1
|
Fliss O, Guay LD, Fliss I, Biron É. Synthesis and structure-activity study of the antimicrobial lipopeptide brevibacillin. RSC Med Chem 2024:d4md00612g. [PMID: 39371433 PMCID: PMC11450366 DOI: 10.1039/d4md00612g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
The antimicrobial lipopeptide brevibacillin is a non-ribosomally synthesized peptide produced by Brevibacillus laterosporus with inhibitory activity against several clinically relevant Gram-positive pathogenic bacteria such as Staphylococcus aureus, Listeria monocytogenes, and Clostridium difficile. In this study, we report the total synthesis of brevibacillin and analogues thereof as well as structure-activity relationship and cytotoxicity studies. Several novel synthetic analogues exhibited high inhibitory activities with minimal inhibitory concentration values in the low micromolar range against several bacteria including Gram-positive L. monocytogenes, S. aureus, Enterococcus faecalis, and Clostridium perfringens as well as Gram-negative Campylobacter coli and Pseudomonas aeruginosa. Of particular interest, four analogues showed a broad spectrum of action and greater antimicrobial activity versus cytotoxicity ratios than native brevibacillin. With a more accessible and efficient production process and improved pharmacological properties, these synthetic analogues are promising candidates to prevent and control the proliferation of various pathogens in the food industry as well as veterinary and human medicine.
Collapse
Affiliation(s)
- Omar Fliss
- Faculté de Pharmacie, Université Laval and Laboratory of Medicinal Chemistry, Centre de recherche du CHU de Québec-Université Laval Québec (QC) G1V 0A6 Canada
- Département des sciences des aliments, Faculté des sciences de l'agriculture et de l'alimentation, Université Laval Québec (QC) G1V 0A6 Canada
- Research Center in Infectious Diseases and Institute of Nutrition and Functional Foods, Université Laval Québec (QC) G1V 0A6 Canada
| | - Louis-David Guay
- Faculté de Pharmacie, Université Laval and Laboratory of Medicinal Chemistry, Centre de recherche du CHU de Québec-Université Laval Québec (QC) G1V 0A6 Canada
- Research Center in Infectious Diseases and Institute of Nutrition and Functional Foods, Université Laval Québec (QC) G1V 0A6 Canada
- PROTEO, The Quebec Network for Research on Protein Function, Engineering, and Applications Québec Canada
| | - Ismail Fliss
- Département des sciences des aliments, Faculté des sciences de l'agriculture et de l'alimentation, Université Laval Québec (QC) G1V 0A6 Canada
- Research Center in Infectious Diseases and Institute of Nutrition and Functional Foods, Université Laval Québec (QC) G1V 0A6 Canada
| | - Éric Biron
- Faculté de Pharmacie, Université Laval and Laboratory of Medicinal Chemistry, Centre de recherche du CHU de Québec-Université Laval Québec (QC) G1V 0A6 Canada
- Research Center in Infectious Diseases and Institute of Nutrition and Functional Foods, Université Laval Québec (QC) G1V 0A6 Canada
- PROTEO, The Quebec Network for Research on Protein Function, Engineering, and Applications Québec Canada
| |
Collapse
|
2
|
Decker T, Rautenbach M, Khan S, Khan W. Antibacterial efficacy and membrane mechanism of action of the Serratia-derived non-ionic lipopeptide, serrawettin W2-FL10. Microbiol Spectr 2024; 12:e0295223. [PMID: 38842361 PMCID: PMC11218446 DOI: 10.1128/spectrum.02952-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 05/14/2024] [Indexed: 06/07/2024] Open
Abstract
The study aimed to investigate the antibacterial activity, cytotoxicity, and mechanism of action of the non-ionic, cyclic lipopeptide, serrawettin W2-FL10 against Staphylococcus aureus. W2-FL10 exhibited potent activity against the Gram-positive bacteria S. aureus, Enterococcus faecalis, Enterococcus faecium, Listeria monocytogenes, and Bacillus subtilis, with minimum inhibitory concentration (MIC) values ranging from 6.3 to 31.3 μg/mL, while no activity was observed against Gram-negative bacteria. Broth microdilution assays showed that W2-FL10 interacted with key cell membrane components, such as lipid phosphatidyl glycerol and lipoteichoic acid of S. aureus. Upon membrane interaction, W2-FL10 dissipated membrane potential within 12 min and increased S. aureus membrane permeability within 28-40 min, albeit at slower rates and higher concentrations than the lytic peptide melittin. The observed membrane permeability, as detected with propidium iodide (PI), may be attributed to transmembrane pores/lesions, possibly dependent on dimer-driven lipopeptide oligomerization in the membrane. Scanning electron microscopy (SEM) imaging also visually confirmed the formation of lesions in the cell wall of one of the S. aureus strains, and cell damage within 1 h of exposure to W2-FL10, corroborating the rapid time-kill kinetics of the S. aureus strains. This bactericidal action against the S. aureus strains corresponded to membrane permeabilization by W2-FL10, indicating that self-promoted uptake into the cytosol may be part of the mode of action. Finally, this lipopeptide exhibited low to moderate cytotoxicity to the Chinese hamster ovarian (CHO) cell line in comparison to the control (emetine) with an optimal lipophilicity range (log D value of 2.5), signifying its potential as an antibiotic candidate. IMPORTANCE Antimicrobial resistance is a major public health concern, urgently requiring antibacterial compounds exhibiting low adverse health effects. In this study, a novel antibacterial lipopeptide analog is described, serrawettin W2-FL10 (derived from Serratia marcescens), with potent activity displayed against Staphylococcus aureus. Mechanistic studies revealed that W2-FL10 targets the cell membrane of S. aureus, causing depolarization and permeabilization because of transmembrane lesions/pores, resulting in the leakage of intracellular components, possible cytosolic uptake of W2-FL10, and ultimately cell death. This study provides the first insight into the mode of action of a non-ionic lipopeptide. The low to moderate cytotoxicity of W2-FL10 also highlights its application as a promising therapeutic agent for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Tanya Decker
- Water Health Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, Johannesburg, Gauteng, South Africa
| | - Marina Rautenbach
- BioPep Peptide Group, Department of Biochemistry, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Sehaam Khan
- Water Health Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, Johannesburg, Gauteng, South Africa
| | - Wesaal Khan
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
3
|
Akinwale AD, Parang K, Tiwari RK, Yamaki J. Mechanistic Study of Antimicrobial Effectiveness of Cyclic Amphipathic Peptide [R 4W 4] against Methicillin-Resistant Staphylococcus aureus Clinical Isolates. Antibiotics (Basel) 2024; 13:555. [PMID: 38927221 PMCID: PMC11201061 DOI: 10.3390/antibiotics13060555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial peptides (AMPs) are being explored as a potential strategy to combat antibiotic resistance due to their ability to reduce susceptibility to antibiotics. This study explored whether the [R4W4] peptide mode of action is bacteriostatic or bactericidal using modified two-fold serial dilution and evaluating the synergism between gentamicin and [R4W4] against Escherichia coli (E. coli) and methicillin-resistant Staphylococcus aureus (MRSA) by a checkered board assay. [R4W4] exhibited bactericidal activity against bacterial isolates (MBC/MIC ≤ 4), with a synergistic effect with gentamicin against E. coli (FICI = 0.3) but not against MRSA (FICI = 0.75). Moreover, we investigated the mechanism of action of [R4W4] against MRSA by applying biophysical assays to evaluate zeta potential, cytoplasmic membrane depolarization, and lipoteichoic acid (LTA) binding affinity. [R4W4] at a 16 mg/mL concentration stabilized the zeta potential of MRSA -31 ± 0.88 mV to -8.37 mV. Also, [R4W4] at 2 × MIC and 16 × MIC revealed a membrane perturbation process associated with concentration-dependent effects. Lastly, in the presence of BODIPY-TR-cadaverine (BC) fluorescence dyes, [R4W4] exhibited binding affinity to LTA comparable with melittin, the positive control. In addition, the antibacterial activity of [R4W4] against MRSA remained unchanged in the absence and presence of LTA, with an MIC of 8 µg/mL. Therefore, the [R4W4] mechanism of action is deemed bactericidal, involving interaction with bacterial cell membranes, causing concentration-dependent membrane perturbation. Additionally, after 30 serial passages, there was a modest increment of MRSA strains resistant to [R4W4] and a change in antibacterial effectiveness MIC [R4W4] and vancomycin by 8 and 4 folds with a slight change in Levofloxacin MIC 1 to 2 µg/mL. These data suggest that [R4W4] warrants further consideration as a potential AMP.
Collapse
Affiliation(s)
- Ajayi David Akinwale
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University School of Pharmacy, Irvine, CA 92618, USA (K.P.)
- Department of Pharmacy Practice, Harry and Diane Rinker Health Science Campus, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University School of Pharmacy, Irvine, CA 92618, USA (K.P.)
| | - Rakesh Kumar Tiwari
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific–Northwest, Western University of Health Sciences, Lebanon, OR 97355, USA
| | - Jason Yamaki
- Department of Pharmacy Practice, Harry and Diane Rinker Health Science Campus, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| |
Collapse
|
4
|
Kim J, Kim J, Yun H, Ganbaatar B, Tahmasebi A, Seo SI, Kim PI, Lee CW. New strain Brevibacillus laterosporus TSA31-5 produces both brevicidine and brevibacillin, exhibiting distinct antibacterial modes of action against Gram-negative and Gram-positive bacteria. PLoS One 2024; 19:e0294474. [PMID: 38558002 PMCID: PMC10984550 DOI: 10.1371/journal.pone.0294474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/01/2023] [Indexed: 04/04/2024] Open
Abstract
The growing prevalence of antibiotic resistance has made it imperative to search for new antimicrobial compounds derived from natural products. In the present study, Brevibacillus laterosporus TSA31-5, isolated from red clay soil, was chosen as the subject for conducting additional antibacterial investigations. The fractions exhibiting the highest antibacterial activity (30% acetonitrile eluent from solid phase extraction) were purified through RP-HPLC. Notably, two compounds (A and B) displayed the most potent antibacterial activity against both Escherichia coli and Staphylococcus aureus. ESI-MS/MS spectroscopy and NMR analysis confirmed that compound A corresponds to brevicidine and compound B to brevibacillin. Particularly, brevicidine displayed notable antibacterial activity against Gram-negative bacteria, with a minimum inhibitory concentration (MIC) range of 1-8 μg/mL. On the other hand, brevibacillin exhibited robust antimicrobial effectiveness against both Gram-positive bacterial strains (MIC range of 2-4 μg/mL) and Gram-negative bacteria (MIC range of 4-64 μg/mL). Scanning electron microscopy analysis and fluorescence assays uncovered distinctive morphological alterations in bacterial cell membranes induced by brevicidine and brevibacillin. These observations imply distinct mechanisms of antibacterial activity exhibited by the peptides. Brevicidine exhibited no hemolysis or cytotoxicity up to 512 μg/mL, comparable to the negative control. This suggests its promising therapeutic potential in treating infectious diseases. Conversely, brevibacillin demonstrated elevated cytotoxicity in in vitro assays. Nonetheless, owing to its noteworthy antimicrobial activity against pathogenic bacteria, brevibacillin could still be explored as a promising antimicrobial agent.
Collapse
Affiliation(s)
- Jeongeun Kim
- Department of Chemistry, Chonnam National University, Gwangju, Republic of Korea
| | - Jueun Kim
- Department of Chemistry, Chonnam National University, Gwangju, Republic of Korea
- Research Center, DAESANG InnoPark, Gangseo-gu, Seoul, Republic of Korea
| | - Hyosuk Yun
- Department of Chemistry, Chonnam National University, Gwangju, Republic of Korea
| | | | - Aminallah Tahmasebi
- Department of Chemistry, Chonnam National University, Gwangju, Republic of Korea
- Department of Agriculture, Minab Higher Education Center, University of Hormozgan, Bandar Abbas, Iran
| | - Sun Il Seo
- Center for Industrialization of Agricultural and Livestock Microorganism, Jeongeup-si, Jeollabuk-do, Republic of Korea
| | - Pyoung Il Kim
- Center for Industrialization of Agricultural and Livestock Microorganism, Jeongeup-si, Jeollabuk-do, Republic of Korea
| | - Chul Won Lee
- Department of Chemistry, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
5
|
Luo Z, Li P, Zhang D, Zhu J, Wang W, Zhao W, Li P, Yuan G. A Novel Antimicrobial Mechanism of Azalomycin F Acting on Lipoteichoic Acid Synthase and Cell Envelope. Molecules 2024; 29:856. [PMID: 38398608 PMCID: PMC10893547 DOI: 10.3390/molecules29040856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Lipoteichoic acid (LTA) plays an essential role in bacterial growth and resistance to antibiotics, and LTA synthetase (LtaS) was considered as an attractive target for combating Gram-positive infections. Azalomycin F, a natural guanidyl-containing polyhydroxy macrolide, can target the LTA of Staphylococcus aureus. Using various technologies including enzyme-linked immunosorbent assay, transmission electron microscope, proteomics, and parallel reaction monitoring, here, the experimental results indicated that azalomycin F can accelerate the LTA release and disrupt the cell envelope, which would also lead to the feedback upregulation on the expressions of LtaS and other related enzymes. Simultaneously, the reconstituted enzyme activity evaluations showed that azalomycin F can significantly inhibit the extracellular catalytic domain of LtaS (eLtaS), while this was vague for LtaS embedded in the liposomes. Subsequently, the fluorescence analyses for five incubation systems containing azalomycin F and eLtaS or the LtaS-embedded liposome indicated that azalomcyin F can spontaneously bind to the active center of LtaS. Combining the mass spectroscopy analyses and the molecular dockings, the results further indicated that this interaction involves the binding sites of substrates and the LTA prolongation, especially the residues Lys299, Phe353, Trp354 and His416. All these suggested that azalomycin F has multiple antibacterial mechanisms against S. aureus. It can not only inhibit LTA biosynthesis through the interactions of its guanidyl side chain with the active center of LtaS but also disrupt the cell envelope through the synergistic effect of accelerating the LTA release, damaging the cell membrane, and electrostatically interacting with LTA. Simultaneously, these antibacterial mechanisms exhibit a synergistic inhibition effect on S. aureus cells, which would eventually cause the cellular autolysis.
Collapse
Affiliation(s)
- Zilong Luo
- Biotechnological Engineering Center for Pharmaceutical Research and Development, Jiangxi Agricultural University, Nanchang 330045, China
| | - Pingyi Li
- Biotechnological Engineering Center for Pharmaceutical Research and Development, Jiangxi Agricultural University, Nanchang 330045, China
| | - Duoduo Zhang
- Biotechnological Engineering Center for Pharmaceutical Research and Development, Jiangxi Agricultural University, Nanchang 330045, China
| | - Jianping Zhu
- Laboratory of Natural Medicine and Microbiological Drug, College of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Wan Wang
- School of Basic Medicine, Nanchang Medical College, Nanchang 330006, China
| | - Wenjia Zhao
- Biotechnological Engineering Center for Pharmaceutical Research and Development, Jiangxi Agricultural University, Nanchang 330045, China
| | - Peibo Li
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Ganjun Yuan
- Biotechnological Engineering Center for Pharmaceutical Research and Development, Jiangxi Agricultural University, Nanchang 330045, China
- Laboratory of Natural Medicine and Microbiological Drug, College of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
6
|
Pino-Hurtado MS, Fernández-Fernández R, Torres C, Robredo B. Searching for Antimicrobial-Producing Bacteria from Soils through an Educational Project and Their Evaluation as Potential Biocontrol Agents. Antibiotics (Basel) 2023; 13:29. [PMID: 38247588 PMCID: PMC10812812 DOI: 10.3390/antibiotics13010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/23/2024] Open
Abstract
Antimicrobial resistance (AMR) is a serious threat to public health due to the lack of effective drugs to combat infectious diseases, which generates the need to search for new antimicrobial substances. In this study, the potential of soil as a source of antimicrobial-producing bacteria (APB) was investigated and the importance of the connection between education and science was emphasized, using service-learning methodologies. Sixty-one soil samples were collected, and 1220 bacterial isolates were recovered. Eighteen of these isolates showed antimicrobial activity against at least 1 of the 12 indicator bacteria tested (including multidrug-resistant and relevant pathogens). The 18 APB were identified by MALDI-TOF and 6 different genera (Bacillus, Brevibacillus, Lysinobacillus, Peribacillus, Streptomyces, and Advenella) and 10 species were identified. The 18 APB were tested for antifungal activity against four phytopathogenic fungi (Botritis cynerea, Lecanicillium fungicola, Trichoderma harzianum, and Cladobotryum mycophilum). Moreover, the antibiotic susceptibility of APB was tested using the disk-diffusion method as well as their β-hemolytic activity (important safety criteria for potential future applications). A total of 10 of the 18 APB were able to inhibit at least 50% of indicator bacteria tested, including methicillin-resistant Staphylococcus aureus (MRSA), among others. A total of 4 of the 18 APB (3 Bacillus pumilus and 1 Bacillus altitudinis) showed inhibitory activity against two of the four fungal pathogens tested (B. cinerea and L. fungicola), as well as against 5-7 of the 12 bacterial pathogen indicators; these 4 isolates showed susceptibility to the antibiotics tested and lacked β-hemolytic activity and were considered promising APB for use as potential biocontrol agents. In addition, one Brevibacillus laterosporus strain had activity against 83% of indicator bacteria tested including Escherichia coli, MRSA and other methicillin-resistant staphylococci, as well as vancomycin-resistant enterococci (but not against fungi). These results show that soil is a source of APB with relevant antibacterial and antifungal activities, and also emphasize the importance of education and science to raise public awareness of the AMR problem and the strategies to control it.
Collapse
Affiliation(s)
- Mario Sergio Pino-Hurtado
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (M.S.P.-H.); (R.F.-F.); (C.T.)
| | - Rosa Fernández-Fernández
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (M.S.P.-H.); (R.F.-F.); (C.T.)
| | - Carmen Torres
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (M.S.P.-H.); (R.F.-F.); (C.T.)
| | - Beatriz Robredo
- Area of Didactic of Experimental Sciences, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain
| |
Collapse
|
7
|
Jeong GJ, Khan F, Tabassum N, Cho KJ, Kim YM. Controlling biofilm and virulence properties of Gram-positive bacteria by targeting wall teichoic acid and lipoteichoic acid. Int J Antimicrob Agents 2023; 62:106941. [PMID: 37536571 DOI: 10.1016/j.ijantimicag.2023.106941] [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: 04/04/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023]
Abstract
Wall teichoic acid (WTA) and lipoteichoic acid (LTA) are structural components of Gram-positive bacteria's peptidoglycan and cell membrane, which are mostly anionic glycopolymers. WTA confers numerous physiological, virulence, and pathogenic features to bacterial pathogens. It controls cell shape, cell division, and the localisation of autolytic enzymes and ion homeostasis. In the context of virulence and pathogenicity, it aids bacterial cell attachment and colonisation and protects against the host defence system and antibiotics. Having such a broad function in pathogenic bacteria's lifecycle, WTA/LTA become one of the potential targets for antibacterial agents to reduce bacterial infection in the host. The number of reports for targeting the WTA/LTA pathway has risen, mostly by focusing on three distinct targets: antivirulence targets, β-lactam potentiator targets, and essential targets. The current review looked at the role of WTA/LTA in biofilm development and virulence in a range of Gram-positive pathogenic bacteria. Furthermore, alternate strategies, such as the application of natural and synthetic compounds that target the WTA/LTA pathway, have been thoroughly discussed. Moreover, the application of nanomaterials and a combination of drugs have also been discussed as a viable method for targeting the WTA/LTA in numerous Gram-positive bacteria. In addition, a future perspective for controlling bacterial infection by targeting the WTA/LTA is proposed.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Kyung-Jin Cho
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
8
|
Haktaniyan M, Sharma R, Bradley M. Size-Controlled Ammonium-Based Homopolymers as Broad-Spectrum Antibacterials. Antibiotics (Basel) 2023; 12:1320. [PMID: 37627740 PMCID: PMC10452032 DOI: 10.3390/antibiotics12081320] [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: 07/31/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Ammonium group containing polymers possess inherent antimicrobial properties, effectively eliminating or preventing infections caused by harmful microorganisms. Here, homopolymers based on monomers containing ammonium groups were synthesized via Reversible Addition Fragmentation Chain Transfer Polymerization (RAFT) and evaluated as potential antibacterial agents. The antimicrobial activity was evaluated against Gram-positive (M. luteus and B. subtilis) and Gram-negative bacteria (E. coli and S. typhimurium). Three polymers, poly(diallyl dimethyl ammonium chloride), poly([2-(methacryloyloxy)ethyl]trimethylammonium chloride), and poly(vinyl benzyl trimethylammonium chloride), were examined to explore the effect of molecular weight (10 kDa, 20 kDa, and 40 kDa) on their antimicrobial activity and toxicity to mammalian cells. The mechanisms of action of the polymers were investigated with dye-based assays, while Scanning Electron Microscopy (SEM) showed collapsed and fused bacterial morphologies due to the interactions between the polymers and components of the bacterial cell envelope, with some polymers proving to be bactericidal and others bacteriostatic, while being non-hemolytic. Among all the homopolymers, the most active, non-Gram-specific polymer was poly([2-(methacryloyloxy)ethyl]trimethylammonium chloride), with a molecular weight of 40 kDa, with minimum inhibitory concentrations between 16 and 64 µg/mL, showing a bactericidal mode of action mediated by disruption of the cytoplasmic membrane. This homopolymer could be useful in biomedical applications such as surface dressings and in areas such as eye infections.
