and it is caused by GAA mutation coding acid alpha-glucosidase in lysosome and glycophagy deficiency. While the initial pathology of Pompe disease is glycogen accumulation in lysosomes, the special role of the lysosomal pathway in glycogen degradation is not fully understood. Hence, we investigated the characteristics of accumulated glycogen and the mechanism underlying glycophagy disturbance in Pompe disease. Skeletal muscle specimens were obtained from the affected sites of patients and mouse models with Pompe disease. Histological analysis, immunoblot analysis, immunofluorescence assay, and lysosome isolation were utilized to analyze the characteristics of accumulated glycogen. Cell culture, lentiviral infection, and the CRISPR/Cas9 approach were utilized to investigate the regulation of glycophagy accumulation. We demonstrated residual glycogen, which was distinguishable from mature glycogen by exposed glycogenin and more α-amylase resistance, accumulated in the skeletal muscle of Pompe disease. Lysosome isolation revealed glycogen-free glycogenin in wild type mouse lysosomes and variously sized glycogenin in Gaa-/- mouse lysosomes. Our study identified that a defect in the degradation of glycogenin-exposed residual glycogen in lysosomes was the fundamental pathological mechanism of Pompe disease. Meanwhile, glycogenin-exposed residual glycogen was absent in other glycogen storage diseases caused by cytoplasmic glycogenolysis deficiencies. In vitro, the generation of residual glycogen resulted from cytoplasmic glycogenolysis. Notably, the inhibition of glycogen phosphorylase led to a reduction in glycogenin-exposed residual glycogen and glycophagy accumulations in cellular models of Pompe disease. Therefore, the lysosomal hydrolysis pathway played a crucial role in the degradation of residual glycogen into glycogenin, which took place in tandem with cytoplasmic glycogenolysis. These findings may offer a novel substrate reduction therapeutic strategy for Pompe disease. © 2024 The Pathological Society of Great Britain and Ireland.


Assuntos
Doença de Depósito de Glicogênio Tipo II , Glicoproteínas , Humanos , Camundongos , Animais , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/patologia , Doença de Depósito de Glicogênio Tipo II/terapia , Glicogênio/análise , Glicogênio/metabolismo , Glucosiltransferases/metabolismo , Músculo Esquelético/patologia , Lisossomos/metabolismo
2.
Am J Med Genet A ; 194(9): e63643, 2024 09.
Artigo em Inglês | MEDLINE | ID: mdl-38656665

RESUMO

The mitochondrial phosphate carrier is critical for adenosine triphosphate synthesis by serving as the primary means for mitochondrial phosphate import across the inner membrane. Variants in the SLC25A3 gene coding mitochondrial phosphate carrier lead to failure in inorganic phosphate transport across mitochondria. The critical dependence on mitochondria as an energy source is especially evident in tissues with high-energy demands such as the heart, muscle; defects in the mitochondrial energy production machinery underlie a wide range of primary mitochondrial disorders that present with cardiac and muscle diseases. The characteristic clinical picture of a prominent early-onset hypertrophic cardiomyopathy and lactic acidosis may be an indication for analysis of the SLC25A3 gene. Here, described a patient with suspicion of infantile Pompe disease due to involvement of heart and muscle and high-level of plasma creatinine kinase but finally diagnosed mitochondrial phosphate-carrier deficiency.


Assuntos
Doença de Depósito de Glicogênio Tipo II , Mitocôndrias , Proteínas de Transporte de Fosfato , Humanos , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/diagnóstico , Doença de Depósito de Glicogênio Tipo II/patologia , Proteínas de Transporte de Fosfato/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Lactente , Doenças Mitocondriais/genética , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/patologia , Mutação/genética , Diagnóstico Diferencial , Masculino , Feminino , Fosfatos/sangue , Fosfatos/metabolismo , Acidose Láctica/genética , Acidose Láctica/diagnóstico
3.
J Inherit Metab Dis ; 47(4): 716-730, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38584574

RESUMO

The aim of this longitudinal cohort study, is to provide more insight into the pattern of brain abnormalities, and possible consequences for cognitive functioning, in patients with classic infantile Pompe disease. We included 19 classic infantile Pompe patients (median age last assessment 8.9 years, range 1.5-22.5 years; 5/19 CRIM negative), treated with ERT. Using MR imaging of the brain (T1, T2, and FLAIR acquisitions), we classified progression of brain abnormalities on a 12-point rating scale at multiple time points throughout follow-up. Additionally we noted specific white matter patterns and examined atrophy. Cognitive development was studied using Wechsler IQ assessments obtained by certified neuropsychologists. The association between age and cognitive functioning, and MRI ratings and cognitive functioning was assessed by linear regression models. All but one patient developed brain abnormalities. The abnormalities progressed in a similar pattern throughout the brain, with early involvement of periventricular white matter, later followed by subcortical white matter, gray matter structures, and juxtacortical U-fibers. We found a significant decline (p < 0.01), with increasing age for full scale IQ, performance IQ and processing speed, but not for verbal IQ (p = 0.17). Each point increment in the 12-point MRI rating scale was associated with a significant decline (3.1-6.0 points) in all the IQ index scores (p < 0.05). The majority of long-term surviving patients in our cohort develop incremental brain MRI abnormalities and decline in cognitive functioning. This highlights the need for new therapies that can cross the blood-brain barrier in order to treat this CNS phenotype.


