Cargando…

5-Azacytidine promotes invadopodia formation and tumor metastasis through the upregulation of PI3K in ovarian cancer cells

The high incidence of metastasis accounts for most of the lethality of ovarian cancer. Invadopodia are small, specialized types of machinery that degrade the extracellular matrix and are thus involved in the invasion and metastasis of cancer cells. The formation of invadopodia is regulated by both g...

Descripción completa

Detalles Bibliográficos
Autores principales: Cao, Dan, Li, Dan, Huang, Yong, Ma, Yu, Zhang, Binglan, Zhao, Chengjian, Deng, Senyi, Luo, Min, Yin, Tao, Wei, Yu-Quan, Wang, Wei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5601130/
https://www.ncbi.nlm.nih.gov/pubmed/28947962
http://dx.doi.org/10.18632/oncotarget.18580
_version_ 1783264332347342848
author Cao, Dan
Li, Dan
Huang, Yong
Ma, Yu
Zhang, Binglan
Zhao, Chengjian
Deng, Senyi
Luo, Min
Yin, Tao
Wei, Yu-Quan
Wang, Wei
author_facet Cao, Dan
Li, Dan
Huang, Yong
Ma, Yu
Zhang, Binglan
Zhao, Chengjian
Deng, Senyi
Luo, Min
Yin, Tao
Wei, Yu-Quan
Wang, Wei
author_sort Cao, Dan
collection PubMed
description The high incidence of metastasis accounts for most of the lethality of ovarian cancer. Invadopodia are small, specialized types of machinery that degrade the extracellular matrix and are thus involved in the invasion and metastasis of cancer cells. The formation of invadopodia is regulated by both genetic and epigenetic factors. However, the ways by which methylation/demethylation regulates the dynamics of invadopodia in ovarian cancer are largely unknown. In this study, we found that the inhibition of methylation by 5-AZ (5-Azacytidine) increased the formation of invadopodia and enhanced degradation of the extracellular matrix in ovarian cancer cells. In mouse xenograft models, treatment with 5-AZ increased the number of metastatic nodules, which suggests an elevated potential for metastasis by demethylation. Further investigation indicated that the inhibition of methylation elevated the transcription of PIK3CA and upregulated genes involved in the PI3K-AKT signaling pathway. In addition, this induction likely occurs though the epigenetic regulation of PIK3CA because analyses of the DNA methylation level of the PIK3CA promoter region found that 5-AZ treatment decreased the methylation of CpG islands in SKOV3 and A2780 cells. Our study demonstrated that epigenetic factors regulate the metastatic potential of ovarian cancer cells and provide rationale for therapies that inhibit PI3K- invadopodia-mediated metastasis.
format Online
Article
Text
id pubmed-5601130
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-56011302017-09-25 5-Azacytidine promotes invadopodia formation and tumor metastasis through the upregulation of PI3K in ovarian cancer cells Cao, Dan Li, Dan Huang, Yong Ma, Yu Zhang, Binglan Zhao, Chengjian Deng, Senyi Luo, Min Yin, Tao Wei, Yu-Quan Wang, Wei Oncotarget Research Paper The high incidence of metastasis accounts for most of the lethality of ovarian cancer. Invadopodia are small, specialized types of machinery that degrade the extracellular matrix and are thus involved in the invasion and metastasis of cancer cells. The formation of invadopodia is regulated by both genetic and epigenetic factors. However, the ways by which methylation/demethylation regulates the dynamics of invadopodia in ovarian cancer are largely unknown. In this study, we found that the inhibition of methylation by 5-AZ (5-Azacytidine) increased the formation of invadopodia and enhanced degradation of the extracellular matrix in ovarian cancer cells. In mouse xenograft models, treatment with 5-AZ increased the number of metastatic nodules, which suggests an elevated potential for metastasis by demethylation. Further investigation indicated that the inhibition of methylation elevated the transcription of PIK3CA and upregulated genes involved in the PI3K-AKT signaling pathway. In addition, this induction likely occurs though the epigenetic regulation of PIK3CA because analyses of the DNA methylation level of the PIK3CA promoter region found that 5-AZ treatment decreased the methylation of CpG islands in SKOV3 and A2780 cells. Our study demonstrated that epigenetic factors regulate the metastatic potential of ovarian cancer cells and provide rationale for therapies that inhibit PI3K- invadopodia-mediated metastasis. Impact Journals LLC 2017-06-20 /pmc/articles/PMC5601130/ /pubmed/28947962 http://dx.doi.org/10.18632/oncotarget.18580 Text en Copyright: © 2017 Cao et al. http://creativecommons.org/licenses/by/3.0/ This article is distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0/) (CC-BY), which permits unrestricted use and redistribution provided that the original author and source are credited.
spellingShingle Research Paper
Cao, Dan
Li, Dan
Huang, Yong
Ma, Yu
Zhang, Binglan
Zhao, Chengjian
Deng, Senyi
Luo, Min
Yin, Tao
Wei, Yu-Quan
Wang, Wei
5-Azacytidine promotes invadopodia formation and tumor metastasis through the upregulation of PI3K in ovarian cancer cells
title 5-Azacytidine promotes invadopodia formation and tumor metastasis through the upregulation of PI3K in ovarian cancer cells
title_full 5-Azacytidine promotes invadopodia formation and tumor metastasis through the upregulation of PI3K in ovarian cancer cells
title_fullStr 5-Azacytidine promotes invadopodia formation and tumor metastasis through the upregulation of PI3K in ovarian cancer cells
title_full_unstemmed 5-Azacytidine promotes invadopodia formation and tumor metastasis through the upregulation of PI3K in ovarian cancer cells
title_short 5-Azacytidine promotes invadopodia formation and tumor metastasis through the upregulation of PI3K in ovarian cancer cells
title_sort 5-azacytidine promotes invadopodia formation and tumor metastasis through the upregulation of pi3k in ovarian cancer cells
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5601130/
https://www.ncbi.nlm.nih.gov/pubmed/28947962
http://dx.doi.org/10.18632/oncotarget.18580
work_keys_str_mv AT caodan 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT lidan 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT huangyong 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT mayu 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT zhangbinglan 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT zhaochengjian 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT dengsenyi 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT luomin 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT yintao 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT weiyuquan 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells
AT wangwei 5azacytidinepromotesinvadopodiaformationandtumormetastasisthroughtheupregulationofpi3kinovariancancercells