Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
Orphanet J Rare Dis ; 11(1): 65, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27189384

ABSTRACT

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.


Subject(s)
Genotype , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Adolescent , Child , Child, Preschool , Cross-Sectional Studies , Diagnosis, Differential , Female , Humans , Infant , Male , Motor Activity , Mutation , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
2.
J Inherit Metab Dis ; 39(3): 383-390, 2016 05.
Article in English | MEDLINE | ID: mdl-26768149

ABSTRACT

BACKGROUND: Though enzyme-replacement therapy (ERT) with alglucosidase alfa has significantly improved the prospects for patients with classic infantile Pompe disease, some 50 % of treated infants do not survive ventilator-free beyond the age of 3 years. We investigated whether higher and more frequent dosing of alglucosidase alfa improves outcome. METHODS: Eight cross-reactive immunological material (CRIM) positive patients were included in the study. All had fully deleterious mutations in both GAA alleles. Four received a dose of 20 mg/kg every other week (eow) and four received 40 mg/kg/week. Survival, ventilator-free survival, left-ventricular mass index (LVMI), motor outcome, infusion-associated reactions (IARs), and antibody formation were evaluated. RESULTS: All eight patients were alive at study end, seven of them remained ventilator-free. The patient who became ventilator dependent was treated with 20 mg/kg eow. Three of the four patients receiving 20 mg/kg eow learned to walk; two of them maintained this ability at study end. All four patients receiving 40 mg/kg/week acquired and maintained the ability to walk at study end (ages of 3.3-5.6 years), even though their baseline motor functioning was poorer. There were no apparent differences between the two dose groups with respect to the effect of ERT on LVMI, the number of IARs and antibody formation. CONCLUSIONS: Our data may suggest that a dose of 40 mg/kg/week improves outcome of CRIM positive patients over that brought by the currently recommended dose of 20 mg/kg eow. Larger studies are needed to draw definite conclusions.


Subject(s)
Glycogen Storage Disease Type II/drug therapy , alpha-Glucosidases/therapeutic use , Child , Child, Preschool , Cross Reactions/physiology , Enzyme Replacement Therapy/methods , Female , Humans , Infant , Infant, Newborn , Male , Treatment Outcome , Ventilators, Mechanical
4.
Gene ; 537(1): 41-5, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24384324

ABSTRACT

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).


Subject(s)
Glucan 1,4-alpha-Glucosidase/genetics , Glucan 1,4-alpha-Glucosidase/metabolism , Glycogen Storage Disease Type II/genetics , Mutation, Missense , Age of Onset , Case-Control Studies , Female , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/epidemiology , HEK293 Cells , Humans , Infant , Male , Middle Aged , Mutation , Pedigree , Phenotype , Pregnancy , Quadriceps Muscle/enzymology , RNA Splicing , Reference Values
6.
JIMD Rep ; 14: 23-7, 2014.
Article in English | MEDLINE | ID: mdl-24272678

ABSTRACT

Hunter disease (Mucopolysaccharidosis type II, MPS II) is an X-linked lysosomal storage disorder caused by deficiency of iduronate-2-sulfatase (IDS). Two main therapies have been reported for MPS II patients: enzyme-replacement therapy (ERT) and hematopoietic stem-cell transplantation (HSCT). Both treatment modalities have been shown to improve some symptoms, but the results with regard to cognitive functioning have been poor. Early initiation of therapy, i.e., before neurological symptoms have manifested, may alter cognitive outcome. The need for early identification makes Hunter disease a candidate for newborn screening (NBS). Our objective was to explore the use of a fluorometric assay that could be applicable for high-throughput analysis of IDS activity in dried blood spots (DBS). The median IDS activity in DBS samples from 1,426 newborns was 377 pmol/punch/17 h (range 78-1111). The IDS activity in one sample was repeatedly under the cutoff value (set at 20% of the median value), which would imply a recall rate of 0.07%. A sample from a clinically diagnosed MPS II individual, included in each 96-well test plate, had IDS activities well below the 20% cutoff value. Coefficients of variation in quality control samples with low, medium, and high IDS activities (190, 304, and 430 pmol/punch/17 h, respectively) were 12% to 16%. This small-scale pilot study shows that newborn screening for Hunter disease using a fluorometric assay in DBS is technically feasible with a fairly low recall rate. NBS may allow for identification of infants with Hunter disease before clinical symptoms become evident enabling early intervention.

