1
|
Hondros AD, Young MM, Jaimes FE, Kinkead J, Thompson RJ, Melander C, Cavanagh J. Two-Component System Sensor Kinase Inhibitors Target the ATP-Lid of PmrB to Disrupt Colistin Resistance in Acinetobacter baumannii. Biochemistry 2025; 64:1317-1327. [PMID: 40056100 DOI: 10.1021/acs.biochem.4c00789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Two-component systems serve as ubiquitous communication modules that enable bacteria to detect and respond to various stimuli by regulating cellular processes such as growth, viability, and, most notably, antimicrobial resistance. Classical two-component systems consist of two proteins: an initial membrane-bound sensor histidine kinase and a DNA-binding response regulator that induces the appropriate response within the cell. Numerous studies have implicated the PmrAB two-component system in facilitating resistance to the last-resort antibiotic polymyxin E (colistin) in Acinetobacter baumannii. As initiators of the signaling pathways that elicit resistance, histidine kinases present ideal targets for developing antibiotic adjuvant drugs. Despite this, due to the membrane-bound nature of the histidine kinase PmrB, in vitro studies on PmrAB have been predominantly limited to the response regulator PmrA. In this work, we counter these limitations by producing a recombinant truncation of the cytosolic portion of PmrB (PmrBc) that retains its ATP binding, autophosphorylation, and phosphotransfer functions. Subsequently, in vivo phosphorylation assays using this protein construct allowed for the evaluation of five compounds (IMD-0354, NDM-265, NDM-455, NDM-463, and NDM-497) that act as PmrBc inhibitors capable of preventing autophosphorylation and phosphotransfer independently. These compounds have been shown to eliminate colistin resistance in vivo. Finally, these results, paired with mass spectrometry and limited proteolysis investigations, enabled us to determine the mechanism of action of these compounds as well as their likely binding site on the ATP-lid of PmrB.
Collapse
Affiliation(s)
- Alexander D Hondros
- Department of Biochemistry & Molecular Biology, Brody School of Medicine East Carolina University, Greenville, North Carolina 27834, United States
| | - Milah M Young
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Felicia E Jaimes
- Department of Biochemistry & Molecular Biology, Brody School of Medicine East Carolina University, Greenville, North Carolina 27834, United States
| | - Jude Kinkead
- Department of Biochemistry & Molecular Biology, Brody School of Medicine East Carolina University, Greenville, North Carolina 27834, United States
| | - Richele J Thompson
- Department of Biochemistry & Molecular Biology, Brody School of Medicine East Carolina University, Greenville, North Carolina 27834, United States
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - John Cavanagh
- Department of Biochemistry & Molecular Biology, Brody School of Medicine East Carolina University, Greenville, North Carolina 27834, United States
| |
Collapse
|
2
|
Aranaga C, Varela R, Falco A, Villa J, Moreno LM, Causse M, Martínez-Martínez L. In Vitro Activity of the Triazinyl Diazepine Compound FTSD2 Against Drug-Resistant Mycobacterium tuberculosis Strains. Pharmaceuticals (Basel) 2025; 18:360. [PMID: 40143137 PMCID: PMC11945624 DOI: 10.3390/ph18030360] [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: 11/22/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Compounds derived from pyrimido-diazepine have shown selective inhibition of the susceptible Mycobacterium tuberculosis strain H37Rv. However, there is a need for studies that evaluate the activity of these compounds against multidrug-resistant strains and clinical isolates. This study aims to evaluate the antitubercular potential of FTSD2 against drug-resistant strains of M. tuberculosis. Methods: The compound 4-(2,4-diamino-8-(4-methoxyphenyl)-8,9-dihydro-7H-pyrimido[4,5-b][1,4]diazepin-6-yl)-N-(2-(4-(dimethylamino)-6-(4-fluorophenyl)amino-1,3,5-triazin-2-yl)amino)ethyl)benzenesulfonamide (FTSD2) was tested against drug-resistant M. tuberculosis strains at minimal inhibitory and bactericidal concentrations (MIC and MBC). Kill curve assays were performed to assess bactericidal activity, and cytotoxicity was evaluated in human monocyte-derived macrophages and the RAW 264.7 murine macrophage cell line. Intracellular death assays, specifically macrophage infection assays, were also conducted to evaluate the effect of FTSD2 on intracellular M. tuberculosis growth. Results: FTSD2 inhibited the growth of drug-resistant M. tuberculosis at MIC and MBC values between 0.5 and 1 mg/L. Kill curve assays demonstrated concentration-dependent bactericidal activity. No cytotoxicity was observed in macrophages at concentrations below 64 mg/L. Additionally, FTSD2 significantly suppressed intracellular M. tuberculosis growth after 192 h. FTSD2 did not inhibit the growth of nontuberculous mycobacteria, including M. avium, M. abscessus, M. fortuitum, M. chelonae, and M. smegmatis at 50 mg/L. Conclusions: FTSD2 exhibits strong potential as a leading compound for the development of new antitubercular drugs, with selective activity against M. tuberculosis and minimal cytotoxic effects on macrophages. Further studies are needed to explore its mechanisms of action and therapeutic potential.
Collapse
Affiliation(s)
- Carlos Aranaga
- Grupo de Investigación en Química y Biotecnología (QUIBIO), Facultad de Ciencias Básicas, Laboratorio de Parasitología y Enfermedades Tropicales, Universidad Santiago de Cali, Santiago de Cali 760035, Colombia;
- Departamento de Química Agrícola, Edafología y Microbiología, Universidad de Córdoba, 14071 Córdoba, Spain;
| | - Ruben Varela
- Grupo de Investigación en Química y Biotecnología (QUIBIO), Facultad de Ciencias Básicas, Laboratorio de Parasitología y Enfermedades Tropicales, Universidad Santiago de Cali, Santiago de Cali 760035, Colombia;
| | - Aura Falco
- Grupo de Investigación en Microbiología, Industria y Medio Ambiente (GIMIA), Facultad de Ciencias Básicas, Universidad Santiago de Cali, Santiago de Cali 760035, Colombia;
| | - Janny Villa
- Grupo de Investigaciones Biomédicas, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín 0500, Colombia;
| | - Leydi M. Moreno
- Grupo de Investigación de Compuestos Heterocíclicos, Departamento de Química, Universidad del Valle, Santiago de Cali 760042, Colombia;
| | - Manuel Causse
- Unidad de Gestión Clínica de Microbiología, Hospital Universitario Reina Sofía, 14004 Córdoba, Spain;
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFECT), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luis Martínez-Martínez
- Departamento de Química Agrícola, Edafología y Microbiología, Universidad de Córdoba, 14071 Córdoba, Spain;
- Unidad de Gestión Clínica de Microbiología, Hospital Universitario Reina Sofía, 14004 Córdoba, Spain;
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFECT), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
3
|
Ji Y, Wang Y, Xu Z, Chen D, Yu Z, Shao Q, Hong X, Liu Z, Cheng D. Small Molecules from Medicinal Plant Iris tectorum as Histidine Kinase Inhibitor to Resensitize β-Lactam-Resistant Escherichia coli. Molecules 2025; 30:663. [PMID: 39942767 PMCID: PMC11820157 DOI: 10.3390/molecules30030663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Due to the widespread use of broad-spectrum antibiotics, the problem of antibiotic resistance has become an increasingly serious global threat. One of the key mechanisms of Escherichia coli resistance to beta-lactam antibiotics is the production of beta-lactamase enzymes, which poses a dilemma for clinicians in selecting antibiotics when faced with resistant bacterial infections. However, research on the reversal of bacterial resistance is limited. METHODS This study involved the preparation of Iris tectorum extract and detection of its effects on antibiotics sensitivity, extended-spectrum beta-lactamase (ESBL) gene expression, and histidine kinase phosphorylation levels in β-lactam antibiotic-resistant Escherichia coli. Additionally, analyses of the active ingredients of Iris tectorum extract were conducted with a liquid chromatography-mass spectrometer, and the binding sites were predicted by molecular docking. RESULTS Iris tectorum extract could restore the sensitivity of Escherichia coli to beta-lactam antibiotics and reduce the expression levels of ESBL genes and histidine phosphorylation levels. The active ingredients of Iris tectorum extract may be irigenin and tectorigenin, and these two small molecules could bind to histidine kinase to inhibit phosphorylation. CONCLUSIONS Iris tectorum extract may serve as an antibiotic adjuvant, restoring the sensitivity of antibiotic-resistant bacteria by inhibiting histidine kinase phosphorylation, thereby alleviating the problem of Escherichia coli resistance to β-lactam antibiotics.
Collapse
Affiliation(s)
- Youqi Ji
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.J.); (Z.X.); (D.C.); (Z.Y.); (Q.S.)
| | - Yinhuan Wang
- Zhejiang Institute for Food and Drug Control, Hangzhou 310052, China;
| | - Zhangkai Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.J.); (Z.X.); (D.C.); (Z.Y.); (Q.S.)
| | - Danlei Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.J.); (Z.X.); (D.C.); (Z.Y.); (Q.S.)
| | - Zhendi Yu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.J.); (Z.X.); (D.C.); (Z.Y.); (Q.S.)
| | - Qingyi Shao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.J.); (Z.X.); (D.C.); (Z.Y.); (Q.S.)
| | - Xin Hong
- Department of Green Pharmaceutical Collaborative Innovation Center, School of Pharmacy, Zhejiang University of Technology, Hangzhou 310014, China;
| | - Zishu Liu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dongqing Cheng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.J.); (Z.X.); (D.C.); (Z.Y.); (Q.S.)
| |
Collapse
|
4
|
Ji S, Li C, Liu M, Liu Y, Jiang L. Targeting New Functions and Applications of Bacterial Two-Component Systems. Chembiochem 2024; 25:e202400392. [PMID: 38967093 DOI: 10.1002/cbic.202400392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/06/2024]
Abstract
Two-component signal transduction systems (TCSs) are regulatory systems widely distributed in eubacteria, archaea, and a few eukaryotic organisms, but not in mammalian cells. A typical TCS consists of a histidine kinase and a response regulator protein. Functional and mechanistic studies on different TCSs have greatly advanced the understanding of cellular phosphotransfer signal transduction mechanisms. In this concept paper, we focus on the His-Asp phosphotransfer mechanism, the ATP synthesis function, antimicrobial drug design, cellular biosensors design, and protein allostery mechanisms based on recent TCS investigations to inspire new applications and future research perspectives.
Collapse
Affiliation(s)
- Shixia Ji
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Magnetic Resonance in Biological System, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430074, China
| | - Conggang Li
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Magnetic Resonance in Biological System, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430074, China
| | - Maili Liu
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Magnetic Resonance in Biological System, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430074, China
| | - Yixiang Liu
- Key Laboratory of Magnetic Resonance in Biological System, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430074, China
| | - Ling Jiang
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
- Key Laboratory of Magnetic Resonance in Biological System, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430074, China
| |
Collapse
|
5
|
Bisht R, Charlesworth PD, Sperandeo P, Polissi A. Breaking Barriers: Exploiting Envelope Biogenesis and Stress Responses to Develop Novel Antimicrobial Strategies in Gram-Negative Bacteria. Pathogens 2024; 13:889. [PMID: 39452760 PMCID: PMC11510100 DOI: 10.3390/pathogens13100889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024] Open
Abstract
Antimicrobial resistance (AMR) has emerged as a global health threat, necessitating immediate actions to develop novel antimicrobial strategies and enforce strong stewardship of existing antibiotics to manage the emergence of drug-resistant strains. This issue is particularly concerning when it comes to Gram-negative bacteria, which possess an almost impenetrable outer membrane (OM) that acts as a formidable barrier to existing antimicrobial compounds. This OM is an asymmetric structure, composed of various components that confer stability, fluidity, and integrity to the bacterial cell. The maintenance and restoration of membrane integrity are regulated by envelope stress response systems (ESRs), which monitor its assembly and detect damages caused by external insults. Bacterial communities encounter a wide range of environmental niches to which they must respond and adapt for survival, sustenance, and virulence. ESRs play crucial roles in coordinating the expression of virulence factors, adaptive physiological behaviors, and antibiotic resistance determinants. Given their role in regulating bacterial cell physiology and maintaining membrane homeostasis, ESRs present promising targets for drug development. Considering numerous studies highlighting the involvement of ESRs in virulence, antibiotic resistance, and alternative resistance mechanisms in pathogens, this review aims to present these systems as potential drug targets, thereby encouraging further research in this direction.
Collapse
Affiliation(s)
| | | | - Paola Sperandeo
- Department of Pharmacological and Biomolecular Sciences, University of Milano, 20133 Milano, Italy; (R.B.); (P.D.C.); (A.P.)
| | | |
Collapse
|
6
|
Majdi C, Meffre P, Benfodda Z. Recent advances in the development of bacterial response regulators inhibitors as antibacterial and/or antibiotic adjuvant agent: A new approach to combat bacterial resistance. Bioorg Chem 2024; 150:107606. [PMID: 38968903 DOI: 10.1016/j.bioorg.2024.107606] [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: 03/25/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
The number of new antibacterial agents currently being discovered is insufficient to combat bacterial resistance. It is extremely challenging to find new antibiotics and to introduce them to the pharmaceutical market. Therefore, special attention must be given to find new strategies to combat bacterial resistance and prevent bacteria from developing resistance. Two-component system is a transduction system and the most prevalent mechanism employed by bacteria to respond to environmental changes. This signaling system consists of a membrane sensor histidine kinase that perceives environmental stimuli and a response regulator which acts as a transcription factor. The approach consisting of developing response regulators inhibitors with antibacterial activity or antibiotic adjuvant activity is a novel approach that has never been previously reviewed. In this review we report for the first time, the importance of targeting response regulators and summarizing all existing studies carried out from 2008 until now on response regulators inhibitors as antibacterial agents or / and antibiotic adjuvants. Moreover, we describe the antibacterial activity and/or antibiotic adjuvants activity against the studied bacterial strains and the mechanism of different response regulator inhibitors when it's possible.