Collapse
Affiliation(s)
- Meltem Haktaniyan
- EaStCHEM, School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh EH9 3FJ, UK; (M.H.); (R.S.)
| | - Richa Sharma
- EaStCHEM, School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh EH9 3FJ, UK; (M.H.); (R.S.)
| | - Mark Bradley
- EaStCHEM, School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh EH9 3FJ, UK; (M.H.); (R.S.)
- Precision Healthcare University Research Institute, Queen Mary University of London, Whitechapel, Empire House, London E1 1HH, UK
| |
Collapse
|
9
|
Liu Y, Ning Y, Chen Z, Han P, Zhi T, Li S, Ma A, Jia Y. Transcriptomics reveals substance biosynthesis and transport on membranes of Listeria monocytogenes affected by antimicrobial lipopeptide brevilaterin B. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.10.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
10
|
Secondary metabolic profiling of Serratia marcescens NP10 reveals new stephensiolides and glucosamine derivatives with bacterial membrane activity. Sci Rep 2023; 13:2360. [PMID: 36759548 PMCID: PMC9911388 DOI: 10.1038/s41598-023-28502-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Secondary metabolic profiling, using UPLC-MSE and molecular networking, revealed the secondary metabolites produced by Serratia marcescens NP10. The NP10 strain co-produced cyclic and open-ring stephensiolides (i.e., fatty acyl chain linked to Thr-Ser-Ser-Ile/Leu-Ile/Leu/Val) and glucosamine derivatives (i.e., fatty acyl chain linked to Val-glucose-butyric/oxo-hexanoic acid), with the structures of sixteen new stephensiolides (L-Y) and three new glucosamine derivatives (L-N) proposed. Genome mining identified sphA (stephensiolides) and gcd (glucosamine derivatives) gene clusters within Serratia genomes available on NBCI using antiSMASH, revealing specificity scores of the adenylation-domains within each module that corroborates MSE data. Of the nine RP-HPLC fractions, two stephensiolides and two glucosamine derivatives exhibited activity against Staphylococcus aureus (IC50 of 25-79 µg/mL). 1H NMR analysis confirmed the structure of the four active compounds as stephensiolide K, a novel analogue stephensiolide U, and glucosamine derivatives A and C. Stephensiolides K and U were found to cause membrane depolarisation and affect the membrane permeability of S. aureus, while glucosamine derivatives A and C primarily caused membrane depolarisation. New members of the stephensiolide and glucosamine derivative families were thus identified, and results obtained shed light on their antibacterial properties and mode of membrane activity.
Collapse
|
11
|
Zhao W, Yang C, Zhang N, Peng Y, Ma Y, Gu K, Liu X, Liu X, Liu X, Liu Y, Li S, Zhao L. Menthone Exerts its Antimicrobial Activity Against Methicillin Resistant Staphylococcus aureus by Affecting Cell Membrane Properties and Lipid Profile. Drug Des Devel Ther 2023; 17:219-236. [PMID: 36721663 PMCID: PMC9884481 DOI: 10.2147/dddt.s384716] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/16/2022] [Indexed: 01/26/2023] Open
Abstract
Objective The characteristic constituents of essential oils from aromatic plants have been widely applied as antimicrobial agents in the last decades. However, their mechanisms of action remain obscure, especially from the metabolic perspective. The aim of the study was to explore the antimicrobial effect and mechanism of menthone, a main component of peppermint oil, against methicillin resistant Staphylococcus aureus (MRSA). Methods An integrated approach including the microbiology and the high-coverage lipidomics was applied. The changes of membrane properties were studies by the fluorescence and electron microscopical observations. The lipid profile was analyzed by ultra-high performance liquid chromatography coupled with quadruple Exactive mass spectrometry (UHPLC-QE-MS). The lipid-related key targets which were associated with the inhibitory effect of menthone against MRSA, were studied by network analysis and molecular docking. Results Menthone exhibited antibacterial activities against MRSA, with minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) of 3,540 and 7,080 μg/mL, respectively. The membrane potential and membrane integrity upon menthone treatment were observed to change strikingly. Further, lipids fingerprinting identified 136 significantly differential lipid species in MRSA cells exposed to menthone at subinhibitory level of 0.1× MIC. These metabolites span 30 important lipid classes belonging to glycerophospholipids, glycolipids, and sphingolipids. Lastly, the correlations of these altered lipids, as well as the potential metabolic pathways and targets associated with menthone treatment were deciphered preliminarily. Conclusion Menthone had potent antibacterial effect on MRSA, and the mechanism of action involved the alteration of membrane structural components and corresponding properties. The interactions of identified key lipid species and their biological functions need to be further determined and verified, for the development of novel antimicrobial strategies against MRSA.
Collapse
Affiliation(s)
- Wenming Zhao
- Department of Spinal Surgery, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, People’s Republic of China,Department of Orthopedics, Zhangye Second People’s Hospital, Zhangye, People’s Republic of China
| | - Chengwei Yang
- Department of Spinal Surgery, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, People’s Republic of China
| | - Ning Zhang
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People’s Republic of China
| | - Yuanyuan Peng
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People’s Republic of China
| | - Ying Ma
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People’s Republic of China
| | - Keru Gu
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People’s Republic of China
| | - Xia Liu
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People’s Republic of China
| | - Xiaohui Liu
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People’s Republic of China
| | - Xijian Liu
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People’s Republic of China
| | - Yumin Liu
- Instrumental Analysis Centre, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Songkai Li
- Department of Spinal Surgery, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, People’s Republic of China,Songkai Li, Department of Spinal Surgery, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, People’s Republic of China, Email
| | - Linjing Zhao
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People’s Republic of China,Correspondence: Linjing Zhao, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People’s Republic of China, Email
| |
Collapse
|
12
|
Clements-Decker T, Kode M, Khan S, Khan W. Underexplored bacteria as reservoirs of novel antimicrobial lipopeptides. Front Chem 2022; 10:1025979. [PMID: 36277345 PMCID: PMC9581180 DOI: 10.3389/fchem.2022.1025979] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Natural products derived from microorganisms play a prominent role in drug discovery as potential anti-infective agents. Over the past few decades, lipopeptides produced by particularly Bacillus, Pseudomonas, Streptomyces, Paenibacillus, and cyanobacteria species, have been extensively studied for their antimicrobial potential. Subsequently, daptomycin and polymyxin B were approved by the Food and Drug Administration as lipopeptide antibiotics. Recent studies have however, indicated that Serratia, Brevibacillus, and Burkholderia, as well as predatory bacteria such as Myxococcus, Lysobacter, and Cystobacter, hold promise as relatively underexplored sources of novel classes of lipopeptides. This review will thus highlight the structures and the newly discovered scaffolds of lipopeptide families produced by these bacterial genera, with potential antimicrobial activities. Additionally, insight into the mode of action and biosynthesis of these lipopeptides will be provided and the application of a genome mining approach, to ascertain the biosynthetic gene cluster potential of these bacterial genera (genomes available on the National Center for Biotechnology Information) for their future pharmaceutical exploitation, will be discussed.
Collapse
Affiliation(s)
| | - Megan Kode
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Sehaam Khan
- Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| | - Wesaal Khan
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- *Correspondence: Wesaal Khan,
| |
Collapse
|
13
|
Lan W, Chen X, Zhao Y, Xie J. Insight into the Antibacterial Mechanism of Ozone water Combined with Tea Polyphenols against
Shewanella putrefaciens
: Membrane Disruption and Oxidative Stress. Int J Food Sci Technol 2022. [DOI: 10.1111/ijfs.16106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Weiqing Lan
- College of Food Science and Technology Shanghai Ocean University Shanghai 201306 China
- Shanghai Aquatic Products Processing and Storage Engineering Technology Research Center Shanghai 201306 China
- National Experimental Teaching Demonstration Center for Food Science and Engineering (Shanghai Ocean University) Shanghai 201306 China
| | - Xuening Chen
- College of Food Science and Technology Shanghai Ocean University Shanghai 201306 China
| | - Yanan Zhao
- College of Food Science and Technology Shanghai Ocean University Shanghai 201306 China
| | - Jing Xie
- College of Food Science and Technology Shanghai Ocean University Shanghai 201306 China
- Shanghai Aquatic Products Processing and Storage Engineering Technology Research Center Shanghai 201306 China
- National Experimental Teaching Demonstration Center for Food Science and Engineering (Shanghai Ocean University) Shanghai 201306 China
| |
Collapse
|
14
|
Liang Q, Cao L, Zhu C, Kong Q, Sun H, Zhang F, Mou H, Liu Z. Characterization of Recombinant Antimicrobial Peptide BMGlv2 Heterologously Expressed in Trichoderma reesei. Int J Mol Sci 2022; 23:ijms231810291. [PMID: 36142214 PMCID: PMC9499586 DOI: 10.3390/ijms231810291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/03/2022] [Accepted: 09/04/2022] [Indexed: 11/16/2022] Open
Abstract
Antimicrobial peptides (AMPs) serve as alternative candidates for antibiotics and have attracted the attention of a wide range of industries for various purposes, including the prevention and treatment of piglet diarrhea in the swine industry. Escherichia coli, Salmonella, and Clostridium perfringens are the most common pathogens causing piglet diarrhea. In this study, the antimicrobial peptide gloverin2 (BMGlv2), derived from Bombyx mandarina, was explored to determine the efficient prevention effect on bacterial piglet diarrhea. BMGlv2 was heterologously expressed in Trichoderma reesei Tu6, and its antimicrobial properties against the three bacteria were characterized. The results showed that the minimum inhibitory concentrations of the peptide against E. coli ATCC 25922, S. derby ATCC 13076, and C. perfringens CVCC 2032 were 43.75, 43.75, and 21.86 μg/mL, respectively. The antimicrobial activity of BMGlv2 was not severely affected by high temperature, salt ions, and digestive enzymes. It had low hemolytic activity against rabbit red blood cells, indicating its safety for use as a feed additive. Furthermore, the measurements of the leakage of bacterial cell contents and scanning electron microscopy of C. perfringens CVCC 2032 indicated that BMGlv2 exerted antimicrobial activity by destroying the cell membrane. Overall, this study showed the heterologous expression of the antimicrobial peptide BMGlv2 in T. reesei and verified its antimicrobial properties against three common pathogenic bacteria associated with piglet diarrhea, which can provide a reference for the applications of AMPs as an alternative product in industrial agriculture.
Collapse
|
15
|
Chen Z, Wang L, Hong D, Liu Y, Han P, Li S, Jia Y. Broad-spectrum cytotoxicity to cancer cells of Brevilaterin C from Brevibacillus laterosporus and its specific mechanism on human epidermal cancer cells. J Cell Biochem 2022; 123:1237-1246. [PMID: 35656936 DOI: 10.1002/jcb.30280] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/18/2022] [Accepted: 05/12/2022] [Indexed: 12/29/2022]
Abstract
Antimicrobial peptides (AMP) from Brevibacillus laterosporus have good prospects as clinical treatments for cancer. Nevertheless, details about their anticancer spectrum and mode of cytotoxicity remain poorly understood. A newly found AMP (named Brevilaterin C) secreted by B. laterosporus S62-9 exhibited strong inhibition on almost cancer cell lines examined at a concentration of 8 µg/ml but was relatively safe for normal cells. We further systematically examined its cytotoxicity and mechanism toward human epidermal cancer cell A431. A dosage of 3 µg/ml of Brevilaterin C could significantly increase lactate dehydrogenase release of tumor cells. Moreover, it could remarkably increase the ratio of apoptosis and reactive oxygen species generation of A431, indicating effective induction of apoptosis. Moreover, the formation of JC-1 aggregates was effectively prevented by a low concentration of Brevilaterin C, indicating its effective induction of A431's apoptosis. Brevilaterin C exhibited broad-spectrum cytotoxicity to cancer cells, indicating a good potential prospect in the medical field.
Collapse
Affiliation(s)
- Zhou Chen
- Lab of Enzyme Engineering, School of Food and Health, Beijing Technology and Business University, Beijing, Haidian District, China
| | - Lulu Wang
- Lab of Enzyme Engineering, School of Food and Health, Beijing Technology and Business University, Beijing, Haidian District, China
| | - Dan Hong
- Lab of Enzyme Engineering, School of Food and Health, Beijing Technology and Business University, Beijing, Haidian District, China
| | - Yangliu Liu
- Lab of Enzyme Engineering, School of Food and Health, Beijing Technology and Business University, Beijing, Haidian District, China
| | - Panpan Han
- Lab of Enzyme Engineering, School of Food and Health, Beijing Technology and Business University, Beijing, Haidian District, China
| | - Siting Li
- Lab of Enzyme Engineering, School of Food and Health, Beijing Technology and Business University, Beijing, Haidian District, China
| | - Yingmin Jia
- Lab of Enzyme Engineering, School of Food and Health, Beijing Technology and Business University, Beijing, Haidian District, China
| |
Collapse
|
16
|
NISIN and gilaburu (Viburnum opulus L.) combination is a cost-effective way to control foodborne Staphylococcus aureus. Food Control 2022. [DOI: 10.1016/j.foodcont.2022.109213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
Chang Y, Bai J, Yu H, Yang X, Chang PS, Nitin N. Synergistic inactivation of Listeria and E. coli using a combination of erythorbyl laurate and mild heating and its application in decontamination of peas as a model fresh produce. Food Microbiol 2022; 102:103869. [PMID: 34809926 DOI: 10.1016/j.fm.2021.103869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 11/04/2022]
Abstract
We investigated the synergistic antimicrobial activity of erythorbyl laurate (EL) and mild heating co-treatment on the Gram-positive Listeria innocua and Gram-negative Escherichia coli O157:H7 bacteria. EL (2 mM) and mild heating (55 °C for 3 min) resulted in 3.1 and 0.5 log colony forming units (CFU)/mL reductions in the number of L. innocua, respectively, compared to a 6.4 log CFU/mL reduction induced by the combined treatment of EL and mild heating in saline. EL (10 mM) and mild heating (55 °C for 3 min) resulted in 1.3 and 0.7 log CFU/mL reductions in the number of E. coli O157:H7, respectively, compared to a 6.2 log CFU/mL reduction with the combined treatment in saline. EL, a membrane-active compound, showed a strong synergistic effect with mild heating, possibly due to enhanced disruption of the bacterial cell membrane. The synergistic antibacterial effect was evaluated using inoculated English peas (Pisum sativum) and this combined treatment (2 mM EL and mild heating against L. innocua and 10 mM EL and mild heating against E. coli O157:H7) resulted in more than 7 log reductions in the numbers of L. innocua and E. coli O157:H7, inoculated on the surface of fresh peas. The treatments did not show significant difference in the color or texture of treated peas compared to the non-treated controls. This is the first report illustrating synergistic activity of EL and mild heating for both the gram positive (L. innocua) and the gram negative (E. coli O157:H7) bacteria on food. Overall, this research will illustrate the development of more effective and rapid antibacterial surface disinfection method for application in the processing of minimally processed foods.
Collapse
Affiliation(s)
- Yoonjee Chang
- Department of Food and Nutrition, Kookmin University, Seoul, 02707, Republic of Korea; Department of Food Science and Technology, University of California, Davis, CA, USA
| | - Jaewoo Bai
- Department of Food Science and Technology, University of California, Davis, CA, USA; Division of Applied Food System, Major in Food Science & Technology, Seoul Women's University, Seoul, 01797, Republic of Korea
| | - Hyunjong Yu
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Xu Yang
- Department of Food Science and Technology, University of California, Davis, CA, USA
| | - Pahn-Shick Chang
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea; Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea; Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Nitin Nitin
- Department of Food Science and Technology, University of California, Davis, CA, USA; Department of Biological and Agricultural Engineering, University of California, Davis, CA, USA.
| |
Collapse
|
18
|
Ning Y, Ma M, Zhang Y, Zhang D, Hou L, Yang K, Fu Y, Wang Z, Jia Y. Antibacterial mechanism of sucrose laurate against Bacillus cereus by attacking multiple targets and its application in milk beverage. Food Res Int 2022; 154:111018. [DOI: 10.1016/j.foodres.2022.111018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 11/04/2022]
|
19
|
Ning Y, Hou L, Ma M, Li M, Zhao Z, Zhang D, Wang Z, Jia Y. Synergistic antibacterial mechanism of sucrose laurate combined with nisin against Staphylococcus aureus and its application in milk beverage. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113145] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Chen Y, Liu L, Wang X, Liao Z, Wang R, Xiong YS, Chen J, jiang G, Wang J, Liao X. Synthesis and antibacterial activity study of ruthenium-based metallodrugs with membrane-disruptive mechanism against Staphylococcus aureus. Dalton Trans 2022; 51:14980-14992. [DOI: 10.1039/d2dt01531e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The wide spread of drug-resistant bacteria, especially methicillin-resistant Staphylococcus aureus (MRSA), have posed a tremendous threat to global health. Of particular concern, resistance to vancomycin, linezolid and daptomycin have already...
Collapse
|
21
|
Daison FA, Kumar N, Balakrishnan S, Venugopal K, Elango S, Sokkar P. Molecular Dynamics Studies on the Bacterial Membrane Pore Formation by Small Molecule Antimicrobial Agents. J Chem Inf Model 2021; 62:40-48. [PMID: 34932333 DOI: 10.1021/acs.jcim.1c01049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antimicrobial peptides (AMPs) act on the membrane bilayer of pathogens, causing leakage in the membrane and cell death. Amphiphilic kaempferol derivatives possessing basic functional groups show excellent antibacterial activities, which has been proven through experimental techniques. These compounds are known to target negatively charged bacterial membranes. However, the detailed mechanism of action and their structure-activity relationship are not clear. In this work, we reported theoretical investigation on the mechanism of action of two previously reported kaempferol derivatives on a DMPC/DMPG mixed bilayer. Despite the rigid structure of the compounds when compared to AMPs, spontaneous pore formation in the membrane was not observed in 400 ns molecular dynamics (MD) simulations. MD simulations with biasing forces resulted in the formation of pores in the bilayer for the derivatives and not for kaempferol. The stability of the pores was assessed by pore closure timescales in unbiased MD simulations, which was found to be 5.3 and 17.0 ns for 2 and 3, respectively. Free energy change for the permeation into the bilayer for kaempferol (1), tertiary amine derivative (2), and arginine derivative (3) was calculated to be -1.5, -48.2, and -100.3 kJ/mol, respectively, which correlate with their antibacterial activity. Furthermore, our results indicate that compound 3 forms a stable toroidal pore in the membrane when multiple molecules are oriented in a transmembrane configuration. Our work sheds light on the mechanism of action of small molecule antimicrobial agents, which can be exploited for the rational design of drug candidates.
Collapse
Affiliation(s)
- Felsis Angelene Daison
- Department of Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, India
| | - Nitheeshkumar Kumar
- Department of Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, India
| | - Siranjeevi Balakrishnan
- Department of Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, India
| | - Kavyashree Venugopal
- Department of Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, India
| | - Sangamithra Elango
- Department of Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, India
| | - Pandian Sokkar
- Department of Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, India
| |
Collapse
|
22
|
Shan MY, Meng FQ, Zhou LB, Lu FX, Bie XM, Zhao HZ, Lu ZX. Surfactin inhibits the growth of Propionibacterium acnes by destroying the cell wall and membrane. Lett Appl Microbiol 2021; 73:684-693. [PMID: 34607389 DOI: 10.1111/lam.13576] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/07/2021] [Accepted: 09/25/2021] [Indexed: 01/08/2023]
Abstract
Propionibacterium acnes plays a major role in acne vulgaris. In the pre-experiment, the growth of P. acnes was inhibited effectively using surfactin; however, the antibacterial mechanism has not been described. Therefore, the aim of this study was to evaluate antibacterial activity and analyse the mechanism of surfactin against P. acnes. Minimum inhibitory concentration, time-killing kinetics and scanning electron microscopy were used to evaluate the activity of surfactin against P. acnes, which showed that 128 μg ml-1 effectively inhibited growth. Cell wall permeability was evaluated by detecting the extracellular alkaline phosphatase activity, which increased to 1·83- and 2·32-fold after incubating with 128 and 256 μg ml-1 of surfactin for 10 h, respectively. Propidium iodide fluorescence, leakage of nucleic acid, protein, K+ , and Ca2+ , membrane potential and the leakage of calcein from small unilamellar vesicles all increased after incubation with surfactin, indicating that its strong biological activities act mainly by altering membrane integrity. In a mouse model of acne, surfactin significantly reduced P. acnes-induced epidermal swelling and erythema. These results indicate that surfactin effectively inhibited the growth of P. acnes by destroying the cell wall and membrane, and is a potential candidate for acne treatment.
Collapse
Affiliation(s)
- M Y Shan
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - F Q Meng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - L B Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - F X Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - X M Bie
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - H Z Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Z X Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
23
|
Zhou L, de Jong A, Yi Y, Kuipers OP. Identification, Isolation, and Characterization of Medipeptins, Antimicrobial Peptides From Pseudomonas mediterranea EDOX. Front Microbiol 2021; 12:732771. [PMID: 34594316 PMCID: PMC8477016 DOI: 10.3389/fmicb.2021.732771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/20/2021] [Indexed: 11/22/2022] Open
Abstract
The plant microbiome is a vastly underutilized resource for identifying new genes and bioactive compounds. Here, we used Pseudomonas sp. EDOX, isolated from the leaf endosphere of a tomato plant grown on a small farm in the Netherlands. To get more insight into its biosynthetic potential, the genome of Pseudomonas sp. EDOX was sequenced and subjected to bioinformatic analyses. The genome sequencing analysis identified strain EDOX as a member of the Pseudomonas mediterranea. In silico analysis for secondary metabolites identified a total of five non-ribosomally synthesized peptides synthetase (NRPS) gene clusters, related to the biosynthesis of syringomycin, syringopeptin, anikasin, crochelin A, and fragin. Subsequently, we purified and characterized several cyclic lipopeptides (CLPs) produced by NRPS, including some of the already known ones, which have biological activity against several plant and human pathogens. Most notably, mass spectrometric analysis led to the discovery of two yet unknown CLPs, designated medipeptins, consisting of a 22 amino acid peptide moiety with varying degrees of activity against Gram-positive and Gram-negative pathogens. Furthermore, we investigated the mode of action of medipeptin A. The results show that medipeptin A acts as a bactericidal antibiotic against Gram-positive pathogens, but as a bacteriostatic antibiotic against Gram-negative pathogens. Medipeptin A exerts its potent antimicrobial activity against Gram-positive bacteria via binding to both lipoteichoic acid (LTA) and lipid II as well as by forming pores in membranes. Collectively, our study provides important insights into the biosynthesis and mode of action of these novel medipeptins from P. mediterranea EDOX.
Collapse
Affiliation(s)
| | | | | | - Oscar P. Kuipers
- Department of Molecular Genetics, University of Groningen, Groningen, Netherlands
| |
Collapse
|
24
|
Ning Y, Han P, Ma J, Liu Y, Fu Y, Wang Z, Jia Y. Characterization of brevilaterins, multiple antimicrobial peptides simultaneously produced by Brevibacillus laterosporus S62-9, and their application in real food system. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Zhao X, Wang X, Shukla R, Kumar R, Weingarth M, Breukink E, Kuipers OP. Brevibacillin 2V Exerts Its Bactericidal Activity via Binding to Lipid II and Permeabilizing Cellular Membranes. Front Microbiol 2021; 12:694847. [PMID: 34335524 PMCID: PMC8322648 DOI: 10.3389/fmicb.2021.694847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/28/2021] [Indexed: 11/14/2022] Open
Abstract
Lipo-tridecapeptides, a class of bacterial non-ribosomally produced peptides, show strong antimicrobial activity against Gram-positive pathogens, including antibiotic-resistant Staphylococcus aureus and Enterococcus spp. However, many of these lipo-tridecapeptides have shown high hemolytic activity and cytotoxicity, which has limited their potential to be developed into antibiotics. Recently, we reported a novel antimicrobial lipo-tridecapeptide, brevibacillin 2V, which showed no hemolytic activity against human red blood cells at a high concentration of 128 mg/L, opposite to other brevibacillins and lipo-tridecapeptides. In addition, brevibacillin 2V showed much lower cytotoxicity than the other members of the brevibacillin family. In this study, we set out to elucidate the antimicrobial mode of action of brevibacillin 2V. The results show that brevibacillin 2V acts as bactericidal antimicrobial agent against S. aureus (MRSA). Further studies show that brevibacillin 2V exerts its bactericidal activity by binding to the bacterial cell wall synthesis precursor Lipid II and permeabilizing the bacterial membrane. Combined solid-state NMR, circular dichroism, and isothermal titration calorimetry assays indicate that brevibacillin 2V binds to the GlcNAc-MurNAc moiety and/or the pentapeptide of Lipid II. This study provides an insight into the antimicrobial mode of action of brevibacillin 2V. As brevibacillin 2V is a novel and promising antibiotic candidate with low hemolytic activity and cytotoxicity, the here-elucidated mode of action will help further studies to develop it as an alternative antimicrobial agent.
Collapse
Affiliation(s)
- Xinghong Zhao
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| | - Xiaoqi Wang
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Rhythm Shukla
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands.,NMR Spectroscopy, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Raj Kumar
- NMR Spectroscopy, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Markus Weingarth
- NMR Spectroscopy, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Eefjan Breukink
- Membrane Biochemistry and Biophysics, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Oscar P Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, Netherlands
| |
Collapse
|
26
|
Wu Y, Nie T, Meng F, Zhou L, Chen M, Sun J, Lu Z, Lu Y. The determination of antibacterial mode for cationic lipopeptides brevibacillins against Salmonella typhimurium by quantum chemistry calculation. Appl Microbiol Biotechnol 2021; 105:5643-5655. [PMID: 34160646 DOI: 10.1007/s00253-021-11398-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/16/2021] [Accepted: 05/24/2021] [Indexed: 01/25/2023]
Abstract
Brevibacillins are broad-spectrum cationic antimicrobial lipopeptides produced by Brevibacillus laterosporus fmb70 CGMCC 18426. The antibacterial mode of brevibacillins against Salmonella typhimurium CICC 21493 was investigated by quantum chemistry calculation in this study. The addition of LPS, Mg2+, and Ca2+ partially reduced the antimicrobial activity of brevibacillin and brevibacillin V against S. typhimurium, which indicated that the two cationic lipopeptides could bind to LPS and displaced the divalent cations on the LPS network. Release of LPS from S. typhimurium by brevibacillin and brevibacillin V resulted in destroying the dense LPS network and increasing the permeability of the outer membrane. Quantum chemistry calculation analysis revealed that Lys7 is the most critical amino acid residue to destroy the outer membrane. The total average N-H charge difference of the three protonated amino groups (Orn3-NH3, Lys7-NH3, and Lys10-NH3) determined the ability of brevibacillin V to bind LPS stronger than brevibacillin. Calcein complete leakage from liposomes and release of DiSC3-5 from the cytoplasmic membrane (CM) indicated that brevibacillin and brevibacillin V may destroy the CM. Brevibacillin and brevibacillin V exhibited their antimicrobial activities through membrane damages, where the OM permeability with high concentration of 64-256 µg/mL and membrane damage of CM with a low concentration of 4 μg/mL. Our finding might be helpful to understand the broad-spectrum antimicrobial mechanism of cationic lipopeptide and to design the novel antimicrobial peptide. KEY POINTS: • Brevibacillin V had stronger affinity for LPS than brevibacillin. • The N-H charge difference was the key of the difference in the affinity to LPS. • Brevibacillins inhibited Salmonella by displacing the divalent cations on the LPS.
Collapse
Affiliation(s)
- Yubo Wu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Ting Nie
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Fanqiang Meng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Libang Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Meirong Chen
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Jing Sun
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, 210023, Jiangsu Province, China
| | - Zhaoxin Lu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, 210023, Jiangsu Province, China.
| |
Collapse
|
27
|
Xia T, Xie F, Bian X, Chen Z, Zhang S, Fang Z, Ye Q, Cai J, Wang Y. Ultrabroad-spectrum, multidrug resistant bacteria-killing, and biocompatible quaternized chitin derivative for infected wound healing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112177. [PMID: 34082977 DOI: 10.1016/j.msec.2021.112177] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 11/17/2022]
Abstract
Wound infections have consistently been recognized as serious threats to human. The design of antimicrobial and biocompatible wound dressings for infected wounds is an area of constant research. Herein, we homogeneously synthesized an ultrabroad-spectrum antimicrobial and biocompatible quaternized chitin derivative (QC-4) in a high-efficiency and sustainable route using aqueous KOH/urea solution. Particularly, QC-4 displayed powerful multidrug resistant bacteria-killing activities even at a very low antimicrobial concentration range from 500 ng/mL to 5 μg/mL, including clinically prevalent multidrug-resistant Escherichia coli (MDR-E. coli), methicillin resistant Staphylococcus aureus (MRSA), multidrug-resistant Pseudomonas aeruginosa (MRPA), and multidrug-resistant Acinetobacter baumannii (MDR-A. baumannii). With the aim to facilitate clinical translation, we validated the biocompatibility and safety of QC-4 both in vitro and in vivo, and further assessed the effects of QC-4 on infected wound healing in a porcine infectious full-thickness skin wound model. QC-4 demonstrated significant reduction of microbial aggregates and enhanced wound-healing effects by promoted re-epithelialization and collagen deposition, which were quite comparable to that of commercial Alginate-Ag dressing and absolutely superior to commercial Chitoclot Bandage dressing. Additionally, we provided clear evidences that QC-4 had a unique mechanism of action by attracting electrostatically to the negatively charged microbial surface, thus damaging the microbial cell wall and membrane. Findings of this work provided robust preclinical rationale for the future translational applications of QC-4 as a novel ultrabroad-spectrum and multidrug resistant bacteria-killing antimicrobial wound dressing for clinical wound management.
Collapse
Affiliation(s)
- Tian Xia
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan 430072, China
| | - Fang Xie
- Hubei Engineering Center of Natural Polymers-based Medical Materials, College of Chemistry & Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Xiaoen Bian
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan 430072, China
| | - Zuhan Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan 430072, China
| | - Shichen Zhang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan 430072, China
| | - Zehong Fang
- Jiangxi Provincial People's Hospital of Nanchang University, Department of General Surgery, Nanchang 330006, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan 430072, China
| | - Jie Cai
- Hubei Engineering Center of Natural Polymers-based Medical Materials, College of Chemistry & Molecular Sciences, Wuhan University, Wuhan 430072, China; Research Institute of Shenzhen, Wuhan University, Shenzhen 518057, China.
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Wuhan 430072, China; Hubei Engineering Center of Natural Polymers-based Medical Materials, College of Chemistry & Molecular Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
28
|
Novel Modifications of Nonribosomal Peptides from Brevibacillus laterosporus MG64 and Investigation of Their Mode of Action. Appl Environ Microbiol 2020; 86:AEM.01981-20. [PMID: 32978140 DOI: 10.1128/aem.01981-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Nonribosomal peptides (NRPs) are a class of secondary metabolites usually produced by microorganisms. They are of paramount importance in different applications, including biocontrol and pharmacy. Brevibacillus spp. are a rich source of NRPs yet have received little attention. In this study, we characterize four novel bogorol variants (bogorols I to L, cationic linear lipopeptides) and four succilins (succilins I to L, containing a succinyl group that is attached to the Orn3/Lys3 in bogorols I to L) from the biocontrol strain Brevibacillus laterosporus MG64. Further investigation revealed that the bogorol family of peptides employs an adenylation pathway for lipoinitiation, different from the usual pattern, which is based on an external ligase and coenzyme A. Moreover, the formation of valinol was proven to be mediated by a terminal reductase domain and a reductase encoded by the bogI gene. Furthermore, succinylation, which is a novel type of modification in the family of bogorols, was discovered. Its occurrence requires a high concentration of the substrate (bogorols), but its responsible enzyme remains unknown. Bogorols display potent activity against both Gram-positive and Gram-negative bacteria. Investigation of their mode of action reveals that bogorols form pores in the cell membrane of both Gram-positive and Gram-negative bacteria. The combination of bogorols and relacidines, another class of NRPs produced by B. laterosporus MG64, displays a synergistic effect on different pathogens, suggesting the great potential of both peptides as well as their producer B. laterosporus MG64 for broad applications. Our study provides a further understanding of the bogorol family of peptides as well as their applications.IMPORTANCE NRPs form a class of secondary metabolites with biocontrol and pharmaceutical potential. This work describes the identification of novel bogorol variants and succinylated bogorols (namely, succilins) and further investigates their biosynthetic pathway and mode of action. Adenylation domain-mediated lipoinitiation of bogorols represents a novel pathway by which NRPs incorporate fatty acid tails. This pathway provides the possibility to engineer the lipid tail of NRPs without identifying a fatty acid coenzyme ligase, which is usually not present in the biosynthetic gene cluster. The terminal reductase domain (TD) and BogI-mediated valinol formation and their effect on the biological activity of bogorols are revealed. Succinylation, which is rarely reported in NRPs, was discovered in the bogorol family of peptides. We demonstrate that bogorols combat bacterial pathogens by forming pores in the cell membrane. We also report the synergistic effect of two natural products (relacidine B and bogorol K) produced by the same strain, which is relevant for competition for a niche.
Collapse
|
29
|
Antibacterial mechanism of brevilaterin B: an amphiphilic lipopeptide targeting the membrane of Listeria monocytogenes. Appl Microbiol Biotechnol 2020; 104:10531-10539. [PMID: 33170327 DOI: 10.1007/s00253-020-10993-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/20/2020] [Accepted: 10/31/2020] [Indexed: 12/14/2022]
Abstract
Antimicrobial peptides (AMPs) are recognized as promising safe alternatives to antibiotics for its low drug-resistance. Brevilaterin B, a newly discovered antimicrobial lipopeptide produced by Brevibacillus laterosporus S62-9, exhibits efficient antibacterial activity on Listeria monocytogenes with a minimum inhibitory concentration of 1 μg mL-1. The present research aimed to investigate the antibacterial mechanism of brevilaterin B against Listeria monocytogenes. Brevilaterin B caused membrane depolarization and the breakup of the cytomembrane as measured by 3,3-dipropylthiadicarbocyanine iodide and transmission electron microscopy, respectively. Using 1,2-dipalmitoyl-sn-glycero-3-phosphocholine and 1,2-dipalmitoyl-sn-glycero-3-phospho-rac-(1-glycerol) sodium salt (7:3) as a model membrane, results proved that brevilaterin B could bind to liposomes, integrate into the lipid bilayer, and consequently increase the permeability of liposomes to calcein. The secondary structure of brevilaterin B also changed from an unstructured coil to a mainly β-sheet conformation as measured by circular dichroism. Brevilaterin B exhibits antibacterial activity by a membrane interaction mechanism, which provides a theoretical basis for using brevilaterin B as a promising natural and effective antimicrobial agent against pathogenic bacteria. KEY POINTS: • Brevilaterin B exhibited antibacterial activity against Listeria monocytogenes. • Brevilaterin B exhibited membrane interaction mechanism. • Brevilaterin B showed conformational change when interacted with liposome.
Collapse
|
30
|
Ning Y, Fu Y, Hou L, Ma M, Wang Z, Li X, Jia Y. iTRAQ-based quantitative proteomic analysis of synergistic antibacterial mechanism of phenyllactic acid and lactic acid against Bacillus cereus. Food Res Int 2020; 139:109562. [PMID: 33509445 DOI: 10.1016/j.foodres.2020.109562] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/14/2020] [Accepted: 07/17/2020] [Indexed: 11/15/2022]
Abstract
Phenyllactic acid (PLA) as a phenolic acid by lactic acid (LA) bacteria shows enhanced antibacterial activity when coexisting with LA, while the antibacterial mechanism of PLA combined with LA was unknown. Hence, the antibacterial mechanism of PLA and LA was investigated against Bacillus cereus. Flow cytometry and TEM analysis demonstrated that single PLA and LA disrupted the membrane integrity and the morphology, while combined PLA and LA synergistically enhanced the damage. iTRAQ-based proteomic analysis suggested that PLA down-regulated kdpB and inhibited K+ transport, disturbed the function of ribosome and expression of competence genes; LA down-regulated periplasmic phosphorus-binding proteins and inhibited phosphorus transport, disturbed the function of ribosome, TCA cycle, as well as purine and pyrimidine metabolism; and combined PLA and LA inhibited K+ and phosphorus transport, and influenced the synthesis of purine and pyrimidine metabolism. The investigation could provide some insights into the application of PLA in food preservation.
Collapse
Affiliation(s)
- Yawei Ning
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Yunan Fu
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Linlin Hou
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Mengge Ma
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Zhixin Wang
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Xingfeng Li
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China.
| | - Yingmin Jia
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
31
|
Schäfer AB, Wenzel M. A How-To Guide for Mode of Action Analysis of Antimicrobial Peptides. Front Cell Infect Microbiol 2020; 10:540898. [PMID: 33194788 PMCID: PMC7604286 DOI: 10.3389/fcimb.2020.540898] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial peptides (AMPs) are a promising alternative to classical antibiotics in the fight against multi-resistant bacteria. They are produced by organisms from all domains of life and constitute a nearly universal defense mechanism against infectious agents. No drug can be approved without information about its mechanism of action. In order to use them in a clinical setting, it is pivotal to understand how AMPs work. While many pore-forming AMPs are well-characterized in model membrane systems, non-pore-forming peptides are often poorly understood. Moreover, there is evidence that pore formation may not happen or not play a role in vivo. It is therefore imperative to study how AMPs interact with their targets in vivo and consequently kill microorganisms. This has been difficult in the past, since established methods did not provide much mechanistic detail. Especially, methods to study membrane-active compounds have been scarce. Recent advances, in particular in microscopy technology and cell biological labeling techniques, now allow studying mechanisms of AMPs in unprecedented detail. This review gives an overview of available in vivo methods to investigate the antibacterial mechanisms of AMPs. In addition to classical mode of action classification assays, we discuss global profiling techniques, such as genomic and proteomic approaches, as well as bacterial cytological profiling and other cell biological assays. We cover approaches to determine the effects of AMPs on cell morphology, outer membrane, cell wall, and inner membrane properties, cellular macromolecules, and protein targets. We particularly expand on methods to examine cytoplasmic membrane parameters, such as composition, thickness, organization, fluidity, potential, and the functionality of membrane-associated processes. This review aims to provide a guide for researchers, who seek a broad overview of the available methodology to study the mechanisms of AMPs in living bacteria.
Collapse
Affiliation(s)
| | - Michaela Wenzel
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
32
|
Famuyide IM, Fasina FO, Eloff JN, McGaw LJ. The ultrastructural damage caused by Eugenia zeyheri and Syzygium legatii acetone leaf extracts on pathogenic Escherichia coli. BMC Vet Res 2020; 16:326. [PMID: 32887606 PMCID: PMC7472583 DOI: 10.1186/s12917-020-02547-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 08/27/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Antibiotics are commonly added to livestock feeds in sub-therapeutic doses as growth promoters and for prophylaxis against pathogenic microbes, especially those implicated in diarrhoea. While this practice has improved livestock production, it is a major cause of antimicrobial resistance in microbes affecting livestock and humans. This has led to the banning of prophylactic antibiotic use in animals in many countries. To compensate for this, alternatives have been sought from natural sources such as plants. While many studies have reported the antimicrobial activity of medicinal plants with potential for use as phytogenic/botanical feed additives, little information exists on their mode of action. This study is based on our earlier work and describes ultrastructural damage induced by acetone crude leaf extracts of Syzygium legatii and Eugenia zeyheri (Myrtaceae) active against diarrhoeagenic E. coli of swine origin using scanning electron microscopy (SEM), transmission electron microscopy (TEM), and fluorescent microscopy (FM). Gas chromatography/mass spectrometry (GC-MS) was used to investigate the chemical composition of plant extracts. RESULTS The extracts damaged the internal and external anatomy of the cytoplasmic membrane and inner structure at a concentration of 0.04 mg/mL. Extracts also led to an increased influx of propidium iodide into treated bacterial cells suggesting compromised cellular integrity and cellular damage. Non-polar compounds such as α-amyrin, friedelan-3-one, lupeol, and β-sitosterol were abundant in the extracts. CONCLUSIONS The extracts of S. legatii and E. zeyheri caused ultrastructural damage to E. coli cells characterized by altered external and internal morphology. These observations may assist in elucidating the mode of action of the extracts.
Collapse
Affiliation(s)
- Ibukun M. Famuyide
- Department of Paraclinical Sciences, Phytomedicine Programme, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort, 0110 South Africa
| | - Folorunso O. Fasina
- grid.49697.350000 0001 2107 2298Dept of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort, 0110 South Africa
- Present Address: Emergency Centre for Transboundary Animal Diseases-Food and Agriculture Organization of the United Nations (ECTAD-FAO), House H. Sida, Ali Hassan Mwinyi Road, Ada Estate, Dar es Salaam, Tanzania
| | - Jacobus N. Eloff
- Department of Paraclinical Sciences, Phytomedicine Programme, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort, 0110 South Africa
| | - Lyndy J. McGaw
- Department of Paraclinical Sciences, Phytomedicine Programme, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort, 0110 South Africa
| |
Collapse
|
33
|
Zhang Y, Xie Y, Tang J, Wang S, Wang L, Zhu G, Li X, Liu Y. Thermal inactivation of Cronobacter sakazakii ATCC 29544 in powdered infant formula milk using thermostatic radio frequency. Food Control 2020. [DOI: 10.1016/j.foodcont.2020.107270] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
34
|
Advances in antimicrobial peptides-based biosensing methods for detection of foodborne pathogens: A review. Food Control 2020. [DOI: 10.1016/j.foodcont.2020.107116] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
35
|
Lin S, Li H, Tao Y, Liu J, Yuan W, Chen Y, Liu Y, Liu S. In Vitro and in Vivo Evaluation of Membrane-Active Flavone Amphiphiles: Semisynthetic Kaempferol-Derived Antimicrobials against Drug-Resistant Gram-Positive Bacteria. J Med Chem 2020; 63:5797-5815. [DOI: 10.1021/acs.jmedchem.0c00053] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shuimu Lin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Hongxia Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yiwen Tao
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Jiayong Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Wenchang Yuan
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yongzhi Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Ying Liu
- Guangdong Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou, Guangdong 510632, P. R. China
| | - Shouping Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| |
Collapse
|
36
|
Tomita S, Hirayasu A, Kajikawa A, Igimi S, Shinohara H, Yokota K. Adsorption of Tolaasins, the Toxins Behind Mushroom Bacterial Blotch, by Microbacterium spp. is Insufficient for Its Detoxification. Curr Microbiol 2020; 77:910-917. [PMID: 31965226 DOI: 10.1007/s00284-020-01884-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/10/2020] [Indexed: 02/03/2023]
Abstract
Tolaasins are lipodepsipeptides secreted by Pseudomonas tolaasii, the causal agent of bacterial blotch on several kinds of cultivated mushrooms. Our previous study reported on tolaasin detoxification by Microbacterium sp. K3-5 as a potential biocontrol of the disease. In this study, the tolaasin-detoxifying activities of various type strains of Microbacterium spp. were evaluated through chemical and biological assays. The bacterial cells of all tested strains of Microbacterium spp. showed tolaasin I-elimination from liquid phase. However, the toxin activities of tolaasins were still retained on the tolaasin-treated bacterial cells of all Microbacterium strains except M. foliorum NBRC 103072T. Furthermore, intact tolaasin I was recovered from the tolaasin-treated bacterial cells of all tested strains except M. foliorum NBRC 103072T. Our data reveal that Microbacterium spp. can be characterized as effective tolaasin I-eliminating bacteria through cell adsorption, but that this adsorption alone is insufficient for actual tolaasin detoxification. The biological degradation process must be needed to carry out the detoxification.
Collapse
Affiliation(s)
- Shun Tomita
- Department of Agricultural Chemistry, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Anna Hirayasu
- Department of Agricultural Chemistry, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Akinobu Kajikawa
- Department of Agricultural Chemistry, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Shizunobu Igimi
- Department of Agricultural Chemistry, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Hirosuke Shinohara
- Department of Agriculture, Tokyo University of Agriculture, Kanagawa, 243-0034, Japan
| | - Kenji Yokota
- Department of Agricultural Chemistry, Tokyo University of Agriculture, Tokyo, 156-8502, Japan.
| |
Collapse
|
37
|
Wu Y, Zhou L, Lu F, Bie X, Zhao H, Zhang C, Lu Z, Lu Y. Discovery of a Novel Antimicrobial Lipopeptide, Brevibacillin V, from Brevibacillus laterosporus fmb70 and Its Application on the Preservation of Skim Milk. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:12452-12460. [PMID: 31674183 DOI: 10.1021/acs.jafc.9b04113] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Increasing cases of infections by foodborne pathogenic bacteria resulted in a great demand to find safe and novel antimicrobial compounds that can be used in the food industry. The isolation and application of antimicrobial peptides including lipopeptides has been increasing tremendously in the past years. In this study, a new bacterial strain called Brevibacillus laterosporus fmb70 (fmb70) was isolated and exhibited strong antimicrobial activities against Gram-positive, Gram-negative bacteria, and fungi. Two major antimicrobial components produced by fmb70 were respectively identified as lipopeptide: brevibacillin V (MW: 1570.12 Da) and brevibacillin (MW: 1583.75 Da), of which brevibacillin V was a new compound. Both of them consisted of 13 amino acids and C6 fatty acyl (FA) chain. Brevibacillin V and brevibacillin showed significant antimicrobial activities against most foodborne pathogenic bacteria and phytopathogenic fungi. They stayed activity at 100 °C and remained 50% of their antimicrobial activities at pH 3 for 22 h. Hemolytic activities of them were lower than 8%. They effectively eliminated the S. aureus GIM 1.142 and L. monocytogenes ATCC 21633 in skim milk. In conclusion, the Brevibacillus laterosporus fmb70 and its major antimicrobial components has remarkable potentials in the food industry.
Collapse
Affiliation(s)
- Yubo Wu
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu Province 210095 , China
| | - Libang Zhou
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu Province 210095 , China
| | - Fengxia Lu
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu Province 210095 , China
| | - Xiaomei Bie
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu Province 210095 , China
| | - Haizhen Zhao
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu Province 210095 , China
| | - Chong Zhang
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu Province 210095 , China
| | - Zhaoxin Lu
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu Province 210095 , China
| | - Yingjian Lu
- College of Food Science and Engineering , Nanjing University of Finance and Economics , Nanjing , Jiangsu Province 210023 , China
| |
Collapse
|
38
|
Synergistic Antimicrobial Activity by Light or Thermal Treatment and Lauric Arginate: Membrane Damage and Oxidative Stress. Appl Environ Microbiol 2019; 85:AEM.01033-19. [PMID: 31253679 DOI: 10.1128/aem.01033-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/22/2019] [Indexed: 12/22/2022] Open
Abstract
The need for more effective antimicrobials is critical for the food industry to improve food safety and reduce spoilage of minimally processed foods. The present study was initiated to develop an efficient and novel antimicrobial approach which combines physical treatments (UV-A or mild heat) and generally recognized as safe lauroyl arginate ethyl (LAE) to inactivate surrogate strains, including Escherichia coli and Listeria innocua Synergistic inactivation of bacteria resulted in an ∼6-log reduction of target bacteria, while individual treatments resulted in <1.5-log inactivation under the same set of conditions. In addition, the synergistic mechanism between LAE and UV-A/mild heat was evaluated by supplementing with a variety of antioxidants for suppressing oxidative stress and measurement of cell membrane damage by nucleic acid release. These results demonstrate that the synergistic antimicrobial activity of LAE and mild physical stresses was suppressed by supplementation with antioxidants. The research also compared LAE with another membrane-targeting lipopeptide antimicrobial agent, polymyxin B, to understand the uniqueness of LAE-induced synergy. Briefly, differences in modes of action between LAE and polymyxin B were characterized by comparing the MIC, damage to liposomes, and oxidative stress generation. These differences in the mode of action between LAE and polymyxin B suggested that both compounds target cell membrane but significantly differ in mechanisms, including membrane disruption and oxidative stress generation. Overall, this study illustrates synergistic antimicrobial activity of LAE with light or mild heat and indicates a novel oxidative stress pathway that enhances the activity of LAE beyond membrane damage.IMPORTANCE This study highlights an effective antimicrobial processing approach using a novel combination of lauroyl arginate ethyl (LAE) and two different physical treatments, light (UV-A) and mild heat. Both combinations demonstrated synergistic inactivation against a model Gram-negative bacterium or a Gram-positive bacterium or both by a >5-log reduction. Further mechanistic study revealed that oxidative stress is responsible for synergistic inactivation between LAE and UV-A, while both membrane damage and oxidative stress are responsible for the synergistic combination between LAE and mild heat. The mode of action of LAE was further compared to that of polymyxin B and analyzed using artificial membrane model systems and the addition of antioxidants. The proposed combination of LAE and common physical treatments may improve food preservation, food safety, and current sanitation processes for the food industry and the inactivation of pathogenic strains in biomedical environments.
Collapse
|
39
|
Yasir M, Dutta D, Willcox MDP. Mode of action of the antimicrobial peptide Mel4 is independent of Staphylococcus aureus cell membrane permeability. PLoS One 2019; 14:e0215703. [PMID: 31356627 PMCID: PMC6663011 DOI: 10.1371/journal.pone.0215703] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/11/2019] [Indexed: 01/30/2023] Open
Abstract
Mel4 is a novel cationic peptide with potent activity against Gram-positive bacteria. The current study examined the anti-staphylococcal mechanism of action of Mel4 and its precursor peptide melimine. The interaction of peptides with lipoteichoic acid (LTA) and with the cytoplasmic membrane using DiSC(3)-5, Sytox green, Syto-9 and PI dyes were studied. Release of ATP and DNA/RNA from cells exposed to the peptides were determined. Bacteriolysis and autolysin-activated cell death were determined by measuring decreases in OD620nm and killing of Micrococcus lysodeikticus cells by cell-free media. Both peptides bound to LTA and rapidly dissipated the membrane potential (within 30 seconds) without affecting bacterial viability. Disturbance of the membrane potential was followed by the release of ATP (50% of total cellular ATP) by melimine and by Mel4 (20%) after 2 minutes exposure (p<0.001). Mel4 resulted in staphylococcal cells taking up PI with 3.9% cells predominantly stained after 150 min exposure, whereas melimine showed 34% staining. Unlike melimine, Mel4 did not release DNA/RNA. Cell-free media from Mel4 treated cells hydrolysed peptidoglycan and produced greater zones of inhibition against M. lysodeikticus lawn than melimine treated samples. These findings suggest that pore formation is unlikely to be involved in Mel4-mediated membrane destabilization for staphylococci, since there was no significant Mel4-induced PI staining and DNA/RNA leakage. It is likely that the S. aureus killing mechanism of Mel4 involves the release of autolysins followed by cell death. Whereas, membrane interaction is the primary bactericidal activity of melimine, which includes membrane depolarization, pore formation, release of cellular contents leading to cell death.
Collapse
Affiliation(s)
- Muhammad Yasir
- School of Optometry and Vision Science, University of New South Wales, Sydney, New South Wales, Australia
- * E-mail:
| | - Debarun Dutta
- School of Optometry and Vision Science, University of New South Wales, Sydney, New South Wales, Australia
- Ophthalmic Research Group, School of Health and Life Sciences, Aston University Birmingham, United Kingdom
| | - Mark D. P. Willcox
- School of Optometry and Vision Science, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Marche MG, Mura ME, Ruiu L. Rapid polymerase chain reaction assays for Brevibacillus laterosporus detection. J Basic Microbiol 2019; 59:853-857. [PMID: 31250936 DOI: 10.1002/jobm.201900188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/25/2019] [Accepted: 06/06/2019] [Indexed: 11/10/2022]
Abstract
The identification of the ubiquitous spore-forming bacterium Brevibacillus laterosporus, whose interest in pharma, agriculture, and other industrial sectors is raising, mostly relies on 16S ribosomal RNA gene sequence analysis. However, due to bacterial gene homology, this method appears insufficient for a proper discrimination of this species, so that the availability of other target genes is necessary. Leveraging the morphological and genetic feature uniqueness of B. laterosporus, a sensitive and reliable detection and quantification method based on polymerase chain reaction (PCR) and quantitative PCR assays, respectively, was developed. Targeting a highly conserved spore surface protein-related gene, B. laterosporus could be easily found in different matrices including soil, food, and insect body. Primer set selectivity was confirmed to be very specific and no false positives or negatives were observed using DNA of different bacterial species as a template. The method developed is also suitable for the rapid identification of newly isolated B. laterosporus strains.
Collapse
Affiliation(s)
| | | | - Luca Ruiu
- Dipartimento di Agraria, University of Sassari, Sassari, Italy
| |
Collapse
|
41
|
Glare TR, Durrant A, Berry C, Palma L, Ormskirk MM, Cox MP. Phylogenetic determinants of toxin gene distribution in genomes of Brevibacillus laterosporus. Genomics 2019; 112:1042-1053. [PMID: 31226484 PMCID: PMC6978878 DOI: 10.1016/j.ygeno.2019.06.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 05/16/2019] [Accepted: 06/17/2019] [Indexed: 11/24/2022]
Abstract
Brevibacillus laterosporus is a globally ubiquitous, spore forming bacterium, strains of which have shown toxic activity against invertebrates and microbes and several have been patented due to their commercial potential. Relatively little is known about this bacterium. Here, we examined the genomes of six published and five newly determined genomes of B. laterosporus, with an emphasis on the relationships between known and putative toxin encoding genes, as well as the phylogenetic relationships between strains. Phylogenetically, strain relationships are similar using average nucleotide identity (ANI) values and multi-gene approaches, although PacBio sequencing revealed multiple copies of the 16S rDNA gene which lessened utility at the strain level. Based on ANI values, the New Zealand isolates were distant from other isolates and may represent a new species. While all of the genomes examined shared some putative toxicity or virulence related proteins, many specific genes were only present in a subset of strains. We examined genomes of 11 Brevibacillus laterosporus, a bacterium which is antagonistic to invertebrates and/or microbes Multiple phylogenetic methods showed New Zealand isolates more distant than all other isolates Each genome could contain 11–13 copies of the 16S rDNA gene, some of which were not identical Many putative toxin encoding genes were present in the genomes, but the toxin complement varied from isolate to isolate Variation in occurrence of toxin-encoding genes indicates the potential to find strains with new combinations of activities
Collapse
Affiliation(s)
- Travis R Glare
- Bio-Protection Research Centre, PO Box 85084, Lincoln University, Lincoln, New Zealand.
| | - Abigail Durrant
- Bio-Protection Research Centre, PO Box 85084, Lincoln University, Lincoln, New Zealand
| | - Colin Berry
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Leopoldo Palma
- Universidad Nacional de Villa María, Instituto A.P. de Ciencias Básicas y Aplicadas, Av. Arturo Jauretche 1555, Villa María 5900, Córdoba, Argentina
| | - M Marsha Ormskirk
- Bio-Protection Research Centre, PO Box 85084, Lincoln University, Lincoln, New Zealand
| | - Murray P Cox
- Statistics and Bioinformatics Group, Institute of Fundamental Sciences, Massey University, Palmerston North 4410, New Zealand
| |
Collapse
|
42
|
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
|
43
|
Azalomycin F5a, a polyhydroxy macrolide binding to the polar head of phospholipid and targeting to lipoteichoic acid to kill methicillin-resistant Staphylococcus aureus. Biomed Pharmacother 2019; 109:1940-1950. [DOI: 10.1016/j.biopha.2018.11.067] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 11/22/2022] Open
|
44
|
Lee J, Kim S, Sim JY, Lee D, Kim HH, Hwang JS, Lee DG, Park ZY, Kim JI. A potent antibacterial activity of new short d-enantiomeric lipopeptide against multi drug resistant bacteria. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1861:34-42. [PMID: 30393205 DOI: 10.1016/j.bbamem.2018.10.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/27/2018] [Accepted: 10/23/2018] [Indexed: 12/27/2022]
Abstract
The emergence of drug-resistant pathogenic bacteria threatens human health. Resistance to existing antibiotics is increasing, while the emergence of new antibiotics is slowing. Cationic antimicrobial peptides (CAMPs) are fascinating alternative antibiotics because they possess a broad spectrum of activity, being active against both Gram-positive and Gram-negative bacteria including those resistant to traditional antibiotics. However, low bioavailability resulting from enzymatic degradation and attenuation by divalent cations like Mg2+ and Ca2+ limits their use as antibiotic agents. Here, we report the design of new CAMPs showing both high antibacterial activity and serum stability under physiological ion concentrations. The peptides were designed by applying two approaches, the use of d-enantiomer and lipidation. Based on the sequence of the CopW (LLWIALRKK-NH2), a nonapeptide derived from coprisin, a series of novel d-form CopW lipopeptides with different acyl chain lengths (C6, C8, C10, C12, C14, and C16) were synthesized and evaluated with respect to their activity and salt sensitivity. Among the analogs, the d-form lipopeptide dCopW3 exhibited MIC values ranging from 1.25 to 5 μM against multidrug-resistant bacteria. Significantly, this compound did not induce bacterial resistance and was highly stable in human serum proteases. The results emphasize the potential of cationic d-form lipopeptide as therapeutically valuable antibiotics for treating drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Jaeho Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Shanghyeon Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Ji-Yeong Sim
- School of Life Sciences, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Daeun Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Ha Hyung Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Jae Sam Hwang
- Department of Agricultural Biology, National Academy of Agricultural Science, RDA, 166, Nongsaengmyeong-ro, Iseo-myeon, Wanju_Gun, Jeollabuk-do 55365, Republic of Korea
| | - Dong Gun Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of National Sciences, Kyungpook National University, 80 Daehakro, Bukgu, Daegu 41566, Republic of Korea
| | - Zee-Yong Park
- School of Life Sciences, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Jae Il Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, 123, Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea.
| |
Collapse
|
45
|
Antimicrobial peptides produced by Brevibacillus spp.: structure, classification and bioactivity: a mini review. World J Microbiol Biotechnol 2018; 34:57. [DOI: 10.1007/s11274-018-2437-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/22/2018] [Indexed: 10/17/2022]
|
46
|
Moon SH, Zhang X, Zheng G, Meeker DG, Smeltzer MS, Huang E. Novel Linear Lipopeptide Paenipeptins with Potential for Eradicating Biofilms and Sensitizing Gram-Negative Bacteria to Rifampicin and Clarithromycin. J Med Chem 2017; 60:9630-9640. [PMID: 29136469 DOI: 10.1021/acs.jmedchem.7b01064] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We report the structure-activity relationship analyses of 17 linear lipopeptide paenipeptin analogues. Analogues 7, 12, and 17 were more potent than the lead compound. Analogue 17 was active against carbapenem-resistant and polymyxin-resistant pathogens. This compound at 40 μg/mL resulted in 3 log and 2.6 log reductions of methicillin-resistant Staphylococcus aureus and Pseudomonas aeruginosa, respectively, in catheter-associated biofilms in vitro. Analogue 17 showed little hemolysis at 32 μg/mL and lysed 11% of red blood cells at 64 μg/mL. Analogues 9 and 16 were nonhemolytic and retained potent P. aeruginosa-specific antimicrobial activity. These two analogues when used alone lacked activity against Acinetobacter baumannii and Klebsiella pneumoniae; however, analogue 9 and 16 at 4 μg/mL decreased the MIC of rifampicin and clarithromycin against the same pathogens from 16 to 32 μg/mL to nanomolar levels (sensitization factor: 2048-8192). Therefore, paenipeptins, alone or in combination with rifampicin or clarithromycin, are promising candidates for treating bacterial infections.
Collapse
Affiliation(s)
- Sun Hee Moon
- Department of Environmental and Occupational Health, ‡Department of Pharmaceutical Sciences, §Department of Microbiology and Immunology, and ∥Department of Orthopedic Surgery, University of Arkansas for Medical Sciences , 4301 West Markham Street, Little Rock, Arkansas 72205, United States
| | - Xuan Zhang
- Department of Environmental and Occupational Health, ‡Department of Pharmaceutical Sciences, §Department of Microbiology and Immunology, and ∥Department of Orthopedic Surgery, University of Arkansas for Medical Sciences , 4301 West Markham Street, Little Rock, Arkansas 72205, United States
| | - Guangrong Zheng
- Department of Environmental and Occupational Health, ‡Department of Pharmaceutical Sciences, §Department of Microbiology and Immunology, and ∥Department of Orthopedic Surgery, University of Arkansas for Medical Sciences , 4301 West Markham Street, Little Rock, Arkansas 72205, United States
| | - Daniel G Meeker
- Department of Environmental and Occupational Health, ‡Department of Pharmaceutical Sciences, §Department of Microbiology and Immunology, and ∥Department of Orthopedic Surgery, University of Arkansas for Medical Sciences , 4301 West Markham Street, Little Rock, Arkansas 72205, United States
| | - Mark S Smeltzer
- Department of Environmental and Occupational Health, ‡Department of Pharmaceutical Sciences, §Department of Microbiology and Immunology, and ∥Department of Orthopedic Surgery, University of Arkansas for Medical Sciences , 4301 West Markham Street, Little Rock, Arkansas 72205, United States
| | - En Huang
- Department of Environmental and Occupational Health, ‡Department of Pharmaceutical Sciences, §Department of Microbiology and Immunology, and ∥Department of Orthopedic Surgery, University of Arkansas for Medical Sciences , 4301 West Markham Street, Little Rock, Arkansas 72205, United States
| |
Collapse
|
47
|
Draft Genome Sequence of Brevibacillus laterosporus OSY-I 1, a Strain That Produces Brevibacillin, Which Combats Drug-Resistant Gram-Positive Bacteria. GENOME ANNOUNCEMENTS 2017; 5:5/41/e01093-17. [PMID: 29025947 PMCID: PMC5637507 DOI: 10.1128/genomea.01093-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Brevibacillus laterosporus OSY-I1 is a Gram-positive spore-forming bacterium isolated from soil. The bacterium produces brevibacillin, an antimicrobial lipopeptide effective against several drug-resistant Gram-positive bacteria. Here, we present the draft genome sequence of the strain OSY-I1 and the gene cluster responsible for the biosynthesis of brevibacillin.
Collapse
|