1
|
Yang P, Liao X. High pressure processing plus technologies: Enhancing the inactivation of vegetative microorganisms. ADVANCES IN FOOD AND NUTRITION RESEARCH 2024; 110:145-195. [PMID: 38906586 DOI: 10.1016/bs.afnr.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
High pressure processing (HPP) is a non-thermal technology that can ensure microbial safety without compromising food quality. However, the presence of pressure-resistant sub-populations, the revival of sub-lethally injured (SLI) cells, and the resuscitation of viable but non-culturable (VBNC) cells pose challenges for its further development. The combination of HPP with other methods such as moderate temperatures, low pH, and natural antimicrobials (e.g., bacteriocins, lactate, reuterin, endolysin, lactoferrin, lactoperoxidase system, chitosan, essential oils) or other non-thermal processes (e.g., CO2, UV-TiO2 photocatalysis, ultrasound, pulsed electric fields, ultrafiltration) offers feasible alternatives to enhance microbial inactivation, termed as "HPP plus" technologies. These combinations can effectively eliminate pressure-resistant sub-populations, reduce SLI or VBNC cell populations, and inhibit their revival or resuscitation. This review provides an updated overview of microbial inactivation by "HPP plus" technologies and elucidates possible inactivation mechanisms.
Collapse
Affiliation(s)
- Peiqing Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P.R. China
| | - Xiaojun Liao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P.R. China; National Engineering Research Center for Fruit & Vegetable Processing, Beijing, P.R. China; Key Laboratory of Fruit & Vegetable Processing, Ministry of Agriculture and Rural Affairs, Beijing, P.R. China; Beijing Key laboratory for Food Non-thermal processing, Beijing, P.R. China.
| |
Collapse
|
2
|
Gao S, Ji Y, Xu S, Jia J, Fan B, Zhang Y, Shen H, Zhou W. Antifungal activity of nisin against clinical isolates of azole-resistant Candida tropicalis. Front Microbiol 2024; 15:1383953. [PMID: 38774506 PMCID: PMC11106359 DOI: 10.3389/fmicb.2024.1383953] [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: 02/08/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
The rapid emergence of invasive infections caused by azole-resistant Candida tropicalis has become a public health concern, and there is an urgent need for alternative treatment strategies. Studies have demonstrated the antibacterial effects of nisin, a well-known peptide naturally produced by Lactococcus lactis subsp. lactis. However, there is scant information about the antifungal effect of nisin against C. tropicalis. The present study aims to investigate the in vitro antifungal activity of nisin against clinical isolates of azole-resistant C. tropicalis strains, as well as its inhibitory effect on biofilm formation. A total of 35 C. tropicalis strains isolated from patients with invasive fungal infections were divided into the azole-resistant group and the azole-sensitive group, containing 21 and 14 strains, respectively. The relative expression levels of the ERG11 and UPC2 genes in the azole-resistant group were higher than those in the azole-sensitive group (p < 0.0001), while no significant differences were observed in the expression levels of the MDR1 and CDR1 genes. The minimum inhibitory concentration of nisin against C. tropicalis ranged from 2 to 8 μg/mL. Nisin treatment inhibited the growth of azole-resistant C. tropicalis, with over a four-fold reduction in OD600 nm values observed at the 8-h time point, while it promoted the transition of C. tropicalis from the spore phase to the hyphal phase, as observed on cryo-scanning electron microscopy. The results of biofilm quantification using crystal violet staining indicated a significant decrease in OD570 nm values in the nisin-treated group compared to the controls (p < 0.0001). Among the 21 azole-resistant C. tropicalis strains, the biofilm formation was inhibited in 17 strains (17/21, 81%), and more than 85% inhibition of biofilm formation was observed in the representative strains. With regard to the molecular mechanisms, the expression of the BCR1 and UPC2 genes in the azole-resistant strains was down-regulated on nisin treatment (p < 0.05). In conclusion, we demonstrated, for the first time, that nisin has antifungal activity and significant anti-biofilm activity against clinical isolates of azole-resistant C. tropicalis strains. Based on the findings, nisin could be a promising alternative antifungal agent for combating azole-resistant C. tropicalis infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wanqing Zhou
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Wolden R, Ovchinnikov KV, Venter HJ, Oftedal TF, Diep DB, Cavanagh JP. The novel bacteriocin romsacin from Staphylococcus haemolyticus inhibits Gram-positive WHO priority pathogens. Microbiol Spectr 2023; 11:e0086923. [PMID: 37905822 PMCID: PMC10715183 DOI: 10.1128/spectrum.00869-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/24/2023] [Indexed: 11/02/2023] Open
Abstract
IMPORTANCE Bacteria produce bacteriocins to inhibit growth of other bacterial species. We have studied the antimicrobial activity of a new bacteriocin produced by the skin bacterium S. haemolyticus. The bacteriocin is effective against several types of Gram-positive bacteria, including highly virulent and antibiotic-resistant strains such as Staphylococcus aureus and Enterococcus faecium. Effective antimicrobials are important for the treatment of infections and the success of major surgery and chemotherapy. Bacteriocins can be part of the solution to the global concern of antimicrobial resistance.
Collapse
Affiliation(s)
- Runa Wolden
- Department of Clinical Medicine, Faculty of Health Sciences, Research Group for Child and Adolescent Health, UiT The Arctic University of Norway, Tromsø, Norway
| | - Kirill V. Ovchinnikov
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Hermoine J. Venter
- Department of Clinical Medicine, Faculty of Health Sciences, Research Group for Child and Adolescent Health, UiT The Arctic University of Norway, Tromsø, Norway
| | - Thomas F. Oftedal
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Dzung B. Diep
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Jorunn Pauline Cavanagh
- Department of Clinical Medicine, Faculty of Health Sciences, Research Group for Child and Adolescent Health, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
4
|
Gupta R, Singh M, Pathania R. Chemical genetic approaches for the discovery of bacterial cell wall inhibitors. RSC Med Chem 2023; 14:2125-2154. [PMID: 37974958 PMCID: PMC10650376 DOI: 10.1039/d3md00143a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 08/10/2023] [Indexed: 11/19/2023] Open
Abstract
Antimicrobial resistance (AMR) in bacterial pathogens is a worldwide health issue. The innovation gap in discovering new antibiotics has remained a significant hurdle in combating the AMR problem. Currently, antibiotics target various vital components of the bacterial cell envelope, nucleic acid and protein biosynthesis machinery and metabolic pathways essential for bacterial survival. The critical role of the bacterial cell envelope in cell morphogenesis and integrity makes it an attractive drug target. While a significant number of in-clinic antibiotics target peptidoglycan biosynthesis, several components of the bacterial cell envelope have been overlooked. This review focuses on various antibacterial targets in the bacterial cell wall and the strategies employed to find their novel inhibitors. This review will further elaborate on combining forward and reverse chemical genetic approaches to discover antibacterials that target the bacterial cell envelope.
Collapse
Affiliation(s)
- Rinki Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee Roorkee - 247 667 Uttarakhand India
| | - Mangal Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee Roorkee - 247 667 Uttarakhand India
| | - Ranjana Pathania
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee Roorkee - 247 667 Uttarakhand India
| |
Collapse
|
5
|
Khan F, Singh P, Joshi AS, Tabassum N, Jeong GJ, Bamunuarachchi NI, Mijakovic I, Kim YM. Multiple potential strategies for the application of nisin and derivatives. Crit Rev Microbiol 2023; 49:628-657. [PMID: 35997756 DOI: 10.1080/1040841x.2022.2112650] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/28/2022] [Accepted: 08/09/2022] [Indexed: 12/22/2022]
Abstract
Nisin is a naturally occurring bioactive small peptide produced by Lactococcus lactis subsp. lactis and belongs to the Type A (I) lantibiotics. Due to its potent antimicrobial activity, it has been broadly employed to preserve various food materials as well as to combat a variety of microbial pathogens. The present review discusses the antimicrobial properties of nisin and different types of their derivatives employed to treat microbial pathogens with a detailed underlying mechanism of action. Several alternative strategies such as combination, conjugation, and nanoformulations have been discussed in order to address several issues such as rapid degradation, instability, and reduced activity due to the various environmental factors that arise in the applications of nisin. Furthermore, the evolutionary relationship of many nisin genes from different nisin-producing bacterial species has been investigated. A detailed description of the natural and bioengineered nisin variants, as well as the underlying action mechanisms, has also been provided. The chemistry used to apply nisin in conjugation with natural or synthetic compounds as a synergetic mode of antimicrobial action has also been thoroughly discussed. The current review will be useful in learning about recent and past research that has been performed on nisin and its derivatives as antimicrobial agents.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
| | - Priyanka Singh
- The Novo Nordisk Foundation, Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Abhayraj S Joshi
- The Novo Nordisk Foundation, Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Nazia Tabassum
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
| | | | - Ivan Mijakovic
- The Novo Nordisk Foundation, Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
- Systems and Synthetic Biology Division, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
6
|
Leena Panigrahi L, Shekhar S, Sahoo B, Arakha M. Adsorption of antimicrobial peptide onto chitosan-coated iron oxide nanoparticles fosters oxidative stress triggering bacterial cell death. RSC Adv 2023; 13:25497-25507. [PMID: 37636508 PMCID: PMC10450573 DOI: 10.1039/d3ra04070d] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/11/2023] [Indexed: 08/29/2023] Open
Abstract
In the prevailing environmental status quo, bacterial resistance has made antibiotics and antimicrobial peptides (AMPs) ineffective, imparting a serious threat and putting a much greater financial burden on the biomedical and food industries. For this reason, the present study investigates the potential of iron oxide nanoparticles (IONPs) coated with chitosan (CS-IONP) as a platform for augmenting the antimicrobial activity of antimicrobial peptides like nisin. Hence, the nisin is allowed to be adsorbed onto chitosan-coated IONPs to formulate nisin-loaded CS-IONP nanoconjugates. The nanoconjugates were characterized by various optical techniques, such as XRD, FTIR, SEM, zeta and DLS. Remarkably, lower concentrations of N-CS-IONP nanoconjugate exhibited significant and broad-spectrum antibacterial potency compared to bare IONPs and nisin against both Gram-positive and Gram-negative bacteria. Biofilm production was also found to be drastically reduced in the presence of nanoconjugates. Further investigation established a relationship between an increase in antibacterial activity and the enhanced generation of reactive oxygen species (ROS). Oxidative stress exhibited due to enhanced ROS generation is a conclusive reason for the rupturing of bacterial membranes and leakage of cytoplasmic contents, eventually leading to the death of the bacteria. Thus, the current study emphasizes the formulation of a novel antimicrobial agent which exploits magnetic nanoparticles modulated with chitosan for enhanced remediation of resistant bacteria due to oxidative stress imparted by the nanoconjugates upon interaction with the bacteria, leading to cell death.
