1
|
Si Z, Sun Y, Tan C, Ooi YJ, Li M, Raju C, Shubi J, Gan YH, Zhu Y, Li P, Chan-Park MB, Pethe K. A cationic main-chain poly(carbonate-imidazolium) potent against Mycobacterium abscessus and other resistant bacteria in mice. Biomaterials 2025; 316:123003. [PMID: 39709850 DOI: 10.1016/j.biomaterials.2024.123003] [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: 08/16/2024] [Revised: 10/23/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024]
Abstract
The incidence of serious lung infections due to Mycobacterium abscessus, a worrying non-tuberculosis mycobacteria (NTM) species, is rising and has in some countries surpassed tuberculosis. NTM are ubiquitous in the environment and can cause serious lung infections in people who are immunocompromised or have pre-existing lung conditions. M. abscessus is intrinsically resistant to most antibiotics. Current treatments involve combination of three or more repurposed antibiotics with the treatment regimen lasting at least 12 months but producing unsatisfactory success rates of less than 50 %. Herein, we report an alternative strategy using a degradable polymer, specifically main-chain cationic carbonate-imidazolium-derived polymer (MCOP-1). MCOP-1 is a non-toxic agent active in a murine lung infection model. MCOP-1 also exhibits excellent efficacy against multi-drug resistant (MDR) ESKAPE bacteria. MCOP-1 damages bacterial membrane and DNA, and serial passaging does not rapidly elicit resistance. Its carbonate linkage is stable enough to allow MCOP-1 to remain intact for long enough to exert its bactericidal effect but is labile over longer time periods to degrade into non-toxic small molecules. These findings underscore the potential of degradable MCOP-1 as a promising therapeutic antimicrobial agent to address the growing incidence of recalcitrant infections due to M. abscessus and MDR ESKAPE bacteria.
Collapse
Affiliation(s)
- Zhangyong Si
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637459, Singapore; Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315300, China
| | - Yan Sun
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
| | - Chongyun Tan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637459, Singapore
| | - Ying Jie Ooi
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637459, Singapore
| | - Ming Li
- Infectious Diseases Translational Research Programme, Department of Biochemistry, National University of Singapore, 117596, Singapore
| | - Cheerlavancha Raju
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637459, Singapore
| | - Jamal Shubi
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637459, Singapore
| | - Yunn-Hwen Gan
- Infectious Diseases Translational Research Programme, Department of Biochemistry, National University of Singapore, 117596, Singapore
| | - Yabin Zhu
- Health Science Center, Ningbo University, 315211, China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics, Northwestern Polytechnical University, 710072, China
| | - Mary B Chan-Park
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637459, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore.
| | - Kevin Pethe
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore; Singapore Centre for Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, 637551, Singapore; National Center for Infectious Diseases (NCID), 308442, Singapore.
| |
Collapse
|
2
|
Ragab A, Ibrahim SA, Aboul-Magd DS, Baren MH. One-pot synthesis of pyrazolo[4,3- d]thiazole derivatives containing α-aminophosphonate as potential Mur A inhibitors against MDR pathogens with radiosterilization and molecular modeling simulation. RSC Adv 2023; 13:34756-34771. [PMID: 38035237 PMCID: PMC10685179 DOI: 10.1039/d3ra07040a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023] Open
Abstract
The present study involves the synthesis of a new series of α-aminophosphonate derivatives in good yields with a simple workup via the Kabachnik-Fields reaction using lithium perchlorate (LiClO4) as a catalyst to facilitate the reaction. All the newly synthesized compounds were confirmed using various physical, spectroscopic, and analytical data, and the obtained results correlated with the proposed molecular structure. The in vitro antimicrobial activities of each compound were evaluated against different clinical isolates. The results indicated that among these derivatives, two compounds (5a and 5b) were the most active and displayed potent activity with MICs in the range from 0.06 to 0.25 μg mL-1 compared with fosfomycin and fluconazole as standard antibiotics. Moreover, the synthesized phosphonates displayed a broad spectrum of bactericidal and fungicidal activities depending on MICs, MBCs/MFCs, and the time-kill kinetics. In addition, the checkerboard assay showed synergistic and partial synergistic activities between the active compounds combined with fosfomycin and fluconazole. Furthermore, the SEM images showed distinct ruptures of the OM integrity of the FOS-R E. coli at their MICs, which was further indicated by the increased EtBr accumulation within the bacterial cells. Moreover, active derivatives revealed MurA inhibitory activity with IC50 values of 3.8 ± 0.39 and 4.5 ± 0.23 μM compared with fosfomycin (IC50 = 12.7 ± 0.27 μM). To our surprise, exposing 5a and 5b compounds to different gamma radiation doses revealed that 7.0 kGy eradicated the microbial load completely. Finally, the results of quantum chemical study supported the binding mode obtained from the docking study performed inside the active site of MurA (PDB: 1UAE), suggesting that these phosphonates may be promising safe candidates for MDR infection therapy clinical trials with no toxic effects on the normal human cells.
