1
|
Paymal S, Barale SS, Supanekar SV, Sonawane KD, Pawar KD. Overexpression, Purification, and Biochemical Characterization of the vanC2 d-Ala-d-Ser Ligase from Enterococcus casseliflavus SSK and Its Inhibition by an Oxadiazole Derivative. ACS OMEGA 2025; 10:14390-14402. [PMID: 40256530 PMCID: PMC12004154 DOI: 10.1021/acsomega.5c00599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/22/2025]
Abstract
The bacterial cell wall and enzymes involved in peptidoglycan biosynthesis are prime targets for the discovery of novel antibacterial agents. Among these enzymes, d-alanine-d-alanine ligases (Ddl) are particularly significant due to their utilization of specific substrates (d-amino acids) essential for bacterial viability. Isozymes of Ddl that utilize alternative substrates such as d-lactate or d-serine are found in vancomycin-resistant Gram-positive bacteria, initially identified in Enterococcus species, and now represent a growing concern in clinical settings. In this study, we isolated and identified vancomycin-resistant Enterococcus casseliflavus (E. casseliflavus) strain SSK and used it for amplification, cloning, and purification of the vanC2 type of d-alanine-d-serine ligase (EcfDdls). Investigations of substrate specificity and enzyme kinetics provided insights into the enzyme's mechanistic action. Evaluation of the inhibitory potential of the previously virtually screened oxadiazole derivative 1-[(5-methyl-1,2-oxazol-3-yl)methyl]-4-{[3-(propan-2-yl)-1,2,4-oxadiazol-5-yl]methyl}piperazine (CID 45805715) was carried out using an inorganic phosphate detection assay, which demonstrated complete enzymatic inhibition of purified EcfDdls. When tested, CID 45805715 significantly inhibited activity of Ddl, with an IC50 of 76.7 μM, compared to 313 μM for the reference compound DCS. Moreover, this compound also exhibited antimicrobial activity against vancomycin-resistant E. casseliflavus strain SSK. Thus, these findings provide valuable insights into the activity and inhibition of vanC2 EcfDdls, offering a promising avenue for addressing vancomycin resistance in enterococci, particularly in nosocomial infections affecting immunocompromised patients.
Collapse
Affiliation(s)
- Sneha
B. Paymal
- Department
of Microbiology, Shivaji University, Vidyanagar, Kolhapur, Maharashtra 416004, India
- Rayat
Institute
of Research and Development (RIRD), Satara, Maharashtra 415001, India
| | - Sagar S. Barale
- Department
of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan 305817, India
| | | | - Kailas D. Sonawane
- Department
of Biochemistry, Shivaji University, Vidyanagar, Kolhapur, Maharashtra 416004, India
| | - Kiran D. Pawar
- School
of
Nanoscience and Biotechnology, Shivaji University, Vidyanagar, Kolhapur, Maharashtra 416004, India
| |
Collapse
|
2
|
Becker R, Pederick JL, Dawes EG, Bruning JB, Abell AD. Structure-guided design and synthesis of ATP-competitive N-acyl-substituted sulfamide d-alanine-d-alanine ligase inhibitors. Bioorg Med Chem 2023; 96:117509. [PMID: 37948922 DOI: 10.1016/j.bmc.2023.117509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/10/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
d-Alanine-d-alanine ligase (Ddl) catalyses the ATP-dependent formation of d-Ala-d-Ala, a critical component in bacterial cell wall biosynthesis and is a validated target for new antimicrobial agents. Here, we describe the structure-guided design, synthesis, and evaluation of ATP-competitive N-acyl-substituted sulfamides 27-36, 42, 46, 47 as inhibitors of Staphylococcus aureus Ddl (SaDdl). A crystal structure of SaDdl complexed with ATP and d-Ala-d-Ala (PDB: 7U9K) identified ATP-mimetic 8 as an initial scaffold for further inhibitor design. Evaluation of 8 in SaDdl enzyme inhibition assays revealed the ability to reduce enzyme activity to 72 ± 8 % (IC50 = 1.6 mM). The sulfamide linker of 8 was extended with 2-(4-methoxyphenyl)ethanol to give 29, to investigate further interactions with the d-Ala pocket of SaDdl, as predicted by molecular docking. This compound reduced enzyme activity to 89 ± 1 %, with replacement of the 4-methoxyphenyl group in 29 with alternative phenyl substituents (27, 28, 31-33, 35, 36) failing to significantly improve on this (80-89 % remaining enzyme activity). Exchanging these phenyl substituents with selected heterocycles (42, 46, 47) did improve activity, with the most active compound (42) reducing SaDdl activity to 70 ± 1 % (IC50 = 1.7 mM), which compares favourably to the FDA-approved inhibitor d-cycloserine (DCS) (IC50 = 0.1 mM). To the best of our knowledge, this is the first reported study of bisubstrate SaDdl inhibitors.
Collapse
Affiliation(s)
- Rouven Becker
- Department of Chemistry, School of Physics, Chemistry and Earth Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia; Institute for Photonics and Advanced Sensing, (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Jordan L Pederick
- Institute for Photonics and Advanced Sensing, (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Edward G Dawes
- Department of Chemistry, School of Physics, Chemistry and Earth Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia; Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, South Australia 5005, Australia
| | - John B Bruning
- Institute for Photonics and Advanced Sensing, (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Andrew D Abell
- Department of Chemistry, School of Physics, Chemistry and Earth Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia; Institute for Photonics and Advanced Sensing, (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, South Australia 5005, Australia.
| |
Collapse
|
3
|
Ding H, Lyu J, Zhang XL, Xiao X, Liu XW. Efficient and versatile formation of glycosidic bonds via catalytic strain-release glycosylation with glycosyl ortho-2,2-dimethoxycarbonylcyclopropylbenzoate donors. Nat Commun 2023; 14:4010. [PMID: 37419914 PMCID: PMC10329021 DOI: 10.1038/s41467-023-39619-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/16/2023] [Indexed: 07/09/2023] Open
Abstract
Catalytic glycosylation is a vital transformation in synthetic carbohydrate chemistry due to its ability to expediate the large-scale oligosaccharide synthesis for glycobiology studies with the consumption of minimal amounts of promoters. Herein we introduce a facile and efficient catalytic glycosylation employing glycosyl ortho-2,2-dimethoxycarbonylcyclopropylbenzoates (CCBz) promoted by a readily accessible and non-toxic Sc(III) catalyst system. The glycosylation reaction involves a novel activation mode of glycosyl esters driven by the ring-strain release of an intramolecularly incorporated donor-acceptor cyclopropane (DAC). The versatile glycosyl CCBz donor enables highly efficient construction of O-, S-, and N-glycosidic bonds under mild conditions, as exemplified by the convenient preparation of the synthetically challenging chitooligosaccharide derivatives. Of note, a gram-scale synthesis of tetrasaccharide corresponding to Lipid IV with modifiable handles is achieved using the catalytic strain-release glycosylation. These attractive features promise this donor to be the prototype for developing next generation of catalytic glycosylation.
Collapse
Affiliation(s)
- Han Ding
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Jian Lyu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Xiao-Lin Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Xiong Xiao
- School of Chemistry and Chemical Engineering, Northwestern Polytechnical University (NPU), Xi'an, 710072, P.R. China.
| | - Xue-Wei Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore.
| |
Collapse
|
4
|
Saidjalolov S, Edoo Z, Fonvielle M, Mayer L, Iannazzo L, Arthur M, Etheve-Quelquejeu M, Braud E. Synthesis of Carbapenems Containing Peptidoglycan Mimetics and Inhibition of the Cross-Linking Activity of a Transpeptidase of l,d Specificity. Chemistry 2021; 27:3542-3551. [PMID: 33336443 DOI: 10.1002/chem.202004831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/15/2020] [Indexed: 11/07/2022]
Abstract
The carbapenem class of β-lactams has been optimized against Gram-negative bacteria producing extended-spectrum β-lactamases by introducing substituents at position C2. Carbapenems are currently investigated for the treatment of tuberculosis as these drugs are potent covalent inhibitors of l,d-transpeptidases involved in mycobacterial cell wall assembly. The optimization of carbapenems for inactivation of these unusual targets is sought herein by exploiting the nucleophilicity of the C8 hydroxyl group to introduce chemical diversity. As β-lactams are structure analogs of peptidoglycan precursors, the substituents were chosen to increase similarity between the drug and the substrate. Fourteen peptido-carbapenems were efficiently synthesized. They were more effective than the reference drug, meropenem, owing to the positive impact of a phenethylthio substituent introduced at position C2 but the peptidomimetics added at position C8 did not further improve the activity. Thus, position C8 can be modified to modulate the pharmacokinetic properties of highly efficient carbapenems.
