1,] 3.vr3. a} F... $712. . . , . . . ,. ‘ .4, ,4. ....4.1f.!41 : égg} . ‘ ‘ : . . a? 3%; a :1 LIBRARY ' a 006 Michigan State University ] This is to certify that the dissertation entitled THE EFFECTS OF CAMPYLOBACTER JEJUNI AND TRICHURIS SUIS ON LOCAL CYTOKINE EXPRESSION IN THE SWINE COLON presented by Kathryn Marie Jones has been accepted towards fulfillment of the requirements for the Doctoral degree in Microbiology and Molecular Genetics I l k l 0 " v.41 , Major Professor’s Sign c/aa/a 005 Date ure MSU is an Affirmative Action/Equal Opportunity Institution 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 2mm THE EFFECTS OF CAMPYLOBACTER JEJUNI AND TRICHURIS SUIS ON LOCAL CYTOKINE EXPRESSION IN THE SWINE COLON By Kathryn Marie Jones A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Microbiology and Molecular Genetics 2005 ABSTRACT THE EFFECTS OF CAMPYLOBACTER JEJUNI AND TRICHURIS SUIS ON LOCAL CYTOKINE EXPRESSION IN THE SWINE COLON By Kathryn Marie Jones Campylobacterjejuni is a significant enteric pathogen of humans, but it is a non- pathogenic resident in the gastrointestinal tract of pigs. Upon infection with the nematode Trichuris suis, C. jejuni opportunistically invades the mucosa in the distal colon, resulting in severe disease and pathology. The central hypothesis of this dissertation project is that natural resistance to C. jejuni infection in swine is due to a proinflammatory response in the intestine. A related hypothesis is that T. suis stimulates an anti-inflammatory response which alters the protective response to C. jejuni. Three experimental designs were used to test these hypotheses. In the first experiment, weaned piglets were inoculated rectally with C. jejuni 81-176pWM1007, then samples of the distal colon and lymphoglandular complexes were taken for quantitation of cytokine mRNA expression by real time PCR. The data showed that C. jejuni stimulated a proinflammatory response in vivo. In the second experiment, weaned piglets were orally inoculated with embryonated T. suis eggs, C. jejuni 33292, or both, then samples of the jejunum, proximal colon, and distal colon were taken for quantitation of cytokine mRNA expression by real time PCR. The data showed that C. jejuni stimulated a proinflammatory response in the jejunum while T. suis stimulated an anti-inflammatory response in the jejunum and proximal colon. When dually infected, T. suis down regulated proinflammatory responses to C. jejuni in the jejunum and proximal colon. In the third experiment, differentiated an undifferentiated swine intestinal epithelial cells (IPEC-l) were infected with C. jejuni 81-176 or 33292 and IL-18 production was measured in culture supematants by ELISA. Only differentiated cells produced significant IL-18 in response to C. jejuni. Taken together, these data indicate that C. jejuni stimulates a local proinflammatory response in vivo, which can be attributed in part to the interaction Of C. jejuni with intestinal epithelial cells. T. suis stimulates an anti-inflammatory response and down regulates proinflammatory cytokines, abrogating the protective response to C. jejuni in vivo. This immunomodulation could facilitate C. jejuni invasion into intestinal epithelial cells and deeper tissues in vivo, resulting in destruction of the epithelium and severe pathology. Cepyright by KATHRYN MARIE JONES 2005 Acknowledgements There were several people who were essential to completion Of this dissertation project and I would like to acknowledge them here. Dr. Linda Mansfield was my mentor and gave me continued guidance and encouragement throughout my graduate studies. My committee members, Drs. John Linz, Katheryn Meek, Richard Schwartz, and Charles Mackenzie, were always willing to Offer guidance and shared their knowledge. My labmates, Lakeisha Cunningham, Dr. Geetha Parthasarathy, and Stacey Wilder gave me emotional support as well as assisted me in my experiments, helped me trouble shoot my problems, and critiqued my writing. Dr. Julia Bell and David Wilson never failed to share their expertise on all things pertaining to Campylobacter jejuni. Drs. Pawin Padungtod, BO Norby, and Jean Gaymer shared their veterinary expertise and technical skills in developing and executing the animal models. Dr. Matti Kiupel performed the staining and microscopy on the animal tissues and shared his expertise in histopathology and immunohistochemistry. Drs. Joseph Urban and Harry Dawson were our collaborators from the USDA and trained me in real time PCR and graciously allowed me to analyze all of my tissue samples in their facility. Drs. John Baker and Vilma Yuzbasiyan-Gurkan assisted me in securing funding that was essential to completing my project. Dr. Shirley Owens trained me in confocal microscopy and shared her expertise in fluorescent microscopy. Christopher Kar, my husband, not only provided unfailing emotional support throughout my graduate studies, but he was also my 24 hour home technical support and made it possible for me to write this dissertation. To all of you and many more who helped me through my degree, I am eternally grateful. TABLE OF CONTENTS List of Tables ....................................................................................... viii List of Figures ......................................................................................... ix Chapter 1: Literature Review C ampylobacter jejuni ........................................................................ 1 Trichuris suis ............................................................................... 15 Rationale for study ......................................................................... 24 References ................................................................................... 29 Chapter 2: Campylobacterjejuni invades distal colonic lymphoglandular complexes causing upregulation of proinflammatory cytokines in a swine rectal challenge model Abstract ....................................................................................... 37 Introduction ................................................................................. 38 Materials and Methods ..................................................................... 41 Results ....................................................................................... 5 1 Discussion ................................................................................... 55 References ................................................................................... 61 Figures ....................................................................................... 64 Chapter 3: T richuris suis down regulates proinflammatory cytokines in the proximal colon of pigs concurrently infected with C. jejuni Abstract ....................................................................................... 75 Introduction ................................................................................. 77 Materials and Methods ..................................................................... 79 Results ....................................................................................... 86 Discussion ................................................................................... 90 References ................................................................................... 98 Figures ...................................................................................... 101 Chapter 4: Campylobacterjejum' stimulates IL-18 secretion from differentiated intestinal epithelial cells Abstract ..................................................................................... 109 Introduction ................................................................................ 1 1 1 Materials and Methods ................................................................... 115 Results ...................................................................................... 122 Discussion ................................................................................. 124 References ................................................................................. 127 vi Figures ...................................................................................... 131 Chapter 5: Summary and conclusions Summary and conclusions .............................................................. 138 References ................................................................................. 1 64 vii LIST OF TABLES Chapter 2 Table 1: Summary Of Campylobacter detection and identification ............... 64 Table 2:Primer and probe sequences for porcine specific cytokines, chemokines, and iNOS .................................................................................. 73 Chapter 3 Table 1: Summary of Campylobacter detection and identification ............... 102 viii LIST OF FIGURES Chapter 2 Figure l: Amplification of the QRDR of C. jejuni gyrA from distal colon DNA from experimentally infected pigs ....................................................... 65 Figure 2: Ala I digest of Campylobacter 23s rRNA gene amplified from pig tissues by PCR .............................................................................. 66 Figrue 3: PCR testing for gfp ............................................................ 67 Figure 4*: Immunohistochemistry of Campylobacter infected IPEC-l cells ...... 68 Figure 5*: Immunohistochemistry of colonic tissues from Campylobacter infected pigs ........................................................................................... 69 Figure 6: PCR testing DNA isolated from LCM samples ............................ 70 Figure 7: Ala I digest of Campylobacter 23s rRNA gene PCR product amplified from microdissected crypts and submucosa ............................................ 71 Figure 8*: Cytokine expression in distal colonic tissues .............................. 72 Chapter 3 Figure 1: Inoculation timeline .......................................................... 101 Figure 2: RFLP analysis of the Campylobacter 23S rRNA gene .................. 103 Figrue 3*: H&E stained sections of colonic tissue ................................... 104 Figure 4*: H&E stained sections of colonic tissue .................................. 105 Figure 5*: H&E sections of LGCs from infected pigs ............................... 106 Figure 6*: Immunohistochemistry from colonic sections ........................... 107 Figure 7*: Cytokine expression in intestinal tissues ................................. 108 Chapter 4 Figure 1: Scanning electron micrograph of polarized IPEC-l cells (12 days) on transwell membranes ..................................................................... 1 3 1 ix Figure 2: IL-18 secretion from differentiated IPEC-l cells ......................... 132 Figrue 3: IL-18 secretion from differentiated IPEC-l cells ......................... 133 Figure 4: IL-1 8 secretion from undifferentiated IPEC-l cells ...................... 134 Figure 5: Optimization of amplification parameters .................................. 135 Figure 6: Determination of optimal number of cycles for amplification .......... 136 Figure 7: IL-18 expression from IPEC-l cells ....................................... 137 *Images in this dissertation are presented in color Chapter 1: Literature Review Campylobacterjejuni Basic Microbiology Campylobacter jejuni is a gram negative, slender, curved rod measuring 0.2-0.5um wide and 0.5-5 um long [1]. It is motile due to the presence of uni- or bipolar flagella. It grows best at 42°C in a microaerophilic atmosphere of 3-15% oxygen and 2-5% carbon dioxide [2]. C. jejuni is nonsaccharolytic and requires complex growth media for laboratory culture [2]. The genome is small, approximately 1.6 to 1.7Mbp, and AT rich with a GC ratio of approximately 30%. In addition, extrachromosomal elements have been discovered, including conjugative plasmids and bacteriophages [2]. C. jejuni is naturally transforrnable, able to take up DNA from the environment and incorporate it into its genome [2-4]. Several genetic tools have been used to identify variants within the species, including pulse field gel electrophoresis, restriction fragment length polymorphism typing of ribosomal and flagellar genes, and amplified fragment length polymorphism analysis [5-9]. These methods have revealed extensive genetic diversity within the species. Clinical Disease C. jejuni is a significant enteric pathogen of humans and is the leading cause of foodborne bacterial illness worldwide. Ingestion of as few as 500 organisms can lead to diarrhea] disease in susceptible humans [1]. Symptoms develop 1 to 7 days after ingestion of bacteria, with an average 24 to 48 hour incubation period [10]. Clinically, fever, abdominal pain, and general malaise precede diarrhea] disease. Diarrhea is profuse and can range from watery to mucoid and bloody in character [11]. C. jejuni colonizes the mucus overlying the epithelium of the distal ileum and colon [1] and colonic biopsies from infected individuals show an acute inflammatory response with infiltration of the epithelium and lamina propria with neutrophils and mononuclear cells [10]. Histologic examination of colonic tissues from experimentally infected colostrum deprived piglets that developed diarrhea] disease after C. jejuni inoculation showed ulcerative colitis with denuded villus tips as well as infiltration of the submucosa with neutrophils [12]. C. jejuni disrupts the absorptive capacity of epithelial cells by invasion, by the effect of toxins, or through induction of an inflammatory response [2]. Gastrointestinal illness due to C. jejuni infection usually resolves afler an average of 10 days [1], but the bacteria can be excreted in the feces for up to 16 days [10]. Due to the fact that illness is usually self limiting, treatment is generally restricted to supportive care with fluid therapy. However, erythromycin is the drug of choice for severe cases or immunosuppressed individuals [11]. The most common clinical manifestation of C. jejuni infection is diarrhea] disease, however, in rare cases extraintestinal complications arise. Local spread of the bacteria can induce cholecystitis, pancreatitis, peritonitis, and massive GI hemorrhage. Systemic spread can cause meningitis, endocarditis, septic arthritis, osteomyelitis, and neonatal sepsis [11]. One notable sequela of C. jejuni infection is Guillain-Barré syndrome (CBS). CBS develops in 1 in every 1000 cases of C. jejuni infection, developing approximately 1-3 weeks after the onset of diarrheal illness. CBS is an ascending peripheral neuropathy that is most commonly associated with Penner serotype 0:19 strains in the US, and type 0:41 in South Africa [1], 13]. Molecular mimicry between C. jejuni LPS and human gangliosides results in the generation of antibodies that attack the myelin sheaths of peripheral nerves, causing nerve paralysis [11, 13]. The combination of C. jejuni causing significant diarrhea] disease and the debilitating sequelae, though rare, warrants intense investigation of this pathogen. Epidemiology Epidemiologic studies have showed a high prevalence of C. jejuni in cases of infectious diarrhea. Diarrhea] disease due to C. jejuni in the U. S. is more prevalent than diarrhea] disease due to Salmonella, Shigella, and E. coli 0157:H7 [11]. The CDC estimates 2.4 million cases of campylobacteriosis each year in the US [11]. Sporadic illnesses account for the majority of reported cases and are generally attributed to consumption of improperly prepared poultry products or other foods contaminated with raw poultry fluids [1]. Seasonal outbreaks occur with bimodal peaks in May and October, and are usually due to consumption of contaminated raw milk or water sources contaminated with animal feces [1]. Children and young adults, particularly young males, are most commonly infected, with most deaths occurring in infants, elderly, and immunosuppressed individuals [1]. The preferred environment for C. jejuni is the intestinal tract of warm blooded animals. It is a commensa] in a variety of wild and domestic mammals and birds, including chickens, cattle, sheep, and swine, and animals can serve as a source of infection for humans [14, 15]. The clinical picture and epidemiology of C. jejuni infections is different in developing countries. Diarrhea] disease due to C. jejuni in developing countries is usually less severe than in developed countries and can range from watery diarrhea, without blood or inflammation, to severe inflammatory, bloody diarrhea accompanied by fever and abdominal pain [16]. Campylobacteriosis does not Show seasonal peaks in developing countries and this could be attributed to climate variations, or to inadequate surveillance for epidemics [16]. In developing countries, Campylobacteriosis is generally a pediatric disease being the most frequently cause of diarrhea] disease in infants less than 6 months of age [16]. In community based studies 40-60% of diarrheic children less than 5 years of age were positive for the organisms on fecal cultures [17]. Additionally, polymicrobial infection is common, with protozoa, viruses, and other enteric bacteria coinfecting individuals [18, 19]. Sources for infection are commonly contact with animals and environmental contamination from animal feces [17, 20, 21]. Virulence Factors C. jejuni causes diarrhea] disease in humans by colonizing the intestinal tract and invading the epithelium which results in decreased absorption due to loss of epithelial function [2, 10]. The events that lead to invasion of the epithelium by C. jejuni include colonization of the mucus layer overlying the epithelium, adherence to epithelial cells, and invasion of epithelial cells. Several virulence factors have been described that play a role in the pathogenesis of C. jejuni infection. Motility: C. jejuni are motile by means of one or two polar flagella [2]. The flagellin protein is encoded by two adjacent genes on the chromosome designated flaA and flaB [2]. Disruption of flaB results in mutants that have flagella of norrna] length, exhibit normal motility, and invade INT-407 cells at wild type levels [22]. In contrast, disruption of the flaA gene results in mutants that are aflagellate or have truncated flagella and are unable to adhere to or invade epithelial cells [22, 23], or colonize the ceca of chicks [24]. Mutants with paralyzed flagella are non motile and non-invasive, but are able to adhere to cells [23]. However, non motile mutants are able to invade epithelial cells in vitro if centrifugation is performed to bring the bacteria into contact with epithelial cells [22]. These data suggest that motility is necessary for invasion [22, 23]. C. jejuni motility is also affected by environmental factors. C. jejuni display normal swarming ability on motility agar of neutral and basic pH (pH=7.3 and 8.5, respectively). However, in an acidic pH of 5.0, swarming ability is decreased despite normal morphology. C. jejuni displays smooth swimming patterns with longer paths and greater velocity in viscous medium that mimics the viscosity of intestinal mucous. Increased viscosity of culture medium also increases adherence to and invasion of Caco-2 cells [25]. The motility of C. jejuni is believed to allow penetration of the intestinal mucous layer in vivo and successful colonization of the host [2]. Iron acquisition: C. jejuni requires iron for optimal growth and prOper cell division. Under iron limiting conditions, C. jejuni produces five proteins ranging in size from 52kDa to 89kDa. Three of these proteins with molecular weights of 74kDa, 76kDa, and 82kDa proteins are surface expressed. Cjejum' is also capable of producing siderophores and of using the exogenous siderophores enterochelin and ferrichrome for iron acquisition [26]. C. jejuni has evolved mechanisms for obtaining iron from its environment necessary for growth. However, in the presence of oxygen iron is able to generate oxygen radicals that can cause damage to DNA, proteins, and membrane lipids [27]. Therefore C. jejuni has also evolved mechanisms to survive in conditions of environemental stress. Alkyl hydroperoxide reductase C (AhpC) is transcriptionally regulated by iron concentrations and has been shown to be involved in resistance to oxidative stress caused by cumene hydroperoxide (CHP) and survival in atmospheric oxygen [28]. C. jejuni also possesses two Fur homolog iron responsive elements, designated fur and perR. PerR is a repressor of ahpC and katA, and perR mutants Show hyperresistance to CHP and hydrogen peroxide [29]. An additional protein, designated Dps, has been identified in C. jejuni. Dps of C. jejuni is structurally similar to the DNA-binding protein from starved cells family of proteins. Dps does not bind DNA, but it does bind to iron. Dps confers resistance to hydrogen peroxide [27]. Chemotaxis: In vivo, C. jejuni colonizes the mucous blanket overlying the epithelium of the intestines [30, 31]. This allows them to associate closely with intestinal epithelial cells [31]. Chemotactic movement is one mechanism by which the bacteria reaches its target niche and has shown to be essential for colonization in vivo [32]. In vitro studies have identified several compounds found in the gastrointestinal environment that affect chemotaxis of C. jejuni. Mucin, the major component of intestinal mucous and a component of bile, attracts C. jejuni [33]. Amino acids and organinc acids, including L- aspartate, L-glutamate, L-serine, L-fucose, pyruvate, succinate, fumarate, citrate, L- malate, and OI-ketoglutarate also attract C. jejuni [32, 33]. These compounds as well as low pH and high osmolarity have been shown in vitro to upregulate the flaA promoter 028, while increased viscosity causes downregulation [34]. It is hypothesized that increased production of FlaA in response to chemoattractants would facilitate maintenance of flagellar filaments, assisting in actions necessary for survival in the host. These actions would include escaping the acid pH of the stomach and chemotaxis toward mucin or amino acids that could be used as energy sources. Decreased motility in a viscous environment simulating the mucous blanket overlying the epithelium would be advantageous because it would signal that the desired niche had been reached [34]. Adherence: Once C. jejuni successfully traverse the intestinal mucus layer and locate the epithelial cells, they must adhere in order to enter the cells and translocate to deeper tissues. Several factors have been identified that mediate adherence. C. jejuni come into close apposition with the cell surface, particularly at the intercellular spaces [35]. Fibronectin is a component of the basement membrane underlying epithelial cells and is also present in regions of cell to cell contact in the GI epithelium [36]. C. jejuni binds to the glycoprotein fibronectin via a 37kDa surface exposed outer membrane protein designated CadF [37]. CadF also mediates adherence to intestinal epithelial cells in vitro, as mutation of CadF reduces adherence by 59% [36]. CadF has also been shown to be important for C. jejuni in vivo as it is necessary for successful colonization of chicken ceca [38]. le is a surface lipoprotein that interacts with Heat shock protein 90a, mediating adherence to HEp-2 epithelial cells. This binding leads to activation of the transcription factors NF -kB and the signaling molecule p38 MAP kinase [39, 40]. The pebIA locus encodes a previously described protein, Cell Binding Factor 1 (CBF -1) that mediates adherence to HeLa cells. Disruption of this locus reduces adherence in vitro by 15 fold, and reduces colonization of mice intestines [41]. Flagella mediate adherence to intestinal epithelial cells in vitro. Aflagellated bacteria, due to mechanical removal by shearing or by mutation, have decreased adherence to INT-407 cells [42]. Invasion: Bacterial invasion into epithelial cells is proposed to contribute to the disease and pathology caused by C. jejuni infection [2]. C. jejuni has been shown to invade intestinal epithelial cells in vitro and in vivo [12, 35, 43, 44]. Studies have indicated a role for both microfilarnents and microtubules in the invasion process. Oelschleger and colleagues found that C. jejuni invasion into INT-407 cells is a microtubule-dependent endocytic process that results in uptake into endosoma] vacuoles. Intemalization requires de novo protein synthesis for the bacteria, but not for the host cell [35]. Hu and Kopecko also showed that microtubules played a role in invasion as depolymerization inhibited C. jejuni internalization into intestinal epithelial cells. Microtubule-based membrane extensions were the initial point of contact between C. jejuni and the host cell. Once internalized, the bacteria associated with microtubules and then migrated to the perinuclear area via association with dynein [45]. Biswas and colleagues analyzed internalization of several human and animal isolates of C. jejuni and found that depolymerization of both microfilaments and microtubules inhibited invasion into INT- 407 cells depending on the strain tested [46]. In addition to host factors involved in invasion, bacteria] factors involved in invasion have been identified. C. jejuni produces proteins that are essential for internalization. Campylobacter invasion antigen (Cia) proteins are secreted by C. jejuni in response to physiologic stimuli, including serum, bile salts, extracellular matrix components, and contact with intestinal epithelial cells in vitro [47, 48]. Eight different proteins have been detected in culture supematants. One of them, designated CiaB, is a 73kDa protein that is necessary for internalization of bacteria into TNT-407 cells. In addition, CiaB itself is translocated into the host cell upon binding of the bacteria [47]. C. jejuni has been shown to encode virulence determinants on extrachromosomal elements. Four virulence genes have been detected on a 35 kb plasmid, designated pVir, found in some clinical isolates of C. jejuni. All four genes are homologous to type IV secretion system components. Three genes, designated comBI, comBZ, and comB3, display significant identity to Helicobacter pylori genes involved in competence for natural transformation and DNA uptake. The fourth gene displays identity to a type IV secretion system component of Agrobacterium designated virBI 1. Mutation of comB3 reduces adherence and invasion of INT-407 cells, and decreases frequency of natural transformation. Mutation of virBI I also reduced adherence and invasion in vitro and reduced virulence in the ferret diarrhea] disease model [49]. Intracellular survival: Intracellular survival of C. jejuni has been demonstrated in intestinal epithelial cells and macrophages [35, 50, 51]. The bacteria can survive within macrophages for up to 7 days [50]. Two genes have been identified to be necessary for survival within cells. The gene katA encodes catalase, which confers resistance to hydrogen peroxide in vitro. Mutation of katA does not affect intracellular survival in HEp-2 cells, but it does eliminate recovery from peritoneal macrophages 72 hours after infection [52]. Superoxide dismutase serves to defend bacterial cells against oxidative damage by catalyzing the breakdown of superoxide radicals to hydrogen peroxide and dioxygen. In C. jejuni, a single gene, designated sodB, encodes this enzyme and has been shown to be required for survival of invasion into INT-407 cells. Mutation of sodB decreases intracellular survival 12 fold [51]. Lipopolysaccharide: Lipopolysaccharide (LPS) is a constituent of the outer membrane of most gram negative bacteria that is necessary for membrane integrity and function. It is composed of 3 components: the lipid A molecule that is embedded in the membrane, the Oligosaccharide core, and the 0 chain. The lipid A molecule contains the endotoxic activity of LPS; the core oligosaccharide is involved in immunomodulation, and the 0 chain is highly variable and confers antigenicity to the molecule [53]. C. jejuni strains produce 2 types of LPS, one that contains all the components and one that lacks the 0 chain and is Similar to the lipooligosaccharides (LOS) of Neisseria spp. and Haemophilus spp. LOS [53,54]. 10 LPS and LOS function in C. jejuni virulence in vitro and in vivo. Carbohydrate moieties of LPS mediate adherence to INT-407 and intestinal mucous in vitro [42]. GalE is a UDP-glucose epimerase that catalyzes the interconversion between UDP-glucose and UDPgalactose, which is a component of C. jejuni LPS. Mutation of galE inhibits adherence and invasion of INT-407 cells and natural transformation efficiency [55]. Sialic acid moieties of C. jejuni LOS confer resistance to complement killing in vitro. Mutation of neuC, a gene involved in the biosynthesis of the Sialic acid N- acetylneuraminic acid (NeuNAc), results in loss of NeuNAc and increased serum sensitivity of C. jejuni [56]. Carbohydrate moieties of LOS can also mimic human gangliosides and play a role in the development of autoimmune disorders. The gene cgtA encodes a N-Acetylgalactosaminyl transferase that is subject to phase variable expression due to slip-strand mispairing in a homopolymeric G tract during DNA replication. This gene plays a role in structural variation in the LOS of C. jejuni 81-176, which can rrrimic GM; or GM3 gangliosides [57]. C. jejuni NCTC 11168 also has phase variable LOS that mimics gangliosides GM; or GM; depending on expression of the wlaN gene. This gene encodes a [3-1,3 galactosyltransferase and is also subject to slip-strand mispairing phase variation , similar to cgtA [58]. In vivo, antibodies directed against C. jejuni LOS molecules that mimic gangliosides can cause autoimmune mediated neuropathy. Rabbits immunized with C. jejuni LOS develop nerve degeneration and paralysis which resembles the clinical signs of GBS in humans [59]. Serum IgG from these rabbits reacts with both GM1 ganglioside and LOS in vitro, indicating a direct role for ganglioside mimicry by C. jejuni LOS in the development of autoimmune disease [59]. Taken 1] together, these studies have demonstrated several roles for LOS in the pathogenesis of C. jejuni infection. Toxins: Toxins have been identified from C. jejuni and are proposed to fimction in cellular destruction and clinical disease. An enterotoxin with immunologic similarity to cholera toxin and the heat labile toxin of E. coli has been described in C. jejuni. Enterotoxin causes elongation of Chinese hamster ovary (CHO) cells, increased intracellular cAMP, and fluid accumulation in a rat ligated intestinal loop model [60, 61]. However, contradictory reports of researchers unable to replicate these results have confounded this evidence [62, 63]. Cytotoxic activity has also been described by assessing rounding of VerO cells or detachment of cells from the growth plate [60, 64]. Cytotoxic activity has been detected in isolates from human patients with invasive diarrhea [64] and from animal isolates [60]. Additionally, cytotoxic activity was detected from poultry isolates tested on chicken lymphocytes using a chromium release assay [65]. Further characterization of enterotoxins and cytotoxins is necessary to elucidate their specific roles in clinical disease. Perhaps the most characterized C. jejuni toxin to date is the cytolethal distending toxin. Cytolethal distending toxin (CDT) causes distension and cell disintegration of a variety of cell types in vitro [66-68]. In C. jejuni, the CDT holotoxin is composed of 3 subunits, Cth, CdtB, and CdtC which are produced from adjacent genes. The individual subunit proteins are approximately 30kDa, 29kDa, and 21kDa, respectively. All three genes are required for toxic activity [69]. The Cth and CdtC subunits are responsible for binding to the host cell, while CdtB enters the host cell and exerts toxic effects [67]. CdtB has significant similarities to DNAse I enzymes and is believed to cause limited damage to DNA resulting in arrest of the cell cycle in the G2 phase [70]. The arrest is mediated by inhibition of CDC2, a kinase subunit required for entry into the M phase of the cell cycle. This arrest results in cell enlargement, nuclear disintegration, and ultimately cell death [68]. A few studies have suggested a function for CDT in induction of an immune response. Mutant C. jejuni lacking the cdtB gene are capable of colonizing NF-kB deficient mice and producing enteritis, but the disease is less severe than that seen from the wild type C. jejuni. In addition, serum IgG2a responses in mice infected with the mutant were lower than mice infected with the wild type strain [7]]. Attenuated disease and pathology with a CdtB mutant has also been demonstrated in a piglet model of infection (Mansfield et al, unpublished results). In vitro, CDT stimulates production of lL-8 from intestinal epithelial cells [72]. These data support the hypothesis that CDT plays a role in development of disease and stimulating an immune response in vivo. Immune responses Humoral immunity is necessary for clearance of C. jejuni from the intestine and resolution of clinical disease. In human infections, C. jejuni stimulates secretion of IgA in feces, urine, saliva, and mammary glands [73]. Flagellin is a strong stimulant for antibody production, as evidenced by the fact that a large proportion of anti-C. jejuni IgG antibodies found in convalescent serum is directed against flagellin [73]. The relatively low incidence Of disease in adults from endemic areas of developing countries is believed l3 to be due to strong humoral immunity stimulated by repeated exposure during childhood [16, 74, 75]. In a swine model of infection, lymphoglandular complexes have been discovered to be a source of IgA in C. jejuni infection. Twenty seven days after oral inoculation with a low dose of C. jejuni, B cell rich germinal centers develop in the LGCs that stain positively for IgA and C. jejuni [76]. These studies suggest that the adaptive immune response to C. jejuni is directed by Th 2 cytokines. Several studies have also demonstrated a role for cytokines in C. jejuni infections. Humans with infectious diarrhea caused by C. jejuni have increased levels Of nitric oxide gas and increased secretion of IL-1 B in the stool during the acute phase of infection [77]. Intestinal colonization of C. jejuni infected mice is significantly reduced when they are given recombinant IL-5 or IL-6 prior to infection. In addition, recombinant IL—6 enhances specific intestinal and systemic IgA production [78]. In vitro, secretion of IL-lB, IL-6, IL-8, and TNFO. is stimulated from intestinal epithelial cells after infection with C. jejuni [79] (Cunningham et al, unpublished results; Parthasarathy et al, unpublished results). In addition, intracellular levels of IL-4, IL-10, IFNy, and TNFOI are increased in C. jejuni infected intestinal epithelial cells [80]. Cultured human monocytes secrete IL-lB, IL-6, IL-8, and TNFOI upon infection, and NF-KB is translocated to the nucleus [81]. Similar to mammalian cells, chick kidney cells and avian macrophages have increased expression Of iNOS with increased release of nitric oxide, as well as increased expression of IL-1 [3, IL- 6, IL-8 [82]. These in vitro studies suggest an important role for proinflammatory cytokines in host resistance to C. jejuni infection. T rich uris suis T. suis life cycle T richuris suis is a nematode parasite that resides in the cecum and colon of pigs and it is closely related to T. muris, which infects mice, and T. trichuria which infects humans. Morphologically it has a slender anterior end that is partially embedded in the intestinal epithelium, and a thicker posterior end that is free in the lumen. Truchuris worms are commonly referred to as whipworrns due to their whip like appearance. There are 5 stages in the life cycle of T. suis from egg to mature adult. Non-infective barrel- shaped, tan eggs with bipolar plugs are shed from the feces of infected pigs. Infective L1 larvae develop, or embryonate, within the egg in the environment. In vitro studies have shown that eggs embryonate in as little as 19 days when cultured at 34°C. Embryonated eggs are ingested by the host and hatch throughout the small intestine, cecum, and colon. Hatched larvae have been detected in the distal small intestine as early as 9 hours after oral infection. The larvae burrow into the mucosa via the crypts of Lieberkilhn and invade the epithelial and goblet cells lining the crypts [83]. As the larvae grow they also invade cells in the lamina propria. The first moult, or shedding of the cuticle, begins approximately 10 days after infection as the larvae enter the L2 stage. The larvae continue to develop within the crypt cells and begin to migrate through the lamina propria towards the surface mucosa] epithelium [83, 84]. The second moult to the L3 stage begins 16 days after infection and the tail begins to protrude into the lumen of the colon[84]. Throughout the moults the larvae are increasing in size and development of the gastrointestinal and reproductive tracts can be observed. The third moult to the L4 stage 15 begins 20 days after infection. On day 32, larvae begin to undergo the last moult to the L5 or adult stage. Sexual maturation completes and fully formed eggs can be detected in the female uterus, and in the feces of the host, by 41 days, allowing the life cycle to commence again [83, 84]. Prevalence Recent studies have found a relatively low prevalence of T. suis in swine herds. T. suis was detected in 5.6% of pigs from intensive pig farms in Guangdong Province, China that do not use a strategic parasite control [85]. In Munsterland, Germany 8% of sows sampled from a 144 breeding farms were found to have T. suis ova in their feces. These farms routinely used anthelminthics in sows [86]. A study of Danish organic swine herds detected T. suis from 4% of weaners, 13% of fatteners, and <1% of sows [87]. Examination of a feral swine population in the US found that 10% of the pigs harbored T. suis [88]. Despite the apparently low prevalence of T. suis in swine herds, economic losses due to poor growth and death warrant further study of disease caused by these nematodes [89]. Mucohemorrhagic diarrhea Mucohemorrhagic diarrhea is a significant health problem in the swine industry that can result from infections with a variety of bacteria and parasitic worms. Treponema hyodysenteriae, Salmonella typhimurium, Salmonella typhisuis, T richuris suis, and Campylobacter spp. have all been found as causative agents. However, in many cases two or more agents act synergistically, causing significant enteric pathology [90, 9]]. l6 T richuris suis infection causes a disease commonly known as “21 day scours” due to the fact that diarrhea] disease commonly occurs approximately 3 weeks after pigs are placed in an area contaminated with infective T. suis eggs [90]. Diarrhea] disease associated with severe infection is profuse, reddish-brown, mucoid, and bloody. In addition to diarrhea, pigs become anorexic, anemic, and can have significant weight loss. Penetration of the nematode into the mucosa of the cecum and proximal colon causes inflammation, hemorrhage, and in severe cases necrosis [44, 89-91]. T. suis was once believed to be innocuous, but further study of these nematodes has revealed that they are an important pathogen in the swine industry. Histological lesions caused by T. suis infection are quite profound. Infection induces thickening of all layers of the intestine, sloughing of degenerate epithelial cells, and dilation of crypts with mucous. The lamina propria and submucosa become infiltrated by inflammatory cells, including neutrophils, lymphocytes, plasma cells and eosinophils [44, 89, 91]. In the distal colon, lymphoglandular complexes become enlarged by increased numbers of lymphocytes and macrophages, and several species of bacteria can be isolated from abscessed LGCs [44, 76, 89, 91]. Microscopic studies have shown that T. suis causes significant pathology both in the proximal colon where the worms reside and in the distal colon far from sites of worm attachment. Disease and pathology caused by T. suis infection is exacerbated by interaction with the microbial component of the swine colon. In 1975 Rutter and Beer demonstrated, through infections of conventionally reared, specific pathogen free and gnotobiotic pigs, that some component of the normal gut flora of pigs acted synergistically with T. suis to cause severe mucohemorrhagic enteritis [92]. Further studies by Mansfield and Urban confirmed Rutter and Beer’s findings, also showing that severe pathology caused by secondary bacterial infection was ameliorated by antibiotic treatment [91]. Additionally, fenbendazole treatment of both naturally and experimentally infected pigs removes all adult T. suis and reverses gross pathology in 7 days (Mansfield et al., unpublished results). These studies demonstrate the complex nature of the polymicrobial infection involving T. suis that results in severe disease. T. suis excretory/secretory product (ESP) The pathologic effects of T. suis infection are mediated both by direct interaction of the worm with host tissues and through the actions of secreted worm products. Adult T. suis produce an excretory/secretory product (ESP) composed of secreted products that function in invasion and feeding in vivo, and excreted products that are byproducts of its metabolism. Crude ESP can be collected from culture media of adult T. suis maintained in vitro after removal from infected pigs [93] and several components have been isolated and partially characterized. A 45kDa zinc metalloprotease was localized by immunocytochemica] staining to the stichosome. The protease displayed proteolytic activity against fibrinogen and elastin and is proposed to have a role in feeding and tissue penetration [93]. Phenol oxidase was localized to the reproductive tract of the female worms and caused brown pigmentation of the worms in an oxygenated environment. Phenol oxidase is proposed to function in tanning of the eggs [94, 95]. Thiol protease activity, localized to the gut of the worm, is hypothesized to function in digestion and absorption of nutrients [96]. Serine protease inhibitors were isolated from culture fluids and soluble worm extracts. These protease inhibitors showed activity against trypsin, chymotrypsin, and elastase, and may fimction in down-regulating host mast cell, and other immune cell, effector mechanisms [97, 98]. Recently, antimicrobial activity which inhibits the grth of C. jejuni, E. coli, and S. aureus in vitro in a dose-dependent manner has been described and may play a role in defending the worm against bacteria in the intestinal environment [99]. A 20kDa glycoprotein isolated from culture media was detected in a Western blot assay using serum from T. suis infected pigs. Serum from uninfected pigs, or pigs infected with the parasites 0esophagostomum dentatum, T richuris spiralis, Ascaris suum, or T oxoplasma gondii did not cross react, indicating that the 20kDa protein was specific for T. suis and could be used in diagnostic tests for infection [100]. In addition to identifying components of ESP, two recent studies have described its effects on host cells. Treatment of intestinal epithelial cells with crude ESP causes a dose dependent cytotoxic effect, and decreased transepithelial electrical resistance of differentiated cells. T. suis ESP treatment of swine intestinal epithelial cells prior to infection with C. jejuni decreases invasion, most likely due in part to its antibacterial activity [101]. Also, ESP has been shown to induce secretion of lL-6 and IL-10 from both differentiated and undifferentiated swine intestinal epithelial cells (Parthasarathy et al, unpublished results). These studies suggest a potential role for ESP in immunomodulation and the development of pathology in vivo. Immunity Preliminary studies have demonstrated that T. suis infection induces a strong immune response in conventionally reared pigs. Humoral immunity develops during 19 primary infection with the development of antibodies directed against a 20kDa glycoprotein isolated from in vitro culture fluids of adult worms. These T. suis specific antibodies were detected in serum from pigs 21 days after natural or experimental infection [100]. In addition, expression of IL-10 mRNA is increased 10 fold over uninfected controls in the mesenteric lymph nodes of pigs experimentally infected with T. suis. Pigs infected on a contaminated dirt lot have evidence of secondary bacterial infection in the distal colon and expression of both IL-10 and IL-12p40 are significantly increased in draining mesenteric lymph nodes compared to uninfected controls [102]. Pigs exposed to low level trickle infections with T. suis eggs over 4 weeks have decreased worm survival and worm fecundity during secondary challenge with 4000 infective eggs [103]. Taken together, these data suggest that T. suis stimulates a Th 2 directed immune response and that protective immunity can be achieved through low level exposure. Additional research is necessary to further characterize the T. suis stimulated immune response. Studies in humans have also described a Th 2 directed immune response to the human whipworm, T. trichuria. People that live in areas endemic for whipworm infection have antigen specific serum antibodies that are predominantly of the IgG] and IgG4 isotypes, with lower levels of IgG2 and IgG3. Antigen specific IgA and IgE are also present, with higher levels of IgE in less heavily infected individuals [104-106]. Whole blood cultures stimulated with T. trichuris ESP antigens or whole worm extracts secrete lL-4, IL-9, IL-10, IL-13, TNFOI, and to a lesser extent IFNy [104, 106]. Additionally, whole blood cultures stimulated with whole worm extracts display a significant proliferative response [106]. Similar to the swine whipworm, only a few studies have 20 been conducted to characterized specific components of the immune response to this nematode and further research is necessary. In contrast to studies in swine and humans, extensive research in murine models has characterized the roles of specific mediators in the immune response to Trichuris infection. Inbred mouse strains have been developed that are either resistant or susceptible to chronic infection with T. muris [107-110]. Resistant strains like BALB/K, BALB/c and C57BL/6 mount a Th 2-directed immune response with increased production of IL-4, IL-5, IL-9, IL-10, IL-13, Trichuris specific IgG], and increased serum levels of IgE [107, 109, 111, 112]. These strains develop significantly greater tissue eosinophilia upon infection than susceptible mice and are able to expel the larval stages of T. muris between 14 and 21 days after infection [107, 112]. Susceptible mouse strains like AKR and B10.BR mount a Th 1 directed immune response with increased production of IFNy and high levels of T richuris specific IgG2a as well as IgGl. These strains are unable to expel the larval stages of T. muris and become chronically infected [107, 111, 112]. These dichotomous strains have allowed researchers to define the roles of specific immune mediators in the disease process. Alteration of the polarized immune responses Of inbred mouse strains to T. muris infection through administration of stimulatory antigens or recombinant cytokines have elucidated the roles of specific cytokines in the immune response. Inoculation of susceptible AKR mice with Schistosoma mansoni egg antigens prior to infection with T. muris results in production of IL-4, IL-5, IL-10, IgG] and IgE and subsequent worm expulsion [113]. Administration of recombinant IL-4 to AKR mice facilitates parasite expulsion while blocking IL-4 activity in BALB/K mice with antibodies inhibits parasite 2] expulsion [114]. Exogenous recombinant IL-9 causes increased intestinal mastocytosis, increased serum IgE, and decreased worm burden in T. suis infected AKR mice [109]. In addition, neutralization of IL-9 with antibodies decreases colonic muscle hypercontractility and inhibits worm expulsion in normally resistant C57BL/6 mice [110]. Exogenous recombinant IL-12 increases production of parasite specific IgG2a and impaired worm expulsion in BALB/K mice [115]. Antibody depletion of IFNy activity in normally susceptible AKR mice infected with T. muris results in parasite expulsion [114]. In addition to using exogenous agents, genetic manipulation of inbred mice to abrogate production of certain cytokines has also proved to be a useful tool for studying the effects of Th 2 cytokines in T. muris infection. IL-lO knockout mice are susceptible to chronic infection with T. muris and have decreased IL-5 and lL-13 production, decreased peripheral eosinophilia, and decreased intestinal mucous production [112]. IL-4 knockout mice infected with T. muris have decreased production of IL-5, IL-9, and IL-13 compared to wild type mice and are unable to generate parasite specific IgGl. IL-13 knockout mice also have impaired cytokine production initially, but by 21 days after infection, IL-4, IL—5, and IL-9 production is at or above the levels produced by infected wild type mice. This demonstrates an early role for IL-13 in promoting the development of Th 2 responses stimulated by T. muris infection [116]. In addition to the roles of specific cytokines and antigen specific antibody in T. muris infection, cell mediated immunity is also important. SCID mice are normally susceptible to chronic T. muris infection, but reconstitution with purified CD4+ T cells from T. muris infected BALB/c mice resulted in expulsion of the parasite in the absence of antigen specific serum antibodies [108]. These studies have demonstrated the critical roles of the Th 2 cytokines 22 IL-4, IL-5, IL-9, IL-10, and IL-13 in resolution of T. muris infection and helped elucidate their specific effects in vivo. 23 Rationale for study Campylobacter jejuni is a significant enteric pathogen in humans causing severe diarrhea] disease and in rare cases debilitating autoimmune disorders [2, 10, 11, 13]. Severe diarrhea] disease and colonic pathology in humans is due to invasion of the intestine by C. jejuni with subsequent inflammation [2, 10]. Because pigs can become colonized with C. jejuni within 24 hours after birth and can carry the bacteria without clinical signs of disease, they serve as a potential source of infection for humans [15, 44]. T. suis infection of pigs harboring C. jejuni, either by natural or experimental infection, results in mucohemorrhagic diarrhea and severe pathology that mimics the clinical disease of Campylobacteriosis in humans [44, 91]. When T. suis is present concurrently, C. jejuni invade epithelial cells, significant inflammation develops, and mucohemorrhagic diarrhea is evident clinically [44]. This suggests that in both human and swine infections, bacterial invasion is the key event in the development of clinical disease and pathology. In the case of dual infection in swine, it is believed that immunomodulation induced by T. suis abrogates the natural resistance to C. jejuni, allowing invasion of intestinal tissues. Studies performed to date to elucidate the in vivo immune responses associated with C. jejuni infection in humans and swine have focused mainly on immunoglobulin production in serum and in lymphoid tissues [73, 76]. In vitro studies have focused mainly on cytokine responses from cultured cells ([72, 79]; Parthasarathy et al, unpublished results; Cunningham et al, unpublished results). The results indicate that intestinal epithelial cells are an important source of proinflammatory cytokines which may function in host resistance in vivo. Indeed, studies have shown a critical role for 24 epithelial cell derived IL-8 in control of experimental Shigellaflexneri infection in rabbits [117]. Characterization of the swine immune response to T. suis is severely limited and has focused on production of a relatively limited panel of cytokines in draining lymph nodes and secreted in the feces ([102]; Parthasarathy et al, unpublished results; Cunningham et al, unpublished results). Comparison of cytokine expression between intestinal tissue at the site of infection and the draining lymph nodes in animal models of nematode infection have shown that the cytokine profiles are not identical [112, 118]. T. suis infection results in significant pathology in the proximal colon due to direct interaction of the worms with the intestinal mucosa, and in the distal colon due to secreted worm products or immunomodulation [44, 91, 92]. Therefore, it is important to study local cytokine responses in the region where the worms reside and also in distant areas where worms are not present to determine the nature of the local immune response and ascertain what effect those cytokines may have on the development of pathology. In this dissertation, we have begun to characterize the swine local immune responses to C. jejuni and T. suis in an effort to expand our knowledge of the immune response to T. suis and to determine the nature of the immunomodulation that occurs during dual infection. Hypothesis The central hypothesis for this dissertation project is that a proinflammatory response plays a role in swine natural resistance to C. jejuni. A related hypothesis is that a T. suis stimulated anti-inflammatory response alters the protective proinflammatory 25 response. Thus T. suis infection would facilitate C. jejuni invasion in the colon and severe pathology would develop. Short term goals The short term goals are to address the following questions: 1. Does C. jejuni that expresses Gfp colonize the distal colons of pigs when inoculated rectally? Does rectally inoculated C. jejuni that expresses Gfp stimulate an immune response in the distal colon? What is the nature of the cytokine response in the distal colon stimulated by rectally inoculated C. jejuni? What is the nature of the local cytokine response in the small and large intestines Of pigs infected orally with C. jejuni, T. suis, or both? Is there evidence of immunomodulation in the colons of dually infected pigs? Do epithelial cell derived cytokines play a role in swine resistance to C. jejuni infection as determined by in vitro infection of swine intestinal epithelial cells? In summary, the goal of this study was to determine the nature of the local cytokine response to C. jejuni and T. suis and to determine whether cytokine dysregulation could play a role in the pathology and disease caused by dual infection. Chapter 2 summarizes the studies on the effects of rectal inoculation with C. jejuni on colonization, localization of the bacteria in the distal colon, and on local cytokine mRNA expression. Proinflammatory cytokines are upregulated in the intestines 26 of animals experimentally or naturally infected with enteric bacteria and they have been shown to be critical for elimination of the bacteria and resolution of disease [117, 119- 121]. We measured mRNA expression of both proinflammatory and anti-inflammatory in the distal colons and LGCS of pigs rectally inoculated with C. jejuni. The nature of the cytokine response to C. jejuni in lymphoid and nonlymphoid tissue in the distal colon is discussed. Chapter 3 summarizes the study on the effect of oral infection with C. jejuni, T. suis, or both on local cytokine expression in the intestines of pigs. Cytokine responses in resistant mice infected with T. muris are predominantly Th 2 directed with increased production of IL-4, IL-5, IL-10, and IL-13 [107, 109, 111, 112]. These cytokines can also be characterized as anti-inflammatory as they downregulate proinflammatory cytokines [122, 123]. We measured mRNA expression of both proinflammatory and anti- inflammatory cytokines in the jejunum, proximal colon, and distal colons of pigs orally inoculated with C. jejuni, T. suis, or both. The nature of the cytokine responses and the differences in responses between the intestinal segments are discussed. Chapter 4 summarizes the in vitro studies on proinflammatory cytokine production from intestinal epithelial cells infected with C. jejuni. C. jejuni stimulates production of IL-IB, IL-6, IL-8, and TNFOL from both swine and human intestinal epithelial cells (Cunningham et al, unpublished results, Parthasarathy et al, unpublished results; [72, 79, 80]. Intestinal epithelial cell derived IL-8 is critical for control of infection in a rabbit ligated ileal loop model of Shigella flexneri infection [117]. We measured secreted lL-18 protein and IL-18 mRNA expression from C. jejuni infected IPEC-l swine intestinal epithelial cells. The nature of the responses from differentiated 27 and undifferentiated IPEC-l cells and the role of transcriptional regulation of IL-18 production are discussed. A proposed model for epithelial cell derived IL-1 8 in resistance to C. jejuni infection in vivo is also discussed. Chapter 5 summarizes the previous chapters and discusses a proposed model for disease and pathology during dual infection with C. jejuni and T. suis due to cytokine dysregulation. 28 10. ll. l2. l3. 14. 15. 16. References Altekruse, S.F., D.L. Swerdlow, and NJ. Stern, Microbial food borne pathogens. Campylobacter jejuni. Vet Clin North Am Food Anim Pract, 1998. 14(1): p. 31-40. Ketley, J.M., Pathogenesis of enteric infection by Campylobacter. Microbiology, 1997. 143 (Pt 1): p. 5-21. Wiesner, R.S., D.R. Hendrixson, and VJ. DiRita, Natural transformation of C ampylobacter jejuni requires components of a type II secretion system. J Bacteriol, 2003. 185(18): p. 5408-18. Wilson, D.L., et al., Variation of the natural transformation frequency of Campylobacter jejuni in liquid shake culture. Microbiology, 2003. 149(Pt 12): p. 3603-15. F ermer, C. and ED. Engvall, Specific PCR identification and diflerentiation of the thermophilic campylobacters, Campylobacterjejuni, C. coli, C. lari, and C. upsaliensis. J Clin Microbiol, 1999. 37(10): p. 3370-3. Moore, J .E., et al., Molecular diversity of Campylobacter coli and C. jejuni isolated from pigs at slaughter by flaA-RF LP analysis and ribotyping. J Vet Med B Infect Dis Vet Public Health, 2002. 49(8): p. 388-93. Cardarelli-Leite, P., et al., Rapid identification of Campylobacter species by restriction fragment length polymorphism analysis of a PCR-amplified fragment of the gene coding for 1 6S rRNA. J Clin Microbiol, 1996. 34(1): p. 62-7. Vacher, S., et al., PCR-restrictionfiagment length polymorphism analysis for detection of point mutations associated with macrolide resistance in Campylobacter spp. Antimicrob Agents Chemother, 2003. 47(3): p. 1125-8. Wassenaar, T.M. and DC. Newell, Genotyping of Campylobacter spp. Appl Environ Microbiol, 2000. 66(1): p. 1-9. Wassenaar, T.M. and MJ. Blaser, Pathophysiology of Campylobacter jejuni infections of humans. Microbes Infect, 1999. 1(12): p. 1023-33. Allos, B.M., Campylobacterjejuni Infections: update on emerging issues and trends. Clin Infect Dis, 2001. 32(8): p. 1201-6. Babakhani, F .K., G.A. Bradley, and LA. Joens, Newborn piglet model for campylobacteriosis. Infect Immun, 1993. 61(8): p. 3466-75. Goodyear, C.S., et al., Monoclonal antibodies raised against Guillain-Barre syndrome-associated C ampylobacter jejuni lipopolysaccharides react with neuronal gangliosides and paralyze muscle- nerve preparations. J Clin Invest, 1999. 104(6): p. 697-708. Altekruse, S.F., et al., Food and animal sources of human Campylobacterjejuni infection. J Am Vet Med Assoc, 1994. 204(1): p. 57-61. Young, C.R., et al., Enteric colonisation following natural exposure to C ampylobacter in pigs. Res Vet Sci, 2000. 68(1): p. 75-8. Coker, A.O., et al., Human campylobacteriosis in developing countries. Emerg Infect Dis, 2002. 8(3): p. 237-44. 29 17. l8. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. R30, M.R., et al., Pathogenicity and convalescent excretion ofCampylobacter in rural Egyptian children. Am J Epidemiol, 2001. 154(2): p. 166-73. Albert, M.J., et al., Case-control stuay of enteropathogens associated with childhood diarrhea in Dhaka, Bangladesh. .1 Clin Microbiol, 1999. 37(11): p. 3458-64. Bichile, L.S., et al., Acute Campylobacterjejuni enteritis in 385 hospitalised patients. J Assoc Physicians India, 1992. 40(3): p. 164-6. Adegbola, R.A., et al., Correlation between human and animal bio-serogroups of Campylobacter isolates in Nigeria. J TrOp Med Hyg, 1990. 93(4): p. 280-3. Coker, A.O., et al., Zoonotic infections in Nigeria: overview from a medical perspective. Acta Trop, 2000. 76(1): p. 59-63. Wassenaar, T.M., N.M. Bleumink-Pluym, and BA. van der Zeijst, Inactivation of Campylobacter jejuniflagellin genes by homologous recombination demonstrates that flaA but not flaB is required for invasion. Embo J, 1991. 10(8): p. 2055-61. Yao, R., et al., Isolation of motile and non-motile insertional mutants of Campylobacter jejuni: the role of motility in adherence and invasion of eukaryotic cells. Mol Microbiol, 1994. 14(5): p. 883- 93. Nachamkin, I., X.H. Yang, and NJ. Stern, Role of Campylobacter jejuni flagella as colonization factors for three-day-old chicks: analysis with flagellar mutants. Appl Environ Microbiol, 1993. 59(5): p. 1269-73. Szymanski, C.M., et al., Campylobacterjejuni motility and invasion of C ac0-2 cells. Infect Immun, 1995. 63(11): p. 4295-300. Field, L.H., et al., Influence of iron on growth, morphology, outer membrane protein composition, and synthesis of siderophores in Campylobacterjejuni. Infect Immun, 1986. 54(1): p. 126-32. lshikawa, T., et al., The iron-binding protein Dps confers hydrogen peroxide stress resistance to Campylobacterjejuni. J Bacteriol, 2003. 185(3): p. 1010-7. Baillon, M.L., et al., An iron-regulated alkyl hydroperoxide reductase (A hpC) confers aerotolerance and oxidative stress resistance to the microaerophilic pathogen Campylobacter jejuni. J Bacteriol, 1999. 181(16): p. 4798-804. van Vliet, A.H., et al., Campylobacterjejuni contains two fur homologs: characterization of iron- responsive regulation of peroxide stress defense genes by the PerR repressor. J Bacteriol, 1999. 181(20): p. 6371-6. Beery, J.T., M.B. Hugdahl, and M.P. Doyle, Colonization of gastrointestinal tracts of chicks by Campylobacterjejuni. Appl Environ Microbiol, 1988. 54(10): p. 2365-70. Lee, A., et al., Mucus colonization as a determinant of pathogenicity in intestinal infection by Campylobacterjejuni: a mouse cecal model. Infect Immun, 1986. 51(2): p. 536-46. Takata, T., S. F uj imoto, and K. Amako, Isolation of nonchemotactic mutants of Campylobacter jejuni and their colonization of the mouse intestinal tract. Infect Immun, 1992. 60(9): p. 3596-600. Hugdahl, M.B., J.T. Beery, and M.P. Doyle, Chemotactic behavior of Campylobacter jejuni. Infect Immun, 1988. 56(6): p. 1560-6. 30 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. Allen, KJ. and M.W. Griffiths, Eflect of environmental and chemotactic stimuli on the activity of the Campylobacterjejuni/lax! sigma(28) promoter. FEMS Microbiol Lett, 2001. 205(1): p. 43-8. Oelschlaeger, T.A., P. Guerry, and DJ. Kopecko, Unusual microtubule-dependent endocytosis mechanisms triggered by C ampylobacter jejuni and Citrobacterfi'eundii. Proc Natl Acad Sci U S A, 1993. 90(14): p. 6884-8. Monteville, M.R., J.E. Yoon, and ME. Konkel, Maximal adherence and invasion of [NT 407 cells by Campylobacterjejuni requires the CadF outer-membrane protein and microfilament reorganization. Microbiology, 2003. 149(Pt 1): p. 153-65. Konkel, M.B., et al., Identification and molecular cloning of a gene encoding a fibronectin- binding protein (CadF)fi'om Campylobacterjejuni. Mol Microbiol, 1997. 24(5): p. 953-63. Ziprin, R.L., et al., The absence of cecal colonization of chicks by a mutant of Campylobacter jejuni not expressing bacterial fibronectin-binding protein. Avian Dis, 1999. 43(3): p. 586-9. J in, 8., et al., leA, a novel surface-exposed lipoprotein specific to Campylobacterjejuni, mediates adherence to host epithelial cells. Mol Microbiol, 2001. 39(5): p. 1225-36. J in, S., et al., leA of Campylobacter jejuni interacts with surface-exposed heat shock protein 90alpha and triggers signalling pathways leading to the activation of NF -kappaB and p38 MAP kinase in epithelial cells. Cell Microbiol, 2003. 5(3): p. 165-74. Pei, 2., et al., Mutation in the peb IA locus of Campylobacter jejuni reduces interactions with epithelial cells and intestinal colonization of mice. Infect Immun, 1998. 66(3): p. 938-43. McSweegan, E. and RI. Walker, Identification and characterization of two Campylobacterjejuni adhesins for cellular and mucous substrates. Infect Immun, 1986. 53(1): p. 141-8. Babakhani, F .K. and LA. Joens, Primary swine intestinal cells as a model for stuaying Campylobacterjejuni invasiveness. Infect Immun, 1993. 61(6): p. 2723-6. Mansfield, L.S., et al., Enhancement of disease and pathology by synergy of Trichuris suis and Campylobacterjejuni in the colon of immunologically naive swine. Am J Trop Med Hyg, 2003. 68(3): p. 70-80. Hu, L. and DJ. Kopecko, Campylobacterjejuni 81-176 associates with microtubules and aynein during invasion of human intestinal cells. Infect Immun, 1999. 67(8): p. 4171-82. Biswas, D., K. Itoh, and C. Sasakawa, Uptake pathways of clinical and healthy animal isolates of Campylobacterjejuni into INT-407 cells. FEMS Immunol Med Microbiol, 2000. 29(3): p. 203-1]. Konkel, M.E., eta1., Bacterial secreted proteins are required for the internaliztion of Campylobacterjejuni into cultured mammalian cells. Mol Microbiol, 1999. 32(4): p. 691-701. Rivera-Amill, V., et al., Secretion of the virulence-associated C ampylobacter invasion antigens fi'om Campylobacterjejuni requires a stimulatory signal. J Infect Dis, 2001. 183(11): p. 1607-16. Bacon, DJ ., et al., Involvement of a plasmid in virulence of Campylobacter jejuni 81-176. Infect Immun, 2000. 68(8): p. 4384-90. Kiehlbauch, J.A., et al., Phagocytosis of Campylobacter jejuni and its intracellular survival in mononuclear phagocytes. Infect Immun, 1985. 48(2): p. 446-5]. 31 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. Pesci, E.C., D.L. Cottle, and CL. Pickett, Genetic, enzymatic, and pathogenic studies of the iron superoxide dismutase of Campylobacter jejuni. Infect Immun, 1994. 62(7): p. 2687-94. Day, W.A., Jr., et al., Role of catalase in Campylobacterjejuni intracellular survival. Infect Immun, 2000. 68(11): p. 6337-45. Moran, A.P., Structure and conserved characteristics of Campylobacter jejuni lipopolysaccharides. J Infect Dis, 1997. 176 Supp12: p. 8115-21. Aspinall, G.O., eta1., Serological diversity and chemical structures of Campylobacter jejuni low- molecular-weight lipopolysaccharides. J Bacteriol, 1992. 174(4): p. 1324-32. Fry, B.N., et al., The galE gene of Campylobacter jejuni is involved in lipopolysaccharide synthesis and virulence. Infect Immun, 2000. 68(5): p. 2594-601. Guerry, P., eta1., Sialylation of lipooligosaccharide cores affects immunogenicity and serum resistance of Campylobacter jejuni. Infect Immun, 2000. 68(12): p. 6656-62. Guerry, P., et al., Phase variation of C ampylobacter jejuni 81-176 lipooligosaccharide aflects ganglioside mimicry and invasiveness in vitro. Infect Immun, 2002. 70(2): p. 787-93. Linton, D., et al., Phase variation of a beta-1,3 galactosyltransferase involved in generation of the ganglioside GMI-like lipo-oligosaccharide of C ampylobacter jejuni. Mol Microbiol, 2000. 37(3): p. 501-14. Yuki, N., et al., Carbohydrate mimicry between human ganglioside GM] and Campylobacter jejuni lipooligosaccharide causes Guillain-Barre syndrome. Proc Natl Acad Sci U S A, 2004. 101(31): p. 11404-9. Johnson, W.M. and H. Lior, Cytotoxic and cytotonic factors produced by Campylobacterjejuni, Campylobacter coli, and Campylobacter laridis. J Clin Microbiol, 1986. 24(2): p. 275-8]. Klipstein, F.A. and RF. Engert, Properties of crude Campylobacterjejuni heat-labile enterotoxin. Infect Immun, 1984. 45(2): p. 314-9. Collins, S., et al., A cytotonic, cholera toxin-like protein produced by C ampylobacter jejuni. Comp Biochem Physiol B, 1992. 103(2): p. 299-303. Wassenaar, T.M., Toxin production by Campylobacter spp. Clin Microbiol Rev, 1997. 10(3): p. 466-76. Klipstcin, F.A., et al., Pathogenic properties of C ampylobacter jejuni: assay and correlation with clinical manifestations. Infect Immun, 1985. 50(1): p. 43-9. Lam, K.M., A cytotoxicity test for the detection of Campylobacter jejuni toxin. Vet Microbiol, 1993. 35(1-2): p. 133-9. Aragon, V., K. Chao, and LA. Dreyfus, Eflect of cytolethal distending toxin on F -actin assembly and cell division in Chinese hamster ovary cells. Infect Immun, 1997. 65(9): p. 3774-80. Lee, RB, et al., Interactions of Campylobacter jejuni cytolethal distending toxin subunits C th and CdtC with HeLa cells. Infect Immun, 2003. 71(9): p. 4883-90. Whitehouse, C.A., et al., Campylobacterjejuni cytolethal distending toxin causes a GZ-phase cell cycle block Infect Immun, 1998. 66(5): p. 1934-40. 32 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. Pickett, C.L., et al., Prevalence of cytolethal distending toxin production in Campylobacterjejuni and relatedness of Campylobacter sp. cdtB gene. Infect Immun, 1996. 64(6): p. 2070-8. Lara-Tejero, M. and J .E. Galan, A bacterial toxin that controls cell cycle progression as a deoxyribonuclease I-like protein. Science, 2000. 290(5490): p. 354-7. Fox, J.G., et al., Gastroenteritis in NF -kappaB-deficient mice is produced with wild-type Camplyobacterjejuni but not with C. jejuni lacking cytolethal distending toxin despite persistent colonization with both strains. Infect Immun, 2004. 72(2): p. 1116-25. Hickey, T.E., et al., Campylobacterjejuni cytolethal distending toxin mediates release of interleukin-8 from intestinal epithelial cells. Infect Immun, 2000. 68(12): p. 6535-41. Cawthraw, S.A., et al., Long-term antibody responses following human infection with Campylobacterjejuni. Clin Exp Immunol, 2002. 130(1): p. 101—6. Calva, J J ., et al., Cohort study of intestinal infection with campylobacter in Mexican children. Lancet, 1988. 1(8584): p. 503-6. Taylor, D.N., et al., Influence of strain characteristics and immunity on the epidemiology of Campylobacter infections in Thailand J Clin Microbiol, 1988. 26(5): p. 863-8. Mansfield, LS. and D.T. Gauthier, Lymphoglandular complexes are important colonic sites for immunoglobulin A induction against Campylobacterjejuni in a swine disease model. Comp Med, 2004. 54(5): p. 514-23. Enocksson, A., et al., Rectal nitric oxide gas and stool cytokine levels during the course of infectious gastroenteritis. Clin Diagn Lab Immunol, 2004. 11(2): p. 250-4. Baqar, S., N.D. Pacheco, and FM. Rollwagen, Modulation of mucosal immunity against Campylobacterjejuni by orally administered cytokines. Antimicrob Agents Chemother, 1993. 37(12): p. 2688-92. Hickey, T.E., eta1., Campylobacterjejuni-stimulated secretion of interleukin-8 by IN T407 cells. Infect Immun, 1999. 67(1): p. 88-93. Al-Salloom, F.S., et al., Campylobacter-stimulated IN T407 cells produce dissociated cytokine profiles. J Infect, 2003. 47(3): p. 217-24. Jones, M.A., et al., Induction of proinflammatory responses in the human monocytic cell line THP-l by Campylobacterjejuni. Infect Immun, 2003. 71(5): p. 2626-33. Smith, C.K., et al., Campylobacterjejuni-induced cytokine responses in avian cells. Infect Immun, 2005. 73(4): p. 2094-100. Beer, RJ., Studies on the biology of the life-cycle of Trichuris suis Schrank, I788. Parasitology, 1973. 67(3): p. 253-62. Beer, RJ., Morphological descriptions of the egg and larval stages of Trichuris suis Schrank, I788. Parasitology, 1973. 67(3): p. 263-78. Wong, Y.B., et al., Survey of intestinal parasites in pigs from intensive farms in Guangdong Province, People’s Republic of China. Vet Parasitol, 2005. 127(3-4): p. 333-6. 33 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. Gerwert, S., K. Failing, and C. Bauer, Husbandry management, worm control practices and gastro-intestinal parasite infections of sows in pig-breeding farms in Munsterland, Germany. Dtsch Tierarztl Wochenschr, 2004. 111(10): p. 398-403. Carstensen, L., M. Vaarst, and A. Roepstorff, Helminth infections in Danish organic swine herds. Vet Parasitol, 2002. 106(3): p. 253-64. Gipson, P.S., et al., Health status of a recently discovered population of feral swine in Kansas. J Wildl Dis, 1999. 35(3): p. 624-7. Hall, G.A., J .M. Rutter, and RJ. Beer, A comparative stuay of the histopathology of the large intestine of conventionally reared, specific pathogen free and gnotobiotic pigs infected with Trichuris suis. J Comp Pathol, 1976. 86(2): p. 285-92. Tubbs, R.C., Four causes of mucohemorrhagic diarrhea you should recognize. Veterinary Medicine, 1987. 82(1): p. 89-90. Mansfield, LS. and J .F . Urban, Jr., The pathogenesis of necrotic proliferative colitis in swine is linked to whipworm induced suppression of mucosal immunity to resident bacteria. Vet Immunol Immunopathol, 1996. 50(1-2): p. 1-17. Rutter, J .M. and RJ. Beer, Synergism between Trichuris suis and the microbial flora of the large intestine causing dysentery in pigs. Infect Immun, 1975. 11(2): p. 395-404. Hill, D.E., et al., Trichuris suis: a zinc metalloprotease from culture fluids of adult parasites. Exp Parasitol, 1993. 77(2): p. 170-8. Fetterer, RH. and DE. Hill, The occurrence of phenol oxidase activity in female Trichuris suis. J Parasitol, 1993. 79(2): p. 155-9. Fetterer, RH. and DE. Hill, Localization of phenol oxidase in female Trichuris suis. J Parasitol, 1994. 80(6): p. 952-9. Hill, DE. and J.A. Sakanari, Trichuris suis: thiol protease activity fiom adult worms. Exp Parasitol, 1997. 85(1): p. 55-62. Rhoads, M.L., R.H. Fetterer, and DE. Hill, Trichuris suis: A secretory serine protease inhibitor. Exp Parasitol, 2000. 94(1): p. 1-7. Rhoads, M.L., et al., Trichuris suis: a secretory chymotrypsin/elastase inhibitor with potential as an immunomodulator. Exp Parasitol, 2000. 95(1): p. 36-44. Abner, S.R., eta1., T richuris suis: detection of antibacterial activity in excretory-secretory products fiom adults. Exp Parasitol, 2001. 99(1): p. 26-36. Hill, D.E., R.D. Romanowski, and IF Urban, Jr., A Trichuris specific diagnostic antigen fiom culture fluids of Trichuris suis adult worms. Vet Parasitol, 1997. 68(1-2): p. 91-102. Abner, S.R., et al., Response of intestinal epithelial cells to Trichuris suis excretory-secretory products and the influence on C ampylobacter jejuni invasion under in vitro conditions. J Parasitol, 2002. 88(4): p. 738-45. Mansfield, L.S., et al., Construction of internal cDNA competitors for measuring IL-10 and IL-12 cytokine gene expression in swine. Vet Immunol Immunopathol, 1998. 65(1): p. 63-74. 34 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. 118. 119. Pedersen, S. and I. Saeed, Acquired immunity to Trichuris suis infection in pigs. Parasitology, 2001.123(Pt1):p.95-101. Faulkner, H., et al., A ge- and infection intensity-dependent cytokine and antiboay production in human trichuriasis: the importance of IgE. J Infect Dis, 2002. 185(5): p. 665-72. Lillywhite, J .E., et al., Humoral immune responses in human infection with the whipworm Trichuris trichiura. Parasite Immunol, 1991. 13(5): p. 491-507. Turner, 1., et al., A comparison of cellular and humoral immune responses to trichuroid derived antigens in human trichuriasis. Parasite Immunol, 2002. 24(2): p. 83-93. Else, K.J. and R.K. Grencis, Cellular immune responses to the murine nematode parasite Trichuris muris. l. Diflerential cytokine production during acute or chronic infection. Immunology, 1991. 72(4): p. 508-13. Else, KJ. and R.K. Grencis, Antibody-independent eflector mechanisms in resistance to the intestinal nematode parasite Trichuris muris. Infect Immun, 1996. 64(8): p. 2950-4. Faulkner, H., et al., Interleukin-9 enhances resistance to the intestinal nematode Trichuris muris. Infect Immun, 1998. 66(8): p. 3832-40. Khan, W.I., et al., Modulation of intestinal muscle contraction by interleukin-9 (IL-9) or IL-9 neutralization: correlation with worm expulsion in murine nematode infections. Infect Immun, 2003. 71(5): p. 2430-8. Else, K.J., L. Hultner, and R.K. Grencis, Cellular immune responses to the murine nematode parasite T richuris muris. II. Diflerential induction of TH-cell subsets in resistant versus susceptible mice. Immunology, 1992. 75(2): p. 232-7. Schopf, L.R., et al., IL-1 0 is critical for host resistance and survival during gastrointestinal helminth infection. J Immunol, 2002. 168(5): p. 2383-92. Curry, A.J., et al., Evidence that cytokine-mediated immune interactions induced by Schistosoma mansoni alter disease outcome in mice concurrently infected with Trichuris muris. J Exp Med, 1995. 181(2): p. 769-74. Else, K.J., et al., Cytokine-mediated regulation of chronic intestinal helminth infection. J Exp Med, 1994. 179(1): p. 347-5]. Bancroft, A.J., et al., Interleukin-12 promotes a chronic intestinal nematode infection. Eur J Immunol, 1997. 27(4): p. 866-70. Bancroft, A.J., A.N. McKenzie, and R.K. Grencis, A critical role for IL-13 in resistance to intestinal nematode infection. J Immunol, 1998. 160(7): p. 3453-61. Sansonetti, RJ., et al., Interleukin-8 controls bacterial transepithelial translocation at the cost of epithelial destruction in experimental shigellosis. Infect Immun, 1999. 67(3): p. 1471-80. Dawson, H.D., et al., Localized multigene expression patterns support an evolving Thl/ThZ-like paradigm in response to infections with Toxoplasma gondii and Ascaris suum. Infect Immun, 2005. 73(2): p. 1116-28. Cho, W.S. and C. Chae, Expression of inflammatory cytokines (T NF -alpha, IL-1, IL-6 and IL-8) in colon of pigs naturally infected with Salmonella typhimurium and S. choleraesuis. J Vet Med A Physio] Pathol Clin Med, 2003. 50(10): p. 484-7. 35 120. 12]. 122. 123. Elhofy, A. and K.L. Bost, Limited interleukin-18 response in Salmonella-infected murine macrophages and in Salmonella-infected mice. Infect Immun, 1999. 67(10): p. 5021-6. Foss, D.L., M.J. Zilliox, and M.P. Murtaugh, Bacterially induced activation of interleukin-1 8 in porcine intestinal mucosa. Vet Immunol Immunopathol, 2001. 78(3-4): p. 263-77. Lugering, N., eta1., Interleukin (IL)-I3 and IL-4 are potent inhibitors of IL-8 secretion by human intestinal epithelial cells. Dig Dis Sci, 1999. 44(3): p. 649-55. Muchamuel, T., et al., IL-l3 protects mice fi'om lipopolysaccharide-induced lethal endotoxemia: correlation with down-modulation of TNF-alpha, IF N-gamma, and IL-12 production. J Immunol, 1997. 158(6): p. 2898-903. 36 Chapter 2: “Campylobacter jejuni invades distal colonic lymphoglandular complexes causing upregulation of proinflammatory cytokines in a swine rectal challenge model” Abstract Campylobacter jejuni is the leading bacterial cause of foodborne illness worldwide. Orally inoculated pigs have been used to study diarrhea] disease, intestinal pathology, and humoral immune responses due to C. jejuni. These studies have shown that C. jejuni mainly targets the distal colon. We used a rectal inoculation technique to target the distal colon in order to assess pathogenesis of C. jejuni infection at specific time points. Weaned piglets were inoculated rectally with 109 cfu of C. jejuni expressing green fluorescent protein (Gfp), and were euthanized 1 or 24 hours after inoculation. Samples of the distal colon were taken for C. jejuni detection and localization, and cytokine analysis. C. jejuni was detected in 93.75% of inoculated pigs. Immunostaining detected organisms associated with surface epithelium, within crypts, and within crypts of lymphoglandular complexes in the distal colon. Several proinflammatory cytokines, including IL-8, IL-18, IL-IB, TNF-Ot, GM-CSF, iNOS, and MCP-l, were significantly upregulated in the distal colon and lymphoglandular complexes 1 hour after inoculation. These results show that rectal inoculation of C. jejuni is a usefiil technique for targeting the distal colon and eliciting cytokine responses. Proinflammatory cytokines are likely important in the pathogenesis of C. jejuni infections in pigs. 37 Introduction Campylobacterjejuni is the most frequently reported bacterial cause of foodborne illness worldwide. The CDC estimates 2.4 million cases of campylobacteriosis occur each year in the US. [1]. Children and young adults are most commonly affected, developing profuse, watery diarrhea with blood, mucus, and leukocytes 1 to 7 days afier ingestion of the organism [2, 3] . In most cases symptoms persist for 5 to 8 days and include abdominal cramping, fever, and malaise [3]. In rare cases death occurs, most commonly in infants, elderly, and immunosuppressed individuals [2]. Most Campylobacter infections in humans result in self-limiting gastrointestinal disease. However, extra intestinal infections can result in local complications including cholecystitis, pancreatitis, peritonitis, and massive GI hemorrhage. Further spread can result in meningitis, endocarditis, septic arthritis, osteomyelitis, and neonatal sepsis [1]. Notable sequelae of C. jejuni infection include Reiters syndrome and Guillain-Barré syndrome (GBS) [1-4] . Reiters syndrome occurs in 1% of people affected with campylobacteriosis. Symptoms commonly arise 7 to 10 days after onset of diarrheal illness and include a sterile, reactive arthritis, urethritis, and conjunctivitis [2]. Guillain- Barré syndrome, an ascending peripheral neuropathy, is an autoimmune disorder that develops in 1 in every 1000 cases of C. jejuni infection and is most commonly associated with Penner serotype 0:19 strains [1-4]. Food sources that have been associated with infection include raw milk, poultry products, shellfish, and fresh produce [2]. Live animals are a significant reservoir for C. jejuni and a source of infection for humans as C. jejuni is a commensal in the gastrointestinal tract of a variety of wild and domestic animal species including poultry, cattle, sheep, and swine [5-8]. Campylobacteriosis has 38 incited an abundance of research aimed at elucidating virulence mechanisms and immune responses in an effort to develop therapies to treat and prevent infection with this pathogen. Immune responses to C. jejuni have been characterized in both human infections and animal models. C. jejuni stimulates a humoral immune response in humans with production of specific IgA in feces, urine, saliva, and mammary glands [9]. Flagellin is an important antigen for stimulating immune responses, as serum taken from patients up to 2 months post infection contained a large proportion of antibodies directed against flagellin [9]. Humoral immunity affords protection against reinfection with homologous strains [10], which is believed to account for the relatively low incidence of diarrhea] disease in adults in developing countries compared to developed countries [11, 12]. Recently, Mansfield and Gauthier demonstrated that lymphoglandular complexes (LGCS) in the distal colons of pigs are targeted by C. jejuni. The LGCS expand significantly after infection due to development of B cell rich germinal centers that produce IgA [13]. Cytokines also play an important role in C. jejuni infections. Mice orally administered recombinant IL-5 or IL-6 have reduced intestinal colonization after infection. In mice given IL-6, specific intestinal and systemic IgA responses are also enhanced [14]. In vitro studies on human cells show that C. jejuni induces IL-8 secretion and NF-KB translocation in intestinal epithelial cells [15-17]. C. jejuni infected monocytes secrete IL-IB, IL-6, IL-8, and TNF-OI, and NF-KB is translocated to the nucleus [18]. Several animal models have emerged to aid in the study of various aspects of C. jejuni infection. Inbred mice have been used to study leukocyte responses, cytokine 39 responses, immunoglobulin responses, and colonization [19-22]. Ferrets have been used to study diarrhea] disease, abortion, colonization, and antibody responses to C. jejuni infection [23]. Pigs have been used to study diarrhea] disease, pathology, and immune responses [13, 24, 25]. Rectal immunization protocols to study immune responses and vaccine efficacy have shown that direct inoculation of infectious agents into the distal colon is an effective, non-invasive method for eliciting and studying local immune responses [26, 27]. In this study we used rectal inoculation of C. jejuni to study the early immunological response to initial invasion events in the distal colon. Our results indicate that rectal inoculation of C. jejuni elicits measurable changes in cytokine expression in the distal colon and lymphoglandular complexes (LGCS). Expression of several proinflammatory cytokines was significantly increased in the LGCS, which have previously been shown to function as antigen sampling structures that are specifically targeted by C. jejuni [13, 25]. 