scholarly journals A Novel Cre Recombinase-MediatedIn VivoMinicircle DNA (CRIM) Vaccine Provides Partial Protection against Newcastle Disease Virus

2019 ◽  
Vol 85 (14) ◽  
Author(s):  
Yanlong Jiang ◽  
Xing Gao ◽  
Ke Xu ◽  
Jianzhong Wang ◽  
Haibin Huang ◽  
...  

ABSTRACTMinicircle DNA (mcDNA), which contains only the necessary components for eukaryotic expression and is thus smaller than traditional plasmids, has been designed for application in genetic manipulation. In this study, we constructed a novel plasmid containing both the Cre recombinase under the phosphoglycerate kinase (PGK) promoter and recombinantlox66andlox71sites located outside the cytomegalovirus (CMV) expression cassette. The strictly controlled synthesis of Cre recombinasein vivomaintained the complete form of the plasmidin vitro, whereas thein vivoproduction of Cre transformed the parental plasmid to mcDNA after transfection. The newly designedCrerecombinase-mediatedin vivomcDNA platform, named CRIM, significantly increased the nuclear entry of mcDNA, followed by increased production of mRNA and protein, using enhanced green fluorescent protein (EGFP) as a model. Similar results were also observed in chickens when the vaccine was delivered by the regulated-delayed-lysisSalmonellastrain χ11218, where significantly increased production of EGFP was observed in chicken livers. Then, we used the HN gene of genotype VII Newcastle disease virus as an antigen model to construct the traditional plasmid pYL43 and the novel mcDNA plasmid pYL47. After immunization, our CRIM vaccine provided significantly increased protection against challenge compared with that of the traditional plasmid, providing us with a novel mcDNA vaccine platform.IMPORTANCEMinicircle DNA (mcDNA) has been considered an attractive alternative to DNA vaccines; however, the relatively high cost and complicated process of purifying mcDNA dramatically restricts the application of mcDNA in the veterinary field. We designed a novelin vivomcDNA platform in which the complete plasmid could spontaneously transform into mcDNAin vivo. In combination with the regulated-delayed-lysisSalmonellastrain, the newly designed mcDNA vaccine provides us with an elegant platform for veterinary vaccine development.

2008 ◽  
Vol 83 (2) ◽  
pp. 584-597 ◽  
Author(s):  
Elena Carnero ◽  
Wenjing Li ◽  
Antonio V. Borderia ◽  
Bruno Moltedo ◽  
Thomas Moran ◽  
...  

ABSTRACT One attractive strategy for the development of a human immunodeficiency virus (HIV) vaccine is the use of viral vectors with a proven safety profile and an absence of preexisting immunity in humans, such as Newcastle disease virus (NDV). Several NDV vaccine vectors have been generated, and their immunogenicities have been investigated with different animal models. However, a systematic study to evaluate the optimal insertion site of the foreign antigens into NDV that results in enhanced immune responses specific to the antigen has not yet been conducted. In this article, we describe the ability of NDV expressing HIV Gag to generate a Gag-specific immune response in mice. We also have determined the optimal insertion site into the NDV genome by generating recombinant NDV-HIVGag viruses in which HIV gag was located at different transcriptional positions throughout the NDV viral genome. All recombinant viruses were viable, grew to similar titers in embryonated chicken eggs, and expressed Gag in a stable manner. Our in vivo experiments revealed that higher HIV Gag protein expression positively correlates with an enhanced CD8+ T-cell-mediated immune response and protective immunity against challenge with vaccinia virus expressing HIV Gag. We also inserted a codon-optimized version of HIV gag in the described best location, between the P and M genes. Virus expressing the codon-optimized version of HIV gag induced a higher expression of the protein and an enhanced immune response against HIV Gag in mice. These results indicate that strategies directed toward increasing antigen expression by NDV result in enhanced immunogenicity and vaccine efficacy.


2004 ◽  
Vol 17 (4) ◽  
pp. 543-547 ◽  
Author(s):  
I. K. Park ◽  
H. L. Jiang ◽  
C. H. Yun ◽  
Y. J. Choi ◽  
S. J. Kim ◽  
...  

BioTechniques ◽  
2020 ◽  
Vol 68 (2) ◽  
pp. 96-100
Author(s):  
Pheik-Sheen Cheow ◽  
Tiong Kit Tan ◽  
Adelene Ai-Lian Song ◽  
Khatijah Yusoff ◽  
Suet Lin Chia

Reverse genetics has been used to generate recombinant Newcastle disease virus with enhanced immunogenic properties for vaccine development. The system, which involves co-transfecting the viral antigenomic plasmid with three helper plasmids into a T7 RNA polymerase-expressing cell to produce viral progenies, poses a great challenge. We have modified the standard transfection method to improve the transfection efficiency of the plasmids, resulting in a higher titer of virus progeny production. Two transfection reagents (i.e., lipofectamine and polyethylenimine) were used to compare the transfection efficiency of the four plasmids. The virus progenies produced were quantitated with flow cytometry analysis of the infectious virus unit. The modified transfection method increased the titer of virus progenies compared with that of the standard transfection method.


2009 ◽  
Vol 83 (11) ◽  
pp. 5943-5946 ◽  
Author(s):  
Yongqi Yan ◽  
Subrat N. Rout ◽  
Shin-Hee Kim ◽  
Siba K. Samal

ABSTRACT To determine the role of untranslated regions (UTRs) in replication and pathogenesis of Newcastle disease virus (NDV), we generated recombinant viruses with deletions in 5′ and 3′ UTRs of the HN mRNA. Deletion of any HN UTR did not noticeably affect in vitro replication of these viruses. However, complete deletion of the 5′ UTR of the HN gene decreased the HN mRNA levels and HN protein contents in virus particles, resulting in attenuation of the virus in chickens. This indicates that the 5′ UTR of HN mRNA plays an important role in replication and pathogenicity of NDV in vivo.


Sign in / Sign up

Export Citation Format

Share Document