1
|
Yoon D, Moon JH, Cho A, Boo H, Cha JS, Lee Y, Yoo J. Structure-Based Insight on the Mechanism of N-Glycosylation Inhibition by Tunicamycin. Mol Cells 2023; 46:337-344. [PMID: 37190766 PMCID: PMC10258461 DOI: 10.14348/molcells.2023.0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 05/17/2023] Open
Abstract
N-glycosylation, a common post-translational modification, is widely acknowledged to have a significant effect on protein stability and folding. N-glycosylation is a complex process that occurs in the endoplasmic reticulum (ER) and requires the participation of multiple enzymes. GlcNAc-1-P-transferase (GPT) is essential for initiating N-glycosylation in the ER. Tunicamycin is a natural product that inhibits N-glycosylation and produces ER stress, and thus it is utilized in research. The molecular mechanism by which GPT triggers N-glycosylation is discussed in this review based on the GPT structure. Based on the structure of the GPT-tunicamycin complex, we also discuss how tunicamycin reduces GPT activity, which prevents N-glycosylation. This review will be highly useful for understanding the role of GPT in the N-glycosylation of proteins, as well as presents a potential for considering tunicamycin as an antibiotic treatment.
Collapse
Affiliation(s)
- Danbi Yoon
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Ju Heun Moon
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Anna Cho
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Hyejoon Boo
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Jeong Seok Cha
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Jiho Yoo
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
2
|
Selected heterocycles for the development of pyrophosphate mimics. Future Med Chem 2022; 14:1219-1222. [PMID: 35861044 DOI: 10.4155/fmc-2022-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
3
|
Dalton E, Morris Z, Ayres N. Synthesis and characterization of sulfated-lactose polyurethane hydrogels. Polym Chem 2022. [DOI: 10.1039/d2py00227b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Polyurethanes (PUs) are widely used due to their durability, flexibility, and biocompatibility. PU hydrogels have been used in biomedical applications tissue engineering, synthetic extracellular matrices, and drug delivery. In this...
Collapse
|
4
|
Prasher P, Sharma M. Medicinal chemistry of pyrophosphate mimics: A mini review. Drug Dev Res 2021; 83:3-15. [PMID: 34506652 DOI: 10.1002/ddr.21877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/21/2022]
Abstract
The pyrophosphate mimicking groups offer rational modification of the pyrophosphate-bearing natural substrates of the overexpressed enzymes that cause the onset of disease progression. Mainly, the modified substrate interacts differently with the enzyme active site eventually causing its deactivation, or provides the therapeutically active products at the completion of the catalytic cycle that contribute toward the inhibition of the target enzyme. Many of the pyrophosphate mimic-containing molecules serve as competitive or allosteric inhibitors of the target enzyme to achieve the desirable properties for the mitigation of the target enzyme's pathophysiology. This review presents an epigrammatic overview of the pyrophosphate mimics in medicinal chemistry.
Collapse
Affiliation(s)
- Parteek Prasher
- UGC Sponsored Centre for Advanced Studies, Department of Chemistry, Guru Nanak Dev University, Amritsar, India.,Department of Chemistry, University of Petroleum & Energy Studies, Dehradun, India
| | - Mousmee Sharma
- UGC Sponsored Centre for Advanced Studies, Department of Chemistry, Guru Nanak Dev University, Amritsar, India.,Department of Chemistry, Uttaranchal University, Dehradun, India
| |
Collapse
|
5
|
Abstract
Iminosugars are naturally occurring carbohydrate analogues known since 1967. These natural compounds and hundreds of their synthetic derivatives prepared over five decades have been mainly exploited to inhibit the glycosidases, the enzymes catalysing the glycosidic bond cleavage, in order to find new drugs for the treatment of type 2 diabetes and other diseases. However, iminosugars are also inhibitors of glycosyltransferases, the enzymes responsible for the synthesis of oligosaccharides and glycoconjugates. The selective inhibition of specific glycosyltransferases involved in cancer or bacterial infections could lead to innovative therapeutic agents. The synthesis and biological properties of all the iminosugars assayed to date as glycosyltransferase inhibitors are reviewed in the present article.
Collapse
Affiliation(s)
- Irene Conforti
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Ecole Nationale Supérieure de Chimie de Montpellier, 8 Rue de l'Ecole Normale, 34296 Montpellier cedex 5, France.
| | - Alberto Marra
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, Ecole Nationale Supérieure de Chimie de Montpellier, 8 Rue de l'Ecole Normale, 34296 Montpellier cedex 5, France.
| |
Collapse
|
6
|
Synthetic Route to Glycosyl β-1C-(phosphino)-phosphonates as Unprecedented Stable Glycosyl Diphosphate Analogs and Their Preliminary Biological Evaluation. Molecules 2020; 25:molecules25214969. [PMID: 33121078 PMCID: PMC7663146 DOI: 10.3390/molecules25214969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 11/16/2022] Open
Abstract
The synthesis of glycosyl-β-1C-(phosphino)-phosphonates is a challenge since it has not yet been described. In this paper, we report an innovative synthetic method for their preparation from Glc-, Man-, and GlcNAc- lactone derivatives. The proposed original strategy involves the addition of the corresponding δ-hexonolactones onto the dianion of (methylphosphino) phosphonate as a key step, followed by dehydration and stereoselective addition of dihydrogen on the resulting double bond. Final deprotection provides the new glycosyl diphosphate analogs in 35%, 36%, and 10% yield over 6 steps from the corresponding δ-hexonolactones. The synthetized compounds were evaluated as inhibitors of phosphatase and diphosphatase activities and found to have complex concentration-dependent activatory and inhibitory properties on alkaline phosphatase. The synthetized tools should be useful to study other enzymes such as transferases.
Collapse
|
7
|
Prasher P, Sharma M. Targeting N-acetylgalactosaminyltransferase for anticancer therapy. Drug Dev Res 2020; 82:3-6. [PMID: 32985012 DOI: 10.1002/ddr.21744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/26/2020] [Accepted: 09/18/2020] [Indexed: 11/12/2022]
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, UGC Sponsored Centre for Advanced Studies, Guru Nanak Dev University, Amritsar, India.,Department of Chemistry, University of Petroleum and Energy Studies, Dehradun, India
| | - Mousmee Sharma
- Department of Chemistry, UGC Sponsored Centre for Advanced Studies, Guru Nanak Dev University, Amritsar, India.,Department of Chemistry, Uttaranchal University, Dehradun, India
| |
Collapse
|
8
|
N-acetylgalactosaminyltransferase: a potential target for colorectal adenocarcinoma. Future Med Chem 2020; 12:1529-1531. [DOI: 10.4155/fmc-2020-0137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
9
|
Mashalidis EH, Lee SY. Structures of Bacterial MraY and Human GPT Provide Insights into Rational Antibiotic Design. J Mol Biol 2020; 432:4946-4963. [PMID: 32199982 DOI: 10.1016/j.jmb.2020.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/07/2020] [Accepted: 03/12/2020] [Indexed: 12/24/2022]
Abstract
The widespread emergence of antibiotic resistance in pathogens necessitates the development of antibacterial agents inhibiting underexplored targets in bacterial metabolism. One such target is phospho-MurNAc-pentapeptide translocase (MraY), an essential integral membrane enzyme that catalyzes the first committed step of peptidoglycan biosynthesis. MraY has long been considered a promising candidate for antibiotic development in part because it is the target of five classes of naturally occurring nucleoside inhibitors with potent in vivo and in vitro antibacterial activity. Although these inhibitors each have a nucleoside moiety, they vary dramatically in their core structures, and they have different activity properties. Until recently, the structural basis of MraY inhibition was poorly understood. Several recent structures of MraY and its human paralog, GlcNAc-1-P-transferase, have provided insights into MraY inhibition that are consistent with known inhibitor activity data and can inform rational drug design for this important antibiotic target.
