1932

Abstract

Enteric viral infections in domestic animals cause significant economic losses. The recent emergence of virulent enteric coronaviruses [porcine epidemic diarrhea virus (PEDV)] in North America and Asia, for which no vaccines are available, remains a challenge for the global swine industry. Vaccination strategies against rotavirus and coronavirus (transmissible gastroenteritis virus) infections are reviewed. These vaccination principles are applicable against emerging enteric infections such as PEDV. Maternal vaccines to induce lactogenic immunity, and their transmission to suckling neonates via colostrum and milk, are critical for early passive protection. Subsequently, in weaned animals, oral vaccines incorporating novel mucosal adjuvants (e.g., vitamin A, probiotics) may provide active protection when maternal immunity wanes. Understanding intestinal and systemic immune responses to experimental rotavirus and transmissible gastroenteritis virus vaccines and infection in pigs provides a basis and model for the development of safe and effective vaccines for young animals and children against established and emerging enteric infections.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-animal-022114-111038
2015-02-16
2024-04-30
Loading full text...

Full text loading...

/deliver/fulltext/animal/3/1/annurev-animal-022114-111038.html?itemId=/content/journals/10.1146/annurev-animal-022114-111038&mimeType=html&fmt=ahah

Literature Cited

  1. US Dep. Agric. 2014. National Animal Health Monitoring System. Washington, DC: US Dep. Agric. http://www.aphis.usda.gov/animal_health/nahms/
  2. Saif LJ, Jackwood D. 1990. Enteric virus vaccines: theoretical considerations, current status, and future approaches. In Viral Diarrheas of Man and Animals, ed. LJ Saif, KW Theil, pp. 313–32. Boca Raton, FL: CRC Press
  3. Cima G. 2013. Fighting a deadly pig disease. Industry, veterinarians trying to contain PED virus, new to the US. J. Am. Vet. Med. Assoc. 243:469–70 [Google Scholar]
  4. Andersen JK, Takamatsu H, Oura CA, Brookes SM, Pullen L, Parkhouse RE. 1999. Systematic characterization of porcine ileal Peyer's patch, I. Apoptosis-sensitive immature B cells are the predominant cell type. Immunology 98:612–21 [Google Scholar]
  5. Lucier MR, Thompson RE, Waire J, Lin AW, Osborne BA, Goldsby RA. 1998. Multiple sites of Vλ diversification in cattle. J. Immunol. 161:5438–44 [Google Scholar]
  6. Parng CL, Hansal S, Goldsby RA, Osborne BA. 1996. Gene conversion contributes to Ig light chain diversity in cattle. J. Immunol. 157:5478–86 [Google Scholar]
  7. Reynaud CA, Garcia C, Hein WR, Weill JC. 1995. Hypermutation generating the sheep immunoglobulin repertoire is an antigen-independent process. Cell 80:115–25 [Google Scholar]
  8. Butler JE, Lager KM, Splichal I, Francis D, Kacskovics I et al. 2009. The piglet as a model for B cell and immune system development. Vet. Immunol. Immunopathol. 128:147–70 [Google Scholar]
  9. Chu RM, Liu CH. 1984. Morphological and functional comparisons of Peyer's patches in different parts of the swine small intestine. Vet. Immunol. Immunopathol. 6:391–403 [Google Scholar]
  10. Yasuda M, Jenne CN, Kennedy LJ, Reynolds JD. 2006. The sheep and cattle Peyer’s patch as a site of B-cell development. Vet. Res. 37:401–15 [Google Scholar]
  11. Pabst O, Herbrand H, Worbs T, Friedrichsen M, Yan S et al. 2005. Cryptopatches and isolated lymphoid follicles: dynamic lymphoid tissues dispensable for the generation of intraepithelial lymphocytes. Eur. J. Immunol. 35:98–107 [Google Scholar]
  12. Iwasaki A, Medzhitov R. 2004. Toll-like receptor control of the adaptive immune responses. Nat. Immunol. 5:987–95 [Google Scholar]
  13. Tohno M, Shimosato T, Moue M, Aso H, Watanabe K et al. 2006. Toll-like receptor 2 and 9 are expressed and functional in gut-associated lymphoid tissues of presuckling newborn swine. Vet. Res. 37:791–812 [Google Scholar]
  14. Guzylack-Piriou L, Balmelli C, McCullough KC, Summerfield A. 2004. Type-A CpG oligonucleotides activate exclusively porcine natural interferon-producing cells to secrete interferon-α, tumour necrosis factor-α and interleukin-12. Immunology 112:28–37 [Google Scholar]
  15. Bailey M, Plunkett FJ, Rothkotter HJ, Vega-Lopez MA, Haverson K, Stokes CR. 2001. Regulation of mucosal immune responses in effector sites. Proc. Nutr. Soc. 60:427–35 [Google Scholar]
  16. Burkey TE, Skjolaas KA, Minton JE. 2009. Board-invited review: porcine mucosal immunity of the gastrointestinal tract. J. Anim. Sci. 87:1493–501 [Google Scholar]
  17. Saif LJ. 1999. Comparative pathogenesis of enteric viral infections of swine. In Mechanisms in the Pathogenesis of Enteric Diseases 2, ed. P Paul, D Francis, pp. 47–59. New York: Kluwer Acad./Plenum Publ.
  18. Saif LJ. 1999. Enteric viral infections of pigs and strategies for induction of mucosal immunity. Adv. Vet. Med. 41:429–46 [Google Scholar]
  19. Bohl EH, Gupta RK, Olquin MV, Saif LJ. 1972. Antibody responses in serum, colostrum, and milk of swine after infection or vaccination with transmissible gastroenteritis virus. Infect. Immun. 6:289–301 [Google Scholar]
  20. Saif LJ, Bohl EH, Gupta RK. 1972. Isolation of porcine immunoglobulins and determination of the immunoglobulin classes of transmissible gastroenteritis viral antibodies. Infect. Immun. 6:600–9 [Google Scholar]
  21. Roux ME, McWilliams M, Phillips-Quagliata JM, Weisz-Carrington P, Lamm ME. 1977. Origin of IgA-secreting plasma cells in the mammary gland. J. Exp. Med. 146:1311–22 [Google Scholar]
  22. Saif LJ, van Cott JL, Brim TA. 1994. Immunity to transmissible gastroenteritis virus and porcine respiratory coronavirus infections in swine. Vet. Immunol. Immunopathol. 43:89–97 [Google Scholar]
  23. Berri M, Meurens F, Lefevre F, Chevaleyre C, Zanello G et al. 2008. Molecular cloning and functional characterization of porcine CCL28: possible involvement in homing of IgA antibody secreting cells into the mammary gland. Mol. Immunol. 45:271–77 [Google Scholar]
  24. Salmon H, Berri M, Gerdts V, Meurens F. 2009. Humoral and cellular factors of maternal immunity in swine. Dev. Comp. Immunol. 33:384–93 [Google Scholar]
  25. Moxley RA, Olson LD. 1989. Clinical evaluation of transmissible gastroenteritis virus vaccines and vaccination procedures for inducing lactogenic immunity in sows. Am. J. Vet. Res. 50:111–18 [Google Scholar]
  26. Saif LJ, Pensaert M, Sestak K, Yeo SG, Jung K. 2012. Coronaviruses. In Diseases of Swine, ed. JJ Zimmerman, LA Karriken, A Ramirez, KJ Schwartz, GW Stevenson, pp. 501–24. Ames, IA: Wiley Blackwell
  27. Song D, Park B. 2012. Porcine epidemic diarrhoea virus: a comprehensive review of molecular epidemiology, diagnosis, and vaccines. Virus Genes 44:167–75 [Google Scholar]
  28. Saif LJ, Fernandez FM. 1996. Group A rotavirus veterinary vaccines. J. Infect. Dis. 174:Suppl. 1S98–106 [Google Scholar]
  29. Park S, Sestak K, Hodgins DC, Shoup DI, Ward LA et al. 1998. Immune response of sows vaccinated with attenuated transmissible gastroenteritis virus (TGEV) and recombinant TGEV spike protein vaccines and protection of their suckling pigs against virulent TGEV challenge exposure. Am. J. Vet. Res. 59:1002–8 [Google Scholar]
  30. Saif LJ. 1985. Passive immunity to coronavirus and rotavirus infections in swine and cattle: enhancement by maternal vaccination. Presented at Int. Semin. Diarrhoeal Dis. South East Asia West. Pac. Reg., Geelong, Australia
  31. Yuan L, Geyer A, Saif LJ. 2001. Short-term immunoglobulin A B-cell memory resides in intestinal lymphoid tissues but not in bone marrow of gnotobiotic pigs inoculated with Wa human rotavirus. Immunology 103:188–98 [Google Scholar]
  32. Knipe DM, Howley PM. 2007. Fields’ Virology. Philadelphia: Wolters Kluwer/Lippincott Williams & Wilkins
  33. Amimo JO, Vlasova AN, Saif LJ. 2013. Prevalence and genetic heterogeneity of porcine group C rotaviruses in nursing and weaned piglets in Ohio, USA and identification of a potential new VP4 genotype. Vet. Microbiol. 164:27–38 [Google Scholar]
  34. Martella V, Banyai K, Lorusso E, Bellacicco AL, Decaro N et al. 2007. Prevalence of group C rotaviruses in weaning and post-weaning pigs with enteritis. Vet. Microbiol. 123:26–33 [Google Scholar]
  35. Amimo JO, Vlasova AN, Saif LJ. 2013. Detection and genetic diversity of porcine group A rotaviruses in historic (2004) and recent (2011 and 2012) swine fecal samples in Ohio: predominance of the G9P[13] genotype in nursing piglets. J. Clin. Microbiol. 51:1142–51 [Google Scholar]
  36. Barrington GM, Parish SM. 2001. Bovine neonatal immunology. Vet. Clin. North Am. Food Anim. Pract. 17:463–76 [Google Scholar]
  37. Chattha KS, Vlasova AN, Kandasamy S, Esseili MA, Siegismund C et al. 2013. Probiotics and colostrum/milk differentially affect neonatal humoral immune responses to oral rotavirus vaccine. Vaccine 31:1916–23 [Google Scholar]
  38. Hodgins DC, Kang SY, deArriba L, Parreno V, Ward LA et al. 1999. Effects of maternal antibodies on protection and development of antibody responses to human rotavirus in gnotobiotic pigs. J. Virol. 73:186–97 [Google Scholar]
  39. Nguyen TV, Yuan L, Azevedo MS, Jeong KI, Gonzalez AM et al. 2006. High titers of circulating maternal antibodies suppress effector and memory B-cell responses induced by an attenuated rotavirus priming and rotavirus-like particle-immunostimulating complex boosting vaccine regimen. Clin. Vaccine Immunol. 13:475–85 [Google Scholar]
  40. Kitikoon P, Nilubol D, Erickson BJ, Janke BH, Hoover TC et al. 2006. The immune response and maternal antibody interference to a heterologous H1N1 swine influenza virus infection following vaccination. Vet. Immunol. Immunopathol. 112:117–28 [Google Scholar]
  41. Loeffen WL, Heinen PP, Bianchi AT, Hunneman WA, Verheijden JH. 2003. Effect of maternally derived antibodies on the clinical signs and immune response in pigs after primary and secondary infection with an influenza H1N1 virus. Vet. Immunol. Immunopathol. 92:23–35 [Google Scholar]
  42. Kimman TG, Westenbrink F, Schreuder BE, Straver PJ. 1987. Local and systemic antibody response to bovine respiratory syncytial virus infection and reinfection in calves with and without maternal antibodies. J. Clin. Microbiol. 25:1097–106 [Google Scholar]
  43. Ellis J, West K, Cortese V, Konoby C, Weigel D. 2001. Effect of maternal antibodies on induction and persistence of vaccine-induced immune responses against bovine viral diarrhea virus type II in young calves. J. Am. Vet. Med. Assoc. 219:351–56 [Google Scholar]
  44. Platt R, Widel PW, Kesl LD, Roth JA. 2009. Comparison of humoral and cellular immune responses to a pentavalent modified live virus vaccine in three age groups of calves with maternal antibodies, before and after BVDV type 2 challenge. Vaccine 27:4508–19 [Google Scholar]
  45. Brar JS, Johnson DW, Muscoplat CC, Shope RE Jr, Meiske JC. 1978. Maternal immunity to infectious bovine rhinotracheitis and bovine viral diarrhea viruses: duration and effect on vaccination in young calves. Am. J. Vet. Res. 39:241–44 [Google Scholar]
  46. Besser TE, Gay CC, McGuire TC, Evermann JF. 1988. Passive immunity to bovine rotavirus infection associated with transfer of serum antibody into the intestinal lumen. J. Virol. 62:2238–42 [Google Scholar]
  47. Besser TE, McGuire TC, Gay CC, Pritchett LC. 1988. Transfer of functional immunoglobulin G (IgG) antibody into the gastrointestinal tract accounts for IgG clearance in calves. J. Virol. 62:2234–37 [Google Scholar]
  48. Sasaki M, Davis CL, Larson BL. 1977. Immunoglobulin IgG1 metabolism in new born calves. J. Dairy Sci. 60:623–26 [Google Scholar]
  49. Chase CC, Hurley DJ, Reber AJ. 2008. Neonatal immune development in the calf and its impact on vaccine response. Vet. Clin. North Am. Food Anim. Pract. 24:87–104 [Google Scholar]
  50. Siegrist CA. 2001. Neonatal and early life vaccinology. Vaccine 19:3331–46 [Google Scholar]
  51. Munoz-Zanzi CA, Thurmond MC, Johnson WO, Hietala SK. 2002. Predicted ages of dairy calves when colostrum-derived bovine viral diarrhea virus antibodies would no longer offer protection against disease or interfere with vaccination. J. Am. Vet. Med. Assoc. 221:678–85 [Google Scholar]
  52. Chappuis G. 1998. Neonatal immunity and immunisation in early age: lessons from veterinary medicine. Vaccine 16:1468–72 [Google Scholar]
  53. Hjelm F, Carlsson F, Getahun A, Heyman B. 2006. Antibody-mediated regulation of the immune response. Scand. J. Immunol. 64:177–84 [Google Scholar]
  54. Nguyen TV, Yuan L, Azevedo MS, Jeong KI, Gonzalez AM et al. 2006. Low titer maternal antibodies can both enhance and suppress B cell responses to a combined live attenuated human rotavirus and VLP-ISCOM vaccine. Vaccine 24:2302–16 [Google Scholar]
  55. Richter MY, Jakobsen H, Haeuw JF, Power UF, Jonsdottir I. 2005. Protective levels of polysaccharide-specific maternal antibodies may enhance the immune response elicited by pneumococcal conjugates in neonatal and infant mice. Infect. Immun. 73:956–64 [Google Scholar]
  56. Hodgins DC, Shewen PE. 2012. Vaccination of neonates: problem and issues. Vaccine 30:1541–59 [Google Scholar]
  57. Siegrist CA. 2003. Mechanisms by which maternal antibodies influence infant vaccine responses: review of hypotheses and definition of main determinants. Vaccine 21:3406–12 [Google Scholar]
  58. Siegrist CA, Cordova M, Brandt C, Barrios C, Berney M et al. 1998. Determinants of infant responses to vaccines in presence of maternal antibodies. Vaccine 16:1409–14 [Google Scholar]
  59. Ellis J, Gow S, West K, Waldner C, Rhodes C et al. 2007. Response of calves to challenge exposure with virulent bovine respiratory syncytial virus following intranasal administration of vaccines formulated for parenteral administration. J. Am. Vet. Med. Assoc. 230:233–43 [Google Scholar]
  60. Kimman TG, Westenbrink F, Straver PJ. 1989. Priming for local and systemic antibody memory responses to bovine respiratory syncytial virus: effect of amount of virus, virus replication, route of administration and maternal antibodies. Vet. Immunol. Immunopathol. 22:145–60 [Google Scholar]
  61. Morein B, Abusugra I, Blomqvist G. 2002. Immunity in neonates. Vet. Immunol. Immunopathol. 87:207–13 [Google Scholar]
  62. Zimmerman AD, Boots RE, Valli JL, Chase CC. 2006. Evaluation of protection against virulent bovine viral diarrhea virus type 2 in calves that had maternal antibodies and were vaccinated with a modified-live vaccine. J. Am. Vet. Med. Assoc. 228:1757–61 [Google Scholar]
  63. Firth MA, Prando Moore D, Pei Y, Shewen PE, Lo RY et al. 2006. Cloning of a gene fragment encoding bovine complement component C3d with expression and characterization of derived fusion proteins. Vet. Immunol. Immunopathol. 114:61–71 [Google Scholar]
  64. Kim D, Niewiesk S. 2014. Synergistic induction of interferon α through TLR-3 and TLR-9 agonists stimulates immune responses against measles virus in neonatal cotton rats. Vaccine 32:265–70 [Google Scholar]
  65. Hoare CM, DeBouck P, Wiseman A. 1997. Immunogenicity of a low-passage, high-titer modified live canine parvovirus vaccine in pups with maternally derived antibodies. Vaccine 15:273–75 [Google Scholar]
