'THEBJS LIBRARY Michigan State University This is to certify that the dissertation entitled EVALUATION OF PROINFLAMMATORY CYTOKINES IN PIGS INFECTED WITH CAMPYLOBACTER JEJUNI AND TRICHURIS SUIS presented by LAKEISHA DIANELE CUNNINGHAM has been accepted towards fulfillment of the requirements for the Ph.D. degree in Microbiology and Molecular Genetics 'ro essor’s Signature (EX/012007 Date MSU is an affinnative-action, equal-opportunity employer .uu —- .I-Q-O-I-I—t—l-1—n-o—D-l-l—n-I-I—n—9-0-I-I-0-0-d-D-o-n-0"-.---nr-I PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 K:IProlecc&Pres/ClRC/DataDue.indd EVALUATION OF PROINFLAMMATORY CYTOKINES IN PIGS INFECTED WITH CAMPYLOBACTER JEJUNI AND TRICHURIS SUIS BY Lakeisha Dianele Cunningham 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 2007 ABSTRACT EVALUATION OF PROINFLAMMATORY CYTOKINES IN PIGS INFECTED WITH CAMPYLOBACTER JEJUNI AND TRICHURIS SUIS By Lakeisha Dianele Cunningham Campylobacterjejuni is a leading cause of gastroenteritis worldwide. In conventionally reared swine, C. jejuni is commonly found in the colon without pathology, but infection and pathology result when the swine whipworm, T. suis, is present. The central hypothesis of this dissertation was that C. jejuni induces natural host resistance via proinflammatory cytokine responses following primary oral infection, but this response is down-regulated in the presence of T. suis thus promoting C. jejuni infection. Specific aims: 1) Evaluate secreted pro- and anti-inflammatory cytokines (IL- 8, IL-l-B, TNF-a, IL-4, IL-6, and IL-10) in the feces of pigs after single or dual challenge infections with C. jejuni and T. suis, 2) Assess IL-8, IL-l-B and TNF-a production from undifferentiated, crypt-like intestinal pig epithelial cells (IPEC-l cells) following C. jejuni infection, and 3) Assess whether IL-8, TNF-a, or IL-l-B treatment of IPEC-l cells have effects on C. jejuni invasion, adherence, and/ or transepithelial electrical resistance (TER) across this mucosa] membrane. In aim 1, weaned piglets were used in short-term (ST, 2 days) and long-term (LT, 23 days) challenge experiments. In the LT experiments, there were 5 groups with two groups given 2500 T. suis orally. At 21 days post-T. suis inoculation, some of the T. suis-infected pigs received C. jejuni, while the remaining pigs received either C. jejuni alone, E. coli DHS-a, or sterile milk. On day 23, all pigs were euthanized and gastrointestinal (GI) tissue samples taken to measure cytokine expression by real time PCR. Campylobacterjejuni stimulated increased proinflammatory cytokine expression in the jejunum while T. suis down-regulated these responses here and in the proximal colon. T richuris suis contributed significantly to diarrheal disease in both single and concurrent infections. Also, enzyme linked irnmunosorbent assay (ELISA) conducted on supernatants from feces indicated that T. suis significantly decreased expression of fecal IL-l—B during concurrent C. jejuni and T. suis infection. In the ST experiment where pigs were inoculated simultaneously with either C. jejuni, T. suis, C. jejuni and T. suis, E. coli DHS-a, or sterile milk none showed significant acute changes in fecal IL-8, IL-l-B, TNF-a, IL-4, IL-6, or IL-10 expression by ELISA. Here, cytokine responses were measurable using fecal ELISAs, but cytokine mRN A expression assays in tissues were more informative. Also, we discovered that secondary invasion of certain epsilon proteobacteria into GI tissues may affect cytokine responses. In aim 2, confluent undifferentiated IPEC-1 cells were infected with C. jejuni and culture supematants analyzed for IL-8, TNF-a, or IL-l-B expression using ELISA. Campylobacterjejuni induced IPEC-1 cells to secrete TNF-a and lL-l-B, but IL-8 was constitutively secreted and not inducible by C. jejuni infection. These data show a pattern of cytokine responses indicative of an innate inflammatory response that we hypothesize to be responsible in part for eliminating these bacteria from the host. In aim 3, the TER of confluent IPEC cells was measured prior to and following treatment with recombinant swine IL-8, TNF- a, or IL-l-B. None of these cytokines had direct effects on C. jejuni interactions with undifferentiated, crypt-like intestinal epithelial cells. Collectively, these data provide evidence for T. suis contributing to cytokine dysregulation during concurrent infection. These immunomodulatory effects may promote C. jejuni infection of the intestinal mucosal epithelium by limiting the natural host resistance to bacterial infection. Copyright by LAKEISHA DIANELE CUNNINGHAM 2007 DEDICATION This dissertation is dedicated to my Lord and Savior, Jesus Christ; my husband, Steven; my daughters, Jadyn and Kailyn and my mother, Carolyn Price. ACKNOWLEDGEMENTS There are several people I would like to acknowledge for making the completion of this degree possible. First, I must acknowledge God for giving me this opportunity and providing me with the strength to persevere and succeed. I sincerely and lovingly thank my husband, Dr. Steven G. Cunningham; my daughters, Jadyn Elyse and Kailyn Danielle; my mother, Carolyn; my father, George and my entire family for their never- ending support, prayers, encouragement, and love throughout my graduate studies. I truly thank my mentor, Dr. Linda S. Mansfield, for supporting and guiding me through this journey of higher learning. I am also grateful to my committee members (Drs. Richard Schwartz, Martha Mulks, John Linz, and Maria Patterson) for their support and guidance throughout my graduate studies. I must also thank my labmates, Drs. Geetha Parthasarathy and Kathryn Jones and Stacey Wilder, for their emotional support, their assistance in conducting the experiments, and for their constructive criticism on my writing. Dr. Julia Bell encouraged me along the way, shared her scientific research expertise, and provided critical review on the writing of this dissertation; she believed in me and will always be my dear friend. David Wilson also provided research assistance and information pertaining to Campylobacterjejuni. Drs. Pawin Padtmgtod and Bo Norby, provided veterinary assistance and advice on statistical analysis. I thank Drs. Geetha Parthasarathy, Kathryn Jones, Pawin Padungtod, and Jeanie Gaymer along with the other MSU Containment Facility staff and Stacey Wilder for their help with the animal experiments and animal care. I also thank, Alice Murphy and Nicholas Barbu for conducting the 16S rRNA PCR assays. I thank Alice Murphy and Sneha Dhital for performing the ELISA to detect antibodies in the plasma of our pigs. Also, I thank Dr. vi Kathryn Jones for conducting the Real-time PCR and 23S rRNA RF LP analysis and histology. I thank Dr. Geetha Parthasarathy for performing the ELISA to measure anti- inflammatory cytokine concentrations in pig fecal samples. In addition, I thank Shirley Xiao for performing the statistical analysis of my data. The studies for this dissertation project were supported by National Institute of Health funding, and Drs. John Baker and Vilma Yuzbasiyan-Gurkan also helped me to secure funding which supported the completion of my graduate studies. I thank the entire Microbiology and Molecular Genetics departmental staff and the entire National Food Safety and Toxicology Center staff for all of their assistance. Additionally, I am genuinely thankful for Kanika Williams and Mrs. Betsy Walker who provided supreme care for my children while I worked late in the lab. I also thank DeMiracIe Woodson and Carolyn Butler for being my prayer partners and my support angels throughout this journey. I am forever grateful to all that contributed in helping me obtain this degree. vii TABLE OF CONENTS List of Tables .................................................................................................................. ix List of Figures ....................................................................................... x Chapter 1 Literature review .................................................................................. 1 Campylobacter jej uni .................................................................... 1 T richuris suis ............................................................................ 36 Rationale ................................................................................. 53 References ................................................................................ 58 Chapter 2 T richuris suis-mediated cytokine dysregulation in concurrent Campylobacterjejuni and T richuris suis infection ............................ 73 Abstract .................................................................................. 73 Introduction .............................................................................. 77 Materials and Methods ................................................................. 82 Results .................................................................................... 97 Discussion .............................................................................. 108 References ............................. . ................................................. 124 Tables and Figures ..................................................................... 132 Chapter 3 Evaluation of proinflammatory cytokine production in intestinal pig epithelial cells infected with Campylobacterjejzmi ................... 160 Abstract ................................................................................. 160 Introduction ............................................................................. 162 Materials and Methods ................................................................ 166 Results ................................................................................... 173 Discussion .............................................................................. 177 References .............................................................................. 187 Tables and Figures ..................................................................... 193 Chapter 4 Summary and Conclusions ..................................................................... 212 References ............................................................................... 23 1 viii LIST OF TABLES Table 2.1. Experimental treatment groups for the short (2 days) and long (23 days) term challenge experiments .......................... 132 Table 2.2. Inoculation timeline for short term challenge experiments (2 days) ................................................. 133 Table 2.3. Inoculation timeline for I long term challenge experiments (23 days) ................................................ 134 Table 2.4. Summary of Campylobacter detection and identification ................... 135 Table 2.5. Short term challenge experiment 16S RNA PCR analysis results for rapid identification of Campylobacter and Helicobacter DNA in pig feces ...................................... 139 Table 2.6. Long term challenge experiment l6S RNA PCR analysis results for rapid identification of Campylobacter and Helicobacter DNA in pig feces ...................................... 140 Tables 2.7. Short term challenge: clinical assessment of diarrhea in weaned, conventionally-reared swine infected with T. suis and/or C. jejuni ........... 145 Tables 2.8. Long term challenge: clinical assessment of diarrhea in weaned, conventionally-reared swine infected with T. suis and/or C. jejuni ........... 146 Table 2.9. Fecal cytokine recovery rate ..................................................... 147 Table 3.1. IPEC-1 cells treatments for cytokine measurement (ELISA) ............... 193 Table 3.2 Recombinant swine cytokine pretreatment concentrations for invasion and adherence assays ............................................................ 194 ix LIST OF FIGURES Figure 2.1. RFLP analysis of the Campylobacter 23S rRNA gene ...................... 136 Figures 2. 2 A and B. (A) Short term challenge experiment and (B) long term challenge experiment ELISA results indicating levels of plasma IgG antibodies detectable against C. jejuni protein antigen ............ 137 Figure 2.3. ELISA results indicating levels of plasma IgG antibodies detectable against protein antigen of various Campylobacter species, Arcobacter butzleri, and Helicobacter hepaticus, suggesting IgG cross-reactivity .................................................................. 138 Figure 2.4. H&E stained sections from the colons of pigs were examined for T. suis larvae ................................................................ 141 Figure 2.5. H&E section fi'om a C. jejuni infected pig ....................................... 142 Figure 2.6. H&E sections of LGCs from infected pigs ....................................... 143 Figure 2.7. Expression of mRNA from a panel of cytokines was measured by Real Time PCR ............................................................... 144 Figure 2.8. Changes in IL-8 levels were quantified in the feces of weaned piglets fi’om the short term challenge experiments. ............................. 148 Figure 2.9. Changes in TNF-u levels were quantified in the feces of weaned piglets fiom the short term challenge experiments ............................... 149 Figure 2.10. Changes in IL-l-B levels were quantified in the feces of weaned piglets from the short term challenge experiments ............................... 150 Figure 2.11. Changes in IL-4 levels were quantified in the feces of weaned piglets fiom the short term challenge experiments ............................... 151 Figure 2.12. Changes in IL-6 levels were quantified in the feces of weaned piglets fi'om the short term challenge experiments ............................... 152 Figure 2.13. Changes in IL-lO levels were quantified in the feces of weaned piglets from the short term challenge experiments ............................... 153 Figure 2.14. Changes in fecal IL-8 levels were measured using ELISA at various time points over a 23-day period ....................................................... 154 Figure 2.15. Changes in fecal TNF-a levels were measured using ELISA at various time points over a 23-day period ....................................................... 155 Figure 2.16. Changes in fecal IL-l-B levels were measured using ELISA at various time points over a 23-day period ..................................................... 156 Figure 2.17. Changes in fecal IL-4 levels were measured using ELISA at various time points overa23-dayperiod.....................................................157 Figure 2.18. Changes in fecal IL-6 levels were measured using ELISA at various time points over a 23-day period ..................................................... 158 Figure 2.19. Changes in fecal IL-lO levels were measured using ELISA at various time points over a 23-day period ..................................................... 159 Figure 3.1. Standard curve for swine TNF-a used in the ELISA ......................... 195 Figure 3.2. Standard curve for swine IL-l-B used in the ELISA .......................... 196 Figure 3.3. Standard curve for swine IL-8 used in the ELISA ............................ 197 Figure 3.4. TNF-a secretion by IPEC-1 monolayers after exposure to C. jejuni 33292 .................................................................................... 198 Figure 3.5. IL-l-B secretion by IPEC-l monolayers after exposure to C. jejuni 33292 .................................................................................... 199 Figure 3.6 (A and B). The two graphs represent experiments conducted at the same time. (A) Positive control: IPEC-l monolayer positively induced to secrete IL-8 following phorbol myristate acetate (PMA 100 ng/ml) and calcium ionophore (CI 100 ng/ml) exposure. (B) IL-8 secretion by IPEC-1 monolayers after exposure to C. jejuni 33292 ................................................ 200 Figure 3.7. IPEC-1 monolayers were pretreated with increasing amounts of swine rTNF-a for 5 hours to determine the effect of swine rTNF-u on C. jejuni 33292 invasion of intestinal epithelial cells. ....................................... 202 Figure 3.8. IPEC-1 monolayers were pretreated with increasing amounts of swine rTNF-a for 5 hours to determine the effect of swine rTNF-a on C. jejuni 33292 adherence to intestinal epithelial cells ....................................... 203 Figure 3.9. IPEC-1 monolayers were pretreated with increasing amounts of swine rTNF-a for 5 hours to determine the effect of swine rTNF-a on E. coli 0157:H43 DEC7A adherence to intestinal epithelial cells .......................... 204 xi Figure 3.10. IPEC-1 monolayers were pretreated with increasing amounts of swine rIL-l-B for 5 hours to determine the effect of swine rIL-l-B on C. jejuni 33292 invasion of intestinal epithelial cells ......................................... 205 Figure 3.11. IPEC-1 monolayers were pretreated with increasing amounts of swine rIL-I-B for 5 hours to determine the effect of swine rIL-l-B on C. jejuni 33292 adherence to intestinal epithelial cells ....................................... 206 Figure 3.12. IPEC-l monolayers were pretreated with increasing amounts of swine rIL-l-B for 5 hours to determine the effect of swine rIL-l-B on E. coli 0157:H43 DEC7A adherence to intestinal epithelial cells ................................................ 207 Figure 3.13. IPEC-l monolayers were pretreated with increasing amounts of swine rIL-8 for 5 hours to determine the effect of swine rIL-8 on C. jejuni 33292 invasion of intestinal epithelial cells ......................................... 208 Figure 3.14. IPEC-1 monolayers were pretreated with increasing amounts of swine rIL—8 for 5 hours to determine the effect of swine rIL-8 on C. jejuni 33292 adherence to intestinal epithelial cells .......................................................... 209 Figure 3.15. IPEC-l monolayers were pretreated with increasing amounts of swine rIL-8 for 5 hours to determine the effect of swine rIL-8 on E. coli 0157:H43 DEC7A adherence to intestinal epithelial cells .......................... 210 Figure 3.16. Effect of swine rlL-8, rIL-l-B, and TNF-u on transepithelial electrical resistance (TER) of IPEC-1 cells ................................................................ 211 xii Chapter 1: Literature Review I. Campylobacter jejuni Basic microbiology Campylobacterjejuni (C. jejuni) is a gram negative bacillus that has a curved or spiral morphology. Its spiral shape and its polar flagellum at one or both ends of the cell mediates a high degree of darting corkscrew motility (81). It is 6.0 pm long and 0.2-0.5 um wide (81). The generation time of C. jejuni is approximately 90 minutes (104). C. jejuni is nonsaccharolytic and requires enriched media and microaerophilic conditions (3-15% 02, 3-5% C02) for adequate growth (81). Therrnophilic conditions at 42°C facilitate the best growth of these organisms; however, they can also grow at 37°C. These organisms are sensitive to low pH (5 5), organic acids, NaCl concentrations exceeding 2 %, and extended periods at temperatures between 10 and 30°C (104). In unfavorable grth conditions, C. jejuni will convert to a viable, coccoid, nonculturable state. It has been suggested that C. jejuni uses this ‘dormant’-like state as an adaptation mechanism to survive in adverse environments(81). The estimated size of the genome is 1730 kb, based on the first two sequenced strains, and its DNA has a high AT content (81,117). Epidemiology C. jejuni is one of the leading causes of acute gastroenteritis worldwide, exceeding diseases caused by Salmonella spp., Shigella spp., or Escherichia coli 0157:H7 (6). It is estimated that there are 2.1 to 2.4 million cases of human campylobacteriosis occurring each year in the United States, and >99% of these reported infections with Campy/abacter are caused by C. jejuni (6, 7). In most cases, human campylobacteriosis is a sporadic illness (7). Fecal oral transmission in food animals can lead to contamination of meat. Documented sources for C. jejuni are improperly cooked poultry, beef, pork, or turkey. Contaminated water, contact with pets, and international travel are also sources of sporadic infection (7, 27, 81). Although the majority of cases are sporadic, there have been reports of outbreaks associated with the consumption of unpasteurized milk and contaminated water (1, 6, 27, 81, 131). C. jejuni is considered to be a commensal in cattle, sheep, swine, fowl, dogs, cats, and rodents (7). It is now confirmed as a human enteric pathogen. The coupling factor relating C. jejuni to human disease is the transmission of C. jejuni to humans through the consumption of foods of animal origin. Most cases of human campylobacteriosis are associated with foodborne or waterborne transmission (20). In developed/ industrialized countries, Campy/abacter gastroenteritis is most typically characterized clinically by diarrhea, fever, and abdominal cramps. The most common source of Campylabacter infections is the consumption and handling of chicken. Studies conducted in the United States, Europe, and Australia indicated that 50-70% of all Campylobacter infections have been attributed to the consumption of chicken (6). A study conducted in the retail market reported a 98% isolation rate of C. jejuni from chicken meat (7). Children (< 1 year of age) and young adults (age 15-44 years), particularly males, are most commonly infected. The reported seasonal occurrence of C. jejuni infection in the United States begins in May and peaks in August (6). In developing countries studied, Campylobacter infections are hyperendemic. Campylobacter infections occur commonly in both children and adults resulting in asymptomatic infection. Infection with disease is more prevalent among young children (<5 years of age) (6, 112). In developing countries the isolation rates of Campylobacter from patients range from 5 to 20%; these estimates of incidence are from laboratories that survey pathogens responsible for diarrhea. According to the World Health Organization (WHO), the estimated incidence rate of campylobacteriosis is 40,000 to 60,000/ 100,000 in children <5 years old (33). In developing countries, environmental contamination and contaminated foods are considered to be major sources of human infection (33). In many of these settings it is common to have animals living in close proximity to people, and this environment likely plays a significant role in the occurrence of campylobacteriosis. In 2002, Coker et al. reported that the incidence of campylobacteriosis was increasing worldwide, and the WHO turned its attentions to this concern. The increased involvement of the WHO will stimulate public health awareness through strengthened diagnostic facilities for campylobacteriosis and will promote the establishment of national surveillance programs in countries where they do not exist (3 3). These efforts are expected to play a significant role in understanding the global epidemiology of human campylobacteriosis. Disease Symptoms of gastroenteritis associated with Campylobacter infection include diarrhea, fever, and abdominal cramps. These symptoms commence within 2-4 days following ingestion of C. jejuni (81). In most cases, illness is self-limiting, and the duration of the illness is usually 1 week or less. C. jejuni-induced diarrhea occurs following ingestion of as few as 800 organisms (25). In human clinical infection studies, fresh blood, mucus, frank pus, and polymorphonuclear leukocytes were found in diarrheic stool samples (25). However, symptoms can be severe, and death can ensue. According to an estimated case/ fatality ratio for all C. jejuni infections, there is one death per one thousand cases. Usually, C. jejuni-related deaths are associated with immunocompromised individuals (6, 7). Clinical manifestations of disease due to C. jejuni vary between developed and developing countries. In developing countries, the clinical manifestations of Campylobacter enteritis are less severe than in developed countries. Watery, nonbloody, noninflammatory diarrhea is a common clinical characteristic of disease in developing countries. In many cases the patients are underweight and malnourished (33). In developed countries, such as the United States, disease is characterized by severe inflammatory diarrhea with bloody stool, fever, and abdominal pain (33). The optimal drug for treatment of Campylobacter infection is erythromycin. Campylobacter continues to have quite a low rate of resistance to erythromycin despite its use over many years. Erythromycin is safe, low in cost, easy to administer, and has a narrow spectrum of activity (6). Macrolides such as azithromycin and clarithromycin, are also effective against C. jejuni infections, but they are more expensive than erythromycin and provide no clinical advantage (6). Other disease manifestations produced by C. jejuni infections include bacteremia, meningitis, proctitis, septic arthritis, septic abortion, carditis, pancreatitis, urinary tract infection, and autoimmune disease(56, 124). In addition to the immediate effects caused by acute Campylobacter infection, there are long-term sequelae associated with campylobacteriosis. Studies have shown that reactive arthritis (ReA), Reiter’s syndrome, Guillain-Barré, and Miller-Fisher syndrome may accompany or follow C. jejuni enteritis (124). These post-infection sequelae result in serious health effects and have a significant economic impact (105). Reactive arthritis (ReA) is a non-purulent joint inflammation which is commonly induced by enteric infections or urogenital tract infections (56). ReA is triggered by agents such as Salmonella, Yersinia enterocolitica, Shigellaflexneri, Chlamydia trachomatis and Campylobacterjejuni (39). Locht et al. observed that ReA patients that had C. jejuni enteritis had more severe gastrointestinal symptoms and prolonged diarrhea than patients who had enterocolitis only (94). Reactive arthritis is usually self-limiting, but a 3-month course of Ciprofloxacin therapy decreases the symptoms for more severe cases (104). The annual incidence of ReA following Campylobacter infection is 4.3 per 100,000 cases (56). Reiter’s syndrome is another reactive arthropathy associated with C. jejuni infection. This syndrome is thought to be an autoimmune response stimulated by infection. It usually occurs 7-10 days after onset of diarrhea, and multiple joints can be affected, particularly the knee joint. This syndrome triggers pain and possible incapacitation which can last for months or become chronic (7). Reiter’s syndrome is reported to occur in approximately 1% of patients with campylobacteriosis (7). The pathogenesis of Reiter’s syndrome is not completely understood. Guillain—Barré Syndrome (GBS) is another serious sequela of Campylobacter infection. It was the first of the postinfectious autoimmune diseases identified as occurring subsequent to C. jejuni colitis and is characterized by acute limb weakness and loss of tendon reflexes (acute flaccid paralysis) (33, 148). C. jejuni serotype 0:19 are the most prevalent strains associated with GBS worldwide (6, 33, 148). In the United States, the mean annual incidence rate of GBS is 1.3 cases per 100,000 population (148). Although C. jejuni infection can trigger GBS, the risk of developing GBS after C. jejuni infection is quite small (approximately 1 case of GBS out of 1000 C. jejuni infections) (6, 7, 148). Approximately 20% of the patients with GBS are left with some long term disability (7). GBS patients that suffer extensive axonal injury have a greater likelihood of need for mechanical ventilation and an increased risk of irreversible neurological damage (6). Approximately 5% of GBS patients die despite advances in supportive respiratory care (7). It is now recognized that the risk of development of GBS does not increase with the severity of C. jejuni infection. Additionally, many GBS-associated C. jejuni infections are asymptomatic. Neurological symptoms of GBS typically occur l-3 weeks after the onset of diarrheal illness, and humoral irnmunopathogenic mechanisms are believed to be involved (6). Similar to Fisher Syndrome (FS), it is believed that molecular mimicry is a possible cause of GBS. Studies have identified the molecular mimicry between GBS-associated C. jejuni lipo-oligosaccharides and peripheral nerve glycolipids or myelin proteins, particularly the GM] ganglioside, as a mechanism for anti-ganglioside antibody induction (6, 72, 159). It is strongly suggested that this association plays an important role in the pathogenesis of Campylobacter-induced GBS (6, 144). Li et al. demonstrated that C. jejuni-infected chickens spontaneously acquired paralytic neuropathy associated with GBS (93). Likewise, Fisher syndrome (FS), also known as Miller Fisher syndrome, is an anatomically localized variant of Guillain-Barré syndrome (GBS) that is characterized by acute onset of ophthalrnoplegia (paralysis of the eye muscles), ataxia, and areflexia (lack of normal reflexes). It is considered a variant of GBS because some patients who present with FS progress to GBS (148). The estimated annual incidence rate of FS is 0.09 per 100,000 population. C. jejuni is the most fiequently identified antecedent pathogen to F S/GBS. It is believed that molecular mimicry is also a possible cause of both F S and GBS. More specifically, studies have shown that anti-Gle IgG, an antiganglioside antibody, is associated with F S (82, 114, 166). This is thought to occur because Gle is enriched in the cranial nerves that innervate the extraocular muscles (166). Fisher syndrome is typically a benign, self-limiting illness in comparison to GBS, and can be treated successfully with plasmapheresis and intravenous immunoglobulin (114). Model Systems 1). Animal models There have been many attempts at establishing a suitable animal model system that would mimic human campylobacteriosis (4, 10, ll, 19, 44, 100, 116, 129, 154). However, the inability to fillly reproduce clinical symptoms of Campylobacter enteritis has proven to be a hindrance in the progress of clarifying the biological significance of C. jejuni virulence factors. Human campylobacteriosis is characterized by watery diarrhea, abdominal pain, fever and the presence of blood and mucus in stools (6, 16, 33, 81, 95). Various experiments conducted have used animals that are not anatomically and physiologically similar to humans; therefore, requiring anatomical manipulation to acquire disease (4, 44, 73). Also, some animal model systems are quite expensive to maintain and/or have proven to yield inconsistent results (4, 73, 129). In the domestic ferret model it has been shown that some form of specific, aspect of immunity is responsible for at least partial resistance to colonization and disease production although the mechanism has not been identified. Likewise, in this domestic ferret model study, humoral immunity did not appear to play a role in preventing colonization but did protect against enteric disease. Even though the use of the domestic ferret model proved to be informative regarding C. jejuni colonization and immunity, further immunological studies using this model are limited due to the lack of available immunologic reagents and genome sequence (19). Furthermore, other animal models have been used to specifically address certain sequelea associated with C. jejuni infection, such as, Guilain— Barre' syndrome (GBS). C. jejuni-infected chickens have been shown to develop spontaneous paralytic neuropathy associated with GBS (93). Overall, an adequate animal model is needed to suitably assess the bacterial mechanisms and clinical manifestations associated with C. jejuni infection. Despite the prior attempts and criticisms, it is known that the pig is anatomically and physiologically similar to the human, and studies have shown it to be a biologically relevant and important animal to use to observe Campylobacterjejum‘ virulence and host immune mechanisms. Newborn colostrum-deprived piglets that have been orally challenged with an invasive strain of C. jejuni have been successfully used in demonstrating the clinical manifestations associated with campylobacteriosis. The piglets developed clinical symptoms and histopathological lesions similar to that seen in human campylobacteriosis (10). Other swine model systems have proven to be of suitable use as well. For example, Mansfield et. al showed that a synergistic relationship between C. jejuni and T richuris suis (T. suis) enhanced disease and pathology in gnotobiotic (germ-free) pigs (101). Likewise, additional swine disease model studies demonstrated that the lyphoglandular complexes are important colonic sites for irnmunoglobulin A induction against C. jejuni (100). Together, these findings have provided significant support in using pigs as a suitable animal model for human campylobacteriosis, and investigators should now be able to use this system to filrther study virulence factors associated with this enteric disease. To date, there are still unanswered questions although the swine disease model has provided a better understanding of some host immunomodulatory responses triggered by C. jejuni and T. suis infections. Therefore, a murine model system has been developed to continue the advancement in understanding the mechanisms involved in C. jejuni infection (99). Current studies have shown that C57BL/6 and congenic IL-IO deficient mice infected with C. jejuni 11168 are stably colonized and exhibit a robust Th1 directed antibody response, but only the congenic IL-10 deficient mice exhibit significant histopathological changes similar to responses seen in human when infected with C. jejuni 11168. These data suggest that colonization of the mouse gastrointestinal tract by C. jejuni 11168 is required but not adequate for enteritis development. Therefore, C57BL/6 and congenic lL-lO deficient mice can serve as suitable models of C. jejuni colonization and enteritis (99). Hence, the findings from these studies using the swine disease model and the existing mouse model will permit further study to characterize the immune responses that are possibly triggered by interactions between host factors of C. jejuni and/or T. suis which may have an effect on the development of enteric pathogenesis. 