Insight into the mechanisms of action of estrogen receptor β in the breast, prostate, colon, and CNS

in Journal of Molecular Endocrinology
Authors:
Prasenjit DeyDepartment of Biology and Biochemistry, Department of Biosciences and Nutrition, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Blvd, Science and Engineering Research Center Bldg 545, Houston, Texas 77204-5056, USA

Search for other papers by Prasenjit Dey in
Current site
Google Scholar
PubMed
Close
,
Rodrigo P A BarrosDepartment of Biology and Biochemistry, Department of Biosciences and Nutrition, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Blvd, Science and Engineering Research Center Bldg 545, Houston, Texas 77204-5056, USA

Search for other papers by Rodrigo P A Barros in
Current site
Google Scholar
PubMed
Close
,
Margaret WarnerDepartment of Biology and Biochemistry, Department of Biosciences and Nutrition, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Blvd, Science and Engineering Research Center Bldg 545, Houston, Texas 77204-5056, USA

Search for other papers by Margaret Warner in
Current site
Google Scholar
PubMed
Close
,
Anders StrömDepartment of Biology and Biochemistry, Department of Biosciences and Nutrition, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Blvd, Science and Engineering Research Center Bldg 545, Houston, Texas 77204-5056, USA

Search for other papers by Anders Ström in
Current site
Google Scholar
PubMed
Close
, and
Jan-Åke GustafssonDepartment of Biology and Biochemistry, Department of Biosciences and Nutrition, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Blvd, Science and Engineering Research Center Bldg 545, Houston, Texas 77204-5056, USA
Department of Biology and Biochemistry, Department of Biosciences and Nutrition, Center for Nuclear Receptors and Cell Signaling, University of Houston, 3605 Cullen Blvd, Science and Engineering Research Center Bldg 545, Houston, Texas 77204-5056, USA

Search for other papers by Jan-Åke Gustafsson in
Current site
Google Scholar
PubMed
Close
View More View Less

Free access

Estrogen and its receptors (ERs) influence many biological processes in physiology and pathology in men and women. ERs are involved in the etiology and/or progression of cancers of the prostate, breast, uterus, ovary, colon, lung, stomach, and malignancies of the immune system. In estrogen-sensitive malignancies, ERβ usually is a tumor suppressor and ERα is an oncogene. ERβ regulates genes in several key pathways including tumor suppression (p53, PTEN); metabolism (PI3K); survival (Akt); proliferation pathways (p45Skp2, cMyc, and cyclin E); cell-cycle arresting factors (p21WAF1, cyclin-dependent kinase inhibitor 1 (CDKN1A)), p27Kip1, and cyclin-dependent kinases (CDKs); protection from reactive oxygen species, glutathione peroxidase. Because they are activated by small molecules, ERs are excellent targets for pharmaceuticals. ERα antagonists have been used for many years in the treatment of breast cancer and more recently pharmaceutical companies have produced agonists which are very selective for ERα or ERβ. ERβ agonists are being considered for preventing progression of cancer, treatment of anxiety and depression, as anti-inflammatory agents and as agents, which prevent or reduce the severity of neurodegenerative diseases.

Abstract

Estrogen and its receptors (ERs) influence many biological processes in physiology and pathology in men and women. ERs are involved in the etiology and/or progression of cancers of the prostate, breast, uterus, ovary, colon, lung, stomach, and malignancies of the immune system. In estrogen-sensitive malignancies, ERβ usually is a tumor suppressor and ERα is an oncogene. ERβ regulates genes in several key pathways including tumor suppression (p53, PTEN); metabolism (PI3K); survival (Akt); proliferation pathways (p45Skp2, cMyc, and cyclin E); cell-cycle arresting factors (p21WAF1, cyclin-dependent kinase inhibitor 1 (CDKN1A)), p27Kip1, and cyclin-dependent kinases (CDKs); protection from reactive oxygen species, glutathione peroxidase. Because they are activated by small molecules, ERs are excellent targets for pharmaceuticals. ERα antagonists have been used for many years in the treatment of breast cancer and more recently pharmaceutical companies have produced agonists which are very selective for ERα or ERβ. ERβ agonists are being considered for preventing progression of cancer, treatment of anxiety and depression, as anti-inflammatory agents and as agents, which prevent or reduce the severity of neurodegenerative diseases.

Introduction

In addition to their well-known role in growth and differentiation of the breast, ovary and uterus in females, and testes and prostate in males, estrogen is also required for proper functioning of the cardiovascular, immune, gastrointestinal, musculoskeletal, and nervous systems as well as skin. The most active estrogen in the body is 17β-estradiol (E2), which is synthesized in the granulosa cells of the ovary, Leydig cells of the testis (Stocco 2012), adipose tissue (Rice et al. 2012), and brain (Boon et al. 2010).

The classical actions of estrogen are mainly mediated via the two nuclear estrogen receptors (ERs), ERα and ERβ, both of which bind to E2 with high affinity in the low nM range. Two metabolites of E2, estrone (E1) and estriol (E3), circulate at high levels at certain phases in the menstrual cycle and during pregnancy (Gruber et al. 2002). E1 and E3 have been thought to be the inactive metabolites of E2, but E3 has significant effects on the immune system (Zang et al. 2002, Zhou et al. 2011) and a closer examination of the physiological functions of these two steroids is warranted.

In addition to estrogens, which are synthesized from testosterone and androstenedione through the action of the enzyme aromatase, there is another class of estrogens which do not have an aromatic A-ring. These are 5-androstene-3β,17β-diol (A5diol), synthesized from the adrenal steroid dehydroepiandrosterone, and 5α-androstane-3β,17β-diol (3β-Adiol), synthesized from the hormone 5α-dihydrotestosterone (DHT). Estrogenic actions of A5diol (Shao et al. 1975) and 3β-Adiol (Stewart et al. 1977) have been known for many years and their very rapid metabolism in the prostate (Isaacs et al. 1980) and pituitary (Guiraud et al. 1979) was reported. However, it was not until the discovery of ERβ that the estrogenic actions of these steroids were understood (Weihua et al. 2002a). Unlike E2, A5diol and 3β-Adiol do not circulate at high concentrations and serve more paracrine and autocrine functions.

ERα and ERβ are transcribed from two genes (ESR1 and ESR2 respectively) that are located on different chromosomes (Gosden et al. 1986, Enmark et al. 1997). ERα was discovered more than 50 years ago by Elwood Jensen and the gene was cloned in 1985 (Walter et al. 1985). ERβ was discovered in 1996 by cross-hybridization of a consensus DNA-binding domain (DBD) probe in a rat prostate cDNA library (Kuiper et al. 1996). ERα protein composed of 595 amino acids (White et al. 1987) is a bit longer than the 530 amino acid-long ERβ. ERα and ERβ are architecturally similar with six regions (A–F) in the primary amino acid sequence, an arrangement found in all members of the nuclear receptor superfamily (Carson-Jurica et al. 1990). Overall there are three functional domains: N-terminal domain (NTD), DNA binding domain (DBD), and ligand-binding domain (LBD). The two ERs share ∼97% similarity in their DBD and 59% in LBD, whereas the NTD is merely 16% similar (Fig. 1; Pettersson & Gustafsson 2001). Although the differences in the LBD are small, they are significant enough to influence the shape of the ligand–binding pocket and that ligands with unique binding affinities for the each receptor have been synthesized (for review see Nilsson et al. (2011)). These ligands are now referred to as specific ER modulators (SERMs). ERβ binds better than ERα to genistein, a phytoestrogen (Kuiper et al. 1998), which has traditionally been regarded as a health-promoting and anti-cancer agent. Two years after the discovery of ERβ, another splice variant was discovered and named ERβcx/2 (Ogawa et al. 1998). Soon after other splice variants – ERβ4, and ERβ5 – were discovered. ERβ4 were identified from a testis library and ERβ5 was identified from the MDA-MB-435 cell line (Moore et al. 1998). All the splice variants have a truncated C-terminus with a unique stretch of amino acids fused to the C-terminus. Because of the truncated LBD, these forms have lost their ability to bind estrogens (Leung et al. 2006b) and so far no ligands have been found for these variants.

Figure 1
Figure 1

Schematic of the structure of the estrogen receptor (ER). The ER is composed of three main domains: the N-terminal domain (NTD), which contains the activation function 1, the DNA-binding domain (DBD), and the ligand-binding domain (LBD), containing the activation function 2. The DBDs of ERα and ERβ are highly conserved and the NTDs are the least conserved. The NTD of ERα is shorter than that of ERβ.

Citation: Journal of Molecular Endocrinology 51, 3; 10.1530/JME-13-0150

In addition to the classical ER-binding site on DNA known as the estrogen response element (ERE), ERs can also be recruited to the response elements on DNA without themselves binding to DNA. Instead, they are tethered to other transcription factors (activator protein 1 (AP1), Sp1, and nuclear factor κB (NFκB); Webb et al. 1999) and influence the transcription of genes normally regulated by these rather ubiquitous transcription factors. ERα and ERβ can have opposite effects at these sites: ERβ-tamoxifen acting through AP1 can activate target genes, whereas ERα-tamoxifen may cause repression of the target genes (Webb et al. 2003).

The action of ERβ at AP1 sites or EREs is determined by the presence of coactivators. In the absence of E2, the co-activator Tip60 enhances ERβ transcriptional activity at AP1 sites, but represses it at EREs. ERβ-selective ligands abolish the repressive actions of ERβ at ERE, while the antagonist, ICI 182 780, enhances the transcription at AP1 sites. In addition to being a key player in reparation of double stranded breaks in DNA (Sun et al. 2010), Tip60 is an interesting coactivator in the relationship between ERβ and AR: TIP60 is an AR coactivator (reviewed in Culig & Santer (2012) and a facilitator of AR transport into the nucleus (Shiota et al. 2010)). If in the absence of E2, ERβ activation at AP1 sites causes proliferation, then the breasts of postmenopausal women should be proliferating but they are not (Cheng et al. 2013), and ERβ could increase proliferation in the prostate but it does not (Attia & Ederveen 2012). Clearly a more detailed analysis of the expression of coactivators in the breast and prostate is needed.

Recent genomic landscaping of ERα and ERβ binding sites has revealed that while ERα is predominantly found at ERE elements (Carroll et al. 2005), whereas ERβ is mostly found at AP1 sites (Zhao et al. 2010). This difference in DNA interaction has revealed a very important difference between the two ERs: the DBD is less important for transcriptional activity in ERβ than it is in ERα. The important consequence of this finding is that although removal of one zinc finger of ERα creates an inactive protein, the same mutation leaves a functional ERβ (Price et al. 2001). Thus, it is not surprising that a mouse in which the second zinc finger (ERβ exon 3) was deleted is normal except for the ability to ovulate (Antal et al. 2008).

Mechanisms of ER action

Ligand-dependent activation of ER

ERs primarily reside in an inactive state in the cytoplasm in complex with heat-shock protein (Hsp) 50, 70, and 90, which stabilizes the receptor (reviewed in Sanchez (2012)). In the classical model of estrogen action, Hsp dissociates and the DBD of the receptor becomes unveiled when the ligand-binding pocket is occupied. Upon binding to DNA, the receptor causes recruitment of the p160 family of co-regulators such as steroid receptor coactivators (SRCs) or silencing mediator of retinoid and thyroid receptor (SMRT/NCOR2) (Xu et al. 2009). The structure of the ligand that enters the ligand-binding pocket influences the conformation adopted by the receptor and thus either coactivators or corepressors are recruited (Paige et al. 1999, Pike et al. 1999).

Ligand-independent mechanism of action

ERs can also be activated by ligand-independent mechanisms via cross-talk with other signaling pathways. Both receptors can be activated via phosphorylation by MAPK and phosphatidylinositide 3-kinase (PI3K; Miller et al. 2011, Riggio et al. 2012, McGlynn et al. 2013). Furthermore, epidermal growth factor receptor (EGFR) and insulin-like growth factor receptor (IGFR), upon activation by their cognate peptide ligands, can activate the serine/threonine kinases, ERK and Akt, which in turn catalyze the phosphorylation and activation of ERs. Posttranscriptional modifications of ERs have been nicely reviewed recently (Le Romancer et al. 2011). Activation of ERα by kinases is thought to play an important role in breast cancer (reviewed in Thomas & Gustafsson (2011)).

Physiological and pathophysiological actions of ERs

The two ERs have distinct functions and act antagonistically in multiple pathways: ERα plays an important role in growth and proliferation (Maggiolini et al. 2001, Ewan et al. 2005, Sun et al. 2006, Teng et al. 2008) while ERβ exerts anti-proliferative, differentiative, and apoptotic actions in breast (Lazennec et al. 2001, Paruthiyil et al. 2004, Strom et al. 2004), prostate (Cheng et al. 2004, Pravettoni et al. 2007, McPherson et al. 2010, Dey et al. 2012), and colon cells (Hartman et al. 2009, Bielecki et al. 2011). Both receptors are required for the maintenance of glucose homeostasis with ERβ predominating in the skeletal muscle and ERα in the adipose tissue (Barros et al. 2009, Barros & Gustafsson 2011).

Breast and ER

In the normal mammary gland, ER is essential for branching morphogenesis and in ERα−/− mice there is a very rudimentary ductal tree (Couse & Korach 1999). In the adult gland, in both rodents (Saji et al. 2000) and women (Cheng et al. 2013), ERα is expressed in <10% of mammary epithelial cells. ERβ is expressed in 70–80% of the mammary epithelial cells and is also expressed in the stroma and immune cells resident in the breast (Clarke et al. 1997, Speirs et al. 2002, Li et al. 2010). ERβ−/− mice revealed that ERβ is not required for ductal growth of the breast but is required for terminal differentiation and for maintaining the organization and differentiation of epithelial tissue (Forster et al. 2002).

Breast cancer and ER

According to the statistical evidence (Siegel et al. 2013), one in eight women will develop breast cancer and the 5-year survival for metastatic breast cancer is 25%. Although initiation and progression of breast cancer involve a complex interplay of many environmental and genetic factors, estrogen plays a very central role (Hankinson et al. 2004). The involvement of estrogen signaling in breast cancer was understood as early as in the 1880s, when surgeons in the USA and Germany were able to treat breast cancer patients by oophorectomy (Love & Philips 2002). Prolonged exposure to estrogen is a well-established risk factor for breast cancer in postmenopausal women (Darbre & Charles 2010, Fernandez & Russo 2010). The combination of multiple gene mutations (BRCA1, TP53, and ERBB2) together with the expression of ERα determines the final course and progression of the tumor (Njiaju & Olopade 2012). About 70% of breast cancers are ERα-positive and respond to anti-estrogen therapies such as tamoxifen, raloxifene, or other SERMs and aromatase inhibitors (Musgrove & Sutherland 2009). However, one-third of the patients treated with tamoxifen become resistant to treatment. The reason for the development of tamoxifen resistance is not yet clear. Loss of ERα expression or mutations in ERα cannot account for all resistance as these occur only in ∼15% of patients (Gutierrez et al. 2005).

The involvement of ERβ in breast cancer is still being investigated. As reviewed by Murphy & Leygue (2012), several studies have demonstrated that ERβ expression in breast cancer, irrespective of presence of ERα, is associated with increased response to endocrine therapy, but others have not found this (Murphy & Watson 2006). The functions of ERβ splice variants in breast cancer are another unresolved issue. ERβ2 (ERβcx) is a C-terminally spliced form of ERβ discovered by Ogawa et al. (1998). Some clinical studies have associated the expression of ERβ2 with poor prognosis and decreased survival of breast cancer patients (Saji et al. 2002); others have found the opposite (Sugiura et al. 2007). Some found that ERβ2 is not indicative of tamoxifen sensitivity (Esslimani-Sahla et al. 2004) while others have found that it is (Vinayagam et al. 2007). Clearly, more work needs to be done in order to clarify these discrepancies. One suggestion which might be helpful is the use of samples bigger than the traditional biopsy. This is because there is heterogeneity in any breast cancer, with many stages of the disease in each sample and this can only be seen in samples of larger size. Thus some regions of the cancer express ERβ and some express ERβcx and these foci of expression can be missed if small samples are used.

Breast cancer cell lines MCF7, T47D, and MDA231 have been used extensively to study the functions of ERα and ERβ. One of the curious results of these studies is that when ER-negative breast cancer cells, MDA-MB-231, are engineered to express ERα or ERβ, both receptors can inhibit proliferation and invasion. The difference is that ERα requires the presence of ligand for its actions but ERβ does not (Lazennec et al. 2001). Studies in the T47D breast cancer cell line revealed that in the presence of E2, ERα regulates expression of cell cycle genes such as c-MYC (MYC), cyclin E, cyclin D, and p45Skp2 (Strom et al. 2004). ERα interacts with a distal half-ERE and an AP1 site in the enhancer of c-myc and transcriptionally upregulates c-myc expression (Dubik & Shiu 1992, Wang et al. 2011). Inhibition of ERα with tamoxifen causes a decrease in cyclin D1 and c-myc expression, which represses downstream targets such as Bcl2 and increases cell death (Butt et al. 2005, Nehra et al. 2010). Very recently, it has been shown that in breast cancer cell lines, ERβ augmented the antiproliferative effects of tamoxifen (Lattrich et al. 2013).

Many studies show that loss of ERβ expression occurs frequently in ductal breast, partly because of promoter hypermethylation. Esslimani-Shaha et al. ascribe the progression of the malignancy and tamoxifen resistance to the decreased expression of ERβ (Shaaban et al. 2003, Esslimani-Sahla et al. 2004, Hopp et al. 2004). However, there are also studies which show high ERβ expression in ductal cancer and in these studies, ERβ expression is associated with poor prognosis (Markey et al. 2009, Kim et al. 2012). The reasons for the reported differences in the contribution of ERβ to breast cancer progression will no doubt be explained as more studies are done and the antibodies used become more standardized.

Despite studies showing beneficial effects of ERβ in breast cancer, a novel mechanism through which ERβ might contribute to aggressiveness in Her2-positive breast cancer has been described by Chen et al. According to this study conducted in several breast cancer cell lines, ERβ expression is associated with high expression of interleukin 8, a cytokine which increases tumor angiogenesis and metastasis (Chen et al. 2011). If it is the relative expression of ERs (ERβ1/ERβ2/ERα) which determines the overall effect of ERβ in breast cancer, expression of all three receptors and their localization must be measured in order to assess the role of ERs in the prognosis of breast cancer.

Prostate and ER

E2 via ERα is required for the development and branching morphogenesis of the prostate (Bosland 2000, Taplin & Ho 2001, Omoto et al. 2005). However, in the adult mouse prostate there is very little ERα expression and most of it is in the stromal compartment. ERβ, on the other hand, is abundantly expressed in the epithelium of the adult mouse and human prostate. It is also expressed in the stroma and the infiltrating immune cells (Schulze & Barrack 1987, Brenner et al. 1990, Schulze & Claus 1990, Prins & Birch 1997, Tsurusaki et al. 2003). The circulating level of E2 in males is similar to that in postmenopausal women, i.e. 30 pg/ml (Taylor et al. 2012). However, in the prostate, the most abundant estrogen is not E2 but 3β-Adiol (Weihua et al. 2002b).

Prostate cancer and ER

Studies on ERβ−/− mice showed prostatic hyperplasia in 5-month-old mice and PIN lesions in mice older than 1 year (Weihua et al. 2002b). This was the first in a series of observations made in mouse models and cancer cell lines, demonstrating an anti-proliferative function of ERβ. In vitro models for studying the role of ERβ in prostate cancer are very artificial because prostate cancer cell lines do not express or express very low levels of ERβ. It is always questionable as to whether results obtained by overexpression of ERβ in a cancer cell line can be extrapolated to clinical situations. Nevertheless, interesting information about the antiproliferative effects of ERβ has been obtained when ERβ was introduced into prostate cancer cell lines. In 2008, Hurtado et al. (2008) found that overexpression of ERβ in LNCaP cells caused G1 cell cycle arrest. More recently we found that when ERβ is expressed in PC3 and 22RV1 cells, there is a decrease in the expression of proliferative and oncogenic factors (p45Skp2, c-myc, and cyclin E) and an increase in cell-cycle arresting factors (p21WAF1, cyclin-dependent kinase inhibitor 1 (CDKN1A), p27Kip1, and cyclin-dependent kinase (Dey et al. 2012)). Thus ERβ can inhibit proliferation in both androgen-dependent (LNCaP and 22RV1) and androgen-independent (PC3) prostate cancer cell lines.

In DU145 cells, ERβ is activated by ICI 182 780, an antiestrogen. Treatment of DU145 cells with ICI 182 780 stabilized the tumor suppressor, Krüppel-like zinc finger transcription factor 5 (KLF5), thereby increasing the expression of the antiproliferative transcription factor, forkhead box protein O1 (FOXO1). The increase in transcription of the FOXO1 gene involves the recruitment of KLF5-ERβ-CBP (CREB-binding protein) complex to the promoter of FOXO1. The increase in transcription can be opposed by E2, which promotes proteasome-dependent degradation of KLF5 (Nakajima et al. 2011). This is one possible mechanism through which ERβ can oppose E2-induced proliferation. Another anti-proliferative route used by ERβ was shown by Leung et al. (2006a) in DU145 cells. In these cells, ICI 182 780 in the presence of ERβ opposed the action of NFκB which led to decreased cell proliferation. In a recent study we have found that ERβ promotes apoptosis in prostate by the upregulation of p53-upregulated modulator of apoptosis (PUMA) via transcriptional activation of FOXO3a (Dey et al. 2013).

Results obtained from prostate cancer cell lines point to an important role of ERβ in repressing key oncogenes (PI3K, p45Skp2, c-myc, and cyclin E; Dey et al. 2012) and increasing expression of antiproliferative genes like PTEN (Lindberg et al. 2011), FOXO3, KLF5, p21WAF1, CDKN1A, and p27Kip1; why has this not translated to a more enthusiastic embrace of ERβ as a target in prostate cancer by urologists? The problem probably lies in the confusion over the phenotype of ERβ−/− mice and the questions raised in laboratories where no prostatic phenotype was observed in ERβ−/− mice (Antal et al. 2008).

As discussed above, exon 3 deletion does not create an ERβ−/− mouse, so some of the controversies are related to inappropriate strategy for creating ERβ−/− mice. A key clinical study published in 2008 should have dispelled all doubts about the beneficial role of ERβ in protecting against prostate cancer. This study showed that expression of the fusion gene TMPRSS2:ERG, which is thought to be responsible for up to 80% of prostate cancers, is repressed by ERβ. The study was a meta-analysis of biopsy samples collected from 455 prostate cancer patients in the Swedish Watchful Waiting cohort (1987–1999) and the USA-based Physicians Health Study cohort (1983–2003) and the effect of ERβ was confirmed in NCI-H660 cancer cells, where the inhibition of TMPRSS2:ERG fusion is prevented by ERβ but not ERα agonists (Setlur et al. 2008). Transmembrane protease, serine 2 (TMPRSS2) is an AR-regulated gene (Paoloni-Giacobino et al. 1997). In certain forms of prostate cancer, there is a chromosomal translocation followed by a gene fusion which results in TMPRSS2 insertion into the promoter region of the erythroblast transformation-specific (ETS) family of transcription factors, ERG and ETV1. Expression of this fusion gene leads to castration resistant prostate cancer (Tomlins et al. 2005, Yu et al. 2010). To date although there are safe and effective ERβ agonists (Malamas et al. 2004), no ERβ agonist has been tested in prostate cancer patients. A similar remarkable effect of ERβ agonists has been observed in the TRAMP mouse (transgenic mouse model for prostate cancer; Gingrich et al. 1996). In this mouse model of prostate cancer, dietary genistein, is an ERβ-dependent pathway, reduced the incidence of prostate cancer and the severity of the invasive cancer was increased in ERβ−/− mice (Slusarz et al. 2012).

One other confounding factor occurring in prostate cancer is the expression of the ERβ splice variant ERβ2. As rodents do not express this splice variant, conclusions about its role in prostate cancer have come from its measurement in prostate cancers (Fujimura et al. 2001, Lee et al. 2013) and from overexpression in cell lines (Dey et al. 2012). So far, there is agreement that the expression of ERβ2 in prostate cancer is associated with a poor prognosis (Leung et al. 2010). The reason for the negative effects of ERβ2 is not clear but if it has actions, which are opposite to those of ERβ (Dey et al. 2012), then it might promote survival under the conditions of hypoxia. One of the mechanisms through which ERβ exerts antiproliferative effects is its facilitation of HIF1α degradation (Mak et al. 2013). The effects of ERβ2 on the stability of HIF1α remain to be determined.

Understanding androgen metabolism in prostate cancer

As is the case with metabolites of estrogen, metabolites of DHT may play important physiological roles. DHT is more a potent androgen than testosterone, so inhibition of the enzyme which catalyzes the conversion of testosterone to DHT should reduce AR activity. However, the use of 5α-reductase inhibitors showed an important negative consequence of inhibition of this enzyme. In the Prostate Cancer Prevention Trial (PCPT), patients treated with the 5α-reductase inhibitor, finasteride, showed a decrease of 25% in prostate cancer prevalence, but a higher rate of Gleason grade (7–10) (37%) over the placebo-treated group (22.2%). A possible explanation for this aberration is that the inhibition of synthesis of DHT also results in the loss of the DHT metabolite, 3β-Adiol, a prostatic ligand of ERβ (Imamov et al. 2004, Briganti 2009). More recently, the long-term treatment of prostate cancer with finasteride has revealed that this drug has no effect on prostate cancer (McCarthy 2013). Treatment of mice with 3β-Adiol resulted in a decreased cell proliferation in the ventral prostate of wild type but not in ERβ knockout animals (Weihua et al. 2002a).

Additional beneficial effects of ERβ in prostate cancer

In addition to its antiproliferative actions, ERβ is also anti-migratory and is an inhibitor of epithelial to mesenchymal transition (EMT). In one of the first cell-based studies demonstrating an anti-migratory effect of ERβ, adenovirus-mediated delivery of ERβ led to a strong decrease in the invasiveness of DU145 cells compared with the control transfected cells (Cheng et al. 2004). Metastasis of ERβ-expressing xenografts of DU145 was blocked by treatment with 3β-Adiol (Guerini et al. 2005). Seven years later, Grubisha et al. (2012) investigated the mechanism through which ERβ, activated by 3β-Adiol, opposes cell migration and motility of DU145 cells. They found that this anti-migratory effect of ERβ is opposed by transforming growth factor β (TGFβ) signal-derived reactive oxygen species (ROS) such as H2O2. Cyclooxygenase 2 (Cox2) in the stromal cells is responsible for the production of H2O2, which, in turn, prevents binding of ERβ to the promoter of E-cadherin. Agents neutralizing H2O2 and Cox2 could reverse the inhibition of ERβ by ROS (Fig. 2; Grubisha et al. 2012).

Figure 2
Figure 2

Pathways regulated by ERβ. ERβ upon binding to its ligand 3β-Adiol/8β-VE2/DPN gets activated and transcriptionally upregulates downstream target genes such as FOXO3a and PHD2. The target genes carry ERβ binding elements such as ERE or AP1-ERE-half-site upstream or downstream of the transcriptional start site. The activated FOXO3a in turn transcriptionally upregulates PUMA, p21, and p27. PHD2 inhibits HIF1α by prolyl hydroxylation of the oxygen-dependent degradation domain (ODDD), which targets HIF1α proteins for proteasomal degradation by promoting their interaction with von Hippel–Lindau (VHL). ERβ also inhibits proliferation of genes such as c-MYC and p45Skp2 by an as yet unknown mechanism. Moreover, some of the EMT and bone metastasis genes, such as β-catenin, SLUG/SNAIL and TWIST, are opposed by ERβ.

Citation: Journal of Molecular Endocrinology 51, 3; 10.1530/JME-13-0150

Mak et al. (2010) showed that ERβ promotes an epithelial phenotype and opposes EMT in both androgen-dependent and androgen-independent cell lines. The authors suggested that TGFβ and hypoxia may cause a decrease in ERβ expression and promote migration and invasion, features of EMT. The paper highlights the point that the Hif1α/VEGF-A/Snail pathway, required for the genesis of EMT, is opposed by ERβ (Mak et al. 2010). Reduced EMT and migration occurs in xenographs of PC3 and 22Rv1 cells when they are induced to overexpress ERβ. We found that ERβ represses the metastasis factor Runx2, the EMT factor Slug and β-catenin (Dey et al. 2012). ERβ is expressed in BPH and low Gleason grade prostate cancer, but is reduced above Gleason grade 3 (Yang et al. 2007, Mak et al. 2010). However, there is a report that ERβ expression re-emerges in metastatic prostate cancer (Lai et al. 2004).

Colon

The Women's Health Initiative Study in 2002 not only showed an increased risk of stroke and heart disease in women on hormone replacement therapy, but it also showed a reduced risk of colon cancer (Rossouw et al. 2002). In agreement with this finding, a study by Cleveland et al. showed that disruption of estrogen signaling increased intestinal neoplasia in Apc (Min/+) mice. Furthermore, it appears that ERβ is important for the tumor suppressive effect, while disruption of ERα signaling has no effect (Cleveland et al. 2009). Several other studies support the tumor suppressive effect of ERβ in colon. One study showed increased induction of mucin-depleted foci in ERβ−/− mice (Saleiro et al. 2010), while another study showed that ERβ−/− mice developed more colitis-associated neoplasia (Saleiro et al. 2012). In agreement with this finding ERβ has been shown to stimulate anti-inflammatory networks in colon cancer cell lines (Edvardsson et al. 2011). Expression of ERβ in colon cancer is correlated to prognosis where a decreased expression is found in higher grade and larger tumors (Konstantinopoulos et al. 2003, Rudolph et al. 2012), and the level is inversely correlated with more advanced Dukes' staging (Jassam et al. 2005). A role of ERα in colon cancer has not been found so far. A hint that it may be involved in progression comes from a study by Armstrong et al. using azoxymethane to induce colon cancer. The study showed that ERβ expression decreases in colonic epithelial cells as the cancer progresses, while expression of ERα increases, suggesting a possible role of ERα in colorectal cancer progression (Armstrong et al. 2013). From the phenotype of ERβ−/− mice, ERβ in the colon appears to decrease proliferation and increase apoptosis, suggesting a tumor suppressive and tumor preventive effect (Wada-Hiraike et al. 2006). ERβ has also been shown to change the micro RNA pool in human colorectal cancer cells (Edvardsson et al. 2013) and to regulate miR-135b and mismatch repair gene expressions in colorectal cells (He et al. 2012).

The incidence of colorectal cancer is much lower in Asian countries than in the Western world, an effect that possibly can be explained by a high dietary intake of phytoestrogens in the form of soy (Lechner et al. 2005). The protective effects of soy are thought to be mediated by binding of genistein, the main phytoestrogen in soy, to ERβ. Genistein binds to ERβ with 20-fold higher affinity than ERα (Kuiper et al. 1997). Genistein is also a tyrosine kinase inhibitor and some of its actions in the colon may be elicited through its inhibition of tyrosine kinase (Wang et al. 2012). Using the Apc (Min/+) mouse model of colorectal cancer, Javid et al. (2005) showed that coumestrol, another phytoestrogen with no tyrosine kinase inhibitory activity, as well as E2 inhibit intestinal tumorigenesis in ovariectomized mice. Both clinical studies and animal experiments suggest that phytoestrogens and E2 have a preventive effect on colorectal cancer by acting through ERβ.

CNS

ERβ and neuro-protection

ERs have been shown to affect the brain in several ways. ERα is responsible for imprinting of the male brain during embryonic development (Naftolin 2008), while ERβ is involved in cortical layering and migration of interneurons in the fetal period and neonatal period (Wang et al. 2003, Fan et al. 2006).

ERβ agonists can modulate tryptophan hydroxylase 1 mRNA expression in the raphe nucleus and cause antidepressant-like effects (Clark et al. 2012, Suzuki et al. 2013). The mechanism involves increase of serotonergic activity by regulating synthesis of serotonin. In addition, serotonin receptors are regulated by estrogens in an ERβ-dependent way (Osterlund 2010). Both ERα and ERβ appear to have protective roles in the brain, where E2 attenuates secondary injury via ERα in the rat brain following subarachnoid hemorrhage (Raval et al. 2013). In the paraventricular nucleus (PVN) and rostroventrolateral medulla, ERβ protects against aldosterone/salt-induced hypertension in female rats by inhibiting mineralocorticoid receptor induced increases in ROS (Xue et al. 2013). Ma et al. (2013) have shown that dietary soy and isoflavones by reducing VEGF are neuroprotective in cerebral ischemia. Not much is known about the ERβ splice variants in the brain with the exception of ERβ5, which is increased in glioma cells (Li et al. 2013) where its expression is induced by hypoxia and HIF1α. Furthermore, in these cells, ERβ5 reduced cell proliferation and AKT signaling and increased PTEN expression and may play a role in repression of tumor growth.

ERβ has profound effects on neuroinflammation and ERβ-selective ligands are very effective in the treatment of neuroinflammation in mouse models (Kumar et al. 2013, Wu et al. 2013). In addition to multiple sclerosis, neuroinflammation plays a role in the etiology and/or progression also of other CNS diseases. These include Parkinson's disease (Koutsilieri et al. 2013), Alzheimer's disease (Fuster-Matanzo et al. 2013), bipolar disorder (Stertz et al. 2013), and autism (Theoharides et al. 2013). In neurodegenerative diseases, one of the mechanisms of progression and damage to healthy neurons in the vicinity of lesions is overactive microglia. Microglia are the policeman of the brain sensing and kill diseased cells and foreign invaders by secreting powerful cytokines (Benjamins 2013). In this process, neighboring normal cells can be damaged. ERβ but not ERα is expressed in microglia and ERβ-selective ligands can prevent microglial activation and secretion of cytokines (Wu et al. 2013). The anti-inflammatory effects of ERβ are not confined to microglia. ERβ is also expressed in T cells, in particular in Th1 cells, where it regulates indolamine-2,3-dioxygenase (IOD), the rate-limiting step in tryptophan degradation (Harden & Egilmez 2012). This enzyme causes cell death by depleting tryptophan and by cytotoxicity of the tryptophan metabolite, kynurenine. IOD is an important mediator of immune tolerance via inhibition of Th1 responses and its loss is associated with autoimmune disease (McGaha et al. 2012). The immunosuppressive property of IOD is essential for immune tolerance and in inhibition of Th1 cells that are responsible for experimental autoimmune encephalitis, a mouse model of multiple sclerosis.

ERs and feeding behavior

E2 has long been recognized as an anorexigenic factor, but the mechanisms behind this effect are still being investigated (Barros & Gustafsson 2011, Eckel 2011). ERα and/or ERβ are expressed in the regions of the hypothalamus that regulate feeding and satiety (Roepke 2009; Table 1).

Table 1

Feeding peptides regulated by estrogen receptors (ERs) in the hypothalamus

Nucleus of the hypothalamusER expressedPeptide expressedFunction
Arcuate nucleusERαCocaine- and amphetamine-regulated transcripts Anorexigenic
α-Melanocyte-stimulating hormoneAnorexigenic
Arcuate nucleusERα and ERβNeuropeptide YOrexigenic
Agouti gene-related proteinOrexigenic
Lateral NoneMelanin-concentrating hormone (MCH) Orexigenic
Orexin neuronsOrexigenic
Dorsomedial ERα and ERβNeuropeptide YOrexigenic
Cocaine- and amphetamine-regulated transcriptsAnorexigenic
Paraventricular nucleusERβUrocortinAnorexigenic (repressed by ERβ)
VentromedialERαBrain-derived neurotrophic factorAnorexigenic
UrocortinAnorexigenic

In the arcuate nucleus, ERα is expressed in the pro-opiomelanocortin neurons which secrete two anorexigenic peptides, α-melanocyte-stimulating hormone, and cocaine- and amphetamine-regulated transcripts (CART). Both ERα and ERβ are expressed in the neuropeptide Y (NPY) neurons (Roepke 2009, de Souza et al. 2011) which secrete NPY and agouti gene-related protein, two orexigenic peptides (Pillot et al. 2011).