  66. Yuan L, Azevedo MS, Gonzalez AM, Jeong KI, Van Nguyen T et al. 2005. Mucosal and systemic antibody responses and protection induced by a prime/boost rotavirus-DNA vaccine in a gnotobiotic pig model. Vaccine 23:3925–36 [Google Scholar]
  67. Endsley JJ, Quade MJ, Terhaar B, Roth JA. 2002. BHV-1-specific CD4+, CD8+, and γδ T cells in calves vaccinated with one dose of a modified live BHV-1 vaccine. Viral Immunol. 15:385–93 [Google Scholar]
  68. Endsley JJ, Ridpath JF, Neill JD, Sandbulte MR, Roth JA. 2004. Induction of T lymphocytes specific for bovine viral diarrhea virus in calves with maternal antibody. Viral Immunol. 17:13–23 [Google Scholar]
  69. Siegrist CA. 2000. Vaccination in the neonatal period and early infancy. Int. Rev. Immunol. 19:195–219 [Google Scholar]
  70. Firth MA, Shewen PE, Hodgins DC. 2005. Passive and active components of neonatal innate immune defenses. Anim. Health Res. Rev. 6:143–58 [Google Scholar]
  71. Hodgins DC, Shewen PE. 2000. Vaccination of neonatal colostrum-deprived calves against Pasteurella haemolytica A1. Can. J. Vet. Res. 64:3–8 [Google Scholar]
  72. Husband AJ, Lascelles AK. 1975. Antibody responses to neonatal immunisation in calves. Res. Vet. Sci. 18:201–7 [Google Scholar]
  73. Yuan L, Saif LJ. 2002. Induction of mucosal immune responses and protection against enteric viruses: rotavirus infection of gnotobiotic pigs as a model. Vet. Immunol. Immunopathol. 87:147–60 [Google Scholar]
  74. Siegrist CA, Aspinall R. 2009. B-cell responses to vaccination at the extremes of age. Nat. Rev. Immunol. 9:185–94 [Google Scholar]
  75. Chattha KS, Firth MA, Hodgins DC, Shewen PE. 2009. Age related variation in expression of CD21 and CD32 on bovine lymphocytes: a cross-sectional study. Vet. Immunol. Immunopathol. 130:70–78 [Google Scholar]
  76. Chattha KS, Firth MA, Hodgins DC, Shewen PE. 2010. Variation in expression of membrane IgM, CD21 (CR2) and CD32 (Fcγ RIIB) on bovine lymphocytes with age: a longitudinal study. Dev. Comp. Immunol. 34:510–17 [Google Scholar]
  77. Chattha KS, Firth MA, Hodgins DC, Shewen PE. 2010. Expression of complement receptor 2 (CD21), membrane IgM and the inhibitory receptor CD32 (FcγRIIb) in the lymphoid tissues of neonatal calves. Vet. Immunol. Immunopathol. 137:99–108 [Google Scholar]
  78. McCauley I, Hartmann PE. 1984. Changes in piglet leucocytes, B lymphocytes and plasma cortisol from birth to three weeks after weaning. Res. Vet. Sci. 37:234–41 [Google Scholar]
  79. Nagahata H, Kojima N, Higashitani I, Ogawa H, Noda H. 1991. Postnatal changes in lymphocyte function of dairy calves. Zentralblatt Vet. B 38:49–54 [Google Scholar]
  80. Chattha KS, Hodgins DC, DeLay J, Antoine N, Shewen PE. 2010. Immunohistochemical investigation of cells expressing CD21, membrane IgM, CD32 and a follicular dendritic cell marker in the lymphoid tissues of neonatal calves. Vet. Immunol. Immunopathol. 137:284–90 [Google Scholar]
  81. Filipp D, Alizadeh-Khiavi K, Richardson C, Palma A, Paredes N et al. 2001. Soluble CD14 enriched in colostrum and milk induces B cell growth and differentiation. PNAS 98:603–8 [Google Scholar]
  82. Nguyen TV, Yuan L, Azevedo MS, Jeong KI, Gonzalez AM, Saif LJ. 2007. Transfer of maternal cytokines to suckling piglets: in vivo and in vitro models with implications for immunomodulation of neonatal immunity. Vet. Immunol. Immunopathol. 117:236–48 [Google Scholar]
  83. Saif LJ, Ward LA, Yuan L, Rosen BI, To TL. 1996. The gnotobiotic piglet as a model for studies of disease pathogenesis and immunity to human rotaviruses. Arch. Virol. Suppl. 12:153–61 [Google Scholar]
  84. Saif L, Yuan L, Ward L, To T. 1997. Comparative studies of the pathogenesis, antibody immune responses, and homologous protection to porcine and human rotaviruses in gnotobiotic piglets. Adv. Exp. Med. Biol. 412:397–403 [Google Scholar]
  85. Vajdy M. 2008. Immunity Against Mucosal Pathogens. Dordrecht, Neth.: Springer
  86. Yuan L, Wen K, Azevedo MS, Gonzalez AM, Zhang W, Saif LJ. 2008. Virus-specific intestinal IFN-γ producing T cell responses induced by human rotavirus infection and vaccines are correlated with protection against rotavirus diarrhea in gnotobiotic pigs. Vaccine 26:3322–31 [Google Scholar]
  87. Yuan L, Ward LA, Rosen BI, To TL, Saif LJ. 1996. Systematic and intestinal antibody-secreting cell responses and correlates of protective immunity to human rotavirus in a gnotobiotic pig model of disease. J. Virol. 70:3075–83 [Google Scholar]
  88. Ward LA, Yuan L, Rosen BI, To TL, Saif LJ. 1996. Development of mucosal and systemic lymphoproliferative responses and protective immunity to human group A rotaviruses in a gnotobiotic pig model. Clin. Diagn. Lab. Immunol. 3:342–50 [Google Scholar]
  89. de Arriba ML, Carvajal A, Pozo J, Rubio P. 2002. Mucosal and systemic isotype-specific antibody responses and protection in conventional pigs exposed to virulent or attenuated porcine epidemic diarrhoea virus. Vet. Immunol. Immunopathol. 85:85–97 [Google Scholar]
  90. Yuan L, Kang SY, Ward LA, To TL, Saif LJ. 1998. Antibody-secreting cell responses and protective immunity assessed in gnotobiotic pigs inoculated orally or intramuscularly with inactivated human rotavirus. J. Virol. 72:330–38 [Google Scholar]
  91. Wang Y, Azevedo M, Saif LJ, Gentsch JR, Glass RI, Jiang B. 2010. Inactivated rotavirus vaccine induces protective immunity in gnotobiotic piglets. Vaccine 28:5432–36 [Google Scholar]
  92. Azevedo MS, Yuan L, Pouly S, Gonzales AM, Jeong KI et al. 2006. Cytokine responses in gnotobiotic pigs after infection with virulent or attenuated human rotavirus. J. Virol. 80:372–82 [Google Scholar]
  93. Zhang W, Wen K, Azevedo MS, Gonzalez A, Saif LJ et al. 2008. Lactic acid bacterial colonization and human rotavirus infection influence distribution and frequencies of monocytes/macrophages and dendritic cells in neonatal gnotobiotic pigs. Vet. Immunol. Immunopathol. 121:222–31 [Google Scholar]
  94. Wen K, Azevedo MS, Gonzalez A, Zhang W, Saif LJ et al. 2009. Toll-like receptor and innate cytokine responses induced by lactobacilli colonization and human rotavirus infection in gnotobiotic pigs. Vet. Immunol. Immunopathol. 127:304–15 [Google Scholar]
  95. Wen K, Li G, Zhang W, Azevedo MS, Saif LJ et al. 2011. Development of γδ T cell subset responses in gnotobiotic pigs infected with human rotaviruses and colonized with probiotic lactobacilli. Vet. Immunol. Immunopathol. 141:267–75 [Google Scholar]
  96. Gonzalez AM, Azevedo MS, Jung K, Vlasova A, Zhang W, Saif LJ. 2010. Innate immune responses to human rotavirus in the neonatal gnotobiotic piglet disease model. Immunology 131:242–56 [Google Scholar]
  97. Gonzalez AM, Nguyen TV, Azevedo MS, Jeong K, Agarib F et al. 2004. Antibody responses to human rotavirus (HRV) in gnotobiotic pigs following a new prime/boost vaccine strategy using oral attenuated HRV priming and intranasal VP2/6 rotavirus-like particle (VLP) boosting with ISCOM. Clin. Exp. Immunol. 135:361–72 [Google Scholar]
  98. Iosef C, Van Nguyen T, Jeong K, Bengtsson K, Morein B et al. 2002. Systemic and intestinal antibody secreting cell responses and protection in gnotobiotic pigs immunized orally with attenuated Wa human rotavirus and Wa 2/6-rotavirus-like-particles associated with immunostimulating complexes. Vaccine 20:1741–53 [Google Scholar]
  99. Yuan L, Geyer A, Hodgins DC, Fan Z, Qian Y et al. 2000. Intranasal administration of 2/6-rotavirus-like particles with mutant Escherichia coli heat-labile toxin (LT-R192G) induces antibody-secreting cell responses but not protective immunity in gnotobiotic pigs. J. Virol. 74:8843–53 [Google Scholar]
  100. Yuan L, Iosef C, Azevedo MS, Kim Y, Qian Y et al. 2001. Protective immunity and antibody-secreting cell responses elicited by combined oral attenuated Wa human rotavirus and intranasal Wa 2/6-VLPs with mutant Escherichia coli heat-labile toxin in gnotobiotic pigs. J. Virol. 75:9229–38 [Google Scholar]
  101. Azevedo MP, Vlasova AN, Saif LJ. 2013. Human rotavirus virus-like particle vaccines evaluated in a neonatal gnotobiotic pig model of human rotavirus disease. Expert Rev. Vaccines 12:169–81 [Google Scholar]
  102. Fukushima Y, Kawata Y, Hara H, Terada A, Mitsuoka T. 1998. Effect of a probiotic formula on intestinal immunoglobulin A production in healthy children. Int. J. Food Microbiol. 42:39–44 [Google Scholar]
  103. Sanz Y, De Palma G. 2009. Gut microbiota and probiotics in modulation of epithelium and gut-associated lymphoid tissue function. Int. Rev. Immunol. 28:397–413 [Google Scholar]
  104. Holscher HD, Czerkies LA, Cekola P, Litov R, Benbow M et al. 2012. Bifidobacterium lactis Bb12 enhances intestinal antibody response in formula-fed infants: a randomized, double-blind, controlled trial. J. Parenter. Enter. Nutr. 36:106S–17S [Google Scholar]
  105. Mullie C, Yazourh A, Thibault H, Odou MF, Singer E et al. 2004. Increased poliovirus-specific intestinal antibody response coincides with promotion of Bifidobacterium longum-infantis and Bifidobacterium breve in infants: a randomized, double-blind, placebo-controlled trial. Pediatr. Res. 56:791–95 [Google Scholar]
  106. Zhang W, Azevedo MS, Wen K, Gonzalez A, Saif LJ et al. 2008. Probiotic Lactobacillus acidophilus enhances the immunogenicity of an oral rotavirus vaccine in gnotobiotic pigs. Vaccine 26:3655–61 [Google Scholar]
  107. Fanaro S, Chierici R, Guerrini P, Vigi V. 2003. Intestinal microflora in early infancy: composition and development. Acta Paediatr. Suppl. 91:48–55 [Google Scholar]
  108. Rinne M, Kalliomaki M, Arvilommi H, Salminen S, Isolauri E. 2005. Effect of probiotics and breastfeeding on the Bifidobacterium and Lactobacillus/Enterococcus microbiota and humoral immune responses. J. Pediatr. 147:186–91 [Google Scholar]
  109. Sjogren YM, Tomicic S, Lundberg A, Bottcher MF, Bjorksten B et al. 2009. Influence of early gut microbiota on the maturation of childhood mucosal and systemic immune responses. Clin. Exp. Allergy 39:1842–51 [Google Scholar]
  110. Chattha KS, Vlasova AN, Kandasamy S, Rajashekara G, Saif LJ. 2013. Divergent immunomodulating effects of probiotics on T cell responses to oral attenuated human rotavirus vaccine and virulent human rotavirus infection in a neonatal gnotobiotic piglet disease model. J. Immunol. 191:2446–56 [Google Scholar]
  111. Kumar A, Vlasova AN, Liu Z, Chattha KS, Kandasamy S et al. 2014. In vivo gut transcriptome responses Lactobacillus rhamnosus GG and Lactobacillus acidophilus in neonatal gnotobiotic piglets. Gut Microbes 5:2152–64 [Google Scholar]
  112. Kaufman DR, De Calisto J, Simmons NL, Cruz AN, Villablanca EJ et al. 2011. Vitamin A deficiency impairs vaccine-elicited gastrointestinal immunity. J. Immunol. 187:1877–83 [Google Scholar]
  113. Sommer A, Katz J, Tarwotjo I. 1984. Increased risk of respiratory disease and diarrhea in children with preexisting mild vitamin A deficiency. Am. J. Clin. Nutr. 40:1090–95 [Google Scholar]
  114. Villamor E, Fawzi WW. 2005. Effects of vitamin A supplementation on immune responses and correlation with clinical outcomes. Clin. Microbiol. Rev. 18:446–64 [Google Scholar]
  115. Mora JR, Iwata M, Eksteen B, Song SY, Junt T et al. 2006. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science 314:1157–60 [Google Scholar]
  116. Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M et al. 2007. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J. Exp. Med. 204:1775–85 [Google Scholar]
  117. Iwata M, Hirakiyama A, Eshima Y, Kagechika H, Kato C, Song SY. 2004. Retinoic acid imprints gut-homing specificity on T cells. Immunity 21:527–38 [Google Scholar]
  118. Vlasova AN, Chattha KS, Kandasamy S, Siegismund CS, Saif LJ. 2013. Prenatally acquired vitamin A deficiency alters innate immune responses to human rotavirus in a gnotobiotic pig model. J. Immunol. 190:4742–53 [Google Scholar]
  119. Chattha KS, Kandasamy S, Vlasova AN, Saif LJ. 2013. Vitamin A deficiency impairs adaptive B and T cell responses to a prototype monovalent attenuated human rotavirus vaccine and virulent human rotavirus challenge in a gnotobiotic piglet model. PLOS ONE 8:e82966 [Google Scholar]
  120. Jee J, Hoet AE, Azevedo MP, Vlasova AN, Loerch SC et al. 2013. Effects of dietary vitamin A content on antibody responses of feedlot calves inoculated intramuscularly with an inactivated bovine coronavirus vaccine. Am. J. Vet. Res. 74:1353–62 [Google Scholar]
  121. Huang YW, Dickerman AW, Piñeyro P, Li L, Fang L et al. 2013. Origin, evolution, and genotyping of emergent porcine epidemic diarrhea virus strains in the United States. mBio 4:e00737–13 [Google Scholar]
  122. Stevenson GW, Hoang H, Schwartz KJ, Burrough ER, Sun D et al. 2013. Emergence of porcine epidemic diarrhea virus in the United States: clinical signs, lesions, and viral genomic sequences. J. Vet. Diagn. Investig. 25:649–54 [Google Scholar]
  123. Hill RE Jr, Foley PL, Carr MY, Elsken LA, Gatewood DM et al. 2003. Regulatory considerations for emergency use of non-USDA licensed vaccines in the United States. Dev. Biol 114:31–52 [Google Scholar]
  124. Schwartz K, Henry S, Tokach L, Potter M, Davidson D, Egnor C. 2014. Exposing sows to PEDV to build herd immunity. Natl. Hog Farmer March 13 [Google Scholar]
  125. Stevenson GW, Hoang H, Schwartz KJ, Burrough ER, Sun D et al. 2013. Emergence of porcine epidemic diarrhea virus in the United States: clinical signs, lesions, and viral genomic sequences. J. Vet. Diagn. Investig 25:5649–54 [Google Scholar]
  126. Sueyoshi M, Tsuda T, Yamazaki K, Yoshida K, Nakazawa M et al. 1995. An immunohistochemical investigation of porcine epidemic diarrhoea. J. Comp. Pathol 113:159–67 [Google Scholar]
/content/journals/10.1146/annurev-animal-022114-111038
Loading
/content/journals/10.1146/annurev-animal-022114-111038
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error