1
|
Zhu X, Ren M, Zhang Z, Meng F, Li Z, Qin Y, Fang Y, Zhang M. Isolation and characterization of quinoa antimicrobial peptides and its effect on the microbial diversity of fresh apple juice. Food Chem 2025; 469:142536. [PMID: 39729667 DOI: 10.1016/j.foodchem.2024.142536] [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: 08/24/2024] [Revised: 11/30/2024] [Accepted: 12/14/2024] [Indexed: 12/29/2024]
Abstract
This study developed antimicrobial peptides (AMPs) from quinoa with high antibacterial activity and stability by mixed-bacteria fermentation. Furthermore, among 9 peptide fractions purified by membrane separation and chromatography, F1 could effectively inhibit the growth and propagation of bacterial microorganisms in apple juice. Subsequently, F1 identified LC-MS/MS as 95 peptides, molecular weights 494.25 Da to 1253.55 Da, notably, AGAAPE peptide (556.25 Da), negatively charged (-1), highly hydrophobic (50 %), with significant inhibitory effects on both Escherichia coli and Staphylococcus aureus (MIC 5 mg/mL). The antimicrobial mechanism of AGAAPE was determined to damage membrane through hydrogen-bond and hydrophobic interactions, resulting in leakage of intramembrane substances and inhibition of intracellular ATPase activity. Moreover, AGAAPE was pH resistant (pH 4-12), thermally stable (121 °C, 30 min), resistant to salt ion interference (Na+, Ca2+), and protease hydrolysis resistant (neutral protease, pepsin, trypsin). Overall, identifying AMPs from quinoa provides a promising new approach for fresh juice preservation.
Collapse
Affiliation(s)
- Xiaoyu Zhu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Mengyao Ren
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhiwei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Fanxing Meng
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zongda Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Yanan Qin
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Yan Fang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China.
| | - Minwei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China.
| |
Collapse
|
2
|
Wu R, Wu Y, Wu P, Li H, She P. Bactericidal and anti-quorum sensing activity of repurposing drug Visomitin against Staphylococcus aureus. Virulence 2024; 15:2415952. [PMID: 39390774 PMCID: PMC11492638 DOI: 10.1080/21505594.2024.2415952] [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: 05/28/2024] [Revised: 09/03/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
With the growing antibiotic resistance in Staphylococcus aureus, it is imperative to develop innovative therapeutic strategies against new targets to reduce selective survival pressures and incidence of resistance. In S. aureus, interbacterial communication relies on a quorum sensing system that regulates gene expression and physiological activities. Here, we identified that Visomitin, an antioxidant small molecule, exhibited bactericidal efficacy against methicillin-resistant S. aureus and its high tolerance phenotypes like intracellular bacteria and persister cells without inducing resistance. Critically, sub-minimal inhibitory concentrations (sub-MICs) of Visomitin could serve as a potent quorum-quencher reducing virulence production (such as haemolysin and staphyloxanthin), along with inhibiting biofilm formation, self-aggregation, and colony spreading of S. aureus. These effects were probably mediated by interfering with the S. aureus accessory gene regulator quorum sensing system. In summary, our findings suggest that Visomitin shows dual antimicrobial effects, including bactericidal effects at the concentrations above MIC and quorum sensing inhibition effects at sub-MICs, which holds promise for treating MRSA-related refractory infections.
Collapse
Affiliation(s)
- Ruolan Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yuan Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Pingyun Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Huilong Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Pengfei She
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
3
|
Qi Y, Shi Q, Ma L, Xu L, Deng Y, Zhou C. Affinity of cefditoren for penicillin-binding proteins in bacteria and its relationship with antibiotic sensitivity. Arch Microbiol 2024; 206:469. [PMID: 39556131 DOI: 10.1007/s00203-024-04194-y] [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: 08/06/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
Penicillin-binding proteins (PBPs) are the targets of β-lactam antibiotics; however, changes in the affinity of PBPs for beta-lactam antibiotics often affect the susceptibility of bacteria to antibiotics. The purpose of this study was to elucidate the mechanism by which cefditoren, an oral third-generation cephalosporin, binds PBPs. The minimal inhibitory concentration (MIC), bactericidal curves, and inhibition zone comparisons were assessed to evaluate the antibacterial activity of cefditoren. PBP1A and PBP2X proteins from Streptococcus pneumoniae were purified, and their ability to bind to cefditoren was investigated via microscale thermophoresis. The Kd of cefditoren toward PBP1A was 0.005 ± 0.004 µM, which was lower than those of other cephalosporins (cefcapene, cefixime and cefdinir). In contrast, the Kd of cefditoren toward PBP2X of S. pneumoniae was 9.70 ± 8.24 µM, which was lower than that of cefixime but higher than those of cefcapene and cefdinir. Additionally, the biotinylated ampicillin (BIO-AMP) method was employed to evaluate the affinity of cefditoren toward PBPs of Haemophilus influenzae, and the results demonstrated that cefditoren and PBP3A/B had the lowest IC50 values (0.060 ± 0.002 µM). These findings indicate that cefditoren has a strong affinity for PBP1A of H. influenzae. Cefditoren has a high affinity toward the PBP1As of S. pneumoniae and PBP1A and PBP3A/B of H. influenzae, which may contribute to the effective antibacterial effects of cefditoren against clinical strains and its low propensity for inducing resistance. The data presented in this article help elucidate the mechanism by which cefditoren, an oral third-generation cephalosporin, binds to PBPs and provide theoretical support for the wider use of cefditoren as an antibiotic therapy.
Collapse
Affiliation(s)
- Yixin Qi
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, Jiangsu, China
| | - Qixue Shi
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, Jiangsu, China
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, Jiangsu, China
| | - Liang Xu
- Nanjing Neiwa Faith Pharmaceutical Co Ltd., No. 36, Shuanggao Road, Nanjing, 211399, Jiangsu, China
| | - Yi Deng
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No.1617, Riyue Avenue, Qinyang District, Chengdu, 611731, China.
| | - Changlin Zhou
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, Jiangsu, China.
| |
Collapse
|
4
|
Sun C, Zhu L, Yang L, Tian Z, Jiao Z, Huang M, Peng J, Guo G. Antimicrobial peptide AMP-17 induces protection against systemic candidiasis and interacts synergistically with fluconazole against Candida albicans biofilm. Front Microbiol 2024; 15:1480808. [PMID: 39552641 PMCID: PMC11564183 DOI: 10.3389/fmicb.2024.1480808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024] Open
Abstract
Candida albicans, a common commensal and opportunistic fungal pathogen in humans, can occasionally progress to disseminated candidiasis which is a serious condition with a high morbidity and fatality rate. The emergence of drug-resistant fungal strains compels us to look for an efficient treatment solution. Our earlier studies have demonstrated that the unique antimicrobial peptide AMP-17 from Musca domestica has a strong antifungal impact on C. albicans in vitro. Here, we verified the therapeutic effects of AMP-17 on systemic candidiasis in vivo and the peptide interacts with fluconazole, a common antifungal medication, to treat systemic candidiasis. In the disseminated candidiasis model of Galleria mellonella and mice challenged with C. albicans, AMP-17 increased the survival rates of infected larvae and mice to 66.7 and 75%, respectively. Furthermore, the peptide lowered the load of C. albicans in the infected larvae and the kidneys of the mice by nearly 90%. Additional histological examination and measurements of plasma cytokines showed that the injection of AMP-17 markedly reduced the inflammatory response and balanced cytokine expression. Furthermore, checkerboard micro dilution experiments demonstrated that AMP-17 and fluconazole worked in synergy to inhibit C. albicans in the biofilm mode. According to morphological studies, AMP-17 and fluconazole together decreased the production of hyphae throughout the C. albicans biofilm formation process, loosening the mature biofilms' structure and lowering the amount of carbohydrates in the extracellular matrix (ECM) of the biofilms. Taken together, these results showed that AMP-17 would be a viable treatment for systemic candidiasis and might be a different approach to combating Candida biofilm, either by itself or in conjunction with fluconazole.
Collapse
Affiliation(s)
- Chaoqin Sun
- School of Basic Medical Sciences, Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guizhou Medical University, Guiyang, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Center of Laboratory Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lijuan Zhu
- School of Basic Medical Sciences, Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guizhou Medical University, Guiyang, China
- Department of Laboratory Medicine, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Longbing Yang
- School of Basic Medical Sciences, Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guizhou Medical University, Guiyang, China
| | - Zhuqing Tian
- School of Basic Medical Sciences, Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guizhou Medical University, Guiyang, China
| | - Zhenlong Jiao
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, China
| | - Mingjiao Huang
- School of Basic Medical Sciences, Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guizhou Medical University, Guiyang, China
| | - Jian Peng
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Guo Guo
- School of Basic Medical Sciences, Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guizhou Medical University, Guiyang, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, China
| |
Collapse
|
5
|
Dar OA, Hashmi AA, Al-Bogami AS, Ahmad A, Wani MY. Heteroleptic cobalt complex augments antifungal activity with fluconazole and causes membrane disruption in Candida albicans. Dalton Trans 2024; 53:11720-11735. [PMID: 38932585 DOI: 10.1039/d4dt01209g] [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: 06/28/2024]
Abstract
Heteroleptic metal complexes containing CuII, CoII, and ZnII, incorporating curcumin and a Schiff base ligand (L), were synthesized and characterized, and their antifungal activity was evaluated. Their antifungal activities were investigated individually and in combination with fluconazole. Utilizing various analytical techniques such as UV-Vis, FT-IR, NMR, ESI-MS, TGA-DTG, elemental analyses, conductance, and magnetic susceptibility measurements, complex C1 ([Cu(Cur)LCl(H2O)]) was assigned a distorted octahedral geometry, while complexes C2 ([Co(Cur)LCl(H2O)]) and C3 ([Zn(Cur)LCl(H2O)]) were assigned octahedral geometries. Among these complexes, C2 exhibited the highest inhibitory activity against both FLC-susceptible and resistant strains of Candida albicans. Furthermore, C2 demonstrated candidicidal activity and synergistic interactions with fluconazole, effectively inhibiting the growth and survival of both FLC-resistant and FLC-sensitive C. albicans strains. The complex displayed a dose-dependent inhibition of drug efflux pumps in FLC-resistant C. albicans strains, indicating its potential to disrupt the cell membrane of these strains. The significant role of membrane efflux transporters in the development of antifungal drug resistance within Candida species has been extensively documented and our findings indicate that complex C2 specifically targets this crucial factor, thereby playing a pivotal role in mitigating drug resistance in C. albicans.
Collapse
Affiliation(s)
- Ovas Ahmad Dar
- Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Athar Adil Hashmi
- Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
| | - Abdullah Saad Al-Bogami
- Department of Chemistry, College of Science, University of Jeddah, 21589 Jeddah, Saudi Arabia.
| | - Aijaz Ahmad
- Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa.
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Mohmmad Younus Wani
- Department of Chemistry, College of Science, University of Jeddah, 21589 Jeddah, Saudi Arabia.
| |
Collapse
|
6
|
Wang Y, Zhang Y, Su R, Wang Y, Qi W. Antimicrobial therapy based on self-assembling peptides. J Mater Chem B 2024; 12:5061-5075. [PMID: 38726712 DOI: 10.1039/d4tb00260a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
The emergence of drug-resistant microorganisms has threatened global health, and microbial infections have severely limited the use of medical materials. For example, the attachment and colonization of pathogenic bacteria to medical implant materials can lead to wound infections, inflammation and complications, as well as implant failure, shortening their lifespan and even resulting in patient death. In the era of antibiotic resistance, antimicrobial drug discovery needs to prioritize unconventional therapies that act on new targets or adopt new mechanisms. In this regard, supramolecular antimicrobial peptides have emerged as attractive therapeutic platforms, both as bactericides for combination antibiotics and as delivery vehicles. By taking advantage of their programmable intermolecular and intramolecular interactions, peptides can be modified to form higher-order structures (including nanofibers and nanoparticles) with unique functionality. This paper begins with an analysis of the relationship between peptide self-assembly and antimicrobial activity, describes in detail the research and development of various self-assembled antimicrobial peptides in recent years, and finally explores different combinatorial strategies for self-assembling antimicrobial peptides.
Collapse
Affiliation(s)
- Yuqi Wang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
| | - Yexi Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
| | - Rongxin Su
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Yuefei Wang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Wei Qi
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
7
|
Wu Y, Sun A, Chen F, Zhao Y, Zhu X, Zhang T, Ni G, Wang R. Synthesis, structure-activity relationship and biological evaluation of indole derivatives as anti-Candida albicans agents. Bioorg Chem 2024; 146:107293. [PMID: 38507998 DOI: 10.1016/j.bioorg.2024.107293] [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: 11/30/2023] [Revised: 02/29/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
In this work, we synthesized a series of indole derivatives to cope with the current increasing fungal infections caused by drug-resistant Candida albicans. All compounds were evaluated for antifungal activities against Candida albicans in vitro, and the structure-activity relationships (SARs) were analyzed. The results indicated that indole derivatives used either alone or in combination with fluconazole showed good activities against fluconazole-resistant Candida albicans. Further mechanisms studies demonstrated that compound 1 could inhibit yeast-to-hypha transition and biofilm formation of Candida albicans, increase the activity of the efflux pump, the damage of mitochondrial function, and the decrease of intracellular ATP content. In vivo studies, further proved the anti-Candida albicans activity of compound 1 by histological observation. Therefore, compound 1 could be considered as a novel antifungal agent.
Collapse
Affiliation(s)
- Yandan Wu
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Aimei Sun
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Fei Chen
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Yin Zhao
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Xianhu Zhu
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Tianbao Zhang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Guanghui Ni
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China.
| | - Ruirui Wang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China.
| |
Collapse
|
8
|
Zhang M, Wang J, Li C, Wu S, Liu W, Zhou C, Ma L. Cathelicidin AS-12W Derived from the Alligator sinensis and Its Antimicrobial Activity Against Drug-Resistant Gram-Negative Bacteria In Vitro and In Vivo. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10250-2. [PMID: 38587584 DOI: 10.1007/s12602-024-10250-2] [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] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Antimicrobial peptides (AMPs) have the potential to treat multidrug-resistant bacterial infections. Cathelicidins are a class of cationic antimicrobial peptides that are found in nearly all vertebrates. Herein, we determined the mature peptide region of Alligator sinensis cathelicidin by comparing its cathelicidin peptide sequence with those of other reptiles and designed nine peptide mutants based on the Alligator sinensis cathelicidin mature peptide. According to the antibacterial activity and cytotoxicity screening, the peptide AS-12W demonstrated broad-spectrum antibacterial activity and exhibited low erythrocyte hemolytic activity. In particular, AS-12W exhibited strong antibacterial activity and rapid bactericidal activity against carbapenem-resistant Pseudomonas aeruginosa in vitro. Additionally, AS-12W effectively removed carbapenem-resistant P. aeruginosa from blood and organs in vivo, leading to improved survival rates in septic mice. Furthermore, AS-12W exhibited good stability and tolerance to harsh conditions such as high heat, high salt, strong acid, and strong alkali, and it also displayed high stability toward trypsin and simulated gastric fluid (SGF). Moreover, AS-12W showed significant anti-inflammatory effects in vitro by inhibiting the production of proinflammatory factors induced by lipopolysaccharide (LPS). Due to its antibacterial mechanism against Escherichia coli, we found that this peptide could neutralize the negative charge on the surface of the bacteria and disrupt the integrity of the bacterial cell membrane. In addition, AS-12W has the ability to bind to the genomic DNA of bacteria and stimulate the production of reactive oxygen species (ROS) within bacteria, which is believed to be the reason for the good antibacterial activity of AS-12W. These results demonstrated that AS-12W exhibits remarkable antibacterial activity, particularly against carbapenem-resistant P. aeruginosa. Therefore, it is a potential candidate for antibacterial drug development.
Collapse
Affiliation(s)
- Meina Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jian Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Chao Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Shaoju Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Changlin Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
9
|
Lu H, Wang C, Lu W, Li X, Wang G, Dong W, Wang X, Chen H, Tan C. Antibacterial efficacy and mechanism of Cyprinus carpio chemokine-derived L-10 against multidrug-resistant Escherichia coli infections. Int J Antimicrob Agents 2024; 63:107104. [PMID: 38325720 DOI: 10.1016/j.ijantimicag.2024.107104] [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: 11/13/2023] [Revised: 01/02/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVES Antimicrobial resistance has raised concerns regarding untreatable infections and poses a growing threat to public health. Rational design of new AMPs is an ideal solution to this threat. METHODS In this study, we designed, modified, and synthesised an excellent AMP, L-10, based on the original sequence of the Cyprinus carpio chemokine. All experimental data were presented as the mean ± standard deviation (SD), and the two-tailed unpaired T-test method was used to analyze all data. RESULTS L-10 exhibited excellent antibacterial activity with negligible toxicity and improved the efficacy of a broad class of antibiotics against MDR Gram-negative pathogens, including tetracycline, meropenem, levofloxacin, and rifampin. Mechanistic studies have suggested that L-10 targets the bacterial membrane components, LPS and PG, to disrupt bacterial membrane integrity, thereby exerting antibacterial effects and enhancing the efficacy of antibiotics. Moreover, in animal infection models, L-10 significantly increased the survival rate of infected animals and effectively reduced the tissue bacterial load and inflammatory factor levels. In addition to its direct antibacterial activity, L-10 dramatically reduced pulmonary pathological alterations in a mouse model of endotoxemia and suppressed LPS-induced proinflammatory cytokines in vitro and in vivo. Lastly, L-10 was successfully expressed in Pichia pastoris and maintained antimicrobial activity against MDR Gram-negative pathogens in vivo and in vitro. CONCLUSION Collectively, these results reveal the potential of L-10 as an ideal candidate against MDR bacterial infections and provide new insights into the design, development, and clinical application of AMPs.
