Your browser doesn't support javascript.
loading
Mucin1 shifts Smad3 signaling from the tumor-suppressive pSmad3C/p21(WAF1) pathway to the oncogenic pSmad3L/c-Myc pathway by activating JNK in human hepatocellular carcinoma cells.
Li, Qiongshu; Liu, Guomu; Yuan, Hongyan; Wang, Juan; Guo, Yingying; Chen, Tanxiu; Zhai, Ruiping; Shao, Dan; Ni, Weihua; Tai, Guixiang.
Affiliation
  • Li Q; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
  • Liu G; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
  • Yuan H; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
  • Wang J; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
  • Guo Y; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
  • Chen T; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
  • Zhai R; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
  • Shao D; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
  • Ni W; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
  • Tai G; Department of Immunology, College of Basic Medical Science, Jilin University, Changchun 130021, China.
Oncotarget ; 6(6): 4253-65, 2015 Feb 28.
Article in En | MEDLINE | ID: mdl-25714018
ABSTRACT
Mucin1 (MUC1) is a transmembrane glycoprotein that acts as an oncogene in human hepatic tumorigenesis. Hepatocellular carcinoma (HCC) cells often gain advantage by reducing the tumor-suppressive activity of transforming growth factor beta (TGF-ß) together with stimulation of its oncogenic activity as in MUC1 expressing HCC cells; however, molecular mechanisms remain largely unknown. Type I TGF-ß receptor (TßRI) and c-Jun NH2-terminal kinase (JNK) differentially phosphorylate Smad3 mediator to create 2 phosphorylated forms COOH-terminally phosphorylated Smad3 (pSmad3C) and linker-phosphorylated Smad3 (pSmad3L). Here, we report that MUC1 overexpression in HCC cell lines suppresses TßRI-mediated pSmad3C signaling which involves growth inhibition by up-regulating p21(WAF1). Instead, MUC1 directly activates JNK to stimulate oncogenic pSmad3L signaling, which fosters cell proliferation by up-regulating c-Myc. Conversely, MUC1 gene silencing in MUC1 expressing HCC cells results in preserved tumor-suppressive function via pSmad3C, while eliminating pSmad3L-mediated oncogenic activity both in vitro and in vivo. In addition, high correlation between MUC1 and pSmad3L/c-Myc but not pSmad3C/p21(WAF1) expression was observed in HCC tissues from patients. Collectively, these results indicate that MUC1 shifts Smad3 signaling from a tumor-suppressive pSmad3C/p21(WAF1) to an oncogenic pSmad3L/c-Myc pathway by directly activating JNK in HCC cells, suggesting that MUC1 is an important target for HCC therapy.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Signal Transduction / Carcinoma, Hepatocellular / Mucin-1 / Smad3 Protein / Liver Neoplasms Limits: Animals / Humans Language: En Journal: Oncotarget Year: 2015 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Signal Transduction / Carcinoma, Hepatocellular / Mucin-1 / Smad3 Protein / Liver Neoplasms Limits: Animals / Humans Language: En Journal: Oncotarget Year: 2015 Document type: Article Affiliation country: