1. Academic Validation
  2. TRIM36 inhibits tumorigenesis through the Wnt/β-catenin pathway and promotes caspase-dependent apoptosis in hepatocellular carcinoma

TRIM36 inhibits tumorigenesis through the Wnt/β-catenin pathway and promotes caspase-dependent apoptosis in hepatocellular carcinoma

  • Cancer Cell Int. 2022 Sep 6;22(1):278. doi: 10.1186/s12935-022-02692-x.
Qing Tong  # 1 2 Mingyu Yi  # 3 Panpan Kong 1 Lin Xu 4 Wukui Huang 2 Yue Niu 2 Xiaojing Gan 2 Huan Zhan 2 Rui Tian 5 Dong Yan 6 7
Affiliations

Affiliations

  • 1 Department of Hepatopancreatobiliary Surgery, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China.
  • 2 The Third Affiliated, Teaching Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
  • 3 The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
  • 4 National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.
  • 5 Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
  • 6 Department of Hepatopancreatobiliary Surgery, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China. [email protected].
  • 7 The Third Affiliated, Teaching Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China. [email protected].
  • # Contributed equally.
Abstract

Background: Hepatocellular carcinoma (HCC) is the most common type of primary liver Cancer and has an extremely poor prognosis. We aimed to determine the latent relationships between TRIM36 regulation of Apoptosis and the Wnt/β-catenin pathway in HCC.

Methods: Immunohistochemistry and western blotting were used to characterize the aberrant expression of TRIM36 in HCC and adjacent tissues. Clinical information was analyzed using Kaplan-Meier and COX methods. RNA-seq of potential targets was conducted to detect the regulation of TRIM36. Apoptosis assays and cellular proliferation, invasion and migration were conducted in a loss- and gain-of-function manner in cultured cells to determine the biological functions of TRIM36. A rescue experiment was conducted to confirm the role of Wnt/β-catenin signaling in TRIM36 regulation. Finally, in vivo experiments were conducted using cell line-derived xenografts in nude mice to validate the central role of TRIM36 in HCC.

Results: TRIM36 expression was significantly downregulated in HCC tissues compared to adjacent non-tumor tissues. TRIM36 repressed the proliferation, migration, and invasion of Huh7 and HCCLM3 cells, whereas it stimulated Apoptosis. Wnt/β-catenin signaling was inhibited by TRIM36, and rescue experiments highlighted its importance in HCC proliferation, migration, and invasion. In vivo experiments further confirmed the effects of sh-TRIM36 on HCC tumorigenesis, inhibition of Apoptosis, and promotion of Wnt/β-catenin signaling.

Conclusion: Our study is the first to indicate that TRIM36 acts as a tumor suppressor in HCC. TRIM36 activates Apoptosis and inhibits cellular proliferation, invasion, and migration via the Wnt/β-catenin pathway, which may serve as an important biomarker and promising therapeutic target for HCC.

Keywords

Apoptosis; Caspase-3; Caspase-7; RNA-seq; TRIM36; Wnt/β-catenin pathway.

Figures