1
|
Yan D, Kong H, Qu Y, Li R, Ampomah-Wireko M, Liu J, Qin S, Wang Z, Li W, Zhang E. Development of Phloroglucinol-Linked Tris-Quaternary Ammonium Salt Antimicrobial Peptide Mimics with Low Cytotoxicity and Broad-Spectrum Antibacterial Activity. J Med Chem 2024; 67:18576-18592. [PMID: 39376107 DOI: 10.1021/acs.jmedchem.4c01935] [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: 10/09/2024]
Abstract
Encouraged by the significantly different toxicities and antibacterial activities of diverse linkers, such as alkyl and aromatic nuclei linkers, and the unique structure of phloroglucinol, we synthesized a series of tris-quaternary ammonium salt (tris-QAS) antibacterial peptide mimics based on the marketed drug phloroglucinol. Among them, 2f displayed excellent activity against Staphylococcus aureus (MIC = 0.5 μg/mL) and high selectivity (SI > 2560). Surprisingly, the cytotoxicity of 2f (CC50 = 152.7 μg/mL) was dramatically better than those of alkyl QAS I and hydroquinone QAS II. Additionally, 2f possessed rapid bactericidal capability, was not prone to inducing bacterial resistance, and also exhibited excellent activity against S. aureus biofilms and persistent bacteria. Mechanistic research and transcriptome analysis revealed that 2f can interfere with the normal metabolism of bacterial cells, and it can specifically bind with phosphatidylglycerol to destroy the cell membrane. Importantly, 2f exhibited potent in vivo antibacterial activity in a mouse subcutaneous methicillin-resistant S. aureus (MRSA) infection model.
Collapse
Affiliation(s)
- Dachao Yan
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Hongtao Kong
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Ye Qu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Ruirui Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Jifeng Liu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenya Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - Wen Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| | - En Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
- Pingyuan Laboratory (Zhengzhou University), Zhengzhou 450001, China
| |
Collapse
|
2
|
Huang HY, Xue RY, Xiao SX, Huang LT, Liao XW, Wang JT, Duan XM, Yu RJ, Xiong YS. AIE-based ruthenium complexes as photosensitizers for specifically photo-inactivate gram-positive bacteria. J Inorg Biochem 2024; 262:112755. [PMID: 39388808 DOI: 10.1016/j.jinorgbio.2024.112755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
The emergence of multidrug-resistant bacterial have caused severe burden for public health. Particularly, Staphylococcus aureus as one of ESKAPE pathogens have induced various infectious diseases and resulted in increasing deaths. Developing new antibacterial agents is still urgent and challenging. Fortunately, in this study, based on aggregation-induced emission (AIE) ruthenium complexes were designed and synthesized, which realized the high efficiency of reactive oxygen species generation and remarkably killed S. aureus unlike conventional antibiotics action. Significantly, owing to good singlet oxygen production ability, Ru1 at only 4 μg/mL of concentration displayed good antibacterial photodynamic therapy effect upon white light irradiation and could deplete essential coenzyme NADH to disrupt intracellular redox balance. Also, the electrostatic interaction between Ru1 and bacteria enhanced the possibility of antibacterial. Under light irradiation, Ru1 could efficiently inhibit the biofilm growth and avoid the development of drug-resistant. Furthermore, Ru1 possessed excellent biocompatibility and displayed remarkable therapy effect in treating mice-wound infections in vivo. These findings indicated that AIE-based ruthenium complexes as new antibacterial agent had great potential in photodynamic therapy of bacteria and addressing the drug-resistance crisis.
Collapse
Affiliation(s)
- Hai-Yan Huang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Run-Yu Xue
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Su-Xin Xiao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Li-Ting Huang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Xiang-Wen Liao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Jin-Tao Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Xue-Min Duan
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Ru-Jian Yu
- School of life science, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China
| | - Yan-Shi Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, Jiangxi, PR China.
| |
Collapse
|
3
|
Vishwakarma A, Narayanan A, Kumar N, Chen Z, Dang F, Menefee J, Dhinojwala A, Joy A. Coacervate Dense Phase Displaces Surface-Established Pseudomonas aeruginosa Biofilms. J Am Chem Soc 2024; 146:26397-26407. [PMID: 39259884 PMCID: PMC11440510 DOI: 10.1021/jacs.4c09311] [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: 09/13/2024]
Abstract
For millions of years, barnacles and mussels have successfully adhered to wet rocks near tide-swept seashores. While the chemistry and mechanics of their underwater adhesives are being thoroughly investigated, an overlooked aspect of marine organismal adhesion is their ability to remove underlying biofilms from rocks and prepare clean surfaces before the deposition of adhesive anchors. Herein, we demonstrate that nonionic, coacervating synthetic polymers that mimic the physicochemical features of marine underwater adhesives remove ∼99% of Pseudomonas aeruginosa (P. aeruginosa) biofilm biomass from underwater surfaces. The efficiency of biofilm removal appears to align with the compositional differences between various bacterial biofilms. In addition, the surface energy influences the ability of the polymer to displace the biofilm, with biofilm removal efficiency decreasing for surfaces with lower surface energies. These synthetic polymers weaken the biofilm-surface interactions and exert shear stress to fracture the biofilms grown on surfaces with diverse surface energies. Since bacterial biofilms are 1000-fold more tolerant to common antimicrobial agents and pose immense health and economic risks, we anticipate that our unconventional approach inspired by marine underwater adhesion will open a new paradigm in creating antibiofilm agents that target the interfacial and viscoelastic properties of established bacterial biofilms.
Collapse
Affiliation(s)
- Apoorva Vishwakarma
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Amal Narayanan
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Nityanshu Kumar
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Zixi Chen
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02120, United States
| | - Francis Dang
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Joshua Menefee
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Ali Dhinojwala
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Abraham Joy
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02120, United States
| |
Collapse
|
4
|
Bahatheg G, Kuppusamy R, Yasir M, Bridge S, Mishra SK, Cranfield CG, StC Black D, Willcox M, Kumar N. Dimeric peptoids as antibacterial agents. Bioorg Chem 2024; 147:107334. [PMID: 38583251 DOI: 10.1016/j.bioorg.2024.107334] [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: 01/29/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Building upon our previous study on peptoid-based antibacterials which showed good activity against Gram-positive bacteria only, herein we report the synthesis of 34 dimeric peptoid compounds and the investigation of their activity against Gram-positive and Gram-negative pathogens. The newly designed peptoids feature a di-hydrophobic moiety incorporating phenyl, bromo-phenyl, and naphthyl groups, combined with variable lengths of cationic units such as amino and guanidine groups. The study also underscores the pivotal interplay between hydrophobicity and cationicity in optimizing efficacy against specific bacteria. The bromophenyl dimeric guanidinium peptoid compound 10j showed excellent activity against S. aureus 38 and E. coli K12 with MIC of 0.8 μg mL-1 and 6.2 μg mL-1, respectively. Further investigation into the mechanism of action revealed that the antibacterial effect might be attributed to the disruption of bacterial cell membranes, as suggested by tethered bilayer lipid membranes (tBLMs) and cytoplasmic membrane permeability studies. Notably, these promising antibacterial agents exhibited negligible toxicity against mammalian red blood cells. Additionally, the study explored the potential of 12 active compounds to disrupt established biofilms of S. aureus 38. The most effective biofilm disruptors were ethyl and octyl-naphthyl guanidinium peptoids (10c and 10 k). These compounds 10c and 10 k disrupted the established biofilms of S. aureus 38 with 51 % at 4x MIC (MIC = 17.6 μg mL-1 and 11.2 μg mL-1) and 56 %-58 % at 8x MIC (MIC = 35.2 μg mL-1 and 22.4 μg mL-1) respectively. Overall, this research contributes insights into the design principles of cationic dimeric peptoids and their antibacterial activity, with implications for the development of new antibacterial compounds.
Collapse
Affiliation(s)
- Ghayah Bahatheg
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia; Department of Chemistry, Faculty of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Rajesh Kuppusamy
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia; School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| | - Muhammad Yasir
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Samara Bridge
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia
| | - Shyam K Mishra
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Charles G Cranfield
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia
| | - David StC Black
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Mark Willcox
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Naresh Kumar
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| |
Collapse
|
5
|
Xue M, Chakraborty S, Gao R, Wang S, Gu M, Shen N, Wei L, Cao C, Sun X, Cai J. Antimicrobial Guanidinylate Polycarbonates Show Oral In Vivo Efficacy Against Clostridioides Difficile. Adv Healthc Mater 2024; 13:e2303295. [PMID: 38321619 PMCID: PMC11144102 DOI: 10.1002/adhm.202303295] [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: 09/27/2023] [Revised: 01/22/2024] [Indexed: 02/08/2024]
Abstract
The emerging antibiotic resistance has been named by the World Health Organization (WHO) as one of the top 10 threats to public health. Notably, methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus faecalis (VREF) are designated as serious threats, whereas Clostridioides difficile (C. difficile) is recognized as one of the most urgent threats to human health and unmet medical need. Herein, they report the design and application of novel biodegradable polymers - the lipidated antimicrobial guanidinylate polycarbonates. These polymers showed potent antimicrobial activity against a panel of bacteria with fast-killing kinetics and low resistance development tendency, mainly due to their bacterial membrane disruption mechanism. More importantly, the optimal polymer showed excellent antibacterial activity against C. difficile infection (CDI) in vivo via oral administration. In addition, compared with vancomycin, the polymer demonstrated a much-prolonged therapeutic effect and virtually diminished recurrence rate of CDI. The convenient synthesis, easy scale-up, low cost, as well as biodegradability of this class of polycarbonates, together with their in vitro broad-spectrum antimicrobial activity and orally in vivo efficacy against CDI, suggest the great potential of lipidated guandinylate polycarbonates as a new class of antibacterial biomaterials to treat CDI and combat emerging antibiotic resistance.
Collapse
Affiliation(s)
- Menglin Xue
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave., Tampa, FL 33620, USA
| | - Soumyadeep Chakraborty
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Ruixuan Gao
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave., Tampa, FL 33620, USA
| | - Shaohui Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Meng Gu
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave., Tampa, FL 33620, USA
| | - Ning Shen
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave., Tampa, FL 33620, USA
| | - Lulu Wei
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave., Tampa, FL 33620, USA
| | - Chuanhai Cao
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave., Tampa, FL 33620, USA
| |
Collapse
|
6
|
Jin Y, Liu Y, Zhu J, Liu H. Pillararenes: a new frontier in antimicrobial therapy. Org Biomol Chem 2024; 22:4202-4211. [PMID: 38727528 DOI: 10.1039/d4ob00396a] [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: 05/30/2024]
Abstract
Pillararenes have gained great interest among researchers in many fields due to their symmetric structure and facile functionalization. In this review, we summarize recent progress for pillararenes as antimicrobial agents, ranging from cationic pillararenes and peptide-modified pillararenes to sugar-functionalized pillararenes. Moreover, their structure-activity relationships are presented, and their mechanisms of action are discussed. As a state-of-the-art technology, their opportunities and outlook are also outlined in this emerging field. Overall, their potent inhibitory activity and high biocompatibility give them potential for the development of novel antimicrobial agents.
Collapse
Affiliation(s)
- Yanqing Jin
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering & Pharmacy, Wuhan Institute of Technology, 693 Xiongchu Avenue, Wuhan 430073, P. R. China.
| | - Yisu Liu
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering & Pharmacy, Wuhan Institute of Technology, 693 Xiongchu Avenue, Wuhan 430073, P. R. China.
| | - Jiang Zhu
- Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical college, Nanchong 637000, Sichuan, P. R. China
| | - Hui Liu
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering & Pharmacy, Wuhan Institute of Technology, 693 Xiongchu Avenue, Wuhan 430073, P. R. China.
| |
Collapse
|
7
|
Gao S, Deng J, Su Z, Liu M, Tang S, Hu T, Qi E, Fu C, Pan GY. Turning Polysaccharides into Injectable and Rapid Self-Healing Antibacterial Hydrogels for Antibacterial Treatment and Bacterial-Infected Wound Healing. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:9082-9096. [PMID: 38619979 DOI: 10.1021/acs.langmuir.4c00451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Great efforts have been devoted to the development of novel and multifunctional wound dressing materials to meet the different needs of wound healing. Herein, we covalently grafted quaternary ammonium groups (QAGs) containing 12-carbon straight-chain alkanes to the dextran polymer skeleton. We then oxidized the resulting product into oxidized quaternized dextran (OQD). The obtained OQD polymer is rich in antibacterial QAGs and aldehyde groups. It can react with glycol chitosan (GC) via the Schiff-base reaction to form a multifunctional GC@OQD hydrogel with good self-healing behavior, hemostasis, injectability, inherent superior antibacterial activity, biocompatibility, and excellent promotion of healing of methicillin-resistant Staphylococcus aureus (MRSA)-infected wounds. The biosafe and nontoxic GC@OQD hydrogel with a three-dimensional porous network structure possesses an excellent swelling rate and water retention capacity. It can be used for hemostasis and treating irregular wounds. The designed GC@OQD hydrogel with inherent antibacterial activity possesses good antibacterial efficacy on both S. aureus (Gram-positive bacteria) and Escherichia coli (Gram-negative bacteria), as well as MRSA bacteria, with antibacterial activity greater than 99%. It can be used for the treatment of wounds infected by MRSA and significantly promotes the healing of wounds. Thus, the multifunctional antibacterial GC@OQD hydrogel has the potential to be applied in clinical practice as a wound dressing.
Collapse
Affiliation(s)
- Shiqi Gao
- School of Pharmacy, Guilin Medical University, Guilin 541100, P. R. China
| | - Jianbin Deng
- School of Pharmacy, Guilin Medical University, Guilin 541100, P. R. China
| | - Zhicheng Su
- School of Pharmacy, Guilin Medical University, Guilin 541100, P. R. China
| | - Mengqi Liu
- School of Pharmacy, Guilin Medical University, Guilin 541100, P. R. China
| | - Songyun Tang
- School of Pharmacy, Guilin Medical University, Guilin 541100, P. R. China
| | - Tingting Hu
- College of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541100, P. R. China
| | - Enfeng Qi
- School of Mathematics and Statistics, Guangxi Normal University, Guilin 541000, P. R. China
| | - Can Fu
- College of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541100, P. R. China
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, Guilin 541100, P. R. China
| | - Guang-Yu Pan
- College of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541100, P. R. China
- Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, Guilin 541100, China
| |
Collapse
|
8
|
Jothi Nayaki S, Roja A, Ravindhiran R, Sivarajan K, Arunachalam M, Dhandapani K. Pillar[ n]arenes in the Fight against Biofilms: Current Developments and Future Perspectives. ACS Infect Dis 2024; 10:1080-1096. [PMID: 38546344 DOI: 10.1021/acsinfecdis.3c00697] [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] [Indexed: 04/13/2024]
Abstract
The global surge in bacterial infections, compounded by the alarming escalation of drug-resistant strains, has evolved into a critical public health crisis. Among the challenges posed, biofilms stand out due to their formidable resistance to conventional antibiotics. This review delves into the burgeoning potential of pillar[n]arenes, distinctive macrocyclic host molecules, as promising anti-biofilm agents. The review is structured into two main sections, each dedicated to exploring distinct facets of pillar[n]arene applications. The first section scrutinizes functionalized pillar[n]arenes with a particular emphasis on cationic derivatives. This analysis reveals their significant efficacy in inhibiting biofilm formation, underscoring the pivotal role of specific chemical attributes in combating microbial communities. The second section of the review shifts its focus to inclusion complexes, elucidating how pillar[n]arenes serve as encapsulation platforms for antibiotics. This encapsulation enhances the stability of antibiotics and enables a controlled release, thereby amplifying their antibacterial activity. The examination of inclusion complexes provides valuable insights into the potential synergy between pillar[n]arenes and traditional antibiotics, offering a novel avenue for overcoming biofilm resistance. This comprehensive review highlights the escalating global threat of bacterial infections and the urgent need for innovative strategies to counteract drug-resistant biofilms. The unique properties of pillar[n]arenes, both as functionalized molecules and as inclusion complex hosts, position them as promising candidates in the quest for effective anti-biofilm agents. The exploration of their distinct mechanisms opens new avenues for research and development in the ongoing battle against bacterial infections and biofilm-related health challenges.
Collapse
Affiliation(s)
- Sekar Jothi Nayaki
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641 043, India
| | - Arivazhagan Roja
- Department of Chemistry, The Gandhigram Rural Institute (Deemed to be University), Dindigul, Tamil Nadu 624 302, India
| | - Ramya Ravindhiran
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641 043, India
| | - Karthiga Sivarajan
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641 043, India
| | - Murugan Arunachalam
- Department of Chemistry, The Gandhigram Rural Institute (Deemed to be University), Dindigul, Tamil Nadu 624 302, India
| | - Kavitha Dhandapani
- Department of Biochemistry, Biotechnology and Bioinformatics, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu 641 043, India
| |
Collapse
|
9
|
Ghosh S, Sen S, Jash M, Ghosh S, Jana A, Roy R, Mukherjee N, Mukherjee D, Sarkar J, Ghosh S. Synergistic Augmentation of Beta-Lactams: Exploring Quinoline-Derived Amphipathic Small Molecules as Antimicrobial Potentiators against Methicillin-Resistant Staphylococcus aureus. ACS Infect Dis 2024; 10:1267-1285. [PMID: 38442370 DOI: 10.1021/acsinfecdis.3c00696] [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] [Indexed: 03/07/2024]
Abstract
The escalation of bacterial resistance against existing therapeutic antimicrobials has reached a critical peak, leading to the rapid emergence of multidrug-resistant strains. Stringent pathways in novel drug discovery hinder our progress in this survival race. A promising approach to combat emerging antibiotic resistance involves enhancing conventional ineffective antimicrobials using low-toxicity small molecule adjuvants. Recent research interest lies in weak membrane-perturbing agents with unique cyclic hydrophobic components, addressing a significant gap in antimicrobial drug exploration. Our study demonstrates that quinoline-based amphipathic small molecules, SG-B-52 and SG-B-22, significantly reduce MICs of selected beta-lactam antibiotics (ampicillin and amoxicillin) against lethal methicillin-resistant Staphylococcus aureus (MRSA). Mechanistically, membrane perturbation, depolarization, and ROS generation drive cellular lysis and death. These molecules display minimal in vitro and in vivo toxicity, showcased through hemolysis assays, cell cytotoxicity analysis, and studies on albino Wistar rats. SG-B-52 exhibits impressive biofilm-clearing abilities against MRSA biofilms, proposing a strategy to enhance beta-lactam antibiosis and encouraging the development of potent antimicrobial potentiators.
Collapse
Affiliation(s)
- Surojit Ghosh
- Smart Healthcare Department, Interdisciplinary Research Platform, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Samya Sen
- iHUB Drishti Foundation, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Moumita Jash
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Satyajit Ghosh
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Aniket Jana
- Smart Healthcare Department, Interdisciplinary Research Platform, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Rajsekhar Roy
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Nabanita Mukherjee
- Smart Healthcare Department, Interdisciplinary Research Platform, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Dipro Mukherjee
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Jayita Sarkar
- Centre for Research and Development of Scientific Instruments (CRDSI), Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India
| | - Surajit Ghosh
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
- Smart Healthcare Department, Interdisciplinary Research Platform, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
- iHUB Drishti Foundation, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| |
Collapse
|
10
|
Chen S, Qin S, Li R, Qu Y, Ampomah-Wireko M, Nininahazwe L, Wang M, Gao C, Zhang E. Design, synthesis and antibacterial evaluation of low toxicity amphiphilic-cephalosporin derivatives. Eur J Med Chem 2024; 268:116293. [PMID: 38447461 DOI: 10.1016/j.ejmech.2024.116293] [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: 11/27/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Global public health is facing a serious problem as a result of the rise in antibiotic resistance and the decline in the discovery of new antibiotics. In this study, two series of amphiphilic-cephalosporins were designed and synthesized, several of which showed good antibacterial activity against both Gram-positive and Gram-negative bacteria. Structure-activity relationships indicated that the length of the hydrophobic alkyl chain significantly affects the antibacterial activity against Gram-negative bacteria. The best compound 2d showed high activity against drug-susceptible Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA) with MICs of 0.5 and 2-4 μg/mL, respectively. Furthermore, 2d remained active in complex mammalian body fluids and had a longer post-antibiotic effect (PAE) than vancomycin. Mechanism studies indicated that compound 2d lacks membrane-damaging properties and can target penicillin-binding proteins to disrupt bacterial cell wall structure, inhibit the metabolic activity and induce the accumulation of reactive oxygen species (ROS) in bacteria. Compound 2d showed minimal drug resistance and was nontoxic to HUVEC and HBZY-1 cells with CC50 > 128 μg/mL. These findings suggest that 2d is a promising drug candidate for treating bacterial infections.
Collapse
Affiliation(s)
- Shengcong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Ruirui Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Ye Qu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Lauraine Nininahazwe
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Meng Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Chen Gao
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - En Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China; Pingyuan Laboratory (Zhengzhou University), PR China.
| |
Collapse
|
11
|
Gao C, Qin S, Wang M, Li R, Ampomah-Wireko M, Chen S, Qu Y, Zhang E. Effective ciprofloxacin cationic antibacterial agent against persister bacteria with low hemolytic toxicity. Eur J Med Chem 2024; 267:116215. [PMID: 38354522 DOI: 10.1016/j.ejmech.2024.116215] [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: 01/05/2024] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/16/2024]
Abstract
With the widespread use of antibiotics, bacterial resistance has developed rapidly. To make matters worse, infections caused by persistent bacteria and biofilms often cannot be completely eliminated, which brings great difficulties to clinical medication. In this work, three series of quinolone pyridinium quaternary ammonium small molecules were designed and synthesized. Most of the compounds showed good antibacterial activity against Gram-positive bacteria (S. aureus and E. faecalis) and Gram-negative bacteria (E. coli and S. maltophilia). The activity of the para-pyridine quaternary ammonium salt was better than that of the meta-pyridine. 3f was the optimal compound with good stability in body fluids and was unlikely to induce bacterial resistance. The hemolysis rate of erythrocytes at 1280 μg/mL for 3f was only 5.1%. Encouragingly, 3f rapidly killed bacteria within 4 h at 4 × MIC concentration and was effective in killing persistent bacteria in biofilms. The antibacterial mechanism experiments showed that 3f could cause disorder of bacterial membrane potential, increase bacterial membrane permeability, dissolve and destroy the membrane. Incomplete bacterial membranes lead to leakage of bacterial genetic material, concomitant production of ROS, and bacterial death due to these multiple effects.
Collapse
Affiliation(s)
- Chen Gao
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Meng Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Ruirui Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Shengcong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Ye Qu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - En Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China; Pingyuan Laboratory (Zhengzhou University), PR China.
| |
Collapse
|
12
|
Li J, Yu Y, Zhou Y, Song J, Yang A, Wang M, Li Y, Wan M, Zhang C, Yang H, Bai Y, Wong WL, Pu H, Feng X. Multi-targeting oligopyridiniums: Rational design for biofilm dispersion and bacterial persister eradication. Bioorg Chem 2024; 144:107163. [PMID: 38306825 DOI: 10.1016/j.bioorg.2024.107163] [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: 10/19/2023] [Revised: 01/09/2024] [Accepted: 01/27/2024] [Indexed: 02/04/2024]
Abstract
The development of effective antibacterial drugs to combat bacterial infections, particularly the biofilm-related infections, remains a challenge. There are two important features of bacterial biofilms, which are well-known critical factors causing biofilms hard-to-treat in clinical, including the dense and impermeable extracellular polymeric substances (EPS) and the metabolically repressed dormant and persistent bacterial population embedded. These characteristics largely increase the difficulty for regular antibiotic treatment due to insufficient penetration into EPS. In addition, the dormant bacteria are insensitive to the growth-inhibiting mechanism of traditional antibiotics. Herein, we explore the potential of a series of new oligopyridinium-based oligomers bearing a multi-biomacromolecule targeting function as the potent bacterial biofilm eradication agent. These oligomers were rationally designed to be "charge-on-backbone" that can offer a special alternating amphiphilicity. This novel and unique feature endows high affinity to bacterial membrane lipids, DNAs as well as proteins. Such a broad multi-targeting nature of molecules not only enables its penetration into EPS, but also plays vital roles in the bactericidal mechanism of action that is highly effective against dormant and persistent bacteria. Our in vitro, ex vivo, and in vivo studies demonstrated that OPc3, one of the most effective derivatives, was able to offer excellent antibacterial potency against a variety of bacteria and effectively eliminate biofilms in zebrafish models and mouse wound biofilm infection models.
Collapse
Affiliation(s)
- Jiaqi Li
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yue Yu
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yu Zhou
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Junfeng Song
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Anming Yang
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Min Wang
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Youzhi Li
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Muyang Wan
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Chunhui Zhang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Yugang Bai
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, Hong Kong, China Hong Kong Special Administrative Region.
| | - Huangsheng Pu
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China; College of Biology, Hunan University, Changsha, Hunan 410082, China; College of Advanced Interdisciplinary Studies & Hunan Provincial Key Laboratory of Novel Nano Optoelectronic Information Materials and Devices, National University of Defense Technology, Changsha, Hunan 410073, China; Nanhu Laser Laboratory, National University of Defense Technology, Changsha 410073, China.
| | - Xinxin Feng
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| |
Collapse
|
13
|
Wang Z, Cheng Q, Lu B, Zhang P, Zhang L, Wu W, Li J, Narain R. Fabrication of antimicrobial cationic hydrogels driven by physically and chemically crosslinking for wound healing. Int J Biol Macromol 2024; 259:129213. [PMID: 38184052 DOI: 10.1016/j.ijbiomac.2024.129213] [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: 09/16/2023] [Revised: 01/01/2024] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
The wound therapy based on antibiotic delivery inevitably leads to the emergence of drug resistance. Hydrogel biomaterials with inherent antibacterial activities have emerged as promising candidates for addressing this issue. However, developing an inherently antibacterial hydrogel through simple and facile strategies to promote localized wound infection healing remains a challenge. In this study, we successfully constructed antimicrobial cationic hydrogels with self-healing and injectable properties through physically and chemically dual-crosslinked networks. The networks were formed by the copolymers poly[(di(ethylene glycol) methyl ether methacrylate)-co-(4-formylphenyl methacrylate)-co-(2-(methacryloyloxy)ethyl]trimethylammonium chloride solution)] (PDFM) and poly[(di(ethylene glycol) methyl ether methacrylate)-co-(2-aminoethyl methacrylate hydrochloride)-co-(2-(((6-(6-methyl-4[1H]pyrimidionylureido) hexyl)carbamoyl)oxy)ethyl methacrylate)] (PDAU). The hydrogel systems effectively facilitate the regeneration and healing of infected wounds through the contact bactericidal feature of quaternary ammonium cations. The presence of Schiff base bonds in the injectable hydrogels imparts remarkable pH responsiveness and self-healing properties. In vitro experiments verified their intrinsic antibacterial activities along with their favorable cytocompatibility and hemocompatibility in both in vitro and in vivo. In addition, the hydrogel significantly accelerated the healing of bacterially infected in a full-thickness skin wound. This facilely prepared dual-crosslinked hydrogel, without antibiotics loading, holds significant prospects for treating infected wounds.
Collapse
Affiliation(s)
- Zhihao Wang
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, PR China; School of Chemistry and Chemical Engineering, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Qiuli Cheng
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, PR China.
| | - Binzhong Lu
- School of Chemistry and Chemical Engineering, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Panpan Zhang
- School of Chemistry and Chemical Engineering, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Leitao Zhang
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Wenlan Wu
- School of Medicine, Henan University of Science & Technology, Luoyang 471023, PR China
| | - Junbo Li
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, PR China.
| | - Ravin Narain
- Department of Chemical and Material Engineering, University of Alberta, Edmonton, Alberta T6G 1H9, Canada
| |
Collapse
|
14
|
Li BC, Lv LX, Huang LL, Zhou JT, Huang XS, Li J, Yang RY, Xu WF. Cycloaspeptide H, a cyclopentapeptide from the endophytic fungus Penicillium virgatum. Nat Prod Res 2024; 38:486-492. [PMID: 36190784 DOI: 10.1080/14786419.2022.2128346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/17/2022] [Indexed: 10/10/2022]
Abstract
One new cyclopentapeptide, cycloaspeptide H (1), featuring a serine residue, along with seven known compounds (2-8), was isolated from the endophytic fungus Penicillium virgatum GDGJ-227. The planar structure of 1 was elucidated by a comprehensive analysis of NMR and MS spectroscopic spectra, and the absolute configuration was determined by single-crystal X-ray diffraction (Cu Kα) analysis. Compounds 7 and 8 displayed antibacterial activities against Bacillus subtilis and Enterobacter aerogenes with MIC values ranging from 12.5 to 50 μg/mL.
Collapse
Affiliation(s)
- Ben-Chao Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, P. R. China
- Guangxi Marine Environmental Monitoring Centre, Beihai, P. R. China
| | - Liu-Xia Lv
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, P. R. China
| | - Li-Li Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, P. R. China
| | - Jia-Tong Zhou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, P. R. China
| | - Xi-Shan Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, P. R. China
| | - Jun Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, P. R. China
| | - Rui-Yun Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, P. R. China
| | - Wei-Feng Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, P. R. China
| |
Collapse
|
15
|
Petit M, Tessier J, Sahli C, Schmitzer AR. Confronting the Threat: Designing Highly Effective bis-Benzimidazolium Agents to Overcome Biofilm Persistence and Antimicrobial Resistance. ACS Infect Dis 2023; 9:2202-2214. [PMID: 37882623 DOI: 10.1021/acsinfecdis.3c00289] [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] [Indexed: 10/27/2023]
Abstract
The objective of this study is to take the initial steps toward developing novel antibiotics to counteract the escalating problem of antimicrobial and bacterial persistence, particularly in relation to biofilms. Our approach involves emulating the structural characteristics of cationic antimicrobial peptides. To circumvent resistance development, we have designed a library of bis-benzimidazolium salts that selectively target the microbial membranes in a nonspecific manner. To explore their structure-activity relationship, we conducted experiments using these compounds on various pathogens known for their resistance to conventional antibiotics, including Gram-positive methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus faecium (VRE), and Gram-negative Escherichia coli (E. coli). Notably, two bis-benzimidazolium salts exhibited robust antimicrobial activity while maintaining a high level of selectivity compared with mammalian cells. Our investigations revealed significant antibiofilm activity, as these compounds rapidly acted against established biofilms. In addition, bis-benzimidazolium compounds exhibited consistent results in resistance development and cross-resistance studies. Consequently, amphiphilic bis-benzimidazolium salts hold promise as potential candidates to combat resistance-associated infections.
Collapse
Affiliation(s)
- Maude Petit
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
| | - Jérémie Tessier
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
- Collège Bois-de-Boulogne, 10555 Ave. de Bois-de-Boulogne, Montréal H4N 1L4, Canada
| | - Célia Sahli
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
- CNRS-UMR 7086, Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), Université Paris Cité, Paris 75013 , France
| | - Andreea R Schmitzer
- Département de Chimie, Faculté des Arts et des Sciences, Université de Montréal, Campus MIL, 1375, Ave. Thérèse Lavoie-Roux, Montréal, Québec H2 V 0B3, Canada
| |
Collapse
|
16
|
Ganesan N, Mishra B, Felix L, Mylonakis E. Antimicrobial Peptides and Small Molecules Targeting the Cell Membrane of Staphylococcus aureus. Microbiol Mol Biol Rev 2023; 87:e0003722. [PMID: 37129495 PMCID: PMC10304793 DOI: 10.1128/mmbr.00037-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
Clinical management of Staphylococcus aureus infections presents a challenge due to the high incidence, considerable virulence, and emergence of drug resistance mechanisms. The treatment of drug-resistant strains, such as methicillin-resistant S. aureus (MRSA), is further complicated by the development of tolerance and persistence to antimicrobial agents in clinical use. To address these challenges, membrane disruptors, that are not generally considered during drug discovery for agents against S. aureus, should be explored. The cell membrane protects S. aureus from external stresses and antimicrobial agents, but membrane-targeting antimicrobial agents are probably less likely to promote bacterial resistance. Nontypical linear cationic antimicrobial peptides (AMPs), highly modified AMPs such as daptomycin (lipopeptide), bacitracin (cyclic peptide), and gramicidin S (cyclic peptide), are currently in clinical use. Recent studies have demonstrated that AMPs and small molecules can penetrate the cell membrane of S. aureus, inhibit phospholipid biosynthesis, or block the passage of solutes between the periplasm and the exterior of the cell. In addition to their primary mechanism of action (MOA) that targets the bacterial membrane, AMPs and small molecules may also impact bacteria through secondary mechanisms such as targeting the biofilm, and downregulating virulence genes of S. aureus. In this review, we discuss the current state of research into cell membrane-targeting AMPs and small molecules and their potential mechanisms of action against drug-resistant physiological forms of S. aureus, including persister cells and biofilms.
Collapse
Affiliation(s)
- Narchonai Ganesan
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Biswajit Mishra
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Department of Medicine, The Miriam Hospital, Providence, Rhode Island, USA
| | - LewisOscar Felix
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Department of Medicine, Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
17
|
Zhou C, Zhou Y, Zheng Y, Yu Y, Yang K, Chen Z, Chen X, Wen K, Chen Y, Bai S, Song J, Wu T, Lei E, Wan M, Cai Q, Ma L, Wong WL, Bai Y, Zhang C, Feng X. Amphiphilic Nano-Swords for Direct Penetration and Eradication of Pathogenic Bacterial Biofilms. ACS APPLIED MATERIALS & INTERFACES 2023; 15:20458-20473. [PMID: 37039625 DOI: 10.1021/acsami.3c03091] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Bacterial biofilms are major causes of persistent and recurrent infections and implant failures. Biofilms are formable by most clinically important pathogens worldwide, such as Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli, causing recalcitrance to standard antibiotic therapy or anti-biofilm strategies due to amphiphilic impermeable extracellular polymeric substances (EPS) and the presence of resistant and persistent bacteria within the biofilm matrix. Herein, we report our design of an oligoamidine-based amphiphilic "nano-sword" with high structural compacity and rigidity. Its rigid, amphiphilic structure ensures effective penetration into EPS, and the membrane-DNA dual-targeting mechanism exerts strong bactericidal effect on the dormant bacterial persisters within biofilms. The potency of this oligoamidine is shown in two distinct modes of application: it may be used as a coating agent for polycaprolactone to fully inhibit surface biofilm growth in an implant-site mimicking micro-environment; meanwhile, it cures model mice of biofilm infections in various ex vivo and in vivo studies.
Collapse
Affiliation(s)
- Cailing Zhou
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yu Zhou
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yaqian Zheng
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Yu
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Kailing Yang
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Zhiyong Chen
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Xianhui Chen
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Kang Wen
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yajie Chen
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Silei Bai
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Junfeng Song
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Tong Wu
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - E Lei
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Muyang Wan
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Qingyun Cai
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Luyan Ma
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wing-Leung Wong
- 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 SAR 999077, China
| | - Yugang Bai
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Chunhui Zhang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Xinxin Feng
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
18
|
Jiang L, Ma Y, Chen Y, Cai M, Wu Z, Xiong Y, Duan X, Liao X, Wang J. Multi-target antibacterial mechanism of ruthenium polypyridine complexes with anthraquinone groups against Staphylococcus aureus. RSC Med Chem 2023; 14:700-709. [PMID: 37122548 PMCID: PMC10131643 DOI: 10.1039/d2md00430e] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/26/2023] [Indexed: 02/09/2023] Open
Abstract
Three new Ru(ii) complexes, [Ru(dtb)2PPAD](PF6)2 (Ru-1), [Ru(dmob)2PPAD](PF6)2 (Ru-2) and [Ru(bpy)2PPAD](PF6)2 (Ru-3) (dtb = 4,4'-di-tert-butyl-2,2'-bipyridine, dmob = 4,4'-dimethyl-2,2'-bipyridine, bpy = 2,2'-bipyridine and PPAD = 2-(pyridine-3-yl)-1H-imidazo[4,5f][1.10]phenanthracene-9,10-dione), were synthesized and characterized by 1H NMR and 13C NMR spectroscopy, HRMS and HPLC. Among them, Ru-1 showed excellent antimicrobial activity against Gram-positive bacteria Staphylococcus aureus (minimum inhibitory concentration (MIC) = 1 μg mL-1) and low hemolytic and cytotoxic activity. In addition, Ru-1 showed obviously rapid bactericidal activity, low resistance rate, bacterial biofilm destroying activity and high biosafety in vivo. Moreover, skin infection models and a mouse model of sepsis indicated that the anti-infective efficacy of Ru-1 was comparable to that of vancomycin. Mechanism exploration results showed that the antibacterial behavior is probably related with targeting of the bacterial cell membrane and inhibiting topoisomerase I.
Collapse
Affiliation(s)
- Li Jiang
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Yuanyuan Ma
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Yiman Chen
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Mengcheng Cai
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Zhixing Wu
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Yanshi Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Xuemin Duan
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Xiangwen Liao
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| | - Jintao Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University Nanchang China
| |
Collapse
|
19
|
Qiu H, Zhao X, Jiang Y, Liang W, Wang W, Jiang X, Jiang M, Wang X, Cui W, Li Y, Tang K, Zhang T, Zhao L, Liang H. Design and synthesis of fascaplysin derivatives as inhibitors of FtsZ with potent antibacterial activity and mechanistic study. Eur J Med Chem 2023; 254:115348. [PMID: 37060755 DOI: 10.1016/j.ejmech.2023.115348] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/20/2023] [Accepted: 04/03/2023] [Indexed: 04/17/2023]
Abstract
The increase in antibiotic resistance has made it particularly urgent to develop new antibiotics with novel antibacterial mechanisms. Inhibition of bacterial cell division by disrupting filamentous temperature-sensitive mutant Z (FtsZ) function is an effective and promising approach. A series of novel fascaplysin derivatives with tunable hydrophobicity were designed and synthesized here. The in vitro bioactivity assessment revealed that these compounds could inhibit the tested Gram-positive bacteria including methicillin-resistant S. aureus (MRSA) (MIC = 0.049-25 μg/mL), B. subtilis (MIC = 0.024-12.5 μg/mL) and S. pneumoniae (MIC = 0.049-50 μg/mL). Among them, compounds B3 (MIC = 0.098 μg/mL), B6 (MIC = 0.098 μg/mL), B8 (MIC = 0.049 μg/mL) and B16 (MIC = 0.098 μg/mL) showed the best bactericidal activities against MRSA and no significant tendency to trigger bacterial resistance as well as rapid bactericidal properties. The cell surface integrity of bacteria was significantly disrupted by hydrophobic tails of fascaplysin derivatives. Further studies revealed that these highly active amphiphilic compounds showed low hemolytic activity and cytotoxicity to mammalian cells. Preliminary mechanistic exploration suggests that B3, B6, B8 and B16 are potent FtsZ inhibitors to promote FtsZ polymerization and inhibit GTPase activity of FtsZ, leading to the death of bacterial cells by inhibiting bacterial division. Molecular docking simulations and structure-activity relationship (SAR) study reveal that appropriate increase in the hydrophobicity of fascaplysin derivatives and the addition of additional hydrogen bonds facilitated their binding to FtsZ proteins. These amphiphilic fascaplysin derivatives could serve as a novel class of FtsZ inhibitors, which not only gives new prospects for the application of compounds containing this skeleton but also provides new ideas for the discovery of new antibiotics.
Collapse
Affiliation(s)
- Hongda Qiu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Xing Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Yinli Jiang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Weida Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Weile Wang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Xingyao Jiang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Mengying Jiang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Xiao Wang
- School of Medicine, Ningbo University, Ningbo, 315211, China.
| | - Wei Cui
- School of Medicine, Ningbo University, Ningbo, 315211, China
| | - Yang Li
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, China
| | - Keqi Tang
- Institute of Mass Spectrometry, School of Materials Science & Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Tao Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, and College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Lingling Zhao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Hongze Liang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
20
|
Chen D, Cadelis MM, Rouvier F, Troia T, Edmeades LR, Fraser K, Gill ES, Bourguet-Kondracki ML, Brunel JM, Copp BR. α,ω-Diacyl-Substituted Analogues of Natural and Unnatural Polyamines: Identification of Potent Bactericides That Selectively Target Bacterial Membranes. Int J Mol Sci 2023; 24:5882. [PMID: 36982955 PMCID: PMC10052977 DOI: 10.3390/ijms24065882] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/14/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
In this study, α-ω-disubstituted polyamines exhibit a range of potentially useful biological activities, including antimicrobial and antibiotic potentiation properties. We have prepared an expanded set of diarylbis(thioureido)polyamines that vary in central polyamine core length, identifying analogues with potent methicillin-resistant Staphylococcus aureus (MRSA), Escherichia coli, Acinetobacter baumannii and Candida albicans growth inhibition properties, in addition to the ability to enhance action of doxycycline towards Gram-negative bacterium Pseudomonas aeruginosa. The observation of associated cytotoxicity/hemolytic properties prompted synthesis of an alternative series of diacylpolyamines that explored aromatic head groups of varying lipophilicity. Examples bearing terminal groups each containing two phenyl rings (15a-f, 16a-f) were found to have optimal intrinsic antimicrobial properties, with MRSA being the most susceptible organism. A lack of observed cytotoxicity or hemolytic properties for all but the longest polyamine chain variants identified these as non-toxic Gram-positive antimicrobials worthy of further study. Analogues bearing either one or three aromatic-ring-containing head groups were either generally devoid of antimicrobial properties (one ring) or cytotoxic/hemolytic (three rings), defining a rather narrow range of head group lipophilicity that affords selectivity for Gram-positive bacterial membranes versus mammalian. Analogue 15d is bactericidal and targets the Gram-positive bacterial membrane.
Collapse
Affiliation(s)
- Dan Chen
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Melissa M. Cadelis
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Florent Rouvier
- UMR MD1 “Membranes et Cibles Therapeutiques”, U1261 INSERM, Faculté de Pharmacie, Aix-Marseille Universite, 27 bd Jean Moulin, 13385 Marseille, France
| | - Thomas Troia
- UMR MD1 “Membranes et Cibles Therapeutiques”, U1261 INSERM, Faculté de Pharmacie, Aix-Marseille Universite, 27 bd Jean Moulin, 13385 Marseille, France
| | - Liam R. Edmeades
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Kyle Fraser
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Evangelene S. Gill
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Marie-Lise Bourguet-Kondracki
- Laboratoire Molécules de Communication et Adaptation des Micro-Organismes, UMR 7245 CNRS, Muséum National d’Histoire Naturelle, 57 Rue Cuvier (C.P. 54), 75005 Paris, France
| | - Jean Michel Brunel
- UMR MD1 “Membranes et Cibles Therapeutiques”, U1261 INSERM, Faculté de Pharmacie, Aix-Marseille Universite, 27 bd Jean Moulin, 13385 Marseille, France
| | - Brent R. Copp
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
21
|
Tuning the Anthranilamide Peptidomimetic Design to Selectively Target Planktonic Bacteria and Biofilm. Antibiotics (Basel) 2023; 12:antibiotics12030585. [PMID: 36978452 PMCID: PMC10044445 DOI: 10.3390/antibiotics12030585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
There is a pressing need to develop new antimicrobials to help combat the increase in antibiotic resistance that is occurring worldwide. In the current research, short amphiphilic antibacterial and antibiofilm agents were produced by tuning the hydrophobic and cationic groups of anthranilamide peptidomimetics. The attachment of a lysine cationic group at the tail position increased activity against E. coli by >16-fold (from >125 μM to 15.6 μM) and greatly reduced cytotoxicity against mammalian cells (from ≤20 μM to ≥150 μM). These compounds showed significant disruption of preformed biofilms of S. aureus at micromolar concentrations.
Collapse
|
22
|
Liu Y, Dong T, Chen Y, Sun N, Liu Q, Huang Z, Yang Y, Cheng H, Yue K. Biodegradable and Cytocompatible Hydrogel Coating with Antibacterial Activity for the Prevention of Implant-Associated Infection. ACS APPLIED MATERIALS & INTERFACES 2023; 15:11507-11519. [PMID: 36852669 DOI: 10.1021/acsami.2c20401] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Implant-associated infection (IAI) caused by pathogens colonizing on the implant surface is a serious issue in the trauma-orthopedic surgery, which often leads to implant failure. The complications of IAI bring a big threat to the clinical practice of implants, accompanied by significant economic cost and long hospitalization time. In this study, we propose an antibiotics-free strategy to address IAI-related challenges by using a biodegradable and cytocompatible hydrogel coating. To achieve this, a novel hydrogel system was developed to combine the synergistic effects of good cell affinity and antibacterial properties. The hydrogel material was prepared by modifying a photocross-linkable gelatin-based polymer (GelMA) with cationic quaternary ammonium salt (QAS) groups via a mild and simple synthesis procedure. By engineering the length of the hydrophobic carbon chain on the QAS group and the degree of functionalization, the resulting GelMA-octylQAS hydrogel exhibited an integration of good mechanical properties, biodegradability, excellent bactericidal activity against various types of bacteria, and high cytocompatibility with mammalian cells. When coated onto the implant via the in situ cross-linking procedure, our hydrogel demonstrated superior antimicrobial ability in the infective model of femoral fracture of rats. Our results suggest that the GelMA-octylQAS hydrogel might provide a promising platform for preventing and treating IAI.
Collapse
Affiliation(s)
- Yanhui Liu
- College of Textiles & Clothing, State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Ting Dong
- College of Textiles & Clothing, State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Yuhang Chen
- Department of Orthopedic Surgery, The First People's Hospital of Foshan, Foshan, Guangdong 528000, China
- Department of Orthopaedic Surgery, Division of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Na Sun
- College of Textiles & Clothing, State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Qi Liu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
| | - Zhenkai Huang
- School of Materials Science and Hydrogen Energy, Foshan University, Foshan 528000, China
| | - Yafeng Yang
- Department of Orthopedics, the Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
| | - Hao Cheng
- Department of Orthopaedic Surgery, Division of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Kan Yue
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
23
|
Li S, Wang M, Chen S, Ampomah-Wireko M, Gao C, Xia Z, Nininahazwe L, Qin S, Zhang E. Development of biaromatic core-linked antimicrobial peptide mimics: Substituent position significantly affects antibacterial activity and hemolytic toxicity. Eur J Med Chem 2023; 247:115029. [PMID: 36549113 DOI: 10.1016/j.ejmech.2022.115029] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/04/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
The development of bacterial resistance to the majority of clinically significant antimicrobials has made it more difficult to treat bacterial infections with conventional antibiotics. As part of ongoing research on antimicrobial peptide mimetics, a series of quaternary ammonium cationic compounds with various linkers were designed and synthesized, with some demonstrating high antibacterial activity against Gram-negative and Gram-positive bacteria. The structure-activity relationship study revealed that the spatial position of substituents had a significant impact on antibacterial activity and hemolytic toxicity. The best compound, 3e, has good antibacterial activity against Staphylococcus aureus [minimum inhibitory concentration (MIC = 1 μg/mL)] and the least hemolytic toxicity [hemolytic concentration (HC50 = 905 μg/mL)], is stable in mammalian body fluids, and rarely induces bacterial resistance. The mechanism study revealed that the membrane action mode may be its potential bactericidal mechanism, and it can effectively cause the accumulation of intracellular reactive oxygen species (ROS) for killing bacteria. Importantly, 3e can effectively reduce the load of methicillin-resistant Staphylococcus aureus (MRSA) in mouse skin and has a higher in vivo bactericidal efficiency than vancomycin. These findings highlight the significance of divergent linkers in quaternary ammonium cations as antimicrobial peptide mimics and the potential of these cations to treat bacterial infections.
Collapse
Affiliation(s)
- Sen Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Meng Wang
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Shengcong Chen
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Chen Gao
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Ziwei Xia
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Lauraine Nininahazwe
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, 450001, PR China.
| | - En Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery and Development, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University, Zhengzhou, 450001, PR China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou, 450001, PR China.
| |
Collapse
|
24
|
Shen BY, Wang MM, Xu SM, Gao C, Wang M, Li S, Ampomah-Wireko M, Chen SC, Yan DC, Qin S, Zhang E. Antibacterial efficacy evaluation and mechanism probe of small lysine chalcone peptide mimics. Eur J Med Chem 2022; 244:114885. [DOI: 10.1016/j.ejmech.2022.114885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/15/2022] [Accepted: 10/24/2022] [Indexed: 11/04/2022]
|
25
|
Dan W, Gao J, Qi X, Wang J, Dai J. Antibacterial quaternary ammonium agents: Chemical diversity and biological mechanism. Eur J Med Chem 2022; 243:114765. [PMID: 36116235 DOI: 10.1016/j.ejmech.2022.114765] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 01/04/2023]
Abstract
Bacterial infections have seriously threatened public health especially with the increasing resistance and the cliff-like decline of the number of newly approved antibacterial agents. Quaternary ammonium compounds (QACs) possess potent medicinal properties with 95 successfully marketed drugs, which also have a long history as antibacterial agents. In this review, we summarize the chemical diversity of antibacterial QACs, divided into chain-like and aromatic ring, reported over the past decade (2012 to mid-2022). Additionally, the structure-activity relationships, mainly covering hydrophobicity, charges and skeleton features, are discussed. In the cases where sufficient information is available, antibacterial mechanisms including biofilm, cell membrane, and intracellular targets are presented. It is hoped that this review will provide sufficient information for medicinal chemists to discover the new generation of antibacterial agents based on QACs.
Collapse
Affiliation(s)
- Wenjia Dan
- School of Life Science and Technology, Weifang Medical University, Shandong, China
| | - Jixiang Gao
- School of Life Science and Technology, Weifang Medical University, Shandong, China
| | - Xiaohui Qi
- School of Life Science and Technology, Weifang Medical University, Shandong, China
| | - Junru Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Shaanxi, China.
| | - Jiangkun Dai
- School of Life Science and Technology, Weifang Medical University, Shandong, China.
| |
Collapse
|
26
|
Kong Q, Li G, Zhang F, Yu T, Chen X, Jiang Q, Wang Y. N-Arylimidazoliums as Highly Selective Biomimetic Antimicrobial Agents. J Med Chem 2022; 65:11309-11321. [PMID: 35930690 DOI: 10.1021/acs.jmedchem.2c00818] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Antibiotic resistance has become one of the greatest health threats in the world. In this study, a charge-dispersed dimerization strategy is described for the antimicrobial peptide (AMP) mimics via a tunable cationic charge to improve the selectivity between prokaryotic microbes and eukaryotic cells. This strategy is demonstrated with a series of charge-dispersed AMP mimics based on N-arylimidazolium skeletons. These N-arylimidazolium AMP mimics show potent antibacterial activity against strains along with a low rate of drug resistance, good hemocompatibility, and low cytotoxicity. In addition to the elimination of planktonic bacteria, N-arylimidazolium AMP mimics can also inhibit biofilm formation and destroy the established biofilm. More importantly, methicillin-resistant Staphylococcus aureus (MRSA)-induced lung-infected mice can be effectively treated by the intravenous administration of N-arylimidazolium AMP mimic, which enable the design of N-arylimidazolium AMP mimics to offer an alternative avenue to eradicate drug-resistant bacterial infection.
Collapse
Affiliation(s)
- Qunshou Kong
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Fanjun Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Tao Yu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Xiaotong Chen
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Qing Jiang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China
| |
Collapse
|
27
|
Do Pham DD, Mojr V, Helusová M, Mikušová G, Pohl R, Dávidová E, Šanderová H, Vítovská D, Bogdanová K, Večeřová R, Sedláková MH, Fišer R, Sudzinová P, Pospíšil J, Benada O, Křížek T, Galandáková A, Kolář M, Krásný L, Rejman D. LEGO-Lipophosphonoxins: A Novel Approach in Designing Membrane Targeting Antimicrobials. J Med Chem 2022; 65:10045-10078. [PMID: 35839126 PMCID: PMC9580004 DOI: 10.1021/acs.jmedchem.2c00684] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
The alarming rise of bacterial antibiotic resistance
requires the
development of new compounds. Such compounds, lipophosphonoxins (LPPOs),
were previously reported to be active against numerous bacterial species,
but serum albumins abolished their activity. Here we describe the
synthesis and evaluation of novel antibacterial compounds termed LEGO-LPPOs,
loosely based on LPPOs, consisting of a central linker module with
two attached connector modules on either side. The connector modules
are then decorated with polar and hydrophobic modules. We performed
an extensive structure–activity relationship study by varying
the length of the linker and hydrophobic modules. The best compounds
were active against both Gram-negative and Gram-positive species including
multiresistant strains and persisters. LEGO-LPPOs act by first depleting
the membrane potential and then creating pores in the cytoplasmic
membrane. Importantly, their efficacy is not affected by the presence
of serum albumins. Low cytotoxicity and low propensity for resistance
development demonstrate their potential for therapeutic use.
Collapse
Affiliation(s)
- Duy Dinh Do Pham
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Viktor Mojr
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Michaela Helusová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 43 Prague 2, Czech Republic
| | - Gabriela Mikušová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10 Prague 6, Czech Republic.,Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 43 Prague 2, Czech Republic
| | - Radek Pohl
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Eva Dávidová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Hana Šanderová
- Institute of Microbiology, Czech Academy of Sciences v.v.i., Vídečská 1083, 142 20 Prague 4, Czech Republic
| | - Dragana Vítovská
- Institute of Microbiology, Czech Academy of Sciences v.v.i., Vídečská 1083, 142 20 Prague 4, Czech Republic
| | - Kateřina Bogdanová
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Renata Večeřová
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Miroslava Htoutou Sedláková
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Radovan Fišer
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 43 Prague 2, Czech Republic
| | - Petra Sudzinová
- Institute of Microbiology, Czech Academy of Sciences v.v.i., Vídečská 1083, 142 20 Prague 4, Czech Republic
| | - Jiří Pospíšil
- Institute of Microbiology, Czech Academy of Sciences v.v.i., Vídečská 1083, 142 20 Prague 4, Czech Republic
| | - Oldřich Benada
- Institute of Microbiology, Czech Academy of Sciences v.v.i., Vídečská 1083, 142 20 Prague 4, Czech Republic
| | - Tomáš Křížek
- Department of Analytical Chemistry, Faculty of Science, Charles University, Albertov 6, 128 43 Prague 2, Czech Republic
| | - Adéla Galandáková
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Milan Kolář
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Libor Krásný
- Institute of Microbiology, Czech Academy of Sciences v.v.i., Vídečská 1083, 142 20 Prague 4, Czech Republic
| | - Dominik Rejman
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences v.v.i., Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| |
Collapse
|
28
|
Li SA, Cadelis MM, Deed RC, Douafer H, Bourguet-Kondracki ML, Michel Brunel J, Copp BR. Valorisation of the diterpene podocarpic acid - Antibiotic and antibiotic enhancing activities of polyamine conjugates. Bioorg Med Chem 2022; 64:116762. [PMID: 35477062 DOI: 10.1016/j.bmc.2022.116762] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/05/2022] [Accepted: 04/18/2022] [Indexed: 11/02/2022]
Abstract
As part of our search for new antimicrobials and antibiotic adjuvants, a series of podocarpic acid-polyamine conjugates have been synthesized. The library of compounds made use of the phenolic and carboxylic acid moieties of the diterpene allowing attachment of polyamines (PA) of different lengths to afford a structurally-diverse set of analogues. Evaluation of the conjugates for intrinsic antimicrobial properties identified two derivatives of interest: a PA3-4-3 (spermine) amide-bonded variant 7a that was a non-cytotoxic, non-hemolytic potent growth inhibitor of Gram-positive Staphylococcus aureus (MRSA) and 9d, a PA3-8-3 carbamate derivative that was a non-toxic selective antifungal towards Cryptococcus neoformans. Of the compound set, only one example exhibited activity towards Gram-negative bacteria. However, in the presence of sub-therapeutic amounts of either doxycycline (4.5 µM) or erythromycin (2.7 μM) several analogues were observed to exhibit weak to modest antibiotic adjuvant properties against Pseudomonas aeruginosa and/or Escherichia coli. The observation of strong cytotoxicity and/or hemolytic properties for subsets of the library, in particular those analogues bearing methyl ester or n-pentylamide functionality, highlighted the fine balance of structural requirements and lipophilicity for antimicrobial activity as opposed to mammalian cell toxicity.
Collapse
Affiliation(s)
- Steven A Li
- School of Chemical Sciences, The University of Auckland, Waipapa Taumata Rau, Private Bag 92019, Auckland 1142, New Zealand
| | - Melissa M Cadelis
- School of Chemical Sciences, The University of Auckland, Waipapa Taumata Rau, Private Bag 92019, Auckland 1142, New Zealand
| | - Rebecca C Deed
- School of Chemical Sciences, The University of Auckland, Waipapa Taumata Rau, Private Bag 92019, Auckland 1142, New Zealand; School of Biological Sciences, The University of Auckland, Waipapa Taumata Rau, Private Bag 92019, Auckland 1142, New Zealand
| | - Hana Douafer
- Aix-Marseille Universite, INSERM, SSA, MCT, Faculté de Pharmacie, 27 bd Jean Moulin, 13385 Marseille, France
| | - Marie-Lise Bourguet-Kondracki
- Laboratoire Molécules de Communication et Adaptation des Micro-organismes, UMR 7245 CNRS, Muséum National d'Histoire Naturelle, 57 rue Cuvier (C.P. 54), 75005 Paris, France
| | - Jean Michel Brunel
- Aix-Marseille Universite, INSERM, SSA, MCT, Faculté de Pharmacie, 27 bd Jean Moulin, 13385 Marseille, France
| | - Brent R Copp
- School of Chemical Sciences, The University of Auckland, Waipapa Taumata Rau, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
29
|
Zha L, Xie Y, Wu C, Lei M, Lu X, Tang W, Zhang J. Novel benzothiazole‒urea hybrids: Design, synthesis and biological activity as potent anti-bacterial agents against MRSA. Eur J Med Chem 2022; 236:114333. [PMID: 35397402 DOI: 10.1016/j.ejmech.2022.114333] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/19/2022] [Accepted: 03/28/2022] [Indexed: 11/26/2022]
Abstract
Novel benzothiazole‒urea hybrids were designed, synthesized and evaluated their anti-bacterial activity. They only exhibited anti-bacterial activity against Gram-positive bacteria, including clinical methicillin-resistant S. aureus (MRSA), compounds 5f, 5i, 8e, 8k and 8l exhibited potent activity (MIC = 0.39 and 0.39/0.78 μM against SA and MRSA, respectively). Crystal violet assay showed that compounds 5f, 8e and 8l not only inhibited the formation of biofilms but also eradicated preformed biofilms. Compound 8l had membrane disruption, little propensity to induce resistance, benign safety and in vivo anti-MRSA efficacy in a mouse model of abdominal infection. Therefore, our data demonstrated the potential to advance benzothiazole‒urea hybrids as a new class of antibiotics.
Collapse
Affiliation(s)
- Liang Zha
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Yunfeng Xie
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Chengyao Wu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Ming Lei
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Xueer Lu
- School of Medicine, Anhui University of Science and Technology, Huainan, 232001, China
| | - Wenjian Tang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| | - Jing Zhang
- Anhui Prevention and Treatment Center for Occupational Disease, Anhui No. 2 Provincial People's Hospital, Hefei, 230022, China.
| |
Collapse
|
30
|
Filatova SM, Guseva MK, Bodrova TG, Parshina DV, Budanova UA, Sebyakin YL. Evolutionary Development and Structural Diversity of Natural Antimicrobial Peptides, Peptidometics, and Cationic Amphiphiles Based on Amino Acids. RUSS J GEN CHEM+ 2022. [DOI: 10.1134/s1070363221130338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Ghosh S, Mukherjee S, Patra D, Haldar J. Polymeric Biomaterials for Prevention and Therapeutic Intervention of Microbial Infections. Biomacromolecules 2022; 23:592-608. [PMID: 35188749 DOI: 10.1021/acs.biomac.1c01528] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The escalating emergence of multidrug-resistant (MDR) pathogens and their ability to colonize into biofilms on a multitude of surfaces have struck global health as a nightmare. The stagnation in the development of antibiotics and the deterioration of clinical pipelines have incited an invigorating search for smart and innovative alternatives in the scientific community. Further, a steep rise in the usage of biomedical devices and implants has resulted in an accelerated occurrence of infections. Toward the goal of mitigation of the aforementioned challenges, antimicrobial polymers have stood out as an astounding option. In this perspective, we highlight our contribution to the field of polymeric biomaterials for tackling antimicrobial resistance (AMR) and infections. Polymers inspired from antimicrobial peptides (AMPs) have been utilized as therapeutic interventions to curb MDR infections and also to rejuvenate obsolete antibiotics. Further, cationic polymers have been used to impart antimicrobial properties to different biomedical surfaces. These cationic polymer-coated surfaces can inactivate pathogens upon contact as well as prevent their biofilm formation. In addition, antimicrobial hydrogels, which are prepared from either inherently antimicrobial polymers or biocide-loaded polymeric hydrogel matrices, have also been engineered. With a brief overview of the progress in the field, detailed elaboration of the polymeric biomaterials for prevention and therapeutic intervention of microbial infections developed by our group is presented. Finally, the challenges in the field of antimicrobial polymers with discussion on the proceedings of polymeric research to alleviate these challenges are discussed.
Collapse
|
32
|
Synergistic antimicrobial and antibiofilm activities of piperic acid and 4-ethylpiperic acid amides in combination with ciprofloxacin. J Antibiot (Tokyo) 2022; 75:236-242. [PMID: 35145264 DOI: 10.1038/s41429-022-00508-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/01/2021] [Accepted: 12/12/2021] [Indexed: 11/08/2022]
Abstract
In the present work, piperic acid and 4-ethylpiperic acid (EPA) amides with amino acids (C1-C8) were bio-evaluated for their antimicrobial activity and biofilm inhibition against Gram-positive and Gram-negative bacterial strains. Among all, EPA-β3,3-Pip(Bzl)-OMe, C2 displayed the potent antimicrobial activity with MIC of 6.25 μg ml-1 against Gram-negative bacteria Escherichia coli. In combination studies, the FIC indices suggested that C1 and C2 have a synergistic effect with ciprofloxacin against E. coli and Bacillus subtilis, whereas C5 exhibited a synergistic effect with ciprofloxacin against all the tested bacteria. The inhibitory effect of amides C1, C2, and C5 on the biofilm formation of test strains was significantly potentiated by co-administration with ciprofloxacin. Furthermore, the effective concentrations of C2 in combination reduced drastically compared to alone for biofilm inhibition. At these concentrations, C2 showed negligible hemolytic and cytotoxic activities.
Collapse
|
33
|
Jeong J, Fujita K. Selective Synthesis of Bisdimethylamine Derivatives from Diols and an Aqueous Solution of Dimethylamine through Iridium‐Catalyzed Borrowing Hydrogen Pathway. ChemCatChem 2021. [DOI: 10.1002/cctc.202101499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Jaeyoung Jeong
- Graduate School of Human and Environmental Studies Kyoto University Sakyo-ku Kyoto 606-8501 Japan
| | - Ken‐ichi Fujita
- Graduate School of Human and Environmental Studies Kyoto University Sakyo-ku Kyoto 606-8501 Japan
| |
Collapse
|
34
|
Novel ocotillol-derived lactone derivatives: design, synthesis, bioactive evaluation, SARs and preliminary antibacterial mechanism. Mol Divers 2021; 26:2103-2120. [PMID: 34661800 DOI: 10.1007/s11030-021-10318-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
A new series of ocotillol-derived lactone derivatives were designed and synthesized to consider their antibacterial activity, structure-activity relationships (SARs), antibacterial mechanism and in vivo antibacterial efficacy. Compound 6d, which exhibited broad antibacterial spectrum, was found to be the most active with minimum inhibitory concentrations (MICs) of 1-2 μg/mL against Gram-positive bacteria and 8-16 μg/mL against Gram-negative bacteria. The subsequent synergistic antibacterial tests displayed that 6d had the ability to improve the susceptibility of MRSA USA300, B. subtilis 168, and E. coli DH5α to kanamycin and chloramphenicol. This active molecule 6d also induced bacterial resistance more slowly than norfloxacin and kanamycin. Furthermore, compound 6d was membrane active and low toxic against mammalian cells, and it could rapidly inhibit the growth of MRSA and E. coli and did not obviously trigger bacterial resistance. Compound 6d also displayed strong in vivo antibacterial activity against S. aureus RN4220 in murine corneal infection models. Additionally, absorption, distribution, metabolism, and excretion properties of this type of compounds have shown drug-likeness with good oral absorption and moderate blood-brain barrier permeability. The obtained results demonstrated that ocotillol-derived compounds are a promising class of antibacterial agents worthy of further study.
Collapse
|
35
|
Gao D, Zhang Y, Bowers DT, Liu W, Ma M. Functional hydrogels for diabetic wound management. APL Bioeng 2021; 5:031503. [PMID: 34286170 PMCID: PMC8272650 DOI: 10.1063/5.0046682] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic wounds often have a slow healing process and become easily infected owing to hyperglycemia in wound beds. Once planktonic bacterial cells develop into biofilms, the diabetic wound becomes more resistant to treatment. Although it remains challenging to accelerate healing in a diabetic wound due to complex pathology, including bacterial infection, high reactive oxygen species, chronic inflammation, and impaired angiogenesis, the development of multifunctional hydrogels is a promising strategy. Multiple functions, including antibacterial, pro-angiogenesis, and overall pro-healing, are high priorities. Here, design strategies, mechanisms of action, performance, and application of functional hydrogels are systematically discussed. The unique properties of hydrogels, including bactericidal and wound healing promotive effects, are reviewed. Considering the clinical need, stimuli-responsive and multifunctional hydrogels that can accelerate diabetic wound healing are likely to form an important part of future diabetic wound management.
Collapse
Affiliation(s)
- Daqian Gao
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Yidan Zhang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Daniel T. Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Wanjun Liu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
36
|
Wei X, Li J, Zhang Y, Zheng Y, Zhang Y, Meng H, Wu G, Hu Y, Gao Y, Huang S, Wang W, Cheng Y, Wu Z, Zhang X. Synergy between Clinical Microenvironment Targeted Nanoplatform and Near-Infrared Light Irradiation for Managing Pseudomonas aeruginosa Infections. ACS APPLIED MATERIALS & INTERFACES 2021; 13:38979-38989. [PMID: 34433249 DOI: 10.1021/acsami.1c08132] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Chronic infections caused by Pseudomonas aeruginosa pose severe threats to human health. Traditional antibiotic therapy has lost its total supremacy in this battle. Here, nanoplatforms activated by the clinical microenvironment are developed to treat P. aeruginosa infection on the basis of dynamic borate ester bonds. In this design, the nanoplatforms expose targeted groups for bacterial capture after activation by an acidic infection microenvironment, resulting in directional transport delivery of the payload to bacteria. Subsequently, the production of hyperpyrexia and reactive oxygen species enhances antibacterial efficacy without systemic toxicity. Such a formulation with a diameter less than 200 nm can eliminate biofilm up to 75%, downregulate the level of cytokines, and finally promote lung repair. Collectively, the biomimetic design with phototherapy killing capability has the potential to be an alternative strategy against chronic infections caused by P. aeruginosa.
Collapse
Affiliation(s)
- Xiaosong Wei
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jie Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yin Zheng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Yanlong Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Huipeng Meng
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Guolin Wu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yuqing Hu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yingchao Gao
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Siyuan Huang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Wenbo Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yijie Cheng
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhongming Wu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
37
|
Nudelman A. Dimeric Drugs. Curr Med Chem 2021; 29:2751-2845. [PMID: 34375175 DOI: 10.2174/0929867328666210810124159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
This review intends to summarize the structures of an extensive number of symmetrical-dimeric drugs, having two monomers linked via a bridging entity while emphasizing the large versatility of biologically active substances reported to possess dimeric structures. The largest number of classes of these compounds consist of anticancer agents, antibiotics/antimicrobials, and anti-AIDS drugs. Other symmetrical-dimeric drugs include antidiabetics, antidepressants, analgesics, anti-inflammatories, drugs for the treatment of Alzheimer's disease, anticholesterolemics, estrogenics, antioxidants, enzyme inhibitors, anti-Parkisonians, laxatives, antiallergy compounds, cannabinoids, etc. Most of the articles reviewed do not compare the activity/potency of the dimers to that of their corresponding monomers. Only in limited cases, various suggestions have been made to justify unexpected higher activity of the dimers vs. the corresponding monomers. These suggestions include statistical effects, the presence of dimeric receptors, binding of a dimer to two receptors simultaneously, and others. It is virtually impossible to predict which dimers will be preferable to their respective monomers, or which linking bridges will lead to the most active compounds. It is expected that the extensive number of articles summarized, and the large variety of substances mentioned, which display various biological activities, should be of interest to many academic and industrial medicinal chemists.
Collapse
Affiliation(s)
- Abraham Nudelman
- Chemistry Department, Bar Ilan University, Ramat Gan 52900, Israel
| |
Collapse
|
38
|
Unlocking the bacterial membrane as a therapeutic target for next-generation antimicrobial amphiphiles. Mol Aspects Med 2021; 81:100999. [PMID: 34325929 DOI: 10.1016/j.mam.2021.100999] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 06/21/2021] [Accepted: 07/16/2021] [Indexed: 11/21/2022]
Abstract
Gram-positive bacteria like Enterococcus faecium and Staphylococcus aureus, and Gram-negative bacteria like Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter Spp. are responsible for most of fatal bacterial infections. Bacteria present a handful of targets like ribosome, RNA polymerase, cell wall biosynthesis, and dihydrofolate reductase. Antibiotics targeting the protein synthesis like aminoglycosides and tetracyclines, inhibitors of RNA/DNA synthesis like fluoroquinolones, inhibitors of cell wall biosynthesis like glycopeptides and β-lactams, and membrane-targeting polymyxins and lipopeptides have shown very good success in combating the bacterial infections. Ability of the bacteria to develop drug resistance is a serious public health challenge as bacteria can develop antimicrobial resistance against newly introduced antibiotics that enhances the challenge for antibiotic drug discovery. Therefore, bacterial membranes present a suitable therapeutic target for development of antimicrobials as bacteria can find it difficult to develop resistance against membrane-targeting antimicrobials. In this review, we present the recent advances in engineering of membrane-targeting antimicrobial amphiphiles that can be effective alternatives to existing antibiotics in combating bacterial infections.
Collapse
|
39
|
Wang M, Feng X, Gao R, Sang P, Pan X, Wei L, Lu C, Wu C, Cai J. Modular Design of Membrane-Active Antibiotics: From Macromolecular Antimicrobials to Small Scorpionlike Peptidomimetics. J Med Chem 2021; 64:9894-9905. [PMID: 33789422 PMCID: PMC8886609 DOI: 10.1021/acs.jmedchem.1c00312] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Infections caused by multidrug-resistant bacteria have emerged in recent decades, leading to escalating interest in host defense peptides (HDPs) to reverse this dangerous trend. Inspired by the modular design in bioengineering, herein we report a new class of small amphiphilic scorpionlike peptidomimetics based on this strategy. These HDP mimics show potent antimicrobial activity against both Gram-positive and Gram-negative bacteria without drug resistance but with a high therapeutic index. The membrane-compromising action mode was suggested to be their potential bactericidal mechanism. Pharmacodynamic experiments were conducted using a murine abscess model of methicillin-resistant Staphylococcus aureus (MRSA) infections. The lead compound 12 showed impressive in vivo therapeutic efficacy with ∼99.998% (4.7log) reduction in skin MRSA burden, a significantly higher bactericidal efficiency than ciprofloxacin, and good biocompatibility. These results highlight the potential of these HDP mimics as novel antibiotic therapeutics.
Collapse
Affiliation(s)
- Minghui Wang
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Xiaoqian Feng
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 511443, China
- Department of Pharmaceutics, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Ruixuan Gao
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Peng Sang
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Xin Pan
- Department of Pharmaceutics, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Lulu Wei
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Chao Lu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 511443, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 511443, China
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| |
Collapse
|
40
|
Gharibi R, Shaker A, Rezapour-Lactoee A, Agarwal S. Antibacterial and Biocompatible Hydrogel Dressing Based on Gelatin- and Castor-Oil-Derived Biocidal Agent. ACS Biomater Sci Eng 2021; 7:3633-3647. [PMID: 34196519 DOI: 10.1021/acsbiomaterials.1c00706] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Favored antibacterial activity associated with excellent biocompatibility, mechanical durability, and exudate handling needs to be addressed by modern dressing to achieve the desired wound healing. This paper deals with developing a new green and facile approach to manufacturing nonleachable antibacterial gelatin-based films for wound dressing. Therefore, a reactive methoxy-silane-functionalized quaternary ammonium compound bearing a fatty amide residue originating from castor oil (Si-CAQ) was initially synthesized. The antibacterial dressings were then fabricated via sol-gel and condensation reactions of the mixture containing gelatin, Si-CAQ, (3-glycidyloxypropyl) trimethoxysilane, and poly(vinyl alcohol). By utilizing bioactive polymers as starting materials and eliminating organic solvents during the dressing preparation, desirable clinical safety could be ensured. The gelatin-based films presented appropriate mechanical properties, such as flexibility and strength, in both dried and hydrated states (tensile strength >6 MPa and elongation >100). It is due to the in situ generations of the inorganic silicon domain in the organic framework via the sol-gel cross-linking process. The prepared dressings exhibited desirable features, including excellent biocompatibility (cell viability >95%), proper wound-exudate-managing characteristics (equilibrium water contact (EWA) 280-350% and water vapor transmission rate (WVTR) 2040-2200 g/m2/day), fluid handling capacity (FHC) (3-3.35 g), as well as commendable hemocompatibility. The promising bactericidal activity of the dressing against Bacillus subtilis, methicillin-resistant Staphylococcus aureus, and Escherichia coli strains with a contact-killing efficacy of 100% could prevent infection development at the wounded area. As evaluated by the wound scratch assay, the desired fibroblast cell growth, migration, and proliferation indicated the capability of the dressing to facilitate the healing process by encouraging fibroblast cell migration to the damaged area. In vivo wound-healing results showed that the prepared biocidal dressing stimulates wound healing and enhances epithelialization, collagen maturation, and vascularization of wounds due to their antibacterial effects and accelerated cellular functions.
Collapse
Affiliation(s)
- Reza Gharibi
- Macromolecular Chemistry II, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany.,Department of Organic Chemistry and Polymer, Faculty of Chemistry, Kharazmi University, 15719-14911 Tehran, Iran
| | - Ali Shaker
- Department of Organic Chemistry and Polymer, Faculty of Chemistry, Kharazmi University, 15719-14911 Tehran, Iran
| | - Alireza Rezapour-Lactoee
- Cellular and Molecular Research Center, Qom University of Medical Sciences, 3736175513 Qom, Iran
| | - Seema Agarwal
- Macromolecular Chemistry II, Bavarian Polymer Institute, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| |
Collapse
|
41
|
Pearce AN, Chen D, Edmeades LR, Cadelis MM, Troudi A, Brunel JM, Bourguet-Kondracki ML, Copp BR. Repurposing primaquine as a polyamine conjugate to become an antibiotic adjuvant. Bioorg Med Chem 2021; 38:116110. [PMID: 33831695 DOI: 10.1016/j.bmc.2021.116110] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 11/17/2022]
Abstract
In our search for new antibiotic adjuvants as a novel strategy to deal with the emergence of multi-drug resistant (MDR) bacteria, a series of succinylprimaquine-polyamine (SPQ-PA) conjugates and derivatives of a cationic amphiphilic nature have been prepared. Evaluation of these primaquine conjugates for intrinsic antimicrobial properties and the ability to restore the antibiotic activity of doxycycline identified two derivatives, SPQ-PA3-8-3 and SPQ-PA3-10-3 that exhibited intrinsic activity against the Gram-positive bacteria Staphylococcus aureus and the yeast Cryptococcus neoformans. None of the analogues were active against the Gram-negative bacterium Pseudomonas aeruginosa. However, in the presence of a sub-therapeutic amount of doxycycline (4.5 µM), both SPQ-PA3-4-3 and SPQ-PA3-10-3 compounds displayed potent antibiotic adjuvant properties against P. aeruginosa, with MIC's of 6.25 µM. A series of derivatives were prepared to investigate the structure-activity relationship that explored the influence of both a simplified aryl lipophilic substituent and variation of the length of the polyamine scaffold on observed intrinsic antimicrobial properties and the ability to potentiate the action of doxycycline against P. aeruginosa.
Collapse
Affiliation(s)
- A Norrie Pearce
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Dan Chen
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Liam R Edmeades
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Melissa M Cadelis
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Azza Troudi
- UMR_MD1, U-1261, Aix Marseille Universite, INSERM, SSA, MCT, 13385 Marseille, France
| | - Jean Michel Brunel
- UMR_MD1, U-1261, Aix Marseille Universite, INSERM, SSA, MCT, 13385 Marseille, France
| | - Marie-Lise Bourguet-Kondracki
- Laboratoire Molécules de Communication et Adaptation des Micro-organismes, UMR 7245 CNRS, Muséum National d'Histoire Naturelle, 57 rue Cuvier (C.P. 54), 75005 Paris, France
| | - Brent R Copp
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
42
|
He D, Tan Y, Li P, Luo Y, Zhu Y, Yu Y, Chen J, Ning N, Zhang S. Surface charge-convertible quaternary ammonium salt-based micelles for in vivo infection therapy. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2020.12.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
43
|
Kaizerman-Kane D, Hadar M, Joseph R, Logviniuk D, Zafrani Y, Fridman M, Cohen Y. Design Guidelines for Cationic Pillar[n]arenes that Prevent Biofilm Formation by Gram-Positive Pathogens. ACS Infect Dis 2021; 7:579-585. [PMID: 33657813 PMCID: PMC8041275 DOI: 10.1021/acsinfecdis.0c00662] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
![]()
Bacterial biofilms are a major threat
to human health, causing
persistent infections that lead to millions of fatalities worldwide
every year. Biofilms also cause billions of dollars of damage annually
by interfering with industrial processes. Recently, cationic pillararenes
were found to be potent inhibitors of biofilm formation in Gram-positive
bacteria. To identify the structural features of pillararenes that
result in antibiofilm activity, we evaluated the activity of 16 cationic
pillar[5]arene derivatives including that of the first cationic water-soluble
pillar[5]arene-based rotaxane. Twelve of the derivatives were potent
inhibitors of biofilm formation by Gram-positive pathogens. Structure
activity analyses of our pillararene derivatives indicated that positively
charged head groups are critical for the observed antibiofilm activity.
Although certain changes in the lipophilicity of the substituents
on the positively charged head groups are tolerated, dramatic elevation
in the hydrophobicity of the substituents or an increase in steric
bulk on these positive charges abolishes the antibiofilm activity.
An increase in the overall positive charge from 10 to 20 did not affect
the activity significantly, but pillararenes with 5 positive charges
and 5 long alkyl chains had reduced activity. Surprisingly, the cavity
of the pillar[n]arene is not essential for the observed activity,
although the macrocyclic structure of the pillar[n]arene core, which
facilitates the clustering of the positive charges, appears important.
Interestingly, the compounds found to be efficient inhibitors of biofilm
formation were nonhemolytic at concentrations that are ∼100-fold
of their MBIC50 (the minimal concentration of a compound
at which at least 50% inhibition of biofilm formation was observed
compared to untreated cells). The structure–activity relationship
guidelines established here pave the way for a rational design of
potent cationic pillar[n]arene-based antibiofilm agents.
Collapse
Affiliation(s)
- Dana Kaizerman-Kane
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Maya Hadar
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Roymon Joseph
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Dana Logviniuk
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Yossi Zafrani
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74000, Israel
| | - Micha Fridman
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Yoram Cohen
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| |
Collapse
|
44
|
Tague AJ, Putsathit P, Riley TV, Keller PA, Pyne SG. Positional Isomers of Biphenyl Antimicrobial Peptidomimetic Amphiphiles. ACS Med Chem Lett 2021; 12:413-419. [PMID: 33738069 PMCID: PMC7957938 DOI: 10.1021/acsmedchemlett.0c00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Small-molecule antimicrobial peptidomimetic amphiphiles represent a promising class of novel antimicrobials with the potential for widespread therapeutic application. To investigate the role of spatial positioning for key hydrophobic and hydrophilic groups on the antimicrobial efficacy and selectivity, positional isomers of the lead biphenyl antimicrobial peptidomimetic compound 1 were synthesized and subjected to microbial growth inhibition and mammalian toxicity assays. Positional isomer 4 exhibited 4-8× increased efficacy against the pathogenic Gram-negative bacteria Pseudomonas aeruginosa and Escherichia coli (MIC = 2 μg/mL), while isomers 2, 3, and 7 exhibited a 4× increase in activity against Acinetobacter baumannii (MIC = 4 μg/mL). Changes in molecular shape had a significant impact on Gram-negative antibacterial efficacy and the resultant spectrum of activity, whereas all structural isomers exhibited significant efficacy (MIC = 0.25-8 μg/mL) against Gram-positive bacterial pathogens (e.g., methicillin-resistant Staphylococcus aureus, Streptococcus pneumoniae, and Enterococcus faecalis).
Collapse
Affiliation(s)
- Andrew J. Tague
- School
of Chemistry and Molecular Bioscience, University
of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Papanin Putsathit
- School
of Medical and Health Sciences, Edith Cowan
University, Joondalup, Western Australia 6027, Australia
| | - Thomas V. Riley
- School
of Medical and Health Sciences, Edith Cowan
University, Joondalup, Western Australia 6027, Australia
- PathWest
Laboratory Medicine, Queen Elizabeth II
Medical Centre, Nedlands, Western Australia 6009, Australia
- School
of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Queen Elizabeth
II Medical Centre, Nedlands, Western Australia 6009, Australia
- Medical,
Molecular and Forensic Sciences, Murdoch
University, Murdoch, Western Australia 6150, Australia
| | - Paul A. Keller
- School
of Chemistry and Molecular Bioscience, University
of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Stephen G. Pyne
- School
of Chemistry and Molecular Bioscience, University
of Wollongong, Wollongong, New South Wales 2522, Australia
| |
Collapse
|
45
|
Simões M, Pereira AR, Simões LC, Cagide F, Borges F. Biofilm control by ionic liquids. Drug Discov Today 2021; 26:1340-1346. [PMID: 33549827 DOI: 10.1016/j.drudis.2021.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/19/2021] [Accepted: 01/29/2021] [Indexed: 10/22/2022]
Abstract
Ionic liquids (ILs) are remarkable chemical compounds with applications in many areas of modern science. They are increasingly recognized as promising compounds to fight microorganisms in both planktonic and biofilm states, contributing to reinvent the antimicrobial pipeline. Biofilm-related infections are particularly challenging given that the scientific community has not yet identified a reliable control strategy. Understanding of the action of ILs in biofilm control is is still in a very early stage. However, given the highly tunable nature and exceptional properties of ILs, they are excellent candidates for biofilm control. Here, we review the major advances in, and challenges tothe use of ILs for effective biofilm control.
Collapse
Affiliation(s)
- Manuel Simões
- LEPABE, Department of Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal.
| | - Ana Rita Pereira
- LEPABE, Department of Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Lúcia Chaves Simões
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
| | - Fernando Cagide
- CIQUP, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal.
| |
Collapse
|
46
|
Design, synthesis, antibacterial activity and toxicity of novel quaternary ammonium compounds based on pyridoxine and fatty acids. Eur J Med Chem 2020; 211:113100. [PMID: 33385851 DOI: 10.1016/j.ejmech.2020.113100] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 12/26/2022]
Abstract
A diverse series of 43 novel "soft antimicrobials" based on quaternary ammonium pyridoxine derivatives which include six-membered acetals and ketals of pyridoxine bound via cleavable linker moieties (amide, ester) with a fragment of fatty carboxylic acid was designed. Nine compounds exhibited in vitro promising antibacterial activity against Gram-positive and Gram-negative bacterial strains with MIC values comparable with reference antiseptics miramistin, benzalkonium chloride and chlorohexidine. On various clinical isolates, the lead compounds 6i and 12a exhibited antibacterial activity comparable with that of benzalkonium chloride while higher than that of miramistin. Moreover, 6i and 12a were able to kill bacteria embedded into the matrix of mono- and dual species biofilms. The treatment of bacterial cells by either 6i and 12a lead to fast depolarization of the membrane suggesting that the membrane is an apparent molecular target of compounds. 6i and 12a were non mutagenic neither in SOS-chromotest nor in Ames test and non-toxic in vivo at acute oral (LD50 > 2000 mg/kg) and cutaneous administration (LD50 > 2500 mg/kg) on mice. Taken together, our data allow suggesting described active compounds as promising starting point for the new antibacterial agents development.
Collapse
|
47
|
Carden RG, Sommers KJ, Schrank CL, Leitgeb AJ, Feliciano JA, Wuest WM, Minbiole KPC. Advancements in the Development of Non-Nitrogen-Based Amphiphilic Antiseptics to Overcome Pathogenic Bacterial Resistance. ChemMedChem 2020; 15:1974-1984. [PMID: 32886856 PMCID: PMC8371456 DOI: 10.1002/cmdc.202000612] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Indexed: 12/23/2022]
Abstract
The prevalence of quaternary ammonium compounds (QACs) as common disinfecting agents for the past century has led bacteria to develop resistance to such compounds. Given the alarming increase in resistant strains, new strategies are required to combat this rise in resistance. Recent efforts to probe and combat bacterial resistance have focused on studies of multiQACs. Relatively unexplored, however, have been changes to the primary atom bearing positive charge in these antiseptics. Here we review the current state of the field of both phosphonium and sulfonium amphiphilic antiseptics, both of which hold promise as novel means to address bacterial resistance.
Collapse
Affiliation(s)
- Robert G Carden
- Department of Chemistry, Villanova University, Villanova, PA, 19085, USA
| | - Kyle J Sommers
- Department of Chemistry, Villanova University, Villanova, PA, 19085, USA
| | | | - Austin J Leitgeb
- Department of Chemistry, Villanova University, Villanova, PA, 19085, USA
| | - Javier A Feliciano
- Department of Chemistry, Villanova University, Villanova, PA, 19085, USA
| | - William M Wuest
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Kevin P C Minbiole
- Department of Chemistry, Villanova University, Villanova, PA, 19085, USA
| |
Collapse
|
48
|
Zhong Y, Xiao H, Seidi F, Jin Y. Natural Polymer-Based Antimicrobial Hydrogels without Synthetic Antibiotics as Wound Dressings. Biomacromolecules 2020; 21:2983-3006. [PMID: 32672446 DOI: 10.1021/acs.biomac.0c00760] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Wound healing is usually accompanied by bacterial infection. The excessive use of synthetic antibiotics leads to drug resistance, posing a significant threat to human health. Hydrogel-based wound dressings aimed at mitigating bacterial infections have emerged as an effective wound treatment. The review presented herein particularly focuses on the hydrogels originating from natural polymers. To further enhance the performance of wound dressings, various strategies and approaches have been developed to endow the hydrogels with excellent broad-spectrum antibacterial activity. Those that are summarized in the current review are the hydrogels with intrinsic or stimuli-triggered bactericidal properties and others that serve as vehicles for loading antibacterial agents without synthetic antibiotics. Specific attention is paid to antimicrobial mechanisms and the antibacterial performance of hydrogels. Practical antibacterial applications to accelerate the wound healing employing these antibiotic-free hydrogels are also introduced along with the discussion on the current challenges and perspectives leading to new technologies.
Collapse
Affiliation(s)
- Yajie Zhong
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources and Joint International Research Lab of Lignocellulosic Functional Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Huining Xiao
- Department of Chemical Engineering, University of New Brunswick, Fredericton, New Brunswick E3B 5A3, Canada
| | - Farzad Seidi
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources and Joint International Research Lab of Lignocellulosic Functional Materials, Nanjing Forestry University, Nanjing 210037, China
| | - Yongcan Jin
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources and Joint International Research Lab of Lignocellulosic Functional Materials, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
49
|
Limwongyut J, Nie C, Moreland AS, Bazan GC. Molecular design of antimicrobial conjugated oligoelectrolytes with enhanced selectivity toward bacterial cells. Chem Sci 2020; 11:8138-8144. [PMID: 34123085 PMCID: PMC8163332 DOI: 10.1039/d0sc03679j] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A series of cationic conjugated oligoelectrolytes (COEs) was designed to understand how variations in molecular dimensions impact the relative activity against bacteria and mammalian cells. These COEs kept a consistent distyrylbenzene framework but differed in the length of linker between the core and the cationic site and the length of substitute on the quaternary ammonium functioned group. Their antimicrobial efficacy, mammalian cell cytotoxicity, hemolytic activity, and cell association were determined. We find that hydrophobicity is a factor that controls the degree of COE association to cells, but in vitro efficacy and cytotoxicity depend on more subtle structural features. COE2-3C-C4butyl was found to be the optimal structure with a minimum inhibitory concentration (MIC) of 4 μg mL−1 against E. coli K12, low cytotoxicity against HepG2 cells and negligible hemolysis of red blood cells, even at 1024 μg mL−1. A time-kill kinetics study of COE2-3C-C4butyl against E. coli K12 demonstrates bactericidal activity. These findings provide the first systematic investigation of how COEs may be modulated to achieve low mammalian cell cytotoxicity with the long-range perspective of finding candidates suitable for developing a broad-spectrum antimicrobial agent. A series of cationic conjugated oligoelectrolytes (COEs) was designed to understand how variations in molecular dimensions impact the relative activity against bacteria and mammalian cells.![]()
Collapse
Affiliation(s)
- Jakkarin Limwongyut
- Center for Polymers and Organic Solids, Department of Chemistry and Biochemistry, University of California Santa Barbara CA 93106 USA
| | - Chenyao Nie
- Departments of Chemistry and Chemical Engineering, National University of Singapore 117543 Singapore
| | - Alex S Moreland
- Center for Polymers and Organic Solids, Department of Chemistry and Biochemistry, University of California Santa Barbara CA 93106 USA
| | - Guillermo C Bazan
- Center for Polymers and Organic Solids, Department of Chemistry and Biochemistry, University of California Santa Barbara CA 93106 USA .,Departments of Chemistry and Chemical Engineering, National University of Singapore 117543 Singapore
| |
Collapse
|
50
|
CPF-C1 analog with effective antimicrobial and antibiofilm activities against Staphylococcus aureus including MRSA. Biochimie 2020; 176:1-11. [PMID: 32590058 DOI: 10.1016/j.biochi.2020.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 11/21/2022]
Abstract
The evolution of Staphylococcus aureus (S. aureus) with the ability to acquire and develop resistance to antibiotics has been described as a distinct strain emergence event. Methicillin-resistant S. aureus (MRSA) is responsible for most global S. aureus bacteremia cases. Bacterial biofilms are one of the primary reasons for drug resistance. Biofilms formed by S. aureus are the most common cause of biofilm-associated infections, which increase the difficulty of treatment. Antimicrobial peptides (AMPs) represent promising candidates for the future treatment of antibiotic-resistant bacterial and biofilm-associated infections. In this study, we designed and synthesized a series of analogs to increase the druggability of the natural antimicrobial peptide CPF-C1. Among the analogs, CPF-2 showed high antimicrobial activity against MRSA and multidrug-resistant S. aureus isolated from clinics. In the serum and physiological salt environment, CPF-2 also exhibited effective antimicrobial activity. Importantly, CPF-2 did not determine resistance and showed no hemolytic activity at the active concentration. Concerning the mechanism of action, CPF-2 produced a rapid bactericidal effect by interrupting the bacterial membranes. Even more surprisingly, CPF-2 showed an excellent ability to prevent and eradicate biofilms caused by S. aureus and MRSA not only in vitro but also in vivo. Our results suggested that CPF-2 has potential as a lead compound to treat infections caused by S. aureus and MRSA, including the associated biofilms.
Collapse
|