Collapse
|
7
|
Lobertti CA, Cabezudo I, Gizzi FO, Blancato V, Magni C, Furlán RLE, García Véscovi E. An allosteric inhibitor of the PhoQ histidine kinase with therapeutic potential against Salmonella infection. J Antimicrob Chemother 2024; 79:1820-1830. [PMID: 38853496 DOI: 10.1093/jac/dkae151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 04/30/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND The upsurge of antimicrobial resistance demands innovative strategies to fight bacterial infections. With traditional antibiotics becoming less effective, anti-virulence agents or pathoblockers, arise as an alternative approach that seeks to disarm pathogens without affecting their viability, thereby reducing selective pressure for the emergence of resistance mechanisms. OBJECTIVES To elucidate the mechanism of action of compound N'-(thiophen-2-ylmethylene)benzohydrazide (A16B1), a potent synthetic hydrazone inhibitor against the Salmonella PhoP/PhoQ system, essential for virulence. MATERIALS AND METHODS The measurement of the activity of PhoP/PhoQ-dependent and -independent reporter genes was used to evaluate the specificity of A16B1 to the PhoP regulon. Autokinase activity assays with either the native or truncated versions of PhoQ were used to dissect the A16B1 mechanism of action. The effect of A16B1 on Salmonella intramacrophage replication was assessed using the gentamicin protection assay. The checkerboard assay approach was used to analyse potentiation effects of colistin with the hydrazone. The Galleria mellonella infection model was chosen to evaluate A16B1 as an in vivo therapy against Salmonella. RESULTS A16B1 repressed the Salmonella PhoP/PhoQ system activity, specifically targeting PhoQ within the second transmembrane region. A16B1 demonstrates synergy with the antimicrobial peptide colistin, reduces the intramacrophage proliferation of Salmonella without being cytotoxic and enhances the survival of G. mellonella larvae systemically infected with Salmonella. CONCLUSIONS A16B1 selectively inhibits the activity of the Salmonella PhoP/PhoQ system through a novel inhibitory mechanism, representing a promising synthetic hydrazone compound with the potential to function as a Salmonella pathoblocker. This offers innovative prospects for combating Salmonella infections while mitigating the risk of antimicrobial resistance emergence.
Collapse
Affiliation(s)
- Carlos A Lobertti
- Instituto de Biología Molecular y Celular de Rosario Consejo Nacional de Investigaciones Científicas y Técnicas and Facultad de Ciencias Bioquímicas y Farmacéuticas, Departamento de Microbiología, Universidad Nacional de Rosario, Rosario S2000EZP, Argentina
| | - Ignacio Cabezudo
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rosario 2000, Argentina
| | - Fernán O Gizzi
- Instituto de Biología Molecular y Celular de Rosario Consejo Nacional de Investigaciones Científicas y Técnicas and Facultad de Ciencias Bioquímicas y Farmacéuticas, Departamento de Microbiología, Universidad Nacional de Rosario, Rosario S2000EZP, Argentina
| | - Víctor Blancato
- Instituto de Biología Molecular y Celular de Rosario Consejo Nacional de Investigaciones Científicas y Técnicas and Facultad de Ciencias Bioquímicas y Farmacéuticas, Departamento de Microbiología, Universidad Nacional de Rosario, Rosario S2000EZP, Argentina
| | - Christian Magni
- Instituto de Biología Molecular y Celular de Rosario Consejo Nacional de Investigaciones Científicas y Técnicas and Facultad de Ciencias Bioquímicas y Farmacéuticas, Departamento de Microbiología, Universidad Nacional de Rosario, Rosario S2000EZP, Argentina
| | - Ricardo L E Furlán
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rosario 2000, Argentina
| | - Eleonora García Véscovi
- Instituto de Biología Molecular y Celular de Rosario Consejo Nacional de Investigaciones Científicas y Técnicas and Facultad de Ciencias Bioquímicas y Farmacéuticas, Departamento de Microbiología, Universidad Nacional de Rosario, Rosario S2000EZP, Argentina
| |
Collapse
|
8
|
Ishikawa T, Eguchi Y, Igarashi M, Okajima T, Mita K, Yamasaki Y, Sumikura K, Okumura T, Tabuchi Y, Hayashi C, Pasqua M, Coluccia M, Prosseda G, Colonna B, Kohayakawa C, Tani A, Haruta JI, Utsumi R. Synthesis and biochemical characterization of naphthoquinone derivatives targeting bacterial histidine kinases. J Antibiot (Tokyo) 2024; 77:522-532. [PMID: 38918599 PMCID: PMC11284088 DOI: 10.1038/s41429-024-00726-2] [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: 11/22/2023] [Revised: 03/25/2024] [Accepted: 03/29/2024] [Indexed: 06/27/2024]
Abstract
