Reactivation of the tumour suppressor RASSF1A in breast cancer by simultaneous targeting of DNA and E2F1 methylation.

<h4>Background</h4>Tumour suppressor genes are often transcriptionally silenced by promoter hypermethylation, and recent research has implicated alterations in chromatin structure as the mechanistic basis for this repression. In addition to DNA methylation, other epigenetic post-translat...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: María F Montenegro, Magali Sáez-Ayala, Antonio Piñero-Madrona, Juan Cabezas-Herrera, José Neptuno Rodríguez-López
Formato: article
Lenguaje:EN
Publicado: Public Library of Science (PLoS) 2012
Materias:
R
Q
Acceso en línea:https://doaj.org/article/17eabdbfda8d4ce0be8e713fa74c05ba
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:17eabdbfda8d4ce0be8e713fa74c05ba
record_format dspace
spelling oai:doaj.org-article:17eabdbfda8d4ce0be8e713fa74c05ba2021-11-18T08:05:00ZReactivation of the tumour suppressor RASSF1A in breast cancer by simultaneous targeting of DNA and E2F1 methylation.1932-620310.1371/journal.pone.0052231https://doaj.org/article/17eabdbfda8d4ce0be8e713fa74c05ba2012-01-01T00:00:00Zhttps://www.ncbi.nlm.nih.gov/pmc/articles/pmid/23251702/?tool=EBIhttps://doaj.org/toc/1932-6203<h4>Background</h4>Tumour suppressor genes are often transcriptionally silenced by promoter hypermethylation, and recent research has implicated alterations in chromatin structure as the mechanistic basis for this repression. In addition to DNA methylation, other epigenetic post-translational modifications that modulate the stability and binding of specific transcription factors to gene promoters have emerged as important mechanisms for controlling gene expression. The aim of this study was to analyse the implications of these mechanisms and their molecular connections in the reactivation of RASSF1A in breast cancer.<h4>Methods</h4>Compounds that modulate the intracellular concentration of adenosine, such as dipyridamole (DIPY), greatly increase the antiproliferative effects of 3-O-(3,4,5-trimethoxybenzoyl)-(-)-catechin (TMCG), a synthetic antifolate derived from the structure of tea catechins. Quantitative real-time PCR arrays and MALDI-TOF mass spectrometry indicated that this combination (TMCG/DIPY) induced apoptosis in breast cancer cells by modulating the methylation levels of DNA and proteins (such as E2F1), respectively. Chromatin immunoprecipitation (ChIP) assays were employed to confirm that this combination induced chromatin remodelling of the RASSF1A promoter and increased the occupancy of E2F1 at the promoter of this tumour suppressor gene.<h4>Results</h4>The TMCG/DIPY combination acted as an epigenetic treatment that reactivated RASSF1A expression and induced apoptosis in breast cancer cells. In addition to modulating DNA methylation and chromatin remodelling, this combination also induced demethylation of the E2F1 transcription factor. The ChIP assay showed enhancement of E2F1 occupancy at the unmethylated RASSF1A promoter after TMCG/DIPY treatment. Interestingly, inhibition of E2F1 demethylation using an irreversible inhibitor of lysine-specific demethylase 1 reduced both TMCG/DIPY-mediated RASSF1A expression and apoptosis in MDA-MB-231 cells, suggesting that DNA and protein demethylation may act together to control these molecular and cellular processes.<h4>Conclusions/significance</h4>This study demonstrates that simultaneous targeting of DNA and E2F1 methylation is an effective epigenetic treatment that reactivates RASSF1A expression and induces apoptosis in breast cancer cells.María F MontenegroMagali Sáez-AyalaAntonio Piñero-MadronaJuan Cabezas-HerreraJuan Cabezas-HerreraJosé Neptuno Rodríguez-LópezPublic Library of Science (PLoS)articleMedicineRScienceQENPLoS ONE, Vol 7, Iss 12, p e52231 (2012)
institution DOAJ
collection DOAJ
language EN
topic Medicine
R
Science
Q
spellingShingle Medicine
R
Science
Q
María F Montenegro
Magali Sáez-Ayala
Antonio Piñero-Madrona
Juan Cabezas-Herrera
Juan Cabezas-Herrera
José Neptuno Rodríguez-López
Reactivation of the tumour suppressor RASSF1A in breast cancer by simultaneous targeting of DNA and E2F1 methylation.
