Your browser doesn't support javascript.
loading
MICAL2 contributes to gastric cancer cell migration via Cdc42-dependent activation of E-cadherin/ß-catenin signaling pathway.
Wang, Qianwen; Qi, Chenxiang; Min, Pengxiang; Wang, Yueyuan; Ye, Fengwen; Xia, Tianxiang; Zhang, Yujie; Du, Jun.
Affiliation
  • Wang Q; Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China.
  • Qi C; Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China.
  • Min P; Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
  • Wang Y; Experimental Teaching Center of Basic Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
  • Ye F; Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China.
  • Xia T; Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China.
  • Zhang Y; Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China.
  • Du J; Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China. dujun@njmu.edu.cn.
Cell Commun Signal ; 20(1): 136, 2022 09 05.
Article in En | MEDLINE | ID: mdl-36064550
ABSTRACT

BACKGROUND:

Gastric cancer is a common and lethal human malignancy worldwide and cancer cell metastasis is the leading cause of cancer-related mortality. MICAL2, a flavoprotein monooxygenase, is an important regulator of epithelial-to-mesenchymal transition. The aim of this study was to explore the effects of MICAL2 on gastric cancer cell migration and determine the underlying molecular mechanisms.

METHODS:

Cell migration was examined by wound healing and transwell assays. Changes in E-cadherin/ß-catenin signaling were determined by qPCR and analysis of cytoplasmic and nuclear protein fractions. E-cadherin/ß-catenin binding was determined by co-immunoprecipitation assays. Cdc42 activity was examined by pulldown assay.

RESULTS:

MICAL2 was highly expressed in gastric cancer tissues. The knockdown of MICAL2 significantly attenuated migratory ability and ß-catenin nuclear translocation in gastric cancer cells while LiCl treatment, an inhibitor of GSK3ß, reversed these MICAL2 knockdown-induced effects. Meanwhile, E-cadherin expression was markedly enhanced in MICAL2-depleted cells. MICAL2 knockdown led to a significant attenuation of E-cadherin ubiquitination and degradation in a Cdc42-dependent manner, then enhanced E-cadherin/ß-catenin binding, and reduced ß-catenin nuclear translocation.

CONCLUSIONS:

Together, our results indicated that MICAL2 promotes E-cadherin ubiquitination and degradation, leading to enhanced ß-catenin signaling via the disruption of the E-cadherin/ß-catenin complex and, consequently, the promotion of gastric cell migration. Video Abstract.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Oxidoreductases / Stomach Neoplasms / Antigens, CD / Cadherins / Cdc42 GTP-Binding Protein / Beta Catenin / Microfilament Proteins Limits: Humans Language: En Journal: Cell Commun Signal Year: 2022 Document type: Article Affiliation country: China

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Oxidoreductases / Stomach Neoplasms / Antigens, CD / Cadherins / Cdc42 GTP-Binding Protein / Beta Catenin / Microfilament Proteins Limits: Humans Language: En Journal: Cell Commun Signal Year: 2022 Document type: Article Affiliation country: China