4.8 Article

The myeloid heat shock transcription factor 1/-catenin axis regulates NLR family, pyrin domain-containing 3 inflammasome activation in mouse liver ischemia/reperfusion injury

期刊

HEPATOLOGY
卷 64, 期 5, 页码 1683-1698

出版社

WILEY
DOI: 10.1002/hep.28739

关键词

-

资金

  1. NIH [R21AI112722, R01DK102110]
  2. National Natural Science Foundation of China [81270558, 81470895]
  3. Dumont Research Foundation

向作者/读者索取更多资源

Heat shock transcription factor 1 (HSF1) has been implicated in the differential regulation of cell stress and disease states. -catenin activation is essential for immune homeostasis. However, little is known about the role of macrophage HSF1--catenin signaling in the regulation of NLRP3 inflammasome activation during ischemia/reperfusion (I/R) injury (IRI) in the liver. This study investigated the functions and molecular mechanisms by which HSF1--catenin signaling influenced NLRP3-mediated innate immune response in vivo and in vitro. Using a mouse model of IR-induced liver inflammatory injury, we found that mice with a myeloid-specific HSF1 knockout (HSF1(M-KO)) displayed exacerbated liver damage based on their increased serum alanine aminotransferase levels, intrahepatic macrophage/neutrophil trafficking, and proinflammatory interleukin (IL)-1 levels compared to the HSF1-proficient (HSF1(FL/FL)) controls. Disruption of myeloid HSF1 markedly increased transcription factor X-box-binding protein (XBP1), NLR family, pyrin domain-containing 3 (NLRP3), and cleaved caspase-1 expression, which was accompanied by reduced -catenin activity. Knockdown of XBP1 in HSF1-deficient livers using a XBP1 small interfering RNA ameliorated hepatocellular functions and reduced NLRP3/cleaved caspase-1 and IL-1 protein levels. In parallel in vitro studies, HSF1 overexpression increased -catenin (Ser552) phosphorylation and decreased reactive oxygen species (ROS) production in bone-marrow-derived macrophages. However, myeloid HSF1 ablation inhibited -catenin, but promoted XBP1. Furthermore, myeloid -catenin deletion increased XBP1 messenger RNA splicing, whereas a CRISPR/CRISPR-associated protein 9-mediated XBP1 knockout diminished NLRP3/caspase-1. Conclusion: The myeloid HSF1--catenin axis controlled NLRP3 activation by modulating the XBP1 signaling pathway. HSF1 activation promoted -catenin, which, in turn, inhibited XBP1, leading to NLRP3 inactivation and reduced I/R-induced liver injury. These findings demonstrated that HSF1/-catenin signaling is a novel regulator of innate immunity in liver inflammatory injury and implied the therapeutic potential for management of sterile liver inflammation in transplant recipients. (Hepatology 2016;64:1683-1698).

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

4.8
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