Signal Transducers and Activators of Transcription as Downstream Targets of Nongenomic Estrogen Receptor Actions
Linda Björnström and
Maria Sjöberg
Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
Address all correspondence and requests for reprints to: Dr. Maria Sjöberg, Department of Cell and Molecular Biology, Karolinska Institute, S-171 Stockholm, Sweden. E-mail: maria.sjoberg{at}cmb.ki.se.
 |
ABSTRACT
|
---|
17ß-Estradiol-activated estrogen receptor
(ER
) and ß (ERß) are able to induce transcriptional activation of signal transducer and activator of transcription (Stat)-regulated promoters via cytoplasmic signal transduction pathways. Stat5 and Stat3 are required for promoter induction, which correlates with cytoplasmic sublocalization of ERs and is independent of intact coactivator binding sites and DNA-binding domains. In endothelial cells, Stat5 and Stat3 are rapidly phosphorylated on both tyrosine and serine residues in response to 17ß-estradiol, and nuclear translocation is subsequently induced. 17ß-Estradiol-induced transactivation of a Stat-regulated promoter requires at least three different signal transduction pathways, including MAPK, Src-kinase, and phosphatidylinositol-3-kinase activities. In conclusion, this work identifies a novel pathway involving an agonist-bound ER-activated phosphorylation cascade, resulting in nuclear transcriptional activation of target transcription factors. These findings reveal novel targets for the development of drugs that modulate a nongenomic-to-genomic ER-dependent mechanism.
 |
INTRODUCTION
|
---|
ESTROGENS ARE POWERFUL mitogens that promote growth and proliferation in many target organs. Their effects are mediated by two related nuclear hormone receptors, estrogen receptor
(ER
) and ß (ERß). These receptors belong to a large superfamily of nuclear hormone receptors that share a well conserved DNA-binding domain (DBD), a structurally conserved ligand binding domain (LBD), and an N-terminal domain with no homology between the different receptors (1, 2, 3). The classical mechanism of activation depends on ligand binding, after which the receptor dimerizes and binds to estrogen response elements (EREs) located within the promoters of estrogen-responsive genes. Ligand binding also induces a conformational change in the LBD of the receptor, which allows the recruitment of coactivator proteins (4, 5).
Estrogens also have very rapid effects in a variety of tissues including bone, brain, mammary gland, and vasculature. For example, estrogens have been shown to stimulate the activities of phospholipase C and adenylate cyclase, resulting in increased production of inositol lipid and cAMP, respectively (6, 7). In addition, they increase intracellular [Ca2+] and activate MAPK (8, 9, 10, 11, 12, 13) and phosphatidylinositol-3-kinase (PI3-kinase) signaling pathways (14, 15, 16, 17). Although it is not fully defined which receptors are responsible for mediating these rapid nongenomic effects of estrogens, some studies point to the existence of a separate membrane receptor unrelated to the classical nuclear receptors, while others implicate a subgroup of the classical ERs that is associated with cellular membranes (for review see Ref. 18).
Stat (signal transducers and activators of transcription) proteins are latent cytoplasmic transcription factors that are activated in response to a large number of cytokines, growth factors, and hormones. The binding of ligand to its cognate cytokine receptor induces activation of receptor-associated members of the Janus kinase (Jak) family, which in turn phosphorylate a conserved tyrosine residue in the C-terminal region of Stats. The tyrosine-phosphorylated Stats undergo dimerization and translocation to the nucleus where they regulate transcription by binding to specific DNA sequences (19). In addition to basic tyrosine phosphorylation, the transcriptional activity of Stats can be regulated by serine phosphorylation of the transactivation domain (20).
Due to the fact that Stats are activated through phosphorylation and because 17ß-estradiol is known to rapidly induce phosphorylation cascades in the cytoplasm, we were interested in investigating the possibility of Stats as downstream targets of nongenomic ER actions. In the present paper, we show that 17ß-estradiol-bound ERs activate cytoplasmic signal transduction pathways in a manner that is independent of receptor-DNA binding, resulting in activation of Stat-regulated promoters via Stat5 and Stat3 proteins. We show that tyrosine and serine phosphorylation of Stat5 and Stat3 are rapidly induced by 17ß-estradiol and that intact MAPK, PI3-kinase, and Src-kinase signaling pathways are required for 17ß-estradiol-induced activation of a Stat-regulated promoter. In summary, the mechanism described in this paper comprises a chain of reactions starting with activation of a cytoplasmic ER by 17ß-estradiol and resulting in nuclear transcriptional activation of target transcription factors, thus representing a novel pathway of nongenomic-to-genomic actions of ERs.
 |
RESULTS
|
---|
17ß-Estradiol Induces Both Tyrosine and Serine Phosphorylation of Stat5 and Activation of the ß-Casein Promoter
17ß-Estradiol is known to rapidly induce cytoplasmic signal transduction pathways, resulting in the phosphorylation of target proteins. This so-called nongenomic action of 17ß-estradiol has been described in various cell types, such as endothelial cells (16). Porcine aortic endothelial (PAE) cells were found to express both endogenous Stat5 and ER
and -ß proteins (data not shown), and we decided to analyze how 17ß-estradiol treatment would affect the phosphorylation status of Stat5 in these cells. Tyrosine and serine phosphorylation of Stat5 upon 50 nM 17ß-estradiol or 15% serum [fetal bovine serum (FBS)] treatment at various time points were analyzed by Western blot analyses of whole-cell extracts, using phospho-specific antibodies against tyrosine-694/699 and serine-725/731 of Stat5a/b, respectively. As shown in Fig. 1A
, both tyrosine and serine phosphorylation of Stat5 were rapidly induced upon 17ß-estradiol treatment and were inhibited in the presence of ICI 182,780, suggesting a requirement for an agonist-bound ER. Total levels of Stat5a and Stat5b proteins in the cell extracts are shown. An induced gel mobility shift upon 17ß-estradiol treatment is observed when immunoblotting with the Stat5b antibody (Fig. 1A
, lower panel). This shift is known to be due to tyrosine phosphorylation of Stat5b, which was confirmed by the recognition of the upper band by the phospho-specific antibody.

View larger version (22K):
[in this window]
[in a new window]
|
Figure 1. 17ß-Estradiol Induces Phosphorylation of Stat5 and Activation of the ß-Casein Reporter Gene
A, Whole-cell extracts prepared from PAE cells, treated for various time points with either no hormone (NH), 50 nM 17ß-estradiol (E2), or 15% FBS, as indicated, were analyzed by Western blot analyses using phospho-specific antibodies against tyrosine-694/699 and serine-725/731 of Stat5a/b. Where indicated, 10-7 M ICI 182,780 (ICI) was added together with hormone. Total Stat5a and Stat5b proteins were detected using polyclonal Stat5a and Stat5b antibodies. The arrow indicates the Stat5b supershift, which corresponds to its tyrosine phosphorylation. B, PAE cells were left untreated or treated with 50 nM 17ß-estradiol (E2) for 30 or 60 min, or 15% FBS for 30 min. Nuclear extracts were prepared and analyzed by Western blot with a polyclonal Stat5 antibody (upper section) or mixed with a biotinylated DNA-binding element (wt) derived from the ß-casein promoter (lower section) as described in Materials and Methods. A mutated DNA binding element was also included (mut). DNA-bound Stat5 was analyzed by Western blot using a polyclonal Stat5 antibody. C, PAE cells were transfected with 1 µg ß-casein-luciferase reporter gene. Cells were treated with either no hormone (NH), 10-8 M 17ß-estradiol (E2), or 15% FBS, as indicated, and the reporter activity was analyzed 24 h thereafter. The luciferase activity was normalized using ß-galactosidase as an internal control. Data are representative of at least three independent experiments performed in duplicate. Mean ± SD are shown. D, COS-7 cells were transfected with 1 µg ß-casein-luciferase reporter gene together with 200 ng ER wt, 10 ng Stat5a, 500 ng Jak2 KN, and 50 ng PRL receptor (Prl-R), as indicated. Cells were treated with either no hormone (NH), 10-8 M 17ß-estradiol (E2), 15% FBS, or 5 µg/ml PRL, as indicated, and further assayed as in panel C.
|
|
Because tyrosine phosphorylation of Stat5 is a prerequisite for its nuclear translocation, we analyzed whether 17ß-estradiol could affect the subcellular localization of Stat5. Nuclear extracts prepared from untreated or 17ß-estradiol-stimulated PAE cells were analyzed by Western blot using a Stat5 antibody. As shown in Fig. 1B
(upper section), there was an increased amount of Stat5 present in the nucleus when the cells had been exposed to 17ß-estradiol for 30 min, indicating that the steroid is able to promote translocation of Stat5 to the nucleus. However, the amount of Stat5 in the nucleus was almost back to basal level after 60 min, demonstrating that the steroid-induced translocation of Stat5 is a rapid and transient process. To investigate whether DNA-binding activity of Stat5 is induced upon 17ß-estradiol stimulation, an in vitro DNA-binding assay using a biotinylated Stat5 DNA-binding site was performed. As shown in Fig. 1B
(lower section), in extracts prepared from cell nuclei of PAE cells treated with 17ß-estradiol or FBS for 30 min, binding of Stat5 to the consensus
-interferon-activated sequence (GAS) element present in the ß-casein promoter was enhanced compared with untreated cells, whereas no interaction with a mutated DNA-binding site was observed.
