Cargando…

O-GlcNAcylation promotes migration and invasion in human ovarian cancer cells via the RhoA/ROCK/MLC pathway

O-GlcNAcylation is a dynamic and reversible post-translational modification associated with the regulation of multiple cellular functions. The addition and removal of O-Linked β-N-acetylglucosamine (O-GlcNAc) on target proteins is catalyzed by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respec...

Descripción completa

Detalles Bibliográficos
Autores principales: Niu, Yichao, Xia, Ye, Wang, Jingyun, Shi, Xiaofei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5364967/
https://www.ncbi.nlm.nih.gov/pubmed/28259907
http://dx.doi.org/10.3892/mmr.2017.6244
_version_ 1782517431011377152
author Niu, Yichao
Xia, Ye
Wang, Jingyun
Shi, Xiaofei
author_facet Niu, Yichao
Xia, Ye
Wang, Jingyun
Shi, Xiaofei
author_sort Niu, Yichao
collection PubMed
description O-GlcNAcylation is a dynamic and reversible post-translational modification associated with the regulation of multiple cellular functions. The addition and removal of O-Linked β-N-acetylglucosamine (O-GlcNAc) on target proteins is catalyzed by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Accumulating evidence suggests that O-GlcNAcylation is associated with the malignancy of several types of human cancer. To investigate the effect of O-GlcNAcylation on ovarian cancer phenotypes, global O-GlcNAc levels were decreased by OGT silencing through RNA interference and increased by inhibiting OGA activity with Thiamet-G. Transwell assay results demonstrated that OGT silencing inhibited the migration and invasion of SKOV3 and 59M ovarian cells in vitro, while Thiamet-G treatment promoted migration and invasion. Furthermore, a pull-down assay and western blot analysis demonstrated that Thiamet-G treatment enhanced RhoA activity and the phosphorylation of the Rho-associated protein kinase (ROCK) substrate, myosin light chain (MLC), while OGT silencing attenuated RhoA activity and MLC phosphorylation. In addition, RhoA silencing via RNA interference and inhibition of ROCK activity with Y-27632 prevented Thiamet-G-induced increases in cell migration and invasion. These data suggest that O-GlcNAcylation augments the motility of ovarian cancer cells via the RhoA/ROCK/MLC signaling pathway. Therefore, O-GlcNAcylation may be a potential target for the diagnosis and treatment of ovarian cancer.
format Online
Article
Text
id pubmed-5364967
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-53649672017-05-15 O-GlcNAcylation promotes migration and invasion in human ovarian cancer cells via the RhoA/ROCK/MLC pathway Niu, Yichao Xia, Ye Wang, Jingyun Shi, Xiaofei Mol Med Rep Articles O-GlcNAcylation is a dynamic and reversible post-translational modification associated with the regulation of multiple cellular functions. The addition and removal of O-Linked β-N-acetylglucosamine (O-GlcNAc) on target proteins is catalyzed by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Accumulating evidence suggests that O-GlcNAcylation is associated with the malignancy of several types of human cancer. To investigate the effect of O-GlcNAcylation on ovarian cancer phenotypes, global O-GlcNAc levels were decreased by OGT silencing through RNA interference and increased by inhibiting OGA activity with Thiamet-G. Transwell assay results demonstrated that OGT silencing inhibited the migration and invasion of SKOV3 and 59M ovarian cells in vitro, while Thiamet-G treatment promoted migration and invasion. Furthermore, a pull-down assay and western blot analysis demonstrated that Thiamet-G treatment enhanced RhoA activity and the phosphorylation of the Rho-associated protein kinase (ROCK) substrate, myosin light chain (MLC), while OGT silencing attenuated RhoA activity and MLC phosphorylation. In addition, RhoA silencing via RNA interference and inhibition of ROCK activity with Y-27632 prevented Thiamet-G-induced increases in cell migration and invasion. These data suggest that O-GlcNAcylation augments the motility of ovarian cancer cells via the RhoA/ROCK/MLC signaling pathway. Therefore, O-GlcNAcylation may be a potential target for the diagnosis and treatment of ovarian cancer. D.A. Spandidos 2017-04 2017-02-24 /pmc/articles/PMC5364967/ /pubmed/28259907 http://dx.doi.org/10.3892/mmr.2017.6244 Text en Copyright: © Niu et al. This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Niu, Yichao
Xia, Ye
Wang, Jingyun
Shi, Xiaofei
O-GlcNAcylation promotes migration and invasion in human ovarian cancer cells via the RhoA/ROCK/MLC pathway
title O-GlcNAcylation promotes migration and invasion in human ovarian cancer cells via the RhoA/ROCK/MLC pathway
title_full O-GlcNAcylation promotes migration and invasion in human ovarian cancer cells via the RhoA/ROCK/MLC pathway
title_fullStr O-GlcNAcylation promotes migration and invasion in human ovarian cancer cells via the RhoA/ROCK/MLC pathway
title_full_unstemmed O-GlcNAcylation promotes migration and invasion in human ovarian cancer cells via the RhoA/ROCK/MLC pathway
title_short O-GlcNAcylation promotes migration and invasion in human ovarian cancer cells via the RhoA/ROCK/MLC pathway
title_sort o-glcnacylation promotes migration and invasion in human ovarian cancer cells via the rhoa/rock/mlc pathway
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5364967/
https://www.ncbi.nlm.nih.gov/pubmed/28259907
http://dx.doi.org/10.3892/mmr.2017.6244
work_keys_str_mv AT niuyichao oglcnacylationpromotesmigrationandinvasioninhumanovariancancercellsviatherhoarockmlcpathway
AT xiaye oglcnacylationpromotesmigrationandinvasioninhumanovariancancercellsviatherhoarockmlcpathway
AT wangjingyun oglcnacylationpromotesmigrationandinvasioninhumanovariancancercellsviatherhoarockmlcpathway
AT shixiaofei oglcnacylationpromotesmigrationandinvasioninhumanovariancancercellsviatherhoarockmlcpathway