Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Glycobiology ; 34(6)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38760939

ABSTRACT

Genetic deficiency of alpha-L-iduronidase causes mucopolysaccharidosis type I (MPS-I) disease, due to accumulation of glycosaminoglycans (GAGs) including chondroitin/dermatan sulfate (CS/DS) and heparan sulfate (HS) in cells. Currently, patients are treated by infusion of recombinant iduronidase or by hematopoietic stem cell transplantation. An alternative approach is to reduce the L-iduronidase substrate, through limiting the biosynthesis of iduronic acid. Our earlier study demonstrated that ebselen attenuated GAGs accumulation in MPS-I cells, through inhibiting iduronic acid producing enzymes. However, ebselen has multiple pharmacological effects, which prevents its application for MPS-I. Thus, we continued the study by looking for novel inhibitors of dermatan sulfate epimerase 1 (DS-epi1), the main responsible enzyme for production of iduronic acid in CS/DS chains. Based on virtual screening of chemicals towards chondroitinase AC, we constructed a library with 1,064 compounds that were tested for DS-epi1 inhibition. Seventeen compounds were identified to be able to inhibit 27%-86% of DS-epi1 activity at 10 µM. Two compounds were selected for further investigation based on the structure properties. The results show that both inhibitors had a comparable level in inhibition of DS-epi1while they had negligible effect on HS epimerase. The two inhibitors were able to reduce iduronic acid biosynthesis in CS/DS and GAG accumulation in WT and MPS-I fibroblasts. Docking of the inhibitors into DS-epi1 structure shows high affinity binding of both compounds to the active site. The collected data indicate that these hit compounds may be further elaborated to a potential lead drug used for attenuation of GAGs accumulation in MPS-I patients.


Subject(s)
Enzyme Inhibitors , Fibroblasts , Glycosaminoglycans , Mucopolysaccharidosis I , Mucopolysaccharidosis I/drug therapy , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology , Humans , Fibroblasts/metabolism , Fibroblasts/drug effects , Glycosaminoglycans/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Carbohydrate Epimerases/metabolism , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/genetics , Molecular Docking Simulation , Antigens, Neoplasm , DNA-Binding Proteins , Neoplasm Proteins
2.
Nat Chem Biol ; 11(9): 728-32, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26147354

ABSTRACT

The gateway to morphine biosynthesis in opium poppy (Papaver somniferum) is the stereochemical inversion of (S)-reticuline since the enzyme yielding the first committed intermediate salutaridine is specific for (R)-reticuline. A fusion between a cytochrome P450 (CYP) and an aldo-keto reductase (AKR) catalyzes the S-to-R epimerization of reticuline via 1,2-dehydroreticuline. The reticuline epimerase (REPI) fusion was detected in opium poppy and in Papaver bracteatum, which accumulates thebaine. In contrast, orthologs encoding independent CYP and AKR enzymes catalyzing the respective synthesis and reduction of 1,2-dehydroreticuline were isolated from Papaver rhoeas, which does not accumulate morphinan alkaloids. An ancestral relationship between these enzymes is supported by a conservation of introns in the gene fusions and independent orthologs. Suppression of REPI transcripts using virus-induced gene silencing in opium poppy reduced levels of (R)-reticuline and morphinan alkaloids and increased the overall abundance of (S)-reticuline and its O-methylated derivatives. Discovery of REPI completes the isolation of genes responsible for known steps of morphine biosynthesis.


Subject(s)
Aldehyde Reductase/metabolism , Carbohydrate Epimerases/metabolism , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Plant , Morphine/biosynthesis , Papaver/metabolism , Plant Proteins/metabolism , Aldehyde Reductase/genetics , Aldo-Keto Reductases , Alkaloids/biosynthesis , Alkaloids/chemistry , Base Sequence , Benzylisoquinolines/chemistry , Benzylisoquinolines/metabolism , Bromoviridae/genetics , Bromoviridae/metabolism , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/genetics , Cytochrome P-450 Enzyme System/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Exons , Gene Fusion , Introns , Ligases/genetics , Ligases/metabolism , Molecular Sequence Data , Morphinans/chemistry , Morphinans/metabolism , Morphine/chemistry , Open Reading Frames , Opium/chemistry , Opium/metabolism , Oxidation-Reduction , Papaver/genetics , Plant Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Stereoisomerism
3.
J Recept Signal Transduct Res ; 36(5): 515-30, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26795501

