Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35162960

ABSTRACT

Inhibition of the human O-linked ß-N-acetylglucosaminidase (hOGA, GH84) enzyme is pharmacologically relevant in several diseases such as neurodegenerative and cardiovascular disorders, type 2 diabetes, and cancer. Human lysosomal hexosaminidases (hHexA and hHexB, GH20) are mechanistically related enzymes; therefore, selective inhibition of these enzymes is crucial in terms of potential applications. In order to extend the structure-activity relationships of OGA inhibitors, a series of 2-acetamido-2-deoxy-d-glucono-1,5-lactone sulfonylhydrazones was prepared from d-glucosamine. The synthetic sequence involved condensation of N-acetyl-3,4,6-tri-O-acetyl-d-glucosamine with arenesulfonylhydrazines, followed by MnO2 oxidation to the corresponding glucono-1,5-lactone sulfonylhydrazones. Removal of the O-acetyl protecting groups by NH3/MeOH furnished the test compounds. Evaluation of these compounds by enzyme kinetic methods against hOGA and hHexB revealed potent nanomolar competitive inhibition of both enzymes, with no significant selectivity towards either. The most efficient inhibitor of hOGA was 2-acetamido-2-deoxy-d-glucono-1,5-lactone 1-naphthalenesulfonylhydrazone (5f, Ki = 27 nM). This compound had a Ki of 6.8 nM towards hHexB. To assess the binding mode of these inhibitors to hOGA, computational studies (Prime protein-ligand refinement and QM/MM optimizations) were performed, which suggested the binding preference of the glucono-1,5-lactone sulfonylhydrazones in an s-cis conformation for all test compounds.


Subject(s)
Antigens, Neoplasm/chemistry , Histone Acetyltransferases/chemistry , Hyaluronoglucosaminidase/chemistry , Hydrazones/chemical synthesis , Lactones/chemistry , beta-Hexosaminidase beta Chain/chemistry , Antigens, Neoplasm/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone Acetyltransferases/metabolism , Humans , Hyaluronoglucosaminidase/metabolism , Hydrazones/chemistry , Hydrazones/pharmacology , Manganese Compounds/chemistry , Models, Molecular , Molecular Conformation , Oxides/chemistry , Structure-Activity Relationship , beta-Hexosaminidase beta Chain/metabolism
3.
Nat Immunol ; 21(7): 802-815, 2020 07.
Article in English | MEDLINE | ID: mdl-32541832

ABSTRACT

Microglia and central nervous system (CNS)-associated macrophages (CAMs), such as perivascular and meningeal macrophages, are implicated in virtually all diseases of the CNS. However, little is known about their cell-type-specific roles in the absence of suitable tools that would allow for functional discrimination between the ontogenetically closely related microglia and CAMs. To develop a new microglia gene targeting model, we first applied massively parallel single-cell analyses to compare microglia and CAM signatures during homeostasis and disease and identified hexosaminidase subunit beta (Hexb) as a stably expressed microglia core gene, whereas other microglia core genes were substantially downregulated during pathologies. Next, we generated HexbtdTomato mice to stably monitor microglia behavior in vivo. Finally, the Hexb locus was employed for tamoxifen-inducible Cre-mediated gene manipulation in microglia and for fate mapping of microglia but not CAMs. In sum, we provide valuable new genetic tools to specifically study microglia functions in the CNS.


Subject(s)
Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Facial Nerve Injuries/pathology , Microglia/metabolism , beta-Hexosaminidase beta Chain/metabolism , Animals , Brain/cytology , Brain/immunology , CRISPR-Cas Systems/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Facial Nerve Injuries/immunology , Gene Knock-In Techniques , Genes, Reporter/genetics , Genetic Loci/genetics , Humans , Intravital Microscopy , Luminescent Agents/chemistry , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Microglia/immunology , NIH 3T3 Cells , RNA-Seq , Single-Cell Analysis , Transfection , beta-Hexosaminidase beta Chain/genetics , Red Fluorescent Protein
4.
Neurobiol Dis ; 134: 104667, 2020 02.
Article in English | MEDLINE | ID: mdl-31682993

ABSTRACT

The favorable outcome of in vivo and ex vivo gene therapy approaches in several Lysosomal Storage Diseases suggests that these treatment strategies might equally benefit GM2 gangliosidosis. Tay-Sachs and Sandhoff disease (the main forms of GM2 gangliosidosis) result from mutations in either the HEXA or HEXB genes encoding, respectively, the α- or ß-subunits of the lysosomal ß-Hexosaminidase enzyme. In physiological conditions, α- and ß-subunits combine to generate ß-Hexosaminidase A (HexA, αß) and ß-Hexosaminidase B (HexB, ßß). A major impairment to establishing in vivo or ex vivo gene therapy for GM2 gangliosidosis is the need to synthesize the α- and ß-subunits at high levels and with the correct stoichiometric ratio, and to safely deliver the therapeutic products to all affected tissues/organs. Here, we report the generation and in vitro validation of novel bicistronic lentiviral vectors (LVs) encoding for both the murine and human codon optimized Hexa and Hexb genes. We show that these LVs drive the safe and coordinate expression of the α- and ß-subunits, leading to supranormal levels of ß-Hexosaminidase activity with prevalent formation of a functional HexA in SD murine neurons and glia, murine bone marrow-derived hematopoietic stem/progenitor cells (HSPCs), and human SD fibroblasts. The restoration/overexpression of ß-Hexosaminidase leads to the reduction of intracellular GM2 ganglioside storage in transduced and in cross-corrected SD murine neural progeny, indicating that the transgenic enzyme is secreted and functional. Importantly, bicistronic LVs safely and efficiently transduce human neurons/glia and CD34+ HSPCs, which are target and effector cells, respectively, in prospective in vivo and ex vivo GT approaches. We anticipate that these bicistronic LVs may overcome the current requirement of two vectors co-delivering the α- or ß-subunits genes. Careful assessment of the safety and therapeutic potential of these bicistronic LVs in the SD murine model will pave the way to the clinical development of LV-based gene therapy for GM2 gangliosidosis.


Subject(s)
Gangliosidoses, GM2/metabolism , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Neural Stem Cells/metabolism , beta-Hexosaminidase alpha Chain/metabolism , beta-Hexosaminidase beta Chain/metabolism , Animals , Gangliosidoses, GM2/genetics , Genetic Vectors , Humans , Lentivirus , Mice , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics
5.
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
6.
Synapse ; 71(7)2017 07.
Article in English | MEDLINE | ID: mdl-28295625

ABSTRACT

Mutations in the glucocerebrosidase 1 (GBA1) gene are related to both Parkinson disease (PD) and Gaucher disease (GD). In both cases, the condition is associated with deficiency of glucocerebrosidase (GCase), the enzyme encoded by GBA1. Ambroxol is a small molecule chaperone that has been shown in mice to cross the blood-brain barrier, increase GCase activity and reduce alpha-synuclein protein levels. In this study, we analyze the effect of ambroxol treatment on GCase activity in healthy nonhuman primates. We show that daily administration of ambroxol results in increased brain GCase activity. Our work further indicates that ambroxol should be investigated as a novel therapy for both PD and neuronopathic GD in humans.


Subject(s)
Ambroxol/administration & dosage , Brain/drug effects , Brain/enzymology , Central Nervous System Agents/administration & dosage , Glucosylceramidase/metabolism , Administration, Oral , Animals , Macaca fascicularis , Male , Preliminary Data , beta-Hexosaminidase beta Chain/metabolism
7.
Sci Rep ; 7: 40518, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28084424

ABSTRACT

Sandhoff disease (SD) is caused by the loss of ß-hexosaminidase (Hex) enzymatic activity in lysosomes resulting from Hexb mutations. In SD patients, the Hex substrate GM2 ganglioside accumulates abnormally in neuronal cells, resulting in neuronal loss, microglial activation, and astrogliosis. Hexb-/- mice, which manifest a phenotype similar to SD, serve as animal models for examining the pathophysiology of SD. Hexb-/- mice reach ~8 weeks without obvious neurological defects; however, trembling begins at 12 weeks and is accompanied by startle reactions and increased limb tone. These symptoms gradually become severe by 16-18 weeks. Immune reactions caused by autoantibodies have been recently associated with the pathology of SD. The inhibition of immune activation may represent a novel therapeutic target for SD. Herein, SD mice (Hexb-/-) were crossed to mice lacking an activating immune receptor (FcRγ-/-) to elucidate the potential relationship between immune responses activated through SD autoantibodies and astrogliosis. Microglial activation and astrogliosis were observed in cortices of Hexb-/- mice during the asymptomatic phase, and were inhibited in Hexb-/- FcRγ-/- mice. Moreover, early astrogliosis and impaired motor coordination in Hexb-/- mice could be ameliorated by immunosuppressants, such as FTY720. Our findings demonstrate the importance of early treatment and the therapeutic effectiveness of immunosuppression in SD.


Subject(s)
Astrocytes/immunology , Astrocytes/pathology , Gliosis/immunology , Gliosis/pathology , Immunity , Receptors, Fc/metabolism , Sandhoff Disease/immunology , Sandhoff Disease/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Fingolimod Hydrochloride/pharmacology , G(M2) Ganglioside/metabolism , Heterozygote , Immunity/drug effects , Immunosuppressive Agents/pharmacology , Mice, Inbred C57BL , Motor Activity/drug effects , Phenotype , Receptors, Fc/deficiency , Sandhoff Disease/physiopathology , Up-Regulation/drug effects , Walking , beta-Hexosaminidase beta Chain/metabolism
8.
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
9.
Mol Ther ; 23(3): 414-22, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25515709

ABSTRACT

G(M2) gangliosidoses are severe neurodegenerative disorders resulting from a deficiency in ß-hexosaminidase A activity and lacking effective therapies. Using a Sandhoff disease (SD) mouse model (Hexb(-/-)) of the G(M2) gangliosidoses, we tested the potential of systemically delivered adeno-associated virus 9 (AAV9) expressing Hexb cDNA to correct the neurological phenotype. Neonatal or adult SD and normal mice were intravenously injected with AAV9-HexB or -LacZ and monitored for serum ß-hexosaminidase activity, motor function, and survival. Brain G(M2) ganglioside, ß-hexosaminidase activity, and inflammation were assessed at experimental week 43, or an earlier humane end point. SD mice injected with AAV9-LacZ died by 17 weeks of age, whereas all neonatal AAV9-HexB-treated SD mice survived until 43 weeks (P < 0.0001) with only three exhibiting neurological dysfunction. SD mice treated as adults with AAV9-HexB died between 17 and 35 weeks. Neonatal SD-HexB-treated mice had a significant increase in brain ß-hexosaminidase activity, and a reduction in G(M2) ganglioside storage and neuroinflammation compared to adult SD-HexB- and SD-LacZ-treated groups. However, at 43 weeks, 8 of 10 neonatal-HexB injected control and SD mice exhibited liver or lung tumors. This study demonstrates the potential for long-term correction of SD and other G(M2) gangliosidoses through early rAAV9 based systemic gene therapy.


Subject(s)
Dependovirus/genetics , G(M2) Ganglioside/metabolism , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Sandhoff Disease/therapy , beta-Hexosaminidase beta Chain/genetics , Age Factors , Animals , Animals, Newborn , Brain/enzymology , Brain/pathology , Disease Models, Animal , Female , Gene Expression , Genetic Vectors/adverse effects , Inflammation/genetics , Inflammation/mortality , Inflammation/pathology , Inflammation/therapy , Injections, Intravenous , Lac Operon , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Lysosomes/enzymology , Lysosomes/pathology , Male , Mice , Mice, Knockout , Motor Activity/genetics , Sandhoff Disease/genetics , Sandhoff Disease/mortality , Sandhoff Disease/pathology , Survival Analysis , beta-Hexosaminidase beta Chain/metabolism
10.
PLoS One ; 8(1): e55856, 2013.
Article in English | MEDLINE | ID: mdl-23383290

ABSTRACT

Sandhoff disease (SD) is a glycosphingolipid storage disease that arises from mutations in the Hexb gene and the resultant deficiency in ß-hexosaminidase activity. This deficiency results in aberrant lysosomal accumulation of the ganglioside GM2 and related glycolipids, and progressive deterioration of the central nervous system. Dysfunctional glycolipid storage causes severe neurodegeneration through a poorly understood pathogenic mechanism. Induced pluripotent stem cell (iPSC) technology offers new opportunities for both elucidation of the pathogenesis of diseases and the development of stem cell-based therapies. Here, we report the generation of disease-specific iPSCs from a mouse model of SD. These mouse model-derived iPSCs (SD-iPSCs) exhibited pluripotent stem cell properties and significant accumulation of GM2 ganglioside. In lineage-directed differentiation studies using the stromal cell-derived inducing activity method, SD-iPSCs showed an impaired ability to differentiate into early stage neural precursors. Moreover, fewer neurons differentiated from neural precursors in SD-iPSCs than in the case of the wild type. Recovery of the Hexb gene in SD-iPSCs improved this impairment of neuronal differentiation. These results provide new insights as to understanding the complex pathogenic mechanisms of SD.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Sandhoff Disease/etiology , Animals , Cell Differentiation/genetics , Disease Models, Animal , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/ultrastructure , Male , Mice , Mice, Knockout , Neurons/metabolism , Neurons/ultrastructure , Phenotype , Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/metabolism
11.
J Biochem ; 153(1): 111-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23127958

ABSTRACT

The adult form of Sandhoff disease with the motor neuron disease phenotype is a rare neurodegenerative disorder caused by mutations in HEXB encoding the ß-subunit of ß-hexosaminidase, yet the properties of mutant ß-subunits of the disease have not been fully determined. We identified a novel mutation (H235Y) in the ß-sheet of the (ß/α)8-barrel domain, in addition to the previously reported P417L mutation that causes aberrant splicing, in a Japanese patient with the motor neuron disease phenotype. Enzyme assays, gel filtration studies and immunoprecipitation studies with HEK293 cells transiently expressing mutant ß-subunits demonstrated that the H235Y mutation abolished both α-ß and ß-ß dimer formation without increasing ß-hexosaminidase activity, whereas other reported mutant ß-subunits (Y456S, P504S or R533H) associated with the motor neuron disease phenotype formed dimers. Structural analysis suggested that the H235Y mutation in the ß-sheet of the (ß/α)8-barrel domain changed the conformation of the ß-subunit by causing a clash with the E288 side chain. In summary, H235Y is the first mutation in the ß-sheet of the (ß/α)8-barrel domain of the ß-subunit that abolishes α-ß and ß-ß dimer formation; the presented patient is the second patient to exhibit the motor neuron disease phenotype with P417L and a non-functional allele of HEXB.


Subject(s)
Motor Neuron Disease/genetics , Mutant Proteins/metabolism , Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , Amino Acid Substitution , Humans , Male , Middle Aged , Models, Molecular , Motor Neuron Disease/metabolism , Motor Neuron Disease/physiopathology , Mutant Proteins/chemistry , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sandhoff Disease/metabolism , Sandhoff Disease/physiopathology , beta-Hexosaminidase beta Chain/chemistry , beta-Hexosaminidase beta Chain/metabolism
12.
PLoS One ; 7(7): e41516, 2012.
Article in English | MEDLINE | ID: mdl-22848519

ABSTRACT

Sandhoff disease (SD) is a lysosomal disorder caused by mutations in the HEXB gene. To date, 43 mutations of HEXB have been described, including 3 large deletions. Here, we have characterized 14 unrelated SD patients and developed a Multiplex Ligation-dependent Probe Amplification (MLPA) assay to investigate the presence of large HEXB deletions. Overall, we identified 16 alleles, 9 of which were novel, including 4 sequence variation leading to aminoacid changes [c.626C>T (p.T209I), c.634C>A (p.H212N), c.926G>T (p.C309F), c.1451G>A (p.G484E)] 3 intronic mutations (c.1082+5G>A, c.1242+1G>A, c.1169+5G>A), 1 nonsense mutation c.146C>A (p.S49X) and 1 small in-frame deletion c.1260_1265delAGTTGA (p.V421_E422del). Using the new MLPA assay, 2 previously described deletions were identified. In vitro expression studies showed that proteins bearing aminoacid changes p.T209I and p.G484E presented a very low or absent activity, while proteins bearing the p.H212N and p.C309F changes retained a significant residual activity. The detrimental effect of the 3 novel intronic mutations on the HEXB mRNA processing was demonstrated using a minigene assay. Unprecedentedly, minigene studies revealed the presence of a novel alternative spliced HEXB mRNA variant also present in normal cells. In conclusion, we provided new insights into the molecular basis of SD and validated an MLPA assay for detecting large HEXB deletions.


Subject(s)
Alternative Splicing/genetics , Base Sequence , Sandhoff Disease/genetics , Sequence Deletion , beta-Hexosaminidase beta Chain/genetics , Female , HEK293 Cells , Humans , Male , Multiplex Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sandhoff Disease/metabolism , beta-Hexosaminidase beta Chain/metabolism
13.
Cell Biochem Funct ; 30(1): 61-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21997228

ABSTRACT

In an attempt to investigate whether the genetic defect in the HEXA and HEXB genes (which causes the absence of the lysosomal ß-N-acetyl-hexosaminidase), are related to the wide inflammation in GM2 gangliosidoses (Tay-Sachs and Sandhoff disease), we have chosen the dendritic cells (DCs) as a study model. Using the RNA interference approach, we generated an in vitro model of HEXs knock-down immunogenic DCs (i-DCs) from CD34(+)-haemopoietic stem cells (CD34(+)-HSCs), thus mimicking the Tay-Sachs (HEXA-/-) and Sandhoff (HEXB-/-) cells. We showed that the absence of ß-N-acetyl-hexosaminidase activity does not alter the differentiation of i-DCs from HSCs, but it is critical for the activation of CD4(+)T cells because knock-down of HEXA or HEXB gene causes a loss of function of i-DCs. Notably, the silencing of the HEXA gene had a stronger immune inhibitory effect, thereby indicating a major involvement of ß-N-acetyl-hexosaminidase A isoenzyme within this mechanism.


Subject(s)
Dendritic Cells/immunology , Stem Cells/immunology , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gangliosidoses, GM2/immunology , Gangliosidoses, GM2/metabolism , Gene Knockdown Techniques , Humans , Inflammation/immunology , Inflammation/metabolism , Stem Cells/cytology , Stem Cells/metabolism , beta-Hexosaminidase alpha Chain/metabolism , beta-Hexosaminidase beta Chain/metabolism
14.
Mol Ther ; 19(6): 1017-24, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21487393

ABSTRACT

To develop a novel enzyme replacement therapy for neurodegenerative Tay-Sachs disease (TSD) and Sandhoff disease (SD), which are caused by deficiency of ß-hexosaminidase (Hex) A, we designed a genetically engineered HEXB encoding the chimeric human ß-subunit containing partial amino acid sequence of the α-subunit by structure-based homology modeling. We succeeded in producing the modified HexB by a Chinese hamster ovary (CHO) cell line stably expressing the chimeric HEXB, which can degrade artificial anionic substrates and GM2 ganglioside in vitro, and also retain the wild-type (WT) HexB-like thermostability in the presence of plasma. The modified HexB was efficiently incorporated via cation-independent mannose 6-phosphate receptor into fibroblasts derived from Tay-Sachs patients, and reduced the GM2 ganglioside accumulated in the cultured cells. Furthermore, intracerebroventricular administration of the modified HexB to Sandhoff mode mice restored the Hex activity in the brains, and reduced the GM2 ganglioside storage in the parenchyma. These results suggest that the intracerebroventricular enzyme replacement therapy involving the modified HexB should be more effective for Tay-Sachs and Sandhoff than that utilizing the HexA, especially as a low-antigenic enzyme replacement therapy for Tay-Sachs patients who have endogenous WT HexB.


Subject(s)
G(M2) Ganglioside/metabolism , beta-Hexosaminidase beta Chain/chemistry , beta-Hexosaminidase beta Chain/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Immunoblotting , Mice , Models, Molecular , Protein Structure, Secondary , Sandhoff Disease/drug therapy , Tay-Sachs Disease/drug therapy , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/therapeutic use
15.
PLoS One ; 5(8)2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20856892

ABSTRACT

BACKGROUND: Sandhoff disease is a lysosomal storage disorder characterized by the absence of ß-hexosaminidase and storage of GM2 ganglioside and related glycolipids. We have previously found that the progressive neurologic disease induced in Hexb(-/-) mice, an animal model for Sandhoff disease, is associated with the production of pathogenic anti-glycolipid autoantibodies. METHODOLOGY/PRINCIPAL FINDINGS: In our current study, we report on the alterations in the thymus during the development of mild to severe progressive neurologic disease. The thymus from Hexb(-/-) mice of greater than 15 weeks of age showed a marked decrease in the percentage of immature CD4(+)/CD8(+) T cells and a significantly increased number of CD4(+)/CD8(-) T cells. During involution, the levels of both apoptotic thymic cells and IgG deposits to T cells were found to have increased, whilst swollen macrophages were prominently observed, particularly in the cortex. We employed cDNA microarray analysis to monitor gene expression during the involution process and found that genes associated with the immune responses were upregulated, particularly those expressed in macrophages. CXCL13 was one of these upregulated genes and is expressed specifically in the thymus. B1 cells were also found to have increased in the thy mus. It is significant that these alterations in the thymus were reduced in FcRγ additionally disrupted Hexb(-/-) mice. CONCLUSIONS/SIGNIFICANCE: These results suggest that the FcRγ chain may render the usually poorly immunogenic thymus into an organ prone to autoimmune responses, including the chemotaxis of B1 cells toward CXCL13.


Subject(s)
Gangliosidoses, GM2/immunology , Gangliosidoses, GM2/pathology , Thymus Gland/immunology , Thymus Gland/pathology , Aging/pathology , Animals , Atrophy/metabolism , Autoantibodies/biosynthesis , Autoimmunity/immunology , Cell Death/immunology , Chemokine CXCL13/genetics , Disease Models, Animal , Disease Progression , Gangliosidoses, GM2/genetics , Gangliosidoses, GM2/metabolism , Gene Expression Profiling , Gene Expression Regulation/immunology , Humans , Infant , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, IgG/deficiency , Receptors, IgG/metabolism , Sandhoff Disease/genetics , Sandhoff Disease/immunology , Sandhoff Disease/metabolism , Sandhoff Disease/pathology , Thymus Gland/metabolism , beta-Hexosaminidase beta Chain/metabolism
16.
Mol Ther ; 18(8): 1519-26, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20571546

ABSTRACT

Human lysosomal beta-hexosaminidase A is a heterodimer composed of alpha- and beta-subunits encoded by HEXA and HEXB, respectively. We genetically introduced an additional N-glycosylation sequon into HEXA, which caused amino acid substitutions (S51 to N and A53 to T) at homologous positions to N84 and T86 in the beta-subunit. The mutant HexA (NgHexA) obtained from a Chinese hamster ovary (CHO) cell line co-expressing the mutated HEXA and wild-type HEXB complementary DNAs was demonstrated to contain an additional mannose-6-phosphate (M6P)-type-N-glycan. NgHexA was more efficiently taken up than the wild-type HexA and delivered to lysosomes, where it degraded accumulated substrates including GM2 ganglioside (GM2) when administered to cultured fibroblasts derived from a Sandhoff disease (SD) patient. On intracerebroventricular (i.c.v.) administration of NgHexA to SD model mice, NgHexA more efficiently restored the HexA activity and reduced the GM2 and GA2 (asialoGM2) accumulated in neural cells of the brain parenchyma than the wild-type HexA. These findings indicate that i.c.v. administration of the modified human HexA with an additional M6P-type N-glycan is applicable for enzyme replacement therapy (ERT) involving an M6P-receptor as a molecular target for HexA deficiencies including Tay-Sachs disease and SD.


Subject(s)
Polysaccharides/metabolism , Sandhoff Disease/metabolism , beta-Hexosaminidase alpha Chain/metabolism , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/therapeutic use , Animals , CHO Cells , Cells, Cultured , Chromatography, Thin Layer , Cricetinae , Cricetulus , G(M2) Ganglioside/metabolism , Glycosylation , Humans , Immunoblotting , Mice , Polysaccharides/chemistry , Sandhoff Disease/drug therapy , Sandhoff Disease/genetics , beta-Hexosaminidase alpha Chain/chemistry , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase alpha Chain/therapeutic use , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/metabolism , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism
17.
J Proteome Res ; 8(6): 2740-51, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19284783

ABSTRACT

Invariant NKT cells are a hybrid cell type of Natural Killer cells and T cells, whose development is dependent on thymic positive selection mediated by double positive thymocytes through their recognition of natural ligands presented by CD1d, a nonpolymorphic, non-MHC, MHC-like antigen presenting molecule. Genetic evidence suggested that beta-glucosylceramide derived glycosphingolipids (GSLs) are natural ligands for NKT cells. N-butyldeoxygalactonojirimycin (NB-DGJ), a drug that specifically inhibits the glucosylceramide synthase, inhibits the endogenous ligands for NKT cells. Furthermore, we and others have found a beta-linked glycosphingolipid, isoglobotriaosylceramide (iGb3), is a stimulatory NKT ligand. The iGb3 synthase knockout mice have a normal NKT development and function, indicating that other ligands exist and remain to be identified. In this study, we have performed a glycosphingolipidomics study of mouse thymus, and studied mice mutants which are deficient in beta-hexosaminidase b or alpha-galactosidase A, two glycosidases that are up- and downstream agents of iGb3 turnover, respectively. Our mass spectrometry methods generated a first database for glycosphingolipids expressed in mouse thymus, which are specifically regulated by rate-limiting glycosidases. Among the identified thymic glycosphingolipids, only iGb3 is a stimulatory ligand for NKT cells, suggesting that large-scale fractionation, enrichment and characterization of minor species of glycosphingolipids are necessary for identifying additional ligands for NKT cells. Our results also provide early insights into cellular lipidomics studies, with a specific focus on the important immunological functions of glycosphingolipids.


Subject(s)
Globosides/metabolism , Glycosphingolipids/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Thymus Gland/cytology , Animals , Galactosyltransferases/metabolism , Gene Expression , Genomics/methods , Globosides/immunology , Glycosphingolipids/immunology , Hydrogen-Ion Concentration , Ligands , Mice , Mice, Knockout , Thymus Gland/immunology , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/metabolism
18.
Mol Genet Metab ; 97(1): 53-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19231264

ABSTRACT

GM2 gangliosidosis is a fatal, progressive neuronopathic lysosomal storage disease resulting from a deficiency of beta-N-acetylhexosaminidase (EC 3.2.1.52) activity. GM2 gangliosidosis occurs with varying degrees of severity in humans and in a variety of animals, including cats. In the current research, European Burmese cats presented with clinical neurological signs and histopathological features typical of a lysosomal storage disease. Thin layer chromatography revealed substantial storage of GM2 ganglioside in brain tissue of affected cats, and assays with a synthetic fluorogenic substrate confirmed the absence of hexosaminidase activity. When the hexosaminidase beta-subunit cDNA was sequenced from affected cats, a 91 base pair deletion constituting the entirety of exon 12 was documented. Subsequent sequencing of introns 11 and 12 revealed a 15 base pair deletion at the 3' end of intron 11 that included the preferred splice acceptor site, generating two minor transcripts from cryptic splice acceptor sites in affected Burmese cats. In the cerebral cortex of affected cats, hexosaminidase beta-subunit mRNA levels were approximately 1.5 times higher than normal (P<0.001), while beta-subunit protein levels were substantially reduced on Western blots.


Subject(s)
Cat Diseases/enzymology , Lysosomal Storage Diseases/veterinary , Nerve Degeneration/complications , Nerve Degeneration/enzymology , beta-Hexosaminidase beta Chain/metabolism , Animals , Base Sequence , Blotting, Western , Cats , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Chromatography, Thin Layer , DNA Mutational Analysis , Europe , Gangliosidoses, GM2/enzymology , Gangliosidoses, GM2/pathology , Lipids/analysis , Lysosomal Storage Diseases/complications , Lysosomal Storage Diseases/enzymology , Molecular Sequence Data , Myanmar
19.
Mol Genet Metab ; 95(4): 236-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18930675

ABSTRACT

Mutations in HEXB, encoding the beta-subunit common to hexosaminidases A and B, cause the neurodegenerative condition, Sandhoff disease. A homozygous missense HEXB mutation (p. D459A) was discovered in six patients with a rare juvenile variant: we show that this disrupts a salt bridge between aspartate D459 and arginine 505 at the subunit interface; R505 mutations are reported in late-onset Sandhoff disease. Identification of D459A contributes to diagnosis and molecular understanding of attenuated Sandhoff disease variants.


Subject(s)
Mutation, Missense , Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/chemistry , beta-Hexosaminidase beta Chain/genetics , Adolescent , Child , Child, Preschool , Female , Genotype , Humans , Male , Pedigree , White People/genetics , beta-Hexosaminidase beta Chain/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...