7.
J Inherit Metab Dis ; 36(5): 787-94, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23053471

ABSTRACT

Pompe disease is a lysosomal storage disorder caused by acid α-glucosidase deficiency and characterized by progressive muscle weakness. Enzyme replacement therapy (ERT) has ameliorated patients' perspectives, but reversal of skeletal muscle pathology remains a challenge. We studied pretreatment biopsies of 22 patients with different phenotypes to investigate to what extent fiber-type distribution and fiber-type-specific damage contribute to clinical diversity. Pompe patients have the same fiber-type distribution as healthy persons, but among nonclassic patients with the same GAA mutation (c.-32-13T>G), those with early onset of symptoms tend to have more type 2 muscle fibers than those with late-onset disease. Further, it seemed that the older, more severely affected classic infantile patients and the wheelchair-bound and ventilated nonclassic patients had a greater proportion of type 2x muscle fibers. However, as in other diseases, this may be caused by physical inactivity of those patients.


Subject(s)
Glycogen Storage Disease Type II/pathology , Muscle Fibers, Skeletal/pathology , Adolescent , Adult , Biopsy , Cross-Sectional Studies , Humans , Phenotype
10.
J Inherit Metab Dis ; 35(3): 505-11, 2012 May.
Article in English | MEDLINE | ID: mdl-22008944

ABSTRACT

Classic infantile Pompe disease is an inherited generalized glycogen storage disorder caused by deficiency of lysosomal acid α-glucosidase. If left untreated, patients die before one year of age. Although enzyme-replacement therapy (ERT) has significantly prolonged lifespan, it has also revealed new aspects of the disease. For up to 11 years, we investigated the frequency and consequences of facial-muscle weakness, speech disorders and dysphagia in long-term survivors. Sequential photographs were used to determine the timing and severity of facial-muscle weakness. Using standardized articulation tests and fibreoptic endoscopic evaluation of swallowing, we investigated speech and swallowing function in a subset of patients. This study included 11 patients with classic infantile Pompe disease. Median age at the start of ERT was 2.4 months (range 0.1-8.3 months), and median age at the end of the study was 4.3 years (range 7.7 months -12.2 years). All patients developed facial-muscle weakness before the age of 15 months. Speech was studied in four patients. Articulation was disordered, with hypernasal resonance and reduced speech intelligibility in all four. Swallowing function was studied in six patients, the most important findings being ineffective swallowing with residues of food (5/6), penetration or aspiration (3/6), and reduced pharyngeal and/or laryngeal sensibility (2/6). We conclude that facial-muscle weakness, speech disorders and dysphagia are common in long-term survivors receiving ERT for classic infantile Pompe disease. To improve speech and reduce the risk for aspiration, early treatment by a speech therapist and regular swallowing assessments are recommended.


Subject(s)
Deglutition Disorders/pathology , Enzyme Therapy/methods , Facial Muscles/pathology , Glycogen Storage Disease Type II/pathology , Muscle Weakness/pathology , Speech Disorders/pathology , Child , Child, Preschool , Deglutition , Deglutition Disorders/diagnosis , Female , Glycogen Storage Disease Type II/diagnosis , Humans , Infant , Larynx/pathology , Male , Muscle Weakness/diagnosis , Pharynx/pathology , Speech , Speech Therapy/methods
11.
Biochim Biophys Acta ; 1812(11): 1542-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21827850

ABSTRACT

The mucopolysaccharidoses (MPS) are prominent among the lysosomal storage diseases. The intra-lysosomal accumulation of glycosaminoglycans (GAGs) in this group of diseases, which are caused by several different enzyme deficiencies, induces a cascade of responses that affect cellular functions and maintenance of the extra-cellular matrix. Against the background of normal tissue-specific processes, this review summarizes and discusses the histological and biochemical abnormalities reported in the bones, joints, teeth and extracellular matrix of MPS patients and animal models. With an eye to the possibilities and limitations of reversing the pathological changes in the various tissues, we address therapeutic challenges, and present a model in which the cascade of pathologic events is depicted in terms of primary and secondary events.


