PLoS Pathogens (Jul 2021)

Pore-forming alpha-hemolysin efficiently improves the immunogenicity and protective efficacy of protein antigens.

  • Jin-Tao Zou,
  • Hai-Ming Jing,
  • Yue Yuan,
  • Lang-Huan Lei,
  • Zhi-Fu Chen,
  • Qiang Gou,
  • Qing-Shan Xiong,
  • Xiao-Li Zhang,
  • Zhuo Zhao,
  • Xiao-Kai Zhang,
  • Hao Zeng,
  • Quan-Ming Zou,
  • Jin-Yong Zhang

DOI
https://doi.org/10.1371/journal.ppat.1009752
Journal volume & issue
Vol. 17, no. 7
p. e1009752

Abstract

Read online

Highly immunogenic exotoxins are used as carrier proteins because they efficiently improve the immunogenicity of polysaccharides. However, their efficiency with protein antigens remains unclear. In the current study, the candidate antigen PA0833 from Pseudomonas aeruginosa was fused to the α-hemolysin mutant HlaH35A from Staphylococcus aureus to form a HlaH35A-PA0833 fusion protein (HPF). Immunization with HPF resulted in increased PA0833-specific antibody titers, higher protective efficacy, and decreased bacterial burden and pro-inflammatory cytokine secretion compared with PA0833 immunization alone. Using fluorescently labeled antigens to track antigen uptake and delivery, we found that HlaH35A fusion significantly improved antigen uptake in injected muscles and antigen delivery to draining lymph nodes. Both in vivo and in vitro studies demonstrated that the increased antigen uptake after immunization with HPF was mainly due to monocyte- and macrophage-dependent macropinocytosis, which was probably the result of HPF binding to ADAM10, the Hla host receptor. Furthermore, a transcriptome analysis showed that several immune signaling pathways were activated by HPF, shedding light on the mechanism whereby HlaH35A fusion improves immunogenicity. Finally, the improvement in immunogenicity by HlaH35A fusion was also confirmed with two other antigens, GlnH from Klebsiella pneumoniae and the model antigen OVA, indicating that HlaH35A could serve as a universal carrier protein to improve the immunogenicity of protein antigens.