Abstract

Considerable attention has been raised on crizotinib- and sunitinib-induced hepatotoxicity, but the underlying mechanisms need further examination. In addition, limited therapeutic strategies exist to reduce the liver damage caused by crizotinib and sunitinib. This study investigated the mechanisms of crizotinib- and sunitinib-induced hepatotoxicity and the potential mitigation through ROS and Nrf2 signaling. Firstly, crizotinib and sunitinib reduced cell viability in human liver cells (L02 cells) and triggered dramatic liver injury in mice. Subsequently, we found that crizotinib and sunitinib activated the oxidative stress response (decreased level of GPx and SOD, and increased MDA content) in vivo. Crizotinib and sunitinib also stimulated hepatocyte mitochondrial apoptosis and necrosis in L02 cells in a dose-dependent manner. In vivo studies further confirmed that crizotinib and sunitinib decreased mitochondrial membrane potential and activated apoptosis-associated proteins (cleaved-PARP, cleaved caspase3, cytochrome c, Bcl2 and Bax). Furthermore, mechanistic investigations demonstrated that crizotinib and sunitinib accumulated ROS and inhibited Nrf2 signaling, and that ROS scavenger NAC and Nrf2 agonist tBHQ alleviated the extent of cell damage and the mitochondrial apoptosis during crizotinib- and sunitinib-induced hepatotoxicity in L02 cells. Collectively, these findings indicated that NAC and tBHQ play the crucial roles in crizotinib- and sunitinib-induced mitochondrial apoptosis via the regulation of oxidative stress.

Highlights

  • Most cancers therapies were previously dominated by chemotherapy

  • The results showed that crizotinib group and sunitinib group both remarkably increased cPARP, c-caspase3 and cytoplasm cytc, and decreased ratio of Bcl2/ Bax that might contribute to the loss of Membrane Potential (MMP) (Figures 3C,D)

  • We explored whether tertiary butylhydroquinone (tBHQ), an antioxidant and the Nrf2 agonist, alleviated crizotinib- and sunitinib-induced hepatotoxicity and mitochondrial apoptosis effectively

Read more

Summary

INTRODUCTION

Most cancers therapies were previously dominated by chemotherapy. with the advent of the first small-molecule targeted drug imatinib, targeted drugs rapidly developed into the mainstream of anti-tumor drug research with the advantages of significant efficacy, higher safety, less damage to normal cells and improvement of survival and life quality for patients. It has been found that TKIs-triggered hepatotoxicity was associated with drug exposure, toxic metabolites, liver transporters, drug metabolism enzyme polymorphism and immune response (Takimoto et al, 2013; Kobayashi et al, 2015; Teo et al, 2015; Ma et al, 2017; Amaya et al, 2018), and closely related to oxidative stress (Xue et al, 2012), mitochondrial damage and apoptosis (Eno et al, 2016; Chen et al, 2018). We used the L02 cell as an in vitro model and C57 mice as an in vivo model to examine whether oxidative stress and mitochondrial apoptosis was relately to crizotinib- and sunitinib-triggered hepatotoxicity. We evaluated the potential protective effects of pretreatment with ROS scavenger n-acetyl-L-cysteine (NAC) and Nrf agonist tertiary butylhydroquinone (tBHQ) on the cell viability and mitochondrial apoptotic pathways in L02 cell elicited by crizotinib and sunitinib

MATERIALS AND METHODS
RESULT
DISCUSSION
CONCLUSION
Findings
ETHICS STATEMENT
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call