Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Eur J Med Genet ; 62(2): 144-148, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29981851

ABSTRACT

Congenital analbuminemia (OMIM # 616000) is an extremely rare autosomal recessive disorder, caused by variations in the albumin gene (ALB), which is generally thought to be a relatively benign condition in adulthood, but seems to be potentially life threatening in the pre- and peri-natal period. The subject of our study was a consanguineous family, in which we identified two analbuminemic individuals. Mutation analysis of ALB revealed that both are homozygous for a previously unreported insertion in exon 9 (c.1098dupT), causing a subsequent frame-shift with the generation of a premature stop codon, and an aberrant truncated putative protein product, p.Val367fsTer12. This variation is present in heterozygous condition in several other members of the family. The phenotype and the molecular genetics of CAA are discussed.


Subject(s)
Hypoalbuminemia/genetics , Mutation , Serum Albumin, Human/genetics , Adult , Aged , Codon, Terminator , Consanguinity , Female , Humans , Hypoalbuminemia/pathology , Male , Pedigree , Phenotype
2.
J Med Chem ; 58(11): 4483-93, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25984755

ABSTRACT

In order to identify structural features of pyrimethamine (5-(4-chlorophenyl)-6-ethylpyrimidine-2,4-diamine) that contribute to its inhibitory activity (IC50 value) and chaperoning efficacy toward ß-N-acetylhexosaminidase, derivatives of the compound were synthesized that differ at the positions bearing the amino, ethyl, and chloro groups. Whereas the amino groups proved to be critical to its inhibitory activity, a variety of substitutions at the chloro position only increased its IC50 by 2-3-fold. Replacing the ethyl group at the 6-position with butyl or methyl groups increased IC50 more than 10-fold. Surprisingly, despite its higher IC50, a derivative lacking the chlorine atom in the para-position was found to enhance enzyme activity in live patient cells a further 25% at concentrations >100 µM, while showing less toxicity. These findings demonstrate the importance of the phenyl group in modulating the chaperoning efficacy and toxicity profile of the derivatives.


Subject(s)
Mutant Proteins/metabolism , Mutation/genetics , Pyrimethamine/chemistry , Pyrimethamine/metabolism , beta-N-Acetylhexosaminidases/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Models, Molecular , Molecular Structure , Mutant Proteins/genetics , Structure-Activity Relationship
3.
Anal Biochem ; 458: 20-6, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24769373

ABSTRACT

GM2 gangliosidosis is a group of inherited neurodegenerative disorders resulting primarily from the excessive accumulation of GM2 gangliosides (GM2) in neuronal cells. As biomarkers for categorising patients and monitoring the effectiveness of developing therapies are lacking for this group of disorders, we sought to develop methodology to quantify GM2 levels in more readily attainable patient samples such as plasma, leukocytes, and cultured skin fibroblasts. Following organic extraction, gangliosides were partitioned into the aqueous phase and isolated using C18 solid-phase extraction columns. Relative quantification of three species of GM2 was achieved using LC/ESI-MS/MS with d35GM1 18:1/18:0 as an internal standard. The assay was linear over the biological range, and all GM2 gangliosidosis patients were demarcated from controls by elevated GM2 in cultured skin fibroblast extracts. However, in leukocytes only some molecular species could be used for differentiation and in plasma only one was informative. A reduction in GM2 was easily detected in patient skin fibroblasts after a short treatment with media from normal cells enriched in secreted ß-hexosaminidase. This method may show promise for measuring the effectiveness of experimental therapies for GM2 gangliosidosis by allowing quantification of a reduction in the primary storage burden.


Subject(s)
Chromatography, High Pressure Liquid , G(M2) Ganglioside/analysis , Tandem Mass Spectrometry , Cell Line , Fibroblasts/chemistry , G(M2) Ganglioside/blood , G(M2) Ganglioside/isolation & purification , Humans , Leukocytes/chemistry , Solid Phase Extraction , Spectrometry, Mass, Electrospray Ionization , beta-N-Acetylhexosaminidases/metabolism
4.
Mol Genet Metab ; 107(1-2): 203-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22784478

ABSTRACT

Deficiencies of lysosomal ß-D-galactosidase can result in GM1 gangliosidosis, a severe neurodegenerative disease characterized by massive neuronal storage of GM1 ganglioside in the brain. Currently there are no available therapies that can even slow the progression of this disease. Enzyme enhancement therapy utilizes small molecules that can often cross the blood brain barrier, but are also often competitive inhibitors of their target enzyme. It is a promising new approach for treating diseases, often caused by missense mutations, associated with dramatically reduced levels of functionally folded enzyme. Despite a number of positive reports based on assays performed with patient cells, skepticism persists that an inhibitor-based treatment can increase mutant enzyme activity in vivo. To date no appropriate animal model, i.e., one that recapitulates a responsive human genotype and clinical phenotype, has been reported that could be used to validate enzyme enhancement therapy. In this report, we identify a novel enzyme enhancement-agent, N-nonyl-deoxygalactonojirimycin, that enhances the mutant ß-galactosidase activity in the lysosomes of a number of patient cell lines containing a variety of missense mutations. We then demonstrate that treatment of cells from a previously described, naturally occurring feline model (that biochemically, clinically and molecularly closely mimics GM1 gangliosidosis in humans) with this molecule, results in a robust enhancement of their mutant lysosomal ß-galactosidase activity. These data indicate that the feline model could be used to validate this therapeutic approach and determine the relationship between the disease stage at which this therapy is initiated and the maximum clinical benefits obtainable.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enzyme Replacement Therapy , Gangliosidosis, GM1/metabolism , Mutant Proteins/metabolism , beta-Galactosidase/metabolism , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/pharmacology , Animals , Cats , Cell Line, Tumor , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Gangliosidosis, GM1/drug therapy , Gangliosidosis, GM1/genetics , Hot Temperature , Humans , Hydrogen-Ion Concentration , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutation , Protein Denaturation/drug effects , Treatment Outcome , beta-Galactosidase/antagonists & inhibitors , beta-Galactosidase/chemistry
5.
PLoS One ; 6(9): e24951, 2011.
Article in English | MEDLINE | ID: mdl-21957468

ABSTRACT

Heparin acetyl-CoA:alpha-glucosaminide N-acetyltransferase (N-acetyltransferase, EC 2.3.1.78) is an integral lysosomal membrane protein containing 11 transmembrane domains, encoded by the HGSNAT gene. Deficiencies of N-acetyltransferase lead to mucopolysaccharidosis IIIC. We demonstrate that contrary to a previous report, the N-acetyltransferase signal peptide is co-translationally cleaved and that this event is required for its intracellular transport to the lysosome. While we confirm that the N-acetyltransferase precursor polypeptide is processed in the lysosome into a small amino-terminal alpha- and a larger ß- chain, we further characterize this event by identifying the mature amino-terminus of each chain. We also demonstrate this processing step(s) is not, as previously reported, needed to produce a functional transferase, i.e., the precursor is active. We next optimize the biochemical assay procedure so that it remains linear as N-acetyltransferase is purified or protein-extracts containing N-acetyltransferase are diluted, by the inclusion of negatively charged lipids. We then use this assay to demonstrate that the purified single N-acetyltransferase protein is both necessary and sufficient to express transferase activity, and that N-acetyltransferase functions as a monomer. Finally, the kinetic mechanism of action of purified N-acetyltransferase was evaluated and found to be a random sequential mechanism involving the formation of a ternary complex with its two substrates; i.e., N-acetyltransferase does not operate through a ping-pong mechanism as previously reported. We confirm this conclusion by demonstrating experimentally that no acetylated enzyme intermediate is formed during the reaction.


Subject(s)
Acetyltransferases/biosynthesis , Acetyltransferases/metabolism , Mucopolysaccharidosis III/enzymology , Protein Processing, Post-Translational , Acetyltransferases/deficiency , Acetyltransferases/isolation & purification , Blotting, Western , Detergents/chemistry , Endoplasmic Reticulum/metabolism , Enzyme Assays , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Protein Transport , Temperature
6.
J Biol Chem ; 286(24): 21413-26, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21518770

ABSTRACT

Mammalian glycosphingolipid (GSL) precursor monohexosylceramides are either glucosyl- or galactosylceramide (GlcCer or GalCer). Most GSLs derive from GlcCer. Substitution of the GSL fatty acid with adamantane generates amphipathic mimics of increased water solubility, retaining receptor function. We have synthesized adamantyl GlcCer (adaGlcCer) and adamantyl GalCer (adaGalCer). AdaGlcCer and adaGalCer partition into cells to alter GSL metabolism. At low dose, adaGlcCer increased cellular GSLs by inhibition of glucocerebrosidase (GCC). Recombinant GCC was inhibited at pH 7 but not pH 5. In contrast, adaGalCer stimulated GCC at pH 5 but not pH 7 and, like adaGlcCer, corrected N370S mutant GCC traffic from the endoplasmic reticulum to lysosomes. AdaGalCer reduced GlcCer levels in normal and lysosomal storage disease (LSD) cells. At 40 µM adaGlcCer, lactosylceramide (LacCer) synthase inhibition depleted LacCer (and more complex GSLs), such that only GlcCer remained. In Vero cell microsomes, 40 µM adaGlcCer was converted to adaLacCer, and LacCer synthesis was inhibited. AdaGlcCer is the first cell LacCer synthase inhibitor. At 40 µM adaGalCer, cell synthesis of only Gb(3) and Gb(4) was significantly reduced, and a novel product, adamantyl digalactosylceramide (adaGb(2)), was generated, indicating substrate competition for Gb(3) synthase. AdaGalCer also inhibited cell sulfatide synthesis. Microsomal Gb(3) synthesis was inhibited by adaGalCer. Metabolic labeling of Gb(3) in Fabry LSD cells was selectively reduced by adaGalCer, and adaGb(2) was produced. AdaGb(2) in cells was 10-fold more effectively shed into the medium than the more polar Gb(3), providing an easily eliminated "safety valve" alternative to Gb(3) accumulation. Adamantyl monohexosyl ceramides thus provide new tools to selectively manipulate normal cellular GSL metabolism and reduce GSL accumulation in cells from LSD patients.


Subject(s)
Gene Expression Regulation , Glycosphingolipids/metabolism , Animals , Cattle , Chlorocebus aethiops , Fibroblasts/metabolism , Humans , Hydrogen-Ion Concentration , Lysosomal Storage Diseases/metabolism , Lysosomes/metabolism , Microsomes/metabolism , Mutation , Recombinant Proteins/metabolism , Vero Cells , alpha-Galactosidase/metabolism
7.
Mol Genet Metab ; 102(1): 6-12, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20926324

ABSTRACT

Late-onset GM2 gangliosidosis is an autosomal recessive, neurodegenerative, lysosomal storage disease, caused by deficiency of ß-hexosaminidase A (Hex A), resulting from mutations in the HEXA (Tay-Sachs variant) or the HEXB (Sandhoff variant) genes. The enzyme deficiency in many patients with juvenile or adult onset forms of the disease results from the production of an unstable protein, which becomes targeted for premature degradation by the quality control system of the smooth endoplasmic reticulum and is not transported to lysosomes. In vitro studies have shown that many mutations in either the α or ß subunit of Hex A can be partially rescued, i.e. enhanced levels of both enzyme protein and activity in lysosomes, following the growth of patient cells in the presence of the drug, pyrimethamine. The objectives of the present clinical trial were to establish the tolerability and efficacy of the treatment of late-onset GM2 gangliosidosis patients with escalating doses of pyrimethamine, to a maximum of 100 mg per day, administered orally in a single daily dose, over a 16-week period . The primary objective, tolerability, was assessed by regular clinical examinations, along with a panel of hematologic and biochemical studies. Although clinical efficacy could not be assessed in this short trial, treatment efficacy was evaluated by repeated measurements of leukocyte Hex A activity, expressed relative to the activity of lysosomal ß-glucuronidase. A total of 11 patients were enrolled, 8 males and 3 females, aged 23 to 50 years. One subject failed the initial screen, another was omitted from analysis because of the large number of protocol violations, and a third was withdrawn very early as a result of adverse events which were not drug-related. For the remaining 8 subjects, up to a 4-fold enhancement of Hex A activity at doses of 50 mg per day or less was observed. Additionally marked individual variations in the pharmacokinetics of the drug among the patients were noted. However, the study also found that significant side effects were experienced by most patients at or above 75 mg pyrimethamine per day. We concluded that pyrimethamine treatment enhances leukocyte Hex A activity in patients with late-onset GM2 gangliosidosis at doses lower than those associated with unacceptable side effects. Further plans are underway to extend these trials and to develop methods to assess clinical efficacy.


Subject(s)
Gangliosidoses, GM2/drug therapy , Pyrimethamine/therapeutic use , Adult , Enzyme Assays , Female , Glucosylceramidase/blood , Hexosaminidase A/blood , Hexosaminidase B/blood , Humans , Male , Middle Aged , Pyrimethamine/adverse effects , Pyrimethamine/blood , Young Adult , beta-Galactosidase/blood
8.
Chembiochem ; 11(14): 2026-33, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20715263

ABSTRACT

A collection of new reversible glycosidase inhibitors of the iminoalditol type featuring N-substituents containing perfluorinated regions has been prepared for evaluation of physicochemical, biochemical and diagnostic properties. The vast variety of feasible oligofluoro moieties allows for modular approaches to customised structures according to the intended applications, which are influenced by the fluorine content as well as the distance of the fluorous moiety from the ring nitrogen. The first examples, in particular in the D-galacto series, exhibited excellent inhibitory activities. A preliminary screen with two human cell lines showed that, at subinhibitory concentrations, they are powerful pharmacological chaperones enhancing the activities of the catalytically handicapped lysosomal D-galactosidase mutants associated with GM1 gangliosidosis and Morquio B disease.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Galactosidases/antagonists & inhibitors , Gangliosidosis, GM1/drug therapy , Sugar Alcohols/chemistry , Sugar Alcohols/pharmacology , Cell Line , Coffee/enzymology , Enzyme Inhibitors/therapeutic use , Escherichia coli/enzymology , Fibroblasts/drug effects , Fibroblasts/enzymology , Galactosidases/metabolism , Halogenation , Humans , Imines/chemistry , Imines/pharmacology , Imines/therapeutic use , Rhizobium/enzymology , Sugar Alcohols/therapeutic use
9.
Beilstein J Org Chem ; 6: 21, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20502610

ABSTRACT

N-Alkylation at the ring nitrogen of the D-galactosidase inhibitor 1-deoxygalactonojirimycin with a functionalised C 6alkyl chain followed by modification with different aromatic substituents provided lipophilic 1-deoxygalactonojirimycin derivatives which exhibit inhibitory properties against ß-glycosidases from E. coli and Agrobacterium sp. as well as green coffee bean α-galactosidase. In preliminary studies, these compounds also showed potential as chemical chaperones for GM1-gangliosidosis related ß-galactosidase mutants.

10.
Carbohydr Res ; 345(10): 1371-6, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20471633

ABSTRACT

Cyclization by double reductive amination of d-xylo-hexos-5-ulose with methyl 6-aminohexanoate gave (methoxycarbonyl)pentyl-1-deoxynojirimycin. Reaction of the terminal carboxylic acid with N-dansyl-1,6-diaminohexane provided the corresponding chain-extended fluorescent derivative. By reaction with bis(6-dansylaminohexyl)amine, the corresponding branched di-N-dansyl compound was obtained. Both compounds are strong inhibitors of d-glucosidases and could also be shown to distinctly improve, at sub-inhibitory concentrations, the activity of beta-glucocerebrosidase in a Gaucher fibroblast (N370S) cell-line through chaperoning of the enzyme to the lysosome.


Subject(s)
1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gaucher Disease/pathology , Nitrogen/chemistry , Phosphatidylcholines/chemistry , 1-Deoxynojirimycin/chemical synthesis , Cell Line , Enzyme Inhibitors/chemical synthesis , Fibroblasts/pathology , Glucosidases/antagonists & inhibitors , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Rhizobium/enzymology , Saccharomyces cerevisiae/enzymology
11.
FEBS J ; 277(7): 1618-38, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20148966

ABSTRACT

Gaucher disease is caused by mutations in the gene that encodes the lysosomal enzyme acid beta-glucosidase (GCase). We have shown previously that the small molecule pharmacological chaperone isofagomine (IFG) binds and stabilizes N370S GCase, resulting in increased lysosomal trafficking and cellular activity. In this study, we investigated the effect of IFG on L444P GCase. Incubation of Gaucher patient-derived lymphoblastoid cell lines (LCLs) or fibroblasts with IFG led to approximately 3.5- and 1.3-fold increases in L444P GCase activity, respectively, as measured in cell lysates. The effect in fibroblasts was increased approximately 2-fold using glycoprotein-enrichment, GCase-immunocapture, or by incubating cells overnight in IFG-free media prior to assay, methods designed to maximize GCase activity by reducing IFG carryover and inhibition in the enzymatic assay. IFG incubation also increased the lysosomal trafficking and in situ activity of L444P GCase in intact cells, as measured by reduction in endogenous glucosylceramide levels. Importantly, this reduction was seen only following three-day incubation in IFG-free media, underscoring the importance of IFG removal to restore lysosomal GCase activity. In mice expressing murine L444P GCase, oral administration of IFG resulted in significant increases (2- to 5-fold) in GCase activity in disease-relevant tissues, including brain. Additionally, eight-week IFG administration significantly lowered plasma chitin III and IgG levels, and 24-week administration significantly reduced spleen and liver weights. Taken together, these data suggest that IFG can increase the lysosomal activity of L444P GCase in cells and tissues. Moreover, IFG is orally available and distributes into multiple tissues, including brain, and may thus merit therapeutic evaluation for patients with neuronopathic and non-neuronopathic Gaucher disease.


Subject(s)
Gaucher Disease/genetics , Imino Pyranoses/chemistry , Lysosomal Storage Diseases/genetics , Mutation , beta-Glucosidase/genetics , Animals , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Glucosylceramidase/metabolism , Humans , Male , Mice , Microscopy, Confocal/methods , Molecular Chaperones/metabolism , Rats , Rats, Sprague-Dawley
12.
Glycobiology ; 20(3): 356-65, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19917668

ABSTRACT

Enzyme enhancement therapy, utilizing small molecules as pharmacological chaperones, is an attractive approach for the treatment of lysosomal storage diseases that are associated with protein misfolding. However, pharmacological chaperones are also inhibitors of their target enzyme. Thus, a major concern with this approach is that, despite enhancing protein folding within, and intracellular transport of the functional mutant enzyme out of the endoplasmic reticulum, the chaperone will continue to inhibit the enzyme in the lysosome, preventing substrate clearance. Here we demonstrate that the in vitro hydrolysis of a fluorescent derivative of lyso-GM2 ganglioside, like natural GM2 ganglioside, is specifically carried out by the beta-hexosaminidase A isozyme, requires the GM2 activator protein as a co-factor, increases when the derivative is incorporated into anionic liposomes and follows similar Michaelis-Menten kinetics. This substrate can also be used to differentiate between lysates from normal and GM2 activator-deficient cells. When added to the growth medium of cells, the substrate is internalized and primarily incorporated into lysosomes. Utilizing adult Tay-Sachs fibroblasts that have been pre-treated with the pharmacological chaperone Pyrimethamine and subsequently loaded with this substrate, we demonstrate an increase in both the levels of mutant beta-hexosaminidase A and substrate-hydrolysis as compared to mock-treated cells.


Subject(s)
Fluorescent Antibody Technique/methods , G(M2) Ganglioside/analysis , G(M2) Ganglioside/metabolism , Cells, Cultured , G(M2) Ganglioside/analogs & derivatives , Humans , Hydrolysis , Kinetics , Liposomes/metabolism , Mass Spectrometry , Tay-Sachs Disease
13.
J Biol Chem ; 284(35): 23502-16, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19578116

ABSTRACT

Gaucher disease (GD), the most prevalent lysosomal storage disease, is caused by a deficiency of glucocerebrosidase (GCase). The identification of small molecules acting as agents for enzyme enhancement therapy is an attractive approach for treating different forms of GD. A thermal denaturation assay utilizing wild type GCase was developed to screen a library of 1,040 Food and Drug Administration-approved drugs. Ambroxol (ABX), a drug used to treat airway mucus hypersecretion and hyaline membrane disease in newborns, was identified and found to be a pH-dependent, mixed-type inhibitor of GCase. Its inhibitory activity was maximal at neutral pH, found in the endoplasmic reticulum, and undetectable at the acidic pH of lysosomes. The pH dependence of ABX to bind and stabilize the enzyme was confirmed by monitoring the rate of hydrogen/deuterium exchange at increasing guanidine hydrochloride concentrations. ABX treatment significantly increased N370S and F213I mutant GCase activity and protein levels in GD fibroblasts. These increases were primarily confined to the lysosome-enriched fraction of treated cells, a finding confirmed by confocal immunofluorescence microscopy. Additionally, enhancement of GCase activity and a reduction in glucosylceramide storage was verified in ABX-treated GD lymphoblasts (N370S/N370S). Hydrogen/deuterium exchange mass spectrometry revealed that upon binding of ABX, amino acid segments 243-249, 310-312, and 386-400 near the active site of GCase are stabilized. Consistent with its mixed-type inhibition of GCase, modeling studies indicated that ABX interacts with both active and non-active site residues. Thus, ABX has the biochemical characteristics of a safe and effective enzyme enhancement therapy agent for the treatment of patients with the most common GD genotypes.


Subject(s)
Ambroxol/chemistry , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Gaucher Disease/enzymology , Glucosylceramidase/antagonists & inhibitors , Ambroxol/pharmacology , Amino Acid Sequence , Binding Sites , Catalytic Domain , Cells, Cultured , Enzyme Inhibitors/pharmacology , Enzyme Stability , Fibroblasts/drug effects , Fibroblasts/enzymology , Gaucher Disease/drug therapy , Glucosylceramidase/chemistry , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Humans , Molecular Conformation , Molecular Sequence Data
14.
Biochem Biophys Res Commun ; 385(4): 576-80, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19486886

ABSTRACT

Platelet activating factor (PAF), an endogenous bioactive phospholipid, has been documented as a pivotal mediator in the inflammatory cascade underlying the pathogenesis of many diseases including necrotizing enterocolitis. Much effort has been directed towards finding an effective in vivo inhibitor of PAF signaling. Here, we report that a small, highly stable, lysosomal lipid transport protein, the GM2 activator protein (GM2AP) is able to inhibit the inflammatory processes otherwise initiated by PAF in a rat model of necrotizing enterocolitis. Based on behavioral observations, gross anatomical observations at necropsy, histopathology and immunocytochemistry, the administration of recombinant GM2AP inhibits the devastating gastrointestinal necrosis resulting from the injection of rats with LPS and PAF. Recombinant GM2AP treatment not only markedly decrease tissue destruction, but also helped to maintain tight junction integrity at the gastrointestinal level as judged by contiguous Zonula Occludens-1 staining of the epithelial layer lining the crypts.


Subject(s)
Enterocolitis, Necrotizing/drug therapy , G(M2) Activator Protein/administration & dosage , Platelet Activating Factor/antagonists & inhibitors , Recombinant Proteins/administration & dosage , Animals , Disease Models, Animal , Enterocolitis, Necrotizing/chemically induced , Enterocolitis, Necrotizing/etiology , Enterocolitis, Necrotizing/pathology , Male , Platelet Activating Factor/metabolism , Platelet Activating Factor/toxicity , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
15.
Mol Genet Metab ; 96(4): 225-32, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19167257

ABSTRACT

Recently, inhibition of L-type Ca(2+) channels, using either Diltiazem or Verapamil, has been reported to partially restore mutant glucocerebrosidase activity in cells from patients with Gaucher disease homozygous for the N370S or L444P alleles, as well as cells from patients with two other lysosomal storage diseases. It was hypothesized that these drugs act on the endoplasmic reticulum, increasing its folding efficiency, inhibited due to altered calcium homeostasis. Several other laboratories have reported that cells carrying either the N370S or the F213I alleles are amenable to enzyme enhancement therapy with pharmacological chaperones, whereas cells homozygous for L444P respond poorly. We found that Verapamil treatment does not enhance mutant enzyme activity in any of the cell lines tested, while Diltiazem moderately increases activity in normal cells, and in N370S/N370S and F213I/L444P, but not in L444P/L444P Gaucher cells, or in either of two adult Tay-Sachs disease cell lines. Since the mode of action of pharmacological chaperones and Diltiazem are believed to be different, we examined the possibility that they could act in concert. Diltiazem co-administered with known chaperones failed to increase enzyme activities above that reached by chaperone-treatment alone in any of the patient cell lines. Thus, we re-examined the possibility that Diltiazem acts as a pharmacological chaperone. We found that, at the acidic pH of lysosomes, Diltiazem was not an inhibitor, nor did its presence increase the heat stability of glucocerebrosidase. However, at neutral pH, found in the endoplasmic reticulum, Diltiazem exhibited both of these properties. Thus Diltiazem exhibits the biochemical characteristics of a glucocerebrosidase pharmacological chaperone.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Diltiazem/pharmacology , Diltiazem/therapeutic use , Gaucher Disease/drug therapy , Gaucher Disease/pathology , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/enzymology , Gaucher Disease/enzymology , Glucosylceramidase/antagonists & inhibitors , Glucosylceramidase/metabolism , Hot Temperature , Humans , Hydrogen-Ion Concentration/drug effects , Imino Pyranoses/pharmacology , Lysosomes/drug effects , Lysosomes/enzymology , Phenotype , Verapamil/pharmacology
16.
Chembiochem ; 9(16): 2650-62, 2008 Nov 03.
Article in English | MEDLINE | ID: mdl-18972510

ABSTRACT

Point mutations in beta-glucocerebrosidase (GCase) can result in a deficiency of both GCase activity and protein in lysosomes thereby causing Gaucher Disease (GD). Enzyme inhibitors such as isofagomine, acting as pharmacological chaperones (PCs), increase these levels by binding and stabilizing the native form of the enzyme in the endoplasmic reticulum (ER), and allow increased lysosomal transport of the enzyme. A high-throughput screen of the 50,000-compound Maybridge library identified two, non-carbohydrate-based inhibitory molecules, a 2,4-diamino-5-substituted quinazoline (IC(50) 5 microM) and a 5-substituted pyridinyl-2-furamide (IC(50) 8 microM). They raised the levels of functional GCase 1.5-2.5-fold in N370S or F213I GD fibroblasts. Immunofluorescence confirmed that treated GD fibroblasts had decreased levels of GCase in their ER and increased levels in lysosomes. Changes in protein dynamics, monitored by hydrogen/deuterium-exchange mass spectrometry, identified a domain III active-site loop (residues 243-249) as being significantly stabilized upon binding of isofagomine or either of these two new compounds; this suggests a common mechanism for PC enhancement of intracellular transport.


Subject(s)
Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Gaucher Disease/enzymology , Glucosylceramidase/antagonists & inhibitors , Animals , Cattle , Cell Line , Deuterium Exchange Measurement , Enzyme Inhibitors/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Gaucher Disease/pathology , Glucosylceramidase/chemistry , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Humans , Hydrogen-Ion Concentration , Lysosomes/drug effects , Lysosomes/enzymology , Mass Spectrometry , Mutation , Protein Conformation/drug effects , Protein Stability/drug effects , Substrate Specificity
17.
Anal Biochem ; 381(2): 276-8, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18619939

ABSTRACT

The low levels of human lysosomal glucocerebrosidase activity expressed in transiently transfected Chinese hamster ovary (CHO) cells were investigated. Reverse transcription PCR (RT-PCR) demonstrated that a significant portion of the transcribed RNA was misspliced owing to the presence of a cryptic splice site in the complementary DNA (cDNA). Missplicing results in the deletion of 179 bp of coding sequence and a premature stop codon. A repaired cDNA was constructed abolishing the splice site without changing the amino acid sequence. The level of glucocerebrosidase expression was increased sixfold. These data demonstrate that for maximum expression of any cDNA construct, the transcription products should be examined.


Subject(s)
Alternative Splicing , Glucosylceramidase/genetics , RNA Splice Sites , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , DNA Mutational Analysis , DNA, Complementary , Gaucher Disease/etiology , Gaucher Disease/genetics , Glucosylceramidase/isolation & purification , Glucosylceramidase/metabolism , Humans , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction
18.
Mol Genet Metab ; 94(2): 212-21, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18353697

ABSTRACT

G(M1) gangliosidosis is an inherited, fatal neurodegenerative disease caused by deficiency of lysosomal beta-d-galactosidase (EC 3.2.1.23) and consequent storage of undegraded G(M1) ganglioside. To characterize the genetic mutation responsible for feline G(M1) gangliosidosis, the normal sequence of feline beta-galactosidase cDNA first was defined. The feline beta-galactosidase open reading frame is 2010 base pairs, producing a protein of 669 amino acids. The putative signal sequence consists of amino acids 1-24 of the beta-galactosidase precursor protein, which contains seven potential N-linked glycosylation sites, as in the human protein. Overall sequence homology between feline and human beta-galactosidase is 74% for the open reading frame and 82% for the amino acid sequence. After normal beta-galactosidase was sequenced, the mutation responsible for feline G(M1) gangliosidosis was defined as a G to C substitution at position 1448 of the open reading frame, resulting in an amino acid substitution at arginine 483, known to cause G(M1) gangliosidosis in humans. Feline beta-galactosidase messenger RNA levels were normal in cerebral cortex, as determined by quantitative RT-PCR assays. Although enzymatic activity is severely reduced by the mutation, a full-length feline beta-galactosidase cDNA restored activity in transfected G(M1) fibroblasts to 18-times normal. beta-Galactosidase protein levels in G(M1) tissues were normal on Western blots, but immunofluorescence analysis demonstrated that the majority of mutant beta-galactosidase protein did not reach the lysosome. Additionally, G(M1) cat fibroblasts demonstrated increased expression of glucose-related protein 78/BiP and protein disulfide isomerase, suggesting that the unfolded protein response plays a role in pathogenesis of feline G(M1) gangliosidosis.


Subject(s)
Cat Diseases/genetics , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/veterinary , Mutation, Missense , beta-Galactosidase/genetics , Amino Acid Substitution , Animals , Cat Diseases/enzymology , Cats , Cell Line , Cells, Cultured , Cloning, Molecular , Endoplasmic Reticulum Chaperone BiP , Fibroblasts/enzymology , Gangliosidosis, GM1/enzymology , Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/metabolism , Molecular Sequence Data , Open Reading Frames , Protein Disulfide-Isomerases/metabolism , Protein Transport , beta-Galactosidase/analysis , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...