1
|
Kim S, Kim NH, Khaleel ZH, Sa DH, Choi D, Ga S, Kim CG, Jang J, Kim K, Kim YJ, Chang SN, Park SM, Park SY, Lee B, Kim J, Lee J, An S, Park JG, Kim YH. Mussel‐Inspired Recombinant Adhesive Protein‐Based Functionalization for Consistent and Effective Antimicrobial Treatment in Chronic Inflammatory Skin Diseases. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202300353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Indexed: 08/07/2024]
Abstract
AbstractChronic inflammatory skin diseases, characterized by a vicious cycle of infection and hyperinflammation, necessitate consistent and effective antimicrobial treatment of target lesions to achieve practical therapeutic outcomes. Antimicrobial dressing materials offer notable advantages over conventional therapeutic drugs, including ease of application, extended contact time, and targeted antimicrobial action, resulting in enhanced efficacy in breaking the vicious cycle. In line with these advantages, this study aims to develop a plug‐and‐playable recombinant adhesive protein (RAP) inspired by the adhesive properties of marine mussels, serving as a durable and effective surface functionalization strategy. By genetically recombining mussel foot protein with antimicrobial peptides, RAP effectively incorporates antimicrobial properties into biomaterials for treating chronic inflammatory skin diseases. The durable adhesion of RAP ensures long‐lasting antimicrobial functionality on target surfaces, MFP making it a promising approach to inhibit chronic inflammation. In addition, when dip‐coated onto cotton gauze, RAP can be utilized as an antimicrobial patch, effectively suppressing chronic inflammation through the inhibition of bacteria‐induced toll‐like receptor signaling. These findings underscore the potential of nature‐inspired protein‐based surface functionalization of biomaterials as a compelling approach to advance the treatment of chronic inflammatory skin diseases.
Collapse
Affiliation(s)
- Suhyeon Kim
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Nano Science and Technology Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Nam Hyeong Kim
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Nano Science and Technology Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Zinah Hilal Khaleel
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Nano Science and Technology Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Deok Hyang Sa
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Nano Science and Technology Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Daekyu Choi
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Seongmin Ga
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Nano Science and Technology Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Chang Geon Kim
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Advanced Bio Convergence Center (ABCC) Pohang Technopark Foundation Pohang Gyeongbuk 37668 Republic of Korea
| | - Jiye Jang
- School of Pharmacy Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Biopharmaceutical Convergence Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Kyeonghyun Kim
- Department of Nano Science and Technology Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Ye Ji Kim
- Department of Nano Science and Technology Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Sukkum Ngullie Chang
- Advanced Bio Convergence Center (ABCC) Pohang Technopark Foundation Pohang Gyeongbuk 37668 Republic of Korea
| | - Seon Min Park
- Advanced Bio Convergence Center (ABCC) Pohang Technopark Foundation Pohang Gyeongbuk 37668 Republic of Korea
| | - Su Yeon Park
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Nano Science and Technology Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Bok‐Soo Lee
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Jin‐Chul Kim
- Natural Products Research Institute Korea Institute of Science and Technology Gangneung Gangwon‐do 25451 Republic of Korea
| | - Jaecheol Lee
- School of Pharmacy Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Biopharmaceutical Convergence Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Imnewrun Inc. 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Seongpil An
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC) Pohang Technopark Foundation Pohang Gyeongbuk 37668 Republic of Korea
- Department of Nano Engineering Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| | - Yong Ho Kim
- SKKU Advanced Institute of Nanotechnology (SAINT) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Nano Science and Technology Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Imnewrun Inc. 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS) Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
- Department of Nano Engineering Sungkyunkwan University (SKKU) 2066 Seobu‐ro, Jangan‐gu Suwon Gyeonggi‐do 16419 Republic of Korea
| |
Collapse
|
2
|
Atomic-Resolution Structures and Mode of Action of Clinically Relevant Antimicrobial Peptides. Int J Mol Sci 2022; 23:ijms23094558. [PMID: 35562950 PMCID: PMC9100274 DOI: 10.3390/ijms23094558] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
Global rise of infections and deaths caused by drug-resistant bacterial pathogens are among the unmet medical needs. In an age of drying pipeline of novel antibiotics to treat bacterial infections, antimicrobial peptides (AMPs) are proven to be valid therapeutics modalities. Direct in vivo applications of many AMPs could be challenging; however, works are demonstrating encouraging results for some of them. In this review article, we discussed 3-D structures of potent AMPs e.g., polymyxin, thanatin, MSI, protegrin, OMPTA in complex with bacterial targets and their mode of actions. Studies on human peptide LL37 and de novo-designed peptides are also discussed. We have focused on AMPs which are effective against drug-resistant Gram-negative bacteria. Since treatment options for the infections caused by super bugs of Gram-negative bacteria are now extremely limited. We also summarize some of the pertinent challenges in the field of clinical trials of AMPs.
Collapse
|
3
|
Sheard DE, Li W, O’Brien-Simpson NM, Separovic F, Wade JD. Peptide Multimerization as Leads for Therapeutic Development. BIOLOGICS 2021; 2:15-44. [DOI: 10.3390/biologics2010002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Multimerization of peptide structures has been a logical evolution in their development as potential therapeutic molecules. The multivalent properties of these assemblies have attracted much attention from researchers in the past and the development of more complex branching dendrimeric structures, with a wide array of biocompatible building blocks is revealing previously unseen properties and activities. These branching multimer and dendrimer structures can induce greater effect on cellular targets than monomeric forms and act as potent antimicrobials, potential vaccine alternatives and promising candidates in biomedical imaging and drug delivery applications. This review aims to outline the chemical synthetic innovations for the development of these highly complex structures and highlight the extensive capabilities of these molecules to rival those of natural biomolecules.
Collapse
Affiliation(s)
- Dean E. Sheard
- School of Chemistry, University of Melbourne, Melbourne, VIC 3010, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Wenyi Li
- ACTV Research Group, Centre for Oral Health Research, The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Neil M. O’Brien-Simpson
- ACTV Research Group, Centre for Oral Health Research, The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Frances Separovic
- School of Chemistry, University of Melbourne, Melbourne, VIC 3010, Australia
| | - John D. Wade
- School of Chemistry, University of Melbourne, Melbourne, VIC 3010, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
4
|
Tummanapalli SS, Willcox MD. Antimicrobial resistance of ocular microbes and the role of antimicrobial peptides. Clin Exp Optom 2021; 104:295-307. [PMID: 32924208 DOI: 10.1111/cxo.13125] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Isolation of antimicrobial-resistant microbes from ocular infections may be becoming more frequent. Infections caused by these microbes can be difficult to treat and lead to poor outcomes. However, new therapies are being developed which may help improve clinical outcomes. This review examines recent reports on the isolation of antibiotic-resistant microbes from ocular infections. In addition, an overview of the development of some new antibiotic therapies is given. The recent literature regarding antibiotic use and resistance, isolation of antibiotic-resistant microbes from ocular infections and the development of potential new antibiotics that can be used to treat these infections was reviewed. Ocular microbial infections are a global public health issue as they can result in vision loss which compromises quality of life. Approximately 70 per cent of ocular infections are caused by bacteria including Chlamydia trachomatis, Staphylococcus aureus, and Pseudomonas aeruginosa and fungi such as Candida albicans, Aspergillus spp. and Fusarium spp. Resistance to first-line antibiotics such as fluoroquinolones and azoles has increased, with resistance of S. aureus isolates from the USA to fluoroquinolones reaching 32 per cent of isolates and 35 per cent being methicillin-resistant (MRSA). Lower levels of MRSA (seven per cent) were isolated by an Australian study. Antimicrobial peptides, which are broad-spectrum alternatives to antibiotics, have been tested as possible new drugs. Several have shown promise in animal models of keratitis, especially treating P. aeruginosa, S. aureus or C. albicans infections. Reports of increasing resistance of ocular isolates to mainstay antibiotics are a concern, and there is evidence that for ocular surface disease this resistance translates into worse clinical outcomes. New antibiotics are being developed, but not by large pharmaceutical companies and mostly in university research laboratories and smaller biotech companies. Antimicrobial peptides show promise in treating keratitis.
Collapse
Affiliation(s)
| | - Mark Dp Willcox
- School of Optometry and Vision Science, The University of New South Wales, Sydney, Australia
| |
Collapse
|
5
|
Zheng H, Ji Z, Roy KR, Gao M, Pan Y, Cai X, Wang L, Li W, Chang CH, Kaweeteerawat C, Chen C, Xia T, Zhao Y, Li R. Engineered Graphene Oxide Nanocomposite Capable of Preventing the Evolution of Antimicrobial Resistance. ACS NANO 2019; 13:11488-11499. [PMID: 31566947 DOI: 10.1021/acsnano.9b04970] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Antimicrobial resistance (AMR) is spreading worldwide and keeps evolving to adapt to antibiotics, causing increasing threats in clinics, which necessitates the exploration of antimicrobial agents for not only killing of resistant cells but also prevention of AMR progression. However, so far, there has been no effective approach. Herein, we designed lanthanum hydroxide and graphene oxide nanocomposites (La@GO) to confer a synergistic bactericidal effect in all tested resistant strains. More importantly, long-term exposure of E. coli (AMR) to subminimum inhibitory concentrations of La@GO does not trigger detectable secondary resistance, while conventional antibiotics and silver nanoparticles lead to a 16- to 64-fold increase in tolerance. The inability of E. coli to evolve resistance to La@GO is likely due to a distinctive extracellular multitarget invasion killing mechanism involving lipid dephosphorylation, lipid peroxidation, and peptidoglycan disruption. Overall, our results highlight La@GO nanocomposites as a promising solution to combating resistant bacteria without inducing the evolution of AMR.
Collapse
Affiliation(s)
- Huizhen Zheng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou , 215123 , Jiangsu , China
| | - Zhaoxia Ji
- Division of Nanomedicine, Department of Medicine, California NanoSystems Institute , University of California , Los Angeles , California 90095 , United States
- Living Proof, Inc. , Cambridge , Massachusetts 02142 , United States
| | - Kevin R Roy
- Department of Genetics, School of Medicine , Stanford University , Palo Alto , California 94304 , United States
| | - Meng Gao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou , 215123 , Jiangsu , China
| | - Yanxia Pan
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou , 215123 , Jiangsu , China
| | - Xiaoming Cai
- School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases , Soochow University , Suzhou , 215123 , Jiangsu , China
| | - Liming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety , Institute of High Energy Physics, Chinese Academy of Sciences (CAS) , Beijing , 100049 , China
| | - Wei Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou , 215123 , Jiangsu , China
| | - Chong Hyun Chang
- Division of Nanomedicine, Department of Medicine, California NanoSystems Institute , University of California , Los Angeles , California 90095 , United States
| | - Chitrada Kaweeteerawat
- National Nanotechnology Center (NANOTEC), NSTDA Characterization and Testing Center (NCTC), National Science and Technology Development Agency (NSTDA) , Klong Nueng , 12120 , Thailand
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China, Chinese Academy of Sciences (CAS) , Beijing , 100190 , China
| | - Tian Xia
- Division of Nanomedicine, Department of Medicine, California NanoSystems Institute , University of California , Los Angeles , California 90095 , United States
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China, Chinese Academy of Sciences (CAS) , Beijing , 100190 , China
| | - Ruibin Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions , Soochow University , Suzhou , 215123 , Jiangsu , China
| |
Collapse
|
6
|
Nelson N, Schwartz DK. Single-Molecule Resolution of Antimicrobial Peptide Interactions with Supported Lipid A Bilayers. Biophys J 2018; 114:2606-2616. [PMID: 29874611 PMCID: PMC6129183 DOI: 10.1016/j.bpj.2018.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/16/2018] [Accepted: 04/12/2018] [Indexed: 01/12/2023] Open
Abstract
The molecular interactions between antimicrobial peptides (AMPs) and lipid A-containing supported lipid bilayers were probed using single-molecule total internal reflection fluorescence microscopy. Hybrid supported lipid bilayers with lipid A outer leaflets and phospholipid (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE)) inner leaflets were prepared and characterized, and the spatiotemporal trajectories of individual fluorescently labeled LL37 and Melittin AMPs were determined as they interacted with the bilayer surfaces comprising either monophosphoryl or diphosphoryl lipid A (from Escherichia coli) to determine the impact of electrostatic interactions. Large numbers of trajectories were obtained and analyzed to obtain the distributions of surface residence times and the statistics of the spatial trajectories. Interestingly, the AMP species were sensitive to subtle differences in the charge of the lipid, with both peptides diffusing more slowly and residing longer on the diphosphoryl lipid A. Furthermore, the single-molecule dynamics indicated a qualitative difference between the behavior of AMPs on hybrid Lipid A bilayers and on those composed entirely of DOPE. Whereas AMPs interacting with a DOPE bilayer exhibited two-dimensional Brownian diffusion with a diffusion coefficient of ∼1.7 μm2/s, AMPs adsorbed to the lipid A surface exhibited much slower apparent diffusion (on the order of ∼0.1 μm2/s) and executed intermittent trajectories that alternated between two-dimensional Brownian diffusion and desorption-mediated three-dimensional flights. Overall, these findings suggested that bilayers with lipid A in the outer leaflet, as it is in bacterial outer membranes, are valuable model systems for the study of the initial stage of AMP-bacterium interactions. Furthermore, single-molecule dynamics was sensitive to subtle differences in electrostatic interactions between cationic AMPs and monovalent or divalent anionic lipid A moieties.
Collapse
Affiliation(s)
- Nathaniel Nelson
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Daniel K Schwartz
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado.
| |
Collapse
|
7
|
Koh JJ, Lin S, Bai Y, Sin WWL, Aung TT, Li J, Chandra V, Pervushin K, Beuerman RW, Liu S. Antimicrobial activity profiles of Amphiphilic Xanthone derivatives are a function of their molecular Oligomerization. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:2281-2298. [PMID: 29782818 DOI: 10.1016/j.bbamem.2018.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/09/2018] [Accepted: 05/16/2018] [Indexed: 01/01/2023]
Abstract
Currently, membrane-targeting small antimicrobial peptidomimetics (SAP) are important in antibiotic development because bacteria appear to develop resistance to these surface-active compounds less readily. However, the molecular membrane-targeting action of SAPs has received little attention. In this study, we investigated the effect of oligomerization of amphiphilic xanthone, a model SAP, on its antimicrobial properties against both Gram-positive and Gram-negative bacteria. First, oligomer formation by an amphiphilic xanthone, compound 2 (also coded as AM052), was investigated via solution-state nuclear magnetic resonance (NMR) spectroscopy. Then, the effects of oligomerization on membrane disruption were further studied via biophysical approaches. The results showed that the antimicrobial activities of SAPs develop in several stages: oligomer formation in aqueous solution, initial binding of oligomers to the membrane-water interface followed by insertion into the membrane bilayer, aggregation of antimicrobial oligomers in the membrane, and induced membrane leakage. Ultimately, the presence of the oligomers in the bacterial membrane leads to decreased membrane fluidity and bacterial cell death. Interestingly, the early formation of large oligomers leads to stronger membrane disruption and more rapid bacterial killing. However, reduced antimicrobial activities against Gram-negative bacteria were observed for compounds that formed larger oligomers because the LPS layer acts as a barrier to large complexes. Taken together, our results suggest that oligomerization of SAPs has a strong impact on their antimicrobial properties.
Collapse
Affiliation(s)
- Jun-Jie Koh
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, 169856, Singapore
| | - Shuimu Lin
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, 169856, Singapore
| | - Yang Bai
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, 169856, Singapore; School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Wendy Wan Ling Sin
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, 169856, Singapore
| | - Thet Tun Aung
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, 169856, Singapore
| | - Jianguo Li
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, 169856, Singapore; Bioinformatics Institute, 138671, Singapore
| | - Verma Chandra
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, 169856, Singapore; Bioinformatics Institute, 138671, Singapore
| | - Konstantin Pervushin
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| | - Roger W Beuerman
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, 169856, Singapore; Duke-NUS Medical School, SRP Neuroscience and Behavioral Disorders, 169857, Singapore.
| | - Shouping Liu
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, 169856, Singapore; Duke-NUS Medical School, SRP Neuroscience and Behavioral Disorders, 169857, Singapore.
| |
Collapse
|
8
|
Kozic M, Fox SJ, Thomas JM, Verma CS, Rigden DJ. Large scale ab initio modeling of structurally uncharacterized antimicrobial peptides reveals known and novel folds. Proteins 2018; 86:548-565. [DOI: 10.1002/prot.25473] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Mara Kozic
- Institute of Integrative Biology, University of Liverpool; Liverpool L69 7ZB U.K
- Agency for Science, Technology and Research (A*STAR), Bioinformatics Institute; Singapore
| | - Stephen J. Fox
- Agency for Science, Technology and Research (A*STAR), Bioinformatics Institute; Singapore
| | - Jens M. Thomas
- Institute of Integrative Biology, University of Liverpool; Liverpool L69 7ZB U.K
| | - Chandra S. Verma
- Agency for Science, Technology and Research (A*STAR), Bioinformatics Institute; Singapore
- Department of Biological Sciences; National University of Singapore; Singapore
- School of Biological Sciences; Nanyang Technological University; Singapore
| | - Daniel J. Rigden
- Institute of Integrative Biology, University of Liverpool; Liverpool L69 7ZB U.K
| |
Collapse
|
9
|
Velkov T, Gallardo-Godoy A, Swarbrick JD, Blaskovich MAT, Elliott AG, Han M, Thompson PE, Roberts KD, Huang JX, Becker B, Butler MS, Lash LH, Henriques ST, Nation RL, Sivanesan S, Sani MA, Separovic F, Mertens H, Bulach D, Seemann T, Owen J, Li J, Cooper MA. Structure, Function, and Biosynthetic Origin of Octapeptin Antibiotics Active against Extensively Drug-Resistant Gram-Negative Bacteria. Cell Chem Biol 2018; 25:380-391.e5. [PMID: 29396290 DOI: 10.1016/j.chembiol.2018.01.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 09/03/2017] [Accepted: 12/29/2017] [Indexed: 01/06/2023]
Abstract
Resistance to the last-resort antibiotic colistin is now widespread and new therapeutics are urgently required. We report the first in toto chemical synthesis and pre-clinical evaluation of octapeptins, a class of lipopeptides structurally related to colistin. The octapeptin biosynthetic cluster consisted of three non-ribosomal peptide synthetases (OctA, OctB, and OctC) that produced an amphiphilic antibiotic, octapeptin C4, which was shown to bind to and depolarize membranes. While active against multi-drug resistant (MDR) strains in vitro, octapeptin C4 displayed poor in vivo efficacy, most likely due to high plasma protein binding. Nuclear magnetic resonance solution structures, empirical structure-activity and structure-toxicity models were used to design synthetic octapeptins active against MDR and extensively drug-resistant (XDR) bacteria. The scaffold was then subtly altered to reduce plasma protein binding, while maintaining activity against MDR and XDR bacteria. In vivo efficacy was demonstrated in a murine bacteremia model with a colistin-resistant P. aeruginosa clinical isolate.
Collapse
Affiliation(s)
- Tony Velkov
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia.
| | | | - James D Swarbrick
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, 3052 VIC, Australia
| | - Mark A T Blaskovich
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Alysha G Elliott
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Meiling Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, 3052 VIC, Australia
| | - Philip E Thompson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, 3052 VIC, Australia
| | - Kade D Roberts
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, 3052 VIC, Australia
| | - Johnny X Huang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Bernd Becker
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mark S Butler
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Lawrence H Lash
- Department of Pharmacology, Wayne State University, School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201, USA
| | - Sónia Troeira Henriques
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Roger L Nation
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, 3052 VIC, Australia
| | - Sivashangarie Sivanesan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, 3052 VIC, Australia
| | - Marc-Antoine Sani
- School of Chemistry, Bio21 Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | - Frances Separovic
- School of Chemistry, Bio21 Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | | | - Dieter Bulach
- Department of Immunology and Microbiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Torsten Seemann
- Department of Immunology and Microbiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jeremy Owen
- School of Biological Sciences, Victoria University, Wellington 6012, New Zealand
| | - Jian Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, 3052 VIC, Australia.
| | - Matthew A Cooper
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
10
|
Manzo G, Serra I, Pira A, Pintus M, Ceccarelli M, Casu M, Rinaldi AC, Scorciapino MA. The singular behavior of a β-type semi-synthetic two branched polypeptide: three-dimensional structure and mode of action. Phys Chem Chem Phys 2018; 18:30998-31011. [PMID: 27805179 DOI: 10.1039/c6cp05464a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Dendrimeric peptides make a versatile group of bioactive peptidomimetics and a potential new class of antimicrobial agents to tackle the pressing threat of multi-drug resistant pathogens. These are branched supramolecular assemblies where multiple copies of the bioactive unit are linked to a central core. Beyond their antimicrobial activity, dendrimeric peptides could also be designed to functionalize the surface of nanoparticles or materials for other medical uses. Despite these properties, however, little is known about the structure-function relationship of such compounds, which is key to unveil the fundamental physico-chemical parameters and design analogues with desired attributes. To close this gap, we focused on a semi-synthetic, two-branched peptide, SB056, endowed with remarkable activity against both Gram-positive and Gram-negative bacteria and limited cytotoxicity. SB056 can be considered the smallest prototypical dendrimeric peptide, with the core restricted to a single lysine residue and only two copies of the same highly cationic 10-mer polypeptide; an octanamide tail is present at the C-terminus. Combining NMR and Molecular Dynamics simulations, we have determined the 3D structure of two analogues. Fluorescence spectroscopy was applied to investigate the water-bilayer partition in the presence of vesicles of variable charge. Vesicle leakage assays were also performed and the experimental data were analyzed by applying an iterative Monte Carlo scheme to estimate the minimum number of bound peptides needed to achieve the release. We unveiled a singular beta hairpin-type structure determined by the peptide chains only, with the octanamide tail available for further functionalization to add new potential properties without affecting the structure.
Collapse
Affiliation(s)
- Giorgia Manzo
- Department of Biomedical Sciences - Biochemistry Unit, University of Cagliari, Monserrato, CA, Italy.
| | - Ilaria Serra
- Department of Chemical and Geological Sciences, University of Cagliari, Monserrato, CA, Italy
| | - Alessandro Pira
- Department of Chemical and Geological Sciences, University of Cagliari, Monserrato, CA, Italy
| | - Manuela Pintus
- Department of Biomedical Sciences - Biochemistry Unit, University of Cagliari, Monserrato, CA, Italy.
| | - Matteo Ceccarelli
- Department of Physics, University of Cagliari, Monserrato, CA, Italy
| | - Mariano Casu
- Department of Physics, University of Cagliari, Monserrato, CA, Italy
| | - Andrea C Rinaldi
- Department of Biomedical Sciences - Biochemistry Unit, University of Cagliari, Monserrato, CA, Italy.
| | | |
Collapse
|
11
|
Koh JJ, Lin S, Beuerman RW, Liu S. Recent advances in synthetic lipopeptides as anti-microbial agents: designs and synthetic approaches. Amino Acids 2017; 49:1653-1677. [PMID: 28823054 DOI: 10.1007/s00726-017-2476-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
Abstract
Infectious diseases impose serious public health burdens and continue to be a global public health crisis. The treatment of infections caused by multidrug-resistant pathogens is challenging because only a few viable therapeutic options are clinically available. The emergence and risk of drug-resistant superbugs and the dearth of new classes of antibiotics have drawn increasing awareness that we may return to the pre-antibiotic era. To date, lipopeptides have been received considerable attention because of the following properties: They exhibit potent antimicrobial activities against a broad spectrum of pathogens, rapid bactericidal activity and have a different antimicrobial action compared with most of the conventional antibiotics used today and very slow development of drug resistance tendency. In general, lipopeptides can be structurally classified into two parts: a hydrophilic peptide moiety and a hydrophobic fatty acyl chain. To date, a significant amount of design and synthesis of lipopeptides have been done to improve the therapeutic potential of lipopeptides. This review will present the current knowledge and the recent research in design and synthesis of new lipopeptides and their derivatives in the last 5 years.
Collapse
Affiliation(s)
- Jun-Jie Koh
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, Singapore, 169856, Singapore
| | - Shuimu Lin
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, Singapore, 169856, Singapore
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Roger W Beuerman
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, Singapore, 169856, Singapore.
- SRP Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, 169857, Singapore.
| | - Shouping Liu
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, Singapore, 169856, Singapore.
- SRP Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
12
|
Li J, Koh JJ, Liu S, Lakshminarayanan R, Verma CS, Beuerman RW. Membrane Active Antimicrobial Peptides: Translating Mechanistic Insights to Design. Front Neurosci 2017; 11:73. [PMID: 28261050 PMCID: PMC5306396 DOI: 10.3389/fnins.2017.00073] [Citation(s) in RCA: 346] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/31/2017] [Indexed: 01/10/2023] Open
Abstract
Antimicrobial peptides (AMPs) are promising next generation antibiotics that hold great potential for combating bacterial resistance. AMPs can be both bacteriostatic and bactericidal, induce rapid killing and display a lower propensity to develop resistance than do conventional antibiotics. Despite significant progress in the past 30 years, no peptide antibiotic has reached the clinic yet. Poor understanding of the action mechanisms and lack of rational design principles have been the two major obstacles that have slowed progress. Technological developments are now enabling multidisciplinary approaches including molecular dynamics simulations combined with biophysics and microbiology toward providing valuable insights into the interactions of AMPs with membranes at atomic level. This has led to increasingly robust models of the mechanisms of action of AMPs and has begun to contribute meaningfully toward the discovery of new AMPs. This review discusses the detailed action mechanisms that have been put forward, with detailed atomistic insights into how the AMPs interact with bacterial membranes. The review further discusses how this knowledge is exploited toward developing design principles for novel AMPs. Finally, the current status, associated challenges, and future directions for the development of AMP therapeutics are discussed.
Collapse
Affiliation(s)
- Jianguo Li
- Ocular Chemistry and Anti-Infectives, Singapore Eye Research InstituteSingapore, Singapore
- Agency for Science, Technology and Research (ASTAR), Bioinformatics InstituteSingapore, Singapore
- Duke-NUS Graduate Medical School, SRP Neuroscience and BDSingapore, Singapore
| | - Jun-Jie Koh
- Ocular Chemistry and Anti-Infectives, Singapore Eye Research InstituteSingapore, Singapore
| | - Shouping Liu
- Ocular Chemistry and Anti-Infectives, Singapore Eye Research InstituteSingapore, Singapore
| | | | - Chandra S. Verma
- Ocular Chemistry and Anti-Infectives, Singapore Eye Research InstituteSingapore, Singapore
- Agency for Science, Technology and Research (ASTAR), Bioinformatics InstituteSingapore, Singapore
- Department of Biological Sciences, National University of SingaporeSingapore, Singapore
- School of Biological Sciences, Nanyang Technological UniversitySingapore, Singapore
| | - Roger W. Beuerman
- Ocular Chemistry and Anti-Infectives, Singapore Eye Research InstituteSingapore, Singapore
- Duke-NUS Graduate Medical School, SRP Neuroscience and BDSingapore, Singapore
| |
Collapse
|
13
|
Li NN, Li JZ, Liu P, Pranantyo D, Luo L, Chen JC, Kang ET, Hu XF, Li CM, Xu LQ. An antimicrobial peptide with an aggregation-induced emission (AIE) luminogen for studying bacterial membrane interactions and antibacterial actions. Chem Commun (Camb) 2017; 53:3315-3318. [DOI: 10.1039/c6cc09408b] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A fluorescence technique to investigate the interactions between bacterial membranes and an AIE luminogen-decorated antimicrobial peptide has been reported.
Collapse
Affiliation(s)
- Ning Ning Li
- Institute for Clean Energy and Advanced Materials
- Faculty of Materials and Energy
- Southwest University
- Chongqing
- China
| | - Jun Zhi Li
- Institute for Clean Energy and Advanced Materials
- Faculty of Materials and Energy
- Southwest University
- Chongqing
- China
| | - Peng Liu
- Department of Chemical & Biomolecular Engineering
- National University of Singapore
- Singapore
| | - Dicky Pranantyo
- Department of Chemical & Biomolecular Engineering
- National University of Singapore
- Singapore
| | - Lei Luo
- College of Pharmaceutical Science
- Southwest University
- Chongqing
- China
| | - Jiu Cun Chen
- Institute for Clean Energy and Advanced Materials
- Faculty of Materials and Energy
- Southwest University
- Chongqing
- China
| | - En-Tang Kang
- Department of Chemical & Biomolecular Engineering
- National University of Singapore
- Singapore
| | - Xue Feng Hu
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu
- China
| | - Chang Ming Li
- Institute for Clean Energy and Advanced Materials
- Faculty of Materials and Energy
- Southwest University
- Chongqing
- China
| | - Li Qun Xu
- Institute for Clean Energy and Advanced Materials
- Faculty of Materials and Energy
- Southwest University
- Chongqing
- China
| |
Collapse
|
14
|
Branched Peptide, B2088, Disrupts the Supramolecular Organization of Lipopolysaccharides and Sensitizes the Gram-negative Bacteria. Sci Rep 2016; 6:25905. [PMID: 27174567 PMCID: PMC4865820 DOI: 10.1038/srep25905] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/25/2016] [Indexed: 12/30/2022] Open
Abstract
Dissecting the complexities of branched peptide-lipopolysaccharides (LPS) interactions provide rationale for the development of non-cytotoxic antibiotic adjuvants. Using various biophysical methods, we show that the branched peptide, B2088, binds to lipid A and disrupts the supramolecular organization of LPS. The disruption of outer membrane in an intact bacterium was demonstrated by fluorescence spectroscopy and checkerboard assays, the latter confirming strong to moderate synergism between B2088 and various classes of antibiotics. The potency of synergistic combinations of B2088 and antibiotics was further established by time-kill kinetics, mammalian cell culture infections model and in vivo model of bacterial keratitis. Importantly, B2088 did not show any cytotoxicity to corneal epithelial cells for at least 96 h continuous exposure or hemolytic activity even at 20 mg/ml. Peptide congeners containing norvaline, phenylalanine and tyrosine (instead of valine in B2088) displayed better synergism compared to other substitutions. We propose that high affinity and subsequent disruption of the supramolecular assembly of LPS by the branched peptides are vital for the development of non-cytotoxic antibiotic adjuvants that can enhance the accessibility of conventional antibiotics to the intracellular targets, decrease the antibiotic consumption and holds promise in averting antibiotic resistance.
Collapse
|
15
|
Koh JJ, Zou H, Lin S, Lin H, Soh RT, Lim FH, Koh WL, Li J, Lakshminarayanan R, Verma C, Tan DTH, Cao D, Beuerman RW, Liu S. Nonpeptidic Amphiphilic Xanthone Derivatives: Structure-Activity Relationship and Membrane-Targeting Properties. J Med Chem 2015; 59:171-93. [PMID: 26681070 DOI: 10.1021/acs.jmedchem.5b01500] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We recently reported the bioinspired synthesis of a highly potent nonpeptidic xanthone, 2c (AM-0016), with potent antibacterial activity against MRSA. Herein, we report a thorough structure-activity relationship (SAR) analysis of a series of nonpeptidic amphiphilic xanthone derivatives in an attempt to identify more potent compounds with lower hemolytic activity and greater membrane selectivity. Forty-six amphiphilic xanthone derivatives were analyzed in this study and structurally classified into four groups based on spacer length, cationic moieties, lipophilic chains, and triarm functionalization. We evaluated and explored the effects of the structures on their membrane-targeting properties. The SAR analysis successfully identified 3a with potent MICs (1.56-3.125 μ/mL) and lower hemolytic activity (80.2 μg/mL for 3a versus 19.7 μg/mL for 2c). Compound 3a displayed a membrane selectivity of 25.7-50.4. Thus, 3a with improved HC50 value and promising selectivity could be used as a lead compound for further structural optimization for the treatment of MRSA infection.
Collapse
Affiliation(s)
- Jun-Jie Koh
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore , 119074 Singapore
| | - Hanxun Zou
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore.,School of Chemistry and Chemical Engineering, South China University of Technology , Guangzhou 510641, China
| | - Shuimu Lin
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore.,School of Chemistry and Chemical Engineering, South China University of Technology , Guangzhou 510641, China
| | - Huifen Lin
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore
| | - Rui Ting Soh
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore
| | - Fang Hui Lim
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore
| | - Wee Luan Koh
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore
| | - Jianguo Li
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore.,Bioinformatics Institute (A*STAR) , 30 Biopolis Street, 07-01 Matrix, 138671 Singapore
| | - Rajamani Lakshminarayanan
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore.,SRP Neuroscience and Behavioural Disorders, Duke-NUS Graduate Medical School , 169857 Singapore
| | - Chandra Verma
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore.,Bioinformatics Institute (A*STAR) , 30 Biopolis Street, 07-01 Matrix, 138671 Singapore.,School of Biological Sciences, Nanyang Technological University , 60 Nanyang Drive, 637551 Singapore.,Department of Biological Sciences, National University of Singapore , 14 Science Drive 4, 117543 Singapore
| | - Donald T H Tan
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore , 119074 Singapore.,Singapore National Eye Centre , 11 Third Hospital Avenue, 168751 Singapore
| | - Derong Cao
- School of Chemistry and Chemical Engineering, South China University of Technology , Guangzhou 510641, China
| | - Roger W Beuerman
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore.,SRP Neuroscience and Behavioural Disorders, Duke-NUS Graduate Medical School , 169857 Singapore
| | - Shouping Liu
- Singapore Eye Research Institute , The Academia, 20 College Road, Discovery Tower Level 6, 169856 Singapore.,SRP Neuroscience and Behavioural Disorders, Duke-NUS Graduate Medical School , 169857 Singapore
| |
Collapse
|
16
|
Saraswathi P, Beuerman RW. Corneal Biofilms: From Planktonic to Microcolony Formation in an Experimental Keratitis Infection with Pseudomonas Aeruginosa. Ocul Surf 2015. [DOI: 10.1016/j.jtos.2015.07.001 [pii]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Koh JJ, Lin H, Caroline V, Chew YS, Pang LM, Aung TT, Li J, Lakshminarayanan R, Tan DTH, Verma C, Tan AL, Beuerman RW, Liu S. N-Lipidated Peptide Dimers: Effective Antibacterial Agents against Gram-Negative Pathogens through Lipopolysaccharide Permeabilization. J Med Chem 2015. [PMID: 26214729 DOI: 10.1021/acs.jmedchem.5b00628] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Treating infections caused by multidrug-resistant Gram-negative pathogens is challenging, and there is concern regarding the toxicity of the most effective antimicrobials for Gram-negative pathogens. We hypothesized that conjugating a fatty acid moiety onto a peptide dimer could maximize the interaction with lipopolysaccharide (LPS) and facilitate the permeabilization of the LPS barrier, thereby improving potency against Gram-negative pathogens. We systematically designed a series of N-lipidated peptide dimers that are active against Gram-negative bacteria, including carbapenem-resistant Enterobacteriaceae (CRE). The optimized lipid length was 6-10 carbons. At these lipid lengths, the N-lipidated peptide dimers exhibited strong LPS permeabilization. Compound 23 exhibited synergy with select antibiotics in most of the combinations tested. 23 and 32 also displayed rapid bactericidal activity. Importantly, 23 and 32 were nonhemolytic at 10 mg/mL, with no cellular or in vivo toxicity. These characteristics suggest that these compounds can overcome the limitations of current Gram-negative-targeted antimicrobials such as polymyxin B.
Collapse
Affiliation(s)
- Jun-Jie Koh
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore , 119074, Singapore
| | - Huifen Lin
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore
| | - Vonny Caroline
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore
| | - Yu Siang Chew
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore
| | - Li Mei Pang
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore
| | - Thet Tun Aung
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore
| | - Jianguo Li
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore.,Bioinformatics Institute (A*STAR) , 30 Biopolis Street, 07-01 matrix, 138671, Singapore
| | - Rajamani Lakshminarayanan
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore.,SRP Neuroscience and Behavioural Disorders, Duke-NUS Medical School , 169857, Singapore
| | - Donald T H Tan
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore , 119074, Singapore.,Singapore National Eye Centre , 11 Third Hospital Avenue, 168751, Singapore
| | - Chandra Verma
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore.,Bioinformatics Institute (A*STAR) , 30 Biopolis Street, 07-01 matrix, 138671, Singapore.,School of Biological Sciences, Nanyang Technological University , 60 Nanyang Drive, 637551, Singapore.,Department of Biological Sciences, National University of Singapore , 14 Science Drive 4, 117543, Singapore
| | - Ai Ling Tan
- Department of Pathology, Singapore General Hospital , 169608, Singapore
| | - Roger W Beuerman
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore.,SRP Neuroscience and Behavioural Disorders, Duke-NUS Medical School , 169857, Singapore
| | - Shouping Liu
- Singapore Eye Research Institute, The Academia , 20 College Road, Discovery Tower Level 6, 169856, Singapore.,SRP Neuroscience and Behavioural Disorders, Duke-NUS Medical School , 169857, Singapore
| |
Collapse
|
18
|
Saraswathi P, Beuerman RW. Corneal Biofilms: From Planktonic to Microcolony Formation in an Experimental Keratitis Infection with Pseudomonas Aeruginosa. Ocul Surf 2015. [PMID: 26220579 DOI: 10.1016/j.jtos.2015.07.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
PURPOSE Microbial biofilms commonly comprise part of the infectious scenario, complicating the therapeutic approach. The purpose of this study was to determine in a mouse model of corneal infection if mature biofilms formed and to visualize the stages of biofilm formation. METHODS A bacterial keratitis model was established using Pseudomonas aeruginosa ATCC 9027 (1 × 10(8) CFU/ml) to infect the cornea of C57BL/6 black mouse. Eyes were examined post-infection (PI) on days 1, 2, 3, 5, and 7, and imaged by slit lamp microscopy, and light, confocal, and electron microscopy to identify the stages of biofilm formation and the time of appearance. RESULTS On PI day 1, Gram staining showed rod-shaped bacteria adherent on the corneal surface. On PI days 2 and 3, bacteria were seen within webs of extracellular polymeric substance (EPS) and glycocalyx secretion, imaged by confocal microscopy. Scanning electron microscopy demonstrated microcolonies of active infectious cells bound with thick fibrous material. Transmission electron microscopy substantiated the formation of classical biofilm architecture with P. aeruginosa densely packed within the extracellular polymeric substances on PI days 5 and 7. CONCLUSION Direct visual evidence showed that biofilms routinely developed on the biotic surface of the mouse cornea. The mouse model can be used to develop new approaches to deal therapeutically with biofilms in corneal infections.
Collapse
Affiliation(s)
| | - Roger W Beuerman
- Singapore Eye Research Institute (SERI), Singapore; Duke-NUS SRP Neuroscience and Behavioural Disorders and Emerging Infectious Diseases, Singapore; Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
19
|
Datta A, Ghosh A, Airoldi C, Sperandeo P, Mroue KH, Jiménez-Barbero J, Kundu P, Ramamoorthy A, Bhunia A. Antimicrobial Peptides: Insights into Membrane Permeabilization, Lipopolysaccharide Fragmentation and Application in Plant Disease Control. Sci Rep 2015; 5:11951. [PMID: 26144972 PMCID: PMC4491704 DOI: 10.1038/srep11951] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/09/2015] [Indexed: 11/09/2022] Open
Abstract
The recent increase in multidrug resistance against bacterial infections has become a major concern to human health and global food security. Synthetic antimicrobial peptides (AMPs) have recently received substantial attention as potential alternatives to conventional antibiotics because of their potent broad-spectrum antimicrobial activity. These peptides have also been implicated in plant disease control for replacing conventional treatment methods that are polluting and hazardous to the environment and to human health. Here, we report de novo design and antimicrobial studies of VG16, a 16-residue active fragment of Dengue virus fusion peptide. Our results reveal that VG16KRKP, a non-toxic and non-hemolytic analogue of VG16, shows significant antimicrobial activity against Gram-negative E. coli and plant pathogens X. oryzae and X. campestris, as well as against human fungal pathogens C. albicans and C. grubii. VG16KRKP is also capable of inhibiting bacterial disease progression in plants. The solution-NMR structure of VG16KRKP in lipopolysaccharide features a folded conformation with a centrally located turn-type structure stabilized by aromatic-aromatic packing interactions with extended N- and C-termini. The de novo design of VG16KRKP provides valuable insights into the development of more potent antibacterial and antiendotoxic peptides for the treatment of human and plant infections.
Collapse
Affiliation(s)
- Aritreyee Datta
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700 054, India
| | - Anirban Ghosh
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700 054, India
| | - Cristina Airoldi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, P.zza della Scienza 2, 2016 Milano, Italy
| | - Paola Sperandeo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, P.zza della Scienza 2, 2016 Milano, Italy
| | - Kamal H Mroue
- Biophysics and Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109-1055, USA
| | - Jesús Jiménez-Barbero
- 1] Infectious Diseases Program, CIC bioGUNE, Parque Tecnologico de Bizkaia, Building 801A, 48160 Derio, Spain [2] IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Pallob Kundu
- Division of Plant Biology, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700 054, India
| | - Ayyalusamy Ramamoorthy
- Biophysics and Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109-1055, USA
| | - Anirban Bhunia
- 1] Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII (M), Kolkata 700 054, India [2] Biophysics and Department of Chemistry, University of Michigan, 930 N. University Avenue, Ann Arbor, Michigan 48109-1055, USA
| |
Collapse
|
20
|
Koh JJ, Lin S, Aung TT, Lim F, Zou H, Bai Y, Li J, Lin H, Pang LM, Koh WL, Salleh SM, Lakshminarayanan R, Zhou L, Qiu S, Pervushin K, Verma C, Tan DTH, Cao D, Liu S, Beuerman RW. Amino acid modified xanthone derivatives: novel, highly promising membrane-active antimicrobials for multidrug-resistant Gram-positive bacterial infections. J Med Chem 2014; 58:739-52. [PMID: 25474410 DOI: 10.1021/jm501285x] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antibiotic resistance is a critical global health care crisis requiring urgent action to develop more effective antibiotics. Utilizing the hydrophobic scaffold of xanthone, we identified three components that mimicked the action of an antimicrobial cationic peptide to produce membrane-targeting antimicrobials. Compounds 5c and 6, which contain a hydrophobic xanthone core, lipophilic chains, and cationic amino acids, displayed very promising antimicrobial activity against multidrug-resistant Gram-positive bacteria, including MRSA and VRE, rapid time-kill, avoidance of antibiotic resistance, and low toxicity. The bacterial membrane selectivity of these molecules was comparable to that of several membrane-targeting antibiotics in clinical trials. 5c and 6 were effective in a mouse model of corneal infection by S. aureus and MRSA. Evidence is presented indicating that 5c and 6 target the negatively charged bacterial membrane via a combination of electrostatic and hydrophobic interactions. These results suggest that 5c and 6 have significant promise for combating life-threatening infections.
Collapse
Affiliation(s)
- Jun-Jie Koh
- Singapore Eye Research Institute , 11 Third Hospital Avenue, 168751, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Patent Highlights. Pharm Pat Anal 2014. [DOI: 10.4155/ppa.14.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development
Collapse
|
22
|
Ravi HK, Stach M, Soares TA, Darbre T, Reymond JL, Cascella M. Electrostatics and flexibility drive membrane recognition and early penetration by the antimicrobial peptide dendrimer bH1. Chem Commun (Camb) 2014; 49:8821-3. [PMID: 23959139 DOI: 10.1039/c3cc44912b] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Molecular dynamics simulations of the polycationic antimicrobial peptide dendrimer (Leu)8(DapLeu)4(DapPhe)2DapLys-NH2 binding to membranes suggest that electrostatic interactions with the polyanionic lipopolysaccharide (LPS) and conformational flexibility of the 2,3-diaminopropanoic acid (Dap) branching units drive its selective insertion into microbial membranes.
Collapse
Affiliation(s)
- Harish K Ravi
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland.
| | | | | | | | | | | |
Collapse
|
23
|
Dimeric unnatural polyproline-rich peptides with enhanced antibacterial activity. Bioorg Med Chem Lett 2013; 24:556-9. [PMID: 24365160 DOI: 10.1016/j.bmcl.2013.12.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 11/24/2022]
Abstract
We report a dimerization strategy to enhance the antibacterial potency of an otherwise weak cationic amphiphilic polyproline helical (CAPH) peptide. Overall, the dimeric CAPHs were more active against Escherichia coli and Staphylococcus aureus than the monomeric counterpart, reaching up to a 60-fold increase in potency. At their minimum inhibitory concentration (MIC), the dimeric peptides demonstrated no hemolytic activity or bacterial membrane disruption as monitored by β-galactosidase release in E. coli. At higher concentrations the dimeric agents were found to induce β-galactosidase release, but maintained negligible hemolytic activity, pointing to a potential shift in the mechanism of action at higher concentrations. Thus, discontinuous dimerization of an unnatural proline-rich peptide was a successful strategy to create potent de novo antibacterial peptides without membrane lysis.
Collapse
|
24
|
Torcato IM, Huang YH, Franquelim HG, Gaspar DD, Craik DJ, Castanho MARB, Henriques ST. The antimicrobial activity of Sub3 is dependent on membrane binding and cell-penetrating ability. Chembiochem 2013; 14:2013-22. [PMID: 24038773 DOI: 10.1002/cbic.201300274] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Indexed: 12/21/2022]
Abstract
Because of their high activity against microorganisms and low cytotoxicity, cationic antimicrobial peptides (AMPs) have been explored as the next generation of antibiotics. Although they have common structural features, the modes of action of AMPs are extensively debated, and a single mechanism does not explain the activity of all AMPs reported so far. Here we investigated the mechanism of action of Sub3, an AMP previously designed and optimised from high-throughput screening with bactenecin as the template. Sub3 has potent activity against Gram-negative and Gram-positive bacteria as well as against fungi, but its mechanism of action has remained elusive. By using AFM imaging, ζ potential, flow cytometry and fluorescence methodologies with model membranes and bacterial cells, we found that, although the mechanism of action involves membrane targeting, Sub3 internalises inside bacteria at lethal concentrations without permeabilising the membrane, thus suggesting that its antimicrobial activity might involve both the membrane and intracellular targets. In addition, we found that Sub3 can be internalised into human cells without being toxic. As some bacteria are able to survive intracellularly and consequently evade host defences and antibiotic treatment, our findings suggest that Sub3 could be useful as an intracellular antimicrobial agent for infections that are notoriously difficult to treat.
Collapse
Affiliation(s)
- Inês M Torcato
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon (Portugal)
| | | | | | | | | | | | | |
Collapse
|
25
|
Li J, Lakshminarayanan R, Bai Y, Liu S, Zhou L, Pervushin K, Verma C, Beuerman RW. Molecular dynamics simulations of a new branched antimicrobial peptide: a comparison of force fields. J Chem Phys 2013; 137:215101. [PMID: 23231260 DOI: 10.1063/1.4768899] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Branched antimicrobial peptides are promising as a new class of antibiotics displaying high activity and low toxicity and appear to work through a unique mechanism of action. We explore the structural dynamics of a covalently branched 18 amino acid peptide (referred to as B2088) in aqueous and membrane mimicking environments through molecular dynamics (MD) simulations. Towards this, we carry out conventional MD simulations and supplement these with replica exchange simulations. The simulations are carried out using four different force fields that are commonly employed for simulating biomolecular systems. These force fields are GROMOS53a6, CHARMM27 with cMAP, CHARMM27 without cMAP and AMBER99sb. The force fields are benchmarked against experimental data available from circular dichroism and nuclear magnetic resonance spectroscopies, and show that CHARMM27 without cMAP correction is the most successful in reproducing the structural dynamics of B2088 both in water and in the presence of micelles. Although the four force fields predict different structures of B2088, they all show that B2088 stabilizes against the head group of the lipid through hydrogen bonding of its Lys and Arg side chains. This leads us to hypothesize that B2088 is unlikely to penetrate into the hydrophobic region of the membrane owing to the high free energy costs of transfer from water, and possibly acts by carpeting and thus disrupting the membrane.
Collapse
Affiliation(s)
- Jianguo Li
- Singapore Eye Research Institute, 11 Third Hospital Avenue, #06-00, Singapore 168751.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Zou H, Koh JJ, Li J, Qiu S, Aung TT, Lin H, Lakshminarayanan R, Dai X, Tang C, Lim FH, Zhou L, Tan AL, Verma C, Tan DTH, Chan HSO, Saraswathi P, Cao D, Liu S, Beuerman RW. Design and Synthesis of Amphiphilic Xanthone-Based, Membrane-Targeting Antimicrobials with Improved Membrane Selectivity. J Med Chem 2013; 56:2359-73. [DOI: 10.1021/jm301683j] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hanxun Zou
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
- School of Chemistry and Chemical
Engineering, State Key Lab of Luminescent Materials and Devices, South
China University of Technology, Guangzhou 510641, China
| | - Jun-Jie Koh
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
- Department of Ophthalmology, Yong
Loo Lin School of Medicine, National University of Singapore, Singapore
119074, Singapore
| | - Jianguo Li
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
- Bioinformatics
Institute, Singapore
138671, Singapore
| | - Shengxiang Qiu
- Program
for Natural Products
Chemical Biology, Key Laboratory of Plant Resources Conservation and
Sustainable Utilization, South China Botanical Garden, the Chinese
Academy of Sciences, Guangzhou, China
| | - Thet Tun Aung
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
| | - Huifen Lin
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
| | - Rajamani Lakshminarayanan
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
- Duke-NUS Medical School, SRP
Neuroscience and Behavioral Disorders, Singapore 169857, Singapore
| | - Xiaoping Dai
- Program
for Natural Products
Chemical Biology, Key Laboratory of Plant Resources Conservation and
Sustainable Utilization, South China Botanical Garden, the Chinese
Academy of Sciences, Guangzhou, China
| | - Charles Tang
- Department of Pathology, Singapore
General Hospital, Singapore 169608, Singapore
| | - Fang Hui Lim
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
- Department of Chemistry, National
University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore
| | - Lei Zhou
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
| | - Ai Ling Tan
- Department of Pathology, Singapore
General Hospital, Singapore 169608, Singapore
| | - Chandra Verma
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
- Bioinformatics
Institute, Singapore
138671, Singapore
| | - Donald T. H. Tan
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
- Department of Ophthalmology, Yong
Loo Lin School of Medicine, National University of Singapore, Singapore
119074, Singapore
| | - Hardy Sze On Chan
- Department of Chemistry, National
University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore
| | | | - Derong Cao
- School of Chemistry and Chemical
Engineering, State Key Lab of Luminescent Materials and Devices, South
China University of Technology, Guangzhou 510641, China
| | - Shouping Liu
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
- Duke-NUS Medical School, SRP
Neuroscience and Behavioral Disorders, Singapore 169857, Singapore
| | - Roger W. Beuerman
- Singapore
Eye Research Institute,
11 Third Hospital Avenue, Singapore 168751, Singapore
- Duke-NUS Medical School, SRP
Neuroscience and Behavioral Disorders, Singapore 169857, Singapore
| |
Collapse
|
27
|
Li J, Liu S, Lakshminarayanan R, Bai Y, Pervushin K, Verma C, Beuerman RW. Molecular simulations suggest how a branched antimicrobial peptide perturbs a bacterial membrane and enhances permeability. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1112-21. [PMID: 23274275 DOI: 10.1016/j.bbamem.2012.12.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 12/03/2012] [Accepted: 12/18/2012] [Indexed: 11/29/2022]
Abstract
A covalently, branched antimicrobial peptide (BAMP) B2088 demonstrating enhanced antimicrobial effects and without additional toxicity when compared to its linear counterpart, has been developed. Atomistic molecular dynamics simulations have been used to investigate the mode of interaction of B2088 with model bacterial and mammalian membranes. These simulations suggest that both long-range electrostatic interactions and short-range hydrogen bonding play important roles in steering B2088 toward the negatively charged membranes. The reason why B2088 is selective towards the bacterial membrane is postulated to be the greater density of negative charges on the bacterial membrane which enables rapid accumulation of B2088 on the bacterial membrane to a high surface concentration, stabilizing it through excess hydrogen bond formation. The majority of hydrogen bonds are seen between the side chains of the basic residues (Arg or Lys) with the PO4 groups of lipids. In particular, formation of the bidentate hydrogen bonds between the guanidinium group of Arg and PO4 groups are found to be more favorable, both geometrically and energetically. Moreover, the planar gaunidinium group and its hydrophobic character enable the Arg side chains to solvate into the hydrophobic membrane. Structural perturbation of the bacterial membrane is found to be concentration dependent and is significant at higher concentrations of B2088, resulting in a large number of water translocations across the bacterial membrane. These simulations enhance our understanding of the action mechanism of a covalently branched antimicrobial peptide with model membranes and provide practical guidance for the design of new antimicrobial peptides.
Collapse
Affiliation(s)
- Jianguo Li
- Singapore Eye Research Institute, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
28
|
Nishi K, Chijiishi M, Katsumoto Y, Nakao T, Fujii K, Chung UI, Noguchi H, Sakai T, Shibayama M. Rubber elasticity for incomplete polymer networks. J Chem Phys 2012; 137:224903. [DOI: 10.1063/1.4769829] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|