Mechanisms of Estrogen Receptor Signaling - CiteSeerX

2 downloads 60 Views 412KB Size Report
Feb 3, 2005 - LDL-R, LDL receptor; NF-ĸЯ, nuclear factor ĸЯ; PI, phospho- inositol; SERM, selective ER modulators; SRE, serum re- sponse element; STAT ...
0888-8809/05/$15.00/0 Printed in U.S.A.

Molecular Endocrinology 19(4):833–842 Copyright © 2005 by The Endocrine Society doi: 10.1210/me.2004-0486

MINIREVIEW

Mechanisms of Estrogen Receptor Signaling: Convergence of Genomic and Nongenomic Actions on Target Genes Linda Bjo¨rnstro¨m and Maria Sjo¨berg Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden Estrogen receptors (ERs) act by regulating transcriptional processes. The classical mechanism of ER action involves estrogen binding to receptors in the nucleus, after which the receptors dimerize and bind to specific response elements known as estrogen response elements (EREs) located in the promoters of target genes. However, ERs can also regulate gene expression without directly binding to DNA. This occurs through protein-protein interactions with other DNA-binding transcription factors in the nucleus. In addition, membraneassociated ERs mediate nongenomic actions of estrogens, which can lead both to altered functions of proteins in the cytoplasm and to regulation of gene expression. The latter two mechanisms of ER action enable a broader range of genes to be

regulated than the range that can be regulated by the classical mechanism of ER action alone. This review surveys our knowledge about the molecular mechanism by which ERs regulate the expression of genes that do not contain EREs, and it gives examples of the ways in which the genomic and nongenomic actions of ERs on target genes converge. Genomic and nongenomic actions of ERs that do not depend on EREs influence the physiology of many target tissues, and thus, increasing our understanding of the molecular mechanisms behind these actions is highly relevant for the development of novel drugs that target specific receptor actions. (Molecular Endocrinology 19: 833–842, 2005)

E

dimerize and bind to specific response elements known as estrogen response elements (EREs) located in the promoters of target genes (1). Hormone binding also induces a conformational change within the ligand binding domain of the receptors, and this conformational change allows coactivator proteins to be recruited (2). However, evidence for signaling pathways that deviate from this classical model has emerged, and it is now accepted that ERs can regulate gene expression by a number of distinct mechanisms. Around one third of the genes in humans that are regulated by ERs do not contain ERE-like sequences (3). The molecular mechanisms by which ERs regulate transcription at alternative response elements are not fully understood but are becoming increasingly clear. ERs can regulate gene expression without binding directly to DNA by modulating the function of other classes of transcription factors through protein-protein interactions in the nucleus (4). The interaction of ERs with the activator protein 1 (AP-1) transcription factor complex is a typical example of such ERE-independent genomic actions. In addition, a number of estrogen-responsive genes that lack EREs contain ERE half-sites, or binding sites for the orphan nuclear hormone receptor SF-1 [SF-1 response elements (SFREs)] that serve as direct ER binding sites (3). ER␣, but not ER␤, is able to bind to SFREs (5).

STROGENS ARE STEROID hormones that regulate growth, differentiation, and function in a broad range of target tissues in the human body. The most potent and dominant estrogen in humans is 17␤estradiol, but lower levels of the estrogens estrone and estriol are also present. The biological effects of estrogens are mediated through estrogen receptor (ER) ␣ and ␤, which are members of a large superfamily of nuclear receptors. These receptors act as ligandactivated transcription factors. The classical mechanism of ER action involves estrogen binding to receptors in the nucleus, after which the receptors First Published Online February 3, 2005 Abbreviations: AF, Activation function; AP-1, activator protein 1; C/EBP␤, CCAAT/enhancer binding protein ␤; CRE, cAMP response element; CREB, CRE binding protein; DBD, DNA binding domain; EGF, epidermal growth factor; eNOS, endothelial nitric oxide synthase; ER, estrogen receptor; ERE, estrogen response element; LDL, low-density lipoprotein; LDL-R, LDL receptor; NF-␬␤, nuclear factor ␬␤; PI, phosphoinositol; SERM, selective ER modulators; SRE, serum response element; STAT, signal transducer and activator of transcription. Molecular Endocrinology is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community. 833

834

Mol Endocrinol, April 2005, 19(4):833–842

Estrogens exert some of their effects through the action of ERs on gene expression, but a number of other effects of estrogens are so rapid that they cannot depend on the activation of RNA and protein synthesis. These actions are known as nongenomic actions and are believed to be mediated through membraneassociated ERs. The actions are frequently associated with the activation of various protein-kinase cascades (6). However, nongenomic actions of estrogens may indirectly influence gene expression, through the activation of signal transduction pathways that eventually act on target transcription factors. The functions of many transcription factors, including AP-1, are regulated through protein kinase-mediated phosphorylation, and these transcription factors may thus be targets of nongenomic actions of estrogens. This signaling pathway can be referred to as nongenomic-togenomic signaling, and it provides for a mechanism, distinct from protein-protein interactions in the nucleus, by which ERs can modulate the functions of transcription factors, and thus regulate the expression of genes that do not contain EREs. This review surveys our knowledge about the molecular mechanisms by which ERs regulate transcription at alternative response elements. We give examples that show that genomic and nongenomic actions of ERs on target genes converge, and we briefly dis-

Bjo¨rnstro¨m and Sjo¨berg • Minireview

cuss the responses of ERs to synthetic ligands as a possibility of designing drugs that target specific receptor actions.

ERE-INDEPENDENT GENOMIC ACTIONS A number of studies have shown that ERs can regulate transcription without binding directly to DNA. The receptors in such cases are tethered through proteinprotein interactions to a transcription factor complex that contacts the DNA, as illustrated in Fig. 1. ERs regulate by this mechanism the expression of a large number of estrogen-responsive genes that do not contain EREs. The mechanism is commonly used by members of the nuclear receptor superfamily and is often referred to as transcriptional cross talk (4). Several genes are activated by 17␤-estradiol through the interaction of ERs with Fos and Jun proteins at AP-1 binding sites. Such genes include those for ovalbumin (7), IGF-I (8), collagenase (9), and cyclin D1 (10, 11). Transcription that depends on AP-1 is also repressed by ERs activated by 17␤-estradiol (12–14). One gene that is repressed by 17␤-estradiol through ER-AP-1 complexes is the choline acetyltransferase gene (15). ERs interact with AP-1 proteins in a com-

Fig. 1. Schematic Illustration of ER Signaling Mechanisms 1. Classical mechanism of ER action. Nuclear E2-ERs bind directly to EREs in target gene promoters. 2. ERE-independent genomic actions. Nuclear E2-ER complexes are tethered through protein-protein interactions to a transcription factor complex (TF) that contacts the target gene promoter. 3. Ligand-independent genomic actions. Growth factors (GF) activate protein-kinase cascades, leading to phosphorylation (P) and activation of nuclear ERs at EREs. 4. Nongenomic actions. Membrane E2-ER complexes activate protein-kinase cascades, leading to altered functions of proteins in the cytoplasm, e.g. activation of eNOS, or to regulation of gene expression through phosphorylation (P) and activation of a TF.

Bjo¨rnstro¨m and Sjo¨berg • Minireview

plex manner, and the involvement of ER activation function (AF) domains depends both on the receptor subtype and on the type of ligand (16). The activation of ER␣-AP-1 complexes that is induced by 17␤-estradiol requires the AF-2 domain of the receptor, which binds p160 coactivators and stabilizes in this way the formation of a multiprotein complex containing c-Jun, ER␣, and transcriptional coactivators at the promoter (17, 18). Genes that contain GC-rich promoter sequences are regulated in a similar manner through the interaction of ERs with the Sp1 transcription factor (19). Increasing numbers of genes are being found that are activated by 17␤-estradiol through ER-Sp1 complexes, including the LDL-R [low-density lipoprotein (LDL) receptor] (20), c-fos (21) and cyclin D1 (22) genes. The actions of ERs at Sp1 binding sites depend on the ligand, the cell type, and the receptor subtype (23), as is the case at for the actions of ERs at AP-1 binding sites (16). Repression of the IL-6 gene by 17␤-estradiol is mediated through the interaction of ERs with two transcription factors, nuclear factor ␬␤ (NF-␬B) and CCAAT/enhancer binding protein ␤ (C/EBP␤) (24, 25). In addition, it has been suggested that the repression of erythropoiesis by 17␤-estradiol involves the interaction of ERs with the GATA-1 transcription factor (26). ERs also regulate genes, such as the ␤-casein gene, that contain signal transducer and activator of transcription (STAT) 5 binding sites, but the outcome of the interaction of ERs with the STAT5 transcription factor is controversial. Some authors have suggested that STAT5-dependent transcription is repressed by ERs activated by 17␤-estradiol, and that this repression requires an intact ER AF-2 domain (27, 28). Other authors have suggested that the transcriptional activity of STAT5 is increased by ERs in a way that does not depend on the ER AF-2 domain (29). ERE-independent genomic actions that involve tethering of ERs to other DNA binding transcription factors do not require an interaction between the receptor and DNA, as is the case in the classical mechanism of ER action. However, the DNA binding domain (DBD) of the receptors is frequently involved, although intact DNA binding activity per se is not required (7–10, 13, 15, 25, 28, 29). Mutational analysis has revealed specific residues within the second zinc finger structure of the ER␤ DBD that discriminate between the classical mechanism of ER action and the modulation of AP-1 and STAT5 activities through tethering (14). This region is well conserved among nuclear receptors, and GR-mediated repression of AP-1 activity depends on residues within the second zinc finger structure of the GR DBD (30). The DBD may be required for proper protein-protein interactions or it may be involved in recruiting additional coregulator proteins to the promoter region.

Mol Endocrinol, April 2005, 19(4):833–842 835

NONGENOMIC ACTIONS OF ESTROGENS Evidence is accumulating that estrogens exert nongenomic actions—actions that are too rapid to be accounted for by the activation of RNA and protein synthesis. Nongenomic actions are a common property of steroid hormones and are frequently associated with the activation of various protein-kinase cascades (6). The nongenomic actions of 17␤-estradiol that have been reported include the mobilization of intracellular calcium (31), and the stimulation of adenylate cyclase activity and cAMP production (32, 33). Activation of the MAPK signaling pathway by 17␤-estradiol has been extensively studied in several cell types, including breast cancer (34), endothelial (35), bone (36, 37), and neuroblastoma (38) cells. 17␤-Estradiol also activates the phosphoinositol (PI) 3-kinase signaling pathway in endothelial (35), breast cancer (39), and liver (40) cells. Some authors have suggested that the nongenomic actions of estrogens are mediated through a subpopulation of the classical ERs, ER␣ and ER␤, that is located at the plasma membrane (33, 41). Other authors disagree (42). In endothelial cells, as in other cell types, ERs have been found in caveolae where they activate endothelial nitric oxide synthase (eNOS) through protein kinase-mediated phosphorylation (43–45). Caveolae are specialized membrane invaginations enriched in the scaffold protein caveolin-1. Caveolae facilitate signal transduction by providing a location for various signaling molecules (46). The only membrane receptors for estrogens that have been found are the classical ERs, ER␣ and ER␤. However, an isoform of ER␣ that is alternatively spliced (of molecular mass 46 kDa) and that has a truncated N-terminal domain has been identified in endothelial cells (47). ERs do not contain a trans-membrane domain, and the ability of ER␣ to associate with the plasma membrane may be due to palmitoylation of the receptor (47, 48). The plasma membrane ERs exist as functional dimers when activated by estrogens (49). It appears that the ligand binding domain of the classical ER␣, when targeted to the plasma membrane, is sufficient for mediating nongenomic actions of estrogens (50). However, other functional domains of the receptors might contribute to the magnitude of these actions by participating in various protein-protein interactions. It is likely that the interaction of ERs with various scaffold or signaling molecules facilitates the activity. ERs at the plasma membrane associate with the scaffold protein caveolin-1 (44, 51), and with a variety of proximal signaling molecules such as G proteins (33, 52), Src kinase, and ras (34, 53), the p85␣ regulatory subunit of PI3-kinase (54) and Shc (55). Furthermore, the scaffold protein MNAR promotes the interaction of ER␣ activated by 17␤-estradiol with Src kinase, leading to an increase in Src kinase activity,

836

Mol Endocrinol, April 2005, 19(4):833–842

and hence activation of the MAPK signaling pathway (56). Membrane ERs also activate membrane tyrosine kinase receptors in various cell types, including breast cancer cells. ER␣ activated by 17␤-estradiol interacts directly with the IGF-I receptor, leading to activation of the IGF-I receptor, and hence activation of the MAPK signaling pathway (57). ER␣ also interacts directly with ErbB2 (HER-2/neu) (58). In addition, ER␣ activated by 17␤-estradiol activates the epidermal growth factor (EGF) receptor by a mechanism that involves activation of G proteins, Src kinase, and matrix metalloproteinases, leading to an increase in MAPK and Akt (protein kinase B) activities (59). The studies described above suggest that the molecular mechanisms underlying the nongenomic actions of estrogens are specific for the cell type. The responses to estrogens may depend on a number of conditions, such as the set of signal transduction molecules and downstream targets present in the target cell, and thus, the responses are likely to be diverse.

NONGENOMIC-TO-GENOMIC SIGNALING: A NOVEL MODE OF REGULATING TRANSCRIPTION Signal transduction pathways may connect the nongenomic actions of estrogens to genomic responses. The functions of many transcription factors are regulated through protein kinase-mediated phosphorylation, and these transcription factors may thus be targets for nongenomic actions of estrogens, as illustrated in Fig. 1. This type of nongenomic-to-genomic signaling is a distinct mechanism by which ERs can regulate transcription at alternative response elements. This mechanism functions in addition to the ERE-independent genomic actions described above that involve protein-protein interactions in the nucleus. The transcription factors Elk-1 (60), C/EBP␤ (61), and CREB [cAMP response element (CRE) binding protein] (62) are all targets for phosphorylation by the MAPK signaling pathway. Two processes have been demonstrated in several cell types. These processes are the phosphorylation of Elk-1 that is induced by 17␤-estradiol, and the activation of serum response elements (SREs) by a mechanism that is dependent on ERs and that requires intact MAPK activity (55, 63–65). C/EBP␤ and CREB are phosphorylated after the MAPK signaling pathway has been activated by 17␤estradiol (65), and the phosphorylation of CREB leads to the expression of genes that contain CREs (65–67). CREB is a target for phosphorylation by the cAMP/ protein kinase A signaling pathway (68), in addition to its MAPK-mediated activation. Thus, CREB may also be activated by 17␤-estradiol through the stimulation of cAMP production. The transcriptional activity of the AP-1 transcription factor complex is regulated through MAPK-mediated phosphorylation (69). Activation of the MAPK signaling

Bjo¨rnstro¨m and Sjo¨berg • Minireview

pathway by 17␤-estradiol results in enhanced AP-1 DNA binding activity and transcriptional activation (40, 67, 70). In contrast, inhibition of the c-jun aminoterminal kinase signaling pathway by 17␤-estradiol results in repression of AP-1 activity (65). The latter observation agrees with previous results showing that the c-jun amino-terminal kinase signaling pathway, and hence AP-1 phosphorylation and transcriptional activation, are inhibited by other ligand-activated nuclear receptors (71, 72). The NF-␬B transcription factor complex is a target for phosphorylation by the Akt kinase (protein kinase B) (73, 74), and activation of the PI3-kinase/Akt signaling pathway by 17␤-estradiol leads to the expression of genes that contain NF-␬B binding sites (75). Tyrosine phosphorylation of the STAT family of transcription factors is required for their nuclear translocation and DNA binding activity (76). It has been suggested that additional serine phosphorylation modulates the maximal transcriptional activity of STATs (77). It has been reported that the phosphorylation of endogenous STAT3 and STAT5 on tyrosine and serine residues is induced by 17␤-estradiol in endothelial cells, and that STAT-dependent transcription, by a mechanism that is dependent on ERs and that requires intact MAPK, PI3-kinase, and Src kinase activities, is activated by 17␤-estradiol (78). These observations disagree with a previous study showing that the tyrosine phosphorylation and DNA binding activity of overexpressed STAT5 is decreased by ERs in COS-1 cells, both in the presence and in the absence of 17␤-estradiol (79). The discrepancies between these studies may be explained by differences in experimental conditions, such as the use of different types of cells and the analysis of endogenous or overexpressed proteins. It is important to note that ER␣ and ER␤ are also targets of phosphorylation by the MAPK signaling pathway (80, 81). Thus, nongenomic actions of estrogens may modulate the functions of ERs themselves, and in this way augment the classical mechanism of ER action. In addition, phosphorylation of the steroid receptor coactivator-3 is induced by 17␤-estradiol, leading to increased transcriptional potency of ERs (82). In the absence of estrogens, other signaling pathways, such as the IGF-I and EGF signaling pathways, can modulate the functions of ERs through phosphorylation of the receptors on certain residues (83), as illustrated in Fig. 1.

CONVERGENCE OF GENOMIC AND NONGENOMIC ACTIONS ON TARGET GENES The studies described above suggest that ERs can modulate the functions of transcription factors, and in this way regulate gene expression, by at least two distinct mechanisms. These mechanisms are proteinprotein interactions in the nucleus and activation of

Bjo¨rnstro¨m and Sjo¨berg • Minireview

signal transduction pathways at the plasma membrane. These mechanisms function in addition to the classical mechanism of ER action that is extensively reviewed elsewhere (1). Natural gene promoters are often regulated by a large number of transcription factors that bind to distinct regulatory sites. Thus, the possible convergence of genomic and nongenomic actions at multiple response elements provides an extremely fine degree of control for the regulation of transcription by ERs, as illustrated in Fig. 2. Cyclin D1 is a well-defined target for estrogens in breast cancer cells, and it is important for the progression of cells through the G1 phase of the cell cycle (84, 85). The cyclin D1 promoter is complex and contains binding sites for several transcription factors, but no ERE-like sequences have been identified (86). It has been suggested that activation of the cyclin D1 gene by 17␤-estradiol results mainly from the interaction of ERs with the Sp1 transcription factor at GC-rich promoter sequences (22), and with c-Jun/ATF-2 heterodimers at a variant CRE (10). However, it has also been reported that activation of the CRE by 17␤estradiol results from activation of the cAMP/protein kinase A signaling pathway (22), suggesting that nongenomic actions are involved. In addition, the cyclin D1 gene is activated by the association of ER␣ that

Mol Endocrinol, April 2005, 19(4):833–842 837

has been activated by 17␤-estradiol with Src kinase and the p85␣ regulatory subunit of PI3-kinase in breast cancer cells (39). In liver (40) and endometrial (87) cells, activation of the cyclin D1 gene by 17␤estradiol is mediated through the AP-1 binding site, and this activation requires intact MAPK activity (40). Furthermore, opposing actions of ER␣ and ER␤ at the cyclin D1 promoter have been observed (11), adding to the complexity of cyclin D1 gene regulation by ERs. The cyclin D1 promoter also contains binding sites for STAT5 and NF-␬B, and these may be targets for ERs through both genomic and nongenomic actions. Regulation of the immediate early c-fos gene by ERs is also mediated through both genomic and nongenomic actions. The interaction of ERs with the Sp1 transcription factor at GC-rich promoter sequences results in activation of the c-fos gene by 17␤-estradiol (21), and this activation also results from activation of the MAPK and PI3-kinase signaling pathways (64, 88). Thus, the nongenomic actions involve activation of both MAPK and PI3-kinase signaling pathways, which target the Elk-1 and the SRF transcription factors, respectively. These transcription factors bind to the SRE located in the c-fos promoter. The c-fos promoter also contains a sis-inducible element recognized by STATs and a cAMP response element, and these may

Fig. 2. Schematic Illustration of How Genomic and Nongenomic Actions of ERs on a Target Gene Promoter May Converge Nuclear E2-ER complexes bind to EREs, and to transcription factor complexes, e.g. AP-1, STATs, ATF-2 (activation transcription factor 2)/c-Jun, Sp1, and NF-␬B, that are bound to their cognate DNA binding sites. Membrane E2-ER complexes activate protein-kinase cascades, leading to phosphorylation (P) of target transcription factors, e.g. AP-1, STATs, Elk-1, SRF (serum response factor), CREB, and NF-␬B. The phosphorylation results in their transcriptional activation and/or in modulation of the transcriptional activities of ER-AP-1, ER-STAT, ER-Sp1, and ER-NF-␬B complexes at the promoter. Protein-kinase cascades also target ERs themselves and steroid receptor coactivator coactivators, resulting in enhanced transcriptional activity of ERs at EREs. The distinct actions of ERs at multiple response elements provide an extremely fine degree of control for the regulation of target gene transcription.

838

Mol Endocrinol, April 2005, 19(4):833–842

be targets for ERs through both genomic and nongenomic actions. Estrogens increase the clearance of LDL and they lower plasma LDL cholesterol levels in postmenopausal women, and these may be the mechanisms by which estrogens reduce the risk of cardiovascular disease (89). The LDL-R gene is activated by 17␤-estradiol in the liver, although the LDL-R promoter does not contain any ERE-like sequences (90). It has been reported that the interaction of ERs with the Sp1 transcription factor at GC-rich promoter sequences gives rise to activation of the LDL-R gene by 17␤-estradiol (20). In addition, tyrosine-kinase activity is required for the 17␤-estradiol-induced activation of the LDL-R gene (91), suggesting that nongenomic actions are involved. The tyrosine-kinase pathway may be connected with Sp1-dependent transcription. The LDL-R promoter also contains an SRE and this may be the target for ERs through nongenomic actions that involve activation of the MAPK and PI3-kinase signaling pathways. Some estrogen-responsive genes contain a combination of direct ER binding sites and binding sites to which ERs indirectly associate through tethering. One such gene is the vascular endothelial growth factor gene that contains a variant ERE that specifically binds ERs activated by 17␤-estradiol, and G/GC-rich sequences that bind ER-Sp1 and ER-Sp3 complexes

Bjo¨rnstro¨m and Sjo¨berg • Minireview

(92, 93). Furthermore, a number of genes that contain ERE half-sites in proximity to Sp1 binding sites, both of which must be occupied for maximal activation, are activated by 17␤-estradiol. Such genes include those for retinoic acid receptor ␣ (94), TGF␣ (95), and progesterone receptor (96). It is evident that the regulation of gene expression by ERs is a multifactorial process, involving both genomic and nongenomic actions that often converge at certain response elements located in the promoters of target genes. The final gene responses, however, may depend on a number of conditions such as the combination of transcription factors bound to a specific gene promoter, the cellular localization of ERs, the expression levels of various coregulator proteins and signal transduction components, and the nature of extracellular stimuli. These variables are highly specific for the cell type. Thus, estrogens may use different signaling pathways depending on the cellular context, and in this way evoke distinct gene responses in different types of target cells, as illustrated in Fig. 3.

RESPONSES TO SYNTHETIC LIGANDS ER actions at alternative response elements have agonist and antagonist profiles that differ from the pro-

Fig. 3. Schematic Illustration of How ERs May Regulate Transcription at a Putative Target Gene Promoter in Different Types of Target Cells The signaling pathways used, i.e. ER actions at EREs or ERE-independent genomic and nongenomic actions of ERs, depend on the cellular localization of ERs, and in this way, the magnitude of the gene response evoked by E2 may differ between target cells.