2). In vitro models Extensive study of the mechanisms by which Campylobacterjejuni causes disease has not only been investigated in viva, but various in vitro model systems have been developed to facilitate the understanding of the mechanisms involved in bacterial- host cell interaction (160). For example, Caco-2 cells is a cultured cell line that has been used in many C. jejuni experiments to specifically observe bacterial interaction with host cells that differentiate at confluency to mature enterocytes (137, 160). This cell line was derived from a human colon adenocarcinoma and has been used in experiments to mimic the intestinal epithelium. As these cells proliferate and differentiate in culture (maturation process of the intestinal epithelium), they form a polarized epithelial monolayer characterized by brush-border microvilli and tight junctions. When studying differentiation and the regulation of intestinal functions, the Caco-2 model is one of the most used models for in vitro study (64, 78). Likewise, INT 407 cells have been a widely used cell line in C. jejuni/host cell interaction studies. This cell line is derived from human small intestine and has been used experimentally to mimic the firnctions of human undifferentiated intestinal epithelium in response to bacterial exposure (160). In addition, primary swine intestinal cells have been used as a model for studying C. jejuni/host cell interaction. It has been shown that clinical C. jejuni isolates (C. jejuni F1474 and T13 193) invade primary swine intestinal cells at a significantly higher frequency than an Eshcherichia coli control strain. Also, C. jejuni colonies recovered fi'om these primary swine intestinal cells invade tissue cultured cells (INT 407) at a significantly higher frequency than the parental strain (C. jejuni T13192) suggesting that C. jejuni invasiveness may be an important in viva virulence attribute (11). Hence, these 10 findings, in part, have influenced the use primary swine intestinal epithelial cells in ongoing C. jejuni/host cell interaction studies. For example, IPEC-1 cells (intestinal pig epithelial cells) are cells derived from neonatal pig intestinal epithelium. These cells have not been cloned, and this leads to some diversity of the population; however, these cells can be induced to differentiate by culturing them for 12-14 days. It has been shown that IPEC-l cells secrete IL-6, lL-lO and IL-18 following C. jejuni exposure (Parathasarathy, dissertation, 2004 and Jones, dissertation, 2005). Also, several other in vitro studies have demonstrated the roles of cytokines in C. jejuni infection. Viable C. jejuni stimulates INT 407 cells to secrete IL-8, and this response has been shown to be mediated via multiple mechanisms such as C. jejuni adherence and/or invasion or C. jejuni cytolethal distending toxin (CDT) treatment (61, 63). Other proinflammatory chemokines, such as GROalpha, GROgamma, macrophage inflammatory protein 1 (MCP-l), and gamma lP-10, are secreted from INT 407 cells following exposure to viable C. jejuni and are believed to play a contributing role in the observed host inflammatory responses (70). Likewise, it has been shown that viable C. jejuni activates the mitogen-activated protein kinase pathways and the transcription factor, NF-KB, which regulates the transcription of inflammatory cytokines and chemokines (97). Viable C. jejuni also stimulates INT 407 cells to produce intracellular IFN-y, IL-10, TNF-a, and IL-4 suggesting that viable C. jejuni is capable of generating a dissociated Th1 and Th2 immune response (5). Likewise, C. jejuni-infected human dendritic cells (DCs) trigger NF ~1cB activation, stimulates IL—l-B, IL-6, IL-8, TNF-q, IL- 10, IL-12 and IFN-y production, and induces DC maturation (69). Also, C. jejuni interaction with cultured human monocytic cells elicits IL-1 -[3, IL-6, IL-8, and TNF ~01 ll induction and triggers NF-KB nuclear translocation (77). In addition, C. jejuni has been shown to invade avian macrophages and primary chick kidney cells and stimulate the production of nitric oxide (NO), IL-l-B, IL-6, IL-8 and K60 (143). Therefore, it is evident that C. jejuni-host cell interactions have been studied using multiple well. established in vitro models. Pathogenesis Intestinal Colonization Colonization is a key factor involved in bacterial survival in the different microenvironments of the gastrointestinal tract of humans and animals and is necessary but not sufficient for production of enteritis (146). Many commensal bacteria asymptomatically colonize the intestinal tracts of animals used as a food source (146, 164). Several studies have demonstrated the importance of C. jejuni colonization to its persistence (79, 98, 110, 149). In order to survive as an intestinal commensal or foodborne pathogen, C. jejuni must be able to adapt to and survive in different microenvironments especially those of the gastrointestinal tract. This capability is likely controlled by a two-component regulatory system (TCR), which is important for the coordinate regulation of gene expression (28). It has been suggested that this novel TCR system plays an important role in the growth and colonization of C. jejuni . This RacR-RacS (reduced ability to colonize) system is believed to be involved in a temperature-dependent signaling pathway. A mutation of the response regulatory gene, racR, reduced the ability of C. jejuni to colonize the chicken intestinal tract and resulted in temperature-dependent l2 changes in protein profile and growth characteristics (28). It is suspected that other TCR systems may be involved in optimal in viva C. jejuni colonization. For example, a dccRS two-component system (diminished Qapacity to golonize) was found to be important for in viva colonization of immunodeficient limited flora (SCID-LF) mice and l-day old chicks, but not relevant for in vitro growth or survival in epithelial cells (98). MacKichan et al. demonstrated that mutations in the dccR response regulon and the dccS sensor kinase reduced colonization in l-day old chicks and SCID-LF mice models and had no significant effect on C. jejuni growth and/or survival in epithelial cells (98). In addition, several genes encoding putative periplasmic and membrane proteins that are regulated by this two-component system have been identified. After comprehensive analyses, the genes involved in the RacR-RacS and dccRS two-component systems (racR and dccRS) proved to be essential for optimal colonization (98). Likewise, it has been suggested that C. jejuni outer membrane glycoproteins may act as adhesins thereby promoting colonization (79). Mutation of the ngH gene involved in C. jejuni N-linked general protein glycosylation showed reduced C. jejuni adherence and invasion of human epithelial Caco-2 cells and significant colonization reduction in l-day old chicks (79). In addition, chemotaxis and motility are believed to be significant virulence factors involved in controlling the mechanisms of C. jejuni colonization of the intestinal tract (These factors will be discussed in greater detail later in the literature review.) (149). Overall, these studies have made significant advances to the knowledge needed for the development of intervention strategies to control transmission of this foodbome pathogen. However, despite progress, further studies are needed to better understand the processes involved in C. jejuni colonization. l3 Virulence factors The characterization of C. jejuni virulence factors has been intensively studied over the past few years because C. jejuni is known to be the most common cause of gastroenteritis worldwide (6, 33). Significant progress has been made in identifying and characterizing virulence factors of C. jejuni; however, the molecular mechanisms involved in pathogenesis remain unclear. Herein, I will discuss a few of the virulence properties that have been recognized and studied by researchers to elucidate the pathogenic mechanisms of C. jejuni. Motility and Chemotaxis As mentioned in the colonization discussion, one of the best-characterized virulence determinants of Campylobacters is flagella-mediated motility. Campylobacter spp. flagella production is necessary for motility (81). It is believed that the spiral-shaped cellular structure and the flagellum, in combination, give Campylobacters an uncommonly high level of motility in viscous environments (81, 91). C. jejuni is motile by the action of one or two polar flagella. It has two copies of the flagellin gene (flaA and flaB). Two unique promoters control flagellin gene expression, and at least one of the genes responds to environmental signals (147). It has been shown through targeted mutagenesis that the flaA gene is the main gene for flagellin production and motility (75, 162). For instance, disruption of flaA produces a short, truncated and nonfirnctional flagellum that has greatly reduced motility, lacks the ability to invade INT 407 cells, and is unable to colonize the ceca of three-day-old chicks (110, 157, 162). However, when 14 there is a disruption of flaB, the flagellum length is nomial, and there is normal flagella function, but slightly decreased motility as compared to the wild type strain (81). Motility of C. jejuni is known to be phase variable, but the mechanism of this variation remains unclear. Specific genes identified as the motility accessory factor (mat) family of flagellin-associated proteins are related to flagellar biosynthesis and phase variation. It has been suggested that these maf genes may confer on C. jejuni flexibility in adapting to changing environmental conditions when flagellar expression and motility are undesirable (80). The maf genes may also be involved in reversible expression of flagella, which could be beneficial in evading the host immune response (1 l l). The reversible expression of flagella in C. jejuni means that the genes involved in flagellum synthesis and mobility can to switched on and off depending on the changing environmental conditions (111). For instance, flagella genes may be initially switched on to allow the flagellum to act as an adhesin and acquire attachment to the host cell. However, once attached and colonization has been established, then flagella formation may be switched off(111). It has also been suggested that the regulation of C. jejuni motility is associated with quorum sensing. C. jejuni produces functional autoinducer-2 (AI-2) cell-signaling activity, and this AI-2 production is regulated by the luxS gene. AI-2 production allows bacteria to assess the population density of other species of bacteria and communicate with these other species of bacteria (75). Studies have demonstrated that inactivation of the quS gene causes C. jejuni to become less motile than the wild type (43). Conversely, quorum sensing has been observed to be related to C. jejuni motility. Quorum sensing in C. jejuni influences flaA transcription and autoagglutination, which is known to be 15 closely associated to the existence of functional flagella in C. jejuni (75). Additional evidence pertaining to the importance of the fla regulon has been acknowledged by the recognition of a F lgS/F lgR two-component signal transduction system. The FlgS sensor and the FlgR response regulator form this system, and it has been shown to be required for the initial stages of chicken cecal colonization, but not for survival and persistence. The signal that triggers this two-component system to turn on the fla regulon has not yet been identified (161). Chemotaxis is also required for effective colonization and is an important virulence property. The ability Campylobacters have to detect chemical gradients along with motility functions enable the cell to move up or down the gradient (81). The importance of chemotaxis has been demonstrated in viva; chemically mutagenized or non-chemotactic C. jejuni failed to colonize suckling mouse intestine (149). Two adjacent genes have been identified to control chemotaxis, a methyl-accepting chemotaxis protein (MCP) and a putative cytochrome c peroxidase. These were identified using signature-tagged transposon mutagenesis of C. jejuni 81-176 followed by testing for chick gastrointestinal tract colonization (59). It is suggested that MCP is required for proper chemotaxis to a specific environmental element, and the putative cytochrome c peroxidase may function to reduce periplasm hydrogen peroxide stress during in viva growth. Loss of either gene function resulted in attenuation of growth throughout the gastrointestinal tract of chicks (60). This particular study was one of the first genetic screens involved in identifying the bacterial gene requirements necessary for establishing a commensalistic relationship with a natural host. Also, it is clear from 16 recent studies in murine models that although colonization is not sufficient for disease following oral C. jejuni infection, it is required for disease to be manifested (99). Adherence Bacterial adherence to target cells plays a critical initiating role in pathogenesis. Bacterial adhesins interact with the intestinal epithelium and help pathogenic bacteria resist peristalsis and the fluid flow of lumenal contents. Once bacteria are adhered to the intestinal epithelium, the bacteria can form colonies, release enzymes and toxins, invade cells, and/or alter intracellular function to their own advantage (68). Therefore, it is suggested that adherence is an important determinant of virulence for various pathogenic bacteria (50). The interaction between adhesins and the host are believed to be a prerequisite for bacterial penetration of the mucosal surface. Several Campylobacter spp. adhesins have been identified and characterized since this genera was discovered. Various in vitro studies have shown that some Campylobacter spp. adhere to cultured cells (106, 127, 141). In 1986, flagella were identified as a potential C. jejuni adhesin by comparing adherence of flagellated and aflagellated variants to INT 407 cells (106). McSweegan et al. showed that removal of the flagella by shearing the bacterial cells reduced adhesion. Lipopolysaccharide (LPS) was also identified as another C. jejuni adhesin. In vitro C. jejuni LPS specifically bound to epithelial cells and intestinal mucus gel (106). These findings support the importance of flagella and LPS in initiating mucosal surface adherence. 17 Other approaches have been used to determine the significance of campylobacter adherence. For example, Yao and colleagues used an insertional mutagenesis method for naturally transformable organisms to characterize non-adherent and non-invasive C. jejuni strains. They generated a series of C. jejuni strain 81-176 mutants with a kanamycin-resistant cassette placed in various regions of the open reading frame of the flaA gene encoding flagellin. By this means, they identified strains that had defects in motility, displayed reductions in invasion levels, and lacked the ability to adhere to the target cells. They suggested that flagellin can serve as a secondary adhesin, and other adhesins mediate a motility-dependent internalization process (162). In addition, these generated mutants were used to identify a cheY gene that was responsible for the non- adherent and non-invasive phenotypes observed in their experiments. Yao et al. also showed that the diploid cheY strain was able to colonize mice, but was attenuated in a ferret disease model (163). The results of the ferret disease model suggested that virulence was reduced due to the presence of excess CheY. This also implied that the 2 copies of cheY may affect subsequent C. jejuni interactions with target epithelial cells in the colon rather than affecting the ability of the organism to survive in the mucus lining of the small intestine (163). As mentioned, cultured cells and animal models have been used extensively to demonstrate C. jejuni adherence to and invasion of epithelial cells (30, 168). Various studies have led to the identification and characterization of other adhesins (76, 84, 122, 141). For example, a Campylobacter adhesion to fibronectin (CadF), a C. jejuni 37 kDa protein, was identified as an adhesin. Various in vitro studies indicated that CadF was a conserved outer membrane protein that mediated the specific binding of C. jejuni to the 18 extracellular matrix component, fibronectin (84). Likewise, other experiments demonstrated the activity of another C. jejuni adhesin identified as a 43-45 kDa C. jejuni major outer membrane protein (MOMP). The interaction between C. jejuni MOMP and cultured INT 407 cell membranes showed significant binding activity, and this interaction suggested that the MOMP firnctioned as an adhesin and mediated bacterial-host tissue contact (141). leA, a C. jejuni specific novel surface-exposed lipoprotein, was another identified adhesin that mediated adherence to host epithelial cells. Insertion and deletion mutants of jlpA demonstrated that adherence to HEp-2 cells can be reduced compared to parental C. jejuni TGH901]. Likewise, anti-GST-leA inhibits C. jejuni adherence to HEp-2. Also, an irnmunoblotting assay showed that leA binds to HEp-2 cells, implying that leA is a C. jejuni adhesin. From these findings, it has been suggested that lipoproteins may play a role in bacterial infection because they induce tumor-necrosis factor and cause macrophages to release interleukin-6. It has been suggested that J lpA may stimulate host cytokine production because it is released into the culture medium. However, this function remains to be established and could have multiple effects on the immune system (76). Furthermore, in vitro and in vivo models were used to characterize PEBl as an adhesin that enhances C. jejuni interactions with epithelial cells. To identify the role of PEBl adhesion activity, pebIA was inactivated by allelic exchange, and used to challenge cultured cells. The inactivation of the pebIA locus significantly reduced C. jejuni adherence toHeLa cells, which suggested that PEBl was an important C. jejuni adhesion (122). This study also provided information about the PEBI adhesin function in C. jejuni invasion and colonization. PebIA locus inactivation significantly reduced C. jejuni invasion of epithelial cells in culture and reduced the rate and duration of mouse 19 intestinal colonization as compared to the C. jejuni wild type isolate. Together, these data indicated that PEBl plays an important role in bacterial-epithelial cell surface interactions (122). In summary, the identification and characterization of these adhesins have proven to be beneficial in establishing a better understanding of C. jejuni host cell interactions. According to these studies, adhesins play important roles in mediating C. jejuni attachment to host epithelial cells. They also suggest that some adhesins may play a significant role in virulence and host resistance and immunity. Further studies will contribute to understanding the relationship between C. jejuni adhesion and infection. Invasion Campylobacter-host cell interactions have been described as intriguing and mechanistically unique (160). Over the years, researchers have been persistently trying to understand the mechanisms by which Campylobacter invade host cells. Both in vitro and in vivo models have been used to establish the invasive potential of C. jejuni (10, 21, 23,29,46,57,61) In vivo studies have shown that C. jejuni invasiveness is clearly correlated with presentation of disease in the host (10, 21). In one particular study, newborn piglets were orally inoculated with a strain of C. jejuni isolated from a patient. The clinical and histopathological damage was similar to that observed in that patient and in other humans with campylobacteriosis. I-Iere, internalized bacteria were only observed within the intestinal cells of these C. jejuni-infected piglets, and there was disruption of the microvilli in the infected area (10). In various studies, cultured mammalian intestinal 20 epithelial cells have been used to demonstrate the process of C. jejuni invasion (86, 87, 107, 135). For example, Konkel and colleagues identified a Campylobacter invasion antigen (CiaB) protein that affects C. jejuni invasion and shows similarities to type III secreted proteins (86). Further studies indicated that maximal invasion of eukaryotic cells and Cia protein secretion depended on intact flagellar synthesis genes, thus suggesting that Cia protein export occurs through the flagellar transport and/or assembly apparatus. Collectively, these findings suggested that the flagellar type III secretion pathway is required for C ia protein export (87). In addition, it has been suggested that the Cia proteins are secreted during colonization, and they may play a significant role in C. jejuni colonization (22). Konkel and colleagues also identified, CadF, a fibronectin— binding protein that is required for the binding of C. jejuni to intestinal epithelial cells. They also found that this interaction enhanced bacterial internalization (84). Monteville et al. found that maximal C. jejuni entry required the binding of CadF to fibronectin which thereby triggered host cell signaling events associated with bacterial uptake. In this study, INT 407 cells were infected with a C. jejuni wild-type isolate and at 30 and 45 minutes post infection, there was an increase in the level of tyrosine phosphorylated paxillin (a focal adhesion signaling molecule)(107). Likewise, ij29 an unknown glutamine-rich protein, was another protein required for C. jejuni invasion, and it was found inside INT 407 cells, suggesting that it may be secreted by a type IV secretion system ( 12). Additional studies have been carried out in order to determine if invasion is a significant virulence factor in facilitating C. jejuni enteritis. Researchers have examined the possible roles of microfilaments and microtubules in the C. jejuni invasion process 21 (23, 71, 89, 113). Treatment of intestinal epithelial cells (INT 407 cells) with cytochalasin-D (a chemical that causes microfilament depolymerization) and colchicines and nocodazole (chemicals that cause microtubule depolymerization) inhibited C. jejuni invasion of INT 407 cells in a dose-dependent manner. There was a more pronounced inhibitory effect of microfilament depolymerization on C. jejuni invasion than that of microtubule depolymerization. Similar observations have been reported on Klebsiella pneumoniae enterohemorrhagic E. coli, enteropathogenic E. coli and E. coli associated with new born meningitis (23, 24). These overall findings suggest that the mechanisms involved in C. jejuni invasion is associated with the combined effect of microfilaments and microtubules of host cells (23). C. jejuni vaccine strain 81-176 has two plasmids (pTet- an R factor plasmid encoding tetracycline resistance and the other, pVir, encoding the cag pathogenicity island usually found in Helicobacter pylori) that have been characterized and are believed to affect the virulence of C. jejuni by encoding a type IV secretion system. Specific mutations in some of the identified plasmid genes (comB3 and virBI I) have been shown to significantly reduce invasion of INT 407 cells (13). Other analyses indicated that there are four additional pVir genes (ij15, ij29, ij32, and ij49) which need further characterization using mutational analyses followed by testing in animal disease models, but they have been found to significantly affect C. jejuni invasion into intestinal epithelial cells. These four genes encode proteins that are envisaged to be soluble and once invasion occurs are located in the cytoplasm. Further studies are being done to elucidate the possible effector protein roles of this putative type IV secretion system (13). 22 The C. jejuni invasion process is multifactorial and influenced by many cellular processes (21). This invasion process appears to be C. jejuni strain dependent and host cell-type dependent. Freshly isolated clinical strains also appear to be more invasive (36). Invasion of the host cell is an important virulence mechanism of C. jejuni, and the molecular mechanisms mediating host cell invasion are gradually being elucidated. Intracellular survival Intracellular survival is definitely a key factor in the pathogenesis of C. jejuni; however, the mechanisms by which C. jejuni persist in the host cell have not yet been defined. Various studies have attempted to demonstrate the entry process, intracellular survival and location of C. jejuni (24, 35, 36, 85, 86). Using transmission electron microscopy and the gentamicin killing assay, DeMelo et a1. observed cellular events and intracellular survival of C. jejuni in Hep-2 cells (3 6). At one hour post infection, DeMelo and colleagues noticed C. jejuni cell surface and microvilli interactions which possibly imply a phagocytic-like mechanism preceding C. jejuni internalization (36). Also at one hour post infection, there was notable evidence of host cytoskeletal rearrangement and the presence of C. jejuni enveloped within endocytic vacuoles in the cytoplasm of the host cell (23, 36, 83). Once internalized and about 9 hours postinfection, phagolysosome fusion occurred, and lysosomes assembled and co-localized in the cytoplasmic matrix. This lysosomal event could possibly explain the loss of C. jejuni viability and the change in the morphological structure from spiral to coccal form at this stage (83). Konkel et al. and Biswas et al. also did studies to firrther characterize C. jejuni internalization and intracellular survival. They concluded that C. jejuni internalization of INT 407 cells was 23 affected by both, host microfilament and microtubule depolymerization (23, 24, 83). In addition, Konkel et al. found that ammonium chloride and methylamine did not inhibit endosomal acidification and did not affect C. jejuni internalization by INT 407 cells (83), but Biswas et al. showed that monenesin significantly inhibited endosome acidification, thereby reducing the number of intracellular C. jejuni (24). Biswas et al. also showed that viable intracellular C. jejuni were reduced in number by treating INT 407 cells with G-strophanthin and monodansylcadaverine which indicated interference of receptor- mediated endocytosis (62). It is known that C. jejuni has the ability to persist and multiply in epithelial cells and phagocytic host cells (35, 36, 85, 123). Konkel et al. showed that intracellular C. jejuni can elicit a cytotoxic effect on INT 407 cells causing deterioration of the cells (85). The weakening effects on the cell monolayers are believed to represent a pathogenic mechanism associated with C. jejuni enteritis (85). Likewise, Hickey et al. showed that C. jejuni strain 81-176 is capable of replicating extensively within human monocytic cell vacuoles. They also suggested that viable intracellular C. jejuni induce apoptotic death via secreted C. jejuni cytolethal distending toxin (62). Similar to the findings of Guerry et al., Siegesmund et al. showed that C. jejuni Cia-secreted proteins are required for maximal apoptosis induction of monocytic cells in vitro (142). It has been suggested that these secreted proteins are associated with a type 111 system of C. jejuni, and are responsible for intestinal epithelial cell invasion, and possibly play a role in C. jejuni enteritis (142). Siegesmund et al. also showed that the apoptotic death of C. jejuni- infected monocytic cells is caspase-l-independent and is not dependent on the concurrent 24 secretion of IL-l-B, which is caspase-l dependent (142). However, it is unclear at this time as to how macrophage apoptosis is associated with C. jejuni-mediated enteritis. There have been several studies which have given insight to the possible mechanisms of intracellular survival (35, 123). For instance, it has been demonstrated how C. jejuni iron superoxide dismutase (SOD) enzyme contributes to intraepithelial cell survival (123). This enzyme catalyzes the breakdown of superoxide radicals to hydrogen peroxide and dioxygen and acts against oxidative stress in the bacterial cell thereby contributing to the defense mechanisms of the bacterial cell (123). To date, only one sod gene has been identified in C. jejuni, which is similar to other reported FeSODs. In one particular study, Pesci et a1. constructed isogenic sod mutants which proved to be significantly more sensitive to intracellular killing than wild-type C. jejuni strains thereby suggesting that SOD plays a role in C. jejuni intracellular survival (123). Likewise, C. jejuni expresses a single catalase enzyme identified as katA (35). It is believed that this catalase also plays a significant role in C. jejuni intracellular survival. Day et al. constructed katA mutants to address this hypothesis, and the findings of this study suggested that catalase plays a significant role in C. jejuni intramacrophage survival by providing resistance to hydrogen peroxide(35). Together, the findings of Pesci et al. and Day et al. suggest that organism persistence inside host cells is in part due to bacterial factors that combat reactive oxygen species and play a role in intracellular survival (35, 123) . 25 Lipopolysaccharide/Lipoolygosaccharide In gram-negative bacteria, lipopolysaccharide (LPS) is a major constituent of the outer membrane, and it is composed of three distinct structural components: lipid A, which anchors the membrane; an oligosaccharide core; and the 0 antigen composed of one or more glycosyl residues that covalently attach to the core (88). LPS molecules that lack O-antigen units and have core oligosaccharides structures limited to 10 saccharide units are referred to as lipooligosaccharides (LOS) (130). It has been suggested that C. jejuni LPS and LOS are excellent targets for the control of infectious diseases and for diagnostic purposes. In vitro and in vivo studies have demonstrated the functions of LPS and LOS in C. jejuni virulence (55, 108, 128, 167). It is believed that C. jejuni LPS and LOS mimic host ganglioside structures, specifically components of the perpherial nerve tissue (55). More specifically, LOS has been shown to play an important role in the pathogenesis of gastroenteritis due to C. jejuni. Phase variation of C. jejuni 81-176 LOS has been demonstrated in vitro and in vivo, and the ability of C. jejuni to undergo phase variation in LOS biosynthesis genes may provide selective advantages in causing gastrointestinal disease (5 5). It has been suggested that phase variation of LOS biosynthesis allows a single bacterial strain to produce a repertoire of LOS molecules that differ in core length and carbohydrate content due to the addition of glycose groups at the nonreducing termini (128). In vitro, slip strand mismatch recombination of cgtA, a gene that encodes N-acetylgalactosaminyl (GalNAc) transferase, resulted in changes in the major core structure from GM; to GM; ganglioside mimicry. Also, site-specific insertional inactivation of the cgtA gene of C. jejuni LOS resulted in a mutant that mimics GM3 and enhances internalization and 26 invasion of intestinal epithelial cells in vitro thereby suggesting that changes in LOS structure may directly affect the ability of C. jejuni to cause gastroenteritis (55). Also, variation in the sialylation of C. jejuni LOS was demonstrated via site-specific mutagenesis of the neuCI gene resulting in the loss of sialic acid in the LOS core, increased irnmunogenicity of the core, and decreased resistance to normal human serum (53, 55, 128, 167). Antigenic phase variation in ganglioside mimicry has also been demonstrated in viva (128). In a volunteer experimental oral infection study, Prendergast et al. found that the LOS of C. jejuni isolates underwent antigenic phase variation in ganglioside mimicry during passage in vivo, and serological testing showed that antiganglioside antibodies were not present following experimental C. jejuni infection or following administration of 3 killed C. jejuni whole-cell vaccine (53, 108, 109, 167). They concluded that their findings were consistent with in vitro reports that have shown that C. jejuni possesses the genetic mechanisms for its LOS to undergo phase variation. They also believe that these LOS phase variation responses could cause difficulties in developing successful vaccines because C. jejuni can synthesize ganglioside mimics and has the ability to convert between different LOS structures (128). These observations also suggest that the ability of C. jejuni to undergo LOS phase variation may be advantageous to bacterial persistence in the host. Hence, the changes that can occur in the major core of C. jejuni due to bacteria] phase variation suggest that C. jejuni LOS core structures are dynamic, and there are indications which imply that there may be differences in the mechanisms by which C. jejuni strains cause diarrhea] disease (55). Further study is needed to elucidate C. jejuni phase variation and how these variations may play a role in C. jejuni-induced gastroenteritis. 27 Additionally, more studies have been conducted to elucidate the association between C. jejuni LOS and GBS (109). It has been demonstrated that molecular mimicry between human ganglioside and C. jejuni LOS play a relevant role in the pathogenesis of GBS (53, 109). Nachamkin et al. found that est-II (or-2,3 and/or d-2,3/ 0,-2,8 sialyltransferase), cgtA (Bl,4-N-aetylgalactosyltransferase) and cgtB ([31,3- galactosyltransferase)are LOS biosynthesis genes involved in C. jejuni LOS core extension and appear to be critical to C. jejuni ganglioside mimic expression associated with GBS-related C. jejuni isolates(21, 167). Also, based on LOS locus analysis and knockout mutants, Godschalk et a]. suggested that specific classes of the LOS biosynthesis gene locus or specific types of genes involved in sialic acid biosynthesis and transfer may be crucial for the induction of neuropathogenic cross-reactive antibodies (53). Various studies have presented evidence implying that the development of auto- antibodies raised against distinct C. jejuni surface LPS and LOS is involved in the pathogenesis of Guillain-Barre syndrome(GBS)(55, 108, 166). In rabbits sensitized with C. jejuni LOS, anti-GM] IgG antibodies were detected and flaccid limb weakness developed, which resembles clinical signs of GBS in humans. The pathological studies of the sciatic nerve specimens of the paralyzed rabbits showed Wallerian-like degeneration identical to that seen in GBS patients (167). In addition, immune naive mice injected with C. jejuni LOS generated monoclonal antibodies (mAb) that reacted with GM] and bound to human peripheral nerves. In general, these findings suggest that carbohydrate mimicry between C. jejuni LOS and human gangliosides are important for the development of GBS. 28 Overall, C. jejuni LPS and LOS appear to play an important role in the pathogenesis of C. jejuni-mediated enteritis and autoimmune diseases such as GBS, and further evaluation of these particular virulence determinants (LPS and LOS) may elucidate specific mechanisms involved in C. jejuni’s ability to persist in host cells causing pathology and disease. Toxins Campylobacter spp. that produce toxins have been isolated from various human and animal sources throughout the world. In attempts to study toxigenicity, researchers usually correlate toxin production with clinical presentation and in vitro analysis (156). Since C. jejuni is one of the leading causes of acute diarrhea in humans in developed countries and has been isolated from healthy and diseased young children in developing countries, I have limited the discussion to this species (156). There are two types of diarrhea induced by C. jejuni: l) a noninflammatory, watery diarrhea without leukocytes or blood and 2) an inflarmnatory, bloody diarrhea containing leukocytes. In both of these clinical presentations, diarrhea] disease caused by C. jejuni is believed to be associated with toxin production. Enterotoxins and cytotoxins are two classes of proteinaceous toxins that differ in their primary mode of action. Enterotoxins are secreted proteins with the ability to bind to a cellular receptor, enter the cell, and elevate intracellular cyclic AMP (CAMP) levels. This elevation of CAMP causes changes in the cellular ion flux, which leads to excessive secretion of fluid, resulting in watery diarrhea (156). Cytotoxins kill target cells by inhibition of cellular protein synthesis and actin filament formation or by forming pores 29 in target cell membranes. Cytotoxins cause inhibition of cellular protein synthesis by inactivating ribosomes by depurination of a nucleotide in the 28S rRNA, and this inhibition leads to cell death. Also, these secreted proteins can damage the intestinal mucosa and cause diarrhea by covalently modifying Rho proteins resulting in the disruption of the actin cytoskeleton. Another cytotoxic mechanism can be directed specifically towards nucleated cells. For instance, the cytotoxin forms pores in the nucleated cell thereby inducing cytokine release, cytoskeleton dysfunction, secretion of granule constituents, and generation of lipid mediators. These profound local and distant effects in host tissues may lead to an overall diminution of the immune responses of the host (156). C. jejuni enterotoxin activity has been demonstrated in cell culture by the elongation of Chinese hamster ovary (CHO) cells, rounding of mouse adrenal tumor cells (Y-l), or by a measurable increase in intracellular CAMP in exposed cells (156). Likewise, Ruiz-Palacios et al. showed that supernatant of a prototype virulent C. jejuni strain causes the induction of fluid secretion in the rat ileal loop test (RILT), but not in the rabbit ileal loop or the infant mouse assay (136, 156). It has been suggested that enterotoxin production is responsible for the watery diarrhea form of campylobacteriosis, as opposed to the other form with inflammatory, bloody diarrhea (156). However, controversy surrounds C. jejuni enterotoxin activity because, to date, no gene encoding this product has been found. Also, the inability to consistently reproduce some of the in vitro and in viva assays used to detect enterotoxin activity continues to fuel the controversy. These concerns will not be resolved until the enterotoxin structural gene has been cloned, sequenced, and the firnctional experiments are proven to be reproducible. 30 C. jejuni cytotoxins are also not well characterized. Several studies have evaluated the correlation between cytotoxin production and clinical manifestations. It has been suggested that cytotoxin-producing Campylobacter strains are more commonly isolated from patients with inflammatory diarrhea than other strains (156). Various cell lines, bacterial culture conditions, and bacterial strains have been used to study Campylobacter spp. cytotoxin production (156). Although diverse methodologies have been used to classify Campylobacter spp. cytotoxin production, to date, some of the research has not been reproducible (125). However, to date, the cytolethal distending toxin (CDT) is the best characterized C. jejuni toxin. In 1996, the cdt genes from C. jejuni strain 81-176 were cloned and sequenced (125). Lara-Tejero et a]. discovered that the cdt genes are required to form a tripartite holotoxin that promotes CDT activity (90). They found that 3 subunits, Cth, CdtB, and CdtC, make up the C. jejuni CDT holotoxin, and all subunits are required for complete toxic activity (90). It has been observed that Cth and CdtC bind the host cell and comprise CdtB. The CdtB subunit has been shown to be the active enzymatic subunit which is imported into the nucleus and functions similar to a DNAse 1 enzyme (32, 88, 92). It is proposed that the CdtB subunit damages the DNA and inhibits the dephosphorylation of Cdc2, a key kinase regulator of cell cycle progression (90). C. jejuni CDT causes elevated intracellular CAMP levels, blocks the Gz/M phase of eukaryotic cells prior to cell division, induces cytoplasm distension, and eventually leads to cell death (32). This cell cycle arrest activity has been demonstrated in experiments using a yeast model system that showed that CdtB induced G2 cell cycle arrest accompanied by degradation of the host cell chromosomal DNA (58). 31 In addition to these findings, it has been suggested that C. jejuni CDT plays a role in host cell immunomodulation. In vitro, CDT stimulates cultured human intestinal epithelial cells to produce IL-8 (63). In an in viva challenge experiment, mutant C. jejuni lacking the cdtB gene successfully colonized NFKB deficient mice producing gastritis but caused much less severe disease than the isogenic parent wild type C. jejuni. This finding suggests that CDT has a proinflamatory effect similar to CDT effects reported in vitro (51). Also, IgG2a levels were lower in mice infected with the CDT mutant than the mice infected with the wild type strain (51). These findings suggest that CDT plays a significant role in stimulating an immune response and in developing disease. Toxin production by Campylobacter spp. has been a very complex virulence property to classify, and the difficulties with Campylobacter spp. toxin classification may reflect significant strain variation in this species. There has been a continuous effort towards developing in vitro and in viva methodologies to determine the modes of action of putative toxins (165). Although there have been several reports of toxin producing Campylobacter spp., the correlation with disease and the mechanisms of action require firrther study. Immune responses The mechanisms by which C. jejuni elicit a host immune response are not well understood. In developing countries, it is believed that healthy infants, children and adults are constantly exposed to Campylobacter antigens in the environment, and consequently, there is early development of serum antibodies to Campylobacter species (33). However, most cases in developed countries occur later in life and are usually 32 primary, sporadic infections with more severe and prolonged clinical manifestations, and the immune response related to disease is not clear (33). Therefore, the host immune responses to campylobacteriosis differ among people in developing and developed countries, and various studies have been conducted to determine the importance of C. jejuni immune responses and host factors that affect the outcome of disease. Humoral immunity is necessary to resolve C. jejuni infection. During C. jejuni infection, the levels of all immunoglobulin classes rise. However, IgA is one of the most important isotypes because it can cross the gut wall and immobilize organisms in the gut (155). In a swine disease model, a significant efflux of immunoglobulin A (IgA) was detected in the enlarged germinal centers of the lymphoglandular complexes (LGCs) of C. jejuni-infected pigs (100). This immune response suggests that there are important colonic sites worthy of further immunological consideration to elucidate mechanisms of resistance or susceptibility to disease. In addition, Cawthraw et a]. detected significant IgA antibody levels in the postinfection serum, saliva and urine of patients with C. jejuni infections (31). Also, serum and salivary antibodies against flagellin were detected suggesting that flagellin is an important surface antigen against which immunity is directed (19). In a domestic ferret model system, humoral immunity does not appear to play a role in preventing colonization but does protect against enteric disease. Taken together, these findings suggest that a Th2-type immune response can in part play a protective role during C. jejuni infection. T-cell-mediated responses to C. jejuni infection appear to be of relevance as well. Rhijn et a]. demonstrated peripheral blood 75 T cell expansion following the exposure of patients to crude sonicates of five different C. jejuni serotypes (153). This expansion was 33 dependent on the presence of CDT/(18+ T cells in cultures or the addition of exogenous IL-2 or IL-15, and 75 TCR mediated the C. jejuni-specific stimulation (153). Also, several in vitro studies have demonstrated the roles of cytokines in C. jejuni infection. Viable C. jejuni stimulates the secretion of IL-8 by INT 407 cells (human intestinal epithelial cells), and this response is mediated via multiple mechanisms, 1) C. jejuni adherence and/or invasion or 2) C. jejuni cytolethal distending toxin (CDT) treatment (61, 63). Other proinflammatory chemokines, such as GROalpha, GROgamma, macrophage inflammatory protein 1, MCP-l, and gamma lP-lO, are secreted by INT 407 cells due to exposure to viable C. jejuni and are believed to play a contributing role in the observed host inflammatory responses (70). Furthermore, it has been shown that viable C. jejuni activates the mitogen-activated protein kinase pathways and the transcription factor NF ~1 0.05). Cytokine levels are expressed as mean (in picograms per milliliter) i standard error of the mean (SEM). Differences were considered statistically significant if P _<_ 0.05. 96 Results C. jejuni isolation and detection We were unable to isolate Campylobacterjejuni from the fecal cultures of any pigs before or after infection (Table 2.4). To enhance sensitivity of detection, we used PCR assays to determine the presence of C. jejuni in the tissue samples taken separately from the jejunum, proximal colon, and distal colon. Polymerase chain reaction (PCR) using specific primers for the QRDR of the gyrA gene of C. jejuni did not detect C. jejuni in any tissue samples from any pig group, except in 1 out of 6 pigs in the T. suis-infected group (Table 2.4). However, Campylobacter 23S rRNA RF LP analysis suggested that C. jejuni, C. coli, and an unidentified Campylobacter species that gave a unique RFLP pattern were present in our animals (Figure 2.1). These findings in pigs handled rigourously as specific pathogen free by various methods lead us to firrther explore these results. Immunologic recognition of C. jejuni To confirm C. jejuni infection in experimentally inoculated pigs and to discriminate between C. jejuni and related epsilon proteobacter infection in preinoculated and control pigs, we pursued ELISA measurements of bacteria specific IgG. Figures 2.2 A and B show that IgG antibodies in the plasma of all the pigs in each treatment group from the 2nd repetition of the short term challenge experiments (days 0, l and 2) reacted with C. jejuni antigen (samples were not available item the first independent experiment). IgG antibodies in the plasma of most pigs in the treatment groups fiom the long term (days 0, 2], 22 and 23) challenge experiments (from 2 independent 97 experiments) also reacted with C. jejuni antigen. The ELISA was conducted using the plasma samples collected 2 days post C. jejuni inoculation on day 2 (short term challenge) and day 23 (long term challenge) of the experiments. Given that we detected high levels of anti-Campylobacter IgG antibodies in the plasma of our pigs and that it has been shown that the induction of an I gG response requires more than 2 days post- Campylobacter infection (about 8—14 days)(23), our data suggest that the pigs had established humoral immune recognition of C. jejuni before being experimentally infected. Therefore, we also wanted to distinguish whether these IgG antibodies measured in the pig plasma were C. jejuni specific or resulted fi'om cross-reactions with other Campylobacter spp. or other environmental Campylobacter-like bacterial antigens. After randomly selecting plasma samples from some of the pigs in the short (days 0, 1 and 2) (2nd repetition only) and long term (days 0, 21, 22 and 23; from 2 independent experiments) challenge experiments, we found that IgG antibodies in the plasma of 8 pigs randomly chosen fiom the C. jejuni-infected groups (positive control) and 6 pigs randomly chosen from the pig groups that received milk alone reacted with C. jejuni, C. lari, C. coli, C. upsaliensis, C. hyointestinalis, A. butzleri, and H. hepaticus antigen by ELISA (Figure 2.3). Conversely, plasma samples taken fiom 2 newborn piglets (< 6 hours old) at the MSU swine farm (negative control) did not elicit any IgG response against Campylobacter spp., A. butzleri, and H. hepaticus antigen. In these studies of our experimental pigs, the greatest IgG response was directed against H. hepaticus antigen (3 related epsilon proteobacteria), suggesting that these bacteria or closely related Helicobacters were present in reasonably high numbers in the pigs prior to inoculation 98 with C. jejuni. Once again, these results stimulated further studies to distinguish whether the pigs carried a Helicobacter spp. Recognition of Campylobacter spp. and Helicobacter spp. in feces In order to confirm the previous ELISA findings, we performed subsequent species-specific PCR assays to identify Campylobacter spp. or Helicobacter spp., which could have possibly had an effect on the cytokine immune response of the pigs. We conducted Campylobacter 16S rRNA PCR analysis and found that Campylobacter species were undetectable in the fecal samples fiom any pig group before or after bacterial infection (Tables 2.5 and 2.6). However, after Helicobacter 16S rRNA PCR analysis, we found that Helicobacter DNA could be detected in the feces of some of the pigs from the uninfected control group, suggesting that they were likely exposed to Helicobacter species (Campylobacter-like bacteria) prior to any experimental exposure to C. jejuni (Tables 2.5 and 2.6). The presence of these closely related bacteria likely contributed to the cross-reacting IgG antibodies detected in the plasma of the pigs by ELISA. The IgG cross-reacting antibodies may have contributed to the immune response against the experimentally-administered C. jejuni, thereby having an effect on the outcome of the cytokine responses elicited throughout the study. In the pig, Helicobacter spp. are found mame in the stomach and small intestines of swine so larger effects are expected in these compartments. 99 Histopathology “Haemotoxylin and Eosin (H&E) stained sections of jejunum, proximal colon, and distal colon of each pig were examined microscopically for the presence of T. suis larvae and histological changes due to infection. T. suis larvae were seen in proximal colon sections from 5 of 6 pigs in group 3, and 5 of 6 pigs in group 4. No larvae were seen in groups 1 or 2 (Figure 2.4). Control animals (Group 1) had no proliferative, inflammatory, or degenerative lesions in any of the tissues examined and were therefore used as a normal comparison for all infection groups. All lesions were confined to the colon. Colonic lesions due to C. jejuni or T. suis infection were similar to those described in germ free piglets (50). Pigs in group 2 (C. jejuni only) developed focal mild crypt distension with sloughed epithelial cells, mucus and bacteria in the crypts in the distal colon (Figure 2.5A) and LGCs (Figure 2.6A). There was no significant inflammatory infiltrate into the lamina propria on day 23 after inoculation. Pigs infected with T. suis only developed mild to moderate colitis with goblet cell hyperplasia and crypt distension with mucus in superficial lamina propria. Lesions were found only in the areas where T. suis larvae were located. A mixed inflammatory infiltrate was present in the lamina propria and included lymphocytes, plasma cells, neutrophils, and eosinophils (Figure 2.5 B). Where present, crypts of LGCs were not distended (Figure 2.6 B). Pigs infected with both C. jejuni and T. suis developed mild colitis with goblet cell hyperplasia and crypt distension with mucus in superficial lamina propria in the areas where T. suis larvae were located. A mixed inflammatory infiltrate in the lamina propria included lymphocytes, plasma cells, neutrophils, and 100 eosinophils. Entrapped crypts of the LGCs were distended with mucus and few sloughed epithelial cells” (Figure 2.6 C). (Jones, dissertation, 2005, reprinted with permission) Clinical observation All piglets were observed prior to infection and prior to fecal sampling to assess clinical signs. Diarrhea was the main clinical sign and was assessed as any evidence of liquid feces ranging from soft stool to pipe stream diarrhea. The results are summarized in Tables 2.7 (short term challenge experiment) and 2.8 (long term challenge experiments). The occurrence of diarrhea was also indicated by subjective evaluation of collected loose feces and large amounts of loose fecal discharge in the cages of the pigs. Over a 2-day period (short term challenge), the occurrence of diarrhea was assessed on days 0, l and 2 during fecal sampling. Pigs given C. jejuni only had 2 episodes of diarrhea, and the dually infected pigs that received C. jejuni and T. suis had 5 episodes of diarrhea. There were no signs of clinical diarrhea in the uninfected pigs, the T. suis only infected pigs, or the noninvasive E. coli DHS-a infected pigs. The occurrence of diarrhea was also assessed on days 0, 21, 22 and 23 of fecal sampling for the long term challenge experiments and more diarrheic episodes were seen. Diarrhea was most prominent in the pigs infected with both T. suis and C. jejuni (19 episodes) and the pigs given T. suis only (20 episodes). Pigs given C. jejuni only had 6 episodes of diarrhea over the observation period. Pigs that were uninfected or received noninvasive E. coli DHS-a had no signs of diarrhea. 101 Cytokine expression following Real-time PCR analysis “We measured expression of mRN A fi'om 15 cytokines, chemokines, and iNOS in each segment of intestine sampled in an effort to determine if T. suis or C. jejuni had a significant effect on cytokine expression, and to determine if the affected cytokines were indicative of a specific response. Cytokine expression patterns were distinct in each segment of the intestine tested (Figure 2.7). In the proximal colon of the T. suis only and dual infection groups, IL-13 expression was increased 3 to 4 fold while IL-12 and IL-8 expression was decreased 2 to 4 fold when compared to uninfected controls. In the dual infection group, expression of IL-5 and MCP-l were each decreased 2 fold compared to controls. These data indicate that T. suis stimulates IL-13 in its local environment while decreasing expression of proinflammatory cytokines. In the distal colon of pigs infected with T. suis only, expression of IL-l-B, IL-6, IFNy, and IL-13 was increased 3 to 4 fold over controls. Infection with C. jejuni only or dual infection with C. jejuni and T. suis did not significantly affect expression of any cytokines, chemokines, or iNOS in the distal colon. These data indicate that a predominately proinflammatory response has been initiated in the distal colons of pigs infected with T. suis only. In the jejunum, expression of MCP-1, TNFa, IL-12p40, IFNy, IL-4, and IL-10 were increased 3 to 5 fold in the C. jejuni only group when compared to controls. Expression of GM-CSF was decreased 5 fold in the dual infection group when compared to controls. These data indicate that Campylobacter species present at low levels in the 102 tissues or related Campylobacter-like bacteria stimulate a predominately proinflammatory response in the jejunum, while T. suis may be down regulating proinflammatory cytokines.” (Jones, dissertation, 2005 reprinted with permission) Cytokine rate recovery from feces Table 2.9 shows the recovery rate of exogenously added recombinant IL-8, IL-1- [3, TNF-a, IL-4, IL-6, and IL-10 present in the feces. In general, these exogenously added recombinant cytokines were recovered from feces at low rates (IO-20% recovery). These findings suggest that the low recovery rate may have an impact on accurately measuring cytokine expression in the feces of the pigs. Additionally, preliminary experiments showed that when water was experimentally evaporated from either solid or loose fecal samples, there were no significant changes in the weight of the samples. Thus, these results suggested that the consistency of the feces would not likely have an effect on adequately measuring changes in fecal cytokine levels (data not shown). Cytokine Secretion—short term challenge (2 days) IL-8, IL-l-B, TNF-a, IL-4, IL-6, and IL-10 concentrations were measured in the feces of all piglets throughout the experiment. However, there were no significant changes in IL-8, IL-l-B, TNF-d, IL-4, IL-6, and IL-10 expression due to any of the administered treatments (IL-8, P e 0.56; IL-l-B, P = 0.27; TNF-a, P = 0.15; IL-4, P = 0.26; IL-6, P = 0.45; IL-10, P = 0.79). The results are shown in Figures 2.8 and 2.9 and 2.10 and 2.11 and 2.12 and 2.13, respectively. These results represent the combined data of two independent experiments. 103 Cytokine secretion—long term challenge (23 days) IL-8 secretion IL-8 was detected in all fecal samples throughout the experiment (23 days). The results are illustrated in Figures 2.14. By day 23, pigs infected with C. jejuni alone showed a significant increase in IL-8 expression compared to levels on day 0 (pre- inoculation; P = 0.02); however, this increased expression was not statistically significant compared to the milk only control group on days 0, 21, 22 and 23 (P > 0.05). The T. suis only infected group showed a significant decrease in IL-8 expression by day 23 compared to levels on day 21 (P = 0.01) and day 22 (P = 0.01), but this decrease was not significantly different (P > 0.05) compared to day 0 and the milk only control group on day 23. Also, E. coli DHS-a did not induce any changes in IL-8 expression. Therefore, the overall changes of IL-8 expression due to the administered treatment over a period of time were insignificant with a P value of 0.08. These results represent the combined data of 2 independent experiments. TNF-a secretion TNF-a was detected in all fecal samples. The results for these experiments are shown in Figure 2.15. The effects of each treatment on TNF-u expression over a period of time showed moderate changes within the treatment groups. The pigs infected with C. jejuni showed a significant increase in TNF-a expression from day 0 to days 22 and 23, and the E. coli DHS-a group showed a significant increase in TNF-a expression fiom day 0 to day 22, but these increases in TNF-a expression were not significantly different in comparison to the milk only control group. From day 0 to day 23, the pigs that were 104 given milk only showed a significant increase in TNF-a. From day 21 to day 22, the T. suis group showed an increase in TNF-u and a decrease from day 22 to day 23, but there were no significant differences compared to day 0 or to the milk only control group. In addition, the pigs infected with C. jejuni and T. suis showed a significant increase in TNF-a expression from day 0 to day 22 and then showed a decrease by day 23; however, these changes were not significant compared to the milk control. Hence, the overall changes in TNF-a expression due to the administered treatment over 23 days were not significant in comparison to the milk only and day 0 control groups with P values > 0.05. IL-l-B secretion IL-l-B was detectable in all stool samples throughout the experiment (23 days). The results are shown in Figure 2.16. By day 23, the pigs infected with T. suis alone showed a significant decrease in IL-l-B expression compared to day 0 and the milk only control group on days 0, 21, 22 and 23. This result indicates that infection with this helminth was successfirl, and triggered down-regulation of a pro-inflammatory cytokine. The T. suis group also showed significantly decreased IL-l-B expression on day 23 compared to the pigs that received C. jejuni. The dual infected group and the E. coli DHS-a group showed a significant decrease in IL-l-B expression by day 23 compared to day 0, but the decrease was not significant when compared to the milk only group. Furthermore, the significant decrease in IL-l-B expression implies that T. suis suppressed the IL-l-B response. These results represent the combined data from 2 independent experiments. 105 IL—4 secretion IL-4 was detectable in all stool samples throughout the experiment (23 days). The results are shown in Figure 2.17. The effects of each treatment on IL-4 expression showed moderate changes within the treatment groups. By day 23, T. suis-infected pigs showed a significant decrease in IL-4 expression compared to IL-4 levels on days 0, 21, and 22. Also, pigs given C. jejuni, E. coli DH5-a, or C. jejuni and T. suis showed a decrease in IL-4 expression by day 23 compared to levels on day 0 within each of the treatment groups. However, the overall changes of IL-4 expression were not statistically significant compared to the milk only control group (P 2 0.05). These results represent the combined data of 2 independent experiments. IL-6 secretion IL-6 was detectable in all stool samples throughout the experiment (23 days). There were no significant changes in IL-6 expression due to the administered treatments (P > 0.05). The results are shown in Figure 2.18. IL-10 secretion IL-10 was detectable in all stool samples throughout the experiment (23 days). The results are shown in Figure 2.19. There were some moderate changes in IL-10 expression within the treatment groups throughout the experiment. Pigs given C. jejuni, T. suis, or C. jejuni and T. suis showed increased IL-10 levels by day 22 compared to day 0. Also, E. coli DHS-u-infected pigs showed an increase in IL-10 expression by day 22 that continued through day 23. These moderate changes in IL-10 expression within the 106 treatment groups were not significant when compared to the IL-10 levels detected in the pigs given milk only (P > 0.05). 107 Discussion In our Campylobacter swine disease model, there have been repeated observations suggesting that there is enhanced disease and pathology in the gut mucosal epithelium and LGCs in response to a concurrent infection with C. jejuni and T. suis (49, 50)(Jones, dissertation, 2005). In this study, clinical signs and histopathology also support an interaction between these two pathogens. The most severe clinical signs and histopathological lesions were observed in dually infected pigs. Lesions were similar to that seen previously with germ free pigs with major changes observed in the proximal colon surrounding the worms and in the distal colon in the LGCs (50). It is thought that cytokine dysregulation plays a significant role in this response. In the present study, we hypothesized that weaned piglets infected with C. jejuni demonstrate an innate protective immune response via pro-inflammatory cytokine up-regulation and simultaneous T. suis infection mediates the down-regulation of the pro-inflammatory cytokine response, thereby promoting C. jejuni infection. In this study, we used standard culture procedures, PCR assays, and immunological testing to determine whether the weaned piglets used in our experiments had been exposed to C. jejuni, other Campylobacter species, or Campylobacter-like organisms (epsilon proteobacteria) at the MSU swine farm facility prior to experimental infection. The piglets apparently had low level exposures to Helicobacter spp. We must consider the possibility that the piglets were also exposed to related Campylobacter species or other environmental Campylobacter-like bacteria from the sow. This was an important factor because it may have caused control pigs to have falsely elevated 108 cytokine responses elicited following exposure to Helicobacter. Also after experimental infections, we attempted to use standard culture methods to identify C. jejuni prior to and throughout the experiment, but in spite of our best efforts, culture alone was not sensitive enough to detect C. jejuni. Others have also found it difficult to recover C. jejuni from feces by culture alone and have used PCR assays and irnmunohistochemical examination to detect C. jejuni (20, 79). Because of problems detecting C. jejuni in pigs after inoculation, some researchers in recent work used culture of supernantants from 5-10 gram samples of feces to enhance the sensitivity (Moller-Nielson, personal communication). To increase the specificity and sensitivity of detection in our studies, we used C. jejuni specific gyrA Real-time PCR (TaqMan) for detection and RF LP analysis of the Campylobacter 23S rRN A gene for thermophilic Campylobacter species differentiation. TaqMan analysis of post infection tissue samples fi'om necropsy gave negative results for most pigs, but a positive result for C. jejuni in distal colon samples fiom 16% (1 of 6) of the pigs that received T. suis-only. In follow-up analysis of post infection samples from necropsy, 23S rRNA RF LP analysis gave positive results for C. jejuni in jejunum samples fi'om the dually infected pigs (16%, 1 of 6) and in proximal colon samples from 16% (1 of 6) of the pigs that received T. suis-only. Likewise, C. jejuni was detected in proximal colon samples from 66% (4 of 6) of the dually-infected pigs. RF LP analysis also showed positive results for C. coli and a possible Campylobacter spp. that gave a unique banding pattern that was not consistent with C. jejuni, C. lari, C. coli, or C. upsaliensis in jejunum, proximal and distal colon samples. These RFLP findings conflicted somewhat with the gyrA TagMan assay results and suggested that multiple Campylobacter species could have been present in the colon of 109 these pigs. RF LP results also implied that the pigs may have been exposed to Campylobacter spp. or Campylobacter-like organisms (epsilon proteobacteria) prior to the experiment. Subsequent immunological testing only partially clarified the issue. It demonstrated the presence of IgG antibodies against C. jejuni protein antigen in the plasma of all pigs in the short and long term challenge experiments. In addition, we detected significant levels of IgG antibodies that reacted with antigen from other Campylobacter species and Campylobacter-like bacteria (Arcobacter butzleri and Helicobacter hepaticus) in the plasma of some of the pigs given C. jejuni (a total of 8 pigs randomly selected from the short and long term challenge) or milk only (a total of 6 pigs randomly selected from the short and long term challenge). These findings further suggest that these pigs may have had a previous exposure to an epsilon proteobacterium possibly C. jejuni or other closely related member of the group with similar outer membrane proteins. It has been shown in humans that IgA and IgG are produced in response to C. jejuni infection, and serum and salivary IgG antibody responses are detectable up to 1 year post-infection while long-term IgA antibody responses are not detectable ( 14, 27). The greatest IgG reaction was obtained with H. hepaticus protein antigen, thereby suggesting that this was the main contaminating bacterium that was present in these pigs. It also suggests that the IgG antibodies detected in the plasma of the pigs cross-reacted with the protein antigen of other Campylobacter species and Campylobacter-like bacteria. Because of the short time period between initial bacterial inoculation (short term challenge-day 0; long term challenge-day 21) and sampling (short term challenge-day 2; long term challenge-day 23), the experimentally inoculated pigs would not have enough time to trigger an IgG response against the administered C. jejuni. 110 As in C. jejuni-infected humans, it has been shown that IgG antibody titers generally peak at approximately 8-14 days postexposure, but in some cases may not peak until 3 to 4 weeks postexposure, and remain elevated and persist at high levels for several weeks or months in patients (23, 45). Taken together, it appears that the pigs had been exposed to the putative contaminating bacterium (epsilon proteobacteria), possibly a Helicobacter spp., at birth or shortly after birth at the MSU swine farm prior to the experiment. Indeed, these ELISA results show increased levels of IgG antibodies reacting with multiple bacterial protein antigens, including C. jejuni, other Campylobacter species, A. butzleri, and H. hepaticus, and could not distinguish between the occurrence of cross reactivity and infection with multiple epsilon proteobacteria. Because no published work exists comparing homologies between outer membrane proteins of bacteria within this group, we cannot distinguish between these possibilities based on these assays. In a final attempt to distinguish the identity of the early bacterial exposure in these pigs, we tested fecal supernatants for the presence of other Campylobacter species and Helicobacter species using 16S rRNA PCR analysis. Here we found that almost all pigs in the short- and long-term challenge experiments tested negative for the presence of Campylobacter species pre- and post-infection. However, several pigs tested positive for the presence of Helicobacter species pre- and post-infection. This data provides the best evidence to suggest that the pigs were exposed to Helicobacter spp. prior to the experiment and that the cytokine responses measured were likely affected by this prior bacterial exposure. This also suggests the possibility that the effects seen actually represent host resistance to secondary bacterial infection. In this scenario, it is likely that these pigs acquired immunity after early exposure to an epsilon proteobacteria such as Helicobacter, and 111 elicited a humoral immune response which provided partial protective immunity to subsequent C. jejuni challenge. For example, it is possible that systemic IgG arising from early exposures may have bound to the experimentally administered C. jejuni or to shed C. jejuni surface material and limited C. jejuni-enterocyte interactions which is the main mechanism for increased inflammatory cytokine production in bacterial infections (38, 39, 42). In the short-term challenge experiments, there were no significant changes in secreted IL-8, IL-l-B, TNF-a, IL-4, IL-6, or IL-10 expression due to any treatment when compared to the uninfected pigs that received milk only. We hypothesized that C. jejuni exposure would trigger an increase in the expression of pro-inflammatory cytokines, such as IL-8, IL-l-B, and TNF-a, and simultaneous T. suis infection would stimulate increased production of anti-inflammatory cytokines, such as IL-4, IL-6, and IL-10. Based on literature and previous findings in our laboratory, we believe that the anti-inflammatory cytokine response can down regulate the pro-inflammatory cytokine response and thereby facilitate C. jejuni infection. Consequently, the dysregulation of cytokine expression due to this concurrent infection could cause immunomodulatory effects that may contribute to the observed disease and pathology, which is similar to that seen in campylobacteriosis. However, because the data here provide some evidence suggesting that these pigs were exposed to Helicobacter spp. or a Campylobacter-like bacteria prior to the experiment, it is reasonable to believe that the administered C. jejuni was a secondary bacteria] challenge. Therefore, in the groups of pigs that received bacteria alone (C. jejuni only or E. coli DHS-a only), the cytokine expressions measured were likely a result of host resistance to secondary bacterial infection. In the short- term challenge experiment, we 112 found that C. jejuni alone did not induce the expected increase in secreted pro- inflammatory cytokine expression in feces. There were also no significant changes in cytokine expression in the pigs that received E. coli DHS-u only. Likewise, in a previous study, Withanage et al. demonstrated that there were no significant changes in the expression of proinflammatory cytokines or chemokines, such as IL-IB, IL-8, or K60, in the gastrointestinal tract of chickens following secondary Salmonella infection. However, there was notable expression of MIP family chemokine and IL-6 in the ileum and cecal tonsils (76). Additionally, in our study, we found that there were increased levels of IgG in the plasma of almost all the pigs regardless of treatment that reacted in the C. jejuni ELISA. It has been shown in previous studies that significant levels of circulating IgG antibodies specific for Salmonella were high in infected chickens following primary infection and remained high with little change following secondary infection (15, 17, 76). In addition, significant levels of specific IgG antibodies have been detected in patients with Campylobacter enteritis, and IgG levels have been reported to be detectable in most patients up to a year post infection (23, 27). Therefore, our results appear to be similar to previous reports that suggest that humoral responses may play a significant role in protective immunity during homologous and heterologous re-challenge (16). The current finding also raises speculation about the possibility that there may be cross-reactive humoral immune responses to Campylobacter species or other episilon proteobacteria that are associated with protective immunity. As for the two groups of pigs that received the T. suis in the short-term challenge (day O-day 2), the results were not surprising. Studies have shown that clinical signs mostly begin 13-14 days following T. suis infection. The severe, bloody diarrhea associated with swine trichuriasis from the 113 20th day onwards is due in part to the burrowing activities of the larval and adult stages in host colonic tissues and secretion of proteolytic enzymes and proteases (ESP). During this phase of T. suis development, the posterior end of the helminth fi'eely protrudes into the lumen while the anterior body is embedded in the mucosa (18). At this phase, the third stage larvae have just moulted to the fourth larval stage. Following the direct mucosal damage by the helminth, localized lesions have been observed surrounding the site of worm attachment. It is at this time that resident bacteria have been demonstrated to amplify the severity of the infection (3, 4, 56, 77). The evidence provided by these reports suggests that T. suis acts synergistically with C. jejuni in producing the pathogenesis of swine mucohemorrhagic colitis. Thus, it is reasonable to believe that results fiom the long-term challenge experiments (23 days) would likely reflect the immune response patterns of natural infections more than that seen in the short-term challenge experiments (2 days). In the long-term challenge experiments (day 0-day 23), we found that T. suis infection significantly decreased secreted IL-l-B by 23 days post-inoculation. Also, on day 23, IL-l-B was down-regulated in the pigs that received T. suis alone compared to the pigs that only received C. jejuni. Furthermore, the dually infected pigs showed moderately decreased IL-l-B expression by day 23, but the response was not significant. The significant decrease seen in IL-l-B expression in response to T. suis alone is characteristic of most helminth infections. In general, helminth infections elicit a Th2 response which induces the production of IL-4, IL-5, IL-6, IL-9, IL-10, and IL-13 during infection (33, 74). This pattern of cytokine expression is characteristic of a Th2 response which predominantly limits Th1 cytokine-induced inflammation (Th1 cytokines- Il-12, 114 IFN-y, IL-8, TNF-a, and IL-l-B) and protects the host by causing worm expulsion, reducing worm fecundity and enhancing host resistance mechanisms (33, 44). Tissue samples were also taken fi'om the pigs in the long-term challenge experiments on day 23 following fecal sampling, and using these samples, Real Time PCR analysis showed increased IL-l3 mRN A (Th2 cytokine) in the proximal colon of T. suis-infected pigs and decreased IL-8 and IL-12 expression (Th1 cytokines) (Jones, dissertation, 2005). This supports the hypothesis that T. suis stimulates Th2 anti-inflammatory cytokine production which can down-regulate the production of Th1 pro-inflammatory cytokines. We have also found in our laboratory that intestinal pig epithelial cells stimulated with T. suis ESP secrete IL-6 and IL-10 (60). Therefore, it is possible that the down regulation of IL-l-B in response to T. suis alone is likely due to IL-4, IL-10 and IL-13 production in response to the helminth infection. In vitro and in vivo studies have demonstrated IL-4, IL-10 and IL- 13 inhibition of intestinal inflammatory responses such as Thl-type cytokine production (22, 46, 48, 68). Taken together, our fecal cytokine ELISA results suggest that T. suis likely plays a significant role in the down-regulation of IL-l-B production, and this response may play a contributing role in altering host conditions which promote secondary bacterial infection. However, these results need to be confirmed in fiirther studies because of the low cytokine recovery rate (IO-20%) determined for these fecal ELISAs. Several factors may have contributed to the low cytokine recovery rate: protein degradation associated with freezer storage, the loss of protein during the fecal preparation due to the cytokines binding to the solid fecal matter, or protein degradation due to protease activity. Because of the drawbacks associated with measuring cytokines in feces using ELISA, we are unable to confidently support the future use of ELISA for 115 this purpose and believe that our results fiom this analysis are notable, but must be confirmed with further experimentation. Nevertheless, in the long-term challenge experiments, cytokine mRN A expression measurements in the tissues of these weaned piglets were more informative. We observed significant changes in cytokine expression in different intestinal compartments based on treatment group, specifically the jejunum, proximal and distal colon. T. suis resides in the proximal colon while C. jejuni has been found in all of these sites. We found that T. suis infection predominantly influenced cytokine expression in the proximal colon with some effects in the distal colon. C. jejuni mainly affected the cytokine expression in the jejunum with minimal changes in the colon. In the proximal colon, T. suis stimulated increased IL-13 expression (3 to 4 fold) in the pigs that received T. suis alone or those that were dually infected, and IL-8 and IL- 12 expression was decreased 2 to 4 fold in both groups when compared to the uninfected control pigs that received milk only. MCP-1 expression was also decreased in the dually- infected pigs. This is similar to other results that show that during intestinal nematode infection, IL-l3 plays a significant role in affecting Th-2-cell-mediated responses which are associated with goblet cell hyperplasia and worm expulsion (11, 37, 52). Th-2 responses are associated with the production of anti-inflammatory cytokines such as IL-4, IL-5, IL-6, IL-9, IL-10 and IL-l3, and these anti-inflammatory cytokines can down regulate Th-l responses (IFN-y, IL-2, IL-12, lymphotoxin, and IgG2a production) which subsequently have a diminishing effect on pro-inflammatory cytokine responses (IL-8, TNF-a, MCP-1, IL-1, and IL-6) (25, 28, 59). These types of cytokine modulations can generally be seen during concurrent bacterial-helrninth infections. Previously reported data from our laboratory showed that pigs rectally challenged with C. jejuni showed 116 increased IL-8 expression at 1 and 24 hours post-inoculation suggesting the importance of IL-8 production in early host resistance to C. jejuni in immunologically naive swine (Jones, dissertation, 2005). It has also been shown that C. jejuni stimulates cultured intestinal epithelial cells (INT 407) to secrete IL-8 and MCP-l (38, 42). Likewise, Helicobacter pylori stimulates cultured gastric epithelial cells to secrete IL-8 and MCP-1 (35, 58, 61). In another enteric bacteria with similar pathogenesis, it has been demonstrated in a rabbit ligated intestinal loop model of Shigellaflexneri infection that IL-8 appears to play a critical role in controlling the spread of infection (67). Because bacteria have been shown to invade and proliferate in the lamina propria and submucosa surrounding the site of worm attachment in previous studies (50), we expected to see increases in proinflammatory cytokines like IL-8 in the proximal colon of pigs given C. jejuni alone. However, no changes in IL-8 were observed in either study design associated with C. jejuni infection alone in this tissue. This may have been affected by the suspected early exposure to Helicobacter spp. in these studies or perhaps the inoculated strain of C. jejuni invaded mame the small intestines based on the proinflammatory responses measured there. Based on the tissue RT-PCR results it is also possible that T. suis contributed to decreased IL-8 expression and may have facilitated bacterial invasion and proliferation in the lamina propria and submucosa, similar to what is observed in the Shigella model. IL-13 production due to T. suis infection likely contributes to the decreased expression of IL-8, given that it has been demonstrated that the production of IL-8 fi‘om intestinal epithelial cells can be inhibited by IL-13 and IL-4 (48). 117 We also found that expression of both IL-12 subunits was significantly decreased in the proximal colon of pigs given T. suis. IL-12 is predominantly produced by monocytes, macrophages, and dendritic cells and plays a role in inflammation and acquired immune responses (36, 71). IL-12 drives the development of Thl-type immune responses by stimulating T cells to produce IFN-y, and in vitro and in vivo studies have shown that IL-12 plays a significant role in developing specific immunity against intracellular pathogens (36, 71). However, IL-l3 has been shown to down-regulate IL-12 production (54). Increased expression of IL-13 has been documented in resistant mice strains infected with T. muris suggesting its critical protective role in nematode infection (1 1). Taken together, decreased expression of both IL-l2 subunits in the proximal colon of T. suis-infected pigs can be attributed to the immunomodulatory effects of IL-l3. Similar to the decreased expression of pro-inflammatory cytokines in the proximal colon, IL-5 expression was significantly decreased in the dually infected pigs. In general, IL-5 is up-regulated along with other classical Th2 cytokines during helminth infections, and IL-5 has been shown to characteristically induce eosinophil production and activation (32). However, experimental studies have shown that C. jejuni intestinal colonization is reduced in Balb/c ByJ female mice that are orally pretreated with IL-5, suggesting that IL-5 may play a protective role for the host (13). It is possible that C. jejuni elicits the down regulation of IL-5 as a mechanism to evade host immune responses and establish infection. In the proximal colon of the pigs that received C. jejuni only, IL-5 expression was decreased, but the decrease was not statistically significant. Even so, the decreased expression of IL-5 observed in the dually-infected pigs and the C. jejuni-infected pigs suggests that this cytokine may play a significant role 118 in the pathology associated with C. jejuni infection in vivo and that firrther studies into this hypothesis are warranted. Unlike the decreased pro-inflammatory cytokine response observed in the proximal colon, IL-l-l3, IL-6, IFN-y and IL-13 were up-regulated in the distal colon of the pigs infected with T. suis only. We speculate that this immune response is likely due in part to the effects of T. suis ESP in the distal colon. The up-regulation of IL-l-B, IL-6, IFN-y is indicative of an inflammatory response. It has been demonstrated in vitro that ESP treatment of differentiated IPEC-1 cells causes a dose dependent cytotoxic response and significantly decreased transepithelial electrical resistance (TER)(1, 2). Also, cultured differentiated and undifferentiated IPEC-l cells secrete IL-6 following ESP exposure (Parthasarathy, 2005). Therefore, it is reasonable to consider that the cytotoxic effects of ESP on intestinal epithelial cells can stimulate an inflammatory cytokine response in the distal colon. Another possible reason for the inflammatory response observed in the distal colon is secondary bacterial infection that is augmented by T. suis infection. In the distal colon of T. suis-infected pigs in previous studies, several resident bacterial species, including C. jejuni, C. coli, an unidentified Campylobacter species, and E. coli, have been cultured fi'om abscessed lymphoglandular complexes (51). It has been shown that C. jejuni and C. coli stimulate cultured human INT-407 intestinal epithelial cells to secrete IL-8 (39). Therefore, it is possible that bacterial invasion of the distal colon tissues in this study initiated an inflammatory response and was responsible for the histopathological changes in the LGCs. It has been shown that some components of the ESP consist of antibacterial agents, and a possible scenario is that the antibacterial effects of ESP could kill residential bacteria thereby making the host environment more 119 conducive to facilitating the proliferation of pathogenic bacteria. Once the ESP antibacterial activity has killed the residential bacteria, these pathogenic secondary bacteria could more effectively interact with the intestinal epithelium and stimulate these cells to produce proinflammatory cytokines. Also, the effects of ESP produced by 4th stage larvae that have traveled to the distal colon could contribute to increased expression of IL- 1 3. It has been shown that T. suis-infected pigs have serum antibodies as early as 21 days post inoculation that recognize a 20kDa glycoprotein isolated fiom ESP collected from adult worms cultured in vitro (41). This indicates that an immune response is likely triggered by the larval stages present in pigs 21 days post-T. suis infection. Altogether, these data suggest that ESP may have direct effects on intestinal epithelial cells and could account in part for the increased cytokine production in vivo at sites distal to worm attachment. In the jejunum, the pigs infected with C. jejuni only showed increased expression of MCP-l, TNF-u, IL-12p40, IFN-y, IL-4, and IL-10. Although we did not confirm the presence of C. jejuni or other Campylobacters in this tissue by culture, results from the histopathology, immunological testing (IgG ELISA), and the 23S rRNA PCR analysis suggest that it is still likely that the inoculated strain of Campylobacter triggered this response. C. jejuni is notoriously difficult to recover from colonized pigs without disease because of low bacterial numbers and the preferred predilection site that is strictly at the base of mucosal crypts. In vitro, C. jejuni induces INT 407 cells to produce intracellular MCP-l, TNF-a, IFN-y, IL-4, and IL-10 (42), and Helicobacter pylori induces gastric epithelial cells to produce IL-8 and MCP-1 (35, 55, 58, 61). Thus, the cytokine changes observed could be due to Campylobacter interaction with the intestinal epithelial cells in 120 the jejunum. Helicobacter responses, if present, would be expected to be similar based on previous studies (34). In the jejunum of the dually-infected pigs, GM-CSF expression was decreased 5 fold compared to the uninfected milk controls, suggesting that the T. suis down-regulated pro-inflammatory cytokine expression similar to the response seen in the proximal colon. Furthermore, clinical diarrhea was a prominent sign of disease throughout the experiments. During fecal sampling in both experiments, diarrhea was observed in pigs that received C. jejuni only, T. suis only or both. However, the dually infected pigs and the T. suis only-infected pigs had more frequent, more severe diarrhea than the other treatment groups, thereby implying that T. suis is mainly responsible for causing the observed diarrhea. Additionally, diarrhea was mostly observed in the long term challenge experiments where T richuris infection had progressed for 21 days. As has been reported frequently in literature reports on T richuris- induced disease, diarrhea usually begins around 21 days post-infection and is commonly called the “21 day scours” (24). In both experiments, some of the C. jejuni-infected pigs had mild diarrhea, which is commonly seen in campylobacterosis (10). In a germ fiee Campylobacter swine disease model experiment of similar design, Mansfield et a]. previously reported that some of the C. jejuni-infected pigs showed clinical signs of mild diarrhea less severe than that in dually-infected or T. suis only-infected pigs (50). Uninfected and E. coli DHS-a infected pigs showed no signs of diarrhea. The observation of severe diarrhea in pigs infected dually or with T. suis alone suggests that T. suis contributed significantly to diarrhea] disease. 121 Additionally, the histopathologic lesions observed in the tissues of pigs infected with T. suis alone or dually infected with T. suis and C. jejuni were consistent with what has been reported in the literature(]0, 50). The lesions in the tissues from the C. jejuni- infected pigs were mild, and there was no crypt distention or goblet cell hypertrophy. The lesions observed in the tissues of T. suis-infected pigs showed signs of mild colitis with crypt distention and increased thickness of all the layers of the colon. However, the lesions in the tissues of dually-infected pigs showed the greatest signs of disease with the greatest crypt distention and increased thickness of all layers of the colon, and greater infiltration of immune cells. Also, in the groups of pigs that were dually-infected or received T. suis alone, histological evaluation (H&E staining) confirmed the presence of T. suis in the tissues (Figure 2.4). Altogether, these results confirmed that all the pigs inoculated with T. suis became infected, and we were able to correlate the presence of the worm with cytokine changes seen in the colon. In summary, we found it necessary to use multiple diagnostic tests to detect and identify Campylobacter species or other epsilon proteobacteria in the feces and intestinal tissues of pigs, and despite this, definitive diagnosis was not possible. However, the data presented here suggest that throughout the intestine, T. suis appears to mediate most changes in pro- and anti- inflammatory cytokine expression during concurrent T. suis and C. jejuni infection, and it is possible that the immunomodulatory effects associated with these changes play a role in promoting a favorable host environment for secondary bacterial infection. One possible scenario is that T. suis interacts with the resident bacteria and down regulates the pro-inflammatory cytokine response which is generally elicited by bacterial infection, and the immunomodulatory effects of these pro- 122 inflammatory cytokines are diminished which could thereby facilitate bacterial invasion of intestinal cells and tissues causing cellular destruction and disease, as seen in human campylobacteriosis. Although our hypotheses cannot be unequivocally accepted, the data presented in this study provide some evidence to support this scenario. The occurrence of frequent, severe diarrhea in pigs infected with C. jejuni and T. suis or T. suis alone is a clear sign of disease and shows that T. suis contributes significantly to this syndrome. We were also able to detect pro- and anti- inflammatory cytokines in the feces of all the pigs regardless of treatment using ELISA, in agreement with data reported in the literature (9, 30, 57, 63, 66). However, measuring cytokine expression in feces by ELISA was found to be insufficient to directly address our hypothesis; unfortunately, this noninvasive method for investigating fecal cytokine expression during infection does not appear to be useful. In this study, the use of RT-PCR to measure cytokine expression in the tissues served as a more accurate and reliable method to address the overall hypothesis. Future studies will be conducted to describe the roles of these pro- and anti- inflammatory cytokines in this concurrent infection. 123 References Abner, S. R., D. E. Hill, J. R. Turner, E. D. Black, P. Bartlett, J. F. Urban, and L. S. Mansfield. 2002. Response of intestinal epithelial cells to Trichuris suis excretory-secretory products and the influence on Campylobacter jejuni invasion under in vitro conditions. J Parasit0188z738-745. Abner, S. R., G. Parthasarathy, D. E. Hill, and L. S. Mansfield. 2001. Trichuris suis: detection of antibacterial activity in excretory-secretory products from adults. Exp Parasitol 99:26-36. Abren, M. T., E. T. Arnold, L. S. Thomas, R. Gonsky, Y. Zhou, B. Hu, and M. Arditi. 2002. TLR4 and MD-2 expression is regulated by immune-mediated signals in human intestinal epithelial cells. J Biol Chem 277:20431-20437. Abren, M. T., P. Vora, E. Faure, L. S. Thomas, E. T. Arnold, and M. Arditi. 2001. Decreased expression of Toll-like receptor-4 and MD-2 correlates with intestinal epithelial cell protection against dysregulated proinflammatory gene expression in response to bacterial lipopolysaccharide. J Immunol 167:1609- 1616. Agnero-Rosenfeld, M. E., X. H. Yang, and l. Nachamkin. 1990. Infection of adult Syrian hamsters with flagellar variants of Campylobacter jejuni. Infect Immun 58:2214-2219. Allos, B. M. 2001. Campylobacter jejuni Infections: update on emerging issues and trends. Clin Infect Dis 32:1201-1206. Altekruse, S. F., N. J. Stern, P. 1. Fields, and D. L. Swerdlow. 1999. Campylobacter jejuni--an emerging foodbome pathogen. Emerg Infect Dis 5:28- 35. Ausubel, F. M., R. Brent, R. E. Kingston, D. D. Moore, J. G. Seidman, J. A. Smith, and K. Struhl. 1997. CTAB procedure for DNA extraction from Campylobacter jejuni. Current Protocols in Molecular Biology 1:2.4.1-2.4.2. Azim, T., R. C. Halder, M. S. Sarker, S. Ahmed, J. Hamadani, A. Chowdhury, F. Qadri, M. A. Salam, R. B. Sack, and M. J. Albert. 1995. Cytokines in the stools of children with complicated shigellosis. Clin Diagn Lab Immunol 2:492-495. 124 10. ll. 12. 13. 14. 15. l6. 17. 18. 19. 20. 21. Babakhani, F. K., G. A. Bradley, and L. A. Joens. 1993. Newborn piglet model for campylobacteriosis. Infect Immun 61:3466-3475. Bancroft, A. J., A. N. McKenzie, and R. K. Grencis. 1998. A critical role for lL-l3 in resistance to intestinal nematode infection. J Immunol 160:3453-3461. Baqar, S., A. L. Bourgeois, L. A. Applebee, A. S. Mourad, M. T. Kleinosky, Z. Mohran, and J. R. Murphy. 1996. Murine intranasal challenge model for the study of Campylobacter pathogenesis and immunity. Infect Immun 64:4933-4939. Baqar, S., N. D. Pacheco, and F. M. Rollwagen. 1993. Modulation of mucosal immunity against Campylobacter jejuni by orally administered cytokines. Antirnicrob Agents Chemother 37:2688-2692. Baqar, S., B. Rice, L. Lee, A. L. Bourgeois, A. N. E] Din, D. R. Tribble, G. P. Heresi, A. S. Mourad, and J. R. Murphy. 2001. Campylobacter jejuni enteritis. Clin Infect Dis 33:901-905. Bea], R. K., C. Powers, P. Wigley, P. A. Barrow, and A. L. Smith. 2004. Temporal dynamics of the cellular, humoral and cytokine responses in chickens during primary and secondary infection with Salmonella enterica serovar Typhimuriurn Avian Pathol 33:25-33. Bea], R. K., P. Wigley, C. Powers, P. A. Barrow, and A. L. Smith. 2006. Cross-reactive cellular and humoral immune responses to Salmonella enterica serovars Typhirnurium and Enteritidis are associated with protection to heterologous re-challenge. Vet Immunol Immunopathol 114:84-93. Bea], R. K., P. Wigley, C. Powers, S. D. Hulme, P. A. Barrow, and A. L. Smith. 2004. Age at primary infection with Salmonella enterica serovar Typhirnurium in the chicken influences persistence of infection and subsequent immunity to re-challenge. Vet Immunol Immunopathol 100: 151-164. Beer, R. J. 1973. Morphological descriptions of the egg and larval stages of Trichuris suis Schrank, 1788. Parasitology 67:263-278. Beer, R. J., L. F. Taffs, D. E. Jacobs, 1. J. Lean, and M. K. Curran. 1971. Evaluation of dichlorvos (V3 formulation) against larval and adult trichuris suis and observations on experimental infection in growing pigs. Vet Rec 88:436-441. Beery, J. T., M. B. Hugdah], and M. P. Doyle. 1988. Colonization of gastrointestinal tracts of chicks by Campylobacter jejuni. Appl Environ Microbiol 54:2365-2370. Bell, J. A., and D. D. Manning. 1990. A domestic ferret model of immunity to Campylobacter jejuni-induced enteric disease. Infect Irnrnun 58: 1848-1852. 125 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. Berg, D. J., N. Davidson, R. Kuhn, W. Muller, S. Menon, G. Holland, L. Thompson-Snipes, M. W. Leach, and D. Rennick. 1996. Enterocolitis and colon cancer in interleukin-lO-deficient mice are associated with aberrant cytokine production and CD4(+) THl-like responses. J Clin Invest 98: 1010-1020. Blaser, M. J ., and D. J. Duncan. 1984. Human serum antibody response to Campylobacter jejuni infection as measured in an enzyme-linked immunosorbent assay. Infect Immun 44:292-298. Bliss, D. 1991. Swine Parasites: A new look. at an old problem. Hoechst-Roussel Agri-Vet Co. Brady, M. T., S. M. O'Neill, J. P. Dalton, and K. H. Mills. 1999. Fasciola hepatica suppresses a protective Th1 response against Bordetella pertussis. Infect Immun 67:5372-5378. Cameron, S., K. Ried, A. Worsley, and D. Topping. 2004. Consumption of foods by young children with diagnosed campylobacter infection - a pilot case- control study. Public Health Nutr 7:85-89. Cawthraw, S. A., R. A. Feldman, A. R. Sayers, and D. G. Newell. 2002. Long- term antibody responses following human infection with Campylobacter jejuni. Clin Exp Immunol 130: 101-106. Chen, C. C., S. Louie, B. McCormick, W. A. Walker, and H. N. Shi. 2005. Concurrent infection with an intestinal helminth parasite impairs host resistance to enteric Citrobacter rodentium and enhances Citrobacter-induced colitis in mice. Infect Immun 73:5468-5481. Dawson, H. D., E. Beshah, S. N ishi, G. Solano-Aguilar, M. Morimoto, A. Zhao, K. B. Madden, T. K. Ledbetter, J. P. Dubey, T. Shea-Donohne, J. K. Lunney, and J. F. Urban, Jr. 2005. Localized multigene expression patterns support an evolving Thl/Th2-like paradigm in response to infections with Toxoplasma gondii and Ascaris suum. Infect Immun 73:1116-1128. Enocksson, A., J. Lundberg, E. Weitzberg, A. Norrby-Teglund, and B. Svenungsson. 2004. Rectal nitric oxide gas and stool cytokine levels during the course of infectious gastroenteritis. Clin Diagn Lab Immunol 11:250-254. Fermer, C., and E. O. Engval]. 1999. Specific PCR identification and differentiation of the thermophilic campylobacters, Campylobacter jejuni, C. coli, C. lari, and C. upsaliensis. J Clin Microbiol 37:3370-3373. 126 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. Finkelman, F. D., E. J. Pearce, J. F. Urban, Jr., and A. Sher. 1991. Regulation and biological function of helrninth-induced cytokine responses. Immunol Today 12:A62-66. Finkelman, F. D., T. Shea-Donohne, J. Goldhill, C. A. Sullivan, S. C. Morris, K. B. Madden, W. C. Gause, and J. F. Urban, Jr. 1997. Cytokine regulation of host defense against parasitic gastrointestinal nematodes: lessons from studies with rodent models. Annu Rev Immunol 15:505-533. Fox, J. G., P. Beck, C. A. Dangler, M. T. Whary, T. C. Wang, H. N. Shi, and C. Nagler-Anderson. 2000. Concurrent enteric helminth infection modulates inflammation and gastric immune responses and reduces helicobacter-induced gastric atrophy. Nat Med 6:536-542. Fntagami, S., T. Hiratsnka, A. Tatsuguchi, K. Suzuki, M. Kusnnoki, Y. Shinji, K. Shinoki, T. Iizumi, T. Akamatsn, H. Nishigaki, K. Wada, K. Miyake, K. Gudis, T. Tsnkni, and C. Sakamoto. 2003. Monocyte chemoattractant protein 1 (MCP-1) released from Helicobacter pylori stimulated gastric epithelial cells induces cyclooxygenase 2 expression and activation in T cells. Gut 52:1257-1264. Gately, M. K., L. M. Renzetti, J. Magram, A. S. Stern, L. Adorini, U. anler, and D. H. Presky. 1998. The interleukin-lZ/interleukin-lZ-reeeptor system: role in normal and pathologic immune responses. Annu Rev Immunol 16:495-521. Grencis, R. K. 2001. Cytokine regulation of resistance and susceptibility to intestinal nematode infection - from host to parasite. Vet Parasit01100:45-50. Hickey, T. E., S. Baqar, A. L. Bourgeois, C. P. Ewing, and P. Guerry. 1999. Campylobacter jejuni-stimulated secretion of interleukin-8 by INT407 cells. Infect Imrnun 67:88-93. Hickey, T. E., A. L. McVeigh, D. A. Scott, R. E. Michielutti, A. Bixby, S. A. Carroll, A. L. Bourgeois, and P. Guerry. 2000. Campylobacter jejuni cytolethal distending toxin mediates release of interleukin-8 fiom intestinal epithelial cells. Infect Immun 68:6535-6541. Hill, D. E., H. R. Gamble, M. L. Rhoads, R. H. Fetterer, and J. F. Urban, Jr. 1993. Trichuris suis: a zinc metalloprotease from culture fluids of adult parasites. Exp Parasitol 77:170-178. Hill, D. E., R. D. Romanowski, and J. F. Urban, Jr. 1997. A Trichuris specific diagnostic antigen from culture fluids of Trichuris suis adult worms. Vet Parasitol 68:91-102. 127 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. Hn, L., and T. E. Hickey. 2005. Campylobacter jejuni induces secretion of proinflammatory chemokines fiom human intestinal epithelial cells. Infect Immun 73:4437-4440. Humphrey, C. D., D. M. Montag, and F. E. Pittman. 1985. Experimental infection of hamsters with Campylobacter jejuni. J Infect Dis 151:485-493. Joseph F. Urban, J., Linda S. Mansfield, Terez Shea-Donahue, W. C.Gause, and F. D. Finkelman. 1999. Mechanisms of Intestinal Immunity to Nematode Parasites and the consequences to invasion by opportunistic bacteria. Seminar Monograph 11: Immunomodulation of the gastrointestinal mucosa. Kaldor, J., H. Pritchard, A. Serpell, and W. Metcalf. 1983. Serum antibodies in Campylobacter enteritis. J Clin Microbiol 18: 1-4. Lane, J. S., K. E. Todd, M. P. Lewis, B. Gloor, S. W. Ashley, H. A. Reber, D. W. McFadden, and C. F. Chandler. 1997. Interleukin-10 reduces the systemic inflammatory response in a murine model of intestinal ischemia/reperfirsion. Surgery 122:288-294. Linton, D., R. J. Owen, and J. Stanley. 1996. Rapid identification by PCR of the genus Campylobacter and of five Campylobacter species enteropathogenic for man and animals. Res Microbiol 147:707-718. Lugering, N., T. Kucharzik, M. Kraft, G. Winde, C. Sorg, R. Stoll, and W. Domschke. 1999. Interleukin (IL)-l3 and IL-4 are potent inhibitors of IL-8 secretion by human intestinal epithelial cells. Dig Dis Sci 44:649-655. Mansfield, L. S., and D. T. Gauthier. 2004. Lymphoglandular complexes are important colonic sites for immunoglobulin A induction against Campylobacter jejuni in a swine disease model. Comp Med 54:514-523. Mansfield, L. S., D. T. Gauthier, S. R. Abner, K. M. Jones, S. R. Wilder, and J. F. Urban. 2003. Enhancement of disease and pathology by synergy of Trichuris suis and Campylobacter jejuni in the colon of immunologically naive swine. Am J Trop Med Hyg 68:70-80. Mansfield, L. S., and J. F. Urban, Jr. 1996. The pathogenesis of necrotic proliferative colitis in swine is linked to whipworm induced suppression of mucosal immunity to resident bacteria. Vet Immunol Immunopathol 50:1-17. McKenzie, G. J., A. Bancroft, R. K. Grencis, and A. N. McKenzie. 1998. A distinct role for interleukin-13 in Th2-cell-mediated immune responses. Curr Biol 8:339-342. 128 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. Mellits, K. H., J. Mullen, M. Wand, G. Armbruster, A. Patel, P. L. Connerton, M. Skelly, and I. F. Connerton. 2002. Activation of the transcription factor NF -kappaB by Campylobacter jejuni. Microbiology 148:2753-2763. Muchamnel, T., S. Menon, P. Pisacane, M. C. Howard, and D. A. Cockayne. 1997. IL-13 protects mice from lipopolysaccharide-induced lethal endotoxemia: correlation with down-modulation of TNF-alpha, IFN-gamma, and IL-12 production. J Immunol 158:2898-2903. Nakano, Y., T. Kasahara, N. Mukaida, Y. C. Ko, M. Nakano, and K. Matsnshima. 1994. Protection against lethal bacterial infection in mice by monocyte-chemotactic and -activating factor. Infect Immun 62:377-383. Neish, A. S., A. T. Gewirtz, H. Zeng, A. N. Young, M. E. Robert, V. Karmali, A. S. Rao, and J. L. Madara. 2000. Prokaryotic regulation of epithelial responses by inhibition of IkappaB-alpha ubiquitination. Science 289: 1560-1563. Nicholls, S., S. Stephens, C. P. Braegger, J. A. Walker-Smith, and T. T. MacDonald. 1993. Cytokines in stools of children with inflammatory bowel disease or infective diarrhoea. J Clin Pathol 46:757-760. Nozawa, Y., K. Nishihara, R. M. Peek, M. Nakano, T. Uji, H. Ajioka, N. Matsuura, and H. Miyake. 2002. Identification of a signaling cascade for interleukin-8 production by Helicobacter pylori in human gastric epithelial cells. Biochem Pharmacol 64:21-30. O'Neil], S. M., M. T. Brady, J. J. Callahan, G. Mulcahy, P. Joyce, K. H. Mills, and J. P. Dalton. 2000. Fasciola hepatica infection downregulates Th1 responses in mice. Parasite Immunol 22:147-155. Parthasarathy, G., and L. S. Mansfield. 2005. Trichuris suis excretory secretory products (ESP) elicit interleukin-6 (IL-6) and IL-10 secretion from intestinal epithelial cells (IPEC-1). Vet Parasitol 131:317-324. Peek, R. M., Jr. 2001. IV. Helicobacter pylori strain-specific activation of signal transduction cascades related to gastric inflammation. Am J Physiol Gastrointest Liver Physiol 280:G525-530. Prescott, J. F., I. K. Barker, K. I. Manninen, and O. P. Miniats. 1981. Campylobacter jejuni colitis in gnotobiotic dogs. Can J Comp Med 45:377-383. Raqib, R., B. Wretlind, J. Andersson, and A. A. Lindberg. 1995. Cytokine secretion in acute shigellosis is correlated to disease activity and directed more to stool than to plasma. J Infect Dis 171:376-384. 129 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. Riley, L. K., C. L. Franklin, R. R. Hook, J r., and C. Besch—Williford. 1996. Identification of murine helicobacters by PCR and restriction enzyme analyses. J Clin Microbiol 34:942-946. Rutter, J. M., and R. J. Beer. 1975. Synergism between Trichuris suis and the microbial flora of the large intestine causing dysentery in pigs. Infect Immun 1 1:395-404. Saiki, T., K. Mitsuyama, A. Toyonaga, H. Ishida, and K. Tanikawa. 1998. Detection of pro- and anti-inflammatory cytokines in stools of patients with inflammatory bowel disease. Scand J Gastroenterol 33:616-622. Sansonetti, P. J., J. Aronde], M. Huerre, A. Harada, and K. Matsnshima. 1999. Interleukin-8 controls bacterial transepithelial translocation at the cost of epithelial destruction in experimental shigellosis. Infect Immun 67: 1471-1480. Schopf, L. R., K. F. Hoffmann, A. W. Cheever, J. F. Urban, Jr., and T. A. Wynn. 2002. IL-10 is critical for host resistance and survival during gastrointestinal helminth infection. J Immunol 168:2383-2392. Summers, R. W., D. E. Elliott, J. F. Urban, Jr., R. Thompson, and J. V. Weinstock. 2005. Trichuris suis therapy in Crohn's disease. Gut 54:87-90. Summers, R. W., D. E. Elliott, J. F. Urban, Jr., R. A. Thompson, and J. V. Weinstock. 2005. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology 128:825-832. Sutterwala, F. S., and D. M. Mosser. 1999. The taming of IL-12: suppressing the production of proinflammatory cytokines. J Leukoc Biol 65:543-551. Swartz, M. N. 2002. Human diseases caused by foodbome pathogens of animal origin. Clin Infect Dis 34 Suppl 3:811 l-122. Turner, J., H. Faulkner, J. Kamgno, K. Else, M. Boussinesq, and J. E. Bradley. 2002. A comparison of cellular and humoral immune responses to trichuroid derived antigens in human trichuriasis. Parasite Immun0124:83-93. Urban, J. F., Jr., K. B. Madden, A. Svetic, A. Cheever, P. P. Trotta, W. C. Gause, I. M. Katona, and F. D. Finkelman. 1992. The importance of Th2 cytokines in protective immunity to nematodes. Immunol Rev 127:205-220. Wilson, D. L., S. R. Abner, T. C. Newman, L. S. Mansfield, and J. E. Linz. 2000. Identification of ciprofloxacin-resistant Campylobacter jejuni by use of a fluorogenic PCR assay. J Clin Microbiol 38:3971-3978. 130 76. 77. 78. 79. Withanage, G. S., P. Wigley, P. Kaiser, P. Mastroeni, H. Brooks, C. Powers, R. Bea], P. Barrow, D. Maskell, and I. McConnell. 2005. Cytokine and chemokine responses associated with clearance of a primary Salmonella enterica serovar Typhimurium infection in the chicken and in protective immunity to rechallenge. Infect Imrnun 73:5173-5182. Xavier, R. J., and D. K. Podolsky. 2000. Microbiology. How to get along-- friendly microbes in a hostile world. Science 289: 1483-1484. Young, C. R., R. Harvey, R. Anderson, D. Nisbet, and L. H. Stanker. 2000. Enteric colonisation following natural exposure to Campylobacter in pigs. Res Vet Sci 68:75-78. Ziprin, R. L., C. R. Young, L. H. Stanker, M. E. Hume, and M. E. Konkel. 1999. The absence of cecal colonization of chicks by a mutant of Campylobacter jejuni not expressing bacterial fibronectin—binding protein. Avian Dis 43:586-589. 13] Table 2.1. Experimental treatment groups for the short (2 days) and long (23 days) term challenge experiments. Treatment Treatment Groups Group 1 C. jejuni (2 x 108 cfu) Group 2 C. jejuni (2 x 108 cfil)and and T. suis (2500 embryonated eggs) Group 3 T. suis (2500 embryonated eggs) Group 4 E. coli DHS-a (2 x 108 cfir) Group 5 Uninfected milk control (2 ml) 132 Table 2.2. Inoculation timeline for short term challenge experiments (2 days): 4-week old weaned, conventionally reared pigs were divided into 5 treatment groups with a total of 10 pigs per group. The inoculations were administered via oral gavage in 2.0 ml of sterile milk. The pigs were given 2 x 108 cfu C. jejuni, 2500 T. suis embryonated eggs, both, 2 X 108 cfir E. coli DHS-a, or milk. Day 0 Day 1 Day 2 l. Rectal swabbing Rectal swabbing 1. Rectal swabbing and fecal sampling and fecal sampling and fecal sampling of all animals of all animals 2. Inoculation 2. All pigs were protocol: euthanized and necropsied. Group 1: C. jejuni Group 2: C. jejuni and T. suis Group 3: T. suis Group 4: E. coli DHS-a Group 5: Milk 133 Table 2.3. Inoculation timeline for long term challenge experiments (23 days): 4-week old weaned, conventionally reared pigs were divided into 5 treatment groups with a total of 1] pigs per group. The inoculations were administered via oral gavage in 2.0 ml of sterile milk. The pigs were given 2 x 108 cfu C. jejuni, 2500 T. suis embryonated eggs, both, 2 x 108 cfu E. coli DHS-a, or milk. Day 0 Day 21 Day 22 Day 23 l. Rectal swabbing 1. Rectal swabbing Rectal 1. Rectal swabbing and and fecal sampling swabbing and fecal sampling and fecal fecal sampling sampling of all animals of all animals 2. Inoculation 2. Inoculation 2. All pigs were protocol: protocol: euthanized and necropsied. Group 1: Milk Group 1: C. jejuni Tissue samples were taken from Group 2: T. suis Group 2: C. jejuni all pigs. Group 3: T. suis Group 3: Milk Group 4: Milk Group 4: E. coli DHS-a Group 5: Milk Group 5: Milk I34 Table 2.4. Summary of Campylobacter detection and identification. Campylobacters were detected and identified fi'om pigs by fecal culture, PCR for the C. jejuni QRDR on intestinal lumen contents obtained from each intestinal region and tissue DNA, and RFLP analysis of the thermophilic CampJvlobacter 23S rRN A gene using tissue DNA. Samples were taken from the pigs in the 2n repetition of the long term challenge experiments only (Jones, dissertation, 2005, reprinted with permission). Campylobacter 23S rRNA RFLP QRDR (tissue DNA) Unidentified Fecal Tissue Campylobacter Infection Culture DNA DNA C. jejuni C. coli sp. Control 0/6 0/6 0/6 0/6 5/6 2/6 Jejunum C. jejuni 0/6 0/6 0/6 0/6 2/6 5/6 T. suis 0/6 0/6 0/6 0/6 3/6 2/6 Dual 0/6 0/6 0/6 1/6 4/6 1/6 Control 0/6 0/6 0/6 1/6 4/6 5/6 Proximal C. jejuni 0/6 0/6 0/6 1/6 2/6 4/6 colon T. suis 0/6 0/6 0/6 1/6 4/6 1/6 Dual 0/6 0/6 0/6 4/6 4/6 2/6 Control 0/6 0/6 0/6 0/6 0/6 6/6 Distal C. jqiuni 0/6 0/6 0/6 0/6 0/6 6/6 colon T. suis 0/6 0/6 1/6 0/6 2/6 3/6 Dual 0/6 0/6 0/6 0/6 2/6 6/6 135 Figure 2.1. RFLP analysis of the Campylobacter 23S rRNA gene. Panel A: AluI digest of proximal colon samples fi'om infected pigs . Lane I: 100 bp DNA ladder; Lane 2: C. jejuni; Lane 3: C. coli; Lanes 4 through 15: DNA from infected pigs. Panel B: Tsp509l digest of proximal colon samples from infected pigs. Lane 1: 100 bp DNA ladder; Lane 2: C. jejuni; Lane 3: C. coli; Lane 4: C. hyoilei; Lanes 5 through 16: DNA from infected pigs. Samples were taken from the pigs in the 2"d repetition of the long term challenge experiments only (Jones, dissertation, 2005, reprinted with permission). 136 1.4 1.2 o 1 In * 0.8 :5 0.6 0 0.4 : 0.2 l 0 I Milk C. jejuni C. jejuni T. suis E. coli control and T. suis Treatment groups B 0.8 0.7 0.6 O in 0.5 Q‘ . 0.4 Q 0 0.3 0.2 0.1 0 Milk C. jejuni C. jejuni T. suis E. coli control and T. suis Treatment groups Figures 2. 2 A and B. (A) Short term challenge experiment and (B) long term challenge experiment ELISA results indicating levels of plasma IgG antibodies detectable against C. jejuni protein antigen. IgG levels were measured in the plasma of all pigs fi'om each treatment group. The data represent the mean i SEM. I37 N l l l l i l l l l l‘ _s '0) l iv .0 co O. D. 450 in fie were Emmi raini- 1‘! --- ”ha/g, F) ---- 6) --- new 6) ---- O o o o N b a) _- Hm: ease reassess se.§e s E§°Q§ g§3e§o§agg tags» 5 ~Q.@8\_ 0 SGgQOE (3737803 0 'Q 0 <50 94’ s 33 as (50 we 8 9° 3.3 $19 -' Q 3.3 as it“ s a 3:6 .753: 00 §~Q Figure 2.3. ELISA results indicating levels of plasma IgG antibodies detectable against protein antigen of various Campylobacter species, Arcobacter butzleri, and Helicobacter hepaticus, suggesting IgG cross-reactivity. IgG levels were measured in the plasma of 8 C. jejuni-infected pigs randomly selected from the short- and long-term challenge experiments (+), 6 uninfected pigs randomly selected fiom both challenge experiments that received milk only(-), and 2 newborn piglets < 6 hours old from the MSU swine farm (control). The data represent the mean :1: SEM. “Images in this dissertation are presented in color.” 138 Table 2.5. Short term challenge experiment 16S RNA PCR analysis results for rapid identification of Campylobacter and Helicobacter DNA in pig feces. Helicobacter ' # Pre-infection Post-infection Pre-infection Post-infection 1 2 3 4 5 6 7 8 9 10 ll 12 l3 14 15 l6 l7 18 19 20 21 22 23 24 25 26 27 28 29 b) O a. + tested positive b. - tested negative c. x represents missing sample 139 Table 2.6. Long term challenge experiment I6S RNA PCR analysis results for rapid identification of Campylobacter and Helicobacter DNA in pig feces. Helicobacter ' # Pre-infection Post-infection Pre-infection Post-infection 6 - 7 8 13 14 15 l6 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 a. + tested positive b. - tested negative c. x represents missing sample 140 Figure 2.4. H&E stained sections from the colons of pigs were examined for T. suis larvae. Panel A: Control; B: C. jejuni only; C: T. suis only; D: Dual infection, 10X magnification. Larvae (arrows) were detected in 5 of 6 pigs from each of the T. suis only and dual infection groups. Samples were taken from the pigs in the 2nd repetition of the long term challenge experiments only (Jones, dissertation, 2005, reprinted with permission). “Images in this dissertation are presented in color.” 141 Figure 2.5. H&E section from a C. jejuni infected pig (panel A) showing sloughed epithelial cells in the crypts (black arrow), and a T. suis infected pig (panel B) showing the mixed inflammatory infiltrate in the lamina propria associated with the larvae (green arrow). Inflammatory cells included eosinophils (black arrow), plasma cells (blue arrow), lymphocytes (red arrow), and neutrophils (yellow arrow) (40X magnification). Samples were taken from the pigs in the 2nd repetition of the long term challenge experiments only (Jones, dissertation, 2005, reprinted with permission). “Images in this dissertation are presented in color.” 142 Figure 2.6. H&E sections of LGCs fi‘om infected pigs. Crypt distension was present in the pigs that received C. jejuni only (panel A) and in the dually-infected pigs (panel C), but crypts were not distended in the pigs that received T. suis only (Panel B). (10X magnification). Samples were taken from the pigs in the 2nd repetition of the long term challenge experiments only (Jones, dissertation, 2005, modified and reprinted with permission). “Images in this dissertation are presented in color.” 143 Je'unum Proximal colon Distal Colon was 1:1 .3 1:1 :3 1: 1: m1:11::11:1::n1: MC.-. m::11:1::1:c: m-a m::11:1::11:11: mm EIEZIIIIEZIDE acnmmju: ms m::1r:11::11:1: m 13::11:H::1:i: W m1::n1::1u1: 1:1 11:1 BID 1: 1: mi 1:11::1:ii:m 1:1 i... a1:1:11:11::11:11:1 :11::11:::11:1m H... DEJDEEDE n... I :JJLIichLi: Figure 2.7. Expression of mRN A from a panel of cytokines was measured by Real Time PCR. The data represent the average fold change in expression fiom infected animals when compared to the milk control animals. Letters indicate statistically significant changes: A: p5 0.05; B: p: 0.01. Tissue samples were taken fiom the pigs in the 2nd repetition of the long term challenge experiments only (Jones, dissertation, 2005, reprinted with permission). “Images in this dissertation are presented in color.” 144 Tables 2.7. Short term challenge: clinical assessment of diarrhea in weaned, conventionally-reared swine infected with T. suis and/or C. jejuni. Experiment Treatment # of pigs with diarrhea group Day 0 Day 1 Day 2 Short (2 days)“ Group 1 2nd repetition c. jejuni 0/6 2/6 0/6 of experiment only Group 2 C. jejuni and T. suis 0/6 1/6 4/6 Group 3 T. suis 06 0/6 0/6 Group 4 E. coli DHS-a 06 0/6 0/6 Group 5 Uninfected 06 0/6 0/6 All animals were assessed for diarrhea during fecal sampling on days 0, 1, and 2 of the short term challenge experiment. a The four pigs from each treatment group in the first repetition of the short term challenge experiment were not assessed for diarrhea. 145 Tables 2.8. Long term challenge: clinical assessment of diarrhea in weaned, conventionally-reared swine infected with T. suis and/or C. jejuni. Experiment Treatment # of pigs with diarrhea group Day 0 Day 21 Day 22 Day 23 Long (23 days) Group 1 Both C. jejuni 0/11 1/11 2/11 3/11 repetitions combined Group 2 C. jejuni andeuis 0/11 6/11 6/11 7/11 Group 3 Tsuis 0/11 5/11 5/11 10/11 Group 4 E. coIiDHS-a 0/11 0/11 0/11 0/11 Group 5 Uninfected 0/11 0/11 0/ 11 0/11 All animals were assessed for diarrhea during fecal sampling on days 0, 21, 22, and 23 of the long term challenge experiments. 146 Table 2.9. Fecal cytokine recovery rate. Cytokine Fecal cytokine recovery rate (%) lL-8 10 TNF-a 20 IL- 1 -[3 10 IL-4 10 IL-6 20 IL- 1 0 10 147 4500‘ 1500 4000 H o 3500- 3 IDay 0 I 3000- H IDay 1 E 2500< . m EJDa 2 Q 2000 y a vi 0 I .3 H 1000 Milk C. jejuni C. jejuni T. suis E. coli Control and T. suis DHS-a Treatment groups Figure 2.8. Changes in IL-8 levels were quantified in the feces of weaned piglets fi'om the short term challenge experiments. The pigs were inoculated via oral gavage with various treatments. Fecal samples were collected on days 0, 1, and 2 for further processing to be used in ELISA. Values shown are means of two identical short term (2 days) challenge experiments with a total of 10 pigs in each group. The data represent the mean 1 standard error of the mean (SEM); P=0.56. 148 H o 3 a lDay 0 E \ IDay 1 tn 9. ElDay 2 1: vi a I E Milk C. jejuni C. jejuni T. suis E. coli Control and T. suis DHs-a Treatment groups Figure 2.9. Changes in TNF-u levels were quantified in the feces of weaned piglets from the short term challenge experiments. The pigs were inoculated via oral gavage with various treatments. Fecal samples were collected on days 0, 1, and 2 for firrther processing to be used in ELISA. Values shown are means of two identical short term (2 days) challenge experiments with a total of 10 pigs in each group. The data represent the mean i SEM; P=0.15. 149 8000‘ 7000 6000 lDa 0 5000 y lDa 1 4000 - Y DDay 2 3000 2000 IL-l-p in pg/ml stool 1000 Milk C. jejuni C. jejuni T. suis E. coli Control and T. suis DHS-a Treatment groups Figure 2.10. Changes in IL-l-B levels were quantified in the feces of weaned piglets from the short term challenge experiments. The pigs were inoculated via oral gavage with various treatments. Fecal samples were collected on days 0, 1, and 2 for further processing to be used in ELISA. Values shown are means of two identical short term (2 days) challenge experiments with a total of 10 pigs in each group. The data represent the mean i SEM; P=0.27. 150 . IDay O IDay 1 DDay 2 IL—4 in pg/ml stool Milk C. jejuni C. jejuni T. suis E. coli Control only and T. suis DH5-alpha Treatment groups Figure 2.11. Changes in IL-4 levels were quantified in the feces of weaned piglets fi'om the short term challenge experiments. The pigs were inoculated via oral gavage with various treatments. Fecal samples were collected on days 0, l, and 2 for firrther processing to be used in ELISA. Values shown are means of two identical short term (2 days) challenge experiments with a total of 10 pigs in each group. The data represent the mean i SEM; P=0.26. 151 4500 1000 500 '6' 4000 , O n 3500» m a 3000 IDay 0 F1 2500 IDa 1 n- 2000 y 3 1500 DDay 2 \D I q H Milk C. jejuni C. jejuni T. suis E. coli Control only and T. suis DH5—alpha Treatment groups Figure 2.12. Changes in IL-6 levels were quantified in the feces of weaned piglets fi'om the short term challenge experiments. The pigs were inoculated via oral gavage with various treatments. Fecal samples were collected on days 0, 1, and 2 for firrther processing to be used in ELISA. Values shown are means of two identical short term (2 days) challenge experiments with a total of 10 pigs in each group. The data represent the mean i SEM; P=0.45. 152 IDay 0 IDay 1 DDay 2 IL-10 in pg/ml stool Milk C. jejuni C. jejuni T. suis E. coli Control only and T. suis DES—alpha Treatment groups Figure 2.13. Changes in IL-10 levels were quantified in the feces of weaned piglets from the short term challenge experiments. The pigs were inoculated via oral gavage with various treatments. Fecal samples were collected on days 0, l, and 2 for further processing to be used in ELISA. Values shown are means of two identical short term (2 days) challenge experiments with a total of 10 pigs in each group. The data represent the mean i SEM; P=0.79. 153 3500.00 ‘ 1000. H 8 3000. ii 2500. IDay 0 a IDay 21 E: 2000. DDay 22 fl 1500' ElDay 23 ‘I'. (D I .:1 H Milk C. jejuni C. jejuni T. suis E. coli Control and T. suis DHs-a Treatment groups Figure 2.14. Changes in fecal IL-8 levels were measured using ELISA at various time points over a 23-day period. Fecal samples were collected from infected, weaned piglets. The values shown are means of two identical long term (23 days) challenge experiments with a total of 1 1 pigs in each group (n=8 in the E. coli DHS-a group). The data represent the mean 1 SEM; P 2 0.05. 154 2500- , 7 . _- __ , ”a 2000 ‘IDay 0 ’IDay 21 _ DDay22 »ElDay23 1500 ‘ 1000 " TNF-a in pg/ml stool ,3". Milk only C. jejuni C. jejuni T.suis E.col.i DHS—a and T.suis Treatment groups Figure 2.15. Changes in fecal TNF-a levels were measured using ELISA at various time points over a 23-day period. Fecal samples were collected fiom infected, weaned piglets. The values shown are means of two identical long term (23 days) challenge experiments with a total of 11 pigs in each group (n=8 in the E. coli DHS-a group). The data represent the mean i SEM; P 2 0.05. 155 § U 50004 if n l a 4000 4 \ l a . 3000 n l a a, 2000 1 I l H ,_'-. 1000 *7 7 H 0 r Milk Control C. jejuni C. jejuni and T. suis E. coli DES—a T. suis Treamnt groups Figure 2.16. Changes in fecal IL-l-B levels were measured using ELISA at various time points over a 23-day period. Fecal samples were collected from infected, weaned piglets. The values shown are means of two identical long term (23 days) challenge experiments with a total of 11 pigs in each group (n=8 in the E. coli DH5-a group). The data represent the mean i SEM. * Represents the significant difference of the values for the T. suis-infected pigs on day 23 in comparison to the values for the milk only control pigs; P S 0.05. 156 450 H g 400 f; 350 IDay o E 300 IDay 21 B 250 ElDay 22 °' 20° IDay 23 g 150 ‘1 100 l A 50 H O . Milk C. jejuni C. jejuni T. suis E. coli Control only and T. suis HHS-alpha Treatment groups Figure 2.17. Changes in fecal IL-4 levels were measured using ELISA at various time points over a 23-day period. Fecal samples were collected from infected, weaned piglets. The values shown are means of two identical long term (23 days) challenge experiments with a total of 1 1 pigs in each group (n=8 in the E. coli DHS-a group). The data represent the mean i SEM; P 2 0.05. 157 IDay 0 IDay 21 EIDay 22 IDay 23 IL-6 in pg/ml stool Milk C. jejuni C. jejuni T. suis E. coli Control only and T. suis DES-alpha Treatment groups Figure 2.18. Changes in fecal IL-6 levels were measured using ELISA at various time points over a 23-day period. Fecal samples were collected from infected, weaned piglets. The values shown are means of two identical long term (23 days) challenge experiments with a total of 11 pigs in each group (n=8 in the E. coli DHS-a group). The data represent the mean 1 SEM; P 2 0.05. 158 IDay O IDay 21 DDay 22 away 23 IL-10 in pg/ml stool Milk C. jejuni C. jejuni T. suis E. coli Control only and T. suis DES—alpha Treatment groups Figure 2.19. Changes in fecal IL-lO levels were measured using ELISA at various time points over a 23-day period. Fecal samples were collected from infected, weaned piglets. The values shown are means of two identical long term (23 days) challenge experiments with a total of 11 pigs in each group (n=8 in the E. coli DHS-a group). The data represent the mean :t SEM; P 2 0.05. 159 Chapter 3: “Evaluation of proinflammatory cytokine production in intestinal pig epithelial cells infected with Campylobacterjejzmi” Abstract C. jejuni is one of the leading causes of gastroenteritis throughout the world. Enterocytes serve as the first line of defense during bacterial infection, and C. jejuni— enterocyte interaction may play a significant role in the production of an acute inflammatory response. It has been shown in vitro that C. jejuni interacts with intestinal epithelial cells and stimulates the production of proinflammatory cytokines and chemokines. Therefore, we hypothesized that cultured undifferentiated, crypt-like intestinal pig epithelial cells (IPEC-l cells) secrete TNF-a, IL-l-B, and IL-8 following C. jejuni infection. To test this hypothesis, we infected cultured IPEC-1 cells with C. jejuni and measured IL-8, TNF-a, and lL-l-B production. Supernatants were collected from IPEC-l cells at O, 8, 24, and 48 hours post-C. jejuni infection, and cytokine expression was measured using an enzyme-linked immunoabsorbant assay (ELISA). C. jejuni infection of IPEC-1 cells significantly induced TNF-a and IL-l-B production. In addition, we found that IPEC-1 cells constitutively secrete IL-8, and IL-8 production was not induced by C. jejuni infection. Our data suggest that induced TNF-a and IL-l-B production and constitutively secreted IL-8 may play a role in an innate inflammatory response that generates or modulates immune responses during C. jejuni infection in vivo. We also pretreated undifferentiated IPEC-l monolayers with recombinant swine lL-8, TNF-a, or IL-l-B to determine the direct effects of these cytokines on C. jejuni adherence, invasion, and the transepithelial electrical resistance of IPEC-l cells. Our 160 findings suggested that neither lL-8 nor lL-l-B pretreatments had a significant effect on C. jejuni invasion, adherence, or the transepithelial electrical resistance of IPEC-1 cells. In addition, we found that TNF-a did not significantly affect C. jejuni adherence to IPEC- ] cells or the transepithelial electrical resistance of IPEC-l cells. Taken together, the data presented here suggest that the induced production of TNF-a and IL-l-B and the constitutive production of IL-8 from intestinal epithelial cells are indicative of an acute innate host immune response that may be responsible in part for eliminating these bacteria from the host in vivo. 16] Introduction Camp}'lobacterjejzmi is a Gram negative, microaerophilic, rod-shaped bacterium which is known throughout the world as one of the leading causes of acute gastroenteritis (34). Documented clinical symptoms related to C. jejuni infection are abdominal cramps, diarrhea, fever, general malaise, backache, headache, and arthralgias (5, 6, ll, 12, 34). In human clinical infection studies, flesh blood, mucus, and polymorphonuclear leukocytes are found in diarrheic stool samples (34). In most cases, illness is self-limiting, and the duration of the ilhiess is usually 1 week or less (34). In addition to the immediate effects of acute Campylobacter infection, there are long-term sequelae associated with campylobacteriosis such as reactive arthritis (ReA), Reiter’s syndrome, Fisher syndrome, and Guillain-Barré Syndrome (41). These post-infectious sequelae result in serious health problems and have a significant economic impact (19, 28, 35, 41, 49, 54). The causative mechanisms of disease by Campylobacter infections are not well understood. However, it is believed that host immune responses, such as cytokine production, may play an important role in the pathogenesis of the disease. Presently, there is limited knowledge about immune responses elicited during C. jejuni infection. In vitro, human intestinal epithelial cells (INT 407), monocytic cells, avian primary chick kidney cells, and an avian macrophage cell line secrete pro- inflammatory cytokines (TNF-a, IFN-y, IL-I-B and IL-6) and chemokines (IL-8, MCP-l, and MIP-l) following infection with viable C. jejuni (4, 25, 27, 31, 43). In these systems, C. jejuni invade and are found within vacuoles. C. jejuni also invades intestinal pig I62 epithelial cells (IPEC-1) with a differentiated phenotype and stimulates IL-6, IL-10, and IL-18 production (Parthasarathy, dissertation, 2004 and Jones, dissertation, 2005). In a Campylobacter swine disease model, weaned piglets infected orally with C. jejuni show MCP-1, TNF-a, IL-12p40, IFNy, IL-4, and IL-10 mRNA expression in the jejunum at 48 hours post-infection, and C. jejuni infection causes lesions in the jejunum and mild crypt distension with sloughing epithelial cells and mucus production. (Jones, dissertation, 2005). These reported findings suggest that cytokines may play a significant role in C. jejuni-induced intestinal inflammation and disease pathology. Further evaluation of C. jejuni-host cell interaction and cytokine expression is necessary to elucidate the immune response elicited during infection. There has been limited data reported which suggest that C. jejuni interaction with or invasion of epithelial cells play a role in stimulating cytokine production (4, 9, 25-27, 43). C. jejuni invasion of intestinal epithelial cells has been observed in vitro and in vivo and has been suggested as playing an important role in the pathogenesis of campylobacteriosis (56, 59). However, to date, a direct correlation between C. jejuni invasion of intestinal epithelial cells and subsequent cytokine production resulting in disease and pathology has not been established. These types of studies cannot be done in human patients. However, in C. jejuni-infected pigs, the bacterial cells have been found, via irnmunohistochemical staining, to be localized in the crypts of the distal colon, the lymphoglandular complexes (LGCs, and within the follicle-associated epithelium of the LGCs (10, 39)(Jones, dissertation, 2005). Also, haemotoxylin and eosin (H&E) staining of intestinal tissue samples from these C. jejuni-infected pigs showed mild crypt distension with sloughed epithelial cells and mucus. Likewise, in these experiments, 163 proinflammatory cytokine production was associated with C. jejuni infection of tissues (Jones, dissertation, 2005). Because it has been shown that C. jejuni-host-cell interaction can trigger an immune response (such as stimulating intestinal epithelial cells to produce pro-inflammatory cytokines and chemokines) and these events are associated with disease and pathology, we hypothesize that pro-inflammatory cytokine production of IECs may also affect epithelial cell integrity and play a role in affecting C. jejuni adherence, invasion, and/or the transepithelial resistance of intestinal epithelial cells. Others have shown that specific cytokines (e. g. TNF-a, IFN-y, and IL-l-B) can alter intestinal epithelial cell function or integrity of the epithelial barrier, thereby facilitating bacterial-enterocyte interactions and causing serious pathological consequences (24, 48, 51). We designed this study to better understand the role played by undifferentiated, crypt-like intestinal epithelial cells in a natural host in immunity and immunomodulation following C. jejuni infection. We used undifferentiated, crypt-like IPEC-1 cells derived from an unsuckled newborn piglet to evaluate pro-inflammatory cytokine production following C. jejuni infection and some of the possible roles these cytokines play in C. jejuni infection. Because C. jejuni-enterocyte interaction has been shown to trigger an acute immune response in vitro and in vivo, we hypothesized that undifferentiated, crypt- like IPEC-I cells secrete TNF-a, IL-l-B, and IL-8 in response to C. jejuni infection. Our findings show that C. jejuni induces these swine intestinal epithelial cells to produce TNF-o. and IL-l-B and that IL-8 is not induced by C. jejuni exposure but is constitutively secreted by these cells. These findings suggest that this early, innate response may participate in stimulating adaptive ThI-type adaptive immune responses which play an 164 important role in bacterial elimination in other hosts. In addition, to elucidate the roles of pro-inflammatory cytokines in C. jejuni-intestinal epithelial cell interaction, we also evaluated how the treatment of IPEC-l cells with recombinant swine TNF-a, IL-l-B, or IL-8 directly affects C. jejuni adherence, invasion and/or the transepithelial electrical resistance. We found that these pro-inflammatory cytokines do not influence C. jejuni adherence and/or invasion of intestinal epithelial cells, nor do they stimulate changes in the monolayer integrity to facilitate C. jejuni infection. 165 Materials and Methods Bacteria and growth conditions C. jejuni strain ATCC 33292 was grown at 37°C, 5% C02 on Bolton agar plates. This strain was originally isolated from a human with enteritis and was rederived by a single passage in a gnotobiotic pig. Escherichia coli DH5-0t was a noninvasive negative control for the invasion assay, and E. coli 0157:H43 was a positive control for the adherence assay. E. coli 0157:H43 (DEC7A) was kindly provided by Dr. Thomas Whittman of Michigan State University. The E. coli strains were grown on Bolton agar at 37°C. All bacterial culture media were obtained fiom Oxoid (Oxoid Inc, Basingstoke, Hampshire, England) unless otherwise stated. All bacteria were taken from frozen stocks, streaked for isolation onto agar and incubated for 24-48 hours. For inoculation, the C. jejuni and E. coli isolated colonies were subcultured for 20 hours for confluent growth, harvested with sterile swabs, and suspended in RPMI 1640 medium (lnvitrogen, lnvitrogen, Frederick, MD) without phenol red, fetal bovine serum (lnvitrogen, Frederick, MD) or insulin-transferrin selenium (lnvitrogen, Frederick, MD). The optical density (OD560) was adjusted to 0.1 to achieve 5 X 108 colony forming units per milliliter. Dilutions of the bacteria were made in RPMI 1640 medium (lnvitrogen, Frederick, MD) without phenol red, fetal bovine serum (F BS; lnvitrogen, Frederick, MD) and insulin- transferrin selenium (ITS; lnvitrogen, Frederick, MD) or Bolton broth (C. jejuni and E. coli strains) as required to achieve a particular bacterial dose. 166 Cultured epithelial cells IPEC-1 (intestinal pig epithelial cells) is a non-immortalized cell line originally derived from the small intestine of an unsuckled neonatal piglet. In this study, these cells were used in an undifferentiated state to represent the rapidly dividing basilar intestinal epithelial crypt cells. Two-to-three days after seeding, they form confluent monolayers but do not have tight junctions or distinct apical or baso-lateral surfaces, which is characteristic of the morphology of intestinal epithelial crypt cells. All culture media and growth supplements were obtained from Invitrogen (Frederick, MD) unless otherwise stated. Growth flasks, flat bottom polystyrene plates, and transwells were obtained from Coming (Costar, Corning, NY). IPEC-1 cells were routinely maintained in RPMI 1640 medium containing 5% fetal bovine serum (FBS) and supplemented with 1% insulin-transferrin selenium (ITS) and grown to a confluent monolayer in T75 flasks at 37°C in a humidified atmosphere with 5% C02. For infection studies, the cells were gently washed with versene and released from the flask with 0.05% Trypsin-EDTA. The cells were resuspended in RPMI 1640 supplemented with 5% FBS and 1% ITS. A subsample (50 ul) of the cells was mixed 1:1 with volume of trypan blue (Sigma, St. Louis, MO), then loaded onto a hemocytometer (Hausser Scientific, Horsham, PA), and viable cells were counted on a Nikon inverted microscope (Mager Scientific, Dexter, MI). Cells were seeded onto fibronectin coated (20 ug/ml) (Sigma, Inc., St. Louis, MO) 12-well Costar tissue culture plates (Costar, Corning, NY) at the appropriate density for each experimental design. The cells were incubated at 37°C in humidified atmosphere with 5% C02 and allowed to grow 2-3 days to form confluent, undifferentiated monolayers. 167 Experimental Design C. jejuni infection of IPEC-l cells for lL-8, IL-l-B, and TN F -a analysis To determine the effect of C. jejuni on IL-8, IL-l-B, and TNF-a secretion from undifferentiated crypt-like cells, confluent IPEC-l monolayers were seeded at a density of 4 x 106 per well and were infected with C. jejuni at multiplicity of infection (MOI) of 40:1 and 200:1 and 400:1. The C. jejuni inocula were prepared by diluting the stock (5 x 108 CF U/ml, OD560 0.1) to the desired multiplicity of infection. The negative control treatment was RPMI 1640 medium without phenol red, PBS or ITS. The positive control treatment for IL-8 and TNF-a induction was phorbol myristate acetate (PMA) and calcium ionophore (CI), each at a final concentration of 100 ng/ml. The positive control treatments for IL-l-fl induction were recombinant swine TNF-a (500 pg/ml per well) or phytohemagglutinin (FHA; 50 11ng per well) (Table 3.1). All treatments were prepared and diluted in RPMI 1640 medium without phenol red, FBS or ITS to eliminate any false positive results. Each treatment was applied in a final volume of one milliliter, and three replicates of each treatment were analyzed for cytokine production. The IPEC-1 cells were exposed to these treatments for O, 8, 24 and 48 hours. The supernatants were collected from the wells at the time intervals described. In order to measure cytokine expression at 0 hours, the growth medium containing RPMI 1640 supplemented with 5% FBS and 1% ITS was removed, and the treatments were immediately applied to the confluent cell monolayer, and the supernatants were immediately collected. All supernatants from each treatment group were centrifuged at 4°C, 10,000 rpm for 5 minutes to remove cell debris, transferred to fresh 1.5 ml tubes, and stored at -80°C. 168 Levels of secreted lL—8, IL-l-B, and TNF-a were measured from the supernatants by enzyme-linked immunoabsorbant assay (ELISA). Assay for IL-8, IL-l-B, and TNF-a secretion IL-8, IL-l-B, and TNF-a were measured using swine sandwich ELISA kits obtained from Biosource International (Camarillo, CA); assays were performed according to the manufacturer’s instructions. Each assay contained a IL-8, IL-l-B, or TNF-a standard protein control. A microplate reader (EL800 Universal Microplate Reader, Bio- Tek Instruments, Winooski, Vermont) was used to measure the absorbance at 450 nm, and the concentration of IL-8, IL-l-B, or TNF-a in each sample was quantified using KCjunior® software (Bio-Tek Instruments, Winooski, VT). Invasion assay To determine the direct effect of exogenous IL-8, IL-l-B, or TNF-a on C. jejuni invasion of IPEC-l cells, confluent monolayers were pretreated with recombinant swine IL-8 (0, 100, 250, 500, 750, or 1000 pg/ml); IL-I-B (0, 3000, 30,000, or 300,0000 pg/ml); or TNF-a (0, 50, 500, or 5000 pg/ml) (Biosource International, Camarillo, CA) in a dose- response design (Table 3.2). The cytokine concentrations used in this experiment were determined by reports from the literature and preliminary data from previously conducted do se-response experiments in our laboratory. Each cytokine was diluted to the appropriate concentration in RPMI 1640 medium without phenol red, FBS or ITS and applied to the cells in a final volume of 1 ml. The monolayers were incubated at 37°C in a humidified atmosphere with 5% CD; for 5 hours. The cytokine pretreatment medium 169 was removed; the monolayers were not washed in order to avoid cell damage or detachment. C. jejuni ATCC 33292 or E. coli DHS-a (noninvasive positive control) inocula were added to the confluent IPEC-1 cells at a MOI of 100:1 (approximately 2 x 108 CFU/ml) and incubated at 37°C in a humidified atmosphere with 5% CO2 for 3 hours to allow bacterial invasion of the cells. The medium was removed and gentamicin (lnvitrogen, Frederick, MD), diluted in RPMI 1640 without phenol red, at a final concentration of 100 ug/ml, was added to the monolayers. The monolayers were incubated for 1 hour to kill the extracellular bacteria, and then the gentamicin was removed. The monolayers were lysed with 0.1% sodium deoxycholate (Sigma, Inc., St. Louis, MO) in 1X Phosphate Buffered Saline (0.01 M). The internalized bacteria were enumerated by serial dilution and spreading on Bolton agar. After 48 hours incubation, the colonies were counted to determine the number of internalized bacteria. All treatments were performed in triplicate. Adherence Assay To determine the direct effect of exogenous IL—8, IL-l-B, or TNF-a on C. jejuni adherence to IPEC-1 cells, confluent monolayers were pretreated with each recombinant swine cytokine as previously described for the invasion assay (Table 3.2). After cytokine pretreatment, C. jejuni ATCC 33292 or E. coli 0157:H43 (DEC7A) (adherence positive control strain) inocula were added to the confluent IPEC-l cells at a MOI of 100:1 (approximately 2 x 108 CF U/ml) and incubated at 37°C in a humidified atmosphere with 5% CO2 for 1 hour to allow bacterial adherence to the cells. The medium was then removed; the monolayers were not washed in order to avoid cell damage or detachment. I70 The monolayers were lysed with 0.1% sodium deoxycholate (Sigma, Inc., St. Louis, MO) in 1X Phosphate Buffered Saline (0.01 M). The cell-associated (adherent) bacteria were enumerated by serial dilution plating on Bolton agar and counting CFUs at 48 hours after incubation. All treatments were performed in triplicate. Transepithelial resistance assay To determine the effect of exogenous recombinant cytokines on IPEC-1 mono layer integrity, we measured the transepithelial resistance (TER) across the monolayers before and after treatment with recombinant swine IL-8, IL-l-B, or TNF-a (Biosource International, Camarillo, CA). IPEC-l cells were seeded at a density of 2 x 106 per well onto 24-Transwell inserts (6.5 mm diameter, 3 pm pore size, Costar, Corning, NY) coated with 20 ug/ml fibronectin in order to measure the TER. The cells were incubated at 37°C in a humidified atmosphere with 5% C02 and allowed to grow 2- 3 days to form confluent monolayers. All treatments were prepared and diluted in RPMI 1640 medium without phenol red, FBS or ITS and applied to the cells in a final volume of 1 mL. The TER across the IPEC-l monolayers was measured before cytokine treatment using an electrode (EVOMX, World Precision Instruments, Sarasota, FL) to assess monolayer integrity. TER values between 150-600 Qcm2 were considered evidence of confluent monolayers. The monolayers were exposed to recombinant swine IL-8 (250 pg/ml), IL-l-B (3000 pg/ml), TNF-a (500 pg/ml), or 0 pg/ml. They were next incubated at 37°C in a humidified atmosphere with 5% CO2 for 5 hours, and then the TER was measured again. All treatments were performed in quadruplicate. 171 Statistical Analysis ELISA data were analyzed using the mixed model procedure in the Statistical Analysis System (SAS)(SAS Institute Inc., Cary, NC) program. The fixed effects in this model were time and treatment. The random effect in this model was the plate. P values S 0.05 were considered significant. Invasion and adherence data were analyzed using the mixed model procedure in the Statistical Analysis System (SAS, SAS Institute Inc., Cary, NC) program. The fixed effects in this model were experiment and treatment. The random effect in this model was the plate. P values S 0.05 were considered significant. Transelectrical resistance (TER) data were analyzed in the Statistical Analysis System (SAS)(SAS Institute Inc., Cary, NC) program. A generalized linear model with one factor (treatment) was used to determine significance. P values 5 0.05 were considered significant. 172 Results Sensitivity of TNF-a, IL-l-B, and lL-8 ELISAs Figures 3.1 to 3.3 show the standard curves for the TNF-a, IL-l-B and IL-8 ELISAs, respectively. The regression curves for these assays were obtained using KCjunior® software (Bio-Tek instruments, Winooski, Vermont). The TNF-a, IL-l-fl and IL-8 regression curves showed significant dose response curves with the concentration range and antibody titer used, respectively, R = 1.000; R = 0.9999; R = 0.9999. The lower limit of detection of the TNF-a and IL-I-B assays is 2 6 pg/ml, and the lower limit of detection of the IL-8 assay is 2 10 pg/ml. Cytokine secretion from C. jejuni-infected IPEC-1 cells Different ratios of bacteria to IPEC-I cells were used in these experiments to determine the time course of induction and optimize the assay (Table 3.1). We exposed undifferentiated IPEC-1 cells to C. jejuni 33292 for 0, 8, 24, and 48 hours to determine levels of TNF-a, IL-l-B, and lL-8 expression following bacterial infection. Figure 3.4 shows that IPEC-1 cells significantly secreted TNF-a post-C. jejuni infection at an optimal MOI of 400:1 at 8 hours (176.97 :h 15.25 pg/ml), 24 hours (354.98 i 15.25 pg/ml), and 48 hours (181.81 :1: 15.25 pg/ml) as compared to the RPMI 1640 medium control at 8 hours (16. 56 :t 18.67 pg/ml), 24 hours (22.83 i 15.25 pg/ml), and 48 hours (28. 07 d: 15.25 pg/ml); all p < 0.0001. We found that C. jejuni-infected IPEC-1 cells showed maximum induction of TNF-a production at a bacterial MOI of 400:1 at 24 hours post-infection. We also note that at 0 hours an elevated level of TNF—a was detected in our RPMI 1640 medium control, and we speculate that this response was 1.73 likely due to physical stress on the IPEC-I cells induced by the rapid manipulation during the experimental technique (described in the materials and methods) used to challenge the cells. We also found that exposure of IPEC-1 cells to C. jejuni 33292 significantly induced IL-l-B (Figure 3.5). By 24 hours post-infection, IL-l-B was induced at a C. jejuni MOI of 400:1(134.27 i 16.12 pg/ml) as compared to the RPMI 1640 medium control (40.47 i 16.12 pg/ml; p = 0.025). In preliminary trials, IPEC-1 cells were tested for positive IL-I-B induction using phytohemagglutinin (PHA 50 ug/ml) or recombinant swine TNF-a (500 pg/ml); over a wide range of time points. Levels of IL-l-B were found to be beneath the level of detection of the assay (TNF-a stimulation) or were not significantly different from the RPMI 1640 control (PHA stimulation). Therefore, induction in this assay was defined as a positive response that was more than 3 fold greater than the control. Because the possible positive control treatments were weak or unavailable, this could be an experimental drawback. Follow-up experiments were conducted to determine if the recombinant TNF- or was active, and results showed that administered recombinant TNF- a actively stimulated IPEC-1 cells to induce IL-8 (data not shown). However, in this case, recombinant swine TNF- u did not effectively stimulate IPEC-1 cells to significantly produce IL-l-B. IPEC-1 cells were not stimulated by C. jejuni 33292 exposure to secrete significant levels of IL-8; however, IL-8 protein was constitutively expressed by IPEC-1 cells. The positive control (PMA/CI) significantly induced lL-8 by 8 hours, and IL-8 levels remained significantly elevated through 48 hours as compared to the RPMI 1640 medium control (Figure 3.6A, all p < 0.0001). Figure 368 shows that IL-8 is 174 constitutively expressed by IPEC-1 cells. From the supernatants of the RPMI 1640 (control), C. jejuni M01 40:1, and C. jejuni M01200:1 treatment groups, we detected IL- 8 production at 24 hours post-treatment and continued detecting increased IL-8 levels through 48 hours. From the supernatants of the C. jejuni 400:1 treatment group, we detected lL-8 production as early as 8 hours post-treatment and continued detecting a rise in IL-8 levels through 48 hours. Although increased IL-8 levels were detected in the supernatants of each treatment group, there were no statistically significant changes in IL-8 secretion in response to C. jejuni infection of IPEC-1 cells (Figure 3.63, p > 0.05). In these experiments, we determined that IL-8 secretion and accumulation was apparently time related and not dependent on the C. jejuni challenge dose given. Effects of cytokine pretreatment on C. jejuni invasion and adherence of IPEC-1 cells IPEC-l cells were pretreated with IL-8, TNF-a, or IL-l-B to determine if these cytokines have a direct effect on the interaction between C. jejuni and crypt-like basal epithelial cells (IPEC-l cells), specifically, effects on the integrity of the epithelial cell barrier. Treatment of IPEC-1 cells with recombinant swine IL-I-B or IL-8 did not have a significant effect on C. jejuni adherence and/or invasion (IL-IB, Figures 3.10 and 3.11; IL-8, Figures 3.13 and 3.14; all p > 0.05). Therefore, C. jejuni 33292 did adhere to and invade IPEC-1 cells regardless of recombinant swine IL-l—B or IL-8 pretreatment of IPEC-1 cells. Also, treatment of IPEC-1 cells with recombinant TNF-a did not have an effect on C. jejuni adherence to IPEC-1 cells (Figure 3.8, p > 0.05), and we could not draw a conclusion about its effects on C. jejuni invasion of IPEC-l cells because of the variability observed in the number of internalized bacteria (Figure 3.7, p > 0.05). E. coli 175 DH5-a (a noninvasive strain) was the control for the invasion assays and neither TNF-a, IL-l-B, nor lL-8 affected E. coli DH5-a interaction with IPEC-1 cells; E. coli DH5-a did not invade IPEC-l cells regardless of cytokine pretreatment of the IECs. E. coli 0157:H43 DEC7A was the control for the adherence assays, and neither TNF-a, IL-l-B, nor IL-8 affected E. coli 0157:H43 DEC7A adherence to IPEC-1 cells (TNF-a, Figure 3.9; IL-IB, Figure 3.12; IL-8, Figure 3.15)(all p > 0.05). Effects of cytokine pretreatment on IPEC-l transepithelial resistance (TER) IPEC-l cells were pretreated with IL-8, TNF-a, or IL-l-B to determine whether these cytokines have a direct effect on the transepithelial electrical resistance of IPEC-l cells. Treatment of IPEC-1 cells with recombinant swine IL-8, TNF-a, or IL-l-B did not have a significant effect on the transepithelial electrical resistance of IPEC-1 cells (Figure 3.16; p > 0.05). IPEC-l cells pretreated with RPMI 1640 medium alone had a TER of ~300 Qcmz. Experimental TER values between 150-600 52cm2 were considered as evidence of confluent monolayers with no evidence of damage to the integrity of the cells post-treatment with recombinant swine cytokines. 176 Discussion C. jejuni invasion or stimulation of intestinal epithelial cells (IECs) has been shown to significantly induce the expression and up-regulation of pro-inflammatory cytokines and chemokines (4, 25-27, 32). In vivo, a robust pro-inflammatory cytokine response is detected in crypt-like epithelial cells (Jones, dissertation, 2005), and Campylobacter organisms are associated with these surface epithelial cells within the crypts and were found within the crypts of lymphoglandular complexes in the distal colon (Jones, dissertation, 2005)(40). Pro-inflammatory cytokines such as IL-8, TNF-a and IL- l-B can act as mediators of inflammation and regulators of immune responses which are associated with an innate immune response that facilitates bacterial elimination during infection (37). In this study, we hypothesized that intestinal pig epithelial cells (IPEC-1 cells), which serve as a model of undifferentiated, crypt-like cells, secrete pro- inflammatory cytokines (specifically IL-8, TNF-a, and IL-l-B) in response to C. jejuni infection when challenged in vitro. We also examined the treatment of confluent IPEC-l monolayers with recombinant swine IL-8, TNF-a, or IL-I-B to determine the direct effects of these cytokines on C. jejuni adherence, invasion, and/or the transepithelial electrical resistance of the IPEC-l cells. IPEC-1 cells were exposed to 5 x 108 CFU/ml of C. jejuni 33292 for 0, 8, 24 and 48 hours. At 8, 24, and 48 hours post-infection, we measured significantly increased levels of TNF-a compared to the uninfected controls. Maximum TNF-a induction was detected 24 hours following bacterial infection at an MOI of 400: 1. Likewise, we measured significant levels of IL-l-B at 24 hours post-infection. Based on the data 177 presented here, C. jejuni challenge induces IPEC-l cells to secrete TNF-a and IL-l-B at significant levels. This finding is comparable to other in vitro reports that demonstrate the production of pro-inflammatory cytokines by intestinal epithelial cells following C. jejuni infection (4, 9, 25, 27). Human intestinal epithelial cells, INT 407 cells, secrete an array of pro-inflammatory chemokines (IL-8, MCP-l, GROalpha, GROgamma, and gammaIP-IO) when stimulated with C. jejuni (4, 27). C. jejuni has also been shown to stimulate INT-407 cells to produce intracellular IFN-y, IL-lO, TNF-a, and IL-4 (4, 47). Induction of pro-inflammatory cytokines to C. jejuni challenge is also observed in viva. Pigs rectally challenged with C. jejuni for 1 hour show significant up-regulation of intracellular TNF-a, IL-18, IL-8, GM-CSF, IL-l-B, iNOS, and MCP-1 in the distal colon and lymphoglandular complexes (LGCs)( Jones, dissertation, 2005). It has also been demonstrated in Balb/c mice that C. jejuni induces a peritoneal inflammatory cytokine response and increased peritoneal phagocyte oxidative activity (31, 47). Given that increased pro-inflammatory cytokine expression elicited by C. jejuni infection of intestinal cells has been demonstrated in vitro and in viva, it was not surprising that we detected significant elevated TNF-a and IL-l-B levels secreted by IPEC-l cells following C. jejuni exposure; the production of these cytokines likely plays a significant role in host innate immunity to this bacteria. Furthermore, in this in vitro, C. jejuni-challenge study, the cytokine response of the undifferentiated IPEC-l cells is consistent with other in viva and in vitro findings (4, 9, 25, 27, 53) (Parthasarathy, dissertation, 2004 and Jones, dissertation, 2005). Because IPEC-1 is a non-irnmortalzied cell line, the cytokine secretion pattern is likely more reflective of the physiology in the host than that represented by an immortalized cell line. 178 This is strengthened by the fact that concurrent studies conducted in our laboratory have shown that C. jejuni also invade differentiated IPEC-1 cells and stimulate these cells to produce cytokines such as IL-18, IL-6, or IL-10 (Parthasarathy, dissertation, 2004 and Jones, dissertation, 2005). Likewise, previous in viva data from our laboratory show that C. jejuni stimulates a proinflammatory cytokine response after invading intestinal tissue and has been identified in the intestinal crypt cells of infected pigs (Kathryn, dissertation, 2005). Our findings support the hypothesis that primary, undifferentiated intestinal pig epithelial cells (IPEC-1) can serve as a more suitable model to mimic the enteric immune response of human undifferentiated intestinal crypt cells as compared to other in vitro systems that involve the use of immortalized cell lines, such as INT 407, T84, and HEp-2 cells. IPEC-1 cells were derived from a newborn unsuckled piglet, and pigs are monogastric and have dietary habits and anatomical and physiological characteristics similar to humans (7). Therefore, the overall similarities between cultured primary intestinal pig epithelial cells (IPEC-1) and human intestinal cells allow for a better evaluation of bacterial virulence, host immune mechanisms, and the possible role played by intestinal epithelial cells as an important source and initiator of enteric immune responses following assault. The functions of IL-l-B and TNF-a are similar and play a significant role in enteric immune responses. TNF-a activates neutrophils, mononuclear phagocytes and eosinophils which can amplify the mucosal‘inflammatory response to efficiently eradicate infectious microorganisms (1). In viva, mice exposed to a primary L. manacytagenes or C. jejuni infection produced significant plasma TNF-a which is believed to enhance innate immune functions(37). Together or independently, TNF-a or IL-l-B can also 179 stimulate inflammatory cells to produce IL—8 which contributes to neutrophil influx, and the combined action of TNF-a, IL-l-B and IL-6 contribute to systemic acute-phase effects which can induce fever (increased body temperature is not conducive for the growth of some pathogenic bacteria)(57). In addition, it is suggested that IL-l-B plays a pivotal role in early events leading to inflammation (50, 57). In vitro, nontransformed epithelial cells rapidly express IL-l-B mRN A, and secreted IL-l-B initiates and enhances the acute inflammatory response following bacterial infection (16, 29, 57). IL-l-B has also been shown to be predominantly expressed in undifferentiated cells located in the basal mucosal crypts following acute experimental colitis . Therefore, in agreement with these findings, the present data suggest that the significant production of TNF-a and IL-l-B in response to C.jejuni infection of IPEC-l cells may play a significant role in initiating and modulating immune responses that facilitate the elimination of C. jejuni infection in viva. IL-8 is a proinflammatory cytokine that is a chemoattractant which induces adherence to vascular endothelium and extravasation into tissues (17). We found that IPEC-1 cells constitutively secrete IL-8 protein, and IL-8 production was not induced by C. jejuni infection. The constitutive expression of IL-8 by the undifferentiated, crypt- like IPEC-1 cells is consistent with other reports showing that various intestinal epithelial cell lines (T84, Caco-2, SW620, and HT29) constitutively secrete IL-8. Also, some of these cell lines can significantly express IL-8 depending on the stimulant (17, 18, 25, 43). Even though IPEC-1 cells were not induced to secrete IL-8 following C. jejuni exposure, the constitutive expression of IL-8 protein constitutes a significant inflammatory response when one considers the number of epithelial cells lining the colon. In viva, the gut epithelium is continuously exposed to a variety of harmless microflora, antigenic dietary 180 constituents, and bacterial pathogens that can damage it by directly invading the mucosa or by toxin production. It is possible that there is a continuous, basal level of inflammation within the intestinal mucosa which can be up-regulated when necessary in response to pathogens and noxious antigens thereby providing protective immunity (16, 60). The constitutive production of IL-8 at low levels along the gut mucosal epithelium could be partially responsible for a consistent low-level. inflammatory cellular infiltration of neutrophils and monocytes which would be favorable to the host (16, 25, 43). Therefore, we suggest that intestinal pig epithelial cells (IPEC-l cells) constitutively secrete IL-8 protein as a molecular inflammatory mediator of innate host defense which may play a role in limiting microbial exposure at the mucosal surface and maintaining tissue integrity for low level insults. 0n the other hand, mature differentiated epithelial cells lining the villus tips of the colon or dendritic cells and macrophages populating the lamina propria immediately underlying the epithelium may be responsible for the IL-8 expression hypothesized to draw neutrophils to the this site. Because others have reported that IL-8 is significantly induced via NF KB activation through lea degradation following Salmonella Dublin, enteroinvasive Escherichia coli, Yersinia enterocolitica, Helicobacter pylori, or C. jejuni invasion or stimulation of IECs (15, 20), we believe it is necessary to discuss a few possible reasons why IL—8 was not significantly induced by C. jejuni infection of IPEC-1 cells in our study. High concentrations of diverse bacteria and bacterial products continually interact with the intestinal mucosal epithelium. Because of this intimate contact, it is critical that the intestinal epithelia facilitate the control and maintenance of a normal healthy state to avoid persistent, dysregulated inflammation. Gut epithelial cells express toll-like 181 receptors (TLRs) which are a family of transmembrane receptors also termed pattem- recognition receptors (PRRs) (33, 42). TLRs play an important role in host innate immunity by recognizing pathogen-associated molecular patterns (PAMPs) present on diverse microbes including Gram-positive and Gram-negative bacteria, fungi, and mycobacteria (33). In general, infection of IECs can lead to activation of the TLRs which then initiates intracellular signaling cascades which can lead to the activation of the NFKB/Rel family transcription factors and cytokine production (2, 3, 38). NFKB has been described as the central regulator of the intestinal epithelial cell innate immune response induced by enteroinvasive bacterial infection (20, 38). NFKB is activated by many extracellular stimuli via TLR interaction, and there are many signaling pathways that lead to NF KB activation which leads to the expression of many genes, including those encoding inflammatory cytokines (14, 38). More specifically, IL-8 production has been associated with TLR4 receptor signaling via LPS stimulation and NFKB activation (2, 3, 13, 21). It has been shown that immune-mediated signals regulate TLR-4 and MD-2 (a TLR4 coreceptor accessory molecule) expression, and changes in TLR-4 and MD-2 expression impact host cell responses to bacterial lipopolysaccharide (LPS) (20, 46, 60). In addition, it has been reported that TLR4 is barely detectable in isolated primary intestinal epithelial cells (3 8). Therefore, it is possible that decreased expression of Toll-like receptor-4 (TLR-4) and MD-2 or inhibition of intracellular signaling via the transcription factor can affect assessment of proinflammatory gene expression in some in vitro systems (3, 33). Hence, if TLR4 and its accessory molecules are only present at low levels or absent from the cell surface of IPEC-1 cells (isolated primary intestinal epithelial cells), then C. jejuni LPS either weakly interacts with TLR4 182 and its accessory molecules or does not bind the receptor complex to induce significant production of IL-8 thereby causing IPEC-1 cells to be unresponsive to this particularly bacterial component (C. jejuni LPS). It is also reasonable to think that C. jejuni-infected IPEC-l cells can significantly produce IL-l-B and TNF-a and do not significantly induce IL-8 expression because induction of these cytokines is likely regulated via different Toll-like receptor signaling or different intracellular signaling pathways. It has been demonstrated that the flagellin of other bacteria, such as Listeria manacytagenes and Salmonella typhimurium , can bind to TLR5 receptors (TLRs that specifically recognize bacterial flagellin) to stimulate TNF-a, IL-8, or lL—l—B production (22, 33, 55), whereas, C. jejuni flagellin has been reported to be a poor stimulator of TLR5 and does not play a significant role in stimulating IL-8 production nor has it been tested to determine if it stimulates TNF—a or IL-l-B production (58). Even though not tested in this study, we suggest that C. jejuni-infected IPEC-1 cells express lL-l-B, TNF-a, or IL-8 via different TLR ligand signaling recognition or other intracellular signaling pathways. Additionally, recent multilocus sequence typing studies of C. jejuni strains in our lab suggest that some strains have LOS variants that may be more stimulating than others. Further studies are needed to elucidate the regulatory mechanisms involved in the interaction between C. jejuni bacterial products or components and intestinal epithelial cell TLR activation, intracellular signaling pathways, and cytokine gene expression and secretion. Secreted cytokines can exert autocrine (self-stimulation), paracrine (adjacent), intracrine (within the cell) and some endocrine (distant) actions (23, 33, 42). Cytokines have been shown to modulate bacterial-enterocyte interactions in vitro. For instance, cytokines can disrupt intestinal epithelial cell function or barrier integrity (24, 48, 51). 183 Increased bacterial adherence has been associated with IFN-y pretreatment of IECs suggesting that IFN-y augments bacterial adherence to the intestinal epithelium (24). In addition, IF N -y and TNF-u act synergistically on the epithelial barrier by causing reduced TER and altering cell morphology (21). Likewise, concurrent studies in our laboratory have shown that recombinant swine IL-4 (rIL-4) pretreatment of differentiated IPEC-1 cells significantly reduced the TER of IPEC-1 cells, increased C. jejuni internalization, and caused differential effects on adherence of two C. jejuni strains (Parthasarathy, dissertation, 2004). Because of the impact cytokines have on enterocytes and the possible roles they play in bacterial-enterocyte interaction, in this study, we pretreated the IPEC-1 cells with recombinant swine IL-8, TNF-a, or IL-l-B to determine if these cytokines would affect C. jejuni invasion, adherence and/or the transepithelial electrical resistance (TER) of IPEC-1 cells. Our results showed that C. jejuni 33292 does adhere to and invade IPEC-l cells, and these data support previous findings from our laboratory (Parthasarathy, dissertation, 2004). However, neither IL-8 nor IL-l-B has a significant direct effect on C. jejuni invasion, adherence, and/or changes in transepithelial electrical resistance of undifferentiated IPEC-1 cells. Also, treatment of IPEC-1 cells with TNF-a did not significantly affect C. jejuni adherence or changes in the TER of the IECs. However, we were unable to draw conclusions about the effects of TNF-a on C. jejuni invasion of IPEC-1 cells because of the variability observed in these experimental results. Additional experiments could be conducted to address this concern; however, the lack of a significant effect under the rigorous experimental conditions employed, suggest efforts might be better placed elsewhere. Even though there was not a direct effect on C. jejuni adherence to or invasion of IPEC-l. cells (in response to 1L-8 or IL-l-B pretreatment) or a 184 direct effect could not be determined following TNF-a pretreatment in vitro, it is still possible that IL-8, TNF-a, or IL-l-B may affect epithelial function in a paracrine-like or another autocrine-like manner during C. jejuni infection in viva. For instance, it has been demonstrated that cytokines can facilitate enterocyte proliferation, maturation, MHC class II expression, cytokine receptor expression or apoptosis in healthy and diseased states (21, 48, 51). In addition, TNF-a and TGF-a have been shown to stimulate immature crypt cell proliferation, and TGF—B and INF-y can inhibit mitotic activity (52). IFN-y can alter immature crypt epithelial cell turnover and upregulate MHC class II expression (51). Therefore, alterations of cytokine concentrations as a consequence of bacterial exposure and intestinal inflammation may contribute to serious pathologic consequences. Further elucidation of the effects of distinct cytokines on epithelial cell growth, phenotype, firnction and bacterial-enterocyte interactions would be of importance in the understanding of cytokine mediated regulation of mucosal immune responses associated with C. jejuni infection. In summary, we showed that undifferentiated, crypt-like intestinal pig cells (IPEC—1) can serve as a suitable in vitro model for studying C. jejuni-enterocyte interaction and host enteric immune responses elicited by C. jejuni during infection. We also found that C. jejuni infection of these cells significantly induces TNF-a and lL-l-B production. In addition, we observed that IPEC-1 cells constitutively secrete IL-8. The data presented here suggest that TNF-a and IL-l-B production and constitutively secreted IL-8 are significant inflammatory responses which are associated with an acute innate immune response and may lead to a healing Thl-type adaptive immune response. It is likely that this inflammatory response can generate and/or modulate the immune response 185 to benefit the host by contributing to the elimination of the bacteria. In addition, we found that neither lL-8 nor IL-l-B had a direct effect on C. jejuni adherence, invasion, or the TER of IPEC-l cells. Likewise, we found that TNF-a did not have a direct effect on C. jejuni adherence to IPEC-l cells or the TER of IPEC—1 cells. Even though these findings showed no direct effect of these cytokines on C. jejuni interactions (specifically adherence, invasion, or changes in IPEC-l TER) with the swine intestinal epithelial cells, they could possibly have other autocrine- or paracrine-like effects which could play a role in host inflammatory immune responses associated with C. jejuni infection. For example, TNF-a and IL-l-B can stimulate intestinal epithelial cells to produce C3, a complement component which is associated with host acute-phase inflammatory responses (21, 44, 45, 48, 51). So, given that our data support the hypothesis that TNF-a, IL-l-B, and IL-8 are cytokines that are important components of the inflammatory immune response to enteric pathogens, we can utilize this information to more specifically determine, characterize, and elucidate the possible roles played by IL-8, TNF-a, and/or IL-l-B during C. jejuni infection. 186 10. 11. References Abram, M., D. Vu kovic, B. Wraber, and M. Doric. 2000. Plasma cytokine response in mice with bacterial infection. Mediators Inflamm 9:229-234. Abreu, M. T., E. T. Arnold, L. S. Thomas, R. Gonsky, Y. Zhou, B. Hu, and M. Arditi. 2002. TLR4 and MD-2 expression is regulated by immune-mediated signals in human intestinal epithelial cells. J Biol Chem 277:20431-20437. Abreu, M. T., P. Vora, E. F aure, L. S. Thomas, E. T. Arnold, and M. Arditi. 2001. Decreased expression of Toll-like receptor-4 and MD-2 correlates with intestinal epithelial cell protection against dysregulated proinflammatory gene expression in response to bacterial lipopolysaccharide. J Immunol 167:1609- 1616. Al-Salloom, F. S., A. A1 Mahmeed, A. Ismaeel, G. A. Botta, and M. Bakhiet. 2003. Campylobacter-stimulated INT407 cells produce dissociated cytokine profiles. J Infect 47:217-224. Allos, B. M. 2001. Campylobacter jejuni Infections: update on emerging issues and trends. Clin Infect Dis 32:1201-1206. Altekruse, S. F ., N. J. Stern, P. 1. Fields, and D. L. Swerdlow. 1999. Campylobacter jejuni--an emerging foodbome pathogen. Emerg Infect Dis 5:28- 35. Babakhani, F. K., G. A. Bradley, and L. A. Joens. 1993. Newborn piglet model for campylobacteriosis. Infect Immun 61:3466-3475. Babakhani, F. K., and L. A. Joens. 1993. Primary swine intestinal cells as a model for studying Campylobacter jejuni invasiveness. Infect Immun 61:2723- 2726. Bakhiet, M., F. S. Al-Salloom, A. Qareiballa, K. Bindayna, I. F arid, and G. A. Botta. 2004. Induction of alpha and beta chemokines by intestinal epithelial cells stimulated with Campylobacter jejuni. J Infect 48:236-244. Baqar, S., B. Rice, L. Lee, A. L. Bourgeois, A. N. El Din, D. R. Tribble, G. P. Heresi, A. S. Mourad, and J. R. Murphy. 2001. Campylobacter jejuni enteritis. Clin Infect Dis 33:901-905. Black, R. E., M. M. Levine, M. L. Clements, T. P. Hughes, and M. J. Blaser. 1988. Experimental Campylobacter jejuni infection in humans. J Infect Dis 157:472-479. 187 12. 13. I4. 15. 16. 17. 18. 19. 20. 21. 22. Blaser, M. J., I. D. Berkowitz, F. M. LaForce, J. Cravens, L. B. Reller, and W. L. Wang. 1979. Campylobacter enteritis: clinical and epidemiologic features. Ann Intern Med 91:179-185. Bruewer, M., A. Luegering, T. Kucharzik, C. A. Parkos, J. L. Madara, A. M. Hopkins, and A. Nusrat. 2003. Proinflammatory cytokines disrupt epithelial barrier firnction by apoptosis-independent mechanisms. J Immunol 171:6164— 6172. Cario, E., I. M. Rosenberg, S. L. Brandwein, P. L. Beck, H. C. Reinecker, and D. K. Podolsky. 2000. Lipopolysaccharide activates distinct signaling pathways in intestinal epithelial cell lines expressing Toll-like receptors. J Immunol 164:966-972. Crabtree, J. E., M. Court, M. A. Aboshkiwa, A. H. Jeremy, M. F. Dixon, and P. A. Robinson. 2004. Gastric mucosal cytokine and epithelial cell responses to Helicobacter pylori infection in Mongolian gerbils. J Pathol 202: 197-207. Darveau, R. P., C. M. Belton, R. A. Reife, and R. J. Lamont. 1998. Local chemokine paralysis, a novel pathogenic mechanism for Porphyromonas gingivalis. Infect Immun 66: 1660-1665. Eckmann, L., H. C. Jung, C. Schurer-Maly, A. Panja, E. Morzycka- Wroblewska, and M. F. Kagnoff. 1993. Differential cytokine expression by human intestinal epithelial cell lines: regulated expression of interleukin 8. Gastroenterology 105: 1689-1697. Eckmann, L., M. F. Kagnoff, and J. Fierer. 1993. Epithelial cells secrete the chemokine interleukin-8 in response to bacterial entry. Infect Immun 61:4569- 4574. Edmonds, J. 1984. Reactive arthritis. Aust N Z J Med 14:81-88. Elewaut, D., J. A. DiDonato, J. M. Kim, F. Truong, L. Eckmann, and M. F. Kagnoff. 1999. NF-kappa B is a central regulator of the intestinal epithelial cell innate immune response induced by infection with enteroinvasive bacteria. J Immunol 163:1457-1466. Fish, S. M., R. Proujansky, and W. W. Reenstra. 1999. Synergistic effects of interferon gamma and tumour necrosis factor alpha on T84 cell function. Gut 45:191-198. Hayashi, F ., K. D. Smith, A. Ozinsky, T. R. Hawn, E. C. Yi, D. R. Goodlett, J. K. Eng, S. Akira, D. M. Underhill, and A. Aderem. 2001. The innate immune 188 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. response to bacterial flagellin is mediated by Toll-like receptor 5. Nature 410: 1099-1 103. Henderson, B., S. Poole, and M. Wilson. 1996. Bacterial modulins: a novel class of virulence factors which cause host tissue pathology by inducing cytokine synthesis. Microbiol Rev 60:316-341. Hess, D. J., M. J. Henry-Stanley, E. A. Erickson, and C. L. Wells. 2002. Effect of tumor necrosis factor alpha, interferon gamma, and interleukin-4 on bacteria- enterocyte interactions. J Surg Res 104:88-94. Hickey, T. E., S. Baqar, A. L. Bourgeois, C. P. Ewing, and P. Guerry. 1999. Campylobacter jejuni-stimulated secretion of interleukin-8 by INT407 cells. Infect Immun 67 :88-93. Hickey, T. E., A. L. McVeigh, D. A. Scott, R. E. Michielutti, A. Bixby, S. A. Carroll, A. L. Bourgeois, and P. Guerry. 2000. Campylobacter jejuni cytolethal distending toxin mediates release of interleukin-8 from intestinal epithelial cells. Infect Imrnun 68:6535-6541. Hu, L., and T. E. Hickey. 2005. Campylobacter jejuni induces secretion of proinflammatory chemokines fiom human intestinal epithelial cells. Infect Immun 73:4437-4440. Hughes, R. A., R. D. Hadden, N. A. Gregson, and K. J. Smith. 1999. Pathogenesis of Guillain-Barre syndrome. J Neuroimmun01100:74-97. Hyland, K. A., D. R. Brown, and M. P. Murtaugh. 2006. Salmonella enterica serovar Choleraesuis infection of the porcine jejunal Peyer’s patch rapidly induces IL-lbeta and lL-8 expression. Vet Immunol Immunopathol 109:1-11. Jones, M. A., S. Totemeyer, D. J. Maskell, C. E. Bryant, and P. A. Barrow. 2003. Induction of proinflammatory responses in the human monocytic cell line THP-l by Campylobacter jejuni. Infect Immun 71:2626-2633. Jung, H. C., L. Eckmann, S. K. Yang, A. Panja, J. F ierer, E. Morzycka- Wroblewska, and M. F. Kagnoff. 1995. A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion. J Clin Invest 95:55-65. Kagnoff, M. F., and L. Eckmann. 1997. Epithelial cells as sensors for microbial infection. J Clin Invest 100:6-10. Kelly, D., and S. Conway. 2005. Bacterial modulation of mucosal innate immunity. Mol Immunol 42:895-901. 189 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. Ketley, J. M. 1997. Pathogenesis of enteric infection by Campylobacter. Microbiology 143 (Pt 1):5-21. Koga, M., M. Gilbert, J. Li, S. Koike, M. Takahashi, K. F urukawa, K. Hirata, and N. Yuki. 2005. Antecedent infections in Fisher syndrome: a common pathogenesis of molecular mimicry. Neurology 64:1605-1611. Konkel, M. E., B. J. Kim, V. Rivera-Amill, and S. G. Garvis. 1999. Bacterial secreted proteins are required for the intemaliztion of Campylobacter jejuni into cultured mammalian cells. Mol Microbiol 32:691-701. Kuby, J. 1997. Immunology. Li, X., and G. R. Stark. 2002. NFkappaB-dependent signaling pathways. Exp Hematol 30:285-296. Mansfield, L. S., and D. T. Gauthier. 2004. Lymphoglandular complexes are important colonic sites for immunoglobulin A induction against Campylobacter jejuni in a swine disease model. Comp Med 54:514-523. Mansfield, L. S., D. T. Gauthier, S. R. Abner, K. M. Jones, S. R. Wilder, and J. F. Urban. 2003. Enhancement of disease and pathology by synergy of Trichuris suis and Campylobacter jejuni in the colon of immunologically naive swine. Am J Trop Med Hyg 68:70-80. McDowell, R. M., and M. D. McElvaine. 1997. Long-term sequelae to foodbome disease. Rev Sci Tech 16:337-341. Medzhitov, R., and C. J aneway, Jr. 2000. The Toll receptor family and microbial recognition. Trends Microbi018:452-456. Mellits, K. H., J. Mullen, M. Wand, G. Armbruster, A. Patel, P. L. Connerton, M. Skelly, and I. F. Connerton. 2002. Activation of the transcription factor NF -kappaB by Campylobacter jejuni. Microbiology 148:2753-2763. Moon, M. R., A. A. Parikh, T. A. Pritts, C. Kane, J. E. Fischer, A. L. Salzman, and P. 0. Hasselgren. 2000. Interleukin-lbeta induces complement component C3 and IL-6 production at the basolateral and apical membranes in a human intestinal epithelial cell line. Shock 13:374-378. Moon, R., A. A. Parikh, C. Szabo, J. E. Fischer, A. L. Salzman, and P. 0. Hasselgren. I997. Complement C3 production in human intestinal epithelial cells is regulated by interleukin lbeta and tumor necrosis factor alpha. Arch Surg 132:1289-1293. 190 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. Neish, A. S., A. T. Gewirtz, H. Zeng, A. N. Young, M. E. Hobert, V. Karmali, A. S. Rao, and J. L. Madara. 2000. Prokaryotic regulation of epithelial responses by inhibition of IkappaB-alpha ubiquitination. Science 289:1560-1563. Pancorbo, P. L., M. A. de Pablo, E. Ortega, A. M. Gallego, C. Alvarez, and G. Alvarez de Cienfuegos. 1999. Evaluation of cytokine production and phagocytic activity in mice infected with Campylobacter jejuni. Curr Microbial 39:129-133. Panja, A., S. Goldberg, L. Eckmann, P. Krishen, and L. Mayer. 1998. The regulation and functional consequence of prainflammatary cytokine binding on human intestinal epithelial cells. J Immunol 161:3675-3684. Peterson, M. C. 1994. Clinical aspects of Campylobacter jejuni infections in adults. West J Med 161:148-152. Radema, S. A., S. J. van Deventer, and A. Cerami. 1991. Interleukin 1 beta is expressed predominantly by enterocytes in experimental colitis. Gastroenterology 100:1180-1186. Ruemmele, F. M., C. Gurbindo, A. M. Mansour, R. Marchand, E. Levy, and E. G. Seidman. 1998. Effects of interferon gamma on growth, apoptosis, and MHC class II expression of immature rat intestinal crypt (IEC-6) cells. J Cell Physiol 176: 120-126. Ruemmele, F. M., and E. G. Seidman. 1998. Cytokine--intestina1 epithelial cell interactions: implications for immune mediated bowel disorders. Zhonghua Min Guo Xiao Er Ke Yi Xue Hui Za Zhi 39:1-8. Smith, C. K., P. Kaiser, L. Rothwell, T. Humphrey, P. A. Barrow, and M. A. Jones. 2005. Campylobacter jejuni-induced cytokine responses in avian cells. Infect Immun 73:2094-2100. Takahashi, M., M. Koga, K. Yokoyama, and N. Yuki. 2005. Epidemiology of Campylobacter jejuni isolated from patients with Guillain-Barre and Fisher syndromes in Japan. J Clin Microbiol 43:335-339. Tallant, T., A. Deb, N. Kar, J. Lupica, M. J. de Veer, and J. A. DiDonato. 2004. Flagellin acting via TLR5 is the major activator of key signaling pathways leading to NF—kappa B and prainflammatary gene program activation in intestinal epithelial cells. BMC Microbial 4:33. Walker, R. 1., M. B. Caldwell, E. C. Lee, P. Guerry, T. J. Trust, and G. M. Ruiz-Palacios. 1986. Pathaphysialogy of Campylobacter enteritis. Microbiol Rev 50:81-94. 191 57. 58. 59. 60. Waterhouse, C. C., and A. W. Stadnyk. 1999. Rapid expression of IL-lbeta by intestinal epithelial cells in vitro. Cell Immunol 193:1-8. Watson, R. 0., and J. E. Galan. 2005. Signal transduction in Campylobacter jejuni-induced cytokine production. Cell Microbial 7 :655-665. Wooldridge, K. G., P. H. Williams, and J. M. Ketley. 1996. Host signal transduction and endocytosis of Campylobacter jejuni. Microb Pathog 21:299- 305. . Xavier, R. J., and D. K. Podolsky. 2000. Microbiology. How to get along-- friendly microbes in a hostile world. Science 289:1483-1484. 192 Table 3.1. IPEC-1 cells treatments for cytokine measurement (ELISA) Treatment Group Treatment 1 Positive controls: IL-8 and TNF-a phorbol myristate acetate (PMA) (100 ng/ml) calcium ionophore (CI) (100 ng/ml) IL- 1 -B phytohemagglutinin (FHA, 50 ug/ml) or recombinant swine TNF-a (rTNF-a, SOOpg/ml) 2 Campylobacterjejuni 33292 (MOI 40:1) 3 Campylobacterjejuni 33292 (MOI 200: I) 4 Campylobacterjejuni 33292 (M01 400: 1) 5 Negative control: RPMI 1640 without phenol red, fetal bovine serum (FBS), or insulin-transferrin-selelium (ITS) I93 Table 3.2 Recombinant swine cytokine pretreatment concentrations for invasion and adherence assays All treatments were suspended in RPMI 1640 without phenol red, fetal bovine serum (F BS) or insulin-transferrin-selenium (ITS). The negative control was identified as medium only. Treatment Recombinant IL-8 Recombinant IL-l-B Recombinant Group (rIL-8) (rIL-l-B) TNF-a (rTNF-a) Negative control: Negative control: Negative control: 1 Medium only Medium only Medium only 0 pg/ml 0 pg/ml 0 pg/ml 2 100 pg/ml 3000 pg/ml 50 pg/ml 3 250 pg/ml 30,000 pg/ml 500 pg/ml 4 500 pg/ml 300,000 pg/ml 5,000 pg/ml 5 750 pg/ml Not applicable Not applicable 6 1000 pg/ml Not applicable Not applicable 194 1.2 1 _— E = 0.8 at v o 2 06 a . .o h ,2 0.4 r 0 Standards 4: — Standard 0.2 - curve I O T I I I 7 0 500 1000 1500 2000 2500 3000 Concentration (pg/ml) 4 Parameter (y = (A - D) / (1 + (x/C)"B) + D) A=19.1384 B=-1.0057 C=47262. 147 D=0.0298, R-Square = 1.000 Figure 3.1. Standard curve for swine TNF-a used in the ELISA. The curve is representative of three identical experiments. I95 1.2 1 + P g 0.8 -— g . ‘ cu U 0.6 "r : a a: E 0.4 -- 0 Standards E — Standard < 0 2 fl curve 0 we 1 1 t : 0 500 1000 1500 2000 2500 3000 Concentration (pg/ml) 4 Parameter (y = (A — D) / (1 + (x/C)"B) + D) A=2.556O B=-I.1 l 16 C=3407.3799 D=0.0077, R-Square = 0.9999 Figure 3.2. Standard curve for swine IL-l-B used in the ELISA. The curve is representative of three identical experiments. I96 0.9 r 0.8 ‘r 0.7 t 0.6 “r 0.5 at 0.4 2* 0 Standards — Standard curve 0.3 *— Ahsorhance (nm) 0.2 “r 0.1 *- 0 , i i l l O 500 1000 1500 2000 2500 Concentration (pg/ml) 4 Parameter (y = (A — D) / (1 + (x/C)"B) + D) A=3.3492 B=-1.3873 C=3570.7411 D=0.0087, R-Square = 0.9999 Figure 3.3. Standard curve for swine IL-8 used in the ELISA. The curve is representative of three identical experiments. 197 400 * 350 :4; 300 B0 hours \ g 250 - * * 138 hours a 200 'd I '3' 150 :i': _ .24 hours ll, 2 ll :l: :l: B48 hours g I“ ' I'l r't 50 ‘ I'l 'l' I'I Ill _d m III '|' r't u't Us I 'I' I'I I't O _. PMA/CI RPMI 1640 C. jejuni C. jejuni C. jejuni MOI 40:1 MOI 200:1 MOI 400:1 Treatment groups Figure 3.4. TNF-a secretion by IPEC-1 monolayers after exposure to C. jejuni 33292. Phorbol myristate acetate (PMA 100 ng/ml) and calcium ionophore (CI 100 ng/ml) were used as the positive control. RPMI 1640 medium was used as the negative control. All treatments were performed in triplicate and are representative of three identical experiments. Values shown represent the mean 3: SD. An asterisk (*) indicates the significant difference between C. jejuni MOI of 400:1 and the RPMI 1640 (uninfected) control at 8, 24, and 48 hours post-infection (P < 0.0001). 198 180 : 160 E 140 I0 hours m ‘9 120 e 100 1118 hours 4.) g 80 , . I I I .24 hours H 60 :l : I I g 40 i: i i 3 ': ' I :48 hours a 20 'I I I 'l : I l rTNF- PHA RPMI 1640 C. jejuni C. jejuni C. jejuni alpha MOI 40:1 MOI 200:1 MOI 400:1 Treatment groups Figure 3.5. IL-l-B secretion by IPEC-1 monolayers after exposure to C. jejuni 33292. Recombinant swine TNF-alpha (500 pg/ml) or phytohemagglutinin (PHA 50 ug/ml) was used as the positive control. RPMI 1640 medium was used as the negative control. All treatments were performed in triplicate and are representative of three identical experiments. Values shown represent the mean 3: SD. An asterisk (*)indicates a significant difference between C. jejuni MOI of 400:1 and the RPMI 1640 (uninfected) control at 24 hours post-infection (P < 0.001). 199 Figure 3.6 (A and B). The two graphs represent experiments conducted at the same time. (A) Positive control: IPEC-l monolayer positively induced to secrete IL-8 following phorbol myristate acetate (PMA 100 ng/ml) and calcium ionophore (CI 100 ng/ml) exposure. RPMI 1640 medium is the negative control. Values shown represent the mean i SD. An asterisk (*)indicates a significant difference between the positive control values at 8, 24, and 48 hours as compared to the RPMI 1640 control, P value was < 0.0001. (B) IL-8 secretion by IPEC-l monolayers after exposure to C. jejuni 33292. RPMI 1640 medium was used as the negative control. All treatments were performed in triplicate, and the values shown represent three identical experiments. Values shown represent the mean i SD. For all values, P was > 0.05 compared to the RPMI 1640 (uninfected) control. 200 Figure 3.6A. (pg/m1) IL-B 9000 8000 7000 6000 5000 4000 3000 2000 1000 O 0 hours Figure 3.68. IL-8 (pg/m) 1200 1000 8 hours 24 hours Time (hours) 48 hours fiPMA/CI IRPMI 1640 RPMI 1640 C. jejuni C. jejuni MOI 40:1 MOI 200:1 Treatment groups 201 C. jejuni MOI 400:1 IO hours m8 hours I24 hours E48 hours .51 7“ i; *1 O 50 500 Internalized bacteria (CPU) N O O Swine rTNF-alpha concentrations (pg/m1) Figure 3.7. IPEC-1 monolayers were pretreated with increasing amounts of swine rTNF-a for 5 hours to determine the effect of swine rTNF-or on C. jejuni 33292 invasion of intestinal epithelial cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were performed in triplicate. Results shown are representative of three experiments. The values shown represent the mean :1: SD. For all values, P was > 0.05 compared to the RPMI 1640 control. 202 Cell-associated(adherent) bacteria x 10*5 0 50 500 5000 Swine rTNF-alpha concentrations (pg/m1) Figure 3.8. IPEC-1 monolayers were pretreated with increasing amounts of swine rTNF—a for 5 hours to determine the effect of swine rTNF-a on C. jejuni 33292 adherence to intestinal epithelial cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were in triplicate. Results shown are representative of three experiments. The values shown represent the mean i SD. For all values, P was > 0.05 compared to the RPMI 1640 control. 203 bacteria x 10‘5 Cell-associated (adherent) 0 50 500 5000 Swine rTNF-alpha concentrations (pg/m1) Figure 3.9. IPEC-I monolayers were pretreated with increasing amounts of swine rTNF-a for 5 hours to determine the effect of swine rTNF-o. on E. coli 0157:H43 DEC7A adherence to intestinal epithelial cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were in triplicate. Results shown are representative of three experiments. The values shown represent the mean i SD. For all values, P was > 0.05 compared to the RPMI 1640 control. 204 1400 B I. 8 1200 .‘l H 1000 0 U u 800 a .n u 600 0 :1 H 400 I a u 200 0 J.) a H 0 3000 30000 300000 Swine rIL-l-beta concentrations (pg/m1) Figure 3.10. IPEC-1 monolayers were pretreated with increasing amounts of swine rIL-I-B for 5 hours to determine the effect of swine rIL-I-B on C. jejuni 33292 invasion of intestinal epithelial cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were in triplicate. Results shown are representative of three experiments. The values shown represent the mean i SD. F or all values, P was > 0.05 compared to the RPMI 1640 control. 205 Cell-associated (adherent) bacteria x 10‘5 0 3000 30000 300000 Swine rIL-l-beta concentrations (pg/m1) Figure 3.1 1. IPEC-I monolayers were pretreated with increasing amounts of swine rIL—I-B for 5 hours to determine the effect of swine rIL-l-B on C. jejuni 33292 adherence to intestinal epithelial cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were in triplicate. Results shown are representative of three experiments. For all values, P was > 0.05 compared to the RPMI 1640 control. 206 Cell-associated(adherent) bacteria x 10‘5 O 3000 30000 300000 Swine rIL-l—beta concentrations (pg/m1) Figure 3.12. IPEC-I monolayers were pretreated with increasing amounts of swine rIL-I-fi for 5 hours to determine the effect of swine rIL-l-B on E. coli 0157:H43 DEC7A adherence to intestinal epithelial cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were in triplicate. Results shown are representative of three experiments. The values shown represent the mean i SD. For all values, P was > 0.05 compared to the RPMI 1640 control. 207 Internalized bacteria (CFU) Swine rIL—8 concentrations (pg/m1) Figure 3.13. IPEC-1 monolayers were pretreated with increasing amounts of swine rIL-8 for 5 hours to determine the effect of swine rIL—8 on C. jejuni 33292 invasion of intestinal epithelial cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were done in triplicate. Results shown are representative of three experiments. The values shown represent the mean i SD. For all values, P was > 0.05 compared to the RPMI 1640 control. 208 Cell-associated (adherent) bacteria x 10‘5 0 100 250 500 750 1000 Swine rIL-8 concentrations (pg/ml) Figure 3.14. IPEC-1 monolayers were pretreated with increasing amounts of swine rIL-8 for 5 hours to determine the effect of swine rIL-8 on C. jejuni 33292 adherence to intestinal epithelial cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were done in triplicate. Results shown are representative of three experiments. The values shown represent the mean :1: SD. For all values, P was > 0.05 compared to the RPMI 1640 control 209 450 400 350 300 250 200 150 bacteria x 10‘5 100 50 Cell-associated (adherent) 0 100 250 500 750 1000 Swine rIL-B concentrations (pg/m1) Figure 3.15. IPEC-1 monolayers were pretreated with increasing amounts of swine rIL-8 for 5 hours to determine the effect of swine rIL-8 on E. coli 0157:H43 DEC7A adherence to intestinal epithelial cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were in triplicate. Results shown are representative of three experiments. The values shown represent the mean i SD. For all values, P was > 0.05 compared to the RPMI 1640 control. 210 TER % Pretreatment 0 pg/ml rIL-B rIL—l—beta I‘I'NF—alpha control 250 pg/ml 3000 pg/ml 500 pg/ml Treatments Figure 3.16. Effect of swine rIL-8, rIL-I-B, and TNF-a on transepithelial electrical resistance (TER) of IPEC-1 cells. The TER was measured before and after treatment. The pretreatment control represents the TER before any treatments were applied to the IPEC-1 cells. RPMI 1640 medium (0 pg/ml) was used as the negative control. All treatments were done in quadruplicate. The values shown represent the mean :1: SD. For all values, P was > 0.05 compared to the pretreatment control and RPMI 1640. 211 Chapter 4: Summary and Conclusions The long term goals of this laboratory project were to elucidate the mechanisms by which concurrent C. jejuni and T. suis infection cause severe disease and pathology in young swine. The identification and characterization of these mechanisms will be beneficial fi‘om both animal and human health perspectives. C. jejuni is a food- bome pathogen that has been identified throughout the world as one of the leading causes of human gastroenteritis (5, 6, 11). There have been significant advances in understanding the mechanisms by which C. jejuni causes disease, but few immunological studies have been pursued. This is especially true of those using a naturally occurring animal model and associated in vitro systems such as swine intestinal epithelial cells. T. suis is a nematode parasite found in the cecum and colon of swine. Young infected pigs can acquire catarrhal enteritis with serious pathology, such as anorexia, diarrhea, anemia, dehydration, retardation of growth, and death (32, 33). C. jejuni is commonly found in the colon of conventionally-reared pigs without signs of pathology; however, simultaneous T. suis infection results in severe disease and pathology (22-24). The severe pathology observed mimics the clinical disease of Campylobacteriasis in humans. For this dissertation project, we addressed a two-fold hypothesis. The central hypothesis is that C. jejuni induces natural host resistance via pro-inflammatory cytokine responses, which arise as a result of infection. A related hypothesis is that pro- inflammatory cytokine responses elicited by C. jejuni can be down-regulated in the presence of T. suis, thereby promoting C. jejuni infection These hypotheses were tested by evaluating pro- and anti-inflammatory cytokine expression in the feces and intestinal 212 tissues of pigs infected with C. jejuni and T. suis and more specifically by assessing the secretion of lL-8, TNF-a, and IL-l-B from cultured C. jejuni-infected intestinal pig epithelial cells (IPEC-1 cells), which served as a model system for undifferentiated, crypt-like cells. Additionally, we evaluated the effects of these secreted cytokines on bacterial-host cell interaction and intestinal mucosal integrity. This dissertation project addressed three specific aims. The first specific aim was to evaluate secreted IL-8, IL-l-B, TNF-a, IL-4, IL-6, and IL-10 expression in the feces of pigs after single or dual challenge infections with C. jejuni and T. suis. Two experimental designs were used to test this specific aim: a short-term challenge experiment (2 days) and a long-term challenge experiment (23 days). In the short-term challenge experiments we assessed the acute pro-inflammatory response to C. jejuni and T. suis infection. In these experiments, conventionally, weaned piglets were assigned to separate treatment groups and were simultaneously infected with 2 x 108 cfu C. jejuni and/or 2500 T. suis embryonated eggs or 2 x 108 cfu E. coli DHS-a. Fecal samples were taken prior to inoculation (day 0), and on days 1 and 2 post-inoculation. The fecal samples were prepared for analysis, and the supernatants were used in an ELISA to measure pro- and anti-inflammatory cytokine responses following concurrent infection. In the long-term challenge we assessed the pro-inflammatory cytokine response to C. jejuni in the presence of adult stage T. suis larvae. The pigs were assigned to separate treatment groups. One group of pigs was orally inoculated with 2500 T. suis embryonated eggs, and 21 days post-T. suis inoculation, these pigs were orally inoculated with 2 X 108 cfu C. jejuni. The other assigned groups were singly inoculated with 2500 T. suis embryonated eggs, 2 x 108 cfu c. jejuni, or 2 x 108 cfir E. coli DHS-a. Fecal 213 samples were taken prior to inoculation (day 0), and on days 21, 22, and 23 post- inoculation. The fecal samples were prepared for analysis, and the supernatants were used in an ELISA to measure pro- and anti-inflammatory cytokine expression during concurrent infection. Also, on day 23 of the long-term challenge experiments, tissue samples were taken from the intestine, and cytokine mRN A expression was measured using real time PCR. The second specific aim was to measure secreted IL-8, TNF-a, and lL-l-B from cultured swine intestinal epithelial cells (IPEC-l cells) following C. jejuni infection. In these in vitro experiments, IPEC-l cells were cultured in an undifferentiated state to mimic intestinal crypt cells and infected with C. jejuni 33292. Culture supernatants were removed at 0, 8, 24, and 48 hours after infection and analyzed for IL-8, TNF-a, and IL-l-B protein expression by ELISA. The third specific aim was to determine whether pro-inflammatory cytokines (specifically, IL-8, TNF-a, and IL-l-B) play a significant role in C. jejuni adherence, invasion, and/or the transepithelial electrical resistance (TER) with persistent exposure. In the adherence and invasion assays, IPEC-l cells were cultured in an undifferentiated state to mimic intestinal crypt cells and treated with various concentrations of recombinant swine IL-8, TNF-a, and IL-l-B for 5 hours. Recombinant swine IL-8, TNF-a, and IL—l-B supernatants were removed, and the pretreated IPEC-1 cells were infected with C. jejuni for 1 hour to allow bacteria to adhere to the cells or 3 hours to allow bacteria to invade the cells. After 1 hour, the adhered (cell-associated) bacteria were enumerated by serial dilution plating. After 3 hours, the gentamycin assay was performed, and intracellular C. jejuni were enumerated by serial dilution plating. Also, IPEC-1 cells were pretreated with recombinant swine IL-8, TNF- a, or IL-l-B to determine if cytokine exposure affects the transepithelial electrical 214 resistance (TER) of these cells. Undifferentiated IPEC-1 cells were grown to confluency, and the TER was measured prior to treating these cells with recombinant swine IL-8, TNF-a, or IL-l-B. Subsequently the IPEC-l cells were treated with recombinant swine IL-8, TNF-a, and IL-l-B for 5 hours, and then the TER was measured. To assess the effects of concurrent T. suis and C. jejuni infection on cytokine expression, we used our established swine infection model system set up as two experimental designs (short term; day O—day 2 and long term; day O-day 23) challenge experiments and measured pro- and anti-inflammatory cytokines in the feces and intestinal tissues of infected pigs. In viva results from the long-term challenge experiments (day O-day 23) showed that pigs that received T. suis alone showed a significant decreased IL-l-B in the feces by 23 days post-inoculation, and the dually- infected pigs showed moderately decreased IL-I-B expression. Also, on day 23, IL-I-[S was significantly decreased in the T. suis-infected pigs compared to IL-l-B expression in the C. jejuni-infected pigs. These and other findings suggest that T. suis likely predominates in driving the cytokine response by down regulating the expression of this pro-inflammatory cytokine during infection. This is not surprising because in most helminth infections a Th2 response is elicited with induced production of IL-4, IL-5, IL- 6, IL-9, IL-10, and IL-13 during infection (13, 38) This pattern of cytokine expression is characteristic of a Th2 response which has been shown to predominantly limit Th1 cytokine-induced inflammation (Th1 cytokines- Il-12, IFN-y, IL-8, TNF-a, and IL-1- B)(l3, 39) and result in warm expulsion. In the short-term challenge experiments, there were no significant changes in secreted lL-8, IL-l-B, TNF-a, IL-4, IL-6, or IL-10 expression. Generally, in pigs that are 215 naturally infected with T. suis, signs of pathology are not seen until 21 days post-T. suis infection, it is reasonable to believe that results from the long term challenge (23 days) would likely reflect the immune response of the natural infections more than the short- term challenge (2 days). This also suggests that immunity to T. suis ova or first stage larvae may be limited. In the swine model experiments, we determined the cytokine recovery rate for these fecal ELISAs to be 10—20%. This low recovery rate may have been due to protein degradation associated with freezer storage, the loss of protein during the fecal preparation due to the cytokines binding to the solid fecal matter, or protein degradation due to protease activity. Because of these possible drawbacks, the fecal ELISA data was only moderately useful and we relied instead on additional data obtained directly by measuring cytokine expression in gastrointestinal tract tissues in areas where the pathogens reside. In the long-term challenge experiments, after fecal sampling on day 23, the pigs were euthanized and tissue samples were taken hour the jejunum, proximal colon, and distal colon of our pigs. In the proximal colon of the two groups of pigs that received T. suis, Real Time PCR analysis showed increased expression of IL-13 mRNA (Th2 cytokine) and decreased IL-8 and IL-12 expression (Th1 cytokines). Compared to the uninfected control pigs, IL-13 expression was upregulated 3 to 4 fold, and IL-8 and IL-12 was decreased 2 to 3 fold and 2 to 4 fold, respectively. Additionally, expression of MCP- ], a chemokine, and IL-5, a Th2 cytokine, were significantly decreased 2 fold in the dual- infected pigs compared to the uninfected control pigs. Also, there were no significant 216 changes in cytokine mRNA expression in the pigs that received C. jejuni alone as compared to the uninfected controls. These data provide evidence that supports the hypothesis that T. suis infection can stimulate the production of a Th2 cytokine (IL-13) which can down-regulate pro- inflammatory cytokines (IL-8 and IL-12) in the proximal colon of pigs. In vitro, it has been shown that IL-13 can decrease IL-8 secretion from human intestinal epithelial cells(21). It has also been shown that IL-13 protects BALB/c mice from LPS-induced lethal endotoxemia, and decreased IL-12 production is correlated with this protective effect (27). In addition, it has been shown in our laboratory that differentiated intestinal pig epithelial cells stimulated with T. suis excretory-secretary products secrete IL-10 (31). In general, IL-10 production has been associated with helminth infections, and more specifically, it has been shown to be a critical component in the development of a polarized Th2-type immune response which has been correlated with host resistance and survival during Trichuris infection (37). Although IL-IO mRN A was not significantly upregulated in any of the tissue samples from the pigs that received T. suis, it is possible that peak expression of IL-10 occurred before tissue sampling on day 23 of the experiment, and by day 23, IL-IO levels had possibly gone back down to baseline levels. Altemately, it is possible that low constant levels of IL-10 could play a role in downregulating pro-inflammtory cytokine expression in order to maintain intestinal mucosal integrity. More to the point, previous data showed that significant production of IL-10 from cultured IPEC-1 cells following T. suis ESP exposure occurred at 24 hours post-infection (31). In vitro and in viva studies have demonstrated IL-10 and IL-13 inhibition of intestinal inflammatory responses such as Th1 -type cytokine production, 217 and this inhibition is believed to diminish the severity of the inflammatory responses thereby reducing clinical disease activity (8, 20, 21, 37). Taken together, the data presented here are a supportive extension of previous findings in our laboratory, and collectively these findings suggest that it is possible that the down regulation of IL-8, IL- 12 and possibly MCP-1 mRN A expression in response to T. suis infection may be due to IL-13 production and possibly IL-10 in response to helminth infection. Therefore, it is reasonable to suggest that T. suis plays a role in the down-regulation of pro-inflammatory cytokine expression, and that this response can alter host conditions which promote secondary bacterial infection. In the distal colon, significant changes in cytokine expression were observed only in the pigs that received T. suis alone. IL-l-B, IL-6, and IFN-y expression were increased 3 to 4 fold, and IL-13 expression was increased 3 fold compared to uninfected controls. lL-l-B, IL-6, and IFN-y production is characteristic of an inflammatory response. The pigs showing these responses had diarrhea and lesions in these sites. This observed inflammatory response distal to the sites of warm attachment (in the cecum and proximal colon) may be the result of secondary bacterial infection. Previous data have shown that T. suis infection plays a role in facilitating secondary bacterial invasion of many species of resident bacteria, including C. jejuni, C. cali, C. lari, Campylobacter spp. (unidentified species), and E. coli (24). In this study, in spite of our best efforts to support the organism in samples during transport, culture alone was not sensitive enough to detect or identify C. jejuni after infection. We found it necessary to use multiple diagnostic tests (TaqMan Real time PCR, RFLP analysis of Campylobacter 23S rRNA gene, 16S rRNA PCR analysis and 218 ELISA) to detect the inoculated strain of C. jejuni after infection. When C. jejuni was detected by TaqMan analysis in the distal colon of l of 6 pigs that received T. suis-only in screens of pigs post inoculation, we realized the possibility that prior infection with this organism or with other Campylobacter spp., or other epsilon proteobacteria may have occurred in these pigs. Suspicions of a contaminating bacterium were firrther aroused when RFLP analysis also showed positive results for C. coli and a Campylobacter spp. that gave a unique banding pattern that was not consistent with C. jejuni, C. lari, C. coli, or C. upsaliensis in jejunum, proximal and distal colon samples from most of the pigs in the treatment groups. ELISA results for a wide range of these bacteria showed that the pigs were most reactive to Helicobacter spp. that may have caused problems in both serological and molecular assays. Helicobacter is a closely related member of the epsilon proteobacteria group with similar outer membrane proteins. Finally, we tested fecal supernatants for the presence of other Campylobacter species and Helicobacter species using 16S rRNA PCR analysis. Here we found that ahnost all pigs in the short- and long- term challenge experiments tested negative for the presence of Campylobacter species pre- and post-infection. However, several pigs tested positive for the presence of Helicobacter species pre- and post-infection. Taken together these data provide the best evidence to suggest that the pigs were exposed to Helicobacter spp. prior to the experiment and that the cytokine responses measured may have been affected by this prior bacterial exposure. We must consider that these results are consistent with the possibility that the increased inflammatory cytokine response seen in the distal colon of the pigs infected with T. suis alone may actually represent host resistance to secondary bacterial infection. 219 It is also possible that the effects of ESP produced by 4th stage larvae that have traveled to the distal colon could contribute to increased expression of IL-13 and IL-6. It has been shown that T. suis-infected pigs have serum antibodies as early as 21 days post inoculation that recognize a 20kDa glycoprotein isolated from ESP collected fi'om adult worms cultured in vitro (17). It has been demonstrated in vitro that T. suis ESP treatment of differentiated IPEC-1 cells causes a dose dependent cytotoxic response and significantly decreased transepithelial electrical resistance (TER)(1, 2). Also, cultured differentiated and undifferentiated IPEC-1 cells secrete IL-6 and IL-10 following ESP exposure (31). Therefore, it is reasonable to consider that the cytotoxic effects of ESP on intestinal epithelial cells can stimulate an inflammatory cytokine response in the distal colon. We should consider that ESP may have direct effects on intestinal epithelial cells and could account in part for the increased cytokine production in viva at sites distal to warm attachment. Interestingly, we also found that there were no significant changes in cytokine expression in the distal colon of pigs that received C. jejuni alone or those that were dually-infected or in the proximal colon of pigs that received C. jejuni alone. In deed, we recognized through the use of multiple diagnostic tests that the pigs from our in viva experiments were likely exposed to Helicobacter spp., a wildtype C. jejuni, other Campylobacter spp., or epsilon proteobacteria prior to the experiments. The timing and source of acquisition is not known, but pigs were farrowed and reared with their mothers in a solid floor shower-in shower-out confinement facility and then were moved directly to the containment colony for these studies. Because our data provides evidence suggesting that these pigs were likely exposed to Helicobacter spp. (epsilon 220 proteobacteria-campylobacter-like organisms) at birth or shortly after birth on the MSU swine farm, it is reasonable to believe that the C. jejuni administered during our experiments served as a secondary bacterial challenge. A possible scenario is that these pigs acquired immunity after early exposure to epsilon proteobacteria such as Helicobacter, and the host cytokine and chemokine responses to the experimental C. jejuni inoculum are more representative of host resistance to secondary bacterial infection associated with protective immunity. Therefore, this scenario affected the interpretation of the results, but it did not negate our findings. Nevertheless, in the jejunum (a division of the small intestine), we found that pigs receiving C. jejuni alone showed significant changes in cytokine expression. The expression of MCP-I, TNF-a, IL-12p40, IFN-y, IL-4, and IL-10 was increased 3 to 5 fold compared to uninfected controls. In vitro, C. jejuni induces INT 407 cells to produce intracellular MCP-1, TNF-a, IFN-y, IL-4, and IL-10 (18), and Helicobacter pylori induces gastric epithelial cells to produce IL-8 and MCP-l (15, 28, 29, 34). Thus, the cytokine changes observed could be due to Campylobacter or Helicobacter interaction with the intestinal epithelial cells in the jejunum. Additionally, it is likely that the jejunal samples taken from our pigs contained some Peyers patches along with surrounding epithelium Peyers patches are lymphoid follicles in the small intestinal mucosa that serve as important components in local and systemic immune responses. Peyers patches are functionally similar to LGCs of the colon. It has been demonstrated in oral- challenged germfiee piglets that C. jejuni targets LGCs and induces robust cytokine responses (22, 23) (Jones, dissertation, 2005). It is possible that C. jejuni or Helicobacter stimulated the jejunal Peyers patch to produce a similar cytokine response, which likely 221 influenced our results. Clearly, the net effect of C. jejuni exposure on the jejunum was elevation of proinflammatory cytokines. To distinguish the exact cell types fi‘om which this response arises, further work should be done to fractionate tissues in the jejunum prior to measuring cytokine responses. The data here suggest that in the absence of T. suis, C. jejuni or epsilon proteobacteria stimulate pro-inflammatory cytokine responses in the jejunum. Furthermore, in the gastrointestinal tract of chickens rechallenged with Salmonella enterica Serovar Typhimurium, it has been shown that significant changes in proinflammatory cytokine and chemokine (IL-6 and MIP family chemokine) expression occur in the ileum and cecal tonsils, which are also segments of the small intestine (40). Additionally, in the jejunum of dually-infected pigs, GM-CSF was down-regulated 5 fold compared to uninfected controls. The pattern of reactivity in the jejunum over the five groups of pigs suggests that T. suis is likely contributing to the down-regulation of pro- inflammatory cytokine expression in the jejunum which is similar to the immunomodulatory effects that occurred in the proximal colon. In addition, we observed that clinical diarrhea was a prominent sign of disease throughout the experiments. Diarrhea was observed in pigs that received C. jejuni only, T. suis only or bath. However, more frequent and more severe diarrhea was observed in the dually infected pigs and the T. suis only-infected pigs compared to the other treatment groups, thereby implying that T. suis is mainly responsible for causing the diarrhea. Additionally, diarrhea was mostly observed in the long-term challenge experiment where T richuris infection had progressed for 21 days. As has been reported frequently in literature reports on T richuris-induced disease, diarrhea usually begins around 21 days post-infection and is commonly called the “21 day scours” (9). In both experiments, 222 some of the C. jejuni-infected pigs had mild diarrhea which is commonly seen in campylobacterosis (7). It has also been reported by Mansfield et al. in a previous C ampylabacter swine disease model experiment that some of the C. jejuni-infected pigs showed clinical signs of mild diarrhea at a less degree of severity than the dually-infected or T. suis only-infected pigs (23). The uninfected pigs and E. coli DHS-u infected pigs showed no signs of diarrhea. The observation of severe diarrhea in pigs infected dually or with T. suis alone has provided some evidence to suggest that T. suis plays a significant role in diarrhea] disease. Furthermore, C. jejuni invasion or stimulation of intestinal epithelial cells has been shown to induce pro-inflammatory cytokine production (16, 18). In viva, C. jejuni invade the lymphoglandular complex (LGCs) entrapped crypts and the follicle-associated epithelium of the LGCs following a primary challenge infection (23)(Jones, dissertation, 2005). A robust pro-inflammatory cytokine response is expressed in the LGCs upon C. jejuni infection (Jones, dissertation, 2005). In viva and in vitro studies have shown that C. jejuni interacts with enterocytes which serve as the first line of defense during bacterial infection (16, 19). My second specific aim was to assess IL-8, IL-l-B and TNF- a production from undifferentiated, crypt-like intestinal pig epithelial cells (IPEC-1 cells) following C. jejuni infection. This primary neonatal cell line was chosen because it complemented our swine model system The cells were grown to form confluent monolayers and infected with C. jejuni for 0, 8, 24, and 48 hours. The supernatants were collected at each time point and analyzed using ELISA. TNF-a was secreted from the IPEC-l cells by 8 hours post-inoculation, and continued production was measured through 48 hours post-inoculation. Likewise, IL-l-B was secreted by the IPEC-1 cells by 223 24 hours post-inoculation. We found that IL-8 was constitutively secreted by the IPEC-1 cells, and was not induced by C. jejuni infection. The data presented here suggest that the induced production of TNF-a and IL-l-B and the constitutive production of IL-8 fiom intestinal epithelial cells are indicative of an acute innate host immune response. It is likely that this inflammatory response can generate and/or modulate the immune response to benefit the host by contributing to the elimination of the bacteria. In the third specific aim, we assessed whether IL-8, TNF-a, or IL-l-B treatment of IPEC-l cells had an effect on C. jejuni invasion, adherence, and/or the transepithelial resistance (TER). IPEC-1 cells were grown on transwell inserts for 2 days to form confluent monolayers. The TER of the confluent monolayer was measured prior to treatment. Then, either IL-8, TNF-a, or IL-l-B were applied to the confluent monolayers and incubated for 5 hours, and the TER was measured again. The data presented fi‘om this work, showed that neither lL-8 nor IL-l-B have a direct effect on C. jejuni invasion, adherence, and/or the TER of IPEC-l cells. Also, treatment of IPEC-1 cells with TNF-a did not significantly affect C. jejuni adherence or changes in the TER of the IECs. This finding suggests that these cytokines do not act in an autocrine-like manner to specifically affect C. jejuni adherence, invasion, and/or the TER of IPEC-1 cells. Even though there was not a direct effect on C. jejuni adherence to or invasion of IPEC-l cells (in response to IL-8 or IL-I-B pretreatment) or a direct effect could not be determined following TNF-a pretreatment in vitro, it is still possible that IL-8, TNF-a, or IL-l-B may affect epithelial function in a paracrine-like or another autocrine-like manner during C. jejuni infection in viva. For instance, it has been demonstrated that cytokines can facilitate enterocyte proliferation, maturation, MHC class II expression, cytokine receptor 224 expression or apoptosis in healthy and diseased states (14, 30, 35). In addition, TNF-a and TGF-a have been shown to stimulate immature crypt cell proliferation, and TGF—B and INF-y can inhibit mitotic activity (36). IFN-y can alter immature crypt epithelial cell turnover and upregulate MHC class II expression (35). Therefore, alterations of cytokine concentrations as a consequence of bacterial exposure and intestinal inflammation may contribute to serious pathologic consequences. Further elucidation of the effects of distinct cytokines on epithelial cell growth, phenotype, firnction and bacterial-enterocyte interactions would be of importance in the understanding of cytokine mediated regulation of mucosal immune responses associated with C. jejuni infection. 225 Future Directions The results fi'om this dissertation project opened other avenues for further research that would enhance understanding of several aspects of this dual infection phenomenon. In chapter 2 we orally infected immunocompetent, conventionally reared pigs that had a complete intestinal microbial population with C. jejuni, T. suis, or both and analyzed fecal cytokine expression and cytokine expression in the jejunum, proximal colon, and distal colon. In this study, we found that our pigs were probably exposed to C. jejuni, other Campylobacter spp. or epsilon proteobacteria prior to the experiments, and this finding significantly impacted the interpretation of our current results. Therefore, it would be imperative to use gnotobiotic pigs for firture studies in order to definitively characterize and gain a better understanding of in viva cytokine profiles and kinetics during concurrent C. jejuni and T. suis infection. Further analysis of cytokine expression using in situ hybridization and cytokine antibody arrays would allow us to determine if there is a correlation between cytokine mRNA expressed in intestinal tissues and secreted cytokine (protein) expression, respectively. During the concurrent infection, analysis of cytokine expression at different times throughout the course of a long term challenge experiment (days 0-45) would allow us to examine acute and long term inflammatory responses (beginning at 24 hours postinfection and continuing throughout the length of the experiment) to C. jejuni infection and would also allow us to examine changes in cytokine expression while T. suis developmental changes were occurring. Extending the evaluation time out to 41 or 45 days would allow a more thorough assessment of cytokine responses throughout the entire prepatent period of T. suis luminal development. In 226 addition, we would examine pathologic changes to correlate lesion progression with cytokine changes. This would entail having a large number of infected pigs so that a subset could be sacrificed at intervals after infection. In general, T. suis infection triggers a Th2 type response involving the cytokine production of IL-4, IL-10, and IL-13 which plays a role in warm expulsion fi'om the host(13, 38). This Th2-type response can down-regulate ThI-type innate host immune responses (e. g. IL-l-fi, TNF—a, INF-y, IL-8, and IL-12) which generally cause bacteria elimination from the host (13, 38, 39). In chapter 2 we discussed our results that suggest that T. suis likely mediates lL-8 and IL-12 down-regulation by producing IL-13 in the proximal colon of pigs, in viva. Even though we did not measure IL-4 or IL-10 mRN A expression in any tissues from the pigs in the in viva experiments, it would be useful to determine the possible roles of these cytokines in vitro. In previously reported data from our laboratory, recombinant swine IL-4 has been shown to weaken tight junctions between IPEC-1 cells, and this could allow bacteria to breach the epithelial barrier and gain access to the basolateral surface of cells. Undifferentiated and differentiated IPEC-1 cells could be treated with IL-4, IL-10 and/or IL-13 in a dose response experiment followed by infecting these cytokine pretreated IPEC-1 cells with C. jejuni. The supernatants of these C. jejuni-infected cells could be used in ELISA to measure lL-8, TNF-a, IL-l-B, IFN-y, and IL-12 production. This experiment could determine if Th2- type cytokines (IL-4, IL-10, and IL-l3) effect Thl-type cytokine production (IL-I-B, TNF-a, and IL-8) from C. jejuni-infected swine intestinal epithelial cells, in vitro. It would also be interesting to challenge these cells with multiple strains of C. jejuni from 227 patients to see if particular gene content or gene expression correlates with enhanced host response. In chapter 3 we measured secreted IL-8, TNF-a, and IL-l-B from undifferentiated crypt-like swine intestinal epithelial cells (IPEC-l cells) following C. jejuni infection. Previous reports from our laboratory have shown that C. jejuni specifically interact with colonic crypts and are associated with mucus near goblet cells (23). It has also been demonstrated that intestinal epithelial cells can be stimulated by C. jejuni to produce proinflammatory cytokines and chemokines (16, 18). We measured secreted IL-l-a and TNF-a from C. jejuni-infected IPEC-1 cells, but only showed that IPEC-1 cells constitutively secrete IL-8. This raises the question as to why IL-8 is not induced by C. jejuni infection of IPEC-1 cells. One way to address this question could be to determine if primary intestinal pig epithelial cells (IPEC-1 cells) lack or have decreased expression of Toll-like receptor 4 and MD-2 (accessory molecule) on the cell surface using PCR analysis or flow cytometry. It has been demonstrated that IL-8 production is associated with TLR4 receptor signaling via LPS stimulation and NFKB activation (3, 4, 10, 12, 14). Once it has been determined whether IPEC-l cells lack or have decreased TLR4 and MD-2 on the cell surface, we could use western blot analysis of intracellular proteins to study the related intracellular signaling pathways of genes encoding proteins involved in inflammation. More specifically, we could evaluate the interaction of TLR4 and MD-2 binding to C. jejuni LPS, and determine if this interaction stimulates the NFKB signaling pathway to induce IL-8 gene expression (3, 4). Also, in chapter 3, our data showed that IL-8, TNF-u, and IL—l-B had no direct effect on C. jejuni interactions (specifically adherence, invasion, or changes in IPEC-I 228 TER) with the swine intestinal epithelial cells, but they could possibly have other autocrine- or paracrine-like effects which could play a role in host inflammatory immune responses associated with C. jejuni infection. For example, it has been shown that TNF-a and IL-l-B can stimulate intestinal epithelial cells to produce C3, a complement component which is associated with host acute-phase inflammatory responses (14, 25, 26, 30, 35). Therefore, it would be interesting know if intestinal pig epithelial cells (IPEC-1 cells) could produce C3. It is known that IL-l-B is an initiator of inflammation and a regulator of immune responses, and it induces acute phase protein responses, such as the production of the acute phase protein complement C3. Undifferentiated and differentiated IPEC-1 cells could be pretreated with IL—l-B in a dose response experiment. Confluent monolayers could be exposed to the different concentrations of IL-l-B. C3 levels could be measured in the supernatants by ELISA, and C3 mRNA levels would be determined by Northern blot analysis. This experiment would determine if IL-l-B acts in an autocrine-like manner to stimulate swine intestinal epithelial cells to produce an acute phase protein, complement C3. In a secondary experiment, the IPEC-1 cells could be pretreated with anti-IL-l-B antibody or IL-lRa (IL-1 receptor antagonist) to reverse C3 production, if any. In summary, we have generated data from this dissertation project that has provided some evidence and understanding of the research defining host immune responses to concurrent T. suis and C. jejuni infections. We demonstrated in viva that T. suis plays a major contributing role in mediating changes in cytokine expression during concurrent T. suis and C. jejuni infection. Using our swine infectious disease model system, we began to define host local immune responses expressed during this concurrent 229 infection. In vitro, we showed that undifferentiated, crypt-like intestinal pig cells (IPEC- 1) can serve as a suitable in vitro model for studying C. jejuni-enterocyte interactions and host enteric immune responses elicited by C. jejuni during infection. We discovered that C. jejuni can stimulate intestinal pig epithelial cells (IPEC-l cells) to produce IL-I-B and TNF-a, and we found that these intestinal pig epithelial cells constitutively secrete IL-8. Hence, these cytokine responses are indicative of an innate inflammatory immune response that may be responsible in some part for eliminating bacteria from the host in viva. In addition, we found that proinflammatory cytokines, such as IL-8, IL- l-B, or TNF-a, do not appear to directly effect C. jejuni interactions with host cells (particularly adherence, invasion, or stimulating changes in the transepithelial electrical resistance (TER) of IPEC-1 cells). Collectively, these data have provided some evidence for T. suis contributing significantly to cytokine dysregulation seen during concurrent infection, and these immunomodulatory effects likely promote C. jejuni infection of the intestinal mucosal epithelium by limiting the natural host resistance to bacterial infection. 230 10. 11. REFERENCES Abner, S. R., D. E. Hill, J. R. Turner, E. D. Black, P. Bartlett, J. F. Urban, and L. S. Mansfield. 2002. Response of intestinal epithelial cells to Trichuris suis excretory-secretory products and the influence on Campylobacter jejuni invasion under in vitro conditions. J Parasit0188: 738- 745. . Abner, S. R., G. Parthasarathy, D. E. Hill, and L. S. Mansfield. 2001. Trichuris suis: detection of antibacterial activity in excretory-secretory products fi'om adults. Exp Parasitol 99:26-36. Abreu, M. T., E. T. Arnold, L. S. Thomas, R. Gonsky, Y. Zhou, B. Hu, and M. Arditi. 2002. TLR4 and MD-2 expression is regulated by immune-mediated signals in human intestinal epithelial cells. J Biol Chem 277 :20431-20437. Abreu, M. T., P. Vora, E. Faure, L. S. Thomas, E. T. Arnold, and M. Arditi. 2001. Decreased expression of Toll-like receptor-4 and MD-2 correlates with intestinal epithelial cell protection against dysregulated proinflammatory gene expression in response to bacterial lipopolysaccharide. J Immunol 167:1609- 1616. ' Allos, B. M. 2001. Campylobacter jejuni Infections: update on emerging issues and trends. Clin Infect Dis 32:1201-1206. Altekruse, S. F., N. J. Stern, P. I. Fields, and D. L. Swerdlow. 1999. Campylobacter jejuni--an emerging foodbome pathogen. Emerg Infect Dis 5: 28- 35. ' Babakhani, F. K., G. A. Bradley, and L. A. Joens. 1993. Newborn piglet model for campylobacteriosis. Infect Immun 61 :3466-3475. Berg, D. J., N. Davidson, R. Kuhn, W. Muller, S. Menon, G. Holland, L. Thompson-Snipes, M. W. Leach, and D. Rennick. 1996. Enterocolitis and colon cancer in interleukin-IO-defiCient mice are associated with aberrant cytokine production and CD4(+) THl-like responses. .1 Clin Invest 98:1010—1020. Bliss, D. 1991. Swine Parasites: A new look at an old problem Hoechst-Roussel - Agri-Vet Co. Bruewer, M., A. Luegering, T. Kucharzik, c. A. Parkos, J. L. Madara, A. M. Hopkins, and A. Nusrat. 2003. Proinflammatory cytokines disrupt epithelial barrier firnction by apoptosis-independent mechanisms. J Immunol 171: 6164- 6172. Coker, A. o., n o. Isokpehi,'B. N. Thomas, K. o. Amisu, and c. L. Obi. 2002. Human campylobacteriosis in developing countries. Emerg Infect Dis 8:237-244. 231 13. 14. 15. 16. 17. 18. 19. 20. 21. Eckmann, L., H. C. Jung, C. Schurer-Maly, A. Panja, E. Morzycka- Wroblewska, and M. F. Kagnoff. 1993. Differential cytokine expression by human intestinal epithelial cell lines: regulated expression of interleukin 8. Gastroenterology 105:1689-1697. F inkelman, F. D., T. Shea-Donahue, J. Goldhill, C. A. Sullivan, S. C. Morris, K. B. Madden, W. C. Gause, and J. F. Urban, Jr. 1997. Cytokine regulation of host defense against parasitic gastrointestinal nematodes: lessons from studies with rodent models. Annu Rev Immunol 15:505-533. Fish, S. M., R. Proujansky, and W. W. Reenstra. 1999. Synergistic effects of interferon gamma and tumour necrosis factor alpha on T84 cell function. Gut 45: 191-198. F utagami, S., T. Hiratsuka, A. Tatsuguchi, K. Suzuki, M. Kusnnoki, Y. Shinji, K. Shinoki, T. Iizumi, T. Akamatsu, H. Nishigaki, K. Wada, K. Miyake, K. Gudis, T. Tsukui, and C. Sakamoto. 2003. Monocyte chemoattractant protein 1 (MCP-l) released from Helicobacter pylori stimulated gastric epithelial cells induces cyclooxygenase 2 expression and activation in T cells. Gut 52: 1257-1264. Hickey, T. E., S. Baqar, A. L. Bourgeois, C. P. Ewing, and P. Guerry. 1999. Campylobacter jejuni-stimulated secretion of interleukin-8 by INT407 cells. Infect Imrnun 67:88-93. Hill, D. E., R. D. Romanowski, and J. F. Urban, Jr. 1997. A Trichuris specific diagnostic antigen from culture fluids of Trichuris suis adult worms. Vet Parasitol 68:91-102. Hu, L., and T. E. Hickey. 2005. Campylobacter jejuni induces secretion of proinflammatory chemokines from human intestinal epithelial cells. Infect Immun 73:4437-4440. Jung, H. C., L. Eckmann, S. K. Yang, A. Panja, J. F ierer, E. Morzycka- Wroblewska, and M. F. Kagnoff. 1995. A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion. J Clin Invest 95:55-65. Lane, J. S., K. E. Todd, M. P. Lewis, B. Gloor, S. W. Ashley, H. A. Reber, D. W. McFadden, and C. F. Chandler. 1997. Interleukin-10 reduces the systemic inflammatory response in a murine model of intestinal ischemia/reperfirsion. Surgery 122:288-294. Lugering, N., T. Kucharzik, M. Kraft, G. Winde, C. Sorg, R. Stall, and W. Domschke. 1999. Interleukin (IL)-l3 and IL-4 are potent inhibitors of IL-8 secretion by human intestinal epithelial cells. Dig Dis Sci 44:649-655. 232 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. Mansfield, L. S., and D. T. Gauthier. 2004. Lymphoglandular complexes are important colonic sites for immunoglobulin A induction against Campylobacter jejuni in a swine disease model. Comp Med 54:514-523. Mansfield, L. S., D. T. Gauthier, S. R. Abner, K. M. Jones, S. R. Wilder, and J. F. Urban. 2003. Enhancement of disease and pathology by synergy of Trichuris suis and Campylobacter jejuni in the colon of immunologically naive swine. Am J Trop Med Hyg 68:70-80. Mansfield, L. S., and J. F. Urban, Jr. 1996. The pathogenesis of necrotic proliferative colitis in swine is linked to whipworm induced suppression of mucosal immunity to resident bacteria. Vet Immunol Immunopathol 50:1-17. Moon, M. R., A. A. Parikh, T. A. Pritts, C. Kane, J. E. Fischer, A. L. Salzman, and P. O. Hasselgren. 2000. Interleukin-lbeta induces complement component C3 and IL-6 production at the basolateral and apical membranes in a human intestinal epithelial cell line. Shock 13:374-378. Moon, R., A. A. Parikh, C. Szabo, J. E. Fischer, A. L. Salzman, and P. O. Hasselgren. 1997. Complement C3 production in human intestinal epithelial cells is regulated by interleukin lbeta and tumor necrosis factor alpha. Arch Surg 132:1289-1293. Muchamuel, T., S. Menon, P. Pisacane, M. C. Howard, and D. A. Cockayne. 1997. IL-13 protects mice fi‘om lipopolysaccharide-induced lethal endotoxemia: correlation with down-modulation of TNF-alpha, IFN-gamma, and IL-12 production. J Immunol 158:2898-2903. N akano, Y., T. Kasahara, N. Mukaida, Y. C. Ko, M. Nakano, and K. Matsushima. 1994. Protection against lethal bacterial infection in mice by monocyte-chemotactic and -activating factor. Infect Immun 62:377-383. Nozawa, Y., K. Nishihara, R. M. Peek, M. Nakano, T. Uji, H. Ajioka, N. Matsuura, and H. Miyake. 2002. Identification of a signaling cascade for interleukin-8 production by Helicobacter pylori in human gastric epithelial cells. Biochem Pharmacol 64:21-30. Panja, A., S. Goldberg, L. Eckmann, P. Krishen, and L. Mayer. 1998. The regulation and firnctional consequence of proinflammatory cytokine binding on human intestinal epithelial cells. J Immunol 161:3675—3684. Parthasarathy, C., and L. S. Mansfield. 2005. Trichuris suis excretory secretory products (ESP) elicit interleukin-6 (IL-6) and IL-10 secretion from intestinal epithelial cells (IPEC-1). Vet Parasitol 131:317-324. 233 32. 33. 34. 35. 36. 37. 38. 39. 40. Pedersen, S., and I. Saeed. 2000. Experimental infection of pigs with three dose levels of Trichuris suis. Parasite 7 :275-281. Pedersen, S., and I. Saeed. 2002. Host age influence on the intensity of experimental Trichuris suis infection in pigs. Parasite 9:75-79. Peek, R. M., Jr. 2001. IV. Helicobacter pylori strain-specific activation of signal transduction cascades related to gastric inflammation. Am J Physiol Gastrointest Liver Physiol 280:G525-530. Ruemmele, F. M., C. Gurbindo, A. M. Mansour, R. Marchand, E. Levy, and E. G. Seidman. 1998. Effects of interferon gamma on growth, apoptosis, and MHC class II expression of immature rat intestinal crypt (IEC-6) cells. J Cell Physiol 176:120-126. Ruemmele, F. M., and E. G. Seidman. 1998. Cytokine--intestinal epithelial cell interactions: implications for immune mediated bowel disorders. Zhonghua Min Guo Xiao Er Ke Yi Xue Hui Za Zhi 39:1-8. Schopf, L. R., K. F. Hoffmann, A. W. Cheever, J. F. Urban, Jr., and T. A. Wynn. 2002. IL-10 is critical for host resistance and survival during gastrointestinal helminth infection. J Immunol 168:2383-2392. Urban, J. F., Jr., K. B. Madden, A. Svetic, A. Cheever, P. P. Trotta, W. C. Gause, I. M. Katona, and F. D. Finkelman. 1992. The importance of Th2 cytokines in protective immunity to nematodes. Immunol Rev 127:205-220. Urban, J. F ., Jr.; Mansfield, LS; Shea-Donahue, T; W. C.Gause, and F inkelman, F D.» 1999. Mechanisms of Intestinal Immunity to Nematode Parasites and the consequences to invasion by opportunistic bacteria. Seminar Monograph 11: Immunomodulation of the gastrointestinal mucosa. Withanage, G. S., P. Wigley, P. Kaiser, P. Mastroeni, H. Brooks, C. Powers, R. Beal, P. Barrow, D. Maskell, and I. McConnell. 2005. Cytokine and chemokine responses associated with clearance of a primary Salmonella enterica serovar Typhimurium infection in the chicken and in protective immunity to rechallenge. Infect Immun 73:5173-5182. 234 HI u131lttljflljlljljljljjljll‘li|