To investigate whether the observed rapid phosphorylation of Stat5 in response to 17ß-estradiol correlated with a downstream event, we analyzed activation of a Stat5-regulated promoter after 17ß-estradiol treatment of PAE cells. As shown in Fig. 1C
, activation of the ß-casein reporter gene was induced 2-fold in response to 17ß-estradiol compared with untreated cells, while FBS treatment resulted in a 2.5-fold induction.
In ER-negative COS-7 cells transiently transfected with the ß-casein reporter gene, no induction response was observed upon treatment with 17ß-estradiol alone. However, 17ß-estradiol induced promoter activity in these cells after cotransfection with ER
expression plasmid (Fig. 1D
). Stat5 is primarily activated by prolactin (PRL) and GH, whereupon tyrosine phosphorylation is induced by the Jak2 kinase. We therefore analyzed the requirement of Jak2 activity for 17ß-estradiol-induced activation of the ß-casein reporter gene. As shown in Fig. 1B
, a dominant negative version of Jak2 (Jak2 KN) transfected into the cells did not interfere with 17ß-estradiol-induced reporter activity in the presence of ER
, nor was the presence of the PRL receptor required since it is not expressed endogenously in these cells. However, as expected, Jak2 KN prevented activation of overexpressed Stat5 in response to PRL. Taken together, these results demonstrate that 17ß-estradiol rapidly induces phosphorylation, nuclear translocation, and DNA-binding activity of Stat5. Furthermore, these data show that an agonist-bound ER is able to induce ß-casein promoter activity through activation of the Stat5 transcription factor, independently of the PRL receptor and the Jak2 kinase.
Cytoplasmic Localization of ER
and ß Is Favored for Activation of the ß-Casein Promoter in Response to 17ß-Estradiol
The involvement of nonnuclear ERs in mediating some of the described nongenomic actions of estrogens has been suggested (12, 21). To investigate whether the cellular localization of ER affected 17ß-estradiol-induced activation of the ß-casein promoter, an ER
mutant with a deletion of important nuclear localization signals in the hinge domain (NLSA) was used (22). As shown in Fig. 2B
, while the wild-type receptor showed nuclear staining, NLSA was predominantly present in the cytoplasm of cells. Interestingly, NLSA transiently transfected into COS-7 cells efficiently induced activation of the ß-casein reporter gene in response to 17ß-estradiol, with an 11-fold induction of the reporter observed while the wild-type receptor showed a 4-fold induction under the same conditions (Fig. 2C
). To analyze the ability of ERß to exhibit the same effect on the ß-casein promoter if rendered cytoplasmic, a tag containing a fatty acylation site was incorporated in-frame upstream of the N terminus of ERß (ERß Mem). This site can be myristoylated and palmitoylated with subsequent membrane localization of the protein (23). As shown in Fig. 2B
, ERß Mem localized to extranuclear sites and, interestingly, was more efficient in activating the ß-casein reporter gene compared with wild-type ERß (Fig. 2C
). Taken together, these results demonstrate that cytoplasmic localization of ER
and -ß is favored for activation of the ß-casein promoter in response to 17ß-estradiol.

View larger version (25K):
[in this window]
[in a new window]
|
Figure 2. Cytoplasmic Localization of ER and -ß Correlates with Efficient Activation of the ß-Casein Reporter Gene in Response to 17ß-Estradiol
A, Schematic representation of ER wt, NLSA, deleted in the hinge domain ( 245307), ERß wt, and ERß Mem. ERß Mem has a fatty acylation site tagged to the N terminus. B, COS-7 cells were transfected with expression vectors for either ER wt, NLSA, ERß wt, or ERß Mem. The subcellular localization of the receptor proteins was analyzed by indirect immunofluorescence as described in Materials and Methods. Cell nuclei are stained with 4,6-diamidino-2-phenylindole. C, COS-7 cells were transfected with 1 µg ß-casein-luciferase reporter gene together with either 200 ng ER wt, NLSA, ERß wt, or ERß Mem. Cells were treated with either no hormone (NH) or 10-8 M 17ß-estradiol (E2), and the reporter activity was analyzed 24 h thereafter. The luciferase activity was normalized using ß-galactosidase as an internal control. Data are representative of at least three independent experiments performed in duplicate. Mean ± SD are shown.
|
|
Stat5 Is Not the Only Target Transcription Factor Activated by 17ß-Estradiol on the ß-Casein Promoter
The ß-casein promoter (-333 to -1) region present in the luciferase reporter construct includes binding sites for several nuclear factors (24). To confirm that Stat5 is the target transcription factor responsible for promoter induction, a consensus Stat5 reporter construct, comprising a six-repeat sequence of the lactogenic hormone response element (LHRE) (25), was used. The LHRE reporter gene was efficiently induced in the presence of 17ß-estradiol in COS-7 cells transiently transfected with NLSA (Fig. 3A
), confirming that endogenous Stat5 is targeted by the 17ß-estradiol-induced pathway. Furthermore, overexpression of a dominant-negative version of Stat5 (MGF
750), which lacks the transactivation domain (26), inhibited 17ß-estradiol-induced activation of the ß-casein reporter gene in a dose-dependent manner (Fig. 3B
). To analyze whether Stat5 is the only target transcription factor induced by 17ß-estradiol on the promoter, the transfection experiments were repeated in mouse fibroblast cells devoid of functional Stat5a and Stat5b (Stat5ab-/- cells) (a gift from J. Ihle). In the presence of NLSA, 17ß-estradiol induced activation of the ß-casein reporter gene in these cells, indicating that Stat5 is not the only transcription factor to be activated. However, overexpression of Stat5 in cells lacking Stat5 proteins markedly increased the 17ß-estradiol-induced response (Fig. 3C
), demonstrating that Stat5 can indeed be induced to activate the reporter in the presence of both NLSA and 17ß-estradiol.

View larger version (19K):
[in this window]
[in a new window]
|
Figure 3. More than One Stat Is Targeted by an Agonist-Bound ER
COS-7 cells were transfected with 1 µg LHRE-tk-luc reporter gene together with 200 ng NLSA (panel A) or 1 µg ß-casein reporter gene together with 200 ng NLSA and MGF 750, 50 ng, 200 ng, and 500 ng, as indicated (panel B). C, Stat5ab-/- cells were transfected with 1 µg ß-casein reporter gene together with 200 ng NLSA and Stat5a, 200 ng and 500 ng, as indicated. Cells were treated with either no hormone (NH) or 10-8 M 17ß-estradiol (E2), as indicated, and the reporter activity was analyzed 24 h thereafter. The luciferase activity was normalized using ß-galactosidase as an internal control. Data are representative of at least three independent experiments performed in duplicate. Mean ± SD are shown.
|
|
Stat3 Is Activated by the 17ß-Estradiol-Induced Pathway
The observation that Stat5 was not the only transcription factor responsible for activating the ß-casein promoter in response to 17ß-estradiol prompted us to test the involvement of other Stat family members with similar characteristics. In a DNA-binding assay, Stat3 was found to bind to the consensus
-interferon-activated sequence element present in the ß-casein promoter in vitro (data not shown) and, furthermore, Stat3 was shown to activate the ß-casein reporter gene in transient transfection experiments (data not shown and Ref. 27). The phosphorylation status of Stat3 upon 50 nM 17ß-estradiol or 15% serum (FBS) treatment was analyzed by Western blot analyses of PAE whole-cell extracts, using phospho-specific antibodies against tyrosine-705 and serine-727 of Stat3, respectively. As shown in Fig. 4A
, both tyrosine and serine phosphorylation of Stat3 were induced within 15 min of 17ß-estradiol treatment and were inhibited in the presence of ICI 182,780. Total levels of Stat3 protein in the cell extracts are shown in Fig. 4A
(lower panel).

