Z Gastroenterol 2020; 58(01): e39
DOI: 10.1055/s-0039-3402204
Poster Visit Session IV Tumors: Saturday, February 15, 2020, 8:30 am – 09:15 am, Lecture Hall P1
Georg Thieme Verlag KG Stuttgart · New York

Hepatic activation of FOXO3 regulates mTORC2-Akt and enhances oxidative damage-associated hepatocellular carcinogenesis

M Lu
1   Klinikum rechts der Isar, Department of Surgery, Munich, Germany
2   Zhongda Hospital, Department of General Surgery, Nanjing, China
,
D Hartmann
1   Klinikum rechts der Isar, Department of Surgery, Munich, Germany
,
R Braren
3   Klinikum rechts der Isar, Institute for diagnostic and interventional Radiology, Munich, Germany
,
A Gupta
3   Klinikum rechts der Isar, Institute for diagnostic and interventional Radiology, Munich, Germany
,
B Wang
1   Klinikum rechts der Isar, Department of Surgery, Munich, Germany
,
Y Wang
1   Klinikum rechts der Isar, Department of Surgery, Munich, Germany
,
C Mogler
4   Klinikum rechts der Isar, Institute of Pathology, Munich, Germany
,
Z Cheng
2   Zhongda Hospital, Department of General Surgery, Nanjing, China
,
T Wirth
5   University of Ulm, Institute of Physiological Chemistry, Ulm, Germany
,
H Friess
1   Klinikum rechts der Isar, Department of Surgery, Munich, Germany
,
J Kleeff
6   University Medical Center Halle, Department of Visceral, Vascular and Endocrine Surgery, Halle (saale), Germany
,
N Hüser
1   Klinikum rechts der Isar, Department of Surgery, Munich, Germany
,
Y Sunami
6   University Medical Center Halle, Department of Visceral, Vascular and Endocrine Surgery, Halle (saale), Germany
› Author Affiliations
Further Information

Publication History

Publication Date:
03 January 2020 (online)

 
 

    Background:

    Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer, accounting for 80 – 90% of cases. Mutations are commonly found in the signaling regulating the PI3K/Akt pathway, leading to oncogenic cell proliferation and survival. Key transcription factors that are negatively regulated downstream of PI3K/Akt are members of the forkhead box O family (FOXO). FOXOs were initially considered as tumor suppressors by inducing cell cycle arrest and apoptosis. However, there is increasing evidence showing that FOXOs, especially FOXO3, can support tumorigenesis.

    Methods:

    To understand the roles of FOXO3 in liver tumorigenesis and hepatocarcinogenesis, we analyzed HCC patient specimens and also established a doxycycline-regulated transgenic mouse model with hepatocyte-specific FOXO3 expression in a constitutively active form.

    Results:

    We found that FOXO3 protein is significantly overexpressed and activated in livers of HCC patients. Hepatic activation of FOXO3 induced extensive hepatic damage and elevated gene expression of several HCC-associated factors. Furthermore, FOXO3 expression enhanced hepatotoxicin-induced tumorigenesis. Mechanistically, FOXO3 activation caused oxidative stress and DNA damage and triggered positive feedback-loop for Akt activation as well as mTORC2 activation. Interestingly, FOXO3 activated not only reactive oxygen species (ROS)-promoting pathways, but also ROS-eliminating systems, which can be associated with the activation of the pentose phosphate pathway.

    Conclusions:

    FOXO3 is a master regulator of ROS. On one side, FOXO3 supports in protecting from ROS and may avoid cellular crisis but FOXO3 can also promote ROS signaling on the other side and support hepatocellular carcinogenesis.


    #