Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 174
Filtrar
1.
Acta Neurol Scand ; 137(1): 85-90, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28832912

RESUMEN

OBJECTIVE: Late-onset Pompe disease (LOPD) is a rare autosomal recessively inherited metabolic myopathy caused by reduced activity of the lysosomal enzyme alpha-glucosidase. In a previous screening study at two large neuromuscular university clinics in Denmark, three patients with LOPD were identified out of 103 patients screened. No systematic screening has been performed at the other neurological departments in the western part of Denmark. Thus, patients with a diagnosis of unspecified myopathy were screened for LOPD. MATERIALS AND METHODS: At seven neurological departments in the western part of Denmark, medical records were evaluated for all patients registered with myopathy diagnosis codes (ICD 10 codes: G 71.0-71.9 and G 72.0-72.9) during the period January 1, 2002, to December 31, 2012. If no specific diagnosis has been reached, patients were invited for screening. Dried blood spot (DBS) test was used to analyze the activity of the enzyme alpha-glucosidase. RESULT: A total of 654 patients were identified. From the medical records, information was obtained concerning symptoms, family history, electromyography, muscle biopsy results and creatine kinase levels. Eighty-seven patients (13.3%) (males 61%) at a mean age of 53.3 years (SD 16.5) fulfilled the criteria for screening. A DBS test was performed in 47 (54%) patients. In all patients, the enzyme activity was within reference values. CONCLUSION: None of the screened patients had a reduced activity of the enzyme alpha-glucosidase. Although the cohort studied was small, our findings do not suggest that LOPD is underdiagnosed in patients with unspecified myopathy in western Denmark.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/epidemiología , Adulto , Dinamarca/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , alfa-Glucosidasas/deficiencia
2.
J Pediatr Gastroenterol Nutr ; 66 Suppl 3: S24-S29, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29762372

RESUMEN

The mucosal maltase enzymes are characterized by an activity that produces glucose from linear glucose polymers, assayed with the disaccharide maltose. The related enzyme isomaltase produces glucose from branched glucose polymers, assayed with palatinose. Maltase and isomaltase activities are part of the 4 disaccharidases assayed from clinical duodenal biopsy homogenates. The reported maltase activities are more difficult to interpret than lactase or sucrase activities because both the sucrase-isomaltase and maltase-glucoamylase proteins have overlapping maltase activities. The early work of Dahlqvist identified 4 maltase activities from human small intestinal mucosa. On one peptide, sucrase (maltase Ib) and isomaltase (maltase Ia) activities shared maltase activities but identified the enzymes as sucrase-isomaltase. On the other peptide, no distinguishing characteristics of the 2 maltase activities (maltases II and III) were detected and the activities identified as maltase-glucoamylase. The nutritional/clinical importance of small intestinal maltase and isomaltase activities are due to their crucial role in the digestion of food starches to absorbable free glucose. This review focuses on the interpretation of biopsy maltase activities in the context of reported lactase, sucrase, maltase, and palatinase biopsy assay activity patterns. We present a classification of mucosal maltase deficiencies and novel primary maltase deficiency (Ib, II, III) and provide a clarification of the role of maltase activity assayed from clinically obtained duodenal biopsies, as a path toward future clinical and molecular genomic investigations.


Asunto(s)
Mucosa Intestinal/enzimología , alfa-Glucosidasas/deficiencia , Animales , Digestión/fisiología , Humanos , Mucosa Intestinal/metabolismo , Mutación , alfa-Glucosidasas/análisis , alfa-Glucosidasas/metabolismo
3.
Clin Chem ; 63(7): 1271-1277, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28450385

RESUMEN

BACKGROUND: Deficiency of the lysosomal enzyme acid α-glucosidase (GAA) causes Pompe disease. Newborn screening for Pompe disease is ongoing, and improved methods for distinguishing affected patients from those with pseudodeficiency, especially in the Asian population, would substantially reduce the number of patient referrals for clinical follow-up. METHODS: We measured the enzymatic activity of GAA in dried blood spots on newborn screening cards (DBS) using a tandem mass spectrometry (MS/MS) assay. The assay displayed a relatively large analytical range compared to the fluorimetric assay with 4-methylumbelliferyl-α-glucoside. DBS from newborns confirmed to have infantile-onset Pompe disease (IOPD, n = 11) or late-onset Pompe disease (LOPD) (n = 12) and those from patients bearing pseudodeficiency alleles with or without Pompe mutations, or Pompe disease carriers (n = 230) were studied. RESULTS: With use of the MS/MS GAA assay in DBS, 96% of the pseudodeficiency newborns and all of the Pompe disease carriers were well separated from the IOPD and LOPD newborns. The fluorimetric assay separated <10% of the pseudodeficiencies from the IOPD/LOPD group. CONCLUSIONS: The relatively large analytical range MS/MS GAA assay but not the fluorimetric assay in DBS provides a robust approach to reduce the number of referrals and should dramatically facilitate newborn screening of Pompe disease.


