4.7 Article

Free-fatty acid receptor-4 (FFA4) modulates ROS generation and COX-2 expression via the C-terminal β-arrestin phosphosensor in Raw 264.7 macrophages

期刊

BIOCHEMICAL PHARMACOLOGY
卷 146, 期 -, 页码 139-150

出版社

PERGAMON-ELSEVIER SCIENCE LTD
DOI: 10.1016/j.bcp.2017.09.008

关键词

FFA4; GPR120; Free-fatty acids; Reactive oxygen species; COX-2

资金

  1. NIH [DK098730]
  2. Diabetes Action Research and Educational Foundation

向作者/读者索取更多资源

Agonism of the G protein-coupled free-fatty acid receptor-4 (FFA4) has been shown to promote numerous anti-inflammatory effects in macrophages that arise due to interaction with beta-arrestin partner proteins. Humans express functionally distinct short and long FFA4 splice variants, such that FFA4-S signals through G alpha q/11 and beta-arrestin, while FFA4-L is intrinsically biased solely towards beta-arrestin signaling. Recently, we and others have shown that phosphorylation of the FFA4 C-terminal tail is responsible for beta-arrestin interactability and signaling. Given the significance of beta-arrestin in the anti-inflammatory function of FFA4, the goal of this study was to examine the role of the C-terminal beta-arrestin phosphosensor in FFA4 signaling induced by PMA and LPS in murine Raw 264.7 macrophages. Our data reveal for the first time that both FFA4 isoforms modulate PMA-induced ROS generation, and that abolishment of the FFA4-S, but not FFA4-L C-terminal phosphosensor, is detrimental to this effect. Furthermore, we show that while both isoforms reduce PMA-induced expression of COX-2, removal of the FFA4-S phosphosensor significantly decreases this response, suggesting that these effects of FFA4-S are beta-arrestin mediated. On the contrary, FFA4-S, as well as the truncated C-terminal congener lacking the beta-arrestin phosphosensor were both able to reduce LPS-induced NF-kappa B activity and ERK1/2 phosphorylation. However, FFA4-L and its corresponding mutant were incapable of modulating either, suggesting that these responses are mediated by G protein coupling. Taken together, our data reveal important structure-function and signaling differences between the two FFA4 isoforms, and for the first time link FFA4 to modulation of ROS in macrophages. (C) 2017 Elsevier Inc. All rights reserved.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据