1
|
Herzog MKM, Peters A, Shayya N, Cazzaniga M, Kaka Bra K, Arora T, Barthel M, Gül E, Maurer L, Kiefer P, Christen P, Endhardt K, Vorholt JA, Frankel G, Heimesaat MM, Bereswill S, Gahan CGM, Claesson MJ, Domingo-Almenara X, Hardt WD. Comparing Campylobacter jejuni to three other enteric pathogens in OligoMM 12 mice reveals pathogen-specific host and microbiota responses. Gut Microbes 2025; 17:2447832. [PMID: 39835346 DOI: 10.1080/19490976.2024.2447832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/22/2025] Open
Abstract
Campylobacter jejuni, non-typhoidal Salmonella spp., Listeria monocytogenes and enteropathogenic/enterohemorrhagic Escherichia coli (EPEC/EHEC) are leading causes of food-borne illness worldwide. Citrobacter rodentium has been used to model EPEC and EHEC infection in mice. The gut microbiome is well-known to affect gut colonization and host responses to many food-borne pathogens. Recent progress has established gnotobiotic mice as valuable models to study how microbiota affect the enteric infections by S. Typhimurium, C. rodentium and L. monocytogenes. However, for C. jejuni, we are still lacking a suitable gnotobiotic mouse model. Moreover, the limited comparability of data across laboratories is often negatively affected by variations between different research facilities or murine microbiotas. In this study, we applied the standardized gnotobiotic OligoMM12 microbiota mouse model and compared the infections in the same facility. We provide evidence of robust colonization and significant pathological changes in OligoMM12 mice following infection with these pathogens. Moreover, we offer insights into pathogen-specific host responses and metabolite signatures, highlighting the advantages of a standardized mouse model for direct comparisons of factors influencing the pathogenesis of major food-borne pathogens. Notably, we reveal for the first time that C. jejuni stably colonizes OligoMM12 mice, triggering inflammation. Additionally, our comparative approach successfully identifies pathogen-specific responses, including the detection of genes uniquely associated with C. jejuni infection in humans. These findings underscore the potential of the OligoMM12 model as a versatile tool for advancing our understanding of food-borne pathogen interactions.
Collapse
Affiliation(s)
- Mathias K-M Herzog
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Audrey Peters
- Department of Life Sciences, MRC Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Nizar Shayya
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Monica Cazzaniga
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Kardokh Kaka Bra
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Trisha Arora
- Omic Sciences Unit, EURECAT - Technology Centre of Catalonia, Reus, Spain
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Ersin Gül
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Luca Maurer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Katharina Endhardt
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Gad Frankel
- Department of Life Sciences, MRC Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Markus M Heimesaat
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Bereswill
- Gastrointestinal Microbiology Research Group, Institute of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - 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
| | - Marcus J Claesson
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | | | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Moya Uribe IA, Terauchi H, Bell JA, Zanetti A, Jantre S, Huebner M, Arshad SH, Ewart SL, Mansfield LS. Fecal microbiota transplants (FMT) of three distinct human communities to germ-free mice exacerbated inflammation and decreased lung function in their offspring. mBio 2025; 16:e0376424. [PMID: 40207915 PMCID: PMC12077122 DOI: 10.1128/mbio.03764-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/21/2025] [Indexed: 04/11/2025] Open
Abstract
Despite explosive rise in allergies, little is known about early life gut microbiota effects on postnatal respiratory function. We hypothesized that Enterobacteriaceae-dominant gut microbiota from eczemic infants increases Type 2 inflammation and decreases lung function in transplanted mice, while Bacteroidaceae-dominant gut microbiota from non-eczemic infants is protective. Fecal microbiota transplants (FMT) from eczemic infants "Infant A" and non-eczemic infants "Infant B" were successfully transplanted into germ-free C57BL/6 mice, passing to offspring unchanged. Infant A and B, Adult C-human-derived (positive control), and Mouse (negative control) microbiotas all in C57BL/6 mice were tested for effects on airway function in nonallergic (phosphate-buffered saline [PBS]) and allergic (house dust mite [HDM]) conditions. Baseline lung mechanics in mice with human microbiotas (HUmicrobiota) were significantly impaired compared to Mouse microbiota controls (MOmicrobiota) with or without HDM; respiratory system resistance (Rrs) was increased (P < 0.05-P < 0.01), and respiratory system compliance (Crs) was decreased (P < 0.05-P < 0.01). HUMicrobiota mice showed a statistically significant impairment compared to MOmicrobiota mice in lung parameters Rrs, Ers, Rn, and G at baseline, and at multiple methacholine (MCh) doses with baseline removed. Impairment manifested as increased small airway resistance and tissue resistance. HDM significantly elevated IL-4, eosinophils, lung inflammation, and mucus cell metaplasia, and decreased macrophages and lung function (P < 0.05) in mice of all microbiotas, yet each HUmicrobiota produced distinct features. Infant B and Adult C mice had elevated basal levels of total IgE compared to MOmicrobiota and Infant A mice (P < 0.05). In HUmicrobiota mice given HDM, only Adult C had elevated IL-5 and IL-13 (P < 0.05), only Adult C and Infant B mice had elevated neutrophils (P < 0.05), and only Infant A had elevated lymphocytes (P < 0.01). IMPORTANCE Fecal microbiota transplants (FMT) of three distinct human communities to germ-free mice exacerbated inflammation and decreased lung function in their offspring. Taxa formerly described to induce an allergic response (agonists) and pro-inflammatory taxa were abundant in HUmicrobiotas compared to MOmicrobiota controls, while taxa formerly described to reduce allergic responses (antagonists) and anti-inflammatory taxa were numerous in MOmicrobiotas and low in HUmicrobiotas. Thus, we largely rejected our hypotheses because data supported multiple pro-inflammatory allergy agonists functioning in a community-wide fashion to impair lung function in the absence of antagonistic anti-inflammatory taxa. Structure of HUmicrobiotas played a key role in determining varied allergic responses and resulting lung impairment, yet, strikingly, all mice with HUmicrobiotas had impaired lung function even in the absence of allergens. Using a comparative approach, we showed that composition of gut microbiota can alter innate/immune regulation in the gut-lung axis to increase baseline lung function responses and the risk of allergic sensitization.
Collapse
Affiliation(s)
- Ivon A. Moya Uribe
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, Michigan, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
- Michigan State University, East Lansing, Michigan, USA
- Comparative Medicine Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Hinako Terauchi
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, Michigan, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
- Michigan State University, East Lansing, Michigan, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Julia A. Bell
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, Michigan, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
- Michigan State University, East Lansing, Michigan, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, USA
| | - Alexander Zanetti
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, Michigan, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Sanket Jantre
- Department of Statistics and Probability, Michigan State University, East Lansing, Michigan, USA
| | - Marianne Huebner
- Department of Statistics and Probability, Michigan State University, East Lansing, Michigan, USA
| | - S. Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- The David Hide Asthma and Allergy Research Centre, Isle of Wight, United Kingdom
- NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Susan L. Ewart
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
- Michigan State University, East Lansing, Michigan, USA
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, USA
| | - Linda S. Mansfield
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, Michigan, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
- Michigan State University, East Lansing, Michigan, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
3
|
Sher AA, Whitehead-Tillery CE, Peer AM, Bell JA, Vocelle DB, Dippel JT, Zhang L, Mansfield LS. Dynamic Spread of Antibiotic Resistance Determinants by Conjugation to a Human-Derived Gut Microbiota in a Transplanted Mouse Model. Antibiotics (Basel) 2025; 14:152. [PMID: 40001396 PMCID: PMC11851821 DOI: 10.3390/antibiotics14020152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/11/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Antibiotic-resistant (AR) bacteria pose an increasing threat to public health, but the dynamics of antibiotic resistance gene (ARG) spread in complex microbial communities are poorly understood. Conjugation is a predominant direct cell-to-cell mechanism for the horizontal gene transfer (HGT) of ARGs. We hypothesized that commensal Escherichia coli donor strains would mediate the conjugative transfer of ARGs to phylogenetically distinct bacteria without antibiotic selection pressure in gastrointestinal tracts of mice carrying a human-derived microbiota with undetectable levels of E. coli. Our objective was to identify a mouse model to study the factors regulating AR transfer by conjugation in the gut. METHODS Two donor E. coli strains were engineered to carry chromosomally encoded red fluorescent protein, and an ARG- and green fluorescent protein (GFP)-encoding broad host range RP4 conjugative plasmid. Mice were orally gavaged with two donor strains (1) E. coli MG1655 or (2) human-derived mouse-adapted E. coli LM715-1 and their colonization assessed by culture over time. Fluorescence-activated cell sorting (FACS) and 16S rDNA sequencing were performed to trace plasmid spread to the microbiota. RESULTS E. coli LM715-1 colonized mice for ten days, while E. coli MG1655 was not recovered after 72 h. Bacterial cells from fecal samples on days 1 and 3 post inoculation were sorted by FACS. Samples from mice given donor E. coli LM715-1 showed an increase in cells expressing green but not red fluorescence compared to pre-inoculation samples. 16S rRNA gene sequencing analysis of FACS GFP positive cells showed that bacterial families Lachnospiraceae, Clostridiaceae, Pseudomonadaceae, Rhodanobacteraceae, Erysipelotrichaceae, Oscillospiraceae, and Butyricicoccaceae were the primary recipients of the RP4 plasmid. CONCLUSIONS Results show this ARG-bearing conjugative RP4 plasmid spread to diverse human gut bacterial taxa within a live animal where they persisted. These fluorescent marker strategies and human-derived microbiota transplanted mice provided a tractable model for investigating the dynamic spread of ARGs within gut microbiota and could be applied rigorously to varied microbiotas to understand conditions facilitating their spread.
Collapse
Affiliation(s)
- Azam A. Sher
- Comparative Enteric Diseases Laboratory, Departments of Large Animal Clinical Sciences and Microbiology, Genetics and Immunology, East Lansing, MI 48824, USA; (A.A.S.); (C.E.W.-T.); (A.M.P.); (J.A.B.)
- Comparative Medicine and Integrative Biology Graduate Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
- BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI 48824, USA
| | - Charles E. Whitehead-Tillery
- Comparative Enteric Diseases Laboratory, Departments of Large Animal Clinical Sciences and Microbiology, Genetics and Immunology, East Lansing, MI 48824, USA; (A.A.S.); (C.E.W.-T.); (A.M.P.); (J.A.B.)
| | - Ashley M. Peer
- Comparative Enteric Diseases Laboratory, Departments of Large Animal Clinical Sciences and Microbiology, Genetics and Immunology, East Lansing, MI 48824, USA; (A.A.S.); (C.E.W.-T.); (A.M.P.); (J.A.B.)
| | - Julia A. Bell
- Comparative Enteric Diseases Laboratory, Departments of Large Animal Clinical Sciences and Microbiology, Genetics and Immunology, East Lansing, MI 48824, USA; (A.A.S.); (C.E.W.-T.); (A.M.P.); (J.A.B.)
| | - Daniel B. Vocelle
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| | - Joshua T. Dippel
- Comparative Enteric Diseases Laboratory, Departments of Large Animal Clinical Sciences and Microbiology, Genetics and Immunology, East Lansing, MI 48824, USA; (A.A.S.); (C.E.W.-T.); (A.M.P.); (J.A.B.)
| | - Lixin Zhang
- Departments of Epidemiology and Biostatistics and Microbiology, Genetics and Immunology, Michigan State University, East Lansing, MI 48824, USA
| | - Linda S. Mansfield
- Comparative Enteric Diseases Laboratory, Departments of Large Animal Clinical Sciences and Microbiology, Genetics and Immunology, East Lansing, MI 48824, USA; (A.A.S.); (C.E.W.-T.); (A.M.P.); (J.A.B.)
- BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
4
|
Hosomi K, Hatanaka N, Hinenoya A, Adachi J, Tojima Y, Furuta M, Uchiyama K, Morita M, Nagatake T, Saika A, Kawai S, Yoshii K, Kondo S, Yamasaki S, Kunisawa J. QcrC is a potential target for antibody therapy and vaccination to control Campylobacter jejuni infection by suppressing its energy metabolism. Front Microbiol 2024; 15:1415893. [PMID: 39015740 PMCID: PMC11250076 DOI: 10.3389/fmicb.2024.1415893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Introduction Campylobacter spp. are a public health concern, yet there is still no effective vaccine or medicine available. Methods Here, we developed a Campylobacter jejuni-specific antibody and found that it targeted a menaquinol cytochrome c reductase complex QcrC. Results The antibody was specifically reactive to multiple C. jejuni strains including clinical isolates from patients with acute enteritis and was found to inhibit the energy metabolism and growth of C. jejuni. Different culture conditions produced different expression levels of QcrC in C. jejuni, and these levels were closely related not only to the energy metabolism of C. jejuni but also its pathogenicity. Furthermore, immunization of mice with recombinant QcrC induced protective immunity against C. jejuni infection. Discussion Taken together, our present findings highlight a possible antibody- or vaccination-based strategy to prevent or control Campylobacter infection by targeting the QcrC-mediated metabolic pathway.
Collapse
Affiliation(s)
- Koji Hosomi
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Noritoshi Hatanaka
- Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
- Asian Health Science Research Institute, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Atsushi Hinenoya
- Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
- Asian Health Science Research Institute, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, NIBIOHN, Osaka, Japan
| | - Yoko Tojima
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Mari Furuta
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Keita Uchiyama
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Makiko Morita
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Functional Anatomy, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Azusa Saika
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Soichiro Kawai
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Ken Yoshii
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Saki Kondo
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Shinji Yamasaki
- Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
- Asian Health Science Research Institute, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Graduate School of Dentistry, Osaka University, Osaka, Japan
- Graduate School of Science, Osaka University, Osaka, Japan
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| |
Collapse
|
5
|
Smith CB, Gao A, Bravo P, Alam A. Microbial Metabolite Trimethylamine N-Oxide Promotes Campylobacter jejuni Infection by Escalating Intestinal Inflammation, Epithelial Damage, and Barrier Disruption. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588895. [PMID: 38645062 PMCID: PMC11030326 DOI: 10.1101/2024.04.10.588895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The interactions between Campylobacter jejuni , a critical foodborne cause of gastroenteritis, and the intestinal microbiota during infection are not completely understood. The crosstalk between C. jejuni and its host is impacted by the gut microbiota through mechanisms of competitive exclusion, microbial metabolites, or immune response. To investigate the role of gut microbiota on C. jejuni pathogenesis, we examined campylobacteriosis in the IL10KO mouse model, which was characterized by an increase in the relative abundance of intestinal proteobacteria, E. coli , and inflammatory cytokines during C. jejuni infection. We also found a significantly increased abundance of microbial metabolite Trimethylamine N-Oxide (TMAO) in the colonic lumens of IL10KO mice. We further investigated the effects of TMAO on C. jejuni pathogenesis. We determined that C. jejuni senses TMAO as a chemoattractant and the administration of TMAO promotes C. jejuni invasion into Caco-2 monolayers. TMAO also increased the transmigration of C. jejuni across polarized monolayers of Caco-2 cells, decreased TEER, and increased C. jejuni -mediated intestinal barrier damage. Interestingly, TMAO treatment and presence during C. jejuni infection of Caco-2 cells synergistically caused an increased inflammatory cytokine expression, specifically IL-1β and IL-8. These results establish that C. jejuni utilizes microbial metabolite TMAO for increased virulence during infection.
Collapse
|
6
|
Gong X, Ma Y, Deng X, Li A, Li X, Kong X, Liu Y, Liu X, Guo K, Yang Y, Li Z, Wei H, Zhou D, Hong Z. Intestinal dysbiosis exacerbates susceptibility to the anti-NMDA receptor encephalitis-like phenotype by changing blood brain barrier permeability and immune homeostasis. Brain Behav Immun 2024; 116:34-51. [PMID: 38030048 DOI: 10.1016/j.bbi.2023.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023] Open
Abstract
Changes in the intestinal microbiota have been observed in patients with anti-N-methyl-D-aspartate receptor encephalitis (NMDARE). However, whether and how the intestinal microbiota is involved in the pathogenesis of NMDARE susceptibility needs to be demonstrated. Here, we first showed that germ-free (GF) mice that underwent fecal microbiota transplantation (FMT) from NMDARE patients, whose fecal microbiota exhibited low short-chain fatty acid content, decreased abundance of Lachnospiraceae, and increased abundance of Verrucomicrobiota, Akkermansia, Parabacteroides, Oscillospirales, showed significant behavioral deficits. Then, these FMT mice were actively immunized with an amino terminal domain peptide from the GluN1 subunit (GluN1356-385) to mimic the pathogenic process of NMDARE. We found that FMT mice showed an increased susceptibility to an encephalitis-like phenotype characterized by more clinical symptoms, greater pentazole (PTZ)-induced susceptibility to seizures, and higher levels of T2 weighted image (T2WI) hyperintensities following immunization. Furthermore, mice with dysbiotic microbiota had impaired blood-brain barrier integrity and a proinflammatory condition. In NMDARE-microbiota recipient mice, the levels of Evan's blue (EB) dye extravasation increased, ZO-1 and claudin-5 expression decreased, and the levels of proinflammatory cytokines (IL-1, IL-6, IL-17, TNF-α and LPS) increased. Finally, significant brain inflammation, mainly in hippocampal and cortical regions, with modest neuroinflammation, immune cell infiltration, and reduced expression of NMDA receptors were observed in NMDARE microbiota recipient mice following immunization. Overall, our findings demonstrated that intestinal dysbiosis increased NMDARE susceptibility, suggesting a new target for limiting the occurrence of the severe phenotype of NMDARE.
Collapse
Affiliation(s)
- Xue Gong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yaru Ma
- Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaolin Deng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Aiqing Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xingjie Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xueying Kong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yue Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xu Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Kundian Guo
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuting Yang
- Precision Medicine Institute, The First Affiliated Hospital Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhongxin Li
- Precision Medicine Institute, The First Affiliated Hospital Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Hong Wei
- Precision Medicine Institute, The First Affiliated Hospital Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhen Hong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan 611730, China; National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Sinha R, LeVeque RM, Callahan SM, Chatterjee S, Stopnisek N, Kuipel M, Johnson JG, DiRita VJ. Gut metabolite L-lactate supports Campylobacter jejuni population expansion during acute infection. Proc Natl Acad Sci U S A 2024; 121:e2316540120. [PMID: 38170751 PMCID: PMC10786315 DOI: 10.1073/pnas.2316540120] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using 6-wk-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni, ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within 2 to 3 d of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP, which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter (lctP) led to identification of a putative thiol-based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides better insights into the pathogenicity of C. jejuni.
Collapse
Affiliation(s)
- Ritam Sinha
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| | - Rhiannon M. LeVeque
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| | - Sean M. Callahan
- Department of Microbiology, University of Tennessee, Knoxville, TN37996
| | - Shramana Chatterjee
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| | - Nejc Stopnisek
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| | - Matti Kuipel
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI48824
| | | | - Victor J. DiRita
- Department of Microbiology, Genetics, & Immunology, Michigan State University, East Lansing, MI48824
| |
Collapse
|
8
|
Calvigioni M, Mazzantini D, Celandroni F, Ghelardi E. Animal and In Vitro Models as Powerful Tools to Decipher the Effects of Enteric Pathogens on the Human Gut Microbiota. Microorganisms 2023; 12:67. [PMID: 38257894 PMCID: PMC10818369 DOI: 10.3390/microorganisms12010067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Examining the interplay between intestinal pathogens and the gut microbiota is crucial to fully comprehend the pathogenic role of enteropathogens and their broader impact on human health. Valid alternatives to human studies have been introduced in laboratory practice to evaluate the effects of infectious agents on the gut microbiota, thereby exploring their translational implications in intestinal functionality and overall health. Different animal species are currently used as valuable models for intestinal infections. In addition, considering the recent advances in bioengineering, futuristic in vitro models resembling the intestinal environment are also available for this purpose. In this review, the impact of the main human enteropathogens (i.e., Clostridioides difficile, Campylobacter jejuni, diarrheagenic Escherichia coli, non-typhoidal Salmonella enterica, Shigella flexneri and Shigella sonnei, Vibrio cholerae, and Bacillus cereus) on intestinal microbial communities is summarized, with specific emphasis on results derived from investigations employing animal and in vitro models.
Collapse
Affiliation(s)
| | | | | | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56127 Pisa, Italy; (M.C.)
| |
Collapse
|
9
|
Sinha R, LeVeque RM, Callahan SM, Chatterjee S, Stopnisek N, Kuipel M, Johnson JG, DiRita VJ. Gut metabolite L-lactate supports Campylobacter jejuni population expansion during acute infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560557. [PMID: 37873437 PMCID: PMC10592923 DOI: 10.1101/2023.10.02.560557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using six-week-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni , ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within two-three days of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP , which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter ( lctP ) led to discovery of a putative thiol based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides new insights into the pathogenicity of C. jejuni . Significance There is a gap in knowledge about the mechanisms by which C. jejuni populations expand during infection. Using an animal model which accurately reflects human infection without the need to alter the host microbiome or the immune system prior to infection, we explored pathophysiological alterations of the gut after C. jejuni infection. Our study identified the gut metabolite L-lactate as playing an important role as a growth substrate for C. jejuni during acute infection. We identified a DNA binding protein, LctR, that binds to the lctP promoter and may repress lctP expression, resulting in decreased lactate transport under low oxygen levels. This work provides new insights about C. jejuni pathogenicity.
Collapse
|
10
|
Molecular Targets in Campylobacter Infections. Biomolecules 2023; 13:biom13030409. [PMID: 36979344 PMCID: PMC10046527 DOI: 10.3390/biom13030409] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Human campylobacteriosis results from foodborne infections with Campylobacter bacteria such as Campylobacter jejuni and Campylobacter coli, and represents a leading cause of bacterial gastroenteritis worldwide. After consumption of contaminated poultry meat, constituting the major source of pathogenic transfer to humans, infected patients develop abdominal pain and diarrhea. Post-infectious disorders following acute enteritis may occur and affect the nervous system, the joints or the intestines. Immunocompromising comorbidities in infected patients favor bacteremia, leading to vascular inflammation and septicemia. Prevention of human infection is achieved by hygiene measures focusing on the reduction of pathogenic food contamination. Molecular targets for the treatment and prevention of campylobacteriosis include bacterial pathogenicity and virulence factors involved in motility, adhesion, invasion, oxygen detoxification, acid resistance and biofilm formation. This repertoire of intervention measures has recently been completed by drugs dampening the pro-inflammatory immune responses induced by the Campylobacter endotoxin lipo-oligosaccharide. Novel pharmaceutical strategies will combine anti-pathogenic and anti-inflammatory effects to reduce the risk of both anti-microbial resistance and post-infectious sequelae of acute enteritis. Novel strategies and actual trends in the combat of Campylobacter infections are presented in this review, alongside molecular targets applied for prevention and treatment strategies.
Collapse
|
11
|
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
|
12
|
Brudvig JM, Cluett MM, Gensterblum-Miller EU, Chen J, Bell JA, Mansfield LS. Th1/Th17-mediated Immunity and Protection from Peripheral Neuropathy in Wildtype and IL10 -/- BALB/c Mice Infected with a Guillain-Barré Syndrome-associated Campylobacter jejuni Strain. Comp Med 2022; 72:63-77. [PMID: 35272743 PMCID: PMC9084571 DOI: 10.30802/aalas-cm-21-000059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/07/2021] [Accepted: 09/03/2021] [Indexed: 11/05/2022]
Abstract
Campylobacter jejuni is an important cause of bacterial gastroenteritis worldwide and is linked to Guillain-Barré syndrome (GBS), a debilitating postinfectious polyneuropathy. The immunopathogenesis of GBS involves the generation of antibodies that are cross reactive to C. jejuni lipooligosaccharide and structurally similar peripheral nerve gangliosides. Both the C. jejuni infecting strain and host factors contribute to GBS development. GBS pathogenesis is associated with Th2-mediated responses in patients. Moreover, induction of IgG1 antiganglioside antibodies in association with colonic Th2-mediated immune responses has been reported in C. jejuni-infected C57BL/6 IL10-/- mice at 4 to 6 wk after infection. We hypothesized that, due to their Th2 immunologic bias, BALB/c mice would develop autoantibodies and signs of peripheral neuropathy after infection with a GBS patient-derived strain of C. jejuni (strain 260.94). WT and IL10-/- BALB/c mice were orally inoculated with C. jejuni 260.94, phenotyped weekly for neurologic deficits, and euthanized after 5 wk. Immune responses were assessed as C. jejuni-specific and antiganglioside antibodies in plasma and cytokine production and histologic lesions in the proximal colon. Peripheral nerve lesions were assessed in dorsal root ganglia and their afferent nerve fibers by scoring immunohistochemically labeled macrophages through morphometry. C. jejuni 260.94 stably colonized both WT and IL10-/- mice and induced systemic Th1/Th17-mediated immune responses with significant increases in C. jejuni-specific IgG2a, IgG2b, and IgG3 plasma antibodies. However, C. jejuni 260.94 did not induce IgG1 antiganglioside antibodies, colitis, or neurologic deficits or peripheral nerve lesions in WT or IL10-/- mice. Both WT and IL10-/- BALB/c mice showed relative protection from development of Th2-mediated immunity and antiganglioside antibodies as compared with C57BL/6 IL10-/- mice. Therefore, BALB/c mice infected with C. jejuni 260.94 are not an effective disease model but provide the opportunity to study the role of immune mechanisms and host genetic background in the susceptibility to post infectious GBS.