Assuntos
Encéfalo , Cognição , Doença de Depósito de Glicogênio Tipo II , Imageamento por Ressonância Magnética , Humanos , Masculino , Doença de Depósito de Glicogênio Tipo II/complicações , Doença de Depósito de Glicogênio Tipo II/patologia , Feminino , Lactente , Encéfalo/patologia , Encéfalo/diagnóstico por imagem , Pré-Escolar , Adolescente , Criança , Estudos Longitudinais , Adulto Jovem , Progressão da Doença , Substância Branca/patologia , Substância Branca/diagnóstico por imagem , Terapia de Reposição de Enzimas , Adulto
4.
Orphanet J Rare Dis ; 19(1): 267, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39010129

RESUMO

BACKGROUND: Late-onset Pompe disease (LOPD) is mainly characterized by progressive limb-girdle muscle weakness and respiratory impairment, whereas stroke and cerebrovascular abnormalities have been insufficiently studied in LOPD. This study aimed to evaluate the frequency and pattern of intracranial artery and brain parenchyma abnormalities in LOPD patients. RESULTS: Neuroimaging data from 30 Chinese adult LOPD patients were collected from our center. Seven patients (7/30) had acute cerebral infarction or hemorrhage. Brain magnetic resonance angiography (MRA) or computed tomography angiography (CTA) revealed artery abnormalities in 23 patients (23/30). Dilative arteriopathy was found in 19 patients (19/30), with vertebrobasilar dolichoectasia found in 17 patients and dilatation of the anterior circulation arteries found in 8 patients. The maximum diameter of the basilar artery was correlated with disease duration (p < 0.05). In addition, aneurysms (7/30) and fenestrations (3/30) were discovered. There were 14 patients with arterial stenosis (14/30), and both anterior and posterior circulation involvement occurred in 9 patients (9/14). Stenosis and dilative arteriopathy simultaneously occurred in 10 patients (10/30). White matter hyperintensities were present in 13 patients (13/28). Microbleeds, predominantly located in the cerebellum and brainstem, were detected in 7 patients (7/22) via susceptibility-weighted imaging. CONCLUSIONS: Intracranial vasculopathy involving both large arteries and small vessels is an important organ damage in LOPD patients. LOPD should be considered a key differential diagnosis in young adults with cryptogenic stroke, and a series of imaging evaluations of the brain and intracranial blood vessels is recommended as a routine workup in adult LOPD patients.


Assuntos
Doença de Depósito de Glicogênio Tipo II , Humanos , Masculino , Feminino , Adulto , Doença de Depósito de Glicogênio Tipo II/patologia , Doença de Depósito de Glicogênio Tipo II/diagnóstico por imagem , Adulto Jovem , Pessoa de Meia-Idade , Angiografia por Ressonância Magnética , Adolescente
5.
Stem Cell Res ; 79: 103459, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38896971

RESUMO

Here we present the generation of HIMRi006-A and HIMRi007-A Pompe disease (PD) patient derived human induced pluripotent stem cell (hiPSC) lines. HIMRi006-A represents an infantile onset disease (IOPD) phenotype caused by a homozygous c.307 T > G mutation in the GAA gene. HIMRi007-A is characterized by heterozygous mutations c.-32-13 T > G/c.1716C > G and is associated with an adult onset of disease symptoms (LOPD). Both lines are generated via lentiviral expression of OCT4, SOX2, KLF4, and c-MYC. The lines display a typical embryonic stem cell morphology, express pluripotency markers, retain a normal karyotype (46, XX/XY) and have the differentiation capacity in all three germ layers. Altogether, both lines provide a resource tool to the community for future in depth molecular studies of PD pathomechanism.


Assuntos
Doença de Depósito de Glicogênio Tipo II , Células-Tronco Pluripotentes Induzidas , Fator 4 Semelhante a Kruppel , Humanos , Doença de Depósito de Glicogênio Tipo II/patologia , Doença de Depósito de Glicogênio Tipo II/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Diferenciação Celular , Linhagem Celular , Masculino , Lactente , Mutação , Feminino
6.
Mol Genet Genomic Med ; 12(7): e2480, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38958145

RESUMO

BACKGROUND: Pompe Disease (PD) is a metabolic myopathy caused by variants in the GAA gene, resulting in deficient enzymatic activity. We aimed to characterize the clinical features and related genetic variants in a series of Mexican patients. METHODS: We performed a retrospective study of clinical records of patients diagnosed with LOPD, IOPD or pseudodeficiency. RESULTS: Twenty-nine patients were included in the study, comprising these three forms. Overall, age of symptom onset was 0.1 to 43 years old. The most frequent variant identified was c.-32-13T>G, which was detected in 14 alleles. Among the 23 different variants identified in the GAA gene, 14 were classified as pathogenic, 5 were likely pathogenic, and 1 was a variant of uncertain significance. Two variants were inherited in cis arrangement and 2 were pseudodeficiency-related benign alleles. We identified two novel variants (c.1615 G>A and c.1076-20_1076-4delAAGTCGGCGTTGGCCTG). CONCLUSION: To the best of our knowledge, this series represent the largest phenotypic and genotypic characterization of patients with PD in Mexico. Patients within our series exhibited a combination of LOPD and IOPD associated variants, which may be related to genetic diversity within Mexican population. Further population-wide studies are required to better characterize the incidence of this disease in Mexican population.


Assuntos
Idade de Início , Doença de Depósito de Glicogênio Tipo II , Mutação , alfa-Glucosidases , Humanos , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/patologia , Masculino , Feminino , Pré-Escolar , Criança , Adulto , alfa-Glucosidases/genética , Lactente , México/epidemiologia , Adolescente , Fenótipo , Estudos Retrospectivos , Estudos de Associação Genética , Alelos , Adulto Jovem
7.
Biomolecules ; 14(5)2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38785980

RESUMO

Autophagy is an evolutionarily conserved lysosome-dependent degradation of cytoplasmic constituents. The system operates as a critical cellular pro-survival mechanism in response to nutrient deprivation and a variety of stress conditions. On top of that, autophagy is involved in maintaining cellular homeostasis through selective elimination of worn-out or damaged proteins and organelles. The autophagic pathway is largely responsible for the delivery of cytosolic glycogen to the lysosome where it is degraded to glucose via acid α-glucosidase. Although the physiological role of lysosomal glycogenolysis is not fully understood, its significance is highlighted by the manifestations of Pompe disease, which is caused by a deficiency of this lysosomal enzyme. Pompe disease is a severe lysosomal glycogen storage disorder that affects skeletal and cardiac muscles most. In this review, we discuss the basics of autophagy and describe its involvement in the pathogenesis of muscle damage in Pompe disease. Finally, we outline how autophagic pathology in the diseased muscles can be used as a tool to fast track the efficacy of therapeutic interventions.


Assuntos
Autofagia , Doença de Depósito de Glicogênio Tipo II , Doença de Depósito de Glicogênio Tipo II/patologia , Doença de Depósito de Glicogênio Tipo II/metabolismo , Humanos , Animais , Glicogênio/metabolismo , Lisossomos/metabolismo , alfa-Glucosidases/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/metabolismo
8.
Mol Metab ; 81: 101899, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38346589

RESUMO

OBJECTIVE: Pompe disease (PD) is caused by deficiency of the lysosomal enzyme acid α-glucosidase (GAA), leading to progressive glycogen accumulation and severe myopathy with progressive muscle weakness. In the Infantile-Onset PD (IOPD), death generally occurs <1 year of age. There is no cure for IOPD. Mouse models of PD do not completely reproduce human IOPD severity. Our main objective was to generate the first IOPD rat model to assess an innovative muscle-directed adeno-associated viral (AAV) vector-mediated gene therapy. METHODS: PD rats were generated by CRISPR/Cas9 technology. The novel highly myotropic bioengineered capsid AAVMYO3 and an optimized muscle-specific promoter in conjunction with a transcriptional cis-regulatory element were used to achieve robust Gaa expression in the entire muscular system. Several metabolic, molecular, histopathological, and functional parameters were measured. RESULTS: PD rats showed early-onset widespread glycogen accumulation, hepato- and cardiomegaly, decreased body and tissue weight, severe impaired muscle function and decreased survival, closely resembling human IOPD. Treatment with AAVMYO3-Gaa vectors resulted in widespread expression of Gaa in muscle throughout the body, normalizing glycogen storage pathology, restoring muscle mass and strength, counteracting cardiomegaly and normalizing survival rate. CONCLUSIONS: This gene therapy holds great potential to treat glycogen metabolism alterations in IOPD. Moreover, the AAV-mediated approach may be exploited for other inherited muscle diseases, which also are limited by the inefficient widespread delivery of therapeutic transgenes throughout the muscular system.


Assuntos
Doença de Depósito de Glicogênio Tipo II , Camundongos , Ratos , Humanos , Animais , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/terapia , Doença de Depósito de Glicogênio Tipo II/patologia , Músculo Esquelético/metabolismo , Glicogênio/metabolismo , Terapia Genética/métodos , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/terapia
9.
J Proteomics ; 291: 105037, 2024 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-38288553

RESUMO

Pompe disease is a lysosomal storage disorder caused by deficiency of acid alpha-glucosidase (GAA), resulting in glycogen accumulation with profound pathology in skeletal muscle. We recently developed an optimized form of lentiviral gene therapy for Pompe disease in which a codon-optimized version of the GAA transgene (LV-GAAco) was fused to an insulin-like growth factor 2 (IGF2) peptide (LV-IGF2.GAAco), to promote cellular uptake via the cation-independent mannose-6-phosphate/IGF2 receptor. Lentiviral gene therapy with LV-IGF2.GAAco showed superior efficacy in heart, skeletal muscle, and brain of Gaa -/- mice compared to gene therapy with untagged LV-GAAco. Here, we used quantitative mass spectrometry using TMT labeling to analyze the muscle proteome and the response to gene therapy in Gaa -/- mice. We found that muscle of Gaa -/- mice displayed altered levels of proteins including those with functions in the CLEAR signaling pathway, autophagy, cytoplasmic glycogen metabolism, calcium homeostasis, redox signaling, mitochondrial function, fatty acid transport, muscle contraction, cytoskeletal organization, phagosome maturation, and inflammation. Gene therapy with LV-GAAco resulted in partial correction of the muscle proteome, while gene therapy with LV-IGF2.GAAco resulted in a near-complete restoration to wild type levels without inducing extra proteomic changes, supporting clinical development of lentiviral gene therapy for Pompe disease. SIGNIFICANCE: Lysosomal glycogen accumulation is the primary cause of Pompe disease, and leads to a cascade of pathological events in cardiac and skeletal muscle and in the central nervous system. In this study, we identified the proteomic changes that are caused by Pompe disease in skeletal muscle of a mouse model. We showed that lentiviral gene therapy with LV-IGF2.GAAco nearly completely corrects disease-associated proteomic changes. This study supports the future clinical development of lentiviral gene therapy with LV-IGF2.GAAco as a new treatment option for Pompe disease.


Assuntos
Doença de Depósito de Glicogênio Tipo II , Animais , Camundongos , Terapia Genética/métodos , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/terapia , Doença de Depósito de Glicogênio Tipo II/patologia , Lentivirus/genética , Lentivirus/metabolismo , Lisossomos/metabolismo , Camundongos Knockout , Músculo Esquelético/metabolismo , Proteoma/metabolismo , Proteômica
10.
Sci Rep ; 13(1): 22822, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-38129558

RESUMO

Quantitative muscle MRI is increasingly important in the non-invasive evaluation of neuromuscular disorders and their progression. Underlying histopathotological alterations, leading to changes in qMRI parameters are incompletely unraveled. Early microstructural differences of unknown origin reflected by Diffusion MRI in non-fat infiltrated muscles were detected in Pompe patients. This study employed a longitudinal approach with a Pompe disease mouse model to investigate the histopathological basis of these changes. Monthly scans of Pompe (Gaa6neo/6neo) and wildtype mice (age 1-8 months) were conducted using diffusion MRI, T2-mapping, and Dixon-based water-fat imaging on a 7 T scanner. Immunofluorescence studies on quadriceps muscles were analyzed for lysosomal accumulations and autophagic buildup and correlated with MRI outcome measures. Fat fraction and water-T2 did not differ between groups and remained stable over time. In Pompe mice, fractional anisotropy increased, while mean diffusivity (MD) and radial diffusivity (RD) decreased in all observed muscles. Autophagic marker and muscle fibre diameter revealed significant negative correlations with reduced RD and MD, while lysosomal marker did not show any change or correlation. Using qMRI, we showed diffusion changes in muscles of presymptomatic Pompe mice without fat-infiltrated muscles and correlated them to autophagic markers and fibre diameter, indicating diffusion MRI reveals autophagic buildup.


Assuntos
Doença de Depósito de Glicogênio Tipo II , Humanos , Camundongos , Animais , Lactente , Doença de Depósito de Glicogênio Tipo II/diagnóstico por imagem , Doença de Depósito de Glicogênio Tipo II/patologia , Fibras Musculares Esqueléticas/patologia , Imagem de Difusão por Ressonância Magnética , Músculo Quadríceps , Modelos Animais de Doenças , Água
11.
Artigo em Chinês | WPRIM | ID: wpr-981813

