101
|
Zhao X, Kuipers OP. BrevicidineB, a New Member of the Brevicidine Family, Displays an Extended Target Specificity. Front Microbiol 2021; 12:693117. [PMID: 34177875 PMCID: PMC8219939 DOI: 10.3389/fmicb.2021.693117] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022] Open
Abstract
The group of bacterial non-ribosomally produced peptides (NRPs) has formed a rich source for drug development. Brevicidine, a bacterial non-ribosomally produced cyclic lipo-dodecapeptide, displays selective antimicrobial activity against Gram-negative pathogens. Here, we show that brevicidineB, which contains a single substitution (Tyr2 to Phe2) in the amino acid sequence of the linear part of brevicidine, has a broadened antimicrobial spectrum, showing bactericidal activity against both Gram-negative (with a MIC value of 2 to 4 mg/L) and Gram-positive (with a MIC value of 2 to 8 mg/L) pathogens. Compared with an earlier reported member of the brevicidine family, the broadened antimicrobial spectrum of brevicidineB is caused by its increased membrane disruptive capacity on Gram-positive pathogens, which was evidenced by fluorescence microscopy assays. In addition, DiSC3(5) and resazurin assays show that brevicidine and brevicidineB exert their antimicrobial activity against Gram-negative bacteria via disrupting the proton motive force of cells. Notably, as a brevicidine family member, brevicidineB also showed neither hemolytic activity nor cytotoxicity at a high concentration of 64 mg/L. This study provides a promising antibiotic candidate (brevicidineB) with a broad antimicrobial spectrum, and provides novel insights into the antimicrobial mode of action of brevicidines.
Collapse
Affiliation(s)
- Xinghong Zhao
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Oscar P Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| |
Collapse
|
102
|
Almeida CV, de Oliveira CFR, Dos Santos EL, Dos Santos HF, Júnior EC, Marchetto R, da Cruz LA, Ferreira AMT, Gomes VM, Taveira GB, Costa BO, Franco OL, Cardoso MH, Macedo MLR. Differential interactions of the antimicrobial peptide, RQ18, with phospholipids and cholesterol modulate its selectivity for microorganism membranes. Biochim Biophys Acta Gen Subj 2021; 1865:129937. [PMID: 34052310 DOI: 10.1016/j.bbagen.2021.129937] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/15/2021] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Antimicrobial peptides (AMPs) are molecules with potential application for the treatment of microorganism infections. We, herein, describe the structure, activity, and mechanism of action of RQ18, an α-helical AMP that displays antimicrobial activity against Gram-positive and Gram-negative bacteria, and yeasts from the Candida genus. METHODS A physicochemical-guided design assisted by computer tools was used to obtain our lead peptide candidate, named RQ18. This peptide was assayed against Gram-positive and Gram-negative bacteria, yeasts, and mammalian cells to determine its selectivity index. The secondary structure and the mechanism of action of RQ18 were investigated using circular dichroism, large unilamellar vesicles, and molecular dynamic simulations. RESULTS RQ18 was not cytotoxic to human lung fibroblasts, peripheral blood mononuclear cells, red blood cells, or Vero cells at MIC values, exhibiting a high selectivity index. Circular dichroism analysis and molecular dynamic simulations revealed that RQ18 presents varying structural profiles in aqueous solution, TFE/water mixtures, SDS micelles, and lipid bilayers. The peptide was virtually unable to release carboxyfluorescein from large unilamellar vesicles composed of POPC/cholesterol, model that mimics the eukaryotic membrane, indicating that vesicles' net charges and the presence of cholesterol may be related with RQ18 selectivity for bacterial and fungal cell surfaces. CONCLUSIONS RQ18 was characterized as a membrane-active peptide with dual antibacterial and antifungal activities, without compromising mammalian cells viability, thus reinforcing its therapeutic application. GENERAL SIGNIFICANCE These results provide further insight into the complex process of AMPs interaction with biological membranes, in special with systems that mimic prokaryotic and eukaryotic cell surfaces.
Collapse
Affiliation(s)
- Claudiane V Almeida
- Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Caio F R de Oliveira
- Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil; Universidade Federal da Grande Dourados, Dourados, Mato Grosso do Sul, Brazil; Oncolytic Anticancer Drugs, Dourados, Mato Grosso do Sul, Brazil
| | - Edson L Dos Santos
- Universidade Federal da Grande Dourados, Dourados, Mato Grosso do Sul, Brazil
| | - Helder F Dos Santos
- Universidade Federal da Grande Dourados, Dourados, Mato Grosso do Sul, Brazil
| | - Edson C Júnior
- Instituto de Química, Departamento de Bioquímica e Química Tecnológica, Universidade Estadual Paulista Júlio de Mesquita Filho, Araraquara, São Paulo, Brazil
| | - Reinaldo Marchetto
- Instituto de Química, Departamento de Bioquímica e Química Tecnológica, Universidade Estadual Paulista Júlio de Mesquita Filho, Araraquara, São Paulo, Brazil
| | - Leticia A da Cruz
- Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Alda Maria T Ferreira
- Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Valdirene M Gomes
- Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Gabriel B Taveira
- Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Bruna O Costa
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Octávio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Marlon H Cardoso
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Maria Lígia R Macedo
- Universidade Federal de Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil.
| |
Collapse
|
103
|
Klobucar K, Côté JP, French S, Borrillo L, Guo ABY, Serrano-Wu MH, Lee KK, Hubbard B, Johnson JW, Gaulin JL, Magolan J, Hung DT, Brown ED. Chemical Screen for Vancomycin Antagonism Uncovers Probes of the Gram-Negative Outer Membrane. ACS Chem Biol 2021; 16:929-942. [PMID: 33974796 DOI: 10.1021/acschembio.1c00179] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The outer membrane of Gram-negative bacteria is a formidable permeability barrier which allows only a small subset of chemical matter to penetrate. This outer membrane barrier can hinder the study of cellular processes and compound mechanism of action, as many compounds including antibiotics are precluded from entry despite having intracellular targets. Consequently, outer membrane permeabilizing compounds are invaluable tools in such studies. Many existing compounds known to perturb the outer membrane also impact inner membrane integrity, such as polymyxins and their derivatives, making these probes nonspecific. We performed a screen of ∼140 000 diverse synthetic compounds, for those that antagonized the growth inhibitory activity of vancomycin at 15 °C in Escherichia coli, to enrich for chemicals capable of perturbing the outer membrane. This led to the discovery that liproxstatin-1, an inhibitor of ferroptosis in human cells, and MAC-0568743, a novel cationic amphiphile, could potentiate the activity of large-scaffold antibiotics with low permeation into Gram-negative bacteria at 37 °C. Liproxstatin-1 and MAC-0568743 were found to physically disrupt the integrity of the outer membrane through interactions with lipopolysaccharide in the outer leaflet of the outer membrane. We showed that these compounds selectively disrupt the outer membrane while minimally impacting inner membrane integrity, particularly at the concentrations needed to potentiate Gram-positive-targeting antibiotics. Further exploration of these molecules and their structural analogues is a promising avenue for the development of outer membrane specific probes.
Collapse
Affiliation(s)
- Kristina Klobucar
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Jean-Philippe Côté
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Shawn French
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Louis Borrillo
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Amelia Bing Ya Guo
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Michael H. Serrano-Wu
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Katie K. Lee
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Brian Hubbard
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Jarrod W. Johnson
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Jeffrey L. Gaulin
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Jakob Magolan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Deborah T. Hung
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Eric D. Brown
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| |
Collapse
|
104
|
Ashrafuzzaman M. Amphiphiles capsaicin and triton X-100 regulate the chemotherapy drug colchicine's membrane adsorption and ion pore formation potency. Saudi J Biol Sci 2021; 28:3100-3109. [PMID: 34025185 PMCID: PMC8117037 DOI: 10.1016/j.sjbs.2021.02.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/07/2021] [Accepted: 02/15/2021] [Indexed: 02/02/2023] Open
Abstract
Chemotherapy drugs (CDs), e.g. colchicine derivative thiocolchicoside (TCC) and taxol, have been found to physically bind with lipid bilayer membrane and induce ion pores. Amphiphiles capsaicin (Cpsn) and triton X-100 (TX100) are known to regulate lipid bilayer physical properties by altering bilayer elasticity and lipid monolayer curvature. Both CDs and amphiphiles are predicted to physically accommodate alongside lipids in membrane to exert their membrane effects. The effects of their binary accommodation in the lipid membrane are yet to be known. Firstly, we have performed experimental studies to inspect whether membrane adsorption of CDs (colchicine or TCC) gets regulated due to any membrane effects of Cpsn or TX100. We find that the aqueous phase presence of these amphiphiles, known to reduce the membrane stiffness, works towards enhancing the membrane adsorption of CDs. Our recently patented technology 'direct detection method' helps address the membrane adsorption mechanisms. Secondly, in electrophysiology records, we measured the amphiphile effects on the potency of ion channel induction due to CDs. We find that amphiphiles increase the CD induced channel induction potency. Specifically, the membrane conductance, apparently due to the ion channel induction by the TCC, increases substantially due to the Cpsn or TX100 induced alterations of the bilayer physical properties. Thus we may conclude that the binary presence of CDs and amphiphiles in lipid membrane may influence considerably in CD's membrane adsorption, as well as the membrane effects, such as ion pore formation.
Collapse
Affiliation(s)
- Md. Ashrafuzzaman
- Biochemistry Department, Science College, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
105
|
El-Beyrouthy J, Freeman E. Characterizing the Structure and Interactions of Model Lipid Membranes Using Electrophysiology. MEMBRANES 2021; 11:319. [PMID: 33925756 PMCID: PMC8145864 DOI: 10.3390/membranes11050319] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 11/16/2022]
Abstract
The cell membrane is a protective barrier whose configuration determines the exchange both between intracellular and extracellular regions and within the cell itself. Consequently, characterizing membrane properties and interactions is essential for advancements in topics such as limiting nanoparticle cytotoxicity. Characterization is often accomplished by recreating model membranes that approximate the structure of cellular membranes in a controlled environment, formed using self-assembly principles. The selected method for membrane creation influences the properties of the membrane assembly, including their response to electric fields used for characterizing transmembrane exchanges. When these self-assembled model membranes are combined with electrophysiology, it is possible to exploit their non-physiological mechanics to enable additional measurements of membrane interactions and phenomena. This review describes several common model membranes including liposomes, pore-spanning membranes, solid supported membranes, and emulsion-based membranes, emphasizing their varying structure due to the selected mode of production. Next, electrophysiology techniques that exploit these structures are discussed, including conductance measurements, electrowetting and electrocompression analysis, and electroimpedance spectroscopy. The focus of this review is linking each membrane assembly technique to the properties of the resulting membrane, discussing how these properties enable alternative electrophysiological approaches to measuring membrane characteristics and interactions.
Collapse
Affiliation(s)
| | - Eric Freeman
- School of Environmental, Civil, Agricultural and Mechanical Engineering, College of Engineering, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
106
|
Effect of membrane potential on entry of lactoferricin B-derived 6-residue antimicrobial peptide into single Escherichia coli cells and lipid vesicles. J Bacteriol 2021; 203:JB.00021-21. [PMID: 33558393 PMCID: PMC8092161 DOI: 10.1128/jb.00021-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The antimicrobial peptide (AMP) derived from lactoferricin B, LfcinB (4-9) (RRWQWR), and lissamine rhodamine B red-labeled peptide (Rh-LfcinB (4-9)) exhibit strong antimicrobial activities, and they can enter Escherichia coli cells without damaging the cell membranes. Thus, these peptides are cell-penetrating peptide (CPP) -type AMPs. In this study, to elucidate the effect of the membrane potential (Δφ) on the action of the CPP-type AMP, Rh-LfcinB (4-9), we investigated the interactions of Rh-LfcinB (4-9) with single E. coli cells and spheroplasts containing calcein in the cytosol using confocal laser scanning microscopy. At low peptide concentrations, Rh-LfcinB (4-9) entered the cytosol of single E. coli cells and spheroplasts without damaging the cell membranes, and the H+-ionophore carbonyl cyanide m-chlorophenyl-hydrazone (CCCP) suppressed its entry. The studies using the time-kill method indicate that these low concentrations of peptide exhibit antimicrobial activity but CCCP inhibits this activity. Next, we investigated the effect of Δφ on the interaction of Rh-LfcinB (4-9) with single giant unilamellar vesicles (GUVs) comprising E. coli polar lipid extracts and containing a fluorescent probe, Alexa Fluor 647 hydrazide. At low concentrations (0.2-0.5 μM), Rh-LfcinB (4-9) showed significant entry to the single GUV lumen without pore formation in the presence of Δφ. The fraction of entry of peptide increased with increasing negative membrane potential, indicating that the rate of peptide entry into the GUV lumen increased with increasing negative membrane potential. These results indicate that Δφ enhances the entry of Rh-LfcinB (4-9) into single E. coli cells, spheroplasts, and GUVs and its antimicrobial activity.IMPORTANCE: Bacterial cells have a membrane potential (Δφ), but the effect of Δφ on action of cell-penetrating peptide-type antimicrobial peptides (AMPs) is not clear. Here, we investigated the effect of Δφ on the action of fluorescent probe-labeled AMP derived from lactoferricin B, Rh-LfcinB (4-9). At low peptide concentrations, Rh-LfcinB (4-9) enters the cytosol of Escherichia coli cells and spheroplasts without damaging their cell membrane, but a protonophore suppresses this entry and its antimicrobial activity. The rate of entry of Rh-LfcinB (4-9) into the giant unilamellar vesicles (GUVs) comprising E. coli lipids without pore formation increases with increasing Δφ. These results indicate that Δφ enhances the antimicrobial activity of Rh-LfcinB (4-9) and hence LfcinB (4-9) by increasing the rate of their entry into the cytosol.
Collapse
|
107
|
Ashrafuzzaman M. The Antimicrobial Peptide Gramicidin S Enhances Membrane Adsorption and Ion Pore Formation Potency of Chemotherapy Drugs in Lipid Bilayers. MEMBRANES 2021; 11:247. [PMID: 33808204 PMCID: PMC8067072 DOI: 10.3390/membranes11040247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/06/2021] [Accepted: 02/17/2021] [Indexed: 01/25/2023]
Abstract
We recently published two novel findings where we found the chemotherapy drugs (CDs) thiocolchicoside (TCC) and taxol to induce toroidal type ion pores and the antimicrobial peptide gramicidin S (GS) to induce transient defects in model membranes. Both CD pores and GS defects were induced under the influence of an applied transmembrane potential (≈100 mV), which was inspected using the electrophysiology record of membrane currents (ERMCs). In this article, I address the regulation of the membrane adsorption and pore formation of CDs due to GS-induced possible alterations of lipid bilayer physical properties. In ERMCs, low micromolar (≥1 μM) GS concentrations in the aqueous phase were found to cause an induction of defects in lipid bilayers, but nanomolar (nM) concentration GS did nothing. For the binary presence of CDs and GS in the membrane-bathing aqueous phase, the TCC pore formation potency is found to increase considerably due to nM concentration GS in buffer. This novel result resembles our recently reported finding that due to the binary aqueous presence of two AMPs (gramicidin A or alamethicin and GS), the pore or defect-forming potency of either AMP increases considerably. To reveal the underlying molecular mechanisms, the influence of GS (0-400 nM) on the quantitative liposome (membrane) adsorption of CD molecules, colchicine and TCC, was tested. I used the recently patented direct detection method, which helps detect the membrane active agents directly at the membrane in the mole fraction relative to its concentrations in aqueous phase. We find that GS, at concentrations known to do nothing to the lipid bilayer electrical barrier properties in ERMCs, increases the membrane adsorption (membrane uptake) of CDs considerably. This phenomenological finding along with the GS effects on CD-induced membrane conductance increase helps predict an important conclusion. The binary presence of AMPs alongside CDs in the lipid membrane vicinity may work toward enhancing the physical adsorption and pore formation potency of CDs in lipid bilayers. This may help understand why CDs cause considerable cytotoxicity.
Collapse
Affiliation(s)
- Md Ashrafuzzaman
- Biochemistry Department, Science College, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
108
|
Yan Y, Li Y, Zhang Z, Wang X, Niu Y, Zhang S, Xu W, Ren C. Advances of peptides for antibacterial applications. Colloids Surf B Biointerfaces 2021; 202:111682. [PMID: 33714188 DOI: 10.1016/j.colsurfb.2021.111682] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/09/2020] [Accepted: 03/05/2021] [Indexed: 01/08/2023]
Abstract
In the past few decades, peptide antibacterial products with unique antibacterial mechanisms have attracted widespread interest. They can effectively reduce the probability of drug resistance of bacteria and are biocompatible, so they possess tremendous development prospects. This review provides recent research and analysis on the basic types of antimicrobial peptides (including poly (amino acid)s, short AMPs, and lipopeptides) and factors to optimize antimicrobial effects. It also summarizes the two most important modes of action of antimicrobial peptides and the latest developments in the application of AMPs, including antimicrobial agent, wound healing, preservative, antibacterial coating and others. Finally, we discuss the remaining challenges to improve the antibacterial peptides and propose prospects in the field.
Collapse
Affiliation(s)
- Yuhan Yan
- School of Chemistry and Materials Science, Ludong University, Yantai, 264025, China
| | - Yuanze Li
- School of Chemistry and Materials Science, Ludong University, Yantai, 264025, China
| | - Zhiwen Zhang
- School of Chemistry and Materials Science, Ludong University, Yantai, 264025, China
| | - Xinhao Wang
- School of Chemistry and Materials Science, Ludong University, Yantai, 264025, China
| | - Yuzhong Niu
- School of Chemistry and Materials Science, Ludong University, Yantai, 264025, China
| | - Shaohua Zhang
- School of Chemistry and Materials Science, Ludong University, Yantai, 264025, China.
| | - Wenlong Xu
- School of Chemistry and Materials Science, Ludong University, Yantai, 264025, China.
| | - Chunguang Ren
- Yantai Institute of Materia Medica, Yantai, 264000, China.
| |
Collapse
|
109
|
Joodaki F, Martin LM, Greenfield ML. Computational Study of Helical and Helix-Hinge-Helix Conformations of an Anti-Microbial Peptide in Solution by Molecular Dynamics and Vibrational Analysis. J Phys Chem B 2021; 125:703-721. [PMID: 33464100 DOI: 10.1021/acs.jpcb.0c07988] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many classical antimicrobial peptides adopt an amphipathic helical structure at a water-membrane interface. Prior studies led to the hypothesis that a hinge near the middle of a helical peptide plays an important role in facilitating peptide-membrane interactions. Here, dynamics and vibrations of a designed hybrid antimicrobial peptide LM7-2 in solution were simulated to investigate its hinge formation. Molecular dynamics simulation results on the basis of the CHARMM36 force field showed that the α-helix LM7-2 bent around two or three residues near the middle of the peptide, stayed in a helix-hinge-helix conformation for a short period of time, and then returned to a helical conformation. High-resolution computational vibrational techniques were applied on the LM7-2 system when it has α-helical and helix-hinge-helix conformations to understand how this structural change affects its inherent vibrations. These studies concentrated on the calculation of frequencies that correspond to backbone amide bands I, II, and III: vibrational modes that are sensitive to changes in the secondary structure of peptides and proteins. To that end, Fourier transforms were applied to thermal fluctuations in C-N-H angles, C-N bond lengths, and C═O bond lengths of each amide group. In addition, instantaneous all-atom normal mode analysis was applied to monitor and detect the characteristic amide bands of each amide group within LM7-2 during the MD simulation. Computational vibrational results indicate that shapes and frequencies of amide bands II and especially III were altered only for amide groups near the hinge. These methods provide high-resolution vibrational information that can complement spectroscopic vibrational studies. They assist in interpreting spectra of similar systems and suggest a marker for the presence of the helix-hinge-helix motif. Moreover, radial distribution functions indicated an increase in the probability of hydrogen bonding between water and a hydrogen atom connected to nitrogen (HN) in such a hinge. The probability of intramolecular hydrogen bond formation between HN and an amide group oxygen atom within LM7-2 was lower around the hinge. No correlation has been found between the presence of a hinge and hydrogen bonds between amide group oxygen atoms and the hydrogen atoms of water molecules. This result suggests a mechanism for hinge formation wherein hydrogen bonds to oxygen atoms of water replace intramolecular hydrogen bonds as the peptide backbone folds.
Collapse
Affiliation(s)
- Faramarz Joodaki
- Department of Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Lenore M Martin
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Michael L Greenfield
- Department of Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States
| |
Collapse
|
110
|
Azmi S, Hussain MK. Analysis of structures, functions, and transgenicity of phytopeptides defensin and thionin: a review. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2021. [DOI: 10.1186/s43088-020-00093-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abstract
Background
Antimicrobial peptides are very primitive innate defense molecules of almost all organisms, from microbes to mammalians and vascular seed-bearing plants. Antimicrobial peptides of plants categorized into cysteine-rich peptides (CRPs) and others and most of the antimicrobial peptides belong to CRPs group. These peptides reported showing the great extent of protecting property against bacteria, fungi, viruses, insect, nematode, and another kind of microbes. To develop a resistant plant against pathogenic fungi, there have been several studies executed to understand the efficiency of transgenicity of these antimicrobial peptides.
Main text
Apart from the intrinsic property of the higher organism for identifying and activating microbial attack defense device, it also involves innate defense mechanism and molecules. In the current review article, apart from the structural and functional characterization of peptides defensin and thionin, we have attempted to provide a succinct overview of the transgenic development of these defense peptides, that are expressed in a constitutive and or over-expressive manner when biotic and abiotic stress inflicted. Transgenic of different peptides show different competence in plants. Most of the transgenic studies made for defensin and thionin revealed the effective transgenic capacity of these peptides.
Conclusion
There have been several studies reported successful development of transgenic plants based on peptides defensin and thionin and observed diverse level of resistance-conferring potency in different plants against phytopathogenic fungi. But due to long regulatory process, there has not been marketed any antimicrobial peptides based transgenic plants yet. However, success report state that possibly in near future transgenic plants of AMPs would be released with devoid of harmful effect, with good efficiency, reproducibility, stability, and least production cost.
Collapse
|
111
|
Benfield AH, Henriques ST. Mode-of-Action of Antimicrobial Peptides: Membrane Disruption vs. Intracellular Mechanisms. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:610997. [PMID: 35047892 PMCID: PMC8757789 DOI: 10.3389/fmedt.2020.610997] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/20/2020] [Indexed: 12/28/2022] Open
Abstract
Antimicrobial peptides are an attractive alternative to traditional antibiotics, due to their physicochemical properties, activity toward a broad spectrum of bacteria, and mode-of-actions distinct from those used by current antibiotics. In general, antimicrobial peptides kill bacteria by either disrupting their membrane, or by entering inside bacterial cells to interact with intracellular components. Characterization of their mode-of-action is essential to improve their activity, avoid resistance in bacterial pathogens, and accelerate their use as therapeutics. Here we review experimental biophysical tools that can be employed with model membranes and bacterial cells to characterize the mode-of-action of antimicrobial peptides.
Collapse
|
112
|
Jia F, Wang J, Zhang L, Zhou J, He Y, Lu Y, Liu K, Yan W, Wang K. Multiple action mechanism and in vivo antimicrobial efficacy of antimicrobial peptide Jelleine-I. J Pept Sci 2020; 27:e3294. [PMID: 33283388 DOI: 10.1002/psc.3294] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/09/2020] [Accepted: 11/19/2020] [Indexed: 12/28/2022]
Abstract
With the extensive use of antibiotics in medicine, agriculture and food chemistry, the emergence of multi-drug resistant bacteria become more and more frequent and posed great threats to human health and life. So novel antimicrobial agents were urgently needed to defend the resistant bacteria. Jelleine-I was a small antimicrobial peptide (AMP) with eight amino acids in its sequence. It was believed to be an ideal template for developing antimicrobial agents. In the present study, the possible action mode against both gram-negative bacteria and gram-positive bacteria and in vivo antimicrobial activity was explored. Our results showed that Jelleine-I exhibits its antimicrobial activity mainly by disrupting the integrity of the cell membrane, which would not be affected by the conventional resistant mechanism. It also aims at some intracellular targets such as genomic DNA to inhibit the growth of microbes. In addition, the result of in vivo antimicrobial activity experiment showed that Jelleine-I performed a good therapeutic effect toward the mice with Escherichia coli infected peritonitis. Notably, Jelleine-I has negligible cytotoxicity toward the tested mammalian cells, indicating excellent cell selectivity between prokaryotic cells and eurkayotic cells. In summary, our results showed that Jelleine-I would be a potential candidate to be developed as a novel antimicrobial agent.
Collapse
Affiliation(s)
- Fengjing Jia
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Jiayi Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Lishi Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Jingjing Zhou
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Yuhang He
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Yaqi Lu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Kexin Liu
- School/Hospital of Stomatology, Lanzhou University, West Donggang Road 199, Lanzhou, 730000, China
| | - Wenjin Yan
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Kairong Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| |
Collapse
|
113
|
Kozon D, Bednarczyk P, Szewczyk A, Jańczewski D. Regulation of Lipid Bilayer Ion Permeability by Antibacterial Polymethyloxazoline-Polyethyleneimine Copolymers. Chembiochem 2020; 22:1020-1029. [PMID: 33124737 DOI: 10.1002/cbic.202000656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/28/2020] [Indexed: 01/08/2023]
Abstract
Amphiphilic antimicrobial polymers display activity against the outer bacterial cell membrane, triggering various physiological effects. We investigated the regulation of ion transport across the lipid bilayer to understand differences in biological activity for a series of amphiphilic polymethyloxazoline - polyethyleneimine copolymers. The results confirmed that the tested structures were able to increase the permeability of the lipid bilayer (LB) membrane or its rupture. Black lipid membrane (BLM) experiments show that the triggered conductance profile and its character is strongly correlated with the polymer structure and zeta potential. The polymer exhibiting the highest antimicrobial activity promotes ion transport by using a unique mechanism and step-like characteristics with well-defined discreet openings and closings. The molecule was incorporated into the membrane in a reproducible way, and the observed channel-like activity could be responsible for the antibacterial activity of this molecule.
Collapse
Affiliation(s)
- Dominika Kozon
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland
| | - Piotr Bednarczyk
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-787, Warsaw, Poland
| | - Adam Szewczyk
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093, Warsaw, Poland
| | - Dominik Jańczewski
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland
| |
Collapse
|
114
|
Antimicrobial and antitumor activity of peptidomimetics synthesized from amino acids. Bioorg Chem 2020; 106:104506. [PMID: 33276980 DOI: 10.1016/j.bioorg.2020.104506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/24/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022]
Abstract
Thirteen cationic peptidomimetics derived from amino acids bearing an alkyl or ethynylphenyl moiety that mimic the structure of cationic antibacterial peptides were designed and synthesized using a simple coupling reaction of an amino acid with a substituted amine. Antibacterial activities of the resulting peptidomimetics against drug-sensitive bacteria, such as Gram-positive Staphylococcus aureus (S. aureus) and Bacillus subtilis, Gram-negative Escherichia coli (E. coli) and Salmonella enterica, and a drug-resistant bacterium, methicillin-resistant S. aureus (MRSA), were systematically evaluated. Most peptidomimetics show significant broad-spectrum antibacterial activity. A-L-Iso-C12 (isoleucine derivative bearing a dodecyl moiety) show MICs of 2.5 μg/mL against S. aureus and 4 μg/mL against MRSA and A-L-Val-C12 (valine derivative bearing a dodecyl moiety) show MICs of 1.67 μg/mL against E. coli and 8.3 μg/mL against MRSA. A-L-Val-C12 showed low cytotoxicity toward L929 cells in comparison with SGC 7901 cells, indicating tumor-directed killing by peptidomimetics while avoiding toxicity to normal cells. The influences of type of amino acid and substituent, length of substituent, and stereochemistry of amino acids on antibacterial activity and cytotoxicity of peptidomimetics were systematically investigated. The results indicate that this series of cationic peptidomimetics derived from amino acids display antitumor activity and may be useful for treatment of bacterial infections.
Collapse
|
115
|
Carrasquel-Ursulaez W, Reeves RD, Dehghany M, Jones C, Schomaker JM, Chanda B. Re-evaluation of the mechanism of cytotoxicity of dialkylated lariat ether compounds. RSC Adv 2020; 10:40391-40394. [PMID: 33732448 PMCID: PMC7963367 DOI: 10.1039/d0ra08494h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The cytotoxicity of dialkylated lariat ethers has been previously attributed to their ionophoric properties. Herein, we provide evidence that these effects are due to loss of membrane integrity rather than ion transport, a finding with important implications for the future design of synthetic ionophores. The cytotoxicity of dialkylated lariat ethers has been previously attributed to their ionophoric properties.![]()
Collapse
Affiliation(s)
| | - Ryan D Reeves
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin 53706, USA
| | - Mahzad Dehghany
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin 53706, USA
| | - Corey Jones
- Department of Chemistry, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | - Baron Chanda
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri 63110, USA
| |
Collapse
|
116
|
Ning Y, Fu Y, Hou L, Ma M, Wang Z, Li X, Jia Y. iTRAQ-based quantitative proteomic analysis of synergistic antibacterial mechanism of phenyllactic acid and lactic acid against Bacillus cereus. Food Res Int 2020; 139:109562. [PMID: 33509445 DOI: 10.1016/j.foodres.2020.109562] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/14/2020] [Accepted: 07/17/2020] [Indexed: 11/15/2022]
Abstract
Phenyllactic acid (PLA) as a phenolic acid by lactic acid (LA) bacteria shows enhanced antibacterial activity when coexisting with LA, while the antibacterial mechanism of PLA combined with LA was unknown. Hence, the antibacterial mechanism of PLA and LA was investigated against Bacillus cereus. Flow cytometry and TEM analysis demonstrated that single PLA and LA disrupted the membrane integrity and the morphology, while combined PLA and LA synergistically enhanced the damage. iTRAQ-based proteomic analysis suggested that PLA down-regulated kdpB and inhibited K+ transport, disturbed the function of ribosome and expression of competence genes; LA down-regulated periplasmic phosphorus-binding proteins and inhibited phosphorus transport, disturbed the function of ribosome, TCA cycle, as well as purine and pyrimidine metabolism; and combined PLA and LA inhibited K+ and phosphorus transport, and influenced the synthesis of purine and pyrimidine metabolism. The investigation could provide some insights into the application of PLA in food preservation.
Collapse
Affiliation(s)
- Yawei Ning
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Yunan Fu
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Linlin Hou
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Mengge Ma
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Zhixin Wang
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Xingfeng Li
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China.
| | - Yingmin Jia
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
117
|
Baron F, Cochet MF, Alabdeh M, Guérin-Dubiard C, Gautier M, Nau F, Andrews SC, Bonnassie S, Jan S. Egg-White Proteins Have a Minor Impact on the Bactericidal Action of Egg White Toward Salmonella Enteritidis at 45°C. Front Microbiol 2020; 11:584986. [PMID: 33133053 PMCID: PMC7578404 DOI: 10.3389/fmicb.2020.584986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 09/18/2020] [Indexed: 02/05/2023] Open
Abstract
Salmonella enterica serovar Enteritidis is noted for its ability to survive the harsh antibacterial activity of egg white which is presumed to explain its occurrence as the major food-borne pathogen associated with the consumption of eggs and egg products. Liquid egg white is a major ingredient for the food industry but, because of its thermal fragility, pasteurization is performed at the modest temperature of 57°C (for 2–6 min). Unfortunately, such treatment does not lead to sufficient reduction in S. Enteritidis contamination, which is a clear health concern when the product is consumed without cooking. However, egg white is able to limit S. Enteritidis growth due to its alkaline pH, iron deficiency and multiple antimicrobial proteins. This anti-Salmonella activity of egg white is temperature dependent and becomes bactericidal once the incubation temperature exceeds 42°C. This property is exploited in the highly promising pasteurization treatment (42–45°C for 1–5 days) which achieves complete killing of S. Enteritidis. However, the precise mechanism and the role of the egg-white proteins are not fully understood. Here, the impact of exposure of S. Enteritidis to egg white-based media, with or without egg-white proteins (>10 kDa), under bactericidal conditions (45°C) was explored by measuring survival and global expression. Surprisingly, the bactericidal activity of egg white at 45°C was only slightly affected by egg-white proteins indicating that they play a minor role in the bactericidal activity observed. Moreover, egg-white proteins had minimal impact on the global-gene-expression response to egg white such that very similar, major regulatory responses (20% genes affected) were observed both with and without egg-white proteins following incubation for 45 min at 45°C. Egg-white proteins caused a significant change in expression for just 64 genes, including the psp and lysozyme-inhibitor responses genes which is suggestive of an early membrane perturbation effect. Such damage was supported by disruption of the proton motive force by egg-white proteins. In summary, the results suggest that low-mass components of egg white are largely responsible for the bactericidal activity of egg white at 45°C.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Simon C Andrews
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Sylvie Bonnassie
- STLO, INRAE, Institut Agro, Rennes, France.,UFR Sciences de la Vie et de l'Environnement, Université de Rennes I, Rennes, France
| | - Sophie Jan
- STLO, INRAE, Institut Agro, Rennes, France
| |
Collapse
|
118
|
Effect of membrane potential on pore formation by the antimicrobial peptide magainin 2 in lipid bilayers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183381. [DOI: 10.1016/j.bbamem.2020.183381] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/28/2020] [Accepted: 05/31/2020] [Indexed: 11/19/2022]
|
119
|
Kmeck A, Tancer RJ, Ventura CR, Wiedman GR. Synergies with and Resistance to Membrane-Active Peptides. Antibiotics (Basel) 2020; 9:antibiotics9090620. [PMID: 32961656 PMCID: PMC7559582 DOI: 10.3390/antibiotics9090620] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/07/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Membrane-active peptides (MAPs) have long been thought of as the key to defeating antimicrobial-resistant microorganisms. Such peptides, however, may not be sufficient alone. In this review, we seek to highlight some of the common pathways for resistance, as well as some avenues for potential synergy. This discussion takes place considering resistance, and/or synergy in the extracellular space, at the membrane, and during interaction, and/or removal. Overall, this review shows that researchers require improved definitions of resistance and a more thorough understanding of MAP-resistance mechanisms. The solution to combating resistance may ultimately come from an understanding of how to harness the power of synergistic drug combinations.
Collapse
|
120
|
Yu TT, Kuppusamy R, Yasir M, Hassan MM, Alghalayini A, Gadde S, Deplazes E, Cranfield C, Willcox MD, Black DS, Kumar N. Design, Synthesis and Biological Evaluation of Biphenylglyoxamide-Based Small Molecular Antimicrobial Peptide Mimics as Antibacterial Agents. Int J Mol Sci 2020; 21:E6789. [PMID: 32947921 PMCID: PMC7555970 DOI: 10.3390/ijms21186789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 01/14/2023] Open
Abstract
There has been an increasing interest in the development of antimicrobial peptides (AMPs) and their synthetic mimics as a novel class of antibiotics to overcome the rapid emergence of antibiotic resistance. Recently, phenylglyoxamide-based small molecular AMP mimics have been identified as potential leads to treat bacterial infections. In this study, a new series of biphenylglyoxamide-based small molecular AMP mimics were synthesised from the ring-opening reaction of N-sulfonylisatin bearing a biphenyl backbone with a diamine, followed by the conversion into tertiary ammonium chloride, quaternary ammonium iodide and guanidinium hydrochloride salts. Structure-activity relationship studies of the analogues identified the octanesulfonyl group as being essential for both Gram-positive and Gram-negative antibacterial activity, while the biphenyl backbone was important for Gram-negative antibacterial activity. The most potent analogue was identified to be chloro-substituted quaternary ammonium iodide salt 15c, which possesses antibacterial activity against both Gram-positive (MIC against Staphylococcus aureus = 8 μM) and Gram-negative bacteria (MIC against Escherichia coli = 16 μM, Pseudomonas aeruginosa = 63 μM) and disrupted 35% of pre-established S. aureus biofilms at 32 μM. Cytoplasmic membrane permeability and tethered bilayer lipid membranes (tBLMs) studies suggested that 15c acts as a bacterial membrane disruptor. In addition, in vitro toxicity studies showed that the potent compounds are non-toxic against human cells at therapeutic dosages.
Collapse
Affiliation(s)
- Tsz Tin Yu
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia; (T.T.Y.); (R.K.); (M.M.H.); (S.G.)
| | - Rajesh Kuppusamy
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia; (T.T.Y.); (R.K.); (M.M.H.); (S.G.)
| | - Muhammad Yasir
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia; (M.Y.); (M.D.P.W.)
| | - Md. Musfizur Hassan
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia; (T.T.Y.); (R.K.); (M.M.H.); (S.G.)
| | - Amani Alghalayini
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia; (A.A.); (E.D.); (C.C.)
| | - Satyanarayana Gadde
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia; (T.T.Y.); (R.K.); (M.M.H.); (S.G.)
| | - Evelyne Deplazes
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia; (A.A.); (E.D.); (C.C.)
| | - Charles Cranfield
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia; (A.A.); (E.D.); (C.C.)
| | - Mark D.P. Willcox
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia; (M.Y.); (M.D.P.W.)
| | - David StC Black
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia; (T.T.Y.); (R.K.); (M.M.H.); (S.G.)
| | - Naresh Kumar
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia; (T.T.Y.); (R.K.); (M.M.H.); (S.G.)
| |
Collapse
|
121
|
Stokes JM, Yang K, Swanson K, Jin W, Cubillos-Ruiz A, Donghia NM, MacNair CR, French S, Carfrae LA, Bloom-Ackermann Z, Tran VM, Chiappino-Pepe A, Badran AH, Andrews IW, Chory EJ, Church GM, Brown ED, Jaakkola TS, Barzilay R, Collins JJ. A Deep Learning Approach to Antibiotic Discovery. Cell 2020; 180:688-702.e13. [PMID: 32084340 DOI: 10.1016/j.cell.2020.01.021] [Citation(s) in RCA: 921] [Impact Index Per Article: 184.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/04/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
Due to the rapid emergence of antibiotic-resistant bacteria, there is a growing need to discover new antibiotics. To address this challenge, we trained a deep neural network capable of predicting molecules with antibacterial activity. We performed predictions on multiple chemical libraries and discovered a molecule from the Drug Repurposing Hub-halicin-that is structurally divergent from conventional antibiotics and displays bactericidal activity against a wide phylogenetic spectrum of pathogens including Mycobacterium tuberculosis and carbapenem-resistant Enterobacteriaceae. Halicin also effectively treated Clostridioides difficile and pan-resistant Acinetobacter baumannii infections in murine models. Additionally, from a discrete set of 23 empirically tested predictions from >107 million molecules curated from the ZINC15 database, our model identified eight antibacterial compounds that are structurally distant from known antibiotics. This work highlights the utility of deep learning approaches to expand our antibiotic arsenal through the discovery of structurally distinct antibacterial molecules.
Collapse
Affiliation(s)
- Jonathan M Stokes
- Department of Biological Engineering, Synthetic Biology Center, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Machine Learning for Pharmaceutical Discovery and Synthesis Consortium, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kevin Yang
- Machine Learning for Pharmaceutical Discovery and Synthesis Consortium, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kyle Swanson
- Machine Learning for Pharmaceutical Discovery and Synthesis Consortium, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wengong Jin
- Machine Learning for Pharmaceutical Discovery and Synthesis Consortium, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andres Cubillos-Ruiz
- Department of Biological Engineering, Synthetic Biology Center, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Nina M Donghia
- Department of Biological Engineering, Synthetic Biology Center, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Craig R MacNair
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Shawn French
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Lindsey A Carfrae
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Zohar Bloom-Ackermann
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Victoria M Tran
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anush Chiappino-Pepe
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Ahmed H Badran
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ian W Andrews
- Department of Biological Engineering, Synthetic Biology Center, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Emma J Chory
- Department of Biological Engineering, Synthetic Biology Center, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Eric D Brown
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Tommi S Jaakkola
- Machine Learning for Pharmaceutical Discovery and Synthesis Consortium, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Regina Barzilay
- Machine Learning for Pharmaceutical Discovery and Synthesis Consortium, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Abdul Latif Jameel Clinic for Machine Learning in Health, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - James J Collins
- Department of Biological Engineering, Synthetic Biology Center, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA 02139, USA; Abdul Latif Jameel Clinic for Machine Learning in Health, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
122
|
Use of a Fluorescence-Based Assay To Measure Escherichia coli Membrane Potential Changes in High Throughput. Antimicrob Agents Chemother 2020; 64:AAC.00910-20. [PMID: 32631824 DOI: 10.1128/aac.00910-20] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/30/2020] [Indexed: 11/20/2022] Open
Abstract
Bacterial membrane potential is difficult to measure using classical electrophysiology techniques due to the small cell size and the presence of the peptidoglycan cell wall. Instead, chemical probes are often used to study membrane potential changes under conditions of interest. Many of these probes are fluorescent molecules that accumulate in a charge-dependent manner, and the resulting fluorescence change can be analyzed via flow cytometry or using a fluorescence microplate reader. Although this technique works well in many Gram-positive bacteria, it generates fairly low signal-to-noise ratios in Gram-negative bacteria due to dye exclusion by the outer membrane. We detail an optimized workflow that uses the membrane potential probe, 3,3'-diethyloxacarbocyanine iodide [DiOC2(3)], to measure Escherichia coli membrane potential changes in high throughput and describe the assay conditions that generate significant signal-to-noise ratios to detect membrane potential changes using a fluorescence microplate reader. A valinomycin calibration curve demonstrates this approach can robustly report membrane potentials over at least an ∼144-mV range with an accuracy of ∼12 mV. As a proof of concept, we used this approach to characterize the effects of some commercially available small molecules known to elicit membrane potential changes in other systems, increasing the repertoire of compounds known to perturb E. coli membrane energetics. One compound, the eukaryotic Ca2+ channel blocker amlodipine, was found to alter E. coli membrane potential and decrease the MIC of kanamycin, further supporting the value of this screening approach. This detailed methodology permits studying E. coli membrane potential changes quickly and reliably at the population level.
Collapse
|
123
|
Rathman BM, Allen JL, Shaw LN, Del Valle JR. Synthesis and biological evaluation of backbone-aminated analogues of gramicidin S. Bioorg Med Chem Lett 2020; 30:127283. [PMID: 32527462 DOI: 10.1016/j.bmcl.2020.127283] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 01/26/2023]
Abstract
We report the parallel synthesis of gramicidin S derivatives featuring backbone N-amino substituents. Analogues were prepared by incorporation of N-amino dipeptide subunits on solid support. Nine backbone-aminated macrocycles were evaluated for growth inhibitory activity against ESKAPE pathogens and hemolytic activity against human red blood cells. Diamination of the Orn residues in the β-strand region of gramicidin S was found to enhance broad-spectrum antimicrobial activity without a corresponding increase in hemolytic activity.
Collapse
Affiliation(s)
- Benjamin M Rathman
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Jessie L Allen
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Lindsey N Shaw
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Juan R Del Valle
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
124
|
Magana M, Pushpanathan M, Santos AL, Leanse L, Fernandez M, Ioannidis A, Giulianotti MA, Apidianakis Y, Bradfute S, Ferguson AL, Cherkasov A, Seleem MN, Pinilla C, de la Fuente-Nunez C, Lazaridis T, Dai T, Houghten RA, Hancock REW, Tegos GP. The value of antimicrobial peptides in the age of resistance. THE LANCET. INFECTIOUS DISEASES 2020; 20:e216-e230. [PMID: 32653070 DOI: 10.1016/s1473-3099(20)30327-3] [Citation(s) in RCA: 676] [Impact Index Per Article: 135.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/29/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022]
Abstract
Accelerating growth and global expansion of antimicrobial resistance has deepened the need for discovery of novel antimicrobial agents. Antimicrobial peptides have clear advantages over conventional antibiotics which include slower emergence of resistance, broad-spectrum antibiofilm activity, and the ability to favourably modulate the host immune response. Broad bacterial susceptibility to antimicrobial peptides offers an additional tool to expand knowledge about the evolution of antimicrobial resistance. Structural and functional limitations, combined with a stricter regulatory environment, have hampered the clinical translation of antimicrobial peptides as potential therapeutic agents. Existing computational and experimental tools attempt to ease the preclinical and clinical development of antimicrobial peptides as novel therapeutics. This Review identifies the benefits, challenges, and opportunities of using antimicrobial peptides against multidrug-resistant pathogens, highlights advances in the deployment of novel promising antimicrobial peptides, and underlines the needs and priorities in designing focused development strategies taking into account the most advanced tools available.
Collapse
Affiliation(s)
- Maria Magana
- Department of Biopathology and Clinical Microbiology, Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Ana L Santos
- Department of Chemistry, Rice University, Houston, TX, USA; Investigación Sanitaria de las Islas Baleares, Palma, Spain
| | - Leon Leanse
- Department of Dermatology, Harvard Medical School, Boston, MA, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Michael Fernandez
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | - Steven Bradfute
- Department of Internal Medicine, Center for Global Health, University of New Mexico, Albuquerque, NM, USA
| | - Andrew L Ferguson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Artem Cherkasov
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Mohamed N Seleem
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, IN, USA
| | - Clemencia Pinilla
- Torrey Pines Institute for Molecular Studies, Port St Lucie, FL, USA
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Penn Institute for Computational Science, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Themis Lazaridis
- Department of Chemistry, The City College of New York, New York, NY, USA; Graduate Programs in Chemistry, Biochemistry, and Physics, The Graduate Center, City University of New York, NY, USA
| | - Tianhong Dai
- Department of Dermatology, Harvard Medical School, Boston, MA, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Robert E W Hancock
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - George P Tegos
- Reading Hospital, Tower Health, West Reading, PA, USA; Micromoria, Venture X Marlborough, Marlborough, MA, USA.
| |
Collapse
|
125
|
French S, Farha M, Ellis MJ, Sameer Z, Côté JP, Cotroneo N, Lister T, Rubio A, Brown ED. Potentiation of Antibiotics against Gram-Negative Bacteria by Polymyxin B Analogue SPR741 from Unique Perturbation of the Outer Membrane. ACS Infect Dis 2020; 6:1405-1412. [PMID: 31566948 DOI: 10.1021/acsinfecdis.9b00159] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Therapeutics targeting Gram-negative bacteria have the challenge of overcoming a formidable outer membrane (OM) barrier. Here, we characterize the action of SPR741, a novel polymyxin B (PMB) analogue shown to potentiate several large-scaffold antibiotics in Gram-negative pathogens. Probing the surface topology of Escherichia coli using atomic force microscopy revealed substantial OM disorder at concentrations of SPR741 that lead to antibiotic potentiation. Conversely, very little cytoplasmic membrane depolarization was observed at these same concentrations, indicating that SPR741 acts predominately on the OM. Truncating the lipopolysaccharide (LPS) core with genetic perturbations uniquely sensitized E. coli to SPR741, suggesting that LPS core residues keep SPR741 at the OM, where it can potentiate a codrug, rather than permit its entry to the cytoplasmic membrane. Further, a promoter activity assay revealed that SPR741 challenge induced the expression of RcsAB, a stress sensor for OM perturbation. Together, these results indicate that SPR741 interacts predominately with the OM, in contrast to the dual action of PMB and colistin at both the outer and cytoplasmic membranes.
Collapse
Affiliation(s)
- Shawn French
- Department of Biochemistry and Biomedical Science and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Maya Farha
- Department of Biochemistry and Biomedical Science and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Michael J. Ellis
- Department of Biochemistry and Biomedical Science and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Zaid Sameer
- Department of Biochemistry and Biomedical Science and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Jean-Philippe Côté
- Department of Biochemistry and Biomedical Science and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Nicole Cotroneo
- Spero Therapeutics, 675 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Troy Lister
- Spero Therapeutics, 675 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Aileen Rubio
- Spero Therapeutics, 675 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Eric D. Brown
- Department of Biochemistry and Biomedical Science and Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
126
|
|
127
|
Ogan A, Yüce‐Dursun B, Abdullah D, Beyler‐Çiğil A, Kahraman MV, Çağlayan P, Birbir M, Mutlu Ö, Gülsoy N. Preparation and antimicrobial properties of LL‐37 peptide immobilized lignin/caprolactone polymer film. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.4942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ayşe Ogan
- Faculty of Arts and Sciences, Department of ChemistryMarmara University Istanbul Turkey
| | - Başak Yüce‐Dursun
- Faculty of Arts and Sciences, Department of ChemistryMarmara University Istanbul Turkey
| | - Deka Abdullah
- Faculty of Arts and Sciences, Department of ChemistryMarmara University Istanbul Turkey
| | - Aslı Beyler‐Çiğil
- Department of Chemistry and Chemical Process Technology SchoolAmasya University Technical Sciences Vocational Amasya Turkey
| | - Memet Vezir Kahraman
- Faculty of Arts and Sciences, Department of ChemistryMarmara University Istanbul Turkey
| | - Pınar Çağlayan
- Faculty of Arts and Sciences, Department of BiologyMarmara University Istanbul Turkey
| | - Meral Birbir
- Faculty of Arts and Sciences, Department of BiologyMarmara University Istanbul Turkey
| | - Özal Mutlu
- Faculty of Arts and Sciences, Department of BiologyMarmara University Istanbul Turkey
| | - Nagihan Gülsoy
- Faculty of Arts and Sciences, Department of BiologyMarmara University Istanbul Turkey
| |
Collapse
|
128
|
Strawberry fatty acyl glycosides enhance disease protection, have antibiotic activity and stimulate plant growth. Sci Rep 2020; 10:8196. [PMID: 32424195 PMCID: PMC7235083 DOI: 10.1038/s41598-020-65125-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
An increasing interest in the development of products of natural origin for crop disease and pest control has emerged in the last decade. Here we introduce a new family of strawberry acyl glycosides (SAGs) formed by a trisaccharide (GalNAc-GalNAc-Glc) and a monounsaturated fatty acid of 6 to 12 carbon atoms linked to the glucose unit. Application of SAGs to Arabidopsis thaliana (hereafter Arabidopsis) plants triggered a transient oxidative burst, callose deposition and defense gene expression, accompanied by increased protection against two phytopathogens, Pseudomonas viridiflava and Botrytis cinerea. SAGs-induced disease protection was also demonstrated in soybean infected with the causal agent of target spot, Corynespora cassiicola. SAGs were shown to exhibit important antimicrobial activity against a wide-range of bacterial and fungal phytopathogens, most probably through membrane destabilization, and the potential use of SAGs as a biofungicide for postharvest disease protection was demonstrated on lemon fruits infected with Penicillium digitatum. Plant growth promotion by application of SAGs was shown by augmented primary root elongation, secondary roots development and increased siliques formation in Arabidopsis, whereas a significant increment in number of seed pods was demonstrated in soybean. Stimulation of radicle development and the induction of an auxin-responsive reporter system (DR5::GUS) in transgenic Arabidopsis plants, suggested that SAGs-stimulated growth at least partly acts through the auxin response pathway. These results indicate that strawberry fatty acid glycosides are promising candidates for the development of environmental-friendly products for disease management in soybean and lemon.
Collapse
|
129
|
Aziz M, Garduno R, Mirani ZA, Baqai R, Sheikh AS, Nazir H, Raza Y, Ayaz M, Kazmi SU. Determination of antimicrobial effect of protamine by transmission electron microscopy and SDS PAGE on Pseudomonas aeruginosa isolates from diabetic foot infection. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 22:827-832. [PMID: 32373306 PMCID: PMC7196347 DOI: 10.22038/ijbms.2019.32414.7989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Objective(s): Diabetic foot infection is one of the major complications of diabetes leading to lower limb amputations. Isolation and identification of bacteria causing diabetic foot infection, determination of antibiotic resistance, antimicrobial potential of protamine by electron microscopy and SDS-PAGE analysis, arethe aims of this study. Materials and Methods: 285 pus samples from diabetic foot infection patients were collected from different hospitals of Karachi and Capital Health Hospital, Halifax, Canada. Clinical history of each patient was recorded. Bacterial isolates were cultured on appropriate media; identification was done by morphology, cultural and biochemical tests. Effect of protamine against multi drug resistant strains of Pseudomona aeruginosa was checked by minimum inhibitory concentration in 96 well micro-titer plates. The isolates were grown in bactericidal concentration of protamine on plates to isolate mutants. Effect of protamine on protein expression was checked by SDS- PAGE and ultra-structural morphological changes by transmission electron microscopy. Results: Results indicated prevalence of foot infection as 92% in diabetic patients. Major bacterial isolates were Staphylococcus aureus 65 (23%), P. aeruginosa 80 (28.1%), Klebsiella spp. 37 (13%), Proteus mirabilis 79 (27.7%), and Escherichia coli 24 (12%). These isolates were highly resistant to different antibiotics. MIC value of protamine was 500 µg/ml against P. aeruginosa. SDS-PAGE analysis revealed that protamine can suppress expression of various virulence proteins and electron micrographs indicated condensation of cytoplasm and accumulation of protamine in cytoplasm without damaging the cell membrane. Conclusion: P. aeruginosa and S. aureus were the major isolates expressing multi-drug resistance and protamine sulfate represented good antimicrobial potential.
Collapse
Affiliation(s)
- Mubashar Aziz
- Department of Pathobiology, Bahauddin Zakariya University, Multan, Pakistan.,Department of Microbiology, University of Karachi, Karachi, Pakistan
| | - Rafael Garduno
- Department of Microbiology, Dalhousie University, Halifax, Canada
| | | | - Rakhshanda Baqai
- Department of Microbiology, University of Karachi, Karachi, Pakistan.,Departmet of Clinical Microbiology & Immunology, Dadabhoy Institute of Higher Education, Karachi, Pakistan
| | - Ahsan Sattar Sheikh
- Institute of Food Science and Nutrition, Bahauddin Zakariya University, Multan, Pakistan
| | | | - Yasir Raza
- Department of Microbiology, University of Karachi, Karachi, Pakistan
| | - Mazhar Ayaz
- Department of Pathobiology, Bahauddin Zakariya University, Multan, Pakistan
| | - Shahana Urooj Kazmi
- Department of Microbiology, University of Karachi, Karachi, Pakistan.,Departmet of Clinical Microbiology & Immunology, Dadabhoy Institute of Higher Education, Karachi, Pakistan
| |
Collapse
|
130
|
Moghal MMR, Hossain F, Yamazaki M. Action of antimicrobial peptides and cell-penetrating peptides on membrane potential revealed by the single GUV method. Biophys Rev 2020; 12:339-348. [PMID: 32152921 PMCID: PMC7242587 DOI: 10.1007/s12551-020-00662-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/23/2020] [Indexed: 02/06/2023] Open
Abstract
Membrane potential plays various key roles in live bacterial and eukaryotic cells. So far, the effects of membrane potential on action of antimicrobial peptides (AMPs) and cell-penetrating peptides (CPPs) have been examined using cells and small lipid vesicles. However, due to the technical drawbacks of these experiments, the effect of membrane potential on the actions of AMPs and CPPs and the elementary processes of interactions of these peptides with cell membranes and vesicle membranes are not well understood. In this short review, we summarize the results of the effect of membrane potential on the action of an AMP, lactoferricin B (LfcinB), and a CPP, transportan 10 (TP10), in vesicle membranes revealed by the single giant unilamellar vesicle (GUV) method. Parts of the actions and their elementary steps of AMPs and CPPs interacting vesicle membranes under membrane potential are clearly revealed using the single GUV method. The experimental methods and their analysis described here can be used to elucidate the effects of membrane potential on various activities of peptides such as AMPs, CPPs, and proteins. Moreover, GUVs with membrane potential are more suitable as a model of cells or artificial cells, as well as GUVs containing small vesicles.
Collapse
Affiliation(s)
- Md Mizanur Rahman Moghal
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, 422-8529, Japan
- Department of Pharmacy, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Farzana Hossain
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, 422-8529, Japan
| | - Masahito Yamazaki
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, 422-8529, Japan.
- Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, 836 Oya, Suruga-ku, Shizuoka, 422-8529, Japan.
- Department of Physics, Faculty of Science, Shizuoka University, Shizuoka, 422-8529, Japan.
| |
Collapse
|
131
|
Krishnakumari V, Binny TM, Adicherla H, Nagaraj R. Escherichia coli Lipopolysaccharide Modulates Biological Activities of Human-β-Defensin Analogues but Not Non-Ribosomally Synthesized Peptides. ACS OMEGA 2020; 5:6366-6375. [PMID: 32258871 PMCID: PMC7114172 DOI: 10.1021/acsomega.9b03770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/04/2020] [Indexed: 06/11/2023]
Abstract
Human-β-defensins (HBD1-3) are antibacterial peptides containing three disulphide bonds. In the present study, the effect of Escherichia coli lipopolysaccharide (LPS) on the antibacterial activities of HBD2-3, C-terminal analogues having a single disulphide bond, Phd1-3, and their corresponding myristoylated analogues MPhd1-3 were investigated. The effect of LPS on the activities of linear amphipathic peptides melittin, LL37 and non-ribosomally synthesized peptides, polymyxin B, alamethicin, gramicidin A, and gramicidin S was also examined. The antibacterial activity of HBD 2-3, Phd1-3, and MPhd1-3 in the presence of LPS against E. coli and Staphylococcus aureus was inhibited. While LPS inhibited the antibacterial activity of LL37, the inhibition of melittin activity was partial. The hemolytic activity exhibited by MPhd1, MPhd3, melittin, and LL37 was inhibited in the presence of LPS. HBD2-3, Phd1-3, and MPhd1-3 also showed endotoxin neutralizing activity. The antibacterial and hemolytic activities of polymyxin B, alamethicin, gramicidin A, and gramicidin S were not inhibited in the presence of LPS. Fluorescence assays employing dansyl cadaverine showed that HBD2-3 and defensin analogues bind to LPS more strongly as compared to alamethicin, gramicidin A, and gramicidin S. Electron microscopy images indicated that peptides disintegrate the structure of LPS. The inhibition of the antibacterial activity of native defensins and analogues in the presence of LPS indicates that the initial interaction with the bacterial surface is similar. The native defensin sequence or structure is also not essential, although cationic charges are necessary for binding to LPS. Hydrophobic interaction is the main driving force for association of non-ribosomally synthesized polymyxin B, alamethicin, gramicidin A, and gramicidin S with LPS. It is likely that these peptides rapidly insert into membranes and do not interact with the bacterial cell surface, whereas cationic peptides such as β-defensin and their analogues, melittin and LL37, first interact with the bacterial cell surface and then the membrane. Our results suggest that evaluating interaction of antibacterial and hemolytic peptides with LPS is a compelling way of elucidating the mechanism of bacterial killing or hemolysis.
Collapse
|
132
|
Fernández de Ullivarri M, Arbulu S, Garcia-Gutierrez E, Cotter PD. Antifungal Peptides as Therapeutic Agents. Front Cell Infect Microbiol 2020; 10:105. [PMID: 32257965 PMCID: PMC7089922 DOI: 10.3389/fcimb.2020.00105] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
Fungi have been used since ancient times in food and beverage-making processes and, more recently, have been harnessed for the production of antibiotics and in processes of relevance to the bioeconomy. Moreover, they are starting to gain attention as a key component of the human microbiome. However, fungi are also responsible for human infections. The incidence of community-acquired and nosocomial fungal infections has increased considerably in recent decades. Antibiotic resistance development, the increasing number of immunodeficiency- and/or immunosuppression-related diseases and limited therapeutic options available are triggering the search for novel alternatives. These new antifungals should be less toxic for the host, with targeted or broader antimicrobial spectra (for diseases of known and unknown etiology, respectively) and modes of actions that limit the potential for the emergence of resistance among pathogenic fungi. Given these criteria, antimicrobial peptides with antifungal properties, i.e., antifungal peptides (AFPs), have emerged as powerful candidates due to their efficacy and high selectivity. In this review, we provide an overview of the bioactivity and classification of AFPs (natural and synthetic) as well as their mode of action and advantages over current antifungal drugs. Additionally, natural, heterologous and synthetic production of AFPs with a view to greater levels of exploitation is discussed. Finally, we evaluate the current and potential applications of these peptides, along with the future challenges relating to antifungal treatments.
Collapse
Affiliation(s)
- Miguel Fernández de Ullivarri
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland
| | - Sara Arbulu
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland
| | - Enriqueta Garcia-Gutierrez
- Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland.,Gut Microbes and Health, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Paul D Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland
| |
Collapse
|
133
|
Cho HS, Yum J, Larivière A, Lévêque N, Le QVC, Ahn B, Jeon H, Hong K, Soundrarajan N, Kim JH, Bodet C, Park C. Opossum Cathelicidins Exhibit Antimicrobial Activity Against a Broad Spectrum of Pathogens Including West Nile Virus. Front Immunol 2020; 11:347. [PMID: 32194564 PMCID: PMC7063992 DOI: 10.3389/fimmu.2020.00347] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/13/2020] [Indexed: 12/14/2022] Open
Abstract
This study aimed to characterize cathelicidins from the gray short-tailed opossum in silico and experimentally validate their antimicrobial effects against various pathogenic bacteria and West Nile virus (WNV). Genome-wide in silico analysis against the current genome assembly of the gray short-tailed opossum yielded 56 classical antimicrobial peptides (AMPs) from eight different families, among which 19 cathelicidins, namely ModoCath1 – 19, were analyzed in silico to predict their antimicrobial domains and three of which, ModoCath1, -5, and -6, were further experimentally evaluated for their antimicrobial activity, and were found to exhibit a wide spectrum of antimicroial effects against a panel of gram-positive and gram-negative bacterial strains. In addition, these peptides displayed low-to-moderate cytotoxicity in mammalian cells as well as stability in serum and various salt and pH conditions. Circular dichroism analysis of the spectra resulting from interactions between ModoCaths and lipopolysaccharides (LPS) showed formation of a helical structure, while a dual-dye membrane disruption assay and scanning electron microscopy analysis revealed that ModoCaths exerted bactericidal effects by causing membrane damage. Furthermore, ModoCath5 displayed potent antiviral activity against WNV by inhibiting viral replication, suggesting that opossum cathelicidins may serve as potentially novel antimicrobial endogenous substances of mammalian origin, considering their large number. Moreover, analysis of publicly available RNA-seq data revealed the expression of eight ModoCaths from five different tissues, suggesting that gray short-tailed opossums may be an interesting source of cathelicidins with diverse characteristics.
Collapse
Affiliation(s)
- Hye-Sun Cho
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, South Korea
| | - Joori Yum
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, South Korea
| | - Andy Larivière
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, LITEC EA 4331, Université de Poitiers, Poitiers, France
| | - Nicolas Lévêque
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, LITEC EA 4331, Université de Poitiers, Poitiers, France
| | - Quy Van Chanh Le
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, South Korea
| | - ByeongYong Ahn
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, South Korea
| | - Hyoim Jeon
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, South Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, South Korea
| | | | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, South Korea
| | - Charles Bodet
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, LITEC EA 4331, Université de Poitiers, Poitiers, France
| | - Chankyu Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, South Korea
| |
Collapse
|
134
|
Boix-Lemonche G, Lekka M, Skerlavaj B. A Rapid Fluorescence-Based Microplate Assay to Investigate the Interaction of Membrane Active Antimicrobial Peptides with Whole Gram-Positive Bacteria. Antibiotics (Basel) 2020; 9:antibiotics9020092. [PMID: 32093104 PMCID: PMC7168298 DOI: 10.3390/antibiotics9020092] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 01/08/2023] Open
Abstract
Background: Membrane-active antimicrobial peptides (AMPs) are interesting candidates for the development of novel antimicrobials. Although their effects were extensively investigated in model membrane systems, interactions of AMPs with living microbial membranes are less known due to their complexity. The aim of the present study was to develop a rapid fluorescence-based microplate assay to analyze the membrane effects of AMPs in whole Staphylococcus aureus and Staphylococcus epidermidis. Methods: Bacteria were exposed to bactericidal and sub-inhibitory concentrations of two membrane-active AMPs in the presence of the potential-sensitive dye 3,3′-dipropylthiadicarbocyanine iodide (diSC3(5)) and the DNA staining dye propidium iodide (PI), to simultaneously monitor and possibly distinguish membrane depolarization and membrane permeabilization. Results: The ion channel-forming gramicidin D induced a rapid increase of diSC3(5), but not PI fluorescence, with slower kinetics at descending peptide concentrations, confirming killing due to membrane depolarization. The pore-forming melittin, at sub-MIC and bactericidal concentrations, caused, respectively, an increase of PI fluorescence in one or both dyes simultaneously, suggesting membrane permeabilization as a key event. Conclusions: This assay allowed the distinction between specific membrane effects, and it could be applied in the mode of action studies as well as in the screening of novel membrane-active AMPs.
Collapse
Affiliation(s)
- Gerard Boix-Lemonche
- Department of Medicine, University of Udine, Piazzale Kolbe, 4, 33100 Udine, Italy
- Correspondence: (G.B.-L.); (B.S.)
| | - Maria Lekka
- Polytechnic Department of Engineering and Architecture, University of Udine, Via delle Scienze 206, 33100 Udine, Italy
| | - Barbara Skerlavaj
- Department of Medicine, University of Udine, Piazzale Kolbe, 4, 33100 Udine, Italy
- Correspondence: (G.B.-L.); (B.S.)
| |
Collapse
|
135
|
Berditsch M, Afonin S, Reuster J, Lux H, Schkolin K, Babii O, Radchenko DS, Abdullah I, William N, Middel V, Strähle U, Nelson A, Valko K, Ulrich AS. Supreme activity of gramicidin S against resistant, persistent and biofilm cells of staphylococci and enterococci. Sci Rep 2019; 9:17938. [PMID: 31784584 PMCID: PMC6884456 DOI: 10.1038/s41598-019-54212-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/06/2019] [Indexed: 01/03/2023] Open
Abstract
Three promising antibacterial peptides were studied with regard to their ability to inhibit the growth and kill the cells of clinical strains of Staphylococcus aureus, Enterococcus faecalis and Enterococcus faecium. The multifunctional gramicidin S (GS) was the most potent, compared to the membranotropic temporin L (TL), being more effective than the innate-defence regulator IDR-1018 (IDR). These activities, compared across 16 strains as minimal bactericidal and minimal inhibitory concentrations (MIC), are independent of bacterial resistance pattern, phenotype variations and/or biofilm-forming potency. For S. aureus strains, complete killing is accomplished by all peptides at 5 × MIC. For E. faecalis strains, only GS exhibits a rapid bactericidal effect at 5 × MIC, while TL and IDR require higher concentrations. The biofilm-preventing activities of all peptides against the six strains with the largest biofilm biomass were compared. GS demonstrates the lowest minimal biofilm inhibiting concentrations, whereas TL and IDR are consistently less effective. In mature biofilms, only GS completely kills the cells of all studied strains. We compare the physicochemical properties, membranolytic activities, model pharmacokinetics and eukaryotic toxicities of the peptides and explain the bactericidal, antipersister and antibiofilm activities of GS by its elevated stability, pronounced cell-penetration ability and effective utilization of multiple modes of antibacterial action.
Collapse
Affiliation(s)
- Marina Berditsch
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany
| | - Sergii Afonin
- KIT, Institute of Biological Interfaces (IBG-2), Karlsruhe, 76021, Germany
| | - Jennifer Reuster
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany
| | - Hannah Lux
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany
| | - Kristina Schkolin
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany
| | - Oleg Babii
- KIT, Institute of Biological Interfaces (IBG-2), Karlsruhe, 76021, Germany
| | - Dmytro S Radchenko
- Enamine Ltd., Kyiv, 02094, Ukraine.,Taras Shevchenko National University of Kyiv, Kyiv, 01601, Ukraine
| | - Issah Abdullah
- University College London (UCL), UCL School of Pharmacy, London, WC1N 1AX, United Kingdom
| | - Nicola William
- University of Leeds, School of Chemistry, Leeds, LS9 2JT, United Kingdom
| | - Volker Middel
- KIT, Institute of Toxicology and Genetics (ITG), Eggenstein-Leopoldshafen, 76344, Germany
| | - Uwe Strähle
- KIT, Institute of Toxicology and Genetics (ITG), Eggenstein-Leopoldshafen, 76344, Germany
| | - Andrew Nelson
- University of Leeds, School of Chemistry, Leeds, LS9 2JT, United Kingdom
| | - Klara Valko
- University College London (UCL), UCL School of Pharmacy, London, WC1N 1AX, United Kingdom
| | - Anne S Ulrich
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany. .,KIT, Institute of Biological Interfaces (IBG-2), Karlsruhe, 76021, Germany.
| |
Collapse
|
136
|
Ruden S, Rieder A, Chis Ster I, Schwartz T, Mikut R, Hilpert K. Synergy Pattern of Short Cationic Antimicrobial Peptides Against Multidrug-Resistant Pseudomonas aeruginosa. Front Microbiol 2019; 10:2740. [PMID: 31849888 PMCID: PMC6901909 DOI: 10.3389/fmicb.2019.02740] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 11/11/2019] [Indexed: 12/18/2022] Open
Abstract
With the rise of various multidrug-resistant (MDR) pathogenic bacteria, worldwide health care is under pressure to respond. Conventional antibiotics are failing and the development of novel classes and alternative strategies is a major priority. Antimicrobial peptides (AMPs) cannot only kill MDR bacteria, but also can be used synergistically with conventional antibiotics. We selected 30 short AMPs from different origins and measured their synergy in combination with polymyxin B, piperacillin, ceftazidime, cefepime, meropenem, imipenem, tetracycline, erythromycin, kanamycin, tobramycin, amikacin, gentamycin, and ciprofloxacin. In total, 403 unique combinations were tested against an MDR Pseudomonas aeruginosa isolate (PA910). As a measure of the synergistic effects, fractional inhibitory concentrations (FICs) were determined using microdilution assays with FICs ranges between 0.25 and 2. A high number of combinations between peptides and polymyxin B, erythromycin, and tetracycline were found to be synergistic. Novel variants of indolicidin also showed a high frequency in synergist interaction. Single amino acid substitutions within the peptides can have a very strong effect on the ability to synergize, making it possible to optimize future drugs toward synergistic interaction.
Collapse
Affiliation(s)
- Serge Ruden
- Institute of Biological Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Annika Rieder
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Irina Chis Ster
- Institute of Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Thomas Schwartz
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Kai Hilpert
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Infection and Immunity, St George's, University of London, London, United Kingdom.,Institute of Microstructure Technology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
137
|
Simpson DH, Hapeshi A, Rogers NJ, Brabec V, Clarkson GJ, Fox DJ, Hrabina O, Kay GL, King AK, Malina J, Millard AD, Moat J, Roper DI, Song H, Waterfield NR, Scott P. Metallohelices that kill Gram-negative pathogens using intracellular antimicrobial peptide pathways. Chem Sci 2019; 10:9708-9720. [PMID: 32015803 PMCID: PMC6977464 DOI: 10.1039/c9sc03532j] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
A range of new water-compatible optically pure metallohelices - made by self-assembly of simple non-peptidic organic components around Fe ions - exhibit similar architecture to some natural cationic antimicrobial peptides (CAMPs) and are found to have high, structure-dependent activity against bacteria, including clinically problematic Gram-negative pathogens. A key compound is shown to freely enter rapidly dividing E. coli cells without significant membrane disruption, and localise in distinct foci near the poles. Several related observations of CAMP-like mechanisms are made via biophysical measurements, whole genome sequencing of tolerance mutants and transcriptomic analysis. These include: high selectivity for binding of G-quadruplex DNA over double stranded DNA; inhibition of both DNA gyrase and topoisomerase I in vitro; curing of a plasmid that contributes to the very high virulence of the E. coli strain used; activation of various two-component sensor/regulator and acid response pathways; and subsequent attempts by the cell to lower the net negative charge of the surface. This impact of the compound on multiple structures and pathways corresponds with our inability to isolate fully resistant mutant strains, and supports the idea that CAMP-inspired chemical scaffolds are a realistic approach for antimicrobial drug discovery, without the practical barriers to development that are associated with natural CAMPS.
Collapse
Affiliation(s)
- Daniel H Simpson
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - Alexia Hapeshi
- Warwick Medical School , University of Warwick , Coventry , CV4 7AL , UK
| | - Nicola J Rogers
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - Viktor Brabec
- The Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Guy J Clarkson
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - David J Fox
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - Ondrej Hrabina
- The Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
- Department of Biophysics , Palacky University , Slechtitelu 27 , 783 71 Olomouc , Czech Republic
| | - Gemma L Kay
- Warwick Medical School , University of Warwick , Coventry , CV4 7AL , UK
| | - Andrew K King
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - Jaroslav Malina
- The Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Andrew D Millard
- Warwick Medical School , University of Warwick , Coventry , CV4 7AL , UK
| | - John Moat
- School of Life Sciences , University of Warwick , Gibbet Hill Campus , Coventry , CV4 7AL , UK
| | - David I Roper
- School of Life Sciences , University of Warwick , Gibbet Hill Campus , Coventry , CV4 7AL , UK
| | - Hualong Song
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | | | - Peter Scott
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| |
Collapse
|
138
|
Li Y, Liu T, Liu Y, Tan Z, Ju Y, Yang Y, Dong W. Antimicrobial activity, membrane interaction and stability of the D-amino acid substituted analogs of antimicrobial peptide W3R6. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 200:111645. [PMID: 31671371 DOI: 10.1016/j.jphotobiol.2019.111645] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 09/30/2019] [Accepted: 10/06/2019] [Indexed: 11/15/2022]
Abstract
Antimicrobial peptide W3R6 was derived from chensinin-1b and showed potential as a novel antibiotics. However, W3R6 was susceptible to protease cleavage, which limited its therapeutic application. To improve the proteolytic resistance of W3R6, D-amino acids were incorporated into its sequence by specific amino acid substitution or whole sequence substitution according to the specificity of the cleavage site. In this study, partially substituted analog D-Arg-W3R6 and completely substituted D-enantiomer D-W3R6 were synthesized. The resistance of D-Arg-W3R6 and D-W3R6 to cleavage by the tested protease increased, particularly of D-W3R6. The antimicrobial activity of D-Arg-W3R6 was almost the same as that of the parent peptide W3R6, but the antimicrobial activity of D-W3R6 was slightly decreased. The hemolytic activity of both D-Arg-W3R6 and D-W3R6 was negligible. The CD spectrum of D-W3R6 exhibited symmetry with that of W3R6 in a membrane-mimetic environment. The membrane interaction between the D-amino acid substituted analogs and a real/mimic bacterial cell membrane was examined. The outer membrane depolarization, inner membrane permeability and dye leakage in three types of liposomes treated with D-Arg-W3R6 and D-W3R6 were not obviously different from W3R6, which could be due to the similar physical and chemical properties. In addition, these three peptides showed the binding ability with LPS micelles detected by ITC, and their ability to disrupt the LPS micelles was examined by DLS experiment and even neutralize the surface negative charge of E. coli cells. These results suggest that D-Arg-W3R6 is a promising antibiotic molecule.
Collapse
Affiliation(s)
- Yi Li
- Department of Respiratory Medicine, Beijing Hospital, Beijing 100730, China
| | - Tong Liu
- School of Life Science, Liaoning Normal University, Dalian 116081, China
| | - Yan Liu
- Ling-Nan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhen Tan
- Department of Respiratory Medicine, Beijing Hospital, Beijing 100730, China
| | - Yang Ju
- Department of Respiratory Medicine, Beijing Hospital, Beijing 100730, China
| | - Yi Yang
- Ling-Nan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Weibing Dong
- School of Life Science, Liaoning Normal University, Dalian 116081, China; Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian 116081, China.
| |
Collapse
|
139
|
Mankoci S, Ewing J, Dalai P, Sahai N, Barton HA, Joy A. Bacterial Membrane Selective Antimicrobial Peptide-Mimetic Polyurethanes: Structure–Property Correlations and Mechanisms of Action. Biomacromolecules 2019; 20:4096-4106. [DOI: 10.1021/acs.biomac.9b00939] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
140
|
|
141
|
Ríos Colombo NS, Chalón MC, Dupuy FG, Gonzalez CF, Bellomio A. The case for class II bacteriocins: A biophysical approach using "suicide probes" in receptor-free hosts to study their mechanism of action. Biochimie 2019; 165:183-195. [PMID: 31381962 DOI: 10.1016/j.biochi.2019.07.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/26/2019] [Indexed: 11/29/2022]
Abstract
Class II bacteriocins are unmodified membrane-active peptides that act over a narrow spectrum of target bacteria. They bind a specific receptor protein on the membrane to form a pore, leading to membrane permeabilization and cell death. However, little is known about the molecular events triggering the pore formation after the bacteriocin recognizes the receptor. It is not clear yet if the pore is the same receptor forced into an open conformation or if the pore results from the bacteriocin insertion and oligomeric assembly in the lipid bilayer. In order to reveal which model is more suitable to explain the toxicity mechanism, in this work we use chimeric peptides, resulting from the fusion of the bitopic membrane protein EtpM with different class II bacteriocins: enterocin CRL35, pediocin PA-1 and microcin V. E. coli strains lacking the specific receptors for these bacteriocins were chosen as expression hosts. As these constructs display a lethal effect when they are heterologously expressed, they are called "suicide probes". The results suggest that, indeed, the specific receptor would act as a docking molecule more than as a structural piece of the pore, as long as the bacteriocin is somehow anchored to the membrane. These set of chimeric peptides also represent an in vivo system that allows to study the interaction of the bacteriocins with real bacterial membranes, instead of model membranes. Hence, the effects of these suicide probes in membrane fluidity and transmembrane potential were also assessed, using fluorescence spectroscopy. The data show that the different suicide probes are able to increase phospholipid order and depolarize the membranes of receptor-free bacterial cells.
Collapse
Affiliation(s)
- N S Ríos Colombo
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461, San Miguel de Tucumán, T4000ILI, Argentina
| | - M C Chalón
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461, San Miguel de Tucumán, T4000ILI, Argentina
| | - F G Dupuy
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461, San Miguel de Tucumán, T4000ILI, Argentina
| | - C F Gonzalez
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, 2033 Mowry road, PO Box 103610, Gainesville, FL, 32610-3610, USA
| | - A Bellomio
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT and Instituto de Química Biológica "Dr. Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT, Chacabuco 461, San Miguel de Tucumán, T4000ILI, Argentina.
| |
Collapse
|
142
|
Balandin SV, Sheremeteva EV, Ovchinnikova TV. Pediocin-Like Antimicrobial Peptides of Bacteria. BIOCHEMISTRY (MOSCOW) 2019; 84:464-478. [PMID: 31234762 DOI: 10.1134/s000629791905002x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Bacteriocins are bacterial antimicrobial peptides that, unlike classical peptide antibiotics, are products of ribosomal synthesis and usually have a narrow spectrum of antibacterial activity against species closely related to the producers. Pediocin-like bacteriocins (PLBs) belong to the class IIa of the bacteriocins of Gram-positive bacteria. PLBs possess high activity against pathogenic bacteria from Listeria and Enterococcus genera. Molecular target for PLBs is a membrane protein complex - bacterial mannose-phosphotransferase. PLBs can be synthesized by components of symbiotic microflora and participate in the maintenance of homeostasis in various compartments of the digestive tract and on the surface of epithelial tissues contacting the external environment. PLBs could give a rise to a new group of antibiotics of narrow spectrum of activity.
Collapse
Affiliation(s)
- S V Balandin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - E V Sheremeteva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - T V Ovchinnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
| |
Collapse
|
143
|
Kamiura R, Matsuda F, Ichihashi N. Survival of membrane-damaged Escherichia coli in a cytosol-mimicking solution. J Biosci Bioeng 2019; 128:558-563. [PMID: 31182278 DOI: 10.1016/j.jbiosc.2019.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 11/19/2022]
Abstract
Selective permeability of cell membrane is critically important for cell survival. The damage caused to cell membrane by pore-forming antimicrobial peptides may result in the loss of selective permeability and leakage of intracellular molecules, eventually leading to cell death. Here, we examined whether the membrane-damaged Escherichia coli cells survive in a cytosol-mimicking solution (CMS), which compensates for the lethal leakage of intracellular molecules. We prepared a CMS comprising 34 low molecular weight compounds from the cytosol and found that the cells were able to grow in CMS even in the presence of a pore-forming peptide, melittin. We confirmed that the melittin-treated cells lost selective membrane permeability by staining with membrane-impermeable dyes, propidium iodide and SYTOX green. Some stained cells maintained the colony formation ability in CMS. These results provide an evidence that E. coli cells can at least partially survive in the CMS even after the temporary impairment of membrane selective permeability. This study demonstrates a technique that allows temporal loss of the selective permeability of the cell membrane while maintaining the viability of cells that may be useful for the introduction of membrane-impermeable molecules into E. coli cells.
Collapse
Affiliation(s)
- Rikuto Kamiura
- Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Fumio Matsuda
- Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Norikazu Ichihashi
- Graduate School of Information Science and Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan; Graduate School of Frontier Bioscience, Osaka University, 1-5 Yamadaoka, Suita, Osaka 565-0871, Japan; Graduate School of Arts and Science, Komaba Institute for Science, Universal Biology Institute, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan.
| |
Collapse
|
144
|
Kutepov IV, Lyashev YD, Artyushkova EB, Solin AV, Serikov VS, Lyashev AY, Chahine AR. Correction of Acute Parodontitis with Indolicidin Analogues. Bull Exp Biol Med 2019; 167:47-49. [PMID: 31177448 DOI: 10.1007/s10517-019-04457-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Indexed: 11/28/2022]
Abstract
We studied the influence of synthetic indolicidin analogues on the development of acute periodontitis. The corrective effect was found in indolicidin analogues Nos. 7 and 8; it manifested in a decrease in the edema of gingival epithelium and lamina propria, a decrease in the relative area of inflammatory infiltrates, and a significant increase in the relative area of normal connective tissue. These changes were revealed as soon as on day 14 and were most pronounced in 21 days after the removal of the ligature. Indolicidin analogues Nos. 7 and 8 demonstrated similar effectiveness on the model of acute periodontitis.
Collapse
Affiliation(s)
- I V Kutepov
- Department of pathophysiology, Kursk State Medical University, Kursk, Russia.
| | - Yu D Lyashev
- Department of pathophysiology, Kursk State Medical University, Kursk, Russia
| | - E B Artyushkova
- Department of pathophysiology, Kursk State Medical University, Kursk, Russia
| | - A V Solin
- Department of pathophysiology, Kursk State Medical University, Kursk, Russia
| | - V S Serikov
- Department of pathophysiology, Kursk State Medical University, Kursk, Russia
| | - A Yu Lyashev
- Department of pathophysiology, Kursk State Medical University, Kursk, Russia
| | - A R Chahine
- ABC Medical Imaging, ABC Laboratories, Beirut, Lebanon
| |
Collapse
|
145
|
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
|
146
|
Tridecaptin M, a New Variant Discovered in Mud Bacterium, Shows Activity against Colistin- and Extremely Drug-Resistant Enterobacteriaceae. Antimicrob Agents Chemother 2019; 63:AAC.00338-19. [PMID: 30936101 PMCID: PMC6535564 DOI: 10.1128/aac.00338-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/12/2019] [Indexed: 12/05/2022] Open
Abstract
The World Health Organization has categorized the Gram-negative superbugs, which are inherently impervious to many antibiotics, as critical priority pathogens due to the lack of effective treatments. The breach in our last-resort antibiotic (i.e., colistin) by extensively drug-resistant and pan-drug-resistant Enterobacteriaceae strains demands the immediate development of new therapies. The World Health Organization has categorized the Gram-negative superbugs, which are inherently impervious to many antibiotics, as critical priority pathogens due to the lack of effective treatments. The breach in our last-resort antibiotic (i.e., colistin) by extensively drug-resistant and pan-drug-resistant Enterobacteriaceae strains demands the immediate development of new therapies. In the present study, we report the discovery of tridecaptin M, a new addition to the family, and its potential against colistin-resistant Enterobacteriaceae in vitro and in vivo. Also, we performed mode-of-action studies using various fluorescent probes and studied the hemolytic activity and mammalian cytotoxicity in two cell lines. Tridecaptin M displayed strong antibacterial activity (MICs of 2 to 8 μg ml−1) against clinical strains of Klebsiella pneumoniae (which were resistant to colistin, carbapenems, third- and fourth-generation cephalosporins, fluoroquinolones, fosfomycin, and other antibiotics) and mcr-1-positive Escherichia coli strains. Unlike polymyxins, tridecaptin M did not permeabilize the outer membrane or cytoplasmic membrane. It blocked ATP synthesis in bacteria by dissipating the proton motive force. The compound exhibited negligible acquired resistance, low in vitro cytotoxicity and hemolytic activity, and no significant acute toxicity in mice. It also showed promising efficacy in a thigh infection model of colistin-resistant K. pneumoniae. Altogether, these results demonstrate the future prospects of this class of antibiotics to address the unmet medical need to circumvent colistin resistance in extensively drug-resistant Enterobacteriaceae infections. The work also emphasizes the importance of natural products in our shrunken drug discovery pipeline.
Collapse
|
147
|
Postantibiotic and Sub-MIC Effects of Exebacase (Lysin CF-301) Enhance Antimicrobial Activity against Staphylococcus aureus. Antimicrob Agents Chemother 2019; 63:AAC.02616-18. [PMID: 30936103 DOI: 10.1128/aac.02616-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/24/2019] [Indexed: 12/13/2022] Open
Abstract
CF-301 (exebacase) is a recombinantly produced bacteriophage-derived lysin (cell wall hydrolase) and is the first agent of this class to enter clinical development in the United States for treating bacteremia including endocarditis due to Staphylococcus aureus Whereas rapid bactericidal activity is the hallmark in vitro and in vivo response to CF-301 at exposures higher than the MIC, prolonged antimicrobial activity, mediated by cell wall damage, is predicted at concentrations less than the MIC. In the current study, a series of in vitro pharmacodynamic parameters, including the postantibiotic effect (PAE), postantibiotic sub-MIC effect (PA-SME), and sub-MIC effect (SME), were studied to determine how short-duration and sub-MIC CF-301 exposures affect the growth of surviving staphylococci and extend its antimicrobial activity. Mean PAE, PA-SME, and SME values up to 4.8, 9.3, and 9.8 h, respectively, were observed against 14 staphylococcal strains tested in human serum; growth delays were extended by 6 h in the presence of daptomycin. Exposures to CF-301 at sub-MIC levels as low as 0.001× to 0.01× MIC (∼1 to 10 ng/ml) resulted in aberrant cell wall ultrastructure, increased membrane permeability, dissipation of membrane potential, and inhibition of virulence phenotypes, including agglutination and biofilm formation. A mouse thigh infection model designed to study the PAE was used to confirm our findings and demonstrate in vivo growth delays of ≥19.3 h. Our findings suggest that at CF-301 concentrations less than the MIC during therapeutic use, sustained reductions in bacterial fitness and virulence may substantially enhance efficacy.
Collapse
|
148
|
Hossain F, Moghal MMR, Islam MZ, Moniruzzaman M, Yamazaki M. Membrane potential is vital for rapid permeabilization of plasma membranes and lipid bilayers by the antimicrobial peptide lactoferricin B. J Biol Chem 2019; 294:10449-10462. [PMID: 31118274 DOI: 10.1074/jbc.ra119.007762] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/17/2019] [Indexed: 11/06/2022] Open
Abstract
Lactoferricin B (LfcinB) is a cationic antimicrobial peptide, and its capacity to damage the bacterial plasma membrane is suggested to be a main factor in LfcinB's antimicrobial activity. However, the specific processes and mechanisms in LfcinB-induced membrane damage are unclear. In this report, using confocal laser-scanning microscopy, we examined the interaction of LfcinB with single Escherichia coli cells and spheroplasts containing the water-soluble fluorescent probe calcein in the cytoplasm. LfcinB induced rapid calcein leakage from single E. coli cells and from single spheroplasts, indicating that LfcinB interacts directly with the plasma membrane and induces its rapid permeabilization. The proton ionophore carbonyl cyanide m-chlorophenylhydrazone suppressed this leakage. Next, we used the single giant unilamellar vesicle (GUV) method to examine LfcinB's interaction with GUVs comprising polar lipid extracts of E. coli containing a water-soluble fluorescent probe, Alexa Fluor 647 hydrazide (AF647). We observed that LfcinB stochastically induces local rupture in single GUVs, causing rapid AF647 leakage; however, higher LfcinB concentrations were required for AF647 leakage from GUVs than from E. coli cells and spheroplasts. To identify the reason for this difference, we examined the effect of membrane potential on LfcinB-induced pore formation, finding that the rate of LfcinB-induced local rupture in GUVs increases greatly with increasing negative membrane potential. These results indicate that membrane potential plays an important role in LfcinB-induced local rupture of lipid bilayers and rapid permeabilization of E. coli plasma membranes. On the basis of these results, we discuss the mode of action of LfcinB's antimicrobial activity.
Collapse
Affiliation(s)
- Farzana Hossain
- From the Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529
| | - Md Mizanur Rahman Moghal
- From the Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529
| | - Md Zahidul Islam
- From the Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529
| | - Md Moniruzzaman
- From the Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529
| | - Masahito Yamazaki
- From the Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529, .,the Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka 422-8529, and.,the Department of Physics, Graduate School of Science, Shizuoka University, Shizuoka 422-8529, Japan
| |
Collapse
|
149
|
Fernandez J, Acosta G, Pulido D, Malý M, Copa-Patiño JL, Soliveri J, Royo M, Gómez R, Albericio F, Ortega P, de la Mata FJ. Carbosilane Dendron-Peptide Nanoconjugates as Antimicrobial Agents. Mol Pharm 2019; 16:2661-2674. [PMID: 31009225 DOI: 10.1021/acs.molpharmaceut.9b00222] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Over the last decades, multidrug-resistant bacteria have emerged and spread, increasing the number of bacteria, against which commonly used antibiotics are no longer effective. It has become a serious public health problem whose solution requires medical research in order to explore novel effective antimicrobial molecules. On the one hand, antimicrobial peptides (AMPs) are regarded as good alternatives because of their generally broad-spectrum activities, but sometimes they can be easily degraded by the organism or be toxic to animal cells. On the other hand, cationic carbosilane dendrons, whose focal point can be functionalized in many different ways, have also shown good antimicrobial activity. In this work, we synthetized first- and second-generation cationic carbosilane dendrons with a maleimide molecule on their focal point, enabling their functionalization with three different AMPs. After different microbiology studies, we found an additive effect between first-generation dendron and AMP3 whose study reveals three interesting effects: (i) bacteria aggregation due to AMP3, which could facilitate bacteria detection or even contribute to antibacterial activity by preventing host cell attack, (ii) bacteria disaggregation capability of second-generation cationic dendrons, and (iii) a higher AMP3 aggregation ability when dendrons were added previously to peptide treatment. These compounds and their different effects observed over bacteria constitute an interesting system for further mechanism studies.
Collapse
Affiliation(s)
- Jael Fernandez
- Instituto de Investigación Química "Andrés M. del Río" (IQAR) , UAH , 28801 Alcalá de Henares , Spain.,Networking Research Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN) , 28029 Madrid , Spain.,Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS , 28034 Madrid , Spain
| | - Gerardo Acosta
- Networking Research Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN) , 28029 Madrid , Spain.,Deparment of Organic and Inorganic Chemistry , University of Barcelona , 08028 Barcelona , Spain.,Institute for Advanced Chemistry of Catalonia-CSIC , 08034 Barcelona , Spain
| | - Daniel Pulido
- Networking Research Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN) , 28029 Madrid , Spain.,Institute for Advanced Chemistry of Catalonia-CSIC , 08034 Barcelona , Spain
| | - Marek Malý
- Faculty of Science , J. E. Purkinje University , České mládeže 8 , 400 96 Ústí nad Labem , Czech Republic
| | | | | | - Miriam Royo
- Networking Research Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN) , 28029 Madrid , Spain.,Institute for Advanced Chemistry of Catalonia-CSIC , 08034 Barcelona , Spain
| | - Rafael Gómez
- Instituto de Investigación Química "Andrés M. del Río" (IQAR) , UAH , 28801 Alcalá de Henares , Spain.,Networking Research Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN) , 28029 Madrid , Spain.,Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS , 28034 Madrid , Spain
| | - Fernando Albericio
- Networking Research Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN) , 28029 Madrid , Spain.,Deparment of Organic and Inorganic Chemistry , University of Barcelona , 08028 Barcelona , Spain.,Institute for Advanced Chemistry of Catalonia-CSIC , 08034 Barcelona , Spain.,School of Chemistry and Physics , University of KwaZulu-Natal , 4001 Durban , South Africa
| | - Paula Ortega
- Instituto de Investigación Química "Andrés M. del Río" (IQAR) , UAH , 28801 Alcalá de Henares , Spain.,Networking Research Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN) , 28029 Madrid , Spain.,Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS , 28034 Madrid , Spain
| | - F Javier de la Mata
- Instituto de Investigación Química "Andrés M. del Río" (IQAR) , UAH , 28801 Alcalá de Henares , Spain.,Networking Research Center on Bioengineering , Biomaterials and Nanomedicine (CIBER-BBN) , 28029 Madrid , Spain.,Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS , 28034 Madrid , Spain
| |
Collapse
|
150
|
Kuppusamy R, Willcox M, Black DS, Kumar N. Short Cationic Peptidomimetic Antimicrobials. Antibiotics (Basel) 2019; 8:antibiotics8020044. [PMID: 31003540 PMCID: PMC6628222 DOI: 10.3390/antibiotics8020044] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/28/2022] Open
Abstract
The rapid growth of antimicrobial resistance against several frontline antibiotics has encouraged scientists worldwide to develop new alternatives with unique mechanisms of action. Antimicrobial peptides (AMPs) have attracted considerable interest due to their rapid killing and broad-spectrum activity. Peptidomimetics overcome some of the obstacles of AMPs such as high cost of synthesis, short half-life in vivo due to their susceptibility to proteolytic degradation, and issues with toxicity. This review will examine the development of short cationic peptidomimetics as antimicrobials.
Collapse
Affiliation(s)
- Rajesh Kuppusamy
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Mark Willcox
- School of Optometry and Vision Science, University of New South Wales, Sydney, NSW 2052, Australia.
| | - David StC Black
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Naresh Kumar
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|