Faculty Opinions recommendation of Membrane-localized estrogen receptor α is required for normal organ development and function.

Author(s):  
Ferdinando Auricchio ◽  
Gabriella Castoria
2014 ◽  
Vol 29 (4) ◽  
pp. 482-490 ◽  
Author(s):  
Ali Pedram ◽  
Mahnaz Razandi ◽  
Michael Lewis ◽  
Stephen Hammes ◽  
Ellis R. Levin

2014 ◽  
Vol 21 (4) ◽  
pp. 555-565 ◽  
Author(s):  
Yi-Lin Chang ◽  
Yu-Kan Hsu ◽  
Tsung-Fan Wu ◽  
Chieh-Ming Huang ◽  
Li-Yin Liou ◽  
...  

Estrogen receptor α (ERA) is a DNA-binding transcription factor that plays an important role in the regulation of cell growth. Previous studies indicated that the expression of ERα in cell lines and tumors derived from oral squamous cell carcinoma (OSCC). The aim of this study was to examine the activity and function of ERα in OSCC cells and the mechanism underlying ERα activation. Immunochemical analyses in benign (n=11) and malignant (n=21) lesions of the oral cavity showed that ERα immunoreactivity was observed in 43% (9/21) of malignant lesions, whereas none of benign lesions showed ERα immunoreactivity. The ERα expression was also found in three OSCC cell lines and its transcriptional activity was correlated with cell growth. Addition of estradiol stimulated cell growth, whereas treatment of tamoxifen or knockdown of ERα expression caused reduced cell growth. Interestingly, the expression and activity of focal adhesion kinase (FAK) were associated with the phosphorylation of ERα at serine 118 in OSCC cells. Elevated expression of FAK in the slow-growing SCC25 cells caused increases in ERα phosphorylation, transcriptional activity, and cell growth rate, whereas knockdown of FAK expression in the rapid-growing OECM-1 cells led to reduced ERα phosphorylation and activity and retarded cell growth. Inhibition of the activity of protein kinase B (AKT), but not ERK, abolished FAK-promoted ERα phosphorylation. These results suggest that OSCC cells expressed functional ERα, whose activity can be enhanced by FAK/AKT signaling, and this was critical for promoting cell growth. Thus, FAK and ERα can serve as the therapeutic targets for the treatment of OSCC.


Endocrinology ◽  
2009 ◽  
Vol 150 (11) ◽  
pp. 5085-5093 ◽  
Author(s):  
So-Youn Kim ◽  
Jeffrey Weiss ◽  
Minghan Tong ◽  
Monica M. Laronda ◽  
Eun-Jig Lee ◽  
...  

Foxl2 is a forkhead transcription factor required for ovary development and ovarian follicle maturation. In this report, we identified and characterized a functional relationship between Foxl2 expression and estrogen receptor (ER)-α signaling. We show that Foxl2 has no effect on classical ERα-mediated transcription, which occurs through canonical estrogen response elements. However, Foxl2 suppresses ERα signaling through nonclassical tethered transcriptional pathways. Specifically, the selective ER modulator tamoxifen stimulates activator protein-1 (AP1)-dependent transcription via the ERα, and this enhancement is blocked by Foxl2. Two lines of evidence suggest that Foxl2 suppression is mediated by physical interactions with ERα rather than direct action at AP1 binding sites. First, ERα is coimmunoprecipitated with Foxl2. Second, activation of a upstream activating sequence (UAS) reporter by Gal4-cJun in the presence of ERα and tamoxifen was blocked by Foxl2, demonstrating suppression in the absence of an AP1 site. Cyclooxygenase-2 (COX2), which is required for ovulation, was identified through expression profiling as a candidate physiological target for nonclassical ERα signaling and thus modulation by ERα/Foxl2 interactions. This possibility was confirmed by two sets of experiments. COX2 protein levels were induced by ERα in the presence of tamoxifen, and protein expression was suppressed by Foxl2. In addition, ERα stimulation of the COX2 promoter was repressed by Foxl2. We conclude that ERα and Foxl2 interact and that Foxl2 selectively suppresses ERα-mediated transcription of AP1-regulated genes. These data provide a potential point of convergence for ERα and Foxl2 to regulate ovarian development and function.


2005 ◽  
Vol 94 (4) ◽  
pp. 347-359 ◽  
Author(s):  
Ioanna Laïos ◽  
Fabrice Journé ◽  
Denis Nonclercq ◽  
Doris Salazar Vidal ◽  
Robert-Alain Toillon ◽  
...  

2014 ◽  
Vol 74 (19) ◽  
pp. 5469-5479 ◽  
Author(s):  
Meritxell Rosell ◽  
Ekaterina Nevedomskaya ◽  
Suzan Stelloo ◽  
Jaya Nautiyal ◽  
Ariel Poliandri ◽  
...  

2006 ◽  
Vol 4 (1) ◽  
pp. nrs.04007 ◽  
Author(s):  
Mariam H. Al-Dhaheri ◽  
Brian G. Rowan

An understanding of posttranslational events in nuclear receptor signaling is crucial for drug design and clinical therapeutic strategies. Phosphorylation is a well-characterized posttranslational modification that regulates subcellular localization and function of nuclear receptors and coregulators. Although the role of single phosphorylation sites in nuclear receptor function has been described, the contribution of combinations of multiple phosphorylation sites to receptor function remains unclear. The development of phosphoantibodies to each phosphorylation site in a nuclear receptor is a powerful tool to address the role of phosphorylation in multiply phosphorylated receptors. However, phosphoantibodies must be rigorously validated prior to use. This review describes the general methodology for design, characterization and validation of phosphoantibodies using the example of eight phosphoantibodies raised against phosphorylation sites in estrogen receptor α (ERα).


Sign in / Sign up

Export Citation Format

Share Document