51
|
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
|
52
|
Qiao Y, Qiu Z, Tian F, Yu L, Zhao J, Zhang H, Zhai Q, Chen W. Pediococcus acidilactici Strains Improve Constipation Symptoms and Regulate Intestinal Flora in Mice. Front Cell Infect Microbiol 2021; 11:655258. [PMID: 33816357 PMCID: PMC8012752 DOI: 10.3389/fcimb.2021.655258] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/23/2021] [Indexed: 01/03/2023] Open
Abstract
Constipation is a prevalent gastrointestinal disorder that seriously reduces the quality of life. Clinical studies have shown that a great change or severe imbalance occurs in the intestinal microbiota of people with constipation. This study explored whether bacteriocin-producing and non-bacteriocin-producing Pediococcus acidilactici strains resulted in differences in the alleviation of constipation and changes in the fecal flora in BALB/c mice. The constipation-related indicators, gastrointestinal regulatory peptides and gut microbiota were identified to evaluate their alleviating effects and underlying mechanisms. The time to the first black-stool defecation and the gastrointestinal transit rate in constipated mice were found to be somewhat improved by four P. acidilactici strains (P > 0.05). Moreover, there were significant differences in the level of most gastrointestinal regulatory peptides in the serum, as well as in the composition and abundance of intestinal microbiota in different groups (P < 0.05). At the phylum level, the relative abundance of Firmicutes was significantly increased, but those of Bacteroidetes and Proteobacteria were significantly reduced after the administration of four P. acidilactici strains for 14 d (P < 0.05). The levels of Bacteroides and genera from Enterobacteriaceae were significantly decreased, whereas Bifidobacterium and Lactobacillus were upregulated when bacteriocin-producing P. acidilactici CCFM18 and CCFM28 strains were provided in the diet (P < 0.05). The results indicated that although constipation-related symptoms were alleviated to only a limited degree, the administration of four P. acidilactici strains effectively regulated the gut flora and provided a potential health benefit to the host, especially the bacteriocin-producing P. acidilactici strains.
Collapse
Affiliation(s)
- Yiteng Qiao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhichang Qiu
- College of Food Science and Engineering, Shandong Agricultural University, Tai'an, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China.,Wuxi Translational Medicine Research Center, Jiangsu Translational Medicine, Research Institute Wuxi Branch, Wuxi, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
53
|
Jung S, Woo C, Fugaban JII, Vazquez Bucheli JE, Holzapfel WH, Todorov SD. Bacteriocinogenic Potential of Bacillus amyloliquefaciens Isolated from Kimchi, a Traditional Korean Fermented Cabbage. Probiotics Antimicrob Proteins 2021; 13:1195-1212. [PMID: 33721203 DOI: 10.1007/s12602-021-09772-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2021] [Indexed: 12/24/2022]
Abstract
Bacteriocin production is considered a favorable property for various beneficial cultures. In addition to their potential as biopreservatives, bacteriocins are also promising alternatives for the control of multidrug-resistant pathogens and the inhibition of some viruses and cancer cells. The objective of this study was to screen and characterize a bacteriocin-producing strain with the aim of its future application for control of Listeria monocytogenes, an important food-borne pathogen. A total of 22 potentially bacteriocinogenic strains active against L. monocytogenes ATCC15313 were isolated from locally produced kimchi through a three-level approach. Pure cultures were obtained according to good microbiological practices and differentiated through RAPD-PCR using the primers OPL01, OPL09, and OPL11. Altogether, 5 strains were selected for further study. Specific focus was given to strain ST05DL based on its specific inhibitory activity against L. monocytogenes ATCC15313, while not affecting different strains belonging to the genera Lactobacillus, Pediococcus, Leuconostoc, and Weissella, most of which are beneficial microorganisms. The strain ST05DL was identified as Bacillus amyloliquefaciens based on its sugar fermentation profile obtained through API50CHB analysis and 16S rRNA partial sequencing. The antimicrobial compound produced by B. amyloliquefaciens ST05DL was found to be sensitive to pepsin and α-chymotrypsin, evidence of its proteinaceous nature. The presence of skim milk, NaCl, Tween 80, glycerol, and SDS did not affect the antimicrobial activity. The addition of 20% cell-free supernatant (CFS) obtained from a 24-h culture of B. amyloliquefaciens ST05DL to an exponentially growing culture of L. monocytogenes ATCC15313 successfully inhibited the test microorganisms during the monitored 10-h incubation. Optimal bacteriocin production by B. amyloliquefaciens ST05DL was observed during the stationary phase at 12 h (800 AU/mL) and remained stable for the next 15 h. The ratio between live and dead cells during this period was 74.37% and 25.66%, respectively, as determined by flow cytometry. The presence of the virulence genes hblA, hblB, hblC, nheA, nheB, and nheC was not detected in the total DNA of B. amyloliquefaciens ST05DL, and the strain was resistant only to ampicillin out of 10 tested antibiotics. Future evaluation of expressed bacteriocin/s by B. amyloliquefaciens ST05DL (amino acid sequence, molecular mass, cytotoxicity, detailed mode of action, etc.), will be the next step in the characterization and its potential application as biopreservative and/or pharmaceutical product.
Collapse
Affiliation(s)
- Sungmin Jung
- ProBacLab, Graduate School of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Chaerin Woo
- ProBacLab, Graduate School of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Joanna Ivy Irorita Fugaban
- ProBacLab, Graduate School of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Jorge Enrique Vazquez Bucheli
- ProBacLab, Graduate School of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Wilhelm Heinrich Holzapfel
- ProBacLab, Graduate School of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Svetoslav Dimitrov Todorov
- ProBacLab, Graduate School of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea.
| |
Collapse
|
54
|
Safety evaluation and bacteriocinogenic potential of Pediococcus acidilactici strains isolated from artisanal cheeses. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2020.110550] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
55
|
Li Y, Hintze KJ, Ward RE. Effect of supplemental prebiotics, probiotics and bioactive proteins on the microbiome composition and fecal calprotectin in C57BL6/j mice. Biochimie 2021; 185:43-52. [PMID: 33609630 DOI: 10.1016/j.biochi.2021.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/11/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023]
Abstract
The composition and metabolic activity of the microbiome affect many aspects of health, and there is current interest in dietary constituents that may affect this system. The purpose of this study was to evaluate the effects of a mix of probiotics, a mix of prebiotics and a bioactive protein fraction on the microbiome, when fed to mice alone and in combination at physiologically relevant doses. Mice were fed the total western diet (TWD) supplemented with prebiotics, probiotics, and bioactive proteins individually and in combination for four weeks. Subsequently, effects on the composition of the gut microbiome, gut short-chain fatty acids (SCFAs) concentration, and gut inflammation were measured. Ruminococcus gnavus was increased in mice gut microbiome after feeding prebiotics. Bifidobacterium longum was increased after feeding probiotics. The treatments significantly affected beta-diversity with minor treatment effects on cecal or fecal SCFAs levels, and the treatments did not affect gut inflammation as measured by fecal calprotectin.
Collapse
Affiliation(s)
- Ye Li
- Nutrition, Dietetics and Food Sciences, Utah State University, 8700 Old Main Hill, Logan, UT, 84322-8700, USA
| | - Korry J Hintze
- Nutrition, Dietetics and Food Sciences, Utah State University, 8700 Old Main Hill, Logan, UT, 84322-8700, USA
| | - Robert E Ward
- Nutrition, Dietetics and Food Sciences, Utah State University, 8700 Old Main Hill, Logan, UT, 84322-8700, USA.
| |
Collapse
|
56
|
Lee JJ, Yong D, Suk KT, Kim DJ, Woo HJ, Lee SS, Kim BS. Alteration of Gut Microbiota in Carbapenem-Resistant Enterobacteriaceae Carriers during Fecal Microbiota Transplantation According to Decolonization Periods. Microorganisms 2021; 9:microorganisms9020352. [PMID: 33578974 PMCID: PMC7916679 DOI: 10.3390/microorganisms9020352] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Fecal microbiota transplantation (FMT) has been suggested as an alternative therapeutic option to decolonize carbapenem-resistant Enterobacteriaceae (CRE). However, the analysis of gut microbiota alteration in CRE carriers during FMT is still limited. Here, gut microbiota changes in CRE carriers were evaluated during FMT according to decolonization periods. The decolonization of 10 CRE carriers was evaluated after FMT, using serial consecutive rectal swab cultures. Alterations of gut microbiota before and after FMT (56 serial samples) were analyzed using high-throughput sequencing. The decolonization rates of CRE carriers were 40%, 50%, and 90% within 1, 3 and 5 months after initial FMT, respectively. Gut microbiota significantly changed after FMT (p = 0.003). Microbiota alteration was different between the early decolonization carriers (EDC) and late decolonization carriers (LDC). Microbiota convergence in carriers to donors was detected in EDC within 4 weeks, and keystone genera within the Bacteroidetes were found in the gut microbiota of EDC before FMT. The relative abundance of Klebsiella was lower in EDC than in LDC, before and after FMT. Our results indicate that FMT is a potential option for CRE decolonization. The gut microbiota of CRE carriers could be used to predict decolonization timing after FMT, and determine repeated FMT necessity.
Collapse
Affiliation(s)
- Jin-Jae Lee
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Korea;
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hallym University, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Korea
| | - Dong Joon Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hallym University, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Korea
| | - Heung-Jeong Woo
- Department of Internal Medicine, Division of Infectious Diseases, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong 18450, Korea;
| | - Seung Soon Lee
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
- Department of Internal Medicine, Division of Infectious Diseases, Hallym University Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon 24252, Korea
- Correspondence: (S.S.L.); (B.-S.K.)
| | - Bong-Soo Kim
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Korea;
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
- Correspondence: (S.S.L.); (B.-S.K.)
| |
Collapse
|
57
|
Yan H, Lu Y, Li X, Yi Y, Wang X, Shan Y, Liu B, Zhou Y, Lü X. Action mode of bacteriocin BM1829 against Escherichia coli and Staphylococcus aureus. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2020.100794] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
58
|
Soltani S, Hammami R, Cotter PD, Rebuffat S, Said LB, Gaudreau H, Bédard F, Biron E, Drider D, Fliss I. Bacteriocins as a new generation of antimicrobials: toxicity aspects and regulations. FEMS Microbiol Rev 2021; 45:fuaa039. [PMID: 32876664 PMCID: PMC7794045 DOI: 10.1093/femsre/fuaa039] [Citation(s) in RCA: 226] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
In recent decades, bacteriocins have received substantial attention as antimicrobial compounds. Although bacteriocins have been predominantly exploited as food preservatives, they are now receiving increased attention as potential clinical antimicrobials and as possible immune-modulating agents. Infections caused by antibiotic-resistant bacteria have been declared as a global threat to public health. Bacteriocins represent a potential solution to this worldwide threat due to their broad- or narrow-spectrum activity against antibiotic-resistant bacteria. Notably, despite their role in food safety as natural alternatives to chemical preservatives, nisin remains the only bacteriocin legally approved by regulatory agencies as a food preservative. Moreover, insufficient data on the safety and toxicity of bacteriocins represent a barrier against the more widespread use of bacteriocins by the food and medical industry. Here, we focus on the most recent trends relating to the application of bacteriocins, their toxicity and impacts.
Collapse
Affiliation(s)
- Samira Soltani
- Food Science Department, Faculty of Agriculture and Food Sciences, Université Laval, G1V 0A6 Québec, Canada
| | - Riadh Hammami
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, 75 Laurier Ave. E, Ottawa, ON K1N 6N5, Canada
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996 Ireland
- APC Microbiome Ireland, Institute and school of Microbiology, University College Cork, Western Road, Cork, T12 YN60, Ireland
| | - Sylvie Rebuffat
- Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Laboratory Molecules of Communication and Adaptation of Microorganisms (MCAM), UMR 7245 CNRS-MNHN, CP 54, 57 rue Cuvier, 75005 Paris, France
| | - Laila Ben Said
- Food Science Department, Faculty of Agriculture and Food Sciences, Université Laval, G1V 0A6 Québec, Canada
| | - Hélène Gaudreau
- Food Science Department, Faculty of Agriculture and Food Sciences, Université Laval, G1V 0A6 Québec, Canada
| | - François Bédard
- Faculty of Pharmacy and Centre de Recherche en Endocrinologie Moléculaire et Oncologique et Génomique Humaine, Université Laval, 2705 Boulevard Laurier, Quebec G1V 4G2, Canada
| | - Eric Biron
- Faculty of Pharmacy and Centre de Recherche en Endocrinologie Moléculaire et Oncologique et Génomique Humaine, Université Laval, 2705 Boulevard Laurier, Quebec G1V 4G2, Canada
| | - Djamel Drider
- Institut Charles Viollette, Université de Lille, EA 7394, 53955 Villeneuve d'Ascq, France
| | - Ismail Fliss
- Food Science Department, Faculty of Agriculture and Food Sciences, Université Laval, G1V 0A6 Québec, Canada
- Institute of Nutrition and Functional Foods, Université Laval, 2440 Boulevard Hochelaga, Québec G1V 0A6, Canada
| |
Collapse
|
59
|
García-Curiel L, Del Rocío López-Cuellar M, Rodríguez-Hernández AI, Chavarría-Hernández N. Toward understanding the signals of bacteriocin production by Streptococcus spp. and their importance in current applications. World J Microbiol Biotechnol 2021; 37:15. [PMID: 33394178 DOI: 10.1007/s11274-020-02973-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/01/2020] [Indexed: 12/25/2022]
Abstract
Microorganisms have developed quorum sensing (QS) systems to detect small signaling molecules that help to control access to additional nutrients and space in highly competitive polymicrobial niches. Many bacterial processes are QS-regulated; two examples are the highly related traits of the natural genetic competence state and the production of antimicrobial peptides such as bacteriocins. The Streptococcus genus is widely studied for its competence and for its ability to produce bacteriocins, as these antimicrobial peptides have significant potential in the treatment of infections caused by multiple-resistant pathogens, a severe public health issue. The transduction of a two-component system controls competence in streptococci: (1) ComD/E, which controls the competence in the Mitis and Anginosus groups, and (2) ComR/S, which performs the same function in the Bovis, Mutans, Salivarius, and Pyogenic groups. The cell-to-cell communication required for bacteriocin production in the Streptococcus groups is controlled mainly by a paralog of the ComD/E system. The relationships between pheromone signals and induction pathways are related to the bacteriocin production systems. In this review, we discuss the recent advances in the understanding of signaling and the induction of bacteriocin biosynthesis by QS regulation in streptococci. This information could aid in the design of better methods for the development and production of these antimicrobial peptides. It could also contribute to the analysis and emerging applications of bacteriocins in terms of their safety, quality, and human health benefits.
Collapse
Affiliation(s)
- Laura García-Curiel
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México
| | - Ma Del Rocío López-Cuellar
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México.
| | - Adriana Inés Rodríguez-Hernández
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México
| | - Norberto Chavarría-Hernández
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México
| |
Collapse
|
60
|
Ribeiro CFA, Silveira GGDOS, Cândido EDS, Cardoso MH, Espínola Carvalho CM, Franco OL. Effects of Antibiotic Treatment on Gut Microbiota and How to Overcome Its Negative Impacts on Human Health. ACS Infect Dis 2020; 6:2544-2559. [PMID: 32786282 DOI: 10.1021/acsinfecdis.0c00036] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The need for new antimicrobial therapies is evident, especially to reduce antimicrobial resistance and minimize deleterious effects on gut microbiota. However, although diverse studies discuss the adverse effects of broad-spectrum antibiotics on the microbiome ecology, targeted interventions that could solve this problem have often been overlooked. The impact of antibiotics on gut microbiota homeostasis is alarming, compromising its microbial community and leading to changes in host health. Recent studies have shown that these impacts can be transient or permanent, causing irreversible damage to gut microbiota. The responses to and changes in the gut microbial community arising from antibiotic treatment are related to its duration, the number of doses, antibiotic class, host age, genetic susceptibility, and lifestyle. In contrast, each individual's native microbiota can also affect the response to treatment as well as respond differently to antibiotic treatment. In this context, the current challenge is to promote the growth of potentially beneficial microorganisms and to reduce the proportion of microorganisms that cause dysbiosis, thus contributing to an improvement in the patient's health. An essential requirement for the development of novel antibiotics will be personalized medicinal strategies that recognize a patient's intestinal and biochemical individuality. Thus, this Review will address a new perspective on antimicrobial therapies through pathogen-selective antibiotics that minimize the impacts on human health due to changes in the gut microbiota from the use of antibiotics.
Collapse
Affiliation(s)
- Camila Fontoura Acosta Ribeiro
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
| | | | - Elizabete de Souza Cândido
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Federal District 71966-700, Brazil
| | - Marlon Henrique Cardoso
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Federal District 71966-700, Brazil
| | - Cristiano Marcelo Espínola Carvalho
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
| | - Octávio Luiz Franco
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Federal District 71966-700, Brazil
| |
Collapse
|
61
|
Pall E, Roman A. Lactoferrin Functionalized Biomaterials: Tools for Prevention of Implant-Associated Infections. Antibiotics (Basel) 2020; 9:E522. [PMID: 32824241 PMCID: PMC7459815 DOI: 10.3390/antibiotics9080522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Tissue engineering is one of the most important biotechnologies in the biomedical field. It requires the application of the principles of scientific engineering in order to design and build natural or synthetic biomaterials feasible for the maintenance of tissues and organs. Depending on the specific applications, the selection of the proper material remains a significant clinical concern. Implant-associated infection is one of the most severe complications in orthopedic implant surgeries. The treatment of these infections is difficult because the surface of the implant serves not only as a substrate for the formation of the biofilm, but also for the selection of multidrug-resistant bacterial strains. Therefore, a promising new approach for prevention of implant-related infection involves development of new implantable, non-antibiotic-based biomaterials. This review provides a brief overview of antimicrobial peptide-based biomaterials-especially those coated with lactoferrin.
Collapse
Affiliation(s)
- Emoke Pall
- Life Science Institute, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca 400372, Romania
| | - Alexandra Roman
- Department of Periodontology, Faculty of Dental Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca 400012, Romania;
| |
Collapse
|
62
|
Rasmussen TS, Koefoed AK, Jakobsen RR, Deng L, Castro-Mejía JL, Brunse A, Neve H, Vogensen FK, Nielsen DS. Bacteriophage-mediated manipulation of the gut microbiome – promises and presents limitations. FEMS Microbiol Rev 2020; 44:507-521. [DOI: 10.1093/femsre/fuaa020] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
ABSTRACT
Gut microbiome (GM) composition and function are linked to human health and disease, and routes for manipulating the GM have become an area of intense research. Due to its high treatment efficacy, the use of fecal microbiota transplantation (FMT) is generally accepted as a promising experimental treatment for patients suffering from GM imbalances (dysbiosis), e.g. caused by recurrent Clostridioides difficile infections (rCDI). Mounting evidence suggests that bacteriophages (phages) play a key role in successful FMT treatment by restoring the dysbiotic bacterial GM. As a refinement to FMT, removing the bacterial component of donor feces by sterile filtration, also referred to as fecal virome transplantation (FVT), decreases the risk of invasive infections caused by bacteria. However, eukaryotic viruses and prophage-encoded virulence factors remain a safety issue. Recent in vivo studies show how cascading effects are initiated when phage communities are transferred to the gut by e.g. FVT, which leads to changes in the GM composition, host metabolome, and improve host health such as alleviating symptoms of obesity and type-2-diabetes (T2D). In this review, we discuss the promises and limitations of FVT along with the perspectives of using FVT to treat various diseases associated with GM dysbiosis.
Collapse
Affiliation(s)
- Torben Sølbeck Rasmussen
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Anna Kirstine Koefoed
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Rasmus Riemer Jakobsen
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Ling Deng
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Josué L Castro-Mejía
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Anders Brunse
- Section of Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Ridebanevej 9, 2nd floor - 1870, Frederiksberg, Denmark
| | - Horst Neve
- Institute of Microbiology and Biotechnology, Max Rubner-Institut, Hermann-Weigmann-Straße 1 - 24103, Kiel, Germany
| | - Finn Kvist Vogensen
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| | - Dennis Sandris Nielsen
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Rolighedsvej 26 4th floor - 1958, Frederiksberg, Denmark
| |
Collapse
|
63
|
Wang Y, Wang J, Bai D, Wei Y, Sun J, Luo Y, Zhao J, Liu Y, Wang Q. Synergistic inhibition mechanism of pediocin PA-1 and L-lactic acid against Aeromonas hydrophila. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183346. [PMID: 32428447 DOI: 10.1016/j.bbamem.2020.183346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/20/2020] [Accepted: 05/07/2020] [Indexed: 01/06/2023]
Abstract
Pediocin PA-1 (PA-1) is a membrane-targeting bacteriocin from lactic acid bacteria, which shows antimicrobial activity against a wide range of Gram-positive pathogens. However, the outer membrane of Gram-negative bacteria does not allow pediocin access to its target. In this work, the synergistic inhibitory mechanism of PA-1 with L-lactic acid against Gram-negative aquaculture and food pathogen Aeromonas hydrophila (A. hydrophila) was analyzed. The combined treatment of 3.5 mmol/L L-lactic acid and 50 μmol/L (or 30 μmol/L) PA-1 had strong bacteriostatic and bactericidal activity against A. hydrophila. Full wavelength scanning and ELISA assay revealed the release of lipopolysaccharide (LPS) from the outer membrane of A. hydrophila caused by L-lactic acid treatment. Laser confocal microscopic imaging of A. hydrophila with FITC-labeled pediocin PA-1 proved the accumulation of PA-1 on lactic acid-treated bacterial cells. PA-1 then caused a rapid dissipation of membrane potential (Δψ) and a proton gradient difference (ΔpH) in lactic acid-treated A. hydrophila. Pediocin PA-1 also caused an increase in the extracellular ATP level. Morphology revealed by SEM and TEM showed that combined treating with lactic acid and PA-1 induced vesicles on the cell surface, the outer and inner membrane disruption, and even cytoplasm leakage and cell lysis. The results proved a potential mechanism of the synergistic inhibition of lactic acid and PA-1 against A. hydrophila, by which L-lactic acid released the outer membrane LPS, making it possible for PA-1 to contact the plasma membrane of A. hydrophila, resulting in the dissipation of proton-motive force in the inner membrane and cell death.
Collapse
Affiliation(s)
- Yang Wang
- Tianjin Key Laboratory of Aqua-ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, 22 Jinjing Road, 300384 Tianjin, China.
| | - Jingru Wang
- Tianjin Key Laboratory of Aqua-ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, 22 Jinjing Road, 300384 Tianjin, China
| | - Dongqing Bai
- Tianjin Key Laboratory of Aqua-ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, 22 Jinjing Road, 300384 Tianjin, China.
| | - Yunlu Wei
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Jingfeng Sun
- Tianjin Key Laboratory of Aqua-ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, 22 Jinjing Road, 300384 Tianjin, China
| | - Yunlong Luo
- Tianjin Key Laboratory of Aqua-ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, 22 Jinjing Road, 300384 Tianjin, China
| | - Jing Zhao
- Tianjin Key Laboratory of Aqua-ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, 22 Jinjing Road, 300384 Tianjin, China
| | - Ying Liu
- Tianjin Key Laboratory of Aqua-ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, 22 Jinjing Road, 300384 Tianjin, China
| | - Qingkui Wang
- Tianjin Key Laboratory of Aqua-ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, 22 Jinjing Road, 300384 Tianjin, China
| |
Collapse
|
64
|
O'Connor PM, Kuniyoshi TM, Oliveira RP, Hill C, Ross RP, Cotter PD. Antimicrobials for food and feed; a bacteriocin perspective. Curr Opin Biotechnol 2020; 61:160-167. [PMID: 31968296 DOI: 10.1016/j.copbio.2019.12.023] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/30/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023]
Abstract
Bacteriocins are natural antimicrobials that have been consumed via fermented foods for millennia and have been the focus of renewed efforts to identify novel bacteriocins, and their producing microorganisms, for use as food biopreservatives and other applications. Bioengineering bacteriocins or combining bacteriocins with multiple modes of action (hurdle approach) can enhance their preservative effect and reduces the incidence of antimicrobial resistance. In addition to their role as food biopreservatives, bacteriocins are gaining credibility as health modulators, due to their ability to regulate the gut microbiota, which is strongly associated with human wellbeing. Indeed the strengthening link between the gut microbiota and obesity make bacteriocins ideal alternatives to Animal Growth Promoters (AGP) in animal feed also. Here we review recent advances in bacteriocin research that will contribute to the development of functional foods and feeds as a consequence of roles in food biopreservation and human/animal health.
Collapse
Affiliation(s)
- Paula M O'Connor
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Taís M Kuniyoshi
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; Biochemical and Pharmaceutical Technology Department, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Lineu Prestes 580, São Paulo 05508-900, Brazil
| | - Ricardo Ps Oliveira
- Biochemical and Pharmaceutical Technology Department, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Lineu Prestes 580, São Paulo 05508-900, Brazil
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Ireland; School of Microbiology, University College Cork, Ireland
| | - Reynolds Paul Ross
- APC Microbiome Ireland, University College Cork, Ireland; School of Microbiology, University College Cork, Ireland
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
65
|
Todorov SD, Kang HJ, Ivanova IV, Holzapfel WH. Bacteriocins From LAB and Other Alternative Approaches for the Control of Clostridium and Clostridiodes Related Gastrointestinal Colitis. Front Bioeng Biotechnol 2020; 8:581778. [PMID: 33042979 PMCID: PMC7517946 DOI: 10.3389/fbioe.2020.581778] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
The gut microbiome is considered as a promising target for future non-conventional therapeutic treatment of inflammatory and infectious diseases. The search for appropriate safe and beneficial (lactic acid bacterial and other) putative probiotic strains and/or their antimicrobial metabolites represents a challenging approach for combating several problematic and emerging infections. The process of selecting suitable strains, especially of lactic acid bacteria (LAB) with superior properties, has been accelerated and intensified during the past two decades, also thanks to recent developments in lab techniques. Currently, special focus is on the potential of antimicrobial metabolites produced by some LAB strains and their application as active therapeutic agents. The vision is to develop a scientific basis for 'biotherapeutics' as alternative to conventional approaches in both human and veterinary medicine. Consequently, innovative and promising applications of LAB to the therapeutic practice are presently emerging. An overview of the existing literature indicates that some antimicrobial metabolites such as bacteriocins, widely produced by different bacterial species including LAB, are promising biotherapeutic agents for controlling infections caused by potential pathogens, such as Clostridium and Clostridiodes. Non-conventional, safe and well designed therapeutic treatments may contribute to the improvement of gut dysbiotic conditions. Thereby gut homeostasis can be restored and inflammatory conditions such as gastrointestinal colitis ameliorated. Combining the knowledge on the production, characterization and application of bacteriocins from probiotic LAB, together with their antibacterial properties, appears to be a promising and novel approach in biotherapy. In this overview, different scenarios for the control of Clostridium spp. by application of bacteriocins as therapeutic agents, also in synergistic combination with antibiotics, will be discussed.
Collapse
Affiliation(s)
- Svetoslav D. Todorov
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, Pohang, South Korea
| | - Hye-Ji Kang
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, Pohang, South Korea
- HEM Inc., Handong Global University, Pohang, South Korea
| | - Iskra V. Ivanova
- Department of General and Applied Microbiology, Faculty of Biology, Sofia University “St. Kliment Ohridski”, Sofia, Bulgaria
| | - Wilhelm H. Holzapfel
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, Pohang, South Korea
- HEM Inc., Handong Global University, Pohang, South Korea
- *Correspondence: Wilhelm H. Holzapfel,
| |
Collapse
|
66
|
Lactobacillus acidophilus JCM 1132 Strain and Its Mutant with Different Bacteriocin-Producing Behaviour Have Various in Situ Effects on the Gut Microbiota of Healthy Mice. Microorganisms 2019; 8:microorganisms8010049. [PMID: 31881756 PMCID: PMC7022661 DOI: 10.3390/microorganisms8010049] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023] Open
Abstract
The production of bacteriocin is considered to be a probiotic trait of lactic acid bacteria (LAB). However, not all strains of LAB harbour bacteriocin genes, even within the same species. Moreover, the effects of bacteriocins on the host gut microbiota and on host physiological indicators are rarely studied. This study evaluated the effects of the bacteriocin-producing Lactobacillus acidophilus strain JCM1132 and its non-producing spontaneous mutant, L. acidophilus CCFM720, on the physiological statuses and gut microbiota of healthy mice. Mice that received the bacteriocin-producing strain JCM1132 exhibited reduced water and food intake. Furthermore, the administration of these strains induced significant changes in the compositional abundance of faecal microbiota at the phylum and genus levels, and some of these changes were more pronounced after one week of withdrawal. The effects of CCFM720 treatment on the gut microbiota seemed to favour the prevention of metabolic diseases to some extent. However, individuals that received JCM1132 treatment exhibited weaker inflammatory responses than those that received CCFM720 treatment. Our results indicate that treatment with bacteriocin-producing or non-producing strains can have different effects on the host. Accordingly, this trait should be considered in the applications of LAB.
Collapse
|
67
|
Umu ÖCO, Gueimonde M, Oostindjer M, Ovchinnikov KV, de los Reyes-Gavilán CG, Arbulu S, Hernández PE, Martínez B, Diep DB, Salazar N. Use of Fecal Slurry Cultures to Study In Vitro Effects of Bacteriocins on the Gut Bacterial Populations of Infants. Probiotics Antimicrob Proteins 2019; 12:1218-1225. [DOI: 10.1007/s12602-019-09614-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
68
|
Gontijo MTP, Silva JDS, Vidigal PMP, Martin JGP. Phylogenetic distribution of the bacteriocin repertoire of lactic acid bacteria species associated with artisanal cheese. Food Res Int 2019; 128:108783. [PMID: 31955749 DOI: 10.1016/j.foodres.2019.108783] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 10/22/2019] [Accepted: 10/26/2019] [Indexed: 12/20/2022]
Abstract
The microbiota contributes to artisanal cheese bioprotection and biopreservation through inter and intraspecific competition. This work aimed to investigate the phylogenetic distribution of the repertoire of bacteriocin structural genes of model lactic acid bacteria (LAB) in order to investigate its respective role in the artisanal cheeses microenvironment. A phylogenetic analysis of the rRNA 16S gene from 445 model strains of LAB was conducted using bayesian inference and the repertoire of bacteriocin genes was predicted from these strains by BAGEL software. Bacterial strains were clustered in five monophyletic clades (A, B, C, D and E) with high posterior probability values (PP > 0.99). One bacteriocin structural gene was predicted for 88.5% of the analyzed strains. The majority of the species encoded different classes of bacteriocins. Greater diversity of bacteriocin genes was found for strains included in clade A, comprising Lactococcus lactis, Streptococcus agalactiae, Streptococcus thermophilus, Streptococcus macedonicus, Enterococcus faecalis and Enterococcus faecium. In addition, Lactococcus lactis presented higher diversity of bacteriocin classes, encoding glycocins, lanthipeptides, sactipeptides, cyclic and linear azole-containing peptides, included in bacteriocins class I, besides class II and III. The results suggest that the distribution of bacteriocin structural genes is related to the phylogenetic clades of LAB species, with a higher frequency in some specific clades. Information comprised in this study contributes to comprehend the bacterial competition mechanisms in the artisanal cheese microenvironment.
Collapse
Affiliation(s)
- Marco Túlio Pardini Gontijo
- Departamento de Microbiologia, Centro de Ciências Biológicas e da Saúde (CCB), Universidade Federal de Viçosa (UFV), Viçosa, 36570-900, Minas Gerais, Brazil.
| | - Jackson de Sousa Silva
- Departamento de Engenharia de Produção, Centro de Ciências e Tecnologia (CCT), Universidade Regional do Cariri (URCA), Juazeiro do Norte, 63040-000 Ceará, Brazil.
| | - Pedro Marcus Pereira Vidigal
- Núcleo de Análise de Biomoléculas (NUBIOMOL), Universidade Federal de Viçosa (UFV), Viçosa, 36570-900, Minas Gerais, Brazil
| | - José Guilherme Prado Martin
- Departamento de Microbiologia, Centro de Ciências Biológicas e da Saúde (CCB), Universidade Federal de Viçosa (UFV), Viçosa, 36570-900, Minas Gerais, Brazil
| |
Collapse
|
69
|
Dean AD, Horsburgh MJ, Vasiev B. Toxin-mediated competition in weakly motile bacteria. J Theor Biol 2019; 480:205-217. [PMID: 31348925 DOI: 10.1016/j.jtbi.2019.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 10/26/2022]
Abstract
Many bacterial species produce toxins that inhibit their competitors. We model this phenomenon by extending classic two-species Lotka-Volterra competition in one spatial dimension to incorporate toxin production by one species. Considering solutions comprising two adjacent single-species colonies, we show how the toxin inhibits the susceptible species near the interface between the two colonies. Moreover, a sufficiently effective toxin inhibits the susceptible species to such a degree that an 'inhibition zone' is formed separating the two colonies. In the special case of truly non-motile bacteria, i.e. with zero bacterial diffusivity, we derive analytical expressions describing the bacterial distributions and size of the inhibition zone. In the more general case of weakly motile bacteria, i.e. small bacterial diffusivity, these two-colony solutions become travelling waves. We employ numerical methods to show that the wavespeed is dependent upon both interspecific competition and toxin strength; precisely which colony expands at the expense of the other depends upon the choice of parameter values. In particular, a sufficiently effective toxin allows the producer to expand at the expense of the susceptible, with a wavespeed magnitude that is bounded above as the toxin strength increases. This asymptotic wavespeed is independent of interspecific competition and due to the formation of the inhibition zone; when the colonies are thus separated, there is no longer direct competition between the two species and the producer can invade effectively unimpeded by its competitor. We note that the minimum toxin strength required to produce an inhibition zone increases rapidly with increasing bacterial diffusivity, suggesting that even moderately motile bacteria must produce very strong toxins if they are to benefit in this way.
Collapse
Affiliation(s)
- Andrew D Dean
- Institute of Integrative Biology, Biosciences Building, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK.
| | - Malcolm J Horsburgh
- Institute of Integrative Biology, Biosciences Building, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - Bakhti Vasiev
- Department of Mathematical Sciences, Mathematical Sciences Building, University of Liverpool, Liverpool L69 7ZL, UK
| |
Collapse
|
70
|
Javůrková VG, Kreisinger J, Procházka P, Požgayová M, Ševčíková K, Brlík V, Adamík P, Heneberg P, Porkert J. Unveiled feather microcosm: feather microbiota of passerine birds is closely associated with host species identity and bacteriocin-producing bacteria. THE ISME JOURNAL 2019; 13:2363-2376. [PMID: 31127178 PMCID: PMC6775979 DOI: 10.1038/s41396-019-0438-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 04/12/2019] [Accepted: 05/03/2019] [Indexed: 11/12/2022]
Abstract
The functional relevance of microbiota is a key aspect for understanding host-microbiota interactions. Mammalian skin harbours a complex consortium of beneficial microorganisms known to provide health and immune-boosting advantages. As yet, however, little is known about functional microbial communities on avian feathers, including their co-evolution with the host and factors determining feather microbiota (FM) diversity. Using 16S rRNA profiling, we investigated how host species identity, phylogeny and geographic origin determine FM in free-living passerine birds. Moreover, we estimated the relative abundance of bacteriocin-producing bacteria (BPB) and keratinolytic feather damaging bacteria (FDB) and evaluated the ability of BPB to affect FM diversity and relative abundance of FDB. Host species identity was associated with feather bacterial communities more strongly than host geographic origin. FM functional properties differed in terms of estimated BPB and FDB relative abundance, with both showing interspecific variation. FM diversity was negatively associated with BPB relative abundance across species, whereas BPB and FDB relative abundance was positively correlated. This study provides the first thorough evaluation of antimicrobial peptides-producing bacterial communities inhabiting the feather integument, including their likely potential to mediate niche-competition and to be associated with functional species-specific feather microbiota in avian hosts.
Collapse
Affiliation(s)
- Veronika Gvoždíková Javůrková
- Faculty of Agrobiology, Food and Natural Resources, Department of Animal Science, Czech University of Life Sciences, Kamýcká 129, 165 00, Prague-Suchdol, Czech Republic.
- Institute of Vertebrate Biology, Czech Academy of Sciences, Květná 8, 603 65, Brno, Czech Republic.
| | - Jakub Kreisinger
- Faculty of Science, Department of Zoology, Charles University, Viničná 7, 128 44, Prague, Czech Republic
| | - Petr Procházka
- Institute of Vertebrate Biology, Czech Academy of Sciences, Květná 8, 603 65, Brno, Czech Republic
| | - Milica Požgayová
- Institute of Vertebrate Biology, Czech Academy of Sciences, Květná 8, 603 65, Brno, Czech Republic
| | - Kateřina Ševčíková
- Faculty of Science, Department of Zoology, Palacký University, 17. listopadu 50, 771 46, Olomouc, Czech Republic
| | - Vojtěch Brlík
- Institute of Vertebrate Biology, Czech Academy of Sciences, Květná 8, 603 65, Brno, Czech Republic
| | - Peter Adamík
- Faculty of Science, Department of Zoology, Palacký University, 17. listopadu 50, 771 46, Olomouc, Czech Republic
| | - Petr Heneberg
- Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague, Czech Republic
| | - Jiří Porkert
- Home address: Gočárova třída 542/12, 500 02, Hradec Králové, Czech Republic
| |
Collapse
|
71
|
Farizano JV, Masías E, Hsu FF, Salomón RA, Freitag NE, Hebert EM, Minahk C, Saavedra L. PrfA activation in Listeria monocytogenes increases the sensitivity to class IIa bacteriocins despite impaired expression of the bacteriocin receptor. Biochim Biophys Acta Gen Subj 2019; 1863:1283-1291. [DOI: 10.1016/j.bbagen.2019.04.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 10/26/2022]
|
72
|
Balandin SV, Sheremeteva EV, Ovchinnikova TV. Pediocin-Like Antimicrobial Peptides of Bacteria. BIOCHEMISTRY (MOSCOW) 2019; 84:464-478. [PMID: 31234762 DOI: 10.1134/s000629791905002x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Bacteriocins are bacterial antimicrobial peptides that, unlike classical peptide antibiotics, are products of ribosomal synthesis and usually have a narrow spectrum of antibacterial activity against species closely related to the producers. Pediocin-like bacteriocins (PLBs) belong to the class IIa of the bacteriocins of Gram-positive bacteria. PLBs possess high activity against pathogenic bacteria from Listeria and Enterococcus genera. Molecular target for PLBs is a membrane protein complex - bacterial mannose-phosphotransferase. PLBs can be synthesized by components of symbiotic microflora and participate in the maintenance of homeostasis in various compartments of the digestive tract and on the surface of epithelial tissues contacting the external environment. PLBs could give a rise to a new group of antibiotics of narrow spectrum of activity.
Collapse
Affiliation(s)
- S V Balandin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - E V Sheremeteva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - T V Ovchinnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
| |
Collapse
|
73
|
Mills S, Stanton C, Lane JA, Smith GJ, Ross RP. Precision Nutrition and the Microbiome, Part I: Current State of the Science. Nutrients 2019; 11:nu11040923. [PMID: 31022973 PMCID: PMC6520976 DOI: 10.3390/nu11040923] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/10/2019] [Accepted: 04/17/2019] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota is a highly complex community which evolves and adapts to its host over a lifetime. It has been described as a virtual organ owing to the myriad of functions it performs, including the production of bioactive metabolites, regulation of immunity, energy homeostasis and protection against pathogens. These activities are dependent on the quantity and quality of the microbiota alongside its metabolic potential, which are dictated by a number of factors, including diet and host genetics. In this regard, the gut microbiome is malleable and varies significantly from host to host. These two features render the gut microbiome a candidate ‘organ’ for the possibility of precision microbiomics—the use of the gut microbiome as a biomarker to predict responsiveness to specific dietary constituents to generate precision diets and interventions for optimal health. With this in mind, this two-part review investigates the current state of the science in terms of the influence of diet and specific dietary components on the gut microbiota and subsequent consequences for health status, along with opportunities to modulate the microbiota for improved health and the potential of the microbiome as a biomarker to predict responsiveness to dietary components. In particular, in Part I, we examine the development of the microbiota from birth and its role in health. We investigate the consequences of poor-quality diet in relation to infection and inflammation and discuss diet-derived microbial metabolites which negatively impact health. We look at the role of diet in shaping the microbiome and the influence of specific dietary components, namely protein, fat and carbohydrates, on gut microbiota composition.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| | - Catherine Stanton
- APC Microbiome Ireland, Teagasc Food Research Centre, Fermoy P61 C996, Co Cork, Ireland.
| | - Jonathan A Lane
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | - Graeme J Smith
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| |
Collapse
|
74
|
Iacob S, Iacob DG, Luminos LM. Intestinal Microbiota as a Host Defense Mechanism to Infectious Threats. Front Microbiol 2019; 9:3328. [PMID: 30761120 PMCID: PMC6362409 DOI: 10.3389/fmicb.2018.03328] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
The intestinal microbiota is a complex microbial community, with diverse and stable populations hosted by the gastrointestinal tract since birth. This ecosystem holds multiple anti-infectious, anti-inflammatory, and immune modulating roles decisive for intestinal homeostasis. Among these, colonization resistance refers to the dynamic antagonistic interactions between commensals and pathogenic flora. Hence, gut bacteria compete for the same intestinal niches and substrates, while also releasing antimicrobial substances such as bacteriocines and changing the environmental conditions. Short chain fatty acids (SCFAs) generated in anaerobic conditions prompt epigenetic regulatory mechanisms that favor a tolerogenic immune response. In addition, the commensal flora is involved in the synthesis of bactericidal products, namely secondary biliary acids or antimicrobial peptides (AMPs) such as cathellicidin-LL37, an immunomodulatory, antimicrobial, and wound healing peptide. Gut microbiota is protected through symbiotic relations with the hosting organism and by quorum sensing, a specific cell-to-cell communication system. Any alterations of these relationships favor the uncontrollable multiplication of the resident pathobionts or external entero-pathogens, prompting systemic translocations, inflammatory reactions, or exacerbations of bacterial virulence mechanisms (T6SS, T3SS) and ultimately lead to gastrointestinal or systemic infections. The article describes the metabolic and immunological mechanisms through which the intestinal microbiota is both an ally of the organism against enteric pathogens and an enemy that favors the development of infections.
Collapse
Affiliation(s)
- Simona Iacob
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Bals", Bucharest, Romania
| | - Diana Gabriela Iacob
- National Institute of Infectious Diseases "Prof. Dr. Matei Bals", Bucharest, Romania
| | - Luminita Monica Luminos
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Bals", Bucharest, Romania
| |
Collapse
|
75
|
Bacteriocins and Bacteriophages: Therapeutic Weapons for Gastrointestinal Diseases? Int J Mol Sci 2019; 20:ijms20010183. [PMID: 30621339 PMCID: PMC6337595 DOI: 10.3390/ijms20010183] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 02/07/2023] Open
Abstract
Bacteriocins are bactericidal peptides, ribosomally synthesized, with an inhibitory activity against diverse groups of undesirable microorganisms. Bacteriocins are produced by both gram-positive and gram-negative bacteria, and to a lesser extent by some archaea. Bacteriophages are viruses that are able to infect bacterial cells and force them to produce viral components, using a lytic or lysogenic cycle. They constitute a large community in the human gut called the phageome, the most abundant part of the gut virome. Bacteriocins and bacteriophages may have an influence on both human health and diseases, thanks to their ability to modulate the gut microbiota and regulate the competitive relationship among the different microorganisms, strains and cells living in the human intestine. In this review, we explore the role of bacteriocins and bacteriophages in the most frequent gastrointestinal diseases by dissecting their interaction with the complex environment of the human gut, analyzing a possible link with extra-intestinal diseases, and speculating on their possible therapeutic application with the end goal of promoting gut health.
Collapse
|
76
|
Liu S, Liu Y, Zhang P. Properties of Listeria monocytogenes on Acquisition of Pediocin Resistance. ANN MICROBIOL 2018. [DOI: 10.1007/s13213-018-1411-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
77
|
George F, Daniel C, Thomas M, Singer E, Guilbaud A, Tessier FJ, Revol-Junelles AM, Borges F, Foligné B. Occurrence and Dynamism of Lactic Acid Bacteria in Distinct Ecological Niches: A Multifaceted Functional Health Perspective. Front Microbiol 2018; 9:2899. [PMID: 30538693 PMCID: PMC6277688 DOI: 10.3389/fmicb.2018.02899] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022] Open
Abstract
Lactic acid bacteria (LAB) are representative members of multiple ecosystems on earth, displaying dynamic interactions within animal and plant kingdoms in respect with other microbes. This highly heterogeneous phylogenetic group has coevolved with plants, invertebrates, and vertebrates, establishing either mutualism, symbiosis, commensalism, or even parasitism-like behavior with their hosts. Depending on their location and environment conditions, LAB can be dominant or sometimes in minority within ecosystems. Whatever their origins and relative abundance in specific anatomic sites, LAB exhibit multifaceted ecological and functional properties. While some resident LAB permanently inhabit distinct animal mucosal cavities, others are provided by food and may transiently occupy the gastrointestinal tract. It is admitted that the overall gut microbiome has a deep impact on health and diseases. Here, we examined the presence and the physiological role of LAB in the healthy human and several animal microbiome. Moreover, we also highlighted some dysbiotic states and related consequences for health, considering both the resident and the so-called "transionts" microorganisms. Whether LAB-related health effects act collectively or follow a strain-specificity dogma is also addressed. Besides the highly suggested contribution of LAB to interplay with immune, metabolic, and even brain-axis regulation, the possible involvement of LAB in xenobiotic detoxification processes and metal equilibrium is also tackled. Recent technological developments such as functional metagenomics, metabolomics, high-content screening and design in vitro and in vivo experimental models now open new horizons for LAB as markers applied for disease diagnosis, susceptibility, and follow-up. Moreover, identification of general and more specific molecular mechanisms based on antioxidant, antimicrobial, anti-inflammatory, and detoxifying properties of LAB currently extends their selection and promising use, either as probiotics, in traditional and functional foods, for dedicated treatments and mostly for maintenance of normobiosis and homeostasis.
Collapse
Affiliation(s)
- Fanny George
- Université de Lille, Inserm, CHU Lille, U995 – LIRIC – Lille Inflammation Research International Center, Lille, France
| | - Catherine Daniel
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR 8204 – CIIL – Center for Infection and Immunity of Lille, Lille, France
| | - Muriel Thomas
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Elisabeth Singer
- Université de Lille, Inserm, CHU Lille, U995 – LIRIC – Lille Inflammation Research International Center, Lille, France
| | - Axel Guilbaud
- Université de Lille, Inserm, CHU Lille, U995 – LIRIC – Lille Inflammation Research International Center, Lille, France
| | - Frédéric J. Tessier
- Université de Lille, Inserm, CHU Lille, U995 – LIRIC – Lille Inflammation Research International Center, Lille, France
| | - Anne-Marie Revol-Junelles
- Laboratoire d’Ingénierie des Biomolécules, École Nationale Supérieure d’Agronomie et des Industries Alimentaires – Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Frédéric Borges
- Laboratoire d’Ingénierie des Biomolécules, École Nationale Supérieure d’Agronomie et des Industries Alimentaires – Université de Lorraine, Vandœuvre-lès-Nancy, France
| | - Benoît Foligné
- Université de Lille, Inserm, CHU Lille, U995 – LIRIC – Lille Inflammation Research International Center, Lille, France
| |
Collapse
|
78
|
Heeney DD, Zhai Z, Bendiks Z, Barouei J, Martinic A, Slupsky C, Marco ML. Lactobacillus plantarum bacteriocin is associated with intestinal and systemic improvements in diet-induced obese mice and maintains epithelial barrier integrity in vitro. Gut Microbes 2018; 10:382-397. [PMID: 30409105 PMCID: PMC6546331 DOI: 10.1080/19490976.2018.1534513] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We investigated the Lactobacillus plantarum bacteriocin plantaricin EF (PlnEF) system for its contributions to L. plantarum mediated benefits in a mouse model of diet-induced obesity. C57BL/6J mice on a high-fat diet (HFD) were administered a rifampicin resistant mutant of L. plantarum NCMIB8826 (NICMB8826-R) or an isogenic ΔplnEFI mutant strain, LM0419, every 48 h for nine weeks. Mice fed wild-type L. plantarum, but not LM0419, reduced their consumption of the HFD starting three weeks into the study and exhibited an overall 10% reduction in weight gain. The responses were independent of glucose homeostasis, as both NCMIB8826-R and LM0419 fed mice had improved oral glucose tolerance compared to sham controls. Although bacteriocins have antibacterial properties, the ileal, cecal, and fecal microbiota and cecocolic metabolomes were unchanged between mice fed either wild-type L. plantarum or the ΔplnEFI mutant. Instead, only mice fed NCMIB8826-R showed an increased production of ZO-1 in ileal tissues. To verify a potential role for the plantaricin EF system in supporting intestinal epithelial function, synthesized PlnEF peptides were applied to Caco-2 cell monolayers challenged with TNF-α and IFN-γ. The combination of PlnE and PlnF were required to prevent sustained cytokine-induced losses to Caco-2 cell para- and transcellular permeability and elevated IL-8 levels. In conclusion, this study shows that probiotic L. plantarum ameliorates the effects of obesogenic diets through a mechanism that involves the plantaricin EF system and likely includes L. plantarum - induced fortification of the intestinal epithelium.
Collapse
Affiliation(s)
- Dustin D. Heeney
- Department of Food Science & Technology, University of California, Davis, CA, USA
| | - Zhengyuan Zhai
- Department of Food Science & Technology, University of California, Davis, CA, USA
| | - Zach Bendiks
- Department of Food Science & Technology, University of California, Davis, CA, USA
| | - Javad Barouei
- Department of Food Science & Technology, University of California, Davis, CA, USA
| | - Alice Martinic
- Department of Nutrition, University of California, Davis, CA, USA
| | - Carolyn Slupsky
- Department of Food Science & Technology, University of California, Davis, CA, USA,Department of Nutrition, University of California, Davis, CA, USA
| | - Maria L. Marco
- Department of Food Science & Technology, University of California, Davis, CA, USA,CONTACT Maria L. Marco Department of Food Science & Technology, University of California, Davis, One Shields Avenue, Davis, CA 95616
| |
Collapse
|
79
|
Martínez B, García P, Rodríguez A. Swapping the roles of bacteriocins and bacteriophages in food biotechnology. Curr Opin Biotechnol 2018; 56:1-6. [PMID: 30098459 DOI: 10.1016/j.copbio.2018.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 07/17/2018] [Indexed: 01/31/2023]
Abstract
To move towards a safer and more sustainable food production chain, natural antimicrobials have been traditionally applied to enhance safety. This is well exemplified by the use of bacteriocins, antimicrobial peptides synthesized by bacteria, as food biopreservatives. However, as knowledge on bacteriocin biology develops, novel functions beyond food preservation emerge and a shift towards health applications is positioning bacteriocins as anti-infectives and modulators of gut microbiota. On the other hand, bacteriophages, viruses infecting bacteria, have been long regarded as a threat for dairy fermentations. However, they may also become allies when specific phages infecting pathogenic or spoilage bacteria are intentionally used. This review summarizes the `dark side' and rather unexplored roles of bacteriocins and phages that, certainly, have much to learn from each other.
Collapse
Affiliation(s)
- Beatriz Martínez
- DairySafe Group, Instituto de Productos Lácteos de Asturias, IPLA-CSIC, Paseo Río Linares s/n, 33300 Villaviciosa, Spain.
| | - Pilar García
- DairySafe Group, Instituto de Productos Lácteos de Asturias, IPLA-CSIC, Paseo Río Linares s/n, 33300 Villaviciosa, Spain
| | - Ana Rodríguez
- DairySafe Group, Instituto de Productos Lácteos de Asturias, IPLA-CSIC, Paseo Río Linares s/n, 33300 Villaviciosa, Spain
| |
Collapse
|
80
|
Garcia-Gutierrez E, Mayer MJ, Cotter PD, Narbad A. Gut microbiota as a source of novel antimicrobials. Gut Microbes 2018; 10:1-21. [PMID: 29584555 PMCID: PMC6363078 DOI: 10.1080/19490976.2018.1455790] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/13/2018] [Accepted: 03/19/2018] [Indexed: 02/08/2023] Open
Abstract
Bacteria, Archaea, Eukarya and viruses coexist in the human gut, and this coexistence is functionally balanced by symbiotic or antagonistic relationships. Antagonism is often characterized by the production of antimicrobials against other organisms occupying the same environmental niche. Indeed, close co-evolution in the gut has led to the development of specialized antimicrobials, which is attracting increased attention as these may serve as novel alternatives to antibiotics and thereby help to address the global problem of antimicrobial resistance. The gastrointestinal (GI) tract is especially suitable for finding novel antimicrobials due to the vast array of microbes that inhabit it, and a considerable number of antimicrobial producers of both wide and narrow spectrum have been described. In this review, we summarize some of the antimicrobial compounds that are produced by bacteria isolated from the gut environment, with a special focus on bacteriocins. We also evaluate the potential therapeutic application of these compounds to maintain homeostasis in the gut and the biocontrol of pathogenic bacteria.
Collapse
Affiliation(s)
- Enriqueta Garcia-Gutierrez
- Gut Health and Food Safety Institute Strategic Programme, Quadram Institute Bioscience, Norwich, UK
- Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland
| | - Melinda J. Mayer
- Gut Health and Food Safety Institute Strategic Programme, Quadram Institute Bioscience, Norwich, UK
| | - Paul D. Cotter
- Food Bioscience Department, Teagasc Food Research Centre, Fermoy, Ireland
- APC Microbiome, Ireland
| | - Arjan Narbad
- Gut Health and Food Safety Institute Strategic Programme, Quadram Institute Bioscience, Norwich, UK
| |
Collapse
|
81
|
Mills S, Ross RP, Hill C. Bacteriocins and bacteriophage; a narrow-minded approach to food and gut microbiology. FEMS Microbiol Rev 2018; 41:S129-S153. [PMID: 28830091 DOI: 10.1093/femsre/fux022] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022] Open
Abstract
Bacteriocins and bacteriophage (phage) are biological tools which exhibit targeted microbial killing, a phenomenon which until recently was seen as a major drawback for their use as antimicrobial agents. However, in an age when the deleterious consequences of broad-spectrum antibiotics on human health have become apparent, there is an urgent need to develop narrow-spectrum substitutes. Indeed, disruption of the microbial communities which exist on and in our bodies can generate immediate and long-term negative effects and this is particularly borne out in the gut microbiota community whose disruption has been linked to a number of disorders reaching as far as the brain. Moreover, the antibiotic resistance crisis has resulted in our inability to treat many bacterial infections and has triggered the search for damage-limiting alternatives. As bacteriocins and phage are natural entities they are relatively easy to isolate and characterise and are also ideal candidates for improving food safety and quality, forfeiting the need for largely unpopular chemical preservatives. This review highlights the efficacy of both antimicrobial agents in terms of gut health and food safety and explores the body of scientific evidence supporting their effectiveness in both environments.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Institute and School of Microbiology, University College Cork, Western Road, Cork T12 YN60, Ireland
| | - R Paul Ross
- APC Microbiome Institute and School of Microbiology, University College Cork, Western Road, Cork T12 YN60, Ireland
| | - Colin Hill
- APC Microbiome Institute and School of Microbiology, University College Cork, Western Road, Cork T12 YN60, Ireland
| |
Collapse
|
82
|
Assessing in vitro digestibility of food biopreservative AS-48. Food Chem 2018; 246:249-257. [DOI: 10.1016/j.foodchem.2017.10.149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/11/2017] [Accepted: 10/31/2017] [Indexed: 11/18/2022]
|
83
|
Sharma C, Rokana N, Chandra M, Singh BP, Gulhane RD, Gill JPS, Ray P, Puniya AK, Panwar H. Antimicrobial Resistance: Its Surveillance, Impact, and Alternative Management Strategies in Dairy Animals. Front Vet Sci 2018; 4:237. [PMID: 29359135 PMCID: PMC5766636 DOI: 10.3389/fvets.2017.00237] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/15/2017] [Indexed: 01/10/2023] Open
Abstract
Antimicrobial resistance (AMR), one among the most common priority areas identified by both national and international agencies, is mushrooming as a silent pandemic. The advancement in public health care through introduction of antibiotics against infectious agents is now being threatened by global development of multidrug-resistant strains. These strains are product of both continuous evolution and un-checked antimicrobial usage (AMU). Though antibiotic application in livestock has largely contributed toward health and productivity, it has also played significant role in evolution of resistant strains. Although, a significant emphasis has been given to AMR in humans, trends in animals, on other hand, are not much emphasized. Dairy farming involves surplus use of antibiotics as prophylactic and growth promoting agents. This non-therapeutic application of antibiotics, their dosage, and withdrawal period needs to be re-evaluated and rationally defined. A dairy animal also poses a serious risk of transmission of resistant strains to humans and environment. Outlining the scope of the problem is necessary for formulating and monitoring an active response to AMR. Effective and commendably connected surveillance programs at multidisciplinary level can contribute to better understand and minimize the emergence of resistance. Besides, it requires a renewed emphasis on investments into research for finding alternate, safe, cost effective, and innovative strategies, parallel to discovery of new antibiotics. Nevertheless, numerous direct or indirect novel approaches based on host-microbial interaction and molecular mechanisms of pathogens are also being developed and corroborated by researchers to combat the threat of resistance. This review places a concerted effort to club the current outline of AMU and AMR in dairy animals; ongoing global surveillance and monitoring programs; its impact at animal human interface; and strategies for combating resistance with an extensive overview on possible alternates to current day antibiotics that could be implemented in livestock sector.
Collapse
Affiliation(s)
- Chetan Sharma
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Namita Rokana
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Mudit Chandra
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Brij Pal Singh
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Rohini Devidas Gulhane
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Jatinder Paul Singh Gill
- School of Public Health and Zoonoses, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Pallab Ray
- Department of Medical Microbiology, Post Graduate Institute for Medical Education and Research (PGIMER), Chandigarh, India
| | - Anil Kumar Puniya
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Harsh Panwar
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| |
Collapse
|
84
|
Yin X, Heeney D, Srisengfa Y, Golomb B, Griffey S, Marco M. Bacteriocin biosynthesis contributes to the anti-inflammatory capacities of probiotic Lactobacillus plantarum. Benef Microbes 2017; 9:333-344. [PMID: 29065706 DOI: 10.3920/bm2017.0096] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Plantaricin EF (PlnEF) is a class IIb bacteriocin produced by Lactobacillus plantarum. We compared L. plantarum NCIMB8826 and LM0419, a plnEFI deletion mutant of that strain lacking plnEF and the gene for the cognate immunity protein plnI, in a 2,4,6-trinitrobenzenesulfonic acid (TNBS) induced mouse model of acute inflammatory bowel disease. Mice fed either L. plantarum NCIMB8826 or LM0419 were not protected against TNBS according to either disease activity or histology (Ameho) scores. Mice consuming NCIMB8826 exhibited intermediate (non-significant) levels of colonic tumour necrosis factor alpha (TNF-α) and interleukin-6 (IL-6) that ranged between the TNBS-treated animals and healthy controls. By comparison, TNF-α and IL-6 quantities were elevated in mice given L. plantarum LM0419 and equivalent to mice given TNBS alone. Both strains survived digestive tract transit in equal numbers and did not result in global changes to the bacterial composition in the intestine according to 16S rRNA gene sequencing either prior to or after TNBS administration. Examination of intestinal taxa showed that mice consuming wild-type L. plantarum, but not LM0419 contained lower proportions of Mucispirillum (Deferribacteres phylum) in the faeces prior to TNBS administration and Parabacteroides (Bacteroidetes phylum) in the caecum after disease induction. Parabacteroides also positively correlated with disease activity and histology scores. These findings suggest a role for PlnEFI production by L. plantarum in benefiting digestive tract health.
Collapse
Affiliation(s)
- X Yin
- 1 Department of Food Science and Technology, University of California, One Shields Avenue, Davis, CA 95616, USA.,2 Department of Plant Pathology, University of California, One Shields Avenue, Davis CA 95616-8751, USA
| | - D Heeney
- 1 Department of Food Science and Technology, University of California, One Shields Avenue, Davis, CA 95616, USA
| | - Y Srisengfa
- 1 Department of Food Science and Technology, University of California, One Shields Avenue, Davis, CA 95616, USA
| | - B Golomb
- 1 Department of Food Science and Technology, University of California, One Shields Avenue, Davis, CA 95616, USA.,3 Bayer U.S. LLC, Crop Science Division, 890 Embarcadero Dr, West Sacramento, CA 95605, USA
| | - S Griffey
- 4 Comparative Pathology Laboratory, School of Veterinary Medicine, University of California, 944 Garrod Dr. 2045 Davis, CA 95616, USA
| | - M Marco
- 1 Department of Food Science and Technology, University of California, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
85
|
Bäuerl C, Umu ÖCO, Hernandez PE, Diep DB, Pérez-Martínez G. A Method to Assess Bacteriocin Effects on the Gut Microbiota of Mice. J Vis Exp 2017. [PMID: 28784971 DOI: 10.3791/56053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Very intriguing questions arise with our advancing knowledge on gut microbiota composition and the relationship with health, particularly relating to the factors that contribute to maintaining the population balance. However, there are limited available methodologies to evaluate these factors. Bacteriocins are antimicrobial peptides produced by many bacteria that may confer a competitive advantage for food acquisition and/or niche establishment. Many probiotic lactic acid bacteria (LAB) strains have great potential to promote human and animal health by preventing the growth of pathogens. They can also be used for immuno-modulation, as they produce bacteriocins. However, the antagonistic activity of bacteriocins is normally determined by laboratory bioassays under well-defined but over-simplified conditions compared to the complex gut environment in humans and animals, where bacteria face multifactorial influences from the host and hundreds of microbial species sharing the same niche. This work describes a complete and efficient procedure to assess the effect of a variety of bacteriocins with different target specificities in a murine system. Changes in the microbiota composition during the bacteriocin treatment are monitored using compositional 16S rDNA sequencing. Our approach uses both the bacteriocin producers and their isogenic non-bacteriocin-producing mutants, the latter giving the ability to distinguish bacteriocin-related from non-bacteriocin-related modifications of the microbiota. The fecal DNA extraction and 16S rDNA sequencing methods are consistent and, together with the bioinformatics, constitute a powerful procedure to find faint changes in the bacterial profiles and to establish correlations, in terms of cholesterol and triglyceride concentration, between bacterial populations and health markers. Our protocol is generic and can thus be used to study other compounds or nutrients with the potential to alter the host microbiota composition, either when studying toxicity or beneficial effects.
Collapse
Affiliation(s)
- Chrstine Bäuerl
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC)
| | - Özgun C O Umu
- Department of Food Safety and Infection Biology, Norwegian University of Life Sciences (NMBU)
| | - Pablo E Hernandez
- Departamento de Nutrición, Bromatología y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid (UCM)
| | - Dzung B Diep
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU)
| | - Gaspar Pérez-Martínez
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC);
| |
Collapse
|
86
|
Pediococcus spp.: An important genus of lactic acid bacteria and pediocin producers. Biotechnol Adv 2017; 35:361-374. [DOI: 10.1016/j.biotechadv.2017.03.004] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 02/19/2017] [Accepted: 03/04/2017] [Indexed: 02/07/2023]
|
87
|
Bacteriocins: antibiotics in the age of the microbiome. Emerg Top Life Sci 2017; 1:55-63. [PMID: 33525813 DOI: 10.1042/etls20160015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/20/2022]
Abstract
Antibiotics have revolutionised the treatment of infectious disease and improved the lives of billions of people worldwide over many decades. With the rise in antimicrobial resistance (AMR) and corresponding lack of antibiotic development, we find ourselves in dire need of alternative treatments. Bacteriocins are a class of bacterially produced, ribosomally synthesised, antimicrobial peptides that may be narrow or broad in their spectra of activity. Animal models have demonstrated the safety and efficacy of bacteriocins in treating a broad range of infections; however, one of the principal drawbacks has been their relatively narrow spectra when compared with small-molecule antibiotics. In an era where we are beginning to appreciate the role of the microbiota in human and animal health, the fact that bacteriocins cause much less collateral damage to the host microbiome makes them a highly desirable therapeutic. This review makes a case for the implementation of bacteriocins as therapeutic antimicrobials, either alone or in combination with existing antibiotics to alleviate the AMR crisis and to lessen the impact of antibiotics on the host microbiome.
Collapse
|
88
|
Umu ÖCO, Rudi K, Diep DB. Modulation of the gut microbiota by prebiotic fibres and bacteriocins. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2017; 28:1348886. [PMID: 28959178 PMCID: PMC5614387 DOI: 10.1080/16512235.2017.1348886] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/24/2017] [Indexed: 12/21/2022]
Abstract
The gut microbiota is considered an organ that co-develops with the host throughout its life. The composition and metabolic activities of the gut microbiota are subject to a complex interplay between the host genetics and environmental factors, such as lifestyle, diet, stress and antimicrobials. It is evident that certain prebiotics, and antimicrobials produced by lactic acid bacteria (LAB), can shape the composition of the gut microbiota and its metabolic activities to promote host health and/or prevent diseases. In this review, we aim to give an overview of the impact of prebiotic fibres, and bacteriocins from LAB, on the gut microbiota and its activities, which affect the physiology and health of the host. These represent two different mechanisms in modulating the gut microbiota, the first involving exploitative competition by which the growth of beneficial bacteria is promoted and the latter involving interference competition by which the growth of pathogens and other unwanted bacteria is prevented. For interference competition in the gut, bacteriocins offer special advantages over traditional antibiotics, in that they can be designed to act towards specific unwanted bacteria and other pathogens, without any remarkable collateral effects on beneficial microbes sharing the same niche.
Collapse
Affiliation(s)
- Özgün C. O. Umu
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Knut Rudi
- 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
| |
Collapse
|