View larger version (23K):
[in this window]
[in a new window]
|
Figure 4. Stat3 Is Targeted by the 17ß-Estradiol-Induced Pathway
A, Whole-cell extracts prepared from PAE cells, treated for 15 min with either no hormone (NH), 50 nM 17ß-estradiol (E2), in the absence or presence of 10-7 M ICI 182,780 (ICI), or 15% FBS, as indicated, were analyzed by Western blot analyses using phospho-specific antibodies against tyrosine-705 and serine-727 of Stat3. Total Stat3 protein was detected using a monoclonal Stat3 antibody. B, COS-7 cells were transfected with 1 µg of either pLucTKS3 or pLucCRP reporter gene together with 200 ng NLSA. Cells were treated with either no hormone (NH), 10-8 M 17ß-estradiol (E2), or 200 ng/ml EGF, as indicated, and the reporter activity was analyzed 24 h thereafter. The luciferase activity was normalized using ß-galactosidase as an internal control. Data are representative of at least three independent experiments performed in duplicate. Mean ± SD are shown. C, PAE cells were left untreated or treated with 50 nM 17ß-estradiol (E2) or 15% FBS for 30 min. Nuclear extracts were prepared and mixed with a biotinylated DNA-binding element derived from the CRP promoter as described in Materials and Methods. DNA-bound Stat3 was analyzed by Western blot using a monoclonal Stat3 antibody. D, Stat5-/- cells were transfected with 1 µg of ß-casein reporter gene together with 200 ng NLSA and Stat3ß, 50 ng, 200 ng, and 500 ng, as indicated. Cells were treated and assayed as described in panel B.
|
|
To determine whether the transcriptional activity of endogenous Stat3 is induced in response to 17ß-estradiol, two Stat3-specific reporter constructs were used. pLucTKS3 contains seven repeated Stat3-specific binding sites derived from the human C-reactive protein (CRP) gene, and pLucCRP contains the true CRP promoter fragment in front of the luciferase gene (28). COS-7 cells were transiently transfected with the pLucTKS3 or pLucCRP reporter gene and NLSA, and the cells were treated with either 17ß-estradiol or, as a control, with epidermal growth factor (EGF), which is known to induce the transcriptional activity of Stat3. As expected, both reporter genes were induced in the presence of EGF. However, in the presence of NLSA and 17ß-estradiol, activation of the two reporter genes was also induced (Fig. 4B
). Furthermore, in an in vitro DNA-binding assay, using the consensus Stat3-binding site present in the CRP promoter, binding of Stat3 to the element was observed in nuclear extracts from PAE cells treated with 17ß-estradiol or FBS for 30 min (Fig. 4C
).
To determine whether the residual ß-casein promoter activity observed in Stat5ab-/- cells is due to Stat3 activity, a dominant negative version of Stat3 (Stat3ß) was transiently cotransfected with NLSA and the ß-casein reporter gene into these cells. Stat3ß is a naturally occurring splice variant with a deletion in the C-terminal transactivation domain (29). As shown in Fig. 4D
, overexpression of Stat3ß efficiently suppressed 17ß-estradiol-induced activation of the reporter, demonstrating that the residual ß-casein promoter activity is due, at least in part, to activation of Stat3. However, the involvement of other Stat proteins cannot be excluded. Taken together, these results demonstrate that, in addition to Stat5, Stat3 is also activated via the pathway induced by 17ß-estradiol-bound ER.
Antiestrogens Block 17ß-Estradiol-Induced Activation of the ß-Casein Promoter
Next, the ability of antiestrogens to block activation of the ß-casein promoter by 17ß-estradiol-activated ER was analyzed. Both the partial ER antagonists, 4-hydroxy tamoxifen (OHT) and raloxifen, as well as the full antagonist ICI 182,780, completely blocked 17ß-estradiol-induced activation of the ß-casein reporter gene (Fig. 5
). OHT and ICI 182,780 are thought to cause ER antagonism via interaction with the LBD, implying that an intact LBD in the agonist-bound conformation is important. 17
-Estradiol, the transcriptionally inactive stereoisomer for 17ß-estradiol, was also tested, but did not induce the reporter. However, the ERß-specific agonist, genistein (30), was able to induce activation of the reporter at 100 nM in the presence of ERß wt (Fig. 5
). In conclusion, these data demonstrate that binding of the natural hormone to ER is essential to induce activation of the ß-casein promoter.

View larger version (9K):
[in this window]
[in a new window]
|
Figure 5. Binding of the Natural Hormone to ER Is Essential for Induction of the ß-Casein Reporter Gene
COS-7 cells were transfected with 1 µg ß-casein reporter gene together with either 200 ng NLSA or ERß wt. Cells were treated with either no hormone (NH), 10-8 M 17ß-estradiol (E2), E2 + 10-7 M OHT, E2 + 10-8 M raloxifen (RAL), E2 + 10-7 M ICI 182,780 (ICI), 10-8 M 17 -estradiol, or 100 nM genistein (Gen), as indicated, and the reporter activity was analyzed 24 h thereafter. The luciferase activity was normalized using ß-galactosidase as an internal control. Data are representative of at least three independent experiments performed in duplicate. Mean ± SD are shown.
|
|
17ß-Estradiol-Induced Activation of the ß-Casein Promoter Is Dependent on Intact Src-Kinase, MAPK, and PI3-Kinase Signaling Pathways
To investigate which signal transduction pathways are required for 17ß-estradiol-induced activation of the ß-casein promoter, COS-7 cells were transiently transfected with the ß-casein reporter gene and NLSA, and 17ß-estradiol-induced reporter activity was analyzed in the presence of various signaling inhibitors. An inhibitory effect was observed when the cells were pretreated with the MAPK kinase (MEK)1/2 inhibitor, UO126, and the two Src-kinase family inhibitors, the pyrazolopyrimidine compound PP1 (31) and genistein, which acts as a tyrosine kinase inhibitor at 100 µM (Ref. 32 and Fig. 6A
), showing that intact MAPK and Src-kinase signaling pathways are required. The involvement of Src-kinase was further demonstrated by transfection of embryonic fibroblast cells derived from Src-/- mice (SYF cells) (33). No, or minimal, activation of the reporter in response to 17ß-estradiol was observed in SYF cells transiently transfected with NLSA. However, coexpression of constitutively active Src-kinase (SrcY527F) restored induction of the reporter gene (Fig. 6B
). Activation of the reporter was also prevented by pretreatment of the cells with the PI3-kinase inhibitor, LY 294002 (Fig. 6A
), suggesting that not only MAPK and Src-kinase, but also PI3-kinase, is involved in the signaling pathway regulating 17ß-estradiol-induced ß-casein promoter activity. The involvement of the p38 MAPK and G protein-coupled receptors was also investigated, but, as shown in Fig. 4A
, activation of the reporter was sustained in the presence of the p38 MAPK inhibitor, SB 203580, and the G
-subunit inhibitor, pertussis toxin (Fig. 6A
). Notably, a similar result was obtained when 17ß-estradiol-induced activation of the consensus LHRE reporter gene was analyzed in the presence of the various signaling inhibitors (data not shown), which confirms that the Src-kinase, MAPK, and PI3-kinase signaling pathways are required for 17ß-estradiol-induced transactivation of Stat5 and excludes the possibility that the kinases themselves activate the ß-casein promoter in a Stat5-independent manner.