Waldiomycin is an inhibitor of histidine kinases (HKs). Although most HK inhibitors target the ATP-binding region, waldiomycin binds to the intracellular dimerization domain (DHp domain) with its naphthoquinone moiety presumed to interact with the conserved H-box region. To further develop inhibitors targeting the H-box, various 2-aminonaphthoquinones with cyclic, aliphatic, or aromatic amino groups and naphtho [2,3-d] isoxazole-4,9-diones were synthesized. These compounds were tested for their inhibitory activity (IC50) against WalK, an essential HK for Bacillus subtilis growth, and their minimum inhibitory concentrations (MIC) against B. subtilis. As a result, 11 novel HK inhibitors were obtained as naphthoquinone derivatives (IC50: 12.6-305 µM, MIC: 0.5-128 µg ml-1). The effect of representative compounds on the expression of WalK/WalR regulated genes in B. subtilis was investigated. Four naphthoquinone derivatives induced the expression of iseA (formerly yoeB), whose expression is negatively regulated by the WalK/WalR system. This suggests that these compounds inhibit WalK in B. subtilis cells, resulting in antibacterial activity. Affinity selection/mass spectrometry analysis was performed to identify whether these naphthoquinone derivatives interact with WalK in a manner similar to waldiomycin. Three compounds were found to competitively inhibit the binding of waldiomycin to WalK, suggesting that they bind to the H-box region conserved in HKs and inhibit HK activity.
Collapse
Affiliation(s)
| | - Yoko Eguchi
- Department of Science and Technology on Food Safety, Faculty of Biology-Oriented Science and Technology, Kindai University, Wakayama, Japan
| | | | - Toshihide Okajima
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan
| | - Kohei Mita
- Graduate School of Education, Okayama University, Okayama, Japan
| | - Yuri Yamasaki
- Graduate School of Education, Okayama University, Okayama, Japan
| | - Kaho Sumikura
- Graduate School of Education, Okayama University, Okayama, Japan
| | - Taisei Okumura
- Graduate School of Education, Okayama University, Okayama, Japan
| | - Yuna Tabuchi
- Graduate School of Education, Okayama University, Okayama, Japan
| | | | - Martina Pasqua
- Istituto Pasteur Italy, Department of Biology and Biotechnology, "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Marco Coluccia
- Istituto Pasteur Italy, Department of Biology and Biotechnology, "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Gianni Prosseda
- Istituto Pasteur Italy, Department of Biology and Biotechnology, "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Bianca Colonna
- Istituto Pasteur Italy, Department of Biology and Biotechnology, "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Chie Kohayakawa
- Department of Lead Exploration Units, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akiyoshi Tani
- Compound Library Screening Center, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Jun-Ichi Haruta
- Department of Lead Exploration Units, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Ryutaro Utsumi
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, Osaka, Japan.
| |
Collapse
|
9
|
Fihn CA, Lembke HK, Gaulin J, Bouchard P, Villarreal AR, Penningroth MR, Crone KK, Vogt GA, Gilbertsen AJ, Ayotte Y, de Oliveira LC, Serrano-Wu MH, Drouin N, Hung DT, Hunter RC, Carlson EE. Evaluation of Expanded 2-Aminobenzothiazole Library for Inhibition of Pseudomonas aeruginosa Virulence Phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.02.539119. [PMID: 37205454 PMCID: PMC10187220 DOI: 10.1101/2023.05.02.539119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Bacterial resistance to antibiotics is a rapidly increasing threat to human health. New strategies to combat resistant organisms are desperately needed. One potential avenue is targeting two-component systems, which are the main bacterial signal transduction pathways used to regulate development, metabolism, virulence, and antibiotic resistance. These systems consist of a homodimeric membrane-bound sensor histidine kinase, and a cognate effector, the response regulator. The high sequence conservation in the catalytic and adenosine triphosphate-binding (CA) domain of histidine kinases and their essential role in bacterial signal transduction could enable broad-spectrum antibacterial activity. Through this signal transduction, histidine kinases regulate multiple virulence mechanisms including toxin production, immune evasion, and antibiotic resistance. Targeting virulence, as opposed to development of bactericidal compounds, could reduce evolutionary pressure for acquired resistance. Additionally, compounds targeting the CA domain have the potential to impair multiple two-component systems that regulate virulence in one or more pathogens. We conducted structure-activity relationship studies of 2-aminobenzothiazole-based inhibitors designed to target the CA domain of histidine kinases. We found these compounds have anti-virulence activities in Pseudomonas aeruginosa, reducing motility phenotypes and toxin production associated with the pathogenic functions of this bacterium.
