TY - JOUR T1 - Targeting clinical epigenetic reprogramming for chemoprevention of metabolic and viral hepatocellular carcinoma JF - Gut JO - Gut SP - 157 LP - 169 DO - 10.1136/gutjnl-2019-318918 VL - 70 IS - 1 AU - Frank Jühling AU - Nourdine Hamdane AU - Emilie Crouchet AU - Shen Li AU - Houssein El Saghire AU - Atish Mukherji AU - Naoto Fujiwara AU - Marine A Oudot AU - Christine Thumann AU - Antonio Saviano AU - Armando Andres Roca Suarez AU - Kaku Goto AU - Ricard Masia AU - Mozhdeh Sojoodi AU - Gunisha Arora AU - Hiroshi Aikata AU - Atsushi Ono AU - Parissa Tabrizian AU - Myron Schwartz AU - Stephen J Polyak AU - Irwin Davidson AU - Christian Schmidl AU - Christoph Bock AU - Catherine Schuster AU - Kazuaki Chayama AU - Patrick Pessaux AU - Kenneth K Tanabe AU - Yujin Hoshida AU - Mirjam B Zeisel AU - François HT Duong AU - Bryan C Fuchs AU - Thomas F Baumert Y1 - 2021/01/01 UR - http://gut.bmj.com/content/70/1/157.abstract N2 - Objective Hepatocellular carcinoma (HCC) is the fastest-growing cause of cancer-related mortality with chronic viral hepatitis and non-alcoholic steatohepatitis (NASH) as major aetiologies. Treatment options for HCC are unsatisfactory and chemopreventive approaches are absent. Chronic hepatitis C (CHC) results in epigenetic alterations driving HCC risk and persisting following cure. Here, we aimed to investigate epigenetic modifications as targets for liver cancer chemoprevention.Design Liver tissues from patients with NASH and CHC were analysed by ChIP-Seq (H3K27ac) and RNA-Seq. The liver disease-specific epigenetic and transcriptional reprogramming in patients was modelled in a liver cell culture system. Perturbation studies combined with a targeted small molecule screen followed by in vivo and ex vivo validation were used to identify chromatin modifiers and readers for HCC chemoprevention.Results In patients, CHC and NASH share similar epigenetic and transcriptomic modifications driving cancer risk. Using a cell-based system modelling epigenetic modifications in patients, we identified chromatin readers as targets to revert liver gene transcription driving clinical HCC risk. Proof-of-concept studies in a NASH-HCC mouse model showed that the pharmacological inhibition of chromatin reader bromodomain 4 inhibited liver disease progression and hepatocarcinogenesis by restoring transcriptional reprogramming of the genes that were epigenetically altered in patients.Conclusion Our results unravel the functional relevance of metabolic and virus-induced epigenetic alterations for pathogenesis of HCC development and identify chromatin readers as targets for chemoprevention in patients with chronic liver diseases. ER -