Asunto(s)
Fluorometría , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Diagnóstico Prenatal , Espectrometría de Masas en Tándem , Humanos , Recién Nacido , alfa-Glucosidasas/sangre , alfa-Glucosidasas/deficiencia
4.
Cochrane Database Syst Rev ; 11: CD011539, 2017 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-29155436

RESUMEN

BACKGROUND: Infantile-onset Pompe disease is a rare and progressive autosomal-recessive disorder caused by a deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). Current treatment involves enzyme replacement therapy (with recombinant human alglucosidase alfa) and symptomatic therapies (e.g. to control secretions). Children who are cross-reactive immunological material (CRIM)-negative require immunomodulation prior to commencing enzyme replacement therapy.Enzyme replacement therapy was developed as the most promising therapeutic approach for Pompe disease; however, the evidence is lacking, especially regarding the optimal dose and dose frequency. OBJECTIVES: To assess the effectiveness, safety and appropriate dose regimen of enzyme replacement therapy for treating infantile-onset Pompe disease. SEARCH METHODS: We searched the Cochrane Cystic Fibrosis and Genetic Disorders Group's Inborn Errors of Metabolism Trials Register, which is compiled from electronic database searches and handsearching of journals and conference abstract books. We also searched the Cochrane Central Register of Controlled Trials (CENTRAL), Embase (Ovid), PubMed and LILACS, and CBM, CNKI, VIP, and WANFANG for literature published in Chinese. In addition, we searched three online registers: WHO International Clinical Trials Registry Platform ClinicalTrials.gov, and www.genzymeclinicalresearch.com. We also searched the reference lists of relevant articles and reviews.Date of last search of the Group's Inborn Errors of Metabolism Trials Register: 24 November 2016. SELECTION CRITERIA: Randomized and quasi-randomized controlled trials of enzyme replacement therapy in children with infantile-onset Pompe disease. DATA COLLECTION AND ANALYSIS: Two authors independently selected relevant trials, assessed the risk of bias and extracted data. We contacted investigators to obtain important missing information. MAIN RESULTS: We found no trials comparing the effectiveness and safety of enzyme replacement therapy to another intervention, no intervention or placebo.We found one trial (18 participants) that fulfilled the selection criteria, comparing different doses of alglucosidase alfa. The trial provided low-quality evidence (this was a small trial, there were no numerical results available by dose group, random sequence generation and allocation concealment were unclear, and there was a lack of blinding). The duration of alglucosidase alfa treatment ranged from 52 weeks (the length of the original study) to up to three years (including the extended phase of the trial), with a median duration of treatment being 2.3 years.The trial only reported that clinical responses including cardiac function and motor development, as well as the proportion of children that were free of invasive ventilation, were similar in the 20 mg/kg every two weeks and the 40 mg/kg every two weeks groups (low-quality evidence). Long-term alglucosidase alfa treatment markedly extended survival as well as ventilation-free survival and improved cardiomyopathy (low-quality evidence). In relation to the number of children experiencing one or more infusion-related events, there was no significant difference between dose groups, risk ratio 0.83 (95% confidence interval 0.40 to 1.76) (low-quality of evidence). However, of note, at 52 weeks, five children in the 20 mg/kg every two weeks dose group experienced a total of 41 mild or moderate (none severe) infusion-related events and the six children in the 40 mg/kg every two weeks dose group experienced a total of 123 infusion-related events. By the end of the extended phase of the trial, five children in the 20 mg/kg every two weeks dose group experienced a total of 47 infusion-related events and the six children in the 40 mg/kg every two weeks dose group experienced a total of 177 infusion-related events. The trial was supported by the Genzyme Corporation. AUTHORS' CONCLUSIONS: The search found no trials comparing the effectiveness and safety of enzyme replacement therapy to another intervention, no intervention or placebo. One small randomized controlled trial provided no robust evidence for which dosing schedule of alglucosidase alfa was more effective to treat infantile-onset Pompe disease. It is not deemed ethical to proceed with new placebo-controlled trials, therefore a randomized controlled trial with a large sample size comparing different dosing schedules of enzyme replacement therapy is needed. The main clinical outcomes (i.e. cardiac function, invasive ventilation, survival, motor development, adverse events (e.g. the development of antibodies)) should be standardized when evaluated and reported.