In the lateral hypothalamus, there are two types of orexigenic neurons, the melanin-concentrating hormone neurons and the orexin neurons (Horvath 2006, Nahon 2006). Neither of the ERs have been identified in either of these neurons, and it is thought that the regulation of feeding and satiety by ERs in the lateral hypothalamus is indirect (Muschamp & Hull 2007). The dorsomedial hypothalamus expresses both ERα and ERβ and secretes both the orexigenic peptide, NPY, and the anorexigenic peptide CART. In the PVN, which responds to glucose and lipids, ERβ is abundant and decreases the expression of urocortin, a potent anorexigenic peptide (Haeger et al. 2006). Stimulation of this nucleus increases food intake (Horvath 2006, Roepke 2009). The ventromedial hypothalamus is mainly regulated by ERα and is an important inhibitor of satiety and food intake (Xu et al. 2003). Inhibition of ERα signaling in this nucleus is associated with hyperphagia, obesity, decreased glucose tolerance, and reduced energy expenditure (Musatov et al. 2007). Brain-derived neurotrophic factor (Gotoh et al. 2013) and urocortin (Chen et al. 2012) are the anorexigenic peptides secreted by neurons of the ventromedial hypothalamus.

Although the precise role of each ER in the regulation of feeding and satiety is still under investigation, it seems that ERα plays a dominant role in anorexia and ERβ in the stimulation of feeding.

Conclusions

This review covers recent advances in estrogen signaling with a focus on ERβ. Since its discovery in 1996, ERβ has changed our understanding of the mechanisms of estrogen action. It appears that the two ERs often have an antagonistic or yin/yang relationship, e.g. in control of cell proliferation where ERα is pro-proliferative whereas ERβ is anti-proliferative. Accordingly, ERβ is a promising potential target for anti-cancer drugs. Furthermore, ERβ is an important mediator of estrogen action in the CNS, again offering promise as a drug target in the treatment of, e.g. multiple sclerosis, Parkinson's disease, and eating disorders.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the review reported.

Funding

This work was supported by The Cancer Prevention and Research Institute of Texas (CPRIT) grant numbers HIRP100680 and RP110444, the Texas Emerging Technology Fund under Agreement no. 300-9-1958, the Robert A Welch Foundation (E-0004), and the Swedish Cancer Fund.

References

  • Antal MC, Krust A, Chambon P & Mark M 2008 Sterility and absence of histopathological defects in nonreproductive organs of a mouse ERβ-null mutant. PNAS 105 24332438. (doi:10.1073/pnas.0712029105).

    • Search Google Scholar
    • Export Citation
  • Armstrong CM, Billimek AR, Allred KF, Sturino JM, Weeks BR & Allred CD 2013 A novel shift in estrogen receptor expression occurs as estradiol suppresses inflammation-associated colon tumor formation. Endocrine-Related Cancer 20 515525. (doi:10.1530/ERC-12-0308).

    • Search Google Scholar
    • Export Citation
  • Attia DM & Ederveen AG 2012 Opposing roles of ERα and ERβ in the genesis and progression of adenocarcinoma in the rat ventral prostate. Prostate 72 10131022. (doi:10.1002/pros.21507).

    • Search Google Scholar
    • Export Citation
  • Barros RP & Gustafsson JA 2011 Estrogen receptors and the metabolic network. Cell Metabolism 14 289299. (doi:10.1016/j.cmet.2011.08.005).

    • Search Google Scholar
    • Export Citation
  • Barros RP, Gabbi C, Morani A, Warner M & Gustafsson JA 2009 Participation of ERα and ERβ in glucose homeostasis in skeletal muscle and white adipose tissue. American Journal of Physiology. Endocrinology and Metabolism 297 E124E133. (doi:10.1152/ajpendo.00189.2009).

    • Search Google Scholar
    • Export Citation
  • Benjamins JA 2013 Direct effects of secretory products of immune cells on neurons and glia. Journal of Neurological Sciences 333 3036. (doi:10.1016/j.jns.2013.06.001).

    • Search Google Scholar
    • Export Citation
  • Bielecki A, Roberts J, Mehta R & Raju J 2011 Estrogen receptor-β mediates the inhibition of DLD-1 human colon adenocarcinoma cells by soy isoflavones. Nutrition and Cancer 63 139150. (doi:10.1080/01635581.2010.516867).

    • Search Google Scholar
    • Export Citation
  • Boon WC, Chow JD & Simpson ER 2010 The multiple roles of estrogens and the enzyme aromatase. Progress in Brain Research 181 209232. (doi:10.1016/S0079-6123(08)81012-6).

    • Search Google Scholar
    • Export Citation
  • Bosland MC 2000 The role of steroid hormones in prostate carcinogenesis. Journal of the National Cancer Institute. Monographs 2000 (27) 3966. (doi:10.1093/oxfordjournals.jncimonographs.a024244).

    • Search Google Scholar
    • Export Citation
  • Brenner RM, West NB & McClellan MC 1990 Estrogen and progestin receptors in the reproductive tract of male and female primates. Biology of Reproduction 42 1119. (doi:10.1095/biolreprod42.1.11).

    • Search Google Scholar
    • Export Citation
  • Briganti A 2009 Oestrogens and prostate cancer: novel concepts about an old issue. European Urology 55 543545. (doi:10.1016/j.eururo.2008.12.004).

    • Search Google Scholar
    • Export Citation
  • Butt AJ, McNeil CM, Musgrove EA & Sutherland RL 2005 Downstream targets of growth factor and oestrogen signalling and endocrine resistance: the potential roles of c-Myc, cyclin D1 and cyclin E. Endocrine-Related Cancer 12 (Suppl 1) S47S59. (doi:10.1677/erc.1.00993).

    • Search Google Scholar
    • Export Citation
  • Carroll JS, Liu XS, Brodsky AS, Li W, Meyer CA, Szary AJ, Eeckhoute J, Shao W, Hestermann EV & Geistlinger TR et al. 2005 Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1. Cell 122 3343. (doi:10.1016/j.cell.2005.05.008).

    • Search Google Scholar
    • Export Citation
  • Carson-Jurica MA, Schrader WT & O'Malley BW 1990 Steroid receptor family: structure and functions. Endocrine Reviews 11 201220. (doi:10.1210/edrv-11-2-201).

    • Search Google Scholar
    • Export Citation
  • Chen Y, Chen L, Li JY, Mukaida N, Wang Q, Yang C, Yin WJ, Zeng XH, Jin W & Shao ZM 2011 ERβ and PEA3 co-activate IL-8 expression and promote the invasion of breast cancer cells. Cancer Biology & Therapy 11 497511. (doi:10.4161/cbt.11.5.14667).

    • Search Google Scholar
    • Export Citation
  • Chen P, Hover CV, Lindberg D & Li C 2012 Central urocortin 3 and type 2 corticotropin-releasing factor receptor in the regulation of energy homeostasis: critical involvement of the ventromedial hypothalamus. Frontiers in Endocrinology 3 180. (doi:10.3389/fendo.2012.00180).

    • Search Google Scholar
    • Export Citation
  • Cheng J, Lee EJ, Madison LD & Lazennec G 2004 Expression of estrogen receptor β in prostate carcinoma cells inhibits invasion and proliferation and triggers apoptosis. FEBS Letters 566 169172. (doi:10.1016/j.febslet.2004.04.025).

    • Search Google Scholar
    • Export Citation
  • Cheng G, Butler R, Warner M, Gustafsson JA, Wilczek B & Landgren BM 2013 Effects of short-term estradiol and norethindrone acetate treatment on the breasts of normal postmenopausal women. Menopause 20 496503. (doi:10.1097/GME.0b013e318276c4ea).

    • Search Google Scholar
    • Export Citation
  • Clark JA, Alves S, Gundlah C, Rocha B, Birzin ET, Cai SJ, Flick R, Hayes E, Ho K & Warrier S et al. 2012 Selective estrogen receptor-β (SERM-β) compounds modulate raphe nuclei tryptophan hydroxylase-1 (TPH-1) mRNA expression and cause antidepressant-like effects in the forced swim test. Neuropharmacology 63 10511063. (doi:10.1016/j.neuropharm.2012.07.004).

    • Search Google Scholar
    • Export Citation
  • Clarke RB, Howell A, Potten CS & Anderson E 1997 Dissociation between steroid receptor expression and cell proliferation in the human breast. Cancer Research 57 49874991.

    • Search Google Scholar
    • Export Citation
  • Cleveland AG, Oikarinen SI, Bynote KK, Marttinen M, Rafter JJ, Gustafsson JA, Roy SK, Pitot HC, Korach KS & Lubahn DB et al. 2009 Disruption of estrogen receptor signaling enhances intestinal neoplasia in Apc(Min/+) mice. Carcinogenesis 30 15811590. (doi:10.1093/carcin/bgp132).

    • Search Google Scholar
    • Export Citation
  • Couse JF & Korach KS 1999 Estrogen receptor null mice: what have we learned and where will they lead us? Endocrine Reviews 20 358417. (doi:10.1210/er.20.3.358).

    • Search Google Scholar
    • Export Citation
  • Culig Z & Santer FR 2012 Androgen receptor co-activators in the regulation of cellular events in prostate cancer. World Journal of Urology 30 297302. (doi:10.1007/s00345-011-0797-6).

    • Search Google Scholar
    • Export Citation
  • Darbre PD & Charles AK 2010 Environmental oestrogens and breast cancer: evidence for combined involvement of dietary, household and cosmetic xenoestrogens. Anticancer Research 30 815827.

    • Search Google Scholar
    • Export Citation
  • Dey P, Jonsson P, Hartman J, Williams C, Strom A & Gustafsson JA 2012 Estrogen receptors β1 and β2 have opposing roles in regulating proliferation and bone metastasis genes in the prostate cancer cell line PC3. Molecular Endocrinology 26 19912003. (doi:10.1210/me.2012.1227).

    • Search Google Scholar
    • Export Citation
  • Dey P, Ström A & Gustafsson J-Å Estrogen receptor β upregulates FOXO3a and causes induction of apoptosis through PUMA in prostate cancer Oncogene 2013 [in press] doi:10.1038/onc.2013.384).

    • Search Google Scholar
    • Export Citation
  • Dubik D & Shiu RP 1992 Mechanism of estrogen activation of c-myc oncogene expression. Oncogene 7 15871594.

  • Eckel LA 2011 The ovarian hormone estradiol plays a crucial role in the control of food intake in females. Physiology & Behavior 104 517524. (doi:10.1016/j.physbeh.2011.04.014).

    • Search Google Scholar
    • Export Citation
  • Edvardsson K, Strom A, Jonsson P, Gustafsson JA & Williams C 2011 Estrogen receptor β induces antiinflammatory and antitumorigenic networks in colon cancer cells. Molecular Endocrinology 25 969979. (doi:10.1210/me.2010-0452).

    • Search Google Scholar
    • Export Citation
  • Edvardsson K, Nguyen-Vu T, Kalasekar SM, Ponten F, Gustafsson JA & Williams C 2013 Estrogen receptor β expression induces changes in the microRNA pool in human colon cancer cells. Carcinogenesis 34 14311441. (doi:10.1093/carcin/bgt067).

    • Search Google Scholar
    • Export Citation
  • Enmark E, Pelto-Huikko M, Grandien K, Lagercrantz S, Lagercrantz J, Fried G, Nordenskjold M & Gustafsson JA 1997 Human estrogen receptor β-gene structure, chromosomal localization, and expression pattern. Journal of Clinical Endocrinology and Metabolism 82 42584265. (doi:10.1210/jc.82.12.4258).

    • Search Google Scholar
    • Export Citation
  • Esslimani-Sahla M, Simony-Lafontaine J, Kramar A, Lavaill R, Mollevi C, Warner M, Gustafsson JA & Rochefort H 2004 Estrogen receptor β (ERβ) level but not its ERβcx variant helps to predict tamoxifen resistance in breast cancer. Clinical Cancer Research 10 57695776. (doi:10.1158/1078-0432.CCR-04-0389).

    • Search Google Scholar
    • Export Citation
  • Ewan KB, Oketch-Rabah HA, Ravani SA, Shyamala G, Moses HL & Barcellos-Hoff MH 2005 Proliferation of estrogen receptor-α-positive mammary epithelial cells is restrained by transforming growth factor-β1 in adult mice. American Journal of Pathology 167 409417. (doi:10.1016/S0002-9440(10)62985-9).

    • Search Google Scholar
    • Export Citation
  • Fan X, Warner M & Gustafsson JA 2006 Estrogen receptor β expression in the embryonic brain regulates development of calretinin-immunoreactive GABAergic interneurons. PNAS 103 1933819343. (doi:10.1073/pnas.0609663103).

    • Search Google Scholar
    • Export Citation
  • Fernandez SV & Russo J 2010 Estrogen and xenoestrogens in breast cancer. Toxicologic Pathology 38 110122. (doi:10.1177/0192623309354108).

    • Search Google Scholar
    • Export Citation
  • Forster C, Makela S, Warri A, Kietz S, Becker D, Hultenby K, Warner M & Gustafsson JA 2002 Involvement of estrogen receptor β in terminal differentiation of mammary gland epithelium. PNAS 99 1557815583. (doi:10.1073/pnas.192561299).

    • Search Google Scholar
    • Export Citation
  • Fujimura T, Takahashi S, Urano T, Ogawa S, Ouchi Y, Kitamura T, Muramatsu M & Inoue S 2001 Differential expression of estrogen receptor β (ERβ) and its C-terminal truncated splice variant ERβcx as prognostic predictors in human prostatic cancer. Biochemical and Biophysical Research Communications 289 692699. (doi:10.1006/bbrc.2001.6038).

    • Search Google Scholar
    • Export Citation
  • Fuster-Matanzo A, Llorens-Martin M, Hernandez F & Avila J 2013 Role of neuroinflammation in adult neurogenesis and Alzheimer disease: therapeutic approaches. Mediators of Inflammation 2013 260925. (doi:10.1155/2013/260925).

    • Search Google Scholar
    • Export Citation
  • Gingrich JR, Barrios RJ, Morton RA, Boyce BF, DeMayo FJ, Finegold MJ, Angelopoulou R, Rosen JM & Greenberg NM 1996 Metastatic prostate cancer in a transgenic mouse. Cancer Research 56 40964102.

    • Search Google Scholar
    • Export Citation
  • Gosden JR, Middleton PG & Rout D 1986 Localization of the human oestrogen receptor gene to chromosome 6q24→q27 by in situ hybridization. Cytogenetics and Cell Genetics 43 218220. (doi:10.1159/000132325).

    • Search Google Scholar
    • Export Citation
  • Gotoh K, Masaki T, Chiba S, Ando H, Fujiwara K, Shimasaki T, Mitsutomi K, Katsuragi I, Kakuma T & Sakata T et al. 2013 Brain-derived neurotrophic factor, corticotropin-releasing factor, and hypothalamic neuronal histamine interact to regulate feeding behavior. Journal of Neurochemistry 125 588598. (doi:10.1111/jnc.12213).

