1
|
Li P, Yin R, Cheng J, Lin J. Bacterial Biofilm Formation on Biomaterials and Approaches to Its Treatment and Prevention. Int J Mol Sci 2023; 24:11680. [PMID: 37511440 PMCID: PMC10380251 DOI: 10.3390/ijms241411680] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Bacterial biofilms can cause widespread infection. In addition to causing urinary tract infections and pulmonary infections in patients with cystic fibrosis, biofilms can help microorganisms adhere to the surfaces of various medical devices, causing biofilm-associated infections on the surfaces of biomaterials such as venous ducts, joint prostheses, mechanical heart valves, and catheters. Biofilms provide a protective barrier for bacteria and provide resistance to antimicrobial agents, which increases the morbidity and mortality of patients. This review summarizes biofilm formation processes and resistance mechanisms, as well as the main features of clinically persistent infections caused by biofilms. Considering the various infections caused by clinical medical devices, we introduce two main methods to prevent and treat biomaterial-related biofilm infection: antibacterial coatings and the surface modification of biomaterials. Antibacterial coatings depend on the covalent immobilization of antimicrobial agents on the coating surface and drug release to prevent and combat infection, while the surface modification of biomaterials affects the adhesion behavior of cells on the surfaces of implants and the subsequent biofilm formation process by altering the physical and chemical properties of the implant material surface. The advantages of each strategy in terms of their antibacterial effect, biocompatibility, limitations, and application prospects are analyzed, providing ideas and research directions for the development of novel biofilm infection strategies related to therapeutic materials.
Collapse
Affiliation(s)
| | | | | | - Jinshui Lin
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan’an University, Yan’an 716000, China; (P.L.); (R.Y.); (J.C.)
| |
Collapse
|
2
|
Zhao B, Osbelt L, Lesker TR, Wende M, Galvez EJC, Hönicke L, Bublitz A, Greweling-Pils MC, Grassl GA, Neumann-Schaal M, Strowig T. Helicobacter spp. are prevalent in wild mice and protect from lethal Citrobacter rodentium infection in the absence of adaptive immunity. Cell Rep 2023; 42:112549. [PMID: 37245209 DOI: 10.1016/j.celrep.2023.112549] [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: 01/14/2023] [Revised: 03/15/2023] [Accepted: 05/04/2023] [Indexed: 05/30/2023] Open
Abstract
Transfer of the gut microbiota from wild to laboratory mice alters the host's immune status and enhances resistance to infectious and metabolic diseases, but understanding of which microbes and how they promote host fitness is only emerging. Our analysis of metagenomic sequencing data reveals that Helicobacter spp. are enriched in wild compared with specific-pathogen-free (SPF) and conventionally housed mice, with multiple species commonly co-colonizing their hosts. We create laboratory mice harboring three non-SPF Helicobacter spp. to evaluate their effect on mucosal immunity and colonization resistance to the enteropathogen Citrobacter rodentium. Our experiments reveal that Helicobacter spp. interfere with C. rodentium colonization and attenuate C. rodentium-induced gut inflammation in wild-type (WT) mice, even preventing lethal infection in Rag2-/- SPF mice. Further analyses suggest that Helicobacter spp. interfere with tissue attachment of C. rodentium, putatively by reducing the availability of mucus-derived sugars. These results unveil pivotal protective functions of wild mouse microbiota constituents against intestinal infection.
Collapse
Affiliation(s)
- Bei Zhao
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Lisa Osbelt
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany; ESF International Graduate School on Analysis, Imaging, and Modelling of Neuronal and Inflammatory Processes, Otto von Guericke University, Magdeburg, Germany
| | - Till Robin Lesker
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Marie Wende
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany; ESF International Graduate School on Analysis, Imaging, and Modelling of Neuronal and Inflammatory Processes, Otto von Guericke University, Magdeburg, Germany
| | - Eric J C Galvez
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Lisa Hönicke
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Arne Bublitz
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | | - Guntram A Grassl
- Department of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Meina Neumann-Schaal
- Bacterial Metabolomics, Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, Braunschweig, Germany; German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; Centre for Individualized Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Center for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|
3
|
Chen GY, Thorup NR, Miller AJ, Li YC, Ayres JS. Cooperation between physiological defenses and immune resistance produces asymptomatic carriage of a lethal bacterial pathogen. SCIENCE ADVANCES 2023; 9:eadg8719. [PMID: 37352357 PMCID: PMC10289649 DOI: 10.1126/sciadv.adg8719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/18/2023] [Indexed: 06/25/2023]
Abstract
Animals evolved two defense strategies to survive infections. Antagonistic strategies include immune resistance mechanisms that operate to kill invading pathogens. Cooperative or physiological defenses mediate host adaptation to the infected state, limiting physiological damage and disease, without killing the pathogen, and have been shown to cause asymptomatic carriage and transmission of lethal pathogens. Here, we demonstrate that physiological defenses cooperate with the adaptive immune response to generate long-term asymptomatic carriage of the lethal enteric murine pathogen, Citrobacter rodentium. Asymptomatic carriage of genetically virulent C. rodentium provided immune resistance against subsequent infections. Immune protection was dependent on systemic antibody responses and pathogen virulence behavior rather than the recognition of specific virulent antigens. Last, we demonstrate that an avirulent strain of C. rodentium in the field has background mutations in genes that are important for LPS structure. Our work reveals insight into how asymptomatic infections can arise mechanistically with immune resistance, mediating exclusion of phenotypically virulent enteric pathogen to promote asymptomatic carriage.
Collapse
Affiliation(s)
- Grischa Y. Chen
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Natalia R. Thorup
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Abigail J. Miller
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Yao-Cheng Li
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Janelle S. Ayres
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
4
|
Harishankar A, Viswanathan VK. Attaching and effacing pathogens modulate host mitochondrial structure and function. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 377:65-86. [PMID: 37268351 PMCID: PMC11321239 DOI: 10.1016/bs.ircmb.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC) are human enteric pathogens that contribute significantly to morbidity and mortality worldwide. These extracellular pathogens attach intimately to intestinal epithelial cells and cause signature lesions by effacing the brush border microvilli, a property they share with other "attaching and effacing" (A/E) bacteria, including the murine pathogen Citrobacter rodentium. A/E pathogens use a specialized apparatus called a type III secretion system (T3SS) to deliver specific proteins directly into the host cytosol and modify host cell behavior. The T3SS is essential for colonization and pathogenesis, and mutants lacking this apparatus fail to cause disease. Thus, deciphering effector-induced host cell modifications is critical for understanding A/E bacterial pathogenesis. Several of the ∼20-45 effector proteins delivered into the host cell modify disparate mitochondrial properties, some via direct interactions with the mitochondria and/or mitochondrial proteins. In vitro studies have uncovered the mechanistic basis for the actions of some of these effectors, including their mitochondrial targeting, interaction partners, and consequent impacts on mitochondrial morphology, oxidative phosphorylation and ROS production, disruption of membrane potential, and intrinsic apoptosis. In vivo studies, mostly relying on the C. rodentium/mouse model, have been used to validate a subset of the in vitro observations; additionally, animal studies reveal broad changes to intestinal physiology that are likely accompanied by mitochondrial alterations, but the mechanistic underpinnings remain undefined. This chapter provides an overview of A/E pathogen-induced host alterations and pathogenesis, specifically focusing on mitochondria-targeted effects.
Collapse
Affiliation(s)
- Anusha Harishankar
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, United States
| | - V K Viswanathan
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, United States; The BIO5 Institute for Collaborative Research, The University of Arizona, Tucson, AZ, United States; Department of Immunobiology, The University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
5
|
Chen GY, Thorup NR, Miller AJ, Li YC, Ayres JS. Cooperation between physiological defenses and immune resistance produces asymptomatic carriage of a lethal bacterial pathogen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.22.525099. [PMID: 36711884 PMCID: PMC9882269 DOI: 10.1101/2023.01.22.525099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Animals have evolved two defense strategies to survive infections. Antagonistic strategies include mechanisms of immune resistance that operate to sense and kill invading pathogens. Cooperative or physiological defenses mediate host adaptation to the infected state, limiting physiological damage and disease, without killing the pathogen, and have been shown to cause asymptomatic carriage and transmission of lethal pathogens. Here we demonstrate that physiological defenses cooperate with the adaptive immune response to generate long-term asymptomatic carriage of the lethal enteric murine pathogen, Citrobacter rodentium. Asymptomatic carriage of genetically virulent C. rodentium provided immune resistance against subsequent infections. Host immune protection was dependent on systemic antibody responses and pathogen virulence behavior, rather than the recognition of specific virulent factor antigens. Finally, we demonstrate that an avirulent strain of C. rodentium in the field has background mutations in two genes that are important for LPS structure. Our work reveals novel insight into how asymptomatic infections can arise mechanistically with immune resistance, mediating exclusion of phenotypically virulent enteric pathogen to promote asymptomatic carriage.
Collapse
Affiliation(s)
- Grischa Y Chen
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Natalia R Thorup
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Abigail J Miller
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Yao-Cheng Li
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Janelle S Ayres
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| |
Collapse
|
6
|
Herzog MKM, Cazzaniga M, Peters A, Shayya N, Beldi L, Hapfelmeier S, Heimesaat MM, Bereswill S, Frankel G, Gahan CG, Hardt WD. Mouse models for bacterial enteropathogen infections: insights into the role of colonization resistance. Gut Microbes 2023; 15:2172667. [PMID: 36794831 PMCID: PMC9980611 DOI: 10.1080/19490976.2023.2172667] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023] Open
Abstract
Globally, enteropathogenic bacteria are a major cause of morbidity and mortality.1-3 Campylobacter, Salmonella, Shiga-toxin-producing Escherichia coli, and Listeria are among the top five most commonly reported zoonotic pathogens in the European Union.4 However, not all individuals naturally exposed to enteropathogens go on to develop disease. This protection is attributable to colonization resistance (CR) conferred by the gut microbiota, as well as an array of physical, chemical, and immunological barriers that limit infection. Despite their importance for human health, a detailed understanding of gastrointestinal barriers to infection is lacking, and further research is required to investigate the mechanisms that underpin inter-individual differences in resistance to gastrointestinal infection. Here, we discuss the current mouse models available to study infections by non-typhoidal Salmonella strains, Citrobacter rodentium (as a model for enteropathogenic and enterohemorrhagic E. coli), Listeria monocytogenes, and Campylobacter jejuni. Clostridioides difficile is included as another important cause of enteric disease in which resistance is dependent upon CR. We outline which parameters of human infection are recapitulated in these mouse models, including the impact of CR, disease pathology, disease progression, and mucosal immune response. This will showcase common virulence strategies, highlight mechanistic differences, and help researchers from microbiology, infectiology, microbiome research, and mucosal immunology to select the optimal mouse model.
Collapse
Affiliation(s)
- Mathias K.-M. Herzog
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Monica Cazzaniga
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Audrey Peters
- Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Nizar Shayya
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Luca Beldi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | | | - Markus M. Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Gad Frankel
- Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Cormac G.M. Gahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Wolf-Dietrich Hardt
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Zhang Y, Si X, Yang L, Wang H, Sun Y, Liu N. Association between intestinal microbiota and inflammatory bowel disease. Animal Model Exp Med 2022; 5:311-322. [PMID: 35808814 PMCID: PMC9434590 DOI: 10.1002/ame2.12255] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/21/2022] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel disease (IBD), which includes Crohn's disease (CD) and ulcerative colitis (UC), has emerged as a global disease with high incidence, long duration, devastating clinical symptoms, and low curability (relapsing immune response and barrier function defects). Mounting studies have been performed to investigate its pathogenesis to provide an ever‐expanding arsenal of therapeutic options, while the precise etiology of IBD is not completely understood yet. Recent advances in high‐throughput sequencing methods and animal models have provided new insights into the association between intestinal microbiota and IBD. In general, dysbiosis characterized by an imbalanced microbiota has been widely recognized as a pathology of IBD. However, intestinal microbiota alterations represent the cause or result of IBD process remains unclear. Therefore, more evidences are needed to identify the precise role of intestinal microbiota in the pathogenesis of IBD. Herein, this review aims to outline the current knowledge of commonly used, chemically induced, and infectious mouse models, gut microbiota alteration and how it contributes to IBD, and dysregulated metabolite production links to IBD pathogenesis.
Collapse
Affiliation(s)
- Yunchang Zhang
- Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xuemeng Si
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Ling Yang
- Department of Food and Bioengineering, Beijing Vocational College of Agriculture, Beijing, China
| | - Hui Wang
- Department of Food and Bioengineering, Beijing Vocational College of Agriculture, Beijing, China
| | - Ye Sun
- Institute of Medical Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
8
|
Cho SH, Park JY, Kim CH. Systemic Lectin-Glycan Interaction of Pathogenic Enteric Bacteria in the Gastrointestinal Tract. Int J Mol Sci 2022; 23:1451. [PMID: 35163392 PMCID: PMC8835900 DOI: 10.3390/ijms23031451] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
Microorganisms, such as bacteria, viruses, and fungi, and host cells, such as plants and animals, have carbohydrate chains and lectins that reciprocally recognize one another. In hosts, the defense system is activated upon non-self-pattern recognition of microbial pathogen-associated molecular patterns. These are present in Gram-negative and Gram-positive bacteria and fungi. Glycan-based PAMPs are bound to a class of lectins that are widely distributed among eukaryotes. The first step of bacterial infection in humans is the adhesion of the pathogen's lectin-like proteins to the outer membrane surfaces of host cells, which are composed of glycans. Microbes and hosts binding to each other specifically is of critical importance. The adhesion factors used between pathogens and hosts remain unknown; therefore, research is needed to identify these factors to prevent intestinal infection or treat it in its early stages. This review aims to present a vision for the prevention and treatment of infectious diseases by identifying the role of the host glycans in the immune response against pathogenic intestinal bacteria through studies on the lectin-glycan interaction.
Collapse
Affiliation(s)
- Seung-Hak Cho
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju 28160, Korea; (S.-H.C.); (J.-y.P.)
| | - Jun-young Park
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju 28160, Korea; (S.-H.C.); (J.-y.P.)
| | - Cheorl-Ho Kim
- Glycobiology Unit, Department of Biological Science, Sung Kyunkwan University, Suwon 16419, Korea
| |
Collapse
|
9
|
Bakheet NZ, A.J AQM, B. FI. Intestinal Anti-Inflammatory Improvement with Fenugreek Seeds as A prebiotic and Synbiotic with Lactobacillus acidophilus in Rats Experimentally Infected with Escherichia coli. THE IRAQI JOURNAL OF VETERINARY MEDICINE 2020. [DOI: 10.30539/ijvm.v44i2.984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Synergistic action of probiotics and prebiotics (synbiotic) has been suggested to be more effective than the two separate components in the prevention and treatment of many intestinal and immune diseases. The present study aimed to examine the anti-inflammatory role of Fenugreek as synbiotic with Lactobacillus acidophilus against Escherichia coli. Twenty four adult males of Wister rats aged 3-4 months and weighted 200-250 gm were used and divided into 4 groups: 1st and 2nd groups were negative and positive control (C and C++) fed with basal diet, the 3rd group (T1) fed diet with Fenugreek seeds (5%) and the 4th group (T2) fed with the synbiotic Fenugreek seeds (5%) and L. acidophilus (5 × 108 CFU/ml) for 45 days. After that, rats in the C++, T1, and T2 had induced enteritis by administrating 1 ml (2.5 × 106 cfu/ml) of enteropathogenic E. coli (EPEC O125:H6). The preventive role of prebiotic and synbiotic was evaluated depending on macro and microscopic duodenum pathological changes in correlation with butyric acid production for 7 days of infection. The results of the macro and microscopic scoring of enteritis revealed that the synergistic effects of the synbiotic in preventing E. coli enteritis was favored by an increase in goblet cells mucin secretion. This anti-inflammatory role was significantly increased by synbiotic and correlated with the production of butyric acid. The synbiotic improved the anti-inflammatory response of intestinal mucosa adaptive immunity via elevation of the immunoglobulin IgA from plasma cells. In conclusion, the inclusion of nutritional supplements containing fibers that constitute a source of butyric acid production, such as Fenugreek seeds, would improve intestinal resistance to inflammation by acting as anti-inflammatory through improving intestinal lymphoid tissues and increasing the production of IgA
Collapse
|
10
|
Fathi J, Ebrahimi F, Nazarian S, Hajizade A, Malekzadegan Y, Abdi A. Production of egg yolk antibody (IgY) against shiga-like toxin (stx) and evaluation of its prophylaxis potency in mice. Microb Pathog 2020; 145:104199. [DOI: 10.1016/j.micpath.2020.104199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/26/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023]
|
11
|
Sharba S, Venkatakrishnan V, Padra M, Winther M, Gabl M, Sundqvist M, Wang J, Forsman H, Linden SK. Formyl peptide receptor 2 orchestrates mucosal protection against Citrobacter rodentium infection. Virulence 2020; 10:610-624. [PMID: 31234710 PMCID: PMC6629182 DOI: 10.1080/21505594.2019.1635417] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Citrobacter rodentium is an attaching and effacing intestinal murine pathogen which shares similar virulence strategies with the human pathogens enteropathogenic- and enterohemorrhagic Escherichia coli to infect their host. C. rodentium is spontaneously cleared by healthy wild-type (WT) mice whereas mice lacking Muc2 or specific immune regulatory genes demonstrate an impaired ability to combat the pathogen. Here we demonstrate that apical formyl peptide receptor 2 (Fpr2) expression increases in colonic epithelial cells during C. rodentium infection. Using a conventional inoculum dose of C. rodentium, both WT and Fpr2−/− mice were infected and displayed similar signs of disease, although Fpr2−/− mice recovered more slowly than WT mice. However, Fpr2−/− mice exhibited increased susceptibility to C. rodentium colonization in response to low dose infection: 100% of the Fpr2−/− and 30% of the WT mice became colonized and Fpr2−/− mice developed more severe colitis and more C. rodentium were in contact with the colonic epithelial cells. In line with the larger amount of C. rodentium detected in the spleen in Fpr2−/− mice, more C. rodentium and enteropathogenic Escherichia coli translocated across an in vitro mucosal surface to the basolateral compartment following FPR2 inhibitor treatment. Fpr2−/− mice also lacked the striated inner mucus layer that was present in WT mice. Fpr2−/− mice had decreased mucus production and different mucin O-glycosylation in the colon compared to WT mice, which may contribute to their defect inner mucus layer. Thus, Fpr2 contributes to protection against infection and influence mucus production, secretion and organization.
Collapse
Affiliation(s)
- S Sharba
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy , Gothenburg , Sweden
| | - V Venkatakrishnan
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy , Gothenburg , Sweden
| | - M Padra
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy , Gothenburg , Sweden
| | - M Winther
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy , Gothenburg , Sweden
| | - M Gabl
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy , Gothenburg , Sweden
| | - M Sundqvist
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy , Gothenburg , Sweden
| | - J Wang
- c Cancer and Inflammation Program , National Cancer Institute at Frederick , Frederick , MD , USA
| | - H Forsman
- b Department of Rheumatology and Inflammation Research , Sahlgrenska Academy , Gothenburg , Sweden
| | - S K Linden
- a Department of Medical Biochemistry and Cell Biology , Sahlgrenska Academy , Gothenburg , Sweden
| |
Collapse
|
12
|
Saha P, Yeoh BS, Xiao X, Golonka RM, Singh V, Wang Y, Vijay-Kumar M. PAD4-dependent NETs generation are indispensable for intestinal clearance of Citrobacter rodentium. Mucosal Immunol 2019; 12:761-771. [PMID: 30710097 PMCID: PMC6519124 DOI: 10.1038/s41385-019-0139-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/07/2019] [Accepted: 01/16/2019] [Indexed: 02/04/2023]
Abstract
Peptidyl arginine deiminase-4 (PAD4) is indispensable for generation of neutrophil extracellular traps (NETs), which can provide antimicrobial effects during host innate immune response; however, the role of PAD4 against gastrointestinal infection is largely unknown. Herein, we challenged PAD4-deficient (Pad4-/-) mice and wild-type (WT) littermates with Citrobacter rodentium (CR), and investigated bacteria clearance and gut pathology. Luminal colonization of CR in Pad4-/- mice peaked between 11-14 days post-infection, whereas WT mice suppressed the infection by 14 days. We demonstrated that Pad4-/- mice were unable to form NETs, whereas WT mice showed increased NETs formation in the colon during infection. Pad4-/- mice showed aggravated CR-associated inflammation as indicated by elevated systemic and colonic pro-inflammatory markers. Histological analysis revealed that transmissible colonic hyperplasia, goblet cell depletion, and apoptotic cell death were more pronounced in the colon of CR-infected Pad4-/- mice. Treating WT mice with deoxyribonuclease I, which can disrupt NETs generation, recapitulated the exacerbated CR infection and gut pathology associated with the loss of PAD4. Administration of the PAD4 inhibitor, Cl-amidine also aggravated CR infection, but to a lesser extent. Taken together, our findings highlight the importance of PAD4 in the mucosal clearance of CR and in resolving gut-associated inflammation.
Collapse
Affiliation(s)
- Piu Saha
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Beng San Yeoh
- Graduate Program in Immunology & Infectious Disease, The Pennsylvania State University, University Park, Philadelphia, PA, 16802, USA
| | - Xia Xiao
- Division of Nephrology, MGH, Harvard Medical School, Boston, MA, 02114, USA
| | - Rachel M Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Vishal Singh
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Yanming Wang
- College of Life Sciences & Medicine, Henan University, Kaifeng, 475004, China
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| |
Collapse
|
13
|
Khanifar J, Hosseini RH, Kazemi R, Ramandi MF, Amani J, Salmanian AH. Prevention of EHEC infection by chitosan nano-structure coupled with synthetic recombinant antigen. J Microbiol Methods 2019; 157:100-107. [PMID: 30633949 DOI: 10.1016/j.mimet.2019.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 01/05/2019] [Accepted: 01/07/2019] [Indexed: 10/27/2022]
Abstract
One of highly effective methods for prevention and control of Entrohemorragic Esherichia coli (EHEC) infections is to use vaccination against extremely immunogenic part of attachment factors. In this study rEIT (EspA, Intimin, Tir) was produced in bacteria and then encapsulated with chitosan nanoparticle as a candidate nanovaccine. A chimeric trivalent recombinant protein which was previously found to provide reasonable immunogenicity against E.coli O157:H7 was used as a base. Mice immunized orally with chitosan based nanoparticle containing rEIT antigen. The rEIT-specific immune responses (IgG and IgA) were measured by indirect ELISA. In challenging tests different groups of immunized mice were infected orally with E.coli O157:H7. The results showed that the recombinant nanovaccine candidate could induce the strong humoral and mucosal immune responses and protect the mice from live EHEC O157:H7 challenge. Higher titers of serum anti rEIT IgG were achieved after the last immunization in all of the groups. Comparison of the amount of IgA titers in serum and feces showed higher values for the latter. In vitro study of binding inhibition assay on Caco-2 cell monolayers by pre-incubated antisera with EHEC bacteria, showed that immunized mice antibody could reduce adhesion properties of E. coli O157:H7. In a challenging study with EHEC bacteria, reduction in number of colonies was observed in all of the immunized groups for over two weeks. Results from the present study prove that nanovaccine candidate with rEIT can reduce signs and symptoms of EHEC infections. This novel approach can be a new strategy for inducing immunity against E. coli O157:H7. This study suggests the use of oral -injection combined vaccination routes comparing to other methods available in order to achieve higher humoral and mucosal immunogenicity levels.
Collapse
Affiliation(s)
- Jaleh Khanifar
- Department of Biology, Faculty of Basic Sciences Tehran Shargh, Payaam Noor University, Biochemistry Research Center, Iran
| | - Reza Haji Hosseini
- Department of Biology, Faculty of Basic Sciences Tehran Shargh, Payaam Noor University, Biochemistry Research Center, Iran
| | | | - Mahdi Fasihi Ramandi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ali Hatef Salmanian
- Department of agricultural Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| |
Collapse
|
14
|
Gioia-Di Chiacchio RM, Cunha MPV, de Sá LRM, Davies YM, Pereira CBP, Martins FH, Munhoz DD, Abe CM, Franzolin MR, Dos Santos LF, Guth BEC, Elias WP, Piazza RMF, Knöbl T. Novel Hybrid of Typical Enteropathogenic Escherichia coli and Shiga-Toxin-Producing E. coli (tEPEC/STEC) Emerging From Pet Birds. Front Microbiol 2018; 9:2975. [PMID: 30574131 PMCID: PMC6291465 DOI: 10.3389/fmicb.2018.02975] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/19/2018] [Indexed: 12/01/2022] Open
Abstract
Exotic psittacine birds have been implicated as reservoir of diarrheagenic Escherichia coli (E. coli), including enteropathogenic E. coli (EPEC) and Shiga-toxin producing E. coli (STEC). Here, we present a genotypic and phenotypic characterization of typical EPEC/STEC hybrid strains isolated from exotic psittacine birds. The strains were positive for eae, bfpA, and stx2f genes, belong to serotype O137:H6 and ST2678. Two strains were subject to whole genome sequencing, confirming the presence of the virulence factors of both E. coli pathotypes. Phenotypical in vitro tests confirmed their ability to adhere to HeLa cells and cause cytotoxicity to Vero cells. The rabbit ileal loop assays showed the attaching and effacing lesion, in addition to inflammatory process and overproduction of intestinal mucus. This is the first report of hybrid typical EPEC/STEC (O137:H6/ST2678) strains isolated from companion psittacine birds and the results suggest zoonotic risks.
Collapse
Affiliation(s)
- Rosely Martins Gioia-Di Chiacchio
- Department of Pathology, School of Veterinary Medicine and Animal Science, São Paulo, Brazil.,School of Veterinary Medicine, Paulista University, São Paulo, Brazil
| | | | | | - Yamê Minieiro Davies
- Department of Pathology, School of Veterinary Medicine and Animal Science, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | - Terezinha Knöbl
- Department of Pathology, School of Veterinary Medicine and Animal Science, São Paulo, Brazil
| |
Collapse
|
15
|
Blanco Crivelli X, Bonino MP, Von Wernich Castillo P, Navarro A, Degregorio O, Bentancor A. Detection and Characterization of Enteropathogenic and Shiga Toxin-Producing Escherichia coli Strains in Rattus spp. from Buenos Aires. Front Microbiol 2018; 9:199. [PMID: 29497406 PMCID: PMC5819574 DOI: 10.3389/fmicb.2018.00199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 01/29/2018] [Indexed: 11/24/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) and Shiga toxin-producing E. coli (STEC) are pathovars of E. coli that impact human health by causing childhood diseases. In this work, 118 synanthropic rodents of the genus Rattus from Buenos Aires, Argentina were evaluated as EPEC and STEC carriers. Rectal swab samples from captured animals were evaluated by conventional PCR to detect the presence of the eae, stx1, stx2, and rfbO157 genes. Twenty-one isolates were obtained (17 EPEC isolates from seven animals and four STEC isolates from the same animal). All EPEC isolates tested negative for the presence of the bfpA gene. One EPEC isolate carried the iha gene, and five EPEC isolates carried the toxB gene. STEC isolates exhibited two different virulence profiles: stx1a/stx2a/stx2c/stx2d/saa/ehxA/subA (3/4) and stx1a/stx2a/saa/ehxA/subA (1/4). EPEC isolate serotypes included O109:H46 (7), O71:H40 (4), O71:NM (2), O138:H40 (1), O108:H21 (1), O88:H25 (1), and O76:NM (1), and STEC isolates belonged to the O108:H11 (4) serotype. Antimicrobial susceptibility testing was carried out, and resistance to tetracycline was observed in one EPEC strain. Our results demonstrate that Rattus spp. may act as carriers of EPEC and STEC strains and may be involved in the epidemiology of diarrheal disease in infancy.
Collapse
Affiliation(s)
- Ximena Blanco Crivelli
- Microbiología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María P Bonino
- Microbiología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Armando Navarro
- Departamento de Salud Pública, Facultad de Medicina, Universidad Nacional de México, Mexico City, Mexico
| | - Osvaldo Degregorio
- Salud Pública, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adriana Bentancor
- Microbiología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
16
|
Hosseini ZS, Amani J, Baghbani Arani F, Nazarian S, Motamedi MJ, Shafighian F. Immunogenicity of the nanovaccine containing intimin recombinant protein in the BALB/c mice. Clin Exp Vaccine Res 2018; 7:51-60. [PMID: 29399580 PMCID: PMC5795045 DOI: 10.7774/cevr.2018.7.1.51] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/29/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
Purpose Escherichia coli O157:H7 is one of the most important pathogens which create hemorrhagic colitis and hemolytic uremic syndrome in human. It is one of the most prevalent causes of diarrhea leading to death of many people every year. The first diagnosed gene in the locus of enterocyte effacement pathogenicity island is eae gene. The product of this gene is a binding protein called intimin belonging to the group of external membrane proteins regarded as a good stimulants of the immune system. Chitosan with its lipophilic property is an environmentally friendly agent able to return to the environment. Materials and Methods Intimin recombinant protein was expressed in pET28a vector with eae gene and purification was performed using Ni-NTA and finally the recombinant protein was approved through western blotting. This protein was encapsulated using chitosan nanoparticles and the size of nanoparticles was measured by Zetasizer. Intimin encapsulated was prescribed for three sessions among three groups of oral, injection, and oral-injection using Chitosan nanoparticles. Challenge was performed for all three groups with 108E. coli O157:H7 bacteria. Results Intimin produced by chitosan nanoparticles improves immunological responses through the adjuvant nature of chitosan nanoparticles. Chitosan may be used as a carrier for transportation of the prescribed vaccine. Among the mice, encapsulated intimin could be able to provide suitable titers of IgG and IgA by the aid of chitosan nanoparticles. Results of mice challenge showed that decreased the bacterial shedding significantly. Conclusion Results showed that the chitosan nanovaccine with intimin protein may be used as a suitable candidate vaccine against E. coli O157:H7.
Collapse
Affiliation(s)
- Zahra Sadat Hosseini
- Department of Genetics and Biotechnology, School of Biological Science, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Fahimeh Baghbani Arani
- Department of Genetics and Biotechnology, School of Biological Science, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Shahram Nazarian
- Department of Biology, Faculty of Science, Imam Hossain University, Tehran, Iran
| | | | - Fatemeh Shafighian
- Pharmaceutical Sciences Branch, Pharmaceutical Sciences Research Center, Islamic Azad University (IAUPS), Tehran, Iran
| |
Collapse
|
17
|
Bouladoux N, Harrison OJ, Belkaid Y. The Mouse Model of Infection with Citrobacter rodentium. ACTA ACUST UNITED AC 2017; 119:19.15.1-19.15.25. [PMID: 29091261 DOI: 10.1002/cpim.34] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Citrobacter rodentium is a murine mucosal pathogen used as a model to elucidate the molecular and cellular pathogenesis of infection with two clinically important human gastrointestinal pathogens, enteropathogenic Escherichia coli (EPEC) and enterohaemorrhagic E. coli (EHEC). C. rodentium infection provides an excellent model to study different aspects of host-pathogen interaction in the gut, including intestinal inflammatory responses during bacteria-induced colitis, mucosal healing and epithelial repair, the induction of mucosal immune responses, and the role of the intestinal microbiota in mediating resistance to colonization by enteric pathogens. This unit provides detailed protocols for growing this bacterium, infecting mice by intragastric inoculation, measuring bacterial loads in feces and organs, and monitoring intestinal pathology induced by infection. Additional protocols describe steps needed to create frozen stocks, establish a growth curve, perform ex vivo organ cultures, isolate immune cells from the large intestine, and measure immune response by flow cytometry. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Nicolas Bouladoux
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland.,NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Oliver J Harrison
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland.,NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| |
Collapse
|
18
|
Cao M, Goodrich-Blair H. Ready or Not: Microbial Adaptive Responses in Dynamic Symbiosis Environments. J Bacteriol 2017; 199:e00883-16. [PMID: 28484049 PMCID: PMC5512229 DOI: 10.1128/jb.00883-16] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In mutually beneficial and pathogenic symbiotic associations, microbes must adapt to the host environment for optimal fitness. Both within an individual host and during transmission between hosts, microbes are exposed to temporal and spatial variation in environmental conditions. The phenomenon of phenotypic variation, in which different subpopulations of cells express distinctive and potentially adaptive characteristics, can contribute to microbial adaptation to a lifestyle that includes rapidly changing environments. The environments experienced by a symbiotic microbe during its life history can be erratic or predictable, and each can impact the evolution of adaptive responses. In particular, the predictability of a rhythmic or cyclical series of environments may promote the evolution of signal transduction cascades that allow preadaptive responses to environments that are likely to be encountered in the future, a phenomenon known as adaptive prediction. In this review, we summarize environmental variations known to occur in some well-studied models of symbiosis and how these may contribute to the evolution of microbial population heterogeneity and anticipatory behavior. We provide details about the symbiosis between Xenorhabdus bacteria and Steinernema nematodes as a model to investigate the concept of environmental adaptation and adaptive prediction in a microbial symbiosis.
Collapse
Affiliation(s)
- Mengyi Cao
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Heidi Goodrich-Blair
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Microbiology, University of Tennessee Knoxville, Knoxville, Tennessee, USA
| |
Collapse
|
19
|
Antibiotic resistance, serogroups, virulence genes, and phylogenetic groups of Escherichia coli isolated from yaks with diarrhea in Qinghai Plateau, China. Gut Pathog 2017; 9:24. [PMID: 28546830 PMCID: PMC5443361 DOI: 10.1186/s13099-017-0174-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/21/2017] [Indexed: 11/15/2022] Open
Abstract
Background Ruminants serve as one of the most important reservoirs for pathogenic Escherichia coli. Infection with E. coli, a foodborne enteropathogen, can lead to asymptomatic infections that can cause life-threatening complications in humans. Therefore, from a clinical and human health perspective, it is important to know which virulence genes, phylogenetic groups, serogroups, and antibiotic resistance patterns are present in E. coli strains in yaks with diarrheic infections. Methods Two-hundred and ninety-two rectal swabs were collected from diarrheic yaks in Qinghai Plateau, China. The antimicrobial sensitivity of each resulting isolate was evaluated according to the disk diffusion method, and different PCR assays were performed for the detection of virulence genes and different phylogroups. Additionally, strains were allocated to different serogroups based on the presence of O antigen via the slide agglutination method. Results Among the E. coli isolates tested, most of the isolates were multidrug resistant (97%) and harbored at least one virulence gene (100%). We observed ten virulence genes (sfa, eaeA, cnf1, etrA, papC, hlyA, aer, faeG, rfc, and sepA), of which sfa was the most commonly found (96.9%). Significant positive associations between some resistance phenotypes and virulence genes were observed (P < 0.05, OR > 1). The majority of the E. coli isolates belonged to phylogroup A (79.5%), and the others belonged to phylogroups B1 (7.5%), D (4.1%), B2 (5.8%), and F (0.7%). Among all the E. coli strains tested, serogroups O91 and O145 were the most prevalent, accounting for 15.4 and 14.4%, respectively. Conclusions Our results suggest that yaks with diarrhea serve as a reservoir of pathogenic E. coli carrying various virulence genes and resistance phenotypes. Therefore, clinicians and relevant authorities must ensure the regulatory use of antimicrobial agents and prevent the spread of these organisms through manure to farm workers and food-processing plants.
Collapse
|
20
|
Chen W, Lu X, Chen Y, Li M, Mo P, Tong Z, Wang W, Wan W, Su G, Xu J, Yu C. Steroid Receptor Coactivator 3 Contributes to Host Defense against Enteric Bacteria by Recruiting Neutrophils via Upregulation of CXCL2 Expression. THE JOURNAL OF IMMUNOLOGY 2017; 198:1606-1615. [PMID: 28053238 DOI: 10.4049/jimmunol.1600300] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 11/30/2016] [Indexed: 01/11/2023]
Abstract
Steroid receptor coactivator 3 (SRC-3) is a transcriptional coactivator that interacts with nuclear receptors and some other transcription factors to enhance their effects on target gene transcription. We reported previously that SRC-3-deficient (SRC-3-/-) mice are extremely susceptible to Escherichia coli-induced septic peritonitis as a result of uncontrolled inflammation and a defect in bacterial clearance. In this study, we observed significant upregulation of SRC-3 in colonic epithelial cells in response to Citrobacter rodentium infection. Based on these findings, we hypothesized that SRC-3 is involved in host defense against attaching and effacing bacterial infection. We compared the responses of SRC-3-/- and wild-type mice to intestinal C. rodentium infection. We found that SRC-3-/- mice exhibited delayed clearance of C. rodentium and more severe tissue pathology after oral infection with C. rodentium compared with wild-type mice. SRC-3-/- mice expressed normal antimicrobial peptides in the colons but exhibited delayed recruitment of neutrophils into the colonic mucosa. Accordingly, SRC-3-/- mice showed a delayed induction of CXCL2 and CXCL5 in colonic epithelial cells, which are responsible for neutrophil recruitment. At the molecular level, we found that SRC-3 can activate the NF-κB signaling pathway to promote CXCL2 expression at the transcriptional level. Collectively, we show that SRC-3 contributes to host defense against enteric bacteria, at least in part via upregulating CXCL2 expression to recruit neutrophils.
Collapse
Affiliation(s)
- Wenbo Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China.,The First Affiliated Hospital of Xiamen University, Xiamen 361001, China; and
| | - Xuqiang Lu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yuan Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Ming Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Pingli Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Zhangwei Tong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Wei Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Wei Wan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Guoqiang Su
- The First Affiliated Hospital of Xiamen University, Xiamen 361001, China; and
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China;
| |
Collapse
|
21
|
Keane OM. Genetic diversity, the virulence gene profile and antimicrobial resistance of clinical mastitis-associated Escherichia coli. Res Microbiol 2016; 167:678-684. [PMID: 27404459 DOI: 10.1016/j.resmic.2016.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/27/2016] [Accepted: 06/29/2016] [Indexed: 12/27/2022]
Abstract
Escherichia coli is a common cause of bovine mastitis, particularly around parturition and early lactation when the host is immunosuppressed. Isolates (n = 37) recovered from cases of clinical mastitis in Ireland were characterised with respect to genotypic diversity, phylogenetic group, virulence gene profile and antimicrobial susceptibility. The isolates were genotypically diverse, belonging to 19 different sequence types. However, the majority (86%) belonged to phylogenetic groups A or B1, groups commonly associated with commensal E. coli. The isolates encoded few virulence genes with iss (increased serum survival, 41%), lpfA (long polar fimbriae, 19%) and astA (enteroaggregative heat-stable toxin, 14%) among the most common virulence genes detected. The only virulence gene to differ in frequency between the phylogenetic groups was lpfA, found exclusively in B1. Resistance to at least one antimicrobial was detected in 16% of isolates. Three isolates were multidrug-resistant, with one resistant to seven antibiotics. There was no relationship between antimicrobial resistance and phylogenetic group. These results indicate that many cases of clinical E. coli mastitis in Ireland may be caused by opportunistic commensal organisms lacking specific virulence genes. However, the organisms represent a reservoir of antimicrobial resistance determinants with the potential to disseminate determinants to other organisms.
Collapse
Affiliation(s)
- Orla M Keane
- Animal & Bioscience Research Department, Teagasc, Grange, Dunsany, Co. Meath, Ireland.
| |
Collapse
|
22
|
Abstract
Erysipelas diagnosed in turkeys, sheep and pigs. Parasitic gastroenteritis reported in cattle on several farms. Unusual presentation of Actinobacillus suis causing spinal abscesses in pigs on a breeder-finisher unit. First APHA diagnosis of oedema disease in pigs in East Anglia for many years. Infectious coryza confirmed in a hobby breeding flock. These are among matters discussed in the Animal and Plant Health Agency's (APHA's) disease surveillance report for November 2014.
Collapse
|
23
|
The Serine Protease Autotransporter Pic Modulates Citrobacter rodentium Pathogenesis and Its Innate Recognition by the Host. Infect Immun 2015; 83:2636-50. [PMID: 25895966 DOI: 10.1128/iai.00025-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bacterial pathogens produce a number of autotransporters that possess diverse functions. These include the family of serine protease autotransporters of Enterobacteriaceae (SPATEs) produced by enteric pathogens such as Shigella flexneri and enteroaggregative Escherichia coli. Of these SPATEs, one termed "protein involved in colonization," or Pic, has been shown to possess mucinase activity in vitro, but to date, its role in in vivo enteric pathogenesis is unknown. Testing a pic null (ΔpicC) mutant in Citrobacter rodentium, a natural mouse pathogen, found that the C. rodentium ΔpicC strain was impaired in its ability to degrade mucin in vitro compared to the wild type. Upon infection of mice, the ΔpicC mutant exhibited a hypervirulent phenotype with dramatically heavier pathogen burdens found in intestinal crypts. ΔpicC mutant-infected mice suffered greater barrier disruption and more severe colitis and weight loss, necessitating their euthanization between 10 and 14 days postinfection. Notably, the virulence of the ΔpicC mutant was normalized when the picC gene was restored; however, a PicC point mutant causing loss of mucinase activity did not replicate the ΔpicC phenotype. Exploring other aspects of PicC function, the ΔpicC mutant was found to aggregate to higher levels in vivo than wild-type C. rodentium. Moreover, unlike the wild type, the C. rodentium ΔpicC mutant had a red, dry, and rough (RDAR) morphology in vitro and showed increased activation of the innate receptor Toll-like receptor 2 (TLR2). Interestingly, the C. rodentium ΔpicC mutant caused a degree of pathology similar to that of wild-type C. rodentium when infecting TLR2-deficient mice, showing that despite its mucinase activity, PicC's major role in vivo may be to limit C. rodentium's stimulation of the host's innate immune system.
Collapse
|
24
|
Lee YS, Yang H, Yang JY, Kim Y, Lee SH, Kim JH, Jang YJ, Vallance BA, Kweon MN. Interleukin-1 (IL-1) signaling in intestinal stromal cells controls KC/ CXCL1 secretion, which correlates with recruitment of IL-22- secreting neutrophils at early stages of Citrobacter rodentium infection. Infect Immun 2015; 83:3257-67. [PMID: 26034212 PMCID: PMC4496604 DOI: 10.1128/iai.00670-15] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 05/28/2015] [Indexed: 01/13/2023] Open
Abstract
Attaching and effacing pathogens, including enterohemorrhagic Escherichia coli in humans and Citrobacter rodentium in mice, raise serious public health concerns. Here we demonstrate that interleukin-1 receptor (IL-1R) signaling is indispensable for protection against C. rodentium infection in mice. Four days after infection with C. rodentium, there were significantly fewer neutrophils (CD11b+ Ly6C+ Ly6G+) in the colons of IL-1R−/− mice than in wild-type mice. Levels of mRNA and protein of KC/CXCL1 were also significantly reduced in colon homogenates of infected IL-1R−/− mice relative to wild-type mice. Of note, infiltrated CD11b+ Ly6C+ Ly6G+ neutrophils were the main source of IL-22 secretion after C. rodentium infection. Interestingly, intestinal stromal cells isolated from IL-1R−/− mice secreted lower levels of KC/CXCL1 than stromal cells from wild-type mice during C. rodentium infection. Similar effects were found when mouse intestinal stromal cells and human nasal polyp stromal cells were treated with IL-1R antagonists (i.e., anakinra) in vitro. These results suggest that IL-1 signaling plays a pivotal role in activating mucosal stromal cells to secrete KC/CXCL1, which is essential for infiltration of IL-22-secreting neutrophils upon bacterial infection.
Collapse
Affiliation(s)
- Yong-Soo Lee
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Hyungjun Yang
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, Canada
| | - Jin-Young Yang
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Yeji Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Su-Hyun Lee
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Ji Heui Kim
- Department of Otolaryngology, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Yong Ju Jang
- Department of Otolaryngology, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Bruce A. Vallance
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, Canada
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
25
|
The secreted effector protein EspZ is essential for virulence of rabbit enteropathogenic Escherichia coli. Infect Immun 2015; 83:1139-49. [PMID: 25561713 DOI: 10.1128/iai.02876-14] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Attaching and effacing (A/E) pathogens adhere intimately to intestinal enterocytes and efface brush border microvilli. A key virulence strategy of A/E pathogens is the type III secretion system (T3SS)-mediated delivery of effector proteins into host cells. The secreted protein EspZ is postulated to promote enterocyte survival by regulating the T3SS and/or by modulating epithelial signaling pathways. To explore the role of EspZ in A/E pathogen virulence, we generated an isogenic espZ deletion strain (ΔespZ) and corresponding cis-complemented derivatives of rabbit enteropathogenic Escherichia coli and compared their abilities to regulate the T3SS and influence host cell survival in vitro. For virulence studies, rabbits infected with these strains were monitored for bacterial colonization, clinical signs, and intestinal tissue alterations. Consistent with data from previous reports, espZ-transfected epithelial cells were refractory to infection-dependent effector translocation. Also, the ΔespZ strain induced greater host cell death than did the parent and complemented strains. In rabbit infections, fecal ΔespZ strain levels were 10-fold lower than those of the parent strain at 1 day postinfection, while the complemented strain was recovered at intermediate levels. In contrast to the parent and complemented mutants, ΔespZ mutant fecal carriage progressively decreased on subsequent days. ΔespZ mutant-infected animals gained weight steadily over the infection period, failed to show characteristic disease symptoms, and displayed minimal infection-induced histological alterations. Terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) staining of intestinal sections revealed increased epithelial cell apoptosis on day 1 after infection with the ΔespZ strain compared to animals infected with the parent or complemented strains. Thus, EspZ-dependent host cell cytoprotection likely prevents epithelial cell death and sloughing and thereby promotes bacterial colonization.
Collapse
|
26
|
Genotypic characterization of canine coronaviruses associated with fatal canine neonatal enteritis in the United States. J Clin Microbiol 2014; 52:4230-8. [PMID: 25253797 DOI: 10.1128/jcm.02158-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Emerging canine coronavirus (CCoV) variants that are associated with systemic infections have been reported in the European Union; however, CCoV-associated disease in the United States is incompletely characterized. The purpose of this study was to correlate the clinicopathological findings and viral antigen distribution with the genotypic characteristics of CCoV in 11 puppies from nine premises in five states that were submitted for diagnostic investigation at Cornell University between 2008 and 2013. CCoV antigen was found in epithelial cells of small intestinal villi in all puppies and the colon in 2 of the 10 puppies where colon specimens were available. No evidence of systemic CCoV infection was found. Comparative sequence analyses of viral RNA extracted from intestinal tissues revealed CCoV-II genotype in 9 out of 11 puppies. Of the nine CCoV-IIs, five were subtyped as group IIa and one as IIb, while three CCoVs could not be subtyped. One of the CCoV-IIa variants was isolated in cell culture. Infection with CCoV alone was found in five puppies, of which two also had small intestinal intussusception. Concurrent infections with either parvovirus (n = 1), attaching-effacing Escherichia coli (n = 4), or protozoan parasites (n = 3) were found in the other six puppies. CCoV is an important differential diagnosis in outbreaks of severe enterocolitis among puppies between 4 days and 21 weeks of age that are housed at high population density. These findings will assist with the rapid laboratory diagnosis of enteritis in puppies and highlight the need for continued surveillance for CCoV variants and intestinal viral diseases of global significance.
Collapse
|
27
|
Papapietro O, Teatero S, Thanabalasuriar A, Yuki KE, Diez E, Zhu L, Kang E, Dhillon S, Muise AM, Durocher Y, Marcinkiewicz MM, Malo D, Gruenheid S. R-spondin 2 signalling mediates susceptibility to fatal infectious diarrhoea. Nat Commun 2013; 4:1898. [PMID: 23695692 PMCID: PMC4844535 DOI: 10.1038/ncomms2816] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 03/27/2013] [Indexed: 12/25/2022] Open
Abstract
Citrobacter rodentium is a natural mouse pathogen widely used as a model for enteropathogenic and enterohemorrhagic Escherichia coli infections in humans. While C. rodentium causes self-limiting colitis in most inbred mouse strains, it induces fatal diarrhea in susceptible strains. The physiological pathways as well as the genetic determinants leading to susceptibility have remained largely uncharacterized. Here we use a forward genetic approach to identify the R-spondin2 gene (Rspo2) as a major determinant of susceptibility to C. rodentium infection. Robust induction of Rspo2 expression during infection in susceptible mouse strains causes a potent Wnt-mediated proliferative response of colonic crypt cells, leading to the generation of an immature and poorly differentiated colonic epithelium with deficiencies in ion-transport components. Our data demonstrate a previously unknown role of R spondins and Wnt signaling in susceptibility to infectious diarrhea and identify Rspo2 as a key molecular link between infection and intestinal homeostasis.
Collapse
Affiliation(s)
- Olivier Papapietro
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada H3A 2B4
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Law RJ, Gur-Arie L, Rosenshine I, Finlay BB. In vitro and in vivo model systems for studying enteropathogenic Escherichia coli infections. Cold Spring Harb Perspect Med 2013; 3:a009977. [PMID: 23457294 DOI: 10.1101/cshperspect.a009977] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) belong to a group of bacteria known as attaching and effacing (A/E) pathogens that cause disease by adhering to the lumenal surfaces of their host's intestinal epithelium. EPEC and EHEC are major causes of infectious diarrhea that result in significant childhood morbidity and mortality worldwide. Recent advances in in vitro and in vivo modeling of these pathogens have contributed to our knowledge of how EPEC and EHEC attach to host cells and subvert host-cell signaling pathways to promote infection and cause disease. A more detailed understanding of how these pathogenic microbes infect their hosts and how the host responds to infection could ultimately lead to new therapeutic strategies to help control these significant enteric pathogens.
Collapse
Affiliation(s)
- Robyn J Law
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | | | | | | |
Collapse
|
29
|
Histopathology. CANINE AND FELINE GASTROENTEROLOGY 2013. [PMCID: PMC7152035 DOI: 10.1016/b978-1-4160-3661-6.00029-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
30
|
|
31
|
Abstract
Gut flora generally contributes to a healthy environment while both commensal and pathogenic bacteria that influence the innate and adaptive immune responses, can cause acute and/or chronic mucosal inflammation. Citrobacter rodentium (C. rodentium) is a member of the family of enteropathogens that provide an excellent in vivo model to investigate the host-pathogen interactions in real-time. It is the etiologic agent for transmissible murine colonic hyperplasia (TMCH) while inflammation following C. rodentium infection is dependent upon the genetic background. Ongoing and completed studies in this model have so far established that Wnt/β-catenin, Notch and PI3K pathways regulate colonic crypt hyperplasia while epithelial-stromal cross-talk, mediated by MEK/ERK/NF-κB signaling, regulates inflammation and/or colitis in susceptible strains. The C. rodentium-induced hyperplastic state also increases the susceptibility to either mutagenic insult or in mice heterozygous for Apc gene. The ability to modulate the host response to C. rodentium infection therefore provides an opportunity to delineate the mechanisms that determine mucosal hyperplasia, intestinal inflammation, and/or neoplasia as disease outcomes.
Collapse
Affiliation(s)
- Shahid Umar
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
32
|
Deng W, Yu HB, de Hoog CL, Stoynov N, Li Y, Foster LJ, Finlay BB. Quantitative proteomic analysis of type III secretome of enteropathogenic Escherichia coli reveals an expanded effector repertoire for attaching/effacing bacterial pathogens. Mol Cell Proteomics 2012; 11:692-709. [PMID: 22661456 DOI: 10.1074/mcp.m111.013672] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Type III secretion systems are central to the pathogenesis and virulence of many important Gram-negative bacterial pathogens, and elucidation of the secretion mechanism and identification of the secreted substrates are critical to our understanding of their pathogenic mechanisms and developing potential therapeutics. Stable isotope labeling with amino acids in cell culture-based mass spectrometry is a quantitative and highly sensitive proteomics tool that we have previously used to successfully analyze the type III secretomes of Citrobacter rodentium and Salmonella enterica serovar Typhimurium. In this report, stable isotope labeling with amino acids in cell culture was used to analyze the type III secretome of enteropathogenic Escherichia coli (EPEC), an important human pathogen, which, together with enterohemorrhagic E. coli and C. rodentium, represents the family of attaching and effacing bacterial pathogens. We not only confirmed all 25 known EPEC type III-secreted proteins and effectors previously identified by conventional molecular and bioinformatical techniques but also identified several new type III-secreted proteins, including two novel effectors, C_0814/NleJ and LifA, that were shown to be translocated into host cells. LifA is a known virulence factor believed to act as a toxin as well as an adhesin, but its mechanism of secretion and function is not understood. With a predicted molecular mass of 366 kDa, LifA is the largest type III effector identified thus far in any pathogen. We further demonstrated that Efa1, ToxB, and Z4332 (homologs of LifA in enterohemorrhagic E. coli) are also type III effectors. This study has comprehensively characterized the type III secretome of EPEC, expanded the repertoire of type III-secreted effectors for the attaching and effacing pathogens, and provided new insights into the mode of function for LifA/Efa1/ToxB/Z4332, an important family of virulence factors.
Collapse
Affiliation(s)
- Wanyin Deng
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | | | | | | | | | | | | |
Collapse
|
33
|
Inoue J, Nishiumi S, Fujishima Y, Masuda A, Shiomi H, Yamamoto K, Nishida M, Azuma T, Yoshida M. Autophagy in the intestinal epithelium regulates Citrobacter rodentium infection. Arch Biochem Biophys 2012; 521:95-101. [PMID: 22475450 DOI: 10.1016/j.abb.2012.03.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/13/2012] [Accepted: 03/14/2012] [Indexed: 10/28/2022]
Abstract
Autophagy, a ubiquitous degradation pathway, is important for the survival and homeostasis of cells. Previous studies have demonstrated the role of autophagy in host defense against bacterial infection, but the importance of autophagy in the intestinal epithelium for the regulation of bacterial infection has not been fully elucidated. In this study, we showed that the essential autophagy protein Atg7 is required for resistance to Citrobacter rodentium infection in the intestinal epithelium. Infected mice in which Atg7 had been conditionally deleted from the intestinal epithelium exhibited greater clinical evidence of disease and higher expression levels of pro-inflammatory cytokine mRNA in the large intestine. Moreover, C. rodentium clearance was reduced in the Atg7 conditional knockout mice. These results demonstrate that autophagy in intestinal epithelial cells plays an important role in host defense against C. rodentium infection and the regulation of C. rodentium infectious colitis.
Collapse
Affiliation(s)
- Jun Inoue
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chu-o-ku, Kobe, Hyogo 650-0017, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Belzer C, Liu Q, Carroll MC, Bry L. THE ROLE OF SPECIFIC IgG AND COMPLEMENT IN COMBATING A PRIMARY MUCOSAL INFECTION OF THE GUT EPITHELIUM. Eur J Microbiol Immunol (Bp) 2011; 1:311-318. [PMID: 22485193 DOI: 10.1556/eujmi.1.2011.4.7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The role of complement and complement-fixing IgG isotypes at mucosal surfaces is ill defined. Previous data have demonstrated that survival of an infection with the attaching and effacing pathogen Citrobacter rodentium requires production of systemic and CD4+ T cell-dependent IgG. We have found that both complement and complement-fixing IgG isotypes are needed to survive a C. rodentium infection. Our results indicate that both IgG and complement C3b enter the gut lumen and bind epithelially adherent, and fecally shed C. rodentium. Furthermore, C3-deficient mice demonstrate a profound survival defect, though means to replenish C3 in systemic or mucosal sites restores the protective capacity of complement in the host. Our data provide evidence that both IgG and complement interact constructively on both sides of the epithelium to fight colonizing mucosal infections.
Collapse
Affiliation(s)
- C Belzer
- Department of Pathology, Harvard University Medical School/Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
35
|
Baines D, Erb S, Lowe R, Turkington K, Sabau E, Kuldau G, Juba J, Masson L, Mazza A, Roberts R. A prebiotic, Celmanax™, decreases Escherichia coli O157:H7 colonization of bovine cells and feed-associated cytotoxicity in vitro. BMC Res Notes 2011; 4:110. [PMID: 21473767 PMCID: PMC3090735 DOI: 10.1186/1756-0500-4-110] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 04/07/2011] [Indexed: 11/20/2022] Open
Abstract
Background Escherichia coli O157:H7 is the most common serovar of enterohemorrhagic E. coli associated with serious human disease outbreaks. Cattle are the main reservoir with E. coli O157:H7 inducing hemorrhagic enteritis in persistent shedding beef cattle, however little is known about how this pathogen affects cattle health. Jejunal Hemorrhage Syndrome (JHS) has unclear etiology but the pathology is similar to that described for E. coli O157:H7 challenged beef cattle suggestive that E. coli O157:H7 could be involved. There are no effective treatments for JHS however new approaches to managing pathogen issues in livestock using prebiotics and probiotics are gaining support. The first objective of the current study was to characterize pathogen colonization in hemorrhaged jejunum of dairy cattle during natural JHS outbreaks. The second objective was to confirm the association of mycotoxigenic fungi in feeds with the development of JHS and also to identify the presence of potential mycotoxins. The third objective was to determine the impact of a prebiotic, Celmanax™, or probiotic, Dairyman's Choice™ paste, on the cytotoxicity associated with feed extracts in vitro. The fourth objective was to determine the impact of a prebiotic or a probiotic on E. coli O157:H7 colonization of mucosal explants and a bovine colonic cell line in vitro. The final objective was to determine if prebiotic and probiotic feed additives could modify the symptoms that preceded JHS losses and the development of new JHS cases. Findings Dairy cattle developed JHS after consuming feed containing several types of mycotoxigenic fungi including Fusarium culmorum, F. poae, F. verticillioides, F. sporotrichioides, Aspergillusflavus, Penicillium roqueforti, P. crustosum, P. paneum and P. citrinum. Mixtures of Shiga toxin - producing Escherichia coli (STEC) colonized the mucosa in the hemorrhaged tissues of the cattle and no other pathogen was identified. The STECs expressed Stx1 and Stx2, but more significantly, Stxs were also present in the blood clot blocking the jejunum. Mycotoxin analysis of the corn crop confirmed the presence of fumonisin, NIV, ZEAR, DON, 15-ADON, 3-ADON, NEO, DAS, HT-2 and T-2. Feed extracts were toxic to enterocytes and 0.1% Celmanax™ removed the cytotoxicity in vitro. There was no effect of Dairyman's Choice™ paste on feed-extract activity in vitro. Fumonisin, T-2, ZEAR and DON were toxic to bovine cells and 0.1% Celmanax™ removed the cytotoxicity in vitro. Celmanax™ also directly decreased E. coli O157:H7 colonization of mucosal explants and a colonic cell line in a dose-dependent manner. There was no effect of Dairyman's Choice™ paste on E. coli O157:H7 colonization in vitro. The inclusion of the prebiotic and probiotic in the feed was associated with a decline in disease. Conclusion The current study confirmed an association between mycotoxigenic fungi in the feed and the development of JHS in cattle. This association was further expanded to include mycotoxins in the feed and mixtures of STECs colonizing the severely hemorrhaged tissues. Future studies should examine the extent of involvement of the different STEC in the infection process. The prebiotic, Celmanax™, acted as an anti-adhesive for STEC colonization and a mycotoxin binder in vitro. Future studies should determine the extent of involvement of the prebiotic in altering disease.
Collapse
Affiliation(s)
- Danica Baines
- Lethbridge Research Centre, 5403 1 Avenue South, P,O, Box 3000, Lethbridge, AB, T1J 4B1, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Petty NK, Feltwell T, Pickard D, Clare S, Toribio AL, Fookes M, Roberts K, Monson R, Nair S, Kingsley RA, Bulgin R, Wiles S, Goulding D, Keane T, Corton C, Lennard N, Harris D, Willey D, Rance R, Yu L, Choudhary JS, Churcher C, Quail MA, Parkhill J, Frankel G, Dougan G, Salmond GPC, Thomson NR. Citrobacter rodentium is an unstable pathogen showing evidence of significant genomic flux. PLoS Pathog 2011; 7:e1002018. [PMID: 21490962 PMCID: PMC3072379 DOI: 10.1371/journal.ppat.1002018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 02/18/2011] [Indexed: 11/18/2022] Open
Abstract
Citrobacter rodentium is a natural mouse pathogen that causes attaching and effacing (A/E) lesions. It shares a common virulence strategy with the clinically significant human A/E pathogens enteropathogenic E. coli (EPEC) and enterohaemorrhagic E. coli (EHEC) and is widely used to model this route of pathogenesis. We previously reported the complete genome sequence of C. rodentium ICC168, where we found that the genome displayed many characteristics of a newly evolved pathogen. In this study, through PFGE, sequencing of isolates showing variation, whole genome transcriptome analysis and examination of the mobile genetic elements, we found that, consistent with our previous hypothesis, the genome of C. rodentium is unstable as a result of repeat-mediated, large-scale genome recombination and because of active transposition of mobile genetic elements such as the prophages. We sequenced an additional C. rodentium strain, EX-33, to reveal that the reference strain ICC168 is representative of the species and that most of the inactivating mutations were common to both isolates and likely to have occurred early on in the evolution of this pathogen. We draw parallels with the evolution of other bacterial pathogens and conclude that C. rodentium is a recently evolved pathogen that may have emerged alongside the development of inbred mice as a model for human disease.
Collapse
Affiliation(s)
- Nicola K. Petty
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
- Department of Biochemistry, University of
Cambridge, Cambridge, United Kingdom
| | - Theresa Feltwell
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Derek Pickard
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Ana L. Toribio
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Maria Fookes
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Kevin Roberts
- Department of Biochemistry, University of
Cambridge, Cambridge, United Kingdom
| | - Rita Monson
- Department of Biochemistry, University of
Cambridge, Cambridge, United Kingdom
| | - Satheesh Nair
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Robert A. Kingsley
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Richard Bulgin
- Centre for Molecular Microbiology and
Infection, Division of Cell and Molecular Biology, Imperial College London,
London, United Kingdom
| | - Siouxsie Wiles
- Centre for Molecular Microbiology and
Infection, Division of Cell and Molecular Biology, Imperial College London,
London, United Kingdom
| | - David Goulding
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Thomas Keane
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Craig Corton
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Nicola Lennard
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - David Harris
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - David Willey
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Richard Rance
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Lu Yu
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Jyoti S. Choudhary
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Carol Churcher
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Michael A. Quail
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Julian Parkhill
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Gad Frankel
- Centre for Molecular Microbiology and
Infection, Division of Cell and Molecular Biology, Imperial College London,
London, United Kingdom
| | - Gordon Dougan
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | | | - Nicholas R. Thomson
- Wellcome Trust Sanger Institute, Wellcome
Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| |
Collapse
|
37
|
Autogenous transcriptional regulation of the regA gene, encoding an AraC-Like, essential virulence regulator in Citrobacter rodentium. J Bacteriol 2011; 193:1777-82. [PMID: 21278290 DOI: 10.1128/jb.01224-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We identified several promoters responsible for the expression of regA, which encodes a global virulence regulator in Citrobacter rodentium. Expression of some of the promoters was strongly autoactivated by RegA in conjunction with bicarbonate. Biochemical and mutational analyses were used to determine the consensus sequence of the RegA-binding sites.
Collapse
|
38
|
The LysR-type transcriptional regulator QseD alters type three secretion in enterohemorrhagic Escherichia coli and motility in K-12 Escherichia coli. J Bacteriol 2010; 192:3699-712. [PMID: 20494990 DOI: 10.1128/jb.00382-10] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 responds to the host-produced epinephrine and norepinephrine, and bacterially produced autoinducer 3 (AI-3), through two-component systems. Further integration of multiple regulatory signaling networks, involving regulators such as the LysR-type transcriptional regulator (LTTR) QseA, promotes effective regulation of virulence factors. These include the production of flagella, a phage-encoded Shiga toxin, and genes within the locus of enterocyte effacement (LEE) responsible for attaching and effacing (AE) lesion formation. Here, we describe a new member of this signaling cascade, an LTTR heretofore renamed QseD (quorum-sensing E. coli regulator D). QseD is present in all enterobacteria but exists almost exclusively in O157:H7 isolates as a helix-turn-helix (HTH) truncated isoform. This "short" isoform (sQseD) is still able to regulate gene expression through a different mechanism than the full-length K-12 E. coli "long" QseD isoform (lQseD). The EHEC Delta qseD mutant exhibits increased expression of all LEE operons and deregulation of AE lesion formation. The loss of qseD in EHEC does not affect motility, but the K-12 Delta qseD mutant is hypermotile. While the lQseD directly binds to the ler promoter, encoding the LEE master regulator, to repress LEE transcription, the sQseD isoform does not. LTTRs bind to DNA as tetramers, and these data suggest that sQseD regulates ler by forming heterotetramers with another LTTR. The LTTRs known to regulate LEE transcription, QseA and LrhA, do not interact with sQseD, suggesting that sQseD acts as a dominant-negative partner with a yet-unidentified LTTR.
Collapse
|
39
|
Bergstrom KSB, Kissoon-Singh V, Gibson DL, Ma C, Montero M, Sham HP, Ryz N, Huang T, Velcich A, Finlay BB, Chadee K, Vallance BA. Muc2 protects against lethal infectious colitis by disassociating pathogenic and commensal bacteria from the colonic mucosa. PLoS Pathog 2010; 6:e1000902. [PMID: 20485566 PMCID: PMC2869315 DOI: 10.1371/journal.ppat.1000902] [Citation(s) in RCA: 465] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 04/08/2010] [Indexed: 12/20/2022] Open
Abstract
Despite recent advances in our understanding of the pathogenesis of attaching and effacing (A/E) Escherichia coli infections, the mechanisms by which the host defends against these microbes are unclear. The goal of this study was to determine the role of goblet cell-derived Muc2, the major intestinal secretory mucin and primary component of the mucus layer, in host protection against A/E pathogens. To assess the role of Muc2 during A/E bacterial infections, we inoculated Muc2 deficient (Muc2−/−) mice with Citrobacter rodentium, a murine A/E pathogen related to diarrheagenic A/E E. coli. Unlike wildtype (WT) mice, infected Muc2−/− mice exhibited rapid weight loss and suffered up to 90% mortality. Stool plating demonstrated 10–100 fold greater C. rodentium burdens in Muc2−/− vs. WT mice, most of which were found to be loosely adherent to the colonic mucosa. Histology of Muc2−/− mice revealed ulceration in the colon amid focal bacterial microcolonies. Metabolic labeling of secreted mucins in the large intestine demonstrated that mucin secretion was markedly increased in WT mice during infection compared to uninfected controls, suggesting that the host uses increased mucin release to flush pathogens from the mucosal surface. Muc2 also impacted host-commensal interactions during infection, as FISH analysis revealed C. rodentium microcolonies contained numerous commensal microbes, which was not observed in WT mice. Orally administered FITC-Dextran and FISH staining showed significantly worsened intestinal barrier disruption in Muc2−/− vs. WT mice, with overt pathogen and commensal translocation into the Muc2−/− colonic mucosa. Interestingly, commensal depletion enhanced C. rodentium colonization of Muc2−/− mice, although colonic pathology was not significantly altered. In conclusion, Muc2 production is critical for host protection during A/E bacterial infections, by limiting overall pathogen and commensal numbers associated with the colonic mucosal surface. Such actions limit tissue damage and translocation of pathogenic and commensal bacteria across the epithelium. Enteropathogenic E. coli (EPEC) and Enterohemorrhagic E. coli (EHEC) are important causes of diarrheal disease and other serious complications worldwide. Despite many studies addressing the pathogenic strategies used by these microbes, how the host protects itself from these pathogens is poorly understood. A critical question we address here is whether the thick mucus layer that overlies the intestinal surface plays a role in host protection. Since EPEC and EHEC do not infect mice efficiently, we used a related mouse pathogen called Citrobacter rodentium to infect and compare responses between wildtype mice and Muc2-deficient mice, which are defective in mucus production. We show that Muc2-deficient mice are extremely susceptible to C. rodentium infection-induced mortality and disease. Muc2-deficient mice were also colonized faster and had higher pathogen burdens throughout the experiment. Resident (non-pathogenic) bacteria were found to interact with C. rodentium and host tissues in Muc2-deficient mice, indicating Muc2 regulates all forms of intestinal microbiota at the gut surface. Deficiency in mucus production also contributed to increased leakiness of the gut, which allowed microbes to enter mucosal tissues. Our study shows that Muc2-dependent mucus production is critical for effective management of both pathogenic and non-pathogenic bacteria during infection by an EPEC/EHEC-like pathogen.
Collapse
Affiliation(s)
- Kirk S. B. Bergstrom
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Vanessa Kissoon-Singh
- Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Deanna L. Gibson
- Department of Biology and Physical Geography, Irving K. Barber School of Arts and Sciences, University of British Columbia-Okanagan, Kelowna, British Columbia, Canada
| | - Caixia Ma
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Marinieve Montero
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Ho Pan Sham
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Natasha Ryz
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Tina Huang
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Anna Velcich
- Department of Oncology, Albert Einstein Cancer Center/Montefiore Medical Center, Bronx, New York, United States of America
| | - B. Brett Finlay
- Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kris Chadee
- Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- * E-mail: (KC); (BAV)
| | - Bruce A. Vallance
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
- * E-mail: (KC); (BAV)
| |
Collapse
|
40
|
Comparative genomics reveal the mechanism of the parallel evolution of O157 and non-O157 enterohemorrhagic Escherichia coli. Proc Natl Acad Sci U S A 2009; 106:17939-44. [PMID: 19815525 DOI: 10.1073/pnas.0903585106] [Citation(s) in RCA: 265] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Among the various pathogenic Escherichia coli strains, enterohemorrhagic E. coli (EHEC) is the most devastating. Although serotype O157:H7 strains are the most prevalent, strains of different serotypes also possess similar pathogenic potential. Here, we present the results of a genomic comparison between EHECs of serotype O157, O26, O111, and O103, as well as 21 other, fully sequenced E. coli/Shigella strains. All EHECs have much larger genomes (5.5-5.9 Mb) than the other strains and contain surprisingly large numbers of prophages and integrative elements (IEs). The gene contents of the 4 EHECs do not follow the phylogenetic relationships of the strains, and they share virulence genes for Shiga toxins and many other factors. We found many lambdoid phages, IEs, and virulence plasmids that carry the same or similar virulence genes but have distinct evolutionary histories, indicating that independent acquisition of these mobile genetic elements has driven the evolution of each EHEC. Particularly interesting is the evolution of the type III secretion system (T3SS). We found that the T3SS of EHECs is composed of genes that were introduced by 3 different types of genetic elements: an IE referred to as the locus of enterocyte effacement, which encodes a central part of the T3SS; SpLE3-like IEs; and lambdoid phages carrying numerous T3SS effector genes and other T3SS-related genes. Our data demonstrate how E. coli strains of different phylogenies can independently evolve into EHECs, providing unique insights into the mechanisms underlying the parallel evolution of complex virulence systems in bacteria.
Collapse
|
41
|
Suwalak S, Voravuthikunchai SP. Morphological and ultrastructural changes in the cell structure of enterohaemorrhagic Escherichia coli O157:H7 following treatment with Quercus infectoria nut galls. JOURNAL OF ELECTRON MICROSCOPY 2009; 58:315-320. [PMID: 19451663 DOI: 10.1093/jmicro/dfp024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Some information is available on the oak (Quercus infectoria) nut gall as an effective medicinal plant against Shiga toxin-producing Escherichia coli (STEC) O157:H7. However, its antibacterial mechanisms have not yet been elucidated. In this study, some antibacterial actions against STEC O157:H7 were investigated by observing cell viability as well as morphological and ultrastructural changes. An ethanolic extract of Q. infectoria demonstrated inhibitory and bactericidal effects on all of the strains tested with minimal inhibition concentrations (MICs) at 0.78-1.56 mg ml(-1) and minimal bactericidal concentrations (MBCs) at 1.56-3.12 mg ml(-1). Cell numbers treated with 4MIC of the extract decreased at least two log-fold within 4 h and were completely killed within 12 h. Scanning electron microscopy illustrated a complete loss of surface appendages and pronounced morphological changes at MIC and 2MIC. The whole cell collapsed at 4MIC. Ultrastructural changes from corresponding transmission electron micrographs further verified that damages in the treated cells increased with the increase in the extract concentrations. At MIC (0.78 mg ml(-1)), there was some evidence that the cytoplasmic membranes of the treated E. coli were bulging and/or ruptured, and the cells appeared to be discharging intracellular materials. At 2MIC, the outer membrane of the treated E. coli which was attached to the cell wall became separated from the wall. Disruption in the outer wall and cytoplasmic membranes, especially at the polar regions of the cells occurred and some vacuolization appeared. At 4MIC, the damage to E. coli cells was extensive, and there was loss of their cellular integrity.
Collapse
Affiliation(s)
- Sakol Suwalak
- Electron Microscopy Unit, Department of Pathology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | | |
Collapse
|
42
|
Escherichia coli O157:H7 strain origin, lineage, and Shiga toxin 2 expression affect colonization of cattle. Appl Environ Microbiol 2009; 75:5074-81. [PMID: 19525271 DOI: 10.1128/aem.00391-09] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Enterohemorrhagic Escherichia coli O157:H7 has evolved into an important human pathogen with cattle as the main reservoir. The recent discovery of E. coli O157:H7-induced pathologies in challenged cattle has suggested that previously discounted bacterial virulence factors may contribute to the colonization of cattle. The objective of the present study was to examine the impact of lineage type, cytotoxin activity, and cytotoxin expression on the amount of E. coli O157:H7 colonization of cattle tissue and cells in vitro. Using selected bovine- and human-origin strains, we determined that lineage type predicted the amount of E. coli O157:H7 strain colonization: lineage I > intermediate lineages > lineage II. All E. coli O157:H7 strain colonization was dose dependent, with threshold colonization at 10(3) to 10(5) CFU and maximum colonization at 10(7) CFU. We also determined that an as-yet-unknown factor of strain origin was the most dominant predictor of the amount of strain colonization in vitro. The amount of E. coli O157:H7 colonization was also influenced by strain cytotoxin activity and the inclusion of cytotoxins from lineage I or intermediate lineage strains increased colonization of a lineage II strain. There was a higher level of expression of the Shiga toxin 1 gene (stx(1)) in human-origin strains than in bovine-origin strains. In addition, lineage I strains expressed higher levels of the Shiga toxin 2 gene (stx(2)). The present study supports a role for strain origin, lineage type, cytotoxin activity, and stx(2) expression in modulating the amount of E. coli O157:H7 colonization of cattle.
Collapse
|
43
|
Chaluvadi MR, Kinloch RD, Nyagode BA, Richardson TA, Raynor MJ, Sherman M, Antonovic L, Strobel HW, Dillehay DL, Morgan ET. Regulation of hepatic cytochrome P450 expression in mice with intestinal or systemic infections of citrobacter rodentium. Drug Metab Dispos 2009; 37:366-74. [PMID: 18971315 PMCID: PMC2631938 DOI: 10.1124/dmd.108.024240] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Accepted: 10/23/2008] [Indexed: 11/22/2022] Open
Abstract
We reported previously that infection of C3H/HeOuJ (HeOu) mice with the murine intestinal pathogen Citrobacter rodentium caused a selective modulation of hepatic cytochrome P450 (P450) gene expression in the liver that was independent of the Toll-like receptor 4. However, HeOu mice are much more sensitive to the pathogenic effects of C. rodentium infection, and the P450 down-regulation was associated with significant morbidity in the animals. Here, we report that oral infection of C57BL/6 mice with C. rodentium, which produced only mild clinical signs and symptoms, produced very similar effects on hepatic P450 expression in this strain. As in HeOu mice, CYP4A mRNAs and proteins were among the most sensitive to down-regulation, whereas CYP4F18 was induced. CYP2D9 mRNA was also induced 8- to 9-fold in the C57BL/6 mice. The time course of P450 regulation followed that of colonic inflammation and bacterial colonization, peaking at 7 to 10 days after infection and returning to normal at 15 to 24 days as the infection resolved. These changes also correlated with the time course of significant elevations in the serum of the proinflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-alpha, as well as of interferon-gamma and IL-2, with serum levels of IL-6 being markedly higher than those of the other cytokines. Intraperitoneal administration of C. rodentium produced a rapid down-regulation of P450 enzymes that was quantitatively and qualitatively different from that of oral infection, although CYP2D9 was induced in both models, suggesting that the effects of oral infection on the liver are not due to bacterial translocation.
Collapse
Affiliation(s)
- Madhusudana R Chaluvadi
- Department of Pharmacology, Emory University School of Medicine, 5119 Rollins Research Center, 1510 Clifton Road, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
La Ragione RM, Best A, Woodward MJ, Wales AD. Escherichia coli O157:H7 colonization in small domestic ruminants. FEMS Microbiol Rev 2008; 33:394-410. [PMID: 19207740 DOI: 10.1111/j.1574-6976.2008.00138.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Enterohaemorrhagic Escherichia coli O157:H7 was first implicated in human disease in the early 1980s, with ruminants cited as the primary reservoirs. Preliminary studies indicated cattle to be the sole source of E. coli O157:H7 outbreaks in humans; however, further epidemiological studies soon demonstrated that E. coli O157:H7 was widespread in other food sources and that a number of transmission routes existed. More recently, small domestic ruminants (sheep and goats) have emerged as important sources of E. coli O157:H7 human infection, particularly with the widespread popularity of petting farms and the increased use of sheep and goat food products, including unpasteurized cheeses. Although the colonization and persistence characteristics of E. coli O157:H7 in the bovine host have been studied intensively, this is not the case for small ruminants. Despite many similarities to the bovine host, the pathobiology of E. coli O157:H7 in small domestic ruminants does appear to differ significantly from that described in cattle. This review aims to critically review the current knowledge regarding colonization and persistence of E. coli O157:H7 in small domestic ruminants, including comparisons with the bovine host where appropriate.
Collapse
Affiliation(s)
- Roberto M La Ragione
- Department of Food and Environmental Safety, Veterinary Laboratories Agency, Addlestone, Surrey, UK.
| | | | | | | |
Collapse
|
45
|
Baines D, Lee B, McAllister T. Heterogeneity in enterohemorrhagicEscherichia coliO157:H7 fecal shedding in cattle is related toEscherichia coliO157:H7 colonization of the small and large intestine. Can J Microbiol 2008; 54:984-95. [DOI: 10.1139/w08-090] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the last decade, Escherichia coli O157:H7 have emerged as important pathogens of the gastrointestinal tract of humans. Healthy cattle have been identified as the primary reservoir, however, the factors affecting heterogeneous E. coli O157:H7 fecal shedding are not fully understood. The aim of this study was to investigate the contribution of E. coli O157:H7 colonization of small and large intestinal sites to the heterogeneity of fecal shedding in cattle. There was a dose-dependant E. coli O157:H7 E318N colonization of duodenum, jejunum, ileum, cecum, ascending colon, spiral colon, descending colon, and the rectoanal junction in vitro with no difference in E. coli O157:H7 colonization of the rectoanal junction and other intestinal sites. There were 10–100 times greater E. coli O157:H7 colonization of intestinal sites from persistent shedding cattle compared with nonpersistent shedding cattle. Novel pathologies were associated with E. coli O157:H7 colonization sites in the small and large intestine. The first pathology, focal petechiae, was present throughout the intestinal tract of cattle that ceased shedding E. coli O157:H7 for 5–12 weeks or in the jejunum, ileum, cecum, and ascending colon of cattle shedding E. coli O157:H7 for 4–5 months. The second pathology, mucosal hemorrhages, was present in the same sites as the focal petechiae in cattle shedding for 5 months and these hemorrhages were in the final stages of repair. Several features of these hemorrhages support this conclusion including the brown appearance, low amount of classic E. coli O157:H7 induced A/E lesions, flattened epithelium, and blunted villi. Although mucosal hemorrhages were present in the jejunum, ileum, cecum, and ascending colon in cattle shedding for 4 months, many other pathologies were also present that were indicative of hemorrhagic enteritis as evidenced by the blood red appearance of hemorrhages, severe edema, and dark red erythema. Escherichia coli O157:H7 were associated with both pathologies suggesting it is the causative agent. The current study supports a relationship between the amount of E. coli O157:H7 colonization in intestinal sites and heterogeneous fecal shedding by cattle.
Collapse
Affiliation(s)
- Danica Baines
- Lethbridge Research Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada
| | - Byron Lee
- Lethbridge Research Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada
| | - Tim McAllister
- Lethbridge Research Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada
| |
Collapse
|
46
|
Foster AP, Otter A, Barlow AM, Pearson GR, Woodward MJ, Higgins RJ. Naturally occurring intestinal lesions in three alpacas (Vicugna pacos) caused by attaching and effacing Escherichia coli. Vet Rec 2008; 162:318-20. [PMID: 18326846 DOI: 10.1136/vr.162.10.318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- A P Foster
- Veterinary Laboratories Agency - Shrewsbury, Kendal Road, Harlescott, Shrewsbury SY1 4HD, UK
| | | | | | | | | | | |
Collapse
|
47
|
Petty NK, Toribio AL, Goulding D, Foulds I, Thomson N, Dougan G, Salmond GPC. A generalized transducing phage for the murine pathogen Citrobacter rodentium. MICROBIOLOGY-SGM 2007; 153:2984-2988. [PMID: 17768241 PMCID: PMC2652034 DOI: 10.1099/mic.0.2007/008888-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A virulent phage (phiCR1) capable of generalized transduction in Citrobacter rodentium was isolated from the environment and characterized. C. rodentium is a natural pathogen of mice, causing transmissible murine colonic hyperplasia. Sequencing of its genome has recently been completed and will soon be fully annotated and published. C. rodentium is an important model organism for infections caused by the human pathogens enteropathogenic and enterohaemorrhagic Escherichia coli (EPEC and EHEC). phiCR1 uses a lipopolysaccharide receptor, has a genome size of approximately 300 kb, and is able to transduce a variety of markers. phiCR1 is the first reported transducing phage for C. rodentium and will be a useful tool for functional genomic analysis of this important natural murine pathogen.
Collapse
Affiliation(s)
- Nicola K. Petty
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | | | | | - Ian Foulds
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | | | - Gordon Dougan
- Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | | |
Collapse
|
48
|
Modulation of intestinal goblet cell function during infection by an attaching and effacing bacterial pathogen. Infect Immun 2007; 76:796-811. [PMID: 17984203 DOI: 10.1128/iai.00093-07] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The attaching and effacing (A/E) bacterial pathogens enteropathogenic Escherichia coli and enterohemorrhagic E. coli and the related mouse pathogen Citrobacter rodentium colonize their hosts' intestines by infecting the apical surfaces of enterocytes, subverting their function, and they ultimately cause diarrhea. Surprisingly, little is known about the interactions of these organisms with goblet cells, which are specialized epithelial cells that secrete the protective molecules Muc2 and trefoil factor 3 (Tff3) into the intestinal lumen. C. rodentium infection leads to dramatic goblet cell depletion within the infected colon, yet it is not clear whether C. rodentium infects goblet cells or if this pathology is pathogen or host mediated. As determined by immunostaining and PCR, both the number of goblet cells and the expression of genes encoding Muc2 and Tff3 were significantly reduced by day 10 postinfection. While electron microscopy and immunostaining revealed that C. rodentium directly infected a fraction of colonic goblet cells, C. rodentium localization did not correlate with goblet cell depletion. To assess the role of the host immune system in these changes, Rag1 knockout (KO) (T- and B-cell-deficient) mice were infected with C. rodentium. Rag1 KO mice did not exhibit the reduction in the number of goblet cells or in mediator (Muc2 and Tff3) expression observed in infected immunocompetent mice. However, reconstitution of Rag1 KO mice with T and B lymphocytes from C57BL/6 mice restored the goblet cell depletion phenotype during C. rodentium infection. In conclusion, these studies demonstrated that while colonic goblet cells can be subject to direct infection and potential subversion by A/E pathogens in vivo, it is the host immune system that primarily modulates the function of these cells during infection.
Collapse
|
49
|
Extensive genomic diversity and selective conservation of virulence-determinants in enterohemorrhagic Escherichia coli strains of O157 and non-O157 serotypes. Genome Biol 2007; 8:R138. [PMID: 17711596 PMCID: PMC2323221 DOI: 10.1186/gb-2007-8-7-r138] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 06/06/2007] [Accepted: 07/10/2007] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Enterohemorrhagic Escherichia coli (EHEC) O157 causes severe food-borne illness in humans. The chromosome of O157 consists of 4.1 Mb backbone sequences shared by benign E. coli K-12, and 1.4 Mb O157-specific sequences encoding many virulence determinants, such as Shiga toxin genes (stx genes) and the locus of enterocyte effacement (LEE). Non-O157 EHECs belonging to distinct clonal lineages from O157 also cause similar illness in humans. According to the 'parallel' evolution model, they have independently acquired the major virulence determinants, the stx genes and LEE. However, the genomic differences between O157 and non-O157 EHECs have not yet been systematically analyzed. RESULTS Using microarray and whole genome PCR scanning analyses, we performed a whole genome comparison of 20 EHEC strains of O26, O111, and O103 serotypes with O157. In non-O157 EHEC strains, although genome sizes were similar with or rather larger than O157 and the backbone regions were well conserved, O157-specific regions were very poorly conserved. Around only 20% of the O157-specific genes were fully conserved in each non-O157 serotype. However, the non-O157 EHECs contained a significant number of virulence genes that are found on prophages and plasmids in O157, and also multiple prophages similar to, but significantly divergent from, those in O157. CONCLUSION Although O157 and non-O157 EHECs have independently acquired a huge amount of serotype- or strain-specific genes by lateral gene transfer, they share an unexpectedly large number of virulence genes. Independent infections of similar but distinct bacteriophages carrying these virulence determinants are deeply involved in the evolution of O157 and non-O157 EHECs.
Collapse
|
50
|
Cookson AL, Bennett J, Thomson-Carter F, Attwood GT. Intimin subtyping ofEscherichia coli: concomitant carriage of multiple intimin subtypes from forage-fed cattle and sheep. FEMS Microbiol Lett 2007; 272:163-71. [PMID: 17521363 DOI: 10.1111/j.1574-6968.2007.00755.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The outer membrane protein, intimin (eae), which mediates bacterial attachment to epithelial cells, is associated with enteropathogenic Escherichia coli and some Shiga toxin-producing E. coli. The eae subtype of E. coli strains isolated from healthy cattle and sheep was identified using a rapid PCR-restriction fragment length polymorphism (RFLP) method to produce profiles that were compared with those generated in silico. The 139 eae-positive E. coli strains were separated into 11 different PCR-RFLP profiles. The most common eae PCR-RFLP type was beta (23.7%), followed by zeta (20.1%), theta (16.5%), iota (12.2%), kappa (8.6%), epsilon (7.2%), gamma (2.9%), nu and beta2 (2.2%) and iota2 (1.4%). Four isolates did not yield a PCR-RFLP amplification product but complete sequencing of the eae gene matched subtype rho. Two different eae variants were isolated from the same swab from 18 different animals and subtype iota was the most 'promiscuous', being isolated with four other eae subtypes from seven separate animals. None of the eae-positive STEC were subtype gamma, which is associated with STEC serogroup O157. This method allowed the rapid identification of eae subtypes and indicates that forage-fed animals possessed a wide diversity of bacterial eae subtypes with a low frequency of eae subtype gamma.
Collapse
Affiliation(s)
- Adrian L Cookson
- Food, Metabolism & Microbiology Section, Food & Health Group, AgResearch, Grasslands Research Centre, Tennent Drive, Palmerston North, New Zealand.
| | | | | | | |
Collapse
|