1
|
Liu D, Li C, Cao T, Lv X, Yue Y, Li S, Cheng Y, Liu F, Huo G, Li B. Bifidobacterium longum K5 Prevents Enterohaemorrhagic Escherichia coli O157:H7 Infection in Mice through the Modulation of the Gut Microbiota. Nutrients 2024; 16:1164. [PMID: 38674854 PMCID: PMC11053520 DOI: 10.3390/nu16081164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7 is a commonly encountered foodborne pathogen that can cause hemorrhagic enteritis and lead to hemolytic uremic syndrome (HUS) in severe cases. Bifidobacterium is a beneficial bacterium that naturally exists in the human gut and plays a vital role in maintaining a healthy balance in the gut microbiota. This study investigated the protective effects of B. longum K5 in a mouse model of EHEC O157:H7 infection. The results indicated that pretreatment with B. longum K5 mitigated the clinical symptoms of EHEC O157:H7 infection and attenuated the increase in myeloperoxidase (MPO) activity in the colon of the mice. In comparison to the model group, elevated serum D-lactic acid concentrations and diamine oxidase (DAO) levels were prevented in the K5-EHEC group of mice. The reduced mRNA expression of tight junction proteins (ZO-1, Occludin, and Claudin-1) and mucin MUC2, as well as the elevated expression of virulence factors Stx1A and Stx2A, was alleviated in the colon of both the K5-PBS and K5-EHEC groups. Additionally, the increase in the inflammatory cytokine levels of TNF-α and IL-1β was inhibited and the production of IL-4 and IL-10 was promoted in the K5-EHEC group compared with the model group. B. longum K5 significantly prevented the reduction in the abundance and diversity of mouse gut microorganisms induced by EHEC O157:H7 infection, including blocking the decrease in the relative abundance of Roseburia, Lactobacillus, and Oscillibacter. Meanwhile, the intervention with B. longum K5 promoted the production of acetic acid and butyric acid in the gut. This study provides insights into the use of B. longum K5 for developing probiotic formulations to prevent intestinal diseases caused by pathogenic bacterial infections.
Collapse
Affiliation(s)
- Deyu Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Chunyan Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Ting Cao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Xiuli Lv
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Yingxue Yue
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Shuang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Yang Cheng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
2
|
Calvigioni M, Mazzantini D, Celandroni F, Ghelardi E. Animal and In Vitro Models as Powerful Tools to Decipher the Effects of Enteric Pathogens on the Human Gut Microbiota. Microorganisms 2023; 12:67. [PMID: 38257894 PMCID: PMC10818369 DOI: 10.3390/microorganisms12010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Examining the interplay between intestinal pathogens and the gut microbiota is crucial to fully comprehend the pathogenic role of enteropathogens and their broader impact on human health. Valid alternatives to human studies have been introduced in laboratory practice to evaluate the effects of infectious agents on the gut microbiota, thereby exploring their translational implications in intestinal functionality and overall health. Different animal species are currently used as valuable models for intestinal infections. In addition, considering the recent advances in bioengineering, futuristic in vitro models resembling the intestinal environment are also available for this purpose. In this review, the impact of the main human enteropathogens (i.e., Clostridioides difficile, Campylobacter jejuni, diarrheagenic Escherichia coli, non-typhoidal Salmonella enterica, Shigella flexneri and Shigella sonnei, Vibrio cholerae, and Bacillus cereus) on intestinal microbial communities is summarized, with specific emphasis on results derived from investigations employing animal and in vitro models.
Collapse
Affiliation(s)
| | | | | | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy; (M.C.)
| |
Collapse
|
3
|
Huang Y, Lin X, Yu S, Chen R, Chen W. Intestinal Engineered Probiotics as Living Therapeutics: Chassis Selection, Colonization Enhancement, Gene Circuit Design, and Biocontainment. ACS Synth Biol 2022; 11:3134-3153. [PMID: 36094344 DOI: 10.1021/acssynbio.2c00314] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Intestinal probiotics are often used for the in situ treatment of diseases, such as metabolic disorders, tumors, and chronic inflammatory infections. Recently, there has been an increased emphasis on intelligent, customized treatments with a focus on long-term efficacy; however, traditional probiotic therapy has not kept up with this trend. The use of synthetic biology to construct gut-engineered probiotics as live therapeutics is a promising avenue in the treatment of specific diseases, such as phenylketonuria and inflammatory bowel disease. These studies generally involve a series of fundamental design issues: choosing an engineered chassis, improving the colonization ability of engineered probiotics, designing functional gene circuits, and ensuring the safety of engineered probiotics. In this review, we summarize the relevant past research, the progress of current research, and discuss the key issues that restrict the widespread application of intestinal engineered probiotic living therapeutics.
Collapse
Affiliation(s)
- Yan Huang
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Xiaojun Lin
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Siyang Yu
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Ruiyue Chen
- Team SZU-China at iGEM 2021, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Weizhao Chen
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.,Shenzhen Key Laboratory for Microbial Gene Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
4
|
Zimmermann C, Wagner AE. Impact of Food-Derived Bioactive Compounds on Intestinal Immunity. Biomolecules 2021; 11:biom11121901. [PMID: 34944544 PMCID: PMC8699755 DOI: 10.3390/biom11121901] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal system is responsible for the digestion and the absorption of nutrients. At the same time, it is essentially involved in the maintenance of immune homeostasis. The strongest antigen contact in an organism takes place in the digestive system showing the importance of a host to develop mechanisms allowing to discriminate between harmful and harmless antigens. An efficient intestinal barrier and the presence of a large and complex part of the immune system in the gut support the host to implement this task. The continuous ingestion of harmless antigens via the diet requires an efficient immune response to reliably identify them as safe. However, in some cases the immune system accidentally identifies harmless antigens as dangerous leading to various diseases such as celiac disease, inflammatory bowel diseases and allergies. It has been shown that the intestinal immune function can be affected by bioactive compounds derived from the diet. The present review provides an overview on the mucosal immune reactions in the gut and how bioactive food ingredients including secondary plant metabolites and probiotics mediate its health promoting effects with regard to the intestinal immune homeostasis.
Collapse
|
5
|
Lee KS, Jeong YJ, Lee MS. Escherichia coli Shiga Toxins and Gut Microbiota Interactions. Toxins (Basel) 2021; 13:toxins13060416. [PMID: 34208170 PMCID: PMC8230793 DOI: 10.3390/toxins13060416] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
Escherichia coli (EHEC) and Shigella dysenteriae serotype 1 are enterohemorrhagic bacteria that induce hemorrhagic colitis. This, in turn, may result in potentially lethal complications, such as hemolytic uremic syndrome (HUS), which is characterized by thrombocytopenia, acute renal failure, and neurological abnormalities. Both species of bacteria produce Shiga toxins (Stxs), a phage-encoded exotoxin inhibiting protein synthesis in host cells that are primarily responsible for bacterial virulence. Although most studies have focused on the pathogenic roles of Stxs as harmful substances capable of inducing cell death and as proinflammatory factors that sensitize the host target organs to damage, less is known about the interface between the commensalism of bacterial communities and the pathogenicity of the toxins. The gut contains more species of bacteria than any other organ, providing pathogenic bacteria that colonize the gut with a greater number of opportunities to encounter other bacterial species. Notably, the presence in the intestines of pathogenic EHEC producing Stxs associated with severe illness may have compounding effects on the diversity of the indigenous bacteria and bacterial communities in the gut. The present review focuses on studies describing the roles of Stxs in the complex interactions between pathogenic Shiga toxin-producing E. coli, the resident microbiome, and host tissues. The determination of these interactions may provide insights into the unresolved issues regarding these pathogens.
Collapse
Affiliation(s)
- Kyung-Soo Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea;
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| | - Yu-Jin Jeong
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea;
- Correspondence: (Y.-J.J.); (M.-S.L.)
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea;
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
- Correspondence: (Y.-J.J.); (M.-S.L.)
| |
Collapse
|
6
|
Ohno H. The impact of metabolites derived from the gut microbiota on immune regulation and diseases. Int Immunol 2020; 32:629-636. [PMID: 32564086 DOI: 10.1093/intimm/dxaa041] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota strongly impacts the physiology and pathology in the host. To understand the complex interactions between host and gut microbiota, an 'integrated omics' approach has been employed, where exhaustive analyses for the different layers of cellular functions, such as epigenomics, transcriptomics and metabolomics, in addition to metagenomics, are combined. With this approach, the mechanisms whereby short-chain fatty acids (SCFAs) regulate host defense and the immune system have been elucidated. In a gnotobiotic mouse model of enterohemorrhagic Escherichia coli infection, Bifidobacterium-derived acetate can protect from infection-mediated death by changing the gene expression profile of colonic epithelial cells. It has also been shown that gut microbiota-derived butyrate enhances colonic regulatory T-cell differentiation through its epigenetic modulatory ability via histone deacetylase inhibition. SCFAs are involved in many other immunomodulatory effects as well as host pathophysiological conditions. Dysbiosis in the gut has been implicated in the pathogenesis of many diseases. Although the causal relationship of gut microbial dysbiosis and/or metabolites with pathogenesis is mostly unknown, mechanistic insights have been elucidated in some cases. Metabolism in the gut microbiota and host liver produces trimethylamine N-oxide, which is known to aggravate atherosclerosis, and a secondary bile acid deoxycholate, which reportedly induces non-alcoholic steatohepatitis-related hepatocellular carcinoma. It has been reported that secondary bile acids could also induce the differentiation of peripherally derived regulatory T cells in the gut. Further studies on the interactions between the host and gut microbiota could lead to the development of new therapeutic strategies as well as in preventive medicine.
Collapse
Affiliation(s)
- Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, Japan.,Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan.,Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, Japan.,Laboratory for Immune Regulation, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba, Japan
| |
Collapse
|
7
|
Complete Genome Sequence of Bifidobacterium longum Strain Jih1, Isolated from Human Feces. Microbiol Resour Announc 2020; 9:9/22/e00319-20. [PMID: 32467270 PMCID: PMC7256257 DOI: 10.1128/mra.00319-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We report the complete genome sequence of Bifidobacterium longum strain Jih1, isolated from human feces. The assembled genome comprised one circular chromosome of 2.37 Mb. The chromosome harbors 1,941 protein-coding genes. We report the complete genome sequence of Bifidobacterium longum strain Jih1, isolated from human feces. The assembled genome comprised one circular chromosome of 2.37 Mb. The chromosome harbors 1,941 protein-coding genes.
