Abstract

Acetaminophen (APAP) toxicity is the leading cause of drug-induced liver failure, which is closely related to mitochondrial dysfunction and oxidative damage. Studies in clinical trials and in animal models have shown that omega-3 polyunsaturated fatty acids (n-3 PUFAs) affect the progression of various types of liver damage. Interestingly, the sex-dependent effect of n-3 PUFAs on human health has also been well documented. However, it is unknown whether supplementation of n-3 PUFAs modulates the pathogenesis of APAP-induced liver failure with sex-specificity. Our results showed that both endogenous and exogenous n-3 PUFAs significantly aggravated the APAP-induced liver injury in male mice, whereas the opposite effects were observed in females. In vivo and in vitro studies demonstrated that estrogen contributes to the gender difference in the regulation of n-3 PUFAs on APAP overdose. We found that n-3 PUFA-mediated regulation of hepatic oxidative stress response and autophagy upon APAP challenge is distinct between male and female mice. Moreover, we provided evidence that β-catenin signaling activation is responsible for the sex-dependent regulation of APAP hepatotoxicity by n-3 PUFAs. Together, these findings indicated that supplementation with n-3 PUFAs displays sex-differential effect on APAP hepatotoxicity and could have profound significance in the clinical management for drug-induced liver injury.

Highlights

  • Acetaminophen (APAP) is currently one of the most widely used antipyretic and analgesic drugs

  • We have demonstrated that n-3 PUFAs exert sexspecific control of APAP hepatotoxicity in mice

  • Our data showed that n-3 PUFAs aggravated APAP-induced liver injury in male mice but ameliorated APAP hepatotoxicity in female mice through differential regulation of autophagy activation

Read more

Summary

Introduction

Acetaminophen (APAP) is currently one of the most widely used antipyretic and analgesic drugs. This drug has been considered safe for decades, an overdose can cause severe liver damage, which may cause acute liver failure [1]. It was reported that both inhibitors of ROS or JNK protected mice from APAP-induced liver injury [4, 5]. Autophagy is a cellular process that can degrade impaired mitochondria, thereby removing accumulated ROS [6]. It has been confirmed that increased autophagy protects mice against APAP-induced liver failure [7, 8]

Objectives
Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.