Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
2.
Mol Ther ; 27(8): 1495-1506, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31208914

ABSTRACT

Neuronopathic glycosphingolipidoses are a sub-group of lysosomal storage disorders for which there are presently no effective therapies. Here, we evaluated the potential of substrate reduction therapy (SRT) using an inhibitor of glucosylceramide synthase (GCS) to decrease the synthesis of glucosylceramide (GL1) and related glycosphingolipids. The substrates that accumulate in Sandhoff disease (e.g., ganglioside GM2 and its nonacylated derivative, lyso-GM2) are distal to the drug target, GCS. Treatment of Sandhoff mice with a GCS inhibitor that has demonstrated CNS access (Genz-682452) reduced the accumulation of GL1 and GM2, as well as a variety of disease-associated substrates in the liver and brain. Concomitant with these effects was a significant decrease in the expression of CD68 and glycoprotein non-metastatic melanoma B protein (Gpnmb) in the brain, indicating a reduction in microgliosis in the treated mice. Moreover, using in vivo imaging, we showed that the monocytic biomarker translocator protein (TSPO), which was elevated in Sandhoff mice, was normalized following Genz-682452 treatment. These positive effects translated in turn into a delay (∼28 days) in loss of motor function and coordination, as measured by rotarod latency, and a significant increase in longevity (∼17.5%). Together, these results support the development of SRT for the treatment of gangliosidoses, particularly in patients with residual enzyme activity.


Subject(s)
Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Quinuclidines/pharmacology , Sandhoff Disease/enzymology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Ligands , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mass Spectrometry , Mice , Mice, Knockout , Molecular Imaging , Receptors, GABA/metabolism , Sandhoff Disease/diagnosis , Sandhoff Disease/genetics , Sandhoff Disease/therapy , Sphingolipids/metabolism , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/metabolism
3.
Curr Gene Ther ; 18(2): 68-89, 2018.
Article in English | MEDLINE | ID: mdl-29618308

ABSTRACT

Tay-Sachs disease, caused by impaired ß-N-acetylhexosaminidase activity, was the first GM2 gangliosidosis to be studied and one of the most severe and earliest lysosomal diseases to be described. The condition, associated with the pathological build-up of GM2 ganglioside, has acquired almost iconic status and serves as a paradigm in the study of lysosomal storage diseases. Inherited as a classical autosomal recessive disorder, this global disease of the nervous system induces developmental arrest with regression of attained milestones; neurodegeneration progresses rapidly to cause premature death in young children. There is no effective treatment beyond palliative care, and while the genetic basis of GM2 gangliosidosis is well established, the molecular and cellular events, from diseasecausing mutations and glycosphingolipid storage to disease manifestations, remain to be fully delineated. Several therapeutic approaches have been attempted in patients, including enzymatic augmentation, bone marrow transplantation, enzyme enhancement, and substrate reduction therapy. Hitherto, none of these stratagems has materially altered the course of the disease. Authentic animal models of GM2 gangliodidosis have facilitated in-depth evaluation of innovative applications such as gene transfer, which in contrast to other interventions, shows great promise. This review outlines current knowledge pertaining the pathobiology as well as potential innovative treatments for the GM2 gangliosidoses.


Subject(s)
Bone Marrow Transplantation , Enzyme Replacement Therapy , Genetic Therapy , Sandhoff Disease/genetics , Sandhoff Disease/therapy , Tay-Sachs Disease/genetics , Tay-Sachs Disease/therapy , Animals , Disease Models, Animal , Glycosphingolipids/metabolism , Humans , Infant , Lysosomes/enzymology , Lysosomes/genetics , Lysosomes/pathology , Mice , Mutation , Rare Diseases , Sandhoff Disease/enzymology , Sandhoff Disease/pathology , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/pathology , beta-N-Acetylhexosaminidases/genetics
4.
Hum Gene Ther ; 27(7): 497-508, 2016 07.
Article in English | MEDLINE | ID: mdl-27199088

ABSTRACT

GM2 gangliosidosis is a group of neurodegenerative diseases caused by ß-hexosaminidase A (HexA) enzyme deficiency. There is currently no cure. HexA is composed of two similar, nonidentical subunits, α and ß, which must interact with the GM2 activator protein (GM2AP), a substrate-specific cofactor, to hydrolyze GM2 ganglioside. Mutations in either subunit or the activator can result in the accumulation of GM2 ganglioside within neurons throughout the central nervous system. The resulting neuronal cell death induces the primary symptoms of the disease: motor impairment, seizures, and sensory impairments. This study assesses the long-term effects of gene transfer in a Sandhoff (ß-subunit knockout) mouse model. The study utilized a modified human ß-hexosaminidase α-subunit (µ-subunit) that contains critical sequences from the ß-subunit that enables formation of a stable homodimer (HexM) and interaction with GM2AP to hydrolyze GM2 ganglioside. We investigated a self-complementary adeno-associated viral (scAAV) vector expressing HexM, through intravenous injections of the neonatal mice. We monitored one cohort for 8 weeks and another cohort long-term for survival benefit, behavioral, biochemical, and molecular analyses. Untreated Sandhoff disease (SD) control mice reached a humane endpoint at approximately 15 weeks, whereas treated mice had a median survival age of 40 weeks, an approximate 2.5-fold survival advantage. On behavioral tests, the treated mice outperformed their knockout age-matched controls and perform similarly to the heterozygous controls. Through the enzymatic and GM2 ganglioside analyses, we observed a significant decrease in the GM2 ganglioside level, even though the enzyme levels were not significantly increased. Molecular analyses revealed a global distribution of the vector between brain and spinal cord regions. In conclusion, the neonatal delivery of a novel viral vector expressing the human HexM enzyme is effective in ameliorating the SD mouse phenotype for long-term. Our data could have implications not only for treatment of SD but also for Tay-Sachs disease (α-subunit deficiency) and similar brain disorders.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Hexosaminidases/genetics , Mutation/genetics , Sandhoff Disease/therapy , Animals , Animals, Newborn , Behavior, Animal , Disease Models, Animal , Mice , Phenotype , Sandhoff Disease/enzymology , Sandhoff Disease/genetics
5.
J Hum Genet ; 61(2): 163-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26582265

ABSTRACT

Sandhoff disease (SD) is an autosomal recessive neurodegenerative lysosomal storage disorder caused by mutations in HEXB gene. Molecular pathology is unknown in Indian patients with SD. The present study is aimed to determine mutations spectrum and molecular pathology leading to SD in 22 unrelated patients confirmed by the deficiency of ß-hexosaminidase-A and total-hexosaminidase in leukocytes. To date, nearly 86 mutations of HEXB have been described, including five large deletions. Over all we have identified 13 mutations in 19 patients, eight of which were novel, including two missense mutations [c.611G>A (p.G204E), c. 634A>T (p.H212Y)], two nonsense mutations [c.333G>A (p.W111X), c.298C>T (p.R100X)], one splice site mutation c.1082+5 G>T, two small in-frame deletions [c.534_541delAGTTTATC (p.V179RfsX10), c.1563_1573delTATGGATGACG (p.M522LfsX2)] and one insertion c.1553_1554insAAGA (p.D518EfsX8). We have also identified previously known, five sequence variations leading to amino acid changes [c.926G>A (p.C309Y), c.1597C>T (p.R533C)], one nonsense mutation c.850 C>T (p.R284X), one splice site mutation c.1417+1 G-A and one insertion c.1591_1592insC (p.R531TfsX22). Mutation was not identified in three patients. We observed from this study that mutation c.850C>T (p.R284X) was identified in 4/19 (21%) patients which is likely to be the most common mutation in the country. This is the first study providing insight into the molecular basis of SD in India.


Subject(s)
Mutation , Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , Child, Preschool , Humans , Infant , Sandhoff Disease/diagnosis , Sandhoff Disease/enzymology , beta-Hexosaminidase beta Chain/metabolism
6.
Zhonghua Er Ke Za Zhi ; 52(4): 313-6, 2014 Apr.
Article in Chinese | MEDLINE | ID: mdl-24915922

ABSTRACT

OBJECTIVE: To explore the clinical features and molecular mutation of HEXB gene in a case with juvenile Sandhoff disease. METHOD: We retrospectively reviewed the clinical, neuroimaging and biochemical findings in this Chinese child with juvenile Sandhoff disease. Hexosaminidase A and hexosaminidase A & B activities were measured in blood leukocytes by fluorometric assay. HEXB gene molecular analysis was performed by PCR and direct sequencing. RESULT: The 9-year-old boy was admitted for psychomotor regression. He presented slowly progressive gait disorder and dysarthria during the last three years. Cranial MRI revealed a marked cerebellar atrophy with normal intensity in the thalamus and basal ganglia. Brain MRS showed normal in the thalamus and basal ganglia. Hexosaminidase A was 69.5 (mg·h) [normal controls 150-360 nmol/(mg·h)], hexosaminidase A & B activity was 119 nmol/(mg·h)[normal controls 600-3 500 nmol/(mg·h)], confirming the diagnosis of Sandhoff disease. The patient was a compound heterozygote for a novel deletion mutation c.1404delT (p. P468P fsX62) and a reported mutation c.1509-26G>A. CONCLUSION: The clinical features of juvenile Sandhoff disease include ataxia, dysarthria and cerebellar atrophy. The enzyme assay and molecular analysis of HEXB gene can confirm the diagnosis of Sandhoff disease. The novel mutation c.1404delT(p. P468P fsX62) is a disease-related mutation.


Subject(s)
Mutation , Sandhoff Disease/diagnosis , Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , Brain/diagnostic imaging , Brain/pathology , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/enzymology , Cerebellar Ataxia/genetics , Child , DNA Mutational Analysis , Heterozygote , Hexosaminidase A/blood , Hexosaminidase A/metabolism , Hexosaminidase B/blood , Hexosaminidase B/metabolism , Humans , Leukocytes/enzymology , Magnetic Resonance Imaging , Male , Radiography , Retrospective Studies , Sandhoff Disease/enzymology
7.
Mol Neurobiol ; 50(1): 159-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24356898

ABSTRACT

Sphingolipidoses are inherited genetic diseases due to mutations in genes encoding proteins involved in the lysosomal catabolism of sphingolipids. Despite a low incidence of each individual disease, altogether, the number of patients involved is relatively high and resolutive approaches for treatment are still lacking. The chaperone therapy is one of the latest pharmacological approaches to these storage diseases. This therapy allows the mutated protein to escape its natural removal and to increase its quantity in lysosomes, thus partially restoring the metabolic functions. Sandhoff disease is an autosomal recessive inherited disorder resulting from ß-hexosaminidase deficiency and characterized by large accumulation of GM2 ganglioside in brain. No enzymatic replacement therapy is currently available, and the use of inhibitors of glycosphingolipid biosynthesis for substrate reduction therapy, although very promising, is associated with serious side effects. The chaperone pyrimethamine has been proposed as a very promising drug in those cases characterized by a residual enzyme activity. In this review, we report the effect of pyrimethamine on the recovery of ß-hexosaminidase activity in cultured fibroblasts from Sandhoff patients.


Subject(s)
Fibroblasts/drug effects , Hexosaminidase B/metabolism , Molecular Chaperones/pharmacology , Pyrimethamine/pharmacology , Sandhoff Disease/drug therapy , Fibroblasts/enzymology , Humans , Molecular Chaperones/therapeutic use , Pyrimethamine/therapeutic use , Sandhoff Disease/enzymology
8.
J Hum Genet ; 58(9): 611-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23759947

ABSTRACT

Sandhoff disease is a GM2 gangliosidosis caused by mutations in HEXB encoding the ß-subunit of ß-hexosaminidase A. ß-Hexosaminidase A exists as a heterodimer consisting of α- and ß-subunits, and requires a GM2 activator protein to hydrolyze GM2. To investigate the molecular pathology in an adult Sandhoff disease patient with an early disease onset, we performed mutation detection, western blot analysis and molecular simulation analysis. The patient had compound heterozygous mutations p.Arg505Gln and p.Ser341ValfsX30. Western blot analysis showed that the amount of mature form of the α- and ß-subunits was markedly decreased in the patient. We then performed docking simulation analysis of the α- and ß-subunits with p.Arg505Gln, the GM2AP/GM2 complex and ß-hexosaminidase A, and GM2 and ß-hexosaminidase A. Simulation analysis showed that p.Arg505Gln impaired each step of molecular conformation of the α- and ß-subunits heterodimer, the activator protein and GM2. The results indicated that p.Ser341ValfsX30 reduced the amount of ß-subunit, and that p.Arg505Gln hampered the maturation of α- and ß-subunits, and hindered the catalytic ability of ß-hexosaminidase A. In conclusion, various methods including simulation analysis were useful to understand the molecular pathology in Sandhoff disease.


Subject(s)
Hexosaminidase A/genetics , Molecular Docking Simulation , Sandhoff Disease/genetics , Adult , Female , G(M2) Activator Protein/chemistry , Hexosaminidase A/chemistry , Hexosaminidase A/metabolism , Humans , Mutation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Sandhoff Disease/enzymology
9.
Mol Ther ; 21(7): 1306-15, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23689599

ABSTRACT

Salutary responses to adeno-associated viral (AAV) gene therapy have been reported in the mouse model of Sandhoff disease (SD), a neurodegenerative lysosomal storage disease caused by deficiency of ß-N-acetylhexosaminidase (Hex). While untreated mice reach the humane endpoint by 4.1 months of age, mice treated by a single intracranial injection of vectors expressing human hexosaminidase may live a normal life span of 2 years. When treated with the same therapeutic vectors used in mice, two cats with SD lived to 7.0 and 8.2 months of age, compared with an untreated life span of 4.5 ± 0.5 months (n = 11). Because a pronounced humoral immune response to both the AAV1 vectors and human hexosaminidase was documented, feline cDNAs for the hexosaminidase α- and ß-subunits were cloned into AAVrh8 vectors. Cats treated with vectors expressing feline hexosaminidase produced enzymatic activity >75-fold normal at the brain injection site with little evidence of an immune infiltrate. Affected cats treated with feline-specific vectors by bilateral injection of the thalamus lived to 10.4 ± 3.7 months of age (n = 3), or 2.3 times as long as untreated cats. These studies support the therapeutic potential of AAV vectors for SD and underscore the importance of species-specific cDNAs for translational research.


Subject(s)
Cat Diseases/enzymology , Cat Diseases/therapy , Sandhoff Disease/enzymology , Sandhoff Disease/therapy , beta-N-Acetylhexosaminidases/metabolism , Animals , Cat Diseases/genetics , Cats , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors/genetics , Sandhoff Disease/genetics , beta-N-Acetylhexosaminidases/genetics
10.
J Neuroinflammation ; 9: 186, 2012 Aug 04.
Article in English | MEDLINE | ID: mdl-22863301

ABSTRACT

This study evaluated whether GM(2) ganglioside storage is necessary for neurodegeneration and neuroinflammation by performing ß-hexosaminidase rescue experiments in neurons of HexB(-/-) mice. We developed a novel mouse model, whereby the expression of the human HEXB gene was targeted to neurons of HexB(-/-) mice by the Thy1 promoter. Despite ß-hexosaminidase restoration in neurons was sufficient in rescuing HexB(-/-) mice from GM(2) neuronal storage and neurodegeneration, brain inflammation persisted, including the presence of large numbers of reactive microglia/macrophages due to persisting GM(2) presence in this cell type. In conclusion, our results suggest that neuroinflammation is not sufficient to elicit neurodegeneration as long as neuronal function is restored.


Subject(s)
Brain/enzymology , Gene Expression Regulation, Enzymologic , Neurodegenerative Diseases/enzymology , Neurons/enzymology , Sandhoff Disease/enzymology , beta-N-Acetylhexosaminidases/genetics , Animals , Brain/pathology , Humans , Inflammation/enzymology , Inflammation/pathology , Mice , Mice, 129 Strain , Mice, Transgenic , Neurodegenerative Diseases/pathology , Neurons/pathology , Sandhoff Disease/genetics , Sandhoff Disease/pathology , beta-N-Acetylhexosaminidases/biosynthesis
11.
Gene ; 506(1): 25-30, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22789865

ABSTRACT

The GM2 gangliosidoses are autosomal recessive lysosomal storage diseases caused by a deficiency of the ß-hexosaminidase A enzyme. This enzyme is composed of two polypeptide chains designated the α- and ß- subunits and it interacts with the GM2 activator protein. The HEXA and HEXB genes encode the α-subunit and the ß-subunit, respectively. Mutations in these genes are causative of Tay-Sachs disease (HEXA) and Sandhoff disease (HEXB). We analyzed the complete HEXA gene in 34 Spanish patients with Tay-Sachs disease and the HEXB gene in 14 Spanish patients with Sandhoff disease. We identified 27 different mutations, 14 of which were novel, in the HEXA gene and 14 different mutations, 8 of which unreported until now, in the HEXB gene, and we attempted to correlate these mutations with the clinical presentation of the patients. We found a high frequency of c.459+5G>A (IVS4+5G>A) mutation in HEXA affected patients, 22 of 68 alleles, which represent the 32.4%. This is the highest percentage found of this mutation in a population. All patients homozygous for mutation c.459+5G>A presented with the infantile form of the disease and, as previously reported, patients carrying mutation p.R178H in at least one of the alleles presented with a milder form. In HEXB affected patients, the novel deletion c.171delG accounts for 21.4% of the mutant alleles (6/28). All patients with this deletion showed the infantile form of the disease. The Spanish GM2 gangliosidoses affected patients show a great mutational heterogeneity as seen in other inherited lisosomal diseases in this country.


Subject(s)
Hexosaminidase A/genetics , Hexosaminidase B/genetics , Mutation , Sandhoff Disease/enzymology , Sandhoff Disease/genetics , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/genetics , Adolescent , Adult , Age of Onset , Child , Child, Preschool , DNA Mutational Analysis , Female , Fibroblasts/enzymology , Gene Frequency , Genetic Association Studies , Heterozygote , Hexosaminidase A/blood , Hexosaminidase A/metabolism , Hexosaminidase B/blood , Hexosaminidase B/metabolism , Homozygote , Humans , Infant , Leukocytes/enzymology , Male , Mutagenesis, Insertional , Mutation, Missense , Sequence Deletion , Spain , Young Adult
12.
PLoS One ; 6(12): e29074, 2011.
Article in English | MEDLINE | ID: mdl-22205997

ABSTRACT

To find a new biomarker of Tay-Sachs disease and Sandhoff disease. The lyso-GM2 ganglioside (lyso-GM2) levels in the brain and plasma in Sandhoff mice were measured by means of high performance liquid chromatography and the effect of a modified hexosaminidase (Hex) B exhibiting Hex A-like activity was examined. Then, the lyso-GM2 concentrations in human plasma samples were determined. The lyso-GM2 levels in the brain and plasma in Sandhoff mice were apparently increased compared with those in wild-type mice, and they decreased on intracerebroventricular administration of the modified Hex B. The lyso-GM2 levels in plasma of patients with Tay-Sachs disease and Sandhoff disease were increased, and the increase in lyso-GM2 was associated with a decrease in Hex A activity. Lyso-GM2 is expected to be a potential biomarker of Tay-Sachs disease and Sandhoff disease.


Subject(s)
G(M2) Ganglioside/analogs & derivatives , Sandhoff Disease/metabolism , Tay-Sachs Disease/metabolism , Adult , Animals , Biomarkers/blood , Biomarkers/metabolism , Brain/metabolism , G(M2) Activator Protein/deficiency , G(M2) Ganglioside/blood , G(M2) Ganglioside/metabolism , Hexosaminidases/blood , Humans , Infant , Mice , Sandhoff Disease/blood , Sandhoff Disease/enzymology , Tay-Sachs Disease/blood , Tay-Sachs Disease/enzymology
13.
Hum Mol Genet ; 20(22): 4371-80, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21852247

ABSTRACT

Sandhoff disease, a GM2 gangliosidosis caused by a deficiency in ß-hexosaminidase, is characterized by progressive neurodegeneration. Although loss of neurons in association with lysosomal storage of glycosphingolipids occurs in patients with this disease, the molecular pathways that lead to the accompanying neurological defects are unclear. Using an authentic murine model of GM2 gangliosidosis, we examined the pattern of neuronal loss in the central nervous system and investigated the effects of gene transfer using recombinant adeno-associated viral vectors expressing ß-hexosaminidase subunits (rAAV2/1-Hex). In 4-month-old Sandhoff mice with neurological deficits, cells staining positively for the apoptotic signature in the TUNEL reaction were found in the ventroposterior medial and ventroposterior lateral (VPM/VPL) nuclei of the thalamus. There was progressive loss of neuronal density in this region with age. Comparable loss of neuronal density was identified in the lateral vestibular nucleus of the brainstem and a small but statistically significant loss was present in the ventral spinal cord. Loss of neurons was not detected in other regions that were analysed. Administration of rAAV2/1-Hex into the brain of Sandhoff mice prevented the decline in neuronal density in the VPM/VPL. Preservation of neurons in the VPM/VPL was variable at the humane endpoint in treated animals, but correlated directly with increased lifespan. Loss of neurons was localized to only a few regions in the Sandhoff brain and was prevented by rAAV-mediated transfer of ß-hexosaminidase gene function at considerable distances from the site of vector administration.


Subject(s)
Brain/metabolism , Brain/pathology , Dependovirus/genetics , Genetic Vectors/genetics , Neurons/pathology , Sandhoff Disease/therapy , beta-N-Acetylhexosaminidases/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , In Situ Nick-End Labeling , Mice , Neurons/metabolism , Sandhoff Disease/enzymology , Sandhoff Disease/metabolism , beta-N-Acetylhexosaminidases/genetics
14.
Ann Neurol ; 69(4): 691-701, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21520232

ABSTRACT

OBJECTIVE: Novel recombinant human lysosomal ß-hexosaminidase A (HexA) was developed for enzyme replacement therapy (ERT) for Tay-Sachs and Sandhoff diseases, ie, autosomal recessive GM2 gangliosidoses, caused by HexA deficiency. METHODS: A recombinant human HexA (Om4HexA) with a high mannose 6-phosphate (M6P)-type-N-glycan content, which was produced by a methylotrophic yeast strain, Ogataea minuta, overexpressing the OmMNN4 gene, was intracerebroventricularly (ICV) administered to Sandhoff disease model mice (Hexb⁻/⁻ mice) at different doses (0.5-2.5 mg/kg), and then the replacement and therapeutic effects were examined. RESULTS: The Om4HexA was widely distributed across the ependymal cell layer, dose-dependently restored the enzyme activity due to uptake via cell surface cation-independent M6P receptor (CI-M6PR) on neural cells, and reduced substrates, including GM2 ganglioside (GM2), asialo GM2 (GA2), and oligosaccharides with terminal N-acetylglucosamine residues (GlcNAc-oligosaccharides), accumulated in brain parenchyma. A significant inhibition of chemokine macrophage inflammatory protein-1 α (MIP-1α) induction was also revealed, especially in the hindbrain (< 63%). The decrease in central neural storage correlated with an improvement of motor dysfunction as well as prolongation of the lifespan. INTERPRETATION: This lysosome-directed recombinant human enzyme drug derived from methylotrophic yeast has the high therapeutic potential to improve the motor dysfunction and quality of life of the lysosomal storage diseases (LSDs) patients with neurological manifestations. We emphasize the importance of neural cell surface M6P receptor as a delivery target of neural cell-directed enzyme replacement therapy (NCDERT) for neurodegenerative metabolic diseases.


Subject(s)
Enzyme Replacement Therapy , Gangliosidoses, GM2/drug therapy , Gangliosidoses, GM2/enzymology , Hexosaminidase A/administration & dosage , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Replacement Therapy/methods , Gangliosidoses, GM2/genetics , Gangliosidoses, GM2/pathology , Hexosaminidase A/genetics , Hexosaminidase B/genetics , Humans , Injections, Intraventricular , Lysosomes/enzymology , Mannose-6-Phosphate Isomerase/administration & dosage , Mice , Mice, Knockout , Receptors, CCR1/antagonists & inhibitors , Recombinant Proteins , Sandhoff Disease/drug therapy , Sandhoff Disease/enzymology , Tay-Sachs Disease/drug therapy , Tay-Sachs Disease/genetics , Treatment Outcome , Yeasts
15.
J Vet Diagn Invest ; 23(2): 338-42, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21398459

ABSTRACT

Polymerase chain reaction (PCR)-based assays combined with microchip electrophoresis were developed and evaluated for diagnosis and genotyping of GM2 gangliosidosis variant 0 (Sandhoff-like disease) in Japanese domestic cats. A preliminary genotyping survey was carried out in the population of Japanese domestic cats (1,015 cats in total) in southern Japan. Three kinds of assays including PCR primer-induced restriction analysis (PIRA) and mutagenically separated (MS)-PCR were carried out using blood-stained Flinders Technology Associates filter papers (FTA cards) as templates. The PCR products were analyzed by both agarose gel and microchip electrophoreses. All assays were sufficient to determine the genotypes of this disease, but MS-PCR offered the most rapid and simplest test, as it does not need the restriction enzyme step required in PCR-PIRA. The use of microchip electrophoresis in combination with FTA cards for sampling could shorten the time required for genotyping and simplify the procedure as well. The genotyping survey in the current study did not find any cats that possessed the mutant allele, suggesting that the prevalence of this allele is low (<0.1%) in southern Japan.


Subject(s)
Cat Diseases/diagnosis , Hexosaminidase B/genetics , Polymerase Chain Reaction/veterinary , Sandhoff Disease/veterinary , Animals , Cat Diseases/enzymology , Cat Diseases/genetics , Cats , DNA/chemistry , DNA/genetics , Electrophoresis, Agar Gel/veterinary , Genotype , Hexosaminidase B/metabolism , Japan , Polymerase Chain Reaction/methods , Polymorphism, Single Nucleotide , Sandhoff Disease/diagnosis , Sandhoff Disease/enzymology , Sandhoff Disease/genetics
16.
J Inherit Metab Dis ; 33 Suppl 3: S355-61, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20821051

ABSTRACT

GM2 gangliosidosis type Sandhoff is caused by a defect of beta-hexosaminidase, an enzyme involved in the catabolism of gangliosides. It has been proposed that substrate reduction therapy using N-butyl-deoxynojirimycin (miglustat) may delay neurological progression, at least in late-onset forms of GM2 gangliosidosis. We report the results of a 3-year treatment with miglustat (100 mg t.i.d) in a patient with chronic Sandhoff disease manifesting with an atypical, spinal muscular atrophy phenotype. The follow-up included serial neurological examinations, blood tests, abdominal ultrasound, and neurophysiologic, cognitive, brain, and muscle MRI studies. We document some minor effects on neurological progression in chronic Sandhoff disease by miglustat treatment, confirming the necessity of phase II therapeutic trials including early-stage patients in order to assess its putative efficacy in chronic Sandhoff disease.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enzyme Inhibitors/therapeutic use , Glucosyltransferases/antagonists & inhibitors , Sandhoff Disease/drug therapy , 1-Deoxynojirimycin/therapeutic use , Disease Progression , Glucosyltransferases/metabolism , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle Weakness/diagnosis , Muscle Weakness/drug therapy , Muscle Weakness/etiology , Muscular Atrophy, Spinal/diagnosis , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/etiology , Neurologic Examination , Predictive Value of Tests , Sandhoff Disease/complications , Sandhoff Disease/diagnosis , Sandhoff Disease/enzymology , Sandhoff Disease/genetics , Time Factors , Treatment Outcome
17.
Neurobiol Dis ; 37(3): 656-60, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20005954

ABSTRACT

Sandhoff disease is an autosomal recessive lysosomal disorder due to mutations in the beta-hexosaminidase beta-chain gene, resulting in beta-hexosaminidases A (alphabeta) and B (betabeta) deficiency and GM2 ganglioside accumulation in the brain. In this study, our aim was to demonstrate that transduction of cerebral endothelial cells cultured in two-chamber culture inserts with a lentiviral vector encoding the hexosaminidases alpha and beta chains could induce a vectorial secretion of hexosaminidases. Therefore, the human cerebral endothelial cell line hCMEC/D3 was infected with the bicistronic vector from the apical compartment, and beta-hexosaminidase activity was measured in transduced cells and in deficient fibroblasts co-cultured in the basal (i.e. brain) compartment. Induced beta-hexosaminidase secretion by transduced hCMEC/D3 cells was sufficient to allow for a 70-90% restoration of beta-hexosaminidase activity in deficient fibroblasts. On the basis of these in vitro data, we propose that brain endothelium be considered as a novel therapeutic target in Sandhoff disease.


Subject(s)
Endothelial Cells/enzymology , Genetic Therapy/methods , Sandhoff Disease/enzymology , Sandhoff Disease/therapy , Transduction, Genetic/methods , beta-N-Acetylhexosaminidases/metabolism , Cell Line, Transformed , Cerebral Arteries/cytology , Cerebral Arteries/enzymology , Cerebrum/blood supply , Cerebrum/enzymology , Cerebrum/physiopathology , Coculture Techniques , Diffusion Chambers, Culture , Endothelial Cells/metabolism , Fibroblasts/enzymology , Fibroblasts/metabolism , G(M2) Ganglioside/metabolism , Genetic Vectors/pharmacology , Genetic Vectors/therapeutic use , Humans , Lentivirus/genetics , Sandhoff Disease/genetics , beta-N-Acetylhexosaminidases/genetics
18.
ChemMedChem ; 4(3): 378-92, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19145603

ABSTRACT

N-Acetylhexosaminidases are of considerable importance in mammals and are involved in various significant biological processes. In humans, deficiencies of these enzymes in the lysosome, resulting from inherited genetic defects, cause the glycolipid storage disorders Tay-Sachs and Sandhoff diseases. One promising therapy for these diseases involves the use of beta-N-acetylhexosaminidase inhibitors as chemical chaperones to enhance the enzyme activity above sub-critical levels. Herein we describe the synthesis and biological evaluation of a potent inhibitor, 2-acetamido-1,4-imino-1,2,4-trideoxy-L-arabinitol (LABNAc), in a high-yielding 11-step procedure from D-lyxonolactone. The N-benzyl and N-butyl analogues were also prepared and found to be potent inhibitors. The enantiomers DABNAc and NBn-DABNAc were synthesised from L-lyxonolactone, and were also evaluated. The L-iminosugar LABNAc and its derivatives were found to be potent noncompetitive inhibitors of some beta-N-acetylhexosaminidases, while the D-iminosugar DABNAc and its derivatives were found to be weaker competitive inhibitors. These results support previous work postulating that D-iminosugar mimics inhibit D-glycohydrolases competitively, and that their corresponding L-enantiomers show noncompetitive inhibition of these enzymes. Molecular modelling studies confirm that the spatial organisation in enantiomeric inhibitors leads to a different overlay with the monosaccharide substrate. Initial cell-based studies suggest that NBn-LABNAc can act as a chemical chaperone to enhance the deficient enzyme's activity to levels that may cause a positive pharmacological effect. LABNAc, NBn-LABNAc, and NBu-LABNAc are potent and selective inhibitors of beta-N-acetylhexosaminidase and may be useful as therapeutic agents for treating adult Tay-Sachs and Sandhoff diseases.


Subject(s)
Imino Sugars/chemical synthesis , Imino Sugars/pharmacology , Sandhoff Disease/drug therapy , Tay-Sachs Disease/drug therapy , beta-N-Acetylhexosaminidases/antagonists & inhibitors , Cell Line , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/metabolism , Humans , Imino Sugars/chemistry , Kinetics , Lactones/chemical synthesis , Lactones/chemistry , Models, Molecular , Sandhoff Disease/enzymology , Sandhoff Disease/genetics , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/genetics , beta-N-Acetylhexosaminidases/metabolism
19.
Appl Environ Microbiol ; 73(15): 4805-12, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17557860

ABSTRACT

Human beta-hexosaminidase A (HexA) is a heterodimeric glycoprotein composed of alpha- and beta-subunits that degrades GM2 gangliosides in lysosomes. GM2 gangliosidosis is a lysosomal storage disease in which an inherited deficiency of HexA causes the accumulation of GM2 gangliosides. In order to prepare a large amount of HexA for a treatment based on enzyme replacement therapy (ERT), recombinant HexA was produced in the methylotrophic yeast Ogataea minuta instead of in mammalian cells, which are commonly used to produce recombinant enzymes for ERT. The problem of antigenicity due to differences in N-glycan structures between mammalian and yeast glycoproteins was potentially resolved by using alpha-1,6-mannosyltransferase-deficient (och1Delta) yeast as the host. Genes encoding the alpha- and beta-subunits of HexA were integrated into the yeast cell, and the heterodimer was expressed together with its isozymes HexS (alphaalpha) and HexB (betabeta). A total of 57 mg of beta-hexosaminidase isozymes, of which 13 mg was HexA (alphabeta), was produced per liter of medium. HexA was purified with immobilized metal affinity column for the His tag attached to the beta-subunit. The purified HexA was treated with alpha-mannosidase to expose mannose-6-phosphate (M6P) residues on the N-glycans. The specific activities of HexA and M6P-exposed HexA (M6PHexA) for the artificial substrate 4MU-GlcNAc were 1.2 +/- 0.1 and 1.7 +/- 0.3 mmol/h/mg, respectively. The sodium dodecyl sulfate-polyacrylamide gel electrophoresis pattern suggested a C-terminal truncation in the beta-subunit of the recombinant protein. M6PHexA was incorporated dose dependently into GM2 gangliosidosis patient-derived fibroblasts via M6P receptors on the cell surface, and degradation of accumulated GM2 ganglioside was observed.


Subject(s)
Recombinant Proteins/biosynthesis , Saccharomycetales/enzymology , Sandhoff Disease/enzymology , Tay-Sachs Disease/enzymology , beta-N-Acetylhexosaminidases/biosynthesis , Amino Acid Sequence , Biotechnology/methods , Cells, Cultured , Fibroblasts/enzymology , Hexosaminidase A , Hexosaminidase B , Humans , Molecular Sequence Data , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Saccharomycetales/genetics , Sandhoff Disease/therapy , Tay-Sachs Disease/therapy , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/therapeutic use
20.
J Neurochem ; 101(5): 1294-302, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17442056

ABSTRACT

Retinal abnormalities are well documented in patients with ganglioside storage diseases. The total content and distribution of retinal glycosphingolipids was studied for the first time in control mice and in Sandhoff disease (SD) and GM1 gangliosidosis mice. Light and electron microscopy of the SD and the GM1 retinas revealed storage in ganglion cells. Similar to previous findings in rat retina, GD3 was the major ganglioside in mouse retina, while GM2 and GM1 were minor species. Total ganglioside content was 44% and 40% higher in the SD and the GM1 retinas, respectively, than in the control retinas. Furthermore, GM2 and GM1 content were 11-fold and 51-fold higher in the SD and the GM1 retinas than in the control retinas, respectively. High concentrations of asialo-GM2 and asialo-GM1 were found in the SD and the GM1 retinas, respectively, but were undetectable in the control retinas. The GSL abnormalities in the SD and the GM1 retinas reflect significant reductions in beta-hexosaminidase and beta-galactosidase enzyme activities, respectively. Although electroretinograms appeared normal in the SD and the GM1 mice, visual evoked potentials were subnormal in both mutants, indicating visual impairments. Our findings present a model system for assessing retinal pathobiology and therapies for the gangliosidoses.


Subject(s)
Gangliosidosis, GM1/genetics , Retina/metabolism , Retina/pathology , Sandhoff Disease/pathology , Sandhoff Disease/physiopathology , Age Factors , Animals , Chromatography, High Pressure Liquid/methods , Disease Models, Animal , Electroretinography , Evoked Potentials, Visual/genetics , Female , Gangliosidoses/metabolism , Gene Expression Regulation, Enzymologic/genetics , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Retina/physiopathology , Retina/ultrastructure , Sandhoff Disease/enzymology , Sandhoff Disease/genetics , beta-Galactosidase/genetics , beta-N-Acetylhexosaminidases/deficiency , beta-N-Acetylhexosaminidases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...