Collapse
Affiliation(s)
- Ahmed Ragab
- Chemistry Department, Faculty of Science (Boys), Al-Azhar University Nasr City Cairo 11884 Egypt
| | - Seham A Ibrahim
- Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| | - Dina S Aboul-Magd
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority Egypt
| | - Mohamed H Baren
- Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| |
Collapse
|
3
|
Zhang H, Chen Q, Xie J, Cong Z, Cao C, Zhang W, Zhang D, Chen S, Gu J, Deng S, Qiao Z, Zhang X, Li M, Lu Z, Liu R. Switching from membrane disrupting to membrane crossing, an effective strategy in designing antibacterial polypeptide. SCIENCE ADVANCES 2023; 9:eabn0771. [PMID: 36696494 PMCID: PMC9876554 DOI: 10.1126/sciadv.abn0771] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/27/2022] [Indexed: 06/17/2023]
Abstract
Drug-resistant bacterial infections have caused serious threats to human health and call for effective antibacterial agents that have low propensity to induce antimicrobial resistance. Host defense peptide-mimicking peptides are actively explored, among which poly-β-l-lysine displays potent antibacterial activity but high cytotoxicity due to the helical structure and strong membrane disruption effect. Here, we report an effective strategy to optimize antimicrobial peptides by switching membrane disrupting to membrane penetrating and intracellular targeting by breaking the helical structure using racemic residues. Introducing β-homo-glycine into poly-β-lysine effectively reduces the toxicity of resulting poly-β-peptides and affords the optimal poly-β-peptide, βLys50HG50, which shows potent antibacterial activity against clinically isolated methicillin-resistant Staphylococcus aureus (MRSA) and MRSA persister cells, excellent biosafety, no antimicrobial resistance, and strong therapeutic potential in both local and systemic MRSA infections. The optimal poly-β-peptide demonstrates strong therapeutic potential and implies the success of our approach as a generalizable strategy in designing promising antibacterial polypeptides.
Collapse
Affiliation(s)
- Haodong Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jiayang Xie
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Zihao Cong
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Chuntao Cao
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wenjing Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Donghui Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Sheng Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jiawei Gu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Shuai Deng
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Zhongqian Qiao
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xinyue Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Maoquan Li
- Department of Interventional and Vascular Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ziyi Lu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
4
|
Dale AG, Porcu A, Mann J, Neidle S. The mechanism of resistance in Escherichia coli to ridinilazole and other antibacterial head-to-head bis-benzimidazole compounds. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02918-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AbstractThe appY gene has been characterised as conferring resistance to a novel series of antimicrobial benzimidazole derivatives in E. coli MC1061 cells when expressed in high copy-number. A microarray approach was used to identify genes involved in the mechanism of appY-mediated antibacterial resistance, that were up- or down-regulated following induction of the gene in the appY knockout strain JW0553. In total, expression of 90 genes was induced and 48 repressed greater than 2.5-fold (P < 0.05), 45 min after appY induction. Over half the genes up-regulated following appY expression had confirmed or putative roles in acid resistance (AR) and response to oxidative and antibiotic stresses. These included the genes for MdtE and MdtF, which form a multi-drug transporter with TolC and have been implicated in resistance to several antibiotics including erythromycin. Amongst the acid resistance genes were gadAB and adiAC encoding the glutamate-dependant (AR2) and arginine-dependant (AR3) acid resistance systems respectively, in addition to the transcriptional activators of these systems gadE and gadX. In agreement with earlier studies, appA, appCB and hyaA-F were also up-regulated following induction of appY. This study has also