1
|
Salnikov E, Adélaïde M, Ramos-Martín F, Saad A, Schauer J, Cremanns M, Rima M, Aisenbrey C, Oueslati S, Naas T, Pfennigwerth N, Gatermann S, Sarazin C, Bechinger B, D'Amelio N. Cathelicidin-BF: A Potent Antimicrobial Peptide Leveraging Charge and Phospholipid Recruitment against Multidrug-Resistant Clinical Bacterial Isolates. J Am Chem Soc 2025; 147:11199-11215. [PMID: 40126422 DOI: 10.1021/jacs.4c17821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Cathelicidin-BF (CatBF) is a LL-37 homologous antimicrobial peptide (AMP) isolated from Bungarus fasciatus with an exceptional portfolio of antimicrobial, antiviral, antifungal, and anticancer activities. Contrary to many AMPs, it showed a good pharmacological profile with a half-life of at least 1 h in serum and efficacy against bacterial infections in mice. To evaluate its potential against resistant nosocomial infections, we assessed its activity against 81 clinically relevant resistant bacterial isolates. CatBF exhibited minimum inhibitory concentrations (MICs) as low as 0.5 μM against carbapenem-resistant Acinetobacter baumannii, Klebsiella pneumoniae, and Escherichia coli. Its wide-ranging activity, unaffected by resistance mechanisms or Gram phenotype, prompted us to investigate its molecular mode of action. NMR spectroscopy, paramagnetic probes, and molecular dynamics (MD) simulations were employed to define its structure, penetration depth, and orientation in various membrane models, including micelles, bicelles, oriented bilayers, and vesicles. We found that CatBF's potent activity relies on its strong charge, allowing membrane neutralization at low peptide/lipid ratios and selective recruitment of charged phospholipids. At higher concentrations, a change in peptide orientation reveals membrane invagination and the formation of transient pores possibly leading to bacterial death. Our findings highlight the potential of CatBF as a model for developing resistance-independent agents to combat multidrug-resistant (MDR) bacterial infections.
Collapse
Affiliation(s)
- Evgeniy Salnikov
- Unité de Génie Enzymatique et Cellulaire, UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens 80039, France
- Institut de Chimie, Université de Strasbourg/CNRS, UMR7177, Strasbourg, 67008, France
| | - Morgane Adélaïde
- Unité de Génie Enzymatique et Cellulaire, UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens 80039, France
| | - Francisco Ramos-Martín
- Unité de Génie Enzymatique et Cellulaire, UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens 80039, France
| | - Ahmad Saad
- Institut de Chimie, Université de Strasbourg/CNRS, UMR7177, Strasbourg, 67008, France
| | - Jennifer Schauer
- Department of Medical Microbiology, Ruhr-University, Bochum, 44801, Germany
| | - Martina Cremanns
- Department of Medical Microbiology, Ruhr-University, Bochum, 44801, Germany
| | - Mariam Rima
- Team RESIST, INSERM U1184, Université Paris-Saclay, Faculté de Médecine, Bacteriology ward, Hôpital de Bicêtre, 3ème étage, 78 rue du Gal Leclerc, Paris 94270, France
| | - Christopher Aisenbrey
- Institut de Chimie, Université de Strasbourg/CNRS, UMR7177, Strasbourg, 67008, France
| | - Saoussen Oueslati
- Team RESIST, INSERM U1184, Université Paris-Saclay, Faculté de Médecine, Bacteriology ward, Hôpital de Bicêtre, 3ème étage, 78 rue du Gal Leclerc, Paris 94270, France
| | - Thierry Naas
- Team RESIST, INSERM U1184, Université Paris-Saclay, Faculté de Médecine, Bacteriology ward, Hôpital de Bicêtre, 3ème étage, 78 rue du Gal Leclerc, Paris 94270, France
| | - Niels Pfennigwerth
- Department of Medical Microbiology, Ruhr-University, Bochum, 44801, Germany
| | - Söeren Gatermann
- Department of Medical Microbiology, Ruhr-University, Bochum, 44801, Germany
| | - Catherine Sarazin
- Unité de Génie Enzymatique et Cellulaire, UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens 80039, France
| | - Burkhard Bechinger
- Institut de Chimie, Université de Strasbourg/CNRS, UMR7177, Strasbourg, 67008, France
- Institut Universitaire de France, Paris 75005, France
| | - Nicola D'Amelio
- Unité de Génie Enzymatique et Cellulaire, UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens 80039, France
| |
Collapse
|
2
|
Liu J, Li A, Li Y, Li J, Geng X, Wan J, Lu Q, Wang Q, Wang M, Yang J. Identification and Characterization of a Novel GAPDH-Derived Antimicrobial Peptide From Jellyfish. J Pept Sci 2025; 31:e70011. [PMID: 40045697 PMCID: PMC11883197 DOI: 10.1002/psc.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025]
Abstract
Marine organisms serve as a rich source of bioactive natural compounds, including antimicrobial agents. Jellyfish, which are ancient marine invertebrates with hundreds of millions of years of evolutionary history, have been in continuous contact with a diverse array of pathogenic microorganisms from seawater, which may give rise to a distinctive innate immune system and related defensive molecules. However, it is difficult and inefficient to isolate active ingredients directly from jellyfish for enrichment, though few jellyfish-sourced antimicrobial peptides (AMPs) have been reported. In this study, we utilized transcriptomic big data with bioinformatic tools to dig deeper into potential antimicrobial components in jellyfish, and identified a new AMP JFP-2826 from Rhopilema esculentum. The 20-mer peptide exhibited an alpha-helix structure and showed antimicrobial activity against selected bacterial strains; more importantly, JFP-2826 demonstrated good selectivity for marine-specific Vibrio including Vibrio vulnificus. Sequence analysis of the full-length protein of JFP-2826 revealed that it is derived from the housekeeping gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH), which is probably produced through enzymatic cleavage of the N-terminal fragment. This suggests that GAPDH of jellyfish might have a newly discovered antimicrobial-related function that is conducted by releasing JFP-2826-like cryptic peptides. JFP-2826 can be subjected to further structural modifications and optimizations to potentially become a potent lead peptide for the development of novel antimicrobial drugs treating infections of marine pathogens.
Collapse
Affiliation(s)
- Jingwen Liu
- School of PharmacyBengbu Medical UniversityBengbuChina
- Naval Medical CenterNaval Medical UniversityShanghaiChina
| | - An Li
- Department of Wound Infection and Drug, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center (Daping Hospital)Army Medical UniversityChongqingChina
| | - Yueyue Li
- Naval Medical CenterNaval Medical UniversityShanghaiChina
| | - Jing Li
- The Third Department of Convalescence of Beidaihe Rehabilitation and Convalescence Center of PLAQinhuangdaoChina
| | - Xiaoyu Geng
- Naval Medical CenterNaval Medical UniversityShanghaiChina
| | - Junyi Wan
- Naval Medical CenterNaval Medical UniversityShanghaiChina
| | - Qianqian Lu
- Naval Medical CenterNaval Medical UniversityShanghaiChina
| | - Qingqing Wang
- School of PharmacyBengbu Medical UniversityBengbuChina
| | - Mingke Wang
- Naval Medical CenterNaval Medical UniversityShanghaiChina
| | - Jishun Yang
- Naval Medical CenterNaval Medical UniversityShanghaiChina
| |
Collapse
|
3
|
Kanaujia KA, Wagh S, Pandey G, Phatale V, Khairnar P, Kolipaka T, Rajinikanth PS, Saraf SA, Srivastava S, Kumar S. Harnessing marine antimicrobial peptides for novel therapeutics: A deep dive into ocean-derived bioactives. Int J Biol Macromol 2025; 307:142158. [PMID: 40107127 DOI: 10.1016/j.ijbiomac.2025.142158] [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: 01/06/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Marine antimicrobial peptides (AMPs) are potent bioactive compounds with broad-spectrum activity against bacteria, viruses, and fungi, offering a promising alternative to traditional antibiotics. These small, cationic, and amphiphilic peptides (3-50 amino acids) are key components of marine organisms' immune defenses, adapted to harsh oceanic environments. Discovered in the 1980s, marine AMPs have garnered interest for their unique structures and potential applications in human health. However, despite the ocean's vast biodiversity, they remain underexplored compared to land-based AMPs. This review emphasizes the therapeutic potential of marine AMPs, including their modes of action, structural variety, and applications in drug development, tissue regeneration, and cancer treatment. Moreover, it discusses their antibacterial, antiviral, antifungal, and antiparasitic properties. Additionally, the review addresses strategies to enhance the therapeutic potential of marine AMPs and the challenges associated with their development. By examining the promising future of marine AMPs, this review aims to pave the way for new approaches to combat antimicrobial resistance and develop innovative treatments for various infectious diseases. The potential of marine AMPs as the "medicine bank of the new millennium" remains vast, providing a valuable resource for future drug discovery and sustainable practices across industries.
Collapse
Affiliation(s)
- Kunal Agam Kanaujia
- Institute of Pharmacy, Dr Rammanohar Lohia Avadh University, Ayodhya, Uttar Pradesh 224133, India
| | - Suraj Wagh
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Tejaswini Kolipaka
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India
| | - Shubhini A Saraf
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli 226002, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Shailendra Kumar
- Department of Microbiology, Dr Rammanohar Lohia Avadh University, Ayodhya, Uttar Pradesh 224133, India.
| |
Collapse
|
4
|
Gajic I, Kekic D, Jankovic M, Tomic N, Skoric M, Petrovic M, Mitic Culafic D, Opavski N, Ristivojevic P, Krstic Ristivojevic M, Lukovic B. Nature's Arsenal: Uncovering Antibacterial Agents Against Antimicrobial Resistance. Antibiotics (Basel) 2025; 14:253. [PMID: 40149065 PMCID: PMC11939603 DOI: 10.3390/antibiotics14030253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Antimicrobial resistance (AMR) poses a significant public health threat, leading to increased mortality. The World Health Organization has established a priority list highlighting critical multidrug-resistant (MDR) pathogens that demand urgent research on antimicrobial treatments. Considering this and the fact that new antibiotics are only sporadically approved, natural antibacterial agents have seen a resurgence in interest as potential alternatives to conventional antibiotics and chemotherapeutics. Natural antibacterials, derived from microorganisms, higher fungi, plants, animals, natural minerals, and food sources, offer diverse mechanisms of action against MDR pathogens. Here, we present a comprehensive summary of antibacterial agents from natural sources, including a brief history of their application and highlighting key strategies for using microorganisms (microbiopredators, such as bacteriophages), plant extracts and essential oils, minerals (e.g., silver and copper), as well as compounds of animal origin, such as milk or even venoms. The review also addresses the role of prebiotics, probiotics, and antimicrobial peptides, as well as novel formulations such as nanoparticles. The mechanisms of action of these compounds, such as terpenoids, alkaloids, and phenolic compounds, are explored alongside the challenges for their application, e.g., extraction, formulation, and pharmacokinetics. Conclusions: Future research should focus on developing eco-friendly, sustainable antimicrobial agents and validating their safety and efficacy through clinical trials. Clear regulatory frameworks are essential for integrating these agents into clinical practice. Despite challenges, natural sources offer transformative potential for combating AMR and promoting sustainable health solutions.
Collapse
Affiliation(s)
- Ina Gajic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.K.); (M.J.); (M.S.); (N.O.)
| | - Dusan Kekic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.K.); (M.J.); (M.S.); (N.O.)
| | - Marko Jankovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.K.); (M.J.); (M.S.); (N.O.)
| | - Nina Tomic
- Group for Biomedical Engineering and Nanobiotechnology, Institute of Technical Sciences of SASA, Kneza Mihaila 35/IV, 11000 Belgrade, Serbia;
| | - Mila Skoric
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.K.); (M.J.); (M.S.); (N.O.)
| | - Milos Petrovic
- University Clinical Hospital Center “Dr. Dragisa Misovic-Dedinje”, Heroja Milana Tepića, 1, 11040 Belgrade, Serbia;
| | | | - Natasa Opavski
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.K.); (M.J.); (M.S.); (N.O.)
| | - Petar Ristivojevic
- Department of Analytical Chemistry, Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11158 Belgrade, Serbia;
| | - Maja Krstic Ristivojevic
- Department of Biochemistry, Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11158 Belgrade, Serbia;
| | - Bojana Lukovic
- College of Health Sciences, Academy of Applied Studies Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
5
|
Ye Z, Xu Z, Ouyang J, Shi W, Li S, Wang X, Lu B, Wang K, Wang Y. Improving the Stability and Anti-Infective Activity of Sea Turtle AMPs Using Multiple Structural Modification Strategies. J Med Chem 2024; 67:22104-22123. [PMID: 39636182 DOI: 10.1021/acs.jmedchem.4c02039] [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: 12/07/2024]
Abstract
Antimicrobial peptides (AMPs) are regarded as promising candidates for combating antimicrobial resistance. Previously we identified an AMP named Cm-CATH2 from the green sea turtle, which exhibited potent antibacterial activity and attractive potential in application. However, natural AMPs including Cm-CATH2 frequently suffer from structural instability and sensitivity to physiological conditions, limiting their effectiveness. Herein, we explored various strategies to enhance the efficacy and stability of Cm-CATH2, including peptide truncation, non-natural amino acid substitutions, disulfide bond-based cyclization, and stapled peptide techniques. The results demonstrated that the truncated NCM4 significantly improved the antimicrobial capability of Cm-CATH2 while also enhancing its anti-inflammatory and antibiofilm activities with minimal cytotoxicity. Further ornithine-substituted peptide oNCM markedly enhanced the stability of NCM4 without compromising its antimicrobial efficacy. This study successfully designed a lead peptide oNCM with significant development potential, while providing valuable insights into the advantages and limitations associated with diverse strategies for enhancing the stability of AMPs.