Collapse
Affiliation(s)
- Saidbakhrom Saidjalolov
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, UMR 8601 CNRS, Université de Paris, 45, rue des saints-pères, Paris, 75006, France
| | - Zainab Edoo
- INSERM UMRS 1138, Sorbonne Universités, UPMC Univ Paris 06, Sorbonne Paris Cité, Université de Paris, Centre de recherche des Cordeliers, Paris, 75006, France
| | - Matthieu Fonvielle
- INSERM UMRS 1138, Sorbonne Universités, UPMC Univ Paris 06, Sorbonne Paris Cité, Université de Paris, Centre de recherche des Cordeliers, Paris, 75006, France
| | - Louis Mayer
- INSERM UMRS 1138, Sorbonne Universités, UPMC Univ Paris 06, Sorbonne Paris Cité, Université de Paris, Centre de recherche des Cordeliers, Paris, 75006, France
| | - Laura Iannazzo
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, UMR 8601 CNRS, Université de Paris, 45, rue des saints-pères, Paris, 75006, France
| | - Michel Arthur
- INSERM UMRS 1138, Sorbonne Universités, UPMC Univ Paris 06, Sorbonne Paris Cité, Université de Paris, Centre de recherche des Cordeliers, Paris, 75006, France
| | - Mélanie Etheve-Quelquejeu
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, UMR 8601 CNRS, Université de Paris, 45, rue des saints-pères, Paris, 75006, France
| | - Emmanuelle Braud
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, UMR 8601 CNRS, Université de Paris, 45, rue des saints-pères, Paris, 75006, France
| |
Collapse
|
5
|
Design and Synthesis of Various 5'-Deoxy-5'-(4-Substituted-1,2,3-Triazol-1-yl)-Uridine Analogues as Inhibitors of Mycobacterium tuberculosis Mur Ligases. Molecules 2020; 25:molecules25214953. [PMID: 33114668 PMCID: PMC7663697 DOI: 10.3390/molecules25214953] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 01/06/2023] Open
Abstract
The synthesis of hitherto unknown 5′-deoxy-5′-(4-substituted-1,2,3-triazol-1-yl)-uridine and its evaluation, through an one-pot screening assay, against MurA-F enzymes involved in Mycobacterium tuberculosis (Mtb), are described. Starting from UDP-N-acetylmuramic acid (UDP-MurNAc), the natural substrate involved in the peptidoglycan biosynthesis, our strategy was to substitute the diphosphate group of UDP-MurNAc by a 1,2,3-triazolo spacer under copper-catalyzed azide-alkyne cycloaddition conditions. The structure-activity relationship was discussed and among the 23 novel compounds developed, N-acetylglucosamine analogues 11c and 11e emerged as the best inhibitors against the Mtb MurA-F enzymes reconstruction pathway with an inhibitory effect of 56% and 50%, respectively, at 100 μM. Both compounds are selective inhibitors of Mtb MurE, the molecular docking and molecular dynamic simulation suggesting that 11c and 11e are occupying the active site of Mtb MurE ligase.
Collapse
|
6
|
MurE inhibitors as antibacterial agents: a review. J INCL PHENOM MACRO 2020. [DOI: 10.1007/s10847-020-01018-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
7
|
Azam MA, Jupudi S. MurD inhibitors as antibacterial agents: a review. CHEMICAL PAPERS 2020. [DOI: 10.1007/s11696-020-01057-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
8
|
Jukič M, Gobec S, Sova M. Reaching toward underexplored targets in antibacterial drug design. Drug Dev Res 2018; 80:6-10. [PMID: 30312991 DOI: 10.1002/ddr.21465] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 01/03/2023]
Abstract
The increase of antimicrobial resistance necessitates the renewal and strong research involvement in antibacterial drug design. In the following work, we comment on the key approaches used in development of new antibacterials, focusing on intracellular therapeutic targets that have been so far mostly underexplored: the enzymes of the Mur pathway MurA to MurF. We identify common obstacles observed during research on MurA, MurB, and Mur ligases inhibitors and their development into potential antibacterial compounds, and discern several approaches and solutions to tackle the whole-cell activity of designed compounds. Furthermore, we consolidate recent literature reports and encourage the further research on Mur enzymes.
Collapse
Affiliation(s)
- Marko Jukič
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Matej Sova
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
9
|
Yu JY, Cheng HJ, Wu HR, Wu WS, Lu JW, Cheng TJ, Wu YT, Fang JM. Structure-based design of bacterial transglycosylase inhibitors incorporating biphenyl, amine linker and 2-alkoxy-3-phosphorylpropanoate moieties. Eur J Med Chem 2018; 150:729-741. [PMID: 29574202 DOI: 10.1016/j.ejmech.2018.03.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/02/2018] [Accepted: 03/12/2018] [Indexed: 01/28/2023]
Abstract
Transglycosylase (TGase) is essential to biosynthesis of peptidoglycan for formation of bacterial cell wall. Moenomycin is a potent TGase inhibitor, but not used in clinic treatment due to its poor pharmacokinetics. The E-F disaccharide, phosphoglycerate and lipid tail in moenomycin are crucial elements for TGase inhibition and antibacterial activity. Based on this scaffold, a series of truncated mimics comprising biphenyl, amine linker and 2-alkoxy-3-phosphorylpropanoate moieties were designed to test their TGase inhibitory activity. In this design, the phosphorylpropanoate group is a surrogate of phosphoglycerate with improved stability. A library of lipid tails can be constructed by a straightforward approach using Cu(I)-catalyzed (3 + 2) cycloaddition reactions, and the as-synthesized triazole ring can provide additional hydrogen bonds in the TGase active site. Our molecular docking experiments reveal that the biphenyl group provides π-π and π-cation interactions to act as a simplified alternative of the C-E disaccharide in moenomycin. To play the role of the oxonium transition state in transglycosylation, the amine linker exists as a positively charged species in physiological condition to attain electrostatic interactions with acidic residues. In this study, two biphenyl-linked 2-alkoxy-3-phosphorylpropanoate compounds (8 and 10) are found to exhibit modest inhibitory activity (IC50 ≈ 150 μM) against the TGase of Acinetobacter baumannii and good antibacterial activity against Staphylococcus aureus (MIC = 6.3 μM).
Collapse
Affiliation(s)
- Jui-Yin Yu
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan, ROC
| | - Hsiu-Jung Cheng
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan, ROC
| | - Huei-Ru Wu
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan, ROC
| | - Wei-Shen Wu
- The Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, ROC
| | - Jui-Wen Lu
- The Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, ROC
| | - Ting-Jen Cheng
- The Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, ROC
| | - Ying-Ta Wu
- The Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, ROC
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan, ROC; The Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, ROC.
| |
Collapse
|
10
|
Urinary Tract Conditions Affect Fosfomycin Activity against Escherichia coli Strains Harboring Chromosomal Mutations Involved in Fosfomycin Uptake. Antimicrob Agents Chemother 2017; 62:AAC.01899-17. [PMID: 29038268 DOI: 10.1128/aac.01899-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/10/2017] [Indexed: 11/20/2022] Open
Abstract
The steps by which Escherichia coli strains harboring mutations related to fosfomycin (FOS) resistance arise and spread during urinary tract infections (UTIs) are far from being understood. The aim of this study was to evaluate the effects of urine, pH, and anaerobiosis on FOS activity against a set of isogenic strains carrying the most prevalent chromosomal mutations conferring FOS resistance (ΔuhpT, ΔglpT, ΔcyaA, and ΔptsI), either singly or in combination. We also studied fosfomycin-resistant E. coli clinical isolates from patients with UTI. Our results demonstrate that urinary tract physiological conditions might have a profound impact on FOS activity against strains with chromosomal FOS resistance mutations. Specifically, acidic pH values and anaerobiosis convert most of the strains categorized as resistant to fosfomycin according to the international guidelines to a susceptible status. Therefore, urinary pH values may have practical interest in the management of UTIs. Finally, our results, together with the high fitness cost associated with FOS resistance mutations, might explain the low prevalence of fosfomycin-resistant E. coli variants in UTIs.
Collapse
|
11
|
Kaminskyy D, Kryshchyshyn A, Lesyk R. 5-Ene-4-thiazolidinones - An efficient tool in medicinal chemistry. Eur J Med Chem 2017; 140:542-594. [PMID: 28987611 PMCID: PMC7111298 DOI: 10.1016/j.ejmech.2017.09.031] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 07/14/2017] [Accepted: 09/17/2017] [Indexed: 02/02/2023]
Abstract
The presented review is an attempt to summarize a huge volume of data on 5-ene-4-thiazolidinones being a widely studied class of small molecules used in modern organic and medicinal chemistry. The manuscript covers approaches to the synthesis of 5-ene-4-thiazolidinone derivatives: modification of the C5 position of the basic core; synthesis of the target compounds in the one-pot or multistage reactions or transformation of other related heterocycles. The most prominent pharmacological profiles of 5-ene derivatives of different 4-thiazolidinone subtypes belonging to hit-, lead-compounds, drug-candidates and drugs as well as the most studied targets have been discussed. Currently target compounds (especially 5-en-rhodanines) are assigned as frequent hitters or pan-assay interference compounds (PAINS) within high-throughput screening campaigns. Nevertheless, the crucial impact of the presence/nature of C5 substituent (namely 5-ene) on the pharmacological effects of 5-ene-4-thiazolidinones was confirmed by the numerous listed findings from the original articles. The main directions for active 5-ene-4-thiazolidinones optimization have been shown: i) complication of the fragment in the C5 position; ii) introduction of the substituents in the N3 position (especially fragments with carboxylic group or its derivatives); iii) annealing in complex heterocyclic systems; iv) combination with other pharmacologically attractive fragments within hybrid pharmacophore approach. Moreover, the utilization of 5-ene-4-thiazolidinones in the synthesis of complex compounds with potent pharmacological application is described. The chemical transformations cover mainly the reactions which involve the exocyclic double bond in C5 position of the main core and correspond to the abovementioned direction of the 5-ene-4-thiazolidinone modification.
