‘ thaw 1. .331... a . $\ t: , a .x.. , 743311. 21.. iv...» . .. afih n‘e x «1.043% "L11. 2. 12;". . . . ‘. \ (a 1. 1:53. is s ‘01 ‘33:»... .l’ 5:1 ‘ . 0|. . 315.....1‘2AVYI; 1333.93.75 V It! 9:. . i. 3315'}! v... (a. ivi, . z... i .. 9. 3 3.5: . .l. 1 1 :3... . .:.\ .1. awsqéfi 215.3%.» . . . x37. him” . ‘ am. . .1. . ‘. 3...... . .r (>0? LIBRARY Mich:- ‘ State Ui'iivmalt'y' This is to certify that the dissertation entitled INTERACTION BETWEEN DENDRITIC CELLS AND CAMPYLOBACTER JEJUNI: ROLE OF TOLL-LIKE RECEPTOR SIGNALING presented by VIJAY ANAND KARUPPANNAN RATHINAM has been accepted towards fulfillment of the requirements for the PhD. degree in COMPARATIVE MEDICINE AND INTEGRATIVE BIOLOGY 05/29/09 Date MSU is an Affirmative Action/Equal Opportunity Employer PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 K /PrqlAcc&Pres/C IRC/DaIeDue indd INTERACTION BETWEEN DENDRITIC CELLS AND CAMPYLOBACTER JEJUNI: ROLE OF TOLL-LIKE RECEPTOR SIGNALING By Vijay Anand Karuppannan Rathinam A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Comparative Medicine and Integrative Biology 2009 ABSTRACT INTERACTION BETWEEN DENDRITIC CELLS AND CAMPYLOBACTER JEJUNI: ROLE OF TOLL-LIKE RECEPTOR SIGNALING By Vijay Anand Karuppannan Rathinam Campy/abacterjejuni is a clinically significant food-bome pathogen that causes enteritis. Dendritic cells (00s) are central to initiating immune responses to pathogens. One objective of this study was to understand the interaction of murine DCs with C. jejuni and its impact on induction of T cell responses mediating resistance. Following infection with C. jejuni, DCs were found to efficiently kill C. jejuni and undergo activation by up regulating the surface expression of maturation markers and by secreting IL-12, lL-6 and TNF-a. Notably, C. jejuni-infected DCs induced Th1-differentiation of naive CD4+ T cells. Next, we investigated the role of toll-like receptor (TLR) signaling in mediating these responses. Upregulation of maturation markers was significantly impaired in both TLR2-I- and TLR4-/- 005 relative to wild type (WT) DCs after C. jejuni challenge. In contrast, TLR4 deficiency, but not TLR2 deficiency, profoundly impaired the cytokine responses following C. jejuni infection. Because TLR4 utilizes both My088 and TRIF adapters for signal transduction, we investigated the role of MyD88 and TRIF in these responses. The expression of maturation markers and cytokines in response to C. jejuni was greatly reduced in the absence of either MyD88 or TRIF. Furthermore, C. jejuni infection induced IRF-3 phosphorylation and IFN-B secretion by 005 in a TLR4-TRIF dependent fashion, further demonstrating activation of this pathway by C. jejuni. lmportantly, TLR2, TLR4, My088, and TRIF deficiencies all markedly impaired Th1-priming ability of C. jejuni-infected DCs. Thus, our results show for the first time that cooperative signaling through MyDB8-dependent and -independent (TRIF) arms of the TLR4 signaling represents a novel mechanism mediating C. jejuni-induced inflammatory activation of DCs. Copyright by Vijay Anand Karuppannan Rathinam 2009 ACKNOWLEDGEMENTS I would like to acknowledge first and foremost Dr. Linda S. Mansfield for being a great advisor and allowing me to pursue my research interests in immunology as well as for providing all the guidance and resources I needed. I am also thankful to her for the freedom I enjoyed in my research. I thank my committee member Dr. Kathleen Hoag for her critical inputs in my research and for helping me in establishing the dendritic cell culture method and other assays I have used. I would also like to sincerely thank my committee members Dr. John Linz, Dr. Katheryn Meek, and Dr. Jon Patterson for their guidance and support. Members of the Mansfield lab made the working environment very friendly and fun-filled. Julia Bell was of great help with statistical consultations and critically reviewing my manuscripts. It has been a pleasure working with Jamie Kopper, Jessica St Charles, JP Jerome, and Andy Flies and I would miss them and all the fun moments we had in the last few years. I also thank other members of the Mansfield lab such as Alice Murphy, Jenna Gettings, Jennifer Olmstead, Amanda Stanton, Eric Smith, Alex Adrian, Bianca Buffa, and Anne Plovanich- Jones for all their help. I thank Vilma Yuzbasiyan-Gurkan for her support and for providing me with the travel funds to attend various conferences. Victoria Hoelzer-Maddox and Margaret Nicholas were of great administrative help. Louis King helped me with the flow cytometry. I thank Andrea Amalfitano and Daniel Appledom for breeding and providing the TLR2, MyD88 and TRIF KO mice I used and for all the exciting immunology discussions. I also thank Dr. Puliyur MohanKumar, Dr. Sheeba MohanKumar, and my friends Ravi, Senthil, Madhu, Sugaleshini, Sarguru, and Madhan for all their help and support. I am grateful to my family for their love and support. My wife Sivapriya Kailasan Vanaja has been a tremendous support and without her encouragement, cooperation and help, I could not have completed this study. vi PUBLICATIONS Chapters 2 and 3 of this dissertation have been published in Microbes and Infection and Infection and Immunity, respectively, as follows: Rathinam, V. A., K. A. Hoag, and L. S. Mansfield. 2008. Dendritic cells from C57BU6 mice undergo activation and induce Th1-effector cell responses against Campylobacterjejuni. Microbes Infect 10:1316-1 324. Rathinam, V. A., D. M. Appledorn, K. A. Hoag, A. Amalfitano, and L. 8. Mansfield. 2009. Campy/abacterjejuni-induced activation of dendritic cells involves cooperative signaling through TLR4-My088 and TLR4—TRIF axes. Infect Immun 77(6): 2499-2507. vii TABLE OF CONTENTS LIST OF TABLES .................................................................................................. ix LIST OF FIGURES .............................................................................................. .x CHAPTER 1. Introduction ........................................................................ 1 Camp ylobacter jejuni ........................................................................ 2 Host defense responses to C. jejuni ................................................... 15 Dendritic ells ................................................................................. 21 Toll-like receptors: Pathogen sensing and initiation of immune responses ..................................................................................... 26 Rationale for this study .................................................................... 31 CHAPTER 2. Dendritic cells from C57BU6 mice undergo activation and induce Th1-effector cell responses against Campy/abacterjejuni .................... 35 Abstract ........................................................................................ 36 Introduction ................................................................................... 37 Materials and methods .................................................................... 39 Results ........................................................................................ 44 Discussion .................................................................................... 49 Figures ......................................................................................... 55 CHAPTER 3. Campy/obacterjejunI-induced activation of dendritic cells involves cooperative signaling through TLR4-MyD88 and TLR4-TRIF axes ..... 68 Abstract ....................................................................................... 69 Introduction .................................................................................. 70 Materials and methods .................................................................... 73 Results ........................................................................................ 79 Discussion .................................................................................... 85 Figures ........................................................................................ 91 CHAPTER 4. Summary and future directions ............................................. 100 Summary ................................................................................... 101 Future directions .......................................................................... 1 13 APPENDIX ....................................................................................... 1 19 BIBLIOGRAPHY ................................................................................. 120 viii LIST OF TABLES Chapter 1 Table 1. TLR ligands ......................................................................... 33 LIST OF FIGURES Chapter 1 Figure 1. TLR2 and TLR4 signaling ..................................................... 34 Chapter 2 Figure 1. Internalization and killing of C. jejuni by BM-DCs ....................... 56 Figure 2. Maturation and cytokine production by BM-DCs infected with C. jejuni ....................................................................... 58 Figure 3. Effect of viability of C. jejuni on activation of DCs ...................... 60 Figure 4. Requirement for contact and internalization of C. jejuni for activation of DCs ........................................................................ 62 Figure 5. Cytokine responses of BM-DCs to various strains of C. jejuni ...... 65 Figure 6. Polarization of CD4+ T cells by C. jejuni-infected BM-DCs ............ 67 Chapter 3 Figure 1. TLR2 and TLR4 signaling are required for C. jejuni-induced maturation of BM-DCs ....................................................................... 92 Figure 2. C. jejuni-induced cytokine responses of BM-DCs is mainly dependent on TLR4 signaling ............................................................. 94 Figure 3. MyD88 and TRIF signaling dependent Upregulation of surface markers and cytokine secretion by C. jejuni-infected BM-DCs ..................... 96 Figure 4. Activation of lRF-3 phosphorylation by C. jejuni-induced TLR4wTRIF signaling ......................................................................................... 97 Figure 5. IFN-B production by C. jejuni-infected BM-DCs .......................... 98 Figure 6. TLR2, TLR4, MyD88, and TRIF signaling are necessary for maximal Th1 polarization by BM-DCs infected with C. jejuni .................................. 99 Chapter 4 Figure 1. Proposed model for the role of TLR signaling in C. jejuni-induced activation of DCs .................................................... 112 xi CHAPTER 1 Introduction CAMPYLOBA C TER JEJUNI Campy/obecterjejuni is one of the leading causes of food-borne bacterial enteritis in humans. C. jejuni is also the second leading cause of traveler’s diarrhea after enterotoxigenic Escherichia coli (146). Although Campy/obacter was described by Theodor Escherich as early as 1886 as a spiral bacterium in the colonic tissues of children who died of what he termed ‘cholera infantum’ (46), it was not recognized as a widespread etiological agent of diarrheal illness until the 1970’s (23). The crucial breakthrough for this discovery was the successful isolation of Campy/obacter from the feces and blood of a woman suffering from severe diarrhea and fever (42). Heretofore, the microaerophilic and fastidious growth requirements of this organism had made it difficult to isolate. C. jejuni belongs to the epsilon subdivision of the Proteobacten'a and is a member of the family Campy/obacteraceae (40). C. jejuni is a non-spore forming Gram-negative and microaerophilic bacterium. It is 0.2 - 0.8 um wide and 0.5 — 5 um long with a spiral, curved or rod shape and is highly motile with a characteristic cork-screw darting motility. C. jejuni has a single unsheathed polar flagellum at one or both ends (40). C. jejuni possesses a relatively small genome of size ~ 1.7 Mb (1.59 -— 1.78 Mb) with low G+C content (~ 30%). C. jejuni exists as a commensal in the gastrointestinal tract of many food animals, especially chickens, and is transmitted to humans by the consumption of contaminated water, undercooked meat, and raw milk (23, 54, 94) and by contact with pet animals and livestock. Epidemiology of C. jejuni infection C. jejuni and C. coli are responsible for about 95% of Campy/obacter infections in humans (100); however, other species of Campy/cheater such as C. fetus, C. upsaliensis, C. hyointestinalis, and C. Iari are also implicated in human intestinal and extraintestinal diseases. The significance of the non-jejuni and non- coli species of Campy/obecters as human pathogens is just beginning to be understood (100). C. jejuni is the most common food-borne enteric bacterium in many developed countries and is the second most common in the US (26). More than 2 million people are estimated to be affected by diarrheal illness due to C. jejuni infection in the US every year (1). Infections with campylobacters account for approximately 17% of hospitalization associated with food-borne pathogens (112). In addition, the economic burden due to campylobacter illnesses is high, up to $8 billion in the US annually (24). The incidence of Campy/cheater infection in immunocompromised individuals such as AIDS patients has been reported to be 40 — 100 times higher than in the immunocompetant population (152). The rate of C. jejuni infection in the US and developed countries is higher in children less than 1 year old. The incidence of C. jejuni infection has been reported to be more frequent in males than in females in all age groups except for the 20-29 year old group, where the trend is reversed (126). Moreover, seasonal variation occurs in the incidence of C. jejuni infection; C. jejuni cases appear to rise during the spring, reach peak levels in summer, and decline thereafter with the smallest number of cases reported during the winter months. The vast majority of C. jejuni infections occurs as sporadic isolated cases not as part of large outbreaks (126). In developing countries, C. jejuni infection is hyperendemic among younger children with 5 — 10 separate episodes of symptomatic infection in the first few years of life owing to poor hygiene (25, 160, 161 ). However, the rate of symptomatic infections of C. jejuni decreases as age increases, which has been shown to correlate with the appearance of C. jejuni-specific IgA antibodies in serum (19). Intestinal manifestations of C. jejuni infection Infection with C. jejuni causes a spectrum of diseases in humans (184). The clinical outcome of C. jejuni infection is likely to depend on multiple factors such as dose and genetic make up of the C. jejuni strain, immune status of the host and the individual’s intestinal microbiota (15). Self-limiting intestinal illness is the most common manifestation in the industrialized nations. As few as 800 colony forming units (cfu) of C. jejuni can cause illness in humans (17) and 100 cfu in IL-10”' mice (110). After an incubation period of 1 — 7 days (usually 24 - 48 h), patients exhibit one or more of the following clinical symptoms: headache, fever, abdominal pain, myalgia, and diarrhea. Abdominal pain and watery to bloody diarrhea are the most frequent symptoms of C. jejuni infection, but vomiting is not very common (18). Diarrhea persists for 3 - 4 days, with frank blood appearing in the feces during the later stages in approximately 15% of all patients and in 30% of hospitalized patients. Patients may continue to experience abdominal pain for several days even after the diarrhea has disappeared. Usually the duration of intestinal illness is less than a week even without antibiotic therapy (172). The relative contributions of innate and adaptive immunity in this resolution of infection are not clear. Relapses, usually characterized by mild illness, can occur in about 20% of the patients (35, 172). It has been reported that C. jejuni continues to be excreted for 16 days on average after the onset of diarrhea (172). In developing countries, infection with C. jejuni can be asymptomatic, and mild non-inflammatory diarrhea is also not uncommon (103, 160). Extraintestinal manifestations of C. jejuni infection Self-limiting intestinal illness, as described above, is the most common clinical presentation in C. jejuni infection. In contrast, patients with immunodeficiencies such as hypogammaglobulinemia and AIDS experience prolonged and recurrent intestinal disease often accompanied by bacteremia and extraintestinal diseases (18, 35, 94). Pancreatitis, cholecystitis, hepatitis, peritonitis, and meningitis have been reported to be caused by C. jejuni (35). Bacteremia during pregnancy can lead to premature labor, perinatal sepsis, neonatal meningitis, and abortion; however, such pregnancy-related complications due to C. jejuni infection are rare (18). Complications due to C. jejuni infection The common post-infectious complications of C. jejuni infection include reactive arthritis (ReA), Reiters Syndrome, and Guillain-Barré Syndrome (688) (18). ReA affecting ankles, knees, wrists and other peripheral joints has been reported to occur in about 1% — 5% of C. jejuni patients (136). ReA is characterized by stiffness, pain, swelling, warmth, and redness of the involved joints. Though the incidence of ReA after C. jejuni infection did not correlate with the human leukocyte antigen (HLA) subtype 827, the possession of this HLA- 827 tissue antigen is implicated in the severity of ReA (18). The molecular basis of the development of ReA following C. jejuni infection remains unknown. 688, an acute progressive peripheral neuropathy, is also a rare autoimmune complication associated with C. jejuni infection. C. jejuni infection is the most common antecedent infection associated with the development of GBS; about 20% of 688 patients have had a previous C. jejuni infection (159). Infections with cytomegalovirus, Epstein-Barr virus, Mycoplasma pneumoniae, and Haemophilus influenzae have also been associated with 688 cases (94, 159). Notably, asymptomatic infection with C. jejuni can also lead to the development of 63$ (98). CBS is estimated to occur in 1 in 1.000 C. jejuni patients (75, 94). The onset of neurological symptoms of GBS mostly occurs about 1 - 3 weeks after infection with C. jejuni (75). 688 is the leading cause of acute flaccid paralysis in polio-free regions of the world. 688 is characterized by paralysis, pain, muscular weakness and respiratory insufficiency in some cases, which warrants the use of ventilators (94). One possible mechanism widely believed to underlie the development of 638 following C. jejuni infection is molecular mimicry between ganglioside structures and lipooligosaccharide (LOS). The vast majority of GBS patients have been reported to have autoantibodies against gangliosides, which are sialic acid- containing glycosphingolipids that are highly concentrated in myelin sheaths of neuronal axons. Autoantibodies have been detected against more than 20 types of human cell surface gangliosides but more frequently against LM1, GM1, GM1b, GM2, GD1a, GalNAc-GD1a, GD1 b, 602, GD3, GT1a and GQ1b (75). It is hypothesized that the antibodies directed against C. jejuni LOS cross react with gangliosides of nerves, initiating a vicious cycle of inflammation that ultimately results in demyelination and nervous tissue damage (75). However, a clear understanding of pathogenic mechanism(s) that contribute to the initiation of CBS is still lacking. Pathogenesis of C. jejuni. After oral ingestion, C. jejuni reaches its predilection sites such as terminal ileum, cecum, and proximal colon, where it adheres to and colonizes the apical surface of the epithelium. In human patients and animal models, invasion of intestinal epithelial cells by C. jejuni has been suggested to play an important role in its pathogenesis (35, 54). This key process depends on several bacterial factors including motility, adhesion, and secreted proteins. C. jejuni exhibits a chemotactic response towards the mucus—particularly the glycoprotein mucin—Iining the intestinal epithelium (69, 97). Consistent with this observation, C. jejuni cells were found to be associated with mucus near goblet cells in pigs orally challenged with C. jejuni (109). Furthermore, the chemotactic property of C. jejuni has been shown to be important for its colonization of the intestinal tract in murine models (158). Previous studies showed that the motility of C. jejuni requires a functional flagellum and FlaA, one of the flagellar proteins (174). The adhesion of C. jejuni to epithelial cells has been shown to be mediated by surface structures such as PEB1 (a homolog of gram-negative ATP-binding transporters), CadF (a fibronectin binding protein), and leA (surface lipoprotein) (79, 80, 118, 131, 132). More importantly, a flagellar export system similar to the type III secretion apparatus of other gram-negative bacteria has been identified in C. jejuni. This flagellar secretion system was found to be essential for secreting Campy/abacter invasion antigens into host cells and thus facilitating invasion (96). Furthermore, a homolog of a type IV secretion system was identified in a large plasmid (pVir) present in some but not all of strains of C. jejuni. The ComB and VirB11 proteins encoded by this plasmid have been shown to contribute to invasion of INT407 intestinal epithelial cells (IEC) in vitro and to virulence in a ferret diarrheal disease model (11). C. jejuni has been shown to breach the mucosal epithelial layer and gain access to the lamina propria by two other routes besides apical invasion of epithelial cells: 1) via paracellular entry by disrupting epithelial tight junctions, and 2) via uptake by M cells (168), after which C. jejuni can enter epithelial cells basolaterally. All of these interactions with epithelial cells trigger intense inflammatory responses characterized by the infiltration of the lamina propria and submucosa with neutrophils and lymphocytes, resulting in tissue damage and pathology. However, the exact role of C. jejuni and its components in eliciting acute inflammatory responses and in inducing tissue damage remains to be elucidated. Colonization of intestinal tract by C. jejuni Colonization of the small and large intestines is a critical step in the pathogenesis of C. jejuni infection. Development of new animal models of C. jejuni in recent years allowed identification of important pathogen factors that mediate the colonization process. These factors include flagellin, a membrane protein peb1A, a member of a two-component regulatory system (RacR) (22), capsule, Png a member of a glycosylation system (83), and a multidrug efflux pump CmeABC (102); some of those factors are described in detail below. Virulence factors of C. jejuni After the genome sequencing of C. jejuni NTCC 11168, there has been a renewal of interest in studying the pathogenesis of C. jejuni infection, which has led to the delineation of some of its virulence mechanisms. The following virulence factors of C. jejuni have been shown to play a key role in pathogenesis. Flagella C. jejuni has a single unsheathed flagellum at one or both ends of the cell. Flagella aid in motility of C. jejuni allowing it to move through the GI tract lumen and into the mucus layer and have been shown to play a role in intestinal epithelial cell attachment and invasion in vitro (55). Besides contributing to motility-related processes, the flagellar apparatus serves as a type III secretion system (96). Toll-like receptor 5 (TLR5) is an innate receptor expressed by host cells that recognizes bacterial flagellins and triggers inflammatory responses (4). Owing to amino acid substitutions at the TLR5 recognition site, C. jejuni flagellins studied to date are not ligands for TLR5, thus contributing to immune evasion by this bacterium (6). However, it is not known whether other pattern recognition receptors for flagellin detection such as Nod-like receptor C4 and neuronal apoptosis inhibitory protein 5 (NAIP5) (155) are activated by flagellin of C. jejuni. Toxins Pathogenic bacteria produce a variety of cytotoxins that play multiple roles in mediating virulence during pathogenesis, including colonization, invasion, and killing of host cells (170). Various strains of C. jejuni have been shown to be cytotoxic to various cell lines; hemolytic activities have also been demonstrated. The C. jejuni genome encodes four cytotoxins including cytolethal distending toxin (CDT), a putative hemolytic cytotoxin (leA), a putative integral membrane protein with a hemolysin domain, and phospholipase A (PldA) (129). With the exception of CDT, the role(s) of these toxins in the disease process, except for CDT, have yet to be elucidated. It has been shown that pore-forming cytolysins of bacteria exhibit a diverse spectrum of pathogenic effects on host cells including hemolysis, cytokine induction, cytoskeletal dysfunction, and killing of host cells (170). There are many reports on the cytotoxic effects of C. jejuni on 10 various cell lines (3, 47, 82, 133). However, no consensus has been reached on cytotoxin production by C. jejuni since different cell lines and strains of C. jejuni were used in these studies. The genome sequence of C. jejuni contains a gene tIyA of 762 bp encoding a putative hemolysin of 253 amino acids with molecular weight of 29 kDa. The predicted amino acid sequence has a high identity to hemolysins of Helicobacter pylori (43% identity), Treponema hyodysenteriae (40% identity), and Mycobacterium tuberculosis (37% identity) (129). The C. jejuni genome also contains the pIdA gene (990 bp) encoding phospholipase A2 of 329 amino acids with a molecular weight of 38.8 kDa. The predicted amino acid sequence of PldA of C. jejuni is highly homologous to phospholipase A of Campy/obacter coli (74% identity), Yersinia enterocolitica (35% identity) and H. pylori (26% identity) ( 129). Preliminary experiments indicated that PldA of C. jejuni is essential for cecal colonization in chicken (191), but it has not been characterized further in vitro or in vivo. Cytolethal distending toxin (CDT) is a holotoxin secreted by C. jejuni, which is composed of three subunits (Cth, CdtB and CdtC). It possesses DNAse I activity and causes cell cycle arrest in the GZIM phase and cell distension and has been implicated in invasion and immunomodulation (62). CDT has been shown to induce lL-8 production by intestinal epithelial cell line INT407. It has also been documented that CDT triggers apoptosis in cultured human 28SC monocytes (61 ). Fox et al. (2004) reported that CDT is required for persistent colonization of C57BL/129 mice as a cdtB mutant of C. jejuni, unlike wild type strain, was not recovered from C57BL/129 mice at 2 and 4 months after 11 oral inoculation. They also suggested that CDT elicits proinflammatory responses in the host and contributes to immune evasion by C. jejuni (48). Capsule Using genetic and biochemical approaches, Karlyshev et al. (2000) demonstrated that C. jejuni possesses capsular polysaccharides (87), until then considered to be high molecular weight lipopolysaccharides. Significant variations in the capsular polysaccharide structure have been observed among various strains of C. jejuni; this variation is partly due to phase variation. The capsular polysaccharide of C. jejuni strain 11168 used in our studies has 6- methyl-D-glycero-a-L-glucoheptose, B-D—glucouronic acid modified with 2-amino- 2-deoxyglycerol, B-D—GalfNAc and B-D—ribose (154), and possesses a novel modification on the GalfNAc (157, 183). Capsular polysaccharides of C. jejuni have been shown to play an important role in the INT407 cell invasion model in vitro. A capsular polysaccharide-deficient strain of C. jejuni (kpsM mutant) displayed a significantly reduced adherence and invasion phenotype upon challenge of cultured INT407 cells (12). Correspondingly, Bacon et al. (2001) showed in a ferret diarrheal disease model that capsular polysaccharides of C. jejuni contribute to virulence. None of the ferrets inoculated with the kps mutant of C. jejuni developed diarrhea whereas the wild type 81 -1 76 capsule-sufficient strain caused diarrhea in 50% of ferrets (12). Furthermore, capsular polysaccharides contributed to bacterial resistance to host anti-microbial mechanisms. C. jejuni capsular polysaccharides appeared to protect the bacterial 12 cells to a certain extent from the bactericidal effects of constitutively expressed epithelial-antimicrobial peptides human B-defensin 1 (hBD) and lysozyme but not hBD—Z and hBD -3 (190). More research is warranted to conclusively understand the role of capsular polysaccharides in host-pathogen interactions and immune evasion by C. jejuni. Lipooligosaccharide (LOS) Lipopolysaccharide (LPS) is a key molecule associated with virulence and immunogenicity of Gram-negative bacteria. The general architecture of LPS is that a lipid A moiety anchored to the cell wall is covalently attached to highly conserved inner and outer cores of sugars (45). A polymer of repeating saccharide subunits called the O-polysaccharide, or O-chain, is attached to the outer core (45). C. jejuni lipid A is antigenically similar to that of enterobacterial pathogens such as Escherichia coli and Salmonella (120). But, unlike other enterobacterial lipid A, C. jejuni lipid A contains 2,3-diamino-2,3-dideoxy-D- glucose (GIcN3N) instead of glucosamine residues. Also, C. jejuni LOS has only a short non-repeating polysaccharide unit instead of the O-polysaccharide typically found in LPS. The inner core region of C. jejuni LOS is comprised of an unique tetrasaccharide (Glc—Hep—Hep—Kdo) and a trisaccharide that also occurs in the inner core of other bacterial species (120). Sialic acid substitutions can occur in the inner and outer core oligosaccharides of C. jejuni LOS, which lead to the molecular mimicry between C. jejuni LOS and ganglioside structures in host nerve tissues (75, 120). Besides playing a role in the development of 688 in 13 infected patients, LOS of C. jejuni induces strong proinflammatory cytokine responses from human dendritic cells (67). Genetic variation in C. jejuni strains The gene content of C. jejuni strains is highly variable, which is partly due to the exchange of DNA between bacterial cells as well as variation that occurs within the genome itself (183). C. jejuni are naturally competent, allowing the transfer of genomic DNA from other C. jejuni strains (179). Furthermore, the C. jejuni genome possesses several homopolymeric tracts (129)— repetitive sequences ofa single nucleotide—that can give rise to slip-strand mispairing during replication. This mechanism can result in variation in surface antigenic structures of C. jejuni, including flagella and LOS. Consequently, the pathogenicity of C. jejuni strains varies significantly: a recent study in our laboratory demonstrated that various strains of C. jejuni that were isolated from different sources, such as chicken and cattle, and belong to six different multi- Iocus sequence types, vary in their ability to colonize and cause disease in IL-10 deficient mice (1 5). 14 HOST DEFENSE RESPONSES TO C. JEJUNI Interaction of C. jejuni with innate cells Intestinal epithelial cells (IE C) Epithelial cells lining the mucosal tracts form the first line of defense against invading pathogens. The innate responses from epithelial cells not only provide a barrier to the invading pathogenic microbes but also educate the antigen-presenting cells to induce appropriate adaptive responses (33). A relatively well-characterized aspect of C. jejuni-host interplay is the C. jejuni-IEC interaction. C. jejuni has been shown to invade IEC by both actin- and microfilament-dependent pathways (16, 124). It has also been shown that lipid rafts or caveolae on the cell membrane, which are enriched for signaling molecules such as receptor tyrosine kinases, play a role in C. jejuni internalization into IEC (175). C. jejuni is capable of surviving inside IEC for a prolonged period of time, and Watson et al. (2007) demonstrated a possible mechanism underlying this survival. They found that after internalization, C. jejuni-containing vacuoles deviate from the canonical endocytic pathway, thereby avoiding fusion with Iysosomes and bacterial degradation. The unique pathway of entry of C. jejuni involving lipid rafts or caveolae is suggested to be central to the escape of C. jejuni-containing vacuoles from being delivered to Iysosomes (1 75). Cultured IEC lines such as INT407 and T84 and human colonic tissue explants infected with C. jejuni secrete lL-8. This cytokine is a chemotactic factor 15 that recruits neutrophils, macrophages, and other immune cells to the site of infection (176). Two mechanisms have been suggested by which C. jejuni may activate lL-8 production by epithelial cells: 1) adhesion to and invasion of cells by C. jejuni (60), “and 2) direct action of CDT on epithelial cells (62). It has been shown that signaling by ERK kinase—a member of the mitogen-activated protein (MAP) kinases—is essential for lL-8 production by intestinal epithelial cells infected with C. jejuni in vitro (176). C. jejuni infection of epithelial cells was shown to activate nuclear translocation of NF-KB, a transcription factor that mediates the production of various chemokines and cytokines involved in innate immunity (113). Consistent with this finding, NF-KB signaling has been shown to be essential for the development of Th1-associated antibody responses against C. jejuni in CS7BL/129 mice (48). The chemokine responses of intestinal epithelial cells after C. jejuni infection have also been analyzed. C. jejuni was shown to up-regulate the expression of macrophage inflammatory protein 1, monocyte chemoattractant protein 1, and gamma interferon-inducible protein 10 in INT407 cells within 4 h of infection (68). Intestinal epithelial cells also secrete antimicrobial peptides such as human beta defensins 2 and 3 in response to C. jejuni infection in vitro; these molecules have been suggested to play a role in limiting the infection (188). Swine intestinal epithelial cells, IPEC-1, secreted lL-1B, TNF-a, lL-6, IL-8 and IL- 18 after C. jejuni infection (G. Parthasarathy, K. Jones, L. Cunningham et al. unpublished data). In a swine rectal challenge model, C. jejuni induced Upregulation of several proinflammatory cytokines such as lL-8, lL-6, lL-18 and 16 TNF-a and iNOS in the distal colon and lymphoglandular complexes from 1 — 48 h after infection (K. Jones, unpublished data). Neutrophils Neutrophilic exudates are a hallmark of C. jejuni-induced intestinal disease in both human beings and animal models (48, 108, 149). However, little is known about the interaction between C. jejuni and neutrophils. Using a green fluorescent protein-tagged C. jejuni strain, Mixter et al. (2003) reported that after intraperitoneal inoculation of BALB/c mice, C. jejuni was mostly associated with CD11b+ Gr1” neutrophils recovered by peritoneal lavage (114). This finding suggests that murine neutrophils can recognize and internalize C. jejuni in vivo. Furthermore, fluorescence associated with intracellular bacteria in these cell types decreased over time indicating the failure of C. jejuni to survive within neutrophils (114). Consistent with this finding, another study showed that C. jejuni strains elicited production of antimicrobial molecules such as oxidative metabolites by neutrophils to varying degrees. Furthermore, opsonization of C. jejuni with human serum increased phagocytosis by neutrophils, which was accompanied by enhanced intracellular production of these oxidative metabolites (167). These studies indicated that complement opsonization of C. jejuni facilitates killing of intracellular bacteria by phagocytes such as neutrophils. 17 Macrophages and monocytes Macrophages have been suggested to play an important role in resistance to C. jejuni as depletion of macrophages resulted in mortality in more than 50% of NMRI mice infected with C. jejuni (14). Consistent with this finding, cultured macrophages, unlike IEC, have been demonstrated to be efficient in killing internalized C. jejuni (70, 173, 175). In line with these findings, infection of cultured murine macrophages with C. jejuni induced more than a 100-fold increase in the expression of inducible nitric oxide synthase (N082) (70), which is involved in the synthesis of an importantanti-microbial molecule, nitric oxide, in phagocytic cells. Furthermore, wild type macrophages capable of producing high levels of inducible nitric oxide were more efficient in killing C. jejuni than N082- deficient macrophages (70). Also, C. jejuni triggered proinflammatory responses from murine macrophages and the human monocytic cell line THP—1, characterized by the production of cytokines such as lL-6 and TNF—a (84). Furthermore, cytokine responses of THP-1 cells were found to be independent of C. jejuni viability and presence of cytolethal distending toxin (84). Like other enteric pathogens such as Salmonella and Shigella, C. jejuni induced apoptosis of infected THP-1 macrophages (148). But C. jejuni-induced apoptosis appeared to be delayed because significant levels of cell death were only observed 24 h after infection. Furthermore, maximal apoptosis induction by C. jejuni required Campy/obacter invasion antigen (CiaB) and proteinase K- and heat-resistant bacterial component(s) but was independent of caspase-1 and caspase-9 activation in host cells (148). In contrast, another study reported that 18 C. jejuni CiaB was dispensable for triggering apoptosis of infected 288C human monocytic cells (61). Therefore, the mechanisms underlying C. jejuni-induced apoptosis remain unclear. Even though apoptosis of C. jejuni-infected macrophages has been suggested to favor the host by eliminating a potential niche for C. jejuni in macrophages (148), the true significance of apoptosis- induction by C. jejuni is not known. Humoral and cell-mediated immunity against C. jejuni The two different outcomes of C. jejuni infection in immunocompetant and immunocompromised patients show that host immune responses are an important determinant of the outcome of C. jejuni infection. Furthermore, oral inoculation of SCID mice—that lack adaptive immune responses—with C. jejuni resulted in persistence of C. jejuni in the intestine and severe inflammation in the cecum and colon (27). This finding suggests that acquired immunity is essential to protect mice of this genotype from C. jejuni-induced intestinal disease and to clear C. jejuni colonization. Humoral immune responses to C. jejuni have been characterized in human infections and mouse models. C. jejuni elicits a strong antibody response in humans: serum lgA levels peak at 7 to 10 days after the onset of symptoms but decline rapidly thereafter, whereas serum IgG levels peak after 3 to 4 weeks of infection and persist long term (76). Furthermore, anti-C. jejuni lgA antibodies have been detected in a variety of samples from infected hosts including milk, saliva, urine and feces (99, 144). The surface exposed components of C. jejuni such as PEB1, capsular polysaccharide antigens, and 19 cytolethal distending toxin were found to be immunogenic, with C. jejuni flagellin being the immunodominant antigen (76). Consistent with human infections, specific lgA, IgM, and lgG antibody responses were elicited against C. jejuni in murine models of infection. IL-10"’+ and lL-10"‘ mice of C57BU6 (108), C3H and non-obese diabetic (NOD) genetic backgrounds (110) and C57BL/129 mice (48) challenged orally with C. jejuni exhibited a predominantly Th1-type associated lgGZb antibody response. However, such robust antibody responses did not protect the mice from C. jejuni-induced disease in the context of IL-10 deficiency (108). In summary, the role of specific immunoglobulins in protection against C. jejuni remains controversial. The higher incidence and persistent nature of C. jejuni infections in individuals with AIDS indicates that cell-mediated immunity plays an important protective role against C. jejuni infection. However, little is known about cell- mediated immunity to C. jejuni infection compared to infection with other enteric bacterial pathogens such as Salmonella and Listeria. It is yet to be determined which component of the cell-mediated immune response i.e., CD4 T cells, CD8 T cells, NK cells or a combination of these, is activated by C. jejuni infection and mediates protection against it. 20 DENDRITIC CELLS Dendritic cells (DCs) are antigen-presenting cells that act as sentinels in body tissues for the recognition of foreign infectious agents. DCs are present as immature cells in peripheral tissues and are efficient in pathogen recognition and uptake because they are equipped with pathogen recognition receptors (PRRs) (89). Upon capturing microbial pathogens, DCs become mature, migrate to draining lymph nodes, and present antigens to naive CD4+ T cells (32, 72, 89, 140). Extensive research in the last decade has demonstrated that DCs play an instructive role in the development of appropriate T cell responses to pathogens (74). Depending upon the nature of the pathogen and pathogen-associated molecular patterns (PAMPs), DCs provide different kinds of signals, particularly cytokine signals, to naive CD4+T cells, which then drive them towards a Th1, Th2, TM? or T regulatory phenotype (86). Furthermore, mucosal 003 have the ability to discriminate and induce tolerance towards self-antigens and intestinal microflora (106, 121 ). On the other hand, pathogens have evolved ‘a multitude of strategies’ (116) to evade and subvert DC functions, which include the suppression of DC maturation and migration and hampering of cytokine production (1 1 1 , 1 16, 164). Thus, given the facts that DCs are central to the induction of antigen specific acquired immune responses and that infectious agents have the ability to interfere with DC functions, it is of paramount importance to understand DC-C. jejuni interactions. 