Cargando…

Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression

E2 attenuates inflammatory responses by suppressing expression of pro-inflammatory genes. Given that inflammation is increasingly being associated with neurodegenerative and psychiatric processes, we sought to elucidate mechanisms by which E2 down-regulates a component of an inflammatory response, c...

Descripción completa

Detalles Bibliográficos
Autores principales: Stacey, Winfred, Bhave, Shreyas, Uht, Rosalie M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5010190/
https://www.ncbi.nlm.nih.gov/pubmed/27588681
http://dx.doi.org/10.1371/journal.pone.0161430
_version_ 1782451643924611072
author Stacey, Winfred
Bhave, Shreyas
Uht, Rosalie M.
author_facet Stacey, Winfred
Bhave, Shreyas
Uht, Rosalie M.
author_sort Stacey, Winfred
collection PubMed
description E2 attenuates inflammatory responses by suppressing expression of pro-inflammatory genes. Given that inflammation is increasingly being associated with neurodegenerative and psychiatric processes, we sought to elucidate mechanisms by which E2 down-regulates a component of an inflammatory response, cyclooxygenase– 2 (COX-2) expression. Although inflammatory processes in the brain are usually associated with microglia and astrocytes, we found that the COX-2 gene (cox-2) was expressed in a neuronal context, specifically in an amygdalar cell line (AR-5). Given that COX-2 has been reported to be in neurons in the brain, and that the amygdala is a site involved in neurodegenerative and neuropsychiatric processes, we investigated mechanisms by which E2 could down-regulate cox-2 expression in the AR-5 line. These cells express estrogen receptors alpha (ERα) and beta (ERβ), and as shown here cox-2. At the level of RNA, E2 and the ERβ selective ligand diarylpropionitrile (DPN) both attenuated gene expression, whereas the ERα selective ligand propyl pyrazole triol (PPT) had no effect. Neither ligand increased ERβ at the cox-2 promoter. Rather, DPN decreased promoter occupancy of NF-κB p65 and histone 4 (H4) acetylation. Treatment with the non-specific HDAC inhibitor Trichostatin A (TSA) counteracted DPN’s repressive effects on cox-2 expression. In keeping with the TSA effect, E2 and DPN increased histone deacetylase one (HDAC1) and switch-independent 3A (Sin3A) promoter occupancy. Lastly, even though E2 increased CpG methylation, DPN did not. Taken together, the pharmacological data indicate that ERβ contributes to neuronal cox-2 expression, as measured by RNA levels. Furthermore, ER ligands lead to increased recruitment of HDAC1, Sin3A and a concomitant reduction of p65 occupancy and Ac-H4 levels. None of the events, however, are associated with a significant recruitment of ERβ at the promoter. Thus, ERβ directs recruitment to the cox-2 promoter, but does so in the absence of being recruited itself.
format Online
Article
Text
id pubmed-5010190
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-50101902016-09-27 Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression Stacey, Winfred Bhave, Shreyas Uht, Rosalie M. PLoS One Research Article E2 attenuates inflammatory responses by suppressing expression of pro-inflammatory genes. Given that inflammation is increasingly being associated with neurodegenerative and psychiatric processes, we sought to elucidate mechanisms by which E2 down-regulates a component of an inflammatory response, cyclooxygenase– 2 (COX-2) expression. Although inflammatory processes in the brain are usually associated with microglia and astrocytes, we found that the COX-2 gene (cox-2) was expressed in a neuronal context, specifically in an amygdalar cell line (AR-5). Given that COX-2 has been reported to be in neurons in the brain, and that the amygdala is a site involved in neurodegenerative and neuropsychiatric processes, we investigated mechanisms by which E2 could down-regulate cox-2 expression in the AR-5 line. These cells express estrogen receptors alpha (ERα) and beta (ERβ), and as shown here cox-2. At the level of RNA, E2 and the ERβ selective ligand diarylpropionitrile (DPN) both attenuated gene expression, whereas the ERα selective ligand propyl pyrazole triol (PPT) had no effect. Neither ligand increased ERβ at the cox-2 promoter. Rather, DPN decreased promoter occupancy of NF-κB p65 and histone 4 (H4) acetylation. Treatment with the non-specific HDAC inhibitor Trichostatin A (TSA) counteracted DPN’s repressive effects on cox-2 expression. In keeping with the TSA effect, E2 and DPN increased histone deacetylase one (HDAC1) and switch-independent 3A (Sin3A) promoter occupancy. Lastly, even though E2 increased CpG methylation, DPN did not. Taken together, the pharmacological data indicate that ERβ contributes to neuronal cox-2 expression, as measured by RNA levels. Furthermore, ER ligands lead to increased recruitment of HDAC1, Sin3A and a concomitant reduction of p65 occupancy and Ac-H4 levels. None of the events, however, are associated with a significant recruitment of ERβ at the promoter. Thus, ERβ directs recruitment to the cox-2 promoter, but does so in the absence of being recruited itself. Public Library of Science 2016-09-02 /pmc/articles/PMC5010190/ /pubmed/27588681 http://dx.doi.org/10.1371/journal.pone.0161430 Text en © 2016 Stacey et al http://creativecommons.org/licenses/by/4.0/ This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Stacey, Winfred
Bhave, Shreyas
Uht, Rosalie M.
Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression
title Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression
title_full Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression
title_fullStr Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression
title_full_unstemmed Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression
title_short Mechanisms by Which 17β-Estradiol (E2) Suppress Neuronal cox-2 Gene Expression
title_sort mechanisms by which 17β-estradiol (e2) suppress neuronal cox-2 gene expression
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5010190/
https://www.ncbi.nlm.nih.gov/pubmed/27588681
http://dx.doi.org/10.1371/journal.pone.0161430
work_keys_str_mv AT staceywinfred mechanismsbywhich17bestradiole2suppressneuronalcox2geneexpression
AT bhaveshreyas mechanismsbywhich17bestradiole2suppressneuronalcox2geneexpression
AT uhtrosaliem mechanismsbywhich17bestradiole2suppressneuronalcox2geneexpression