Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters











Publication year range
1.
Chembiochem ; 25(15): e202400081, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38830828

ABSTRACT

Mucopolysaccharidosis type IIIB (MPS IIIB) is an autosomal inherited disease caused by mutations in gene encoding the lysosomal enzyme N-acetyl-alpha-glucosaminidase (NAGLU). These mutations result in reduced NAGLU activity, preventing it from catalyzing the hydrolysis of the glycosaminoglycan heparan sulfate (HS). There are currently no approved treatments for MPS IIIB. A novel approach in the treatment of lysosomal storage diseases is the use of pharmacological chaperones (PC). In this study, we used a drug repurposing approach to identify and characterize novel potential PCs for NAGLU enzyme. We modeled the interaction of natural and artificial substrates within the active cavity of NAGLU (orthosteric site) and predicted potential allosteric sites. We performed a virtual screening for both the orthosteric and the predicted allosteric site against a curated database of human tested molecules. Considering the binding affinity and predicted blood-brain barrier permeability and gastrointestinal absorption, we selected atovaquone and piperaquine as orthosteric and allosteric PCs. The PCs were evaluated by their capacity to bind NAGLU and the ability to restore the enzymatic activity in human MPS IIIB fibroblasts These results represent novel PCs described for MPS IIIB and demonstrate the potential to develop novel therapeutic alternatives for this and other protein deficiency diseases.


Subject(s)
Acetylglucosaminidase , Mucopolysaccharidosis III , Humans , Mucopolysaccharidosis III/drug therapy , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/pathology , Acetylglucosaminidase/metabolism , Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/chemistry , Acetylglucosaminidase/genetics , Allosteric Site/drug effects , Allosteric Regulation/drug effects
2.
Org Biomol Chem ; 20(3): 619-629, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34940771

ABSTRACT

We report the synthesis of seven-membered iminosugars derived from a 3S-acetamido-4R,5R,6S-trihydroxyazepane scaffold and their evaluation as inhibitors of functionally related exo-N-acetylhexosaminidases including human O-GlcNAcase (OGA), human lysosomal ß-hexosaminidase (HexAB), and Escherichia coli NagZ. Capitalizing on the flexibility of azepanes and the active site tolerances of hexosaminidases, we explore the effects of epimerization of stereocenters at C-3, C-5 and C-6 and C-alkylation at the C-2 or C-7 positions. Accordingly, epimerization at C-6 (L-ido) and at C-5 (D-galacto) led to selective HexAB inhibitors whereas introduction of a propyl group at C-7 on the C-3 epimer furnished a potent NagZ inhibitor.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Imino Sugars/pharmacology , beta-N-Acetylhexosaminidases/antagonists & inhibitors , Acetylglucosaminidase/metabolism , Alkylation , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Escherichia coli/enzymology , Humans , Imino Sugars/chemical synthesis , Imino Sugars/chemistry , Molecular Conformation , beta-N-Acetylhexosaminidases/metabolism
3.
Pharmacol Res ; 165: 105467, 2021 03.
Article in English | MEDLINE | ID: mdl-33515704

ABSTRACT

Cardiovascular diseases (CVDs) are the leading cause of death globally. While the major focus of pharmacological and non-pharmacological interventions has been on targeting disease pathophysiology and limiting predisposing factors, our understanding of the cellular and molecular mechanisms underlying the pathogenesis of CVDs remains incomplete. One mechanism that has recently emerged is protein O-GlcNAcylation. This is a dynamic, site-specific reversible post-translational modification of serine and threonine residues on target proteins and is controlled by two enzymes: O-linked ß-N-acetylglucosamine transferase (OGT) and O-linked ß-N-acetylglucosaminidase (OGA). Protein O-GlcNAcylation alters the cellular functions of these target proteins which play vital roles in pathways that modulate vascular homeostasis and cardiac function. Through this review, we aim to give insights on the role of protein O-GlcNAcylation in cardiovascular diseases and identify potential therapeutic targets in this pathway for development of more effective medicines to improve patient outcomes.


Subject(s)
Cardiovascular Agents/administration & dosage , Cardiovascular Diseases/drug therapy , Drug Delivery Systems/methods , Enzyme Inhibitors/administration & dosage , Protein Processing, Post-Translational/drug effects , Acetylglucosamine/antagonists & inhibitors , Acetylglucosamine/metabolism , Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/metabolism , Acylation/drug effects , Acylation/physiology , Animals , Antigens, Neoplasm/metabolism , Cardiovascular Diseases/metabolism , Glycosylation/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Humans , Hyaluronoglucosaminidase/antagonists & inhibitors , Hyaluronoglucosaminidase/metabolism , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational/physiology , beta-N-Acetylhexosaminidases/antagonists & inhibitors , beta-N-Acetylhexosaminidases/metabolism
4.
Carbohydr Res ; 499: 108201, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33243428

ABSTRACT

We investigated the inhibition kinetics of VhGlcNAcase, a GH20 exo-ß-N-acetylglucosaminidase (GlcNAcase) from the marine bacterium Vibrio campbellii (formerly V. harveyi) ATCC BAA-1116, using TMG-chitotriomycin, a natural enzyme inhibitor specific for GH20 GlcNAcases from chitin-processing organisms, with p-nitrophenyl N-acetyl-ß-d-glucosaminide (pNP-GlcNAc) as the substrate. TMG-chitotriomycin inhibited VhGlcNAcase with an IC50 of 3.0 ± 0.7 µM. Using Dixon plots, the inhibition kinetics indicated that TMG-chitotriomycin is a competitive inhibitor, with an inhibition constant Ki of 2.2 ± 0.3 µM. Isothermal titration calorimetry experiments provided the thermodynamic parameters for the binding of TMG-chitotriomycin to VhGlcNAcase and revealed that binding was driven by both favorable enthalpy and entropy changes (ΔH° = -2.5 ± 0.1 kcal/mol and -TΔS° = -5.8 ± 0.3 kcal/mol), resulting in a free energy change, ΔG°, of -8.2 ± 0.2 kcal/mol. Dissection of the entropic term showed that a favorable solvation entropy change (-TΔSsolv° = -16 ± 2 kcal/mol) is the main contributor to the entropic term.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Sugar Alcohols/pharmacology , Thermodynamics , Vibrio/enzymology , Acetylglucosaminidase/metabolism , Carbohydrate Conformation , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Kinetics , Sugar Alcohols/chemical synthesis , Sugar Alcohols/chemistry
5.
Org Biomol Chem ; 18(29): 5639-5651, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32724966

ABSTRACT

Synthesis of novel homoazanucleosides and their peptidyl analogs as hybrid molecules comprised of amino acids, an iminosugar and natural nucleobases is reported for the first time. A pluripotent amino-substituted chiral polyhydroxypyrrolidine, possessing orthogonally different functional groups on either arm of the pyrrolidine ring, served as an ideal substrate for the synthesis of the proposed peptidyl homoazanucleosides. The acid sensitive primary benzyloxy group, on one arm of the pyrrolidine ring, after selective deprotection, was utilized for the introduction of nucleobases to obtain the homoazanucleosides. The amino group on the other side offered the opportunity to be coupled with amino acids to deliver the desired peptidyl homoazanucleosides. Glycosidase inhibition studies revealed that the acetamido derivatives of homoazanucleosides were found to be sub-millimolar inhibitors of ß-N-acetyl-glucosaminidase.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Aza Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Nucleosides/pharmacology , Peptides/pharmacology , Acetylglucosaminidase/metabolism , Aza Compounds/chemical synthesis , Aza Compounds/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Fabaceae/enzymology , Models, Molecular , Molecular Conformation , Nucleosides/chemical synthesis , Nucleosides/chemistry , Peptides/chemical synthesis , Peptides/chemistry
6.
FEBS J ; 287(22): 4982-4995, 2020 11.
Article in English | MEDLINE | ID: mdl-32145141

ABSTRACT

Vibrio spp. play a vital role in the recycling of chitin in oceans, but several Vibrio strains are highly infectious to aquatic animals and humans. These bacteria require chitin for growth; thus, potent inhibitors of chitin-degrading enzymes could serve as candidate drugs against Vibrio infections. This study examined NAG-thiazoline (NGT)-mediated inhibition of a recombinantly expressed GH20 ß-N-acetylglucosaminidase, namely VhGlcNAcase from Vibrio campbellii (formerly V. harveyi) ATCC BAA-1116. NGT strongly inhibited VhGlcNAcase with an IC50 of 11.9 ± 1.0 µm and Ki 62 ± 3 µm, respectively. NGT was also found to completely inhibit the growth of V. campbellii strain 650 with an minimal inhibitory concentration value of 0.5 µm. ITC data analysis showed direct binding of NGT to VhGlcNAcase with a Kd of 32 ± 1.2 µm. The observed ΔG°binding of -7.56 kcal·mol-1 is the result of a large negative enthalpy change and a small positive entropic compensation, suggesting that NGT binding is enthalpy-driven. The structural complex shows that NGT fully occupies the substrate-binding pocket of VhGlcNAcase and makes an exclusive hydrogen bond network, as well as hydrophobic interactions with the conserved residues around the -1 subsite. Our results strongly suggest that NGT could serve as an excellent scaffold for further development of antimicrobial agents against Vibrio infections. DATABASE: Structural data are available in PDB database under the accession number 6K35.


Subject(s)
Acetylglucosamine/analogs & derivatives , Acetylglucosaminidase/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Thiazoles/pharmacology , Vibrio/enzymology , Acetylglucosamine/chemistry , Acetylglucosamine/pharmacology , Acetylglucosaminidase/chemistry , Acetylglucosaminidase/metabolism , Animals , Bacterial Infections/microbiology , Bacterial Infections/prevention & control , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Kinetics , Models, Molecular , Oligosaccharides/metabolism , Protein Domains , Substrate Specificity , Thermodynamics , Thiazoles/chemistry , Vibrio/drug effects , Vibrio/genetics , Vibrio/growth & development
7.
Int J Biol Macromol ; 142: 503-512, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31593714

ABSTRACT

ß-N-acetylglucosaminidases (GlcNAcases) play a crucial role in the metabolism of glycan-conjugated proteins/lipids in humans. Elevated levels of serum GlcNAcases have been associated with certain types of cancer, and GlcNAcases therefore serve as drug targets. Here, we employed virtual screening to identify two novel GlcNAcase inhibitors from the National Cancer Institute (NCI) Drug Library using a bacterial GH-20 GlcNAcase (VhGlcNAcase) as a search model. NSC73735 was shown to be most potent with IC50 of 12.7 ±â€¯1.2 µM, agreeing with Kd of 0.94 ±â€¯0.2 µM obtained by ITC. Molecular docking refinement indicated that Trp582 the key residue that interacted with all the inhibitor molecules. Docking NSC7373 into the active site of human O-GlcNAcase (hOGA) yielded reasonably good fit with the estimated Kd of 44.7 µM, indicating its possibility to be a true binding partner. NSC73735 was shown to significantly suppress both cell growth and GlcNAcase activity of five cancer cell lines (U937, THP-1, MCF-7, HepG2 and PC-3) that express endogenous GlcNAcases. The cell cytotoxicity assay indicated the inherent effects of the lead compound on GlcNAcase expression with cancer cell proliferation, and therefore this novel GlcNAcase inhibitor may serve as a virtuous candidate for further development of highly potent anti-tumor agents.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Acetylglucosaminidase/chemistry , Cell Line, Tumor , Drug Evaluation, Preclinical , Humans , Protein Conformation , User-Computer Interface
8.
Bioprocess Biosyst Eng ; 42(4): 611-619, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30673842

ABSTRACT

ß-N-Acetylglucosaminidases (NAGase) can remove N-acetylglucosamine (GlcNAc) from the non-reducing end of chitin or chitosan. GlcNAc has many important physiological functions in organism, which can be used for the treatment of rheumatoid arthritis clinically and be used as food antioxidant, infant food additive and diabetic sweetener. Thus, it is very important to develop genetic-engineering strains with high-yield NAGase to hydrolyze chitin into GlcNAc. Here, the NAGase gene of Bacillus subtilis 168 (BsnagZ) was synthesized according to the codon bias of Pichia pastoris and expressed in P. pastoris. The expression level of BsNagZ in P. pastoris increased over the induced time and the highest activity reached 0.76 U/mL at the 7th day. The recombinant BsNagZ was purified for characterization. The optimal temperature and pH are 60 °C and 6.0, respectively. It can both keep over 80% activities after pre-incubation at 55 °C for one hour and at 4 °C for 12 h from pH 4.5 to 10.0. To further improve the expression level of BsNagZ, a recombinant strain with four copy BsnagZs was screened using a high concentration of zeocin. The highest BsNagZ activity reached 3.2 U/mL at the 12th day, which was fourfold higher than that of single-copy strain. Combined with commercial chitinase CtnSg, GlcNAc can be produced by recombinant BsNagZ when used colloidal chitin as the substrate. Our study highlights that the NAGase was first successfully expressed in P. pastoris and GlcNAc can be produced via NAGase hydrolyzing the colloidal chitin.


Subject(s)
Acetylglucosamine/chemistry , Acetylglucosaminidase , Bacillus subtilis/genetics , Bacterial Proteins , Gene Expression , Pichia , Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/chemistry , Acetylglucosaminidase/genetics , Bacillus subtilis/enzymology , Bacterial Proteins/biosynthesis , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
9.
Chem Commun (Camb) ; 54(75): 10630-10633, 2018 Sep 25.
Article in English | MEDLINE | ID: mdl-30178799
10.
SAR QSAR Environ Res ; 29(9): 647-660, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30160524

ABSTRACT

A structure-based approach is applied for the development of inhibitors of bacterial N-acetyglucosaminidase (autolysin). Autolysins are enzymes involved in the degradation of peptidoglycan and therefore participate in bacterial cell growth and different lysis phenomena. Several studies indicate that by the inhibition of autolysins, and consequently of bacterial cell division, antibacterial activity can be obtained, thus paving the road to a novel group of therapeutics against human pathogens. As crystal structures of the autolysin E (AtlE)-ligand complexes were obtained in our laboratories, fragment-based virtual screening was the method of choice for the initial studies. Fragment libraries from various databases were merged to increase the number of compounds for the virtual screening. Twenty-four commercially available virtual hits were selected and subjected to quantitative analysis of binding interactions using the surface plasmon resonance technique. Twelve fragments showed fragment-AtlE interactions. For F1, the top hit of the virtual screening, a KD of 228 µM was determined, while other fragments displayed non-stoichiometric binding. Blind docking of potential binders uncovers three possible allosteric sites. Ligands of N-acetyglucosaminidase identified in our study represent valuable information for the further development of AtlE inhibitors, which could in future represent antibacterial agents acting by a novel mode of action.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical , N-Acetylmuramoyl-L-alanine Amidase/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Models, Molecular , Molecular Docking Simulation , Small Molecule Libraries
11.
Int J Biol Macromol ; 115: 1165-1173, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29730005

ABSTRACT

Exo-ß-N-acetylglucosaminidases (GlcNAcases) are hydrolytic enzymes involved in the metabolism of chitin in bacteria and in eukaryotic glycosphingolipid metabolism, with genetic defects in human GlcNAcases (HexA and HexB) resulting in Tay-Sachs and Sandhoff diseases, respectively. Here, we determined the effects of three known inhibitors of exo-ß-N-acetylglucosaminidases (PUGNAc, NHAcCAS and NHAcDNJ) on a GH20 exo-ß-N-GlcNAcase (VhGlcNAcase) from the pathogenic bacterium Vibrio harveyi, in dose-response experiments. The inhibitors were shown to modify the kinetic parameters (both Km and kcat), yielding significant decreases in the overall efficiency of the enzyme in hydrolyzing the natural substrate diNAG. Molecular interactions between the inhibitors and the enzyme were investigated by isothermal calorimetry (ITC), and were confirmed using molecular docking. VhGlcNAcase was strongly inhibited by these compounds, with PUGNAc having the lowest IC50 value, of 1.2 µM. Molecular docking suggested that the inhibitors mimicked reaction intermediates, with enzyme-inhibitor interactions being similar to those of the enzyme with diNAG. The equilibrium dissociation constants (Kd) obtained from ITC were 0.19 µM for PUGNAc, 12.9 µM for NHAcCAS and 25.6 µM for NHAcDNJ, confirming that PUGNAc was the most potent inhibitor. The ITC data indicated that the binding of the enzyme to the inhibitors was driven by enthalpy. The negative heat capacity change (ΔCp) of -0.34 ±â€¯0.05 kcal·mol-1·K-1 indicates that hydrophobic interactions make a substantial contribution to the molecular interactions between PUGNAc and the enzyme. Our results suggest that PUGNAc is a highly potent inhibitor, and suggest its usefulness as a scaffold for potential drugs targeting GlcNAcase-related metabolic diseases.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Vibrio/enzymology , Acetylglucosaminidase/chemistry , Kinetics , Molecular Docking Simulation , Protein Conformation , Thermodynamics
12.
Anim Reprod Sci ; 195: 96-101, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29807827

ABSTRACT

This study investigated how the inhibition of certain enzymes present in ide sperm influences sperm motility and the percentage of fertilised eggs. The enzymes studied were acid phosphatase (AcP), ß-N-acetylglucosaminidase (ß-NAGase) and lactate dehydrogenase (LDH). None of the inhibitors affected ide sperm motility parameters. The addition of gossypol (a LDH inhibitor) caused a considerable increase in the percentage of fertilised eggs (92-95% compared to 63% in the control). The inhibition of AcP caused a considerable decrease in fertility rate - at the highest inhibitor dose, the percentage of fertilised eggs decreased to 26%. A similar effect was seen after the addition of acetamide (a ß-NAGase inhibitor), but in this case the highest dose caused complete inhibition of fertilisation. The results presented here indicate the importance of AcP and ß-NAGase in the process of ide fertilisation.


Subject(s)
Acetylglucosaminidase/metabolism , Cyprinidae/physiology , Enzyme Inhibitors/pharmacology , L-Lactate Dehydrogenase/metabolism , Spermatozoa/enzymology , Zygote/physiology , Acetamides/pharmacology , Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/genetics , Animals , Fertility , Gossypol/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/genetics , Male , Molybdenum/pharmacology , Semen
13.
ACS Infect Dis ; 3(6): 421-427, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28448118

ABSTRACT

N-Acetylglucosaminidases (GlcNAcases) play an important role in the remodeling and recycling of bacterial peptidoglycan by degrading the polysaccharide backbone. Genetic deletions of autolysins can impair cell division and growth, suggesting an opportunity for using small molecule autolysin inhibitors both as tools for studying the chemical biology of autolysins and also as antibacterial agents. We report here the synthesis and evaluation of a panel of diamides that inhibit the growth of Bacillus subtilis. Two compounds, fgkc (21) and fgka (5), were found to be potent inhibitors (MIC 3.8 ± 1.0 and 21.3 ± 0.1 µM, respectively). These compounds inhibit the B. subtilis family 73 glycosyl hydrolase LytG, an exo GlcNAcase. Phenotypic analysis of fgkc (21)-treated cells demonstrates a propensity for cells to form linked chains, suggesting impaired cell growth and division.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Azo Compounds/chemical synthesis , Bacillus subtilis/drug effects , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , N-Glycosyl Hydrolases/antagonists & inhibitors , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Anti-Bacterial Agents/pharmacology , Azo Compounds/pharmacology , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Bacillus subtilis/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Division/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression , Hydrolysis , Microbial Sensitivity Tests , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Peptidoglycan/chemistry , Structure-Activity Relationship
14.
J Cereb Blood Flow Metab ; 37(8): 2938-2951, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27864466

ABSTRACT

Inflammatory responses are accountable for secondary injury induced by acute ischemic stroke (AIS). Previous studies indicated that O-GlcNAc modification (O-GlcNAcylation) is involved in the pathology of AIS, and increase of O-GlcNAcylation by glucosamine attenuated the brain damage after ischemia/reperfusion. Inhibition of ß-N-acetylglucosaminidase (OGA) with thiamet G (TMG) is an alternative option for accumulating O-GlcNAcylated proteins. In this study, we investigate the neuroprotective effect of TMG in a mouse model of experimental stroke. Our results indicate that TMG administration either before or after middle cerebral artery occlusion (MCAO) surgery dramatically reduced infarct volume compared with that in untreated controls. TMG treatment ameliorated the neurological deficits and improved clinical outcomes in neurobehavioral tests by modulating the expression of pro-inflammatory and anti-inflammatory cytokines. Additionally, TMG administration reduced the number of Iba1+ cells in MCAO mice, decreased expression of the M1 markers, and increased expression of the M2 markers in vivo. In vitro, M1 polarization of BV2 cells was inhibited by TMG treatment. Moreover, TMG decreased the expression of iNOS and COX2 mainly by suppressing NF-κB p65 signaling. These results suggest that TMG exerts a neuroprotective effect and could be useful as an anti-inflammatory agent for ischemic stroke therapy.


Subject(s)
Macrophages/drug effects , Microglia/drug effects , Neuroprotective Agents/therapeutic use , Pyrans/therapeutic use , Stroke/drug therapy , Thiazoles/therapeutic use , Transcription Factor RelA/metabolism , Acetylglucosamine/metabolism , Acetylglucosaminidase/antagonists & inhibitors , Animals , Biomarkers/metabolism , Disease Models, Animal , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , N-Acetylglucosaminyltransferases/metabolism , Neuroprotective Agents/administration & dosage , Pyrans/administration & dosage , Signal Transduction/drug effects , Stroke/metabolism , Stroke/pathology , Thiazoles/administration & dosage
15.
J Biochem ; 159(2): 191-200, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26330565

ABSTRACT

Vibrio harveyi is a bioluminescent marine bacterium that utilizes chitin as its sole source of energy. In the course of chitin degradation, the bacterium primarily secretes an endochitinase A (VhChiA) to hydrolyze chitin, generating chitooligosaccharide fragments that are readily transported into the cell and broken down to GlcNAc monomers by an exo ß-N-acetylglucosaminidase (VhGlcNAcase). Here we report that sodium salts, especially sodium azide, inhibit two classes of these chitin-degrading enzymes (VhChiA and VhGlcNAcase) with distinct modes of action. Kinetic analysis of the enzymatic hydrolysis of pNP-glycoside substrates reveals that sodium azide inhibition of VhChiA has a mixed-type mode, but that it inhibits VhGlcNAcase competitively. We propose that azide anions inhibit chitinase activity by acting as strong nucleophiles that attack Cγ of the catalytic Glu or Cß of the neighbouring Asp residues. Azide anions may bind not only to the catalytic centre, but also to the other subsites in the substrate-binding cleft of VhChiA. In contrast, azide anions may merely occupy the small-binding pocket of VhGlcNAcase, thereby blocking the accessibility of its active site by short-chain substrates.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Azides/pharmacology , Bacterial Proteins/antagonists & inhibitors , Chitinases/antagonists & inhibitors , Vibrio/enzymology , Acetylglucosamine/metabolism , Acetylglucosaminidase/isolation & purification , Anions/pharmacology , Bacterial Proteins/isolation & purification , Catalysis , Catalytic Domain/drug effects , Chitin/analogs & derivatives , Chitin/metabolism , Chitinases/isolation & purification , Chitosan , Hydrolysis , Kinetics , Oligosaccharides , Protein Binding
16.
Org Biomol Chem ; 13(30): 8196-204, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26108188

ABSTRACT

The reversible O-linked attachment of single ß-D-N-acetylglucosamine (GlcNAc) moieties to serine/threonine residues in target proteins is a frequently occurring post-translational modification affecting the functionality of many cellular systems. In this report we present experimental evidence suggesting that the epidermal growth factor receptor (EGFR) is subjected to O-GlcNAcylation in human carcinoma epidermoid A431 cells and human lung carcinoma A549 cells. However, no signal was detected in human cervix adenocarcinoma HeLa cells or in mouse EGFR-T17 fibroblasts ectopically expressing the human EGFR. We detected a positive O-GlcNAcylation signal in the immunoprecipitated EGFR by Western blotting using two distinct specific anti-O-GlcNAc antibodies even after N-deglycosylation of the receptor using peptide-N-glycosidase F (PNGase F). Conversely, the presence of EGFR was detected by Western blotting using an anti-EGFR antibody in the immunocomplex of O-GlcNAcylated proteins immunoprecipitated with an anti-O-GlcNAc antibody. These signals were enhanced when the O-linked ß-N-acetylglucosaminidase (OGA) inhibitor Thiamet G was added to prevent the deglycosylation of the GlcNAc moiety(ies). Moreover, we also detected a positive signal in the immunoprecipitated and N-deglycosylated EGFR using PNGase F, and tunicamycin when the cells were metabolically labeled with azido-GlcNAc (GlcNAz), biotinylated and probed with a streptavidin-labeled peroxidase. Finally, EGFR and O-linked ß-N-acetylglucosamine transferase (OGT) co-immunoprecipitate, and incubation of the immunoprecipitated EGFR with the immunoprecipitated OGT in the presence of uridine 5'-diphospho-N-acetylglucosamine (UDP-GlcNAc) resulted in a significant enhancement of the EGFR O-GlcNAcylation signal as detected by Western blotting using an anti-O-GlcNAc antibody. We conclude that the human EGFR is subjected to O-GlcNAcylation in the A431 and A549 tumor cell lines.


Subject(s)
Acetylglucosamine/metabolism , ErbB Receptors/metabolism , Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/metabolism , Animals , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Glycosylation/drug effects , Humans , Immunoblotting , Immunoprecipitation , Mice , Staining and Labeling
17.
J Biosci Bioeng ; 119(2): 131-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25193875

ABSTRACT

The baculovirus-silkworm recombinant protein expression system is an excellent method for achieving high-level expression and post-translational modifications, especially glycosylation. However, the presence of paucimannosidic-type N-glycan in glycoproteins restricts their clinical use. Paucimannosidic-type N-glycan is produced by insect-specific membrane-binding-type ß-N-acetylglucosaminidase (GlcNAcase). In the silkworm, BmGlcNAcase1, BmGlcNAcase2, and BmFDL are membrane-binding-type GlcNAcases. We investigated the localization of these GlcNAcases and found that BmFDL and BmGlcNAcase2 were mainly located in the fat body and hemolymph, respectively. The fat body is the main tissue of recombinant protein expression by baculovirus, and many glycoproteins are secreted into the hemolymph. These results suggest that inhibition of BmFDL and BmGlcNAcase2 could increase GlcNAc-type N-glycan levels. We therefore injected a GlcNAcase inhibitor into silkworms to investigate changes in the N-glycan structure of the glycoprotein expressed by baculovirus; modest levels of GlcNAc-type N-glycan were observed (0.8% of total N-glycan). Next, we generated a transgenic silkworm in which RNA interference (RNAi) reduced the BmFDL transcript level and enzyme activity to 25% and 50%, respectively, of that of the control silkworm. The proportion of GlcNAc-type N-glycan increased to 4.3% in the RNAi-transgenic silkworm. We conclude that the structure of N-glycan can be changed by inhibiting the GlcNAcases in silkworm.


Subject(s)
Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/metabolism , Bombyx/enzymology , Glycoproteins/biosynthesis , Glycoproteins/chemistry , Polysaccharides/chemistry , Protein Processing, Post-Translational , Acetylglucosaminidase/isolation & purification , Animals , Animals, Genetically Modified , Baculoviridae/genetics , Bombyx/genetics , Bombyx/metabolism , Fat Body/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Hemolymph/metabolism , Polysaccharides/metabolism , Protein Transport , RNA Interference , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
FEBS Lett ; 589(1): 110-6, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25436416

ABSTRACT

NAG-thiazoline (NGT) and its derivatives are well-known inhibitors against most ß-acetylglucosaminidases (ß-GlcNAcases) except for insect and bacterial chitinolytic ß-GlcNAcases, including the molting-indispensable OfHex1 from the insect Ostrinia furnacalis. Here, we report the co-crystal structure of OfHex1 in complex with NGT. This structure reveals a large active pocket in OfHex1 that may account for the poor inhibitory activity of NGT. To test this hypothesis, a bulky substituent was designed and synthesized on the thiazoline ring of NGT. The resulting compound (NMAGT) was determined to be a submicromolar inhibitor of OfHex1 with a Ki value of 0.13 µM, which is 600-fold lower than Ki value of NGT. Molecular dynamics simulation analysis supported the good fit of NMAGT to the active pocket.


Subject(s)
Acetylglucosamine/analogs & derivatives , Acetylglucosaminidase/chemistry , Enzyme Inhibitors/chemistry , Insect Proteins , Moths/enzymology , Thiazoles/chemistry , Acetylglucosamine/chemistry , Acetylglucosaminidase/antagonists & inhibitors , Animals , Insect Proteins/antagonists & inhibitors , Insect Proteins/chemistry , Molecular Dynamics Simulation
19.
Sci Rep ; 4: 6188, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25155420

ABSTRACT

Selective inhibition of function-specific ß-GlcNAcase has great potential in terms of drug design and biological research. The symmetrical bis-naphthalimide M-31850 was previously obtained by screening for specificity against human glycoconjugate-lytic ß-GlcNAcase. Using protein-ligand co-crystallization and molecular docking, we designed an unsymmetrical dyad of naphthalimide and thiadiazole, Q2, that changes naphthalimide specificity from against a human glycoconjugate-lytic ß-GlcNAcase to against insect and bacterial chitinolytic ß-GlcNAcases. The crystallographic and in silico studies reveal that the naphthalimide ring can be utilized to bind different parts of these enzyme homologs, providing a new starting point to design specific inhibitors. Moreover, Q2-induced closure of the substrate binding pocket is the structural basis for its 13-fold increment in inhibitory potency. Q2 is the first non-carbohydrate inhibitor against chitinolytic ß-GlcNAcases. This study provides a useful example of structure-based rationally designed inhibitors as potential pharmaceuticals or pesticides.


Subject(s)
Acetylglucosaminidase/chemistry , Enzyme Inhibitors/chemistry , Naphthalimides/chemistry , Thiadiazoles/chemistry , Acetylglucosaminidase/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Catalytic Domain , Crystallography, X-Ray , Drug Design , Humans , Insect Proteins/antagonists & inhibitors , Insect Proteins/chemistry , Kinetics , Molecular Docking Simulation , Protein Binding , Sequence Homology, Amino Acid , Substrate Specificity
20.
Aquat Toxicol ; 154: 163-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24907676

ABSTRACT

N-acetyl-ß-D-glucosaminidase (EC 3.2.1.52, NAGase), hydrolyzes dimers or trimers of N-acetyl-ß-D-glucosamine (NAG) into monomers and is shown to be important for the reproduction of male animals. NAGase is purified from the spermary of Nile tilapia, and its enzyme activity can be strongly inhibited by mercuric chloride (HgCl2). In this paper, we determined the kinetics of HgCl2-mediated inhibition of NAGase, and our results showed that it was irreversible inhibition with an IC50 value at 2.70±0.02 µM. Moreover, Hg(2+) reduced the thermal and pH stability of the enzyme. We determined the inhibition kinetics of Hg(2+) by using the kinetic method of substrate reaction. With this inhibition model, the microscopic rate constants for the reaction of Hg(2+) with free enzyme (k1) and the enzyme-substrate complex ( [Formula: see text] ) were determined to be 4.42×10(-4) mM(-1) s(-1) and 7.06×10(-5) mM(-1) s(-1), respectively, indicating that the presence of substrate can protect NAGase from Hg(2+) inhibition.


Subject(s)
Acetylglucosaminidase/metabolism , Cichlids/physiology , Mercuric Chloride/toxicity , Water Pollutants, Chemical/toxicity , Acetylglucosaminidase/antagonists & inhibitors , Animals , Enzyme Stability/drug effects , Inhibitory Concentration 50 , Ions/toxicity , Kinetics , Male
SELECTION OF CITATIONS
SEARCH DETAIL