Abstract

Helicobacter pylori (H. pylori) is a gram negative bacterium that infects more than 50% of humanity and is associated with gastritis, peptic ulcer and gastric cancer. Although CD4+ T cells are recruited to the gastric mucosa, the host is unable to clear the bacteria. Previously, we demonstrated that H. pylori infection upregulates the expression of the T cell co-inhibitory molecule B7-H1 while simultaneously downregulating the expression of T cell co-stimulatory molecule B7-H2 on gastric epithelial cells (GEC), which together affect the Treg and Th17 cell balance and foster bacterial persistence. Because B7-H3, another member of the B7 family of co-inhibitory receptors, has been found to have important immunoregulatory roles and in cancer, in this study we examined the expression of B7-H3 molecules on GEC and how the expression is regulated by H. pylori during infection. Our study showed that both human and murine GEC constitutively express B7-H3 molecules, but their expression levels increased during H. pylori infection. We further demonstrated that H. pylori uses its type 4 secretion system (T4SS) components CagA and cell wall peptidoglycan (PG) fragment to upregulate B7-H3. Th17 cells and Treg cells which are increased during H. pylori infection also had an effect on B7-H3 induction. The underlying cell signaling pathway involves modulation of p38MAPK pathway. Since B7-H3 were shown to up-regulate Th2 responses, the phenotype of T cell subpopulations in mice infected with H. pylori PMSS1 or SS1 strains were characterized. A mixed Th1/Th2 response in H. pylori infected mice was observed. Consistent with previous findings, increased Treg cells and decreased Th17 cells in MLN of PMSS1 infected mice compared to SS1 infected mice was observed. Human biopsy samples collected from gastritis biopsies and gastric tumors showed a strong association between increased B7-H3 and Th2 responses in H. pylori strains associated with gastritis. T cell: GEC co-cultures and anti-B7-H3 blocking Ab confirmed that the induction of Th2 is mediated by B7-H3 and associated exclusively with an H. pylori gastritis strain not cancer or ulcer strains. In conclusion, these studies revealed a novel regulatory mechanism employed by H. pylori to influence the type of T cell response that develops within the infected gastric mucosa.

Highlights

  • Helicobacter pylori (H. pylori) colonizes the human gastric mucosa and may induce gastritis, peptic ulcer and two forms of neoplasia: gastric adenocarcinoma and mucosa-associated lymphoid tissue (MALT) lymphoma [1]

  • We further demonstrated that H. pylori uses its type 4 secretion system (T4SS) components CagA and cell wall peptidoglycan (PG) fragment to upregulate B7-H3

  • In addition to CagA, H. pylori translocates via the T4SS its cell wall peptidoglycan (PG) fragments, which are recognized by intracellular pattern recognition receptor NOD1 and activates MAPKs and NFkB pathways [14,15,16]

Read more

Summary

Introduction

Helicobacter pylori (H. pylori) colonizes the human gastric mucosa and may induce gastritis, peptic ulcer and two forms of neoplasia: gastric adenocarcinoma and mucosa-associated lymphoid tissue (MALT) lymphoma [1]. Upon attachment of H. pylori to gastric epithelial cells (GEC), CagA is injected via the T4SS and becomes phosphorylated in the tyrosine residue of their EPIYA motifs by host Src kinases and c-Ab1 [6,7,8,9,10]. Both phosphorylated and unphosphorylated forms of CagA can interact with a range of host cell signaling proteins and activates them, which results in several physiological changes in GECs [11,12,13]. In addition to CagA, H. pylori translocates via the T4SS its cell wall peptidoglycan (PG) fragments, which are recognized by intracellular pattern recognition receptor NOD1 and activates MAPKs and NFkB pathways [14,15,16]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call