research

Construction and characterization of H5N1-recombinant fowlpox viruses co-expressing host cytokines

Abstract

Possessing a large double stranded DNA genome up to 300 kb, fowlpox virus (FWPV) has been developed to express avian influenza virus (AIV) antigens since the late 1980s. A more advanced approach would be to coexpress host cytokines from such recombinants. This thesis describes the strategy to construct H5N1-recombinant FWPV (rFWPV) coexpressing chicken Interleukin 12 (IL-12) or Interleukin 15 (IL-15), and discusses the immunogenicity of the recombinants following inoculation into specific-pathogen-free (SPF) chickens. Previously cloned and sequenced cDNAs encoding full-length H5 and N1 of influenza strain A/Chicken/Malaysia/5858/2004 genes were amplified by PCR and inserted into plasmid pEFL29, under the control of a copy of the vaccinia virus p7.5 early/late promoter. The expression cassettes were recombined into the genome of the FP9 strain of FWPV at the fpv002 locus. Recombinant viruses were produced by transfection of the plasmid into chicken embryo fibroblasts (CEFs) after infection with FP9, and isolated by six fold plaque purification on CEFs using X-Gal selection. Chicken IL-12 or IL-15 genes, under control of a synthetic/hybrid poxvirus promoter, were inserted into a ‘transient dominant selection’ recombination plasmid, pPC1.X. The cytokine expression cassettes were then recombined, at the fpPC1 (fpv030) locus, into rFWPV already carrying AIV genes. Following three rounds of passage in CEFs in the presence of mycophenolic acid (MPA), recombinant viruses carrying the gpt gene were isolated. These unstable recombinants were plaque-purified in the absence of MPA until they lost the gpt gene spontaneously, verified by their failure to replicate in the presence of MPA. Recombinant proteins were successfully detected using western blotting and indirect immunofluorescence assay (IFAT). Parental and rFWPV (105 PFU) were inoculated subcutaneously into one-day-old SPF chickens. Sera from chickens immunized with rFWPV/H5 and rFWPV/H5/IL-15 demonstrated viral neutralizing activities, based on the haemagglutation inhibition (HI) test, in which reached a peak at Week 3. A competitive enzyme-linked immunosorbent (ELISA) assay detected N1-specific antibodies induced by rFWPV/N1 and rFWPV/N1/IL-12 at Weeks 4 and 5. Non-specific cellular immune responses were assessed by flow cytometric analysis to enumerate CD4+ and CD8+ T-lymphocytes in peripheral blood. Results of Experiment 2 showed chickens vaccinated with rFWPV/H5, rFWPV/H5/IL-15, rFWPV/N1 and rFWPV/N1/IL-12 demonstrated a higher increase in CD8+ than CD4+ T cell population, relative to control and chickens vaccinated with parental FWPV. Weekly weighing showed that chickens vaccinated with rFWPV/H5/IL-15 had the highest body weight compared to other groups, while the rFWPV/N1/IL-12 group showed the significantly lowest body weight. In summary, this study showed diverse immunogenicity of H5N1-rFWPV coexpressing IL-12 or IL-15. It also demonstrated a weight sparing effect of co-expressing IL-15 in rFWPV vaccines. The results provide the basis for future homologous challenge studies, using live H5N1 virus to evaluate the protective efficacy of the rFWPV vaccines

    Similar works