tug-mm; 71‘ oliglq‘v‘v gas-um msnv-u-uyn-vmgn ‘!"i. «y‘- 2’ m. -u.., n u m. m .u c1-.vglr .cm .u. . . ." nn “.9."- .x m“ UBRARY Michigan State U 3 -3 awn:*\l Ul IIVUI any This is to certify that the dissertation entitled DESIGN AND EVALUATION OF IMMUNOGENIC Escherichia coli HEAT- STABLE ENTEROTOXIN (STa) AND'CHARACTERIZATION OF THE IMMUNE RESPONSE IN LABORATORY ANIMALS presented by Nasr-Eldin Mohamed M. Aref has been accepted towards fulfillment of the requirements for the PhD. degree in Lane Animal Clinical Sciences W/uwg/ Major Professor’s Signature 03’.» 07- 0g Date MSU is an affinnative-action, equal-opportunity employer ..-—-—-—-—.—.—.-o--o-o---.—--—-—-.—-—._-—-n—.— .— .uauo—a-o-o-I-o-n-u-o-I-u-u-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 K1/Prolecc8Pres/CIRCIDateDue.nndd DESIGN AND EVALUATION OF IMMUNOGENIC ESCHERICHIA COLI HEAT- STABLE ENTEROTOXIN (STa) AND CHARACTERIZATION OF THE IMMUNE RESPONSE IN LABORATORY ANIMALS BY Nasr—Eldin Mohamed M. Aref A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Large Animal Clinical Sciences 2008 ABSTRACT DESIGN AND EVALUATION OF IMMUNOGENIC Escherichia coli HEAT- STABLE ENTEROTOXIN (STa) AND CHARACTERIZATION OF THE IMMUNE RESPONSE IN LABORATORY ANIMALS By Nasr-Eldin Mohamed M. Aref Strains of Enterotoxigenic Escherichia coli (ETEC) that produce heat-stable enterotox'm (STa) are an important cause of diarrheal disease in humans and animals. They are responsible for a significant proportion of diarrheal cases among infants, neonatal animals, and travelers going from non-endemic to endemic areas. The development of effective strategies to reduce the incidence and severity of ETEC- caused diarrhea has been hampered by the lack of an effective vaccine or immunotherapeutic agents against this enteric pathogen. The lack of an effective vaccine against the ETEC is largely due to the diverse antigenic structure of the ETEC strains and the poor immunogenicity of the STa enterotoxin, which is the predominant enterotoxin in 75% of ETEC strains and is the immediate mediator of ETEC-caused diarrhea. Attempts to induce an appropriate immune response against the STa involve the design of immunogenic STa-carrier conjugates. However, based on the protocols reported in previous studies, only low titers of STa-antisera have been obtained. In this study, we used highly purified E. coli STa to produce STa-carrier conjugates through covalently cross-linking the STa to a modified bovine serum albumin (BSA) carrier protein. Four different conjugation protocols were evaluated. The most effective STa-conjugate was selected based on its retention of STa biological activity, conjugation efficiency, and stability. The selected STa-BSA conjugate was used to immunize rabbits for antibody production. This STa-conjugate induced a high immune response against the native STa. The harvested rabbit sera showed a neutralization capacity of 3 x 104 STa mouse units/ml serum using a suckling mouse assay, and a specific binding titer of 10'6 using indirect ELISA. These levels of STa neutralization and binding capacity are higher than what has been reported in previous studies. This study demonstrated the feasibility of designing an effective immunogenic ETEC-STa conjugate that can be used for the development of immunotherapeutic reagents and /or a potential vaccine against the ETEC-STa. Copyright by Nasr—Eldin Mohamed M. Aref 2008 DEDICATION To my almighty and beloved ALLAH who has been my shield, my strength, and my very present help in time of need. Thank you my LORD for helping me throughout this dissertation and for giving me a hope beyond hopes. ACKNOWLEDGMENTS I am indebted to my guidance committee members, their contribution made this work possible. A special thanks to my major advisor, Dr. A. Mahdi Saeed, for his continuous intellectual advisement, encouragement, and support. He is a great mentor, colleague and friend. No words can express my sincere appreciation to my Ph.D. guidance committee members, Drs. Patricia Ganey, Robert Roth, Ronald Erskine and Daniel Grooms, for their great mentorship. Their suggestions and constructive feedback while conducting various parts of my research and writing my dissertation were vital for the completion of my project. I gratefully acknowledge Dr. Joseph Leykam and Eric Lund, Research Technology Support Facility at Michigan State University for their help on mass spectroscopy and amino acid sequencing. I would like to thank the staff in the Containment Facility, Michigan State University for their help. In particular, Cathy Tyler and Randy Shoemaker, for their assistance in taking care of animals used in this research. Many thanks to our laboratory staff, Kristin Evon and Nicole Crisp, our summer interns, Jessica Malay and Amanda Audo, and our undergraduate students, Laya Kevan, Alyssa DiFilippo, Kyle Korolowicz, Narges Rahman, Krystal Snodgrass, Tressa Fountain, Cassy Nguyen, Josephine Wee, Ashley Walters, Amy LaRose, Symone Coleman, I Rin, Diane Sinawi and Hiu-Lam Lau, for their great assistance throughout this research. vi Many friends and colleagues at Michigan State University provided substantial advice, support and encouragement during this work, in particular, Drs. Seongbeom Cho, Mokhtar Arshad and Muhammad Younus. I would also like to thank the administrative staff at the College of Veterinary Medicine and National Food Safety Toxicology Center, Michigan State University, in particular, Faith Peterson, Jennifer Sysak, Susan Dies and Margaret Nicolas, for their support. My special gratitude and sincere appreciation goes to my wife and children for their continuous prayers, encouragement, support, patience and tolerance during this critical stage in my life. Special thanks are extended to Assiut University, Egyptian High Education Ministry, Egyptian Cultural Educational Bureau (Washington DC.) and Microbiological Research Unit-National Institute of Health (MRU-NIH-USA) for their financial support. vii TABLE OF CONTENTS LIST OF TABLES ............................................................................ xi LIST OF FIGURES ........................................................................... xii LIST OF ABBREVIATIONS .............................................................. xv CHAPTER I: Introduction Statement of the Issue and Rationale ................................................. 1 General Objectives ....................................................................... 3 Specific Aims ............................................................................. 4 Potential Benefit of the Study .......................................................... 4 References ................................................................................. 6 CHAPTER II: Review of Literature Magnitude and impact of diarrheal diseases ................................................ 8 General overview of Escherichia coli ....................................................... 9 Virulence Attributes of ETEC ................................................................. 9 Structure, Classification, and Antigenic Types of ETEC ................................. 11 Pilus or F imbrial Antigen (F antigen) ................................................ 11 Somatic (0) antigen ..................................................................... 13 Capsular (K) antigens ..................................................................... 14 Enterotoxins of Enterotoxigenic E. coli ................................................... 15 Heat-Stable Enterotoxins (STs) ............................................................... 15 Differences between STa and STb ........................................................... 16 Enterotoxigenic E. coli STa (STI) ........................................................... 17 The STa family of toxins ................................................................ 18 Structural determinants and biochemical properties of STa ........................ 19 Antigencity and antigenic determinants of E. coli STa ............................... 21 Structural-Function Relationship of E. coli STa ...................................... 22 Receptor of ETEC-STa: Guanylyl Cyclase C (GC-C) ................................. 23 Intestinal fluid and electrolyte homeostasis ............................................. 29 Intestinal absorptive mechanisms ...................................................... 29 Intestinal secretory mechanisms ........................................................ 30 Role of cyclic nucleotide in intestinal homeostasis ........................ ‘ .......... 3 O Pathogenesis of ETEC Diarrhea ........................................................... 31 Infection with ETEC ..................................................................... 31 Intestinal colonization ................................................................... 32 Elaboration of enterotoxin: Pathophysiology of STa action ....................... 33 Potential systemic effect of ETEC-STa diarrhea ..................................... 37 Current Approaches for Controlling ETEC-STa Diarrhea .......................... 38 References ...................................................................................... 4] viii CHAPTER III: PURIFCIATION AND CHARACTERIZATION OF E. coli HEAT-STABLE ENTEROTOXIN Introduction .............................................................................. 56 Materials and methods ............................................................... 57 Animals .............................................................................. 57 Reagents ............................................................................ 57 Verifying the ETEC K99+ Strain ............................................... 57 Bacteria] Strain ................................................................. 57 DNA Extraction (Template) by boiling lysis .............................. 58 Primer selection and preparation ............................................ 58 PCR program .................................................................. 58 PCR reaction .................................................................. 59 Agarose gel electrophoresis analysis of PCR product .................... 59 Purification and characterization of E. coli STa ....................................... 59 Seed culture and frozen stock preparation of ETEC ................................... 60 Batch medium (asparagine salt medium) and growth condition ..................... 60 Preparation of cell free filtrate ............................................................ 61 Amberlite XAD-2 Batch Adsorption Chromatography ............................... 61 Acetone Fractionation ..................................................................... 62 Reversed-Phase Batch Adsorption Chromatography .................................. 62 Preparative RP—HPLC ..................................................................... 63 STa assessment for biological activity ................................................... 63 Criteria for homogeneity of the purified STa ........................................... 64 Results and Discussion ....................................................................... 64 Conclusion ...................................................................................... 67 References ...................................................................................... 72 CHAPTER IV: DESIGN AND CHARACTERIZATION OF AN IMMUNOGENIC E. coli HEAT-STABLE ENTEROTOXIIN (STa) Introduction ................................................................................... 74 Background and principle of peptide-carrier conjugation ................................ 75 Cross-linking reagents .................................................................. 75 Carrier proteins ........................................................................... 76 Materials and methods ....................................................................... 78 Reagents ..................................................................................... 78 Chemical modification of bovine serum albumin................................... 79 Succinylation of BSA ............................................................... 79 Hyper-succinylation of BSA ...................................................... 80 Coupling of E. coli STa to modified BSA .......................................... 81 Protocol 1: DMF method. ......................................................... 81 Protocol 2: Imidazole method .................................................... 82 Protocol 3: Hyper-succinylation method ........................................ 83 Protocol 4: Conventional method ................................................. 83 Dialysis ..................................................................................... 84 Gel permeation chromatography ........................................................ 84 Size exclusion chromatography ......................................................... 85 ix Amino acid compositional analysis ................................................... 85 Matrix assisted laser desorption ionization-time of flight mass spectroscopy... 85 Protein Assay ............................................................................. 86 STa-carrier conjugate activity bioassay ............................................... 86 Results ........................................................................................... 86 Discussion ....................................................................................... 88 References ...................................................................................... 98 CAHPT ER V. ANTIBODY PRODUCTION AND CHARACTERIZATION THE IMMUNE RESPONSE AGAINST E. coli HEAT-STABLE ENTEROTOXIN Introduction .................................................................................... 1 03 Materials and methods ...................................................................... 104 Reagents and instruments ............................................................... 104 Animals .................................................................................... 105 Immunization procedures ............................................................... 105 Animal bleeding .......................................................................... 105 STa— Serum Neutralization Assay ...................................................... 106 Kinetics of rabbit immune response to E. coli heat-stable enterotoxin ............ 107 Antibody-capture ELISA for screening sera .................................... 107 Avidity ELISA ...................................................................... 108 Statistical analysis ............................................................................ 109 Results ........................................................................................... 109 Discussion ....................................................................................... 111 References ....................................................................................... 126 Summary and conclusions .................................................................. 129 LIST OF TABLES Table 1. Difference between E. coli Heat-Stable Enterotoxin A (STa) and Heat- Stable Enterotoxin B (STb) ................................................................... 16 TableIZ. The STa Family of Toxins .......................................................... 18 Table 3. PCR primers used to detect STa gene .............................................. 58 Table 4. PCR running conditions for detection of STa gene .............................. 58 Table 5. PCR reaction for detection of STa gene ........................................... 59 Table 6. Composition of optimal minimal medium for STa elaboration ................ 60 Table 7. Summary of the purification scheme of E. coli heat-stable enterotoxin per batch ............................................................................................... 71 Table 8. Summary of conjugation experiments: Evaluation of four different conjugation protocols ........................................................................... 92 Table 9. Amino acid compositional analysis of E. coli heat-stable enterotoxin peptide— BSA carrier molecule ................................................................. 93 Table 10. Approximate contribution of STa molecules to one molecule of modified BSA: calculation of the conjugation ratio .................................................... 93 Table 11. E. coli STa-specific serum antibody end titer: Mean OD i SD value of group 1, 2 and 3 rabbits after 24 weeks post immunization at various serum dilutions ........................................................................................................................ 1 24 Table 12. Summary of STa- ELISA binding and neutralization end titers of rabbit sera immunized with STa-suBSA conjugate after the primary immunization and during the boosting intervals .................................................................. 124 Table 13. Summary of the development of STa antibody affinity after multiple boosters with the STa conjugate: Five molar ammonium thiocyanate elution ......... 125 Table 14. Neutralization capacity of sera from animals immunized with several STa-immuogenes and the end titers of the STa-neutralizing antibodies as reported in the literature .................................................................................. 125 xi LIST OF FIGURES Figure 1: Structural determinants of Heat-Stable Enterotoxin of Enterotoxigenic E. coli .............................................................................................. 20 Figure 2. Intramolecular cysteine—disulfide linkage for STaP and STaH ................ 24 Figure 3. Domain structure of guanylyl cyclases ........................................... 26 Figure 4. Pathophysiology of E. coli heat-stable enterotoxin diarrhea. ................. 36 Figure 5. Agarose gel electrophoresis of PCR product analysis .......................... 68 Figure 6. Growth kinetic of ETEC on 36L batch ASM under different pH using Bellco bioreactor .................................................................................. 68 Figure 7. Revere-phase-high performance liquid chromatography elution profile of 60% HPLC-grade methanol- MCI gel -STa-rich fraction on preparative C8 Vaydac column .............................................................................................. 69 Figure 8. Elution profile of biologically active E. coli heat-stable enterotoxin peaks on analytic aquapore reverse-phase C 8 Perkin Elmer column ........................... 69 Figure 9. Matrix assisted laser desorption ionization-time of flight mass spectroscopy profile of E. coli heat-stable enterotoxin .................................... 70 Figure 10. Illustration showing the conjugate of bovine serum albumin carrier protein- E. coli STa peptide ................................................................... 79 Figure 11. The action of succinic anhydride upon the amino groups of a protein. . . 80 Figure 12. Size exclusion chromatography profile of native and succinylated BSA molecule ........................................................................................... 94 Figure 13. Matrix- assisted laser desorption ionization-time of flight mass spectroscopy profile of succinylated BSA molecule ....................................... 94 Figure 14. Matrix- assisted laser desorption ionization-time of flight mass spectroscopy profile of E. coli heat-stable enterotoxin peptide-succinylated BSA carrier conjugate. ................................................................................ 95 Figure 15. Matrix- assisted laser desorption ionization-time of flight mass spectroscopy profile of E. coli heat-stable enterotoxin peptide-hypersuccinylated BSA carrier conjugate ........................................................................... 96 xii Figure 16. Calculation of A molecular weight and conjugation ratio of the E. coli heat-stable enterotoxin peptide- BSA carrier conjugate .................................. 97 Figure 17. Illustration of antibody capture indirect ELISA protocol ..................... 114 Figure 18. E. coli STa-specific serum antibody neutralization bioassay: Group 1 rabbits ............................................................................................. 115 Figure 19. Neutralization capacity of E. coli STa-specific serum antibody ............. 115 Figure 20. E. coli STa-specific serum antibody neutralization bioassay: All rabbits ............................................................................................. 116 Figure 21. E. coli STa ELISA Optimization: Screening serum dilution for optimal E. coli STa-STa antibody interaction ........................................................ 117 Figure 22. E. coli STa -specific serum antibody: 104 serum dilution of group 1 rabbits ............................................................................................. 117 Figure 23. E. coli STa-specific serum antibody: 10'4 serum dilution of group 2 rabbits ..................................................... . ........................................ 118 Figure 24. E. coli STa-specific serum antibody: 104 serum dilution of group 3 rabbits .............................................................................................. 118 Figure 25. Mean OD value of group 1 rabbits after 20 weeks post immunization at various serum dilutions .......................................................................... 119 Figure 26. . Mean OD value of group 2 rabbits after 20 weeks post immunization at various serum dilutions .. ............................................................................................. 119 Figure 27. . Mean OD value of group 1 rabbits after 24 weeks post immunization at various serum dilutions ......................................................................... 120 Figure 28. End titer of the E. coli STa-specific serum antibody: 24 weeks post immunization from 8 rabblts 120 Figure 29. E. coli STa-specific serum antibody end titer: Mean OD value of group 1, 2 and 3 rabbits after 24 weeks post immunization at various serum dilutions ....... 121 Figure 30. Time-course evaluation of the E. coli STa-specific serum antibody avidity using ammonium thiocyanate dose response ....................................... 122 Figure 31. Five molar thiocyanate elution profile of E. coli STa-STa serum antibody complex: Mean OD of treated serum from group 1, 2 and 3 of rabbits .................. 123 xiii Figure 32. Avidity index E. coli STa-specific serum antibody of the group, 1, 2 and 3 of rabbits ....................................................................................... 123 xiv LIST OF ABBREVIATIONS ANPR = Atrial natriuretic peptide receptors BSA = Bovine serum albumin cAMP = Cyclic adenosine monophosphate 2CIATP = 2 choloradenosine trihosphate CFA = Colonization fimbriae of human ETEC strains CFT R = Cystic fibrosis transmembrane conductance regulator cGMP = Cyclic guanosine monophosphate CI' = Cholride ion CS = Coli surface antigen CT = Cholera toxin EAEC = Enteroaggressive E. coli EAST] = Enteroaggregative heat-stable enterotoxin ECD = Extracellular domain EDAC =1—ethyl-3[3-dimethylaminopropyl] carbodiimide hydrochloride ETEC = Enterotoxigenic E. coli GC-C = Guanylyl cyclase type C GTP = Guanosine triphosphate HCO3' = Bicarbonate ion HPLC = High performance liquid chromatography ICD = Intracellular domain K+ =Potassium ion LT = Heat-labile enterotoxin XV LT = Heat-labile toxin MU = Mouse unit MWCO = Molecular weight cutoff Na+ = Sodium ion NMR = Nuclear magnetic resonance PBS = Phosphate buffer solution PDE3 = phosphodiesterase PKC = Protein Kinase SMA = Suckling mouse assay STs = Heat-stable enterotoxins STb = Heat-stable enterotoxin B STa = Heat-stable enterotoxin isolated primarily from bovine STp = Heat-stable enterotoxin isolated primarily from porcine STh = Heat-stable enterotoxin isolated primarily from human SuBSA = Succinylated bovine serum albumin TD = Transmembrane domain TFA = Triflouroacetic acid Amino Acid Abbreviations A ALA Alanine CYS Cysteine ASP Aspartic Acid GLU Glutamic Acid '11P! U G PHE Phynylalanine xvi G GLY Glycine HIS Histidine I ILE Isoleucine “ LYS Lysine LEU Leucine MET Methionine zgt-x ASN Asparagine PRO Proline "U GLN Glutamine we ARG Arginine SER Serine U) T THR Threonine V VAL Valine W TRP Tryptophan Y TYR Tyrosine xvii CHAPTER I INTRODUCTION Statement of the Issue and Rationale Enterotoxigenic Escherichia coli (ETEC) is a major enteropathogen that causes potentially fatal diarrhea in both human and animal neonates (Moon and Bunn 1993, Tacket et al. 1994). It is also responsible for a large proportion of diarrheal disease among adult travelers (Tacket et a1. 1994). Therefore, strategies to reduce the incidence and severity of ETEC diarrhea have been considered an important public health priority (Tacket et a1. 1994). A large proportion of ETEC diarrhea is caused by heat-stable enterotoxin (STa), a small peptide (2kD), which is an important virulence determinant in enterotoxin-mediated diseases (Sears & Kaper 1996 and Giannella & Elizabeth 2003). Upon infection, the STa producing- ETEC adheres to the epithelium of the small intestine via one or more colonization factor antigens or pili surface proteins. Once established, ETEC elaborates heat-stable enterotoxin (STa), which acts on a specific intestinal membrane bound receptor, guanylyl cyclase C, initiating a cascade of altered metabolic pathways. This may result in secretory diarrhea among affected adults but can cause fatal dehydration in neonates. Methods for the treatment and control of ETEC diarrhea are still a matter of debate among veterinarians, livestock producers and in the animal industry in general. The use of sub-therapeutic doses of antibiotics may help protect animals from some, but not all, of these bacterial strains. Moreover, the use of antimicrobials at sub- therapeutic levels has been linked to the problem of emerging antibiotic resistance among several bacterial species, including ETEC strains. “en- While there are several reagents that are in use against ETEC diarrheal disease in animals, most of these reagents are based on surface structures of the ETE strains. However, the development of a broad-spectrum vaccine against ETEC remains elusive (Walker et al. 2007). Two major technical problems contribute to this deficiency. The first involves the production of immunogenic preparations of antigens with the ability to confer broad-spectrum protection against ETEC infections. The second is the challenge of achieving effective mucosal immunization (Walker et a1. 2007) due to the multiplicity, antigenic diversity, and high prevalence of unidentifiable forms of specific colonization antigens responsible for mucosal adherence (Deneke et a1. 1981, Levine et al. 1980, Thomas and Rowe 1982). Against this background, there is an urgent need to define a new common antigenic determinant that could provide broad protection against ETEC-STa—induced diarrhea. Saeed et a1. (1985) demonstrated that calf scour could be experimentally induced by a highly purified STa preparation, supporting the notion that ETEC STa is the immediate mediator of diarrhea in claves. Additionally, several studies have demonstrated a significant correlation between STa-producing ETEC strains and diarrhea, and that 75% of ETEC strains produce STa either alone or in combination with heat-labile enterotoxin (LT) (Wolf 1997). Thus, the inclusion of STa in colonization factor-based ETEC vaccines or the production of neutralizing STa- antibodies would potentially offer immune protection against ETEC—caused diarrhea. However, this approach has been a challenge, partly because of the haptenic nature of STa (molecular weight <2 kDa), which fails to elicit an antibody response (De Weck 1974 and Pereira et a1. 2001 and Boedeker 2005). Additionally, the correlation between STa toxicity and antigenicity (Takeda et a1. 1993) hampers the ability to produce a safe STa/CFAs vaccine. However, it was hypothesized that the poor immunogencity associated with the STa molecule could be improved by conjugation of the STa to a suitable macromolecule (carrier protein) (Elanger 1980 and Pauillac et al. 1998). Therefore, antibody-based therapy (passive immunization) targeting the STa antigen could be used to reduce the impact of ETEC-STa induced diarrhea and avoid the safety issue associated with active immunization with CFA/toxin based—vaccine. Attempts to conjugate the STa to a carrier protein have been reported (Clements, l 990; Houghten et al. 1984, 1985; Klipstein et a1. 1982, 1983; Sanchez et al. 1986, l 988), however no sufficient details were presented on the efficiency and /or the Characteristics of these conjugates. In this study, STa was purified to homogeneity and its properties were ChaJacterized. The purified STa was then covalently cross-linked to modified BSA using 4 different protocols of peptide-carrier conjugation. We have characterized the STa-conjugates for biological activity, conjugation efficiency, and stability of the conjugates. The best STa-conjugate was then used for immunological studies. \General Objectives ' Purify and characterize E. coli STa. I Design and characterize an immunogenic E. coli STa. I Produce and characterize specific antibodies against E. coli STa in laboratory animal models. Specific Aims 1. Production of high performance liquid chromatography - purified E. coli STa I Verify bacterial strain as an ETEC STa-positive strain by identification of the STa encoding gene. I Adoption of bioreactor E. coli culture method for the production of STa and study the growth kinetics at different pH levels. I Biological and molecular confirmation of STa specificity and identity. II. Design and characterization of an immunogenic E. coli STa . I Evaluate four different protocols of peptide conjugation to carrier protein. I Molecular characterization of the most effective STa— conjugates. III- Production of specific antibodies with high neutralizing capacity against E coli STa I Study the neutralization capacity of harvested polyclonal antibodies against E. coli STa. I Study the kinetics of the E. coli STa antigen-antibody interaction: - ELISA STa-IgG binding capacity assay. - Determination affinity and avidity index of STa antibodies. L’O. tential Benefits of the Study Enterotoxigenic E. coli (ETEC) that produces STa is a major cause of severe diarrhea in neonatal animals, children, and adult travelers. The production of specific aIltibodies with a high neutralization capacity against STa can help to reduce the incidence and economic losses associated with ETEC STa-induced diarrhea. An effective immunogenic STa may have potential as an ETEC vaccine that may confer broad protection against ETEC-STa-induced diarrhea. References Boedeker, E. C. 2005. Vaccines for enterotoxigenic Escherichia coli: current status. Current Opinion in Gastroenterology. 21 (1):] 5-1 9. Clements, J. D. 1990. Construction of a nontoxic fusion peptide for immunization against Escherichia coli strains that produce heat-labile and heat-stable enterotoxins. Infect Immun 58, 1 159-1 1 66. De Week, A. L. 1974. Low molecular weight antigens. In: M. Sela (Ed.), The Antigens, Vol. II. Academic Press, New York, Chapter 3. Deneke, C.F; Throne, GM. and Gorbach. S.L. 1981 Serotypes of attachment pili of enterotoxigenic Eschericia coli isolated from humans. Infect immun 32: 1254- 1260. Elanger, BE. 1980. The preparation of antigenic-hapten-cam'er conjugates: a survey. Methods Enzymol. 70 (A), 85. Giannella, R. A. and Elizabeth, A. M. 2003. E. coli heat-stable enterotoxin and guanylyl cyclase C: New functions and suspected actions. Trans Amr Clin Climato Ass. 114, 67-85. Houghten, R. A., Engert, R. F ., Ostresh, J. M., Hoffman, S. R. & Klipstein, F. A. 1985. A completely synthetic toxoid vaccine containing Escherichia coli heat- stable toxin and antigenic determinants of the heat-labile toxin B subunit. Infect Immun 48, 735- 740. Houghten, R. A., Ostresh, J. M. & Klipstein, F. A. 1984. Chemical synthesis of an octadecapeptide with the biological and immunological properties of human heat-stable Escherichia coli enterotoxin. Eur J Biochem 145, 157-162. Klipstein, F. A., Engert, R. F. & Clements, J. D. 1982. Development of a vaccine of cross-linked heat-stable and heat-labile enterotoxins that protects against Escherichia coli producing either enterotoxin. Infect Immun 3 7, 55 0-55 7. Klipstein, F. A., Engert, R. F ., Clements, J. D. & Houghten, R. A.1983. Vaccine for enterotoxigenic Escherichia coli based on synthetic heat-stable toxin crossed- linked to the B subunit of heat-labile toxin. J Infect Dis 147, 318-326. Levine M.M; Rennels, M.B. Daya, V. and Hughes, T.P. 1980. Hemagglutination and colonization factors in enterotoxigenic and enteropathogenic Eschercia coli that cause diarrhea. J. Infect. Dis. 141: 733- 73 7. Moon, H.W. and Bunn, TO. 1993. Vaccinne for preventing enetrotoxigenic Escherichia coli infections in farm animals. Vacine, 11: 213-220. Pauillac, S; Naar, J; Branaa, P. and Chinain M. 1998. An improved method for the production of antibodies to lipophlic carboxlic hapten using small amount of hapten-carrier conjugate. Journal of Immunological Methods 220: 105-1 14. Pereira, C.M; Guth, B.E.C; Aleida, ME. and Castilho, BA. 2001. Antibody response against Escherichia coli heat-stable enterotoxin expressed as fusions to flagellin. Microbiology, 147, 861 -86 7. Saeed, A. M; Magnuson, N. S; Gay, C. C. and Greenberg, R. N. 1985. Characterization of heat-stable enterotoxin from a hypertoxigenic Escherichia coli for cattle. Microbiology and Therapy, 15: 221 -229. Sanchez, J ., Svennerholm, A-M. & Holmgren, J. 1988. Genetic fusion of a non-toxic heat-stable enterotoxin-related decapeptide antigen to cholera toxin B-subunit. FEBS Lett 241, 110-114. Sanchez, J ., Uhlin, B. E., Grundstrom, T., Holmgren, J. & Hirst, T. R. 1986. Immunoactive chimeric ST-LT enterotoxins of Escherichia coli generated by in vitro gene fusion. F EBS Lett 208, 194-198. Sears, C. L; and Kaper. J. B. 1996. Enteric bacterial toxins: mechanisms of action and linkage to intestinal secretion. Microbiol. Rev. 60:167-215. Tacket, C.O; Reid, R.H; Boedeker, E.C; Losonsky, G; Nataro, J .P; Bhagat, H. and Edelman, R. 1994. Enteral immunization and challenge of volunteers given enterotoxigenic E. coli CF A/II encapsulated in biodegradable microspheres. Vaccine 12:1270-1274. Takeda, T; Nair, G. B; Suzuki, K; Zhe, H. X; Yokoo, Y; Hemelhof, W; Butzler, J. B; Takeda, Y; and Shimonishi, Y. 1993. Epitope mapping and characterization of antigenic determinants of heat-stable enterotoxin (STh) of enterotoxigenic Escherichia coli by using monoclonal antibodies. Infection and Immunity 61 :289-294. Thomas, L.V. and Rowe, B. 1982. The occurrence of colonisation factors (CFAI, CF All and E8775) in enterotoxigenic Escherichia coli from various copuntries in south east Asia. Med Microbiol Immunol 1 71: 85-90. Walker, R.I; Steele, D; Aguado, T. and Ad Hoc ETEC Technical Expert Committee 2007. Analysis of strategies to successfully vaccinate infants in developing countries against ETEC disease. Vaccine 25, 2545-25 66. Wolf, MK. 1997. Occurrence, distribution and association of O and H serogroups, colonization factor antigens, and toxin of enterotoxigenic Escxheichia coli. Cli Microbiol Rev 10: 5 69-584. CHAPTER II REVIEW OF LITERATURES Magnitude and impact of diarrheal diseases Diarrhea] diseases are one of the major causes of human death on a global scale. They are the leading cause of childhood death, resulting in 18% of all deaths in children under the age of five (Bryce, 2005). In some populous developing areas, they are responsible for more years of potential life lost than all other causes combined (WHO, 1990). According to the Global Burden of Disease, in 1990, diarrheal diseases were a leading cause of disability-adjusted life year (DALY), second only to lower respiratory infections (est. 99.2 million DALYs lost) (Murray, 1997). Additionally, the National Animal Health Monitoring System (NAHMS, 1994 &1996) survey and the USDA (1997) identified diarrheal diseases as the most common infectious cause of neonatal calf mortalities (est.75% of mortalities). Decades of research have been dedicated to obtaining a better understanding of the neonatal calf diarrhea complex. However, despite improvements in the identification of the infectious agents, management practices, and treatment and prevention strategies, the complex remains the most common and costly (est. $120 million annually) disease affecting newborn calves in the United States (NAHMS, 1996). The list of enteropathogens that can potentially cause diarrhea is quite large and diverse. There are six major pathogens that cause diarrhea in newborn animals: enterotoxigenic Escherichia coli, Rotavirus, Coronavirus, Cryptosporidium parvum, Salmonella spp., and Clostridium perfringens type C (Snodgrass et al. 1986). Diarrhea caused by Enterotoxigenic Escherichia coli (ETEC) is problematic in both animal and human populations. In animal neonates it causes severe profuse watery diarrhea, potentially fatal dehydration and metabolic acidosis (Argenzio, 1985). Additionally, ETEC is a major cause of traveler’s diarrhea (Gorbach et al. 1975) and a leading enterpathogen responsible for gastroenteritis outbreaks on cruise ships (Addiss et al. 1989 and Koo et al. 1996). M] Overview of Escherichia coli Escherichia coli is a common member of the normal flora of the large intestine. Most E. coli strains are avirulent and remain benign commensals because they lack specific genetic elements encoding for virulence factors. However, strains become virulent upon acquiring bacteriophage plasmid DNA encoding enterotoxins or invasion factors. The mechanisms by which virulent E. coli strains acquire such genetic elements seem to be horizontal genetic transfer including phage transudation, transposition, conjugation of plasmids and simple recombination (Seifert and DiRita, 2006). Virulent E. coli strains cause a variety of disease conditions including plain, watery diarrhea, inflarrunatory dysentery, hemolytic uremic syndrome, septicemia, pneumonia and meningitis (Salyers and Witt 1994). Virulence Attributes of ETEC Pathogenicity of E. coli is a complex multifactorial mechanism involving a large number of virulence factors that vary according to the pathotype. They include attachment functions, host cell surface modifying factors, invasins, and many different toxins as well as secretion systems which export toxins and other virulence factors and pilot them to the target host cells. These virulence attributes are often organized into large genetic blocks on the chromosome, known as pathogenicity islands, on a large plasmid, or on phage DNA and can be transmitted horizontally between strains (Seifert and DiRita, 2006). In this regard, E. coli becomes virulent upon the acquisition of plasmids that encode genes for two virulence determinants, namely colonization factors (adhesins), and enterotoxins. Both enterotoxins and CPS work in concert to cause diarrhea in both humans and animals. Two classification schemes are currently used to classify the different strains of E. coli, serotyping and virotyping (Lior 1996 and Levine 1987). Serotyping of E. coli occupies a central place in the history of these pathogens and is mainly based on antigenic differences in the highly variable bacterial surface molecules such as lipopolysaccharides (O antigens) and flagella . (H antigen) (Lior 1996). Specific combinations of O and H antigens define the serotype of an E. coli strain and are widely used for tracing outbreaks of enteric diseases. More than 170 different “0”- specific somatic antigens, 56 different types of flagellar “H” antigens and 103 different capsular “K” antigens in E. coli have been characterized (Nataro and Kaper, 1998). Some E. coli, particularly the ETEC strains, produce mannose resistant (MR) fimbriae that may be used for serological identification. Some of these MR fimbriae (e.g. K88 and K99) were once identified as K antigens before their chemical composition was known. They are presently defined as F (fimbrial) antigens, which also include the mannose sensitive type 1 fimbriae group (Levine et al. 1983). Virotyping is a phenotypic classification scheme based on virulence characteristics of E. coli that includes patterns of bacterial attachments on host cells, the effect of attachments on host cells, production of toxins and invasiveness. 10 Currently there are six virotypes: Enterotoxigenic E. coli (ETEC), Enteropathogenic E. coli or Attaching and Effacing E. coli (EPEH, AEEC), Enterohemorrhagic E. coli, Shiga toxin producing E. coli or Virotoxigenic E. coli (EHEC, STEC, VTEC), Enteroaggregative E. coli (EAEC), Enteroinvasive E. coli (EIEC) and diffusely adherent E. coli (DAEC) (Nataro and Kaper, 1998). In this literature review, we will largely cover the different characteristics of ETEC strains. Structure, Classification, and Antigenic Types of ETEC ETEC organisms are Gram-negative, short rods, not visibly different fiom the E. coli found in the normal flora of the large intestine (Bettelheim 1994). Virulence- associated attributes are fimbriae and enterotoxins. Genes for both attributes are carried on a plasmid and play a crucial role in the pathogenesis of calf scours (Levine et al. 1983 and Nataro and Kaper 1998). All ETEC strains contain plasmids, but this is not a distinguishing feature unless gene probe techniques are used to detect specific virulence-associated genes on these plasmids (N ataro and Kaper, 1998). Pilus or Fimbrial Antigen (E antigen) A common trait of ETEC strains is the expression of several surface- associated proteins, antigenically unrelated to O and H antigens, called F imbriae (Pili) antigens. Fimbriae, which can be seen in electron micrographs as fuzzy coat, are filamentous structures that protrude as hair-like structures from the bacterial surface which are much thinner (3-4 nm) and usually more rigid than flagella (Follett and Gordon, 1980). They are composed of proteins (pilin) that are tightly packed into an array that is shaped like a helical cylinder and their assembly occurs on the outer cell membrane of the bacteria through a specialized pathway of the general secretion 11 system, the alternate chaperone/ usher pathway (Pitek et al. 2004 & Planet et al. 2006) Commensal E. coli strains usually produce so-called common pili, while pathogenic ETEC possess specialized host-specific pili (Levine et al. 1983). Virulence-associated fimbriae are antigenically unrelated to common pili. They are too small to be seen by light microscopy and act as ligands to bind specific complex carbohydrate receptors on the epithelial cell surfaces of the small intestine. Since this interaction results in colonization of the intestine by ETEC, with subsequent multiplication on the gut surface, these pili are termed adhesins or colonization factors antigens (CFAs). A variety of CFs has been described in ETEC strains of animal origin. F4 (K88), F5 (K99), F6 (987P), F41, F42, F165, F17 and F18 are produced by E. coli that cause acute diarrhea in domestic animals. ETEC strains expressing K-99 are pathogenic for calves, lambs and pig, whereas K-88 expressing organisms are able to cause disease only in pigs (Cassels & Wolf 1995). Human ETEC strains also possess an array of colonization fimbriae (CFAI, CFAII and CFAIV) (de Craaf & Gaastra 1994) Nearly all ETEC from calves produce F5 (CFA-K99) and STa (Gyles 1996). In addition to K99, several other fimbriae designated as F41, F92b, Att25, and F210 have been identified on calf ETEC but are much less common (Morris et al. 1983). F41 has been characterized and it mediates attachment to calf enteroytes in vitro and to the small intestine of newborn lambs (Morris et al. 1983). There may be a relatively small group of other fimbriae that also mediate adhesion to calf enterocytes, 12 although K99 is the most common attachment factor on bovine ETEC. This is not surprising, since a similar situation occurs in ETEC isolated from pigs, where K88 and 987P, and at least two other fimbriae known as K99 and F41, also mediate adhesion to pig enterocytes (Moon et al. 1977). Age-dependent resistance to diarrhea caused by F5 positive ETEC develops rapidly in calves (Runnels et al. 1980). Genes coding for the production of CFAs reside on the ETEC virulence transmissible plasmids, usually on the same plasmids that carry the genes for one or both of the two types of E. coli enterotoxin, heat-labile enterotoxin (LT) and heat- stable enterotoxin (ST). Most F4 (K88) strains produce LT while most F5 (K99) and F6 (987P) ETEC strains produce ST. Two types of STs are produced, the majority of ETEC strains produce STa thought a few produce STb. Therefore, most cases of ETEC diarrhea in newborn calves are caused by E. coli possessing a F5 (K99) and an ST gene (Gyles, 1996). Somatic (O) Antigen Somatic antigens are specific heat-stable and composed of polysaccharide chains linked to the core lipopolysaccharide (LPS) complex common to all Gram- negative bacteria. “0” antigen specificity is determined by sugar or amino-sugar composition and by the sequence of these outer polysaccharide chains (Morris et al. 1983). In normal smooth strains, the core LPS is buried beneath the 0 antigen while O-minus mutants, called rough strains, the core LPS is exposed. “O” antigens are not single antigens, but they are composed of several antigenic components and, therefore, are called 0 group antigens. Different 0 groups may share some of these 13 antigenic components; hence, there is considerable cross-reactivity among E. coli 0 antigens; also, many 0 groups of E. coli are cross-reactive or identical with specific 0 groups of Shigella, Salmonella, or Klebsiella (Gyles, 1996). O antigens form the basis of serotyping classification of E. coli. More than 170 different O-specific antigens have been defined but only groups 8, 9, 20, 26, 101 and 141 are common on calf ETEC (Nataro and Kaper, 1998). The role of “0” antigen in the pathogenesis of diarrhea is not clear, however, it is assumed that strains that possess these antigens offer unknown advantageous conditions for the plasmids that carry the genetic elements encoding for enterotoxin and fimbrial production (Evans et al. 1977). Capsular (K) antigens Capsular antigens are acidic polysaccharides that encapsulate the somatic antigen on some but not all ETEC (Acres, 1985). They appear as a dense mat of fibrous projections under electron microscope (Hadad and Gyles, 1982a). The process of E. coli serotyping has encountered difficulty because K antigens prevent the agglutination of the “O” antigens by the homologous sera. They are further subdivided into three groups based on their heat stability; (i) The L-type K antigens are completely destroyed by heat at 100 °C for 1 hour, thus rendering the 0 antigen agglutinable in O antisera. They retain no antigenicity and lose their ability to combine with homologous K antisera. (ii) The B-type K antigens are also destroyed by heat at 100 °C for 1 hour and lose their antigenicity. However, they retain their ability to bind or combine with their homologous antisera. (iii) A-type K antigens are not inactivated by heat at 100 °C, but require heat at 121 °C for 2.5 hr before the O 14 antigen becomes agglutinable. There are 103 different K antigens recognized in E. coli, however only 11 have been identified commonly on calf ETEC strains (Orskov et al. 1977). The role of K antigens in infection by ETEC is not known, but they appear to be important in extra-intestinal infections. Enterotoxins of Epterotoxigenic E. coli Two enterotoxins have been identified among the extracellular products of E. coli isolated from human and other mammals with diarrheal diseases- one is a high molecular- weight type (86.500 kD), namely heat-labile enterotoxin (LT) and the other is a low-molecular weight type called heat-stable enterotoxin (STs). The LT is closely related to cholera toxin (CT), sharing approximately 80% protein sequence identity in the A and B subunits and like CT toxin, stimulates adenylate cyclase and is inactivated by heating. On the other hand, STs completely differ from CT, they act more rapidly, and resist inactivation by heating (Levine et al. 1983; Saeed et al 1984; Sears & Kaper 1996 and Nataro & Kaper, 1998). Heat— Stable Enterotoxins (STs) ETEC strains produce a diverse family of closely related potent peptides toxins that induce secretory diarrhea in humans and animals. STs were initially defined by their resistance to inactivation by boiling for 30 min (Stine and Nataro, 2006). They are classified into two structurally, functionally and immunologically unrelated subtypes, STa (STI) and STb (STII). Table 1 shows the basic differences between STa and STb (Sears and Kaper, 1996 & Nataro and Kaper, 1998). 15 Table 1. Difference between E. coli Heat-Stable Enterotoxin A (STa) and Heat-Stable Enterotoxin B (STb) Properties STa = STl STb = STII Size <2kDa 5.1kDa Number of amino acid residues 18-19 amino acids 48 amino acids Mechanism of synthesis and Pre-pro form 72 amino-acid precursor followed by two consecutive peptidase cleavages 71 amino-acid precursor processed into 48 amino acid mature toxin secreted extracellularly without secretion before extracellular diffusion of furth . 18-19 amino acid mature toxin er p rocessmg Number of cysteine Six cysteine residues F our cysteine residues residues Three S-S bonds Two S-S bonds Toxic domain Hydrophobic: l l-l4 amino acid espgcialy Ala 13 Charged amino acids: especially lys-22, lys 23, arg 29 and asp-30 Solubility Methanol soluble Methanol Insoluble Inactive in suckling mice but active . . Active in mice, infant, piglet and In rats and ligated piglet mtestrnal Actrvrty calves segment. Have no effect on human small intestine. Trypsin effect Resistant Sensitive Does not act on cyclic nucleotides. Mechanism of action Act on guanylyl cyclase C Ca, PGE2 and serotonin may be its mode of action Loss of villus epithelial cells and Effect on partial villus atrophy (Nagy and enterocytes: No effect Fekete 1999) STaP (procine isolates) and STaH Prototypes (human isolate) None Producing strains ETEC and other bacteria Only ETEC 16 Genes encoding for both classes are found predominantly on plasmids, and some STa-encoding genes have been found on transposons. So and colleagues (1979, 1980 & 1981) demonstrated that the STa gene is embedded within a functional transposon, and the presence of STa on widely variable plasmids supports this as the dominant mechanism for ongoing transfer. Enterotoxigenic E. coli STa (ST 1) There are two slightly different toxin prototypes under this designation: STIa (STp) and Sle (STh). Both variants are structurally, functionally and antigenically related. They share a core 13 amino acid-sequence that is necessary and sufficient for enterotoxic activity but differ in their N-terminal sequence. While STp is produced by porcine, bovine and human ETEC, STh is onlyproduced by human isolates (Nataro and Kaper, 1998). A large family of STa-related toxins has been also identified in various organisms. All share similar size (ranging from 15-30 amino acids) and a highly conserved 13-amino-acid C- terminal region, which is essential for their toxicity (Table 2) (Stine and Nataro, 2006). The biochemical, physiOlogical and immunological properties of the various STa enterotoxins are remarkably similar. They have four conserved amino acids (N-P-A-C) (Stine and Nataro, 2006) that mediate binding to the membrane guanylyl cyclase-C (Greenbreg et al. 1997). Most of them have six cysteine residues located in the same relative positions and linked intrarnolecularly by three disulfide bonds, which suggests that these enterotoxins have similar tertiary structure (Shimonishi et al. 1987). The secretory potency and heat 17 stability of the STa enterotoxins are determined by conserved core sequence (Yoshimura et al. 1985 and Shimonishi et al. 1987). Table 2. The STa Family of Toxins Toxin and No. Sequence Reference host Amino acids STaH ETEC 19 N-S-S-N-Y-C-C-E-L-C-C-N-P-A-C-T-G-C-Y Aimoto et al 1982 STaP ETEC 18 N-T-F-Y-C-C-E-L-C—C-N-P-A-C—A-G-C-Y Takao et al 1983 STa ETEC 18 N-T-F-Y-C-C-E-L—C-C-N-P-A-C-A-G-C-Y Saeed et a1 1984 (bovine) C itrobacter 18 N-T-F-Y-C-C-E-L-C-C-N-P-A-C-A-G-C-Y Guarino et a1 1989 Freundii Yersinia 30 S-S-D-Y-D-C-C-D-Y-C-C-N-P-A-C-A-G-C Takao et a1 1985 enterocolitica V. cholera l7 l-D-C-C-E-l-C-C-N-P-A- C-F-G-C-L-N Yoshimura et a1 Non-01 1986 V.cholera non- 18 L-l-D-C-C-E-l-C-C-N-P-A-C-F—G-C-L-N Arita et al 1991 01 Hataka V. mimicus l7 l-D-C-C-E-l-C-C-N-P—A-C-F-G-C-L-N Arita et al 1991 E. coli EAST -l 38 ...A-S-S-Y-A-S-C-l-W-C-T---T-A-C-A-S—C-H-G Savarino et a1 1993 Conus l3 E-C-C-N-P—A-C-G-R-H-Y-S-C Gray et a1 1981 geographus I3 Guanylin '15 P—G-T-C-E-l-C-C-AY-A-A-C-T-G-C Greenberg et al (human) 1997 Adopted from Nataro & Stine (2006) 18 Structural determinants and biochemical properties of STa (Eigpre l) STa is a small peptide (molecular weight of less than 2 kDa) that has 18- or 19- amino acids (Sato & Shimonishi, 2004). STa is produced as a 72 amino-acid precursor that is cleaved by signal peptidase l to a 53-amino- acid-peptide (pro-STa) (Rasheed et al. 1990). Prior to secretion by the bacteria, three intramolecular disulfide bonds crucial to toxin activity are formed in the periplasm by DsbA, chromosomally encoded protein (Yamanaka et al. 1994). A second undefined proteolytic event occurs extracellularly to process the pro-STa to produce biologically active STa of 18 or 19 amino acids which is released by diffusion across the outer membrane (Rasheed et al. 1990) The structural characteristics of biologically active STa were extensively studied (Gariepy et al. 1986; and Osaki et al. 1991) on weakly toxic and nontoxic STa analogs (Sato et al. 1994). Using X-ray diffraction analysis, Sato and Shimonishi, (2004) studied the molecular structure of the toxic domain of fully active STa molecule, which consists of the native sequence from Cys5 to Cysl7 and revealed that the peptide molecule forms a ring-shaped peptide hexamer. The outer surface of this peptide is occupied exclusively by the central portion of the side chains of Leus, Asnl ', Pro”, and Ala”, proposed to be a binding site to the receptor protein, guanylyl cyclase C, GC-C while the inner surface of the peptide hexamer consists of a single invariant Gly residue (Osaki et al. 1991; Sato and Shimonishi, 2004). Overall the STa molecule has a hydrophobic nature with certain structural elements that are functionally critical. One of which is the occurrence of six cysteine residues participating in three disulfide bridges (Garpiey et a1. 1987; Okamoto 1987; 19 Figure 1. Structural Determinants of Escherichia coli STa ’ ETEC STa ‘ Arrrino acid sequence _ Small MW Structural Determinants ]—— 2kDa INTFYCCELCCNPACAGCY”; 1 Non immunogenic Hydrophohrc NMR & X ray Crystallography ‘ 6-Cysteine residues | L 3-beta turns 1 3-disulfide bonds Bl turn 132 turn B3 turn Cys6- Asn1 1- CycM- Cys9 Cysl4 Cysl7 l Cyss-Cysl0 I Cysé-Cysl4 l Cys9-Cys17 11-14 residues NPAC esp Ala13 Stability and binding activity sites 20 Osaki et al. 1991; Sato et al. 1994 and Sato & Shimonishi, 2004) suggesting that this toxin has tertiary structure (Shimonishi et al. 1987). Listed in descending order of importance in terms of their respective contribution to enterotoxic activity, the disulfide linkages between Cys6 and Cys”, CysS and Cys'o, and Cys9 and Cysl7 are all necessary for stabilizing the STa molecule and expressing full STa toxic activity (Garpiey et al. 1987). In addition, the second B-turn at residues 11 to 14 (especially Ala”), is proposed to be crucial to the STa toxicity and its interaction with GC-C (Osaki et al. 1991 and Sato et al. 1994). Amino acid sequence of STa purified from several strains of bovine ETEC was reported by Saeed and colleagues (1983, 1985a & b). STa from bovine ETEC was found to consist of 18 amino acids and was similar to STa purified from ETEC of human and porcine origin. ETEC (STa) is methanol and acetone soluble. It resists proteolytic (pronase and trypsin) enzymes (Jacks and Wu, 1974), heat and acids but not alkalines. The toxin loses biological activity on treatment with reducing agents, such as p-mercapto- ethanol, dithiothreitol or after performic acid oxidation suggesting the presence of disulfide bridges (Eldeib et al. 1986). Antigenicity and antigenic determinants of E. coli STa E. coli STa is poorly immunogenic because it has low molecular weight (Gyles 1971; Evans et al. 1973; and Alderete & Robertson, 1978). The antigenic determinants of STa have been analyzed (Takeda et al. 1993). Three distinct antigenic sites of STa have been recognized, one near the N-terminus, another in the core functional region, and the third in the C-terminal, Asn-Tyr‘l-Leu8 -Asn”-Pro'2-Alal3- 21 Cys'4-Tyr18. Characterization of various STa epitopes revealed that the N—terminus residues, which are not essential for the biological activity of STa in suckling mice (Aimoto et al. 1982; Shimonishi et al. 1987 and Yoshimura et al. 1985 & 1987), but possess an important antigenic determinant (Takeda et a1. 1993). Structural-Function Relatioyhip of E. coli S'I‘ja Four characteristic structural features of the STa molecule greatly contribute to its function: hydrophobicity, single invariant glycine residue, conserved 6-cysteine residues, and geometrical shape of this molecule (Sato and Shimonishi, 2004). Hydrophobicity of the STa creates a favorable situation for an interaction with GC-C on biological membranes. The side chains of the amino acids of the binding region of ST (Asnl 1, Pro‘2 and Ala”), which are constant for numerous bacteria, form a prominent, isolated cluster that projects from the surface to the outside of the molecule and allows binding with GC-C because of their hydrophobic nature. Site directed mutagenesis studied the contribution of these residues to the toxic activity of the STa molecule and found that replacement of Ala at position 13 by Gly or Leu residue led to loss of enterotoxigenic activity of the toxin (Sato and Shimonishi, 2004) suggesting that Ala13 has a key role in the interaction of STa with its receptor domain. Other amino acid residues were tested for their role in STa-receptor binding (Takeda et al. 1991 & 1993 and Yoshino et a1. 1994). The inner surface of the peptide hexamer is occupied by a single invariant Gly. The backbone conformation in this region is unique for the Gly residue and other amino acid substitution may disrupt the formation of hexamer structure by steric hindrance (Sato and Shimonishi, 2004). 22 STa has 6-cystiene residues connected with each other though three intramolecular disulfide bonds (Figure 2) which are crucial for the spatial structure of STa and for the expression of toxicity. Yamasaki et al. (1990) studied the contribution of each disulfide bond to the toxic pr0perty of STa. The authors found that peptides with only one disulfide bond are not biologically active, but peptides with a disulfide bond in the second B-turn and one other disulfide bond had distinct activity. A peptide that lacks a disulfide bond in the second [3 turn is not toxic. Clearly, disulfide bonds are essential to stabilize the spatial structure of STa are necessary for STa toxicity and stability. The finding that native ST forms a self-associated hexamer is consistent with other structure-activity relationships, which are difficult to rationalize using the monomeric structure of STa analogous (Sato and Shimonishi, 2004). Mpg" of ETEC-STa: valyl Cyclase C (GC-C) The major receptor for STa is a membrane-bound (particulate) guanylyl cyclase C (GC-C), (Field et al. 1978; Hughes et al. 1978; Schulz et al. 1990 and Vaandrager, 2002, Giannella and Elizabeth 2003) which belongs to a group of receptor-linked enzymes that catalyzes the conversion of guanosine 5'-triphosphate (GTP) to second messenger cyclic guanosine 5'-monophosphate (cGMP) in response to diverse signals, such as peptide hormones. In addition to GC-C, this family includes also the atrial natriuretic peptide receptors (GC-A & GC-B) (Lowe et al. 1990 a&b) and the olfactory (GC-D) (Yang et al. 1996) and retinal cyclases, (GC-E, & GC-F) (Oliveira et al. 1994 and Yang et a1. 1996). 23 Amide backbone Side chains binding site l l l l Cyss-Cys‘S-Glu-CLeu8 -Cys9 -Cysl0 -Asn"-Pro12 - Alau’ -Cys"—Ala'5-Glyl6-Cysl7 I J l l l l lSt B turn 2nd B turn 3"I B turn Crucial Structure of STa STaP: N-T-F-Y-C51-C6-E-L-C9-CLLN-P-A-CI‘ll-A-G-Cl7-Y STaH: N-S-S-N-Y-p6-C7-E-L-C”-91l-N-P-A-Clls-T-G-Cls-Y Figure 2. Intramolecular cysteine-disuflide linkage for STaP and STaH (Sato et al. 1994) 24 All these GCs are receptor-linked enzymes with one membrane-spanning region and share a conserved intracellular catalytic domain (ICD) but they differ in their extracellular ligand-binding domains (ECD) because they are activated by different signals (Lucas et al. 2000; Schulz et a1. 1990 and Vaandrager, 2002) (Figure 3). The size and nature of the STa receptors have been an area of intensive investigation resulting in the identification of at least one definitive STa receptors located in the apical membrane of the intestinal epithelial cells (Schulz et al. 1990; Thompson & Giannella, 1990; and Cohen et al. 1993). Binding of ligands (STa, guanylin and uroguanylin) to the ECD stimulates the intracellular enzymatic activity. Molecular cloning from various mammalian species revealed that the primary structure of GC-C (120-kDa protein unglycosylated and a 140 to 160-kDa protein after N-linked glycosylation) is composed of an extracellular N—terminal receptor domain (ECD), a single transmembrane domain (TMD) and a cytosolic domain (ICD). ICD comprises a kinase homology domain (KHD) which is linked by an or- helical putative dimerization domain to the C-terminal catalytic domain (Schulz et al. 1990; de Sauvage et a1. 1991; Garbers, 1999; Lucas et al. 2000). GC—C contains a unique ECD comprising approximately 40% of the total protein, which contains 8-10 N-linked glycosylation sites and functions as the binding site for GCC-ligands (Schulz et al. 1990 and Lucas et al. 2000). The ligand recognition sites and the importance of carbohydrate moieties on GC-C were studied by site directed mutational analysis of the ECD and revealed that the N-linked carbohydrate moiety at N379 plays an essential role in stabilizing the functional structure of ECD. 25 ANPCR ch Amino terminal nun—nun— .— ECD TMD 1H KHD Hinge region Catalytic Domain Carboxyl terminal Figure. 3. Domain structure of guanylyl cyclases (Lucas et al. 2000) 26 It also revealed that the ligand specifically binds to the region residue 387- 393. Both the N—linked carbohydrate moiety at N379 and residue 387 to residue 393 regions are located close to the transmembrane portion of GC-C on the external cellular surface which explain their pivotal role in ligand binding and signaling processes (Hasegawa et a1. 1999 and Hasegawa & Shimonishi, 2005). Solubilization and STa receptors cross-linking experiments with intestinal tissues have been repeatedly identified several species of both small and large STa- binding proteins, 45 to 80kD and 120-160kD respectively (Cohen et a1 1993; de Sauvage et al. 1992, Hakki et al. 1993; Ivens et al. 1990; Thompson & Giannella, 1990 and Vaandrager & de Jonge 1994). Study of biogenesis, cellular localization and functional activation of STa receptors (Runder et al. 1996) demonstrated that only the full length of STa receptor is translocated to the cell surface and is thus the only species available to interact directly with the extracellular environment. The smaller protein forms are likely to represent premature termination products of STa receptor that are not transported to the cell surface. The generation of antibodies to GC-C furthermore suggested that low molecular weight STa binding proteins were proteolytic fragments of GC-C (de Sauvage et al. 1991 and Vaandrager et al. 1993). The study of STa receptor biogenesis also showed that ligand stimulation of GC-C does not lead to recruitment of additional receptor to the cell surface. This proves that STa receptor is rapidly internalized and recycled back to the cell surface after binding STa (Runder et al. 1996 and Urbanski et al. 1995). STa-GCC receptor binding kinetics has been studied (Giannella et al. 1983 Frantz et al. 1984; Carpick & Gariepy 1991 & 1993; and Al-Majali et al. 1999 a&b). 27 Giannella (1995) showed that the binding constant of STa is two log orders less than that of CT for its receptor and accounts for the rapid reversibility of STa-induced secretion and the relative "permanency" of CT-induced secretion (Fasano, 1999). Previous studies of affinity chromatography fail to recover the functional STa receptors from affinity matrices which contain STa covalently cross-linked to a solid support (Thompson, 1987 and Hugues & Waldman 1992). This suggests that the STa- receptor interaction is an irreversible process. However, Hugues et al. (1992) showed that bile salts completely dissociated ST-receptor interaction despite the low recovery of total ST binding activity, thus supporting the theory of reversibility of STa- receptor interaction. Several groups of workers have studied the age-dependence, density and distribution of STa receptors in different models including human, pig, rat, rabbit and calf intestines (Cohen et al. 1986; Forte et al. 1989; Jaso-Friedman et al. 1992; Katwa and White 1992; Krause et al. 1994 and Al-Majali et al. 1999 a, b&c). In all of these models, a consistent increase in STa receptor density along the brush border membrane was observed in immature host. This coincides with the period of high susceptibility to STa-induced diarrhea that occurs in the early life of human and animals. Study of distribution and characterization of STa-specific receptors on enterocytes and brush border membrane vesicles (BBMVs) from different intestinal segments of newborn calves found that higher density and affinity of STa receptors on enterocytes and BBMVs derived from the ileum than other intestinal segments (Al-Majali et a1. 