Outer membrane vesicles from genetically engineered Salmonella enterica serovar Typhimurium presenting Helicobacter pylori antigens UreB and CagA induce protection against Helicobacter pylori infection in mice

doi: 10.4121/013c1924-6914-441f-ad7b-da69d55d15da.v1
The doi above is for this specific version of this collection, which is currently the latest. Newer versions may be published in the future. For a link that will always point to the latest version, please use
doi: 10.4121/013c1924-6914-441f-ad7b-da69d55d15da
Datacite citation style:
Qiong, Qiong (2024): Outer membrane vesicles from genetically engineered Salmonella enterica serovar Typhimurium presenting Helicobacter pylori antigens UreB and CagA induce protection against Helicobacter pylori infection in mice. Version 1. 4TU.ResearchData. collection. https://doi.org/10.4121/013c1924-6914-441f-ad7b-da69d55d15da.v1
Other citation styles (APA, Harvard, MLA, Vancouver, Chicago, IEEE) available at Datacite
Collection

Helicobacter pylori is a globally prevalent infection that is highly related to chronic gastritis and even causes the development of gastric carcinoma. With the rapid increase of antibiotic resistance, scientists have begun to search for better vaccine design strategies to eradicate H. pylori colonization. However, while current strategies prefer to formulate vaccines with a single H. pylori antigen, their potential has not yet been fully realized. Outer membrane vesicles (OMVs) are a potential vaccine platform since they could deliver multiple antigens. In this study, we engineered three crucial H. pylori antigen proteins (UreB, CagA, and VacA) onto the surface of OMVs derived from Salmonella enterica serovar Typhimurium mutant strains using the hemoglobin protease (Hbp) autotransporter system. In various knockout strategies, we found that OMVs isolated from the ΔrfbP ΔfliC ΔfljB ΔompA mutants could cause distinct increases in immunoglobulin G (IgG) and A (IgA) levels and effectively trigger T helper 1- and 17-biased cellular immune responses, which perform a vital role in protecting against H. pylori. Next, OMVs derived from ΔrfbP ΔfliC ΔfljB ΔompA mutants were used as a vector to deliver different combinations of H. pylori antigens. The antibody and cytokine levels and challenge experiments in mice model indicated that co-delivering UreB and CagA could protect against H. pylori and antigen-specific T cell responses. In summary, adopting OMVs derived from the ΔrfbP ΔfliC ΔfljB ΔompA mutant strain as the vector while importing UreB and CagA as antigens using the Hbp autotransporter system would greatly benefit controlling H. pylori infection.

history
  • 2024-05-02 first online, published, posted
publisher
4TU.ResearchData

DATASETS