Asunto(s)
Terapia de Reemplazo Enzimático/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , alfa-Glucosidasas/administración & dosificación , Desarrollo Infantil , Terapia de Reemplazo Enzimático/efectos adversos , Humanos , Lactante , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Respiración Artificial/estadística & datos numéricos , alfa-Glucosidasas/efectos adversos , alfa-Glucosidasas/deficiencia
5.
Physiol Genomics ; 48(11): 785-794, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27614205

RESUMEN

Pompe disease, caused by deficiency of acid alpha-glucosidase (GAA), leads to widespread glycogen accumulation and profound neuromuscular impairments. There has been controversy, however, regarding the role of central nervous system pathology in Pompe motor dysfunction. We hypothesized that absence of GAA protein causes progressive activation of neuropathological signaling, including pathways associated with cell death. To test this hypothesis, genomic data (Affymetrix Mouse Gene Array 2.0ST) from the midcervical spinal cord in 6 and 16 mo old Pompe (Gaa-/-) mice were evaluated (Broad Institute Molecular Signature Database), along with spinal cord histology. The midcervical cord was selected because it contains phrenic motoneurons, and phrenic-diaphragm dysfunction is prominent in Pompe disease. Several clinically important themes for the neurologic etiology of Pompe disease emerged from this unbiased genomic assessment. First, pathways associated with cell death were strongly upregulated as Gaa-/- mice aged, and motoneuron apoptosis was histologically verified. Second, proinflammatory signaling was dramatically upregulated in the Gaa-/- spinal cord. Third, many signal transduction pathways in the Gaa-/- cervical cord were altered in a manner suggestive of impaired synaptic function. Notably, glutamatergic signaling pathways were downregulated, as were "synaptic plasticity pathways" including genes related to neuroplasticity. Fourth, many genes and pathways related to cellular metabolism are dysregulated. Collectively, the data unequivocally confirm that systemic absence of GAA induces a complex neuropathological cascade in the spinal cord. Most importantly, the results indicate that Pompe is a neurodegenerative condition, and this underscores the need for early therapeutic intervention capable of targeting the central nervous system.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Médula Espinal/patología , Transcriptoma/genética , alfa-Glucosidasas/deficiencia , Animales , Muerte Celular , Vértebras Cervicales/patología , Perfilación de la Expresión Génica , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Inflamación/patología , Ratones , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , alfa-Glucosidasas/metabolismo
6.
J Inherit Metab Dis ; 38(2): 305-14, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24715333

RESUMEN

BACKGROUND: Enzyme-replacement therapy (ERT) in Pompe disease--an inherited metabolic disorder caused by acid α-glucosidase deficiency and characterized in infants by generalized muscle weakness and cardiomyopathy--can be complicated by immune responses. Infants that do not produce any endogenous acid α-glucosidase, so-called CRIM-negative patients, reportedly develop a strong response. We report the clinical outcome of our Dutch infants in relation to their CRIM status and immune response. METHODS: Eleven patients were genotyped and their CRIM status was determined. Antibody formation and clinical outcome were assessed for a minimum of 4 years. RESULTS: ERT was commenced between 0.1 and 8.3 months of age, and patients were treated from 0.3 to 13.7 years. All patients developed antibodies. Those with a high antibody titer (above 1:31,250) had a poor response. The antibody titers varied substantially between patients and did not strictly correlate with the patients' CRIM status. Patients who started ERT beyond 2 months of age tended to develop higher titers than those who started earlier. All three CRIM-negative patients in our study succumbed by the age of 4 years seemingly unrelated to the height of their antibody titer. CONCLUSION: Antibody formation is a common response to ERT in classic infantile Pompe disease and counteracts the effect of treatment. The counteracting effect seems determined by the antibody:enzyme molecular stoichiometry. The immune response may be minimized by early start of ERT and by immune modulation, as proposed by colleagues. The CRIM-negative status itself seems associated with poor outcome.