1999 a&b). Moreover, ileal villous epithelial cells have approximately twice as many receptors as crypt cells for the enterotoxin (Giannella et 28 al. 1983 and Frantz et al. 1984). This strongly explains why STa act more rapidly than LT. The endogenous agonist for GC-C is 15- amino- acid hormone called guanylin, which contains four cysteines and is less potent than STa in activating GC- C and stimulating chloride secretion (Carpick & Gariepy 1993; Currie et al. 1992 and Forte & Currie 1995). Guanylin presumably plays a role in basal gut homeostasis, and STa opportunistically utilize GC-C to alter ion transport in the gut. The STa enterotoxin brings its secretory effect through the GC/CGMP pathway. Intestinal fluid and electrolyte homeostasis Most of enteropathogens, including ETEC, target normal processes of intestinal water and electrolyte transport. Consequently, an understanding of the physiology of intestinal fluid and ion transport is essential. Fluid is both secreted, to provide an environment that promotes the digestion and absorption of ingested nutrients, and absorbed, to avoid excessive fluid loss and resulting dehydration (Beme & Levy 1996 and Montrose et al. 1999). Intestinal absorptive mechanisms Sodium ions play a central role in net nutrient and water intestinal absorption (Chang & Bookstein 2000). Glucose, amino acids and other nutrients are absorbed, through an ion selective channel, or by a process coupled transport of Na+ via a Naif/H)r cation exchanger and Cl'/HC03 anion exchanger. All of these pathways involve Na/K-ATPase pumps on the basolateral membrane of the enterocyts, (Chang & Bookstein 2000). 29 Intestinal secretory mechanisms The primary driving force for secretory fluid fluxes in the intestine is the movement of Cl' from the blood circulation to the intestinal lumen. Chloride accumulate in the cytosol above its electrochemical equilibrium in response to the sodium concentration gradient established by the basolateral Na, K- ATPase and exit across the apical membrane when Cl‘ channels at that site Open in response to changes in second messengers. The most important of these Cl' channels is the cystic fibrosis transmembrane conductance regulator (CFTR) (Barrett and Bertelsen, 2003). Role of cyclic nucleotides in intestinal homeostasis The important role of both cAMP and cGMP in the control of intestinal epithelial ion transport has been documented (Rao et al. 1981 and Donowitz and Welsh 1986). Cyclic AMP (cAMP) is generated from ATP in response to the activation of adenylate cyclase while cGMP is generated from GTP through the action of guanylyl cyclase. Both cyclases are membrane bound enzyme and are activated in the presence of appropriate agonists. The cAMP-dependent Cl' secretion is induced through protein kinase A (PKA) (Berger et al. 1993, Shlatz et al. 1979) while cGMP-induced Cl' secretion has been documented to occur via a protein kinase GII (PKGII) pathway (Selveraj et al. 2000). Both PKA and PKGII appear to be capable of phosphorylating and thereby activating of CF TR in the presence of appropriate levels of intracellular signaling. Absorptive mechanisms may also be affected by changes in cyclic nucleotide, although in this case it can be inhibitory. Na+/H+ exchangers (NHE3) are also regulated by phosphorylation events via cAMP-induced PKA action; however this 30 activation has an inhibitory effect on NHE3 with the subsequent inhibition of Na absorption. Therefore, activation of PKA leads to inhibition of NHE3 and activation of CFTR stimulating Cl' secretion and inhibiting neutral Na+/Cl' absorption (Yun et al. 1995). Pathogenesis of ETEC-STa Diarrlle_a_ ETEC diarrhea occurs in the first few days of life, particularly in the most colostrum-deprived calves. Three criteria must be met for ETEC to produce illness: the ETEC must express adhesive factors that allow it to adhere to the small intestine; the ETEC must possess the plasmid that encodes for the enterotoxin(s), and sufficient numbers of the ETEC must be ingested. The pathogenesis of ETEC diarrhea involves three steps: establishment of infection, intestinal colonization, and finally elaboration of diarrheagenic enterotoxin(s) leading to an acute profuse watery diarrhea and potentially fatal dehydration, metabolic acidosis and electrolyte imbalances (Argenzio 1985). Infection with ETEC Calves contract infection with E. coli from the environment during or shortly after birth, often by fecal-oral route of transmission (Smith, 1965 and Butler & Clarke, 1994). Under normal circumstances, nonpathogenic types of E. coli are among the first bacterial species to gain access the intestinal tract, and they are prevalent throughout the intestine by the end of the first day of life with marked increase in number from the proximal to the distal portion of the small intestine (Smith, 1965 and Acres, 1985). In healthy calves, the majority of these nonpathogenic E. coli are suspended in intestinal contents and are constantly 31 propelled caudally by peristaltic and flow of ingesta and their number rarely exceed 107/g intestinal content. However, when ETEC are ingested they multiply and colonize the small intestine in larger numbers. Rapid establishment of E. coli infection is favored by several characteristics of newborn calves including a relatively high abomasal pH, sluggish intestinal motility and the absence of competing microflora (Acres, 1985). Intestinal colonization The ability to produce enterotoxin alone is not sufficient for ETEC to cause diarrhea. The bacteria must also able to colonize the mucosal surface of epithelial cells of the small intestine. This depends on adhesive factors that are composed of nonflagellar, flimentous fimbriae that are protein in nature. These fimbriae bind to specific receptors in the membrane of cells of the small intestine and allow ETEC to overcome the peristaltic cleansing mechanisms of the intestine. In normal calves, the majority of E. coli are in the lurnenal content and only 10-20 % attached to the mucosa. In contrast, in claves with enteric colibacillosis the situation is reversed and 80 % to 90% of ETEC are attached the mucosa (Hadad and Gyles, 1982a&b). The dynamics of this process have been studied in colostrums-deprived calves where it appears that colonization begins at the ileal-cecal junction within 3 hours of infection and progress anteriorly to involve up to 60% of the small intestine by 16 hours post infection. The precise molecular mechanism of attachment is not well understood, however bacterial fimbrial are definitely involved in this process (Smith & Huggins, 1978 and Hadad & Gyles, 1982a&b). Both the surface of ETEC bacterial cells and 32 intestinal epithelial cell possess a negative net charge which tends to repel each other. To overcome this physicochemical property, adhesive organelles, fimbriae K99, protrude from the bacteria cell surface and initiate attachment by extending across the zone of repulsion to reach specific complex receptors on the epithelial cell surfaces of the small intestine and act as a ligand to bind the bacterial cell to the intestinal surface (Acres, 1985 and Donnenberg, 2000). Receptors are probably sugar residues located in the cell membrane of the epithelial cells and those which bind to K99 or F41 fimbriae appears to contain sialic acid (Paris et al. 1980). Following primary attachment, ETEC multiply and form rnicrocolonies that cover the intestinal villi (Chan et al. 1982 and Bellamy & Acres, 1979). Colonization of the posterior half of the intestine is the central event in the ileum and there may be as many as 109-10l0 ETEC/g of intestinal contents (Smith and Huggins, 1978). The degree of intestinal colonization varies among ETEC strains and is probably affected by serotype as well as physiological and nutritional factors in the small intestine (Chan et al. 1982, Isaacson, 1980 and Bellamy & Acres, 1979). Maximum colonization seems to occur with strains that are fimbriated and encapsulated (Smith & Huggins, 1978 and Hadad & Gyles, 1982b). In addition, some strains posses only one type of fimbriae whereas others posses more than one type which may intensify colonization and increase virulence (Orskov et al. 1977). Elaboration of eJnterotoxin: Pathophysiology of STa action When large numbers of ETEC colonize the small intestine, sufficient STa is produced to cause diarrhea. Presumably, enterotoxin is released by the bacteria and diffuses toward the brush border where it binds to receptor sites on the membrane of 33 the intestinal epithelial cells. Enterotoxin alters normal movement of ions and water across the intestinal mucosa by exerting a hormone-like effect on the enterocytes (Moon, 1974 & 1978). Fluid and electrolytes constantly move across the intestinal mucosa in two directions: from the intestinal lumen to the blood (absorption), and from blood to intestinal lumen (secretion). Secretory and absorptive fluxes occur simultaneously, and the net movement of fluid is the difference between the two processes (Finco et al. 1973 and Moon 1994). The villus-crypt unit is the functional apparatus through which these fluxes occur (F inco et al. 1973). Immature enterocytes in the crypt are the main secretory cells, whereas more mature villous enterocytes are responsible for digestion and absorption. In normal calves, absorptive fluxes exceed secretory fluxes creating a state of net absorption (F inco et al. 1973 and Acres, 1985). STa appears to act by stimulating secretion as well as reducing absorption, and this dual effect reverses the normal pattern causing a net secretion of fluid into the small intestine and resulting in hypersecretory diarrhea (Rao et al. 1980).The total volume of each unidirectional flux in normal calves 4-6 week of age is between 80-144 liters/day (Bywater, 1973). Calves infected with ETEC may lose between 1.0-2.7 liters of diarrhea] fluid in 24 hours, which represents only 1-2% of the normal unidirectional flux volume (Fisher and Martinez, 1975). This illustrates that only a small percentage change of the normal capacity of the small intestine is required to cause severe diarrhea. STa induces secretory diarrhea by activating guanylyl cyclase-C and increasing cGMP (Figure 4). Disruption of the gene encoding GC-C in mice resulted in resistance to ST-induced diarrhea, demonstrating that GC-C is absolutely required 34 for ST-induced intestinal secretion (Mann et al. 1997 and Schulz et al. 1997). Cystic fibrosis transmembrane conductance regulator (CFTR), chloride channel, is a key component mediating the enterotoxigenic effect of STa (Golin—Bisello et a1. 2005). In the absence of functional CFTR, ST and cGMP analogs fail to induce diarrhea (Quinton, 1990). Cyclic GMP activates CFTR and promotes chloride efflux, which presumably drives water transport into the lumen of the intestine. The intermediate steps involved in the CFTR phosphorylation are controversial, though the roles for both cGMP-dependent kinases and cAMP-dependent kinases have been reported (Sears and Kaper, 1996). GMP-dependent kinases (PKG) appear to be the principal molecular target of cGMP in the signal sequence leading to CFTR activation. Both PKG-Ia and PKG-H phosphorylate CFTR, in vitro, with similar kinetics, suggesting the absence of a specific PKG-mediated function in this process (French et a1. 1995). However, PKG- II., but not PKG-Ia colocalizes with GC-C in brush borders of enterocytes and activates CFTR in excised membrane patches of various cell lines transfected with CFTR (Lohmann et al. 1997) suggesting that PKG-II is a major physiological mediator of CFTR activation in small intestine (Pfeifer et a1. 1996; Vaandrager et al. 1997 & 2000). The role cAMP-dependent kinases (PKA) are also reported. They may be activated either directly by cGMP or indirectly by local accumulation of cAMP in response to inhibition of PDE3 by cGMP and leads to CFTR activation. Vaandrager et al (2000) reported that STa induce electrogenic chloride secretion in the colon and jejunum of PKG II-deficient mice. 35 Lumen V 1, fl STa Mucosa Brush border Serosa Figure 4. Pathophysiology of E. coli STa diarrhea. “Enterotoxigenic E. coli, containing plasmids encoding a member of the homologous peptide family of STs colonize the intestine after the consumption of contaminated food and/or water. Bacterial colonization leads to production of ST in the gut lumen, which specifically recognizes and binds to the extracellular domain of GC-C, expressed in the brush border membranes of intestinal mucosa cells from the duodenum to the rectum. Interaction of ST to the extracellular domain of GC-C is translated across the plasma membrane into activation of the cytoplasmic catalytic domain resulting in the production and accumulation of [cGMP]. This cyclic nucleotide binds to and activates PKG II, also localized in the intestinal cell brush border membrane. Also, cGMP may activate PKA, either directly or by inhibiting a cAMP-specific PDE and inducing the accumulation of cAMP. The CFTR that is colocalized with GC-C and PKG II in brush border membranes is a substrate for that protein kinase and PKA. CFTR is a chloride channel, and its phosphorylation by PKA or PKG results in a persistent open state, permitting chloride to flow down its concentration gradient from the intracellular to the extracellular compartment. Other ion channels and transporters in the cell maintain the electroneutrality of ST-induced chloride efflux. Vectoral water flux from the basolateral to the apical surface is driven by these ionic conductances, resulting in the accumulation of fluid and electrolytes in the intestinal lumen and secretory diarrhea” (Lucas et al. 2000) 36 In addition to CF TR activation, recent evidence suggests a role for inhibition of brush border membrane electroneutral sodium absorption, possibly mediated by a Nail/H+ exchanger, in mechanisms underlying STa-induced fluid and electrolyte secretion (V aandrager et al. 2000). Lucas and colleagues (2001 & 2005) reported that STa reduced fluid absorption mediated by a Na+/H+ exchanger but does not act by causing increased fluid secretion. Hence, the STa-induced diarrhea is the result of an increase in net fluid secretion due to stimulation of chloride secretion in response to CFTR activation and electrogenic chloride transport. Whether ST affects the Na+/H+ exchange in the small intestine and hence modifies acidification in the lumen is still unknown (Laohachai et al. 2003). Potential systemic effect of ETEC-STa diarrhea . The systemic effects of ETEC-STa diarrhea, which eventually contribute to deaths, are precipitated by a single event, the loss of extracellular fluid. (ECF). Four major potentially fatal can be identified due to the loss of ECF: dehydration, electrolyte imbalances, metabolic acidosis and negative energy balance (Fisher, 1965, Barber et a1. 1975; Moon et al. 1978; Argenzio, 1985; Booth and Naylor, 1987; Groutides and Michell 1990 and Grove-White, 1996). Metabolic acidosis is an important consequence of ETEC diarrhea and a nmnber of factors contribute to its development. A major factor is the loss of bicarbonate ions in feces and additionally, an increase in the production of lactic acid in poorly perfused tissue and reduction in renal hydrogen ions excretion secondary to dehydration (Booth and Naylor, 1987). 37 Hyperkalemia is a complex sequence to ETEC diarrhea and occurs despite a significant loss of potassium ions (K+) in the feces. This paradoxical situation is the result of acidosis and compromised renal function. A shift of potassium ions from intracellular to ECF compartment in exchange with the excess extracellular hydrogen ions occurs to maintain cell electroneutrality (Lewis and Phillips, 1972). This redistribution of K+ ions in response to acidosis causes a reduction in the resting membrane potential leading to serious and eventually lethal effects on cardiac muscle fimction (Fisher 1965, Fisher and McEwan, 1967 and Phillips and Knox, 1969). Hyponatremia and hypochloremia result from significant fecal loss of sodium and chloride ions. Hypoglycemia frequently occurs in the last stage severe ETEC diarrhea near death. Anorexia, decreased absorption of nutrients, minimal glycogen reserves, inhibited gluconeogenesis, and increased anaerobic glycolysis in poorly perfused tissues may all contribute to the negative energy balance (Hall et al. 1992). Current Approaches for Controlling ETEC-STa- induced Diarrhea Several approaches for the treatment and control of ETEC diarrhea are briefly described below: Antibiotic-based therapy: The use of sub-therapeutic doses of antibiotics may help protect animals from some but not all of the bacterial strains. However, the use of antimicrobial agents at sub-therapeutic levels has been linked to the problem of emerging antibiotic resistance among several bacterial species, including ETEC strains. Mechanism-based therapy: Therapeutic trails were also explored in an attempt to approach ETEC-STa diarrhea through its putative receptors (Mechanism-based 38 therapy). Mann et al. 1997 and Schulz et a1. 1997 showed that disruption of the gene encoding GC-C in mice resulted in resistance to ST-induced diarrhea. However disruption of genes involved in the regulation of intestinal secretion may have adverse effect on other host’s cell function. One study showed the GCC knockout- mice were resistant to STa—induced diarrhea but they suffered of over 33 percent stunted growth compared to normal mice (Pfeifer et al. 1996). Another study targeted the path of the signaling cascade mediated by STa using 2-chloroadenosine triphosphate (2-chloroATP) to interrupt STa stimulation of chloride current and water secretion in Caco-2 cells (Zhang et al. 1999). However, models of STa-induced secretion developed in vivo are particularly unsuited to examine the therapeutic efficacy of prodrug (2C1Ado) that requires time-dependent transport and metabolic conversion to the active moiety of 2-chloroATP. Moreover, the pharmacokinetic barriers to maintain this active moiety in enterocytes in vivo limit examination of the utility of this approach to prevent STa-induced diarrhea. Immunity-based therapy Active immunization Active immunization through vaccination is an effective way to control infectious agents. However, the current strategy for development of a broad-spectrum vaccine against ETEC targeting the pili antigen has been challenged by the numerous intestinal colonization factors expressed by ETEC and continuous antigenic drift of pili antigens (Boedeker, 2005 and Walker et al. 2007). 39 Passive immunization Antibody-based therapy, which includes pathogen-specific and non pathogen- specific modalities, plays an important role in modern therapy against several diseases, including ETEC. However, due to the lack of common ETEC antigen candidates, there are no immunotherapeutic reagents that can confer broad protection against the wide array of ETEC strains. The use of STa toxin as a common antigen to confer a broad protection against 75% of ETEC is rationalized. However, antibodies raised against STa toxin have been limited by several factors including the poor immunogenicity of the STa, and the fact that the design of an effective immunogenic STa is dependant on a successful conjugation reaction. Antibody-based therapy against STa is also challenged by the large amount of STa antibodies required to neutralize the local effect of the STa (Svennerholm and Steele, 2004). 40 References Acres, S. D. 1985. Enterotoxigenic Escherichia coli infections in newborn calves: A review. Journal of Dairy Science, 68, 229-256. Addiss, D. G; Yashuk, J. C; Clapp, D. E. and Blake, P. A.1989. Outbreaks of diarrheal illness on passenger cruise ships, 1975-1985. Epidemiol Infect. 103: 63- 72. Argenzio, R. A. 1985. Pathophysiology of neonatal calf diarrhea. Vet Clin of Noth American. Food Animal Practice. 1: 461-469. Alderete, J. F and Robertson, D. C.1978. Purification and chemical characterization of the heat-stable enterotoxin produced by porcine strains of enterotoxigenic Escherichia coli. Infection and Immunity 19:1021-1030. Al-Majali, A. M; Robinson, J. P; Asem, E. K; Lamar, C; Freeman, M. J; and Saeed, A. M. 1999a. Use of flow cytometry to measure the interaction between Escherichia coli heat-stable enterotoxin and its intestinal receptor in mice. J Immunol Methods 222(1-2): 65- 72. Al-Majali, A. M; Robinson, J. P; Asem, E. K; Lamar, C. H; Freeman, M. J; and Saeed, A. M. 1999b. Characterization of the interaction of Escherichia coli heat-stable enterotoxixn (STa) with its intestinal putative receptor in various age groups of mice, using flow cytometry and binding assays. Lab Anim Sci.3: 25 4-259. Al-Majali, A, M; Robinson, J. P; Asem, E. K; Lamar, C; Freeman, M. J; and Saeed; A. M. 1999c. Age-dependent variation in the density and affinity of Escherichia coli heat-stable enterotoxin receptors in mice. Adv Exp Med Biol.4 73:13 7-45. Aimoto, S; Takae, T; Shimonishi, Y; Hara, S; Takeda, T; Takeda, Y; and Miwatani. T. 1982. Amino-acid sequence of heat —stable enterotoxin produced by human enterotoxigenic Escherichia coli, Eur. J. Biochem. 129: 257-263. Arita, M; Honda, T; Miwatani, T; Takeda, T; Takao, T; and Shimonishi. Y. 1991. Purification and characterization of a heat-stable enterotoxin produced by Vibrio mimicus. FEMS Microbiol. Lett. 63: 105-110. Barber, D. M. L; Doxey, D. L; and Maclennan, W. 1975. Biochemical studies of the collapse syndrome in suckled calves. Vet. Rec 29: 424-428. Barrett, K. M. and Bertelsen, L. S. 2003. Physiological regulation of gastrointestinal ion transport. In Microbial pathogenesis and the intestinal epithelial cell. Ed. by G. Hecht, ASM Press Washington D. C. 41 Bellamy, J. EC; and Acres, S. D. 1979. Enterotoxigenic colibacillosis in colostrums- fed calves: pathologic changes. American Journal of Veterinary Research, 40, 1 391 -1 3 9 7. Berger, H.A; Travis, SM. and Welsh, M.J. 1993. Regulation of the cystic fibrosis transmembrane conductance regulator Cl channel by specific protein kinases and protein phosphatases. J Biol Chem 268: 2037-2047. Beme, E. W. and Levy, M. N.1996. Principles of physiology. p 43 7-498. Mosby, St Louis. Bettelheim, K. A. 1994. Biochemical characteristics of Escherichia coli. P3-30 In Gyles (ed). Escherichia coli in domestic animals and humans. CAB International. Wallingfard. UK. Boedeker, E. C. 2005. Vaccines for enterotoxigenic Escherichia coli: current status. Current Opinion in Gastraenteralagy. 