American Association for Cancer Research
Browse
00085472can202052-sup-245427_3_supp_7005600_qqfjsx.xls (46 kB)

Supplementary Table 2 from E2F1 and E2F2-Mediated Repression of CPT2 Establishes a Lipid-Rich Tumor-Promoting Environment

Download (46 kB)
dataset
posted on 2023-03-31, 04:44 authored by Francisco González-Romero, Daniela Mestre, Igor Aurrekoetxea, Colm J. O'Rourke, Jesper B. Andersen, Ashwin Woodhoo, Miguel Tamayo-Caro, Marta Varela-Rey, Marta Palomo-Irigoyen, Beatriz Gómez-Santos, Diego Sáenz de Urturi, Maitane Núñez-García, Juan L. García-Rodríguez, Larraitz Fernández-Ares, Xabier Buqué, Ainhoa Iglesias-Ara, Irantzu Bernales, Virginia Gutierrez De Juan, Teresa C. Delgado, Naroa Goikoetxea-Usandizaga, Richard Lee, Sanjay Bhanot, Igotz Delgado, Maria J. Perugorria, Gaizka Errazti, Lorena Mosteiro, Sonia Gaztambide, Idoia Martinez de la Piscina, Paula Iruzubieta, Javier Crespo, Jesus M. Banales, Maria L. Martínez-Chantar, Luis Castaño, Ana M. Zubiaga, Patricia Aspichueta

Supplementary table 2

Funding

Ayudas para apoyar grupos de investigación del sistema Universitario Vasco

MCIU

AEI

FEDER

MINECO

Horizon 2020

EITB

La Caixa Fundation

Spanish Carlos III Health Institute

Miguel Servet Program

AMMF-The Cholangiocarcinoma Charity, Euskadi

Severo Ochoa Excellence Accreditation

History

ARTICLE ABSTRACT

Lipid metabolism rearrangements in nonalcoholic fatty liver disease (NAFLD) contribute to disease progression. NAFLD has emerged as a major risk for hepatocellular carcinoma (HCC), where metabolic reprogramming is a hallmark. Identification of metabolic drivers might reveal therapeutic targets to improve HCC treatment. Here, we investigated the contribution of transcription factors E2F1 and E2F2 to NAFLD-related HCC and their involvement in metabolic rewiring during disease progression. In mice receiving a high-fat diet (HFD) and diethylnitrosamine (DEN) administration, E2f1 and E2f2 expressions were increased in NAFLD-related HCC. In human NAFLD, E2F1 and E2F2 levels were also increased and positively correlated. E2f1−/− and E2f2−/− mice were resistant to DEN–HFD-induced hepatocarcinogenesis and associated lipid accumulation. Administration of DEN–HFD in E2f1−/− and E2f2−/− mice enhanced fatty acid oxidation (FAO) and increased expression of Cpt2, an enzyme essential for FAO, whose downregulation is linked to NAFLD-related hepatocarcinogenesis. These results were recapitulated following E2f2 knockdown in liver, and overexpression of E2f2 elicited opposing effects. E2F2 binding to the Cpt2 promoter was enhanced in DEN–HFD-administered mouse livers compared with controls, implying a direct role for E2F2 in transcriptional repression. In human HCC, E2F1 and E2F2 expressions inversely correlated with CPT2 expression. Collectively, these results indicate that activation of the E2F1–E2F2–CPT2 axis provides a lipid-rich environment required for hepatocarcinogenesis. These findings identify E2F1 and E2F2 transcription factors as metabolic drivers of hepatocellular carcinoma, where deletion of just one is sufficient to prevent disease.