Hepatocyte-specific high-mobility group box 1 deletion worsens the injury in liver ischemia/reperfusion: a role for intracellular high-mobility group box 1 in cellular protection.

 Back to publications

2013

Hepatology. 2013 Dec 21. doi: 10.1002/hep.26976. [Epub ahead of print]

Hepatocyte-specific high-mobility group box 1 deletion worsens the injury in liver ischemia/reperfusion: a role for intracellular high-mobility group box 1 in cellular protection.

Huang H;Nace GW;McDonald KA;Tai S;Klune JR;Rosborough BR;Ding Q;Loughran P;Zhu X;Beer-Stolz;Chang EB;Billiar T;Tsung A

Department Of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA. Center for Biologic Imaging, Department of Cell Biology, University of Pittsburgh Medical Center, Pittsburgh, PA. Department of Medicine, University of Chicago, Chicago, IL.

Service type: Knockout mice

Abstract

High mobility group box-1 (HMGB1) is an abundant chromatin associated nuclear protein and released into the extracellular milieu during liver ischemia/reperfusion (I/R), signaling the activation of pro-inflammatory cascades. Since the intracellular function of HMGB1 during the sterile inflammation of I/R is currently unknown, we sought to determine the role of intracellular HMGB1 in hepatocytes following liver I/R. When hepatocyte specific HMGB1 knockout (HMGB1-HC-KO) and control mice were subjected to a non-lethal warm liver I/R, it was found that HMGB1-HC-KO mice had significantly greater hepatocellular injury after I/R compared to control mice. Additionally, there was significantly greater DNA damage and decreased chromatin accessibility to repair with lack of HMGB1. Furthermore, lack of hepatocyte HMGB1 led to excessive poly (ADP-ribose) polymerase-1 (PARP-1) activation, exhausting NAD(+) and ATP stores, exacerbating mitochondrial instability and damage, and consequently leading to increased cell death. We found that this was also associated with significantly more oxidative stress in HMGB1-HC-KO mice compared to control. Increased nuclear instability led to a resultant increase in the release of histones with subsequently more inflammatory cytokine production and organ damage through the activation of TLR9. Conclusion: Therefore, the lack of HMGB1 within hepatocytes leads to the increased susceptibility to cellular death after oxidative stress conditions. (Hepatology 2013;).

View Publication