Asunto(s)
Anticuerpos/sangre , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , alfa-Glucosidasas/uso terapéutico , Factores de Edad , Biomarcadores/sangre , Células Cultivadas , Preescolar , Progresión de la Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/inmunología , Enfermedad del Almacenamiento de Glucógeno Tipo II/mortalidad , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Países Bajos , Fenotipo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Factores de Riesgo , Factores de Tiempo , Transfección , Resultado del Tratamiento , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética , alfa-Glucosidasas/inmunología
7.
Mol Genet Metab ; 111(2): 92-100, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24044919

RESUMEN

Pompe disease is caused by a deficiency of acid α-glucosidase (GAA; EC, 3.2.1.20), and the infantile-onset form is rapidly fatal if left untreated. However, recombinant human GAA (rhGAA) enzyme replacement therapy (ERT) extends survival for infantile Pompe patients. Although cross-reactive immunologic material (CRIM)-negative patients, who lack detectable endogenous GAA, mount an immune response to rhGAA that renders the therapy ineffective, timely induction of immune tolerance in these patients may improve clinical outcomes. Previously, CRIM status has been determined by Western blot analysis in cultured skin fibroblasts, a process that can take a few weeks. We present a blood-based CRIM assay that can yield results within 48 to 72 h. Results from this assay have been confirmed by GAA Western blot analysis in fibroblasts or by GAA sequencing in a small number of Pompe disease patients. Rapid classification of CRIM status will assist in identifying the most effective treatment course and minimizing treatment delays in patients with infantile-onset Pompe disease.


Asunto(s)
Linfocitos B/inmunología , Bioensayo , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , alfa-Glucosidasas/inmunología , Linfocitos B/química , Western Blotting , Células Cultivadas , Reacciones Cruzadas , Fibroblastos/química , Fibroblastos/inmunología , Enfermedad del Almacenamiento de Glucógeno Tipo II/sangre , Enfermedad del Almacenamiento de Glucógeno Tipo II/inmunología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Inmunosupresores/uso terapéutico , Recién Nacido , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Factores de Tiempo , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/uso terapéutico
8.
Am J Physiol Regul Integr Comp Physiol ; 307(10): R1251-9, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25231351

RESUMEN

Pompe disease is due to a deficiency in acid-α-glucosidase (GAA) and results in debilitating skeletal muscle wasting, characterized by the accumulation of glycogen and autophagic vesicles. Given the role of lysosomes as a platform for mTORC1 activation, we examined mTORC1 activity in models of Pompe disease. GAA-knockdown C2C12 myoblasts and GAA-deficient human skin fibroblasts of infantile Pompe patients were found to have decreased mTORC1 activation. Treatment with the cell-permeable leucine analog L-leucyl-L-leucine methyl ester restored mTORC1 activation. In vivo, Pompe mice also displayed reduced basal and leucine-stimulated mTORC1 activation in skeletal muscle, whereas treatment with a combination of insulin and leucine normalized mTORC1 activation. Chronic leucine feeding restored basal and leucine-stimulated mTORC1 activation, while partially protecting Pompe mice from developing kyphosis and the decline in muscle mass. Leucine-treated Pompe mice showed increased spontaneous activity and running capacity, with reduced muscle protein breakdown and glycogen accumulation. Together, these data demonstrate that GAA deficiency results in reduced mTORC1 activation that is partly responsible for the skeletal muscle wasting phenotype. Moreover, mTORC1 stimulation by dietary leucine supplementation prevented some of the detrimental skeletal muscle dysfunction that occurs in the Pompe disease mouse model.


Asunto(s)
Suplementos Dietéticos , Dipéptidos/farmacología , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Complejos Multiproteicos/metabolismo , Músculo Esquelético/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , alfa-Glucosidasas/deficiencia , Animales , Línea Celular , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Humanos , Insulina/farmacología , Cifosis/enzimología , Cifosis/patología , Cifosis/fisiopatología , Cifosis/prevención & control , Lisosomas/efectos de los fármacos , Lisosomas/enzimología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Atrofia Muscular/enzimología , Atrofia Muscular/patología , Atrofia Muscular/fisiopatología , Atrofia Muscular/prevención & control , Mioblastos/efectos de los fármacos , Mioblastos/enzimología , Interferencia de ARN , Transfección , alfa-Glucosidasas/genética
9.
Acta Myol ; 32(2): 82-4, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24399863

