期刊论文详细信息
Molecular Therapy: Oncolytics
AAMP promotes colorectal cancer metastasis by suppressing SMURF2-mediated ubiquitination and degradation of RhoA
Jiansheng Xie1  Rongjie Zhao2  Yuhui Wu2  Bofang Liu2  Weidong Han2  Weiqiang Lin3 
[1]Corresponding author: Weidong Han, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China.
[2]Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
[3]Laboratory of Cancer Biology, Institute of Clinical Science, The Fourth Affiliated Hospital, College of Medicine, Zhejiang University, Yiwu, Zhejiang 322000, China
关键词: AAMP;    colorectal cancer;    metastasis;    RhoA;    SMURF2;    ubiquitination;   
DOI  :  
来源: DOAJ
【 摘 要 】
Metastasis is considered the leading cause of cancer death due to the limited possibilities to therapeutically target this process. How the ubiquitination machinery contributes to metastasis remains underexplored. Angio-associated migratory cell protein (AAMP), a ubiquitously expressed protein involved in cell migration, has been reported to play oncogenic roles in breast and non-small cell lung cancer (NSCLC). However, the role of AAMP in colorectal cancer (CRC) has not been demonstrated. Here, we report that AAMP is aberrantly upregulated in metastatic CRC and that AAMP upregulation is correlated with the poor survival of CRC patients. AAMP knockdown significantly attenuated the migration and invasion of CRC cells, while AAMP overexpression led to the opposite effects. Mechanistically, we identified Ras homolog family member A (RhoA) as a target of AAMP. Smad ubiquitin regulatory factor (SMURF) 2 was previously found to be a CRC suppressor. Notably, we discovered here that SMURF2 acted as an E3 ubiquitin ligase to mediate the ubiquitination and degradation of RhoA. AAMP stabilized RhoA by binding to it and suppressing its SMURF2-mediated ubiquitination and degradation. Subsequently, the level of active RhoA was increased, thereby accelerating CRC cell migration and invasion. These findings indicate a new potential antitumor target for CRC.
【 授权许可】

Unknown   

  文献评价指标  
  下载次数:0次 浏览次数:0次