Main

Ischemia-reperfusion injury (I/R) is responsible for primary liver dysfunction and failure after transplantation, liver resection, or hemorrhagic shock.1 I/R injury is triggered when the liver is transiently deprived of oxygen and reoxygenated, and this type of injury can occur in several clinical settings, such as those associated with low-flow states, a diverse range of surgical procedures, or organ procurement for transplantation.2 Jaeschke et al.3 described and characterized two distinct phases of liver injury. The early phase of injury occurs during the initial few hours after reperfusion and involves the production of reactive oxygen species by Kupffer cells, which also produce pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-1 (IL-1), and eventually give rise to a later, inflammation-mediated, phase of injury.3 I/R injury is detrimental to liver graft function. Unfortunately, there is no treatment available to prevent hepatic I/R injury. A better understanding of the intrinsic mechanisms that mediate the intricate cell survival signaling pathway should allow the development of a novel therapeutic approach to minimize the adverse effects of I/R injury.

Nuclear factor-kappa B (NF-κB) and activating protein-1 (AP-1) are important transcription factors that control the expression of cytokines and factors involved in the regulation of I/R injury. Previous studies have indicated that NF-κB is activated during liver I/R injury and that the inhibition of its activation during I/R reduces hepatocyte necrosis in rats.4, 5, 6, 7 The inflammatory response is tightly regulated by pro-inflammatory mediators such as p38 mitogen-activated protein kinases (MAPKs) and c-Jun-N-terminal kinase (JNK). The activation of JNK is crucial to I/R injury and leads to caspase-3 activation and hepatic apoptosis.Full size image

To determine the interaction regions, deletion mutants of TRAF1 and ASK1 were constructed and analyzed for their ability to interact with each other (Figure 8c). HepG2 cells were co-transfected with full-length or truncated TRAF1 expression vectors with Myc tags and with Flag-tagged ASK1 expression vector. The cell lysates were immunoprecipitated with anti-Myc or an anti-Flag antibody and then analyzed by western blotting using anti-Flag or anti-Myc antibody, respectively. The results showed that residues 188–409 of TRAF1 were responsible for the interaction (Figures 8d and f). Similarly, we co-transfected HepG2 cells with a series of Flag-tagged ASK1 constructs and full-length Myc-tagged TRAF1 and performed IP, and we confirmed that only the construct consisting of ASK1 C terminus (residues 941–1375) lost the ability to bind to TRAF1 (Figures 8e and g), indicating that all of the other ASK1 domains contributed to this interaction.

To investigate the putative relationship between TRAF1 and ASK1, we investigated whether TRAF1 colocalizes with ASK1 in vitro. HepG2 cells were co-transfected with GFP-fusion proteins of full-length TRAF1 or the TRAF1 N terminus (residues 1–187) and a mCherry-fusion protein of ASK1. The subcellular localization of the proteins was visualized by confocal microscopy and image processing. Full-length TRAF1 and the N terminus of TRAF1were perfectly colocalized with ASK1 in the cytoplasm, although the TRAF1 N terminus was unable to interact with ASK1 in vitro. (Figure 8h). To further confirm the TRAF1–ASK1 interaction in vivo, we used fluorescence resonance energy transfer (FRET), in which the energy transfer between the donor and acceptor depends on the distance and dipole orientations of the two partners. We co-transfected 293T cells with vectors expressing CFP-fused ASK1 and YFP-fused full-length TRAF1 or TRAF1 N terminus (residues 1–187) as the donor–acceptor pair. To measure FRET in cells, we used the dequenching approach in which the mean fluorescence intensities of the donor (CFP) fluorophores were recorded before and after acceptor (YFP) photobleaching. The average FRET efficiency was calculated, and the intensities of ASK1-CFP fluorescence increased by 8.69% with coexpressed full-length TRAF1-YFP, but only by 1.64% with coexpressed truncated TRAF1. The detected FRET suggests that the C-terminal domain of TRAF1 is required for the association of the two proteins (Figures 8i and j).

The kinase activity of ASK1 is crucial for its function in hepatocytes

Owing to the direct interaction of TRAF1 with ASK1 and the observation that the levels of phosphorylated ASK1 were lower in TRAF1-deficient mice, we hypothesized that the inhibition of the physical binding between TRAF1 and ASK1 blocks the effect of TRAF1 on ischemic hepatocytes. We then constructed recombinant adenoviruses expressing mutant ASK1 that could not bind to TRAF1. Transfection of cultured hepatocytes with the vector carrying this mutant ASK1 did not increase the H/R-induced cell damage, as measured by an MTT assay and an LDH release assay (Figure 8k), indicating that the dissociation of ASK1 from TRAF1 eliminates the damage caused by excessive wild-type TRAF1.

We transfected TRAF1-deficient hepatocytes with AdASK1 and observed that the overexpression of ASK1 exacerbated the cell injury, abrogating the hepatoprotective effect of targeting TRAF1 (Figure 8l). Finally, we examined whether the activity of ASK1 is required for its effect on hepatocyte survival. Accordingly, hepatocytes isolated from TRAF1 TG or NTG mice were transfected with adenoviruses expressing dominant-negative ASK1 (Addn-ASK1). Surprisingly, higher MTT values and less LDH release were observed in the Addn-ASK group, indicating that H/R-induced cell injury was suppressed by dnASK1 (Figure 8m). The above data demonstrated that TRAF1–ASK1 signaling has an important role in I/R-associated hepatocyte injury.