Bjo¨rnstro¨m and Sjo¨berg • Minireview

files of the classical mechanism of ER action. The different responses to synthetic ligands may enable the use of such compounds to dissociate between distinct signaling pathways, and hence between gene responses. It has been suggested that estrogen-responsive genes containing AP-1 binding sites contribute to the difference in response that particular tissues make to selective ER modulators (SERMs). These differences, it is suggested, arise through differences in the activation of ER␣ and ER␤ in the nucleus (97). Differential activation of ER␣-Sp1 and of ER␤-Sp1 complexes at GC-rich promoters takes place also in the presence of SERMs (98). More recent studies, however, suggest that some of the responses to SERMs are mediated through nongenomic actions, which subsequently lead to genomic responses (70, 99). It may be possible, therefore, to develop of novel tissue-specific drugs that modulate the function of ERs at specific cellular locations. It has been claimed that estren, a synthetic estrogen, exerts only nongenomic actions (50). However, estren also affects ER actions in the nucleus (70), and it activates transcription at EREs, although the transcriptional potency of estren is lower that of 17␤estradiol (100). The full estrogen antagonist ICI 182,780, on the other hand, is unable to exert nongenomic actions mediated through classical receptors, and this antagonist blocks completely the nongenomic actions of estrogens (40, 50, 55, 63–65, 67, 78). However, ICI 182,780 is a potent agonist to both ER␣ and ER␤ when the receptors are tethered to the AP-1 (13, 14, 97), Sp1 (98, 101), and STAT5 (29) transcription factors in the nucleus. Thus, not all signaling pathways are inhibited by this antagonist. CONCLUDING REMARKS It is evident that many genes are regulated by ERs and that these genes are of two types: those that contain EREs and those that do not. The latter genes contain binding sites for a variety of heterogeneous transcription factors. ER actions at alternative response elements enable a broader range of genes to be regulated than the range that can be regulated by the classical mechanism of ER action alone. It is important to note that the nongenomic actions of estrogens target many of the transcription factors, such as AP-1, that participate in protein-protein interactions with ERs in the nucleus. The possible convergence of genomic and nongenomic actions on target genes is an attractive mechanism by which ERs can finely regulate gene expression. The relative contributions of genomic and nongenomic actions to certain gene responses in vivo, however, remain to be elucidated. The molecular mechanisms underlying the effects of estrogens are likely to be specific for the cell type, and thus, the gene responses are likely to be diverse.

Mol Endocrinol, April 2005, 19(4):833–842 839

It appears that different functional domains of ERs are involved in different receptor actions. Thus, naturally occurring splice variants such as ER␤cx (102), ER␤␦3 (103), and the 46-kDa ER␣ (47), although unable to regulate transcription at EREs, may act through alternative signaling pathways in their target tissues. If this is the case, it will have important biological significance. Further knowledge about the molecular mechanisms by which ERs regulate transcription at alternative response elements may enable drug design to target specific receptor actions. Drugs that have been designed to differently affect ER actions at specific cellular locations in a manner that depends on the cell type would widely expand the pharmacological possibilities open to physicians. Acknowledgments Received December 7, 2004. Accepted January 25, 2005. Address all correspondence and requests for reprints to: Maria Sjo¨berg, Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden. E-mail: [email protected].

REFERENCES 1. Nilsson S, Makela S, Treuter E, Tujague M, Thomsen J, Andersson G, Enmark E, Pettersson K, Warner M, Gustafsson JA 2001 Mechanisms of estrogen action. Physiol Rev 81:1535–1565 2. Rosenfeld MG, Glass CK 2001 Coregulator codes of transcriptional regulation by nuclear receptors. J Biol Chem 276:36865–36868 3. O’Lone R, Frith MC, Karlsson EK, Hansen U 2004 Genomic targets of nuclear estrogen receptors. Mol Endocrinol 18:1859–1875 4. Gottlicher M, Heck S, Herrlich P 1998 Transcriptional cross-talk, the second mode of steroid hormone receptor action. J Mol Med 76:480–489 5. Vanacker JM, Pettersson K, Gustafsson JA, Laudet V 1999 Transcriptional targets shared by estrogen receptor-related receptors (ERRs) and estrogen receptor (ER) ␣, but not by ER␤. EMBO J 18:4270–4279 6. Losel R, Wehling M 2003 Nongenomic actions of steroid hormones. Nat Rev Mol Cell Biol 4:46–56 7. Gaub MP, Bellard M, Scheuer I, Chambon P, SassoneCorsi P 1990 Activation of the ovalbumin gene by the estrogen receptor involves the fos-jun complex. Cell 63:1267–1276 8. Umayahara Y, Kawamori R, Watada H, Imano E, Iwama N, Morishima T, Yamasaki Y, Kajimoto Y, Kamada T 1994 Estrogen regulation of the insulin-like growth factor I gene transcription involves an AP-1 enhancer. J Biol Chem 269:16433–16442 9. Webb P, Lopez GN, Uht RM, Kushner PJ 1995 Tamoxifen activation of the estrogen receptor/AP-1 pathway: potential origin for the cell-specific estrogen-like effects of antiestrogens. Mol Endocrinol 9:443–456 10. Sabbah M, Courilleau D, Mester J, Redeuilh G 1999 Estrogen induction of the cyclin D1 promoter: involvement of a cAMP response-like element. Proc Natl Acad Sci USA 96:11217–11222 11. Liu MM, Albanese C, Anderson CM, Hilty K, Webb P, Uht RM, Price Jr RH, Pestell RG, Kushner PJ 2002 Opposing action of estrogen receptors ␣ and ␤ on

840 Mol Endocrinol, April 2005, 19(4):833–842

12.

13.

14.

15.

16. 17.

18.

19. 20.

21.

22.

23. 24.

25. 26. 27.

28.

cyclin D1 gene expression. J Biol Chem 277: 24353–24360 Philips A, Teyssier C, Galtier F, Rivier-Covas C, Rey JM, Rochefort H, Chalbos D 1998 FRA-1 expression level modulates regulation of activator protein-1 activity by estradiol in breast cancer cells. Mol Endocrinol 12: 973–985 Jakacka M, Ito M, Weiss J, Chien PY, Gehm BD, Jameson JL 2001 Estrogen receptor binding to DNA is not required for its activity through the nonclassical AP1 pathway. J Biol Chem 276:13615–13621 Bjo¨rnstro¨m L, Sjo¨berg M 2002 Mutations in the estrogen receptor DNA-binding domain discriminate between the classical mechanism of action and cross-talk with Stat5b and activating protein 1 (AP-1). J Biol Chem 277:48479–48483 Schmitt M, Bausero P, Simoni P, Queuche D, Geoffroy V, Marschal C, Kempf J, Quirin-Stricker C 1995 Positive and negative effects of nuclear receptors on transcription activation by AP-1 of the human choline acetyltransferase proximal promoter. J Neurosci Res 40: 152–164 Kushner PJ, Agard DA, Greene GL, Scanlan TS, Shiau AK, Uht RM, Webb P 2000 Estrogen receptor pathways to AP-1. J Steroid Biochem Mol Biol 74:311–317 Webb P, Nguyen P, Valentine C, Lopez GN, Kwok GR, McInerney E, Katzenellenbogen BS, Enmark E, Gustafsson JA, Nilsson S, Kushner PJ 1999 The estrogen receptor enhances AP-1 activity by two distinct mechanisms with different requirements for receptor transactivation functions. Mol Endocrinol 13:1672–1685 Teyssier C, Belguise K, Galtier F, Chalbos D 2001 Characterization of the physical interaction between estrogen receptor ␣ and JUN proteins. J Biol Chem 276: 36361–36369 Porter W, Saville B, Hoivik D, Safe S 1997 Functional synergy between the transcription factor Sp1 and the estrogen receptor. Mol Endocrinol 11:1569–1580 Li C, Briggs MR, Ahlborn TE, Kraemer FB, Liu J 2001 Requirement of Sp1 and estrogen receptor ␣ interaction in 17␤-estradiol-mediated transcriptional activation of the low density lipoprotein receptor gene expression. Endocrinology 142:1546–1553 Duan R, Porter W, Safe S 1998 Estrogen-induced c-fos protooncogene expression in MCF-7 human breast cancer cells: role of estrogen receptor Sp1 complex formation. Endocrinology 139:1981–1990 Castro-Rivera E, Samudio I, Safe S 2001 Estrogen regulation of cyclin D1 gene expression in ZR-75 breast cancer cells involves multiple enhancer elements. J Biol Chem 276:30853–30861 Safe S 2001 Transcriptional activation of genes by 17 ␤-estradiol through estrogen receptor-Sp1 interactions. Vitam Horm 62:231–252 Ray A, Prefontaine KE, Ray P 1994 Down-modulation of interleukin-6 gene expression by 17 ␤-estradiol in the absence of high affinity DNA binding by the estrogen receptor. J Biol Chem 269:12940–12946 Stein B, Yang MX 1995 Repression of the interleukin-6 promoter by estrogen receptor is mediated by NF-␬B and C/EBP ␤. Mol Cell Biol 15:4971–4979 Blobel GA, Sieff CA, Orkin SH 1995 Ligand-dependent repression of the erythroid transcription factor GATA-1 by the estrogen receptor. Mol Cell Biol 15:3147–3153 Stoecklin E, Wissler M, Schaetzle D, Pfitzner E, Groner B 1999 Interactions in the transcriptional regulation exerted by Stat5 and by members of the steroid hormone receptor family. J Steroid Biochem Mol Biol 69:195–204 Faulds MH, Pettersson K, Gustafsson JA, Haldosen LA 2001 Cross-talk between ERs and signal transducer and activator of transcription 5 is E2 dependent and involves two functionally separate mechanisms. Mol Endocrinol 15:1929–1940

Bjo¨rnstro¨m and Sjo¨berg • Minireview

29. Bjo¨rnstro¨m L, Kilic E, Norman M, Parker MG, Sjo¨berg M 2001 Cross-talk between Stat5b and estrogen receptor-␣ and -␤ in mammary epithelial cells. J Mol Endocrinol 27:93–106 30. Heck S, Kullmann M, Gast A, Ponta H, Rahmsdorf HJ, Herrlich P, Cato AC 1994 A distinct modulating domain in glucocorticoid receptor monomers in the repression of activity of the transcription factor AP-1. EMBO J 13:4087–4095 31. 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 96:4686–4691 32. 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 91:8517–8521 33. 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 ER␤ expressed in Chinese hamster ovary cells. Mol Endocrinol 13:307–319 34. 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 15:1292–1300 35. 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 103:401–406 36. Endoh H, Sasaki H, Maruyama K, Takeyama K, Waga I, Shimizu T, Kato S, Kawashima H 1997 Rapid activation of MAP kinase by estrogen in the bone cell line. Biochem Biophys Res Commun 235:99–102 37. Jessop HL, Sjoberg M, Cheng MZ, Zaman G, WheelerJones CP, Lanyon LE 2001 Mechanical strain and estrogen activate estrogen receptor ␣ in bone cells. J Bone Miner Res 16:1045–1055 38. Watters JJ, Campbell JS, Cunningham MJ, Krebs EG, Dorsa DM 1997 Rapid membrane effects of steroids in neuroblastoma cells: effects of estrogen on mitogen activated protein kinase signalling cascade and c-fos immediate early gene transcription. Endocrinology 138: 4030–4033 39. 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 20:6050–6059 40. Marino M, Acconcia F, Bresciani F, Weisz A, Trentalance A 2002 Distinct nongenomic signal transduction pathways controlled by 17␤-estradiol regulate DNA synthesis and cyclin D(1) gene transcription in HepG2 cells. Mol Biol Cell 13:3720–3729 41. Pappas TC, Gametchu B, Watson CS 1995 Membrane estrogen receptors identified by multiple antibody labeling and impeded-ligand binding. FASEB J 9:404–410 42. Toran-Allerand CD, Guan X, MacLusky NJ, Horvath TL, Diano S, Singh M, Connolly Jr ES, Nethrapalli IS, Tinnikov AA 2002 ER-X: a novel, plasma membrane-associated, putative estrogen receptor that is regulated during development and after ischemic brain injury. J Neurosci 22:8391–8401 43. 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 263:257–262 44. 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:E44–E52

Bjo¨rnstro¨m and Sjo¨berg • Minireview

45. Chambliss KL, Yuhanna IS, Anderson RG, Mendelsohn ME, Shaul PW 2002 ER␤ has nongenomic action in caveolae. Mol Endocrinol 16:938–946 46. Shaul PW, Anderson RG 1998 Role of plasmalemmal caveolae in signal transduction. Am J Physiol 275: L843–L851 47. Li L, Haynes MP, Bender JR 2003 Plasma membrane localization and function of the estrogen receptor ␣ variant (ER46) in human endothelial cells. Proc Natl Acad Sci USA 100:4807–4812 48. Acconcia F, Ascenzi P, Fabozzi G, Visca P, Marino M 2004 S-Palmitoylation modulates human estrogen receptor-␣ functions. Biochem Biophys Res Commun 316:878–883 49. Razandi M, Pedram A, Merchenthaler I, Greene GL, Levin ER 2004 Plasma membrane estrogen receptors exist and functions as dimers. Mol Endocrinol 18: 2854–2865 50. Kousteni S, Bellido T, Plotkin LI, O’Brien 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 104:719–730 51. Razandi M, Oh P, Pedram A, Schnitzer J, Levin ER 2002 ERs associate with and regulate the production of caveolin: implications for signaling and cellular actions. Mol Endocrinol 16:100–115 52. Wyckoff MH, Chambliss KL, Mineo C, Yuhanna IS, Mendelsohn ME, Mumby SM, Shaul PW 2001 Plasma membrane estrogen receptors are coupled to endothelial nitric-oxide synthase through G␣(i). J Biol Chem 276:27071–27076 53. 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 17:2008–2018 54. Simoncini T, Hafezi-Moghadam A, Brazil DP, Ley K, Chin WW, Liao JK 2000 Interaction of oestrogen receptor with the regulatory subunit of phosphatidylinositol3-OH kinase. Nature 407:538–541 55. Song RX, McPherson RA, Adam L, Bao Y, Shupnik M, Kumar R, Santen RJ 2002 Linkage of rapid estrogen action to MAPK activation by ER␣-Shc association and Shc pathway activation. Mol Endocrinol 16:116–127 56. Wong CW, McNally C, Nickbarg E, Komm BS, Cheskis BJ 2002 Estrogen receptor-interacting protein that modulates its nongenomic activity-crosstalk with Src/ Erk phosphorylation cascade. Proc Natl Acad Sci USA 99:14783–14788 57. 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 275:18447–18453 58. Chung YL, Sheu ML, Yang SC, Lin CH, Yen SH 2002 Resistance to tamoxifen-induced apoptosis is associated with direct interaction between Her2/neu and cell membrane estrogen receptor in breast cancer. Int J Cancer 97:306–312 59. Razandi M, Pedram A, Park ST, Levin ER, Oh P, Schnitzer J 2003 Proximal events in signaling by plasma membrane estrogen receptors ERs associate with and regulate the production of caveolin: implications for signaling and cellular actions. J Biol Chem 278:2701–2712 60. Cruzalegui FH, Cano E, Treisman R 1999 ERK activation induces phosphorylation of Elk-1 at multiple S/T-P motifs to high stoichiometry. Oncogene 18:7948–7957 61. Buck M, Poli V, van der Geer P, Chojkier M, Hunter T 1999 Phosphorylation of rat serine 105 or mouse threonine 217 in C/EBP ␤ is required for hepatocyte proliferation induced by TGF ␣. Mol Cell 4:1087–1092

Mol Endocrinol, April 2005, 19(4):833–842 841

62. Bonni A, Brunet A, West AE, Datta SR, Takasu MA, Greenberg ME 1999 Cell survival promoted by the RasMAPK signaling pathway by transcription-dependent and -independent mechanisms. Science 286: 1358–1362 63. 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 276:27873–27880 64. 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 276:11590–11598 65. Kousteni S, Han L, Chen JR, Almeida M, Plotkin LI, Bellido T, Manolagas SC 2003 Kinase-mediated regulation of common transcription factors accounts for the bone-protective effects of sex steroids. J Clin Invest 111:1651–1664 66. Zhou Y, Watters JJ, Dorsa DM 1996 Estrogen rapidly induces the phosphorylation of the cAMP response element binding protein in rat brain. Endocrinology 137: 2163–2166 67. Dos Santos EG, Dieudonne MN, Pecquery R, Le Moal V, Giudicelli Y, Lacasa D 2002 Rapid nongenomic E2 effects on p42/p44 MAPK, activator protein-1, and cAMP response element binding protein in rat white adipocytes. Endocrinology 143:930–940 68. Shaywitz AJ, Greenberg ME 1999 CREB: a stimulusinduced transcription factor activated by a diverse array of extracellular signals. Annu Rev Biochem 68:821–861 69. Karin M 1995 The regulation of AP-1 activity by mitogen-activated protein kinases. J Biol Chem 270: 16483–16486 70. Bjo¨rnstro¨m L, Sjo¨berg M 2004 Estrogen receptor-dependent activation of AP-1 via non-genomic signalling. Nucl Recept 2:3 71. Caelles C, Gonzalez-Sancho JM, Munoz A 1997 Nuclear hormone receptor antagonism with AP-1 by inhibition of the JNK pathway. Genes Dev 11:3351–3364 72. Gonzalez MV, Jimenez B, Berciano MT, Gonzalez-Sancho JM, Caelles C, Lafarga M, Munoz A 2000 Glucocorticoids antagonize AP-1 by inhibiting the Activation/ phosphorylation of JNK without affecting its subcellular distribution. J Cell Biol 150:1199–1208 73. Romashkova JA, Makarov SS 1999 NF-␬B is a target of AKT in anti-apoptotic PDGF signalling. Nature 401: 86–90 74. Ozes ON, Mayo LD, Gustin JA, Pfeffer SR, Pfeffer LM, Donner DB 1999 NF-␬B activation by tumour necrosis factor requires the Akt serine-threonine kinase. Nature 401:82–85 75. Kawagoe J, Ohmichi M, Takahashi T, Ohshima C, Mabuchi S, Takahashi K, Igarashi H, Mori-Abe A, Saitoh M, Du B, Ohta T, Kimura A, Kyo S, Inoue M, Kurachi H 2003 Raloxifene inhibits estrogen-induced up-regulation of telomerase activity in a human breast cancer cell line. J Biol Chem 278:43363–43372 76. Darnell Jr JE 1997 STATs and gene regulation. Science 277:1630–1635 77. Decker T, Kovarik P 2000 Serine phosphorylation of STATs. Oncogene 19:2628–2637 78. Bjo¨rnstro¨m L, Sjo¨berg M 2002 Signal transducers and activators of transcription as downstream targets of nongenomic estrogen receptor actions. Mol Endocrinol 16:2202–2214 79. 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 13:330–343 80. Kato S, Endoh H, Masuhiro Y, Kitamoto T, Uchiyama S, Sasaki H, Masushige S, Gotoh Y, Nishida E, Kawashima

842 Mol Endocrinol, April 2005, 19(4):833–842

81.

82.

83.

84. 85.

86. 87.

88.

89.

90.

91.

92.

H, Metzger D, Chambon P 1995 Activation of the estrogen receptor through phosphorylation by mitogenactivated protein kinase. Science 270:1491–1494 Tremblay A, Tremblay GB, Labrie F, Giguere V 1999 Ligand-independent recruitment of SRC-1 to estrogen receptor ␤ through phosphorylation of activation function AF-1. Mol Cell 3:513–519 Wu RC, Qin J, Yi P, Wong J, Tsai SY, Tsai MJ, O’Malley BW 2004 Selective phosphorylations of the SRC-3/AIB1 coactivator integrate genomic reponses to multiple cellular signaling pathways. Mol Cell 15:937–949 Driggers PH, Segars JH 2002 Estrogen action and cytoplasmic signaling pathways. Part II: the role of growth factors and phosphorylation in estrogen signaling. Trends Endocrinol Metab 13:422–427 Planas-Silva MD, Weinberg RA 1997 Estrogen-dependent cyclin E-cdk2 activation through p21 redistribution. Mol Cell Biol 17:4059–4069 Prall OW, Sarcevic B, Musgrove EA, Watts CK, Sutherland RL 1997 Estrogen-induced activation of Cdk4 and Cdk2 during G1-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin-dependent kinase inhibitor association with cyclin E-Cdk2. J Biol Chem 272:10882–10894 Herber B, Truss M, Beato M, Muller R 1994 Inducible regulatory elements in the human cyclin D1 promoter. Oncogene 9:2105–2107 Shiozawa T, Miyamoto T, Kashima H, Nakayama K, Nikaido T, Konishi I 2004 Estrogen-induced proliferation of normal endometrial glandular cells is initiated by transcriptional activation of cyclin D1 via binding of c-Jun to an AP-1 sequence. Oncogene 23:8603–8610 Duan R, Xie W, Li X, McDougal A, Safe S 2002 Estrogen regulation of c-fos gene expression through phosphatidylinositol-3-kinase-dependent activation of serum response factor in MCF-7 breast cancer cells. Biochem Biophys Res Commun 294:384–394 Walsh BW, Schiff I, Rosner B, Greenberg L, Ravnikar V, Sacks FM 1991 Effects of postmenopausal estrogen replacement on the concentrations and metabolism of plasma lipoproteins. N Engl J Med 325:1196–1204 Croston GE, Milan LB, Marschke KB, Reichman M, Briggs MR 1997 Androgen receptor-mediated antagonism of estrogen-dependent low density lipoprotein receptor transcription in cultured hepatocytes. Endocrinology 138:3779–3786 Distefano E, Marino M, Gillette JA, Hanstein B, Pallottini V, Bruning J, Krone W, Trentalance A 2002 Role of tyrosine kinase signaling in estrogen-induced LDL receptor gene expression in HepG2 cells. Biochim Biophys Acta 1580:145–149 Mueller MD, Vigne JL, Minchenko A, Lebovic DI, Leitman DC, Taylor RN 2000 Regulation of vascular endothelial growth factor (VEGF) gene transcription by estrogen receptors ␣ and ␤. Proc Natl Acad Sci USA 97:10972–10977

Bjo¨rnstro¨m and Sjo¨berg • Minireview

93. Stoner M, Wang F, Wormke M, Nguyen T, Samudio I, Vyhlidal C, Marme D, Finkenzeller G, Safe S 2000 Inhibition of vascular endothelial growth factor expression in HEC1A endometrial cancer cells through interactions of estrogen receptor ␣ and Sp3 proteins. J Biol Chem 275:22769–22779 94. Rishi AK, Shao ZM, Baumann RG, Li XS, Sheikh MS, Kimura S, Bashirelahi N, Fontana JA 1995 Estradiol regulation of the human retinoic acid receptor ␣ gene in human breast carcinoma cells is mediated via an imperfect half-palindromic estrogen response element and Sp1 motifs. Cancer Res 55:4999–5006 95. Vyhlidal C, Samudio I, Kladde MP, Safe S 2000 Transcriptional activation of transforming growth factor ␣ by estradiol: requirement for both a GC-rich site and an estrogen response element half-site. J Mol Endocrinol 24:329–338 96. Petz LN, Nardulli AM 2000 Sp1 binding sites and an estrogen response element half-site are involved in regulation of the human progesterone receptor A promoter. Mol Endocrinol 14:972–985 97. Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Kushner PJ, Scanlan TS 1997 Differential ligand activation of estrogen receptors ER␣ and ER␤ at AP1 sites. Science 277:1508–1510 98. Saville B, Wormke M, Wang F, Nguyen T, Enmark E, Kuiper G, Gustafsson JA, Safe S 2000 Ligand-, cell-, and estrogen receptor subtype (␣/␤)-dependent activation at GC-rich (Sp1) promoter elements. J Biol Chem 275:5379–5387 99. Singleton DW, Feng Y, Burd CJ, Khan SA 2003 Nongenomic activity and subsequent c-fos induction by estrogen receptor ligands are not sufficient to promote deoxyribonucleic acid synthesis in human endometrial adenocarcinoma cells. Endocrinology 144:121–128 100. Moverare S, Dahllund J, Andersson N, Islander U, Carlsten H, Gustafsson JA, Nilsson S, Ohlsson C 2003 Estren is a selective estrogen receptor modulator with transcriptional activity. Mol Pharmacol 64:1428–1433 101. Zou A, Marschke KB, Arnold KE, Berger EM, Fitzgerald P, Mais DE, Allegretto EA 1999 Estrogen receptor ␤ activates the human retinoic acid receptor ␣-1 promoter in response to tamoxifen and other estrogen receptor antagonists, but not in response to estrogen. Mol Endocrinol 13:418–430 102. Ogawa S, Inoue S, Watanabe T, Orimo A, Hosoi T, Ouchi Y, Muramatsu M 1998 Molecular cloning and characterization of human estrogen receptor ␤cx: a potential inhibitor of estrogen action in human. Nucleic Acids Res 26:3505–3512 103. Price Jr RH, Butler CA, Webb P, Uht R, Kushner P, Handa RJ 2001 A splice variant of estrogen receptor ␤ missing exon 3 displays altered subnuclear localization and capacity for transcriptional activation. Endocrinology 142:2039–2049

Molecular Endocrinology is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community.