1
|
Bermúdez-Puga S, Dias M, Lima Reis I, Freire de Oliveira T, Yokomizo de Almeida SR, Mendes MA, Moore SJ, Almeida JR, Proaño-Bolaños C, Pinheiro de Souza Oliveira R. Microscopic and metabolomics analysis of the anti-Listeria activity of natural and engineered cruzioseptins. Biochimie 2024; 225:168-175. [PMID: 38823620 DOI: 10.1016/j.biochi.2024.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/04/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Listeria monocytogenes is a human opportunistic foodborne pathogen that produces life-threatening infections with a high mortality rate. The control of Listeria in the food production environment and effective clinical management of human listeriosis are challenging due to the emergence of antibiotic resistance. Hence we evaluate the in vitro anti-Listeria activity of two synthetic cruzioseptins reproducing their natural sequences CZS-9, and CZS-12, and one engineered sequence based on CZS-1, named [K4K15]CZS-1. The assessment of the in vitro potential of cruzioseptins, highlighted the promising antibacterial effect of [K4K15]CZS-1 in very low concentrations (0.91 μM) and its thermal stability at high-temperature conditions, is compatible with the food industry. Microscopic and metabolomic analyses suggest cruzioseptin induces anti-Listeria bioactivity through membrane disruption and changes in the intracellular metabolome. We also report that [K4K15]CZS-1 is not resistant to peptidases/proteases emphasizing a key advantage for their use as a food preservative. However, there is a need for further structural and functional optimisations for the potential clinical application as an antibiotic. In conclusion, [K4K15]CZS-1 stand out as membrane-active peptides with the ability to induce shifts in the bacteria metabolome and inspire the development of strategies for the prevention of L. monocytogenes emergence and dissemination.
Collapse
Affiliation(s)
- Sebastián Bermúdez-Puga
- Microbial Biomolecules Laboratory, Faculty of Pharmaceutical Sciences, University of São Paulo, Rua do Lago 250, São Paulo, 05508-000, SP, Brazil
| | - Meriellen Dias
- Dempster MS Lab, Chemical Engineering Department of Polytechnic School of University of São Paulo, Rua do Lago 250, São Paulo, 05508-080, SP, Brazil
| | - Iara Lima Reis
- Microbial Biomolecules Laboratory, Faculty of Pharmaceutical Sciences, University of São Paulo, Rua do Lago 250, São Paulo, 05508-000, SP, Brazil
| | - Taciana Freire de Oliveira
- Microbial Biomolecules Laboratory, Faculty of Pharmaceutical Sciences, University of São Paulo, Rua do Lago 250, São Paulo, 05508-000, SP, Brazil
| | | | - Maria Anita Mendes
- Dempster MS Lab, Chemical Engineering Department of Polytechnic School of University of São Paulo, Rua do Lago 250, São Paulo, 05508-080, SP, Brazil
| | - Simon J Moore
- School of Biological and Behavioural Sciences, Queen Mary University of London, UK
| | - José R Almeida
- Biomolecules Discovery Group, Universidad Regional Amazónica Ikiam, Km 7 Via Muyuna, Tena, Napo, Ecuador; School of Pharmacy, University of Reading, Reading, RG6 6UB, UK
| | - Carolina Proaño-Bolaños
- Biomolecules Discovery Group, Universidad Regional Amazónica Ikiam, Km 7 Via Muyuna, Tena, Napo, Ecuador
| | - Ricardo Pinheiro de Souza Oliveira
- Microbial Biomolecules Laboratory, Faculty of Pharmaceutical Sciences, University of São Paulo, Rua do Lago 250, São Paulo, 05508-000, SP, Brazil.
| |
Collapse
|
2
|
Chen M, Xiao Z, Yan C, Tang X, Fang M, Wang Z, Zhang D. Centrosomal protein of 192 kDa (Cep192) fragment possesses bactericidal and parasiticidal activities in Larimichthys crocea. Int J Biol Macromol 2024; 254:127744. [PMID: 38287570 DOI: 10.1016/j.ijbiomac.2023.127744] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 01/31/2024]
Abstract
A novel AMP Lc1773, derived from centrosomal protein of 192 kDa (Cep192), was isolated from Larimichthys crocea using a Bacillus subtilis system. After cDNA libraries construction, repeating selection of B. subtilis system, extraction of extracellular protein, and expression of recombinant protein, we found that B. subtilis 1773, extracellular protein, and rLc1773 had a strong potential to kill Vibrio. parahaemolyticus and V. vulnificus. Further analysis of the antibacterial mechanism revealed that rLc1773 not only disrupted the integrity of bacterial membrane (as confirmed by SEM, TEM, and confocal microscopy observation, and flow cytometry assays), resulting in bacterial cell membrane pore conformation, bacterial rupture, and leakage of cellular contents, but also targeted to block protein synthesis rather than damage nucleic acids (as confirmed by SDS-PAGE, enzyme expression, and gel retardation assays). In addition, rLc1773 had the ability to kill parasite Scuticociliatida in a high rate and low concentration. Critically, the antibacterial activity of rLc1773 had good thermal stability and UV radiation tolerance, but it was affected by pH 9-11 and diverse enzyme to some extent. Lc1773 had neither hemolysis on fish, shrimp, and rabbit erythrocytes,nor significant cytotoxicity. To our knowledge, Cep192 fragment was first demonstrated to possess bactericidal and parasiticidal activities.
Collapse
Affiliation(s)
- Meiling Chen
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Zhiqun Xiao
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Chunmei Yan
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Xin Tang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Ming Fang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China
| | - Zhiyong Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Dongling Zhang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Jimei University, Xiamen, China.
| |
Collapse
|
3
|
Gambato S, Bellotto O, Mardirossian M, Di Stasi A, Gennaro R, Pacor S, Caporale A, Berti F, Scocchi M, Tossi A. Designing New Hybrid Antibiotics: Proline-Rich Antimicrobial Peptides Conjugated to the Aminoglycoside Tobramycin. Bioconjug Chem 2023. [PMID: 37379329 PMCID: PMC10360068 DOI: 10.1021/acs.bioconjchem.2c00467] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Resistance to aminoglycoside antibiotics is a serious problem, typically arising from inactivating enzymes, reduced uptake, or increased efflux in the important pathogens for which they are used as treatment. Conjugating aminoglycosides to proline-rich antimicrobial peptides (PrAMPs), which also target ribosomes and have a distinct bacterial uptake mechanism, might mutually benefit their individual activities. To this aim we have developed a strategy for noninvasively modifying tobramycin to link it to a Cys residue and through this covalently link it to a Cys-modified PrAMP by formation of a disulfide bond. Reduction of this bridge in the bacterial cytosol should release the individual antimicrobial moieties. We found that the conjugation of tobramycin to the well-characterized N-terminal PrAMP fragment Bac7(1-35) resulted in a potent antimicrobial capable of inactivating not only tobramycin-resistant bacterial strains but also those less susceptible to the PrAMP. To a certain extent, this activity also extends to the shorter and otherwise poorly active fragment Bac7(1-15). Although the mechanism that allows the conjugate to act when its individual components do not is as yet unclear, results are very promising and suggest this may be a way of resensitizing pathogens that have developed resistance to the antibiotic.
Collapse
Affiliation(s)
- Stefano Gambato
- Department of Life Sciences, University of Trieste, Via L. Giorgeri, 5, 34127 Trieste, Italy
| | - Ottavia Bellotto
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgeri, 1, 34127 Trieste, Italy
| | - Mario Mardirossian
- Department of Life Sciences, University of Trieste, Via L. Giorgeri, 5, 34127 Trieste, Italy
| | - Adriana Di Stasi
- Department of Life Sciences, University of Trieste, Via L. Giorgeri, 5, 34127 Trieste, Italy
| | - Renato Gennaro
- Department of Life Sciences, University of Trieste, Via L. Giorgeri, 5, 34127 Trieste, Italy
| | - Sabrina Pacor
- Department of Life Sciences, University of Trieste, Via L. Giorgeri, 5, 34127 Trieste, Italy
| | - Andrea Caporale
- CNR, Institute of Crystallography, SS 14 Km 163.5 c/o Area Science Park, Basovizza, 34149 Trieste, Italy
- CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", 80134 Napoli, Italy
| | - Federico Berti
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgeri, 1, 34127 Trieste, Italy
| | - Marco Scocchi
- Department of Life Sciences, University of Trieste, Via L. Giorgeri, 5, 34127 Trieste, Italy
| | - Alessandro Tossi
- Department of Life Sciences, University of Trieste, Via L. Giorgeri, 5, 34127 Trieste, Italy
| |
Collapse
|
4
|
Langer MK, Rahman A, Dey H, Anderssen T, Blencke HM, Haug T, Stensvåg K, Strøm MB, Bayer A. Investigation of tetrasubstituted heterocycles reveals hydantoins as a promising scaffold for development of novel antimicrobials with membranolytic properties. Eur J Med Chem 2023; 249:115147. [PMID: 36739750 DOI: 10.1016/j.ejmech.2023.115147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/19/2022] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
Mimics of antimicrobial peptides (AMPs) have been proposed as a promising class of antimicrobial agents. We report the analysis of five tetrasubstituted, cationic, amphipathic heterocycles as potential AMP mimics. The analysis showed that the heterocyclic scaffold had a strong influence on the haemolytic activity of the compounds, and the hydantoin scaffold was identified as a promising template for drug lead development. Subsequently, a total of 20 hydantoin derivatives were studied for their antimicrobial potency and haemolytic activity. We found 19 of these derivatives to have very low haemolytic toxicity and identified three lead structures, 2dA, 6cG, and 6dG with very promising broad-spectrum antimicrobial activity. Lead structure 6dG displayed minimum inhibitory concentration (MIC) values as low as 1 μg/mL against Gram-positive bacteria and 4-16 μg/mL against Gram-negative bacteria. Initial mode of action (MoA) studies performed on the amine derivative 6cG, utilizing a luciferase-based biosensor assay, suggested a strong membrane disrupting effect on the outer and inner membrane of Escherichia coli. Our findings show that the physical properties and structural arrangement induced by the heterocyclic scaffolds are important factors in the design of AMP mimics.
Collapse
Affiliation(s)
- Manuel K Langer
- Department of Chemistry, UiT - The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Ataur Rahman
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT - The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Hymonti Dey
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT - The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Trude Anderssen
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Hans-Matti Blencke
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT - The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Tor Haug
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT - The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Klara Stensvåg
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT - The Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Morten B Strøm
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, NO-9037, Tromsø, Norway.
| | - Annette Bayer
- Department of Chemistry, UiT - The Arctic University of Norway, NO-9037, Tromsø, Norway.
| |
Collapse
|
5
|
Novel Arginine- and Proline-Rich Candidacidal Peptides Obtained through a Bioinformatic Approach. Antibiotics (Basel) 2023; 12:antibiotics12030472. [PMID: 36978339 PMCID: PMC10044544 DOI: 10.3390/antibiotics12030472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Antimicrobial resistance is a major public health concern worldwide. Albeit to a lesser extent than bacteria, fungi are also becoming increasingly resistant to antifungal drugs. Moreover, due to the small number of antifungal classes, therapy options are limited, complicating the clinical management of mycoses. In this view, antimicrobial peptides (AMPs) are a potential alternative to conventional drugs. Among these, Proline-rich antimicrobial peptides (PrAMPs), almost exclusively of animal origins, are of particular interest due to their peculiar mode of action. In this study, a search for new arginine- and proline-rich peptides from plants has been carried out with a bioinformatic approach by sequence alignment and antimicrobial prediction tools. Two peptide candidates were tested against planktonic cells and biofilms of Candida albicans and Candida glabrata strains, including resistant isolates. These peptides showed similar potent activity, with half-maximal effective concentration values in the micromolar range. In addition, some structural and functional features, revealing peculiar mechanistic behaviors, were investigated.
Collapse
|
6
|
Rattanadilog Na Phuket T, Charoensapsri W, Amparyup P, Imjongjirak C. Antibacterial activity and immunomodulatory role of a proline-rich antimicrobial peptide SpPR-AMP1 against Vibrio campbellii infection in shrimp Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2023; 132:108479. [PMID: 36513322 DOI: 10.1016/j.fsi.2022.108479] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Antimicrobial peptides (AMPs) constitute one of the most promising sources of natural molecules used for the design of effective antimicrobial agents alternative to antibiotics. Previously, we have showed that a crab proline-rich AMP designated as SpPR-AMP1 is a potent AMP that exhibited antimicrobial activity against both Gram-positive and Gram-negative bacteria. Here, we demonstrated the importance of SpPR-AMP1 peptide in treating a virulent acute hepatopancreatic necrosis disease (AHPND) Vibrio campbellii VH-639 isolate and eliciting the innate immune response to counter the AHPND infection in shrimp Litopenaeus vannamei. SpPR-AMP1 exhibited a strong antimicrobial activity against V. campbellii VH-639 at MIC value of 0.195-0.39 μM. Scanning electron microscopy (SEM) revealed the membrane disruption potential of SpPR-AMP1 against the V. campbellii VH-639 cells. The in vivo effect of SpPR-AMP1 in shrimp L.vannamei was investigated and the results showed that SpPR-AMP1 was capable of modulating the innate immune response by stimulating the expression levels of AMP transcripts in shrimp hemocytes. Moreover, treatments with SpPR-AMP1 could promote the resistance of shrimp against V. campbellii VH-639 infection as demonstrated by a significant increase in shrimp survival rate and decrease in both the bacterial load and the expression levels of bacterial PirA and PirB toxin gene transcripts in the infected shrimp. These results suggest the potential of SpPR-AMP1 peptide with the combined antimicrobial and immunoenhancing capabilities as promising antimicrobial agent to treat V. campbellii VH-639 causing AHPND infection in shrimp aquaculture.
Collapse
Affiliation(s)
- Thitiya Rattanadilog Na Phuket
- Department of Food Technology, Faculty of Science, Chulalongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand
| | - Walaiporn Charoensapsri
- Marine Biotechnology Research Team, Integrative Aquaculture Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani, 12120, Thailand; Center of Excellence for Marine Biotechnology, Department of Marine Science, Faculty of Science, Chulalongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand
| | - Piti Amparyup
- Marine Biotechnology Research Team, Integrative Aquaculture Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani, 12120, Thailand; Center of Excellence for Marine Biotechnology, Department of Marine Science, Faculty of Science, Chulalongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand.
| | - Chanprapa Imjongjirak
- Department of Food Technology, Faculty of Science, Chulalongkorn University, 254 Phayathai Road, Bangkok, 10330, Thailand.
| |
Collapse
|
7
|
Shen S, Sun Y, Ren F, Blair JMA, Siasat P, Fan S, Hu J, He J. Characteristics of antimicrobial peptide OaBac5mini and its bactericidal mechanism against Escherichia coli. Front Vet Sci 2023; 10:1123054. [PMID: 36908510 PMCID: PMC9995905 DOI: 10.3389/fvets.2023.1123054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Antimicrobial peptides (AMPs) play an important role in defending against the attack of pathogenic microorganisms. Among them, the proline-rich antibacterial peptides (PrAMPs) have been attracting close attention due to their simple structure, strong antibacterial activity, and low cell toxicity. OaBac5mini is an active fragment of the sheep-derived OaBac5 belonging to the PrAMPs family. Methods In this study, the antibacterial activity of OaBac5mini was investigated by testing the MICs against different stains of E. coli and S. aureus as well as the time-kill curve. The bactericidal mechanism was explored by determining the effect of OaBac5mini on the cell membrane. The stability and biosafety were also evaluated. Results The susceptibility test demonstrated that OaBac5mini showed potent antibacterial activity against the multidrug-resistant (MDR) E. coli isolates. It is noticeable that the absence of inner membrane protein SbmA in E. coli ATCC 25922 caused the MIC of OaBac5mini to increase 4-fold, implying OaBac5mini can enter into the cytoplasm via SbmA and plays its antibacterial activity. Moreover, the antibacterial activity of OaBac5mini against E. coli ATCC 25922 was not remarkably affected by the serum salts except for CaCl2 at a physiological concentration, pH, temperature, repeated freeze-thawing and proteases (trypsin < 20 μg/mL, pepsin or proteinase K). Time-kill curve analysis showed OaBac5mini at the concentration of 200 μg/mL (8 × MICs) could effectively kill E. coli ATCC 25922 after co-incubation for 12 h. In addition, OaBac5mini was not hemolytic against rabbit red blood cells and also was not cytotoxic to porcine small intestinal epithelial cells (IPEC-J2). Bioinformatic analysis indicated that OaBac5mini is a linear peptide with 8 net positive charges. Furthermore, OaBac5mini significantly increased the outer membrane permeability and impaired the inner membrane integrity and ultrastructure of E. coli ATCC25922. Conclusion OaBac5mini is a stable and potent PrAMP that kills E. coli by two different modes of action - inhibiting intracellular target(s) and damaging cell membrane.
Collapse
Affiliation(s)
- Shanshan Shen
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China.,College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Yawei Sun
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Fei Ren
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Jessica M A Blair
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Pauline Siasat
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shuaiqi Fan
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Junping He
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| |
Collapse
|
8
|
Langer MK, Rahman A, Dey H, Anderssen T, Zilioli F, Haug T, Blencke HM, Stensvåg K, Strøm MB, Bayer A. A concise SAR-analysis of antimicrobial cationic amphipathic barbiturates for an improved activity-toxicity profile. Eur J Med Chem 2022; 241:114632. [DOI: 10.1016/j.ejmech.2022.114632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/15/2022] [Accepted: 07/24/2022] [Indexed: 11/03/2022]
|
9
|
Dewangan RP, Verma DP, Verma NK, Gupta A, Pant G, Mitra K, Habib S, Ghosh JK. Spermine-Conjugated Short Proline-Rich Lipopeptides as Broad-Spectrum Intracellular Targeting Antibacterial Agents. J Med Chem 2022; 65:5433-5448. [PMID: 35297625 DOI: 10.1021/acs.jmedchem.1c01809] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Toward the design of new proline-rich peptidomimetics, a short peptide segment, present in several proline-rich antimicrobial peptides (AMPs), was selected. Fatty acids of varying lengths and spermine were conjugated at the N- and C-terminals of the peptide, respectively. Spermine-conjugated lipopeptides, C10-PR-Spn and C12-PR-Spn, exhibited minimum inhibitory concentrations within 1.5-6.2 μM against the tested pathogens including resistant bacteria and insignificant hemolytic activity against human red blood cells up to 100 μM concentrations and demonstrated resistance against trypsin digestion. C10-PR-Spn and C12-PR-Spn showed synergistic antimicrobial activity against multidrug-resistant methicillin-resistant Staphylococcus aureus with several tested antibiotics. These lipopeptides did not permeabilize bacterial membrane-mimetic lipid vesicles or damage the Escherichia coli membrane like the nonmembrane-lytic AMP, buforin-II. The results suggested that C10-PR-Spn and C12-PR-Spn could interact with the 70S ribosome of E. coli and inhibit its protein synthesis. C10-PR-Spn and C12-PR-Spn demonstrated superior clearance of bacteria from the spleen, liver, and kidneys of mice, infected with S. aureus ATCC 25923 compared to levofloxacin.
Collapse
Affiliation(s)
- Rikeshwer Prasad Dewangan
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Devesh Pratap Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Neeraj Kumar Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Ankit Gupta
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Garima Pant
- Electron Microscopy Unit, SAIF Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Kalyan Mitra
- Electron Microscopy Unit, SAIF Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Saman Habib
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Jimut Kanti Ghosh
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| |
Collapse
|
10
|
Landon C, Zhu Y, Mustafi M, Madinier JB, Lelièvre D, Aucagne V, Delmas AF, Weisshaar JC. Real-Time Fluorescence Microscopy on Living E. coli Sheds New Light on the Antibacterial Effects of the King Penguin β-Defensin AvBD103b. Int J Mol Sci 2022; 23:ijms23042057. [PMID: 35216173 PMCID: PMC8880245 DOI: 10.3390/ijms23042057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
Abstract
(1) Antimicrobial peptides (AMPs) are a promising alternative to conventional antibiotics. Among AMPs, the disulfide-rich β-defensin AvBD103b, whose antibacterial activities are not inhibited by salts contrary to most other β-defensins, is particularly appealing. Information about the mechanisms of action is mandatory for the development and approval of new drugs. However, data for non-membrane-disruptive AMPs such as β-defensins are scarce, thus they still remain poorly understood. (2) We used single-cell fluorescence imaging to monitor the effects of a β-defensin (namely AvBD103b) in real time, on living E. coli, and at the physiological concentration of salts. (3) We obtained key parameters to dissect the mechanism of action. The cascade of events, inferred from our precise timing of membrane permeabilization effects, associated with the timing of bacterial growth arrest, differs significantly from the other antimicrobial compounds that we previously studied in the same physiological conditions. Moreover, the AvBD103b mechanism does not involve significant stereo-selective interaction with any chiral partner, at any step of the process. (4) The results are consistent with the suggestion that after penetrating the outer membrane and the cytoplasmic membrane, AvBD103b interacts non-specifically with a variety of polyanionic targets, leading indirectly to cell death.
Collapse
Affiliation(s)
- Céline Landon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; (Y.Z.); (M.M.); (J.C.W.)
- Center for Molecular Biophysics, CNRS, 45071 Orléans, France; (J.-B.M.); (D.L.); (V.A.); (A.F.D.)
- Correspondence:
| | - Yanyu Zhu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; (Y.Z.); (M.M.); (J.C.W.)
| | - Mainak Mustafi
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; (Y.Z.); (M.M.); (J.C.W.)
| | - Jean-Baptiste Madinier
- Center for Molecular Biophysics, CNRS, 45071 Orléans, France; (J.-B.M.); (D.L.); (V.A.); (A.F.D.)
| | - Dominique Lelièvre
- Center for Molecular Biophysics, CNRS, 45071 Orléans, France; (J.-B.M.); (D.L.); (V.A.); (A.F.D.)
| | - Vincent Aucagne
- Center for Molecular Biophysics, CNRS, 45071 Orléans, France; (J.-B.M.); (D.L.); (V.A.); (A.F.D.)
| | - Agnes F. Delmas
- Center for Molecular Biophysics, CNRS, 45071 Orléans, France; (J.-B.M.); (D.L.); (V.A.); (A.F.D.)
| | - James C. Weisshaar
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; (Y.Z.); (M.M.); (J.C.W.)
| |
Collapse
|
11
|
Translocation of non-lytic antimicrobial peptides and bacteria penetrating peptides across the inner membrane of the bacterial envelope. Curr Genet 2021; 68:83-90. [PMID: 34750687 PMCID: PMC8801401 DOI: 10.1007/s00294-021-01217-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/28/2021] [Accepted: 10/06/2021] [Indexed: 11/02/2022]
Abstract
The increase in multidrug-resistant pathogenic bacteria has become a problem worldwide. Currently there is a strong focus on the development of novel antimicrobials, including antimicrobial peptides (AMP) and antimicrobial antisense agents. While the majority of AMP have membrane activity and kill bacteria through membrane disruption, non-lytic AMP are non-membrane active, internalize and have intracellular targets. Antimicrobial antisense agents such as peptide nucleic acids (PNA) and phosphorodiamidate morpholino oligomers (PMO), show great promise as novel antibacterial agents, killing bacteria by inhibiting translation of essential target gene transcripts. However, naked PNA and PMO are unable to translocate across the cell envelope of bacteria, to reach their target in the cytosol, and are conjugated to bacteria penetrating peptides (BPP) for cytosolic delivery. Here, we discuss how non-lytic AMP and BPP-PMO/PNA conjugates translocate across the cytoplasmic membrane via receptor-mediated transport, such as the cytoplasmic membrane transporters SbmA, MdtM/YjiL, and/or YgdD, or via a less well described autonomous process.
Collapse
|
12
|
Sultana A, Luo H, Ramakrishna S. Antimicrobial Peptides and Their Applications in Biomedical Sector. Antibiotics (Basel) 2021; 10:1094. [PMID: 34572676 PMCID: PMC8465024 DOI: 10.3390/antibiotics10091094] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 01/10/2023] Open
Abstract
In a report by WHO (2014), it was stated that antimicrobial resistance is an arising challenge that needs to be resolved. This resistance is a critical issue in terms of disease or infection treatment and is usually caused due to mutation, gene transfer, long-term usage or inadequate use of antimicrobials, survival of microbes after consumption of antimicrobials, and the presence of antimicrobials in agricultural feeds. One of the solutions to this problem is antimicrobial peptides (AMPs), which are ubiquitously present in the environment. These peptides are of concern due to their special mode of action against a wide spectrum of infections and health-related problems. The biomedical field has the highest need of AMPs as it possesses prominent desirable activity against HIV-1, skin cancer, breast cancer, in Behcet's disease treatment, as well as in reducing the release of inflammatory cells such as TNFα, IL-8, and IL-1β, enhancing the production of anti-inflammatory cytokines such as IL-10 and GM-CSF, and in wound healing properties. This review has highlighted all the major functions and applications of AMPs in the biomedical field and concludes the future potential of AMPs.
Collapse
Affiliation(s)
- Afreen Sultana
- Center for Nanotechnology & Sustainability, Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore;
| | - Hongrong Luo
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, China;
| | - Seeram Ramakrishna
- Center for Nanotechnology & Sustainability, Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore;
| |
Collapse
|
13
|
Pacor S, Benincasa M, Musso MV, Krce L, Aviani I, Pallavicini A, Scocchi M, Gerdol M, Mardirossian M. The proline-rich myticalins from Mytilus galloprovincialis display a membrane-permeabilizing antimicrobial mode of action. Peptides 2021; 143:170594. [PMID: 34118363 DOI: 10.1016/j.peptides.2021.170594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022]
Abstract
Bivalve mollusks are continuously exposed to potentially pathogenic microorganisms living in the marine environment. Not surprisingly, these filter-feeders developed a robust innate immunity to protect themselves, which includes a broad panel of antimicrobial peptides. Among these, myticalins represent a recently discovered family of linear cationic peptides expressed in the gills of Mytilus galloprovincialis. Even though myticalins and insect and mammalian proline-rich antimicrobial peptides (PrAMPs) share a similar amino acid composition, we here show that none of the tested mussel peptides use a non-lytic mode of action relying on the bacterial transporter SbmA. On the other hand, all the tested myticalins perturbed and permeabilized the membranes of E. coli BW25113, as shown by flow-cytometry and atomic force microscopy. Circular dichroism spectra revealed that most myticalins did not adopt recognizable secondary structures in the presence of amphipathic environments, such as biological membranes. To explore possible uses of myticalins for biotech, we assessed their biocompatibility with a human cell line. Non-negligible cytotoxic effects displayed by myticalins indicate that their optimization would be required before their further use as lead compounds in the development of new antibiotics.
Collapse
Affiliation(s)
- Sabrina Pacor
- Department of Life Sciences, Via Licio Giorgieri 5, University of Trieste, 34127 Trieste, Italy.
| | - Monica Benincasa
- Department of Life Sciences, Via Licio Giorgieri 5, University of Trieste, 34127 Trieste, Italy.
| | - Maria Valentina Musso
- Department of Life Sciences, Via Licio Giorgieri 5, University of Trieste, 34127 Trieste, Italy.
| | - Lucija Krce
- Department of Physics, Faculty of Science, University of Split, Soba B3-18, Ruđera Boškovića 33, 21000 Split, Croatia.
| | - Ivica Aviani
- Department of Physics, Faculty of Science, University of Split, Soba B3-18, Ruđera Boškovića 33, 21000 Split, Croatia.
| | - Alberto Pallavicini
- Department of Life Sciences, Via Licio Giorgieri 5, University of Trieste, 34127 Trieste, Italy.
| | - Marco Scocchi
- Department of Life Sciences, Via Licio Giorgieri 5, University of Trieste, 34127 Trieste, Italy.
| | - Marco Gerdol
- Department of Life Sciences, Via Licio Giorgieri 5, University of Trieste, 34127 Trieste, Italy.
| | - Mario Mardirossian
- Department of Medical Sciences, University of Trieste, Laboratorio Clinica Odontostomatologica, Piazza dell'Ospitale 1, 34125 Trieste, Italy.
| |
Collapse
|
14
|
Harvesting of Antimicrobial Peptides from Insect (Hermetia illucens) and Its Applications in the Food Packaging. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11156991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
About one-third of the total food produced is wasted, rising the concern to adopt proper management. Simultaneously with the increase in population, demand for food is increasing which may lead to scarcity. Adequate packaging is one of the ways to avoid deterioration of food and prevent wastage. In recent years, active packaging has attained interest due to its commendable results in food preservation. Several studies proved that the embodiment of antimicrobial components into the packaging material has the ability to prevent microbial contamination. Antimicrobial peptides (AMP) are newly discovered antimicrobial agents for impregnation into packaging material. Among various sources for AMP, insects have shown great resistivity against a wide spectrum of microorganisms. Insects feed on substances consisting of a varying range of contaminations, which often results in infections. Insects synthesise AMPs to fight such infections and survive in that atmosphere. The disease-causing agents in humans are the same as those found in insects. Hence, AMPs extracted from insects have the potential to fight the microorganisms that act as hazards to human health. This review highlights the harvesting and synthesis of AMPs from Hermetia illucens, which is a promising source for AMP and its applications in the food packaging industry.
Collapse
|
15
|
Armas F, Di Stasi A, Mardirossian M, Romani AA, Benincasa M, Scocchi M. Effects of Lipidation on a Proline-Rich Antibacterial Peptide. Int J Mol Sci 2021; 22:7959. [PMID: 34360723 PMCID: PMC8347091 DOI: 10.3390/ijms22157959] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 01/04/2023] Open
Abstract
The emergence of multidrug-resistant bacteria is a worldwide health problem. Antimicrobial peptides have been recognized as potential alternatives to conventional antibiotics, but still require optimization. The proline-rich antimicrobial peptide Bac7(1-16) is active against only a limited number of Gram-negative bacteria. It kills bacteria by inhibiting protein synthesis after its internalization, which is mainly supported by the bacterial transporter SbmA. In this study, we tested two different lipidated forms of Bac7(1-16) with the aim of extending its activity against those bacterial species that lack SbmA. We linked a C12-alkyl chain or an ultrashort cationic lipopeptide Lp-I to the C-terminus of Bac7(1-16). Both the lipidated Bac-C12 and Bac-Lp-I forms acquired activity at low micromolar MIC values against several Gram-positive and Gram-negative bacteria. Moreover, unlike Bac7(1-16), Bac-C12, and Bac-Lp-I did not select resistant mutants in E. coli after 14 times of exposure to sub-MIC concentrations of the respective peptide. We demonstrated that the extended spectrum of activity and absence of de novo resistance are likely related to the acquired capability of the peptides to permeabilize cell membranes. These results indicate that C-terminal lipidation of a short proline-rich peptide profoundly alters its function and mode of action and provides useful insights into the design of novel broad-spectrum antibacterial agents.
Collapse
Affiliation(s)
- Federica Armas
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (F.A.); (A.D.S.); (M.M.); (M.B.)
- Area Science Park, Padriciano, 34149 Trieste, Italy
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Adriana Di Stasi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (F.A.); (A.D.S.); (M.M.); (M.B.)
| | - Mario Mardirossian
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (F.A.); (A.D.S.); (M.M.); (M.B.)
- Department of Medical Sciences, University of Trieste, 34129 Trieste, Italy
| | | | - Monica Benincasa
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (F.A.); (A.D.S.); (M.M.); (M.B.)
| | - Marco Scocchi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (F.A.); (A.D.S.); (M.M.); (M.B.)
| |
Collapse
|
16
|
Loffredo MR, Savini F, Bobone S, Casciaro B, Franzyk H, Mangoni ML, Stella L. Inoculum effect of antimicrobial peptides. Proc Natl Acad Sci U S A 2021; 118:e2014364118. [PMID: 34021080 PMCID: PMC8166072 DOI: 10.1073/pnas.2014364118] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The activity of many antibiotics depends on the initial density of cells used in bacterial growth inhibition assays. This phenomenon, termed the inoculum effect, can have important consequences for the therapeutic efficacy of the drugs, because bacterial loads vary by several orders of magnitude in clinically relevant infections. Antimicrobial peptides are a promising class of molecules in the fight against drug-resistant bacteria because they act mainly by perturbing the cell membranes rather than by inhibiting intracellular targets. Here, we report a systematic characterization of the inoculum effect for this class of antibacterial compounds. Minimum inhibitory concentration values were measured for 13 peptides (including all-D enantiomers) and peptidomimetics, covering more than seven orders of magnitude in inoculated cell density. In most cases, the inoculum effect was significant for cell densities above the standard inoculum of 5 × 105 cells/mL, while for lower densities the active concentrations remained essentially constant, with values in the micromolar range. In the case of membrane-active peptides, these data can be rationalized by considering a simple model, taking into account peptide-cell association, and hypothesizing that a threshold number of cell-bound peptide molecules is required in order to cause bacterial killing. The observed effect questions the clinical utility of activity and selectivity determinations performed at a fixed, standardized cell density. A routine evaluation of the dependence of the activity of antimicrobial peptides and peptidomimetics on the inoculum should be considered.
Collapse
Affiliation(s)
- Maria Rosa Loffredo
- Laboratory affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Filippo Savini
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Sara Bobone
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Bruno Casciaro
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Maria Luisa Mangoni
- Laboratory affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy;
| | - Lorenzo Stella
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, 00133 Rome, Italy;
| |
Collapse
|
17
|
Manju Devi S, Raj N, Sashidhar RB. Efficacy of short-synthetic antifungal peptides on pathogenic Aspergillus flavus. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 174:104810. [PMID: 33838711 DOI: 10.1016/j.pestbp.2021.104810] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 06/12/2023]
Abstract
The efficacies of three short synthetic antifungal peptides were tested for their inhibitory action on pathogenic fungi, Aspergillus flavus. The sequences of the short synthetic peptides are PPD1- FRLHF, 66-10-FRLKFH, 77-3- FRLKFHF, respectively. These test peptides inhibited fungal growth and showed a membranolytic activity. The fungal biomass and ergosterol levels were significantly low in peptides treated samples. Further, the fungal cell wall component chitin was also found to be lower in peptides treated samples. Scanning electron microscopic images also showed highly wrinkled fungal mycelia. Significant membrane permeabilisation as well as potassium ion leakage was also observed in fungal samples treated with peptides. To assess the membrane damage, the uptake of Sytox green dye was employed. At tested concentration, peptides induced fungal membrane damage as evidenced by the green fluorescence. Further, at tested concentration, these peptides induced an oxidative stress in A.flavus as evidenced by an increase in the ROS production, malondialdehyde levels, increase in the antioxidant enzymes - superoxide dismutase, catalase with concomitant decrease in the reduced glutathione content. Additionally, a growth dependent reduction in aflatoxin levels were also observed in peptides treated samples. Docking studies on the interaction of the peptides with a trans-membrane protein calcium ATPase of A. flavus showed that all the peptides were able to bind to the protein with high z rank score. The activity of the calcium ATPase was significantly decreased in peptides treated fungal samples, thereby validating the docking results. Among all the tested peptides, 77-3 peptide exhibited the maximal membrane damage property.
Collapse
Affiliation(s)
- S Manju Devi
- Department of Biochemistry, University College of Science, Osmania University, Hyderabad 500007, Telangana State, India
| | - Navya Raj
- Department of Health Informatics, College of Health Sciences, Saudi Electronic University, Dammam, Saudi Arabia
| | - R B Sashidhar
- Department of Biochemistry, University College of Science, Osmania University, Hyderabad 500007, Telangana State, India.
| |
Collapse
|
18
|
Upert G, Luther A, Obrecht D, Ermert P. Emerging peptide antibiotics with therapeutic potential. MEDICINE IN DRUG DISCOVERY 2021; 9:100078. [PMID: 33398258 PMCID: PMC7773004 DOI: 10.1016/j.medidd.2020.100078] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/15/2020] [Accepted: 12/27/2020] [Indexed: 02/09/2023] Open
Abstract
This review covers some of the recent progress in the field of peptide antibiotics with a focus on compounds with novel or established mode of action and with demonstrated efficacy in animal infection models. Novel drug discovery approaches, linear and macrocyclic peptide antibiotics, lipopeptides like the polymyxins as well as peptides addressing targets located in the plasma membrane or in the outer membrane of bacterial cells are discussed.
Collapse
Key Words
- ADMET, absorption, distribution, metabolism and excretion – toxicity in pharmacokinetics
- AMP, antimicrobial peptide
- AMR, antimicrobial resistance
- ATCC, ATCC cell collection
- Antibiotic
- BAM, β-barrel assembly machinery
- CC50, cytotoxic concentration to kill 50% of cells
- CD, circular dichroism
- CFU, colony forming unit
- CLSI, clinical and laboratory standards institute
- CMS, colistin methane sulfonate
- DMPC, 1,2-dimyristoyl-sn-glycero-3-phosphocholine
- ESKAPE, acronym encompassing six bacterial pathogens (often carrying antibiotic resistance): Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumonia, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp
- FDA, U. S. Food and Drug Administration
- HABP, hospital acquired bacterial pneumonia
- HDP, host-defense peptide
- HEK293, human embryonic kidney 293 cells
- HK-2, human kidney 2 cells (proximal tubular cell line)
- HepG2, human hepatocellular carcinoma cell line
- Hpg, 4-hydroxy-phenyl glycine
- ITC, isothermal titration calorimetry
- KPC, Klebsiella pneumoniae metallo-β-lactamase C resistant
- LPS, lipopolysaccharide
- LptA, lipopolysaccharide transport protein A
- LptC, lipopolysaccharide transport protein C
- LptD, lipopolysaccharide transport protein D
- MDR, multidrug-resistant
- MH-I, Müller-Hinton broth I
- MH-II, Müller-Hinton broth II (cation adjusted)
- MIC, minimal inhibitory concentration
- MRSA, methicilline-resistant S. aureus
- MSSA, methicilline-sensitive S. aureus
- MoA, mechanism (mode) of action
- NDM-1, New Delhi metallo-β-lactamase resistant
- NOAEL, no adverse effect level
- ODL, odilorhabdin
- OMPTA (outer membrane targeting antibiotic)
- OMPTA, outer membrane targeting antibiotic
- Omp, outer membrane protein
- PBMC, peripheral mononuclear blood cell
- PBP, penicillin-binding protein
- PBS, phosphate-buffered saline
- PK, pharmacokinetics
- POPC, 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine
- POPG, 2-oleoyl-1-palmitoyl-sn-glycero-3-phospho-(1-glycerol)
- PrAMPs, polyproline antimicrobial peptides
- RBC, red blood cell
- SAR, structure-activity relationship
- SPR, surface plasmon resonance
- SPase I, signal peptidase I
- VABP, ventilator associated bacterial pneumonia
- VIM-1, beta-lactamase 2 (K. pneumoniae)
- VISA, vancomycin-intermediate S. aureus
- VRE, vancomycin-resistant enterococcus
- WHO, World Health Organization
- WT, wild type
- WTA, wall teichoic acid
- XDR, extremely drug-resistant
- antimicrobial peptide
- antimicrobial resistance
- bid, bis in die (two times a day)
- i.p., intraperitoneal
- i.v., intravenous
- lipopeptide
- mITT population, minimal intend-to-treat population
- peptide antibiotic
- s.c., subcutaneous
Collapse
Affiliation(s)
- Gregory Upert
- Polyphor Ltd, Hegenheimermattweg 125, 4123 Allschwil, Switzerland
| | - Anatol Luther
- Bachem AG, Hauptstrasse 114, 4416 Bubendorf, Switzerland
| | - Daniel Obrecht
- Polyphor Ltd, Hegenheimermattweg 125, 4123 Allschwil, Switzerland
| | - Philipp Ermert
- Polyphor Ltd, Hegenheimermattweg 125, 4123 Allschwil, Switzerland
| |
Collapse
|
19
|
Casciaro B, Cappiello F, Verrusio W, Cacciafesta M, Mangoni ML. Antimicrobial Peptides and their Multiple Effects at Sub-Inhibitory Concentrations. Curr Top Med Chem 2021; 20:1264-1273. [PMID: 32338221 DOI: 10.2174/1568026620666200427090912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/06/2020] [Accepted: 03/16/2020] [Indexed: 01/10/2023]
Abstract
The frequent occurrence of multidrug-resistant strains to conventional antimicrobials has led to a clear decline in antibiotic therapies. Therefore, new molecules with different mechanisms of action are extremely necessary. Due to their unique properties, antimicrobial peptides (AMPs) represent a valid alternative to conventional antibiotics and many of them have been characterized for their activity and cytotoxicity. However, the effects that these peptides cause at concentrations below the minimum growth inhibitory concentration (MIC) have yet to be fully analyzed along with the underlying molecular mechanism. In this mini-review, the ability of AMPs to synergize with different antibiotic classes or different natural compounds is examined. Furthermore, data on microbial resistance induction are reported to highlight the importance of antibiotic resistance in the fight against infections. Finally, the effects that sub-MIC levels of AMPs can have on the bacterial pathogenicity are summarized while showing how signaling pathways can be valid therapeutic targets for the treatment of infectious diseases. All these aspects support the high potential of AMPs as lead compounds for the development of new drugs with antibacterial and immunomodulatory activities.
Collapse
Affiliation(s)
- Bruno Casciaro
- Center For Life Nano Science @ Sapienza, Italian Institute of Technology, Rome 00161, Italy
| | - Floriana Cappiello
- Laboratory affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| | - Walter Verrusio
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Mauro Cacciafesta
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Maria Luisa Mangoni
- Laboratory affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| |
Collapse
|
20
|
Kumar R, Ali SA, Singh SK, Bhushan V, Mathur M, Jamwal S, Mohanty AK, Kaushik JK, Kumar S. Antimicrobial Peptides in Farm Animals: An Updated Review on Its Diversity, Function, Modes of Action and Therapeutic Prospects. Vet Sci 2020; 7:vetsci7040206. [PMID: 33352919 PMCID: PMC7766339 DOI: 10.3390/vetsci7040206] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Antimicrobial peptides (AMPs) are the arsenals of the innate host defense system, exhibiting evolutionarily conserved characteristics that are present in practically all forms of life. Recent years have witnessed the emergence of antibiotic-resistant bacteria compounded with a slow discovery rate for new antibiotics that have necessitated scientific efforts to search for alternatives to antibiotics. Research on the identification of AMPs has generated very encouraging evidence that they curb infectious pathologies and are also useful as novel biologics to function as immunotherapeutic agents. Being innate, they exhibit the least cytotoxicity to the host and exerts a wide spectrum of biological activity including low resistance among microbes and increased wound healing actions. Notably, in veterinary science, the constant practice of massive doses of antibiotics with inappropriate withdrawal programs led to a high risk of livestock-associated antimicrobial resistance. Therefore, the world faces tremendous pressure for designing and devising strategies to mitigate the use of antibiotics in animals and keep it safe for posterity. In this review, we illustrate the diversity of farm animal-specific AMPs, and their biochemical foundations, mode of action, and prospective application in clinics. Subsequently, we present the data for their systematic classification under the major and minor groups, antipathogenic action, and allied bioactivities in the host. Finally, we address the limitations of their clinical implementation and envision areas for further advancement.
Collapse
|
21
|
Wang Y, Wang M, Shan A, Feng X. Avian host defense cathelicidins: structure, expression, biological functions, and potential therapeutic applications. Poult Sci 2020; 99:6434-6445. [PMID: 33248558 PMCID: PMC7704953 DOI: 10.1016/j.psj.2020.09.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/14/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022] Open
Abstract
Host defense peptides (HDP) are multifunctional effectors of the innate immune system, which has antimicrobial and pleiotropic immunomodulatory functions. Although there is a very sophisticated superposition of adaptive immune systems in vertebrates, this system is still essential. As an important family of HDP, cathelicidins are also known for their broad-spectrum antibacterial activity against bacteria, fungi, and enveloped viruses. It has been found in humans and other species, including cattle, pigs, sheep, goats, chickens, rabbits, and some kind of fish. Among them, cathelicidins in birds were described for the first time in 2005. This review focuses on the structure, biological activities, expression, and regulation of avian cathelicidin, especially main effects of host defense cathelicidin on potential therapeutic applications. According to the results obtained both in vitro and in vivo, good perspectives have been opened for cathelicidin. Nevertheless, further studies are needed to better characterize the mechanisms of action underlying the beneficial effects of cathelicidin as novel therapeutic alternatives to antibiotics.
Collapse
Affiliation(s)
- Yingjie Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Min Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Xingjun Feng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, People's Republic of China.
| |
Collapse
|
22
|
Sola R, Mardirossian M, Beckert B, Sanghez De Luna L, Prickett D, Tossi A, Wilson DN, Scocchi M. Characterization of Cetacean Proline-Rich Antimicrobial Peptides Displaying Activity against ESKAPE Pathogens. Int J Mol Sci 2020; 21:E7367. [PMID: 33036159 PMCID: PMC7582929 DOI: 10.3390/ijms21197367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/27/2022] Open
Abstract
Proline-rich antimicrobial peptides (PrAMPs) may be a valuable weapon against multi-drug resistant pathogens, combining potent antimicrobial activity with low cytotoxicity. We have identified novel PrAMPs from five cetacean species (cePrAMPs), and characterized their potency, mechanism of action and in vitro cytotoxicity. Despite the homology between the N-terminal of cePrAMPs and the bovine PrAMP Bac7, some differences emerged in their sequence, activity spectrum and mode of action. CePrAMPs with the highest similarity with the Bac7(1-35) fragment inhibited bacterial protein synthesis without membrane permeabilization, while a second subgroup of cePrAMPs was more membrane-active but less efficient at inhibiting bacterial translation. Such differences may be ascribable to differences in presence and positioning of Trp residues and of a conserved motif seemingly required for translation inhibition. Unlike Bac7(1-35), which requires the peptide transporter SbmA for its uptake, the activity of cePrAMPs was mostly independent of SbmA, regardless of their mechanism of action. Two peptides displayed a promisingly broad spectrum of activity, with minimal inhibiting concentration MIC ≤ 4 µM against several bacteria of the ESKAPE group, including Pseudomonas aeruginosa and Enterococcus faecium. Our approach has led us to discover several new peptides; correlating their sequences and mechanism of action will provide useful insights for designing optimized future peptide-based antibiotics.
Collapse
Affiliation(s)
- Riccardo Sola
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (R.S.); (L.S.D.L.); (D.P.); (A.T.); mscocchi.units.it (M.S.)
| | - Mario Mardirossian
- Department of Medical Sciences, University of Trieste, 34125 Trieste, Italy;
| | - Bertrand Beckert
- Institute for Biochemistry and Molecular Biology, University of Hamburg, 20146 Hamburg, Germany; (B.B.); (D.N.W.)
| | - Laura Sanghez De Luna
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (R.S.); (L.S.D.L.); (D.P.); (A.T.); mscocchi.units.it (M.S.)
| | - Dennis Prickett
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (R.S.); (L.S.D.L.); (D.P.); (A.T.); mscocchi.units.it (M.S.)
| | - Alessandro Tossi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (R.S.); (L.S.D.L.); (D.P.); (A.T.); mscocchi.units.it (M.S.)
| | - Daniel N. Wilson
- Institute for Biochemistry and Molecular Biology, University of Hamburg, 20146 Hamburg, Germany; (B.B.); (D.N.W.)
| | - Marco Scocchi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy; (R.S.); (L.S.D.L.); (D.P.); (A.T.); mscocchi.units.it (M.S.)
| |
Collapse
|
23
|
Wu F, Tan C. Dead bacterial absorption of antimicrobial peptides underlies collective tolerance. J R Soc Interface 2020; 16:20180701. [PMID: 30958185 DOI: 10.1098/rsif.2018.0701] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The collective tolerance towards antimicrobial peptides (APs) is thought to occur primarily through mechanisms associated with live bacterial cells. In contrast to the focus on live cells, we discover that the LL37 antimicrobial peptide kills a subpopulation of Escherichia coli, forming dead cells that absorb the remaining LL37 from the environment. Combining mathematical modelling with population and single-cell experiments, we show that bacteria absorb LL37 at a timing that coincides with the permeabilization of their cytoplasmic membranes. Furthermore, we show that one bacterial strain can absorb LL37 and protect another strain from killing by LL37. Finally, we demonstrate that the absorption of LL37 by dead bacteria can be reduced using a peptide adjuvant. In contrast to the known collective tolerance mechanisms, we show that the absorption of APs by dead bacteria is a dynamic process that leads to emergent population behaviour.
Collapse
Affiliation(s)
- Fan Wu
- Department of Biomedical Engineering, University of California Davis , Davis, CA 95616 , USA
| | - Cheemeng Tan
- Department of Biomedical Engineering, University of California Davis , Davis, CA 95616 , USA
| |
Collapse
|
24
|
Doolin T, Gross S, Siryaporn A. Physical Mechanisms of Bacterial Killing by Histones. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1267:117-133. [PMID: 32894480 DOI: 10.1007/978-3-030-46886-6_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibiotic resistance is a global epidemic, becoming increasingly pressing due to its rapid spread. There is thus a critical need to develop new therapeutic approaches. In addition to searching for new antibiotics, looking into existing mechanisms of natural host defense may enable researchers to improve existing defense mechanisms, and to develop effective, synthetic drugs guided by natural principles. Histones, primarily known for their role in condensing mammalian DNA, are antimicrobial and share biochemical similarities with antimicrobial peptides (AMPs); however, the mechanism by which histones kill bacteria is largely unknown. Both AMPs and histones are similar in size, cationic, contain a high proportion of hydrophobic amino acids, and possess the ability to form alpha helices. AMPs, which mostly kill bacteria through permeabilization or disruption of the biological membrane, have recently garnered significant attention for playing a key role in host defenses. This chapter outlines the structure and function of histone proteins as they compare to AMPs and provides an overview of their role in innate immune responses, especially regarding the action of specific histones against microorganisms and their potential mechanism of action against microbial pathogens.
Collapse
Affiliation(s)
- Tory Doolin
- Department of Developmental and Cell Biology, UC Irvine, Irvine, CA, USA
| | - Steven Gross
- Department of Developmental and Cell Biology, UC Irvine, Irvine, CA, USA. .,Department of Physics & Astronomy, UC Irvine, Irvine, CA, USA.
| | - Albert Siryaporn
- Department of Physics & Astronomy, UC Irvine, Irvine, CA, USA. .,Department of Molecular Biology & Biochemistry, UC Irvine, Irvine, CA, USA.
| |
Collapse
|
25
|
Theansungnoen T, Jangpromma N, Anwised P, Daduang S, Fukumori Y, Taoka A, Klaynongsruang S. Membranolytic Effects of KT2 on Gram-Negative Escherichia coli Evaluated by Atomic Force Microscopy. APPL BIOCHEM MICRO+ 2019. [DOI: 10.1134/s0003683819050144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
26
|
Zhang R, Wang Z, Tian Y, Yin Q, Cheng X, Lian M, Zhou B, Zhang X, Yang L. Efficacy of Antimicrobial Peptide DP7, Designed by Machine-Learning Method, Against Methicillin-Resistant Staphylococcus aureus. Front Microbiol 2019; 10:1175. [PMID: 31191493 PMCID: PMC6546875 DOI: 10.3389/fmicb.2019.01175] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/08/2019] [Indexed: 02/05/2023] Open
Abstract
Antimicrobial peptides (AMPs) provide a promising strategy against infections involving multidrug-resistant pathogens. In previous studies, we designed a short 12 amino acid AMP DP7, using a machine-learning method based on an amino acid activity contribution matrix. DP7 shows broad-spectrum antimicrobial activities both in vitro and in vivo. Here, we aim to investigate the efficacy of DP7 against multidrug resistant Staphylococcus aureus (S. aureus) and reveal the potential mechanisms. First, by measuring the killing kinetics of DP7 against S. aureus and comparing these results with antibiotics with different antimicrobial mechanisms, we hypothesize that DP7, in addition to its known ability to induce cell wall cation damage, can also exert a full killing effect. With FITC-conjugated or biotin-labeled DP7, we tracked DP7's attachment, membrane permeation and subsequent intracellular distribution in S. aureus. These results indicated that the possible targets of DP7 were within the bacterial cells. Transcriptome sequencing of S. aureus exposed to DP7 identified 333 differentially expressed genes (DEGs) influenced by DP7, involving nucleic acid metabolism, amino acid biosynthesis, cell wall destruction and pathogenesis, respectively, indicating the comprehensive killing efficacy of DP7. In addition, the genome sequencing results of the induced DP7 resistant strain S. aureus DP7-R revealed two-point mutations in the mprF and guaA gene. Moreover, in a murine model for MRSA blood stream infection, intravenously treating mice with DP7 showed a good protective effect on mice. In conclusion, DP7 is an effective bactericide for S. aureus, which deserves further study for clinical application and drug development.
Collapse
Affiliation(s)
- Rui Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Zhenling Wang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yaomei Tian
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qi Yin
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xingjun Cheng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Mao Lian
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Bailing Zhou
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xueyan Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Holfeld L, Knappe D, Hoffmann R. Proline-rich antimicrobial peptides show a long-lasting post-antibiotic effect on Enterobacteriaceae and Pseudomonas aeruginosa. J Antimicrob Chemother 2019; 73:933-941. [PMID: 29309652 DOI: 10.1093/jac/dkx482] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/21/2017] [Indexed: 01/28/2023] Open
Abstract
Background Proline-rich antimicrobial peptides (PrAMPs) represent a promising class of potential therapeutics to treat multiresistant infections. They inhibit bacterial protein translation at the 70S ribosome by either blocking the peptide-exit tunnel (oncocin type) or trapping release factors (apidaecin type). Objectives Besides direct concentration-dependent antibacterial effects, the post-antibiotic effect (PAE) is the second most important criterion of antimicrobial pharmacodynamics to be determined in vitro. Here, PAEs of 10 PrAMPs and three antibiotics against three Escherichia coli strains, Klebsiella pneumoniae ATCC 10031 and Pseudomonas aeruginosa ATCC 27853 were studied after 1 h of exposure. Methods A robust high-throughput screening to determine PAEs was established, i.e. liquid handling by a 96-channel pipetting system and continuous incubation and absorbance measurement in a microplate reader. Results Prolonged PAEs (≥4 h) were detected for all peptides at their MIC values against all strains; PAEs were even >10 h for Api88, Api137, Bac7(1-60) and A3-APO. The PAEs increased further at 4 × MIC. Aminoglycosides gentamicin and kanamycin usually showed lower PAEs (≤4 h) at MIC, but PAEs increased to > 10 h at 4 × MIC. Bacteriostatic chloramphenicol exhibited the shortest PAEs (<4 h). Conclusions The PAEs of PrAMPs studied against Enterobacteriaceae and P. aeruginosa for the first time were typically 4-fold stronger than for conventional antibiotics. Together with their fast and irreversible uptake by bacteria, the observed prolonged PAE of PrAMPs helps to explain their high in vivo efficacy despite unfavourable pharmacokinetics.
Collapse
Affiliation(s)
- Luzia Holfeld
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, Deutscher Platz 5, Leipzig, Germany.,Center for Biotechnology and Biomedicine, Universität Leipzig, Deutscher Platz 5, Leipzig, Germany
| | - Daniel Knappe
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, Deutscher Platz 5, Leipzig, Germany.,Center for Biotechnology and Biomedicine, Universität Leipzig, Deutscher Platz 5, Leipzig, Germany
| | - Ralf Hoffmann
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, Universität Leipzig, Deutscher Platz 5, Leipzig, Germany.,Center for Biotechnology and Biomedicine, Universität Leipzig, Deutscher Platz 5, Leipzig, Germany
| |
Collapse
|
28
|
Devi MS, Sashidhar RB. Antiaflatoxigenic effects of selected antifungal peptides. Peptides 2019; 115:15-26. [PMID: 30776385 DOI: 10.1016/j.peptides.2019.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 12/11/2022]
Abstract
Aflatoxins are potent carcinogenic mycotoxins produced as secondary metabolites mainly by the fungi Aspergillus flavus and Aspergillus parasiticus. Control measures to curtail the contamination of aflatoxin in food products is still a challenge. Although there are several reports on the antifungal peptides, there is no specific study on the action of antifungal peptides on aflatoxin synthesis. This work details the effect of four antimicrobial peptides (AMPs) - PPD1 (FRLHF), 66-10 (FRLKFH), 77-3 (FRLKFHF) and D4E1 (FKLRAKIKVRLRAKIKL) on the aflatoxin production by A. flavus and A. parasiticus. Results of the investigations suggests that AMPs at near minimum inhibitory concentrations (MIC) were effectively inhibiting aflatoxins, without hindering the growth of the fungi. These AMPs, at concentrations near MIC, induced membrane permeabilisation, without inducing cellular leakage. The involvement of oxidative stress for the aflatoxin synthesis was reversed by the antioxidant nature of the peptides as evidenced by the 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulphonic acid (ABTS) assay, reactive oxygen species production, malondialdehyde and antioxidant enzymes analysis. Quantitative real time polymerase chain reaction (RT-qPCR) analysis of the aflatoxin gene cluster showed that 'aflR' and its downstream genes expressions were significantly down regulated. Conidiation of the fungi were negatively influenced by the peptides as evidenced by scanning electron microscopy analysis and RT-qPCR. mRNA levels of Manganese-superoxide dismutase (Mn-SOD) showed a decrease in the expression in RT-qPCR. The effect of these peptides on aflatoxin inhibition provides insight into their use as novel antiaflatoxigenic molecules.
Collapse
Affiliation(s)
- Manju S Devi
- Department of Biochemistry, University College of Science, Osmania University, Hyderabad-500 007, Telangana State, India
| | - R B Sashidhar
- Department of Biochemistry, University College of Science, Osmania University, Hyderabad-500 007, Telangana State, India.
| |
Collapse
|
29
|
Armas F, Pacor S, Ferrari E, Guida F, Pertinhez TA, Romani AA, Scocchi M, Benincasa M. Design, antimicrobial activity and mechanism of action of Arg-rich ultra-short cationic lipopeptides. PLoS One 2019; 14:e0212447. [PMID: 30789942 PMCID: PMC6383929 DOI: 10.1371/journal.pone.0212447] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/01/2019] [Indexed: 01/01/2023] Open
Abstract
The increasing emergence of multidrug-resistant microorganisms represents one of the greatest challenges in the clinical management of infectious diseases, and requires the development of novel antimicrobial agents. To this aim, we de novo designed a library of Arg-rich ultra-short cationic antimicrobial lipopeptides (USCLs), based on the Arg-X-Trp-Arg-NH2 peptide moiety conjugated with a fatty acid, and investigated their antibacterial potential. USCLs exhibited an excellent antimicrobial activity against clinically pathogenic microorganisms, in particular Gram-positive bacteria, including multidrug resistant strains, with MIC values ranging between 1.56 and 6.25 μg/mL. The capability of the two most active molecules, Lau-RIWR-NH2 and Lau-RRIWRR-NH2, to interact with the bacterial membranes has been predicted by molecular dynamics and verified on liposomes by surface plasmon resonance. Both compounds inhibited the growth of S. aureus even at sub MIC concentrations and induced cell membranes permeabilization by producing visible cell surface alterations leading to a significant decrease in bacterial viability. Interestingly, no cytotoxic effects were evidenced for these lipopeptides up to 50–100 μg/mL in hemolysis assay, in human epidermal model and HaCaT cells, thus highlighting a good cell selectivity. These results, together with the simple composition of USCLs, make them promising lead compounds as new antimicrobials.
Collapse
Affiliation(s)
- Federica Armas
- Department of Life Sciences, University of Trieste, Trieste, Italy
- Area Science Park, Padriciano, Trieste, Italy
| | - Sabrina Pacor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Elena Ferrari
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Filomena Guida
- Department of Life Sciences, University of Trieste, Trieste, Italy
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Thelma A. Pertinhez
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Transfusion Medicine Unit, AUSL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | | | - Marco Scocchi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Monica Benincasa
- Department of Life Sciences, University of Trieste, Trieste, Italy
- * E-mail:
| |
Collapse
|
30
|
Intracellular Antimicrobial Peptides Targeting the Protein Synthesis Machinery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1117:73-89. [PMID: 30980354 DOI: 10.1007/978-981-13-3588-4_6] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
While antimicrobial peptides (AMPs) are well-known for their disruptive effects on bacterial membranes, the mechanism of many intracellular AMPs is still being elucidated. In the recent years, it has been demonstrated that the subclass of proline-rich AMPs (PrAMPs) can pass through the bacterial membrane and kill bacteria by inhibiting protein synthesis. PrAMPs are a product of the innate immune system and are secreted in response to bacterial infection. So far PrAMPs have been identified in many arthropods, such as beetles, wasps, and flies, as well as some mammals, such as sheep, cows, and goats. PrAMPs show high potency against Gram-negative bacteria, while exhibiting low toxicity in eukaryotes, suggesting that they may represent a promising avenue for the development of future antimicrobial agents to combat the increase of multidrug-resistant bacterial pathogens. Structural and biochemical data have revealed the PrAMP binding sites on the ribosome as well as insight into their mechanisms of action. While the binding site of all so far investigated PrAMPs is situated within nascent polypeptide exit tunnel, the mechanism of action is distinct between class I and II PrAMPs. Specifically, class I PrAMPs, such as Bac7, Onc112, pyrrhocoricin, and metalnikowin, block the delivery of aa-tRNA by EF-Tu to the ribosomal A-site, whereas the class II PrAMPs, such as apidaecin 1b and Api137, act during translation termination and inhibit protein synthesis by trapping of release factors on the 70S ribosome following hydrolysis of the nascent polypeptide chain.
Collapse
|
31
|
Panteleev PV, Bolosov IA, Kalashnikov AÀ, Kokryakov VN, Shamova OV, Emelianova AA, Balandin SV, Ovchinnikova TV. Combined Antibacterial Effects of Goat Cathelicidins With Different Mechanisms of Action. Front Microbiol 2018; 9:2983. [PMID: 30555455 PMCID: PMC6284057 DOI: 10.3389/fmicb.2018.02983] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022] Open
Abstract
Being essential components of innate immune system, animal antimicrobial peptides (AMPs) also known as host-defense peptides came into sharp focus as possible alternatives to conventional antibiotics due to their high efficacy against a broad range of MDR pathogens and low rate of resistance development. Mammalian species can produce a set of co-localized AMPs with different structures and mechanisms of actions. Here we examined the combined antibacterial effects of cathelicidins, structurally diverse family of host-defense peptides found in vertebrate species. As a model we have used structurally distinct cathelicidins expressed in the leukocytes of goat Capra hircus. The recombinant analogs of natural peptides were obtained by heterologous expression in bacterial system and biological activities as well as the major mechanisms of antibacterial action of the peptides were investigated. As the result, the marked synergistic effect against wide panel of bacterial strains including extensively drug-resistant ones was observed for the pair of membranolytic α-helical amphipathic peptide ChMAP-28 and Pro-rich peptide mini-ChBac7.5Nα targeting a bacterial ribosome. ChMAP-28 was shown to damage the outer bacterial membrane at sub-inhibitory concentrations that could facilitate Pro-rich peptide translocation into the cell. Finally, resistance changes under a long-term continuous selective pressure of each individual peptide and the synergistic combination of both peptides were tested against Escherichia coli strains. The combination was shown to keep a high activity after the 26-days selection experiment in contrast to mini-ChBac7.5Nα used alone and the reference antibiotic polymyxin B. We identified the point mutation leading to amino acid substitution V102E in the membrane transport protein SbmA of the mini-ChBac7.5Nα-resistant strain obtained by selection. The experiments revealed that the presence of sub-inhibitory concentrations of ChMAP-28 restored the activity of mini-ChBac7.5Nα against this strain and clinical isolate with a weak sensitivity to mini-ChBac7.5Nα. The obtained results suggest a potential medical application of synergistic combinations of natural cathelicidins, which allows using a lower therapeutic dose and minimizes the risk of resistance development.
Collapse
Affiliation(s)
- Pavel V Panteleev
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ilia A Bolosov
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexander À Kalashnikov
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Olga V Shamova
- Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Anna A Emelianova
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Sergey V Balandin
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Tatiana V Ovchinnikova
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
32
|
Hornerin contains a Linked Series of Ribosome-Targeting Peptide Antibiotics. Sci Rep 2018; 8:16158. [PMID: 30385807 PMCID: PMC6212518 DOI: 10.1038/s41598-018-34467-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/15/2018] [Indexed: 11/15/2022] Open
Abstract
Cationic intrinsically disordered antimicrobial peptides (CIDAMPs) belong to a novel class of epithelial peptide antibiotics with microbicidal activity against various pathogens, including Pseudomonas aeruginosa, Escherichia coli, Staphylococcus aureus and Candida albicans. Here we show that treatment of distinct bacteria with different hornerin (HRNR)-derived CIDAMPs cause formation of unique cytoplasmic protein aggregates, suggesting a common intracellular mode of action. We further found that, unlike most amphipathic antimicrobial peptides, HRNR traverses bacterial membranes energy-dependently and accumulates within the cytoplasm. Strikingly, certain structurally different, HRNR-based CIDAMPs were found to bind to an identical panel of distinct bacterial ribosomal proteins, thereby manifesting features of several known classes of antibiotics. This may cause the formation of aberrant proteins and toxic protein aggregates in HRNR-treated pathogens which eventually may induce its death. Our study reveals evidence that structurally distinct CIDAMPs of an abundant body surface protein simultaneously target multiple sites of the bacterial protein synthesis machinery.
Collapse
|
33
|
Fragments of the Nonlytic Proline-Rich Antimicrobial Peptide Bac5 Kill Escherichia coli Cells by Inhibiting Protein Synthesis. Antimicrob Agents Chemother 2018; 62:AAC.00534-18. [PMID: 29844040 DOI: 10.1128/aac.00534-18] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/19/2018] [Indexed: 11/20/2022] Open
Abstract
Unlike most antimicrobial peptides (AMPs), the main mode of action of the subclass of proline-rich antimicrobial peptides (PrAMPs) is not based on disruption of the bacterial membrane. Instead, PrAMPs exploit the inner membrane transporters SbmA and YjiL/MdtM to pass through the bacterial membrane and enter the cytosol of specific Gram-negative bacteria, where they exert an inhibitory effect on protein synthesis. Despite sharing a high proline and arginine content with other characterized PrAMPs, the PrAMP Bac5 has a low sequence identity with them. Here we investigated the mode of action of three N-terminal Bac5 fragments, Bac5(1-15), Bac5(1-25), and Bac5(1-31). We show that Bac5(1-25) and Bac5(1-31) retained excellent antimicrobial activity toward Escherichia coli and low toxicity toward eukaryotic cells, whereas Bac5(1-15) was inactive. Bac5(1-25) and Bac5(1-31) inhibited bacterial protein synthesis in vitro and in vivo Competition assays suggested that the binding site of Bac5 is within the ribosomal tunnel, where it prevents the transition from the initiation to the elongation phase of translation, as reported for other PrAMPs, such as the bovine PrAMP Bac7. Surprisingly, unlike Bac7, Bac5(1-25) exhibited species-specific inhibition, being an excellent inhibitor of protein synthesis on E. coli ribosomes but a poor inhibitor on Thermus thermophilus ribosomes. This indicates that while Bac5 most likely has an overlapping binding site with Bac7, the mode of interaction is distinct, suggesting that Bac5 fragments may be interesting alternative lead compounds for the development of new antimicrobial agents.
Collapse
|
34
|
Ciociola T, Giovati L, Giovannelli A, Conti S, Castagnola M, Vitali A. The activity of a mammalian proline-rich peptide against Gram-negative bacteria, including drug-resistant strains, relies on a nonmembranolytic mode of action. Infect Drug Resist 2018; 11:969-979. [PMID: 30046246 PMCID: PMC6054295 DOI: 10.2147/idr.s165179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background A peptide of 2,733 Da named SP-E, previously isolated from pig saliva and already described for its antifungal activity and absence of toxicity against mammalian cells, is characterized by a high content of proline residues (70% of entire sequence), that confer structural features probably related to peptide activity. Purpose The aim of this study was to evaluate the activity of SP-E against Gram-negative bacteria, including drug-resistant clinical isolates. Methods SP-E and shorter fragments of the same peptide were tested in vitro against the selected bacteria by colony forming unit assays. Scanning electron microscopy and confocal microscopy were also applied. SP-E potential therapeutic activity was evaluated in vivo in a Galleria mellonella model of bacterial infection. Results SP-E proved to be active against the tested bacteria with EC50 values in the micro-molar range. Though maintaining antibacterial properties, the shorter peptides showed lower activity in respect to the parental molecule. Kinetics of killing action and nonmembranolytic internalization within Escherichia coli and Pseudomonas aeruginosa cells strongly suggested a cytosolic mechanism of action involving one or more intracellular molecular targets. A single injection of SP-E exerted a therapeutic effect in G. mellonella larvae infected with P. aeruginosa. Conclusion The biological properties of SP-E strongly back this peptide as a new promising multitasking antimicrobial molecule.
Collapse
Affiliation(s)
- Tecla Ciociola
- Department of Medicine and Surgery, University of Parma, Parma,
| | - Laura Giovati
- Department of Medicine and Surgery, University of Parma, Parma,
| | - Angela Giovannelli
- Institute of Biochemistry and Clinical Biochemistry, Catholic University, Rome
| | - Stefania Conti
- Department of Medicine and Surgery, University of Parma, Parma,
| | - Massimo Castagnola
- Institute of Biochemistry and Clinical Biochemistry, Catholic University, Rome.,Institute for the Chemistry of Molecular Recognition, C.N.R., c/o Institute of Biochemistry and Clinical Biochemistry, Catholic University, Rome, Italy
| | - Alberto Vitali
- Institute for the Chemistry of Molecular Recognition, C.N.R., c/o Institute of Biochemistry and Clinical Biochemistry, Catholic University, Rome, Italy
| |
Collapse
|
35
|
Important Late-Stage Symbiotic Role of the Sinorhizobium meliloti Exopolysaccharide Succinoglycan. J Bacteriol 2018; 200:JB.00665-17. [PMID: 29632097 DOI: 10.1128/jb.00665-17] [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: 11/04/2017] [Accepted: 04/06/2018] [Indexed: 12/18/2022] Open
Abstract
Sinorhizobium meliloti enters into beneficial symbiotic interactions with Medicago species of legumes. Bacterial exopolysaccharides play critical signaling roles in infection thread initiation and growth during the early stages of root nodule formation. After endocytosis of S. meliloti by plant cells in the developing nodule, plant-derived nodule-specific cysteine-rich (NCR) peptides mediate terminal differentiation of the bacteria into nitrogen-fixing bacteroids. Previous transcriptional studies showed that the intensively studied cationic peptide NCR247 induces expression of the exo genes that encode the proteins required for succinoglycan biosynthesis. In addition, genetic studies have shown that some exo mutants exhibit increased sensitivity to the antimicrobial action of NCR247. Therefore, we investigated whether the symbiotically active S. meliloti exopolysaccharide succinoglycan can protect S. meliloti against the antimicrobial activity of NCR247. We discovered that high-molecular-weight forms of succinoglycan have the ability to protect S. meliloti from the antimicrobial action of the NCR247 peptide but low-molecular-weight forms of wild-type succinoglycan do not. The protective function of high-molecular-weight succinoglycan occurs via direct molecular interactions between anionic succinoglycan and the cationic NCR247 peptide, but this interaction is not chiral. Taken together, our observations suggest that S. meliloti exopolysaccharides not only may be critical during early stages of nodule invasion but also are upregulated at a late stage of symbiosis to protect bacteria against the bactericidal action of cationic NCR peptides. Our findings represent an important step forward in fully understanding the complete set of exopolysaccharide functions during legume symbiosis.IMPORTANCE Symbiotic interactions between rhizobia and legumes are economically important for global food production. The legume symbiosis also is a major part of the global nitrogen cycle and is an ideal model system to study host-microbe interactions. Signaling between legumes and rhizobia is essential to establish symbiosis, and understanding these signals is a major goal in the field. Exopolysaccharides are important in the symbiotic context because they are essential signaling molecules during early-stage symbiosis. In this study, we provide evidence suggesting that the Sinorhizobium meliloti exopolysaccharide succinoglycan also protects the bacteria against the antimicrobial action of essential late-stage symbiosis plant peptides.
Collapse
|
36
|
Mardirossian M, Pérébaskine N, Benincasa M, Gambato S, Hofmann S, Huter P, Müller C, Hilpert K, Innis CA, Tossi A, Wilson DN. The Dolphin Proline-Rich Antimicrobial Peptide Tur1A Inhibits Protein Synthesis by Targeting the Bacterial Ribosome. Cell Chem Biol 2018; 25:530-539.e7. [PMID: 29526712 PMCID: PMC6219704 DOI: 10.1016/j.chembiol.2018.02.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 01/08/2018] [Accepted: 02/05/2018] [Indexed: 12/22/2022]
Abstract
Proline-rich antimicrobial peptides (PrAMPs) internalize into susceptible bacteria using specific transporters and interfere with protein synthesis and folding. To date, mammalian PrAMPs have so far been identified only in artiodactyls. Since cetaceans are co-phyletic with artiodactyls, we mined the genome of the bottlenose dolphin Tursiops truncatus, leading to the identification of two PrAMPs, Tur1A and Tur1B. Tur1A, which is orthologous to the bovine PrAMP Bac7, is internalized into Escherichia coli, without damaging the membranes, using the inner membrane transporters SbmA and YjiL/MdM. Furthermore, like Bac7, Tur1A also inhibits bacterial protein synthesis by binding to the ribosome and blocking the transition from the initiation to the elongation phase. By contrast, Tur1B is a poor inhibitor of protein synthesis and may utilize another mechanism of action. An X-ray structure of Tur1A bound within the ribosomal exit tunnel provides a basis to develop these peptides as novel antimicrobial agents.
Collapse
Affiliation(s)
- Mario Mardirossian
- Gene Center, Department for Biochemistry and Center for Integrated Protein Sciences, Munich (CiPSM), University of Munich, 81377 Munich, Germany
| | - Natacha Pérébaskine
- ARNA Laboratory, University of Bordeaux, Inserm U1212, CNRS UMR 5320, IECB, 33607 Pessac, France
| | - Monica Benincasa
- Antimicrobial Peptides Laboratory, Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Stefano Gambato
- Antimicrobial Peptides Laboratory, Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Sven Hofmann
- Institute of Infection and Immunity, St George's, University of London, SW17 0RE London, UK
| | - Paul Huter
- Gene Center, Department for Biochemistry and Center for Integrated Protein Sciences, Munich (CiPSM), University of Munich, 81377 Munich, Germany; Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Claudia Müller
- Gene Center, Department for Biochemistry and Center for Integrated Protein Sciences, Munich (CiPSM), University of Munich, 81377 Munich, Germany; Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Kai Hilpert
- Institute of Infection and Immunity, St George's, University of London, SW17 0RE London, UK
| | - C Axel Innis
- ARNA Laboratory, University of Bordeaux, Inserm U1212, CNRS UMR 5320, IECB, 33607 Pessac, France
| | - Alessandro Tossi
- Antimicrobial Peptides Laboratory, Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Daniel N Wilson
- Gene Center, Department for Biochemistry and Center for Integrated Protein Sciences, Munich (CiPSM), University of Munich, 81377 Munich, Germany; Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany.
| |
Collapse
|
37
|
Polikanov YS, Aleksashin NA, Beckert B, Wilson DN. The Mechanisms of Action of Ribosome-Targeting Peptide Antibiotics. Front Mol Biosci 2018; 5:48. [PMID: 29868608 PMCID: PMC5960728 DOI: 10.3389/fmolb.2018.00048] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/23/2018] [Indexed: 12/31/2022] Open
Abstract
The ribosome is one of the major targets in the cell for clinically used antibiotics. However, the increase in multidrug resistant bacteria is rapidly reducing the effectiveness of our current arsenal of ribosome-targeting antibiotics, highlighting the need for the discovery of compounds with new scaffolds that bind to novel sites on the ribosome. One possible avenue for the development of new antimicrobial agents is by characterization and optimization of ribosome-targeting peptide antibiotics. Biochemical and structural data on ribosome-targeting peptide antibiotics illustrates the large diversity of scaffolds, binding interactions with the ribosome as well as mechanism of action to inhibit translation. The availability of high-resolution structures of ribosomes in complex with peptide antibiotics opens the way to structure-based design of these compounds as novel antimicrobial agents.
Collapse
Affiliation(s)
- Yury S Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States.,Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, IL, United States
| | - Nikolay A Aleksashin
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Bertrand Beckert
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Daniel N Wilson
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
38
|
Domalaon R, Sanchak Y, Koskei LC, Lyu Y, Zhanel GG, Arthur G, Schweizer F. Short Proline-Rich Lipopeptide Potentiates Minocycline and Rifampin against Multidrug- and Extensively Drug-Resistant Pseudomonas aeruginosa. Antimicrob Agents Chemother 2018; 62:e02374-17. [PMID: 29437631 PMCID: PMC5914005 DOI: 10.1128/aac.02374-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/27/2018] [Indexed: 12/11/2022] Open
Abstract
A series of 16 short proline-rich lipopeptides (SPRLPs) were constructed to mimic longer naturally existing proline-rich antimicrobial peptides. Antibacterial assessment revealed that lipopeptides containing hexadecanoic acid (C16) possess optimal antibacterial activity relative to others with shorter lipid components. SPRLPs were further evaluated for their potential to serve as adjuvants in combination with existing antibiotics to enhance antibacterial activity against drug-resistant Pseudomonas aeruginosa Out of 16 prepared SPRLPs, C12-PRP was found to significantly potentiate the antibiotics minocycline and rifampin against multidrug- and extensively drug-resistant (MDR/XDR) P. aeruginosa clinical isolates. This nonhemolytic C12-PRP is comprised of the heptapeptide sequence PRPRPRP-NH2 acylated to dodecanoic acid (C12) at the N terminus. The adjuvant potency of C12-PRP was apparent by its ability to reduce the MIC of minocycline and rifampin below their interpretative susceptibility breakpoints against MDR/XDR P. aeruginosa An attempt to optimize C12-PRP through peptidomimetic modification was performed by replacing all l- to d-amino acids. C12-PRP demonstrated that it was amenable to optimization, since synergism with minocycline and rifampin were retained. Moreover, C12-PRP displayed no cytotoxicity against human liver carcinoma HepG2 and human embryonic kidney HEK-293 cell lines. Thus, the SPRLP C12-PRP is a lead adjuvant candidate that warrants further optimization. The discovery of agents that are able to resuscitate the activity of existing antibiotics against drug-resistant Gram-negative pathogens, especially P. aeruginosa, is of great clinical interest.
Collapse
Affiliation(s)
- Ronald Domalaon
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yaroslav Sanchak
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Linet Cherono Koskei
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- School of Biological Sciences, University of Nairobi, Nairobi, Kenya
| | - Yinfeng Lyu
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - George G Zhanel
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gilbert Arthur
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Frank Schweizer
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
39
|
Chongsiriwatana NP, Lin JS, Kapoor R, Wetzler M, Rea JAC, Didwania MK, Contag CH, Barron AE. Intracellular biomass flocculation as a key mechanism of rapid bacterial killing by cationic, amphipathic antimicrobial peptides and peptoids. Sci Rep 2017; 7:16718. [PMID: 29196622 PMCID: PMC5711933 DOI: 10.1038/s41598-017-16180-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/08/2017] [Indexed: 12/19/2022] Open
Abstract
Many organisms rely on antimicrobial peptides (AMPs) as a first line of defense against pathogens. In general, most AMPs are thought to kill bacteria by binding to and disrupting cell membranes. However, certain AMPs instead appear to inhibit biomacromolecule synthesis, while causing less membrane damage. Despite an unclear understanding of mechanism(s), there is considerable interest in mimicking AMPs with stable, synthetic molecules. Antimicrobial N-substituted glycine (peptoid) oligomers ("ampetoids") are structural, functional and mechanistic analogs of helical, cationic AMPs, which offer broad-spectrum antibacterial activity and better therapeutic potential than peptides. Here, we show through quantitative studies of membrane permeabilization, electron microscopy, and soft X-ray tomography that both AMPs and ampetoids trigger extensive and rapid non-specific aggregation of intracellular biomacromolecules that correlates with microbial death. We present data demonstrating that ampetoids are "fast killers", which rapidly aggregate bacterial ribosomes in vitro and in vivo. We suggest intracellular biomass flocculation is a key mechanism of killing for cationic, amphipathic AMPs, which may explain why most AMPs require micromolar concentrations for activity, show significant selectivity for killing bacteria over mammalian cells, and finally, why development of resistance to AMPs is less prevalent than developed resistance to conventional antibiotics.
Collapse
Affiliation(s)
- Nathaniel P Chongsiriwatana
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, United States
| | - Jennifer S Lin
- Department of Bioengineering, Stanford University, Stanford, California, United States
| | - Rinki Kapoor
- Biophysics Program, Stanford University, Stanford, California, United States
| | - Modi Wetzler
- Department of Bioengineering, Stanford University, Stanford, California, United States
| | - Jennifer A C Rea
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, United States
| | - Maruti K Didwania
- Department of Bioengineering, Stanford University, Stanford, California, United States
| | - Christopher H Contag
- Departments of Microbiology and Immunology, Pediatrics, and Radiology, Stanford University, Stanford, California, United States
| | - Annelise E Barron
- Department of Bioengineering, Stanford University, Stanford, California, United States.
| |
Collapse
|
40
|
Selective phenylalanine to proline substitution for improved antimicrobial and anticancer activities of peptides designed on phenylalanine heptad repeat. Acta Biomater 2017; 57:170-186. [PMID: 28483698 DOI: 10.1016/j.actbio.2017.05.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/19/2017] [Accepted: 05/04/2017] [Indexed: 01/09/2023]
Abstract
Introducing cell-selectivity in antimicrobial peptides (AMPs) without compromising the antimicrobial and anti-endotoxin properties is a crucial step towards the development of new antimicrobial agents. A peptide designed on phenylalanine heptad repeat possesses significant cytotoxicity along with desired antimicrobial and anti-endotoxin properties. Amino acid substitutions at 'a' and/or 'd' positions of heptad repeats of AMPs could alter their helical structure in mammalian membrane-mimetic environments and cytotoxicity towards mammalian cells. Since proline is a helix breaker, effects of selective proline substitution(s) at 'a' and/or 'd' positions of a 15-residue peptide designed on phenylalanine heptad repeat (FR-15) were investigated. Proline-substituted FR-15 variants were highly selective toward bacteria and fungi over hRBCs and murine 3T3 cells and also retained their antibacterial activities at high salt, serum and elevated temperatures. These non-cytotoxic variants also inhibited LPS-induced production of pro-inflammatory cytokines/chemokines in human monocytes, THP-1, RAW 264.7 and in BALB/c mice. The two non-cytotoxic variants (FR8P and FR11P) showed potent anti-cancer activity against highly metastatic human breast cancer cell line MDA-MB-231 with IC50 values less than 10μM. At sub-IC50 concentrations, FR8P and FR11P also showed anti-migratory and anti-invasive effects against MDA-MB-231 cells. FR8P and FR11P induced cellular apoptosis by triggering intrinsic apoptotic pathway through depolarization of mitochondrial membrane potential and activation of caspases. Overall the results demonstrated the utilization of selective phenylalanine to proline substitution in a heptad repeat of phenylalanine residues for the design of cell-selective, broad-spectrum AMPs with significant anti-cancer properties. STATEMENT OF SIGNIFICANCE We have demonstrated a methodology to design cell-selective potent antimicrobial and anti-endotoxin peptides by utilizing phenylalanine zipper as a template and replacement of phenylalanine residue(s) from "a" and/or "d" position(s) with proline residue(s) produced non-cytotoxic AMPs with improved antibacterial properties against the drug-resistant strains of bacteria. The work showed that the 'a' and 'd' positions of the phenylalanine heptad repeat could be replaced by an appropriate amino acid to control cytotoxicity of the peptide without compromising its potency in antimicrobial and anti-endotoxin properties. The direct bacterial membrane targeting mechanism of proline substituted analogs of parent peptide makes difficult for bacteria to grow resistance against them. The peptides designed could be lead molecules in the area of sepsis as they possess significant anti-LPS activities for in vitro and in vivo. Interestingly since cancer cells and bacterial cell membranes possess the structural resemblances, the cancer cells are also targets for these peptides making them lead molecules in this field. However, unlike in bacteria where the peptides showed membrane permeabilization property to lyse them, the peptides induced apoptosis in MDA-MB-231 breast cancer cells to inhibit their proliferation and growth. The results are significant because it reveals that "a" and "d" positions of a phenylalanine zipper can be utilized as switches to design cell-selective, antimicrobial, anti-endotoxin and anticancer peptides.
Collapse
|
41
|
The Mechanism of Killing by the Proline-Rich Peptide Bac7(1-35) against Clinical Strains of Pseudomonas aeruginosa Differs from That against Other Gram-Negative Bacteria. Antimicrob Agents Chemother 2017; 61:AAC.01660-16. [PMID: 28137800 DOI: 10.1128/aac.01660-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/15/2017] [Indexed: 12/13/2022] Open
Abstract
Pseudomonas aeruginosa infections represent a serious threat to worldwide health. Proline-rich antimicrobial peptides (PR-AMPs), a particular group of peptide antibiotics, have demonstrated in vitro activity against P. aeruginosa strains. Here we show that the mammalian PR-AMP Bac7(1-35) is active against some multidrug-resistant cystic fibrosis isolates of P. aeruginosa By confocal microscopy and cytometric analyses, we investigated the mechanism of killing against P. aeruginosa strain PAO1 and three selected isolates, and we observed that the peptide inactivated the target cells by disrupting their cellular membranes. This effect is deeply different from that previously described for PR-AMPs in Escherichia coli and Salmonella enterica serovar Typhimurium, where these peptides act intracellularly after having been internalized by means of the transporter SbmA without membranolytic effects. The heterologous expression of SbmA in PAO1 cells enhanced the internalization of Bac7(1-35) into the cytoplasm, making the bacteria more susceptible to the peptide but at the same time more resistant to the membrane lysis, similarly to what occurs in E. coli The results evidenced a new mechanism of action for PR-AMPs and indicate that Bac7 has multiple and variable modes of action that depend on the characteristics of the different target species and the possibility to be internalized by bacterial transporters. This feature broadens the spectrum of activity of the peptide and makes the development of peptide-resistant bacteria a more difficult process.
Collapse
|
42
|
Abstract
Antimicrobial peptides (AMPs) are expressed in various living organisms as first-line host defenses against potential harmful encounters in their surroundings. AMPs are short polycationic peptides exhibiting various antimicrobial activities. The principal antibacterial activity is attributed to the membrane-lytic mechanism which directly interferes with the integrity of the bacterial cell membrane and cell wall. In addition, a number of AMPs form a transmembrane channel in the membrane by self-aggregation or polymerization, leading to cytoplasm leakage and cell death. However, an increasing body of evidence has demonstrated that AMPs are able to exert intracellular inhibitory activities as the primary or supportive mechanisms to achieve efficient killing. In this review, we focus on the major intracellular targeting activities reported in AMPs, which include nucleic acids and protein biosynthesis and protein-folding, protease, cell division, cell wall biosynthesis, and lipopolysaccharide inhibition. These multifunctional AMPs could serve as the potential lead peptides for the future development of novel antibacterial agents with improved therapeutic profiles.
Collapse
|
43
|
Arasu A, Kumaresan V, Palanisamy R, Arasu MV, Al-Dhabi NA, Ganesh MR, Arockiaraj J. Bacterial membrane binding and pore formation abilities of carbohydrate recognition domain of fish lectin. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 67:202-212. [PMID: 27729229 DOI: 10.1016/j.dci.2016.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/06/2016] [Accepted: 10/06/2016] [Indexed: 06/06/2023]
Abstract
Antimicrobial peptides (AMPs) are innate molecules that are found in a wide variety of species ranging from bacteria to humans. In recent years, excessive usage of antibiotics resulted in development of multi-drug resistant pathogens which made researchers to focus on AMPs as potential substitute for antibiotics. Lily type mannose-binding lectin is an extended super-family of structurally and evolutionarily related sugar binding proteins. These lectins are well-known AMPs which play important roles in fish defense mechanism. Here, we report a full-length lily type lectin-2 (LTL-2) identified from the cDNA library of striped murrel, Channa striatus (Cs). CsLTL-2 protein contained B-lectin domain along with three carbohydrate binding sites which is a prominent characteristic functional feature of LTL. The mRNA transcripts of CsLTL-2 were predominantly expressed in gills and considerably up-regulated upon infection with fungus (Aphanomyces invadans) and bacteria (Aeromonas hydrophila). To evaluate the antimicrobial activity of the carbohydrate binding region of CsLTL-2, the region was synthesized (QP13) and its bactericidal activity was analyzed. In addition, QP13 was labeled with fluorescein isothiocyanate (FITC) and its binding affinity with the bacterial cell membranes was analyzed. Minimum inhibitory concentration assay revealed that QP13 inhibited the growth of Escherichia coli at a concentration of 80 μM/ml. Confocal microscopic observation showed that FITC tagged QP13 specifically bound to the bacterial membrane. Fluorescence assisted cell sorter (FACS) assay showed that QP13 reduced the bacterial cell count drastically. Therefore, the mechanism of action of QP13 on E. coli cells was determined by propidium iodide internalization assay which confirmed that QP13 induced bacterial membrane disruption. Moreover, the peptide did not show any cytotoxicity towards fish peripheral blood leucocytes. Taken together, these results support the potentiality of QP13 that can be used as an antimicrobial agent against the tested pathogens.
Collapse
Affiliation(s)
- Abirami Arasu
- Division of Fisheries Biotechnology & Molecular Biology, Department of Biotechnology, Faculty of Science and Humanities, SRM University, Kattankulathur, 603 203, Chennai, Tamil Nadu, India; Department of Microbiology, SRM Arts & Science College, Kattankulathur, 603 203, Chennai, India
| | - Venkatesh Kumaresan
- Division of Fisheries Biotechnology & Molecular Biology, Department of Biotechnology, Faculty of Science and Humanities, SRM University, Kattankulathur, 603 203, Chennai, Tamil Nadu, India
| | - Rajesh Palanisamy
- Division of Fisheries Biotechnology & Molecular Biology, Department of Biotechnology, Faculty of Science and Humanities, SRM University, Kattankulathur, 603 203, Chennai, Tamil Nadu, India
| | - Mariadhas Valan Arasu
- Department of Botany and Microbiology, Addiriyah Chair for Environmental Studies, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Naif Abdullah Al-Dhabi
- Department of Botany and Microbiology, Addiriyah Chair for Environmental Studies, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Munuswamy-Ramanujam Ganesh
- Interdisciplinary Institute of Indian System of Medicine, SRM University, Kattankulathur, 603 203, Chennai, Tamil Nadu, India
| | - Jesu Arockiaraj
- Division of Fisheries Biotechnology & Molecular Biology, Department of Biotechnology, Faculty of Science and Humanities, SRM University, Kattankulathur, 603 203, Chennai, Tamil Nadu, India.
| |
Collapse
|
44
|
Rončević T, Gajski G, Ilić N, Goić-Barišić I, Tonkić M, Zoranić L, Simunić J, Benincasa M, Mijaković M, Tossi A, Juretić D. PGLa-H tandem-repeat peptides active against multidrug resistant clinical bacterial isolates. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:228-237. [DOI: 10.1016/j.bbamem.2016.11.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/20/2016] [Accepted: 11/22/2016] [Indexed: 12/20/2022]
|
45
|
Graf M, Mardirossian M, Nguyen F, Seefeldt AC, Guichard G, Scocchi M, Innis CA, Wilson DN. Proline-rich antimicrobial peptides targeting protein synthesis. Nat Prod Rep 2017; 34:702-711. [DOI: 10.1039/c7np00020k] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Proline-rich antimicrobial peptides (PrAMPs) bind within the exit tunnel of the ribosome and inhibit translation elongation. Structures of ribosome-bound PrAMPs reveal the interactions with ribosomal components and could pave the way for the development of novel peptide-based antimicrobial agents.
Collapse
Affiliation(s)
- Michael Graf
- Gene Center
- Department for Biochemistry and Center for Integrated Protein Sciences Munich (CiPS-M)
- University of Munich
- 81377 Munich
- Germany
| | - Mario Mardirossian
- Gene Center
- Department for Biochemistry and Center for Integrated Protein Sciences Munich (CiPS-M)
- University of Munich
- 81377 Munich
- Germany
| | - Fabian Nguyen
- Gene Center
- Department for Biochemistry and Center for Integrated Protein Sciences Munich (CiPS-M)
- University of Munich
- 81377 Munich
- Germany
| | | | - Gilles Guichard
- Université de Bordeaux
- CNRS
- Institut Polytechnique de Bordeaux
- UMR 5248
- Institut de Chimie et Biologie des Membranes et des Nano-objets (CBMN)
| | - Marco Scocchi
- Department of Life Sciences
- University of Trieste
- Trieste
- Italy
| | - C. Axel Innis
- Univ. Bordeaux
- ARNA Laboratory
- Inserm U1212
- CNRS UMR 5320
- IECB
| | - Daniel N. Wilson
- Gene Center
- Department for Biochemistry and Center for Integrated Protein Sciences Munich (CiPS-M)
- University of Munich
- 81377 Munich
- Germany
| |
Collapse
|
46
|
D'Este F, Benincasa M, Cannone G, Furlan M, Scarsini M, Volpatti D, Gennaro R, Tossi A, Skerlavaj B, Scocchi M. Antimicrobial and host cell-directed activities of Gly/Ser-rich peptides from salmonid cathelicidins. FISH & SHELLFISH IMMUNOLOGY 2016; 59:456-468. [PMID: 27818338 DOI: 10.1016/j.fsi.2016.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/28/2016] [Accepted: 11/02/2016] [Indexed: 06/06/2023]
Abstract
Cathelicidins, a major family of vertebrate antimicrobial peptides (AMPs), have a recognized role in the first line of defense against infections. They have been identified in several salmonid species, where the putative mature peptides are unusually long and rich in serine and glycine residues, often arranged in short multiple repeats (RLGGGS/RPGGGS) intercalated by hydrophobic motifs. Fragments of 24-40 residues, spanning specific motifs and conserved sequences in grayling or brown, rainbow and brook trout, were chemically synthesized and examined for antimicrobial activity against relevant Gram-positive and Gram-negative salmonid pathogens, as well as laboratory reference strains. They were not active in complete medium, but showed varying potency and activity spectra in diluted media. Bacterial membrane permeabilization also occurred only under these conditions and was indicated by rapid propidium iodide uptake in peptide-treated bacteria. However, circular dichroism analyses indicated that they did not significantly adopt ordered conformations in membrane-like environments. The peptides were not hemolytic or cytotoxic to trout cells, including freshly purified head kidney leukocytes (HKL) and the fibroblastic RTG-2 cell line. Notably, when exposed to them, HKL showed increased metabolic activity, while a growth-promoting effect was observed on RTG-2 cells, suggesting a functional interaction of salmonid cathelicidins with host cells similar to that shown by mammalian ones. The three most active peptides produced a dose-dependent increase in phagocytic uptake by HKL simultaneously stimulated with bacterial particles. The peptide STF(1-37), selected for further analyses, also enhanced phagocytic uptake in the presence of autologous serum, and increased intracellular killing of live E. coli. Furthermore, when tested on HKL in combination with the immunostimulant β-glucan, it synergistically potentiated both phagocytic uptake and the respiratory burst response, activities that play a key role in fish immunity. Collectively, these data point to a role of salmonid cathelicidins as modulators of fish microbicidal mechanisms beyond a salt-sensitive antimicrobial activity, and encourage further studies also in view of potential applications in aquaculture.
Collapse
Affiliation(s)
- Francesca D'Este
- Department of Medical and Biological Sciences, Piazzale Kolbe 4, 33100 Udine, Italy
| | - Monica Benincasa
- Department of Life Sciences, University of Trieste, Via Giorgieri 5, 34127 Trieste, Italy
| | - Giuseppe Cannone
- Department of Life Sciences, University of Trieste, Via Giorgieri 5, 34127 Trieste, Italy
| | - Michela Furlan
- Department of Life Sciences, University of Trieste, Via Giorgieri 5, 34127 Trieste, Italy
| | - Michele Scarsini
- Department of Medical and Biological Sciences, Piazzale Kolbe 4, 33100 Udine, Italy
| | - Donatella Volpatti
- Department of Agricultural, Food, Environmental and Animal Sciences, Via Sondrio 2/a, 33100 Udine, Italy
| | - Renato Gennaro
- Department of Life Sciences, University of Trieste, Via Giorgieri 5, 34127 Trieste, Italy
| | - Alessandro Tossi
- Department of Life Sciences, University of Trieste, Via Giorgieri 5, 34127 Trieste, Italy
| | - Barbara Skerlavaj
- Department of Medical and Biological Sciences, Piazzale Kolbe 4, 33100 Udine, Italy
| | - Marco Scocchi
- Department of Life Sciences, University of Trieste, Via Giorgieri 5, 34127 Trieste, Italy.
| |
Collapse
|
47
|
The polymyxin B-induced transcriptomic response of a clinical, multidrug-resistant Klebsiella pneumoniae involves multiple regulatory elements and intracellular targets. BMC Genomics 2016; 17:737. [PMID: 27801293 PMCID: PMC5088521 DOI: 10.1186/s12864-016-3070-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background The emergence of multidrug-resistant Klebsiella pneumoniae is a major public health concern. Many K. pneumoniae infections can only be treated when resorting to last-line drugs such as polymyxin B (PB). However, resistance to this antibiotic is also observed, although insufficient information is described on its mode of action as well as the mechanisms used by resistant bacteria to evade its effects. We aimed to study PB resistance and the influence of abiotic stresses in a clinical K. pneumoniae strain using whole transcriptome profiling. Results We sequenced 12 cDNA libraries of K. pneumoniae Kp13 bacteria, from two biological replicates of the original strain Kp13 (Kp13) and five derivative strains: induced high-level PB resistance in acidic pH (Kp13pH), magnesium deprivation (Kp13Mg), high concentrations of calcium (Kp13Ca) and iron (Kp13Fe), and a control condition with PB (Kp13PolB). Our results show the involvement of multiple regulatory loci that differentially respond to each condition as well as a shared gene expression response elicited by PB treatment, and indicate the participation of two-regulatory components such as ArcA-ArcB, which could be involved in re-routing the K. pneumoniae metabolism following PB treatment. Modules of co-expressed genes could be determined, which correlated to growth in acid stress and PB exposure. We hypothesize that polymyxin B induces metabolic shifts in K. pneumoniae that could relate to surviving against the action of this antibiotic. Conclusions We obtained whole transcriptome data for K. pneumoniae under different environmental conditions and PB treatment. Our results supports the notion that the K. pneumoniae response to PB exposure goes beyond damaged membrane reconstruction and involves recruitment of multiple gene modules and intracellular targets. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3070-y) contains supplementary material, which is available to authorized users.
Collapse
|
48
|
Schneider VAF, Coorens M, Ordonez SR, Tjeerdsma-van Bokhoven JLM, Posthuma G, van Dijk A, Haagsman HP, Veldhuizen EJA. Imaging the antimicrobial mechanism(s) of cathelicidin-2. Sci Rep 2016; 6:32948. [PMID: 27624595 PMCID: PMC5021996 DOI: 10.1038/srep32948] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 08/16/2016] [Indexed: 11/10/2022] Open
Abstract
Host defence peptides (HDPs) have the potential to become alternatives to conventional antibiotics in human and veterinary medicine. The HDP chicken cathelicidin-2 (CATH-2) has immunomodulatory and direct killing activities at micromolar concentrations. In this study the mechanism of action of CATH-2 against Escherichia coli (E. coli) was investigated in great detail using a unique combination of imaging and biophysical techniques. Live-imaging with confocal fluorescence microscopy demonstrated that FITC-labelled CATH-2 mainly localized at the membrane of E. coli. Upon binding, the bacterial membrane was readily permeabilized as was shown by propidium iodide influx into the cell. Concentration- and time-dependent effects of the peptide on E. coli cells were examined by transmission electron microscopy (TEM). CATH-2 treatment was found to induce dose-dependent morphological changes in E. coli. At sub-minimal inhibitory concentrations (sub-MIC), intracellular granulation, enhanced vesicle release and wrinkled membranes were observed, while membrane breakage and cell lysis occurred at MIC values. These effects were visible within 1–5 minute of peptide exposure. Immuno-gold TEM showed CATH-2 binding to bacterial membranes. At sub-MIC values the peptide rapidly localized intracellularly without visible membrane permeabilization. It is concluded that CATH-2 has detrimental effects on E. coli at concentrations that do not immediately kill the bacteria.
Collapse
Affiliation(s)
- Viktoria A F Schneider
- Department of Infectious Diseases and Immunology, Division Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Maarten Coorens
- Department of Infectious Diseases and Immunology, Division Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Soledad R Ordonez
- Department of Infectious Diseases and Immunology, Division Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Johanna L M Tjeerdsma-van Bokhoven
- Department of Infectious Diseases and Immunology, Division Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - George Posthuma
- Department of Cell Biology, Cell Microscopy Core, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Albert van Dijk
- Department of Infectious Diseases and Immunology, Division Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Henk P Haagsman
- Department of Infectious Diseases and Immunology, Division Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Edwin J A Veldhuizen
- Department of Infectious Diseases and Immunology, Division Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
49
|
Benincasa M, Lagatolla C, Dolzani L, Milan A, Pacor S, Liut G, Tossi A, Cescutti P, Rizzo R. Biofilms from Klebsiella pneumoniae: Matrix Polysaccharide Structure and Interactions with Antimicrobial Peptides. Microorganisms 2016; 4:microorganisms4030026. [PMID: 27681920 PMCID: PMC5039586 DOI: 10.3390/microorganisms4030026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/20/2016] [Accepted: 08/02/2016] [Indexed: 01/15/2023] Open
Abstract
Biofilm matrices of two Klebsiella pneumoniae clinical isolates, KpTs101 and KpTs113, were investigated for their polysaccharide composition and protective effects against antimicrobial peptides. Both strains were good biofilm producers, with KpTs113 forming flocs with very low adhesive properties to supports. Matrix exopolysaccharides were isolated and their monosaccharide composition and glycosidic linkage types were defined. KpTs101 polysaccharide is neutral and composed only of galactose, in both pyranose and furanose ring configurations. Conversely, KpTs113 polysaccharide is anionic due to glucuronic acid units, and also contains glucose and mannose residues. The susceptibility of the two strains to two bovine cathelicidin antimicrobial peptides, BMAP-27 and Bac7(1–35), was assessed using both planktonic cultures and biofilms. Biofilm matrices exerted a relevant protection against both antimicrobials, which act with quite different mechanisms. Similar protection was also detected when antimicrobial peptides were tested against planktonic bacteria in the presence of the polysaccharides extracted from KpTs101 and KpTs113 biofilms, suggesting sequestering adduct formation with antimicrobials. Circular dichroism experiments on BMAP-27 in the presence of increasing amounts of either polysaccharide confirmed their ability to interact with the peptide and induce an α-helical conformation.
Collapse
Affiliation(s)
- Monica Benincasa
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 1, Trieste I-34127, Italy.
| | - Cristina Lagatolla
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 1, Trieste I-34127, Italy.
| | - Lucilla Dolzani
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 1, Trieste I-34127, Italy.
| | - Annalisa Milan
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 1, Trieste I-34127, Italy.
| | - Sabrina Pacor
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 1, Trieste I-34127, Italy.
| | - Gianfranco Liut
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 1, Trieste I-34127, Italy.
| | - Alessandro Tossi
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 1, Trieste I-34127, Italy.
| | - Paola Cescutti
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 1, Trieste I-34127, Italy.
| | - Roberto Rizzo
- Department of Life Sciences, University of Trieste, Via L. Giorgieri 1, Trieste I-34127, Italy.
| |
Collapse
|
50
|
Balandin SV, Ovchinnikova TV. Antimicrobial peptides of invertebrates. Part 2. biological functions and mechanisms of action. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2016. [DOI: 10.1134/s106816201604004x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|