Collapse
Affiliation(s)
- Danylo Kaminskyy
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv-10, 79010, Ukraine
| | - Anna Kryshchyshyn
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv-10, 79010, Ukraine
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, Lviv-10, 79010, Ukraine.
| |
Collapse
|
12
|
Identification of Ideal Multi-targeting Bioactive Compounds Against Mur Ligases of Enterobacter aerogenes and Its Binding Mechanism in Comparison with Chemical Inhibitors. Interdiscip Sci 2017; 11:135-144. [PMID: 29086207 DOI: 10.1007/s12539-017-0261-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 08/02/2017] [Accepted: 09/11/2017] [Indexed: 10/18/2022]
Abstract
Enterobacter aerogenes have been reported as important opportunistic and multi-resistant bacterial pathogens for humans during the last three decades in hospital wards. The emergence of drug-resistant E. aerogenes demands the need for developing new drugs. Peptidoglycan is an important component of the cell wall of bacteria and the peptidoglycan biochemical pathway is considered as the best source of antibacterial targets. Within this pathway, four Mur ligases MurC, MurD, MurE, and MurF are responsible for the successive additions of L-alanine and suitable targets for developing novel antibacterial drugs. As an inference from this fact, we modeled the three-dimensional structure of above Mur ligases using best template structures available in PDB and analyzed its common binding features. Structural refinement and energy minimization of the predicted Mur ligases models is also being done using molecular dynamics studies. The models of Mur ligases were further investigated for in silico docking studies using bioactive plant compounds from the literature. Interestingly, these results indicate that four plant compounds Isojuripidine, Atroviolacegenin, Porrigenin B, and Nummularogenin showing better docking results in terms of binding energy and number of hydrogen bonds. All these four compounds are spirostan-based compounds with differences in side chains and the amino acid such as ASN, LYS, THR, HIS, ARG (polar) and PHE, GLY, VAL, ALA, MET (non-polar) playing active role in binding site of all four Mur ligases. Overall, in the predicted model, the four plant compounds with its binding features could pave way to design novel multi-targeted antibacterial plant-based bioactive compounds specific to Mur ligases for the treatment of Enterobacter infections.
Collapse
|
13
|
Extra precision docking, free energy calculation and molecular dynamics studies on glutamic acid derivatives as MurD inhibitors. Comput Biol Chem 2017; 69:55-63. [DOI: 10.1016/j.compbiolchem.2017.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 04/18/2017] [Accepted: 05/20/2017] [Indexed: 01/28/2023]
|
14
|
Fang C, Nagy-Staroń A, Grafe M, Heermann R, Jung K, Gebhard S, Mascher T. Insulation and wiring specificity of BceR-like response regulators and their target promoters inBacillus subtilis. Mol Microbiol 2017; 104:16-31. [DOI: 10.1111/mmi.13597] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Chong Fang
- Department Biology I; Ludwig-Maximilians-Universität München; Großhaderner Str. 2-4 Martinsried 82152 Germany
| | - Anna Nagy-Staroń
- Department Biology I; Ludwig-Maximilians-Universität München; Großhaderner Str. 2-4 Martinsried 82152 Germany
- Institute of Science and Technology (IST) Austria, Am Campus 1; Klosterneuburg A-3400 Austria
| | - Martin Grafe
- Department Biology I; Ludwig-Maximilians-Universität München; Großhaderner Str. 2-4 Martinsried 82152 Germany
| | - Ralf Heermann
- Department Biology I; Ludwig-Maximilians-Universität München; Großhaderner Str. 2-4 Martinsried 82152 Germany
- Munich Center for Integrated Protein Science (CiPSM) at the Department Biology I; Ludwig-Maximilians-Universität München; Großhaderner Str. 2-4 Martinsried 82152 Germany
| | - Kirsten Jung
- Department Biology I; Ludwig-Maximilians-Universität München; Großhaderner Str. 2-4 Martinsried 82152 Germany
- Munich Center for Integrated Protein Science (CiPSM) at the Department Biology I; Ludwig-Maximilians-Universität München; Großhaderner Str. 2-4 Martinsried 82152 Germany
| | - Susanne Gebhard
- Department Biology I; Ludwig-Maximilians-Universität München; Großhaderner Str. 2-4 Martinsried 82152 Germany
- Milner Centre for Evolution, Department of Biology and Biochemistry; University of Bath; Claverton Down Bath BA2 7AY UK
| | - Thorsten Mascher
- Department Biology I; Ludwig-Maximilians-Universität München; Großhaderner Str. 2-4 Martinsried 82152 Germany
- Institute of Microbiology; Technische Universität (TU) Dresden; 01062 Dresden Germany
| |
Collapse
|
15
|
Lee TK, Meng K, Shi H, Huang KC. Single-molecule imaging reveals modulation of cell wall synthesis dynamics in live bacterial cells. Nat Commun 2016; 7:13170. [PMID: 27774981 PMCID: PMC5078992 DOI: 10.1038/ncomms13170] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 09/08/2016] [Indexed: 12/03/2022] Open
Abstract
The peptidoglycan cell wall is an integral organelle critical for bacterial cell shape and stability. Proper cell wall construction requires the interaction of synthesis enzymes and the cytoskeleton, but it is unclear how the activities of individual proteins are coordinated to preserve the morphology and integrity of the cell wall during growth. To elucidate this coordination, we used single-molecule imaging to follow the behaviours of the two major peptidoglycan synthases in live, elongating Escherichia coli cells and after perturbation. We observed heterogeneous localization dynamics of penicillin-binding protein (PBP) 1A, the synthase predominantly associated with cell wall elongation, with individual PBP1A molecules distributed between mobile and immobile populations. Perturbations to PBP1A activity, either directly through antibiotics or indirectly through PBP1A's interaction with its lipoprotein activator or other synthases, shifted the fraction of mobile molecules. Our results suggest that multiple levels of regulation control the activity of enzymes to coordinate peptidoglycan synthesis. The bacterial cell wall is important for cell shape and stability, but how the activities of the biosynthetic machinery are coordinated are not clear. Here the authors use single-molecule imaging and chemical perturbations to determine factors that affect the localization dynamics of penicillin-binding proteins (PBP)1A and PBP1B.
Collapse
Affiliation(s)
- Timothy K Lee
- Department of Bioengineering, Stanford University, Stanford, California 94305, USA.,Program in Biomedical Informatics, Stanford University, Stanford, California 94305, USA
| | - Kevin Meng
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Handuo Shi
- Department of Bioengineering, Stanford University, Stanford, California 94305, USA
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, California 94305, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
16
|
Abstract
Lactic acid bacteria (LAB) are important starter, commensal, or pathogenic microorganisms. The stress physiology of LAB has been studied in depth for over 2 decades, fueled mostly by the technological implications of LAB robustness in the food industry. Survival of probiotic LAB in the host and the potential relatedness of LAB virulence to their stress resilience have intensified interest in the field. Thus, a wealth of information concerning stress responses exists today for strains as diverse as starter (e.g., Lactococcus lactis), probiotic (e.g., several Lactobacillus spp.), and pathogenic (e.g., Enterococcus and Streptococcus spp.) LAB. Here we present the state of the art for LAB stress behavior. We describe the multitude of stresses that LAB are confronted with, and we present the experimental context used to study the stress responses of LAB, focusing on adaptation, habituation, and cross-protection as well as on self-induced multistress resistance in stationary phase, biofilms, and dormancy. We also consider stress responses at the population and single-cell levels. Subsequently, we concentrate on the stress defense mechanisms that have been reported to date, grouping them according to their direct participation in preserving cell energy, defending macromolecules, and protecting the cell envelope. Stress-induced responses of probiotic LAB and commensal/pathogenic LAB are highlighted separately due to the complexity of the peculiar multistress conditions to which these bacteria are subjected in their hosts. Induction of prophages under environmental stresses is then discussed. Finally, we present systems-based strategies to characterize the "stressome" of LAB and to engineer new food-related and probiotic LAB with improved stress tolerance.
Collapse
|
17
|
Auletta S, Galli F, Lauri C, Martinelli D, Santino I, Signore A. Imaging bacteria with radiolabelled quinolones, cephalosporins and siderophores for imaging infection: a systematic review. Clin Transl Imaging 2016; 4:229-252. [PMID: 27512687 PMCID: PMC4960278 DOI: 10.1007/s40336-016-0185-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/17/2016] [Indexed: 12/20/2022]
Abstract
Bacterial infections are still one of the main causes of patient morbidity and mortality worldwide. Nowadays, many imaging techniques, like computed tomography or magnetic resonance imaging, are used to identify inflammatory processes, but, although they recognize anatomical modifications, they cannot easily distinguish bacterial infective foci from non bacterial infections. In nuclear medicine, many efforts have been made to develop specific radiopharmaceuticals to discriminate infection from sterile inflammation. Several compounds (antimicrobial peptides, leukocytes, cytokines, antibiotics…) have been radiolabelled and tested in vitro and in vivo, but none proved to be highly specific for bacteria. Indeed factors, including the number and strain of bacteria, the infection site, and the host condition may affect the specificity of tested radiopharmaceuticals. Ciprofloxacin has been proposed and intensively studied because of its easy radiolabelling method, broad spectrum, and low cost, but at the same time it presents some problems such as low stability or the risk of antibiotic resistance. Therefore, in the present review studies with ciprofloxacin and other radiolabelled antibiotics as possible substitutes of ciprofloxacin are reported. Among them we can distinguish different classes, such as cephalosporins, fluoroquinolones, inhibitors of nucleic acid synthesis, inhibitors of bacterial cell wall synthesis and inhibitors of protein synthesis; then also others, like siderophores or maltodextrin-based probes, have been discussed as bacterial infection imaging agents. A systematic analysis was performed to report the main characteristics and differences of each antibiotic to provide an overview about the state of the art of imaging infection with radiolabelled antibiotics.
Collapse
Affiliation(s)
- S. Auletta
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, St. Andrea Hospital, “Sapienza” University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy
| | - F. Galli
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, St. Andrea Hospital, “Sapienza” University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy
| | - C. Lauri
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, St. Andrea Hospital, “Sapienza” University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy
| | - D. Martinelli
- Microbiology Unit, Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
| | - I. Santino
- Microbiology Unit, Department of Clinical and Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, St. Andrea Hospital, “Sapienza” University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy
| |
Collapse
|
18
|
The Staphylococcus aureus Methicillin Resistance Factor FmtA Is a d-Amino Esterase That Acts on Teichoic Acids. mBio 2016; 7:e02070-15. [PMID: 26861022 PMCID: PMC4752606 DOI: 10.1128/mbio.02070-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED The methicillin resistance factor encoded by fmtA is a core member of the Staphylococcus aureus cell wall stimulon, but its function has remained elusive for the past two decades. First identified as a factor that affects methicillin resistance in S. aureus strains, FmtA was later shown to interact with teichoic acids and to localize to the cell division septum. We have made a breakthrough in understanding FmtA function. We show that FmtA hydrolyzes the ester bond between d-Ala and the backbone of teichoic acids, which are polyglycerol-phosphate or polyribitol-phosphate polymers found in the S. aureus cell envelope. FmtA contains two conserved motifs found in serine active-site penicillin-binding proteins (PBPs) and β-lactamases. The conserved SXXK motif was found to be important for the d-amino esterase activity of FmtA. Moreover, we show that deletion of fmtA (ΔfmtA) led to higher levels of d-Ala in teichoic acids, and this effect was reversed by complementation of ΔfmtA with fmtA. The positive charge on d-Ala partially masks the negative charge of the polyol-phosphate backbone of teichoic acids; hence, a change in the d-Ala content will result in modulation of their charge. Cell division, biofilm formation, autolysis, and colonization are among the many processes in S. aureus affected by the d-Ala content and overall charge of the cell surface teichoic acids. The esterase activity of FmtA and the regulation of fmtA suggest that FmtA functions as a modulator of teichoic acid charge, thus FmtA may be involved in S. aureus cell division, biofilm formation, autolysis, and colonization. IMPORTANCE Teichoic acids are involved in cell division, cell wall synthesis, biofilm formation, attachment of bacteria to artificial surfaces, and colonization. However, the function of teichoic acids is not fully understood. Modification by glycosylation and/or d-alanylation of the polyol-phosphate backbone of teichoic acids is important in the above cell processes. The intrinsic negative charge of teichoic acid backbone plays a role in the charge and/or pH of the bacterial surface, and d-alanylation represents a means through which bacteria modulate the charge or the pH of their surfaces. We discovered that FmtA removes d-Ala from teichoic acids. We propose FmtA may provide a temporal and spatial regulation of the bacterial cell surface charge in two ways, by removing the d-Ala from LTA to make it available to wall teichoic acid (WTA) in response to certain conditions and by removing it from WTA to allow the cell to reset its surface charge to a previous condition.
Collapse
|
19
|
Guo Z, Han J, Yang XY, Cao K, He K, Du G, Zeng G, Zhang L, Yu G, Sun Z, He QY, Sun X. Proteomic analysis of the copper resistance of Streptococcus pneumoniae. Metallomics 2015; 7:448-54. [PMID: 25608595 DOI: 10.1039/c4mt00276h] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Streptococcus pneumoniae is a Gram-positive bacterial pathogen causing a variety of diseases, including otitis media, bacteraemia and meningitis. Although copper is an essential trace metal for bacterial growth, high intracellular levels of free-copper are toxic. Copper resistance has emerged as an important virulence determinant of microbial pathogens. In this study, we determined the minimum inhibition concentration of copper for the growth inhibition of S. pneumoniae. Two-dimensional-electrophoresis coupled with mass spectrometry was applied to identify proteins involved in copper resistance of S. pneumoniae. In total, forty-four proteins with more than 1.5-fold alteration in expression (p < 0.05) were identified. Quantitative reverse transcription PCR was used to confirm the proteomic results. Bioinformatics analysis showed that the differentially expressed proteins were mainly involved in the cell wall biosynthesis, protein biosynthesis, purine biosynthesis, pyrimidine biosynthesis, primary metabolic process, and the nitrogen compound metabolic process. Many up-regulated proteins in response to the copper treatment directly or indirectly participated in the cell wall biosynthesis, indicating that the cell wall is a critical determinant in copper resistance of S. pneumoniae.
Collapse
Affiliation(s)
- Zhong Guo
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Höfler C, Heckmann J, Fritsch A, Popp P, Gebhard S, Fritz G, Mascher T. Cannibalism stress response in Bacillus subtilis. MICROBIOLOGY-SGM 2015; 162:164-176. [PMID: 26364265 DOI: 10.1099/mic.0.000176] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
When faced with carbon source limitation, the Gram-positive soil organism Bacillus subtilis initiates a survival strategy called sporulation, which leads to the formation of highly resistant endospores that allow B. subtilis to survive even long periods of starvation. In order to avoid commitment to this energy-demanding and irreversible process, B. subtilis employs another strategy called 'cannibalism' to delay sporulation as long as possible. Cannibalism involves the production and secretion of two cannibalism toxins, sporulation delaying protein (SDP) and sporulation killing factor (SKF), which are able to lyse sensitive siblings. The lysed cells are thought to then provide nutrients for the cannibals to slow down or even prevent them from entering sporulation. In this study, we uncovered the role of the cell envelope stress response (CESR), especially the Bce-like antimicrobial peptide detoxification modules, in the cannibalism stress response during the stationary phase. SDP and SKF specifically induce Bce-like systems and some extracytoplasmic function σ factors in stationary-phase cultures, but only the latter provide some degree of protection. A full Bce response is only triggered by mature toxins, and not by toxin precursors. Our study provides insights into the close relationship between stationary-phase survival and the CESR of B. subtilis.
Collapse
Affiliation(s)
- Carolin Höfler
- Department Biology I, Ludwig-Maximilians-Universität München, Großhaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Judith Heckmann
- Department Biology I, Ludwig-Maximilians-Universität München, Großhaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Anne Fritsch
- Department Biology I, Ludwig-Maximilians-Universität München, Großhaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Philipp Popp
- Department Biology I, Ludwig-Maximilians-Universität München, Großhaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Susanne Gebhard
- Department Biology I, Ludwig-Maximilians-Universität München, Großhaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Georg Fritz
- Department Biology I, Ludwig-Maximilians-Universität München, Großhaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Thorsten Mascher
- Department Biology I, Ludwig-Maximilians-Universität München, Großhaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
21
|
Kouidmi I, Levesque RC, Paradis-Bleau C. The biology of Mur ligases as an antibacterial target. Mol Microbiol 2014; 94:242-53. [DOI: 10.1111/mmi.12758] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2014] [Indexed: 01/19/2023]
Affiliation(s)
- Imène Kouidmi
- Department of Microbiology, Infectiology and Immunology; Université de Montreal; Montreal Quebec Canada
| | - Roger C. Levesque
- Institut de biologie intégrative et des systèmes; Université Laval; Montreal Quebec Canada
| | - Catherine Paradis-Bleau
- Department of Microbiology, Infectiology and Immunology; Université de Montreal; Montreal Quebec Canada
| |
Collapse
|
22
|
UDP-N-acetylmuramic acid l-alanine ligase (MurC) inhibition in a tolC mutant Escherichia coli strain leads to cell death. Antimicrob Agents Chemother 2014; 58:6165-71. [PMID: 25114134 DOI: 10.1128/aac.02890-14] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Mur ligases play an essential role in the biosynthesis of bacterial peptidoglycan and hence are attractive antibacterial targets. A screen of the AstraZeneca compound library led to the identification of compound A, a pyrazolopyrimidine, as a potent inhibitor of Escherichia coli and Pseudomonas aeruginosa MurC. However, cellular activity against E. coli or P. aeruginosa was not observed. Compound A was active against efflux pump mutants of both strains. Experiments using an E. coli tolC mutant revealed accumulation of the MurC substrate and a decrease in the level of product upon treatment with compound A ,: indicating inhibition of MurC enzyme in these cells. Such a modulation was not observed in the E. coli wild-type cells. Further, overexpression of MurC in the E. coli tolC mutant led to an increase in the compound A MIC by ≥16-fold, establishing a correlation between MurC inhibition and cellular activity. In addition, estimation of the intracellular compound A level showed an accumulation of the compound over time in the tolC mutant strain. A significant compound A level was not detected in the wild-type E. coli strain even upon treatment with high concentrations of the compound. Therefore, the lack of MIC and absence of MurC inhibition in wild-type E. coli were possibly due to suboptimal compound concentration as a consequence of a high efflux level and/or poor permeativity of compound A.
Collapse
|
23
|
Manabe K, Nishizawa S, Shiratori S. Porous surface structure fabricated by breath figures that suppresses Pseudomonas aeruginosa biofilm formation. ACS APPLIED MATERIALS & INTERFACES 2013; 5:11900-11905. [PMID: 24171453 DOI: 10.1021/am4035762] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
As colonizers of medical-device surfaces, Pseudomonas aeruginosa strains present a serious source of infection and are of major concern. In this study, we fabricated films with porous surfaces by breath figures that disturb mergence by bacterial attachment, thereby impeding biofilm development. Previous studies have shown that microtopography prevents the development of P. aeruginosa biofilms. Accordingly we indented surfaces with patterns of micrometer-sized pores using breath figures at ordinary temperatures and pressures. The antimicrobial effect of surface figures was experimentally investigated by controlling the surface structure. The results suggested that pores of 5-11 μm in diameter effectively inhibit bacterial activity. It appears that biofilm development is precluded by the decreased contact area between the films and bacteria.
Collapse
Affiliation(s)
- Kengo Manabe
- Graduate School of Science and Technology, School of Integrated Design Engineering, Keio University , 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | | | | |
Collapse
|
24
|
Mann PA, Müller A, Xiao L, Pereira PM, Yang C, Ho Lee S, Wang H, Trzeciak J, Schneeweis J, dos Santos MM, Murgolo N, She X, Gill C, Balibar CJ, Labroli M, Su J, Flattery A, Sherborne B, Maier R, Tan CM, Black T, Önder K, Kargman S, Monsma FJ, Pinho MG, Schneider T, Roemer T. Murgocil is a highly bioactive staphylococcal-specific inhibitor of the peptidoglycan glycosyltransferase enzyme MurG. ACS Chem Biol 2013; 8:2442-51. [PMID: 23957438 DOI: 10.1021/cb400487f] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Modern medicine is founded on the discovery of penicillin and subsequent small molecules that inhibit bacterial peptidoglycan (PG) and cell wall synthesis. However, the discovery of new chemically and mechanistically distinct classes of PG inhibitors has become exceedingly rare, prompting speculation that intracellular enzymes involved in PG precursor synthesis are not 'druggable' targets. Here, we describe a β-lactam potentiation screen to identify small molecules that augment the activity of β-lactams against methicillin-resistant Staphylococcus aureus (MRSA) and mechanistically characterize a compound resulting from this screen, which we have named murgocil. We provide extensive genetic, biochemical, and structural modeling data demonstrating both in vitro and in whole cells that murgocil specifically inhibits the intracellular membrane-associated glycosyltransferase, MurG, which synthesizes the lipid II PG substrate that penicillin binding proteins (PBPs) polymerize and cross-link into the cell wall. Further, we demonstrate that the chemical synergy and cidality achieved between murgocil and the β-lactam imipenem is mediated through MurG dependent localization of PBP2 to the division septum. Collectively, these data validate our approach to rationally identify new target-specific bioactive β-lactam potentiation agents and demonstrate that murgocil now serves as a highly selective and potent chemical probe to assist our understanding of PG biosynthesis and cell wall biogenesis across Staphylococcal species.
Collapse
Affiliation(s)
- Paul A. Mann
- Infectious
Disease Research, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Anna Müller
- Institute
of Medical Microbiology, Immunology and Parasitology—Pharmaceutical
Microbiology Section, University of Bonn, Bonn, Germany
| | - Li Xiao
- Computational
Chemistry, Global Structure Chemistry, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Pedro M. Pereira
- Laboratory
of Bacterial Cell Biology, Instituto de Tecnologia Química
e Biológica, Universidade Nova de Lisboa, Avenida da República, 2781-901 Oeiras, Portugal
| | - Christine Yang
- Medicinal
Chemistry, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Sang Ho Lee
- Infectious
Disease Research, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Hao Wang
- Infectious
Disease Research, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Joanna Trzeciak
- Infectious
Disease Research, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Jonathan Schneeweis
- In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Margarida Moreira dos Santos
- Laboratory
of Bacterial Cell Biology, Instituto de Tecnologia Química
e Biológica, Universidade Nova de Lisboa, Avenida da República, 2781-901 Oeiras, Portugal
| | - Nicholas Murgolo
- Research
Solutions, Bioinformatics, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Xinwei She
- Informatics
IT, Merck Inc., Boston, Massachusetts 02110, United States
| | - Charles Gill
- In Vivo Pharmacology, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Carl J. Balibar
- Infectious
Disease Research, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Marc Labroli
- Medicinal
Chemistry, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Jing Su
- Medicinal
Chemistry, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Amy Flattery
- In Vivo Pharmacology, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Brad Sherborne
- Computational
Chemistry, Global Structure Chemistry, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Richard Maier
- Procomcure Biotech GmbH, Krems a.d. Donau, Austria
- Division of Molecular
Dermatology, Department of Dermatology, Paracelsus Medical University, Salzburg, Austria
| | - Christopher M. Tan
- Infectious
Disease Research, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Todd Black
- Infectious
Disease Research, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Kamil Önder
- Procomcure Biotech GmbH, Krems a.d. Donau, Austria
- Division of Molecular
Dermatology, Department of Dermatology, Paracelsus Medical University, Salzburg, Austria
| | - Stacia Kargman
- In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Frederick J Monsma
- In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| | - Mariana G. Pinho
- Laboratory
of Bacterial Cell Biology, Instituto de Tecnologia Química
e Biológica, Universidade Nova de Lisboa, Avenida da República, 2781-901 Oeiras, Portugal
| | - Tanja Schneider
- Institute
of Medical Microbiology, Immunology and Parasitology—Pharmaceutical
Microbiology Section, University of Bonn, Bonn, Germany
- German Centre for Infection Research (DZIF), partner site
Bonn-Cologne, Bonn, Germany
| | - Terry Roemer
- Infectious
Disease Research, Merck Research Laboratories, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
25
|
Kaur N, Khokhar M, Jain V, Bharatam PV, Sandhir R, Tewari R. Identification of druggable targets for Acinetobacter baumannii via subtractive genomics and plausible inhibitors for MurA and MurB. Appl Biochem Biotechnol 2013; 171:417-36. [PMID: 23846799 DOI: 10.1007/s12010-013-0372-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 06/24/2013] [Indexed: 11/28/2022]
Abstract
Emergence of the multidrug-resistant pathogens has rendered the current therapies ineffective thereby, resulting in the need for new drugs and drug targets. The accumulating protein sequence data has initiated a drift from classical drug discovery protocols to structure-based drug designing. In the present study, in silico subtractive genomics approach was implemented to find a set of potential drug targets present in an opportunist bacterial pathogen, Acinetobacter baumannii (A. baumannii). Out of the 43 targets identified, further studies for protein model building and lead-inhibitor identification were carried out on two cell-essential targets, MurA and MurB enzymes (of A. baumannii designated as MurAAb and MurBAb) involved in the peptidoglycan biosynthesis pathway of bacteria. The homology model built for each of them was further refined and validated using various available programs like PROCHECK, Errat, ProSA energy plots, etc. Compounds showing activity against MurA and MurB enzymes of other organisms were collected from the literature and were docked into the active site of MurAAb and MurBAb enzymes. Three inhibitors namely, T6361, carbidopa, and aesculin, showed maximum Glide score, hydrogen bonding interactions with the key amino acid residues of both the enzymes and acceptable ADME properties. Furthermore, molecular dynamics simulation studies on MurAAb-T6361 and MurBAb-T6361 complexes suggested that the ligand has a high binding affinity with both the enzymes and the hydrogen bonding with the key residues were stable in the dynamic condition also. Therefore, these ligands have been propsed as dual inhibitors and promising lead compounds for the drug design against MurAAb and MurBAb enzymes.
Collapse
Affiliation(s)
- Navkiran Kaur
- Centre for Microbial Biotechnology, Panjab University, Sector 14, Chandigarh 160014, India
| | | | | | | | | | | |
Collapse
|
26
|
Shih HW, Chang YF, Li WJ, Meng FC, Huang CY, Ma C, Cheng TJR, Wong CH, Cheng WC. Effect of the Peptide Moiety of Lipid II on Bacterial Transglycosylase. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201204038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
27
|
Shih HW, Chang YF, Li WJ, Meng FC, Huang CY, Ma C, Cheng TJR, Wong CH, Cheng WC. Effect of the peptide moiety of Lipid II on bacterial transglycosylase. Angew Chem Int Ed Engl 2012; 51:10123-6. [PMID: 22952114 DOI: 10.1002/anie.201204038] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Hao-Wei Shih
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Taipei 115, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yoshida T, Nasu H, Namba E, Ubukata O, Yamashita M. Discovery of a compound that acts as a bacterial PyrG (CTP synthase) inhibitor. J Med Microbiol 2012; 61:1280-1285. [PMID: 22700553 DOI: 10.1099/jmm.0.046052-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PyrG (CTP synthase) catalyses the conversion of UTP to CTP, an essential step in the pyrimidine metabolic pathway in a variety of bacteria, including those causing community-acquired respiratory tract infections (RTIs). In this study, a luminescence-based ATPase assay of PyrG was developed and used to evaluate the inhibitory activity of 2-(3-[3-oxo-1,2-benzisothiazol-2(3H)-yl]phenylsulfonylamino) benzoic acid (compound G1). Compound G1 inhibited PyrG derived from Streptococcus pneumoniae with a 50 % inhibitory concentration value of 0.091 µM, and the inhibitory activity of compound G1 was 13 times higher than that of acivicin (1.2 µM), an established PyrG inhibitor. The results of saturation transfer difference analysis using nuclear magnetic resonance spectroscopy suggested that these compounds compete with ATP and/or UTP for binding to Strep. pneumoniae PyrG. Finally, compound G1 was shown to have antimicrobial activity against several different bacteria causing RTIs, such as Staphylococcus aureus and Haemophilus influenzae, suggesting that it is a prototype chemical compound that could be harnessed as an antimicrobial drug with a novel structure to target bacterial PyrG.
Collapse
Affiliation(s)
- Tatsuhiko Yoshida
- Biological Research Laboratories IV, Daiichi Sankyo Co. Ltd, 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Hatsumi Nasu
- Biological Research Laboratories IV, Daiichi Sankyo Co. Ltd, 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Eiko Namba
- Biological Research Laboratories IV, Daiichi Sankyo Co. Ltd, 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Osamu Ubukata
- Core Technology Research Laboratories, Daiichi Sankyo Co. Ltd, 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Makoto Yamashita
- Biological Research Laboratories IV, Daiichi Sankyo Co. Ltd, 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| |
Collapse
|
29
|
Interaction with lipid II induces conformational changes in bovicin HC5 structure. Antimicrob Agents Chemother 2012; 56:4586-93. [PMID: 22687503 DOI: 10.1128/aac.00295-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bovicin HC5 is a lantibiotic produced by Streptococcus bovis HC5 that targets the cell wall precursor lipid II. An understanding of the modes of action against target bacteria can help broadening the clinical applicability of lantibiotics in human and veterinary medicine. In this study, the interaction of bovicin HC5 with lipid II was examined using tryptophan fluorescence and circular dichroism spectroscopy with model membrane systems that do or do not allow pore formation by bovicin HC5. In the presence of lipid II, a blue-shift of 12 nm could be observed for the fluorescence emission maximum of the tryptophan residue for all of the membrane systems tested. This change in fluorescence emission was paralleled by a decrease in accessibility toward acrylamide and phospholipids carrying a spin-label at the acyl chain; the tryptophan residue of bovicin HC5 was located near the twelfth position of the membrane phospholipid acyl chains. Moreover, the binding of lipid II by bovicin HC5 induced remarkable conformational changes in the bovicin HC5 structure. The interaction of bovicin HC5 with lipid II was highly stable even at pH 2.0. These results indicate that bovicin HC5 interacts directly with lipid II and that the topology of this interaction changes under different conditions, which is relevant for the biological activity of the peptide. To our knowledge, bovicin HC5 is the only bacteriocin described thus far that is able to interact with its target in extreme pH values, and this fact might be related to its unique structure and stability.
Collapse
|
30
|
Development of anti-infectives using phage display: biological agents against bacteria, viruses, and parasites. Antimicrob Agents Chemother 2012; 56:4569-82. [PMID: 22664969 DOI: 10.1128/aac.00567-12] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The vast majority of anti-infective therapeutics on the market or in development are small molecules; however, there is now a nascent pipeline of biological agents in development. Until recently, phage display technologies were used mainly to produce monoclonal antibodies (MAbs) targeted against cancer or inflammatory disease targets. Patent disputes impeded broad use of these methods and contributed to the dearth of candidates in the clinic during the 1990s. Today, however, phage display is recognized as a powerful tool for selecting novel peptides and antibodies that can bind to a wide range of antigens, ranging from whole cells to proteins and lipid targets. In this review, we highlight research that exploits phage display technology as a means of discovering novel therapeutics against infectious diseases, with a focus on antimicrobial peptides and antibodies in clinical or preclinical development. We discuss the different strategies and methods used to derive, select, and develop anti-infectives from phage display libraries and then highlight case studies of drug candidates in the process of development and commercialization. Advances in screening, manufacturing, and humanization technologies now mean that phage display can make a significant contribution in the fight against clinically important pathogens.
Collapse
|
31
|
Tuson HH, Auer GK, Renner LD, Hasebe M, Tropini C, Salick M, Crone WC, Gopinathan A, Huang KC, Weibel DB. Measuring the stiffness of bacterial cells from growth rates in hydrogels of tunable elasticity. Mol Microbiol 2012; 84:874-91. [PMID: 22548341 DOI: 10.1111/j.1365-2958.2012.08063.x] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although bacterial cells are known to experience large forces from osmotic pressure differences and their local microenvironment, quantitative measurements of the mechanical properties of growing bacterial cells have been limited. We provide an experimental approach and theoretical framework for measuring the mechanical properties of live bacteria. We encapsulated bacteria in agarose with a user-defined stiffness, measured the growth rate of individual cells and fit data to a thin-shell mechanical model to extract the effective longitudinal Young's modulus of the cell envelope of Escherichia coli (50-150 MPa), Bacillus subtilis (100-200 MPa) and Pseudomonas aeruginosa (100-200 MPa). Our data provide estimates of cell wall stiffness similar to values obtained via the more labour-intensive technique of atomic force microscopy. To address physiological perturbations that produce changes in cellular mechanical properties, we tested the effect of A22-induced MreB depolymerization on the stiffness of E. coli. The effective longitudinal Young's modulus was not significantly affected by A22 treatment at short time scales, supporting a model in which the interactions between MreB and the cell wall persist on the same time scale as growth. Our technique therefore enables the rapid determination of how changes in genotype and biochemistry affect the mechanical properties of the bacterial envelope.
Collapse
Affiliation(s)
- Hannah H Tuson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Yoshida T, Nasu H, Namba E, Ubukata O, Yamashita M. Discovery of a compound which acts as a bacterial UMP kinase PyrH inhibitor. FEMS Microbiol Lett 2012; 330:121-6. [PMID: 22428584 DOI: 10.1111/j.1574-6968.2012.02546.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 02/28/2012] [Accepted: 03/01/2012] [Indexed: 11/29/2022] Open
Abstract
PyrH is a member of the UMP kinase family that catalyses the conversion of UMP to UDP, an essential step in the pyrimidine metabolic pathway in a variety of bacteria including those causing community-acquired respiratory tract infections (RTIs). In this study, we have developed a luminescence-based kinase assay of PyrH and evaluated the inhibitory activity of PYRH-1 (sodium {3-[4-tert-butyl-3-(9H-xanthen-9-ylacetylamino)phenyl]-1-cyclohexylmethylpropoxycarbonyloxy}acetate). PYRH-1 inhibits PyrH derived from both Streptococcus pneumoniae and Haemophilus influenzae with IC(50) (concentration of inhibitor giving a 50% decrease in enzyme activity) values of 48 and 75 μM, respectively, whose inhibitory activity against S. pneumoniae PyrH was far higher compared with that of UTP (IC(50) = 710 μM), an allosteric PyrH inhibitor. The molecular interaction analysis by surface plasmon resonance suggested that PYRH-1 directly interacts with S. pneumoniae PyrH at one-to-one molar ratio. Finally, PYRH-1 was shown to have antimicrobial activity against several different bacteria causing RTIs, such as S. pneumoniae, Staphylococcus aureus, H. influenzae (acrA knockout strain), suggesting that PYRH-1 is a prototype chemical compound that can be harnessed as an antimicrobial drug with a novel mode of action by targeting bacterial PyrH.
Collapse
Affiliation(s)
- Tatsuhiko Yoshida
- Biological Research Laboratories IV, Daiichi Sankyo Co., Ltd, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
33
|
Sarabia F, Vivar-García C, García-Ruiz C, Martín-Ortiz L, Romero-Carrasco A. Exploring the Chemistry of Epoxy Amides for the Synthesis of the 2′′-epi-Diazepanone Core of Liposidomycins and Caprazamycins. J Org Chem 2012; 77:1328-39. [DOI: 10.1021/jo202061t] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Francisco Sarabia
- Department of Organic Chemistry, Faculty
of Sciences, University of Malaga, 29071
Malaga, Spain
| | - Carlos Vivar-García
- Department of Organic Chemistry, Faculty
of Sciences, University of Malaga, 29071
Malaga, Spain
| | - Cristina García-Ruiz
- Department of Organic Chemistry, Faculty
of Sciences, University of Malaga, 29071
Malaga, Spain
| | - Laura Martín-Ortiz
- Department of Organic Chemistry, Faculty
of Sciences, University of Malaga, 29071
Malaga, Spain
| | - Antonio Romero-Carrasco
- Department of Organic Chemistry, Faculty
of Sciences, University of Malaga, 29071
Malaga, Spain
| |
Collapse
|
34
|
Hao H, Cheng G, Dai M, Wu Q, Yuan Z. Inhibitors targeting on cell wall biosynthesis pathway of MRSA. MOLECULAR BIOSYSTEMS 2012; 8:2828-38. [DOI: 10.1039/c2mb25188d] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
35
|
KONG DEXIN, ZHU WEILIANG, WU DALEI, SHEN XU, JIANG HUALIANG. COMPARISON OF THREE 3D-QSAR METHODS USING A NOVEL CLASS OF MURF INHIBITORS. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2011. [DOI: 10.1142/s0219633607002812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
MurF was considered as an attractive target for new antibacterial discovery. In this paper, three QSAR methods were employed, viz. comparative molecular field analysis (CoMFA), comparative molecular similarity indices analysis (CoMSIA) and hologram QSAR (HQSAR), to derive highly predictive QSAR models for designing novel MurF inhibitors and comparing different 3D-QSAR/alignment methods. QSAR models with high predictive ability for MurF inhibitors were successfully constructed in terms of cross-validation q2, standard error and predictive coefficient r2, which were around 0.70, 0.55 and 0.99, respectively. All the models from different methods were in good agreement with each other. Compounds with indeterminate activities were used as a test set; results showed that CoMSIA had the best predictive ability, followed by HQSAR and CoMFA. Based on these models, some key features for designing new MurF inhibitors were identified. A virtual database screen process was proposed based on the combination of these models.
Collapse
Affiliation(s)
- DE-XIN KONG
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- Shandong Provincial Research Center for Bioinformatics Engineering and Technique, Shandong University of Technology, Zibo, Shandong 255049, China
| | - WEI-LIANG ZHU
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - DA-LEI WU
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - XU SHEN
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - HUA-LIANG JIANG
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| |
Collapse
|
36
|
Shih HW, Chen KT, Cheng TJR, Wong CH, Cheng WC. A new synthetic approach toward bacterial transglycosylase substrates, Lipid II and Lipid IV. Org Lett 2011; 13:4600-3. [PMID: 21797279 DOI: 10.1021/ol201806d] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A new synthetic approach toward the bacterial transglycosylase substrates, Lipid II (1) and Lipid IV (2), is described. The key disaccharide was synthesized using the concept of relative reactivity value (RRV) and elaborated to Lipid II and Lipid IV by conjugation with the appropriate oligopeptides and pyrophosphate lipids. Interestingly, the results from our HPLC-based functional TGase assay suggested Lipid IV has a higher affinity for the enzyme than Lipid II.
Collapse
Affiliation(s)
- Hao-Wei Shih
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
37
|
Abstract
The discovery of novel small-molecule antibacterial drugs has been stalled for many years. The purpose of this review is to underscore and illustrate those scientific problems unique to the discovery and optimization of novel antibacterial agents that have adversely affected the output of the effort. The major challenges fall into two areas: (i) proper target selection, particularly the necessity of pursuing molecular targets that are not prone to rapid resistance development, and (ii) improvement of chemical libraries to overcome limitations of diversity, especially that which is necessary to overcome barriers to bacterial entry and proclivity to be effluxed, especially in Gram-negative organisms. Failure to address these problems has led to a great deal of misdirected effort.
Collapse
Affiliation(s)
- Lynn L Silver
- LL Silver Consulting, LLC, 955 S. Springfield Ave., Unit C403, Springfield, NJ 07081, USA.
| |
Collapse
|
38
|
Gautam A, Vyas R, Tewari R. Peptidoglycan biosynthesis machinery: a rich source of drug targets. Crit Rev Biotechnol 2010; 31:295-336. [PMID: 21091161 DOI: 10.3109/07388551.2010.525498] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The range of antibiotic therapy for the control of bacterial infections is becoming increasingly limited because of the rapid rise in multidrug resistance in clinical bacterial isolates. A few diseases, such as tuberculosis, which were once thought to be under control, have re-emerged as serious health threats. These problems have resulted in intensified research to look for new inhibitors for bacterial pathogens. Of late, the peptidoglycan (PG) layer, the most important component of the bacterial cell wall has been the subject of drug targeting because, first, it is essential for the survivability of eubacteria and secondly, it is absent in humans. The last decade has seen tremendous inputs in deciphering the 3-D structures of the PG biosynthetic enzymes. Many inhibitors against these enzymes have been developed using virtual and high throughput screening techniques. This review discusses the mechanistic and structural properties of the PG biosynthetic enzymes and inhibitors developed in the last decade.
Collapse
Affiliation(s)
- Ankur Gautam
- Department of Biotechnology, Panjab University, Chandigarh, India
| | | | | |
Collapse
|
39
|
Cheng TJR, Wu YT, Yang ST, Lo KH, Chen SK, Chen YH, Huang WI, Yuan CH, Guo CW, Huang LY, Chen KT, Shih HW, Cheng YSE, Cheng WC, Wong CH. High-throughput identification of antibacterials against methicillin-resistant Staphylococcus aureus (MRSA) and the transglycosylase. Bioorg Med Chem 2010; 18:8512-29. [PMID: 21075637 DOI: 10.1016/j.bmc.2010.10.036] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/11/2010] [Accepted: 10/14/2010] [Indexed: 12/01/2022]
Abstract
To identify new transglycosylase inhibitors with potent anti-methicillin-resistant Staphylococcus aureus (MRSA) activities, a high-throughput screening against Staphylococcus aureus was conducted to look for antibacterial cores in our 2M compound library that consists of natural products, proprietary collection, and synthetic molecules. About 3600 hits were identified from the primary screening and the subsequent confirmation resulted in a total of 252 compounds in 84 clusters which showed anti-MRSA activities with MIC values as low as 0.1 μg/ml. Subsequent screening targeting bacterial transglycosylase identified a salicylanilide-based core that inhibited the lipid II polymerization and the moenomycin-binding activities of transglycosylase. Among the collected analogues, potent inhibitors with the IC(50) values below 10 μM against transglycosylase were identified. The non-carbonhydrate scaffold reported in this study suggests a new direction for development of bacterial transglycosylase inhibitors.
Collapse
Affiliation(s)
- Ting-Jen Rachel Cheng
- Genomics Research Center, Academia Sinica, 128 Sec 2 Academia Road, Nankang, Taipei 115, Taiwan. Genomics Research Center, Academia Sinica, 128 Sec 2 Academia Road, Nankang, Taipei 115, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Liu CY, Guo CW, Chang YF, Wang JT, Shih HW, Hsu YF, Chen CW, Chen SK, Wang YC, Cheng TJR, Ma C, Wong CH, Fang JM, Cheng WC. Synthesis and evaluation of a new fluorescent transglycosylase substrate: lipid II-based molecule possessing a dansyl-C20 polyprenyl moiety. Org Lett 2010; 12:1608-11. [PMID: 20187630 DOI: 10.1021/ol100338v] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The preparation of a novel fluorescent lipid II-based substrate for transglycosylases (TGases) is described. This substrate has characteristic structural features including a shorter lipid chain, a fluorophore tag at the end of the lipid chain rather than on the peptide chain, and no labeling with a radioactive atom. This fluorescent substrate is readily utilized in TGase activity assays to characterize TGases and also to evaluate the activities of TGase inhibitors.
Collapse
Affiliation(s)
- Chen-Yu Liu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Dube S, Nanda K, Rani R, Kaur NJ, Nagpal JK, Upadhyay DJ, Cliffe IA, Saini KS, Purnapatre KP. UDP-N-acetylglucosamine enolpyruvyl transferase from Pseudomonas aeruginosa. World J Microbiol Biotechnol 2010. [DOI: 10.1007/s11274-010-0338-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
42
|
Shih HW, Chen KT, Chen SK, Huang CY, Cheng TJR, Ma C, Wong CH, Cheng WC. Combinatorial approach toward synthesis of small molecule libraries as bacterial transglycosylase inhibitors. Org Biomol Chem 2010; 8:2586-93. [DOI: 10.1039/c000622j] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
43
|
Basavannacharya C, Robertson G, Munshi T, Keep NH, Bhakta S. ATP-dependent MurE ligase in Mycobacterium tuberculosis: biochemical and structural characterisation. Tuberculosis (Edinb) 2009; 90:16-24. [PMID: 19945347 DOI: 10.1016/j.tube.2009.10.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 10/10/2009] [Accepted: 10/26/2009] [Indexed: 01/30/2023]
Abstract
New therapies are required against Mycobacterium tuberculosis and its cell wall peptidoglycan biosynthesis is a potential therapeutic target. UDP-MurNAc-tripeptide ligase (MurE) is a member of the ATP-dependent ligase family, which incorporate amino acids including meso-diaminopimelic acid (m-DAP) into peptidoglycan during synthesis in a species-specific manner. In the present study, we have cloned, over-expressed, and characterised MurE from M. tuberculosis (Mtb-MurE). The crystal structure has been determined at 3.0A resolution in the presence of the substrate UDP-MurNAc-l-Ala-d-Glu (UAG). The activity of the enzyme was measured through estimating inorganic phosphate released in a non-radioactive high-throughput colourimetric assay. UDP-MurNAc-l-Ala-d-Glu-m-DAP (UMT) formation coupled to inorganic phosphate release was confirmed by HPLC and mass spectrometric analyses. Kinetic constants were determined for a range of natural substrates using optimised conditions. From our findings, it is evident that Mtb-MurE is highly specific in adding m-DAP to UDP-MurNAc-dipeptide and ATP-hydrolysis is an absolute requirement for its activity.
Collapse
Affiliation(s)
- Chandrakala Basavannacharya
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, Malet Street, London WC1E 7HX, UK
| | | | | | | | | |
Collapse
|
44
|
Antibiotics from microbes: converging to kill. Curr Opin Microbiol 2009; 12:520-7. [PMID: 19695947 DOI: 10.1016/j.mib.2009.07.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Revised: 07/06/2009] [Accepted: 07/08/2009] [Indexed: 11/20/2022]
Abstract
As genetically encoded small molecules, antibiotics are phenotypes that have resulted from mutation and natural selection. Advances in genetics, biochemistry, and bioinformatics have connected hundreds of antibiotics to the gene clusters that encode them, allowing these molecules to be analyzed using the tools of evolutionary biology. This review surveys examples of convergent evolution from microbially produced antibiotics, including the convergence of distinct gene clusters on similar phenotypes and the merger of distinct gene clusters into a single functional unit. Examining antibiotics through an evolutionary lens highlights the versatility of biosynthetic pathways, reveals lessons for combating antibiotic resistance, and provides an entry point for studying the natural roles of these natural products.
Collapse
|
45
|
Huber J, Donald RG, Lee SH, Jarantow LW, Salvatore MJ, Meng X, Painter R, Onishi RH, Occi J, Dorso K, Young K, Park YW, Skwish S, Szymonifka MJ, Waddell TS, Miesel L, Phillips JW, Roemer T. Chemical Genetic Identification of Peptidoglycan Inhibitors Potentiating Carbapenem Activity against Methicillin-Resistant Staphylococcus aureus. ACTA ACUST UNITED AC 2009; 16:837-48. [DOI: 10.1016/j.chembiol.2009.05.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 05/25/2009] [Accepted: 05/28/2009] [Indexed: 10/20/2022]
|
46
|
Abstract
AbstractThe development of antimicrobial resistance by bacteria is inevitable and is considered as a major problem in the treatment of bacterial infections in the hospital and in the community. Despite efforts to develop new therapeutics that interact with new targets, resistance has been reported even to these agents. In this review, an overview is given of the many therapeutic possibilities that exist for treatment of bacterial infections and how bacteria become resistant to these therapeutics.
