51
|
Moazzezy N, Rismani E, Rezaei M, Karam MRA, Rafati S, Bouzari S, Oloomi M. Computational evaluation of modified peptides from human neutrophil peptide 1 (HNP-1). J Biomol Struct Dyn 2020; 40:1163-1171. [PMID: 32981420 DOI: 10.1080/07391102.2020.1823249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The development of bacterial resistance toward antibiotics has been led to pay attention to the antimicrobial peptides (AMPs). The common mechanism of AMPs is disrupting the integrity of the bacterial membrane. One of the most accessible targets for α-defensins human neutrophil peptide-1 (HNP-1) is lipid II. In the present study, we performed homology modeling and geometrical validation of human neutrophil defensin 1. Then, the conformational and physicochemical properties of HNP-1 derived peptides 2Abz14S29, 2Abz23S29, and HNP1ΔC18A, as well as their interaction with lipid II were studied computationally. The overall quality of the predicted model of full protein was -5.14, where over 90% of residues were in the most favored and allowed regions in the Ramachandran plot. Although HNP-1 and HNP1ΔC18A were classified as unstable peptides, 2Abz14S29 and 2Abz23S29 were stable, based on the instability index values. Molecular docking showed similar interaction pattern of peptides and HNP-1 to lipid II. Molecular dynamic simulations revealed the overall stability of conformations, though the fluctuations of amino acids in the modified peptides were relatively higher than HNP-1. Further, the binding affinity constant (Kd) of HNP-1 and 2Abz23S29 in complex with lipid II was 10 times stronger than 2Abz14S29 and HNP1ΔC18A. Overall, computational studies of conformational and interaction patterns have signified how derived peptides could have displayed relatively similar antimicrobial results compared to HNP-1 in the reported experimental studies. Chemical modifications not only have improved the physicochemical properties of derived peptides compared to HNP-1, but also they have retained the similar pattern and binding affinity of peptides. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Neda Moazzezy
- Molecular Biology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Rismani
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Maryam Rezaei
- Molecular Biology Department, Pasteur Institute of Iran, Tehran, Iran
| | | | - Sima Rafati
- Immunotherapy and Leishmania Vaccine Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Saeid Bouzari
- Molecular Biology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mana Oloomi
- Molecular Biology Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
52
|
Overview of Host Defense Peptides and Their Applications for Plastic and Reconstructive Surgeons. Plast Reconstr Surg 2020; 146:91-103. [PMID: 32590651 DOI: 10.1097/prs.0000000000006910] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Host defense peptides are a family of endogenous short peptides that are found in all living beings and play a critical role in innate immunity against infection. METHODS A nonsystematic review of host defense peptides was conducted with specific interest in properties and applications relevant to plastic and reconstructive surgery. RESULTS In addition to their direct antimicrobial actions against pathogens, including multidrug-resistant bacteria, they also demonstrate important functions in immunomodulation, tumor cell lysis, and tissue regeneration. These properties have made them a topic of clinical interest for plastic surgeons because of their potential applications as novel antibiotics, wound healing medications, and cancer therapies. The rising clinical interest has led to a robust body of literature describing host defense peptides in great depth and breadth. Numerous mechanisms have been observed to explain their diverse functions, which rely on specific structural characteristics. However, these peptides remain mostly experimental, with limited translation to clinical practice because of numerous failures to achieve acceptable results in human trials. CONCLUSIONS Despite the broad ranging potential of these peptides for use in the field of plastic and reconstructive surgery, they are rarely discussed in the literature or at scientific meetings. In this review, the authors provide a summary of the background, structure, function, bacterial resistance, and clinical applications of host defense peptides with the goal of stimulating host defense peptide-based innovation within the field of plastic and reconstructive surgery.
Collapse
|
53
|
Magana M, Pushpanathan M, Santos AL, Leanse L, Fernandez M, Ioannidis A, Giulianotti MA, Apidianakis Y, Bradfute S, Ferguson AL, Cherkasov A, Seleem MN, Pinilla C, de la Fuente-Nunez C, Lazaridis T, Dai T, Houghten RA, Hancock REW, Tegos GP. The value of antimicrobial peptides in the age of resistance. THE LANCET. INFECTIOUS DISEASES 2020; 20:e216-e230. [PMID: 32653070 DOI: 10.1016/s1473-3099(20)30327-3] [Citation(s) in RCA: 676] [Impact Index Per Article: 135.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/29/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022]
Abstract
Accelerating growth and global expansion of antimicrobial resistance has deepened the need for discovery of novel antimicrobial agents. Antimicrobial peptides have clear advantages over conventional antibiotics which include slower emergence of resistance, broad-spectrum antibiofilm activity, and the ability to favourably modulate the host immune response. Broad bacterial susceptibility to antimicrobial peptides offers an additional tool to expand knowledge about the evolution of antimicrobial resistance. Structural and functional limitations, combined with a stricter regulatory environment, have hampered the clinical translation of antimicrobial peptides as potential therapeutic agents. Existing computational and experimental tools attempt to ease the preclinical and clinical development of antimicrobial peptides as novel therapeutics. This Review identifies the benefits, challenges, and opportunities of using antimicrobial peptides against multidrug-resistant pathogens, highlights advances in the deployment of novel promising antimicrobial peptides, and underlines the needs and priorities in designing focused development strategies taking into account the most advanced tools available.
Collapse
Affiliation(s)
- Maria Magana
- Department of Biopathology and Clinical Microbiology, Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Ana L Santos
- Department of Chemistry, Rice University, Houston, TX, USA; Investigación Sanitaria de las Islas Baleares, Palma, Spain
| | - Leon Leanse
- Department of Dermatology, Harvard Medical School, Boston, MA, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Michael Fernandez
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | - Steven Bradfute
- Department of Internal Medicine, Center for Global Health, University of New Mexico, Albuquerque, NM, USA
| | - Andrew L Ferguson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Artem Cherkasov
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Mohamed N Seleem
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, IN, USA
| | - Clemencia Pinilla
- Torrey Pines Institute for Molecular Studies, Port St Lucie, FL, USA
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Penn Institute for Computational Science, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Themis Lazaridis
- Department of Chemistry, The City College of New York, New York, NY, USA; Graduate Programs in Chemistry, Biochemistry, and Physics, The Graduate Center, City University of New York, NY, USA
| | - Tianhong Dai
- Department of Dermatology, Harvard Medical School, Boston, MA, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Robert E W Hancock
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - George P Tegos
- Reading Hospital, Tower Health, West Reading, PA, USA; Micromoria, Venture X Marlborough, Marlborough, MA, USA.
| |
Collapse
|
54
|
Li J, Fernández-Millán P, Boix E. Synergism between Host Defence Peptides and Antibiotics Against Bacterial Infections. Curr Top Med Chem 2020; 20:1238-1263. [DOI: 10.2174/1568026620666200303122626] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/22/2020] [Accepted: 02/07/2020] [Indexed: 01/10/2023]
Abstract
Background:Antimicrobial resistance (AMR) to conventional antibiotics is becoming one of the main global health threats and novel alternative strategies are urging. Antimicrobial peptides (AMPs), once forgotten, are coming back into the scene as promising tools to overcome bacterial resistance. Recent findings have attracted attention to the potentiality of AMPs to work as antibiotic adjuvants.Methods:In this review, we have tried to collect the currently available information on the mechanism of action of AMPs in synergy with other antimicrobial agents. In particular, we have focused on the mechanisms of action that mediate the inhibition of the emergence of bacterial resistance by AMPs.Results and Conclusion:We find in the literature many examples where AMPs can significantly reduce the antibiotic effective concentration. Mainly, the peptides work at the bacterial cell wall and thereby facilitate the drug access to its intracellular target. Complementarily, AMPs can also contribute to permeate the exopolysaccharide layer of biofilm communities, or even prevent bacterial adhesion and biofilm growth. Secondly, we find other peptides that can directly block the emergence of bacterial resistance mechanisms or interfere with the community quorum-sensing systems. Interestingly, the effective peptide concentrations for adjuvant activity and inhibition of bacterial resistance are much lower than the required for direct antimicrobial action. Finally, many AMPs expressed by innate immune cells are endowed with immunomodulatory properties and can participate in the host response against infection. Recent studies in animal models confirm that AMPs work as adjuvants at non-toxic concentrations and can be safely administrated for novel combined chemotherapies.
Collapse
Affiliation(s)
- Jiarui Li
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| | - Pablo Fernández-Millán
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| |
Collapse
|
55
|
Parducho KR, Beadell B, Ybarra TK, Bush M, Escalera E, Trejos AT, Chieng A, Mendez M, Anderson C, Park H, Wang Y, Lu W, Porter E. The Antimicrobial Peptide Human Beta-Defensin 2 Inhibits Biofilm Production of Pseudomonas aeruginosa Without Compromising Metabolic Activity. Front Immunol 2020; 11:805. [PMID: 32457749 PMCID: PMC7225314 DOI: 10.3389/fimmu.2020.00805] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
Biofilm production is a key virulence factor that facilitates bacterial colonization on host surfaces and is regulated by complex pathways, including quorum sensing, that also control pigment production, among others. To limit colonization, epithelial cells, as part of the first line of defense, utilize a variety of antimicrobial peptides (AMPs) including defensins. Pore formation is the best investigated mechanism for the bactericidal activity of AMPs. Considering the induction of human beta-defensin 2 (HBD2) secretion to the epithelial surface in response to bacteria and the importance of biofilm in microbial infection, we hypothesized that HBD2 has biofilm inhibitory activity. We assessed the viability and biofilm formation of a pyorubin-producing Pseudomonas aeruginosa strain in the presence and absence of HBD2 in comparison to the highly bactericidal HBD3. At nanomolar concentrations, HBD2 - independent of its chiral state - significantly reduced biofilm formation but not metabolic activity, unlike HBD3, which reduced biofilm and metabolic activity to the same degree. A similar discrepancy between biofilm inhibition and maintenance of metabolic activity was also observed in HBD2 treated Acinetobacter baumannii, another Gram-negative bacterium. There was no evidence for HBD2 interference with the regulation of biofilm production. The expression of biofilm-related genes and the extracellular accumulation of pyorubin pigment, another quorum sensing controlled product, did not differ significantly between HBD2 treated and control bacteria, and in silico modeling did not support direct binding of HBD2 to quorum sensing molecules. However, alterations in the outer membrane protein profile accompanied by surface topology changes, documented by atomic force microscopy, was observed after HBD2 treatment. This suggests that HBD2 induces structural changes that interfere with the transport of biofilm precursors into the extracellular space. Taken together, these data support a novel mechanism of biofilm inhibition by nanomolar concentrations of HBD2 that is independent of biofilm regulatory pathways.
Collapse
Affiliation(s)
- Kevin R. Parducho
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Brent Beadell
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Tiffany K. Ybarra
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Mabel Bush
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Erick Escalera
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Aldo T. Trejos
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Andy Chieng
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Marlon Mendez
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Chance Anderson
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Hyunsook Park
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| | - Yixian Wang
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, CA, United States
| | - Wuyuan Lu
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Edith Porter
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
56
|
Abstract
Defensins are a major family of host defense peptides expressed predominantly in neutrophils and epithelial cells. Their broad antimicrobial activities and multifaceted immunomodulatory functions have been extensively studied, cementing their role in innate immunity as a core host-protective component against bacterial, viral and fungal infections. More recent studies, however, paint defensins in a bad light such that they are "alleged" to promote viral and bacterial infections in certain biological settings. This mini review summarizes the latest findings on the potential pathogenic properties of defensins against the backdrop of their protective roles in antiviral and antibacterial immunity. Further, a succinct description of both tumor-proliferative and -suppressive activities of defensins is also given to highlight their functional and mechanistic complexity in antitumor immunity. We posit that given an enabling environment defensins, widely heralded as the "Swiss army knife," can function as a "double-edged sword" in host immunity.
Collapse
Affiliation(s)
- Dan Xu
- Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Sciences and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Wuyuan Lu
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
57
|
Grimsey E, Collis DWP, Mikut R, Hilpert K. The effect of lipidation and glycosylation on short cationic antimicrobial peptides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183195. [PMID: 32130974 DOI: 10.1016/j.bbamem.2020.183195] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 01/16/2023]
Abstract
The global health threat surrounding bacterial resistance has resulted in antibiotic researchers shifting their focus away from 'traditional' antibiotics and concentrating on other antimicrobial agents, including antimicrobial peptides. These low molecular weight "mini-proteins" exhibit broad-spectrum activity against bacteria, including multi-drug resistant strains, viruses, fungi and protozoa and constitute a major element of the innate-immune system of many multicellular organisms. Some naturally occurring antimicrobial peptides are lipidated and/or glycosylated and almost all antimicrobial peptides in clinical use are either lipopeptides (Daptomycin and Polymyxin E and B) or glycopeptides (Vancomycin). Lipidation, glycosylation and PEGylation are an option for improving stability and activity in serum and for reducing the rapid clearing via the kidneys and liver. Two broad-spectrum antimicrobial peptides NH2-RIRIRWIIR-CONH2 (A1) and NH2-KRRVRWIIW-CONH2 (B1) were conjugated via a linker, producing A2 and B2, to individual fatty acids of C8, C10, C12 and C14 and in addition, A2 was conjugated to either glucose, N-acetyl glucosamine, galactose, mannose, lactose or polyethylene glycol (PEG). Antimicrobial activity against two Gram-positive strains (methicillin resistant Staphylococcus aureus (MRSA) and vancomycin resistant Enterococcus faecalis (VRE)) and three Gram-negative strains (Salmonella typhimurium, E. coli and Pseudomonas aeruginosa) were determined. Activity patterns for the lipidated versions are very complex, dependent on sequence, bacteria and fatty acid. Two reciprocal effects were measured; compared to the parental peptides, some combinations led to a 16-fold improvement whereas other combinations let to a 32-fold reduction in antimicrobial activity. Glycosylation decreased antimicrobial activity by 2 to 16-fold in comparison to A1, respectively on the sugar-peptide combination. PEGylation rendered the peptide inactive. Antimicrobial activity in the presence of 25% human serum of A1 and B1 was reduced 32-fold and 8-fold, respectively. The longer chain fatty acids almost completely restored this activity; however, these fatty acids increased hemolytic activity. B1 modified with C8 increased the therapeutic index by 2-fold for four bacterial strains. Our results suggest that finding the right lipid-peptide combination can lead to improved activity in the presence of serum and potentially more effective drug candidates for animal studies. Glycosylation with the optimal sugar and numbers of sugars at the right peptide position could be an alternative route or could be used in addition to lipidation to counteract solubility and toxicity issues.
Collapse
Affiliation(s)
- Elizabeth Grimsey
- Institute for Infection and Immunity, St. George's University of London, London, UK
| | | | - Ralf Mikut
- Karlsruhe Institute of Technology (KIT), Institute for Automation and Applied Informatics (IAI), Eggenstein-Leopoldshafen, Germany
| | - Kai Hilpert
- Institute for Infection and Immunity, St. George's University of London, London, UK.
| |
Collapse
|
58
|
Contreras G, Shirdel I, Braun MS, Wink M. Defensins: Transcriptional regulation and function beyond antimicrobial activity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 104:103556. [PMID: 31747541 DOI: 10.1016/j.dci.2019.103556] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 05/20/2023]
Abstract
Defensins are one the largest group of antimicrobial peptides and are part of the innate defence. Defensins are produced by animals, plants and fungi. In animals and plants, defensins can be constitutively or differentially expressed both locally or systemically which confer defence before and a stronger response after infection. Immune signalling pathways regulate the gene expression of defensins. These pathways include cellular receptors, which recognise pathogen-associated molecular patterns and are found both in plants and animals. After recognition, signalling pathways and, subsequently, transcriptional factors are activated. There is an increasing number of novel functions in defensins, such as immunomodulators and immune cell attractors. Identification of defensin triggers could help us to elucidate other new functions. The present article reviews the different elicitors of defensins with a main focus on human, fish and marine invertebrate defensins.
Collapse
Affiliation(s)
- Gabriela Contreras
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany.
| | - Iman Shirdel
- Marine Sciences Faculty, Tarbiat Modares University, Noor, Iran
| | - Markus Santhosh Braun
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
59
|
Mookherjee N, Anderson MA, Haagsman HP, Davidson DJ. Antimicrobial host defence peptides: functions and clinical potential. Nat Rev Drug Discov 2020; 19:311-332. [DOI: 10.1038/s41573-019-0058-8] [Citation(s) in RCA: 850] [Impact Index Per Article: 170.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2019] [Indexed: 12/18/2022]
|
60
|
Lin GY, Chang CF, Lan CY. The interaction Between Carbohydrates and the Antimicrobial Peptide P-113Tri is Involved in the Killing of Candida albicans. Microorganisms 2020; 8:microorganisms8020299. [PMID: 32098211 PMCID: PMC7074873 DOI: 10.3390/microorganisms8020299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/14/2022] Open
Abstract
The emergence of drug resistance to Candida albicans is problematic in the clinical setting. Therefore, developing new antifungal drugs is in high demand. Our previous work indicated that the antimicrobial peptide P-113Tri exhibited higher antifungal activity against planktonic cells, biofilm cells, and clinical isolates of Candida species compared to its parental peptide P-113. In this study, we further investigated the difference between these two peptides in their mechanisms against C. albicans. Microscopic examination showed that P-113 rapidly gained access to C. albicans cells. However, most of the P-113Tri remained on the cell surface. Moreover, using a range of cell wall-defective mutants and competition assays, the results indicated that phosphomannan and N-linked mannan in the cell wall are important for peptide binding to C. albicans cells. Furthermore, the addition of exogenous phosphosugars reduced the efficacy of the peptide, suggesting that negatively charged phosphosugars also contributed to the peptide binding to the cell wall polysaccharides. Finally, using a glycan array, P-113Tri, but not P-113, can bind to other glycans commonly present on other microbial and mammalian cells. Together, these results suggest that P-113 and P-113Tri have fundamental differences in their interaction with C. albicans and candidacidal activities.
Collapse
Affiliation(s)
- Guan-Yu Lin
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Chuan-Fa Chang
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Chung-Yu Lan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan;
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: ; Tel.: +886-3-574-2473; Fax: +886-3-571-5934
| |
Collapse
|
61
|
Chauhan J, Yu W, Cardinale S, Opperman TJ, MacKerell AD, Fletcher S, de Leeuw EP. Optimization of a Benzothiazole Indolene Scaffold Targeting Bacterial Cell Wall Assembly. Drug Des Devel Ther 2020; 14:567-574. [PMID: 32103898 PMCID: PMC7024799 DOI: 10.2147/dddt.s226313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/30/2019] [Indexed: 11/23/2022] Open
Abstract
Background The bacterial cell envelope is comprised of the cell membrane and the cell wall. The bacterial cell wall provides rigidity to the cell and protects the organism from potential harmful substances also. Cell wall biosynthesis is an important physiological process for bacterial survival and thus has been a primary target for the development of antibacterials. Antimicrobial peptides that target bacterial cell wall assembly are abundant and many bind to the essential cell wall precursor molecule Lipid II. Methods We describe the structure-to-activity (SAR) relationship of an antimicrobial peptide-derived small molecule 7771–0701 that acts as a novel agent against cell wall biosynthesis. Derivatives of compound 7771–0701 (2-[(1E)-3-[(2E)-5,6-dimethyl-3-(prop-2-en-1-yl)-1,3-benzothiazol-2-ylidene]prop-1-en-1-yl]-1,3,3-trimethylindol-1-ium) were generated by medicinal chemistry guided by Computer-Aided Drug Design and NMR. Derivatives were tested for antibacterial activity and Lipid II binding. Results Our results show that the N-alkyl moiety is subject to change without affecting functionality and further show the functional importance of the sulfur in the scaffold. The greatest potency against Gram-positive bacteria and Lipid II affinity was achieved by incorporation of a bromide at the R3 position of the benzothiazole ring. Conclusion We identify optimized small molecule benzothiazole indolene scaffolds that bind to Lipid II for further development as antibacterial therapeutics.
Collapse
Affiliation(s)
- Jay Chauhan
- Institute of Human Virology & Department of Biochemistry and Molecular Biology of the University of Maryland Baltimore School of Medicine, Baltimore, MD 21201, USA
| | - Wenbo Yu
- Computer-Aided Drug Design Center, University of Maryland, School of Pharmacy, Baltimore, MD 21201, USA
| | | | | | - Alexander D MacKerell
- Computer-Aided Drug Design Center, University of Maryland, School of Pharmacy, Baltimore, MD 21201, USA.,Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, MD 21201, USA
| | - Steven Fletcher
- Institute of Human Virology & Department of Biochemistry and Molecular Biology of the University of Maryland Baltimore School of Medicine, Baltimore, MD 21201, USA
| | - Erik Ph de Leeuw
- Institute of Human Virology & Department of Biochemistry and Molecular Biology of the University of Maryland Baltimore School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
62
|
Raheem N, Straus SK. Mechanisms of Action for Antimicrobial Peptides With Antibacterial and Antibiofilm Functions. Front Microbiol 2019; 10:2866. [PMID: 31921046 PMCID: PMC6927293 DOI: 10.3389/fmicb.2019.02866] [Citation(s) in RCA: 233] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022] Open
Abstract
The antibiotic crisis has led to a pressing need for alternatives such as antimicrobial peptides (AMPs). Recent work has shown that these molecules have great potential not only as antimicrobials, but also as antibiofilm agents, immune modulators, anti-cancer agents and anti-inflammatories. A better understanding of the mechanism of action (MOA) of AMPs is an important part of the discovery of more potent and less toxic AMPs. Many models and techniques have been utilized to describe the MOA. This review will examine how biological assays and biophysical methods can be utilized in the context of the specific antibacterial and antibiofilm functions of AMPs.
Collapse
Affiliation(s)
- Nigare Raheem
- Department of Chemistry, The University of British Columbia, Vancouver, BC, Canada
| | - Suzana K Straus
- Department of Chemistry, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
63
|
Ruden S, Rieder A, Chis Ster I, Schwartz T, Mikut R, Hilpert K. Synergy Pattern of Short Cationic Antimicrobial Peptides Against Multidrug-Resistant Pseudomonas aeruginosa. Front Microbiol 2019; 10:2740. [PMID: 31849888 PMCID: PMC6901909 DOI: 10.3389/fmicb.2019.02740] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 11/11/2019] [Indexed: 12/18/2022] Open
Abstract
With the rise of various multidrug-resistant (MDR) pathogenic bacteria, worldwide health care is under pressure to respond. Conventional antibiotics are failing and the development of novel classes and alternative strategies is a major priority. Antimicrobial peptides (AMPs) cannot only kill MDR bacteria, but also can be used synergistically with conventional antibiotics. We selected 30 short AMPs from different origins and measured their synergy in combination with polymyxin B, piperacillin, ceftazidime, cefepime, meropenem, imipenem, tetracycline, erythromycin, kanamycin, tobramycin, amikacin, gentamycin, and ciprofloxacin. In total, 403 unique combinations were tested against an MDR Pseudomonas aeruginosa isolate (PA910). As a measure of the synergistic effects, fractional inhibitory concentrations (FICs) were determined using microdilution assays with FICs ranges between 0.25 and 2. A high number of combinations between peptides and polymyxin B, erythromycin, and tetracycline were found to be synergistic. Novel variants of indolicidin also showed a high frequency in synergist interaction. Single amino acid substitutions within the peptides can have a very strong effect on the ability to synergize, making it possible to optimize future drugs toward synergistic interaction.