RESUMO

OBJECTIVE@#To explore the clinical features, lysosomal enzymatic [acid α-glucosidase (GAA)] activities and genetic variants in a child with late-onset Pompe disease (LOPD).@*METHODS@#Clinical data of a child who had presented at the Genetic Counseling Clinic of West China Second University Hospital in August 2020 was retrospectively analyzed. Blood samples were collected from the patient and her parents for the isolation of leukocytes and lymphocytes as well as DNA extraction. The activity of lysosomal enzyme GAA in leukocytes and lymphocytes was analyzed with or without addition of inhibitor of GAA isozyme. Potential variants in genes associated with neuromuscular disorders were analyzed, in addition with conservation of the variant sites and protein structure. The remaining samples from 20 individuals undergoing peripheral blood lymphocyte chromosomal karyotyping were mixed and used as the normal reference for the enzymatic activities.@*RESULTS@#The child, a 9-year-old female, had featured delayed language and motor development from 2 years and 11 months. Physical examination revealed unstable walking, difficulty in going upstairs and obvious scoliosis. Her serum creatine kinase was significantly increased, along with abnormal electromyography, whilst no abnormality was found by cardiac ultrasound. Genetic testing revealed that she has harbored compound heterozygous variants of the GAA gene, namely c.1996dupG (p.A666Gfs*71) (maternal) and c.701C>T (p.T234M) (paternal). Based on the guidelines from the American College of Medical Genetics and Genomics, the c.1996dupG (p.A666Gfs*71) was rated as pathogenic (PVS1+PM2_Supporting+PM3), whilst the c.701C>T (p.T234M) was rated as likely pathogenic (PM1+PM2_Supporting+PM3+PM5+PP3). The GAA in the leukocytes from the patient, her father and mother were respectively 76.1%, 91.3% and 95.6% of the normal value without the inhibitor, and 70.8%, 112.9% and 128.2% of the normal value with the inhibitor, whilst the activity of GAA in their leukocytes had decreased by 6 ~ 9 times after adding the inhibitor. GAA in lymphocytes of the patient, her father and mother were 68.3%, 59.0% and 59.5% of the normal value without the inhibitor, and 41.0%, 89.5% and 57.7% of the normal value with the inhibitor, the activity of GAA in lymphocytes has decreased by 2 ~ 5 times after adding the inhibitor.@*CONCLUSION@#The child was diagnosed with LOPD due to the c.1996dupG and c.701C>T compound heterozygous variants of the GAA gene. The residual activity of GAA among LOPD patients can range widely and the changes may be atypical. The diagnosis of LOPD should not be based solely on the results of enzymatic activity but combined clinical manifestation, genetic testing and measurement of enzymatic activity.


Assuntos
Humanos , Criança , Masculino , Feminino , Doença de Depósito de Glicogênio Tipo II/patologia , Estudos Retrospectivos , alfa-Glucosidases/genética , Mães , Lisossomos/patologia , Mutação
12.
Arq. neuropsiquiatr ; 76(4): 247-251, Apr. 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-888382

RESUMO

ABSTRACT Pompe disease is an inherited disease caused by acid alpha-glucosidase (GAA) deficiency. A single center observational study aimed at assessing the prevalence of late-onset Pompe disease in a high-risk Brazilian population, using the dried blood spot test to detect GAA deficiency as a main screening tool. Dried blood spots were collected for GAA activity assay from 24 patients with "unexplained" limb-girdle muscular weakness without vacuolar myopathy in their muscle biopsy. Samples with reduced enzyme activity were also investigated for GAA gene mutations. Of the 24 patients with dried blood spots, one patient (4.2%) showed low GAA enzyme activity (NaG/AaGIA: 40.42; %INH: 87.22%). In this patient, genetic analysis confirmed two heterozygous mutations in the GAA gene (c.-32-13T>G/p.Arg854Ter). Our data confirm that clinicians should look for late-onset Pompe disease in patients whose clinical manifestation is an "unexplained" limb-girdle weakness even without vacuolar myopathy in muscle biopsy.


RESUMO A doença de Pompe é uma doença hereditária causada pela deficiência da enzima alfa-glicosidase ácida (GAA). Estudo observacional foi realizado, em um único centro, para determinar a prevalência da doença de Pompe de início tardio (LOPD) em uma população brasileira de alto risco, usando teste em gota seca (DBS) como ferramenta principal de triagem para detectar a deficiência da GAA. DBS foi coletado para avaliar a atividade da GAA em 24 pacientes com fraqueza muscular de cinturas "não explicada" sem miopatia vacuolar na biópsia muscular. As amostras com atividade enzimática reduzida foram também submetidas a análise de mutações no gene GAA. Dos 24 pacientes com DBS, baixa atividade da enzima GAA (NaG/AaGIA: 40.42; %INH: 87.22%) foi encontrada em um paciente (4.2%). Nessa paciente, a análise genética confirmou duas mutações em heterozigose composta no gene GAA (c.-32-13T > G/p.Arg854Ter). Nossos resultados confirmam que LOPD deve ser investigada quando a manifestação clínica é uma fraqueza muscular de cinturas "não explicada", mesmo na ausência de miopatia vacuolar na biópsia muscular.


