Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Mol Genet Metab ; 109(4): 377-81, 2013 Aug.
Article En | MEDLINE | ID: mdl-23786846

Three major clinical subgroups are usually distinguished in Mucopolysaccharidosis type I: Hurler (MPS IH, severe presentation), Hurler-Scheie (MPS IH/S, intermediate) and Scheie (MPS IS, mild). To facilitate treatment with hematopoietic stem-cell transplantation, early diagnosis is important for MPS IH patients. Although screening for MPS I in newborns would allow detection at an early age, it may be difficult to predict the phenotype on the basis of the genotype in these infants. Extra diagnostic tools are thus required. Based on the hypothesis that distinct MPS I phenotypes may result from differences in residual α-l-iduronidase (IDUA) activity, we modified the common IDUA assay using the substrate 4-methylumbelliferyl-α-l-iduronide to allow quantification of low IDUA activity in MPS I fibroblasts. Enzyme incubation was performed with high protein concentrations at different time points up to 8h. Mean residual IDUA activity was 0.18% (range 0-0.6) of the control value in MPS IH fibroblasts (n=5); against 0.27% (range 0.2-0.3) in MPS IH/S cells (n=3); and 0.79% (range 0.3-1.8) in MPS IS fibroblasts (n=5). These results suggest that residual IDUA activity and severity of the MPS I phenotype are correlated. Two MPS IS patients with rare (E276K/E276K) or indefinite (A327P/unknown) IDUA genotypes had residual IDUA activity in the MPS IS range, illustrating the usefulness of our approach. IDUA(E276K) was very unstable at 37°C, but more stable at 23°C, suggesting thermal instability. We conclude that this procedure for determining residual IDUA activity in fibroblasts of MPS I patients may be helpful to predict MPS I phenotype.


Fibroblasts/enzymology , Hymecromone/analogs & derivatives , Iduronidase/metabolism , Mucopolysaccharidosis I/diagnosis , Cell Line , Early Diagnosis , Humans , Hymecromone/metabolism , Infant, Newborn , Mucopolysaccharidosis I/enzymology , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology , Mutation
2.
J Inherit Metab Dis ; 36(2): 293-307, 2013 Mar.
Article En | MEDLINE | ID: mdl-23371450

Mucopolysaccharidosis IVA (MPS IVA; Morquio A syndrome) is an autosomal recessive lysosomal storage disorder resulting from a deficiency of N-acetylgalactosamine-6-sulfate sulfatase (GALNS) activity. Diagnosis can be challenging and requires agreement of clinical, radiographic, and laboratory findings. A group of biochemical genetics laboratory directors and clinicians involved in the diagnosis of MPS IVA, convened by BioMarin Pharmaceutical Inc., met to develop recommendations for diagnosis. The following conclusions were reached. Due to the wide variation and subtleties of radiographic findings, imaging of multiple body regions is recommended. Urinary glycosaminoglycan analysis is particularly problematic for MPS IVA and it is strongly recommended to proceed to enzyme activity testing even if urine appears normal when there is clinical suspicion of MPS IVA. Enzyme activity testing of GALNS is essential in diagnosing MPS IVA. Additional analyses to confirm sample integrity and rule out MPS IVB, multiple sulfatase deficiency, and mucolipidoses types II/III are critical as part of enzyme activity testing. Leukocytes or cultured dermal fibroblasts are strongly recommended for enzyme activity testing to confirm screening results. Molecular testing may also be used to confirm the diagnosis in many patients. However, two known or probable causative mutations may not be identified in all cases of MPS IVA. A diagnostic testing algorithm is presented which attempts to streamline this complex testing process.


Glycosaminoglycans/urine , Mucopolysaccharidosis IV/diagnosis , Mucopolysaccharidosis IV/enzymology , Algorithms , Fibroblasts/enzymology , Humans , Leukocytes/enzymology , Mucolipidoses/diagnosis , Mucopolysaccharidosis IV/genetics , Mucopolysaccharidosis IV/urine , Multiple Sulfatase Deficiency Disease/diagnosis , Mutation , Pathology, Molecular/methods
3.
Curr Protoc Hum Genet ; 79: 17.14.1-17.14.9, 2013 Oct 18.
Article En | MEDLINE | ID: mdl-24510650

Mucopolysaccharidosis type II (MPS II) is an X-linked lysosomal storage disorder caused by a deficiency of iduronate 2-sulfatase (IDS). Progressive, intralysosomal accumulation of the glycosaminoglycans (GAGs) dermatan and heparan sulfate in almost all tissues leads to multi-organ involvement in affected males but to virtual absence of symptoms in heterozygote female carriers due to preferential inactivation of the mutant allele. Diagnosis of MPS II in males is based on IDS analysis in leukocytes, fibroblasts, plasma, or dried blood spots (DBS), whereas IDS activities may be within the normal range in heterozygote females. The advent of fluorometric and mass spectrometry methods for enzyme analysis in DBS has simplified the diagnostic approach for MPS II males. Molecular analysis of the IDS gene confirms the diagnosis of MPS II in males and is the only diagnostic test to confirm carrier status in females. This unit provides detailed analytical protocols for measurement of IDS activity in DBS and plasma using a fluorometric assay.


Dried Blood Spot Testing/methods , Fluorometry/methods , Glycoproteins/blood , Mucopolysaccharidosis II/diagnosis , Dermatan Sulfate/metabolism , Female , Glycoproteins/deficiency , Glycoproteins/genetics , Heparitin Sulfate/metabolism , Humans , Lysosomes/pathology , Male , Mucopolysaccharidosis II/genetics
4.
Mol Genet Metab ; 104(1-2): 144-8, 2011.
Article En | MEDLINE | ID: mdl-21831684

Dried blood spot (DBS) methods are currently available for identification of a range of lysosomal storage disorders (LSDs). These disorders are generally characterized by a deficiency of activity of a lysosomal enzyme and by a broad spectrum of phenotypes. Diagnosis of LSD patients is often delayed, which is of particular concern as therapeutic outcomes (e.g. enzyme replacement therapy) are generally more favorable in early disease stages. Experts in the field of LSDs diagnostics and screening programs convened and reviewed experiences with the use of DBS methods, and discuss the diagnostic challenges, possible applications and quality programs in this paper. Given the easy sampling and shipping and stability of samples, DBS has evident advantages over other laboratory methods and can be particularly helpful in the early identification of affected LSD patients through neonatal screening, high-risk population screening or family screening.


Dried Blood Spot Testing/methods , Dried Blood Spot Testing/trends , Lysosomal Storage Diseases/blood , Lysosomal Storage Diseases/diagnosis , Dried Blood Spot Testing/standards , Humans , Quality Control , Reference Standards , Research Report
5.
Ann Neurol ; 68(6): 876-87, 2010 Dec.
Article En | MEDLINE | ID: mdl-21061399

OBJECTIVE: Mucopolysaccharidosis (MPS) IIIA (Sanfilippo syndrome type A) is a lysosomal storage disorder caused by deficiency of the enzyme sulfamidase. Information on the natural course of MPS IIIA is scarce, but is much needed in view of emerging therapies. METHODS: Clinical history and molecular defects of all 110 MPS IIIA patients identified by enzymatic studies in the Netherlands were collected and included in this study. RESULTS: First clinical signs, mainly consisting of delayed speech development and behavioral problems, were noted between the ages of 1 and 6 years. Other symptoms included sleeping and hearing problems, recurrent upper airway infections, diarrhea, and epilepsy. The clinical course varied remarkably and could be correlated with the molecular defects. The frequent pathogenic mutations p.R245H, p.Q380R, p.S66W, and c.1080delC were associated with the classical severe phenotype. Patients compound heterozygous for the p.S298P mutation in combination with 1 of the mutations associated with the classical severe phenotype had a significantly longer preservation of psychomotor functions and a longer survival. Two patients homozygous for the p.S298P mutation, and 4 patients from 3 families heterozygous for 3 missense variants not reported previously (p.T421R, p.P180L, and p.L12Q), showed a remarkably attenuated phenotype. INTERPRETATION: We report the natural history and mutational analysis in a large unbiased cohort of MPS IIIA patients. We demonstrate that the clinical spectrum of MPS IIIA is much broader than previously reported. A significant genotype-phenotype correlation was established in this cohort.


Genetic Association Studies , Genotype , Phenotype , Adolescent , Adult , Behavioral Symptoms/etiology , Cells, Cultured , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Epilepsy/etiology , Female , Fibroblasts , Hearing Disorders/etiology , Humans , Hydrolases/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Mucopolysaccharidosis III/complications , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Mutation/genetics , Pregnancy , Regression Analysis , Severity of Illness Index , Skin/pathology , Sleep Wake Disorders/etiology , Vision Disorders/etiology , Young Adult
6.
Hum Mutat ; 31(5): E1348-60, 2010 May.
Article En | MEDLINE | ID: mdl-20232353

Mucopolysaccharidosis III D (Sanfilippo disease type D, MPS IIID) is a rare autosomal recessive lysosomal storage disorder previously described in only 20 patients. MPS IIID is caused by a deficiency of N-acetylglucosamine-6-sulphate sulphatase (GNS), one of the enzymes required for the degradation of heparan sulphate. So far only seven mutations in the GNS gene have been reported. The clinical phenotype of 12 new MPS IIID patients from 10 families was studied. Mutation analysis of GNS was performed in 16 patients (14 index cases). Clinical signs and symptoms of the MPS IIID patients appeared to be similar to previously described patients with MPS III. Early development was normal with onset of behavioral problems around the age of 4 years, followed by developmental stagnation, deterioration of verbal communication and subsequent deterioration of motor functions. Sequence analysis of the coding regions of the gene encoding GNS (GNS) resulted in the identification of 15 novel mutations: 3 missense mutations, 1 nonsense mutation, 4 splice site mutations, 3 frame shift mutations, 3 large deletions and 1 in-frame small deletion. They include the first missense mutations and a relatively high proportion of large rearrangements, which warrants the inclusion of quantitative techniques in routine mutation screening of the GNS gene.


Mucopolysaccharidosis III/genetics , Sulfatases/deficiency , Sulfatases/genetics , Adolescent , Adult , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Male , Mutation/genetics , Phenotype , Young Adult
7.
Hum Mutat ; 30(6): 918-25, 2009 Jun.
Article En | MEDLINE | ID: mdl-19479962

Mucopolysaccharidosis (MPS) type IIIC or Sanfilippo syndrome type C is a rare autosomal recessive disorder caused by the deficiency of the lysosomal membrane enzyme, heparan sulfate acetyl-CoA (AcCoA): alpha-glucosaminide N-acetyltransferase (HGSNAT; EC 2.3.1.78), which catalyzes transmembrane acetylation of the terminal glucosamine residues of heparan sulfate prior to their hydrolysis by alpha-N-acetylglucosaminidase. Lysosomal storage of undegraded heparan sulfate in the cells of affected patients leads to neuronal death, causing neurodegeneration and severely impaired development accompanied by mild visceral and skeletal abnormalities, including mild dwarfism, coarse facies, and joint stiffness. To date, 50 HGSNAT mutations have been identified in MPS IIIC patients: 40 were previously published and 10 novel mutations are reported here. The mutations span the entire structure of the gene and include 13 splice-site mutations, 11 insertions and deletions, 8 nonsense mutations, and 18 missense mutations (http://chromium.liacs.nl/LOVD2/home.php?select_db=HGSNAT). In addition, four polymorphisms result in amino acid changes that do not affect activity of the enzyme. In this work we discuss the spectrum of MPS IIIC mutations, their clinical presentation and distribution within the patient population, and speculate how the mutations may affect the structure and function of HGSNAT.


Acetyltransferases/genetics , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/genetics , Mutation/genetics , Acetyltransferases/chemistry , Amino Acid Sequence , Humans , Molecular Sequence Data , Mucopolysaccharidosis III/diagnosis , Mucopolysaccharidosis III/pathology
8.
Neurobiol Dis ; 29(2): 306-15, 2008 Feb.
Article En | MEDLINE | ID: mdl-17988881

Batten disease (neuronal ceroid lipofuscinoses, NCLs) are a group of inherited childhood diseases that result in severe brain atrophy, blindness and seizures, leading to premature death. To date, eight different genes have been identified, each associated with a different form. Linkage analysis indicated a CLN5 form in a colony of affected New Zealand Borderdale sheep. Sequencing studies established the disease-causing mutation to be a substitution at a consensus splice site (c.571+1G>A), leading to the excision of exon 3 and a truncated putative protein. A molecular diagnostic test has been developed based on the excision of exon 3. Sequence alignments support the gene product being a soluble lysosomal protein. Western blotting of isolated storage bodies indicates the specific storage of subunit c of mitochondrial ATP synthase. This flock is being expanded as a large animal model for mechanistic studies and trial therapies.


Exons/genetics , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Point Mutation , Animals , DNA Mutational Analysis/methods , Disease Models, Animal , Genetic Linkage , Islets of Langerhans/pathology , Islets of Langerhans/ultrastructure , Leukocytes/pathology , Microscopy, Electron, Transmission , Molecular Sequence Data , Neuronal Ceroid-Lipofuscinoses/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Neuronal Ceroid-Lipofuscinoses/physiopathology , Sheep Diseases , Sheep, Domestic
9.
Muscle Nerve ; 37(4): 530-6, 2008 Apr.
Article En | MEDLINE | ID: mdl-17994551

Adult polyglucosan body disease (APBD) is characterized by the accumulation of insoluble glucose polymers within the central and peripheral nervous systems. A common missense mutation in the glycogen branching enzyme (GBE1) gene has been identified in Ashkenazi patients with APBD. We report on a non-Jewish patient with APBD on whom we performed proton magnetic resonance spectroscopic imaging of the brain. GBE activity in fibroblasts was markedly reduced, and a novel heterozygous mutation was identified in the GBE1 gene. Our findings widen the spectrum of APBD genotypes, underline the importance of performing GBE analysis in all APBD patients, and suggest that brain white matter degeneration in APBD may result from tissue damage involving axons and myelin.


1,4-alpha-Glucan Branching Enzyme/genetics , Glucans/metabolism , Glycogen Storage Disease Type IV/genetics , Magnetic Resonance Spectroscopy , Nervous System Diseases/genetics , Adult , Brain/metabolism , Brain/pathology , Electromyography , Family Health , Female , Glycogen Storage Disease Type IV/metabolism , Glycogen Storage Disease Type IV/pathology , Humans , Mutation, Missense , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Protons
10.
Biochem Biophys Res Commun ; 361(2): 445-50, 2007 Sep 21.
Article En | MEDLINE | ID: mdl-17662246

Glycogen branching enzyme deficiency (glycogen storage disease type IV, GSD-IV) is a rare autosomal recessive disorder of the glycogen synthesis with high mortality. Two female newborns showed severe hypotonia at birth and both died of cardiorespiratory failure, at 4 and 12 weeks, respectively. In both patients, muscle biopsies showed deposits of PAS-positive diastase-resistant material and biochemical analysis in cultured fibroblasts showed markedly reduced glycogen branching enzyme activity. Direct sequencing of GBE1 gene revealed that patient 1 was homozygous for a novel c.691+5 g>c in intron 5 (IVS5+5 g>c). RT-PCR analysis of GBE1 transcripts from fibroblasts cDNA showed that this mutation produce aberrant splicing. Patient 2 was homozygous for a novel c.1643G>A mutation leading to a stop at codon 548 in exon 13 (p.W548X). These data underscore that in GSD-IV a severe phenotype correlates with null mutations, and indicate that RNA analysis is necessary to characterize functional consequences of intronic mutations.


Glycogen Storage Disease Type IV/genetics , Mutation/genetics , 1,4-alpha-Glucan Branching Enzyme/chemistry , 1,4-alpha-Glucan Branching Enzyme/genetics , Amino Acid Sequence , Base Sequence , Biopsy , Blotting, Western , DNA Mutational Analysis , DNA, Complementary , Exons/genetics , Fatal Outcome , Female , Fibroblasts/enzymology , Humans , Infant, Newborn , Introns/genetics , Molecular Sequence Data , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology
11.
Am J Hum Genet ; 79(5): 807-19, 2006 Nov.
Article En | MEDLINE | ID: mdl-17033958

Mucopolysaccharidosis IIIC (MPS IIIC, or Sanfilippo C syndrome) is a lysosomal storage disorder caused by the inherited deficiency of the lysosomal membrane enzyme acetyl-coenzyme A: alpha -glucosaminide N-acetyltransferase (N-acetyltransferase), which leads to impaired degradation of heparan sulfate. We report the narrowing of the candidate region to a 2.6-cM interval between D8S1051 and D8S1831 and the identification of the transmembrane protein 76 gene (TMEM76), which encodes a 73-kDa protein with predicted multiple transmembrane domains and glycosylation sites, as the gene that causes MPS IIIC when it is mutated. Four nonsense mutations, 3 frameshift mutations due to deletions or a duplication, 6 splice-site mutations, and 14 missense mutations were identified among 30 probands with MPS IIIC. Functional expression of human TMEM76 and the mouse ortholog demonstrates that it is the gene that encodes the lysosomal N-acetyltransferase and suggests that this enzyme belongs to a new structural class of proteins that transport the activated acetyl residues across the cell membrane.


Acetyltransferases/genetics , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/genetics , Mutation , Acetyltransferases/chemistry , Acetyltransferases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 8/genetics , Cloning, Molecular , DNA Mutational Analysis , DNA, Complementary/genetics , Exons , Female , Gene Expression , Humans , Male , Mice , Molecular Sequence Data , Pedigree , Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection
12.
Transfusion ; 46(10): 1705-11, 2006 Oct.
Article En | MEDLINE | ID: mdl-17002626

BACKGROUND: Hydroxyethyl starch (HES) solutions have largely replaced conventional plasma expanders such as human albumin and colloidal fluids. Only a few side effects have been reported and mainly concern pruritus or blood coagulation disorders. Excessive HES exposure can result in diffuse tissue storage and accumulation with foamy appearing macrophages which produce the enzyme chitotriosidase (CT). In case of massive tissue storage, this enzyme activity can reach levels comparable to those of Gaucher disease. STUDY DESIGN AND METHODS: In this single-center retrospective analysis of 11 consecutive patients receiving large amounts of HES for chronic plasmapheresis, plasma CT activity was investigated. Five patients receiving chronic intermittent plasmapheresis with conventional plasma expanders served as controls. Plasma CT activity was measured and plotted against creatinine clearance. Where available, marrow aspirate was analyzed with light microscopy to detect foamy macrophages. One patient developed a lysosomal storage disease and was examined extensively. RESULTS: Conventional plasma expanders did not alter plasma CT activity. In patients with impaired renal function, frequent plasma replacement with HES resulted in an increase in plasma CT activity. In the patient with the acquired lysosomal storage disease, massive tissue infiltration with activated foamy macrophages was observed. The phagocytic capacity in this patient, however, did not seem to be altered. CONCLUSION: Patients with impaired renal function receiving large amounts of HES exhibit an increase in plasma CT activity. Because excessive HES exposure can result in an acquired lysosomal storage disease, this should be avoided in chronic plasmapheresis procedures.


Hydroxyethyl Starch Derivatives/adverse effects , Kidney Diseases/therapy , Lysosomal Storage Diseases/chemically induced , Plasma Substitutes/adverse effects , Plasmapheresis/adverse effects , Adult , Aged , Bone Marrow/enzymology , Bone Marrow/pathology , Female , Foam Cells/enzymology , Foam Cells/pathology , Gaucher Disease/enzymology , Gaucher Disease/pathology , Hexosaminidases/blood , Humans , Hydroxyethyl Starch Derivatives/administration & dosage , Kidney Diseases/blood , Kidney Diseases/enzymology , Kidney Diseases/pathology , Lysosomal Storage Diseases/blood , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/pathology , Male , Middle Aged , Plasma Substitutes/administration & dosage , Retrospective Studies
13.
Mol Genet Metab ; 88(1): 22-8, 2006 May.
Article En | MEDLINE | ID: mdl-16359900

We have established a new method for the enzymatic diagnosis of glycogen storage disease type II (Pompe disease or acid maltase deficiency) using mixed leukocytes. The method employs glycogen and 4-methylumbelliferyl-alpha-D-glucopyranoside (4MU-alphaGlc) as substrates for measuring the lysosomal acid alpha-glucosidase (acid alphaGlu) activity, and incorporates acarbose to eliminate the interference of unrelated alpha-glucosidases (predominantly maltase-glucoamylase). It is shown that 3.0 micromol/L acarbose completely inhibits the maltase-glucoamylase activity at pH 4.0, but the lysosomal acid alphaGlu activity by less than 5%. With this method, we determined the acid alphaGlu activity in mixed leukocytes from 25 patients with glycogen storage disease type II (2 infantile and 23 late-onset cases), one GAA2/GAA2 homozygote and 30 healthy subjects. In the assay with glycogen as substrate, the addition of acarbose created a clear separation between the patient and the control ranges. In the assay with 4MU-alphaGlc as substrate, the two ranges were fully separated but remained very close despite the use of acarbose. The separation of the patient and normal ranges was improved considerably by taking the ratio of acarbose-inhibited over uninhibited activity. A GAA2/GAA2 homozygote was correctly diagnosed with 4MU-alphaGlc but misdiagnosed as patient when glycogen was used as substrate. We conclude that the inclusion of 3.0 micromol/L acarbose in the assays with glycogen and 4MU-alphaGlc substrates at pH 4.0 allows for the specific measurement of lysosomal acid alphaGlu activity in mixed leukocytes, thus enabling a reliable diagnosis of glycogen storage disease type II in this specimen.


Glucan 1,4-alpha-Glucosidase/deficiency , Glycogen Storage Disease Type II/diagnosis , Leukocytes/chemistry , Acarbose , Glucan 1,4-alpha-Glucosidase/antagonists & inhibitors , Humans , Hydrogen-Ion Concentration , Infant , alpha-Glucosidases
14.
Hum Mol Genet ; 13(23): 2893-906, 2004 Dec 01.
Article En | MEDLINE | ID: mdl-15459177

Neuronal ceroid lipofuscinoses (NCL) comprise the most common group of childhood encephalopathies caused by mutations in eight genetic loci, CLN1-CLN8. Here, we have developed a novel mouse model for the human vLINCL (CLN5) by targeted deletion of exon 3 of the mouse Cln5 gene. The Cln5-/- mice showed loss of vision and accumulation of autofluorescent storage material in the central nervous system (CNS) and peripheral tissues without prominent brain atrophy. The ultrastructure of the storage material accurately replicated the abnormalities in human patients revealing mixture of lamellar profiles including fingerprint profiles as well as curvilinear and rectilinear bodies in electronmicroscopic analysis. Prominent loss of a subset of GABAergic interneurons in several brain areas was seen in the Cln5-/- mice. Transcript profiling of the brains of the Cln5-/- mice revealed altered expression in several genes involved in neurodegeneration, as well as in defense and immune response, typical of age-associated changes in the CNS. Downregulation of structural components of myelin was detected and this agrees well with the hypomyelination seen in the human vLINCL patients. In general, the progressive pathology of the Cln5-/- brain mimics the symptoms of the corresponding neurodegenerative disorder in man. Since the Cln5-/- mice do not exhibit significant brain atrophy, these mice could serve as models for studies on molecular processes associated with advanced aging.


Aging , Brain/pathology , Disease Models, Animal , Membrane Proteins/physiology , Neuronal Ceroid-Lipofuscinoses/genetics , Animals , Base Sequence , Brain/enzymology , Brain/physiopathology , DNA Primers , Gene Expression Profiling , Humans , Immunohistochemistry , Lysosomal Membrane Proteins , Lysosomes/enzymology , Membrane Proteins/genetics , Mice , Mice, Knockout , Neuronal Ceroid-Lipofuscinoses/pathology , Reverse Transcriptase Polymerase Chain Reaction , gamma-Aminobutyric Acid/physiology
15.
Pediatrics ; 113(5): e448-57, 2004 May.
Article En | MEDLINE | ID: mdl-15121988

OBJECTIVE: Recent reports warn that the worldwide cell culture capacity is insufficient to fulfill the increasing demand for human protein drugs. Production in milk of transgenic animals is an attractive alternative. Kilogram quantities of product per year can be obtained at relatively low costs, even in small animals such as rabbits. We tested the long-term safety and efficacy of recombinant human -glucosidase (rhAGLU) from rabbit milk for the treatment of the lysosomal storage disorder Pompe disease. The disease occurs with an estimated frequency of 1 in 40,000 and is designated as orphan disease. The classic infantile form leads to death at a median age of 6 to 8 months and is diagnosed by absence of alpha-glucosidase activity and presence of fully deleterious mutations in the alpha-glucosidase gene. Cardiac hypertrophy is characteristically present. Loss of muscle strength prevents infants from achieving developmental milestones such as sitting, standing, and walking. Milder forms of the disease are associated with less severe mutations and partial deficiency of alpha-glucosidase. METHODS: In the beginning of 1999, 4 critically ill patients with infantile Pompe disease (2.5-8 months of age) were enrolled in a single-center open-label study and treated intravenously with rhAGLU in a dose of 15 to 40 mg/kg/week. RESULTS: Genotypes of patients were consistent with the most severe form of Pompe disease. Additional molecular analysis failed to detect processed forms of alpha-glucosidase (95, 76, and 70 kDa) in 3 of the 4 patients and revealed only a trace amount of the 95-kDa biosynthetic intermediate form in the fourth (patient 1). With the more sensitive detection method, 35S-methionine incorporation, we could detect low-level synthesis of -glucosidase in 3 of the 4 patients (patients 1, 2, and 4) with some posttranslation modification from 110 kDa to 95 kDa in 1 of them (patient 1). One patient (patient 3) remained totally deficient with both detection methods (negative for cross-reactive immunologic material [CRIM negative]). The alpha-glucosidase activity in skeletal muscle and fibroblasts of all 4 patients was below the lower limit of detection (<2% of normal). The rhAGLU was tolerated well by the patients during >3 years of treatment. Anti-rhAGLU immunoglobulin G titers initially increased during the first 20 to 48 weeks of therapy but declined thereafter. There was no consistent difference in antibody formation comparing CRIM-negative with CRIM-positive patients. Muscle alpha-glucosidase activity increased from <2% to 10% to 20% of normal in all patients during the first 12 weeks of treatment with 15 to 20 mg/kg/week. For optimizing the effect, the dose was increased to 40 mg/kg/week. This resulted, 12 weeks later, in normal alpha-glucosidase activity levels, which were maintained until the last measurement in week 72. Importantly, all 4 patients, including the patient without any endogenous alpha-glucosidase (CRIM negative), revealed mature 76- and 70-kDa forms of -glucosidase on Western blot. Conversion of the 110-kDa precursor from milk to mature 76/70-kDa alpha-glucosidase provides evidence that the enzyme is targeted to lysosomes, where this proteolytic processing occurs. At baseline, patients had severe glycogen storage in the quadriceps muscle as revealed by strong periodic acid-Schiff--positive staining and lacework patterns in hematoxylin and eosin--stained tissue sections. The muscle pathology correlated at each time point with severity of signs. Periodic acid-Schiff intensity diminished and number of vacuoles increased during the first 12 weeks of treatment. Twelve weeks after dose elevation, we observed signs of muscle regeneration in 3 of the 4 patients. Obvious improvement of muscular architecture was seen only in the patient who learned to walk. Clinical effects were significant. All patients survived beyond the age of 4 years, whereas untreated patients succumb at a median age of 6 to 8 months. The characteristic cardiac hypertrophy present at start of treatment diminished significantly. The left ventricular mass index decreased from 171 to 599 g/m2 (upper limit of normal 86.6 g/m2 for infants from 0 to 1 year) to 70 to 160 g/m2 during 84 weeks of treatment. In addition, we found a significant change of slope for the diastolic thickness of the left ventricular posterior wall against time at t = 0 for each separate patient. Remarkably, the younger patients (patients 1 and 3) showed no significant respiratory problems during the first 2 years of life. One of the younger patients recovered from a life-threatening bronchiolitis at the age of 1 year without sequelae, despite borderline oxygen saturations at inclusion. At the age of 2, however, she became ventilator dependent after surgical removal of an infected Port-A-Cath. She died at the age of 4 years and 3 months suddenly after a short period of intractable fever of >42 degrees C, unstable blood pressure, and coma. The respiratory course of patient 1 remained uneventful. The 2 older patients, who both were hypercapnic (partial pressure of carbon dioxide: 10.6 and 9.8 kPa; normal range: 4.5-6.8 kPa) at start of treatment, became ventilator dependent before the first infusion (patient 2) and after 10 weeks of therapy (patient 4). Patient 4 was gradually weaned from the ventilator after 1 year of high-dose treatment and was eventually completely ventilator-free for 5 days, but this situation could not be maintained. Currently, both patients are completely ventilator dependent. The most remarkable progress in motor function was seen in the younger patients (patients 1 and 3). They achieved motor milestones that are unmet in infantile Pompe disease. Patient 1 learned to crawl (12 months), walk (16 months), squat (18 months), and climb stairs (22 months), and patient 3 learned to sit unsupported. The Alberta Infant Motor Scale score for patients 2, 3, and 4 remained far below p5. Patient 1 followed the p5 of normal. CONCLUSION: Our study shows that a safe and effective medicine can be produced in the milk of mammals and encourages additional development of enzyme replacement therapy for the several forms of Pompe disease. Restoration of skeletal muscle function and prevention of pulmonary insufficiency require dosing in the range of 20 to 40 mg/kg/week. The effect depends on residual muscle function at the start of treatment. Early start of treatment is required.


Glycogen Storage Disease Type II/drug therapy , Milk/enzymology , Transgenes , alpha-Galactosidase/therapeutic use , Animals , Animals, Genetically Modified , Cardiomegaly/etiology , Child, Preschool , Female , Glycogen Storage Disease Type II/pathology , Glycogen Storage Disease Type II/physiopathology , Humans , Infant , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Psychomotor Performance , Rabbits/genetics , Recombinant Proteins/therapeutic use , Respiratory Insufficiency/etiology , Survival Analysis , Treatment Outcome , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism
16.
Ann Neurol ; 55(4): 495-502, 2004 Apr.
Article En | MEDLINE | ID: mdl-15048888

Pompe's disease is an autosomal recessive myopathy. The characteristic lysosomal storage of glycogen is caused by acid alpha-glucosidase deficiency. Patients with late-onset Pompe's disease present with progressive muscle weakness also affecting pulmonary function. In search of a treatment, we investigated the feasibility of enzyme replacement therapy with recombinant human alpha-glucosidase from rabbit milk. Three patients (aged 11, 16, and 32 years) were enrolled in the study. They were all wheelchair-bound and two of them were ventilator dependent with a history of deteriorating pulmonary function. After 3 years of treatment with weekly infusions of alpha-glucosidase, the patients had stabilized pulmonary function and reported less fatigue. The youngest and least affected patient showed an impressive improvement of skeletal muscle strength and function. After 72 weeks of treatment, he could walk without support and finally abandoned his wheelchair. Our findings demonstrate that recombinant human alpha-glucosidase from rabbit milk has a therapeutic effect in late-onset Pompe's disease. There is good reason to continue the development of enzyme replacement therapy for Pompe's disease and to explore further the production of human therapeutic proteins in the milk of mammals.


Glycogen Storage Disease Type II/drug therapy , alpha-Glucosidases/therapeutic use , Adolescent , Adult , Animals , Child , Female , Follow-Up Studies , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/physiopathology , Humans , Linear Models , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Pilot Projects , Rabbits , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Recovery of Function/drug effects , Recovery of Function/physiology , alpha-Glucosidases/pharmacology
17.
Brain Pathol ; 13(4): 574-81, 2003 Oct.
Article En | MEDLINE | ID: mdl-14655761

We describe the neuropathological and biochemical autopsy findings in 3 patients with autosomal dominant adult neuronal ceroid lipofuscinosis (ANCL, Parry type; MIM 162350), from a family with 6 affected individuals in 3 generations. Throughout the brain of these patients, there was abundant intraneuronal lysosomal storage of autofluorescent lipopigment granules. Striking loss of neurons in the substantia nigra was found. In contrast, little neuronal cell loss occurred in other cerebral areas, despite massive neuronal inclusions. Visceral storage was present in gut, liver, cardiomyocytes, skeletal muscle, and in the skin eccrine glands. The storage material showed highly variable immunoreactivity with antiserum against subunit c of mitochondrial ATP synthase, but uniform strong immunoreactivity for saposin D (sphingolipid activating protein D). Protein electrophoresis of isolated storage material revealed a major protein band of about 14 kDa, recognized in Western blotting by saposin D antiserum (but not subunit c of mitochondrial ATPase (SCMAS) antiserum). Electron microscopy showed ample intraneuronal granular osmiophilic deposits (GRODs), as occurs in CLN1 and congenital ovine NCL. These forms of NCL are caused by the deficiencies of palmitoyl protein thioesterase 1 and cathepsin D, respectively. However, activities of these enzymes were within normal range in our patients. Thus we propose that a gene distinct from the cathepsin D and CLN1-CLN8 genes is responsible for this autosomal dominant form of ANCL.


Lipids , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/pathology , Palmitoyl-CoA Hydrolase/metabolism , Adult , Blotting, Western , Cathepsin D/metabolism , Electrophoresis, Polyacrylamide Gel , Family Health , Female , Glycoproteins/metabolism , Humans , Immunohistochemistry , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Male , Microscopy, Electron , Middle Aged , Mitochondrial Proton-Translocating ATPases/metabolism , Neuronal Ceroid-Lipofuscinoses/enzymology , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Neurons/ultrastructure , Palmitoyl-CoA Hydrolase/deficiency , Pedigree , Peptide Hydrolases/metabolism , Pigments, Biological/metabolism , Saposins
18.
Pediatrics ; 112(2): 332-40, 2003 Aug.
Article En | MEDLINE | ID: mdl-12897283

OBJECTIVE: Infantile Pompe's disease is a lethal cardiac and muscular disorder. Current developments toward enzyme replacement therapy are promising. The aim of our study is to delineate the natural course of the disease to verify endpoints of clinical studies. METHODS: A total of 20 infantile patients diagnosed by the collaborative Dutch centers and 133 cases reported in literature were included in the study. Information on clinical history, physical examination, and diagnostic parameters was collected. RESULTS: The course of Pompe's disease is essentially the same in the Dutch and the general patient population. Symptoms start at a median age of 1.6 months in both groups. The median age of death is 7.7 and 6 months, respectively. Five percent of the Dutch patients and 8% of all reported patients survive beyond 1 year of age. Only 2 patients from literature became older than 18 months. A progressive cardiac hypertrophy is characteristic for infantile Pompe's disease. The diastolic thickness of the left ventricular posterior wall and cardiac weight at autopsy increase significantly with age. Motor development is severely delayed and major developmental milestones are generally not achieved. For the Dutch patient group, growth deviates significantly from normal despite start of nasogastric tube feeding. Levels of aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, creatine kinase, or creatine kinase-myocardial band isoenzyme are typically elevated, although aspartate aminotransferase, alanine aminotransferase, and lactate dehydrogenase increase significantly with age. The patients have fully deleterious mutations. Acid alpha-glucosidase activity is severely deficient. CONCLUSIONS: Survival, decrease of the diastolic thickness of the left ventricular posterior wall, and achievement of major motor milestones are valid endpoints for therapeutic studies of infantile Pompe's disease. Mutation analysis and measurement of the alpha-glucosidase activity should be part of the enrollment program.


Cardiomegaly/etiology , Glycogen Storage Disease Type II/physiopathology , Blood Chemical Analysis , Brain/pathology , Cardiomegaly/diagnosis , Child Development , Disease Progression , Glycogen Storage Disease Type II/complications , Glycogen Storage Disease Type II/mortality , Humans , Infant , Infant, Newborn/growth & development , Infant, Premature , Mutation , Netherlands/epidemiology , Survival Analysis , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
19.
J Comp Neurol ; 455(3): 368-77, 2003 Jan 13.
Article En | MEDLINE | ID: mdl-12483688

Palmitoyl protein thioesterase 1 (PPT1) is a depalmitoylating enzyme whose deficiency leads to infantile neuronal ceroid lipofuscinosis. The disease is characterized by early loss of vision and massive neuronal death. Although PPT1 is expressed in many tissues, a deficiency of PPT1 damages neurons only in the cerebral and cerebellar cortexes and retina; other cell types remain relatively unaffected. We previously demonstrated that PPT1 is present in the synaptosomes and synaptic vesicles of neurons. To understand the crucial role of PPT1 for neuronal cells, we further investigated the expression and targeting of PPT1 in retinal, hippocampal, and cortical neurons during their maturation in culture. We found that PPT1 activity increases by neuronal maturation and is highest in retinal neuron cultures. In retinal neurons the expression of PPT1 precedes that of the synaptic vesicle protein 2 and synaptophysin, indicating a significant role for PPT1 in the early development of neuronal cells. We also found by quantitative confocal immunofluorescence microscopy that PPT1 is targeted preferably to axons in mature neurons, as indicated by its colocalization with the axonal marker microtubule-associated protein 1. In axons PPT1 is targeted specifically to axonal varicosities and presynaptic terminals, as indicated by its significant colocalization with growth-associated protein 43 and synaptophysin. Axonal localization of PPT1 was confirmed by double labeling with synaptophysin and postembedding immunoelectron microscopy. The polarized axonal targeting of PPT1 may well indicate a role for PPT1 in the exocytotic pathway of neurons.


Axons/enzymology , Cerebral Cortex/growth & development , Hippocampus/growth & development , Neurons/enzymology , Retina/growth & development , Thiolester Hydrolases/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Blotting, Western , Cell Culture Techniques , Cerebral Cortex/chemistry , Cerebral Cortex/ultrastructure , Hippocampus/chemistry , Hippocampus/ultrastructure , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron , Microscopy, Immunoelectron , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/ultrastructure , Presynaptic Terminals/metabolism , Retina/chemistry , Retina/ultrastructure , Synaptophysin/metabolism
20.
J Lipid Res ; 43(7): 1096-104, 2002 Jul.
Article En | MEDLINE | ID: mdl-12091494

Skin fibroblast cultures from patients with inherited lysosomal enzymopathies, alpha-N-acetylgalactosaminidase (alpha-NAGA) and alpha-galactosidase A deficiencies (Schindler and Fabry disease, respectively), and from normal controls were used to study in situ degradation of blood group A and B glycosphingolipids. Glycosphingolipids A-6-2 (GalNAc (alpha 1-->3)[Fuc alpha 1-->2]Gal(beta1-->4)GlcNAc(beta 1-->3)Gal(beta 1--> 4)Glc (beta 1-->1')Cer, IV(2)-alpha-fucosyl-IV(3)-alpha-N-acetylgalactosaminylneolactotetraosylceramide), B-6-2 (Gal(alpha 1-->3)[Fuc alpha 1--> 2] Gal (beta 1-->4)GlcNAc(beta 1-->3)Gal(beta 1-->4)Glc(beta 1-->1')Cer, IV(2)- alpha-fucosyl-IV(3)-alpha-galactosylneolactotetraosylceramide), and globoside (GalNAc(beta 1-->3)Gal(alpha 1-->4)Gal(beta 1-->4)Glc(beta 1-->1') Cer, globotetraosylceramide) were tritium labeled in their ceramide moiety and used as natural substrates. The degradation rate of glycolipid A-6-2 was very low in fibroblasts of all the alpha-NAGA-deficient patients (less than 7% of controls), despite very heterogeneous clinical pictures, ruling out different residual enzyme activities as an explanation for the clinical heterogeneity. Strongly elevated urinary excretion of blood group A glycolipids was detected in one patient with blood group A, secretor status (five times higher than upper limit of controls), in support of the notion that blood group A-active glycolipids may contribute as storage compounds in blood group A patients. When glycolipid B-6-2 was fed to alpha-galactosidase A-deficient cells, the degradation rate was surprisingly high (50% of controls), while that of globotriaosylceramide was reduced to less than 15% of control average, presumably reflecting differences in the lysosomal enzymology of polar glycolipids versus less-polar ones. Relatively high-degree degradation of substrates with alpha-D-Galactosyl moieties hints at a possible contribution of other enzymes.


ABO Blood-Group System/chemistry , ABO Blood-Group System/metabolism , Fabry Disease/metabolism , Glycosphingolipids/metabolism , Adolescent , Adult , Cell Line , Child , Child, Preschool , Fabry Disease/blood , Fabry Disease/enzymology , Fabry Disease/pathology , Fibroblasts , Glycosphingolipids/blood , Glycosphingolipids/urine , Hexosaminidases/deficiency , Humans , Skin , alpha-N-Acetylgalactosaminidase
...