Controlled Release of Bordetella Bronchiseptica Dermonecrotoxin(BBD) Vaccine from BBD-Loaded Chitosan Microspheres In Vitro

  • Jiang, Hu-Lin (School of Agricultural Biotechnology, Seoul National University) ;
  • Park, In-Kyu (School of Agricultural Biotechnology, Seoul National University) ;
  • Shin, Na-Ri (College of Veterinary Medicine and the School of Agricultural Biotechnology, Seoul National University) ;
  • Yoo, Han-Sang (College of Veterianry Medicine and the School of Agricultural Biotechnology, Seoul National University) ;
  • Akaike, Toshihiro (Department of Biomolecular Engineering, Tokyo Institute of Technolog) ;
  • Cho, Chong-Su (School of Agricultural Biotechnology, Seoul National University)
  • Published : 2004.03.01

Abstract

Chitosan microspheres were prepared by ionic gelation process with sodium sulfate for nasal vaccine delivery. Bordetella Bronchiseptica Dermonecrotoxin (BBD) as a major virulence factor of a causative agent of atrophic rhinitis (AR) was loaded to the chitosan microspheres for vaccination. Morphology of BBD-loaded chitosan microspheres was observed as spherical shapes. The average particle sizes of the BBD-loaded chitosan microspheres were about $2.69$\mid${\;}\mu\textrm{m}$. More BBD was released with an increase of molecular weight of chitosan and with an increase of medium pH in vitro due to weaker intermolecular interaction between chitosan and BBD. Tumor necrosis $factor-{\alpha}{\;}(TNF{\alpha})$ and nitric oxide (NO) from RAW264.7 cells stimulated with BBD-loaded chitosan microspheres were gradually secreted, suggesting that released BBD from chitosan microspheres had immune stimulating activity of AR vaccine.

Keywords

References

  1. Artursson, P., Lindmark, T., Davis, S. S., and ilium, L., Effect of chitosan on permeability of monolayers of intestinal epithelial cells (Caco-2). Pharm. Res., 11, 1358-1361 (1994) https://doi.org/10.1023/A:1018967116988
  2. Eldridge, J. H., Hammond, C. J., Meulbrock, J. A., Staas, J. K., Gilley, R. M., and Rice, T. R., Controlled vaccine release in the gut-associated lymphoid tissues. I. Orally administered biodegradable microspheres target the peyer's patches. J. Cont. ReI., 11, 205-214 (1990) https://doi.org/10.1016/0168-3659(90)90133-E
  3. Eyles, J., Sharp, G., Williamson, E., Spiers, I., and Alpar, H., Intranasal administration of poly-lactic acid microsphere co-encapsulated Yersinia pestis subunits confers protection from pneumonic plague in the mouse. Vaccine, 16, 698-707 (1998) https://doi.org/10.1016/S0264-410X(97)00249-1
  4. Green, L. C., Wagner, D. A., Glogowski, J., Skipper, PL., J. Wishnok, S., and Tannenbaum, S. R., Analysis of nitrate, nitrite, and $[^{15}N]$ nitrate in biological fluids. Anal. Biochem., 126, 131-138 (1992) https://doi.org/10.1016/0003-2697(82)90118-X
  5. Hasebe, H., Occurrence and epizootiological surveys of infectious atrophic rhinitis in swine. Nippon Vet. Zootech. Col. Bull., 19, 92-102 (1971)
  6. He, P., Davis, S. S, and ilium, L., In vitro evaluation of the mucoadhesive properties of chitosan microspheres. Int. J. Pharm., 166, 75-88 (1998) https://doi.org/10.1016/S0378-5173(98)00027-1
  7. Ilium, L., Farraj, N. F., and Davis, S. S., Chitosan as a novel nasal delivery system for peptide drugs. Pharm. Res., 11, 1186-1189 (1994) https://doi.org/10.1023/A:1018901302450
  8. Ilium, L., Jabbal-Gill, I., Hinchcliffe, M., Fisher, A. N., and Davis, S. S., Chitosan as a novel nasal delivery system for vaccines. Adv. Drug Deliv. Rev., 51, 81-96 (2001) https://doi.org/10.1016/S0169-409X(01)00171-5
  9. Janes, K. A., Calvo. P., and Alonso, M. J., Polysaccharide colloidal particles as delivery systems for macromolecules. Adv. Drug Deliv. Rev., 47, 83-97 (2001) https://doi.org/10.1016/S0169-409X(00)00123-X
  10. Kreuter, J. and Speiser, P. P., New adjutants on poly (methylmethacrylate) base. Infect Immunol., 13, 204-210 (1976)
  11. Kreuter, J. and Speiser, .P P., In vitro studies of poly (methylmethacrylate) adjutants. J. Pharm. Sci., 65, 1624-1627 (1976) https://doi.org/10.1002/jps.2600651115
  12. Kreuter, J. and Liehl, E., Long-term studies of microencapsulated and adsorbed influenza vaccine nanoparticles. J. Pharm. Sci., 70, 367-371 (1981) https://doi.org/10.1002/jps.2600700406
  13. Kreuter, J., Liehl, E., and Berg, V., Influence of hydrophobicity on the adjuvant effect of particlate polymeric adjutants. Vaccine, 6, 253-256 (1988) https://doi.org/10.1016/0264-410X(88)90220-4
  14. Lika, I., Vine, T., Nishimura, K., and Azuma, I., Simulation of non-specific host resistance against Sendai virus and E. coli infection by chitin in mice. Vaccine, 5, 270-274 (1987) https://doi.org/10.1016/0264-410X(87)90150-2
  15. Lowry, O. and Rosebrough, N., Protein measurement with the folin phenol reagent. J. Biol. Chem., 193-265 (1951)
  16. Lydyard, P. and Grossi, C., The lymphoid system. Immunology, 5, 31-42 (1998)
  17. Mi, F. L., Shyu, S. S., Chen, C. T., and Schoung, J. Y., Porous chitosan microsphere for controlling the antigen release of Newcastle disease vaccine: preparation of antigen-adsorbed microsphere and in vitro release, Biomaterials, 20, 1603-1612 (1999) https://doi.org/10.1016/S0142-9612(99)00064-2
  18. Saiki, I., Murata, J., Tokura, S., and Azuma, I., Inhibition by sulfated chitin derivatives of advasion through extracellular matrix and enzymatic degradation metastatic melanoma cells. Cancer Res., 50, 3631-3637 (1990)
  19. Shin, N. R., Kim, J. M., and Yoo, H. S., Korean J. Vet. Res., 40, 551-561 (2000)
  20. Suzuki, K., Ogawa, Y., Hashimoto, K., Suzuki, S., and Suzuki, M., Protecting elect of chitin and chitosan on experimentally induced murine candidasis. Microbial. Immunol., 28, 903-904 (1984)
  21. Van der Lubben, I. M., Kersten, G., Fretz, M. M., Beuvery, C., Verhoef, J. C., and Junginger, H. E., Chitosan microparticles for mucosal vaccination against diphtheria:oral and nasal efficacy studies in mice. Vaccine, 21, 1400-1408 (2003) https://doi.org/10.1016/S0264-410X(02)00686-2