Collapse
|
8
|
Koyanagi Y, Suzuki R, Ihara K, Miyagi H, Isogai H, Yoneyama H, Isogai E. Intestinal Clostridium species lower host susceptibility to enterohemorrhagic Escherichia coli O157:H7 infection. Pathog Dis 2020; 77:5538763. [PMID: 31344225 DOI: 10.1093/femspd/ftz036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/22/2019] [Indexed: 12/28/2022] Open
Abstract
Susceptibility to enterohemorrhagic Escherichia coli (EHEC) infection varies among humans. The intestinal microbiota seems to play an essential role in host defense against EHEC; thus, we hypothesized that indigenous bacteria, such as Clostridium ramosum and Clostridium perfringens, could influence the susceptibility to EHEC infection. To evaluate the effect of indigenous bacteria on EHEC infection, germ-free mice were precolonized with each indigenous bacterium, and then infected with EHEC O157:H7. Precolonization with C. ramosum or C. perfringens completely prevented death from EHEC infection througout a test period. Precolonization with C. ramosum also reduced the level of secreted Shiga toxin (Stx) 2 and prevented histopathological changes in the kidneys in a similar way to precolonization with Bifidobacterium longum, which is used as a model for preventing EHEC infection. In contrast, the mice precolonized with C. perfringens showed mild renal injuries. When evaluated using an in vitro co-culturing system, again C. ramosum inhibited the growth and Stx production of EHEC more potently than C. perfringens. These results indicate that C. ramosum and C. perfringens suppressed EHEC infection; however, the extent of their preventive effects differed. Therefore, the susceptibility to EHEC infection and its severity can depend on the functional bacteria present in the intestinal microbiota of individuals.
Collapse
Affiliation(s)
- Yukako Koyanagi
- Laboratory of Animal Microbiology, Department of Microbial Biotechnology, Graduate School of Agricultural Science, Tohoku University, 468-1, Aramaki Aza Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Rie Suzuki
- Laboratory of Animal Microbiology, Department of Microbial Biotechnology, Graduate School of Agricultural Science, Tohoku University, 468-1, Aramaki Aza Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Kohei Ihara
- Laboratory of Animal Microbiology, Department of Microbial Biotechnology, Graduate School of Agricultural Science, Tohoku University, 468-1, Aramaki Aza Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Hikaru Miyagi
- Laboratory of Animal Microbiology, Department of Microbial Biotechnology, Graduate School of Agricultural Science, Tohoku University, 468-1, Aramaki Aza Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Hiroshi Isogai
- Animal Research Center, Sapporo Medical University, Minami 1, Nishi 16, Sapporo 060-8556, Japan
| | - Hiroshi Yoneyama
- Laboratory of Animal Microbiology, Department of Microbial Biotechnology, Graduate School of Agricultural Science, Tohoku University, 468-1, Aramaki Aza Aoba, Aoba-ku, Sendai 980-0845, Japan
| | - Emiko Isogai
- Laboratory of Animal Microbiology, Department of Microbial Biotechnology, Graduate School of Agricultural Science, Tohoku University, 468-1, Aramaki Aza Aoba, Aoba-ku, Sendai 980-0845, Japan
| |
Collapse
|
9
|
Aw W, Fukuda S. Protective effects of bifidobacteria against enteropathogens. Microb Biotechnol 2019; 12:1097-1100. [PMID: 31284329 PMCID: PMC6801159 DOI: 10.1111/1751-7915.13460] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 06/21/2019] [Indexed: 12/21/2022] Open
Abstract
Recent major advances in metagenomics and metabolomics technologies have enabled us to collect more data on the gut microbiome and metabolome to evaluate its influence on host health. In this short opinion article, we have chosen to focus on summarizing the protective mechanisms of bifidobacteria, a highly regarded probiotic, and it's metabolite: acetate; against enteropathogens, specifically in the E. coli O157:H7 mice model. We advocate for using a novel approach metabologenomics, which is an integration of metagenomic and metabolomic information on a systems biology-wide approach to better understand this interplay between gut microbiome and host metabolism.
Collapse
Affiliation(s)
- Wanping Aw
- Institute for Advanced BiosciencesKeio University246‐2 Mizukami, KakuganjiTsuruokaYamagata997‐0052Japan
| | - Shinji Fukuda
- Institute for Advanced BiosciencesKeio University246‐2 Mizukami, KakuganjiTsuruokaYamagata997‐0052Japan
- Intestinal Microbiota ProjectKanagawa Institute of Industrial Science and Technology3‐25‐13 Tonomachi, Kawasaki‐kuKawasakiKanagawa210‐0821Japan
- Transborder Medical Research CenterUniversity of Tsukuba1‐1‐1 TennodaiTsukubaIbaraki305‐8575Japan
- PRESTOJapan Science and Technology Agency4‐1‐8 Honcho KawaguchiSaitama332‐0012Japan
| |
Collapse
|
10
|
Inhibition of enterohemorrhagic Escherichia coli O157:H7 infection in a gnotobiotic mouse model with pre-colonization by Bacteroides strains. Biomed Rep 2019; 10:175-182. [PMID: 30906546 PMCID: PMC6403472 DOI: 10.3892/br.2019.1193] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/04/2019] [Indexed: 12/11/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 has been known to cause outbreaks of hemorrhagic colitis and hemolytic uremic syndrome. We previously demonstrated that intestinal flora contribute to the prevention of EHEC infection in a mouse model. However, it has not yet been determined whether Bacteroides, a predominant genus in the human intestine, contributes to the prevention of EHEC infection. The aim of the present study was to investigate the effect of Bacteroides fragilis (B. fragilis) and Bacteroides vulgatus (B. vulgatus) on EHEC O157:H7 infection in vivo using gnotobiotic mice. These strains were inoculated into germ-free mice to create a gnotobiotic mouse model. EHEC was inoculated into the mice, which were then monitored for 7 days for any change in symptoms. The mice that had been pre-colonized with the Bacteroides strains did not develop lethal EHEC infection, although several inflammatory symptoms were observed in the B. vulgatus pre-colonized group. However, no inflammatory symptoms were identified in the B. fragilis pre-colonized group. Moreover, B. fragilis exerted an inhibitory effect on enterocyte-like cell apoptosis. B. fragilis protected HT29 cells from apoptosis caused by Shiga toxin. In conclusion, the findings of the present study demonstrated that colonization by Bacteroides strains can inhibit EHEC infection.
Collapse
|
11
|
Wang H, Zhang S, Shen Q, Zhu MJ. A metabolomic explanation on beneficial effects of dietary Goji on intestine inflammation. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
12
|
Gut AM, Vasiljevic T, Yeager T, Donkor ON. Salmonella infection - prevention and treatment by antibiotics and probiotic yeasts: a review. MICROBIOLOGY-SGM 2018; 164:1327-1344. [PMID: 30136920 DOI: 10.1099/mic.0.000709] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Global Salmonella infection, especially in developing countries, is a health and economic burden. The use of antibiotic drugs in treating the infection is proving less effective due to the alarming rise of antibiotic-resistant strains of Salmonella, the effects of antibiotics on normal gut microflora and antibiotic-associated diarrhoea, all of which bring a growing need for alternative treatments, including the use of probiotic micro-organisms. However, there are issues with probiotics, including their potential to be opportunistic pathogens and antibiotic-resistant carriers, and their antibiotic susceptibility if used as complementary therapy. Clinical trials, animal trials and in vitro investigations into the prophylactic and therapeutic efficacies of probiotics have demonstrated antagonistic properties against Salmonella and other enteropathogenic bacteria. Nonetheless, there is a need for further studies into the potential mechanisms, efficacy and mode of delivery of yeast probiotics in Salmonella infections. This review discusses Salmonella infections and treatment using antibiotics and probiotics.
Collapse
Affiliation(s)
- Abraham Majak Gut
- 1Institute for Sustainable Industries and Livable Cities, College of Health and Biomedicine, Victoria University, Werribee Campus, PO Box 14428, Melbourne, Victoria 8001, Australia
| | - Todor Vasiljevic
- 1Institute for Sustainable Industries and Livable Cities, College of Health and Biomedicine, Victoria University, Werribee Campus, PO Box 14428, Melbourne, Victoria 8001, Australia
| | - Thomas Yeager
- 2Institute for Sustainable Industries and Livable Cities, College of Engineering and Science, Victoria University, Werribee Campus, PO Box 14428, Melbourne, Victoria 8001, Australia
| | - Osaana N Donkor
- 1Institute for Sustainable Industries and Livable Cities, College of Health and Biomedicine, Victoria University, Werribee Campus, PO Box 14428, Melbourne, Victoria 8001, Australia
| |
Collapse
|
13
|
Hampe CS, Roth CL. Probiotic strains and mechanistic insights for the treatment of type 2 diabetes. Endocrine 2017; 58:207-227. [PMID: 29052181 DOI: 10.1007/s12020-017-1433-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 09/20/2017] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The intestinal microbial composition appears to differ between healthy controls and individuals with Type 2 diabetes (T2D). This observation has led to the hypothesis that perturbations of the intestinal microbiota may contribute to the development of T2D. Manipulations of the intestinal microbiota may therefore provide a novel approach in the prevention and treatment of T2D. Indeed, fecal transplants have shown promising results in both animal models for obesity and T2D and in human clinical trials. To avoid possible complications associated with fecal transplants, probiotics are considered as a viable alternative therapy. An important, however often underappreciated, characteristic of probiotics is that individual strains may have different, even opposing, effects on the host. This strain specificity exists also within the same species. A comprehensive understanding of the underlying mechanisms at the strain level is therefore crucial for the selection of suitable probiotic strains. PURPOSE The aim of this review is to discuss the mechanisms employed by specific probiotic strains of the Lactobacillus and the Bifidobacterium genuses, which showed efficacy in the treatment of obesity and T2D. Some probiotic strains employ recurring beneficial effects, including the production of anti-microbial lactic acid, while other strains display highly unique features, such as hydrolysis of tannins. CONCLUSION A major obstacle in the evaluation of probiotic strains lays in the great number of strains, differences in detection methodology and measured outcome parameters. The understanding of further research should be directed towards the development of standardized evaluation methods to facilitate the comparison of different studies.
Collapse
Affiliation(s)
- Christiane S Hampe
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, 98109, USA.
| | - Christian L Roth
- Center for Integrative Brain Research, Seattle Children's Hospital & Research Institute, Seattle, WA, 98101, USA
- Pediatric Endocrinology, Seattle Children's Hospital & Research Institute, Seattle, WA, 98101, USA
| |
Collapse
|
14
|
Current Status of the Preharvest Application of Pro- and Prebiotics to Farm Animals to Enhance the Microbial Safety of Animal Products. Microbiol Spectr 2017; 5. [PMID: 28185614 DOI: 10.1128/microbiolspec.pfs-0012-2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The selection of microorganisms that act as probiotics and feed additives that act as prebiotics is an ongoing research effort, but a sizable range of commercial pro-, pre- and synbiotic (combining pro- and prebiotics) products are already available and being used on farms. A survey of the composition of commercial products available in the United States revealed that Lactobacillus acidophilus, Enterococcus faecium, and Bacillus subtilis were the three most common species in probiotic products. Of the nearly 130 probiotic products (also called direct-fed microbials) for which information was available, about 50 also contained yeasts or molds. The focus on these particular bacteria and eukaryotes is due to long-standing ideas about the benefits of such strains, research data on effectiveness primarily in laboratory or research farm settings, and regulations that dictate which microorganisms or feed additives can be administered to farm animals. Of the direct-fed microbials, only six made a claim relating to food safety or competitive exclusion of pathogens. None of the approximately 50 prebiotic products mentioned food safety in their descriptions. The remainder emphasized enhancement of animal performance such as weight gain or overall animal health. The reason why so few products carry food safety-related claims is the difficulties in establishing unambiguous cause and effect relationships between the application of such products in varied and constantly changing farm environments and improved food safety of the end product.
Collapse
|
15
|
Cordonnier C, Thévenot J, Etienne-Mesmin L, Alric M, Livrelli V, Blanquet-Diot S. Probiotic and enterohemorrhagic Escherichia coli: An effective strategy against a deadly enemy? Crit Rev Microbiol 2016; 43:116-132. [PMID: 27798976 DOI: 10.1080/1040841x.2016.1185602] [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: 12/15/2022]
Abstract
Enterohemorrhagic Escherichia coli (EHEC) are major food-borne pathogens that constitute a serious public health threat. Currently, there is no specific treatment available for EHEC infections in human creating an urgent need for the development of alternative therapeutic strategies. Among them, one of the most promising approaches is the use of probiotic microorganisms. Even if many studies have shown the antagonistic effects of probiotic bacteria or yeast on EHEC survival, virulence, adhesion on intestinal epithelium or pathogen-induced inflammatory responses, mechanisms mediating their beneficial effects remain unclear. This review describes EHEC pathogenesis and novel therapeutic strategies, with a particular emphasis on probiotics. The interests and limits of a probiotic-based approach and the way it might be incorporated into global health strategies against EHEC infections will be discussed.
Collapse
Affiliation(s)
- Charlotte Cordonnier
- a EA 4678 CIDAM, "Conception, Ingénierie et Développement de l'Aliment et du Médicament", Centre de Recherche en Nutrition Humaine Auvergne , Université d'Auvergne , Clermont-Ferrand , France.,b M2iSH, "Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte" , UMR Inserm/Université d'Auvergne U1071, USC-INRA 2018, Centre de Recherche en Nutrition Humaine Auvergne, Université d'Auvergne , Clermont-Ferrand , France
| | - Jonathan Thévenot
- a EA 4678 CIDAM, "Conception, Ingénierie et Développement de l'Aliment et du Médicament", Centre de Recherche en Nutrition Humaine Auvergne , Université d'Auvergne , Clermont-Ferrand , France.,b M2iSH, "Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte" , UMR Inserm/Université d'Auvergne U1071, USC-INRA 2018, Centre de Recherche en Nutrition Humaine Auvergne, Université d'Auvergne , Clermont-Ferrand , France
| | - Lucie Etienne-Mesmin
- a EA 4678 CIDAM, "Conception, Ingénierie et Développement de l'Aliment et du Médicament", Centre de Recherche en Nutrition Humaine Auvergne , Université d'Auvergne , Clermont-Ferrand , France.,b M2iSH, "Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte" , UMR Inserm/Université d'Auvergne U1071, USC-INRA 2018, Centre de Recherche en Nutrition Humaine Auvergne, Université d'Auvergne , Clermont-Ferrand , France
| | - Monique Alric
- a EA 4678 CIDAM, "Conception, Ingénierie et Développement de l'Aliment et du Médicament", Centre de Recherche en Nutrition Humaine Auvergne , Université d'Auvergne , Clermont-Ferrand , France
| | - Valérie Livrelli
- b M2iSH, "Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte" , UMR Inserm/Université d'Auvergne U1071, USC-INRA 2018, Centre de Recherche en Nutrition Humaine Auvergne, Université d'Auvergne , Clermont-Ferrand , France.,c Service de Bactériologie , CHU Clermont-Ferrand , Clermont-Ferrand , France
| | - Stéphanie Blanquet-Diot
- a EA 4678 CIDAM, "Conception, Ingénierie et Développement de l'Aliment et du Médicament", Centre de Recherche en Nutrition Humaine Auvergne , Université d'Auvergne , Clermont-Ferrand , France
| |
Collapse
|
16
|
Kwak MJ, Kwon SK, Yoon JK, Song JY, Seo JG, Chung MJ, Kim JF. Evolutionary architecture of the infant-adapted group of Bifidobacterium species associated with the probiotic function. Syst Appl Microbiol 2016; 39:429-439. [PMID: 27524178 DOI: 10.1016/j.syapm.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 01/11/2023]
Abstract
Bifidobacteria, often associated with the gastrointestinal tract of animals, are well known for their roles as probiotics. Among the dozens of Bifidobacterium species, Bifidobacterium bifidum, B. breve, and B. longum are the ones most frequently isolated from the feces of infants and known to help the digestion of human milk oligosaccharides. To investigate the correlation between the metabolic properties of bifidobacteria and their phylogeny, we performed a phylogenomic analysis based on 452 core genes of forty-four completely sequenced Bifidobacterium species. Results show that a major evolutionary event leading to the clade of the infant-adapted species is linked to carbohydrate metabolism, but it is not the only factor responsible for the adaptation of bifidobacteria to the gut. The genome of B. longum subsp. infantis, a typical bifidobacterium in the gut of breast-fed infants, encodes proteins associated with several kinds of species-specific metabolic pathways, including urea metabolism and biosynthesis of riboflavin and lantibiotics. Our results demonstrate that these metabolic features, which are associated with the probiotic function of bifidobacteria, are species-specific and highly correlate with their phylogeny.
Collapse
Affiliation(s)
- Min-Jung Kwak
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Soon-Kyeong Kwon
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jae-Kyung Yoon
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ju Yeon Song
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jae-Gu Seo
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - Myung Jun Chung
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - Jihyun F Kim
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Strategic Initiative for Microbiomes in Agriculture and Food, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
17
|
Affiliation(s)
- Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| |
Collapse
|
18
|
Hostetter SJ, Helgerson AF, Paton JC, Paton AW, Cornick NA. Therapeutic use of a receptor mimic probiotic reduces intestinal Shiga toxin levels in a piglet model of hemolytic uremic syndrome. BMC Res Notes 2014; 7:331. [PMID: 24890228 PMCID: PMC4049369 DOI: 10.1186/1756-0500-7-331] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 05/26/2014] [Indexed: 12/31/2022] Open
Abstract
Background Hemolytic uremic syndrome (HUS) is a systemic and potentially fatal complication of gastroenteritis secondary to Shiga toxin-producing enterohemorrhagic Escherichia coli (EHEC) infection characterized by microangiopathic hemolytic anemia, thrombocytopenia, and acute renal damage. Shiga toxin (Stx), the toxin principle in HUS, is produced locally within the gut following EHEC colonization and is disseminated via the vasculature. Clinical development of HUS currently has no effective treatment and is a leading cause of renal failure in children. Novel post-exposure therapies are currently needed for HUS; therefore, the purpose of this study was to investigate the efficacy of a Stx receptor mimic probiotic in a porcine model of HUS. Edema disease, an infection of swine caused by host adapted Shiga toxin-producing Escherichia coli (STEC) and mediated by Shiga toxin 2e (Stx2e), shares many pathogenic similarities to HUS. In this study, three-week old piglets were inoculated with STEC and 24 hours later treated twice daily with a probiotic expressing an oligosaccharide receptor mimic for Stx2e to determine if the probiotic could reduce intestinal toxin levels. Methods Piglets were orally inoculated with 1010 CFU of STEC strain S1191 eight days after weaning. Beginning day 1 post-inoculation, piglets were treated orally twice daily with 5 × 1011 CFU of either the receptor mimic probiotic or a sham probiotic for 10 days. Intestinal Stx2e levels were assessed daily via Vero cell assay. The efficacy of the probiotic at reducing intestinal Stx2e, vascular lesions, and clinical disease was evaluated with repeated measures ANOVA and Fisher’s exact test as appropriate. Results The probiotic significantly reduced intestinal Stx2e, as reflected by decreased fecal toxin titers on days 3–8 post-inoculation (p < 0.01). Despite this reduction in intestinal toxin levels, however, the probiotic failed to reduce the incidence of vascular necrosis in target organs and had no effect on clinical disease. Conclusions The data suggest that post-exposure treatment with a Stx-binding probiotic is effective in reducing intestinal toxin burden. Future studies could target this approach for possible development of post-exposure interventions.
Collapse
Affiliation(s)
- Shannon J Hostetter
- Department of Veterinary Pathology, Iowa State University, 1600 S, 16th Street, Ames, IA 50011-1250, USA.
| | | | | | | | | |
Collapse
|
19
|
Commensal bacteria mediated defenses against pathogens. Curr Opin Immunol 2014; 29:16-22. [PMID: 24727150 DOI: 10.1016/j.coi.2014.03.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 03/13/2014] [Indexed: 12/20/2022]
Abstract
Commensal bacterial communities residing within the intestinal lumen of mammals have evolved to flourish in this microenvironment. To preserve this niche, commensal bacteria act with the host to prevent colonization by invasive pathogens that induce inflammation and disrupt the intestinal niche commensal bacteria occupy. Thus, it is mutually beneficial to the host and commensal bacteria to inhibit a pathogen's ability to establish an infection. Commensal bacteria express factors that support colonization, maximize nutrient uptake, and produce metabolites that confer a survival advantage over pathogens. Further, commensal bacteria stimulate the host's immune defenses and drive tonic expression of anti-microbial factors. In combination, these mechanisms preserve the niche for commensal bacteria and assist the host in preventing infection.
Collapse
|
20
|
Fukuda S, Toh H, Taylor TD, Ohno H, Hattori M. Acetate-producing bifidobacteria protect the host from enteropathogenic infection via carbohydrate transporters. Gut Microbes 2013; 3:449-54. [PMID: 22825494 DOI: 10.4161/gmic.21214] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The human gut harbors a large and diverse community of commensal bacteria. Among them, Bifidobacterium is known to exhibit various probiotic effects including protection of hosts from infectious diseases. We recently discovered that genes encoding an ATP-binding-cassette-type carbohydrate transporter present in certain bifidobacteria contribute to protecting gnotobiotic mice from death induced by enterohemorrhagic Escherichia coli O157:H7. We elucidated the molecular mechanism on lethal infection in mice associated with several bifidobacterial strains by a multi-omics approach combining genomics, transcriptomics and metabolomics. The combined data clearly show that acetate produced by protective bifidobacteria acts in vivo to promote defense functions of the host epithelial cells and thereby protects the host from lethal infection. As demonstrated here, our multi-omics approach provides a powerful strategy for evaluation of host-microbial interactions in the complex gut ecosystem.
Collapse
Affiliation(s)
- Shinji Fukuda
- Laboratory for Epithelial Immunobiology, RIKEN Research Center for Allergy and Immunology, Yokohama, Japan
| | | | | | | | | |
Collapse
|
21
|
Pang G, Xie J, Chen Q, Hu Z. How functional foods play critical roles in human health. FOOD SCIENCE AND HUMAN WELLNESS 2012. [DOI: 10.1016/j.fshw.2012.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
22
|
Long-term changes in human colonic Bifidobacterium populations induced by a 5-day oral amoxicillin-clavulanic acid treatment. PLoS One 2012; 7:e50257. [PMID: 23209691 PMCID: PMC3507739 DOI: 10.1371/journal.pone.0050257] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 10/17/2012] [Indexed: 01/01/2023] Open
Abstract
The objective of this study was to assess the possible modifications due to amoxicillin-clavulanic acid (AMC) treatment on total bacteria and on Bifidobacterium species balance in human colonic microbiota. Eighteen healthy volunteers (19 to 36 years old) were given a 875/125 mg dose of AMC twice a day for 5 days. Fecal samples were obtained before and after antibiotic exposure. After total DNA extraction, total bacteria and bifidobacteria were specifically quantified using real-time PCR. Dominant species were monitored over time using bacterial and bifidobacterial Temporal Temperature Gradient gel Electrophoresis (TTGE). At the end of AMC exposure, total bacterial concentrations as well as bifidobacteria concentrations were significantly reduced compared to before AMC exposure:10.7±0.1 log10 16S rRNA gene copies/g vs 11.1±0.1 log10 (p = 0.003) and 8.1±0.5 log10 16S rRNA gene copies/g vs 9.4±0.3 log10 (p = 0.003), respectively. At the same time, the mean similarity percentages of TTGE bacteria and TTGE bifidobacteria profiles were significantly reduced compared to before AMC exposure: 51.6%±3.5% vs 81.4%±2.1% and 55.8%±7.6% vs 84.5%±4.1%, respectively. Occurrence of B. adolescentis, B. bifidum and B. pseudocatenulatum/B. catenulatum species significantly decreased. Occurrence of B. longum remained stable. Moreover, the number of distinct Bifidobacterium species per sample significantly decreased (1.5±0.3 vs 2.3±0.3; p = 0.01). Two months after AMC exposure, the mean similarity percentage of TTGE profiles was 55.6% for bacteria and 62.3% for bifidobacteria. These results clearly demonstrated that a common antibiotic treatment may qualitatively alter the colonic microbiota. Such modifications may have potential long-term physiological consequences.
Collapse
|
23
|
Zeinhom M, Tellez AM, Delcenserie V, El-Kholy AM, El-Shinawy SH, Griffiths MW. Yogurt containing bioactive molecules produced by Lactobacillus acidophilus La-5 exerts a protective effect against enterohemorrhagic Escherichia coli in mice. J Food Prot 2012; 75:1796-805. [PMID: 23043828 DOI: 10.4315/0362-028x.jfp-11-508] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
An active fraction extracted from Lactobacillus acidophilus La5 cell-free spent medium (LAla-5AF) was incorporated in a dairy matrix and tested to assess its antivirulent effect against enterohemorrhagic Escherichia coli (EHEC). Mice in experimental groups were fed for 4 days with yogurt supplemented with LAla-5AF. On the fifth day, mice were challenged with a single dose (10(7) CFU per mouse) of E. coli O157:H7. The clinical manifestations of the infection were significantly less severe in mice fed the yogurt supplemented with LAla-5AF. EHEC attachment and colonization was attenuated by LAla-5AF. Tumor necrosis factor alpha production was down-regulated, which might indicate a protective effect in the kidney during EHEC infection. To investigate the mechanisms associated with the in vivo effects observed, LAla-5AF was tested by reverse transcription real-time PCR to confirm its effects on the expression of several virulence genes of EHEC O157. The results showed that these fractions were able to down-regulate several virulence genes of EHEC, including stxB2, qseA, luxS, tir, ler, eaeA, and hlyB.
Collapse
Affiliation(s)
- Mohamed Zeinhom
- Food Hygiene & Control Department, Faculty of Veterinary Medicine, Beni-Suef University, Egypt
| | | | | | | | | | | |
Collapse
|
24
|
Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM, Topping DL, Suzuki T, Taylor TD, Itoh K, Kikuchi J, Morita H, Hattori M, Ohno H. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011. [PMID: 21270894 DOI: 10.1083/nature09646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The human gut is colonized with a wide variety of microorganisms, including species, such as those belonging to the bacterial genus Bifidobacterium, that have beneficial effects on human physiology and pathology. Among the most distinctive benefits of bifidobacteria are modulation of host defence responses and protection against infectious diseases. Nevertheless, the molecular mechanisms underlying these effects have barely been elucidated. To investigate these mechanisms, we used mice associated with certain bifidobacterial strains and a simplified model of lethal infection with enterohaemorrhagic Escherichia coli O157:H7, together with an integrated 'omics' approach. Here we show that genes encoding an ATP-binding-cassette-type carbohydrate transporter present in certain bifidobacteria contribute to protecting mice against death induced by E. coli O157:H7. We found that this effect can be attributed, at least in part, to increased production of acetate and that translocation of the E. coli O157:H7 Shiga toxin from the gut lumen to the blood was inhibited. We propose that acetate produced by protective bifidobacteria improves intestinal defence mediated by epithelial cells and thereby protects the host against lethal infection.
Collapse
Affiliation(s)
- Shinji Fukuda
- Laboratory for Epithelial Immunobiology, RIKEN Research Center for Allergy and Immunology, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011; 469:543-7. [PMID: 21270894 DOI: 10.1038/nature09646] [Citation(s) in RCA: 1586] [Impact Index Per Article: 122.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/04/2010] [Indexed: 02/06/2023]
Abstract
The human gut is colonized with a wide variety of microorganisms, including species, such as those belonging to the bacterial genus Bifidobacterium, that have beneficial effects on human physiology and pathology. Among the most distinctive benefits of bifidobacteria are modulation of host defence responses and protection against infectious diseases. Nevertheless, the molecular mechanisms underlying these effects have barely been elucidated. To investigate these mechanisms, we used mice associated with certain bifidobacterial strains and a simplified model of lethal infection with enterohaemorrhagic Escherichia coli O157:H7, together with an integrated 'omics' approach. Here we show that genes encoding an ATP-binding-cassette-type carbohydrate transporter present in certain bifidobacteria contribute to protecting mice against death induced by E. coli O157:H7. We found that this effect can be attributed, at least in part, to increased production of acetate and that translocation of the E. coli O157:H7 Shiga toxin from the gut lumen to the blood was inhibited. We propose that acetate produced by protective bifidobacteria improves intestinal defence mediated by epithelial cells and thereby protects the host against lethal infection.
Collapse
|
26
|
Nakanishi Y, Fukuda S, Chikayama E, Kimura Y, Ohno H, Kikuchi J. Dynamic omics approach identifies nutrition-mediated microbial interactions. J Proteome Res 2010; 10:824-36. [PMID: 21058740 DOI: 10.1021/pr100989c] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
"Omics" studies reported to date have dealt with either thoroughly characterized single species or poorly explored meta-microbial communities. However, these techniques are capable of producing highly informative data for the analysis of interactions between two organisms. We examined the bacterial interaction between Escherichia coli O157:H7 (O157) and Bifidobacterium longum (BL) as a pathogenic-commensal bacterial model creating a minimum microbial ecosystem in the gut using dynamic omics approaches, consisting of improved time-lapse 2D-nuclear magnetic resonance (NMR) metabolic profiling, transcriptomic, and proteomic analyses. Our study revealed that the minimum ecosystem was established by bacterial adaptation to the changes in the extracellular environment, primarily by O157, but not by BL. Additionally, the relationship between BL and O157 could be partially regarded as that between a producer and a consumer of nutrients, respectively, especially with regard to serine and aspartate metabolism. Taken together, our profiling system can provide a new insight into the primary metabolic dynamics in microbial ecosystems.
Collapse
Affiliation(s)
- Yumiko Nakanishi
- Graduate School of Nanobiosciences, Yokohama City University, Yokohama, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Rahal EA, Kazzi N, Kanbar A, Abdelnoor AM, Matar GM. Role of rifampicin in limiting Escherichia coli O157:H7 Shiga-like toxin expression and enhancement of survival of infected BALB/c mice. Int J Antimicrob Agents 2010; 37:135-9. [PMID: 21129928 DOI: 10.1016/j.ijantimicag.2010.10.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 09/06/2010] [Accepted: 10/06/2010] [Indexed: 10/18/2022]
Abstract
The sequelae of infection with Escherichia coli O157:H7 include the potentially fatal haemolytic uraemic syndrome. The pathobiological process of E. coli O157:H7 is chiefly dependent on the production of Shiga-like toxins I and II (SLT-I and -II). Antibiotic treatment is currently refrained from since it may lead to enhanced release of SLTs from the bacterium. In this study, the potential utility of rifampicin in treating E. coli O157:H7 infections was assessed both in vitro and in vivo. Five strains of E. coli O157:H7 were tested by reverse transcriptase polymerase chain reaction (RT-PCR) for the transcription of the SLT-I- and SLT-II-encoding genes (stx1 and stx2, respectively). Treatment of bacterial strains with the rifampicin minimal inhibitory concentration (MIC), minimal bactericidal concentration (MBC), or the MIC followed by the MBC led to the inhibition of stx1 and stx2 gene transcription. Treatment with the MIC or with the MIC followed by the MBC was also capable of limiting toxin release. SLT-I and SLT-II detection by reverse passive latex agglutination showed an effective decrease in toxin titres following treatment with the MIC of rifampicin or with the MIC followed by the MBC. Treatment of cultures with the MBC alone was not as effective in decreasing toxin titres. The efficacy of rifampicin in treating E. coli O157:H7-infected BALB/c mice was also assessed. Rifampicin treatment resulted in enhanced mouse survival and limited the weight loss of infected animals. In conclusion, both in vitro and in vivo tests showed that rifampicin may be useful in treating E. coli O157:H7 infection.
Collapse
Affiliation(s)
- Elias A Rahal
- Department of Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | | | | |
Collapse
|
28
|
Probiotic Lactobacillus reuteri ameliorates disease due to enterohemorrhagic Escherichia coli in germfree mice. Infect Immun 2010; 79:185-91. [PMID: 20974822 DOI: 10.1128/iai.00880-10] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Strains of enterohemorrhagic Escherichia coli (EHEC) are a group of Shiga toxin-producing food-borne pathogens that cause severe hemorrhagic colitis and can lead to hemolytic-uremic syndrome (HUS), a life-threatening condition that principally affects children and for which there is no effective treatment. We used a germfree mouse model of renal and enteric disease due to EHEC to determine if probiotic Lactobacillus reuteri ATCC PTA 6475 is effective in suppressing disease symptoms caused by EHEC. When germfree Swiss Webster mice are monocolonized with EHEC, they develop disease characterized by weight loss, cecal luminal fluid accumulation, and renal tubular necrosis. When L. reuteri was administered 1 day prior to EHEC challenge and every other day thereafter, EHEC colonization was suppressed and mice were significantly protected from the manifestations of disease. Protection from disease did not require the induction of the antimicrobial compound reuterin in L. reuteri prior to treatment. The twice-daily administration of L. reuteri appeared more effective than every-other-day administration. These data indicated that L. reuteri partially protects mice from disease manifestations of EHEC.
Collapse
|