1
|
Poddar SM, Chakraborty J, Gayathri P, Srinivasan R. Disruption of salt bridge interactions in the inter-domain cleft of the tubulin-like protein FtsZ of Escherichia coli makes cells sensitive to the cell division inhibitor PC190723. Cytoskeleton (Hoboken) 2024. [PMID: 39230425 DOI: 10.1002/cm.21924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024]
Abstract
FtsZ forms a ring-like assembly at the site of division in bacteria. It is the first protein involved in the formation of the divisome complex to split the cell into two halves, indicating its importance in bacterial cell division. FtsZ is an attractive target for developing new anti-microbial drugs to overcome the challenges of antibiotic resistance. The most potent inhibitor against FtsZ is PC190723, which is effective against all strains and species of Staphylococcus, including the methicillin- and multi-drug-resistant Staphylococcus aureus and strains of Bacillus. However, FtsZs from bacteria such as E. coli, Streptococcus, and Enterococcus were shown to be resistant to this inhibitor. In this study, we provide further evidence that the three pairwise bridging interactions, between residues S227 and G191, R307 and E198 and D299 and R202, between S7, S9, S10 β-strands and the H7 helix occlude the inhibitor from binding to E. coli FtsZ. We generated single, double and triple mutations to disrupt those bridges and tested the effectiveness of PC190723 directly on Z-ring assembly in vivo. Our results show that the disruption of S227-G191 and R307-E198 bridges render EcFtsZ highly sensitive to PC190723 for Z-ring assembly. Ectopic expression of the double mutants, FtsZ S227I R307V results in hypersensitivity of the susceptible E. coli imp4213 strain to PC190723. Our studies could further predict the effectiveness of PC190723 or its derivatives towards FtsZs of other bacterial genera.
Collapse
Affiliation(s)
- Sakshi Mahesh Poddar
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
- Homi Bhabha National Institutes (HBNI), Training School Complex, Mumbai, India
| | | | - Pananghat Gayathri
- Biology, Indian Institute of Science Education and Research, Pune, India
| | - Ramanujam Srinivasan
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
- Homi Bhabha National Institutes (HBNI), Training School Complex, Mumbai, India
| |
Collapse
|
2
|
Bryan EJ, Qiao Q, Wang Y, Roberge JY, LaVoie EJ, Pilch DS. A FtsZ Inhibitor That Can Utilize Siderophore-Ferric Iron Uptake Transporter Systems for Activity against Gram-Negative Bacterial Pathogens. Antibiotics (Basel) 2024; 13:209. [PMID: 38534644 DOI: 10.3390/antibiotics13030209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/28/2024] Open
Abstract
The global threat of multidrug-resistant Gram-negative bacterial pathogens necessitates the development of new and effective antibiotics. FtsZ is an essential and highly conserved cytoskeletal protein that is an appealing antibacterial target for new antimicrobial therapeutics. However, the effectiveness of FtsZ inhibitors against Gram-negative species has been limited due in part to poor intracellular accumulation. To address this limitation, we have designed a FtsZ inhibitor (RUP4) that incorporates a chlorocatechol siderophore functionality that can chelate ferric iron (Fe3+) and utilizes endogenous siderophore uptake pathways to facilitate entry into Gram-negative pathogens. We show that RUP4 is active against both Klebsiella pneumoniae and Acinetobacter baumannii, with this activity being dependent on direct Fe3+ chelation and enhanced under Fe3+-limiting conditions. Genetic deletion studies in K. pneumoniae reveal that RUP4 gains entry through the FepA and CirA outer membrane transporters and the FhuBC inner membrane transporter. We also show that RUP4 exhibits bactericidal synergy against K. pneumoniae when combined with select antibiotics, with the strongest synergy observed with PBP2-targeting β-lactams or MreB inhibitors. In the aggregate, our studies indicate that incorporation of Fe3+-chelating moieties into FtsZ inhibitors is an appealing design strategy for enhancing activity against Gram-negative pathogens of global clinical significance.
Collapse
Affiliation(s)
- Eric J Bryan
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Qi Qiao
- Department of Molecular Design and Synthesis, Rutgers University Biomedical Innovation Cores, Piscataway, NJ 08854, USA
| | - Yuxuan Wang
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Jacques Y Roberge
- Department of Molecular Design and Synthesis, Rutgers University Biomedical Innovation Cores, Piscataway, NJ 08854, USA
| | - Edmond J LaVoie
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Daniel S Pilch
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| |
Collapse
|
3
|
Suigo L, Monterroso B, Sobrinos-Sanguino M, Alfonso C, Straniero V, Rivas G, Zorrilla S, Valoti E, Margolin W. Benzodioxane-benzamides as promising inhibitors of Escherichia coli FtsZ. Int J Biol Macromol 2023; 253:126398. [PMID: 37634788 DOI: 10.1016/j.ijbiomac.2023.126398] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/02/2023] [Accepted: 08/16/2023] [Indexed: 08/29/2023]
Abstract
The conserved process of cell division in bacteria has been a long-standing target for antimicrobials, although there are few examples of potent broad-spectrum compounds that inhibit this process. Most currently available compounds acting on division are directed towards the FtsZ protein, a self-assembling GTPase that is a central element of the division machinery in most bacteria. Benzodioxane-benzamides are promising candidates, but poorly explored in Gram-negatives. We have tested a number of these compounds on E. coli FtsZ and found that many of them significantly stabilized the polymers against disassembly and reduced the GTPase activity. Reconstitution in crowded cell-like conditions showed that FtsZ bundles were also susceptible to these compounds, including some compounds that were inactive on protofilaments in dilute conditions. They efficiently killed E. coli cells defective in the AcrAB efflux pump. The activity of the compounds on cell growth and division generally showed a good correlation with their effect in vitro, and our experiments are consistent with FtsZ being the target in vivo. Our results uncover the detrimental effects of benzodioxane-benzamides on permeable E. coli cells via its central division protein, implying that lead compounds may be found within this class for the development of antibiotics against Gram-negative bacteria.
Collapse
Affiliation(s)
- Lorenzo Suigo
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milano, Italy
| | - Begoña Monterroso
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Marta Sobrinos-Sanguino
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Carlos Alfonso
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Valentina Straniero
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milano, Italy
| | - Germán Rivas
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Silvia Zorrilla
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain.
| | - Ermanno Valoti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milano, Italy.
| | - William Margolin
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas, Houston 77030, TX, USA.
| |
Collapse
|
4
|
Sharma AK, Poddar SM, Chakraborty J, Nayak BS, Kalathil S, Mitra N, Gayathri P, Srinivasan R. A mechanism of salt bridge-mediated resistance to FtsZ inhibitor PC190723 revealed by a cell-based screen. Mol Biol Cell 2023; 34:ar16. [PMID: 36652338 PMCID: PMC10011733 DOI: 10.1091/mbc.e22-12-0538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Bacterial cell division proteins, especially the tubulin homologue FtsZ, have emerged as strong targets for developing new antibiotics. Here, we have utilized the fission yeast heterologous expression system to develop a cell-based assay to screen for small molecules that directly and specifically target the bacterial cell division protein FtsZ. The strategy also allows for simultaneous assessment of the toxicity of the drugs to eukaryotic yeast cells. As a proof-of-concept of the utility of this assay, we demonstrate the effect of the inhibitors sanguinarine, berberine, and PC190723 on FtsZ. Though sanguinarine and berberine affect FtsZ polymerization, they exert a toxic effect on the cells. Further, using this assay system, we show that PC190723 affects Helicobacter pylori FtsZ function and gain new insights into the molecular determinants of resistance to PC190723. On the basis of sequence and structural analysis and site-specific mutations, we demonstrate that the presence of salt bridge interactions between the central H7 helix and β-strands S9 and S10 mediates resistance to PC190723 in FtsZ. The single-step in vivo cell-based assay using fission yeast enabled us to dissect the contribution of sequence-specific features of FtsZ and cell permeability effects associated with bacterial cell envelopes. Thus, our assay serves as a potent tool to rapidly identify novel compounds targeting polymeric bacterial cytoskeletal proteins like FtsZ to understand how they alter polymerization dynamics and address resistance determinants in targets.
Collapse
Affiliation(s)
- Ajay Kumar Sharma
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Sakshi Mahesh Poddar
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Joyeeta Chakraborty
- Biology, Indian Institute of Science Education and Research, Pune 411008, India
| | - Bhagyashri Soumya Nayak
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Srilakshmi Kalathil
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Nivedita Mitra
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| | - Pananghat Gayathri
- Biology, Indian Institute of Science Education and Research, Pune 411008, India
| | - Ramanujam Srinivasan
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Centre for Interdisciplinary Sciences, National Institute of Science Education and Research, Bhubaneswar 752050, India.,Homi Bhabha National Institutes, Anushakti Nagar, Mumbai 400094, India
| |
Collapse
|
5
|
Rosado-Lugo JD, Sun Y, Banerjee A, Cao Y, Datta P, Zhang Y, Yuan Y, Parhi AK. Evaluation of 2,6-difluoro-3-(oxazol-2-ylmethoxy)benzamide chemotypes as Gram-negative FtsZ inhibitors. J Antibiot (Tokyo) 2022; 75:385-395. [PMID: 35618784 DOI: 10.1038/s41429-022-00531-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 01/21/2023]
Abstract
FtsZ inhibitors represent a new drug class as no drugs using this mode of action (MOA) have been approved by regulators. 3-alkoxy substituted 2,6-difluorobenzamide scaffold is one of the most studied FtsZ inhibitors among which the most promising anti-MRSA candidate TXA709 is in clinical trial. In this paper, we present the screening and evaluation of a benzamide class that is functionalized at the alkoxy fragment targeting Gram-negative bacteria. The variations in 3-alkoxy substitutions, specifically the hydroxylated alkyl residues to the secondary and stereogenic pseudo-benzylic carbon of their methyleneoxy linker, are particularly active against K. pneumoniae ATCC 10031 in marked contrast to the derivatives related to PC190723, all of which were inactive against Gram-negative bacteria. The two lead molecules TXA6101 and TXY6129 inhibit the polymerization of E. coli FtsZ in a concentration-dependent manner and induce changes in the morphology of E. coli and K. pneumoniae consistent with inhibition of cell division. These classes of compounds, however, were found to be substrates for efflux pumps in Gram-negative bacteria.
Collapse
Affiliation(s)
- Jesus D Rosado-Lugo
- TAXIS Pharmaceuticals, Inc., R&D Department, 9 Deer Park Drive, Suite J-15, Monmouth Junction, NJ, 08852, USA
| | - Yangsheng Sun
- TAXIS Pharmaceuticals, Inc., R&D Department, 9 Deer Park Drive, Suite J-15, Monmouth Junction, NJ, 08852, USA
| | - Anamika Banerjee
- TAXIS Pharmaceuticals, Inc., R&D Department, 9 Deer Park Drive, Suite J-15, Monmouth Junction, NJ, 08852, USA
| | - Yanlu Cao
- TAXIS Pharmaceuticals, Inc., R&D Department, 9 Deer Park Drive, Suite J-15, Monmouth Junction, NJ, 08852, USA
| | - Pratik Datta
- TAXIS Pharmaceuticals, Inc., R&D Department, 9 Deer Park Drive, Suite J-15, Monmouth Junction, NJ, 08852, USA
| | - Yongzheng Zhang
- TAXIS Pharmaceuticals, Inc., R&D Department, 9 Deer Park Drive, Suite J-15, Monmouth Junction, NJ, 08852, USA
| | - Yi Yuan
- TAXIS Pharmaceuticals, Inc., R&D Department, 9 Deer Park Drive, Suite J-15, Monmouth Junction, NJ, 08852, USA
| | - Ajit K Parhi
- TAXIS Pharmaceuticals, Inc., R&D Department, 9 Deer Park Drive, Suite J-15, Monmouth Junction, NJ, 08852, USA.
| |
Collapse
|
6
|
Zhong DX, She MT, Guo XC, Zheng BX, Huang XH, Zhang YH, Ser HL, Wong WL, Sun N, Lu YJ. Design and synthesis of quinolinium-based derivatives targeting FtsZ for antibacterial evaluation and mechanistic study. Eur J Med Chem 2022; 236:114360. [PMID: 35421657 DOI: 10.1016/j.ejmech.2022.114360] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 01/03/2023]
Abstract
The discovery of small molecular inhibitors targeting essential and conserved bacterial drug targets such as FtsZ protein is a promising approach to fight against multi-drug resistant bacteria. In the present study, two new series of FtsZ inhibitors based on a 1-methylquinolinium scaffold were synthesized. The inhibitors possess a variety of substituent groups including the cyclic or linear amine skeleton at the 2- and 4-position of the quinolinium ring for structure-activity relationship study. In general, the inhibitors bearing a cyclic amine substituent at the 4-position of the quinolinium ring showed better antibacterial activity (MIC down to 0.25 μg/mL) than that at the 2-position, especially against Gram-positive bacteria. Among the twenty FtsZ inhibitors examined in various assays, A3 was identified to exhibit excellent antibacterial activity against S. aureus (MIC = 0.5-1 μg/mL), S. epidermidis (MIC = 0.25 μg/mL) and E. faecium (MIC = 1-8 μg/mL). More importantly, A3 showed low hemolytic toxicity (IC5 = 64 μg/mL) and was found not readily to induce drug resistance. A3 at 2-8 μg/mL promoted the polymerization of FtsZ and interrupted the bacterial division. Furthermore, the ligand-FtsZ interaction study conducted with circular dichroism and molecular docking revealed that A3 induced secondary structure changes of FtsZ protein upon binding to the interdomain cleft of the protein. A3 is thus a potent inhibitor of FtsZ and shows potential to be used as a new antibacterial agent against drug-resistant bacteria.
Collapse
Affiliation(s)
- Dong-Xiao Zhong
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Meng-Ting She
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Xiao-Chun Guo
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Bo-Xin Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Xuan-He Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Yi-Han Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Hooi-Leng Ser
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Wing-Leung Wong
- The State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China.
| | - Ning Sun
- The State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China; Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, PR China.
| | - Yu-Jing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China; Engineering Research Academy of High Value Utilization of Green Plants, Meizhou, 514021, PR China; Golden Health (Guangdong) Biotechnology Co., Ltd, Foshan, 28225, PR China.
| |
Collapse
|
7
|
Pradhan P, Margolin W, Beuria TK. Targeting the Achilles Heel of FtsZ: The Interdomain Cleft. Front Microbiol 2021; 12:732796. [PMID: 34566937 PMCID: PMC8456036 DOI: 10.3389/fmicb.2021.732796] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/16/2021] [Indexed: 02/03/2023] Open
Abstract
Widespread antimicrobial resistance among bacterial pathogens is a serious threat to public health. Thus, identification of new targets and development of new antibacterial agents are urgently needed. Although cell division is a major driver of bacterial colonization and pathogenesis, its targeting with antibacterial compounds is still in its infancy. FtsZ, a bacterial cytoskeletal homolog of eukaryotic tubulin, plays a highly conserved and foundational role in cell division and has been the primary focus of research on small molecule cell division inhibitors. FtsZ contains two drug-binding pockets: the GTP binding site situated at the interface between polymeric subunits, and the inter-domain cleft (IDC), located between the N-terminal and C-terminal segments of the core globular domain of FtsZ. The majority of anti-FtsZ molecules bind to the IDC. Compounds that bind instead to the GTP binding site are much less useful as potential antimicrobial therapeutics because they are often cytotoxic to mammalian cells, due to the high sequence similarity between the GTP binding sites of FtsZ and tubulin. Fortunately, the IDC has much less sequence and structural similarity with tubulin, making it a better potential target for drugs that are less toxic to humans. Over the last decade, a large number of natural and synthetic IDC inhibitors have been identified. Here we outline the molecular structure of IDC in detail and discuss how it has become a crucial target for broad spectrum and species-specific antibacterial agents. We also outline the drugs that bind to the IDC and their modes of action.
Collapse
Affiliation(s)
- Pinkilata Pradhan
- Institute of Life Sciences, Nalco Square, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - William Margolin
- Department of Microbiology and Molecular Genetics, McGovern Medical School, Houston, TX, United States
| | | |
Collapse
|
8
|
Chen CC, Zhang YQ, Zhong DX, Huang XH, Zhang YH, Jiang WH, Li M, Chen Q, Wong WL, Lu YJ. The study of 9,10-dihydroacridine derivatives as a new and effective molecular scaffold for antibacterial agent development. Biochem Biophys Res Commun 2021; 546:40-45. [PMID: 33561747 DOI: 10.1016/j.bbrc.2021.01.096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/28/2021] [Indexed: 10/22/2022]
Abstract
The emergence of worldwide spreading drug-resistant bacteria has been a serious threat to public health during the past decades. The development of new and effective antibacterial agents to address this critical issue is an urgent action. In the present study, we investigated the antibacterial activity of two 9,10-dihydroacridine derivatives and their mechanism. Both compounds were found possessing strong antibacterial activity against some selected Gram-positive bacteria including MRSA, VISA and VRE. The biological study suggests that the compounds promoted FtsZ polymerization and also disrupted Z-ring formation at the dividing site and consequently, the bacterial cell division is interrupted and causing cell death.
Collapse
Affiliation(s)
- Cui-Cui Chen
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Yi-Qi Zhang
- College of Marine Science, Shandong University, Weihai, 264209, PR China
| | - Dong-Xiao Zhong
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Xuan-He Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Yi-Han Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Wen-Hao Jiang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Ming Li
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Qian Chen
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, 510006, PR China.
| | - Wing-Leung Wong
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China.
| | - Yu-Jing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
9
|
Chai WC, Whittall JJ, Song D, Polyak SW, Ogunniyi AD, Wang Y, Bi F, Ma S, Semple SJ, Venter H. Antimicrobial Action and Reversal of Resistance in MRSA by Difluorobenzamide Derivatives Targeted at FtsZ. Antibiotics (Basel) 2020; 9:E873. [PMID: 33291418 PMCID: PMC7762090 DOI: 10.3390/antibiotics9120873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 01/21/2023] Open
Abstract
The bacterial cell division protein, FtsZ, has been identified as a target for antimicrobial development. Derivatives of 3-methoxybenzamide have shown promising activities as FtsZ inhibitors in Gram-positive bacteria. We sought to characterise the activity of five difluorobenzamide derivatives with non-heterocyclic substituents attached through the 3-oxygen. These compounds exhibited antimicrobial activity against methicillin resistant Staphylococcus aureus (MRSA), with an isopentyloxy-substituted compound showing modest activity against vancomycin resistant Enterococcus faecium (VRE). The compounds were able to reverse resistance to oxacillin in highly resistant clinical MRSA strains at concentrations far below their MICs. Three of the compounds inhibited an Escherichia coli strain lacking the AcrAB components of a drug efflux pump, which suggests the lack of Gram-negative activity can partly be attributed to efflux. The compounds inhibited cell division by targeting S. aureus FtsZ, producing a dose-dependent increase in GTPase rate which increased the rate of FtsZ polymerization and stabilized the FtsZ polymers. These compounds did not affect the polymerization of mammalian tubulin and did not display haemolytic activity or cytotoxicity. These derivatives are therefore promising compounds for further development as antimicrobial agents or as resistance breakers to re-sensitive MRSA to beta-lactam antibiotics.
Collapse
Affiliation(s)
- Wern Chern Chai
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, SA 5000 Adelaide, Australia; (W.C.C.); (J.J.W.); (S.W.P.); (S.J.S.)
| | - Jonathan J. Whittall
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, SA 5000 Adelaide, Australia; (W.C.C.); (J.J.W.); (S.W.P.); (S.J.S.)
| | - Di Song
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (D.S.); (Y.W.); (F.B.); (S.M.)
| | - Steven W. Polyak
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, SA 5000 Adelaide, Australia; (W.C.C.); (J.J.W.); (S.W.P.); (S.J.S.)
| | - Abiodun D. Ogunniyi
- Australia Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy Campus, SA 5371 Roseworthy, Australia;
| | - Yinhu Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (D.S.); (Y.W.); (F.B.); (S.M.)
- School of Pharmacy, Liaocheng University, Liaocheng 252000, China
| | - Fangchao Bi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (D.S.); (Y.W.); (F.B.); (S.M.)
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (D.S.); (Y.W.); (F.B.); (S.M.)
| | - Susan J. Semple
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, SA 5000 Adelaide, Australia; (W.C.C.); (J.J.W.); (S.W.P.); (S.J.S.)
- Quality Use of Medicines and Pharmacy Research Centre, Clinical and Health Sciences, University of South Australia, SA 5000 Adelaide, Australia
| | - Henrietta Venter
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, SA 5000 Adelaide, Australia; (W.C.C.); (J.J.W.); (S.W.P.); (S.J.S.)
| |
Collapse
|
10
|
Khare S, Hsin J, Sorto NA, Nepomuceno GM, Shaw JT, Shi H, Huang KC. FtsZ-Independent Mechanism of Division Inhibition by the Small Molecule PC190723 in Escherichia coli. ACTA ACUST UNITED AC 2020; 3:e1900021. [PMID: 32648693 DOI: 10.1002/adbi.201900021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/07/2019] [Indexed: 11/12/2022]
Abstract
While cell division is a critical process in cellular proliferation, very few antibiotics have been identified that target the bacterial cell-division machinery. Recent studies have shown that the small molecule PC190723 inhibits cell division in several Gram-positive bacteria, with a hypothesized mechanism of action involving direct targeting of the tubulin homolog FtsZ, which is essential for division in virtually all bacterial species. Here, it is shown that PC190723 also inhibits cell division in the Gram-negative bacterium Escherichia coli if the outer membrane permeability barrier is compromised genetically or chemically. The results show that the equivalent FtsZ mutations conferring PC190723 resistance in Staphylococcus aureus do not protect E. coli against PC190723, and that suppressors of PC190723 sensitivity in E. coli, which do not generically decrease outer membrane permeability, do not map to FtsZ or other division proteins. These suppressors display a wide range of morphological and growth phenotypes, and one exhibits a death phenotype in the stationary phase similar to that of a mutant with disrupted lipid homeostasis. Finally, a complementing FtsZ-msfGFP fusion is used to show that PC190723 does not affect the Z-ring structure. Taken together, the findings suggest that PC190723 inhibits growth and division in E. coli without targeting FtsZ. This study highlights the importance of utilizing a combination of genetic, chemical, and single-cell approaches to dissect the mechanisms of action of new antibiotics, which are not necessarily conserved across bacterial species.
Collapse
Affiliation(s)
- Somya Khare
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Jen Hsin
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Nohemy A Sorto
- Department of Chemistry, University of California at Davis, Davis, CA, 95616, USA
| | | | - Jared T Shaw
- Department of Chemistry, University of California at Davis, Davis, CA, 95616, USA
| | - Handuo Shi
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA.,Department of Microbiology and Immunology, Stanford University, Stanford, CA, 94305, USA.,Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
| |
Collapse
|
11
|
Straniero V, Suigo L, Casiraghi A, Sebastián-Pérez V, Hrast M, Zanotto C, Zdovc I, De Giuli Morghen C, Radaelli A, Valoti E. Benzamide Derivatives Targeting the Cell Division Protein FtsZ: Modifications of the Linker and the Benzodioxane Scaffold and Their Effects on Antimicrobial Activity. Antibiotics (Basel) 2020; 9:antibiotics9040160. [PMID: 32260339 PMCID: PMC7235863 DOI: 10.3390/antibiotics9040160] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 01/21/2023] Open
Abstract
Filamentous temperature-sensitive Z (FtsZ) is a prokaryotic protein with an essential role in the bacterial cell division process. It is widely conserved and expressed in both Gram-positive and Gram-negative strains. In the last decade, several research groups have pointed out molecules able to target FtsZ in Staphylococcus aureus, Bacillus subtilis and other Gram-positive strains, with sub-micromolar Minimum Inhibitory Concentrations (MICs). Conversely, no promising derivatives active on Gram-negatives have been found up to now. Here, we report our results on a class of benzamide compounds, which showed comparable inhibitory activities on both S. aureus and Escherichia coli FtsZ, even though they proved to be substrates of E. coli efflux pump AcrAB, thus affecting the antimicrobial activity. These surprising results confirmed how a single molecule can target both species while maintaining potent antimicrobial activity. A further computational study helped us decipher the structural features necessary for broad spectrum activity and assess the drug-like profile and the on-target activity of this family of compounds.
Collapse
Affiliation(s)
- Valentina Straniero
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milano, Italy; (L.S.); (A.C.)
- Correspondence: (V.S.); (E.V.); Tel.: +39-0250319361 (V.S.); +39-0250319334 (E.V.)
| | - Lorenzo Suigo
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milano, Italy; (L.S.); (A.C.)
| | - Andrea Casiraghi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milano, Italy; (L.S.); (A.C.)
| | - Victor Sebastián-Pérez
- Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain;
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| | - Martina Hrast
- Pharmacy Faculty, University of Ljubljana, Aškerčeva cesta, 7, 1000 Ljubljana, Slovenia;
| | - Carlo Zanotto
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Vanvitelli, 32, 20129 Milano, Italy; (C.Z.); (A.R.)
| | - Irena Zdovc
- Veterinary Faculty, University of Ljubljana, Gerbičeva, 60, 1000 Ljubljana, Slovenia;
| | - Carlo De Giuli Morghen
- Department of Chemical – Pharmaceutical and Biomolecular Technologies, Catholic University “Our Lady of Good Counsel”, Rr. Dritan Hoxha, 1025 Tirana, Albania;
| | - Antonia Radaelli
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Vanvitelli, 32, 20129 Milano, Italy; (C.Z.); (A.R.)
| | - Ermanno Valoti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Luigi Mangiagalli, 25, 20133 Milano, Italy; (L.S.); (A.C.)
- Correspondence: (V.S.); (E.V.); Tel.: +39-0250319361 (V.S.); +39-0250319334 (E.V.)
| |
Collapse
|
12
|
Casiraghi A, Suigo L, Valoti E, Straniero V. Targeting Bacterial Cell Division: A Binding Site-Centered Approach to the Most Promising Inhibitors of the Essential Protein FtsZ. Antibiotics (Basel) 2020; 9:E69. [PMID: 32046082 PMCID: PMC7167804 DOI: 10.3390/antibiotics9020069] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 11/16/2022] Open
Abstract
Binary fission is the most common mode of bacterial cell division and is mediated by a multiprotein complex denominated the divisome. The constriction of the Z-ring splits the mother bacterial cell into two daughter cells of the same size. The Z-ring is formed by the polymerization of FtsZ, a bacterial protein homologue of eukaryotic tubulin, and it represents the first step of bacterial cytokinesis. The high grade of conservation of FtsZ in most prokaryotic organisms and its relevance in orchestrating the whole division system make this protein a fascinating target in antibiotic research. Indeed, FtsZ inhibition results in the complete blockage of the division system and, consequently, in a bacteriostatic or a bactericidal effect. Since many papers and reviews already discussed the physiology of FtsZ and its auxiliary proteins, as well as the molecular mechanisms in which they are involved, here, we focus on the discussion of the most compelling FtsZ inhibitors, classified by their main protein binding sites and following a medicinal chemistry approach.
Collapse
Affiliation(s)
| | | | | | - Valentina Straniero
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via Luigi Mangiagalli, 25, 20133 Milano, Italy; (A.C.); (L.S.); (E.V.)
| |
Collapse
|
13
|
Li Y, Sun N, Ser HL, Long W, Li Y, Chen C, Zheng B, Huang X, Liu Z, Lu YJ. Antibacterial activity evaluation and mode of action study of novel thiazole-quinolinium derivatives. RSC Adv 2020; 10:15000-15014. [PMID: 35497125 PMCID: PMC9052103 DOI: 10.1039/d0ra00691b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
Abstract
A new series of thiazole-quinolinium derivatives perturb the polymerization of FtsZ with strong antibacterial activities.
Collapse
Affiliation(s)
- Ying Li
- School of Biomedical and Pharmaceutical Sciences
- Guangdong University of Technology
- Guangzhou 510006
- PR China
| | - Ning Sun
- School of Biomedical and Pharmaceutical Sciences
- Guangdong University of Technology
- Guangzhou 510006
- PR China
- The State Key Laboratory of Chemical Biology and Drug Discovery
| | - Hooi-Leng Ser
- School of Biomedical and Pharmaceutical Sciences
- Guangdong University of Technology
- Guangzhou 510006
- PR China
- Novel Bacteria and Drug Discovery (NBDD) Research Group
| | - Wei Long
- School of Biomedical and Pharmaceutical Sciences
- Guangdong University of Technology
- Guangzhou 510006
- PR China
| | - Yanan Li
- Department of Pharmacy
- The Fifth Affiliated Hospital of Sun Yat-sen University
- Zhuhai
- P. R. China
| | - Cuicui Chen
- School of Biomedical and Pharmaceutical Sciences
- Guangdong University of Technology
- Guangzhou 510006
- PR China
| | - Boxin Zheng
- School of Biomedical and Pharmaceutical Sciences
- Guangdong University of Technology
- Guangzhou 510006
- PR China
| | - Xuanhe Huang
- School of Biomedical and Pharmaceutical Sciences
- Guangdong University of Technology
- Guangzhou 510006
- PR China
| | - Zhihua Liu
- The State Key Laboratory of Chemical Biology and Drug Discovery
- Department of Applied Biology and Chemical Technology
- The Hong Kong Polytechnic University
- Kowloon
- China
| | - Yu-Jing Lu
- School of Biomedical and Pharmaceutical Sciences
- Guangdong University of Technology
- Guangzhou 510006
- PR China
| |
Collapse
|
14
|
Straniero V, Sebastián-Pérez V, Hrast M, Zanotto C, Casiraghi A, Suigo L, Zdovc I, Radaelli A, De Giuli Morghen C, Valoti E. Benzodioxane-Benzamides as Antibacterial Agents: Computational and SAR Studies to Evaluate the Influence of the 7-Substitution in FtsZ Interaction. ChemMedChem 2019; 15:195-209. [PMID: 31750973 DOI: 10.1002/cmdc.201900537] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/08/2019] [Indexed: 01/24/2023]
Abstract
FtsZ is a crucial prokaryotic protein involved in bacterial cell replication. It recently arose as a promising target in the search for antimicrobial agents able to fight antimicrobial resistance. In this work, going on with our structure-activity relationship (SAR) study, we developed variously 7-substituted 1,4-benzodioxane compounds, linked to the 2,6-difluorobenzamide by a methylenoxy bridge. Compounds exhibit promising antibacterial activities not only against multidrug-resistant Staphylococcus aureus, but also on mutated Escherichia coli strains, thus enlarging their spectrum of action toward Gram-negative bacteria as well. Computational studies elucidated, through a validated FtsZ binding protocol, the structural features of new promising derivatives as FtsZ inhibitors.
Collapse
Affiliation(s)
- Valentina Straniero
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Luigi Mangiagalli 25, 20133, Milano, Italy
| | | | - Martina Hrast
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Carlo Zanotto
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Via Vanvitelli 32, 20129, Milano, Italy
| | - Andrea Casiraghi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Luigi Mangiagalli 25, 20133, Milano, Italy
| | - Lorenzo Suigo
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Luigi Mangiagalli 25, 20133, Milano, Italy
| | - Irena Zdovc
- Faculty of Veterinary Medicine, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Antonia Radaelli
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Via Vanvitelli 32, 20129, Milano, Italy
| | | | - Ermanno Valoti
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via Luigi Mangiagalli 25, 20133, Milano, Italy
| |
Collapse
|
15
|
Carro L. Recent Progress in the Development of Small-Molecule FtsZ Inhibitors as Chemical Tools for the Development of Novel Antibiotics. Antibiotics (Basel) 2019; 8:E217. [PMID: 31717975 PMCID: PMC6963470 DOI: 10.3390/antibiotics8040217] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/26/2019] [Accepted: 11/05/2019] [Indexed: 01/20/2023] Open
Abstract
Antibiotics are potent pharmacological weapons against bacterial pathogens, nevertheless their efficacy is becoming compromised due to the worldwide emergence and spread of multidrug-resistant bacteria or "superbugs". Antibiotic resistance is rising to such dangerous levels that the treatment of bacterial infections is becoming a clinical challenge. Therefore, urgent action is needed to develop new generations of antibiotics that will help tackle this increasing and serious public health problem. Due to its essential role in bacterial cell division, the tubulin-like protein FtsZ has emerged as a promising target for the development of novel antibiotics with new mechanisms of action. This review highlights the medicinal chemistry efforts towards the identification of small-molecule FtsZ inhibitors with antibacterial activity in the last three years.
Collapse
Affiliation(s)
- Laura Carro
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain;
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| |
Collapse
|
16
|
Kusuma KD, Payne M, Ung AT, Bottomley AL, Harry EJ. FtsZ as an Antibacterial Target: Status and Guidelines for Progressing This Avenue. ACS Infect Dis 2019; 5:1279-1294. [PMID: 31268666 DOI: 10.1021/acsinfecdis.9b00055] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The disturbing increase in the number of bacterial pathogens that are resistant to multiple, or sometimes all, current antibiotics highlights the desperate need to pursue the discovery and development of novel classes of antibacterials. The wealth of knowledge available about the bacterial cell division machinery has aided target-driven approaches to identify new inhibitor compounds. The main division target being pursued is the highly conserved and essential protein FtsZ. Despite very active research on FtsZ inhibitors for several years, this protein is not yet targeted by any commercial antibiotic. Here, we discuss the suitability of FtsZ as an antibacterial target for drug development and review progress achieved in this area. We use hindsight to highlight the gaps that have slowed progress in FtsZ inhibitor development and to suggest guidelines for concluding that FtsZ is actually the target of these molecules, a key missing link in several studies. In moving forward, a multidisciplinary, communicative, and collaborative process, with sharing of research expertise, is critical if we are to succeed.
Collapse
|
17
|
Straniero V, Zanotto C, Straniero L, Casiraghi A, Duga S, Radaelli A, De Giuli Morghen C, Valoti E. 2,6-Difluorobenzamide Inhibitors of Bacterial Cell Division Protein FtsZ: Design, Synthesis, and Structure-Activity Relationships. ChemMedChem 2017; 12:1303-1318. [PMID: 28586174 DOI: 10.1002/cmdc.201700201] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/30/2017] [Indexed: 11/08/2022]
Abstract
A wide variety of drug-resistant microorganisms are continuously emerging, restricting the therapeutic options for common bacterial infections. Antimicrobial agents that were originally potent are now no longer helpful, due to their weak or null activity toward these antibiotic-resistant bacteria. In addition, none of the recently approved antibiotics affect innovative targets, resulting in a need for novel drugs with innovative antibacterial mechanisms of action. The essential cell division protein filamentous temperature-sensitive Z (FtsZ) has emerged as a possible target, thanks to its ubiquitous expression and its homology to eukaryotic β-tubulin. In the latest years, several compounds were shown to interact with this prokaryotic protein and selectively inhibit bacterial cell division. Recently, our research group developed interesting derivatives displaying good antibacterial activities against methicillin-resistant Staphylococcus aureus, as well as vancomycin-resistant Enterococcus faecalis and Mycobacterium tuberculosis. The aim of the present study was to summarize the structure-activity relationships of differently substituted heterocycles, linked by a methylenoxy bridge to the 2,6-difluorobenzamide, and to validate FtsZ as the real target of this class of antimicrobials.
Collapse
Affiliation(s)
- Valentina Straniero
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, via Mangiagalli 25, 20133, Milano, Italy
| | - Carlo Zanotto
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, via Vanvitelli 32, 20129, Milano, Italy
| | - Letizia Straniero
- Department of Biomedical Sciences, Humanitas University, via Manzoni 113, 2, 0089, Rozzano-Milano, Italy
| | - Andrea Casiraghi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, via Mangiagalli 25, 20133, Milano, Italy
| | - Stefano Duga
- Department of Biomedical Sciences, Humanitas University, via Manzoni 113, 2, 0089, Rozzano-Milano, Italy.,Humanitas Clinical and Research Center, via Manzoni 56, 20089, Rozzano-Milano, Italy
| | - Antonia Radaelli
- Department of Pharmacological and Biomolecular Science, Università degli Studi di Milano, via Balzaretti 9, 20133, Milano, Italy.,Cellular and Molecular Pharmacology Section, National Research Council (CNR), Institute of Neurosciences, Università degli Studi di Milano, via Vanvitelli 32, 20129, Milano, Italy
| | | | - Ermanno Valoti
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, via Mangiagalli 25, 20133, Milano, Italy
| |
Collapse
|
18
|
Sun N, Lu YJ, Chan FY, Du RL, Zheng YY, Zhang K, So LY, Abagyan R, Zhuo C, Leung YC, Wong KY. A Thiazole Orange Derivative Targeting the Bacterial Protein FtsZ Shows Potent Antibacterial Activity. Front Microbiol 2017; 8:855. [PMID: 28553278 PMCID: PMC5426085 DOI: 10.3389/fmicb.2017.00855] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/27/2017] [Indexed: 01/02/2023] Open
Abstract
The prevalence of multidrug resistance among clinically significant bacteria calls for the urgent development of new antibiotics with novel mechanisms of action. In this study, a new small molecule exhibiting excellent inhibition of bacterial cell division with potent antibacterial activity was discovered through cell-based screening. The compound exhibits a broad spectrum of bactericidal activity, including the methicillin-resistant Staphylococcus aureus, vancomycin-resistant Enterococcus and NDM-1 Escherichia coli. The in vitro and in vivo results suggested that this compound disrupts the dynamic assembly of FtsZ protein and Z-ring formation through stimulating FtsZ polymerization. Moreover, this compound exhibits no activity on mammalian tubulin polymerization and shows low cytotoxicity on mammalian cells. Taken together, these findings could provide a new chemotype for development of antibacterials with FtsZ as the target.
Collapse
Affiliation(s)
- Ning Sun
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chirosciences, The Hong Kong Polytechnic UniversityHong Kong, Hong Kong
| | - Yu-Jing Lu
- Institute of Natural Medicine and Green Chemistry, School of Chemical Engineering and Light Industry, Guangdong University of TechnologyGuangzhou, China
| | - Fung-Yi Chan
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chirosciences, The Hong Kong Polytechnic UniversityHong Kong, Hong Kong
| | - Ruo-Lan Du
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chirosciences, The Hong Kong Polytechnic UniversityHong Kong, Hong Kong
| | - Yuan-Yuan Zheng
- Institute of Natural Medicine and Green Chemistry, School of Chemical Engineering and Light Industry, Guangdong University of TechnologyGuangzhou, China
| | - Kun Zhang
- Institute of Natural Medicine and Green Chemistry, School of Chemical Engineering and Light Industry, Guangdong University of TechnologyGuangzhou, China
| | - Lok-Yan So
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chirosciences, The Hong Kong Polytechnic UniversityHong Kong, Hong Kong
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La JollaCA, USA
| | - Chao Zhuo
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Yun-Chung Leung
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chirosciences, The Hong Kong Polytechnic UniversityHong Kong, Hong Kong
| | - Kwok-Yin Wong
- Department of Applied Biology and Chemical Technology and the State Key Laboratory of Chirosciences, The Hong Kong Polytechnic UniversityHong Kong, Hong Kong
| |
Collapse
|
19
|
Hu Z, Zhang S, Zhou W, Ma X, Xiang G. Synthesis and antibacterial activity of 3-benzylamide derivatives as FtsZ inhibitors. Bioorg Med Chem Lett 2017; 27:1854-1858. [DOI: 10.1016/j.bmcl.2017.02.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 02/05/2017] [Accepted: 02/15/2017] [Indexed: 11/26/2022]
|
20
|
Araújo-Bazán L, Ruiz-Avila LB, Andreu D, Huecas S, Andreu JM. Cytological Profile of Antibacterial FtsZ Inhibitors and Synthetic Peptide MciZ. Front Microbiol 2016; 7:1558. [PMID: 27752253 PMCID: PMC5045927 DOI: 10.3389/fmicb.2016.01558] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/16/2016] [Indexed: 11/26/2022] Open
Abstract
Cell division protein FtsZ is the organizer of the cytokinetic ring in almost all bacteria and a target for the discovery of new antibacterial agents that are needed to counter widespread antibiotic resistance. Bacterial cytological profiling, using quantitative microscopy, is a powerful approach for identifying the mechanism of action of antibacterial molecules affecting different cellular pathways. We have determined the cytological profile on Bacillus subtilis cells of a selection of small molecule inhibitors targeting FtsZ on different binding sites. FtsZ inhibitors lead to long undivided cells, impair the normal assembly of FtsZ into the midcell Z-rings, induce aberrant ring distributions, punctate FtsZ foci, membrane spots and also modify nucleoid length. Quantitative analysis of cell and nucleoid length combined, or the Z-ring distribution, allows categorizing FtsZ inhibitors and to distinguish them from antibiotics with other mechanisms of action, which should be useful for identifying new antibacterial FtsZ inhibitors. Biochemical assays of FtsZ polymerization and GTPase activity combined explain the cellular effects of the FtsZ polymer stabilizing agent PC190723 and its fragments. MciZ is a 40-aminoacid endogenous inhibitor of cell division normally expressed during sporulation in B. subtilis. Using FtsZ cytological profiling we have determined that exogenous synthetic MciZ is an effective inhibitor of B. subtilis cell division, Z-ring formation and localization. This finding supports our cell-based approach to screen for FtsZ inhibitors and opens new possibilities for peptide inhibitors of bacterial cell division.
Collapse
Affiliation(s)
- Lidia Araújo-Bazán
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas Madrid, Spain
| | - Laura B Ruiz-Avila
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas Madrid, Spain
| | - David Andreu
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra Barcelona, Spain
| | - Sonia Huecas
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas Madrid, Spain
| | - José M Andreu
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas Madrid, Spain
| |
Collapse
|
21
|
Abstract
Filamenting temperature-sensitive mutant Z (FtsZ), an essential cell division protein in bacteria, has recently emerged as an important and exploitable antibacterial target. Cytokinesis in bacteria is regulated by the assembly dynamics of this protein, which is ubiquitously present in eubacteria. The perturbation of FtsZ assembly has been found to have a deleterious effect on the cytokinetic machinery and, in turn, upon cell survival. FtsZ is highly conserved among prokaryotes, offering the possibility of broad-spectrum antibacterial agents, while its limited sequence homology with tubulin (an essential protein in eukaryotic mitosis) offers the possibility of selective toxicity. This review aims to summarize current knowledge regarding the mechanism of action of FtsZ, and to highlight existing attempts toward the development of clinically useful inhibitors.
Collapse
|
22
|
Mikuni S, Kodama K, Sasaki A, Kohira N, Maki H, Munetomo M, Maenaka K, Kinjo M. Screening for FtsZ Dimerization Inhibitors Using Fluorescence Cross-Correlation Spectroscopy and Surface Resonance Plasmon Analysis. PLoS One 2015; 10:e0130933. [PMID: 26154290 PMCID: PMC4496089 DOI: 10.1371/journal.pone.0130933] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/01/2015] [Indexed: 01/16/2023] Open
Abstract
FtsZ is an attractive target for antibiotic research because it is an essential bacterial cell division protein that polymerizes in a GTP-dependent manner. To find the seed chemical structure, we established a high-throughput, quantitative screening method combining fluorescence cross-correlation spectroscopy (FCCS) and surface plasmon resonance (SPR). As a new concept for the application of FCCS to polymerization-prone protein, Staphylococcus aureus FtsZ was fragmented into the N-terminal and C-terminal, which were fused with GFP and mCherry (red fluorescent protein), respectively. By this fragmentation, the GTP-dependent head-to-tail dimerization of each fluorescent labeled fragment of FtsZ could be observed, and the inhibitory processes of chemicals could be monitored by FCCS. In the first round of screening by FCCS, 28 candidates were quantitatively and statistically selected from 495 chemicals determined by in silico screening. Subsequently, in the second round of screening by FCCS, 71 candidates were also chosen from 888 chemicals selected via an in silico structural similarity search of the chemicals screened in the first round of screening. Moreover, the dissociation constants between the highest inhibitory chemicals and Staphylococcus aureus FtsZ were determined by SPR. Finally, by measuring the minimum inhibitory concentration, it was confirmed that the screened chemical had antibacterial activity against Staphylococcus aureus, including methicillin-resistant Staphylococcus aureus (MRSA).
Collapse
Affiliation(s)
- Shintaro Mikuni
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Kota Kodama
- Creative Research Institution, Hokkaido University, Sapporo, Japan
| | - Akira Sasaki
- Bio-Analytical Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Ibaraki, Japan
| | - Naoki Kohira
- Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Hideki Maki
- Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Masaharu Munetomo
- Information Initiative Center and Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical Science, Hokkaido University, Sapporo, Japan
| | - Masataka Kinjo
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
- * E-mail:
| |
Collapse
|
23
|
Li XZ, Plésiat P, Nikaido H. The challenge of efflux-mediated antibiotic resistance in Gram-negative bacteria. Clin Microbiol Rev 2015; 28:337-418. [PMID: 25788514 PMCID: PMC4402952 DOI: 10.1128/cmr.00117-14] [Citation(s) in RCA: 946] [Impact Index Per Article: 105.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The global emergence of multidrug-resistant Gram-negative bacteria is a growing threat to antibiotic therapy. The chromosomally encoded drug efflux mechanisms that are ubiquitous in these bacteria greatly contribute to antibiotic resistance and present a major challenge for antibiotic development. Multidrug pumps, particularly those represented by the clinically relevant AcrAB-TolC and Mex pumps of the resistance-nodulation-division (RND) superfamily, not only mediate intrinsic and acquired multidrug resistance (MDR) but also are involved in other functions, including the bacterial stress response and pathogenicity. Additionally, efflux pumps interact synergistically with other resistance mechanisms (e.g., with the outer membrane permeability barrier) to increase resistance levels. Since the discovery of RND pumps in the early 1990s, remarkable scientific and technological advances have allowed for an in-depth understanding of the structural and biochemical basis, substrate profiles, molecular regulation, and inhibition of MDR pumps. However, the development of clinically useful efflux pump inhibitors and/or new antibiotics that can bypass pump effects continues to be a challenge. Plasmid-borne efflux pump genes (including those for RND pumps) have increasingly been identified. This article highlights the recent progress obtained for organisms of clinical significance, together with methodological considerations for the characterization of MDR pumps.
Collapse
Affiliation(s)
- Xian-Zhi Li
- Human Safety Division, Veterinary Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Patrick Plésiat
- Laboratoire de Bactériologie, Faculté de Médecine-Pharmacie, Centre Hospitalier Régional Universitaire, Université de Franche-Comté, Besançon, France
| | - Hiroshi Nikaido
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| |
Collapse
|
24
|
Artola M, Ruiz-Avila LB, Vergoñós A, Huecas S, Araujo-Bazán L, Martín-Fontecha M, Vázquez-Villa H, Turrado C, Ramírez-Aportela E, Hoegl A, Nodwell M, Barasoain I, Chacón P, Sieber SA, Andreu JM, López-Rodríguez ML. Effective GTP-replacing FtsZ inhibitors and antibacterial mechanism of action. ACS Chem Biol 2015; 10:834-43. [PMID: 25486266 DOI: 10.1021/cb500974d] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Essential cell division protein FtsZ is considered an attractive target in the search for antibacterials with novel mechanisms of action to overcome the resistance problem. FtsZ undergoes GTP-dependent assembly at midcell to form the Z-ring, a dynamic structure that evolves until final constriction of the cell. Therefore, molecules able to inhibit its activity will eventually disrupt bacterial viability. In this work, we report a new series of small molecules able to replace GTP and to specifically inhibit FtsZ, blocking the bacterial division process. These new synthesized inhibitors interact with the GTP-binding site of FtsZ (Kd = 0.4-0.8 μM), display antibacterial activity against Gram-positive pathogenic bacteria, and show selectivity against tubulin. Biphenyl derivative 28 stands out as a potent FtsZ inhibitor (Kd = 0.5 μM) with high antibacterial activity [MIC (MRSA) = 7 μM]. In-depth analysis of the mechanism of action of compounds 22, 28, 33, and 36 has revealed that they act as effective inhibitors of correct FtsZ assembly, blocking bacterial division and thus leading to filamentous undivided cells. These findings provide a compelling rationale for the development of compounds targeting the GTP-binding site as antibacterial agents and open the door to antibiotics with novel mechanisms of action.
Collapse
Affiliation(s)
- Marta Artola
- Departamento de Química Orgánica
I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | | | - Albert Vergoñós
- Centro de Investigaciones Biológicas, CSIC, E-28040 Madrid, Spain
| | - Sonia Huecas
- Centro de Investigaciones Biológicas, CSIC, E-28040 Madrid, Spain
| | | | - Mar Martín-Fontecha
- Departamento de Química Orgánica
I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Henar Vázquez-Villa
- Departamento de Química Orgánica
I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Carlos Turrado
- Departamento de Química Orgánica
I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Erney Ramírez-Aportela
- Centro de Investigaciones Biológicas, CSIC, E-28040 Madrid, Spain
- Instituto de Química-Física Rocasolano, CSIC, E-28006 Madrid, Spain
| | - Annabelle Hoegl
- Center for Integrated Protein Science Munich, Technische Universität München, Department of Chemistry, D-85747 Garching, Germany
| | - Matthew Nodwell
- Center for Integrated Protein Science Munich, Technische Universität München, Department of Chemistry, D-85747 Garching, Germany
| | - Isabel Barasoain
- Centro de Investigaciones Biológicas, CSIC, E-28040 Madrid, Spain
| | - Pablo Chacón
- Instituto de Química-Física Rocasolano, CSIC, E-28006 Madrid, Spain
| | - Stephan A. Sieber
- Center for Integrated Protein Science Munich, Technische Universität München, Department of Chemistry, D-85747 Garching, Germany
| | - Jose M. Andreu
- Centro de Investigaciones Biológicas, CSIC, E-28040 Madrid, Spain
| | - María L. López-Rodríguez
- Departamento de Química Orgánica
I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| |
Collapse
|
25
|
Chiodini G, Pallavicini M, Zanotto C, Bissa M, Radaelli A, Straniero V, Bolchi C, Fumagalli L, Ruggeri P, De Giuli Morghen C, Valoti E. Benzodioxane-benzamides as new bacterial cell division inhibitors. Eur J Med Chem 2014; 89:252-65. [PMID: 25462242 DOI: 10.1016/j.ejmech.2014.09.100] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 09/09/2014] [Accepted: 09/15/2014] [Indexed: 11/30/2022]
Abstract
A SAR study was performed on 3-substituted 2,6-difluorobenzamides, known inhibitors of the essential bacterial cell division protein FtsZ, through a series of modifications first of 2,6-difluoro-3-nonyloxybenzamide and then of its 3-pyridothiazolylmethoxy analogue PC190723. The study led to the identification of chiral 2,6-difluorobenzamides bearing 1,4-benzodioxane-2-methyl residue at the 3-position as potent antistaphylococcal compounds.
Collapse
Affiliation(s)
- Giuseppe Chiodini
- Dipartimento di Scienze Farmaceutiche, Università di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Marco Pallavicini
- Dipartimento di Scienze Farmaceutiche, Università di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Carlo Zanotto
- Department of Medical Biothechnologies and Translational Medicine, Università di Milano, Via Vanvitelli 32, I-20129 Milano, Italy
| | - Massimiliano Bissa
- Department of Pharmacological and Biomolecular Sciences, Università di Milano, Via Balzaretti 9, I-2013 Milano, Italy
| | - Antonia Radaelli
- Department of Pharmacological and Biomolecular Sciences, Università di Milano, Via Balzaretti 9, I-2013 Milano, Italy; CNR Institute of Neurosciences, Cellular and Molecular Pharmacology Section, Università di Milano, Via Vanvitelli 32, I-20129 Milano, Italy
| | - Valentina Straniero
- Dipartimento di Scienze Farmaceutiche, Università di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Cristiano Bolchi
- Dipartimento di Scienze Farmaceutiche, Università di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Laura Fumagalli
- Dipartimento di Scienze Farmaceutiche, Università di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Paola Ruggeri
- Dipartimento di Scienze Farmaceutiche, Università di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Carlo De Giuli Morghen
- Department of Medical Biothechnologies and Translational Medicine, Università di Milano, Via Vanvitelli 32, I-20129 Milano, Italy; CNR Institute of Neurosciences, Cellular and Molecular Pharmacology Section, Università di Milano, Via Vanvitelli 32, I-20129 Milano, Italy
| | - Ermanno Valoti
- Dipartimento di Scienze Farmaceutiche, Università di Milano, Via Mangiagalli 25, I-20133 Milano, Italy.
| |
Collapse
|