Collapse
Affiliation(s)
- Ellene H Mashalidis
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive,Durham, NC 27710, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive,Durham, NC 27710, USA.
| |
Collapse
|
10
|
Moukha-Chafiq O, Reynolds RC, Wilson JC, Snowden TS. Parallel Solution Phase Synthesis and Preliminary Biological Activity of a 5'-Substituted Cytidine Analog Library. ACS COMBINATORIAL SCIENCE 2019; 21:628-634. [PMID: 31365223 DOI: 10.1021/acscombsci.9b00072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A 109-membered library of 5'-substituted cytidine analogs was synthesized, via funding through the NIH Roadmap Initiative and the Pilot Scale Library (PSL) Program. Reaction core compounds contained -NH2 (2) and -COOH (44 and 93) groups that were coupled to a diversity of reactants in a parallel, solution phase format to produce the target library. The assorted reactants included -NH2, -CHO, -SO2Cl, and -COOH functional groups, and condensation with the intermediate core materials 2 and 44 followed by acidic hydrolysis produced 3-91 in good yields and high purity. Linkage of the amino terminus of d-phenylalanine methyl ester to the free 5'-COOH of 44 and NaOH treatment led to core library -COOH precursor 93. In a libraries from libraries approach, compound 93 served as the vital building block for our unique library of dipeptidyl cytidine analogs 94-114 through amide coupling of the -COOH group with numerous commercial amines followed by acidic deprotection. Initial screening of the complete final library through the MLPCN program revealed a modest number of hits over diverse biological processes. These hits might be considered as starting points for hit-to-lead optimization and development studies.
Collapse
Affiliation(s)
- Omar Moukha-Chafiq
- Chemistry Department, Drug Discovery Division, Southern Research, 2000 Ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Robert C. Reynolds
- Department of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, NP 2540 J, 1720 Second Avenue South, Birmingham, Alabama 35294-3300, United States
| | - Jacob C. Wilson
- Department of Chemistry and Biochemistry, The University of Alabama, 250 Hackberry Lane, Tuscaloosa, Alabama 35487-0336, United States
| | - Timothy S. Snowden
- Department of Chemistry and Biochemistry, The University of Alabama, 250 Hackberry Lane, Tuscaloosa, Alabama 35487-0336, United States
| |
Collapse
|
11
|
Nakamura H, Tsukano C, Yoshida T, Yasui M, Yokouchi S, Kobayashi Y, Igarashi M, Takemoto Y. Total Synthesis of Caprazamycin A: Practical and Scalable Synthesis of syn-β-Hydroxyamino Acids and Introduction of a Fatty Acid Side Chain to 1,4-Diazepanone. J Am Chem Soc 2019; 141:8527-8540. [PMID: 31067040 DOI: 10.1021/jacs.9b02220] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The first total synthesis of caprazamycin A (1), a representative liponucleoside antibiotic, is described. Diastereoselective aldol reactions of aldehydes 12 and 25-27, derived from uridine, with diethyl isocyanomalonate 13 and phenylcarbamate 21 were investigated using thiourea catalysts 14 or bases to synthesize syn-β-hydroxyamino acid derivatives. The 1,4-diazepanone core of 1 was constructed using a Mitsunobu reaction, and the fatty acid side chain was introduced using a stepwise sequence based on model studies. Notably, global deprotection was realized using palladium black and formic acid without hydrogenating the olefin in the uridine unit.
Collapse
Affiliation(s)
- Hugh Nakamura
- Graduate School of Pharmaceutical Sciences , Kyoto University , Yoshida, Sakyo-ku, Kyoto 606-8501 , Japan
| | - Chihiro Tsukano
- Graduate School of Pharmaceutical Sciences , Kyoto University , Yoshida, Sakyo-ku, Kyoto 606-8501 , Japan
| | - Takuma Yoshida
- Graduate School of Pharmaceutical Sciences , Kyoto University , Yoshida, Sakyo-ku, Kyoto 606-8501 , Japan
| | - Motohiro Yasui
- Graduate School of Pharmaceutical Sciences , Kyoto University , Yoshida, Sakyo-ku, Kyoto 606-8501 , Japan
| | - Shinsuke Yokouchi
- Graduate School of Pharmaceutical Sciences , Kyoto University , Yoshida, Sakyo-ku, Kyoto 606-8501 , Japan
| | - Yusuke Kobayashi
- Graduate School of Pharmaceutical Sciences , Kyoto University , Yoshida, Sakyo-ku, Kyoto 606-8501 , Japan
| | - Masayuki Igarashi
- Institute of Microbial Chemistry (BIKAKEN), Tokyo , 3-14-23 Kamiosaki , Shinagawa-ku, Tokyo 141-0021 , Japan
| | - Yoshiji Takemoto
- Graduate School of Pharmaceutical Sciences , Kyoto University , Yoshida, Sakyo-ku, Kyoto 606-8501 , Japan
| |
Collapse
|
12
|
Madec AGE, Schocker NS, Sanchini S, Myratgeldiyev G, Das D, Imperiali B. Facile Solid-Phase Synthesis and Assessment of Nucleoside Analogs as Inhibitors of Bacterial UDP-Sugar Processing Enzymes. ACS Chem Biol 2018; 13:2542-2550. [PMID: 30080379 DOI: 10.1021/acschembio.8b00477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The privileged uptake of nucleosides into cells has generated interest in the development of nucleoside-analog libraries for mining new inhibitors. Of particular interest are applications in the discovery of substrate mimetic inhibitors for the growing number of identified glycan-processing enzymes in bacterial pathogens. However, the high polarity and the need for appropriate protecting group strategies for nucleosides challenges the development of synthetic approaches. Here, we report an accessible, user-friendly synthesis that branches from a common solid phase-immobilized uridinyl-amine intermediate, which can be used as a starting point for diversity-oriented synthesis. We demonstrate the generation of five series of uridinyl nucleoside analogs for investigating inhibitor structure-activity relationships. This library was screened for inhibition of representative enzymes from three functional families including a phosphoglycosyl transferase, a UDP-aminosugar acetyltransferase, and a glycosyltransferase. These candidates were taken from the Gram-negative bacteria Campylobacter concisus and Campylobacter jejuni and the Gram-positive bacterium Clostridium difficile, respectively. Inhibition studies show that specific compound series preferentially inhibit selected enzymes, with IC50 values ranging from 35 ± 7 μM to 174 ± 21 μM. Insights from the screen provide a strong foundation for further structural elaboration, to improve potency, which will be enabled by the same synthetic strategy. The solid-phase strategy was also used to synthesize pseudouridine analogs of lead compounds. Finally, the compounds were found to be nontoxic to mammalian cells, further supporting the opportunities for future development.
Collapse
Affiliation(s)
- Amaël G. E. Madec
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Nathaniel S. Schocker
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Silvano Sanchini
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Gadam Myratgeldiyev
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Debasis Das
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Barbara Imperiali
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
13
|
Komor R, Pastuch-Gawolek G, Krol E, Szeja W. Synthesis and Preliminary Evaluation of Biological Activity of Glycoconjugates Analogues of Acyclic Uridine Derivatives. Molecules 2018; 23:molecules23082017. [PMID: 30104510 PMCID: PMC6222857 DOI: 10.3390/molecules23082017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/09/2018] [Accepted: 08/11/2018] [Indexed: 12/16/2022] Open
Abstract
Herein we present the methodology for obtaining glycosyltransferase inhibitors, analogues of natural enzyme substrates of donor-type: UDP-glucose and UDP-galactose. The synthesis concerned glycoconjugates, nucleoside analogues containing an acyclic ribose mimetic linked to a uracil moiety in their structure. The biological activity of the synthesised compounds was determined on the basis of their ability to inhibit the model enzyme action of β-1,4-galactosyltransferase from bovine milk. The obtained results allowed to expand and supplement the existing library of synthetic compounds that are able to regulate the biological activity of enzymes from the GT class.