Collapse
Affiliation(s)
- Jean M Brudvig
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Matthew M Cluett
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Elizabeth U Gensterblum-Miller
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michiga
| | - James Chen
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michiga
| | - Julia A Bell
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michiga
| | - Linda S Mansfield
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michigan; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan;,
| |
Collapse
|
13
|
Malik A, Brudvig JM, Gadsden BJ, Ethridge AD, Mansfield LS. Campylobacter jejuni induces autoimmune peripheral neuropathy via Sialoadhesin and Interleukin-4 axes. Gut Microbes 2022; 14:2064706. [PMID: 35442154 PMCID: PMC9037470 DOI: 10.1080/19490976.2022.2064706] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Campylobacter jejuni is a leading cause of gastroenteritis that has been causally linked with development of the autoimmune peripheral neuropathy Guillain Barré Syndrome (GBS). Previously, we showed that C. jejuni isolates from human enteritis patients induced Type1/17-cytokine dependent colitis in interleukin-10 (IL-10)-/- mice, while isolates from GBS patients colonized these mice without colitis but instead induced autoantibodies that cross-reacted with the sialylated oligosaccharide motifs on the LOS of GBS-associated C. jejuni and the peripheral nerve gangliosides. We show here that infection of IL-10-/- mice with the GBS but not the colitis isolate led to sciatic nerve inflammation and abnormal gait and hind limb movements, with character and timing consistent with this syndrome in humans. Autoantibody responses and associated nerve histologic changes were dependent on IL-4 production by CD4 T cells. We further show that Siglec-1 served as a central antigen presenting cell receptor mediating the uptake of the GBS isolates via interaction with the sialylated oligosaccharide motifs found specifically on the LOS of GBS-associated C. jejuni, and the ensuing T cell differentiation and autoantibody elicitation. Sialylated oligosaccharide motifs on the LOS of GBS-associated C. jejuni therefore acted as both the Siglec-1-ligand for phagocytosis, as well as the epitope for autoimmunity. Overall, we present a mouse model of an autoimmune disease induced directly by a bacterium that is dependent upon Siglec-1 and IL-4. We also demonstrate the negative regulatory role of IL-10 in C. jejuni induced autoimmunity and provide IL-4 and Siglec-1 blockade as potential therapeutic interventions against GBS.
Collapse
Affiliation(s)
- Ankit Malik
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Jean M. Brudvig
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, MIUSA
| | - Barbie J. Gadsden
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, MIUSA
| | - Alexander D. Ethridge
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Linda S. Mansfield
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
14
|
Korchagina AA, Koroleva E, Tumanov AV. Innate Lymphoid Cells in Response to Intracellular Pathogens: Protection Versus Immunopathology. Front Cell Infect Microbiol 2021; 11:775554. [PMID: 34938670 PMCID: PMC8685334 DOI: 10.3389/fcimb.2021.775554] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/03/2021] [Indexed: 12/23/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous group of cytokine-producing lymphocytes which are predominantly located at mucosal barrier surfaces, such as skin, lungs, and gastrointestinal tract. ILCs contribute to tissue homeostasis, regulate microbiota-derived signals, and protect against mucosal pathogens. ILCs are classified into five major groups by their developmental origin and distinct cytokine production. A recently emerged intriguing feature of ILCs is their ability to alter their phenotype and function in response to changing local environmental cues such as pathogen invasion. Once the pathogen crosses host barriers, ILCs quickly activate cytokine production to limit the spread of the pathogen. However, the dysregulated ILC responses can lead to tissue inflammation and damage. Furthermore, the interplay between ILCs and other immune cell types shapes the outcome of the immune response. Recent studies highlighted the important role of ILCs for host defense against intracellular pathogens. Here, we review recent advances in understanding the mechanisms controlling protective and pathogenic ILC responses to intracellular pathogens. This knowledge can help develop new ILC-targeted strategies to control infectious diseases and immunopathology.
Collapse
Affiliation(s)
- Anna A Korchagina
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ekaterina Koroleva
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
15
|
Wymore Brand M, Sahin O, Hostetter JM, Trachsel J, Zhang Q, Wannemuehler MJ. Campylobacter jejuni persistently colonizes gnotobiotic altered Schaedler flora C3H/HeN mice and induces mild colitis. FEMS Microbiol Lett 2021; 367:5937419. [PMID: 33098301 DOI: 10.1093/femsle/fnaa163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 10/05/2020] [Indexed: 11/13/2022] Open
Abstract
Campylobacter jejuni is a major cause of food-borne human bacterial gastroenteritis but animal models for C. jejuni mediated disease remain limited because C. jejuni poorly colonizes immunocompetent, conventionally-reared (Conv-R) mice. Thus, a reliable rodent model (i.e. persistent colonization) is desirable in order to evaluate C. jejuni-mediated gastrointestinal disease and mechanisms of pathogenicity. As the nature and complexity of the microbiota likely impacts colonization resistance for C. jejuni, Conv-R and gnotobiotic C3H/HeN mice were used to evaluate the persistence of C. jejuni colonization and development of disease. A total of four C. jejuni isolates readily and persistently colonized ASF mice and induced mild mucosal inflammation in the proximal colon, but C. jejuni did not stably colonize nor induce lesions in Conv-R mice. This suggests that the pathogenesis of C. jejuni is influenced by the microbiota, and that ASF mice offer a reproducible model to study the influence of the microbiota on the ability of C. jejuni to colonize the gut and to mediate gastroenteritis.
Collapse
Affiliation(s)
- Meghan Wymore Brand
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| | - Orhan Sahin
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| | - Jesse M Hostetter
- Department of Veterinary Pathology, College of Veterinary Medicine, University of Georgia, 501 D. W. Brooks Drive, Athens, GA 30602, USA
| | - Julian Trachsel
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| | - Qijing Zhang
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, 1800 Christensen Drive, Ames, IA 50011, USA
| |
Collapse
|
16
|
Muraoka WT, Korchagina AA, Xia Q, Shein SA, Jing X, Lai Z, Weldon K, Wang LJ, Chen Y, Kummer LW, Mohrs M, Vivier E, Koroleva EP, Tumanov AV. Campylobacter infection promotes IFNγ-dependent intestinal pathology via ILC3 to ILC1 conversion. Mucosal Immunol 2021; 14:703-716. [PMID: 33214656 PMCID: PMC8084871 DOI: 10.1038/s41385-020-00353-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 08/12/2020] [Accepted: 10/16/2020] [Indexed: 02/04/2023]
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous family of immune regulators that protect against mucosal pathogens but can also promote intestinal pathology. Although the plasticity between ILCs populations has been described, the role of mucosal pathogens in inducing ILC conversion leading to intestinal pathology remains unclear. Here we demonstrate that IFNγ-producing ILCs are responsible for promoting intestinal pathology in a mouse model of enterocolitis caused by Campylobacter jejuni, a common human enteric pathogen. Phenotypic analysis revealed a distinct population of IFNγ-producing NK1.1-T-bet+ILCs that accumulated in the intestine of C. jejuni-infected mice. Adoptive transfer experiments demonstrated their capacity to promote intestinal pathology. Inactivation of T-bet in NKp46+ ILCs ameliorated disease. Transcriptome analysis and cell-fate mapping experiments revealed that IFNγ-producing NK1.1-ILCs correspond to ILC1 profile and develop from RORγt+ progenitors. Collectively, we identified a distinct population of NK1.1-ex-ILC3s that promotes intestinal pathology through IFNγ production in response to C. jejuni infection.
Collapse
Affiliation(s)
- Wayne T. Muraoka
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA,Trudeau Institute, Saranac Lake, NY, USA,Contributed equally,Current address: US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA
| | - Anna A. Korchagina
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA,Contributed equally
| | - Qingqing Xia
- Trudeau Institute, Saranac Lake, NY, USA,Current address: Brooke Army Medical Center, Ft. Sam Houston, TX, USA
| | - Sergey A. Shein
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Xi Jing
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, USA
| | - Korri Weldon
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, USA
| | - Li-Ju Wang
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, USA
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, USA
| | | | - Markus Mohrs
- Trudeau Institute, Saranac Lake, NY, USA,Current address: Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Eric Vivier
- Innate Pharma and Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Ekaterina P. Koroleva
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Alexei V. Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX, USA,Correspondence: Alexei Tumanov (), 7703 Floyd Curl Dr. San Antonio, TX 78229, (210) 450-8157
| |
Collapse
|
17
|
Peroral Clove Essential Oil Treatment Ameliorates Acute Campylobacteriosis-Results from a Preclinical Murine Intervention Study. Microorganisms 2021; 9:microorganisms9040735. [PMID: 33807493 PMCID: PMC8066448 DOI: 10.3390/microorganisms9040735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/23/2022] Open
Abstract
Campylobacter (C.) jejuni infections pose progressively emerging threats to human health worldwide. Given the rise in antibiotic resistance, antibiotics-independent options are required to fight campylobacteriosis. Since the health-beneficial effects of clove have been known for long, we here analyzed the antimicrobial and immune-modulatory effects of clove essential oil (EO) during acute experimental campylobacteriosis. Therefore, microbiota-depleted interleukin-10 deficient (IL-10-/-) mice were perorally infected with C. jejuni and treated with clove EO via drinking water starting on day 2 post-infection. On day 6 post-infection, lower small- and large-intestinal pathogen loads could be assessed in clove EO as compared to placebo treated mice. Although placebo mice suffered from severe campylobacteriosis as indicated by wasting and bloody diarrhea, clove EO treatment resulted in a better clinical outcome and in less severe colonic histopathological and apoptotic cell responses in C. jejuni infected mice. Furthermore, lower colonic numbers of macrophages, monocytes, and T lymphocytes were detected in mice from the verum versus the placebo cohort that were accompanied by lower intestinal, extra-intestinal, and even systemic proinflammatory cytokine concentrations. In conclusion, our preclinical intervention study provides first evidence that the natural compound clove EO constitutes a promising antibiotics-independent treatment option of acute campylobacteriosis in humans.
Collapse
|
18
|
Heimesaat MM, Backert S, Alter T, Bereswill S. Human Campylobacteriosis-A Serious Infectious Threat in a One Health Perspective. Curr Top Microbiol Immunol 2021; 431:1-23. [PMID: 33620646 DOI: 10.1007/978-3-030-65481-8_1] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Zoonotic Campylobacter species-mainly C. jejuni and C. coli-are major causes of food-borne bacterial infectious gastroenteritis worldwide. Symptoms of intestinal campylobacteriosis include abdominal pain, diarrhea and fever. The clinical course of enteritis is generally self-limiting, but some infected individuals develop severe post-infectious sequelae including autoimmune disorders affecting the nervous system, the joints and the intestinal tract. Moreover, in immunocompromised individuals, systemic spread of the pathogens may trigger diseases of the circulatory system and septicemia. The socioeconomic costs associated with Campylobacter infections have been calculated to several billion dollars annually. Poultry meat products represent major sources of human infections. Thus, a "One World-One Health" approach with collective efforts of public health authorities, veterinarians, clinicians, researchers and politicians is required to reduce the burden of campylobacteriosis. Innovative intervention regimes for the prevention of Campylobacter contaminations along the food chain include improvements of information distribution to strengthen hygiene measures for agricultural remediation. Given that elimination of Campylobacter from the food production chains is not feasible, novel intervention strategies fortify both the reduction of pathogen contamination in food production and the treatment of the associated diseases in humans. This review summarizes some current trends in the combat of Campylobacter infections including the combination of public health and veterinary preventive approaches with consumer education. The "One World-One Health" perspective is completed by clinical aspects and molecular concepts of human campylobacteriosis offering innovative treatment options supported by novel murine infection models that are based on the essential role of innate immune activation by bacterial endotoxins.
Collapse
Affiliation(s)
- Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité-University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Steffen Backert
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg, Erlangen, Germany
| | - Thomas Alter
- Department of Veterinary Medicine, Institute of Food Safety and Food Hygiene, Free University Berlin, Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité-University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
19
|
Jing X, Korchagina AA, Shein SA, Muraoka WT, Koroleva E, Tumanov AV. IL-23 Contributes to Campylobacter jejuni-Induced Intestinal Pathology via Promoting IL-17 and IFNγ Responses by Innate Lymphoid Cells. Front Immunol 2021; 11:579615. [PMID: 33488580 PMCID: PMC7815532 DOI: 10.3389/fimmu.2020.579615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
Human pathogen Campylobacter jejuni is a significant risk factor for the development of long-term intestinal dysfunction although the cellular and molecular mechanisms remain scantily defined. IL-23 is an emerging therapeutic target for the treatment of inflammatory intestinal diseases, however its role in C. jejuni-driven intestinal pathology is not fully understood. IL-10 deficient mice represent a robust model to study the pathogenesis of C. jejuni infection because C. jejuni infection of mice lacking IL-10 results in symptoms and pathology that resemble human campylobacteriosis. To determine the role of IL-23 in C. jejuni-driven intestinal inflammation, we studied the disease pathogenesis in IL-23-/- mice with inhibited IL-10Rα signaling. These mice exhibited reduced intestinal pathology independent from bacterial clearance. Further, levels of IFNγ, IL-17, IL-22, TNF, and IL-6 were reduced and associated with reduced accumulation of neutrophils, monocytes and macrophages in the colon. Flow cytometry analysis revealed reduced production of IL-17 and IFNγ by group 1 and 3 innate lymphoid cells. Thus, our data suggest that IL-23 contributes to intestinal inflammation in C. jejuni infected mice by promoting IL-17 and IFNγ production by innate lymphoid cells.
Collapse
Affiliation(s)
- Xi Jing
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Anna A Korchagina
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Sergey A Shein
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Wayne T Muraoka
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Trudeau Institute, Saranac Lake, NY, United States
| | - Ekaterina Koroleva
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
20
|
Mousavi S, Bereswill S, Heimesaat MM. Murine Models for the Investigation of Colonization Resistance and Innate Immune Responses in Campylobacter Jejuni Infections. Curr Top Microbiol Immunol 2021; 431:233-263. [PMID: 33620654 DOI: 10.1007/978-3-030-65481-8_9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Human infections with the food-borne pathogen Campylobacter jejuni are progressively increasing worldwide and constitute a significant socioeconomic burden to mankind. Intestinal campylobacteriosis in humans is characterized by bloody diarrhea, fever, abdominal pain, and severe malaise. Some individuals develop chronic post-infectious sequelae including neurological and autoimmune diseases such as reactive arthritis and Guillain-Barré syndrome. Studies unraveling the molecular mechanisms underlying campylobacteriosis and post-infectious sequelae have been hampered by the scarcity of appropriate experimental in vivo models. Particularly, conventional laboratory mice are protected from C. jejuni infection due to the physiological colonization resistance exerted by the murine gut microbiota composition. Additionally, as compared to humans, mice are up to 10,000 times more resistant to C. jejuni lipooligosaccharide (LOS) constituting a major pathogenicity factor responsible for the immunopathological host responses during campylobacteriosis. In this chapter, we summarize the recent progress that has been made in overcoming these fundamental obstacles in Campylobacter research in mice. Modification of the murine host-specific gut microbiota composition and sensitization of the mice to C. jejuni LOS by deletion of genes encoding interleukin-10 or a single IL-1 receptor-related molecule as well as by dietary zinc depletion have yielded reliable murine infection models resembling key features of human campylobacteriosis. These substantial improvements pave the way for a better understanding of the molecular mechanisms underlying pathogen-host interactions. The ongoing validation and standardization of these novel murine infection models will provide the basis for the development of innovative treatment and prevention strategies to combat human campylobacteriosis and collateral damages of C. jejuni infections.
Collapse
Affiliation(s)
- Soraya Mousavi
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University of Berlin, Berlin Institute of Health, Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University of Berlin, Berlin Institute of Health, Berlin, Germany
| | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University of Berlin, Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
21
|
Preclinical Evaluation of Oral Urolithin-A for the Treatment of Acute Campylobacteriosis in Campylobacter jejuni Infected Microbiota-Depleted IL-10 -/- Mice. Pathogens 2020; 10:pathogens10010007. [PMID: 33374868 PMCID: PMC7823290 DOI: 10.3390/pathogens10010007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022] Open
Abstract
Human campylobacteriosis represents an infectious enteritis syndrome caused by Campylobacter species, mostly Campylobacter jejuni. Given that C. jejuni infections are rising worldwide and antibiotic treatment is usually not indicated, novel treatment options for campylobacteriosis are needed. Urolithin-A constitutes a metabolite produced by the human gut microbiota from ellagitannins and ellagic acids in berries and nuts which have been known for their health-beneficial including anti-inflammatory effects since centuries. Therefore, we investigated potential pathogen-lowering and immunomodulatory effects following oral application of synthetic urolithin-A during acute campylobacteriosis applying perorally C. jejuni infected, microbiota-depleted IL-10-/- mice as preclinical inflammation model. On day 6 post infection, urolithin-A treated mice harbored slightly lower pathogen loads in their ileum, but not colon as compared to placebo counterparts. Importantly, urolithin-A treatment resulted in an improved clinical outcome and less pronounced macroscopic and microscopic inflammatory sequelae of infection that were paralleled by less pronounced intestinal pro-inflammatory immune responses which could even be observed systemically. In conclusion, this preclinical murine intervention study provides first evidence that oral urolithin-A application is a promising treatment option for acute C. jejuni infection and paves the way for future clinical studies in human campylobacteriosis.
Collapse
|
22
|
Fu Y, Almansour A, Bansal M, Alenezi T, Alrubaye B, Wang H, Sun X. Microbiota attenuates chicken transmission-exacerbated campylobacteriosis in Il10 -/- mice. Sci Rep 2020; 10:20841. [PMID: 33257743 PMCID: PMC7705718 DOI: 10.1038/s41598-020-77789-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/17/2020] [Indexed: 11/08/2022] Open
Abstract
Campylobacter jejuni is a prevalent foodborne pathogen mainly transmitting through poultry. It remains unknown how chicken-transmitted C. jejuni and microbiota impact on human campylobacteriosis. Campylobacter jejuni AR101 (Cj-P0) was introduced to chickens and isolated as passage 1 (Cj-P1). Campylobacter jejuni Cj-P1-DCA-Anaero was isolated from Cj-P0-infected birds transplanted with DCA-modulated anaerobic microbiota. Specific pathogen free Il10-/- mice were gavaged with antibiotic clindamycin and then infected with Cj-P0, Cj-P1, or Cj-P1-DCA-Anaero, respectively. After 8 days post infection, Il10-/- mice infected with Cj-P1 demonstrated severe morbidity and bloody diarrhea and the experiment had to be terminated. Cj-P1 induced more severe histopathology compared to Cj-P0, suggesting that chicken transmission increased C. jejuni virulence. Importantly, mice infected with Cj-P1-DCA-Anaero showed attenuation of intestinal inflammation compared to Cj-P1. At the cellular level, Cj-P1 induced more C. jejuni invasion and neutrophil infiltration into the Il10-/- mouse colon tissue compared to Cj-P0, which was attenuated with Cj-P1-DCA-Anaero. At the molecular level, Cj-P1 induced elevated inflammatory mediator mRNA accumulation of Il17a, Il1β, and Cxcl1 in the colon compared to Cj-P0, while Cj-P1-DCA-Anaero showed reduction of the inflammatory gene expression. In conclusion, our data suggest that DCA-modulated anaerobes attenuate chicken-transmitted campylobacteriosis in mice and it is important to control the elevation of C. jejuni virulence during chicken transmission process.
Collapse
Affiliation(s)
- Ying Fu
- CEMB, University of Arkansas, Fayetteville, AR, 72701, USA
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W Maple St. O-409, Fayetteville, AR, 72701, USA
| | - Ayidh Almansour
- CEMB, University of Arkansas, Fayetteville, AR, 72701, USA
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W Maple St. O-409, Fayetteville, AR, 72701, USA
| | - Mohit Bansal
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W Maple St. O-409, Fayetteville, AR, 72701, USA
| | - Tahrir Alenezi
- CEMB, University of Arkansas, Fayetteville, AR, 72701, USA
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W Maple St. O-409, Fayetteville, AR, 72701, USA
| | - Bilal Alrubaye
- CEMB, University of Arkansas, Fayetteville, AR, 72701, USA
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W Maple St. O-409, Fayetteville, AR, 72701, USA
| | - Hong Wang
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W Maple St. O-409, Fayetteville, AR, 72701, USA
| | - Xiaolun Sun
- CEMB, University of Arkansas, Fayetteville, AR, 72701, USA.
- Center of Excellence for Poultry Science, University of Arkansas, 1260 W Maple St. O-409, Fayetteville, AR, 72701, USA.
| |
Collapse
|
23
|
Sher AA, Jerome JP, Bell JA, Yu J, Kim HY, Barrick JE, Mansfield LS. Experimental Evolution of Campylobacter jejuni Leads to Loss of Motility, rpoN (σ54) Deletion and Genome Reduction. Front Microbiol 2020; 11:579989. [PMID: 33240235 PMCID: PMC7677240 DOI: 10.3389/fmicb.2020.579989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/14/2020] [Indexed: 12/18/2022] Open
Abstract
Evolution experiments in the laboratory have focused heavily on model organisms, often to the exclusion of clinically relevant pathogens. The foodborne bacterial pathogen Campylobacter jejuni belongs to a genus whose genomes are small compared to those of its closest genomic relative, the free-living genus Sulfurospirillum, suggesting genome reduction during the course of evolution to host association. In an in vitro experiment, C. jejuni serially passaged in rich medium in the laboratory exhibited loss of flagellar motility-an essential function for host colonization. At early time points the motility defect was often reversible, but after 35 days of serial culture, motility was irreversibly lost in most cells in 5 independently evolved populations. Population re-sequencing revealed disruptive mutations to genes in the flagellar transcriptional cascade, rpoN (σ54)-therefore disrupting the expression of the genes σ54 regulates-coupled with deletion of rpoN in all evolved lines. Additional mutations were detected in virulence-related loci. In separate in vivo experiments, we demonstrate that a phase variable (reversible) motility mutant carrying an adenine deletion within a homopolymeric tract resulting in truncation of the flagellar biosynthesis gene fliR was deficient for colonization in a C57BL/6 IL-10-/- mouse disease model. Re-insertion of an adenine residue partially restored motility and ability to colonize mice. Thus, a pathogenic C. jejuni strain was rapidly attenuated by experimental laboratory evolution and demonstrated genomic instability during this evolutionary process. The changes observed suggest C. jejuni is able to evolve in a novel environment through genome reduction as well as transition, transversion, and slip-strand mutations.
Collapse
Affiliation(s)
- Azam A. Sher
- Comparative Enteric Diseases Laboratory, East Lansing, MI, United States
- Comparative Medicine and Integrative Biology, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
- BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI, United States
| | - John P. Jerome
- Comparative Enteric Diseases Laboratory, East Lansing, MI, United States
- BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Julia A. Bell
- Comparative Enteric Diseases Laboratory, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Julian Yu
- Comparative Enteric Diseases Laboratory, East Lansing, MI, United States
- BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Hahyung Y. Kim
- Comparative Enteric Diseases Laboratory, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Jeffrey E. Barrick
- BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI, United States
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States
| | - Linda S. Mansfield
- Comparative Enteric Diseases Laboratory, East Lansing, MI, United States
- BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
24
|
The Host-Specific Intestinal Microbiota Composition Impacts Campylobacter coli Infection in a Clinical Mouse Model of Campylobacteriosis. Pathogens 2020; 9:pathogens9100804. [PMID: 33003421 PMCID: PMC7600086 DOI: 10.3390/pathogens9100804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/04/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
Human Campylobacter-infections are progressively rising globally. However, the molecular mechanisms underlying C. coli–host interactions are incompletely understood. In this study, we surveyed the impact of the host-specific intestinal microbiota composition during peroral C. coli infection applying an established murine campylobacteriosis model. Therefore, microbiota-depleted IL-10−/− mice were subjected to peroral fecal microbiota transplantation from murine versus human donors and infected with C. coli one week later by gavage. Irrespective of the microbiota, C. coli stably colonized the murine gastrointestinal tract until day 21 post-infection. Throughout the survey, C. coli-infected mice with a human intestinal microbiota displayed more frequently fecal blood as their murine counterparts. Intestinal inflammatory sequelae of C. coli-infection could exclusively be observed in mice with a human intestinal microbiota, as indicated by increased colonic numbers of apoptotic epithelial cells and innate as well as adaptive immune cell subsets, which were accompanied by more pronounced pro-inflammatory cytokine secretion in the colon and mesenteric lymph nodes versus mock controls. However, in extra-intestinal, including systemic compartments, pro-inflammatory responses upon pathogen challenge could be assessed in mice with either microbiota. In conclusion, the host-specific intestinal microbiota composition has a profound effect on intestinal and systemic pro-inflammatory immune responses during C. coli infection.
Collapse
|
25
|
Perruzza L, Jaconi S, Lombardo G, Pinna D, Strati F, Morone D, Seehusen F, Hu Y, Bajoria S, Xiong J, Kumru OS, Joshi SB, Volkin DB, Piantanida R, Benigni F, Grassi F, Corti D, Pizzuto MS. Prophylactic Activity of Orally Administered FliD-Reactive Monoclonal SIgA Against Campylobacter Infection. Front Immunol 2020; 11:1011. [PMID: 32582158 PMCID: PMC7296071 DOI: 10.3389/fimmu.2020.01011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
Campylobacter infection is one of the most common causes of bacterial gastroenteritis worldwide and a major global health threat due to the rapid development of antibiotic resistance. Currently, there are no vaccines approved to prevent campylobacteriosis, and rehydration is the main form of therapy. Secretory immunoglobulin A (SIgA) is the main antibody class found in mucous secretions, including human milk, and serves as the first line of defense for the gastrointestinal epithelium against enteric pathogens. In this study, we describe the prophylactic activity of orally delivered recombinant SIgA generated from two human monoclonal antibodies (CAA1 and CCG4) isolated for their reactivity against the flagellar-capping protein FliD, which is essential for bacteria motility and highly conserved across Campylobacter species associated with severe enteritis. In an immunocompetent weaned mouse model, a single oral administration of FliD-reactive SIgA CAA1 or CCG4 at 2 h before infection significantly enhances Campylobacter clearance at early stages post-infection, reducing the levels of inflammation markers associated with epithelial damage and polymorphonuclear (PMN) cells infiltration in the cecum lamina propria. Our data indicate that the prophylactic activity of CAA1 and CCG4 is not only dependent on the specificity to FliD but also on the use of the SIgA format, as the immunoglobulin G (IgG) versions of the same antibodies did not confer a comparable protective effect. Our work emphasizes the potential of FliD as a target for the development of vaccines and supports the concept that orally administered FliD-reactive SIgA can be developed to prevent or mitigate the severity of Campylobacter infections as well as the development of post-infection syndromes.
Collapse
Affiliation(s)
- Lisa Perruzza
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Stefano Jaconi
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Gloria Lombardo
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Debora Pinna
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Francesco Strati
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Diego Morone
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Frauke Seehusen
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Yue Hu
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Jian Xiong
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Ozan Selahattin Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Sangeeta Bagai Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - David Bernard Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Renato Piantanida
- Department of Otolaryngology-Head and Neck Surgery, Ospedale Regionale di Lugano, Lugano, Switzerland
| | - Fabio Benigni
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Fabio Grassi
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Davide Corti
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | | |
Collapse
|
26
|
Mousavi S, Bereswill S, Heimesaat MM. Novel Clinical Campylobacter jejuni Infection Models Based on Sensitization of Mice to Lipooligosaccharide, a Major Bacterial Factor Triggering Innate Immune Responses in Human Campylobacteriosis. Microorganisms 2020; 8:E482. [PMID: 32231139 PMCID: PMC7232424 DOI: 10.3390/microorganisms8040482] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022] Open
Abstract
: Human Campylobacter jejuni infections inducing campylobacteriosis including post-infectious sequelae such as Guillain-Barré syndrome and reactive arthritis are rising worldwide and progress into a global burden of high socioeconomic impact. Intestinal immunopathology underlying campylobacteriosis is a classical response of the innate immune system characterized by the accumulation of neutrophils and macrophages which cause tissue destruction, barrier defects and malabsorption leading to bloody diarrhea. Clinical studies revealed that enteritis and post-infectious morbidities of human C. jejuni infections are strongly dependent on the structure of pathogenic lipooligosaccharides (LOS) triggering the innate immune system via Toll-like-receptor (TLR)-4 signaling. Compared to humans, mice display an approximately 10,000 times weaker TLR-4 response and a pronounced colonization resistance (CR) against C. jejuni maintained by the murine gut microbiota. In consequence, investigations of campylobacteriosis have been hampered by the lack of experimental animal models. We here summarize recent progress made in the development of murine C. jejuni infection models that are based on the abolishment of CR by modulating the murine gut microbiota and by sensitization of mice to LOS. These advances support the major role of LOS driven innate immunity in pathogenesis of campylobacteriosis including post-infectious autoimmune diseases and promote the preclinical evaluation of novel pharmaceutical strategies for prophylaxis and treatment.
Collapse
|
27
|
Brooks PT, Bell JA, Bejcek CE, Malik A, Mansfield LS. An antibiotic depleted microbiome drives severe Campylobacter jejuni-mediated Type 1/17 colitis, Type 2 autoimmunity and neurologic sequelae in a mouse model. J Neuroimmunol 2019; 337:577048. [PMID: 31678855 DOI: 10.1016/j.jneuroim.2019.577048] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/01/2019] [Accepted: 09/02/2019] [Indexed: 10/26/2022]
Abstract
The peripheral neuropathy Guillain-Barré Syndrome can follow Campylobacter jejuni infection when outer core lipooligosaccharides induce production of neurotoxic anti-ganglioside antibodies. We hypothesized that gut microbiota depletion with an antibiotic would increase C. jejuni colonization, severity of gastroenteritis, and GBS. Microbiota depletion increased C. jejuni colonization, invasion, and colitis with Type 1/17 T cells in gut lamina propria. It also stimulated Type 1/17 anti-C. jejuni and -antiganglioside-antibodies, Type 2 anti-C. jejuni and -antiganglioside antibodies, and neurologic phenotypes. Results indicate that both C. jejuni strain and gut microbiota affect development of inflammation and GBS and suggest that probiotics following C. jejuni infection may ameliorate inflammation and autoimmune disease.
Collapse
Affiliation(s)
- Phillip T Brooks
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Comparative Medicine Integrative Biology Graduate Program, Michigan State University, East Lansing, MI, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Julia A Bell
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Departments of Microbiology and Molecular Genetics and Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Christopher E Bejcek
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Departments of Microbiology and Molecular Genetics and Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Ankit Malik
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Departments of Microbiology and Molecular Genetics and Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Linda S Mansfield
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA; Departments of Microbiology and Molecular Genetics and Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
28
|
Lou H, Li X, Guo F, Ding M, Hu Y, Chen H, Yan J. Evaluations of Alkyl hydroperoxide reductase B cell antigen epitope as a potential epitope vaccine against Campylobacter jejuni. Saudi J Biol Sci 2019; 26:1117-1122. [PMID: 31516338 PMCID: PMC6734151 DOI: 10.1016/j.sjbs.2019.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/28/2019] [Accepted: 05/28/2019] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE The present study aimed to screen and find alkyl hydroperoxide reductase (AhpC) B cell dominant epitope of Campylobacter jejuni (C. jejuni). MATERIALS AND METHODS Bio-informatic algorithms were used to predict B cell epitopes of AhpC. The AhpC protein and chemically synthesized antigenic epitopes of C. jejuni were considered as antigens, and the AhpC antibody was used as the primary antibody, ELISA and dot blot were used to analyze and screen the dominant epitope. The specific IgG of mice serum and IL-4 in splenocyte culture supernatant were detected by ELISA. The protective efficacy was evaluated by animal disease index and tissue histopathological staining of the jejunum. RESULTS Seven epitopes of AhpC were predicted, one epitope (AhpC4-16) was found to recognize the antibodies of AhpC and had strong antigenicity by ELISA and dot blot analysis. In epitope AhpC4-16 immunized mice, specific IgG of serum and IL-4 in splenocyte culture supernatant were significantly higher. The illness index decreased significantly, the protective rate was 66.67%. Histopathology displayed that the jejunum morphology was better than the control group. CONCLUSIONS These findings suggested that epitope AhpC4-16 showed effective protective role against C. jejuni and is a candidate epitope of vaccine against this pathogen.
Collapse
Affiliation(s)
- Hongqiang Lou
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou 310058, China
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua 321000, China
| | - Xusheng Li
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua 321000, China
| | - Fangming Guo
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua 321000, China
| | - Mingxing Ding
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua 321000, China
| | - Ye Hu
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua 321000, China
| | - Haohao Chen
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua 321000, China
| | - Jie Yan
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
29
|
Burnham PM, Hendrixson DR. Campylobacter jejuni: collective components promoting a successful enteric lifestyle. Nat Rev Microbiol 2019; 16:551-565. [PMID: 29892020 DOI: 10.1038/s41579-018-0037-9] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Campylobacter jejuni is the leading cause of bacterial diarrhoeal disease in many areas of the world. The high incidence of sporadic cases of disease in humans is largely due to its prevalence as a zoonotic agent in animals, both in agriculture and in the wild. Compared with many other enteric bacterial pathogens, C. jejuni has strict growth and nutritional requirements and lacks many virulence and colonization determinants that are typically used by bacterial pathogens to infect hosts. Instead, C. jejuni has a different collection of factors and pathways not typically associated together in enteric pathogens to establish commensalism in many animal hosts and to promote diarrhoeal disease in the human population. In this Review, we discuss the cellular architecture and structure of C. jejuni, intraspecies genotypic variation, the multiple roles of the flagellum, specific nutritional and environmental growth requirements and how these factors contribute to in vivo growth in human and avian hosts, persistent colonization and pathogenesis of diarrhoeal disease.
Collapse
Affiliation(s)
- Peter M Burnham
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David R Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
30
|
Effects of antibiotic resistance (AR) and microbiota shifts on Campylobacter jejuni-mediated diseases. Anim Health Res Rev 2019; 18:99-111. [PMID: 29665882 DOI: 10.1017/s1466252318000014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Campylobacter jejuni is an important zoonotic pathogen recently designated a serious antimicrobial resistant (AR) threat. While most patients with C. jejuni experience hemorrhagic colitis, serious autoimmune conditions can follow including inflammatory bowel disease (IBD) and the acute neuropathy Guillain Barré Syndrome (GBS). This review examines inter-relationships among factors mediating C. jejuni diarrheal versus autoimmune disease especially AR C. jejuni and microbiome shifts. Because both susceptible and AR C. jejuni are acquired from animals or their products, we consider their role in harboring strains. Inter-relationships among factors mediating C. jejuni colonization, diarrheal and autoimmune disease include C. jejuni virulence factors and AR, the enteric microbiome, and host responses. Because AR C. jejuni have been suggested to affect the severity of disease, length of infections and propensity to develop GBS, it is important to understand how these interactions occur when strains are under selection by antimicrobials. More work is needed to elucidate host-pathogen interactions of AR C. jejuni compared with susceptible strains and how AR C. jejuni are maintained and evolve in animal reservoirs and the extent of transmission to humans. These knowledge gaps impair the development of effective strategies to prevent the emergence of AR C. jejuni in reservoir species and human populations.
Collapse
|
31
|
He Z, Gharaibeh RZ, Newsome RC, Pope JL, Dougherty MW, Tomkovich S, Pons B, Mirey G, Vignard J, Hendrixson DR, Jobin C. Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut 2019; 68:289-300. [PMID: 30377189 PMCID: PMC6352414 DOI: 10.1136/gutjnl-2018-317200] [Citation(s) in RCA: 267] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/12/2018] [Accepted: 10/04/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Campylobacter jejuni produces a genotoxin, cytolethal distending toxin (CDT), which has DNAse activity and causes DNA double-strand breaks. Although C. jejuni infection has been shown to promote intestinal inflammation, the impact of this bacterium on carcinogenesis has never been examined. DESIGN Germ-free (GF) ApcMin/+ mice, fed with 1% dextran sulfate sodium, were used to test tumorigenesis potential of CDT-producing C. jejuni. Cells and enteroids were exposed to bacterial lysates to determine DNA damage capacity via γH2AX immunofluorescence, comet assay and cell cycle assay. To examine the interplay of CDT-producing C. jejuni, gut microbiome and host in tumorigenesis, colonic RNA-sequencing and faecal 16S rDNA sequencing were performed. Rapamycin was administrated to investigate the prevention of CDT-producing C. jejuni-induced tumorigenesis. RESULTS GF ApcMin/+ mice colonised with human clinical isolate C. jejuni81-176 developed significantly more and larger tumours when compared with uninfected mice. C. jejuni with a mutated cdtB subunit, mutcdtB, attenuated C. jejuni-induced tumorigenesis in vivo and decreased DNA damage response in cells and enteroids. C. jejuni infection induced expression of hundreds of colonic genes, with 22 genes dependent on the presence of cdtB. The C. jejuni-infected group had a significantly different microbial gene expression profile compared with the mutcdtB group as shown by metatranscriptomic data, and different microbial communities as measured by 16S rDNA sequencing. Finally, rapamycin could diminish the tumorigenic capability of C. jejuni. CONCLUSION Human clinical isolate C. jejuni 81-176 promotes colorectal cancer and induces changes in microbial composition and transcriptomic responses, a process dependent on CDT production.
Collapse
Affiliation(s)
- Zhen He
- Department of Medicine, University of Florida, Gainesville, Florida, USA,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Raad Z Gharaibeh
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Rachel C Newsome
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jllian L Pope
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | | | - Sarah Tomkovich
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Benoit Pons
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, ENVT, INP- Purpan, UPS, Toulouse, France
| | - Gladys Mirey
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, ENVT, INP- Purpan, UPS, Toulouse, France
| | - Julien Vignard
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, ENVT, INP- Purpan, UPS, Toulouse, France
| | - David R Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, Florida, USA,Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, USA,Department of Infectious Diseases and Immunology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
32
|
Liu J, Parrish JR, Hines J, Mansfield L, Finley RL. A proteome-wide screen of Campylobacter jejuni using protein microarrays identifies novel and conformational antigens. PLoS One 2019; 14:e0210351. [PMID: 30633767 PMCID: PMC6329530 DOI: 10.1371/journal.pone.0210351] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023] Open
Abstract
Campylobacter jejuni (C. jejuni) is a foodborne intestinal pathogen and major cause of gastroenteritis worldwide. C. jejuni proteins that are immunogenic have been sought for their potential use in the development of biomarkers, diagnostic assays, or subunit vaccines for humans or livestock. To identify new immunogenic C. jejuni proteins, we used a native protein microarray approach. A protein chip, with over 1400 individually purified GST-tagged C. jejuni proteins, representing over 86% of the proteome, was constructed to screen for antibody titers present in test sera raised against whole C. jejuni cells. Dual detection of GST signals was incorporated as a way of normalizing the variation of protein concentrations contributing to the antibody staining intensities. We detected strong signals to 102 C. jejuni antigens. In addition to antigens recognized by antiserum raised against C. jejuni, parallel experiments were conducted to identify antigens cross-reactive to antiserum raised against various serotypes of E. coli or Salmonella or to healthy human sera. This led to the identification of 34 antigens specifically recognized by the C. jejuni antiserum, only four of which were previously known. The chip approach also allowed identification of conformational antigens. We demonstrate in the case of Cj1621 that antigen signals are lost to denaturing conditions commonly used in other approaches to identify immunogens. Antigens identified in this study include those possessing sequence features indicative of cell surface localization, as well as those that do not. Together, our results indicate that the unbiased chip-based screen can help reveal the full repertoire of host antibodies against microbial proteomes.
Collapse
Affiliation(s)
- Jiayou Liu
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Jodi R Parrish
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Julie Hines
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Linda Mansfield
- Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| | - Russell L Finley
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America.,Department of Microbiology, Immunology, and Biochemistry Wayne State University School of Medicine, Detroit, Michigan, United States of America
| |
Collapse
|
33
|
Ren F, Li X, Tang H, Jiang Q, Yun X, Fang L, Huang P, Tang Y, Li Q, Huang J, Jiao XA. Insights into the impact of flhF inactivation on Campylobacter jejuni colonization of chick and mice gut. BMC Microbiol 2018; 18:149. [PMID: 30348090 PMCID: PMC6196472 DOI: 10.1186/s12866-018-1318-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/15/2018] [Indexed: 11/23/2022] Open
Abstract
Background Campylobacter jejuni (C. jejuni) is a leading cause of foodborne gastroenteritis worldwide. This bacterium lacks many of the classical virulence factors, and flagellum-associated persistent colonization has been shown to be crucial for its pathogenesis. The flagellum plays a multifunctional role in C. jejuni pathogenesis, and different flagellar elements make diverse contributions. The flhF gene encodes the flagellar biosynthesis regulator, which is important for flagellar biosynthesis. In this study, the influence of flhF on C. jejuni colonization was systematically studied, and the possible mechanisms were also analyzed. Results The flhF gene has a significant influence on C. jejuni colonization, and its inactivation resulted in severe defects in the commensal colonization of chicks, with approximately 104- to 107-fold reductions (for NCTC 11168 and a C. jejuni isolate respectively) observed in the bacterial caecal loads. Similar effects were observed in mice where the flhF mutant strain completely lost the ability to continuously colonize mice, which cleared the isolate at 7 days post inoculation. Characterization of the phenotypic properties of C. jejuni that influence colonization showed that the adhesion and invasion abilities of the C. jejuni flhF mutant were reduced to approximately 52 and 27% of that of the wild-type strain, respectively. The autoagglutination and biofilm-formation abilities of the flhF mutant strain were also significantly decreased. Further genetic investigation revealed that flhF is continuously upregulated during the infection process, which indicates a close association of this gene with C. jejuni pathogenesis. The transcription of some other infection-related genes that are not directly involved in flagellar assembly were also influenced by its inactivation, with the flagellar coexpressed determinants (Feds) being apparently affected. Conclusions Inactivation of flhF has a significant influence on C. jejuni colonization in both birds and mammals. This defect may be caused by the decreased adhesion, invasion, autoagglutination and biofilm-formation abilities of the flhF mutant strain, as well as the influence on the transcription of other infection related genes, which provides insights into this virulence factor and the flagellum mediated co-regulation of C. jejuni pathogenesis. Electronic supplementary material The online version of this article (10.1186/s12866-018-1318-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fangzhe Ren
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China
| | - Xiaofei Li
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China
| | - Haiyan Tang
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China
| | - Qidong Jiang
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China
| | - Xi Yun
- Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Lin Fang
- Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Pingyu Huang
- Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Yuanyue Tang
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China.,Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Qiuchun Li
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China.,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China.,Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China
| | - Jinlin Huang
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China. .,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China. .,Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China.
| | - Xin-An Jiao
- Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 East Wenhui Road, Yangzhou, 225009, Jiangsu, China. .,Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou, 225009, Jiangsu, China. .,Joint International Research Laboratory of Agriculture and Agri-product Safety, Ministry of Education of China, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
34
|
Abstract
Background: Acute diarrheal disease caused by viral, bacterial and parasitic infections are a major global health problem with substantial mortality and morbidity in children under five years of age in lower and middle income countries. However, a number of these infections also impact large segments of populations in upper income countries, as well as individuals who travel overseas for work, business or pleasure. Campylobacter has been and continues to be a leading cause of disease burden globally across all income countries. Aims: The aim of this review is to describe recent understanding in burden of disease, consider the current landscape of Campylobacter vaccine development, and address the challenges that need to be overcome. Sources: Relevant data from the literature as well as clinical trials described in European and US registries were used to conduct this review. Content: Despite advances in population health, food security, improved sanitation, water quality and the reduction of poverty, Campylobacter infections continue to plague global populations. The emerging recognition of chronic health consequences attributed to this pathogen is changing the potential valuation of preventive interventions. Advancing development of new vaccines is a present opportunity and holds promise.
Collapse
Affiliation(s)
- Frédéric Poly
- a Enteric Diseases Department , Naval Medical Research Center , Silver Spring , MD , USA
| | - Alexander J Noll
- a Enteric Diseases Department , Naval Medical Research Center , Silver Spring , MD , USA
| | - Mark S Riddle
- b F. Edward Hébert School of Medicine , Uniformed Services University , Bethesda , MD , USA
| | - Chad K Porter
- a Enteric Diseases Department , Naval Medical Research Center , Silver Spring , MD , USA
| |
Collapse
|
35
|
Gut-brain actions underlying comorbid anxiety and depression associated with inflammatory bowel disease. Acta Neuropsychiatr 2018; 30:275-296. [PMID: 28270247 DOI: 10.1017/neu.2017.3] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
UNLABELLED IntroductionInflammatory bowel disease (IBD) is a chronic relapsing and remitting disorder characterised by inflammation of the gastrointestinal tract. There is a growing consensus that IBD is associated with anxiety- and depression-related symptoms. Psychological symptoms appear to be more prevalent during active disease states with no difference in prevalence between Crohn's disease and ulcerative colitis. Behavioural disturbances including anxiety- and depression-like symptoms have also been observed in animal models of IBD. RESULTS The likely mechanisms underlying the association are discussed with particular reference to communication between the gut and brain. The close bidirectional relationship known as the gut-brain axis includes neural, hormonal and immune communication links. Evidence is provided for a number of interacting factors including activation of the inflammatory response system in the brain, the hypothalamic-pituitary-adrenal axis, and brain areas implicated in altered behaviours, changes in blood brain barrier integrity, and an emerging role for gut microbiota and response to probiotics in IBD.DiscussionThe impact of psychological stress in models of IBD remains somewhat conflicted, however, it is weighted in favour of stress or early stressful life events as risk factors in the development of IBD, stress-induced exacerbation of inflammation and relapse. CONCLUSION It is recommended that patients with IBD be screened for psychological disturbance and treated accordingly as intervention can improve quality of life and may reduce relapse rates.
Collapse
|
36
|
Dann SM, Le CHY, Hanson EM, Ross MC, Eckmann L. Giardia Infection of the Small Intestine Induces Chronic Colitis in Genetically Susceptible Hosts. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:548-559. [PMID: 29898958 PMCID: PMC7351291 DOI: 10.4049/jimmunol.1700824] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 05/07/2018] [Indexed: 01/01/2023]
Abstract
The lumen-dwelling protozoan Giardia is an important parasitic cause of diarrheal disease worldwide. Infection can persist over extended periods with minimal intestinal inflammation, suggesting that Giardia may attenuate host responses to ensure its survival, although clearance eventually occurs in most cases. IL-10 is an anti-inflammatory regulator critical for intestinal homeostasis and controlling host responses to bacterial exposure, yet its potential role in coordinating antiprotozoal host defense in the intestine is not known. In this study, we found that murine infection with the natural enteric pathogen Giardia muris induced a transient IL-10 response after 2-4 wk at the primary site of infection in the upper small intestine, but parasite colonization and eradication were not affected by the absence of the cytokine in gene-targeted mice. However, IL-10 was critical for controlling infection-associated immunological sequelae in the colon because severe and persistent diarrhea and colitis were observed in IL-10-deficient mice within 1-2 wk postinfection but not in uninfected littermate controls. Inflammation was characterized by epithelial hyperplasia, neutrophil and macrophage expansion, and Th1 induction and could be prevented by blockade of IL-12/IL-23 p40 but not depletion of CD11c+ dendritic cells. Furthermore, the intestinal microbiota underwent characteristic shifts in composition and was required for disease because antibiotics and loss of TLR signaling in MyD88-deficient mice protected against colitis. Together, our data suggest that transient infection by a luminal and seemingly noninflammatory pathogen can trigger sustained colitis in genetically susceptible hosts, which has broader implications for understanding postinfectious syndromes and other chronic intestinal inflammatory conditions.
Collapse
Affiliation(s)
- Sara M Dann
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Christine H Y Le
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Elaine M Hanson
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Matthew C Ross
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| |
Collapse
|
37
|
Brooks PT, Brakel KA, Bell JA, Bejcek CE, Gilpin T, Brudvig JM, Mansfield LS. Transplanted human fecal microbiota enhanced Guillain Barré syndrome autoantibody responses after Campylobacter jejuni infection in C57BL/6 mice. MICROBIOME 2017; 5:92. [PMID: 28789710 PMCID: PMC5547673 DOI: 10.1186/s40168-017-0284-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/05/2017] [Indexed: 05/10/2023]
Abstract
BACKGROUND Campylobacter jejuni is the leading antecedent infection to the autoimmune neuropathy Guillain-Barré syndrome (GBS), which is accompanied by an autoimmune anti-ganglioside antibody attack on peripheral nerves. Previously, we showed that contrasting immune responses mediate C. jejuni induced colitis and autoimmunity in interleukin-10 (IL-10)-deficient mice, dependent upon the infecting strain. Strains from colitis patients elicited T helper 1 (TH1)-dependent inflammatory responses while strains from GBS patients elicited TH2-dependent autoantibody production. Both syndromes were exacerbated by antibiotic depletion of the microbiota, but other factors controlling susceptibility to GBS are unknown. METHODS Using 16S rRNA gene high-throughput sequencing, we examined whether structure of the gut microbial community alters host (1) gastrointestinal inflammation or (2) anti-ganglioside antibody responses after infection with C. jejuni strains from colitis or GBS patients. We compared these responses in C57BL/6 mice with either (1) stable human gut microbiota (Humicrobiota) transplants or (2) conventional mouse microbiota (Convmicrobiota). RESULTS Inoculating germ-free C57BL/6 wild-type (WT) mice with a mixed human fecal slurry provided a murine model that stably passed its microbiota over >20 generations. Mice were housed in specific pathogen-free (SPF) facilities, while extra precautions of having caretakers wear sterile garb along with limited access ensured that no mouse pathogens were acquired. Humicrobiota conferred many changes upon the WT model in contrast to previous results, which showed only colonization with no disease after C. jejuni challenge. When compared to Convmicrobiota mice for susceptibility to C. jejuni enteric or GBS patient strains, infected Humicrobiota mice had (1) 10-100 fold increases in C. jejuni colonization of both strains, (2) pathologic change in draining lymph nodes but only mild changes in colon or cecal lamina propria, (3) significantly lower Th1/Th17-dependent anti-C. jejuni responses, (4) significantly higher IL-4 responses at 5 but not 7 weeks post infection (PI), (5) significantly higher Th2-dependent anti-C. jejuni responses, and (6) significantly elevated anti-ganglioside autoantibodies after C. jejuni infection. These responses in Humicrobiota mice were correlated with a dominant Bacteroidetes and Firmicutes microbiota. CONCLUSIONS These data demonstrate that Humicrobiota altered host-pathogen interactions in infected mice, increasing colonization and Th-2 and autoimmune responses in a C. jejuni strain-dependent manner. Thus, microbiota composition is another factor controlling susceptibility to GBS.
Collapse
Affiliation(s)
- Phillip T Brooks
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Kelsey A Brakel
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Julia A Bell
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Christopher E Bejcek
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
| | - Trey Gilpin
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
| | - Jean M Brudvig
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI, USA
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Linda S Mansfield
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI, USA.
- Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, MI, USA.
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA.
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA.
- Department of Microbiology and Molecular Genetics, Michigan State University, 181 Food Safety Building; 1129 Farm Lane, East Lansing, MI, 48824, USA.
| |
Collapse
|
38
|
Heimesaat MM, Grundmann U, Alutis ME, Fischer A, Bereswill S. Small Intestinal Pro-Inflammatory Immune Responses Following Campylobacter Jejuni Infection of Secondary Abiotic IL-10 -/- Mice Lacking Nucleotide-Oligomerization-Domain-2. Eur J Microbiol Immunol (Bp) 2017; 7:138-145. [PMID: 28690881 PMCID: PMC5495086 DOI: 10.1556/1886.2017.00005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/13/2017] [Indexed: 01/08/2023] Open
Abstract
Host immune responses are crucial for combating enteropathogenic infections including Campylobacter jejuni. Within 1 week following peroral C. jejuni infection, secondary abiotic IL-10–/– mice develop severe immunopathological sequelae affecting the colon (ulcerative enterocolitis). In the present study, we addressed whether pathogen-induced pro-inflammatory immune responses could also be observed in the small intestines dependent on the innate receptor nucleotide-oligomerization-domain-protein 2 (Nod2). Within 7 days following peroral infection, C. jejuni stably colonized the gastrointestinal tract of both IL-10–/– mice lacking Nod2 (Nod2–/– IL-10–/–) and IL-10–/– controls displaying bloody diarrhea with similar frequencies. Numbers of apoptotic and regenerating epithelial cells increased in the small intestines of C. jejuni-infected mice of either genotype that were accompanied by elevated ileal T and B lymphocyte counts. Notably, ileal T cell numbers were higher in C. jejuni-infected Nod2–/– IL-10–/– as compared to IL-10–/– counterparts. Furthermore, multifold increased concentrations of pro-inflammatory cytokines including IFN-γ, TNF, and MCP-1 could be measured in small intestinal ex vivo biopsies derived from C. jejuni-infected mice of either genotype. In conclusion, C. jejuni-induced pro-inflammatory immune responses affected the small intestines of both Nod2–/– IL-10–/– and IL-10–/– mice, whereas ileal T lymphocyte numbers were even higher in the former.
Collapse
Affiliation(s)
- Markus M Heimesaat
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Ursula Grundmann
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Marie E Alutis
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - André Fischer
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Stefan Bereswill
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| |
Collapse
|
39
|
Stahl M, Graef FA, Vallance BA. Mouse Models for Campylobacter jejuni Colonization and Infection. Methods Mol Biol 2017; 1512:171-188. [PMID: 27885607 DOI: 10.1007/978-1-4939-6536-6_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Relevant animal models for Campylobacter jejuni infection have been difficult to establish due to C. jejuni's inability to cause disease in many common animal research models. Fortunately, recent work has proven successful in developing several new and relevant mouse models of C. jejuni infection, including the SIGIRR-deficient mouse strain that develops acute enterocolitis in response to C. jejuni. Here we describe how to properly infect mice with C. jejuni, as well as a number of accompanying histological techniques to aid in studying C. jejuni colonization and infection in mice.
Collapse
Affiliation(s)
- Martin Stahl
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, BC, V6H 3V4, Canada
| | - Franziska A Graef
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, BC, V6H 3V4, Canada
| | - Bruce A Vallance
- Division of Gastroenterology, Department of Pediatrics, Child and Family Research Institute, University of British Columbia, Vancouver, BC, V6H 3V4, Canada.
| |
Collapse
|
40
|
St Charles JL, Bell JA, Gadsden BJ, Malik A, Cooke H, Van de Grift LK, Kim HY, Smith EJ, Mansfield LS. Guillain Barré Syndrome is induced in Non-Obese Diabetic (NOD) mice following Campylobacter jejuni infection and is exacerbated by antibiotics. J Autoimmun 2016; 77:11-38. [PMID: 27939129 DOI: 10.1016/j.jaut.2016.09.003] [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: 01/06/2016] [Revised: 08/25/2016] [Accepted: 09/04/2016] [Indexed: 01/10/2023]
Abstract
Campylobacter jejuni is a leading cause of bacterial gastroenteritis linked to several serious autoimmune sequelae such as the peripheral neuropathies Guillain Barré syndrome (GBS) and Miller Fisher syndrome (MFS). We hypothesized that GBS and MFS can result in NOD wild type (WT) mice or their congenic interleukin (IL)-10 or B7-2 knockouts secondary to C. jejuni infection. Mice were gavaged orally with C. jejuni strains HB93-13 and 260.94 from patients with GBS or CF93-6 from a patient with MFS and assessed for clinical neurological signs and phenotypes, anti-ganglioside antibodies, and cellular infiltrates and lesions in gut and peripheral nerve tissues. Significant increases in autoantibodies against single gangliosides (GM1, GQ1b, GD1a) occurred in infected NOD mice of all genotypes, although the isotypes varied (NOD WT had IgG1, IgG3; NOD B7-2-/- had IgG3; NOD IL-10-/- had IgG1, IgG3, IgG2a). Infected NOD WT and NOD IL-10-/- mice also produced anti-ganglioside antibodies of the IgG1 isotype directed against a mixture of GM1/GQ1b gangliosides. Phenotypic tests showed significant differences between treatment groups of all mouse genotypes. Peripheral nerve lesions with macrophage infiltrates were significantly increased in infected mice of NOD WT and IL-10-/- genotypes compared to sham-inoculated controls, while lesions with T cell infiltrates were significantly increased in infected mice of the NOD B7-2-/- genotype compared to sham-inoculated controls. In both infected and sham inoculated NOD IL-10-/- mice, antibiotic treatment exacerbated neurological signs, lesions and the amount and number of different isotypes of antiganglioside autoantibodies produced. Thus, inducible mouse models of post-C. jejuni GBS are feasible and can be characterized based on evaluation of three factors-onset of GBS clinical signs/phenotypes, anti-ganglioside autoantibodies and nerve lesions. Based on these factors we characterized 1) NOD B-7-/- mice as an acute inflammatory demyelinating polyneuropathy (AIDP)-like model, 2) NOD IL-10-/- mice as an acute motor axonal neuropathy (AMAN)-like model best employed over a limited time frame, and 3) NOD WT mice as an AMAN model with mild clinical signs and lesions. Taken together these data demonstrate that C. jejuni strain genotype, host genotype and antibiotic treatment affect GBS disease outcomes in mice and that many disease phenotypes are possible.
Collapse
Affiliation(s)
- J L St Charles
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI 48824, USA; Comparative Medicine and Integrative Biology Graduate Program, Michigan State University, East Lansing, MI 48824, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - J A Bell
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI 48824, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - B J Gadsden
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI 48824, USA; Comparative Medicine and Integrative Biology Graduate Program, Michigan State University, East Lansing, MI 48824, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - A Malik
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI 48824, USA; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - H Cooke
- College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - L K Van de Grift
- College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - H Y Kim
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI 48824, USA
| | - E J Smith
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI 48824, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - L S Mansfield
- Comparative Enteric Diseases Laboratory, Michigan State University, East Lansing, MI 48824, USA; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA; College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
41
|
Fiebiger U, Bereswill S, Heimesaat MM. Dissecting the Interplay Between Intestinal Microbiota and Host Immunity in Health and Disease: Lessons Learned from Germfree and Gnotobiotic Animal Models. Eur J Microbiol Immunol (Bp) 2016; 6:253-271. [PMID: 27980855 PMCID: PMC5146645 DOI: 10.1556/1886.2016.00036] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 02/06/2023] Open
Abstract
This review elaborates the development of germfree and gnotobiotic animal models and their application in the scientific field to unravel mechanisms underlying host-microbe interactions and distinct diseases. Strictly germfree animals are raised in isolators and not colonized by any organism at all. The germfree state is continuously maintained by birth, raising, housing and breeding under strict sterile conditions. However, isolator raised germfree mice are exposed to a stressful environment and exert an underdeveloped immune system. To circumvent these physiological disadvantages depletion of the bacterial microbiota in conventionally raised and housed mice by antibiotic treatment has become an alternative approach. While fungi and parasites are not affected by antibiosis, the bacterial microbiota in these "secondary abiotic mice" have been shown to be virtually eradicated. Recolonization of isolator raised germfree animals or secondary abiotic mice results in a gnotobiotic state. Both, germfree and gnotobiotic mice have been successfully used to investigate biological functions of the conventional microbiota in health and disease. Particularly for the development of novel clinical applications germfree mice are widely used tools, as summarized in this review further focusing on the modulation of bacterial microbiota in laboratory mice to better mimic conditions in the human host.
Collapse
Affiliation(s)
| | | | - Markus M. Heimesaat
- Gastrointestinal Microbiology Research Group, Institute of Microbiology and Hygiene, Charité – University Medicine Berlin, Campus Benjamin Franklin
| |
Collapse
|
42
|
Yu ZT, Nanthakumar NN, Newburg DS. The Human Milk Oligosaccharide 2'-Fucosyllactose Quenches Campylobacter jejuni-Induced Inflammation in Human Epithelial Cells HEp-2 and HT-29 and in Mouse Intestinal Mucosa. J Nutr 2016; 146:1980-1990. [PMID: 27629573 PMCID: PMC5037868 DOI: 10.3945/jn.116.230706] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 08/09/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Campylobacter jejuni causes diarrhea worldwide; young children are most susceptible. Binding of virulent C. jejuni to the intestinal mucosa is inhibited ex vivo by α1,2-fucosylated carbohydrate moieties, including human milk oligosaccharides (HMOSs). OBJECTIVE The simplest α1,2-fucosylated HMOS structure, 2'-fucosyllactose (2'-FL), can be predominant at ≤5 g/L milk. Although 2'-FL inhibits C. jejuni binding ex vivo and in vivo, the effects of 2'FL on the cell invasion central to C. jejuni pathogenesis have not been tested. Clinical isolates of C. jejuni infect humans, birds, and ferrets, limiting studies on its mammalian pathobiology. METHODS Human epithelial cells HEp-2 and HT-29 infected with the virulent C. jejuni strain 81-176 human isolate were treated with 5 g 2'-FL/L, and the degree of infection and inflammatory response was measured. Four-week-old male wild-type C57BL/6 mice were fed antibiotics to reduce their intestinal microbiota and were inoculated with C. jejuni strain 81-176. The sensitivity of the resulting acute transient enteric infection and immune response to inhibition by 2'-FL ingestion was tested. RESULTS In HEp-2 and HT-29 cells, 2'-FL attenuated 80% of C. jejuni invasion (P < 0.05) and suppressed the release of mucosal proinflammatory signals of interleukin (IL) 8 by 60-70%, IL-1β by 80-90%, and the neutrophil chemoattractant macrophage inflammatory protein 2 (MIP-2) by 50% (P < 0.05). Ingestion of 2'-FL by mice reduced C. jejuni colonization by 80%, weight loss by 5%, histologic features of intestinal inflammation by 50-70%, and induction of inflammatory signaling molecules of the acute-phase mucosal immune response by 50-60% (P < 0.05). This acute model did not induce IL-17 (adaptive T cell response), a chronic response. CONCLUSIONS In human cells in vitro (HEp-2, HT-29) and in a mouse infection model that recapitulated key pathologic features of C. jejuni clinical disease, 2'-FL inhibited pathogenesis and its sequelae. These data strongly support the hypothesis that 2'-FL represents a new class of oral agent for prevention, and potentially for treatment, of specific enteric infectious diseases.
Collapse
Affiliation(s)
- Zhuo-Teng Yu
- Department of Biology, Boston College, Chestnut Hill, MA; and
| | - N Nanda Nanthakumar
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - David S Newburg
- Department of Biology, Boston College, Chestnut Hill, MA; and
| |
Collapse
|
43
|
Vaezirad MM, Keestra-Gounder AM, de Zoete MR, Koene MG, Wagenaar JA, van Putten JPM. Invasive behavior of Campylobacter jejuni in immunosuppressed chicken. Virulence 2016; 8:248-260. [PMID: 27574876 PMCID: PMC5411237 DOI: 10.1080/21505594.2016.1221559] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Campylobacter jejuni is a predominant cause of gastroenteritis in humans but rather harmless in chickens. The basis of this difference is unknown. We investigated the effect of the chicken immune defense on the behavior of C. jejuni using glucocorticoid (GC)-treated and mock-treated 17-day old Ross 308 chicken bearing in mind that GCs have immunosuppressive effects and dampen the innate immune response. The effect of GC administration on the behavior of C. jejuni was compared with that on infection with Salmonella Enteritidis to address possible microbe-associated differences. Our results revealed that GC treatment fastened the intestinal colonization of C. jejuni (p < 0.001) and enhanced its dissemination to the liver (p = 0.007). The effect of GC on intestinal colonization of S. Enteritidis was less pronounced (p = 0.033) but GC did speed up the spread of this pathogen to the liver (p < 0.001). Cytokine transcript analysis showed an up to 30-fold reduction in baseline levels of IL-8 mRNA in the cecal (but not spleen) tissue at Day 1 after GC treatment (p < 0.005). Challenge with C. jejuni strongly increased intestinal IL-8, IL-6, and iNOS transcript levels in the non-GC treated animals but not in the GC-treated birds (P < 0.005). In vitro assays with chicken macrophages showed that GC dampened the TLR agonist- and C. jejuni induced-inflammatory gene transcription and production of nitric oxide (P < 0.005). Together, the results support the hypothesis that C. jejuni has the intrinsic ability to invade chicken tissue and that an effective innate immune response may limit its invasive behavior.
Collapse
Affiliation(s)
- Mahdi M Vaezirad
- a Department of Infectious Diseases & Immunology , Utrecht University , Utrecht , the Netherlands.,b University of Birjand , Birjand , Iran
| | | | - Marcel R de Zoete
- a Department of Infectious Diseases & Immunology , Utrecht University , Utrecht , the Netherlands
| | - Miriam G Koene
- c Central Veterinary Institute of Wageningen UR , Lelystad , the Netherlands
| | - Jaap A Wagenaar
- a Department of Infectious Diseases & Immunology , Utrecht University , Utrecht , the Netherlands.,c Central Veterinary Institute of Wageningen UR , Lelystad , the Netherlands
| | - Jos P M van Putten
- a Department of Infectious Diseases & Immunology , Utrecht University , Utrecht , the Netherlands
| |
Collapse
|
44
|
Insights into Campylobacter jejuni colonization and enteritis using a novel infant rabbit model. Sci Rep 2016; 6:28737. [PMID: 27357336 PMCID: PMC4928045 DOI: 10.1038/srep28737] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/07/2016] [Indexed: 01/26/2023] Open
Abstract
A lack of relevant disease models for Campylobacter jejuni has long been an obstacle to research into this common enteric pathogen. Here we used an infant rabbit to study C. jejuni infection, which enables us to define several previously unknown but key features of the organism. C. jejuni is capable of systemic invasion in the rabbit, and developed a diarrhea symptom that mimicked that observed in many human campylobacteriosis. The large intestine was the most consistently colonized site and produced intestinal inflammation, where specific cytokines were induced. Genes preferentially expressed during C. jejuni infection were screened, and acs, cj1385, cj0259 seem to be responsible for C. jejuni invasion. Our results demonstrates that the infant rabbit can be used as an alternative experimental model for the study of diarrheagenic Campylobacter species and will be useful in exploring the pathogenesis of other related pathogens.
Collapse
|
45
|
Kopper JJ, Patterson JS, Mansfield LS. Metronidazole—but not IL-10 or prednisolone—rescues Trichuris muris infected C57BL/6 IL-10 deficient mice from severe disease. Vet Parasitol 2015; 212:239-52. [DOI: 10.1016/j.vetpar.2015.07.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 07/24/2015] [Accepted: 07/31/2015] [Indexed: 10/23/2022]
|
46
|
Stahl M, Vallance BA. Insights into Campylobacter jejuni colonization of the mammalian intestinal tract using a novel mouse model of infection. Gut Microbes 2015; 6:143-8. [PMID: 25831043 PMCID: PMC4615362 DOI: 10.1080/19490976.2015.1016691] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A lack of relevant disease models for Campylobacter jejuni has long been an obstacle to research into this common enteric pathogen. We recently published that mice deficient in Single IgG Interleukin-1 related receptor (SIGIRR), a repressor of MyD88-dependent innate immune signaling, were highly susceptible to enteric infection by murine bacterial pathogens. Subsequently, we successfully employed these mice as an animal model for the human pathogen C. jejuni and gained substantial new insights into infection by this pathogen. The infected mice developed significant intestinal inflammation, primarily via TLR4 stimulation. Furthermore, the resulting gastroenteritis was dependent on C. jejuni pathogenesis as bacterial strains suffering mutations in key virulence factors were attenuated in causing disease. The ability to infect SIGIRR-deficient mice with C. jejuni sheds new light onto how these bacteria colonize the mucus layer of the intestinal tract, invade epithelial cells, and raises new prospects for studying the virulence strategies and pathogenesis of C. jejuni.
Collapse
Affiliation(s)
- Martin Stahl
- Division of Gastroenterology; BC's Children's Hospital; The Child and Family Research Institute and The University of British Columbia; Vancouver, BC Canada
| | - Bruce A Vallance
- Division of Gastroenterology; BC's Children's Hospital; The Child and Family Research Institute and The University of British Columbia; Vancouver, BC Canada,Correspondence to: Bruce A Vallance;
| |
Collapse
|
47
|
Stahl M, Ries J, Vermeulen J, Yang H, Sham HP, Crowley SM, Badayeva Y, Turvey SE, Gaynor EC, Li X, Vallance BA. A novel mouse model of Campylobacter jejuni gastroenteritis reveals key pro-inflammatory and tissue protective roles for Toll-like receptor signaling during infection. PLoS Pathog 2014; 10:e1004264. [PMID: 25033044 PMCID: PMC4102570 DOI: 10.1371/journal.ppat.1004264] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/06/2014] [Indexed: 12/21/2022] Open
Abstract
Campylobacter jejuni is a major source of foodborne illness in the developed world, and a common cause of clinical gastroenteritis. Exactly how C. jejuni colonizes its host's intestines and causes disease is poorly understood. Although it causes severe diarrhea and gastroenteritis in humans, C. jejuni typically dwells as a commensal microbe within the intestines of most animals, including birds, where its colonization is asymptomatic. Pretreatment of C57BL/6 mice with the antibiotic vancomycin facilitated intestinal C. jejuni colonization, albeit with minimal pathology. In contrast, vancomycin pretreatment of mice deficient in SIGIRR (Sigirr−/−), a negative regulator of MyD88-dependent signaling led to heavy and widespread C. jejuni colonization, accompanied by severe gastroenteritis involving strongly elevated transcription of Th1/Th17 cytokines. C. jejuni heavily colonized the cecal and colonic crypts of Sigirr−/− mice, adhering to, as well as invading intestinal epithelial cells. This infectivity was dependent on established C. jejuni pathogenicity factors, capsular polysaccharides (kpsM) and motility/flagella (flaA). We also explored the basis for the inflammatory response elicited by C. jejuni in Sigirr−/− mice, focusing on the roles played by Toll-like receptors (TLR) 2 and 4, as these innate receptors were strongly stimulated by C. jejuni. Despite heavy colonization, Tlr4−/−/Sigirr−/− mice were largely unresponsive to infection by C. jejuni, whereas Tlr2−/−/Sigirr−/− mice developed exaggerated inflammation and pathology. This indicates that TLR4 signaling underlies the majority of the enteritis seen in this model, whereas TLR2 signaling had a protective role, acting to promote mucosal integrity. Furthermore, we found that loss of the C. jejuni capsule led to increased TLR4 activation and exaggerated inflammation and gastroenteritis. Together, these results validate the use of Sigirr−/− mice as an exciting and relevant animal model for studying the pathogenesis and innate immune responses to C. jejuni. Research into the key virulence strategies of the bacterial pathogen Campylobacter jejuni, as well as the host immune responses that develop against this microbe have, in many ways, been limited by the lack of relevant animal models. Here we describe the use of Sigirr deficient (−/−) mice as a model for C. jejuni pathogenesis. Not only do Sigirr−/− mice develop significant intestinal inflammation in response to colonization by C. jejuni, but the ability of this pathogen to trigger gastroenteritis was dependent on key virulence factors. We also found that the induction of the inflammatory and Th1/Th17 immune responses to infection in these mice depended on specific Toll-like receptors, principally TLR4, which we identified as the main driver of inflammation. In contrast, TLR2 signaling was found to protect mucosal integrity, with Tlr2−/−/Sigirr−/− mice suffering exaggerated mucosal damage and inflammation. Notably, we found that C. jejuni's capsule helped conceal it from the host's immune system as its loss led to significantly increased activation of host TLRs and exaggerated gastroenteritis. Our research shows that the increased sensitivity of Sigirr−/− mice can be used to generate a unique and exciting model that facilitates the study of C. jejuni pathogenesis as well as host immunity to this enteric pathogen.
Collapse
Affiliation(s)
- Martin Stahl
- Division of Gastroenterology, British Columbia Children's Hospital, the Child and Family Research Institute and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Jenna Ries
- Division of Gastroenterology, British Columbia Children's Hospital, the Child and Family Research Institute and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Jenny Vermeulen
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hong Yang
- Department of Pediatrics, British Columbia Children's Hospital and Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ho Pan Sham
- Division of Gastroenterology, British Columbia Children's Hospital, the Child and Family Research Institute and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Shauna M. Crowley
- Division of Gastroenterology, British Columbia Children's Hospital, the Child and Family Research Institute and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Yuliya Badayeva
- Division of Gastroenterology, British Columbia Children's Hospital, the Child and Family Research Institute and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Stuart E. Turvey
- Department of Pediatrics, British Columbia Children's Hospital and Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Erin C. Gaynor
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiaoxia Li
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
| | - Bruce A. Vallance
- Division of Gastroenterology, British Columbia Children's Hospital, the Child and Family Research Institute and the University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
48
|
Campylobacter jejuni is not merely a commensal in commercial broiler chickens and affects bird welfare. mBio 2014; 5:e01364-14. [PMID: 24987092 PMCID: PMC4161246 DOI: 10.1128/mbio.01364-14] [Citation(s) in RCA: 188] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Campylobacter jejuni is the leading cause of bacterial food-borne infection; chicken meat is its main source. C. jejuni is considered commensal in chickens based on experimental models unrepresentative of commercial production. Here we show that the paradigm of Campylobacter commensalism in the chicken is flawed. Through experimental infection of four commercial breeds of broiler chickens, we show that breed has a significant effect on C. jejuni infection and the immune response of the animals, although these factors have limited impact on the number of bacteria in chicken ceca. All breeds mounted an innate immune response. In some breeds, this response declined when interleukin-10 was expressed, consistent with regulation of the intestinal inflammatory response, and these birds remained healthy. In another breed, there was a prolonged inflammatory response, evidence of damage to gut mucosa, and diarrhea. We show that bird type has a major impact on infection biology of C. jejuni. In some breeds, infection leads to disease, and the bacterium cannot be considered a harmless commensal. These findings have implications for the welfare of chickens in commercial production where C. jejuni infection is a persistent problem. Importance: Campylobacter jejuni is the most common cause of food-borne bacterial diarrheal disease in the developed world. Chicken is the most common source of infection. C. jejuni infection of chickens had previously not been considered to cause disease, and it was thought that C. jejuni was part of the normal microbiota of birds. In this work, we show that modern rapidly growing chicken breeds used in intensive production systems have a strong inflammatory response to C. jejuni infection that can lead to diarrhea, which, in turn, leads to damage to the feet and legs on the birds due to standing on wet litter. The response and level of disease varied between breeds and is related to regulation of the inflammatory immune response. These findings challenge the paradigm that C. jejuni is a harmless commensal of chickens and that C. jejuni infection may have substantial impact on animal health and welfare in intensive poultry production:
Collapse
|
49
|
Contrasting immune responses mediate Campylobacter jejuni-induced colitis and autoimmunity. Mucosal Immunol 2014; 7:802-17. [PMID: 24220299 PMCID: PMC4112758 DOI: 10.1038/mi.2013.97] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 10/10/2013] [Accepted: 10/12/2013] [Indexed: 02/04/2023]
Abstract
Campylobacter jejuni is a leading cause of foodborne enteritis that has been linked to the autoimmune neuropathy, Guillain Barré syndrome (GBS). C57BL/6 interleukin (IL)-10(+/+) and congenic IL-10(-/-) mice serve as C. jejuni colonization and colitis models, respectively, but a mouse model for GBS is lacking. We demonstrate that IL-10(-/-) mice infected with a C. jejuni colitogenic human isolate had significantly upregulated type 1 and 17 but not type 2 cytokines in the colon coincident with infiltration of phagocytes, T cells and innate lymphoid cells (ILCs). Both ILC and T cells participated in interferon-γ (IFN-γ), IL-17, and IL-22 upregulation but in a time- and organ-specific manner. T cells were, however, necessary for colitis as mice depleted of Thy-1(+) cells were protected while neither Rag1(-/-) nor IL-10R blocked Rag1(-/-) mice developed colitis after infection. Depleting IFN-γ, IL-17, or both significantly ameliorated colitis and drove colonic responses toward type 2 cytokine and antibody induction. In contrast, C. jejuni GBS patient strains induced mild colitis associated with blunted type 1/17 but enhanced type 2 responses. Moreover, the type 2 but not type 1/17 antibodies cross-reacted with peripheral nerve gangliosides demonstrating autoimmunity.
Collapse
|
50
|
Stephenson HN, Mills DC, Jones H, Milioris E, Copland A, Dorrell N, Wren BW, Crocker PR, Escors D, Bajaj-Elliott M. Pseudaminic acid on Campylobacter jejuni flagella modulates dendritic cell IL-10 expression via Siglec-10 receptor: a novel flagellin-host interaction. J Infect Dis 2014; 210:1487-98. [PMID: 24823621 PMCID: PMC4195440 DOI: 10.1093/infdis/jiu287] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Introduction. Campylobacter jejuni is a leading cause of bacterial gastroenteritis worldwide. At present the identity of host-pathogen interactions that promote successful bacterial colonisation remain ill defined. Herein, we aimed to investigate C. jejuni-mediated effects on dendritic cell (DC) immunity. Results. We found C. jejuni to be a potent inducer of human and murine DC interleukin 10 (IL-10) in vitro, a cellular event that was MyD88- and p38 MAPK-signalling dependent. Utilizing a series of C. jejuni isogenic mutants we found the major flagellin protein, FlaA, modulated IL-10 expression, an intriguing observation as C. jejuni FlaA is not a TLR5 agonist. Further analysis revealed pseudaminic acid residues on the flagella contributed to DC IL-10 expression. We identified the ability of both viable C. jejuni and purified flagellum to bind to Siglec-10, an immune-modulatory receptor. In vitro infection of Siglec-10 overexpressing cells resulted in increased IL-10 expression in a p38-dependent manner. Detection of Siglec-10 on intestinal CD11c+ CD103+ DCs added further credence to the notion that this novel interaction may contribute to immune outcome during human infection. Conclusions. We propose that unlike the Salmonella Typhimurium flagella-TLR5 driven pro-inflammatory axis, C. jejuni flagella instead promote an anti-inflammatory axis via glycan-Siglec-10 engagement.
Collapse
Affiliation(s)
- Holly N Stephenson
- Infectious Diseases and Microbiology Unit, Institute of Child Health, University College London
| | - Dominic C Mills
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine
| | - Hannah Jones
- Infectious Diseases and Microbiology Unit, Institute of Child Health, University College London
| | - Enea Milioris
- Infectious Diseases and Microbiology Unit, Institute of Child Health, University College London
| | - Alastair Copland
- Infectious Diseases and Microbiology Unit, Institute of Child Health, University College London
| | - Nick Dorrell
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine
| | - Brendan W Wren
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine
| | | | - David Escors
- Rayne Institute, University College London, United Kingdom Navarrabiomed-Fundacion Miguel Servet, Pamplona, Navarra, Spain
| | - Mona Bajaj-Elliott
- Infectious Diseases and Microbiology Unit, Institute of Child Health, University College London
| |
Collapse
|