Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Hum Mutat ; 40(11): 1954-1967, 2019 11.
Article in English | MEDLINE | ID: mdl-31254424

ABSTRACT

Pompe disease is an autosomal recessive lysosomal storage disorder caused by disease-associated variants in the acid alpha-glucosidase (GAA) gene. The current Pompe mutation database provides a severity rating of GAA variants based on in silico predictions and expression studies. Here, we extended the database with clinical information of reported phenotypes. We added additional in silico predictions for effects on splicing and protein function and for cross reactive immunologic material (CRIM) status, minor allele frequencies, and molecular analyses. We analyzed 867 patients and 562 GAA variants. Based on their combination with a GAA null allele (i.e., complete deficiency of GAA enzyme activity), 49% of the 422 disease-associated variants could be linked to classic infantile, childhood, or adult phenotypes. Predictions and immunoblot analyses identified 131 CRIM negative and 216 CRIM positive variants. While disease-associated missense variants were found throughout the GAA protein, they were enriched up to seven-fold in the catalytic site. Fifteen percent of disease-associated missense variants were predicted to affect splicing. This should be confirmed using splicing assays. Inclusion of clinical severity rating in the Pompe mutation database provides an invaluable tool for diagnosis, prognosis of disease progression, treatment regimens, and the future development of personalized medicine for Pompe disease.


Subject(s)
Databases, Genetic , Genetic Association Studies , Genetic Predisposition to Disease , Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/genetics , Mutation , Alleles , Computational Biology/methods , Gene Frequency , Genetic Association Studies/methods , Humans , Phenotype , Polymorphism, Single Nucleotide , Severity of Illness Index
2.
Eur J Hum Genet ; 27(6): 919-927, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30737479

ABSTRACT

Analyses in our diagnostic DNA laboratory include genes involved in autosomal recessive (AR) lysosomal storage disorders such as glycogenosis type II (Pompe disease) and mucopolysaccharidosis type I (MPSI, Hurler disease). We encountered 4 cases with apparent homozygosity for a disease-causing sequence variant that could be traced to one parent only. In addition, in a young child with cardiomyopathy, in the absence of other symptoms, a diagnosis of Pompe disease was considered. Remarkably, he presented with different enzymatic and genotypic features between leukocytes and skin fibroblasts. All cases were examined with microsatellite markers and SNP genotyping arrays. We identified one case of total uniparental disomy (UPD) of chromosome 17 leading to Pompe disease and three cases of segmental uniparental isodisomy (UPiD) causing Hurler-(4p) or Pompe disease (17q). One Pompe patient with unusual combinations of features was shown to have a mosaic segmental UPiD of chromosome 17q. The chromosome 17 UPD cases amount to 11% of our diagnostic cohort of homozygous Pompe patients (plus one case of pseudoheterozygosity) where segregation analysis was possible. We conclude that inclusion of parental DNA is mandatory for reliable DNA diagnostics. Mild or unusual phenotypes of AR diseases should alert physicians to the possibility of mosaic segmental UPiD. SNP genotyping arrays are used in diagnostic workup of patients with developmental delay. Our results show that even small Regions of Homozygosity that include telomeric areas are worth reporting, regardless of the imprinting status of the chromosome, as they might indicate segmental UPiD.


Subject(s)
Glycogen Storage Disease Type II/genetics , Mucopolysaccharidosis I/genetics , Polymorphism, Single Nucleotide , Uniparental Disomy , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male
3.
PLoS One ; 13(12): e0208854, 2018.
Article in English | MEDLINE | ID: mdl-30532252

