首页> 外文期刊>Journal of neurosurgical sciences >Gold nanoparticle-adjuvanted S protein induces a strong antigen-specific IgG response against severe acute respiratory syndrome-related coronavirus infection, but fails to induce protective antibodies and limit eosinophilic infiltration in lungs
【24h】

Gold nanoparticle-adjuvanted S protein induces a strong antigen-specific IgG response against severe acute respiratory syndrome-related coronavirus infection, but fails to induce protective antibodies and limit eosinophilic infiltration in lungs

机译:金纳米粒子辅助的S蛋白诱导强烈的抗原特异性IgG反应,针对严重急性呼吸综合征相关的冠状病毒感染,但不能诱导保护性抗体并限制肺部嗜酸性嗜嗜酸性渗透

获取原文
获取原文并翻译 | 示例
       

摘要

The spike (S) protein of coronavirus, which binds to cellular receptors and mediates membrane fusion for cell entry, is a candidate vaccine target for blocking coronavirus infection. However, some animal studies have suggested that inadequate immunization against severe acute respiratory syndrome coronavirus (SARS-CoV) induces a lung eosinophilic immunopathology upon infection. The present study evaluated two kinds of vaccine adjuvants for use with recombinant S protein: gold nanoparticles (AuNPs), which are expected to function as both an antigen carrier and an adjuvant in immunization; and Toll-like receptor (TLR) agonists, which have previously been shown to be an effective adjuvant in an ultraviolet-inactivated SARS-CoV vaccine. All the mice immunized with more than 0.5 mu g S protein without adjuvant escaped from SARS after infection with mouse-adapted SARS-CoV; however, eosinophilic infiltrations were observed in the lungs of almost all the immunized mice. The AuNP-adjuvanted protein induced a strong IgG response but failed to improve vaccine efficacy or to reduce eosinophilic infiltration because of highly allergic inflammatory responses. Whereas similar virus titers were observed in the control animals and the animals immunized with S protein with or without AuNPs, Type 1 interferon and pro-inflammatory responses were moderate in the mice treated with S protein with and without AuNPs. On the other hand, the TLR agonist-adjuvanted vaccine induced highly protective antibodies without eosinophilic infiltrations, as well as Th1/17 cytokine responses. The findings of this study will support the development of vaccines against severe pneumonia-associated coronaviruses.
机译:与细胞受体结合的冠状病毒的穗蛋白质和介导细胞入口的膜融合,是用于阻断冠状病毒感染的候选疫苗靶标。然而,一些动物研究表明,对严重急性呼吸综合征冠状病毒(SARS-COV)免疫不足诱导肺嗜酸性免疫病理学。本研究评估了两种疫苗佐剂,用于重组的蛋白质:金纳米颗粒(AUNP),预期用作抗原载体和免疫中的佐剂;和类似的受体(TLR)激动剂,其先前已被证明是紫外线灭活SARS-COV疫苗中的有效佐剂。在用小鼠适应的SARS-COV感染后,所有没有佐剂的蛋白质免疫没有佐剂免疫的小鼠。然而,在几乎所有免疫小鼠的肺部中观察到嗜酸性渗透。 AUNP辅助蛋白质诱导强烈的IgG响应,但由于高度过敏反应,未能改善疫苗疗效或降低嗜酸性渗透。然而,在对照动物中观察到类似的病毒滴度,并且用蛋白质免疫的动物免疫有或没有AUNPS,型干扰素和促炎反应在用S蛋白质处理的小鼠中适度,具有和无肛周。另一方面,TLR激动剂 - 佐剂疫苗诱导高度保护性抗体而没有嗜酸性渗透,以及TH1 / 17细胞因子反应。该研究的结果将支持对严重肺炎相关冠状病毒的疫苗的发展。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号