Subject(s)
Bone and Bones/cytology , Joints/growth & development , Mucopolysaccharidoses/physiopathology , Mucopolysaccharidoses/therapy , Tooth/growth & development , Animals , Humans
12.
Mol Genet Metab ; 104(1-2): 129-36, 2011.
Article in English | MEDLINE | ID: mdl-21763167

ABSTRACT

Respiratory insufficiency is a serious threat to patients with Pompe disease, a neuromuscular disorder caused by lysosomal acid alpha-glucosidase deficiency. Innovative therapeutic options which may stabilize pulmonary function have recently become available. We therefore determined proportion and severity of pulmonary involvement in patients with Pompe disease, the rate of progression of pulmonary dysfunction, and predictive factors for poor respiratory outcome. In a single-center, prospective, cohort study, we measured vital capacity (VC) in sitting and supine positions, as well as maximum inspiratory (MIP) and expiratory (MEP) mouth pressures, and end expiratory CO(2) in 17 children and 75 adults with Pompe disease (mean age 42.7 years, range 5-76 years). Seventy-four percent of all patients, including 53% of the children, had some degree of respiratory dysfunction. Thirty-eight percent had obvious diaphragmatic weakness. Males appeared to have more severe pulmonary involvement than females: at a group level, their mean VC was significantly lower than that of females (p<0.001), they used mechanical ventilation more often than females (p=0.042) and the decline over the course of the disease was significantly different between males and females (p=0.003). Apart from male gender, severe skeletal muscle weakness and long disease duration were the most important predictors of poor respiratory status. During follow-up (average 1.6 years, range 0.5-4.2 years), three patients became ventilator dependent. Annually, there were average decreases in VC in upright position of 0.9% points (p=0.09), VC in supine position of 1.2% points (p=0.049), MIP of 3.2% points (p=0.018) and MEP of 3.8% points (p<0.01). We conclude that pulmonary dysfunction in Pompe disease is much more common than generally thought. Males, patients with severe muscle weakness, and those with advanced disease duration seem most at risk.


Subject(s)
Disease Progression , Glycogen Storage Disease Type II/pathology , Glycogen Storage Disease Type II/physiopathology , Lung/physiopathology , Adolescent , Adult , Aged , Capnography , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Longitudinal Studies , Male , Middle Aged , Muscle Strength/physiology , Prognosis , Spirometry , Supine Position , Treatment Outcome , Vital Capacity/physiology , Young Adult
13.
Mol Genet Metab ; 102(1): 44-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20947400

ABSTRACT

Derivatives of 4-methylumbelliferone (4MU) are favorite substrates for the measurement of lysosomal enzyme activities in a wide variety of cell and tissue specimens. Hydrolysis of these artificial substrates at acidic pH leads to the formation of 4-methylumbelliferone, which is highly fluorescent at a pH above 10. When used for the assay of enzyme activities in dried blood spots the light emission signal can be very low due to the small sample size so that the patient and control ranges are not widely separated. We have investigated the hypothesis that quenching of the fluorescence by hemoglobin leads to appreciable loss of signal and we show that the precipitation of hemoglobin with trichloroacetic acid prior to the measurement of 4-methylumbelliferone increases the height of the output signal up to eight fold. The modified method provides a clear separation of patients' and controls' ranges for ten different lysosomal enzyme assays in dried blood spots, and approaches the conventional leukocyte assays in outcome quality.


Subject(s)
Hemoglobins/analysis , Hymecromone/analogs & derivatives , Lysosomal Storage Diseases/diagnosis , Adolescent , Adult , Case-Control Studies , Clinical Enzyme Tests , Enzyme Assays , Enzymes/blood , Fluorescence , Fractional Precipitation , Humans , Hymecromone/blood , Hymecromone/chemistry , Indicators and Reagents , Infant , Leukocytes/enzymology , Lysosomal Storage Diseases/blood , Trichloroacetic Acid/chemistry
14.
Neuromuscul Disord ; 20(12): 775-82, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20817528

ABSTRACT

Pompe disease is a rare neuromuscular disorder caused by deficiency of acid α-glucosidase. Treatment with recombinant human α-glucosidase recently received marketing approval based on prolonged survival of affected infants. The current open-label study was performed to evaluate the response in older children (age 5.9-15.2 years). The five patients that we studied had limb-girdle muscle weakness and three of them also had decreased pulmonary function in upright and supine position. They received 20-mg/kg recombinant human α-glucosidase every two weeks over a 3-year period. No infusion-associated reactions were observed. Pulmonary function remained stable (n = 4) or improved slightly (n = 1). Muscle strength increased. Only one patient approached the normal range. Patients obtained higher scores on the Quick Motor Function Test. None of the patients deteriorated. Follow-up data of two unmatched historical cohorts of adults and children with Pompe disease were used for comparison. They showed an average decline in pulmonary function of 1.6% and 5% per year. Data on muscle strength and function of untreated children were not available. Further studies are required.


Subject(s)
Glucan 1,4-alpha-Glucosidase/therapeutic use , Glycogen Storage Disease Type II/therapy , Muscle, Skeletal/physiopathology , Adolescent , Child , Child, Preschool , Enzyme Replacement Therapy , Female , Glycogen Storage Disease Type II/physiopathology , Humans , Male , Recombinant Proteins/therapeutic use , Treatment Outcome
15.
J Inherit Metab Dis ; 32(3): 416-23, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19387865

ABSTRACT

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.


Subject(s)
Acarbose/metabolism , Glucan 1,4-alpha-Glucosidase/analysis , Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/enzymology , Glycogen/metabolism , Leukocytes/enzymology , Acarbose/pharmacology , Diagnostic Techniques, Neurological , Glucan 1,4-alpha-Glucosidase/antagonists & inhibitors , Glucan 1,4-alpha-Glucosidase/metabolism , Glycogen/pharmacology , Glycogen Storage Disease Type II/blood , Glycogen Storage Disease Type II/pathology , Humans , Infant, Newborn , Leukocytes/pathology , Substrate Specificity
16.
Neuromuscul Disord ; 19(2): 113-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19084399

ABSTRACT

To determine the rate of disease progression in patients with late-onset Pompe disease, we collected longitudinal data on pulmonary function and skeletal muscle strength in 16 patients whose symptoms had started in childhood or adulthood. The mean duration of follow-up was 16 years (range 4-29 years). During the follow-up period, eight patients (50%) became wheelchair bound and three (19%) became ventilator dependent. At a group level, pulmonary function deteriorated by 1.6% per year, and proximal muscle weakness progressed gradually. At the individual level, however, the rate and extent of progression varied highly between patients. In two thirds of patients, pulmonary function and muscle strength declined simultaneously and to the same extent. The remaining one third of patients showed a variable, sometimes rapidly progressive course, leading to early respirator or wheelchair dependency. These individual differences, especially in pulmonary dysfunction, indicate the need for regular monitoring every 6-12 months depending on the rate of disease progression.


Subject(s)
Glycogen Storage Disease Type II/epidemiology , Muscle Weakness/epidemiology , Respiratory Paralysis/epidemiology , Activities of Daily Living , Adult , Age of Onset , Aged , Comorbidity , Cost of Illness , Disability Evaluation , Disease Progression , Female , Follow-Up Studies , Glycogen Storage Disease Type II/physiopathology , Humans , Male , Middle Aged , Monitoring, Physiologic/standards , Muscle Weakness/physiopathology , Muscle, Skeletal/physiopathology , Respiratory Paralysis/physiopathology , Time Factors , Ventilators, Mechanical/statistics & numerical data , Wheelchairs/statistics & numerical data
17.
J Neurol Sci ; 275(1-2): 46-50, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18757064

ABSTRACT

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.


Subject(s)
Glucan 1,4-alpha-Glucosidase/genetics , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/physiopathology , Heart Diseases/etiology , Mutation/genetics , Adult , Age Factors , Aged , Child , Electrocardiography/methods , Family Health , Female , Genotype , Heart Diseases/genetics , Humans , Male , Middle Aged , Retrospective Studies , Ultrasonography/methods
18.
Neuromuscul Disord ; 18(6): 447-52, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18508267

ABSTRACT

Pompe disease (type 2 glycogenosis, acid maltase deficiency) is a disorder affecting skeletal and cardiac muscle, caused by deficiency of acid alpha-glucosidase. In 2006 enzyme therapy with recombinant human alpha-glucosidase received marketing approval based on studies in infants. Results in older children and adults are awaited. Earlier we reported on the 3-year follow-up data of enzyme therapy in two adolescents and one adult. In the present study these patients were followed for another 5 years. Two severely affected patients, wheelchair and ventilator dependent, who had shown stabilization of pulmonary and muscle function in the first 3 years, maintained this stabilization over the 5-year extension period. In addition patients became more independent in daily life activities and quality of life improved. The third moderately affected patient had shown a remarkable improvement in muscle strength and regained the ability to walk over the first period. He showed further improvement of strength and reached normal values for age during the extension phase. The results indicate that both long-term follow-up and timing of treatment are important topics for future studies.


Subject(s)
Glycogen Storage Disease Type II/drug therapy , alpha-Glucosidases/therapeutic use , Adolescent , Adult , Animals , CHO Cells/drug effects , Child , Cricetinae , Cricetulus , Female , Glycogen Storage Disease Type II/pathology , Humans , Longitudinal Studies , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Treatment Outcome
19.
Hum Mutat ; 29(6): E27-36, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18429042

ABSTRACT

We characterized 29 unrelated patients presenting with the severe form of Pompe disease (Glycogen Storage Disease Type II, acid maltase deficiency) and identified 26 pathogenic mutations divided over 28 different genotypes. Among the eight new mutations, five were exonic point mutations (c.572A>G, c.1124G>T, c.1202A>G, c.1564C>G and c.1796C>A) leading to codon changes (p.Y191C, p.R375L, p.Q401R, p.P522A and p.S599Y); two were intronic point mutations (c.-32-3C>A and c.1636+5G>C) affecting mRNA processing; one was a single base deletion (c.742delC) generating a truncated protein (p.L248PfsX20). A comprehensive evaluation, based on different methodological approaches, confirmed the detrimental effect of the eight mutations on the protein and its function. Structural alterations potentially induced by the five missense mutations were also predicted through visual inspection of the atomic model of the GAA protein, in terms of both function and spatial orientation of specific residues as well as disturbance generated by amino acid substitutions. Although the remarkable heterogeneity of the mutational spectrum in Pompe disease was already known, our data demonstrate and confirm the power of molecular and functional analysis in predicting the natural course of Pompe disease.


Subject(s)
Glycogen Storage Disease Type II/genetics , Mutation , alpha-Glucosidases/genetics , Animals , COS Cells , Child, Preschool , Chlorocebus aethiops , DNA Mutational Analysis , Exons , Gene Deletion , Humans , Infant , Introns , Models, Molecular , Mutation, Missense , Point Mutation , alpha-Glucosidases/chemistry
20.
Mol Genet Metab ; 93(3): 275-81, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18078773

ABSTRACT

Pompe disease is an autosomal recessive disorder of glycogen metabolism caused by a deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). It presents at any age, with variable rates of progression ranging from a rapidly progressive course, often fatal by one-year of age, to a more slowly, but nevertheless relentlessly progressive course, resulting in significant morbidity and premature mortality. In infants, early initiation of enzyme replacement therapy is needed to gain the maximum therapeutic benefit, underscoring the need for early diagnosis. Several new methods for measuring GAA activity have been developed. The Pompe Disease Diagnostic Working Group met to review data generated using the new methods, and to establish a consensus regarding the application of the methods for the laboratory diagnosis of Pompe disease. Skin fibroblasts and muscle biopsy have traditionally been the samples of choice for measuring GAA activity. However, new methods using blood samples are rapidly becoming adopted because of their speed and convenience. Measuring GAA activity in blood samples should be performed under acidic conditions (pH 3.8-4.0), using up to 2 mM of the synthetic substrate 4-methylumbelliferyl-alpha-D-glucoside or glycogen (50 mg/mL), in the presence of acarbose (3-9 microM) to inhibit the isoenzyme maltase-glucoamylase. The activity of a reference enzyme should also be measured to confirm the quality of the sample. A second test should be done to support the diagnosis of Pompe disease until a program for external quality assurance and proficiency testing of the enzymatic diagnosis in blood is established.


Subject(s)
Glucan 1,4-alpha-Glucosidase/blood , Glycogen Storage Disease Type II/diagnosis , Clinical Laboratory Techniques , Humans , Infant
SELECTION OF CITATIONS
SEARCH DETAIL