Differential crosstalk between estrogen receptor (ER)α and ERβ and the thyroid hormone receptor isoforms results in flexible regulation of the consensus ERE

2001 ◽  
Vol 95 (1-2) ◽  
pp. 9-17 ◽  
Author(s):  
Nandini Vasudevan ◽  
Noriyuki Koibuchi ◽  
William W Chin ◽  
Donald W Pfaff
2001 ◽  
Vol 67 (1) ◽  
pp. 106-113 ◽  
Author(s):  
Fabienne Bury ◽  
Jean-Luc Carré ◽  
Sonia Vega ◽  
M. Said Ghandour ◽  
Angeles Rodriguez-Peña ◽  
...  

Mitochondrion ◽  
2006 ◽  
Vol 6 (3) ◽  
pp. 143-148 ◽  
Author(s):  
Fionnuala Morrish ◽  
Norman E. Buroker ◽  
Ming Ge ◽  
Xue-Han Ning ◽  
Jesus Lopez-Guisa ◽  
...  

2004 ◽  
Vol 378 (2) ◽  
pp. 549-557 ◽  
Author(s):  
Keiko NAKANO ◽  
Akio MATSUSHITA ◽  
Shigekazu SASAKI ◽  
Hiroko MISAWA ◽  
Kozo NISHIYAMA ◽  
...  

The molecular mechanism involved in the liganded thyroid hormone receptor suppression of the TSHβ (thyroid-stimulating hormone β, or thyrotropin β) gene transcription is undetermined. One of the main reasons is the limitation of useful cell lines for the experiments. We have developed an assay system using non-pituitary CV1 cells and studied the negative regulation of the TSHβ gene. In CV1 cells, the TSHβ–CAT (chloramphenicol acetyltransferase) reporter was stimulated by Pit1 and GATA2 and suppressed by T3 (3,3´,5-tri-iodothyronine)-bound thyroid hormone receptor. The suppression was dependent on the amounts of T3 and the receptor. Unliganded receptor did not stimulate TSHβ activity, suggesting that the receptor itself is not an activator. Analyses using various receptor mutants revealed that the intact DNA-binding domain is crucial to the TSHβ gene suppression. Co-activators and co-repressors are not necessarily essential, but are required for the full suppression of the TSHβ gene. Among the three receptor isoforms, β2 exhibited the strongest inhibition and its protein level was the most predominant in a thyrotroph cell line, TαT1, in Western blotting. The dominant-negative effects of various receptor mutants measured on the TSHβ–CAT reporter were not simple mirror images of those in the positive regulation under physiological T3 concentration.


Author(s):  
Jean-Luc Carr� ◽  
Corinne Demerens ◽  
Angeles Rodr�guez-Pe�a ◽  
Herv� H. Floch ◽  
Guy Vincendon ◽  
...  

2008 ◽  
Vol 8 (1) ◽  
pp. 8 ◽  
Author(s):  
Lucas Bleicher ◽  
Ricardo Aparicio ◽  
Fabio M Nunes ◽  
Leandro Martinez ◽  
Sandra M Gomes Dias ◽  
...  

2003 ◽  
Vol 203 (1-2) ◽  
pp. 65-75 ◽  
Author(s):  
Peter M. Sadow ◽  
Olivier Chassande ◽  
Eugene K. Koo ◽  
Karine Gauthier ◽  
Jacques Samarut ◽  
...  

2007 ◽  
Vol 27 (17) ◽  
pp. 5933-5948 ◽  
Author(s):  
Theresa J. Peterson ◽  
Sudipan Karmakar ◽  
Margaret C. Pace ◽  
Tong Gao ◽  
Carolyn L. Smith

ABSTRACT Multiple factors influence estrogen receptor α (ERα) transcriptional activity. Current models suggest that the silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) corepressor functions within a histone deactylase-containing protein complex that binds to antiestrogen-bound ERα and contributes to negative regulation of gene expression. In this report, we demonstrate that SMRT is required for full agonist-dependent ERα activation. Chromatin immunoprecipitation assays demonstrate that SMRT, like ERα and the SRC-3 coactivator, is recruited to an estrogen-responsive promoter in estrogen-treated MCF-7 cells. Depletion of SMRT, but not histone deacetylases 1 or 3, negatively impacts estradiol-stimulated ERα transcriptional activity, while exogenous expression of SMRT's receptor interaction domains blocks ERα activity, indicating a functional interaction between this corepressor and agonist-bound ERα. Stimulation of estradiol-induced ERα activity by SMRT overexpression occurred in HeLa and MCF-7 cells, but not HepG2 cells, indicating that these positive effects are cell type specific. Similarly, the ability of SMRT depletion to promote the agonist activity of tamoxifen was observed for HeLa but not MCF-7 cells. Furthermore, impairment of agonist-stimulated activity by SMRT depletion is specific to ERα and not observed for receptors for vitamin D, androgen, or thyroid hormone. Nuclear receptor corepressor (N-CoR) depletion increased the transcriptional activity of all four tested receptors. SMRT is required for full expression of the ERα target genes cyclin D1, BCL-2, and progesterone receptor but not pS2, and its depletion significantly attenuated estrogen-dependent proliferation of MCF-7 cells. Taken together, these data indicate that SMRT, in conjunction with gene-specific and cell-dependent factors, is required for positively regulating agonist-dependent ERα transcriptional activity.


Sign in / Sign up

Export Citation Format

Share Document