1. Academic Validation
  2. NEK2 promotes the migration and proliferation of ESCC via stabilization of YAP1 by phosphorylation at Thr-143

NEK2 promotes the migration and proliferation of ESCC via stabilization of YAP1 by phosphorylation at Thr-143

  • Cell Commun Signal. 2022 Jun 15;20(1):87. doi: 10.1186/s12964-022-00898-0.
Wei Su  # 1 2 Hao Hu  # 1 2 Qiurong Ding 3 Min Wang 4 Yan Zhu 1 2 Zhaochao Zhang 1 2 Zihan Geng 1 2 Shengli Lin 5 6 Pinghong Zhou 7 8
Affiliations

Affiliations

  • 1 Zhongshan Hospital Fudan University Endoscopy Center, Shanghai, 200032, China.
  • 2 Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, 200032, China.
  • 3 Institute of Nutrition Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
  • 4 Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, 200062, China.
  • 5 Zhongshan Hospital Fudan University Endoscopy Center, Shanghai, 200032, China. [email protected].
  • 6 Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, 200032, China. [email protected].
  • 7 Zhongshan Hospital Fudan University Endoscopy Center, Shanghai, 200032, China. [email protected].
  • 8 Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, 200032, China. [email protected].
  • # Contributed equally.
Abstract

Background: Esophageal Squamous Cell Carcinoma (ESCC) was characterized as a regional-prevalent and aggressive tumor with high morbidity and mortality. NIMA-related kinase 2 (NEK2) is an interesting oncogene, the alteration of which leads to patients-beneficial outcomes. We aimed to explore the role of NEK2 in ESCC and excavate its mechanism.

Methods: RNA-seq data were downloaded from TCGA and GEO and analyzed by R software. The protein levels were detected by immunohistochemistry (IHC) or western blot (WB), and mRNA expression was detected by qRT-PCR. The in vitro role of proliferation and migration was detected by Transwell migration assay and by colony formation assay, respectively. The in vivo roles were explored using a subcutaneous xenograft tumor model, where immunofluorescence (IF) and IHC were employed to investigate expression and localization. The interaction between proteins was detected by immunoprecipitation. The stability of proteins was measured by WB in the presence of cycloheximide.

Results: A higher level of NEK2 was found in ESCC than normal esophageal epithelia in GEO, TCGA, and tissue microarray, which was associated with worse prognoses. The NEK2 knockdown impaired the proliferation and migration of ESCC, which also downregulated YAP1 and EMT markers like N-Cadherin and Vimentin in vitro. On the contrary, NEK2 overexpression enhanced the migration of ESCC and elevated the levels of YAP1, N-Cadherin, and Vimentin. Additionally, the overexpression of YAP1 in NEK2 knocked down ESCCs partly rescued the corresponding decrease in migration. The knockdown of NEK2 played an anti-tumor role in vivo and was accompanied by a lower level and nucleus shuffling of YAP1. In mechanism, NEK2 interacted with YAP1 and increased the stability of both endogenous and exogenous YAP1 by preventing ubiquitination. Moreover, the computer-predicted phosphorylation site of YAP1, Thr-143, reduced the ubiquitination of HA-YAP1, strengthened its stability, and thus influenced the migration in vitro.

Conclusions: NEK2 is a prognostic oncogene highly expressed in ESCC and promotes the progression of ESCC in vitro and in vivo. Mechanistically, NEK2-mediated phosphorylation of YAP1 at Thr-143 protects it from Proteasome degradation and might serve as a promising therapeutic target in ESCC. Video Abstract.

Keywords

Epithelial-mesenchymal transition; Esophageal squamous cell carcinoma; NEK2; Phosphorylation; YAP1.

Figures
Products