scholarly journals Helicobacter pylori-Induced Th17 Responses Modulate Th1 Cell Responses, Benefit Bacterial Growth, and Contribute to Pathology in Mice

2010 ◽  
Vol 184 (9) ◽  
pp. 5121-5129 ◽  
Author(s):  
Yun Shi ◽  
Xiao-Fei Liu ◽  
Yuan Zhuang ◽  
Jin-Yu Zhang ◽  
Tao Liu ◽  
...  
2018 ◽  
Vol 9 (5) ◽  
Author(s):  
Yi-pin Lv ◽  
Yong-sheng Teng ◽  
Fang-yuan Mao ◽  
Liu-sheng Peng ◽  
Jin-yu Zhang ◽  
...  

1999 ◽  
Vol 21 (4) ◽  
pp. 803-846 ◽  
Author(s):  
A. Di Leo ◽  
C. Messa ◽  
F. Russo ◽  
M. Linsalata ◽  
L. Amati ◽  
...  

mBio ◽  
2019 ◽  
Vol 10 (2) ◽  
Author(s):  
Aleksandra Altobelli ◽  
Michael Bauer ◽  
Karelia Velez ◽  
Timothy L. Cover ◽  
Anne Müller

ABSTRACT The gastric bacterium Helicobacter pylori causes a persistent infection that is directly responsible for gastric ulcers and gastric cancer in some patients and protective against allergic and other immunological disorders in others. The two outcomes of the Helicobacter-host interaction can be modeled in mice that are infected as immunocompetent adults and as neonates, respectively. Here, we have investigated the contribution of the Helicobacter immunomodulator VacA to H. pylori-specific local and systemic immune responses in both models. We found that neonatally infected mice are colonized at higher levels than mice infected as adults and fail to generate effector T-cell responses to the bacteria; rather, T-cell responses in neonatally infected mice are skewed toward Foxp3-positive (Foxp3+) regulatory T cells that are neuropilin negative and express RORγt. We found these peripherally induced regulatory T cells (pTregs) to be enriched, in a VacA-dependent manner, not only in the gastric mucosa but also in the lungs of infected mice. Pulmonary pTreg accumulation was observed in mice that have been infected neonatally with wild-type H. pylori but not in mice that have been infected as adults or mice infected with a VacA null mutant. Finally, we traced VacA to gastric lamina propria myeloid cells and show that it suppressed interleukin-23 (IL-23) expression by dendritic cells and induced IL-10 and TGF-β expression in macrophages. Taken together, the results are consistent with the idea that H. pylori creates a tolerogenic environment through its immunomodulator VacA, which skews T-cell responses toward Tregs, favors H. pylori persistence, and affects immunity at distant sites. IMPORTANCE Helicobacter pylori has coexisted with humans for at least 60.000 years and has evolved persistence strategies that allow it to evade host immunity and colonize its host for life. The VacA protein is expressed by all H. pylori strains and is required for high-level persistent infection in experimental mouse models. Here, we show that VacA targets myeloid cells in the gastric mucosa to create a tolerogenic environment that facilitates regulatory T-cell differentiation, while suppressing effector T-cell priming and functionality. Tregs that are induced in the periphery during H. pylori infection can be found not only in the stomach but also in the lungs of infected mice, where they are likely to affect immune responses to allergens.


2011 ◽  
Vol 42 (2) ◽  
pp. 364-373 ◽  
Author(s):  
Radha Gopal ◽  
Yinyao Lin ◽  
Nataša Obermajer ◽  
Samantha Slight ◽  
Nikhil Nuthalapati ◽  
...  
Keyword(s):  
Th1 Cell ◽  

2021 ◽  
Vol 160 (6) ◽  
pp. S-109
Author(s):  
Stella G. Hoft ◽  
Jose Saenz ◽  
Christine Noto ◽  
Kevin Bockerstett ◽  
Nicholas M. Jackson ◽  
...  

2003 ◽  
Vol 202 (2) ◽  
pp. 197-207 ◽  
Author(s):  
Jean E Crabtree ◽  
Michelle Court ◽  
Mohamed A Aboshkiwa ◽  
Anthony HT Jeremy ◽  
Michael F Dixon ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document