1
|
Radford GA, Vrbanac L, de Nys RT, Worthley DL, Wright JA, Hasty J, Woods SL. Towards Understanding Tumour Colonisation by Probiotic Bacterium E. coli Nissle 1917. Cancers (Basel) 2024; 16:2971. [PMID: 39272829 PMCID: PMC11394440 DOI: 10.3390/cancers16172971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The last decade has seen a rapid increase in studies utilising a genetically modified probiotic, Escherichia coli Nissle 1917 (EcN), as a chassis for cancer treatment and detection. This approach relies on the ability of EcN to home to and selectively colonise tumours over normal tissue, a characteristic common to some bacteria that is thought to result from the low-oxygen, nutrient-rich and immune-privileged niche the tumour provides. Pre-clinical studies have used genetically modified EcN to deliver therapeutic payloads that show efficacy in reducing tumour burden as a result of high-tumour and low off-target colonisation. Most recently, the EcN chassis has been expanded into an effective tumour-detection tool. These advances provide strong justification for the movement of genetically modified EcN into clinical oncology trials. What is currently unknown in the field is a deep mechanistic understanding of how EcN distributes to and localises within tumours. This review summarises the existing EcN literature, with the inclusion of research undertaken with other tumour-homing and pathogenic bacteria, to provide insights into possible mechanisms of EcN tumour homing for future validation. Understanding exactly how and why EcN colonises neoplastic tissue will inform the design and testing of the next generation of EcN chassis strains to address biosafety and containment concerns and optimise the detection and treatment of cancer.
Collapse
Affiliation(s)
- Georgette A Radford
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Laura Vrbanac
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Rebekah T de Nys
- Precision Cancer Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| | | | - Josephine A Wright
- Precision Cancer Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| | - Jeff Hasty
- Synthetic Biology Institute, University of California, San Diego, CA 92093, USA
- Department of Bioengineering, University of California, San Diego, CA 92093, USA
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, CA 92093, USA
- Center for Microbiome Innovation, University of California, San Diego, CA 92093, USA
| | - Susan L Woods
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| |
Collapse
|
2
|
Hudcovic T, Petr Hermanova P, Kozakova H, Benada O, Kofronova O, Schwarzer M, Srutkova D. Priority order of neonatal colonization by a probiotic or pathogenic Escherichia coli strain dictates the host response to experimental colitis. Front Microbiol 2024; 15:1393732. [PMID: 39206364 PMCID: PMC11349737 DOI: 10.3389/fmicb.2024.1393732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
The alarming prevalence of inflammatory bowel disease (IBD) in early childhood is associated with imbalances in the microbiome, the immune response, and environmental factors. Some pathogenic Escherichia coli (E. coli) strains have been found in IBD patients, where they may influence disease progression. Therefore, the discovery of new harmful bacterial strains that have the potential to drive the inflammatory response is of great importance. In this study, we compared the immunomodulatory properties of two E. coli strains of serotype O6: the probiotic E. coli Nissle 1917 and the uropathogenic E. coli O6:K13:H1. Using the epithelial Caco-2 cell line, we investigated the different abilities of the strains to adhere to and invade epithelial cells. We confirmed the potential of E. coli Nissle 1917 to modulate the Th1 immune response in a specific manner in an in vitro setting by stimulating mouse bone marrow-derived dendritic cells (BM-DCs). In gnotobiotic in vivo experiments, we demonstrated that neonatal colonization with E. coli Nissle 1917 achieves a stable high concentration in the intestine and protects mice from the progressive effect of E. coli O6:K13:H1 in developing ulcerative colitis in an experimental model. In contrast, a single-dose treatment with E. coli Nissle 1917 is ineffective in achieving such high concentrations and does not protect against DSS-induced ulcerative colitis in mice neonatally colonized with pathobiont E. coli O6:K13:H1. Despite the stable coexistence of both E. coli strains in the intestinal environment of the mice, we demonstrated a beneficial competitive interaction between the early colonizing E. coli Nissle 1917 and the late-arriving strain O6:K13:H1, suggesting its anti-inflammatory potential for the host. This study highlights the importance of the sequence of bacterial colonization, which influences the development of the immune response in the host gut and potentially impacts future quality of life.
Collapse
Affiliation(s)
- Tomas Hudcovic
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Nový Hrádek, Czechia
| | - Petra Petr Hermanova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Nový Hrádek, Czechia
| | - Hana Kozakova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Nový Hrádek, Czechia
| | - Oldrich Benada
- Laboratory of Molecular Structure Characterization, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Olga Kofronova
- Laboratory of Molecular Structure Characterization, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Nový Hrádek, Czechia
| | - Dagmar Srutkova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Nový Hrádek, Czechia
| |
Collapse
|
3
|
Mihailović M, Soković Bajić S, Arambašić Jovanović J, Brdarić E, Dinić S, Grdović N, Uskoković A, Rajić J, Đorđević M, Tolinački M, Golić N, Živković M, Vidaković M. Beneficial Effects of Probiotic Lactobacillus paraplantarum BGCG11 on Pancreatic and Duodenum Function in Diabetic Rats. Int J Mol Sci 2024; 25:7697. [PMID: 39062940 PMCID: PMC11277547 DOI: 10.3390/ijms25147697] [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: 06/04/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Diabetes mellitus, as a chronic metabolic disorder, significantly impacts the pancreas and among other organs, affects duodenal function. Emerging evidence suggests that probiotics can exert beneficial effects on gut health and metabolism. In our previous research, we evaluated the probiotic Lactobacillus paraplantarum BGCG11 primarily for its protective properties against diabetic rats' damaged liver and kidneys. In this work, we further examined the effects of probiotic strain BGCG11 on the function of the duodenum and pancreas in diabetic rats. We explored the potential mechanisms underlying the probiotic's effects, focusing on general indicators of diabetes, the architecture and morphology of pancreatic islets, duodenal integrity (measuring the transfer of fluid and serum zonulin level), and the modulation of gut microbiota composition. Our findings reveal the protective and regulatory roles of L. paraplantarum BGCG11 in mitigating diabetes-induced pancreatic and duodenal dysfunction regardless of its application time (pre- or post-treatment), highlighting its therapeutic potential in managing diabetes-related gastrointestinal complications.
Collapse
Affiliation(s)
- Mirjana Mihailović
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Svetlana Soković Bajić
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Jelena Arambašić Jovanović
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Emilija Brdarić
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Svetlana Dinić
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Nevena Grdović
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Aleksandra Uskoković
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Jovana Rajić
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Marija Đorđević
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| | - Maja Tolinački
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Nataša Golić
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Milica Živković
- Institute of Molecular Genetics and Genetics Engineering, University of Belgrade, Vojvode Stepe 444a, P.O. Box 23, 11010 Belgrade, Serbia (E.B.); (M.T.); (N.G.)
| | - Melita Vidaković
- Institute for Biological Research “Siniša Stanković”, University of Belgrade, Bulevar Despota Stefana 142, 10060 Belgrade, Serbia; (M.M.); (J.A.J.); (S.D.); (N.G.); (A.U.); (J.R.); (M.Đ.)
| |
Collapse
|
4
|
Willer T, Han Z, Pielsticker C, Rautenschlein S. In vitro investigations on interference of selected probiotic candidates with Campylobacter jejuni adhesion and invasion of primary chicken derived cecal and Caco-2 cells. Gut Pathog 2024; 16:30. [PMID: 38907359 PMCID: PMC11191211 DOI: 10.1186/s13099-024-00623-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Campylobacter (C.) jejuni is one of the most important bacterial foodborne pathogens worldwide. Probiotics such as Lactobacillus or Bacillus species are considered one option for reducing the colonization rate and magnitude in poultry, the most frequent source of human infections. Due to the lack of suitable avian in vitro models such as chicken intestinal cell lines, especially those derived from the cecum, most in vitro studies on C. jejuni host interaction have been conducted with human intestinal cell lines. In this study, we compared C. jejuni-cell interactions between primary chicken cecal cells and the human intestinal cell line Caco-2, which is derived from colorectal adenocarcinoma, and investigated possible interfering effects of selected probiotic candidates. RESULTS We detected differences in adhesion and invasion between the two tested gut cell types and between different C. jejuni strains. The probiotic inhibition of C. jejuni adhesion and invasion of human and avian gut cells was affected by host cell type, investigated C. jejuni strain and time points of probiotic treatment. Additionally, our results suggest a possible correlation between C. jejuni invasion and the detected increase in IL-6 mRNA expression. CONCLUSIONS Our results indicate distinct differences between avian and human gut cells in their interaction with C. jejuni. Therefore, data obtained in one host species on C. jejuni-host interaction may not easily be transferrable to another one. The factors influencing the variable efficacy of probiotic intervention in chicken and human derived cells should be investigated further.
Collapse
Affiliation(s)
- Thomas Willer
- Clinic for Poultry, University of Veterinary Medicine Hannover, Foundation, Buenteweg 17, 30559, Hanover, Germany
| | - Zifeng Han
- Clinic for Poultry, University of Veterinary Medicine Hannover, Foundation, Buenteweg 17, 30559, Hanover, Germany
| | - Colin Pielsticker
- Clinic for Poultry, University of Veterinary Medicine Hannover, Foundation, Buenteweg 17, 30559, Hanover, Germany
| | - Silke Rautenschlein
- Clinic for Poultry, University of Veterinary Medicine Hannover, Foundation, Buenteweg 17, 30559, Hanover, Germany.
| |
Collapse
|
5
|
Alhubail M, McBain AJ, O'Neill CA. A survey of multiple candidate probiotic bacteria reveals specificity in the ability to modify the effects of key wound pathogens. Microbiol Spectr 2024; 12:e0034724. [PMID: 38700333 PMCID: PMC11237428 DOI: 10.1128/spectrum.00347-24] [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: 02/06/2024] [Accepted: 04/12/2024] [Indexed: 05/05/2024] Open
Abstract
We have evaluated the inhibitory effects of supernatants and lysates derived from several candidate probiotics, on the growth and biofilm formation of wound pathogens, and their ability to protect human primary epidermal keratinocytes from the toxic effects of pathogens. Supernatants (neutralized and non-neutralized) and lysates (via sonication) from Lactiplantibacillus plantarum, Limosilactobacillus reuteri, Bifidobacterium longum, Lacticaseibacillus rhamnosus GG, and Escherichia coli Nissle 1917 were tested for their inhibitory effects against Staphylococcus aureus, Streptococcus pyogenes, Escherichia coli, Pseudomonas aeruginosa, and Acinetobacter baumanni. The supernatants of L. plantarum, L. rhamnosus, B. longum, and L. rhamnosus GG reduced the growth of S. aureus, E. coli, and A. baumanni. B. longum additionally inhibited P. aeruginosa growth. However, neutralized Lactobacillus supernatants did not inhibit growth and in some cases were stimulatory. Lysates of L. plantarum and L. reuteri inhibited S. pyogenes while B. longum lysates inhibited E. coli and S. aureus growth. E. coli Nissle 1917 lysates enhanced the growth of S. pyogenes and P. aeruginosa. Biofilm formation by E. coli was reduced by lysates of L. reuteri and neutralized supernatants of all candidate probiotics. P. aeruginosa biofilm formation was reduced by E. coli Nissle supernatant but increased by L. plantarum, L. reuteri, and Bifidobacterium longum lysates. L. reuteri decreased the toxic effects of S. aureus on keratinocytes while E. coli Nissle 1917 lysates protected keratinocytes from S. pyogenes toxicity. In conclusion, lactobacilli and E. coli Nissle lysates confer inhibitory effects on pathogenic growth independently of acidification and may beneficially alter the outcome of interactions between host cell-pathogen in a species-specific manner.IMPORTANCEOne of the attributes of probiotics is their ability to inhibit pathogens. For this reason, many lactobacilli have been investigated for their effects as potential topical therapeutics against skin pathogens. However, this field is in its infancy. Even though probiotics are known to be safe when taken orally, the potential safety concerns when applied to potentially compromised skin are unknown. For this reason, we believe that extracts of probiotics will offer advantages over the use of live bacteria. In this study, we have surveyed five candidate probiotics, when used as extracts, in terms of their effects against common wound pathogens. Our data demonstrate that some probiotic extracts promote the growth of pathogens and highlight the need for careful selection of species and strains when probiotics are to be used topically.
Collapse
Affiliation(s)
- Muna Alhubail
- Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Andrew J. McBain
- Faculty of Biology, School of Health Sciences, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Catherine A. O'Neill
- Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
6
|
Liu Y, Cheng YY, Thompson J, Zhou Z, Vivas EI, Warren MF, Rey FE, Anantharaman K, Venturelli OS. Shaping human gut community assembly and butyrate production by controlling the arginine dihydrolase pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.10.523442. [PMID: 37986770 PMCID: PMC10659395 DOI: 10.1101/2023.01.10.523442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The arginine dihydrolase pathway (arc operon) present in a subset of diverse human gut species enables arginine catabolism. This specialized metabolic pathway can alter environmental pH and nitrogen availability, which in turn could shape gut microbiota inter-species interactions. By exploiting synthetic control of gene expression, we investigated the role of the arc operon in probiotic Escherichia coli Nissle 1917 on human gut community assembly and health-relevant metabolite profiles in vitro and in the murine gut. By stabilizing environmental pH, the arc operon reduced variability in community composition across different initial pH perturbations. The abundance of butyrate producing bacteria were altered in response to arc operon activity and butyrate production was enhanced in a physiologically relevant pH range. While the presence of the arc operon altered community dynamics, it did not impact production of short chain fatty acids. Dynamic computational modeling of pH-mediated interactions reveals the quantitative contribution of this mechanism to community assembly. In sum, our framework to quantify the contribution of molecular pathways and mechanism modalities on microbial community dynamics and functions could be applied more broadly.
Collapse
Affiliation(s)
- Yiyi Liu
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison WI 53706
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Yu-Yu Cheng
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Jaron Thompson
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison WI 53706
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Zhichao Zhou
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
| | - Eugenio I. Vivas
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
- Gnotobiotic Animal Core Facility, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Matthew F. Warren
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
| | | | - Ophelia S. Venturelli
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison WI 53706
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
- Department of Bacteriology, University of Wisconsin-Madison, WI 53706
| |
Collapse
|
7
|
Ninyio N, Schmitt K, Sergon G, Nilsson C, Andersson S, Scherbak N. Stable expression of HIV-1 MPER extended epitope on the surface of the recombinant probiotic bacteria Escherichia Coli Nissle 1917 using CRISPR/Cas9. Microb Cell Fact 2024; 23:39. [PMID: 38311724 PMCID: PMC10840157 DOI: 10.1186/s12934-023-02290-0] [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: 10/17/2023] [Accepted: 12/28/2023] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Mucosal vaccines have the potential to induce protective immune responses at the sites of infection. Applying CRISPR/Cas9 editing, we aimed to develop a probiotic-based vaccine candidate expressing the HIV-1 envelope membrane-proximal external region (MPER) on the surface of E. coli Nissle 1917. RESULTS The HIV-1 MPER epitope was successfully introduced in the porin OmpF of the E. coli Nissle 1917 (EcN-MPER) and the modification was stable over 30 passages of the recombinant bacteria on the DNA and protein level. Furthermore, the introduced epitope was recognized by a human anti-HIV-1 gp41 (2F5) antibody using both live and heat-killed EcN-MPER, and this antigenicity was also retained over 30 passages. Whole-cell dot blot suggested a stronger binding of anti-HIV-1 gp41 (2F5) to heat-killed EcN-MPER than their live counterpart. An outer membrane vesicle (OMV) - rich extract from EcN-MPER culture supernatant was equally antigenic to anti-HIV-1 gp41 antibody which suggests that the MPER antigen could be harboured in EcN-MPER OMVs. Using quantitative ELISA, we determined the amount of MPER produced by the modified EcN to be 14.3 µg/108 cfu. CONCLUSIONS The CRISPR/Cas9 technology was an effective method for establishment of recombinant EcN-MPER bacteria that was stable over many passages. The developed EcN-MPER clone was devoid of extraneous plasmids and antibiotic resistance genes which eliminates the risk of plasmid transfer to animal hosts, should this clone be used as a vaccine. Also, the EcN-MPER clone was recognised by anti-HIV-1 gp41 (2F5) both as live and heat-killed bacteria making it suitable for pre-clinical evaluation. Expression of OmpF on bacterial surfaces and released OMVs identifies it as a compelling candidate for recombinant epitope modification, enabling surface epitope presentation on both bacteria and OMVs. By applying the methods described in this study, we present a potential platform for cost-effective and rational vaccine antigen expression and administration, offering promising prospects for further research in the field of vaccine development.
Collapse
Affiliation(s)
- Nathaniel Ninyio
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Katharina Schmitt
- School of Science and Technology, Life Science Center, Örebro University, Örebro, Sweden
- Institute of Virology, Saarland University Medical Center, 66421, Homburg, Germany
| | - Gladys Sergon
- School of Science and Technology, Life Science Center, Örebro University, Örebro, Sweden
| | - Charlotta Nilsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Microbiology, Public Health Agency of Sweden, Solna, Sweden
| | - Sören Andersson
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Public Health Analysis and Data Management, Unit for Vaccination Programmes, Public Health Agency of Sweden, Solna, Sweden
| | - Nikolai Scherbak
- School of Science and Technology, Life Science Center, Örebro University, Örebro, Sweden.
| |
Collapse
|
8
|
Hu M, Zhang T, Miao M, Li K, Luan Q, Sun G. Expectations for employing Escherichia coli Nissle 1917 in food science and nutrition. Crit Rev Food Sci Nutr 2024:1-9. [PMID: 38189668 DOI: 10.1080/10408398.2023.2301416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
As a promising probiotic strain, Escherichia coli Nissle 1917 (EcN) has been demonstrated to confer beneficial effects on intestinal health, immune function, and pathogen prevention. Additionally, EcN has also been widely studied due to its clear genomic information, tractable gene regulation, and simple growth conditions. This review summarizes the various applications potential of EcN in food science and nutrition, including inflammation prevention, tumor-targeting therapy, antibacterial agents for food, and nutrient production with a focus on specific case studies. Moreover, we highlight the major challenges of employing EcN in food science and nutrition, including regulatory approval, stability during food processing, and consumer acceptance. Finally, we conclude with a discussion on perspectives related to employing EcN in food science and nutrition.
Collapse
Affiliation(s)
- Miaomiao Hu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Ming Miao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Kewen Li
- Baolingbao Biology Co., Ltd, Yucheng, Shandong, China
| | - Qingmin Luan
- Baolingbao Biology Co., Ltd, Yucheng, Shandong, China
| | - Guilian Sun
- Baolingbao Biology Co., Ltd, Yucheng, Shandong, China
| |
Collapse
|
9
|
Dong MM, Song L, Xu JQ, Zhu L, Xiong LB, Wei DZ, Wang FQ. Improved cryptic plasmids in probiotic Escherichia coli Nissle 1917 for antibiotic-free pathway engineering. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12662-6. [PMID: 37405431 DOI: 10.1007/s00253-023-12662-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 07/06/2023]
Abstract
The engineered probiotic Escherichia coli Nissle 1917 (EcN) is expected to be employed in the diagnosis and treatment of various diseases. However, the introduced plasmids typically require antibiotics to maintain genetic stability, and the cryptic plasmids in EcN are usually eliminated to avoid plasmid incompatibility which may change the inherent probiotic characteristics. Here, we provided a simple design to minimize the genetic change of probiotics by eliminating native plasmids and reintroducing the recombinants carrying functional genes. Specific insertion sites in the vectors showed significant differences in the expression of fluorescence proteins. Selected integration sites were applied in the de novo synthesis of salicylic acid, leading to a titer of 142.0 ± 6.0 mg/L in a shake flask with good production stability. Additionally, the design successfully realized the biosynthesis of ergothioneine (45 mg/L) by one-step construction. This work expands the application scope of native cryptic plasmids to the easy construction of functional pathways. KEY POINTS: • Cryptic plasmids of EcN were designed to express exogenous genes • Insertion sites with different expression intensities in cryptic plasmids were provided • Target products were stably produced by engineering cryptic plasmids.
Collapse
Affiliation(s)
- Miao-Miao Dong
- State Key Laboratory of Bioreactor Engineering, Newworld Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Lu Song
- State Key Laboratory of Bioreactor Engineering, Newworld Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Jia-Qi Xu
- State Key Laboratory of Bioreactor Engineering, Newworld Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Lin Zhu
- State Key Laboratory of Bioreactor Engineering, Newworld Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Liang-Bin Xiong
- State Key Laboratory of Bioreactor Engineering, Newworld Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China.
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| | - Dong-Zhi Wei
- State Key Laboratory of Bioreactor Engineering, Newworld Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Feng-Qing Wang
- State Key Laboratory of Bioreactor Engineering, Newworld Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
10
|
Fang X, Kang L, Qiu YF, Li ZS, Bai Y. Yersinia enterocolitica in Crohn’s disease. Front Cell Infect Microbiol 2023; 13:1129996. [PMID: 36968108 PMCID: PMC10031030 DOI: 10.3389/fcimb.2023.1129996] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
Increasing attention is being paid to the unique roles gut microbes play in both physiological and pathological processes. Crohn’s disease (CD) is a chronic, relapsing, inflammatory disease of the gastrointestinal tract with unknown etiology. Currently, gastrointestinal infection has been proposed as one initiating factor of CD. Yersinia enterocolitica, a zoonotic pathogen that exists widely in nature, is one of the most common bacteria causing acute infectious gastroenteritis, which displays clinical manifestations similar to CD. However, the specific role of Y. enterocolitica in CD is controversial. In this Review, we discuss the current knowledge on how Y. enterocolitica and derived microbial compounds may link to the pathogenesis of CD. We highlight examples of Y. enterocolitica-targeted interventions in the diagnosis and treatment of CD, and provide perspectives for future basic and translational investigations on this topic.
