Skip to main content
Top
Published in: BMC Cancer 1/2019

Open Access 01-12-2019 | Hepatocellular Carcinoma | Research article

Oncogenic Akt-FOXO3 loop favors tumor-promoting modes and enhances oxidative damage-associated hepatocellular carcinogenesis

Authors: Miao Lu, Daniel Hartmann, Rickmer Braren, Aayush Gupta, Baocai Wang, Yang Wang, Carolin Mogler, Zhangjun Cheng, Thomas Wirth, Helmut Friess, Jörg Kleeff, Norbert Hüser, Yoshiaki Sunami

Published in: BMC Cancer | Issue 1/2019

Login to get access

Abstract

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 in a ‘carrot and stick’ manner; on one side avoiding cellular crisis while also supporting hepatocellular carcinogenesis. Clinically, we suggest analyzing FOXO3 activation status in patients with liver diseases, in addition to PI3K/Akt signaling. Personalized therapy of FOXO3 inhibition may be a reasonable, depending on the activation status of FOXO3.
Literature
1.
go back to reference Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRef Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRef
2.
go back to reference Ringelhan M, Pfister D, O'Connor T, Pikarsky E, Heikenwalder M. The immunology of hepatocellular carcinoma. Nat Immunol. 2018;19:222–32.CrossRef Ringelhan M, Pfister D, O'Connor T, Pikarsky E, Heikenwalder M. The immunology of hepatocellular carcinoma. Nat Immunol. 2018;19:222–32.CrossRef
3.
go back to reference Vanhaesebroeck B, Stephens L, Hawkins P. PI3K signalling: the path to discovery and understanding. Nat Rev Mol Cell Biol. 2012;13:195–203.CrossRef Vanhaesebroeck B, Stephens L, Hawkins P. PI3K signalling: the path to discovery and understanding. Nat Rev Mol Cell Biol. 2012;13:195–203.CrossRef
4.
go back to reference Matter MS, Decaens T, Andersen JB, Thorgeirsson SS. Targeting the mTOR pathway in hepatocellular carcinoma: current state and future trends. J Hepatol. 2014;60:855–65.CrossRef Matter MS, Decaens T, Andersen JB, Thorgeirsson SS. Targeting the mTOR pathway in hepatocellular carcinoma: current state and future trends. J Hepatol. 2014;60:855–65.CrossRef
5.
go back to reference Hornsveld M, Dansen TB, Derksen PW, Burgering BMT. Re-evaluating the role of FOXOs in cancer. Semin Cancer Biol. 2018;50:90–100.CrossRef Hornsveld M, Dansen TB, Derksen PW, Burgering BMT. Re-evaluating the role of FOXOs in cancer. Semin Cancer Biol. 2018;50:90–100.CrossRef
6.
go back to reference Coomans de Brachène A, Demoulin JB. FOXO transcription factors in cancer development and therapy. Cell Mol Life Sci. 2016;73:1159–72.CrossRef Coomans de Brachène A, Demoulin JB. FOXO transcription factors in cancer development and therapy. Cell Mol Life Sci. 2016;73:1159–72.CrossRef
7.
go back to reference Salih DA, Brunet A. FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol. 2008;20:126–36.CrossRef Salih DA, Brunet A. FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol. 2008;20:126–36.CrossRef
8.
go back to reference van der Vos KE, Coffer PJ. The extending network of FOXO transcriptional target genes. Antioxid Redox Signal. 2011;14:579–92.CrossRef van der Vos KE, Coffer PJ. The extending network of FOXO transcriptional target genes. Antioxid Redox Signal. 2011;14:579–92.CrossRef
9.
