1
|
Whitaker EE, Mecum NE, Cott RC, Goode DJ. Expression of MHC II in DRG neurons attenuates paclitaxel-induced cold hypersensitivity in male and female mice. PLoS One 2024; 19:e0298396. [PMID: 38330029 PMCID: PMC10852343 DOI: 10.1371/journal.pone.0298396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Chemotherapy is often a life-saving treatment, but the development of intractable pain caused by chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting toxicity that restricts cancer survival rates. Recent reports demonstrate that paclitaxel (PTX) robustly increases anti-inflammatory CD4+ T cells in the dorsal root ganglion (DRG), and that T cells and anti-inflammatory cytokines are protective against CIPN. However, the mechanism by which CD4+ T cells are activated, and the extent cytokines released by CD4+ T cells target DRG neurons are unknown. Here, we are the first to detect major histocompatibility complex II (MHCII) protein in mouse DRG neurons and to find CD4+ T cells breaching the satellite glial cell barrier to be in close proximity to neurons, together suggesting CD4+ T cell activation and targeted cytokine release. MHCII protein is primarily expressed in small nociceptive neurons in male and female mouse DRG but increased after PTX in small nociceptive neurons in only female DRG. Reducing one copy of MHCII in small nociceptive neurons decreased anti-inflammatory IL-10 and IL-4 producing CD4+ T cells in naïve male DRG and increased their hypersensitivity to cold. Administration of PTX to male and female mice that lacked one copy of MHCII in nociceptive neurons decreased anti-inflammatory CD4+ T cells in the DRG and increased the severity of PTX-induced cold hypersensitivity. Collectively, our results demonstrate expression of MHCII protein in mouse DRG neurons, which modulates cytokine producing CD4+ T cells in the DRG and attenuates cold hypersensitivity during homeostasis and after PTX treatment.
Collapse
Affiliation(s)
- Emily E. Whitaker
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States of America
| | - Neal E. Mecum
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States of America
| | - Riley C. Cott
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States of America
| | - Diana J. Goode
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, United States of America
| |
Collapse
|
2
|
Noda M, Danshiitsoodol N, Kanno K, Uchida T, Sugiyama M. The Exopolysaccharide Produced by Lactobacillus paracasei IJH-SONE68 Prevents and Ameliorates Inflammatory Responses in DSS-Induced Ulcerative Colitis. Microorganisms 2021; 9:microorganisms9112243. [PMID: 34835369 PMCID: PMC8621803 DOI: 10.3390/microorganisms9112243] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an autoimmune disease characterized by chronic inflammation of the gastrointestinal tract. IBD includes Crohn’s disease (CD) and ulcerative colitis (UC). CD can occur in any part of the gastrointestinal tract, whereas UC mainly occurs in the colon and rectum. We previously demonstrated that a novel exopolysaccharide (EPS) produced by a plant-derived bacterium, Lactobacillus paracasei IJH-SONE68, prevents and improves the inflammation in contact dermatitis model mice via oral administration. To evaluate the preventive effect of the EPS against other inflammatory diseases, in the present study, we employed dextran sulfate sodium (DSS)-induced UC model mice. The stool consistency, hematochezia, and colonic atrophy of the mice were improved by the orally administered EPS. We also evaluated the cytokine transcription. Overexpression of the mouse macrophage inflammatory protein 2 mRNA in the colon as a functional homolog of human interleukin-8 was decreased by the orally administered EPS. However, the expression of interleukin-10, which is known as an anti-inflammatory cytokine, was stimulated in the EPS-administrated group. Based on these results, we conclude that the IJH-SONE68-derived EPS is a promising lead material for the development of drugs useful in treating inflammatory diseases such as UC.
Collapse
Affiliation(s)
- Masafumi Noda
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.D.)
| | - Narandalai Danshiitsoodol
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.D.)
| | - Keishi Kanno
- Department of General Internal Medicine, Hiroshima University Hospital, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan;
- Department of Clinical Pharmaceutical and Therapeutics, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan;
| | - Tomoyuki Uchida
- Department of Clinical Pharmaceutical and Therapeutics, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8551, Japan;
- Sone Farm Co., Ltd., Shinjuku, Shinjuku-ku, Tokyo 160-0022, Japan
| | - Masanori Sugiyama
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.D.)
- Correspondence: ; Tel.: +81-82-257-5280
| |
Collapse
|
3
|
The Host Cellular Immune Response to Infection by Campylobacter Spp. and Its Role in Disease. Infect Immun 2021; 89:e0011621. [PMID: 34031129 DOI: 10.1128/iai.00116-21] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Campylobacter spp. are the leading cause of bacterium-derived gastroenteritis worldwide, impacting 96 million individuals annually. Unlike other bacterial pathogens of the gastrointestinal tract, Campylobacter spp. lack many of the classical virulence factors that are often associated with the ability to induce disease in humans, including an array of canonical secretion systems and toxins. Consequently, the clinical manifestations of human campylobacteriosis and its resulting gastrointestinal pathology are believed to be primarily due to the host immune response toward the bacterium. Further, while gastrointestinal infection is usually self-limiting, numerous postinfectious disorders can occur, including the development of Guillain-Barré syndrome, reactive arthritis, and irritable bowel syndrome. Because gastrointestinal disease likely results from the host immune response, the development of these postinfectious disorders may be due to dysregulation or misdirection of the same inflammatory response. As a result, it is becoming increasingly important to the Campylobacter field, and human health, that the cellular immune responses toward Campylobacter be better understood, including which immunological events are critical to the development of disease and the postinfectious disorders mentioned above. In this review, we collectively cover the cellular immune responses across susceptible hosts to Campylobacter jejuni infection, along with the tissue pathology and postinfectious disorders which may develop.
Collapse
|
4
|
Abstract
Campylobacter jejuni and Campylobacter coli can be frequently isolated from poultry and poultry-derived products, and in combination these two species cause a large portion of human bacterial gastroenteritis cases. While birds are typically colonized by these Campylobacter species without clinical symptoms, in humans they cause (foodborne) infections at high frequencies, estimated to cost billions of dollars worldwide every year. The clinical outcome of Campylobacter infections comprises malaise, diarrhea, abdominal pain and fever. Symptoms may continue for up to two weeks and are generally self-limiting, though occasionally the disease can be more severe or result in post-infection sequelae. The virulence properties of these pathogens have been best-characterized for C. jejuni, and their actions are reviewed here. Various virulence-associated bacterial determinants include the flagellum, numerous flagellar secreted factors, protein adhesins, cytolethal distending toxin (CDT), lipooligosaccharide (LOS), serine protease HtrA and others. These factors are involved in several pathogenicity-linked properties that can be divided into bacterial chemotaxis, motility, attachment, invasion, survival, cellular transmigration and spread to deeper tissue. All of these steps require intimate interactions between bacteria and host cells (including immune cells), enabled by the collection of bacterial and host factors that have already been identified. The assortment of pathogenicity-associated factors now recognized for C. jejuni, their function and the proposed host cell factors that are involved in crucial steps leading to disease are discussed in detail.
Collapse
|
5
|
Low-Dose Exposure to Ganglioside-Mimicking Bacteria Tolerizes Human Macrophages to Guillain-Barré Syndrome-Associated Antigens. mBio 2021; 13:e0385221. [PMID: 35100875 PMCID: PMC8805021 DOI: 10.1128/mbio.03852-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Early in life, commensal bacteria play a major role in immune development, helping to guide the host response toward harmful stimuli while tolerating harmless antigens to prevent autoimmunity. Guillain-Barré syndrome (GBS) is an autoimmune disease caused by errant immune attack of antibody-bound ganglioside receptors on host nerve cells, resulting in paralysis. Lipooligosaccharides enveloping the prevalent enteric pathogen, Campylobacter jejuni, frequently mimic human gangliosides and can trigger GBS by stimulating the autoimmune response. In low- to middle-income countries, young children are consistently exposed to C. jejuni, and it is not known if this impacts GBS susceptibility later in life. Using a macrophage model, we examined the effect of training these cells with low doses of ganglioside-mimicking bacteria prior to challenge with GBS-associated antigens. This training caused decreased production of proinflammatory cytokines, suggesting tolerance induction. We then screened Campylobacter isolates from 154 infant fecal samples for GM1 ganglioside mimicry, finding that 23.4% of strains from both symptomatic and asymptomatic infants displayed GM1-like structures. Training macrophages with one of these asymptomatic carrier isolates also induced tolerance against GBS-associated antigens, supporting that children can be exposed to the tolerizing antigen early in life. RNA interference of Toll-like receptor 2 (TLR2) and TLR4 suggests that these receptors are not involved in tolerance associated with decreases in tumor necrosis factor (TNF), interleukin-6 (IL-6), or IL-1β levels. The results of this study suggest that exposure to ganglioside-mimicking bacteria early in life occurs naturally and impacts host susceptibility to GBS development. IMPORTANCE In this study, we demonstrated that it is possible to tolerize immune cells to potentially dampen the autoreactive proinflammatory immune response against Guillain-Barré syndrome (GBS)-associated antigens. The innate immune response functions to arm the host against bacterial attack, but it can be tricked into recognizing the host's own cells when infectious bacteria display sugar structures that mimic human glycans. It is this errant response that leads to the autoimmunity and paralysis associated with GBS. By presenting immune cells with small amounts of the bacterial glycan mimic, we were able to suppress the proinflammatory immune response upon subsequent high exposure to glycan-mimicking bacteria. This suggests that individuals who have already been exposed to the glycan mimics in small amounts are less sensitive to autoimmune reactions against these glycans, and this could be a factor in determining susceptibility to GBS.
Collapse
|
6
|
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
|
7
|
Toll-Like Receptor-4 Is Involved in Mediating Intestinal and Extra-Intestinal Inflammation in Campylobacter coli-Infected Secondary Abiotic IL-10 -/- Mice. Microorganisms 2020; 8:microorganisms8121882. [PMID: 33261211 PMCID: PMC7761268 DOI: 10.3390/microorganisms8121882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 01/01/2023] Open
Abstract
Human Campylobacter infections are emerging worldwide and constitute significant health burdens. We recently showed that the immunopathological sequelae in Campylobacter jejuni-infected mice were due to Toll-like receptor (TLR)-4 dependent immune responses induced by bacterial lipooligosaccharide (LOS). Information regarding the molecular mechanisms underlying Campylobacter coli-host interactions are scarce, however. Therefore, we analyzed C. coli-induced campylobacteriosis in secondary abiotic IL-10−/− mice with and without TLR4. Mice were infected perorally with a human C. coli isolate or with a murine commensal Escherichia coli as apathogenic, non-invasive control. Independent from TLR4, C. coli and E. coli stably colonized the gastrointestinal tract, but only C. coli induced clinical signs of campylobacteriosis. TLR4−/− IL-10−/− mice, however, displayed less frequently fecal blood and less distinct histopathological and apoptotic sequelae in the colon versus IL-10−/− counterparts on day 28 following C. coli infection. Furthermore, C. coli-induced colonic immune cell responses were less pronounced in TLR4−/− IL-10−/− as compared to IL-10−/− mice and accompanied by lower pro-inflammatory mediator concentrations in the intestines and the liver of the former versus the latter. In conclusion, our study provides evidence that TLR4 is involved in mediating C. coli-LOS-induced immune responses in intestinal and extra-intestinal compartments during murine campylobacteriosis.
Collapse
|
8
|
Gupta N, Kumar A. Designing an efficient multi-epitope vaccine against Campylobacter jejuni using immunoinformatics and reverse vaccinology approach. Microb Pathog 2020; 147:104398. [PMID: 32771659 DOI: 10.1016/j.micpath.2020.104398] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 01/28/2023]
Abstract
Campylobacterjejuni causes acute diarrhea as a leading cause of morbidity and mortality in children especially in the developing countries of Asia and Africa. C.jejuni has been identified as a member of the priority pathogens category due to the sudden emergence of multidrug-resistant isolates. Therefore, it is important to develop a protective vaccine against this pathogen. In the present study, the Reverse vaccinology approach was used to identify vaccine targets from the proteome of diarrheagenic C. jejuni strain NCTC11168 for the development of chimeric vaccine candidates against C. jejuni. Pathogen proteins that have adhesin like properties and role in virulence but not present in the human host were selected for the design of a multi-epitope vaccine. MHC class I & II and B-cell epitopes present in the selected vaccine target proteins were screened using different immunoinformatics tools. The commonly predicted epitopes from their corresponding different servers were selected and further shortlisted based on their immunogenicity, antigenicity, and toxicity analysis. Shortlisted peptides were joined by GPGPG linkers to design a multi-epitope vaccine construct. Immune-modulating adjuvant monophosphoryl lipid sequence was added with the vaccine construct's N terminal using EAAAK linkers to enhance the immunogenicity. The designed vaccine construct was evaluated by antigenicity, allergenicity, solubility, and physicochemical analysis using various bioinformatics tools. A three-dimensional model of vaccine construct was modeled by the Phyre2 server and refinement by the GalaxyRefine tool. Constructed model quality was validated by the ProSA-web error-detection tool and the Ramachandran plot. After that vaccine model was docked with TLR-4 protein and complex stability confirmed by molecular dynamics simulation studies. Finally, In-silico cloning of vaccine constructs into a vector was performed to ensuring its effective expression in the microbial system.
Collapse
Affiliation(s)
- Nayan Gupta
- Department of Biotechnology, Faculty of Engineering & Technology, Rama University Uttar Pradesh, Kanpur, 209217, India
| | - Ajay Kumar
- Department of Biotechnology, Faculty of Engineering & Technology, Rama University Uttar Pradesh, Kanpur, 209217, India.
| |
Collapse
|
9
|
Callahan S, Doster R, Kelley BR, Gaddy JA, Johnson JG. Induction of neutrophil extracellular traps by Campylobacter jejuni. Cell Microbiol 2020; 22:e13210. [PMID: 32329205 PMCID: PMC7354212 DOI: 10.1111/cmi.13210] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/14/2022]
Abstract
Campylobacter jejuni is the leading cause of bacterial-derived gastroenteritis worldwide and can lead to several post-infectious inflammatory disorders. Despite the prevalence and health impacts of the bacterium, interactions between the host innate immune system and C. jejuni remain poorly understood. To expand on earlier work demonstrating that neutrophils traffic to the site of infection in an animal model of campylobacteriosis, we identified significant increases in several predominantly neutrophil-derived proteins in the faeces of C. jejuni-infected patients, including lipocalin-2, myeloperoxidase and neutrophil elastase. In addition to demonstrating that these proteins significantly inhibited C. jejuni growth, we determined they are released during formation of C. jejuni-induced neutrophil extracellular traps (NETs). Using quantitative and qualitative methods, we found that purified human neutrophils are activated by C. jejuni and exhibit signatures of NET generation, including presence of protein arginine deiminase-4, histone citrullination, myeloperoxidase, neutrophil elastase release and DNA extrusion. Production of NETs correlated with C. jejuni phagocytosis/endocytosis and invasion of neutrophils suggesting that host- and bacterial-mediated activities are responsible for NET induction. Further, NET-like structures were observed within intestinal tissue of C. jejuni-infected ferrets. Finally, induction of NETs significantly increased human colonocyte cytotoxicity, indicating that NET formation during C. jejuni infection may contribute to observed tissue pathology. These findings provide further understanding of C. jejuni-neutrophil interactions and inflammatory responses during campylobacteriosis.
Collapse
Affiliation(s)
- Sean Callahan
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Ryan Doster
- Division of Infectious Diseases, Department of Medicine Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Brittni R. Kelley
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Jennifer A. Gaddy
- Division of Infectious Diseases, Department of Medicine Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeremiah G. Johnson
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
10
|
Mnich ME, van Dalen R, van Sorge NM. C-Type Lectin Receptors in Host Defense Against Bacterial Pathogens. Front Cell Infect Microbiol 2020; 10:309. [PMID: 32733813 PMCID: PMC7358460 DOI: 10.3389/fcimb.2020.00309] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
Antigen-presenting cells (APCs) are present throughout the human body—in tissues, at barrier sites and in the circulation. They are critical for processing external signals to instruct both local and systemic responses toward immune tolerance or immune defense. APCs express an extensive repertoire of pattern-recognition receptors (PRRs) to detect and transduce these signals. C-type lectin receptors (CLRs) comprise a subfamily of PRRs dedicated to sensing glycans, including those expressed by commensal and pathogenic bacteria. This review summarizes recent findings on the recognition of and responses to bacteria by membrane-expressed CLRs on different APC subsets, which are discussed according to the primary site of infection. Many CLR-bacterial interactions promote bacterial clearance, whereas other interactions are exploited by bacteria to enhance their pathogenic potential. The discrimination between protective and virulence-enhancing interactions is essential to understand which interactions to target with new prophylactic or treatment strategies. CLRs are also densely concentrated at APC dendrites that sample the environment across intact barrier sites. This suggests an–as yet–underappreciated role for CLR-mediated recognition of microbiota-produced glycans in maintaining tolerance at barrier sites. In addition to providing a concise overview of identified CLR-bacteria interactions, we discuss the main challenges and potential solutions for the identification of new CLR-bacterial interactions, including those with commensal bacteria, and for in-depth structure-function studies on CLR-bacterial glycan interactions. Finally, we highlight the necessity for more relevant tissue-specific in vitro, in vivo and ex vivo models to develop therapeutic applications in this area.
Collapse
Affiliation(s)
- Malgorzata E Mnich
- Medical Microbiology, UMC Utrecht, Utrecht University, Utrecht, Netherlands.,GSK, Siena, Italy
| | - Rob van Dalen
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Nina M van Sorge
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
11
|
Kløve S, Genger C, Mousavi S, Weschka D, Bereswill S, Heimesaat MM. Toll-Like Receptor-4 Dependent Intestinal and Systemic Sequelae Following Peroral Campylobacter coli Infection of IL10 Deficient Mice Harboring a Human Gut Microbiota. Pathogens 2020; 9:E386. [PMID: 32443576 PMCID: PMC7281621 DOI: 10.3390/pathogens9050386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
Zoonotic Campylobacter, including C. jejuni and C. coli, are among the most prevalent agents of food-borne enteritis worldwide. The immunopathological sequelae of campylobacteriosis are caused by Toll-like Receptor-4 (TLR4)-dependent host immune responses, induced by bacterial lipooligosaccharide (LOS). In order to investigate C. coli-host interactions, including the roles of the human gut microbiota and TLR4, upon infection, we applied a clinical acute campylobacteriosis model, and subjected secondary abiotic, TLR4-deficient IL10-/- mice and IL10-/- controls to fecal microbiota transplantation derived from human donors by gavage, before peroral C. coli challenge. Until day 21 post-infection, C. coli could stably colonize the gastrointestinal tract of human microbiota-associated (hma) mice of either genotype. TLR4-deficient IL10-/- mice, however, displayed less severe clinical signs of infection, that were accompanied by less distinct apoptotic epithelial cell and innate as well as adaptive immune cell responses in the colon, as compared to IL10-/- counterparts. Furthermore, C. coli infected IL10-/-, as opposed to TLR4-deficient IL10-/-, mice displayed increased pro-inflammatory cytokine concentrations in intestinal and, strikingly, systemic compartments. We conclude that pathogenic LOS might play an important role in inducing TLR4-dependent host immune responses upon C. coli infection, which needs to be further addressed in more detail.
Collapse
|
12
|
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
|
13
|
Hameed A. Human Immunity Against Campylobacter Infection. Immune Netw 2019; 19:e38. [PMID: 31921468 PMCID: PMC6943174 DOI: 10.4110/in.2019.19.e38] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
Campylobacter is a worldwide foodborne pathogen, associated with human gastroenteritis. The efficient translocation of Campylobacter and its ability to secrete toxins into host cells are the 2 key features of Campylobacter pathophysiology which trigger inflammation in intestinal cells and contribute to the development of gastrointestinal symptoms, particularly diarrhoea, in humans. The purpose of conducting this literature review is to summarise the current understanding of: i) the human immune responses involved in the elimination of Campylobacter infection and ii) the resistance potential in Campylobacter against these immune responses. This review has highlighted that the intestinal epithelial cells are the preliminary cells which sense Campylobacter cells by means of their cell-surface and cytosolic receptors, activate various receptors-dependent signalling pathways, and recruit the innate immune cells to the site of inflammation. The innate immune system, adaptive immune system, and networking between these systems play a crucial role in bacterial clearance. Different cellular constituents of Campylobacter, mainly cell membrane lipooligosaccharides, capsule, and toxins, provide protection to Campylobacter against the human immune system mediated killing. This review has also identified gaps in knowledge, which are related to the activation of following during Campylobacter infection: i) cathelicidins, bactericidal permeability-increasing proteins, chemokines, and inflammasomes in intestinal epithelial cells; ii) siglec-7 receptors in dendritic cell; iii) acute phase proteins in serum; and iv) T-cell subsets in lymphoid nodules. This review evaluates the existing literature to improve the understanding of human immunity against Campylobacter infection and identify some of the knowledge gaps for future research.
Collapse
Affiliation(s)
- Amber Hameed
- Division of Life Sciences, University of Northampton, Northampton NN1 5PH, UK
| |
Collapse
|
14
|
Zheng G, Wen N, Pan M, Huang Y, Li Z. Biologically active 1,25-dihydroxyvitamin D3 protects against experimental sepsis by negatively regulating the Toll-like receptor 4/myeloid differentiation primary response gene 88/Toll-IL-1 resistance-domain-containing adapter-inducing interferon-β signaling pathway. Int J Mol Med 2019; 44:1151-1160. [PMID: 31524226 DOI: 10.3892/ijmm.2019.4266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 06/18/2019] [Indexed: 11/06/2022] Open
Abstract
The hormonally active form of vitamin D (VD), 1,25‑dihydroxyvitamin D3, has been reported to be a key immunoregulator in the reduction of inflammation. In this study, we investigated the effects of VD in an experimental sepsis cell model, and the underlying mechanisms. The sepsis cell model was first established in monocytes, isolated from newborns and healthy adults, which were stimulation with lipopolysaccharide (LPS). We observed that cell viability was significantly impaired in the monocytes after LPS stimulation, using a Cell Counting Kit‑8 and trypan blue assays. Additionally, ELISA revealed that LPS stimulation significantly elevated the expression of interleukin 6 (IL‑6), IL‑10 and tumor necrosis factor‑α (TNF‑α). The expression levels of Toll‑like receptor (TLR4), myeloid differentiation primary response gene 88 (MyD88), and Toll‑IL‑1 resistance‑domain‑containing adapter‑inducing interferon‑β (TRIF) mRNA were also significantly elevated under LPS stimulation using reverse transcription‑quantitative PCR and western blot analysis. VD treatment could significantly suppress the effects of LPS simulation on monocytes by negatively regulating inflammatory cytokines and TLR4/MyD88/TRIF signaling. Furthermore, a regulatory feedback mechanism was proposed to involve TLR4, MyD88 and TRIF in the sepsis cell model. In conclusion, VD may effectively decrease the release of inflammatory cytokines by inhibiting the TLR4/MyD88/TRIF signaling pathway, could be considered as a potential therapeutic agent for the treatment of sepsis.
Collapse
Affiliation(s)
- Ge Zheng
- Department of Pediatrics, Ruian People's Hospital, Ruian, Zhejiang 325200, P.R. China
| | - Na Wen
- Department of Pediatrics, Ruian People's Hospital, Ruian, Zhejiang 325200, P.R. China
| | - Minli Pan
- Department of Pediatrics, Ruian People's Hospital, Ruian, Zhejiang 325200, P.R. China
| | - Yumao Huang
- Department of Pediatrics, Ruian People's Hospital, Ruian, Zhejiang 325200, P.R. China
| | - Zhishu Li
- Department of Pediatrics, Ruian People's Hospital, Ruian, Zhejiang 325200, P.R. China
| |
Collapse
|
15
|
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: 7] [Impact Index Per Article: 1.2] [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
|
16
|
Suzawa M, Muhammad NM, Joseph BS, Bland ML. The Toll Signaling Pathway Targets the Insulin-like Peptide Dilp6 to Inhibit Growth in Drosophila. Cell Rep 2019; 28:1439-1446.e5. [DOI: 10.1016/j.celrep.2019.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/28/2019] [Accepted: 07/03/2019] [Indexed: 01/08/2023] Open
|
17
|
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
|
18
|
Brunner K, John CM, Phillips NJ, Alber DG, Gemmell MR, Hansen R, Nielsen HL, Hold GL, Bajaj-Elliott M, Jarvis GA. Novel Campylobacter concisus lipooligosaccharide is a determinant of inflammatory potential and virulence. J Lipid Res 2018; 59:1893-1905. [PMID: 30049709 DOI: 10.1194/jlr.m085860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/29/2018] [Indexed: 12/15/2022] Open
Abstract
The pathogenicity of Campylobacter concisus, increasingly found in the human gastrointestinal (GI) tract, is unclear. Some studies indicate that its role in GI conditions has been underestimated, whereas others suggest that the organism has a commensal-like phenotype. For the enteropathogen C. jejuni, the lipooligosaccharide (LOS) is a main driver of virulence. We investigated the LOS structure of four C. concisus clinical isolates and correlated the inflammatory potential of each isolate with bacterial virulence. Mass spectrometric analyses of lipid A revealed a novel hexa-acylated diglucosamine moiety with two or three phosphoryl substituents. Molecular and fragment ion analysis indicated that the oligosaccharide portion of the LOS had only a single phosphate and lacked phosphoethanolamine and sialic acid substitution, which are hallmarks of the C. jejuni LOS. Consistent with our structural findings, C. concisus LOS and live bacteria induced less TNF-α secretion in human monocytes than did C. jejuni Furthermore, the C. concisus bacteria were less virulent than C. jejuni in a Galleria mellonella infection model. The correlation of the novel lipid A structure, decreased phosphorylation, and lack of sialylation along with reduced inflammatory potential and virulence support the significance of the LOS as a determinant in the relative pathogenicity of C. concisus.
Collapse
Affiliation(s)
- Katja Brunner
- Infection, Immunity and Inflammation Programme, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Constance M John
- Center for Immunochemistry, Veterans Affairs Medical Center, San Francisco, CA.,Department of Laboratory Medicine University of California, San Francisco, CA
| | - Nancy J Phillips
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA
| | - Dagmar G Alber
- Infection, Immunity and Inflammation Programme, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Matthew R Gemmell
- Center for Genome-Enabled Biology and Medicine, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Richard Hansen
- Department of Paediatric Gastroenterology, Royal Hospital for Children, Glasgow, United Kingdom
| | - Hans L Nielsen
- Department of Infectious Diseases Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark
| | - Georgina L Hold
- St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Mona Bajaj-Elliott
- Infection, Immunity and Inflammation Programme, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Gary A Jarvis
- Center for Immunochemistry, Veterans Affairs Medical Center, San Francisco, CA .,Department of Laboratory Medicine University of California, San Francisco, CA
| |
Collapse
|
19
|
Rodríguez Y, Rojas M, Pacheco Y, Acosta-Ampudia Y, Ramírez-Santana C, Monsalve DM, Gershwin ME, Anaya JM. Guillain-Barré syndrome, transverse myelitis and infectious diseases. Cell Mol Immunol 2018; 15:547-562. [PMID: 29375121 PMCID: PMC6079071 DOI: 10.1038/cmi.2017.142] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023] Open
Abstract
Guillain-Barré syndrome (GBS) and transverse myelitis (TM) both represent immunologically mediated polyneuropathies of major clinical importance. Both are thought to have a genetic predisposition, but as of yet no specific genetic risk loci have been clearly defined. Both are considered autoimmune, but again the etiologies remain enigmatic. Both may be induced via molecular mimicry, particularly from infectious agents and vaccines, but clearly host factor and co-founding host responses will modulate disease susceptibility and natural history. GBS is an acute inflammatory immune-mediated polyradiculoneuropathy characterized by tingling, progressive weakness, autonomic dysfunction, and pain. Immune injury specifically takes place at the myelin sheath and related Schwann-cell components in acute inflammatory demyelinating polyneuropathy, whereas in acute motor axonal neuropathy membranes on the nerve axon (the axolemma) are the primary target for immune-related injury. Outbreaks of GBS have been reported, most frequently related to Campylobacter jejuni infection, however, other agents such as Zika Virus have been strongly associated. Patients with GBS related to infections frequently produce antibodies against human peripheral nerve gangliosides. In contrast, TM is an inflammatory disorder characterized by acute or subacute motor, sensory, and autonomic spinal cord dysfunction. There is interruption of ascending and descending neuroanatomical pathways on the transverse plane of the spinal cord similar to GBS. It has been suggested to be triggered by infectious agents and molecular mimicry. In this review, we will focus on the putative role of infectious agents as triggering factors of GBS and TM.
Collapse
Affiliation(s)
- Yhojan Rodríguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yovana Pacheco
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Davis, USA, CA
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia.
| |
Collapse
|
20
|
Al-Banna NA, Cyprian F, Albert MJ. Cytokine responses in campylobacteriosis: Linking pathogenesis to immunity. Cytokine Growth Factor Rev 2018; 41:75-87. [PMID: 29550265 DOI: 10.1016/j.cytogfr.2018.03.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/08/2018] [Indexed: 12/15/2022]
Abstract
Campylobacter jejuni is an important enteric pathogen that causes diarrheas of different degrees of severity and several extra-intestinal manifestations, including Guillain-Barre syndrome. The variability of disease outcomes is thought to be linked to the immune response induced by C. jejuni. The virulence factors of C. jejuni induce a pro-inflammatory response, that is initiated by the intestinal epithelial cells, propagated by innate immune cells and modulated by the cells of the adaptive immune response. This review focuses on cytokines, that are reported to orchestrate the induction and propagation of pro-inflammatory immune response, and also those that are involved in control and resolution of inflammation. We describe the functional roles of a number of cytokines in modulating anti-Campylobacter immune responses: 1. cytokines of innate immunity (TNF-α, IL-6, and IL-8) as initiators of inflammatory response, 2. cytokines of antigen-presenting cells (IL-1β, IL-12, and IL-23) as promoters of pro-inflammatory response, 3. cytokines produced by T cells (IFN-γ, IL-17, IL-22) as activators of T cells, and 4. anti-inflammatory cytokines (IL-4 and IL-10) as inhibitors of pro-inflammatory responses. We highlight the roles of cytokines as potential therapeutic agents that are under investigation. In the end, we pose several questions that remain unanswered in our quest to understand Campylobacter immunity.
Collapse
Affiliation(s)
- Nadia A Al-Banna
- Department of Basic Medical Sciences, College of Medicine, QU Health Cluster, Qatar University, Doha, Qatar.
| | - Farhan Cyprian
- Department of Basic Medical Sciences, College of Medicine, QU Health Cluster, Qatar University, Doha, Qatar.
| | - M John Albert
- Department of Microbiology, Faculty of Medicine, Kuwait University, Jabriya, Kuwait.
| |
Collapse
|
21
|
Korneev KV, Kondakova AN, Sviriaeva EN, Mitkin NA, Palmigiano A, Kruglov AA, Telegin GB, Drutskaya MS, Sturiale L, Garozzo D, Nedospasov SA, Knirel YA, Kuprash DV. Hypoacylated LPS from Foodborne Pathogen Campylobacter jejuni Induces Moderate TLR4-Mediated Inflammatory Response in Murine Macrophages. Front Cell Infect Microbiol 2018; 8:58. [PMID: 29535976 PMCID: PMC5835049 DOI: 10.3389/fcimb.2018.00058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/12/2018] [Indexed: 11/13/2022] Open
Abstract
Toll-like receptor 4 (TLR4) initiates immune response against Gram-negative bacteria upon specific recognition of lipid A moiety of lipopolysaccharide (LPS), the major component of their cell wall. Some natural differences between LPS variants in their ability to interact with TLR4 may lead to either insufficient activation that may not prevent bacterial growth, or excessive activation which may lead to septic shock. In this study we evaluated the biological activity of LPS isolated from pathogenic strain of Campylobacter jejuni, the most widespread bacterial cause of foodborne diarrhea in humans. With the help of hydrophobic chromatography and MALDI-TOF mass spectrometry we showed that LPS from a C. jejuni strain O2A consists of both hexaacyl and tetraacyl forms. Since such hypoacylation can result in a reduced immune response in humans, we assessed the activity of LPS from C. jejuni in mouse macrophages by measuring its capacity to activate TLR4-mediated proinflammatory cytokine and chemokine production, as well as NFκB-dependent reporter gene transcription. Our data support the hypothesis that LPS acylation correlates with its bioactivity.
Collapse
Affiliation(s)
- Kirill V. Korneev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Anna N. Kondakova
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina N. Sviriaeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Nikita A. Mitkin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Angelo Palmigiano
- CNR Institute for Polymers Composites and Biomaterials, Catania, Italy
| | - Andrey A. Kruglov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- German Rheumatism Research Center, Leibniz Institute, Berlin, Germany
| | - Georgy B. Telegin
- Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Russia
| | - Marina S. Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Luisa Sturiale
- CNR Institute for Polymers Composites and Biomaterials, Catania, Italy
| | - Domenico Garozzo
- CNR Institute for Polymers Composites and Biomaterials, Catania, Italy
| | - Sergei A. Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- German Rheumatism Research Center, Leibniz Institute, Berlin, Germany
| | - Yuriy A. Knirel
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry V. Kuprash
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
22
|
Kim S, Vela A, Clohisey SM, Athanasiadou S, Kaiser P, Stevens MP, Vervelde L. Host-specific differences in the response of cultured macrophages to Campylobacter jejuni capsule and O-methyl phosphoramidate mutants. Vet Res 2018; 49:3. [PMID: 29316981 PMCID: PMC5759256 DOI: 10.1186/s13567-017-0501-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 12/15/2017] [Indexed: 01/10/2023] Open
Abstract
Campylobacter jejuni is the leading cause of bacterial food-borne gastroenteritis worldwide and human infections are frequently associated with handling and consumption of contaminated poultry. The polysaccharide capsule of C. jejuni plays important roles in colonisation of the chicken gut, invasion of epithelial cells and serum resistance and is subject to modification with O-methyl phosphoramidate (MeOPN) in most strains. In this study, the cytokine responses of mouse bone marrow-derived macrophages (mBMMs), chicken bone marrow-derived macrophages (chBMMs) and human monocyte-derived macrophages (hMDMs) were measured following infection with C. jejuni 11168H wild-type (WT) or isogenic mutants lacking either the capsule (Δcj1439) or its MeOPN modification (Δcj1417). Consistent with previous observations using murine bone marrow-derived dendritic cells, mutants lacking the capsule or MeOPN elicited enhanced transcription of IL-6 and IL-10 in mBMMs compared to wild-type C. jejuni. However, the lack of capsule and MeOPN did not alter IL-6 and IL-10 expression in chBMMs and hMDMs compared to C. jejuni WT. Phagocytosis assays showed the acapsular mutant was not impaired in uptake or net intracellular survival after phagocytosis in both chicken and human macrophages; however, the phagocytosis of the MeOPN mutant was significantly decreased in both chicken and human macrophages. In conclusion, differences in the response of macrophages of varying host origin to Campylobacter were detected. The absence of MeOPN modification on the capsule of C. jejuni did not alter the levels of innate cytokine expression in both chicken and human macrophages compared to the 11168H WT, but affected phagocytosis by host macrophages.
Collapse
Affiliation(s)
- Sungwon Kim
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Andrea Vela
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Sara M Clohisey
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | | | - Pete Kaiser
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Mark P Stevens
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Lonneke Vervelde
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.
| |
Collapse
|
23
|
Campylobacter jejuni and associated immune mechanisms: short-term effects and long-term implications for infants in low-income countries. Curr Opin Infect Dis 2018; 30:322-328. [PMID: 28157786 DOI: 10.1097/qco.0000000000000364] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Campylobacter jejuni is recognized as one of the most common causes of food-borne gastrointestinal illness worldwide, resulting in a self-limiting dysentery in developed countries. However, it is increasingly gaining attention due to its association with postinfectious complications such as Guillain-Barré Syndrome and recently recognized importance in early childhood diarrhea in developing countries. We hypothesize that the inflammation mediated by C. jejuni infection causes environmental enteric dysfunction, and with contribution from diet and the host, microbiome may be responsible for growth faltering in children and developmental disability. RECENT FINDINGS Diet plays a major role in the impact of C. jejuni infection, both by availability of micronutrients for the bacteria and host as well as shaping the microbiome that affords resistance. Early childhood repeated exposure to the bacterium results in inflammation that affords long-term immunity but, in the short term, can lead to malabsorption, oral vaccine failure, cognitive delay and increased under-5 mortality. SUMMARY As interest in C. jejuni increases, our understanding of its virulence mechanisms has improved. However, much work remains to be done to fully understand the implications of immune-mediated inflammation and its potential role in diseases such as environmental enteric dysfunction.
Collapse
|
24
|
V K, E M, K B, R H, V R, D S, A K, M L, M L, A L, Z Š, M L. TLR4 and TLR21 expression, MIF, IFN-β, MD-2, CD14 activation, and sIgA production in chickens administered with EFAL41 strain challenged with Campylobacter jejuni. Folia Microbiol (Praha) 2016; 62:89-97. [PMID: 27696326 DOI: 10.1007/s12223-016-0475-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 09/21/2016] [Indexed: 01/26/2023]
Abstract
The protective effect of Enterococcus faecium EFAL41 on chicken's caecum in relation to the TLR (TLR4 and TLR21) activation and production of luminal IgA challenged with Campylobacter jejuni CCM6191 was assessed. The activation of MIF, IFN-β, MD-2 and CD14 was followed-up after bacterial infection. Day-old chicks (40) were divided into four groups (n = 10): control (C), E. faecium AL41 (EFAL41), C. jejuni (CJ) and combined E. faecium AL41+C. jejuni (EFAL41+CJ). Relative mRNA expression of TLR4, TLR21 and CD14 was upregulated in the probiotic strain and infected (combined) group on day 4 and 7 post infection (p.i.). The caecal relative MD-2 mRNA expression was upregulated on day 4 p.i. in the EFAL41+CJ and CJ groups. MIF and IFN-β reached the highest levels in the combined groups on day 7 p.i. The concentration of the sIgA in intestinal flush was upregulated in EFAL41+CJ group on day 4 p.i. The results demonstrated that E. faecium EFAL41 probiotic strain can modulate the TLRs expression and modify the activation of MIF, IFN-β, MD-2 and CD14 molecules in the chickens caecum challenged with C. jejuni CCM 6191. The counts of EFAL41 were sufficient and high, similarly the counts of enterococci in both, caecum and faeces but without reduction of Campylobacter counts.
Collapse
Affiliation(s)
- Karaffová V
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic.
| | - Marcinková E
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic
| | - Bobíková K
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic
| | - Herich R
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic
| | - Revajová V
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic
| | - Stašová D
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic
| | - Kavuľová A
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic
| | - Levkutová M
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic
| | - Levkut M
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic
| | - Lauková A
- Institute of Animal Physiology, Slovak Academy of Sciences, Soltésovej 4-6, 04001, Kosice, Slovak Republic
| | - Ševčíková Z
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic
| | - Levkut M
- University of Veterinary Medicine and Pharmacy, Komenského 73, 041 81, Košice, Slovak Republic.,Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10, Bratislava, Slovak Republic
| |
Collapse
|
25
|
The immunobiology of Campylobacter jejuni: Innate immunity and autoimmune diseases. Immunobiology 2015; 221:535-43. [PMID: 26709064 DOI: 10.1016/j.imbio.2015.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 12/05/2015] [Accepted: 12/06/2015] [Indexed: 12/26/2022]
Abstract
The Gram-negative bacterium Campylobacter jejuni causes gastroenteritis and Guillain-Barré syndrome in humans. Recent advances in the immunobiology of C. jejuni have been made. This review summarizes C. jejuni-binding innate receptors and highlights the role of innate immunity in autoimmune diseases. This human pathogen produces a variety of glycoconjugates, including human ganglioside-like determinants and multiple activators of Toll-like receptors (TLRs). Furthermore, C. jejuni targets MyD88, NLRP3 inflammasome, TIR-domain-containing adapter-inducing interferon-β (TRIF), sialic acid-binding immunoglobulin-like lectins (Siglecs), macrophage galactose-type lectin (MGL), and immunoglobulin-like receptors (TREM2, LMIR5/CD300b). The roles of these innate receptors and signaling molecules have been extensively studied. MyD88-mediated TLR activation or inflammasome-dependent IL-1β secretion is essential for autoimmune induction. TRIF mediates the production of type I interferons that promote humoral immune responses and immunoglobulin class-switching. Siglec-1 and Siglec-7 interact directly with gangliosides. Siglec-1 activation enhances phagocytosis and inflammatory responses. MGL internalizes GalNAc-containing glycoconjugates. TREM2 is well-known for its role in phagocytosis. LMIR5 recognizes C. jejuni components and endogenous sulfoglycolipids. Several lines of evidence from animal models of autoimmune diseases suggest that simultaneous activation of innate immunity in the presence of autoreactive lymphocytes or antigen mimicry may link C. jejuni to immunopathology.
Collapse
|
26
|
Toll-like receptors recognize distinct proteinase-resistant glycoconjugates in Campylobacter jejuni and Escherichia coli. Mol Immunol 2015; 64:195-203. [DOI: 10.1016/j.molimm.2014.11.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/20/2014] [Accepted: 11/25/2014] [Indexed: 12/18/2022]
|
27
|
Transcriptomic and proteomic analyses reveal key innate immune signatures in the host response to the gastrointestinal pathogen Campylobacter concisus. Infect Immun 2014; 83:832-45. [PMID: 25486993 DOI: 10.1128/iai.03012-14] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pathogenic species within the genus Campylobacter are responsible for a considerable burden on global health. Campylobacter concisus is an emergent pathogen that plays a role in acute and chronic gastrointestinal disease. Despite ongoing research on Campylobacter virulence mechanisms, little is known regarding the immunological profile of the host response to Campylobacter infection. In this study, we describe a comprehensive global profile of innate immune responses to C. concisus infection in differentiated THP-1 macrophages infected with an adherent and invasive strain of C. concisus. Using RNA sequencing (RNA-seq), quantitative PCR (qPCR), mass spectrometry, and confocal microscopy, we observed differential expression of pattern recognition receptors and robust upregulation of DNA- and RNA-sensing molecules. In particular, we observed IFI16 inflammasome assembly in C. concisus-infected macrophages. Global profiling of the transcriptome revealed the significant regulation of a total of 8,343 transcripts upon infection with C. concisus, which included the activation of key inflammatory pathways involving CREB1, NF-κB, STAT, and interferon regulatory factor signaling. Thirteen microRNAs and 333 noncoding RNAs were significantly regulated upon infection, including MIR221, which has been associated with colorectal carcinogenesis. This study represents a major advance in our understanding of host recognition and innate immune responses to infection by C. concisus.
Collapse
|
28
|
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
|
29
|
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: 61] [Impact Index Per Article: 5.5] [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
|
30
|
Huizinga R, van Rijs W, Bajramovic JJ, Kuijf ML, Laman JD, Samsom JN, Jacobs BC. Sialylation of Campylobacter jejuni endotoxin promotes dendritic cell-mediated B cell responses through CD14-dependent production of IFN-β and TNF-α. THE JOURNAL OF IMMUNOLOGY 2013; 191:5636-45. [PMID: 24166974 DOI: 10.4049/jimmunol.1301536] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Campylobacter jejuni is the most common bacterial cause of human gastroenteritis and often precedes development of Guillain-Barré syndrome (GBS), a life-threatening paralytic disease. The incorporation of the carbohydrate sialic acid into C. jejuni lipooligosaccharides (LOS) is associated with increased severity of gastroenteritis and with induction of GBS; however, the underlying mechanisms remain completely unknown. In this study, we demonstrate that sialic acids in C. jejuni endotoxin enhance the rapid production of IFN-β and TNF-α by human dendritic cells (DCs). Using neutralizing Abs and receptors it was shown that these DC-derived cytokines promote the proliferation of human mucosal B cells in a T cell-independent manner. The production of both IFN-β and TNF-α by DCs in response to LOS requires CD14, and the amplified response of DCs to sialylated C. jejuni LOS is CD14 dependent. Together, these results indicate that sialylation of C. jejuni LOS increases DC activation and promotes subsequent B cell responses through CD14-driven production of IFN-β and TNF-α. This enhanced DC/B cell response may explain the increased pathogenicity of sialylated C. jejuni and may be key to the initiation of B cell-mediated autoimmunity in GBS.
Collapse
Affiliation(s)
- Ruth Huizinga
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
31
|
Deciphering the contribution of human meningothelial cells to the inflammatory and antimicrobial response at the meninges. Infect Immun 2013; 81:4299-310. [PMID: 24002066 DOI: 10.1128/iai.00477-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have investigated the response of primary human meningothelial cells to Neisseria meningitidis. Through a transcriptome analysis, we provide a comprehensive examination of the response of meningothelial cells to bacterial infection. A wide range of chemokines are elicited which act to attract and activate the main players of innate and adaptive immunity. We showed that meningothelial cells expressed a high level of Toll-like receptor 4 (TLR4), and, using a gene silencing strategy, we demonstrated the contribution of this pathogen recognition receptor in meningothelial cell activation. Secretion of interleukin-6 (IL-6), CXCL10, and CCL5 was almost exclusively TLR4 dependent and relied on MyD88 and TRIF adaptor cooperation. In contrast, IL-8 induction was independent of the presence of TLR4, MyD88, and TRIF. Transcription factors NF-κB p65, p38 mitogen-activated protein kinase (MAPK), Jun N-terminal protein kinase (JNK1), IRF3, and IRF7 were activated after contact with bacteria. Interestingly, the protein kinase IRAK4 was found to play a minor role in the meningothelial cell response to Neisseria infection. Our work highlights the role of meningothelial cells in the development of an immune response and inflammation in the central nervous system (CNS) in response to meningococcal infection. It also sheds light on the complexity of intracellular signaling after TLR triggering.
Collapse
|
32
|
Rasmussen JJ, Vegge CS, Frøkiær H, Howlett RM, Krogfelt KA, Kelly DJ, Ingmer H. Campylobacter jejuni carbon starvation protein A (CstA) is involved in peptide utilization, motility and agglutination, and has a role in stimulation of dendritic cells. J Med Microbiol 2013; 62:1135-1143. [PMID: 23682166 DOI: 10.1099/jmm.0.059345-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Campylobacter jejuni is the most frequent cause of severe gastroenteritis in the developed world. The major symptom of campylobacteriosis is inflammatory diarrhoea. The molecular mechanisms of this infection are poorly understood compared to those of less frequent disease-causing pathogens. In a previous study, we identified C. jejuni proteins that antibodies in human campylobacteriosis patients reacted with. One of the immunogenic proteins identified (Cj0917) displays homology to carbon starvation protein A (CstA) from Escherichia coli, where this protein is involved in the starvation response and peptide uptake. In contrast to many bacteria, C. jejuni relies on amino acids and organic acids for energy, but in vivo it is highly likely that peptides are also utilized, although their mechanisms of uptake are unknown. In this study, Biolog phenotype microarrays have been used to show that a ΔcstA mutant has a reduced ability to utilize a number of di- and tri-peptides as nitrogen sources. This phenotype was restored through genetic complementation, suggesting CstA is a peptide uptake system in C. jejuni. Furthermore, the ΔcstA mutant also displayed reduced motility and reduced agglutination compared to WT bacteria; these phenotypes were also restored through complementation. Murine dendritic cells exposed to UV-killed bacteria showed a reduced IL-12 production, but the same IL-10 response when encountering C. jejuni ΔcstA compared to the WT strain. The greater Th1 stimulation elicited by the WT as compared to ΔcstA mutant cells indicates an altered antigenic presentation on the surface, and thus an altered recognition of the mutant. Thus, we conclude that C. jejuni CstA is important not only for peptide utilization, but also it may influence host-pathogen interactions.
Collapse
Affiliation(s)
- J J Rasmussen
- Department of Microbiology and Infection Control, Statens Serum Institut, 2300 Copenhagen S, Denmark
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - C S Vegge
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - H Frøkiær
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - R M Howlett
- Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - K A Krogfelt
- Department of Microbiology and Infection Control, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | - D J Kelly
- Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - H Ingmer
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| |
Collapse
|
33
|
Stephenson HN, John CM, Naz N, Gundogdu O, Dorrell N, Wren BW, Jarvis GA, Bajaj-Elliott M. Campylobacter jejuni lipooligosaccharide sialylation, phosphorylation, and amide/ester linkage modifications fine-tune human Toll-like receptor 4 activation. J Biol Chem 2013; 288:19661-72. [PMID: 23629657 DOI: 10.1074/jbc.m113.468298] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Campylobacter jejuni is a leading cause of acute gastroenteritis. C. jejuni lipooligosaccharide (LOS) is a potent activator of Toll-like receptor (TLR) 4-mediated innate immunity. Structural variations of the LOS have been previously reported in the oligosaccharide (OS) moiety, the disaccharide lipid A (LA) backbone, and the phosphorylation of the LA. Here, we studied LOS structural variation between C. jejuni strains associated with different ecological sources and analyzed their ability to activate TLR4 function. MALDI-TOF MS was performed to characterize structural variation in both the OS and LA among 15 different C. jejuni isolates. Cytokine induction in THP-1 cells and primary monocytes was correlated with LOS structural variation in each strain. Additionally, structural variation was correlated with the source of each strain. OS sialylation, increasing abundance of LA d-glucosamine versus 2,3-diamino-2,3-dideoxy-d-glucose, and phosphorylation status all correlated with TLR4 activation as measured in THP-1 cells and monocytes. Importantly, LOS-induced inflammatory responses were similar to those elicited by live bacteria, highlighting the prominent contribution of the LOS component in driving host immunity. OS sialylation status but not LA structure showed significant association with strains clustering with livestock sources. Our study highlights how variations in three structural components of C. jejuni LOS alter TLR4 activation and consequent monocyte activation.
Collapse
Affiliation(s)
- Holly N Stephenson
- Infectious Diseases and Microbiology Unit, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Al-Banna NA, Raghupathy R, Albert MJ. Induction of cytokines in different organs after intranasal inoculation of Campylobacter jejuni in mice. Gut Pathog 2012; 4:23. [PMID: 23244595 PMCID: PMC3531258 DOI: 10.1186/1757-4749-4-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 12/06/2012] [Indexed: 11/30/2022] Open
Abstract
Background Cytokine production and histopathological changes occur in the lungs of mice after intranasal inoculation with Campylobacter jejuni, but the levels of cytokines in different organs to which C. jejuni disseminates have not been studied. Findings Adult BALB/c mice were intranasally inoculated with C. jejuni 81–176 (test) or phosphate-buffered saline (control) (n=16 per group). The levels of cytokines in the organs (spleen, liver, and small and large intestines) to which C. jejuni disseminated were measured by ELISA. Two cytokine patterns were observed. First, increased proinflammatory cytokines, TNF-α, IL-1, and IL-2, were followed by anti-inflammatory cytokines, IL-4 and IL-10 in the spleen and large intestine. Second, in the liver and small intestine, there was a predominant production of anti-inflammatory cytokines, IL-4 and IL-10, with some increase in IL-2 levels. In the spleen and intestines, the levels of pro- and anti-inflammatory cytokines were concurrently increased. Conclusion Dissemination of C. jejuni is associated with the production of different cytokine profiles in different tissues, with the proinflammatory response appearing in the spleen and large intestine at an earlier time point than in the liver and small intestine. The organs produce different cytokine profiles in response to C. jejuni dissemination. These preliminary findings should be confirmed with a study involving a larger group of animals.
Collapse
Affiliation(s)
- Nadia A Al-Banna
- Department of Microbiology, Faculty of Medicine, Kuwait University, PO Box 24923, Safat 13110, Kuwait.
| | | | | |
Collapse
|
35
|
Zhang X, Beduhn M, Zheng X, Lian D, Chen D, Li R, Siu LKS, Marleau A, French PW, Ichim TE, Min WP. Induction of alloimmune tolerance in heart transplantation through gene silencing of TLR adaptors. Am J Transplant 2012; 12:2675-88. [PMID: 22823145 DOI: 10.1111/j.1600-6143.2012.04196.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Toll-like receptors (TLRs) activate biochemical pathways that evoke activation of innate immunity, which leads to dendritic cell (DC) maturation and initiation of adaptive immune responses that provoke allograft rejection. We aimed to prolong allograft survival by selectively inhibiting expression of the common adaptors of TLR signaling, namely MyD88 and TRIF, using siRNA. In vitro we demonstrated that blocking expression of MyD88 and TRIF led to reduced DC maturation. In vivo treatment of recipients with MyD88 and TRIF siRNA significantly prolonged allograft survival in the BALB/c > C57BL6 cardiac transplant model. Moreover, the combination of MyD88 and TRIF siRNA along with a low dose of rapamycin further extended the allograft survival (88.8 ± 7.1 days). Tissue histopathology demonstrated an overall reduction in lymphocyte interstitium infiltration, vascular obstruction and hemorrhage in mice treated with MyD88 and TRIF siRNA vector plus rapamycin. Furthermore, treatment was associated with an increase in the numbers of CD4(+) CD25(+) FoxP3(+) regulatory T cells and Th2 deviation. To our knowledge, this study is the first demonstration of prolonging the survival of allogeneic heart grafts through gene silencing of TLR signaling adaptors, highlighting the therapeutic potential of siRNA in clinical transplantation.
Collapse
Affiliation(s)
- X Zhang
- Departments of Surgery, Pathology, and Oncology, University of Western Ontario, London, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Otto B, Haag LM, Fischer A, Plickert R, Kühl AA, Göbel UB, Heimesaat MM, Bereswill S. Campylobacter jejuni induces extra-intestinal immune responses via Toll-like-receptor-4 signaling in conventional IL-10 deficient mice with chronic colitis. Eur J Microbiol Immunol (Bp) 2012; 2:210-9. [PMID: 24688768 PMCID: PMC3962757 DOI: 10.1556/eujmi.2.2012.3.7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 06/27/2012] [Indexed: 11/19/2022] Open
Abstract
Campylobacter jejuni is one of the predominant causes for foodborne bacterial infections worldwide. We investigated whether signaling of C. jejuni-lipoproteins and -lipooligosaccharide via Toll-like-receptor (TLR) -2 and -4, respectively, is inducing intestinal and extra-intestinal immune responses following infection of conventional IL-10(-/-) mice with chronic colitis. At day 3 following oral infection, IL-10(-/-) mice lacking TLR-2 or TLR-4 harbored comparable C. jejuni strain ATCC 43431 loads in their colon. Interestingly, infected TLR-4(-/-) IL-10(-/-) mice displayed less compromized epithelial barrier function as indicated by lower translocation rates of live gut commensals into mesenteric lymphnodes (MLNs), and exhibited less distinct B lymphocyte responses in their colonic mucosa as compared to naїve IL-10(-/-) controls. Furthermore, in extra-intestinal compartments such as MLNs and spleens, abundance of myeloid cells was less distinct whereas relative percentages of activated T helper cells and cytotoxic T cells were higher in spleens and dendritic cells more abundant in MLNs of infected IL-10(-/-) animals lacking TLR-4 as compared to IL-10(-/-) controls. Taken together, in conventionally colonized IL-10(-/-) mice, TLR-4, but not TLR-2, is involved in mediating extra-intestinal pro-inflammatory immune responses following C. jejuni infection. Thus, conventional IL-10(-/-) mice are well suited to further dissect mechanisms underlying Campylobacter infections in vivo.
Collapse
Affiliation(s)
- B. Otto
- Department of Microbiology and Hygiene, Charité – University
Medicine BerlinBerlinGermany
| | - L.-M. Haag
- Department of Microbiology and Hygiene, Charité – University
Medicine BerlinBerlinGermany
| | - A. Fischer
- Department of Microbiology and Hygiene, Charité – University
Medicine BerlinBerlinGermany
| | - R. Plickert
- Department of Microbiology and Hygiene, Charité – University
Medicine BerlinBerlinGermany
| | - A. A. Kühl
- Department of Internal Medicine, Rheumatology and Clinical
Immunology / Research Center Immuno-Sciences (RCIS), Charité – University
Medicine BerlinBerlinGermany
| | - U. B. Göbel
- Department of Microbiology and Hygiene, Charité – University
Medicine BerlinBerlinGermany
| | - M. M. Heimesaat
- Department of Microbiology and Hygiene, Charité – University
Medicine BerlinBerlinGermany
| | - S. Bereswill
- Department of Microbiology and Hygiene, Charité – University
Medicine BerlinBerlinGermany
| |
Collapse
|
37
|
Teo I, Toms SM, Marteyn B, Barata TS, Simpson P, Johnston KA, Schnupf P, Puhar A, Bell T, Tang C, Zloh M, Matthews S, Rendle PM, Sansonetti PJ, Shaunak S. Preventing acute gut wall damage in infectious diarrhoeas with glycosylated dendrimers. EMBO Mol Med 2012; 4:866-81. [PMID: 22887873 PMCID: PMC3491821 DOI: 10.1002/emmm.201201290] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 06/15/2012] [Accepted: 06/22/2012] [Indexed: 01/25/2023] Open
Abstract
Intestinal pathogens use the host's excessive inflammatory cytokine response, designed to eliminate dangerous bacteria, to disrupt epithelial gut wall integrity and promote their tissue invasion. We sought to develop a non-antibiotic-based approach to prevent this injury. Molecular docking studies suggested that glycosylated dendrimers block the TLR4-MD-2-LPS complex, and a 13.6 kDa polyamidoamine (PAMAM) dendrimer glucosamine (DG) reduced the induction of human monocyte interleukin (IL)-6 by Gram-negative bacteria. In a rabbit model of shigellosis, PAMAM-DG prevented epithelial gut wall damage and intestinal villous destruction, reduced local IL-6 and IL-8 expression, and minimized bacterial invasion. Computational modelling studies identified a 3.3 kDa polypropyletherimine (PETIM)-DG as the smallest likely bioactive molecule. In human monocytes, high purity PETIM-DG potently inhibited Shigella Lipid A-induced IL-6 expression. In rabbits, PETIM-DG prevented Shigella-induced epithelial gut wall damage, reduced local IL-6 and IL-8 expression, and minimized bacterial invasion. There was no change in β-defensin, IL-10, interferon-β, transforming growth factor-β, CD3 or FoxP3 expression. Small and orally delivered DG could be useful for preventing gut wall tissue damage in a wide spectrum of infectious diarrhoeal diseases. –>See accompanying article http://dx.doi.org/10.1002/emmm.201201668
Collapse
Affiliation(s)
- Ian Teo
- Departments of Medicine, Infectious Diseases & Immunity, Imperial College London, Hammersmith Hospital, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Connell S, Meade KG, Allan B, Lloyd AT, Kenny E, Cormican P, Morris DW, Bradley DG, O'Farrelly C. Avian resistance to Campylobacter jejuni colonization is associated with an intestinal immunogene expression signature identified by mRNA sequencing. PLoS One 2012; 7:e40409. [PMID: 22870198 PMCID: PMC3411578 DOI: 10.1371/journal.pone.0040409] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 06/06/2012] [Indexed: 12/12/2022] Open
Abstract
Campylobacter jejuni is the most common cause of human bacterial gastroenteritis and is associated with several post-infectious manifestations, including onset of the autoimmune neuropathy Guillain-Barré syndrome, causing significant morbidity and mortality. Poorly-cooked chicken meat is the most frequent source of infection as C. jejuni colonizes the avian intestine in a commensal relationship. However, not all chickens are equally colonized and resistance seems to be genetically determined. We hypothesize that differences in immune response may contribute to variation in colonization levels between susceptible and resistant birds. Using high-throughput sequencing in an avian infection model, we investigate gene expression associated with resistance or susceptibility to colonization of the gastrointestinal tract with C. jejuni and find that gut related immune mechanisms are critical for regulating colonization. Amongst a single population of 300 4-week old chickens, there was clear segregation in levels of C. jejuni colonization 48 hours post-exposure. RNAseq analysis of caecal tissue from 14 C. jejuni-susceptible and 14 C. jejuni-resistant birds generated over 363 million short mRNA sequences which were investigated to identify 219 differentially expressed genes. Significantly higher expression of genes involved in the innate immune response, cytokine signaling, B cell and T cell activation and immunoglobulin production, as well as the renin-angiotensin system was observed in resistant birds, suggesting an early active immune response to C. jejuni. Lower expression of these genes in colonized birds suggests suppression or inhibition of a clearing immune response thus facilitating commensal colonization and generating vectors for zoonotic transmission. This study describes biological processes regulating C. jejuni colonization of the avian intestine and gives insight into the differential immune mechanisms incited in response to commensal bacteria in general within vertebrate populations. The results reported here illustrate how an exaggerated immune response may be elicited in a subset of the population, which alters host-microbe interactions and inhibits the commensal state, therefore having wider relevance with regard to inflammatory and autoimmune disease.
Collapse
Affiliation(s)
- Sarah Connell
- Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, Ireland.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Klaas M, Oetke C, Lewis LE, Erwig LP, Heikema AP, Easton A, Willison HJ, Crocker PR. Sialoadhesin promotes rapid proinflammatory and type I IFN responses to a sialylated pathogen, Campylobacter jejuni. THE JOURNAL OF IMMUNOLOGY 2012; 189:2414-22. [PMID: 22851711 DOI: 10.4049/jimmunol.1200776] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sialoadhesin (Sn) is a macrophage (Mφ)-restricted receptor that recognizes sialylated ligands on host cells and pathogens. Although Sn is thought to be important in cellular interactions of Mφs with cells of the immune system, the functional consequences of pathogen engagement by Sn are unclear. As a model system, we have investigated the role of Sn in Mφ interactions with heat-killed Campylobacter jejuni expressing a GD1a-like, sialylated glycan. Compared to Sn-expressing bone marrow-derived macrophages (BMDM) from wild-type mice, BMDM from mice either deficient in Sn or expressing a non-glycan-binding form of Sn showed greatly reduced phagocytosis of sialylated C. jejuni. This was accompanied by a strong reduction in MyD88-dependent secretion of TNF-α, IL-6, IL-12, and IL-10. In vivo studies demonstrated that functional Sn was required for rapid TNF-α and IFN-β responses to i.v.-injected sialylated C. jejuni. Bacteria were captured within minutes after i.v. injection and were associated with Mφs in both liver and spleen. In the spleen, IFN-β-reactive cells were localized to Sn⁺ Mφs and other cells in the red pulp and marginal zone. Together, these studies demonstrate that Sn plays a key role in capturing sialylated pathogens and promoting rapid proinflammatory cytokine and type I IFN responses.
Collapse
Affiliation(s)
- Mariliis Klaas
- Division of Cell Signalling and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Campylobacter jejuni induces acute enterocolitis in gnotobiotic IL-10-/- mice via Toll-like-receptor-2 and -4 signaling. PLoS One 2012; 7:e40761. [PMID: 22808254 PMCID: PMC3393706 DOI: 10.1371/journal.pone.0040761] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 06/12/2012] [Indexed: 12/31/2022] Open
Abstract
Background Campylobacter jejuni is a leading cause of foodborne bacterial enterocolitis worldwide. Investigation of immunopathology is hampered by a lack of suitable vertebrate models. We have recently shown that gnotobiotic mice as well as conventional IL-10−/− animals are susceptible to C. jejuni infection and develop intestinal immune responses. However, clinical symptoms of C. jejuni infection were rather subtle and did not reflect acute bloody diarrhea seen in human campylobacteriosis. Methodology/Principal Findings In order to overcome these limitations we generated gnotobiotic IL-10−/− mice by quintuple antibiotic treatment starting right after weaning. The early treatment was essential to prevent these animals from chronic colitis. Following oral infection C. jejuni colonized the gastrointestinal tract at high levels and induced acute enterocolitis within 7 days as indicated by bloody diarrhea and pronounced histopathological changes of the colonic mucosa. Immunopathology was further characterized by increased numbers of apoptotic cells, regulatory T-cells, T- and B-lymphocytes as well as elevated TNF-α, IFN-γ, and MCP-1 concentrations in the inflamed colon. The induction of enterocolitis was specific for C. jejuni given that control animals infected with a commensal E. coli strain did not display any signs of disease. Most strikingly, intestinal immunopathology was ameliorated in mice lacking Toll-like-receptors-2 or -4 indicating that C. jejuni lipoproteins and lipooligosaccharide are essential for induction and progression of immunopathology. Conclusion/Significance Gnotobiotic IL-10−/− mice develop acute enterocolitis following C. jejuni infection mimicking severe episodes of human campylobacteriosis and are thus well suited to further dissect mechanisms underlying Campylobacter infections in vivo.
Collapse
|
41
|
Gentle ME, Rose A, Bugeon L, Dallman MJ. Noncanonical Notch signaling modulates cytokine responses of dendritic cells to inflammatory stimuli. THE JOURNAL OF IMMUNOLOGY 2012; 189:1274-84. [PMID: 22753939 DOI: 10.4049/jimmunol.1103102] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dendritic cell (DC)-derived cytokines play a key role in specifying adaptive immune responses tailored to the type of pathogen encountered and the local tissue environment. However, little is known about how DCs perceive the local environment. We investigated whether endogenous Notch signaling could affect DC responses to pathogenic stimuli. We demonstrate that concurrent Notch and TLR stimulation results in a unique cytokine profile in mouse bone-marrow derived DCs characterized by enhanced IL-10 and IL-2, and reduced IL-12 expression compared with TLR ligation alone. Unexpectedly, modulation of cytokine production occurred through a noncanonical Notch signaling pathway, independent of γ-secretase activity. Modulation required de novo protein synthesis, and PI3K, JNK, and ERK activity were necessary for enhanced IL-2 expression, whereas modulation of IL-10 required only PI3K activity. Further, we show that this γ-secretase-independent Notch pathway can induce PI3K activity. In contrast, expression of the canonical Notch target gene Hes1 was suppressed in DCs stimulated with Notch and TLR ligands simultaneously. Thus, our data suggest that Notch acts as an endogenous signal that modulates cytokine expression of DCs through a noncanonical pathway and therefore has the potential to tailor the subsequent adaptive immune response in a tissue- and/or stage-dependent manner.
Collapse
Affiliation(s)
- Madeleine E Gentle
- Division of Cell and Molecular Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | | | | | | |
Collapse
|
42
|
Huizinga R, Easton AS, Donachie AM, Guthrie J, van Rijs W, Heikema A, Boon L, Samsom JN, Jacobs BC, Willison HJ, Goodyear CS. Sialylation of Campylobacter jejuni lipo-oligosaccharides: impact on phagocytosis and cytokine production in mice. PLoS One 2012; 7:e34416. [PMID: 22470569 PMCID: PMC3314637 DOI: 10.1371/journal.pone.0034416] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 02/28/2012] [Indexed: 11/27/2022] Open
Abstract
Background Guillain-Barré syndrome (GBS) is a post-infectious polyradiculoneuropathy, frequently associated with antecedent Campylobacter jejuni (C. jejuni) infection. The presence of sialic acid on C. jejuni lipo-oligosaccharide (LOS) is considered a risk factor for development of GBS as it crucially determines the structural homology between LOS and gangliosides, explaining the induction of cross-reactive neurotoxic antibodies. Sialylated C. jejuni are recognised by TLR4 and sialoadhesin; however, the functional implications of these interactions in vivo are unknown. Methodology/Principal Findings In this study we investigated the effects of bacterial sialylation on phagocytosis and cytokine secretion by mouse myeloid cells in vitro and in vivo. Using fluorescently labelled GM1a/GD1a ganglioside-mimicking C. jejuni strains and corresponding (Cst-II-mutant) control strains lacking sialic acid, we show that sialylated C. jejuni was more efficiently phagocytosed in vitro by BM-MΦ, but not by BM-DC. In addition, LOS sialylation increased the production of IL-10, IL-6 and IFN-β by both BM-MΦ and BM-DC. Subsequent in vivo experiments revealed that sialylation augmented the deposition of fluorescent bacteria in splenic DC, but not macrophages. In addition, sialylation significantly amplified the production of type I interferons, which was independent of pDC. Conclusions/Significance These results identify novel immune stimulatory effects of C. jejuni sialylation, which may be important in inducing cross-reactive humoral responses that cause GBS.
Collapse
Affiliation(s)
- Ruth Huizinga
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alistair S. Easton
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Anne M. Donachie
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jim Guthrie
- Department of Bacteriology, Southern General Hospital, Glasgow, United Kingdom
| | - Wouter van Rijs
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Astrid Heikema
- Department of Microbiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Janneke N. Samsom
- Division Gastroenterology and Nutrition, Department of Paediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bart C. Jacobs
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hugh J. Willison
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Carl S. Goodyear
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
The Campylobacter jejuni NCTC11168 capsule prevents excessive cytokine production by dendritic cells. Med Microbiol Immunol 2011; 201:137-44. [PMID: 21863342 DOI: 10.1007/s00430-011-0214-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Indexed: 02/06/2023]
Abstract
Campylobacter jejuni is the leading cause of human bacterial gastroenteritis worldwide, mainly caused by handling and consumption of contaminated poultry. However, the immune response to infection is poorly understood. Here, the impact of the C. jejuni capsule, flagella and the N-linked glycosylation system on cytokine production by dendritic cells was investigated. Bone marrow-derived murine dendritic cells (BMDCs) infected with C. jejuni lacking the N-linked glycosylation system produced similar amounts of cytokines compared to cells infected with C. jejuni 11168H wild-type (WT) cultures. C. jejuni flagellin FlaA mutants elicited reduced IL-6 and IL-10 production in BMDCs compared to C. jejuni WT and this reduction was more pronounced in TLR4(-/-) BMDCs. An acapsular C. jejuni mutant as well as a mutant lacking the O-methyl phosphoramidate modification of the capsule elicited a higher cytokine response in BMDCs. Experiments with TLR4(-/-) BMDCs revealed that this increased cytokine production was not solely dependent on signalling through TLR4. Therefore, the C. jejuni capsule is important to prevent excessive cytokine production by BMDCs and even minor changes in capsule composition such as the lack of the O-methyl phosphoramidate modification can lead to increased cytokine production.
Collapse
|
44
|
Novel murine infection models provide deep insights into the "ménage à trois" of Campylobacter jejuni, microbiota and host innate immunity. PLoS One 2011; 6:e20953. [PMID: 21698299 PMCID: PMC3115961 DOI: 10.1371/journal.pone.0020953] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 05/13/2011] [Indexed: 12/11/2022] Open
Abstract
Background Although Campylobacter jejuni-infections have a high prevalence worldwide and represent a significant socioeconomic burden, it is still not well understood how C. jejuni causes intestinal inflammation. Detailed investigation of C. jejuni-mediated intestinal immunopathology is hampered by the lack of appropriate vertebrate models. In particular, mice display colonization resistance against this pathogen. Methodology/Principal Findings To overcome these limitations we developed a novel C. jejuni-infection model using gnotobiotic mice in which the intestinal flora was eradicated by antibiotic treatment. These animals could then be permanently associated with a complete human (hfa) or murine (mfa) microbiota. After peroral infection C. jejuni colonized the gastrointestinal tract of gnotobiotic and hfa mice for six weeks, whereas mfa mice cleared the pathogen within two days. Strikingly, stable C. jejuni colonization was accompanied by a pro-inflammatory immune response indicated by increased numbers of T- and B-lymphocytes, regulatory T-cells, neutrophils and apoptotic cells, as well as increased concentrations of TNF-α, IL-6, and MCP-1 in the colon mucosa of hfa mice. Analysis of MyD88−/−, TRIF−/−, TLR4−/−, and TLR9−/− mice revealed that TLR4- and TLR9-signaling was essential for immunopathology following C. jejuni-infection. Interestingly, C. jejuni-mutant strains deficient in formic acid metabolism and perception induced less intestinal immunopathology compared to the parental strain infection. In summary, the murine gut flora is essential for colonization resistance against C. jejuni and can be overcome by reconstitution of gnotobiotic mice with human flora. Detection of C. jejuni-LPS and -CpG-DNA by host TLR4 and TLR9, respectively, plays a key role in immunopathology. Finally, the host immune response is tightly coupled to bacterial formic acid metabolism and invasion fitness. Conclusion/Significance We conclude that gnotobiotic and “humanized” mice represent excellent novel C. jejuni-infection and -inflammation models and provide deep insights into the immunological and molecular interplays between C. jejuni, microbiota and innate immunity in human campylobacteriosis.
Collapse
|
45
|
Campylobacter jejuni lipooligosaccharides modulate dendritic cell-mediated T cell polarization in a sialic acid linkage-dependent manner. Infect Immun 2011; 79:2681-9. [PMID: 21502591 DOI: 10.1128/iai.00009-11] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Carbohydrate mimicry between Campylobacter jejuni lipooligosaccharides (LOS) and host neural gangliosides plays a crucial role in the pathogenesis of Guillain-Barré syndrome (GBS). Campylobacter jejuni LOS may mimic various gangliosides, which affects the immunogenicity and the type of neurological deficits in GBS patients. Previous studies have shown the interaction of LOS with sialic acid-specific siglec receptors, although the functional consequences remain unknown. Cells that express high levels of siglecs include dendritic cells (DCs), which are crucial for initiation and differentiation of immune responses. We confirm that α2,3-sialylated GD1a/GM1a mimic and α2,8-sialylated GD1c mimic LOS structures interact with recombinant Sn and siglec-7, respectively. Although the linkage of the terminal sialic acid of LOS did not regulate expression of DC maturation markers, it displayed clear opposite expression levels of interleukin-12 (IL-12) and OX40L, molecules involved in DC-mediated Th cell differentiation. Accordingly, targeting DC-expressed siglec-7 with α2,8-linked sialylated LOS resulted in Th1 responses, whereas Th2 responses were induced by targeting with LOS containing α2,3-linked sialic acid. Thus, our data demonstrate for the first time that depending on the sialylated composition of Campylobacter jejuni LOS, specific Th differentiation programs are initiated, possibly through targeting of distinct DC-expressed siglecs.
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW The incidence of gastrointestinal infections continues to increase and infectious colitis contributes to significant morbidity and mortality worldwide. The purpose of this review is to highlight the recent advances in knowledge of pathogens causing infectious colitis. We describe the various pathogens and specifically focus on enterohemorrhagic Escherichia coli (EHEC) O157:H7, Salmonella, Shigella, Campylobacter, and Entamoeba histolytica infections, and their impact on long-term effects, including postinfectious irritable bowel syndrome and inflammatory bowel disease. RECENT FINDINGS Salmonella, Campylobacter, and EHEC outbreaks continue to occur with disturbing regularity. Peanut butter and peppers were recently responsible for outbreaks of nontyphoid Salmonella. Recent research has identified Salmonella genes required for colonization of various hosts and transposon-mediated differential hybridization was recently used to identify genes required during infection in different animal models. A number of other strains of EHEC in addition to O157:H7 are emerging as serious threats to food safety in the USA. Campylobacter jejuni isolates are of interest because of absence of genes encoding for classical enterotoxins, and lack of plasmids encoding genes promoting bacterial invasion. Recent research has identified that the organism is able to invade and replicate in infected epithelia via Toll-like receptor (TLR)-2 and TLR-4. Also patients with infectious colitis, in particular Salmonella and Campylobacter, are at increased risk of postinfectious irritable bowel syndrome and inflammatory bowel disease on long-term follow-up. The paradigm of Entamoeba histolytica infection is changing with recent reports of detection of E. dispar deoxyribonucleic acid sequences, previously considered nonpathogenic. SUMMARY There has been an explosion in the understanding of the epidemiology, pathobiology, and mechanisms underlying infectious colitis. Additional studies to address prevention strategies and strict screening modalities for these infections are necessary.
Collapse
|
47
|
Edwards LA, Nistala K, Mills DC, Stephenson HN, Zilbauer M, Wren BW, Dorrell N, Lindley KJ, Wedderburn LR, Bajaj-Elliott M. Delineation of the innate and adaptive T-cell immune outcome in the human host in response to Campylobacter jejuni infection. PLoS One 2010; 5:e15398. [PMID: 21085698 PMCID: PMC2976761 DOI: 10.1371/journal.pone.0015398] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 09/04/2010] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Campylobacter jejuni is the most prevalent cause of bacterial gastroenteritis worldwide. Despite the significant health burden this infection presents, molecular understanding of C. jejuni-mediated disease pathogenesis remains poorly defined. Here, we report the characterisation of the early, innate immune response to C. jejuni using an ex-vivo human gut model of infection. Secondly, impact of bacterial-driven dendritic cell activation on T-cell mediated immunity was also sought. METHODOLOGY Healthy, control paediatric terminal ileum or colonic biopsy tissue was infected with C. jejuni for 8-12 hours. Bacterial colonisation was followed by confocal microscopy and mucosal innate immune responses measured by ELISA. Marked induction of IFNγ with modest increase in IL-22 and IL-17A was noted. Increased mucosal IL-12, IL-23, IL-1β and IL-6 were indicative of a cytokine milieu that may modulate subsequent T-cell mediated immunity. C. jejuni-driven human monocyte-derived dendritic cell activation was followed by analyses of T cell immune responses utilising flow cytometry and ELISA. Significant increase in Th-17, Th-1 and Th-17/Th-1 double-positive cells and corresponding cytokines was observed. The ability of IFNγ, IL-22 and IL-17 cytokines to exert host defence via modulation of C. jejuni adhesion and invasion to intestinal epithelia was measured by standard gentamicin protection assay. CONCLUSIONS Both innate and adaptive T cell-immunity to C. jejuni infection led to the release of IFNγ, IL-22 and IL-17A; suggesting a critical role for this cytokine triad in establishing host anti-microbial immunity during the acute and effectors phase of infection. In addition, to their known anti-microbial functions; IL-17A and IL-17F reduced the number of intracellular C. jejuni in intestinal epithelia, highlighting a novel aspect of how IL-17 family members may contribute to protective immunity against C. jejuni.
Collapse
Affiliation(s)
- Lindsey A. Edwards
- Infectious Diseases and Microbiology, Institute of Child Health, London, United Kingdom
| | - Kiran Nistala
- Rheumatology, Institute of Child Health, London, United Kingdom
| | - Dominic C. Mills
- Pathogen Molecular Biology Department, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Holly N. Stephenson
- Infectious Diseases and Microbiology, Institute of Child Health, London, United Kingdom
| | - Matthias Zilbauer
- Infectious Diseases and Microbiology, Institute of Child Health, London, United Kingdom
- Paediatric Gastroenterology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Brendan W. Wren
- Pathogen Molecular Biology Department, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Nick Dorrell
- Pathogen Molecular Biology Department, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Keith J. Lindley
- Autoimmunity and Surgery Units, Institute of Child Health, London, United Kingdom
| | | | - Mona Bajaj-Elliott
- Infectious Diseases and Microbiology, Institute of Child Health, London, United Kingdom
| |
Collapse
|
48
|
Hu Z, Qiu L, Xiao Z, Wang J, Yu Q, Li J, Feng H, Guo C, Zhang J. Effects of esculentoside A on autoimmune syndrome induced by Campylobacter jejuni in mice and its modulation on T-lymphocyte proliferation and apoptosis. Int Immunopharmacol 2010; 10:65-71. [DOI: 10.1016/j.intimp.2009.09.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 09/08/2009] [Accepted: 09/30/2009] [Indexed: 11/30/2022]
|
49
|
Abstract
PURPOSE OF REVIEW The aim of this review is to highlight recent advances in knowledge of bacterial enteric infections. We focus on understanding of enterohemorrhagic Escherichia coli O157:H7 and Campylobacter jejuni infections, and to link these acute events with long-term consequences in a susceptible host, including irritable bowel syndrome and chronic inflammatory bowel diseases. RECENT FINDINGS Enterohemorrhagic E. coli and C. jejuni are zoonotic infections that are acquired from exposure to tainted food (undercooked hamburger and chicken, respectively) and contaminated drinking water. Noninvasive E. coli O157:H7 elaborates Shiga-like toxins and protein effectors that are injected, via a molecular syringe that is encoded by a bacterial type 3 secretion system, into infected eukaryotic cells. Less is known about the precise virulence properties of enteroinvasive Campylobacter strains, but both enteric pathogens are able to disrupt polarized epithelial monolayers resulting in increased uptake of macromolecules and antigens. SUMMARY An improved understanding of the epidemiology, pathobiology and mechanisms underlying infectious enterocolitides will provide the basis for developing new intervention strategies including, for example, the use of probiotics, to interrupt the infectious process.
Collapse
Affiliation(s)
- Philip M Sherman
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | | | | |
Collapse
|
50
|
Abstract
Campylobacter infection in humans is accompanied by severe inflammation of the intestinal mucosa, in contrast to colonization of chicken. The basis for the differential host response is unknown. Toll-like receptors (TLRs) sense and respond to microbes in the body and participate in the induction of an inflammatory response. Thus far, the interaction of Campylobacter with chicken TLRs has not been studied. Here, we investigated the potential of four Campylobacter strains to activate human TLR1/2/6, TLR4, TLR5, and TLR9 and chicken TLR2t2/16, TLR4, TLR5, and TLR21. Live bacteria showed no or very limited potential to activate TLR2, TLR4, and TLR5 of both the human and chicken species, with minor but significant differences between Campylobacter strains. In contrast, lysed bacteria induced strong NF-kappaB activation through human TLR1/2/6 and TLR4 and chicken TLR2t2/16 and TLR4 but not via TLR5 of either species. Interestingly, C. jejuni induced TLR4-mediated beta interferon in human but not chicken cells. Furthermore, isolated chromosomal Campylobacter DNA was unable to activate human TLR9 in our system, whereas chicken TLR21 was activated by DNA from all of the campylobacters tested. Our data are the first comparison of TLR-induced immune responses in humans and chickens. The results suggest that differences in bacterial cell wall integrity and in TLR responses to Campylobacter LOS and/or DNA may contribute to the distinct clinical manifestation between the species.
Collapse
|