Arginase 2 deletion leads to enhanced M1 macrophage activation and upregulated polyamine metabolism in response to Helicobacter pylori infection

We reported that arginase 2 (ARG2) deletion results in increased gastritis and decreased bacterial burden during Helicobacter pylori infection in mice. Our studies implicated a potential role for inducible nitric oxide (NO) synthase (NOS2), as Arg2 − / − mice exhibited increased NOS2 levels in gastr...

Full description

Saved in:
Bibliographic Details
Published inAmino acids Vol. 48; no. 10; pp. 2375 - 2388
Main Authors Hardbower, Dana M., Asim, Mohammad, Murray-Stewart, Tracy, Casero, Robert A., Verriere, Thomas, Lewis, Nuruddeen D., Chaturvedi, Rupesh, Piazuelo, M. Blanca, Wilson, Keith T.
Format Journal Article
LanguageEnglish
Published Vienna Springer Vienna 01.10.2016
Springer Nature B.V
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:We reported that arginase 2 (ARG2) deletion results in increased gastritis and decreased bacterial burden during Helicobacter pylori infection in mice. Our studies implicated a potential role for inducible nitric oxide (NO) synthase (NOS2), as Arg2 − / − mice exhibited increased NOS2 levels in gastric macrophages, and NO can kill H. pylori . We now bred Arg2 − / − to Nos2 − / − mice, and infected them with H. pylori . Compared to wild-type mice, both Arg2 − / − and Arg2 − / − ;Nos2 − / − mice exhibited increased gastritis and decreased colonization, the latter indicating that the effect of ARG2 deletion on bacterial burden was not mediated by NO. While Arg2 − / − mice demonstrated enhanced M1 macrophage activation, Nos2 − / − and Arg2 − / − ;Nos2 − / − mice did not demonstrate these changes, but exhibited increased CXCL1 and CXCL2 responses. There was an increased expression of the Th1/Th17 cytokines, interferon gamma and interleukin 17, in gastric tissues and splenic T-cells from Arg2 − / − , but not Nos2 − / − or Arg2 − / − ;Nos2 − / − mice. Gastric tissues from infected Arg2 − / − mice demonstrated increased expression of arginase 1, ornithine decarboxylase, adenosylmethionine decarboxylase 1, spermidine/spermine N 1 -acetyltransferase 1, and spermine oxidase, along with increased spermine levels. These data indicate that ARG2 deletion results in compensatory upregulation of gastric polyamine synthesis and catabolism during H. pylori infection, which may contribute to increased gastric inflammation and associated decreased bacterial load. Overall, the finding of this study is that ARG2 contributes to the immune evasion of H. pylori by restricting M1 macrophage activation and polyamine metabolism.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:0939-4451
1438-2199
DOI:10.1007/s00726-016-2231-2