Collapse
Affiliation(s)
- Conrad A. Fihn
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, Minnesota 55455, United States
| | - Hannah K. Lembke
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55454, United States
| | - Jeffrey Gaulin
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Patricia Bouchard
- NMX Research and Solution Inc., 500 Cartier Boulevard W., Suite 6000, Laval, Quebec, Canada, H1Y 2R1
| | - Alex R. Villarreal
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Mitchell R. Penningroth
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Kathryn K. Crone
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55455, United States
| | - Grace A. Vogt
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Adam J. Gilbertsen
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Yann Ayotte
- NMX Research and Solution Inc., 500 Cartier Boulevard W., Suite 6000, Laval, Quebec, Canada, H1Y 2R1
| | | | | | - Nathalie Drouin
- NMX Research and Solution Inc., 500 Cartier Boulevard W., Suite 6000, Laval, Quebec, Canada, H1Y 2R1
| | - Deborah T. Hung
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Ryan C. Hunter
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Ave Se Minneapolis, Minnesota 55455, United States
| | - Erin E. Carlson
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55454, United States
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
10
|
Pathak RK, Kim JM. Identification of histidine kinase inhibitors through screening of natural compounds to combat mastitis caused by Streptococcus agalactiae in dairy cattle. J Biol Eng 2023; 17:59. [PMID: 37752501 PMCID: PMC10523694 DOI: 10.1186/s13036-023-00378-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Mastitis poses a major threat to dairy farms globally; it results in reduced milk production, increased treatment costs, untimely compromised genetic potential, animal deaths, and economic losses. Streptococcus agalactiae is a highly virulent bacteria that cause mastitis. The administration of antibiotics for the treatment of this infection is not advised due to concerns about the emergence of antibiotic resistance and potential adverse effects on human health. Thus, there is a critical need to identify new therapeutic approaches to combat mastitis. One promising target for the development of antibacterial therapies is the transmembrane histidine kinase of bacteria, which plays a key role in signal transduction pathways, secretion systems, virulence, and antibiotic resistance. RESULTS In this study, we aimed to identify novel natural compounds that can inhibit transmembrane histidine kinase. To achieve this goal, we conducted a virtual screening of 224,205 natural compounds, selecting the top ten based on their lowest binding energy and favorable protein-ligand interactions. Furthermore, molecular docking of eight selected antibiotics and five histidine kinase inhibitors with transmembrane histidine kinase was performed to evaluate the binding energy with respect to top-screened natural compounds. We also analyzed the ADMET properties of these compounds to assess their drug-likeness. The top two compounds (ZINC000085569031 and ZINC000257435291) and top-screened antibiotics (Tetracycline) that demonstrated a strong binding affinity were subjected to molecular dynamics simulations (100 ns), free energy landscape, and binding free energy calculations using the MM-PBSA method. CONCLUSION Our results suggest that the selected natural compounds have the potential to serve as effective inhibitors of transmembrane histidine kinase and can be utilized for the development of novel antibacterial veterinary medicine for mastitis after further validation through clinical studies.
Collapse
Affiliation(s)
- Rajesh Kumar Pathak
- Department of Animal Science and Technology, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea.
| |
Collapse
|
11
|
Subtractive genomics profiling for potential drug targets identification against Moraxella catarrhalis. PLoS One 2022; 17:e0273252. [PMID: 36006987 PMCID: PMC9409589 DOI: 10.1371/journal.pone.0273252] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/04/2022] [Indexed: 01/12/2023] Open
Abstract
Moraxella catarrhalis (M. catarrhalis) is a gram-negative bacterium, responsible for major respiratory tract and middle ear infection in infants and adults. The recent emergence of the antibiotic resistance M. catarrhalis demands the prioritization of an effective drug target as a top priority. Fortunately, the failure of new drugs and host toxicity associated with traditional drug development approaches can be avoided by using an in silico subtractive genomics approach. In the current study, the advanced in silico genome subtraction approach was applied to identify potential and pathogen-specific drug targets against M. catarrhalis. We applied a series of subtraction methods from the whole genome of pathogen based on certain steps i.e. paralogous protein that have extensive homology with humans, essential, drug like, non-virulent, and resistant proteins. Only 38 potent drug targets were identified in this study. Eventually, one protein was identified as a potential new drug target and forwarded to the structure-based studies i.e. histidine kinase (UniProt ID: D5VAF6). Furthermore, virtual screening of 2000 compounds from the ZINC database was performed against the histidine kinase that resulted in the shortlisting of three compounds as the potential therapeutic candidates based on their binding energies and the properties exhibited using ADMET analysis. The identified protein gives a platform for the discovery of a lead drug candidate that may inhibit it and may help to eradicate the otitis media caused by drug-resistant M. catarrhalis. Nevertheless, the current study helped in creating a pipeline for drug target identification that may assist wet-lab research in the future.
Collapse
|
12
|
Chen H, Yu C, Wu H, Li G, Li C, Hong W, Yang X, Wang H, You X. Recent Advances in Histidine Kinase-Targeted Antimicrobial Agents. Front Chem 2022; 10:866392. [PMID: 35860627 PMCID: PMC9289397 DOI: 10.3389/fchem.2022.866392] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/16/2022] [Indexed: 11/15/2022] Open
Abstract
The prevalence of antimicrobial-resistant pathogens significantly limited the number of effective antibiotics available clinically, which urgently requires new drug targets to screen, design, and develop novel antibacterial drugs. Two-component system (TCS), which is comprised of a histidine kinase (HK) and a response regulator (RR), is a common mechanism whereby bacteria can sense a range of stimuli and make an appropriate adaptive response. HKs as the sensor part of the bacterial TCS can regulate various processes such as growth, vitality, antibiotic resistance, and virulence, and have been considered as a promising target for antibacterial drugs. In the current review, we highlighted the structural basis and functional importance of bacterial TCS especially HKs as a target in the discovery of new antimicrobials, and summarize the latest research progress of small-molecule HK-inhibitors as potential novel antimicrobial drugs reported in the past decade.
Collapse
Affiliation(s)
- Hongtong Chen
- Laboratory of Pharmacology/Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengqi Yu
- School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Han Wu
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Guoqing Li
- Laboratory of Pharmacology/Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Congran Li
- Laboratory of Pharmacology/Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Hong
- Beijing Institute of Collaborative Innovation, Beijing, China
| | - Xinyi Yang
- Laboratory of Pharmacology/Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Wang
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
- Institute of National Security, Minzu University of China, Beijing, China
| | - Xuefu You
- Laboratory of Pharmacology/Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Khan K, Jalal K, Khan A, Al-Harrasi A, Uddin R. Comparative Metabolic Pathways Analysis and Subtractive Genomics Profiling to Prioritize Potential Drug Targets Against Streptococcus pneumoniae. Front Microbiol 2022; 12:796363. [PMID: 35222301 PMCID: PMC8866961 DOI: 10.3389/fmicb.2021.796363] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/28/2021] [Indexed: 02/01/2023] Open
Abstract
Streptococcus pneumoniae is a notorious pathogen that affects ∼450 million people worldwide and causes up to four million deaths per annum. Despite availability of antibiotics (i.e., penicillin, doxycycline, or clarithromycin) and conjugate vaccines (e.g., PCVs), it is still challenging to treat because of its drug resistance ability. The rise of antibiotic resistance in S. pneumoniae is a major source of concern across the world. Computational subtractive genomics is one of the most applied techniques in which the whole proteome of the bacterial pathogen is gradually reduced to a limited number of potential therapeutic targets. Whole-genome sequencing has greatly reduced the time required and provides more opportunities for drug target identification. The goal of this work is to evaluate and analyze metabolic pathways in serotype 14 of S. pneumonia to identify potential drug targets. In the present study, 47 potent drug targets were identified against S. pneumonia by employing the computational subtractive genomics approach. Among these, two proteins are prioritized (i.e., 4-oxalocrotonate tautomerase and Sensor histidine kinase uniquely present in S. pneumonia) as novel drug targets and selected for further structure-based studies. The identified proteins may provide a platform for the discovery of a lead drug candidate that may be capable of inhibiting these proteins and, therefore, could be helpful in minimizing the associated risk related to the drug-resistant S. pneumoniae. Finally, these enzymatic proteins could be of prime interest against S. pneumoniae to design rational targeted therapy.
Collapse
Affiliation(s)
- Kanwal Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Khurshid Jalal
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
14
|
Expression, Purification, and Characterization of the Recombinant, Two-Component, Response Regulator ArlR from Fusobacterium nucleatum. Appl Biochem Biotechnol 2022; 194:2093-2107. [PMID: 35029789 DOI: 10.1007/s12010-021-03785-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2021] [Indexed: 11/02/2022]
Abstract
Fusobacterium nucleatum is associated with the incidence and development of multiple diseases, such as periodontitis and colorectal cancer (CRC). Until now, studies have proved only a few proteins to be associated with such pathogenic diseases. The two-component system is one of the most prevalent forms of bacterial signal transduction related to intestinal diseases. Here, we report a novel, recombinant, two-component, response regulator protein ArlR from the genome of F. nucleatum strain ATCC 25,586. We optimized the expression and purification conditions of ArlR; in addition, we characterized the interaction of this response regulator protein with the corresponding histidine kinase and DNA sequence. The full-length ArlR was successfully expressed in six E. coli host strains. However, optimum expression conditions of ArlR were present only in E. coli strain BL21 CodonPlus (DE3) RIL that was later induced with isopropyl β-D-1-thiogalactopyranoside (IPTG) for 8 h at 25 °C. The SDS-PAGE analysis revealed the molecular weight of the recombinant protein as 27.3 kDa with approximately 90% purity after gel filtration chromatography. Because ArlR was biologically active after its purification, it accepted the corresponding phosphorylated histidine kinase phosphate group and bound to the analogous DNA sequence. The binding constant between ArlR and the corresponding histidine kinase was about 2.1 μM, whereas the binding constant between ArlR and its operon was 6.4 μM. Altogether, these results illustrate an effective expression and purification method for the novel two-component system protein ArlR.
Collapse
|
15
|
Ma P, Phillips-Jones MK. Membrane Sensor Histidine Kinases: Insights from Structural, Ligand and Inhibitor Studies of Full-Length Proteins and Signalling Domains for Antibiotic Discovery. Molecules 2021; 26:molecules26165110. [PMID: 34443697 PMCID: PMC8399564 DOI: 10.3390/molecules26165110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/02/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022] Open
Abstract
There is an urgent need to find new antibacterial agents to combat bacterial infections, including agents that inhibit novel, hitherto unexploited targets in bacterial cells. Amongst novel targets are two-component signal transduction systems (TCSs) which are the main mechanism by which bacteria sense and respond to environmental changes. TCSs typically comprise a membrane-embedded sensory protein (the sensor histidine kinase, SHK) and a partner response regulator protein. Amongst promising targets within SHKs are those involved in environmental signal detection (useful for targeting specific SHKs) and the common themes of signal transmission across the membrane and propagation to catalytic domains (for targeting multiple SHKs). However, the nature of environmental signals for the vast majority of SHKs is still lacking, and there is a paucity of structural information based on full-length membrane-bound SHKs with and without ligand. Reasons for this lack of knowledge lie in the technical challenges associated with investigations of these relatively hydrophobic membrane proteins and the inherent flexibility of these multidomain proteins that reduces the chances of successful crystallisation for structural determination by X-ray crystallography. However, in recent years there has been an explosion of information published on (a) methodology for producing active forms of full-length detergent-, liposome- and nanodisc-solubilised membrane SHKs and their use in structural studies and identification of signalling ligands and inhibitors; and (b) mechanisms of signal sensing and transduction across the membrane obtained using sensory and transmembrane domains in isolation, which reveal some commonalities as well as unique features. Here we review the most recent advances in these areas and highlight those of potential use in future strategies for antibiotic discovery. This Review is part of a Special Issue entitled “Interactions of Bacterial Molecules with Their Ligands and Other Chemical Agents” edited by Mary K. Phillips-Jones.
Collapse
Affiliation(s)
- Pikyee Ma
- Laboratory of Biomolecular Research, Paul Scherrer Institute, CH-5232 Villigen, Switzerland;
| | - Mary K. Phillips-Jones
- National Centre for Macromolecular Hydrodynamics, School of Biosciences, University of Nottingham, Sutton Bonington LE12 5RD, UK
- Correspondence:
| |
Collapse
|
16
|
Lin Y, Chen J, Zhou X, Li Y. Inhibition of Streptococcus mutans biofilm formation by strategies targeting the metabolism of exopolysaccharides. Crit Rev Microbiol 2021; 47:667-677. [PMID: 33938347 DOI: 10.1080/1040841x.2021.1915959] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Dental caries is one of the most prevalent and costly biofilm-associated infectious diseases affecting most of the world's population. In particular, dental caries is driven by dysbiosis of the dental biofilm adherent to the enamel surface. Specific types of acid-producing bacteria, especially Streptococcus mutans, colonize the dental surface and cause damage to the hard tooth structure in the presence of fermentable carbohydrates. Streptococcus mutans has been established as the major cariogenic pathogen responsible for human dental caries, with a high ability to form biofilms. The exopolysaccharide (EPS) matrix, mainly contributed by S. mutans, has been considered as a virulence determinant of cariogenic biofilm. As EPS is an important virulence factor, targeting EPS metabolism could be useful in preventing cariogenic biofilm formation. This review summarizes plausible strategies targeting S. mutans biofilms by degrading EPS structure, inhibiting EPS production, and disturbing the EPS metabolism-related gene expression and regulatory systems.
Collapse
Affiliation(s)
- Yongwang Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiamin Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|