1
|
Wang S, Huang C, Gong Y, Chen J, Li L, Wang Y, Xu T, Bi W, Liu M, Sun J, Xu Y. Co-assembly of antimicrobial polypeptoids/carbon dots for internal-external cooperated sterilization. J Colloid Interface Sci 2025; 690:137292. [PMID: 40096802 DOI: 10.1016/j.jcis.2025.137292] [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: 01/19/2025] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
Bacterial infections have emerged as a significant global public health challenge that requires urgent attention. In the research of popular antimicrobial agents, antimicrobial peptide mimics with good properties have the disadvantage of high toxicity, and nanomaterials with metal-doped carbon dots as the most representative have the problems of easy agglomeration and insufficient bactericidal effect. Herein, combined therapeutic strategy was proposed to reach the best compromise and sterilization effects. We employed an electrostatic co-assembly strategy to combine nanomaterials iron-doped carbon dots (Fe-CDs) and antimicrobial polypeptoids Poly(N-allylglycine) modified with thiol-terminated amines (PNAG66-NH2), resulting in the creation of the antimicrobial composite Fe-CDs-PNAG66-NH2. Through electrostatic adsorption, the composite disrupts the electrostatic environment of the bacterial outer membrane, alters its permeability, and triggers an increase in intracellular reactive oxygen species (ROS) to rapidly kill 99.999% of Gram-negative Escherichia coli (E. coli) and Gram-positive Staphylococcus aureus (S. aureus) within 10 min. It exhibited negligible cytotoxicity to normal cells. Furthermore, in vivo experiments demonstrated that Fe-CDs-PNAG66-NH2 accelerated the healing of infected wounds, reduced inflammation. The present study demonstrates that the efficient bactericidal properties of the complexes are triggered by the synergistic action of nanomaterials and antimicrobial polypeptoids, which provides a new strategy to achieve safe and efficient broad-spectrum bactericidal activity in antimicrobial aspects.
Collapse
Affiliation(s)
- Shang Wang
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao 266071, PR China
| | - Chao Huang
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao 266071, PR China; Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, PR China
| | - Yiyu Gong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, PR China
| | - Junrong Chen
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao 266071, PR China
| | - Limin Li
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao 266071, PR China
| | - Yanjing Wang
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao 266071, PR China
| | - Tingqiang Xu
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao 266071, PR China
| | - Weilin Bi
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao 266071, PR China
| | - Miao Liu
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao 266071, PR China
| | - Jing Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin 130012, PR China.
| | - Yuanhong Xu
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao 266071, PR China.
| |
Collapse
|
2
|
Liu X, Song M, Liu Y, Yang S, Chen S, Kang J, Shen J, Zhu K. Rational Design of Natural Xanthones Against Gram-negative Bacteria. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411923. [PMID: 39888292 PMCID: PMC11984908 DOI: 10.1002/advs.202411923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/21/2024] [Indexed: 02/01/2025]
Abstract
Most antibiotics are ineffective against Gram-negative bacteria owing to the existence of the outer membrane (OM) barrier. The rational design of compounds to expand their antibacterial spectra of antibiotics solely targeting Gram-positive pathogens remains challenging. Here, the design of skeletons from natural products to penetrate the OM are deciphered. Structure-activity relationship analysis shows the optimization of the model of natural xanthones α-mangostin endows the broad-spectrum antibacterial activity. Mechanistic studies demonstrate the lead compound A20 penetrates the OM in a self-promoted pathway through electronic and hydrophobic interactions with lipopolysaccharides and phospholipids in OM. A20 displays rapid bactericidal activity by targeting the cofactor heme in the respiratory complex. The therapeutic efficacy of A20 is demonstrated in two animal models infected with multidrug-resistant Gram-negative bacterial pathogens. The findings elucidate the structural property and self-promoted transportation of a class of antibacterial compounds, to facilitate the design and discovery of antibacterial agents against increasingly prevalent Gram-negative pathogens associated with infections.
Collapse
Affiliation(s)
- Xiaojia Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Meirong Song
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Ying Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Shuyu Yang
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Shang Chen
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Jijun Kang
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural UniversityNo.2 Yuanmingyuan West RoadBeijing100193China
| |
Collapse
|
3
|
Liu Y, Zhu J, Liu Z, Zhi Y, Mei C, Wang H. Flavonoids as Promising Natural Compounds for Combating Bacterial Infections. Int J Mol Sci 2025; 26:2455. [PMID: 40141099 PMCID: PMC11942443 DOI: 10.3390/ijms26062455] [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: 10/22/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 03/28/2025] Open
Abstract
The increasing emergence and dissemination of multidrug-resistant (MDR) bacterial pathogens have intensified the need for new antibiotics and alternative therapeutic strategies. Flavonoids, a diverse group of bioactive natural compounds found in plants, have shown significant promise as antibacterial agents. Flavonoids inhibit bacterial growth through various mechanisms, including disruption of cell wall synthesis, prevention of biofilm formation, disruption of cell membrane integrity, and inhibition of bacterial efflux pumps. These actions not only reduce bacterial viability but also enhance the efficacy of conventional antibiotics, offering a potential solution to antibiotic resistance. However, challenges such as poor bioavailability limit their clinical application. Recent advances in nanotechnology-based drug delivery systems, chemical modifications, and formulation techniques have shown promise in improving flavonoid bioavailability and therapeutic efficacy. This review evaluates the antibacterial mechanisms of flavonoids, explores their potential synergistic effects with antibiotics, and highlights strategies to overcome bioavailability issues. Our findings underscore the importance of continued research on flavonoids as promising candidates for innovative antibacterial therapies aimed at combating MDR bacterial infections.
Collapse
Affiliation(s)
- Ying Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (Y.L.); (Z.L.); (Y.Z.)
| | - Jiajia Zhu
- Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China;
| | - Zhenyi Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (Y.L.); (Z.L.); (Y.Z.)
| | - Yan Zhi
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (Y.L.); (Z.L.); (Y.Z.)
| | - Chen Mei
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (Y.L.); (Z.L.); (Y.Z.)
| | - Hongjun Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (Y.L.); (Z.L.); (Y.Z.)
| |
Collapse
|
4
|
Lei R, Yang C, Zhu T, Zhu X, Zhu Z, Cui H, Pei H, Li J, Mao Y, Lan C. Multifunctional cyclic biomimetic peptides: Self-assembling nanotubes for effective treatment of sepsis. Int J Biol Macromol 2025; 288:138522. [PMID: 39672431 DOI: 10.1016/j.ijbiomac.2024.138522] [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: 01/19/2024] [Revised: 11/18/2024] [Accepted: 12/05/2024] [Indexed: 12/15/2024]
Abstract
Antibiotic abuse has led to an increasingly serious risk of antimicrobial resistance, developing alternative antimicrobials to combat this alarming issue is urgently needed. Rhesus theta defensin-1 (RTD-1) is a theta-defensin contributing to broad-spectrum bactericidal activity via the mechanisms of membrane perturbation. Intriguingly, human defensin-6 (HD6), an enteric defensin secreted by Paneth cells without direct bactericidal effect, could self-assembled into fibrous networks to trap enteric pathogens for assistance of innate immunity. The direct bactericidal action of RTD-1 and the bacterial trapping of HD6 inspire a promising antimicrobial paradigm for unique antibacterial strategies. In this study, we utilized the principle of alternating arrangement of D- and L-amino acids in cyclic peptides, which endows them with the potential to self-assemble into nanotubes, mimic the antimicrobial processes of RTD-1 and HD6. We designed and synthesized five cyclic biomimetic peptides (CBPs), among these biomimetics, CBP-4, which possessed a nanotube-like structure, demonstrated the ability to directly and rapidly disrupt the cell membranes of Gram-positive S. aureus and MRSA, while also targeting the surfaces of Gram-negative E. coil using its nanofibrous network to capture bacteria, preventing invasion and migration, and indirectly killing the bacteria. Moreover, CBP-4 eliminated pathogens, inhibited excessive inflammatory responses caused by infections, and maintained immune system homeostasis in septic mice. By fully emulating the antimicrobial mechanisms of both RTD-1 and HD6, CBP-4 showed promising potential for anti-infectious therapies.
Collapse
Affiliation(s)
- Ruyi Lei
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Chujun Yang
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Tao Zhu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xingqiang Zhu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhiqiang Zhu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongwei Cui
- Department of General ICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hui Pei
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jiye Li
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yujing Mao
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chao Lan
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
5
|
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 2025; 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] [MESH Headings] [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
|
6
|
Wei MZ, Wang ZJ, Zhu YY, Zu WB, Zhao YL, Luo XD. Oleanolic acid derivatives against drug-resistant bacteria and fungi by multi-targets to avoid drug resistance. Eur J Med Chem 2024; 280:116940. [PMID: 39388902 DOI: 10.1016/j.ejmech.2024.116940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/28/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
Mixed infections caused by drug-resistant bacteria and fungi pose a severe threat to human health, and multi-target drugs may provide an effective approach to combat drug-resistant pathogens. Therefore, this study aimed to investigate the efficacies of some oleanolic acid (OA) derivatives against multidrug-resistant (MDR) bacteria and fungi using in vitro and in vivo experiments. Novel amphiphilic OA derivatives were designed and optimised, in which compounds G1 and J1 exhibited effective antimicrobial activity (MICs = 1-2 μg/mL), high selectivity against MDR strains, rapid bactericidal activity, and good predictive pharmacokinetics. Mechanistically, both compounds prevented drug resistance by disrupting the bacterial cell membrane, inserting into the DNA, and binding to DNA gyrase. Additionally, J1 reduced microbial count in a mouse MRSA skin infection model and accelerated wound healing much better than vancomycin. Conclusively, this study presents a new class of potential drugs for resistant bacteria and fungi.
Collapse
Affiliation(s)
- Mei-Zhen Wei
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Wen-Biao Zu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, People's Republic of China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, People's Republic of China.
| |
Collapse
|
7
|
Cheng X, Zhang Y, Zhang Y, Chen Y, Chen J, Wang W, Zhu G. Multiple strategies of HSP antimicrobial peptide optimization to enhance antimicrobial activity. Amino Acids 2024; 56:66. [PMID: 39589573 PMCID: PMC11599297 DOI: 10.1007/s00726-024-03428-z] [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: 07/15/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
Antimicrobial peptides (AMPs) have caught the attention of researchers over the last couple of years due to their unique membrane lytic mechanism for combating antibiotic resistance, which differs from the molecular targets of traditional antibiotics. Although natural AMPs exhibit potential antimicrobial activity against a wide range of microorganisms, some drawbacks, such as toxicity, low antibacterial activity, and high production costs limit their clinical application. To enhance the antimicrobial activity of a series of HSP peptides derived from the natural peptide HSP-1, this study optimized them using a variety of strategies, including net charge, hydrophobic moment, hydrophobicity, and helicity. Optimizing the antimicrobial action of HSP peptides depended mostly on net charge, hydrophobic moment, and hydrophobicity rather than helicity. HSP-M4 may be designed to combat microbial infections because the antimicrobial activity and cytotoxicity assays showed that they exhibited low cytotoxicity and prominent antimicrobial activity, respectively.
Collapse
Affiliation(s)
- Xiaozhong Cheng
- Anhui Province Green Food Collaborative Technology Service Center for Rural Revitalization, Hefei Normal University, Hefei, 230601, China.
| | - Yonghuang Zhang
- Department of Pharmacy, Hefei Binhu Hospital, Hefei, 230601, China
| | - Yan Zhang
- Anhui Province Green Food Collaborative Technology Service Center for Rural Revitalization, Hefei Normal University, Hefei, 230601, China
| | - Yajun Chen
- Anhui Province Green Food Collaborative Technology Service Center for Rural Revitalization, Hefei Normal University, Hefei, 230601, China
| | - Jianli Chen
- Shimadzu (China) Co., Ltd, Wuhan, 430000, China
| | - Wei Wang
- Anhui Province Green Food Collaborative Technology Service Center for Rural Revitalization, Hefei Normal University, Hefei, 230601, China.
| | - Guilan Zhu
- Anhui Province Green Food Collaborative Technology Service Center for Rural Revitalization, Hefei Normal University, Hefei, 230601, China.
| |
Collapse
|
8
|
Wang Z, Shen W, Li Y, Wang X, Zhong X, Wang X. Multi-omics Analysis of Klebsiella pneumoniae Revealed Opposing Effects of Rutin and Luteolin on Strain Growth. Curr Microbiol 2024; 82:9. [PMID: 39585437 DOI: 10.1007/s00284-024-03982-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
The emergence of pathogenic bacteria resistant to conventional antibiotics is becoming increasingly challenging. Plant-derived flavonoids are potential alternatives to antibiotics, owing to their antimicrobial properties. However, the molecular mechanisms through which they inhibit the growth of pathogenic microorganisms remain unclear. Therefore, Klebsiella pneumoniae ATCC700603 was separately incubated in two flavonoids to elucidate their inhibitory mechanism. Metabolomic and transcriptomic analyses were performed after 4-h incubation. In total, 5483 genes and 882 metabolites were identified. Compared to the untreated control, rutin and luteolin activated 507 and 374 differentially expressed genes (DEGs), respectively. However, the number of differential abundant metabolites (DAMs) remained the same. The top 10 correlated DEGs and DAMs were identified within each comparative group after a correlation analysis. Rutin induced the accumulation of unique metabolites and suppressed gene expression whereas luteolin did not. Our results explain the disparate effects of these two flavonoids and demonstrate the inhibitory mechanism of rutin on strain growth.
Collapse
Affiliation(s)
- Zhibin Wang
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wanxia Shen
- Citrus Research Institute, Southwest University, Beibei, Chongqing, 400715, China
| | - Yuejiao Li
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaoyun Wang
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaolin Zhong
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xing Wang
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
9
|
Vrbnjak K, Sewduth RN. Recent Advances in Peptide Drug Discovery: Novel Strategies and Targeted Protein Degradation. Pharmaceutics 2024; 16:1486. [PMID: 39598608 PMCID: PMC11597556 DOI: 10.3390/pharmaceutics16111486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
Recent technological advancements, including computer-assisted drug discovery, gene-editing techniques, and high-throughput screening approaches, have greatly expanded the palette of methods for the discovery of peptides available to researchers. These emerging strategies, driven by recent advances in bioinformatics and multi-omics, have significantly improved the efficiency of peptide drug discovery when compared with traditional in vitro and in vivo methods, cutting costs and improving their reliability. An added benefit of peptide-based drugs is the ability to precisely target protein-protein interactions, which are normally a particularly challenging aspect of drug discovery. Another recent breakthrough in this field is targeted protein degradation through proteolysis-targeting chimeras. These revolutionary compounds represent a noteworthy advancement over traditional small-molecule inhibitors due to their unique mechanism of action, which allows for the degradation of specific proteins with unprecedented specificity. The inclusion of a peptide as a protein-of-interest-targeting moiety allows for improved versatility and the possibility of targeting otherwise undruggable proteins. In this review, we discuss various novel wet-lab and computational multi-omic methods for peptide drug discovery, provide an overview of therapeutic agents discovered through these cutting-edge techniques, and discuss the potential for the therapeutic delivery of peptide-based drugs.
Collapse
Affiliation(s)
- Katarina Vrbnjak
- VIB-KU Leuven Center for Cancer Biology (VIB), 3000 Leuven, Belgium
| | | |
Collapse
|
10
|
Liu F, Yang S, Zhang L, Zhang M, Bi Y, Wang S, Wang X, Wang Y. Design, synthesis and biological evaluation of amphiphilic benzopyran derivatives as potent antibacterial agents against multidrug-resistant bacteria. Eur J Med Chem 2024; 277:116784. [PMID: 39178727 DOI: 10.1016/j.ejmech.2024.116784] [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: 07/13/2024] [Revised: 08/10/2024] [Accepted: 08/17/2024] [Indexed: 08/26/2024]
Abstract
Antimicrobial resistance has emerged as a significant threat to global public health. To develop novel, high efficiency antibacterial alternatives to combat multidrug-resistant bacteria, A total of thirty-two novel amphiphilic benzopyran derivatives by mimicking the structure and function of antimicrobial peptides were designed and synthesized. Among them, the most promising compounds 4h and 17e displayed excellent antibacterial activity against Gram-positive bacteria (MICs = 1-4 μg/mL) with weak hemolytic activity and good membrane selectivity. Additionally, compounds 4h and 17e had rapid bactericidal properties, low resistance frequency, good plasma stability, and strong capabilities of inhibiting and eliminating bacterial biofilms. Mechanistic studies revealed that compounds 4h and 17e could effectively disrupt the integrity of bacterial cell membranes, and accompanied by an increase in intracellular reactive oxygen species and the leakage of proteins and DNA, ultimately leading to bacterial death. Notably, compound 4h exhibited comparable in vivo antibacterial potency in a mouse septicemia model infected by Staphylococcus aureus ATCC43300, as compared to vancomycin. These findings indicated that 4h might be a promising antibacterial candidate to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Fangquan Liu
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Siyu Yang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Lei Zhang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Meiyue Zhang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Ying Bi
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Shuo Wang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Xuekun Wang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, China.
| | - Yinhu Wang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, 252059, China.
| |
Collapse
|
11
|
Liu X, Liu Y, Song M, Zhu K, Shen J. A Rhein-Based Derivative Targets Staphylococcus aureus. Antibiotics (Basel) 2024; 13:882. [PMID: 39335055 PMCID: PMC11428220 DOI: 10.3390/antibiotics13090882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
The rise in antibiotic-resistant bacteria highlights the need for novel antimicrobial agents. This study presents the design and synthesis of a series of rhein (RH)-derived compounds with improved antimicrobial properties. The lead compound, RH17, exhibited a potent antibacterial activity against Staphylococcus aureus (S. aureus) isolates, with minimum inhibitory concentrations (MICs) ranging from 8 to 16 μg/mL. RH17 disrupted bacterial membrane stability, hindered metabolic processes, and led to an increase in reactive oxygen species (ROS) production. These mechanisms were confirmed through bacterial growth inhibition assays, membrane function assessments, and ROS detection. Notably, RH17 outperformed the parent compound RH and demonstrated bactericidal effects in S. aureus. The findings suggest that RH17 is a promising candidate for further development as an antimicrobial agent against Gram-positive pathogens, addressing the urgent need for new therapies.
