NLRP3 inflammasome activation in hepatocytes results in pyroptotic cell death, release of NLRP3 particles and liver fibrosis

2018 ◽  
Vol 68 ◽  
pp. S31-S32
Author(s):  
S. Schuster ◽  
F.A. Guerra ◽  
C.D. Johnson ◽  
C.J. Calvente ◽  
D. Povero ◽  
...  
2021 ◽  
Vol 22 (22) ◽  
pp. 12413
Author(s):  
Shuang Ge ◽  
Wei Yang ◽  
Haiqiang Chen ◽  
Qi Yuan ◽  
Shi Liu ◽  
...  

Chronic liver disease mediated by the activation of hepatic stellate cells (HSCs) leads to liver fibrosis. The signal adaptor MyD88 of Toll-like receptor (TLR) signaling is involved during the progression of liver fibrosis. However, the specific role of MyD88 in myeloid cells in liver fibrosis has not been thoroughly investigated. In this study, we used a carbon tetrachloride (CCl4)-induced mouse fibrosis model in which MyD88 was selectively depleted in myeloid cells. MyD88 deficiency in myeloid cells attenuated liver fibrosis in mice and decreased inflammatory cell infiltration. Furthermore, deficiency of MyD88 in macrophages inhibits the secretion of CXC motif chemokine 2 (CXCL2), which restrains the activation of HSCs characterized by NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome activation. Moreover, targeting CXCL2 by CXCR2 inhibitors attenuated the activation of HSCs and reduced liver fibrosis. Thus, MyD88 may represent a potential candidate target for the prevention and treatment of liver fibrosis.


2020 ◽  
Vol 204 (9) ◽  
pp. 2514-2522 ◽  
Author(s):  
Rajendra Karki ◽  
Ein Lee ◽  
Bhesh R. Sharma ◽  
Balaji Banoth ◽  
Thirumala-Devi Kanneganti

2020 ◽  
Vol 217 (7) ◽  
Author(s):  
Sannula Kesavardhana ◽  
Thirumala-Devi Kanneganti

ZBP1 triggers NLRP3 inflammasome activation/pyroptosis, apoptosis, and necroptosis; the specific ligand for ZBP1 activation remains ambiguous. Recent studies, including Devos et al. in this issue of JEM (https://doi.org/10.1084/jem.20191913), collectively suggest that ZBP1 sensing Z-nucleic acids is critical for cell death/inflammatory disease.


2016 ◽  
Vol 25 (8) ◽  
pp. 1501-1516 ◽  
Author(s):  
Ishaq A. Viringipurampeer ◽  
Andrew L. Metcalfe ◽  
Abu E. Bashar ◽  
Olena Sivak ◽  
Anat Yanai ◽  
...  

2017 ◽  
Vol 8 (8) ◽  
pp. e2984-e2984 ◽  
Author(s):  
Fátima Martín-Sánchez ◽  
Juan José Martínez-García ◽  
María Muñoz-García ◽  
Miriam Martínez-Villanueva ◽  
José A Noguera-Velasco ◽  
...  

2017 ◽  
Vol 2017 ◽  
pp. 1-17 ◽  
Author(s):  
Zhen Qiu ◽  
Shaoqing Lei ◽  
Bo Zhao ◽  
Yang Wu ◽  
Wating Su ◽  
...  

The reactive oxygen species- (ROS-) induced nod-like receptor protein-3 (NLRP3) inflammasome triggers sterile inflammatory responses and pyroptosis, which is a proinflammatory form of programmed cell death initiated by the activation of inflammatory caspases. NLRP3 inflammasome activation plays an important role in myocardial ischemia/reperfusion (MI/R) injury. Our present study investigated whether diabetes aggravated MI/R injury through NLRP3 inflammasome-mediated pyroptosis. Type 1 diabetic rat model was established by intraperitoneal injection of streptozotocin (60 mg/kg). MI/R was induced by ligating the left anterior descending artery (LAD) for 30 minutes followed by 2 h reperfusion. H9C2 cardiomyocytes were exposed to high glucose (HG, 30 mM) conditions and hypoxia/reoxygenation (H/R) stimulation. The myocardial infarct size, CK-MB, and LDH release in the diabetic rats subjected to MI/R were significantly higher than those in the nondiabetic rats, accompanied with increased NLRP3 inflammasome activation and increased pyroptosis. Inhibition of inflammasome activation with BAY11-7082 significantly decreased the MI/R injury.In vitrostudies showed similar effects, as BAY11-7082 or the ROS scavenger N-acetylcysteine, attenuated HG and H/R-induced H9C2 cell injury. In conclusion, hyperglycaemia-induced NLRP3 inflammasome activation may be a ROS-dependent process in pyroptotic cell death, and NLRP3 inflammasome-induced pyroptosis aggravates MI/R injury in diabetic rats.


Sign in / Sign up

Export Citation Format

Share Document