RESUMEN

Glycogen storage disease type II - also called Pompe disease or acid maltase deficiency - is an autosomal recessive metabolic disorder, caused by an accumulation of glycogen in the lysosome due to deficiency of the lysosomal acid alpha-glucosidase enzyme. Pompe disease is transmitted as an autosomal recessive trait and is caused by mutations in the gene encoding the acid α-glucosidase (GAA), located on chromosome 17q25.2-q25.3. The different disease phenotypes are related to the levels of residual GAA activity in muscles. The clinical spectrum ranging from the classical form with early onset and severe phenotype to not-classical form with later onset and milder phenotype is described.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Debilidad Muscular/diagnóstico , alfa-Glucosidasas , Edad de Inicio , Diagnóstico Diferencial , Predisposición Genética a la Enfermedad , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/epidemiología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Humanos , Lisosomas/metabolismo , Debilidad Muscular/etiología , Debilidad Muscular/fisiopatología , Mutación , Pronóstico , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética
10.
Acta Myol ; 32(2): 95-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24399866

RESUMEN

Glycogen-storage disease type II, also named Pompe disease, is caused by the deficiency of the enzyme acid alpha-glucosidase, which originates lysosomal glycogen accumulation leading to progressive neuromuscular damage. Early-onset Pompe disease shows a debilitating and frequently fulminating course. To date, more than 300 mutations have been described; the majority of them are unique to each affected individual. Most early-onset phenotypes are associated with frameshift mutations leading to a truncated alpha-glucosidase protein with loss of function. Founder effects are responsible from many cases from few highprevalence world regions. Herein we described two apparently unrelated cases affected with classical early-onset Pompe disease, both pertaining to a small region from Central Mexico (the State of San Luis Potosí), the same novel homozygous frameshift mutation at gene GAA (c.1987delC) was demonstrated in both cases. This GAA gene deletion implies a change of glutamine to serine at codon 663, and a new reading frame that ends after 33 base pairs, which leads to the translation of a truncated protein. This report contributes to widen the knowledge on the effect of pathogenic mutations in Pompe disease. Here we postulate the existence of a founder effect.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , alfa-Glucosidasas , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/etiología , Resultado Fatal , Efecto Fundador , Predisposición Genética a la Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo II/complicaciones , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Homocigoto , Humanos , Lactante , Masculino , México , Mutación , Radiografía , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética
11.
Acta Myol ; 32(2): 91-4, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24399865

RESUMEN

Glycogenosis II (GSD II) is an autosomal recessive lysosomal storage disorder resulting from acid alpha-glucosidase deficiency, subsequent accumulation of glycogen in tissues, impairment of autophagic processes and progressive cardiac, motor and respiratory failure. The late-onset form is characterized by wide variability in residual enzyme activity, age of onset, rate of disease progression and phenotypical spectrum. Although the pathological process mainly affects the skeletal muscle, several other tissues may be involved in the course of the disease; therefore GSD II should be regarded as a multisystem disorder in which glycogen accumulation is present in skeletal and smooth muscle, heart, brain, liver, spleen, salivary glands, kidney and blood vessels. In this review, we briefly summarize the main non-muscle targets of the pathological process in late-onset GSD II. Further studies aimed at evaluating the extra-muscle involvement in this group of patients will help to better define clinical features and prognostic factors and to delineate the natural history of the disease.


Asunto(s)
Enfermedades Óseas , Enfermedad del Almacenamiento de Glucógeno Tipo II/complicaciones , Enfermedades del Sistema Nervioso , Enfermedades Vasculares , Edad de Inicio , Enfermedades Óseas/diagnóstico , Enfermedades Óseas/etiología , Enfermedades Óseas/fisiopatología , Progresión de la Enfermedad , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Humanos , Enfermedades del Sistema Nervioso/diagnóstico , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/fisiopatología , Fenotipo , Pronóstico , Enfermedades Vasculares/diagnóstico , Enfermedades Vasculares/etiología , Enfermedades Vasculares/fisiopatología , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética
12.
Am J Med Genet C Semin Med Genet ; 160C(1): 69-79, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22252989

RESUMEN

Enzyme replacement therapy (ERT) with alglucosidase alpha, approved by the FDA in 2006, has expanded possibilities for individuals with Pompe disease (glycogen storage disease type II, GSDII, or acid maltase deficiency). Children with infantile Pompe disease are surviving beyond infancy, some achieving independent walking and functional levels never before possible. Individuals with late-onset Pompe disease are experiencing motor and respiratory improvement and/or stabilization with slower progression of impairments. A new phenotype is emerging for those with infantile Pompe disease treated with ERT. This new phenotype appears to be distinct from the late-onset phenotype rather than a shift from infantile to late-onset phenotype that might be expected from a simple diminution of symptoms with ERT. Questions arise regarding the etiology of the distinct distribution of weakness in this new phenotype, with increasing questions regarding exercise and musculoskeletal management. Answers require an increased understanding of the muscle pathology in Pompe disease, how that muscle pathology may be impacted by ERT, and the potential impact of, and need for, other clinical interventions. This article reviews the current state of knowledge regarding the pathology of muscle involvement in Pompe disease and the potential change in muscle pathology with ERT; the newly emerging musculoskeletal and gross motor phenotype of infantile Pompe disease treated with ERT; updated recommendations regarding musculoskeletal management in Pompe disease, particularly in children now surviving longer with residual weakness impacting development and integrity of the musculoskeletal system; and the potential impact and role of exercise in infantile Pompe survivors treated with ERT.


Asunto(s)
Ejercicio Físico , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Músculo Esquelético/patología , Edad de Inicio , Niño , Preescolar , Terapia de Reemplazo Enzimático/efectos adversos , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Humanos , Actividad Motora/fisiología , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/metabolismo
13.
Am J Med Genet C Semin Med Genet ; 160C(1): 13-21, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22253254

RESUMEN

Macroautophagy (often referred to as autophagy) is an evolutionarily conserved intracellular system by which macromolecules and organelles are delivered to lysosomes for degradation and recycling. Autophagy is robustly induced in response to starvation in order to generate nutrients and energy through the lysosomal degradation of cytoplasmic components. Constitutive, basal autophagy serves as a quality control mechanism for the elimination of aggregated proteins and worn-out or damaged organelles, such as mitochondria. Research during the last decade has made it clear that malfunctioning or failure of this system is associated with a wide range of human pathologies and age-related diseases. Our recent data provide strong evidence for the role of autophagy in the pathogenesis of Pompe disease, a lysosomal glycogen storage disease caused by deficiency of acid alpha-glucosidase (GAA). Large pools of autophagic debris in skeletal muscle cells can be seen in both our GAA knockout model and patients with Pompe disease. In this review, we will focus on these recent data, and comment on the not so recent observations pointing to the involvement of autophagy in skeletal muscle damage in Pompe disease.


Asunto(s)
Autofagia/fisiología , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Lisosomas/enzimología , Mitocondrias/patología , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/metabolismo , Animales , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Humanos , Lisosomas/patología , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/patología , alfa-Glucosidasas/genética
14.
Mol Genet Metab ; 105(3): 428-32, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22217428

RESUMEN

This report describes the cognitive development of 17 children with infantile Pompe disease who participated in a 52-week clinical trial of enzyme replacement therapy (ERT) via biweekly infusion of Myozyme® (alglucosidase alfa). Subjects were six months of age or younger (adjusted for gestational age) upon initiation of ERT. The Mental Scale of the Bayley Scales of Infant Development-Second Edition (BSID-II) was administered to obtain a Mental Development Index (MDI) at baseline and weeks 12, 26, 38, and 52 of ERT to assess cognitive development in this treated cohort. Data regarding motor development were also obtained at the same visits and these were used to determine correlations between cognitive and motor development. Over the course of the study, two subgroups of subjects emerged: high responders who were sitting independently and/or ambulating by week 52 (n=13) and limited responders who showed minimal motor gains throughout the first year of ERT (n=4). In the high responder group, MDI scores on the BSID-II remained stable throughout the study and were within normal limits. Positive correlations between cognitive and motor development were also present. These data suggest that the cognitive function of infants up to 18 months of age with Pompe disease is unaffected by the possible presence of glycogen in the central nervous system. Continued investigation of the cognitive development of older survivors is warranted.


Asunto(s)
Cognición , Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , alfa-Glucosidasas/uso terapéutico , Estudios de Cohortes , Femenino , Glucógeno/análisis , Enfermedad del Almacenamiento de Glucógeno Tipo II/psicología , Humanos , Lactante , Recién Nacido , Masculino , Destreza Motora , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo
15.
Mol Genet Metab ; 106(3): 281-6, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22578805

RESUMEN

BACKGROUND: Pompe disease is caused by a deficiency in acid α-glucosidase (GAA) and results in progressive, debilitating, and often life-threatening symptoms. Newborn screening has led to the early diagnosis of Pompe disease, but the best algorithm for screening has not yet been established. MATERIALS AND METHODS: GAA and neutral α-glucosidase (NAG) activities in dried blood spots (DBSs) were assayed using 4-methylumbelliferyl-ß-d-glucopyranoside as the substrate. We also measure α-galactosidase A (GLA) activity in DBSs for comparison. A total of 473,738 newborns were screened for Pompe disease, and the data were analyzed retrospectively to determine the best screening algorithm. RESULTS: The fluorescence assay used in the screening possessed good reproducibility, but the NAG/GAA ratio was superior in separating the true-positive from the false-positive cases. An NAG/GAA cutoff ratio≥60 produces a positive predictive value (PPV) of 63.4%, and in our sample, only two cases of later-onset Pompe disease would have been missed. The GLA/GAA ratio is not as effective as the NAG/GAA ratio. CONCLUSION: A suitable control enzyme can improve the performance of newborn screening. Newborn screening for Pompe disease can be performed using the NAG/GAA ratio as a cutoff even in the presence of GAA partial deficiency.


Asunto(s)
Algoritmos , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Tamizaje Neonatal/métodos , Secuencia de Bases , Glucósidos/química , Glucósidos/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Humanos , Himecromona/análogos & derivados , Himecromona/química , Himecromona/metabolismo , Recién Nacido , Datos de Secuencia Molecular , Reproducibilidad de los Resultados , Taiwán , alfa-Galactosidasa/química , alfa-Galactosidasa/metabolismo , alfa-Glucosidasas/química , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/metabolismo
16.
Semin Neurol ; 32(5): 506-11, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23677658

RESUMEN

Glycogen storage disease type II, also known as Pompe's disease or acid maltase deficiency, is caused by a deficiency in acid α-glucosidase. Severe enzyme deficiency results in infantile Pompe's disease with multiorgan involvement; a partial deficiency produces a less severe phenotype mainly consisting of a myopathy, with a later age of onset. Treatment is now available with intravenous infusion of recombinant acid α-glucosidase. Such treatment results in marked improvement in patients with infantile Pompe's disease, and modest improvement or stabilization in patients with late-onset Pompe's disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , alfa-Glucosidasas/deficiencia , Edad de Inicio , Progresión de la Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Humanos , Resultado del Tratamiento , alfa-Glucosidasas/metabolismo
17.
Curr Neurol Neurosci Rep ; 12(1): 70-5, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22002767

RESUMEN

Late-onset glycogenosis type II (glycogen storage disease type II [GSDII]) is a rare autosomal disorder caused by deficiency of acid maltase, a lysosomal enzyme that hydrolyzes glycogen to glucose. Recently, both infantile and adult GSDII patients have been treated with enzyme replacement therapy (ERT), and a number of studies including large cohorts of GSDII patients have recently demonstrated that ERT is effective in modifying the natural course of the disease. The opportunity of this new treatment gave new hope to patients, but also an important impulse to the research on every feature of the disease, leading to a deeper knowledge on the response to treatment, on clinical manifestations, and on pathophysiologic aspects such as the role of autophagy and immune status.


Asunto(s)
Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , alfa-Glucosidasas/uso terapéutico , Animales , Glucógeno/metabolismo , Humanos , Músculo Esquelético/citología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética
18.
Mol Genet Metab ; 103(1): 12-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21320792

RESUMEN

The high frequency (3.3-3.9%) of acid α-glucosidase pseudodeficiency, c.[1726G>A; 2065G>A] homozygote (AA homozygote), in Asian populations complicates newborn screening for Pompe disease (glycogen storage disease type II or acid maltase deficiency) on dried blood spots, since AA homozygotes have a considerably low enzyme activity. We observed that hemoglobin in the enzyme reaction solution strongly interferes with the fluorescence of 4-methylumbelliferone released from 4-methylumbelliferyl α-D-glucopyranoside (4MU-αGlc) by acid α-glucosidase. Therefore, we have searched for a method to effectively eliminate hemoglobin in the reaction solution. Hemoglobin precipitation with barium hydroxide and zinc sulfate (Ba/Zn method) carried out after the enzyme reaction considerably enhances the fluorescence intensity while it does not reduce the intensity to any extent as can occur with conventional deproteinization agents like trichloroacetic acid. The Ba/Zn method greatly improved the separation between 18 Japanese patients with Pompe disease and 70 unaffected AA homozygotes in a population of Japanese newborns in the assay with 4MU-αGlc on dried blood spots. No overlap was observed between both groups. We further examined acid α-glucosidase activity in fibroblasts from 11 Japanese patients and 57 Japanese unaffected individuals including 31 c.[1726G; 2065G] homozygotes, 18 c.[1726G; 2065G]/[1726A; 2065A] heterozygotes and 8 AA homozygotes to confirm that fibroblasts can be used for definitive diagnosis. The patients were reliably distinguished from three control groups. These data provide advanced information for the development of a simple and reliable newborn screening program with dried blood spots for Pompe disease in Asian populations.


Asunto(s)
Pruebas Enzimáticas Clínicas/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/sangre , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Pruebas Hematológicas/métodos , Tamizaje Neonatal , alfa-Glucosidasas/sangre , Adulto , Niño , Fibroblastos/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Homocigoto , Humanos , Lactante , Recién Nacido , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética
19.
Orv Hetil ; 152(39): 1569-75, 2011 Sep 25.
Artículo en Húngaro | MEDLINE | ID: mdl-21920843

RESUMEN

UNLABELLED: Pompe's disease is an autosomal recessive disease caused by deficiency of acid-alpha-glucosidase. AIMS AND METHODS: Authors analyzed the phenotype of 11 Hungarian patients with Pompe's disease and evaluated clinical parameters and response to enzyme replacement therapy during a long-term follow-up in 8 patients. RESULTS: One patient with atypical infantile form presented with cardiomyopathy and a very slow progression of motor deficits; after 2 years of enzyme replacement therapy no disability was present at the age 6 years. Another patient was asymptomatic at the age of 2.5 years. The adult onset form was characterized by slight to prominent limb-girdle myopathy with an age of onset between 20 and 50 years. In 3 of such cases respiratory insufficiency was also present. CONCLUSIONS: Hungarian patients with Pompe's disease presented with a wide phenotypic variability ranging from atypical early childhood form with slowly progressive course to late-onset limb-girdle myopathy with variable courses. Enzyme replacement therapy resulted in significant improvement in motor and respiratory functions in most of the patients.


Asunto(s)
Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Insuficiencia Respiratoria/etiología , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/uso terapéutico , Adulto , Edad de Inicio , Dióxido de Carbono/metabolismo , Niño , Preescolar , Progresión de la Enfermedad , Terapia de Reemplazo Enzimático/métodos , Femenino , Volumen Espiratorio Forzado , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Humanos , Hungría , Masculino , Persona de Mediana Edad , Oxígeno/metabolismo , Fenotipo , Insuficiencia Respiratoria/enzimología , Insuficiencia Respiratoria/fisiopatología , Factores de Tiempo , alfa-Glucosidasas/genética
20.
Hum Mol Genet ; 17(24): 3897-908, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18782848

RESUMEN

The role of autophagy, a catabolic lysosome-dependent pathway, has recently been recognized in a variety of disorders, including Pompe disease, the genetic deficiency of the glycogen-degrading lysosomal enzyme acid-alpha glucosidase. Accumulation of lysosomal glycogen, presumably transported from the cytoplasm by the autophagic pathway, occurs in multiple tissues, but pathology is most severe in skeletal and cardiac muscle. Skeletal muscle pathology also involves massive autophagic buildup in the core of myofibers. To determine if glycogen reaches the lysosome via autophagy and to ascertain whether autophagic buildup in Pompe disease is a consequence of induction of autophagy and/or reduced turnover due to defective fusion with lysosomes, we generated muscle-specific autophagy-deficient Pompe mice. We have demonstrated that autophagy is not required for glycogen transport to lysosomes in skeletal muscle. We have also found that Pompe disease involves induction of autophagy but manifests as a functional deficiency of autophagy because of impaired autophagosomal-lysosomal fusion. As a result, autophagic substrates, including potentially toxic aggregate-prone ubiquitinated proteins, accumulate in Pompe myofibers and may cause profound muscle damage.


Asunto(s)
Autofagia , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Músculo Esquelético/patología , Enfermedades Musculares/etiología , Enfermedades Musculares/patología , Proteínas/efectos adversos , Proteínas/metabolismo , Ubiquitinación , Animales , Autofagia/genética , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Proteínas/genética , Ubiquitinación/genética , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda