Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Publication year range
1.
J Neuromuscul Dis ; 7(1): 15-31, 2020.
Article in English | MEDLINE | ID: mdl-31796685

ABSTRACT

Pompe disease (glycogen storage disease type II) is caused by mutations in acid α-glucosidase (GAA) resulting in lysosomal pathology and impairment of the muscular and cardio-pulmonary systems. Enzyme replacement therapy (ERT), the only approved therapy for Pompe disease, improves muscle function by reducing glycogen accumulation but this approach entails several limitations including a short drug half-life and an antibody response that results in reduced efficacy. To address these limitations, new treatments such as gene therapy are under development to increase the intrinsic ability of the affected cells to produce GAA. Key components to gene therapy strategies include the choice of vector, promoter, and the route of administration. The efficacy of gene therapy depends on the ability of the vector to drive gene expression in the target tissue and also on the recipient's immune tolerance to the transgene protein. In this review, we discuss the preclinical and clinical studies that are paving the way for the development of a gene therapy strategy for patients with early and late onset Pompe disease as well as some of the challenges for advancing gene therapy.


Subject(s)
Dependovirus , Genetic Therapy , Glycogen Storage Disease Type II/therapy , Animals , Humans
2.
Int J Clin Pharmacol Ther ; 47 Suppl 1: S42-7, 2009.
Article in English | MEDLINE | ID: mdl-20040311

ABSTRACT

Pompe disease is caused by an inherited deficiency of acid a-glucosidase (GAA), a lysosomal enzyme that catalyzes the breakdown of glycogen to glucose. In the absence of GAA, enlarged, glycogen-laden lysosomes accumulate in multiple tissues, although the major clinical manifestations are seen in cardiac and skeletal muscle. For many years, it was believed that the rupture of glycogen-filled lysosomes was the major cause of the profound muscle damage observed in patients with Pompe disease. Here, we present evidence that a failure of productive autophagy in muscle tissue contributes strongly to disease pathology in both patients with Pompe disease and GAA-knockout mice. In the GAA-knockout mouse model, progressive accumulation of autophagic vesicles is restricted to Type II-rich muscle fibers. Not only does this build-up of autophagosomes disrupt the contractile apparatus in the muscle fibers, it also interferes with enzyme replacement therapy by acting as a sink for the recombinant enzyme and preventing its efficient delivery to the lysosomes. Our data indicate that a re-examination of the presumed pathological mechanism in Pompe disease is necessary, and suggest that successful treatment of patients with Pompe disease will require consideration of the dramatic failure of autophagy that occurs in this disease.


Subject(s)
Autophagy/physiology , Glycogen Storage Disease Type II/pathology , Muscle, Skeletal/pathology , alpha-Glucosidases/therapeutic use , Animals , Enzyme Replacement Therapy/methods , Glycogen Storage Disease Type II/physiopathology , Glycogen Storage Disease Type II/therapy , Humans , Mice , Mice, Knockout , Muscle, Skeletal/ultrastructure , alpha-Glucosidases/deficiency , alpha-Glucosidases/genetics
3.
J Struct Biol ; 162(3): 500-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18468456

ABSTRACT

The large size of the multinucleated muscle fibers of skeletal muscle makes their examination for structural and pathological defects a challenge. Sections and single fibers are accessible to antibodies and other markers but imaging of such samples does not provide a three-dimensional view of the muscle. Regrettably, bundles of fibers cannot be stained or imaged easily. Two-photon microscopy techniques overcome these obstacles. Second harmonic generation (SHG) by myosin filaments and two-photon excited fluorescence (2PEF) of mitochondrial and lysosomal components provides detailed structural information on unstained tissue. Furthermore, the infrared exciting light can penetrate several layers of muscle fibers and the minimal processing is particularly valuable for fragile biopsies. Here we demonstrate the usefulness of SHG, combined with 2PEF, to reveal enlarged lysosomes and accumulations of non-contractile material in muscles from the mouse model for the lysosomal storage disorder Pompe disease (PD), and in biopsies from adult and infant PD patients. SHG and 2PEF also detect sarcomeric defects that may presage the loss of myofibrils in atrophying muscle and signify loss of elasticity. The combination of SHG and 2PEF should be useful in the analysis and diagnosis of a wide range of skeletal muscle pathologies.


Subject(s)
Muscle, Skeletal/metabolism , Sarcomeres/pathology , Adult , Animals , Autophagy , Glycogen Storage Disease Type II/metabolism , Humans , Infant , Infant, Newborn , Mice , Mice, Knockout , Microscopy, Fluorescence/methods , Mitochondria/metabolism , Muscle Contraction , alpha-Glucosidases/metabolism
4.
Mol Genet Metab ; 94(4): 448-455, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18538603

ABSTRACT

Pompe disease results in the accumulation of lysosomal glycogen in multiple tissues due to a deficiency of acid alpha-glucosidase (GAA). Enzyme replacement therapy for Pompe disease was recently approved in Europe, the U.S., Canada, and Japan using a recombinant human GAA (Myozyme, alglucosidase alfa) produced in CHO cells (CHO-GAA). During the development of alglucosidase alfa, we examined the in vitro and in vivo properties of CHO cell-derived rhGAA, an rhGAA purified from the milk of transgenic rabbits, as well as an experimental version of rhGAA containing additional mannose-6-phosphate intended to facilitate muscle targeting. Biochemical analyses identified differences in rhGAA N-termini, glycosylation types and binding properties to several carbohydrate receptors. In a mouse model of Pompe disease, glycogen was more efficiently removed from the heart than from skeletal muscle for all enzymes, and overall, the CHO cell-derived rhGAA reduced glycogen to a greater extent than that observed with the other enzymes. The results of these preclinical studies, combined with biochemical characterization data for the three molecules described within, led to the selection of the CHO-GAA for clinical development and registration as the first approved therapy for Pompe disease.


Subject(s)
Glycogen Storage Disease Type II/therapy , alpha-Glucosidases/chemistry , alpha-Glucosidases/pharmacology , Animals , Antibodies/blood , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Glycogen/metabolism , Glycogen Storage Disease Type II/immunology , Glycogen Storage Disease Type II/metabolism , Humans , Lectins, C-Type/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Protein Binding , Rabbits , Receptor, IGF Type 2/metabolism , Receptors, Cell Surface/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
5.
Acta Myol ; 26(1): 45-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17915569

ABSTRACT

In Pompe disease, a deficiency of lysosomal acid alpha-glucosidase, glycogen accumulates in multiple tissues, but clinical manifestations are mainly due to skeletal and cardiac muscle involvement. A major advance has been the development of enzyme replacement therapy (ERT), which recently became available for Pompe patients. Based on clinical and pre-clinical studies, the effective clearance of skeletal muscle glycogen appears to be more difficult than anticipated. Skeletal muscle destruction and resistance to therapy remain unsolved problems. We have found that the cellular pathology in Pompe disease spreads to affect both the endocytic and autophagic pathways, leading to excessive autophagic buildup in therapy resistant muscle fibers of knockout mice. Furthermore, the autophagic buildup had a profound effect on the trafficking and processing of the therapeutic enzyme along the endocytic pathway. These findings may explain why ERT often falls short of reversing the disease process, and point to new avenues for the development of pharmacological intervention.


Subject(s)
Autophagy , Glycogen Storage Disease Type II/physiopathology , Animals , Disease Models, Animal , Glycogen Storage Disease Type II/pathology , Humans , Mice , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/ultrastructure
6.
Diabetes ; 40(1): 118-22, 1991 Jan.
Article in English | MEDLINE | ID: mdl-2015966

ABSTRACT

The nucleotide sequence of the insulin gene was determined in American Pima Indians and Micronesian Nauruans, two populations in whom the prevalence of non-insulin-dependent (type II) diabetes mellitus is the highest in the world. The insulin gene was amplified by the polymerase chain reaction to generate single-stranded DNA suitable for direct sequencing. The nucleotide sequences of the coding and adjacent regions of the insulin gene in six Pima Indians and two Nauruans with type II diabetes were identical to previously published insulin gene sequences of nondiabetic subjects.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Ethnicity , Indians, North American , Insulin/genetics , Adult , Arizona , Base Sequence , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Exons , Female , Humans , Insulin/blood , Insulin/isolation & purification , Male , Micronesia , Middle Aged , Molecular Sequence Data , Oligonucleotide Probes , Polymerase Chain Reaction/methods , Polymorphism, Genetic , Prevalence , Proinsulin/blood , RNA, Messenger/genetics , RNA, Messenger/isolation & purification
7.
Diabetes ; 41(4): 408-15, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1607067

ABSTRACT

Denaturing gradient gel electrophoresis (DGGE) has been used to screen for mutations in the insulin receptor gene. Each of the 22 exons was amplified by the polymerase chain reaction (PCR). For each exon, one of the two PCR primers contained a guanine-cytosine (GC) clamp at its 5' end. The DNA was analyzed by electrophoresis through a polyacrylamide gel containing a gradient of denaturants. Two geometries for the gels were compared; the gradient of denaturants was oriented either parallel or perpendicular to the electric field. The sensitivity of the technique was evaluated by determining whether DGGE succeeded in detecting known mutations and polymorphisms in the insulin receptor gene. With parallel gels, 12 of 16 sequence variants were detected. The use of perpendicular gels increased the sensitivity of detection so that all 16 sequence variants were successfully detected when DNA was analyzed by a combination of perpendicular and parallel gels. Furthermore, DGGE was used to investigate a patient with leprechaunism whose insulin receptor genes had not previously been studied. Two mutant alleles were identified in this patient. The allele inherited from the father had a mutation substituting alanine for Val-28; in the allele inherited from the mother, arginine was substituted for Gly-366.


Subject(s)
Electrophoresis, Polyacrylamide Gel/methods , Mutation/genetics , Receptor, Insulin/genetics , Alanine/analysis , Alleles , Arginine/analysis , Base Sequence , Child , DNA/analysis , DNA/genetics , Exons , Female , Gene Amplification , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genetic Testing , Genetic Variation , Humans , Molecular Sequence Data , Nucleic Acid Denaturation , Polymerase Chain Reaction , Polymorphism, Genetic
8.
Hum Gene Ther ; 8(12): 1469-80, 1997 Aug 10.
Article in English | MEDLINE | ID: mdl-9287147

ABSTRACT

Polymyositis is regarded as an autoimmune inflammatory muscle disease. A major subgroup of patients have autoantibodies to cellular histidyl-transfer RNA synthetase (HRS). We have analyzed the role of the autoantigen HRS in the induction of murine myositis in a comparative study of inoculation of BALB/c mice with recombinant HRS protein versus naked DNA coding for HRS. Adult BALB/c mice produced antibodies to human HRS following inoculation with HRS protein and adjuvant, but myositis was not observed. Alternatively, expression plasmid DNA constructs encoding full-length and truncated human HRS were inoculated intramuscularly in gene transfer studies. DNA-inoculated mice produced relatively low anti-HRS antibody titers. However, in contrast to recombinant HRS protein-inoculated mice, HRS gene transfer induced pathology with evidence of cellular infiltration of perivascular and endomysial regions of the inoculated muscle. Multiple inoculations of a plasmid construct encoding a hybrid molecule consisting of HRS and the transferrin receptor cytoplasmic tail induced the highest levels of antibodies and persisting cellular infiltration. Unlike HRS, expression of influenza virus hemagglutinin (HA) following inoculation of an HA plasmid did not induce myositis. Transfer of naked DNA constructs expressing HRS is likely to provide valuable information on the autoimmune response to this protein and its role in the development of myositis.


Subject(s)
Histidine-tRNA Ligase/genetics , Histidine-tRNA Ligase/immunology , Immunization , Myositis/immunology , Animals , Disease Models, Animal , Drug Administration Schedule , Female , Gene Transfer Techniques , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Histidine-tRNA Ligase/pharmacology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Molecular Sequence Data , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Myositis/chemically induced , Polymyositis/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology
9.
Hum Gene Ther ; 12(5): 527-38, 2001 Mar 20.
Article in English | MEDLINE | ID: mdl-11268285

ABSTRACT

Pompe disease is a lethal cardioskeletal myopathy in infants and results from genetic deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). Genetic replacement of the cDNA for human GAA (hGAA) is one potential therapeutic approach. Three months after a single intramuscular injection of 10(8) plaque-forming units (PFU) of E1-deleted adenovirus encoding human GAA (Ad-hGAA), the activity in whole muscle lysates of immunodeficient mice is increased to 20 times the native level. Direct transduction of a target muscle, however, may not correct all deficient cells. Therefore, the amount of enzyme that can be transferred to deficient cells from virally transduced cells was studied. Fibroblasts from an affected patient were transduced with AdhGAA, washed, and plated on transwell culture dishes to serve as donors of recombinant enzyme. Deficient fibroblasts were plated as acceptor cells, and were separated from the donor monolayer by a 22-microm pore size filter. Enzymatic and Western analyses demonstrate secretion of the 110-kDa precursor form of hGAA from the donor cells into the culture medium. This recombinant, 110-kDa species reaches the acceptor cells, where it can be taken up by mannose 6-phosphate receptor-mediated endocytosis. It then trafficks to lysosomes, where Western analysis shows proteolytic processing to the 76- and 70-kDa lysosomal forms of the enzyme. Patient fibroblasts receiving recombinant hGAA by this transfer mechanism reach levels of enzyme activity that are comparable to normal human fibroblasts. Skeletal muscle cell cultures from an affected patient were also transduced with Ad-hGAA. Recombinant hGAA is identified in a lysosomal location in these muscle cells by immunocytochemistry, and enzyme activity is transferred to deficient skeletal muscle cells grown in coculture. Transfer of the precursor protein between muscle cells again occurs via mannose 6-phosphate receptors, as evidenced by competitive inhibition with 5 mM mannose 6-phosphate. In vivo studies in GAA-knockout mice demonstrate that hepatic transduction with adenovirus encoding either murine or human GAA can provide a depot of recombinant enzyme that is available to heart and skeletal muscle through this mechanism. Taken together, these data show that the mannose 6-phosphate receptor pathway provides a useful strategy for cell-to-cell distribution of virally derived recombinant GAA.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , alpha-Glucosidases/genetics , Adenoviridae/genetics , Animals , Blotting, Western , Cells, Cultured , Coculture Techniques , DNA, Complementary/metabolism , Fibroblasts/metabolism , Humans , Immunohistochemistry , Lysosomes/metabolism , Mannosephosphates/metabolism , Mice , Mice, Knockout , Mice, Nude , Muscle, Skeletal/cytology , Myocardium/metabolism , Placenta/metabolism , Receptor, IGF Type 2/metabolism , Recombinant Proteins/metabolism , Time Factors , Transduction, Genetic
10.
J Clin Endocrinol Metab ; 71(1): 164-9, 1990 Jul.
Article in English | MEDLINE | ID: mdl-2196279

ABSTRACT

Mutations in the insulin gene can impair the bioactivity of the insulin molecule. Previously, two classes of mutations have been identified: 1) those that impair posttranslational processing of proinsulin to insulin, and 2) those that alter the structure of the insulin molecule, thereby reducing the affinity of the molecule for the insulin receptor. We have investigated two apparently unrelated patients, both of which have mutations that inhibit the conversion of proinsulin to insulin. By directly sequencing genomic DNA amplified by polymerase chain reaction, we have demonstrated that both patients are heterozygous for the same point mutation converting codon 65 from an arginine (CGT) to a histidine (CAT) codon. Because Arg65 is one of the two dibasic amino acids at the site of proteolytic cleavage between the insulin A-chain and C-peptide, this mutation explains the impairment in the cleavage of proinsulin to insulin. Interestingly, the same His65 mutation has been identified in the insulin gene of a Japanese kindred with familial hyperproinsulinemia. Thus, this mutation has occurred in three apparently unrelated kindreds from two different racial groups. This observation is consistent with the hypothesis that the dinucleotide sequence CpG, the first two nucleotides in the arginine (CGT) codon, is a "hot spot" for mutations.


Subject(s)
Arginine , DNA/analysis , Histidine , Insulin/genetics , Proinsulin/blood , Proinsulin/genetics , Alleles , Amino Acid Sequence , Base Sequence , Blood Glucose/analysis , Codon , Exons , Gene Amplification , Genes , Humans , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Insulin/biosynthesis , Male , Middle Aged , Molecular Sequence Data , Mutation , Nucleic Acid Hybridization , Peptide Hydrolases , Polymerase Chain Reaction , Proinsulin/metabolism
11.
Gene ; 138(1-2): 219-22, 1994 Jan 28.
Article in English | MEDLINE | ID: mdl-8125304

ABSTRACT

Two isoforms of lck/yes-related novel (LYN) protein tyrosine kinase (PTK) appear to play a role in B-cell-IgM and FcERI receptor signaling. The cDNAs lynA and lynB encoding these two forms were isolated and sequenced; they were derived from rat mucosal mast cell and human myeloid cell lines. The nucleotide (nt) and deduced amino acid (aa) sequences share 94 and 97% identity between rat and mouse lyn, respectively, and 88 and 96% identity between rat and human lyn. In all three species, a region of 20 aa is uniformly inserted at an identical site and its sequence is highly conserved. This suggests an important regulatory role for this region mediated by this PTK.


Subject(s)
Protein-Tyrosine Kinases/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA Primers , DNA, Complementary/metabolism , Humans , Leukemia, Basophilic, Acute , Leukemia, Promyelocytic, Acute , Molecular Sequence Data , Open Reading Frames , Polymerase Chain Reaction , Protein-Tyrosine Kinases/genetics , Rats , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured , src-Family Kinases
12.
Gene ; 155(2): 299-304, 1995 Apr 03.
Article in English | MEDLINE | ID: mdl-7721108

ABSTRACT

The human isoleucyl-tRNA synthetase (IRS)-encoding cDNA, whose primary structure we report here, has an open reading frame (ORF) which encodes a protein of 1262 amino acids (aa) with strong homology to IRS from yeast (53.5%) and Tetrahymena (51.0%) and contains all the major consensus motifs of class-I hydrophobic amino-acyl-tRNA synthetases (aaRS; MRS, LRS, VRS, IRS). However, the human enzyme has an unusually long C-terminal extension composed, in part, of a twice-repeated motif which shows no homology to any reported protein. We also report the presence of a coiled-coil-like motif in the C-terminal half of the protein. The mRNA has an additional exon in the 5'-untranslated region (UTR) which is alternatively spliced, giving rise to two types of mRNA, both of which are expressed in several human tissues. The longer of the two transcripts contains predicted secondary structure in the 5'-UTR which may reduce the translational efficiency of this mRNA. Two possible regulatory elements in the 5'-UTR, an interferon-stimulated response element (ISRE)-like sequence and a short ORF, have been identified. Because human IRS has previously been shown to be the target of antibodies in autoimmune disease, we discuss the role of protein structural features in the development of an autoimmune response to IRS.


Subject(s)
Alternative Splicing , Isoleucine-tRNA Ligase/genetics , Amino Acid Sequence , Base Sequence , DNA, Complementary/genetics , Humans , Isoleucine-tRNA Ligase/chemistry , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Sequence Analysis
13.
Neurology ; 44(6): 1097-100, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8208408

ABSTRACT

Late-onset muscle weakness is rare in glycolytic disorders. There are two reports in the literature of phosphofructokinase (PFK)-deficient Ashkenazi Jews with severe vacuolar myopathy manifesting in late adulthood. The genetic abnormality in these patients is unknown. We report a third patient with a similar syndrome: early-onset exercise intolerance in young childhood and progressive weakness in a limb-girdle distribution appearing at 57 years of age, leading to severe incapacity. Muscle histology showed diffuse vacuolar changes, and muscle fibers contained excess glycogen-like material. Muscle biochemistry was diagnostic for PFK deficiency. DNA analysis from the patient and his family showed that he was homozygous for a recently identified point mutation at the exon 5/intron 5 junction (a G-to-A change); two other family members were heterozygous for this mutation. It is not clear whether late-onset weakness is the natural course for all PFK-deficient patients or whether the exon 5 mutation carries increased risk for this severe myopathy.


Subject(s)
Muscular Diseases/enzymology , Muscular Diseases/genetics , Phosphofructokinase-1/deficiency , Point Mutation , Base Sequence , Exons , Glycolysis , Humans , Introns , Male , Middle Aged , Molecular Sequence Data , Phosphofructokinase-1/genetics
14.
Neurology ; 56(12): 1739-45, 2001 Jun 26.
Article in English | MEDLINE | ID: mdl-11425943

ABSTRACT

BACKGROUND: The authors previously reported the generation of a knockout mouse model of Pompe disease caused by the inherited deficiency of lysosomal acid alpha-glucosidase (GAA). The disorder in the knockout mice (GAA-/-) resembles the human disease closely, except that the clinical symptoms develop late relative to the lifespan of the animals. In an attempt to accelerate the course of the disease in the knockouts, the authors increased the level of cytoplasmic glycogen by overexpressing glycogen synthase (GSase) or GlutI glucose transporter. METHODS: GAA-/- mice were crossed to transgenic mice overexpressing GSase or GlutI in skeletal muscle. RESULTS: Both transgenics on a GAA knockout background (GS/GAA-/- and GlutI/GAA-/-) developed a severe muscle wasting disorder with an early age at onset. This finding, however, is not the major focus of the study. Unexpectedly, the mice bearing the GSase transgene, but not those bearing the GlutI transgene, accumulated structurally abnormal polysaccharide (polyglucosan) similar to that observed in patients with Lafora disease, glycogenosis type IV, and glycogenosis type VII. Ultrastructurally, the periodic acid-Schiff (PAS)-positive polysaccharide inclusions were composed of short, amorphous, irregular branching filaments indistinguishable from classic polyglucosan bodies. The authors show here that increased level of GSase in the presence of normal glycogen branching enzyme (GBE) activity leads to polyglucosan accumulation. The authors have further shown that inactivation of lysosomal acid alpha-glucosidase in the knockout mice does not contribute to the process of polyglucosan formation. CONCLUSIONS: An imbalance between GSase and GBE activities is proposed as the mechanism involved in the production of polyglucosan bodies. The authors may have inadvertently created a "muscle polyglucosan disease" by simulating the mechanism for polyglucosan formation.


Subject(s)
Genetic Engineering , Glucans/genetics , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/pathology , Muscles/pathology , 1,4-alpha-Glucan Branching Enzyme/metabolism , Animals , Disease Models, Animal , Glycogen Storage Disease Type IV/metabolism , Glycogen Synthase/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron , Muscles/ultrastructure
15.
Neuromuscul Disord ; 10(4-5): 283-91, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10838256

ABSTRACT

Glycogen storage disease type II (GSDII) is a recessively inherited disorder caused by defects in lysosomal acid alpha-glucosidase. In an attempt to reproduce the range of clinical manifestations of the human illness we have created null alleles at the acid alpha-glucosidase locus (GAA) with several gene targeting strategies. In each knockout strain, enzyme activity was completely abolished and glycogen accumulated at indistinguishable rates. The phenotypes, however, differed strikingly. Acid alpha-glucosidase deficiency on a 129xC57BL/6 background resulted in a severe phenotype with progressive cardiomyopathy and profound muscle wasting similar to that in patients with glycogen storage disease type II. On a 129/C57BL/6xFVB background, homozygous mutants developed a milder phenotype with a later age of onset. Females were more affected than males irrespective of genetic background. As in humans with glycogen storage disease type II, therefore, other genetic loci affect the phenotypic expression of a single gene mutation.


Subject(s)
Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , alpha-Glucosidases/deficiency , alpha-Glucosidases/genetics , Age Factors , Animals , Disease Models, Animal , Disease Progression , Gene Targeting , Genetic Therapy/methods , Glycogen Storage Disease Type II/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Mutagenesis/genetics , Phenotype , RNA, Messenger/metabolism , Treatment Outcome
16.
Vopr Med Khim ; 26(4): 557-61, 1980.
Article in Russian | MEDLINE | ID: mdl-7456400

ABSTRACT

A method is described for estimation of prostaglandins in human urine. The prostaglandins were isolated from urine using Amberlite HAD-2 and fractionated on a column with silicic acid. For the fractionation of the samples specific markers were required for each column. Two methods of column chromatography were compared. The radioimmunoassay was shown to be an accurate and highly reliable method. The advantages of the method are that PGE2 is converted into PGF2 alpha using sodium borhydride and the latter is estimated in a reaction with the specific antiserum supplied with the Kit.


Subject(s)
Prostaglandins/urine , Chromatography/methods , Humans , Prostaglandins E/urine , Prostaglandins F/urine , Prostaglandins G/urine , Radioimmunoassay
SELECTION OF CITATIONS
SEARCH DETAIL