confirmed that over-expression of mdtEF confers resistance to these antibacterial benzimidazoles, indicating that the observation of appY conferring resistance to these compounds, proceeds through an appY-mediated up-regulation of this efflux transporter. To assess the importance of the AppY enzyme to acid stress responses, the percentage survival of bacteria in acidified media (pH ≤ 2) was measured. From an initial input of 1 × 106 CFU/ml, the wild-type strain MG1655 showed 7.29% and 0.46% survival after 2 and 4 h, respectively. In contrast, strain JW0553 in which appY is deleted was completely killed by the treatment. Transformation of JW0553 with a plasmid carrying appY returned survival to wild-type levels (7.85% and 1.03% survival at 2 and 4 h). Further dissection of the response by prior induction of each of the three AR systems has revealed that AR1 and AR3 were most affected by the absence of appY. This work highlights an important and previously unidentified role for the AppY enzyme in mediating the responses to several stress conditions. It is likely that the appY gene fits into a complex transcriptional regulatory network involving σS and gadE and gadX. Further work to pinpoint its position in such a hierarchy and to assess the contribution of appY to oxidative stress responses should help determine its full significance. This work is also consistent with recent studies in C. difficile showing that the mechanism of action of ridinilazole involves AT-rich DNA minor groove binding.
Collapse
|
5
|
Xie J, Zhou M, Qian Y, Cong Z, Chen S, Zhang W, Jiang W, Dai C, Shao N, Ji Z, Zou J, Xiao X, Liu L, Chen M, Li J, Liu R. Addressing MRSA infection and antibacterial resistance with peptoid polymers. Nat Commun 2021; 12:5898. [PMID: 34625571 PMCID: PMC8501045 DOI: 10.1038/s41467-021-26221-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 09/17/2021] [Indexed: 01/21/2023] Open
Abstract
Methicillin-Resistant Staphylococcus aureus (MRSA) induced infection calls for antibacterial agents that are not prone to antimicrobial resistance. We prepare protease-resistant peptoid polymers with variable C-terminal functional groups using a ring-opening polymerization of N-substituted N-carboxyanhydrides (NNCA), which can provide peptoid polymers easily from the one-pot synthesis. We study the optimal polymer that displays effective activity against MRSA planktonic and persister cells, effective eradication of highly antibiotic-resistant MRSA biofilms, and potent anti-infectious performance in vivo using the wound infection model, the mouse keratitis model, and the mouse peritonitis model. Peptoid polymers show insusceptibility to antimicrobial resistance, which is a prominent merit of these antimicrobial agents. The low cost, convenient synthesis and structure diversity of peptoid polymers, the superior antimicrobial performance and therapeutic potential in treating MRSA infection altogether imply great potential of peptoid polymers as promising antibacterial agents in treating MRSA infection and alleviating antibiotic resistance. Antibiotic resistance is a major issue in medicine and new antimicrobials for treating resistant infection are needed. Here, the authors report on antibacterial peptoid polymers, prepared via NNCA ring-opening polymerization, demonstrating antibacterial function against MRSA in vitro and in in vivo infection models.
Collapse
Affiliation(s)
- Jiayang Xie
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Min Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Yuxin Qian
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Zihao Cong
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Sheng Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Wenjing Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Weinan Jiang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Chengzhi Dai
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Ning Shao
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Zhemin Ji
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Jingcheng Zou
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Ximian Xiao
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Longqiang Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Minzhang Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China
| | - Jin Li
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 200237, Shanghai, China. .,Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, 200237, Shanghai, China.
