Your browser doesn't support javascript.
loading
KCTD17-mediated Ras stabilization promotes hepatocellular carcinoma progression.
Jung, Young Hoon; Lee, Yun Ji; Dao, Tam; Jung, Kyung Hee; Yu, Junjie; Oh, Ah-Reum; Jeong, Yelin; Gi, HyunJoon; Kim, Young Un; Ryu, Dongryeol; Carrer, Michele; Pajvani, Utpal B; Lee, Sang Bae; Hong, Soon-Sun; Kim, KyeongJin.
Afiliación
  • Jung YH; Department of Biomedical Sciences, College of Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Lee YJ; Program in Biomedical Science & Engineering, College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Dao T; Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Jung KH; Department of Biomedical Sciences, College of Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Yu J; Program in Biomedical Science & Engineering, College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Oh AR; Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Jeong Y; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
  • Gi H; Department of Biomedical Sciences, College of Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Kim YU; Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Ryu D; Department of Medicine, Columbia University, New York, NY 10032, USA.
  • Carrer M; Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China.
  • Pajvani UB; Department of Biomedical Sciences, College of Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Lee SB; Program in Biomedical Science & Engineering, College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Hong SS; Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea.
  • Kim K; Department of Biomedical Sciences, College of Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea.
Clin Mol Hepatol ; 2024 Aug 05.
Article en En | MEDLINE | ID: mdl-39098817
ABSTRACT
Background/

Aims:

Potassium channel tetramerization domain containing 17 (KCTD17) protein, an adaptor for the cullin3 (Cul3) ubiquitin ligase complex, has been implicated in various human diseases; however, its role in hepatocellular carcinoma (HCC) remains elusive. Here, we aimed to elucidate the clinical features of KCTD17, and investigate the mechanisms by which KCTD17 affects HCC progression.

Methods:

We analyzed transcriptomic data from patients with HCC. Hepatocyte-specific KCTD17 deficient mice were treated with diethylnitrosamine (DEN) to assess its effect on HCC progression. Additionally, we tested KCTD17-directed antisense oligonucleotides for their therapeutic potential in vivo.

Results:

Our investigation revealed the upregulation of KCTD17 expression in both tumors from patients with HCC and mouse models of HCC, in comparison to non-tumor controls. We identified the leucine zipper-like transcriptional regulator 1 (Lztr1) protein, a previously identified Ras destabilizer, as a substrate for KCTD17-Cul3 complex. KCTD17-mediated Lztr1 degradation led to Ras stabilization, resulting in increased proliferation, migration, and wound healing in liver cancer cells. Hepatocyte-specific KCTD17 deficient mice or liver cancer xenograft models were less susceptible to carcinogenesis or tumor growth. Similarly, treatment with KCTD17-directed antisense oligonucleotides (ASO) in a mouse model of HCC markedly lowered tumor volume as well as Ras protein levels, compared to those in control ASO-treated mice.

Conclusions:

KCTD17 induces the stabilization of Ras and downstream signaling pathways and HCC progression and may represent a novel therapeutic target for HCC.
Palabras clave

Texto completo: 1 Base de datos: MEDLINE Idioma: En Revista: Clin Mol Hepatol Año: 2024 Tipo del documento: Article

Texto completo: 1 Base de datos: MEDLINE Idioma: En Revista: Clin Mol Hepatol Año: 2024 Tipo del documento: Article