Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Type of study
Publication year range
1.
Elife ; 122023 Dec 21.
Article in English | MEDLINE | ID: mdl-38127423

ABSTRACT

Germline CTLA-4 deficiency causes severe autoimmune diseases characterized by dysregulation of Foxp3+ Tregs, hyper-activation of effector memory T cells, and variable forms autoimmune cytopenia including gradual loss of B cells. Cancer patients with severe immune-related adverse events (irAE) after receiving anti-CTLA-4/PD-1 combination immunotherapy also have markedly reduced peripheral B cells. The immunological basis for B cell loss remains unexplained. Here, we probe the decline of B cells in human CTLA-4 knock-in mice by using anti-human CTLA-4 antibody Ipilimumab conjugated to a drug payload emtansine (Anti-CTLA-4 ADC). The anti-CTLA-4 ADC-treated mice have T cell hyper-proliferation and their differentiation into effector cells which results in B cell depletion. B cell depletion is mediated by both CD4 and CD8 T cells and at least partially rescued by anti-TNF-alpha antibody. These data revealed an unexpected antagonism between T and B cells and the importance of regulatory T cells in preserving B cells.


Subject(s)
Abatacept , B-Lymphocytes , T-Lymphocytes, Regulatory , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Abatacept/pharmacology , Animals , Mice , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Lymphocyte Depletion , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Apoptosis/drug effects , Immunoglobulins/blood , Immunoglobulins/immunology , CHO Cells , Cricetulus , Mice, Inbred C57BL , Male , Female
2.
Sci Transl Med ; 15(685): eabm5663, 2023 03.
Article in English | MEDLINE | ID: mdl-36857433

ABSTRACT

Immune checkpoint inhibitors (ICIs), such as nivolumab and ipilimumab, not only elicit antitumor responses in a wide range of human cancers but also cause severe immune-related adverse events (irAEs), including death. A largely unmet medical need is to treat irAEs without abrogating the immunotherapeutic effect of ICIs. Although abatacept has been used to treat irAEs, it risks neutralizing the anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4) monoclonal antibodies administered for cancer therapy, thereby reducing the efficacy of anti-CTLA-4 immunotherapy. To avoid this caveat, we compared wild-type abatacept and mutants of CTLA-4-Ig for their binding to clinically approved anti-CTLA-4 antibodies and for their effect on both irAEs and immunotherapy conferred by anti-CTLA-4 and anti-PD-1 antibodies. Here, we report that whereas abatacept neutralized the therapeutic effect of anti-CTLA-4 antibodies, the mutants that bound to B7-1 and B7-2, but not to clinical anti-CTLA-4 antibodies, including clinically used belatacept, abrogated irAEs without affecting cancer immunotherapy. Our data demonstrate that anti-CTLA-4-induced irAEs can be corrected by provision of soluble CTLA-4 variants and that the clinically available belatacept may emerge as a broadly applicable drug to abrogate irAEs while preserving the therapeutic efficacy of CTLA-4-targeting ICIs.


Subject(s)
Immune Checkpoint Inhibitors , Immunotherapy , Humans , Abatacept , Ipilimumab , Nivolumab
3.
bioRxiv ; 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-36909522

ABSTRACT

Germline CTLA-4 deficiency causes severe autoimmune diseases characterized by dysregulation of Foxp3+ Tregs, hyper-activation of effector memory T cells, and variable forms autoimmune cytopenia including gradual loss of B cells. Cancer patients with severe immune-related adverse events (irAE) after receiving anti-CTLA-4/PD-1 combination immunotherapy also have markedly reduced peripheral B cells. The immunological basis for B cell loss remains unexplained. Here we probe the decline of B cells in human CTLA-4 knock-in mice by using antihuman CTLA-4 antibody Ipilimumab conjugated to a drug payload emtansine (Anti-CTLA-4 ADC). The anti-CTLA-4 ADC-treated mice have T cell hyper-proliferation and their differentiation into effector cells which results in B cell depletion. B cell depletion is mediated by both CD4 and CD8 T cells and at least partially rescued by anti-TNF-alpha antibody. These data revealed an unexpected antagonism between T and B cells and the importance of regulatory T cells in preserving B cells.