| |
Collapse
|
6
|
Zha GF, Preetham HD, Rangappa S, Sharath Kumar KS, Girish YR, Rakesh KP, Ashrafizadeh M, Zarrabi A, Rangappa KS. Benzimidazole analogues as efficient arsenals in war against methicillin-resistance staphylococcus aureus (MRSA) and its SAR studies. Bioorg Chem 2021; 115:105175. [PMID: 34298242 DOI: 10.1016/j.bioorg.2021.105175] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/11/2021] [Indexed: 12/19/2022]
Abstract
Small molecule based inhibitors development is a growing field in medicinal chemistry. In recent years, different heterocyclic derivatives have been designed to counter the infections caused by multi-drug resistant bacteria. Indeed, small molecule inhibitors can be employed as an efficient antibacterial agents with different mechanism of action. Methicillin-resistant Staphylococcus aureus (MRSA) is becoming lethal to mankind due to easy transmission mode, rapid resistance development to existing antibiotics and affect difficult-to-treat skin and filmsy diseases. Benzimidazoles are a class of heterocyclic compounds which have capability to fight against MRSA. High biocompatibility of benzimidazoles, synergistic behaviour with antibiotics and their tunable physico-chemical properties attracted the researchers to develop new benzimidazole based antibacterial agents. The present review focus on recent developments of benzimidazole-hybrid molecules as anti MRSA agents and the results of in-vitro and in-vivo studies with possible mechanism of action and discussing structure-activity relationship (SAR) in different directions. Benzimdazoles act as DNA binding agents, enzyme inhibitors, anti-biofilm agents and showed synergistic effect with available antibiotics to achieve antibacterial activity against MRSA. This cumulative figures would help to design new benzimidazole-based MRSA growth inhibitors.
Collapse
Affiliation(s)
- Gao-Feng Zha
- Scientific Research Centre, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhan 518107, China.
| | - Habbanakuppe D Preetham
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysuru 570 006, India
| | - Shobith Rangappa
- Adichunchanagiri Institute for Molecular Medicine, Adichunchanagiri Institute of Medical Sciences, Adichunchanagiri University, B. G. Nagar, Nagamangala Taluk, Mandya District 571448, India
| | | | - Yarabahally R Girish
- Centre for Research and Innovations, School of Natural Sciences, BGSIT, Adichunchanagiri University, B. G. Nagara, Mandya, 571448, India
| | - Kadalipura P Rakesh
- School of Material Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
| | | |
Collapse
|
7
|
Bisbenzimidazole Derivatives as Potential Antimicrobial Agents: Design, Synthesis, Biological Evaluation and Pharmacophore Analysis. Pharm Chem J 2021. [DOI: 10.1007/s11094-021-02389-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
8
|
Gregory-Bass R, Winfield LL. Preliminary Analysis of Anti-proliferative, Apoptotic, and Anti-migratory Effects llw-3-6 in Skov-3 Ovarian Cystadenocarcinoma Cell Line. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817666200129142949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Ovarian cancer and ovarian related diseases affect reproductive health.
Therapeutic molecules are needed to improve treatment outcomes and overcome drug resistance.
The benzimidazole-based sulphonamide LLW-3-6 has both anti-apoptotic and anti-proliferative
effects when used to treat prostate, breast, and brain cancer cells.
Objective:
The study described herein evaluates the anti-proliferative and anti-migratory effects of
LLW-3-6 in SKOV-3 ovarian cystadenocarcinoma cell line.
Methods:
Studies were conducted using SKOV-3 cells treated with LLW-3-6. The cell line was
propagated and proliferative activity was evaluated by hemocytometric and MTT colorimetric
assays. Cellular apoptosis was assessed using caspase-3 spectrophotometric analysis. Lastly, a
scratch wound assay was conducted at several concentrations and time points to assess the effect of
LLW-3-6 on migration.
Results & Conclusion:
Proliferative studies suggest, SKOV-3 cells exposed to LLW-3-6 in culture
resulted in decreased growth and proliferation of cells in a time and dose-dependent manner. The
apoptotic effect of this agent was noted with the confirmed presence of Caspase-3 in a dose and
time-dependent manner as well. Preliminary studies also suggest an anti-migratory effect of LLW-3-
6, confirmed by scratch wound analysis.
Conclusion:
LLW-3-6 is potentially a chemotherapeutic option for decreasing proliferation and
inducing apoptosis in ovarian carcinomas. Additional biological analysis are ongoing to further
assess the utility of the molecule and its mechanism of action.
Collapse
Affiliation(s)
- Rosalind Gregory-Bass
- Department of Environmental and Health Sciences, Spelman College, Atlanta, GA, United States
| | - Leyte L. Winfield
- Department of Chemistry & Biochemistry, Spelman College, Atlanta, GA, United States
| |
Collapse
|
9
|
Yadav N, Agarwal D, Kumar S, Dixit AK, Gupta RD, Awasthi SK. In vitro antiplasmodial efficacy of synthetic coumarin-triazole analogs. Eur J Med Chem 2018; 145:735-745. [PMID: 29366931 DOI: 10.1016/j.ejmech.2018.01.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/05/2018] [Accepted: 01/05/2018] [Indexed: 11/24/2022]
Abstract
Twenty two diverse coumarin-triazole derivatives were synthesized by alkylation of 7-hydroxy-4-methyl-coumarin followed by click chemistry at 7-position. These compounds were evaluated for their in vitro antiplasmodial activity against chloroquine sensitive strain of Plasmodium falciparum (3D7). Compound 9 (7-[1-(2, 4-dimethoxy-phenyl)-1H- [1-3] triazol-4-ylmethoxy]-4-methyl-chromen-2-one) was found most active with IC50 value 0.763 ± 0.0124 μg/mL. Further, the structure of compound 20 was characterized by single crystal X-ray diffraction. In view of impressive results, we considered it worthwhile to validate the results of in vitro antiplasmodial activity by assessing whether these compounds are capable of hampering the catalytic activity of DNA gyrase, thus preventing its supercoiling function.
Collapse
Affiliation(s)
- Neesha Yadav
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Drishti Agarwal
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Satyanand Kumar
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - A K Dixit
- V S S D College, Kanpur University, Kanpur, India
| | - Rinkoo D Gupta
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India
| | - Satish K Awasthi
- Chemical Biology Laboratory, Department of Chemistry, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
10
|
CopperII phthalocyanine as an efficient and reusable catalyst for the N-arylation of nitrogen containing heterocycles. Tetrahedron Lett 2017. [DOI: 10.1016/j.tetlet.2017.06.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Picconi P, Hind C, Jamshidi S, Nahar K, Clifford M, Wand ME, Sutton JM, Rahman KM. Triaryl Benzimidazoles as a New Class of Antibacterial Agents against Resistant Pathogenic Microorganisms. J Med Chem 2017. [PMID: 28650661 DOI: 10.1021/acs.jmedchem.7b00108] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A new class of nontoxic triaryl benzimidazole compounds, derived from existing classes of DNA minor groove binders, were designed, synthesized, and evaluated for their antibacterial activity against multidrug resistant (MDR) Gram-positive and Gram-negative species. Molecular modeling experiments suggest that the newly synthesized class cannot be accommodated within the minor groove of DNA due to a change in the shape of the molecules. Compounds 8, 13, and 14 were found to be the most active of the series, with MICs in the range of 0.5-4 μg/mL against the MDR Staphylococci and Enterococci species. Compound 13 showed moderate activity against the MDR Gram-negative strains, with MICs in the range of 16-32 μg/mL. Active compounds showed a bactericidal mode of action, and a mechanistic study suggested the inhibition of bacterial gyrase as the mechanism of action (MOA) of this chemical class. The MOA was further supported by the molecular modeling study.
Collapse
Affiliation(s)
- Pietro Picconi
- Institute of Pharmaceutical Science, King's College London , London SE1 1DB, U.K
| | - Charlotte Hind
- National Infections Service, Porton Down, Public Health England , Salisbury SP4 0JG, Wiltshire U.K
| | - Shirin Jamshidi
- Institute of Pharmaceutical Science, King's College London , London SE1 1DB, U.K
| | - Kazi Nahar
- Institute of Pharmaceutical Science, King's College London , London SE1 1DB, U.K
| | - Melanie Clifford
- National Infections Service, Porton Down, Public Health England , Salisbury SP4 0JG, Wiltshire U.K
| | - Matthew E Wand
- National Infections Service, Porton Down, Public Health England , Salisbury SP4 0JG, Wiltshire U.K
| | - J Mark Sutton
- National Infections Service, Porton Down, Public Health England , Salisbury SP4 0JG, Wiltshire U.K
| | | |
Collapse
|
12
|
Ranjan N, Story S, Fulcrand G, Leng F, Ahmad M, King A, Sur S, Wang W, Tse-Dinh YC, Arya DP. Selective Inhibition of Escherichia coli RNA and DNA Topoisomerase I by Hoechst 33258 Derived Mono- and Bisbenzimidazoles. J Med Chem 2017; 60:4904-4922. [DOI: 10.1021/acs.jmedchem.7b00191] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Nihar Ranjan
- Laboratory
of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Sandra Story
- NUBAD LLC, 900B West Faris
Road, Greenville, South Carolina 29605, United States
| | - Geraldine Fulcrand
- Department
of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
- Biomolecular
Sciences Institute, Florida International University, Miami, Florida 33199, United States
| | - Fenfei Leng
- Department
of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
- Biomolecular
Sciences Institute, Florida International University, Miami, Florida 33199, United States
| | - Muzammil Ahmad
- Genome
Instability and Chromatin Remodeling Section, Lab of Genetics, National
Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, Maryland 21224, United States
| | - Ada King
- NUBAD LLC, 900B West Faris
Road, Greenville, South Carolina 29605, United States
| | - Souvik Sur
- Laboratory
of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Weidong Wang
- Genome
Instability and Chromatin Remodeling Section, Lab of Genetics, National
Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, Maryland 21224, United States
| | - Yuk-Ching Tse-Dinh
- Department
of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
- Biomolecular
Sciences Institute, Florida International University, Miami, Florida 33199, United States
| | - Dev P. Arya
- Laboratory
of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
- NUBAD LLC, 900B West Faris
Road, Greenville, South Carolina 29605, United States
| |
Collapse
|
13
|
The Current Case of Quinolones: Synthetic Approaches and Antibacterial Activity. Molecules 2016; 21:268. [PMID: 27043501 PMCID: PMC6274096 DOI: 10.3390/molecules21040268] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 11/17/2022] Open
Abstract
Quinolones are broad-spectrum synthetic antibacterial drugs first obtained during the synthesis of chloroquine. Nalidixic acid, the prototype of quinolones, first became available for clinical consumption in 1962 and was used mainly for urinary tract infections caused by Escherichia coli and other pathogenic Gram-negative bacteria. Recently, significant work has been carried out to synthesize novel quinolone analogues with enhanced activity and potential usage for the treatment of different bacterial diseases. These novel analogues are made by substitution at different sites--the variation at the C-6 and C-8 positions gives more effective drugs. Substitution of a fluorine atom at the C-6 position produces fluroquinolones, which account for a large proportion of the quinolones in clinical use. Among others, substitution of piperazine or methylpiperazine, pyrrolidinyl and piperidinyl rings also yields effective analogues. A total of twenty six analogues are reported in this review. The targets of quinolones are two bacterial enzymes of the class II topoisomerase family, namely gyrase and topoisomerase IV. Quinolones increase the concentration of drug-enzyme-DNA cleavage complexes and convert them into cellular toxins; as a result they are bactericidal. High bioavailability, relative low toxicity and favorable pharmacokinetics have resulted in the clinical success of fluoroquinolones and quinolones. Due to these superior properties, quinolones have been extensively utilized and this increased usage has resulted in some quinolone-resistant bacterial strains. Bacteria become resistant to quinolones by three mechanisms: (1) mutation in the target site (gyrase and/or topoisomerase IV) of quinolones; (2) plasmid-mediated resistance; and (3) chromosome-mediated quinolone resistance. In plasmid-mediated resistance, the efflux of quinolones is increased along with a decrease in the interaction of the drug with gyrase (topoisomerase IV). In the case of chromosome-mediated quinolone resistance, there is a decrease in the influx of the drug into the cell.
Collapse
|
14
|
Song D, Ma S. Recent Development of Benzimidazole-Containing Antibacterial Agents. ChemMedChem 2016; 11:646-59. [PMID: 26970352 DOI: 10.1002/cmdc.201600041] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 02/20/2016] [Indexed: 11/11/2022]
Abstract
Clinically significant antibiotic resistance is one of the greatest challenges of the twenty-first century. However, new antibacterial agents are currently being developed at a much slower pace than our growing need for such drugs. Given their diverse biological activities and clinical applications, many bioactive heterocyclic compounds containing a benzimidazole nucleus have been the focus of interest for many researchers. The benzimidazole nucleus is a structural isostere of naturally occurring nucleotides. This advantage allows benzimidazoles to readily interact with the various biopolymers found in living systems. In view of this situation, much attention has been given to the exploration of benzimidazole-based antibacterial agents, leading to the discovery of many new chemical entities with intriguing profiles. In this minireview we summarize novel benzimidazole derivatives active against various bacterial strains. In particular, we outline the relationship between the structures of variously modified benzimidazoles and their antibacterial activity.
Collapse
Affiliation(s)
- Di Song
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, 250012, P.R. China
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, 250012, P.R. China.
| |
Collapse
|
15
|
Li M, Lai L, Zhao Z, Chen T. Aquation Is a Crucial Activation Step for Anticancer Action of Ruthenium(II) Polypyridyl Complexes to Trigger Cancer Cell Apoptosis. Chem Asian J 2015; 11:310-20. [DOI: 10.1002/asia.201501048] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 10/12/2015] [Indexed: 02/01/2023]
Affiliation(s)
- Meng Li
- Department of Chemistry; Jinan University; Guangzhou 510631 P. R China
| | - Lanhai Lai
- Department of Chemistry; Jinan University; Guangzhou 510631 P. R China
| | - Zhennan Zhao
- Department of Chemistry; Jinan University; Guangzhou 510631 P. R China
| | - Tianfeng Chen
- Department of Chemistry; Jinan University; Guangzhou 510631 P. R China
| |
Collapse
|
16
|
Mann J, Taylor PW, Dorgan CR, Johnson PD, Wilson FX, Vickers R, Dale AG, Neidle S. The discovery of a novel antibiotic for the treatment of Clostridium difficile infections: a story of an effective academic-industrial partnership. MEDCHEMCOMM 2015; 6:1420-1426. [PMID: 26949507 PMCID: PMC4756575 DOI: 10.1039/c5md00238a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/30/2015] [Indexed: 12/13/2022]
Abstract
The story of the discovery of the bis-benzimidazole derivative SMT19969, which is currently in clinical trials against the pathogen Clostridium difficile.
Academic drug discovery is playing an increasingly important role in the identification of new therapies for a wide range of diseases. There is no one model that guarantees success. We describe here a drug discovery story where chance, the ability to capitalise on chance, and the assembling of a range of expertise, have all played important roles in the discovery and subsequent development of an antibiotic chemotype based on the bis-benzimidazole scaffold, with potency against a number of current therapeutically challenging diseases. One compound in this class, SMT19969, has recently entered Phase 2 human clinical trials for the treatment of Clostridium difficile infections.
Collapse
Affiliation(s)
- John Mann
- UCL School of Pharmacy , University College London , London WC1N 1AX , UK .
| | - Peter W Taylor
- UCL School of Pharmacy , University College London , London WC1N 1AX , UK .
| | | | | | | | | | - Aaron G Dale
- UCL School of Pharmacy , University College London , London WC1N 1AX , UK .
| | - Stephen Neidle
- UCL School of Pharmacy , University College London , London WC1N 1AX , UK .
| |
Collapse
|
17
|
Neidle S. A Personal History of Quadruplex-Small Molecule Targeting. CHEM REC 2015; 15:691-710. [DOI: 10.1002/tcr.201500011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Indexed: 01/15/2023]
Affiliation(s)
- Stephen Neidle
- UCL School of Pharmacy; University College London; 29-39 Brunswick Square London WC1N 1AX UK
| |
Collapse
|
18
|
Abstract
DNA topoisomerases are enzymes that control the topology of DNA in all cells. There are two types, I and II, classified according to whether they make transient single- or double-stranded breaks in DNA. Their reactions generally involve the passage of a single- or double-strand segment of DNA through this transient break, stabilized by DNA-protein covalent bonds. All topoisomerases can relax DNA, but DNA gyrase, present in all bacteria, can also introduce supercoils into DNA. Because of their essentiality in all cells and the fact that their reactions proceed via DNA breaks, topoisomerases have become important drug targets; the bacterial enzymes are key targets for antibacterial agents. This article discusses the structure and mechanism of topoisomerases and their roles in the bacterial cell. Targeting of the bacterial topoisomerases by inhibitors, including antibiotics in clinical use, is also discussed.
Collapse
|
19
|
Li C, Zhang WT, Wang XS. CuI-Catalyzed C–N Bond Formation and Cleavage for the Synthesis of Benzimidazo[1,2-a]quinazoline Derivatives. J Org Chem 2014; 79:5847-51. [DOI: 10.1021/jo5007398] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Chao Li
- School of Chemistry and Chemical
Engineering, Jiangsu Key Laboratory of Green Synthesis for Functional
Materials, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P. R. China
| | - Wen-Ting Zhang
- School of Chemistry and Chemical
Engineering, Jiangsu Key Laboratory of Green Synthesis for Functional
Materials, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P. R. China
| | - Xiang-Shan Wang
- School of Chemistry and Chemical
Engineering, Jiangsu Key Laboratory of Green Synthesis for Functional
Materials, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P. R. China
| |
Collapse
|
20
|
Mayer C, Janin YL. Non-quinolone inhibitors of bacterial type IIA topoisomerases: a feat of bioisosterism. Chem Rev 2013; 114:2313-42. [PMID: 24313284 DOI: 10.1021/cr4003984] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Claudine Mayer
- Unité de Microbiologie Structurale, Département de Biologie Structurale et Chimie, Institut Pasteur , 25 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | | |
Collapse
|
21
|
Moreira JB, Mann J, Neidle S, McHugh TD, Taylor PW. Antibacterial activity of head-to-head bis-benzimidazoles. Int J Antimicrob Agents 2013; 42:361-6. [DOI: 10.1016/j.ijantimicag.2013.04.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 04/22/2013] [Accepted: 04/26/2013] [Indexed: 11/30/2022]
|
22
|
Ramsey DM, Amirul Islam M, Turnbull L, Davis RA, Whitchurch CB, McAlpine SR. Psammaplysin F: A unique inhibitor of bacterial chromosomal partitioning. Bioorg Med Chem Lett 2013; 23:4862-6. [DOI: 10.1016/j.bmcl.2013.06.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 06/17/2013] [Accepted: 06/27/2013] [Indexed: 12/29/2022]
|
23
|
Kim SO, Sakchaisri K, N. R. T, Soung NK, Jang JH, Kim YS, Lee KS, Kwon YT, Asami Y, Ahn JS, Erikson RL, Kim BY. STK295900, a dual inhibitor of topoisomerase 1 and 2, induces G(2) arrest in the absence of DNA damage. PLoS One 2013; 8:e53908. [PMID: 23349762 PMCID: PMC3551932 DOI: 10.1371/journal.pone.0053908] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 12/04/2012] [Indexed: 11/18/2022] Open
Abstract
STK295900, a small synthetic molecule belonging to a class of symmetric bibenzimidazoles, exhibits antiproliferative activity against various human cancer cell lines from different origins. Examining the effect of STK295900 in HeLa cells indicates that it induces G(2) phase arrest without invoking DNA damage. Further analysis shows that STK295900 inhibits DNA relaxation that is mediated by topoisomerase 1 (Top 1) and topoisomerase 2 (Top 2) in vitro. In addition, STK295900 also exhibits protective effect against DNA damage induced by camptothecin. However, STK295900 does not affect etoposide-induced DNA damage. Moreover, STK295900 preferentially exerts cytotoxic effect on cancer cell lines while camptothecin, etoposide, and Hoechst 33342 affected both cancer and normal cells. Therefore, STK295900 has a potential to be developed as an anticancer chemotherapeutic agent.
Collapse
Affiliation(s)
- Sun-Ok Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
- Department of Biochemistry, College of Natural Sciences, ChungNam National University, Yuseonggu, Daejeon, Korea
| | - Krisada Sakchaisri
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Thimmegowda N. R.
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Nak Kyun Soung
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Jae-Hyuk Jang
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, ChungNam National University, Yuseonggu, Daejeon, Korea
| | - Kyung Sang Lee
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yong Tae Kwon
- World Class University (WCU), Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, Korea
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, School of Pharmacy, Universigy of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yukihiro Asami
- Chemical Biology Department, RIKEN Advanced Science Institute, Wako-shi, Saitama, Japan
| | - Jong Seog Ahn
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Raymond Leo Erikson
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Bo Yeon Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| |
Collapse
|
24
|
Special Challenges to the Rational Design of Antibacterial Agents. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2013. [DOI: 10.1016/b978-0-12-417150-3.00018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|