Collapse
Affiliation(s)
- Zifan Ye
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhouye Xu
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jianhong Ouyang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wenzhuang Shi
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shuangyu Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xu Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Binjuan Lu
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kang Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yipeng Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Chai J, Wu J, Yang J, Ye T, Gao Y, Zeng B, Xiong W, Kotsyfakis M, Dijkgraaf I, Liu J, Chen X, Xu X. Cath-HG improves the survival rates and symptoms in LPS-induced septic mice due to its multifunctional properties. Int Immunopharmacol 2024; 143:113332. [PMID: 39395379 DOI: 10.1016/j.intimp.2024.113332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024]
Abstract
The clinical syndrome of sepsis arises from severe infection, triggering an abnormal immune response that can lead to multiple organ dysfunction and ultimately the death of the host. Current therapies for sepsis are often limited in efficacy and fail to target the complex interplay of infection, inflammation and coagulation, leading to high mortality rates, which underscores the urgent need for novel therapeutics to combat sepsis. We previously identified Cath-HG, a compound capable of alleviating platelet dysfunction by suppressing GPVI-mediated platelet activation, thereby improving the survival of septic mice subjected to cecal ligation and puncture. Here, we further explored the antimicrobial, anti-inflammatory, LPS-neutralizing and anticoagulant properties of Cath-HG, as well as its protective effects in LPS-induced septic mice. Our results demonstrated that Cath-HG can bind to LPS, aggregate bacteria, and disrupt bacterial cell membranes, subsequently resulting in microbial death. Unlike most other Cathelicidins, Cath-HG displayed anticoagulation properties by regulating the enzymes plasmin, thrombin, β-tryptase, chymase and tissue plasminogen activator. In septic mice, Cath-HG provided protection against sepsis induced by LPS injection and exhibited bactericidal killing, LPS neutralization and inhibition of coagulation and MAPK signal transduction. Furthermore, Cath-HG obviously reduced the expression of pro-inflammatory cytokines and improved the pathological manifestations of tissue injury across multiple organs. Thus, Cath-HG emerges as a promising drug candidate for protecting against sepsis.
Collapse
Affiliation(s)
- Jinwei Chai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jiena Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianxi Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tiaofei Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yihan Gao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Baishuang Zeng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weichen Xiong
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Michail Kotsyfakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013 Heraklion, Crete, Greece
| | - Ingrid Dijkgraaf
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, Netherlands
| | - Junfang Liu
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Xueqing Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
7
|
Dzurová L, Holásková E, Pospíšilová H, Schneider Rauber G, Frébortová J. Cathelicidins: Opportunities and Challenges in Skin Therapeutics and Clinical Translation. Antibiotics (Basel) 2024; 14:1. [PMID: 39858288 PMCID: PMC11762488 DOI: 10.3390/antibiotics14010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/27/2025] Open
Abstract
Cathelicidins are a group of cationic, amphipathic peptides that play a vital role in the innate immune response of many vertebrates, including humans. Produced by immune and epithelial cells, they serve as natural defenses against a wide range of pathogens, including bacteria, viruses, and fungi. In humans, the cathelicidin LL-37 is essential for wound healing, maintaining skin barrier integrity, and combating infections. Cathelicidins of different origins have shown potential in treating various skin conditions, including melanoma, acne, and diabetic foot ulcers. Despite their promising therapeutic potential, cathelicidins face significant challenges in clinical application. Many peptide-based therapies have failed in clinical trials due to unclear efficacy and safety concerns. Additionally, the emergence of bacterial resistance, which contradicts initial claims of non-resistance, further complicates their development. To successfully translate cathelicidins into effective clinical treatments, therefore, several obstacles must be addressed, including a better understanding of their mechanisms of action, sustainable large-scale production, optimized formulations for drug delivery and stability, and strategies to overcome microbial resistance. This review examines the current knowledge of cathelicidins and their therapeutic applications and discusses the challenges that hinder their clinical use and must be overcome to fully exploit their potential in medicine.
Collapse
Affiliation(s)
- Lenka Dzurová
- Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, 77900 Olomouc, Czech Republic; (E.H.); (H.P.); (J.F.)
| | | | | | | | | |
Collapse
|
8
|
Michira BB, Wang Y, Mwangi J, Wang K, Asmamaw D, Tadese DA, Gao J, Khalid M, Lu QM, Lai R, Li J. A Tachyplesin Antimicrobial Peptide from Theraphosidae Spiders with Potent Antifungal Activity Against Cryptococcus neoformans. Microorganisms 2024; 12:2648. [PMID: 39770850 PMCID: PMC11728142 DOI: 10.3390/microorganisms12122648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
The venoms of Theraphosidae spiders have evolved into diverse natural pharmacopeias through selective pressures. Cryptococcus neoformans is a global health threat that frequently causes life-threatening meningitis and fungemia, particularly in immunocompromised patients. In this study, we identify a novel anti-C. neoformans peptide, QS18 (QCFKVCFRKRCFTKCSRS), from the venom gland of China's native spider species Chilobrachys liboensis by utilizing bioinformatic tools. QS18 shares over 50% sequence similarity with tachyplesin peptides, previously identified only in horseshoe crab hemocytes, expanding the known repertoire of the tachyplesin family to terrestrial arachnids. The oxidative folding of QS18 notably enhances its antifungal activity and stability, resulting in a minimum inhibitory concentration of 1.4 µM. The antimicrobial mechanism of QS18 involves cell membrane disruption. QS18 exhibits less than 5% hemolysis in human erythrocytes, indicating microbial selectivity and a favorable safety profile for therapeutic use. Furthermore, mouse model studies highlight QS18's ability as an antifungal agent with notable anti-inflammatory activity. Our study demonstrates QS18 as both a promising template for spider venom peptide research and a novel candidate for the development of peptide antifungals.
Collapse
Affiliation(s)
- Brenda B. Michira
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Yi Wang
- Center for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China;
| | - James Mwangi
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Kexin Wang
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- Medical College of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Demeke Asmamaw
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Dawit Adisu Tadese
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Jinai Gao
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Mehwish Khalid
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Qiu-Min Lu
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Ren Lai
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| | - Juan Li
- Key Laboratory of Genetic Evolution & Animal Models, Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, and Sino-African Joint Research Center, New Cornerstone Science Laboratory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China; (B.B.M.); (J.M.); (K.W.); (D.A.); (D.A.T.); (J.G.); (M.K.); (Q.-M.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China
| |
Collapse
|
9
|
Chatterjee D, Sivashanmugam K. Immunomodulatory peptides: new therapeutic horizons for emerging and re-emerging infectious diseases. Front Microbiol 2024; 15:1505571. [PMID: 39760081 PMCID: PMC11695410 DOI: 10.3389/fmicb.2024.1505571] [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: 10/03/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
The emergence and re-emergence of multi-drug-resistant (MDR) infectious diseases have once again posed a significant global health challenge, largely attributed to the development of bacterial resistance to conventional anti-microbial treatments. To mitigate the risk of drug resistance globally, both antibiotics and immunotherapy are essential. Antimicrobial peptides (AMPs), also referred to as host defense peptides (HDPs), present a promising therapeutic alternative for treating drug-resistant infections due to their various mechanisms of action, which encompass antimicrobial and immunomodulatory effects. Many eukaryotic organisms produce HDPs as a defense mechanism, for example Purothionin from Triticum aestivum plant, Defensins, Cathelicidins, and Histatins from humans and many such peptides are currently the focus of research because of their antibacterial, antiviral and anti-fungicidal properties. This article offers a comprehensive review of the immunomodulatory activities of HDPs derived from eukaryotic organisms including humans, plants, birds, amphibians, reptiles, and marine species along with their mechanisms of action and therapeutic benefits.
Collapse
|
10
|
Cordero Gil TDLÁ, Moleón MS, Marelli BE, Siroski PA. Host defense peptides in crocodilians - A comprehensive review. Peptides 2024; 182:171312. [PMID: 39471969 DOI: 10.1016/j.peptides.2024.171312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/01/2024] [Accepted: 10/23/2024] [Indexed: 11/01/2024]
Abstract
Amphibians and reptiles, like all animals, are prone to periodic infections. However, crocodilians stand out for their remarkable ability to remain generally healthy and infection-free despite frequent exposure to a wide variety of microorganisms in their habitats and often sustaining significant injuries. These animals have evolved highly active immune mechanisms that provide rapid and effective defense. This is evidenced by the superior hemolytic capacity of their plasma compared to that of other organisms. To date, several host defense peptides (HDPs) have been identified in crocodilians, including cathelicidins, beta-defensins, hepcidins, leucrocins, hemocidins, and omwaprins. These peptides exhibit potent and broad-spectrum antimicrobial, antibiofilm, antifungal, and anticancer activities. Due to the relatively low but diverse evolutionary rate of crocodilians, the HDPs found in this species offer valuable insights into proteins and mechanisms of action that are highly conserved across many animals related to immune defense. The potential applications of HDPs in modern medicine represent a promising strategy for developing new therapeutic agents. Their novelty and the vast variability with which peptide sequences can be designed and modified expand the field of application for HDPs almost infinitely. This review addresses the urgent need for innovative and more effective drugs to combat the rise of antimicrobialresistant infections and evaluates the potential of crocodilian HDPs. It presents recent advances in the identification of crocodilian HDPs, particularly antimicrobial peptides (AMPs), including previously underexplored topics such as the sequential and structural conformation of different peptide types in crocodilians and the use of bioinformatics tools to enhance native peptides.
Collapse
Affiliation(s)
- Trinidad de Los Ángeles Cordero Gil
- Laboratorio de Ecología Molecular Aplicada (ICiVET-UNL), CONICET, Esperanza, Santa Fe S3080, Argentina; Laboratorio de Zoología Aplicada: Anexo Vertebrados (FHUC-UNL/MMA), Santa Fe 3000, Argentina; Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), UNL, CONICET, Esperanza, Santa Fe S3080, Argentina.
| | - María Soledad Moleón
- Laboratorio de Ecología Molecular Aplicada (ICiVET-UNL), CONICET, Esperanza, Santa Fe S3080, Argentina; Laboratorio de Zoología Aplicada: Anexo Vertebrados (FHUC-UNL/MMA), Santa Fe 3000, Argentina; Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), UNL, CONICET, Esperanza, Santa Fe S3080, Argentina
| | - Belkis Ester Marelli
- Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), UNL, CONICET, Esperanza, Santa Fe S3080, Argentina
| | - Pablo Ariel Siroski
- Laboratorio de Ecología Molecular Aplicada (ICiVET-UNL), CONICET, Esperanza, Santa Fe S3080, Argentina; Laboratorio de Zoología Aplicada: Anexo Vertebrados (FHUC-UNL/MMA), Santa Fe 3000, Argentina; Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), UNL, CONICET, Esperanza, Santa Fe S3080, Argentina; Ministerio de Medio Ambiente y Cambio Climático, Santa Fe 3000, Argentina
| |
Collapse
|
11
|
Qi WX, Liu F, Liu FF, Ren HY, Zhang BX, Yu XQ, Rao XJ. Discovery and characterization of a novel Lepidoptera-specific antimicrobial peptide from the fall armyworm, Spodoptera frugiperda (Lepidoptera: Noctuidae). INSECT SCIENCE 2024. [PMID: 39552199 DOI: 10.1111/1744-7917.13471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/23/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Antimicrobial peptides (AMPs) are critical components of innate immunity in diverse organisms, including plants, vertebrates, and insects. This study identified and characterized a novel Lepidoptera-specific AMP, named lepidoptin, from the invasive pest Spodoptera frugiperda (Lepidoptera: Noctuidae). Lepidoptin is a 116-amino acid protein containing a signal peptide and a novel β-sandwich domain that is distinct from previously reported AMPs. Temporal and spatial expression analyses revealed a significant upregulation of the lepidoptin gene in vivo and in cultured SF9 cells in response to pathogens. Molecular docking analysis identified a specific binding cavity. Enzyme-linked immunosorbent assay and binding assays confirmed that lepidoptin can bind to pathogen-associated molecular patterns, bacteria, and fungi. Recombinant lepidoptin exhibited potent antibacterial activity by inducing bacterial agglutination, inhibiting bacterial growth, increasing bacterial membrane permeability, and preventing biofilm formation. Lepidoptin also showed antifungal activity against the entomopathogenic fungus Beauveria bassiana by inhibiting spore germination, increasing fungal cell permeability, and increasing reactive oxygen species. Injection of recombinant lepidoptin into S. frugiperda larvae increased survival after B. bassiana infection, whereas knockdown of lepidoptin by RNA interference decreased larval survival. In addition, lepidoptin showed antimicrobial activity against the plant pathogen Fusarium graminearum by inhibiting spore germination and alleviating disease symptoms in wheat seedlings and cherry tomatoes. This study demonstrates the remarkable dual functionality of lepidoptin in enhancing S. frugiperda immunity and controlling plant pathogens, making it a promising candidate for biocontrol strategies in both pest management and plant disease prevention.
Collapse
Affiliation(s)
- Wen-Xuan Qi
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, China
- Key Laboratory of Agri-products Quality and Biosafety (Anhui Agricultural University), Ministry of Education, Hefei, China
| | - Feng Liu
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, China
- Key Laboratory of Agri-products Quality and Biosafety (Anhui Agricultural University), Ministry of Education, Hefei, China
| | - Fang-Fang Liu
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, China
- Key Laboratory of Agri-products Quality and Biosafety (Anhui Agricultural University), Ministry of Education, Hefei, China
| | - Hai-Yan Ren
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, China
- Key Laboratory of Agri-products Quality and Biosafety (Anhui Agricultural University), Ministry of Education, Hefei, China
| | - Bang-Xian Zhang
- School of Biological Science and Food Engineering, Chuzhou University, Chuzhou, China
| | - Xiao-Qiang Yu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiang-Jun Rao
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, China
- Key Laboratory of Agri-products Quality and Biosafety (Anhui Agricultural University), Ministry of Education, Hefei, China
| |
Collapse
|
12
|
Samat R, Sen S, Jash M, Ghosh S, Garg S, Sarkar J, Ghosh S. Venom: A Promising Avenue for Antimicrobial Therapeutics. ACS Infect Dis 2024; 10:3098-3125. [PMID: 39137302 DOI: 10.1021/acsinfecdis.4c00314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Venom in medicine is well documented in the chronicles of ancient Greece and the Roman Empire and persisted into the Renaissance and even into the modern era. Venoms were not always associated with detrimental consequences. Since ancient times, the curative capacity of venom has been recognized, portraying venom as a metaphor for pharmacy and medicine. Venom proteins and peptides' antimicrobial potential has not undergone systematic exploration despite the huge literature on natural antimicrobials. In light of the escalating challenge of antimicrobial resistance and the diminishing effectiveness of antibiotics, there is a pressing need for innovative antimicrobials capable of effectively addressing illnesses caused by multidrug-resistant microorganisms. This review adds to our understanding of the effectiveness of different venom components against a host of pathogenic microorganisms. The aim is to illuminate the various antimicrobials present in venom and venom peptides, thereby emphasizing the unexplored medicinal potential for antimicrobial properties. We have presented a concise summary of the molecular examination of the venom peptides' functioning processes, as well as the current clinical and preclinical progress of venom antimicrobial peptides.
Collapse
Affiliation(s)
- Ramkamal Samat
- Smart Healthcare Department, Interdisciplinary Research Platform, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Samya Sen
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
- iHUB Drishti Foundation, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Moumita Jash
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Satyajit Ghosh
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Shubham Garg
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Jayita Sarkar
- Centre for Research and Development of Scientific Instruments (CRDSI), Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| | - Surajit Ghosh
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
- Smart Healthcare Department, Interdisciplinary Research Platform, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
- iHUB Drishti Foundation, Indian Institute of Technology, Jodhpur, Rajasthan 342030, India
| |
Collapse
|
13
|
Ye Z, Fu L, Li S, Chen Z, Ouyang J, Shang X, Liu Y, Gao L, Wang Y. Synergistic collaboration between AMPs and non-direct antimicrobial cationic peptides. Nat Commun 2024; 15:7319. [PMID: 39183339 PMCID: PMC11345435 DOI: 10.1038/s41467-024-51730-x] [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: 01/03/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Non-direct antimicrobial cationic peptides (NDACPs) are components of the animal innate immune system. But their functions and association with antimicrobial peptides (AMPs) are incompletely understood. Here, we reveal a synergistic interaction between the AMP AW1 and the NDACP AW2, which are co-expressed in the frog Amolops wuyiensis. AW2 enhances the antibacterial activity of AW1 both in vitro and in vivo, while mitigating the development of bacterial resistance and eradicating biofilms. AW1 and AW2 synergistically damage bacterial membranes, facilitating cellular uptake and interaction of AW2 with the intracellular target bacterial genomic DNA. Simultaneously, they trigger the generation of ROS in bacteria, contributing to cell death upon reaching a threshold level. Moreover, we demonstrate that this synergistic antibacterial effect between AMPs and NDACPs is prevalent across diverse animal species. These findings unveil a robust and previously unknown correlation between AMPs and NDACPs as a widespread antibacterial immune defense strategy in animals.
Collapse
Affiliation(s)
- Zifan Ye
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Lei Fu
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Shuangyu Li
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Ziying Chen
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Jianhong Ouyang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xinci Shang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yanli Liu
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Lianghui Gao
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China.
| | - Yipeng Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.
| |
Collapse
|
14
|
Hernández-Arvizu EE, Asada M, Kawazu SI, Vega CA, Rodríguez-Torres A, Morales-García R, Pavón-Rocha AJ, León-Ávila G, Rivas-Santiago B, Mosqueda J. Antiparasitic Evaluation of Aquiluscidin, a Cathelicidin Obtained from Crotalus aquilus, and the Vcn-23 Derivative Peptide against Babesia bovis, B. bigemina and B. ovata. Pathogens 2024; 13:496. [PMID: 38921794 PMCID: PMC11206629 DOI: 10.3390/pathogens13060496] [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: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Babesiosis is a growing concern due to the increased prevalence of this infectious disease caused by Babesia protozoan parasites, affecting various animals and humans. With rising worries over medication side effects and emerging drug resistance, there is a notable shift towards researching babesiacidal agents. Antimicrobial peptides, specifically cathelicidins known for their broad-spectrum activity and immunomodulatory functions, have emerged as potential candidates. Aquiluscidin, a cathelicidin from Crotalus aquilus, and its derivative Vcn-23, have been of interest due to their previously observed antibacterial effects and non-hemolytic activity. This work aimed to characterize the effect of these peptides against three Babesia species. Results showed Aquiluscidin's significant antimicrobial effects on Babesia species, reducing the B. bigemina growth rate and exhibiting IC50 values of 14.48 and 20.70 μM against B. ovata and B. bovis, respectively. However, its efficacy was impacted by serum presence in culture, and it showed no inhibition against a B. bovis strain grown in serum-supplemented medium. Conversely, Vcn-23 did not demonstrate babesiacidal activity. In conclusion, Aquiluscidin shows antibabesia activity in vitro and its efficacy is affected by the presence of serum in the culture medium. Nevertheless, this peptide represents a candidate for further investigation of its antiparasitic properties and provides insights into potential alternatives for the treatment of babesiosis.
Collapse
Affiliation(s)
- Edwin Esaú Hernández-Arvizu
- Immunology and Vaccine Research Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico; (E.E.H.-A.); (R.M.-G.); (A.J.P.-R.)
- PhD Program in Natural Sciences, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico
| | - Masahito Asada
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medcine, Inadacho, Nishi 2-13, Obihiro 080-8555, Hokkaido, Japan; (M.A.); (S.-I.K.)
| | - Shin-Ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medcine, Inadacho, Nishi 2-13, Obihiro 080-8555, Hokkaido, Japan; (M.A.); (S.-I.K.)
| | - Carlos Agustín Vega
- Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico; (C.A.V.); (A.R.-T.)
| | - Angelina Rodríguez-Torres
- Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico; (C.A.V.); (A.R.-T.)
| | - Rodrigo Morales-García
- Immunology and Vaccine Research Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico; (E.E.H.-A.); (R.M.-G.); (A.J.P.-R.)
| | - Aldo J. Pavón-Rocha
- Immunology and Vaccine Research Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico; (E.E.H.-A.); (R.M.-G.); (A.J.P.-R.)
| | - Gloria León-Ávila
- Department of Zoology, National School of Biological Sciences, National Polytechnic Institute, Carpio y Plan de Ayala S/N, C.P. 11340, Casco de Santo Tomas, Mexico City 11340, Mexico;
| | - Bruno Rivas-Santiago
- Medical Research Unit Zacatecas-Instituto Mexicano del Seguro Social, Zacatecas 98053, Mexico;
| | - Juan Mosqueda
- Immunology and Vaccine Research Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico; (E.E.H.-A.); (R.M.-G.); (A.J.P.-R.)
| |
Collapse
|
15
|
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
|
16
|
Thomas AM, Antony SP. Marine Antimicrobial Peptides: An Emerging Nightmare to the Life-Threatening Pathogens. Probiotics Antimicrob Proteins 2024; 16:552-578. [PMID: 37022565 DOI: 10.1007/s12602-023-10061-x] [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] [Accepted: 03/08/2023] [Indexed: 04/07/2023]
Abstract
The emergence of multidrug-resistant pathogens due to improper usage of conventional antibiotics has created a global health crisis. Alternatives to antibiotics being an urgent need, the scientific community is forced to search for new antimicrobials. This exploration has led to the discovery of antimicrobial peptides, a group of small peptides occurring in different phyla such as Porifera, Cnidaria, Annelida, Arthropoda, Mollusca, Echinodermata, and Chordata, as a component of their innate immune system. The marine environment, possessing immense diversity of organisms, is undoubtedly one of the richest sources of unique potential antimicrobial peptides. The distinctiveness of marine antimicrobial peptides lies in their broad-spectrum activity, mechanism of action, less cytotoxicity, and high stability, which form the benchmark for developing a potential therapeutic. This review aims to (1) synthesise the available information on the distinctive antimicrobial peptides discovered from marine organisms, particularly over the last decade, and (2) discuss the distinctiveness of marine antimicrobial peptides and their prospects.
Collapse
Affiliation(s)
- Anne Maria Thomas
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - Swapna P Antony
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India.
| |
Collapse
|
17
|
Wang M, Liao Z, Zhangsun D, Wu Y, Luo S. Engineering Enhanced Antimicrobial Properties in α-Conotoxin RgIA through D-Type Amino Acid Substitution and Incorporation of Lysine and Leucine Residues. Molecules 2024; 29:1181. [PMID: 38474693 DOI: 10.3390/molecules29051181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Antimicrobial peptides (AMPs), acknowledged as host defense peptides, constitute a category of predominant cationic peptides prevalent in diverse life forms. This study explored the antibacterial activity of α-conotoxin RgIA, and to enhance its stability and efficacy, D-amino acid substitution was employed, resulting in the synthesis of nine RgIA mutant analogs. Results revealed that several modified RgIA mutants displayed inhibitory efficacy against various pathogenic bacteria and fungi, including Candida tropicalis and Escherichia coli. Mechanistic investigations elucidated that these polypeptides achieved antibacterial effects through the disruption of bacterial cell membranes. The study further assessed the designed peptides' hemolytic activity, cytotoxicity, and safety. Mutants with antibacterial activity exhibited lower hemolytic activity and cytotoxicity, with Pep 8 demonstrating favorable safety in mice. RgIA mutants incorporating D-amino acids exhibited notable stability and adaptability, sustaining antibacterial properties across diverse environmental conditions. This research underscores the potential of the peptide to advance innovative oral antibiotics, offering a novel approach to address bacterial infections.
Collapse
Affiliation(s)
- Minghe Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Zhouyuji Liao
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Dongting Zhangsun
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Yong Wu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Sulan Luo
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| |
Collapse
|
18
|
Li W, Ma Y, Ou L, Xu C, Wei Y, Yang K, Yuan B. Asymmetric disturbance and permeabilization of bilayer membranes by 3-nm carbon dots. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133382. [PMID: 38163412 DOI: 10.1016/j.jhazmat.2023.133382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/15/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
Small-sized fluorescent carbon dots (CDs) are gaining increasing attention in the field of biomedical applications. The environmental and biological compatibility of positively charged CDs has been extensively investigated; however, the potential cytotoxicity caused by negatively and particularly neutrally charged small CDs has been significantly overlooked. In this study, we conducted a comprehensive investigation into the cellular membrane disruption effect of weakly negatively charged 3-nm CDs using a combination of various biophysical techniques. Our findings demonstrate that even at a low concentration of 0.5 μg mL-1, these CDs induce significant perturbations on the cellular membrane, resulting in increased membrane permeability due to asymmetric disruption of the bilayer structure. Furthermore, CDs exhibit distinct mechanisms at different concentrations, including prompt insertion into the bilayer at low concentrations (<20 μg mL-1) and a synergistic effect after a threshold time at high concentrations (e.g., 25-200 μg mL-1). Moreover, these CDs possess specific antibacterial properties against Acinetobacter baumannii (with a minimum inhibitory concentration of 50 μg mL-1) while showing minimal hemolytic or cytotoxic effects on mammalian cells. This study provides comprehensive insights into the biophysical aspects of cellular membrane toxicity caused by small weakly negatively charged CDs and contributes to assessing their potential biomedical applications.
Collapse
Affiliation(s)
- Wenwen Li
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China; Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| | - Yurong Ma
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 215123 Suzhou, China
| | - Luping Ou
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Cheng Xu
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| | - Yushuang Wei
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China; Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China.
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China.
| |
Collapse
|
19
|
Hu C. Marine natural products and human immunity: novel biomedical resources for anti-infection of SARS-CoV-2 and related cardiovascular disease. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:12. [PMID: 38282092 PMCID: PMC10822835 DOI: 10.1007/s13659-024-00432-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/17/2024] [Indexed: 01/30/2024]
Abstract
Marine natural products (MNPs) and marine organisms include sea urchin, sea squirts or ascidians, sea cucumbers, sea snake, sponge, soft coral, marine algae, and microalgae. As vital biomedical resources for the discovery of marine drugs, bioactive molecules, and agents, these MNPs have bioactive potentials of antioxidant, anti-infection, anti-inflammatory, anticoagulant, anti-diabetic effects, cancer treatment, and improvement of human immunity. This article reviews the role of MNPs on anti-infection of coronavirus, SARS-CoV-2 and its major variants (such as Delta and Omicron) as well as tuberculosis, H. Pylori, and HIV infection, and as promising biomedical resources for infection related cardiovascular disease (irCVD), diabetes, and cancer. The anti-inflammatory mechanisms of current MNPs against SARS-CoV-2 infection are also discussed. Since the use of other chemical agents for COVID-19 treatment are associated with some adverse effects in cardiovascular system, MNPs have more therapeutic advantages. Herein, it's time to protect this ecosystem for better sustainable development in the new era of ocean economy. As huge, novel and promising biomedical resources for anti-infection of SARS-CoV-2 and irCVD, the novel potential mechanisms of MNPs may be through multiple targets and pathways regulating human immunity and inhibiting inflammation. In conclusion, MNPs are worthy of translational research for further clinical application.
Collapse
Affiliation(s)
- Chunsong Hu
- Department of Cardiovascular Medicine, Jiangxi Academy of Medical Science, Nanchang University, Hospital of Nanchang University, No. 461 Bayi Ave, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
20
|
Zhong C, Deng X, Jiang A, Liu Y, Liu Y, Fu J, Cao G. The Cell Envelope Integrity Protein Homologue D0Y85_RS06240 of Stenotrophomonas Confers Multiantibiotic Resistance. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:7547514. [PMID: 38283082 PMCID: PMC10821804 DOI: 10.1155/2024/7547514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/19/2023] [Accepted: 01/13/2024] [Indexed: 01/30/2024]
Abstract
Background The potential role of cell envelope integrity proteins in mediating antibiotic resistance is not well understood. In this study, we investigated whether the cell envelope integrity protein D0Y85_RS06240 from the multiantibiotic resistant strain Stenotrophomonas sp. G4 mediates antibiotic resistance. Methods Bioinformatics analysis was conducted to identify proteins related to the D0Y85_RS06240 protein. The D0Y85_RS06240 gene was heterologously expressed in Escherichia coli, both antibiotic MICs and the effect of efflux pump inhibitors on antibiotic MICs were determined by the broth microdilution method. A combination of antibiotic and efflux pump inhibitor was used to investigate bacterial killing kinetics, and binding of D0Y85_RS06240 to antibiotic molecules was predicted by molecular docking analysis. Results Sequence homology analysis revealed that D0Y85_RS06240 was related to cell envelope integrity proteins. The D0Y85_RS06240 heterologous expression strains were resistant to multiple antibiotics, including colistin, tetracycline, and cefixime. However, the efflux pump inhibitor N-methylpyrrolidone (NMP) reduced the antibiotic MICs of the D0Y85_RS06240 heterologous expression strain, and bacterial killing kinetics revealed that NMP enhanced the bactericidal rate of tetracycline to the drug-resistant bacteria. Molecular docking analysis indicated that D0Y85_RS06240 could bind colistin, tetracycline, and cefixime. Conclusion The cell envelope integrity protein D0Y85_RS06240 in Stenotrophomonas sp. G4 mediates multiantibiotic resistance. This study lays the foundation for an in-depth analysis of D0Y85_RS06240-mediated antibiotic resistance mechanisms and the use of D0Y85_RS06240 as a target for the treatment of multiantibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Chuanqing Zhong
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Ji'nan 250101, China
| | - Xiaoqiang Deng
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Ji'nan 250101, China
| | - Aihua Jiang
- Jinan Urban and Rural Water Bureau, Ji'nan 250099, China
| | - Yayu Liu
- Biomedical Sciences College, Shandong Medicinal Biotechnology Centre, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji'nan 250117, China
| | - Yuanyuan Liu
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Ji'nan 250101, China
| | - Jiafang Fu
- Biomedical Sciences College, Shandong Medicinal Biotechnology Centre, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji'nan 250117, China
| | - Guangxiang Cao
- Biomedical Sciences College, Shandong Medicinal Biotechnology Centre, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji'nan 250117, China
| |
Collapse
|
21
|
Hernández-Arvizu EE, Silis-Moreno TM, García-Arredondo JA, Rodríguez-Torres A, Cervantes-Chávez JA, Mosqueda J. Aquiluscidin, a Cathelicidin from Crotalus aquilus, and the Vcn-23 Derivative Peptide, Have Anti-Microbial Activity against Gram-Negative and Gram-Positive Bacteria. Microorganisms 2023; 11:2778. [PMID: 38004789 PMCID: PMC10673557 DOI: 10.3390/microorganisms11112778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Anti-microbial peptides play a vital role in the defense mechanisms of various organisms performing functions that range from the elimination of microorganisms, through diverse mechanisms, to the modulation of the immune response, providing protection to the host. Among these peptides, cathelicidins, a well-studied family of anti-microbial peptides, are found in various animal species, including reptiles. Due to the rise in anti-microbial resistance, these compounds have been suggested as potential candidates for developing new drugs. In this study, we identified and characterized a cathelicidin-like peptide called Aquiluscidin (Aq-CATH) from transcripts obtained from the skin and oral mucosa of the Querétaro's dark rattlesnake, Crotalus aquilus. The cDNA was cloned, sequenced, and yielded a 566-base-pair sequence. Using bioinformatics, we predicted that the peptide precursor contains a signal peptide, a 101-amino-acid conserved cathelin domain, an anionic region, and a 34-amino-acid mature peptide in the C-terminal region. Aq-CATH and a derived 23-amino-acid peptide (Vcn-23) were synthesized, and their anti-microbial activity was evaluated against various species of bacteria in in vitro assays. The minimal inhibitory concentrations against bacteria ranged from 2 to 8 μg/mL for both peptides. Furthermore, at concentrations of up to 50 μM, they exhibited no significant hemolytic activity (<2.3% and <1.2% for Aquiluscidin and Vcn-23, respectively) against rat erythrocytes and displayed no significant cytotoxic activity at low concentrations (>65% cell viability at 25 µM). Finally, this study represents the first identification of an antimicrobial peptide in Crotalus aquilus, which belongs to the cathelicidin family and exhibits the characteristic features of these peptides. Both Aq-CATH and its derived molecule, Vcn-23, displayed remarkable inhibitory activity against all tested bacteria, highlighting their potential as promising candidates for further antimicrobial research.
Collapse
Affiliation(s)
- Edwin Esaú Hernández-Arvizu
- Immunology and Vaccine Research Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico; (E.E.H.-A.)
- Ph.D. Program in Natural Sciences, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico
| | - Teresa Monserrat Silis-Moreno
- Immunology and Vaccine Research Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico; (E.E.H.-A.)
| | - José Alejandro García-Arredondo
- Departamento de Investigación Química y Farmacológica de Productos Naturales, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario S/N, Queretaro 76010, Mexico;
| | - Angelina Rodríguez-Torres
- Natural Sciences College, Autonomous University of Querétaro, Queretaro 76230, Mexico; (A.R.-T.); (J.A.C.-C.)
- Cuerpo Academico Salud Animal y Microbiología Ambiental, Autonomous University of Queretaro, Queretaro 76230, Mexico
| | | | - Juan Mosqueda
- Immunology and Vaccine Research Laboratory, Natural Sciences College, Autonomous University of Queretaro, Queretaro 76230, Mexico; (E.E.H.-A.)
- Cuerpo Academico Salud Animal y Microbiología Ambiental, Autonomous University of Queretaro, Queretaro 76230, Mexico
| |
Collapse
|
22
|
León Madrazo A, Segura Campos MR. Antibacterial properties of peptides from chia (Salvia hispanica L.) applied to pork meat preservation. J Food Sci 2023; 88:4194-4217. [PMID: 37655475 DOI: 10.1111/1750-3841.16754] [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: 05/08/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 09/02/2023]
Abstract
Chia-derived peptides might represent a novel alternative to conventional preservatives in food. Despite the antibacterial potential of these molecules, their food application is still limited. This study aimed to evaluate chia-derived peptides' antibacterial and antibiofilm potential in food preservation. The peptides YACLKVK, KLKKNL, KLLKKYL, and KKLLKI were synthesized, and their antibacterial activity against Listeria monocytogenes, Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, and Salmonella Enteritidis was evaluated through microdilution tests. A bacterial killing kinetic assay determined bacterial growth over time. The ability to prevent and eradicate S. aureus biofilm was assessed by crystal violet staining. The hemolytic and cytotoxic activities were determined in human red blood cells and fibroblasts using free hemoglobin detection and (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assays, respectively. Finally, a microbial challenge was performed on meat samples inoculated with L. monocytogenes and S. Enteritidis to determine their inhibitory effects on pork meat. Results showed the potential antibacterial activity of these peptides, with minimum inhibitory concentrations ranging from 0.23 to 5.58 mg/mL. Biofilm inhibition percentages were above 40%, and eradication percentages were lower than 20%. In vitro assays in human red blood cells and fibroblasts demonstrated that peptides are not hemolytic or cytotoxic agents. In microbiological challenge testing, KKLLKI showed the most promising antibacterial effects against S. Enteritidis on refrigerated pork meat samples. These findings suggest that chia-derived peptides have the potential as natural food preservatives due to their antibacterial and antibiofilm properties. Notably, KKLLKI demonstrated promising antibacterial effects against Salmonella spp. on a complex food matrix, such as pork meat. PRACTICAL APPLICATION: Chia-derived peptides can be a safer alternative to synthetic preservatives in the food industry because the latter may be detrimental to human health. Salmonella spp. growth on chilled pork meat was shown to be inhibited by the peptide KKLLKI, indicating that the use of these peptides may offer a more secure and natural alternative to synthetic preservatives.
