1
|
Li L, Zeng Y, Tian M, Cao H, Qiu Z, Guo G, Shen F, Wang Y, Peng J. The antimicrobial peptide Cec4 has therapeutic potential against clinical carbapenem-resistant Klebsiella pneumoniae. Microbiol Spectr 2025:e0273824. [PMID: 40377314 DOI: 10.1128/spectrum.02738-24] [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: 10/30/2024] [Accepted: 04/08/2025] [Indexed: 05/18/2025] Open
Abstract
The rapid increase in carbapenem-resistant Klebsiella pneumoniae (CRKP) infections, along with the cross-resistance of CRKP to other antibiotics, has created an urgent need for novel therapeutic agents. Among the potential options for next-generation antibiotics, antimicrobial peptides (AMPs) show great promise. In this study, we aimed to elucidate the mechanisms underlying the antibacterial activity against CRKP of an antibacterial peptide named Cecropin-4 (Cec4), which we successfully identified previously. Our results demonstrate that Cec4 not only exhibits rapid antibacterial activity but also effectively inhibits and eradicates bacterial biofilm at a low concentration of 8 µg/mL. Additionally, when used in combination with traditional antibiotics, Cec4 enhances their antibacterial effect. Microscopy techniques, including transmission electron microscopy (TEM), confocal laser scanning microscopy, and scanning electron microscopy (SEM), found that Cec4 destroyed bacteria's cell membrane integrity and increased the membrane permeability (flow cytometry instrument technology further characterization of Cec4 against K. pneumoniae bacteria antibacterial effect). Furthermore, in vitro experiments demonstrated that Cec4 binds to bacterial DNA and RNA of CRKP. Moreover, in vivo studies using a mouse skin wound model confirmed the efficacy of Cec4, and transcriptomic analysis shed light on the molecular mechanisms underlying its antibacterial activity. Based on our findings, Cec4 appears to be a promising candidate for combating CRKP infections.IMPORTANCEThe rapid increase in carbapenem-resistant Klebsiella pneumoniae (CRKP) infections and the serious cross-resistance to multiple antibiotics make the development of new therapeutic drugs urgent. Antimicrobial peptides (AMPs) have attracted much attention as a potential option for the next generation of antibiotics. Previous studies have identified the antimicrobial peptide Cecropin-4 (Cec4), and this study further explored its antimicrobial mechanism against CRKP. Studies have found that Cec4 shows high antibacterial activity at low concentrations, can inhibit and eradicate bacterial biofilms, and can also enhance the efficacy of traditional antibiotics. Its mechanism of action, such as destroying cell membranes and binding nucleic acid, has been revealed by various techniques, and its effectiveness has been confirmed in vivo, providing a promising candidate drug for combating CRKP infection.
Collapse
Affiliation(s)
- Lu Li
- Department of Intensive Care Unit/Centre for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Yang Zeng
- Department of Intensive Care Unit/Centre for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Minfang Tian
- Department of Intensive Care Unit/Centre for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
- Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Huijun Cao
- Department of Intensive Care Unit/Centre for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Zhilang Qiu
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Guo Guo
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Feng Shen
- Department of Intensive Care Unit/Centre for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Yuping Wang
- Department of Intensive Care Unit/Centre for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Jian Peng
- Department of Intensive Care Unit/Centre for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
- Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
- The Engineering Research Center of Health Medicine Biotechnology of Institution of Higher Education of Guizhou Province, Guiyang, Guizhou, People's Republic of China
| |
Collapse
|
2
|
Hadjigol S, Shabani S, Jafari VF, Barlow A, Qiao GG, O'Brien-Simpson NM. Lipidated SNAPP-Stars Target and Kill Multidrug-Resistant Bacteria within Minutes. ACS APPLIED MATERIALS & INTERFACES 2025; 17:25163-25181. [PMID: 40237536 DOI: 10.1021/acsami.5c03839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
The fast emergence of bacteria resistance has already threatened global health, and immediate action is required before the emergence of another global pandemic. Despite substantial progress in the chemical synthesis of novel antimicrobial compounds and advancements in understanding antimicrobial resistance, there has been only a handful of new antibiotics coming to the market. Structurally Nanoengineered Antimicrobial Peptide Polymers (SNAPP-stars) are a new class of antimicrobials. Here, we show that lipidation of lysine-valine 16-armed SNAPP-star, S16 (lipo-SNAPP-star) where the N-terminal arms are conjugated with different fatty acids (caproic, C6, lauric, C12, and stearic acid, C18) enhanced the antimicrobial activity toward S. aureus and MRSA. Lipidation enhanced activity by targeting the SNAPP-stars to the bacterial surface by binding to peptidoglycan, leading to greater inner membrane disruption and depolarization. Lipo-SNAPP-stars killed bacteria in under a minute, whereas vancomycin took >16 h. Lipo-SNAPP-stars were found to preferentially target and kill MRSA rather than S. aureus in a mixed bacteria model. Lipid chain length affected activity, with C6-S16 having greater activity compared to C12-S16 > C18-S16. Lauric and stearic acid enhanced SNAPP-star binding to the bacterial surface and membrane depolarization but impeded SNAPP-stars' ability to transit through the peptidoglycan layer to disrupt the inner membrane. Microbial flow cytometry showed that lipidation aided binding to bacteria via lipoteichoic acid and specifically to peptidoglycan. Further, lipid length enhanced bacterial binding with C18-S16 > C12-S16 > C6-S16 = S16, which contrasts the activity order of C6-S16 > S16 ≫ C12-S16 ≫ C18-S16. Our data demonstrate that lipidation enhances antimicrobial activity by targeting and binding an antimicrobial to peptidoglycan, but increasing lipid length reduces activity by retaining the antimicrobial in the outer layer. Lipidation of SNAPP-stars did not increase cytotoxicity, with C6-S16 having an improved therapeutic index compared to S16. Our data show how lipidation of SNAPP-stars enhances its antimicrobial activity, resulting in a highly biocompatible antimicrobial that targets and kills the "superbug" MRSA within minutes.
Collapse
Affiliation(s)
- Sara Hadjigol
- ACTV Research Group, Division of Basic and Clinical Oral Sciences, The Melbourne Dental School, Royal Dental Hospital, The University of Melbourne, 720 Swanston Street, Carlton, Melbourne, Victoria 3010, Australia
| | - Sadegh Shabani
- Polymer Science Group, Department of Chemical & Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Vianna F Jafari
- Polymer Science Group, Department of Chemical & Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anders Barlow
- Materials Characterisation and Fabrication Platform, Melbourne School of Engineering, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Greg G Qiao
- Polymer Science Group, Department of Chemical & Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Neil M O'Brien-Simpson
- ACTV Research Group, Division of Basic and Clinical Oral Sciences, The Melbourne Dental School, Royal Dental Hospital, The University of Melbourne, 720 Swanston Street, Carlton, Melbourne, Victoria 3010, Australia
| |
Collapse
|
3
|
Liu S, HuiXin E, Xing B. Harnessing from Nature - Evolving Potential of Antimicrobial Peptide. Chembiochem 2025; 26:e202400983. [PMID: 39871592 DOI: 10.1002/cbic.202400983] [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: 12/01/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 01/29/2025]
Abstract
Antimicrobial peptides (AMPs) are recognized as one of the most ancient components of innate immunity, playing a pivotal role as the first line of host defense systems. These evolutionarily conserved molecules have been identified in various organisms, from prokaryotes to humans. AMPs establish a delicate balanced relationship between host and microbes, by simultaneously regulating the biological activities of pathogens and commensal microbes. Given the escalating global concern over antibiotic resistance, there is an urgent need to explore alternative strategies to combat challenging infectious diseases. AMPs have emerged as promising candidates employed in clinical practice due to their sustainable bactericidal properties. Witnessed by deep understanding of AMPs actions toward host and bacteria, the potential applications of AMPs extend far beyond infection control. Emerging developments harnessed natural capabilities of AMPs to optimize their roles in modulating host signaling, treating diverse diseases, advancing biosensing and bioimaging technologies. In this Concept paper, we provide a comprehensive overview of the diversity and properties of AMPs. Additionally, we elaborate on the mechanisms underlying AMP activity and bacterial responses counteracting AMPs' functions. Most importantly, we discuss potential biomedical applications of AMPs and offer perspectives on their future development.
Collapse
Affiliation(s)
- Songhan Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - EveliasYan HuiXin
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Bengang Xing
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, P. R. China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| |
Collapse
|
4
|
Noe MM, Rodríguez JA, Barredo Vacchelli GR, Camperi SA, Franchi AN, Turina AV, Perillo MA, Nolan V. Whey-Derived Antimicrobial Anionic Peptide Interaction with Model Membranes and Cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:242-252. [PMID: 39757468 DOI: 10.1021/acs.langmuir.4c03391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
The present work focuses on one of the possible target mechanisms of action of the anionic antimicrobial peptide β-lg125-135 derived from trypsin hydrolysis of β-lactoglobulin. After confirmation of bactericidal activity against a pathogenic Gram(+) strain and demonstration of the innocuousness on a eukaryotic cell line, we investigated the interaction of β-lg125-135 with monolayers and bilayers of dpPC and dpPC:dpPG as model membranes of eukaryotic and bacterial membranes, respectively. In monolayers, compared to zwitterionic dpPC, in the negatively charged dpPC-dpPG, β-lg125-135 injected into the subphase penetrated up to higher surface pressures and showed greater extents of penetration with increasing concentration in the subphase. Additionally, the rate constants for β-lg125-135 adsorption and desorption were 1 order of magnitude higher, and the resultant thermodynamic association constant was 1 order of magnitude lower. In turn, the compression isotherms of monolayers prepared with the β-lg125-135 present in the mixture spread over the air-water interface, remained in the monolayer and showed positive deviations from ideality, a greater decrease in the surface compressibility modulus, and an increase in the surface potential of both interfaces, more pronounced on dpPC:dpPG. In SUVs, fluorescence anisotropy (FA) assays using DPH and TMA-DPM indicated that β-lg125-135 tended to disrupt the gel phase of dpPC bilayers. Conversely, in dpPC:dpPG, the peptide increased the FA of both probes. These results reflect a relatively high tendency of the β-lg125-135 to approach the negative interface, with a favorable electrostatic orientation but low stability and short residence time. Once inside the membrane, it stiffens dpPG-containing bilayers.
Collapse
Affiliation(s)
- Melania M Noe
- Departamento de Química, Catedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), Córdoba 5000, Argentina
- Instituto de Ciencia y Tecnología de los Alimentos (ICTA), Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Jésica A Rodríguez
- Cátedra de Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Universidad de Buenos Aires (UBA) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
| | - Gabriela R Barredo Vacchelli
- Cátedra de Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Universidad de Buenos Aires (UBA) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
| | - Silvia A Camperi
- Cátedra de Biotecnología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Universidad de Buenos Aires (UBA) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
| | - Anahí N Franchi
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), Córdoba 5000, Argentina
- Departamento de Fisiología, Catedra de Biología Celular, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Anahí V Turina
- Departamento de Química, Catedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), Córdoba 5000, Argentina
- Instituto de Ciencia y Tecnología de los Alimentos (ICTA), Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - María A Perillo
- Departamento de Química, Catedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), Córdoba 5000, Argentina
- Instituto de Ciencia y Tecnología de los Alimentos (ICTA), Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Verónica Nolan
- Departamento de Química, Catedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), Córdoba 5000, Argentina
- Instituto de Ciencia y Tecnología de los Alimentos (ICTA), Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| |
Collapse
|
5
|
Yücesoy SN, Ak T, Öner S, Serdaroğlu S. Serum Catestatin Levels in Patients with Acne Vulgaris: Single-Center Prospective Study. Skin Appendage Disord 2024; 10:490-496. [PMID: 39659650 PMCID: PMC11627537 DOI: 10.1159/000539224] [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: 02/21/2024] [Accepted: 05/03/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Recent studies showed that antimicrobial peptides have an essential role in the pathogenesis of acne vulgaris. This study aims to investigate serum catestatin levels in acne vulgaris patients and focuses on the change in serum levels after systemic isotretinoin therapy. Methods This prospective study includes 101 acne vulgaris patients and 28 healthy controls. Serum catestatin levels were measured and compared between acne vulgaris and control group patients. Also, serum catestatin levels were measured again at the 24th week of isotretinoin therapy. Results The serum catestatin levels in patients with acne vulgaris were statistically higher than in the control group (p < 0.001). In addition, serum catestatin levels were associated with the severity of acne vulgaris. A significant decrease in serum catestatin levels was detected after 24 weeks of systemic isotretinoin treatment. Conclusion Catestatin was found to be significantly higher in the serum of patients with acne vulgaris compared to the control group. Although, the demonstration that catestatin levels decrease with isotretinoin treatment may indicate that it may have an important role in the pathogenesis of acne and could be used as a biomarker, future studies are needed to establish the role of catestatin in the pathogenesis of acne vulgaris.
Collapse
Affiliation(s)
- Sera Nur Yücesoy
- Department of Dermatology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Tumay Ak
- Department of Internal Medicine, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Sena Öner
- Department of Biochemistry, Biruni University, Istanbul, Turkey
| | - Server Serdaroğlu
- Department of Dermatology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
6
|
Li S, Peng L, Chen L, Que L, Kang W, Hu X, Ma J, Di Z, Liu Y. Discovery of Highly Bioactive Peptides through Hierarchical Structural Information and Molecular Dynamics Simulations. J Chem Inf Model 2024; 64:8164-8175. [PMID: 39466714 DOI: 10.1021/acs.jcim.4c01006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Peptide drugs play an essential role in modern therapeutics, but the computational design of these molecules is hindered by several challenges. Traditional methods like molecular docking and molecular dynamics (MD) simulation, as well as recent deep learning approaches, often face limitations related to computational resource demands, complex binding affinity assessments, extensive data requirements, and poor model interpretability. Here, we introduce PepHiRe, an innovative methodology that utilizes the hierarchical structural information in peptide sequences and employs a novel strategy called Ladderpath, rooted in algorithmic information theory, to rapidly generate and enhance the efficiency and clarity of novel peptide design. We applied PepHiRe to develop BH3-like peptide inhibitors targeting myeloid cell leukemia-1, a protein associated with various cancers. By analyzing just eight known bioactive BH3 peptide sequences, PepHiRe effectively derived a hierarchy of subsequences used to create new BH3-like peptides. These peptides underwent screening through MD simulations, leading to the selection of five candidates for synthesis and subsequent in vitro testing. Experimental results demonstrated that these five peptides possess high inhibitory activity, with IC50 values ranging from 28.13 ± 7.93 to 167.42 ± 22.15 nM. Our study explores a white-box model driven technique and a structured screening pipeline for identifying and generating novel peptides with potential bioactivity.
Collapse
Affiliation(s)
- Shu Li
- Centre of Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR 999078, China
| | - Lu Peng
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Liuqing Chen
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Linjie Que
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
| | - Wenqingqing Kang
- Centre of Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR 999078, China
| | - Xiaojun Hu
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Jun Ma
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Zengru Di
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
| | - Yu Liu
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
| |
Collapse
|
7
|
Yadav N, Chauhan VS. Advancements in peptide-based antimicrobials: A possible option for emerging drug-resistant infections. Adv Colloid Interface Sci 2024; 333:103282. [PMID: 39276418 DOI: 10.1016/j.cis.2024.103282] [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: 02/03/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024]
Abstract
In recent years, multidrug-resistant pathogenic microorganisms (MDROs) have emerged as a severe threat to human health, exhibiting robust resistance to traditional antibiotics. This has created a formidable challenge in modern medicine as we grapple with limited options to combat these resilient bacteria. Despite extensive efforts by scientists to develop new antibiotics targeting these pathogens, the quest for novel antibacterial molecules has become increasingly arduous. Fortunately, nature offers a potential solution in the form of cationic antimicrobial peptides (AMPs) and their synthetic counterparts. AMPs, naturally occurring peptides, have displayed promising efficacy in fighting bacterial infections by disrupting bacterial cell membranes, hindering their survival and reproduction. These peptides, along with their synthetic mimics, present an exciting alternative in combating antibiotic resistance. They hold the potential to emerge as a formidable tool against MDROs, offering hope for improved strategies to protect communities. Extensive research has explored the diversity, history, and structure-properties relationship of AMPs, investigating their amphiphilic nature for membrane disruption and mechanisms of action. However, despite their therapeutic promise, AMPs face several documented limitations. Among these challenges, poor pharmacokinetic properties stand out, impeding the attainment of therapeutic levels in the body. Additionally, some AMPs exhibit toxicity and susceptibility to protease cleavage, leading to a short half-life and reduced efficacy in animal models. These limitations pose obstacles in developing effective treatments based on AMPs. Furthermore, the high manufacturing costs associated with AMPs could significantly hinder their widespread use. In this review, we aim to present experimental and theoretical insights into different AMPs, focusing specifically on antibacterial peptides (ABPs). Our goal is to offer a concise overview of peptide-based drug candidates, drawing from a wide array of literature and peer-reviewed studies. We also explore recent advancements in AMP development and discuss the challenges researchers face in moving these molecules towards clinical trials. Our main objective is to offer a comprehensive overview of current AMP and ABP research to guide the development of more precise and effective therapies for bacterial infections.
Collapse
Affiliation(s)
- Nitin Yadav
- Gandhi Institute of Technology and Management, Gandhi Nagar, Rushikonda, Visakhapatnam, Andhra Pradesh 530045, India; Molecular Medicine, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India; Biotide Solutions LLP, B-23, Geetanjali Enclave, Malviya Nagar, New Delhi 110017, India.
| | - Virander S Chauhan
- Gandhi Institute of Technology and Management, Gandhi Nagar, Rushikonda, Visakhapatnam, Andhra Pradesh 530045, India; Molecular Medicine, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India; Biotide Solutions LLP, B-23, Geetanjali Enclave, Malviya Nagar, New Delhi 110017, India.
| |
Collapse
|
8
|
Taheri-Araghi S. Synergistic action of antimicrobial peptides and antibiotics: current understanding and future directions. Front Microbiol 2024; 15:1390765. [PMID: 39144233 PMCID: PMC11322369 DOI: 10.3389/fmicb.2024.1390765] [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: 02/23/2024] [Accepted: 07/05/2024] [Indexed: 08/16/2024] Open
Abstract
Antibiotic resistance is a growing global problem that requires innovative therapeutic approaches and strategies for administering antibiotics. One promising approach is combination therapy, in which two or more drugs are combined to combat an infection. Along this line, the combination of antimicrobial peptides (AMPs) with conventional antibiotics has gained attention mainly due to the complementary mechanisms of action of AMPs and conventional antibiotics. In this article, we review both in vitro and in vivo studies that explore the synergy between AMPs and antibiotics. We highlight several mechanisms through which synergy is observed in in vitro experiments, including increasing membrane permeability, disrupting biofilms, directly potentiating antibiotic efficacy, and inhibiting resistance development. Moreover, in vivo studies reveal additional mechanisms such as enhanced/modulated immune responses, reduced inflammation, and improved tissue regeneration. Together, the current literature demonstrates that AMP-antibiotic combinations can substantially enhance efficacy of antibiotic therapies, including therapies against resistant bacteria, which represents a valuable enhancement to current antimicrobial strategies.
Collapse
Affiliation(s)
- Sattar Taheri-Araghi
- Department of Physics and Astronomy, California State University, Northridge, CA, United States
| |
Collapse
|
9
|
Kong X, Vishwanath V, Neelakantan P, Ye Z. Harnessing antimicrobial peptides in endodontics. Int Endod J 2024; 57:815-840. [PMID: 38441321 DOI: 10.1111/iej.14043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 06/13/2024]
Abstract
Endodontic therapy includes various procedures such as vital pulp therapy, root canal treatment and retreatment, surgical endodontic treatment and regenerative endodontic procedures. Disinfection and tissue repair are crucial for the success of these therapies, necessitating the development of therapeutics that can effectively target microbiota, eliminate biofilms, modulate inflammation and promote tissue repair. However, no current endodontic agents can achieve these goals. Antimicrobial peptides (AMPs), which are sequences of amino acids, have gained attention due to their unique advantages, including reduced susceptibility to drug resistance, broad-spectrum antibacterial properties and the ability to modulate the immune response of the organism effectively. This review systematically discusses the structure, mechanisms of action, novel designs and limitations of AMPs. Additionally, it highlights the efforts made by researchers to overcome peptide shortcomings and emphasizes the potential applications of AMPs in endodontic treatments.
Collapse
Affiliation(s)
- Xinzi Kong
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Vijetha Vishwanath
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Prasanna Neelakantan
- Department of Endodontics, University of the Pacific Arthur A. Dugoni School of Dentistry, San Francisco, California, USA
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R., China
| |
Collapse
|
10
|
Marissen J, Reichert L, Härtel C, Fortmann MI, Faust K, Msanga D, Harder J, Zemlin M, Gomez de Agüero M, Masjosthusmann K, Humberg A. Antimicrobial Peptides (AMPs) and the Microbiome in Preterm Infants: Consequences and Opportunities for Future Therapeutics. Int J Mol Sci 2024; 25:6684. [PMID: 38928389 PMCID: PMC11203687 DOI: 10.3390/ijms25126684] [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: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial peptides (AMPs) are crucial components of the innate immune system in various organisms, including humans. Beyond their direct antimicrobial effects, AMPs play essential roles in various physiological processes. They induce angiogenesis, promote wound healing, modulate immune responses, and serve as chemoattractants for immune cells. AMPs regulate the microbiome and combat microbial infections on the skin, lungs, and gastrointestinal tract. Produced in response to microbial signals, AMPs help maintain a balanced microbial community and provide a first line of defense against infection. In preterm infants, alterations in microbiome composition have been linked to various health outcomes, including sepsis, necrotizing enterocolitis, atopic dermatitis, and respiratory infections. Dysbiosis, or an imbalance in the microbiome, can alter AMP profiles and potentially lead to inflammation-mediated diseases such as chronic lung disease and obesity. In the following review, we summarize what is known about the vital role of AMPs as multifunctional peptides in protecting newborn infants against infections and modulating the microbiome and immune response. Understanding their roles in preterm infants and high-risk populations offers the potential for innovative approaches to disease prevention and treatment.
Collapse
Affiliation(s)
- Janina Marissen
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Lilith Reichert
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Mats Ingmar Fortmann
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Kirstin Faust
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Delfina Msanga
- Department of Pediatrics, Bugando Hospital, Catholic University of Health and Allied Sciences, Mwanza 33109, Tanzania;
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, 24105 Kiel, Germany;
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| | - Alexander Humberg
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| |
Collapse
|
11
|
Ostan NKH, Cole GB, Wang FZ, Reichheld SE, Moore G, Pan C, Yu R, Lai CCL, Sharpe S, Lee HO, Schryvers AB, Moraes TF. A secreted bacterial protein protects bacteria from cationic antimicrobial peptides by entrapment in phase-separated droplets. PNAS NEXUS 2024; 3:pgae139. [PMID: 38633880 PMCID: PMC11022072 DOI: 10.1093/pnasnexus/pgae139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/26/2024] [Indexed: 04/19/2024]
Abstract
Mammalian hosts combat bacterial infections through the production of defensive cationic antimicrobial peptides (CAPs). These immune factors are capable of directly killing bacterial invaders; however, many pathogens have evolved resistance evasion mechanisms such as cell surface modification, CAP sequestration, degradation, or efflux. We have discovered that several pathogenic and commensal proteobacteria, including the urgent human threat Neisseria gonorrhoeae, secrete a protein (lactoferrin-binding protein B, LbpB) that contains a low-complexity anionic domain capable of inhibiting the antimicrobial activity of host CAPs. This study focuses on a cattle pathogen, Moraxella bovis, that expresses the largest anionic domain of the LbpB homologs. We used an exhaustive biophysical approach employing circular dichroism, biolayer interferometry, cross-linking mass spectrometry, microscopy, size-exclusion chromatography with multi-angle light scattering coupled to small-angle X-ray scattering (SEC-MALS-SAXS), and NMR to understand the mechanisms of LbpB-mediated protection against CAPs. We found that the anionic domain of this LbpB displays an α-helical secondary structure but lacks a rigid tertiary fold. The addition of antimicrobial peptides derived from lactoferrin (i.e. lactoferricin) to the anionic domain of LbpB or full-length LbpB results in the formation of phase-separated droplets of LbpB together with the antimicrobial peptides. The droplets displayed a low rate of diffusion, suggesting that CAPs become trapped inside and are no longer able to kill bacteria. Our data suggest that pathogens, like M. bovis, leverage anionic intrinsically disordered domains for the broad recognition and neutralization of antimicrobials via the formation of biomolecular condensates.
Collapse
Affiliation(s)
- Nicholas K H Ostan
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gregory B Cole
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Flora Zhiqi Wang
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sean E Reichheld
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Gaelen Moore
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chuxi Pan
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ronghua Yu
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - Simon Sharpe
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Hyun O Lee
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anthony B Schryvers
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Trevor F Moraes
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
12
|
Blumstein DT, Johnson NA, Katz ND, Kharpatin S, Ortiz‐Ross X, Parra E, Reshke A. Biological lessons for strategic resistance management. Evol Appl 2023; 16:1861-1871. [PMID: 38143901 PMCID: PMC10739074 DOI: 10.1111/eva.13616] [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: 04/04/2023] [Accepted: 10/28/2023] [Indexed: 12/26/2023] Open
Abstract
Biological resistance to pesticides, vaccines, antibiotics, and chemotherapies creates huge costs to society, including extensive morbidity and mortality. We simultaneously face costly resistance to social changes, such as those required to resolve human-wildlife conflicts and conserve biodiversity and the biosphere. Viewing resistance as a force that impedes change from one state to another, we suggest that an analysis of biological resistance can provide unique and potentially testable insights into understanding resistance to social changes. We review key insights from managing biological resistance and develop a framework that identifies seven strategies to overcome resistance. We apply this framework to consider how it might be used to understand social resistance and generate potentially novel hypotheses that may be useful to both enhance the development of strategies to manage resistance and modulate change in socio-ecological systems.
Collapse
Affiliation(s)
- Daniel T. Blumstein
- Department of Ecology and Evolutionary BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Norman A. Johnson
- Department of BiologyUniversity of MassachusettsAmherstMassachusettsUSA
| | - Nurit D. Katz
- Department of Ecology and Evolutionary BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Samuel Kharpatin
- Department of Ecology and Evolutionary BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Xochitl Ortiz‐Ross
- Department of Ecology and Evolutionary BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Eliseo Parra
- Department of Ecology and Evolutionary BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Amanda Reshke
- Department of Ecology and Evolutionary BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
13
|
Lee S, Schefter BR, Taheri-Araghi S, Ha BY. Modeling selectivity of antimicrobial peptides: how it depends on the presence of host cells and cell density. RSC Adv 2023; 13:34167-34182. [PMID: 38020026 PMCID: PMC10663724 DOI: 10.1039/d3ra06030f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
Antimicrobial peptides (AMPs), naturally-occurring peptide antibiotics, are known to attack bacteria selectively over the host cells. The emergence of drug-resistant bacteria has spurred much effort in utilizing optimized (more selective) AMPs as new peptide antibiotics. Cell selectivity of these peptides depends on various factors or parameters such as their binding affinity for cell membranes, peptide trapping in cells, peptide coverages on cell membranes required for membrane rupture, and cell densities. In this work, using a biophysical model of peptide selectivity, we show this dependence quantitatively especially for a mixture of bacteria and host cells. The model suggests a rather nontrivial dependence of the selectivity on the presence of host cells, cell density, and peptide trapping. In a typical biological setting, peptide trapping works in favor of host cells; the selectivity increases with increasing host-cell density but decreases with bacterial cell density. Because of the cell-density dependence of peptide activity, the selectivity can be overestimated by two or three orders of magnitude. The model also clarifies how the cell selectivity of AMPs differs from their membrane selectivity.
Collapse
Affiliation(s)
- Suemin Lee
- Department of Physics and Astronomy, University of Waterloo Waterloo Ontario N2L 3G1 Canada
| | - Bethany R Schefter
- Department of Physics and Astronomy, University of Western Ontario London Ontario N6A 3K7 Canada
| | - Sattar Taheri-Araghi
- Department of Physics and Astronomy, California State University Northridge CA 91330 USA
| | - Bae-Yeun Ha
- Department of Physics and Astronomy, University of Waterloo Waterloo Ontario N2L 3G1 Canada
| |
Collapse
|
14
|
Lau JZ, Kuo SH, Belo Y, Malach E, Maron B, Caraway HE, Oh MW, Zhang Y, Ismail N, Lau GW, Hayouka Z. Antibacterial efficacy of an ultra-short palmitoylated random peptide mixture in mouse models of infection by carbapenem-resistant Klebsiella pneumoniae. Antimicrob Agents Chemother 2023; 67:e0057423. [PMID: 37819119 PMCID: PMC10648864 DOI: 10.1128/aac.00574-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/18/2023] [Indexed: 10/13/2023] Open
Abstract
Indiscriminate use of antibiotics has imposed a selective pressure for the rapid rise in bacterial resistance, creating an urgent need for novel therapeutics for managing bacterial infectious diseases while counteracting bacterial resistance. Carbapenem-resistant Klebsiella pneumoniae strains have become a major challenge in modern medicine due to their ability to cause an array of severe infections. Recently, we have shown that the 20-mer random peptide mixtures are effective therapeutics against three ESKAPEE pathogens. Here, we evaluated the toxicity, biodistribution, bioavailability, and efficacy of the ultra-short palmitoylated 5-mer phenylalanine:lysine (FK5P) random peptide mixtures against multiple clinical isolates of carbapenem-resistant K. pneumoniae and K. oxytoca. We demonstrate the FK5P rapidly and effectively killed various strains of K. pneumoniae, inhibited the formation of biofilms, and disrupted mature biofilms. FK5P displayed strong toxicity profiles both in vitro and in mice, with prolonged favorable biodistribution and a long half-life. Significantly, FK5P reduced the bacterial burden in mouse models of acute pneumonia and bacteremia and increased the survival rate in a mouse model of bacteremia. Our results demonstrate that FK5P is a safe and promising therapy against Klebsiella species as well as other ESKAPEE pathogens.
Collapse
Affiliation(s)
- Jonathan Z. Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Shanny Hsuan Kuo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Yael Belo
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Einav Malach
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Bar Maron
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Hannah E. Caraway
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Myung Whan Oh
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Yi Zhang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Nahed Ismail
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Gee W. Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
15
|
Yang S, Duncan GA. Synthetic mucus biomaterials for antimicrobial peptide delivery. J Biomed Mater Res A 2023; 111:1616-1626. [PMID: 37199137 PMCID: PMC10524183 DOI: 10.1002/jbm.a.37559] [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: 03/07/2023] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023]
Abstract
Despite the promise of antimicrobial peptides (AMPs) as treatments for antibiotic-resistant infections, their therapeutic efficacy is limited due to the rapid degradation and low bioavailability of AMPs. To address this, we have developed and characterized a synthetic mucus (SM) biomaterial capable of delivering LL37 AMPs and enhancing their therapeutic effect. LL37 is an AMP that exhibits a wide range of antimicrobial activity against bacteria, including Pseudomonas aeruginosa. LL37 loaded SM hydrogels demonstrated controlled release with 70%-95% of loaded LL37 over 8 h due to charge-mediated interactions between mucins and LL37 AMPs. Compared to treatment with LL37 alone where antimicrobial activity was reduced after 3 h, LL37-SM hydrogels inhibited P. aeruginosa (PAO1) growth over 12 h. LL37-SM hydrogel treatment reduced PAO1 viability over 6 h whereas a rebound in bacterial growth was observed when treated with LL37 only. These data demonstrate LL37-SM hydrogels enhance antimicrobial activity by preserving LL37 AMP activity and bioavailability. Overall, this work establishes SM biomaterials as a platform for enhanced AMP delivery for antimicrobial applications.
Collapse
Affiliation(s)
- Sydney Yang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Gregg A Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
16
|
Tang R, Tan H, Dai Y, Li L, Huang Y, Yao H, Cai Y, Yu G. Application of antimicrobial peptides in plant protection: making use of the overlooked merits. FRONTIERS IN PLANT SCIENCE 2023; 14:1139539. [PMID: 37538059 PMCID: PMC10394246 DOI: 10.3389/fpls.2023.1139539] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/07/2023] [Indexed: 08/05/2023]
Abstract
Pathogen infection is one of the major causes of yield loss in the crop field. The rapid increase of antimicrobial resistance in plant pathogens has urged researchers to develop both new pesticides and management strategies for plant protection. The antimicrobial peptides (AMPs) showed potential on eliminating plant pathogenic fungi and bacteria. Here, we first summarize several overlooked advantages and merits of AMPs, which includes the steep dose-response relations, fast killing ability, broad synergism, slow resistance selection. We then discuss the possible application of AMPs for plant protection with above merits, and highlight how AMPs can be incorporated into a more efficient integrated management system that both increases the crop yield and reduce resistance evolution of pathogens.
Collapse
|
17
|
Witzany C, Rolff J, Regoes RR, Igler C. The pharmacokinetic-pharmacodynamic modelling framework as a tool to predict drug resistance evolution. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001368. [PMID: 37522891 PMCID: PMC10433423 DOI: 10.1099/mic.0.001368] [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: 03/20/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023]
Abstract
Pharmacokinetic-pharmacodynamic (PKPD) models, which describe how drug concentrations change over time and how that affects pathogen growth, have proven highly valuable in designing optimal drug treatments aimed at bacterial eradication. However, the fast rise of antimicrobial resistance calls for increased focus on an additional treatment optimization criterion: avoidance of resistance evolution. We demonstrate here how coupling PKPD and population genetics models can be used to determine treatment regimens that minimize the potential for antimicrobial resistance evolution. Importantly, the resulting modelling framework enables the assessment of resistance evolution in response to dynamic selection pressures, including changes in antimicrobial concentration and the emergence of adaptive phenotypes. Using antibiotics and antimicrobial peptides as an example, we discuss the empirical evidence and intuition behind individual model parameters. We further suggest several extensions of this framework that allow a more comprehensive and realistic prediction of bacterial escape from antimicrobials through various phenotypic and genetic mechanisms.
Collapse
Affiliation(s)
| | - Jens Rolff
- Evolutionary Biology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| | - Roland R. Regoes
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Claudia Igler
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
- School of Biological Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
18
|
Dalla Torre C, Sannio F, Battistella M, Docquier JD, De Zotti M. Peptaibol Analogs Show Potent Antibacterial Activity against Multidrug Resistant Opportunistic Pathogens. Int J Mol Sci 2023; 24:ijms24097997. [PMID: 37175704 PMCID: PMC10178204 DOI: 10.3390/ijms24097997] [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: 04/05/2023] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
New classes of antibacterial drugs are urgently needed to address the global issue of antibiotic resistance. In this context, peptaibols are promising membrane-active peptides since they are not involved in innate immunity and their antimicrobial activity does not involve specific cellular targets, therefore reducing the chance of bacterial resistance development. Trichogin GA IV is a nonhemolytic, natural, short-length peptaibol active against Gram-positive bacteria and resistant to proteolysis. In this work, we report on the antibacterial activity of cationic trichogin analogs. Several peptides appear non-hemolytic and strongly active against many clinically relevant bacterial species, including antibiotic-resistant clinical isolates, such as Staphylococcus aureus, Acinetobacter baumannii, and extensively drug-resistant Pseudomonas aeruginosa, against which there are only a limited number of antibiotics under development. Our results further highlight how the modification of natural peptides is a valuable strategy for obtaining improved antibacterial agents with potential therapeutic applications.
Collapse
Affiliation(s)
- Chiara Dalla Torre
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, I-35131 Padova, Italy
| | - Filomena Sannio
- Dipartimento di Biotecnologie Mediche, University of Siena, Viale Bracci 16, I-53100 Siena, Italy
| | - Mattia Battistella
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, I-35131 Padova, Italy
| | - Jean-Denis Docquier
- Dipartimento di Biotecnologie Mediche, University of Siena, Viale Bracci 16, I-53100 Siena, Italy
- Lead Discovery Siena s.r.l., Via Fiorentina 1, I-53100 Siena, Italy
- Laboratoire de Bactériologie Moléculaire, Centre d'Ingénierie des Protéines-UR InBioS, University of Liège, Allée du Six Août 11, B-4000 Liège, Belgium
| | - Marta De Zotti
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, I-35131 Padova, Italy
| |
Collapse
|
19
|
Jangir PK, Ogunlana L, Szili P, Czikkely M, Shaw LP, Stevens EJ, Yu Y, Yang Q, Wang Y, Pál C, Walsh TR, MacLean CR. The evolution of colistin resistance increases bacterial resistance to host antimicrobial peptides and virulence. eLife 2023; 12:e84395. [PMID: 37094804 PMCID: PMC10129329 DOI: 10.7554/elife.84395] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 04/05/2023] [Indexed: 04/26/2023] Open
Abstract
Antimicrobial peptides (AMPs) offer a promising solution to the antibiotic resistance crisis. However, an unresolved serious concern is that the evolution of resistance to therapeutic AMPs may generate cross-resistance to host AMPs, compromising a cornerstone of the innate immune response. We systematically tested this hypothesis using globally disseminated mobile colistin resistance (MCR) that has been selected by the use of colistin in agriculture and medicine. Here, we show that MCR provides a selective advantage to Escherichia coli in the presence of key AMPs from humans and agricultural animals by increasing AMP resistance. Moreover, MCR promotes bacterial growth in human serum and increases virulence in a Galleria mellonella infection model. Our study shows how the anthropogenic use of AMPs can drive the accidental evolution of resistance to the innate immune system of humans and animals. These findings have major implications for the design and use of therapeutic AMPs and suggest that MCR may be difficult to eradicate, even if colistin use is withdrawn.
Collapse
Affiliation(s)
- Pramod K Jangir
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Lois Ogunlana
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Petra Szili
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research NetworkSzegedHungary
- Doctoral School of Multidisciplinary Medical Sciences, University of SzegedSzegedHungary
| | - Marton Czikkely
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research NetworkSzegedHungary
| | - Liam P Shaw
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Emily J Stevens
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Yang Yu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural UniversityGuangzhouChina
| | - Qiue Yang
- Fujian Provincial Key Laboratory of Soil Environmental Health and RegulaWon, College of Resources and Environment, Fujian Agriculture and Forestry UniversityFuzhouChina
| | - Yang Wang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural UniversityBeijingChina
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research NetworkSzegedHungary
| | - Timothy R Walsh
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Craig R MacLean
- Department of Biology, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
20
|
Yang S, Duncan G. Synthetic Mucus Biomaterials for Antimicrobial Peptide Delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531025. [PMID: 36945438 PMCID: PMC10028879 DOI: 10.1101/2023.03.07.531025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Despite the promise of antimicrobial peptides (AMPs) as treatments for antibiotic-resistant infections, their therapeutic efficacy is limited due to the rapid degradation and low bioavailability of AMPs. To address this, we have developed and characterized a synthetic mucus (SM) biomaterial capable of delivering AMPs and enhancing their therapeutic effect. LL37 loaded SM hydrogels demonstrated controlled release of LL37 over 8 hours as a result of charge-mediated interactions between mucins and LL37 AMPs. Compared to treatment with LL37 alone where antimicrobial activity was reduced after 3 hours, LL37-SM hydrogels inhibited Pseudomonas aeruginosa PAO1 growth over 12 hours. LL37-SM hydrogel treatment reduced PAO1 viability over 6 hours whereas a rebound in bacterial growth was observed when treated with LL37 only. These data demonstrate LL37-SM hydrogels enhance antimicrobial activity by preserving LL37 AMP activity and bioavailability. Overall, this work establishes SM biomaterials as a platform for enhanced AMP delivery for antimicrobial applications.
Collapse
Affiliation(s)
- Sydney Yang
- University of Maryland, Fischell Department of Bioengineering, College Park, MD
| | - Gregg Duncan
- University of Maryland, Fischell Department of Bioengineering, College Park, MD
| |
Collapse
|
21
|
Kang DD, Park J, Park Y. Therapeutic Potential of Antimicrobial Peptide PN5 against Multidrug-Resistant E. coli and Anti-Inflammatory Activity in a Septic Mouse Model. Microbiol Spectr 2022; 10:e0149422. [PMID: 36129300 PMCID: PMC9603901 DOI: 10.1128/spectrum.01494-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/01/2022] [Indexed: 12/31/2022] Open
Abstract
Antibiotic-resistant bacteria have become a public health problem. Thus, antimicrobial peptides (AMPs) have been evaluated as substitutes for antibiotics. Herein, we investigated PN5 derived from Pinus densiflora (pine needle). PN5 exhibited antimicrobial activity without causing cytotoxic effects. Based on these results, we examined the mode of action of PN5 against Gram-negative and -positive bacteria. PN5 exhibited membrane permeabilization ability, had antimicrobial stability in the presence of elastase, a proteolytic enzyme, and did not induce resistance in bacteria. Bacterial lipopolysaccharide (LPS) induces an inflammatory response in RAW 264.7 macrophages. PN5 suppressed proinflammatory cytokines mediated by NF-κB and mitogen-activated protein kinase signaling. In C57BL/6J mice treated with LPS and d-galactosamine, PN5 exhibited anti-inflammatory activity in inflamed mouse livers. Our results indicate that PN5 has antimicrobial and anti-inflammatory activities and thus may be useful as an antimicrobial agent to treat septic shock caused by multidrug-resistant (MDR) Escherichia coli without causing further resistance. IMPORTANCE Antibiotic-resistant bacteria are a global health concern. There is no effective treatment for antibiotic-resistant bacteria, and new alternatives are being suggested. The present study found antibacterial and anti-inflammatory activities of PN5 derived from Pinus densiflora (pine needle), and further investigated the therapeutic effect in a mouse septic model. As a mechanism of antibacterial activity, PN5 exhibited the membrane permeabilization ability of the toroidal model, and treated strains did not develop drug resistance during serial passages. PN5 showed immunomodulatory properties of neutralizing LPS in a mouse septic model. These results indicate that PN5 could be a new and promising therapeutic agent for bacterial infectious disease caused by antibiotic-resistant strains.
Collapse
Affiliation(s)
- Da Dam Kang
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Jonggwan Park
- Department of Bioinformatics, Kongju National University, Kongju, South Korea
| | - Yoonkyung Park
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
- Research Center for Proteinaceous Materials (RCPM), Chosun University, Gwangju, South Korea
| |
Collapse
|
22
|
Maron B, Rolff J, Friedman J, Hayouka Z. Antimicrobial Peptide Combination Can Hinder Resistance Evolution. Microbiol Spectr 2022; 10:e0097322. [PMID: 35862981 PMCID: PMC9430149 DOI: 10.1128/spectrum.00973-22] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/24/2022] [Indexed: 12/29/2022] Open
Abstract
Antibiotic-resistant microbial pathogens are becoming a major threat to human health. Therefore, there is an urgent need to develop new alternatives to conventional antibiotics. One such promising alternative is antimicrobial peptides (AMPs), which are produced by virtually all organisms and typically inhibit bacteria via membrane disruption. However, previous studies demonstrated that bacteria can rapidly develop AMP resistance. Here, we study whether combination therapy, known to be able to inhibit the evolution of resistance to conventional antibiotics, can also hinder the evolution of AMP resistance. To do so, we evolved the opportunistic pathogen Staphylococcus aureus in the presence of individual AMPs, AMP pairs, and a combinatorial antimicrobial peptide library. Treatment with some AMP pairs indeed hindered the evolution of resistance compared with individual AMPs. In particular, resistance to pairs was delayed when resistance to the individual AMPs came at a cost of impaired bacterial growth and did not confer cross-resistance to other tested AMPs. The lowest level of resistance evolved during treatment with the combinatorial antimicrobial peptide library termed random antimicrobial peptide mixture, which contains more than a million different peptides. A better understanding of how AMP combinations affect the evolution of resistance is a crucial step in order to design "resistant proof" AMP cocktails that will offer a sustainable treatment option for antibiotic-resistant pathogens. IMPORTANCE The main insights gleaned from this study are the following. (i) AMP combination treatment can delay the evolution of resistance in S. aureus. Treatment with some AMP pairs resulted in significantly lower resistance then treatment with either of the individual AMPs. Treatment with a random AMP library resulted in no detectable resistance. (ii) The rate at which resistance to combination arises correlates with the cost of resistance to individual AMPs and their cross-resistance. In particular, combinations to which the least resistance arose involved AMPs with high fitness cost of resistance and low cross-resistance. (iii) No broad-range AMP resistance evolved. Strains that evolved resistance to some AMPs typically remained sensitive to other AMPs, alleviating concerns regarding the evolution of resistance to immune system AMPs in response to AMP treatment.
Collapse
Affiliation(s)
- Bar Maron
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot, Israel
- Department of Plant Pathology and Microbiology, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Jens Rolff
- Institute of Biology, Evolutionary Biology, Freie University, Berlin, Germany
| | - Jonathan Friedman
- Department of Plant Pathology and Microbiology, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
23
|
The EnvZ/OmpR Two-Component System Regulates the Antimicrobial Activity of TAT-RasGAP 317-326 and the Collateral Sensitivity to Other Antibacterial Agents. Microbiol Spectr 2022; 10:e0200921. [PMID: 35579440 PMCID: PMC9241736 DOI: 10.1128/spectrum.02009-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The rapid emergence of antibiotic-resistant bacteria poses a serious threat to public health worldwide. Antimicrobial peptides (AMPs) are promising antibiotic alternatives; however, little is known about bacterial mechanisms of AMP resistance and the interplay between AMP resistance and the bacterial response to other antimicrobials. In this study, we identified Escherichia coli mutants resistant to the TAT-RasGAP317-326 antimicrobial peptide and found that resistant bacteria show collateral sensitivity to other AMPs and antibacterial agents. We determined that resistance to TAT-RasGAP317-326 peptide arises through mutations in the histidine kinase EnvZ, a member of the EnvZ/OmpR two-component system responsible for osmoregulation in E. coli. In particular, we found that TAT-RasGAP317-326 binding and entry is compromised in E. coli peptide-resistant mutants. We showed that peptide resistance is associated with transcriptional regulation of a number of pathways and EnvZ-mediated resistance is dependent on the OmpR response regulator but is independent of the OmpC and OmpF outer membrane porins. Our findings provide insight into the bacterial mechanisms of TAT-RasGAP317-326 resistance and demonstrate that resistance to this AMP is associated with collateral sensitivity to other antibacterial agents. IMPORTANCE Antimicrobial peptides (AMP) are promising alternatives to classical antibiotics in the fight against antibiotic resistance. Resistance toward antimicrobial peptides can occur, but little is known about the mechanisms driving this phenomenon. Moreover, there is limited knowledge on how AMP resistance relates to the bacterial response to other antimicrobial agents. Here, we address these questions in the context of the antimicrobial peptide TAT-RasGAP317-326. We show that resistant Escherichia coli strains can be selected and do not show resistance to other antimicrobial agents. Resistance is caused by a mutation in a regulatory pathway, which lowers binding and entry of the peptide in E. coli. Our results highlight a mechanism of resistance that is specific to TAT-RasGAP317-326. Further research is required to characterize these mechanisms and to evaluate the potential of antimicrobial combinations to curb the development of antimicrobial resistance.
Collapse
|
24
|
Adaptation to simulated microgravity in Streptococcus mutans. NPJ Microgravity 2022; 8:17. [PMID: 35654802 PMCID: PMC9163064 DOI: 10.1038/s41526-022-00205-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 05/13/2022] [Indexed: 11/08/2022] Open
Abstract
Long-term space missions have shown an increased incidence of oral disease in astronauts’ and as a result, are one of the top conditions predicted to impact future missions. Here we set out to evaluate the adaptive response of Streptococcus mutans (etiological agent of dental caries) to simulated microgravity. This organism has been well studied on earth and treatment strategies are more predictable. Despite this, we are unsure how the bacterium will respond to the environmental stressors in space. We used experimental evolution for 100-days in high aspect ratio vessels followed by whole genome resequencing to evaluate this adaptive response. Our data shows that planktonic S. mutans did evolve variants in three genes (pknB, SMU_399 and SMU_1307c) that can be uniquely attributed to simulated microgravity populations. In addition, collection of data at multiple time points showed mutations in three additional genes (SMU_399, ptsH and rex) that were detected earlier in simulated microgravity populations than in the normal gravity controls, many of which are consistent with other studies. Comparison of virulence-related phenotypes between biological replicates from simulated microgravity and control orientation cultures generally showed few changes in antibiotic susceptibility, while acid tolerance and adhesion varied significantly between biological replicates and decreased as compared to the ancestral populations. Most importantly, our data shows the importance of a parallel normal gravity control, sequencing at multiple time points and the use of biological replicates for appropriate analysis of adaptation in simulated microgravity.
Collapse
|
25
|
Ali W, Elsahn A, Ting DSJ, Dua HS, Mohammed I. Host Defence Peptides: A Potent Alternative to Combat Antimicrobial Resistance in the Era of the COVID-19 Pandemic. Antibiotics (Basel) 2022; 11:475. [PMID: 35453226 PMCID: PMC9032040 DOI: 10.3390/antibiotics11040475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 12/07/2022] Open
Abstract
One of the greatest challenges facing the medical community today is the ever-increasing trajectory of antimicrobial resistance (AMR), which is being compounded by the decrease in our antimicrobial armamentarium. From their initial discovery to the current day, antibiotics have seen an exponential increase in their usage, from medical to agricultural use. Benefits aside, this has led to an exponential increase in AMR, with the fear that over 10 million lives are predicted to be lost by 2050, according to the World Health Organisation (WHO). As such, medical researchers are turning their focus to discovering novel alternatives to antimicrobials, one being Host Defence Peptides (HDPs). These small cationic peptides have shown great efficacy in being used as an antimicrobial therapy for currently resistant microbial variants. With the sudden emergence of the SARS-CoV-2 variant and the subsequent global pandemic, the great versatility and potential use of HDPs as an alternative to conventional antibiotics in treating as well as preventing the spread of COVID-19 has been reviewed. Thus, to allow the reader to have a full understanding of the multifaceted therapeutic use of HDPs, this literature review shall cover the association between COVID-19 and AMR whilst discussing and evaluating the use of HDPs as an answer to antimicrobial resistance (AMR).
Collapse
Affiliation(s)
| | | | | | | | - Imran Mohammed
- Section of Ophthalmology, Larry A. Donoso Laboratory for Eye Research, Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, University of Nottingham, Queens Medical Centre, Eye and ENT Building, Nottingham NG7 2UH, UK; (W.A.); (A.E.); (D.S.J.T.); (H.S.D.)
| |
Collapse
|
26
|
Recombination resolves the cost of horizontal gene transfer in experimental populations of Helicobacter pylori. Proc Natl Acad Sci U S A 2022; 119:e2119010119. [PMID: 35298339 PMCID: PMC8944584 DOI: 10.1073/pnas.2119010119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Horizontal gene transfer (HGT)—the transfer of DNA between lineages—is responsible for a large proportion of the genetic variation that contributes to evolution in microbial populations. While HGT can bring beneficial genetic innovation, the transfer of DNA from other species or strains can also have deleterious effects. In this study, we evolve populations of the bacteria Helicobacter pylori and use DNA sequencing to identify over 40,000 genetic variants transferred by HGT. We measure the cost of many of these and find that both strongly beneficial mutations and deleterious mutations are genetic variants transferred by natural transformation. Importantly, we also show how recombination that separates linked beneficial and deleterious mutations resolves the cost of HGT. Horizontal gene transfer (HGT) is important for microbial evolution, yet we know little about the fitness effects and dynamics of horizontally transferred genetic variants. In this study, we evolve laboratory populations of Helicobacter pylori, which take up DNA from their environment by natural transformation, and measure the fitness effects of thousands of transferred genetic variants. We find that natural transformation increases the rate of adaptation but comes at the cost of significant genetic load. We show that this cost is circumvented by recombination, which increases the efficiency of selection by decoupling deleterious and beneficial genetic variants. Our results show that adaptation with HGT, pervasive in natural microbial populations, is shaped by a combination of selection, recombination, and genetic drift not accounted for in existing models of evolution.
Collapse
|
27
|
Nuti N, Rottmann P, Stucki A, Koch P, Panke S, Dittrich PS. A Multiplexed Cell-Free Assay to Screen for Antimicrobial Peptides in Double Emulsion Droplets. Angew Chem Int Ed Engl 2022; 61:e202114632. [PMID: 34989471 PMCID: PMC9303939 DOI: 10.1002/anie.202114632] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Indexed: 12/17/2022]
Abstract
The global surge in bacterial resistance against traditional antibiotics triggered intensive research for novel compounds, with antimicrobial peptides (AMPs) identified as a promising candidate. Automated methods to systematically generate and screen AMPs according to their membrane preference, however, are still lacking. We introduce a novel microfluidic system for the simultaneous cell-free production and screening of AMPs for their membrane specificity. On our device, AMPs are cell-free produced within water-in-oil-in-water double emulsion droplets, generated at high frequency. Within each droplet, the peptides can interact with different classes of co-encapsulated liposomes, generating a membrane-specific fluorescent signal. The double emulsions can be incubated and observed in a hydrodynamic trapping array or analyzed via flow cytometry. Our approach provides a valuable tool for the discovery and development of membrane-active antimicrobials.
Collapse
Affiliation(s)
- Nicola Nuti
- Department of Biosystems Science and EngineeringBioanalytics GroupETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Philipp Rottmann
- Department of Biosystems Science and EngineeringBioprocess LaboratoryETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Ariane Stucki
- Department of Biosystems Science and EngineeringBioanalytics GroupETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Philipp Koch
- Department of Biosystems Science and EngineeringBioprocess LaboratoryETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Sven Panke
- Department of Biosystems Science and EngineeringBioprocess LaboratoryETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Petra S. Dittrich
- Department of Biosystems Science and EngineeringBioanalytics GroupETH ZürichMattenstrasse 264058BaselSwitzerland
| |
Collapse
|
28
|
Caraway HE, Lau JZ, Maron B, Oh MW, Belo Y, Brill A, Malach E, Ismail N, Hayouka Z, Lau GW. Antimicrobial Random Peptide Mixtures Eradicate Acinetobacter baumannii Biofilms and Inhibit Mouse Models of Infection. Antibiotics (Basel) 2022; 11:antibiotics11030413. [PMID: 35326876 PMCID: PMC8944503 DOI: 10.3390/antibiotics11030413] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 12/11/2022] Open
Abstract
Antibiotic resistance is one of the greatest crises in human medicine. Increased incidents of antibiotic resistance are linked to clinical overuse and overreliance on antibiotics. Among the ESKAPE pathogens, Acinetobacter baumannii, especially carbapenem-resistant isolates, has emerged as a significant threat in the context of blood, urinary tract, lung, and wound infections. Therefore, new approaches that limit the emergence of antibiotic resistant A. baumannii are urgently needed. Recently, we have shown that random peptide mixtures (RPMs) are an attractive alternative class of drugs to antibiotics with strong safety and pharmacokinetic profiles. RPMs are antimicrobial peptide mixtures produced by incorporating two amino acids at each coupling step, rendering them extremely diverse but still defined in their overall composition, chain length, and stereochemistry. The extreme diversity of RPMs may prevent bacteria from evolving resistance rapidly. Here, we demonstrated that RPMs rapidly and efficiently kill different strains of A. baumannii, inhibit biofilm formation, and disrupt mature biofilms. Importantly, RPMs attenuated bacterial burden in mouse models of acute pneumonia and soft tissue infection and significantly reduced mouse mortality during sepsis. Collectively, our results demonstrate RPMs have the potential to be used as powerful therapeutics against antibiotic-resistant A. baumannii.
Collapse
Affiliation(s)
- Hannah E. Caraway
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA; (H.E.C.); (J.Z.L.); (M.W.O.)
| | - Jonathan Z. Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA; (H.E.C.); (J.Z.L.); (M.W.O.)
| | - Bar Maron
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel; (B.M.); (Y.B.); (A.B.); (E.M.)
| | - Myung Whan Oh
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA; (H.E.C.); (J.Z.L.); (M.W.O.)
| | - Yael Belo
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel; (B.M.); (Y.B.); (A.B.); (E.M.)
| | - Aya Brill
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel; (B.M.); (Y.B.); (A.B.); (E.M.)
| | - Einav Malach
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel; (B.M.); (Y.B.); (A.B.); (E.M.)
| | - Nahed Ismail
- Department of Pathology, College of Medicine, University of Illinois at Chicago, 840 South Wood Street, Chicago, IL 60612, USA;
| | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel; (B.M.); (Y.B.); (A.B.); (E.M.)
- Correspondence: (Z.H.); (G.W.L.)
| | - Gee W. Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA; (H.E.C.); (J.Z.L.); (M.W.O.)
- Correspondence: (Z.H.); (G.W.L.)
| |
Collapse
|
29
|
Schefter BR, Nourbakhsh S, Taheri-Araghi S, Ha BY. Modeling Cell Selectivity of Antimicrobial Peptides: How Is the Selectivity Influenced by Intracellular Peptide Uptake and Cell Density. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 3:626481. [PMID: 35047907 PMCID: PMC8757749 DOI: 10.3389/fmedt.2021.626481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/20/2021] [Indexed: 01/18/2023] Open
Abstract
Antimicrobial peptides (AMPs) are known to attack bacteria selectively over their host cells. Many attempts have been made to use them as a template for designing peptide antibiotics for fighting drug-resistant bacteria. A central concept in this endeavor is “peptide selectivity,” which measures the “quality” of peptides. However, the relevance of selectivity measurements has often been obscured by the cell-density dependence of the selectivity. For instance, the selectivity can be overestimated if the cell density is larger for the host cell. Furthermore, recent experimental studies suggest that peptide trapping in target bacteria magnifies the cell-density dependence of peptide activity. Here, we propose a biophysical model for peptide activity and selectivity, which assists with the correct interpretation of selectivity measurements. The resulting model shows how cell density and peptide trapping in cells influence peptide activity and selectivity: while these effects can alter the selectivity by more than an order of magnitude, peptide trapping works in favor of host cells at high host-cell densities. It can be used to correct selectivity overestimates.
Collapse
Affiliation(s)
- Bethany R Schefter
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
| | - Shokoofeh Nourbakhsh
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
| | - Sattar Taheri-Araghi
- Department of Physics and Astronomy, California State University, Northridge, CA, United States
| | - Bae-Yeun Ha
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
30
|
Dittrich PS, Nuti N, Rottmann P, Stucki A, Koch P, Panke S. A Multiplexed Cell‐Free Assay to Screen for Antimicrobial Peptides in Double Emulsion Droplets. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202114632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Petra S Dittrich
- Eidgenossische Technische Hochschule Zurich Biosystems and Engineering Mattenstrasse 26 4058 Basel SWITZERLAND
| | - Nicola Nuti
- ETH Zurich: Eidgenossische Technische Hochschule Zurich Biosystems Science and Engineering SWITZERLAND
| | - Philipp Rottmann
- ETH Zurich: Eidgenossische Technische Hochschule Zurich Biosystems Science and Engineering SWITZERLAND
| | - Ariane Stucki
- ETH Zurich: Eidgenossische Technische Hochschule Zurich Biosystems Science and Engineering SWITZERLAND
| | - Philipp Koch
- ETH Zurich: Eidgenossische Technische Hochschule Zurich Biosystems Science and Engineering SWITZERLAND
| | - Sven Panke
- ETH Zurich: Eidgenossische Technische Hochschule Zurich Biosysystems Science and Engineering SWITZERLAND
| |
Collapse
|
31
|
Alizon S, Turner PE. Can we eradicate viral pathogens? J Evol Biol 2021; 34:1851-1854. [PMID: 34907625 DOI: 10.1111/jeb.13958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Samuel Alizon
- MIVEGEC, CNRS, IRD, Université de Montpellier, Montpellier, France
| | - Paul E Turner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, USA.,Program in Microbiology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
32
|
Timmons PB, Hewage CM. Conformation and membrane interaction studies of the potent antimicrobial and anticancer peptide palustrin-Ca. Sci Rep 2021; 11:22468. [PMID: 34789753 PMCID: PMC8599514 DOI: 10.1038/s41598-021-01769-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/03/2021] [Indexed: 01/13/2023] Open
Abstract
Palustrin-Ca (GFLDIIKDTGKEFAVKILNNLKCKLAGGCPP) is a host defence peptide with potent antimicrobial and anticancer activities, first isolated from the skin of the American bullfrog Lithobates catesbeianus. The peptide is 31 amino acid residues long, cationic and amphipathic. Two-dimensional NMR spectroscopy was employed to characterise its three-dimensional structure in a 50/50% water/2,2,2-trifluoroethanol-\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$d_{3}$$\end{document}d3 mixture. The structure is defined by an \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\alpha$$\end{document}α-helix that spans between Ile\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{6}$$\end{document}6-Ala\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{26}$$\end{document}26, and a cyclic disulfide-bridged domain at the C-terminal end of the peptide sequence, between residues 23 and 29. A molecular dynamics simulation was employed to model the peptide’s interactions with sodium dodecyl sulfate micelles, a widely used bacterial membrane-mimicking environment. Throughout the simulation, the peptide was found to maintain its \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\alpha$$\end{document}α-helical conformation between residues Ile\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{6}$$\end{document}6-Ala\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$^{26}$$\end{document}26, while adopting a position parallel to the surface to micelle, which is energetically-favourable due to many hydrophobic and electrostatic contacts with the micelle.
Collapse
Affiliation(s)
- Patrick B Timmons
- UCD School of Biomolecular and Biomedical Science, UCD Centre for Synthesis and Chemical Biology, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Chandralal M Hewage
- UCD School of Biomolecular and Biomedical Science, UCD Centre for Synthesis and Chemical Biology, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
33
|
You C, Jirků M, Corcoran DL, Parker W, Jirků-Pomajbíková K. Altered gut ecosystems plus the microbiota's potential for rapid evolution: A recipe for inevitable change with unknown consequences. Comput Struct Biotechnol J 2021; 19:5969-5977. [PMID: 34849201 PMCID: PMC8598968 DOI: 10.1016/j.csbj.2021.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/08/2021] [Accepted: 10/27/2021] [Indexed: 11/20/2022] Open
Abstract
In a single human gut, which is estimated to produce 1000-times more bacteria in a single day than the entire human population on Earth as of 2020, the potential for evolution is vast. In addition to the sheer volume of reproductive events, prokaryotes can transfer most genes horizontally, greatly accelerating their potential to evolve. In the face of this evolutionary potential, Westernization has led to profound changes in the ecosystem of the gut, including increased chronic inflammation in many individuals and dramatically reduced fiber consumption and decreased seasonal variation in the diet of most individuals. Experimental work using a variety of model systems has shown that bacteria will evolve within days to weeks when faced with substantial environmental changes. However, studies evaluating the effects of inflammation of the gut on the microbiota are still in their infancy and generally confounded by the effects of the microbiota on the immune system. At the same time, experimental data indicate that complete loss of fiber from the diet constitutes an extinction-level event for the gut microbiota. However, these studies evaluating diet may not apply to Westernized humans who typically have reduced but not absent levels of fiber in their diet. Thus, while it is expected that the microbiota will evolve rapidly in the face of Westernization, experimental studies that address the magnitude of that evolution are generally lacking, and it remains unknown to what extent this evolutionary process affects disease and the ability to treat the disease state.
Collapse
Affiliation(s)
- Celina You
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Global Health Institute, Duke University and Duke University School of Medicine, Durham, NC, USA
| | - Milan Jirků
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 370 05 České Budějovice, Czech Republic
| | - David L. Corcoran
- Genomic Analysis and Bioinformatics Shared Resource, Duke Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC, USA
| | - William Parker
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Global Health Institute, Duke University and Duke University School of Medicine, Durham, NC, USA
| | - Kateřina Jirků-Pomajbíková
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 370 05 České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, 370 05 České Budějovice, Czech Republic
| |
Collapse
|
34
|
Host Defense Peptides: Dual Antimicrobial and Immunomodulatory Action. Int J Mol Sci 2021; 22:ijms222011172. [PMID: 34681833 PMCID: PMC8538224 DOI: 10.3390/ijms222011172] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022] Open
Abstract
The rapid rise of multidrug-resistant (MDR) bacteria has once again caused bacterial infections to become a global health concern. Antimicrobial peptides (AMPs), also known as host defense peptides (HDPs), offer a viable solution to these pathogens due to their diverse mechanisms of actions, which include direct killing as well as immunomodulatory properties (e.g., anti-inflammatory activity). HDPs may hence provide a more robust treatment of bacterial infections. In this review, the advent of and the mechanisms that lead to antibiotic resistance will be described. HDP mechanisms of antibacterial and immunomodulatory action will be presented, with specific examples of how the HDP aurein 2.2 and a few of its derivatives, namely peptide 73 and cG4L73, function. Finally, resistance that may arise from a broader use of HDPs in a clinical setting and methods to improve biocompatibility will be briefly discussed.
Collapse
|
35
|
Timmons PB, Hewage CM. Biophysical study of the structure and dynamics of the antimicrobial peptide maximin 1. J Pept Sci 2021; 28:e3370. [PMID: 34569121 DOI: 10.1002/psc.3370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/18/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022]
Abstract
Maximin 1 is a cationic, amphipathic antimicrobial peptide found in the skin secretions and brains of the Chinese red belly toad Bombina maxima. The 27 amino acid residue-long peptide is biologically interesting as it possesses a variety of biological activities, including antibacterial, antifungal, antiviral, antitumour and spermicidal activities. Its three-dimensional structural model was obtained in a 50/50% water/2,2,2-trifluoroethanol-d3 mixture using two-dimensional NMR spectroscopy. Maximin 1 was found to adopt an α-helical structure from residue Ile2 to Ala26 . The peptide is amphipathic, showing a clear separation between polar and non-polar residues. The interactions with sodium dodecyl sulfate micelles, a widely-used bacterial membrane-mimicking environment, were modelled using molecular dynamics simulations. The peptide maintains an α-helical conformation, occasionally displaying a flexibility around the Gly9 and Gly16 residues, which is likely responsible for the peptide's low haemolytic activity. It is found to preferentially adopt a position parallel to the micellar surface, establishing a number of hydrophobic and electrostatic interactions with the micelle.
Collapse
Affiliation(s)
- Patrick B Timmons
- UCD School of Biomolecular and Biomedical Science,UCD Centre for Synthesis and Chemical Biology, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Chandralal M Hewage
- UCD School of Biomolecular and Biomedical Science,UCD Centre for Synthesis and Chemical Biology, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
36
|
Cisse A, Marquette A, Altangerel M, Peters J, Bechinger B. Investigation of the Action of Peptides on Lipid Membranes. J Phys Chem B 2021; 125:10213-10223. [PMID: 34464136 DOI: 10.1021/acs.jpcb.1c06388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Calorimetric and incoherent neutron scattering methods were employed to investigate the action of magainin 2 and PGLa peptides on the phase behavior and molecular dynamics of lipids mimicking cytoplasmic membranes of Gram-negative bacteria. The impact of the peptides, tested individually and cooperatively by differential scanning calorimetry, presented a broadened peak, sometimes with a second shoulder, depicting the phase transition temperature around 21 °C. Neutron scattering revealed a small but significant variation of the membrane dynamics due to the peptides in both in-plane and out-of-plane directions. Although we did not find a clear hint for synergy in the interplay of the two peptides, the calorimetric and neutron data give compatible results in terms of a decrease of the enthalpy due to the presence of the peptides, which destabilize the membrane. The dynamics in the two directions was differentiated when the individual peptides were added to the membranes, but the impact was smaller when both peptides were added together.
Collapse
Affiliation(s)
- Aline Cisse
- Univ. Grenoble-Alpes, CNRS, LiPhy, 38000 Grenoble, France.,Institut Laue-Langevin, 38000 Grenoble, France
| | - Arnaud Marquette
- University of Strasbourg/CNRS, Chemistry Institute, Membrane Biophysics and NMR, UMR7177 Strasbourg, France
| | - Munkhtuguldur Altangerel
- Univ. Grenoble-Alpes, CNRS, LiPhy, 38000 Grenoble, France.,Institut Laue-Langevin, 38000 Grenoble, France
| | - Judith Peters
- Univ. Grenoble-Alpes, CNRS, LiPhy, 38000 Grenoble, France.,Institut Laue-Langevin, 38000 Grenoble, France.,Institut Universitaire de France, 75231 Paris, France
| | - Burkhard Bechinger
- University of Strasbourg/CNRS, Chemistry Institute, Membrane Biophysics and NMR, UMR7177 Strasbourg, France.,Institut Universitaire de France, 75231 Paris, France
| |
Collapse
|
37
|
Miao F, Li Y, Tai Z, Zhang Y, Gao Y, Hu M, Zhu Q. Antimicrobial Peptides: The Promising Therapeutics for Cutaneous Wound Healing. Macromol Biosci 2021; 21:e2100103. [PMID: 34405955 DOI: 10.1002/mabi.202100103] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/31/2021] [Indexed: 12/12/2022]
Abstract
Chronic wound infections have caused an increasing number of deaths and economic burden, which necessitates wound treatment options. Hitherto, the development of functional wound dressings has achieved reasonable progress. Antibacterial agents, growth factors, and miRNAs are incorporated in different wound dressings to treat various types of wounds. As an effective antimicrobial agent and emerging wound healing therapeutic, antimicrobial peptides (AMPs) have attracted significant attention. The present study focuses on the application of AMPs in wound healing and discusses the types, properties and formulation strategies of AMPs used for wound healing. In addition, the clinical trial and the current status of studies on "antimicrobial peptides and wound healing" are elaborated through bibliometrics. Also, the challenges and opportunities for further development and utilization of AMP formulations in wound healing are discussed.
Collapse
Affiliation(s)
- Fengze Miao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China.,National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.,Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Ying Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China
| | - Yong Zhang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.,Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Yue Gao
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.,Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Menghong Hu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China.,National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.,Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, 200443, China
| |
Collapse
|
38
|
Patiño MI, Restrepo LM, Becerra NY, van der Mei HC, van Kooten TG, Sharma PK. Nonviral Expression of LL-37 in a Human Skin Equivalent to Prevent Infection in Skin Wounds. Hum Gene Ther 2021; 32:1147-1157. [PMID: 33980038 DOI: 10.1089/hum.2021.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Inefficient autologous tissue recovery in skin wounds increases the susceptibility of patients to infections caused by multidrug resistant microorganisms, resulting in a high mortality rate. Genetic modification of skin cells has become an important field of study because it could lead to the construction of more functional skin grafts, through the overexpression of antimicrobial peptides that would prevent early contamination and infection with bacteria. In this study, we produce and evaluate human skin equivalents (HSEs) containing transfected human primary fibroblasts and keratinocytes by polyplexes to express the antimicrobial peptide LL-37. The effect of LL-37 on the metabolic activity of normal HSEs was evaluated before the construction of the transfected HSEs, and the antimicrobial efficacy against Pseudomonas aeruginosa and Staphylococcus aureus was evaluated. Subsequently, the levels of LL-37 in the culture supernatants of transfected HSEs, as well as the local expression, were determined. It was found that LL-37 treatment significantly promoted the cellular proliferation of HSEs. Furthermore, HSEs that express elevated levels of LL-37 were shown to possess histological characteristics close to the normal skin and display enhanced antimicrobial activity against S. aureus in vitro. These findings demonstrate that HSEs expressing LL-37 through nonviral modification of skin cells are a promising approach for the prevention of bacterial colonization in wounds.
Collapse
Affiliation(s)
- Maria Isabel Patiño
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia.,Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Luz Marina Restrepo
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Natalia Yiset Becerra
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Henny C van der Mei
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Theo G van Kooten
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Prashant K Sharma
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
39
|
Rodríguez-Rojas A, Baeder DY, Johnston P, Regoes RR, Rolff J. Bacteria primed by antimicrobial peptides develop tolerance and persist. PLoS Pathog 2021; 17:e1009443. [PMID: 33788905 PMCID: PMC8041211 DOI: 10.1371/journal.ppat.1009443] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 04/12/2021] [Accepted: 03/02/2021] [Indexed: 12/21/2022] Open
Abstract
Antimicrobial peptides (AMPs) are key components of innate immune defenses. Because of the antibiotic crisis, AMPs have also come into focus as new drugs. Here, we explore whether prior exposure to sub-lethal doses of AMPs increases bacterial survival and abets the evolution of resistance. We show that Escherichia coli primed by sub-lethal doses of AMPs develop tolerance and increase persistence by producing curli or colanic acid, responses linked to biofilm formation. We develop a population dynamic model that predicts that priming delays the clearance of infections and fuels the evolution of resistance. The effects we describe should apply to many AMPs and other drugs that target the cell surface. The optimal strategy to tackle tolerant or persistent cells requires high concentrations of AMPs and fast and long-lasting expression. Our findings also offer a new understanding of non-inherited drug resistance as an adaptive response and could lead to measures that slow the evolution of resistance. Animals and plants defend themselves with ancient molecules called antimicrobial peptides (AMPs) against pathogens. As more and more bacterial diseases have become drug resistant, these AMPs are considered as promising alternatives. In natural situation such as on the skin, bacteria are often exposed to low concentrations of AMPs that do no kill. Here we show that the bacterium Escherichia coli when exposed to such low concentrations becomes recalcitrant to killing concentrations of the same AMPs. We report the ways in which the bacteria alter their surface to do so. We then use a mathematical model to show that these effects caused by low concentrations can drive the evolution of resistance. From the perspective of an organism using AMPs in self-defense, the best option is to deploy high concentrations of AMPs for long. Our findings also offer a new understanding of similar drug resistance mechanisms.
Collapse
Affiliation(s)
| | | | - Paul Johnston
- Berlin Center for Genomics in Biodiversity Research, Berlin, Germany
- Leibniz-Institute of Freshwater Ecology and Inland Fisheries (IGB), Berlin, Germany
| | - Roland R. Regoes
- Institute of Integrative Biology, Zürich, Switzerland
- * E-mail: (RRR); (JR)
| | - Jens Rolff
- Freie Universität Berlin, Institut für Biologie, Evolutionary Biology, Berlin, Germany
- Berlin Center for Genomics in Biodiversity Research, Berlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research (BBIB), Berlin, Germany
- * E-mail: (RRR); (JR)
| |
Collapse
|
40
|
Bennett RC, Oh MW, Kuo SH, Belo Y, Maron B, Malach E, Lin J, Hayouka Z, Lau GW. Random Peptide Mixtures as Safe and Effective Antimicrobials against Pseudomonas aeruginosa and MRSA in Mouse Models of Bacteremia and Pneumonia. ACS Infect Dis 2021; 7:672-680. [PMID: 33650856 DOI: 10.1021/acsinfecdis.0c00871] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antibiotic resistance is a daunting challenge in modern medicine, and novel approaches that minimize the emergence of resistant pathogens are desperately needed. Antimicrobial peptides are newer therapeutics that attempt to do this; however, they fall short because of low to moderate antimicrobial activity, low protease stability, susceptibility to resistance development, and high cost of production. The recently developed random peptide mixtures (RPMs) are promising alternatives. RPMs are synthesized by incorporating a defined proportion of two amino acids at each coupling step rather than just one, making them highly variable but still defined in their overall composition, chain length, and stereochemistry. Because RPMs have extreme diversity, it is unlikely that bacteria would be capable of rapidly evolving resistance. However, their efficacy against pathogens in animal models of human infectious diseases remained uncharacterized. Here, we demonstrated that RPMs have strong safety and pharmacokinetic profiles. RPMs rapidly killed both Pseudomonas aeruginosa and Staphylococcus aureus efficiently and disrupted preformed biofilms by both pathogens. Importantly, RPMs were efficacious against both pathogens in mouse models of bacteremia and acute pneumonia. Our results demonstrate that RPMs are potent broad-spectrum therapeutics against antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Richard C. Bennett
- Department of Pathobiology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61802, United States
| | - Myung Whan Oh
- Department of Pathobiology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61802, United States
| | - Shanny Hsuan Kuo
- Department of Pathobiology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61802, United States
| | - Yael Belo
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Bar Maron
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Einav Malach
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Jingjun Lin
- Department of Pathobiology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61802, United States
| | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Gee W. Lau
- Department of Pathobiology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61802, United States
| |
Collapse
|
41
|
Lim DSW, Yuan Y, Zhang Y. pH-Degradable Polymers as Impermanent Antimicrobial Agents for Environmental Sustainability. ACS APPLIED BIO MATERIALS 2021; 4:1544-1551. [DOI: 10.1021/acsabm.0c01402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Diane S. W. Lim
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos #07-01, Singapore 138669, Singapore
| | - Yuan Yuan
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos #07-01, Singapore 138669, Singapore
| | - Yugen Zhang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos #07-01, Singapore 138669, Singapore
| |
Collapse
|
42
|
Attieh Z, Mouawad C, Rejasse A, Jehanno I, Perchat S, Hegna IK, Økstad OA, Kallassy Awad M, Sanchis-Borja V, El Chamy L. The fliK Gene Is Required for the Resistance of Bacillus thuringiensis to Antimicrobial Peptides and Virulence in Drosophila melanogaster. Front Microbiol 2020; 11:611220. [PMID: 33391240 PMCID: PMC7775485 DOI: 10.3389/fmicb.2020.611220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/18/2020] [Indexed: 11/13/2022] Open
Abstract
Antimicrobial peptides (AMPs) are essential effectors of the host innate immune system and they represent promising molecules for the treatment of multidrug resistant microbes. A better understanding of microbial resistance to these defense peptides is thus prerequisite for the control of infectious diseases. Here, using a random mutagenesis approach, we identify the fliK gene, encoding an internal molecular ruler that controls flagella hook length, as an essential element for Bacillus thuringiensis resistance to AMPs in Drosophila. Unlike its parental strain, that is highly virulent to both wild-type and AMPs deficient mutant flies, the fliK deletion mutant is only lethal to the latter's. In agreement with its conserved function, the fliK mutant is non-flagellated and exhibits highly compromised motility. However, comparative analysis of the fliK mutant phenotype to that of a fla mutant, in which the genes encoding flagella proteins are interrupted, indicate that B. thuringiensis FliK-dependent resistance to AMPs is independent of flagella assembly. As a whole, our results identify FliK as an essential determinant for B. thuringiensis virulence in Drosophila and provide new insights on the mechanisms underlying bacteria resistance to AMPs.
Collapse
Affiliation(s)
- Zaynoun Attieh
- UR-EGP, Faculté des Sciences, Université Saint-Joseph de Beyrouth, Beirut, Lebanon
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Carine Mouawad
- UR-EGP, Faculté des Sciences, Université Saint-Joseph de Beyrouth, Beirut, Lebanon
| | - Agnès Rejasse
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Isabelle Jehanno
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Stéphane Perchat
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Ida K. Hegna
- Department of Pharmacy, Centre for Integrative Microbial Evolution (CIME), Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Ole A. Økstad
- Department of Pharmacy, Centre for Integrative Microbial Evolution (CIME), Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | | | - Vincent Sanchis-Borja
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Laure El Chamy
- UR-EGP, Faculté des Sciences, Université Saint-Joseph de Beyrouth, Beirut, Lebanon
| |
Collapse
|
43
|
O'Connor LMJ, Fugère V, Gonzalez A. Evolutionary Rescue Is Mediated by the History of Selection and Dispersal in Diversifying Metacommunities. Front Ecol Evol 2020. [DOI: 10.3389/fevo.2020.517434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Rapid evolution can sometimes prevent population extirpation in stressful environments, but the conditions leading to “evolutionary rescue” in metacommunities are unclear. Here we studied the eco-evolutionary response of microbial metacommunities adapting to selection by the antibiotic streptomycin. Our experiment tested how the history of antibiotic selection and contrasting modes of dispersal influenced diversification and subsequent evolutionary rescue in microbial metacommunities undergoing adaptive radiation. We first tracked the change in diversity and density of Pseudomonas fluorescens morphotypes selected on a gradient of antibiotic stress. We then examined the recovery of these metacommunities following abrupt application of a high concentration of streptomycin lethal to the ancestral organisms. We show that dispersal increases diversity within the stressed metacommunities, that exposure to stress alters diversification dynamics, and that community composition, dispersal, and past exposure to stress mediate the speed at which evolutionary rescue occurs, but not the final outcome of recovery in abundance and diversity. These findings extend recent experiments on evolutionary rescue to the case of metacommunities undergoing adaptive diversification, and should motivate new theory on this question. Our findings are also relevant to evolutionary conservation biology and research on antimicrobial resistance.
Collapse
|
44
|
Shen Z, Guo Z, Zhou L, Wang Y, Zhang J, Hu J, Zhang Y. Biomembrane induced in situ self-assembly of peptide with enhanced antimicrobial activity. Biomater Sci 2020; 8:2031-2039. [PMID: 32083626 DOI: 10.1039/c9bm01785b] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Antimicrobial peptides (AMPs) as biocides are of great interest because they have the ability to combat antibiotic resistance. Normally, natural AMPs need to be rationally designed or modified for practical use as an antibiotic. Here, a novel AMP, termed FF8, which is a cationic octapeptide composed of arginine, lysine, and phenylalanine, was designed. The FF8 was found to self-assemble into nanofibers when induced by a negatively charged lipid membrane or pH is above 9.4. The fibers on the membrane broke the lipid membrane, forming pores and significantly reducing its fluidity. FF8 also exhibited enhanced antibacterial activity by significantly increasing the permeability of the inner and outer membranes of Escherichia coli (E. coli) and maintaining the pores of the inner membrane of cells, which caused continuous membrane leakage. Because of its high antibacterial activity, cytocompatibility, and cost-effectiveness, FF8 is a promising antibacterial material.
Collapse
Affiliation(s)
- Zhiwei Shen
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Guo
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Limin Zhou
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China and Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Yujiao Wang
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinjin Zhang
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China and Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Jun Hu
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China and Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Yi Zhang
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China and Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| |
Collapse
|
45
|
Mücke PA, Ostrzinski A, Hammerschmidt S, Maaß S, Becher D. Proteomic Adaptation of Streptococcus pneumoniae to the Antimicrobial Peptide Human Beta Defensin 3 (hBD3) in Comparison to Other Cell Surface Stresses. Microorganisms 2020; 8:microorganisms8111697. [PMID: 33143252 PMCID: PMC7694020 DOI: 10.3390/microorganisms8111697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
The antimicrobial peptide human Beta defensin 3 (hBD3) is an essential part of the innate immune system and is involved in protection against respiratory pathogens by specifically permeabilizing bacterial membranes. The Gram-positive bacterium Streptococcus pneumoniae causes serious diseases including pneumonia, meningitis, and septicemia, despite being frequently exposed to human defense molecules, including hBD3 during colonization and infection. Thus, the question arises how pneumococci adapt to stress caused by antimicrobial peptides. We addressed this subject by analyzing the proteome of S. pneumoniae after treatment with hBD3 and compared our data with the proteomic changes induced by LL-37, another crucial antimicrobial peptide present in the human respiratory tract. As antimicrobial peptides usually cause membrane perturbations, the response to the membrane active cationic detergent cetyltrimethylammonium bromide (CTAB) was examined to assess the specificity of the pneumococcal response to antimicrobial peptides. In brief, hBD3 and LL-37 induce a similar response in pneumococci and especially, changes in proteins with annotated transporter and virulence function have been identified. However, LL-37 causes changes in the abundance of cell surface modification proteins that cannot be observed after treatment with hBD3. Interestingly, CTAB induces unique proteomic changes in S. pneumoniae. Though, the detergent seems to activate a two-component system that is also activated in response to antimicrobial peptide stress (TCS 05). Overall, our data represent a novel resource on pneumococcal adaptation to specific cell surface stresses on a functional level. This knowledge can potentially be used to develop strategies to circumvent pneumococcal resistance to antimicrobial peptides.
Collapse
Affiliation(s)
- Pierre-Alexander Mücke
- Department of Microbial Proteomics, Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Anne Ostrzinski
- Department of Microbial Proteomics, Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Sandra Maaß
- Department of Microbial Proteomics, Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Dörte Becher
- Department of Microbial Proteomics, Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| |
Collapse
|
46
|
Hill T, Unckless RL. Recurrent evolution of high virulence in isolated populations of a DNA virus. eLife 2020; 9:e58931. [PMID: 33112738 PMCID: PMC7685711 DOI: 10.7554/elife.58931] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/28/2020] [Indexed: 12/30/2022] Open
Abstract
Hosts and viruses are constantly evolving in response to each other: as a host attempts to suppress a virus, the virus attempts to evade and suppress the host's immune system. Here, we describe the recurrent evolution of a virulent strain of a DNA virus, which infects multiple Drosophila species. Specifically, we identified two distinct viral types that differ 100-fold in viral titer in infected individuals, with similar differences observed in multiple species. Our analysis suggests that one of the viral types recurrently evolved at least four times in the past ~30,000 years, three times in Arizona and once in another geographically distinct species. This recurrent evolution may be facilitated by an effective mutation rate which increases as each prior mutation increases viral titer and effective population size. The higher titer viral type suppresses the host-immune system and an increased virulence compared to the low viral titer type.
Collapse
Affiliation(s)
- Tom Hill
- The Department of Molecular Biosciences, University of KansasLawrenceUnited States
| | - Robert L Unckless
- The Department of Molecular Biosciences, University of KansasLawrenceUnited States
| |
Collapse
|
47
|
Bansal S, Su WC, Budamagunta M, Xiao W, Ajena Y, Liu R, Voss JC, Carney RP, Parikh AN, Lam KS. Discovery and mechanistic characterization of a structurally-unique membrane active peptide. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183394. [PMID: 32562695 PMCID: PMC7478859 DOI: 10.1016/j.bbamem.2020.183394] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 05/21/2020] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
Abstract
Membrane active peptides (MAPs) have gained wide interest due to their far reaching applications in drug discovery and drug delivery. The search for new MAPs, however, has been largely skewed with bias selecting for physicochemical parameters believed to be important for membrane activity, such as alpha helicity, cationicity and hydrophobicity. Here we carry out a search-and-find strategy to screen a 100,000-membered one-bead-one-compound (OBOC) combinatorial peptide library for lead compounds, agnostic of those physicochemical constraints. Such a synthetic strategy also permits expansion of our peptide repertoire to include unnatural amino acids. Using this approach, we discovered a structurally unique lead peptide LBF14, a linear 14-mer peptide, that induces gross morphological disruption of membranes, irrespective of membrane composition. Further, we demonstrate that the unique insertion mechanism of the peptide, visualized by spinning disc confocal microscopy and further analyzed by electron paramagnetic resonance measurements, may be the cause of this large scale membrane deformation. We also demonstrate the robustness, reproducibility, and potential application of this technique to discover and characterize new membrane active peptides that display activity by local insertion and subsequent allosteric effects leading to global membrane disruption.
Collapse
Affiliation(s)
- Shivani Bansal
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, United States of America; Department of Chemistry, University of California, Davis, United States of America
| | - Wan-Chih Su
- Department of Chemistry, University of California, Davis, United States of America; Department of Biomedical Engineering, University of California, Davis, United States of America
| | - Madhu Budamagunta
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, United States of America
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, United States of America
| | - Yousif Ajena
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, United States of America
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, United States of America
| | - John C Voss
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, United States of America
| | - Randy P Carney
- Department of Biomedical Engineering, University of California, Davis, United States of America
| | - Atul N Parikh
- Department of Biomedical Engineering, University of California, Davis, United States of America
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, United States of America; Department of Chemistry, University of California, Davis, United States of America.
| |
Collapse
|
48
|
Rodríguez-Rojas A, Nath A, El Shazely B, Santi G, Kim JJ, Weise C, Kuropka B, Rolff J. Antimicrobial Peptide Induced-Stress Renders Staphylococcus aureus Susceptible to Toxic Nucleoside Analogs. Front Immunol 2020; 11:1686. [PMID: 33133056 PMCID: PMC7550632 DOI: 10.3389/fimmu.2020.01686] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Cationic antimicrobial peptides (AMPs) are active immune effectors of multicellular organisms and are also considered as new antimicrobial drug candidates. One of the problems encountered when developing AMPs as drugs is the difficulty of reaching sufficient killing concentrations under physiological conditions. Here, using pexiganan, a cationic peptide derived from a host defense peptide of the African clawed frog and the first AMP developed into an antibacterial drug, we studied whether sub-lethal effects of AMPs can be harnessed to devise treatment combinations. We studied the pexiganan stress response of Staphylococcus aureus at sub-lethal concentrations using quantitative proteomics. Several proteins involved in nucleotide metabolism were elevated, suggesting a metabolic demand. We then show that Staphylococcus aureus is highly susceptible to antimetabolite nucleoside analogs when exposed to pexiganan, even at sub-inhibitory concentrations. These findings could be used to enhance pexiganan potency while decreasing the risk of resistance emergence, and our findings can likely be extended to other antimicrobial peptides.
Collapse
Affiliation(s)
| | - Arpita Nath
- Institut für Biologie, Evolutionary Biology, Freie Universität Berlin, Berlin, Germany
| | - Baydaa El Shazely
- Institut für Biologie, Evolutionary Biology, Freie Universität Berlin, Berlin, Germany
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Greta Santi
- Institut für Biologie, Evolutionary Biology, Freie Universität Berlin, Berlin, Germany
| | - Joshua Jay Kim
- Institut für Biologie, Evolutionary Biology, Freie Universität Berlin, Berlin, Germany
| | - Christoph Weise
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Benno Kuropka
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Jens Rolff
- Institut für Biologie, Evolutionary Biology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
49
|
Bonnel C, Legrand B, Simon M, Clavié M, Masnou A, Jumas-Bilak E, Kang YK, Licznar-Fajardo P, Maillard LT, Masurier N. Tailoring the Physicochemical Properties of Antimicrobial Peptides onto a Thiazole-Based γ-Peptide Foldamer. J Med Chem 2020; 63:9168-9180. [DOI: 10.1021/acs.jmedchem.0c00077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Clément Bonnel
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, UM, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 15 Avenue Charles Flahault, 34093 Montpellier Cedex 5, France
| | - Baptiste Legrand
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, UM, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 15 Avenue Charles Flahault, 34093 Montpellier Cedex 5, France
| | - Matthieu Simon
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, UM, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 15 Avenue Charles Flahault, 34093 Montpellier Cedex 5, France
| | - Margaux Clavié
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, UM, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 15 Avenue Charles Flahault, 34093 Montpellier Cedex 5, France
| | - Agnès Masnou
- Laboratoire HydroSciences Montpellier, UMR 5569, CNRS, UM, IRD, Département d’Hygiène Hospitalière−CHU Montpellier, 34095 Montpellier, France
| | - Estelle Jumas-Bilak
- Laboratoire HydroSciences Montpellier, UMR 5569, CNRS, UM, IRD, Département d’Hygiène Hospitalière−CHU Montpellier, 34095 Montpellier, France
| | - Young Kee Kang
- Department of Chemistry, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Patricia Licznar-Fajardo
- Laboratoire HydroSciences Montpellier, UMR 5569, CNRS, UM, IRD, Département d’Hygiène Hospitalière−CHU Montpellier, 34095 Montpellier, France
| | - Ludovic T. Maillard
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, UM, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 15 Avenue Charles Flahault, 34093 Montpellier Cedex 5, France
| | - Nicolas Masurier
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, UM, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 15 Avenue Charles Flahault, 34093 Montpellier Cedex 5, France
| |
Collapse
|
50
|
Revilla-Guarinos A, Zhang Q, Loderer C, Alcántara C, Müller A, Rahnamaeian M, Vilcinskas A, Gebhard S, Zúñiga M, Mascher T. ABC Transporter DerAB of Lactobacillus casei Mediates Resistance against Insect-Derived Defensins. Appl Environ Microbiol 2020; 86:e00818-20. [PMID: 32414796 PMCID: PMC7357469 DOI: 10.1128/aem.00818-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/09/2020] [Indexed: 01/28/2023] Open
Abstract
Bce-like systems mediate resistance against antimicrobial peptides in Firmicutes bacteria. Lactobacillus casei BL23 encodes an "orphan" ABC transporter that, based on homology to BceAB-like systems, was proposed to contribute to antimicrobial peptide resistance. A mutant lacking the permease subunit was tested for sensitivity against a collection of peptides derived from bacteria, fungi, insects, and humans. Our results show that the transporter specifically conferred resistance against insect-derived cysteine-stabilized αβ defensins, and it was therefore renamed DerAB for defensin resistance ABC transporter. Surprisingly, cells lacking DerAB showed a marked increase in resistance against the lantibiotic nisin. This could be explained by significantly increased expression of the antimicrobial peptide resistance determinants regulated by the Bce-like systems PsdRSAB (formerly module 09) and ApsRSAB (formerly module 12). Bacterial two-hybrid studies in Escherichia coli showed that DerB could interact with proteins of the sensory complex in the Psd resistance system. We therefore propose that interaction of DerAB with this complex in the cell creates signaling interference and reduces the cell's potential to mount an effective nisin resistance response. In the absence of DerB, this negative interference is relieved, leading to the observed hyperactivation of the Psd module and thus increased resistance to nisin. Our results unravel the function of a previously uncharacterized Bce-like orphan resistance transporter with pleiotropic biological effects on the cell.IMPORTANCE Antimicrobial peptides (AMPs) play an important role in suppressing the growth of microorganisms. They can be produced by bacteria themselves-to inhibit competitors-but are also widely distributed in higher eukaryotes, including insects and mammals, where they form an important component of innate immunity. In low-GC-content Gram-positive bacteria, BceAB-like transporters play a crucial role in AMP resistance but have so far been primarily associated with interbacterial competition. Here, we show that the orphan transporter DerAB from the lactic acid bacterium Lactobacillus casei is crucial for high-level resistance against insect-derived AMPs. It therefore represents an important mechanism for interkingdom defense. Furthermore, our results support a signaling interference from DerAB on the PsdRSAB module that might prevent the activation of a full nisin response. The Bce modules from L. casei BL23 illustrate a biological paradox in which the intrinsic nisin detoxification potential only arises in the absence of a defensin-specific ABC transporter.
Collapse
Affiliation(s)
| | - Qian Zhang
- Institut für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| | - Christoph Loderer
- Institut für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| | - Cristina Alcántara
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Ariane Müller
- Institut für Zoologie, Technische Universität Dresden, Dresden, Germany
| | - Mohammad Rahnamaeian
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Department of Bioresources, Giessen, Germany
| | - Andreas Vilcinskas
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Department of Bioresources, Giessen, Germany
- Institute for Insect Biotechnology, Justus Liebig University Giessen, Giessen, Germany
| | - Susanne Gebhard
- Department of Biology and Biochemistry, Milner Centre for Evolution, University of Bath, United Kingdom
| | - Manuel Zúñiga
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), Valencia, Spain
| | - Thorsten Mascher
- Institut für Mikrobiologie, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|