go back to reference Eijkelenboom A, Burgering BM. FOXOs: signalling integrators for homeostasis maintenance. Nat Rev Mol Cell Biol. 2013;14:83–97.CrossRef Eijkelenboom A, Burgering BM. FOXOs: signalling integrators for homeostasis maintenance. Nat Rev Mol Cell Biol. 2013;14:83–97.CrossRef
10.
go back to reference Qian Z, Ren L, Wu D, Yang X, Zhou Z, Nie Q, et al. Overexpression of FoxO3a is associated with glioblastoma progression and predicts poor patient prognosis. Int J Cancer. 2017;140:2792–804.CrossRef Qian Z, Ren L, Wu D, Yang X, Zhou Z, Nie Q, et al. Overexpression of FoxO3a is associated with glioblastoma progression and predicts poor patient prognosis. Int J Cancer. 2017;140:2792–804.CrossRef
11.
go back to reference Kumazoe M, Takai M, Bae J, Hiroi S, Huang Y, Takamatsu K, et al. FOXO3 is essential for CD44 expression in pancreatic cancer cells. Oncogene. 2017;36:2643–54.CrossRef Kumazoe M, Takai M, Bae J, Hiroi S, Huang Y, Takamatsu K, et al. FOXO3 is essential for CD44 expression in pancreatic cancer cells. Oncogene. 2017;36:2643–54.CrossRef
12.
go back to reference Hornsveld M, Smits LMM, Meerlo M, van Amersfoort MJA, Groot Koerkamp MJA, van Leenen D, et al. FOXO transcription factors both suppress and support breast Cancer progression. Cancer Res. 2018;78:2356–69.CrossRef Hornsveld M, Smits LMM, Meerlo M, van Amersfoort MJA, Groot Koerkamp MJA, van Leenen D, et al. FOXO transcription factors both suppress and support breast Cancer progression. Cancer Res. 2018;78:2356–69.CrossRef
13.
go back to reference Sunami Y, Leithäuser F, Gul S, Fiedler K, Güldiken N, Espenlaub S, et al. Hepatic activation of IKK/NFκB signaling induces liver fibrosis via macrophage-mediated chronic inflammation. Hepatology. 2012;56:1117–28.CrossRef Sunami Y, Leithäuser F, Gul S, Fiedler K, Güldiken N, Espenlaub S, et al. Hepatic activation of IKK/NFκB signaling induces liver fibrosis via macrophage-mediated chronic inflammation. Hepatology. 2012;56:1117–28.CrossRef
14.
go back to reference Sunami Y, Ringelhan M, Kokai E, Lu M, O’Connor T, Lorentzen A, et al. Canonical NF-κB signaling in hepatocytes acts as a tumor-suppressor in hepatitis B virus surface antigen-driven hepatocellular carcinoma by controlling the unfolded protein response. Hepatology. 2016;63:1592–607.CrossRef Sunami Y, Ringelhan M, Kokai E, Lu M, O’Connor T, Lorentzen A, et al. Canonical NF-κB signaling in hepatocytes acts as a tumor-suppressor in hepatitis B virus surface antigen-driven hepatocellular carcinoma by controlling the unfolded protein response. Hepatology. 2016;63:1592–607.CrossRef
15.
go back to reference Schips TG, Wietelmann A, Höhn K, Schimanski S, Walther P, Braun T, et al. FoxO3 induces reversible cardiac atrophy and autophagy in a transgenic mouse model. Cardiovasc Res. 2011;91:587–97.CrossRef Schips TG, Wietelmann A, Höhn K, Schimanski S, Walther P, Braun T, et al. FoxO3 induces reversible cardiac atrophy and autophagy in a transgenic mouse model. Cardiovasc Res. 2011;91:587–97.CrossRef
16.
go back to reference Schmidt-Strassburger U, Schips TG, Maier HJ, Kloiber K, Mannella F, Braunstein KE, et al. Expression of constitutively active FoxO3 in murine forebrain leads to a loss of neural progenitors. FASEB J. 2012;26:4990–5001.CrossRef Schmidt-Strassburger U, Schips TG, Maier HJ, Kloiber K, Mannella F, Braunstein KE, et al. Expression of constitutively active FoxO3 in murine forebrain leads to a loss of neural progenitors. FASEB J. 2012;26:4990–5001.CrossRef