Collapse
Affiliation(s)
- Anaí León Madrazo
- Faculty of Chemical Engineering, Autonomous University of Yucatán, Merida, Mexico
| | | |
Collapse
|
23
|
Lu X, Yang M, Zhou S, Yang S, Chen X, Khalid M, Wang K, Fang Y, Wang C, Lai R, Duan Z. Identification and Characterization of RK22, a Novel Antimicrobial Peptide from Hirudinaria manillensis against Methicillin Resistant Staphylococcus aureus. Int J Mol Sci 2023; 24:13453. [PMID: 37686259 PMCID: PMC10487658 DOI: 10.3390/ijms241713453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Staphylococcus aureus (S. aureus) infections are a leading cause of morbidity and mortality, which are compounded by drug resistance. By manipulating the coagulation system, S. aureus gains a significant advantage over host defense mechanisms, with hypercoagulation induced by S. aureus potentially aggravating infectious diseases. Recently, we and other researchers identified that a higher level of LL-37, one endogenous antimicrobial peptide with a significant killing effect on S. aureus infection, resulted in thrombosis formation through the induction of platelet activation and potentiation of the coagulation factor enzymatic activity. In the current study, we identified a novel antimicrobial peptide (RK22) from the salivary gland transcriptome of Hirudinaria manillensis (H. manillensis) through bioinformatic analysis, and then synthesized it, which exhibited good antimicrobial activity against S. aureus, including a clinically resistant strain with a minimal inhibitory concentration (MIC) of 6.25 μg/mL. The RK22 peptide rapidly killed S. aureus by inhibiting biofilm formation and promoting biofilm eradication, with good plasma stability, negligible cytotoxicity, minimal hemolytic activity, and no significant promotion of the coagulation system. Notably, administration of RK22 significantly inhibited S. aureus infection and the clinically resistant strain in vivo. Thus, these findings highlight the potential of RK22 as an ideal treatment candidate against S. aureus infection.
Collapse
Affiliation(s)
- Xiaoyu Lu
- School of Life Sciences, Tianjin University, Tianjin 300072, China;
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
| | - Min Yang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Shengwen Zhou
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Shuo Yang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xiran Chen
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Mehwish Khalid
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Kexin Wang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- School of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Yaqun Fang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
| | - Chaoming Wang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Ren Lai
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650107, China
- National Resource for Non-Human Primates, Kunming Primate Research Center, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650107, China
- Sino-African Joint Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Zilei Duan
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
| |
Collapse
|
24
|
Zhou M, Yao Y, Ma S, Zou M, Chen Y, Cai S, Zhao F, Wu H, Xiao F, Abudushalamu G, Fan X, Wu G. Dual-targeted and dual-sensitive self-assembled protein nanocarrier delivering hVEGI-192 for triple-negative breast cancer. Int J Biol Macromol 2023:125475. [PMID: 37353129 DOI: 10.1016/j.ijbiomac.2023.125475] [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] [Received: 03/22/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/25/2023]
Abstract
Breast cancer is a highly prevalent malignancy worldwide among women with an increasing incidence in recent years. Triple-negative breast cancer (TNBC), a specific type of breast cancer, occurs primarily in young women and exhibits large tumor size, high clinical stage, and extremely poor prognosis with a high rate of lymph node, liver, and lung metastases. TNBC is insensitive to endocrine therapy and trastuzumab treatment, and there is an urgent need for effective therapeutics and treatment guidelines. However, investigations into antiangiogenic agents for the treatment of TNBC are ongoing. In this study, we successfully engineered a self-assembled protein nanocarrier TfRBP9-hVEGI-192-ELP fusion protein (TVEFP) to deliver the therapeutic protein, human vascular endothelial growth inhibitor (hVEGI-192). This was found to be effective in inhibiting tumor angiogenesis in vivo. The protein nanocarrier effectively inhibited the progression of TNBC in vivo and showed the behavior of self-assembly, thermoresponsiveness, enzyme stimulation-responsiveness, tumor-targeting, biocompatibility, and biodegradability. Near-infrared imaging studies showed that fluorescent dye-stained TVEFP effectively aggregated at the tumor site. The TVEFP nanocarrier significantly expands the application of the therapeutic protein hVEGI-192 and improves the imaging and biotherapeutic effects in TNBC, chiefly based on anti-angiogenesis effects.
Collapse
Affiliation(s)
- Meiling Zhou
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yuming Yao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Shuo Ma
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Mingyuan Zou
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yaya Chen
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Shijie Cai
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Fengfeng Zhao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Huina Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Feng Xiao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - GuliNazhaer Abudushalamu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiaobo Fan
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Guoqiu Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Laboratory Medcine, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
25
|
Zhang LM, Yang M, Zhou SW, Zhang H, Feng Y, Shi L, Li DS, Lu QM, Zhang ZH, Zhao M. Blapstin, a Diapause-Specific Peptide-Like Peptide from the Chinese Medicinal Beetle Blaps rhynchopetera, Has Antifungal Function. Microbiol Spectr 2023; 11:e0308922. [PMID: 37140456 PMCID: PMC10269622 DOI: 10.1128/spectrum.03089-22] [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: 08/07/2022] [Accepted: 04/10/2023] [Indexed: 05/05/2023] Open
Abstract
Drug resistance against bacteria and fungi has become common in recent years, and it is urgent to discover novel antimicrobial peptides to manage this problem. Many antimicrobial peptides from insects have been reported to have antifungal activity and are candidate molecules in the treatment of human diseases. In the present study, we characterized an antifungal peptide named blapstin that was isolated from the Chinese medicinal beetle Blaps rhynchopetera used in folk medicine. The complete coding sequence was cloned from the cDNA library prepared from the midgut of B. rhynchopetera. It is a 41-amino-acid diapause-specific peptide (DSP)-like peptide stabilized by three disulfide bridges and shows antifungal activity against Candida albicans and Trichophyton rubrum with MICs of 7 μM and 5.3 μM, respectively. The C. albicans and T. rubrum treated with blapstin showed irregular and shrunken cell membranes. In addition, blapstin inhibited the activity of C. albicans biofilm and showed little hemolytic or toxic activity on human cells and it is highly expressed in the fat body, followed by the hemolymph, midgut, muscle, and defensive glands. These results indicate that blapstin may help insects fight against fungi and showed a potential application in the development of antifungal reagents. IMPORTANCE Candida albicans is one of the conditional pathogenic fungi causing severe nosocomial infections. Trichophyton rubrum and other skin fungi are the main pathogens of superficial cutaneous fungal diseases, especially in children and the elderly. At present, antibiotics such as amphotericin B, ketoconazole, and fluconazole are the main drugs for the clinical treatment of C. albicans and T. rubrum infections. However, these drugs have certain acute toxicity. Long-term use can increase kidney damage and other side effects. Therefore, obtaining broad-spectrum antifungal drugs with high efficiency and low toxicity for the treatment of C. albicans and T. rubrum infections is a top priority. Blapstin is an antifungal peptide which shows activity against C. albicans and T. rubrum. The discovery of blapstin provides a novel clue for our understanding of the innate immunity of Blaps rhynchopetera and provides a template for designing antifungal drugs.
Collapse
Affiliation(s)
- La-Mei Zhang
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- College of Forestry, Nanjing Forestry University, Nanjing, Jiangsu, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| | - Min Yang
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Sheng-Wen Zhou
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Hao Zhang
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Ying Feng
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| | - Lei Shi
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| | - Dong-Sheng Li
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Sino-African Joint Research Center, Chinese Academy of Science, Wuhan, Hubei, China
| | - Qiu-Min Lu
- Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
- Sino-African Joint Research Center, Chinese Academy of Science, Wuhan, Hubei, China
| | - Zhong-He Zhang
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| | - Min Zhao
- Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, China
- Key Laboratory of Breeding and Utilization of Resource Insects, National Forestry and Grassland Administration, Kunming, China
| |
Collapse
|
26
|
Past, Present, and Future of Naturally Occurring Antimicrobials Related to Snake Venoms. Animals (Basel) 2023; 13:ani13040744. [PMID: 36830531 PMCID: PMC9952678 DOI: 10.3390/ani13040744] [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: 12/15/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023] Open
Abstract
This review focuses on proteins and peptides with antimicrobial activity because these biopolymers can be useful in the fight against infectious diseases and to overcome the critical problem of microbial resistance to antibiotics. In fact, snakes show the highest diversification among reptiles, surviving in various environments; their innate immunity is similar to mammals and the response of their plasma to bacteria and fungi has been explored mainly in ecological studies. Snake venoms are a rich source of components that have a variety of biological functions. Among them are proteins like lectins, metalloproteinases, serine proteinases, L-amino acid oxidases, phospholipases type A2, cysteine-rich secretory proteins, as well as many oligopeptides, such as waprins, cardiotoxins, cathelicidins, and β-defensins. In vitro, these biomolecules were shown to be active against bacteria, fungi, parasites, and viruses that are pathogenic to humans. Not only cathelicidins, but all other proteins and oligopeptides from snake venom have been proteolyzed to provide short antimicrobial peptides, or for use as templates for developing a variety of short unnatural sequences based on their structures. In addition to organizing and discussing an expressive amount of information, this review also describes new β-defensin sequences of Sistrurus miliarius that can lead to novel peptide-based antimicrobial agents, using a multidisciplinary approach that includes sequence phylogeny.
Collapse
|
27
|
Coppola D, Buonocore C, Palisse M, Tedesco P, de Pascale D. Exploring Oceans for Curative Compounds: Potential New Antimicrobial and Anti-Virulence Molecules against Pseudomonas aeruginosa. Mar Drugs 2022; 21:9. [PMID: 36662182 PMCID: PMC9865402 DOI: 10.3390/md21010009] [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: 11/21/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Although several antibiotics are already widely used against a large number of pathogens, the discovery of new antimicrobial compounds with new mechanisms of action is critical today in order to overcome the spreading of antimicrobial resistance among pathogen bacteria. In this regard, marine organisms represent a potential source of a wide diversity of unique secondary metabolites produced as an adaptation strategy to survive in competitive and hostile environments. Among the multidrug-resistant Gram-negative bacteria, Pseudomonas aeruginosa is undoubtedly one of the most important species due to its high intrinsic resistance to different classes of antibiotics on the market and its ability to cause serious therapeutic problems. In the present review, we first discuss the general mechanisms involved in the antibiotic resistance of P. aeruginosa. Subsequently, we list the marine molecules identified up until now showing activity against P. aeruginosa, dividing them according to whether they act as antimicrobial or anti-virulence compounds.
Collapse
Affiliation(s)
- Daniela Coppola
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton 55, 80133 Naples, Italy
| | - Carmine Buonocore
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton 55, 80133 Naples, Italy
| | - Morgan Palisse
- Département des Sciences de la Vie et de la Terre, Université de Caen Normandie, Boulevard Maréchal Juin CS, CEDEX, 14032 Caen, France
| | - Pietro Tedesco
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton 55, 80133 Naples, Italy
| | - Donatella de Pascale
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Ammiraglio Ferdinando Acton 55, 80133 Naples, Italy
| |
Collapse
|
28
|
Shi J, Wu J, Chen Q, Shen Y, Mi K, Yang H, Mu L. A Frog-Derived Cathelicidin Peptide with Dual Antimicrobial and Immunomodulatory Activities Effectively Ameliorates Staphylococcus aureus-Induced Peritonitis in Mice. ACS Infect Dis 2022; 8:2464-2479. [PMID: 36378028 DOI: 10.1021/acsinfecdis.2c00260] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
As antimicrobial resistance poses an increasing threat to public health, it is urgent to develop new antimicrobial agents. In this paper, we identify a novel 30-residue peptide (Nv-CATH, NCNFLCKVKQRLRSVSSTSHIGMAIPRPRG) from the skin of the frog Nanorana ventripunctata, which belongs to the cathelicidin family. Nv-CATH exhibited broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria. Nv-CATH significantly protected mice from lethal infections caused by Staphylococcus aureus. Furthermore, the peptide suppressed excessive and harmful inflammatory responses by repressing the production of NO, IL-6, TNF-α, and IL-1β. The NF-κB-NLRP3 and MAPK inflammatory signaling pathways were involved in the protection in vitro and in vivo. Nv-CATH also modulated macrophage/monocyte and neutrophil trafficking to the infection site by stimulating CXCL1, CXCL2, and CCL2 production in macrophages. Nv-CATH augmented immunocyte-mediated bacterial killing by modestly promoting neutrophils' phagocytosis and PMA-induced NET formation. Thus, Nv-CATH protects mice against bacterial infection by antimicrobial-immunomodulatory duality. The combination of these two characteristics makes Nv-CATH a promising molecule template for the development of novel antimicrobial and antibiotic-resistant agents.
Collapse
Affiliation(s)
- Jie Shi
- Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jing Wu
- Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Qian Chen
- Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yan Shen
- Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Kai Mi
- Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Hailong Yang
- Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Lixian Mu
- Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| |
Collapse
|
29
|
Wu D, Fu L, Wen W, Dong N. The dual antimicrobial and immunomodulatory roles of host defense peptides and their applications in animal production. J Anim Sci Biotechnol 2022; 13:141. [PMID: 36474280 PMCID: PMC9724304 DOI: 10.1186/s40104-022-00796-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022] Open
Abstract
Host defense peptides (HDPs) are small molecules with broad-spectrum antimicrobial activities against infectious bacteria, viruses, and fungi. Increasing evidence suggests that HDPs can also indirectly protect hosts by modulating their immune responses. Due to these dual roles, HDPs have been considered one of the most promising antibiotic substitutes to improve growth performance, intestinal health, and immunity in farm animals. This review describes the antimicrobial and immunomodulatory roles of host defense peptides and their recent applications in animal production.
Collapse
Affiliation(s)
- Di Wu
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Linglong Fu
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Weizhang Wen
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Na Dong
- grid.22935.3f0000 0004 0530 8290State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| |
Collapse
|
30
|
Wang J, Yuan T, He X, Yi Z, Li H, Gao W, Li Q. Production, characterization, and application of phage-derived PK34 recombinant anti-microbial peptide. Appl Microbiol Biotechnol 2022; 107:163-174. [DOI: 10.1007/s00253-022-12306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 12/02/2022]
|
31
|
Santana FL, Estrada K, Alford MA, Wu BC, Dostert M, Pedraz L, Akhoundsadegh N, Kalsi P, Haney EF, Straus SK, Corzo G, Hancock REW. Novel Alligator Cathelicidin As-CATH8 Demonstrates Anti-Infective Activity against Clinically Relevant and Crocodylian Bacterial Pathogens. Antibiotics (Basel) 2022; 11:1603. [PMID: 36421248 PMCID: PMC9686568 DOI: 10.3390/antibiotics11111603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2024] Open
Abstract
Host defense peptides (HDPs) represent an alternative way to address the emergence of antibiotic resistance. Crocodylians are interesting species for the study of these molecules because of their potent immune system, which confers high resistance to infection. Profile hidden Markov models were used to screen the genomes of four crocodylian species for encoded cathelicidins and eighteen novel sequences were identified. Synthetic cathelicidins showed broad spectrum antimicrobial and antibiofilm activity against several clinically important antibiotic-resistant bacteria. In particular, the As-CATH8 cathelicidin showed potent in vitro activity profiles similar to the last-resort antibiotics vancomycin and polymyxin B. In addition, As-CATH8 demonstrated rapid killing of planktonic and biofilm cells, which correlated with its ability to cause cytoplasmic membrane depolarization and permeabilization as well as binding to DNA. As-CATH8 displayed greater antibiofilm activity than the human cathelicidin LL-37 against methicillin-resistant Staphylococcus aureus in a human organoid model of biofilm skin infection. Furthermore, As-CATH8 demonstrated strong antibacterial effects in a murine abscess model of high-density bacterial infections against clinical isolates of S. aureus and Acinetobacter baumannii, two of the most common bacterial species causing skin infections globally. Overall, this work expands the repertoire of cathelicidin peptides known in crocodylians, including one with considerable therapeutic promise for treating common skin infections.
Collapse
Affiliation(s)
- Felix L. Santana
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Karel Estrada
- Unidad de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico
| | - Morgan A. Alford
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Bing C. Wu
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Melanie Dostert
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Lucas Pedraz
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Noushin Akhoundsadegh
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Pavneet Kalsi
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Evan F. Haney
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Suzana K. Straus
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Gerardo Corzo
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Mexico
| | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
32
|
Zhang M, Ouyang J, Fu L, Xu C, Ge Y, Sun S, Li X, Lai S, Ke H, Yuan B, Yang K, Yu H, Gao L, Wang Y. Hydrophobicity Determines the Bacterial Killing Rate of α-Helical Antimicrobial Peptides and Influences the Bacterial Resistance Development. J Med Chem 2022; 65:14701-14720. [DOI: 10.1021/acs.jmedchem.2c01238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Minghui Zhang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu215123, China
| | - Jianhong Ouyang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu215123, China
| | - Lei Fu
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing100875, China
| | - Cheng Xu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou215006Jiangsu, China
| | - Yuke Ge
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou215006Jiangsu, China
| | - Shuqing Sun
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou215006Jiangsu, China
| | - Xiangyuan Li
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing100875, China
| | - Shian Lai
- Department of Molecular Chemistry and Biochemistry, Faculty of Science and Engineering, Doshisha University, Kyotanabe, Kyoto610-0394, Japan
| | - Hengte Ke
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu215123, China
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan, Guangdong523808, China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou215006Jiangsu, China
| | - Haining Yu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, Liaoning116024, China
| | - Lianghui Gao
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing100875, China
| | - Yipeng Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu215123, China
| |
Collapse
|
33
|
Synergistic Membrane Disturbance Improves the Antibacterial Performance of Polymyxin B. Polymers (Basel) 2022; 14:polym14204316. [PMID: 36297894 PMCID: PMC9611124 DOI: 10.3390/polym14204316] [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: 09/12/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 01/24/2023] Open
Abstract
Drug-resistant Gram-negative bacteria pose a serious threat to public health, and polymyxin B (PMB) is clinically used as a last-line therapy for the treatment of infections caused by these pathogens. However, the appearance of PMB resistance calls for an effort to develop new approaches to improve its antibacterial performance. In this work, a new type of nanocomposite, composed of PMB molecules being chemically decorated on the surface of graphene oxide (GO) nanosheets, was designed, which showed potent antibacterial ability through synergistically and physically disturbing the bacterial membrane. The as-fabricated PMB@GO nanocomposites demonstrated an enhanced bacterial-killing efficiency, with a minimum inhibitory concentration (MIC) value half of that of free PMB (with an MIC value as low as 0.5 μg mL-1 over Escherichia coli), and a bacterial viability less than one fourth of that of PMB (with a bacterial reduction of 60% after 3 h treatment, and 90% after 6 h incubation). Furthermore, the nanocomposite displayed moderate cytotoxicity or hemolysis effect, with cellular viabilities over 85% at concentrations up to 16 times the MIC value. Studies on antibacterial mechanism revealed that the synergy between PMB molecules and GO nanosheets greatly facilitated the vertical insertion of the nanocomposite into the lipid membrane, leading to membrane disturbance and permeabilization. Our results demonstrate a physical mechanism for improving the antibacterial performance of PMB and developing advanced antibacterial agents for better clinic uses.
Collapse
|
34
|
Yuan B, Lu X, Yang M, He Q, Cha Z, Fang Y, Yang Y, Xu L, Yan J, Lai R, Wang A, Yu X, Duan Z. A designed antimicrobial peptide with potential ability against methicillin resistant Staphylococcus aureus. Front Microbiol 2022; 13:1029366. [PMID: 36299717 PMCID: PMC9589885 DOI: 10.3389/fmicb.2022.1029366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive pathogenic bacterium, which persistently colonizes the anterior nares of approximately 20-30% of the healthy adult population, and up to 60% is intermittently colonized. With the misuse and overuse of antibiotics, large-scale drug-resistant bacteria, including methicillin-resistant S. aureus (MRSA), have been appeared. MRSA is among the most prevalent pathogens causing community-associated infections. Once out of control, the number of deaths caused by antimicrobial resistance may exceed 10 million annually by 2050. Antimicrobial peptides (AMPs) are regarded as the best solution, for they are not easy to develop drug resistance. Based on our previous research, here we designed a new antimicrobial peptide named GW18, which showed excellent antimicrobial activity against S. aureus, even MRSA, with the hemolysis less than 5%, no cytotoxicity, and no acute toxicity. Notably, administration of GW18 significantly decreased S. aureus infection in mouse model. These findings identify GW18 as the ideal candidate against S. aureus infection.
Collapse
Affiliation(s)
- Bingqian Yuan
- School of Life Sciences, Tianjin University, Tianjin, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Center, and Engineering Laboratory of Peptides, Kunming Institute of Zoology, Kunming, China
- The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaoyu Lu
- School of Life Sciences, Tianjin University, Tianjin, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Center, and Engineering Laboratory of Peptides, Kunming Institute of Zoology, Kunming, China
- The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Min Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Center, and Engineering Laboratory of Peptides, Kunming Institute of Zoology, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Qiyi He
- College of Life Science, Chongqing Normal University, Chongqing, China
| | - Zhuocen Cha
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital, Kunming, Yunnan, China
| | - Yaqun Fang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Center, and Engineering Laboratory of Peptides, Kunming Institute of Zoology, Kunming, China
| | - Yan Yang
- Yunnan Provincial Academy of Science and Technology, Kunming, China
| | - Lei Xu
- Yunnan Provincial Academy of Science and Technology, Kunming, China
| | - Jingting Yan
- Yunnan Provincial Academy of Science and Technology, Kunming, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Center, and Engineering Laboratory of Peptides, Kunming Institute of Zoology, Kunming, China
| | - Aili Wang
- Center for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, China
| | - Xiaodong Yu
- College of Life Science, Chongqing Normal University, Chongqing, China
| | - Zilei Duan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Center, and Engineering Laboratory of Peptides, Kunming Institute of Zoology, Kunming, China
| |
Collapse
|
35
|
Wang J, Jiang B, Wang K, Dai J, Dong C, Wang Y, Zhang P, Li M, Xu W, Wei L. A cathelicidin antimicrobial peptide from Hydrophis cyanocinctus inhibits Zika virus infection by downregulating expression of a viral entry factor. J Biol Chem 2022; 298:102471. [PMID: 36089062 PMCID: PMC9530963 DOI: 10.1016/j.jbc.2022.102471] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 11/05/2022] Open
Abstract
Zika virus (ZIKV) is a re-emerging flavivirus that causes conditions such as microcephaly and testis damage. The spread of ZIKV has become a major public health concern. Recent studies indicated that antimicrobial peptides are an ideal source for screening antiviral candidates with broad-spectrum antiviral activities, including against ZIKV. We herein found that Hc-CATH, a cathelicidin antimicrobial peptide identified from the sea snake Hydrophis cyanocinctus in our previous work, conferred protection against ZIKV infection in host cells and showed preventative efficacy and therapeutic efficacy in C57BL/6J mice, Ifnar1−/− mice, and pregnant mice. Intriguingly, we revealed that Hc-CATH decreased the susceptibility of host cells to ZIKV by downregulating expression of AXL, a TAM (TYRO3, AXL and MERTK) family kinase receptor that mediates ZIKV infection, and subsequently reversed the negative regulation of AXL on host’s type I interferon response. Furthermore, we showed that the cyclo-oxygenase-2/prostaglandin E2/adenylyl cyclase/protein kinase A pathway was involved in Hc-CATH-mediated AXL downregulation, and Hc-CATH in addition directly inactivated ZIKV particles by disrupting viral membrane. Finally, while we found Hc-CATH did not act on the late stage of ZIKV infection, structure–function relationship studies revealed that α-helix and phenylalanine residues are key structural requirements for its protective efficacy against initial ZIKV infection. In summary, we demonstrate that Hc-CATH provides prophylactic and therapeutic efficacy against ZIKV infection via downregulation of AXL, as well as inactivating the virion. Our findings reveal a novel mechanism of cathelicidin against viral infection and highlight the potential of Hc-CATH to prevent and treat ZIKV infection.
Collapse
Affiliation(s)
- Jing Wang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Bingyan Jiang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Kezhen Wang
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jianfeng Dai
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Chunsheng Dong
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Yipeng Wang
- Department of Biopharmaceuticals, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Peng Zhang
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Li
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China.
| | - Lin Wei
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
36
|
Hunting for Novel Routes in Anticancer Drug Discovery: Peptides against Sam-Sam Interactions. Int J Mol Sci 2022; 23:ijms231810397. [PMID: 36142306 PMCID: PMC9499636 DOI: 10.3390/ijms231810397] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 01/10/2023] Open
Abstract
Among the diverse protein binding modules, Sam (Sterile alpha motif) domains attract attention due to their versatility. They are present in different organisms and play many functions in physiological and pathological processes by binding multiple partners. The EphA2 receptor contains a Sam domain at the C-terminus (EphA2-Sam) that is able to engage protein regulators of receptor stability (including the lipid phosphatase Ship2 and the adaptor Odin). Ship2 and Odin are recruited by EphA2-Sam through heterotypic Sam-Sam interactions. Ship2 decreases EphA2 endocytosis and consequent degradation, producing chiefly pro-oncogenic outcomes in a cellular milieu. Odin, through its Sam domains, contributes to receptor stability by possibly interfering with ubiquitination. As EphA2 is upregulated in many types of tumors, peptide inhibitors of Sam-Sam interactions by hindering receptor stability could function as anticancer therapeutics. This review describes EphA2-Sam and its interactome from a structural and functional perspective. The diverse design strategies that have thus far been employed to obtain peptides targeting EphA2-mediated Sam-Sam interactions are summarized as well. The generated peptides represent good initial lead compounds, but surely many efforts need to be devoted in the close future to improve interaction affinities towards Sam domains and consequently validate their anticancer properties.
Collapse
|
37
|
Chen Z, Zhang J, Ming Z, Tong H, Wu J, Chen Q, Wang Y, Luo F, Wang Y, Feng T. As-Cathelicidin4 enhances the immune response and resistance against Aeromonas hydrophila in caridean shrimp. JOURNAL OF FISH DISEASES 2022; 45:743-754. [PMID: 35100453 DOI: 10.1111/jfd.13588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 06/14/2023]
Affiliation(s)
- Zhiqiang Chen
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinyu Zhang
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Zhihao Ming
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Hao Tong
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Jiahui Wu
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Qiaoqiao Chen
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Yintao Wang
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Fangmei Luo
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Yipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Tingting Feng
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| |
Collapse
|
38
|
Zhang K, Liu Y, Tang Y. Screening of TNFR1 Binding Peptides from Deinagkistrodon acutus Venom through Phage Display. Toxins (Basel) 2022; 14:toxins14020155. [PMID: 35202182 PMCID: PMC8878721 DOI: 10.3390/toxins14020155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
The venomous species Deinagkistrodon acutus has been used as anti-inflammatory medicine in China for a long time. It has been proven to have anti-inflammatory activity, but its specific anti-inflammatory components have not yet been fully elucidated. Tumor necrosis factor receptor-1 (TNFR1), which participates in important intracellular signaling pathways, mediates apoptosis, and functions as a regulator of inflammation, is often used as the target to develop anti-inflammatory drugs. The small peptides of snake venom have the advantages of weak immunogenicity and strong activity. To obtain the specific TNFR1 binding peptides, we constructed a T7 phage library of D. acutus venom glands, and then performed biopanning against TNFR1 on the constructed library. After biopanning three times, several sequences with potential binding capacity were obtained and one 41-amino acid peptide was selected through a series of biological analyses including sequence length, solubility, and simulated affinity, named DAvp-1. After synthesis, the binding capacity of DAvp-1 and TNFR1 was verified using surface plasmon resonance technology (SPR). Conclusively, by applying phage display technology, this work depicts the successful screening of a promising peptide DAvp-1 from D. acutus venom that binds to TNFR1. Additionally, our study emphasizes the usefulness of phage display technology for studies on screening natural product components.
Collapse
Affiliation(s)
- Kangran Zhang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China;
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Liu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China;
- Correspondence: (Y.L.); (Y.T.)
| | - Yezhong Tang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China;
- Correspondence: (Y.L.); (Y.T.)
| |
Collapse
|
39
|
Zhu Y, Hao W, Wang X, Ouyang J, Deng X, Yu H, Wang Y. Antimicrobial peptides, conventional antibiotics, and their synergistic utility for the treatment of drug-resistant infections. Med Res Rev 2022; 42:1377-1422. [PMID: 34984699 DOI: 10.1002/med.21879] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/09/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022]
Abstract
Antimicrobial peptides (AMPs), also known as host defense peptides (HDPs), are important effector immune defense molecules in multicellular organisms. AMPs exert their antimicrobial activities through several mechanisms; thus far, induction of drug resistance through AMPs has been regarded as unlikely. Therefore, they have great potential as new generation antimicrobial agents. To date, more than 30 AMP-related drugs are in the clinical trial phase. In recent years, studies show that some AMPs and conventional antibiotics have synergistic effects. The combined use of AMPs and antibiotics can kill drug-resistant pathogens, prevent drug resistance, and significantly improve the therapeutic effects of antibiotics. In this review, we discuss the progress in synergistic studies on AMPs and conventional antibiotics. An overview of the current understanding of the functional scope of AMPs, ongoing clinical trials, and challenges in the development processes are also presented.
Collapse
Affiliation(s)
- Yiyun Zhu
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Weijing Hao
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xia Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jianhong Ouyang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xinyi Deng
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Haining Yu
- Department of Bioscience and Biotechnology, Dalian University of Technology, Dalian, Liaoning, China
| | - Yipeng Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
40
|
Wu J, Guo R, Chai J, Xiong W, Tian M, Lu W, Xu X. The Protective Effects of Cath-MH With Anti-Propionibacterium Acnes and Anti-Inflammation Functions on Acne Vulgaris. Front Pharmacol 2021; 12:788358. [PMID: 34955858 PMCID: PMC8696257 DOI: 10.3389/fphar.2021.788358] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/08/2021] [Indexed: 12/01/2022] Open
Abstract
Acne vulgaris is a common adolescent skin condition which is mainly caused by Propionibacterium acnes overcolonization and subsequent inflammation. Our previous studies have demonstrated that Cath-MH, an antimicrobial peptide from the skin of the frog Microhyla heymonsivogt, possesses potential antimicrobial, LPS-binding, and anti-septicemic properties. However, its protective effects and potential mechanisms against acne vulgaris are still unclear. In the present study, its anti-P. acnes effects were measured by two-fold broth dilution method, agglutination assay, scanning electron microscopy and confocal laser scanning microscopy experiments. Its treatment potential for acne vulgaris was further evaluated in mice ear inoculated by P. acnes. In addition, the binding ability between Cath-MH and LTA was measured by the Circular Dichroism and antibacterial assay. Moreover, the anti-inflammatory efficiency of Cath-MH was evaluated in LTA- and LPS-induced RAW 264.7 macrophage cells. Cath-MH was found to kill P. acnes with a MIC value of about 1.56 μM by membrane disruption mechanism. It also exhibited agglutination activity against P. acnes. Cath-MH was able to bind LTA as well as LPS, inhibit LTA/LPS-stimulated TLR2/4 expression, and subsequently decreased the inflammatory response in RAW 264.7 cells. As expected, Cath-MH alleviated the formation of edema and the infiltration of inflammatory cells in acne mouse model with concurrent suppression of P. acnes growth and inflammatory cytokines expression in vivo. The potent P. acnes inhibition activity combined with powerful anti-inflammatory effect of Cath-MH indicates its potential as a novel therapeutic option for acne vulgaris.
Collapse
Affiliation(s)
- Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ruiyin Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Weichen Xiong
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Maolin Tian
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wancheng Lu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
Duan Y, Ouyang J, Mo G, Hao W, Zhang P, Yang H, Liu X, Wang R, Cao B, Wang Y, Yu H. Defensing role of novel piscidins from largemouth bass (Micropterus salmoides) with evidence of bactericidal activities and inducible expressional delineation. Microbiol Res 2021; 256:126953. [PMID: 34972023 DOI: 10.1016/j.micres.2021.126953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022]
Abstract
Micropterus salmoides is an economical important species of freshwater-cultured fish, the in-depth knowledge of its immune system is in urgent development to cope with serious infectious diseases. Piscidin is an important antimicrobial peptide (AMP) family existing in almost all teleosts. However, no piscidin has been reported in largemouth bass. In this study, three novel piscidins (MSPiscidin-1, -2, and -3) were firstly identified and characterized from the largemouth bass. The predicted mature peptides of MSPiscidin-1, -2, and -3 (consists of 24, 27, 25 amino acid residues, respectively) all adopted an amphipathic α-helical conformation representative of cationic AMPs that are important for membrane permeabilization and antibacterial activity. MSPiscidin-2 and -3 indeed displayed strong, broad-spectrum, and highly efficient antimicrobial activities in vitro against aquatic pathogens, but MSPiscidin-1 didn't show direct antimicrobial activity. MSPiscidin-2 and -3 killed bacteria mainly by inducing membrane permeabilization, in addition, they also can interact with bacterial genomic DNA, which might influence the DNA replication and transcription. Besides, MSPiscidin-2 and -3 could effectively inhibit the formation of the bacterial biofilm and eliminate the preformed biofilms. In vivo, MSPiscidin-1-3 genes showed an inducible expression pattern in the tested tissues upon Vibrio harveyi infection, which further indicated the key roles of piscidins in innate immunity in largemouth bass. Overall, this study will supplement the understanding of M. salmoides innate immune system and provide candidates for the design of novel peptide antibacterial agents used in aquaculture.
Collapse
Affiliation(s)
- Yuxin Duan
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Jianhong Ouyang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Guoxiang Mo
- College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Weijing Hao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Peng Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| | - Huaixin Yang
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Xiaowei Liu
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Runying Wang
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Biyin Cao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Haining Yu
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| |
Collapse
|
42
|
Antimicrobial Activity of Snake β-Defensins and Derived Peptides. Toxins (Basel) 2021; 14:toxins14010001. [PMID: 35050978 PMCID: PMC8777785 DOI: 10.3390/toxins14010001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/20/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022] Open
Abstract
β-defensins are antimicrobial peptides presenting in vertebrate animals. They participate in innate immunity, but little is known about them in reptiles, including snakes. Although several β-defensin genes were described in Brazilian snakes, their function is still unknown. The peptide sequence from these genes was deduced, and synthetic peptides (with approximately 40 amino acids and derived peptides) were tested against pathogenic bacteria and fungi using microbroth dilution assays. The linear peptides, derived from β-defensins, were designed applying the bioisosterism strategy. The linear β-defensins were more active against Escherichia coli, Micrococcus luteus, Citrobacter freundii, and Staphylococcus aureus. The derived peptides (7–14 mer) showed antibacterial activity against those bacteria and on Klebsiella pneumoniae. Nonetheless, they did not present activity against Candida albicans, Cryptococcus neoformans, Trychophyton rubrum, and Aspergillus fumigatus showing that the cysteine substitution to serine is deleterious to antifungal properties. Tryptophan residue showed to be necessary to improve antibacterial activity. Even though the studied snake β-defensins do not have high antimicrobial activity, they proved to be attractive as template molecules for the development of antibiotics.
Collapse
|
43
|
Wan MC, Qin W, Lei C, Li QH, Meng M, Fang M, Song W, Chen JH, Tay F, Niu LN. Biomaterials from the sea: Future building blocks for biomedical applications. Bioact Mater 2021; 6:4255-4285. [PMID: 33997505 PMCID: PMC8102716 DOI: 10.1016/j.bioactmat.2021.04.028] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 02/08/2023] Open
Abstract
Marine resources have tremendous potential for developing high-value biomaterials. The last decade has seen an increasing number of biomaterials that originate from marine organisms. This field is rapidly evolving. Marine biomaterials experience several periods of discovery and development ranging from coralline bone graft to polysaccharide-based biomaterials. The latter are represented by chitin and chitosan, marine-derived collagen, and composites of different organisms of marine origin. The diversity of marine natural products, their properties and applications are discussed thoroughly in the present review. These materials are easily available and possess excellent biocompatibility, biodegradability and potent bioactive characteristics. Important applications of marine biomaterials include medical applications, antimicrobial agents, drug delivery agents, anticoagulants, rehabilitation of diseases such as cardiovascular diseases, bone diseases and diabetes, as well as comestible, cosmetic and industrial applications.
Collapse
Affiliation(s)
- Mei-chen Wan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Wen Qin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Chen Lei
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Qi-hong Li
- Department of Stomatology, The Fifth Medical Centre, Chinese PLA General Hospital (Former 307th Hospital of the PLA), Dongda Street, Beijing, 100071, PR China
| | - Meng Meng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Ming Fang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Wen Song
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Ji-hua Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Franklin Tay
- College of Graduate Studies, Augusta University, Augusta, GA, 30912, USA
| | - Li-na Niu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, PR China
| |
Collapse
|
44
|
Xiong W, Li J, Feng Y, Chai J, Wu J, Hu Y, Tian M, Lu W, Xu X, Zou M. Brevinin-2GHk, a Peptide Derived from the Skin of Fejervarya limnocharis, Inhibits Zika Virus Infection by Disrupting Viral Integrity. Viruses 2021; 13:v13122382. [PMID: 34960651 PMCID: PMC8708736 DOI: 10.3390/v13122382] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/25/2021] [Indexed: 12/11/2022] Open
Abstract
Several years have passed since the Zika virus (ZIKV) pandemic reoccurred in 2015–2016. However, there is still a lack of proved protective vaccines or effective drugs against ZIKV. The peptide brevinin-2GHk (BR2GK), pertaining to the brevinin-2 family of antimicrobial peptides, has been reported to exhibit only weak antibacterial activity, and its antiviral effects have not been investigated. Thus, we analyzed the effect of BR2GK on ZIKV infection. BR2GK showed significant inhibitory activity in the early and middle stages of ZIKV infection, with negligible cytotoxicity. Furthermore, BR2GK was suggested to bind with ZIKV E protein and disrupt the integrity of the envelope, thus directly inactivating ZIKV. In addition, BR2GK can also penetrate the cell membrane, which may contribute to inhibition of the middle stage of ZIKV infection. BR2GK blocked ZIKV E protein expression with an IC50 of 3.408 ± 0.738 μΜ. In summary, BR2GK was found to be a multi-functional candidate and a potential lead compound for further development of anti-ZIKV drugs.
Collapse
Affiliation(s)
- Weichen Xiong
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jingyan Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yifei Feng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yunrui Hu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Maolin Tian
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wancheng Lu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Correspondence: (X.X.); (M.Z.)
| | - Min Zou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (W.X.); (J.L.); (Y.F.); (J.C.); (J.W.); (Y.H.); (M.T.); (W.L.)
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Correspondence: (X.X.); (M.Z.)
| |
Collapse
|
45
|
Simora RMC, Wang W, Coogan M, El Husseini N, Terhune JS, Dunham RA. Effectiveness of Cathelicidin Antimicrobial Peptide against Ictalurid Catfish Bacterial Pathogens. JOURNAL OF AQUATIC ANIMAL HEALTH 2021; 33:178-189. [PMID: 34121235 DOI: 10.1002/aah.10131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 06/12/2023]
Abstract
One of the major goals in aquaculture is to protect fish against infectious diseases as disease outbreaks could lead to economic losses if not controlled. Antimicrobial peptides (AMPs), a class of highly conserved peptides known to possess direct antimicrobial activities against invading pathogens, were evaluated for their ability to protect Channel Catfish Ictalurus punctatus and hybrid catfish (female Channel Catfish × male Blue Catfish I. furcatus) against infection caused by the fish pathogen Aeromonas hydrophila ML09-119. To identify effective peptides, the minimum inhibitory concentrations against bacterial pathogens Edwardsiella ictaluri S97-773, Edwardsiella piscicida E22-10, A. hydrophila ML09-119, Aeromonas veronii 03X03876, and Flavobacterium columnare GL-001 were determined in vitro. In general and overall, cathelicidins derived from alligator and sea snake exhibited more potent and rapid antimicrobial activities against the tested catfish pathogens as compared to cecropin and pleurocidin AMPs and ampicillin, the antibiotic control. When the peptides (2.5 µg of peptide/g of fish) were injected into fish and simultaneously challenged with A. hydrophila through immersion, increased survival rates in Channel Catfish and hybrid catfish were observed in both cathelicidin (alligator and sea snake) treatments as compared to other peptides and the infected control (P < 0.001) with alligator cathelicidin being the overall best treatment. Bacterial numbers in the kidney and liver of Channel Catfish and hybrid catfish also decreased (P < 0.05) for cathelicidin-injected groups at 24 and 48 h after challenge infection. These results show the potential of cathelicidin to protect catfish against bacterial infections and suggest that an approach overexpressing the peptide in transgenic fish, which is the long-term goal of this research program, may provide a method of decreasing bacterial disease problems in catfish as delivering the peptides via individual injection or feeding would not be economically feasible.
Collapse
Affiliation(s)
- Rhoda Mae C Simora
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, Alabama, 36849, USA
- College of Fisheries and Ocean Sciences, University of the Philippines Visayas, Miagao, Iloilo, 5023, Philippines
| | - Wenwen Wang
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, Alabama, 36849, USA
| | - Michael Coogan
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, Alabama, 36849, USA
| | - Nour El Husseini
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, Alabama, 36849, USA
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
| | - Jeffery S Terhune
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, Alabama, 36849, USA
| | - Rex A Dunham
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, Alabama, 36849, USA
| |
Collapse
|
46
|
Luo X, Ouyang J, Wang Y, Zhang M, Fu L, Xiao N, Gao L, Zhang P, Zhou J, Wang Y. A novel anionic cathelicidin lacking direct antimicrobial activity but with potent anti-inflammatory and wound healing activities from the salamander Tylototriton kweichowensis. Biochimie 2021; 191:37-50. [PMID: 34438004 DOI: 10.1016/j.biochi.2021.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/08/2021] [Accepted: 08/18/2021] [Indexed: 12/26/2022]
Abstract
Cathelicidin is a family of antimicrobial peptides (AMPs) existing in vertebrates, which play multiple functions in host responses against environmental stresses. All cathelicidins identified to date are cationic, no anionic member with net negative charges has been reported. In the present study, a novel anionic cathelicidin (TK-CATH) with a net charge of -3 was identified from the skin of the salamander, T. kweichowensis. Unlike most other cathelicidin members, it didn't exhibit direct antimicrobial activity. However, it demonstrated strong anti-inflammatory activity. It effectively inhibited the LPS-induced pro-inflammatory cytokine gene expression and protein production in amphibian leukocytes and mouse macrophages by inhibiting the LPS-activated mitogen-activated protein kinase (MAPK) signaling pathways. Besides, TK-CATH showed potent wound healing activity. It could effectively induce the production of several cytokines, chemokines and growth factors relating to wound healing, promote the motility and proliferation of keratinocytes, and accelerate the skin wound healing in a mouse full-thickness wound model. These results imply that TK-CATH participates in both the inflammatory phase and new tissue formation phase of wound repair process. Meanwhile, TK-CATH exhibited weak but effective free radical scavenging activity and low cytotoxicity. All the results above indicate that TK-CATH is a multifunctional peptide in the skin of the salamander T. kweichowensis. It may play important roles in host immune responses against bacterial infection and skin wound repair.
Collapse
Affiliation(s)
- Xuanjin Luo
- Biology Department, Guizhou Normal University, Guiyang, 550000, Guizhou, China
| | - Jianhong Ouyang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yan Wang
- Biology Department, Guizhou Normal University, Guiyang, 550000, Guizhou, China
| | - Minghui Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Lei Fu
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Ning Xiao
- Guiyang Nursing Vacational College, Guiyang, 550014, Guizhou, China
| | - Lianghui Gao
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Peng Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, China
| | - Jiang Zhou
- Biology Department, Guizhou Normal University, Guiyang, 550000, Guizhou, China.
| | - Yipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
47
|
Creane SE, Carlile SR, Downey D, Weldon S, Dalton JP, Taggart CC. The Impact of Lung Proteases on Snake-Derived Antimicrobial Peptides. Biomolecules 2021; 11:biom11081106. [PMID: 34439773 PMCID: PMC8394243 DOI: 10.3390/biom11081106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 11/16/2022] Open
Abstract
Respiratory infections are a leading cause of global morbidity and mortality and are of significant concern for individuals with chronic inflammatory lung diseases. There is an urgent need for novel antimicrobials. Antimicrobial peptides (AMPs) are naturally occurring innate immune response peptides with therapeutic potential. However, therapeutic development has been hindered by issues with stability and cytotoxicity. Availing of direct drug delivery to the affected site, for example the lung, can reduce unwanted systemic side effects and lower the required dose. As cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) lungs typically exhibit elevated protease levels, the aim of this study was to assess their impact on snake-derived AMPs. Peptide cleavage was determined using SDS-PAGE and antimicrobial and anti-inflammatory activities of neutrophil elastase (NE)-incubated peptides were assessed using a radial diffusion assay (RDA) and an in vitro LPS-induced inflammation model, respectively. Although the snake-derived AMPs were found to be susceptible to cleavage by lung proteases including NE, several retained their function following NE-incubation. This facilitated the design of novel truncated derivatives that retained functionality following NE incubation. Snake-derived AMPs are tractable candidate treatments for use in environments that feature elevated NE levels, such as the CF airways.
Collapse
Affiliation(s)
- Shannice E. Creane
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (S.E.C.); (S.R.C.); (S.W.)
| | - Simon R. Carlile
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (S.E.C.); (S.R.C.); (S.W.)
| | - Damian Downey
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK;
| | - Sinéad Weldon
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (S.E.C.); (S.R.C.); (S.W.)
| | - John P. Dalton
- Zoology Department, School of Natural Sciences, Centre for One Health, Ryan Institute, National University of Ireland Galway, H91 TK33 Galway, Ireland;
- School of Biological Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (S.E.C.); (S.R.C.); (S.W.)
- Correspondence:
| |
Collapse
|
48
|
Recent developments on production, purification and biological activity of marine peptides. Food Res Int 2021; 147:110468. [PMID: 34399466 DOI: 10.1016/j.foodres.2021.110468] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/18/2021] [Accepted: 05/23/2021] [Indexed: 12/11/2022]
Abstract
Marine peptides are one of the richest sources of structurally diverse bioactive compounds and a considerable attention has been drawn towards their production and bioactivity. However, there is a paucity in consolidation of emerging trends encompassing both production techniques and biological application. Herein, we intend to review the recent advancements on different production, purification and identification technologies used for marine peptides along with presenting their potential health benefits. Bibliometric analysis revealed a growing number of scientific publications on marine peptides (268 documents per year) with both Asia (37.2%) and Europe (33.1%) being the major contributors. Extraction and purification by ultrafiltration and enzymatic hydrolysis, followed by identification by chromatographic techniques coupled with an appropriate detector could yield a high content of peptides with improved bioactivity. Moreover, the multifunctional health benefits exerted by marine peptides including anti-microbial, antioxidant, anti-hypertension, anti-diabetes and anti-cancer along with their structure-activity relationship were presented. The future perspective on marine peptide research should focus on finding improved separation and purification technologies with enhanced selectivity and resolution for obtaining more novel peptides with high yield and low cost. In addition, by employing encapsulation strategies such as nanoemulsion and nanoliposome, oral bioavailability and bioactivity of peptides can be greatly enhanced. Also, the potential health benefits that are demonstrated by in vitro and in vivo models should be validated by conducting human clinical trials for a technology transfer from bench to bedside.
Collapse
|
49
|
Yang Y, Han X, Chen Y, Wu J, Li M, Yang H, Xu W, Wei L. EGCG Induces Pro-inflammatory Response in Macrophages to Prevent Bacterial Infection through the 67LR/p38/JNK Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:5638-5651. [PMID: 33993695 DOI: 10.1021/acs.jafc.1c01353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Extensive studies focused on the therapeutic efficacy of epigallocatechin-3-gallate (EGCG) against bacterial infection. However, little is known about its prophylactic efficacy against bacterial infection. Herein, we found that EGCG showed an effective prophylactic efficacy against bacterial infection with a broad spectrum, including Gram-negative, Gram-positive, and drug-resistant bacteria. Pretreatment with EGCG through intraperitoneal injection, intravenous injection, or intragastric administration significantly reduced the bacterial load, inflammatory response, and mortality in mouse abdominal infection models induced by bacterial inoculation or cecal ligation and puncture. Pretreatment with EGCG by intraperitoneal injection significantly increased the numbers of neutrophils and monocytes/macrophages in the abdominal cavity and peripheral blood of mice, and depletion of neutrophils and monocytes/macrophages by specific antibodies or chemical drugs obviously increased the bacterial load in mice. Of note, EGCG did not directly induce neutrophil and macrophage migration, and it just induced phagocyte migration in the presence of macrophages in a co-cultured system, implying that EGCG-induced phagocyte migration relies on its immunoregulatory effects on macrophages. EGCG markedly induced the production of cytokines and chemokines in macrophages and mouse peritoneal lavage, including tumor necrosis factor-α (TNF-α), interleukin-1 β (IL-1β), IL-6, CXC chemokine ligands 1 and 2 (CXCL1 and 2), and monocyte chemotactic protein-1 (MCP-1). EGCG significantly induced the phosphorylation of p38 and JNK mitogen-activated protein kinases (MAPKs) in macrophages, and inhibition of p38 and JNK MAPKs markedly reduced EGCG-induced chemokine and cytokine production. Anti-67-kDa laminin receptor (67LR) antibody treatment significantly reduced EGCG-induced chemokine production and p38 and JNK phosphorylation in macrophages. Together, EGCG showed an obvious prophylactic efficacy against bacterial infection by inducing a pro-inflammatory response in macrophages through the 67LR/p38/JNK signaling pathway, supporting the further development of EGCG as a potent prophylaxis for bacterial infection and providing new clues to understand the healthcare function of green tea.
Collapse
Affiliation(s)
- Yang Yang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Xiaoyang Han
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Yue Chen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jing Wu
- School of Basic Medical Sciences, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Min Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Hailong Yang
- School of Basic Medical Sciences, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Wei Xu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Lin Wei
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
50
|
Antimicrobial, anti-biofilm properties of three naturally occurring antimicrobial peptides against spoilage bacteria, and their synergistic effect with chemical preservatives in food storage. Food Control 2021. [DOI: 10.1016/j.foodcont.2020.107729] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|