View larger version (22K):
[in this window]
[in a new window]
|
Figure 6. Intact MAPK, PI3-Kinase, and Src-Kinase Signaling Pathways Are Important for 17ß-Estradiol-Induced Activation of the ß-Casein Reporter Gene
A, COS-7 cells were transfected with 1 µg ß-casein reporter gene together with 200 ng NLSA. Cells were pretreated with 20 µM PP1, 100 µM genistein, 50 µM UO126, 50 µM LY 294002 or 20 µM SB 203580 for 1 h, or 100 ng/ml pertussis toxin (PT) for 16 h, before the addition of 10-8 M 17ß-estradiol (E2), and the reporter activity was analyzed 6 h thereafter. B, SYF cells were transfected with 1 µg ß-casein-luciferase reporter gene together with 200 ng NLSA and 500 ng SrcY527F, as indicated. Cells were treated with either no hormone (NH) or 10-8 M 17ß-estradiol (E2), and the reporter activity was analyzed 24 h thereafter. The luciferase activity was normalized using ß-galactosidase as an internal control. Data are representative of at least three independent experiments performed in duplicate. Mean and ± SD are shown. C, Whole-cell extracts from endothelial cells were treated for various time points with either no hormone (NH), 50 nM 17ß-estradiol (E2), or 15% FBS, and activated ERK1/2 was analyzed by Western blot using a MAPK p42/44 phospho-specific antibody (upper panel). Total ERK1/2 protein was detected with a control MAPK p42/44 antibody (lower panel). Where indicated, cells were pretreated with 1 µM ICI 182,780 for 45 min, 50 µM ERK1/2 inhibitor, UO126, or 20 µM tyrosine kinase inhibitor, PP1, for 1 h before the addition of hormone.
|
|
To determine whether the MAPK pathway is induced by 17ß-estradiol in PAE cells, whole-cell extracts were analyzed by Western blot using antibodies against activated ERK1/2. As shown in Fig. 6C
, ERK1/2 was indeed activated upon 17ß-estradiol treatment, but not in the presence of ICI 182,780, suggesting a requirement for an agonist-bound ER. Also shown in Fig. 6C
, activation of ERK1/2 was prevented both by the MEK1/2 inhibitor, UO126, as expected, and by the Src-kinase inhibitor, PP1, suggesting a role for Src-kinase upstream of MAPK in the signal transduction pathway induced by 17ß-estradiol. Total levels of ERK1/2 proteins in the cell extracts are shown in Fig. 6C
(lower panel). All together, these results demonstrate that the Src-kinase, MAPK, and PI3-kinase signaling pathways act downstream of 17ß-estradiol. In contrast, neither the p38 MAPK nor the G protein-mediated signal transduction pathways are involved in mediating 17ß-estradiol-induced ß-casein promoter activity.
Serine Phosphorylation of Stat5 Is Required for Optimal Transcriptional Activity in Response to 17ß-Estradiol
In addition to tyrosine phosphorylation, serine phosphorylation of Stat5 was also rapidly induced by 17ß-estradiol (Fig. 1A
). Together with the establishment of serine phosphorylation as a regulator of Stat5 transcriptional activity in T lymphocytes (34), this observation prompted us to analyze the importance of specific serine residues within Stat5 for 17ß-estradiol-induced activation of the ß-casein promoter. Stat5ab-/- cells were transiently transfected with wild-type Stat5a or different point mutants, in which serine residues 725 and/or 779 in the C-terminal transactivation domain had been changed to alanines (S725A, S779A, and S725/779) (35), together with the ß-casein reporter gene and NLSA. Interestingly, the S725A, S779A, and S725/779A mutants all showed a lower induction of the reporter in response to 17ß-estradiol compared with wild-type Stat5a (Fig. 7
). As expected, the Y694F mutant, in which the critical tyrosine residue 694 had been changed to phenylalanine, was unable to induce activation above basal level (Fig. 7
). In conclusion, these results demonstrate that tyrosine phosphorylation is a prerequisite for 17ß- estradiol-induced transactivation of Stat5a, whereas intact serine residues in the transactivation domain are important for optimal transcriptional activity.

View larger version (18K):
[in this window]
[in a new window]
|
Figure 7. Intact Serine Residues Within the Transactivation Domain of Stat5a Are Required for Optimal ß-Casein Reporter Activity in Response to 17ß-Estradiol
Stat5ab -/- cells were transfected with 1 µg ß-casein reporter gene together with 200 ng NLSA and either 500 ng Stat5a or point mutants of Stat5a, S725A, S779A, S725/779A, or Y694F, as indicated. Cells were treated with either no hormone (NH) or 10-8 M 17ß-estradiol (E2), and the reporter activity was analyzed 24 h thereafter. The luciferase activity was normalized using ß-galactosidase as an internal control. Data are representative of at least three independent experiments performed in duplicate. Mean ± SD are shown.
|
|
DNA-Binding Activity of ER Is Dispensable, whereas the LBD Is Sufficient for Mediating 17ß-Estradiol-Induced Activation of the ß-Casein Promoter
To analyze the contribution of different domains within ER to mediate activation of the ß-casein promoter, COS-7 cells were transiently transfected with the ß-casein reporter gene and various deletion and point mutants of ER
. Figure 8A
shows a schematic representation of the ER constructs used in the experiment. Figure 8B
shows how the wild-type ER
, in the presence of 17ß-estradiol, activated the reporter gene 4-fold compared with control cells without transfected ER. The importance of the DBD was analyzed by using a DNA-binding mutant in which two cysteine residues in the second zinc-finger had been changed to alanines. These mutations have previously been shown to inhibit DNA binding to an ERE (36). As expected, the DBD was found to be dispensable for activation of the reporter, demonstrating that the observed activity is not mediated through classical transcriptional activation by ER. The N-terminal domain was found to be dispensable, as shown using the N-terminal-deletion ER
mutant (182599), while deletion of the C-terminal domain (1339) prevented reporter activation. An ER with mutations that abrogate interactions with nuclear hormone receptor coactivators, such as the ER
AF2 mutant (L543A/L544A) (37) or the ER
(K366A) (38), did not block activation of the reporter. However, a 50% reduction in activation was observed with the AF2 mutant, while the K366A mutant was as active as the wild-type receptor. An ER
single-point mutant that is unable to bind 17ß-estradiol (G525R) (39) did not activate the reporter above background, indicating that the activity is mediated by hormone-bound ER. Furthermore, mutations in the helix 11 dimerization domain (L508/512/515E) was deleterious to the ability of ER
to activate the reporter. This dimerization-defective mutant receptor is unable to bind DNA and is transcriptionally inactive (40). Deletion of the C-terminal-most F domain of ER
did not affect reporter activation. Tyr 541, located in the N-terminal end of helix 12, has been shown to be important for maintaining the receptor in an inactive state in the absence of hormone (41). However, the constitutively active ER
mutant, Y541-E, showed the same 17ß-estradiol-induced activation as that found for the wild-type receptor. Finally, NLSA, which contains a deletion of nuclear localization signals in the hinge domain and which is cytoplasmic in the absence of hormone (Fig. 2B
and Ref. 22), activated the reporter gene 11-fold compared with control cells (see also Fig. 2C
). Taken together, these results demonstrate that activation of the ß-casein promoter by 17ß-estradiol is mediated through the ligand binding domain(s) of an ER homodimer. Furthermore, cytoplasmic localization of the receptor aids efficient induction whereas interaction with coactivators in the nucleus is not essential.

View larger version (19K):
[in this window]
[in a new window]
|
Figure 8. Identification of Domains in ER That Are Important for Activation of the ß-Casein Reporter Gene
A, Schematic representation of the different ER constructs used. DBDm, C241A/C244A, defective in DNA-binding, 1339, C-terminal deletion mutant, 182599, N-terminal deletion mutant, AF-2 m, L543A/L544A, activation function 2 mutated with two point mutations in the amphipathic helix 12, K366A, defective in coactivator interaction, G525R, defective in ligand binding, DIMm, L508/512/515E, dimerization-defective mutant, F, the F-domain in the very C-terminal deleted, Y547-E, mutation in a conserved tyrosine residue, NLSA, 245307, disrupted localization. B, COS-7 cells were transfected with 1 µg ß-casein reporter gene together with 200 ng ER wt or different point- and deletion mutants of ER . Cells were treated with either no hormone (NH) or 10-8 M 17ß-estradiol (E2), and the reporter activity was analyzed 24 h thereafter. The luciferase activity was normalized using ß-galactosidase as an internal control. Data are representative of at least three independent experiments performed in duplicate. Mean ± SD are shown.
|
|
 |
DISCUSSION
|
---|
In this study we have identified a novel pathway involving ER-dependent activation of cytoplasmic signal transduction pathways, resulting in phosphorylation and subsequent transactivation of a target transcription factor. Two members of the Stat protein family, Stat5 and Stat3, are activated by the nongenomic action of ER described here.
17ß-Estradiol was shown to rapidly induce tyrosine and serine phosphorylation, nuclear translocation, and DNA-binding activity of Stat5 in endothelial cells (Fig. 1
, A and B) and to activate the ß-casein promoter in an ER-dependent manner (Fig. 1
, C and D). Phosphorylation of Stat5 and induction of promoter activity were abrogated in the presence of antiestrogens (Figs. 1A
and 5
), suggesting a requirement for an agonist-bound ER. In addition to the true ß-casein promoter, the consensus Stat5 LHRE reporter gene and two Stat3-regulated reporter genes were efficiently induced (Figs. 3A
and 4B
), demonstrating that both Stat5 and Stat3 are targeted by the signal transduction pathway induced by 17ß-estradiol-bound ER. Tyrosine and serine phosphorylation and DNA-binding activity of Stat3 were also shown to be induced by 17ß-estradiol (Fig. 4
, A and C). The ability of 17ß-estradiol to promote nuclear translocation of Stats has not been described previously, whereas it has been demonstrated with glucocorticoids (42) and progestins (43).
An intact MAPK signaling pathway was found to be required for 17ß-estradiol-induced activation of the ß-casein promoter, as shown by abrogated activity in the presence of the MEK1/2 inhibitor, UO126 (Fig. 6A
). As further evidence for the involvement of the MAPK pathway, ERK1/2 was shown to be rapidly phosphorylated upon 17ß-estradiol treatment of endothelial cells (Fig. 6C
), which is in agreement with previous studies that showed 17ß-estradiol-induced activation of ERK1/2 in endothelial cells (16) as well as in other cells and tissues (Refs. 8 , 9 , 12 , 13 , 44 , and 45 and our unpublished data). Consistent with the studies by Migliaccio et al. (8, 9) and Kousteni et al. (12), activation of ERK1/2 by 17ß-estradiol was found to be dependent on initial Src-kinase activity (Fig. 6C
). Src-kinase was also found to be required for 17ß-estradiol-induced activation of the ß-casein promoter, since the activity was completely abrogated in the presence of the Src-kinase inhibitors, PP1 and genistein, and was not observed in cells lacking functional Src-kinase (Fig. 6
, A and B). Phosphotyrosine 537 of human ER
(corresponding to tyrosine 541 of mouse ER
) was recently shown to interact with the SH2 domain of Src-kinase (46). However, tyrosine 541 was dispensable for 17ß-estradiol-induced activation of the ß-casein promoter (Fig. 8
), suggesting that Src-kinase is activated downstream of ER by a distinct mechanism in this pathway. In addition to MAPK and Src-kinase activities, an intact PI3-kinase signaling pathway was also found to be required (Fig. 6A
). Recent reports have shown that rapid, nongenomic activation of endothelial nitric oxide synthase by 17ß-estradiol and ER
in endothelial cells involves the PI3-kinase signaling pathway (16, 17, 47) and that 17ß-estradiol increases PI3-kinase activity in MCF-7 cells (14). Furthermore, simultaneous PI3-kinase and MAPK activity has been shown to contribute to serine phosphorylation of Stat3, resulting in optimal transcriptional activity (48, 49). Although some of the described nongenomic actions of estrogens are mediated by modulation of G protein-coupled receptors (50), G protein-mediated signal transduction was shown not to be part of the 17ß-estradiol-induced pathway targeting Stats (Fig. 6A
).
Cytoplasmic sublocalization of ERs was shown to correlate with a more efficient activation of the ß-casein promoter (Fig. 2C
). In vivo, however, it is unclear whether the endogenous receptor exhibits this action through a specific pool of membrane-bound receptors or receptors that "pass by" the membrane during nucleocytoplasmic events (22). We believe, however, that the presence of ERs for a certain amount of time within plasma membrane domains facilitates signal transduction. Several members of the MAPK signaling pathway, as well as Src-kinase and PI3-kinase, are found clustered in caveolae, which are specialized membrane invaginations enriched in the scaffolding protein, caveolin-1 (51). ER
has been shown to coimmunoprecipitate with caveolin-1 (52), and both ER
(21) and Stat3 have been detected in caveolae (53). It is thus conceivable that the presence of ERs, Stats, and various signal transduction proteins in a defined cellular compartment, e.g. caveolae, facilitates signal transduction induced by an agonist-bound ER. This idea is supported by the finding that targeting of ERß to cellular membranes results in more efficient activation of the ß-casein promoter (Fig. 2C
). In contrast to NLSA, which located to the nucleus after several hours of exposure to hormone (22), the cellular localization of ERß Mem was not affected by the addition of hormone (data not shown), further verifying that the initial action is mediated by an agonist-bound receptor present in the cytoplasm.
In transient transfection experiments, intact serine residues within the transactivation domain of Stat5a were shown to be required for optimal 17ß-estradiol-induced activation of the ß-casein promoter (Fig. 7
). In addition to basic tyrosine phosphorylation, serine phosphorylation has been shown to affect the transcriptional activities of Stat1 and Stat3 in particular, and also that of Stat5 under certain conditions (34, 35, 54), although the mechanism has not been fully defined. However, members of the MAPK family have been implicated in the phosphorylation of serine residues conserved among the Stats (55, 56, 57, 58). Apart from the two characterized serine phosphorylation sites in Stat5a, residues 725 and 779 (57, 59), it is possible that additional serine residues within the molecule are preferentially phosphorylated and involved in 17ß-estradiol-induced transactivation.
Our analysis of functional domains in ER that are required for mediating activation of the ß-casein promoter, via the nongenomic mechanism described in this paper, revealed that the LBD is essential, whereas the DBD and the activation function 2 (AF-2) coactivator binding regions are dispensable. The findings, that an ER which is unable to bind ligand is inactive (Fig. 8
) and that the activity is abrogated in the presence of ER antagonists (Fig. 5
), confirm that the activity is mediated by an agonist-bound ER. The conformational change in the constitutively active Y541-E mutant, which is thought to generate an interacting surface allowing recruitment of coactivators in the absence of hormone (41), does not induce activation of the reporter (Fig. 8
), implying that the surface targeting signal transduction pathways is different. The dimerization-defective mutant, which contains three mutated leucine residues in helix 11, is unable to activate the reporter (Fig. 8
). This could be due to either a requirement for an ER dimer or, alternatively, the domain mutated being required for proper protein-protein interactions mediating the induction of signal transduction pathways.
Recently, we reported that ER
and -ß can act as coactivators for PRL-activated Stat5 on the ß-casein promoter and that ERs are capable of interacting with Stat5 via the DBD/hinge domain (60). The deletion mutant NLSA is unable to interact with Stat5 and does not potentiate the transcriptional activity of PRL-activated Stat5 (our unpublished data). However, it markedly induces activation of the ß-casein promoter via the mechanism described in this paper, demonstrating that the transcriptional activity of Stat5 can be modulated via distinct ER-mediated actions.
Taken together, this study identifies a novel mechanism of gene activation by ERs in which 17ß-estradiol initiates cytoplasmic signaling events upon binding to the receptor, resulting in phosphorylation and subsequent transactivation of a target transcription factor. Interestingly, induction of the signaling cascade requires an agonist-bound receptor, but may dispense with other functional domains except for the LBD. We are now able to discriminate between three diverse actions of ERs: classical regulation of ERE-containing genes, cross-talk with PRL-activated Stat5b on the ß-casein promoter (60), and, as put forward in this paper, activation of Stats via induction of cytoplasmic signaling pathways. In contrast to the two latter actions, the classical mechanism of ER activation requires binding of coactivator proteins. This suggests that different conformations of the receptor are involved in various activities, potentially enabling ligand design to target specific actions of the receptor for therapeutic use.
 |
MATERIALS AND METHODS
|
---|
Plasmids
The JAK2 KN (Lys882Glu) was provided by Olli Silvennoinen (Tampere, Finland) (61). The pLucCRP and pLucTKS3 reporter plasmids and the Stat3ß expression plasmid were provided by Richard Jove (Tampa, FL) (28). The Stat5a and Stat5a mutants (S725A, S779A, S725/779A, and Y694F) were provided by Nancy Hynes (Basel, Switzerland) (59) and Hallgeir Rui (Bethesda, MD) (35). The pSG5 L508A/L512A/L515A was provided by Janet Valentine (London, UK) (40). The following plasmids have been described previously: the ß-casein (-344 to -1) luciferase reporter plasmid and the Stat5a MGF
750 (26, 62); the long form of the Prl-R (63); the SrcY527F (64); the mouse ER
expression vector pMT2-MOR (36); the ER
DNA-binding mutant pMT2-MOR C241A/C244A (36); the ER
N-terminal deletion mutant pMT2-MOR 182595 (36); the ER
C-terminal deletion mutant pMT2-MOR 1339 (65); the ER
AF-2 mutant pMT2-MOR L543A/L544A (66); the pMT2-MOR K366A (38); the pMT2-MOR Y541F (41); the pMT2-MOR
245307 (NLSA) (22); and the human ERß expression vector, pSG5-ERß (67). The ER
F mutant was prepared using the Gene Editor in vitro Site-Directed Mutagenesis System (Promega Corp., Madison, WI) with an oligonucleotide primer designed to delete the F domain (amino acids 550595) of pMT2-MOR. The ERß LYN mutant was generated by subcloning a fatty acylation site into the coding sequence of pSG5-ERß wt. The DNA sequences of the new constructs were verified by automated sequencing.
Estrogens, Growth Factors, and Inhibitors
17ß-Estradiol, the inactive stereoisomer, 17
-estradiol, genistein, 4-hydroxy tamoxifen, ovine PRL, recombinant human EGF, and pertussis toxin were purchased from Sigma (St. Louis, MO). The pure ER antagonist, ICI 182,780, was obtained from Tocris Cookson, Inc. (Ballwin, MO). The MEK1/2 inhibitor, UO126, and the PI3-kinase inhibitor, LY294002, were purchased from Cell Signaling Technology (Beverly, MA). The Src-family tyrosine kinase inhibitor, PP1, and the p38MAPK Inhibitor, SB203580, were obtained from BIOMOL Research Laboratories, Inc. (Plymouth Meeting, PA). Raloxifen was a gift from Stefan Nilsson (Karo Bio AB, Huddinge, Sweden).
Cell Culture and Transient Transfection Techniques
COS-7 cells, Stat5ab-/- cells (a gift from J. Ihle), and SYF cells (purchased from ATCC, Manassas, VA) were cultured in DMEM supplemented with 10% FBS (Life Technologies, Inc., Buckinghamshire, UK). PAE cells (a gift from Stefan Wennström) were cultured in DMEM/F-12 (Life Technologies, Inc.) supplemented with 10% FBS. For transient transfection assays, PAE cells, COS-7 cells, Stat5ab-/- cells, and SYF cells were seeded in phenol-red free DMEM (Life Technologies, Inc.) supplemented with 5% dextran-charcoal-stripped (DCS) FBS in 24-well microtiter plates, 24 h before transfection. Cells were transfected with 1 µg reporter plasmid, 250 ng cytomegalovirus-ß-galactosidase plasmid as an internal control, and various expression plasmids, as indicated in the figure legends, together with an empty expression vector to a total of 2 µg DNA per well, using a modified calcium phosphate coprecipitation method (68). The transfection medium was changed after 24 h to a phenol red-free DMEM supplemented with 0,05% DCS FBS. The hormones were added as indicated in the figures. After 24 h, the cells were harvested in lysis buffer (10 mM Tris-HCl, pH 8.0; 1 mM EDTA; 150 mM NaCl; and 0.65% Nonidet P-40). Extracts were assayed for luciferase and ß-galactosidase activity in a microplate reader (Lucy-1; Anthos, Salzburg, Austria). For immunofluorescence studies, COS-7 cells were seeded onto poly-L-lysine precoated coverslips placed in six-well tissue culture plates in phenol red-free DMEM supplemented with 5% DCS FBS. Cells were transfected as described above and then processed for histological studies.
Indirect Immunofluorescence
For histological studies, cells were washed with PBS and fixed in 3% paraformaldehyde solution [3% (wt/vol) paraformaldehyde, 0.1 mM CaCl2 and 0.1 mM MgCl2, pH 7.4, in PBS] for 20 min. The cells were then washed with PBS, permeabilized in 0.2% Triton X-100/PBS for 4 min, and then washed with 10 mg/ml BSA in PBS. ER
and -ß proteins were detected using the ER
polyclonal antibody H-184 (Santa Cruz Biotechnology, Inc., Santa Cruz, CA) and an ERß polyclonal antibody (Upstate Biotechnology, Inc., Lake Placid, NY), respectively. The antibodies were diluted 1:100 in 10 mg/ml BSA in PBS, and 40 µl samples were placed on parafilm in a box containing moist Whatman 3MM (Clifton, NJ). Coverslips were inverted onto the antibody and incubated for 1 h at room temperature. They were then washed with 10 mg/ml BSA in PBS, and the above procedure was repeated with an appropriate fluorescein isothiocyanate-conjugated secondary antibody (DAKO A/S, Glostrop, Denmark). Cell nuclei were stained with 4,6-diamidino2-phenylindole. The coverslips were subsequently mounted on slides with Vectashield Mounting Medium (Vector Laboratories, Inc., Burlingame, CA). In all cases, untransfected or transfected cells incubated without a primary antibody were included as controls. Slides were examined using a DMRXA microscope (Leica Corp., Deerfield, IL), and digitally imaged using a Hamamatsu C488040 CCD camera (Hamamatsu Photonics Norden AB, Solna, Sweden), the Openlab software package (Improvision, Sollentuna, Sweden), and Adobe Photoshop software.
Western Blotting
PAE cells were seeded in six-well tissue culture plates in phenol red-free DMEM supplemented with 5% DCS FBS. After 48 h, cells were washed three times with PBS before incubation for 24 h in serum-free medium. Cells were then washed twice before addition of hormone and various inhibitors for the times indicated in the figure legends. Cells were lysed in hot Laemmelli lysis buffer directly in the petri dish, and proteins were separated by electrophoresis through a 10% polyacrylamide gel. Proteins were transferred to Hybond-C membranes (Amersham International, Buckinghamshire, UK). The membranes were probed using either phospho-Stat5A/B (Y694/699), phospho-Stat5A/B (S726/731) (Upstate Biotechnology, Inc.), Stat5a (L-20), Stat5b (C-17), Stat3 (F-2) (Santa Cruz Biotechnology, Inc.), phospho-Stat3 (Tyr705), phospho-Stat3 (Ser727), phospho-p44/42 MAPK (Thr202/Tyr204), or p44/p42 MAPK (Cell Signaling Technology) antibodies. Proteins were detected using the ECL chemiluminescence system (Amersham Pharmacia Biotech).
DNA Affinity Purification
Biotinylated oligonucleotide (5'-AGATTTCTAGGAATTCAAATC-3'), derived from the ß-casein promoter and including a Stat5 response element, was annealed to an antisense strand. An oligonucleotide with two point mutations in the consensus Stat5 response element (5'-AGATTTCTATTAATTCAAATC-3') and an oligonucleotide derived from the human CRP promoter and including a Stat3 response element (5'-CCTCTTCCCGAAGCTCT-3') were prepared in a similar manner. PAE cells were plated in phenol red-free DMEM supplemented with 5% DCS FBS. After 48 h, cells were washed three times with PBS before incubation for 24 h in serum-free medium. Cells were then washed twice before addition of hormone at various time points, as indicated in the figure legends, and nuclear extracts were prepared as previously described (60). Nuclear extracts (containing a total of
300 µg protein) were mixed with 2 µg double-stranded biotinylated DNA oligo in 800 µl A-buffer (8 mM trisphosphate, pH 7.4; 120 mM KCl; 8% glycerol; 4 mM dithiothreitol; 100 µM Na orthovanadate; 1 mM phenylmethylsulfonylfluoride; 10,000 U/ml aprotinin) and incubated at 4 C for 1 h on a rotator. Streptavidin MagneSphere Paramagnetic Particles (Promega Corp.) resuspended in A-buffer were added to a final volume of 1 ml, and incubation was continued for 15 min. After washing, bound proteins were eluted in loading buffer and separated by electrophoresis through a 10% polyacrylamide gel. Proteins were transferred to Hybond-C membranes (Amersham International) and detected by Western blotting with the Stat5 (C-17) or Stat3 (F-2) antibody (Santa Cruz Biotechnology, Inc.). Proteins were detected using the ECL chemiluminescence system.
 |
ACKNOWLEDGMENTS
|
---|
We thank Janet Valentine, Nancy Hynes, James Ihle, Stefan Wennström, James Darnell, Richard Jove, Hallgeir Rui, Olli Silvennoinen, and Stefan Nilsson for generous gifts of reagents. We also thank Janet Valentine, Thomas Perlmann, and Christer Höög for critical reading of the manuscript.
 |
FOOTNOTES
|
---|
This work was supported by the Swedish Cancer Society, the Karolinska Institute, the Swedish Medical Society, the M. Bergwall Foundation, the H&G Jaensson Foundation, and the Royal Swedish Academy of Sciences.
Abbreviations: AF-2, Activation function 2; CRP, C-reactive protein; DBD, DNA-binding domain; DCS, dextran-charcoal-stripped; EGF, epidermal growth factor; ER, estrogen receptor; ERß wt, wild-type ERß; ERß Mem, N terminus of ERß; ERE, estrogen response element; FBS, fetal bovine serum; Jak, Janus kinase; LBD, ligand-binding domain; LHRE, lactogenic hormone response element; MEK, MAPK kinase; NLSA, nuclear localization signals in the hinge domain; OHT, 4-hydroxytamoxifen; PAE, porcine aortic endothelial; PI3-kinase, phosphatidylinositol 3-kinase; PRL, prolactin; Stat, signal transducer and activator of transcription.
Received for publication February 18, 2002.
Accepted for publication June 24, 2002.
 |
REFERENCES
|
---|
- Parker MG, Arbuckle N, Dauvois S, Danielian P, White R 1993 Structure and function of the estrogen receptor. Ann NY Acad Sci 684:119126[Abstract]
- Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans RE 1995 The nuclear receptor superfamily: the second decade. Cell 83:835839[Medline]
- Beato M, Herrlich P, Schutz G 1995 Steroid-hormone receptorsmany actors in search of a plot. Cell 83:851857[Medline]
- Bevan C, Parker M 1999 The role of coactivators in steroid hormone action. Exp Cell Res 253:349356[CrossRef][Medline]
- Glass CK, Rosenfeld MG 2000 The coregulator exchange in transcriptional functions of nuclear receptors. Genes Dev 14:121141[Free Full Text]
- Aronica SM, Kraus WL, Katzenellenbogen BS 1994 Estrogen action via the cAMP signaling pathway: stimulation of adenylate cyclase and cAMP-regulated gene transcription. Proc Natl Acad Sci USA 91:85178521[Abstract]
- Razandi M, Pedram A, Greene GL, Levin ER 1999 Cell membrane and nuclear estrogen receptors (ERs) originate from a single transcript: studies of ER
and ERß expressed in Chinese hamster ovary cells. Mol Endocrinol 13:307319[Abstract/Free Full Text]
- Migliaccio A, Di Domenico M, Castoria G, de Falco A, Bontempo P, Nola E, Auricchio F 1996 Tyrosine kinase/p21ras/MAP-kinase pathway activation by estradiol-receptor complex in MCF-7 cells. EMBO J 15:12921300[Abstract]
- Migliaccio A, Piccolo D, Castoria G, Di Domenico M, Bilancio A, Lombardi M, Gong W, Beato M, Auricchio F 1998 Activation of the Src/p21ras/Erk pathway by progesterone receptor via cross-talk with estrogen receptor. EMBO J 17:20082018[Free Full Text]
- Improta-Brears T, Whorton AR, Codazzi F, York JD, Meyer T, McDonnell DP 1999 Estrogen-induced activation of mitogen-activated protein kinase requires mobilization of intracellular calcium. Proc Natl Acad Sci USA 96:46864691[Abstract/Free Full Text]
- Kahlert S, Nuedling S, van Eickels M, Vetter H, Meyer R, Grohe C 2000 Estrogen receptor
rapidly activates the IGF-1 receptor pathway. J Biol Chem 275:1844718453[Abstract/Free Full Text]
- Kousteni S, Bellido T, Plotkin LI, OBrien CA, Bodenner DL, Han L, Han K, DiGregorio GB, Katzenellenbogen JA, Katzenellenbogen BS, Roberson PK, Weinstein RS, Jilka RL, Manolagas SC 2001 Nongenotropic, sex-nonspecific signaling through the estrogen or androgen receptors: dissociation from transcriptional activity. Cell 104:719730[Medline]
- de Jager T, Pelzer T, Muller-Botz S, Imam A, Muck J, Neyses L 2001 Mechanisms of estrogen receptor action in the myocardium. Rapid gene activation via the ERK1/2 pathway and serum response elements. J Biol Chem 276:2787327880[Abstract/Free Full Text]
- Castoria G, Migliaccio A, Bilancio A, Di Domenico M, de Falco A, Lombardi M, Fiorentino R, Varricchio L, Barone MV, Auricchio F 2001 PI3-kinase in concert with Src promotes the S-phase entry of oestradiol-stimulated MCF-7 cells. EMBO J 20:60506059[Abstract/Free Full Text]
- Kim HP, Lee JY, Jeong JK, Bae SW, Lee HK, Jo I 1999 Nongenomic stimulation of nitric oxide release by estrogen is mediated by estrogen receptor
localized in caveolae. Biochem Biophys Res Commun 263:257262[CrossRef][Medline]
- Chen Z, Yuhanna IS, Galcheva-Gargova Z, Karas RH, Mendelsohn ME, Shaul PW 1999 Estrogen receptor
mediates the nongenomic activation of endothelial nitric oxide synthase by estrogen. J Clin Invest 103:401406[Abstract/Free Full Text]
- Simoncini T, Hafezi-Moghadam A, Brazil DP, Ley K, Chin WW, Liao JK 2000 Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol-3-OH kinase. Nature 407:538541[CrossRef][Medline]
- Kelly MJ, Levin ER 2001 Rapid actions of plasma membrane estrogen receptors. Trends Endocrinol Metab 12:152156[CrossRef][Medline]
- Darnell Jr JE 1997 STATs and gene regulation. Science 277:16301635[Abstract/Free Full Text]
- Decker T, Kovarik P 2000 Serine phosphorylation of STATs. Oncogene 19:26282637[CrossRef][Medline]
- Chambliss KL, Yuhanna IS, Mineo C, Liu P, German Z, Sherman TS, Mendelsohn ME, Anderson RG, Shaul PW 2000 Estrogen receptor
and endothelial nitric oxide synthase are organized into a functional signaling module in caveolae. Circ Res 87:E44E52
- Dauvois S, White R, Parker MG 1993 The antiestrogen ICI 182780 disrupts estrogen receptor nucleocytoplasmic shuttling. J Cell Sci 106:13771388[Abstract/Free Full Text]
- Milligan G, Parenti M, Magee AI 1995 The dynamic role of palmitoylation in signal transduction. Trends Biochem Sci 20:181187[CrossRef][Medline]
- Groner B, Gouilleux F 1995 Prolactin-mediated gene activation in mammary epithelial cells. Curr Opin Genet Dev 5:587594[CrossRef][Medline]
- Goffin V, Kinet S, Ferrag F, Binart N, Martial JA, Kelly PA 1996 Antagonistic properties of human prolactin analogs that show paradoxical agonistic activity in the Nb2 bioassay. J Biol Chem 271:1657316579[Abstract/Free Full Text]
- Moriggl R, Gouilleux-Gruart V, Jahne R, Berchtold S, Gartmann C, Liu X, Hennighausen L, Sotiropoulos A, Groner B, Gouilleux F 1996 Deletion of the carboxyl-terminal transactivation domain of MGF-Stat5 results in sustained DNA binding and a dominant negative phenotype. Mol Cell Biol 16:56915700[Abstract]
- OFarrell AM, Liu Y, Moore KW, Mui AL 1998 IL-10 inhibits macrophage activation and proliferation by distinct signaling mechanisms: evidence for Stat3-dependent and -independent pathways. EMBO J 17:10061018[Abstract/Free Full Text]
- Turkson J, Bowman T, Garcia R, Caldenhoven E, De Groot RP, Jove R 1998 Stat3 activation by Src induces specific gene regulation and is required for cell transformation. Mol Cell Biol 18:25452552[Abstract/Free Full Text]
- Caldenhoven E, van Dijk TB, Solari R, Armstrong J, Raaijmakers JA, Lammers JW, Koenderman L, de Groot RP 1996 STAT3ß, a splice variant of transcription factor STAT3, is a dominant negative regulator of transcription. J Biol Chem 271:1322113227[Abstract/Free Full Text]
- Barkhem T, Carlsson B, Nilsson Y, Enmark E, Gustafsson J, Nilsson S 1998 Differential response of estrogen receptor
and estrogen receptor ß to partial estrogen agonists/antagonists. Mol Pharmacol 54:105112[Abstract/Free Full Text]
- Hanke JH, Gardner JP, Dow RL, Changelian PS, Brissette WH, Weringer EJ, Pollok BA, Connelly PA 1996 Discovery of a novel, potent, and Src family-selective tyrosine kinase inhibitor. Study of Lck- and FynT-dependent T cell activation. J Biol Chem 271:695701[Abstract/Free Full Text]
- Akiyama T, Ogawara H 1991 Use and specificity of genistein as inhibitor of protein-tyrosine kinases. Methods Enzymol 201:362370[Medline]
- Klinghoffer RA, Sachsenmaier C, Cooper JA, Soriano P 1999 Src family kinases are required for integrin but not PDGFR signal transduction. EMBO J 18:24592471[Abstract/Free Full Text]
- Beadling C, Ng J, Babbage JW, Cantrell DA 1996 Interleukin-2 activation of STAT5 requires the convergent action of tyrosine kinases and a serine/threonine kinase pathway distinct from the Raf1/ERK2 MAP kinase pathway. EMBO J 15:19021913[Abstract]
- Yamashita H, Nevalainen MT, Xu J, LeBaron MJ, Wagner KU, Erwin RA, Harmon JM, Hennighausen L, Kirken RA, Rui H 2001 Role of serine phosphorylation of Stat5a in prolactin-stimulated ß-casein gene expression. Mol Cell Endocrinol 183:151163[CrossRef][Medline]
- Lahooti H, White R, Danielian PS, Parker MG 1994 Characterization of ligand-dependent phosphorylation of the estrogen receptor. Mol Endocrinol 8:182188[Abstract]
- Cavailles V, Dauvois S, LHorset F, Lopez G, Hoare S, Kushner PJ, Parker MG 1995 Nuclear factor RIP140 modulates transcriptional activation by the estrogen receptor. EMBO J 14:37413751[Abstract]
- Henttu PM, Kalkhoven E, Parker MG 1997 AF-2 activity and recruitment of steroid receptor coactivator 1 to the estrogen receptor depend on a lysine residue conserved in nuclear receptors. Mol Cell Biol 17:18321839[Abstract]
- Danielian PS, White R, Hoare SA, Fawell SE, Parker MG 1993 Identification of residues in the estrogen receptor that confer differential sensitivity to estrogen and hydroxytamoxifen. Mol Endocrinol 7:232240[Abstract]
- Valentine JE, Kalkhoven E, White R, Hoare S, Parker MG 2000 Mutations in the estrogen receptor ligand binding domain discriminate between hormone-dependent transactivation and transrepression. J Biol Chem 275:2532225329[Abstract/Free Full Text]
- White R, Sjoberg M, Kalkhoven E, Parker MG 1997 Ligand-independent activation of the oestrogen receptor by mutation of a conserved tyrosine. EMBO J 16:14271435[Abstract/Free Full Text]
- Wyszomierski SL, Yeh J, Rosen JM 1999 Glucocorticoid receptor/signal transducer and activator of transcription 5 (STAT5) interactions enhance STAT5 activation by prolonging STAT5 DNA binding and tyrosine phosphorylation. Mol Endocrinol 13:330343[Abstract/Free Full Text]
- Richer JK, Lange CA, Manning NG, Owen G, Powell R, Horwitz KB 1998 Convergence of progesterone with growth factor and cytokine signaling in breast cancer. Progesterone receptors regulate signal transducers and activators of transcription expression and activity. J Biol Chem 273:3131731326[Abstract/Free Full Text]
- Duan R, Xie W, Burghardt RC, Safe S 2001 Estrogen receptor-mediated activation of the serum response element in MCF-7 cells through MAPK-dependent phosphorylation of Elk-1. J Biol Chem 276:1159011598[Abstract/Free Full Text]
- Jessop HL, Sjoberg M, Cheng MZ, Zaman G, Wheeler-Jones CP, Lanyon LE 2001 Mechanical strain and estrogen activate estrogen receptor
in bone cells. J Bone Miner Res 16:10451055[Medline]
- Migliaccio A, Castoria G, Di Domenico M, de Falco A, Bilancio A, Lombardi M, Barone MV, Ametrano D, Zannini MS, Abbondanza C, Auricchio F 2000 Steroid-induced androgen receptor-oestradiol receptor ß-Src complex triggers prostate cancer cell proliferation. EMBO J 19:54065417[Abstract/Free Full Text]
- Haynes MP, Sinha D, Russell KS, Collinge M, Fulton D, Morales-Ruiz M, Sessa WC, Bender JR 2000 Membrane estrogen receptor engagement activates endothelial nitric oxide synthase via the PI3-kinase-Akt pathway in human endothelial cells. Circ Res 87:677682[Abstract/Free Full Text]
- Su L, Rickert RC, David M 1999 Rapid STAT phosphorylation via the B cell receptor. Modulatory role of CD19. J Biol Chem 274:3177031774[Abstract/Free Full Text]
- Duarte RF, Frank DA 2000 SCF and G-CSF lead to the synergistic induction of proliferation and gene expression through complementary signaling pathways. Blood 96:34223430[Abstract/Free Full Text]
- Kelly MJ, Wagner EJ 1999 Estrogen modulation of G-protein-coupled receptors. Trends Endocrinol Metab 10:369374[CrossRef][Medline]
- Okamoto T, Schlegel A, Scherer PE, Lisanti MP 1998 Caveolins, a family of scaffolding proteins for organizing "preassembled signaling complexes" at the plasma membrane. J Biol Chem 273:54195422[Free Full Text]
- Schlegel A, Wang C, Katzenellenbogen BS, Pestell RG, Lisanti MP 1999 Caveolin-1 potentiates estrogen receptor
(ER
) signaling. caveolin-1 drives ligand-independent nuclear translocation and activation of ER
. J Biol Chem 274:3355133556[Abstract/Free Full Text]
- Ju H, Venema VJ, Liang H, Harris MB, Zou R, Venema RC 2000 Bradykinin activates the Janus-activated kinase/signal transducers and activators of transcription (JAK/STAT) pathway in vascular endothelial cells: localization of JAK/STAT signalling proteins in plasmalemmal caveolae. Biochem J 351:257264[CrossRef][Medline]
- Wen Z, Zhong Z, Darnell Jr JE 1995 Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell 82:241250[Medline]
- Chung J, Uchida E, Grammer TC, Blenis J 1997 STAT3 serine phosphorylation by ERK-dependent and -independent pathways negatively modulates its tyrosine phosphorylation. Mol Cell Biol 17:65086516[Abstract]
- Ng J, Cantrell D 1997 STAT3 is a serine kinase target in T lymphocytes. Interleukin 2 and T cell antigen receptor signals converge upon serine 727. J Biol Chem 272:2454224549[Abstract/Free Full Text]
- Yamashita H, Xu J, Erwin RA, Farrar WL, Kirken RA, Rui H 1998 Differential control of the phosphorylation state of proline-juxtaposed serine residues Ser725 of Stat5a and Ser730 of Stat5b in prolactin-sensitive cells. J Biol Chem 273:3021830224[Abstract/Free Full Text]
- Pircher TJ, Petersen H, Gustafsson JA, Haldosen LA 1999 Extracellular signal-regulated kinase (ERK) interacts with signal transducer and activator of transcription (STAT) 5a. Mol Endocrinol 13:555565[Abstract/Free Full Text]
- Beuvink I, Hess D, Flotow H, Hofsteenge J, Groner B, Hynes NE 2000 Stat5a serine phosphorylation. Serine 779 is constitutively phosphorylated in the mammary gland, and serine 725 phosphorylation influences prolactin-stimulated in vitro DNA binding activity. J Biol Chem 275:1024710255[Abstract/Free Full Text]
- Björnström L, Kilic E, Norman M, Parker MG, Sjöberg M 2001 Cross-talk between Stat5b and estrogen receptor-
and -ß in mammary epithelial cells. J Mol Endocrinol 27:93106[Abstract/Free Full Text]
- Paukku K, Valgeirsdottir S, Saharinen P, Bergman M, Heldin CH, Silvennoinen O 2000 Platelet-derived growth factor (PDGF)-induced activation of signal transducer and activator of transcription (Stat) 5 is mediated by PDGF ß-receptor and is not dependent on c-src, fyn, jak1 or jak2 kinases. Biochem J 345:759766[CrossRef][Medline]
- Gouilleux F, Wakao H, Mundt M, Groner B 1994 Prolactin induces phosphorylation of Tyr694 of Stat5 (MGF), a prerequisite for DNA binding and induction of transcription. EMBO J 13:43614369[Abstract]
- Lesueur L, Edery M, Paly J, Clark J, Kelly PA, Djiane J 1990 Prolactin stimulates milk protein promoter in CHO cells cotransfected with prolactin receptor cDNA. Mol Cell Endocrinol 71:R7R12
- Erpel T, Superti-Furga G, Courtneidge SA 1995 Mutational analysis of the Src SH3 domain: the same residues of the ligand binding surface are important for intra- and intermolecular interactions. EMBO J 14:963975[Abstract]
- Lees JA, Fawell SE, Parker MG 1989 Identification of two transactivation domains in the mouse oestrogen receptor. Nucleic Acids Res 17:54775488[Abstract]
- Danielian PS, White R, Lees JA, Parker MG 1992 Identification of a conserved region required for hormone dependent transcriptional activation by steroid hormone receptors [published erratum appears in EMBO J 1992 Jun;11(6):2366]. EMBO J 11:10251033[Abstract]
- Cowley SM, Parker MG 1999 A comparison of transcriptional activation by ER
and ERß. J Steroid Biochem Mol Biol 69:165175[CrossRef][Medline]
- Chen C, Okayama H 1987 High-efficiency transformation of mammalian cells by plasmid DNA. Mol Cell Biol 7:27452752[Medline]