Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Cell Rep ; 43(5): 114117, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38630590

ABSTRACT

Endoplasmic reticulum-plasma membrane (ER-PM) junctions mediate Ca2+ flux across neuronal membranes. The properties of these membrane contact sites are defined by their lipid content, but little attention has been given to glycosphingolipids (GSLs). Here, we show that GM1-ganglioside, an abundant GSL in neuronal membranes, is integral to ER-PM junctions; it interacts with synaptic proteins/receptors and regulates Ca2+ signaling. In a model of the neurodegenerative lysosomal storage disease, GM1-gangliosidosis, pathogenic accumulation of GM1 at ER-PM junctions due to ß-galactosidase deficiency drastically alters neuronal Ca2+ homeostasis. Mechanistically, we show that GM1 interacts with the phosphorylated N-methyl D-aspartate receptor (NMDAR) Ca2+ channel, thereby increasing Ca2+ flux, activating extracellular signal-regulated kinase (ERK) signaling, and increasing the number of synaptic spines without increasing synaptic connectivity. Thus, GM1 clustering at ER-PM junctions alters synaptic plasticity and worsens the generalized neuronal cell death characteristic of GM1-gangliosidosis.


Subject(s)
Calcium Signaling , Endoplasmic Reticulum , G(M1) Ganglioside , Gangliosidosis, GM1 , Receptors, N-Methyl-D-Aspartate , Animals , Humans , Mice , Calcium/metabolism , Cell Membrane/metabolism , Dendritic Spines/metabolism , Disease Models, Animal , Endoplasmic Reticulum/metabolism , G(M1) Ganglioside/metabolism , Gangliosidosis, GM1/metabolism , Gangliosidosis, GM1/pathology , Neuronal Plasticity , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Male , Female
2.
PLoS One ; 18(11): e0294437, 2023.
Article in English | MEDLINE | ID: mdl-38019733

ABSTRACT

Site-directed Enzyme Enhancement Therapy (SEE-Tx®) technology is a disease-agnostic drug discovery tool that can be applied to any protein target of interest with a known three-dimensional structure. We used this proprietary technology to identify and characterize the therapeutic potential of structurally targeted allosteric regulators (STARs) of the lysosomal hydrolase ß-galactosidase (ß-Gal), which is deficient due to gene mutations in galactosidase beta 1 (GLB1)-related lysosomal storage disorders (LSDs). The biochemical HaloTag cleavage assay was used to monitor the delivery of wildtype (WT) ß-Gal and four disease-related ß-Gal variants (p.Ile51Thr, p.Arg59His, p.Arg201Cys and p.Trp273Leu) in the presence and absence of two identified STAR compounds. In addition, the ability of STARs to reduce toxic substrate was assessed in a canine fibroblast cell model. In contrast to the competitive pharmacological chaperone N-nonyl-deoxygalactonojirimycin (NN-DGJ), the two identified STAR compounds stabilized and substantially enhanced the lysosomal transport of wildtype enzyme and disease-causing ß-Gal variants. In addition, the two STAR compounds reduced the intracellular accumulation of exogenous GM1 ganglioside, an effect not observed with the competitive chaperone NN-DGJ. This proof-of-concept study demonstrates that the SEE-Tx® platform is a rapid and cost-effective drug discovery tool for identifying STARs for the treatment of LSDs. In addition, the HaloTag assay developed in our lab has proved valuable in investigating the effect of STARs in promoting enzyme transport and lysosomal delivery. Automatization and upscaling of this assay would be beneficial for screening STARs as part of the drug discovery process.


Subject(s)
Gangliosidosis, GM1 , Lysosomal Storage Diseases , Animals , Dogs , Gangliosidosis, GM1/drug therapy , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , 1-Deoxynojirimycin/pharmacology , beta-Galactosidase/metabolism
3.
Curr Gene Ther ; 22(4): 352-365, 2022.
Article in English | MEDLINE | ID: mdl-35249485

ABSTRACT

BACKGROUND: GM1 gangliosidosis (GM1) is an autosomal recessive disorder characterized by the deficiency of beta-galactosidase (ß-gal), a ubiquitous lysosomal enzyme that catalyzes the hydrolysis of GM1 ganglioside. OBJECTIVE: The study aims to explore the application of the AAV9-coGLB1 for effective treatment in a GM1 gangliosidosis mutant mouse model. METHODS: We designed a novel adeno-associated virus 9 (AAV9) vector expressing ß-gal (AAV9- coGLB1) to treat GM1 gangliosidosis. The vector, injected via the caudal vein at 4 weeks of age, drove the widespread and sustained expression of ß-gal for up to 32 weeks in the Glb1G455R/G455R mutant mice (GM1 mice). RESULTS: The increased levels of ß-gal reduced the pathological damage occurring in GM1 mice. Histological analyses showed that myelin deficits and neuron-specific pathology were reduced in the cerebral cortex region of AAV9-coGLB1-treated mice. Immunohistochemical staining showed that the accumulation of GM1 ganglioside was also reduced after gene therapy. The reduction of the storage in these regions was accompanied by a decrease in activated microglia. In addition, AAV9 treatment reversed the blockade of autophagic flux in GM1 mice. CONCLUSION: These results show that AAV9-coGLB1 reduces the pathological signs of GM1 gangliosidosis in a mouse model.


Subject(s)
Gangliosidosis, GM1 , Animals , Central Nervous System , Dependovirus/genetics , Disease Models, Animal , G(M1) Ganglioside , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , Gangliosidosis, GM1/therapy , Inflammation/genetics , Inflammation/therapy , Lysosomes/genetics , Lysosomes/pathology , Mice
4.
Metab Brain Dis ; 36(3): 499-508, 2021 03.
Article in English | MEDLINE | ID: mdl-33394287

ABSTRACT

Lysosomal storage diseases comprise different forms of autosomal recessive disorders from which GM1 gangliosidosis has categorized by the accumulation of complex glycolipids associated with a range of progressive neurologic phenotypes. GM1 gangliosidosis is an inherited disorder that progressively destroys nerve cells (neurons) in the brain and spinal cord. GM1 has three main types of onsets, namely infantile (type I), juvenile (type II), and adult (type III) forms. This study provides a series of computational methods that examine the mutations that occurred in GLB1 protein. Initially, the mutational analysis started with 689 amino acid variants for a sequence-based screening and it was done with quite a few In-silico tools to narrow down the most significant variants by utilizing the standard tools; namely, Evolutionary analysis (77 variants), Pathogenicity prediction (44 variants), Stability predictions (30 variants), Biophysical functions (19 variants) and according to the binding site of protein structure with PDB ID 3THC, seven variants (Y83D, Y83H, Y270S, Y270D, W273R, W273D, and Y333H) were narrowed down. Structure based analysis was performed to understand the interacting profile of the native protein and variants with Miglustat; which is the currently used FDA drug as an alternative to enzyme replacement therapy. Molecular Docking study was done to analyze the protein interaction with Miglustat (ligand), as a result native (3THC) structure had a binding affinity of -8.18 kcal/mol and two variant structures had an average binding affinities of -2.61 kcal/mol (Y83D) and - 7.63 kcal/mol (Y270D). Finally, Molecular Dynamics Simulation was performed to know the mutational activity of the protein structures on Miglustat for 50,000 ps. The Y83D variant showed higher deviation than native protein and Y270D in all trajectory analysis. The analysis was done to the protein structures to check the structural variations happened through simulations. This study aids to understand the most deleterious mutants, the activity of the drug to the protein structure and also gives an insight on the stability of the drug with the native and selected variants.


Subject(s)
Gangliosidosis, GM1/metabolism , Mutation , Phenotype , beta-Galactosidase/metabolism , Amino Acid Sequence , DNA Mutational Analysis , Gangliosidosis, GM1/genetics , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , beta-Galactosidase/genetics
5.
Hum Gene Ther ; 31(21-22): 1169-1177, 2020 11.
Article in English | MEDLINE | ID: mdl-33045869

ABSTRACT

GM1 gangliosidosis is a rare neurodegenerative lysosomal storage disease caused by loss-of-function mutations in the gene encoding beta-galactosidase (ß-gal). There are no approved treatments for GM1 gangliosidosis. Previous studies in animal models have demonstrated that adeno-associated viral (AAV) vector-mediated gene transfer to the brain can restore ß-gal expression and prevent the onset of neurological signs. We developed an optimized AAV vector expressing human ß-gal and evaluated the efficacy of a single intracerebroventricular injection of this vector into the cerebrospinal fluid (CSF) of a murine disease model. The AAV vector administration into the CSF increased ß-gal activity in the brain, reduced neuronal lysosomal storage lesions, prevented the onset of neurological signs and gait abnormalities, and increased survival. These findings demonstrate the potential therapeutic activity of this vector and support its subsequent development for the treatment of GM1 gangliosidosis.


Subject(s)
Brain/metabolism , Cerebrospinal Fluid/metabolism , Dependovirus/genetics , Gangliosidosis, GM1/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , beta-Galactosidase/physiology , Animals , Brain/pathology , Cerebrospinal Fluid/cytology , Disease Models, Animal , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , Gangliosidosis, GM1/pathology , Genetic Vectors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , beta-Galactosidase/administration & dosage , beta-Galactosidase/genetics
6.
Stem Cell Reports ; 14(5): 909-923, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32302553

ABSTRACT

GM1 gangliosidosis is a lysosomal storage disease caused by loss of lysosomal ß-galactosidase activity and characterized by progressive neurodegeneration due to massive accumulation of GM1 ganglioside in the brain. Here, we generated induced pluripotent stem cells (iPSCs) derived from patients with GM1 gangliosidosis, and the resultant neurons showed impaired neurotransmitter release as a presynaptic function and accumulation of GM1 ganglioside. Treatment of normal neurons with GM1 ganglioside also disturbed presynaptic function. A high-content drug-screening system was then established and identified two compounds as drug candidates for GM1 gangliosidosis. Treatment of the patient-derived neurons with the candidate agents activated autophagy pathways, reducing GM1 ganglioside accumulation in vitro and in vivo, and restoring the presynaptic dysfunction. Our findings thus demonstrated the potential value of patient-derived iPSC lines as cellular models of GM1 gangliosidosis and revealed two potential therapeutic agents for future clinical application.


Subject(s)
Autophagy , G(M1) Ganglioside/metabolism , Gangliosidosis, GM1/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Cells, Cultured , Drug Development/methods , Gangliosidosis, GM1/pathology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurons/cytology , Neurons/drug effects , Synapses/drug effects , Synapses/metabolism
7.
Hum Genet ; 139(5): 657-673, 2020 May.
Article in English | MEDLINE | ID: mdl-32219518

ABSTRACT

GM1-gangliosidosis, a lysosomal storage disorder, is associated with ~ 161 missense variants in the GLB1 gene. Affected patients present with ß-galactosidase (ß-Gal) deficiency in lysosomes. Loss of function in ER-retained misfolded enzymes with missense variants is often due to subcellular mislocalization. Deoxygalactonojirimycin (DGJ) and its derivatives are pharmaceutical chaperones that directly bind to mutated ß-Gal in the ER promoting its folding and trafficking to lysosomes and thus enhancing its activity. An Emirati child has been diagnosed with infantile GM1-gangliosidosis carrying the reported p.D151Y variant. We show that p.D151Y ß-Gal in patient's fibroblasts retained < 1% residual activity due to impaired processing and trafficking. The amino acid substitution significantly affected the enzyme conformation; however, p.D151Y ß-Gal was amenable for partial rescue in the presence of glycerol or at reduced temperature where activity was enhanced with ~ 2.3 and 7 folds, respectively. The butyl (NB-DGJ) and nonyl (NN-DGJ) derivatives of DGJ chaperoning function were evaluated by measuring their IC50s and ability to stabilize the wild-type ß-Gal against thermal degradation. Although NN-DGJ showed higher affinity to ß-Gal, it did not show a significant enhancement in p.D151Y ß-Gal activity. However, NB-DGJ promoted p.D151Y ß-Gal maturation and enhanced its activity up to ~ 4.5% of control activity within 24 h which was significantly increased to ~ 10% within 6 days. NB-DGJ enhancement effect was sustained over 3 days after washing it out from culture media. We therefore conclude that NB-DGJ might be a promising therapeutic chemical chaperone in infantile GM1 amenable variants and therefore warrants further analysis for its clinical applications.


Subject(s)
1-Deoxynojirimycin/pharmacology , Fibroblasts/metabolism , Gangliosidosis, GM1/metabolism , Mutant Proteins/metabolism , Mutation , Protein Processing, Post-Translational/drug effects , beta-Galactosidase/metabolism , 1-Deoxynojirimycin/chemistry , Child, Preschool , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Gangliosidosis, GM1/drug therapy , Gangliosidosis, GM1/pathology , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Male , Molecular Chaperones/pharmacology , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Conformation , Protein Transport , beta-Galactosidase/chemistry , beta-Galactosidase/genetics
8.
J Biol Chem ; 295(39): 13532-13555, 2020 09 25.
Article in English | MEDLINE | ID: mdl-31481471

ABSTRACT

Autosomal recessive mutations in the galactosidase ß1 (GLB1) gene cause lysosomal ß-gal deficiency, resulting in accumulation of galactose-containing substrates and onset of the progressive and fatal neurodegenerative lysosomal storage disease, GM1 gangliosidosis. Here, an enzyme replacement therapy (ERT) approach in fibroblasts from GM1 gangliosidosis patients with recombinant human ß-gal (rhß-gal) produced in Chinese hamster ovary cells enabled direct and precise rhß-gal delivery to acidified lysosomes. A single, low dose (3 nm) of rhß-gal was sufficient for normalizing ß-gal activity and mediating substrate clearance for several weeks. We found that rhß-gal uptake by the fibroblasts is dose-dependent and saturable and can be competitively inhibited by mannose 6-phosphate, suggesting cation-independent, mannose 6-phosphate receptor-mediated endocytosis from the cell surface. A single intracerebroventricularly (ICV) administered dose of rhß-gal (100 µg) resulted in broad bilateral biodistribution of rhß-gal to critical regions of pathology in a mouse model of GM1 gangliosidosis. Weekly ICV dosing of rhß-gal for 8 weeks substantially reduced brain levels of ganglioside and oligosaccharide substrates and reversed well-established secondary neuropathology. Of note, unlike with the ERT approach, chronic lentivirus-mediated GLB1 overexpression in the GM1 gangliosidosis patient fibroblasts caused accumulation of a prelysosomal pool of ß-gal, resulting in activation of the unfolded protein response and endoplasmic reticulum stress. This outcome was unsurprising in light of our in vitro biophysical findings for rhß-gal, which include pH-dependent and concentration-dependent stability and dynamic self-association. Collectively, our results highlight that ICV-ERT is an effective therapeutic intervention for managing GM1 gangliosidosis potentially more safely than with gene therapy approaches.


Subject(s)
Enzyme Replacement Therapy , Gangliosidosis, GM1/therapy , beta-Galactosidase/metabolism , Animals , Gangliosidosis, GM1/metabolism , Gangliosidosis, GM1/pathology , Mice
9.
Bioorg Med Chem ; 26(20): 5462-5469, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30270003

ABSTRACT

(5aR)-5a-C-pentyl-4-epi-isofagomine 1 is a powerful inhibitor of lysosomal ß-galactosidase and a remarkable chaperone for mutations associated with GM1-gangliosidosis and Morquio disease type B. We report herein an improved synthesis of this compound and analogs (5a-C-methyl, pentyl, nonyl and phenylethyl derivatives), and a crystal structure of a synthetic intermediate that confirms its configuration resulting from the addition of a Grignard reagent. These compounds were evaluated as glycosidase inhibitors and their potential as chaperones for mutant lysosomal galactosidases determined. Based on these results and on docking studies, the 5-C-pentyl derivative 1 was selected as the optimal structure for further investigations: this compound induces the maturation of mutated ß-galactosidase in fibroblasts of a GM1-gangliosidosis patient and promote the decrease of keratan sulfate and oligosaccharide load in patient cells. Compound 1 is clearly capable of restoring ß-galactosidase activity and of promoting maturation of the protein, which should result in significant clinical benefit. These properties strongly support the development of compound 1 for the treatment of GM1-gangliosidosis and Morquio disease type B patients harboring ß-galactosidase mutations sensitive to pharmacological chaperoning.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gangliosidosis, GM1/drug therapy , Imino Pyranoses/chemistry , Imino Pyranoses/pharmacology , Mucopolysaccharidosis IV/drug therapy , beta-Galactosidase/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Gangliosidosis, GM1/enzymology , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , Humans , Imino Pyranoses/chemical synthesis , Imino Pyranoses/therapeutic use , Molecular Docking Simulation , Mucopolysaccharidosis IV/enzymology , Mucopolysaccharidosis IV/genetics , Mucopolysaccharidosis IV/metabolism , Mutation/drug effects , Structure-Activity Relationship , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
10.
J Lipid Res ; 59(12): 2446-2455, 2018 12.
Article in English | MEDLINE | ID: mdl-30266834

ABSTRACT

Mass spectrometry imaging (MSI) is a tool to rapidly map the spatial location of analytes without the need for tagging or a reporter system. Niemann-Pick disease type C1 (NPC1) is a neurodegenerative, lysosomal storage disorder characterized by accumulation of unesterified cholesterol and sphingolipids in the endo-lysosomal system. Here, we use MSI to visualize lipids including cholesterol in cerebellar brain tissue from the NPC1 symptomatic mouse model and unaffected controls. To complement the imaging studies, a data-processing pipeline was developed to generate consensus mass spectra, thereby using both technical and biological image replicates to assess differences. The consensus spectra are used to determine true differences in lipid relative abundance; lipid distributions can be determined in an unbiased fashion without prior knowledge of location. We show the cerebellar distribution of gangliosides GM1, GM2, and GM3, including variants of lipid chain length. We also performed MALDI-MSI of cholesterol. Further analysis of lobules IV/V and X of the cerebellum gangliosides indicates regional differences. The specificity achieved highlights the power of MSI, and this new workflow demonstrates a universal approach for addressing reproducibility in imaging experiments applied to NPC1.


Subject(s)
Mass Spectrometry/methods , Niemann-Pick Disease, Type C/metabolism , Animals , Cholesterol/metabolism , Gangliosides/metabolism , Gangliosidoses, GM2/metabolism , Gangliosidosis, GM1/metabolism , Lipid Metabolism/physiology , Mice , Mice, Inbred BALB C , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Sphingolipids/metabolism
11.
Immunol Res ; 66(1): 187-199, 2018 02.
Article in English | MEDLINE | ID: mdl-28940167

ABSTRACT

Aeromonas hydrophila is responsible for causing fatal infections in freshwater fishes. Besides chemical/antibiotic treatment and whole-cell vaccine, no subunit vaccine is currently available for A. hydrophila. Outer membrane proteins of gram-negative bacteria have been reported as effective vaccine candidates. Peptide antigens elicit focused immune responses against immunodominant stretches of the antigen. We have attempted to characterize the immunogenicity of linear B-cell epitopes of outer membrane protein (OmpC) of A. hydrophila identified using in silico tools, in conjugation with heat-labile enterotoxin B (LTB) subunit of Escherichia coli as a carrier protein. Antisera against the fusion protein harboring 323-336 residues of the AhOmpC (raised in mice) showed maximum cross-reactivity with the parent protein OmpC and LTB. The fusion protein displayed efficient GM1 ganglioside receptor binding, retaining the adjuvanicity of LTB. Antibody isotype profile and in vitro T-cell response analysis, cytokine ELISA, and array analysis collectively revealed a Th2-biased mixed T-helper cell response. Agglutination assay and flow cytometry analysis validated the ability of anti-fusion protein antisera to recognize the surface exposed epitopes on Aeromonas cells, demonstrating its neutralization potential. Oral immunization studies in Labeo rohita resulted in the generation of long-lasting humoral immune response, and the antisera could cross-react with the fusion protein as well as both the fusion partners. Considering significant similarity among OmpC of different enteric bacteria, the use of A. hydrophila OmpC epitope323-336 in fusion with LTB could have a broader scope in vaccine design.


Subject(s)
Aeromonas hydrophila/physiology , Bacterial Toxins/immunology , Cyprinidae/immunology , Enterotoxins/immunology , Epitopes, B-Lymphocyte/immunology , Escherichia coli Proteins/immunology , Fish Diseases/immunology , Gram-Negative Bacterial Infections/immunology , Porins/immunology , Recombinant Fusion Proteins/immunology , Th2 Cells/immunology , Vaccines, Subunit/immunology , Animals , Antibodies, Bacterial/metabolism , Bacterial Toxins/genetics , Bacterial Vaccines/immunology , Cells, Cultured , Computers, Molecular , Enterotoxins/genetics , Epitopes, B-Lymphocyte/genetics , Escherichia coli Proteins/genetics , Gangliosidosis, GM1/metabolism , Porins/genetics , Protein Binding , Recombinant Fusion Proteins/genetics
12.
Mol Ther ; 25(4): 892-903, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28236574

ABSTRACT

GM1 gangliosidosis is a fatal neurodegenerative disease that affects individuals of all ages. Favorable outcomes using adeno-associated viral (AAV) gene therapy in GM1 mice and cats have prompted consideration of human clinical trials, yet there remains a paucity of objective biomarkers to track disease status. We developed a panel of biomarkers using blood, urine, cerebrospinal fluid (CSF), electrodiagnostics, 7 T MRI, and magnetic resonance spectroscopy in GM1 cats-either untreated or AAV treated for more than 5 years-and compared them to markers in human GM1 patients where possible. Significant alterations were noted in CSF and blood of GM1 humans and cats, with partial or full normalization after gene therapy in cats. Gene therapy improved the rhythmic slowing of electroencephalograms (EEGs) in GM1 cats, a phenomenon present also in GM1 patients, but nonetheless the epileptiform activity persisted. After gene therapy, MR-based analyses revealed remarkable preservation of brain architecture and correction of brain metabolites associated with microgliosis, neuroaxonal loss, and demyelination. Therapeutic benefit of AAV gene therapy in GM1 cats, many of which maintain near-normal function >5 years post-treatment, supports the strong consideration of human clinical trials, for which the biomarkers described herein will be essential for outcome assessment.


Subject(s)
Biomarkers , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , Genetic Therapy , Animals , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Biomarkers/urine , Cats , Dependovirus/classification , Dependovirus/genetics , Disease Models, Animal , Electroencephalography , Gangliosidosis, GM1/mortality , Gangliosidosis, GM1/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Hypocalcemia/metabolism , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Treatment Outcome
13.
J Peripher Nerv Syst ; 22(1): 4-12, 2017 03.
Article in English | MEDLINE | ID: mdl-27801990

ABSTRACT

The outcome of Guillain-Barré syndrome (GBS) remains unchanged since plasma exchange and intravenous immunoglobulin (IVIg) were introduced over 20 years ago. Pathogenesis studies on GBS have identified the terminal component of complement cascade as a key disease mediator and therapeutic target. We report the first use of terminal complement pathway inhibition with eculizumab in humans with GBS. In a randomised, double-blind, placebo-controlled trial, 28 subjects eligible on the basis of GBS disability grade of at least 3 were screened, of whom 8 (29%) were randomised. Five received eculizumab for 4 weeks, alongside standard IVIg treatment. The safety outcomes, monitored via adverse events capture, showed eculizumab to be well-tolerated and safe when administered in conjunction with IVIg. Primary and secondary efficacy outcomes in the form of GBS disability scores (GBS DS), MRC sum scores, Rasch overall disability scores, and overall neuropathy limitation scores are reported descriptively. For the primary efficacy outcome at 4 weeks after recruitment, two of two placebo- and two of five eculizumab-treated subjects had improved by one or more grades on the GBS DS. Although the small sample size precludes a statistically meaningful analysis, these pilot data indicate further studies on complement inhibition in GBS are warranted.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Guillain-Barre Syndrome/drug therapy , Immunologic Factors/therapeutic use , Adult , Aged , Disability Evaluation , Dose-Response Relationship, Drug , Double-Blind Method , Female , Gangliosidoses, GM2/metabolism , Gangliosidosis, GM1/metabolism , Humans , Longitudinal Studies , Male , Middle Aged , Treatment Outcome
14.
Chem Commun (Camb) ; 52(32): 5497-515, 2016 Apr 25.
Article in English | MEDLINE | ID: mdl-27043200

ABSTRACT

Lysosomal storage disorders (LSDs) are often caused by mutations that destabilize native folding and impair the trafficking of enzymes, leading to premature endoplasmic reticulum (ER)-associated degradation, deficiencies of specific hydrolytic functions and aberrant storage of metabolites in the lysosomes. Enzyme replacement therapy (ERT) and substrate reduction therapy (SRT) are available for a few of these conditions, but most remain orphan. A main difficulty is that virtually all LSDs involve neurological decline and neither proteins nor the current SRT drugs can cross the blood-brain barrier. Twenty years ago a new therapeutic paradigm better suited for neuropathic LSDs was launched, namely pharmacological chaperone (PC) therapy. PCs are small molecules capable of binding to the mutant protein at the ER, inducing proper folding, restoring trafficking and increasing enzyme activity and substrate processing in the lysosome. In many LSDs the mutated protein is a glycosidase and the accumulated substrate is an oligo- or polysaccharide or a glycoconjugate, e.g. a glycosphingolipid. Although it might appear counterintuitive, substrate analogues (glycomimetics) behaving as competitive glycosidase inhibitors are good candidates to perform PC tasks. The advancements in the knowledge of the molecular basis of LSDs, including enzyme structures, binding modes, trafficking pathways and substrate processing mechanisms, have been put forward to optimize PC selectivity and efficacy. Moreover, the chemical versatility of glycomimetics and the variety of structures at hand allow simultaneous optimization of chaperone and pharmacokinetic properties. In this Feature Article we review the advancements made in this field in the last few years and the future outlook through the lessons taught by three archetypical LSDs: Gaucher disease, GM1-gangliosidosis and Fabry disease.


Subject(s)
Carbohydrates/chemistry , Fabry Disease/drug therapy , Gangliosidosis, GM1/drug therapy , Gaucher Disease/drug therapy , Molecular Chaperones/therapeutic use , Molecular Mimicry , Fabry Disease/metabolism , Gangliosidosis, GM1/metabolism , Gaucher Disease/metabolism , Humans
15.
Sci Rep ; 6: 20997, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26880066

ABSTRACT

The role of first-stage ß-amyloid aggregation in the development of the Alzheimer disease, is widely accepted but still unclear. Intimate interaction with the cell membrane is invoked. We designed Neutron Reflectometry experiments to reveal the existence and extent of the interaction between ß-amyloid (Aß) peptides and a lone customized biomimetic membrane, and their dependence on the aggregation state of the peptide. The membrane, asymmetrically containing phospholipids, GM1 and cholesterol in biosimilar proportion, is a model for a raft, a putative site for amyloid-cell membrane interaction. We found that the structured-oligomer of Aß(1-42), its most acknowledged membrane-active state, is embedded as such into the external leaflet of the membrane. Conversely, the Aß(1-42) unstructured early-oligomers deeply penetrate the membrane, likely mimicking the interaction at neuronal cell surfaces, when the Aß(1-42) is cleaved from APP protein and the membrane constitutes a template for its further structural evolution. Moreover, the smaller Aß(1-6) fragment, the N-terminal portion of Aß, was also used. Aß N-terminal is usually considered as involved in oligomer stabilization but not in the peptide-membrane interaction. Instead, it was seen to remove lipids from the bilayer, thus suggesting its role, once in the whole peptide, in membrane leakage, favouring peptide recruitment.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Biological Mimicry , Lipid Bilayers , Alzheimer Disease , Biomimetics , Cell Membrane , Gangliosidosis, GM1/metabolism , Humans , Neutrons , Protein Binding , Protein Multimerization
16.
Am J Med Genet A ; 170(3): 634-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26646981

ABSTRACT

Background GM1 gangliosidosis is a lysosomal storage disorder caused by mutations in GLB1, encoding ß-galactosidase. The range of severity is from type I infantile disease, lethal in early childhood, to type III adult onset, resulting in gradually progressive neurological symptoms in adulthood. The intermediate group of patients has been recently classified as having type II late infantile subtype with onset of symptoms at one to three years of age or type II juvenile subtype with symptom onset at 2-10 years. To characterize disease severity and progression, six Late infantile and nine juvenile patients were evaluated using magnetic resonance imaging (MRI), and MR spectroscopy (MRS). Since difficulties with ambulation (gross motor function) and speech (expressive language) are often the first reported symptoms in type II GM1, patients were also scored in these domains. Deterioration of expressive language and ambulation was more rapid in the late infantile patients. Fourteen MRI scans in six Late infantile patients identified progressive atrophy in the cerebrum and cerebellum. Twenty-six MRI scans in nine juvenile patients revealed greater variability in extent and progression of atrophy. Quantitative MRS demonstrated a deficit of N-acetylaspartate in both the late infantile and juvenile patients with greater in the late infantile patients. This correlates with clinical measures of ambulation and expressive language. The two subtypes of type II GM1 gangliosidosis have different clinical trajectories. MRI scoring, quantitative MRS and brain volume correlate with clinical disease progression and may serve as important minimally-invasive outcome measures for clinical trials.


Subject(s)
Atrophy/diagnosis , Gangliosidosis, GM1/diagnosis , Speech Disorders/diagnosis , beta-Galactosidase/genetics , Adolescent , Age of Onset , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Atrophy/genetics , Atrophy/metabolism , Atrophy/pathology , Cerebellum/metabolism , Cerebellum/pathology , Cerebrum/metabolism , Cerebrum/pathology , Child , Child, Preschool , Disease Progression , Female , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , Gangliosidosis, GM1/pathology , Gene Expression , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mobility Limitation , Severity of Illness Index , Speech , Speech Disorders/genetics , Speech Disorders/metabolism , Speech Disorders/pathology , Young Adult , beta-Galactosidase/deficiency
17.
Brain Dev ; 38(2): 175-80, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26259553

ABSTRACT

INTRODUCTION: Galactosialidosis is a rare lysosomal storage disease caused by a combined deficiency of GM1 ß-galactosidase (ß-gal) and neuraminidase secondary to a defect of a lysosomal enzyme protective protein/cathepsin A (PPCA) and mutation in CTSA gene. Three subtypes are recognized: early infantile, late infantile, and juvenile/adult. There is no specific therapy for patients with galactosialidosis at this time. OBJECTIVES: The aim of this study was to determine the chaperone effect of N-octyl-4-epi-ß-valienamine (NOEV) on ß-gal proteins in skin fibroblasts of PPCA-deficit patients. METHODS: ß-Gal and neuraminidase activities were measured for the diagnosis of the patients with galactosialidosis. Western blotting for PPCA protein and direct sequencing for CTSA gene were performed. Cultured skin fibroblast were treated with NOEV. RESULTS: We report four novel patients with galactosialidosis: one had the early infantile form and the other three had the juvenile/adult form. We found that NOEV stabilized ß-gal activity in lysate from cultured skin fibroblasts from these patients. Treatment with NOEV significantly enhanced ß-gal activity in cultured skin fibroblasts in the absence of PPCA. CONCLUSIONS: Our results indicate the possibility that NOEV chaperone therapy might have a beneficial effect, at least in part, for patients with galactosialidosis.


Subject(s)
Gangliosidosis, GM1/drug therapy , Hexosamines/pharmacology , Adolescent , Adult , Cathepsin A/metabolism , Cells, Cultured , Child, Preschool , Fibroblasts/drug effects , Gangliosidosis, GM1/enzymology , Gangliosidosis, GM1/metabolism , Gangliosidosis, GM1/pathology , Humans , Infant, Newborn , Molecular Chaperones/pharmacology , Mutation , beta-Galactosidase/metabolism
19.
Hum Mol Genet ; 24(15): 4353-64, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25964428

ABSTRACT

GM1 gangliosidosis (GM1) is an autosomal recessive lysosomal storage disease where GLB1 gene mutations result in a reduction or absence of lysosomal acid ß-galactosidase (ßgal) activity. ßgal deficiency leads to accumulation of GM1-ganglioside in the central nervous system (CNS). GM1 is characterized by progressive neurological decline resulting in generalized paralysis, extreme emaciation and death. In this study, we assessed the therapeutic efficacy of an adeno-associated virus (AAV) 9-mßgal vector infused systemically in adult GM1 mice (ßGal(-/-)) at 1 × 10(11) or 3 × 10(11) vector genomes (vg). Biochemical analysis of AAV9-treated GM1 mice showed high ßGal activity in liver and serum. Moderate ßGal levels throughout CNS resulted in a 36-76% reduction in GM1-ganglioside content in the brain and 75-86% in the spinal cord. Histological analyses of the CNS of animals treated with 3 × 10(11) vg dose revealed increased presence of ßgal and clearance of lysosomal storage throughout cortex, hippocampus, brainstem and spinal cord. Storage reduction in these regions was accompanied by a marked decrease in astrogliosis. AAV9 treatment resulted in improved performance in multiple tests of motor function and behavior. Also the majority of GM1 mice in the 3 × 10(11) vg cohort retained ambulation and rearing despite reaching the humane endpoint due to weight loss. Importantly, the median survival of AAV9 treatment groups (316-576 days) was significantly increased over controls (250-264 days). This study shows that moderate widespread expression of ßgal in the CNS of GM1 gangliosidosis mice is sufficient to achieve significant biochemical impact with phenotypic amelioration and extension in lifespan.


Subject(s)
Central Nervous System/metabolism , Gangliosidosis, GM1/genetics , Genetic Therapy , beta-Galactosidase/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain Stem/metabolism , Brain Stem/pathology , Central Nervous System/pathology , Dependovirus/genetics , Disease Models, Animal , Gangliosides/metabolism , Gangliosidosis, GM1/metabolism , Gangliosidosis, GM1/therapy , Genetic Vectors , Humans , Mice , Spinal Cord/metabolism , Spinal Cord/pathology , beta-Galactosidase/biosynthesis , beta-Galactosidase/blood
20.
J Pathol ; 237(1): 98-110, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25925601

ABSTRACT

GM1 gangliosidosis (GM1) is an inherited neurodegenerative disorder caused by mutations in the lysosomal ß-galactosidase (ß-gal) gene. Insufficient ß-gal activity leads to abnormal accumulation of GM1 gangliosides in tissues, particularly in the central nervous system, resulting in progressive neurodegeneration. Here, we report an in vitro human GM1 model, based on induced pluripotent stem cell (iPSC) technology. Neural progenitor cells differentiated from GM1 patient-derived iPSCs (GM1-NPCs) recapitulated the biochemical and molecular phenotypes of GM1, including defective ß-gal activity and increased lysosomes. Importantly, the characterization of GM1-NPCs established that GM1 is significantly associated with the activation of inflammasomes, which play a critical role in the pathogenesis of various neurodegenerative diseases. Specific inflammasome inhibitors potently alleviated the disease-related phenotypes of GM1-NPCs in vitro and in vivo. Our data demonstrate that GM1-NPCs are a valuable in vitro human GM1 model and suggest that inflammasome activation is a novel target pathway for GM1 drug development.


Subject(s)
Gangliosidosis, GM1/metabolism , Induced Pluripotent Stem Cells/metabolism , Inflammasomes/metabolism , Neural Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Line , Cell Shape , Cellular Reprogramming , Gangliosidosis, GM1/immunology , Gangliosidosis, GM1/pathology , Genotype , Humans , Immunologic Factors/pharmacology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/transplantation , Inflammasomes/antagonists & inhibitors , Inflammasomes/immunology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Lysosomes/metabolism , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neural Stem Cells/immunology , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Phenotype , Signal Transduction , Time Factors , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...