1
|
Sanchez C, Vargas-Cuebas GG, Michaud ME, Allen RA, Morrison-Lewis KR, Siddiqui S, Minbiole KPC, Wuest WM. Highly Effective Biocides against Pseudomonas aeruginosa Reveal New Mechanistic Insights Across Gram-Negative Bacteria. ACS Infect Dis 2024; 10:3868-3879. [PMID: 39440866 PMCID: PMC11555683 DOI: 10.1021/acsinfecdis.4c00433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/28/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Pseudomonas aeruginosa is a major nosocomial pathogen that persists in healthcare settings despite rigorous disinfection protocols due to intrinsic mechanisms conferring resistance. We sought to systematically assess cationic biocide efficacy against this pathogen using a panel of multidrug-resistant P. aeruginosa clinical isolates. Our studies revealed widespread resistance to commercial cationic disinfectants that are the current standard of care, raising concerns about their efficacy. To address this shortcoming, we highlight a new class of quaternary phosphonium compounds that are highly effective against all members of the panel. To understand the difference in efficacy, mechanism of action studies were carried out, which identified a discrete inner-membrane selective target. Resistance selection studies implicated the SmvRA efflux system (a transcriptionally regulated, inner membrane-associated efflux system) as a major determinant of resistance. This system is also implicated in resistance to two commercial bolaamphiphile antiseptics, octenidine and chlorhexidine, which was further validated herein. In sum, this work highlights, for the first time, a discrete inner-membrane specific mechanism for the bolaamphiphile class of disinfectants that contrasts with the prevailing model of indiscriminate membrane interactions of commercial amphiphiles paving the way for future innovations in disinfectant research.
Collapse
Affiliation(s)
- Christian
A. Sanchez
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Germán G. Vargas-Cuebas
- Department
of Microbiology and Immunology, Emory University, Atlanta, Georgia 30322, United States
| | - Marina E. Michaud
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Ryan A. Allen
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | | | - Shehreen Siddiqui
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Kevin P. C. Minbiole
- Department
of Chemistry, Villanova University, Villanova, Pennsylvania 19085, United States
| | - William M. Wuest
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
2
|
Jaikishan S, Lavainne M, Ravald HK, Scobbie K, Dusa F, Maheswari R, Turpeinen J, Eikemans I, Chen R, Rantala J, Aseyev V, Maier NN, Wiedmer SK. Fragment-based approach to study fungicide-biomimetic membrane interactions. SOFT MATTER 2024. [PMID: 39012330 DOI: 10.1039/d4sm00648h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
In this study, the molecular interactions of the allylamine-type fungicide butenafine and a set of substructures ("fragments") with liposomes mimicking biological membranes were studied to gain a better understanding of the structural factors governing membrane affinity and perturbation. Specifically, drug/fragment-membrane interactions were investigated using an interdisciplinary approach involving micro differential scanning calorimetry, open-tubular capillary electrochromatography, nanoplasmonic sensing, and quartz crystal microbalance. By incubating the drug and the fragment compounds with liposomes with varying lipid composition or by externally adding the compounds to preformed liposomes, a detailed mechanistic picture on the underlying drug/fragment-membrane interactions was obtained. The nature and the degree of ionisation of polar head groups of the lipids had a major influence on the nature of drug-membrane interactions, and so had the presence and relative concentration of cholesterol within the membranes. The in-depth understanding of drug/fragment-membranes interactions established by the presented interdisciplinary fragment-based approach may be useful in guiding the design and early-stage evaluation of prospective antifungal drug candidates, and the discovery of agents with improved membrane penetrating characteristics in general.
Collapse
Affiliation(s)
- Shishir Jaikishan
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Marine Lavainne
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Henri K Ravald
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Kieran Scobbie
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Filip Dusa
- Institute of Analytical Chemistry, Czech Academy of Sciences, Veveří 97, Brno 60200, Czech Republic
| | - Rekha Maheswari
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Jenni Turpeinen
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Ian Eikemans
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Rui Chen
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Julia Rantala
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Vladimir Aseyev
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Norbert N Maier
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| | - Susanne K Wiedmer
- Department of Chemistry, A.I. Virtasen aukio 1, POB 55, 00014 University of Helsinki, Finland.
| |
Collapse
|
3
|
Tang Z, Feng J, Challa M, Rowthu SR, Xiong S, Zou C, Li J, Verma CS, Peng H, He X, Huang C, He Y. Discovery of novel Thymol-TPP antibiotics that eradicate MRSA persisters. Eur J Med Chem 2024; 270:116381. [PMID: 38604097 DOI: 10.1016/j.ejmech.2024.116381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
The high prevalence of methicillin-resistant Staphylococcus aureus (MRSA) strains and the formation of non-growing, dormant "persisters" subsets help bacteria evade antibiotic treatment and enhance bacterial resistance, which poses a serious threat to human life and health. It is urgent to discover novel antibacterial therapies effective against MRSA persisters. Thymol is a common nutraceutical with weak antibacterial and antitumor activities. A series of Thymol triphenylphosphine (TPP) conjugates (TPP-Thy3) was designed and synthesized. These compounds showed significantly improved inhibitory activity against Gram-positive bacteria compared with Thymol. Among them, Thy3d displayed a low probability of resistance selection and showed excellent biocompatibility. Interestingly, Thy3d elicited a rapid killing effect of MRSA persisters (99.999%) at high concentration. Fluorescence experiments, electron microscopy, molecular dynamics simulation and bilayer experiment confirmed that Thy3d conjugates exerted potent antimicrobial activity by disrupting the integrity of the membrane of bacterial even the persister. Furthermore, Thy3d exhibited considerable efficacy in a mouse model of subcutaneous murine MRSA infection. In summary, TPP-Thy3 conjugates are a series of novel antibacterial agents and could serve as a new therapeutic strategy for combating antibiotic resistance.
Collapse
Affiliation(s)
- Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Jizhou Feng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Mahesh Challa
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Sankara Rao Rowthu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Shuxin Xiong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Cheng Zou
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Jianguo Li
- Singapore Eye Research Institute, Singapore, 169856, Singapore; Bioinformatics Institute, A*STAR, 30 Biopolis Street, Matrix, 138671, Singapore
| | - Chandra Shekhar Verma
- Bioinformatics Institute, A*STAR, 30 Biopolis Street, Matrix, 138671, Singapore; Department of Biological Sciences, National University of Singapore, 117543, Singapore; School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Haibo Peng
- Chongqing Academy of Science and Technology, Chongqing, 401123, China
| | - Xiaoli He
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Chao Huang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China.
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China; BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
4
|
Tang Z, Jiang W, Li S, Huang X, Yang Y, Chen X, Qiu J, Xiao C, Xie Y, Zhang X, Li J, Verma CS, He Y, Yang A. Design and evaluation of tadpole-like conformational antimicrobial peptides. Commun Biol 2023; 6:1177. [PMID: 37980400 PMCID: PMC10657444 DOI: 10.1038/s42003-023-05560-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 11/08/2023] [Indexed: 11/20/2023] Open
Abstract
Antimicrobial peptides are promising alternatives to conventional antibiotics. Herein, we report a class of "tadpole-like" peptides consisting of an amphipathic α-helical head and an aromatic tail. A structure-activity relationship (SAR) study of "tadpole-like" temporin-SHf and its analogs revealed that increasing the number of aromatic residues in the tail, introducing Arg to the α-helical head and rearranging the peptide topology dramatically increased antimicrobial activity. Through progressive structural optimization, we obtained two peptides, HT2 and RI-HT2, which exhibited potent antimicrobial activity, no hemolytic activity and cytotoxicity, and no propensity to induce resistance. NMR and molecular dynamics simulations revealed that both peptides indeed adopted "tadpole-like" conformations. Fluorescence experiments and electron microscopy confirmed the membrane targeting mechanisms of the peptides. Our studies not only lead to the discovery of a series of ultrashort peptides with potent broad-spectrum antimicrobial activities, but also provide a new strategy for rational design of novel "tadpole-like" antimicrobial peptides.
Collapse
Affiliation(s)
- Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Wuqiao Jiang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Shuangli Li
- National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xue Huang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Yi Yang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Xiaorong Chen
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Jingyi Qiu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Chuyu Xiao
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Ying Xie
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Xu Zhang
- National Centre for Magnetic Resonance in Wuhan, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jianguo Li
- Bioinformatics Institute, A∗STAR, 30 Biopolis Street, Matrix, Singapore, 138671, Singapore
- Singapore Eye Research Institute, Singapore, 169856, Singapore
| | - Chandra Shekhar Verma
- Bioinformatics Institute, A∗STAR, 30 Biopolis Street, Matrix, Singapore, 138671, Singapore
- Department of Biological Sciences, National University of, Singapore, 117543, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Yun He
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China.
- BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China.
| |
Collapse
|
5
|
Gonçalves RA, Holmberg K, Lindman B. Cationic surfactants: A review. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2023.121335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
6
|
Vemuri GN, Hughes JR, Iovine PM. Synthesis and characterization of terpene-derived cationic bolaamphiphiles. Tetrahedron 2022. [DOI: 10.1016/j.tet.2022.133008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
7
|
Li N, Liu X, Zhang M, Zhang Z, Zhang B, Wang X, Wang J, Tu P, Jiang Y, Shi SP. Characterization of a coumarin C-/ O-prenyltransferase and a quinolone C-prenyltransferase from Murraya exotica. Org Biomol Chem 2022; 20:5535-5542. [PMID: 35788620 DOI: 10.1039/d2ob01054b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prenyltransferases (PTs) play important roles in the biosynthesis and structural diversification of natural products. In the present study, two new PTs were characterized from a medicinal plant Murraya exotica. MePT1 unprecedentedly catalyses the formation of two C-geranylated products 8/6-C-geranylumbelliferone together with a trace product 7-O-geranylumbelliferone from umbelliferone. MePT2 regio-specifically catalyses the formation of C-3 dimethylallylated products from quinolone alkaloids. This is the first report that a plant PT catalyses the simultaneous formation of C- and O-prenylated products, and a plant PT specifically utilizes quinolone alkaloids as prenyl acceptors. The results not only provide important insight into the functional diversity of plant PTs and the biosynthesis of the prenylated coumarins, quinolone and carbazole alkaloids in Murraya plants, but also pave the way for the overproduction of the prenylated coumarins and alkaloids using metabolic engineering approaches.
Collapse
Affiliation(s)
- Na Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100102, China. .,School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Xiao Liu
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Mingliang Zhang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100102, China. .,School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Zekun Zhang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100102, China. .,School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Beibei Zhang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100102, China. .,School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Xiaohui Wang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Juan Wang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100102, China. .,State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| | - She-Po Shi
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100102, China.
| |
Collapse
|
8
|
MacDermott-Opeskin HI, Gupta V, O’Mara ML. Lipid-mediated antimicrobial resistance: a phantom menace or a new hope? Biophys Rev 2022; 14:145-162. [PMID: 35251360 PMCID: PMC8880301 DOI: 10.1007/s12551-021-00912-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/14/2021] [Indexed: 02/06/2023] Open
Abstract
Abstract The proposition of a post-antimicrobial era is all the more realistic with the continued rise of antimicrobial resistance. The development of new antimicrobials is failing to counter the ever-increasing rates of bacterial antimicrobial resistance. This necessitates novel antimicrobials and drug targets. The bacterial cell membrane is an essential and highly conserved cellular component in bacteria and acts as the primary barrier for entry of antimicrobials into the cell. Although previously under-exploited as an antimicrobial target, the bacterial cell membrane is attractive for the development of novel antimicrobials due to its importance in pathogen viability. Bacterial cell membranes are diverse assemblies of macromolecules built around a central lipid bilayer core. This lipid bilayer governs the overall membrane biophysical properties and function of its membrane-embedded proteins. This mini-review will outline the mechanisms by which the bacterial membrane causes and controls resistance, with a focus on alterations in the membrane lipid composition, chemical modification of constituent lipids, and the efflux of antimicrobials by membrane-embedded efflux systems. Thorough insight into the interplay between membrane-active antimicrobials and lipid-mediated resistance is needed to enable the rational development of new antimicrobials. In particular, the union of computational approaches and experimental techniques for the development of innovative and efficacious membrane-active antimicrobials is explored.
Collapse
Affiliation(s)
- Hugo I. MacDermott-Opeskin
- Research School of Chemistry, College of Science, The Australian National University, Canberra, ACT 2601 Australia
| | - Vrinda Gupta
- Research School of Chemistry, College of Science, The Australian National University, Canberra, ACT 2601 Australia
| | - Megan L. O’Mara
- Research School of Chemistry, College of Science, The Australian National University, Canberra, ACT 2601 Australia
| |
Collapse
|
9
|
Marine Origin Ligands of Nicotinic Receptors: Low Molecular Compounds, Peptides and Proteins for Fundamental Research and Practical Applications. Biomolecules 2022; 12:biom12020189. [PMID: 35204690 PMCID: PMC8961598 DOI: 10.3390/biom12020189] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
The purpose of our review is to briefly show what different compounds of marine origin, from low molecular weight ones to peptides and proteins, offer for understanding the structure and mechanism of action of nicotinic acetylcholine receptors (nAChRs) and for finding novel drugs to combat the diseases where nAChRs may be involved. The importance of the mentioned classes of ligands has changed with time; a protein from the marine snake venom was the first excellent tool to characterize the muscle-type nAChRs from the electric ray, while at present, muscle and α7 receptors are labeled with the radioactive or fluorescent derivatives prepared from α-bungarotoxin isolated from the many-banded krait. The most sophisticated instruments to distinguish muscle from neuronal nAChRs, and especially distinct subtypes within the latter, are α-conotoxins. Such information is crucial for fundamental studies on the nAChR revealing the properties of their orthosteric and allosteric binding sites and mechanisms of the channel opening and closure. Similar data are provided by low-molecular weight compounds of marine origin, but here the main purpose is drug design. In our review we tried to show what has been obtained in the last decade when the listed classes of compounds were used in the nAChR research, applying computer modeling, synthetic analogues and receptor mutants, X-ray and electron-microscopy analyses of complexes with the nAChRs, and their models which are acetylcholine-binding proteins and heterologously-expressed ligand-binding domains.
Collapse
|
10
|
Bekbossynova A, Zharylgap A, Filchakova O. Venom-Derived Neurotoxins Targeting Nicotinic Acetylcholine Receptors. Molecules 2021; 26:molecules26113373. [PMID: 34204855 PMCID: PMC8199771 DOI: 10.3390/molecules26113373] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 01/02/2023] Open
Abstract
Acetylcholine was the first neurotransmitter described. The receptors targeted by acetylcholine are found within organisms spanning different phyla and position themselves as very attractive targets for predation, as well as for defense. Venoms of snakes within the Elapidae family, as well as those of marine snails within the Conus genus, are particularly rich in proteins and peptides that target nicotinic acetylcholine receptors (nAChRs). Such compounds are invaluable tools for research seeking to understand the structure and function of the cholinergic system. Proteins and peptides of venomous origin targeting nAChR demonstrate high affinity and good selectivity. This review aims at providing an overview of the toxins targeting nAChRs found within venoms of different animals, as well as their activities and the structural determinants important for receptor binding.
Collapse
|
11
|
Torres MDT, Cao J, Franco OL, Lu TK, de la Fuente-Nunez C. Synthetic Biology and Computer-Based Frameworks for Antimicrobial Peptide Discovery. ACS NANO 2021; 15:2143-2164. [PMID: 33538585 PMCID: PMC8734659 DOI: 10.1021/acsnano.0c09509] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Antibiotic resistance is one of the greatest challenges of our time. This global health problem originated from a paucity of truly effective antibiotic classes and an increased incidence of multi-drug-resistant bacterial isolates in hospitals worldwide. Indeed, it has been recently estimated that 10 million people will die annually from drug-resistant infections by the year 2050. Therefore, the need to develop out-of-the-box strategies to combat antibiotic resistance is urgent. The biological world has provided natural templates, called antimicrobial peptides (AMPs), which exhibit multiple intrinsic medical properties including the targeting of bacteria. AMPs can be used as scaffolds and, via engineering, can be reconfigured for optimized potency and targetability toward drug-resistant pathogens. Here, we review the recent development of tools for the discovery, design, and production of AMPs and propose that the future of peptide drug discovery will involve the convergence of computational and synthetic biology principles.
Collapse
Affiliation(s)
- Marcelo D T Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jicong Cao
- Synthetic Biology Group, MIT Synthetic Biology Center, Department of Biological Engineering and Electrical Engineering and Computer Science, Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Octavio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, DF 70790160, Brazil
- S-inova Biotech, Universidade Católica Dom Bosco, Campo Grande, MS 79117010, Brazil
| | - Timothy K Lu
- Synthetic Biology Group, MIT Synthetic Biology Center, Department of Biological Engineering and Electrical Engineering and Computer Science, Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - 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, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
12
|
Hughes JR, Miller AS, Wallace CE, Vemuri GN, Iovine PM. Biomedically Relevant Applications of Bolaamphiphiles and Bolaamphiphile-Containing Materials. Front Chem 2021; 8:604151. [PMID: 33553103 PMCID: PMC7855593 DOI: 10.3389/fchem.2020.604151] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/15/2020] [Indexed: 12/28/2022] Open
Abstract
Bolaamphiphiles (BAs) are structurally segmented molecules with rich assembly characteristics and diverse physical properties. Interest in BAs as standalone active agents or as constituents of more complex therapeutic formulations has increased substantially in recent years. The preorganized amphiphilicity of BAs allows for a range of biological activities including applications that rely on multivalency. This review summarizes BA-related research in biomedically relevant areas. In particular, we review BA-related literature in four areas: gene delivery, antimicrobial materials, hydrogels, and prodrugs. We also discuss several distinguishing characteristics of BAs that impact their utility as biomedically relevant compounds.
Collapse
Affiliation(s)
| | | | | | | | - Peter M. Iovine
- Department of Chemistry and Biochemistry, University of San Diego, San Diego, CA, United States
| |
Collapse
|
13
|
Lin S, Wade JD, Liu S. De Novo Design of Flavonoid-Based Mimetics of Cationic Antimicrobial Peptides: Discovery, Development, and Applications. Acc Chem Res 2021; 54:104-119. [PMID: 33346639 DOI: 10.1021/acs.accounts.0c00550] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The alarming rate at which pathogens are developing resistance to conventional antibiotics represents one of the major global challenges to public health care. The prevalence of multidrug-resistant microorganisms is a major impetus for the discovery and development of new antimicrobials. Nature has, to date, been the source of most of the antibiotics discovered and used, including cationic antimicrobial peptides (CAMPs). CAMPs are key components of the innate immune system that are widely found in humans, animals, plants, and microorganisms and that serve as a first line of defense for the host. The attractive features of CAMPs have led to their recognition as potential new antimicrobials. However, they possess several inherent flaws that limit their clinical application including low stability, poor oral bioavailability, poor in vivo efficacy, and a high production cost. To address these issues, small molecule-based peptidomimetic antimicrobials have been designed to biomimic the structural features and biological function of CAMPs. Plant-derived flavonoids (e.g., xanthones and flavones) are active components in traditional herbal medicines and have been reported to contain a variety of significant pharmacological actions including antibacterial, antiviral, antioxidant, and anticancer activities. Over the past decade, we have developed a new chemical strategy to design, discover, and develop xanthone- or flavone-based peptidomimetics and have designed, synthesized, and biologically evaluated a library of approximately 450 new xanthone or flavone derivatives. The designed, structurally diverse compounds can be generally classified into two subfamilies, namely, peptidic and nonpeptidic amphiphilic xanthone or flavone derivatives. In this Account, we describe our efforts on the design, synthesis, biological property evaluation, and mechanism of action model studies of synthetic mimics of CAMPs. The flavonoid compounds are an important component of these rationally designed mimics because they function as hydrophobic aromatic moieties conjugated with different length lipid moieties, behave like an unnatural hydrophobic residue, and provide a rigid scaffold, with the reduced conformational flexibility more likely to provide an active conformation. The mimics can effectively disrupt the integrity of the bacterial membranes. Our endeavors encompass design principles, chemical synthesis, in vitro screening, structural optimization, extensive structural-activity relationship analysis, and a mechanism of action study through biophysical technologies including NMR spectroscopy techniques and computer dynamics simulations, drug resistance assays, in vivo pharmaceutical kinetics (PK) analyses, and in vivo efficacy evaluations of selected promising compounds against drug-resistant bacteria and fungi. Our major contributions to the discovery and development of flavonoid-based mimics as antimicrobials include effectively addressing several limitations associated with CAMPs and have led to promising compounds with a notable potential for further development as new therapeutic antimicrobial agents for the treatment of drug-resistant bacteria- or fungi-induced infections.
Collapse
Affiliation(s)
- Shuimu Lin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - John D. Wade
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria 3010, Australia
- School of Chemistry, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Shouping Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| |
Collapse
|
14
|
Morini M, Pedroni V, Alarcón L, Verde A, Mendioroz P, Appignanesi G, Sierra M. Interaction of hydroxy-xanthones with phosphatidylcholines: The effector decreases compressibility and increases the fluidity of membranes. Chem Phys Lipids 2020; 231:104938. [DOI: 10.1016/j.chemphyslip.2020.104938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 06/16/2020] [Accepted: 06/22/2020] [Indexed: 11/16/2022]
|
15
|
Hotarat W, Nutho B, Wolschann P, Rungrotmongkol T, Hannongbua S. Delivery of Alpha-Mangostin Using Cyclodextrins through a Biological Membrane: Molecular Dynamics Simulation. Molecules 2020; 25:molecules25112532. [PMID: 32485931 PMCID: PMC7321106 DOI: 10.3390/molecules25112532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
α-Mangostin (MGS) exhibits various pharmacological activities, including antioxidant, anticancer, antibacterial, and anti-inflammatory properties. However, its low water solubility is the major obstacle for its use in pharmaceutical applications. To increase the water solubility of MGS, complex formation with beta-cyclodextrins (βCDs), particularly with the native βCD and/or its derivative 2,6-dimethyl-β-CD (DMβCD) is a promising technique. Although there have been several reports on the adsorption of βCDs on the lipid bilayer, the release of the MGS/βCDs inclusion complex through the biological membrane remains unclear. In this present study, the release the MGS from the two different βCDs (βCD and DMβCD) across the lipid bilayer was investigated. Firstly, the adsorption of the free MGS, free βCDs, and inclusion complex formation was studied by conventional molecular dynamics simulation. The MGS in complex with those two βCDs was able to spontaneously release free MGS into the inner membrane. However, both MGS and DMβCD molecules potentially permeated into the deeper region of the interior membrane, whereas βCD only adsorbed at the outer membrane surface. The interaction between secondary rim of βCD and the 1-palmitoeyl-2-oleoyl-glycero-3-phosphocholine (POPC) phosphate groups showed the highest number of hydrogen bonds (up to 14) corresponding to the favorable location of βCD on the POPC membrane. Additionally, the findings suggested that electrostatic energy was the main driving force for βCD adsorption on the POPC membrane, while van der Waals interactions played a predominant role in DMβCD adsorption. The release profile of MGS from the βCDs pocket across the lipid bilayer exhibited two energy minima along the reaction coordinate associated with the permeation of the MGS molecule into the deeper region of the POPC membrane.
Collapse
Affiliation(s)
- Wiparat Hotarat
- Center of Excellence in Computational Chemistry (CECC), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (W.H.); (B.N.)
| | - Bodee Nutho
- Center of Excellence in Computational Chemistry (CECC), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (W.H.); (B.N.)
| | - Peter Wolschann
- Department of Pharmaceutical Chemistry, University of Vienna, 1090 Vienna, Austria;
- Institute of Theoretical Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Thanyada Rungrotmongkol
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Molecular Sensory Science Center, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (T.R.); (S.H.); Tel.: +66(0)2218-5418 (T.R.); +66(0)2218-7603 (S.H.)
| | - Supot Hannongbua
- Center of Excellence in Computational Chemistry (CECC), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (W.H.); (B.N.)
- Correspondence: (T.R.); (S.H.); Tel.: +66(0)2218-5418 (T.R.); +66(0)2218-7603 (S.H.)
| |
Collapse
|
16
|
Torres MD, Sothiselvam S, Lu TK, de la Fuente-Nunez C. Peptide Design Principles for Antimicrobial Applications. J Mol Biol 2019; 431:3547-3567. [DOI: 10.1016/j.jmb.2018.12.015] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 12/19/2018] [Accepted: 12/22/2018] [Indexed: 02/08/2023]
|
17
|
Juretić D, Simunić J. Design of α-helical antimicrobial peptides with a high selectivity index. Expert Opin Drug Discov 2019; 14:1053-1063. [DOI: 10.1080/17460441.2019.1642322] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Davor Juretić
- Mediterranean Institute for Life Sciences, Split, Croatia
- Department of Physics, Faculty of Science, University of Split, Split, Croatia
| | - Juraj Simunić
- Division of molecular biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
18
|
De Novo Design and In Vitro Testing of Antimicrobial Peptides against Gram-Negative Bacteria. Pharmaceuticals (Basel) 2019; 12:ph12020082. [PMID: 31163671 PMCID: PMC6631481 DOI: 10.3390/ph12020082] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/26/2019] [Accepted: 05/30/2019] [Indexed: 12/13/2022] Open
Abstract
Antimicrobial peptides (AMPs) have been identified as a potentially new class of antibiotics to combat bacterial resistance to conventional drugs. The design of de novo AMPs with high therapeutic indexes, low cost of synthesis, high resistance to proteases and high bioavailability remains a challenge. Such design requires computational modeling of antimicrobial properties. Currently, most computational methods cannot accurately calculate antimicrobial potency against particular strains of bacterial pathogens. We developed a tool for AMP prediction (Special Prediction (SP) tool) and made it available on our Web site (https://dbaasp.org/prediction). Based on this tool, a simple algorithm for the design of de novo AMPs (DSP) was created. We used DSP to design short peptides with high therapeutic indexes against gram-negative bacteria. The predicted peptides have been synthesized and tested in vitro against a panel of gram-negative bacteria, including drug resistant ones. Predicted activity against Escherichia coli ATCC 25922 was experimentally confirmed for 14 out of 15 peptides. Further improvements for designed peptides included the synthesis of D-enantiomers, which are traditionally used to increase resistance against proteases. One synthetic D-peptide (SP15D) possesses one of the lowest values of minimum inhibitory concentration (MIC) among all DBAASP database short peptides at the time of the submission of this article, while being highly stable against proteases and having a high therapeutic index. The mode of anti-bacterial action, assessed by fluorescence microscopy, shows that SP15D acts similarly to cell penetrating peptides. SP15D can be considered a promising candidate for the development of peptide antibiotics. We plan further exploratory studies with the SP tool, aiming at finding peptides which are active against other pathogenic organisms.
Collapse
|
19
|
Fernandes C, Carraro ML, Ribeiro J, Araújo J, Tiritan ME, Pinto MMM. Synthetic Chiral Derivatives of Xanthones: Biological Activities and Enantioselectivity Studies. Molecules 2019; 24:E791. [PMID: 30813236 PMCID: PMC6412826 DOI: 10.3390/molecules24040791] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/17/2019] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Many naturally occurring xanthones are chiral and present a wide range of biological and pharmacological activities. Some of them have been exhaustively studied and subsequently, obtained by synthesis. In order to obtain libraries of compounds for structure activity relationship (SAR) studies as well as to improve the biological activity, new bioactive analogues and derivatives inspired in natural prototypes were synthetized. Bioactive natural xanthones compromise a large structural multiplicity of compounds, including a diversity of chiral derivatives. Thus, recently an exponential interest in synthetic chiral derivatives of xanthones (CDXs) has been witnessed. The synthetic methodologies can afford structures that otherwise could not be reached within the natural products for biological activity and SAR studies. Another reason that justifies this trend is that both enantiomers can be obtained by using appropriate synthetic pathways, allowing the possibility to perform enantioselectivity studies. In this work, a literature review of synthetic CDXs is presented. The structures, the approaches used for their synthesis and the biological activities are described, emphasizing the enantioselectivity studies.
Collapse
Affiliation(s)
- Carla Fernandes
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Edifício do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal.
| | - Maria Letícia Carraro
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - João Ribeiro
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Joana Araújo
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Maria Elizabeth Tiritan
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Edifício do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal.
- Cooperativa de Ensino Superior, Politécnico e Universitário (CESPU), Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde (IINFACTS), Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal.
| | - Madalena M M Pinto
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Edifício do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal.
| |
Collapse
|
20
|
Sivaranjani M, Leskinen K, Aravindraja C, Saavalainen P, Pandian SK, Skurnik M, Ravi AV. Deciphering the Antibacterial Mode of Action of Alpha-Mangostin on Staphylococcus epidermidis RP62A Through an Integrated Transcriptomic and Proteomic Approach. Front Microbiol 2019; 10:150. [PMID: 30787919 PMCID: PMC6372523 DOI: 10.3389/fmicb.2019.00150] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/21/2019] [Indexed: 11/25/2022] Open
Abstract
Background: Alpha-mangostin (α-MG) is a natural xanthone reported to exhibit rapid bactericidal activity against Gram-positive bacteria, and may therefore have potential clinical application in healthcare sectors. This study sought to identify the impact of α-MG on Staphylococcus epidermidis RP62A through integrated advanced omic technologies. Methods: S. epidermidis was challenged with sub-MIC (0.875 μg/ml) of α-MG at various time points and the differential expression pattern of genes/proteins were analyzed in the absence and presence of α-MG using RNA sequencing and LC-MS/MS experiments. Bioinformatic tools were used to categorize the biological processes, molecular functions and KEGG pathways of differentially expressed genes/proteins. qRT-PCR was employed to validate the results obtained from these analyses. Results: Transcriptomic and proteomic profiling of α-MG treated cells indicated that genes/proteins affected by α-MG treatment were associated with diverse cellular functions. The greatest reduction in expression was observed in transcription of genes conferring cytoplasmic membrane integrity (yidC2, secA and mscL), cell division (ftsY and divlB), teichoic acid biosynthesis (tagG and dltA), fatty-acid biosynthesis (accB, accC, fabD, fabH, fabI, and fabZ), biofilm formation (icaA) and DNA replication and repair machinery (polA, polC, dnaE, and uvrA). Those with increased expression were involved in oxidative (katA and sodA) and cellular stress response (clpB, clpC, groEL, and asp23). The qRT-PCR analysis substantiated the results obtained from transcriptomic and proteomic profiling studies. Conclusion: Combining transcriptomic and proteomic methods provided comprehensive information about the antibacterial mode of action of α-MG. The obtained results suggest that α-MG targets S. epidermidis through multifarious mechanisms, and especially prompts that loss of cytoplasmic membrane integrity leads to rapid onset of bactericidal activity.
Collapse
Affiliation(s)
| | - Katarzyna Leskinen
- Department of Bacteriology and Immunology, Medicum, Research Programs Unit, Immunobiology Research Program, University of Helsinki, Helsinki, Finland
| | | | - Päivi Saavalainen
- Department of Bacteriology and Immunology, Medicum, Research Programs Unit, Immunobiology Research Program, University of Helsinki, Helsinki, Finland
| | | | - Mikael Skurnik
- Department of Bacteriology and Immunology, Medicum, Research Programs Unit, Immunobiology Research Program, University of Helsinki, Helsinki, Finland
- Division of Clinical Microbiology, Helsinki University Hospital, HUSLAB, Helsinki, Finland
| | | |
Collapse
|
21
|
Araújo J, Fernandes C, Pinto M, Tiritan ME. Chiral Derivatives of Xanthones with Antimicrobial Activity. Molecules 2019; 24:E314. [PMID: 30654546 PMCID: PMC6359477 DOI: 10.3390/molecules24020314] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
According to the World Health Organization, the exacerbated use of antibiotics worldwide is increasing multi-resistant infections, especially in the last decade. Xanthones are a class of compounds receiving great interest in drug discovery and development that can be found as natural products or obtained by synthesis. Many derivatives of xanthones are chiral and associated with relevant biological activities, including antimicrobial. The aim of this review is to compile information about chiral derivatives of xanthones from natural sources and their synthesized examples with antimicrobial activity.
Collapse
Affiliation(s)
- Joana Araújo
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Carla Fernandes
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Center of Marine and Environmental Research (CIIMAR), University of Porto, Edificio do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal.
| | - Madalena Pinto
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Center of Marine and Environmental Research (CIIMAR), University of Porto, Edificio do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal.
| | - Maria Elizabeth Tiritan
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Center of Marine and Environmental Research (CIIMAR), University of Porto, Edificio do Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4050-208 Matosinhos, Portugal.
- Institute of Research and Advanced Training in Health Sciences and Technologies, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal.
| |
Collapse
|
22
|
Design and synthesis of oligo-lipidated arginyl peptide (OLAP) dimers with enhanced physicochemical activity, peptide stability and their antimicrobial actions against MRSA infections. Amino Acids 2018; 50:1329-1345. [PMID: 30066172 DOI: 10.1007/s00726-018-2607-6] [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: 04/23/2018] [Accepted: 06/21/2018] [Indexed: 10/28/2022]
Abstract
Multi-drug resistant pathogens have been of increasing concern today. There is an urgent need for the discovery of more potent antibiotics. Cationic antimicrobial peptides (CAMPs) are known to be effective antimicrobial agents against resistant pathogens. However, poor activity under physiological conditions is one of the major limitations of CAMPS in clinical applications. In this study, a series of oligo-lipidated arginyl peptide OLAP dimers comprised of a saturated fatty acid chain (with m number of carbon units) and p repeating units of arginyl fatty acid chains (with n number of carbon units) were designed and studied for their antimicrobial activities as well as their physico-chemical property in various physiological conditions, such as in human serum albumin and high salt conditions. Our results showed that OLAP-11 exhibits potent antimicrobial activity against Gram-positive bacteria with improved physico-chemical activity in various physiological conditions. OLAP-11 is also less susceptible to human serum and trypsin degradation. The HPLC-MS analysis showed that the lipid-arginine bond is very stable. SYTOX Green assay and scanning electron microscopy both show that the OLAP-11 killed bacteria via inner membrane disruption. In addition, OLAP-11 is inner membrane targeting, making it difficult for bacteria to develop resistance. Overall, the design of the OLAP dimers provides an alternative approach to improve the physicochemical activity, peptide stability of CAMPs with potent inner membrane disruption and low in vitro toxicity to increase their potential for clinical applications in the future.
Collapse
|
23
|
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
|
24
|
Lau QY, Li J, Sani MA, Sinha S, Li Y, Ng FM, Kang C, Bhattacharjya S, Separovic F, Verma C, Chia CSB. Elucidating the bactericidal mechanism of action of the linear antimicrobial tetrapeptide BRBR-NH 2. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1517-1527. [PMID: 29758185 DOI: 10.1016/j.bbamem.2018.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 05/09/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022]
Abstract
Linear antimicrobial peptides, with their rapid bactericidal mode of action, are well-suited for development as topical antibacterial drugs. We recently designed a synthetic linear 4-residue peptide, BRBR-NH2, with potent bactericidal activity against Staphylococcus aureus (MIC 6.25 μM), the main causative pathogen of human skin infections with an unknown mechanism of action. Herein, we describe a series of experiments conducted to gain further insights into its mechanism of action involving electron microscopy, artificial membrane dye leakage, solution- and solid-state NMR spectroscopy followed by molecular dynamics simulations. Experimental results point towards a SMART (Soft Membranes Adapt and Respond, also Transiently) mechanism of action, suggesting that the peptide can be developed as a topical antibacterial agent for treating drug-resistant Staphylococcus aureus infections.
Collapse
Affiliation(s)
- Qiu Ying Lau
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, Nanos #03-01, Singapore 138669, Singapore
| | - Jianguo Li
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix #07-01, Singapore 138671, Singapore; Singapore Eye Research Institute, The Academia, 20 College Road, Singapore 168751, Singapore
| | - Marc-Antoine Sani
- School of Chemistry, Bio21 Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Sheetal Sinha
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; Advanced Environmental Biotechnology Centre, Nanyang Environment and Water Research Institute, Nanyang Technological University, 1 Cleantech Loop, Singapore 637141, Singapore; Interdisciplinary Graduate School, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Yan Li
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, Nanos #03-01, Singapore 138669, Singapore
| | - Fui Mee Ng
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, Nanos #03-01, Singapore 138669, Singapore
| | - CongBao Kang
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, Nanos #03-01, Singapore 138669, Singapore
| | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Frances Separovic
- School of Chemistry, Bio21 Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Chandra Verma
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix #07-01, Singapore 138671, Singapore; School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Cheng San Brian Chia
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, Nanos #03-01, Singapore 138669, Singapore.
| |
Collapse
|
25
|
Li J, Beuerman RW, Verma CS. Molecular Insights into the Membrane Affinities of Model Hydrophobes. ACS OMEGA 2018; 3:2498-2507. [PMID: 30023836 PMCID: PMC6044992 DOI: 10.1021/acsomega.7b01759] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/19/2018] [Indexed: 06/08/2023]
Abstract
Membrane-active antibiotics are of great interest in fighting bacterial resistance. α-Mangostin is a membrane-active molecule, but there are no details of its mechanism of action at the atomistic level. We have employed free-energy simulations and microsecond-long conventional molecular dynamics simulations to study the mode of interaction of α-mangostin with a model bacterial membrane and compare it with the mechanisms of three hydrophobic molecules (ciprofloxacin, xanthone, and tetracycline). We find that α-mangostin is thermodynamically more favored to insert into the membrane compared to the other three molecules. Apart from tetracycline, which is largely hydrophilic, the other three molecules aggregate in water; however, only α-mangostin can penetrate into the lipid tail region of the membrane. When it reaches a high concentration in the lipid tail region, α-mangostin can form tubular clusters that span the two head group regions of the membrane, resulting in a large number of water translocations along the transmembrane aggregates. Structure-activity relationship analysis revealed two structural properties that characterize α-mangostin, namely, the two isoprenyl groups and the polar groups present in the aromatic rings, which result in "disruptive amphiphilicity" and hence its excellent membrane activity.
Collapse
Affiliation(s)
- Jianguo Li
- Singapore
Eye Research Institute, The Academia, 20 College Road, 169856, Singapore
- Bioinformatics
Institute (A*-STAR), 30 Biopolis Street, #07-01 Matrix, 138671 Singapore
| | - Roger W. Beuerman
- Singapore
Eye Research Institute, The Academia, 20 College Road, 169856, Singapore
- Department
of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Lower Kent Ridge Road, 119074 Singapore
- Duke-NUS,
SRP Neuroscience & Behavioural Disorders, 8 College Road, 169857, Singapore
| | - Chandra S. Verma
- Singapore
Eye Research Institute, The Academia, 20 College Road, 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
| |
Collapse
|
26
|
A novel α-conopeptide Eu1.6 inhibits N-type (Ca V2.2) calcium channels and exhibits potent analgesic activity. Sci Rep 2018; 8:1004. [PMID: 29343689 PMCID: PMC5772529 DOI: 10.1038/s41598-017-18479-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/06/2017] [Indexed: 01/13/2023] Open
Abstract
We here describe a novel α-conopeptide, Eu1.6 from Conus eburneus, which exhibits strong anti-nociceptive activity by an unexpected mechanism of action. Unlike other α-conopeptides that largely target nicotinic acetylcholine receptors (nAChRs), Eu1.6 displayed only weak inhibitory activity at the α3β4 and α7 nAChR subtypes and TTX-resistant sodium channels, and no activity at TTX-sensitive sodium channels in rat dorsal root ganglion (DRG) neurons, or opiate receptors, VR1, KCNQ1, L- and T-type calcium channels expressed in HEK293 cells. However, Eu1.6 inhibited high voltage-activated N-type calcium channel currents in isolated mouse DRG neurons which was independent of GABAB receptor activation. In HEK293 cells expressing CaV2.2 channels alone, Eu1.6 reversibly inhibited depolarization-activated Ba2+ currents in a voltage- and state-dependent manner. Inhibition of CaV2.2 by Eu1.6 was concentration-dependent (IC50 ~1 nM). Significantly, systemic administration of Eu1.6 at doses of 2.5–5.0 μg/kg exhibited potent analgesic activities in rat partial sciatic nerve injury and chronic constriction injury pain models. Furthermore, Eu1.6 had no significant side-effect on spontaneous locomotor activity, cardiac and respiratory function, and drug dependence in mice. These findings suggest α-conopeptide Eu1.6 is a potent analgesic for the treatment of neuropathic and chronic pain and opens a novel option for future analgesic drug design.
Collapse
|
27
|
|
28
|
Dutertre S, Nicke A, Tsetlin VI. Nicotinic acetylcholine receptor inhibitors derived from snake and snail venoms. Neuropharmacology 2017. [PMID: 28623170 DOI: 10.1016/j.neuropharm.2017.06.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The nicotinic acetylcholine receptor (nAChR) represents the prototype of ligand-gated ion channels. It is vital for neuromuscular transmission and an important regulator of neurotransmission. A variety of toxic compounds derived from diverse species target this receptor and have been of elemental importance in basic and applied research. They enabled milestone discoveries in pharmacology and biochemistry ranging from the original formulation of the receptor concept, the first isolation and structural analysis of a receptor protein (the nAChR) to the identification, localization, and differentiation of its diverse subtypes and their validation as a target for therapeutic intervention. Among the venom-derived compounds, α-neurotoxins and α-conotoxins provide the largest families and still represent indispensable pharmacological tools. Application of modified α-neurotoxins provided substantial structural and functional details of the nAChR long before high resolution structures were available. α-bungarotoxin represents not only a standard pharmacological tool and label in nAChR research but also for unrelated proteins tagged with a minimal α-bungarotoxin binding motif. A major advantage of α-conotoxins is their smaller size, as well as superior selectivity for diverse nAChR subtypes that allows their development into ligands with optimized pharmacological and chemical properties and potentially novel drugs. In the following, these two groups of nAChR antagonists will be described focusing on their respective roles in the structural and functional characterization of nAChRs and their development into research tools. In addition, we provide a comparative overview of the diverse α-conotoxin selectivities that can serve as a practical guide for both structure activity studies and subtype classification. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
Affiliation(s)
- Sébastien Dutertre
- Institut des Biomolécules Max Mousseron, UMR 5247, Université Montpellier - CNRS, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
| | - Annette Nicke
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Nußbaumstr. 26, 80336 Munich, Germany.
| | - Victor I Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str.16/10, Moscow 117999, Russian Federation
| |
Collapse
|
29
|
Lee Y, Choi KH, Park KM, Lee JM, Park BJ, Park KD. In Situ Forming and H 2O 2-Releasing Hydrogels for Treatment of Drug-Resistant Bacterial Infections. ACS APPLIED MATERIALS & INTERFACES 2017; 9:16890-16899. [PMID: 28474514 DOI: 10.1021/acsami.7b03870] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Various types of commercialized wound dressings (e.g., films, foams, gels, and nanofiber meshes) have been clinically used as a physical barrier against bacterial invasion and as wound-healing materials. Although these dressings can protect the wounded tissue from the external environment, they cannot treat the wounds that are already infected with bacteria. Herein, we report in situ H2O2-releasing hydrogels as an active wound dressing with antibacterial properties for treatment of drug-resistant bacterial infection. In this study, H2O2 was used for two major purposes: (1) in situ gel formation via a horseradish peroxidase (HRP)/H2O2-triggered cross-linking reaction, and (2) antibacterial activity of the hydrogel via its oxidative effects. We found that there were residual H2O2 in the matrix after in situ HRP-catalyzed gelling, and varying the feed amount of H2O2 (1-10 mM; used to make hydrogels) enabled control of H2O2 release kinetics within a range of 2-509 μM. In addition, although the gelatin-hydroxyphenyl propionic acid (GH) gel called "GH 10" (showing the greatest H2O2 release, 509 μM) slightly decreased cell viability (to 82-84%) of keratinocyte (HaCaT) and fibroblast (L-929) cells in in vitro assays, none of the hydrogels showed significant cytotoxicity toward tissues in in vivo skin irritation tests. When the H2O2-releasing hydrogels that promote in vivo wound healing, were applied to various bacterial strains in vitro and ex vivo, they showed strong killing efficiency toward Gram-positive bacteria including Staphylococcus aureus, S. epidermidis, and clinical isolate of methicillin-resistant S. aureus (MRSA, drug-resistant bacteria), where the antimicrobial effect was dependent on the concentration of the H2O2 released. The present study suggests that our hydrogels have great potential as an injectable/sprayable antimicrobial dressing with biocompatibility and antibacterial activity against drug-resistant bacteria including MRSA for wound and infection treatment.
Collapse
Affiliation(s)
- Yunki Lee
- Department of Molecular Science and Technology, Ajou University , Suwon 443-749, Republic of Korea
| | - Kyong-Hoon Choi
- Department of Electrical and Biological Physics, Kwangwoon University , Seoul 138-701, Republic of Korea
| | - Kyung Min Park
- Division of Bioengineering, College of Life Sciences and Bioengineering, Incheon National University , Incheon 22012, Republic of Korea
| | - Jong-Min Lee
- College of Medicine, Dongguk University , Goyang 10326, Republic of Korea
| | - Bong Joo Park
- Department of Electrical and Biological Physics, Kwangwoon University , Seoul 138-701, Republic of Korea
| | - Ki Dong Park
- Department of Molecular Science and Technology, Ajou University , Suwon 443-749, Republic of Korea
| |
Collapse
|
30
|
Abstract
Mangosteen (Garcinia mangostana Linn.) is a well-known tropical tree indigenous to Southeast Asia. Its fruit's pericarp abounds with a class of isoprenylated xanthones which are referred as mangostins. Numerous in vitro and in vivo studies have shown that mangostins and their derivatives possess diverse pharmacological activities, such as antibacterial, antifungal, antimalarial, anticarcinogenic, antiatherogenic activities as well as neuroprotective properties in Alzheimer's disease (AD). This review article provides a comprehensive review of the pharmacological activities of mangostins and their derivatives to reveal their promising utilities in the treatment of certain important diseases, mainly focusing on the discussions of the underlying molecular targets/pathways, modes of action, and relevant structure-activity relationships (SARs). Meanwhile, the pharmacokinetics (PK) profile and recent toxicological studies of mangostins are also described for further druggability exploration in the future.
Collapse
|
31
|
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: 372] [Impact Index Per Article: 46.5] [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
|
32
|
Lin S, Koh JJ, Aung TT, Lim F, Li J, Zou H, Wang L, Lakshminarayanan R, Verma C, Wang Y, Tan DTH, Cao D, Beuerman RW, Ren L, Liu S. Symmetrically Substituted Xanthone Amphiphiles Combat Gram-Positive Bacterial Resistance with Enhanced Membrane Selectivity. J Med Chem 2017; 60:1362-1378. [DOI: 10.1021/acs.jmedchem.6b01403] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Shuimu Lin
- School
of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Singapore
Eye Research Institute, The Academia, 20 College Road, Discovery Tower
Level 6, 169856, Singapore
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Jun-Jie Koh
- 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
| | - Fanghui Lim
- 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
| | - Hanxun Zou
- Singapore
Eye Research Institute, The Academia, 20 College Road, Discovery Tower
Level 6, 169856, Singapore
| | - Lin Wang
- School
of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Rajamani Lakshminarayanan
- Singapore
Eye Research Institute, The Academia, 20 College Road, Discovery Tower
Level 6, 169856, Singapore
- SRP
Neuroscience and Behavioral 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
| | - Yingjun Wang
- School
of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Donald T. H. Tan
- Singapore
Eye Research Institute, The Academia, 20 College Road, Discovery Tower
Level 6, 169856, Singapore
- Singapore National Eye Center, 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 Behavioral Disorders, Duke-NUS Graduate Medical School, 169857, Singapore
| | - Li Ren
- School
of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Shouping Liu
- Singapore
Eye Research Institute, The Academia, 20 College Road, Discovery Tower
Level 6, 169856, Singapore
- SRP
Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School, 169857, Singapore
| |
Collapse
|
33
|
Amphiphilic xanthones as a potent chemical entity of anti-mycobacterial agents with membrane-targeting properties. Eur J Med Chem 2016; 123:684-703. [PMID: 27517813 DOI: 10.1016/j.ejmech.2016.07.068] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022]
Abstract
Tuberculosis (TB) remains a deadly disease and infects one-third of the world's population. Given the low success rates encountered in clinical development, there is an urgent need to identify structurally novel antimicrobials for tuberculosis. The present report details the anti-mycobacterial activities, structure-activity relationships (SARs) and mechanism of action of amphiphilic xanthone derivatives. The xanthones exhibited potent MIC, rapid time-kill and no cross-resistance with the current anti-TB drugs. Evidence is presented that these compounds disrupted the inner membrane and led to ATP depletion. Amphiphilic xanthone derivatives exhibited superior metabolic stability, low cytotoxicity and low activity against the common cytochrome P450. Compound 5 was selected for an in vivo pharmacokinetic study. Its bioavailability at an oral dose of 2 mg/kg was 15%. The xanthones thuse provide valuable insight for the development of a new class of membrane targeting antimycobacterial agents that may assist in overcoming the limitations of the current TB medications.
Collapse
|
34
|
Li Z, Chee PL, Owh C, Lakshminarayanan R, Loh XJ. Safe and efficient membrane permeabilizing polymers based on PLLA for antibacterial applications. RSC Adv 2016. [DOI: 10.1039/c6ra04531f] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Highly active antibacterial poly(N,N-dimethylaminoethyl methacrylate)-block-poly(l-lactic acid)-block-poly(N,N-dimethylaminoethyl methacrylate) conjugated with poly(ethylene glycol) (D-PLLA-D@PEG) copolymers were synthesized.
Collapse
Affiliation(s)
- Zibiao Li
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science Technology and Research)
- Singapore 138634
- Singapore
| | - Pei Lin Chee
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science Technology and Research)
- Singapore 138634
- Singapore
| | - Cally Owh
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science Technology and Research)
- Singapore 138634
- Singapore
| | | | - Xian Jun Loh
- Institute of Materials Research and Engineering
- A*STAR (Agency for Science Technology and Research)
- Singapore 138634
- Singapore
- Department of Materials Science and Engineering
| |
Collapse
|
35
|
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
|
36
|
Deng D, Li Y, Xue J, Wang J, Ai G, Li X, Gu Y. Gold nanoparticle-based beacon to detect STAT5b mRNA expression in living cells: a case optimized by bioinformatics screen. Int J Nanomedicine 2015; 10:3231-44. [PMID: 25987838 PMCID: PMC4422291 DOI: 10.2147/ijn.s81754] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Messenger RNA (mRNA), a single-strand ribonucleic acid with functional gene information is usually abnormally expressed in cancer cells and has become a promising biomarker for the study of tumor progress. Hairpin DNA-coated gold nanoparticle (hDAuNP) beacon containing a bare gold nanoparticle (AuNP) as fluorescence quencher and thiol-terminated fluorescently labeled stem-loop-stem oligonucleotide sequences attached by Au-S bond is currently a new nanoscale biodiagnostic platform capable of mRNA detection, in which the design of the loop region sequence is crucial for hybridizing with the target mRNA. Hence, in this study, to improve the sensitivity and selectivity of hDAuNP beacon simultaneously, the loop region of hairpin DNA was screened by bioinformatics strategy. Here, signal transducer and activator of transcription 5b (STAT5b) mRNA was selected and used as a practical example. The results from the combined characterizations using optical techniques, flow cytometry assay, and cell microscopic imaging showed that after optimization, the as-prepared hDAuNP beacon had higher selectivity and sensitivity for the detection of STAT5b mRNA in living cells, as compared with our previous beacon. Thus, the bioinformatics method may be a promising new strategy for assisting in the designing of the hDAuNP beacon, extending its application in the detection of mRNA expression and the resultant mRNA-based biological processes and disease pathogenesis.
Collapse
Affiliation(s)
- Dawei Deng
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yang Li
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jianpeng Xue
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jie Wang
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Guanhua Ai
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xin Li
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yueqing Gu
- Department of Biomedical Engineering, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|