Collapse
|
47
|
MurF inhibitors with antibacterial activity: effect on muropeptide levels. Antimicrob Agents Chemother 2009; 53:3240-7. [PMID: 19470511 DOI: 10.1128/aac.00166-09] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
MurF catalyzes the last cytoplasmic step of bacterial cell wall synthesis and is essential for bacterial survival. Our previous studies used a pharmacophore model of a MurF inhibitor to identify additional inhibitors with improved properties. We now present the characterization of two such inhibitors, the diarylquinolines DQ1 and DQ2. DQ1 inhibited Escherichia coli MurF (50% inhibitory concentration, 24 microM) and had modest activity (MICs, 8 to 16 microg/ml) against lipopolysaccharide (LPS)-defective E. coli and wild-type E. coli rendered permeable with polymyxin B nonapeptide. DQ2 additionally displayed activity against gram-positive bacteria (MICs, 8 to 16 microg/ml), including methicillin (meticillin)-susceptible and -resistant Staphylococcus aureus isolates and vancomycin-susceptible and -resistant Enterococcus faecalis and Enterococcus faecium isolates. Treatment of LPS-defective E. coli cells with >or=2x MIC of DQ1 resulted in a 75-fold-greater accumulation of the MurF substrate compared to the control, a 70% decline in the amount of the MurF product, and eventual cell lysis, consistent with the inhibition of MurF within bacteria. DQ2 treatment of S. aureus resulted in similar effects on the MurF substrate and product quantities. At lower levels of DQ1 (<or=1x MIC), the level of accumulation of the substrate was less pronounced (15-fold greater compared to the amount for the control). However, a 50% increase in the amount of the MurF product compared to the control was reproducibly observed, consistent with the possible upregulation of muropeptide biosynthesis upon partial inhibition of this pathway. The overexpression of cloned MurF appeared to partly alleviate the DQ1-mediated inhibition of muropeptide synthesis. The identification of MurF inhibitors such as DQ1 and DQ2 that disrupt cell wall biosynthesis suggests that MurF remains a viable target for an antibacterial agent.
Collapse
|
48
|
Paradis-Bleau C, Lloyd A, Sanschagrin F, Clarke T, Blewett A, Bugg TDH, Levesque RC. Phage display-derived inhibitor of the essential cell wall biosynthesis enzyme MurF. BMC BIOCHEMISTRY 2008; 9:33. [PMID: 19099588 PMCID: PMC2626591 DOI: 10.1186/1471-2091-9-33] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Accepted: 12/19/2008] [Indexed: 11/10/2022]
Abstract
BACKGROUND To develop antibacterial agents having novel modes of action against bacterial cell wall biosynthesis, we targeted the essential MurF enzyme of the antibiotic resistant pathogen Pseudomonas aeruginosa. MurF catalyzes the formation of a peptide bond between D-Alanyl-D-Alanine (D-Ala-D-Ala) and the cell wall precursor uridine 5'-diphosphoryl N-acetylmuramoyl-L-alanyl-D-glutamyl-meso-diaminopimelic acid (UDP-MurNAc-Ala-Glu-meso-A2pm) with the concomitant hydrolysis of ATP to ADP and inorganic phosphate, yielding UDP-N-acetylmuramyl-pentapeptide. As MurF acts on a dipeptide, we exploited a phage display approach to identify peptide ligands having high binding affinities for the enzyme. RESULTS Screening of a phage display 12-mer library using purified P. aeruginosa MurF yielded to the identification of the MurFp1 peptide. The MurF substrate UDP-MurNAc-Ala-Glumeso-A2pm was synthesized and used to develop a sensitive spectrophotometric assay to quantify MurF kinetics and inhibition. MurFp1 acted as a weak, time-dependent inhibitor of MurF activity but was a potent inhibitor when MurF was pre-incubated with UDP-MurNAc-Ala-Glu-meso-A2pm or ATP. In contrast, adding the substrate D-Ala-D-Ala during the pre-incubation nullified the inhibition. The IC50 value of MurFp1 was evaluated at 250 microM, and the Ki was established at 420 microM with respect to the mixed type of inhibition against D-Ala-D-Ala. CONCLUSION MurFp1 exerts its inhibitory action by interfering with the utilization of D-Ala-D-Ala by the MurF amide ligase enzyme. We propose that MurFp1 exploits UDP-MurNAc-Ala-Glu-meso-A2pm-induced structural changes for better interaction with the enzyme. We present the first peptide inhibitor of MurF, an enzyme that should be exploited as a target for antimicrobial drug development.
Collapse
|
49
|
Humljan J, Kotnik M, Contreras-Martel C, Blanot D, Urleb U, Dessen A, Šolmajer T, Gobec S. Novel Naphthalene-N-sulfonyl-d-glutamic Acid Derivatives as Inhibitors of MurD, a Key Peptidoglycan Biosynthesis Enzyme,. J Med Chem 2008; 51:7486-94. [DOI: 10.1021/jm800762u] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jan Humljan
- Drug Discovery, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia, Laboratoire des Protéines Membranaires, Institut de Biologie Structurale Jean-Pierre Ebel, CEA, CNRS, UJF, UMR5075, 41 Rue Jules Horowitz, F-38027 Grenoble, France, Enveloppes Bactériennes et Antibiotiques, IBBMC, UMR 8619 CNRS, Univ Paris-Sud, 91405 Orsay, France, and Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Miha Kotnik
- Drug Discovery, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia, Laboratoire des Protéines Membranaires, Institut de Biologie Structurale Jean-Pierre Ebel, CEA, CNRS, UJF, UMR5075, 41 Rue Jules Horowitz, F-38027 Grenoble, France, Enveloppes Bactériennes et Antibiotiques, IBBMC, UMR 8619 CNRS, Univ Paris-Sud, 91405 Orsay, France, and Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Carlos Contreras-Martel
- Drug Discovery, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia, Laboratoire des Protéines Membranaires, Institut de Biologie Structurale Jean-Pierre Ebel, CEA, CNRS, UJF, UMR5075, 41 Rue Jules Horowitz, F-38027 Grenoble, France, Enveloppes Bactériennes et Antibiotiques, IBBMC, UMR 8619 CNRS, Univ Paris-Sud, 91405 Orsay, France, and Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Didier Blanot
- Drug Discovery, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia, Laboratoire des Protéines Membranaires, Institut de Biologie Structurale Jean-Pierre Ebel, CEA, CNRS, UJF, UMR5075, 41 Rue Jules Horowitz, F-38027 Grenoble, France, Enveloppes Bactériennes et Antibiotiques, IBBMC, UMR 8619 CNRS, Univ Paris-Sud, 91405 Orsay, France, and Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Uroš Urleb
- Drug Discovery, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia, Laboratoire des Protéines Membranaires, Institut de Biologie Structurale Jean-Pierre Ebel, CEA, CNRS, UJF, UMR5075, 41 Rue Jules Horowitz, F-38027 Grenoble, France, Enveloppes Bactériennes et Antibiotiques, IBBMC, UMR 8619 CNRS, Univ Paris-Sud, 91405 Orsay, France, and Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Andréa Dessen
- Drug Discovery, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia, Laboratoire des Protéines Membranaires, Institut de Biologie Structurale Jean-Pierre Ebel, CEA, CNRS, UJF, UMR5075, 41 Rue Jules Horowitz, F-38027 Grenoble, France, Enveloppes Bactériennes et Antibiotiques, IBBMC, UMR 8619 CNRS, Univ Paris-Sud, 91405 Orsay, France, and Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Tom Šolmajer
- Drug Discovery, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia, Laboratoire des Protéines Membranaires, Institut de Biologie Structurale Jean-Pierre Ebel, CEA, CNRS, UJF, UMR5075, 41 Rue Jules Horowitz, F-38027 Grenoble, France, Enveloppes Bactériennes et Antibiotiques, IBBMC, UMR 8619 CNRS, Univ Paris-Sud, 91405 Orsay, France, and Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- Drug Discovery, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia, Laboratoire des Protéines Membranaires, Institut de Biologie Structurale Jean-Pierre Ebel, CEA, CNRS, UJF, UMR5075, 41 Rue Jules Horowitz, F-38027 Grenoble, France, Enveloppes Bactériennes et Antibiotiques, IBBMC, UMR 8619 CNRS, Univ Paris-Sud, 91405 Orsay, France, and Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
50
|
Tomasić T, Zidar N, Rupnik V, Kovac A, Blanot D, Gobec S, Kikelj D, Masic LP. Synthesis and biological evaluation of new glutamic acid-based inhibitors of MurD ligase. Bioorg Med Chem Lett 2008; 19:153-7. [PMID: 19014883 DOI: 10.1016/j.bmcl.2008.10.129] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 10/28/2008] [Accepted: 10/29/2008] [Indexed: 12/18/2022]
Abstract
Mur ligases catalyze the biosynthesis of the UDP-MurNAc-pentapeptide precursor of peptidoglycan, an essential polymer of bacterial cell-wall. They constitute attractive targets for the development of novel antibacterial agents. Here we report on the synthesis of a series of 2,4-diaminoquinazolines, quinazoline-2,4(1H,3H)-diones, 5-benzylidenerhodanines and 5-benzylidenethiazolidine-2,4-diones and their inhibitory activities against MurD from Escherichia coli. Compounds (R)-27 and (S)-27 showed inhibitory activity against MurD with IC(50) values of 174 and 206 microM, respectively, which makes them promising starting points for optimization.
Collapse
Affiliation(s)
- Tihomir Tomasić
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | | | | | | | | | | | | | | |
Collapse
|