Collapse
Affiliation(s)
- Lipsa Leena Panigrahi
- Center for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar 751003 Odisha India
| | | | - Banishree Sahoo
- Center for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar 751003 Odisha India
| | - Manoranjan Arakha
- Center for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Bhubaneswar 751003 Odisha India
| |
Collapse
|
7
|
Identification, Purification, Characterization and Biopreservation Potential of Antimicrobial Peptide of Pediococcus acidilactici NCDC 252. Int J Pept Res Ther 2023. [DOI: 10.1007/s10989-022-10485-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
8
|
Bacteriocin Production by Bacillus Species: Isolation, Characterization, and Application. Probiotics Antimicrob Proteins 2022; 14:1151-1169. [PMID: 35881232 DOI: 10.1007/s12602-022-09966-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 12/25/2022]
Abstract
Antibiotic resistance is a problem that has been increasing lately; therefore, it is important to find new alternatives to treat infections induced by pathogens that cannot be eliminated with available products. Small antimicrobial peptides (AMPs) known as bacteriocin could be an alternative to antibiotics because they have shown to be effective against a great number of multidrug-resistant microbes. In addition to its high specificity against microbial pathogens and its low cytotoxicity against human cells, most bacteriocin present tolerance to enzyme degradation and stability to temperature and pH alterations. Bacteriocins are small peptides with a great diversity of structures and functions; however, their mechanisms of action are still not well understood. In this review, bacteriocin produced by Bacillus species will be described, especially its mechanisms of action, culture conditions used to improve its production and state-of-the-art methodologies applied to identify them. Bacteriocin utilization as food preservatives and as new molecules to treat cancer also will be discussed.
Collapse
|
9
|
Maurizzi E, Bigi F, Quartieri A, De Leo R, Volpelli LA, Pulvirenti A. The Green Era of Food Packaging: General Considerations and New Trends. Polymers (Basel) 2022; 14:polym14204257. [PMID: 36297835 PMCID: PMC9610407 DOI: 10.3390/polym14204257] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
Recently, academic research and industries have gained awareness about the economic, environmental, and social impacts of conventional plastic packaging and its disposal. This consciousness has oriented efforts towards more sustainable materials such as biopolymers, paving the way for the “green era” of food packaging. This review provides a schematic overview about polymers and blends of them, which are emerging as promising alternatives to conventional plastics. Focus was dedicated to biopolymers from renewable sources and their applications to produce sustainable, active packaging with antimicrobial and antioxidant properties. In particular, the incorporation of plant extracts, food-waste derivatives, and nano-sized materials to produce bio-based active packaging with enhanced technical performances was investigated. According to recent studies, bio-based active packaging enriched with natural-based compounds has the potential to replace petroleum-derived materials. Based on molecular composition, the natural compounds can diversely interact with the native structure of the packaging materials, modulating their barriers, optical and mechanical performances, and conferring them antioxidant and antimicrobial properties. Overall, the recent academic findings could lead to a breakthrough in the field of food packaging, opening the gates to a new generation of packaging solutions which will be sustainable, customised, and green.
Collapse
Affiliation(s)
- Enrico Maurizzi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Correspondence:
| | - Francesco Bigi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Andrea Quartieri
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Riccardo De Leo
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Luisa Antonella Volpelli
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Interdepartmental Research Centre for the Improvement of Agro-Food Biological Resources (BIOGEST-SITEIA), University of Modena and Reggio Emilia, 42124 Reggio Emilia, Italy
| | - Andrea Pulvirenti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Interdepartmental Research Centre for the Improvement of Agro-Food Biological Resources (BIOGEST-SITEIA), University of Modena and Reggio Emilia, 42124 Reggio Emilia, Italy
| |
Collapse
|
10
|
Bucheli JEV, Fugaban JII, Holzapfel WH, Todorov SD. Combined Action of Antibiotics and Bacteriocins against Vancomycin-Resistant Enterococci. Microorganisms 2022; 10:microorganisms10071423. [PMID: 35889141 PMCID: PMC9324536 DOI: 10.3390/microorganisms10071423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/06/2022] [Accepted: 07/13/2022] [Indexed: 02/01/2023] Open
Abstract
Antibiotics have been one of the most important discoveries in the area of applied medical microbiology; however, as a result of various factors, we are currently facing a dramatic and relatively dangerous increase in the number of cases of antibiotic resistance, and the need for new types of antimicrobials continues to grow. New approaches are needed to combat antibiotic-resistant pathogens. Bacteriocins, as part of the group of antimicrobial peptides, can be considered as alternatives and/or complements to known antibiotics. Their narrow spectra of activity can be explored for the control of various pathogens, such as vancomycin-resistant enterococci (VRE), as single therapies or in combination with known antibiotics. In the present study, we isolated bacteriocins from different lactic acid bacteria (LAB) strains, including Enterococcus and Pediococcus, and explored the possible synergistic inhibition of growth by bacteriocins and vancomycin. It was observed in the growth dynamics with previously selected VRE strains that the bacteriocins had a high specificity and a promising inhibitory effect against the VRE strains, and these results were validated by a propidium iodide viability test using flow cytometry. The data obtained indicate that the selected bacteriocins can be used to control VRE in the food industry or even as an alternative treatment to combat infections with antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Jorge Enrique Vazquez Bucheli
- Human Effective Microbes Laboratory, Department of Advanced Convergence, Handong Global University, Pohang 37554, Korea; (J.E.V.B.); (W.H.H.)
- ProBacLab, Department of Advanced Convergence, Handong Global University, Pohang 37554, Korea;
| | | | - Wilhelm Heinrich Holzapfel
- Human Effective Microbes Laboratory, Department of Advanced Convergence, Handong Global University, Pohang 37554, Korea; (J.E.V.B.); (W.H.H.)
| | - Svetoslav Dimitrov Todorov
- ProBacLab, Department of Advanced Convergence, Handong Global University, Pohang 37554, Korea;
- Correspondence: ; Tel.: +82-10-3490-3152
| |
Collapse
|
11
|
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]
|
12
|
Anti-adhesion and Anti-inflammatory Potential of the Leaderless Class IIb Bacteriocin Enterocin DD14. Probiotics Antimicrob Proteins 2022; 14:613-619. [PMID: 35604525 PMCID: PMC9125348 DOI: 10.1007/s12602-022-09954-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2022] [Indexed: 12/01/2022]
Abstract
In this study, we investigate the interactions between the leaderless class IIb bacteriocin, enterocin DD14 (EntDD14), or the methicillin or the combination of these antibacterials, and two methicillin-resistant Staphylococcus aureus strains (MRSA-S1 and USA 300) which are respectively a clinical strain and a reference strain. The results obtained showed that EntDD14 alone or in combination with the antibiotic could significantly prevent the adhesion of these pathogenic bacteria to human cells. On the other hand, we investigated the anti-inflammatory effect of EntDD14 on the secretion of pro-inflammatory interleukins, including IL-6 and IL-8. The results show that EntDD14 is able to decrease significantly the secretion of both interleukins on Caco-2 cells following their treatments with lipopolysaccharides. These novel data provide insightful informations to support applications of bacteriocins as therapeutic agents capable as well to defeat pathogenic bacteria and concomitantly limit their inflammatory reactions.
Collapse
|
13
|
Amaning Danquah C, Minkah PAB, Osei Duah Junior I, Amankwah KB, Somuah SO. Antimicrobial Compounds from Microorganisms. Antibiotics (Basel) 2022; 11:285. [PMID: 35326749 PMCID: PMC8944786 DOI: 10.3390/antibiotics11030285] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/27/2022] [Accepted: 02/07/2022] [Indexed: 02/06/2023] Open
Abstract
Antimicrobial resistance is an exigent public health concern owing to the emergence of novel strains of human resistant pathogens and the concurrent rise in multi-drug resistance. An influx of new antimicrobials is urgently required to improve the treatment outcomes of infectious diseases and save lives. Plant metabolites and bioactive compounds from chemical synthesis have found their efficacy to be dwindling, despite some of them being developed as drugs and used to treat human infections for several decades. Microorganisms are considered untapped reservoirs for promising biomolecules with varying structural and functional antimicrobial activity. The advent of cost-effective and convenient model organisms, state-of-the-art molecular biology, omics technology, and machine learning has enhanced the bioprospecting of novel antimicrobial drugs and the identification of new drug targets. This review summarizes antimicrobial compounds isolated from microorganisms and reports on the modern tools and strategies for exploiting promising antimicrobial drug candidates. The investigation identified a plethora of novel compounds from microbial sources with excellent antimicrobial activity against disease-causing human pathogens. Researchers could maximize the use of novel model systems and advanced biomolecular and computational tools in exploiting lead antimicrobials, consequently ameliorating antimicrobial resistance.
Collapse
Affiliation(s)
- Cynthia Amaning Danquah
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana;
| | - Prince Amankwah Baffour Minkah
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana;
- Global Health and Infectious Disease Research Group, Kumasi Centre for Collaborative Research in Tropical Medicine, College of Health Sciences, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana
| | - Isaiah Osei Duah Junior
- Department of Optometry and Visual Science, College of Science, Kwame Nkrumah University of Science and Technology, PMB, Kumasi, Ghana;
| | - Kofi Bonsu Amankwah
- Department of Biomedical Sciences, University of Cape Coast, PMB, Cape Coast, Ghana;
| | - Samuel Owusu Somuah
- Department of Pharmacy Practice, School of Pharmacy, University of Health and Allied Sciences, PMB, Ho, Ghana;
| |
Collapse
|
14
|
Wang Q, Peng B, Song M, Abdullah, Li J, Miao J, Feng K, Chen F, Zhai X, Cao Y. Effects of Antibacterial Peptide F1 on Bacterial Liposome Membrane Integrity. Front Nutr 2021; 8:768890. [PMID: 34869536 PMCID: PMC8633404 DOI: 10.3389/fnut.2021.768890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/01/2021] [Indexed: 11/26/2022] Open
Abstract
Previous studies from our lab have shown that the antimicrobial peptide F1 obtained from the milk fermentation by Lactobacillus paracasei FX-6 derived from Tibetan kefir was different from common antimicrobial peptides; specifically, F1 simultaneously inhibited the growth of Gram-negative and Gram-positive bacteria. Here, we present follow-on work demonstrating that after the antimicrobial peptide F1 acts on either Escherichia coli ATCC 25922 (E. coli) or Staphylococcus aureus ATCC 63589 (S. aureus), their respective bacterial membranes were severely deformed. This deformation allowed leakage of potassium and magnesium ions from the bacterial membrane. The interaction between the antimicrobial peptide F1 and the bacterial membrane was further explored by artificially simulating the bacterial phospholipid membranes and then extracting them. The study results indicated that after the antimicrobial peptide F1 interacted with the bacterial membranes caused significant calcein leakage that had been simulated by different liposomes. Furthermore, transmission electron microscopy observations revealed that the phospholipid membrane structure was destroyed and the liposomes presented aggregation and precipitation. Quartz Crystal Microbalance with Dissipation (QCM-D) results showed that the antimicrobial peptide F1 significantly reduced the quality of liposome membrane and increased their viscoelasticity. Based on the study's findings, the phospholipid membrane particle size was significantly increased, indicating that the antimicrobial peptide F1 had a direct effect on the phospholipid membrane. Conclusively, the antimicrobial peptide F1 destroyed the membrane structure of both Gram-negative and Gram-positive bacteria by destroying the shared components of their respective phospholipid membranes which resulted in leakage of cell contents and subsequently cell death.
Collapse
Affiliation(s)
- Qun Wang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Bo Peng
- College of Food Science, South China Agricultural University, Guangzhou, China.,Guangdong Haitian Innovation Technology Co., Ltd., Foshan, China
| | - Mingyue Song
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Abdullah
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jun Li
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jianyin Miao
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Konglong Feng
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Feilong Chen
- College of Food Science, South China Agricultural University, Guangzhou, China.,Evonik Rexim Nanning Co., Ltd., Nanning, China
| | | | - Yong Cao
- College of Food Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
15
|
Pokhrel R, Bhattarai N, Baral P, Gerstman BS, Park JH, Handfield M, Chapagain PP. Lipid II Binding and Transmembrane Properties of Various Antimicrobial Lanthipeptides. J Chem Theory Comput 2021; 18:516-525. [PMID: 34874159 DOI: 10.1021/acs.jctc.1c00666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
There has been an alarming rise in antibacterial resistant infections in recent years due to the widespread use of antibiotics, and there is a dire need for the development of new antibiotics utilizing novel modes of action. Lantibiotics are promising candidates to engage in the fight against resistant strains of bacteria due to their unique modes of action, including interference with cell wall synthesis by binding to lipid II and creating pores in bacterial membranes. In this study, we use atomic-scale molecular dynamics computational studies to compare both the lipid II binding ability and the membrane interactions of five lanthipeptides that are commonly used in antimicrobial research: nisin, Mutacin 1140 (MU1140), gallidermin, NVB302, and NAI107. Among the five peptides investigated, nisin is found to be the most efficient at forming water channels through a membrane, whereas gallidermin and MU1140 are found to be better at binding the lipid II molecules. Nisin's effectiveness in facilitating water transport across the membrane is due to the creation of several different water trajectories along with no significant water delay points along the paths. The shorter peptide deoxyactagardine B (NVB302) was found to not form a water channel. These detailed observations provide insights into the dual mechanisms of the action of lantibiotic peptides and can facilitate the design and development of novel lanthipeptides by strategic placement of different residues.
Collapse
Affiliation(s)
| | | | | | | | - Jae H Park
- Oragenics Inc., Alachua, Florida 32615, United States
| | | | | |
Collapse
|
16
|
Listeriolysin S: A bacteriocin from Listeria monocytogenes that induces membrane permeabilization in a contact-dependent manner. Proc Natl Acad Sci U S A 2021; 118:2108155118. [PMID: 34599102 PMCID: PMC8501752 DOI: 10.1073/pnas.2108155118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 11/18/2022] Open
Abstract
Listeria monocytogenes (Lm) is a bacterial pathogen that causes listeriosis, a foodborne disease characterized by gastroenteritis, meningitis, bacteremia, and abortions in pregnant women. The most severe human listeriosis outbreaks are associated with a subset of Lm hypervirulent clones that encode the bacteriocin Listeriolysin S (LLS), which modifies the gut microbiota and allows efficient Lm gut colonization and invasion of deeper organs. Our present work identifies the killing mechanism displayed by LLS to outcompete gut commensal bacteria, demonstrating that it induces membrane permeabilization and membrane depolarization of target bacteria. Moreover, we show that LLS is a thiazole/oxazole–modified microcin that displays a contact-dependent inhibition mechanism. Listeriolysin S (LLS) is a thiazole/oxazole–modified microcin (TOMM) produced by hypervirulent clones of Listeria monocytogenes. LLS targets specific gram-positive bacteria and modulates the host intestinal microbiota composition. To characterize the mechanism of LLS transfer to target bacteria and its bactericidal function, we first investigated its subcellular distribution in LLS-producer bacteria. Using subcellular fractionation assays, transmission electron microscopy, and single-molecule superresolution microscopy, we identified that LLS remains associated with the bacterial cell membrane and cytoplasm and is not secreted to the bacterial extracellular space. Only living LLS-producer bacteria (and not purified LLS-positive bacterial membranes) display bactericidal activity. Applying transwell coculture systems and microfluidic-coupled microscopy, we determined that LLS requires direct contact between LLS-producer and -target bacteria in order to display bactericidal activity, and thus behaves as a contact-dependent bacteriocin. Contact-dependent exposure to LLS leads to permeabilization/depolarization of the target bacterial cell membrane and adenosine triphosphate (ATP) release. Additionally, we show that lipoteichoic acids (LTAs) can interact with LLS and that LTA decorations influence bacterial susceptibility to LLS. Overall, our results suggest that LLS is a TOMM that displays a contact-dependent inhibition mechanism.
Collapse
|
17
|
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
|
18
|
El-Saber Batiha G, Hussein DE, Algammal AM, George TT, Jeandet P, Al-Snafi AE, Tiwari A, Pagnossa JP, Lima CM, Thorat ND, Zahoor M, El-Esawi M, Dey A, Alghamdi S, Hetta HF, Cruz-Martins N. Application of natural antimicrobials in food preservation: Recent views. Food Control 2021. [DOI: 10.1016/j.foodcont.2021.108066] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
19
|
Bacteriocins from Lactic Acid Bacteria. A Powerful Alternative as Antimicrobials, Probiotics, and Immunomodulators in Veterinary Medicine. Animals (Basel) 2021; 11:ani11040979. [PMID: 33915717 PMCID: PMC8067144 DOI: 10.3390/ani11040979] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/20/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
In the search for an alternative treatment to reduce antimicrobial resistance, bacteriocins shine a light on reducing this problem in public and animal health. Bacteriocins are peptides synthesized by bacteria that can inhibit the growth of other bacteria and fungi, parasites, and viruses. Lactic acid bacteria (LAB) are a group of bacteria that produce bacteriocins; their mechanism of action can replace antibiotics and prevent bacterial resistance. In veterinary medicine, LAB and bacteriocins have been used as antimicrobials and probiotics. However, another critical role of bacteriocins is their immunomodulatory effect. This review shows the advances in applying bacteriocins in animal production and veterinary medicine, highlighting their biological roles.
Collapse
|
20
|
Yang P, Rao L, Zhao L, Wu X, Wang Y, Liao X. High pressure processing combined with selected hurdles: Enhancement in the inactivation of vegetative microorganisms. Compr Rev Food Sci Food Saf 2021; 20:1800-1828. [PMID: 33594773 DOI: 10.1111/1541-4337.12724] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/28/2020] [Accepted: 01/21/2021] [Indexed: 12/15/2022]
Abstract
High pressure processing (HPP) as a nonthermal processing (NTP) technology can ensure microbial safety to some extent without compromising food quality. However, for vegetative microorganisms, the existence of pressure-resistant subpopulations, the revival of sublethal injury (SLI) state cells, and the resuscitation of viable but nonculturable (VBNC) state cells may constitute potential food safety risks and pose challenges for the further development of HPP application. HPP combined with selected hurdles, such as moderately elevated or low temperature, low pH, natural antimicrobials (bacteriocin, lactate, reuterin, endolysin, lactoferrin, lactoperoxidase system, chitosan, essential oils), or other NTP (CO2 , UV-TiO2 photocatalysis, ultrasound, pulsed electric field, ultrafiltration), have been highlighted as feasible alternatives to enhance microbial inactivation (synergistic or additive effect). These combinations can effectively eliminate the pressure-resistant subpopulation, reduce the population of SLI or VBNC state cells and inhibit their revival or resuscitation. This review provides an updated overview of the microbial inactivation by the combination of HPP and selected hurdles and restructures the possible inactivation mechanisms.
Collapse
Affiliation(s)
- Peiqing Yang
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing of Ministry of Agriculture and Rural Affairs, Beijing Key Laboratory for Food Non-Thermal Processing, China Agricultural University, Beijing, 100083, China
| | - Lei Rao
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing of Ministry of Agriculture and Rural Affairs, Beijing Key Laboratory for Food Non-Thermal Processing, China Agricultural University, Beijing, 100083, China
| | - Liang Zhao
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing of Ministry of Agriculture and Rural Affairs, Beijing Key Laboratory for Food Non-Thermal Processing, China Agricultural University, Beijing, 100083, China
| | - Xiaomeng Wu
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing of Ministry of Agriculture and Rural Affairs, Beijing Key Laboratory for Food Non-Thermal Processing, China Agricultural University, Beijing, 100083, China
| | - Yongtao Wang
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing of Ministry of Agriculture and Rural Affairs, Beijing Key Laboratory for Food Non-Thermal Processing, China Agricultural University, Beijing, 100083, China
| | - Xiaojun Liao
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing of Ministry of Agriculture and Rural Affairs, Beijing Key Laboratory for Food Non-Thermal Processing, China Agricultural University, Beijing, 100083, China
| |
Collapse
|
21
|
Cui Y, Luo L, Wang X, Lu Y, Yi Y, Shan Y, Liu B, Zhou Y, Lü X. Mining, heterologous expression, purification, antibactericidal mechanism, and application of bacteriocins: A review. Compr Rev Food Sci Food Saf 2020; 20:863-899. [PMID: 33443793 DOI: 10.1111/1541-4337.12658] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/04/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Bacteriocins are generally considered as low-molecular-weight ribosomal peptides or proteins synthesized by G+ and G- bacteria that inhibit or kill other related or unrelated microorganisms. However, low yield is an important factor restricting the application of bacteriocins. This paper reviews mining methods, heterologous expression in different systems, the purification technologies applied to bacteriocins, and identification methods, as well as the antibacterial mechanism and applications in three different food systems. Bioinformatics improves the efficiency of bacteriocins mining. Bacteriocins can be heterologously expressed in different expression systems (e.g., Escherichia coli, Lactobacillus, and yeast). Ammonium sulfate precipitation, dialysis membrane, pH-mediated cell adsorption/desorption, solvent extraction, macroporous resin column, and chromatography are always used as purification methods for bacteriocins. The bacteriocins are identified through electrophoresis and mass spectrum. Cell envelope (e.g., cell permeabilization and pore formation) and inhibition of gene expression are common antibacterial mechanisms of bacteriocins. Bacteriocins can be added to protect meat products (e.g., beef and sausages), dairy products (e.g., cheese, milk, and yogurt), and vegetables and fruits (e.g., salad, apple juice, and soybean sprouts). The future research directions are also prospected.
Collapse
Affiliation(s)
- Yanlong Cui
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Lingli Luo
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xin Wang
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yingying Lu
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yanglei Yi
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yuanyuan Shan
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Bianfang Liu
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yuan Zhou
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xin Lü
- Lab of Bioresources, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| |
Collapse
|
22
|
Shan L, Wenling Q, Mauro P, Stefano B. Antibacterial Agents Targeting the Bacterial Cell Wall. Curr Med Chem 2020; 27:2902-2926. [PMID: 32003656 DOI: 10.2174/0929867327666200128103653] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 11/22/2022]
Abstract
The introduction of antibiotics to treat bacterial infections either by killing or blocking their growth has been accompanied by the studies of mechanism that allows the drugs to kill the bacteria or to stop their proliferation. In such a scenario, the emergence of antibacterial agents active on the bacterial cell wall has been of fundamental importance in the fight against bacterial agents responsible for severe diseases. As a matter of fact, the cell wall, which plays many roles during the lifecycle, is an essential constituent of most bacteria. This overview focuses on the intracellular steps of peptidoglycan biosynthesis and the research of new antibacterial agents based on the enzymes involved in these early steps of the formation of cell membrane components.
Collapse
Affiliation(s)
- Li Shan
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 401331 Chongqing, China
| | - Qin Wenling
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 401331 Chongqing, China
| | - Panunzio Mauro
- Isof-CNR Chemistry Department, Via Selmi, 2, 40126 Bologna, Italy
| | - Biondi Stefano
- BioVersys AG, C/o Technologiepark Basel, Hochbergerstrasse 60c, CH- 4057 Basel, Switzerland
| |
Collapse
|
23
|
O'Sullivan JN, O'Connor PM, Rea MC, O'Sullivan O, Walsh CJ, Healy B, Mathur H, Field D, Hill C, Ross RP. Nisin J, a Novel Natural Nisin Variant, Is Produced by Staphylococcus capitis Sourced from the Human Skin Microbiota. J Bacteriol 2020; 202:e00639-19. [PMID: 31740495 PMCID: PMC6964739 DOI: 10.1128/jb.00639-19] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/05/2019] [Indexed: 02/04/2023] Open
Abstract
The skin microbiota is thought to play a key role in host protection from infection. Nisin J is a novel nisin variant produced by Staphylococcus capitis APC 2923, a strain isolated from the toe web space area in a screening study performed on the human skin microbiota. Whole-genome sequencing and mass spectrometry of the purified peptide confirmed that S. capitis APC 2923 produces a 3,458-Da bacteriocin, designated nisin J, which exhibited antimicrobial activity against a range of Gram-positive pathogens, including methicillin-resistant Staphylococcus aureus (MRSA) and Cutibacterium acnes The gene order in the nisin J gene cluster (nsjFEGBTCJP) differs from that of other nisin variants in that it is lacking the nisin regulatory genes, nisRK, as well as the nisin immunity gene nisI Nisin J has 9 amino acid changes compared to prototypical nisin A, with 8 amino acid substitutions, 6 of which are not present in other nisin variants (Ile4Lys, Met17Gln, Gly18Thr, Asn20Phe, Met21Ala, Ile30Gly, Val33His, and Lys34Thr), and an extra amino acid close to the C terminus, rendering nisin J the only nisin variant to contain 35 amino acids. This is the first report of a nisin variant produced by a Staphylococcus species and the first nisin producer isolated from human skin.IMPORTANCE This study describes the characterization of nisin J, the first example of a natural nisin variant, produced by a human skin isolate of staphylococcal origin. Nisin J displays inhibitory activity against a wide range of bacterial targets, including MRSA. This work demonstrates the potential of human commensals as a source for novel antimicrobials that could form part of the solution to antibiotic resistance across a broad range of bacterial pathogens.
Collapse
Affiliation(s)
- Julie N O'Sullivan
- Teagasc Food Research Centre, Fermoy, County Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paula M O'Connor
- Teagasc Food Research Centre, Fermoy, County Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Mary C Rea
- Teagasc Food Research Centre, Fermoy, County Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Orla O'Sullivan
- Teagasc Food Research Centre, Fermoy, County Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Calum J Walsh
- Teagasc Food Research Centre, Fermoy, County Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Brian Healy
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Harsh Mathur
- Teagasc Food Research Centre, Fermoy, County Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Des Field
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Colin Hill
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- Teagasc Food Research Centre, Fermoy, County Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
24
|
Karas JA, Chen F, Schneider-Futschik EK, Kang Z, Hussein M, Swarbrick J, Hoyer D, Giltrap AM, Payne RJ, Li J, Velkov T. Synthesis and structure-activity relationships of teixobactin. Ann N Y Acad Sci 2019; 1459:86-105. [PMID: 31792983 DOI: 10.1111/nyas.14282] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 11/04/2019] [Accepted: 11/10/2019] [Indexed: 12/15/2022]
Abstract
The discovery of antibiotics has led to the effective treatment of bacterial infections that were otherwise fatal and has had a transformative effect on modern medicine. Teixobactin is an unusual depsipeptide natural product that was recently discovered from a previously unculturable soil bacterium and found to possess potent antibacterial activity against several Gram positive pathogens, including methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococci. One of the key features of teixobactin as an antibiotic lead is that resistance could not be generated in a laboratory setting. This is proposed to be a result of a mechanism of action that involves binding to essential cell wall synthesis building blocks, lipid II and lipid III. Since the initial isolation report in 2015, significant efforts have been made to understand its unique mechanism of action, develop efficient synthetic routes for its production, and thus enable the generation of analogues for structure-activity relationship studies and optimization of its pharmacological properties. Our review provides a comprehensive treatise on the progress in understanding teixobactin chemistry, structure-activity relationships, and mechanisms of antibacterial activity. Teixobactin represents an exciting starting point for the development of new antibiotics that can be used to combat multidrug-resistant bacterial ("superbug") infections.
Collapse
Affiliation(s)
- John A Karas
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - Fan Chen
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - Elena K Schneider-Futschik
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia.,Lung Health Research Centre, Department of Pharmacology & Therapeutics, the University of Melbourne, Parkville, Victoria, Australia
| | - Zhisen Kang
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - Maytham Hussein
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - James Swarbrick
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - Daniel Hoyer
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia.,The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia.,Department of Molecular Medicine, the Scripps Research Institute, La Jolla, California
| | - Andrew M Giltrap
- School of Chemistry, the University of Sydney, Sydney, New South Wales, Australia
| | - Richard J Payne
- School of Chemistry, the University of Sydney, Sydney, New South Wales, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
25
|
Galván Márquez IJ, McKay B, Wong A, Cheetham JJ, Bean C, Golshani A, Smith ML. Mode of action of nisin on Escherichia coli. Can J Microbiol 2019; 66:161-168. [PMID: 31743042 DOI: 10.1139/cjm-2019-0315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nisin is a class I polycyclic bacteriocin produced by the bacterium Lactococcus lactis, which is used extensively as a food additive to inhibit the growth of foodborne Gram-positive bacteria. Nisin also inhibits growth of Gram-negative bacteria when combined with membrane-disrupting chelators such as citric acid. To gain insight into nisin's mode of action, we analyzed chemical-genetic interactions and identified nisin-sensitive Escherichia coli strains in the Keio library of knockout mutants. The most sensitive mutants fell into two main groups. The first group accords with the previously proposed mode of action based on studies with Gram-positive bacteria, whereby nisin interacts with factors involved in cell wall, membrane, envelope biogenesis. We identified an additional, novel mode of action for nisin based on the second group of sensitive mutants that involves cell cycle and DNA replication, recombination, and repair. Further analyses supported these two distinct modes of action.
Collapse
Affiliation(s)
- Imelda J Galván Márquez
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.,Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Bruce McKay
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.,Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Alex Wong
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.,Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - James J Cheetham
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.,Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Cody Bean
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.,Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Ashkan Golshani
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.,Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Myron L Smith
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.,Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
26
|
Pietrysiak E, Smith S, Ganjyal GM. Food Safety Interventions to Control
Listeria monocytogenes
in the Fresh Apple Packing Industry: A Review. Compr Rev Food Sci Food Saf 2019; 18:1705-1726. [DOI: 10.1111/1541-4337.12496] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 05/13/2019] [Accepted: 07/29/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Ewa Pietrysiak
- School of Food Science Washington State Univ. P.O. Box 646376 Pullman WA 99164‐6376 U.S.A
| | - Stephanie Smith
- School of Food Science Washington State Univ. P.O. Box 646376 Pullman WA 99164‐6376 U.S.A
| | - Girish M Ganjyal
- School of Food Science Washington State Univ. P.O. Box 646376 Pullman WA 99164‐6376 U.S.A
| |
Collapse
|
27
|
Malin JJ, de Leeuw E. Therapeutic compounds targeting Lipid II for antibacterial purposes. Infect Drug Resist 2019; 12:2613-2625. [PMID: 31692545 PMCID: PMC6711568 DOI: 10.2147/idr.s215070] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022] Open
Abstract
Resistance against commonly used antibiotics has emerged in all bacterial pathogens. In fact, there is no antibiotic currently in clinical use against which resistance has not been reported. In particular, rapidly increasing urbanization in developing nations are sites of major concern. Additionally, the widespread practice by physicians to prescribe antibiotics in cases of viral infections puts selective pressure on antibiotics that still remain effective and it will only be a matter of time before resistance develops on a large scale. The biosynthesis pathway of the bacterial cell wall is well studied and a validated target for the development of antibacterial agents. Cell wall biosynthesis involves two major processes; 1) the biosynthesis of cell wall teichoic acids and 2) the biosynthesis of peptidoglycan. Key molecules in these pathways, including enzymes and precursor molecules are attractive targets for the development of novel antibacterial agents. In this review, we will focus on the major class of natural antibacterial compounds that target the peptidoglycan precursor molecule Lipid II; namely the glycopeptides, including the novel generation of lipoglycopeptides. We will discuss their mechanism-of-action and clinical applications. Further, we will briefly discuss additional peptides that target Lipid II such as the lantibiotic nisin and defensins. We will highlight recent developments and future perspectives.
Collapse
Affiliation(s)
- Jakob J Malin
- University of Cologne, Department I of Internal Medicine, Division of Infectious Diseases, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Erik de Leeuw
- Institute of Human Virology and Department of Molecular Biology & Biochemistry of the University of Maryland, Baltimore School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
28
|
Nagendra Prasad HS, Karthik CS, Manukumar HM, Mallesha L, Mallu P. New approach to address antibiotic resistance: Miss loading of functional membrane microdomains (FMM) of methicillin-resistant Staphylococcus aureus (MRSA). Microb Pathog 2018; 127:106-115. [PMID: 30503959 DOI: 10.1016/j.micpath.2018.11.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 12/13/2022]
Abstract
The synthesized potent piperazine analog ChDiPiCa was characterised by various spectroscopic techniques and for the first time evaluated functional membrane microdomain (FMM) disassembly in methicillin-resistant Staphylococcus aureus (MRSA). The ChDiPiCa showed excellent in vitro biocidal activity against MRSA at 26 μg/mL compared to the antibiotic streptomycin and bacitracin 14 μg/mL and 13 μg/mL at 10 μg concentration respectively. The membrane damaging property was confirmed by the SEM analysis. Further, we addressed the new approach for the first time to overcome antibiotic resistance of MRSA through membrane microdomain miss loading to lipids. By which, the ChDiPiCa confirms the significant activity in miss loading of FMM of MRSA which is validated by the fatty acid profile and lipid analysis. The result shows that, altered saturated (Lauric acid and Myristic acid), mono unsaturated (Oleic acid), and poly unsaturated (Linoleic acid and Linolenic acid) fatty acids and hypothesises, altered the membrane functional lipids. For the better understanding of miss loading of FMM by the ChDiPiCa, the in-silico molecular docking studies was analyzed and confirmed the predicted role. This suggests the way to develop ChDiPiCa in medicinal chemistry as anti-MRSA candidates and also this report opens up new window to treat microbial pathogens and infections.
Collapse
Affiliation(s)
- H S Nagendra Prasad
- Department of Chemistry, Sri Jayachamarajendra College of Engineering, JSS Science and Technology University, Mysuru, 570 006, Karnataka, India
| | - C S Karthik
- Department of Chemistry, Sri Jayachamarajendra College of Engineering, JSS Science and Technology University, Mysuru, 570 006, Karnataka, India
| | - H M Manukumar
- Department of Chemistry, Sri Jayachamarajendra College of Engineering, JSS Science and Technology University, Mysuru, 570 006, Karnataka, India
| | - L Mallesha
- PG Department of Chemistry, JSS College of Arts, Commerce and Science, Mysuru, 570025, Karnataka, India
| | - P Mallu
- Department of Chemistry, Sri Jayachamarajendra College of Engineering, JSS Science and Technology University, Mysuru, 570 006, Karnataka, India.
| |
Collapse
|
29
|
Wu Y, An J, Liu Y, Wang Y, Ren W, Fang Z, Sun L, Gooneratne R. Mode of action of a novel anti-Listeria bacteriocin (CAMT2) produced by Bacillus amyloliquefaciens ZJHD3-06 from Epinephelus areolatus. Arch Microbiol 2018; 201:61-66. [PMID: 30203187 DOI: 10.1007/s00203-018-1553-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 10/28/2022]
Abstract
Bacteriocin CAMT2, produced by Bacillus amyloliquefaciens ZJHD3-06, has been shown to exhibit protective activity against important food spoilage and food-borne bacterial pathogens. This study was conducted to investigate the mode of action of bacteriocin CAMT2 against highly pathogenic Listeria monocytogenes ATCC 19111. The addition of bacteriocin CAMT2 at 64 AU/ml inhibited L. monocytogenes ATCC 19111. An efflux of K+ ions, lactic acid dehydrogenase and an increase in extracellular electrical conductivity was observed in CAMT2-treated L. monocytogenes. Electron microscopy showed morphological alterations such as uneven cell surface, accumulation of cell debris and bacterial lysis. These results show that bacteriocin CAMT2 inhibit L. monocytogenes by increasing cell permeability and inducing membrane damage, hence it has the great application potentials in ensuring food safety.
Collapse
Affiliation(s)
- Yaqian Wu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, Zhanjiang, 524088, China
| | - Junying An
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, Zhanjiang, 524088, China
| | - Ying Liu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, Zhanjiang, 524088, China.
| | - Yaling Wang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, Zhanjiang, 524088, China
| | - Wenbin Ren
- Zhongkai University of Agriculture and Engineering, Guangzhou, 510230, China
| | - Zhijia Fang
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, Zhanjiang, 524088, China
| | - Lijun Sun
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, Zhanjiang, 524088, China.
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Lincoln University, Lincoln, Canterbury, 7647, New Zealand
| |
Collapse
|
30
|
Goyal C, Malik RK, Pradhan D. Purification and characterization of a broad spectrum bacteriocin produced by a selected Lactococcus lactis strain 63 isolated from Indian dairy products. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2018; 55:3683-3692. [PMID: 30150828 PMCID: PMC6098757 DOI: 10.1007/s13197-018-3298-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 10/06/2017] [Accepted: 06/17/2018] [Indexed: 10/28/2022]
Abstract
Lactococcus lactis subsp. lactis strain 63 was isolated from Indian dairy products, produced a bacteriocin with broad spectrum inhibition against several of food pathogens like Listeria monocytogenes and Bacillus cereus as well as Gram negative bacteria viz., E. coli, Yersinia, Citrobacter, Proteus, Enterobacter, Klebsiella and Serratia strains. Bacteriocin production was higher in GM-17 and MRS as compared to TYGE broth and enriched skim milk broth and reached the maximum level during the early stationary phase. The bacteriocin was purified by performing ammonium sulfate precipitation. The bacteriocin was able to survive 90 °C/10 min but not 100 °C/10 min. Complete inactivation of bacteriocin was observed after autoclaving. The bacteriocin maintained its activity over a wide range of pH (3-9). The antimicrobial compound produced by the isolate 63, was sensitive to papain, pepsin, trypsin and amylase but was resistant to detergents like SDS and urea. Tween 20, Tween-80 as well as Triton X-100 enhanced its activity. Since the treatment with proteolytic enzymes resulted in loss of activity, this shows that the proteinaceous nature of the antimicrobial substance. Tentative molecular weight of the bacteriocin was found to be between 3.5 and 5 kDa by Tricine SDS-PAGE. Finally, we confirmed the presence of gene for nisin, and the sequence thus obtained, was identical to the sequences previously described for nisin Z. Lactococcus lactis subsp. lactis 63 or its bacteriocin, which has a wide inhibitory spectrum, has the potential for use as a starter or protective culture in the manufacture of fermented products.
Collapse
Affiliation(s)
- Chhaya Goyal
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132 001 India
| | - R. K. Malik
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132 001 India
| | - Diwas Pradhan
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana 132 001 India
| |
Collapse
|
31
|
Punekar AS, Samsudin F, Lloyd AJ, Dowson CG, Scott DJ, Khalid S, Roper DI. The role of the jaw subdomain of peptidoglycan glycosyltransferases for lipid II polymerization. Cell Surf 2018; 2:54-66. [PMID: 30046666 PMCID: PMC6053601 DOI: 10.1016/j.tcsw.2018.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/12/2018] [Accepted: 06/12/2018] [Indexed: 12/27/2022] Open
Abstract
Bacterial peptidoglycan glycosyltransferases (PGT) catalyse the essential polymerization of lipid II into linear glycan chains required for peptidoglycan biosynthesis. The PGT domain is composed of a large head subdomain and a smaller jaw subdomain and can be potently inhibited by the antibiotic moenomycin A (MoeA). We present an X-ray structure of the MoeA-bound Staphylococcus aureus monofunctional PGT enzyme, revealing electron density for a second MoeA bound to the jaw subdomain as well as the PGT donor site. Isothermal titration calorimetry confirms two drug-binding sites with markedly different affinities and positive cooperativity. Hydrophobic cluster analysis suggests that the membrane-interacting surface of the jaw subdomain has structural and physicochemical properties similar to amphipathic cationic α -helical antimicrobial peptides for lipid II recognition and binding. Furthermore, molecular dynamics simulations of the drug-free and -bound forms of the enzyme demonstrate the importance of the jaw subdomain movement for lipid II selection and polymerization process and provide molecular-level insights into the mechanism of peptidoglycan biosynthesis by PGTs.
Collapse
Affiliation(s)
- Avinash S. Punekar
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Firdaus Samsudin
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Adrian J. Lloyd
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| | | | - David J. Scott
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Leicestershire LE12 5RD, United Kingdom
- ISIS Neutron and Muon Spallation Source and Research Complex at Harwell, Rutherford Appleton Laboratory, Oxfordshire, United Kingdom
| | - Syma Khalid
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - David I. Roper
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom
| |
Collapse
|
32
|
Fernández MV, Agüero MV, Jagus RJ. Green tea extract: A natural antimicrobial with great potential for controlling native microbiota,
Listeria innocua
and
Escherichia coli
in fresh‐cut beet leaves. J Food Saf 2017. [DOI: 10.1111/jfs.12374] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- María Verónica Fernández
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científica y Técnicas (CONICET), Instituto de Tecnologías y Ciencias de la Ingeniería (INTECIN), Facultad de IngenieríaBuenos Aires Argentina
- Departamento de Ingeniería Química, Laboratorio de Microbiología Industrial: Tecnología de alimentosBuenos Aires Argentina
- Peruilh Foundation, Facultad de Ingeniería, Universidad de Buenos AiresBuenos Aires Argentina
| | - María Victoria Agüero
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científica y Técnicas (CONICET), Instituto de Tecnologías y Ciencias de la Ingeniería (INTECIN), Facultad de IngenieríaBuenos Aires Argentina
- Departamento de Ingeniería Química, Laboratorio de Microbiología Industrial: Tecnología de alimentosBuenos Aires Argentina
| | - Rosa Juana Jagus
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científica y Técnicas (CONICET), Instituto de Tecnologías y Ciencias de la Ingeniería (INTECIN), Facultad de IngenieríaBuenos Aires Argentina
- Departamento de Ingeniería Química, Laboratorio de Microbiología Industrial: Tecnología de alimentosBuenos Aires Argentina
| |
Collapse
|
33
|
Klahn P, Brönstrup M. Bifunctional antimicrobial conjugates and hybrid antimicrobials. Nat Prod Rep 2017; 34:832-885. [PMID: 28530279 DOI: 10.1039/c7np00006e] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Covering: up to the end of 2016Novel antimicrobial drugs are continuously needed to counteract bacterial resistance development. An innovative molecular design strategy for novel antibiotic drugs is based on the hybridization of an antibiotic with a second functional entity. Such conjugates can be grouped into two major categories. In the first category (antimicrobial hybrids), both functional elements of the hybrid exert antimicrobial activity. Due to the dual targeting, resistance development can be significantly impaired, the pharmacokinetic properties can be superior compared to combination therapies with the single antibiotics, and the antibacterial potency is often enhanced in a synergistic manner. In the second category (antimicrobial conjugates), one functional moiety controls the accumulation of the other part of the conjugate, e.g. by mediating an active transport into the bacterial cell or blocking the efflux. This approach is mostly applied to translocate compounds across the cell envelope of Gram-negative bacteria through membrane-embedded transporters (e.g. siderophore transporters) that provide nutrition and signalling compounds to the cell. Such 'Trojan Horse' approaches can expand the antibacterial activity of compounds against Gram-negative pathogens, or offer new options for natural products that could not be developed as standalone antibiotics, e.g. due to their toxicity.
Collapse
Affiliation(s)
- P Klahn
- Department for Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany. and Institute for Organic Chemistry, Technische Universität Braunschweig, Hagenring 30, 38106 Braunschweig, Germany.
| | - M Brönstrup
- Department for Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany.
| |
Collapse
|
34
|
Ge X, Teng K, Wang J, Zhao F, Zhang J, Zhong J. Identification of Key Residues in the NisK Sensor Region for Nisin Biosynthesis Regulation. Front Microbiol 2017; 8:106. [PMID: 28184221 PMCID: PMC5266694 DOI: 10.3389/fmicb.2017.00106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/16/2017] [Indexed: 11/13/2022] Open
Abstract
Histidine kinase (HK) NisK is well known to sense lantibiotic nisin for regulating the biosynthesis of nisin. NisK possesses two trans-membrane segments and a large extracellular region and nisin contains 34 amino acids with five lanthionine rings. Unlike most peptide sensing HK with multi trans-membrane segments, NisK is a representative of a group of rarely reported HK that sense peptide as ligand. To reveal how NisK senses nisin molecule to regulate nisin biosynthesis, we constructed a reporter Lactococcus lactis strain with nisRK constitutively expressed and a reporter gene lacZ expressed under the control of promoter P nisA . We showed that the extracellular region of NisK was involved in recognizing nisin. Conserved residues in this group of HK were found in the extracellular region of NisK and mutagenesis of these residues in the reporter strain revealed that several hydrophobic residues including two aromatic residues are crucial for NisK sensing nisin and regulating nisin biosynthesis. Substitutions of hydrophobic regions in NisK extracellular domain showed that the first strand that was rich of hydrophobic amino acids was involved in regulating nisin biosynthesis. A negatively charged residue in the first βstrand also contributed to nisin biosynthesis. Protein binding analyses demonstrated that nisin could not interact with key NisK mutants, indicating these site in the extracellular region of NisK was involved in recognizing nisin.
Collapse
Affiliation(s)
- Xiaoxuan Ge
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijing, China; University of Chinese Academy of SciencesBeijing, China
| | - Kunling Teng
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences Beijing, China
| | - Jian Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences Beijing, China
| | - Fangyuan Zhao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijing, China; University of Chinese Academy of SciencesBeijing, China
| | - Jie Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijing, China; University of Chinese Academy of SciencesBeijing, China
| | - Jin Zhong
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijing, China; University of Chinese Academy of SciencesBeijing, China
| |
Collapse
|
35
|
Aloui H, Khwaldia K. Natural Antimicrobial Edible Coatings for Microbial Safety and Food Quality Enhancement. Compr Rev Food Sci Food Saf 2016; 15:1080-1103. [DOI: 10.1111/1541-4337.12226] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/27/2016] [Accepted: 08/04/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Hajer Aloui
- Laboratoire des Substances Naturelles (LSN, LR10 INRAP02), Inst. Natl. de Recherche et d'Analyse Physico-chimique (INRAP); Pôle Technologique de Sidi Thabet; 2020 Sidi Thabet Tunisia
| | - Khaoula Khwaldia
- Laboratoire des Substances Naturelles (LSN, LR10 INRAP02), Inst. Natl. de Recherche et d'Analyse Physico-chimique (INRAP); Pôle Technologique de Sidi Thabet; 2020 Sidi Thabet Tunisia
| |
Collapse
|
36
|
Ahire JJ, Dicks LMT. Nisin Incorporated With 2,3-Dihydroxybenzoic Acid in Nanofibers Inhibits Biofilm Formation by a Methicillin-Resistant Strain of Staphylococcus aureus. Probiotics Antimicrob Proteins 2016; 7:52-9. [PMID: 25319566 DOI: 10.1007/s12602-014-9171-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The aim of the present study was to determine the effect of nisin, 2,3-dihydroxybenzoic acid (DHBA) and a combination of nisin and DHBA incorporated into nanofibers prepared from poly(D,L-lactide) (PDLLA) and poly(ethylene oxide) (PEO) on biofilm formation of a methicillin-resistant strain of Staphylococcus aureus (strain Xen 31). Biofilm formation decreased by 88% after 24 h of exposure to nanofibers containing nisin and DHBA (NDF), compared to a 63% decrease when exposed to nanofibers containing only DHBA (DF) and a 3% decrease when exposed to nanofibers containing only nisin (NF). Planktonic cell numbers of biofilms exposed to nanofibers without nisin or DHBA (CF) and NF increased from no detectable OD(595nm) readings to 0.35 and 0.3, respectively, within the first 8 h of exposure, followed by a steady decline over the following 16 h. Planktonic cells of biofilms treated with DF increased from no detectable OD(595nm) readings to 0.05 after 8 h of exposure and remained more-or-less constant for the duration of the experiment. Planktonic cells of biofilms exposed to NDF increased from OD(595nm) 0.03 after 8 h of exposure and to 0.2 over the following 16 h. Biofilm formation increased with increasing concentrations of FeCl3·6H2O, which suggests that iron is required for S. aureus Xen 31 to form a biofilm. However, when exposed to NDF, biofilm formation decreased significantly in the presence of increasing concentrations of iron. This suggests that NDF may be used to prevent biofilm formation of MRSA and control infection.
Collapse
Affiliation(s)
- Jayesh J Ahire
- Department of Microbiology, University of Stellenbosch, Private Bag X1, Matieland, 7602, Stellenbosch, South Africa
| | | |
Collapse
|
37
|
Ng V, Chan WC. New Found Hope for Antibiotic Discovery: Lipid II Inhibitors. Chemistry 2016; 22:12606-16. [PMID: 27388768 DOI: 10.1002/chem.201601315] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Indexed: 12/14/2022]
Abstract
Research into antibacterial agents has recently gathered pace in light of the disturbing crisis of antimicrobial resistance. The development of modern tools offers the opportunity of reviving the fallen era of antibacterial discovery through uncovering novel lead compounds that target vital bacterial cell components, such as lipid II. This paper provides a summary of the role of lipid II as well as an overview and insight into the structural features of macrocyclic peptides that inhibit this bacterial cell wall component. The recent discovery of teixobactin, a new class of lipid II inhibitor has generated substantial research interests. As such, the significant progress that has been achieved towards its development as a promising antibacterial agent is discussed.
Collapse
Affiliation(s)
- Vivian Ng
- School of Pharmacy, Centre of Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Weng C Chan
- School of Pharmacy, Centre of Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| |
Collapse
|
38
|
Zhou L, van Heel AJ, Montalban-Lopez M, Kuipers OP. Potentiating the Activity of Nisin against Escherichia coli. Front Cell Dev Biol 2016; 4:7. [PMID: 26904542 PMCID: PMC4745983 DOI: 10.3389/fcell.2016.00007] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 01/18/2016] [Indexed: 11/13/2022] Open
Abstract
Lantibiotics are antimicrobial (methyl)lanthionine-containing peptides produced by various Gram-positive bacteria. The model lantibiotic, nisin, binds lipid II in the cell membrane. Additionally, after binding it can insert into the membrane creating a pore. Nisin can efficiently inhibit the growth of Gram-positive bacteria and resistance is rarely observed. However, the activity of lantibiotics is at least 100-fold lower against certain Gram-negative bacteria. This is caused by the fact that Gram-negative bacteria have an outer membrane that hinders the peptides to reach lipid II, which is located in the inner membrane. Improving the activity of lantibiotics against Gram-negative bacteria could be achieved if the outer membrane traversing efficiency is increased. Here, several anti-Gram-negative peptides (e.g., apidaecin 1b, oncocin), or parts thereof, were fused to the C-terminus of either a truncated version of nisin containing the first three/five rings or full length nisin. The activities of these fusion peptides were tested against Gram-negative pathogens. Our results showed that when an eight amino acids (PRPPHPRL) tail from apidaecin 1b was attached to nisin, the activity of nisin against Escherichia coli CECT101 was increased more than two times. This research presents a new and promising method to increase the anti-Gram-negative activity of lantibiotics.
Collapse
Affiliation(s)
- Liang Zhou
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen Groningen, Netherlands
| | - Auke J van Heel
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen Groningen, Netherlands
| | - Manuel Montalban-Lopez
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen Groningen, Netherlands
| | - Oscar P Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen Groningen, Netherlands
| |
Collapse
|
39
|
Lactocin 160, a Bacteriocin Produced by Vaginal Lactobacillus rhamnosus, Targets Cytoplasmic Membranes of the Vaginal Pathogen, Gardnerella vaginalis. Probiotics Antimicrob Proteins 2016; 1:67-74. [PMID: 20445810 DOI: 10.1007/s12602-008-9003-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Bacterial vaginosis (BV) is a commonly occurring vaginal infection that is associated with a variety of serious risks related to the reproductive health of women. Conventional antibiotic treatment for this condition is frequently ineffective because the antibiotics tend to inhibit healthy vaginal microflora along with the pathogens. Lactocin 160, a bacteriocin produced by healthy vaginal lactobacilli, is a promising alternative to antibiotics; this compound specifically inhibits the BV-associated vaginal pathogens such as Gardnerella vaginalis and Prevotella bivia without affecting the healthy microflora. This study investigates the molecular mechanism of action for lactocin 160 and reveals that this compound targets the cytoplasmic membrane of G. vaginalis, causing the efflux of ATP molecules and dissipation of the proton motive force.
Collapse
|
40
|
Roces C, Rodríguez A, Martínez B. Cell Wall-active Bacteriocins and Their Applications Beyond Antibiotic Activity. Probiotics Antimicrob Proteins 2016; 4:259-72. [PMID: 26782186 DOI: 10.1007/s12602-012-9116-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Microorganisms synthesize several compounds with antimicrobial activity in order to compete or defend themselves against others and ensure their survival. In this line, the cell wall is a major protective barrier whose integrity is essential for many vital bacterial processes. Probably for this reason, it represents a 'hot spot' as a target for many antibiotics and antimicrobial peptides such as bacteriocins. Bacteriocins have largely been recognized by their pore-forming ability that collapses the selective permeability of the cytoplasmic membrane. However, in the last few years, many bacteriocins have been shown to inhibit cell wall biosyntheis alone, or in a concerted action with pore formation like nisin. Examples of cell wall-active bacteriocins are found in both Gram-negative and Gram-positive bacteria and include a wide diversity of structures such as nisin-like and mersacidin-like lipid II-binding bacteriocins, two-peptide lantibiotics, and non-modified bacteriocins. In this review, we summarize the current knowledge on these antimicrobial peptides as well as the role, composition, and biosynthesis of the bacterial cell wall as their target. Moreover, even though bacteriocins have been a matter of interest as natural food antimicrobials, we propose them as suitable tools to provide new means to improve biotechnologically relevant microorganisms.
Collapse
Affiliation(s)
- Clara Roces
- DairySafe Group, Department of Technology and Biotechnology of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Paseo Río Linares s/n., 33300, Villaviciosa, Asturias, Spain
| | - Ana Rodríguez
- DairySafe Group, Department of Technology and Biotechnology of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Paseo Río Linares s/n., 33300, Villaviciosa, Asturias, Spain
| | - Beatriz Martínez
- DairySafe Group, Department of Technology and Biotechnology of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Paseo Río Linares s/n., 33300, Villaviciosa, Asturias, Spain.
| |
Collapse
|
41
|
Optimization of the antioxidant and antimicrobial response of the combined effect of nisin and avocado byproducts. Lebensm Wiss Technol 2016. [DOI: 10.1016/j.lwt.2015.07.048] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
42
|
Klahn P, Brönstrup M. New Structural Templates for Clinically Validated and Novel Targets in Antimicrobial Drug Research and Development. Curr Top Microbiol Immunol 2016; 398:365-417. [PMID: 27704270 DOI: 10.1007/82_2016_501] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The development of bacterial resistance against current antibiotic drugs necessitates a continuous renewal of the arsenal of efficacious drugs. This imperative has not been met by the output of antibiotic research and development of the past decades for various reasons, including the declining efforts of large pharma companies in this area. Moreover, the majority of novel antibiotics are chemical derivatives of existing structures that represent mostly step innovations, implying that the available chemical space may be exhausted. This review negates this impression by showcasing recent achievements in lead finding and optimization of antibiotics that have novel or unexplored chemical structures. Not surprisingly, many of the novel structural templates like teixobactins, lysocin, griselimycin, or the albicidin/cystobactamid pair were discovered from natural sources. Additional compounds were obtained from the screening of synthetic libraries and chemical synthesis, including the gyrase-inhibiting NTBI's and spiropyrimidinetrione, the tarocin and targocil inhibitors of wall teichoic acid synthesis, or the boronates and diazabicyclo[3.2.1]octane as novel β-lactamase inhibitors. A motif that is common to most clinically validated antibiotics is that they address hotspots in complex biosynthetic machineries, whose functioning is essential for the bacterial cell. Therefore, an introduction to the biological targets-cell wall synthesis, topoisomerases, the DNA sliding clamp, and membrane-bound electron transport-is given for each of the leads presented here.
Collapse
Affiliation(s)
- Philipp Klahn
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany.
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany.
| |
Collapse
|
43
|
Tai HF, Foo HL, Abdul Rahim R, Loh TC, Abdullah MP, Yoshinobu K. Molecular characterisation of new organisation of plnEF and plw loci of bacteriocin genes harbour concomitantly in Lactobacillus plantarum I-UL4. Microb Cell Fact 2015; 14:89. [PMID: 26077560 PMCID: PMC4467070 DOI: 10.1186/s12934-015-0280-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 06/02/2015] [Indexed: 12/05/2022] Open
Abstract
Background Bacteriocin-producing Lactic acid bacteria (LAB) have vast applications in human and animal health, as well as in food industry. The structural, immunity, regulatory, export and modification genes are required for effective bacteriocin biosynthesis. Variations in gene sequence, composition and organisation will affect the antimicrobial spectrum of bacteriocin greatly. Lactobacillus plantarum I-UL4 is a novel multiple bacteriocin producer that harbours both plw and plnEF structural genes simultaneous which has not been reported elsewhere. Therefore, molecular characterisation of bacteriocin genes that harboured in L. plantarum I-UL4 was conducted in this study. Results and discussion Under optimised conditions, 8 genes (brnQ1, napA1, plnL, plnD, plnEF, plnI, plnG and plnH) of plnEF locus and 2 genes (plw and plwG) of plw locus were amplified successfully from genomic DNA extracted from L. plantarum I-UL4 using specific primers designed from 24 pln genes selected randomly from reported plw, plS, pln423 and plnEF loci. DNA sequence analysis of the flanking region of the amplified genes revealed the presence of two pln loci, UL4-plw and UL4-plnEF loci, which were chromosomally encoded as shown by Southern hybridisation. UL4-plw locus that contained three ORFs were arranged in one operon and possessed remarkable amino acid sequence of LMG2379-plw locus, suggesting it was highly conserved. Interestingly, the UL4-plnEF locus appeared to be a composite pln locus of JDM1-plnEF and J51-plnEF locus in terms of genetic composition and organisation, whereby twenty complete and one partial open reading frames (ORFs) were aligned and organised successfully into five operons. Furthermore, a mutation was detected in plnF structural gene which has contributed to a longer bacteriocin peptide. Conclusions Plantaricin EF and plantaricin W encoded by plnEF and plnW loci are classified as class I bacteriocin and class II bacteriocin molecules respectively. The concurrent presence of two pln loci encoding bacteriocins from two different classes has contributed greatly to the broad inhibitory spectrum of L. plantarum I-UL4. The new genetic composition and organisation of plnEF locus and concurrent presence of plnEF and plnW loci indicated that L. plantarum I-UL4 is a novel multiple bacteriocin producer that possesses vast potentials in various industries.
Collapse
Affiliation(s)
- Hui Fong Tai
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Hooi Ling Foo
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia. .,Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Raha Abdul Rahim
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia. .,Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Teck Chewn Loh
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia. .,Institute of Tropical Agriculture, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Mohd Puad Abdullah
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia. .,Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia. .,Institute of Tropical Agriculture, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Kimura Yoshinobu
- Department of Biofunctional Chemistry, Graduate School of Environmental and Life Sciemce, Okayama University, Okayama, Japan.
| |
Collapse
|
44
|
Escano J, Smith L. Multipronged approach for engineering novel peptide analogues of existing lantibiotics. Expert Opin Drug Discov 2015; 10:857-70. [PMID: 26004576 DOI: 10.1517/17460441.2015.1049527] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Lantibiotics are a class of ribosomally and post-translationally modified peptide antibiotics that are active against a broad spectrum of Gram-positive bacteria. Great efforts have been made to promote the production of these antibiotics, so that they can one day be used in our antimicrobial arsenal to combat multidrug-resistant bacterial infections. AREAS COVERED This review provides a synopsis of lantibiotic research aimed at furthering our understanding of the structural limitation of lantibiotics as well as identifying structural regions that can be modified to improve the bioactivity. In vivo, in vitro and chemical synthesis of lantibiotics has been useful for engineering novel variants with enhanced activities. These approaches have provided novel ways to further our understanding of lantibiotic function and have advanced the objective to develop lantibiotics for the treatment of infectious diseases. EXPERT OPINION Synthesis of lantibiotics with enhanced activities will lead to the discovery of new promising drug candidates that will have a long lasting impact on the treatment of Gram-positive infections. The current body of literature for producing structural variants of lantibiotics has been more of a 'proof-of-principle' approach and the application of these methods has not yet been fully utilized.
Collapse
Affiliation(s)
- Jerome Escano
- Texas A&M University, Department of Biological Sciences, College Station , TX 77843 , USA
| | | |
Collapse
|
45
|
Fletcher S, Yu W, Huang J, Kwasny SM, Chauhan J, Opperman TJ, MacKerell AD, de Leeuw EPH. Structure-activity exploration of a small-molecule Lipid II inhibitor. Drug Des Devel Ther 2015; 9:2383-94. [PMID: 25987836 PMCID: PMC4422293 DOI: 10.2147/dddt.s79504] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We have recently identified low-molecular weight compounds that act as inhibitors of Lipid II, an essential precursor of bacterial cell wall biosynthesis. Lipid II comprises specialized lipid (bactoprenol) linked to a hydrophilic head group consisting of a peptidoglycan subunit (N-acetyl glucosamine [GlcNAc]–N-acetyl muramic acid [MurNAc] disaccharide coupled to a short pentapeptide moiety) via a pyrophosphate. One of our lead compounds, a diphenyl-trimethyl indolene pyrylium, termed BAS00127538, interacts with the MurNAc moiety and the isoprenyl tail of Lipid II. Here, we report on the structure–activity relationship of BAS00127538 derivatives obtained by in silico analyses and de novo chemical synthesis. Our results indicate that Lipid II binding and bacterial killing are related to three features: the diphenyl moiety, the indolene moiety, and the positive charge of the pyrylium. Replacement of the pyrylium moiety with an N-methyl pyridinium, which may have importance in stability of the molecule, did not alter Lipid II binding or antibacterial potency.
Collapse
Affiliation(s)
- Steven Fletcher
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, MD, USA
| | - Wenbo Yu
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, MD, USA ; Computer-Aided Drug Design Center, University of Maryland, School of Pharmacy, Baltimore, MD, USA
| | - Jing Huang
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, MD, USA ; Computer-Aided Drug Design Center, University of Maryland, School of Pharmacy, Baltimore, MD, USA
| | | | - Jay Chauhan
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, MD, USA
| | | | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, University of Maryland, School of Pharmacy, Baltimore, MD, USA ; Computer-Aided Drug Design Center, University of Maryland, School of Pharmacy, Baltimore, MD, USA
| | - Erik P H de Leeuw
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
46
|
|
47
|
Patra M, Wenzel M, Prochnow P, Pierroz V, Gasser G, Bandow JE, Metzler-Nolte N. An organometallic structure-activity relationship study reveals the essential role of a Re(CO) 3 moiety in the activity against gram-positive pathogens including MRSA. Chem Sci 2015; 6:214-224. [PMID: 28553471 PMCID: PMC5433042 DOI: 10.1039/c4sc02709d] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/24/2014] [Indexed: 01/30/2023] Open
Abstract
The worrying appearance of microbial resistance to antibiotics is a worldwide problem which needs to be tackled urgently. Microbial resistance to the common classes of antibiotics involving purely organic compounds unfortunately develops very rapidly and in most cases, resistance was detected soon after or even before release of the antibiotic to the market. Therefore, novel concepts for antibiotics must be investigated, and metal-containing compounds hold particular promise in that area. Taking a trimetallic complex (1a) which contains a ferrocenyl (Fc), a CpMn(CO)3 (cymantrene) and a [(dpa)Re(CO)3] residue as the lead structure, a systematic structure-activity relationship (SAR) study against various gram-positive pathogenic bacteria including methicillin-resistant Staphylococcus aureus (MRSA) strains was performed. The [(dpa)Re(CO)3] moiety was discovered to be the essential unit for the observed antibacterial activity of 1a. The ferrocenyl and CpMn(CO)3 units can be replaced one by one or both together by organic moieties such as a phenyl ring without loss of antibacterial activity. The most potent mono-metallic complex (9c') has an antibacterial activity comparable to the well-established organic drugs amoxicillin and norfloxacin and importantly, only moderate cytotoxicity against mammalian cells. Microbiological studies on membrane potential, membrane permeabilization, and cell wall integrity revealed that 9c' targets the bacterial membrane and disturbs cell wall integrity, but shows more efficient membrane permeabilization than the lead structure 1a.
Collapse
Affiliation(s)
- Malay Patra
- Lehrstuhl für Anorganische Chemie I - Bioanorganische Chemie , Fakultät für Chemie und Biochemie , Ruhr-Universität Bochum , Universitätsstrasse 150 , D-44801 Bochum , Germany .
| | - Michaela Wenzel
- Ruhr-Universität Bochum , Biologie der Mikroorganismen , Arbeitsgruppe Mikrobielle Antibiotikaforschung , Universitätsstrasse 150 , D-44801 Bochum , Germany . ; ; Tel: +49-234-32-23102
| | - Pascal Prochnow
- Ruhr-Universität Bochum , Biologie der Mikroorganismen , Arbeitsgruppe Mikrobielle Antibiotikaforschung , Universitätsstrasse 150 , D-44801 Bochum , Germany . ; ; Tel: +49-234-32-23102
| | - Vanessa Pierroz
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 Zurich , Switzerland
| | - Gilles Gasser
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 Zurich , Switzerland
| | - Julia E Bandow
- Ruhr-Universität Bochum , Biologie der Mikroorganismen , Arbeitsgruppe Mikrobielle Antibiotikaforschung , Universitätsstrasse 150 , D-44801 Bochum , Germany . ; ; Tel: +49-234-32-23102
| | - Nils Metzler-Nolte
- Lehrstuhl für Anorganische Chemie I - Bioanorganische Chemie , Fakultät für Chemie und Biochemie , Ruhr-Universität Bochum , Universitätsstrasse 150 , D-44801 Bochum , Germany .
| |
Collapse
|
48
|
LeBel G, Vaillancourt K, Frenette M, Gottschalk M, Grenier D. Suicin 90-1330 from a nonvirulent strain of Streptococcus suis: a nisin-related lantibiotic active on gram-positive swine pathogens. Appl Environ Microbiol 2014; 80:5484-92. [PMID: 24973067 PMCID: PMC4136082 DOI: 10.1128/aem.01055-14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/20/2014] [Indexed: 01/20/2023] Open
Abstract
Streptococcus suis serotype 2 is known to cause severe infections (meningitis, endocarditis, and septicemia) in pigs and is considered an emerging zoonotic agent. Antibiotics have long been used in the swine industry for disease treatment/prevention and growth promoters. This pattern of utilization resulted in the spread of antibiotic resistance in S. suis worldwide. Interestingly, pigs may harbor S. suis in their tonsils without developing diseases, while North American strains belonging to the sequence type 28 (ST28) are nonvirulent in animal models. Consequently, the aim of this study was to purify and characterize a bacteriocin produced by a nonvirulent strain of S. suis serotype 2, with a view to a potential therapeutic and preventive application. S. suis 90-1330 belonging to ST28 and previously shown to be nonvirulent in an animal model exhibited antibacterial activity toward all S. suis pathogenic isolates tested. The bacteriocin produced by this strain was purified to homogeneity by cationic exchange and reversed-phase fast protein liquid chromatography. Given its properties (molecular mass of <4 kDa, heat, pH and protease stability, and the presence of modified amino acids), the bacteriocin, named suicin 90-1330, belongs to the lantibiotic class. Using a DNA-binding fluorophore, the bacteriocin was found to possess a membrane permeabilization activity. When tested on other swine pathogens, the suicin showed activity against Staphylococcus hyicus and Staphylococcus aureus, whereas it was inactive against all Gram-negative bacteria tested. Amino acid sequencing of the purified bacteriocin showed homology (90.9% identity) with nisin U produced by Streptococcus uberis. The putative gene cluster involved in suicin production was amplified by PCR and sequence analysis revealed the presence of 11 open reading frames, including the structural gene and those required for the modification of amino acids, export, regulation, and immunity. Further studies will evaluate the ability of suicin 90-1330 or the producing strain to prevent experimental S. suis infections in pigs.
Collapse
Affiliation(s)
- Geneviève LeBel
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Quebec City, Quebec, Canada
| | - Katy Vaillancourt
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Quebec City, Quebec, Canada
| | - Michel Frenette
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Quebec City, Quebec, Canada Centre de Recherche en Infectiologie Porcine et Avicole, Fonds de Recherche du Québec-Nature et Technologies, Quebec City, Quebec, Canada
| | - Marcelo Gottschalk
- Centre de Recherche en Infectiologie Porcine et Avicole, Fonds de Recherche du Québec-Nature et Technologies, Quebec City, Quebec, Canada Groupe de Recherche sur les Maladies Infectieuses du Porc, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Daniel Grenier
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Quebec City, Quebec, Canada Centre de Recherche en Infectiologie Porcine et Avicole, Fonds de Recherche du Québec-Nature et Technologies, Quebec City, Quebec, Canada
| |
Collapse
|
49
|
Snyder AB, Worobo RW. Chemical and genetic characterization of bacteriocins: antimicrobial peptides for food safety. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2014; 94:28-44. [PMID: 23818338 DOI: 10.1002/jsfa.6293] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 06/24/2013] [Accepted: 07/01/2013] [Indexed: 05/16/2023]
Abstract
Antimicrobial peptides are produced across all domains of life. Among these diverse compounds, those produced by bacteria have been most successfully applied as agents of biocontrol in food and agriculture. Bacteriocins are ribosomally synthesized, proteinaceous compounds that inhibit the growth of closely related bacteria. Even within the subcategory of bacteriocins, the peptides vary significantly in terms of the gene cluster responsible for expression, and chemical and structural composition. The polycistronic gene cluster generally includes a structural gene and various combinations of immunity, secretion, and regulatory genes and modifying enzymes. Chemical variation can exist in amino acid identity, chain length, secondary and tertiary structural features, as well as specificity of active sites. This diversity posits bacteriocins as potential antimicrobial agents with a range of functions and applications. Those produced by food-grade bacteria and applied in normally occurring concentrations can be used as GRAS-status food additives. However, successful application requires thorough characterization.
Collapse
Affiliation(s)
- Abigail B Snyder
- Department of Food Science, Cornell University, Geneva, NY, 14456, USA
| | | |
Collapse
|
50
|
Effect of Combined Natural Antimicrobials on Spoilage Microorganisms and Listeria Innocua in a Whey Cheese “Ricotta”. FOOD BIOPROCESS TECH 2013. [DOI: 10.1007/s11947-013-1243-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|