Your browser doesn't support javascript.
loading
ECRG4 acts as a tumor suppressor in nasopharyngeal carcinoma by suppressing the AKT/GSK3ß/ß-catenin signaling pathway.
Yang, Zhengyuan; Ye, Xiajun; Zhang, Yujie; Huang, Yiteng; Chen, Jian; Zeng, Yunzhu; Chen, Jiongyu.
Afiliação
  • Yang Z; Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515031 People's Republic of China.
  • Ye X; Department of Medical Affairs, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515031 People's Republic of China.
  • Zhang Y; Department of Chemical Engineering, Guangdong Technion-Israel Institute of Technology, No. 241 Daxue Road, Shantou, 515031 People's Republic of China.
  • Huang Y; Health Care Center, The First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, 515041 People's Republic of China.
  • Chen J; Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. Raoping Road, Shantou, 515031 People's Republic of China.
  • Zeng Y; Department of Pathology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515031 Guangdong Province People's Republic of China.
  • Chen J; Oncological Research Lab, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515031 Guangdong Province People's Republic of China.
Cytotechnology ; 74(2): 231-243, 2022 Apr.
Article em En | MEDLINE | ID: mdl-35464163
ABSTRACT
Nasopharyngeal carcinoma (NPC) is a malignant tumor with a poor prognosis. Studies have shown that esophageal carcinoma related gene 4 (ECRG4) is hypermethylated and significantly downregulated in NPC tissues. However, the role of ECRG4 in NPC, and in particular the underlying molecular mechanism, is largely unclear. In this study, using immunohistochemical staining of ECRG4 in NPC and normal specimens, we confirmed that ECRG4 was downregulated in human NPC tissues. In addition, various biological and molecular studies were carried out and the results showed that ECRG4 exerted anticancer effect in NPC, including inhibiting cell growth, migration, and invasion of NPC cells in vitro. Moreover, restoring ECRG4 expression suppressed the in vivo tumorigenesis of CNE2 cells. ECRG4 inhibited AKT/GSK3ß/ß-catenin signaling, as well as the downstream targets of ß-catenin. LiCl treatment, which reduced GSK3ß phosphorylation and upregulated ß-catenin expression, restored the invasive ability of ECRG4-overexpressing NPC cells. Furthermore, we showed that the DNA methylation inhibitor 5-aza-dC reduced ECRG4 methylation and the invasive ability of negative control cells, but not that of ECRG4-overexpressing cells, suggesting that the inhibitory effect of 5-aza-dC depends on low expression of ECRG4. Collectively, our results demonstrated that ECRG4 downregulation contributed to NPC growth and invasion by activating AKT/GSK3ß/ß-catenin signaling pathway. ECRG4 could be a promising therapeutic target for the treatment of NPC. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-022-00520-8.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2022 Tipo de documento: Article