Collapse
Affiliation(s)
- Serge Ruden
- Institute of Biological Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Annika Rieder
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Irina Chis Ster
- Institute of Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Thomas Schwartz
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Kai Hilpert
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Infection and Immunity, St George's, University of London, London, United Kingdom.,Institute of Microstructure Technology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
64
|
Song J, Wang J, Zhan N, Sun T, Yu W, Zhang L, Shan A, Zhang A. Therapeutic Potential of Trp-Rich Engineered Amphiphiles by Single Hydrophobic Amino Acid End-Tagging. ACS APPLIED MATERIALS & INTERFACES 2019; 11:43820-43834. [PMID: 31687796 DOI: 10.1021/acsami.9b12706] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
End-tagging with a single hydrophobic residue contributes to improve the cell selectivity of antimicrobial peptides (AMPs), but systematic studies have been lacking. Thus, this study aimed to systematically investigate how end-tagging with hydrophobic residues at the C-terminus and Gly capped at the N-terminus of W4 (RWRWWWRWR) affects the bioactivity of W4 variants. Among all the hydrophobic residues, only Ala end-tagging improved the antibacterial activity of W4. Meanwhile, Gly capped at the N-terminus could promote the helical propensity of the end-tagged peptides in dodecylphosphocholine micelles, increasing their antimicrobial activities. Of these peptides, GW4A (GRWRWWWRWRA) showed the best antibacterial activity against the 19 species of bacteria tested (GMMIC = 1.86 μM) with low toxicity, thus possessing the highest cell selectivity (TIall = 137.63). It also had rapid sterilization, good salt and serum resistance, and LPS-neutralizing activity. Antibacterial mechanism studies showed that the short peptide GW4A killed bacteria by destroying cell membrane integrity and causing cytoplasmic leakage. Overall, these findings suggested that systematic studies on terminal modifications promoted the development of peptide design theory and provided a potential method for optimization of effective AMPs.
Collapse
Affiliation(s)
- Jing Song
- Institute of Animal Nutrition , Northeast Agricultural University , Harbin 150030 , Heilongjiang , P. R. China
| | - Jiajun Wang
- Institute of Animal Nutrition , Northeast Agricultural University , Harbin 150030 , Heilongjiang , P. R. China
| | - Na Zhan
- Institute of Animal Nutrition , Northeast Agricultural University , Harbin 150030 , Heilongjiang , P. R. China
| | - Taotao Sun
- Institute of Animal Nutrition , Northeast Agricultural University , Harbin 150030 , Heilongjiang , P. R. China
| | - Weikang Yu
- Institute of Animal Nutrition , Northeast Agricultural University , Harbin 150030 , Heilongjiang , P. R. China
| | - Licong Zhang
- Institute of Animal Nutrition , Northeast Agricultural University , Harbin 150030 , Heilongjiang , P. R. China
| | - Anshan Shan
- Institute of Animal Nutrition , Northeast Agricultural University , Harbin 150030 , Heilongjiang , P. R. China
| | - Aizhong Zhang
- College of Animal Science and Veterinary Medicine , Bayi Agricultural University , Daqing 163000 , Heilongjiang , P. R. China
| |
Collapse
|
65
|
Franco AR, Fernandes EM, Rodrigues MT, Rodrigues FJ, Gomes ME, Leonor IB, Kaplan DL, Reis RL. Antimicrobial coating of spider silk to prevent bacterial attachment on silk surgical sutures. Acta Biomater 2019; 99:236-246. [PMID: 31505301 DOI: 10.1016/j.actbio.2019.09.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 02/08/2023]
Abstract
Microbial infections from post-surgery or other medical-related procedure is a serious health problem. Nowadays, the research is focused on the development of new drug-free materials with antibacterial properties to prevent or minimize the risk of infections. Spider silk is known for its unique biomechanical properties allied with biocompatibility. Recombinant DNA technology allows to bioengineering spider silk with antimicrobial peptides (AMP). Thus, our goal was to bioengineered spider silk proteins with AMP (6mer-HNP1) as an antibacterial drug-free coating for commercial silk sutures (Perma-Hand®) for decreasing bacterial infections. Perma-Hand® sutures were coated with 6mer-HNP1 by dip coating. In vitro tests, using human fetal lung fibroblasts (MRC5), showed that coated sutures sustained cell viability, and also, the contact with red blood cells (RBCs) demonstrate blood compatibility. Also, the coatings inhibited significantly the adherence and formation of biofilm, where sutures coated with 6mer-HNP1 produced a 1.5 log reduction of Methicillin-Resistant Staphylococcus aureus (MRSA) and a 2 log reduction of Escherichia coli (E. coli) compared to the uncoated Perma-Hand® suture. The mechanical properties of Perma-Hand® sutures were not affected by the presence of bioengineered spider silk proteins. Thus, the present work demonstrated that using spider silk drug-free coatings it is possible to improve the antibacterial properties of the commercial sutures. Furthermore, a new class of drug-free sutures for reducing post-implantation infections can be developed. STATEMENT OF SIGNIFICANCE: Microbial infections from post-surgery or other medical-related procedure is a serious health problem. Developing new drug-free materials with antibacterial properties is an approach to prevent or minimize the risk of infections. Spider silk is known for its unique biomechanical properties allied with biocompatibility. Recombinant DNA technology allow to bioengineering spider silk with antimicrobial peptides (AMP). Our goal is bioengineered spider silk proteins with AMP as an antibacterial coating for silk sutures. The coatings showed exceptional antibacterial properties and maintained intrinsic mechanical features. In vitro studies showed a positive effect of the coated sutures on the cell behavior. With this new drug-free bioengineered spider silk coating is possible to develop a new class of drug-free sutures for reducing post-implantation infections.
Collapse
|
66
|
Chen H, Guo A, Lu Z, Tan S, Wang J, Gao J, Zhang S, Huang X, Zheng J, Xi J, Yi K. Agrobacterium tumefaciens-mediated transformation of a hevein-like gene into asparagus leads to stem wilt resistance. PLoS One 2019; 14:e0223331. [PMID: 31589638 PMCID: PMC6779260 DOI: 10.1371/journal.pone.0223331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/18/2019] [Indexed: 11/18/2022] Open
Abstract
Asparagus stem wilt, is a significant and devastating disease, typically leading to extensive economic losses in the asparagus industry. To obtain transgenic plants resistant to stem wilt, the hevein-like gene, providing broad spectrum bacterial resistance was inserted into the asparagus genome through Agrobacterium tumefaciens-mediated transformation. The optimal genetic transformation system for asparagus was as follows: pre-culture of embryos for 2 days, inoculation using a bacterial titre of OD600 = 0.6, infection time 10 min and co-culturing for 4 days using an Acetosyringone concentration of 200 μmol/L. Highest transformation frequencies reached 21% and ten transgenic asparagus seedlings carrying the hevein-like gene were identified by polymerase chain reaction. Moreover, integration of the hevein-like gene in the T1 generation of transgenic plants was confirmed by southern blot hybridization. Analysis showed that resistance to stem wilt was enhanced significantly in the transgenic plants, in comparison to non- transgenic plants. The results provide additional data for genetic improvement and are of importance for the development of new disease-resistant asparagus varieties.
Collapse
Affiliation(s)
- Helong Chen
- Institute of Tropical Agriculture and Forestry, Hainan University, Haikou, China
- Institute of Tropical Bioscience and Biotechnology/Hainan Academy of Tropical Agricultural Resource, Chinese Academy of Tropical Agricultural Sciences /Key Laboratory of Biology and Genetic Resources of Tropical Crops, Ministry of Agriculture, Haikou, China
- * E-mail: (CHL); (YKX)
| | - Anping Guo
- Institute of Tropical Agriculture and Forestry, Hainan University, Haikou, China
- Institute of Tropical Bioscience and Biotechnology/Hainan Academy of Tropical Agricultural Resource, Chinese Academy of Tropical Agricultural Sciences /Key Laboratory of Biology and Genetic Resources of Tropical Crops, Ministry of Agriculture, Haikou, China
| | - Zhiwei Lu
- South Subtropical Crops Institute, Chinese Academy of Tropical Agricultural Sciences/ Zhanjiang City Key Laboratory for Tropical Crops Genetic Improvement, Zhanjiang, Guangdong, China
| | - Shibei Tan
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, Hainan, China
| | - Jian Wang
- Institute of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Jianming Gao
- Institute of Tropical Bioscience and Biotechnology/Hainan Academy of Tropical Agricultural Resource, Chinese Academy of Tropical Agricultural Sciences /Key Laboratory of Biology and Genetic Resources of Tropical Crops, Ministry of Agriculture, Haikou, China
| | - Shiqing Zhang
- Institute of Tropical Bioscience and Biotechnology/Hainan Academy of Tropical Agricultural Resource, Chinese Academy of Tropical Agricultural Sciences /Key Laboratory of Biology and Genetic Resources of Tropical Crops, Ministry of Agriculture, Haikou, China
| | - Xing Huang
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, Hainan, China
| | - Jinlong Zheng
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, Hainan, China
| | - Jingen Xi
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, Hainan, China
| | - Kexian Yi
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, Hainan, China
- * E-mail: (CHL); (YKX)
| |
Collapse
|
67
|
Pinto IB, dos Santos Machado L, Meneguetti BT, Nogueira ML, Espínola Carvalho CM, Roel AR, Franco OL. Utilization of antimicrobial peptides, analogues and mimics in creating antimicrobial surfaces and bio-materials. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.107237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
68
|
Non-Lytic Antibacterial Peptides That Translocate Through Bacterial Membranes to Act on Intracellular Targets. Int J Mol Sci 2019; 20:ijms20194877. [PMID: 31581426 PMCID: PMC6801614 DOI: 10.3390/ijms20194877] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/12/2019] [Accepted: 09/14/2019] [Indexed: 11/28/2022] Open
Abstract
The advent of multidrug resistance among pathogenic bacteria has attracted great attention worldwide. As a response to this growing challenge, diverse studies have focused on the development of novel anti-infective therapies, including antimicrobial peptides (AMPs). The biological properties of this class of antimicrobials have been thoroughly investigated, and membranolytic activities are the most reported mechanisms by which AMPs kill bacteria. Nevertheless, an increasing number of works have pointed to a different direction, in which AMPs are seen to be capable of displaying non-lytic modes of action by internalizing bacterial cells. In this context, this review focused on the description of the in vitro and in vivo antibacterial and antibiofilm activities of non-lytic AMPs, including indolicidin, buforin II PR-39, bactenecins, apidaecin, and drosocin, also shedding light on how AMPs interact with and further translocate through bacterial membranes to act on intracellular targets, including DNA, RNA, cell wall and protein synthesis.
Collapse
|
69
|
Malin JJ, de Leeuw E. Therapeutic compounds targeting Lipid II for antibacterial purposes. Infect Drug Resist 2019; 12:2613-2625. [PMID: 31692545 PMCID: PMC6711568 DOI: 10.2147/idr.s215070] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022] Open
Abstract
Resistance against commonly used antibiotics has emerged in all bacterial pathogens. In fact, there is no antibiotic currently in clinical use against which resistance has not been reported. In particular, rapidly increasing urbanization in developing nations are sites of major concern. Additionally, the widespread practice by physicians to prescribe antibiotics in cases of viral infections puts selective pressure on antibiotics that still remain effective and it will only be a matter of time before resistance develops on a large scale. The biosynthesis pathway of the bacterial cell wall is well studied and a validated target for the development of antibacterial agents. Cell wall biosynthesis involves two major processes; 1) the biosynthesis of cell wall teichoic acids and 2) the biosynthesis of peptidoglycan. Key molecules in these pathways, including enzymes and precursor molecules are attractive targets for the development of novel antibacterial agents. In this review, we will focus on the major class of natural antibacterial compounds that target the peptidoglycan precursor molecule Lipid II; namely the glycopeptides, including the novel generation of lipoglycopeptides. We will discuss their mechanism-of-action and clinical applications. Further, we will briefly discuss additional peptides that target Lipid II such as the lantibiotic nisin and defensins. We will highlight recent developments and future perspectives.
Collapse
Affiliation(s)
- Jakob J Malin
- University of Cologne, Department I of Internal Medicine, Division of Infectious Diseases, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Erik de Leeuw
- Institute of Human Virology and Department of Molecular Biology & Biochemistry of the University of Maryland, Baltimore School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
70
|
Bolatchiev A, Baturin V, Bazikov I, Maltsev A, Kunitsina E. Effect of antimicrobial peptides HNP-1 and hBD-1 on Staphylococcus aureus strains in vitro and in vivo. Fundam Clin Pharmacol 2019; 34:102-108. [PMID: 31313350 DOI: 10.1111/fcp.12499] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022]
Abstract
The aims of this study were: (i) To investigate the activity of recombinant AMPs HNP-1 and hBD-1 in combination with cefotaxime against Staphylococcus aureus strains (MSSA and MRSA) in vitro using checkerboard method; (ii) To investigate the activity of HNP-1 and hBD-1 encapsulated in silicon nanoparticles (niosomes) in the treatment of MRSA-infected wound in rats. For this S. aureus strains (MSSA and MRSA) were isolated from patients with diabetic foot infection. Cefotaxime, recombinant HNP-1 and hBD-1 (in all possible combinations with each other) were used for testing by the checkerboard method. Two niosomal topical gels with HNP-1/hBD-1 were prepared to treat MRSA-infected wounds in rats. Gels were administered once a day, the control group-without treatment. Wound healing rate was calculated on the 4th, 9th and 16th days of the experiment and compared using one-way ANOVA with Bonferroni correction. MIC of HNP-1 for MSSA and MRSA was the same-1 mg/L. MIC of hBD-1 for MSSA and MRSA was also the same-0.5 mg/L. Topical gels with niosomal HNP-1 (or hBD-1) showed a significantly faster wound healing in comparison with the control. The data obtained open up prospects for use of AMPs encapsulated in silica nanoparticles for the development of new antibiotics.
Collapse
Affiliation(s)
- Albert Bolatchiev
- Department of Clinical Pharmacology, Stavropol State Medical University, Stavropol, Russia
| | - Vladimir Baturin
- Department of Clinical Pharmacology, Stavropol State Medical University, Stavropol, Russia
| | - Igor Bazikov
- Department of Microbiology, Stavropol State Medical University, Stavropol, Russia
| | - Alexander Maltsev
- Department of Microbiology, Stavropol State Medical University, Stavropol, Russia
| | - Elena Kunitsina
- Department of Clinical Microbiology, The Center of Clinical Pharmacology and Pharmacotherapy, Stavropol, Russia
| |
Collapse
|
71
|
Koehbach J, Craik DJ. The Vast Structural Diversity of Antimicrobial Peptides. Trends Pharmacol Sci 2019; 40:517-528. [DOI: 10.1016/j.tips.2019.04.012] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 01/08/2023]
|
72
|
Chauhan J, Kwasny SM, Fletcher S, Opperman TJ, de Leeuw EPH. Optimization of a small-molecule Lipid II binder. Bioorg Med Chem Lett 2019; 29:1849-1853. [PMID: 31126852 DOI: 10.1016/j.bmcl.2019.04.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 11/16/2022]
Abstract
Lipid II is an essential precursor of bacterial cell wall biosynthesis and an attractive target for antibiotics. Lipid II is comprised of specialized lipid (bactoprenol) linked to a hydrophilic head group consisting of a peptidoglycan subunit (N-acetylglucosamine (GlcNAc)-N-acetylmuramic acid (MurNAc) disaccharide coupled to a short pentapeptide moiety) via a pyrophosphate. We previously identified a (E)-2,4-bis(4-bromophenyl)-6-(4-(dimethylamino)styryl)pyrylium boron tetrafluoride salt, termed 6jc48-1, that interacts with the MurNAc moiety, the phosphate cage and the isoprenyl tail of Lipid II. Here, we report on the structure-activity relationship of 6jc48-1 derivatives obtained by de novo chemical synthesis. Our results indicate that bacterial killing is positively driven by bi-phenyl stacking with peptidoglycan units. Replacement of bromides by fluorides resulted in activity against S. aureus without affecting Lipid II binding and cytotoxicity. Antibacterial activity was affected negatively by extended interaction of the scaffold with Lipid II isoprenyl units.
Collapse
Affiliation(s)
- Jay Chauhan
- Institute of Human Virology & Department of Biochemistry and Molecular Biology of the University of Maryland Baltimore School of Medicine, 725 West Lombard Street, Baltimore, MD 21201, USA
| | - Steven M Kwasny
- Microbiotix, Inc., One Innovation Drive, Worcester, MA 01605, USA
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, 20 Penn Street, Baltimore, MD 21201, USA
| | | | - Erik P H de Leeuw
- Institute of Human Virology & Department of Biochemistry and Molecular Biology of the University of Maryland Baltimore School of Medicine, 725 West Lombard Street, Baltimore, MD 21201, USA.
| |
Collapse
|
73
|
Grein F, Schneider T, Sahl HG. Docking on Lipid II-A Widespread Mechanism for Potent Bactericidal Activities of Antibiotic Peptides. J Mol Biol 2019; 431:3520-3530. [PMID: 31100388 DOI: 10.1016/j.jmb.2019.05.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/30/2019] [Accepted: 05/08/2019] [Indexed: 12/21/2022]
Abstract
Natural product antibiotics usually target the major biosynthetic pathways of bacterial cells and the search for new targets outside these pathways has proven very difficult. Cell wall biosynthesis maybe the most prominent antibiotic target, and ß-lactams are among the clinically most relevant antibiotics. Among cell wall biosynthesis inhibitors, glycopeptide antibiotics are a second group of important drugs, which bind to the peptidoglycan building block lipid II and prevent the incorporation of the monomeric unit into polymeric cell wall. However, lipid II acts as a docking molecule for many more naturally occurring antibiotics from diverse chemical classes and likely is the most targeted molecule in antibacterial mechanisms. We summarize current knowledge on lipid II binding antibiotics and explain, on the levels of mechanisms and resistance development, why lipid II is such a prominent target, and thus provide insights for the design of new antibiotic drugs.
Collapse
Affiliation(s)
- Fabian Grein
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany.
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, Germany; German Center for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Hans-Georg Sahl
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, Germany
| |
Collapse
|
74
|
Zharkova MS, Orlov DS, Golubeva OY, Chakchir OB, Eliseev IE, Grinchuk TM, Shamova OV. Application of Antimicrobial Peptides of the Innate Immune System in Combination With Conventional Antibiotics-A Novel Way to Combat Antibiotic Resistance? Front Cell Infect Microbiol 2019; 9:128. [PMID: 31114762 PMCID: PMC6503114 DOI: 10.3389/fcimb.2019.00128] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 04/10/2019] [Indexed: 01/10/2023] Open
Abstract
Rapidly growing resistance of pathogenic bacteria to conventional antibiotics leads to inefficiency of traditional approaches of countering infections and determines the urgent need for a search of fundamentally new anti-infective drugs. Antimicrobial peptides (AMPs) of the innate immune system are promising candidates for a role of such novel antibiotics. However, some cytotoxicity of AMPs toward host cells limits their active implementation in medicine and forces attempts to design numerous structural analogs of the peptides with optimized properties. An alternative route for the successful AMPs introduction may be their usage in combination with conventional antibiotics. Synergistic antibacterial effects have been reported for a number of such combinations, however, the molecular mechanisms of the synergy remain poorly understood and little is known whether AMPs cytotoxicy for the host cells increases upon their application with antibiotics. Our study is directed to examination of a combined action of natural AMPs with different structure and mode of action (porcine protegrin 1, caprine bactenecin ChBac3.4, human alpha- and beta-defensins (HNP-1, HNP-4, hBD-2, hBD-3), human cathelicidin LL-37), and egg white lysozyme with varied antibiotic agents (gentamicin, ofloxacin, oxacillin, rifampicin, polymyxin B, silver nanoparticles) toward selected bacteria, including drug-sensitive and drug-resistant strains, as well as toward some mammalian cells (human erythrocytes, PBMC, neutrophils, murine peritoneal macrophages and Ehrlich ascites carcinoma cells). Using “checkerboard titrations” for fractional inhibitory concentration indexes evaluation, it was found that synergy in antibacterial action mainly occurs between highly membrane-active AMPs (e.g., protegrin 1, hBD-3) and antibiotics with intracellular targets (e.g., gentamicin, rifampcin), suggesting bioavailability increase as the main model of such interaction. In some combinations modulation of dynamics of AMP-bacterial membrane interaction in presence of the antibiotic was also shown. Cytotoxic effects of the same combinations toward normal eukaryotic cells were rarely synergistic. The obtained data approve that combined application of antimicrobial peptides with antibiotics or other antimicrobials is a promising strategy for further development of new approach for combating antibiotic-resistant bacteria by usage of AMP-based therapeutics. Revealing the conventional antibiotics that increase the activity of human endogenous AMPs against particular pathogens is also important for cure strategies elaboration.
Collapse
Affiliation(s)
- Maria S Zharkova
- Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, FSBSI Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Dmitriy S Orlov
- Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, FSBSI Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Olga Yu Golubeva
- Laboratory of Nanostructures Research, Institute of Silicate Chemistry of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Oleg B Chakchir
- Nanobiotechnology Laboratory, Saint Petersburg National Research Academic University of the Russian Academy of Science, Saint Petersburg, Russia
| | - Igor E Eliseev
- Nanobiotechnology Laboratory, Saint Petersburg National Research Academic University of the Russian Academy of Science, Saint Petersburg, Russia
| | - Tatyana M Grinchuk
- Laboratory of Intracellular Signaling, Institute of Cytology of the Russian Academy of Science, Saint Petersburg, Russia
| | - Olga V Shamova
- Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, FSBSI Institute of Experimental Medicine, Saint Petersburg, Russia
| |
Collapse
|
75
|
Chung LK, Raffatellu M. G.I. pros: Antimicrobial defense in the gastrointestinal tract. Semin Cell Dev Biol 2019; 88:129-137. [PMID: 29432952 PMCID: PMC6087682 DOI: 10.1016/j.semcdb.2018.02.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 01/11/2023]
Abstract
The gastrointestinal tract is a complex environment in which the host immune system interacts with a diverse array of microorganisms, both symbiotic and pathogenic. As such, mobilizing a rapid and appropriate antimicrobial response depending on the nature of each stimulus is crucial for maintaining the balance between homeostasis and inflammation in the gut. Here we focus on the mechanisms by which intestinal antimicrobial peptides regulate microbial communities during dysbiosis and infection. We also discuss classes of bacterial peptides that contribute to reducing enteric pathogen outgrowth. This review aims to provide a comprehensive overview on the interplay of diverse antimicrobial responses with enteric pathogens and the gut microbiota.
Collapse
Affiliation(s)
- Lawton K Chung
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, La Jolla, CA, 92093-0704, United States
| | - Manuela Raffatellu
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, La Jolla, CA, 92093-0704, United States; Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), La Jolla CA, United States.
| |
Collapse
|
76
|
Hu H, Di B, Tolbert WD, Gohain N, Yuan W, Gao P, Ma B, He Q, Pazgier M, Zhao L, Lu W. Systematic mutational analysis of human neutrophil α-defensin HNP4. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:835-844. [PMID: 30658057 DOI: 10.1016/j.bbamem.2019.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 12/19/2018] [Accepted: 01/13/2019] [Indexed: 12/28/2022]
Abstract
Defensins are a family of cationic antimicrobial peptides of innate immunity with immunomodulatory properties. The prototypic human α-defensins, also known as human neutrophil peptides 1-3 or HNP1-3, are extensively studied for their structure, function and mechanisms of action, yet little is known about HNP4 - the much less abundant "distant cousin" of HNP1-3. Here we report a systematic mutational analysis of HNP4 with respect to its antibacterial activity against E. coli and S. aureus, inhibitory activity against anthrax lethal factor (LF), and binding activity for LF and HIV-1 gp120. Except for nine conserved and structurally important residues (6xCys, 1xArg, 1xGlu and 1xGly), the remaining 24 residues of HNP4 were each individually mutated to Ala. The crystal structures of G23A-HNP4 and T27A-HNP4 were determined, both exhibiting a disulfide-stabilized canonical α-defensin dimer identical to wild-type HNP4. Unlike HNP1-3, HNP4 preferentially killed the Gram-negative bacterium, a property largely attributable to three clustered cationic residues Arg10, Arg11 and Arg15. The cationic cluster was also important for HNP4 killing of S. aureus, inhibition of LF and binding to LF and gp120. However, F26A, while functionally inconsequential for E. coli killing, was far more deleterious than any other mutations. Similarly, N-methylation of Leu20 to destabilize the HNP4 dimer had little effect on E. coli killing, but significantly reduced the ability of HNP4 to kill S. aureus, inhibit LF, and bind to LF and gp120. Our findings unveil the molecular determinants of HNP4 function, completing the atlas of structure and function relationships for all human neutrophil α-defensins.
Collapse
Affiliation(s)
- Han Hu
- Key Laboratory of Fermentation Engineering, Ministry of Education, College of Bioengineering, Hubei University of Technology, Wuhan, China; Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA
| | - Bin Di
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA
| | - William D Tolbert
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA
| | - Neelakshi Gohain
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA
| | - Weirong Yuan
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA
| | - Pan Gao
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA
| | - Bohan Ma
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Marzena Pazgier
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA.
| | - Le Zhao
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Wuyuan Lu
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
77
|
Carbohydrate-Dependent and Antimicrobial Peptide Defence Mechanisms Against Helicobacter pylori Infections. Curr Top Microbiol Immunol 2019; 421:179-207. [PMID: 31123890 DOI: 10.1007/978-3-030-15138-6_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The human stomach is a harsh and fluctuating environment for bacteria with hazards such as gastric acid and flow through of gastric contents into the intestine. H. pylori gains admission to a stable niche with nutrient access from exudates when attached to the epithelial cells under the mucus layer, whereof adherence to glycolipids and other factors provides stable and intimate attachment. To reach this niche, H. pylori must overcome mucosal defence mechanisms including the continuously secreted mucus layer, which provides several layers of defence: (1) mucins in the mucus layer can bind H. pylori and transport it away from the gastric niche with the gastric emptying, (2) mucins can inhibit H. pylori growth, both via glycans that can have antibiotic like function and via an aggregation-dependent mechanism, (3) antimicrobial peptides (AMPs) have antimicrobial activity and are retained in a strategic position in the mucus layer and (4) underneath the mucus layer, the membrane-bound mucins provide a second barrier, and can function as releasable decoys. Many of these functions are dependent on H. pylori interactions with host glycan structures, and both the host glycosylation and concentration of antimicrobial peptides change with infection and inflammation, making these interactions dynamic. Here, we review our current understanding of mucin glycan and antimicrobial peptide-dependent host defence mechanisms against H. pylori infection.
Collapse
|
78
|
Leeuw EPHD, Lee SH, Kim WH, Kwasny SM, Opperman TJ, Lillehoj HS. Pleiotropic Anti-Infective Effects of Defensin-Derived Antimicrobial Compounds. Avian Dis 2018; 62:381-387. [PMID: 31119922 DOI: 10.1637/11912-061118-reg.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 11/05/2022]
Abstract
We identified low-molecular weight compounds derived from the antimicrobial peptide human neutrophil peptide-1 that bind to Lipid II, an essential precursor of bacterial cell wall biosynthesis. These compounds act as antibacterials on multiple biosynthesis pathways with specificity against gram-positive organisms. Here, we have tested a small subset of our most promising leads against the bacterium Clostridium perfringens and sporozoites of Eimeria tenella, an intracellular protozoan parasite that causes intestinal disease in poultry. We found one compound, 1611-0203 (2-{2,3,5,6-tetrafluoro-4-[2,3,5,6-tetrafluoro-4-(2-hydroxyphenoxy)phenyl]phenoxy}phenol), specifically to inhibit growth of both agents out of all compounds tested. Additionally, compound 1611-0203 inhibits Staphylococcus aureus and Enterococcus spp. Mechanism-of-action studies further reveal that 1611-0203 affects cell wall biosynthesis and inhibits additional biosynthetic pathways. Combined, our results indicate that compounds such as 1611-0203 have therapeutic potential to act as anti-infectives against various organisms simultaneously.
Collapse
Affiliation(s)
- Erik P H de Leeuw
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore School of Medicine, Baltimore, MD 21201,
| | - Sung Hyen Lee
- United States Department of Agriculture, Animal Biosciences and Biotechnology Laboratory, Beltsville, MD 20705
| | - Woo H Kim
- United States Department of Agriculture, Animal Biosciences and Biotechnology Laboratory, Beltsville, MD 20705
| | | | | | - Hyun S Lillehoj
- United States Department of Agriculture, Animal Biosciences and Biotechnology Laboratory, Beltsville, MD 20705,
| |
Collapse
|
79
|
Franco AR, Palma Kimmerling E, Silva C, Rodrigues FJ, Leonor IB, Reis RL, Kaplan DL. Silk‐Based Antimicrobial Polymers as a New Platform to Design Drug‐Free Materials to Impede Microbial Infections. Macromol Biosci 2018; 18:e1800262. [DOI: 10.1002/mabi.201800262] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/03/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Albina R. Franco
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory 4805 Braga/Guimarães Portugal
| | | | - Carla Silva
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory 4805 Braga/Guimarães Portugal
| | - Fernando J. Rodrigues
- ICVS/3B's – PT Government Associate Laboratory 4805 Braga/Guimarães Portugal
- Life and Health Sciences Research Institute, School of Health SciencesUniversity of Minho 4805 Braga Portugal
| | - Isabel B. Leonor
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory 4805 Braga/Guimarães Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory 4805 Braga/Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of Minho Avepark 4805‐017 Barco Guimarães Portugal
| | - David L. Kaplan
- Department of Biomedical EngineeringTufts University Medford MA 02155 USA
| |
Collapse
|
80
|
Did cis- and trans-defensins derive from a common ancestor? Immunogenetics 2018; 71:61-69. [DOI: 10.1007/s00251-018-1086-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/21/2018] [Indexed: 02/06/2023]
|
81
|
Shen W, He P, Xiao C, Chen X. From Antimicrobial Peptides to Antimicrobial Poly(α-amino acid)s. Adv Healthc Mater 2018; 7:e1800354. [PMID: 29923332 DOI: 10.1002/adhm.201800354] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/22/2018] [Indexed: 01/17/2023]
Abstract
Conventional small-molecule antibiotics are facing a significant challenge of the rapidly developed drug resistance of pathogens. In contrast, antimicrobial peptides (AMPs), an important component for innate host defenses, are now under intensive investigation as a promising antimicrobial agent for combating drug resistant pathogens. Most AMPs can effectively kill a broad spectrum of pathogens via physical disruption of microbial cellular membranes, which is identified to be difficult to develop resistance. However, the clinical applications of AMPs are still greatly limited by several inherent impediments, such as high cost of production, potential hemolysis or toxicity, and liability to proteinase degradation. Recently, cationic poly(α-amino acid)s with structures mimicking the AMPs are found to have excellent antimicrobial activity. These polymers, termed "antimicrobial poly(α-amino acid)s (APAAs)," have some advantages over AMPs, such as easy production and modification, prolonged antimicrobial activity, low cytotoxicity, and enhanced stability to protease degradation. Here, a brief introduction of mechanisms and affecting factors of microbial killing by AMPs is first presented, followed by a systematic illustration of recent advances in design and preparation of biomimetic APAAs and a perspective in this field.
Collapse
Affiliation(s)
- Wei Shen
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
- University of Chinese Academy of Sciences; 19A Yuquan Road Beijing 100049 P. R. China
| | - Pan He
- School of Materials Science and Engineering; Changchun University of Science and Technology; Changchun 130022 P. R. China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| |
Collapse
|
82
|
Zheng J, Huang Y, Islam D, Wen XY, Wu S, Streutker C, Luo A, Li M, Khang J, Han B, Zhong N, Li Y, Yu K, Zhang H. Dual effects of human neutrophil peptides in a mouse model of pneumonia and ventilator-induced lung injury. Respir Res 2018; 19:190. [PMID: 30268129 PMCID: PMC6162902 DOI: 10.1186/s12931-018-0869-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Pneumonia is a major cause of high morbidity and mortality in critically illness, and frequently requires support with mechanical ventilation. The latter can lead to ventilator-induced lung injury characterized by neutrophil infiltration. The cationic human neutrophil peptides (HNP) stored in neutrophils can kill microorganisms, but excessive amount of HNP released during phagocytosis may contribute to inflammatory responses and worsen lung injury. Based on our previous work, we hypothesized that blocking the cell surface purinergic receptor P2Y6 will attenuate the HNP-induced inflammatory responses while maintaining their antimicrobial activity in pneumonia followed by mechanical ventilation. METHODS Plasma HNP levels were measured in patients with pneumonia who received mechanical ventilation and in healthy volunteers. FVB littermate control and HNP transgenic (HNP+) mice were randomized to receive P. aeruginosa intranasally. The P2Y6 antagonist (MRS2578) or vehicle control was given after P. aeruginosa instillation. Additional mice underwent mechanical ventilation at either low pressure (LP) or high pressure (HP) ventilation 48 h after pneumonia, and were observed for 24 h. RESULTS Plasma HNP concentration increased in patients with pneumonia as compared to healthy subjects. The bacterial counts in the bronchoalveolar lavage fluid (BALF) were lower in HNP+ mice than in FVB mice 72 h after P. aeruginosa instillation. However, upon receiving HP ventilation, HNP+ mice had higher levels of cytokines and chemokines in BALF than FVB mice. These inflammatory responses were attenuated by the treatment with MRS2578 that did not affect the microbial effects of HNP. CONCLUSIONS HNP exerted dual effects by exhibiting antimicrobial activity in pneumonia alone condition while enhancing inflammatory responses in pneumonia followed by HP mechanical ventilation. Blocking P2Y6 can attenuate the inflammation without affecting the antibacterial property of HNP. The P2Y6 receptor may be a novel therapeutic target in attenuation of the leukocyte-mediated excessive host responses in inflammatory lung diseases.
Collapse
Affiliation(s)
- Junbo Zheng
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Yongbo Huang
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
| | - Diana Islam
- Keenan Research Center for Biomedical Science of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Xiao-Yan Wen
- Keenan Research Center for Biomedical Science of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Sulong Wu
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
| | - Catherine Streutker
- Keenan Research Center for Biomedical Science of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Alice Luo
- Keenan Research Center for Biomedical Science of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Manshu Li
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China.,Keenan Research Center for Biomedical Science of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Julie Khang
- Keenan Research Center for Biomedical Science of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Bing Han
- Keenan Research Center for Biomedical Science of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Nanshan Zhong
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
| | - Yimin Li
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China.
| | - Kaijiang Yu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, China.
| | - Haibo Zhang
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China. .,Keenan Research Center for Biomedical Science of St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada. .,Interdepartmental Division of Critical Care Medicine, Departments of Anesthesia and Physiology, University of Toronto, Toronto, ON, M5B 1T8, Canada.
| |
Collapse
|
83
|
Sultan I, Rahman S, Jan AT, Siddiqui MT, Mondal AH, Haq QMR. Antibiotics, Resistome and Resistance Mechanisms: A Bacterial Perspective. Front Microbiol 2018; 9:2066. [PMID: 30298054 PMCID: PMC6160567 DOI: 10.3389/fmicb.2018.02066] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/13/2018] [Indexed: 12/28/2022] Open
Abstract
History of mankind is regarded as struggle against infectious diseases. Rather than observing the withering away of bacterial diseases, antibiotic resistance has emerged as a serious global health concern. Medium of antibiotic resistance in bacteria varies greatly and comprises of target protection, target substitution, antibiotic detoxification and block of intracellular antibiotic accumulation. Further aggravation to prevailing situation arose on observing bacteria gradually becoming resistant to different classes of antibiotics through acquisition of resistance genes from same and different genera of bacteria. Attributing bacteria with feature of better adaptability, dispersal of antibiotic resistance genes to minimize effects of antibiotics by various means including horizontal gene transfer (conjugation, transformation, and transduction), Mobile genetic elements (plasmids, transposons, insertion sequences, integrons, and integrative-conjugative elements) and bacterial toxin-antitoxin system led to speedy bloom of antibiotic resistance amongst bacteria. Proficiency of bacteria to obtain resistance genes generated an unpleasant situation; a grave, but a lot unacknowledged, feature of resistance gene transfer.
Collapse
Affiliation(s)
- Insha Sultan
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Safikur Rahman
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | | | | | | |
Collapse
|
84
|
Lei R, Hou J, Chen Q, Yuan W, Cheng B, Sun Y, Jin Y, Ge L, Ben-Sasson SA, Chen J, Wang H, Lu W, Fang X. Self-Assembling Myristoylated Human α-Defensin 5 as a Next-Generation Nanobiotics Potentiates Therapeutic Efficacy in Bacterial Infection. ACS NANO 2018; 12:5284-5296. [PMID: 29856606 DOI: 10.1021/acsnano.7b09109] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The increasing prevalence of antibacterial resistance globally underscores the urgent need to the update of antibiotics. Here, we describe a strategy for inducing the self-assembly of a host-defense antimicrobial peptide (AMP) into nanoparticle antibiotics (termed nanobiotics) with significantly improved pharmacological properties. Our strategy involves the myristoylation of human α-defensin 5 (HD5) as a therapeutic target and subsequent self-assembly in aqueous media in the absence of exogenous excipients. Compared with its parent HD5, the C-terminally myristoylated HD5 (HD5-myr)-assembled nanobiotic exhibited significantly enhanced broad-spectrum bactericidal activity in vitro. Mechanistically, it selectively killed Escherichia coli ( E. coli) and methicillin-resistant Staphylococcus aureus (MRSA) through disruption of the cell wall and/or membrane structure. The in vivo results further demonstrated that the HD5-myr nanobiotic protected against skin infection by MRSA and rescued mice from E. coli-induced sepsis by lowering the systemic bacterial burden and alleviating organ damage. The self-assembled HD5-myr nanobiotic also showed negligible hemolytic activity and substantially low toxicity in animals. Our findings validate this design rationale as a simple yet versatile strategy for generating AMP-derived nanobiotics with excellent in vivo tolerability. This advancement will likely have a broad impact on antibiotic discovery and development efforts aimed at combating antibacterial resistance.
Collapse
Affiliation(s)
- Ruyi Lei
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310003 , China
| | - Jinchao Hou
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310003 , China
| | - Qixing Chen
- The Children's Hospital, School of Medicine , Zhejiang University , Hangzhou 310052 , China
| | - Weirong Yuan
- Institute of Human Virology and Department of Biochemistry and Molecular Biology , University of Maryland School of Medicine , Baltimore , Maryland 21201 , United States
| | - Baoli Cheng
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310003 , China
| | - Yaqi Sun
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310003 , China
| | - Yue Jin
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310003 , China
| | - Lujie Ge
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310003 , China
| | - Shmuel A Ben-Sasson
- Department of Developmental Biology, Institute for Medical Research Israel-Canada , The Hebrew University-Hadassah Medical School , Jerusalem 91120 , Israel
| | - Jiong Chen
- Laboratory of Biochemistry and Molecular Biology , Ningbo University , Ningbo 315211 , China
| | - Hangxiang Wang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310003 , China
| | - Wuyuan Lu
- Institute of Human Virology and Department of Biochemistry and Molecular Biology , University of Maryland School of Medicine , Baltimore , Maryland 21201 , United States
| | - Xiangming Fang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou 310003 , China
| |
Collapse
|
85
|
Zou G, de Leeuw E. Neutralization of Pseudomonas auruginosa Exotoxin A by human neutrophil peptide 1. Biochem Biophys Res Commun 2018; 501:454-457. [PMID: 29738776 DOI: 10.1016/j.bbrc.2018.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/02/2018] [Indexed: 01/01/2023]
Abstract
Pseudomonas aeruginosa produces a large number of virulence factors, including the extracellular protein, Exotoxin A (ETA). Human Neutrophil Peptide 1 (HNP1) neutralizes the Exotoxin A. HNP1 belongs to the family of α-defensins, small effector peptides of the innate immune system that combat against microbial infections. Neutralization of bacterial toxins such as ETA by HNP1 is a novel biological function in addition to direct killing of bacteria. In this study, we report on the interaction between HNP-1 and Exotoxin A at the molecular level to allow for the design and development of potent antibacterial peptides as alternatives to classical antibiotics.
Collapse
Affiliation(s)
- Guozhang Zou
- U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Erik de Leeuw
- Institute of Human Virology of the University of Maryland Baltimore School of Medicine, 725 West Lombard Street, Baltimore, MD 21201, USA.
| |
Collapse
|
86
|
Kumar P, Kizhakkedathu JN, Straus SK. Antimicrobial Peptides: Diversity, Mechanism of Action and Strategies to Improve the Activity and Biocompatibility In Vivo. Biomolecules 2018; 8:E4. [PMID: 29351202 PMCID: PMC5871973 DOI: 10.3390/biom8010004] [Citation(s) in RCA: 759] [Impact Index Per Article: 108.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 02/06/2023] Open
Abstract
Antibiotic resistance is projected as one of the greatest threats to human health in the future and hence alternatives are being explored to combat resistance. Antimicrobial peptides (AMPs) have shown great promise, because use of AMPs leads bacteria to develop no or low resistance. In this review, we discuss the diversity, history and the various mechanisms of action of AMPs. Although many AMPs have reached clinical trials, to date not many have been approved by the US Food and Drug Administration (FDA) due to issues with toxicity, protease cleavage and short half-life. Some of the recent strategies developed to improve the activity and biocompatibility of AMPs, such as chemical modifications and the use of delivery systems, are also reviewed in this article.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada.
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, 2350 Health Sciences Mall, Life Sciences Centre, Vancouver, BC V6T 1Z3, Canada.
| | - Jayachandran N Kizhakkedathu
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada.
- Centre for Blood Research, Department of Pathology and Laboratory Medicine, University of British Columbia, 2350 Health Sciences Mall, Life Sciences Centre, Vancouver, BC V6T 1Z3, Canada.
| | - Suzana K Straus
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada.
| |
Collapse
|
87
|
Pachón-Ibáñez ME, Smani Y, Pachón J, Sánchez-Céspedes J. Perspectives for clinical use of engineered human host defense antimicrobial peptides. FEMS Microbiol Rev 2018; 41:323-342. [PMID: 28521337 PMCID: PMC5435762 DOI: 10.1093/femsre/fux012] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/28/2017] [Indexed: 12/15/2022] Open
Abstract
Infectious diseases caused by bacteria, viruses or fungi are among the leading causes of death worldwide. The emergence of drug-resistance mechanisms, especially among bacteria, threatens the efficacy of all current antimicrobial agents, some of them already ineffective. As a result, there is an urgent need for new antimicrobial drugs. Host defense antimicrobial peptides (HDPs) are natural occurring and well-conserved peptides of innate immunity, broadly active against Gram-negative and Gram-positive bacteria, viruses and fungi. They also are able to exert immunomodulatory and adjuvant functions by acting as chemotactic for immune cells, and inducing cytokines and chemokines secretion. Moreover, they show low propensity to elicit microbial adaptation, probably because of their non-specific mechanism of action, and are able to neutralize exotoxins and endotoxins. HDPs have the potential to be a great source of novel antimicrobial agents. The goal of this review is to provide an overview of the advances made in the development of human defensins as well as the cathelicidin LL-37 and their derivatives as antimicrobial agents against bacteria, viruses and fungi for clinical use.
Collapse
Affiliation(s)
- María Eugenia Pachón-Ibáñez
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville
| | - Younes Smani
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville
| | - Jerónimo Pachón
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville.,Department of Medicine, University of Seville, Seville, Spain
| | - Javier Sánchez-Céspedes
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville.,Department of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
88
|
Lewis ML, Surewaard BGJ. Neutrophil evasion strategies by Streptococcus pneumoniae and Staphylococcus aureus. Cell Tissue Res 2017; 371:489-503. [PMID: 29204747 DOI: 10.1007/s00441-017-2737-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/06/2017] [Indexed: 02/05/2023]
Abstract
Humans are well equipped to defend themselves against bacteria. The innate immune system employs diverse mechanisms to recognize, control and initiate a response that can destroy millions of different microbes. Microbes that evade the sophisticated innate immune system are able to escape detection and could become pathogens. The pathogens Streptococcus pneumoniae and Staphylococcus aureus are particularly successful due to the development of a wide variety of virulence strategies for bacterial pathogenesis and they invest significant efforts towards mechanisms that allow for neutrophil evasion. Neutrophils are a primary cellular defense and can rapidly kill invading microbes, which is an indispensable function for maintaining host health. This review compares the key features of Streptococcus pneumoniae and Staphylococcus aureus in epidemiology, with a specific focus on virulence mechanisms utilized to evade neutrophils in bacterial pathogenesis. It is important to understand the complex interactions between pathogenic bacteria and neutrophils so that we can disrupt the ability of pathogens to cause disease.
Collapse
Affiliation(s)
- Megan L Lewis
- Department of Physiology & Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Bas G J Surewaard
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada. .,Department of Medical Microbiology, University Medical Centre, Utrecht, Netherlands.
| |
Collapse
|
89
|
Kudryashova E, Seveau SM, Kudryashov DS. Targeting and inactivation of bacterial toxins by human defensins. Biol Chem 2017; 398:1069-1085. [PMID: 28593905 DOI: 10.1515/hsz-2017-0106] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/18/2017] [Indexed: 11/15/2022]
Abstract
Defensins, as a prominent family of antimicrobial peptides (AMP), are major effectors of the innate immunity with a broad range of immune modulatory and antimicrobial activities. In particular, defensins are the only recognized fast-response molecules that can neutralize a broad range of bacterial toxins, many of which are among the deadliest compounds on the planet. For a decade, the mystery of how a small and structurally conserved group of peptides can neutralize a heterogeneous group of toxins with little to no sequential and structural similarity remained unresolved. Recently, it was found that defensins recognize and target structural plasticity/thermodynamic instability, fundamental physicochemical properties that unite many bacterial toxins and distinguish them from the majority of host proteins. Binding of human defensins promotes local unfolding of the affected toxins, destabilizes their secondary and tertiary structures, increases susceptibility to proteolysis, and leads to their precipitation. While the details of toxin destabilization by defensins remain obscure, here we briefly review properties and activities of bacterial toxins known to be affected by or resilient to defensins, and discuss how recognized features of defensins correlate with the observed inactivation.
Collapse
|
90
|
Kim CH, Lee YJ, Go HJ, Oh HY, Lee TK, Park JB, Park NG. Defensin-neurotoxin dyad in a basally branching metazoan sea anemone. FEBS J 2017; 284:3320-3338. [DOI: 10.1111/febs.14194] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/27/2017] [Accepted: 08/07/2017] [Indexed: 11/26/2022]
Affiliation(s)
- Chan-Hee Kim
- Department of Biotechnology; College of Fisheries Sciences; Pukyong National University; Busan Korea
| | - Ye Jin Lee
- Department of Biotechnology; College of Fisheries Sciences; Pukyong National University; Busan Korea
| | - Hye-Jin Go
- Department of Biotechnology; College of Fisheries Sciences; Pukyong National University; Busan Korea
| | - Hye Young Oh
- Department of Biotechnology; College of Fisheries Sciences; Pukyong National University; Busan Korea
| | - Tae Kwan Lee
- Department of Biotechnology; College of Fisheries Sciences; Pukyong National University; Busan Korea
| | - Ji Been Park
- Department of Biotechnology; College of Fisheries Sciences; Pukyong National University; Busan Korea
| | - Nam Gyu Park
- Department of Biotechnology; College of Fisheries Sciences; Pukyong National University; Busan Korea
| |
Collapse
|
91
|
Müller A, Klöckner A, Schneider T. Targeting a cell wall biosynthesis hot spot. Nat Prod Rep 2017; 34:909-932. [PMID: 28675405 DOI: 10.1039/c7np00012j] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Covering: up to 2017History points to the bacterial cell wall biosynthetic network as a very effective target for antibiotic intervention, and numerous natural product inhibitors have been discovered. In addition to the inhibition of enzymes involved in the multistep synthesis of the macromolecular layer, in particular, interference with membrane-bound substrates and intermediates essential for the biosynthetic reactions has proven a valuable antibacterial strategy. A prominent target within the peptidoglycan biosynthetic pathway is lipid II, which represents a particular "Achilles' heel" for antibiotic attack, as it is readily accessible on the outside of the cytoplasmic membrane. Lipid II is a unique non-protein target that is one of the structurally most conserved molecules in bacterial cells. Notably, lipid II is more than just a target molecule, since sequestration of the cell wall precursor may be combined with additional antibiotic activities, such as the disruption of membrane integrity or disintegration of membrane-bound multi-enzyme machineries. Within the membrane bilayer lipid II is likely organized in specific anionic phospholipid patches that form a particular "landing platform" for antibiotics. Nature has invented a variety of different "lipid II binders" of at least 5 chemical classes, and their antibiotic activities can vary substantially depending on the compounds' physicochemical properties, such as amphiphilicity and charge, and thus trigger diverse cellular effects that are decisive for antibiotic activity.
Collapse
Affiliation(s)
- Anna Müller
- Institute of Pharmaceutical Microbiology, University of Bonn, Bonn, Germany.
| | | | | |
Collapse
|
92
|
Mathew B, Nagaraj R. Variations in the interaction of human defensins with Escherichia coli: Possible implications in bacterial killing. PLoS One 2017; 12:e0175858. [PMID: 28423004 PMCID: PMC5397029 DOI: 10.1371/journal.pone.0175858] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/31/2017] [Indexed: 01/08/2023] Open
Abstract
Human α and β-defensins are cationic antimicrobial peptides characterized by three disulfide bonds with a triple stranded β-sheet motif. It is presumed that interaction with the bacterial cell surface and membrane permeabilization by defensins is an important step in the killing process. In this study, we have compared interactions of three human α-defensins HNP3, HNP4, HD5 and human β-defensins HBD1-4 that are active against Escherichia coli, with its cell surface and inner membrane as well as negatively charged model membranes. We have also included the inactive α-defensin HD6 in the study. Among the α-defensins, HNP4, HD5 and HD6 were more effective in increasing the zeta potential as compared to HNP3. Among the β-defensins, HBD1 was the least effective in increasing the zeta potential. The zeta potential modulation data indicate variations in the surface charge neutralizing ability of α- and β-defensins. Comparison of E. coli inner membrane and model membrane permeabilizing abilities indicated that HD5, HD6 and HBD1 do not permeabilize membranes. Although HBD4 does not permeabilize model membranes, considerable damage to the inner membrane of E. coli is observed. Our data indicate that mammalian defensins do not kill E. coli by a simple mechanism involving membrane permeabilization though their antibacterial potencies are very similar.
Collapse
Affiliation(s)
- Basil Mathew
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | |
Collapse
|
93
|
Chairatana P, Nolan EM. Human α-Defensin 6: A Small Peptide That Self-Assembles and Protects the Host by Entangling Microbes. Acc Chem Res 2017; 50:960-967. [PMID: 28296382 DOI: 10.1021/acs.accounts.6b00653] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human α-defensin 6 (HD6) is a 32-residue cysteine-rich peptide that contributes to innate immunity by protecting the host at mucosal sites. This peptide is produced in small intestinal Paneth cells, stored as an 81-residue precursor peptide named proHD6 in granules, and released into the lumen. One unusual feature of HD6 is that it lacks the broad-spectrum antimicrobial activity observed for other human α-defensins, including the Paneth cell peptide human α-defensin 5 (HD5). HD6 exhibits unprecedented self-assembly properties, which confer an unusual host-defense function. HD6 monomers self-assemble into higher-order oligomers termed "nanonets", which entrap microbes and prevent invasive gastrointestinal pathogens such as Salmonella enterica serovar Typhimurium and Listeria monocytogenes from entering host cells. One possible advantage of this host-defense mechanism is that HD6 helps to keep microbes in the lumen such that they can be excreted or attacked by other components of the immune system, such as recruited neutrophils. In this Account, we report our current understanding of HD6 and focus on work published since 2012 when Bevins and co-workers described the discovery of HD6 nanonets in the literature. First, we present studies that address the biosynthesis, storage, and maturation of HD6, which demonstrate that nature uses a propeptide strategy to spatially and temporally control the formation of HD6 nanonets in the small intestine. The propeptide is stored in Paneth cell granules, and proteolysis occurs during or following release into the lumen, which affords the 32-residue mature peptide that self-assembles. We subsequently highlight structure-function studies that provide a foundation for understanding the molecular basis for why HD6 exhibits unusual self-assembly properties compared with other characterized defensins. The disposition of hydrophobic residues in the HD6 primary structure differs from that of other human α-defensins and is an important structural determinant for oligomerization. Lastly, we consider functional studies that illuminate how HD6 contributes to mucosal immunity. We recently discovered that in addition to blocking bacterial invasion into host epithelial cells by Gram-negative and Gram-positive gastrointestinal pathogens, HD6 suppresses virulence traits displayed by the opportunistic human fungal pathogen Candida albicans. In particular, we found that C. albicans biofilm formation, which causes complications in the treatment of candidiasis, is inhibited by HD6. This observation suggests that HD6 may contribute to intestinal homeostasis by helping to keep C. albicans in its commensal state. We intend for this Account to inspire further biochemical, biophysical, and biological investigations that will advance our understanding of HD6 in mucosal immunity and the host-microbe interaction.
Collapse
Affiliation(s)
- Phoom Chairatana
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
94
|
López-Pérez PM, Grimsey E, Bourne L, Mikut R, Hilpert K. Screening and Optimizing Antimicrobial Peptides by Using SPOT-Synthesis. Front Chem 2017; 5:25. [PMID: 28447030 PMCID: PMC5388751 DOI: 10.3389/fchem.2017.00025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/29/2017] [Indexed: 11/14/2022] Open
Abstract
Peptide arrays on cellulose are a powerful tool to investigate peptide interactions with a number of different molecules, for examples antibodies, receptors or enzymes. Such peptide arrays can also be used to study interactions with whole cells. In this review, we focus on the interaction of small antimicrobial peptides with bacteria. Antimicrobial peptides (AMPs) can kill multidrug-resistant (MDR) human pathogenic bacteria and therefore could be next generation antibiotics targeting MDR bacteria. We describe the screen and the result of different optimization strategies of peptides cleaved from the membrane. In addition, screening of antibacterial activity of peptides that are tethered to the surface is discussed. Surface-active peptides can be used to protect surfaces from bacterial infections, for example implants.
Collapse
Affiliation(s)
| | - Elizabeth Grimsey
- Institute for Infection and Immunity, St. George's University of LondonLondon, UK
| | - Luc Bourne
- Institute for Infection and Immunity, St. George's University of LondonLondon, UK
| | - Ralf Mikut
- Karlsruhe Institute of Technology (KIT), Institute for Applied Computer Science (IAI)Eggenstein-Leopoldshafen, Germany
| | - Kai Hilpert
- TiKa Diagnostics LtdLondon, UK
- Institute for Infection and Immunity, St. George's University of LondonLondon, UK
| |
Collapse
|
95
|
Min HJ, Yun H, Ji S, Rajasekaran G, Kim JI, Kim JS, Shin SY, Lee CW. Rattusin structure reveals a novel defensin scaffold formed by intermolecular disulfide exchanges. Sci Rep 2017; 7:45282. [PMID: 28345637 PMCID: PMC5366907 DOI: 10.1038/srep45282] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/23/2017] [Indexed: 12/14/2022] Open
Abstract
Defensin peptides are essential for innate immunity in humans and other living systems, as they provide protection against infectious pathogens and regulate the immune response. Here, we report the solution structure of rattusin (RTSN), an α-defensin-related peptide, which revealed a novel C2-symmetric disulfide-linked dimeric structure. RTSN was synthesized by solid-phase peptide synthesis (SPPS) and refolded by air oxidation in vitro. Dimerization of the refolded RTSN (r-RTSN) resulted from five intermolecular disulfide (SS) bond exchanges formed by ten cysteines within two protomer chains. The SS bond pairings of r-RTSN were determined by mass analysis of peptide fragments cleaved by trypsin digestion. In addition to mass analysis, nuclear magnetic resonance (NMR) experiments for a C15S mutant and r-RTSN confirmed that the intermolecular SS bond structure of r-RTSN showed an I-V', II-IV', III-III', IV-II', V-I' arrangement. The overall structure of r-RTSN exhibited a cylindrical array, similar to that of β-sandwich folds, with a highly basic surface. Furthermore, fluorescence spectroscopy results suggest that r-RTSN exerts bactericidal activity by damaging membrane integrity. Collectively, these results provide a novel structural scaffold for designing highly potent peptide-based antibiotics suitable for use under various physiological conditions.
Collapse
Affiliation(s)
- Hye Jung Min
- Department of Chemistry, Chonnam National University, Gwangju 61186, South Korea.,Department of Pharmaceutical Cosmetics, Kwangju Women's University, Gwangju 62396, South Korea
| | - Hyosuk Yun
- Department of Chemistry, Chonnam National University, Gwangju 61186, South Korea
| | - Sehyeon Ji
- Department of Chemistry, Chonnam National University, Gwangju 61186, South Korea
| | - Ganesan Rajasekaran
- Department of Medical Science, Graduate School and Department of Cellular and Molecular Medicine, School of Medicine, Chosun University, Gwangju 61452, South Korea
| | - Jae Il Kim
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Jeong-Sun Kim
- Department of Chemistry, Chonnam National University, Gwangju 61186, South Korea
| | - Song Yub Shin
- Department of Medical Science, Graduate School and Department of Cellular and Molecular Medicine, School of Medicine, Chosun University, Gwangju 61452, South Korea
| | - Chul Won Lee
- Department of Chemistry, Chonnam National University, Gwangju 61186, South Korea
| |
Collapse
|
96
|
Abstract
Antimicrobial peptides (AMPs) are expressed in various living organisms as first-line host defenses against potential harmful encounters in their surroundings. AMPs are short polycationic peptides exhibiting various antimicrobial activities. The principal antibacterial activity is attributed to the membrane-lytic mechanism which directly interferes with the integrity of the bacterial cell membrane and cell wall. In addition, a number of AMPs form a transmembrane channel in the membrane by self-aggregation or polymerization, leading to cytoplasm leakage and cell death. However, an increasing body of evidence has demonstrated that AMPs are able to exert intracellular inhibitory activities as the primary or supportive mechanisms to achieve efficient killing. In this review, we focus on the major intracellular targeting activities reported in AMPs, which include nucleic acids and protein biosynthesis and protein-folding, protease, cell division, cell wall biosynthesis, and lipopolysaccharide inhibition. These multifunctional AMPs could serve as the potential lead peptides for the future development of novel antibacterial agents with improved therapeutic profiles.
Collapse
|
97
|
Malin J, Shetty AC, Daugherty SC, de Leeuw EP. Effect of a small molecule Lipid II binder on bacterial cell wall stress. Infect Drug Resist 2017; 10:69-73. [PMID: 28280373 PMCID: PMC5338996 DOI: 10.2147/idr.s126254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We have recently identified small molecule compounds that act as binders of Lipid II, an essential precursor of bacterial cell wall biosynthesis. Lipid II comprised a hydrophilic head group that includes a peptidoglycan subunit composed of N-acetylglucosamine (GlcNAc) and N-acetylmuramic acid (MurNAc) coupled to a short pentapeptide moiety. This headgroup is coupled to a long bactoprenol chain via a pyrophosphate group. Here, we report on the cell wall activity relationship of dimethyl-3-methyl(phenyl)amino-ethenylcyclohexylidene-propenyl-3-ethyl-1,3-benzothiazolium iodide (compound 5107930) obtained by functional and genetic analyses. Our results indicate that compounds bind to Lipid II and cause specific upregulation of the vancomycin-resistance associated gene vraX. vraX is implicated in the cell wall stress stimulon that confers glycopeptide resistance. Our small molecule Lipid II inhibitor retained activity against strains of Staphylococcus aureus mutated in genes encoding the cell wall stress stimulon. This suggests the feasibility of developing this new scaffold as a therapeutic agent in view of increasing glycopeptide resistance.
Collapse
Affiliation(s)
- Jakob Malin
- Institute of Human Virology; Department of Biochemistry and Molecular Biology
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland Baltimore School of Medicine, Baltimore, MD, USA
| | - Sean C Daugherty
- Institute for Genome Sciences, University of Maryland Baltimore School of Medicine, Baltimore, MD, USA
| | - Erik Ph de Leeuw
- Institute of Human Virology; Department of Biochemistry and Molecular Biology
| |
Collapse
|
98
|
Shafee TMA, Lay FT, Phan TK, Anderson MA, Hulett MD. Convergent evolution of defensin sequence, structure and function. Cell Mol Life Sci 2017; 74:663-682. [PMID: 27557668 PMCID: PMC11107677 DOI: 10.1007/s00018-016-2344-5] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/27/2016] [Accepted: 08/15/2016] [Indexed: 02/06/2023]
Abstract
Defensins are a well-characterised group of small, disulphide-rich, cationic peptides that are produced by essentially all eukaryotes and are highly diverse in their sequences and structures. Most display broad range antimicrobial activity at low micromolar concentrations, whereas others have other diverse roles, including cell signalling (e.g. immune cell recruitment, self/non-self-recognition), ion channel perturbation, toxic functions, and enzyme inhibition. The defensins consist of two superfamilies, each derived from an independent evolutionary origin, which have subsequently undergone extensive divergent evolution in their sequence, structure and function. Referred to as the cis- and trans-defensin superfamilies, they are classified based on their secondary structure orientation, cysteine motifs and disulphide bond connectivities, tertiary structure similarities and precursor gene sequence. The utility of displaying loops on a stable, compact, disulphide-rich core has been exploited by evolution on multiple occasions. The defensin superfamilies represent a case where the ensuing convergent evolution of sequence, structure and function has been particularly extreme. Here, we discuss the extent, causes and significance of these convergent features, drawing examples from across the eukaryotes.
Collapse
Affiliation(s)
- Thomas M A Shafee
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Fung T Lay
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Thanh Kha Phan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Marilyn A Anderson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
99
|
Genomic Landscape of Intrahost Variation in Group A Streptococcus: Repeated and Abundant Mutational Inactivation of the fabT Gene Encoding a Regulator of Fatty Acid Synthesis. Infect Immun 2016; 84:3268-3281. [PMID: 27600505 DOI: 10.1128/iai.00608-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/08/2016] [Indexed: 01/03/2023] Open
Abstract
To obtain new information about Streptococcus pyogenes intrahost genetic variation during invasive infection, we sequenced the genomes of 2,954 serotype M1 strains recovered from a nonhuman primate experimental model of necrotizing fasciitis. A total of 644 strains (21.8%) acquired polymorphisms relative to the input parental strain. The fabT gene, encoding a transcriptional regulator of fatty acid biosynthesis genes, contained 54.5% of these changes. The great majority of polymorphisms were predicted to deleteriously alter FabT function. Transcriptome-sequencing (RNA-seq) analysis of a wild-type strain and an isogenic fabT deletion mutant strain found that between 3.7 and 28.5% of the S. pyogenes transcripts were differentially expressed, depending on the growth temperature (35°C or 40°C) and growth phase (mid-exponential or stationary phase). Genes implicated in fatty acid synthesis and lipid metabolism were significantly upregulated in the fabT deletion mutant strain. FabT also directly or indirectly regulated central carbon metabolism genes, including pyruvate hub enzymes and fermentation pathways and virulence genes. Deletion of fabT decreased virulence in a nonhuman primate model of necrotizing fasciitis. In addition, the fabT deletion strain had significantly decreased survival in human whole blood and during phagocytic interaction with polymorphonuclear leukocytes ex vivo We conclude that FabT mutant progeny arise during infection, constitute a metabolically distinct subpopulation, and are less virulent in the experimental models used here.
Collapse
|
100
|
Chauhan J, Cardinale S, Fang L, Huang J, Kwasny SM, Pennington MR, Basi K, diTargiani R, Capacio BR, MacKerell AD, Opperman TJ, Fletcher S, de Leeuw EPH. Towards Development of Small Molecule Lipid II Inhibitors as Novel Antibiotics. PLoS One 2016; 11:e0164515. [PMID: 27776124 PMCID: PMC5077133 DOI: 10.1371/journal.pone.0164515] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/25/2016] [Indexed: 12/28/2022] Open
Abstract
Recently we described a novel di-benzene-pyrylium-indolene (BAS00127538) inhibitor of Lipid II. BAS00127538 (1-Methyl-2,4-diphenyl-6-((1E,3E)-3-(1,3,3-trimethylindolin-2-ylidene)prop-1-en-1-yl)pyryl-1-ium) tetrafluoroborate is the first small molecule Lipid II inhibitor and is structurally distinct from natural agents that bind Lipid II, such as vancomycin. Here, we describe the synthesis and biological evaluation of 50 new analogs of BAS00127538 designed to explore the structure-activity relationships of the scaffold. The results of this study indicate an activity map of the scaffold, identifying regions that are critical to cytotoxicity, Lipid II binding and range of anti-bacterial action. One compound, 6jc48-1, showed significantly enhanced drug-like properties compared to BAS00127538. 6jc48-1 has reduced cytotoxicity, while retaining specific Lipid II binding and activity against Enterococcus spp. in vitro and in vivo. Further, this compound showed a markedly improved pharmacokinetic profile with a half-life of over 13 hours upon intravenous and oral administration and was stable in plasma. These results suggest that scaffolds like that of 6jc48-1 can be developed into small molecule antibiotic drugs that target Lipid II.
Collapse
Affiliation(s)
- Jamal Chauhan
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, Maryland, United States of America
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Steven Cardinale
- Microbiotix, Inc., One Innovation Drive, Worcester, Massachusetts, United States of America
| | - Lei Fang
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, Maryland, United States of America
- Computer-Aided Drug Design Center, University of Maryland, School of Pharmacy, Baltimore, Maryland, United States of America
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jing Huang
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, Maryland, United States of America
- Computer-Aided Drug Design Center, University of Maryland, School of Pharmacy, Baltimore, Maryland, United States of America
| | - Steven M. Kwasny
- Microbiotix, Inc., One Innovation Drive, Worcester, Massachusetts, United States of America
| | - M. Ross Pennington
- U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland, United States of America
| | - Kelly Basi
- U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland, United States of America
| | - Robert diTargiani
- U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland, United States of America
| | - Benedict R. Capacio
- U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland, United States of America
| | - Alexander D. MacKerell
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, Maryland, United States of America
- Computer-Aided Drug Design Center, University of Maryland, School of Pharmacy, Baltimore, Maryland, United States of America
| | - Timothy J. Opperman
- Microbiotix, Inc., One Innovation Drive, Worcester, Massachusetts, United States of America
| | - Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, Maryland, United States of America
| | - Erik P. H. de Leeuw
- Institute of Human Virology & Department of Biochemistry and Molecular Biology of the University of Maryland Baltimore School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|