21 Dendritic cell development The development of DCs is one of the most complex phenomena in Iimmunobiology because of the existence of multiple subsets of DCs and the flexibility of DC developmental pathways. DCs and related cell types, monocytes and macrophages, develop from progenitor cells in the bone marrow through intricate pathways. The latest findings by Liu et al. (2009) provide evidence for various stages in DC development in vivo and also for the stage at which DC and monocyte developmental pathways diverge (104). According to their model, a myeloid progenitor (MP) population gives rise to macrophage and DC precursors (MDP) that differentiate into common DC precursors (CDP) and monocytes. The point of divergence between DCs and monocytes thus appears to be at the MDP stage. CDP differentiates into plasmacytoid DCs (pDCs) and conventional DCs precursors (pre-cDC), which later give rise to cDCs. Furthermore, the terminal differentiation of cDCs in the peripheral lymphoid tissues is controlled by regulatory T cells in a FMS-like tyrosine kinase 3 (Flt3)—dependent manner (104). Pathogen sensing by DCs DCs are equipped with a set of germ-line encoded receptors known as pattern recognition receptors (PRRs) that recognize highly conserved components associated with pathogens. PRR expressed by DCs include Toll-like receptors (TLRs), nucleotide oligomerization domain (NOD)-like receptors (NLRs), and C-type lectin-Iike receptors (CLRs) (4). Various combinations of these PRR combined with their intracellular and surface locations allow DCs to 22 recognize and respond to almost any kind of pathogen: viral, bacterial, fungal, or parasitic. Highly orchestrated signaling cascades triggered by the PRR induce inflammatory and innate immune responses that regulate antigen presentation and the ensuing adaptive immune responses (4, 74). Pathogen sensing by DCs and the signaling pathways involved are described in detail later in the chapter with the main focus on TLR signaling. Antigen presentation by DCs Antigens in the cytosol and phagocytic vesicles are processed in two different ways (77). In the first pathway, endogenous proteins in the cytosol—for example, viral proteins synthesized by cellular machinery—are degraded by the proteosome complex of the cell into peptides that are translocated into the lumen of the endoplasmic reticulum (ER) by transporters associated with antigen processing (TAP), a heterodimer of TAP1 and TAP2. Then, the peptide binds to the partly folded MHC class I molecule—which is present as a complex with TAP and a chaperone molecule in the ER—resulting in complete folding of the MHC class I molecule that is subsequently released from the complex. This MHC-I complex with the peptide is then translocated onto the cell surface through the Golgi apparatus and presented to CD8+ T cells. In the second pathway, following internalization, pathogens and their products are subjected to limited proteolysis in phagosomes by proteases such as cathepsins B, D, S and L to generate short peptides. The peptides thus generated are loaded onto the peptide groove in the MHC class II molecule after the release of the CLIP fragment from the MHC 23 class II molecule by HLA-DM (an MHC class Il-like molecule in the lysosome). The MHC-II molecules with the antigenic peptides are exported to the cell surface and presented to CD4+ T cells (77). Besides these classical pathways of antigen presentation, a new pathway of cross presentation has been identified. Cross-presentation is a mechanism by which DCs present exogenous antigens derived from the extracellular milieu in the context of an MHC-l molecule instead of the conventional MHC-ll (143). Cross presentation represents a novel mechanism of generating effector CDB T cell responses by DCs to exogenous antigens, such as bacterial pathogens. Recently it has also been shown that cross presentation of apoptotic cells by D05 is critical for the induction of tolerance against self-antigens (143). DC-driven polarization of naive CD4*T cells A large body of evidence clearly shows that DCs are the key cell type that determines the nature of effector CD4“ T cell responses such as Th1, Th2, regulatory T cells, or the recently described Th17 responses (86, 116, 138). This capacity of DCs is primarily due to their ability to differentially recognize various pathogens and convey distinct combinations of signals to naive T cells. Potent activation of T cell responses by DCs requires delivery of three signals to naive CD4’ T cells: signals 1 and 2 are MHC-ll plus antigen complex and costimulatory molecules, respectively. lmportantly, DCs deliver a third instructive signal, mostly cytokines, which are largely responsible for inducing the effector T cell phenotype (86). In general, upon recognition of intracellular bacteria such as Salmonella 24 lyphimurium, DCs express lL-12, lL-18, or CD70 promoting IFNay-secreting Th1 cells. Infection with parasitic helminths induces DCs to up-regulate molecules like OX40L, which primes Th2-differentiation of naive CD4+ T cells (86). Furthermore, intensive research during the last few years demonstrated that DCs are/also capable of inducing Th17 cells, a novel subset of effector T helper cells which play an important role in mucosal immunity and autoimmune responses (9). In response to ATP produced by bacteria in the GI tract, a CD70"'9"CD11c'°‘” subset of DCs in the lamina propria secretes IL-6, lL-23 and TGF-B; the combination of these cytokines leads to development of lL-17- producing Th17 cells from naive CD4” T cells (9). 25 TOLL-LIKE RECEPTORS: PATHOGEN SENSING AND INITIATION OF IMMUNE RESPONSES Toll-like receptors (TLRs) are a type of pattern recognition receptor that recognize conserved molecular patterns associated with a broad range of pathogens and initiate protective immune and inflammatory responses against them. Various cell types, particularly professional antigen-presenting cells, such as dendritic cells, macrophages, and B cells, express TLRs (74). TLRs are type I integral membrane glycoproteins with extracellular and cytoplasmic domains. Extracellular domains are characterized by varying numbers of Ieucine-rich- repeat motifs, 24-29 amino acids in length. The cytoplasmic signaling domain is homologous to that of the IL-1 receptor (IL-1 R) and contains a Toll/IL-1 R (TIR) domain (91 ). Each TLR identifies a different microbial structure (Table 1); for example, TLR2 recognizes peptidoglycan and lipopeptides, TLR4 recognizes lipopolysaccharide (LPS), and TLR5 recognizes flagellin. Upon detecting pathogens, TLRs initiate intracellular signaling cascades through adapter molecules, which ultimately result in the production of proinflammatory cytokines and other immune responses (91). A growing body of evidence suggests that TLR2 and TLR4 signaling pathways play an essential role in the detection of invading bacterial pathogens and in mounting protective immune responses against them (63, 88, 162). 26 TLR4 and TLR2 signaling MyD88-dependent pathway Upon binding LPS, TLR4 undergoes dimerization leading to the recruitment of an adapter protein MyD88 via another molecule TIRAP. Similarly, recognition of specific ligands by TLR2 in combination with TLR1 or TLR6 recruits MyD88 via TIRAP. MyD88 activates lL-1R-associated kinase-4 (IRAK4), which then recruits IRAK1 (4). The downstream target molecule for IRAK1 is TRAF6, which is then polyubiquitinylated and activated by ch13 and Uev1A, leading to the activation of TAK1. In combination with TAB1, TABZ and TAB3, TAK1 activates the IKK complex (lKKa + IKKB + lKKy/NEMO), which catalyzes the phosphorylation of IKB proteins. Under normal conditions, IKB sequesters NF- KB in the cytoplasm thus preventing its activation and nuclear translocation. The phosphorylation of “(3 leads to its degradation, releasing NF-kB that translocates to the nucleus to activate transcription of a number of inflammatory genes. The IKK complex also activates MAP kinases leading to the activation of transcription factors such as AP-1 and c-Jun. Additionally, the My088-IRAK4-TRAF6 complex phosphorylates the IRF5 transcription factor, which then translocates to the nucleus to activate genes involved in proinflammatory responses. Thus, MyD88 signaling triggers robust Inflammatory responses through the activation of various transcription factors (91). 27 MyD88-independent pathway TLR4 also signals through a MyD88-independent pathway, which is mediated by TRIF, another adapter molecule (4). Following ligand binding, the TLR4 cytoplasmic domain recruits the adapter protein TRIF through TRAM. Medzhitov and colleagues recently demonstrated that MyD88 and TRIF signaling are sequentially activated by TLR4 (85). According to their model, activation of TLR4 by LPS first triggers TIRAP-My088 signaling from the cell membrane. Then TLR4-LPS is endocytosed, leading to displacement of TlRAP-MyD88 from TLR4 and termination of MyD88 signaling. TRIF is then recruited to the cytoplasmic domain of TLR4 in the endosomes via TRAM. leading to a second phase of signaling from TLR4. TRIF recruits TRAF3, which forms a complex with TBK1 and IKKi. This complex phosphorylates a transcription factor unique to the TRIF pathway, lRF-3, which mediates the expression of type I IFN and IFN- inducible genes. TRIF also interacts with TRAF6 and RIP1, which mediate NF-KB and MAP kinase activation as described above in the MyD88-dependent pathway (90). Thus, TLR4-TRIF and TLR4-My088 pathways activate a number of overlapping cellular responses, but activation of type I interferon responses is unique to the TRIF arm of TLR4 signaling. TLR4, TLR2, MyD88, and TRIF signaling in host defense As sensors of invading pathogens, TLRs expressed by DCs have been shown to play an instructive role in the development of adaptive T-cell immunity. 28 In murine models of Bordetella pertussis infection, TLR4 signaling was shown to activate antigen-specific regulatory T cells by inducing lL-10 production from DCs, which confers resistance by limiting inflammatory pathology (63). Similarly, TLR2 was described to have a protective role in Listeria monocytogenes infection in mice; infected TLR2-deficient mice had increased bacterial burdens and reduced survival rates (162). Furthermore, the maturation of and TNF-a production by BM-DCs following Francisella tularensis infection were found to be mediated via TLR2 (88). Host defense responses to various pathogens have been shown to be dependent on MyD88. For instance, MyDB8-deficient mice infected with Leishmania major failed to produce IL-12 and developed a Th2-type response instead of a protective Th1-type response and were therefore severely compromised in their resistance to infection (122). Furthermore, the production of TNF-a, lL-12 and nitric oxide and the expression of co-stimulatory molecules by BM-DCs and macrophages following L. monocytogenes infection were almost abolished in the absence of MyD88 (162). Correspondingly, MyDB8-deficient mice were highly susceptible to L. monocytogenes infection (162). In contrast to My088, the role of TRIF signaling in mediating protection against bacterial pathogens is just beginning to be understood. Recent studies have shown that TRIF is essential for pulmonary host defense against Gram-negative bacteria such as Pseudomonas aeruginosa and Escherichia coli (78, 137). However, the role of TLR2, TLR4, MyD88, and TRIF signaling in host defense against C. jejuni infection remains largely unknown and thus formed a large part of the focus of my work. 29 TLR recognition of C. jejuni C. jejuni has been reported to possess multiple Iipoproteins, including CjaA, HisJ, and Omp18 as well as leA. Even though none of these have been demonstrated to be recognized by TLR2, the presence of multiple lipoproteins suggests that C. jejuni has potential ligands that can activate TLR2 signaling. Indeed, Friss et al. (2009) recently reported that TLR2 signaling was necessary for C. jejuni-induced lL-6 production by cultured IEC (50). C. jejuni flagellin is not recognized by TLR5, and Andersen-Nissan et al. (2005) reported that this immune evasion is due to mutation in the TLR5 recognition site of C. jejuni flagellin (6). Furthermore, owing to the low frequency of CpG motifs in its genomic DNA, C. jejuni is a poor inducer of TLR9 signaling (38). Accordingly, C. jejuni DNA was found to be dispensable for triggering lL-8 secretion from IEC in vitro (187). It has also been shown that My088 signaling is necessary for the innate inflammatory responses of intestinal epithelial cells and antigen-presenting cells following infection with C. jejuni (177, 187). 30 RATIONALE FOR THIS STUDY Our understanding of the interaction between C. jejuni and DCs, a key cell type that bridges innate and adaptive immunity, is very limited. Hu et al. (2006) reported that human monocyte-derived DCs recognize C. jejuni, undergo maturation, and secrete proinflammatory cytokines (67). However, the ability of C. jejuni-infected DCs to activate T cell responses—a primary function of DCs-is not known. Furthermore, the TLRs that recognize C. jejuni and mediate DC responses against C. jejuni remain unknown. Also, the relative contribution of MyD88—dependent and -independent pathways to the activation of DCs has yet to be determined. The main objective of this study was to address these knowledge gaps. This study was designed with the following specific aims to investigate the interplay of C. jejuni with murine DCs and its impact on the development of subsequent T helper responses and to understand the mechanism of activation of C. jejuni-infected murine DCs. Specific Aim 1: To characterize the interaction of C. jejuni with murine bone- marrow derived-DCs (BM-DCs) with respect to internalization and processing of C. jejuni and maturation of DCs. Hypothesis: Murine BM-DCs take up C. jejuni, process it, and undergo maturation in vitro. 31 Specific Aim 2: To determine whether the C. jejuni-stimulated murine BM-DCs produce T helper (Th) cell-polarizing signals and induce Th polarization in vitro. Hypothesis: C. jejuni-stimulated murine BM-DCs produce Th1-polarizing signal(s) and thereby induce Th1-type immune response in vitro. Specific Aim 3: To determine the role of TLR2, TLR4, MyD88, and TRIF in the activation of BM-DCs by C. jejuni. Hypothesis: C. jejuni-induced activation of bone marrow-derived DCs (BM-DCs) is mediated by TLR2 and TLR4, and that MyD88 and/or TRIF signaling is necessary for this activation. The findings for specific aims 1 and 2 are described in chapter 2. Chapter 2 also describes the bacterial factors that contribute to C. jejuni-induced activation of DCs. The findings from specific aim 3 are covered in chapter 3. Finally, chapter 4 summarizes the findings reported in chapters 2 and 3 and discusses future investigations suggested by this study. 32 Table 1. TLR ligands TLRs Ligands Peptidoglycan of Gram-positive bacteria; porins of Neisseria; lipoarabinomannan of Mycobacteria; TLR2 phospholipomannan of Candida albicans; hemagglutinin protein of measles virus TLR2 and TLR1 Triacyl Iipoprotein Diacyl Iipoprotein; zymosan; lipoteichoic acids from group TLR2 and TLR6 B Streptococcus TLR3 Double-stranded RNA from viruses Lipid A of LPS; DnaK from Francisella tularensis; TLR4 fusion protein from respiratory syncytial virus; envelope protein from mammary tumor virus TLR5 Flagellin TLR7 Single-stranded RNA TLR8 Single-stranded RNA TLR9 CpG DNA, hemozoin Profilin like protein, uropathogenic E. coli TLR1 1 components 33 Activation of Type I interferon Inflammatory genes response Figure 1. TLR2 and TLR4 signaling 34 CHAPTER 2 Dendritic cells from C57BU6 mice undergo activation and induce Th1- effector cell responses against Campylobacterjejuni Rathinam, V. A., K. A. Hoag, and L. 8. Mansfield. 2008. Dendritic cells from C57BU6 mice. undergo activation and induce Th1-effector cell responses against Campy/obacterjejuni. Microbes Infect 10: 1 316-1 324. 35 ABSTRACT Food-borne Campy/obacterjejuni is an important cause of enteritis. We showed that CS7BL/6 mice and congenic interleukin (lL)-10"' mice serve as models of C. jejuni colonization and enteritis, respectively. Thus, C57BU6 mice are resistant to C. jejuni-induced disease. Because dendritic cells (DCs) are central to regulating adaptive immune responses, we investigated the interaction of C. jejuni with murine bone marrow-derived DCs (BM-DCs) to assess bacterial killing, DC activation, and the ability of C. jejuni-infected BM-DCs to stimulate Campy/abacter-specific T cell responses in vitro. BM-DCs challenged with C. jejuni efficiently internalized and killed C. jejuni 11168 and significantly up- regulated surface MHC-II, CD40, CD80 and CD86 demonstrating a mature phenotype. Infected BM-DCs secreted significant amounts of tumor necrosis factor-a, lL-6 and lL-12p70. Formalin-killed C. jejuni also induced maturation of BM—DCs with similar cytokine production but at a significantly lower magnitude than live bacteria. Maximal activation of murine BM-DCs required internalization of C. jejuni; attachment alone was not sufficient to elicit significant responses. Also, various strains of C. jejuni elicited different magnitudes of cytokine production from BM-DCs. Finally, in a coculture system, C. jejuni-infected BM- DCs induced high-level interferon-y production from CD4“ T cells indicating Th1 polarization. Thus, DCs from resistant C57BU6 mice initiate T cell responses against C. jejuni. 36 INTRODUCTION Campylobacterjejuni is an common bacterial agent of enteritis worldwide. Campylobacteriosis usually presents as self-limiting intestinal illness with the clinical spectrum ranging from mild watery to severe bloody diarrhea. However, persistent and systemic illnesses can develop in immunocompromised patients (1 71 ). C. jejuni invades the intestinal mucosa (108, 166) and resides inside intestinal epithelial cells (IEC) (166) and in the lamina propria causing pathology predominantly in colon (166) and, in mice the cecum (108). DCs densely populate the lamina propria, are in close proximity to the epithelium, and can form transepithelial dendrites to sample the lumen for microbes (123). We observed C. jejuni associated with mononuclear cells morphologically similar to DOS in the lamina propria and submucosa of infected C57BU6 IL-10"' mice with enteritis (108). The strategic location of DCs in the subepithelial area and the invasive nature of C. jejuni (108, 166) together with our recent observations suggest that DCs in the lamina propria are likely candidates for capture and processing of C. jejuni for antigen presentation. Furthermore, Johanesen and Dwinell demonstrated that infection of cultured IEC with C. jejuni upregulates secretion of the DC chemokine CCL20, suggesting this as a mechanism of increased DC recruitment into the intestinal mucosa (81 ). Therefore, it is crucial to understand DC-C. jejuni interactions and the role of DCs in stimulating host defense against C. jejuni. 37 Human monocyte-derived DCs were recently shown to undergo maturation and secrete proinflammatory cytokines subsequent to C. jejuni 81-176 challenge in vitro (67); but to our knowledge, the resultant antigen-presenting capacity of DCs for CD4+ T cell polarization has not been investigated. We hypothesized that murine bone marrow-derived DCs (BM-DCs) internalize C. jejuni, which induces BM-DCs to undergo activation and initiate a Th1-type immune response. In this study, we investigated activation of BM-DCs by C. jejuni, mechanisms mediating the activation and the ability of C. jejuni-infected BM-DCs to stimulate T cell responses. 38 MATERIALS AND METHODS Mice CS7BU6J mice purchased from The Jackson Laboratory (Bar Harbor, ME) were bred, maintained, and monitored as described (108), and used at 8—12 weeks of age. Animal protocols were approved by MSU Institutional Animal Care & Use Committee conforming to NIH guidelines. Bacteria and inoculum preparation C. jejuni strains were grown on Bolton agar (BA) plates at 37 °C in 10% C02. Bacterial growth was resuspended in R10.2 medium (RPMI 1640 Dutch modification [Sigma-Aldrich, St. Louis, MO] with 2 mM L-glutamine [lnvitrogen, Grand Island, NY] and 10% fetal bovine serum [FBS; lnvitrogenl) to an optical density of 0.12 at 560 nm (~2 x 108 colony forming units [CFU]/ml). Where needed, bacteria were inactivated by treatment with 2% formaldehyde in Hank’s Balanced Salt Solution (HBSS) for 30 min at 37 °C. CFU in the inocula and bacterial killing were confirmed by limiting dilution on BA plates. Generation of BM-DCs BM-DCs were generated as described with minor modifications (21, 105, 186). Bone-marrow cells from femurs and tibiae were cultured in bacteriological- grade Petri-dishes in 10 ml (2.5 x 105 cells/ml) of R10 medium (R10.2 medium containing 50 uM 2-mercaptoethanol, 100 U/ml penicillin, 100 uglml streptomycin [lnvitrogen] and 20 ng/ml murine granulocyte-macrophage colony-stimulating 39 factor [Peprotech, Rocky Hill, NJ]). Non-adherent cells in the culture were discarded on days 3 and 6. On day 3, 10 ml of fresh medium was added; on days 6 and 8, 10 ml of medium was exchanged for fresh medium. DCs were collected and used on day 9. The proportion of DCs in the culture was consistently >90% as determined by flow cytometric analysis of CD110 and MHC-ll markers. Gentamicin killing assay This assay was performed as described with minor modifications (173). Bacteria diluted in R10.2 medium were added to 2 x 105 BM-DCs per well in a 24-well plate (BD Falcon, Bradford, MA) at multiplicity of infection (MOI) of 10 or 100 bacteria per BM-DC. After 1 h of incubation, non-phagocytosed bacteria were removed by washing the cells and resuspending in R10 medium containing 250 ”9”!“ gentamicin but without penicillin and streptomycin. After 1 h, cells were washed and resuspended in R10 medium without antibiotics. Viable intracellular bacteria were enumerated at 2, 4 and 8 h post-infection (p.i.) by lysing the cells with 0.5 ml of 0.1% Triton X-100 in phosphate-buffered saline for 15 min and spreading serial dilutions of lysate on BA plates. Colonies were counted 72 h after incubation at 37°C with 10% C02. Assessment of maturation and cytokine secretion by C. jejuni-infected BM- DCs BM-DCs (2 x 105 cells/ml) were treated with c. jejuni (MOI 10:1 [in cytokine experiments only] or 100:1 ), R10.2 medium (negative control) or 0.1 40 uglml Salmonella enterica serovar Typhimurium lipopolysaccharide (LPS; positive control; Sigma-Aldrich). In some experiments, to inhibit internalization of C. jejuni, BM-DCs were preincubated with cytochalasin D (1 jig/ml) and nocodazole (20 uM; Sigma-Aldrich) or DMSO (solvent control, 0.1%) for 1 h and treated as described above. In other experiments, BM-DCs were also treated with formalin-killed bacteria. One h after infection, gentamicin (250 pglml) was added to all wells to kill extracellular bacteria. DCs were analyzed by flow cytometry at 24 h pi For cytokine analysis, supernatants were collected at 4, 24 and 48 h p.i. unless otherwise indicated. Flow cytometry Antibodies were from BD Pharmingen, San Diego, CA unless otherwise noted, including R-PE-conjugated anti-CD11c (HL3), F lTC-conjugated anti-MHC- ll (l-A") (AF6-120.1), FlTC-conjugated anti-CD40 (3/23), FITC-conjugated anti- C080 (16-10A1), PE-Cy5-conjugated anti-CD80 (Biolegend, San Diego, CA), F ITO-conjugated anti-CD86 (GL1), PE-Cy5-conjugated anti-CD86 (Biolegend) and FITC-conjugated anti-CD4 (RM4—5) along with corresponding isotype controls. Cells were incubated with Fc block (anti-mouse Fchl/lll) for 10 min at 4 °C. Fluorochrome-conjugated antibodies were added to cells and incubated for 30 min at 4 °C. The cells were washed twice with HBSS containing 1% FBS and 0.1% sodium azide and analyzed with a FACS Vantage flow cytometer (BD Biosciences). 41 DC-CD4’T cell coculture CD4+ T cells were negatively selected from splenocytes using the B0” IMag Mouse CD4 T Lymphocyte Enrichment Set-DM (BD Pharrningen) according to manufacturer’s instructions. The proportion of CD4+ T cells was enriched to > 90% as measured by flow cytometry. BM-DCs in complete DMEM containing 25 mM HEPES, 10% FBS, 2 mM L-glutamine, 50 uM 2-mercaptoethanol, 1 mM sodium pyruvate, and 1% MEM non-essential amino acids seeded in 24—well plates (105 cells/well) were treated with one of the following: C. jejuni (MOI of 100:1), R10.2 medium, S. Iyphimurium LPS (0.1 ug/ml; inducer of IFN-y producing Th1 cells), or zymosan (10 ug/ml; inducer of IL-4 production from Th cells; Sigma-Aldrich). One h after infection, gentamicin (250 jig/ml) was added to all wells. At 24 h p.i., CD4+ T cells in complete DMEM supplemented with 100 U/ml penicillin and 100 pg/ml streptomycin were added to DCs at a DC-to-T cell ratio of 1:10. After coculturing for 72 h, supernatants were collected for measurement of IFva, IL-4 and IL-10. ELISA Cytokines in culture supernatants were measured using Ready-SET-Go! ELISA Sets (eBioscience, San Diego, CA) according to manufacturer’s instructions. Assay detection limits (in pg/ml) were lL-12, 15; lL-10, 30; lL-4, 4; IL- 6, 4; TNF-a, 8; IFN-y, 0.8. 42 Statistical analysis One-way or two-way analysis of variance followed by the Holm-Sidak correction for multiple comparisons was performed using SigmaStat 3.1 (Systat Software, San Jose, CA). P values < 0.05 were considered significant. 43 RESULTS BM-DCs internalize and kill C. jejuni Within 2 h p.i., BM-DCs internalized C. jejuni in a dose-dependent manner (Fig. 1A). Approximately 5 i 0.8 x 103 and 7.38 i 0.2 x 104 viable intracellular bacteria were recovered at M0ls of 10:1 and 100:1, respectively, 2 h pi (Fig 1A), which decreased rapidly to 1 a 0.18 x 103 and 1.3 a 0.0076 x 104 for M0ls of 10:1 and 100:1, respectively, by 4 h p.i.; no viable bacteria were recovered at 8 h p.i. at either MOI. In contrast, CFU of C. jejuni inoculated into R10 medium without antibiotics and DCs increased by several-fold at 8 h p.i. (Fig. 1B). BM-DCs undergo maturation following C. jejuni infection In experiments to test BM-DC maturation, mean fluorescence intensities (MFI) indicating the levels of expression of MHC-II, CD40, C080 and CD86 were significantly higher by 2.1-, 1.9-, 3.7-, and 2.5-fold, respectively, in C. jejuni- infected BM-DCs compared to uninfected cells (Fig. 2A). Furthermore, infection with C. jejuni resulted in a significant (P < 0.05) increase in the proportion of BM- DCs expressing these markers (Fig. 2A). In all cases, these levels were comparable to those induced by LPS. C. jejuni-infected BM-DCs secrete IL-12p70 and proinflammatory cytokines C. jejuni induced a dose-dependent increase in lL-12p70 secretion at 24 and 48 h p.i. (Fig. 2B). Peak levels of lL-12p70 secretion were observed with the higher dose of C. jejuni at 24 h p.i. In contrast, the lower dose of C. jejuni induced 44 markedly lower amounts of lL-12p70 at 24 and 48 h p.i. C. jejuni also induced dose-dependent production of lL-6 and TNF-a by BM-DCs at 4, 24 and 48 h p.i. (Fig. 2B). At 4 h p.i., moderate production of lL-6 was induced by the higher dose of C. jejuni, which increased at later time points (24 and 48 h). At the lower dose of bacteria, a similar trend was observed in IL-6 secretion but at a significantly lower magnitude (Fig. 2B). BM-DCs challenged with C. jejuni did not produce detectable quantities of lL-10 at any of the three time points studied (data not shown). C. jejuni viability is required for maximal activation of BM-DCs Infection with killed C. jejuni also resulted in upregulation of maturation markers; however, live C. jejuni induced significantly higher (P < 0.05) levels of expression of CD80 and CD86 than formalin-killed C. jejuni. Similarly, a significantly higher proportion (P < 0.05) of live C. jejuni-exposed DCs expressed CD40 and CD86 than DCs treated with formalin-killed C. jejuni (Fig. 3A). However, the level of MHC-II expression and proportion of DCs expressing MHC- II or CD80 were not significantly different (P > 0.05) between live- and formalin- killed C. jejuni-exposed DCs (Fig. 3A). Bacterial viability also affected cytokine secretion. At 24 and 48 h p.i., live C. jejuni-exposed DCs secreted 4.7- and 3.2-fold higher levels of lL-12 and 4.5- and 4.2-fold higher levels of IL-6 than DCs exposed to formalin-killed C. jejuni (Fig. 33). Live C. jejuni elicited significantly greater (P < 0.05) TNF-a production from DCs than formalin-killed C. jejuni at all time points. Like live C. jejuni, 45 formalin-killed C. jejuni did not elicit detectable IL-10 secretion from DCs at 4, 24 or 48 h p.i. (data not shown). Full activation of BM-DCs requires contact of C. jejuni with DCs and bacterial internalization C. jejuni entry into IEC has been suggested to involve both actin filaments and microtubules (125). Therefore, we used a combination of cytochalasin D (1 uglml) and nocodazole (20 pM), inhibitors of actin and microtubule polymerization respectively, to inhibit phagocytosis of C. jejuni by DCs. This treatment of DCs resulted in about 80% inhibition of uptake of C. jejuni as assessed by the gentamicin killing assay, which was markedly higher than that observed with either inhibitor alone (Fig. 1C). Preincubation with inhibitors alone caused a significant increase in the MFI of MHC-ll and CD80 and in the proportion of CD40+ and C086" DCs as shown previously (115). Comparison of inhibitor-treated and non-treated DCs infected with C. jejuni showed that pretreatment with cytochalasin D and nocodazole abolished the increase in MHC-ll expression level and markedly reduced CD40, C080 and CD86 expression levels and the proportion of DCs expressing CD40, CD80 and CD86 (Fig. 4A-B). Blocking DC phagocytosis of C. jejuni with cytochalasin D and nocodazole completely inhibited IL-12 secretion and significantly (P < 0.05) reduced TNF-oi secretion, but did not inhibit IL'6 production. The effect of inhibitors on Salmonella LPS-induced expression of 46 surface markers and cytokines was comparable to that observed for C. jejuni- induced responses. To test whether activation of DCs depends on direct contact with C. jejuni or whether a diffusible bacterial product is sufficient to stimulate DC responses, C. jejuni and DCs were separated by a 0.2 uM transwell insert, which allowed passage of bacterial products across the membrane but not intact bacteria. The presence of a membrane between DCs and C. jejuni resulted in a significant (P < 0.05) reduction in MHC-ll and CD80 expression levels and in the number of CD40“, 0080*, and CD86+ DCs. Furthermore, membrane presence abolished the increase in CD86 expression level and in the proportion of DCs expressing MHC-II after C. jejuni challenge (Fig. 4C-D). lmportantly, the prevention of direct contact between C. jejuni and DOS resulted in abrogation of lL-12, TNF-a and IL- 6 production (Fig. 4E). However, the presence of membrane only minimally affected DC expression of surface markers, TNF-a and lL-6 in response to Salmonella LPS. Conversely, Salmonella LPS-induced lL-12 secretion was significantly reduced by the presence of transwells (Fig. 4C-E). Various strains of C. jejuni differ in their ability to elicit cytokine responses from BM-DCs . We investigated whether different strains of C. jejuni elicit different cytokine responses from BM-DCs. lL-12 production by DCs exposed to various strains ranged from 107.9 i 1.3 to 310.5 :1: 8.6 pg/ml at 24 h p.i. and 70.2 i 2.4 to 231.7 i 7.3 pg/ml at 48 h p.i. (Fig. 5A). At both time-points, strain 33560 induced 47 the highest levels whereas strain NW induced the lowest levels of IL-12. Peak levels of lL-12 secretion were observed at 24 h p.i. with all strains, consistent with previous experiments with-strain 11168. All C. jejuni strains elicited TNF-a and IL-6 secretion; maximum levels of TNF-a and lL-6 were observed with strain 33560 and minimum levels were detected with strains NW or D0835 (Fig. 5B-C). The highest levels of TNF-a and IL-6 were detected at 24 and 48 h p.i. respectively for all strains. Strain 33560 induced a minimal amount of lL-10 (36.6 :I: 1.6 pg/ml) only at 24 h pi; no other strains induced detectable amounts of lL-10 (data not shown). C. jejuni-infected BM-DCs induce Th1 polarization of CD4”T cells Because Th-polarizing ability of C. jejuni-infected DCs is not known, an in vitro DC-CD4+ T cell coculture system was employed to assess this function. Negative control groups (T cells + medium alone, T cells + medium alone-treated DCs, and DOS + C. jejuni) did not produce detectable quantities of IFN-y (Fig. 6A). However, co-culture of C. jejuni-infected BM-DCs with CD4+ T cells induced marked production of IFNdy that was significantly higher (P < 0.05) than observed with all negative controls and comparable to that induced by S. typhimurium LPS- treated BM-DCs. In contrast, C. jejuni-infected BM-DCs did not elicit detectable lL-4 (Fig. 6B) or lL-10 (data not shown) secretion by CD4+ T cells. It is evident from this experiment that BM-DCs infected with C. jejuni induce Th1 polarization of CD4+ T cells in vitro. 48 DISCUSSION Pathogen-host cell interactions in C. jejuni infection remain largely unknown. We demonstrate here that C. jejuni induces phenotypic and functional activation of DCs from resistant C57BL/6 mice. C. jejuni viability and internalization were necessary for maximal activation of DCs. Furthermore, murine DCs primed C. jejuni-specific Th1-effector responses in vitro, consistent with this murine model where C. jejuni was limited to colonization of the gastrointestinal tract (108). Together these findings support a role for DCs in regulating innate and adaptive immune responses during C. jejuni infection. Our data show that murine DCs efficiently internalize and kill C. jejuni within 8 h p.i. and that C. jejuni internalization is mediated by both actin filaments and microtubules as suggested previously (125). Wassenaar et al. (173) reported failure of C. jejuni to survive inside activated human macrophages and suggested that “infra-phagocytic survival is not a common phenomenon” in C. jejuni infection. While C. jejuni persists within cultured IEC (175), our data and that of Hu et al. (67) suggest that near complete killing of C. jejuni by DCs likely rules out the possibility that DCs serve as transporters of C. jejuni for extra-intestinal dissemination. In our study, immature murine DCs infected with C. jejuni undergo maturation by upregulating surface expression of MHC-ll, CD40, and costimulatory molecules. This implies that C. jejuni-infected murine DCs are capable of engaging both T-cell receptors and CD28 molecules on naive Th cells, thereby delivering essential signals for induction of anti-microbial T cell 49 responses. In our studies, most of the unstimulated immature DCs (> 70 %) expressed CD80 but only a small fraction of unstimulated DCs (< 20—35 %) expressed C086. As a result of this basal difference, the magnitude of change due to C. jejuni stimulation and/or other treatment conditions, like presence of transwells, is distinct for CD80 and C086. That is, the magnitude of increase in proportion of cells expressing CD80 following C. jejuni infection is lower than that observed with CD86 (1.1—1.2-fold increase for C080 vs. 2.5—3-fold increase for C086 in our studies). Thus, we show for the first time the effect of C. jejuni viability on DC maturation. When compared to live C. jejuni, killed C. jejuni induced comparable levels of MHC-ll expression but significantly lower levels of costimulatory molecule expression, which may be due in part to very low amounts of TNF-a—a known stimulus inducing DC maturation—elicited by nonviable C. jejuni. We observed a C. jejuni dose- and time-dependent secretion of IL-12 by DCs. IL-12 secreted by DCs enhances CD4+ T cell proliferation and IFN-y production. Furthermore, recent studies demonstrated that lL-12 mediates DC activation of natural killer (NK) cells, contributing to initial resistance to bacterial pathogens (57, 92). This ability to elicit lL-12 suggests that C. jejuni may also stimulate NK cells, leading to mucosal resistance. C. jejuni-infected DCs secreted TNF-a and lL-6 suggesting that DCs may initiate or amplify the proinflammatory reaction to C. jejuni enhancing recruitment of neutrophils and macrophages to the intestinal mucosa. Indeed, neutrophilic exudates are a prominent feature of C. jejuni-induced disease in humans and 50 animals (48, 108, 149). DC-mediated recruitment of neutrophils to intestinal mucosa may contribute to C. jejuni killing as recently suggested in Aspergillus fumigatus infection (52). Additionally, killed C. jejuni elicited significantly lower levels of lL-12 and proinflammatory cytokines than live C. jejuni. Our data taken together with previous reports (60, 67, 84) demonstrate that, in contrast to IEC, phagocytes such as DCs and THP-1 monocytic cells do not require live C. jejuni to secrete proinflammatory cytokines. However, C. jejuni viability is necessary for maximal induction of cytokine secretion by murine DCs, unlike human DCs and monocytes. We also observed that the maturation and cytokine responses of DCs from C3H/He0uJ mice to C. jejuni were similar to that observed with DCs from C57BU6 mice (V. Rathinam and L. Mansfield, unpublished data). Blocking phagocytosis of C. jejuni resulted in inhibition of IL-12 and TNF-a but not lL-6 production by murine DCs, consistent with previous reports on Neisseria meningitidis and Streptococcus pneumoniae (31, 165). Helicobacter pylori-induced 0C secretion of lL-12 but not lL-8 also depended on phagocytosis (57). C. jejuni internalization was also essential for optimal surface expression of DC maturation markers. Transwell experiments showed that diffusible products secreted by C. jejuni induced only partial upregulation of maturation markers and did not elicit secretion of proinflammatory cytokines. Collectively, these findings show that internalization of C. jejuni was necessary for complete maturation of DCs and secretion of lL-12 and TNF-a but was dispensable for lL-6 production. A possible explanation for this requirement is that, following C. jejuni uptake, additional intracellular signaling pathways are activated from phagosomes that 51 are required for complete activation of DCs. As shown previously, our data also suggest that the Salmonella LPS-induced responses of DCs depend on internalization of LPS (34). To the best of our knowledge, this is the first report describing the requirements of C. jejuni contact and internalization for DC activation. We show that murine 0C secretion of lL-12, TNF-a, and lL-6 varies among C. jejuni strains. This result is likely due to variation in antigenic surface structures among these strains of C. jejuni (130). Similar variation in induced levels of lL-8 and CCL20 secretion was observed in IEC challenged with different clinical isolates of C. jejuni (60, 81). Moreover, separate studies in our lab showed that these C. jejuni strains exhibited varying colonization and disease phenotypes in C57BU6 lL-10"' mice (Bell et al., unpublished data). No detectable IL-10 production was observed from C. jejuni 11168-infected DCs. We suspect that lL-10 secretion by C. jejuni-infected DCs may require, along with a microbial stimulus, ligation of CD40 on DCs with CD40L that occurs during encounter with NK cells and T cells (147). We show for the first time that C. jejuni-infected DCs elicit high levels of IFN-y secretion from CD4+ T cells demonstrating a Th1 polarization. This finding correlates with the secretion of lL-12 from C. jejuni-infected DCs observed here and is further strengthened by in vivo studies demonstrating Th1-associated lgG2b antibody responses in IL-10+’+ and lL-10"' mice of C57BU6 (108), C3H and non-obese diabetic (NOD) genetic backgrounds (108, 110) and in C57BL/129 mice (48) challenged orally with C. jejuni. Consistent with our data, 52 two earlier studies documented elevated plasma levels of IFN-y in a naturally infected human patient and in C57BU6 mice intraperitoneally inoculated with C. jejuni (2, 13). IFN-y secreted by effector CD4+ T cells could activate infected macrophages to augment microbicidal activities for killing of ingested C. jejuni. Wassenaar et al. (173) and Iovine et al. (71) showed that IFN-1y treated human or murine macrophages were enhanced for killing clinical isolates of C. jejuni. Similarly, DC-induction of IFN-y from CD4+ T cells has been reported for infections with various pathogens including H. pylori and A. fumigatus (51, 57). Thus, adaptive immunity plays a role in clearing C. jejuni infection in mouse models (27). Th1 effector responses initiated by DCs may contribute to mucosal resistance to C. jejuni infection in C57BU6 mice. However, excessive innate or T-cell mediated inflammatory responses in the intestine triggered by DCs in the absence of immunoregulatory elements, like lL-10, presumably contribute to immune pathology as evident in our CS7BL/6 lL-10"' enteritis model (108). Further investigations are needed to establish these mechanisms in vivo. Currently, we are investigating the role of toll-like receptor signaling in the activation of DCs in response to C. jejuni. ACKNOWLEDGEMENTS We thank Alice Murphy for breeding mice and Dr. Julia Bell for critical manuscript review. This project was funded in part with federal funds from NIAID, NIH, Department of Health and Human Services, under Contract No. N01-Al- 53 30058 and Grant No. K26 RR023080-01. Dr. Rathinam was supported by funds from the MSU-CVM. 54 FIGURES Figure 1. (A) Internalization and killing of C. jejuni by BM-DCs. BM-DCs (2 x 105) in a 24-well plate were infected with C. jejuni at MOls of 10:1 (2.1 x 10‘5 CFU; A) and 100:1 (1.91 x 107 CFU; A). After 1 h of infection, non-phagocytosed bacteria were removed or killed by washing followed by gentamicin treatment. Subsequently BM-DCs were lysed at 2, 4, and 8 h p.i. and lysates cultured on BA plates to enumerate viable intracellular bacteria. (B) Viability of C. jejuni in R10.2 medium without BM-DCs. R10.2 medium without BM-DCs (as a control) was inoculated with 2.1 x 106 CFU or 1.91 x 107 CFU of C. jejuni and the bacterial viability was assessed at 2, 4, and 8 h post-inoculation. Data are from three wells in an experiment and are expressed as mean i SEM. One experiment representative of three independent experiments is shown. (C) Inhibition of BM- DC internalization of C. jejuni by cytochalasin 0 (CCD) and nocodazole (NCD). BM-DCs, left untreated or preincubated with 1 jig/ml CCD, 20 uM NCD, 1 jig/ml CCD and 20 0M NCD or DMSO (0.1%, vehicle control)°for 1 h, were inoculated with C. jejuni (MOI of 100:1). The internalized bacteria were determined at 2 h p.i. by using gentamicin killing assay as described above. lnternalized bacteria in each treatment group were presented as the % of that observed in DC + C. jejuni group (without inhibitors). Data are from two wells in an experiment and are expressed as mean :2 SEM. One experiment representative of two independent experiments is shown. 55 + C. jejuni1.91 x 107 CFU I + C.jejuni2.1 x 106 CFU LogtoCFU O -* N 00 A 01 O) 0 2 4 6 8 10 Time post-infection (h) + c. jejuni1.91 x 107 CFU . —A— c. jejuni2.1 x 106 CFU L0910CFU T —r T 0 2 4 6 8 10 Time post-infection (h) C DC+Q DC + DMSO + Cj DC + CCD + Cj DC + NCD + Cj DC + CCD + NCD + C] 0 50 100 150 % Relative internalization Figure 1. Internalization and killing of C. jejuni by BM-DCs 56 Figure 2. Maturation and cytokine production by BM-DCs infected with C. jejuni. Data are from three wells in an experiment and are expressed as mean i SEM. One experiment representative of two independent experiments is shown. (A) Maturation: BM-DCs (2 x 105 cells/ml) seeded in 6-well plates (B0 Falcon) were infected with C. jejuni at a MOI of 100:1, treated with LPS (0.1 ug/ml) or R10.2 medium alone. After 24 h, cells were double-stained with R-PE-conjugated anti-CD11c and FITC-conjugated anti-MHC-ll (l-Ab), anti-CD40, anti-CD80 or anti-CD86 antibodies or appropriate isotype controls and subjected to flow cytometric analysis. The mean fluorescence intensities of MHC-ll, CD40, CD80, and C086 markers on CD11c+ cells and the percentage of C011c" cells expressing these maturation markers are shown. Asterisks indicate P < 0.05 for 1) C. jejuni-infected DCs or LPS-treated DCs vs. medium alone-treated DCs. (B) Cytokine production: BM-DCs (2 x 105 cells per well of a 24-well plate) in R10 medium without antibiotics were treated with C. jejuni at MOI of 10:1 and 100:1 or LPS (0.1 ug/ml) or R10.2 medium alone. The levels of lL-12, lL-6 and TNF-a cytokines in the culture supernatants at 4, 24 and 48 h p.i. were measured by cytokine-specific sandwich ELISA. Data from LPS treatment group was included in the statistical analysis but the significance levels are not shown in figure. 57 > Mean fluorescence intensity on m Medium alone - C. jejuni :1 LPS ._L O O N-hODCD 000 O % of CD11c+ cells 0 MHC-IICD40 CD80 CD86 500 lL-12 6 5 _ 4 E. 3 C 2 1 0 50 lL-6 40 ll: lT—n E 30 E: Medium alone 20 [:31 C. jejuni10:1 10 . . - C. jejuni100:1 P ITI l:l LPS Figure 2. Maturation and cytokine production by BM-DCs infected with C. jejuni 58 Figure 3. Effect of viability of C. jejuni on activation of DCs. BM-DCs were treated with approximately equal numbers of live or formalin-killed C. jejuni (MOI of 100:1), LPS (0.1 ug/ml), or R10.2 medium alone. After 24 h, the expression of cell surface markers was analyzed by flow cytometry. (A) Mean fluorescence intensities of MHC-ll, C040, C080, and C086 markers on CD11c+ cells and the percentage of CD11c+ cells expressing these maturation markers are shown. (B) The levels of lL-12, lL-6 and TNF-ot cytokines in the culture supernatants at 4, 24 and 48 h p.i. were measured by cytokine-specific sandwich ELISA. Asterisks indicate significance P < 0.05. Data are from three wells in an experiment and are expressed as mean i SEM. One experiment representative of two independent experiments is shown. 59 Mean fluorescence intensity w pg/ml Medium alone Live C. jejuni ‘EEIU Formalin-killed C. jejuni LPS 10000 120 8000 . £100 . 5 8 80 g? F 6000 Z ‘2. a g ¢ 2 .- 60 i f / 4000 a... A D i 2 fl 2 r o 40 1 2 fit 2 f ‘- 4 2.. f“ 2000 f" 2 ° 4 y ya -. 1 .0 2° 1» 1% 1:“ MHC-IICD40 CD80 086 MHC-IICD4O 0080 D86 160 140 120 100 80 g, 60 ‘ c 40 20 ng/ml 4h 24h 48h Time post-infection Figure 3. Effect of viability of C. jejuni on activation of DCs 60 Figure 4. Requirement for contact and internalization of C. jejuni for activation of DCs. BM-DCs, preincubated with 1 pg/ml CCD and 20 uM NCD or DMSO (0.1% vehicle control) for 1 h, were treated with C. jejuni (MOI of 100:1), LPS (0.1 ug/ml) or R10.2 medium alone. In experiments with transwells, BM-DCs were added to wells of cell culture plates and 0.2 pM Anopore membrane cell culture inserts were placed in wells. Then, C. jejuni at an MOI of 100:1, R10.2 medium alone or 0.1 ug/ml of S. typhimurium LPS was added to the top Chamber. After 24 h of incubation, maturation and cytokine responses of BM-DCs were analyzed as described before. (A-D) The mean fluorescence intensities of MHC-II, C040, C080, and CD86 markers on CD11c+ cells and the percentage of CD11c+ cells expressing these maturation markers are shown. For the experiment with actin and microtubule inhibitors, MFI and percentage of CD11c cells positive for markers in the medium alone group were considered basal values and the increase in these parameters in the other two treatment groups was expressed as fold increase over basal value. (E) The levels of cytokines secreted at 24 p.i. Data are from three wells in an experiment and are expressed as mean i SEM. One experiment representative of two independent experiments is shown. 61 moo Lo co=m>=om .9 .22:me .0 Lo co=m~=mEo§ new. 85:8 .2 EoEoLSUmm .v 059”. was 6 5282 m5 wn: .6 E282 .9. .ON .om .ov com szo c 82500 D 9953:. oz I .6 Ease: was .6 E285. .o.o ... i r a. 4.6 m... .48 w s. ed a. 2.. .ed .3 .3 3 9.8 ma... 6 E282 o .F .m .m .e .m .c s OVDO md we; 6 E282 .. .o.o m m e. .3 m. .3 .3 $012 om ma... 6 E285. .o m . . .. .e . .m u .c .512 sneo +Ol lClO l0 °/o ui OSBOJOU! p|0:l CD |:Ill\l u! eseeioug piod 62 moo Co c0888 .2 .22:me .0 .0 52828525 new 89:8 .2 EwEegzcmi .v 939“. ma... 6 528.2 ma... 6 5282 we: 6 528.2 . III- C .V . r O ...\ m 8885 55s V\\\x v _ ...w m u 82 + 80 U . m .m ....._m . 8 A 88582 I . w s- M ..n. 1.. re ..L.. .8 r.— , a. .. Z. . 8 8-525 8-... 8...: mo... 6 5282 m5 6 528.2 ms: 6 528s. was 6 5285. w 0 C j, . O _ 2 2 O i T8 .8 .8 8 2 CV 2 0v ,2 0% 5 CV 28 .8 8 28 u 78 .8 u . V. 8 e u 28 88 82 . 8mo o? 88 02 - :61: SF 0 ma... 6 528.2 ma: 6 528.2 ma: 6 528.2 ma... 6 528.2 . O . . O O . O 2 .88 F .88 www : 88 88 88¢ . .8 .88 .88 .88 288 c8 .88 .88 .08. I I ... .wwmm" u u .882 u .88. . u. .88 . 88 88 O88" 88 83 .512 88. 8885 .2; U 8885 592; I zdozczcoo .v 2:9”. SII93 +01 ICIO 4° % lliisueiui eoueoseionlj ueew 63 Figure 5. Cytokine responses of BM-DCs to various strains of C. jejuni. BM- DCs were infected with the one of following strains of C. jejuni: 11168, D2600, 00835, NW, D0121 and 33560 or treated with LPS (0.1 ug/ml) or R10.2 medium alone. At 24 and 48 h p.i., the concentrations of IL-12 (A), TNF—a (B) and IL-6 (C) and in the culture supernatants were measured by sandwich ELISA. Data are from three wells in an experiment and are expressed as mean i SEM. One experiment representative of two independent experiments is shown. DCs treated with all C. jejuni strains or LPS were significantly different (P < 0.05) from medium alone-treated DCs within each time point. 64 IL-1 2 350 - 24 h 300 1:] 48 h 200 1 50 1 00 50 991ml 6%) %&\§°\\b% Q30 00656) \$\?;Q\q’:5 (566% Q9 6C6 0\ 0\ c',\ O\ 4000 TNF-a 3000 2000 log/ml 1 000 ®6M0‘§3\$Q>Q>°9 -oxso> I :8 v “3.9. cc 0.qu NP!- mn: 950 6 528.2 ‘0. N O 68 O N O (D e 2 co Vf‘ Slleo +3l lOO lo % 0.90.098 (UPI-CO.— 9.00 “2 N ma... 6 528.2 .51 a .. . .8 .8 .e m. o.o a a. .0... 8.. . . .8 88 8.8 8: 6 528.2 .1 . . .8 o... a .8... .o.. a e . m.P .08 m8 omoo 8: 6 528.2 o... .8... 3.0 .8... .8... .o.. 8.. .... i e . . .8 .8 a. 7 V .m 800 8 ma... 6 528.2 0.0 8... o.. 8.. 0.8 .7 e . . I .1. .N .8 L. .m 88 8 mm.— 6 528.2 0.0 . md . 0.. . m... 0.8 = o I:IW U! BSBBJOUI mod __-OI_2 war. 6 528.2 __-OT=2 SIIGO +0L lClO 1° °/o o u! BSBBJOUI p|O_-_| SII9° +3l L03 10 °/o SSBBJOU! mod 0 u! BSBBJOUI mod to < 92 Figure 2. C. jejuni-induced cytokine responses of BM-DCs is mainly dependent on TLR4 signaling. wr, TLR2‘" and TLR4"‘ BM-DCs were treated with C. jejuni, Salmonella serovar Typhimurium LPS (0.1 pg/ml), or medium alone. (A, B) Cytokine levels in the culture supernatant at 24 h p.i. were analyzed by ELISA. Asterisks indicate P = 0.05 for WT vs. TLR2”' or TLR4'I' BM-DCs. (C) HEK-Blue-2 cells were treated with medium-alone, C. jejuni or Pam3CSK4 (100 ng/ml). After 23 h of incubation, the level of secreted alkaline phosphatase in the culture supernatant was quantified by incubating the supernatant with QUANTI- Blue and reading the plate at 630 nm. Asterisks indicate P s 0.05 for medium vs. C. jejuni or Pam3CSK4. Data are from three wells in an experiment and are expressed as mean 2 SEM. One experiment representative of two independent experiments is shown. 93 -WT A C3 TLR4'/' 250 “-"2 IL-6 30 TNF-a 200 . 4° ‘ 2.5 I — — 30 « — 2.0 5 150 . 5 5 8100‘ l 8’20J 81:3] ,, 50 ‘ l 10 ‘ 0 5 i * ,, I . o - .* * o - * 0.0 . , Medium Cj LPS Medium Cj LPS Medium Cj LPS - wr B III TLR2'/' 300 IL-12 _ lL-6 5 TNF-a 250 ‘ 30 I * 4 . * * — 200 ‘ — * — ‘ $1504 $20 ‘ g3 0'100 « i. . 5 10 , = 2 ‘ 50‘ 1 I o . o . . o - Medium 0] LPS Medium Ci LPS Medium Cj LPS C e 1.2 O 3 "g 1.04 ,, * g .3 0.8 o o 0.6 08 2"; 0.4- c u. 02 3 0.0 g ,\ A . . Q 1‘ 66$) '08) 0% \“ C’J-\?.a((\"5 Figure 2. C. jejuni-induced cytokine responses of BM-DCs is mainly dependent on TLR4 signaling. 94 Figure 3. MyD88 and TRIF signaling dependent upregulation of surface markers and cytokine secretion by C. jejuni-infected BM-DCs. BM-DCs derived from WT, MyD884', and TRIF"' mice were treated with C. jejuni, Salmonella serovar Typhimurium LPS (0.1 pg/ml) or medium alone. After 24 h, surface expression of maturation markers (A) and secretion of cytokines (B) were analyzed by flow cytometry and ELISA, respectively. Data for BM-DCs from 057BLI6 WT mice are the same as in Figure 1C-D and 2B. Asterisks indicate P s 0.05 for WT vs. TRIF/'or My088"' BM-DCs. Data are from three wells in an experiment and are expressed as mean i SEM. One experiment representative of two independent experiments is shown. 95 800-55 68.68.58.226. .0 3 5.6688 8:26.26 6:8 8.86:8... oomtzw .0 538.2665: .coocoaoo 92.8.5.8 n=m._. 9.8 802.2 .m 859”. smqeeswq 8.... 6 528.2 «I— fld . .8.d .8..m . CONII .88 PPPOOOOO 8.: 6 528.2 8.... 6 528.2 2 w u . or u 8.m .m .8 M .8w $802.2 nU . v -\-n__m_._. a . m . om ts I 8-82. 8-... 8.... 6 528.2 8.... 6 528.2 8.... 6 528.2 . o % . ._ - . o . . 8 8.. w _. . . . O . . 8.. m .. 8 .8 . . 00 w . m m. . . . om m . v . . 8.8 8 88 888 88 8.... 6 528.2 8.... 6 528.2 8.: 6 528.2 . .. . ._ . o.o . . 0.0 .. .. . .m . _. . _. . _. 8.. r. . ooF .. ... . ooF . . .. . m... . a. . m... . o.m . o8 8.8 8.8 9.8 888 88 NT.: 8.... 6 528.2 K. com CD . od .o... .m.. ON Slleo +0l lClO l0 % U! v—Vi v “20.00.053.90. OOOONNFFOO __-O_.=2 ij U! GSBGJOU! p|0:| aseeloul mod 96 WT TRIF"' My088"' WT TLR4"‘ C. jejuni — + — + — + ._ + _ + leF-3l ‘3 .- a . .' . l r. lRF-aL —- —- ,— .._.. .....~| f; Tubulin L— ———————— ——-l "S i Figure 4. Activation of IRF-3 phosphorylation by C. jejuni-induced TLR4— TRIF signaling. WT, TLR44', MyD88"', and TRIF”' BM-DCs treated with c. jejuni or medium alone were lysed at 1 h p.i. The lysates were electrophoresed on 10% polyacrylamide gels and transferred to nitrocellulose membrane. Blots were then probed with appropriate antibodies and scanned with Ll-COR Odyssey scanner. One experiment that is representative of two independent experiments is shown. .97 A B §§ DE: 8 IFN-B mRNA IFN-[3 IFN-B a. E 50 2 act 2 '3 300 . 40 . E E 5 20 5 30‘ I \ ‘ g \ § § 200 8 83 2o . m g 100 . 1° ‘ 10 I gel 0 o - o - 5 3 cl LPS Medium ci LPS Medium ci LPS h a O x U- o — wr — 2.5 h :3 5 h -- TRIF’I’ - WT / .,. :1 TLR4” I: My088 Figure 5. IFN-B production by C. jejuni-infected BM-DCs. (A) IFN-[3 mRNA expression. Total RNA was extracted from BM-DCs treated with C. jejuni, Salmonella serovar Typhimurium LPS, or medium alone at 2.5 and 5 h post- treatment. IFN-B mRNA levels were quantified by Q-PCR. IFN-[3 mRNA levels in C. jejuni and LPS treatment groups were expressed relative to that observed in the medium-alone treated control. (B) BM-DCs derived from WT, TLR4"', MyD884', and TRIF" mice were treated with C. jejuni, Salmonella serovar Typhimurium LPS (0.1 pg/ml) or medium alone. IFN-[3 levels in the culture supernatant at 24 h p.i. were analyzed by ELISA. Asterisks indicate P s 0.05 for WT vs. TRIFJ', My088"'. or TLR44' BM-DCs. Data are from three wells in an experiment and are expressed as mean 2 SEM. 98 IFN-y 'FN’Y 1200 500 400. E3004 E 800‘ e E Ecool 200‘ 400‘ 0 0 ' : .t 06063,, c‘xo d\ c‘xo c‘xo ox 0&8; ,, 0g ,, ch , c\ Ox‘o“ «‘0 C;\ V00 002:“ “(‘2 \,0 {4“ 0 ‘1‘?) “.P £63 0“" fl: ‘0' Figure 6. TLR2, TLR4, MyDBB, and TRIF signaling are necessary for maximal Th1 polarization by BM-DCs infected with c. jejuni. WT, TLR2'", TLR44', MyD88"', and TRIF‘" BM-DCs were treated with c. jejuni or medium alone and CD4+ T cells were added to each well at 24 h p.i. IFN-y level in the culture supernatant after 72 h of coculturing was analyzed by ELISA. Asterisks indicate P s 0.05 for WT vs. TLR2'I', TLR44'. MyD88/2 or TRIF" BM-DCs infected with C. jejuni. Data are from three wells in an experiment and are expressed as mean :I: SEM. 99 CHAPTER 4 Summary and future directions 100 SUMMARY Following its recognition as a human pathogen in the 19703, Campylobacterjejuni was rapidly recognized as one of the most common bacterial causes of gastroenteritis in humans in both industrialized and developing countries (23). The Centers for Disease Control and Prevention estimates that more than 2.4 million people in the US (0.8% of the population) are affected annually with campylobacteriosis, the disease caused by C. jejuni (1). C. jejuni exists as a commensal in the intestinal tracts of food animals, particularly chickens, and is transmitted to humans through contaminated meat, milk, and water (23, 54, 94). The most common symptoms include fever, abdominal cramps, and watery to bloody diarrhea. However, patients with immunodeficiencies, including AIDS patients, develop protracted and serious diseases, such as bacteremia, peritonitis, hepatitis and meningitis following C. jejuni infection (23). Because of the molecular mimicry between certain Campylobacter cell components and host tissues (185), a small proportion of patients develop debilitating and life-threatening autoimmune complications such as Guillain-Barré syndrome (GBS) and Reiter’s syndrome (23). GBS is an acute progressive neuropathy characterized by bilateral ascending paralysis and occurs in 1 in 1000 cases of C. jejuni. The pathogenesis of C. jejuni and the host defense mechanisms controlling the infection remain poorly understood. The early events associated with pathogenesis of C. jejuni include adherence to and colonization of epithelial cells of terminal ileum, cecum, and proximal colon. A growing body of evidence 101 indicates that these processes are complex and multifactorial: several surface structures of C. jejuni including PEB1, outer membrane protein CadF, capsular polysaccharides and leA (surface lipoprotein) contribute to adherence and invasion of epithelial cells by C. jejuni (79, 80, 118, 132, 182). Interaction of C. jejuni with intestinal epithelial cells has been shown to elicit a variety of innate responses (60, 107, 113, 176). C. jejuni infection of cultured intestinal epithelial cell lines (INT407 and T84) results in secretion of IL- 8 (60, 107) as well as the antimicrobial peptides human B—defensins 2 and 3 (188). It also stimulates the secretion of several other chemokines including macrophage inflammatory proteins (MlP)-10t and MlP-30t, monocyte chemoattractant protein 1, and gamma interferon-inducible protein 10 (68, 81). Furthermore, C. jejuni infection triggers signaling through NF-tcB and MAP kinase pathways, which mediate the production of various chemokines and cytokines involved in innate immunity (29, 113, 176). While innate responses of intestinal epithelial cells to C. jejuni and the signaling pathways involved have been extensively investigated, less is known about the interplay between C. jejuni and host immune cells, particularly dendritic cells (DCs), which are necessary to elicit protective immunity. DCs are professional antigen presenting cells involved in immunosurveillance of body tissues. DCs densely populate the lamina propria of the small and large intestine and are capable of forming transepithelial dendrites by a CXgCR1-dependent mechanism to sample the intestinal lumen for commensals or pathogens (123). An extensive body of evidence demonstrates 102 that DCs play a role in educating the immune system for the development of appropriate T cell responses to pathogens (86). Conversely, pathogens have evolved many mechanisms to subvert DC functions as a means of immune evasion (36, 116). Given the facts that DCs are central to the induction of antigen-specific acquired immune responses and that certain pathogens can interfere with DC functions, it is pivotal to understand DC-C. jejuni interactions. C. r. jejuni has been shown to elicit maturation and proinflammatory responses from in vitro cultured human DCs (67). However, at this time, the role of DCs in inducing adaptive immune responses against C. jejuni is not known. ’ i; Recently, we showed that CS7BU6 wild type and congenic lL-10 deficient mice can serve as murine models of C. jejuni colonization and enteritis, respectively (108). In this model, while C. jejuni infection induced enteritis in IL- 10 deficient mice, wild type mice were resistant to C. jejuni-induced disease but stably colonized. These models will allow exploration of the basis of protective immune responses that result in either healing or disease mediated by immunopathology after a primary C. jejuni infection, which otherwise would be difficult to investigate in humans. To be able to focus on the role of specific immune components in controlling C. jejuni infection, we first need to understand the overall immune responses elicited by C. jejuni in these models. In this context, a study describing the role of DCs in inducing the innate and adaptive immune responses to C. jejuni in mouse models is warranted. The objective of these studies was to gain insight into the interaction of murine DC with C. jejuni and the effect of this interaction on development of Th cell responses in vitro. We 103 hypothesized that murine bone marrow-derived DCs (BM-DCs) internalize C. jejuni, which then undergo activation and initiate a Th1-type immune response. As a primary goal, we wanted to determine whether C. jejuni 11168 is efficiently phagocytosed and killed by murine BM-DCs and whether it is capable of surviving intracellularly. It remains controversial in the literature whether C. jejuni escapes killing by phagocytic cells and translocates from the intestinal tracts to systemic organs inside these cells. In this study, the intracellular survival of the bacteria was assessed at 2, 4 and 8 h post-infection using a standard gentamicin killing assay. We show that C. jejuni 11168 is efficiently internalized and killed by murine DCs within 8 h post-infection. These findings are consistent with those of Hu et al., (2006) (67) who reported that ~ 99% of phagocytosed C. jejuni 81-176 were killed by human 003 within 24 h post-infection. Similar results were obtained with activated human macrophages. In contrast, intracellular pathogens such as S. typhimurium and Mycobacterium tuberculosis resist killing by DOS and survive intracellularly for an extended period of time; this survival ability was implicated in the systemic spread of these bacteria (20, 28). Under steady state conditions, 005 are present in the immature state but rapidly undergo functional transition to the mature state following exposure to microbial products (86, 116). Mature DCs are characterized by enhanced surface-display of MHC-ll-antigen complexes and costimulatory molecules, which provide activating signals 1 and 2, respectively, to naive T cells (86). Accumulating evidence has demonstrated that DCs play a major role in eliciting anti-microbial T-cell immunity vs. tolerance towards self-antigens and 104 commensals; this role is suggested to depend mechanistically in part on their maturation status after antigenic exposure (106). In the current study, flow cytometric analysis revealed that immature murine DCs infected with C. jejuni 11168 undergo maturation by up-regulating the surface expression of MHC-ll, CD40, 0080, and CD86 molecules. Similarly, an earlier study showed that human DCs become mature following C. jejuni infection as demonstrated by the increased expression of CD40, CD80, and CD86 (67). The increased DC expression of MHC-ll and co-stimulatory molecules after challenge suggests that C. jejuni-infected DCs would provide essential signals 1 and 2 to stimulate efficient anti-C. jejuni T cell responses. In order to stimulate appropriate CD4” T cell responses, antigen presenting cells must provide instructive cytokine signals to na'l've Th cells (106). Several lines of evidence indicate that cytokine signals from DCs have a major influence on the differentiation of naive Th cells to effector cells (86, 116). DC- and macrophage-derived lL-12 is crucial for the optimal generation of Th1-type immune responses to a variety of pathogens (86, 116). In this study we observed a C. jejuni 11168 dose- and time-dependent secretion of lL-12 by infected DCs. lL-12 has been shown to mediate the cross-talk between DOS and innate natural killer (NK) cells, inducing lFva production by NK cells (57, 92, 93, 169). This DC- mediated NK cell activation occurs early and locally at the site of infection, mediating the initial resistance to bacterial infections (92, 169). Hu et al., (2006) also showed that human monocyte-derived DCs secreted lL-12 after challenge with C. jejuni 81-176 (67). With such elicitation of lL-12, it is likely that C. jejuni 105 also stimulates NK cell activation in' the host, although this event has not been studied. TNF-a and lL-6 are multifunctional proinflammatory mediators induced in many microbial infections (95). In this study, murine DCs secreted TNF-a and IL- 6 when infected with C. jejuni 11168. TNF-a secreted by C. jejuni-infected DCs may act in an autocrine or paracrine manner to enhance DC—maturation and migration (163). Additionally, TNF-ot may act together with IFN-y in activating local anti-microbial mechanisms in macrophages (101). Likewise, human DCs were also shown to elicit robust proinflammatory responses by secreting TNF-a, IL-1B, lL-6 and lL-8 following C. jejuni 81-176 infection (67). Similarly, a number of in vitro studies demonstrated that C. jejuni induces proinflammatory cytokine and chemokine production from a variety of cells such as intestinal epithelial cells, macrophages, or monocytic cell lines (60, 68, 84, 107, 177). IL-10 is an immunoregulatory cytokine critical in preventing exaggerated immune and inflammatory responses against normal enteric flora and pathogens (134). In our mouse model, lL-10"' mice manifested moderate to severe colitis from 2—35 days following primary C. jejuni 11168 challenge, whereas wild type C57BU6 mice were resistant to disease despite uniform stable colonization (108). Therefore, it is evident that IL-10 plays an important immunoregulatory role in C. jejuni infection in mice that protects against tissue damage due to unchecked inflammatory responses. In the current study, no detectable amounts of lL-10 production were observed from C. jejuni-infected DCs at all time points examined. Our in vitro experiments suggest that DCs are unlikely to be the primary source of lL-10 in mice infected with C. jejuni. A recent report by Perona- 106 Wright et al., (2006) also demonstrated that DCs may not necessarily be the most important source of IL-10 required for immunoregulation in vivo. These authors suggested that non-hematopoietic and innate cells such as epithelial cells may be significant contributors of lL-10 (134). Because immunity to C. jejuni has been suggested to be strain-specific (54) and cytokines are integral components of cell-mediated immune resistance, we next investigated whether different strains of C. jejuni, which were isolated from various sources, elicit differential cytokine production from murine DCs. C. jejuni strains 11168 and 33560 induced higher levels of lL-12, TNF-a and IL-6, whereas other four strains elicited relatively lower amounts of these cytokines. In particular, strain NW was the least potent inducer of these cytokines. These findings indicate that DCs can be activated by various strains of C. jejuni to secrete IL-12 and proinflammatory cytokines in a strain-specific manner and support the conclusion that strain-to-strain variation influences host-pathogen interactions in C. jejuni infection and thus the disease development (15). DCs have the ability to express different cytokines and membrane-bound molecules and play a decisive role in the differentiation of naive T cells to effector populations (86). The Th-polarizing ability of C. jejuni 11168-infected BM-DCs was evaluated in an in vitro DC-CD4" T cell coculture system; BM-DCs infected with C. jejuni 11168 induced Th1 but not Th2 priming of CD4‘ T cells in vitro. Having demonstrated that 005 recognize C. jejuni, undergo activation, and induce Th1-effector differentiation of naive CD4“ T cells, we sought to understand the mechanisms, both bacterial and host, responsible for these 107 responses. We investigated the roles of viability of C. jejuni, bacterial contact with DCs, and bacterial internalization by DCs in inducing activation of DC maturation and cytokine production. Formalin-killed C. jejuni also elicited maturation and cytokine responses from DCs but at a significantly lower magnitude compared to live bacteria. Inhibition of C. jejuni phagocytosis by DCs resulted in significant impairment of expression of maturation markers, IL-12, and TNF-a but not lL-6. This result suggests that recognition of C. jejuni by surface receptors on DCs is sufficient to induce lL-6 secretion by DCs. Furthermore, when direct contact between C. jejuni and DOS was prevented using transwell inserts, the maturation and cytokine responses of DCs to C. jejuni were impaired. Overall, all the bacterial factors analyzed in this study, such as C. jejuni viability, contact of C. jejuni cells with D03, and internalization of C. jejuni by DCs, are necessary for maximal activation of DCs in response to C. jejuni. Next, the role of TLR signaling in mediating DC responses to C. jejuni was investigated. TLRs are germ-line encoded pattern recognition receptors that recognize various conserved molecules of pathogens, including LPS, flagellin, and nucleic acids (4). Antigen-presenting cells, particularly DCs, express a wide array of TLRs on their surfaces as well as intracellularly. Following recognition of their ligands, TLRs induce innate inflammatory responses that regulate the subsequent development of adaptive immune responses (74). TLR2 (in association with TLR1 or TLR6), TLR4, TLR5, and TLR9 are involved in sensing of bacterial pathogens. In this study, .I focused on the role of TLR2 and TLR4 recognition of C. jejuni for two main reasons: 1) C. jejuni possesses potential 108 ligands for these receptors, namely LOS and lipoproteins (80, 120); 2) it has been reported that TLR5 and TLR9 recognition of C. jejuni is weak because of the nature of their agonists, flagellin and DNA respectively, in C. jejuni (6, 37). The surface expression of MHC-ll, 0080, and CD86 molecules was substantially reduced in TLR2- or TLR4-deficient DCs compared to WT 003 following infection with C. jejuni. The production of lL-6, TNF-ot and IL-12 was profoundly impaired in infected TLR4-deficient 003 whereas TLR2 deficiency resulted in only minimal reduction in lL-6 and TNF-ot secretion and about 40-50% inhibition in the lL-12 response to C. jejuni. Overall, it appears that signaling through both of these receptors is necessary for optimal maturation of DCs following C. jejuni infection, whereas cytokine responses of 005 to C. jejuni are mainly dependent on TLR4. Such a major role of TLR4 in C. jejuni-induced activation of DCs is consistent with the structure of lipid A of C. jejuni: lipid A of C. jejuni is hexa-acylated and diphosphorylated and thus possesses the essential configuration to stimulate TLR4 signaling (45, 120). In most cases, recognition of LPS by TLR4 involves two additional host factors such as CD14 and MD-2, which act as the co-receptors (4). It would be of interest to determine whether TLR4 recognition of C. jejuni LOS is also dependent on CD14 and MD—2. Surprisingly, both MyD88-dependent and -independent (TRIF) pathways downstream of TLR4 were activated by C. jejuni, and signaling through both of these pathways was found be required for maximal maturation and cytokine responses to C. jejuni. Besides stimulating distinct transcription factors, both MyD88 and TRIF have also been shown to activate the same set of transcription 109 factors, including NF-KB, that mediate the transcription of genes involved in innate responses. As a result, TRIF and MyD88 pathways induce a variety of overlapping cellular responses (4). However, TRIF also activates a unique pathway leading to activation of transcription factor lRF-3 that induces expression of type I interferon genes (181). Indeed, C. jejuni signaling through TLR4-TRIF was found to trigger phosphorylation of downstream lRF-3 and secretion of IFN-B. Stimulation of TLR4 by E. coli LPS triggers IFN-[3 production by D05, and the secreted IFN-l3 augments activation of DCs through lRF-7. This finding suggests that IFN-[3 secreted by C. jejuni-infected DCs may amplify their maturation and IL-12 responses (53). Impairment of C. jejuni-induced maturation and cytokine profiles in the absence of TLR2, TLR4, My088, and TRIF was reflected in the Th1 cell activating ability of C. jejuni-infected DCs. C. jejuni-infected DCs deficient in these components elicited significantly reduced amounts of IFN-y from naive CD4+ T cells, indicating that TLR-mediated delivery of costimulatory and cytokine signals are essential for maximal Th1 polarization. Consistent with these findings, Watson et al., (2007) (177) reported that mice deficient in MyD88 following oral inoculation with C. jejuni were compromised in their ability to eliminate the bacteria. MyD88”' mice harbored higher numbers of C. jejuni in intestinal tissues than WT mice, and the extraintestinal spread of C. jejuni into systemic sites such as spleen and liver was observed mainly in MyD88"' mice. Based on the results of my studies, we propose the following model (Fig. 1) for the role of TLR signaling in C. jejuni-induced activation of DCs: TLR4 110 recognizes C. jejuni, most likely the LOS of C. jejuni, and activates both MyD88 and TRIF signaling that cooperate in inducing DC maturation and cytokine responses. It is possible that these two signaling pathways activated by C. jejuni converge at the level of transcription factors such as NFKB resulting in sustained activation and nuclear translocation. Furthermore, activation of the TLR4-TRIF pathway induces lRF-3 activation and IFN-B production in response to C. jejuni. Besides stimulating TLR4 signaling, C. jejuni, possibly through its Iipoprotein leA, activates TLR2 signaling, which further augments some of the responses of DCs via My088. In conclusion, this study provides important mechanistic insights into the interaction between C. jejuni and DCs, which are the players in tailoring pathogen-specific adaptive immune responses. Identification of TLR3 responsible for C. jejuni recognition and DC activation in this study would advance current understanding of host defense mechanisms that confer protective immunity to infection with C. jejuni and form a basis for future investigations to dissect immunoregulation during C. jejuni infection. 111 C. jejuni Figure 1. Proposed model for the role of TLR signaling in C. jejuni-induced activation of DCs. 112 FUTURE DIRECTIONS Does C. jejuni activate DCs in vivo? An important question that arises from this study is whether C. jejuni induces the activation of DCs present in the intestinal lamina propria. The in vitro findings of this study combined with our laboratory’s previous observation—that C. jejuni-specific immunohistochemical staining was found associated with DC- Iike cells in the intestinal mucosa (108)-—suggest the hypothesis that DCs in the intestinal mucosa recognize. C. jejuni and undergo activation. The use of confocal microscopy and immunohistochemistry will allow us to investigate this in vivo phenomenon. Localization of C. jejuni in the 005 can be assessed by performing dual immunostaining for CD11c (dendritic cell marker) and C. jejuni antigens in the frozen sections of ileocecocolic junctions and colon tissues from mice orally infected with C. jejuni. Furthermore, the phenotypic maturation of DCs in tissues from C. jejuni-infected mice can be assessed by immunohistochemistry using antibodies against CDBO, CD86 and CD40. An alternate approach is to use multicolor flow cytometry: single cell suspensions obtained from fresh tissues of control and infected mice can be stained with fluorochrome—conjugated anti- CD11c and anti-C080, anti-CD86, or anti-CD40 antibodies and analyzed by flow cytometry. However, this method is cumbersome and has practical difficulties since the numbers of DCs that can be harvested from intestinal tissue may be too low to obtain reliable results. 113 Distinct subsets of DCs exist in the intestinal lamina propria, which include CD11c”CD11b"CD8a', CD11c+CD1 1 b'CD8a‘, and CD11c"CD11b'CDBa' 00$ (73). Additionally, a population of DCs characterized by the expression of a chemokine receptor, CX3CR1, has been identified in the lamina propria (123). These CX3CR1+ DCs directly sample the intestinal lumen for microbes by forming transepithelial dendrites. It is important to understand which subset(s) of lamina propria DCs participates in C. jejuni recognition in vivo. This also can be analyzed by using a combination of antibodies specific for each subset of DCs in immunohistochemistry and flow cytometry. 114 Does NOD1 recognition of C. jejuni contribute to TLR-independent induction of Th1 responses by DCs? A surprising finding from this study is that priming of Th1 cells by C. jejuni- infected DCs was preserved to a certain extent in the absence of TLR signaling. This indicates that C. jejuni stimulates TLR-independent pathway(s) in DOS, which then contribute to the induction of Th1 cells by DCs. Signaling through cytoplasmic nucleotide-binding oligomerization domain (NOD)—1 receptor may play a role in this process. NOD1 belongs to a family of cytoplasmic pattern recognition receptors and is mainly expressed in antigen-presenting cells such as DCs and epithelial cells (156). NOD1 recognizes y—D-glutamyl-meso- diaminopimelic acid (iE-DAP) derived from peptidoglycan (PGN) of Gram- negative bacteria. Sensing of iE-DAP by NOD-1 results in induction of inflammatory cytokine genes via activation of MAP kinases and NF-KB (156). Zilbauer et al. (2007) recently reported that NOD1 recognition of C. jejuni is required for maximal expression of lL-8 and human B-defensin, an antimicrobial peptide, by Caco-2 intestinal epithelial cells. Furthermore, the knock—down of NOD1 expression in IEC resulted in increased numbers of intracellular C. jejuni (189). On the basis of these published reports, I hypothesize that cytosolic NOD1 receptor signaling triggered by C. jejuni in 003 contributes to Th1 polarization by C. jejuni-infected DCs. Even though C. jejuni has been shown not to escape the phagolysosome into the cytoplasm in phagocytic cells (175) such as macrophages and dendritic cells, it is possible that derivative products of PGN 115 from C. jejuni being degraded in the phagolysosome can leak into cytoplasm and stimulate NOD1 signaling as reported previously for Listeria monocytogenes (59). The experimental approach to determine whether NOD1 can mediate Th1- induction by C. jejuni-infected DCs in the absence of TLR signaling would be to coculture C57BU6 wild type DCs, MyD88"' TRIF"' DCs, and MyD88"' TRIF"' NOD1"' DCs infected with C. jejuni with naive CD4+ T cells and analyze the IFN-y levels in the supernatants by the methods described in the previous chapters. 116 Identification of TLR ligands in C. jejuni Having demonstrated that C. jejuni activates signaling through TLR2 and TLR4, the next step is to identify the ligands in C. jejuni that trigger TLR2 and TLR4 pathways. Furthermore, identification of C. jejuni components that induce potent innate immune responses through TLR signaling would aid in the development of vaccines, particularly subunit vaccines, against C. jejuni. As described in chapter 3, LOS of C. jejuni is the most likely candidate to be recognized by TLR4. Supporting evidence for this speculation comes from the structure of lipid A of C. jejuni LOS. Lipid A is the primary immunostimulatory component of LPS/LOS, and the number and length of acyl chains in lipid A are critical for this function (5). C. jejuni LOS, like Escherichia coli LPS, possesses diphosphorylated hexa-acyl lipid A that was reported to be essential for optimal stimulation of cellular immune responses (120). Indeed, it has been shown that C. jejuni LOS exhibits comparable endotoxic activity in biological test systems relative to that observed with prototype enterobacterial LPS preparations (120). The putative ligands for TLR2 signaling by C. jejuni are lipoproteins. Literature shows that C. jejuni genome encodes for several lipoproteins, namely CjaA, HisJ, Omp18, and leA of which two (JIpA; CjaA) have been characterized (79, 80). However, to date none of these molecules has been demonstrated to be recognized by TLR2. Here, the hypothesis to be tested is that LOS and lipoproteins, particularly leA, of C. jejuni induce phenotypic and functional activation of DCs. The experimental approach is to isolate the LOS from C. jejuni using the Darveau-Hancock method (39) or by using a commercial LPS 117 extraction kit (lntron Biotechnology, Korea). Genes encoding lipoproteins would be cloned into and expressed in E. coli as His-tagged proteins and purified using nickel columns. DCs would be stimulated with various doses of purified LOS and lipoproteins and be assessed by flow cytometry and ELISA for maturation and cytokine production, respectively. A caveat in using purified components from bacteria is that they are often contaminated with other undesirable components that invalidate the results of experiments designed to examine the role of specific bacterial mediators (4, 64). It has been well documented that LPS preparations are often contaminated with lipoprotein and vice versa, which would confound the results obtained with such preparations. However, this problem can be overcome to certain extent by using both WT and TLR4"' DCs in experiments to test LOS preparations and WT and TLR2"’ DCs in experiments to test Iipoprotein preparations. 118 APPENDIX Besides my research projects, I have contributed to the following publications from our laboratory: Bell, J. A., J. L. St. Charles, A. J. Murphy, V. A. Rathinam, A. E. Plovanich- Jones, E. L. Stanley, J. E. Wolf, J. R. Gettings, T. S. Whittam, and L. S. Mansfield. 2009. Multiple factors interact to produce responses resembling spectrum of human disease in Campylobacterjejuni infected CS7BL/6 IL10"' mice. BMC Microbiol 9(1):57. Mansfield, L. S., J. S. Patterson, B. R. Fierro, A. J. Murphy, V. A. Rathinam, J. J. Kopper, N. l. Barbu, T. J. Onifade, and J. A. Bell. 2008. Genetic background of lL-10-l- mice alters host-pathogen interactions with Campylobacterjejuni and influences disease phenotype. Microb Pathog 45:241-257. Mansfield, L. S., J. A. Bell, D. L. Wilson, A. J. Murphy, H. M. Elsheikha, V. A. Rathinam, B. R. Fierro, J. E. Linz, and V. B. Young. 2007. C57BU6 and congenic interleukin-10-deficient mice can serve as models of Campylobacter jejuni colonization and enteritis. Infect lmmun 75:1099-1115. My contributions include oral inoculation of mice with C. jejuni, clinical assessment of infected mice, blood collection, necropsy, and tissue harvesting. 119 BIBLIOGRAPHY http://www.cdc.gov/nczved/dfbmd/disease_listing/campylobacter_gi.html. Abram, M., D. Vu kovic, B. Wraber, and M. Doric. 2000. Plasma cytokine response in mice with bacterial infection. Mediators lnflamm 9:229-234. ' Akhtar, S. Q., and F. Huq. 1989. Effect of Campylobacterjejuni extracts and culture supernatants on cell culture. J Trop Med Hyg 92:80-85. Akira, S., S. Uematsu, and O. Takeuchi. 2006. Pathogen recognition and innate immunity. Cell 124:783-801. Alexander, C., and E. T. Riefschel. 2001. Invited review: Bacterial lipopolysaccharides and innate immunity. Journal of Endotoxin Research 7:167-202. Andersen-Nissen, E., K. D. Smith, K. L. Strobe, S. L. Barrett, B. T. Cookson, S. M. Logan, and A. Aderem. 2005. Evasion of Toll-like receptor 5 by flagellated bacteria. Proc Natl Acad Sci U S A 102:9247- 9252. ' Appledorn, D. M., S. Patial, A. McBride, S. Godbehere, N. Van Rooijen, N. Parameswaran, and A. Amalfitano. 2008. Adenovirus vector-induced innate inflammatory mediators, MAPK signaling, as well as adaptive immune responses are dependent upon both TLR2 and TLR9 in vivo. J Immunol 181:2134-2144. Ashtekar, A. R., P. Zhang, J. Katz, C. C. S. Deivanayagam, P. Rallabhandi, S. N. Vogel, and S. M. Michalek. 2008. TLR4-mediated activation of dendritic cells by the heat shock protein DnaK from Francisella tularensis. J Leukoc Biol 84:1434-1446. Atarashl, K., J. Nishimura, T. Shima, Y. Umesaki, M. Yamamoto, M. Onoue, H. Yagita, N. lshii, R. Evans, K. Honda, and K. Takeda. 2008. ATP drives lamina propria TH17 cell differentiation. Nature 455:808-812. 120 10. 11. 12. 13. 14. 15. 16. 17. 18. Au, W. C., P. A. Moore, W. Lowther, Y. T. Juang, and P. M. Pitha. 1995. Identification of a member of the interferon regulatory factor family that binds to the interferon-stimulated response element and activates expression of interferon-induced genes. Proc Natl Acad Sci U S A 92:11657-11661. Bacon, D. J., R. A. Aim, D. H. Burr, L. Hu, D. J. Kopecko, C. P. Ewing, T. J. Trust, and P. Guerry. 2000. Involvement of a plasmid in virulence of Camp ylobacter jejuni 81 -1 76. Infect lmmun 68:4384-4390. Bacon, D. J., C. M. Szymanski, D. H. Burr, R. P. Silver, R. A. Aim, and P. Guerry. 2001. A phase-variable capsule is involved in virulence of Campylobacterjejuni 81 -1 76. Mol Microbiol 40:769-777. Baqar, S., B. Rice, L. Lee, A. L. Bourgeois, A. N. El Din, D. R. Tribble, G. P. Heresi, A. S. Mourad, and J. R. Murphy. 2001. Campylobacter jejuni enteritis. Clin Infect Dis 33:901-905. Bar, W. 1988. Role of murine macrophages and complement in experimental Campylobacter infection. J Med Microbiol 26:55-59. Bell, J., J. St Charles, A. Murphy, V. Rathinam, A. Plovanich-Jones, E. Stanley, J. Wolf, J. Gettings, T. Whittam, and L. Mansfield. 2009. Multiple factors interact to produce responses resembling spectrum of human disease in Campylobacterjejuni infected CS7BL/6 lL-10"' mice. BMC Microbiol 9:57. Biswas, D., K. ltoh, and C. Sasakawa. 2003. Role of microfilaments and microtubules in the invasion of INT-407 cells by Campylobacterjejuni. Microbiol Immunol 47:469-473. Black, R. E., M. M. Levine, M. L. Clements, T. P. Hughes, and M. J. Blaser. 1988. Experimental Campylobacterjejuni infection in humans. J Infect Dis 157:472-479. Blaser, M. J., and J. Engberg. 2008. Clinical aspects of Campylobacter jejuni and Campylobacter coli infections, p. 99-121. In I. Nachamkin, C. M. Szymanski, and M. J. Blaser (ed.), Campylobacter, 3rd ed. ASM Press, Washington, DC. 121 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. Blaser, M. J., R. 5. Black, D. J. Duncan, and J. Amer. 1985. Campylobacterjejuni-specific serum antibodies are elevated in healthy Bangladeshi children. J Clin Microbiol 21:164-167. Bodnar, K. A., N. V. Serbina, and J. L. Flynn. 2001. Fate of Mycobacterium tuberculosis within murine dendritic cells. Infect lmmun 69:800-809. Brandt, K., S. Bulfone-Paus, D. C. Foster, and R. Ruckert. 2003. Interleukin-21 inhibits dendritic cell activation and maturation. Blood 102:4090-4098. Bras, A. M., S. Chatterjee, B. W. Wren, D. G. Newell, and J. M. Ketley. 1999. A novel Campylobacterjejuni two-component regulatory system important for temperature-dependent growth and colonization. J Bacteriol 1 81 :3298-3302. ' Butzler, J. P. 2004. Campylobacter, from obscurity to celebrity. Clin Microbiol Infect 10:868-876. Buzby, J. C., B. M. Alice, and T. Roberts. 1997. The economic burden of Campylobacter-associated Guillain-Barré syndrome. J Infect Dis 176 Suppl 2:8192-197. Calva, J. J., G. M. Ruiz-Palacios, A. B. Lopez-Vidal, A. Ramos, and R. Bojalil. 1988. Cohort study of intestinal infection with Campylobacter in Mexican children. Lancet 1 :503-506. Centers-for-Disease-Control-and-Prevention. 2008. Preliminary FoodNet data on the incidence of infection with pathogens transmitted commonly through food—10 states, 2007. MMWR Morb Mortal Wkly Rep 57:366-370. Chang, C., and J. F. Miller. 2006. Campylobacterjejuni colonization of mice with limited enteric flora. Infect lmmun 74:5261-5271. Cheminay, C., M. Schoen, M. Hensel, A. Wandersee-Steinhauser, U. Ritter, H. Korner, M. Rollinghoff, and J. Hein. 2002. Migration of Salmonella typhimurium-harboring bone marrow-derived dendritic cells towards the chemokines CCL19 and CCL21. Microb Pathog 32:207-218. 122 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. Chen, M. L., Z. Ge, J. G. Fox, and D. B. Schauer. 2006. Disruption of tight junctions and induction of proinflammatory cytokine responses in colonic epithelial cells by Campylobacterjejuni. Infect lmmun 74:6581- 6589. Coccia, E. M. 2008. IFN regulation and functions in myeloid dendritic cells. Cytokine Growth Factor Rev 19:21-32. Colino, J., and C. M. Snapper. 2003. Two distinct mechanisms for induction of dendritic cell apoptosis in response to intact Streptococcus pneumoniae. J Immunol 171 :2354-2365. Colonna, M. 2004. Alerting dendritic cells to pathogens: The importance of Toll-like receptor signaling of stromal cells. Proc Natl Acad Sci U S A 101:16083-16084. Coombes, J. L., and F. Powrie. 2008. Dendritic cells in intestinal immune regulation. Nat Rev Immunol 8:435-446. Cowan, D. B., S. Noria, C. Stamm, L. M. Garcia, D. N. Poutias, P. J. del Nido, and F. X. McGowan, Jr. 2001. Lipopolysaccharide internalization activates endotoxin-dependent signal transduction in Cardiomyocytes. Circ Res 88:491-498. Crushell, E., S. Harty, F. Sharif, and B. Bourke. 2004. Enteric campylobacter: purging its secrets? Pediatr Res 55:3-12. Cutler, C. W., R. Jotwani, and B. Pulendran. 2001. Dendritic cells: immune saviors or Achilles' heel? Infect Immun 69:4703-4708. Dalpke, A., J. Frank, M. Peter, and K. Heeg. 2006. Activation of Toll-like receptor 9 by DNA from different bacterial species. Infect. Immun. 74:940- 946. Dalpke, A., J. Frank, M. Peter, and K. Heeg. 2006. Activation of Toll-like receptor 9 by DNA from different bacterial species. Infect lmmun 74:940- 946. 123 39. 40. 41. 42. 43. 45. 46. 47. 48. Darveau, R. P., and R. E. Hancock. 1983. Procedure for isolation of bacterial lipopolysaccharides from both smooth and rough Pseudomonas aeruginosa and Salmonella typhimurium strains. J Bacteriol 155:831-838. Debruyne, L., D. Gevers, and P. Vandamme. 2008. Taxonomy of the family Campylobacteraceae, p. 3-25. In I. Nachamkin, C. M. Szymanski, and M. J. Blaser (ed.), Campylobacter, 3rd ed. ASM Press, Washington, DC Decker, T., M. Muller, and S. Stockinger. 2005. The Yin and Yang of type I interferon activity in bacterial infection. Nat Rev Immunol 5:675-687. Dekeyser, P., M. Gossuin-Detrain, J. P. Butzler, and J. Sternon. 1972. Acute enteritis due to related vibrio: first positive stool cultures. J Infect Dis 125:390-392. Doyle, S. E., S. A. Vaidya, R. O'Connell, H. Dadgostar, P. W. Dempsey, T.-T. Wu, G. Rao, R. Sun, M. E. Haberland, R. L. Modlln, and G. Cheng. 2002. IRF3 mediates a TLR3/TLR4—specific antiviral gene program. Immunity 17:251-263. DuPont, H. L. 2006. Travellers' diarrhoea: contemporary approaches to therapy and prevention. Drugs 66:303-314. Erridge, C., E. Bennett-Guerrero, and I. R. Poxton. 2002. Structure and function of lipopolysaccharides. Microbes Infect 4:837-851. Escherich, T. 1886. Beitrage zur Kenntniss der Darmbacterien. lll. Ueber das Vorkommen von Vibrionen im Darrncanal und den Stuhlgangen der Sauglinge. (Articles adding to the knowledge of intestinal bacteria. Ill. On the existence of vibrios in the intestines and feces of babies). Miinchener Med Wochenschrift 33:815—817. Florin, l., and F. Antillon. 1992. Production of enterotoxin and cytotoxin in Campylobacterjejuni strains isolated in Costa Rica. J Med Microbiol 37:22-29. Fox, J. G., A. B. Rogers, M. T. Whary, Z. Ge, N. S. Taylor, S. Xu, B. H. Horwitz, and S. E. Erdman. 2004. Gastroenteritis in NF-kB-deficient mice is produced with wild-type Camp/yobacterjejuni but not with C. jejuni 124 49. 50. 51. 52. 53. 54. 55. 56. lacking cytolethal distending toxin despite persistent colonization with both strains. Infect lmmun 72:1116-1125. Freudenberg, M. A., T. Merlin, C. Kalis, Y. Chvatchko, H. Stubig, and C. Galanos. 2002. Cutting Edge: A murine, IL-12-independent pathway of IFN-g induction by Gram-negative bacteria based on STAT4 activation by type I IFN and lL-18 signaling. J Immunol 169:1665-1668. Friis, L. M., M. Keelan, and D. E. Taylor. 2009. Campylobacterjejuni drives MyD88-independent interleukin-6 secretion via Toll-like receptor 2. Infect lmmun 77:1553-1560. Gafa, V., R. Lande, M. C. Gagliardi, M. Severa, E. Giacomini, M. E. Remoli, R. Nisini, C. Ramoni, P. Di Francesco, D. Aldebert, R. Grillot, and E. M. Coccia. 2006. Human dendritic cells following Aspergillus fumigatus infection express the CCR7 receptor and a differential pattern of interleukin-12 (IL-12), lL-23, and IL-27 cytokines, which lead to a Th1 response. Infect lmmun 74:1480-1489. Gafa, V., M. E. Remoli, E. Giacomini, M. C. Gagliardi, R. Lande, M. Severa, R. Grillot, and E. M. Coccia. 2007. In vitro infection of human dendritic cells by Aspergillus fumigatus conidia triggers the secretion of chemokines for neutrophil and Th1 lymphocyte recruitment. Microbes Infect 9:971 -980. Gautier, G., M. Humbert, F. Deauvieau, M. Scuiller, J. Hiscott, E. E. Bates, G. Trinchieri, C. Caux, and P. Garrone. 2005. A type I interferon autocrine-paracrine loop is involved in Toll-like receptor-induced interleukin-12p70 secretion by dendritic cells. J Exp Med 201:1435-1446. Girard, M. P., D. Steele, C. L. Chaignat, and M. P. Kieny. 2006. A review of vaccine research and development: human enteric infections. Vaccine 24:2732-2750. Grant, C. C., M. E. Konkel, W. Cieplak, Jr., and L. S. Tompkins. 1993. Role of flagella in adherence, internalization, and translocation of Campylobacterjejuni in nonpolarized and polarized epithelial cell cultures. Infect lmmun 61:1764-1771. Hacker, H., R. M. Vabulas, O. Takeuchi. K. Hoshino, S. Akira, and H. Wagner. 2000. Immune cell activation by bacterial CpG-DNA through 125 57. 58. 59. 60. 61. 62. 63. 64. myeloid differentiation marker 88 and tumor necrosis factor receptor- associated factor (TRAF)6. J Exp Med 192:595-600. Hafsi, N., P. Voland, S. Schwendy, R. Rad, W. Reindl, M. Gerhard, and C. Prinz. 2004. Human dendritic cells respond to Helicobacter pylori, promoting NK cell and Th1-effector responses in vitro. J Immunol 173:1249-1257. Hellman, J., M. M. Tehan, and H. A. S. Warren. 2003. Murein Iipoprotein, peptidoglycan-associated Iipoprotein, and outer membrane protein A are present in purified rough and smooth lipopolysaccharides. J Infect Dis 188:286-289. Herskovits, A. A., V. Auerbuch, and D. A. Portnoy. 2007. Bacterial ligands ggenerated in a phagosome are targets of the cytosolic innate immune system. PLoS Pathog 3:e51. Hickey, T. E., S. Baqar, A. L. Bourgeois, C. P. Ewing, and P. Guerry. 1999. Campylobacterjejuni-stimuIated secretion of interleukin-8 by INT407 cells. Infect Immun 67:88-93. Hickey, T. E., G. Majam, and P. Guerry. 2005. Intracellular survival of Campylobacterjejuni in human monocytic cells and induction of apoptotic death by cytholethal distending toxin. Infect lmmun 73:5194—5197. Hickey, T. E., A. L. McVeigh, D. A. Scott, R. E. Michielutti, A. Bixby, S. A. Carroll, A. L. Bourgeois, and P. Guerry. 2000. Campylobacterjejuni cytolethal distending toxin mediates release of interleukin-8 from intestinal epithelial cells. Infect Immun 68:6535-6541. Higgins, S. C., E. C. Lavelle, C. McCann, B. Keogh, E. McNeela, P. Byrne, B. O'Gorman, A. Jarnicki, P. McGuirk, and K. H. Mills. 2003. Toll-like receptor 4-mediated innate lL-10 activates antigen-specific regulatory T cells and confers resistance to Bordetella pertussis by inhibiting inflammatory pathology. J Immunol 171 :31 19-3127. Hirata, N., Y. Yanagawa, T. Ebihara, T. Seya, S. Uematsu, S. Aklra, F. Hayashi, K. lwabuchi, and K. Onoé. 2008. Selective synergy in anti- inflammatory cytokine production upon cooperated signaling via TLR4 and TLR2 in murine conventional dendritic cells. Mol Immunol 45:2734. 126 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. Hoebe, K., X. Du, P. Georgel, E. Janssen, K. Tabeta, S. 0. Kim, J. Goode, P. Lin, N. Mann, S. Mudd, K. Crozat, S. Sovath, J. Han, and B. Beutler. 2003. Identification of Lps2 as a key transducer of MyD88- independent TIR signalling. Nature 424:743-748. Hoshino, K., T. Kaisho, T. Iwabe, O. Takeuchi, and S. Akira. 2002. Differential involvement of IFN-b in Toll-like receptor-stimulated dendritic cell activation. Int Immunol 14:1225-1231. Hu, L., M. D. Bray, M. Osorlo, and D. J. Kopecko. 2006. Campylobacter jejuni induces maturation and cytokine production in human dendritic cells. Infect lmmun 74:2697-2705. Hu, L., and T. E. Hickey. 2005. Campylobacterjejuni induces secretion of proinfiammatory chemokines from human intestinal epithelial cells. Infect lmmun 73:4437-4440. Hugdahl, M. B., J. T. Beery, and M. P. Doyle. 1988. Chemotactic behavior of Campylobacterjejuni. Infect lmmun 56:1560-1566. Iovine, N. M., S. Pursnani, A. Voldman, G. Wasserman, M. J. Blaser, and Y. Weinrauch. 2008. Reactive nitrogen species contribute to innate host defense against Campylobacterjejuni. Infect Immun 76:986-993. Iovine, N. M., S. Pursnani, A. Voldman, G. Wasserman, M. J. Blaser, and Y. Weinrauch. 2008. Reactive nitrogen species contribute to innate host defense against Campylobacterjejuni. Infect. lmmun.:lAl.01063- 01007. ltano, A. A., and M. K. Jenkins. 2003. Antigen presentation to naive CD4 T cells in the lymph node. Nat Immunol 4:733-739. lwasaki, A. 2007. Mucosal Dendritic Cells. Annu Rev Immunol 25:381- 418. lwasaki, A., and R. Medzhitov. 2004. Toll-like receptor control of the adaptive immune responses. Nat Immunol 5:987-995. 127 75. 76. 77. 78. 79. 80. 81. 82. 83. Jacobs, B. C., A. Belkum, and H. P. Endtz. 2008. Guillain-Barré Syndrome and Campylobacter Infection, p. 245-261. In I. Nachamkin, C. M. Szymanski, and M. J. Blaser (ed.), Campylobacter, 3rd ed. ASM Press, Washington, DC. Janssen, R., K. A. Krogfelt, S. A. Cawthraw, W. van Pelt, J. A. Wagenaar, and R. J. Owen. 2008. Host-pathogen interactions in Campylobacter infections: the host perspective. Clin Microbiol Rev 21 :505-518. Jensen, P. E. 2007. Recent advances in antigen processing and presentation. Nat Immunol 8:1041-1048. Jeyaseelan, S., S. K. Young, M. B. Fessler, Y. Liu, K. C. Malcolm, M. Yamamoto, S. Akira, and G. S. Worthen. 2007. Toll/lL-1 receptor domain-containing adaptor inducing IFN-b (TRIF)-mediated signaling contributes to innate immune responses in the lung during Escherichia coli pneumonia. J Immunol 178:3153-3160. Jin, S., A. Joe, J. Lynett, E. K. Hani, P. Sherman, and V. L. Chen. 2001. leA, a novel surface-exposed Iipoprotein specific to Campylobacter jejuni, mediates adherence to host epithelial cells. Mol Microbiol 39:1225- 1236. Jin, S., Y. C. Song, A. Emili, P. M. Sherman, and V. L. Chen. 2003. leA of Campylobacterjejuni interacts with surface-exposed heat shock protein 90a and triggers signalling pathways leading to the activation of NF-kB and p38 MAP kinase in epithelial cells. Cell Microbiol 5:165-174. Johanesen, P. A., and M. B. Dwinell. 2006. Flagellin-independent regulation of chemokine host defense in Campylobacterjejuni-infected intestinal epithelium. Infect lmmun 74:3437-3447. Johnson, W. M., and H. Lior. 1986. Cytotoxic and cytotonic factors produced by Campylobacterjejuni, Campylobacter coli, and Campylobacter Iaridis. J Clin Microbiol 24:275-281. Jones, M. A., K. L. Marston, C. A. Woodall, D. J. Maskell, D. Linton, A. V. Karlyshev, N. Dorrell, B. W. Wren, and P. A. Barrow. 2004. Adaptation of Campylobacterjejuni NCTC11168 to high-level colonization of the avian gastrointestinal tract. Infect lmmun 72:3769-3776. 128 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. Jones, M. A., S. Totemeyer, D. J. Maskell, C. E. Bryant, and P. A. Barrow. 2003. Induction of proinflammatory responses in the human monocytic cell line THP-1 by Campylobacterjejuni. Infect lmmun 71 :2626- 2633. Kagan, J. C., T. Su, 1'. Horng, A. Chow, S. Akira, and R. Medzhitov. 2008. TRAM couples endocytosis of Toll- like receptor 4 to the induction of intelferon- [beta]. Nat Immunol 9: 361-368. Kapsenberg, M. L. 2003. Dendritic-cell control of pathogen-driven T-cell polarization. Nat Rev Immunol 3:984-993. Karlyshev, A. V., D. Linton, N. A. Gregson, A. J. Lastovica, and B. W. Wren. 2000. Genetic and biochemical evidence of a Campylobacterjejuni capsular polysaccharide that accounts for Penner serotype specificity. Mol Microbiol 35:529-541. Katz, J., P. Zhang, M. Martin, S. N. Vogel, and S. M. Michalek. 2006. Toll-like receptor 2 is required for inflammatory responses to Francisella tularensis LVS. Infect lmmun 74:2809-281 6. Kawai, T., and S. Akira. 2005. Pathogen recognition with Toll-like receptors. Curr Opin Immunol 17:338-344. Kawai, T., and S. Akira. 2006. TLR signaling. Cell Death Differ 13:816- 825. Kawai, T., and S. Akira. 2007. TLR signaling. Semin in Immunol 19:24- 32. Kikuchi, T., C. L. Hahn, S. Tanaka, S. E. Barbour, H. A. Schenkein, and J. G. Tew. 2004. Dendritic cells stimulated with Actinobacillus actinomycetemcomitans elicit rapid gamma interferon responses by natural killer cells. Infect lmmun 72:5089-5096. Kikuchi, T., D. L. Willis, M. Liu, D. B. Purkall, S. Sukumar, S. E. Barbour, H. A. Schenkein, and J. G. Tew. 2005. Dendritic-NK cell interactions in P. gingivalis-specific responses. J Dent Res 84:858-862. 129 94. 95. 96. 97. 98. 99. 100. 101. 102. Kist, M., and S. Bereswill. 2001. Campylobacterjejuni. Contrib Microbiol 8:150-165. KoIb-Maurer, A., A. Unkmeir, U. Kammerer, C. Hubner, T. Leimbach, A. Stade, E. Kampgen, M. Frosch, and G. Dietrich. 2001. Interaction of Neisseria meningitidis with human dendritic cells. Infect lmmun 69:6912- 6922. Konkel, M. E., B. J. Kim, V. Rivera-Amill, and S. G. Garvis. 1999. Bacterial secreted proteins are required for the intemaliztion of Campylobacterjejuni into cultured mammalian cells. Mol Microbiol 32:691- 701. Konkel, M. E., M. R. Monteville, V. Rivera-Amill, and L. A. Joens. 2001. The pathogenesis of Campylobacterjejuni-mediated enteritis. Curr Issues lntest Microbiol 2:55-71. Kuroki, S., T. Haruta, M. Yoshioka, Y. Kobayashl, M. Nukina, and H. Nakanishi. 1991. Guillain-Barré syndrome associated with Campylobacter infection. Pediatr Infect Dis J 10:149-151. Lane, E. M., R. A. Batchelor, A. L. Bourgeois, D. H. Burr, and J. G. Olson. 1987. Urine and faecal lgA response during naturally acquired infection with Campylobacterjejuni. Lancet 1:1141. Lastovica, A. J., and B. M. Alice. 2008. Clinical significance of Campylobacter and related species, other than Campylobacterjejuni and Campylobacter coli, p. 123-149. In I. Nachamkin, C. M. Szymanski, and M. J. Blaser (ed.), Campylobacter, 3rd ed. ASM Press, Washington, DC Leenen, P. J., B. P. Canono, D. A. Drevets, J. S. Voerman, and P. A. Campbell. 1994. TNF-alpha and IFN-gamma stimulate a macrophage precursor cell line to kill Listeria monocytogenes in a nitric oxide- independent manner. J Immunol 153:5141-5147. Lin, J., O. Sahin, L. 0. Michel, and Q. Zhang. 2003. Critical role of multidrug efflux pump CmeABC in bile resistance and in vivo colonization of Campylobacterjejuni. Infect lmmun 71 :4250-4259. 130 103. 104. 105. 106. 107. 108. 109. 110. Lindblom, G. B., C. Ahren, J. Changalucha, R. Gabone, B. Kaijser, L. A. Nilsson, E. Sjogren, A. M. Svennerholm, and M. Temu. 1995. Campylobacterjejuni/coli and enterotoxigenic Escherichia coli (ETEC) in faeces from children and adults in Tanzania. Scand J Infect Dis 27:589- 593. Llu, K., G. D. Victora, T. A. Schwlckert, P. Guermonprez, M. M. Meredith, K. Yao, F.-F. Chu, G. J. Randolph, A. Y. Rudensky, and M. Nussenzweig. 2009. In vivo analysis of dendritic cell development and homeostasis. Science 324:392-397. Lutz, M. B., N. Kukutsch, A. L. Ogilvie, S. Rossner, F. Koch, N. Romani, and G. Schuler. 1999. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. J Immunol Methods 223:77—92. Lutz, M. B., and G. Schuler. 2002. Immature, semi-mature and fully mature dendritic cells: which signals induce tolerance or immunity? Trends Immunol 23:445-449. Maccallum, A. J., D. Harris, G. Haddock, and P. H. Everest. 2006. Campylobacterjejuni-infected human epithelial cell lines vary in their ability to secrete interleukin-8 compared to in vitro-infected primary human intestinal tissue. Microbiology 152:3661-3665. Mansfield, L. S., J. A. Bell, D. L. Wilson, A. J. Murphy, H. M. Elsheikha, V. A. Rathinam, B. R. Fierro, J. E. Linz, and V. B. Young. 2007. CS7BL/6 and congenic interleukin-10-deficient mice can serve as models of Campylobacterjejuni colonization and enteritis. Infect Immun 75:1099—1115. Mansfield, L. S., D. T. Gauthier, S. R. Abner, K. M. Jones, S. R. Wilder, and J. F. Urban. 2003. Enhancement of disease and pathology by synergy of Trichuris suis and Campylobacterjejuni in the colon of immunologically naive swine. Am J Trop Med Hyg 68:70-80. Mansfield, L. S., J. S. Patterson, B. R. Fierro, A. J. Murphy, V. A. Rathinam, J. J. Kopper, N. I. Barbu, T. J. Onifade, and J. A. Bell. 2008. Genetic background of lL-10"' mice alters host-pathogen interactions with Campylobacterjejuni and influences disease phenotype. Microb Pathog 45:241-257. 131 111. 112. 113. 114. 115. 116. 117. 118. 119. McGuirk, P., C. McCann, and K. H. Mills. 2002. Pathogen-specific T regulatory 1 cells induced in the respiratory tract by a bacterial molecule that stimulates interleukin 10 production by dendritic cells: a novel strategy for evasion of protective T helper type 1 responses by Bordetella pertussis. J Exp Med 195:221-231. Mead, P. S., L. Slutsker, V. Dietz, L. F. McCaig, J. S. Bresee, C. Shapiro, P. M. Griffin, and R. V. Tauxe. 1999. Food-related illness and death in the United States. Emerg Infect Dis 5:607-625. Mellits, K. H., J. Mullen, M. Wand, G. Armbruster, A. Patel, P. L. Connerton, M. Skelly, and I. F. Connerton. 2002. Activation of the transcription factor NF-kB by Campylobacterjejuni. Microbiology 148:2753-2763. Mixter, P. F., J. D. Klena, G. A. Flom, A. M. Siegesmund, and M. E. Konkel. 2003. In vivo tracking of Campylobacterjejuni by using a novel recombinant expressing green fluorescent protein. Appl Environ Microbiol 69:2864-2874. Mlzumoto, N., J. Gao, H. Matsushima, Y. Ogawa, H. Tanaka, and A. Takashima. 2005. Discovery of novel immunostimulants by dendritic-cell- based functional screening. Blood 106:3082-3089. Moll, H. 2003. Dendritic cells and host resistance to infection. Cell Microbiol 5:493-500. Monteleone, I., A. M. Platt, E. Jaensson, W. W. Agace, and A. M. Mowat. 2008. IL-10-dependent partial refractoriness to Toll-like receptor stimulation modulates gut mucosal dendritic cell function. Eur J Immunol 38:1533-1547. Monteville, M. R., J. E. Yoon, and M. E. Konkel. 2003. Maximal adherence and invasion of INT 407 cells by Campylobacterjejuni requires the CadF outer-membrane protein and microfilament reorganization. Microbiology 149:153-165. Montoya, M., G. Schiavonl, F. Mattel, I. Gresser, F. Belardelli, P. Borrow, and D. F. Tongh. 2002. Type I interferons produced by dendritic cells promote their phenotypic and functional activation. Blood 99:3263— 3271. 132 120. 121. 122. 123. 124. 125. 126. 127. 128. Moran, A. P. 1997. Structure and conserved characteristics of Campylobacterjejuni lipopolysaccharides. J Infect Dis 176 Suppl 2:S115- 121. Mowat, A. M. 2003. Anatomical basis of tolerance and immunity to Intestinal antigens. Nat Rev Immunol 3:331-341. Muraille, E., C. De Trez, M. Brait, P. De Baetselier, 0. Leo, and Y. Carlier. 2003. Genetically resistant mice lacking MyDB8-adapter protein display a high susceptibility to Leishmania major infection associated with a polarized Th2 response. J Immunol 170:4237-4241. Niess, J. H., S. Brand, X. Gu, L. Landsman, S. Jung, B. A. McCormick, ; J. M. Vyas, M. Boes, H. L. Ploegh, J. G. Fox, D. R. Littman, and H.-C. k Reinecker. 2005. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 307:254-258. Oelschlaeger, T. A., P. Guerry, and D. J. Kopecko. 1993. Unusual microtuble-dependent endocytosis mechanisms triggered by Campylobacterjejuni and Citrobacter freundii. Proc Natl Acad Sci U S A 90:6884-6888. Oelschlaeger, T. A., P. Guerry, and D. J. Kopecko. 1993. Unusual Microtuble-Dependent Endocytosis Mechanisms Triggered by Campylobacterjejuni and Citrobacter freundii. Proceedings of the National Academy of Sciences 90:6884-6888. Olson, C. K., S. Ethelberg, W. Pelt, and R. V. Tauxe. 2008. Epidemiology of Campylobacterjejuni Infections in Industrialized Nations, p. 163-189. In I. Nachamkin, C. M. Szymanski, and M. J. Blaser (ed.), Campylobacter, 3rd ed. ASM Press, Washington, DC. Ouyang, X., H. Negishi, R. Takeda. Y. Fujita, T. Tanlguchi, and K. Honda. 2007. Cooperation between MyD88 and TRIF pathways in TLR synergy via IRF5 activation. Biochem Biophys Res Commun 354:1045- 1051. Overbergh, L., D. Valckx, M. Waer, and C. Mathieu. 1999. Quantification of murine cytokine mRNAs using real time quantitative reverse transcriptase PCR. Cytokine 11:305-312. 133 129. 130. 131. 132. 133. 134. 135. 136. 137. Parkhill, J., B. W. Wren, K. Mungall, J. M. Ketley, C. Churcher, D. Basham, T. Chillingworth, R. M. Davies, T. Feltwell, S. Holroyd, K. Jagels, A. V. Karlyshev, S. Moule, M. J. Pallen, C. W. Penn, M. A. Quail, M. A. Rajandream, K. M. Rutherford, A. H. M. van Vliet, S. Whitehead, and B. G. Barrell. 2000. The genome sequence of the food- borne pathogen Campylobacterjejuni reveals hypervariable sequences. Nature 403:665-668. Pearson, B. M., C. Pin, J. Wright, K. I'Anson, T. Humphrey, and J. M. Wells. 2003. Comparative genome analysis of Campylobacterjejuni using whole genome DNA microarrays. FEBS Lett 554:224-230. Pei, Z., and M. J. Blaser. 1993. PEB1, the major cell-binding factor of Campylobacterjejuni, is a homolog of the binding component in gram- negative nutrient transport systems. J Biol Chem 268:18717-18725. Pei, 2., C. Burucoa, B. Grlgnon, S. Baqar, X. Z. Huang, D. J. Kopecko, A. L. Bourgeois, J. L. Fauchere, and M. J. Blaser. 1998. Mutation in the peb1A locus of Campylobacterjejuni reduces interactions with epithelial cells and intestinal colonization of mice. Infect Immun 66:938-943. Perez-Perez, G. l., D. L. Cohn, R. L. Guerrant, C. M. Patton, L. B. Reller, and M. J. Blaser. 1989. Clinical and immunologic significance of cholera-like toxin and cytotoxin production by Campylobacter species in patients with acute inflammatory diarrhea in the USA. J Infect Dis 160:460-468. Perona-erght, G., S. J. Jenkins, A. Crawford, D. Gray, E. J. Pearce, and A. S. MacDonald. 2006. Distinct sources and targets of IL-10 during dendritic cell-driven Th1 and Th2 responses in vivo. Eur J Immunol 36:2367-2375. Pfaffl, M. W. 2001. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 29:e45. Pope, J. E., A. Krizova, A. X. Garg, H. Thiessen-Philbrook, and J. M. Ouimet. 2007. Campylobacter reactive arthritis: a systematic review. Semin Arthritis Rheum 37:48-55. Power, M. R., B. Li, M. Yamamoto, S. Akira, and T.-J. Lin. 2007. A role of Toll-IL-1 receptor domain-containing adaptor-inducing IFN-b in the host 134 138. 139. 140. 141. 142. 143. 144. 145. 146. response to Pseudomonas aeruginosa lung infection in mice. J Immunol 1 78:3170-3176. Pulendran, B. 2004. Modulating TH1/T H2 responses with microbes, dendritic cells, and pathogen recognition receptors. Immunol Res 29:187- 1 96. Rad, R., L. Brenner, A. Krug, P. Voland, J. Mages, R. Lang, S. Schwendy, W. Reindl, A. Dossumbekova, W. Ballhorn, H. Wagner, R. M. Schmid, S. Bauer, and C. Prinz. 2007. Toll-like receptor-dependent activation of antigen-presenting cells affects adaptive immunity to Helicobacter pylori. Gastroenterology 1 33:150-163 e153. Randolph, G. J., V. Angeli, and M. A. Swartz. 2005. Dendritic-cell trafficking to lymph nodes through lymphatic vessels. Nat Rev Immunol 5:61 7-628. - Rathinam, V. A., K. A. Hoag, and L. S. Mansfield. 2008. Dendritic cells from CS7BU6 mice undergo activation and induce Th1-effector cell responses against Campylobacterjejuni. Microbes Infect 1 0:1316—1324. Remoli, M. E., E. Giacomini, G. Lutfalla, E. Dondi, G. Oreflci, A. Battistini, G. Uze, S. Pellegrini, and E. M. Coccia. 2002. Selective expression of type I IFN genes in human dendritic cells infected with Mycobacterium tuberculosis. J Immunol 169:366-374. Rock, K. L., and L. Shen. 2005. Cross-presentation: underlying mechanisms and role in immune surveillance. Immunol Rev 207:166-183. Ruiz-Palacios, G. M., J. J. Calva, L. K. Pickering, Y. Lopez-Vidal, P. Volkow, H. Pezzarossi, and M. S. West. 1990. Protection of breast-fed infants against Campylobacter diarrhea by antibodies in human milk. J Pediatr 1 16:707-713. Schnare, M., A. C. Holt, K. Takeda, S. Akira, and R. Medzhitov. 2000. Recognition of CpG DNA is mediated by signaling pathways dependent on the adaptor protein My088. Curr Biol 10:1139. Scott, D. A. 1997. Vaccines against Campylobacterjejuni. J Infect Dis 176 Suppl 2:8183-188. 135 147. 148. 149. 150. 151. 152. 153. 154. Scott, K., M. Manunta, C. Germain, P. Smith, M. Jones, P. Mitchell, D. Dessi, K. Branigan Bamford, R. I. Lechler, P. L. Fiori, G. R. Foster, and G. Lombardi. 2005. Qualitatively distinct patterns of cytokines are released by human dendritic cells in response to different pathogens. Immunology 1 16:245-254. Siegesmund, A. M., M. E. Konkel, J. D. Klena, and P. F. Mixter. 2004. Campylobacterjejuni infection of differentiated THP-1 macrophages results in interleukin 1-b release and caspase—1-independent apoptosis. Microbiology 150:561-569. Skirrow, M. B., and M. J. Blaser. 2000. Clinical aspects of Campylobacter infection, p. 69-88. In I. Nachamkin and M. J. Blaser (ed.), Campylobacter, 2nd ed. ASM Press, Washington, DC Skokos, D., and M. C. Nussenzweig. 2007. CD8 DCs induce lL-12- independent Th1 differentiation through Delta 4 Notch-like ligand in response to bacterial LPS. J Exp Med 204:1525-1531. Scares, H., H. Waechter, N. Glaichenhaus, E. Mougneau, H. Yagita, O. Mizenlna, D. Dudziak, M. C. Nussenzweig, and R. M. Stelnman. 2007. A subset of dendritic cells induces CD4+ T cells to produce IFN-g by an IL- 12-independent but CD70-dependent mechanism in vivo. J Exp Med 204: 1 095-1 106. Sorvillo, F. J., L. E. Lieb, and S. H. Waterman. 1991. Incidence of campylobacteriosis among patients with AIDS in Los Angeles County. J Acquir Immune Deflc Syndr 4:598-602. Spiller, S., S. Dreher, G. Meng, A. Grabiec, W. Thomas, T. Hartung, K. Pfeffer, H. Hochrein, H. Brade, W. Bessler, H. Wagner, and C. J. Kirschning. 2007. Cellular recognition of trimyristoylated peptide or enterobacterial lipopolysaccharide via both TLR2 and TLR4. J Biol Chem 282:13190-13198. St Michael, F., C. M. Szymanski, J. LI, K. H. Chan, N. H. Khleu, S. Larocque, W. W. Wakarchuk, J. R. Brisson, and M. A. Monteiro. 2002. The structures of the lipooligosaccharide and capsule polysaccharide of Campylobacterjejuni genome sequenced strain NCTC 11168. Eur J Biochem 269:5119-5136. 136 155. 156. 157. 158. 159. 160. 161. 162. 163. Steiner, T. S. 2007. How flagellin and Toll-like receptor 5 contribute to enteric infection. Infect Immun 75:545-552. Strober, W., P. J. Murray, A. Kitani, and T. Watanabe. 2006. Signalling pathways and molecular interactions of NOD1 and N002. Nat Rev Immunol 6:9. Szymanski, C. M., F. S. Michael, H. C. Jarrell, J. Li, M. Gilbert, S. Larocque, E. Vlnogradov, and J.-R. Brisson. 2003. Detection of conserved N-Iinked glycans and phase-variable Iipooligosaccharides and capsules from Campylobacter cells by mass spectrometry and high resolution magic angle spinning NMR spectroscopy. J Biol Chem 278:24509-24520. Takata, T., S. Fujimoto, and K. Amako. 1992. Isolation of nonchemotactic mutants of Campylobacterjejuni and their colonization of the mouse intestinal tract. Infect Immun 60:3596-3600. Tam, C. C., S. J. O'Brien, I. Petersen, A. Islam, A. Hayward, and L. C. Rodrigues. 2007. Guillain-Barré syndrome and preceding infection with Campylobacter, Influenza and Epstein-Barr virus in the general practice research database. PLoS ONE 2:6344. Taylor, D. N., P. Echeverria, C. Pitarangsi, J. Seriwatana, L. Bodhidatta, and M. J. Blaser. 1988. Influence of strain characteristics and immunity on the epidemiology of Campylobacter infections in Thailand. J Clin Microbiol 26:863-868. Taylor, D. N., D. M. Perlman, P. D. Echeverria, U. Lexomboon, and M. J. Blaser. 1993. Campylobacter immunity and quantitative excretion rates in Thai children. J Infect Dis 168:754-758. Torres, D., M. Barrier, F. Bihl, V. J. Quesniaux, I. Maillet, S. Akira, B. Ryffel, and F. Erard. 2004. Toll-like receptor 2 is required for optimal control of Listeria monocytogenes infection. Infect Immun 72:2131-2139. Trevejo, J. M., M. W. Marino, N. Philpott, R. Josien, E. C. Richards, K. B. Elkon, and E. Falck-Pedersen. 2001. TNF-a-dependent maturation of local dendritic cells is critical for activating the adaptive immune response to virus infection. Proc Natl Acad Sci U S A 98:12162-12167. 137 164. 165. 166. 167. 168. 169. 170. 171. 172. 173. Urban, B. C., and D. J. Roberts. 2002. Malaria, monocytes, macrophages and myeloid dendritic cells: sticking of infected erythrocytes switches off host cells. Curr Opin Immunol 14:458-465. Uronen-Hansson, H., L. Steeghs, J. Allen, G. L. J. Dixon, M. Osman, P. van der Ley, S. Y. C. Wong, R. Callard, and N. Klein. 2004. Human dendritic cell activation by Neisseria meningitidis: phagocytosis depends on expression of lipooligosaccharide (LOS) by the bacteria and is required for optimal cytokine production. Cellular Microbiology 6:625. van Spreeuwel, J. P., G. C. Duursma, C. J. Meijer, R. Bax, P. C. Rosekrans, and J. Lindeman. 1985. Campylobacter colitis: histological immunohistochemical and ultrastructural findings. Gut 26:945-951. Walan, A., C. Dahlgren, E. Kihlstrom, O. Stendahl, and R. Lock. 1992. Phagocyte killing of Campylobacterjejuni in relation to oxidative activation. Apmis 100:424-430. Walker, R. I., E. A. Schmauder—Chock, J. L. Parker, and D. Burr. 1988. Selective association and transport of Campylobacterjejuni through M cells of rabbit Peyer's patches. Can J Microbiol 34:1142-1147. Walzer, T., M. Dalod, S. H. Robbins, L. Zitvogel, and E. Vivier. 2005. Natural-killer cells and dendritic cells: 'I'union fait la force”. Blood 106:2252-2258. Wassenaar, T. M. 1997. Toxin production by Campylobacter spp. Clin Microbiol Rev 10:466-476. Wassenaar, T. M., and M. J. Blaser. 1999. Pathophysiology of Campylobacterjejuni infections of humans. Microbes and Infection 1 :1023-1033. Wassenaar, T. M., and M. J. Blaser. 1999. Pathophysiology of Campylobacterjejuni infections of humans. Microbes Infect 1 :1023-1033. Wassenaar, T. M., M. Engelskirchen, S. Park, and A. Lastovica. 1997. Differential uptake and killing potential of Campylobacterjejuni by human peripheral monocytes/macrophages. Med Microbiol Immunol (Berl) 186: 1 39-1 44. 138 174. 175. 176. 177. 178. 179. 180. 181. 182. Wassenaar, T. M., B. A. van der Zeijst, R. Ayling, and D. G. Newell. 1993. Colonization of chicks by motility mutants of Campylobacterjejuni demonstrates the importance of flagellin A expression. J Gen Microbiol 139 Pt 6:1171-1175. Watson, R. O., and J. E. Galan. 2008. Campylobacterjejuni survives within epithelial cells by avoiding delivery to Iysosomes. PLoS Pathogens 4:e14. Watson, R. O., and J. E. Galan. 2005. Signal transduction in Campylobacterjejuni-induced cytokine production. Cell Microbiol 7:655- 665. Watson, R. O., V. Novik, D. Hofreuter, M. Lara-Tejero, and J. E. Galan. 2007. A MyDBB-deficient mouse model reveals a role for Nramp1 in Campylobacterjejuni infection. Infect Immun 75:1994-2003. Weinstein, S. L., A. J. Finn, S. H. Dave, F. Meng, C. A. Lowell, J. S. Sanghera, and A. L. DeFranco. 2000. Phosphatidylinositol 3-kinase and mTOR mediate lipopolysaccharide-stimulated nitric oxide production in macrophages via interferon-b. J Leukoc Biol 67:405-414. Wilson, D. L., J. A. Bell, V. B. Young, S. R. Wilder, L. S. Mansfield, and J. E. Linz. 2003. Variation of the natural transformation frequency of Campylobacterjejuni in liquid shake culture. Microbiology 149:3603-3615. Yamamoto, M., S. Sato, H. Hemmi, K. Hoshino, T. Kaisho, H. Sanjo, O. Takeuchi, M. Suglyama, M. Okabe, K. Takeda, and S. Aklra. 2003. Role of adaptor TRIF in the MyDB8-independent Toll-like receptor signaling pathway. Science 301:640-643. Yamamoto, M., S. Sato, K. Mori, K. Hoshino, O. Takeuchi, K. Takeda, and S. Akira. 2002. Cutting Edge: A novel Toll/lL-1 receptor domain- containing adapter that preferentially activates the IFN-b promoter in the Toll-like receptor signaling. J Immunol 169:6668-6672. Young, K. T., L. M. Davis, and V. J. DiRita. 2007. Campylobacterjejuni: molecular biology and pathogenesis. Nat Rev Micro 5:665. 139 183. 184. 185. 186. 187. 188. 189. 190. 191. Young, K. T., L. M. Davis, and V. J. DiRita. 2007. Campylobacterjejuni: molecular biology and pathogenesis. Nat Rev Micro 5:665-679. Young, V. B., and L. S. Mansfield. 2005. Campylobacter Infection - Clinical Context, p. 1-12. In J. M. Ketley, M. E. Konkel (ed.), Campylobacter. Molecular and Cellular Biology, vol. Horizon Bioscience, Norfolk, UK. Yuki, N., and M. Odaka. 2005. Ganglioside mimicry as a cause of Guillain-Barre syndrome. Curr Opin NeuroI 18:557-561. Zhao, C., M. W. Wood, E. E. Galyov, U. E. Hopken, M. Lipp, H. C. Bodmer, D. F. Tough, and R. W. Carter. 2006. Salmonella typhimurium infection triggers dendritic cells and macrophages to adopt distinct migration patterns in vivo. Eur J Immunol 36:2939-2950. Zheng, J., J. Meng, S. Zhao, R. Singh, and W. Song. 2008. Campylobacter-induced interleukin-8 secretion in polarized human intestinal epithelial cells requires Campylobacter-secreted cytolethal distending toxin- and Toll-like receptor-mediated activation of NF-kB. Infect Immun 76:4498-4508. Zilbauer, M., N. Dorrell, P. K. Boughan, A. Harris, B. W. Wren, N. J. Klein, and M. Bajaj-Elliott. 2005. Intestinal innate immunity to Campylobacterjejuni results in induction of bactericidal human beta- defensins 2 and 3. Infect lmmun 73:7281-7289. Zilbauer, M., N. Dorrell, A. Elmi, K. J. Lindley, S. SchulIer, H. E. Jones, N. J. Klein, G. Nunez, B. W. Wren, and M. Bajaj-Elliott. 2007. A major role for intestinal epithelial nucleotide oligomerization domain 1 (NOD1) in eliciting host bactericidal immune responses to Campylobacterjejuni. Cell Microbiol 9:2404-2416. Zilbauer, M., N. Dorrell, B. W. Wren, and M. Bajaj-Elliott. 2008. Campylobacterjejuni-mediated disease pathogenesis: an update. Trans R Soc Trop Med Hyg 102:123—129. Ziprin, R. L., C. R. Young, J. A. Byrd, L. H. Stanker, M. E. Hume, S. A. Gray, B. J. Kim, and M. E. Konkel. 2001. Role of Campylobacterjejuni potential virulence genes in cecal colonization. Avian Dis 45:549-557. 140