Collapse
Affiliation(s)
| | | | | | | | - Yu Bai
- *Correspondence: Zhao-Shen Li, ; Yu Bai,
| |
Collapse
|
11
|
Tumor Colonization and Therapy by Escherichia coli Nissle 1917 Strain in Syngeneic Tumor-Bearing Mice Is Strongly Affected by the Gut Microbiome. Cancers (Basel) 2022; 14:cancers14246033. [PMID: 36551519 PMCID: PMC9776137 DOI: 10.3390/cancers14246033] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
In the past, different bacterial species have been tested for cancer therapy in preclinical and clinical studies. The success of bacterial cancer therapy is mainly dependent on the ability of the utilized bacteria to overcome the host immune defense system to colonize the tumors and to initiate tumor-specific immunity. In recent years, several groups have demonstrated that the gut microbiome plays an important role of modulation of the host immune response and has an impact on therapeutic responses in murine models and in cohorts of human cancer patients. Here we analyzed the impact of the gut microbiome on tumor colonization and tumor therapy by the Escherichia coli Nissle 1917 (EcN) strain. This EcN strain is a promising cancer therapy candidate with probiotic properties. In our study, we observed significantly better tumor colonization by EcN after antibiotic-induced temporal depletion of the gut microbiome and after two intranasal applications of the EcN derivate (EcN/pMUT-gfp Knr) in 4T1 tumor-bearing syngeneic BALB/c mice. In addition, we demonstrated significant reduction in tumor growth and extended survival of the EcN-treated mice in contrast to phosphate-buffered saline (PBS)-treated tumor-bearing control animals. Multispectral imaging of immune cells revealed that depletion of the gut microbiome led to significantly lower infiltration of cytotoxic and helper T cells (CD4 and CD8 cells) in PBS tumors of mice pretreated with antibiotics in comparison with antibiotic untreated PBS-or EcN treated mice. These findings may help in the future advancement of cancer treatment strategies using E. coli Nissle 1917.
Collapse
|
12
|
Deletion of the cheZ gene results in the loss of swimming ability and the decrease of adhesion ability to Caco-2 cells in Escherichia coli Nissle 1917. Folia Microbiol (Praha) 2022; 68:395-402. [DOI: 10.1007/s12223-022-01019-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
|
13
|
The Growth Medium Affects the Viability of IPEC-J2 Animal Cell Line in the Presence of Probiotic Bacteria. Appl Microbiol 2022. [DOI: 10.3390/applmicrobiol2040058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: The IPEC-J2 cell line is frequently used as an in vitro model to study the bioactivity of live probiotics. However, lactic acid bacteria (LB) acidify the medium, and the impact of pH and lactic acid accumulation on cell viability seem to be underestimated. Methods: IPEC-J2 viability was assessed by neutral red and flow cytometry in the presence of eight probiotics at concentrations between 106 and 109 bacteria/mL in maintenance and buffered media. Results: It was shown that a high inoculation level led to higher cytotoxic effects on IPEC-J2 cells after 22 h of incubation and that viability losses were more related to a combination of low pH and lactic acid than to the probiotics themselves. Furthermore, with LB at 106 and 107 bacteria/mL, the addition of phosphates to the media significantly reduced the drop in the pH and preserved the IPEC-J2 viability between 100% and 69%, compared to a highly variable viability between 100% and 17.5% in the unbuffered media. Conclusions: Under certain in vitro conditions, probiotics can lead to the deterioration of animal cells, and pH neutralization is an essential parameter in the cell–probiotic system in order to preserve cell viability and to better evaluate the bioactive properties of live probiotics.
Collapse
|
14
|
Cheng SY, Lin TH, Chen PT. Integration of Multiple Phage Attachment Sites System to Create the Chromosomal T7 System for Protein Production in Escherichia coli Nissle 1917. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10239-10247. [PMID: 35960546 DOI: 10.1021/acs.jafc.2c04614] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Escherichia coli Nissle 1917 (EcN) is a probiotic used to treat gastrointestinal diseases. The probiotic and endotoxin-free characteristics of EcN support its potential to be developed into a microbial expression system. With this aim, in this study, the powerful T7 expression system was constructed in the cryptic plasmid-free EcN (EcNP) to generate the T7 expression host ENL6P. The concept of multiple copies of gene expression cassettes regulated by the chromosomal T7 promoter was promoted due to plasmid instability issues with protein production in ENL6P. The integration of multiple phage attachment sites (IMPACT) system, which combined Cre-lox72, CRIM, and lambda red recombinase systems, was designed to simplify the manipulation and achieve the multiple φ80 bacterial attachment sites (attB) in ENL6P to generate the new strain ENL6PP4 with four φ80 attB sites. The strain can simultaneously integrate four copies of gene expression cassettes in the chromosome to produce recombinant proteins. The IMPACT systems incorporated several tools in gene editing to rapidly achieve more robust and stable microbial strains for research and various industrial applications.
Collapse
Affiliation(s)
- Shu-Yun Cheng
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan 710, Taiwan
| | - Tzu-Han Lin
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan 710, Taiwan
| | - Po-Ting Chen
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan 710, Taiwan
| |
Collapse
|
15
|
Wong-Chew RM, de Castro JAA, Morelli L, Perez M, Ozen M. Gut immune homeostasis: the immunomodulatory role of Bacillus clausii, from basic to clinical evidence. Expert Rev Clin Immunol 2022; 18:717-729. [PMID: 35674642 DOI: 10.1080/1744666x.2022.2085559] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The gut microbiota affects the development of the gut immune system in early life. Perturbations to microbiota structure and composition during this period can have long-term consequences on the health of the individual, through its effects on the immune system. Research in the last few decades has shown that probiotic administration can reverse these effects in strain- and environment-specific ways. Bacillus clausii (B. clausii) has been in use for many decades as a safe and efficacious probiotic, but its mode of action has not yet been completely elucidated. AREAS COVERED In this review, we discuss how the gut immune system works, the factors that affect its functioning, and the plethora of research highlighting its role in various diseases. We also discuss the known modes of action of Bacillus probiotics, and highlight the preclinical and clinical evidence that reveal how B. clausii acts to bolster gut defense. EXPERT OPINION We anticipate that the treatment and/or prevention of dysbiosis will be central to managing human health and disease in the future. Discovering the pathophysiology of autoimmune diseases, infections, allergies, and some cancers will aid our understanding of the key role played by microbial communities in these diseases.
Collapse
Affiliation(s)
- Rosa María Wong-Chew
- Facultad de Medicina, División de Investigación, Universidad Nacional Autónoma de México, Coyoacán, Cdmx
| | - Jo-Anne A de Castro
- Department of Pediatrics de la Salle Medical and Health Sciences Institute (DLSMHSI), Dasmariñas Cavite, Philippines; Department of Microbiology and Parasitology, Pamantasan ng Lunsod ng Maynila (PLM), College of Medicine Intramuros, Manila, Philippines
| | - Lorenzo Morelli
- Faculty of Agriculture, Food and Environmental Sciences, Università Cattolica del Sacro Cuore Piacenza - Cremona, Italy
| | | | - Metehan Ozen
- Division of Pediatric Infectious Diseases, Acıbadem Mehmet Ali Aydınlar University, School of Medicine, Istanbul Turkey
| |
Collapse
|
16
|
Foster-Nyarko E, Pallen MJ. The microbial ecology of Escherichia coli in the vertebrate gut. FEMS Microbiol Rev 2022; 46:fuac008. [PMID: 35134909 PMCID: PMC9075585 DOI: 10.1093/femsre/fuac008] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Escherichia coli has a rich history as biology's 'rock star', driving advances across many fields. In the wild, E. coli resides innocuously in the gut of humans and animals but is also a versatile pathogen commonly associated with intestinal and extraintestinal infections and antimicrobial resistance-including large foodborne outbreaks such as the one that swept across Europe in 2011, killing 54 individuals and causing approximately 4000 infections and 900 cases of haemolytic uraemic syndrome. Given that most E. coli are harmless gut colonizers, an important ecological question plaguing microbiologists is what makes E. coli an occasionally devastating pathogen? To address this question requires an enhanced understanding of the ecology of the organism as a commensal. Here, we review how our knowledge of the ecology and within-host diversity of this organism in the vertebrate gut has progressed in the 137 years since E. coli was first described. We also review current approaches to the study of within-host bacterial diversity. In closing, we discuss some of the outstanding questions yet to be addressed and prospects for future research.
Collapse
Affiliation(s)
- Ebenezer Foster-Nyarko
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Mark J Pallen
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, United Kingdom
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TU, United Kingdom
| |
Collapse
|
17
|
Moutinho TJ, Neubert BC, Jenior ML, Papin JA. Quantifying cumulative phenotypic and genomic evidence for procedural generation of metabolic network reconstructions. PLoS Comput Biol 2022; 18:e1009341. [PMID: 35130271 PMCID: PMC8853471 DOI: 10.1371/journal.pcbi.1009341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 02/17/2022] [Accepted: 01/19/2022] [Indexed: 01/26/2023] Open
Abstract
Genome-scale metabolic network reconstructions (GENREs) are valuable tools for understanding microbial metabolism. The process of automatically generating GENREs includes identifying metabolic reactions supported by sufficient genomic evidence to generate a draft metabolic network. The draft GENRE is then gapfilled with additional reactions in order to recapitulate specific growth phenotypes as indicated with associated experimental data. Previous methods have implemented absolute mapping thresholds for the reactions automatically included in draft GENREs; however, there is growing evidence that integrating annotation evidence in a continuous form can improve model accuracy. There is a need for flexibility in the structure of GENREs to better account for uncertainty in biological data, unknown regulatory mechanisms, and context-specificity associated with data inputs. To address this issue, we present a novel method that provides a framework for quantifying combined genomic, biochemical, and phenotypic evidence for each biochemical reaction during automated GENRE construction. Our method, Constraint-based Analysis Yielding reaction Usage across metabolic Networks (CANYUNs), generates accurate GENREs with a quantitative metric for the cumulative evidence for each reaction included in the network. The structuring of CANYUNs allows for the simultaneous integration of three data inputs while maintaining all supporting evidence for biochemical reactions that may be active in an organism. CANYUNs is designed to maximize the utility of experimental and annotation datasets and to ultimately assist in the curation of the reference datasets used for the automatic construction of metabolic networks. We validated CANYUNs by generating an E. coli K-12 model and compared it to the manually curated reconstruction iML1515. Finally, we demonstrated the use of CANYUNs to build a model by generating an E. coli Nissle CANYUNs model using novel phenotypic data that we collected. This method may address key challenges for the procedural construction of metabolic networks by leveraging uncertainty and redundancy in biological data.
Collapse
Affiliation(s)
- Thomas J. Moutinho
- University of Virginia, Department of Biomedical Engineering, Charlottesville, Virginia, United States of America
| | - Benjamin C. Neubert
- University of Virginia, Department of Biomedical Engineering, Charlottesville, Virginia, United States of America
| | - Matthew L. Jenior
- University of Virginia, Department of Biomedical Engineering, Charlottesville, Virginia, United States of America
| | - Jason A. Papin
- University of Virginia, Department of Biomedical Engineering, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
18
|
Bacteria and bacterial derivatives as delivery carriers for immunotherapy. Adv Drug Deliv Rev 2022; 181:114085. [PMID: 34933064 DOI: 10.1016/j.addr.2021.114085] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023]
Abstract
There is growing interest in the role of microorganisms in human health and disease, with evidence showing that new types of biotherapy using engineered bacterial therapeutics, including bacterial derivatives, can address specific mechanisms of disease. The complex interactions between microorganisms and metabolic/immunologic pathways underlie many diseases with unmet medical needs, suggesting that targeting these interactions may improve patient treatment. Using tools from synthetic biology and chemical engineering, non-pathogenic bacteria or bacterial products can be programmed and designed to sense and respond to environmental signals to deliver therapeutic effectors. This review describes current progress in biotherapy using live bacteria and their derivatives to achieve therapeutic benefits against various diseases.
Collapse
|
19
|
Helmy YA, Kassem II, Rajashekara G. Immuno-modulatory effect of probiotic E. coli Nissle 1917 in polarized human colonic cells against Campylobacter jejuni infection. Gut Microbes 2022; 13:1-16. [PMID: 33382951 PMCID: PMC7781529 DOI: 10.1080/19490976.2020.1857514] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Campylobacter jejuni is among the leading causes of bacterial foodborne illness. Poultry is the major reservoir and source of human campylobacteriosis. Currently, there is no effective and practical method to decrease C. jejuni colonization in chickens or to reduce human infections. Additionally, antibiotic-resistant infections pose a serious public health concern; therefore, antibiotic-alternative approaches are needed to reduce transmission of C. jejuni including resistant bacteria from chickens to humans. Here, we evaluated the effect of E. coli Nissle 1917 (EcN) on innate responses of polarized HT-29 cells and consequently on C. jejuni 81176 infections in HT-29 cells. Pre-treatment of HT-29 cells with EcN for 4 h had a significant effect on the invasion of different C. jejuni strains (2 h post-infection) (P < .05) and no intracellular C. jejuni (24 h post-infection) were recovered. To further understand how EcN mediates its impact on C. jejuni's survival inside the cells, we used Human Antibacterial RT2 ProfilerTM PCR arrays to profile gene expression in HT-29 cells after treatment with EcN with or without C. jejuni 81-176 infection. Our results suggest that pre-treatment of the HT-29 cells with EcN induced the anti-inflammatory cytokines and activated the anti-apoptotic Akt signaling which likely to protect the cells against the proinflammatory and apoptosis responses induced by C. jejuni. EcN also positively affected the expression of genes involved in cellular maintenance, growth, development, and proliferation. Further, EcN modulated the expression of genes involved in protective innate immunity, such as TLRs, ERK1/2, p38 MAPK, Ap1, JNK, IL1B, IL17A, and NF-κB signaling.
Collapse
Affiliation(s)
- Yosra A. Helmy
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, the Ohio State University, Wooster, Ohio, USA
| | - Issmat I. Kassem
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, the Ohio State University, Wooster, Ohio, USA,College of Agricultural and Environmental Sciences, Center for Food Safety, University of Georgia, Griffin, Georgia, USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, the Ohio State University, Wooster, Ohio, USA,Corresponding author Gireesh Rajashekara Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH44691, USA
| |
Collapse
|
20
|
Ghani R, Mullish BH, Roberts LA, Davies FJ, Marchesi JR. The potential utility of fecal (or intestinal) microbiota transplantation in controlling infectious diseases. Gut Microbes 2022; 14:2038856. [PMID: 35230889 PMCID: PMC8890388 DOI: 10.1080/19490976.2022.2038856] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
The intestinal microbiota is recognized to play a role in the defense against infection, but conversely also acts as a reservoir for potentially pathogenic organisms. Disruption to the microbiome can increase the risk of invasive infection from these organisms; therefore, strategies to restore the composition of the gut microbiota are a potential strategy of key interest to mitigate this risk. Fecal (or Intestinal) Microbiota Transplantation (FMT/IMT), is the administration of minimally manipulated screened healthy donor stool to an affected recipient, and remains the major 'whole microbiome' therapeutic approach at present. Driven by the marked success of using FMT in the treatment of recurrent Clostridioides difficile infection, the potential use of FMT in treating other infectious diseases is an area of active research. In this review, we discuss key examples of this treatment based on recent findings relating to the interplay between microbiota and infection, and potential further exploitations of FMT/IMT.
Collapse
Affiliation(s)
- Rohma Ghani
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Benjamin H. Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Lauren A. Roberts
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Frances J. Davies
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Julian R. Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
21
|
Seco EM, Fernández LÁ. Efficient markerless integration of genes in the chromosome of probiotic E. coli Nissle 1917 by bacterial conjugation. Microb Biotechnol 2021; 15:1374-1391. [PMID: 34755474 PMCID: PMC9049610 DOI: 10.1111/1751-7915.13967] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/22/2021] [Accepted: 10/23/2021] [Indexed: 01/30/2023] Open
Abstract
The probiotic strain Escherichia coli Nissle 1917 (EcN) is a common bacterial chassis in synthetic biology developments for therapeutic applications given its long track record of safe administration in humans. Chromosomal integration of the genes of interest (GOIs) in the engineered bacterium offers significant advantages in genetic stability and to control gene dose, but common methodologies relying on the transformation of EcN are inefficient. In this work, we implement in EcN the use of bacterial conjugation in combination with markerless genome engineering to efficiently insert multiple GOIs at different loci of EcN chromosome, leaving no antibiotic resistance genes, vector sequences or scars in the modified bacterium. The resolution of cointegrants that leads to markerless insertion of the GOIs requires expression of I-SceI endonuclease and its efficiency is enhanced by λ Red proteins. We show the potential of this strategy by integrating different genes encoding fluorescent and bioluminescent reporters (i.e. GFP, mKate2, luxCDABE) both individually and sequentially. We also demonstrate its application for gene deletions in EcN (ΔflhDC) and to replace the endogenous regulation of chromosomal locus (i.e. flhDC) by heterologous regulatory elements (e.g. tetR-Ptet) in order to have an ectopic control of gene expression in EcN with an external inducer to alter bacterial behaviour (e.g. flagellar motility). Whole-genome sequencing confirmed the introduction of the designed modifications without off-target alterations in the genome. This straightforward approach accelerates the generation of multiple modifications in EcN chromosome for the generation of living bacterial therapeutics.
Collapse
Affiliation(s)
- Elena M Seco
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Campus UAM Cantoblanco, Madrid, 28049, Spain
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Campus UAM Cantoblanco, Madrid, 28049, Spain
| |
Collapse
|
22
|
Jones CV, Jarboe BG, Majer HM, Ma AT, Beld J. Escherichia coli Nissle 1917 secondary metabolism: aryl polyene biosynthesis and phosphopantetheinyl transferase crosstalk. Appl Microbiol Biotechnol 2021; 105:7785-7799. [PMID: 34546406 DOI: 10.1007/s00253-021-11546-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 10/20/2022]
Abstract
Escherichia coli Nissle 1917 (EcN) is a Gram-negative bacterium that is used to treat inflammatory bowel diseases. The probiotic character of EcN is not well-understood, but its ability to produce secondary metabolites plays an important role in its activity. The EcN genome encodes for an aryl polyene (APE) biosynthetic gene cluster (BGC), and APE products have a role in biofilm formation. We show here that this unusual polyketide assembly line synthase produces four APE molecules which are likely cis/trans isomers. Within the APE BGC, two acyl carrier proteins are involved in biosynthesis. Acyl carrier proteins require activation by post-translational modification with a phosphopantetheinyl transferase (PPTase). Through analysis of single, double, and triple mutants of three PPTases, the PPTase-BGC crosstalk relationship in EcN was characterized. Understanding PPTase-BGC crosstalk is important for the engineering of secondary metabolite production hosts and for targeting of PPTases with new antibiotics. KEY POINTS: • Escherichia coli Nissle 1917 biosynthesizes four aryl polyene isoforms. • Phosphopantetheinyl transferase crosstalk is important for biosynthesis.
Collapse
Affiliation(s)
- Courtney V Jones
- Department of Microbiology and Immunology, Center for Advanced Microbial Processing and Center for Genomics Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N 15th St, Philadelphia, PA, 19102, USA
| | - Brianna G Jarboe
- Department of Microbiology and Immunology, Center for Advanced Microbial Processing and Center for Genomics Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N 15th St, Philadelphia, PA, 19102, USA
| | - Haley M Majer
- Department of Microbiology and Immunology, Center for Advanced Microbial Processing and Center for Genomics Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N 15th St, Philadelphia, PA, 19102, USA
| | - Amy T Ma
- Department of Microbiology and Immunology, Center for Advanced Microbial Processing and Center for Genomics Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N 15th St, Philadelphia, PA, 19102, USA
| | - Joris Beld
- Department of Microbiology and Immunology, Center for Advanced Microbial Processing and Center for Genomics Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N 15th St, Philadelphia, PA, 19102, USA.
| |
Collapse
|
23
|
Jayashree S, Sivakumar R, Karthikeyan R, Gunasekaran P, Rajendhran J. Genome-wide identification of probiotic Escherichia coli Nissle 1917 (EcN) fitness genes during adhesion to the intestinal epithelial cells Caco-2. Gene 2021; 803:145890. [PMID: 34375634 DOI: 10.1016/j.gene.2021.145890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/22/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022]
Abstract
Escherichia coli Nissle 1917 (EcN) is an efficient probiotic strain extensively used worldwide because of its several health benefits. Adhesion to the intestinal cells is one of the prerequisites for a probiotic strain. To identify the genes essential for the adhesion of EcN on the intestinal cells, we utilized a quantitative genetic footprinting approach called transposon insertion sequencing (INSeq). A transposon insertion mutant library of EcN comprising of ~17,000 mutants was used to screen the adherence to the intestinal epithelial cells, Caco-2. The transposon insertion sites were identified from the input and output population by employing next-generation sequencing using the Ion torrent platform. Based on the relative abundance of reads in the input and output pools, we identified 113 candidate genes that are essential for the fitness of EcN during the adhesion and colonization on the Caco-2 cells. Functional categorization revealed that these fitness genes are associated with carbohydrate transport and metabolism, cell wall/membrane/envelope biogenesis, post-translational modification, stress response, motility and adhesion, and signal transduction. To further validate the genes identified in our INSeq analysis, we constructed individual knock-out mutants in five genes (cyclic di-GMP phosphodiesterase (gmp), hda, uidC, leuO, and hypothetical protein-coding gene). We investigated their ability to adhere to Caco-2 cells. Evaluation of these mutants showed reduced adhesion on Caco-2 cells, confirming their role in adhesion. Understanding the functions of these genes may provide novel insights into molecular regulation during colonization of probiotic bacteria to the intestinal cells, and useful to develop designer probiotic strains.
Collapse
Affiliation(s)
| | - Ramamoorthy Sivakumar
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Raman Karthikeyan
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | | | - Jeyaprakash Rajendhran
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, India.
| |
Collapse
|
24
|
Serra DO, Hengge R. Bacterial Multicellularity: The Biology of Escherichia coli Building Large-Scale Biofilm Communities. Annu Rev Microbiol 2021; 75:269-290. [PMID: 34343018 DOI: 10.1146/annurev-micro-031921-055801] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Biofilms are a widespread multicellular form of bacterial life. The spatial structure and emergent properties of these communities depend on a polymeric extracellular matrix architecture that is orders of magnitude larger than the cells that build it. Using as a model the wrinkly macrocolony biofilms of Escherichia coli, which contain amyloid curli fibers and phosphoethanolamine (pEtN)-modified cellulose as matrix components, we summarize here the structure, building, and function of this large-scale matrix architecture. Based on different sigma and other transcription factors as well as second messengers, the underlying regulatory network reflects the fundamental trade-off between growth and survival. It controls matrix production spatially in response to long-range chemical gradients, but it also generates distinct patterns of short-range matrix heterogeneity that are crucial for tissue-like elasticity and macroscopic morphogenesis. Overall, these biofilms confer protection and a potential for homeostasis, thereby reducing maintenance energy, which makes multicellularity an emergent property of life itself. Expected final online publication date for the Annual Review of Microbiology, Volume 75 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Diego O Serra
- Instituto de Biología Molecular y Celular de Rosario (IBR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario (UNR), 2000 Rosario, Argentina
| | - Regine Hengge
- Institut für Biologie/Mikrobiologie, Humboldt-Universität zu Berlin, 10115 Berlin, Germany;
| |
Collapse
|
25
|
Damodharan K, Palaniyandi SA, Suh JW, Yang SH. Probiotic Characterization of Lactobacillus paracasei subsp. paracasei KNI9 Inhibiting Adherence of Yersinia enterocolitica on Caco-2 Cells In Vitro. Probiotics Antimicrob Proteins 2021; 12:600-607. [PMID: 31289994 DOI: 10.1007/s12602-019-09535-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The study was aimed to characterize the probiotic properties of Lactobacillus paracasei subsp. paracasei strain KNI9 and its antagonistic activity against Yersinia enterocolitica subsp. enterocolitica. The strain KNI9 was susceptible to antibiotics such as chloramphenicol, tetracycline, erythromycin, and streptomycin recommended by European food safety authority (EFSA). Strain KNI9 exhibited tolerance to simulated oro-gastrointestinal (OGT) condition, adherence to Caco-2 cells, and antimicrobial activity against intestinal enteric pathogens such as Yersinia enterocolitica subsp. enterocolitica, Shigella boydii, and Listeria monocytogenes. Furthermore, the strain KNI9 inhibited the adherence and invasiveness of Y. enterocolitica subsp. enterocolitica to Caco-2 cell line. These results indicate that the L. paracasei subsp. paracasei KNI9 could be further developed into a potential probiotic strain after appropriate in vivo studies.
Collapse
Affiliation(s)
- Karthiyaini Damodharan
- Center for Nutraceutical and Pharmaceutical Materials, College of Natural Science, Myongji University, Cheoin-gu, Yongin, Gyeonggi-Do, 17058, Republic of Korea
| | - Sasikumar Arunachalam Palaniyandi
- Department of Biotechnology, Mepco Schlenk Engineering College, Mepco Nagar, Mepco Engineering College Post, Sivakasi, Tamilnadu, 626005, India
| | - Joo-Won Suh
- Center for Nutraceutical and Pharmaceutical Materials, College of Natural Science, Myongji University, Cheoin-gu, Yongin, Gyeonggi-Do, 17058, Republic of Korea.
| | - Seung Hwan Yang
- Department of Biotechnology, Chonnam National University, Yeosu, Chonnam, 59626, Republic of Korea.
| |
Collapse
|
26
|
Chiang CJ, Chao YP, Ali A, Day CH, Ho TJ, Wang PN, Lin SC, Padma VV, Kuo WW, Huang CY. Probiotic Escherichia coli Nissle inhibits IL-6 and MAPK-mediated cardiac hypertrophy during STZ-induced diabetes in rats. Benef Microbes 2021; 12:283-293. [PMID: 34030609 DOI: 10.3920/bm2020.0094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Escherichia coli Nissle (EcN), a probiotic bacterium protects against several disorders. Multiple reports have studied the pathways involved in cardiac hypertrophy. However, the effects of probiotic EcN against diabetes-induced cardiac hypertrophy remain to be understood. We administered five weeks old Wistar male (271±19.4 g body weight) streptozotocin-induced diabetic rats with 109 cfu of EcN via oral gavage every day for 24 days followed by subjecting the rats to echocardiography to analyse the cardiac parameters. Overexpressed interleukin (IL)-6 induced the MEK5/ERK5, JAK2/STAT3, and MAPK signalling cascades in streptozotocin-induced diabetic rats. Further, the upregulation of calcineurin, NFATc3, and p-GATA4 led to the elevation of hypertrophy markers, such as atrial and B-type natriuretic peptides. In contrast, diabetic rats supplemented with probiotic EcN exhibited significant downregulated IL-6. Moreover, the MEK5/ERK5 and JAK2/STAT3 cascades involved during eccentric hypertrophy and MAPK signalling, including phosphorylated MEK, ERK, JNK, and p-38, were significantly attenuated in diabetic rats after supplementation of EcN. Western blotting and immunofluorescence revealed the significant downregulation of NFATc3 and downstream mediators, thereby resulting in the impairment of cardiac hypertrophy. Taken together, the findings demonstrate that supplementing probiotic EcN has the potential to show cardioprotective effects by inhibiting diabetes-induced cardiomyopathies.
Collapse
Affiliation(s)
- C J Chiang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, 91 Hsueh-Shih Rd., Taichung 40402, Taiwan
| | - Y P Chao
- Department of Chemical Engineering, Feng Chia University, No. 100 Wenhwa Rd., Seatwen, Taichung 40724, Taiwan
| | - A Ali
- Department of Biological Science and Technology, China Medical University, 91 Hsueh-Shih Rd., Taichung 40402, Taiwan
| | - C H Day
- Department of Nursing, MeiHo University, 23, Pingguang Rd., Neipu, Pingtung 912, Taiwan
| | - T J Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, 707 Section 3 Chung-Yang Road, Hualien 97002, Taiwan.,Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan.,School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, 701 Jhongyang Road Section 3, Hualien 97004, Taiwan
| | - P N Wang
- Department of Chemical Engineering, Feng Chia University, No. 100 Wenhwa Rd., Seatwen, Taichung 40724, Taiwan
| | - S C Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, 91 Hsueh-Shih Rd., Taichung 40402, Taiwan
| | - V V Padma
- Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - W W Kuo
- Department of Biological Science and Technology, China Medical University, 91 Hsueh-Shih Rd., Taichung 40402, Taiwan.,Ph.D. Program for Biotechnology Industry, China Medical University, Taichung 406, Taiwan
| | - C Y Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, 91 Hsueh-Shih Rd., Taichung 40402, Taiwan.,Department of Biotechnology, Asia University, 500 Liufeng Rd., Wufeng, 41354 Taichung, Taiwan.,Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan
| |
Collapse
|
27
|
Lan YJ, Tan SI, Cheng SY, Ting WW, Xue C, Lin TH, Cai MZ, Chen PT, Ng IS. Development of Escherichia coli Nissle 1917 derivative by CRISPR/Cas9 and application for gamma-aminobutyric acid (GABA) production in antibiotic-free system. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2021.107952] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
28
|
Kan A, Gelfat I, Emani S, Praveschotinunt P, Joshi NS. Plasmid Vectors for in Vivo Selection-Free Use with the Probiotic E. coli Nissle 1917. ACS Synth Biol 2021; 10:94-106. [PMID: 33301298 PMCID: PMC7813132 DOI: 10.1021/acssynbio.0c00466] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Escherichia
coli Nissle 1917 (EcN) is a probiotic
bacterium, commonly employed to treat certain gastrointestinal disorders.
It is fast emerging as an important target for the development of
therapeutic engineered bacteria, benefiting from the wealth of knowledge
of E. coli biology and ease of manipulation.
Bacterial synthetic biology projects commonly utilize engineered plasmid
vectors, which are simple to engineer and can reliably achieve high
levels of protein expression. However, plasmids typically require
antibiotics for maintenance, and the administration of an antibiotic
is often incompatible with in vivo experimentation
or treatment. EcN natively contains plasmids pMUT1 and pMUT2, which
have no known function but are stable within the bacteria. Here, we
describe the development of the pMUT plasmids into a robust platform
for engineering EcN for in vivo experimentation,
alongside a CRISPR-Cas9 system to remove the native plasmids. We systematically
engineered both pMUT plasmids to contain selection markers, fluorescent
markers, temperature sensitive expression, and curli secretion systems
to export a customizable functional material into the extracellular
space. We then demonstrate that the engineered plasmids were maintained
in bacteria as the engineered bacteria pass through the mouse GI tract
without selection, and that the secretion system remains functional,
exporting functionalized curli proteins into the gut. Our plasmid
system presents a platform for the rapid development of therapeutic
EcN bacteria.
Collapse
Affiliation(s)
- Anton Kan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - Ilia Gelfat
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Sivaram Emani
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
- Harvard College, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Pichet Praveschotinunt
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Neel S. Joshi
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
29
|
Kelly VW, Liang BK, Sirk SJ. Living Therapeutics: The Next Frontier of Precision Medicine. ACS Synth Biol 2020; 9:3184-3201. [PMID: 33205966 DOI: 10.1021/acssynbio.0c00444] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Modern medicine has long studied the mechanism and impact of pathogenic microbes on human hosts, but has only recently shifted attention toward the complex and vital roles that commensal and probiotic microbes play in both health and dysbiosis. Fueled by an enhanced appreciation of the human-microbe holobiont, the past decade has yielded countless insights and established many new avenues of investigation in this area. In this review, we discuss advances, limitations, and emerging frontiers for microbes as agents of health maintenance, disease prevention, and cure. We highlight the flexibility of microbial therapeutics across disease states, with special consideration for the rational engineering of microbes toward precision medicine outcomes. As the field advances, we anticipate that tools of synthetic biology will be increasingly employed to engineer functional living therapeutics with the potential to address longstanding limitations of traditional drugs.
Collapse
Affiliation(s)
- Vince W. Kelly
- Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Benjamin K. Liang
- Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Shannon J. Sirk
- Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
30
|
Zhao Y, Wang C, Goel A. Role of gut microbiota in epigenetic regulation of colorectal Cancer. Biochim Biophys Acta Rev Cancer 2020; 1875:188490. [PMID: 33321173 DOI: 10.1016/j.bbcan.2020.188490] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) remains one of the most commonly diagnosed cancers and a leading cause of cancer-related deaths worldwide. The stepwise accumulation of epigenetic alterations in the normal colorectal epithelium has been reported to act as a driving force for the initiation and promotion of tumorigenesis in CRC. From a mechanistic standpoint, emerging evidence indicates that within the colorectal epithelium, the diverse gut microbiota can interact with host cells to regulate multiple physiological processes. In fact, recent studies have found that the gut microbiota represents a potential cause of carcinogenesis, invasion, and metastasis via DNA methylation, histone modifications, and non-coding RNAs - providing an epigenetic perspective for the connection between the gut microbiota and CRC. Herein, we comprehensively review the recent research that provides a comprehensive yet succinct evidence connecting the gut microbiota to CRC at an epigenetic level, including carcinogenic mechanisms of cancer-related microbiota, and the potential for utilizing the gut microbiota as CRC biomarkers. These scientific findings highlight a promising future for manipulating the gut microbiota to improve clinical outcomes in patients suffering from CRC.
Collapse
Affiliation(s)
- Yinghui Zhao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China; Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
31
|
Mayorgas A, Dotti I, Salas A. Microbial Metabolites, Postbiotics, and Intestinal Epithelial Function. Mol Nutr Food Res 2020; 65:e2000188. [DOI: 10.1002/mnfr.202000188] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/31/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Aida Mayorgas
- Department of Gastroenterology, Hospital Clínic ‐ IDIBAPS C/Rosselló, 149‐153, 3rd Floor Barcelona 08036 Spain
| | - Isabella Dotti
- Department of Gastroenterology, Hospital Clínic ‐ IDIBAPS C/Rosselló, 149‐153, 3rd Floor Barcelona 08036 Spain
| | - Azucena Salas
- Department of Gastroenterology, Hospital Clínic ‐ IDIBAPS C/Rosselló, 149‐153, 3rd Floor Barcelona 08036 Spain
| |
Collapse
|
32
|
Abdelhamid AG, El-Dougdoug NK. Controlling foodborne pathogens with natural antimicrobials by biological control and antivirulence strategies. Heliyon 2020; 6:e05020. [PMID: 32995651 PMCID: PMC7511826 DOI: 10.1016/j.heliyon.2020.e05020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/02/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
Foodborne diseases represent a global health threat besides the great economic losses encountered by the food industry. These hazards necessitate the implementation of food preservation methods to control foodborne pathogens, the causal agents of human illnesses. Until now, most control methods rely on inhibiting the microbial growth or eliminating the pathogens by applying lethal treatments. Natural antimicrobials, which inhibit microbial growth, include traditional chemicals, naturally occurring antimicrobials, or biological preservation (e.g. beneficial microbes, bacteriocins, or bacteriophages). Although having great antimicrobial effectiveness, challenges due to the adaptation of foodborne pathogens to such control methods are becoming apparent. Such adaptation enables the survival of the pathogens in foods or food-contact environments. This imperative concern inspires contemporary research and food industry sector to develop technologies which do not target microbial growth but disarming microbial virulence factors. These technologies, referred to as "antivirulence", render the microbe non-capable of causing the disease with very limited or no opportunities for the pathogenic microorganisms to develop resistance. For the sake of safer and fresh-like foods, with no effect on the sensory properties of foods, a combination of two or more natural antimicrobials or with other stressors, is now widespread, to preserve foods. This review introduces and critically describes the traditional versus the emerging uses of natural antimicrobials for controlling foodborne pathogens in foods. Development of biological control strategies using natural antimicrobials proved to be effective in inhibiting microbial growth in foods and allowing improved food safety. In the meanwhile, discovery of new antivirulence agents could be a transformative strategy in food preservation in the far future.
Collapse
Affiliation(s)
- Ahmed G. Abdelhamid
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, 13511, Egypt
| | - Noha K. El-Dougdoug
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, 13511, Egypt
| |
Collapse
|
33
|
Terminal Ileitis due to Yersinia Infection: An Underdiagnosed Situation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1240626. [PMID: 32566652 PMCID: PMC7273408 DOI: 10.1155/2020/1240626] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/29/2020] [Indexed: 01/08/2023]
Abstract
Endoscopy is currently the gold standard for the diagnosis of inflammatory bowel disease (IBD). The presence of macroscopic lesions along with the microscopic detection of inflammatory infiltration in the terminal ileum often leads the gastroenterologist to the diagnosis of Crohn's disease (CD). However, some of these cases could be, in fact, an infection caused by Yersinia spp., accompanied or not with CD, which could be easily diagnosed with the identification of serum antibodies against Yersinia outer protein antigens (YOP antigens). Since Yersiniosis is considered to be an uncommon situation, food and water are not usually checked for the possibility of contamination by Yersinia. Therefore, it is reasonable to assume that the true prevalence of Yersinia infection in patients with terminal ileitis is probably underestimated. In this article, we review the most important data regarding the various aspects of Yersinia infection with special focus on its pathophysiology and diagnosis. We recommend testing for serum antibodies against YOP antigens in all patients with an endoscopic and histological image of terminal ileitis in order to identify Yersiniosis in conjunction or not with terminal ileum CD.
Collapse
|
34
|
Behrouzi A, Mazaheri H, Falsafi S, Tavassol ZH, Moshiri A, Siadat SD. Intestinal effect of the probiotic Escherichia coli strain Nissle 1917 and its OMV. J Diabetes Metab Disord 2020; 19:597-604. [PMID: 32550212 DOI: 10.1007/s40200-020-00511-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/26/2020] [Indexed: 12/15/2022]
Abstract
Several investigations have been conducted during the past years to examine the correlation between dysbiosis and both intestinal and extra-intestinal diseases such as inflammatory bowel disease (IBD) and ulcerative colitis (UC). E. coli Nissle 1917 (EcN) is a nonpathogenic gram-negative strain utilized in numerous gastrointestinal issues, consisting of diarrhea, uncomplicated diverticular malady, IBD and specifically UC. Many investigations have been done to examine the capability of assertive bacteria, inclusive of commensal and probiotic strains to enhance IBD in clinical testing. Bacterial secreted factors have been investigated to detect the EcN agents that facilitate the regulation of tight junction. These agents candiffuse smoothly through the mucin layer before reaching intestinal epithelial cells. Outer membrane vesicles (OMVs) are known as intercellular communicasomes as they facilitate the distal transfer of active compounds between cells. A few investigations have detailed immune-modulatory attributes for EcN through various systems that could be liable for its clinical viability in IBD. Today, the function of gut microbiota extracellular vesicles in health and disease has become a focus of attention as they serve as vehicles for the transmission of microorganisms to distal tissues of many bacterial effectors.
Collapse
Affiliation(s)
- Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hoora Mazaheri
- Department of Molecular Biology, Pasteur Institute of Iran , Tehran, Iran
| | - Sarvenaz Falsafi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Zahra Hoseini Tavassol
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Arfa Moshiri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.,Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Jiménez-Avalos JA, Arrevillaga-Boni G, González-López L, García-Carvajal ZY, González-Avila M. Classical methods and perspectives for manipulating the human gut microbial ecosystem. Crit Rev Food Sci Nutr 2020; 61:234-258. [PMID: 32114770 DOI: 10.1080/10408398.2020.1724075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A healthy Human Gut Microbial Ecosystem (HGME) is a necessary condition for maintaining the orderly function of the whole body. Major alterations in the normal gut microbial composition, activity and functionality (dysbiosis) by an environmental or host-related disruptive event, can compromise metabolic, inflammatory, and neurological processes, causing disorders such as obesity, inflammatory bowel disease, colorectal cancer, and depressive episodes. The restore or the maintaining of the homeostatic balance of Gut Microbiota (GM) populations (eubiosis) is possible through diet, the use of probiotics, prebiotics, antibiotics, and even Fecal Microbiota Transplantation (FMT). Although these "classic methods" represent an effective and accepted way to modulate GM, the complexity of HGME requires new approaches to control it in a more appropriate way. Among the most promising emergent strategies for modulating GM are the use of engineered nanomaterials (metallic nanoparticles (NP), polymeric-NP, quantum dots, micelles, dendrimers, and liposomes); phagotherapy (i.e., phages linked with the CRISPR/Cas9 system), and the use of antimicrobial peptides, non-antibiotic drugs, vaccines, and immunoglobulins. Here we review the current state of development, implications, advantages, disadvantages, and perspectives of the different approaches for manipulating HGME.
Collapse
Affiliation(s)
- Jorge Armando Jiménez-Avalos
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Gerardo Arrevillaga-Boni
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | | | - Zaira Yunuen García-Carvajal
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Marisela González-Avila
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| |
Collapse
|
36
|
Li Y, Rebuffat S. The manifold roles of microbial ribosomal peptide-based natural products in physiology and ecology. J Biol Chem 2020; 295:34-54. [PMID: 31784450 PMCID: PMC6952617 DOI: 10.1074/jbc.rev119.006545] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The ribosomally synthesized and posttranslationally modified peptides (RiPPs), also called ribosomal peptide natural products (RPNPs), form a growing superfamily of natural products that are produced by many different organisms and particularly by bacteria. They are derived from precursor polypeptides whose modification by various dedicated enzymes helps to establish a vast array of chemical motifs. RiPPs have attracted much interest as a source of potential therapeutic agents, and in particular as alternatives to conventional antibiotics to address the bacterial resistance crisis. However, their ecological roles in nature are poorly understood and explored. The present review describes major RiPP actors in competition within microbial communities, the main ecological and physiological functions currently evidenced for RiPPs, and the microbial ecosystems that are the sites for these functions. We envision that the study of RiPPs may lead to discoveries of new biological functions and highlight that a better knowledge of how bacterial RiPPs mediate inter-/intraspecies and interkingdom interactions will hold promise for devising alternative strategies in antibiotic development.
Collapse
Affiliation(s)
- Yanyan Li
- Laboratory Molecules of Communication and Adaptation of Microorganisms (MCAM, UMR 7245 CNRS-MNHN), National Museum of Natural History (MNHN), CNRS, CP 54, 57 rue Cuvier 75005, Paris, France.
| | - Sylvie Rebuffat
- Laboratory Molecules of Communication and Adaptation of Microorganisms (MCAM, UMR 7245 CNRS-MNHN), National Museum of Natural History (MNHN), CNRS, CP 54, 57 rue Cuvier 75005, Paris, France.
| |
Collapse
|
37
|
Wosinska L, Cotter PD, O'Sullivan O, Guinane C. The Potential Impact of Probiotics on the Gut Microbiome of Athletes. Nutrients 2019; 11:E2270. [PMID: 31546638 PMCID: PMC6835687 DOI: 10.3390/nu11102270] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/05/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022] Open
Abstract
There is accumulating evidence that physical fitness influences the gut microbiome and as a result, promotes health. Indeed, exercise-induced alterations in the gut microbiome can influence health parameters crucial to athletic performance, specifically, immune function, lower susceptibility to infection, inflammatory response and tissue repair. Consequently, maintenance of a healthy gut microbiome is essential for an athlete's health, training and performance. This review explores the effect of exercise on the microbiome while also investigating the effect of probiotics on various potential consequences associated with over-training in athletes, as well as their associated health benefits.
Collapse
Affiliation(s)
- Laura Wosinska
- Department of Biological Sciences, Cork Institute of Technology, Bishopstown, T12 P928 Cork, Ireland.
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
- APC Microbiome Ireland, T12 YT20 Cork, Ireland.
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
- APC Microbiome Ireland, T12 YT20 Cork, Ireland.
| | - Orla O'Sullivan
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
- APC Microbiome Ireland, T12 YT20 Cork, Ireland.
| | - Caitriona Guinane
- Department of Biological Sciences, Cork Institute of Technology, Bishopstown, T12 P928 Cork, Ireland.
| |
Collapse
|
38
|
Sarkar S, Hutton ML, Vagenas D, Ruter R, Schüller S, Lyras D, Schembri MA, Totsika M. Intestinal Colonization Traits of Pandemic Multidrug-Resistant Escherichia coli ST131. J Infect Dis 2019; 218:979-990. [PMID: 29471349 PMCID: PMC6093498 DOI: 10.1093/infdis/jiy031] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 02/01/2018] [Indexed: 01/09/2023] Open
Abstract
Background Epidemiological studies point to the gut as a key reservoir of multidrug resistant Escherichia coli multilocus sequence type 131 (ST131), a globally dominant pathogenic clone causing urinary tract and bloodstream infections. Here we report a detailed investigation of its intestinal lifestyle. Methods Clinical ST131 isolates and type 1 fimbriae null mutants were assessed for colonization of human intestinal epithelia and in mouse intestinal colonization models. Mouse gut tissue underwent histologic analysis for pathology and ST131 localization. Key findings were corroborated in mucus-producing human cell lines and intestinal biopsy specimens. Results ST131 strains adhered to and invaded human intestinal epithelial cells more than probiotic and commensal strains. The reference ST131 strain EC958 established persistent intestinal colonization in mice, and expression of type 1 fimbriae mediated higher colonization levels. Bacterial loads were highest in the distal parts of the mouse intestine and did not cause any obvious pathology. Further analysis revealed that EC958 could bind to both mucus and underlying human intestinal epithelia. Conclusions ST131 strains can efficiently colonize the mammalian gut and persist long term. Type 1 fimbriae enhance ST131 intestinal colonization, suggesting that mannosides, currently developed as therapeutics for bladder infections and Crohn’s disease, could also be used to limit intestinal ST131 reservoirs.
Collapse
Affiliation(s)
- Sohinee Sarkar
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Queensland, Australia
| | - Melanie L Hutton
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia
- Department of Microbiology, Monash University, Clayton, Australia
| | - Dimitrios Vagenas
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Queensland, Australia
| | - Rinaldo Ruter
- Gut Health and Food Safety Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Stephanie Schüller
- Gut Health and Food Safety Programme, Quadram Institute Bioscience, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Dena Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, Australia
- Department of Microbiology, Monash University, Clayton, Australia
| | - Mark A Schembri
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane
| | - Makrina Totsika
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Queensland, Australia
- Correspondence: M. Totsika, PhD, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, QLD 4059, Australia ()
| |
Collapse
|
39
|
Splichal I, Donovan SM, Splichalova Z, Neuzil Bunesova V, Vlkova E, Jenistova V, Killer J, Svejstil R, Skrivanova E, Splichalova A. Colonization of Germ-Free Piglets with Commensal Lactobacillus amylovorus, Lactobacillus mucosae, and Probiotic E. coli Nissle 1917 and Their Interference with Salmonella Typhimurium. Microorganisms 2019; 7:microorganisms7080273. [PMID: 31434337 PMCID: PMC6722580 DOI: 10.3390/microorganisms7080273] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 01/12/2023] Open
Abstract
Non-typhoid Salmonellae are worldwide spread food-borne pathogens that cause diarrhea in humans and animals. Their multi-drug resistances require alternative ways to combat this enteric pathogen. Mono-colonization of a gnotobiotic piglet gastrointestinal tract with commensal lactobacilli Lactobacillus amylovorus and Lactobacillus mucosae and with probiotic E. coli Nissle 1917 and their interference with S. Typhimurium infection was compared. The impact of bacteria and possible protection against infection with Salmonella were evaluated by clinical signs, bacterial translocation, intestinal histology, mRNA expression of villin, claudin-1, claudin-2, and occludin in the ileum and colon, and local intestinal and systemic levels of inflammatory cytokines IL-8, TNF-α, and IL-10. Both lactobacilli colonized the gastrointestinal tract in approximately 100× lower density compare to E. coli Nissle and S. Typhimurium. Neither L. amylovorus nor L. mucosae suppressed the inflammatory reaction caused by the 24 h infection with S. Typhimurium. In contrast, probiotic E. coli Nissle 1917 was able to suppress clinical signs, histopathological changes, the transcriptions of the proteins, and the inductions of the inflammatory cytokines. Future studies are needed to determine whether prebiotic support of the growth of lactobacilli and multistrain lactobacilli inoculum could show higher protective effects.
Collapse
Affiliation(s)
- Igor Splichal
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic
| | - Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA
| | - Zdislava Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic
| | - Vera Neuzil Bunesova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, 165 00 Prague, Czech Republic
| | - Eva Vlkova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, 165 00 Prague, Czech Republic
| | - Vera Jenistova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic
| | - Jiri Killer
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, 165 00 Prague, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Roman Svejstil
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, 165 00 Prague, Czech Republic
| | - Eva Skrivanova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, 165 00 Prague, Czech Republic
| | - Alla Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic.
| |
Collapse
|
40
|
Zainuddin HS, Bai Y, Mansell TJ. CRISPR-based curing and analysis of metabolic burden of cryptic plasmids in Escherichia coli Nissle 1917. Eng Life Sci 2019; 19:478-485. [PMID: 32625025 DOI: 10.1002/elsc.201900003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/28/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022] Open
Abstract
E. coli Nissle 1917 (EcN) has long been used as an over-the-counter probiotic and has shown potential to be used as a live biotherapeutic. It contains two stably replicating cryptic plasmids, pMUT1, and pMUT2, the function of which is unclear but the presence of which may increase the metabolic burden on the cell, particularly in the context of added recombinant plasmids. In this work, we present a clustered regularly interspaced short palindromic repeats-Cas9-based method of curing cryptic plasmids, producing strains cured of one or both plasmids. We then assayed heterologous protein production from three different recombinant plasmids in wild-type and cured EcN derivatives and found that production of reporter proteins was not significantly different across strains. In addition, we replaced pMUT2 with an engineered version containing an inserted antibiotic resistance reporter gene and demonstrated that the engineered plasmid was stable over 90 generations without selection. These findings have broad implications for the curing of cryptic plasmids and for stable heterologous expression of proteins in this host. Specifically, curing of cryptic plasmids may not be necessary for optimal heterologous expression in this host.
Collapse
Affiliation(s)
- Halimatun S Zainuddin
- Department of Chemical and Biological Engineering Iowa State University Sweeney Hall Ames IA USA
| | - Yanfen Bai
- Department of Chemical and Biological Engineering Iowa State University Sweeney Hall Ames IA USA
| | - Thomas J Mansell
- Department of Chemical and Biological Engineering Iowa State University Sweeney Hall Ames IA USA
| |
Collapse
|
41
|
Mirsepasi-Lauridsen HC, Vallance BA, Krogfelt KA, Petersen AM. Escherichia coli Pathobionts Associated with Inflammatory Bowel Disease. Clin Microbiol Rev 2019; 32:e00060-18. [PMID: 30700431 PMCID: PMC6431131 DOI: 10.1128/cmr.00060-18] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gut bacteria play a key role in initiating and maintaining the inflammatory process in the gut tissues of inflammatory bowel disease (IBD) patients, by supplying antigens or other stimulatory factors that trigger immune cell activation. Changes in the composition of the intestinal microbiota in IBD patients compared to that in healthy controls and a reduced diversity of intestinal microbial species are linked to the pathogenesis of IBD. Adherent invasive Escherichia coli (AIEC) has been linked to Crohn's disease (CD) patients, while diffusely adherent E. coli (DAEC) has been associated with ulcerative colitis (UC). Bacteriological analysis of intestinal biopsy specimens and fecal samples from IBD patients shows an increased number of E. coli strains belonging to the B2 phylogenetic group, which are typically known as extraintestinal pathogenic E. coli (ExPEC). Results from studies of both cell cultures and animal models reveal pathogenic features of these E. coli pathobionts, which may link them to IBD pathogenesis. This suggests that IBD-associated E. coli strains play a facilitative role during IBD flares. In this review, we explain IBD-associated E. coli and its role in IBD pathogenesis.
Collapse
Affiliation(s)
| | - Bruce Andrew Vallance
- Division of Gastroenterology, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen Angeliki Krogfelt
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
- Department of Viral and Microbiological Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | - Andreas Munk Petersen
- Department of Gastroenterology, Hvidovre University Hospital, Copenhagen, Denmark
- Department of Clinical Microbiology, Hvidovre University Hospital, Copenhagen, Denmark
| |
Collapse
|
42
|
Dawley C, Gibson KE. Virus–Bacteria Interactions: Implications for Prevention and Control of Human Enteric Viruses from Environment to Host. Foodborne Pathog Dis 2019; 16:81-89. [DOI: 10.1089/fpd.2018.2543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Cailin Dawley
- Department of Food Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas
| | - Kristen E. Gibson
- Department of Food Science, Division of Agriculture, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
43
|
Geldart KG, Kommineni S, Forbes M, Hayward M, Dunny GM, Salzman NH, Kaznessis YN. Engineered E. coli Nissle 1917 for the reduction of vancomycin-resistant Enterococcus in the intestinal tract. Bioeng Transl Med 2018; 3:197-208. [PMID: 30377660 PMCID: PMC6195901 DOI: 10.1002/btm2.10107] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/22/2018] [Accepted: 07/09/2018] [Indexed: 12/22/2022] Open
Abstract
Vancomycin-resistant Enterococcus (VRE) poses a serious threat in hospitals where they densely colonize the intestinal tracts of patients. In vulnerable hosts, these pathogens may translocate to the bloodstream and become lethal. The ability to selectively reduce VRE in the intestinal tracts of patients could potentially prevent many of these translocation events and reduce the spread of the pathogen. Herein, we have engineered Escherichia. coli Nissle 1917 to produce and secrete three antimicrobial peptides, Enterocin A, Enterocin B, and Hiracin JM79, to specifically target and kill Enterococcus. These peptides exhibited potent activity against both Enterococcus faecium and Enterococcus faecalis, the two most prominent species responsible for VRE infections. We first discuss the optimization of the system used to express and secrete the peptides. We then show that by simultaneously expressing these peptides, both E. faecium and E. faecalis were drastically inhibited. We then demonstrate a suppression of the development of resistance when supernatant from the E. coli producer strains was used to treat E. faecium. Finally, we tested the efficacy of the probiotic in a VRE colonization model in mice. These studies showed that administration of the engineered probiotic significantly reduced the levels of both E. faecium and E. faecalis in the feces of male Balb/cJ mice.
Collapse
Affiliation(s)
- Kathryn G. Geldart
- General Probiotics Inc.St. PaulMN 55114
- Department of Microbiology and ImmunologyUniversity of MinnesotaMinneapolisMN 55455
| | | | | | - Michael Hayward
- Department of PediatricsMedical College of WisconsinMilwaukeeWI 53226
| | - Gary M. Dunny
- Department of Microbiology and ImmunologyUniversity of MinnesotaMinneapolisMN 55455
| | - Nita H. Salzman
- Department of PediatricsMedical College of WisconsinMilwaukeeWI 53226
- Department of Microbiology and ImmunologyMedical College of WisconsinMilwaukeeWI 53226
| | | |
Collapse
|
44
|
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
|
45
|
Barbaro MR, Fuschi D, Cremon C, Carapelle M, Dino P, Marcellini MM, Dothel G, De Ponti F, Stanghellini V, Barbara G. Escherichia coli Nissle 1917 restores epithelial permeability alterations induced by irritable bowel syndrome mediators. Neurogastroenterol Motil 2018; 30:e13388. [PMID: 29956419 DOI: 10.1111/nmo.13388] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/03/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intestinal permeability is altered in a subgroup of irritable bowel syndrome (IBS) patients and may contribute to symptom development. The aim of this study was to evaluate the in vitro effect of the probiotic Escherichia coli Nissle 1917 (EcN) on Caco-2 permeability alterations induced by mediators released by IBS mucosal biopsies compared to asymptomatic controls (AC). METHODS Caco-2 cells were used as an in vitro model of intestinal permeability. Seven AC and 28 well-phenotyped IBS (9 IBS-D, 8 IBS-C, and 11 IBS-M) patients were enrolled. Mucosal mediators spontaneously released (SUP) by IBS and AC biopsies were collected. Two concentrations of EcN (108 and 106 ) were applied to Caco-2 with or without SUP or SLIGRL (a protease-activated receptor-2 activating peptide), tumor necrosis factor-α, and interferon-γ. Paracellular permeability was assessed by evaluating the flow of sulfonic-acid conjugated to fluorescein through Caco-2 monolayer. KEY RESULTS EcN 108 significantly reinforced Caco-2 monolayer compared to cells incubated with medium alone. IBS SUP induced a significant increase in paracellular permeability compared to AC SUP, independently of IBS bowel habit. EcN 108 induced a significant recovery of permeability rate compared to IBS SUP. Permeability increase induced by IBS SUP significantly correlated with severity and frequency of abdominal pain and abdominal distension. The co-incubation of EcN and IBS SUP abolished the above significant correlations. CONCLUSIONS AND INFERENCES EcN reinforces the integrity of Caco-2 monolayer and reverts the increase of permeability induced by mediators released by IBS biopsies. Future studies should investigate EcN therapeutic potentials in IBS.
Collapse
Affiliation(s)
- M R Barbaro
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - D Fuschi
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - C Cremon
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - M Carapelle
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - P Dino
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - M M Marcellini
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - G Dothel
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - F De Ponti
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - V Stanghellini
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| | - G Barbara
- Department of Medical and Surgical Sciences (DIMEC), Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum - University of Bologna and S. Orsola-Malpighi Hospital, Bologna, Italy
| |
Collapse
|
46
|
Vinusha KS, Deepika K, Johnson TS, Agrawal GK, Rakwal R. Proteomic studies on lactic acid bacteria: A review. Biochem Biophys Rep 2018; 14:140-148. [PMID: 29872746 PMCID: PMC5986552 DOI: 10.1016/j.bbrep.2018.04.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/02/2018] [Accepted: 04/17/2018] [Indexed: 02/07/2023] Open
Abstract
Probiotics are amongst the most common microbes in the gastro-intestinal tract of humans and other animals. Prominent among probiotics are Lactobacillus and Bifidobacterium. They offer wide-ranging health promoting benefits to the host which include reduction in pathological alterations, stimulation of mucosal immunity and interaction with mediators of inflammation among others. Proteomics plays a vital role in understanding biological functions of a cell. Proteomics is also slowly and steadily adding to the existing knowledge on role of probiotics. In this paper, the proteomics of probiotics, with special reference to lactic acid bacteria is reviewed with a view to understand i) proteome map, ii) mechanism of adaptation to harsh gut environment such as low pH and bile acid, iii) role of cell surface proteins in adhering to intestinal epithelial cells, and iv) as a tool to answer basic cell functions. We have also reviewed various analytical methods used to carry out proteome analysis, in which 2D-MS and LC-MS/MS approaches were found to be versatile methods to perform high-throughput sample analyses even for a complex gut samples. Further, we present future road map of understanding gut microbes combining meta-proteomics, meta-genomics, meta-transcriptomics and -metabolomics.
Collapse
Affiliation(s)
- K Sri Vinusha
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - K Deepika
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - T Sudhakar Johnson
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - Ganesh K Agrawal
- Research Laboratory for Biotechnology and Biochemistry (RLABB), GPO Box 13265, Kathmandu, Nepal.,GRADE Academy Private Limited, Adarsh Nagar-13, Birgunj, Nepal
| | - Randeep Rakwal
- Research Laboratory for Biotechnology and Biochemistry (RLABB), GPO Box 13265, Kathmandu, Nepal.,GRADE Academy Private Limited, Adarsh Nagar-13, Birgunj, Nepal.,Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8574, Japan.,Global Research Center for Innovative Life Science, Peptide Drug Innovation, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 4-41 Ebara 2-chome, Shinagawa, Tokyo 142-8501, Japan
| |
Collapse
|
47
|
Zilelidou EA, Skandamis PN. Growth, detection and virulence of Listeria monocytogenes in the presence of other microorganisms: microbial interactions from species to strain level. Int J Food Microbiol 2018; 277:10-25. [PMID: 29677551 DOI: 10.1016/j.ijfoodmicro.2018.04.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 01/28/2023]
Abstract
Like with all food microorganisms, many basic aspects of L. monocytogenes life are likely to be influenced by its interactions with bacteria living in close proximity. This pathogenic bacterium is a major concern both for the food industry and health organizations since it is ubiquitous and able to withstand harsh environmental conditions. Due to the ubiquity of Listeria monocytogenes, various strains may contaminate foods at different stages of the supply chain. Consequently, simultaneous exposure of consumers to multiple strains is also possible. In this context even strain-to-strain interactions of L. monocytogenes play a significant role in fundamental processes for the life of the pathogen, such as growth or virulence, and subsequently compromise food safety, affect the evolution of a potential infection, or even introduce bias in the detection by classical enrichment techniques. This article summarizes the impact of microbial interactions on the growth and detection of L. monocytogenes primarily in foods and food-associated environments. Furthermore it provides an overview of L. monocytogenes virulence in the presence of other microorganisms.
Collapse
Affiliation(s)
- Evangelia A Zilelidou
- Agricultural University of Athens, Department of Food Science and Human Nutrition, Laboratory of Food Quality Control and Hygiene, Iera odos 75, 11855 Athens, Greece
| | - Panagiotis N Skandamis
- Agricultural University of Athens, Department of Food Science and Human Nutrition, Laboratory of Food Quality Control and Hygiene, Iera odos 75, 11855 Athens, Greece.
| |
Collapse
|
48
|
Hoffmann S, Walter S, Blume AK, Fuchs S, Schmidt C, Scholz A, Gerlach RG. High-Throughput Quantification of Bacterial-Cell Interactions Using Virtual Colony Counts. Front Cell Infect Microbiol 2018; 8:43. [PMID: 29497603 PMCID: PMC5818393 DOI: 10.3389/fcimb.2018.00043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/31/2018] [Indexed: 11/13/2022] Open
Abstract
The quantification of bacteria in cell culture infection models is of paramount importance for the characterization of host-pathogen interactions and pathogenicity factors involved. The standard to enumerate bacteria in these assays is plating of a dilution series on solid agar and counting of the resulting colony forming units (CFU). In contrast, the virtual colony count (VCC) method is a high-throughput compatible alternative with minimized manual input. Based on the recording of quantitative growth kinetics, VCC relates the time to reach a given absorbance threshold to the initial cell count using a series of calibration curves. Here, we adapted the VCC method using the model organism Salmonella enterica sv. Typhimurium (S. Typhimurium) in combination with established cell culture-based infection models. For HeLa infections, a direct side-by-side comparison showed a good correlation of VCC with CFU counting after plating. For MDCK cells and RAW macrophages we found that VCC reproduced the expected phenotypes of different S. Typhimurium mutants. Furthermore, we demonstrated the use of VCC to test the inhibition of Salmonella invasion by the probiotic E. coli strain Nissle 1917. Taken together, VCC provides a flexible, label-free, automation-compatible methodology to quantify bacteria in in vitro infection assays.
Collapse
Affiliation(s)
| | - Steffi Walter
- Project Group 5, Robert Koch Institute, Wernigerode, Germany
| | - Anne-Kathrin Blume
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle, Germany
| | - Stephan Fuchs
- Division 13: Nosocomial Pathogens and Antibiotic Resistances, Robert Koch Institute, Wernigerode, Germany
| | | | | | | |
Collapse
|
49
|
Multifaceted Defense against Listeria monocytogenes in the Gastro-Intestinal Lumen. Pathogens 2017; 7:pathogens7010001. [PMID: 29271903 PMCID: PMC5874727 DOI: 10.3390/pathogens7010001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/18/2017] [Accepted: 12/20/2017] [Indexed: 02/07/2023] Open
Abstract
Listeria monocytogenes is a foodborne pathogen that can cause febrile gastroenteritis in healthy subjects and systemic infections in immunocompromised individuals. Despite the high prevalence of L. monocytogenes in the environment and frequent contamination of uncooked meat and poultry products, infections with this pathogen are relatively uncommon, suggesting that protective defenses in the general population are effective. In the mammalian gastrointestinal tract, a variety of defense mechanisms prevent L. monocytogenes growth, epithelial penetration and systemic dissemination. Among these defenses, colonization resistance mediated by the gut microbiota is crucial in protection against a range of intestinal pathogens, including L. monocytogenes. Here we review defined mechanisms of defense against L. monocytogenes in the lumen of the gastro-intestinal tract, with particular emphasis on protection conferred by the autochthonous microbiota. We suggest that selected probiotic species derived from the microbiota may be developed for eventual clinical use to enhance resistance against L. monocytogenes infections.
Collapse
|
50
|
Chiu L, Bazin T, Truchetet ME, Schaeverbeke T, Delhaes L, Pradeu T. Protective Microbiota: From Localized to Long-Reaching Co-Immunity. Front Immunol 2017; 8:1678. [PMID: 29270167 PMCID: PMC5725472 DOI: 10.3389/fimmu.2017.01678] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/15/2017] [Indexed: 12/17/2022] Open
Abstract
Resident microbiota do not just shape host immunity, they can also contribute to host protection against pathogens and infectious diseases. Previous reviews of the protective roles of the microbiota have focused exclusively on colonization resistance localized within a microenvironment. This review shows that the protection against pathogens also involves the mitigation of pathogenic impact without eliminating the pathogens (i.e., “disease tolerance”) and the containment of microorganisms to prevent pathogenic spread. Protective microorganisms can have an impact beyond their niche, interfering with the entry, establishment, growth, and spread of pathogenic microorganisms. More fundamentally, we propose a series of conceptual clarifications in support of the idea of a “co-immunity,” where an organism is protected by both its own immune system and components of its microbiota.
Collapse
Affiliation(s)
- Lynn Chiu
- University of Bordeaux, CNRS, ImmunoConcept, UMR 5164, Bordeaux, France
| | - Thomas Bazin
- University of Bordeaux, INRA, Mycoplasmal and Chlamydial Infections in Humans, EA 3671, Bordeaux, France.,Department of Hepato-Gastroenterology, Bordeaux Hospital University Center, Pessac, France
| | | | - Thierry Schaeverbeke
- University of Bordeaux, INRA, Mycoplasmal and Chlamydial Infections in Humans, EA 3671, Bordeaux, France.,Department of Rheumatology, Bordeaux Hospital University Center, Bordeaux, France
| | - Laurence Delhaes
- Department of Parasitology and Mycology, Bordeaux Hospital University Center, Bordeaux, France.,University of Bordeaux, INSERM, Cardio-Thoracic Research Centre of Bordeaux, U1045, Bordeaux, France
| | - Thomas Pradeu
- University of Bordeaux, CNRS, ImmunoConcept, UMR 5164, Bordeaux, France
| |
Collapse
|