40 Materials and Methods Animals Four week old male and female outbred Landrace-Yorkshire cross pigs were obtained from the Michigan State University (MSU) swine farm. Animals were housed in the MSU Research containment facility in groups of 4 for social support. Individual infection groups were housed in separate rooms to eliminate cross contamination. All animal housing and handling complied with MSU animal use guidelines and National Institutes of Health guidelines for humane use of laboratory animals. Bacteria C. jejuni strain 81-176 which had been electroporated with the plasmid pWM1007 [28] was used for all experiments. The plasmid pWM1007 contains the gin gene under the influence of a constitutively active consensus Campylobacter promoter. Bacteria were grown, from frozen stocks, on Bolton (Oxoid Inc., Basingstoke, Hampshire, England) agar plates without antibiotics at 37°C, 5% C02 in humidified air for 48 to 72 hours. Isolated colonies were subcultured onto Bolton agar with 50 ug/mL of kanamycin to select for gfp-expressing clones. Four milliliter (4 mL) aliquots of Bolton broth without antibiotics, in a 6-well polystyrene tissue culture plate (Costar®, Corning, Corning, NY), were inoculated from isolated colonies from agar plates with kanamycin. The broth was incubated at 37°C, 5% C02 in humidified air at 200 RPM until the early log phase of growth, as determined by optical density (OD). The culture was diluted 1:100 in 75 mL fresh Bolton broth without antibiotics and incubated under the previously described 41 conditions until an OD6oo of approximately 0.1 was reached. Serial dilutions and plate counts from previous experiments have shown that this OD corresponds to the early log phase of grth and a concentration of approximately 1x108 chL. The bacteria were collected and pelleted by centrifugation and resuspended in serum free medium (SFM) (Sigma Inc., St. Louis, M0) for rectal inoculation of pigs. A subsarnple of the inoculum were examined microscopically using a Nikon Eclipse E600 with an Epi-fluorescence system (Mager Scientific, Dexter, M1) for darting motility and fluorescence prior to infecting pigs. Also, a sample from the prepared inocula was serially diluted and plated to determine actual concentration. Each pig was inoculated with approximately 2x109 cfu C. jejuni in a total volume of 10 mL SF M. Experimental design and inoculation procedure Pigs were divided into 4 groups as follows: Group 1: Control, euthanized after 1 hour; Group 2: Infected, euthanized after 1 hour; Group 3: Control, euthanized afler 24 hours; Group 4: Infected, euthanized after 24 hours. All infected pigs received C. jejuni resuspended in 10 mL SFM. All control pigs received 10 mL SFM without bacteria. All pigs were tested for the presence of C. jejuni both before and afier inoculation by culture of rectal swabs or fecal samples. Rectal swabs or fecal samples were placed in Cary Blair (Difco Laboratories, Detroit, MI) transport medium for later culture in the laboratory. After samples were taken for C. jejuni culture, the pigs were anesthetized with intramuscular injections of 4.4 mg/kg Telazol (Fort Dodge, Overland Park, KS) and 2.2 mg/kg xylazine (Butler, Columbus, OH). Once the pigs were fully anesthetized, they were placed in lateral recumbency on heating pads and covered with towels to maintain 42 body temperature throughout the procedure. For each pig, the perineum was cleaned with three scrubs, alternating betadine and isopropyl alcohol for each scrub, and a purse string suture pattern was placed around the anus using 0 silk suture and left untied. A sterile 14 French feeding tube was inserted into the anus and advanced 10 to 15 cm into the distal colon. The appropriate inoculum was slowly injected through the tube, which was immediately removed. The purse string was tightened, and the hindquarters were elevated slightly. All pigs remained anesthetized for at least 1 hour, after which they were either euthanized or allowed to recover from anesthesia, depending on group assignment. Pigs that were allowed to recover from anesthesia were reanesthetized, as previously described, 24 hours after inoculation. All pigs were euthanized with an intravenous injection of 86 mg/kg sodium pentobarbital (Fatal Plus®, Vortech Pharmaceuticals, Dearbom, MI). Sample collection The colons of euthanized pigs were removed from the abdomen and Opened. A separate set of sterile instruments was used for each group to eliminate cross contamination between infection groups. Full thickness samples of the distal colon and individual LGCS were collected. Samples were fixed in 3.7% formaldehyde for histology and irnmunohistochemistry, embedded in OCT compound for laser capture microdissection, or placed in cryovials and snap frozen in liquid nitrogen for nucleic acid isolation. 43 C. jejuni isolation and identification Rectal swabs or fecal samples were streaked onto Preston selective agar (Oxoid Inc., Basingstoke, Hampshire, England) and incubated at 42°C, 5% C02 in humidified air for 48 hours. Colonies isolated from rectal swabs or fecal samples of experimentally infected pigs were scored according to 5 criteria: 1) the bacteria grew on Campylobacter selective media under microaerophilic conditions (5% C02 at 42°C); 2) colonies were round, raised, had pink pigmentation, and were 1-2mm in diameter-morphology that was consistent with Campylobacter spp.; 3) microscopic examination revealed curved motile rods; 4) bacteria were positive for PCR amplification of the quinolone resistance determining region (QRDR) of the gyrA gene of C. jejuni [29]; and 5) bacteria fluoresced green when examined at 488 nm wavelength on a Nikon Eclipse E600 with an Epi-fluorescence system (Mager Scientific, Dexter, MI). Pigs were considered positive for the strain of C. jejuni used for infections if all 5 criteria were met. We found culture alone to be inadequate for detection of C. jejuni from experimentally inoculated animals, therefore we used 3 different PCR techniques to detect C. jejuni from tissue DNA. Total DNA was isolated from approximately 25 mg of frozen tissue using a Qiagen DNEasy tissue kit (Qiagen, Valencia, CA) following the manufacturer’s protocol. The DNA was quantified spectrophotometrically (Beckman, Fullerton, CA). To detect C. jejuni from experimental animals, the C. jejuni QRDR target was amplified by PCR from SOOng of total DNA isolated from full thickness tissue samples using the previously designed primers JL238 (5‘ TGG GTG CTG TTA TAG GTC GT 3') and JL239 (5' GCT CAT GAG AAA GT1" TAC TC 3') [29]. Amplicons were electrophoresed on a 1.8% agarose gel in 1X Tris-Acetate-EDTA (TAE) buffer with 44 0.2 ug/mL ethidium bromide. The fragments were visualized by UV illumination on an Alpha Imager Gel documentation system (Alpha Innotech Corporation, San Leandro, CA). A Restriction Fragment Length Polymorphism (RFLP) assay to detect and distinguish C. jejuni, C. coli, C. lari, and C. upsaliensis by analysis of a portion of the 23S rRNA gene was used to detect thermophilic campylobacters in experimental animals [30]. The target was amplified from 500ng of total pig tissue DNA using primers THERMI (5'-TAT TCC AAT ACC AAC ATT AGT-3') and THERM4 (5'-CTT CGC TAA TGC TAA CCC-3'). The amplicons were digested overnight with either AIuI or T sp5091 following the manufacturers protocols (New England Biolabs, Beverly, MD), and the resultant fragments were separated electrophoretically on a 2% agarose gel in 1X Tris-Acetate-EDTA (TAE) buffer with 0.2 ug/mL ethidium bromide. The fragments were visualized by UV illumination on an Alpha Imager Gel documentation system (Alpha Innotech Corporation, San Leandro, CA). Primers were designed to amplify a 543 bp fragment from the gfp gene (Genbank Number AF 292556) using Squeb soflware (Genetics Computer Group, Madison, WI). The forward primer sequence is 5'AGT GGA GAG GGT GAA GGT GAT G 3'; the reverse primer sequence is 5' AAG GGC AGA TTG TGT GGA CAG G 3'. A 25 [IL PCR reaction with 1X PCR buffer (Tris-HCL, pH 8.3, 50 mM KCl), 1.0 mM MgCl2, 0.05 mM each dATP, dTTP, dCTP, dGTP, 37.5 ng of each primer, 0.625 U AmpliTaq Gold DNA polymerase (Applied Biosystems, Foster City, CA), and 500ng total tissue DNA was used to amplify the target. Amplification parameters were initial denaturation at 94°C for 10 minutes, followed by denaturation for 1 minute at 94°C, annealing for 1 minute at 45 62°C, extension for 1 minute at 72°C for 39 cycles, and a final extension at 72°C for 10 minutes. The amplicons were electrophoresed on a 1.8% agarose gel, stained, and visualized as previously described. Histology and Immunohistochemistry Fixed tissue samples were embedded in paraffin and 5 pm sections were cut and adhered to charged slides. Slides were routinely dehydrated in progressively higher concentrations of ethanol, deparaffinized and rehydrated. For histological evaluation, slides were stained with Mayer's hematoxylin and eosin and cover slipped. For irnmunostaining, sections of selected tissues were incubated in preheated antigen retrieval buffer (Dako, Carpinteria, CA) for 20 minutes at 100° C. Slides were removed from the steamer and left in the retrieval solution at room temperature to cool down for 20 minutes, rinsed in distilled water and transferred to Tris-HCL buffer. Endogenous peroxidase was blocked for 15 minutes with 3% hydrogen peroxide. Non-specific immunoglobulin binding blocking was done by a 10 minute incubation with a protein- blocking agent (Dako, Carpinteria, CA) prior to application of the primary antibody. A rabbit polyclonal anti-C. jejuni antibody (Biogenesis, New Fields, UK) was used as the primary antibody at a concentration of 1:100. Sections were stained with a Dako autostainer using a labeled streptavidin-immunoperoxidase staining procedure (Dako Corp., Carpinteria, CA). The immunoreaction was visualized with ABC (Dako Corp., Carpinteria, CA). Sections were counterstained with Mayer’s hematoxylin, then dehydrated, cleared and mounted. Sections of distal colons from pigs in which infection with C. jejuni had been confirmed by PCR served as positive controls. 46 Determination of antibody specificity Healthy, conventionally reared pigs have been shown to have more than one species of Campylobacter residing as commensals in their intestines [7]. Due to the likelihood that the pigs used in this study also harbored other Campylobacter species and the possibility of cross reactivity with those species, we tested the specificity of the polyclonal antibody for C. jejuni. IPEC-l cells are a non-immortalized swine intestinal epithelial cell line derived from neonatal pigs. All cell culture reagents were obtained from Invitrogen unless otherwise indicated. IPEC-l cells were grown in DMEM/F12 media supplemented with 5% fetal bovine serum (FBS) and 1% Insulin-Transferrin-Selenium(ITS) in Costar culture flasks (Costar®, Corning, Corning, NY). Cells were washed in versene and detached with 0.05% Trypsin-EDTA, then seeded at a density of 5x105 cells per well onto glass chamber slides coated with 20ug/mL fibronectin (Sigma, St. Louis, MO). The cells were incubated at 37°C, 5% C02 until confluent (2-3 days). C. jejuni 81-176 expressing gfp, C. coli 43134, C. lari 43675, and C. upsaliensis 43954 were streaked for isolation onto Bolton agar plates (Oxoid Inc., Basingstoke, Hampshire, England) and incubated for 72 hours at 37°C, 5% C02. Isolated colonies were streaked for lawn growth and incubated for 20 to 24 hours. Lawns were harvested from plates with sterile swabs, resuspended in DMEM/F12 supplemented with 5%F BS and 1%ITS, and adjusted to an 0Deoo of 0.1. IPEC-l cells were inoculated with an M01 100:] of each Campylobacter species and incubated for 3 hours at 37°C, 5% C02, The supematants were removed and 47 the cells were fixed in 10% formaldehyde, then stained as previously described with the anti-C. jejuni antibody. Laser Capture Microdissection Five micron sections were cut from tissue samples embedded in OCT compound on a Reichert-Jung cryotome (Reichert-Jung , Bensheim, Germany) and adhered to poly- L-lysine (Sigma, St. Louis, MO) coated slides. Slides were rinsed in ethanol, stained with Mayer’s Haemotoxylin, dehydrated in progressively higher concentrations of ethanol, then rinsed in xylene. Crypt units and submucosa] samples, avoiding the surface mucosa, were microdissected using a Pixcell II laser capture Microdissection system (Arcturus, Mounrainview CA). Samples were adhered to CapSure HS LCM Caps (Arcturus, Mounrainview CA) and stored at -20°C for later DNA isolation. For DNA extraction, caps were incubated in Proteinase K digestion buffer (1mg/ml Proteinase K (Qiagen, Valencia, CA) / 1% Tween 20 (Sigma) in TE Buffer pH.8.0) overnight at 37°C. Samples were heated to 95°C for 10 minutes to inactivate the Proteinase K and stored at -20°C. To determine if the inoculating strain was present in the crypts and submucosa, IOuL of DNA were used as a template in a PCR reaction with primers designed in our laboratory to detect the gp gene. RFLP analysis of the Campylobacter 23S rRNA gene was also performed to determine if other Campylobacter strains were present in the crypts and submucosa[3 O]. 48 Cytokine Analysis To determine the effect of rectal inoculation of C. jejuni on local immune responses in the distal colon and LGCS, mRNA expression of 14 cytokines, chemokines and iNOS was measured using a real time PCR assay. Total RNA was isolated from frozen tissue samples from each pig using Trizol reagent (GibcoBRL Life Technologies). Briefly, 3mm3 tissue samples were homogenized in Trizol using a Polytron® tissue homogenizer (Kinematica, Cincinnati, OH). RNA was extracted with chloroform, precipitated with isopropanol, and washed with ethanol. Pellets were air dried, then resuspended in DEPC treated H2O (GibcoBRL Life Technologies). DNA contamination was removed by treatment with approximately 27 Kunitz units of DNase I for 15 minutes. Samples were repurified on a Qiagen RNEasy column (Qiagen, Valencia, CA). RNA concentration and integrity were determined using an Agilent Bioanalyzer 2100 (Agilent Technologies, Palo Alto, CA). For each sample, 10 pg of total RNA was reverse transcribed with random primers using a Stratagene First Strand RT-PCR kit (Stratagene, La Jolla, CA). First strand cDNA was used as the template in real time PCR reactions. Real time PCR primers and probes designed using the Primer Express (Applied Biosystems, Foster City, CA) software package were used to analyze 14 cytokines, chemokines, and iNOS (Table 2). The 18S rRNA probe was obtained from Applied Biosystems and was used as a constitutively active gene for normalization. PCR was performed using a commercially available kit (Brilliant kit, Stratagene, La Jolla, CA) on an ABI PRISM 79OOHT (Applied Biosystems). Amplification conditions were as follows 50°C for 2 minutes; 95°C for 10 minutes; 40 cycles of 95°C 15 seconds and 60°C for 1 minute. Fluorescence signals measured during amplification were processed post 49 amplification. Threshold cycle (Ct) values were determined using values of 0.2 for TET, 0.1 for VIC, and 0.05 for FAM reporter dyes. Statistical Analysis The cytokine mRNA expression data for each tissue was analyzed separately using Statview 5.0 for Macintosh (Abacus Concepts, Berkeley, Calif). The CI value for the 18S ribosomal subunit was subtracted from the C, value for each cytokine message to normalize for RNA content prior to statistical analysis. One-way analysis of variance (ANOVA) was performed to determine the statistically significant differences between treatments. A Fisher’s least squares difference test was performed to determine differences between infected and control animals at each time point. P values of 0.05 or less were considered statistically significant. 50 Results Campylobacter isolation and identification C. jejuni was not isolated from any fecal samples taken prior to infection (data not shown). One hour afler infection, C. jejuni was not recovered from any infected or control pigs (Table 1). Twenty four hours after inoculation, C. jejuni was recovered from 1 of 4 infected pigs and 0 of 4 control pigs. All C. jejuni isolates recovered from pigs were confirmed to be the inoculating strain by examination using fluorescence microscopy. PCR analysis of total DNA isolated from pig tissues proved to be a more sensitive assay for the detection of C. jejuni from experimentally inoculated pigs. One hour after inoculation, C. jejuni was detected in all 4 infected pigs and none of the control pigs using primers specific for the QRDR of the gyrA gene of C. jejuni. Twenty four hours after inoculation, C. jejuni was detected in 1 of 4 infected pigs and 1 of 4 control pigs (Figure l). The infected pig that was positive on culture 24 hours after inoculation was the same pig that was positive by amplification of the QRDR target. Analysis of the Campylobacter 23S rRNA RFLP demonstrated that both C. jejuni and C. coli were present in our experimental animals (Figure 2). One hour after inoculation, 3 of 4 infected pigs had patterns indicative of C. jejuni and the remaining pig had a pattern indicative of C. coli. All control pigs had patterns indicative of C. coli. Additionally, 24 hours afier inoculation, all infected and control animals had patterns indicative of C. coli only. Neither C. lari nor C. upsaliensis were detected from any pig. These data strongly suggest that all of the pigs were colonized by C. coli prior to 51 infection and at one hour after experimental infection C. jejuni is the predominant species, but by 24 hours C. coli is predominant. Detection of the gp gene proved to be the most sensitive assay for detection of C. jejuni from experimentally inoculated pigs. One hour after inoculation, the mp target was detected fi'om all infected pigs and none of the controls (Figure 3). Twenty four hours after inoculation, the target was detected from 2 Of 4 infected pigs and none of the controls. Histology and Immunohistochemistry Distal colonic tissue samples were examined microscopically to determine if lesions were induced by rectal inoculation with C. jejuni. Additionally, the location of Campylobacter organisms in the samples was determined by immunohistochemistry using a rabbit polyclonal anti-C. jejuni antibody. Control pigs showed no degenerative, proliferative, or inflammatory lesions. Most experimentally infected pigs also showed no degenerative, proliferative, or inflammatory lesions (data not shown). Tests to determine the specificity of the polyclonal antibody used for immunocytochemical staining showed strong cross reactivity with C. lari as well as C. jejuni, but no cross reactivity with C. coli, C. hyointestinalis, or C. upsaliensis (Figure 4). Microscopic examination of tissues stained with the polyclonal antibody showed positively stained rods along the surface epithelium, within the mucus of the crypts and the crypts of the LGCS, and associated with goblet cells (Figure 5A through D). Staining patterns were similar at both 1 and 24 hours and were detected in both infected and control animals. 52 Interestingly, one pig had more severe lesions at 1 hour post inoculation characterized by multifocal distention of crypts with mucus, neutrophils, and small curved rods that stained positively on IHC (Figure 5E). Crypts within the LGCS were also distended with mucus, neutrophils, and positively staining curved rods. Lymphocytes, neutrophils, and plasma cells had infiltrated the epithelium and superficial propria, and there was evidence of epithelial degeneration. Immunocytochemical staining of rod shaped bacteria in the distal colonic crypts suggested that Campylobacters were located in the crypts. However, cross reactivity of the antibody with C. lari as well as C. jejuni necessitated a more specific technique to determine if the bacteria in the crypts were the same strain used for inoculation. We used laser capture microdissection to specifically isolate bacteria and enterocytes from the crypts and submucosa to test by PCR for C. jejuni specific targets. Laser Capture Microdissection Laser capture microdissection was performed to isolate crypt units and submucosa, avoiding the surface epithelium, to determine if the C. jejuni strain used for inoculation was present. A PCR using primers specific for the gfi) gene amplified the target only from the infected pig that was tested, while the control animal was negative (Figure 6). Additionally, RFLP analysis of the Campylobacter 23s rRNA gene was performed to determine if other Campylobacter species were present in crypts and submucosa. The infected animal had banding patterns consistent with C. jejuni and C. coli. A faint pattern that could be consistent with C. upsaliensis was also seen. The control animal had banding patterns that were consistent with C. coli, and an additional 53 band at ~150bp that did not fit the patterns Of C. jejuni, C. coli, C. lari, or C. upsaliensis (Figure 7). Cytokine mRNA expression We measured mRNA expression of 14 cytokines, chemokines, and iNOS in both the distal colon and LGCS to determine if C. jejuni had a significant impact on cytokine expression (Table 2). One hour after infection, expression of IL-18, TNFor, and IL-8 was significantly upregulated 2 to 3 fold over controls in the distal colon and IL-8 remained upregulated 24 hours after infection. At the same time in the LGCS expression Of IL-18, TNFOI, IL-8, GM-CSF, IL-IB, iNOS, MCP-l, IL-6, and IL-10 was upregulated 3 to 20 fold over controls (Figure 8). Downregulation of mRNA expression was not seen for any target measured. These data indicates that a proinflammatory response was initiated by C. jejuni infection, and the response was more robust in LGCS compared to the distal colon. 54 Discussion Rectal inoculation of C. jejuni stimulates a proinflammatory cytokine response in the colon characterized by increased expression of IL-IB, IL-6, IL-8, IL-18, GM-CSF, iNOS, MCP-l, and TNFOI. In samples of the distal colon excluding LGCS, expression of IL-18, TNFOL, and IL-8 was significantly upregulated 1 hour after inoculation, and IL-8 remained upregulated 24 hours after inoculation. These data correlate with cytokine production from in vitro infections of intestinal epithelial cells and monocytes with C. jejuni. Swine intestinal epithelial cells, IPEC-l, secrete IL-IB, TNFOI, IL-6, IL-8 and IL- 18 upon infection with C. jejuni (Parthasarathy and Mansfield, unpublished data; Jones et al, unpublished data; Cunningham and Mansfield, unpublished data). Human intestinal epithelial cells, INT-407, have been shown to secrete IL-8 in response to C. jejuni [15, 16]. In addition, intracellular production of IFNy, TNFOI, IL-4, and IL-10 is increased with C. jejuni infection [31]. In our model immunohistocherrristry detected Campylobacter organisms associated with surface epithelial cells, goblet cells, and within the mucus of distal colon crypts. Histologically, no inflammatory changes were seen in infected pigs, therefore the significantly increased cytokines are likely due to the interaction of C. jejuni with intestinal epithelial cells. C. jejuni targets lymphoglandular complexes in the distal colons of pigs after oral inoculation, stimulating an immunoglobulin response [13]. LGCS from C. jejuni infected pigs are composed of the follicle associated epithelium, B cell rich germinal centers, T cells, and cells with macrophage morphology [13, 25]. In vitro human monocytes secrete IL-10, IL-6, IL-8, and TNF-a upon infection with C. jejuni [18]. In our rectal challenge model, IHC 55 demonstrated Campylobacter organisms within the entrapped crypts of the LGCS and within the follicle associated with leukocytes. In the LGCS IL-10, IL-6, IL-8, IL-18, GM- CSF, iNOS, MCP-l, and TNFOL were significantly upregulated. In addition, the response was more robust than in the non-lymphoid portions of the distal colon with 3 to 20 fold increases in cytokine expression compared to 2 to 3.5 fold increases in the distal colon. In contrast to the distal colon, the increased cytokines in the LGC are likely due primarily to the interaction of C. jejuni with leukocytes, which are capable of producing an expanded cytokine profile and greater levels of cytokines [15, 18]. Ultimately, both tissues showed induction of a proinflammatory response. The enhanced response in LGCS supports other data showing that C. jejuni specifically targets the lymphoid tissue [13, 25]. Proinflammatory cytokines are induced by a variety of enteric pathogens. Salmonella, Shigella, and Campylobacter increase rectal nitric oxide gas and stool IL-1 [3 levels in humans during the acute phase of infection [32]. Shigella stimulates production of IL-IB, IL-8, and IL-18 in animal models of infection. These cytokines are critical for initiating inflammation that limits invasion of the bacteria into the lamina propria and ultimately eliminates the bacteria from the host [33, 34]. Salmonella stimulates expression of IL-1, IL-6, IL-8, IL-18, and TNFOI from intestinal tissues of pigs [35, 36]. In mice, wild type Salmonella decreases expression of 1L-18 in Peyer's patches and mesenteric lymph nodes, which is believed to be a mechanism for evading the immune response and establishing an infection [37]. Helicobacter pylori stimulates increased expression of IL-10, IL-6, IL-18 and TNFOI in the gastric mucosa of humans and nonhuman primates [3 8-40]. Given that several enteric bacterial pathogens induce 56 proinflammatory cytokines in vivo, and C. jejuni induces proinflammatory cytokines in vitro, it is not surprising that expression of several proinflammatory cytokines was increased in the distal colons and LGCS of rectally infected pigs as these cytokines likely play a role in resistance to C. jejuni infection. In our model, rectally inoculated C. jejuni behaved in a similar manner to orally inoculated bacteria. Oral inoculation studies have demonstrated C. jejuni within entrapped crypts and in the follicle associated epithelium of distal colonic LGCS of germ free piglets [25], and within superficial mucosa] epithelial cells of the colons of colostrums deprived piglets [24]. These bacteria have traversed the gut and encountered the acidic pH of the stomach and bile in the small intestine, two environmental conditions that can affect the virulence of the organism [41]. The C. jejuni flaA promoter can be upregulated by acidic pH, bovine bile, and deoxycholate which suggests that these stimuli enhance production of flagella, an important virulence determinant [41, 42]. Additionally, bile stimulates the synthesis of Cia proteins, which are essential for C. jejuni invasion into intestinal epithelial cells [41]. However, these environmental stimuli are not absolutely necessary for C. jejuni virulence. In vitro, routinely cultured bacteria invade intestinal epithelial cells [15, 28, 43] and elicit cytokine responses [15-17]. In vivo, ferrets rectally infected with C. jejuni become colonized and develop diarrhea similar to orally infected animals [23]. Our data correlates with these findings, demonstrating Campylobacter organisms associated with epithelial cells and goblet cells, within distal colonic crypts, and within crypts of the LGCS at both 1 hour and 24 hours after inoculation. PCR analysis of microdissected crypts confirmed that C. jejuni 81-176 expressing gfp was present in infected pigs and not controls. Thus it was not entirely 57 unexpected that cytokine expression was significantly increased in the distal colon and LGCS Positive staining with immunohistochemistry on the mucosal surface, in the crypts, and in the entrapped crypts Of the LGCs of rectally infected pigs suggests that C. jejuni is present in these areas. PCR analysis of DNA from full thickness tissue samples and microdissected crypts and submocosa confirm that the inoculating strain of C. jejuni had invaded the crypts and submucosa of infected pigs. Positive staining was also seen in control pigs but PCR analysis of DNA samples did not corroborate the presence of C. jejuni specifically. Cross reaction of the polyclonal antibody with closely related Campylobacter species could account for positive staining in control pigs. All control pigs harbored C. coli, as determined by RFLP analysis, but our tests to determine antibody specificity showed cross reaction only with C. lari (Figure 5). C. lari has been detected by culture in the feces of healthy pigs [7] as well as from abscessed LGCs from pigs with mucohemorrhagic diarrhea due to Trichuris suis infection [44]. All control and infected animals from our rectal inoculation study were negative by PCR for the C. lari Ipr gene (data not shown) [45]. Interestingly, Mansfield and Urban isolated an unidentified Campylobacter species from abscessed LGCs as well as C. jejuni, C. coli and C. lari [44]. These bacteria were all part of the resident flora of conventionally reared pigs prior to T. suis infection, and could also have been present in the pigs used in our experiment. In light of this, the changes in cytokine expression in the distal colon and LGCS could also be due in part to perturbation of the resident bacterial population upon introduction of exogenous Campylobacter. 58 Due to the low incidence of C. jejuni cultured from pigs after infection, we used three different PCR methods to detect C. jejuni in DNA isolated from pig tissue samples: amplification of the C. jejuni QRDR, RFLP of Campylobacter 23S rRNA gene, and amplification of the gfp gene. The RF LP analysis gave valuable information about the species of Campylobacter present in the distal colons of the experimentally infected piglets, but it was not as sensitive for detection of the inoculation strain as was expected. Therefore, we developed a PCR assay to detect the gfp gene that would only be present in the samples if the inoculating strain was present in the tissue. The PCR protocols for amplification of C. jejuni QRDR and Campylobacter 23S rRNA gene were originally designed for use on bacterial cultures, but we were able to adapt the protocols and successfully amplify Campylobacter targets from mixed DNA samples containing both pig and bacterial DNA. We found these assays to be more rapid and sensitive for Campylobacter detection and identification in infection studies than traditional culture methods. Overall, we believe that culture alone is insufficient to detect and distinguish Campylobacters that are present in the gut of conventionally reared pigs. PCR analyses were more sensitive and specific for detection and identification and did not rely on the ability to recover viable bacteria, as would be needed for traditional speciation by biochemical means. The single pig that was infected for only one hour but had significant microscopic lesions was an unusual finding. Clearly there was an inflammatory response occurring in this animal as was evident by infiltration of inflammatory cells into the lamina propria and the existence of multifocal crypt abscesses (Figure 5E). It is unlikely that the abscessation was caused by the C. jejuni inoculum as this pig was only infected for one 59 hour. However, it is interesting to note that there were Campylobacter organisms associated with the abscessed crypts. Whether the organisms were from the inoculum or from the resident population the pathology supports the hypothesis that Campylobacter spp. are opportunistic pathogens in the distal colons of pigs. Campylobacter organisms invade abscessed LGCs in the distal colons of both immunocompetent pigs infected with the whipworm T suis [44] and germ free piglets infected with T. suis and C. jejuni [25]. In this dual infection model, it is hypothesized that the whipworm infection facilitates secondary infection by resident gut flora through dysregulation of the immune response, or by direct effects on enterocytes that permit bacterial invasion [44] (Parthasarthy, unpublished results). It is possible that in the rectally inoculated animal from our study, some undetermined agent facilitated Campylobacter invasion similar to T. suis facilitation of C. jejuni invasion. Rectal inoculation proved to be a useful tool for studying the pathogenesis of C. jejuni infection in a swine model. Rectal inoculation allowed targeted delivery of a specific amount of bacteria to the distal colon and a specific time frame to study the effects on cytokine expression in the distal colon. The fact that significant changes in cytokine expression were measured after rectal inoculation with C. jejuni confirms that the bacteria are capable of eliciting a response when delivered by a non conventional route. This model allows for delivery of the agent of interest to a discrete area of the intestine and assessment of responses at more accurate time points. We believe that this model will continue to be useful for studying the complex interaction between pathogen and host in vivo. 60 10. ll. 12. l3. 14. 15. l6. 17. References Allos, B.M., Campylobacterjejuni Infections: update on emerging issues and trends. Clin Infect Dis, 2001. 32(8): p. 1201-6. Altekruse, S.F., D.L. Swerdlow, and NJ. Stern, Microbial food borne pathogens. C ampylobacter jejuni. Vet Clin North Am Food Anim Pract, 1998. 14(1): p. 31-40. Ketley, J.M., Pathogenesis of enteric infection by Campylobacter. Microbiology, 1997. 143 (Pt 1): p. 5-21. Hughes, R.A. and J .H. Rees, Clinical and epidemiologic features of Guillain-Barre syndrome. J Infect Dis, 1997. 176 Suppl 2: p. 892-8. Torre, E. and M. Tello, Factors influencing fecal shedding of C ampylobacter jejuni in dogs without diarrhea. Am J Vet Res, 1993. 54(2): p. 260-2. Wolfs, T.F., et al., Neonatal sepsis by Campylobacterjejuni: genetically proven transmission from a household puppy. Clin Infect Dis, 2001. 32(5): p. E97-9. Young, C.R., et al., Enteric colonisation following natural exposure to Campylobacter in pigs. Res Vet Sci, 2000. 68(1): p. 75-8. Altekruse, S.F., et al., Food and animal sources of human C ampylobacter jejuni infection. J Am Vet Med Assoc, 1994. 204(1): p. 57-61. Cawthraw, S.A., et al., Long-term antibody responses following human infection with Campylobacterjejuni. Clin Exp Immunol, 2002. 130(1): p. 101-6. Panigrahi, P., et al., Human immune response to Campylobacterjejuni proteins expressed in vivo. Infect Immun, 1992. 60(11): p. 4938-44. Calva, J .J ., et al., Cohort study of intestinal infection with campylobacter in Mexican children. Lancet, 1988. 1(8584): p. 503-6. Taylor, D.N., et al., Influence of strain characteristics and immunity on the epidemiology of Campylobacter infections in Thailand. J Clin Microbiol, 1988. 26(5): p. 863-8. Mansfield, LS. and D.T. Gauthier, Lymphoglandular complexes are important colonic sites for immunoglobulin A induction against C ampylobacter jejuni in a swine disease model. Comp Med, 2004. 54(5): p. 514-23. Baqar, 8., ND. Pacheco, and F.M. Rollwagen, Modulation of mucosal immunity against Campylobacterjejuni by orally administered cytokines. Antimicrob Agents Chemother, 1993. 37(12): p. 2688-92. Hickey, T.E., et al., C ampylobacter jejuni-stimulated secretion of interleukin-8 by INT407 cells. Infect Immun, 1999. 67(1): p. 88-93. Hickey, T.E., et al., Campylobacterjejuni cytolethal distending toxin mediates release of interleukin-8 from intestinal epithelial cells. Infect Immun, 2000. 68(12): p. 6535-41. Mellits, K.H., et al., Activation of the transcription factor NF -kappaB by Campylobacterjejuni. Microbiology, 2002. 148(Pt 9): p. 2753-63. 61 l8. 19. 20. 2]. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. Jones, M.A., et al., Induction ofproinflammatory responses in the human monocytic cell line THP-l by Campylobacterjejuni. Infect Immun, 2003. 71(5): p. 2626-33. Baqar, S., et al., Murine intranasal challenge model for the study of Campylobacter pathogenesis and immunity. Infect Immun, 1996. 64(12): p. 4933-9. Fox, J.G., et al., Gastroenteritis in NF -kappaB-deficient mice is produced with wild-type Camplyobacterjejuni but not with C. jejuni lacking cytolethal distending toxin despite persistent colonization with both strains. Infect Immun, 2004. 72(2): p. 1116-25. Pancorbo, P.L., et al., Evaluation of cytokine production and phagocytic activity in mice infected with Campylobacterjejuni. Curr Microbiol, 1999. 39(3): p. 129-33. Pancorbo, P.L., et al., Potential intervention of Campylobacter jejuni in the modulation of murine immune response. Curr Microbiol, 2001. 43(3): p. 209-14. Bell, J .A. and DD. Manning, A domestic ferret model of immunity to Campylobacterjejuni- induced enteric disease. Infect Immun, 1990. 58(6): p. 1848-52. Babakhani, F.K., G.A. Bradley, and LA. Joens, Newborn piglet model for campylobacteriosis. Infect Immun, 1993. 61(8): p. 3466-75. Mansfield, L.S., et al., Enhancement of disease and pathology by synergy of Trichuris suis and C ampylobacter jejuni in the colon of immunologically naive swine. Am J Trop Med Hyg, 2003. 68(3): p. 70-80. Abolhassani, M., et al., Mycobacterium bovis BCG induces similar immune responses and protection by rectal and parenteral immunization routes. Infect Immun, 2000. 68(10): p. 565 7-62. Zhao, J ., et al., Improved protection of rhesus macaques against intrarectal simian immunodeficiency virus SI V (mac25 I ) challenge by a replication-competent Ad5hr-Sl V env/rev and A d5hr-Sl Vgag recombinant priming/gpl20 boosting regimen J Virol, 2003. 77(15): p. 83 54-65. Miller, W.G., et al., Detection on surfaces and in Caco-2 cells of Campylobacter jejuni cells transformed with new gfp, yfp, and cfp marker plasmids. Appl Environ Microbiol, 2000. 66(12): p. 5426-36. Wilson, D.L., et al., Identification of ciprofloxacin-resistant C ampylobacter jejuni by use of a fluorogenic PCR assay. J Clin Microbiol, 2000. 38(11): p. 3971-8. F ermer, C. and E0. Engvall, Specific PCR identification and differentiation of the thermophilic campylobacters, Campylobacterjejuni, C. coli, C. lari, and C. upsaliensis. J Clin Microbiol, 1999. 37(10): p. 3370-3. Al-Salloom, F.S., et al., Campylobacter-stimulated IN T407 cells produce dissociated cytokine profiles. J Infect, 2003. 47(3): p. 217-24. Enocksson, A., eta1., Rectal nitric oxide gas and stool cytokine levels during the course of infectious gastroenteritis. Clin Diagn Lab Immunol, 2004. 11(2): p. 250-4. Sansonetti, P.J., et al., Interleukin-8 controls bacterial transepithelial translocation at the cost of epithelial destruction in experimental shigellosis. Infect Immun, 1999. 67(3): p. 1471-80. Sansonetti, P.J., et al., Caspase-I activation of IL-1 beta and IL-18 are essential for Shigella flexneri-induced inflammation Immunity, 2000. 12(5): p. 581-90. 62 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. Cho, W.S. and C. Chae, Expression of inflammatory cytokines (T NF -alpha, IL- 1, IL-6 and IL-8) in colon of pigs naturally infected with Salmonella typhimurium and S. choleraesuis. J Vet Med A Physio] Pathol Clin Med, 2003. 50(10): p. 484-7. Foss, D.L., M.J. Zilliox, and M.P. Murtaugh, Bacterially induced activation ofinterleukin-18 in porcine intestinal mucosa. Vet Immunol Immunopathol, 2001. 78(34): p. 263-77. Elhofy, A. and K.L. Bost, Limited interleukin-I8 response in Salmonella-infected murine macrophages and in Salmonella-infected mice. Infect Immun, 1999. 67(10): p. 5021-6. Fera, M.T., et al., Correlation between Helicobacter pylori infection and IL-18 mRNA expression in human gastric biopsy specimens. Ann N Y Acad Sci, 2002. 963: p. 326-8. Harris, P.R., et al., Inflammatory cytokine mRNA expression during early and persistent Helicobacter pylori infection in nonhuman primates. J Infect Dis, 2000. 181(2): p. 783-6. Tomita, T., et al., Expression of Interleukin-1 8, a Th1 cytokine, in human gastric mucosa is increased in Helicobacter pylori infection. J Infect Dis, 2001. 183(4): p. 620-7. Rivera-Amill, V., et al., Secretion of the virulence-associated Campylobacter invasion antigens fiom Campylobacterjejuni requires a stimulatory signal. J Infect Dis, 2001. 183(11): p. 1607-16. Allen, K.J. and M.W. Griffiths, Eflect of environmental and chemotactic stimuli on the activity of the Campylobacterjejuni/MA sigma(28) promoter. FEMS Microbiol Lett, 2001. 205(1): p. 43-8. Abner, S.R., et al., Response of intestinal epithelial cells to Trichuris suis excretory-secretory products and the influence on C ampylobacter jejuni invasion under in vitro conditions. J Parasitol, 2002. 88(4): p. 738-45. Mansfield, LS. and J .F. Urban, Jr., The pathogenesis of necrotic proliferative colitis in swine is linked to whipworm induced suppression of m ucosal immunity to resident bacteria. Vet Immunol Immunopathol, 1996. 50(1-2): p. 1-17. Wemo, A.M., eta1., Fatal case of Campylobacter lari prosthetic joint infection and bacteremia in an immunocompetent patient. J Clin Microbiol, 2002. 40(3): p. 1053-5. 63 Inoculation Culture C. jejuni gfp PCR 23S rRNA RFLP QRDR C. jejuni C. coli C.jejuni 00f4 3of4 40f4 3of4 lof4 1hr Control 0 of 4 0 of 4 O of 4 0 of 4 4 of 4 1hr Cjejuni lof4 lof4 20f4 00f4 4of4 24 hr Control 0 of 4 1 of 4 0 of 4 0 of 4 4 of 4 24hr Table 1: Summary of Campylobacter detection and identification. Campylobacters were detected by culture of feces, or PCR for C. jejuni, gfi), or Campylobacter 23S rRNA targets using total DNA isolated from distal colonic tissue samples as the template. 64 Figure l: Amplification of the QRDR of C. jejuni gyrA from distal colon DNA from experimentally infected pigs. Lane 1: 100bp DNA ladder; Lane 2: DNA from C. jejuni expressing gfp; Lanes 3-6: 24 hour infected pigs; Lanes 7-10: 1 hour infected pigs; Lanes 11-18: uninfected controls. 65 1 2 3 4 s 6 7 8 910111213141516171819 Figure 2: Alu I digest of Campylobacter 23s rRNA gene amplified from pig tissues by PCR. Lane 1: 100bp DNA ladder; Lane 2: C. jejuni; Lane 3: C. coli ; Lanes 4-7 : 24 hour infected pigs ; Lanes 8-11 : 1 hour infected pigs ; Lanes 12-19 : uninfected controls. 66 12 3 45 678 9101112131415161718 Figure 3: PCR testing for gfp. A 543 bp amplified fragment of the gfl gene was amplified from total DNA isolated from distal colonic pig tissues. Lane 1: 100bp DNA ladder; Lane 2: DNA from C. jejuni expressing gfp; Lanes 3-6: 24 hour infected pigs; Lanes 7-10: 1 hour infected pigs; Lanes 11-18: uninfected controls. 67 Figure 4: Immunohistochemistry of Campylobacter infected IPEC-l cells. Immunohistochemistry was performed on IPEC-l cells infected with one of five Campylobacter species to determine antibody specificity. A: C. jejuni 81-176 expressing gfp; B: C. coli 43134; C: C. lari 43675; D: C. upsaliensis 43954 ; E : C. hyointestinalis 35217. (100X magnification) 68 Figure 6: Immunohistochemistry of distal colon samples using the polyclonal antibody detected Campylobacter spp. organisms associated with the brush border and in the lumen (A), within the crypts (B), within the crypts of the LGCs and associated with inflammatory cells (C), and associated with goblet cells, possibly within the cytoplasm (D) (100X magnification). One pig had organisms within crypt abscesses (E) (20X magnification). 69 12345 Figure 6: PCR testing DNA isolated from LCM samples. Primers designed to amplify a fragment of the gfp gene were used to test DNA isolated from LCM samples. Lane 1: 100bp DNA ladder; Lane 2: DNA from C. jejuni expressing gfp; Lane 3: DNA from a full thickness tissue sample taken from an infected animal; Lane 4: DNA from microdissected crypts and submucosa from an infected animal; Lane 5: DNA from microdissected crypts and submucosa from a control animal. 70 112345678' Figure 7: Alu I digest of Campylobacter 23s rRNA gene PCR product amplified from microdissected crypts and submucosa. Lane 1: 100bp DNA ladder; Lane 2: C. jejuni 81- 176 expressing gfp; Lane 3: C. lari 43675; Lane 4: C. upsaliensis 43954; Lane 5: C. coli 43134; Lane 6: DNA from a full thickness tissue sample taken from an infected animal; Lane 7: DNA from microdissected crypts and submucosa from an infected animal; Lane 8: DNA from microdissected crypts and submucosa from a control animal. 71 GM-CSF iNOS IL-1B lL-18 TNF-a lL-8 MCP-1 IL-6 1L-12p35 lL-1 2p40 IFNy IL-4 IL-5 IL-10 IL-13 Distal Colon LlLllJLlLllelLliflLlLlLl 9V 9@171F][TEN—11117FH‘IIJITVIFIHIT 94.—l 55.. Figure 8: Cytokine expression in distal colonic tissues. Expression of mRNA from a panel of cytokines was measured by Real Time PCR. The data represents the average fold change in expression from infected animals when compared to control animals. Letters denote statistically significant changes. A: p50.05; B: p500]. 72 €253 N 2.3 2: of EEH-» Am 35% m anew 89a .6 353 v 28 2 355 N 339% E 808E 33 8658:. EEO .&E @8003 88m 801:0 833m mfim ”m 9.53% .1» “L .. M., gum 106 aofiéews: xo: v anew ua 35$ w Anouaomiwafi v83 m 955 ”0 35m MN 955 “m Baum : 9.80 u< ESE .mnsohm deacon“: :5 95 £838 MO 385 05 5 25:390 @0qu 3338a .53me U ”gamma 3&8 33:5 Eco—oboa 2: .8288 3:28 82m Fmaonoomeogg no 953..— ...4..... 3.. km". 107 Distal Colon 5 no _ O o — GM-CSF iNOS MCP-l TNF-a IL-1 [3 IL-6 IL-8 IL-18 IL—12p35 lL-lZp40 IFNy lL—4 IL-5 flDflflmDflflfll—H—H—lflflg LJLJLJLJLJLILJLILJLJLILJLJLI ”MIDDDDDEDDDEDDED IL-l0 IL-13 mjDDDDDDDDDDDDDD' WTHUDDHHHEDUHDHI MWDJIJ[JIJIJIJIJIJIJIJIJIJIJIJ[J3 mun[JE]D[]E]D[JE]D[]EJD[]Ef WTHHHHHHHHHHHDDH M119! '3 gimp! '3 ‘ : Figure 7: Cytokine expression in intestinal tissues. Expression of mRNA from a panel of cytokines was measured by Real Time PCR. The data represents the average fold change in expression from infected animals when compared to control animals. Letters indicate statistically significant changes: A: p: 0.05; B: p: 0.01 108 Chapter 4: “Campylobacterjejuni stimulates IL-18 secretion from differentiated intestinal epithelial cells” Abstract Campylobacter jejuni is the leading bacterial cause of foodborne illness worldwide. Clinical signs of campylobacteriosis mimicking human disease have been reproduced in piglets. Upon infection, the bacteria penetrate the intestinal mucus layer, and adhere to and invade intestinal epithelial cells, resulting in inflammation. We have previously shown that C. jejuni induces proinflammatory cytokines in the distal colon and lymphoglandular complexes of rectally inoculated piglets one hour after infection. Additionally, C. jejuni has been shown to stimulate secretion of the proinflammatory cytokines IL-IB, TNFoc, IL-6, and lL-8 from intestinal epithelial cells and monocytes. Interleukin 18 is a pleiotrophic cytokine that is constitutively expressed in intestinal tissues and plays a critical role in initiating an immune response to pathogenic bacteria. Because intestinal epithelial cells play an important role in the pathogenesis of C. jejuni infection in vivo, we hypothesized that C. jejuni induces IL-18 from cultured swine intestinal epithelial cells. Differentiated and undifferentiated IPEC-l cells were infected apically with C. jejuni strain ATCC 33292 or 81-176. Supematants were collected at O, 1, 3, 6, 8, and 24 hours after infection and analyzed for IL—18 protein by ELISA. RNA was isolated from cells 8 hours after infection to assay for IL-18 mRNA expression by RT- PCR. Differentiated cells secreted significantly higher levels of IL-18 protein as early as 3 hours after infection, compared to uninfected controls. Undifferentiated cells did not 109 secrete significant levels of IL-18 at any time point. Expression of IL-18 mRNA was not significantly increased by C. jejuni 8 hours after infection from either differentiated or undifferentiated cells. These results indicate that differentiated intestinal epithelial cells are a significant source of IL-l8. Epithelial cell derived IL-18 may play an important role in the pathogenesis of C. jejuni infection in vivo. 110 Introduction Campylobacterjejuni (C. jejuni) is the most frequent bacterial cause of foodborne illness worldwide, with the CDC estimating 2.4 million cases of campylobacteriosis each year in the US. [1]. Clinical signs include a prodrome of fever, abdominal pain, and malaise followed by the onset of profuse, watery diarrhea with blood, mucus, and leukocytes [2]. Microscopic examination of colonic tissues from humans and experimentally infected animals demonstrates that C. jejuni infection stimulates an acute inflammatory response with infiltration of the epithelium and lamina propria with neutrophils and mononuclear cells [3-5]. Immunohistochemical staining of colonic tissues shows C. jejuni associated with epithelial cells and goblet cells, within deep crypts of the colon, and within lymphoglandular complexes (LGCs) in entrapped crypts and in the follicle associated epithelium [3, 4]. The pathogenesis of C. jejuni infection involves colonization of the intestinal mucous overlying the epithelium, adherence to intestinal epithelial cells, and invasion of epithelial cells [2, 5]. C. jejuni is chemoattracted to mucin, a major component of intestinal mucous, and polar flagella combined with its spiral morphology facilitate efficient movement through the mucous to come into contact with the epithelium [2, 6, 7]. C. jejuni binds to epithelial cells, intercellular junctions, and the basement membrane via adhesins, including CadF, le, CBF-l, and flagella [8-13]. Bound bacteria are internalized by microtubule- and microfilament-dependent processes [14-16]. Epithelial cell destruction can occur due to the effects of invasion and toxins produced by C. jejuni, including enterotoxin, cytotoxins, and cytolethal distending toxin 111 [17-21]. Cellular destruction and loss of epithelial function leads to decreased absorption which manifests clinically as diarrhea [2, 5]. Elimination of the bacteria and resolution of disease is dependent upon induction of a humoral immune response and cytokine production. C. jejuni stimulates specific serum IgG as well as secretory IgA in feces, urine, saliva, and mammary glands [22]. In a swine model of infection, anti-C. jejuni IgA has been localized to B cell rich germinal centers of colonic lymphoglandular complexes [23]. Oral pretreatment of mice with recombinant IL-5 or IL-6 prior to oral challenge with C. jejuni significantly reduces intestinal colonization. In addition, recombinant IL-6 enhances specific intestinal and systemic IgA production [24]. In vitro, C. jejuni stimulates secretion of IL-IB, TNFa, IL-6, and IL-8 from intestinal epithelial cells and monocytes [25-27] (Parthasarathy and Mansfield, unpublished data; Cunningham and Mansfield, unpublished data). These studies indicate that a humoral immune response is necessary for resolution of infection and that proinflammatory cytokines may be important for host resistance to C. jejuni infection in viva. IL-18 is a pleiotrophic cytokine capable of initiating and regulating both innate and adaptive immune responses in viva [28-31]. Mouse peritoneal macrophages and human peripheral blood mononuclear cells produce TNFa, IL-la, IL-1 [3, IL-6, IL-8, and MCP-l upon stimulation with IL-18 [32, 33]. Naive T cells proliferate and produce IFNy when stimulated with IL-12 and IL-18 [29, 34, 35]. NK cell cytotoxicity and Th 1 cell differentiation are enhanced by stimulation with IL-12 and IL-18 [29, 34, 35]. In the presence of lL-2, IL-18 stimulates IL-13 production from NK cells and T cells [35]. IL- 18 is produced by a variety of cell types and is widely distributed through tissues in viva 112 [35, 36]. IL-18 is both transcriptionally and post translationally regulated. The IL-18 promoter region is responsive to lipopolysaccharide (LPS) and increased mRNA expression can be induced in LPS stimulated swine macrophages and splenocytes in vitro [36, 37]. Immature IL-18 protein requires cleavage by IL-lB converting enzyme (ICE), also known as Caspase-l, for activation and secretion from cells [38]. In addition, IL-18 binding protein regulates IL-18 activity in viva [39]. IL-l8 mRNA and protein are constitutively expressed in a variety of cell types, including intestinal cells, suggesting a role for IL-18 as a first line of host defense against pathogens [3 5]. IL-18 is constitutively expressed in intestinal tissues and plays a critical role in initiating an immune response to pathogenic bacteria [34, 35, 40, 41]. Also, as discussed in Chapter 2, C. jejuni stimulates significant expression of lL-18 and other proinflammatory cytokines in the distal colon and lymphoglandular complexes 1 hour after rectal inoculation. Because enterocytes are the first line of defense against enteric bacteria, we hypothesized that intestinal epithelial cells are a source of IL-18 during C. jejuni infections. To test this hypothesis, differentiated and undifferentiated cultured Intestinal Pig Epithelial Cells (IPEC-l) were infected with C. jejuni for O, l, 3, 6, 8, and 24 hours. IL-18 production was analyzed using two methods. IL-18 secretion in culture supematants was measured by ELISA. IL-l8 mRNA expression in cells was measured by RT-PCR. Differentiated cells secreted significant levels of IL-18 protein as early as 3 hours after infection. Undifferentiated cells did not secrete significant levels of IL-18 at any time point. Expression of IL-1 8 mRNA was not significantly increased by C. jejuni at any time from either differentiated or undifferentiated cells. These results indicate that 113 differentiated intestinal epithelial cells are a significant source of IL-l8. Epithelial cell- derived IL-18 may be important for host resistance to C. jejuni infection in viva. I“ ‘fliK.-.’- .l I. 114 Materials and Methods Cells IPEC-l is a non-immortalized undifferentiated cell line derived from the small intestine of neonatal pigs. These cells will differentiate, developing tight junctions and distinct apical and basolateral surfaces, when grown on porous supports for 10 to 14 days [42]. The transepithelial electrical resistance (TER) can be measured across the monolayer to indicate whether the cells have achieved a differentiated state. Differentiated IPEC-l cells represent the mature enterocytes located on the villus tips in the intestines. When IPEC-l cells are seeded at the appropriate density onto flat bottom polystyrene plates and grown for only 2-3 days, the cells form confluent monolayers that remain undifferentiated. Undifferentiated cells represent the rapidly dividing cells found in the base of the crypts in the intestine. We used IPEC-l cells in both differentiated and undifferentiated states to determine if IL-18 production was stimulated upon C. jejuni infection. All culture media and growth supplements were obtained from Invitrogen unless otherwise stated (Invitrogen, Frederick, MD). Growth flasks and transwells were obtained from Corning (Costar, Corning, NY). Cells were routinely cultured at 37°C, 5% C02 in DMEM/F12 supplemented with 5% Fetal Bovine Serum (FBS) and 1% Insulin- Transferrin-Selenium (ITS) in T75 flasks. To seed cells for infection studies, the cells were gently washed with versene and released from the flask with 0.05% Trypsin-EDTA. Detached cells were collected and pelleted by centrifugation at 1200rpm for 5 minutes. 115 The supernatant was removed and the cells were resuspended in RPMI supplemented with 5%FBS. A subsarnple of the cells was mixed with a 1:1 volume of trypan blue (Sigma, St. Louis, MO), then loaded onto a hemocytometer (Hausser Scientific, Horsharn, PA) and viable cells were counted on a Nikon inverted microscope (Mager Scientific, Dexter, MI). Cells to be tested in a differentiated state were seeded at a density of 3x105 cells onto 6.5mm diameter, 3.0um pore size transwell inserts which had been previously coated with 20ug/mL fibronectin (Sigma, St. Louis, MO). Cells were incubated for 12- 14 days to allow differentiation. Prior to infection TER was measured from each well using an electrode to confirm confluency and tight junction formation (EVOMX, World Precision Instruments, Sarasota, FL). TER values greater than 6OOQ/cm2 indicated that the cells were differentiated. Cells to be infected in the undifferentiated state were seeded at a density of 3x105 cells per well onto 24 well plates that had previously been coated with 20ug/mL of fibronectin (Sigma, St. Louis, MO). The cells were incubated for 2-3 days to allow formation of a confluent monolayer. Scanning Electron Microscopy Differentiated IPEC-l cells grown on transwells for 12 days were examined by scanning electron microscopy to verify their state of differentiation. The cells were seeded onto transwells and incubated for 12 days as previously described. The medium was removed, the membrane was excised, and the cells were fixed in 4% glutaraldehyde in 0.1M sodium phosphate buffer. The cells were further processed and examined at the 116 Center for Microscopy at Michigan State University (East Lansing, MI). The cells were fixed in 1% osmium tetroxide, dehydrated through and ethanol gradient, then dried in a critical point dryer (Balzers, Lichenstein). Dehydrated membranes were mounted on aluminum stubs and sputter coated with 7nm of gold (Emscope SCSOO sputter coater, UK). The cells were visualized on a scanning electron microscope (JEOL 64OOV, Japan). Bacteria Campylobacterjejuni strains ATCC 33292 and 81-176 were used for infections of [PEG] cells. Both strains were originally isolated from humans with enteritis. The C. jejuni strain ATCC 33292 used in these infections had been rederived by passage through a pig in our laboratory and stored as second passage stocks. C. jejuni strain 81-176 was kindly donated by Carol Pickett. Bacterial cultures were streaked for isolation onto Bolton Agar plates and incubated for 48 to 72 hours at 37°C, 5% C02. Isolated colonies were subcultured for lawn growth and grown for approximately 20 hours. Lawns were harvested with sterile swabs and resuspended in RPMI without phenol red or FBS. The OD560 was measured on a Beckman DU 530 UVN is spectrophotometer (Beckman Coulter, Inc, Fullerton, CA) and subsequently adjusted to an OD560 of 0.1 in RPMI. Previous studies in our lab have shown that this measurement corresponds to approximately 5x108 cfu/mL. A subsarnple of each inoculum was examined microscopically to assess morphology and darting motility of the bacteria. 117 Experimental Design To determine the effect of C. jejuni on IL-18 secretion, IPEC-l cells were exposed to each strain of bacteria apically at a multiplicity of infection (MOI) of approximately 40:1. Previous studies in our laboratory have shown this to be the optimal concentration of C. jejuni for stimulation of IL-6 and IL-10 secretion from IPEC-l cells. RPMI without phenol red or F BS was used as the negative control and Concanavalin A (Con A) was used at a concentration of lOOug/mL as the positive control. Supematants were removed at O, l, 3, 6, 8, or 24 hours after infection then centrifuged for 10 minutes at 6000rpm and 4°C to pellet bacteria. The supematants were transferred to polypropylene tubes (DOT Scientific, Burton, MI) and stored at -80°C until ELISA was performed to measure the concentration of IL-18 protein. For differentiated IPEC-l cells, supematants from the upper and lower chamber were stored and analyzed separately to determine if IL-18 was secreted from the apical or basolateral surface. Expression of IL- 18 mRNA was measured from IPEC-l cells in both differentiated and undifferentiated states by RT-PCR of total RNA extracted from cells infected for 8 hours. ELISA Secreted IL-18 protein was measured from culture supematants by an indirect ELISA developed in our laboratory. Recombinant porcine IL-18 and anti-swine IL-18 primary antibody were obtained from R and D systems (Minneapolis, MN). Biotin conjugated anti-goat IgG secondary antibody, Extravidin®, bovine serum albumin (BSA), and the ready to use peroxide containing tetramethyl benzidine (TMB) substrate were obtained from Sigma (Sigma, St. Louis, MO). ELISA plates were washed 3 times 118 with Tris-buffered saline with 0.5% Tween-20 (TTBS) between incubations and all incubations were for 1 hour at room temperature unless otherwise indicated. Prior to measuring IL-18 from experimentally infected IPEC-l cells, the optimal concentrations for the primary antibody, secondary antibody, and Extravidin were determined experimentally (data not shown). For preparation of the standards, recombinant porcine IL-18 was diluted in RPMI 1640 without phenol red to lOOng/mL, SOng/mL, 25ng/mL, 12.5ng/mL, 6.25ng/mL, 3.125ng/mL, and 1.56ng/mL final concentrations. Supematants and standards were coated in duplicate onto Nunc Immuno Maxisorp 96 well plates (V WR, West Chester, PA) overnight at 4°C. Plates were blocked for 90 minutes with 3% BSA at 37°C, then bound IL-18 was detected by 2 hour incubation with anti-swine IL-1 8 antibody diluted to 750ng/mL in 1%BSA in TTBS. Primary antibody was detected by biotin conjugated secondary antibody diluted 1:10,000 in 1%BSA in TTBS. The biotin conjugate was reacted with Extravidin® diluted to lug/mL in 1%BSA in TTBS, followed by color development for approximately 15 minutes with the peroxide containing TMB substrate. Color development was stopped by addition of 6N H2804 after approximately 15 minutes. Absorbance was measured at 450nm on an EL8OO Universal Microplate Reader and the concentration of IL-18 protein in each sample was calculated by KCjunior® software (Bio-Tek instruments, Winooski, Vermont). Cytokine expression Total RNA was isolated from IPEC-l cells using the Trizol® method. Briefly, cells were lysed in Trizol® reagent (Invitrogen, Frederick, MD), RNA was extracted with chloroform, precipitated with isopropanol, and washed with ethanol. Pellets were air 119 17.3.,7 dried, then resuspended in DEPC-treated H20 (Invitrogen, Frederick, MD). Concentration of RNA was determined spectrophotometrically (Beckman). Samples were checked for degradation by electrophoresis on a 1.8% agarose gel in 1X TAE buffer with 0.2ug/mL ethidium bromide. Gels were visualized by UV illumination on an Alpha Imager 2000 Documentation and Analysis system (Alpha Innotech Corporation, San Leandro, CA). RNA samples were stored at -80°C. For each sample, 5ug of total RNA was reverse transcribed with Oligo dT primers using a Stratagene (Stratagene, La Jolla, CA) First Strand RT-PCR kit. First strand cDNA was used as the template in further PCR reactions Swine IL-18 and Bz-microglobulin expression was measured from IPEC-l cDNA samples using previously described primers [35, 43]. The primer sequences for IL-18 were forward 5' TAT GCC TGA TI‘C TGA CTG TT 3’ and reverse 5' ATG AAG ACT CAA ACT GTA TCT 3' [3 5]. The primer sequences for Bz-microglobulin were forward 5' CTG CTC TCA CTG TCT GG 3', and reverse 5' ATC GAG AGT CAC GTG CT 3' [43]. For each target, a 25 “L PCR reaction consisted of IX Perkin Elmer PCR buffer (Tris-HCL, pH 8.3, 50 mM KCI), 1.0mM MgClz, 0.05mM each dNTP, 37.5ng of each primer, 0.625U Perkin Elmer AmpliTaq Gold, and luL of first strand cDNA in DEPC treated water. Each sample was amplified in triplicate. Prior to measuring expression in our sample set, the optimal number of amplification cycles for each target was determined for measurement of expression during the exponential phase of amplification. We measured expression from positive and negative controls every 2 cycles from 20 to 40 cycles and plotted the values on a graph (Figures 5 and 6). All samples were amplified with a 10 minute hot start at 94°C followed 120 i. 71“.“ haul by 39 cycles of 94°C for 1 minute, 55°C for 1 minute, 72°C for 1 minute, with a final extension time of 10 minutes at 72°C. The PCR product was electrophoresed on 1.8% agarose gels in 1X TAE with 0.2ug/mL ethidium bromide. The gels were visualized by UV illumination on an Alpha Imager 2000 Documentation and Analysis system, and density of individual bands was measured using the Line Densitometry fimction. Bz-microglobulin was used as the constitutively expressed gene for normalization of cytokine expression. Ratios of specific cytokine to housekeeping gene were calculated by dividing the density values obtained from the IL-18 target by those obtained from the Bz-microglobulin target [3 5]. Statistical Analysis The data were analyzed in Excel (Microsoft, Redmond, WA). An F test for sample variance was performed to compare means of infected cells to uninfected cells. The appropriate T test was then used to determine the effect of treatment on IL-18 production at each time point. P values of 0.05 or less were considered statistically significant. 121 Results Scanning Electron Microscopy (SEM) A representative SEM image of IPEC-l cells grown on transwells for 12 days is shown in figure 1. TER values measured from 12 day old IPEC-l cells ranged from 600 to 800 Q/cmz. The cell monolayers were confluent and cells had developed microvilli on their apical surfaces, indicating that they had achieved a differentiated state. IL-l8 secretion IPEC-l cells in a differentiated state secreted significant amounts of IL-18 into the culture supernatant by 3 hours after C. jejuni infection (Figure 2). In the upper chamber, secretion was virtually undetectable immediately after infection (0 hr) and 1 hour after infection. Secretion peaked at approximately 12 ng/mL 3 hours after infection, and remained elevated at both 6 and 8 hours after infection. By 24 hours, IL-18 was still detectable, but the levels had decreased significantly. In the lower chamber, lL-l8 secretion was very low and was not significantly induced by C. jejuni infection at any time point (Figure 3). IL-18 secretion in response to C. jejuni infection was not significantly different between strains. In contrast, IPEC-l cells in an undifferentiated state did not secrete significant amounts of IL-18 in response to C. jejuni at any time (Figure 4). 122 'Afi._ml l' IL-18 mRNA expression IL-18 expression was measured by RT—PCR from IPEC-l cells in both the differentiated and undifferentiated state 8 hours after infection with C. jejuni. Prior to measuring expression from experimental samples, the optimal cycle number to use was determined by measuring the density of the IL-18 and BZ-microglobulin targets amplified from stimulated (lOOng each of Calcium ionophore and phorbol myristate acetate) and unstimulated cells (DMEM or RPMI medium only) over several cycles (Figure 5). The optimal cycle number for IL-18 measurement from experimental samples was selected from the linear portion of the amplification curve for each target (Figure 6). The optimal cycle numbers selected for measurement of target expression from experimental samples were 31 for IL-18 and 27 for BZ-microglobulin. IL-18 and BZ-microglobulin targets were amplified from IPEC-l cells in both differentiation states with the exception of IPEC-l cells in the differentiated state treated with Con A (data not shown). IL-18 expression was not significantly impacted by C. jejuni infection at any time point in IPEC-l cells in either differentiation state (Figure 7). 123 Discussion We hypothesized that intestinal epithelial cells infected with C. jejuni are a source of IL-18. Furthermore, early release of IL-18 from epithelial cells assaulted by C. jejuni may serve to initiate a protective inflammatory response in viva. Our experiments show that C. jejuni stimulates lL-18 secretion from IPEC-l cells that have achieved a differentiated state. Secretion peaked at 3 hours and remained elevated at 6 and 8 hours after infection before decreasing significantly by 24 hours after infection. In vitro, C. jejuni stimulates secretion of a panel of proinflammatory cytokines from intestinal epithelial cells and monocytes, including IL-IB, TNFa, IL-6, and IL-8 [25-27] (Parthasarathy and Mansfield, unpublished data; Cunningham and Mansfield, unpublished data). Epithelial cell-derived cytokines play an important role in controlling the spread of pathogenic bacteria in viva [44]. IL-18 facilitates neutrophil accumulation in tissues of mice during lethal endotoxemia, and it has also been shown to activate neutrophils, inducing granule release and enhancing the respiratory burst [45, 46]. Neutrophil infiltration of the lamina propria and submucosa is a consistent finding in animal models of C. jejuni infection [3, 4, 47]. The effects of IL-18 on neutrophil functions likely serve to kill invading C. jejuni and contain the infection. IL-18 also stimulates production of IL-8, TNFa, and IL-IB, which would sustain the inflammatory response and activate other mechanisms for eliminating bacterial pathogens [32, 33, 45, 46]. IL-18 secretion from differentiated IPEC-l cells was apparently not transcriptionally regulated. IL-18 mRNA was constitutively expressed in these cells, but 124 expression was not significantly increased by infection with C. jejuni. The IL-18 promoter sequence contains LPS response elements and IL-18 expression is induced in LPS stimulated swine alveolar macrophages, adherent splenocytes, and non adherent splenocytes [36, 37]. Differentiated IPEC-l cells stimulated with 200ug/mL of LPS fiom E. coli 026:B6 secrete significant amounts of IL-6 (Parthasarathy et al, unpublished results), but not IL-18 (data not shown). This concurs with data from Okazawa and colleagues who found that LPS stimulation of the human colonic cell line HT-29 did not result in increased intracellular accumulation of IL-18 or increased secretion [48]. These data indicate that LPS-induced transcriptional regulation does not increase production of IL-18 from intestinal epithelial cells. Constitutive expression of IL-18 protein in the intestine suggests that rapid production of IL-18 is important for early responses to pathogens [35, 36]. Therefore, it is likely that post translational modification of IL-l8, via cleavage with Caspase-l, rather than increased expression of mRNA, is the main mechanism for IL-18 secretion in the intestine. The data from differentiated IPEC-l cells support that hypothesis. In contrast to differentiated IPEC-l cells, C. jejuni infection did not stimulate increased production of IL-18 protein or mRNA from undifferentiated IPEC-l cells at any time assayed over 24 hours. Similar to differentiated cells, undifferentiated cells express IL-18 mRNA constitutively (data not shown). Undifferentiated cells are capable of secreting 7ng/mL of IL-18 after 8 hours of stimulation with Con A (Figure 4). It has been demonstrated that undifferentiated IPEC-l cells secrete IL-10 in response to Con A, but not in response to C. jejuni, while differentiated IPEC-l cells do secrete lL-10 in response to C. jejuni infection (Parthasarathy et al, unpublished results). IL-1 [3 stimulated 125 differentiated HT-29 human intestinal epithelial cells have impaired production of IL-8 when compared to undifferentiated cells. This effect is due to decreased c-Jun N-terminal kinase (JNK) and MB kinase (IKK) activity with subsequently reduced NF -KB DNA binding after stimulation of the IL-1 [3 receptor [49]. Impaired secretion of IL-18 (and IL- 10) from undifferentiated IPEC-l cells may be due to altered intracellular signaling pathways, but further study is necessary to elucidate this. IL-l8 mRNA and immature protein are constitutively expressed in swine intestinal epithelial cells and gut associated lymphoid tissues throughout the intestine [35, 36]. Salmonella, Shigella, Yersenia, and Helicobacter infections induce increased expression of IL-18 mRNA and maturation of IL—18 protein to the active form, which are critical for resolution of infection [34, 36, 41, 50-52]. We have also shown, as discussed in Chapter 2, that C. jejuni induces IL-18 and other proinflammatory cytokines in the distal colon and lymphoglandular complexes of rectally inoculated pigs. We propose that in our swine model of C. jejuni infection, IL-l8 released from C. jejuni infected epithelial cells stimulates production of IL-IB, TNFOL, IL-6, IL-8, and MCP-l from epithelial cells and leukocytes locally, inducing an inflammatory response. Also, neutrophil infiltration of the epithelium and lamina propria with subsequent activation of their bacterial killing mechanisms by IL-18 may be critical for reducing pathogen numbers. 126 10. ll. l2. l3. 14. 15. 16. References Allos, B.M., Campylobacterjejuni Infections: update on emerging issues and trends. Clin Infect Dis, 2001. 32(8): p. l201-6. Ketley, J.M., Pathogenesis of enteric infection by Campylobacter. Microbiology, 1997. 143 (Pt I): p. 5-21. Babakhani, F.K., G.A. Bradley, and LA. Joens, Newborn piglet model for campylobacteriosis. Infect Immun, 1993. 61(8): p. 3466-75. Mansfield, L.S., et al., Enhancement of disease and pathology by synergy of Trichuris suis and Campylobacterjejuni in the colon of immunologically naive swine. Am J Trop Med Hyg, 2003. 68(3): p. 70-80. Wassenaar, T.M. and M.J. Blaser, Pathophysialogy of C ampylobacter jejuni infections of humans. Microbes Infect, 1999. 1(12): p. 1023-33. Hugdahl, M.B., J.T. Beery, and M.P. Doyle, C hemotactic behavior of Campylobacter jejuni. Infect Immun, 1988. 56(6): p. 1560-6. Szymanski, C.M., et al., Campylobacterjejuni motility and invasion of Caca-2 cells. Infect Immun, 1995. 63(11): p. 4295-300. J in, S., et al., leA, a novel surface-exposed lipoprotein specific to Campylobacterjejuni, mediates adherence to host epithelial cells. Mol Microbiol, 2001. 39(5): p. 1225-36. J in, S., et al., leA of Campylobacter jejuni interacts with surface-exposed heat shock protein 90alpha and triggers signalling pathways leading to the activation of NF -kappaB and p38 MAP kinase in epithelial cells. Cell Microbiol, 2003. 5(3): p. 165-74. Konkel, M.B., et al., Identification and molecular cloning of a gene encoding a fibronectin- binding protein (CadF) from Campylobacterjejuni. Mol Microbiol, 1997. 24(5): p. 953-63. McSweegan, E. and RJ. Walker, Identification and characterization of two Campylobacterjejuni adhesinsfor cellular and mucous substrates. Infect Immun, 1986. 53(1): p. 141-8. Monteville, M.R., J .E. Yoon, and ME. Konkel, Maximal adherence and invasion of IN T 407 cells by Campylobacterjejuni requires the CadF outer-membrane protein and microfilament reorganization. Microbiology, 2003. 149(Pt l): p. 153-65. Pei, Z., et al., Mutation in the peblA locus of Campylobacter jejuni reduces interactions with epithelial cells and intestinal colonization of mice. Infect Immun, 1998. 66(3): p. 93 8—43. Biswas, D., K. Itoh, and C. Sasakawa, Uptake pathways of clinical and healthy animal isolates of Campylobacterjejuni into INT-407 cells. FEMS Immunol Med Microbiol, 2000. 29(3): p. 203-1 1. Hu, L. and DJ. Kopecko, Campylobacterjejuni 81-1 76 associates with microtubules and afvnein during invasion of human intestinal cells. Infect Immun, 1999. 67(8): p. 4171-82. Oelschlaeger, T.A., P. Guerry, and DJ. Kopecko, Unusual microtubule-dependent endocytosis mechanisms triggered by C ampylobacter jejuni and C itrabacter freundii. Proc Natl Acad Sci U S A, 1993. 90(14): p. 6884-8. 127 17. I8. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. Aragon, V., K. Chao, and LA. Dreyfus, Eflect of cytolethal distending toxin on F -actin assembly and cell division in Chinese hamster ovary cells. Infect Immun, 1997. 65(9): p. 3774-80. Johnson, W.M. and H. Lior, Cytotoxic and cytotonic factors produced by Campylobacterjejuni, Campylobacter coli, and Campylobacter laridis. J Clin Microbiol, 1986. 24(2): p. 275-81. Klipstein, F.A. and RF. Engert, Properties of crude Campylobacterjejuni heat-labile enterotoxin. Infect Immun, 1984. 45(2): p. 314-9. Lara-Tejero, M., A bacterial toxin that causes DNA damage to modulate cellular responses. ScientificWorldJoumal, 2001. l: p. 190-1. Lee, R.B., et al., Interactions of Campylobacter jejuni cytolethal distending toxin subunits C th and CdtC with HeLa cells. Infect Immun, 2003. 71(9): p. 4883-90. Cawthraw, S.A., et al., Long-term antibody responses following human infection with Campylobacterjejuni. Clin Exp Immunol, 2002. 130(1): p. 101-6. Mansfield, LS. and D.T. Gauthier, Lymphoglandular complexes are important colonic sites for immunoglobulin A induction against Campylobacterjejuni in a swine disease model. Comp Med, 2004. 54(5): p. 514-23. Baqar, 8., ND. Pacheco, and F .M. Rollwagen, Modulation of mucosal immunity against Campylobacterjejuni by orally administered cytokines. Antimicrob Agents Chemother, 1993. 37(12): p. 2688-92. Hickey, T.E., et al., Campylobacterjejuni-stimulated secretion of interleukin-8 by IN T407 cells. Infect Immun, 1999. 67(1): p. 88-93. Hickey, T.E., et al., Campylobacterjejuni cytolethal distending toxin mediates release of interleukin-8 from intestinal epithelial cells. Infect Immun, 2000. 68(12): p. 6535-41. Jones, M.A., et al., Induction of proinflammatory responses in the human monocytic cell line THP-I by Campylobacterjejuni. Infect Immun, 2003. 71(5): p. 2626-33. Akira, S., The role of IL-1 8 in innate immunity. Curr Opin Immunol, 2000. 12(1): p. 59-63. Dinarello, C.A., et al., Overview of interleukin-I 8: more than an interferon-gamma inducing factor. J Leukoc Biol, 1998. 63(6): p. 658-64. Gracie, J.A., S.E. Robertson, and LB. Mclnnes, Interleukin-I8. J Leukoc Biol, 2003. 73(2): p. 213-24. Nakanishi, K., et al., Interleukin-I8 is a unique cytokine that stimulates both Th1 and Th2 responses depending on its cytokine milieu. Cytokine Growth Factor Rev, 2001. 12(1): p. 53-72. Netea, M.G., et al., Interleukin-I8 induces production of proinflammatory cytokines in mice: no intermediate role for the cytokines of the tumor necrosis factor family and interleukin-I beta. Eur J Immunol, 2000. 30(10): p. 3057-60. Puren, A.J., et al., Interleukin-I8 (IFNgamma-inducing factor) induces IL-8 and IL-I beta via INF alpha production fi'om non-CDI4+ human blood mononuclear cells. J Clin Invest, 1998. 101(3): p. 711-2]. Biet, F., C. Locht, and L. Kremer, Immunoregulatoryfunctions of interleukin I8 and its role in defense against bacterial pathogens. .1 Mol Med, 2002. 80(3): p. 147-62. 128 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. Fournout, S., et al., Cloning, chromosomal location, and tissue expression of the gene for pig interleukin-I8. Immunogenetics, 2000. 51(4-5): p. 358-65. Foss, D.L., M.J. Zilliox, and M.P. Murtaugh, Bacterially induced activation of interleukin-1 8 in porcine intestinal mucosa. Vet Immunol Immunopathol, 2001. 78(3-4): p. 263-77. Kim, Y.M., et al., Roles of IFN consensus sequence binding protein and PU. I in regulating IL-I8 gene expression. J Immunol, 1999. 163(4): p. 2000-7. Dinarello, C .A., Interleukin-I beta, interleukin-I8, and the interleukin-I beta converting enzyme. Ann N Y Acad Sci, 1998. 856: p. 1-1 I. Dinarello, C.A., Novel targets for interleukin I8 binding protein. Ann Rheum Dis, 2001. 60 Suppl 3: p. iiil8-24. Fox, J.G., et al., Gastroenteritis in NF -kappaB-deficient mice is produced with wild-type Camplyobacterjejuni but not with C. jejuni lacking cytolethal distending toxin despite persistent colonization with both strains. Infect Immun, 2004. 72(2): p. 1116-25. Sansonetti, P.J., et al., Caspase-l activation of IL-I beta and IL-18 are essential for Shigella flexneri-induced inflammation. Immunity, 2000. 12(5): p. 581-90. Abner, S.R., et al., Response of intestinal epithelial cells to Trichuris suis excretory-secretory products and the influence on Campylobacterjejuni invasion under in vitro conditions. J Parasitol, 2002. 88(4): p. 738-45. Reddy, N.R., B.N. Wilkie, and B.A. Mallard, Construction of an internal control to quantitate multiple porcine cytokine mRNAs by RT-PCR. Biotechniques, 1996. 21(5): p. 868-70, 872, 875. Sansonetti, P.J., et al., Interleukin-8 controls bacterial transepithelial translocation at the cost of epithelial destruction in experimental shigellosis. Infect Immun, 1999. 67(3): p. 1471-80. Leung, B.P., et al., A role for IL-I8 in neutrophil activation. J Immunol, 2001. 167(5): p. 2879-86. Netea, M.G., et al., Neutralization of IL-1 8 reduces neutrophil tissue accumulation and protects mice against lethal Escherichia coli and Salmonella typhimurium endotoxemia. J Immunol, 2000. 164(5): p. 2644-9. F ox, J .G., et al., Campylobacterjejuni infection in the ferret: an animal model of human campylobacteriosis. Am J Vet Res, 1987. 48(1): p. 85-90. Okazawa, A., et al., Human intestinal epithelial cell-derived interleukin (IL)-I8, along with IL-2, IL-7 and IL-1 5, is a potent synergistic factor for the proliferation of intraepithelial lymphocytes. Clin Exp Immunol, 2004. 136(2): p. 269-76. Bocker, U., et al., Cellular diflerentiation causes a selective down-regulation of interleukin (IL)- ! beta-mediated NF -kappaB activation and IL-8 gene expression in intestinal epithelial cells. J Biol Chem, 2000. 275(16): p. 12207-13. Elhofy, A. and K.L. Bost, Limited interleukin-I8 response in Salmonella-infected murine macrophages and in Salmonella-infected mice. Infect Immun, 1999. 67(10): p. 5021-6. Fera, M.T., et al., Correlation between Helicobacter pylori infection and IL-I8 mRNA expression in human gastric biopsy specimens. Ann N Y Acad Sci, 2002. 963: p. 326-8. 129 52. Tomita, T., et al., Expression of Interleukin-I 8, a Th] cytokine, in human gastric mucosa is increased in Helicobacter pylori infection. J Infect Dis, 2001. 183(4): p. 620-7. 130 Figure 1: Scanning electron micrograph of polarized IPEC-l cells (12 days) on transwell membranes. Top panel is overview of cells; bar represents 20 pm. The apical finger-like microvilli are seen in the bottom panel; bar represents 2 pm. Micrographs are representative of three samples each. I31 lL-18 secretion from lPEC-1 cells- upper chamber nglmL a IOI'Ir I1hr m3hr 56hr EB” .2417 81-176 3292 RPMI Figure 2: IL-18 secretion from differentiated IPEC-l cells. Apical secretion of IL-18 in response to apical infection with C. jejuni was measured by ELISA from the upper chamber of differentiated cells. Secretion from C. jejuni infected cells was significantly greater than medium only controls at 3, 6, 8, and 24 hours afier infection. (p50.05). 132 IL-18 secretion from IPEC-1 cells- lower chamber 30 25 - ~— 20 15 --————- —-——*fi-~ nglmL 10 ~—- , ,, 81-176 3292 1.--. _ , J II. \ Figure 3: IL-18 secretion from differentiated IPEC-l cells. Basolateral secretion of IL-18 in response to apical infection with C. jejuni was measured by ELISA from the lower chamber of differentiated cells. Secretion from C. jejuni infected cells was not significantly greater than medium only controls at any time after infection. (pS0.05). 133 30 25 ngImL 1 lL-18 secretion from lPEC-1 cells: undifferentiated cells 2O «— 15~ 1o W IIIIII S\\\\ ConA 81 -1 76 33292 RPMI um] Figure 4: IL-1 8 secretion from undifferentiated IPEC-l cells. IL-18 secretion in response to C. jejuni infection of undifferentiated IPEC-l cells was measured by ELISA. Undifferentiated IPEC-l cells secreted an average of 7ng/mL of IL-1 8 in response to Concanavalin A, however IL-1 8 secretion from C. jejuni infected cells was not significantly greater than medium only controls at any time after infection. (p50.05). 134 1r— Figure 5: Optimization of amplification parameters. We titrated the cycle number needed to measure IL-18 and B2-microglobulin expression within the linear portion of the amplification curve. Samples were amplified in triplicate, electrophoresed on an agarose gel, and density of the bands was measured. Panel A: Cycle titration for IL-18 expression. Lane 1: 100bp DNA ladder; Lanes 2-4: 28 cycles; Lanes 5-7: 30 cycles; Lanes 8-10: 32 cycles; Lanes 11-13: 34 cycles; Lanes 14-16: 36 cycles; Lanes 17-19: 38 cycles. Panel B : Cycle titration for BZ-microglobulin expression. Lane 1: 100bp DNA ladder; Lanes 2-4: 24 cycles; Lanes 5-7: 26 cycles; Lanes 8-10: 28 cycles; Lanes 1 l-13: 30 cycles; Lanes 14-16: 32 cycles; Lanes 17-18: 34 cycles. 135 Cycle titration for IL-18 expression 1400 1200 1000 g 000 i a 600 400 200 0 Cycle titration for 32- microglobulin 1600.0 1400.0 1200.0 1000.0 4 W I 800 o 7 +CVPMA 1 ‘ ‘ ,+, ,fll 6000 4000 2000 0.0 24 26 20 30 32 34 Cycle Nunber Figure 6: Determination of optimal number of cycles for amplification. The average density of the bands from each cycle was plotted on a graph. The optimal cycle number for measurement of target expression from experimentally infected samples was chosen from the linear portion of the amplification curve. IL-18 expression was measured at 31 cycles while BZ-microglobulin expression was measured at 27 cycles. 136 lL-18 expression from differentiated lPEC-1 0.4 -* ~‘ lL-18zb2 ratio 0.2 - 7-7 0.8+ 7 ,fi 7- w L, -7 ,, 7- ,7— 0.6 -- 7 - —— 7 if ,, . Con A 81 -1 76 33292 RPM I lL-18 expression from undifferentiated cells Ratio lL-1B:82-M CVHM B1-176bep 33292 Figure 7: IL-18 expression from IPEC-l cells. IL-18 expression was measured 8 hours after infection from both differentiated and undifferentiated IPEC-l cells. The data from each sample was normalized to BZ-microglobulin expression and is presented as the ratio of IL-18 expression over to BZ-microglobulin expression. Neither IL-18 nor to [32- microglobulin were measured from Con A stimulated differentiated cells. IL-1 8 expression was not significantly changed in C. jejuni infected cells when compared to medium only controls for either cell type. 137 g Chapter 5: Summary and Conclusions Pigs are an excellent animal model for studying C. jejuni infection in humans for several reasons. Physiologically, the swine digestive tract is similar to humans and outbred populations that are heterogeneous, similar to the human population, are readily available at low cost from production facilities. Colostrum-deprived piglets become colonized and develop mucohemorrhagic diarrhea and intestinal pathology that mimics human disease after oral inoculation with C. jejuni [1, 2]. In addition, an extensive array of swine specific reagents is commercially available for the analysis of immune mediators. All together, these facts make pigs quite suitable for use in research on the pathogenesis of C. jejuni infection. We used a dual infection model of concurrent C. jejuni and T. suis infection in pigs to study the immune responses associated with mucohemorrhagic diarrhea. Campylobacter jejuni is a commensal in the colons of healthy pigs, but upon infection with the whipworm Trichuris suis, these two agents synergize to produce mucohemorrhagic diarrhea and severe pathology [2-4]. The long term goals of the laboratory are to elucidate the mechanisms by which dual infection with these two agents causes severe disease and pathology. One hypothesis is that T. suis infection facilitates C. jejuni invasion by modulation of the host immune response. The central hypothesis for this dissertation project is that a proinflammatory response plays a role in swine natural resistance to C. jejuni. A related hypothesis is T. suis infection alters the protective proinflammatory response resulting in C. jejuni invasion in the colon and severe pathology. These hypotheses were tested, generally, by assessing local cytokine expression in the intestines of C. jejuni and T. suis infected pigs, and more specifically by 138 assessing IL-18 secretion from C. jejuni infected IPEC-l cells, which serve as a model system for the swine intestinal epithelium. This dissertation project was designed to address three main objectives. The first objective was to determine the nature of the local immune response to C. jejuni in the distal colon and LGC of pigs. To address this issue, weaned piglets were inoculated rectally with C. jejuni 81-176 that produced Gfp. The pigs were killed 1 hour or 24 hours after inoculation and samples from the distal colon and LGCs were taken for microscopic examination for lesions, for immunohistochemical staining with anti-C. jejuni antibody to localize the bacteria in the colon, and for measurement of cytokine mRNA expression by real time PCR. The second objective was to determine the nature of the local immune response in different segments of the intestine when pigs were infected with C. jejuni, T. suis, or both. To address this issue, weaned piglets were inoculated by oral gavage with embryonated T. suis eggs. Twenty one days after infection with T. suis, pigs were inoculated by oral gavage with C. jejuni strain ATCC 33292. Two days after inoculation with C. jejuni, all pigs were killed and samples from the jejunum, proximal colon, and distal colon were taken for microscopic examination for lesions, for immunohistochemical staining with anti-C. jejuni antibody to localize the bacteria, and for measurement of cytokine mRNA expression by real time PCR. The third and final objective was to determine if epithelial cell derived cytokines play a role in swine resistance to C. jejuni infection as determined by in vitro infection of swine intestinal epithelial cells. To address this issue, IPEC-l cells in both a differentiated and undifferentiated state were infected separately with C. jejuni strains 81-176 and ATCC 33292. Culture supematants were removed 0, 1, 3, 6, 8, and 24 hours afier infection and 139 .— -4. -2' LI)“. .011 analyzed for IL-18 protein levels by ELISA. Total RNA was isolated from cells infected for 8 hours and IL-18 mRNA expression was measured by RT-PCR. Collectively the purpose of these experiments was to characterize the nature of the local immune responses to C. jejuni and T. suis and to determine whether cytokine dysregulation could play a role in the pathology and disease caused by dual infection. Severe colonic pathology is characteristic of C. jejuni infection in humans, therefore characterizing the immune response in the colon is critical for understanding the pathogenesis of this disease. In a swine infection model C. jejuni targets distal colonic LGCs, stimulating germinal center expansion with IgA secreting B cells [2, 5]. To assess the effect of C. jejuni infection on cytokine expression in the distal colon and LGCs, I developed a rectal challenge model of infection. Weaned piglets inoculated rectally with 2x109 cfu of C. jejuni had 2-3 fold increases in expression of IL-18, TNFoc, and IL-8 in distal colon samples excluding lymphoid tissue 1 hour after infection, with a sustained increase in IL-8 expression 24 hours after infection. Concurrently, 3 to 20 fold increases in mRNA expression of IL-lB, IL-6, IL-8, IL-10, IL-18, GM-CSF, iNOS, MCP-l, and TNFa were measured from LGCs 1 hour after infection. Expression of IL-4, IL-5, IL- 12p35, IL-l2p40, and IL-13 was unaffected by C. jejuni infection. These data show that C. jejuni stimulates a proinflammatory response in vivo, and the more robust response in LGCs supports the hypothesis that these secondary lymphoid tissues are important sites for induction of an immune response directed against this pathogen. C. jejuni colonizes the intestinal mucus in the colon, associating with epithelial cells, goblet cells, and invading the deep crypts [1, 2]. In this thesis, I demonstrated through immunohistochemistry and laser capture microdissection that rectally inoculated 140 C. jejuni is capable of migrating to the same locations as orally inoculated bacteria. This occurs despite the fact that they did not travel through the acidic pH of the stomach or the bile rich environment of the small intestine, two environmental conditions that have been shown to stimulate C. jejuni virulence in vitro [6, 7]. These data correlate with those found by Bell and Manning who showed that rectal inoculation of naive ferrets resulted in colonization and diarrheal disease similar to their orally inoculated counterparts [8]. Although rectal inoculation in ferrets has already demonstrated the colonization and disease causing potential of C. jejuni infection by this nontraditional route, we used this technique in swine because they are more similar to humans physiologically and we were able to analyze the local cytokine responses in LGCs which have been characterized in pigs. Ultimately, these data show that rectal inoculation is a useful technique for studying in vivo effects of infectious agents with better definition of the affected area and more precise timing of responses. The availability of a genetically modified strain of C. jejuni that behaved identically to the wild type parent strain facilitated detection and identification in a non germ free or specific pathogen free animal infection model. In the rectal challenge study I used a strain of C. jejuni that had been electroporated with a plasmid containing the gfp gene under the influence of a constitutively active promoter. This strain was reisolated from the feces of a pig 24 hours after rectal inoculation, and was quickly identified by examination with an epifluorescent microscope. The original goal was to use this strain to allow for easy detection of the bacteria in frozen sections of tissue by fluorescence microscopy. Mixter and colleagues used a strain of C. jejuni expressing gfp to infect mice intraperitoneally and were able to track the bacteria by flow cytometry for Gfp. They 141 proposed this as a useful technique for detecting the bacteria in vivo [9]. While detection of fluorescent C. jejuni was successful in the mouse peritoneal infection model, in the swine model examination of unstained tissue sections microscopically for the fluorescent bacteria was unsuccessful. The primary impediment to accurate visualization of the strain used for inoculation was auto fluorescence of the intestinal tissue that masked the fluorescent bacteria. I tried to overcome this problem by treating the tissues with propidium iodide to stain nucleic acids to produce background staining of a different wavelength than Gfp fluorescence to facilitate visualization of the bacteria. Unfortunately, due to the small size of the bacteria and the presence of debris in the intestine that did not stain with propidium iodide, the bacteria could not be distinguished definitively. In fact, small fluorescent particles of the same size as the bacteria were present in control pigs as well as infected animals. This phenomenon has also been encountered in cecal and intestinal samples from chicks infected with C. jejuni with the same gfp construct (Anna Bates, personal communication). The presence of fluorescent particles in the intestines severely limited the usefulness of fluorescently tagged C. jejuni for direct visualization in our system and necessitated the use of IHC, LCM, and DNA tests to detect and localize the inoculating strain used in the rectal challenge experiment. However, the gfp marker was very useful for distinguishing the inoculating strain from naturally occurring Campylobacters in experimentally infected pigs. IL-18 became a focal cytokine in this work because it has been shown to play an important early role in the immune response to other enteric infections with similar pathogenesis. IL-18 is a pleiotrophic cytokine that functions in both innate and acquired immunity [10, 11]. In mammals studied to date, its mRNA and protein are both 142 constitutively expressed throughout intestinal tissues, including gut associated lymphoid tissues and intestinal epithelial cells [12, 13]. IL-18 production early in the response to Shigella and Salmonella infections is critical for stimulation of a Th 1 directed immune response with increased production of IFNy, which is essential for resolution of disease [14-16]. In vitro studies have shown that C. jejuni stimulates secretion of IL-1 B, IL-6, IL- 8, and TNFa from swine intestinal epithelial cells (Cunningham et al, unpublished data; Parthasarathy et al, unpublished data). In addition to initiating a Th 1 directed immune response, IL-18 stimulates proinflammatory cytokine production from leukocytes in vitro [14-18]. Therefore, we hypothesized that C. jejuni stimulates secretion of IL-18 from cultured swine intestinal epithelial cells and that this functions to stimulate further immune responses that kill the bacteria. Differentiated IPEC-l cells secreted significant amounts of IL-18 as early as 3 hours after infection. IL-18 levels remained high at 6 and 8 hours after infection, and then decreased by 24 hours. In contrast, undifferentiated IPEC-l cells did not secrete significant amounts of IL-18 in response to C. jejuni at any time, despite the fact that they secreted IL-18 in response to the positive control, Con A. Additionally, mRNA expression of IL-18 was not significantly altered by C. jejuni 8 hours after infection of IPEC-l cells in either differentiation state. Taken together, these data show that C. jejuni does induce IL-18 production from differentiated cells. Increased secretion in differentiated IPEC-l cells does not correlate with increased mRNA expression, suggesting that transcriptional regulation does not play a role in the IL-18 response of these C. jejuni infected intestinal epithelial cells. The increased expression of IL-18 seen in the rectal challenge model may be due to the intraepithelial lymphocytes, macrophages, or other leukocytes present in the lamina propria and submucosa of the 143 distal colon and the follicles of the LGCs in live pigs with the full anatomic complexity of the gastrointestinal tract. Increased secretion from differentiated IPEC-l cells is likely due to increased cleavage of stored immature protein by Caspase-l , facilitating its release from the cell. These data suggest that IL-18 functions in host defense against C. jejuni and that intestinal epithelial cells may be an important source of IL-18 in combating this infection in viva. Immunomodulation during concurrent infections can determine whether host pathology and disease are enhanced or abrogated. Pigs are naturally resistant to C. jejuni, which is likely due in part to a proinflammatory response initiated early in life during a primary infection. Upon infection with the swine whipworm T. suis in the proximal colon, C. jejuni invade tissues surrounding the site of worm attachment and LGCs in the distal colon [2, 3]. We hypothesized that C. jejuni invasion secondary to T. suis infection is due to downregulation of a protective proinflammatory immune response. To address this hypothesis, weaned piglets were orally inoculated with embryonated T. suis eggs. Twenty one days after inoculation, pigs were orally infected with C. jejuni. Two days after C. jejuni infection, pigs were killed and we measured cytokine mRNA expression in the jejunum, proximal colon, and distal colon of pigs infected with T. suis, C. jejuni, or both. In the proximal colons of both groups of pigs that received T. suis, IL-13 expression was upregulated 3 to 4 fold over uninfected controls. Concurrently, expression of cytokines known to be important for clearance of bacterial infections like IL-8 was decreased 2 to 3 fold, and IL-12 expression was decreased 2 to 4 fold compared to uninfected controls. Additionally, expression of both the chemokine MCP-l and the Th 2 cytokine IL-S decreased 2 fold in the dual infection group compared to mrinfected 144 controls. At the same time, there were no significant differences in expression of any cytokines in the proximal colons of pigs infected with C. jejuni only compared to uninfected controls. These data suggest a role for T. suis stimulated immunomodulation in the proximal colon. T. suis infection increased expression of IL-13 while concurrently decreasing the expression of proinflammatory cytokines in the proximal colon. IL-13 has been shown to decrease secretion of IL-8 from stimulated human intestinal epithelial cells in vitro [19]. Exogenous recombinant IL-13 administered to mice concurrently with a lethal dose of LPS decreases production of IL-12p40 and protects the mice from lethal toxemia [20]. Additionally, egg antigens from the intestinal helminth Schistosoma mansoni stimulate IL-10 production from dendritic cells, while decreasing production of IL-12 [21]. Based on T. muris infection studies in mice, we expected to see increased expression of IL-10 in the tissues of these T. suis infected pigs along with increased expression of IL-13. IL-10 expression was not increased in any tissue sampled from orally infected pigs, however a concurrent study on secreted cytokines in the feces of these pigs showed increased IL-10 in T. suis infected pigs on day 22 (Parthasarathy et al, unpublished results). Increased expression of IL-10 may not have been measured because we examined tissues 23 days after T. suis infection, which was after the increase of fecal cytokine levels was measured. Also, T. muris infection studies in mice generally focus on cytokine production in draining lymph nodes, which does not correlate exactly with local cytokine expression in the tissues. Nevertheless, increased production of IL-10 on day 22 likely had immunomodulatory effects on cytokine expression on day 23. The combined actions of T. suis stimulated IL-10 and IL-13 are likely responsible for decreased 145 expression of IL-8, IL-12, and possibly MCP-l as well. Based on data from previous dual infection studies, we expected to see increased numbers of C. jejuni in the crypts and tissues of dual infected pigs. C. jejuni was detected by RFLP analysis in 4 of 6 pigs from the dual infection group, and l of 6 pigs from each of the single infection groups and the controls. Increased prevalence of C. jejuni in the proximal colon of dual infected pigs compared to pigs that received C. jejuni only suggests that T. suis facilitates its proliferation and possibly invasion. Detection of C. jejuni from control animals suggests that C. jejuni was likely present in low numbers as a commensal in these pigs. Taken together, these data suggest that decreased proinflammatory cytokine expression resulting from T. suis infection facilitates proliferation of C. jejuni. In the distal colon, cytokine expression was only altered significantly in the T. suis only group. Three to four fold increases in expression of IL-10, IL-6 and IFNy were measured, while expression of IL-13 was upregulated 3 fold compared to uninfected controls. Increased expression of IL-lB, IL-6 and IFNy is indicative of an inflammatory response, possibly due to secondary bacterial infection. C. jejuni was not detected by RFLP in the distal colons of any pigs, however, it has previously been demonstrated that T. suis facilitates secondary invasion of many species of resident bacteria, including C. coli, an unidentified Campylobacter spp., and E. coli [3]. Invasive strains of C. coli stimulate IL-8 secretion from cultured human intestinal epithelial cells [22]. Analysis of distal colon tissues by RFLP of the Campylobacter 23S rRNA gene detected C. coli in 2 of 6 pigs, and an unidentified species of Campylobacter in 3 of 6 pigs. Thus it is possible that either of these species invaded tissues in the distal colon and initiated an inflammatory response. It has also been shown that oral inoculation of pigs with E. coli 146 DHSa causes increased secretion of proinflammatory cytokines in the feces (Cunningham et al, unpublished results). Larval stages of T. suis produce ESP in vivo, as is evidenced by the fact that serum antibodies directed against a 20kDa glycoprotein isolated from adult worm culture fluids can be detected as early as 21 days after inoculation with embryonated eggs [23]. In addition, T. suis ESP stimulates production of IL-6 and IL-10 from IPEC-l swine intestinal epithelial cells in vitro. Therefore, increased expression of IL-13 in the distal colon of T. suis infected pigs could be attributed to the effects of excretory/secretory product (ESP) produced by the 4th stage larvae in the proximal colon that have traveled to the distal colon. Taken together, these data indicate that ESP is highly stimulatory and likely has effects on intestinal epithelial cells which would increase cytokine production in vivo at sites distal to worm attachment. Interestingly, although the cytokine response in the proximal colon was similar between the T. suis and dual infection groups, the responses diverged in the distal colon. There were no significant changes in cytokine expression in the distal colon of the dual infection group. This was unexpected, as previous studies have shown that T. suis facilitates invasion of C. jejuni and resident bacteria into the LGCs in the distal colon [2, 3], and distal colonic samples from our pigs likely included LGCs. In our rectal challenge model we showed that C. jejuni stimulates proinflammatory cytokines in LGCs 1 hour after infection, therefore we expected to see a strong cytokine response from the dual infected pigs. Tissues for cytokine expression from orally inoculated pigs were taken 48 hours after C. jejuni infection, which corresponded to 23 days after T. suis infection. Our rectal challenge model showed that proinflammatory cytokine responses were seen primarily 1 hour after infection, therefore it is likely that proinflammatory cytokines had 147 returned to baseline levels by the time samples were taken from orally infected pigs. The inability to recover C. jejuni from fecal culture of these pigs 48 hours after oral inoculation indicated that low levels of C. jejuni were present. The oral inoculum was 108 cfu of C. jejuni, while the inoculum used in the rectal challenge experiment was 2x109 cfu of C. jejuni. Therefore the low level of C. jejuni present in the distal colon 48 hours may have been too low to stimulate a significant proinflammatory response. It is more likely that the absence of a measurable proinflammatory cytokine response in the distal colons of dual infected pigs is due to sampling after the response has waned. Indeed, gnotobiotic piglets orally inoculated with only 106 cfu of C. jejuni 33292 developed transient fever that lasted only for the first 24 hours after infection, suggesting that C. jejuni stimulated a proinflammatory response with increased production of IL-6 that caused fever [2]. Another issue that may have affected distal colonic cytokine responses is the larval stage present in the pigs. T. suis undergoes 4 molts from L1 larvae to L5 adults in about 41 days [24, 25]. Mucohemorrhagic diarrhea associated with T. suis infection can occur as early as 14 days in heavy infection, but on average occurs around 21 days [26, 27]. This corresponds to the emergence of the posterior end of L4 larvae into the lumen [24, 25]. The severity of diarrheal disease and pathology increases as worms mature to adults, reproducing and excreting significant amounts of ESP [27] (Linda Mansfield, personal communication). Progression of disease and pathology likely correlates with intensified cytokine responses. Though T. suis larvae were present in the proximal colons of dual infected pigs, and significant histologic changes were seen, lesions were confined to the areas where worms were present. The time that the pigs in our study were sampled 148 corresponded to the beginning of the molt from L3 to L4 larvae, and ESP production would have been low. Therefore, it is possible that in the dual infection groups, distal colonic effects of T. suis would not yet have been manifested. Analysis of cytokine expression at later time points, including 30 days and 41 days after T. suis infection would determine whether an inflammatory response is induced in the distal colon as disease progresses. C. jejuni infection alone stimulated significant changes in the jejunum of orally infected animals. In the jejunum of C. jejuni infected pigs expression of MCP-l, TNFat, IL-12p40, IFNy, IL-4, and IL-10 were increased 3 to 5 fold over uninfected controls. C. jejuni stimulates intracellular production of MCP-l, TNFoc, IFNy, IL-4, and IL-10 from cultured human intestinal epithelial cells [28, 29]. Additionally, jejunal samples likely contained samples of peyers patches as well as surrounding epithelium. It has been demonstrated in our rectal challenge model, and in an oral challenge model of germ free piglets [2] that C. jejuni targets LGCs and stimulates robust cytokine responses. It is likely that C. jejuni stimulated a similar response in the jejunal Peyers patch, which would have influenced our results. These results indicate that C. jejuni stimulates a proinflammatory response in the jejunum in the absence of T. suis. In the jejunum of pigs infected with C. jejuni and T. suis concurrently, GM-CSF is downregulated 5 fold compared to uninfected controls, and the rest of the cytokines measured were not significantly different that uninfected controls. This suggests that T. suis exerts immunomodulatory effects in the jejunum as well as the proximal colon, down regulating C. jejuni induced proinflammatory cytokines. These effects could be due to systemic IL- 13 and IL-10 produced by lymphocytes stimulated by worm antigens in draining lymph 149 nodes. Examination of cytokine expression in draining lymph nodes and peripheral leukocytes would be necessary to confirm this. Increased expression of proinflammatory cytokines in the distal colon and LGCs of rectally inoculated pigs and in the jejunum of orally inoculated pigs indicated that C. jejuni stimulated a proinflammatory response in viva. However, the fact that the pigs in these studies were immunocompetent, conventionally reared animals with a complete intestinal microbial population raises the question of whether some of the cytokine responses are the result of direct actions of C. jejuni on intestinal tissues, or indirect actions due to perturbation of intestinal flora homeostasis by the introduction of exogenous bacteria. Pigs orally infected with a non-pathogenic laboratory strain of E. coli DHSa have increased secretion of IL-6 in the feces 48 hours after inoculation, but this strain does not invade swine intestinal epithelial cells in vitro (Parthasarathy et al, unpublished results). Introduction of exogenous bacteria could increase the total intestinal bacterial load above normal levels. Additionally, any exogenous strain could compete with resident strains for resources or provide an essential growth factor, causing an imbalance in the intestinal environment and facilitating overgrowth of resident strains. These factors could stimulate inflammatory responses in an effort by the host to reduce total bacterial numbers and possibly eliminate the exogenous strain, restoring balance to the microbial population. Taking this into account, exogenous C. jejuni may have stimulated a host inflammatory response indirectly. However, in vitro data definitively shows that C. jejuni infection alone stimulates secretion of a similar panel of proinflammatory cytokines from cultured intestinal epithelial cells, suggesting that the 150 responses seen in viva can be attributed to both direct and indirect effects of C. jejuni on intestinal tissues. Collectively, the data from this dissertation project support the hypothesis that cytokine dysregulation during dual infection with T. suis and C. jejuni is a major cause of the severe disease and pathology observed. From this analysis of local cytokine expression in pigs infected singly or concurrently with C. jejuni and T. suis, we propose the following model for the roles of specific cytokines in disease progression in vivo. A proinflammatory response is stimulated by C. jejuni early in the infection, from both intestinal epithelial cells and LGCs. IL-1 8 released from intestinal epithelial cells early in the immune response induces IL-lB, IL-8, and TNFa from leukocytes, amplifying and sustaining inflammation. Additionally, epithelial cell derived IL-18 recruits and activates neutrophils to engulf and eliminate extracellular bacteria. With the addition of T. suis, IL- 13 is upregulated while IL-8 and IL-12 are downregulated. IL-13 and IL-4 stimulate mucin production from intestinal epithelial cells [30, 31] which provides a favorable environment for proliferation of C. jejuni [32, 33]. Piglets infected with a low dose of C. jejuni, 106 cfu, develop transient fever but no diarrhea, while piglets infected with high doses of C. jejuni, 4-5x109 cfu, develop diarrheal disease and pathology [1, 2, 34]. Overgrowth of C. jejuni in a mucus rich environment could be responsible in part for the mucohemorrhagic diarrhea seen in dual infection. Also, IL-13 downregulation of IL-8 is likely to abrogate neutrophil recruitment and containment of bacteria in the epithelial layer [35, 36], allowing invasion by significantly increased numbers of C. jejuni into deeper tissues. 151 Other cytokines may enhance invasion of C. jejuni. IL-4 has also been shown to decrease transepithelial electrical resistance of differentiated IPEC-l cells and increase invasion of C. jejuni (Parthasarathy et al, unpublished results). Although increased IL-4 expression was not detected in our oral challenge study, concurrent analysis of secreted cytokines in the feces showed increased secretion on day 22 in dual infected pigs (Parthasarathy et al, unpublished results). In vivo, T. suis induced IL-4 may weaken tight junctions between epithelial cells allowing bacteria access to the basolateral surface of cells. These bacteria that have breached the epithelial barrier could bind fibronectin in the extracellular matrix via CadF [37] and gain access to the lamina propria and submucosa. Another factor to consider is the secreted worm products. ESP has a dose dependent cytotoxic effect on undifferentiated epithelial cells. Additionally, ESP exposed to both apical and basolateral surfaces of differentiated cells significantly decreases transepithelial electrical resistance and damages perijunctional complexes, making cellular junctions more permissive [34]. The combined effects of ESP on differentiated and undifferentiated epithelial cells would result destruction of the epithelial barrier in viva, allowing invasion of C. jejuni into deeper tissues, increasing the severity of disease. Ultimately, the combined actions of T. suis stimulated IL-4 and IL-13 would provide a favorable environment for C. jejuni proliferation, while dampening the host response that would protect against invasion by overwhelming amounts of C. jejuni. 152 Future Directions The analysis of local cytokine expression in intestinal tissues from C. jejuni and T. suis infected pigs expanded our knowledge of the swine immune response to these two agents and shed some light on cytokine dysregulation that can occur during infection with these two agents. These data would be enhanced by exploring several aspects of this dual infection phenomenon. In chapter 2 we analyzed cytokine expression in the distal colon and LGCs of pigs rectally inoculated with C. jejuni. Further analysis of cytokine expression at different times, for example 4, 8, and 12 hours after infection, would be instructive as to the in vivo kinetics of cytokine expression in response to C. jejuni. Additionally, since our data showed that the cytokine response was more robust in the LGCs compared to the distal colon, identification of cytokine producing cells by in situ hybridization or flow cytometry in these two tissues would elucidate the specific cells activated by C. jejuni. Concurrent analysis of intracellular protein within the cells by immunohistochemistry, and of proteins secreted into the feces by ELISA would allow comparison of changes in expression and changes in secretion. In chapter 3 we analyzed cytokine expression in the jejunum, proximal colon, and distal colon of pigs orally infected with C. jejuni, T. suis, or both. As with the rectally inoculated pigs, further analysis of cytokine expression at different times with concurrent analysis of intracellular and secreted proteins would expand our knowledge of in viva cytokine kinetics. Analysis of cytokine expression 4, 8, 12, and 24 hours after infection would allow us to determine if proinflammatory responses were induced in C. jejuni infected pigs early in the response to oral infection. In addition, examination of cytokine 153 expression at later time points, for example on day 30, day 41, and day 48 after T. suis infection, would help us to asses cytokine changes that may coincide with developmental changes in T. suis. Concurrent examination of pathologic changes would correlate lesion progression with cytokine changes. Previous studies of immunoglobulin production from piglets orally inoculated with C. jejuni and the rectal challenge study from this dissertation demonstrated the importance of LGCs as sites for induction of immune responses to C. jejuni. To further characterize the cytokine response to C. jejuni infection, identification of specific cell types that are reactive to C. jejuni and characterization of their cytokine responses should be undertaken. To achieve this, multiple LGCs would be collected from pigs orally or rectally infected with C. jejuni. Individually, each LGC would be gently disrupted to make single cell suspensions and an aliquot would be tested for the presence of C. jejuni by PCR amplification of an appropriate gene, for example the QRDR of the gyrA gene. LGCs that do not contain C. jejuni would serve as internal negative controls for each pig. The cells would then be stimulated in vitro with C. jejuni and subsequently stained for specific cell markers to detect CD4+ cells, CD8+ cells, B-cells, and macrophages. The cells would be separated by flow cytometry and assayed for expression of a panel of proinflammatory and anti-inflammatory cytokines by real time PCR. This study would determine which cell types are involved in the immune response to C. jejuni and also provide information about the distribution of C. jejuni in vivo. In chapter 4 we measured IL- 1 8 secretion from differentiated and undifferentiated IPEC-l cells infected with C. jejuni. The difference in responses between the two differentiation states raises the question of whether cellular maturation coincides with 154 intracellular signaling pathways that allow differentiated cells to secrete IL-18 in response to C. jejuni, while undifferentiated cells do not. This question could be addressed by examination of C. jejuni activated intracellular signaling molecules using western blot analysis of intracellular proteins. We may find that at some point in the signal transduction cascade a key activator of signaling molecules specific to the IL-18 pathway is absent or repressed in undifferentiated cells, indicating that the differentiation process includes synthesis of this activator or derepression of its expression. We also found that E. coli LPS does not stimulate IL-l8 secretion from differentiated IPEC-l cells. Previous studies have shown that adherence, invasion, and Cdt are virulence factors of C. jejuni that stimulate IL-8 secretion from intestinal epithelial cells [22, 38]. The effect of these and other specific virulence factors on IL-18 secretion from swine intestinal epithelial cells should be explored to determine the mechanisms that C. jejuni uses to stimulate host cytokine responses in the gut. One factor that affected interpretation of the results from our in vivo studies, discussed in chapters 2 and 3, was the presence of other Campylobacters in conventionally reared pigs. It has been reported that healthy pigs can harbor C. jejuni, C. coli, and C. lari as commensals [4]. In my studies C. coli was detected by RFLP analysis of the Campylobacter 23S rRNA gene in both rectally and orally infected control pigs. An unidentified Campylobacter species that had an RF LP pattern that was different from all the other thermophilic Campylobacters detected by this assay was found in at least one intestinal segment from virtually 100% of orally infected pigs. Immunohistochemistry using antibody raised against whole C. jejuni bacteria positively stained organisms in the colons of all experimental pigs. Antibody validation tests 155 revealed cross reactivity with C. lari, but preliminary PCR analysis of tissue DNA did not detect C. lari in any pigs regardless of infection route. These data suggest that another Campylobacter species was present in the colons of our experimental animals that occupied the same niche as C. jejuni. A detailed analysis of the resident Campylobacter strains in experimental animals would help to determine whether the changes in cytokine expression were due to direct interaction of C. jejuni with colonic tissues, to the interaction of C. jejuni with the unidentified Campylobacter resulting in it invading tissues, or to the interaction of C. jejuni with the resident bacterial population causing a perturbation of environmental homeostasis. To address this issue we would clone and sequence l6S rRNA genes from as many Campylobacter species amplified from total pig intestinal DNA as possible, then compare the sequences obtained from our pigs to sequences in bacterial ribosomal databases. The results from this experiment may reveal that C. lari is present in the pigs, that a variant of C. jejuni, C. coli, or C. lari is present that gives a unique RFLP pattern using the 23S rRNA gene, or that an as yet uncharacterized Campylobacter is present in the pigs. This analysis is currently underway. The data gathered from these experiments could also be enhanced by quantifying Campylobacters in the feces of experimental animals and comparing bacterial loads between controls and infected animals. We may find that the total bacterial load is unchanged, but the ratios of different organisms are different and might indicate a dose dependent effect of Campylobacter infection on local intestinal cytokine production. In summary, the data generated from this dissertation contributed to the body of research defining host immune responses to C. jejuni and T. suis infections. We 156 developed a swine rectal challenge model that proved to be a useful technique for studying the complexity of the host immune response to C. jejuni, while allowing the precise timing and specific targeting of tissues afforded by in vitro models. We began to define the host local immune response to T. suis and the T. suis induced immunomodulation that may decrease natural resistance to C. jejuni, facilitating invasion. Finally, through in vitro infection studies we discovered that C. jejuni infected intestinal epithelial cells are a source of early IL-18 production, which may be critical in initiating a protective response to infection in viva. Collectively this data gave us evidence for T. suis downregulation of proinflammatory responses that are detrimental to swine colonized by the opportunistic pathogen C. jejuni. 157 The results obtained from this dissertation opened several avenues for further research and generated new questions to be addressed. Experimental designs of studies to address three of these questions will be discussed in detail. Cytokine responses of intestinal epithelial cells to multi-species Campylobacter infection Healthy, conventionally reared pigs can harbor multiple Campylobacter species in their colons without evidence of disease [4]. In our swine rectal challenge model, inoculation of C. jejuni into the distal colons of conventionally reared pigs with naturally acquired C. coli stimulated increased expression of proinflammatory cytokines. The hypothesis for this study is that swine intestinal epithelial cells exposed to C. coli will secrete proinflammatory cytokines when given a secondary challenge with C. jejuni. Differentiated and undifferentiated IPEC-l cells will be exposed to C. coli for 0, 1, 3, 6, 8, and 24 hours. The supematants will be removed and saved for cytokine assay by ELISA. Then the cells will be exposed to C. jejuni expressing Gfp, which are also resistant to kanamycin, for l, 3, 6, 8, and 24 hours. The supematants will be removed and saved for cytokine assay by ELISA, and then the cell monolayers will be treated with 100mg/mL gentamicin for 1 hour to kill extracellular bacteria. The gentamicin will be removed, then the cells will by lysed in 0.1% sodium deoxycholate and the cell lysates will be plated on Bolton agar with kanamycin to select for the inoculating strain of C. jejuni. Supematants from C. coli only infected cells as well as cells infected with C. coli, then subsequently infected with C. jejuni will be assayed for IL-IB, IL-6, IL-8, lL-18, 158 and TNFor by ELISA. These experiments will determine 1) if C. coli stimulates proinflammatory cytokine production from IPEC-l cells; 2) if C. jejuni stimulates a proinflammatory response in cells previously exposed to C. coli; and 3) if C. jejuni invasion into IPEC-l cells is altered by prior exposure of the cells to C. coli. 159 The effects of IL-13 on proinflammatory cytokine production and C. jejuni invasion in viva T. suis induced IL-13 downregulates expression of proinflammatory cytokines in the proximal colons of pigs concurrently infected with C. jejuni (Jones et al, discussed in Chapter 3). In a rectal challenge model, expression of proinflammatory cytokines is significantly increased in pigs inoculated with C. jejuni. The hypothesis for this study is that IL-13 will downregulate proinflammatory cytokines in the distal colon and facilitate invasion of epithelial cells by C. jejuni. Using a rectal challenge model, the distal colons of pigs will be pretreated with recombinant IL-13, serum free media, or no treatment for 5 hours, then all pigs will be infected with C. jejuni expressing Gfp rectally. Three hours after C. jejuni infection, the pigs will be killed and the distal colons will be removed. Intestinal epithelial cells will be isolated by the enzymatic digestion technique described by Babakhani and Joens [1]. Briefly, the colonic sections will be washed with PBS, then the enterocytes will be detached by incubation with 0.25% trypsin-lmM EDTA at 37°C on a rotator. Detached cells will be washed three times with MEM supplemented with 1% FBS, then the cells will be incubated for 1 hour with lOOug/mL gentamicin in MEM supplemented with 1% FBS to kill extracellular bacteria. The cells will then be fixed with paraforrnaldehyde, and cells containing internalized fluorescent C. jejuni can be separated and counted by flow cytometry. The cells could then be stained for intracellular IL-8 protein with specific antibodies. These experiments will determine 1) if recombinant IL- 13 facilitates increased invasion of intestinal epithelial cells with C. jejuni in vivo; 2) if C. 160 jejuni invasion stimulates IL-8 production from intestinal epithelial cells; and 3) if C. jejuni stimulated IL-8 secretion is decreased by IL-13 pretreatment. 161 The effects of IL-18 on C. jejuni invasion of intestinal epithelial cells in vitro IL-18 is a pleiotrophic cytokine that is induced in viva during infections with enteric bacteria, and is critical for host resistance to the pathogens. C. jejuni stimulates IL-18 secretion from differentiated IPEC-l cells in vitro. In addition, IL-18 expression is significantly increased in the distal colons and LGCs of pigs one hour after rectal inoculation with C. jejuni. The hypothesis of this study is that early production of IL-18 from intestinal epithelial cells after infection with C. jejuni is important for resistance to invasion. Two experimental designs will be used to test this hypothesis. In the first experiment, differentiated IPEC-l cells will be pretreated with different doses of recombinant IL-18 for 5 hours. The supernatant will be removed and saved for later cytokine assay. The pretreated cells will then be infected with C. jejuni for 3 hours, and then treated with lOOpg/mL gentamicin for 1 hour to kill extracellular bacteria. The gentamicin will be removed, the cells will be lysed with 0.1% sodium deoxycholate, and cell lysates will be plated to enumerate internalized bacteria. In the second experiment, differentiated IPEC-l cells will be treated with anti-IL-18 antibody and concurrently infected with C. jejuni. Supematants will be removed 1, 3, 6, 8, and 24 hours after infection and saved for later cytokine assay. The infected monolayers will be treated with gentamicin to kill extracellular bacteria, then lysed and plated to enumerate internalized bacteria. The supematants from both experiments will be analyzed by ELISA for IL-IB, IL-6, and IL-8 which can all be induced by IL-18 stimulation [17, 18]. These experiments will determine 1) if pretreatment with recombinant IL-l8 enhances IPEC-l resistance to invasion; 2) if blocking of IL-18 with antibodies during C. jejuni infection facilitates 162 invasion; and 3) if IL-l8, either exogenous or recombinant, induces increased secretion of IL-1 [3, IL-6, and IL-8 during C. jejuni infection. I63 10. ll. 12. l3. I4. 15. 16. References Babakhani, F.K., G.A. Bradley, and LA. Joens, Newborn piglet model for campylobacteriosis. Infect Immun, 1993. 61(8): p. 3466-75. Mansfield, L.S., et al., Enhancement of disease and pathology by synergy of Trichuris suis and C ampylobacter jejuni in the colon of immunologically naive swine. Am J Trop Med Hyg, 2003. 68(3): p. 70-80. Mansfield, LS. and J .F. Urban, Jr., The pathogenesis of necrotic proliferative colitis in swine is linked to whipworm induced suppression of mucosal immunity to resident bacteria. Vet Immunol Immunopathol, 1996. 50(1-2): p. 1-17. Young, C.R., et al., Enteric colonisation following natural exposure to Campylobacter in pigs. Res Vet Sci, 2000. 68(1): p. 75-8. Mansfield, LS. and D.T. Gauthier, Lymphoglandular complexes are important colonic sites for immunoglobulin A induction against Campylobacterjejuni in a swine disease model. Comp Med, 2004. 54(5): p. 514-23. Allen, K.J. and M.W. Griffiths, Eflect of environmental and chemotactic stimuli on the activity of the Campylobacterjejuni/MA sigma(28) promoter. FEMS Microbiol Lett, 2001. 205(1): p. 43-8. Rivera-Ami“, V., et al., Secretion of the virulence-associated Campylobacter invasion antigens from Campylobacterjejuni requires a stimulatory signal. J Infect Dis, 2001. 183(11): p. 1607-16. Bell, J .A. and DD. Manning, A domestic ferret model of immunity to Campylobacterjejuni- induced enteric disease. Infect Immun, 1990. 58(6): p. 1848-52. Mixter, P.F., et al., In vivo tracking of C ampylobacter jejuni by using a novel recombinant expressing green fluorescent protein. Appl Environ Microbiol, 2003. 69(5): p. 2864-74. Akira, S., The role of IL-18 in innate immunity. Curr Opin Immunol, 2000. 12(1): p. 59-63. Dinarello, C.A., et al., Overview of interleukin-1 8: more than an interferon-gamma inducing factor. J Leukoc Biol, 1998. 63(6): p. 658-64. Foss, D.L., M.J. Zilliox, and M.P. Murtaugh, Bacterially induced activation of interleukin-I 8 in porcine intestinal mucosa. Vet Immunol Immunopathol, 2001. 78(34): p. 263-77. Foumout, S., et al., Cloning, chromosomal location, and tissue expression of the gene for pig interleukin-I8. Immunogenetics, 2000. 51(4-5): p. 358-65. Biet, F ., C. Locht, and L. Kremer, Immunoregulatory functions of interleukin I 8 and its role in defense against bacterial pathogens. J Mol Med, 2002. 80(3): p. 147-62. Elhofy, A. and K.L. Bost, Limited interleukin-I8 response in Salmonella-infected murine macrophages and in Salmonella-infected mice. Infect Immun, 1999. 67(10): p. 5021-6. Sansonetti, P.J., et al., Caspase—I activation of IL-1 beta and lL-I8 are essential for Shigella flexneri-induced inflammation Immunity, 2000. 12(5): p. 581-90. 164 I7. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. Netea, M.G., et al., Interleukin- I 8 induces production of proinflammatory cytokines in mice: no intermediate role for the cytokines of the tumor necrosis factor family and interleukin-I beta. Eur J Immunol, 2000. 30(10): p. 3057-60. Puren, A.J., et al., Interleukin-I8 (IFNgamma-inducing factor) induces IL-8 and IL-I beta via T NF alpha production from non-CD14 + human blood mononuclear cells. J Clin Invest, 1998. 101(3): p. 711-21. Lugering, N., et al., Interleukin (IL)-I3 and IL-4 are potent inhibitors of IL-8 secretion by human intestinal epithelial cells. Dig Dis Sci, 1999. 44(3): p. 649-55. Muchamuel, T., et al., IL-I3 protects mice from lipopolysaccharide-induced lethal endotoxemia: correlation with down-modulation of TNF-alpha, IF N-gamma, and IL- I 2 production. J Immunol, 1997. 158(6): p. 2898-903. Kane, C.M., et al., Helminth antigens modulate TLR-initiated dendritic cell activation. J Immunol, 2004. 173(12): p. 7454-61. Hickey, T.E., et al., Campylobacterjejuni cytolethal distending toxin mediates release of interleukin-8fi'am intestinal epithelial cells. Infect Immun, 2000. 68(12): p. 6535-41. Hill, D.E., R.D. Romanowski, and J .F . Urban, Jr., A Trichuris specific diagnostic antigen from culture fluids of Trichuris suis adult worms. Vet Parasitol, 1997. 68(1-2): p. 91-102. Beer, RJ., Morphological descriptions of the egg and larval stages of Trichuris suis Schrank, I788. Parasitology, 1973. 67(3): p. 263-78. Beer, RJ., Studies on the biology of the life-cycle of Trichuris suis Schrank, I788. Parasitology, 1973. 67(3): p. 253-62. Batte, E.G., et al., Pathophysiology of swine trichuriasis. Am J Vet Res, 1977. 38(7): p. 1075-9. Rutter, J .M. and RJ. Beer, Synergism between Trichuris suis and the microbial flora of the large intestine causing dysentery in pigs. Infect Immun, 1975. 11(2): p. 395-404. Al-Salloom, F .S., et al., Campylobacter-stimulated IN T407 cells produce dissociated cytokine profiles. J Infect, 2003. 47(3): p. 217-24. Bakhiet, M., et al., Induction of alpha and beta chemokines by intestinal epithelial cells stimulated with Campylobacterjejuni. J Infect, 2004. 48(3): p. 236-44. Blanchard, C., et al., IL-4 and IL-I3 up—regulate intestinal trefoil factor expression: requirement for STA T6 and de novo protein synthesis. J Immunol, 2004. 172(6): p. 3775-83. Iwashita, J ., et al., mRNA of MUCZ is stimulated by IL-4, IL-I3 or TNF-alpha through a mitogen- activated protein kinase pathway in human colon cancer cells. Immunol Cell Biol, 2003. 81(4): p. 275-82. Beery, J .T., M.B. Hugdahl, and M.P. Doyle, Colonization of gastrointestinal tracts of chicks by Campylobacterjejuni. Appl Environ Microbiol, 1988. 54(10): p. 2365-70. McSweegan, E. and R.I. Walker, Identification and characterization of two Campylobacterjejuni adhesinsfor cellular and mucous substrates. Infect Immun, 1986. 53(1): p. 141-8. 165 34. 35. 36. 37. 38. Abner, S.R., et al., Response of intestinal epithelial cells to Trichuris suis excretory-secretory products and the influence on Campylobacterjejuni invasion under in vitro conditions. J Parasitol, 2002. 88(4): p. 738-45. Netea, M.G., et al., Neutralization of lL-I 8 reduces neutrophil tissue accumulation and protects mice against lethal Escherichia coli and Salmonella typhimurium endotoxemia. J Immunol, 2000. 164(5): p. 2644-9. Sansonetti, P.J., et al., Interleukin-8 controls bacterial transepithelial translocation at the cost of epithelial destruction in experimental shigellosis. Infect Immun, 1999. 67(3): p. 1471-80. Konkel, M.B., et al., Identification and molecular cloning of a gene encoding a fibronectin- binding protein (CadF) from Campylobacterjejuni. Mol Microbiol, 1997. 24(5): p. 953-63. Hickey, T.E., et al., Campylobacterjejuni-stimulated secretion of interleukin-8 by IN T407 cells. Infect Immun, 1999. 67(1): p. 88-93. 166