Collapse
Affiliation(s)
- Xiaojia Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Yuan Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Meirong Song
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| |
Collapse
|
12
|
Yang S, Liu F, Leng Y, Zhang M, Zhang L, Wang X, Wang Y. Development of Xanthoangelol-Derived Compounds with Membrane-Disrupting Effects against Gram-Positive Bacteria. Antibiotics (Basel) 2024; 13:744. [PMID: 39200044 PMCID: PMC11350758 DOI: 10.3390/antibiotics13080744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Infections caused by multidrug-resistant pathogens have emerged as a serious threat to public health. To develop new antibacterial agents to combat such drug-resistant bacteria, a class of novel amphiphilic xanthoangelol-derived compounds were designed and synthesized by mimicking the structure and function of antimicrobial peptides (AMPs). Among them, compound 9h displayed excellent antimicrobial activity against the Gram-positive strains tested (MICs = 0.5-2 μg/mL), comparable to vancomycin, and with low hemolytic toxicity and good membrane selectivity. Additionally, compound 9h demonstrated rapid bactericidal effects, low resistance frequency, low cytotoxicity, and good plasma stability. Mechanistic studies further revealed that compound 9h had good membrane-targeting ability and was able to destroy the integrity of bacterial cell membranes, causing an increase in intracellular ROS and the leakage of DNA and proteins, thus accelerating bacterial death. These results make 9h a promising antimicrobial candidate to combat bacterial infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuekun Wang
- State Key Laboratory for Macromolecule Drugs and Large-Scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China; (S.Y.); (F.L.); (Y.L.); (M.Z.); (L.Z.)
| | - Yinhu Wang
- State Key Laboratory for Macromolecule Drugs and Large-Scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China; (S.Y.); (F.L.); (Y.L.); (M.Z.); (L.Z.)
| |
Collapse
|
13
|
Zhang Y, Luo M, Shi X, Li A, Zhou W, Yin Y, Wang H, Wong WL, Feng X, He Q. Pyrgos[ n]cages: Redefining antibacterial strategy against drug resistance. SCIENCE ADVANCES 2024; 10:eadp4872. [PMID: 39058779 PMCID: PMC11277403 DOI: 10.1126/sciadv.adp4872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Amid rising antibiotic resistance, the quest for advanced antibacterial agents to surpass microbial adaptation is paramount. This study introduces Pyrgos[n]cages (n = 1 to 4), pioneering multidecker cationic covalent organic cages engineered to combat drug-resistant bacteria via a dual-targeting approach. Synthesized through successive photocatalytic bromination and cage-forming reactions, these architectures stand out for their dense positive charge distribution, exceptional stability, and substantial rigidity. Pyrgos[n]cages exhibit potent bactericidal activity by disrupting bacterial membrane potential and binding to DNA. Notably, these structures show unparalleled success in eradicating both extracellular and intracellular drug-resistant pathogens in diverse infection scenarios, with antibacterial efficiency markedly increasing over 100-fold as the decker number rises from 1 to 3. This study provides an advance in antibacterial tactics and underscores the transformative potential of covalent organic cages in devising enduring countermeasures against antibiotic-resistant microbial threats.
Collapse
Affiliation(s)
- Yi Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Miaomiao Luo
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xiangling Shi
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Aimin Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Wei Zhou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yuyao Yin
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing 100044, China
| | - Hui Wang
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing 100044, 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
| | - Xinxin Feng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Qing He
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
14
|
Gupta S, Luxami V, Paul K. Bacterial cell death to overcome drug resistance with multitargeting bis-naphthalimides as potent antibacterial agents against Enterococcus faecalis. J Mater Chem B 2024; 12:5645-5660. [PMID: 38747306 DOI: 10.1039/d3tb02804f] [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: 06/13/2024]
Abstract
The increasing frequency of drug-resistant pathogens poses serious health issues to humans around the globe, leading to the development of new antibacterial agents to conquer drug resistance and bacterial infections. In view of this, we have synthesized a series of bis-naphthalimides to respond to awful drug resistance. Bioactivity assay and structure-activity relationship disclosed that compounds 5d and 5o exhibit potent antibacterial activity against E. faecalis, outperforming the marketed antibiotics. These drug candidates not only inhibit the biofilm formation of E. faecalis but also display rapid bactericidal properties, thus delaying the development of drug resistance within 20 passages. To explore the mechanism of antibacterial activity against E. faecalis, biofunctional examination was carried out which unveiled that 5d and 5o effectively disrupt bacterial cell membranes, causing the leakage of cytoplasmic contents and metabolic activity loss. Concurrently, 5d and 5o effectively intercalate with DNA to block DNA replication, causing the build-up of excessive reactive oxygen species and inhibiting the glutathione activity, ultimately leading to oxidative damage of E. faecalis and cell death. In addition, these compounds readily bind with HSA with a high binding constant, indicating that these drug candidates could be easily delivered to the target site. The above finding manifested that these newly synthesized bis-naphthalimides with multitargeting antibacterial properties offer a new prospect to overcome drug resistance.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India.
| | - Vijay Luxami
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India.
| | - Kamaldeep Paul
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India.
| |
Collapse
|
15
|
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
|
16
|
Cebrián R, Lucas R, Fernández-Cantos MV, Slot K, Peñalver P, Martínez-García M, Párraga-Leo A, de Paz MV, García F, Kuipers OP, Morales JC. Synthesis and antimicrobial activity of aminoalkyl resveratrol derivatives inspired by cationic peptides. J Enzyme Inhib Med Chem 2023; 38:267-281. [PMID: 36600674 PMCID: PMC9828810 DOI: 10.1080/14756366.2022.2146685] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Antimicrobial resistance is a global concern, far from being resolved. The need of new drugs against new targets is imminent. In this work, we present a family of aminoalkyl resveratrol derivatives with antibacterial activity inspired by the properties of cationic amphipathic antimicrobial peptides. Surprisingly, the newly designed molecules display modest activity against aerobically growing bacteria but show surprisingly good antimicrobial activity against anaerobic bacteria (Gram-negative and Gram-positive) suggesting specificity towards this bacterial group. Preliminary studies into the action mechanism suggest that activity takes place at the membrane level, while no cross-resistance with traditional antibiotics is observed. Actually, some good synergistic relations with existing antibiotics were found against Gram-negative pathogens. However, some cytotoxicity was observed, despite their low haemolytic activity. Our results show the importance of the balance between positively charged moieties and hydrophobicity to improve antimicrobial activity, setting the stage for the design of new drugs based on these molecules.
Collapse
Affiliation(s)
- Rubén Cebrián
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands,Department of Clinical Microbiology, Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospital Clínico San Cecilio, Granada, Spain,CONTACT Rubén Cebrián University Hospital San Cecilio,Clinical Microbiology Department, Av. de la Innovación s/n, 18061, Granada, Spain
| | - Ricardo Lucas
- Department of Organic and Pharmaceutical Chemistry, School of Pharmacy, University of Seville, Seville, Spain
| | - María Victoria Fernández-Cantos
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Koen Slot
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Pablo Peñalver
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Armilla, Granada, Spain
| | - Marta Martínez-García
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Antonio Párraga-Leo
- Department of Organic and Pharmaceutical Chemistry, School of Pharmacy, University of Seville, Seville, Spain
| | - María Violante de Paz
- Department of Organic and Pharmaceutical Chemistry, School of Pharmacy, University of Seville, Seville, Spain
| | - Federico García
- Department of Clinical Microbiology, Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospital Clínico San Cecilio, Granada, Spain
| | - Oscar P. Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands,Oscar P. Kuipers University of Groningen, Faculty of Science and Engineering, Department of Genetics, Nijenborgh 7, 9747AG, Groningen, The Netherlands
| | - Juan Carlos Morales
- Department of Biochemistry and Molecular Pharmacology, Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Armilla, Granada, Spain,Juan Carlos Morales Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avda. del Conocimiento 17, Armilla, 18016Granada, Spain
| |
Collapse
|
17
|
Yu Q, Cai Q, Liang W, Zhong K, Liu J, Li H, Chen Y, Li H, Fang S, Zhong R, Liu S, Lin S. Design of phenothiazine-based cationic amphiphilic derivatives incorporating arginine residues: Potential membrane-active broad-spectrum antimicrobials combating pathogenic bacteria in vitro and in vivo. Eur J Med Chem 2023; 260:115733. [PMID: 37643545 DOI: 10.1016/j.ejmech.2023.115733] [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: 06/13/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Multidrug-resistant bacteria infections pose an increasingly serious threat to human health, and the development of antimicrobials is far from meeting the clinical demand. It is urgent to discover and develop novel antibiotics to combat bacterial resistance. Currently, the development of membrane active antimicrobial agents is an attractive strategy to cope with antimicrobial resistance issues. In this study, the synthesis and biological evaluation of cationic amphiphilic phenothiazine-based derivatives were reported. Among them, the most promising compound 30 bearing a n-heptyl group and two arginine residues displayed potent bactericidal activity against both Gram-positive (MICs = 1.56 μg/mL) and Gram-negative bacteria (MICs = 3.125-6.25 μg/mL). Compound 30 showed low hemolysis activity (HC50 = 281.4 ± 1.6 μg/mL) and low cytotoxicity (CC50 > 50 μg/mL) toward mammalian cells, as well as excellent salt resistance. Compound 30 rapidly killed bacteria by acting on the bacterial cell membrane and appeared less prone to resistance. Importantly, compound 30 showed potent in vivo efficacy in a murine model of bacterial keratitis. Hence, the results suggested compound 30 has a promising prospect as a broad-spectrum antibacterial agent for the treatment of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Qian Yu
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qiongna Cai
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wanxin Liang
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kewen Zhong
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiayong Liu
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haizhou Li
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yongzhi Chen
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hongxia Li
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shanfang Fang
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Rongcui Zhong
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shouping Liu
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Shuimu Lin
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
18
|
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
|
19
|
Yuan J, Wang J, Li X, Zhang Y, Xian J, Wang C, Zhang J, Wu C. Amphiphilic small molecule antimicrobials: From cationic antimicrobial peptides (CAMPs) to mechanism-related, structurally-diverse antimicrobials. Eur J Med Chem 2023; 262:115896. [PMID: 39491431 DOI: 10.1016/j.ejmech.2023.115896] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 11/05/2024]
Abstract
Bacterial infections are characterized by their rapid and widespread proliferation, leading to significant morbidity. Despite the availability of a variety of antimicrobial drugs, the resistance exhibited by pathogenic microorganisms towards these drugs demonstrates a consistent upward trajectory year after year. This trend can be attributed to the abuse or misuse of antibiotics. Although antimicrobial peptides can avoid the emergence of drug resistance to a certain extent, their clinical application has been hindered by factors such as their high production cost, poor in vivo stability, and potential cytotoxicity. Consequently, there arises an urgent need for the development of novel antimicrobial drugs. Small-molecule amphiphatic antimicrobials have a good prospect for research and development. These peptides hold the potential to address several issues, including the high cost of antimicrobial peptide production, poor in vivo stability, and cytotoxicity. Moreover, they exhibit the capability to overcome bacterial resistance, thereby considerably satisfying market demands and clinical needs. This paper reviews recent research pertaining to small molecule host-defending amphiphatic antimicrobials with cationic amphiphilic structures. It focuses on the design concepts, inherent relationships, drug-like properties, antimicrobial activities, application prospects, and emerging screening methods for novel antimicrobial. This review assumes paramount importance in mitigating the current shortcomings of antimicrobial agents. It also provides potential new ideas and methodologies for the research and development of antimicrobial agents.
Collapse
Affiliation(s)
- Jiani Yuan
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Xiaoxue Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ya Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jinghong Xian
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Chengyong Wu
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
20
|
Wang R, Zhou X, Chen J, Chen Y, Xiong Y, Duan X, Liao X, Wang J. Ruthenium polypyridine complexes containing prenyl groups as antibacterial agents against Staphylococcus aureus through a membrane-disruption mechanism. Arch Pharm (Weinheim) 2023; 356:e2300175. [PMID: 37421212 DOI: 10.1002/ardp.202300175] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 07/10/2023]
Abstract
Four new ruthenium polypyridyl complexes with prenyl groups, [Ru(bpy)2 (MHIP)](PF6 )2 (Ru(II)-1), [Ru(dtb)2 (MHIP)](PF6 )2 (Ru(II)-2), [Ru(dmb)2 (MHIP)](PF6 )2 (Ru(II)-3), and [Ru(dmob)2 (MHIP)](PF6 )2 (Ru(II)-4) (bpy = 2,2'-bipyridine, dtb = 4,4'-di-tert-butyl-2,2'-bipyridine, dmb = 4,4'-dimethyl-2,2'-bipyridine, dmob = 4,4'-dimethoxy-2,2'-bipyridine, and MHIP = 2-(2,6-dimethylhepta-1,5-dien-1-yl)-1H-imidazo[4,f][1,10]phenanthroline), were synthesized and characterized. Their antibacterial activities against Staphylococcus aureus were assessed, and the minimum inhibition concentration (MIC) value of Ru(II)-2 against S. aureus was only 0.5 µg/mL, showing the best antibacterial activity among them. S. aureus could be quickly killed by Ru(II)-2 in 30 min and Ru(II)-2 displayed an obvious inhibitive effect on the formation of a biofilm, which was essential to avoid the development of drug-resistance. Meanwhile, Ru(II)-2 exhibited a stable MIC value against antibiotic-resistant bacteria. The antibacterial mechanism of Ru(II)-2 was probably related to depolarization of the cell membrane, and a change of permeability was associated with the formation of reactive oxygen species, leading to leakage of nucleic acid and bacterial death. Furthermore, Ru(II)-2 hardly showed toxicity to mammalian cells and the Galleria mellonella worm. Finally, murine infection studies also illustrated that Ru(II)-2 was highly effective against S. aureus in vivo.
Collapse
Affiliation(s)
- Runbin Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Xiaomin Zhou
- Shenzhen Second People's Hospital, Shenzhen, China
| | - Jingjing Chen
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Yushou Chen
- 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
|
21
|
Li G, Lv K, Cheng Q, Xing H, Xue W, Zhang W, Lin Q, Ma D. Enhanced Bacterial-Infected Wound Healing by Nitric Oxide-Releasing Topological Supramolecular Nanocarriers with Self-Optimized Cooperative Multi-Point Anchoring. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206959. [PMID: 36793143 PMCID: PMC10104656 DOI: 10.1002/advs.202206959] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Polymeric systems that provide cationic charges or biocide-release therapeutics are used to treat the bacteria-infected wound. However, most antibacterial polymers based on topologies with restricted molecular dynamics still do not satisfy the clinical requirements due to their limited antibacterial efficacy at safe concentrations in vivo. Here a NO-releasing topological supramolecular nanocarrier with rotatable and slidable molecular entities is reported to provide conformational freedom to promote the interactions between the carrier and the pathogenic microbes, hence greatly improving the antibacterial performance. With improved contacting-killing and efficient delivery of NO biocide from the molecularly dynamic cationic ligand design, the NO-loaded topological nanocarrier achieves excellent antibacterial and anti-biofilm effects via destroying the bacterial membrane and DNA. MRSA-infected rat model is also brought out to demonstrate its wound-healing effect with neglectable toxicity in vivo. Introducing flexible molecular motions into therapeutic polymeric systems is a general design to enhance the healing of a range of diseases.
Collapse
Affiliation(s)
- Guowei Li
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan UniversityGuangzhou510630China
- Key Laboratory of Biomaterials of Guangdong Higher Education InstitutesDepartment of Biomedical EngineeringJinan UniversityGuangzhou510632China
| | - Kai Lv
- Key Laboratory of Biomaterials of Guangdong Higher Education InstitutesDepartment of Biomedical EngineeringJinan UniversityGuangzhou510632China
| | - Qikun Cheng
- Key Laboratory of Biomaterials of Guangdong Higher Education InstitutesDepartment of Biomedical EngineeringJinan UniversityGuangzhou510632China
| | - Hui Xing
- Key Laboratory of Biomaterials of Guangdong Higher Education InstitutesDepartment of Biomedical EngineeringJinan UniversityGuangzhou510632China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education InstitutesDepartment of Biomedical EngineeringJinan UniversityGuangzhou510632China
| | - Wu Zhang
- The First Affiliated Hospital of Jinan UniversityJinan UniversityGuangzhou510630China
- School of Stomatology of Jinan UniversityJinan UniversityGuangzhou510632China
| | - Qianming Lin
- School of Biomedical EngineeringSun Yat‐sen University, Shenzhen CampusShenzhen518107China
- School of Biomedical EngineeringSun Yat‐sen UniversityGuangzhou510006China
| | - Dong Ma
- Key Laboratory of Biomaterials of Guangdong Higher Education InstitutesDepartment of Biomedical EngineeringJinan UniversityGuangzhou510632China
- MOE Key Laboratory of Tumor Molecular BiologyJinan UniversityGuangzhou510632China
| |
Collapse
|
22
|
Zeng C, Avula SR, Meng J, Zhou C. Synthesis and Biological Evaluation of Piperazine Hybridized Coumarin Indolylcyanoenones with Antibacterial Potential. Molecules 2023; 28:molecules28062511. [PMID: 36985486 PMCID: PMC10056909 DOI: 10.3390/molecules28062511] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
A class of piperazine hybridized coumarin indolylcyanoenones was exploited as new structural antibacterial frameworks to combat intractable bacterial resistance. Bioactive assessment discovered that 4-chlorobenzyl derivative 11f showed a prominent inhibition on Pseudomonas aeruginosa ATCC 27853 with a low MIC of 1 μg/mL, which was four-fold more effective than norfloxacin. Importantly, the highly active 11f with inconspicuous hemolysis towards human red blood cells displayed quite low proneness to trigger bacterial resistance. Preliminary explorations on its antibacterial behavior disclosed that 11f possessed the ability to destroy bacterial cell membrane, leading to increased permeability of inner and outer membranes, the depolarization and fracture of membrane, and the effusion of intracellular components. Furthermore, bacterial oxidative stress and metabolic turbulence aroused by 11f also accelerated bacterial apoptosis. In particular, 11f could not only effectively inset into DNA, but also bind with DNA gyrase through forming supramolecular complex, thereby affecting the biological function of DNA. The above findings of new piperazine hybridized coumarin indolylcyanoenones provided an inspired possibility for the treatment of resistant bacterial infections.
Collapse
Affiliation(s)
- Chunmei Zeng
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Srinivasa Rao Avula
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Jiangping Meng
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, College of Pharmacy, Chongqing University of Arts and Sciences, Chongqing 402160, China
- Correspondence: (J.M.); (C.Z.)
| | - Chenghe Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
- Correspondence: (J.M.); (C.Z.)
| |
Collapse
|
23
|
Zhai X, Wu G, Tao X, Yang S, Lv L, Zhu Y, Dong D, Xiang H. Success stories of natural product-derived compounds from plants as multidrug resistance modulators in microorganisms. RSC Adv 2023; 13:7798-7817. [PMID: 36909750 PMCID: PMC9994607 DOI: 10.1039/d3ra00184a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/01/2023] [Indexed: 03/14/2023] Open
Abstract
Microorganisms evolve resistance to antibiotics as a function of evolution. Antibiotics have accelerated bacterial resistance through mutations and acquired resistance through a combination of factors. In some cases, multiple antibiotic-resistant determinants are encoded in these genes, immediately making the recipient organism a "superbug". Current antimicrobials are no longer effective against infections caused by pathogens that have developed antimicrobial resistance (AMR), and the problem has become a crisis. Microorganisms that acquire resistance to chemotherapy (multidrug resistance) are a major obstacle for successful treatments. Pharmaceutical industries should be highly interested in natural product-derived compounds, as they offer new sources of chemical entities for the development of new drugs. Phytochemical research and recent experimental advances are discussed in this review in relation to the antimicrobial efficacy of selected natural product-derived compounds as well as details of synergistic mechanisms and structures. The present review recognizesand amplifies the importance of compounds with natural origins, which can be used to create safer and more effective antimicrobial drugs by combating microorganisms that are resistant to multiple types of drugs.
Collapse
Affiliation(s)
- Xiaohan Zhai
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University Dalian China
| | - Guoyu Wu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University Dalian China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University Dalian China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University Dalian China
| | - Linlin Lv
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University Dalian China
| | - Yanna Zhu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University Dalian China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University Dalian China
| | - Hong Xiang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University Dalian China
| |
Collapse
|
24
|
Lu H, Cheng Z, Hu Y, Tang LV. What Can De Novo Protein Design Bring to the Treatment of Hematological Disorders? BIOLOGY 2023; 12:166. [PMID: 36829445 PMCID: PMC9952452 DOI: 10.3390/biology12020166] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Protein therapeutics have been widely used to treat hematological disorders. With the advent of de novo protein design, protein therapeutics are not limited to ameliorating natural proteins but also produce novel protein sequences, folds, and functions with shapes and functions customized to bind to the therapeutic targets. De novo protein techniques have been widely used biomedically to design novel diagnostic and therapeutic drugs, novel vaccines, and novel biological materials. In addition, de novo protein design has provided new options for treating hematological disorders. Scientists have designed protein switches called Colocalization-dependent Latching Orthogonal Cage-Key pRoteins (Co-LOCKR) that perform computations on the surface of cells. De novo designed molecules exhibit a better capacity than the currently available tyrosine kinase inhibitors in chronic myeloid leukemia therapy. De novo designed protein neoleukin-2/15 enhances chimeric antigen receptor T-cell activity. This new technique has great biomedical potential, especially in exploring new treatment methods for hematological disorders. This review discusses the development of de novo protein design and its biological applications, with emphasis on the treatment of hematological disorders.
Collapse
Affiliation(s)
| | | | | | - Liang V. Tang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
25
|
Cai Q, Yu Q, Liang W, Li H, Liu J, Li H, Chen Y, Fang S, Zhong R, Liu S, Lin S. Membrane-Active Nonivamide Derivatives as Effective Broad-Spectrum Antimicrobials: Rational Design, Synthesis, and Biological Evaluation. J Med Chem 2022; 65:16754-16773. [PMID: 36510819 DOI: 10.1021/acs.jmedchem.2c01604] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Antibiotic resistance is emerging as a "global public health concern". To address the growing epidemic of multidrug-resistant pathogens, the development of novel antimicrobials is urgently needed. In this study, by biomimicking cationic antibacterial peptides, we designed and synthesized a series of new membrane-active nonivamide and capsaicin derivatives as peptidomimetic antimicrobials. Through modulating charge/hydrophobicity balance and rationalizing structure-activity relationships of these peptidomimetics, compound 51 was identified as the lead compound. Compound 51 exhibited potent antibacterial activity against both Gram-positive bacteria (MICs = 0.39-0.78 μg/mL) and Gram-negative bacteria (MICs = 1.56-6.25 μg/mL), with low hemolytic activity and low cytotoxicity. Compound 51 displayed a faster bactericidal action through a membrane-disruptive mechanism and avoided bacterial resistance development. Furthermore, compound 51 significantly reduced the microbial burden in a murine model of keratitis infected by Staphylococcus aureus or Pseudomonas aeruginosa. Hence, this design strategy can provide a promising and effective solution to overcome antibiotic resistance.
Collapse
Affiliation(s)
- Qiongna Cai
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Qian Yu
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Wanxin Liang
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Haizhou Li
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiayong Liu
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Hongxia Li
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yongzhi Chen
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shanfang Fang
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Rongcui Zhong
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shouping Liu
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shuimu Lin
- The Fifth Affiliated Hospital & Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
26
|
Zhang P, Tangadanchu VKR, Zhou C. Identification of Novel Antifungal Skeleton of Hydroxyethyl Naphthalimides with Synergistic Potential for Chemical and Dynamic Treatments. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238453. [PMID: 36500547 PMCID: PMC9739515 DOI: 10.3390/molecules27238453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
The invasion of pathogenic fungi poses nonnegligible threats to the human health and agricultural industry. This work exploited a family of hydroxyethyl naphthalimides as novel antifungal species with synergistic potential of chemical and dynamic treatment to combat the fungal resistance. These prepared naphthalimides showed better antifungal potency than fluconazole towards some tested fungi including Aspergillus fumigatus, Candida tropicalis and Candida parapsilosis 22019. Especially, thioether benzimidazole derivative 7f with excellent anti-Candida tropicalis efficacy (MIC = 4 μg/mL) possessed low cytotoxicity, safe hemolysis level and less susceptibility to induce resistance. Biochemical interactions displayed that 7f could form a supramolecular complex with DNA to block DNA replication, and constitute a biosupermolecule with cytochrome P450 reductase (CPR) from Candida tropicalis to hinder CPR biological function. Additionally, 7f presented strong lipase affinity, which facilitated its permeation into cell membrane. Moreover, 7f with dynamic antifungal potency promoted the production and accumulation of reactive oxygen species (ROS) in cells, which destroyed the antioxidant defence system, led to oxidative stress with lipid peroxidation, loss of glutathione, membrane dysfunction and metabolic inactivation, and eventually caused cell death. The chemical and dynamic antifungal synergistic effect initiated by hydroxyethyl naphthalimides was a reasonable treatment window for prospective development.
Collapse
Affiliation(s)
- Pengli Zhang
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Drug Discovery and Development Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Vijai Kumar Reddy Tangadanchu
- Department of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- Correspondence: (V.K.R.T.); (C.Z.)
| | - Chenghe Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
- Correspondence: (V.K.R.T.); (C.Z.)
| |
Collapse
|
27
|
Zhang PL, Laiche MH, Li YL, Gao WW, Lin JM, Zhou CH. An unanticipated discovery of novel naphthalimidopropanediols as potential broad-spectrum antibacterial members. Eur J Med Chem 2022; 241:114657. [PMID: 35964427 DOI: 10.1016/j.ejmech.2022.114657] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 12/14/2022]
Abstract
Constructing a new antibacterial structural framework is an effective strategy to combat drug resistance. This work discovered a class of naphthalimidopropanediols (NIOLs) as a novel structural type of potential broad-spectrum antibacterial agents. Especially, NIOLs 9u, 12i, 15 against Staphylococcus aureus and NIOLs 9l, 13a against Pseudomonas aeruginosa showed excellent inhibitory activities, and they displayed high membrane selectivity from an electrostatic distinction on the membranes between bacteria and mammalian cells. These highly active NIOLs could effectually inhibit the bacterial growths, and relieve the resistance developments. Moreover, the facts of membrane depolarization, outer/inner membrane permeabilization and leakage of intracellular materials, demonstrated that these NIOLs could target and destroy the S. aureus or P. aeruginosa membranes. In particular, they could disrupt the antioxidant defense systems of S. aureus or P. aeruginosa through up-regulation of reactive oxygen species. Simultaneously, they could render the metabolic inactivation of the tested strains, and eradicate the formed biofilms and efficiently kill the strains within the biofilms. The in vitro and in vivo cytotoxicity assay indicated that these compounds possessed low toxicity. These findings of novel NIOLs as potential broad-spectrum antibacterial members provided a bright hope for conquering drug resistance.
Collapse
Affiliation(s)
- Peng-Li Zhang
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China
| | - Mouna Hind Laiche
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China
| | - Yan-Liang Li
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China
| | - Wei-Wei Gao
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China.
| | - Jian-Mei Lin
- Department of Infections, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Cheng-He Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
28
|
Liang W, Yu Q, Zheng Z, Liu J, Cai Q, Liu S, Lin S. Design and Synthesis of Phenyl Sulfide-Based Cationic Amphiphiles as Membrane-Targeting Antimicrobial Agents against Gram-Positive Pathogens. J Med Chem 2022; 65:14221-14236. [PMID: 36256884 DOI: 10.1021/acs.jmedchem.2c01437] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Due to the emergence of antimicrobial resistance and the lack of new antibacterial agents, it has become urgent to discover and develop new antibacterial agents against multidrug-resistant pathogens. Antimicrobial peptides (AMPs) serve as the first line of defense for the host. In this work, we have designed, synthesized, and biologically evaluated a series of phenyl sulfide derivatives by biomimicking the structural features and biological functions of AMPs. Among these derivatives, the most promising compound 17 exhibited potent antibacterial activity against Gram-positive bacteria (minimum inhibitory concentrations = 0.39-1.56 μg/mL), low hemolytic activity (HC50 > 200 μg/mL), and high membrane selectivity. In addition, 17 can rapidly kill Gram-positive bacteria within 0.5 h through membrane-targeting action and avoid antibiotic resistance. More importantly, 17 showed high in vivo efficacy against Staphylococcus aureus in a murine corneal infection model. Therefore, 17 has great potential as a lead compound for the treatment of Gram-positive bacterial infections.
Collapse
Affiliation(s)
- Wanxin Liang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Qian Yu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Zixian Zheng
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiayong Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Qiongna Cai
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Shouping Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Shuimu Lin
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
29
|
Liu J, Li H, He Q, Chen K, Chen Y, Zhong R, Li H, Fang S, Liu S, Lin S. Design, synthesis, and biological evaluation of tetrahydroquinoline amphiphiles as membrane-targeting antimicrobials against pathogenic bacteria and fungi. Eur J Med Chem 2022; 243:114734. [PMID: 36088756 DOI: 10.1016/j.ejmech.2022.114734] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/15/2022]
Abstract
The rising prevalence of drug-resistant pathogens is one of the biggest threats to human health. The development of new antibiotics that can overcome drug resistance is in urgent need. Herein, we designed and synthesized a series of amphiphilic tetrahydroquinoline derivatives as small-molecule-based antimicrobial peptidomimetics. Two lead compounds 36 and 52 which contained the tetrahydroquinoline core, hydrophobic alkyl chains (n-nonyl or isoprenyl group), different spacer lengths (n = 4 or 8), and cationic guanidine moiety, showed poor hemolytic activity, low cytotoxicity, and potent broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria, as well as fungi. The further biological evaluation revealed that compounds 36 and 52 can kill bacteria and fungi rapidly via membrane-targeting action and avoid drug resistance development. More importantly, compounds 36 and 52 exhibited similarly potent in vivo antimicrobial activities in a murine corneal infection caused by Staphylococcus aureus ATCC29213 or Pseudomonas aeruginosa ATCC9027, as compared to vancomycin or gatifloxacin. These results suggest that compounds 36 and 52 have great potential as new broad-spectrum antimicrobial agents to combat microbial resistance.
Collapse
Affiliation(s)
- Jiayong Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hongxia Li
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Qile He
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kaiting Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yongzhi Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Rongcui Zhong
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haizhou Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shanfang Fang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shouping Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Shuimu Lin
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
30
|
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: 2.3] [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
|
31
|
Aloe emodin-conjugated sulfonyl hydrazones as novel type of antibacterial modulators against S. aureus 25923 through multifaceted synergistic effects. Bioorg Chem 2022; 127:106035. [PMID: 35870413 DOI: 10.1016/j.bioorg.2022.106035] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 12/24/2022]
Abstract
Aloe emodin-conjugated sulfonyl hydrazones were designed and synthesized as novel type of antibacterial modulators. Aloe emodin benzenesulfonyl hydrazone 5a (AEBH-5a) was preponderant for the treatment of S. aureus 25923 (MIC = 0.5 μg/mL) over norfloxacin and presented high selectivity between bacterial membranes and mammalian membranes. Especially, AEBH-5a could eliminate the formed biofilms and relieve the development of S. aureus 25923 resistance. The antibacterial mechanism of AEBH-5a from extracellularity to intracellularity illustrated that AEBH-5a could destroy bacterial membrane integrity, leading to the leakage of protein and nucleic acid. Besides, AEBH-5a could not only interact with DNA and induce oxidative stress but also inhibit lactate dehydrogenase (LDH) activity as well as render metabolic inactivation. In silico ADME studies prediction of AEBH-5a revealed a favorable bioavailability score and prominent drug-likeness profile. This research showed that the multifaceted synergistic effect initiated by aloe emodin-conjugated sulfonyl hydrazones is a reasonable and effective tactic to combat menacing bacterial infections.
Collapse
|
32
|
Liu T, Zhang Y, Liu J, Peng J, Jia X, Xiao Y, Zheng L, Dong Y. Evaluation of the Acute and Sub-Acute Oral Toxicity of Jaranol in Kunming Mice. Front Pharmacol 2022; 13:903232. [PMID: 35847023 PMCID: PMC9280858 DOI: 10.3389/fphar.2022.903232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Jaranol has shown a wide range of pharmacological activities; however, no study has yet examined in vivo toxicity. The study aimed to investigate the oral acute and sub-acute toxicity of jaranol in mice. Methods: The acute toxicity was determined by a single oral dose of jaranol (2000 mg/kg). Therein animal behaviour and mortality rate were observed for 14 days. The jaranol (50, 100 and 200 mg/kg BW·d−1) was given by gavage for 28 days daily in the sub-acute study. The mouse body weight (BW), organ weight, food, water intake, biochemical, haematological parameters, and histopathology were studied in acute and sub-acute toxicity. Results: During the acute toxicity test, a single oral dose (2000 mg/kg) jaranol did not cause significant alteration in majority of the hematological indices. However, jaranol decreased the level of serum alanine aminotransferase and aspartate aminotransferase. Those results showed that the oral lethal dose 50 (LD50) of jaranol was higher than 2000 mg/kg BW, regardless of sex. In repeated daily oral doses (50, 100 and 200 mg/kg BW·d−1), no mortality was recorded in the various experimental groups. The jaranol reduced body weight gain (200 mg/kg BW·d−1), the relative spleen weight (all doses) and serum alanine aminotransferase activity (200 mg/kg BW·d−1). On the other hand, jaranol significantly elevated red blood cell count (100 and 200 mg/kg BW·d−1) and serum creatinine levels (200 mg/kg BW·d−1). Histological study revealed that spleen bleeding was identified in 200 mg/kg jaranol-treated mice. Conclusion: Jaranol was relatively safe in Kunming Mice when repetitively administered orally in small doses for a prolonged period of time. We recommend more chronic toxicity studies and clinical trials on jaranol to ensure that its use is free of potential toxicity to humans.
Collapse
Affiliation(s)
- Tianlong Liu
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yao Zhang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Jing Liu
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Junwen Peng
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xin Jia
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
- Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Hohhot, China
| | - Yunfeng Xiao
- Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Hohhot, China
- Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, China
| | - Lanbing Zheng
- Department of Psychiatry, Inner Mongolia Mental Health Center, Hohhot, China
- *Correspondence: Yu Dong, ; Lanbing Zheng,
| | - Yu Dong
- Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
- Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Hohhot, China
- *Correspondence: Yu Dong, ; Lanbing Zheng,
| |
Collapse
|
33
|
Svenson J, Molchanova N, Schroeder CI. Antimicrobial Peptide Mimics for Clinical Use: Does Size Matter? Front Immunol 2022; 13:915368. [PMID: 35720375 PMCID: PMC9204644 DOI: 10.3389/fimmu.2022.915368] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
The search for efficient antimicrobial therapies that can alleviate suffering caused by infections from resistant bacteria is more urgent than ever before. Infections caused by multi-resistant pathogens represent a significant and increasing burden to healthcare and society and researcher are investigating new classes of bioactive compounds to slow down this development. Antimicrobial peptides from the innate immune system represent one promising class that offers a potential solution to the antibiotic resistance problem due to their mode of action on the microbial membranes. However, challenges associated with pharmacokinetics, bioavailability and off-target toxicity are slowing down the advancement and use of innate defensive peptides. Improving the therapeutic properties of these peptides is a strategy for reducing the clinical limitations and synthetic mimics of antimicrobial peptides are emerging as a promising class of molecules for a variety of antimicrobial applications. These compounds can be made significantly shorter while maintaining, or even improving antimicrobial properties, and several downsized synthetic mimics are now in clinical development for a range of infectious diseases. A variety of strategies can be employed to prepare these small compounds and this review describes the different compounds developed to date by adhering to a minimum pharmacophore based on an amphiphilic balance between cationic charge and hydrophobicity. These compounds can be made as small as dipeptides, circumventing the need for large compounds with elaborate three-dimensional structures to generate simplified and potent antimicrobial mimics for a range of medical applications. This review highlight key and recent development in the field of small antimicrobial peptide mimics as a promising class of antimicrobials, illustrating just how small you can go.
Collapse
Affiliation(s)
| | - Natalia Molchanova
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Christina I. Schroeder
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| |
Collapse
|
34
|
Abstract
We report the development of peptidomimetic antibiotics derived from a natural antimicrobial peptide, human α-defensin 5. By engaging multiple bacterial targets, the lead compound is efficacious in vitro and in vivo against bacteria with highly inducible antibiotic resistance, promising a useful therapeutic agent for the treatment of infections caused by antibiotic-resistant bacteria. Antibiotics with multiple mechanisms of action and broad-spectrum are urgently required to combat the growing health threat posed by resistant pathogenic microorganisms. Combining computational and medicinal chemistry tools, we used the structure of human α-defensin 5 (HD5) to design a class of peptidomimetic antibiotics with improved activity against both gram-negative and gram-positive bacteria. The most promising lead, compound 10, showed potent killing of multiple drug-resistant gram-negative bacteria isolated from patients. Compound 10 exhibited a multiplex mechanism of action through targeting membrane components—outer membrane protein A and lipopolysaccharide, as well as a potential intracellular target—70S ribosome, thus causing membrane perturbation and inhibition of protein synthesis. In vivo efficacy, stability, and safety of compound 10 were also validated. This human defensin-inspired synthetic peptidomimetic could help solve the serious problem of drug resistance to conventional antibiotics.
Collapse
|
35
|
Liu X, Shen J, Zhu K. Antibacterial activities of plant-derived xanthones. RSC Med Chem 2022; 13:107-116. [PMID: 35308024 PMCID: PMC8864485 DOI: 10.1039/d1md00351h] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/20/2021] [Indexed: 07/26/2023] Open
Abstract
The increasing threat to global health posed by antibiotic resistance remains a serious concern. This troublesome scenario has steered a need for the discovery and evaluation of novel antibacterial agents. Natural products are the main sources of antimicrobials used in clinical practice, serving as a rich reservoir for the discovery of new antibiotics. Pharmaceutical phenolics especially xanthones widely exist in the plant kingdom, and are important plant metabolites. They possess versatile biological activities, including antiviral, antibacterial, neurotrophic, and anticancer. In the present study, we focus on the antibacterial activities of phytoxanthones and summarize their structures and sources, categories and drug-likeness evaluations, and antibacterial activities. A total of 226 different plant xanthones are identified through the NETs screening, and most of them are distributed in Clusiaceae family. These phytoxanthones are divided into four groups according to the intrinsic structural properties, including the most common simple xanthones and the majority of biprenylated ones. Moreover, their physicochemical parameters are calculated and the structure-activity relationships are discussed as well. These results indicate that the biprenylated xanthone derivatives may be promising antibacterial candidates and that the natural products of plants may be a poorly understood repository for the discovery of novel antibacterial agents.
Collapse
Affiliation(s)
- Xiaojia Liu
- National Center for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, China Agricultural University Beijing 00193 China
| | - Jianzhong Shen
- National Center for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, China Agricultural University Beijing 00193 China
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety and Beijing Laboratory for Food Quality and Safety, China Agricultural University Beijing 100193 China
| | - Kui Zhu
- National Center for Veterinary Drug Safety Evaluation, College of Veterinary Medicine, China Agricultural University Beijing 00193 China
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety and Beijing Laboratory for Food Quality and Safety, China Agricultural University Beijing 100193 China
| |
Collapse
|
36
|
Luong HX, Ngan HD, Thi Phuong HB, Quoc TN, Tung TT. Multiple roles of ribosomal antimicrobial peptides in tackling global antimicrobial resistance. ROYAL SOCIETY OPEN SCIENCE 2022; 9:211583. [PMID: 35116161 PMCID: PMC8790363 DOI: 10.1098/rsos.211583] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/20/2021] [Indexed: 05/03/2023]
Abstract
In the last century, conventional antibiotics have played a significant role in global healthcare. Antibiotics support the body in controlling bacterial infection and simultaneously increase the tendency of drug resistance. Consequently, there is a severe concern regarding the regression of the antibiotic era. Despite the use of antibiotics, host defence systems are vital in fighting infectious diseases. In fact, the expression of ribosomal antimicrobial peptides (AMPs) has been crucial in the evolution of innate host defences and has been irreplaceable to date. Therefore, this valuable source is considered to have great potential in tackling the antimicrobial resistance (AMR) crisis. Furthermore, the possibility of bacterial resistance to AMPs has been intensively investigated. Here, we summarize all aspects related to the multiple applications of ribosomal AMPs and their derivatives in combating AMR.
Collapse
Affiliation(s)
- Huy Xuan Luong
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam
- PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi 12116, Vietnam
| | | | | | - Thang Nguyen Quoc
- Nuclear Medicine Unit, Vinmec Healthcare System, Hanoi 10000, Vietnam
| | - Truong Thanh Tung
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam
- PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi 12116, Vietnam
| |
Collapse
|
37
|
Panjla A, Kaul G, Chopra S, Titz A, Verma S. Short Peptides and Their Mimetics as Potent Antibacterial Agents and Antibiotic Adjuvants. ACS Chem Biol 2021; 16:2731-2745. [PMID: 34779605 DOI: 10.1021/acschembio.1c00626] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Antimicrobial resistance (AMR) has been increasing unrelentingly worldwide, thus negatively impacting human health. The discovery and development of novel antibiotics is an urgent unmet need of the hour. However, it has become more challenging, requiring increasingly time-consuming efforts with increased commercial risks. Hence, alternative strategies are urgently needed to potentiate the existing antibiotics. In this context, short cationic peptides or peptide-based antimicrobials that mimic the activity of naturally occurring antimicrobial peptides (AMPs) could overcome the disadvantages of AMPs having evolved as potent antibacterial agents. Besides their potent antibacterial efficacy, short peptide conjugates have also gained attention as potent adjuvants to conventional antibiotics. Such peptide antibiotic combinations have become an increasingly cost-effective therapeutic option to tackle AMR. This Review summarizes the recent progress for peptide-based small molecules as promising antimicrobials and as adjuvants for conventional antibiotics to counter multidrug resistant (MDR) pathogens.
Collapse
Affiliation(s)
- Apurva Panjla
- Department of Chemistry, IIT Kanpur, Kanpur-208016, Uttar Pradesh, India
| | - Grace Kaul
- Department of Microbiology, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sidharth Chopra
- Department of Microbiology, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124, Standort Hannover-Braunschweig, Germany
| | - Sandeep Verma
- Department of Chemistry, IIT Kanpur, Kanpur-208016, Uttar Pradesh, India
- Center for Nanoscience, IIT Kanpur, Kanpur-208016, Uttar Pradesh, India
| |
Collapse
|
38
|
Rezende SB, Oshiro KGN, Júnior NGO, Franco OL, Cardoso MH. Advances on chemically modified antimicrobial peptides for generating peptide antibiotics. Chem Commun (Camb) 2021; 57:11578-11590. [PMID: 34652348 DOI: 10.1039/d1cc03793e] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Antimicrobial peptides (AMPs) are pinpointed as promising molecules against antibiotic-resistant bacterial infections. Nevertheless, there is a discrepancy between the AMP sequences generated and the tangible outcomes in clinical trials. AMPs' limitations include enzymatic degradation, chemical/physical instability and toxicity toward healthy human cells. These factors compromise AMPs' bioavailability, resulting in limited therapeutic potential. To overcome such obstacles, peptidomimetic approaches, including glycosylation, PEGylation, lipidation, cyclization, grafting, D-amino acid insertion, stapling and dendrimers are promising strategies to fine-tune AMPs. Here we focused on chemical modifications applied for AMP optimization and how they have helped these peptide-based antibiotic candidates' design and translational potential.
Collapse
Affiliation(s)
- Samilla B Rezende
- S-Inova Biotech, Universidade Católica Dom Bosco (UCDB), Campo Grande, MS, Brazil
| | - Karen G N Oshiro
- S-Inova Biotech, Universidade Católica Dom Bosco (UCDB), Campo Grande, MS, Brazil.,Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília (UnB), Brasília, DF, Brazil
| | - Nelson G O Júnior
- Centro de Análises Proteômicas e Bioquímicas Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília (UCB), Brasília, DF, Brazil.
| | - Octávio L Franco
- S-Inova Biotech, Universidade Católica Dom Bosco (UCDB), Campo Grande, MS, Brazil.,Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília (UnB), Brasília, DF, Brazil.,Centro de Análises Proteômicas e Bioquímicas Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília (UCB), Brasília, DF, Brazil.
| | - Marlon H Cardoso
- S-Inova Biotech, Universidade Católica Dom Bosco (UCDB), Campo Grande, MS, Brazil.,Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília (UnB), Brasília, DF, Brazil.,Centro de Análises Proteômicas e Bioquímicas Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília (UCB), Brasília, DF, Brazil.
| |
Collapse
|
39
|
Host Defense Peptides: Dual Antimicrobial and Immunomodulatory Action. Int J Mol Sci 2021; 22:ijms222011172. [PMID: 34681833 PMCID: PMC8538224 DOI: 10.3390/ijms222011172] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022] Open
Abstract
The rapid rise of multidrug-resistant (MDR) bacteria has once again caused bacterial infections to become a global health concern. Antimicrobial peptides (AMPs), also known as host defense peptides (HDPs), offer a viable solution to these pathogens due to their diverse mechanisms of actions, which include direct killing as well as immunomodulatory properties (e.g., anti-inflammatory activity). HDPs may hence provide a more robust treatment of bacterial infections. In this review, the advent of and the mechanisms that lead to antibiotic resistance will be described. HDP mechanisms of antibacterial and immunomodulatory action will be presented, with specific examples of how the HDP aurein 2.2 and a few of its derivatives, namely peptide 73 and cG4L73, function. Finally, resistance that may arise from a broader use of HDPs in a clinical setting and methods to improve biocompatibility will be briefly discussed.
Collapse
|
40
|
Zhong R, Li H, Li H, Fang S, Liu J, Chen Y, Liu S, Lin S. Development of Amphiphilic Coumarin Derivatives as Membrane-Active Antimicrobial Agents with Potent In Vivo Efficacy against Gram-Positive Pathogenic Bacteria. ACS Infect Dis 2021; 7:2864-2875. [PMID: 34505771 DOI: 10.1021/acsinfecdis.1c00246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Increases in drug-resistant pathogens are becoming a serious detriment to human health. To combat pathogen infections, a new series of amphiphilic coumarin derivatives were designed and synthesized as antimicrobial agents with membrane-targeting action. We herein report a lead compound, 25, that displayed potent antibacterial activity against Gram-positive bacteria, including MRSA. Compound 25 exhibited weak hemolytic activity and low toxicity to mammalian cells and can kill Gram-positive bacteria quickly (within 0.5 h) by directly disrupting the bacterial cell membranes. Additionally, compound 25 demonstrated excellent efficacy in a murine corneal infection caused by Staphylococcus aureus. These results suggest that 25 has great potential to be a potent antimicrobial agent for treating drug-resistant Gram-positive bacterial infections.
Collapse
Affiliation(s)
- Rongcui Zhong
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Haizhou Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Hongxia Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Shanfang Fang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Jiayong Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yongzhi Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Shouping Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Shuimu Lin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| |
Collapse
|
41
|
Song M, Liu Y, Li T, Liu X, Hao Z, Ding S, Panichayupakaranant P, Zhu K, Shen J. Plant Natural Flavonoids Against Multidrug Resistant Pathogens. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100749. [PMID: 34041861 PMCID: PMC8336499 DOI: 10.1002/advs.202100749] [Citation(s) in RCA: 206] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/10/2021] [Indexed: 02/05/2023]
Abstract
The increasing emergence and dissemination of multidrug resistant (MDR) bacterial pathogens accelerate the desires for new antibiotics. Natural products dominate the preferred chemical scaffolds for the discovery of antibacterial agents. Here, the potential of natural flavonoids from plants against MDR bacteria, is demonstrated. Structure-activity relationship analysis shows the prenylation modulates the activity of flavonoids and obtains two compounds, α-mangostin (AMG) and isobavachalcone (IBC). AMG and IBC not only display rapid bactericidal activity against Gram-positive bacteria, but also restore the susceptibility of colistin against Gram-negative pathogens. Mechanistic studies generally show such compounds bind to the phospholipids of bacterial membrane, and result in the dissipation of proton motive force and metabolic perturbations, through distinctive modes of action. The efficacy of AMG and IBC in four models associated with infection or contamination, is demonstrated. These results suggest that natural products of plants may be a promising and underappreciated reservoir to circumvent the existing antibiotic resistance.
Collapse
Affiliation(s)
- Meirong Song
- National Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Ying Liu
- National Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Tingting Li
- National Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Xiaojia Liu
- National Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Zhihui Hao
- Center of Research and Innovation of Chinese Traditional Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Shuangyang Ding
- Beijing Key Laboratory of Detection Technology for Animal‐Derived Food Safety and Beijing Laboratory for Food Quality and SafetyChina Agricultural UniversityBeijing100193China
| | - Pharkphoom Panichayupakaranant
- Department of Pharmacognosy and Pharmaceutical BotanyFaculty of Pharmaceutical SciencesPrince of Songkla UniversityHat‐Yai90112Thailand
| | - Kui Zhu
- National Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Center of Research and Innovation of Chinese Traditional Veterinary MedicineChina Agricultural UniversityBeijing100193China
| | - Jianzhong Shen
- National Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural UniversityBeijing100193China
- Beijing Key Laboratory of Detection Technology for Animal‐Derived Food Safety and Beijing Laboratory for Food Quality and SafetyChina Agricultural UniversityBeijing100193China
| |
Collapse
|
42
|
Tu W, Xue K, Lou S, Zhu C, Yu Z. Self-assembly of virulent amyloid-derived peptides into nanoantibacterials. NANOSCALE 2021; 13:9864-9872. [PMID: 34037034 DOI: 10.1039/d1nr01622a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Current strategies for the design of antibacterial peptides show limitations in the development of assembled antibacterial peptides due to the challenges in simultaneously balancing the antibacterial activity and assembling behavior. Herein, we report on one strategy for the design of antibacterial peptides derived from virulent amyloids and investigate their self-assembly into nanostructures with remarkable antibacterial activity. The peptides were either directly truncated from virulent amyloid peptide PSM α3 or mutated from the original sequence by replacing the lysine and phenylalanine residues with arginine or tryptophan, leading to three undecapeptides. Conformational and morphological results indicated the formation of nanotubes and twisted nanoribbons by the truncated peptide and the mutated peptide, respectively, predominately driven by anti-parallel β-sheets. Bacterial culturing experiments revealed that the two mutated peptides possessed remarkable antibacterial activity against both Gram-positive and Gram-negative bacteria by disrupting the bacterial membrane at a concentration above their critical aggregation concentrations, thus leading to two nanoantibacterials. Our findings demonstrate that biomimetic peptides originated from virulent amyloids exhibit great potential in the development of assembled antibacterial peptides, thus providing a new strategy for simultaneously addressing the antibacterial activity and pharmacokinetics of natural antibacterial peptides in the future.
Collapse
Affiliation(s)
- Wenlu Tu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Weijin Road 94, Tianjin 300071, China.
| | | | | | | | | |
Collapse
|
43
|
Shi J, Chen C, Wang D, Tong Z, Wang Z, Liu Y. Amphipathic Peptide Antibiotics with Potent Activity against Multidrug-Resistant Pathogens. Pharmaceutics 2021; 13:438. [PMID: 33804947 PMCID: PMC8063935 DOI: 10.3390/pharmaceutics13040438] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/15/2021] [Accepted: 03/20/2021] [Indexed: 11/16/2022] Open
Abstract
The emergence and prevalence of multidrug-resistant (MDR) bacteria have posed a serious threat to public health. Of particular concern are methicillin-resistant Staphylococcus aureus (MRSA) and blaNDM, mcr-1 and tet(X)-positive Gram-negative pathogens. The fact that few new antibiotics have been approved in recent years exacerbates this global crisis, thus, new alternatives are urgently needed. Antimicrobial peptides (AMPs) originated from host defense peptides with a wide range of sources and multiple functions, are less prone to achieve resistance. All these characteristics laid the foundation for AMPs to become potential antibiotic candidates. In this study, we revealed that peptide WW307 displayed potent antibacterial and bactericidal activity against MDR bacteria, including MRSA and Gram-negative bacteria carrying blaNDM-5, mcr-1 or tet(X4). In addition, WW307 exhibited great biofilm inhibition and eradication activity. Safety and stability experiments showed that WW307 had a strong resistance against various physiological conditions and displayed relatively low toxicity. Mechanistic experiments showed that WW307 resulted in membrane damage by selectively targeting bacterial membrane-specific components, including lipopolysaccharide (LPS), phosphatidylglycerol (PG), and cardiolipin (CL). Moreover, WW307 dissipated membrane potential and triggered the production of reactive oxygen species (ROS). Collectively, these results demonstrated that WW307 represents a promising candidate for combating MDR pathogens.
Collapse
Affiliation(s)
- Jingru Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.S.); (C.C.); (D.W.); (Z.T.)
| | - Chen Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.S.); (C.C.); (D.W.); (Z.T.)
| | - Dejuan Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.S.); (C.C.); (D.W.); (Z.T.)
| | - Ziwen Tong
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.S.); (C.C.); (D.W.); (Z.T.)
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.S.); (C.C.); (D.W.); (Z.T.)
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.S.); (C.C.); (D.W.); (Z.T.)
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|