Collapse
Affiliation(s)
- Hao Lu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Chenchen Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Wenjia Lu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Xiaodan Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Gaoyan Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Wenqi Dong
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China.
| |
Collapse
|
10
|
Chen P, Ye T, Li C, Praveen P, Hu Z, Li W, Shang C. Embracing the era of antimicrobial peptides with marine organisms. Nat Prod Rep 2024; 41:331-346. [PMID: 37743806 DOI: 10.1039/d3np00031a] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Covering: 2018 to Jun of 2023The efficiency of traditional antibiotics has been undermined by the proliferation of antibiotic-resistant pathogenic microorganisms, necessitating the pursuit of innovative therapeutic agents. Antimicrobial peptides (AMPs), which are part of host defence peptides found ubiquitously in nature, exhibiting a wide range of activity towards bacteria, fungi, and viruses, offer a highly promising candidate solution. The efficacy of AMPs can frequently be augmented via alterations to their amino acid sequences or structural adjustments. Given the vast reservoir of marine life forms and their distinctive ecosystems, marine AMPs stand as a burgeoning focal point in the quest for alternative peptide templates extracted from natural sources. Advances in identification and characterization techniques have accelerated the discoveries of marine AMPs, thereby stimulating AMP customization, optimization, and synthesis research endeavours. This review presents an overview of recent discoveries related to the intriguing qualities of marine AMPs. Emphasis will be placed upon post-translational modifications (PTMs) of marine AMPs and how they may impact functionality and potency. Additionally, this review considers ways in which marine PTM might support larger-scale, heterologous AMP manufacturing initiatives, providing insights into translational applications of these important biomolecules.
Collapse
Affiliation(s)
- Pengyu Chen
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Ting Ye
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| | - Chunyuan Li
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| | - Praveen Praveen
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science La Trobe University, Victoria, 3086, Australia.
| | - Zhangli Hu
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| | - Wenyi Li
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science La Trobe University, Victoria, 3086, Australia.
| | - Chenjing Shang
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
11
|
Roy S, Bhogapurapu B, Chandra S, Biswas K, Mishra P, Ghosh A, Bhunia A. Host antimicrobial peptide S100A12 disrupts the fungal membrane by direct binding and inhibits growth and biofilm formation of Fusarium species. J Biol Chem 2024; 300:105701. [PMID: 38301897 PMCID: PMC10891332 DOI: 10.1016/j.jbc.2024.105701] [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: 07/12/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024] Open
Abstract
Fungal keratitis is the foremost cause of corneal infections worldwide, of which Fusariumspp. is the common etiological agent that causes loss of vision and warrants surgical intervention. An increase in resistance to the available drugs along with severe side effects of the existing antifungals demands for new effective antimycotics. Here, we demonstrate that antimicrobial peptide S100A12 directly binds to the phospholipids of the fungal membrane, disrupts the structural integrity, and induces generation of reactive oxygen species in fungus. In addition, it inhibits biofilm formation by Fusariumspp. and exhibits antifungal property against Fusariumspp. both in vitro and in vivo. Taken together, our results delve into specific effect of S100A12 against Fusariumspp. with an aim to investigate new antifungal compounds to combat fungal keratitis.
Collapse
Affiliation(s)
- Sanhita Roy
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India; Dr. Chigurupati Nageswara Rao Ocular Pharmacology Research Centre, LV Prasad Eye Institute, Hyderabad, India.
| | - Bharathi Bhogapurapu
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | - Sreyanki Chandra
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India; Dr. Chigurupati Nageswara Rao Ocular Pharmacology Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | - Karishma Biswas
- Department of Chemical Sciences, Bose Institute, Unified Academic Campus, Kolkata, India
| | - Priyasha Mishra
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India; Graduate Studies, Manipal Academy of Higher Education, Manipal, India
| | - Abhijit Ghosh
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India; Dr. Chigurupati Nageswara Rao Ocular Pharmacology Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | - Anirban Bhunia
- Department of Chemical Sciences, Bose Institute, Unified Academic Campus, Kolkata, India
| |
Collapse
|
12
|
Shang L, Chen C, Sun R, Guo J, Liu J, Wang M, Zhang L, Fei C, Xue F, Liu Y, Gu F. Engineered Peptides Harboring Cation Motifs Against Multidrug-Resistant Bacteria. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5522-5535. [PMID: 38266749 DOI: 10.1021/acsami.3c15913] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Multidrug-resistant (MDR) pathogens pose a serious threat to the health and life of humans, necessitating the development of new antimicrobial agents. Herein, we develop and characterize a panel of nine amino acid peptides with a cation end motif. Bioactivity analysis revealed that the short peptide containing "RWWWR" as a central motif harboring mirror structure "KXR" unit displayed not only high activity against MDR planktonic bacteria but also a clearance rate of 92.33% ± 0.58% against mature biofilm. Mechanically, the target peptide (KLR) killed pathogens by excessively accumulating reactive oxygen species and physically disrupting membranes, thereby enhancing its robustness for controlling drug resistance. In the animal model of sepsis infection by MDR bacteria, the peptide KLR exhibited strong therapeutic effects. Collectively, this study provided the dominant structure of short antimicrobial peptides (AMPs) to replenish our arsenals for combating bacterial infections and illustrated what could be harnessed as a new agent for fighting MDR bacteria.
Collapse
Affiliation(s)
- Lu Shang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Chan Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Rui Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Juan Guo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Jing Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Mi Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Lifang Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Chenzhong Fei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Feiqun Xue
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Yingchun Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Feng Gu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| |
Collapse
|
13
|
Wang L, Liu M, Qi Y, Wang J, Shi Q, Xie X, Zhou C, Ma L. hsdSA regulated extracellular vesicle-associated PLY to protect Streptococcus pneumoniae from macrophage killing via LAPosomes. Microbiol Spectr 2024; 12:e0099523. [PMID: 38018988 PMCID: PMC10783081 DOI: 10.1128/spectrum.00995-23] [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: 03/06/2023] [Accepted: 08/01/2023] [Indexed: 11/30/2023] Open
Abstract
IMPORTANCE S. pneumoniae is a major human pathogen that undergoes a spontaneous and reversible phase variation that allows it to survive in different host environments. Interestingly, we found hsdSA , a gene that manipulated the phase variation, promoted the survival and replication of S. pneumoniae in macrophages by regulating EV production and EV-associated PLY. More importantly, here we provided the first evidence that higher EV-associated PLY (produced by D39) could form LAPosomes that were single membrane compartments containing S. pneumoniae, which are induced by integrin β1/NOX2/ROS pathway. At the same time, EV-associated PLY increased the permeability of lysosome membrane and induced an insufficient acidification to escape the host killing, and ultimately prolonged the survival of S. pneumoniae in macrophages. In contrast, lower EV-associated PLY (produced by D39ΔhsdSA ) activated ULK1 recruitment to form double-layered autophagosomes to eliminate bacteria.
Collapse
Affiliation(s)
- Liping Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mengyuan Liu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yixin Qi
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jian Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qixue Shi
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaolin Xie
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Changlin Zhou
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lingman Ma
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
14
|
Goki NH, Tehranizadeh ZA, Saberi MR, Khameneh B, Bazzaz BSF. Structure, Function, and Physicochemical Properties of Pore-forming Antimicrobial Peptides. Curr Pharm Biotechnol 2024; 25:1041-1057. [PMID: 37921126 DOI: 10.2174/0113892010194428231017051836] [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: 06/24/2023] [Revised: 08/28/2023] [Accepted: 09/08/2023] [Indexed: 11/04/2023]
Abstract
Antimicrobial peptides (AMPs), a class of antimicrobial agents, possess considerable potential to treat various microbial ailments. The broad range of activity and rare complete bacterial resistance to AMPs make them ideal candidates for commercial development. These peptides with widely varying compositions and sources share recurrent structural and functional features in mechanisms of action. Studying the mechanisms of AMP activity against bacteria may lead to the development of new antimicrobial agents that are more potent. Generally, AMPs are effective against bacteria by forming pores or disrupting membrane barriers. The important structural aspects of cytoplasmic membranes of pathogens and host cells will also be outlined to understand the selective antimicrobial actions. The antimicrobial activities of AMPs are related to multiple physicochemical properties, such as length, sequence, helicity, charge, hydrophobicity, amphipathicity, polar angle, and also self-association. These parameters are interrelated and need to be considered in combination. So, gathering the most relevant available information will help to design and choose the most effective AMPs.
Collapse
Affiliation(s)
- Narjes Hosseini Goki
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeinab Amiri Tehranizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Saberi
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahman Khameneh
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Sedigheh Fazly Bazzaz
- Department of Pharmaceutical Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Suchi SA, Lee DY, Kim YK, Kang SS, Bilkis T, Yoo JC. Synergistic Effect, Improved Cell Selectivity, and Elucidating the Action Mechanism of Antimicrobial Peptide YS12. Int J Mol Sci 2023; 24:13522. [PMID: 37686328 PMCID: PMC10487915 DOI: 10.3390/ijms241713522] [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: 06/06/2023] [Revised: 08/15/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Antimicrobial peptides (AMPs) have attracted considerable attention as potential substitutes for traditional antibiotics. In our previous research, a novel antimicrobial peptide YS12 derived from the Bacillus velezensis strain showed broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria. In this study, the fractional inhibitory concentration index (FICI) indicated that combining YS12 with commercial antibiotics produced a synergistic effect. Following these findings, the combination of YS12 with an antibiotic resulted in a faster killing effect against bacterial strains compared to the treatment with the peptide YS12 or antibiotic alone. The peptide YS12 maintained its antimicrobial activity under different physiological salts (Na+, Mg2+, and Fe3+). Most importantly, YS12 exhibited no cytotoxicity towards Raw 264.7 cells and showed low hemolytic activity, whereas positive control melittin indicated extremely high toxicity. In terms of mode of action, we found that peptide YS12 was able to bind with LPS through electrostatic interaction. The results from fluorescent measurement revealed that peptide YS12 damaged the integrity of the bacterial membrane. Confocal laser microscopy further confirmed that the localization of peptide YS12 was almost in the cytoplasm of the cells. Peptide YS12 also exhibited anti-inflammatory activity by reducing the release of LPS-induced pro-inflammatory mediators such as TNF-α, IL-1β, and NO. Collectively, these properties strongly suggest that the antimicrobial peptide YS12 may be a promising candidate for treating microbial infections and inflammation.
Collapse
Affiliation(s)
- Suzia Aktar Suchi
- Department of Pharmacy, College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Young Kyun Kim
- Department of Pharmacy, College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Seong Soo Kang
- Department of Veterinary Medicine and BK21 Four Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Tahmina Bilkis
- Department of Biomedical Sciences, Chosun University, Gwangju 61452, Republic of Korea
| | - Jin Cheol Yoo
- Department of Pharmacy, College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
16
|
Agüero-Chapin G, Antunes A, Mora JR, Pérez N, Contreras-Torres E, Valdes-Martini JR, Martinez-Rios F, Zambrano CH, Marrero-Ponce Y. Complex Networks Analyses of Antibiofilm Peptides: An Emerging Tool for Next-Generation Antimicrobials' Discovery. Antibiotics (Basel) 2023; 12:antibiotics12040747. [PMID: 37107109 PMCID: PMC10135022 DOI: 10.3390/antibiotics12040747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Microbial biofilms cause several environmental and industrial issues, even affecting human health. Although they have long represented a threat due to their resistance to antibiotics, there are currently no approved antibiofilm agents for clinical treatments. The multi-functionality of antimicrobial peptides (AMPs), including their antibiofilm activity and their potential to target multiple microbes, has motivated the synthesis of AMPs and their relatives for developing antibiofilm agents for clinical purposes. Antibiofilm peptides (ABFPs) have been organized in databases that have allowed the building of prediction tools which have assisted in the discovery/design of new antibiofilm agents. However, the complex network approach has not yet been explored as an assistant tool for this aim. Herein, a kind of similarity network called the half-space proximal network (HSPN) is applied to represent/analyze the chemical space of ABFPs, aiming to identify privileged scaffolds for the development of next-generation antimicrobials that are able to target both planktonic and biofilm microbial forms. Such analyses also considered the metadata associated with the ABFPs, such as origin, other activities, targets, etc., in which the relationships were projected by multilayer networks called metadata networks (METNs). From the complex networks' mining, a reduced but informative set of 66 ABFPs was extracted, representing the original antibiofilm space. This subset contained the most central to atypical ABFPs, some of them having the desired properties for developing next-generation antimicrobials. Therefore, this subset is advisable for assisting the search for/design of both new antibiofilms and antimicrobial agents. The provided ABFP motifs list, discovered within the HSPN communities, is also useful for the same purpose.
Collapse
Affiliation(s)
- Guillermin Agüero-Chapin
- CIIMAR/CIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Porto, Portugal
- Department of Biology, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Agostinho Antunes
- CIIMAR/CIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208 Porto, Portugal
- Department of Biology, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - José R Mora
- Universidad San Francisco de Quito (USFQ), Colegio de Ciencias e Ingenierías "El Politécnico", Instituto de Simulación Computacional (ISC-USFQ), Diego de Robles y vía Interoceánica, Quito 170157, Pichincha, Ecuador
| | - Noel Pérez
- Universidad San Francisco de Quito (USFQ), Colegio de Ciencias e Ingenierías "El Politécnico", Instituto de Simulación Computacional (ISC-USFQ), Diego de Robles y vía Interoceánica, Quito 170157, Pichincha, Ecuador
| | - Ernesto Contreras-Torres
- Universidad San Francisco de Quito (USFQ), Grupo de Medicina Molecular y Traslacional (MeM&T), Colegio de Ciencias de la Salud (COCSA), Escuela de Medicina, Edificio de Especialidades Médicas and Instituto de Simulación Computacional (ISC-USFQ), Diego de Robles y vía Interoceánica, Quito 170157, Pichincha, Ecuador
| | | | - Felix Martinez-Rios
- Facultad de Ingeniería, Universidad Panamericana, Augusto Rodin No. 498, Insurgentes Mixcoac, Benito Juárez, Ciudad de México 03920, Mexico
| | - Cesar H Zambrano
- Universidad San Francisco de Quito (USFQ), Colegio de Ciencias e Ingenierías "El Politécnico", Instituto de Simulación Computacional (ISC-USFQ), Diego de Robles y vía Interoceánica, Quito 170157, Pichincha, Ecuador
| | - Yovani Marrero-Ponce
- Universidad San Francisco de Quito (USFQ), Grupo de Medicina Molecular y Traslacional (MeM&T), Colegio de Ciencias de la Salud (COCSA), Escuela de Medicina, Edificio de Especialidades Médicas and Instituto de Simulación Computacional (ISC-USFQ), Diego de Robles y vía Interoceánica, Quito 170157, Pichincha, Ecuador
- Departamento de Ciencias de la Computación, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, Baja California, Mexico
| |
Collapse
|
17
|
Jin X, Hu X, Jiang S, Zhao T, Zha Y, Wei S, Zhao J, Wang M, Zhang Y. Temporin-GHb-Derived Peptides Exhibit Potent Antibacterial and Antibiofilm Activities against Staphylococcus aureus In Vitro and Protect Mice from Acute Infectious Pneumonia. ACS Infect Dis 2023; 9:840-855. [PMID: 36862073 DOI: 10.1021/acsinfecdis.2c00544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
With the continuous development of drug resistance in bacteria to traditional antibiotics, the demand for novel antibacterial agents is urgent. Antimicrobial peptides (AMPs) are promising candidates because of their unique mechanism of action and low tendency to induce drug resistance. Previously, we cloned temporin-GHb (hereafter referred to simply as "GHb") from Hylarana guentheri. In this study, a series of derived peptides were designed, namely, GHbR, GHbK, GHb3K, GHb11K, and GHbK4R. The five derived peptides had stronger antibacterial activities against Staphylococcus aureus than the parent peptide GHb and could effectively inhibit the formation of biofilms and eradicate mature biofilms in vitro. GHbR, GHbK, GHb3K, and GHbK4R exerted bactericidal effects by disrupting membrane integrity. However, GHb11K exhibited bacteriostatic efficacy with toroidal pore formation on the cell membrane. In comparison to GHbK4R, GHb3K showed much lower cytotoxicity against A549 alveolar epithelial cells, with an IC50 > 200 μM, which was much higher than its minimal inhibitory concentration (MIC = 3.1 μM) against S. aureus. The anti-infection potential of GHbK4R and GHb3K was investigated in vivo. Compared with vancomycin, the two peptides displayed significant efficacy in a mouse model of acute pneumonia infected with S. aureus. Both GHbK4R and GHb3K also had no obvious toxicity to normal mice after intraperitoneal administration (15 mg/kg) for 8 days. Our results indicate that GHb3K and GHbK4R might be promising candidates for the treatment of bacterial pneumonia infected with S. aureus.
Collapse
|
18
|
Guo X, Yan T, Rao J, An Y, Yue X, Miao X, Wang R, Sun W, Cai J, Xie J. Novel Feleucin-K3-Derived Peptides Modified with Sulfono-γ-AA Building Blocks Targeting Pseudomonas aeruginosa and Methicillin-Resistant Staphylococcus aureus Infections. J Med Chem 2023; 66:1254-1272. [PMID: 36350686 DOI: 10.1021/acs.jmedchem.2c01396] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The prevalence of multidrug-resistant bacterial infections has led to dramatically increased morbidity and mortality. Antimicrobial peptides (AMPs) have great potential as new therapeutic agents to reverse this dangerous trend. Herein, a series of novel AMP Feleucin-K3 analogues modified with unnatural peptidomimetic sulfono-γ-AA building blocks were designed and synthesized. The structure-activity, structure-toxicity, and structure-stability relationships were investigated to discover the optimal antimicrobial candidates. Among them, K122 exhibited potent and broad-spectrum antimicrobial activity and high selectivity. K122 had a rapid bactericidal effect and a low tendency to induce resistance. Surprisingly, K122 showed excellent effectiveness against bacterial pneumonia. For biofilm and local skin infections, K122 significantly decreased the bacterial load and improved tissue injury at a dose of only 0.25 mg/kg, which was 160 times lower than the concentration deemed to be safe for local dermal applications. In summary, K122 is an outstanding candidate for the treatment of multidrug-resistant bacteria and biofilm infections.
Collapse
Affiliation(s)
- Xiaomin Guo
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou730000, China
| | - Tiantian Yan
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou730000, China
| | - Jing Rao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou730000, China
| | - Yingying An
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou730000, China
| | - Xin Yue
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou730000, China
| | - Xiaokang Miao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou730000, China
| | - Rui Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou730000, China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100050, China
| | - Wangsheng Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou730000, China
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, Florida33620, United States
| | - Junqiu Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou730000, China
| |
Collapse
|
19
|
Silva PSE, Guindo AS, Oliveira PHC, de Moraes LFRN, Boleti APDA, Ferreira MA, de Oliveira CFR, Macedo MLR, Rossato L, Simionatto S, Migliolo L. Evaluation of the Synthetic Multifunctional Peptide Hp-MAP3 Derivative of Temporin-PTa. Toxins (Basel) 2023; 15:42. [PMID: 36668862 PMCID: PMC9866994 DOI: 10.3390/toxins15010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 01/06/2023] Open
Abstract
In recent years, antimicrobial peptides isolated from amphibian toxins have gained attention as new multifunctional drugs interacting with different molecular targets. We aimed to rationally design a new peptide from temporin-PTa. Hp-MAP3 (NH2-LLKKVLALLKKVL-COOH), net charge (+4), hydrophobicity (0.69), the content of hydrophobic residues (69%), and hydrophobic moment (0.73). For the construction of the analog peptide, the physicochemical characteristics were reorganized into hydrophilic and hydrophobic residues with the addition of lysines and leucines. The minimum inhibitory concentration was 2.7 to 43 μM against the growth of Gram-negative and positive bacteria, and the potential for biofilm eradication was 173.2 μM. Within 20 min, the peptide Hp-MAP3 (10.8 μM) prompted 100% of the damage to E. coli cells. At 43.3 μM, eliminated 100% of S. aureus within 5 min. The effects against yeast species of the Candida genus ranged from 5.4 to 86.6 μM. Hp-MAP3 presents cytotoxic activity against tumor HeLa at a concentration of 21.6 μM with an IC50 of 10.4 µM. Furthermore, the peptide showed hemolytic activity against murine erythrocytes. Structural studies carried out by circular dichroism showed that Hp-MAP3, while in the presence of 50% trifluoroethanol or SDS, an α-helix secondary structure. Finally, Amphipathic Hp-MAP3 building an important model for the design of new multifunctional molecules.
Collapse
Affiliation(s)
- Patrícia Souza e Silva
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | - Alexya Sandim Guindo
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | - Pedro Henrique Cardoso Oliveira
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | | | - Ana Paula de Araújo Boleti
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | - Marcos Antonio Ferreira
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | - Caio Fernando Ramalho de Oliveira
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Unidade de Tecnologia de Alimentos e da Saúde Pública, Universidade Federal de Mato Grosso do Sul, Campo Grande 79070-900, Mato Grosso do Sul, Brazil
| | - Maria Ligia Rodrigues Macedo
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Unidade de Tecnologia de Alimentos e da Saúde Pública, Universidade Federal de Mato Grosso do Sul, Campo Grande 79070-900, Mato Grosso do Sul, Brazil
| | - Luana Rossato
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados UFGD, Dourados 79825-070, Mato Grosso do Sul, Brazil
| | - Simone Simionatto
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados UFGD, Dourados 79825-070, Mato Grosso do Sul, Brazil
| | - Ludovico Migliolo
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| |
Collapse
|
20
|
Peng C, Liu Y, Shui L, Zhao Z, Mao X, Liu Z. Mechanisms of Action of the Antimicrobial Peptide Cecropin in the Killing of Candida albicans. LIFE (BASEL, SWITZERLAND) 2022; 12:life12101581. [PMID: 36295016 PMCID: PMC9604627 DOI: 10.3390/life12101581] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022]
Abstract
The development of drug resistance has caused fungal infections to become a global health concern. Antimicrobial peptides (AMPs) offer a viable solution to these pathogens due to their resistance to drug resistance and their diverse mechanisms of actions, which include direct killing and immunomodulatory properties. The peptide Cecropin, which is expressed by genetically engineered bacteria, has antifungal effects on Candida albicans. The minimal inhibitory concentration (MIC) and the minimal fungicidal concentration (MFC) of Candida albicans were 0.9 μg/mL and 1.8 μg/mL, respectively, detected by the micro-broth dilution method. According to the killing kinetics, the MFC of Cecropin could kill Candida albicans in 40 min. The electron microscope indicated that Cecropin could cause the cell wall to become rough and nicked, eventually killing Candida albicans. The effects of Cecropin on the cell membrane of treated C. albicans, using the 1,6-diphenyl-1,3,5-hexatriene and propidium iodide protocol, showed that they could change the permeability and fluidity, destroy it, and lead to cell necrosis. In addition, Cecropin can also induce cells to produce excessive reactive oxygen species, causing changes in the mitochondrial membrane potential. Therefore, this study provides a certain theoretical basis for the antifungal infection of new antifungal agents.
Collapse
|
21
|
Wang X, Hong X, Chen F, Wang KJ. A truncated peptide Spgillcin177–189 derived from mud crab Scylla paramamosain exerting multiple antibacterial activities. Front Cell Infect Microbiol 2022; 12:928220. [PMID: 36061863 PMCID: PMC9435603 DOI: 10.3389/fcimb.2022.928220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Antimicrobial peptides (AMPs) may be the most promising substitute for antibiotics due to their effective bactericidal activity and multiple antimicrobial modes against pathogenic bacteria. In this study, a new functional gene named Spgillcin was identified in Scylla paramamosain, which encoded 216 amino acids of mature peptide. In vivo, Spgillcin was dominantly expressed in the gills of male and female crabs, offering the highest expression level among all tested organs or tissues. The expression pattern of Spgillcin was significantly altered when challenged by Staphylococcus aureus, indicating a positive immune response. In vitro, a functional truncated peptide Spgillcin177–189 derived from the amino acid sequence of Spgillcin was synthesized and showed a broad-spectrum and potent antibacterial activity against several bacterial strains, including the clinical isolates of multidrug-resistant (MDR) strains, with a range of minimum inhibitory concentrations from 1.5 to 48 μM. Spgillcin177–189 also showed rapid bactericidal kinetics for S. aureus and Pseudomonas aeruginosa but did not display any cytotoxicity to mammalian cells and maintained its antimicrobial activity in different conditions. Mechanistic studies indicated that Spgillcin177–189 was mainly involved in the disruption of cell membrane integrity where the membrane components lipoteichoic acid and lipopolysaccharide could significantly inhibit the antimicrobial activity in a dose-dependent manner. In addition, Spgillcin177–189 could change the membrane permeability and cause the accumulation of intracellular reactive oxygen species. No resistance was generated to Spgillcin177–189 when the clinical isolates of methicillin-resistant S. aureus and MDR P. aeruginosa were treated with Spgillcin177–189 and then subjected to a long term of continuous culturing for 50 days. In addition, Spgillcin177–189 exerted a strong anti-biofilm activity by inhibiting biofilm formation and was also effective at killing extracellular S. aureus in the cultural supernatant of RAW 264.7 cells. Taken together, Spgillcin177–189 has strong potential as a substitute for antibiotics in future aquaculture and medical applications.
Collapse
Affiliation(s)
- Xiaofei Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Xiao Hong
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- *Correspondence: Ke-Jian Wang,
| |
Collapse
|
22
|
Freitas ED, Bataglioli RA, Oshodi J, Beppu MM. Antimicrobial peptides and their potential application in antiviral coating agents. Colloids Surf B Biointerfaces 2022; 217:112693. [PMID: 35853393 PMCID: PMC9262651 DOI: 10.1016/j.colsurfb.2022.112693] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 11/24/2022]
Abstract
Coronavirus pandemic has evidenced the importance of creating bioactive materials to mitigate viral infections, especially in healthcare settings and public places. Advances in antiviral coatings have led to materials with impressive antiviral performance; however, their application may face health and environmental challenges. Bio-inspired antimicrobial peptides (AMPs) are suitable building blocks for antimicrobial coatings due to their versatile design, scalability, and environmentally friendly features. This review presents the advances and opportunities on the AMPs to create virucidal coatings. The review first describes the fundamental characteristics of peptide structure and synthesis, highlighting the recent findings on AMPs and the role of peptide structure (α-helix, β-sheet, random, and cyclic peptides) on the virucidal mechanism. The following section presents the advances in AMPs coating on medical devices with a detailed description of the materials coated and the targeted pathogens. The use of peptides in vaccine formulations is also reported, emphasizing the molecular interaction of peptides with different viruses and the current clinical stage of each formulation. The role of several materials (metallic particles, inorganic materials, and synthetic polymers) in the design of antiviral coatings is also presented, discussing the advantages and the drawbacks of each material. The final section offers future directions and opportunities for using AMPs on antiviral coatings to prevent viral outbreaks.
Collapse
Affiliation(s)
- Emanuelle D Freitas
- School of Chemical Engineering, Department of Materials and Bioprocess Engineering, University of Campinas, Campinas, São Paulo 13083-852, Brazil
| | - Rogério A Bataglioli
- School of Chemical Engineering, Department of Materials and Bioprocess Engineering, University of Campinas, Campinas, São Paulo 13083-852, Brazil
| | - Josephine Oshodi
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Marisa M Beppu
- School of Chemical Engineering, Department of Materials and Bioprocess Engineering, University of Campinas, Campinas, São Paulo 13083-852, Brazil.
| |
Collapse
|
23
|
Xu L, She P, Liu Y, Liu S, Li Z, Li Y, Hussain Z, Wu Y. A novel bactericidal small molecule, STK-35, and its derivative, STK-66, as antibacterial agents against Gram-negative pathogenic bacteria in vitro and in vivo. Lett Appl Microbiol 2022; 75:655-666. [PMID: 35218030 DOI: 10.1111/lam.13682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/29/2022]
Abstract
Due to the increasing rate of antibiotic resistance and the emergence of persister cells of Gram-negative pathogenic bacteria, the development of new antibacterial agents is urgently needed to deal with this problem. Our results indicated that both newly identified small molecule STK-35 and its derivative STK-66 exhibited effective antibacterial properties against a variety of Gram-negative pathogens including A. baumannii, E. coli, K. pneumoniae and P. aeruginosa. The minimal inhibitory concentrations (MICs) and minimal bactericidal concentrations (MBCs) ranges were 0.0625-8 μg mL-1 and 0.125-16 μg mL-1 respectively, while no hemolytic activity and mammalian cell cytotoxicity were observed. The time-killing assays showed STK-35/66 had strong bactericidal activity against Gram-negative pathogens. STK-35/66 also showed different degrees of synergistic antibacterial activity with conventional antibiotics and exhibited persister cells killing activity. Moreover, STK-35/66 effectively eradicated the pre-formed biofilms of P. aeruginosa and A. baumannii. In addition, STK-35/66 significantly increased the survival rate of E. coli infected mice and induced a decrease in bacterial load of the peritonitis model. In nutshell, these results suggested that STK-35/66 possessed antimicrobial activity against Gram-negative pathogenic bacteria in vitro and in vivo, which could be considered as potential substitutes for the treatment of Gram-negative pathogenic infections after further structure optimization.
Collapse
Affiliation(s)
- Lanlan Xu
- Department of Laboratory Medicine, Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Pengfei She
- Department of Laboratory Medicine, Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Yaqian Liu
- Department of Laboratory Medicine, Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Shasha Liu
- Department of Laboratory Medicine, Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Zehao Li
- Department of Laboratory Medicine, Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Yimin Li
- Department of Laboratory Medicine, Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Zubair Hussain
- Department of Laboratory Medicine, Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Yong Wu
- Department of Laboratory Medicine, Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China.,Department of Laboratory Medicine, The First Hospital of Changsha, Changsha, 410013, Hunan, China
| |
Collapse
|
24
|
Shang L, Wu Y, Wei N, Yang F, Wang M, Zhang L, Fei C, Liu Y, Xue F, Gu F. Novel Arginine End-Tagging Antimicrobial Peptides to Combat Multidrug-Resistant Bacteria. ACS APPLIED MATERIALS & INTERFACES 2022; 14:245-258. [PMID: 34964342 DOI: 10.1021/acsami.1c19305] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The emergence of multidrug-resistant microorganisms has been termed one of the most common global health threats, emphasizing the discovery of new antibacterial agents. To address this issue, we engineered peptides harboring "RWWWR" as a central motif plus arginine (R) end-tagging and then tested them in vitro and in vivo. Our results demonstrate that Pep 6, one of the engineered peptides, shows great potential in combating Escherichia coli bacteremia and the Staphylococcus aureus skin burn infection model, which induces a 62-90% reduction in bacterial burden. Remarkably, after long serial passages of S. aureus and E. coli for 30 days, Pep 6 is still highly efficient in killing pathogens, compared with 64- and 128-fold increase in minimal inhibitory concentrations (MICs) for vancomycin and polymyxin B, respectively. We also found that Pep 6 exhibited robust biofilm-inhibiting activity and eliminated 61.33% of the mature methicillin-resistant Staphylococcus aureus (MRSA) biofilm with concentration in the MIC level. These results suggest that the RWWWR motif and binding of arginine end-tagging could be harnessed as a new agent for combating multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Lu Shang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Yuting Wu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Nan Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Fayu Yang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Mi Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Lifang Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Chenzhong Fei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Yingchun Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Feiqun Xue
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| | - Feng Gu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai 200241, China
| |
Collapse
|
25
|
Yang L, Tian Z, Zhou L, Zhu L, Sun C, Huang M, Peng J, Guo G. In vitro Antifungal Activity of a Novel Antimicrobial Peptide AMP-17 Against Planktonic Cells and Biofilms of Cryptococcus neoformans. Infect Drug Resist 2022; 15:233-248. [PMID: 35115792 PMCID: PMC8800587 DOI: 10.2147/idr.s344246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/08/2022] [Indexed: 12/30/2022] Open
Abstract
Background Cryptococcus neoformans is a common human fungal pathogen in immunocompromised people, as well as a prevalent cause of meningitis in HIV-infected individuals. With the emergence of clinical fungal resistance and the shortage of antifungal drugs, it is urgent to discover novel antifungal agents. AMP-17, a novel antimicrobial peptide from Musca domestica, has antifungal activity against C. neoformans. However, its antifungal and anti-biofilm activities remain unclear. Thus, this study aimed to evaluate the antifungal activity of AMP-17 against planktonic cells and biofilms of C. neoformans. Methods The minimum inhibitory concentration (MIC), the biofilm inhibitory and eradicating concentration (BIC and BEC) were determined by the broth microdilution assay or the 2, 3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide (XTT) reduction assay, respectively. The inhibitory and killing activities of AMP-17 against C. neoformans were investigated through the time-inhibition/killing kinetic curves. The potential antifungal mechanism of AMP-17 was detected by flow cytometry, scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). The efficiency of AMP-17 against biofilm formation or preformed biofilm was evaluated by crystal violet staining and XTT reduction assays. The morphology of pre-biofilms was tested by optical microscopy (OM) and CLSM. Results AMP-17 exhibited in vitro antifungal activity against C. neoformans planktonic cells and biofilms, with MICs of 4~16 μg/ml, BIC80 and BEC80 of 16~32 μg/ml, 64~128 μg/ml, respectively. In addition, the 2× and 4× MIC of AMP-17 exhibited similar inhibition levels compared to the 2× and 4× MIC of the clinical drugs FLC and AMB in C. neoformans growth. Moreover, the time-kill results showed that AMP-17 (8× MIC) did not significantly eliminate colony forming units (CFU) after 6 h of treatment; however, there was 2.9-log reduction in CFU of C. neoformans. Furthermore, increasing of the permeability of the fungal cell membrane was observed with the treatment of AMP-17, since the vast change as fungal leakage and cell membrane disruption. However, the DNA binding assay of AMP-17 indicated that the peptide did not target DNA. Besides, AMP-17 was superior in inhibiting and eradicating biofilms of C. neoformans compared with FLC. Conclusion AMP-17 exhibited potential in vitro antifungal activity against the planktonic cells and biofilms of C. neoformans, and it may disrupt fungal cell membranes through multi-target interactions, which provides a promising therapeutic strategy and experimental basis for Cryptococcus-associated infections.
Collapse
Affiliation(s)
- Longbing Yang
- School of Basic Medical Sciences, The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
| | - Zhuqing Tian
- School of Basic Medical Sciences, The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
| | - Luoxiong Zhou
- School of Basic Medical Sciences, The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
- School of Public Health, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
| | - Lijuan Zhu
- School of Basic Medical Sciences, The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
| | - Chaoqin Sun
- School of Basic Medical Sciences, The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
| | - Mingjiao Huang
- School of Basic Medical Sciences, The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
| | - Jian Peng
- School of Basic Medical Sciences, The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
| | - Guo Guo
- School of Basic Medical Sciences, The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, 550025, People’s Republic of China
- Correspondence: Guo Guo, Building Wuben, School of Basic Medical Sciences, Guizhou Medical University, College Town, Gui’an New District, Guiyang, 550025, People’s Republic of China, Tel/fax +86 851 882 59268, Email
| |
Collapse
|
26
|
Potent antibacterial and antibiofilm activities of TICbf-14, a peptide with increased stability against trypsin. J Microbiol 2021; 60:89-99. [PMID: 34964945 DOI: 10.1007/s12275-022-1368-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/17/2021] [Accepted: 10/01/2021] [Indexed: 10/19/2022]
Abstract
The poor stability of peptides against trypsin largely limits their development as potential antibacterial agents. Here, to obtain a peptide with increased trypsin stability and potent antibacterial activity, TICbf-14 derived from the cationic peptide Cbf-14 was designed by the addition of disulfide-bridged hendecapeptide (CWTKSIPPKPC) loop. Subsequently, the trypsin stability and antimicrobial and antibiofilm activities of this peptide were evaluated. The possible mechanisms underlying its mode of action were also clarified. The results showed that TICbf-14 exhibited elevated trypsin inhibitory activity and effectively mitigated lung histopathological damage in bacteria-infected mice by reducing the bacterial counts, further inhibiting the systemic dissemination of bacteria and host inflammation. Additionally, TICbf-14 significantly repressed bacterial swimming motility and notably inhibited biofilm formation. Considering the mode of action, we observed that TICbf-14 exhibited a potent membrane-disruptive mechanism, which was attributable to its destructive effect on ionic bridges between divalent cations and LPS of the bacterial membrane. Overall, TICbf-14, a bifunctional peptide with both antimicrobial and trypsin inhibitory activity, is highly likely to become an ideal candidate for drug development against bacteria.
