Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Neurochem ; 163(5): 375-390, 2022 12.
Article in English | MEDLINE | ID: mdl-36227633

ABSTRACT

Phosphacan, a chondroitin sulfate proteoglycan, is a repulsive cue of cerebellar granule cells. This study aims to explore the molecular mechanism. The glycosylphosphatidylinositol-anchored neural adhesion molecule TAG-1 is a binding partner of phosphacan, suggesting that the repulsive effect of phosphacan is possibly because of its interaction with TAG-1. The repulsive effect was greatly reduced on primary cerebellar granule cells of TAG-1-deficient mice. Surface plasmon resonance analysis confirmed the direct interaction of TAG-1 with chondroitin sulfate C. On postnatal days 1, 4, 7, 11, 15, and 20 and in adulthood, phosphacan was present in the molecular layer and internal granular layer, but not in the external granular layer. In contrast, transient TAG-1 expression was observed exclusively within the premigratory zone of the external granular layer on postnatal days 1, 4, 7, and 11. Boyden chamber cell migration assay demonstrated that phosphacan exerted its repulsive effect on the spontaneous and brain-derived neurotrophic factor (BDNF)-induced migration of cerebellar granule cells. The BDNF-induced migration was inhibited by MK-2206, an Akt inhibitor. The pre-treatment with a raft-disrupting agent, methyl-ß-cyclodextrin, also inhibited the BDNF-induced migration, suggesting that lipid rafts are involved in the migration of cerebellar granule cells. In primary cerebellar granule cells obtained on postnatal day 7 and cultured for 7 days, the ganglioside GD3 and TAG-1 preferentially localized in the cell body, whereas the ganglioside GD1b and NB-3 localized in not only the cell body but also neurites. Pre-treatment with the anti-GD3 antibody R24, but not the anti-GD1b antibody GGR12, inhibited the spontaneous and BDNF-induced migration, and attenuated BDNF-induced Akt activation. These findings suggest that phosphacan is responsible for the repulsion of TAG-1-expressing cerebellar granule cells via GD3 rafts to attenuate BDNF-induced migration signaling.


Subject(s)
Cell Adhesion Molecules, Neuronal , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Animals , Mice , Rats , Brain-Derived Neurotrophic Factor/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cerebellum/metabolism , Membrane Microdomains/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism
2.
PLoS One ; 16(7): e0253912, 2021.
Article in English | MEDLINE | ID: mdl-34197550

ABSTRACT

Protein kinase C-delta (PKCδ) has a caspase-3 recognition sequence in its structure, suggesting its involvement in apoptosis. In addition, PKCδ was recently reported to function as an anti-cancer factor. The generation of a PKCδ knockout mouse model indicated that PKCδ plays a role in B cell homeostasis. However, the Pkcrd gene, which is regulated through complex transcription, produces multiple proteins via alternative splicing. Since gene mutations can result in the loss of function of molecular species required for each tissue, in the present study, conditional PKCδ knockout mice lacking PKCδI, II, IV, V, VI, and VII were generated to enable tissue-specific deletion of PKCδ using a suitable Cre mouse. We generated PKCδ-null mice that lacked whole-body expression of PKCδ. PKCδ+/- parental mice gave birth to only 3.4% PKCδ-/- offsprings that deviated significantly from the expected Mendelian ratio (χ2(2) = 101.7, P < 0.001). Examination of mice on embryonic day 11.5 (E11.5) showed the proportion of PKCδ-/- mice implanted in the uterus in accordance with Mendelian rules; however, approximately 70% of the fetuses did not survive at E11.5. PKCδ-/- mice that survived until adulthood showed enlarged spleens, with some having cardiac and pulmonary abnormalities. Our findings suggest that the lack of PKCδ may have harmful effects on fetal development, and heart and lung functions after birth. Furthermore, our study provides a reference for future studies on PKCδ deficient mice that would elucidate the effects of the multiple protein variants in mice and decipher the roles of PKCδ in various diseases.


Subject(s)
Elastic Tissue/pathology , Fetal Development/genetics , Lung/pathology , Pneumonia/genetics , Protein Kinase C-delta/deficiency , Animals , Disease Models, Animal , Elastic Tissue/immunology , Female , Humans , Hyperplasia/genetics , Hyperplasia/pathology , Lung/immunology , Male , Mice , Mice, Knockout , Pneumonia/immunology , Pneumonia/pathology , Protein Kinase C-delta/genetics
3.
Chemistry ; 26(59): 13400-13407, 2020 Oct 21.
Article in English | MEDLINE | ID: mdl-32662545

ABSTRACT

Efficient catalytic arsa-Wittig reactions have been developed by using 1-phenylarsolane as a catalyst. A wide array of aldehydes was converted to the corresponding olefins in high yields with moderate to excellent E stereoselectivity in the presence of a catalytic amount of 1-phenylarsolane. Moreover, density functional theory calculations were carried out to afford insight into the E/Z selectivity.

4.
Angew Chem Int Ed Engl ; 58(34): 11686-11690, 2019 Aug 19.
Article in English | MEDLINE | ID: mdl-31251829

ABSTRACT

Dibenzo[b,f]arsepins possessing severely distorted cores compared to those of other heteropins were synthesized. These derivatives exhibited dual photoluminescence in the green-to-red region (500-700 nm) and the near-ultraviolet region (<380 nm), which could be attributed to the planarization of the arsepin core in the lowest singlet excited (S1 ) state. The computational approach for the assessment of the aromatic indices revealed that the dibenzoarsepins studied show aromaticity (8π system) in the S1 states in line with Baird's rule. The lone pair electrons of the arsenic atoms play a crucial role in the aromaticity in the S1 states.

5.
Methods Mol Biol ; 1804: 285-291, 2018.
Article in English | MEDLINE | ID: mdl-29926414

ABSTRACT

The antibody preparation method to the glycolipid is basically same to the method to the protein. However, because the immunogenicity of the carbohydrate moieties of glycolipid is generally low in the mouse, the development of the anti-glycolipid antibody using purified glycolipid as an immunogen was not yet established. Here, we describe a method using a purified ganglioside adsorbed onto Salmonella minnesota for the efficient production of an anti-ganglioside mouse monoclonal antibody that recognizes the carbohydrate moieties of the ganglioside.


Subject(s)
Biochemistry/methods , Gangliosides/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Cell Fusion , Hybridomas , Immunization , Mice , Salmonella/metabolism , Spleen/cytology
6.
Chemistry ; 24(35): 8797-8803, 2018 Jun 21.
Article in English | MEDLINE | ID: mdl-29719074

ABSTRACT

2,3,4,5-Tetraaryl-1-phenylarsoles were synthesized by utilizing safely generated diiodophenylarsine and zirconacyclopentadienes. The obtained peraryl arsoles showed aggregation-induced emission (AIE), where intense emission was observed in the solid states (quantum yields up to 0.61), whereas the corresponding solutions were very weakly emissive. The optical and electronic properties were examined by experimental and computational methods. It was elucidated that the aryl groups at the 2,5-positions affected the frontier orbitals and the aromaticity of the arsole core. On the other hand, those at the 1,3,5-positions were perpendicular to the luminophore and effective for a restriction of aggregation-caused quenching. Because the lone pair of the arsenic atom has a sufficient coordination ability due to the low aromaticity of the arsole moiety, a gold(I) chloride complex of 1,2,3,4,5-pentaphenylarsole was synthesized. The complex formation caused a blue shift of the emission from the bare ligand. Interestingly, the complex showed luminescent mechanochromism; grinding the crystals with a blue emission (λem =445 nm) gave amorphous samples with a greenish-blue emission (λem =496 nm).

7.
Mol Genet Metab ; 120(1-2): 116-120, 2017.
Article in English | MEDLINE | ID: mdl-27756537

ABSTRACT

In Fabry disease, large amounts of globotriaosylceramide (Gb3) and related glycosphingolipids accumulate in organs due to a deficiency of α-galactosidase A (GLA) activity. Enzyme replacement therapy (ERT) with recombinant GLA is now available, and it has been reported that ERT is beneficial for patients with Fabry disease, especially those who start treatment at an early stage of the disease. However, it seems that the efficacy of ERT differs with each organ, and Gb3 accumulated in the kidneys shows resistance to ERT when it is started at a late stage. In this study, we examined the differences in cleavage of Gb3 isoforms, and lyso-Gb3 and its analogues in the kidneys, liver, and heart in young Fabry mice subjected to ERT. The results revealed that recurrent administration of recombinant GLA had prominent effects in terms of degradation of Gb3 and its derivatives accumulated in the organs. However, particular Gb3 isoforms, i.e., Gb3 (C20:0) and Gb3 (C24OH), accumulated in the kidneys largely escaped from degradation. Such Gb3 isoforms may gradually accumulate in the kidneys from a young age, which results in a reduction in the efficacy of ERT for Fabry disease.


Subject(s)
Fabry Disease/drug therapy , Isoenzymes/therapeutic use , Kidney/metabolism , Trihexosylceramides/chemistry , alpha-Galactosidase/therapeutic use , Animals , Disease Models, Animal , Drug Resistance , Enzyme Replacement Therapy , Fabry Disease/metabolism , Humans , Mice
8.
PLoS One ; 8(11): e78588, 2013.
Article in English | MEDLINE | ID: mdl-24236025

ABSTRACT

Fabry disease is an X-linked genetic disorder caused by defects in the α-galactosidase A (GLA) gene, and heterogeneous mutations lead to quantitative and/or qualitative defects in GLA protein in male patients with Fabry disease. Random X-chromosomal inactivation modifies the clinical and biochemical features of female patients with Fabry disease. Functional polymorphisms have been frequently reported in recent times, and these increase the difficulty of understanding the pathogenetic basis of the disease. To date, GLA protein level has been measured using an enzyme-linked immunosorbent assay (ELISA). However, ELISA is not highly sensitive due to the high background noise. In this paper, we introduce a novel application of the immuno-polymerase chain reaction (PCR) method (termed Multiple Simultaneous Tag [MUSTag]) for measurement of the GLA protein level in blood samples. We compared the sensitivities of the MUSTag method with plates or magnetic beads with those of ELISA for recombinant human GLA and found that the apparent maximal sensitivity was higher for the former than for the latter. We then measured the GLA concentrations in serum and plasma from male patients with classic Fabry disease (Male Fabry), females with Fabry disease (Female Fabry), male subjects harboring the functional polymorphism p.E66Q (E66Q), and control (Control) subjects. Our results revealed that compared to the MUSTag plate and ELISA, the MUSTag beads assay afforded a clearer estimation of the GLA protein levels in the serum and plasma with minimal or no background noise, although all the methods could differentiate between the Male Fabry, E66Q, and Control groups. The Female Fabry group showed characteristic heterogeneity, which was consistent with the X-linked inheritance. This novel method is expected to be useful for the sensitive determination of GLA level in blood and elucidation of the pathogenetic basis of Fabry disease.


Subject(s)
Fabry Disease/diagnosis , Molecular Diagnostic Techniques , alpha-Galactosidase/blood , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Fabry Disease/blood , Fabry Disease/enzymology , Female , Humans , Male , Polymerase Chain Reaction , Sensitivity and Specificity
9.
Blood ; 122(19): 3340-8, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24002447

ABSTRACT

Membrane rafts are spatially and functionally heterogenous in the cell membrane. We observed that lysenin-positive sphingomyelin (SM)-rich rafts are identified histochemically in the central region of adhered platelets where fibrin and myosin are colocalized on activation by thrombin. The clot retraction of SM-depleted platelets from SM synthase knockout mouse was delayed significantly, suggesting that platelet SM-rich rafts are involved in clot retraction. We found that fibrin converted by thrombin translocated immediately in platelet detergent-resistant membrane (DRM) rafts but that from Glanzmann's thrombasthenic platelets failed. The fibrinogen γ-chain C-terminal (residues 144-411) fusion protein translocated to platelet DRM rafts on thrombin activation, but its mutant that was replaced by A398A399 at factor XIII crosslinking sites (Q398Q399) was inhibited. Furthermore, fibrin translocation to DRM rafts was impaired in factor XIII A subunit-deficient mouse platelets, which show impaired clot retraction. In the cytoplasm, myosin translocated concomitantly with fibrin translocation into the DRM raft of thrombin-stimulated platelets. Furthermore, the disruption of SM-rich rafts by methyl-ß-cyclodextrin impaired myosin activation and clot retraction. Thus, we propose that clot retraction takes place in SM-rich rafts where a fibrin-αIIbß3-myosin complex is formed as a primary axis to promote platelet contraction.


Subject(s)
Blood Platelets/metabolism , Clot Retraction/genetics , Factor XIII/metabolism , Fibrin/metabolism , Myosins/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Sphingomyelins/metabolism , Animals , Blood Coagulation/drug effects , Blood Coagulation/genetics , Blood Platelets/cytology , Blood Platelets/drug effects , Clot Retraction/drug effects , Factor XIII/genetics , Fibrin/genetics , Gene Expression , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Myosins/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Protein Transport , Signal Transduction , Thrombin/pharmacology , Transferases (Other Substituted Phosphate Groups)/deficiency , Transferases (Other Substituted Phosphate Groups)/genetics
10.
Mol Genet Metab ; 105(4): 615-20, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22305854

ABSTRACT

Recently, male subjects harboring the c.196G>C nucleotide change which leads to the E66Q enzyme having low α-galactosidase A (GLA) activity have been identified at an unexpectedly high frequency on Japanese and Korean screening for Fabry disease involving dry blood spots and plasma/serum samples. Individuals with the E66Q enzyme have been suspected to have the later-onset Fabry disease phenotype leading to renal and cardiac disease. However, there has been no convincing evidence for this. To determine whether c.196G>C (E66Q) is disease-causing or not, we performed biochemical, pathological and structural studies. It was predicted that the E66Q amino acid substitution causes a small conformational change on the molecular surface of GLA, which leads to instability of the enzyme protein. However, biochemical studies revealed that subjects harboring the E66Q enzyme exhibited relatively high residual enzyme activity in white blood cells, and that there was no accumulation of globotriaosylceramide in cultured fibroblasts or an increased level of plasma globotriaosylsphingosine in these subjects. An electron microscopic examination did not reveal any pathological changes specific to Fabry disease in biopsied skin tissues from a male subject with the E66Q enzyme. These results strongly suggest that the c.196G>C is not a pathogenic mutation but is a functional polymorphism.


Subject(s)
Fabry Disease/enzymology , Fabry Disease/genetics , Mutation/genetics , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism , Aged , Aged, 80 and over , Amino Acid Substitution , Asian People , Cells, Cultured , Child, Preschool , DNA Mutational Analysis , Fabry Disease/pathology , Fibroblasts/cytology , Fibroblasts/enzymology , Heterozygote , Humans , Immunoblotting , Male , Microscopy, Electron , Middle Aged , Models, Molecular , Phenotype , Real-Time Polymerase Chain Reaction , Skin/cytology , Skin/enzymology , Trihexosylceramides/blood , alpha-Galactosidase/chemistry
11.
Mol Med ; 18: 76-82, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22033676

ABSTRACT

To economically produce recombinant human α-galactosidase A (GLA) with a cell culture system that does not require bovine serum, we chose methylotrophic yeast cells with the OCH1 gene, which encodes α-1,6-mannosyltransferase, deleted and over-expressing the Mnn4p (MNN4) gene, which encodes a positive regulator of mannosylphosphate transferase, as a host cell line. The enzyme (yr-hGLA) produced with the gene-manipulated yeast cells has almost the same enzymological parameters as those of the recombinant human GLA produced with cultured human fibroblasts (agalsidase alfa), which is currently used for enzyme replacement therapy for Fabry disease. However, the basic structures of their sugar chains are quite different. yr-hGLA has a high content of phosphorylated N-glycans and is well incorporated into the kidneys, the main target organ in Fabry disease, where it cleaves the accumulated glycosphingolipids. A glycoprotein production system involving this gene-manipulated yeast cell line will be useful for the development of a new enzyme replacement therapy for Fabry disease.


Subject(s)
Fabry Disease/drug therapy , Kidney/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Yeasts/metabolism , alpha-Galactosidase/metabolism , alpha-Galactosidase/therapeutic use , Animals , Fabry Disease/metabolism , Female , Humans , Male , Mice , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics , Yeasts/genetics , alpha-Galactosidase/genetics , alpha-Galactosidase/pharmacokinetics
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.
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
14.
Biochem Biophys Res Commun ; 399(4): 716-20, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20692233

ABSTRACT

Fabry disease is a genetic disease caused by a deficiency of alpha-galactosidase A (GLA), which leads to systemic accumulation of glycolipids, predominantly globotriaosylceramide (Gb3). With the introduction and spread of enzyme replacement therapy (ERT) with recombinant GLAs for this disease, a useful biomarker for assessing the response to ERT is strongly required. We measured the tissue level of lyso-globotriaosylsphingosine (lyso-Gb3) in Fabry mice by means of high performance liquid chromatography, and compared it with the Gb3 level. The results revealed a marked increase in the lyso-Gb3 level in most tissues of Fabry mice, and which decreased after the administration of a recombinant GLA as in the case of Gb3, which is usually used as a biomarker of Fabry disease. The response was more impressive for lyso-Gb3 compared with for Gb3, especially in kidney tissues, in which a defect significantly influences the morbidity and mortality in patients with this disease. The plasma level of lyso-Gb3 also decreased after the injection of the enzyme, and it was well related to the degradation of tissue lyso-Gb3. Thus, lyso-Gb3 is expected to be a useful new biomarker for assessing the response to ERT for Fabry disease.


Subject(s)
Fabry Disease/therapy , Glycolipids/analysis , Sphingolipids/analysis , alpha-Galactosidase/administration & dosage , Animals , Biomarkers/analysis , Biomarkers/blood , Enzyme Replacement Therapy , Fabry Disease/pathology , Glycolipids/blood , Kidney/drug effects , Kidney/pathology , Mice , Prognosis , Recombinant Proteins/administration & dosage , Sphingolipids/blood
15.
Am J Hum Genet ; 85(5): 569-80, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19853240

ABSTRACT

A modified alpha-N-acetylgalactosaminidase (NAGA) with alpha-galactosidase A (GLA)-like substrate specificity was designed on the basis of structural studies and was produced in Chinese hamster ovary cells. The enzyme acquired the ability to catalyze the degradation of 4-methylumbelliferyl-alpha-D-galactopyranoside. It retained the original NAGA's stability in plasma and N-glycans containing many mannose 6-phosphate (M6P) residues, which are advantageous for uptake by cells via M6P receptors. There was no immunological cross-reactivity between the modified NAGA and GLA, and the modified NAGA did not react to serum from a patient with Fabry disease recurrently treated with a recombinant GLA. The enzyme cleaved globotriaosylceramide (Gb3) accumulated in cultured fibroblasts from a patient with Fabry disease. Furthermore, like recombinant GLA proteins presently used for enzyme replacement therapy (ERT) for Fabry disease, the enzyme intravenously injected into Fabry model mice prevented Gb3 storage in the liver, kidneys, and heart and improved the pathological changes in these organs. Because this modified NAGA is hardly expected to cause an allergic reaction in Fabry disease patients, it is highly promising as a new and safe enzyme for ERT for Fabry disease.


Subject(s)
Enzyme Replacement Therapy/methods , Fabry Disease/drug therapy , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/therapeutic use , Amino Acid Substitution , Animals , Binding Sites , CHO Cells , Catalysis , Cells, Cultured , Cricetinae , Cricetulus , Culture Media, Conditioned/chemistry , DNA, Complementary/metabolism , Disease Models, Animal , Drug Stability , Fabry Disease/enzymology , Fabry Disease/metabolism , Fibroblasts/drug effects , Fluorescent Dyes/metabolism , Galactosides/metabolism , Genetic Vectors , Humans , Hydrogen-Ion Concentration , Hymecromone/analogs & derivatives , Hymecromone/metabolism , Immunohistochemistry , Kidney/drug effects , Kidney/pathology , Kidney/ultrastructure , Liver/drug effects , Liver/pathology , Liver/ultrastructure , Mice , Mice, Knockout , Models, Molecular , Molecular Weight , Myocardium/pathology , Myocardium/ultrastructure , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use , Retroviridae/genetics , Transfection , Trihexosylceramides/metabolism , alpha-N-Acetylgalactosaminidase/genetics , alpha-N-Acetylgalactosaminidase/isolation & purification
16.
Mol Genet Metab ; 96(4): 233-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19181556

ABSTRACT

Enzyme enhancement therapy (EET) for Fabry disease involving imino sugars has been developed and attracted interest. It is thought that imino sugars act as pharmacological chaperones for wild-type and mutant alpha-galactosidases (GLAs) in cells, but the mechanisms underlying the molecular interactions between the imino sugars and the enzyme have not been clarified yet. We examined various kinds of imino sugars and found that galactostatin bisulfite (GBS) inhibited GLA in vitro and increased the enzyme activity in cultured Fabry fibroblasts as in the case of 1-deoxygalactonojirimycin (DGJ). Then, we analyzed the molecular interactions between the imino sugars and recombinant human GLA by means of isothermal titration calorimetry and surface plasmon resonance biosensor assays, and first determined the thermodynamic and binding-kinetics parameters of imino sugar and GLA complex formation. The results revealed that DGJ bound to the enzyme more strongly than GBS, the binding of DGJ to the enzyme protein being enthalpy-driven. In the case of GBS, the reaction was mainly enthalpy-driven, but there was a possibility that entropy-driven factors were involved in the binding. Structural analysis in silico revealed that both the chemicals fit into the active-site pocket and undergo hydrogen bonding with residues comprising the active-site pocket including the catalytic ones. The side chain of GBS was oriented towards the entrance of the active-site pocket, and thus it could be in contact with residues comprising the wall of the active-site pocket. Thermodynamic, kinetic and structural studies should provide us with a lot of information for improving EET for Fabry disease.


Subject(s)
Fabry Disease/enzymology , Imino Sugars/pharmacology , alpha-Galactosidase/antagonists & inhibitors , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/pharmacology , Animals , CHO Cells , Catalytic Domain , Cells, Cultured , Cricetinae , Cricetulus , Fabry Disease/pathology , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Galactosamine/analogs & derivatives , Galactosamine/chemistry , Galactosamine/pharmacology , Humans , Imino Sugars/chemistry , Kinetics , Models, Molecular , Thermodynamics , alpha-Galactosidase/metabolism
17.
Biol Pharm Bull ; 31(9): 1691-5, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18758061

ABSTRACT

To examine the uptake of a recombinant human alpha-L-iduronidase (laronidase) by cultured fibroblasts from a patient with mucopolysaccharidosis I (MPS I) and its effect on the cleavage of accumulated substrates, we performed enzymological, Western blotting, immunocytochemical and morphological studies. Laronidase was incorporated into the MPS I cells dose-dependently mainly via mannose 6-phosphate (M6P) receptors. Then the incorporated enzyme was transported to lysosomes and processed to the mature form, the pathological changes of the cells being improved. Furthermore, we compared the uptake of laronidase by cultured mouse osteoblasts with that by cultured mouse fibroblasts. The enzyme was incorporated into the cultured mouse osteoblasts mainly via M6P receptors, although mannose (Man) receptors were partially involved in the uptake of the enzyme, as in the cultured fibroblasts. But the uptake by the former was apparently lower than that by the latter. The administration of a high dose of the enzyme or development of a recombinant alpha-L-iduronidase containing many M6P residues is required for further improvement of enzyme replacement therapy for skeletal disorders caused by MPS I.


Subject(s)
Fibroblasts/metabolism , Iduronidase/metabolism , Osteoblasts/metabolism , Animals , Cells, Cultured , Fibroblasts/ultrastructure , Humans , Kinetics , Mice , Mucopolysaccharidosis I/metabolism , Osteoblasts/ultrastructure , Receptor, IGF Type 2/metabolism , Recombinant Proteins/metabolism
18.
Clin Chim Acta ; 391(1-2): 68-73, 2008 May.
Article in English | MEDLINE | ID: mdl-18328816

ABSTRACT

BACKGROUND: Recently, enzyme enhancement therapy (EET) for Pompe disease involving imino sugars, which act as potential inhibitors of acid alpha-glucosidases in vitro, to improve the stability and/or transportation of mutant acid alpha-glucosidases in cells was studied and attracted interest. However, the mechanism underlying the molecular interaction between the imino sugars and the enzyme has not been clarified yet. METHODS: We examined the inhibitory and binding effects of four imino sugars on a recombinant human acid alpha-glucosidase, alglucosidase alfa, by means of inhibition assaying and isothermal titration calorimetry (ITC). Furthermore, we built structural models of complexes of the catalytic domain of the enzyme with the imino sugars bound to its active site by homology modeling, and examined the molecular interaction between them. RESULTS: All of the imino sugars examined exhibited a competitive inhibitory action against the enzyme, 1-deoxynojirimycin (DNJ) exhibiting the strongest action among them. ITC revealed that one compound molecule binds to one enzyme molecule and that DNJ most strongly binds to the enzyme among them. Structural analysis revealed that the active site of the enzyme is almost completely occupied by DNJ. CONCLUSION: These biochemical and structural analyses increased our understanding of the molecular interaction between a human acid alpha-glucosidase and imino sugars.


Subject(s)
Glycogen Storage Disease Type II/enzymology , Imino Sugars/metabolism , alpha-Glucosidases/metabolism , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/metabolism , 1-Deoxynojirimycin/pharmacology , Binding Sites , Catalytic Domain , Drug Interactions , Glycoside Hydrolase Inhibitors , Humans , Imino Sugars/chemistry , Imino Sugars/pharmacology , Models, Molecular , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Thermodynamics
19.
Neurosignals ; 16(2-3): 226-34, 2008.
Article in English | MEDLINE | ID: mdl-18253060

ABSTRACT

To characterize biomarkers in neural tumors, we analyzed the acidic lipid fractions of 13 neural tumor cell lines using enzyme-linked immunoabsorbent assay (ELISA) and high-performance thin-layer chromatography (HPTLC) immunostaining. Sulfated glucuronosyl glycosphingolipids (SGGLs) are cell surface molecules that are endowed with the Human Natural Killer-1 (HNK-1) carbohydrate epitope. These glycosphingolipids (GSLs) were expressed in all cell lines with concentrations ranging from 210 to 330 ng per 2 x 10(6) cells. Sulfoglucuronosyl paragloboside (SGPG) was the prominent species with lesser amounts of sulfoglucuronosyl lactosaminyl paragloboside (SGLPG) in these tumor cell lines as assessed by quantitative HPTLC immunostaining. Among the gangliosides surveyed, GD3 and 9-O-acetylated GD3 (OAc-GD3) were expressed in all tumor cell lines. In contrast, fucosyl-GM1 was not found to restrict to small cell lung carcinoma cells. In addition, we have analyzed serum antibody titers against SGPG, GD3, and OAc-GD3 in patients with neural tumors by ELISA and HPTLC immunostaining. All sera had high titers of antibodies of the IgM isotype against SGPG (titers over 1:3,200), especially in tumors such as meningiomas, germinomas, orbital tumors, glioblastomas, medulloblastomas, and subependymomas. Serum in a patient with subependymomas also had a high anti-SGGL antibody titer of the IgG and IgA types (titers over 12,800). The titer of anti-GD3 antibody was also elevated in patients with subependymomas and medulloblastomas; the latter cases also had a high titer of antibody against OAc-GD3. Our data indicate that certain GSL antigens, especially SGGLs, GD3, and OAc-GD3, are expressed in neural tumor cells and may be considered as tumor-associated antigens that represent important biomarkers for neural tumors. Furthermore, antibody titers in sera of patients with these tumors may be of diagnostic value for monitoring the presence of tumor cells and tumor progression.


Subject(s)
Antibodies, Neoplasm/blood , Antigens, Neoplasm/blood , Glycosphingolipids/physiology , Nervous System Neoplasms/immunology , Animals , Antigens, Neoplasm/immunology , Biomarkers/blood , Cattle , Cell Line, Tumor , Glycosphingolipids/immunology , Humans , Nervous System Neoplasms/blood
20.
J Hum Genet ; 52(12): 1018-1025, 2007.
Article in English | MEDLINE | ID: mdl-17965825

ABSTRACT

Peripheral neuropathy is one of the important manifestations of Fabry disease. Enzyme replacement therapy with presently available recombinant alpha-galactosidases does not always improve the Fabry neuropathy. But the reason has not been determined yet. We established a Schwann cell line from Fabry mice, characterized it, and then examined the uptake of alpha-galactosidase by cells and its effect on the degradation of accumulated substrate. The cells exhibited a distinct Schwann cell morphology and biochemical phenotype (alpha-Galactosidase activity was deficient, and numerous cytoplasmic inclusion bodies were present in the cells). A recombinant alpha-galactosidase added to the culture medium was incorporated into the cultured Fabry Schwann cells dose dependently. But the increase in cell-associated enzyme activity was less than that in the cases of human and mouse Fabry fibroblasts. The administration of a high dose of the enzyme improved the pathological changes in cells, although a low dose of it did not. Cellular uptake of the enzyme was strongly inhibited in the presence of mannose 6-phosphate. This suggests that the enzyme is incorporated via cation-independent mannose 6-phosphate receptors in Schwann cells. The low expression of cation-independent mannose 6-phosphate receptors in Schwann cells must be one of the reasons their uptake of the present enzymes was low. The administration of a high dose of the enzyme or the development of an enzyme containing many mannose 6-phosphate residues is required to improve Fabry neuropathy.


Subject(s)
Fabry Disease/drug therapy , Schwann Cells/metabolism , alpha-Galactosidase/pharmacokinetics , Animals , Biological Transport , Dose-Response Relationship, Drug , Fabry Disease/enzymology , Fabry Disease/metabolism , Humans , Mice , Mice, Inbred C57BL , Receptor, IGF Type 2 , Recombinant Proteins , Schwann Cells/drug effects , alpha-Galactosidase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...