ABSTRACT

The majority of children and adults with Pompe disease in the population of European descent carry the leaky splicing GAA variant c.-32-13T>G (IVS1) in combination with a fully deleterious GAA variant on the second allele. The phenotypic spectrum of this patient group is exceptionally broad, with symptom onset ranging from early infancy to late adulthood. In addition, the response to enzyme replacement therapy (ERT) varies between patients. The insertion/deletion (I/D) polymorphism of the angiotensin I-converting enzyme (ACE) has been suggested to be a modifier of disease onset and/or response to ERT. Here, we have investigated the effect of the ACE I/D polymorphism in a relatively large cohort of 131 children and adults with Pompe disease, of whom 112 were followed during treatment with ERT for 5 years. We assessed the use of wheelchair and mechanical ventilation, muscle strength assessed via manual muscle testing and hand-held dynamometry (HHD), distance walked on the six-minute walk test (6MWT), forced vital capacity (FVC) in sitting and supine position and daily-life activities assessed by R-PAct. Cross sectional analysis at first visit showed no differences between the genotypes with respect to age at first symptoms, diagnosis, wheelchair use, or ventilator use. Also response to ERT over 5 years assessed by linear mixed model analyses showed no significant differences between ACE groups for any of the outcome measures. The patient cohort contained 24 families with 54 siblings. Differences in ACE genotype could neither explain inter nor intra familial differences. We conclude that the ACE I/D polymorphism does not explain the large variation in disease severity and response to ERT observed among Pompe patients with the same c.-32-13T>G GAA variant.


Subject(s)
Enzyme Replacement Therapy , Glycogen Storage Disease Type II , Models, Biological , Peptidyl-Dipeptidase A , Polymorphism, Genetic , Adolescent , Adult , Age Factors , Aged , Child , Child, Preschool , Female , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/physiopathology , Humans , Infant , Infant, Newborn , Male , Middle Aged , Muscle Strength , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Peptidyl-Dipeptidase A/therapeutic use , Walking
5.
Dev Med Child Neurol ; 59(10): 1063-1070, 2017 10.
Article in English | MEDLINE | ID: mdl-28543354

ABSTRACT

AIM: Mucopolysaccharidosis type II (MPS II) is caused by variants in the iduronate-2-sulphatase gene (IDS). Patients can be either neuronopathic with intellectual disability, or non-neuronopathic. Few studies have reported on the IDS genotype-phenotype relationship and on the molecular effects involved. We addressed this in a cohort study of Dutch patients with MPS II. METHOD: Intellectual performance was assessed for school performance, behaviour, and intelligence. Urinary glycosaminoglycans were quantified by mass spectrometry. IDS variants were analysed in expression studies for enzymatic activity and processing by immunoblotting. RESULTS: Six patients had a non-neuronopathic phenotype and 11 a neuronopathic phenotype, three of whom had epilepsy. Total deletion of IDS invariably resulted in the neuronopathic phenotype. Phenotypes of seven known IDS variants were consistent with the literature. Expression studies of nine variants were novel and showed impaired IDS enzymatic activity, aberrant intracellular processing, and elevated urinary excretion of heparan sulphate and dermatan sulphate irrespective of the MPS II phenotype. INTERPRETATION: We speculate that very low or cell-type-specific IDS residual activity is sufficient to prevent the neuronal phenotype of MPS II. Whereas the molecular effects of IDS variants do not distinguish between MPS II phenotypes, the IDS genotype is a strong predictor.


Subject(s)
Genetic Variation , Glycoproteins/genetics , Glycoproteins/metabolism , Mucopolysaccharidosis II/genetics , Mucopolysaccharidosis II/psychology , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Educational Status , Epilepsy/enzymology , Epilepsy/genetics , Epilepsy/psychology , Genetic Association Studies , Glycosaminoglycans/urine , Humans , Immunoblotting , Intelligence , Mass Spectrometry , Middle Aged , Mucopolysaccharidosis II/drug therapy , Mucopolysaccharidosis II/metabolism , Netherlands , Phenotype , Young Adult
6.
Genet Med ; 19(1): 90-97, 2017 01.
Article in English | MEDLINE | ID: mdl-27362911

ABSTRACT

PURPOSE: To determine the effect of antibodies against recombinant human acid α-glucosidase (rhGAA) on treatment efficacy and safety, and to test whether the GAA genotype is involved in antibody formation. METHODS: We used enzyme-linked immunosorbent assay (ELISA) to determine anti-rhGAA antibody titers at baseline and at 6, 12, and 36 months of rhGAA treatment. We measured the capacity of antibodies to neutralize rhGAA enzymatic activity or cellular uptake and the effects on infusion-associated reactions (IARs), muscle strength, and pulmonary function. RESULTS: Of 73 patients, 45 developed antibodies. Maximal titers were high (≥1:31,250) in 22% of patients, intermediate (1:1,250-1:31,250) in 40%, and none or low (0-1:1,250) in 38%. The common IVS1/delex18 GAA genotype was absent only in the high-titer group. The height of the titer positively correlated with the occurrence and number of IARs (P ≤ 0.001). On the group level, antibody titers did not correlate with treatment efficacy. Eight patients (11%) developed very high maximal titers (≥156,250), but only one patient showed high sustained neutralizing antibodies that probably interfered with treatment efficacy. CONCLUSIONS: In adults with Pompe disease, antibody formation does not interfere with rhGAA efficacy in the majority of patients, is associated with IARs, and may be attenuated by the IVS1/delex18 GAA genotype.Genet Med 19 1, 90-97.


Subject(s)
Antibody Formation/immunology , Glycogen Storage Disease Type II/immunology , Recombinant Proteins/administration & dosage , alpha-Glucosidases/administration & dosage , Adult , Aged , Antibody Formation/genetics , Enzyme Replacement Therapy/adverse effects , Female , Genotype , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Humans , Male , Middle Aged , Recombinant Proteins/adverse effects , Recombinant Proteins/genetics , Recombinant Proteins/immunology , alpha-Glucosidases/adverse effects , alpha-Glucosidases/genetics , alpha-Glucosidases/immunology
8.
Hum Mutat ; 36(1): 57-68, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25243733

ABSTRACT

Identification of pathogenic variants in monogenic diseases is an important aspect of diagnosis, genetic counseling, and prediction of disease severity. Pathogenic mechanisms involved include changes in gene expression, RNA processing, and protein translation. Variants affecting pre-mRNA splicing are difficult to predict due to the complex mechanism of splicing regulation. A generic approach to systematically detect and characterize effects of sequence variants on splicing would improve current diagnostic practice. Here, it is shown that such approach is feasible by combining flanking exon RT-PCR, sequence analysis of PCR products, and exon-internal quantitative RT-PCR for all coding exons. Application of this approach to one novel and six previously published variants in the acid-alpha glucosidase (GAA) gene causing Pompe disease enabled detection of a total of 11 novel splicing events. Aberrant splicing included cryptic splice-site usage, intron retention, and exon skipping. Importantly, the extent of leaky wild-type splicing correlated with disease onset and severity. These results indicate that this approach enables sensitive detection and in-depth characterization of variants affecting splicing, many of which are still unrecognized or poorly understood. The approach is generic and should be adaptable for application to other monogenic diseases to aid in improved diagnostics.


Subject(s)
Glycogen Storage Disease Type II/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction/methods , alpha-Glucosidases/genetics , Adolescent , Adult , Child , Exons , Humans , Infant , Infant, Newborn , Introns , Middle Aged , Mutation , RNA Splicing
9.
J Inherit Metab Dis ; 38(2): 305-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24715333

ABSTRACT

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.


Subject(s)
Antibodies/blood , Enzyme Replacement Therapy , Glycogen Storage Disease Type II/drug therapy , alpha-Glucosidases/therapeutic use , Age Factors , Biomarkers/blood , Cells, Cultured , Child, Preschool , Disease Progression , Female , Genetic Predisposition to Disease , Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/immunology , Glycogen Storage Disease Type II/mortality , Humans , Infant , Infant, Newborn , Male , Mutation , Netherlands , Phenotype , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Risk Factors , Time Factors , Transfection , Treatment Outcome , alpha-Glucosidases/deficiency , alpha-Glucosidases/genetics , alpha-Glucosidases/immunology
10.
Orphanet J Rare Dis ; 8: 51, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23557332

ABSTRACT

BACKGROUND: Mucopolysaccharidosis type VI (Maroteaux-Lamy syndrome; MPS VI) is an autosomal recessive lysosomal storage disorder in which deficiency of N-acetylgalactosamine 4-sulfatase (arylsulfatase B; ARSB) leads to the storage of glycosaminoglycans (GAGs) in connective tissue. The genotype-phenotype correlation has been addressed in several publications but the picture is not complete. Since 2007, enzyme-replacement therapy (ERT) has been available for patients with MPS VI in the Netherlands. The purpose of our study was to learn more about the genotype-phenotype correlations in MPS VI and the antibody response to ERT with galsulfase (recombinant human arylsulfatase B). METHODS: We identified ARSB mutations in 12 patients and used site-directed mutagenesis to study their effect. Antibody levels to galsulfase were measured using ELISA and a semi-quantitative immunoprecipitation method. We assessed the in vitro inhibitory effect of antibodies on galsulfase uptake and their effect on clinical outcome. RESULTS: Five patients had a rapidly progressive phenotype and seven a slowly progressive phenotype. In total 9 pathogenic mutations were identified including 4 novel mutations (N301K, V332G, A237D, and c.1142 + 2 T > C) together composing 8 pathogenic genotypes. Most mutations appeared not to affect the synthesis of ARSB (66 kD precursor), but to hamper its maturation (43 kD ARSB). Disease severity was correlated with urinary GAG excretion. All patients developed antibodies to galsulfase within 26 weeks of treatment. It was demonstrated that these antibodies can inhibit the uptake of galsulfase in vitro. CONCLUSIONS: The clinical phenotypes and the observed defects in the biosynthesis of ARSB show that some of the mutations that we identified are clearly more severe than others. Patients receiving galsulfase as enzyme-replacement therapy can develop antibodies towards the therapeutic protein. Though most titers are modest, they can exceed a level at which they potentially affect the clinical outcome of enzyme-replacement therapy.


Subject(s)
Antibody Formation/immunology , Mucopolysaccharidosis VI/genetics , Mucopolysaccharidosis VI/pathology , N-Acetylgalactosamine-4-Sulfatase/immunology , Adolescent , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Genotype , Humans , Immunoprecipitation , Infant , Male , Mucopolysaccharidosis VI/immunology , Mutagenesis, Site-Directed , Phenotype , Recombinant Proteins/immunology
11.
JIMD Rep ; 7: 39-48, 2013.
Article in English | MEDLINE | ID: mdl-23430493

ABSTRACT

Pompe disease (PD) is a recessive metabolic disorder characterized by acid α-glucosidase (GAA) deficiency, which results in lysosomal accumulation of glycogen in all tissues, especially in skeletal muscles. PD clinical course is mainly determined by the nature of the GAA mutations. Although ~400 distinct GAA sequence variations have been described, the genotype-phenotype correlation is not always evident.In this study, we describe the first clinical and genetic analysis of Colombian PD patients performed in 11 affected individuals. GAA open reading frame sequencing revealed eight distinct mutations related to PD etiology including two novel missense mutations, c.1106 T > C (p.Leu369Pro) and c.2236 T > C (p.Trp746Arg). In vitro functional studies showed that the structural changes conferred by both mutations did not inhibit the synthesis of the 110 kD GAA precursor form but affected the processing and intracellular transport of GAA. In addition, analysis of previously described variants located at this position (p.Trp746Gly, p.Trp746Cys, p.Trp746Ser, p.Trp746X) revealed new insights in the molecular basis of PD. Notably, we found that p.Trp746Cys mutation, which was previously described as a polymorphism as well as a causal mutation, displayed a mild deleterious effect. Interestingly and by chance, our study argues in favor of a remarkable Afro-American and European ancestry of the Colombian population. Taken together, our report provides valuable information on the PD genotype-phenotype correlation, which is expected to facilitate and improve genetic counseling of affected individuals and their families.

12.
Orphanet J Rare Dis ; 7: 88, 2012 Nov 12.
Article in English | MEDLINE | ID: mdl-23147228

ABSTRACT

BACKGROUND: Due partly to physicians' unawareness, many adults with Pompe disease are diagnosed with great delay. Besides, it is not well known which factors influence the rate of disease progression, and thus disease outcome. We delineated the specific clinical features of Pompe disease in adults, and mapped out the distribution and severity of muscle weakness, and the sequence of involvement of the individual muscle groups. Furthermore, we defined the natural disease course and identified prognostic factors for disease progression. METHODS: We conducted a single-center, prospective, observational study. Muscle strength (manual muscle testing, and hand-held dynamometry), muscle function (quick motor function test), and pulmonary function (forced vital capacity in sitting and supine positions) were assessed every 3-6 months and analyzed using repeated-measures ANOVA. RESULTS: Between October 2004 and August 2009, 94 patients aged between 25 and 75 years were included in the study. Although skeletal muscle weakness was typically distributed in a limb-girdle pattern, many patients had unfamiliar features such as ptosis (23%), bulbar weakness (28%), and scapular winging (33%). During follow-up (average 1.6 years, range 0.5-4.2 years), skeletal muscle strength deteriorated significantly (mean declines of -1.3% point/year for manual muscle testing and of -2.6% points/year for hand-held dynamometry; both p<0.001). Longer disease duration (>15 years) and pulmonary involvement (forced vital capacity in sitting position <80%) at study entry predicted faster decline. On average, forced vital capacity in supine position deteriorated by 1.3% points per year (p=0.02). Decline in pulmonary function was consistent across subgroups. Ten percent of patients declined unexpectedly fast. CONCLUSIONS: Recognizing patterns of common and less familiar characteristics in adults with Pompe disease facilitates timely diagnosis. Longer disease duration and reduced pulmonary function stand out as predictors of rapid disease progression, and aid in deciding whether to initiate enzyme replacement therapy, or when.


Subject(s)
Glycogen Storage Disease Type II/diagnosis , Adult , Aged , Disease Progression , Female , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/enzymology , Glycogen Storage Disease/pathology , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/pathology , Humans , Male , Middle Aged , Prospective Studies , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
13.
Mol Genet Metab ; 107(3): 485-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23000108

ABSTRACT

INTRODUCTION: Most adults with Pompe disease are compound heterozygotes in which one acid α-glucosidase (GAA) allele harbors the c.-32-13T>G mutation, causing partial loss of GAA, and the other allele harbors a fully deleterious mutation. The fibroblast GAA activity in these patients is usually between 5% and 25% of the average in healthy individuals. In some adult patients, however, the fibroblast GAA activity is much lower and is in the range that is normally observed in classic-infantile Pompe disease. We investigated the genotype-phenotype correlation in three such adult patients and measured the GAA activity as well as the glycogen content in muscle and fibroblasts in order to better understand the clinical course. METHODS: DNA was sequenced and GAA activity and glycogen content were measured in leukocytes, fibroblasts and muscle. Muscle biopsies were microscopically analyzed and the biosynthesis of GAA in fibroblasts was analyzed by immunoblotting. GAA activity and glycogen content in fibroblasts and muscle tissue in healthy controls, adult patients with Pompe disease and classic-infantile patients were compared with those of the three index patients. RESULTS: One patient had genotype c.525delT/c.671G>A (r.0/p.Arg224Gln). Two affected brothers had genotype c.569G>A/c.1447G>A (p.Arg190His/p.Gly483Arg). In all three cases the GAA activity and the glycogen content in fibroblasts were within the same range as in classic-infantile Pompe disease, but the activity and glycogen content in muscle were both within the adult range. In fibroblasts, the first step of GAA synthesis appeared unaffected but lysosomal forms of GAA were not detectable with immunoblotting. CONCLUSION: Some adult patients with mutations other than c.-32-13T>G can have very low GAA activity in fibroblasts but express higher activity in muscle and store less glycogen in muscle than patients with classic-infantile Pompe disease. This might explain why these patients have a slowly progressive course of Pompe disease.


Subject(s)
Fibroblasts/enzymology , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/genetics , Glycogen/metabolism , Muscle, Skeletal/enzymology , alpha-Glucosidases/metabolism , Adult , Alleles , Fibroblasts/pathology , Genetic Association Studies , Genotype , Glycogen Storage Disease Type II/pathology , Heterozygote , Humans , Infant, Newborn , Male , Middle Aged , Muscle, Skeletal/pathology , Mutation , Phenotype , alpha-Glucosidases/genetics
14.
Hum Mutat ; 33(8): 1161-5, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22644586

ABSTRACT

Pompe disease is an autosomal recessive lysosomal glycogen storage disorder, characterized by progressive muscle weakness. Deficiency of acid α-glucosidase (EC; 3.2.1.20/3) can be caused by numerous pathogenic variants in the GAA gene. The Pompe Disease Mutation Database at http://www.pompecenter.nl aims to list all variants and their effect. This update reports on 94 variants. We examined 35 novel and 34 known mutations by site-directed mutagenesis and transient expression in COS-7 cells or HEK293T cells. Each of these mutations was given a severity rating using a previously published system, based on the level of acid α-glucosidase activity in medium and transfected cells and on the quantity and quality of the different molecular mass species in the posttranslational modification and transport of acid α-glucosidase. This approach enabled to classify 55 missense mutations as pathogenic and 13 as likely nonpathogenic. Based on their nature and the use of in silico analysis (Alamut® software), 12 of the additional 25 novel mutations were predicted to be pathogenic including 4 splicing mutations, 6 mutations leading to frameshift, and 2 point mutations causing stop codons. Seven of the additional mutations were considered nonpathogenic (4 silent and 3 occurring in intron regions), and 6 are still under investigation.


Subject(s)
Databases, Genetic , Glycogen Storage Disease Type II/genetics , alpha-Glucosidases/genetics , Genetic Predisposition to Disease , Humans , Mutation
15.
Am J Med Genet C Semin Med Genet ; 160C(1): 59-68, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22253258

ABSTRACT

Pompe disease is an autosomal recessive lysosomal glycogen storage disorder that is caused by acid α-glucosidase (GAA) deficiency and is due to pathogenic sequence variations in the corresponding GAA gene. The correlation between genotypes and phenotypes is strict, in that patients with the most severe phenotype, classic infantile Pompe disease, have two pathogenic mutations, one in each GAA allele, that prevent the formation of GAA or totally obliterates its function. All patients with less progressive phenotypes have at least one sequence variation that allows normal or low level synthesis of GAA leading to the formation of analytically measurable, low level GAA activity in most cases. There is an overall trend of finding higher GAA enzyme levels in patients with onset of symptoms in adulthood when compared to patients who show clinical manifestations in early childhood, aged 0-5 years, with a rapidly progressive course, but who lack the severe characteristics of classic infantile Pompe disease. However, several cases have been reported of adult-onset disease with very low GAA activity, which in all those cases corresponds with the GAA genotype. The clinical diversity observed within a large group of patients with functionally the same GAA genotype and the same c.-32-13C > T haplotype demonstrates that modifying factors can have a substantial effect on the clinical course of Pompe disease, disturbing the GAA genotype-phenotype correlation. The present day challenge is to identify these factors and explore them as therapeutic targets.


Subject(s)
Genetic Association Studies , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/metabolism , Glycogen/metabolism , alpha-Glucosidases/genetics , Age of Onset , Glycogen Storage Disease Type II/pathology , Haplotypes , Humans , Mutation , Structure-Activity Relationship , alpha-Glucosidases/metabolism
16.
Mol Genet Metab ; 103(1): 12-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21320792

ABSTRACT

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.


Subject(s)
Clinical Enzyme Tests/methods , Glycogen Storage Disease Type II/blood , Glycogen Storage Disease Type II/diagnosis , Hematologic Tests/methods , Neonatal Screening , alpha-Glucosidases/blood , Adult , Child , Fibroblasts/metabolism , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/genetics , Homozygote , Humans , Infant , Infant, Newborn , alpha-Glucosidases/deficiency , alpha-Glucosidases/genetics
17.
Mol Genet Metab ; 101(4): 338-45, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20826098

ABSTRACT

Clinical trials have demonstrated beneficial effects of enzyme replacement therapy (ERT) with alglucosidase alfa in infants, children and adults with Pompe disease. Recent studies have shown that high antibody titers can occur in patients receiving ERT and counteract the effect of treatment. This particularly occurs in those patients with classic-infantile Pompe disease that do not produce any endogenous acid α-glucosidase (CRIM-negative). It is still unclear to what extent antibody formation affects the outcome of ERT in adults with residual enzyme activity. We present the case of a patient with adult-onset Pompe disease. He was diagnosed at the age of 39years by enzymatic testing (10.7% residual activity in fibroblasts) and DNA analysis (genotype: c.-32-13T>G/p.Trp516X). Infusion-associated reactions occurred during ERT and the patient's disease progressed. Concurrently, the antibody titer rose to a similarly high level as reported for some CRIM-negative patients with classic-infantile Pompe disease. Using newly developed immunologic-assays we could calculate that approximately 40% of the administered alglucosidase alfa was captured by circulating antibodies. Further, we could demonstrate that uptake of alglucosidase alfa by cultured fibroblasts was inhibited by admixture of the patient's serum. This case demonstrates that also patients with an appreciable amount of properly folded and catalytically active endogenous acid α-glucosidase can develop antibodies against alglucosidase alfa that affect the response to ERT.


Subject(s)
Antibodies/blood , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/immunology , alpha-Glucosidases/immunology , alpha-Glucosidases/therapeutic use , Adult , Antibodies/immunology , Enzyme Replacement Therapy , Female , Fibroblasts/drug effects , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/physiopathology , Humans , Immunologic Tests , Male , Middle Aged , Muscle Strength/physiology , Respiratory Function Tests , Treatment Outcome , alpha-Glucosidases/adverse effects
18.
Hum Mutat ; 31(7): 858-65, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20578233

ABSTRACT

The molecular genetic diagnosis of inherited metabolic disorders is challenging. The diseases are rare, and most show locus heterogeneity. Hence, testing of the genes associated with IMDs is time consuming and often not easily available. We report a resequencing array that allows the simultaneous resequencing of up to 92 genes associated with IMDs. To validate the array, DNA samples from 51 patients with 52 different known variants (including point variants, small insertion, and deletions [indels]) in seven genes (C14ORF133, GAA, NPC1, NPC2, VPS33B, WFS1, and SLC19A2) were amplified by PCR and hybridized to the array. A further patient cohort with 48 different mutations in NPC1 were analyzed blind. Out of 76 point variants, 73 were identified using automated software analysis followed by manual review. Ten insertion and deletion variants were detected in the extra tiling using mutation specific probes, with 11 heterozygous deletions and 3 heterozygous insertions. In summary, we identified 96% (95% confidence interval [CI] 89-99%) of point variants added to the array, but the pickup rate reduced to 83% (95% CI 75-89%) when insertions/deletions were included. Although the methodology has strengths and weaknesses, application of this technique could expedite diagnosis in most patients with multilocus IMDs.


