Immunogenicity of a DNA vaccine for coxsackievirus B3 in mice: protective effects of capsid proteins against viral challenge

Vaccine ◽  
2005 ◽  
Vol 23 (14) ◽  
pp. 1672-1679 ◽  
Author(s):  
Joo-Young Kim ◽  
Eun-Seok Jeon ◽  
Byung-Kwan Lim ◽  
Sun-Mi Kim ◽  
Sun-Ku Chung ◽  
...  
2007 ◽  
Vol 293 (1) ◽  
pp. H69-H76 ◽  
Author(s):  
Yi-Xin Wang ◽  
Valdeci da Cunha ◽  
Jon Vincelette ◽  
Kathy White ◽  
Sharlene Velichko ◽  
...  

The present study tested the hypothesis that murine (m)IFN-β or mIFN-α2can eliminate cardiac viral load and protect cardiomyocytes from injury in animals infected with coxsackievirus B3 (CVB3). CVB3-inoculated male Balb/c mice exhibited signs of illness, including lethargy, progressive weight loss, and death (10% on day 3 and 100% on day 8). Cardiac viral load was high [4,277 ± 1,009 plaque-forming units and 25 ± 5 copies CVB3/hypoxanthine guanine phosphoribosyl transferase 1 mRNA] on day 4. The cardiac tissue exhibited severe inflammatory infiltration and myocyte damage with an average myocarditis integrated pathology score of 2.1 ± 0.2 on day 7. Most of the mice infected with CVB3 also developed epicarditis, and 55% had intraventricular thrombi present. Treatment with mIFN-β [2.5 to 10 million international units (MIU)/kg] dose-dependently improved the general health status in CVB3-inoculated mice, as evidenced by reduction in weight loss, prevention of death, elimination of cardiac viral load, protection of myocytes from injury, decrease in inflammatory cell infiltration, and attenuation of intraventricular thrombus formation. Treatment with 10 MIU/kg mIFN-α2resulted in a similar level of efficacy as that induced by 5 MIU/kg mIFN-β, with the exception that mIFN-α2did not reduce cardiac CVB3 mRNA. However, mIFN-α2, but not any dose group of mIFN-β, significantly attenuated CVB3-induced epicarditis. These data demonstrate antiviral effects for both mIFN-β and mIFN-α2, which lead to protection of the mice from CVB3-induced myocarditis. However, the potential mechanisms leading to a differential host response for the two isoforms of mIFN remain to be elucidated.


2013 ◽  
Vol 20 (11) ◽  
pp. 1743-1751 ◽  
Author(s):  
Maowei Wang ◽  
Yan Yue ◽  
Chunsheng Dong ◽  
Xiaoyun Li ◽  
Wei Xu ◽  
...  

ABSTRACTCoxsackievirus B3 (CVB3), a small single-stranded RNA virus, belongs to thePicornaviridaefamily. Its infection is the most common cause of myocarditis, with no vaccine available. Gastrointestinal mucosa is the major entry port for CVB3; therefore, the induction of local immunity in mucosal tissues may help control initial viral infections and alleviate subsequent myocardial injury. Here we evaluated the ability of high-mobility group box 1 (HMGB1) encapsulated in chitosan particles to enhance the mucosal immune responses induced by the CVB3-specific mucosal DNA vaccine chitosan-pVP1. Mice were intranasally coimmunized with 4 doses of chitosan-pHMGB1 and chitosan-pVP1 plasmids, at 2-week intervals, and were challenged with CVB3 4 weeks after the last immunization. Compared with chitosan-pVP1 immunization alone, coimmunization with chitosan-pHMGB1 significantly (P< 0.05) enhanced CVB3-specific fecal secretory IgA levels and promoted mucosal T cell immune responses. In accordance, reduced severity of myocarditis was observed in coimmunized mice, as evidenced by significantly (P< 0.05) reduced viral loads, decreased myocardial injury, and increased survival rates. Flow cytometric analysis indicated that HMGB1 enhanced dendritic cell (DC) recruitment to mesenteric lymph nodes and promoted DC maturation, which might partly account for its mucosal adjuvant effect. This strategy may represent a promising approach to candidate vaccines against CVB3-induced myocarditis.


PLoS ONE ◽  
2014 ◽  
Vol 9 (1) ◽  
pp. e86578 ◽  
Author(s):  
Xiaoting Wang ◽  
Yang Dai ◽  
Song Zhao ◽  
Jianxia Tang ◽  
Hongjun Li ◽  
...  

Life Sciences ◽  
2013 ◽  
Vol 92 (24-26) ◽  
pp. 1186-1194 ◽  
Author(s):  
Yuan-Yuan Zhang ◽  
Jin-Nan Li ◽  
Harry Hua-Xiang Xia ◽  
Shi-Lun Zhang ◽  
Jiang Zhong ◽  
...  

Parasitology ◽  
2005 ◽  
Vol 132 (1) ◽  
pp. 67-71 ◽  
Author(s):  
Y. ZHU ◽  
J. SI ◽  
D. A. HARN ◽  
M. XU ◽  
J. REN ◽  
...  

The protective efficacy of a Schistosoma japonicum, Chinese strain, triose-phosphate isomerase (TPI) plasmid DNA vaccine was examined in naïve pigs. Pigs were vaccinated with the TPI DNA-plasmid alone, or in conjunction with IL-12 as pcDNA3.1-P35, pcDNA3.1-P40 plasmids via intramuscular injection. Control pigs were immunized with equivalent amounts of pcDNA3.1. Pigs were immunized 3 times at 21-day intervals and challenged 30 days after the final boost. Forty-five days post-challenge, pigs were sacrificed and perfused to compare adult worm burdens, female worm burdens, liver egg burdens and granuloma size. We found that pigs vaccinated with SjCTPI DNA alone had adult worm burdens reduced by 48·3% and that a further decrease in adult worm burdens was not seen in the group vaccinated with SjCTPI DNA in conjunction with IL-12 (46·2% reduction). The SjCTPI DNA vaccines had a more pronounced effect on reducing female worm burdens i.e. 53·6% SjCTPI alone and 59·6% for SjCTPI+IL-12. Vaccination with SjCTPI-DNA reduced liver eggs by 49·4% and this response was significantly enhanced by the addition of IL-12 (65·8% reduction in liver eggs). In addition to the dramatic protective effects seen in vaccinated pigs, we also noted that granuloma size was reduced by 42% in both groups. Thus, vaccination of pigs and other large animals in China with SjCTPI DNA vaccine will likely reduce transmission by reducing adult worm burdens and worm egg output and simultaneously reduce hepatic egg-associated pathology.


Sign in / Sign up

Export Citation Format

Share Document