ABSTRACT

WbpP encoding UDP-GlcNAC C4 epimerase is responsible for the activation of virulence factor in marine pathogen Vibrio vulnificus (V. vulnificus) and it is linked to many aquatic diseases, thus making it a potential therapeutic target. There are few reported compounds that include several natural products and synthetic compounds targeting Vibrio sp, but specific inhibitor targeting WbpP are unavailable. Here, we performed structure-based virtual screening using chemical libraries such as Binding, TOSLab and Maybridge to identify small molecule inhibitors of WbpP with better drug-like properties. Deficient structural information forced to model the structure and the stable protein structure was obtained through 30 ns of MD simulations. Druggability regions are focused for new lead compounds and our screening protocol provides fast docking of entire small molecule library with screening criteria of ADME/Lipinski filter/Docking followed by re-docking of top hits using a method that incorporates both ligand and protein flexibility. Docking conformations of lead molecules interface displays strong H-bond interactions with the key residues Gly101, Ser102, Val195, Tyr165, Arg298, Val209, Ser142, Arg233 and Gln200. Subsequently, the top-ranking compounds were prioritized using the molecular dynamics simulation-based conformation and stability studies. Our study suggests that the proposed compounds may aid as a starting point for the rational design of novel therapeutic agents.


Subject(s)
Carbohydrate Epimerases/chemistry , Foodborne Diseases/drug therapy , Lead/chemistry , Vibrio vulnificus/chemistry , Aquatic Organisms/genetics , Aquatic Organisms/microbiology , Aquatic Organisms/pathogenicity , Binding Sites , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/metabolism , Drug Design , Foodborne Diseases/microbiology , Humans , Lead/therapeutic use , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship , Vibrio vulnificus/drug effects , Vibrio vulnificus/pathogenicity
4.
Proc Natl Acad Sci U S A ; 110(27): E2510-7, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776209

ABSTRACT

A cell-based phenotypic screen for inhibitors of biofilm formation in mycobacteria identified the small molecule TCA1, which has bactericidal activity against both drug-susceptible and -resistant Mycobacterium tuberculosis (Mtb) and sterilizes Mtb in vitro combined with rifampicin or isoniazid. In addition, TCA1 has bactericidal activity against nonreplicating Mtb in vitro and is efficacious in acute and chronic Mtb infection mouse models both alone and combined with rifampicin or isoniazid. Transcriptional analysis revealed that TCA1 down-regulates genes known to be involved in Mtb persistence. Genetic and affinity-based methods identified decaprenyl-phosphoryl-ß-D-ribofuranose oxidoreductase DprE1 and MoeW, enzymes involved in cell wall and molybdenum cofactor biosynthesis, respectively, as targets responsible for the activity of TCA1. These in vitro and in vivo results indicate that this compound functions by a unique mechanism and suggest that TCA1 may lead to the development of a class of antituberculosis agents.


Subject(s)
Antitubercular Agents/pharmacology , Benzothiazoles/pharmacology , Mycobacterium tuberculosis/drug effects , Thiophenes/pharmacology , Tuberculosis, Pulmonary/drug therapy , Alcohol Oxidoreductases , Amino Acid Sequence , Animals , Antitubercular Agents/administration & dosage , Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Benzothiazoles/administration & dosage , Benzothiazoles/chemistry , Biofilms/drug effects , Biofilms/growth & development , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/chemistry , Carbohydrate Epimerases/genetics , Drug Resistance, Bacterial , Female , Genes, Bacterial , High-Throughput Screening Assays , Isoniazid/administration & dosage , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Molecular Sequence Data , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/chemistry , Oxidoreductases/genetics , Rifampin/administration & dosage , Thiophenes/administration & dosage , Thiophenes/chemistry , Tuberculosis, Pulmonary/microbiology
5.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 4): 658-68, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23519675

ABSTRACT

ADP-L-glycero-D-manno-heptose 6-epimerase (AGME), the product of the rfaD gene, is the last enzyme in the heptose-biosynthesis pathway; it converts ADP-D-glycero-D-manno-heptose (ADP-D,D-Hep) to ADP-L-glycero-D-manno-heptose (ADP-L,D-Hep). AGME contains a catalytic triad involved in catalyzing hydride transfer with the aid of NADP(+). Defective lipopolysaccharide is found in bacterial mutants lacking this gene. Therefore, it is an interesting target enzyme for a novel epimerase inhibitor for use as a co-therapy with antibiotics. The crystal structure of AGME from Burkholderia thailandensis (BtAGME), a surrogate organism for studying the pathogenicity of melioidosis caused by B. pseudomallei, has been determined. The crystal structure determined with co-purified NADP(+) revealed common as well as unique structural properties of the AGME family when compared with UDP-galactose 4-epimerase homologues. They form a similar architecture with conserved catalytic residues. Nevertheless, there are differences in the substrate- and cofactor-binding cavities and the oligomerization domains. Structural comparison of BtAGME with AGME from Escherichia coli indicates that they may recognize their substrate in a `lock-and-key' fashion. Unique structural features of BtAGME are found in two regions. The first region is the loop between ß8 and ß9, affecting the binding affinity of BtAGME for the ADP moiety of ADP-D,D-Hep. The second region is helix α8, which induces decamerization at low pH that is not found in other AGMEs. With the E210G mutant, it was observed that the resistance of the wild type to acid-induced denaturation is related to the decameric state. An in silico study was performed using the Surflex-Dock GeomX module of the SYBYL-X 1.3 software to predict the catalytic mechanism of BtAGME with its substrate, ADP-D,D-Hep. In the in silico study, the C7'' hydroxymethyl group of ADP-D,D-Hep is predicted to form hydrogen bonds to Ser116 and Gln293. With the aid of these interactions, the hydroxyl of Tyr139 forms a hydrogen bond to O6″ of ADP-D,D-Hep and the proton at C6″ orients closely to C4 of NADP(+). Therefore, the in silico study supports a one-base mechanism as a major catalytic pathway, in which Tyr139 solely functions as a catalytic acid/base residue. These results provide a new insight into the development of an epimerase inhibitor as an antibiotic adjuvant against melioidosis.


Subject(s)
Burkholderia/enzymology , Carbohydrate Epimerases/chemistry , Carbohydrate Epimerases/metabolism , Adjuvants, Pharmaceutic/chemistry , Adjuvants, Pharmaceutic/therapeutic use , Animals , Carbohydrate Epimerases/antagonists & inhibitors , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Humans , Melioidosis/drug therapy , Melioidosis/enzymology , Melioidosis/microbiology , Protein Binding , Structure-Activity Relationship , Substrate Specificity
6.
Bioengineered ; 13(3): 5551-5563, 2022 03.
Article in English | MEDLINE | ID: mdl-35184647

ABSTRACT

Eriodictyol is a natural flavonoid with many pharmacological effects, such as anti-oxidation, anti-inflammation, anti-tumor, and neuroprotection. Besides, it has been reported that flavonoids play an important role in protein glycosylation. The fucosylation structure is closely associated with processes of various tumor metastases. TSTA3 is involved in the de novo synthesis and can convert cellular GDP-D-mannose into GDP-L-fucose. It was predicted on the STITCH database that eriodictyol interacted with TSTA3. In addition, literature has confirmed that TSTA3 is upregulated in CRC and can regulate the proliferation and migration of breast cancer cells. Herein, the precise effects of eriodictyol on the clone-forming, proliferative, migratory and invasive abilities of CRC cells as well as EMT process were assessed. Moreover, the correlation among eriodictyol, TSTA3, and fucosylation in these malignant behaviors of CRC cells was evaluated, in order to elucidate the underlying mechanism. The current work discovered that eriodictyol inhibited the viability, clone-formation, proliferation, migration, invasion, and EMT of CRC cells, and that these inhibitory effects of eriodictyol on the malignant behavior of CRC cells were reversed by TSTA3 overexpression. Additionally, eriodictyol suppresses fucosylation by downregulating the TSTA3 expression. Results confirmed that fucosylation inhibitor (2-F-Fuc) inhibited clone formation, proliferation, migration, invasion, as well as EMT of CRC cells and eriodictyol treatment further reinforced the suppressing effects of 2-F-Fuc on the malignant behavior of CRC cells. We conclude that eriodictyol suppresses the clone-forming, proliferative, migrative and invasive abilities of CRC cells as well as represses the EMT process by downregulating TSTA3 expression to restrain fucosylation.


Subject(s)
Carbohydrate Epimerases , Colorectal Neoplasms , Ketone Oxidoreductases , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/drug therapy , Epithelial-Mesenchymal Transition , Flavanones , Glycosylation , Guanosine Diphosphate Fucose/metabolism , Guanosine Diphosphate Fucose/pharmacology , Humans , Ketone Oxidoreductases/antagonists & inhibitors , Ketone Oxidoreductases/metabolism
7.
Biotechnol Prog ; 37(1): e3061, 2021 01.
Article in English | MEDLINE | ID: mdl-32748555

ABSTRACT

Antibody-dependent cellular cytotoxicity (ADCC) is the primary mechanism of actions for several marketed therapeutic antibodies (mAbs) and for many more in clinical trials. The ADCC efficacy is highly dependent on the ability of therapeutic mAbs to recruit effector cells such as natural killer cells, which induce the apoptosis of targeted cells. The recruitment of effector cells by mAbs is negatively affected by fucose modification of N-Glycans on the Fc; thus, utilization of afucosylated mAbs has been a trend for enhanced ADCC therapeutics. Most of afucosylated mAbs in clinical or commercial manufacturing were produced from Fut8-/- Chinese hamster ovary cells (CHO) host cells, generally generating low yields compared to wildtype CHO host. This study details the generation and characterization of two engineered CHOZN® cell lines, in which the enzyme involved in guanosine diphosphate (GDP)-fucose synthesis, GDP mannose-4,6-dehydratase (Gmds) and GDP-L-fucose synthase (FX), was knocked out. The top host cell lines for each of the knockouts, FX-/- and Gmds-/-, were selected based on growth robustness, bulk MSX selection tolerance, production titer, fucosylation level, and cell stability. We tested the production of two proprietary IgG1 mAbs in the engineered host cells, and found that the titers were comparable to CHOZN® cells. The mAbs generated from either KO cell line exhibited loss of fucose modification, leading to significantly boosted FcγRIIIa binding and ADCC effects. Our data demonstrated that both FX-/- and Gmds-/- host cells could replace Fut8-/- CHO cells for clinical manufacturing of antibody therapeutics.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Carbohydrate Epimerases/antagonists & inhibitors , Fucose/metabolism , Guanosine Diphosphate/metabolism , Hydro-Lyases/antagonists & inhibitors , Ketone Oxidoreductases/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Base Sequence , CHO Cells , CRISPR-Cas Systems , Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/metabolism , Cricetinae , Cricetulus , Glycosylation , Humans , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Immunoglobulin G/immunology , Ketone Oxidoreductases/genetics , Ketone Oxidoreductases/metabolism , Receptors, IgG/metabolism
8.
Bioorg Med Chem ; 18(2): 896-908, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19969466

ABSTRACT

High-throughput screening of 201,368 compounds revealed that 1-(3-(5-ethyl-5H-[1,2,4]triazino[5,6-b]indol-3-ylthio)propyl)-1H-benzo[d]imidazol-2(3H)-one (SID 7975595) inhibited RmlC a TB cell wall biosynthetic enzyme. SID 7975595 acts as a competitive inhibitor of the enzyme's substrate and inhibits RmlC as a fast-on rate, fully reversible inhibitor. An analog of SID 7975595 had a K(i) of 62nM. Computer modeling showed that the binding of the tethered two-ringed system into the active site occurred at the thymidine binding region for one ring system and the sugar region for the other ring system.


Subject(s)
Benzimidazoles/pharmacology , Carbohydrate Epimerases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Mycobacterium tuberculosis/enzymology , Aorta/cytology , Aorta/drug effects , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Catalytic Domain , Cell Survival/drug effects , Computer Simulation , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Humans , Indoles/chemical synthesis , Indoles/chemistry , Models, Chemical , Structure-Activity Relationship
9.
Cell Chem Biol ; 24(12): 1467-1478.e5, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29033318

ABSTRACT

Fucosylation is a glycan modification critically involved in cancer and inflammation. Although potent fucosylation inhibitors are useful for basic and clinical research, only a few inhibitors have been developed. Here, we focus on a fucose analog with an alkyne group, 6-alkynyl-fucose (6-Alk-Fuc), which is used widely as a detection probe for fucosylated glycans, but is also suggested for use as a fucosylation inhibitor. Our glycan analysis using lectin and mass spectrometry demonstrated that 6-Alk-Fuc is a potent and general inhibitor of cellular fucosylation, with much higher potency than the existing inhibitor, 2-fluoro-fucose (2-F-Fuc). The action mechanism was shown to deplete cellular GDP-Fuc, and the direct target of 6-Alk-Fuc is FX (encoded by TSTA3), the bifunctional GDP-Fuc synthase. We also show that 6-Alk-Fuc halts hepatoma invasion. These results highlight the unappreciated role of 6-Alk-Fuc as a fucosylation inhibitor and its potential use for basic and clinical science.


Subject(s)
Alkynes/pharmacology , Antineoplastic Agents/pharmacology , Carbohydrate Epimerases/antagonists & inhibitors , Carcinoma, Hepatocellular/drug therapy , Enzyme Inhibitors/pharmacology , Fucose/pharmacology , Guanosine Diphosphate Fucose/biosynthesis , Ketone Oxidoreductases/antagonists & inhibitors , Liver Neoplasms/drug therapy , Alkynes/chemistry , Antineoplastic Agents/chemistry , Carbohydrate Epimerases/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Fucose/chemistry , HEK293 Cells , HeLa Cells , Humans , Ketone Oxidoreductases/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology
10.
Structure ; 8(5): 453-62, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10896473

ABSTRACT

BACKGROUND: ADP-L-glycero--mannoheptose 6-epimerase (AGME) is required for lipopolysaccharide (LPS) biosynthesis in most genera of pathogenic and non-pathogenic Gram-negative bacteria. It catalyzes the interconversion of ADP-D-glycero-D-mannoheptose and ADP-L-glycero-D-mannoheptose, a precursor of the seven-carbon sugar L-glycero-mannoheptose (heptose). Heptose is an obligatory component of the LPS core domain; its absence results in a truncated LPS structure resulting in susceptibility to hydrophobic antibiotics. Heptose is not found in mammalian cells, thus its biosynthetic pathway in bacteria presents a unique target for the design of novel antimicrobial agents. RESULTS: The structure of AGME, in complex with NADP and the catalytic inhibitor ADP-glucose, has been determined at 2.0 A resolution by multiwavelength anomalous diffraction (MAD) phasing methods. AGME is a homopentameric enzyme, which crystallizes with two pentamers in the asymmetric unit. The location of 70 crystallographically independent selenium sites was a key step in the structure determination process. Each monomer comprises two domains: a large N-terminal domain, consisting of a modified seven-stranded Rossmann fold that is associated with NADP binding; and a smaller alpha/beta C-terminal domain involved in substrate binding. CONCLUSIONS: The first structure of an LPS core biosynthetic enzyme leads to an understanding of the mechanism of the conversion between ADP-D-glycero--mannoheptose and ADP-L-glycero-D-mannoheptose. On the basis of its high structural similarity to UDP-galactose epimerase and the three-dimensional positions of the conserved residues Ser116, Tyr140 and Lys144, AGME was classified as a member of the short-chain dehydrogenase/reductase (SDR) superfamily. This study should prove useful in the design of mechanistic and structure-based inhibitors of the AGME catalyzed reaction.


Subject(s)
Bacterial Proteins/chemistry , Carbohydrate Epimerases/chemistry , Models, Molecular , Protein Structure, Quaternary , Adenosine Diphosphate Glucose/chemistry , Adenosine Diphosphate Glucose/pharmacology , Binding Sites , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/metabolism , Catalysis , Crystallography, X-Ray , Escherichia coli/enzymology , Lipopolysaccharides/biosynthesis , NADP/metabolism , Protein Structure, Tertiary , Reproducibility of Results , Selenium/chemistry , Selenium/metabolism , X-Ray Diffraction/methods
11.
Structure ; 3(12): 1323-32, 1995 Dec 15.
Article in English | MEDLINE | ID: mdl-8747459

ABSTRACT

BACKGROUND: Glucosamine 6-phosphate deaminase from Escherichia coli is an allosteric hexameric enzyme which catalyzes the reversible conversion of D-glucosamine 6-phosphate into D-fructose 6-phosphate and ammonium ion and is activated by N-acetyl-D-glucosamine 6-phosphate. Mechanistically, it belongs to the group of aldoseketose isomerases, but its reaction also accomplishes a simultaneous amination/deamination. The determination of the structure of this protein provides fundamental knowledge for understanding its mode of action and the nature of allosteric conformational changes that regulate its function. RESULTS: The crystal structure of glucosamine 6-phosphate deaminase with bound phosphate ions is presented at 2.1 A resolution together with the refined structures of the enzyme in complexes with its allosteric activator and with a competitive inhibitor. The protein fold can be described as a modified NAD-binding domain. CONCLUSIONS: From the similarities between the three presented structures, it is concluded that these represent the enzymatically active R state conformer. A mechanism for the deaminase reaction is proposed. It comprises steps to open the pyranose ring of the substrate and a sequence of general base-catalyzed reactions to bring about isomerization and deamination, with Asp72 playing a key role as a proton exchanger.


Subject(s)
Aldose-Ketose Isomerases , Bacterial Proteins/chemistry , Carbohydrate Epimerases/chemistry , Escherichia coli/enzymology , Glucose-6-Phosphate/analogs & derivatives , Models, Molecular , Protein Conformation , Allosteric Regulation , Bacterial Proteins/metabolism , Binding Sites , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/metabolism , Catalysis , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Fructosephosphates/biosynthesis , Glucosamine/analogs & derivatives , Glucosamine/metabolism , Glucosephosphates/metabolism , Macromolecular Substances , NAD/metabolism , Phosphates/metabolism , Sorbitol/analogs & derivatives , Sorbitol/chemistry , Sorbitol/pharmacology , Sugar Phosphates/chemistry , Sugar Phosphates/pharmacology
12.
Sci Rep ; 6: 23274, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26980148

ABSTRACT

The bifunctional enzyme UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE) plays a key role in sialic acid production. It is different from the non-hydrolyzing enzymes for bacterial cell wall biosynthesis, and it is feed-back inhibited by the downstream product CMP-Neu5Ac. Here the complex crystal structure of the N-terminal epimerase part of human GNE shows a tetramer in which UDP binds to the active site and CMP-Neu5Ac binds to the dimer-dimer interface. The enzyme is locked in a tightly closed conformation. By comparing the UDP-binding modes of the non-hydrolyzing and hydrolyzing UDP-GlcNAc epimerases, we propose a possible explanation for the mechanistic difference. While the epimerization reactions of both enzymes are similar, Arg113 and Ser302 of GNE are likely involved in product hydrolysis. On the other hand, the CMP-Neu5Ac binding mode clearly elucidates why mutations in Arg263 and Arg266 can cause sialuria. Moreover, full-length modelling suggests a channel for ManNAc trafficking within the bifunctional enzyme.


Subject(s)
N-Acetylneuraminic Acid/biosynthesis , Allosteric Regulation , Amino Acid Sequence , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/chemistry , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Cytidine Monophosphate/analogs & derivatives , Cytidine Monophosphate/chemistry , Enzyme Inhibitors/chemistry , Humans , Hydrogen Bonding , Hydrolysis , Kinetics , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Sialic Acids/chemistry , Uridine Diphosphate/chemistry
13.
Carbohydr Res ; 419: 18-28, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26598987

ABSTRACT

Neisseria meningitidis serogroup A non-hydrolyzing uridine 5'-diphosphate-N-acetylglucosamine (UDP-GlcNAc) 2-epimerase (NmSacA) catalyzes the interconversion between UDP-GlcNAc and uridine 5'-diphosphate-N-acetylmannosamine (UDP-ManNAc). It is a key enzyme involved in the biosynthesis of the capsular polysaccharide [-6ManNAcα1-phosphate-]n of N. meningitidis serogroup A, one of the six serogroups (A, B, C, W-135, X, and Y) that account for most cases of N. meningitidis-caused bacterial septicemia and meningitis. N. meningitidis serogroup A is responsible for large epidemics in the developing world, especially in Africa. Here we report that UDP-ManNAc could be used as a substrate for C-terminal His6-tagged recombinant NmSacA (NmSacA-His6) in the absence of UDP-GlcNAc. NmSacA-His6 was activated by UDP-GlcNAc and inhibited by 2-acetamidoglucal and UDP. Substrate specificity study showed that NmSacA-His6 could tolerate several chemoenzymatically synthesized UDP-ManNAc derivatives as substrates although its activity was much lower than non-modified UDP-ManNAc. Homology modeling and molecular docking revealed likely structural determinants of NmSacA substrate specificity. This is the first detailed study of N. meningitidis serogroup A UDP-GlcNAc 2-epimerase.


Subject(s)
Neisseria meningitidis/enzymology , Uridine Diphosphate N-Acetylglucosamine/chemistry , Uridine Diphosphate N-Acetylglucosamine/metabolism , Amino Acid Sequence , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/chemistry , Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/metabolism , Catalytic Domain , Cloning, Molecular , Enzyme Activation/drug effects , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Hexosamines/metabolism , Hexosamines/pharmacology , Molecular Docking Simulation , Molecular Sequence Data , Neisseria meningitidis/genetics , Substrate Specificity , Uridine Diphosphate/metabolism , Uridine Diphosphate/pharmacology
14.
J Biomol Screen ; 21(6): 579-89, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27028606

ABSTRACT

Nonessential enzymes in the staphylococcal wall teichoic acid (WTA) pathway serve as highly validated ß-lactam potentiation targets. MnaA (UDP-GlcNAc 2-epimerase) plays an important role in an early step of WTA biosynthesis by providing an activated form of ManNAc. Identification of a selective MnaA inhibitor would provide a tool to interrogate the contribution of the MnaA enzyme in the WTA pathway as well as serve as an adjuvant to restore ß-lactam activity against methicillin-resistant Staphylococcus aureus (MRSA). However, development of an epimerase functional assay can be challenging since both MnaA substrate and product (UDP-GlcNAc/UDP-ManNAc) share an identical molecular weight. Herein, we developed a nuclear magnetic resonance (NMR) functional assay that can be combined with other NMR approaches to triage putative MnaA inhibitors from phenotypic cell-based screening campaigns. In addition, we determined that tunicamycin, a potent WTA pathway inhibitor, inhibits both S. aureus MnaA and a functionally redundant epimerase, Cap5P.


Subject(s)
Cell Wall/drug effects , Magnetic Resonance Spectroscopy/methods , Methicillin-Resistant Staphylococcus aureus/drug effects , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Epimerases/chemistry , Cell Wall/chemistry , Humans , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Teichoic Acids/chemistry , Teichoic Acids/metabolism , Uridine Diphosphate Sugars/chemistry , Uridine Diphosphate Sugars/metabolism , beta-Lactam Resistance/drug effects , beta-Lactamases/chemistry , beta-Lactamases/drug effects
15.
Biochim Biophys Acta ; 570(2): 352-62, 1979 Oct 11.
Article in English | MEDLINE | ID: mdl-497230

ABSTRACT

D-Ribulose-5-phosphate 3-epimerase (EC 5.1.3.1) was purified 760-fold from calf liver by adsorption on DEAE-cellulose, chromatography on DEAE-Sephadex, chromatography on D-ribose 5-phosphate-Sepharose and gel filtration on Biogel P200. The purified enzyme of specific activity 617 units/mg was obtained in 28% yield and gave a single band on polyacrylamide gel electrophoresis. It had a molecular weight of 45 000 and appeared to contain two identical peptide chains of 22 900 daltons. The Km for D-ribulose 5-phosphate was 0.19 +/- 0.07 mM (S.E.). It was inhibited by reagents reacting with sulphydryl groups, by sulphate ion, and by D-deoxyribose 5-phosphate. The pH-stability and pH-activity curves were determined.


Subject(s)
Carbohydrate Epimerases/isolation & purification , Liver/enzymology , Pentosephosphates/metabolism , Ribulosephosphates/metabolism , Animals , Carbohydrate Epimerases/antagonists & inhibitors , Cattle , Chromatography, Gel , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Kinetics
16.
Biochim Biophys Acta ; 787(1): 45-52, 1984 May 31.
Article in English | MEDLINE | ID: mdl-6426523

ABSTRACT

Incubation of anticapsin with the purified glucosamine synthetase (2-amino-2-deoxy-D-glucose-6-phosphate ketol-isomerase, amino transferring, EC 5.3.1.19) from Escherichia coli, Pseudomonas aeruginosa, Arthrobacter aurescens and Bacillus thuringiensis led to the formation of an inactive enzyme irreversibly modified. The inactivation reaction followed pseudo-first-order kinetics. The rate of the inactivation reaction at various concentrations of anticapsin exhibited saturation kinetics, implying that anticapsin binds reversibly to the enzyme prior to inactivation. The determined Kinact is in the range of 10(-5) M (B. thuringiensis) and 10(-6) M (E. coli, P. aeruginosa, A. aurescens ). The addition of glutamine protected the amidotransferase from inactivation by anticapsin . The anticapsin was demonstrated to be a mixed type or competitive inhibitor with respect to glutamine with a Ki value of 10(-6) to 10(-7) M. Reaction of anticapsin with the enzyme exhibits the characteristics of affinity labelling of the glutamine binding site. Chemical modification of the enzyme thiol group with various reagents, 5,5'-dithiobis-(2-nitrobenzoic) acid, 6,6'- dithiodinicotinic acid, 1,1'- dithiodiformamidine , N-ethylmaleimide and iodoacetamide, resulted in an inactive enzyme.


Subject(s)
Alanine/analogs & derivatives , Arthrobacter/enzymology , Bacillus thuringiensis/enzymology , Carbohydrate Epimerases/antagonists & inhibitors , Escherichia coli/enzymology , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/antagonists & inhibitors , Pseudomonas aeruginosa/enzymology , Alanine/pharmacology , Binding, Competitive , Glutamine/analogs & derivatives , Glutamine/pharmacology , Kinetics , Protein Binding
17.
Biochim Biophys Acta ; 828(3): 247-54, 1985 Apr 29.
Article in English | MEDLINE | ID: mdl-3921053

ABSTRACT

Synthetic derivatives of N3-fumaroyl-L-2,3-diaminopropanoic acid constitute the novel group of glutamine analogs. They are powerful, competitive inhibitors of the glucosamine synthetase (2-amino-2-deoxy-D-glucose-6-phosphate ketol-isomerase (amino-transferring), EC 5.3.1.19) from Candida albicans with respect to glutamine and uncompetitive with respect to D-fructose 6-phosphate. Some of the compounds tested irreversibly inactivate glucosamine synthetase with Kinact values of 10(-4) to 10(-6) M. The addition of glutamine protects enzyme from the inactivation, while the absence of D-fructose 6-phosphate lowers the rate of inactivation. An ordered, sequential mechanism is suggested for binding of the inhibitors to the glutamine-binding site. A number of tested compounds act as active-site-directed, irreversible inhibitors. It is suggested that derivatives of N3-fumaroyl-L-2,3-diaminopropanoic acid should be classified as mechanism-based enzyme inactivators. Structural requirements for an effective inactivator containing N3-fumaroyl-L-2,3-diaminopropanoic acid moiety are discussed.


Subject(s)
Alanine/analogs & derivatives , Candida albicans/enzymology , Carbohydrate Epimerases/antagonists & inhibitors , Fumarates/pharmacology , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/antagonists & inhibitors , beta-Alanine/analogs & derivatives , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/isolation & purification , Kinetics , Structure-Activity Relationship , beta-Alanine/pharmacology
18.
J Mol Biol ; 212(1): 211-35, 1990 Mar 05.
Article in English | MEDLINE | ID: mdl-2319597

ABSTRACT

The active site and mechanism of D-xylose isomerase have been probed by determination of the crystal structures of the enzyme bound to various substrates, inhibitors and cations. Ring-opening is an obligatory first step of the reaction and is believed to be the rate-determining step for the aldose to ketose conversion. The structure of a complex with a cyclic thio-glucose has been determined and it is concluded that this is an analogue of the Michaelis complex. At -10 degrees C substrates in crystals are observed in the extended chain form. The absence of an appropriately situated base for either the cyclic or extended chain forms from the substrate binding site indicates that the isomerisation does not take place by an enediol or enediolate mechanism. Binding of a trivalent cation places an additional charge at the active site, producing a substrate complex that is analogous to a possible transition state. Of the two binding sites for divalent cations, [1] is permanently occupied under catalytic conditions and is co-ordinated to four carboxylate groups. In the absence of substrate it is exposed to solvent, and in the Michaelis complex analogue, site [1] is octahedrally coordinated, with ligands to O-3 and O-4 of the thiopyranose. In the complex with an open-chain substrate it remains octahedrally co-ordinated, with ligands to O-2 and O-4. Binding at a second cation site [2] is also necessary for catalysis and this site is believed to bind Co2+ more strongly than site [1]. This site is octahedrally co-ordinated to three carboxylate groups (bidentate co-ordination to one of them), an imidazole and a solvent molecule. It is proposed that during the hydride shift the C-O-1 and C-O-2 bonds of the substrate are polarized by the close approach of the site [2] cation. In the transition-state analogue this cation is observed at a site [2'], 1.0 A from site [2] and about 2.7 A from O-1 and O-2 of the substrate. It is likely that co-ordination of the cation to O-1 and O-2 would be concomitant with ionisation of the sugar hydroxyl group. The polarisation of C-O-1 and C-O-2 is assisted by the co-ordination of O-2 to cation [1] and O-1 to a lysine side-chain.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Aldose-Ketose Isomerases , Arthrobacter/enzymology , Carbohydrate Epimerases/metabolism , Apoenzymes/metabolism , Binding Sites/physiology , Carbohydrate Epimerases/antagonists & inhibitors , Carbohydrate Metabolism , Crystallization , Hydrogen Bonding , Isomerism , Macromolecular Substances , Metals/metabolism , Models, Molecular , Protein Conformation , Substrate Specificity , X-Ray Diffraction
19.
Chem Biol ; 9(1): 113-29, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11841944

ABSTRACT

The biological study of O-linked glycosylation is particularly problematic, as chemical tools to control this modification are lacking. An inhibitor of the UDP-GlcNAc 4-epimerase that synthesizes UDP-GalNAc, the donor initiating O-linked glycosylation, would be a powerful reagent for reversibly inhibiting O-linked glycosylation. We synthesized a 1338 member library of uridine analogs directed to the epimerase by virtue of substrate mimicry. Screening of the library identified an inhibitor with a K(i) value of 11 microM. Tests against related enzymes confirmed the compound's specificity for the UDP-GlcNAc 4-epimerase. Inhibitors of a key step of O-linked glycan biosynthesis can be discovered from a directed library screen. Progeny thereof may be powerful tools for controlling O-linked glycosylation in cells.


Subject(s)
Carbohydrate Epimerases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Carbohydrate Epimerases/metabolism , Enzyme Inhibitors/metabolism , Glycosylation , Humans , Peptide Library , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Uridine/metabolism
20.
Comput Biol Med ; 58: 110-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25637777

ABSTRACT

BACKGROUND: Tuberculosis remains one of the deadliest infectious diseases in humans. It has caused more than 100 million deaths since its discovery in 1882. Currently, more than 5 million people are infected with TB bacterium each year. The cell wall of Mycobacterium tuberculosis plays an important role in maintaining the ability of mycobacteria to survive in a hostile environment. Therefore, we report a virtual screening (VS) study aiming to identify novel inhibitors that simultaneously target RmlB and RmlC, which are two essential enzymes for the synthesis of the cell wall of M. tuberculosis. METHODS: A hybrid VS method that combines drug-likeness prediction, pharmacophore modeling and molecular docking studies was used to indentify inhibitors targeting RmlB and RmlC. RESULTS: The pharmacophore models HypoB and HypoC of RmlB inhibitors and RmlC inhibitors, respectively, were developed based on ligands complexing with their corresponding receptors. In total, 20 compounds with good absorption, distribution, metabolism, excretion, and toxicity properties were carefully selected using the hybird VS method. DISCUSSION: We have established a hybrid VS method to discover novel inhibitors with new scaffolds. The molecular interactions of the selected potential inhibitors with the active-site residues are discussed in detail. These compounds will be further evaluated using biological activity assays and deserve consideration for further structure-activity relationship studies.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Carbohydrate Epimerases/antagonists & inhibitors , Cell Wall/drug effects , Drug Discovery/methods , Mycobacterium tuberculosis/drug effects , Antitubercular Agents/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carbohydrate Epimerases/chemistry , Carbohydrate Epimerases/metabolism , Cell Wall/metabolism , Molecular Docking Simulation , Mycobacterium tuberculosis/cytology , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL