Oncotarget

Priority Research Papers:

Peretinoin, an acyclic retinoid, suppresses steatohepatitis and tumorigenesis by activating autophagy in mice fed an atherogenic high-fat diet

Hikari Okada, Riuta Takabatake, Masao Honda _, Kai Takegoshi, Taro Yamashita, Mikiko Nakamura, Takayoshi Shirasaki, Yoshio Sakai, Tetsuro Shimakami, Naoto Nagata, Toshinari Takamura, Takuji Tanaka and Shuichi Kaneko

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:39978-39993. https://doi.org/10.18632/oncotarget.18116

Metrics: PDF 2487 views  |   HTML 3463 views  |   ?  


Abstract

Hikari Okada1,*, Riuta Takabatake1,*, Masao Honda1,2, Kai Takegoshi1, Taro Yamashita1, Mikiko Nakamura1, Takayoshi Shirasaki2, Yoshio Sakai1, Tetsuro Shimakami1, Naoto Nagata3, Toshinari Takamura4, Takuji Tanaka5 and Shuichi Kaneko1,4

1 Department of Gastroenterology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan

2 Department of Advanced Medical Technology, Kanazawa University Graduate School of Health Medicine, Kanazawa, Japan

3 Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Japan

4 Department of Disease Control and Homeostasis, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan

5 The Tohkai Cytopathology Institute, Cancer Research and Prevention, Gifu, Japan

* These authors have contributed equally to this work

Correspondence to:

Masao Honda, email:

Keywords: acyclic retinoid, non-alcoholic fatty liver disease (NAFLD), hepatocellular carcinoma (HCC), autophagy

Received: January 25, 2017 Accepted: May 12, 2017 Published: May 23, 2017

Abstract

The pathogenesis of non-alcoholic steatohepatitis (NASH) is still unclear and the prevention of the development of hepatocellular carcinoma (HCC) has not been established. We established an atherogenic and high-fat diet mouse model that develops hepatic steatosis, inflammation, fibrosis, and liver tumors at a high frequency. Using two NASH-HCC mouse models, we showed that peretinoin, an acyclic retinoid, significantly improved liver histology and reduced the incidence of liver tumors. Interestingly, we found that peretinoin induced autophagy in the liver of mice, which was characterized by the increased co-localized expression of microtubule-associated protein light chain 3B-II and lysosome-associated membrane protein 2, and increased autophagosome formation and autophagy flux in the liver. These findings were confirmed using primary mouse hepatocytes. Among representative autophagy pathways, the autophagy related (Atg) 5-Atg12-Atg16L1 pathway was impaired; especially, Atg16L1 was repressed at both the mRNA and protein level. Decreased Atg16L1 mRNA expression was also found in the liver of patients with NASH according to disease progression. Promoter analysis revealed that peretinoin activated the promoter of Atg16L1 by increasing the expression of CCAAT/enhancer-binding-protein-alpha. Interestingly, Atg16L1 overexpression in HepG2 cells inhibited palmitate-induced NF-kB activation and interleukin-6-induced STAT3 activation. We showed that Atg16L1 induced the de-phosphorylation of Gp130, a receptor subunit of interleukin-6 family cytokines, which subsequently repressed phosphorylated-STAT3 (Tyr705) levels, and this process might be independent of autophagy function. Thus, peretinoin prevents the progression of NASH and the development of HCC through activating the autophagy pathway by increased Atg16L1 expression, which is an essential regulator of autophagy and anti-inflammatory proteins.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 18116