1:15-19. Booth, A. J. and Naylor, J. M. 1987. Correction of metabolic acidosis in diarrheal calves by oral administration of electrolyte solution with or without bicarbonate. J Am Vet Med Ass. 191: 62-68. _ Bryce, J. 2005. WHO estimates of the causes of death in children; Lancet, 365: p 1 14 7. Butler, D. G. and Clarke, R. C. 1994. Diarrhea and dysentery in calves. Escherichia coli in domestic animals and humans, p. 91-116. Editor: CL. Gyles. CAB International, Walingford, UK. Bywater, R. J. 1973. Pathophysiological aspects of unidirectional water and sodium transport in calf intestine. Ann. Rech Vet. 4. 125. Carpick, B.W. and Gariepy, J. 1991. Structural characterization of functionally important regionsof Escherichia coli heat-stable enterotoxin STIb. Biochemistry 3 0: 4803-4809. Carpick, B.W. and Gariepy, J .1993. Escherichia coli heat-stable enterotoxin is a long- lived super agonist of guanylin. Infection and Immunity, 61 :4 71 0-4715. Cassels, F. J. and Wolf, M. K.1995. Colonization factors of diarrheagenic E.coli and their intestinal receptors. J Ind Microbiol 33 :2894-2898. Chan, R; Acres, S. D; and Costerton, J. W. 1982. Use of specific antibody to demonstrate glycocalyx, K99 pili, and spatial relationship of K99+ enterotoxigenic Escherichia coli in the ileum of colotrum-fed calves. Infection and immunity, 3 7, I 1 70-1 180. 42 Chang E. B. and Bookstein, C. 2000. Mechanisms of intestinal absorption and secretion: an abbreviated review and update. In: Domschke W, Stoll R, eds. Intestinal mucosa and its diseases-pathaphysiolagy and clinics (F alk Symposium 110). The Netherlands: Kluwer Academic publishers, 1999. Cohen, M. B; Moyer, M. S; Luttrel, M; and Giannella, R. A.1986.The immature rat small intestine exhibits an increased sensitivity and response to Escherichia coli heat-stable enterotoxin. Pediatr Res 20:555-5 60. Cohen, ME; Jensen, N.J; Hawkins, J .A; Mann, E.A; Thompson, MR; Lentze, M.J. and Giannella, RA. 1993. Receptors for Escherichia coli heat-stable enterotoxin in human intestine and in a human intestinal cell line (Caco-2). J. Cell Physio. 1 5 6: 138-144. Currie, M. G; Fok, K. F; Kato, J; Moore, R. J; Hmara, F. K; Duffin, K. L; and Smith, C. E. 1992. Guanylin: An endogenous activator of intestinal guanylate cyclase. Prac Nat Acad Sci USA 89:94 7-951. de Craaf, F. K; and Gaastra, W. 1994. F imbriae of enterotoxigenic Escherichia coli. P. 58-83 InP. K. Klemm (ed.), F imbriae: adhesion, genetics, biogenesis and vaccines. CRC Press, Inc, Boca Ratan, F la. ~ de Sauvage F. J; Camerato, T. R; and Goeddel, D. V. 1991. Primary structure and functional expression of the human receptor for Escherichia coli heat-stable enterotoxin J Biol Chem 266: 1 7912-1 7918. de Sauvage, F. J; Horuk, R; Bennett, G; Quan, C; Burnier, J. P; and Goeddel. D. V. 1992. Characterization of the recombinant human receptor for Escherichia coli heat-stable enterotoxin. J. Biol. Chem. 26 7.64 79-6482. Donnenberg, M. S. 2000. Pathogenic strategies of enteric bacteria. Nature 406: 768- 774. Donowitz, M. and Welsh, M.J. 1986. Ca“ and cyclic AMP in regulation of intestinal Na+, KL and Cl' transport. Annu Rev Physiol 48: 135-150. Eldeib, M. M; Dove, C. R; Parker, C. D; Veum, T. L; Zinn, G. M; and White, A.A.1986. Reversal of the biological activity of Escherichia coli heat-stable enterotoxin by disulfide-reducing agents. Infection and Immunity, 51: 24-30. Evans, D. G; Evans, D. J; and Pierce, N. F. 1973. Differences in response of rabbit small intestine to heat-labile and heat-stable enterotoxins of Escherichia coli cultures. Infection and Immunity 7:8 73-880. 43 Evans, D. G; Evans, D. J; DuPont H. L; Orskve, F; and Orskov, I. 1977. Patterns of loss of enterotoxigenicity by Escherichia coli isolated from adults with diarrhea: suggestive evidence for interrelationship with serotype. Infection and Immunity, I 7, 105. Faris, A; Lindahl, M; and Wadstrom, T. 1980. GM2-like glycoconjugate as possible erythrocyte receptor for CFA/I and K99 haemagglutinins of enterotoxigenic Escherichia coli. FEMS Microbiological Letters, 7, 265. Fasano, A.1999. Cellular microbiology: can we learn cell physiology from microorganisms? Am J Physio] Cell physio] 276: C 765-C 776. Field, M; Graf, L. H. J; Laird, W. J; and Smith, P. L. 1978. Heat-stable enterotoxin of Escherichia Cali: In vitro effects on guanylate cyclase activity, cyclic GMP concentration and ion transport in small intestine. Prac. Nat] Acad Sci USA 75 :2800-2804. F inco, D. R; Duncan, J. R; and Schall, W. D. 1973. Chronic enteric disease and hypoproteinnemia in dogs. Journal of American Veterinary Medical Association, 163, 262-271. Fisher, E. D. 1965. Death in neonatal calf diarrhea. British VetJ. 121: 132-138. Fisher, E. D; and McEwan, A. D. 1967. Death in neonatal calves diarrhea. Part II: The role of oxygen and potassium. British Vet J 123: 4- 7. Fisher, E. D; and Martinez, A. A. 1975 Studies of neonatal calf diarrhea British Veterinary Journal, 131, 190. F ollett, E. AC; and Gordon, J. 1980. An electron microscope study of Vibrio flagella. J Gen Microbial 32: 235-239. Forte, L. R; and Currie, M. G. 1995. Guanylin: A peptide regulator of epithelial transport. FASEB J 9:643-650. Forte, L. R; Krause, W. J; and Freeman, R. H. 1989. Escherichia coli heat-stable enterotoxins receptors: Localization in opossum kidney, intestine and testis. Am J Physiol 275: F 874-881. Frantz, J. C; Jaso-Friedman, L; and Robertson, D. C. 1984 Binding of Escherichia coli heat-stable enterotoxin to rat intestinal cells and brush border membranes. Infect Immun. F eb;43 (2) .' 622-3 0. 44 French, P. J; Bijman, J; Edixhoven, M; Vaandrager; A. B; Scholte, B. J; Lohman, S. M; Naim, A. C. and de Jonge, H. R. 1995. Isotype-specific activation of cystic fibrosis transmembrane conductance regulator-chloride channels by cGMP- dependent protein kinase II. J. Biol Chem 270: 26626-26631. Garbers, D. L.1999. The guanylyl cyclase receptors. Methods 19: 477-484. Gariepy, J; Lane, A; Freeman, F; Wilbur, D; Robien, W; Schoolnik, G. K; and Jardetzky, O. 1986. Structure of the toxic domain of the Escherichia coli heat- stable enterotoxin STI. Biochemistry 25: 7854- 7866. Garpiey, J; Judd A. K; and Schoolnik G. K. 1987. Importance of disulfide bridges in the structure and activity of Escherichia coli enterotoxin STIb. Prac. Natl. Acd. Sci. USA 84: 8907-89011. Giannella, R. A; Luttrell, M; and Thompspon, M .1983 Binding of Escherichia coli heat-stable enterotoxin to receptors on rat intestinal cells. American Journal of Physiology. 245: 6492-0498. Giannella, R. A. 1995. Escherichia coli heat-stable enterotoxins, guanylins and their receptors: what are they and what do they do? J.Lab. Clin. Med. 125: 1 73- 181. ' Giannella, R. A. and Elizabeth, A. M. 2003. E. coli heat stable enterotoxin and guanylyl cyclase C: New functions and suspected actions. Trans Amr Clin Climato Ass. 1 I 4, 6 7-85. Golin—Bisello, F; Bradbury, N. and Ameen, N. 2005. STa and cGMP stimulate CF TR translocation to the surface of villus enterocytes in rat jejunum and is regulated by protein kinase G. Am J Physiol Cell Physiol 289: 708- 716. Gorbach, S. L; Kaen, B. H; Evans, D. G; and Bessudo, D. 1975. Travelers’ diarrhea and toxigenic E. coli. N Engl J Med 1975; 292: 933-93 6. Gray, W. R; Luque, A; Olivera, B. M; Barrett J; and Cruz; L. J .1981. Peptide toxins from Conus geographyus venom J. Biol. Chem. 156: 4 734-4 7. Greenberg, R. N; Hill, M; Krytzer, J; Krause, W. J; Eber, S. L; Hamra, F. K. and Forte, L. R.1997. Comparison effects of uroguanylin, guanylin, and Escherichia coli heat-stable enterotoxin STa in mouse intestine and kidney: evidence that uroguanylin is an intestinal natriuretic hormone. J. Investig. Med. 45: 276-282. Groutides, C. P; and Michell, A. R. 1990. Change in plasma composition in calves surviving or dying from diarrhea. Britich Vet J 146:205-21 0. 45 Grove-White D. H. 1996. Pathophysiology and treatment of metabolic acidosis in diarrheic calf. Proceeding of XIX Buiatrics Congress of Vet Association (BC VA) Edinburg, Calf Diseases 1 : 102-107. Guarino, A; Giannella, R. and Thompson, M. R. 1989. Citrobacter freundii produces an 18-amino acid heat-stable enterotoxin identical to the 18-amino acid Escherichia coli heat-stable enterotoxin (STIa). Infect. Immun. 5 7: 649-652. Gyles, C. L. 1971. Heat-labile and heat-stable forms of enterotoxin from Escherichia coli strains enteropathogenic for pigs. Ann NY Acad Sci. 1 76:314-322. Gyles, C. L. 1996. Escherichia coli in domestic animals and humans. p 31-72 CAB International. Wallingfard. UK. Hadad, J. J. and Gyles, C. L. 1982a. Scanning and transmission electron microscopic study of the small intestine of colostrums-fed calves infected with selected strains of Escherichia coli. American Journal of Veterinary Research, 43, 41 - 49. Hadad, J. J. and Gyles, C. L.1982b. The role of K antigens of enteropathogenic Escherichia coli in colonization of the small intestine of claves. Canadian Journal of Comparative Medicine, 46, 21 -26. Hakki, S; Crane, M; Hugues, M; O'Hanley, P; and Waldman, S. A. 1993. Solubilization and characterization of functionally coupled Escherichia coli heat-stable toxin receptors and particulate guanylate cyclase associated with the cytoskeleton compartment of intestinal membranes. Int J Biochem 25 : 557-566. Hall, G. A; Jones, P. W; and Morgen, J. H. 1992. Calf Diarrhea. In Bovine Medicine Diseases and Husbandry of Cattle by Andrews A. H. 1” Ed Oxford Blackwell Scientific Pp. 154-180. Hasegawa, M. and Shimonishi, Y. 2005. Recognition and signal transduction mechanism of Escherichia coli heat-stable enterotoxin and its receptor, guanylyl cyclase C. J. Peptide Res, 65 .' 261-2 71. Hasegawa, M; Hidaka, Y; Wada, A; Hirayama, T; and Shimonishi, Y. 1999 The relevance of N-linked glycosylation to the binding of a ligand to guanylate cyclase C. Eur J Biochem 263: 338-346. Hughes, J. M; Murad, F; Chang, B; and Guerrant, R. L.1978. Role of cyclic GMP in the action of heat-stable enterotoxin of Escherichia coli. Nature (Land) 271: 755- 756. 46 Hugues, M; and Waldman, S. A. 1992. Molecular mechanisms underlying activation of particulate guanylyl cyclase and GMP accumulation induced by Escherichia coli heat-stable enterotoxin. Biochem Life Science Adv 10:103- 1 12. Hugues, M; Crane, M. R; Thomas, B. R; Robertson, D; Gazzano, H’ O'Hanley, P; and Waldman S. A. 1992. Affinity purification of functional receptors for Escherichia coli heat-stable enterotoxin from rat intestine. Biochemistry. Jan 14:31:12-16. ' Isaacson, R. E. 1980. Factors affecting expression of Escherichia coli K99 pilus. Infection and Immunity, 28, 190-194. Ivens, K; Gazzano, H; O'Hanley, P. and Waldman, S. A. 1990. Heterogeneity of intestinal receptors for Escherichia coli heat-stable enterotoxin. Infect Immun 58: I81 7-1820. Jacks, T. M. and Wu, B. J. 1974. Biochemical properties of Escherichia coli low molecular weight heat-stable enterotoxin. Infection and Immunity 9342-34 7. Jaso-Friedman, L; Drefus, L. A; Whipp, S. C; and Robertson, D. C. 1992. Effect of age on activation of porcine guanylate cyclase and binding of Escherichia coli heat-stable enterotoxin (STa) to porcine intestinal cells and brush border membranes. Am J Vet Res 53 :2251 -2258. Katwa, L. C; and White, A. A. 1992. Presence of functional receptors for Escherichia coli heat-stable enterotoxin in the gastrointestinal tract of chicken. Infect immune 60:3546—3551. Koo, D; Maloney, K; and Tauxe, R. 1996. Epidemiology of diarrhea] diseases outbreaks on cruise ships, 1986 through 1993. JAMA, 275: 545-54 7. Krause, W. J; Cullingford, G. L; Freeman, R. H; Ber, S. L; Richardson, K. C; F ok, K. F; Currie, M. G; and Forte LR 1994. Distribution of heat-stable enterotoxin/guanylin receptors in the intestinal tract of manand other mammals. J. Anat 184:404-417. Laohachai, K. N; Bahadi, R; Hardo, M. B; Hardo, P. G; and Kourie, J. I. 2003. The role of bacterial and non-bacterial toxins in the induction of changes in membrane transport: implication for diarrhea. Toxican 42: 687- 707. Levine, M. M. 1987. Escherichia coli that cause diarrhea: Enterotoxigenic, enteropathogenic, enteroinvasive, enterohemorrhagic and enterodherent. Journal of Infectious Diseases 155: 33 7-389. 47 Levine, M. M. Kaper, J. B; Black, R. E; and Clements, M. L. 1983. New knowledge on pathogenesis of bacterial enteric infections as applied to vaccine development. Microbiological Reviews 4 7:51 0-5 5 0. Lewis, L. D; and Phillips, R. W. 1972. Water and electrolyte losses in neonatal calves with acute diarrhea: A complete balance study. Cornell Vet. 62: 596-60 7. Lior, H. 1996. Classification of Escherichia coli. pp. 31-72. In C.L. Gyles (ed). Escherichia coli in domestic animals and humans. CAB International. Wallingford. UK. Lohmann, S. M; Vaandrager, A. B; Smolenski A, Walter, U. and De Jonge H. R. 1997. Distinct and specific functions of cGMP- dependent kinase. Trends Biochem Sci 22: 307-312. Lowe, D. G; Camerato, T. R. and Goeddel, D. V. 1990a. cDNA sequence of the human atrial natriuretic peptide clearance receptor. Nucleic Acids Res 18: 3412 Lowe, D. G; Klisak , I; Sparkes, R. S; Mohandas, T. and Goeddel, D. V.1990b. Chromosomal distribution of three members of the human natriuretic peptide receptor/guanylyl cyclase gene family. Genamics. Lucas, K. A; Pitari, G. M; Kazerounian, S; Ruiz-Stewart, 1; Park, J; Schulz, S; Chepenik, K. P; and Waldman, S. A. 2000. Guanylyl Cyclase and Signaling by Cyclic GMP. Pharmaca] Rev 52: 3 75-413. Lucas, M. L. 2001. A reconsideration of the evidence for Escherichia coli STa (heat- stable) enterotoxin-driven fluid secretion: a new view of STa action and a new paradigm for fluid absorption. Journal of Applied Microbiology 90: 7-26. Lucas, M. L; Thom, M. M. M; Bradley, J. M; O’Reilly, M and Nelson, Y. B. 2005. Escherichia coli heat-stable enterotoxin and the upper small intestine: Lack of evidence in vivo for net fluid secretion. J Membrane Bio]. 206: 29-42. Mann, E. A; Jump, M.L; Wu, J; Yee, B. and Giannella, R. A. 1997. Mice lacking the guanylyl cyclase C receptor are resistant to STa-induced intestinal secretion. Biochem Biophys Res Commun 239:463-466. Montrose, M. H; Keely, S. J. and Barrett, K. E. 1999. Secretion and absorption. small intestine and colon, p. 320- 355. In T. Yarnada, DH Alpers, L. Latine, C. Owyang, and D. W. Powell (ed) Textbook of Gastraenteralogy, 3lrd ed Lippincatt, Williams and Wilkins, Philadelphia, PA. Moon, H.W. 1994. Pathophysiology of viral diarrhea. In: Viral infections. of the gastrointestinal tract, p. 27-52. editor: A.Z. Kapikian, CIP, New York. 48 Moon, H. W. 1974. Pathogenesis of enteric diseases caused by Escherichia coli. In: Advances in veterinary science and comparative medicine, pp. 179-211. Editors: C.A. Brandly and CE. Cornelius. Academic Press, New York. Moon, H.W. 1978. Mechanisms in the pathogenesis of diarrhea: A review. Journal of American Veterinary Medicine Association, 172, 443-448. Moon, H. W; Nagy, B; Isaacson, R. E; and Orskov, I. 1977. Occurrence of K99 antigen on Escherichia coli isolated from pigs and colonization of pig ileum by K99+ enterotoxigenic E. coli from calves and pigs. Infection and Immunity 15: 614-620. Morris, J. A; Thome, C. J; Wells, G. A; Scott, A. C. and Sojka, W. J. 1983. The production of F 41 fimbriae by piglet strain of enterotoxigenic Escherichia coli that lack K88, K99 and 987P fimbriae. Journal of Genreal Microbiology, 129, 2753. Murray, C. J. L.1997. Global mortality, disability, and the contribution of risk factors: Global Burden of Disease Study; Lancet,349: 143 6. Nagy, B. and F ekete, P. Z. 1999. Enterotoxigenic Escherichia coli in farm animals. Vet Res. 30 (2-3) :25 9-84. NAHMS 1994. USDA Part III: Beef cow/calf health and health managements. US. Dept of Agriculture Animal and Plant Inspection Service, NAHMS, Veterinary Services, Fort Collins, Colorado, 37. NAHMS 1996. USDA Part 11: Changes in the US. Dairy Industry. 1991-1996. US. Dept of Agriculture Animal and Plant Inspection Service, NAHMS Veterinary Services, Fort Collins, Colorado. 17-21. Nataro, J. P. and Kaper, J. B. 1998. Diarrheogenic Escherichia coli. Clin Microbialo Rev 11: 142-301. Okamoto, K; Yukitake, J; Kawarnoto, Y; and Miyama, A. 1987. Substitution of cysteine residues of Escherichia coli heat-stable enterotoxin by oligonucleotide-directed mutagenesis. Infect. Immun. 55: 2121-2125. Oliveira, L; Minion, P; Viegas-Pequignot, E; Rozet, J. M; Dollfus, H. and Pittler, S. J. 1994. Human retinal guanylate cyclase (GUC2D) maps to chromosome 17p13.1. Genomics 22: 478-481. Orskov, I; Orskov, F; Jann, B; and Jann, K. 1977. Serology, chemistry, and genetics of O and K antigens of Escherichia coli. Bacteriological Reviews, 41, 667. 49 Osaki, H; Sato, T; Kubota, H; Hata, Y; Katsube, Y; and Shimonishi, Y.1991. Molecular structure of the toxic domain of heat-stable produced by a pathogenic strain of Escherichia coli. J.Bial. Chem. 266:5 934-5 94 1 . Pfeifer, A; Aszodi, A; Seidler, U; Ruth, P; Hofinann, F; and F assler, R. 1996. Intestinal secretory defects and dwarfism in mice-lacking cGMP-dependent kinase II. Science (Wash DC) 274:2082-2086. Phillips, R.W. and Knox, K.L.l969. Diarrheie acidosis in calves. J Comp Lab Med 3:1 -3. Pitek, R; Zalewska, B; Bury, K; and Kur, J. 2004. The chaperone-usher pathway of bacterial adhesin biogenesis -from molecular mechanism to strategies of anti- bacterial prevention and modern vaccine design, International Review Conference on Biotechnology, Vienna,Austria. Planet, P. J; Figurski, D. H; and DeSalle 2006. Function, evolution and classification of macromolecular transport systems. In Evolution of Microbial pathogen, Edited by Seifert HS. and DiRita V.J. ASM Press, Washington DC. Quinton P. M.1990. Cystic fibrosis: A disease in electrolyte transport. FASEB J 4: 2709-2 71 7. Rao, M.C; Guandalini, S; Smith, P. L; and Field, M. 1980. Mode of action of heat- stable Escherichia coli enterotoxin. Biochemical et Biophysica, 632, 35-46. Rao, M.C; Field, M; Orellana, S.A; Robertson, DC. and Giannella, RA. 1981. Comparison of the biological actions of three purified heat-stable enterotoxins: effects on ion transport and guanylate cyclase activity in rabbit ileum in vitro. Infect immune 33: 1 6 7. Rasheed, J. K; Guzman-Verduzco, L-M; and Kupersztoch, Y. M. 1990. Two precursors of the heat-stable enterotoxin of Escherichia coli: evidence of extracellular processing. Mol. Microbiol. 4: 265-273. Runder, X. L., Nicehitta, C. and Almenoff, J. S. 1996. Biogenesis, cellular localization and functional activation of heat-stable enterotoxin receptors(Guanylyl cyclase C). Biochemistry 35: 10680-10686. Runnels, P. L; Moon, H. W. and Schneider, R. A.1980. Development of resistance with host age to adhesion of K99+ Escherichia coli to isolated intestinal epithelial cells. Infection and Immunity 28: 298-3 00. Saeed, A. M; Srianganathan, N; Cosand, W; and Burger, D. 1983. Purification and characterization of heat-stable enterotoxin from bovine enterotoxigenic Escherichia coli. Infection and Immunity, 40: 701-701. 50 Saeed, A. M; Magnuson, N. S; Sriranganathan, N; Burger, D; Cosand, W. 1984. Molecular homogeneity of heat-stable enterotoxins produced by bovine enterotoxigenic Escherichia coli. Infect Immun 45:242-247. Saeed, M. A; and Greenberg, R.N. 1985a. Preparative purification of Escherichia coli Heat-stable Enterotoxin. Anal. Bia.151: 431-43 7. Saeed, A. M; Magnuson, N. S; Gay, C. C. and Greenberg, R. N. 1985b. Characterization of heat-stable enterotoxin from a hypertoxigenic Escherichia coli for cattle. Microbiology and Therapy, 15: 221 -229. Salyers, A. A; and Witt, D. D.1994. Bacterial pathogenesis: A molecular approach, ASM Press, Washington DC. Sato,T; and Shimonishi, Y. 2004 Structural features of Escherichia coli heat-stable enterotoxin that activates membrane-associated guanylyl cyclase. J. Peptide Res 63:200-206. Sato, T., Ozaki, H; Kitagawa, Y; Katsube, Y; and Shimonishi, Y. 1994. Structural characteristics for biological activity of heat-stable enterotoxin produced by enterotoxigenic Escherichia coli: X-ray crystallography of weakly toxic and nontoxic analogs. Biochemisty 33: 8641-8650. Savarino, S.J; Fasano, A; Watson, J; Martin, B. M; Levine, M. M. Guandalini, S; and Guerry, P. 1993. Enteroaggregative Escherichia coli heat-stable enterotoxin 1 represents another subfamily of E. coli heat-stable toxin. Proc. Natl. Acad Sci. USA 90:3 093 -3 09 7. Schulz, S; Green, C. K; Yuen, P. S. T; and Garbers, D. L. 1990. Guanylyl cyclase is a heat-stable enterotoxin receptor. Cell 63: 941-948. Schulz, S; Lopez, M. J; Kuhn, M; and Garbers D. L. 1997. Disruption of the guanylyl cyclase-C gene leads to a paradoxical phenotype of viable but heat-stable enterotoxin-resistant mice. J C [in Invest 100: 1590-1595. Sears, C. L; and Kaper. J. B.1996. Enteric bacterial toxins: mechanisms of action and linkage to intestinal secretion. Microbial. Rev. 60:167-215. Seifert, H. S. and DiRita, V.J. 2006. Evolution of Microbial pathogen, ASM Press, Washington DC. Selveraj, N. G; Prasad, R; Goldstein, L. and Rao, M. C. 2000. Evidence for the presence of cGMP-dependent protein kinase-II in human distal colon and in T84, the colonic cell line. Biochem. Biophys. Acta 1498: 32-43. 51 Shimonishi, Y; Hidaka, Y, Koizumi, M; Hane, M; Arimoto, S; Takeda, T;, Miwatani, T; and Takeda, Y. 1987. Mode of disulfide bond formation of heat-stable enterotoxin (STh) produced by human strain of enterotoxigenic Esherichia coli. FEBS Lett. 215: 165-1 70. Shlatz L. J; Kimberg, D. V. and Cattieu, K. A. 1979. Phosphorylation of specific rat intestinal microvillus and basal-lateral membrane proteins by cyclic mucleotides. Gastaenterology 76: 293. Smith, H. W. 1965. The development of flora of the alimentary tract in young animals. Journal of Pathological Bacteriology, 90, 495. Smith, H.W; and Huggins, M. B. 1978. The influence of plasmid-deterrnined and other characteristics of enteropathogenic Escherichia coli on their ability to proliferate in the alimentary tracts of piglets, claves and lambs. Journal of Medical Biochemistry, 11, 471. Snodgrass, D. R; Terzolo, H. R; Campbell, D; Sherwood, I; Menzies, J. D; and Synge, B. A. 1986. Aetiology of diarrhoea in young calves. Vet Record, 1 19:31-34. So, M; and McCarthy B. J. 1980. Nucleotide sequence of the bacterial transposon Tn1681 encoding a heat-stable (ST) toxin and its identification in enterotoxigenic Escherichia coli strains. Prac Nat] Acad Sci USA 77:4 01 1- 4015. So, M; Heffron, F; and McCarthy B. J. 1979. The Ecali gene encoding a heat-stable toxin is a bacterial transposon flanked by inverted repeats of IS 1. Nature 277: 453-45 6. So, M; R. Atchison, Falkow S, Moseley and McCarthy B. J. 1981. A study of the dissemination of Tn1681: a bacterial transposon encoding a heat-stable enterotoxin among genetic Escherichia coli isolates. Cod Spring Harbor Symp. Quant. Biol. 45: (Pt. I ).' 53-58. Stine, CC; and Nataro, J. P. 2006. The evolution of bacterial toxins. In: Evolution of Microbial pathogen, Eited by HS. Seifert and VJ. DiRita 2006 ASM Press, Washington DC. Svennerholm, A. Steele D. 2004. Progress in enteric vaccine development. Best Practice & Research Clinical Gastroenteralog, I8: 421 — 445. Takao, T; T.Hitouji, T; Aimoto, A; Shimonishi, Y; Hara, S; Takeda, T; Takeda, Y; and Miwatani. T. 1983. Amino acid sequence of a heat-stable enterotoxin isolated from enterotoxigenic E. coli strain 18D. FEBS Lett. 152: 1-5. 52 Takao, T; Tominaga, T; Shimonishi, Y; Hara, S; Inoue, T; and Miyama. A.1985. Isolation, primary structure and synthesis of heat-stable enterotoxin produced by Yersinia enteracalitica. Eur. J. Biach. I52: I99-206. Takeda, T; Nair, G. B; Suzuki, K; Zhe, H. X; Yokoo, Y; Hemelhof, W; Butzler, J. B; Takeda, Y; and Shimonishi, Y. 1993. Epitope mapping and characterization of antigenic determinants of heat-stable enterotoxin (STh) of enterotoxigenic Escherichia coli by using monoclonal antibodies. Infection and Immunity 61 :289-294. Takeda, Y; Yamasaki, S; Hirayama, T; and Shimonishi, Y. 1991. Heat-stable enterotoxins produced by enteric bacteria. In: Molecular pathogenesis of gastrointestinal Infections (F ederatian European Microbiological Societies Symposium Series, No 58) (Wadstrom T, Makela PH, Svennerhalm AM and Wolf-Watz H., eds) pp 125-138. Plenum Press, NY Thompson, M. R. 1987. Escherichia coli heat-stable enterotoxins and their receptors. Pathal Immunapatha] Res 6:1. 3-1 I 6 Thompson, M.R., and Giannella, R. A. 1990. Different cross linking agents identify distinctly different putative Escherichia coli heat-stable enterotoxin rat intestinal cell receptor proteins. J. Receptor Res. 10: 97-11 7. Urbanski, R; Carrithers, S. L. and Waldman, S. A. 1995. Intemalization of E. coli ST mediated by guanylyl cyclase C in T84 human colon carcinoma cells. Biachim Biophys Acta 1245: 29-3 6. USDA, NASS&APHIS 1997. Cattle and Calves Death Loss. Vaandrager, A. B; Schulz, S; De Jonge, H. R; and Garbers, D. L. 1993. Guanylyl cyclase C is an N-linked glycoprotein receptor that accounts for multiple heat- stable enterotoxin-binding proteins in the intestine. J Biol Chem 268: 21 74- 21 79. Vaandrager, A. B. and de Jonge, H. R. 1994. Effect of cyclic GMP on intestinal transport. Adv Pharmaca] 26:253-283. Vaandrager, A. B. Bot, A. G; and de Jonge, H. R. 1997. Guanosine 3',5'-cyclic monophosphate-dependent protein kinase II mediates heat-stable enterotoxin- provoked chloride secretion in rat intestine. Gastraenteralagy 112: 437-443. Vaandrager, A. B; Bot, A. G; Ruth, P; Pfeifer, A; Hofrnann, F. and de Jonge, H. R. 2000. Differential role of cyclic GMP-dependent protein kinase II in ion transport in murine small intestine and colon. Gastraenterolagy 118: 108- I 1 4. 53 Vaandrager, A. B. 2002. Structure and function of the heat-stable enterotoxin receptors/guanylyl cyclase C. Molecular and Cellular Biochemistry 230: 73- 83. Walker, R.I; Steele, D; Aguado, T. and Ad Hoc ETEC Technical Expert Committee. 2007. Analysis of strategies to successfully vaccinate infants in developing countries against ETEC disease. Vaccine 25, 2545-2566. World Health Organization 1990. Tropical diseases. ASM News, 1990, 56: 358-359. Yamanaka, H; Kameyama, M; Baba, T; Fujii, Y; and Okamoto, K. 1994. Maturation pathway of Escherichia coli heat-stable enterotoxin 1. Requirement of DsbA for disulfide bond formation. J. Bacteriol. 1 79: 2906-2913. Yamasaki, S; Sato, T; Hidaka, Y; Ozaki, H; Ito, H; Hirayama, T; Takeda, T; Sugimura, T; Tai, A; and Shimonishi, Y. 1990. Structural-activity relation ship of Escherichia coli heat-stable enterotoxin: role of Ala residue at position 14 in toxin-receptor interaction. Bulletin of Chemistry Society of Japan, 63, 2063. Yang, R.B; F iille H. J; and Garbers, D.L. 1996. Chromosomal localization and genomic organization of genes encoding guanylyl cyclase receptors expressed in olfactory sensory neurons and retina. Genomics. 31: 3 6 7-3 72. Yoshimura, S; Ikemura, Y; Watanabe, H; Aimoto, S; himonishi, Y; Hara, S; Takeda, T; Miwatani, T; and Takeda, Y. 1985. Essential structure for enterotoxigenic activity of heat-stable enterotoxin produced by enterotoxigenic Escherichia coli. FEBS Lett. 181: 138-142. Yoshimura, S., Hidaka, Y; Aimoto, Shimonishi, S; Takeda T; Miwatani, T; and Takeda, Y. 1987. Structural-activity relationship of a heat-stable enterotoxin produced by Yersinia enteracalitica. Bull Chem Soc Jpn 60:2481-2489. Yoshimura, S., Takao, T; Shimonishi, Y; Hara, S; Arita, M; Takeda, T; Irnaishi, H; Honda, T; and Miwatani, T. 1986. A heat-stable enterotoxin of Vibrio cholerae non-OI: Chemical synthesis, and biological and physicochemical properties. Biopalymers 25 (Suppl. ): S69-S83. Yoshino K, Huang; X; Miyachi, M; Hong, Y-M; Takao, T; Nakao, H; Takeda, T; and Shimonishi, Y. 1994. Amino acid sequence of a novel heat-stable enterotoxin produced by a yst gene-negative strain of Yersinia enterocalitica. Lett Pept Sci 1: 95-105. Yun, C. H. C; Tse, C. M; Nath, S. K; Levine, S. A; Brant, S. R. and Donowitz, M. 1995. Mammalian Na+/H+ exchanger gene family: structure and function studies. Am J Physiol 269: G] -GI 1. 54 Zhang, W; Mannan I; Schulz, S; Parkinson . J; Alekseev, A. E; Gomez L. A; Terzic; A. and Waldman SA. 1999. Interruption of transmembrane signaling as a novel antisecretory strategy to treat enterotoxigenic diarrhea. FASEB J 13: 913-922. 55 CHAPTER III PURIFICATION AND CHARACTERIZATION OF HEAT-STABLE ENTEROTOXIN (STa) OF ENTEROTOIXIGENIC Escherichia coli Introduction Enterotoxigenic strains of Escherichia coli (ETEC) have been implicated as causative agents of diarrhea in human infants (Guerrant et al. 975 and Sack et al. 1975), diarrhea in neonatal domestic animals (Smith and Halls, 1967 and Whipp et al. 1975) and in “travelers” diarrhea (Jiang et al. 2000). ETEC strains are capable of producing at least two distinct forms of extracellular enterotoxins. One form is called heat-labile enterotoxin (LT), which is structurally and functionally similar to cholera toxin (CT) (Gyles 1971 and Evans et al. 1973),. and the other form is heat-stable enterotoxin (STa), which is a low-molecular-weight (< 2kD) peptide that is non- immunogenic and is not neutralized by antibodies specific to CT (Gyles 1971 and Evans et al. 1973 and Naline et al. 1974). Seventy five percent of tested ETEC strains have been found to produce STa alone or in combination of LT (Wolf, 1997). Therefore, STa is considered to be an important virulence determinant in enterotoxin- mediated diarrheal diseases caused by E. coli (Sears & Kaper, 1996 and Giannella & Elizabeth, 2003). The purification and characterization of STa have been complicated by the complexity of various growth media used for toxin production and by laborious purification procedures such as salt precipitation, ultrafiltration, ion exchange and molecular sieve chromatography. These procedures are time consuming and have a 56 relatively low recovery yield of homogenous STa (Staples et al. 1980, Saeed et a1. 1983, and Saeed & Greenberg 1985). In this study, we adopted the growth medium and purification methods reported by Staples et al. (1980) and modified by Saeed et al. (1983) and Saeed and Greenberg (1985) under controlled growth conditions using a 36 L Bellco bioreactor. Material and methods Animals Swiss Webster Mice: A group of 20 Swiss-Webster (fifteen females and five males) was used to establish a colony as a source of suckling mice for STa bioassay. Exhausted females and males were continuously replaced with younger animals to ensure production efficiency of infant mice litters by the colony. Reagents All reagents were obtained from commercial sources and were of analytical grade. Mobile phases used for purification of STa include HPLC-grade methanol, triflouroacetic acid, as well as the other chemical ingredients listed under this section. Verifying the ETEC K99+ Strain Basic PCR protocol described by Olsvick and Strockbine (1993) and Salvadori et al. (2003) was used to detect the STa gene and verify the strain as STa- producing E. coli. Bacteria] Strains: An ETEC strain was isolated from a clinically diarrheic neonatal calf and was provided by A. M. Saeed (Molecular Epidemiology Laboratory, National Food Safety Toxicology Center (NFST), Michigan State University (MSU), 57 East Lansing MI. A control strain (K-12 E. coli) was kindly obtained from the Bacterial Evolution Laboratory, NF ST, MSU, East Lansing- MI. DNA Extraction (Template) by boiling lysis: ETEC and K-12 strains were grown on Tripticase Soy Agar slants overnight at 37°C. A uniform bacterial colony from both strains was taken and suspended in 1 ml sterile Milli Q water and boiled for 10 min, then left in ice for 5 min, followed by eentrifugation at 13,000 rpm for 4 min. The supernatant was taken and kept at -20 °C until use (Holmes and Quigley,198 l ). Primer selection and preparation: Two different sizes of STa primer, 244bp and 127bp (Table 3), were obtained from Integrated DNA Technology Inc. (Coralville, IA). Both primers were prepared according to the manufacturer’s instructions. Table 3. PCR primers used to detect STa gene Primer Sequence (5-3) Base Reference pair STa 5’- TCC GTG AAA CAA CAT GAC GG-3’ 244 Salvadori et al. 5’- ATA ACA TCC AGC ACA GGC AG-3’ 2003 STI 5’-TTA ATA GCA CCC GGT ACA AGC AGG-3’ 127 Olsvick, and 5’-CTT GAC TCT TCA AAA GAG AAA ATT AC-3’ Strockbine 1993 PCR program: PCR running conditions for detection of the STa gene are presented in Table 4 using PCT-100 Programmable Thermal Controller (MJ Research Inc). Table 4. PCR running conditions for detection of STa gene Sig) Temperature (°C) Time (min) Pre-denature 95 5 Denature 95 1 Annealing 60 1 Extension 72 1 Final extension 72 10 Storage 4 24 hours Number of PCR cycles 29 before storage 58 PCR reaction: The PCR reaction was performed as described in table 5 using Fisher exACTGene Complete PCR kit. Table 5. PCR reaction for detection of STa gene Component Amount/sample # of Amount/two rxns (11') Samples Volume (pl) 10x buffer A 2 2.5 5 dNTP (1 OmM) Q4 25 1 Sta-thouM) 0.4 2.5 1 Sta-R (20 PM) 0.4 2.5 1 25 mM MgClz 0 0 0 Fisher Taq 0.1 2.5 1 polymerase Ultrapure water 14.7 2. 5 36.75 Total volume 20 45 Template 2 Agarose Gel Electrophoresis Analysis of PCR Products: The analysis of the PCR products was performed in 2% agarose gel electrophoresis using the Horizontal Gel Electrophoresis System, Life Technology (Cat # 11068-012). Briefly, two percent of agarose was prepared (1.5 gm/75 m1 leAE electrophoretic sequence grade) and ethidium bromide was added at concentration 3 ul/ 50 ml. The reagent was poured into the electrophoretic chamber and filled with leAE. Five volumes of PCR product were mixed with 1 volume of gel loading buffer and loaded into the wells along with a 1.5 kb ladder. The agarose gel was left to run at the appropriate voltage (100-160 volts) for 30-45 min and then examined under UV light. Purification and Characterization of E. coli STa STa was purified according to the protocol reported by Staples et al. (1980) and modified by Saeed et al. (1983) and Saeed and Greenberg (1985). 59 Seed culture and frozen stock of E T E C preparation Casamino acid-yeast extract-salts (CAYE) seed culture was used for optimal growth of the ETEC strain (Giannella, 1976). The ETEC strain was grown on 500 m1 of CAYE, incubated at 39 0C for 24 hours on a rotary shaker at 120 rpm, then mixed with glycerol at a final concentration of 15%, then aliquoted into 10 ml samples and frozen at -80 °C (frozen stock). Batch medium (Asparagine salt medium) and growth conditions (Staples et al. 1980): Table 6. Composition of optimal minimal medium for STa production (g/L) NaCl 2.52 Na2SO4 0.14 Na acetate 10.00 MgSO4 0.05 K2HPO4.3H20 8.12 Man 1% 0.5 ml Asparagine 5.00 F eCl3 1% 0.5 ml We used the Asparagine-salt medium (ASM) reported by Staples et a1. (1980) that was compared with other grth media by Saeed et a1. (1983) and found to offer several advantages, including high level of STa production along with minimal contaminating proteins that facilitated the STa-purification process. Each batch consisted of 30 liters of culture-innoculated ASM grown in a 36L omni vessel under different pH conditions (7.4, 8 and 8.6) using a Bellco bioreactor (Bellco Glass Inc. Vineland NJ). Preeulture was prepared by inoculating 10 ml of frozen stock of ETEC into 1L of CAYE broth and was incubated at 39 °C for 24 hours on a rotary shaker at 120 rpm. The preculture medium was then transferred into 30 liters of batch medium and kept at 39°C under continuous agitation at 120 rpm, aeration and oxygenation 60 were at a rate of 5 L/min and 600 ml/min, respectively, through a sintered metal dispersion ring. Foam, speed of agitation, temperature and P02 were controlled using respective Bellco control modules. Samples were taken every two hours to determine the growth kinetics under various pH levels. Preparation of cell free filtrate After 24 hours of incubation, the growth medium was immediately filtered by tangential flow filtration through a 02-11 cassette in Millipore Pellicon System (Millipore Crop, Bedford, MA). Cell free filtrate was kept on ice throughout the time of filtration to minimize bacterial grth and enzymatic activity. Samples from the cell-free filtrate were collected for determination of total protein and STa content using suckling mouse assay. Amberlite XAD-2 Batch Adsorption Chromatograph Cell free filtrate was desalted and the hydrophobic STa was concentrated using Amberlite XAD-2 batch adsorption chromatography. Amberlite XAD-2 resin was first washed extensively with purified water to remove any preservative and powdery contaminants. Then 500g were suspended into 15 L of cell free filtrate in a 20- L carboy and kept overnight at 4 °C under gentle stirring. Resin was poured from the carboy into a 40 cm long glass column and washed with 5 L of Milli Q water. The contaminants loosely bound to the resin were eluted with 1L of 1% acetic acid in 20% methanol/water (v/v). A stepwise elution system was applied to elute the STa starting with 1L of 1% acetic acid in 80% methanol/water (v/v) followed by 1L of 1% acetic acid in 99% methanol/water (v/v) and finally 1 L of 50% acetone/water (v/v). The last three fractions were pooled and concentrated by flash evaporation & freeze-drying. 61 The resin was degassed for 5 minutes after each solvent was added to drain completely before further addition of solvent. Samples were collected for determination of the total protein and testing for STa biological activity in suckling mouse. Acetone Fractionation Lyophilized crude STa was dissolved in 20 ml of 25% of acetic acid. Acetone was added to bring the final volume to 100-150 ml. After standing 1 hour at 4 °C, the sample was centrifuged at 10,000g for 30 minutes at 4 oC. The supernatant fraction was evaporated to remove the acetone and was then freeze-dried. Samples were collected for protein determination and STa biological activity. Reversed-Phase Batch Adsorption Chromatography (MCI-gel) An intermediate purification step was applied to the acetone STa-rich fraction to achieve a further level of STa purity. The lyophilized crude STa was solubilized in 100 ml of 0.1% of 20% HPLC-grade methanol. To this solution, 100g of Reverse- Phase Methacrylate Adsorbent Polymer Resin (340 °A 30 um Mitsubishi Chemical Corporation, Cat # CHP2MGY-01L) was slowly added under gentle mixing and the slurry was kept at 4 °C for 2 hours under gentle shaking. The slurry was poured into a 10-mm-i.d.x 25-cm-long glass column. The column was washed with 300ml of 0.1%TFA/1120 (v/v). Stepwise elution of the proteins was performed with 100 ml of 0.1%TFA of 20, 40, 60, 80 &100% MeOH (v/v). Fractions were collected separately from each elution step. The methanol and TFA were evaporated and the residues were tested for protein and STa biological activity. 62 Preparative Reverse-Phase High Performance Liquid Chromatography (RP- HPLC) RP-HPLC was performed on Waters Associate Liquid Chromatography System equipped with multi-solvent delivery pumps, an automated gradient programmer 6OOS controller, Model 486 tunable absorbance detector using 7 pm, 300 A, 25cm x 10 mm i.d Vydac C8 preparative columns (Sorbent Technologies, Inc, Atlanta, GA). Samples from RP-methacrylate adsorbent polymer resin were applied on RP-C8 column and STa was eluted by gradient system with 0.1% TFA in water as solvent A & 0.1%TFA in 80% methanol as solvent B (0-30% 5min and 30-80% in 80 min). The UV absorbing peaks were detected at 214 nm. Peaks were collected separately and the methanol was evaporated and then freeze-dried. The freeze-dried substance was reconstituted into physiological saline and tested for protein content and STa biological activity. S T a assessment for biological activity Detecting and quantifying STa biological activity was done using a reference standard in vivo model test, suckling mouse assay, according to Dean et al. (1972) and Giannella, (1976). Newborn Swiss Albino suckling mice (2-3 days old) were randomly divided into groups (three each). Samples from RP-HPLC were serially diluted 1/ 100; 1/ 10,000 & 100,000 and 10 ul of 0.2% Evans blue (w/v) was added per ml. Each suckling mouse was inoculated orally by IOO-ul sample using a 1 ml syringe and a 20-p-diameter polyethylene tube. Each sample dilution was tested in triplicate. After 2-hour incubation at room temperature, the mice were euthanized by carbon 63 dioxide in a C02 chamber and the intestine (not including the stomach) from each newborn mouse was removed and weighed. The ratios of intestinal weight to remaining body weight of the three mice were determined. Animals with no dye in the intestine or with dye within the peritoneal cavity at autopsy were discarded. One unit of ST activity (one mouse unit) is defined as the minimal amount of toxin that produces an intestinal weight/carcass ratio of 2 0.083. Criteria for homogeneity of purified S T a Homogeneity of the purified STa from preparative runs was validated by analytic aquapore RP-300A Perkin Elmer C8 column. Additionally, the exact molecular weight of STa was determined by. matrix-assisted laser desorption ionization-time of flight mass spectroscopy (MALDI-TOF/MS). The purified STa was then submitted for amino acid sequencing. Results and Discussion Detection of S T a gene PCR amplification verified that the tested strain carried the gene encoding for STa after analyzing the product on gel electrophoresis. Two amplicon bands of 127 bp and 244 bp were detected under UV light for the tested strain, which were not apparent for the control strain (E. coli K-12) (Figure 5). Culture analysis Growth kinetics experiments were conducted on 30L batch cultures under various pH values (7.4, 8 and 8.5). Samples were taken every two hours and the growth pattern was determined by counting the total cell count (CFU/ml) using a 64 robotic spiral plate and computer linked camera (Q counter). As Figure 6 indicates, tested ETEC growth was maximal in medium in which the initial pH was adjusted to 7.4. This level of grth was associated with higher level of crude STa as verified by the suckling mouse assay. Purification and Characterization of E. coli STa Table 7 shows the summary of the purification scheme of STa for the ETEC E. coli in 30 L batch culture. Amberlite XAD-2 Batch Adsorption Chromatography This step yields a high specific activity of the crude STa (8.70 x 103 MU/mg protein) compared with the STa specific activity in the cell free filtrate (1.22x103 MU/mg protein). Acetone fractionation Acetone fractionation resulted in further purification of the STa by removing additional amount of non-STa protein that was precipitated in acetone. Samples were taken for protein determination and STa biological activity. Specific activity of STa increased to 88.7 x 103 MU/mg protein. V Reversed Phase-Batch Adsorption Chromatography (MCI-gel) Specific activity of the STa at this step of purification increased from 88.7 x 103 to 112 x 103 MU/mg protein. This step allowed for a larger sample load on preparative RP-HPLC. Up to 15 mg of the crude STa cleaned by this procedure could be used as a single load in RP-HPLC without overloading the column or losing the resolution. This has led to a considerable reduction in the number of HPLC runs needed to purify STa. 65 Preparative Reverse-Phase HPLC Chromatography (RP-HPL C): Sixty percent methanol MCI-gel STa-rich fractions were loaded on a preparative C8 column for further purification. Figures 7 and 8 describe the elution profiles of STa. Elution with an increasing methanol gradient resulted in number of absorbance peaks at 214 nm (Figure 7), the last of which was found to contain enterotoxin activity. The enterotoxin peak began to elute at approximately 5 5-60% methanol after 35 minutes retention time. This peak was collected and after methanol evaporation, was freeze-dried. It was then reconstituted into physiological saline and tested for STa biological activity and protein concentration. Further improvement in the STa specific activity (885 x 104 MU/mg) was achieved in this step. The biological activity was demonstrated to be 0.113 ng perone mouse unit of STa minimal effective dose (MED) in 2-3 day-old inoculated Swiss Webster suckling mice. Criteria for homogeneity of purified STa I Analytic C8 column: Pooled peaks from several preparative RP-HPLC runs were tested on an analytic aquapore RP-300A Perkin Elmer C8 column to demonstrate a single symmetrical peak (Figure 8). I Mr-value determination using Matrix assisted laser desorption ionization- time of flighflmass spectroscopy: A lyophilized HPLC-purified sample was analyzed by MALDI-TOF/MS to determine the molecular weight and the result is shown in Figure 9. The observed signal at 100% MS intensity with m/c = 1972.1 indicates that the Mr of the purified product is 1972.1 Da, which is compatible with the Mr (1969-1972) value calculated from amino acid composition of the STa, confirming the purity and identity of the purified 66 product as the STa molecule. This was in agreement with the findings of Takao et al. (1983). I Amino acid sequence: Further confirmation of the homogeneity and identity of the purified product was performed by determination of amino acid sequence. A lyophilized HPLC-purified sample was submitted for amino acid sequence analysis and the results showed the 18 amino acid residues of the STa molecule were matching the reported sequence. Conclusion This protocol includes concentrating the cell free filtrate using Amberlite XAD-2 batch adsorption chromatography (BAC), acetone fractionation, and Methacrylate polymer resin BAC and finally through RP-HPLC. Chemical analysis of the purified preparations matched the reported structure for this type of enterotoxin. The biological activity was demonstrated to be less than 0.2 ng per one Mouse Unit ‘of the STa in 2-3 day-old inoculated Swiss Webster suckling mice. In summary, purification of STa to homogeneity was accomplished and the purity of the produced STa was documented through amino acid sequencing, and mass spectroscopy. 67 28 sees B 2: 0 EB 582 3 8m .m EB 83 an 82 .< 33m 55, eves as: sea :8 .m 3:80 .m ESE 3 SN 5? Baa anon :8 .m EEC .4 BEE 35 E: can: as: gas :8 .m 3550 .m 55E 35 e? Baa anon :8 .m aoazo .m cause <28 secs 3 82 ._ 88855 oozom was: mm Bodegas down: 35¢. :83 gem co omhm mo 26:2 €380 H2605 mow mo $223502“. 6w 8989‘. .m oSwE Esta—=92: Leta 2.5: A: :5 s 5:5 l G.» :5 c 6:5 11. ex :5 m 8:511 8de 2:25..» mgws. r mo+mon.w mom—co.“ gen .N gmgd SE36 Ill/{MD 68 5:28 Lea—m :28; w 0 895-355“ Baggy. £335“ :0 mason 5288528 03933: :8 .m 26.8 mzeflwofifi mo pica nous—m .w 2:?» 5:38 ouch; mo 3:8ng :0 cozoafi autumn—1w - Em H02 493508 3590.5: $8 mo oEoE 20:20 Emwuwoumaofio 25c: vegetated nwfiomgmougom .N. 8sz are . . Seared “V m. L; : new m .. 69 Exoaobfio 2 383.3: :8 .m we 2on Ecomoboomm mama Ema mo 08:-eoseNEom 83983 59: 62%.? x532 d PEME 08v cord coon Saw 8.2038: 8,8 camp «N. 3r 3 a 2“""§”‘"é“ .. ,1“ -«w—a and-..“ 3. on 8 8 .2 . 8. £885 .x. 70 Table 7. Summary of the purification of E. coli STa per batch iRP-HPLC Volume . Total PM“ SP A“ MED Purification Step Titer 7 Cone MU x ml MU/ 10 3 ng fold /mg 10 /mg Cell Free 3 -2 filtrate 30 x 10 10 3 24,660 1.22 822 1 Amberlite 4 XAD_2 BAC 120 10 1.2 1,378 8.70 114.9 7.13 New?" . 6O 10'5 6 676.8 88.7 11.28 72.67 Fractionation 60%MCI-gel F 30 10‘5 3 267.6 112.10 8.92 91.88 80 10'6 80 90.4 8849.56 0.113 7253.28 MU = Mouse Unit = minimal amount of toxin producing intestinal weight to remaining body weight ratio 3 0.085 Sp Ac = Specific activity = total mouse unit/protein concentration MED = Minimal effective dose = protein concentration per mg/total mouse unit per million BAC = Batch absorption chromatography Purification fold = specific activity of STa from each step/ specific activity of STa in the cell free filtrate Protein assay was done by Lowery method (Lowery 1951) using Perkin Elmer Spectrophotometer 71 References Dean, A; Chang, Y-C; Williams, R; and Harden, L B. 1972. Test for Escherichia coli enterotoxin using infant mice: Application in a study of diarrhea in children in Honolulu. The Journal of Infectious Diseases 125: 40 7-41 I . Evans, D.G; Evans, DJ; and Perce, NP. 1973. Differences in the response of rabbit small intestine to heat-labile and heat-stable enterotoxins of E. coli. Infection and Immunity. 7:8 73-880. Giannella, R. A. 1976. Suckling mouse model for detection of heat-stable Escherichia coli enterotoxin: Characteristics of the model. Infection and Immunity I4: 95- 99. Giannella, R. A. and Elizabeth, A. M. 2003. E. coli heat stable enterotoxin and guanylyl cyclase C: New functions and suspected actions. Trans Amr Clin Climato Ass. 1 14, 6 7-85. Guerrant R.L; Moore, R.A; Kirshenfed, RM. and Sande, M. S. 1975. Role of toxigenic and invasive bacteria in acute diarrhea of childhood. N. Egl. J. Med 293 :5 6 7-5 73. Gyles, C. L. 1971. Heat-labile and heat-stable forms of enterotoxin from Escherichia coli strains enteropathogenic for pigs. Ann NY Acad Sci. 1 76:314-322. Holmes, D. S. and Quigley, M. 1981. A rapid boiling method for the preparation of bacterial plasmids. Anal. Biochem. I 14, 193—1 9. Jiang, Z.D; Mathewson, J .J ; Ericsson, C.D; Svennerholm, A.M; Pulido, C. and DuPont H.L. 2000. Characterization of enterotoxigenic Escherichia coli strains in patients with travelers’ diarrhea acquired in Guadalajara, Mexico, 1992-1997. J Infect Dis. 181: 779-82. Naline. D.R; Bhattachaijee AK. and Richardson, SH. 1974. Cholera-like toxin effect of culture filtrates of Escherichia coli. J. Infec Dis. 130: 595-601. Olsvick, O and Strockbine NA. 1993. PCR detection of heat-stable, heat-labile and Shiga-like toxin genes in Escherichia coli. In Diagnostic Molecular Biology by Persing, DH; Smith, T.S; T enover, FC. and White, T.J. American Society for Microbiol Washinghton DC. Sack, R.B; Hirschhorn, N; Brownlee, I; Cash, R.A; Woodward, (W.E and Sack, DA. 1975. Enterotoxigenic Escherichia coli-associated diarrheal disease in Apache children. N.Engl.J. Med. 292:1041-1045. 72 Saeed, A. M; Srianganathan, N; Cosand, W; and Burger, D. 1983. Purification and characterization of heat-stable enterotoxin from bovine enterotoxigenic Escherichia coli. Infection and Immunity, 40: 701-701. Saeed, M. A; and Greenberg, RN. 1985. Preparative purification of Escherichia coli heat-stable enterotoxin. Anal. Bio.151: 431-43 7. Salvadori, M.R. Valadares, G.F; Leite D.D; Blanco, J. and Yano, T. 2003. Virulence factors of Escherichia coli isolated from calves with diarrhea in Barzil. Brazillian J Microbiology, 3 4: 23 0-235. Sears, C. L; and Kaper. J. B. 1996. Enteric bacterial toxins: mechanisms of action and linkage to intestinal secretion. Microbiol. Rev. 60:167-215. Smith, H.W; and Halls, S. 1967. Observation by the ligated intestinal segment and oral inoculation methods on Escherichia coli infections in pigs, calves, lambs and rabbits. J.Pathol.bacteriol. 93 :499-529. Staples, 8.]; Asher, SE; and Giannella, RA. 1980. Purification and characterization of heat-stable enterotoxin produced by a strain of E. coli pathogenic for man J. Biol. Chem. 155, 10: 4 71 6—4 721. Takao, T; Hitouji, T; Aimoto, S; Shimonishi Y; Hara, S; Takeda, Y. and Miwatani, T. 1983. Amino acid sequence of heat-stable enterotoxin isolated from enterotoxigenic Escherichia coli strain 18 D. FEBS Letters, 152: 1-5. Whipp, S.C; Moon H.W. and Lyon, NC. 1975. Heat-stable Escherichia coli enterotoxin production in vivo. Infect. Immun 12:240-244. Wolf, MK. 1997. Occurrence, distribution and associations of O and H serogroups colonization factor antigens and toxins of enterotoxigenic Escherichia coli. Rev Infect Dis. 1: 918-926. 73 CHAPTER IV DESIGN AND CHARACTERIZATION OF AN IMMUNOGENIC Escherichia coli HEAT-STABLE ENTEROTOXIN (STa)I Introduction Heat-stable enterotoxin (STa) is an important virulence determinant in enterotoxin-mediated diseases caused by E. coli (Sears & Kaper, 1996; Nataro & Kaper, 1998 and Giannella & Elizabeth, 2003). The immune system has difficulty in eliciting antibody against STa because of its small molecular weight (<2 kDa) (De Week, 1974 and Pereira et al. 2001). However, proteins of molecular size LOGO-3,000 Da can be made immunogenic by conjugation to a suitable macromolecule (carrier protein) (Elanger, 1980 and Pauillac et al. 1998). A wide range of carrier proteins and several procedures for conjugation have been reported for the preparation of peptide- carrier conjugates using various cross-linkers. The selection depends on the chemistry of conjugation and the location of antigenic determinants (epitopes) within the native peptide. Conjugation of STa to several types of carrier protein have been reported, however, no sufficient details were presented on the efficiency and the characteristics of these conjugates (Houghten et al. 1984, 1985; Klipstein et al. 1982, 1983; Sanchez et al. 1986, 1988; Clements, 1990 and Thompson and Giannella, 1990). In this study, purified STa peptide was covalently cross-linked to the modified bovine serum albumin (BSA) through the amino terminal of the STa to preserve the biologically active moiety that is perceived to be associated with carboxyl terminus of the peptide. ' Manuscript of this chapter is under preparation 74 This study aims to design and characterize an efficient immunogenic STa conjugates based on the evaluation of four different conjugation protocols. Background and principles of peptide -carrier conjugation Conjugation of proteins involves the linkage of two or more molecules to form a novel complex which has the combined properties or features of its individual components (Hermanson, 1996). Ideally, there are two critical criteria that should be met in any conjugation procedure. The first is that the coupling process should provide a high yield of well-defined and reproducible composition without inactivation of the peptide molecule. Secondly, it should yield a stable linkage between the peptide and carrier (Pauillac et al. 1998 and Fuentes et al. 2005). The strategies of the coupling process rely on the presence of functional moieties, which can be used to facilitate the conjugation of the peptide to a carrier protein. These moieties include carboxylic acids, amines, thiols, anhydrides, N- maleimides, imino esters and others. Depending on the functional moiety used, the reactive group may be the e-amino group of a lysine residue, a-amino of an N- terrninal (NH3,), sufhydryl [SH] group of cysteine, or the carboxylic acid [COO'] groups (Asp, Glu, or alpha-carboxyl). These moieties are capable of forming covalent bonds between two protein species (Dick & Beurret 1989 and Wong, 1991). Cross- linking reagents Cross-linkers contain reactive ends to specific functional groups (primary amines, sulfhydryls, etc.) on protein molecules (Wong, 1991 and Hermanson, 1996). Several cross-linkers are available and can be used to covalently link one molecule to 75 another. Most commonly used are carbodiimide derivatives, glutaraldehyde (GA), and m-maleimidobenzoyl—N-hydroxysuccinimide ester (MBS). Carbodiimide derivatives, both the water soluble derivative, 1-ethyl-3-(3- dimethylaminopropyl carbodiimide hydrochloride (EDAC) and the organic soluble derivative, N,N-dicyclohexylcarbodiimide (DCC) are commonly used. Carbodiimides are heterobifunctional reagents that are mainly used for coupling —COOH and —NH2 groups (Goodfriend et al. 1964 and Kurzer et al. 1967). They can activate the side chain carboxylic groups of aspartic and glutamic acid, as well as the carboxyl terminal group, to make reactive sites for coupling with primary amines (Yamada et al. 1981). Glutaraldehyde is a bifunctional coupling reagent that links two compounds through their amino groups (Habeeb & Hiramoto, 1968; Richard & Knowles, 1968 and Russel and Hopwood, 1976). Although glutaraldehyde provides a highly flexible spacer between the peptide and carrier protein for favorable presentation to the immune system, it is a very reactive compound and will react with Cys, Tyr and His to a limited extent and the result is a poorly defined conjugate (Molin et al. 1978). The m-Maleimidobenzoyl-N-hydroxysuccinimide ester (MBS) is a hetero- bifunctional reagent that can be used to link peptides to carrier proteins via cysteines (Carlson et al. 1978; King et al. 1978 and Yoshitaki et al. 1979). The coupling takes place with the thiol group (SH) of cysteine residues. Carrier Proteins Carrier proteins are macromolecules capable of stimulating the immune cells to elicit an antibody response (Harlow and Lane, 1988). Peptides lack the T-cell 76 epitopes, which are essential for priming CD4+ T helper cells. The carrier molecule provides the T-cell antigenic determinants for T-cell signaling, proliferation and release of mediators which activate specific B cells to stimulate antibody production in response to the immunogenic peptide-carrier conjugate (De Silva et al. 1999). Structurally, peptide-carrier conjugates have native antigenic determinants of the carrier as well as the peptide epiotpes. Introducing this conjugate into a suitable host generates peptide-specific antibodies (Brinkley, 1992) as well as carrier antibodies. Peptides have been coupled to several carrier proteins. The most commonly selected carriers are bovine serum albumin (BSA) and keyhole limpet hemacyanin (KLH). BSA is one of the most widely used carriers for the design of peptide-carrier conjugates. It is highly antigenic (M.W. ~67 kD) and can be easily modified to introduce a new moiety for specific coupling procedures (Habeeb 1967a&b). However, BSA is used as a blocking agent in many experimental immunoassays (ELISA), so antibodies raised against peptide-BSA conjugates will limit the application of the standard ELISA procedure and different blocking agents are needed. Although KLH is large (1.3 x 107 Da) and immunogenic, it may precipitate during cross-linking making it difficult to use in some cases. Moreover, the serum antibody response to this carrier ofien obscures that of the antigen of interest (De Silva et al. 1999) resulting in a reduced antigen-specific antibody concentration in the IgG fraction. Ovalbumin (OVA), rabbit serum albumin (RSA) thyroglobulin (TG) or synthetic carriers such as multiple antigenic peptides (MAPS) can also be used as carrier proteins (Wong, 1991 and Hermanson, 1996). 77 It is important to recognize that the immune system reacts to the peptide- protein carrier as a whole and that there will be a portion of response directed against the conjugated peptide as well as the carrier protein. Thus, coupling should be done to keep the antigen of interest in as native a condition as possible (Harlow and Lane, 1988). De Silva et al. (1999) showed that during preparation of peptide-carrier conjugates, one must strive to preserve the integrity of the antigen structure since many applications involving the antibodies require discrimination with very minimal conformational differences. In this study, BSA was modified in order to incorporate extra functional carboxyl groups and used as a carrier to cross-link the STa molecules through their amino terminals (Figure 10). Four different conjugation protocols were evaluated to design a successful conjugate that retained high biological activity for further use in the study of its immunogenicity. Materials and methods Reagents All reagents were obtained from commercial sources and were of analytical grade. Bovine serum albumin (BSA), succinic anhydride (SA), dioxane, N,N- dicyclohexylcarbodiimide (DCC), 1-ethyl-3-(3-dimethylaminopropyl carbodiimide hydrochloride (EDAC), N-methy-imidazole, dimethyformamide (DMF), 2—(N- morpholino) ethanesulfonic acid buffer (MES), triethylamine (ET3N), p-nitrophenol, sodium azide (NaNg) and phosphate buffer saline (PBS) tablets were obtained from Sigma Chemical Co. (St. Louis, MO). STa was purified and a detailed methodology was described in the previous chapter. 78 Carbodiimide C-terminal N—terminal Figure 10. Illustration showing the conjugate of bovine serum albumin carrier protein- E. coli STa peptide Procedure for covalently cross-linking STa with modified BSA Chemical modification of bovine serum albumin Bovine serum albumin was chemically modified to introduce new carboxyl moieties using two different protocols: I Succinylation I Hyper-succinylation Succinylation of Bovine Serum Albumin Basis of reaction: Succinic anhydride (SA) reacts rapidly with the e-amino groups of lysines and the a-amino groups of the N -termini of proteins at pH 7-9, forming an amide bond by replacing the amino group with a carboxyl (Riordan & Vallee 1964, Gounaris & Perlmann 1976 and Merck & Co., Inc 1996) (Figure 11). Thus, introducing a succinic anhydride moiety on BSA will afford a protein derivative with more carboxyl groups and hence increase the possibility to link STa 79 from its amino terminal and preserve its antigenic determinants associated with the carboxyl terminal of the molecule. (:1 3 4 + O _—... protein 2 1 2 3 4 0 NH 1 NH2 \[r CH2CH2 ----- COOH \j 0 Coverts amine to acid Figure 11. The action of succinic anhydride upon the amino groups of a protein Procedure: The methods followed were those described by Habeeb (1967a, b). Briefly, one gram of BSA was dissolved in 200 ml of 0.2 M borate buffer, pH 9.3. A 20 ml solution of dioxane with 5.4 g succinic anhydride was added in small aliquots over a period 30 minutes, the reaction mixture was stirred magnetically while maintaining the pH at 9.3 through the addition of 3 M NaOH. Following the last addition of succinic anhydride, the acylation reaction was allowed to continue for 45 minutes. The solution was then dialyzed at 4°C against several changes of 0.01 M triethylamine using dialysis tubing with a M.W. cutoff of 12-14 KD. The dialyzed preparation was first freeze-dried and then further dried in a dessecator over phosphorous pentoxide (P205). Samples were taken and reconstituted in PBS buffer (pH 6.8) for size exclusion chromatography and mass spectroscopy. Hyper-succinylated Bovine Serum Albumin (HS-BSA) Basis of reaction: An extensive modification of the BSA by introducing a large number of succinyl moieties [COO'] on the carrier protein could be achieved in 80 the hyper-succinylation reaction. The hyper-succinylation reaction was carried out in two steps. The first step involves the production of hyperaminated BSA by conversion of all free carboxyl groups on the BSA (aspartic and glutamic acids) into amino groups. The second step was the production of hypersuccinylated BSA by addition of succinic anhydride to the hyperaminated BSA to convert all amino groups (newly introduced, free, and N—terminal) into carboxyl groups. Procedure: Native BSA was treated with lmM of ethylenediamine at pH 4.75 in the presence of 10 mM EDAC. This hyperaminated protein molecule was then treated with 100 moles of succinic anhydride at pH 8.0 for two hours to produce a hyper-succinylated BSA molecule (Fuentes et al. 2005). Coupling of E. coli STa to modified BSA Four different conjugation protocols were used to covalently cross-linking STa to modified BSA. They were evaluated on the basis of stability of covalent bond, retained STa biological activity and the conjugation efficiency. Protocol 1: Using Dimethyformamide (DMF) as a solubilizer for the peptide and carrier protein Atassi (1981) described the use of DMF to solubilize several synthetic peptides before cross-linking them to carrier proteins. This protocol enhances the coupling of the amino terminal ends of synthetic peptides to the carrier protein. In this study, the carrier protein (suBSA) was solubilized in DMF and then treated with p- nitrophenol to activate the carboxyl groups on the carrier. Procedure: Applying this procedure, we have used the purified native STa peptide and have solubilized it in DMF prior to addition to the modified suBSA. The mixture was kept stirring overnight at room temperature. This design was meant to encourage 81 the STa coupling through its amino terminals based on nucleophilic attack at the reactive ester groups of the modified suBSA forming amide linkages. In a typical reaction, we used 140 mg of suBSA suspended in 10 ml of anhydrous DMF and stirred it magnetically in a tight-capped foil-wrapped bottle for 3-4 hours. A solution of p—nitrophenol (65mg/0.5m1) in DMF was added and magnetic stirring continued for 15 minutes. A solution of DCC (50 mg/O.5ml) was added to suBSA at a molar ratio of 120:] and the reaction was allowed to continue stirring at room temperature for 3 hours. One hundred mg of STa in 1 ml DMF was added to the activated suBSA at a molar ratio of 24:1. Shortly after, 1 ml of triethylamine was added. The reaction mixture was stirred overnight, at room temperature, while protected from direct light. The next day, 30 ml of Milli Q water was added and the mixture was dialyzed extensively against distilled water at 4°C using a dialysis membrane with a 12-14 kD M.W. cutoff and then freeze-dried. Samples were taken for measurement of the STa biological activity of the conjugate using suckling mice assay (SMA), protein determination, biochemical and molecular characterization. Protocol 2: Imidazole-based protocol One coupling procedure (Dean et al. 1990) described the use of imidazole to stabilize the carrier protein and minimize the formation of polymers due to the acylation process when the cross linker and the peptides intended for cross-linking are added. Procedure: We used 0.5 M N-methyl-imidazole pH (6.0) to dissolve the STa peptide and the carrier protein suBSA at a molar ratio (100:1). After the addition of EDAC (molar ratio: 50 mol EDAC/mol STa), the mixture was stirred for 30 minutes 82 at room temperature followed by dialysis (M.W. cutoff 12-14 kD) against distilled water at 4°C. Protocol 3: Hyper-succinylated BSA-based protocol Fuentes et al. (2005) reported that an increase in the numbers of succinyl groups [COO'] on the carrier protein can enhance the cross linking of the peptides from their amino terminal. The procedure for hyper-succinylation of BSA was described above. Procedure: In this coupling protocol, 3 mg hypersuccinylated-BSA was dissolved in 2.5 ml of 5 mM sodium phosphate buffer pH 7 and mixed with 2.5 ml of the STa peptide (0.5 mg/ml) in dioxane. EDAC was then added gradually to reach a concentration of 100 mM. After that, the conjugated composite was dialyzed using a tube with a M.W. cutoff 12-14 kD against distilled water at 4°C. Protocol 4: Conventional peptide-carrier coupling protocol In this coupling procedure, suBSA, EDAC and MES buffer were used (Uptima interchim, 2007). Procedure: SuBSA carrier protein was dissolved in 0.1M MES buffer pH 5 to a final concentration of 10mg/ml. Two milligrams of STa peptide were added to 2 mg of suBSA carrier protein. Then, EDAC (10 mg /ml in cold distilled water) was added at a ratio of 0.5 mg of EDAC per mg of total protein. The reaction was kept stirring for 2-3 hours at room temperature before dialysis at 4 °C against PBS using a tube of 12-14 kD M.W. cutoff. 83 Dialysis The products of BSA modification and STa-modified BSA conjugation reactions were subjected to extensive dialysis to remove the small molecular weight reactants (<14 kD). Dialysis tubing with nominal M.W. cut-off 12-14 kD was purchased from Fisher Scientific (Pittsburg, PA). STa-SuBSA conjugate was subjected to extensive dialysis against Milli Q purified water using a dialysis membrane of 12-14kD M.W. cutoff. Molecular species of l4kD or higher were retained inside the dialysis tube and all other reactants below 14 kD including uncoupled (free) toxin were dialyzed out. Gel Filtration Chromatography (GFC) PD-lO columns, Sephadex G-25M packed columns, of a nominal molecular mass exclusion limit of 5000 for protein were purchased from GE. Healthcare (Buckingharnshire, UK). These columns are designed to separate proteins based on their molecular weight. The columns were equilibrated and developed by following the manufacturer’s instructions. The dialyzed STa conjugate samples were passed through PD-lO Sephadex G-25 GFC column to purify the STa-suBSA conjugates from free STa, then assessed for biological activity and protein concentration. A freeze-dried STa-suBSA conjugate sample was reconstituted into 2.5 ml of PBS and passed through a PD-lO column to separate the unconjugated STa peptide from the portion that was successfully cross linked to the carrier. The STa-carrier conjugate was then eluted with 3.5 ml PBS and the effluent was collected and tested for biological activity in a suckling mouse assay. 84 Size Exclusion-High Performance Liquid Chromatography SE-HPLC was performed to compare the molecular size of both native and modified BSA. Bio-Sil® SEC-125 HPLC 300 x 7.8mm filtration column (Cat # 125- 0060) was purchased from BioRad (Hercules, CA) and hooked to the Waters Associate Liquid Chromatography System equipped with multi-solvent delivery pumps, an automated gradient programmer 6008 controller and a tunable absorbance detector (Model 486). The column was equilibrated with 0.05 M sodium phosphate (diabasic), 0.05M sodium phosphate (monobasic), 0.15M NaCl and 0.01 M NaN3 at pH 6.8. Isocratic elution system was applied at flow rate 1 ml/min. The peak absorbance was monitored at UV 280 nm. Amino acid compositional analysis To determine the conjugation ratio of STa to the modified BSA, a freeze-dried STa-suBSA conjugate sample was subjected to amino acid compositional analysis at the Research Technology Support Facility (RTSF) at Michigan State University. Matrix-assisted laser desorption ionization/time of flight mass spectroscopy (MALDI-TOF/MS) Two hundreds micrograms of freeze-dried STa-suBSA conjugate sample were subjected to MALDI-TOF/MS analysis at the MSU-RTSF laboratory to determine precisely the molecular weight of the conjugate and use this figure to calculate the number of STa molecules that covalently cross-linked to one BSA molecule (conjugation ratio). 85 Protein Assay Protein assays were performed according to the Lowery method (Lowry et al. 1951), using a Perkin Elmer Lambda 3A UV/U S Spectrophotometer. STa-suBSA conjugates activity bioassay The biological activity of the STa-suBSA conjugates was determined using the suckling mouse assay as described by Dean et al. (1972), Giannella, (1976) and modified by Saeed et al. (1983). M Table 8 shows the summary of conjugation experiments and their evaluation. More details on the DMF conjugation protocol are presented below. Characteristics of the modified BSA The results from SE-HPLC showed that modified BSA with succinic anhydride was eluted faster (RT= 2.99 min) than native BSA (RT= 4.20 min), suggesting that the molecular weight of suBSA had undergone a significance change (Figure 12). Change in the molecular weight of the succinylated BSA was also confirmed by MALDI-TOF/ MS. It was found that suBSA has a M.W. of 72.40 kD in comparison to native BSA which has a M.W. of ~67.00 kD (Figure 13). Characteristics of E. coli STa-suBSA conjugates Dialysis and Gel Filtration Chromatography A summary of the results is presented in Table 8. The conjugation protocol based on the DMF method showed a higher rate of conjugation efficiency and higher level of retained STa biological activity compared to other conjugates (Table 8). 86 Further characterization of the DMF-based conjugation protocol is described belowAmino acid compositional analysis A dialyzed conjugate sample based on the DMF protocol was subjected to amino acid compositional analysis. Table 9 shows the pmole concentration and retention time of each amino acid residue. Calculation of S T a Peptide to suBSA Ratio The conjugation ratio is defined as the number of STa molecules covalently cross-linked to one molecule of succinylated BSA. The amino acid composition of the conjugate was empirically determined by measuring the pmoles of each amino acid detected in a known volume of sample. Well-recovered residues, arginine and methionine, were used to quantify the concentration of each residue (pmole) in the conjugate sample. The number of STa molecules in the conjugate sample was calculated using the Arg & Meth residues not present within the sequence of the STa peptide. Table 10 gives the approximate number of coupled STa molecules to one molecule of modified BSA. Based on the data of amino acid compositional analysis, it was found that approximately 4-5 STa molecules were coupled to each molecule of suBSA. S T a Conjugate Analysis by MLDI-TOFMS Lyophilized conjugate samples based on DMF and HS-BSA protocols were subjected to MALDI-TOF/ MS to accurately determine the molecular weight (Mr Value). Both samples showed median Mr Values over 80 kDa (Figure 14 & 15). Figure 16 shows the molecular weight differences for the modified BSA before and after STa peptide conjugation (AM.W. = 8342.5 Da). This difference was attributed to 87 the contribution of the STa molecules. Based on this data, the median number of STa molecules successfully crossed linked to one molecule of suBSA was calculated from the following equation: A M.W. /STa M.W. = 8342.5/ 1959 = 4 -5. Four to five STa molecules were successfully crossed linked to one molecule of modified BSA based on DMF and HS-BSA protocols. S T a conjugate activity bioassay & Conjugation efficiency We concluded that the STa biological activity and the conjugation efficiency of the conjugate were highest in the DMF protocol. A summary of conjugation efficiency based on the tested protocols expressed largely by the conjugation ratios and the retained STa biological activities of the conjugates is presented in Table 8. Discussion Numerous attempts have been made to render STa immunogenic, including chemical coupling and genetic fusions to appropriate carrier proteins (Houghten et al. 1984, 1985, Sanchez et al. 1986, 1988, Clements, 1990). However, results of these studies showed limited success since the uncontrolled cross-linking process led to the loss of the biological activity of STa as a part of the conjugation process (Pereira et al. 2001). Additionally, these studies showed no sufficient details on the efficiency and the characteristics of the produced STa conjugates. The objective of this study was to design and characterize a well-defined, stable and active STa conjugate for further study of its immunogenicity in laboratory animal models. We have evaluated several conjugation protocols to achieve a stable biologically active STa conjugate through carefully planned cross-linking of the STa 88 peptide to a modified carrier protein using BSA, carbodiimide derivatives and different solvents. Given the perceived molecular structure of the STa peptide and the desire to crosslink it through its amino terminus, we have selected carbodiimide coupling reagents. Other coupling reagents, glutaraldehyde and m-Maleimidobenzoyl-N- hydroxysuccinimide ester (MBS), may affect the 3-demientional structure of the STa peptide. Glutaraldehyde binds non—specific amino groups and this leads to polymerization of peptide and/ or carrier protein, which results in a poorly defined conjugate (Molin et al. 1978). Cysteine residues on the STa peptide play a crucial role in the biological activity and the stability of STa peptide. Thus, using MBS as a hetero-bifimctional reagent targeting thiol group on cysteine residue (Carlson et al. 1978; King et al. 1978 and Yoshitaki et al. 1979) may disrupt the disulfide bonds and affect the biological moieties on the STa peptide. BSA is widely used as a carrier protein in conjugation reactions because it is highly antigenic and can be easily modified to introduce a new moiety for specific coupling procedures (Habeeb 1967a&b). Therefore, the use of carbodiimide and BSA, in our conjugation reaction was justified based on a thorough understanding of the molecular structure of the STa peptide. In this study, BSA was modified by introducing succinic moieties, and its modification was confirmed using size exclusion chromatography and MALDI- TOF/MS. The data showed a 5000 Da difference in the molecular size between the modified and native BSA molecules, indicating an 8% increases in the M.W. of the modified BSA. This suggests that an extensive modification of the free amino groups 89 was achieved with the addition of succinic anhydride (Habeeb, 1967b). The subsequent step in the design of STa-BSA conjugate was the cross-linking of STa to the modified BSA. This reaction was initiated by incubation of the modified BSA with p—nitrophenol and DCC for three hours to provide reactive ester groups that could easily attach the STa from its amino terminal, forming amide linkages. The use of DMF was shown to enhance the solubility of reactants including peptides and carrier proteins ((Lateef, 2007). We believe that the use of DMF as a solvent reagent may have facilitated the solubility of the hydrophobic STa molecules, solving a problem encountered with the other solvents and coupling media. The STa-conjugate was tested for its protein content and biological activity. Based on the protein estimation, there was a conjugation efficiency of 52-64%, which is higher than previously reported (Frantz & Robertson 19981; F rantz et al. 1987; and Thompson and Giannella, 1990). Moreover, this conjugate showed a higher biological activity than any activity reported in the previous STa-conjugates (Table 8). Based on these results, it is clear that most of the biological activity of the STa introduced into this reaction was retained in the conjugate even after extensive dialysis, GFC and SEC. Covalent attachment of STa molecules to modified BSA was documented by amino acid composition analysis and MALDI-TOF/ MS. A median value for the conjugation ratio of 4-5:1 STazsuBSA has been determined based on amino acid analysis and MALDI-TOF/MS (Figure 16). Based on the results of the biological activity of this conjugate, we believe strongly that STa molecules may have been more efficiently oriented on the BSA carrier molecule via linkage through their amino 9O terminals. Such orientation, achieved through the DMF protocol, has preserved the biologically active moiety of the STa and may offer an explanation for the relatively low yield of STa conjugate produced by other protocols in this study. The ineffective preservation and presentation of the STa biologically active moiety on previously studied STa conjugates may also explain the sub-optimal immune response against STa in laboratory animals (Alderete and Robertson 1978; Lockwood and Robertson 1984; Houghten et al. 1984, 1985; Sanchez et al. 1986, 1988; Clements, 1990; Lowenadler et al. 1991 and Pereira et al. 2001). In summary, we have designed a well-defined STa-conjugate based on a thorough understanding of the molecular structure of the STa peptide. Afier careful evaluation of several peptide-carrier conjugation protocols, we have defined the conditions for a conjugate that expressed a high STa biological activity in suckling mice. Its stability and biochemical attributes were characterized using GFC, amino acid analysis and MALDI-TOF/mass spectroscopy. 91 3388...-.. 182.... 583 H . IL IIII>II4§1IIAII :P. .\ .A .k 1 O 5 o. 5...... 3.5.... 5........... 1.. .15.........§..?£J.§.&3f+~1¥2¥5K1$.5§..5 ON 8.3. 5...MW .55.. w .88 m. mNmN ,8 . 55...... 3.. , r m 35.5.. m 9. 55.5. N SEN... .. _. .. o. 0.82. .. 5. mom 5.. 5 ... ._ ., . moo % .. . .5. . . ._. .,. . 0.5. 5 .. .. . .8 5. $35 I... 5 005 b.3055 .x. .11. mémoow ozfiméo: :8 .m 0:... mo 0:8 :oflwmsEoo was 5303 33032: < mo nous—HSHU .3 oSmE Emma—.38 8E8 oQ 5823.0 02.2 <95 3.. £5 ’ Jfl 4 = i 3558 3 How Do mm a 08383 05 I; 40 4 088:0:0 :95 some 8 0000a 83 9.2 Ed mow pm ~28 0002050 20 08m , H. . .. - M.0 C 83% 0836 28.5002: 00_ .EE . 2 0.. 2 a 8:385 23 :03 be some 8 000?». we? 3&va 20028 8 3:20 05 8-523. Ax..n..0.0 88033 023on £030 .8 E 00_ -mmm E 00.0 53> 3:5 Bow :3? b0 32% 33 Cuban wE 00 -503 o . - . ow 0883 98 3-525. $8.0 .x Law—WW 0mm WWW. mewmw “flow -mmm 320.0 0:3 88: 58 fig? . .9 58 we 50 00 mm as 02335 b0 8 3320 £25 :08 8 825 mm? 03 ESE can 8-523. $3.0-me -55 “wow cowmwsfiooomfimammonm $20.0 H33 880 500 :33 2E8?“ 085:0 28-003031 00“ P. v “a images £3 28 Gowns 9:88 823 0.0 mm .5003 828080 28.0 E a: 84.9221 2: 55 0880 mm? Baa «aim 2:. be 8 cocoa 05 8-593 $8.0 .20 5:00 £5 380 :50 :83 114 STa-IgG neutralization 0.140 0.120 0.100 0.080 0.060 0.040 0.020 0.000 GWIRBW wk1 wk3 wk6 wk12 wk17 wk20 wk24 Week Post immnization Figure 18. E. coli STa-specific serum antibody neutralization bioassay: Group 1 (rabbits. 1, 2, and 3). Straight horizontal line at 0.083 Gut weight: remaining body weight ratio, signifies the cut off value for STa- positive SAM. Neutrallztion Capacity of IgG 100000 10000 ‘ i w 1000 ‘8 a g 100 O z 10 0 5 10 15 20 25 30 weeks post Immunization Figure 19. Neutralization capacity of E. coli STa-specific serum antibody. Points represent the mean of neutralization titers from sera of 3 rabbits 115 STa-IgG neutralization bioassay 0.16 0.14 -+- Rabbil#1 _._ Rabbit# 2 _.— Rabbit#3 0.12 3 °-1 —-— Rabbit#4 g o 08 _._ Rabbi1# 5 E V j + Rabbit#6 + Rabbit#7 ' 7 —-— Rabbit# 8 —I- RabbiflfiQ —I-— Rabbit#10 wk1 wk 12 wk 17 wk 20 wk 24 weeks postinmunization Figure 20. E. coli STa-specific serum antibody neutralization bioassay: All rabbits Note: Three rabbits showed STa neutralizing antibodies by 12 weeks, another 3 rabbits showed STa neutralizing antibodies by 17 weeks and finally 2 rabbits started to show STa neutralizing antibodies by 20 weeks post immunization GW= Gut weight, RBW= Remaining body weight 116 Ablnecxess OD405nm 10‘3 10‘4 10‘5 10‘6 Reciprocal serum dilution Figure 21. E. coli STa-binding ELISA optimization: screening serum dilution for optimal E .coli STa-STa antibody interaction ST a-igG Ending Assay 10M serum dilution &seine wk3 wk6 wk 12 wki5 wk 17 wk20 wk24 weeks Fl Figure 22. E. coli STa-specific serum antibody: 10‘4 serum dilution of group 1 rabbits 117 —I— Rabbiw 4 + Rabbi1# 5 + Rabbii# 6 OD 405 nm Baseline wk 3 wk 6 wk 12 wk 17 wk 20 wk 24 Weeks Pl Figure 23. E. coli STa-specific serum antibody: ‘10'4 serum dilution of group 2 rabbits 2.500 + Rabbiflf- 7 + Rabbiflf 8 2 .000 1 .500 1.000 OD 405 nm 0.500 0.000 Baseline wk 3 wk 6 wk 12 wk 17 wk 20 wk 24 weeks Pi Figure 24. E. coli STa-specific serum antibody: 10'4 serum dilution of group 3 rabbits 118 i E ; 2.500 — 1 +61 PI Rabbit 2.000 - +G1 Baseline 2 1.500 — In 0 v o _ O 1.000 0.500 — 0.000 a . . . . . J . 10"3 10M 10"5 10"6 Reciprocal of serum Dilution Figure 25. Mean OD value of group 1 rabbits after 20 weeks post immunization at various serum dilutions 2.5 . +G2 Rabbit 2 +G2 Baseline E 1.5 — In O <- 0 - O 1 0.5 - 0 , . I 14"". 10"3 10M 10"5 10"6 Reciprocal of serum dilution Figure 26. Mean OD value of group 2 rabbits after 20 weeks post immunization at various serum dilutions 119 +63 Pl Rabbits 2.000 - +63 Baseline g 1.500 - 8 1.000 - 0.500 — 0.000 K t, = 2.500 - 10M 10"5 Reciprocal of serum Dilution 10‘6 Figure 27. Mean OD value of group 3_ rabbits afier 24 weeks post immunization at various serum dilutions OD 405 nm 2.500 2.000 .5 0' o 0 § 0.500 0.000 10"3 10"4 10"5 Reciprocal of Serum Dilution -I— Rabbit# 1 —¢-— Rabbil# 2 -l- Rabbit# 3 —l- Rabbifll 4 -I— Rabbit# 5 + Rabbi’di 6 --—- Rabbit# 7 -—l— Rabbit# 8 10‘6 Figure 28. End titer of E. coli STa- specific serum antibody: 24 weeks post immunization from 8 rabbits 120 2.500 — 2 000 + Group 1 ' q A + Group 2 +Group 3 + Baseline E 1.500 ~ C In 0 V o o 1.000 . 0.500 - 0.000 . . T . . T a 10"3 10M 10"5 10"6 Reciprocal of Serum Dilution Figure 29. E. coli STa-specific serum antibody end titer: Mean OD value of group 1, 2 and 3 rabbits after 24 weeks post immunization at various serum dilutions 121 OD 405 nm 2.5 + umtreated 0 3 6 12 17 24 weeks postimmunization Figure 30. Time-course evaluation of the avidity of E. coli STa-specific serum antibody using ammonium thiocyanate dose response 122 OD 405 nm 0 3 6 17 24 weeks postimmunization Figure 31. Five molar thiocyanate elution profile of E. coli STa- STa serum antibody complex: Mean OD of treated serum from group 1, 2 and 3 of rabbits Avidity index of three groups 0 3 6 17 24 weeks post im m unlzation Figure 32. Avidity index of E. coli STa-specific serum antibody from group 1, 2 and 3 of rabbits using avidity ELISA procedure 123 Table 11: E. coli STa-specific serum antibody end titer: Mean OD 1: SD value of group 1, 2 and 3 rabbits after 24— week post immunization at various serum dilutions Grow/serum 10'3 10“ 10'5 10“ dilution G1 1.933 :t 1.923 :t 1.237 i 0.379 :1: 0.013 0.014 0.568 0.352 G2 1.964 t 1.969 :1: 1.550 :t 0.374 :t 0.035 0.031 0.010 0.173 G3 1.936 1 1.656 :1: 0.354 d: 0.040 :t 0.006 0.035 0.015 0.010 Baseline 0.864 :1: 0.232 d: 0.033 :t 0.039 :1: 0.318 0.155 0.048 0.060 Table 12. Summary of STa- ELISA binding and neutralization end titers of rabbit sera immunized with STa-suBSA conjugate after the primary immunization and during the boosting intervals. Data were generated by STa binding ELISA and STa neutralization using suckling mouse assay. Anti-STa Response Neutralization Blee din Protein Specific g ELISA Neutralized Assay Activity Titer STa MU/ml mg/ml serum (Titerl Baseline 0 0 64.5 - Week 3 PI 0 0 66.8 - Week 6 PI 0 0 65.7 - Week 12 PI 10,000 2000 83.5 23.95 Week 15 PI 100,000 15,000 69.6 215.52 Week 20 PI 1,000,000 20,000 65.00 307.69 Week 24 PI 1,000,000 20,000 65.00 307.69 Week 28 PI 1,000,000 30,000 65.00 461.54 124 Table 13. Summary of the development of STa antibody avidity after multiple boosters with the STa conjugate using 5 M ammonium thiocyanate ELISA dissociation assay. Baseline Week 3 Week 6 1273‘ Week 24 Avidity Index (AI) % afier measurement OD at 405 nm Gm 1 0.017 0.043 0.001 0.578 0.927 P (4.70%) (8.91%) (0.03%) (25.94%) (48.21%) Gm 2 0.043 0 0.022 0.027 0.762 P (3%) (0%) (2.24%) (1.53%) (38.71%) Gm“ 3 0.04 0 0.095 0.151 0.352 P (0%) (0%) (1 1.36%) (12.05%) (21.30%) Table 14. Neutralization capacity of sera from animals immunized with several STa- immuogenes and the end titers of the STa-neutralizing antibodies as reported in the literature. . Neutralization Max1mum Amount of . References neutralization titer ‘ SMU cap acity Total MU/ml serum Liiwenadler et al. . 1991 1.55 30 1665 Lockwood and , Robertson 1984 1'100 8 800 Frantz and , . Robertson 1981 1.5000 to l.10,000 1 5000 to 10,000 Alderete and _ - Robertson 1978 1'2’300 1 2’500 1:50 for immunogene using 1 50 . native STa fusion Pereira et al. protein 2001 1:4000 using mutated STa fusion 1 4,000 protein This study 1:30.000 1 30,000 125 References Alderete J .F. and Robertson, DC. 1978. Purification and chemical characterization of the heat-stable enterotoxin produced by porcine strains of enterotoxigenic Escherichia coli. Infection and Immunity 19: 1021-1030. Boedeker, E. C. 2005. Vaccines for enterotoxigenic Escherichia coli: current status. Current Opinion in Gastroenterology. 21 (1):] 5-1 9. Cardenas, L and Clements, J .D. 1993. Development of mucosal protection against the heat-stable enterotoxin (ST) of Escherichia coli by oral immunization with a genetic fusion delivered by a bacterial vector. Infect Immun. 61: 4629—463 6. Clements, J. D. 1990. Construction of a nontoxic fusion peptide for immunization against Escherichia coli strains that produce heat-labile and heat-stable enterotoxins. Infect Immun 5 8, 1 159-1 1 66. Ferreira, M.U. and Katzin, A.M. 1995. The assessment of antibody distribution by hocyanate elution: a simple dose-response approach. Journal of Immunological Methods 1 8 7:29 7-3 05. Frantz, J. C and Robertson, DC. 1981. Immunological properties of Escherichia coli heat-stable enterotoxins: development of a radioimmunoassay specific for heat- stable enterotoxins with suckling mouse activity. Infection and Immunity. 33: 193-198. Frantz, J .C; Bhatnagar, P.K; Brown, A.L; Garrett, L.K.and Hughes, J .L. 1987. Investigation of synthetic Escherichia coli heat-stable enterotoxin as an immunogen for swine and cattle. Infection and Immunity. 55: 1077—1084. Goldblatt, D. 2001. Affinity of antigen-antibody interactions. Encyclopedia of Life Sceinences. John Wiley &Sons, Ltd. [-5 Gordon, L.K. 1981. A reliable method for repetitively bleeding rabbits from the central artery of the ear. Journal of immunological methods, 44: 241 -24. Gyles, C. L. (1996). In Escherichia coli in domestic animals and humans. pp. 31-72 CAB International. Wallingford. UK. Houghten, R. A., Engert, R. F., Ostresh, J. M., Hoffman, S. R. & Klipstein, F. A. 1985. A completely synthetic toxoid vaccine containing Escherichia coli heat- stable toxin and antigenic determinants of the heat-labile toxin B subunit. Infect Immun 48, 735- 740. 126 Houghten, R. A., Ostresh, J. M. & Klipstein, F. A. 1984. Chemical synthesis of an octadecapeptide with the biological and immunological properties of human heat-stable Escherichia coli enterotoxin. Eur J Biochem 145, 157-162. Klipstein, F. A., Engert, R. F. & Clements, J. D. 1982. Development of a vaccine of cross-linked heat-stable and heat-labile enterotoxins that protects against Escherichia coli producing either enterotoxin. Infect Immun 3 7, 55 0-55 7. Klipstein, F. A., Engert, R. F ., Clements, J. D. & Houghten, R. A.1983. Vaccine for enterotoxigenic Escherichia coli based on synthetic heat-stable toxin crossed- linked to the B subunit of heat-labile toxin. J Infect Dis 147, 318-326. Kosek, M., Bern, C., Guerrant, KL 2003. The magnitude of global burden of diarrhoea] disease from studies published 1992-2000. Bulletin of the World Health Organization, 81: p19 7-204. Lefl