Collapse
Affiliation(s)
- Roman Komor
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland.
| | - Gabriela Pastuch-Gawolek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland.
- Biotechnology Center, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland.
| | - Ewelina Krol
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland.
| | - Wieslaw Szeja
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland.
| |
Collapse
|
14
|
Krol E, Pastuch-Gawolek G, Chaubey B, Brzuska G, Erfurt K, Szewczyk B. Novel Uridine Glycoconjugates, Derivatives of 4-Aminophenyl 1-Thioglycosides, as Potential Antiviral Compounds. Molecules 2018; 23:molecules23061435. [PMID: 29899276 PMCID: PMC6100568 DOI: 10.3390/molecules23061435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/02/2018] [Accepted: 06/10/2018] [Indexed: 01/19/2023] Open
Abstract
A novel series of uridine glycoconjugates, derivatives of 4-aminophenyl 1-thioglycosides, was designed and synthesized. All compounds were evaluated in vitro for their antiviral activity against hepatitis C virus (HCV) and classical swine fever virus (CSFV), two important human and animal viral pathogens for which new or improved therapeutic options are needed. The antiviral activity of all synthesized compounds was confirmed using pseudo-plaque reduction assays in which a significant arrest of CSFV and HCV growth was observed in the presence of these compounds. Two of the synthesized compounds, 9 and 12, displayed a significant inhibitory effect on HCV and CSFV propagation with IC50 values of 4.9 and 13.5 µM for HCV and 4.2 and 4 µM for CSFV, respectively, with low cytotoxicity. Using various infection and replication models, we have shown that both compounds were able to significantly reduce viral genome replication by up to 90% with IC50 values in the low micromolar range. A structure activity analysis of the synthesized compounds showed that the high antiviral activity was attributed to the hydrophobicity of glycoconjugates and the introduction of elements capable to coordinate metal ions into the spacer connecting the sugar and uridine moiety, which can be useful in the development of new antiviral compounds in the future.
Collapse
Affiliation(s)
- Ewelina Krol
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland.
| | - Gabriela Pastuch-Gawolek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland.
- Biotechnology Center, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland.
| | - Binay Chaubey
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland.
- Functional Genomics Lab., Centre for Advanced Study, Department of Botany, University of Calcutta, 35, Ballygunge Circular Road, 700019 Kolkata, India.
| | - Gabriela Brzuska
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland.
| | - Karol Erfurt
- Department of Chemical Organic Technology and Petrochemistry, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland.
| | - Boguslaw Szewczyk
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland.
| |
Collapse
|
15
|
Yoo J, Mashalidis EH, Kuk ACY, Yamamoto K, Kaeser B, Ichikawa S, Lee SY. GlcNAc-1-P-transferase-tunicamycin complex structure reveals basis for inhibition of N-glycosylation. Nat Struct Mol Biol 2018; 25:217-224. [PMID: 29459785 PMCID: PMC5840018 DOI: 10.1038/s41594-018-0031-y] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/17/2018] [Indexed: 12/19/2022]
Abstract
N-linked glycosylation is a predominant post-translational modification of protein in eukaryotes, and its dysregulation is the etiology of several human disorders. The enzyme UDP-N-acetylglucosamine:dolichyl-phosphate N-acetylglucosaminephosphotransferase (GlcNAc-1-P-transferase, GPT) catalyzes the first and committed step of N-linked glycosylation in the endoplasmic reticulum membrane, and it is the target of the natural product tunicamycin. Tunicamycin has potent antibacterial activity by inhibiting the bacterial cell wall synthesis enzyme MraY, but its usefulness as an antibiotic is limited by off-target inhibition of human GPT. Our understanding of how tunicamycin inhibits N-linked glycosylation and efforts to selectively target MraY are hampered by a lack of structural information. Here we present crystal structures of human GPT in complex with tunicamycin. Our structural and functional analyses reveal the difference between GPT and MraY in their mechanisms of inhibition by tunicamycin. We demonstrate that this difference could be exploited for the design of MraY-specific inhibitors as potential antibiotics.
Collapse
Affiliation(s)
- Jiho Yoo
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | | | - Alvin C Y Kuk
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Kazuki Yamamoto
- Faculty of Pharmaceutical Science, Hokkaido University, Sapporo, Japan
| | - Benjamin Kaeser
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Satoshi Ichikawa
- Faculty of Pharmaceutical Science, Hokkaido University, Sapporo, Japan
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
16
|
Affiliation(s)
- Kazuki Yamamoto
- Faculty
of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Fumika Yakushiji
- Faculty
of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
- Center
for Research and Education on Drug Discovery, Faculty of Pharmaceutical
Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Takanori Matsumaru
- Faculty
of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
- Center
for Research and Education on Drug Discovery, Faculty of Pharmaceutical
Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Satoshi Ichikawa
- Faculty
of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
- Center
for Research and Education on Drug Discovery, Faculty of Pharmaceutical
Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
17
|
Malkowski SN, Dishuck CF, Lamanilao GG, Embry CP, Grubb CS, Cafiero M, Peterson LW. Design, Modeling and Synthesis of 1,2,3-Triazole-Linked Nucleoside-Amino Acid Conjugates as Potential Antibacterial Agents. Molecules 2017; 22:molecules22101682. [PMID: 28994722 PMCID: PMC6151744 DOI: 10.3390/molecules22101682] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 09/27/2017] [Accepted: 10/03/2017] [Indexed: 02/01/2023] Open
Abstract
Copper-catalyzed azide-alkyne cycloadditions (CuAAC or click chemistry) are convenient methods to easily couple various pharmacophores or bioactive molecules. A new series of 1,2,3-triazole-linked nucleoside-amino acid conjugates have been designed and synthesized in 57–76% yields using CuAAC. The azido group was introduced on the 5′-position of uridine or the acyclic analogue using the tosyl-azide exchange method and alkylated serine or proparylglycine was the alkyne. Modeling studies of the conjugates in the active site of LpxC indicate they have promise as antibacterial agents.
Collapse
Affiliation(s)
- Sarah N Malkowski
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Carolyn F Dishuck
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Gene G Lamanilao
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Carter P Embry
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Christopher S Grubb
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Mauricio Cafiero
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Larryn W Peterson
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| |
Collapse
|
18
|
Pastuch-Gawolek G, Plesniak M, Komor R, Byczek-Wyrostek A, Erfurt K, Szeja W. Synthesis and preliminary biological assay of uridine glycoconjugate derivatives containing amide and/or 1,2,3-triazole linkers. Bioorg Chem 2017; 72:80-88. [DOI: 10.1016/j.bioorg.2017.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 11/16/2022]
|
19
|
Pastuch-Gawolek G, Chaubey B, Szewczyk B, Krol E. Novel thioglycosyl analogs of glycosyltransferase substrates as antiviral compounds against classical swine fever virus and hepatitis C virus. Eur J Med Chem 2017; 137:247-262. [PMID: 28601004 DOI: 10.1016/j.ejmech.2017.05.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/29/2017] [Accepted: 05/24/2017] [Indexed: 12/15/2022]
Abstract
Hepatitis C virus (HCV) and classical swine fever virus (CSFV) are important pathogens for which new therapeutic approaches are in high demand. Herein, we report the synthesis of newly designed thioglycosyl analogs of glycosyltransferase substrates which were evaluated using cell-based assays for cytotoxicity and antiviral activity against both viruses. The antiviral activity of synthesized compounds against CSFV and HCV was confirmed using pseudo-plaque reduction assays where a significant arrest of viral growth was observed in the presence of selected compounds. We showed that compounds 13 and 14 exerted the most significant inhibitory effect on in vitro CSFV and HCV infections in the series. Glycoconjugates 13 and 14 not only inhibited both viral propagation with IC50 values in low micromolar range, but efficiently suppressed the production of viral proteins in a dose-dependent manner. In addition, studies using in vitro HCV infection and replication models have shown that both compounds are able to significantly reduce viral genomic replication. We demonstrated that compounds 13 and 14 showed a strong inhibition, up to 90% of replication which inscribe them in the promising alternative approach for the development of new anti-CSFV and anti-HCV drugs.
Collapse
Affiliation(s)
- Gabriela Pastuch-Gawolek
- Silesian University of Technology, Faculty of Chemistry, Chair of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Krzywoustego 4, 44-100 Gliwice, Poland; Biotechnology Center, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland
| | - Binay Chaubey
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland; Functional Genomics Lab., Centre for Advanced Study, Department of Botany, University of Calcutta, 35, Ballygunge Circular Road, 700019 Kolkata, India
| | - Boguslaw Szewczyk
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland
| | - Ewelina Krol
- Department of Recombinant Vaccines, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland.
| |
Collapse
|
20
|
Merino P, Delso I, Tejero T, Ghirardello M, Juste-Navarro V. Nucleoside Diphosphate Sugar Analogues that Target Glycosyltransferases. ASIAN J ORG CHEM 2016. [DOI: 10.1002/ajoc.201600396] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Pedro Merino
- Department of Synthesis and Structure of Biomolecules; Institute of Chemical Synthesis and Homogeneous Catalysis (ISQCH); University of Zaragoza, CSIC; Zaragoza, Aragón 50009 Spain
| | - Ignacio Delso
- NMR Service, Center of Chemistry and Materials of Aragon (CEQMA); University of Zaragoza, CSIC; Zaragoza, Aragón 50009 Spain
| | - Tomás Tejero
- Department of Synthesis and Structure of Biomolecules; Institute of Chemical Synthesis and Homogeneous Catalysis (ISQCH); University of Zaragoza, CSIC; Zaragoza, Aragón 50009 Spain
| | - Mattia Ghirardello
- Department of Synthesis and Structure of Biomolecules; Institute of Chemical Synthesis and Homogeneous Catalysis (ISQCH); University of Zaragoza, CSIC; Zaragoza, Aragón 50009 Spain
| | - Verónica Juste-Navarro
- Department of Synthesis and Structure of Biomolecules; Institute of Chemical Synthesis and Homogeneous Catalysis (ISQCH); University of Zaragoza, CSIC; Zaragoza, Aragón 50009 Spain
| |
Collapse
|
21
|
Serpi M, Ferrari V, Pertusati F. Nucleoside Derived Antibiotics to Fight Microbial Drug Resistance: New Utilities for an Established Class of Drugs? J Med Chem 2016; 59:10343-10382. [PMID: 27607900 DOI: 10.1021/acs.jmedchem.6b00325] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Novel antibiotics are urgently needed to combat the rise of infections due to drug-resistant microorganisms. Numerous natural nucleosides and their synthetically modified analogues have been reported to have moderate to good antibiotic activity against different bacterial and fungal strains. Nucleoside-based compounds target several crucial processes of bacterial and fungal cells such as nucleoside metabolism and cell wall, nucleic acid, and protein biosynthesis. Nucleoside analogues have also been shown to target many other bacterial and fungal cellular processes although these are not well characterized and may therefore represent opportunities to discover new drugs with unique mechanisms of action. In this Perspective, we demonstrate that nucleoside analogues, cornerstones of anticancer and antiviral treatments, also have great potential to be repurposed as antibiotics so that an old drug can learn new tricks.
Collapse
Affiliation(s)
- Michaela Serpi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University , Redwood Building, King Edward VII Avenue, CF10 3NB Cardiff, United Kingdom
| | - Valentina Ferrari
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University , Redwood Building, King Edward VII Avenue, CF10 3NB Cardiff, United Kingdom
| | - Fabrizio Pertusati
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University , Redwood Building, King Edward VII Avenue, CF10 3NB Cardiff, United Kingdom
| |
Collapse
|
22
|
Structural Investigation of Park's Nucleotide on Bacterial Translocase MraY: Discovery of Unexpected MraY Inhibitors. Sci Rep 2016; 6:31579. [PMID: 27531195 PMCID: PMC4987650 DOI: 10.1038/srep31579] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/26/2016] [Indexed: 11/17/2022] Open
Abstract
Systematic structural modifications of the muramic acid, peptide, and nucleotide moieties of Park’s nucleotide were performed to investigate the substrate specificity of B. subtilis MraY (MraYBS). It was found that the simplest analogue of Park’s nucleotide only bearing the first two amino acids, l-alanine-iso-d-glutamic acid, could function as a MraYBS substrate. Also, the acid group attached to the Cα of iso-d-glutamic acid was found to play an important role for substrate activity. Epimerization of the C4-hydroxyl group of muramic acid and modification at the 5-position of the uracil in Park’s nucleotide were both found to dramatically impair their substrate activity. Unexpectedly, structural modifications on the uracil moiety changed the parent molecule from a substrate to an inhibitor, blocking the MraYBS translocation. One unoptimized inhibitor was found to have a Ki value of 4 ± 1 μM against MraYBS, more potent than tunicamycins.
Collapse
|
23
|
Synthesis and Evaluation of Bicyclo[3.1.0]hexane-Based UDP-Galf Analogues as Inhibitors of the Mycobacterial Galactofuranosyltransferase GlfT2. Molecules 2016; 21:molecules21081053. [PMID: 27529206 PMCID: PMC6272867 DOI: 10.3390/molecules21081053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/05/2016] [Accepted: 08/06/2016] [Indexed: 11/17/2022] Open
Abstract
UDP-galactofuranose (UDP-Galf) is the donor substrate for both bifunctional galactofuranosyltransferases, GlfT1 and GlfT2, which are involved in the biosynthesis of mycobacterial galactan. In this paper, a group of UDP-Galf mimics were synthesized via reductive amination of a bicyclo[3.1.0]hexane-based amine by reacting with aromatic, linear, or uridine-containing aldehydes. These compounds were evaluated against GlfT2 using a coupled spectrophotometric assay, and were shown to be weak inhibitors of the enzyme.
Collapse
|
24
|
Huang Y, Chai Q, Warmin MR, Ayres N. Lactose‐containing hydrogels for enzyme stabilization. ACTA ACUST UNITED AC 2016. [DOI: 10.1002/pola.28127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yongshun Huang
- Department of Chemistrythe University of CincinnatiP.O. Box 210172Cincinnati Ohio45221
| | - Qinyuan Chai
- Department of Chemistrythe University of CincinnatiP.O. Box 210172Cincinnati Ohio45221
| | - Mary R. Warmin
- Department of Chemistrythe University of CincinnatiP.O. Box 210172Cincinnati Ohio45221
| | - Neil Ayres
- Department of Chemistrythe University of CincinnatiP.O. Box 210172Cincinnati Ohio45221
| |
Collapse
|
25
|
Structural insights into inhibition of lipid I production in bacterial cell wall synthesis. Nature 2016; 533:557-560. [PMID: 27088606 PMCID: PMC4882255 DOI: 10.1038/nature17636] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 03/01/2016] [Indexed: 12/30/2022]
Abstract
Antibiotic-resistant bacterial infection is a serious threat to public health. Peptidoglycan biosynthesis is a well-established target for antibiotic development. MraY (phospho-MurNAc-pentapeptide translocase) catalyzes the first and an essential membrane step of peptidoglycan biosynthesis. It is considered a very promising target for the development of new antibiotics, as many naturally occuring nucleoside inhibitors with antibacterial activity target this enzyme1-4. However, antibiotics targeting MraY have not been developed for clinical use mainly due to a lack of structural insight into inhibition of this enzyme. Here we present the crystal structure of MraY from Aquifex aeolicus (MraYAA) in complex with its naturally occurring inhibitor, muraymycin D2 (MD2). Upon binding MD2, MraYAA undergoes remarkably large conformational rearrangements near the active site, which lead to the formation of a nucleoside-binding pocket and a peptide-binding site. MD2 binds the nucleoside-binding pocket like a two-pronged plug inserting into a socket. Additional interactions it makes in the adjacent peptide-binding site anchor MD2 to and enhance its affinity for MraYAA. Surprisingly, MD2 does not interact with three acidic residues or the Mg2+ cofactor required for catalysis, suggesting that MD2 binds to MraYAA in a manner that overlaps with, but is distinct from its natural substrate, UDP-MurNAc-pentapeptide. We have deciphered the chemical logic of MD2 binding to MraYAA, including how it avoids the need for pyrophosphate and sugar moieties, which are essential features for substrate binding. The conformational plasticity of MraY could be the reason that it is the target of many structurally distinct inhibitors. These findings can inform the design of new inhibitors targeting MraY as well as its paralogs, WecA and TarO.
Collapse
|
26
|
Walvoort MTC, Lukose V, Imperiali B. A Modular Approach to Phosphoglycosyltransferase Inhibitors Inspired by Nucleoside Antibiotics. Chemistry 2015; 22:3856-64. [PMID: 26662170 DOI: 10.1002/chem.201503986] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Indexed: 11/09/2022]
Abstract
Phosphoglycosyltransferases (PGTs) represent "gatekeeper" enzymes in complex glycan assembly pathways by catalyzing transfer of a phosphosugar from an activated nucleotide diphosphosugar to a membrane-resident polyprenol phosphate. The unique structures of selected nucleoside antibiotics, such as tunicamycin and mureidomycin A, which are known to inhibit comparable biochemical transformations, are exploited as the foundation for the development of modular synthetic inhibitors of PGTs. Herein we present the design, synthesis, and biochemical evaluation of two readily manipulatable modular scaffolds as inhibitors of monotopic bacterial PGTs. Selected compounds show IC50 values down to the 40 μm range, thereby serving as lead compounds for future development of selective and effective inhibitors of diverse PGTs of biological and medicinal interest.
Collapse
Affiliation(s)
- Marthe T C Walvoort
- Departments of Chemistry and Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Vinita Lukose
- Departments of Chemistry and Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Barbara Imperiali
- Departments of Chemistry and Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
27
|
Bella M, Koóš M, Lin CH. Towards inhibitors of glycosyltransferases: A novel approach to the synthesis of 3-acetamido-3-deoxy-D-psicofuranose derivatives. Beilstein J Org Chem 2015; 11:1547-52. [PMID: 26425214 PMCID: PMC4578356 DOI: 10.3762/bjoc.11.170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/20/2015] [Indexed: 12/16/2022] Open
Abstract
A novel synthetic strategy leading to 3-acetamido-3-deoxy-D-psicofuranose 9 is presented. The latter compound, after some manipulations, was transformed into fully protected 3-acetamido-3-deoxy-D-psicofuranose 11 as a potential substrate for the synthesis of N-acetylglucosaminyltransferase inhibitors designed by computational methods. After the attempted thioglycosylation of 11 with EtSH in the presence of BF3·OEt2, 2-methyloxazoline derivatives 13 and 14 were isolated.
Collapse
Affiliation(s)
- Maroš Bella
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, SK-845 38, Bratislava, Slovakia
| | - Miroslav Koóš
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, SK-845 38, Bratislava, Slovakia
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Sec. 2, Taipei 11529, Taiwan
| |
Collapse
|
28
|
Wang S, Cuesta-Seijo JA, Striebeck A, Lafont D, Palcic MM, Vidal S. Design of Glycosyltransferase Inhibitors: Serine Analogues as Pyrophosphate Surrogates? Chempluschem 2015; 80:1525-1532. [DOI: 10.1002/cplu.201500282] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Indexed: 01/07/2023]
Affiliation(s)
- Shuai Wang
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires; Laboratoire de Chimie Organique 2-Glycochimie, UMR 5246; CNRS and Université Claude Bernard Lyon 1; 43 Boulevard du 11 Novembre 1918 69622 Villeurbanne France
| | | | | | - Dominique Lafont
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires; Laboratoire de Chimie Organique 2-Glycochimie, UMR 5246; CNRS and Université Claude Bernard Lyon 1; 43 Boulevard du 11 Novembre 1918 69622 Villeurbanne France
| | - Monica M. Palcic
- Carlsberg Laboratory; Gamle Carlsberg Vej 10 1799 Copenhagen V Denmark
- Department of Biochemistry and Microbiology; University of Victoria; Victoria BC V8W 3P6 Canada
| | - Sébastien Vidal
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires; Laboratoire de Chimie Organique 2-Glycochimie, UMR 5246; CNRS and Université Claude Bernard Lyon 1; 43 Boulevard du 11 Novembre 1918 69622 Villeurbanne France
| |
Collapse
|
29
|
Vasconcelos-Dos-Santos A, Oliveira IA, Lucena MC, Mantuano NR, Whelan SA, Dias WB, Todeschini AR. Biosynthetic Machinery Involved in Aberrant Glycosylation: Promising Targets for Developing of Drugs Against Cancer. Front Oncol 2015; 5:138. [PMID: 26161361 PMCID: PMC4479729 DOI: 10.3389/fonc.2015.00138] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/02/2015] [Indexed: 12/22/2022] Open
Abstract
Cancer cells depend on altered metabolism and nutrient uptake to generate and keep the malignant phenotype. The hexosamine biosynthetic pathway is a branch of glucose metabolism that produces UDP-GlcNAc and its derivatives, UDP-GalNAc and CMP-Neu5Ac and donor substrates used in the production of glycoproteins and glycolipids. Growing evidence demonstrates that alteration of the pool of activated substrates might lead to different glycosylation and cell signaling. It is already well established that aberrant glycosylation can modulate tumor growth and malignant transformation in different cancer types. Therefore, biosynthetic machinery involved in the assembly of aberrant glycans are becoming prominent targets for anti-tumor drugs. This review describes three classes of glycosylation, O-GlcNAcylation, N-linked, and mucin type O-linked glycosylation, involved in tumor progression, their biosynthesis and highlights the available inhibitors as potential anti-tumor drugs.
Collapse
Affiliation(s)
| | - Isadora A Oliveira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Miguel Clodomiro Lucena
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Natalia Rodrigues Mantuano
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Stephen A Whelan
- Department of Biochemistry, Cardiovascular Proteomics Center, Boston University School of Medicine , Boston, MA , USA
| | - Wagner Barbosa Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| | - Adriane Regina Todeschini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brasil
| |
Collapse
|
30
|
Huang Y, Shaw MA, Mullins ES, Kirley TL, Ayres N. Synthesis and anticoagulant activity of polyureas containing sulfated carbohydrates. Biomacromolecules 2014; 15:4455-66. [PMID: 25329742 PMCID: PMC4261991 DOI: 10.1021/bm501245v] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
Polyurea-based synthetic glycopolymers
containing sulfated glucose,
mannose, glucosamine, or lactose as pendant groups have been synthesized
by step-growth polymerization of hexamethylene diisocyanate and corresponding
secondary diamines. The obtained polymers were characterized by gel
permeation chromatography, nuclear magnetic resonance spectroscopy,
and Fourier transform infrared spectroscopy. The nonsulfated polymers
showed similar results to the commercially available biomaterial polyurethane
TECOFLEX in a platelet adhesion assay. The average degree of sulfation
after reaction with SO3 was calculated from elemental analysis
and found to be between three and four −OSO3 groups
per saccharide. The blood-compatibility of the synthetic polymers
was measured using activated partial thromboplastin time, prothrombin
time, thrombin time, anti-IIa, and anti-Xa assays. Activated partial
thromboplastin time, prothrombin time, and thrombin time results indicated
that the mannose and lactose based polymers had the highest anticoagulant
activities among all the sulfated polymers. The mechanism of action
of the polymers appears to be mediated via an anti-IIa pathway rather
than an anti-Xa pathway.
Collapse
Affiliation(s)
- Yongshun Huang
- Department of Chemistry and ‡Materials Science and Engineering Program, The University of Cincinnati , Cincinnati, Ohio 45221, United States
| | | | | | | | | |
Collapse
|
31
|
Moukha-chafiq O, Reynolds RC. Parallel solution-phase synthesis and general biological activity of a uridine antibiotic analog library. ACS COMBINATORIAL SCIENCE 2014; 16:232-7. [PMID: 24661222 PMCID: PMC4025591 DOI: 10.1021/co4001452] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
![]()
A small library of ninety four uridine antibiotic analogs was synthesized,
under the Pilot Scale Library (PSL) Program of the NIH Roadmap initiative,
from amine 2 and carboxylic acids 33 and 77 in solution-phase fashion. Diverse aldehyde, sulfonyl chloride,
and carboxylic acid reactant sets were condensed to 2, leading after acid-mediated hydrolysis, to the targeted compounds 3–32 in good yields and high purity. Similarly,
treatment of 33 with diverse amines and sulfonamides
gave 34–75. The coupling of the amino
terminus of d-phenylalanine methyl ester to the free 5′-carboxylic
acid moiety of 33 followed by sodium hydroxide treatment
led to carboxylic acid analog 77. Hydrolysis of this
material gave analog 78. The intermediate 77 served as the precursor for the preparation of novel dipeptidyl
uridine analogs 79–99 through peptide
coupling reactions to diverse amine reactants. None of the described
compounds show significant anticancer or antimalarial acivity. A number
of samples exhibited a variety of promising inhibitory, agonist, antagonist,
or activator properties with enzymes and receptors in primary screens
supplied and reported through the NIH MLPCN program.
Collapse
Affiliation(s)
- Omar Moukha-chafiq
- Southern Research Institute, Drug Discovery Division, Birmingham, Alabama 35205, United States
| | - Robert C. Reynolds
- Southern Research Institute, Drug Discovery Division, Birmingham, Alabama 35205, United States
| |
Collapse
|
32
|
Datta D, Samanta A, Dasgupta S, Pathak T. Synthesis of 5′-carboxymethylsulfonyl-5′-deoxyribonucleosides under mild hydrolytic conditions: a new class of acidic nucleosides as inhibitors of ribonuclease A. RSC Adv 2014. [DOI: 10.1039/c3ra45084h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
33
|
Wang S, Cuesta-Seijo JA, Lafont D, Palcic MM, Vidal S. Design of glycosyltransferase inhibitors: pyridine as a pyrophosphate surrogate. Chemistry 2013; 19:15346-57. [PMID: 24108680 DOI: 10.1002/chem.201301871] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/25/2013] [Indexed: 12/12/2022]
Abstract
A series of ten glycosyltransferase inhibitors has been designed and synthesized by using pyridine as a pyrophosphate surrogate. The series was prepared by conjugation of carbohydrate, pyridine, and nucleoside building blocks by using a combination of glycosylation, the Staudinger-Vilarrasa amide-bond formation, and azide-alkyne click chemistry. The compounds were evaluated as inhibitors of five metal-dependent galactosyltransferases. Crystallographic analyses of three inhibitors complexed in the active site of one of the enzymes confirmed that the pyridine moiety chelates the Mn(2+) ion causing a slight displacement (2 Å) from its original position. The carbohydrate head group occupies a different position than in the natural uridine diphosphate (UDP)-Gal substrate with little interaction with the enzyme.
Collapse
Affiliation(s)
- Shuai Wang
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, Laboratoire de Chimie Organique 2, Glycochimie, UMR 5246, CNRS and Université Claude Bernard Lyon 1, 43 Boulevard du 11 Novembre 1918, 6922 Villeurbanne (France), Fax: (+33) 472-448-109
| | | | | | | | | |
Collapse
|
34
|
Paszkowska J, Kral K, Bieg T, Nawrot U, Szeja W, Wandzik I. Synthesis and preliminary biological evaluations of 5'-substituted derivatives of uridine as glycosyltransferase inhibitors. Molecules 2013; 18:8018-27. [PMID: 23884133 PMCID: PMC6270623 DOI: 10.3390/molecules18078018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 06/26/2013] [Indexed: 11/20/2022] Open
Abstract
New derivatives of uridine which contain a β-ketoenol motif were synthesized, characterized and biologically tested. Synthesized compounds 1–4 showed no activity against bovine milk β-1,4-galactosyltransferase I at concentrations up to 2.0 mM and were not active against Candida albicans and Aspergilus fumigatus up to the maximum tested concentration of 1,000 µg/mL.
Collapse
Affiliation(s)
- Jadwiga Paszkowska
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland; E-Mails: (J.P.); (K.K.); (T.B.); (W.S.)
| | - Katarzyna Kral
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland; E-Mails: (J.P.); (K.K.); (T.B.); (W.S.)
| | - Tadeusz Bieg
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland; E-Mails: (J.P.); (K.K.); (T.B.); (W.S.)
| | - Urszula Nawrot
- Department of Microbiology, Wroclaw Medical University, T. Chałubińskiego 4, 50-368 Wrocław, Poland; E-Mail:
| | - Wiesław Szeja
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland; E-Mails: (J.P.); (K.K.); (T.B.); (W.S.)
| | - Ilona Wandzik
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland; E-Mails: (J.P.); (K.K.); (T.B.); (W.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +48-32-2372028; Fax: +48-32-2372094
| |
Collapse
|
35
|
Schaefer K, Sindhuwinata N, Hackl T, Kötzler MP, Niemeyer FC, Palcic MM, Peters T, Meyer B. A nonionic inhibitor with high specificity for the UDP-Gal donor binding site of human blood group B galactosyltransferase: design, synthesis, and characterization. J Med Chem 2013; 56:2150-4. [PMID: 23406460 DOI: 10.1021/jm300642a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
9-(5-O-α-D-galactopyranosyl)-D-arabinityl-1,3,7-trihydropurine-2,6,8-trione (1) was designed and synthesized as a nonionic inhibitor for the donor binding site of human blood group B galactosyltransferase (GTB). Enzymatic characterization showed 1 to be extremely specific, as the highly homologous human N-acetylgalactosaminyltransferase (GTA) is not inhibited. The binding epitope of 1 demonstrates a high involvement of the arabinityl linker, whereas the galactose residue is only making contact to the protein via its C-2 site, which is very important for the discrimination between galactose and N-acetylgalactosamine, the substrate transferred by GTA. The approach can generate highly specific glycosyltransferase inhibitors.
Collapse
Affiliation(s)
- Katrin Schaefer
- Organic Chemistry, Department of Chemistry, Faculty of Sciences, University of Hamburg, Martin Luther King Platz 6, 20146 Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Mugunthan G, Sriram D, Yogeeswari P, Kartha KPR. Synthesis and Preliminary Screening ofO-β-D-Galactopyranosides as Potential Mimic of UDP-GalpagainstMycobacterium tuberculosis. J Carbohydr Chem 2012. [DOI: 10.1080/07328303.2011.654017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
37
|
Developing inhibitors of glycan processing enzymes as tools for enabling glycobiology. Nat Chem Biol 2012; 8:683-94. [PMID: 22810773 DOI: 10.1038/nchembio.1029] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Glycoconjugates are ubiquitous biomolecules found in all kingdoms of life. These diverse structures are metabolically responsive and occur in a cell line- and protein-specific manner, conferring tissue type-specific properties. Glycans have essential roles in diverse processes, including, for example, intercellular signaling, inflammation, protein quality control, glucohomeostasis and cellular adhesion as well as cell differentiation and proliferation. Many mysteries remain in the field, however, and uncovering the physiological roles of various glycans remains a key pursuit. Realizing this aim necessitates the ability to subtly and selectively manipulate the series of different glycoconjugates both in cells and in vivo. Selective small-molecule inhibitors of glycan processing enzymes hold great potential for such manipulation as well as for determining the function of 'orphan' carbohydrate-processing enzymes. In this review, we discuss recent advances and existing inhibitors, the prospects for small-molecule inhibitors and the challenges associated with generating high-quality chemical probes for these families of enzymes. The coordinated efforts of chemists, biochemists and biologists will be crucial for creating and characterizing inhibitors that are useful tools both for advancing a basic understanding of glycobiology in mammals as well as for validating new potential therapeutic targets within this burgeoning field.
Collapse
|
38
|
A neutral diphosphate mimic crosslinks the active site of human O-GlcNAc transferase. Nat Chem Biol 2011; 8:72-7. [PMID: 22082911 PMCID: PMC3241908 DOI: 10.1038/nchembio.711] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 08/31/2011] [Indexed: 12/29/2022]
Abstract
Glycosyltransferases (Gtfs) catalyze the formation of a diverse array of glycoconjugates. Small molecule inhibitors to manipulate Gtf activity in cells have long been sought as tools to understand Gtf function. Success has been limited due to challenges in designing inhibitors that mimic the negatively-charged diphosphate substrates. Here we report the mechanism of action of a small molecule that inhibits O-GlcNAc transferase (OGT), an essential human enzyme that modulates cell signaling pathways by catalyzing a unique intracellular post translational modification, β-O-GlcNAcylation. The molecule contains a five heteroatom dicarbamate core that functions as a neutral diphosphate mimic. One dicarbamate carbonyl reacts with an essential active site lysine that anchors the diphosphate of the nucleotide-sugar substrate. The lysine adduct reacts again with a nearby cysteine to crosslink the OGT active site. While this unprecedented mechanism reflects the unique architecture of the OGT active site, related dicarbamate scaffolds may inhibit other enzymes that bind diphosphate containing substrates.
Collapse
|
39
|
Auberger N, Frlan R, Al-Dabbagh B, Bouhss A, Crouvoisier M, Gravier-Pelletier C, Le Merrer Y. Synthesis and biological evaluation of potential new inhibitors of the bacterial transferase MraY with a β-ketophosphonate structure. Org Biomol Chem 2011; 9:8301-12. [PMID: 22042341 DOI: 10.1039/c1ob06124k] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Stable analogs of bacterial transferase MraY substrate or product with a pyrophosphate surrogate in their structure are described. β-ketophosphonates were designed as pyrophosphate bioisosteres and were investigated as UDP-GlcNAc mimics. The developed strategy allows introduction of structural diversity at a late stage of the synthesis. The biological activity of the synthesized compounds was evaluated on the MraY enzyme.
Collapse
Affiliation(s)
- Nicolas Auberger
- Université Paris Descartes, UMR 8601 CNRS, Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, 45 rue des Saints Pères, 75006 Paris, France
| | | | | | | | | | | | | |
Collapse
|
40
|
Descroix K, Wagner GK. The first C-glycosidic analogue of a novel galactosyltransferase inhibitor. Org Biomol Chem 2011; 9:1855-63. [PMID: 21267505 DOI: 10.1039/c0ob00630k] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Structural analogues and mimics of the natural sugar-nucleotide UDP-galactose (UDP-Gal) are sought after as chemical tools for glycobiology and drug discovery. We have recently developed a novel class of galactosyltransferase (GalT) inhibitors derived from UDP-Gal, bearing an additional substituent at the 5-position of the uracil base. Herein we report the first C-glycosidic derivative of this new class of GalT inhibitors. We describe a practical convergent synthesis of the new UDP-C-Gal derivative, including a systematic study into the use of radical chemistry for the preparation of galactosyl ethylphosphonate, a key synthetic intermediate. The new inhibitor showed activity against a bacterial UDP-Gal 4'-epimerase at micromolar concentrations. This is the first example of a base-modified UDP-sugar as an inhibitor of a UDP-sugar-dependent enzyme which is not a glycosyltransferase, and these results may therefore have implications for the design of inhibitors of these enzymes in the future.
Collapse
Affiliation(s)
- Karine Descroix
- School of Pharmacy, University of East Anglia, Norwich, UK NR4 7TJ
| | | |
Collapse
|
41
|
Vembaiyan K, Pearcey JA, Bhasin M, Lowary TL, Zou W. Synthesis of sugar-amino acid-nucleosides as potential glycosyltransferase inhibitors. Bioorg Med Chem 2010; 19:58-66. [PMID: 21167722 DOI: 10.1016/j.bmc.2010.11.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 11/18/2010] [Accepted: 11/22/2010] [Indexed: 11/29/2022]
Abstract
Sugar-amino acid-nucleosides (SAAN) were synthesized to mimic glycosyl nucleotide donors based on the hypothesis that a basic amino acid may interact with carboxylate groups of the enzyme in a manner similar to the diphosphate metal ion complex. C-Glycoside analogues of the d-galactopyranose or l-arabinofuranose ring systems, and four amino acids (lysine, glutamine, tryptophan, and histidine), were chosen for this study. The targets were synthesized and tested against GlfT2, a galactofuranosyltransferase essential for cell wall galactan biosynthesis in Mycobacterium tuberculosis. The inhibition assay showed that analogues containing histidine and tryptophan are moderate inhibitors of GlfT2.
Collapse
Affiliation(s)
- Kannan Vembaiyan
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada K1A 0R6
| | | | | | | | | |
Collapse
|
42
|
Zhang GL, Zhang LH, Ye XS. Concise syntheses of selective inhibitors against α-1,3-galactosyltransferase. Org Biomol Chem 2010; 8:5062-8. [PMID: 20820649 DOI: 10.1039/c0ob00042f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Several iminosugar-based uridine diphosphate galactose (UDP-Gal) mimetics 1-4 including d- and l-epimers were designed and synthesized by concise routes, and these synthetic compounds were evaluated for the inhibition of α-1,3- and β-1,4-galactosyltransferases in vitro. The experimental data demonstrated that l-epimer 2 displayed the strongest inhibitory activity with moderate selectivity against α-1,3-galactosyltransferase.
Collapse
Affiliation(s)
- Guo-Liang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, and School of Pharmaceutical Sciences, Peking University, Xue Yuan Road #38, Beijing 100191, China
| | | | | |
Collapse
|
43
|
Niewiadomski S, Beebeejaun Z, Denton H, Smith TK, Morris RJ, Wagner GK. Rationally designed squaryldiamides - a novel class of sugar-nucleotide mimics? Org Biomol Chem 2010; 8:3488-99. [PMID: 20532300 DOI: 10.1039/c004165c] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sugar-nucleotides such as GDP-mannose, GDP-fucose and UDP-glucose are important biomolecules with a central role in carbohydrate and glycoconjugate biosynthesis, metabolism and cell signalling. Analogues and mimics of naturally occurring sugar-nucleotides are sought after as chemical tools and inhibitor candidates for sugar-nucleotide-dependent enzymes including glycosyltransferases. Many sugar-nucleotides bind to their target glycosyltransferases via coordination of the diphosphate group to a divalent metal cofactor in the active site. The identification of uncharged, chemically stable surrogates for the diphosphate group, with the ability to coordinate to a divalent metal, is therefore an important design criteria for the development of sugar-nucleotide mimics. Here, we describe the rational design and synthesis of a novel class of sugar-nucleotide mimics based on a squaryldiamide scaffold, an uncharged phosphate isostere. We demonstrate by comprehensive NMR titration experiments that the new sugar-nucleotide mimics coordinate efficiently to Mg(2+), and provide results from biological studies with a therapeutically relevant mannosyltransferase from Trypanosoma brucei. Our findings suggest that squaryldiamides are a promising template for the development of sugar-nucleotide mimics, and illustrate the considerable potential of the squarylamide group as a fragment for inhibitor design.
Collapse
|
44
|
Trunkfield AE, Gurcha SS, Besra GS, Bugg TD. Inhibition of Escherichia coli glycosyltransferase MurG and Mycobacterium tuberculosis Gal transferase by uridine-linked transition state mimics. Bioorg Med Chem 2010; 18:2651-63. [PMID: 20226679 PMCID: PMC3034214 DOI: 10.1016/j.bmc.2010.02.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Revised: 01/07/2010] [Accepted: 02/16/2010] [Indexed: 11/01/2022]
Abstract
Glycosyltransferase MurG catalyses the transfer of N-acetyl-d-glucosamine to lipid intermediate I on the bacterial peptidoglycan biosynthesis pathway, and is a target for development of new antibacterial agents. A transition state mimic was designed for MurG, containing a functionalised proline, linked through the alpha-carboxylic acid, via a spacer, to a uridine nucleoside. A set of 15 functionalised prolines were synthesised, using a convergent dipolar cycloaddition reaction, which were coupled via either a glycine, proline, sarcosine, or diester linkage to the 5'-position of uridine. The library of 18 final compounds were tested as inhibitors of Escherichia coli glycosyltransferase MurG. Ten compounds showed inhibition of MurG at 1mM concentration, the most active with IC(50) 400microM. The library was also tested against Mycobacterium tuberculosis galactosyltransferase GlfT2, and one compound showed effective inhibition at 1mM concentration.
Collapse
Affiliation(s)
- Amy E. Trunkfield
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Sudagar S. Gurcha
- Department of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Gurdyal S. Besra
- Department of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Timothy D.H. Bugg
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| |
Collapse
|
45
|
Chiesa K, Shvoryna A, Bernet B, Vasella A. Oligonucleotide Analogues with Integrated Bases and Backbones. Part 24. Helv Chim Acta 2010. [DOI: 10.1002/hlca.201000011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Tóth M, Somsák L. 5'-Uridyl derivatives of N-glycosyl allophanic acid and biuret. Carbohydr Res 2010; 345:163-7. [PMID: 19896644 DOI: 10.1016/j.carres.2009.08.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 08/24/2009] [Accepted: 08/28/2009] [Indexed: 10/20/2022]
Abstract
(2',3'-O-Isopropylidene-5'-uridyl) 4-(2,3,4,6-tetra-O-acetyl-beta-d-glycopyranosyl)allophanates were obtained in the reactions of 2',3'-O-isopropylidene-uridine and O-peracetylated beta-d-gluco-, galacto- and xylopyranosylamines, and OCNCOCl. 2,3,4,6-Tetra-O-acetyl-beta-d-glucopyranosyl isocyanate and N-(2',3'-O-isopropylidene-5'-uridyl)urea gave 1-(2,3,4,6-tetra-O-acetyl-beta-d-glucopyranosyl)-5-(2',3'-O-isopropylidene-5'-uridyl)biuret. Deprotection of the beta-d-gluco configured allophanate and biuret was carried out by standard methods.
Collapse
Affiliation(s)
- Marietta Tóth
- Department of Organic Chemistry, University of Debrecen, POB 20, H-4010 Debrecen, Hungary
| | | |
Collapse
|
47
|
Zhang Y, Ren S, Sun D, Tao J. Synthesis and Evaluation of the Analogues for Galactosyl Donors as Inhibitors ofβ-1,4-Galactosyltransferase. SYNTHETIC COMMUN 2010. [DOI: 10.1080/00397910902730887] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Yanxia Zhang
- a School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China , Qingdao, China
| | - Sumei Ren
- a School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China , Qingdao, China
| | - Dongkui Sun
- a School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China , Qingdao, China
| | - Jiang Tao
- a School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China , Qingdao, China
| |
Collapse
|
48
|
Duez S, Coudray L, Mouray E, Grellier P, Dubois J. Towards the synthesis of bisubstrate inhibitors of protein farnesyltransferase: Synthesis and biological evaluation of new farnesylpyrophosphate analogues. Bioorg Med Chem 2010; 18:543-56. [DOI: 10.1016/j.bmc.2009.12.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Accepted: 12/04/2009] [Indexed: 01/16/2023]
|
49
|
Rowan AS, Nicely NI, Cochrane N, Wlassoff WA, Claiborne A, Hamilton CJ. Nucleoside triphosphate mimicry: a sugar triazolyl nucleoside as an ATP-competitive inhibitor of B. anthracis pantothenate kinase. Org Biomol Chem 2009; 7:4029-36. [PMID: 19763307 PMCID: PMC6074028 DOI: 10.1039/b909729e] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The synthesis of a library of nucleoside triphosphate mimetics is described where the Mg(2+) chelated triphosphate sidechain is replaced by an uncharged methylene-triazole linked monosaccharide sidechain. The compounds have been evaluated as inhibitors of Bacillus anthracis pantothenate kinase and a competitive inhibitor has been identified with a K(i) that is 3-fold lower than the K(m) value of ATP.
Collapse
Affiliation(s)
- Andrew S Rowan
- School of Chemistry and Chemical Engineering, David Keir Building, Stranmillis Road, Queen's University Belfast, Belfast, BT9 5AG
| | | | | | | | | | | |
Collapse
|
50
|
Synthesis of 2-deoxy-hexopyranosyl derivatives of uridine as donor substrate analogues for glycosyltransferases. Bioorg Chem 2009; 37:211-6. [PMID: 19765794 DOI: 10.1016/j.bioorg.2009.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 07/30/2009] [Accepted: 08/11/2009] [Indexed: 11/24/2022]
Abstract
A series of 2-deoxy-hexopyranosyl derivatives of uridine have been synthesized as analogues of UDP-sugar. These compounds were tested as inhibitors against bovine beta-1,4-galactosyltransferase I in fluorescent assays and showed no significant inhibition.
Collapse
|