Abstract

Helicobacter pylori is a gram-negative, microaerophilic, and spiral-shaped bacterium and causes gastrointestinal diseases in human. IL-1β is a representative cytokine produced in innate immune cells and is considered to be a key factor in the development of gastrointestinal malignancies. However, the mechanism of IL-1β production by neutrophils during H. pylori infection is still unknown. We designed this study to identify host and bacterial factors involved in regulation of H. pylori-induced IL-1β production in neutrophils. We found that H. pylori-induced IL-1β production is abolished in NLRP3-, ASC-, and caspase-1/11-deficient neutrophils, suggesting essential role for NLRP3 inflammasome in IL-1β response against H. pylori. Host TLR2, but not TLR4 and Nod2, was also required for transcription of NLRP3 and IL-1β as well as secretion of IL-1β. H. pylori lacking cagL, a key component of the type IV secretion system (T4SS), induced less IL-1β production in neutrophils than did its isogenic WT strain, whereas vacA and ureA were dispensable. Moreover, T4SS was involved in caspase-1 activation and IL-1β maturation in H. pylori-infected neutrophils. We also found that FlaA is essential for H. pylori-mediated IL-1β production in neutrophils, but not dendritic cells. TLR5 and NLRC4 were not required for H. pylori-induced IL-1β production in neutrophils. Instead, bacterial motility is essential for the production of IL-1β in response to H. pylori. In conclusion, our study shows that host TLR2 and NLRP3 inflammasome and bacterial T4SS and motility are essential factors for IL-1β production by neutrophils in response to H. pylori.

Highlights

  • Helicobacter pylori (H. pylori) is a gram-negative, microaerophilic, and spiral bacterium that colonizes the human gastric mucosa

  • These findings suggest that the NLRP3/ASC/caspase-1 axis is essential for H. pyloriinduced Interleukin 1β (IL-1β) production in neutrophils

  • We identified that NLRP3 inflammasome is a key host factor in neutrophils for production of IL-1β in response to H. pylori by showing that secretion and cleavage of IL-1β were abolished in NLRP3, caspase-1/11, and ASC-deficient neutrophils

Read more

Summary

Introduction

Helicobacter pylori (H. pylori) is a gram-negative, microaerophilic, and spiral bacterium that colonizes the human gastric mucosa. H. pylori-Induced IL-1β Production in Neutrophils gastric mucosa-associated lymphoid tissue (MALT) lymphoma [1]. For this reason, it was classified by the World Health Organization as a class I carcinogen in 1994 [2]. Increased expression of IL8, IL-1β, and COX-2 genes was observed in patients with chronic gastritis infected with H. pylori compared with H. pylori negative patients [10]. These findings suggest that IL-1β may play a crucial role in the development of H. pylori-induced gastric inflammation and cancer

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