This application addresses NCI PQ6 on how circadian processes affect tumor development by studying the role of circadian dysfunction of nuclear receptor pathways in non-alcoholic fatty liver disease (NAFLD)- induced hepatocellular carcinoma (HCC). HCC, previously considered a rare cancer in the Western world, has increased 3-fold in incidence since the 1980s, and is currently the fastest rising cause of cancer-related death in the U.S, due to an increase in incidence in combination of lack of understanding of its mechanism. The increased HCC risk is coupled with the prevalence of obesity-related NAFLD, which has recently become the leading risk factor for HCC. However, no efficient approaches for prevention, early diagnosis, and treatment of NAFLD-induced HCCs are currently available. The increased risk of NAFLD-related HCC is coupled with population-wide chronic circadian disruption, a common risk factor of obesity, NAFLD, and cancer in humans. We have recently established a jet-lagged mouse model following a human nightshift schedule. We found that chronic jet-lag induces metabolic syndrome, NAFLD, and HCC in wild-type mice following a pathophysiological pathway strikingly similar to that observed in obese humans, with NAFLD progressing to nonalcoholic steatohepatitis (NASH) and fibrosis prior to HCC detection. We identified intrahepatic cholestasis as the key pathophysiological mechanism promoting NAFLD-induced HCC, and demonstrated that circadian dysregulation of nuclear receptor FXR and CAR pathways is an essential oncogenic mechanism that drives cholestasis and toxic bile acid signaling to promote the progression from NAFLD to NASH, fibrosis, and eventually HCC. In this application, we propose 3 aims. 1) Define the circadian profiles of FXR and CAR controlled hepatic gene networks and jet-lag induced deregulation signatures of these gene networks. 2) Define the role of Ctnnb1 and Tp53 and other oncogenic mutations in the progression from jet-lag induced liver premalignant lesions to HCC and the circadian profiles of gene signatures as well as serum biomarkers associated with hepatocarcinogenesis. 3) Test the predicted abilities of FXR agonist obeticholic acid (OCA) and CAR inverse agonist androstanol (ANDR) to prevent jet-lag induced cholestasis and HCC. These studies will significantly improve our understanding of the role of circadian dysfunction in spontaneous carcinogenesis in general, and NFALD-induced hepatocarcinogenesis in particular. They will also illuminate new and exciting avenues to develop novel chronotherapy strategies for prevention and treatment of NAFLD-induced HCC in humans.

Public Health Relevance

This project addresses fundamental mechanisms that connect disruption of circadian rhythm to the lethal liver cancer hepatocellular carcinoma (HCC). It will address the roles of circadian dysregulation of the nuclear xenobiotic receptor CAR and the bile acid receptor FXR in the induction of HCC, and will also test the predicted abilities of drugs targeting these receptors to prevent circadian disruption induced liver pathologies and HCC. The results of these studies will have outstanding potential for near-term therapeutic options in HCC prevention and treatment in humans.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA230848-02
Application #
9787413
Study Section
Special Emphasis Panel (ZCA1)
Program Officer
Watson, Joanna M
Project Start
2018-09-18
Project End
2023-08-31
Budget Start
2019-09-01
Budget End
2020-08-31
Support Year
2
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Baylor College of Medicine
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
051113330
City
Houston
State
TX
Country
United States
Zip Code
77030