description <h4>Background</h4>Tumour suppressor genes are often transcriptionally silenced by promoter hypermethylation, and recent research has implicated alterations in chromatin structure as the mechanistic basis for this repression. In addition to DNA methylation, other epigenetic post-translational modifications that modulate the stability and binding of specific transcription factors to gene promoters have emerged as important mechanisms for controlling gene expression. The aim of this study was to analyse the implications of these mechanisms and their molecular connections in the reactivation of RASSF1A in breast cancer.<h4>Methods</h4>Compounds that modulate the intracellular concentration of adenosine, such as dipyridamole (DIPY), greatly increase the antiproliferative effects of 3-O-(3,4,5-trimethoxybenzoyl)-(-)-catechin (TMCG), a synthetic antifolate derived from the structure of tea catechins. Quantitative real-time PCR arrays and MALDI-TOF mass spectrometry indicated that this combination (TMCG/DIPY) induced apoptosis in breast cancer cells by modulating the methylation levels of DNA and proteins (such as E2F1), respectively. Chromatin immunoprecipitation (ChIP) assays were employed to confirm that this combination induced chromatin remodelling of the RASSF1A promoter and increased the occupancy of E2F1 at the promoter of this tumour suppressor gene.<h4>Results</h4>The TMCG/DIPY combination acted as an epigenetic treatment that reactivated RASSF1A expression and induced apoptosis in breast cancer cells. In addition to modulating DNA methylation and chromatin remodelling, this combination also induced demethylation of the E2F1 transcription factor. The ChIP assay showed enhancement of E2F1 occupancy at the unmethylated RASSF1A promoter after TMCG/DIPY treatment. Interestingly, inhibition of E2F1 demethylation using an irreversible inhibitor of lysine-specific demethylase 1 reduced both TMCG/DIPY-mediated RASSF1A expression and apoptosis in MDA-MB-231 cells, suggesting that DNA and protein demethylation may act together to control these molecular and cellular processes.<h4>Conclusions/significance</h4>This study demonstrates that simultaneous targeting of DNA and E2F1 methylation is an effective epigenetic treatment that reactivates RASSF1A expression and induces apoptosis in breast cancer cells.
format article
author María F Montenegro
Magali Sáez-Ayala
Antonio Piñero-Madrona
Juan Cabezas-Herrera
Juan Cabezas-Herrera
José Neptuno Rodríguez-López
author_facet María F Montenegro
Magali Sáez-Ayala
Antonio Piñero-Madrona
Juan Cabezas-Herrera
Juan Cabezas-Herrera
José Neptuno Rodríguez-López
author_sort María F Montenegro
title Reactivation of the tumour suppressor RASSF1A in breast cancer by simultaneous targeting of DNA and E2F1 methylation.
title_short Reactivation of the tumour suppressor RASSF1A in breast cancer by simultaneous targeting of DNA and E2F1 methylation.
title_full Reactivation of the tumour suppressor RASSF1A in breast cancer by simultaneous targeting of DNA and E2F1 methylation.
title_fullStr Reactivation of the tumour suppressor RASSF1A in breast cancer by simultaneous targeting of DNA and E2F1 methylation.
title_full_unstemmed Reactivation of the tumour suppressor RASSF1A in breast cancer by simultaneous targeting of DNA and E2F1 methylation.
title_sort reactivation of the tumour suppressor rassf1a in breast cancer by simultaneous targeting of dna and e2f1 methylation.
publisher Public Library of Science (PLoS)
publishDate 2012
url https://doaj.org/article/17eabdbfda8d4ce0be8e713fa74c05ba
work_keys_str_mv AT mariafmontenegro reactivationofthetumoursuppressorrassf1ainbreastcancerbysimultaneoustargetingofdnaande2f1methylation
AT magalisaezayala reactivationofthetumoursuppressorrassf1ainbreastcancerbysimultaneoustargetingofdnaande2f1methylation
AT antoniopineromadrona reactivationofthetumoursuppressorrassf1ainbreastcancerbysimultaneoustargetingofdnaande2f1methylation
AT juancabezasherrera reactivationofthetumoursuppressorrassf1ainbreastcancerbysimultaneoustargetingofdnaande2f1methylation
AT juancabezasherrera reactivationofthetumoursuppressorrassf1ainbreastcancerbysimultaneoustargetingofdnaande2f1methylation
AT joseneptunorodriguezlopez reactivationofthetumoursuppressorrassf1ainbreastcancerbysimultaneoustargetingofdnaande2f1methylation
_version_ 1718422213987663872