1
|
Brudvig JM, Cluett MM, Gensterblum-Miller EU, Chen J, Bell JA, Mansfield LS. Th1/Th17-mediated Immunity and Protection from Peripheral Neuropathy in Wildtype and IL10 -/- BALB/c Mice Infected with a Guillain-Barré Syndrome-associated Campylobacter jejuni Strain. Comp Med 2022; 72:63-77. [PMID: 35272743 PMCID: PMC9084571 DOI: 10.30802/aalas-cm-21-000059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/07/2021] [Accepted: 09/03/2021] [Indexed: 11/05/2022]
Abstract
Campylobacter jejuni is an important cause of bacterial gastroenteritis worldwide and is linked to Guillain-Barré syndrome (GBS), a debilitating postinfectious polyneuropathy. The immunopathogenesis of GBS involves the generation of antibodies that are cross reactive to C. jejuni lipooligosaccharide and structurally similar peripheral nerve gangliosides. Both the C. jejuni infecting strain and host factors contribute to GBS development. GBS pathogenesis is associated with Th2-mediated responses in patients. Moreover, induction of IgG1 antiganglioside antibodies in association with colonic Th2-mediated immune responses has been reported in C. jejuni-infected C57BL/6 IL10-/- mice at 4 to 6 wk after infection. We hypothesized that, due to their Th2 immunologic bias, BALB/c mice would develop autoantibodies and signs of peripheral neuropathy after infection with a GBS patient-derived strain of C. jejuni (strain 260.94). WT and IL10-/- BALB/c mice were orally inoculated with C. jejuni 260.94, phenotyped weekly for neurologic deficits, and euthanized after 5 wk. Immune responses were assessed as C. jejuni-specific and antiganglioside antibodies in plasma and cytokine production and histologic lesions in the proximal colon. Peripheral nerve lesions were assessed in dorsal root ganglia and their afferent nerve fibers by scoring immunohistochemically labeled macrophages through morphometry. C. jejuni 260.94 stably colonized both WT and IL10-/- mice and induced systemic Th1/Th17-mediated immune responses with significant increases in C. jejuni-specific IgG2a, IgG2b, and IgG3 plasma antibodies. However, C. jejuni 260.94 did not induce IgG1 antiganglioside antibodies, colitis, or neurologic deficits or peripheral nerve lesions in WT or IL10-/- mice. Both WT and IL10-/- BALB/c mice showed relative protection from development of Th2-mediated immunity and antiganglioside antibodies as compared with C57BL/6 IL10-/- mice. Therefore, BALB/c mice infected with C. jejuni 260.94 are not an effective disease model but provide the opportunity to study the role of immune mechanisms and host genetic background in the susceptibility to post infectious GBS.
Collapse
Affiliation(s)
- Jean M Brudvig
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Matthew M Cluett
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Elizabeth U Gensterblum-Miller
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michiga
| | - James Chen
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michiga
| | - Julia A Bell
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michiga
| | - Linda S Mansfield
- Comparative Enteric Diseases Laboratory, Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan; Comparative Medicine and Integrative Biology, Michigan State University, East Lansing, Michigan; College of Veterinary Medicine, Michigan State University, East Lansing, Michigan; Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan;,
| |
Collapse
|
2
|
Quantitative trait loci and transcriptome signatures associated with avian heritable resistance to Campylobacter. Sci Rep 2021; 11:1623. [PMID: 33436657 PMCID: PMC7804197 DOI: 10.1038/s41598-020-79005-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Campylobacter is the leading cause of bacterial foodborne gastroenteritis worldwide. Handling or consumption of contaminated poultry meat is a key risk factor for human campylobacteriosis. One potential control strategy is to select poultry with increased resistance to Campylobacter. We associated high-density genome-wide genotypes (600K single nucleotide polymorphisms) of 3000 commercial broilers with Campylobacter load in their caeca. Trait heritability was modest but significant (h2 = 0.11 ± 0.03). Results confirmed quantitative trait loci (QTL) on chromosomes 14 and 16 previously identified in inbred chicken lines, and detected two additional QTLs on chromosomes 19 and 26. RNA-Seq analysis of broilers at the extremes of colonisation phenotype identified differentially transcribed genes within the QTL on chromosome 16 and proximal to the major histocompatibility complex (MHC) locus. We identified strong cis-QTLs located within MHC suggesting the presence of cis-acting variation in MHC class I and II and BG genes. Pathway and network analyses implicated cooperative functional pathways and networks in colonisation, including those related to antigen presentation, innate and adaptive immune responses, calcium, and renin–angiotensin signalling. While co-selection for enhanced resistance and other breeding goals is feasible, the frequency of resistance-associated alleles was high in the population studied and non-genetic factors significantly influenced Campylobacter colonisation.
Collapse
|
3
|
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
|
4
|
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
|
5
|
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: 25] [Impact Index Per Article: 3.6] [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
|
6
|
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
|
7
|
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
|
8
|
Biomarkers of Gastrointestinal Host Responses to Microbial Infections. Mol Microbiol 2016. [DOI: 10.1128/9781555819071.ch46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
9
|
Papadopoulos A, Gorvel JP. Subversion of mouse dendritic cell subset function by bacterial pathogens. Microb Pathog 2015; 89:140-9. [PMID: 26453826 DOI: 10.1016/j.micpath.2015.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/24/2015] [Accepted: 10/04/2015] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) play an important role as sentinels of the immune system in initiating and controlling the quality of adaptive immune responses. Located at entry points of the host they can sense and alert the body from dangers such as infection by pathogenic bacteria. Considering their strategic localization it is not surprising that DCs have evolved in a series of DC subtypes, which are well adapted to their microenvironment. Nowadays, the advent of the identification of specific DC subtypes has opened the way for the study of pathogen-DCs interactions and the involved mechanisms of these interactions. Due to key aspect of DCs, several bacterial pathogens have taken advantage of these cells and developed mechanisms to subvert DC function and thereby evade the immune system. This review brings recent insights into DC-pathogenic bacteria cross-talk using the mouse model of infection with an emphasis on DC subtypes.
Collapse
Affiliation(s)
- Alexia Papadopoulos
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France
| | - Jean-Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France.
| |
Collapse
|
10
|
Klančnik A, Pogačar MŠ, Raspor P, Abram M, Možina SS, Vučković D. Virulence genes and cytokine profile in systemic murine Campylobacter coli infection. Virulence 2015; 6:581-90. [PMID: 26039573 DOI: 10.1080/21505594.2015.1042642] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Campylobacter coli are one of the most common bacteria in bacterial gastroenteritis and acute enterocolitis in humans. However, relatively little is known regarding the mechanisms of pathogenesis and host response to C. coli infections. To investigate the influence of genetic changes, we first used PCR to demonstrate the presence of the known virulence genes cadF, virB11, cdtB, cdtC and ceuE in the clinical isolate C. coli 26536, which was isolated from the liver of infected BALB/c mice. Sequence analyses of the cadF, virB11, cdtB and ceuE genes in C. coli 26536 confirmed the stability in these virulence genes during their transmission through the host. We further investigated C. coli infection for the bacterial clearance from the liver and spleen of infected mice, and for their immune response. C. coli persisted well in both organs, with better survival in the liver. We also determined the levels of several pro-inflammatory cytokines (i.e., interleukin [IL]-6, IL-12, interferon-γ, tumor necrosis factor-α) and the anti-inflammatory cytokine IL-10 in plasma and in liver homogenates from the infected mice, using enzyme-linked immunosorbent assays. The lowest levels among these cytokines were for tumor necrosis factor-α in the plasma and IL-6 in the liver on days 1, 3 and 8 post-infection. The most pronounced production was for IL-10, in both plasma (days 1 and 8 post-infection) and liver (day 8 post-infection), which suggests that it has a role in healing of the organ inflammation. Our findings showed dynamic relationships between pro- and anti-inflammatory cytokines and thus contribute toward clarification of the healing processes involved in the resolution of C. coli infections.
Collapse
Affiliation(s)
- Anja Klančnik
- a Department of Food Science and Technology ; Biotechnical Faculty; University of Ljubljana ; Ljubljana , Slovenia
| | | | - Peter Raspor
- d Institute of Food, Nutrition and Health; Faculty of Health Sciences; University of Primorska ; Izola , Slovenia
| | - Maja Abram
- c Department of Microbiology ; Medical Faculty; University of Rijeka ; Rijeka , Croatia
| | - Sonja Smole Možina
- a Department of Food Science and Technology ; Biotechnical Faculty; University of Ljubljana ; Ljubljana , Slovenia
| | - Darinka Vučković
- c Department of Microbiology ; Medical Faculty; University of Rijeka ; Rijeka , Croatia
| |
Collapse
|
11
|
Fimlaid KA, Lindow JC, Tribble DR, Bunn JY, Maue AC, Kirkpatrick BD. Peripheral CD4+ T cell cytokine responses following human challenge and re-challenge with Campylobacter jejuni. PLoS One 2014; 9:e112513. [PMID: 25397604 PMCID: PMC4232357 DOI: 10.1371/journal.pone.0112513] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/06/2014] [Indexed: 01/22/2023] Open
Abstract
Campylobacter jejuni is a leading cause of human gastroenteritis worldwide; however, our understanding of the human immune response to C. jejuni infection is limited. A previous human challenge model has shown that C. jejuni elicits IFNγ production by peripheral blood mononuclear cells, a response associated with protection from clinical disease following re-infection. In this study, we investigate T lymphocyte profiles associated with campylobacteriosis using specimens from a new human challenge model in which C. jejuni-naïve subjects were challenged and re-challenged with C. jejuni CG8421. Multiparameter flow cytometry was used to investigate T lymphocytes as a source of cytokines, including IFNγ, and to identify cytokine patterns associated with either campylobacteriosis or protection from disease. Unexpectedly, all but one subject evaluated re-experienced campylobacteriosis after re-challenge. We show that CD4+ T cells make IFNγ and other pro-inflammatory cytokines in response to infection; however, multifunctional cytokine response patterns were not found. Cytokine production from peripheral CD4+ T cells was not enhanced following re-challenge, which may suggest deletion or tolerance. Evaluation of alternative paradigms or models is needed to better understand the immune components of protection from campylobacteriosis.
Collapse
Affiliation(s)
- Kelly A. Fimlaid
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, Vermont, 05405, United States of America
- University of Vermont College of Medicine, Vaccine Testing Center and Unit of Infectious Diseases, Burlington, Vermont, United States of America
- * E-mail:
| | - Janet C. Lindow
- University of Vermont College of Medicine, Vaccine Testing Center and Unit of Infectious Diseases, Burlington, Vermont, United States of America
| | - David R. Tribble
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Janice Y. Bunn
- University of Vermont College of Mathematics, Burlington, Vermont, United States of America
| | - Alexander C. Maue
- Naval Medical Research Center, Enteric Diseases Department, Silver Spring, Maryland, United States of America
| | - Beth D. Kirkpatrick
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, Vermont, 05405, United States of America
- University of Vermont College of Medicine, Vaccine Testing Center and Unit of Infectious Diseases, Burlington, Vermont, United States of America
| |
Collapse
|
12
|
Bouwman LI, de Zoete MR, Bleumink-Pluym NMC, Flavell RA, van Putten JPM. Inflammasome activation by Campylobacter jejuni. THE JOURNAL OF IMMUNOLOGY 2014; 193:4548-57. [PMID: 25267974 PMCID: PMC4201959 DOI: 10.4049/jimmunol.1400648] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The Gram-negative pathogen Campylobacter jejuni is the most common cause of bacterial foodborne disease worldwide. The mechanisms that lead to bacterial invasion of eukaryotic cells and massive intestinal inflammation are still unknown. In this study, we report that C. jejuni infection of mouse macrophages induces upregulation of pro-IL-1β transcript and secretion of IL-1β without eliciting cell death. Immunoblotting indicated cleavage of caspase-1 and IL-1β in infected cells. In bone marrow-derived macrophages from different knockout mice, IL-1β secretion was found to require NLRP3, ASC, and caspase-1/11 but not NLRC4. In contrast to NLRP3 activation by ATP, C. jejuni activation did not require priming of these macrophages. C. jejuni also activated the NLRP3 inflammasome in human macrophages as indicated by the presence of ASC foci and caspase-1-positive cells. Analysis of a vast array of C. jejuni mutants with defects in capsule formation, LPS biosynthesis, chemotaxis, flagella synthesis and flagellin (-like) secretion, type 6 secretion system needle protein, or cytolethal distending toxin revealed a direct correlation between the number of intracellular bacteria and NLRP3 inflammasome activation. The C. jejuni invasion-related activation of the NLRP3 inflammasome without cytotoxicity and even in nonprimed cells extends the known repertoire of bacterial inflammasome activation and likely contributes to C. jejuni-induced intestinal inflammation.
Collapse
Affiliation(s)
- Lieneke I Bouwman
- Department of Infectious Diseases and Immunology, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Marcel R de Zoete
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520; and Howard Hughes Medical Institute, Yale University, New Haven, CT 06520
| | - Nancy M C Bleumink-Pluym
- Department of Infectious Diseases and Immunology, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520; and Howard Hughes Medical Institute, Yale University, New Haven, CT 06520
| | - Jos P M van Putten
- Department of Infectious Diseases and Immunology, Utrecht University, 3584 CL Utrecht, the Netherlands;
| |
Collapse
|
13
|
Ganesan S, Rathinam VAK, Bossaller L, Army K, Kaiser WJ, Mocarski ES, Dillon CP, Green DR, Mayadas TN, Levitz SM, Hise AG, Silverman N, Fitzgerald KA. Caspase-8 modulates dectin-1 and complement receptor 3-driven IL-1β production in response to β-glucans and the fungal pathogen, Candida albicans. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:2519-2530. [PMID: 25063877 PMCID: PMC4134963 DOI: 10.4049/jimmunol.1400276] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Inflammasomes are central mediators of host defense to a wide range of microbial pathogens. The nucleotide-binding domain and leucine-rich repeat containing family (NLR), pyrin domain-containing 3 (NLRP3) inflammasome plays a key role in triggering caspase-1-dependent IL-1β maturation and resistance to fungal dissemination in Candida albicans infection. β-Glucans are major components of fungal cell walls that trigger IL-1β secretion in both murine and human immune cells. In this study, we sought to determine the contribution of β-glucans to C. albicans-induced inflammasome responses in mouse dendritic cells. We show that the NLRP3-apoptosis-associated speck-like protein containing caspase recruitment domain protein-caspase-1 inflammasome is absolutely critical for IL-1β production in response to β-glucans. Interestingly, we also found that both complement receptor 3 (CR3) and dectin-1 play a crucial role in coordinating β-glucan-induced IL-1β processing as well as a cell death response. In addition to the essential role of caspase-1, we identify an important role for the proapoptotic protease caspase-8 in promoting β-glucan-induced cell death and NLRP3 inflammasome-dependent IL-1β maturation. A strong requirement for CR3 and caspase-8 also was found for NLRP3-dependent IL-1β production in response to heat-killed C. albicans. Taken together, these results define the importance of dectin-1, CR3, and caspase-8, in addition to the canonical NLRP3 inflammasome, in mediating β-glucan- and C. albicans-induced innate responses in dendritic cells. Collectively, these findings establish a novel link between β-glucan recognition receptors and the inflammatory proteases caspase-8 and caspase-1 in coordinating cytokine secretion and cell death in response to immunostimulatory fungal components.
Collapse
Affiliation(s)
- Sandhya Ganesan
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Vijay A K Rathinam
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lukas Bossaller
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Kelly Army
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - William J Kaiser
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Edward S Mocarski
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher P Dillon
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Tanya N Mayadas
- Center for excellence in Vascular biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Stuart M Levitz
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Amy G Hise
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
- Department of Medicine, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Neal Silverman
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Katherine A Fitzgerald
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
14
|
Kim JJ, Hwang YH, Kang KY, Kim I, Kim JB, Park JH, Yoo YC, Yee ST. Enhanced dendritic cell maturation by the B-chain of Korean mistletoe lectin (KML-B), a novel TLR4 agonist. Int Immunopharmacol 2014; 21:309-19. [PMID: 24859056 DOI: 10.1016/j.intimp.2014.05.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/24/2014] [Accepted: 05/06/2014] [Indexed: 01/04/2023]
Abstract
Korean mistletoe lectin (KML) is composed of A and B sub-chains. The B-chain binds to cell surfaces, whereas the A-chain hinders translation because it is a RIP (ribosome inactivating protein) inducing apoptosis. Although KML has various biological and immunological activities, its potential use in cancer therapy or as an adjuvant therapy is limited by its toxicity to normal cells. This study was conducted to determine whether the B-chain of KML (KML-B) has immunoadjuvant activity and cytotoxicity activity. To evaluate the immunomodulatory activities of B chain KML, in vitro experiments employing bone marrow-derived dendritic cells (BMDCs) were performed. Dendritic cells (DCs) are a unique group of white blood cells that are able to capture and process antigens for presentation to T cells, which constitute primary immune response. In the present study, KML-B was found to be non-cytotoxic to BMDCs. Furthermore, the expressions of co-stimulatory molecules (CD40, CD80, CD86, and MHC II) and the secretions of cytokines (IL-1β, IL-6, IL-12p70, and TNF-α) were increased in BMDCs by KML-B. In addition, other indicators (antigen-uptake and CCR7 expression) of BMDC maturation were changed by KML-B, and the ability of KML-B to enhance various functions by BMDCs was found to be dependent on TLR4 expression. Moreover, BMDCs matured by KML-B induced naïve CD4(+) T cell differentiation toward Th1 cells directly and indirectly. These experiments confirm that KML-B exhibits potent immunomodulatory properties and suggest that KML-B be considered a potential dendritic cell-based cancer therapy and immunoadjuvant.
Collapse
Affiliation(s)
- Jong-Jin Kim
- Department of Biology, Sunchon National University, Suncheon, Republic of Korea
| | - Yun-Ho Hwang
- Department of Pharmacy, Sunchon National University, Suncheon, Republic of Korea
| | - Kyung-Yun Kang
- Department of Pharmacy, Sunchon National University, Suncheon, Republic of Korea
| | - Inbo Kim
- School of Life Science, Handong Global University, Pohang, Republic of Korea
| | - Jong-Bae Kim
- School of Life Science, Handong Global University, Pohang, Republic of Korea
| | - Jong-Hwan Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Yung-Choon Yoo
- Department of Microbiology, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Sung-Tae Yee
- Department of Biology, Sunchon National University, Suncheon, Republic of Korea; Department of Pharmacy, Sunchon National University, Suncheon, Republic of Korea.
| |
Collapse
|
15
|
Samuelson DR, Eucker TP, Bell JA, Dybas L, Mansfield LS, Konkel ME. The Campylobacter jejuni CiaD effector protein activates MAP kinase signaling pathways and is required for the development of disease. Cell Commun Signal 2013; 11:79. [PMID: 24144181 PMCID: PMC3833307 DOI: 10.1186/1478-811x-11-79] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/07/2013] [Indexed: 12/22/2022] Open
Abstract
Background Enteric pathogens utilize a distinct set of proteins to modulate host cell signaling events that promote host cell invasion, induction of the inflammatory response, and intracellular survival. Human infection with Campylobacter jejuni, the causative agent of campylobacteriosis, is characterized by diarrhea containing blood and leukocytes. The clinical presentation of acute disease, which is consistent with cellular invasion, requires the delivery of the Campylobacter invasion antigens (Cia) to the cytosol of host cells via a flagellar Type III Secretion System (T3SS). We identified a novel T3SS effector protein, which we termed CiaD that is exported from the C. jejuni flagellum and delivered to the cytosol of host cells. Results We show that the host cell kinases p38 and Erk 1/2 are activated by CiaD, resulting in the secretion of interleukin-8 (IL-8) from host cells. Additional experiments revealed that CiaD-mediated activation of p38 and Erk 1/2 are required for maximal invasion of host cells by C. jejuni. CiaD contributes to disease, as evidenced by infection of IL-10 knockout mice. Noteworthy is that CiaD contains a Mitogen-activated protein (MAP) kinase-docking site that is found within effector proteins produced by other enteric pathogens. These findings indicate that C. jejuni activates the MAP kinase signaling pathways Erk 1/2 and p38 to promote cellular invasion and the release of the IL-8 pro-inflammatory chemokine. Conclusions The identification of a novel T3SS effector protein from C. jejuni significantly expands the knowledge of virulence proteins associated with C. jejuni pathogenesis and provides greater insight into the mechanism utilized by C. jejuni to invade host cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael E Konkel
- School of Molecular Biosciences, Washington State University, College of Veterinary Medicine, Life Sciences Bldg, Room 302c, Pullman, WA 99164-7520, USA.
| |
Collapse
|
16
|
Campylobacter jejuni outer membrane vesicles play an important role in bacterial interactions with human intestinal epithelial cells. Infect Immun 2012; 80:4089-98. [PMID: 22966047 DOI: 10.1128/iai.00161-12] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Campylobacter jejuni is the most prevalent cause of food-borne gastroenteritis in the developed world; however, the molecular basis of pathogenesis is unclear. Secretion of virulence factors is a key mechanism by which enteric bacterial pathogens interact with host cells to enhance survival and/or damage the host. However, C. jejuni lacks the virulence-associated secretion systems possessed by other enteric pathogens. Many bacterial pathogens utilize outer membrane vesicles (OMVs) for delivery of virulence factors into host cells. In the absence of prototypical virulence-associated secretion systems, OMVs could be an important alternative for the coordinated delivery of C. jejuni proteins into host cells. Proteomic analysis of C. jejuni 11168H OMVs identified 151 proteins, including periplasmic and outer membrane-associated proteins, but also many determinants known to be important in survival and pathogenesis, including the cytolethal distending toxin (CDT). C. jejuni OMVs contained 16 N-linked glycoproteins, indicating a delivery mechanism by which these periplasm-located yet immunogenic glycoproteins can interact with host cells. C. jejuni OMVs possess cytotoxic activity and induce a host immune response from T84 intestinal epithelial cells (IECs), which was not reduced by OMV pretreatment with proteinase K or polymyxin B prior to coincubation with IECs. Pretreatment of IECs with methyl-beta-cyclodextrin partially blocks OMV-induced host immune responses, indicating a role for lipid rafts in host cell plasma membranes during interactions with C. jejuni OMVs. OMVs isolated from a C. jejuni 11168H cdtA mutant induced interleukin-8 (IL-8) to the same extent as did wild-type OMVs, suggesting OMV induction of IL-8 is independent of CDT.
Collapse
|
17
|
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
|
18
|
Rathinam VAK, Vanaja SK, Waggoner L, Sokolovska A, Becker C, Stuart LM, Leong JM, Fitzgerald KA. TRIF licenses caspase-11-dependent NLRP3 inflammasome activation by gram-negative bacteria. Cell 2012; 150:606-19. [PMID: 22819539 DOI: 10.1016/j.cell.2012.07.007] [Citation(s) in RCA: 578] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 06/04/2012] [Accepted: 07/03/2012] [Indexed: 12/14/2022]
Abstract
Systemic infections with Gram-negative bacteria are characterized by high mortality rates due to the "sepsis syndrome," a widespread and uncontrolled inflammatory response. Though it is well recognized that the immune response during Gram-negative bacterial infection is initiated after the recognition of endotoxin by Toll-like receptor 4, the molecular mechanisms underlying the detrimental inflammatory response during Gram-negative bacteremia remain poorly defined. Here, we identify a TRIF pathway that licenses NLRP3 inflammasome activation by all Gram-negative bacteria. By engaging TRIF, Gram-negative bacteria activate caspase-11. TRIF activates caspase-11 via type I IFN signaling, an event that is both necessary and sufficient for caspase-11 induction and autoactivation. Caspase-11 subsequently synergizes with the assembled NLRP3 inflammasome to regulate caspase-1 activation and leads to caspase-1-independent cell death. These events occur specifically during infection with Gram-negative, but not Gram-positive, bacteria. The identification of TRIF as a regulator of caspase-11 underscores the importance of TLRs as master regulators of inflammasomes during Gram-negative bacterial infection.
Collapse
Affiliation(s)
- Vijay A K Rathinam
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
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
|
20
|
Szymanski CM, Gaynor E. How a sugary bug gets through the day: recent developments in understanding fundamental processes impacting Campylobacter jejuni pathogenesis. Gut Microbes 2012; 3:135-44. [PMID: 22555465 PMCID: PMC3370946 DOI: 10.4161/gmic.19488] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Campylobacter jejuni is a highly prevalent yet fastidious bacterial pathogen that poses a significant health burden worldwide. Lacking many hallmark virulence factors, it is becoming increasingly clear that C. jejuni pathogenesis involves different strategies compared with other well-characterized enteric organisms. This includes the involvement of basic biological processes and cell envelope glycans in a number of aspects related to pathogenesis. The past few years have seen significant progress in the understanding of these pathways and how they relate to C. jejuni fundamental biology, stress survival, colonization, and virulence attributes. This review focuses on recent studies in three general areas where "pathogenesis" and "basic biology" overlap: physiology, stress responses and glycobiology.
Collapse
Affiliation(s)
- Christine M. Szymanski
- Alberta Glycomics Centre and Department of Biological Sciences; University of Alberta; Edmonton, Canada,Correspondence to: Christine M. Szymanski, or Erin Gaynor,
| | - Erin Gaynor
- Department of Microbiology and Immunology; University of British Columbia; Vancouver, Canada,Correspondence to: Christine M. Szymanski, or Erin Gaynor,
| |
Collapse
|
21
|
Activation of innate immune responses in a pathogen-mimicking manner by amphiphilic polyanhydride nanoparticle adjuvants. Biomaterials 2011; 32:6815-22. [DOI: 10.1016/j.biomaterials.2011.05.063] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 05/20/2011] [Indexed: 11/22/2022]
|
22
|
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
|
23
|
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
|
24
|
Panchanathan R, Duan X, Shen H, Rathinam VAK, Erickson LD, Fitzgerald KA, Choubey D. Aim2 deficiency stimulates the expression of IFN-inducible Ifi202, a lupus susceptibility murine gene within the Nba2 autoimmune susceptibility locus. THE JOURNAL OF IMMUNOLOGY 2010; 185:7385-93. [PMID: 21057088 DOI: 10.4049/jimmunol.1002468] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Murine Aim2 and p202 proteins (encoded by the Aim2 and Ifi202 genes) are members of the IFN-inducible p200 protein family. Both proteins can sense dsDNA in the cytoplasm. However, upon sensing dsDNA, only the Aim2 protein through its pyrin domain can form an inflammasome to activate caspase-1 and induce cell death. Given that the p202 protein has been predicted to inhibit the activation of caspase-1 by the Aim2 protein and that increased levels of the p202 protein in female mice of certain strains are associated with lupus susceptibility, we compared the expression of Aim2 and Ifi202 genes between Aim2-deficient and age-matched wild-type mice. We found that the Aim2 deficiency in immune cells stimulated the expression of Ifi202 gene. The increased levels of the p202 protein in cells were associated with increases in the expression of IFN-β, STAT1, and IFN-inducible genes. Moreover, after knockdown of Aim2 expression in the murine macrophage cell line J774.A1, IFN-β treatment of cells robustly increased STAT1 protein levels (compared with those of control cells), increased the activating phosphorylation of STAT1 on Tyr-701, and stimulated the activity of an IFN-responsive reporter. Notably, the expression of Aim2 in non-lupus-prone (C57BL/6 and B6.Nba2-C) and lupus-prone (B6.Nba2-ABC) splenic cells and in a murine macrophage cell line that overexpressed p202 protein was found to be inversely correlated with Ifi202. Collectively, our observations demonstrate an inverse correlation between Aim2 and p202 expressions. We predict that defects in Aim2 expression within immune cells contribute to increased susceptibility to lupus.
Collapse
|
25
|
The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nat Immunol 2010; 11:395-402. [PMID: 20351692 PMCID: PMC2887480 DOI: 10.1038/ni.1864] [Citation(s) in RCA: 1055] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 03/08/2010] [Indexed: 12/02/2022]
Abstract
Inflammasomes regulate the activity of capase-1 and maturation of interleukin-1β and interleukin-18. Recently, AIM2 was shown to bind DNA and engage ASC to form a caspase-1 activating inflammasome. Using Aim2-deficient mice, we reveal a central role for AIM2 in regulating caspase-1-dependent maturation of IL-1β and IL-18, as well as pyroptosis in response to synthetic dsDNA. AIM2 is essential for inflammasome activation in response to Fransicella tularensis, vaccinia virus, mouse cytomegalovirus and plays a partial role in sensing Listeria monocytogenes. Moreover, production of IL-18 and NK cell-dependent IFN-γ production, events critical in early control of virus replication were dependent on AIM2 during mCMV infection in vivo. Collectively, these observations reveal the importance of AIM2 in sensing both bacterial and viral pathogens and triggering innate immunity.
Collapse
|
26
|
Lee CH, Nawar HF, Mandell L, Liang S, Hajishengallis G, Connell TD. Enhanced antigen uptake by dendritic cells induced by the B pentamer of the type II heat-labile enterotoxin LT-IIa requires engagement of TLR2. Vaccine 2010; 28:3696-705. [PMID: 20332049 DOI: 10.1016/j.vaccine.2010.03.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 03/01/2010] [Accepted: 03/09/2010] [Indexed: 11/19/2022]
Abstract
The potent mucosal adjuvant properties of the type II heat-labile enterotoxin LT-IIa of Escherichia coli are dependent upon binding of the B pentamer of the enterotoxin (LT-IIa-B(5)) to ganglioside receptors on immunocompetent cells. To evaluate the immunomodulatory activities of LT-IIa-B(5), in vitro experiments employing bone marrow-derived dendritic cells (BMDC) were performed. Uptake of OVA-FITC, a model antigen (Ag), was enhanced by treatment of BMDC with LT-IIa-B5, but not by treatment of cells with the B pentamer of cholera toxin (CTB). Expression of co-stimulatory molecules (CD40, CD80, CD86, and MHC-II) and cytokines (IL-12p40, TNF-alpha, and IFN-gamma) was increased in BMDC treated with LT-IIa-B(5). The capacity of LT-IIa-B(5) to enhance Ag uptake and to induce expression of co-stimulatory receptors and cytokines by BMDC was dependent upon expression of TLR2 by the cell. Increased Ag uptake induced by LT-IIa-B(5) was correlated with increased Ag-specific proliferation of CD4(+) T cells in an in vitro syngeneic DO11.10 CD4(+) T cell proliferation assay. These experiments confirm that LT-IIa-B(5) exhibits potent immunomodulatory properties which may be exploitable as a non-toxic mucosal adjuvant.
Collapse
Affiliation(s)
- Chang Hoon Lee
- The Department of Microbiology and Immunology, The University at Buffalo, NY 14214, USA
| | | | | | | | | | | |
Collapse
|
27
|
van Putten JPM, van Alphen LB, Wösten MMSM, de Zoete MR. Molecular mechanisms of campylobacter infection. Curr Top Microbiol Immunol 2010; 337:197-229. [PMID: 19812984 DOI: 10.1007/978-3-642-01846-6_7] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Campylobacter jejuni is the principal bacterial foodborne pathogen. A major challenge still is to identify the virulence strategies exploited by C. jejuni. Recent genomics, proteomics, and metabolomics approaches indicate that C. jejuni displays extensive inter- and intrastrain variation. The diverse behavior enables bacterial adaptation to different environmental conditions and directs interactions with the gut mucosa. Here, we report recent progress in understanding the molecular mechanisms and functional consequences of the phenotype diversity. The results suggest that C. jejuni actively penetrates the intestinal mucus layer, secretes proteins mainly via its flagellar apparatus, is engulfed by intestinal cells, and can disrupt the integrity of the epithelial lining. C. jejuni stimulates the proinflammatory pathway and the production of a large repertoire of cytokines, chemokines, and innate effector molecules. Novel experimental infection models suggest that the activation of the innate immune response is important for the development of intestinal pathology.
Collapse
Affiliation(s)
- Jos P M van Putten
- Department of Infectious Diseases & Immunology, Utrecht University, Yalelaan 1, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
28
|
Assessment of the duration of protection in Campylobacter jejuni experimental infection in humans. Infect Immun 2010; 78:1750-9. [PMID: 20086085 DOI: 10.1128/iai.01021-09] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A human Campylobacter jejuni infection model provided controlled exposure to assess vaccine efficacy and investigate protective immunity for this important diarrheal pathogen. A well-characterized outbreak strain, C. jejuni 81-176, was investigated using a volunteer experimental infection model to evaluate the dose range and duration of protection. Healthy Campylobacter-seronegative adults received C. jejuni strain 81-176 via oral inoculation of 10(5), 10(7), or 10(9) CFU (5 adults/dose), which was followed by clinical and immunological monitoring. Based on dose range clinical outcomes, the 10(9)-CFU dose (n = 31) was used to assess homologous protection at 28 to 49 days (short-term veterans [STV]; n = 8) or 1 year (long-term veterans [LTV]; n = 7) after primary infection. An illness dose effect was observed for naïve subjects (with lower doses, 40 to 60% of the subjects were ill; with the 10(9)-CFU dose, 92% of the subjects were ill) along with complete protection for the STV group and attenuated illness for the LTV group (57%). Partial resistance to colonization was seen in STV (25% of the subjects were not infected; 3-log-lower maximum excretion level). Systemic and mucosal immune responses were robust in naïve subjects irrespective of the dose or the severity of illness. In contrast, in STV there was a lack of circulating antibody-secreting cells (ASC), reflecting the local mucosal effector responses. LTV exhibited comparable ASC responses to primary infection, and anamnestic fecal IgA responses likely contributed to self-resolving illness prior to antibiotic treatment. Campylobacter antigen-dependent production of gamma interferon by peripheral blood mononuclear cells was strongly associated with protection from illness, supporting the hypothesis that TH1 polarization has a primary role in acquired immunity to C. jejuni. This study revealed a C. jejuni dose-related increase in campylobacteriosis rates, evidence of complete short-term protection that waned with time, and immune response patterns associated with protection.
Collapse
|
29
|
Campylobacter jejuni-induced activation of dendritic cells involves cooperative signaling through Toll-like receptor 4 (TLR4)-MyD88 and TLR4-TRIF axes. Infect Immun 2009; 77:2499-507. [PMID: 19332531 DOI: 10.1128/iai.01562-08] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Campylobacter jejuni is an important cause of human enteritis and has been linked to the development of autoimmune diseases. Recently we showed that infection of murine dendritic cells (DCs) with C. jejuni resulted in DC activation and induction of Campylobacter-specific Th1-effector responses. Toll-like receptor (TLR) signaling through myeloid differentiation factor 88 (MyD88) and/or Toll-interleukin 1 (IL-1) receptor domain-containing adaptor-inducing beta interferon (IFN-beta) (TRIF) is critical in inducing immunity against pathogens. In this study, we investigated the role of TLR2, TLR4, MyD88, and TRIF signaling in C. jejuni-induced inflammatory activation of DCs. DC upregulation of major histocompatibility complex class II and costimulatory molecules after C. jejuni challenge was profoundly impaired by TLR2, TLR4, MyD88, and TRIF deficiencies. Similarly, C. jejuni-induced secretion of IL-12, IL-6, and tumor necrosis factor alpha was significantly inhibited in TLR2(-/-), TLR4(-/-), MyD88(-/-), and TRIF(-/-) DCs compared to that in wild-type DCs; however, the magnitude of inhibition was greater in MyD88(-/-), TRIF(-/-), and TLR4(-/-) DCs than in TLR2(-/-) DCs. Furthermore, C. jejuni induced interferon regulatory factor 3 phosphorylation and IFN-beta secretion by DCs in a TLR4-TRIF-dependent fashion, further demonstrating activation of this pathway by C. jejuni. Importantly, TLR2, TLR4, MyD88, and TRIF deficiencies all markedly impaired the Th1-priming ability of C. jejuni-infected DCs. Thus, our results show that cooperative signaling through the TLR4-MyD88 and TLR4-TRIF axes represents a novel mechanism mediating C. jejuni-induced inflammatory responses of DCs. To our knowledge, such a mechanism has not been demonstrated previously for an intact bacterium.
Collapse
|