Collapse
|
27
|
Guo HN, Tong YC, Wang HL, Zhang J, Li ZX, Abbas Z, Yang TT, Liu MY, Chen PY, Hua ZC, Yan XN, Cheng Q, Ahmat M, Wang JY, Zhang LL, Wei XB, Liao XD, Zhang RJ. Novel Hybrid Peptide Cathelicidin 2 (1-13)-Thymopentin (TP5) and Its Derived Peptides with Effective Antibacterial, Antibiofilm, and Anti-Adhesion Activities. Int J Mol Sci 2021; 22:11681. [PMID: 34769113 PMCID: PMC8583881 DOI: 10.3390/ijms222111681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 02/08/2023] Open
Abstract
The increasing numbers of infections caused by multidrug-resistant (MDR) pathogens highlight the urgent need for new alternatives to conventional antibiotics. Antimicrobial peptides have the potential to be promising alternatives to antibiotics because of their effective bactericidal activity and highly selective toxicity. The present study was conducted to investigate the antibacterial, antibiofilm, and anti-adhesion activities of different CTP peptides (CTP: the original hybrid peptide cathelicidin 2 (1-13)-thymopentin (TP5); CTP-NH2: C-terminal amidated derivative of cathelicidin 2 (1-13)-TP5; CTPQ: glutamine added at the C-terminus of cathelicidin 2 (1-13)-TP5) by determining the minimal inhibitory concentrations (MICs), minimal bactericidal concentrations (MBCs), propidium iodide uptake, and analysis by scanning electron microscopy, transmission electron microscopy, and confocal laser scanning microscopy). The results showed that CTPs had broad-spectrum antibacterial activity against different gram-positive and gram-negative bacteria, with MICs against the tested strains varying from 2 to 64 μg/mL. CTPs at the MBC (2 × MIC 64 μg/mL) showed strong bactericidal effects on a standard methicillin-resistant Staphylococcus aureus strain ATCC 43300 after co-incubation for 6 h through disruption of the bacterial membrane. In addition, CTPs at 2 × MIC also displayed effective inhibition activity of several S. aureus strains with a 40-90% decrease in biofilm formation by killing the bacteria embedded in the biofilms. CTPs had low cytotoxicity on the intestinal porcine epithelial cell line (IPEC-J2) and could significantly decrease the rate of adhesion of S. aureus ATCC 43300 on IPEC-J2 cells. The current study proved that CTPs have effective antibacterial, antibiofilm, and anti-adhesion activities. Overall, this study contributes to our understanding of the possible antibacterial and antibiofilm mechanisms of CTPs, which might be an effective anti-MDR drug candidate.
Collapse
Affiliation(s)
- He-Nan Guo
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Yu-Cui Tong
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Hui-Li Wang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Jing Zhang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Zhong-Xuan Li
- College of Bioengineering, Sichuan University of Science & Engineering, Zigong 643000, China;
| | - Zaheer Abbas
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Tian-Tian Yang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Meng-Yao Liu
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Pei-Yao Chen
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Zheng-Chang Hua
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Xiao-Na Yan
- College of Animal Science and Technology, Hebei Normal University of Science & Technology, Qinhuangdao 066004, China;
| | - Qiang Cheng
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Marhaba Ahmat
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Jun-Yong Wang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| | - Lu-Lu Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; (L.-L.Z.); (X.-B.W.)
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xu-Biao Wei
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; (L.-L.Z.); (X.-B.W.)
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiu-Dong Liao
- Mineral Nutrition Research Division, State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China;
| | - Ri-Jun Zhang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (H.-N.G.); (Y.-C.T.); (H.-L.W.); (J.Z.); (Z.A.); (T.-T.Y.); (M.-Y.L.); (P.-Y.C.); (Z.-C.H.); (Q.C.); (M.A.); (J.-Y.W.)
| |
Collapse
|
28
|
Jadi PK, Sharma P, Bhogapurapu B, Roy S. Alternative Therapeutic Interventions: Antimicrobial Peptides and Small Molecules to Treat Microbial Keratitis. Front Chem 2021; 9:694998. [PMID: 34458234 PMCID: PMC8386189 DOI: 10.3389/fchem.2021.694998] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/02/2021] [Indexed: 01/10/2023] Open
Abstract
Microbial keratitis is a leading cause of blindness worldwide and results in unilateral vision loss in an estimated 2 million people per year. Bacteria and fungus are two main etiological agents that cause corneal ulcers. Although antibiotics and antifungals are commonly used to treat corneal infections, a clear trend with increasing resistance to these antimicrobials is emerging at rapid pace. Extensive research has been carried out to determine alternative therapeutic interventions, and antimicrobial peptides (AMPs) are increasingly recognized for their clinical potential in treating infections. Small molecules targeted against virulence factors of the pathogens and natural compounds are also explored to meet the challenges and growing demand for therapeutic agents. Here we review the potential of AMPs, small molecules, and natural compounds as alternative therapeutic interventions for the treatment of corneal infections to combat antimicrobial resistance. Additionally, we have also discussed about the different formats of drug delivery systems for optimal administration of drugs to treat microbial keratitis.
Collapse
Affiliation(s)
- Praveen Kumar Jadi
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | - Prerana Sharma
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
- Department of Animal Sciences, University of Hyderabad, Hyderabad, India
| | - Bharathi Bhogapurapu
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | - Sanhita Roy
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| |
Collapse
|
29
|
Mangmee S, Reamtong O, Kalambaheti T, Roytrakul S, Sonthayanon P. Antimicrobial Peptide Modifications against Clinically Isolated Antibiotic-Resistant Salmonella. Molecules 2021; 26:molecules26154654. [PMID: 34361810 PMCID: PMC8348142 DOI: 10.3390/molecules26154654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 11/16/2022] Open
Abstract
Antimicrobial peptides are promising molecules to address the global antibiotic resistance problem, however, optimization to achieve favorable potency and safety is required. Here, a peptide-template modification approach was employed to design physicochemical variants based on net charge, hydrophobicity, enantiomer, and terminal group. All variants of the scorpion venom peptide BmKn-2 with amphipathic α-helical cationic structure exhibited an increased antibacterial potency when evaluated against multidrug-resistant Salmonella isolates at a MIC range of 4–8 µM. They revealed antibiofilm activity in a dose-dependent manner. Sheep red blood cells were used to evaluate hemolytic and cell selectivity properties. Peptide Kn2-5R-NH2, dKn2-5R-NH2, and 2F-Kn2-5R-NH2 (variants with +6 charges carrying amidated C-terminus) showed stronger antibacterial activity than Kn2-5R (a variant with +5 charges bearing free-carboxyl group at C-terminus). Peptide dKn2-5R-NH2 (d-enantiomer) exhibited slightly weaker antibacterial activity with much less hemolytic activity (higher hemolytic concentration 50) than Kn2-5R-NH2 (l-enantiomer). Furthermore, peptide Kn2-5R with the least hydrophobicity had the lowest hemolytic activity and showed the highest specificity to Salmonella (the highest selectivity index). This study also explained the relationship of peptide physicochemical properties and bioactivities that would fulfill and accelerate progress in peptide antibiotic research and development.
Collapse
Affiliation(s)
- Suthee Mangmee
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (S.M.); (O.R.)
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (S.M.); (O.R.)
| | - Thareerat Kalambaheti
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, Pathumthani 12120, Thailand;
| | - Piengchan Sonthayanon
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand; (S.M.); (O.R.)
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Correspondence: or ; Tel.: +66-2-354-9100 (ext. 1440)
| |
Collapse
|
30
|
Almeida CV, de Oliveira CFR, Dos Santos EL, Dos Santos HF, Júnior EC, Marchetto R, da Cruz LA, Ferreira AMT, Gomes VM, Taveira GB, Costa BO, Franco OL, Cardoso MH, Macedo MLR. Differential interactions of the antimicrobial peptide, RQ18, with phospholipids and cholesterol modulate its selectivity for microorganism membranes. Biochim Biophys Acta Gen Subj 2021; 1865:129937. [PMID: 34052310 DOI: 10.1016/j.bbagen.2021.129937] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/15/2021] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Antimicrobial peptides (AMPs) are molecules with potential application for the treatment of microorganism infections. We, herein, describe the structure, activity, and mechanism of action of RQ18, an α-helical AMP that displays antimicrobial activity against Gram-positive and Gram-negative bacteria, and yeasts from the Candida genus. METHODS A physicochemical-guided design assisted by computer tools was used to obtain our lead peptide candidate, named RQ18. This peptide was assayed against Gram-positive and Gram-negative bacteria, yeasts, and mammalian cells to determine its selectivity index. The secondary structure and the mechanism of action of RQ18 were investigated using circular dichroism, large unilamellar vesicles, and molecular dynamic simulations. RESULTS RQ18 was not cytotoxic to human lung fibroblasts, peripheral blood mononuclear cells, red blood cells, or Vero cells at MIC values, exhibiting a high selectivity index. Circular dichroism analysis and molecular dynamic simulations revealed that RQ18 presents varying structural profiles in aqueous solution, TFE/water mixtures, SDS micelles, and lipid bilayers. The peptide was virtually unable to release carboxyfluorescein from large unilamellar vesicles composed of POPC/cholesterol, model that mimics the eukaryotic membrane, indicating that vesicles' net charges and the presence of cholesterol may be related with RQ18 selectivity for bacterial and fungal cell surfaces. CONCLUSIONS RQ18 was characterized as a membrane-active peptide with dual antibacterial and antifungal activities, without compromising mammalian cells viability, thus reinforcing its therapeutic application. GENERAL SIGNIFICANCE These results provide further insight into the complex process of AMPs interaction with biological membranes, in special with systems that mimic prokaryotic and eukaryotic cell surfaces.
Collapse
Affiliation(s)
- Claudiane V Almeida
- Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Caio F R de Oliveira
- Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil; Universidade Federal da Grande Dourados, Dourados, Mato Grosso do Sul, Brazil; Oncolytic Anticancer Drugs, Dourados, Mato Grosso do Sul, Brazil
| | - Edson L Dos Santos
- Universidade Federal da Grande Dourados, Dourados, Mato Grosso do Sul, Brazil
| | - Helder F Dos Santos
- Universidade Federal da Grande Dourados, Dourados, Mato Grosso do Sul, Brazil
| | - Edson C Júnior
- Instituto de Química, Departamento de Bioquímica e Química Tecnológica, Universidade Estadual Paulista Júlio de Mesquita Filho, Araraquara, São Paulo, Brazil
| | - Reinaldo Marchetto
- Instituto de Química, Departamento de Bioquímica e Química Tecnológica, Universidade Estadual Paulista Júlio de Mesquita Filho, Araraquara, São Paulo, Brazil
| | - Leticia A da Cruz
- Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Alda Maria T Ferreira
- Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Valdirene M Gomes
- Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Gabriel B Taveira
- Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Bruna O Costa
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Octávio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Marlon H Cardoso
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Maria Lígia R Macedo
- Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil.
| |
Collapse
|
31
|
Silveira GGOS, Torres MDT, Ribeiro CFA, Meneguetti BT, Carvalho CME, de la Fuente-Nunez C, Franco OL, Cardoso MH. Antibiofilm Peptides: Relevant Preclinical Animal Infection Models and Translational Potential. ACS Pharmacol Transl Sci 2021; 4:55-73. [PMID: 33615161 DOI: 10.1021/acsptsci.0c00191] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Indexed: 12/21/2022]
Abstract
Biofilm-forming bacteria may be 10-1000 times more resistant to antibiotics than planktonic bacteria and represent about 75% of bacterial infections in humans. Antibiofilm treatments are scarce, and no effective therapies have been reported so far. In this context, antibiofilm peptides (ABPs) represent an exciting class of agents with potent activity against biofilms both in vitro and in vivo. Moreover, murine models of bacterial biofilm infections have been used to evaluate the in vivo effectiveness of ABPs. Therefore, here we highlight the translational potential of ABPs and provide an overview of the different clinically relevant murine models to assess ABP efficacy, including wound, foreign body, chronic lung, and oral models of infection. We discuss key challenges to translate ABPs to the clinic and the pros and cons of the existing murine biofilm models for reliable assessment of the efficacy of ABPs.
Collapse
Affiliation(s)
- Gislaine G O S Silveira
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil
| | - Marcelo D T Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Camila F A Ribeiro
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil
| | - Beatriz T Meneguetti
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil
| | - Cristiano M E Carvalho
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Octávio L Franco
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal 71966-700, Brazil
| | - Marlon H Cardoso
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal 71966-700, Brazil
| |
Collapse
|
32
|
Wojda I, Staniec B, Sułek M, Kordaczuk J. The greater wax moth Galleria mellonella: biology and use in immune studies. Pathog Dis 2020; 78:ftaa057. [PMID: 32970818 PMCID: PMC7683414 DOI: 10.1093/femspd/ftaa057] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/28/2020] [Indexed: 01/04/2023] Open
Abstract
The greater wax moth Galleria mellonella is an invertebrate that is increasingly being used in scientific research. Its ease of reproduction, numerous offspring, short development cycle, and finally, its known genome and immune-related transcriptome provide a convenient research model for investigation of insect immunity at biochemical and molecular levels. Galleria immunity, consisting of only innate mechanisms, shows adaptive plasticity, which has recently become the subject of intensive scientific research. This insect serves as a mini host in studies of the pathogenicity of microorganisms and in vivo tests of the effectiveness of single virulence factors as well as new antimicrobial compounds. Certainly, the Galleria mellonella species deserves our attention and appreciation for its contribution to the development of research on innate immune mechanisms. In this review article, we describe the biology of the greater wax moth, summarise the main advantages of using it as a model organism and present some of the main techniques facilitating work with this insect.
Collapse
Affiliation(s)
- Iwona Wojda
- Maria Curie Sklodowska University, Institute of Biological Sciences, Department of Immunobiology, Akademicka 19, 20-033 Lublin, Poland
| | - Bernard Staniec
- Maria Curie Sklodowska University, Institute of Biological Sciences, Department of Zoology and Nature Protection, Akademicka 19, 20-033 Lublin, Poland
| | - Michał Sułek
- Maria Curie Sklodowska University, Institute of Biological Sciences, Department of Immunobiology, Akademicka 19, 20-033 Lublin, Poland
| | - Jakub Kordaczuk
- Maria Curie Sklodowska University, Institute of Biological Sciences, Department of Immunobiology, Akademicka 19, 20-033 Lublin, Poland
| |
Collapse
|
33
|
Antifungal In Vitro Activity of Pilosulin- and Ponericin-Like Peptides from the Giant Ant Dinoponera quadriceps and Synergistic Effects with Antimycotic Drugs. Antibiotics (Basel) 2020; 9:antibiotics9060354. [PMID: 32585881 PMCID: PMC7344683 DOI: 10.3390/antibiotics9060354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/17/2020] [Accepted: 06/20/2020] [Indexed: 02/06/2023] Open
Abstract
Venoms from ants comprise a rich source of bioactive peptides, including antimicrobial peptides. From the proteome and peptidome of the giant ant Dinoponera quadriceps venom, members of five known classes of antimicrobial peptides were disclosed (e.g., dermaseptin-, defensin-, ICK-, pilosulin- and ponericin-like types). Based on comparative analysis, these family members have structural determinants that indicate they could display antimicrobial activities. In previous works, pilosulin- and ponericin-like peptides were demonstrated to be active against bacteria, fungi, and parasites. Herein, the antifungal activity of ponericin- and pilosulin-like peptides were assessed, aiming at the expansion of the knowledge about AMPs in predatory ants and the development of new microbicide strategies to deal with difficult-to-treat fungal infections. Synthetic pilosulin- (Dq-2562, Dq-1503, and Dq-1319) and ponericin-like (Dq-3162) peptides were evaluated for their fungicide and fungistatic activities against different species of Candida, including a drug-resistant clinical strain. The MICs and MLCs were determined for all peptides individually and in combination with general antifungal drugs by the microdilution method. The time-kill kinetic curves were set up by means of a luminescent reagent, of which the light signal is proportional to the number of viable cells. The candicidal synergism observed by the combination of subinhibitory concentrations of peptides and general antimycotic drugs were quantified by the checkerboard test and fluorescent dye permeation assay. The influence of ergosterol on the antifungal activity was verified by supplementation of culture medium. The pilosulin- (Dq-2562 and Dq-1503) and ponericin-like (Dq-3162) were the most active peptides, displaying a broad spectrum of antifungal activity in vitro, with MICs in the range of 0.625 to 10 µM. The combination of peptides and conventional antimycotic drugs displayed a synergistic reduction in the MIC values of individual peptides and drugs, while soluble ergosterol in the culture medium increased the MICs. The fungicide and fungistatic activity of the individual peptides and peptides in combination with antimycotics were time-dependent with a rapid onset of action and long-lasting effect, which involved membrane disruption as an underlying mechanism of their action. Altogether, pilosulin- and ponericin-like peptides from the giant ant D. quadriceps venom display a broad-spectrum of candicidal activity, what allows their inclusion in the row of the antifungal peptides and gives support for further studies on the development of strategies to fight candidiasis.
Collapse
|