scholarly journals Increase toll‐like receptor 4 expression after ischemia/reperfusion contributes to myocardial apoptosis: role of PI3Kγ/NFκB pathway

2010 ◽  
Vol 24 (S1) ◽  
Author(s):  
Hu Xu ◽  
Xuemei Xu ◽  
Jingchao Zhang ◽  
Zhaoliang Su ◽  
Claudio M. Martin ◽  
...  
2005 ◽  
Vol 201 (3) ◽  
pp. S101
Author(s):  
Hassan Albadawi ◽  
Fateh Entabi ◽  
Mark Conrad ◽  
David Stone ◽  
Michael Watkins

Hepatology ◽  
2013 ◽  
Vol 58 (1) ◽  
pp. 374-387 ◽  
Author(s):  
Gary W. Nace ◽  
Hai Huang ◽  
John R. Klune ◽  
Raymond E. Eid ◽  
Brian R. Rosborough ◽  
...  

2008 ◽  
Vol 144 (2) ◽  
pp. 206
Author(s):  
David J. Kaczorowski ◽  
Atsu Nakao ◽  
Raghuveer Vallabhaneni ◽  
Brian S. Zuckerbraun ◽  
Kenneth R. McCurry ◽  
...  

2015 ◽  
Vol 99 (4) ◽  
pp. 1193-1199 ◽  
Author(s):  
Heather E. Merry ◽  
Patrick Phelan ◽  
Mathew R. Doak ◽  
Minqing Zhao ◽  
Billanna Hwang ◽  
...  

2011 ◽  
Vol 300 (3) ◽  
pp. H913-H921 ◽  
Author(s):  
Hu Xu ◽  
Yongwei Yao ◽  
Zhaoliang Su ◽  
Yunbo Yang ◽  
Raymond Kao ◽  
...  

High-mobility group box 1 (HMGB1) is a nuclear protein that has been implicated in the myocardial inflammation and injury induced by ischemia-reperfusion (I/R). The purpose of the present study was to assess the role of HMGB1 in myocardial apoptosis induced by I/R. In vivo, myocardial I/R induced an increase in myocardial HMGB1 expression and apoptosis. Inhibition of HMGB1 (A-box) ameliorated the I/R-induced myocardial apoptosis. In vitro, isolated cardiac myocytes were challenged with anoxia-reoxygenation (A/R; in vitro correlate to I/R). A/R-challenged myocytes also generated HMGB1 and underwent apoptosis. Inhibition of HMGB1 attenuated the A/R-induced myocyte apoptosis. Exogenous HMGB1 had no effect on myocyte apoptosis. However, inhibition of HMGB1 attenuated myocyte TNF-α production after the A/R was challenged; surprisingly, HMGB1 itself did not induce myocyte TNF-α production. Exogenous TNF-α induced a moderate proapoptotic effect on the myocytes, an effect substantially potentiated by coadministration of HMGB1. It is generally accepted that apoptosis induced by TNF-α is regulated by the balance of activation of c-Jun NH2-terminal kinase (JNK) and NF-κB. Indeed, in the present study, TNF-α increased the phosphorylation status of JNK and p65, a subunit of NF-κB; HMGB1 greatly potentiated TNF-α-induced JNK phosphorylation. Furthermore, inhibition of JNK (SP-600125) prevented the myocyte apoptosis induced by a TNF-α/HMGB1 cocktail. Finally, A/R increased HMGB1 production in both wild-type and toll-like receptor 4-deficient myocytes; however, deficiency in toll-like receptor 4 diminished A/R-induced myocyte apoptosis, TNF-α, and JNK activation. Our results indicate that myocyte-derived HMGB1 and TNF-α work in concert to promote I/R-induced myocardial apoptosis through JNK activation.


2014 ◽  
Vol 306 (8) ◽  
pp. F801-F811 ◽  
Author(s):  
Hailin Zhao ◽  
Jessica Santiváñez Perez ◽  
Kaizhi Lu ◽  
Andrew J. T. George ◽  
Daqing Ma

Toll-like receptor-4 (TLR-4) has been increasingly recognized as playing a critical role in the pathogenesis of ischemia-reperfusion injury (IRI) of renal grafts. This review provides a detailed overview of the new understanding of the involvement of TLR-4 in ischemia-reperfusion injury of renal grafts and its clinical significance in renal transplantation. TLR-4 not only responds to exogenous microbial motifs but can also recognize molecules which are released by stressed and necrotic cells, as well as degraded products of endogenous macromolecules. Upregulation of TLR-4 is found in tubular epithelial cells, vascular endothelial cells, and infiltrating leukocytes during renal ischemia-reperfusion injury, which is induced by massive release of endogenous damage-associated molecular pattern molecules such as high-mobility group box chromosomal protein 1. Activation of TLR-4 promotes the release of proinflammatory mediators, facilitates leukocyte migration and infiltration, activates the innate and adaptive immune system, and potentiates renal fibrosis. TLR-4 inhibition serves as the target of pharmacological agents, which could attenuate ischemia-reperfusion injury and associated delayed graft function and allograft rejection. There is evidence in the literature showing that targeting TLR-4 could improve long-term transplantation outcomes. Given the pivotal role of TLR-4 in ischemia-reperfusion injury and associated delayed graft function and allograft rejection, inhibition of TLR-4 using pharmacological agents could be beneficial for long-term graft survival.


2013 ◽  
Vol 57 (5) ◽  
pp. 77S
Author(s):  
Ali Navi ◽  
Rebekah Yu ◽  
Xu Shi-Wen ◽  
Sidney Shaw ◽  
George Hamilton ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document