4.
ACS Chem Biol ; 17(11): 3013-3023, 2022 11 18.
Article in English | MEDLINE | ID: mdl-35316032

ABSTRACT

Lysosome-targeting chimeras (LYTACs) offer an opportunity for the degradation of extracellular and membrane-associated proteins of interest. Here, we report an efficient chemoenzymatic method that enables a single-step and site-specific conjugation of high-affinity mannose-6-phosphate (M6P) glycan ligands to antibodies without the need of protein engineering and conventional click reactions that would introduce "unnatural" moieties, yielding homogeneous antibody-M6P glycan conjugates for targeted degradation of membrane-associated proteins. Using trastuzumab and cetuximab as model antibodies, we showed that the wild-type endoglycosidase S (Endo-S) could efficiently perform the antibody deglycosylation and simultaneous transfer of an M6P-glycan from a synthetic M6P-glycan oxazoline to the deglycosylated antibody in a one-pot manner, giving structurally well-defined antibody-M6P glycan conjugates. A two-step procedure, using wild-type Endo-S2 for deglycosylation followed by transglycosylation with an Endo-S2 mutant (D184M), was also efficient to provide M6P glycan-antibody conjugates. The chemoenzymatic approach was highly specific for Fc glycan remodeling when both Fc and Fab domains were glycosylated, as exemplified by the selective Fc-glycan remodeling of cetuximab. SPR binding analysis indicated that the M6P conjugates possessed a nanomolar range of binding affinities for the cation-independent mannose-6-phosphate receptor (CI-MPR). Preliminary cell-based assays showed that the M6P-trastuzumab and M6P-cetuximab conjugates were able to selectively degrade the membrane-associated HER2 and EGFR, respectively. This modular glycan-remodeling strategy is expected to find wide applications for antibody-based lysosome-targeted degradation of extracellular and membrane proteins.


Subject(s)
Antibodies , Polysaccharides , Proteolysis , Cetuximab , Ligands , Antibodies/chemistry , Polysaccharides/metabolism , Trastuzumab
5.
J Biol Chem ; 298(2): 101464, 2022 02.
Article in English | MEDLINE | ID: mdl-34864059

ABSTRACT

Wall teichoic acid (WTA) polymers are covalently affixed to the Gram-positive bacterial cell wall and have important functions in cell elongation, cell morphology, biofilm formation, and ß-lactam antibiotic resistance. The first committed step in WTA biosynthesis is catalyzed by the TagA glycosyltransferase (also called TarA), a peripheral membrane protein that produces the conserved linkage unit, which joins WTA to the cell wall peptidoglycan. TagA contains a conserved GT26 core domain followed by a C-terminal polypeptide tail that is important for catalysis and membrane binding. Here, we report the crystal structure of the Thermoanaerobacter italicus TagA enzyme bound to UDP-N-acetyl-d-mannosamine, revealing the molecular basis of substrate binding. Native MS experiments support the model that only monomeric TagA is enzymatically active and that it is stabilized by membrane binding. Molecular dynamics simulations and enzyme activity measurements indicate that the C-terminal polypeptide tail facilitates catalysis by encapsulating the UDP-N-acetyl-d-mannosamine substrate, presenting three highly conserved arginine residues to the active site that are important for catalysis (R214, R221, and R224). From these data, we present a mechanistic model of catalysis that ascribes functions for these residues. This work could facilitate the development of new antimicrobial compounds that disrupt WTA biosynthesis in pathogenic bacteria.


Subject(s)
Bacterial Proteins , Glycosyltransferases , Lipoproteins , Staphylococcus aureus , Teichoic Acids , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Wall/metabolism , Glycosyltransferases/chemistry , Glycosyltransferases/metabolism , Lipoproteins/chemistry , Lipoproteins/metabolism , Staphylococcus aureus/metabolism , Substrate Specificity , Teichoic Acids/chemistry , Teichoic Acids/metabolism , Uridine Diphosphate/metabolism
6.
Cancer Cell Int ; 19: 322, 2019.
Article in English | MEDLINE | ID: mdl-31827396

ABSTRACT

BACKGROUND: Autologous tumor-infiltrating lymphocytes (Tils) immunotherapy is a promising treatment in patients with advanced hepatocellular cancer. Although Tils treatment has shown great promise, their persistence and the efficacy after adoptive-transfer are insufficient and remain a challenge. Studies have demonstrated that IL-15 and Akt inhibitor can regulate T cell differentiation and memory. Here, we constructed S-15 (Super human IL-15), a fusion protein consisting of human IL-15, the sushi domain of the IL-15 receptor α chain and human IgG-Fc. Herein we compared the effects of S-15 with IL-2 or in combination with Akti on the expansion and activation of Tils. METHODS: Hepatocellular cancer tissues were obtained from 6 patients, Tils were expanded using IL-2, IL-2/S-15, IL-2/Akti or in combination IL-2/S-15/Akti. At day 10, anti-CD3 antibody was added to the culture media and expanded to day 25. The composition, exhaustion and T-cell differentiation markers (CD45RA/CCR7) were analyzed by flow cytometry. RESULTS: We found that IL-2/S-15/Akti expanded Tils and showed the highest percentage of central memory CD45RA-CCR7+ phenotype prior to anti-CD3 antibody activation and after anti-CD3 antibody activation. T cells cultured with IL-2/S-15/Akti exhibited a mixture of CD4+, CD8+, and CD3+CD4-CD8- T cells; S-15 in combination with Akt inhibitor downregulated the expression of PD-1+Tim-3+ on Tils and decreased the Tregs in Tils. Additionally, the Tils expanded in the presence of the Akt inhibitor and S-15 showed enhanced antitumor activity as indicated by the increase in IFN-γ producing tumor infiltrating CD8+ T cells and without comprising the Tils expansion. CONCLUSION: Our study elucidates that IL-2/S-15/Akti expanded Tils and represent a viable source for the cellular therapy for patients with hepatocellular cancer.

7.
J Org Chem ; 84(11): 6697-6708, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31083938

ABSTRACT

A library of 2(a),3(a/e)-difluorosialic acids and their C-5 and/or C-9 derivatives were chemoenzymatically synthesized. Pasteurella multocida sialic acid aldolase (PmAldolase), but not its Escherichia coli homologue (EcAldolase), was found to catalyze the formation of C5-azido analogue of 3-fluoro(a)-sialic acid. In comparison, both PmAldolase and EcAldolase could catalyze the synthesis of 3-fluoro(a/e)-sialic acids and their C-9 analogues although PmAldolase was generally more efficient. The chemoenzymatically synthesized 3-fluoro(a/e)-sialic acid analogues were purified and chemically derivatized to form the desired difluorosialic acids and derivatives. Inhibition studies against several bacterial sialidases and a recombinant human cytosolic sialidase hNEU2 indicated that sialidase inhibition was affected by the C-3 fluorine stereochemistry and derivatization at C-5 and/or C-9 of the inhibitor. Opposite to that observed for influenza A virus sialidases and hNEU2, compounds with axial fluorine at C-3 were better inhibitors (up to 100-fold) against bacterial sialidases compared to their 3F-equatorial counterparts. While C-5-modified compounds were less-efficient antibacterial sialidase inhibitors, 9-N3-modified 2,3-difluoro-Neu5Ac showed increased inhibitory activity against bacterial sialidases. 9-Azido-9-deoxy-2-(e)-3-(a)-difluoro- N-acetylneuraminic acid [2(e)3(a)DFNeu5Ac9N3] was identified as an effective inhibitor with a long effective duration selectively against pathogenic bacterial sialidases from Clostridium perfringens (CpNanI) and Vibrio cholerae.


Subject(s)
Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Pasteurella multocida/enzymology , Sialic Acids/pharmacology , Carbohydrate Conformation , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Neuraminidase/metabolism , Sialic Acids/chemical synthesis , Sialic Acids/chemistry
8.
Bioorg Med Chem ; 24(8): 1696-705, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26968649

ABSTRACT

ß1-3-N-Acetylglucosaminyltransferases (ß3GlcNAcTs) and ß1-4-galactosyltransferases (ß4GalTs) have been broadly used in enzymatic synthesis of N-acetyllactosamine (LacNAc)-containing oligosaccharides and glycoconjugates including poly-LacNAc, and lacto-N-neotetraose (LNnT) found in the milk of human and other mammals. In order to explore oligosaccharides and derivatives that can be synthesized by the combination of ß3GlcNAcTs and ß4GalTs, donor substrate specificity studies of two bacterial ß3GlcNAcTs from Helicobacter pylori (Hpß3GlcNAcT) and Neisseria meningitidis (NmLgtA), respectively, using a library of 39 sugar nucleotides were carried out. The two ß3GlcNAcTs have complementary donor substrate promiscuity and 13 different trisaccharides were produced. They were used to investigate the acceptor substrate specificities of three ß4GalTs from Neisseria meningitidis (NmLgtB), Helicobacter pylori (Hpß4GalT), and bovine (Bß4GalT), respectively. Ten of the 13 trisaccharides were shown to be tolerable acceptors for at least one of these ß4GalTs. The application of NmLgtA in one-pot multienzyme (OPME) synthesis of two trisaccharides including GalNAcß1-3Galß1-4GlcßProN3 and Galß1-3Galß1-4Glc was demonstrated. The study provides important information for using these glycosyltransferases as powerful catalysts in enzymatic and chemoenzymatic syntheses of oligosaccharides and derivatives which can be useful probes and reagents.


Subject(s)
Galactosyltransferases/metabolism , Helicobacter pylori/enzymology , N-Acetylglucosaminyltransferases/metabolism , Neisseria meningitidis/enzymology , Carbohydrate Conformation , Oligosaccharides/biosynthesis , Oligosaccharides/chemistry , Substrate Specificity
9.
Chem Sci ; 7(4): 2827-2831, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-28138383

ABSTRACT

O-Sulfated sialyl Lewis x antigens play important roles in nature. However, due to their structural complexity, they are not readily accessible by either chemical or enzymatic synthetic processes. Taking advantage of a bacterial sialyltransferase mutant that can catalyze the transfer of different sialic acid forms from the corresponding sugar nucleotide donors to Lewis x antigens which are fucosylated glycans as well as an efficient one-pot multienzyme (OPME) sialylation system, O-sulfated sialyl Lewis x antigens containing different sialic acid forms and O-sulfation at different locations were systematically synthesized by chemoenzymatic methods.

10.
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
11.
Chem Commun (Camb) ; 51(22): 4595-8, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25686901

ABSTRACT

Arabidopsis thaliana glucuronokinase (AtGlcAK) was cloned and shown to be able to use various uronic acids as substrates to produce the corresponding uronic acid-1-phosphates. AtGlcAK or Bifidobacterium infantis galactokinase (BiGalK) was used with a UDP-sugar pyrophosphorylase, an inorganic pyrophosphatase, with or without a glycosyltransferase for highly efficient synthesis of UDP-uronic acids and glucuronides. These improved cost-effective one-pot multienzyme (OPME) systems avoid the use of nicotinamide adenine dinucleotide (NAD(+))-cofactor in dehydrogenase-dependent UDP-glucuronic acid production processes and can be broadly applied for synthesizing various glucuronic acid-containing molecules.


Subject(s)
Adenosine Diphosphate/biosynthesis , Glucuronides/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Uronic Acids/metabolism , Adenosine Diphosphate/chemistry , Arabidopsis/enzymology , Bifidobacterium/enzymology , Carbohydrate Conformation , Carbohydrate Sequence , Glucuronides/chemistry , Molecular Sequence Data , Uronic Acids/chemistry
12.
Carbohydr Res ; 389: 100-11, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24680514

ABSTRACT

Modifications at the glycerol side chain of sialic acid in sialosides modulate their recognition by sialic acid-binding proteins and sialidases. However, limited work has been focused on the synthesis and functional studies of sialosides with C7-modified sialic acids. Here we report chemical synthesis of C4-modified ManNAc and mannose and their application as sialic acid precursors in a highly efficient one-pot three-enzyme system for chemoenzymatic synthesis of α2-3- and α2-6-linked sialyl para-nitrophenyl galactosides in which the C7-hydroxyl group in sialic acid (N-acetylneuraminic acid, Neu5Ac, or 2-keto-3-deoxynonulosonic acid, Kdn) was systematically substituted by -F, -OMe, -H, and -N3 groups. Substrate specificity study of bacterial and human sialidases using the obtained sialoside library containing C7-modified sialic acids showed that sialosides containing C7-deoxy Neu5Ac were selective substrates for all bacterial sialidases tested but not for human NEU2. The information obtained from sialidase substrate specificity can be used to guide the design of new inhibitors that are selective against bacterial sialidases.


Subject(s)
Enzyme Assays , Neuraminidase/metabolism , Sialic Acids/chemical synthesis , Sialic Acids/metabolism , Bacteria/enzymology , Chemistry Techniques, Synthetic , Enzyme Inhibitors/pharmacology , Humans , Neuraminidase/antagonists & inhibitors , Sialic Acids/chemistry , Substrate Specificity
13.
Appl Microbiol Biotechnol ; 98(3): 1127-34, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23661084

ABSTRACT

The biological activities of heparan sulfate (HS) and heparin (HP) are closely related to their molecular structures. Both Pasteurella multocida heparosan synthase 2 (PmHS2) and Escherichia coli K5 KfiA have been used for enzymatic and chemoenzymatic synthesis of HS and HP oligosaccharides and their derivatives. We show here that cloning using the pET15b vector and expressing PmHS2 as an N-His6-tagged fusion protein improve its expression level in E. coli. Investigation of the donor substrate specificity of the N-acetylglucosaminyltransferase activities of P. multocida heparosan synthase 2 (PmHS2) and E. coli K5 KfiA indicates the substrate promiscuities of PmHS2 and KfiA. Overall, both PmHS2 and KfiA can use uridine 5'-diphosphate-N-acetylglucosamine (UDP-GlcNAc) and some of its C2'- and C6'-derivatives as donor substrates for their α1-4-GlcNAcT activities. Nevertheless, PmHS2 has a broader tolerance towards substrate modifications. Other than the UDP-sugars that can be used by KfiA, additional C6'-derivatives of UDP-GlcNAc, UDP-glucose, and UDP-N-acetylgalactosamine (UDP-GalNAc) are tolerable substrates for the α1-4-GlcNAcT activity of PmHS2. The substrate promiscuities of PmHS2 and KfiA will allow efficient chemoenzymatic synthesis of diverse HS and HP oligosaccharide derivatives which may have improved or altered activities compared to their natural counterparts.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Glycosyltransferases/metabolism , N-Acetylglucosaminyltransferases/metabolism , Pasteurella multocida/enzymology , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Gene Expression , Glycosyltransferases/genetics , N-Acetylglucosaminyltransferases/genetics , Pasteurella multocida/genetics , Substrate Specificity
14.
PLoS One ; 8(5): e63452, 2013.
Article in English | MEDLINE | ID: mdl-23700425

ABSTRACT

O-Linked ß-N-acetylglucosaminyl transferase (OGT) plays an important role in the glycosylation of proteins, which is involved in various cellular events. In human, three isoforms of OGT (short OGT [sOGT]; mitochondrial OGT [mOGT]; and nucleocytoplasmic OGT [ncOGT]) share the same catalytic domain, implying that they might adopt a similar catalytic mechanism, including sugar donor recognition. In this work, the sugar-nucleotide tolerance of sOGT was investigated. Among a series of uridine 5'-diphosphate-N-acetylglucosamine (UDP-GlcNAc) analogs tested using the casein kinase II (CKII) peptide as the sugar acceptor, four compounds could be used by sOGT, including UDP-6-deoxy-GlcNAc, UDP-GlcNPr, UDP-6-deoxy-GalNAc and UDP-4-deoxy-GlcNAc. Determined values of Km showed that the substitution of the N-acyl group, deoxy modification of C6/C4-OH or epimerization of C4-OH of the GlcNAc in UDP-GlcNAc decreased its affinity to sOGT. A molecular docking study combined with site-directed mutagenesis indicated that the backbone carbonyl oxygen of Leu653 and the hydroxyl group of Thr560 in sOGT contributed to the recognition of the sugar moiety via hydrogen bonds. The close vicinity between Met501 and the N-acyl group of GlcNPr, as well as the hydrophobic environment near Met501, were responsible for the selective binding of UDP-GlcNPr. These findings illustrate the interaction of OGT and sugar nucleotide donor, providing insights into the OGT catalytic mechanism.


Subject(s)
Acetylglucosamine/analogs & derivatives , Acetylglucosamine/chemistry , N-Acetylglucosaminyltransferases/chemistry , Amino Acid Substitution , Catalytic Domain , Humans , Hydrogen Bonding , Kinetics , Molecular Docking Simulation , Mutagenesis, Site-Directed , N-Acetylglucosaminyltransferases/genetics , Protein Binding , Substrate Specificity
15.
Org Biomol Chem ; 10(30): 6112-20, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22641268

ABSTRACT

Sialidases or neuraminidases catalyze the hydrolysis of terminal sialic acid residues from sialyl oligosaccharides and glycoconjugates. Despite successes in developing potent inhibitors specifically against influenza virus neuraminidases, the progress in designing and synthesizing selective inhibitors against bacterial and human sialidases has been slow. Guided by sialidase substrate specificity studies and sialidase crystal structural analysis, a number of 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (DANA or Neu5Ac2en) analogues with modifications at C9 or at both C5 and C9 were synthesized. Inhibition studies of various bacterial sialidases and human cytosolic sialidase NEU2 revealed that Neu5Gc9N(3)2en and Neu5AcN(3)9N(3)2en are selective inhibitors against V. cholerae sialidase and human NEU2, respectively.


Subject(s)
Cytosol/enzymology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Vibrio cholerae/enzymology , Chemistry Techniques, Synthetic , Drug Design , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Neuraminic Acids/chemical synthesis , Neuraminic Acids/chemistry , Neuraminic Acids/pharmacology , Neuraminidase/chemistry , Protein Conformation
16.
Chem Commun (Camb) ; 48(27): 3357-9, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22361713

ABSTRACT

A library of α2-3- and α2-6-linked sialyl galactosides containing C9-modified sialic acids was synthesized from C6-modified mannose derivatives using an efficient one-pot three-enzyme system. These sialosides were used in a high-throughput sialidase substrate specificity assay to elucidate the importance of C9-OH in sialidase recognition.


Subject(s)
Bacterial Proteins/chemistry , Galactosides/chemistry , Neuraminidase/chemistry , Sialic Acids/chemistry , Sialyltransferases/chemistry , High-Throughput Screening Assays , Humans , Mannose/analogs & derivatives , Mannose/chemistry , Substrate Specificity
17.
Chem Commun (Camb) ; 48(21): 2728-30, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22306833

ABSTRACT

A promiscuous UDP-sugar pyrophosphorylase (BLUSP) was cloned from Bifidobacterium longum strain ATCC55813 and used efficiently with a Pasteurella multocida inorganic pyrophosphatase (PmPpA) with or without a monosaccharide 1-kinase for one-pot multienzyme synthesis of UDP-galactose, UDP-glucose, UDP-mannose, and their derivatives. Further chemical diversification of a UDP-mannose derivative resulted in the formation of UDP-N-acetylmannosamine.


Subject(s)
Bifidobacterium/enzymology , UTP-Glucose-1-Phosphate Uridylyltransferase/metabolism , Uridine Diphosphate Sugars/biosynthesis , Galactokinase/metabolism , Inorganic Pyrophosphatase/metabolism , Pasteurella multocida/enzymology , Uridine Diphosphate Galactose/biosynthesis , Uridine Diphosphate Galactose/chemistry , Uridine Diphosphate Glucose/biosynthesis , Uridine Diphosphate Glucose/chemistry , Uridine Diphosphate Sugars/chemistry
18.
Chem Commun (Camb) ; 46(33): 6066-8, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20625591

ABSTRACT

Two bacterial beta1-4-galactosyltransferases, NmLgtB and Hp1-4GalT, exhibit promiscuous and complementary acceptor substrate specificity. They have been used in an efficient one-pot multienzyme system to synthesize LacNAc, lactose, and their derivatives including those containing negatively charged 6-O-sulfated GlcNAc and C2-substituted GlcNAc or Glc, from monosaccharide derivatives and inexpensive Glc-1-P.


Subject(s)
Galactosides/chemical synthesis , Helicobacter pylori/enzymology , N-Acetyllactosamine Synthase/chemistry , Neisseria meningitidis/enzymology , Pasteurella multocida/enzymology , Recombinant Fusion Proteins/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Cloning, Molecular , Escherichia coli/genetics , Galactosides/chemistry , N-Acetyllactosamine Synthase/genetics , Recombinant Fusion Proteins/genetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL