The Oncolytic Avian Reovirus p17 Protein Inhibits Invadopodia Formation in Murine Melanoma Cancer Cells by Suppressing the FAK/Src Pathway and the Formation of theTKs5/NCK1 Complex

Author:

Hsu Chao-Yu12,Li Jyun-Yi3,Yang En-Ying3,Liao Tsai-Ling24ORCID,Wen Hsiao-Wei5ORCID,Tsai Pei-Chien26,Ju Tz-Chuen3,Lye Lon-Fye7,Nielsen Brent L.8ORCID,Liu Hung-Jen236910ORCID

Affiliation:

1. Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan

2. Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan

3. Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan

4. Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan

5. Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan

6. Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan

7. Department of Medical Research, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan

8. Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA

9. The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan

10. Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan

Abstract

To explore whether the p17 protein of oncolytic avian reovirus (ARV) mediates cell migration and invadopodia formation, we applied several molecular biological approaches for studying the involved cellular factors and signal pathways. We found that ARV p17 activates the p53/phosphatase and tensin homolog (PTEN) pathway to suppress the focal adhesion kinase (FAK)/Src signaling and downstream signal molecules, thus inhibiting cell migration and the formation of invadopodia in murine melanoma cancer cell line (B16-F10). Importantly, p17-induced formation of invadopodia could be reversed in cells transfected with the mutant PTENC124A. p17 protein was found to significantly reduce the expression levels of tyrosine kinase substrate 5 (TKs5), Rab40b, non-catalytic region of tyrosine kinase adaptor protein 1 (NCK1), and matrix metalloproteinases (MMP9), suggesting that TKs5 and Rab40b were transcriptionally downregulated by p17. Furthermore, we found that p17 suppresses the formation of the TKs5/NCK1 complex. Coexpression of TKs5 and Rab40b in B16-F10 cancer cells reversed p17-modulated suppression of the formation of invadopodia. This work provides new insights into p17-modulated suppression of invadopodia formation by activating the p53/PTEN pathway, suppressing the FAK/Src pathway, and inhibiting the formation of the TKs5/NCK1 complex.

Funder

Ministry of Science and Technology of Taiwan

the Ministry of Education (MOE) in Taiwan

National Chung Hsing University, and Taichung Veterans General Hospital

Publisher

MDPI AG

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3