Assuntos
Humanos , Masculino , Feminino , Adulto , Doença de Depósito de Glicogênio Tipo II/diagnóstico , Distrofia Muscular do Cíngulo dos Membros/diagnóstico , Distrofia Muscular do Cíngulo dos Membros/sangue , alfa-Glucosidases/sangue , Biópsia , Doença de Depósito de Glicogênio Tipo II/patologia , Doença de Depósito de Glicogênio Tipo II/sangue , Prevalência , Distrofia Muscular do Cíngulo dos Membros/patologia
13.
Rev. lab. clín ; 9(1): 17-20, ene.-mar. 2016.
Artigo em Espanhol | IBECS (Espanha) | ID: ibc-150651

RESUMO

Introducción. La enfermedad de Pompe, también denominada déficit de maltasa ácida o glucogenosis tipo ii, es un trastorno metabólico autosómico recesivo caracterizado por un acúmulo anormal de glucógeno lisosomal, causado por la deficiencia de la enzima α-glucosidasa ácida (GAA). Según la edad de inicio y el grado de afectación orgánica, la enfermedad de Pompe se suele subdividir en 2 tipos: neonatal (infantile-onset) y tardío (late-onset). El diagnóstico clínico se confirma mediante la ausencia virtual (forma infantil) o marcada reducción (forma del adulto) de la actividad enzimática GAA en diferentes muestras biológicas. Material y métodos. El objetivo de este trabajo consiste en establecer los valores de referencia para la actividad enzimática GAA intralinfocitaria que constituye el método gold standard para el diagnóstico de la enfermedad. Resultados. Los resultados obtenidos en una población de sujetos control difieren de los publicados en otros trabajos, lo que refleja la importancia de establecer valores de referencia dentro de cada laboratorio (AU)


Introduction. Pompe disease also called acid maltase deficiency or glycogenosis type ii is an autosomal recessive metabolic disorder characterized by an abnormal accumulation of lysosomal glycogen caused by deficiency of the enzyme acid α-glucosidase (GAA). According to the age of onset and the extent of organ involvement, Pompe disease is usually divided into 2 subtypes: Neonatal (infantile-onset) and adult (late-onset). The clinical diagnosis is confirmed by the virtual absence (infantile form) or markedly reduced (adult form) GAA enzyme activity in different biological samples. Material and methods. The aim of this work is to establish reference values for intralymphocyte alpha-glucosidase acid activity, which is the 'gold standard' method for the diagnosis of Pompe disease. Results. The results obtained in a population of control subjects are different from those published in other studies. This reflects the importance of establishing reference values within each laboratory (AU)


Assuntos
Humanos , Masculino , Feminino , Adulto , Ativação Enzimática , Ativação Enzimática/fisiologia , Valores de Referência , alfa-Glucosidases/análise , Doença de Depósito de Glicogênio Tipo II/diagnóstico , Doença de Depósito de Glicogênio Tipo II/patologia , Espectrofotometria/métodos , Espectrofotometria/normas , Espectrofotometria , 28599
14.
Arq. neuropsiquiatr ; 71(5): 284-289, maio 2013. tab, graf
Artigo em Inglês | LILACS | ID: lil-674216

RESUMO

Pompe disease (PD) can be diagnosed by measuring alpha-glucosidase levels or by identifying mutations in the gene enzyme. Muscle biopsies can aid diagnosis in doubtful cases. Methods: A review of muscle biopsy from 19 cases of PD (infantile, 6 cases; childhood, 4 cases; and juvenile/adult, 9 cases). Results: Vacuoles with or without glycogen storage were found in 18 cases. All cases had increased acid phosphatase activity. The vacuole frequency varied (almost all fibers in the infantile form to only a few in the juvenile/adult form). Atrophy of type 1 and 2 fibers was frequent in all forms. Atrophic angular fibers in the NADH-tetrazolium reductase and nonspecific esterase activity were observed in 4/9 of the juvenile/adult cases. Conclusion: Increased acid phosphatase activity and vacuoles were the primary findings. Most vacuoles were filled with glycogen, and the adult form of the disease had fewer fibers with vacuoles than the infantile or childhood forms. .


O diagnóstico da doença de Pompe (PD) pode ser feito pela dosagem da enzima alfa-glicosidase ou pela mutação do seu gene codificador. A biópsia muscular pode ajudar em casos duvidosos. Métodos: Revisão das biópsias musculares de 19 casos de PD (forma infantil, 6 casos; infantil tardia, 4; e juvenil/adulto, 9). Resultados: Encontrados vacúolos em 18 casos, com ou sem depósito de glicogênio. Todos mostraram aumento da fosfatase ácida. Os vacúolos estavam presentes na maioria das fibras nas formas infantis, menos frequentes nas formas juvenil e mais raros nas formas do adulto. A atrofia de fibras dos tipos 1 e 2 ocorreram em todas as formas. Fibras atróficas na NADH-tetrazolium redutase e esterase não específica foram observadas em 4/9 das formas infantil tardia/adulta. Conclusões: Os dados mais frequentes foram vacúolos, preenchidos por glicogênio com atividade aumentada da fosfatase ácida. A forma adulta apresenta menor número de vacúolos que as formas infantil e infantil tardia. .


Assuntos
Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Adulto Jovem , Doença de Depósito de Glicogênio Tipo II/patologia , Músculo Esquelético/patologia , Distribuição por Idade , Biópsia , Eletromiografia , Doença de Depósito de Glicogênio Tipo II/enzimologia , Músculo Esquelético/enzimologia , Estudos Retrospectivos , Distribuição por Sexo , Fatores de Tempo , Vacúolos/enzimologia , Vacúolos/patologia
15.
Rev. neurol. (Ed. impr.) ; 61(9): 416-420, 1 nov., 2015. graf
Artigo em Espanhol | IBECS (Espanha) | ID: ibc-145395

RESUMO

Introducción. La enfermedad de Pompe es un trastorno generalizado progresivo producido por el déficit de la enzima alfaglucosidasa ácida (AGA) de los lisosomas. Se presentan tres casos manifestados de forma muy diferente y tratados con terapia enzimática sustitutiva (TES), con evolución favorable. Casos clínicos. Caso 1: varón de 3 meses, con debilidad y rechazo de la alimentación, hepatomegalia leve, ligera macroglosia e hipotonía, y aumento de las enzimas musculares. Caso 2: varón de 5 meses, con retraso del desarrollo motor, sordera neurosensorial grave, trastornos respiratorios de repetición de evolución tórpida, hipotonía y leve elevación de la creatincinasa. Caso 3: varón de 22 años con disnea progresiva, con antecedentes de elevación de la creatincinasa y las transaminasas, e hipercolesterolemia. Sufrió insuficiencia respiratoria grave que precisó intubación endotraqueal. La biopsia muscular presentó depósitos de glucógeno sugestivos de la enfermedad. En los tres casos, el estudio electromiográfico dio un patrón característico, con descargas pseudomiotónicas, y se confirmó el déficit de AGA en los linfocitos. Se encontró una mutación en un caso y dos mutaciones en los otros dos. Todos recibieron TES con evolución favorable: desaparición de las alteraciones cardíacas en el caso 1, mejoría en los hitos motores en los dos casos infantiles y retirada del respirador en el caso 3. Conclusión. La enfermedad de Pompe tiene una amplia variabilidad en la expresión clínica. La TES consigue una buena respuesta, especialmente en las formas infantiles. La supervivencia a largo plazo de las formas infantiles tratadas permitirá conocer más aspectos del curso de la enfermedad (AU)


Introduction. Pompe disease is a generalized progressive disease caused by a deficiency of the lysosome enzyme acid alphaglucosidase (GAA). We present three cases with different clinical symptomatology and treated with enzyme replacement therapy (ERT) with positive evolution. Case reports. Case 1: three-month old male, with weakness and rejecting meals; mild hepatomegaly, discrete macroglossia and muscular hypotony; and increased muscular enzymes. Case 2: five-month old male, with delayed motor development, severe neurosensory deafness, and respiratory disorder of difficult evolution; muscular hypotony; and mild increase in creatine kinase. Case 3: 22-year old male, with progressive dyspnea, with history of increased creatine kinase and transaminases, and hypercholesterolemia. He suffered from severe respiratory failure requiring endotraqueal intubation Muscular biopsy showed glycogen storage suggestive of Pompe disease. In the three cases, the EMG showed a characteristic pattern with pseudomyotonic discharges and the deficiency in GAA was confirmed in lymphocytes. One single mutation was observed in one case and two in the other two cases. Every patient received ERT showing a favorable evolution; with disappearance of cardiac disorders in case 1, improvement in motor development in both infants and no longer need for mechanical ventilation in case 3. Conclusion. Pompe disease has a wide variability in clinical expression. ERT achieves a good response, especially in infant forms of the disease. The survival of treated patients with these Pompe disease forms will allow knowing further the course of the disease (AU)


Assuntos
Adolescente , Criança , Humanos , Masculino , Doença de Depósito de Glicogênio Tipo II/genética , Doença de Depósito de Glicogênio Tipo II/metabolismo , Terapia Enzimática/métodos , Terapia Enzimática/normas , Músculo Esquelético/anormalidades , Hepatomegalia/patologia , Surdez/diagnóstico , Dispneia/genética , Qualidade de Vida/psicologia , Doença de Depósito de Glicogênio Tipo II/complicações , Doença de Depósito de Glicogênio Tipo II/patologia , Terapia Enzimática/classificação , Terapia Enzimática , Músculo Esquelético/lesões , Hepatomegalia/metabolismo , Surdez/complicações , Dispneia/metabolismo
16.
Arq. bras. cardiol ; 73(5): 435-40, Nov. 1999. ilus
Artigo em Inglês, Português | LILACS | ID: lil-261165

RESUMO

This is the report of a five-month-old child presenting clinical evidence of Pompe's disease: severe hypotonicity, hyporeflexia and congestive heart failure. The ECG showed a short PR interval, the chest radiography disclosed marked cardiomegaly, and the echocardiogram revealed marked left ventricular hypertrophy - the most typical finding of this disease. A skeletal muscle biopsy led to final diagnosis, because in the histopathologic study marked increased glycogen accumulation was evident. Death occurred two months after symptom onset.


Assuntos
Humanos , Feminino , Lactente , Doença de Depósito de Glicogênio Tipo II/diagnóstico , Ecocardiografia , Eletrocardiografia , Doença de Depósito de Glicogênio Tipo II/patologia
17.
Rev. cuba. pediatr ; 46(1): 107-114, ene.-feb.1974. ilus, tab, graf
Artigo em Espanhol | CUMED | ID: cum-25640

RESUMO

Se presenta un nuevo caso de enfermedad de Pompe, que a pesar de reunir las características clínicas y E.C.G. propias del síndrome, no fue diagnosticado en vida. Se insiste en el valor del acortamiento del PR y la presencia de un síndrome de W.P.W. como elementos diagnósticos complementarios. Se concluye que de sospecharse una glicogenosis tipo II, debe practicarse un estudio en sangre periférica utilizando la coloración de P.A.S. para la búsqueda de linfocitos con inclusiones de glicógeno(AU)


Assuntos
Humanos , Doença de Depósito de Glicogênio Tipo II/diagnóstico , Doença de Depósito de Glicogênio Tipo II/patologia , Testes Hematológicos
18.
Acta méd. colomb ; 18(3): 172-6, mayo-jun. 1993. ilus, tab
Artigo em Espanhol | LILACS | ID: lil-183298

RESUMO

La enfermedad de Pompe es una de las glicogenosis o enferemdad de depósito del glucógeno, que se transmite con carácter autosómico recesivo y es producida por la deficiencia de la enzima maltasa ácida que degrada glicógeno en los lisosomas. Se dintinguen tres tipos de enferemdad de Pompe : el tipo infantil que se acracteriza por la acumulación de glicógeno en el hígado, músculo esquelético y cardíaco y células del sistema nervioso central; los pacientes fallecen usaulamente antes de los dos años por falla cardiorespiratoria; el fenotipo juvenil se presenta en niños o jóvenes, no siempre involucra el sistema nervioso central o el músculo cardíaco y los pacientes fallecen generalmente durante la segunda década de vida; el tipo adulto cursa con distrofia muscular, algunas veces asociada con deficiencia respiratoria por compromiso de los músculos diagragma e intercostales. El pronóstico depende del grado de la falla respiratoria, la cual es causa principal de la muerte de estos pacientes. En este trabajo se informa el caso de un niño de seis meses de edad, afectado por cardiomegalia, retardo psicomotor e hipotonía. La confirmación bioquímica de la enfermedad se hizo determinando la actividad de la alfa-glucosidasa en leucocitos por dos métodos diferentes, usando como sustrato maltasa o el sustrato fluorescente 4-metil-umbeliferil-glucósido. La actividad de la maltasa ácida en leucocitos, usando maltosa como sustrato fue de 6,36Umol glucosa/min/g, con un valor de referencia entre 16-63 Umoles/glucosa/min/g de proteína. Con el sustrato fluorogénico, la activida en el paciente fue de 34.48 nanomoles/h/mg, de proteína, para un valor de referencia entre 194 y 258. La actividad en leucocitos tanto del padre como de la madre corresponde a un 60 por ciento del valor normal, hallado en personas en cuya familia no se sospecha dicha enfermedad.


Assuntos
Humanos , Masculino , Lactente , Doença de Depósito de Glicogênio Tipo II/cirurgia , Doença de Depósito de Glicogênio Tipo II/classificação , Doença de Depósito de Glicogênio Tipo II/complicações , Doença de Depósito de Glicogênio Tipo II/diagnóstico , Doença de Depósito de Glicogênio Tipo II/dietoterapia , Doença de Depósito de Glicogênio Tipo II/epidemiologia , Doença de Depósito de Glicogênio Tipo II/etiologia , Doença de Depósito de Glicogênio Tipo II/fisiopatologia , Doença de Depósito de Glicogênio Tipo II/mortalidade , Doença de Depósito de Glicogênio Tipo II/patologia , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Doença de Depósito de Glicogênio Tipo II , Doença de Depósito de Glicogênio Tipo II/terapia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa