Clin Exp Vaccine Res.  2014 Jul;3(2):176-184. 10.7774/cevr.2014.3.2.176.

A single immunization with recombinant rabies virus (ERAG3G) confers complete protection against rabies in mice

Affiliations
  • 1Viral Disease Division, Animal and Plant Quarantine Agency, MAFRA, Anyang, Korea. yangdk@korea.kr
  • 2The United Graduated School of Veterinary Science, Gifu University, Gifu, Japan.

Abstract

PURPOSE
New alternative bait rabies vaccines applicable to pet dogs and wild animals are needed to eradicate rabies in Korea. In this study, recombinant rabies virus, ERAG3G strain was constructed using reverse genetic system and the safety, efficacy and immunogenicity of the ERAG3G strain was evaluated in mice and dogs.
MATERIALS AND METHODS
Using the full-length genome mutated amino acid at position 333 of glycoprotein of rabies virus (RABV) and helper plasmids, the ERAG3G strain was rescued in BHK/T7-9 cells successfully. Mice were inoculated with the ERAG3G strain for safety and efficacy. Safety and immunogenicity of the dog inoculated with the ERAG3G strain (1 mL, 10(8.0) FAID50/mL) via intramuscular route was evaluated for 28 days after inoculation.
RESULTS
The ERAG3G strain rescued by reverse genetic system was propagated well in the mouse neuroblastoma cells revealing titer of 10(8.5) FAID50/mL and was not pathogenic to 4- or 6-week-old mice that received by intramuscular or intracranical route. Immunization with the ERAG3G strain conferred complete protection from lethal RABV in mice. Dogs inoculated with the vaccine candidate via intramuscular route showed high neutralizing antibody titer ranging from 2.62 to 23.9 IU/mL at 28 days postinoculation.
CONCLUSION
Our findings suggest that the ERAG3G strain plays an important role in inducing protective efficacy in mice and causes to arise anti-rabies neutralizing antibody in dogs.

Keyword

Rabies virus; Recombinant rabies virus; Vaccine; Animals

MeSH Terms

Animals
Animals, Wild
Antibodies, Neutralizing
Dogs
Genome
Glycoproteins
Immunization*
Korea
Mice*
Neuroblastoma
Plasmids
Rabies Vaccines
Rabies virus*
Rabies*
Antibodies, Neutralizing
Glycoproteins
Rabies Vaccines

Figure

  • Fig. 1 Vector map for the construction of rERA and ERAG3G strain. These full-length cDNA plasmids and three helper plasmids (N, P, and L plasmid) were transfected into BHK/T7-9 cells producing RNA polymerase, respectively.

  • Fig. 2 Cytopathic effects observed in BHK/T7-9 cell inoculated with rERA (A, ×100) and ERAG3G (B, ×200) and immunofluorescence rERA (C, ×200) and ERAG3G (D, ×100) strain by indirect fluorescent assay test using monoclonal antibodies against rabies virus (RABV) in BHK/T7-9 cells. The RABV specific fluorescent appeared in the cytoplasm of the infected cell.

  • Fig. 3 Rabies virus particles in cytoplasm of BHK-T7 cells inoculated with rERA (A, ×50,000) and ERAG3G (B, ×50,000) strains showing bullet shape.

  • Fig. 4 Change in body weight in 4-week-old (A, B) and 6-week-old mice (C, D) inoculated with ERA, rERA, and ERAG3G strains via intramuscular (IM; A, C) or intracranial (IC; B, D) route.

  • Fig. 5 Safety test in 4-week-old (A, B) and 6-week-old mice (C, D) inoculated with ERA, rERA, and ERAG3G strains via intramuscular (IM; A, C) or intracranial (IC; B, D) route. The 4-week-old mice inoculated with ERA and rERA strains respectively died at 12 or 13 days postinoculation. Fifty percent of the 6-week-old mice inoculated with ERA and rERA IM were alive and all of the mice inoculated with ERA and rERA IC died at 13 days postinoculation. On the contrary, both 4- and 6-week-old mice inoculated with ERAG3G IM or IC were 100% alive as control group.

  • Fig. 6 Change of body weight (A) and survival rate (B) in 4- and 6-week-old mice immunized with ERAG3G strain via intramuscular (IM) or intracranial (IC) route and challenged with highly pathogenic rabies virus strain (CVSN2c). The average body weight of ERAG3G group mice increased for 17 days after challenge and survival rates did not change for 17 days after challenge.

  • Fig. 7 Immune response in dogs inoculated with the ERAG3G strain via intramuscular route. All dogs immunized with the ERAG3G strain induced high neutralizing antibody titer >0.5 IU/mL against rabies virus. VN, virus neutralizing.


Cited by  4 articles

Oral immunization of mice with recombinant rabies vaccine strain (ERAG3G) induces complete protection
Dong-Kun Yang, Ha-Hyun Kim, Sung-Suk Choi, Jong-Taek Kim, Woong-Ho Jeong, Jae-Young Song
Clin Exp Vaccine Res. 2015;4(1):107-113.    doi: 10.7774/cevr.2015.4.1.107.

A recombinant rabies virus (ERAGS) for use in a bait vaccine for swine
Dong-Kun Yang, Ha-Hyun Kim, Sung-Suk Choi, Seong Heon Lee, In-Soo Cho
Clin Exp Vaccine Res. 2016;5(2):169-174.    doi: 10.7774/cevr.2016.5.2.169.

Safety and immunogenicity of recombinant rabies virus (ERAGS) in mice and raccoon dogs
Dong-Kun Yang, Ha-Hyun Kim, Sung-Suk Choi, Jong-Tack Kim, Kang-Bok Lee, Seong Heon Lee, In-Soo Cho
Clin Exp Vaccine Res. 2016;5(2):159-168.    doi: 10.7774/cevr.2016.5.2.159.

Safety and Immunogenicity of a Recombinant Rabies Virus Strain (ERAG3G) in Korean Raccoon Dogs
Dong-Kun Yang, Ha-Hyun Kim, Hyun-Ye Jo, Hee-Won Kim, Sung-Suk Choi, In-Soo Cho
J Bacteriol Virol. 2015;45(3):250-255.    doi: 10.4167/jbv.2015.45.3.250.


Reference

1. Johnson N, Black C, Smith J, et al. Rabies emergence among foxes in Turkey. J Wildl Dis. 2003; 39:262–270.
Article
2. Yang DK, Kim SY, Oh YI, et al. Epidemiological characteristics of rabies in South Korea from January 2004 to March 2011. J Bacteriol Virol. 2011; 41:165–171.
Article
3. Orciari LA, Niezgoda M, Hanlon CA, et al. Rapid clearance of SAG-2 rabies virus from dogs after oral vaccination. Vaccine. 2001; 19:4511–4518.
Article
4. Faber M, Dietzschold B, Li J. Immunogenicity and safety of recombinant rabies viruses used for oral vaccination of stray dogs and wildlife. Zoonoses Public Health. 2009; 56:262–269.
Article
5. Baer GM, Abelseth MK, Debbie JG. Oral vaccination of foxes against rabies. Am J Epidemiol. 1971; 93:487–490.
Article
6. Muller TF, Schuster P, Vos AC, Selhorst T, Wenzel UD, Neubert AM. Effect of maternal immunity on the immune response to oral vaccination against rabies in young foxes. Am J Vet Res. 2001; 62:1154–1158.
Article
7. Cliquet F, Gurbuxani JP, Pradhan HK, et al. The safety and efficacy of the oral rabies vaccine SAG2 in Indian stray dogs. Vaccine. 2007; 25:3409–3418.
Article
8. Fehlner-Gardiner C, Rudd R, Donovan D, Slate D, Kempf L, Badcock J. Comparing ONRAB(R) AND RABORAL V-RG(R) oral rabies vaccine field performance in raccoons and striped skunks, New Brunswick, Canada, and Maine, USA. J Wildl Dis. 2012; 48:157–167.
Article
9. Taylor J, Meignier B, Tartaglia J, et al. Biological and immunogenic properties of a canarypox-rabies recombinant, ALVAC-RG (vCP65) in non-avian species. Vaccine. 1995; 13:539–549.
Article
10. Rupprecht CE, Blass L, Smith K, et al. Human infection due to recombinant vaccinia-rabies glycoprotein virus. N Engl J Med. 2001; 345:582–586.
Article
11. Franka R, Wu X, Jackson FR, et al. Rabies virus pathogenesis in relationship to intervention with inactivated and attenuated rabies vaccines. Vaccine. 2009; 27:7149–7155.
Article
12. Inoue K, Shoji Y, Kurane I, Iijima T, Sakai T, Morimoto K. An improved method for recovering rabies virus from cloned cDNA. J Virol Methods. 2003; 107:229–236.
Article
13. Ito N, Takayama-Ito M, Yamada K, Hosokawa J, Sugiyama M, Minamoto N. Improved recovery of rabies virus from cloned cDNA using a vaccinia virus-free reverse genetics system. Microbiol Immunol. 2003; 47:613–617.
Article
14. Yang DK, Oh YI, Cho SD, et al. Molecular identification of the vaccine strain from the inactivated rabies vaccine. J Bacteriol Virol. 2011; 41:47–54.
Article
15. Cliquet F, Aubert M, Sagne L. Development of a fluorescent antibody virus neutralisation test (FAVN test) for the quantitation of rabies-neutralising antibody. J Immunol Methods. 1998; 212:79–87.
Article
16. Fu ZF. The rabies situation in Far East Asia. Dev Biol (Basel). 2008; 131:55–61.
17. Streicker DG, Altizer SM, Velasco-Villa A, Rupprecht CE. Variable evolutionary routes to host establishment across repeated rabies virus host shifts among bats. Proc Natl Acad Sci U S A. 2012; 109:19715–19720.
Article
18. Cliquet F, Aubert M. Elimination of terrestrial rabies in Western European countries. Dev Biol (Basel). 2004; 119:185–204.
19. Schnell MJ, Mebatsion T, Conzelmann KK. Infectious rabies viruses from cloned cDNA. EMBO J. 1994; 13:4195–4203.
Article
20. Takayama-Ito M, Inoue K, Shoji Y, et al. A highly attenuated rabies virus HEP-Flury strain reverts to virulent by single amino acid substitution to arginine at position 333 in glycoprotein. Virus Res. 2006; 119:208–215.
Article
21. Tuffereau C, Leblois H, Benejean J, Coulon P, Lafay F, Flamand A. Arginine or lysine in position 333 of ERA and CVS glycoprotein is necessary for rabies virulence in adult mice. Virology. 1989; 172:206–212.
Article
22. Bankovskiy D, Safonov G, Kurilchuk Y. Immunogenicity of the ERA G 333 rabies virus strain in foxes and raccoon dogs. Dev Biol (Basel). 2008; 131:461–466.
Full Text Links
  • CEVR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr