Unknown

Dataset Information

0

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


ABSTRACT: 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 (HSF1M-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 (HSF1FL/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.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).

SUBMITTER: Yue S 

PROVIDER: S-EPMC5074868 | biostudies-literature | 2016 Nov

REPOSITORIES: biostudies-literature

altmetric image

Publications

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

Yue Shi S   Zhu Jianjun J   Zhang Ming M   Li Changyong C   Zhou Xingliang X   Zhou Min M   Ke Michael M   Busuttil Ronald W RW   Ying Qi-Long QL   Kupiec-Weglinski Jerzy W JW   Xia Qiang Q   Ke Bibo B  

Hepatology (Baltimore, Md.) 20160829 5


Heat shock transcription factor 1 (HSF1) has been implicated in the differential regulation of cell stress and disease states. β-catenin activation is essential for immune homeostasis. However, little is known about the role of macrophage HSF1-β-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-β-catenin signaling influenced NLRP3-mediated i  ...[more]

Similar Datasets

| S-EPMC6307946 | biostudies-literature
| S-EPMC8349068 | biostudies-literature
| S-EPMC6189699 | biostudies-other
| S-EPMC2692047 | biostudies-literature
| S-EPMC6116402 | biostudies-literature
| S-EPMC6351190 | biostudies-literature
| S-EPMC6829519 | biostudies-literature
| S-EPMC2928523 | biostudies-literature
| S-EPMC7142395 | biostudies-literature
| S-EPMC4528191 | biostudies-literature