Cargando…

RGS5 decreases the proliferation of human ovarian carcinoma-derived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia

Regulator of G-protein signaling 5 (RGS5), a tissue-specific signal-regulating molecule, plays a key role in the development of the vasculature. It was recently found that RGS5 is abundantly expressed in epithelial ovarian cancer (EOC) compared with the normal ovaries. However, the distribution of R...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Dan, Xu, Yan, Feng, Lu, Yin, Pin, Song, Shuang Shuang, Wu, Feng, Yan, Ping, Liang, Zhiqing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6278583/
https://www.ncbi.nlm.nih.gov/pubmed/30365142
http://dx.doi.org/10.3892/or.2018.6811
_version_ 1783378398514511872
author Wang, Dan
Xu, Yan
Feng, Lu
Yin, Pin
Song, Shuang Shuang
Wu, Feng
Yan, Ping
Liang, Zhiqing
author_facet Wang, Dan
Xu, Yan
Feng, Lu
Yin, Pin
Song, Shuang Shuang
Wu, Feng
Yan, Ping
Liang, Zhiqing
author_sort Wang, Dan
collection PubMed
description Regulator of G-protein signaling 5 (RGS5), a tissue-specific signal-regulating molecule, plays a key role in the development of the vasculature. It was recently found that RGS5 is abundantly expressed in epithelial ovarian cancer (EOC) compared with the normal ovaries. However, the distribution of RGS5 in EOC and its significance require further investigation. The aim of the present study was to investigate the expression of RGS5 in EOC, as well as its association with cancer differentiation, metastasis and clinicopathological parameters. Immunohistochemistry (IHC), western blotting, RT-PCR, wound-healing, cell proliferation and flow cytometric assays were the methods used in the present study. RGS5 was highly expressed in the cytoplasm of ovarian carcinoma cells and in microvascular structures. The expression of RGS5 in EOC was negatively associated with peritoneal metastasis (P=0.004), but it was not found to be associated with age, tumor size, clinical stage or lymph node metastasis (P>0.05). EOC patients with high RGS5 expression had a prolonged progression-free survival (72.34±8.41 vs. 43.56±5.41 months, P<0.001). High expression of RGS5 was correlated with significantly lower microvascular density (MVD) as indicated by the expression of CD34, whereas the opposite was observed in tissues with low RGS5 expression (P<0.05). Hypoxia increased RGS5 expression in ovarian carcinoma-derived endothelial cells (ODMECs), whereas the proliferative capacity of ODMECs exhibited a significant increase following RNAi-mediated reduction of RGS5 expression. These data indicated that RGS5 plays a key role in angiogenesis in ovarian carcinoma. In addition, RGS5 downregulated the expression of the downstream proteins CDC25A, CDK2 and cyclin E, which are mediated by the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway, causing ODMEC arrest in the G1 phase of the cell cycle under hypoxic conditions. Collectively, our data indicated that RGS5 is crucial for the occurrence and development of ovarian cancer, and that RGS5 and its signaling pathway may serve as anti-angiogenesis targets for the treatment of ovarian cancer.
format Online
Article
Text
id pubmed-6278583
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-62785832018-12-17 RGS5 decreases the proliferation of human ovarian carcinoma-derived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia Wang, Dan Xu, Yan Feng, Lu Yin, Pin Song, Shuang Shuang Wu, Feng Yan, Ping Liang, Zhiqing Oncol Rep Articles Regulator of G-protein signaling 5 (RGS5), a tissue-specific signal-regulating molecule, plays a key role in the development of the vasculature. It was recently found that RGS5 is abundantly expressed in epithelial ovarian cancer (EOC) compared with the normal ovaries. However, the distribution of RGS5 in EOC and its significance require further investigation. The aim of the present study was to investigate the expression of RGS5 in EOC, as well as its association with cancer differentiation, metastasis and clinicopathological parameters. Immunohistochemistry (IHC), western blotting, RT-PCR, wound-healing, cell proliferation and flow cytometric assays were the methods used in the present study. RGS5 was highly expressed in the cytoplasm of ovarian carcinoma cells and in microvascular structures. The expression of RGS5 in EOC was negatively associated with peritoneal metastasis (P=0.004), but it was not found to be associated with age, tumor size, clinical stage or lymph node metastasis (P>0.05). EOC patients with high RGS5 expression had a prolonged progression-free survival (72.34±8.41 vs. 43.56±5.41 months, P<0.001). High expression of RGS5 was correlated with significantly lower microvascular density (MVD) as indicated by the expression of CD34, whereas the opposite was observed in tissues with low RGS5 expression (P<0.05). Hypoxia increased RGS5 expression in ovarian carcinoma-derived endothelial cells (ODMECs), whereas the proliferative capacity of ODMECs exhibited a significant increase following RNAi-mediated reduction of RGS5 expression. These data indicated that RGS5 plays a key role in angiogenesis in ovarian carcinoma. In addition, RGS5 downregulated the expression of the downstream proteins CDC25A, CDK2 and cyclin E, which are mediated by the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway, causing ODMEC arrest in the G1 phase of the cell cycle under hypoxic conditions. Collectively, our data indicated that RGS5 is crucial for the occurrence and development of ovarian cancer, and that RGS5 and its signaling pathway may serve as anti-angiogenesis targets for the treatment of ovarian cancer. D.A. Spandidos 2019-01 2018-10-22 /pmc/articles/PMC6278583/ /pubmed/30365142 http://dx.doi.org/10.3892/or.2018.6811 Text en Copyright: © Wang 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
Wang, Dan
Xu, Yan
Feng, Lu
Yin, Pin
Song, Shuang Shuang
Wu, Feng
Yan, Ping
Liang, Zhiqing
RGS5 decreases the proliferation of human ovarian carcinoma-derived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia
title RGS5 decreases the proliferation of human ovarian carcinoma-derived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia
title_full RGS5 decreases the proliferation of human ovarian carcinoma-derived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia
title_fullStr RGS5 decreases the proliferation of human ovarian carcinoma-derived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia
title_full_unstemmed RGS5 decreases the proliferation of human ovarian carcinoma-derived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia
title_short RGS5 decreases the proliferation of human ovarian carcinoma-derived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia
title_sort rgs5 decreases the proliferation of human ovarian carcinoma-derived primary endothelial cells through the mapk/erk signaling pathway in hypoxia
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6278583/
https://www.ncbi.nlm.nih.gov/pubmed/30365142
http://dx.doi.org/10.3892/or.2018.6811
work_keys_str_mv AT wangdan rgs5decreasestheproliferationofhumanovariancarcinomaderivedprimaryendothelialcellsthroughthemapkerksignalingpathwayinhypoxia
AT xuyan rgs5decreasestheproliferationofhumanovariancarcinomaderivedprimaryendothelialcellsthroughthemapkerksignalingpathwayinhypoxia
AT fenglu rgs5decreasestheproliferationofhumanovariancarcinomaderivedprimaryendothelialcellsthroughthemapkerksignalingpathwayinhypoxia
AT yinpin rgs5decreasestheproliferationofhumanovariancarcinomaderivedprimaryendothelialcellsthroughthemapkerksignalingpathwayinhypoxia
AT songshuangshuang rgs5decreasestheproliferationofhumanovariancarcinomaderivedprimaryendothelialcellsthroughthemapkerksignalingpathwayinhypoxia
AT wufeng rgs5decreasestheproliferationofhumanovariancarcinomaderivedprimaryendothelialcellsthroughthemapkerksignalingpathwayinhypoxia
AT yanping rgs5decreasestheproliferationofhumanovariancarcinomaderivedprimaryendothelialcellsthroughthemapkerksignalingpathwayinhypoxia
AT liangzhiqing rgs5decreasestheproliferationofhumanovariancarcinomaderivedprimaryendothelialcellsthroughthemapkerksignalingpathwayinhypoxia