17.
go back to reference Heid I, Steiger K, Trajkovic-Arsic M, Settles M, Eßwein MR, Erkan M, et al. Co-clinical assessment of tumor cellularity in pancreatic Cancer. Clin Cancer Res. 2017;23:1461–70.CrossRef Heid I, Steiger K, Trajkovic-Arsic M, Settles M, Eßwein MR, Erkan M, et al. Co-clinical assessment of tumor cellularity in pancreatic Cancer. Clin Cancer Res. 2017;23:1461–70.CrossRef
18.
go back to reference Hagenbuchner J, Kuznetsov A, Hermann M, Hausott B, Obexer P, Ausserlechner MJ. FOXO3-induced reactive oxygen species are regulated by BCL2L11 (Bim) and SESN3. J Cell Sci. 2012;125:1191–203.CrossRef Hagenbuchner J, Kuznetsov A, Hermann M, Hausott B, Obexer P, Ausserlechner MJ. FOXO3-induced reactive oxygen species are regulated by BCL2L11 (Bim) and SESN3. J Cell Sci. 2012;125:1191–203.CrossRef
19.
go back to reference Chen CC, Jeon SM, Bhaskar PT, Nogueira V, Sundararajan D, Tonic I, et al. FoxOs inhibit mTORC1 and activate Akt by inducing the expression of Sestrin3 and Rictor. Dev Cell. 2010;18:592–604.CrossRef Chen CC, Jeon SM, Bhaskar PT, Nogueira V, Sundararajan D, Tonic I, et al. FoxOs inhibit mTORC1 and activate Akt by inducing the expression of Sestrin3 and Rictor. Dev Cell. 2010;18:592–604.CrossRef
20.
go back to reference Yu JS, Cui W. Proliferation, survival and metabolism: the role of PI3K/AKT/mTOR signaling in pluripotency and cell fate determination. Development. 2016;143:3050–60.CrossRef Yu JS, Cui W. Proliferation, survival and metabolism: the role of PI3K/AKT/mTOR signaling in pluripotency and cell fate determination. Development. 2016;143:3050–60.CrossRef
21.
go back to reference Ferber EC, Peck B, Delpuech O, Bell GP, East P, Schulze A. FOXO3a regulates reactive oxygen metabolism by inhibiting mitochondrial gene expression. Cell Death Differ. 2012;19:968–79.CrossRef Ferber EC, Peck B, Delpuech O, Bell GP, East P, Schulze A. FOXO3a regulates reactive oxygen metabolism by inhibiting mitochondrial gene expression. Cell Death Differ. 2012;19:968–79.CrossRef
22.
go back to reference Patra KC, Hay N. The pentose phosphate pathway and cancer. Trends Biochem Sci. 2014;39:347–54.CrossRef Patra KC, Hay N. The pentose phosphate pathway and cancer. Trends Biochem Sci. 2014;39:347–54.CrossRef
23.
go back to reference Stanton RC. Glucose-6-phosphate dehydrogenase, NADPH, and cell survival. IUBMB Life. 2012;64:362–9.CrossRef Stanton RC. Glucose-6-phosphate dehydrogenase, NADPH, and cell survival. IUBMB Life. 2012;64:362–9.CrossRef
24.
go back to reference Essaghir A, Dif N, Marbehant CY, Coffer PJ, Demoulin JB. The transcription of FOXO genes is stimulated by FOXO3 and repressed by growth factors. J Biol Chem. 2009;284:10334–42.CrossRef Essaghir A, Dif N, Marbehant CY, Coffer PJ, Demoulin JB. The transcription of FOXO genes is stimulated by FOXO3 and repressed by growth factors. J Biol Chem. 2009;284:10334–42.CrossRef
25.
go back to reference Wang X, Ding J, Feng Y, Weng L, Zhao G, Xiang J, et al. Targeting of growth factors in the treatment of hepatocellular carconima: the potentials of polysaccharides. Oncol Lett. 2017;13:1509–17.CrossRef Wang X, Ding J, Feng Y, Weng L, Zhao G, Xiang J, et al. Targeting of growth factors in the treatment of hepatocellular carconima: the potentials of polysaccharides. Oncol Lett. 2017;13:1509–17.CrossRef
26.
go back to reference Paik JH, Kollipara R, Chu G, Ji H, Xiao Y, Ding Z, et al. FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell. 2007;128:309–23.CrossRef Paik JH, Kollipara R, Chu G, Ji H, Xiao Y, Ding Z, et al. FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell. 2007;128:309–23.CrossRef
27.
go back to reference Santamaría CM, Chillón MC, García-Sanz R, Pérez C, Caballero MD, Ramos F, et al. High FOXO3a expression is associated with a poorer prognosis in AML with normal cytogenetics. Leuk Res. 2009;33:1706–9.CrossRef Santamaría CM, Chillón MC, García-Sanz R, Pérez C, Caballero MD, Ramos F, et al. High FOXO3a expression is associated with a poorer prognosis in AML with normal cytogenetics. Leuk Res. 2009;33:1706–9.CrossRef
28.
go back to reference Benito A, Polat IH, Noé V, Ciudad CJ, Marin S, Cascante M. Glucose-6-phosphate dehydrogenase and transketolase modulate breast cancer cell metabolic reprogramming and correlate with poor patient outcome. Oncotarget. 2017;8:106693–706.CrossRef Benito A, Polat IH, Noé V, Ciudad CJ, Marin S, Cascante M. Glucose-6-phosphate dehydrogenase and transketolase modulate breast cancer cell metabolic reprogramming and correlate with poor patient outcome. Oncotarget. 2017;8:106693–706.CrossRef
30.
go back to reference Pickering CR, Zhou JH, Lee JJ, Drummond JA, Peng SA, Saade RE, et al. Mutational landscape of aggressive cutaneous squamous cell carcinoma. Clin Cancer Res. 2014;20:6582–92.CrossRef Pickering CR, Zhou JH, Lee JJ, Drummond JA, Peng SA, Saade RE, et al. Mutational landscape of aggressive cutaneous squamous cell carcinoma. Clin Cancer Res. 2014;20:6582–92.CrossRef
31.
go back to reference Carbajo-Pescador S, Mauriz JL, García-Palomo A, González-Gallego J. FoxO proteins: regulation and molecular targets in liver cancer. Curr Med Chem. 2014;21:1231–46.CrossRef Carbajo-Pescador S, Mauriz JL, García-Palomo A, González-Gallego J. FoxO proteins: regulation and molecular targets in liver cancer. Curr Med Chem. 2014;21:1231–46.CrossRef
32.
go back to reference Wang Q, Chen X, Hay N. Akt as a target for cancer therapy: more is not always better (lessons from studies in mice). Br J Cancer. 2017;117:159–63.CrossRef Wang Q, Chen X, Hay N. Akt as a target for cancer therapy: more is not always better (lessons from studies in mice). Br J Cancer. 2017;117:159–63.CrossRef
Metadata
Title
Oncogenic Akt-FOXO3 loop favors tumor-promoting modes and enhances oxidative damage-associated hepatocellular carcinogenesis
Authors
Miao Lu
Daniel Hartmann
Rickmer Braren
Aayush Gupta
Baocai Wang
Yang Wang
Carolin Mogler
Zhangjun Cheng
Thomas Wirth
Helmut Friess
Jörg Kleeff
Norbert Hüser
Yoshiaki Sunami
Publication date
01-12-2019
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2019
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-6110-6

Other articles of this Issue 1/2019

BMC Cancer 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine