1
|
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
|
2
|
Nagib M, Sayed AM, Korany AH, Abdelkader K, Shari FH, Mackay WG, Rateb ME. Human Defensins: Structure, Function, and Potential as Therapeutic Antimicrobial Agents with Highlights Against SARS CoV-2. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10436-8. [PMID: 39693007 DOI: 10.1007/s12602-024-10436-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
The human defensins are a group of cationic antimicrobial peptides that range in size from 2 to 5 kDa and share a common structural motif of six disulphide-linked cysteines. Several naturally occurring human α- and β-defensins have been identified over the past two decades. They have a wide variety of antimicrobial effects, and their potential to avoid the development of resistance to antimicrobial treatment makes them attractive as therapeutic agents. Human defensins have recently been the focus of medical and molecular biology studies due to their promising application in medicine and the pharmaceutical industry. This work aims to provide a comprehensive summary of the current developments of human defensins, including their identification, categorization, molecular features, expression, modes of action, and potential application in medical settings. Current obstacles and future opportunities for using human defensins are also covered. Furthermore, we shed light on the potential of this class as an antiviral agent, particularly against SARS CoV-2, by providing an in silico-based investigation of their plausible mechanisms of action.
Collapse
Affiliation(s)
- Maryam Nagib
- School of Computing Engineering and Physical Sciences, University of the West of Scotland, Paisley, Scotland, PA12BE, UK
| | - Ahmed M Sayed
- Department of Pharmacognosy, College of Pharmacy, Almaaqal University, Basrah, 61014, Iraq
| | - Ahmed H Korany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Nahda University, Beni Suef, 62513, Egypt
| | - Karim Abdelkader
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62511, Egypt
| | - Falah H Shari
- Department of Clinical Biochemistry, College of Pharmacy, Almaaqal University, Basrah, 61014, Iraq
| | - William G Mackay
- School of Health and Life Sciences, University of the West of Scotland, Blantyre, Glasgow, G72 0LH, UK
| | - Mostafa E Rateb
- School of Computing Engineering and Physical Sciences, University of the West of Scotland, Paisley, Scotland, PA12BE, UK.
| |
Collapse
|
3
|
Lakshmaiah Narayana J, Mechesso AF, Rather IIG, Zarena D, Luo J, Xie J, Wang G. Origami of KR-12 Designed Antimicrobial Peptides and Their Potential Applications. Antibiotics (Basel) 2024; 13:816. [PMID: 39334990 PMCID: PMC11429261 DOI: 10.3390/antibiotics13090816] [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: 07/31/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
This review describes the discovery, structure, activity, engineered constructs, and applications of KR-12, the smallest antibacterial peptide of human cathelicidin LL-37, the production of which can be induced under sunlight or by vitamin D. It is a moonlighting peptide that shows both antimicrobial and immune-regulatory effects. Compared to LL-37, KR-12 is extremely appealing due to its small size, lack of toxicity, and narrow-spectrum antimicrobial activity. Consequently, various KR-12 peptides have been engineered to tune peptide activity and stability via amino acid substitution, end capping, hybridization, conjugation, sidechain stapling, and backbone macrocyclization. We also mention recently discovered peptides KR-8 and RIK-10 that are shorter than KR-12. Nano-formulation provides an avenue to targeted delivery, controlled release, and increased bioavailability. In addition, KR-12 has been covalently immobilized on biomaterials/medical implants to prevent biofilm formation. These constructs with enhanced potency and stability are demonstrated to eradicate drug-resistant pathogens, disrupt preformed biofilms, neutralize endotoxins, and regulate host immune responses. Also highlighted are the safety and efficacy of these peptides in various topical and systemic animal models. Finaly, we summarize the achievements and discuss future developments of KR-12 peptides as cosmetic preservatives, novel antibiotics, anti-inflammatory peptides, and microbiota-restoring agents.
Collapse
Affiliation(s)
- Jayaram Lakshmaiah Narayana
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore 560078, India
| | - Abraham Fikru Mechesso
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
| | - Imran Ibni Gani Rather
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
| | - D Zarena
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
- College of Engineering, Jawaharlal Nehru Technological University, Anantapur 515002, India
| | - Jinghui Luo
- Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Guangshun Wang
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
4
|
Wu R, Pettersson C, Demirel I. Testosterone increases the virulence traits of uropathogenic Escherichia coli. Front Microbiol 2024; 15:1422747. [PMID: 38863749 PMCID: PMC11165178 DOI: 10.3389/fmicb.2024.1422747] [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: 04/24/2024] [Accepted: 05/15/2024] [Indexed: 06/13/2024] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is the most common cause of urinary tract infections (UTIs) in humans. Testosterone negatively impacts UTIs by affecting the immune response, leading to higher susceptibility of chronic cystitis in individuals with elevated testosterone levels, regardless of gender. Current research is mostly focused on how testosterone affects the host response to UPEC, but not so much is known about how testosterone directly affect UPEC virulence. The aim of the present study was to investigate the impact of testosterone exposure on the virulence of UPEC. We found that testosterone directly increases UPEC growth, endotoxin release and biofilm formation. We also found that testosterone-stimulated CFT073 increased colonization and invasion of bladder epithelial cells. Testosterone-stimulated CFT073 also increased the release of IL-1β and LDH from bladder epithelial cells. Additionally, by using a Caenorhabditis elegans survival assay we also showed that testosterone decreased the survival of CFT073 infected C. elegans worms. Taken together, our findings show that testosterone directly increases the virulence traits of UPEC.
Collapse
Affiliation(s)
- Rongrong Wu
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Carolina Pettersson
- School of Medical Sciences, Örebro University, Örebro, Sweden
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Isak Demirel
- School of Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
5
|
Fontanot A, Ellinger I, Unger WWJ, Hays JP. A Comprehensive Review of Recent Research into the Effects of Antimicrobial Peptides on Biofilms-January 2020 to September 2023. Antibiotics (Basel) 2024; 13:343. [PMID: 38667019 PMCID: PMC11047476 DOI: 10.3390/antibiotics13040343] [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: 03/11/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/29/2024] Open
Abstract
Microbial biofilm formation creates a persistent and resistant environment in which microorganisms can survive, contributing to antibiotic resistance and chronic inflammatory diseases. Increasingly, biofilms are caused by multi-drug resistant microorganisms, which, coupled with a diminishing supply of effective antibiotics, is driving the search for new antibiotic therapies. In this respect, antimicrobial peptides (AMPs) are short, hydrophobic, and amphipathic peptides that show activity against multidrug-resistant bacteria and biofilm formation. They also possess broad-spectrum activity and diverse mechanisms of action. In this comprehensive review, 150 publications (from January 2020 to September 2023) were collected and categorized using the search terms 'polypeptide antibiotic agent', 'antimicrobial peptide', and 'biofilm'. During this period, a wide range of natural and synthetic AMPs were studied, of which LL-37, polymyxin B, GH12, and Nisin were the most frequently cited. Furthermore, although many microbes were studied, Staphylococcus aureus and Pseudomonas aeruginosa were the most popular. Publications also considered AMP combinations and the potential role of AMP delivery systems in increasing the efficacy of AMPs, including nanoparticle delivery. Relatively few publications focused on AMP resistance. This comprehensive review informs and guides researchers about the latest developments in AMP research, presenting promising evidence of the role of AMPs as effective antimicrobial agents.
Collapse
Affiliation(s)
- Alessio Fontanot
- Department of Medical Microbiology & Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (A.F.); (W.W.J.U.)
- Department of Pediatrics, Laboratory of Pediatrics, Erasmus University Medical Center Rotterdam, Sophia Children’s Hospital, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Isabella Ellinger
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Währinger Gürtel 18–20, 1090 Vienna, Austria;
| | - Wendy W. J. Unger
- Department of Medical Microbiology & Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (A.F.); (W.W.J.U.)
- Department of Pediatrics, Laboratory of Pediatrics, Erasmus University Medical Center Rotterdam, Sophia Children’s Hospital, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - John P. Hays
- Department of Medical Microbiology & Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (A.F.); (W.W.J.U.)
| |
Collapse
|
6
|
Von Vietinghoff S, Shevchuk O, Dobrindt U, Engel DR, Jorch SK, Kurts C, Miethke T, Wagenlehner F. The global burden of antimicrobial resistance - urinary tract infections. Nephrol Dial Transplant 2024; 39:581-588. [PMID: 37891013 DOI: 10.1093/ndt/gfad233] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Indexed: 10/29/2023] Open
Abstract
Antimicrobial resistance (AMR) has emerged as a significant global healthcare problem. Antibiotic use has accelerated the physiologic process of AMR, particularly in Gram-negative pathogens. Urinary tract infections (UTIs) are predominantly of a Gram-negative nature. Uropathogens are evolutionarily highly adapted and selected strains with specific virulence factors, suggesting common mechanisms in how bacterial cells acquire virulence and AMR factors. The simultaneous increase in resistance and virulence is a complex and context-dependent phenomenon. Among known AMR mechanisms, the plenitude of different β-lactamases is especially prominent. The risk for AMR in UTIs varies in different patient populations. A history of antibiotic consumption and the physiology of urinary flow are major factors that shape AMR prevalence. The urinary tract is in close crosstalk with the microbiome of other compartments, including the gut and genital tracts. In addition, pharmacokinetic properties and the physiochemical composition of urinary compartments can contribute to the emergence of AMR. Alternatives to antibiotic treatment and a broader approach to address bacterial infections are needed. Among the various alternatives studied, antimicrobial peptides and bacteriophage treatment appear to be highly promising approaches. We herein summarize the present knowledge of clinical and microbiological AMR in UTIs and discuss innovative approaches, namely new risk prediction tools and the use of non-antibiotic approaches to defend against uropathogenic microbes.
Collapse
Affiliation(s)
- Sibylle Von Vietinghoff
- University Hospital Bonn, Medical Clinic 1, Section for Nephrology and University Bonn, Germany
| | - Olga Shevchuk
- University Duisburg-Essen, University Hospital Essen, Institute of Experimental Immunology and Imaging, Department of Immunodynamics, Essen, Germany
| | - Ulrich Dobrindt
- University of Münster, Institute of Hygiene, Münster, Germany
| | - Daniel Robert Engel
- University Duisburg-Essen, University Hospital Essen, Institute of Experimental Immunology and Imaging, Department of Immunodynamics, Essen, Germany
| | | | | | - Thomas Miethke
- Medical Faculty of Mannheim University of Heidelberg, Institute for Medical Microbiology and Hygiene, Heidelberg, Germany
- Medical Faculty of Mannheim, Heidelberg University, Institute for Medical Microbiology and Hygiene, Mannheim, Germany
| | - Florian Wagenlehner
- Justus-Liebig University Giessen, Clinic for Urology, Paediatric Urology and Andrology, Giessen, Germany
| |
Collapse
|
7
|
Huang S, Su G, Jiang S, Chen L, Huang J, Yang F. New N-Terminal Fatty-Acid-Modified Melittin Analogs with Potent Biological Activity. Int J Mol Sci 2024; 25:867. [PMID: 38255940 PMCID: PMC10815238 DOI: 10.3390/ijms25020867] [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: 11/15/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Melittin, a natural antimicrobial peptide, has broad-spectrum antimicrobial activity. This has resulted in it gaining increasing attention as a potential antibiotic alternative; however, its practical use has been limited by its weak antimicrobial activity, high hemolytic activity, and low proteolytic stability. In this study, N-terminal fatty acid conjugation was used to develop new melittin-derived lipopeptides (MDLs) to improve the characteristics of melittin. Our results showed that compared with native melittin, the antimicrobial activity of MDLs was increased by 2 to 16 times, and the stability of these MDLs against trypsin and pepsin degradation was increased by 50 to 80%. However, the hemolytic activity of the MDLs decreased when the length of the carbon chain of fatty acids exceeded 10. Among the MDLs, the newly designed analog Mel-C8 showed optimal antimicrobial activity and protease stability. The antimicrobial mechanism studied revealed that the MDLs showed a rapid bactericidal effect by interacting with lipopolysaccharide (LPS) or lipoteichoic acid (LTA) and penetrating the bacterial cell membrane. In conclusion, we designed and synthesized a new class of MDLs with potent antimicrobial activity, high proteolytic stability, and low hemolytic activity through N-terminal fatty acid conjugation.
Collapse
Affiliation(s)
- Sheng Huang
- Animal Nutrition Institute, Chongqing Academy of Animal Science, Chongqing 402460, China; (S.H.); (L.C.)
| | - Guoqi Su
- Animal Nutrition Institute, Chongqing Academy of Animal Science, Chongqing 402460, China; (S.H.); (L.C.)
| | - Shan Jiang
- Animal Nutrition Institute, Chongqing Academy of Animal Science, Chongqing 402460, China; (S.H.); (L.C.)
| | - Li Chen
- Animal Nutrition Institute, Chongqing Academy of Animal Science, Chongqing 402460, China; (S.H.); (L.C.)
| | - Jinxiu Huang
- Animal Nutrition Institute, Chongqing Academy of Animal Science, Chongqing 402460, China; (S.H.); (L.C.)
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing 402460, China
| | - Feiyun Yang
- Animal Nutrition Institute, Chongqing Academy of Animal Science, Chongqing 402460, China; (S.H.); (L.C.)
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing 402460, China
| |
Collapse
|
8
|
Mwangi J, Kamau PM, Thuku RC, Lai R. Design methods for antimicrobial peptides with improved performance. Zool Res 2023; 44:1095-1114. [PMID: 37914524 PMCID: PMC10802102 DOI: 10.24272/j.issn.2095-8137.2023.246] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/20/2023] [Indexed: 11/03/2023] Open
Abstract
The recalcitrance of pathogens to traditional antibiotics has made treating and eradicating bacterial infections more difficult. In this regard, developing new antimicrobial agents to combat antibiotic-resistant strains has become a top priority. Antimicrobial peptides (AMPs), a ubiquitous class of naturally occurring compounds with broad-spectrum antipathogenic activity, hold significant promise as an effective solution to the current antimicrobial resistance (AMR) crisis. Several AMPs have been identified and evaluated for their therapeutic application, with many already in the drug development pipeline. Their distinct properties, such as high target specificity, potency, and ability to bypass microbial resistance mechanisms, make AMPs a promising alternative to traditional antibiotics. Nonetheless, several challenges, such as high toxicity, lability to proteolytic degradation, low stability, poor pharmacokinetics, and high production costs, continue to hamper their clinical applicability. Therefore, recent research has focused on optimizing the properties of AMPs to improve their performance. By understanding the physicochemical properties of AMPs that correspond to their activity, such as amphipathicity, hydrophobicity, structural conformation, amino acid distribution, and composition, researchers can design AMPs with desired and improved performance. In this review, we highlight some of the key strategies used to optimize the performance of AMPs, including rational design and de novo synthesis. We also discuss the growing role of predictive computational tools, utilizing artificial intelligence and machine learning, in the design and synthesis of highly efficacious lead drug candidates.
Collapse
Affiliation(s)
- James Mwangi
- Key Laboratory of Bioactive Peptides of Yunnan Province, Engineering Laboratory of Peptides of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Centre for Non-Human Primates, Kunming Primate Research Centre, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Centre, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Peter Muiruri Kamau
- Key Laboratory of Bioactive Peptides of Yunnan Province, Engineering Laboratory of Peptides of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Centre for Non-Human Primates, Kunming Primate Research Centre, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Centre, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Rebecca Caroline Thuku
- Key Laboratory of Bioactive Peptides of Yunnan Province, Engineering Laboratory of Peptides of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Centre for Non-Human Primates, Kunming Primate Research Centre, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Centre, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ren Lai
- Key Laboratory of Bioactive Peptides of Yunnan Province, Engineering Laboratory of Peptides of Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Centre for Non-Human Primates, Kunming Primate Research Centre, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Sino-African Joint Research Centre, New Cornerstone Science Institute, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- Centre for Evolution and Conservation Biology, Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou, Guangdong 511458, China. E-mail:
| |
Collapse
|
9
|
Tripathi AK, Singh J, Trivedi R, Ranade P. Shaping the Future of Antimicrobial Therapy: Harnessing the Power of Antimicrobial Peptides in Biomedical Applications. J Funct Biomater 2023; 14:539. [PMID: 37998108 PMCID: PMC10672284 DOI: 10.3390/jfb14110539] [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: 09/25/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023] Open
Abstract
Antimicrobial peptides (AMPs) have emerged as a promising class of bioactive molecules with the potential to combat infections associated with medical implants and biomaterials. This review article aims to provide a comprehensive analysis of the role of antimicrobial peptides in medical implants and biomaterials, along with their diverse clinical applications. The incorporation of AMPs into various medical implants and biomaterials has shown immense potential in mitigating biofilm formation and preventing implant-related infections. We review the latest advancements in biomedical sciences and discuss the AMPs that were immobilized successfully to enhance their efficacy and stability within the implant environment. We also highlight successful examples of AMP coatings for the treatment of surgical site infections (SSIs), contact lenses, dental applications, AMP-incorporated bone grafts, urinary tract infections (UTIs), medical implants, etc. Additionally, we discuss the potential challenges and prospects of AMPs in medical implants, such as effectiveness, instability and implant-related complications. We also discuss strategies that can be employed to overcome the limitations of AMP-coated biomaterials for prolonged longevity in clinical settings.
Collapse
Affiliation(s)
- Amit Kumar Tripathi
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.T.); (P.R.)
| | - Jyotsana Singh
- Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Rucha Trivedi
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.T.); (P.R.)
| | - Payal Ranade
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.T.); (P.R.)
| |
Collapse
|
10
|
Mourenza A, Ganesan R, Camarero JA. Resistance is futile: targeting multidrug-resistant bacteria with de novo Cys-rich cyclic polypeptides. RSC Chem Biol 2023; 4:722-735. [PMID: 37799576 PMCID: PMC10549238 DOI: 10.1039/d3cb00015j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/27/2023] [Indexed: 10/07/2023] Open
Abstract
The search for novel antimicrobial agents to combat microbial pathogens is intensifying in response to rapid drug resistance development to current antibiotic therapeutics. The use of disulfide-rich head-to-tail cyclized polypeptides as molecular frameworks for designing a new type of peptide antibiotics is gaining increasing attention among the scientific community and the pharmaceutical industry. The use of macrocyclic peptides, further constrained by the presence of several disulfide bonds, makes these peptide frameworks remarkably more stable to thermal, biological, and chemical degradation showing better activities when compared to their linear analogs. Many of these novel peptide scaffolds have been shown to have a high tolerance to sequence variability in those residues not involved in disulfide bonds, able to cross biological membranes, and efficiently target complex biomolecular interactions. Hence, these unique properties make the use of these scaffolds ideal for many biotechnological applications, including the design of novel peptide antibiotics. This article provides an overview of the new developments in the use of several disulfide-rich cyclic polypeptides, including cyclotides, θ-defensins, and sunflower trypsin inhibitor peptides, among others, in the development of novel antimicrobial peptides against multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Alvaro Mourenza
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy Los Angeles CA90033 USA +1-(323) 442-1417
| | - Rajasekaran Ganesan
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy Los Angeles CA90033 USA +1-(323) 442-1417
| | - Julio A Camarero
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy Los Angeles CA90033 USA +1-(323) 442-1417
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California Los Angeles CA90033 USA
| |
Collapse
|
11
|
Wei Z, Rolle MW, Camesano TA. Characterization of LL37 Binding to Collagen through Peptide Modification with a Collagen-Binding Domain. ACS OMEGA 2023; 8:35370-35381. [PMID: 37779975 PMCID: PMC10536065 DOI: 10.1021/acsomega.3c05328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023]
Abstract
Collagen-based biomaterials loaded with antimicrobial peptides (AMPs) present a promising approach for promoting wound healing while providing protection against infections. In our previous work, we modified the AMP LL37 by incorporating a collagen-binding domain (cCBD) as an anchoring unit for collagen-based wound dressings. We demonstrated that cCBD-modified LL37 (cCBD-LL37) exhibited improved retention on collagen after washing with PBS. However, the binding mechanism of cCBD-LL37 to collagen remained to be elucidated. In this study, we found that cCBD-LL37 showed a slightly higher affinity for collagen compared to LL37. Our results indicated that cCBD inhibited cCBD-LL37 binding to collagen but did not fully eliminate the binding. This suggests that cCBD-LL37 binding to collagen may involve more than just one-site-specific binding through the collagen-binding domain, with non-specific interactions also playing a role. Electrostatic studies revealed that both LL37 and cCBD-LL37 interact with collagen via long-range electrostatic forces, initiating low-affinity binding that transitions to close-range or hydrophobic interactions. Circular dichroism analysis showed that cCBD-LL37 exhibited enhanced structural stability compared to LL37 under varying ionic strengths and pH conditions, implying potential improvements in antimicrobial activity. Moreover, we demonstrated that the release of LL37 and cCBD-LL37 into the surrounding medium was influenced by the electrostatic environment, but cCBD could enhance the retention of peptide on collagen scaffolds. Collectively, these results provide important insights into cCBD-modified AMP-binding mechanisms and suggest that the addition of cCBD may enhance peptide structural stability and retention under varying electrostatic conditions.
Collapse
Affiliation(s)
- Ziqi Wei
- Department
of Chemical Engineering, Worcester Polytechnic
Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| | - Marsha W. Rolle
- Department
of Biomedical Engineering, Worcester Polytechnic
Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| | - Terri A. Camesano
- Department
of Chemical Engineering, Worcester Polytechnic
Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| |
Collapse
|
12
|
Lindblad A, Wu R, Persson K, Demirel I. The Role of NLRP3 in Regulation of Antimicrobial Peptides and Estrogen Signaling in UPEC-Infected Bladder Epithelial Cells. Cells 2023; 12:2298. [PMID: 37759520 PMCID: PMC10526908 DOI: 10.3390/cells12182298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The NLRP3 inflammasome, estrogen and antimicrobial peptides have all been found to have a vital role in the protection of the bladder urothelium. However, the interdependence between these protective factors during a bladder infection is currently unknown. Our aim was to investigate the role of NLRP3 in the regulation of antimicrobial peptides and estrogen signaling in bladder epithelial cells during a UPEC infection. Human bladder epithelial cells and CRISPR/Cas9-generated NLRP3-deficient cells were stimulated with the UPEC strain CFT073 and estradiol. The gene and protein expression were evaluated with microarray, qRT-PCR, western blot and ELISA. Microarray results showed that the expression of most antimicrobial peptides was reduced in CFT073-infected NLRP3-deficient cells compared to Cas9 control cells. Conditioned medium from NLRP3-deficient cells also lost the ability to suppress CFT073 growth. Moreover, NLRP3-deficient cells had lower basal release of Beta-defensin-1, Beta-defensin-2 and RNase7. The ability of estradiol to induce an increased expression of antimicrobial peptides was also abrogated in NLRP3-deficient cells. The decreased antimicrobial peptide expression might be linked to the observed reduced expression and activity of estradiol receptor beta in NLRP3-deficient cells. This study suggests that NLRP3 may regulate the release and expression of antimicrobial peptides and affect estrogen signaling in bladder epithelial cells.
Collapse
Affiliation(s)
| | | | | | - Isak Demirel
- School of Medical Sciences, Örebro University, 701 82 Örebro, Sweden; (A.L.); (R.W.); (K.P.)
| |
Collapse
|
13
|
Liu C, Henning-Knechtel A, Österlund N, Wu J, Wang G, Gräslund RAO, Kirmizialtin S, Luo J. Oligomer Dynamics of LL-37 Truncated Fragments Probed by α-Hemolysin Pore and Molecular Simulations. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206232. [PMID: 37170734 DOI: 10.1002/smll.202206232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 04/01/2023] [Indexed: 05/13/2023]
Abstract
Oligomerization of antimicrobial peptides (AMPs) is critical in their effects on pathogens. LL-37 and its truncated fragments are widely investigated regarding their structures, antimicrobial activities, and application, such as developing new antibiotics. Due to the small size and weak intermolecular interactions of LL-37 fragments, it is still elusive to establish the relationship between oligomeric states and antimicrobial activities. Here, an α-hemolysin nanopore, mass spectrometry (MS), and molecular dynamic (MD) simulations are used to characterize the oligomeric states of two LL-37 fragments. Nanopore studies provide evidence of trapping events related to the oligomer formation and provide further details on their stabilities, which are confirmed by MS and MD simulations. Furthermore, simulation results reveal the molecular basis of oligomer dynamics and states of LL-37 fragments. This work provides unique insights into the relationship between the oligomer dynamics of AMPs and their antimicrobial activities at the single-molecule level. The study demonstrates how integrating methods allows deciphering single molecule level understanding from nanopore sensing approaches.
Collapse
Affiliation(s)
- Chang Liu
- Department of Biology and Chemistry, Paul Scherrer Institute, Villigen, 5232, Switzerland
| | - Anja Henning-Knechtel
- Science Division, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, 129188, UAE
| | - Nicklas Österlund
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 106 91, Sweden
| | - Jinming Wu
- Department of Biology and Chemistry, Paul Scherrer Institute, Villigen, 5232, Switzerland
| | - Guangshun Wang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | | | - Serdal Kirmizialtin
- Science Division, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, 129188, UAE
| | - Jinghui Luo
- Department of Biology and Chemistry, Paul Scherrer Institute, Villigen, 5232, Switzerland
| |
Collapse
|
14
|
Lee YCJ, Javdan B, Cowan A, Smith K. More than skin deep: cyclic peptides as wound healing and cytoprotective compounds. Front Cell Dev Biol 2023; 11:1195600. [PMID: 37325572 PMCID: PMC10267460 DOI: 10.3389/fcell.2023.1195600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
The prevalence and cost of wounds pose a challenge to patients as well as the healthcare system. Wounds can involve multiple tissue types and, in some cases, become chronic and difficult to treat. Comorbidities may also decrease the rate of tissue regeneration and complicate healing. Currently, treatment relies on optimizing healing factors rather than administering effective targeted therapies. Owing to their enormous diversity in structure and function, peptides are among the most prevalent and biologically important class of compounds and have been investigated for their wound healing bioactivities. A class of these peptides, called cyclic peptides, confer stability and improved pharmacokinetics, and are an ideal source of wound healing therapeutics. This review provides an overview of cyclic peptides that have been shown to promote wound healing in various tissues and in model organisms. In addition, we describe cytoprotective cyclic peptides that mitigate ischemic reperfusion injuries. Advantages and challenges in harnessing the healing potential for cyclic peptides from a clinical perspective are also discussed. Cyclic peptides are a potentially attractive category of wound healing compounds and more research in this field could not only rely on design as mimetics but also encompass de novo approaches as well.
Collapse
Affiliation(s)
- Ying-Chiang J. Lee
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Bahar Javdan
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Alexis Cowan
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Keith Smith
- Merck & Co., Inc., Kenilworth, NJ, United States
| |
Collapse
|
15
|
Sowers A, Wang G, Xing M, Li B. Advances in Antimicrobial Peptide Discovery via Machine Learning and Delivery via Nanotechnology. Microorganisms 2023; 11:1129. [PMID: 37317103 PMCID: PMC10223199 DOI: 10.3390/microorganisms11051129] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 06/16/2023] Open
Abstract
Antimicrobial peptides (AMPs) have been investigated for their potential use as an alternative to antibiotics due to the increased demand for new antimicrobial agents. AMPs, widely found in nature and obtained from microorganisms, have a broad range of antimicrobial protection, allowing them to be applied in the treatment of infections caused by various pathogenic microorganisms. Since these peptides are primarily cationic, they prefer anionic bacterial membranes due to electrostatic interactions. However, the applications of AMPs are currently limited owing to their hemolytic activity, poor bioavailability, degradation from proteolytic enzymes, and high-cost production. To overcome these limitations, nanotechnology has been used to improve AMP bioavailability, permeation across barriers, and/or protection against degradation. In addition, machine learning has been investigated due to its time-saving and cost-effective algorithms to predict AMPs. There are numerous databases available to train machine learning models. In this review, we focus on nanotechnology approaches for AMP delivery and advances in AMP design via machine learning. The AMP sources, classification, structures, antimicrobial mechanisms, their role in diseases, peptide engineering technologies, currently available databases, and machine learning techniques used to predict AMPs with minimal toxicity are discussed in detail.
Collapse
Affiliation(s)
- Alexa Sowers
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
- School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA
| | - Guangshun Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198, USA
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Bingyun Li
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
16
|
Yang M, Liu S, Zhang C. Antimicrobial peptides with antiviral and anticancer properties and their modification and nanodelivery systems. CURRENT RESEARCH IN BIOTECHNOLOGY 2023. [DOI: 10.1016/j.crbiot.2023.100121] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
|
17
|
Muhammad T, Strömstedt AA, Gunasekera S, Göransson U. Transforming Cross-Linked Cyclic Dimers of KR-12 into Stable and Potent Antimicrobial Drug Leads. Biomedicines 2023; 11:biomedicines11020504. [PMID: 36831040 PMCID: PMC9953701 DOI: 10.3390/biomedicines11020504] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Is it possible to enhance structural stability and biological activity of KR-12, a truncated antimicrobial peptide derived from the human host defense peptide LL-37? Based on the mapping of essential residues in KR-12, we have designed backbone-cyclized dimers, cross-linked via a disulfide bond to improve peptide stability, while at the same time improving on-target activity. Circular dichroism showed that each of the dimers adopts a primarily alpha-helical conformation (55% helical content) when bound to lyso-phosphatidylglycerol micelles, indicating that the helical propensity of the parent peptide is maintained in the new cross-linked cyclic form. Compared to KR-12, one of the cross-linked dimers showed 16-fold more potent antimicrobial activity against human pathogens Pseudomonas aeruginosa, Staphylococcus aureus, and Candida albicans and 8-fold increased activity against Escherichia coli. Furthermore, these peptides retained antimicrobial activity at physiologically relevant conditions, including in the presence of salts and in human serum, and with selective Gram-negative antibacterial activity in rich growth media. In addition to giving further insight into the structure-activity relationship of KR-12, the current work demonstrates that by combining peptide stabilization strategies (dimerization, backbone cyclization, and cross-linking via a disulfide bond), KR-12 can be engineered into a potent antimicrobial peptide drug lead with potential utility in a therapeutic context.
Collapse
|
18
|
Decker AP, Su Y, Mishra B, Verma A, Lushnikova T, Xie J, Wang G. Peptide Stability Is Important but Not a General Requirement for Antimicrobial and Antibiofilm Activity In Vitro and In Vivo. Mol Pharm 2023; 20:738-749. [PMID: 36485036 DOI: 10.1021/acs.molpharmaceut.2c00918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Peptide stability to proteases has been a major requirement for developing peptide therapeutics. This study investigates the effects of peptide stability on antimicrobial and antibiofilm activity under various conditions. For this purpose, two human cathelicidin-derived peptides differing in stability to proteases were utilized. While GF-17, a peptide derived from the major antimicrobial region of human LL-37, can be rapidly cleaved by proteases, the engineered peptide 17BIPHE2 is resistant to multiple proteases. In the standard antimicrobial susceptibility, killing kinetics, and membrane permeabilization assays conducted in vitro using planktonic bacteria, these two peptides displayed similar potency. The two peptides were also similarly active against methicillin-resistant Staphylococcus aureus (MRSA) USA300 prior to biofilm formation. However, 17BIPHE2 was superior to GF-17 in disrupting preformed biofilms probably due to both enhanced stability and slightly higher DNA binding capacity. In a wax moth model, 17BIPHE2 better protected insects from MRSA infection-caused death than GF-17, consistent with the slower degradation of 17BIPHE2 than GF-17. Here, peptide antimicrobial activity was found to be critical for in vivo efficacy. When incorporated in the nanofiber/microneedle delivery device, GF-17 and 17BIPHE2 displayed a similar effect in eliminating MRSA in murine chronic wounds, underscoring the advantage of nanofibers in protecting the peptide from degradation. Since nanoformulation can ease the requirement of peptide stability, it opens the door to a direct use of natural peptides or their cocktails for antimicrobial treatment, accelerating the search of effective antibiofilm peptides to treat chronic wounds.
Collapse
Affiliation(s)
- Aaron P Decker
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Biswajit Mishra
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Atul Verma
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Tamara Lushnikova
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Guangshun Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| |
Collapse
|