Subject(s)
Metabolic Diseases/genetics , Mutation , Oligonucleotide Array Sequence Analysis/methods , Sequence Analysis, DNA/methods , Carrier Proteins/genetics , Genetic Predisposition to Disease , Glycoproteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Metabolic Diseases/diagnosis , Niemann-Pick C1 Protein , Polymerase Chain Reaction , Reproducibility of Results , Research Design , Vesicular Transport Proteins/genetics , alpha-Glucosidases/genetics
19.
Blood ; 115(26): 5329-37, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20385789

ABSTRACT

Pompe disease (acid alpha-glucosidase deficiency) is a lysosomal glycogen storage disorder characterized in its most severe early-onset form by rapidly progressive muscle weakness and mortality within the first year of life due to cardiac and respiratory failure. Enzyme replacement therapy prolongs the life of affected infants and supports the condition of older children and adults but entails lifelong treatment and can be counteracted by immune responses to the recombinant enzyme. We have explored the potential of lentiviral vector-mediated expression of human acid alpha-glucosidase in hematopoietic stem cells (HSCs) in a Pompe mouse model. After mild conditioning, transplantation of genetically engineered HSCs resulted in stable chimerism of approximately 35% hematopoietic cells that overexpress acid alpha-glucosidase and in major clearance of glycogen in heart, diaphragm, spleen, and liver. Cardiac remodeling was reversed, and respiratory function, skeletal muscle strength, and motor performance improved. Overexpression of acid alpha-glucosidase did not affect overall hematopoietic cell function and led to immune tolerance as shown by challenge with the human recombinant protein. On the basis of the prominent and sustained therapeutic efficacy without adverse events in mice we conclude that ex vivo HSC gene therapy is a treatment option worthwhile to pursue.


Subject(s)
Genetic Therapy/methods , Glycogen Storage Disease Type II/therapy , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , alpha-Glucosidases/genetics , Animals , Cells, Cultured , Chimerism , Gene Expression , Genetic Vectors/genetics , Glycogen/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic System/metabolism , Humans , Mice , Mice, Knockout , Motor Activity , Transduction, Genetic
20.
J Hum Genet ; 54(6): 324-30, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19343043

ABSTRACT

To elucidate the mechanism underlying transport and processing defects from the viewpoint of enzyme folding, we constructed three-dimensional models of human acid alpha-glucosidase encompassing 27 relevant amino acid substitutions by means of homology modeling. Then, we determined in each separate case the number of affected atoms, the root-mean-square distance value and the solvent-accessible surface area value. The analysis revealed that the amino acid substitutions causing a processing or transport defect responsible for Pompe disease were widely spread over all of the five domains comprising the acid alpha-glucosidase. They were distributed from the core to the surface of the enzyme molecule, and the predicted structural changes varied from large to very small. Among the structural changes, we paid particular attention to G377R and G483R. These two substitutions are predicted to cause electrostatic changes in neighboring small regions on the molecular surface. The quality control system of the endoplasmic reticulum apparently detects these very small structural changes and degrades the mutant enzyme precursor (G377R), but also the cellular sorting system might be misled by these minor changes whereby the precursor is secreted instead of being transported to lysosomes (G483R).


Subject(s)
Glycogen Storage Disease Type II/enzymology , Mutant Proteins/chemistry , Protein Processing, Post-Translational , Protein Transport , alpha-Glucosidases/chemistry , Amino Acid Sequence , Amino Acid Substitution , Endoplasmic Reticulum/metabolism , Glycogen Storage Disease Type II/genetics , Humans , Lysosomes/metabolism , Models, Molecular , Molecular Sequence Data , Mutant Proteins/metabolism , Protein Conformation , Sequence Homology, Amino Acid , alpha-Glucosidases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...