    • Search Google Scholar
    • Export Citation
  • Gruber CJ, Tschugguel W, Schneeberger C & Huber JC 2002 Production and actions of estrogens. New England Journal of Medicine 346 340352. (doi:10.1056/NEJMra000471).

    • Search Google Scholar
    • Export Citation
  • Grubisha MJ, Cifuentes ME, Hammes SR & Defranco DB 2012 A local paracrine and endocrine network involving TGFβ, Cox-2, ROS, and estrogen receptor β influences reactive stromal cell regulation of prostate cancer cell motility. Molecular Endocrinology 26 940954. (doi:10.1210/me.2011-1371).

    • Search Google Scholar
    • Export Citation
  • Guerini V, Sau D, Scaccianoce E, Rusmini P, Ciana P, Maggi A, Martini PG, Katzenellenbogen BS, Martini L & Motta M et al. 2005 The androgen derivative 5α-androstane-3β,17β-diol inhibits prostate cancer cell migration through activation of the estrogen receptor β subtype. Cancer Research 65 54455453. (doi:10.1158/0008-5472.CAN-04-1941).

    • Search Google Scholar
    • Export Citation
  • Guiraud JM, Morfin R, Ducouret B, Samperez S & Jouan P 1979 Pituitary metabolism of 5α-androstane-3β-17β-diol: intense and rapid conversion into 5α-androstane-3β,6α,17β-triol and 5α-androstane-3β,7α, 17β-triol. Steroids 34 241248. (doi:10.1016/0039-128X(79)90076-X).

    • Search Google Scholar
    • Export Citation
  • Gutierrez MC, Detre S, Johnston S, Mohsin SK, Shou J, Allred DC, Schiff R, Osborne CK & Dowsett M 2005 Molecular changes in tamoxifen-resistant breast cancer: relationship between estrogen receptor, HER-2, and p38 mitogen-activated protein kinase. Journal of Clinical Oncology 23 24692476. (doi:10.1200/JCO.2005.01.172).

    • Search Google Scholar
    • Export Citation
  • Haeger P, Andres ME, Forray MI, Daza C, Araneda S & Gysling K 2006 Estrogen receptors α and β differentially regulate the transcriptional activity of the Urocortin gene. Journal of Neuroscience 26 49084916. (doi:10.1523/JNEUROSCI.0476-06.2006).

    • Search Google Scholar
    • Export Citation
  • Hankinson SE, Colditz GA & Willett WC 2004 Towards an integrated model for breast cancer etiology: the lifelong interplay of genes, lifestyle, and hormones. Breast Cancer Research 6 213218. (doi:10.1186/bcr921).

    • Search Google Scholar
    • Export Citation
  • Harden JL & Egilmez NK 2012 Indoleamine 2,3-dioxygenase and dendritic cell tolerogenicity. Immunological Investigations 41 738764. (doi:10.3109/08820139.2012.676122).

    • Search Google Scholar
    • Export Citation
  • Hartman J, Edvardsson K, Lindberg K, Zhao C, Williams C, Strom A & Gustafsson JA 2009 Tumor repressive functions of estrogen receptor β in SW480 colon cancer cells. Cancer Research 69 61006106. (doi:10.1158/0008-5472.CAN-09-0506).

    • Search Google Scholar
    • Export Citation
  • He YQ, Sheng JQ, Ling XL, Fu L, Jin P, Yen L & Rao J 2012 Estradiol regulates miR-135b and mismatch repair gene expressions via estrogen receptor-β in colorectal cells. Experimental & Molecular Medicine 44 723732. (doi:10.3858/emm.2012.44.12.079).

    • Search Google Scholar
    • Export Citation
  • Hopp TA, Weiss HL, Parra IS, Cui Y, Osborne CK & Fuqua SA 2004 Low levels of estrogen receptor β protein predict resistance to tamoxifen therapy in breast cancer. Clinical Cancer Research 10 74907499. (doi:10.1158/1078-0432.CCR-04-1114).

    • Search Google Scholar
    • Export Citation
  • Horvath TL 2006 Synaptic plasticity in energy balance regulation. Obesity 14 (Suppl 5) 228S233S. (doi:10.1038/oby.2006.314).

  • Hurtado A, Pinos T, Barbosa-Desongles A, Lopez-Aviles S, Barquinero J, Petriz J, Santamaria-Martinez A, Morote J, de Torres I & Bellmunt J et al. 2008 Estrogen receptor β displays cell cycle-dependent expression and regulates the G1 phase through a non-genomic mechanism in prostate carcinoma cells. Cellular Oncology 30 349365. (doi:10.3233/CLO-2009-0495).

    • Search Google Scholar
    • Export Citation
  • Imamov O, Lopatkin NA & Gustafsson JA 2004 Estrogen receptor β in prostate cancer. New England Journal of Medicine 351 27732774. (doi:10.1056/NEJM200412233512622).

    • Search Google Scholar
    • Export Citation
  • Isaacs JT, McDermott IR & Coffey DS 1980 The identification and characterization of the C19O3 steroid metabolites of 5α-androstane-3β,17β-diol produced by the canine prostate: 5α-androstane-3β,6α,17β-triol and 5α-androstane-3β,7α,17β-triol. Steroids 35 139156. (doi:10.1016/0039-128X(80)90099-9).

    • Search Google Scholar
    • Export Citation
  • Jassam N, Bell SM, Speirs V & Quirke P 2005 Loss of expression of oestrogen receptor β in colon cancer and its association with Dukes' staging. Oncology Reports 14 1721.

    • Search Google Scholar
    • Export Citation
  • Javid SH, Moran AE, Carothers AM, Redston M & Bertagnolli MM 2005 Modulation of tumor formation and intestinal cell migration by estrogens in the Apc(Min/+) mouse model of colorectal cancer. Carcinogenesis 26 587595. (doi:10.1093/carcin/bgh346).

    • Search Google Scholar
    • Export Citation
  • Kim TJ, Lee A, Choi YJ, Song BJ, Yim HW & Kang CS 2012 Prognostic significance of high expression of ER-β in surgically treated ER-positive breast cancer following endocrine therapy. Journal of Breast Cancer 15 7986. (doi:10.4048/jbc.2012.15.1.79).

    • Search Google Scholar
    • Export Citation
  • Konstantinopoulos PA, Kominea A, Vandoros G, Sykiotis GP, Andricopoulos P, Varakis I, Sotiropoulou-Bonikou G & Papavassiliou AG 2003 Oestrogen receptor β (ERβ) is abundantly expressed in normal colonic mucosa, but declines in colon adenocarcinoma paralleling the tumour's dedifferentiation. European Journal of Cancer 39 12511258. (doi:10.1016/S0959-8049(03)00239-9).

    • Search Google Scholar
    • Export Citation
  • Koutsilieri E, Lutz MB & Scheller C 2013 Autoimmunity, dendritic cells and relevance for Parkinson's disease. Journal of Neural Transmission 120 7581. (doi:10.1007/s00702-012-0842-7).

    • Search Google Scholar
    • Export Citation
  • Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S & Gustafsson JA 1996 Cloning of a novel receptor expressed in rat prostate and ovary. PNAS 93 59255930. (doi:10.1073/pnas.93.12.5925).

    • Search Google Scholar
    • Export Citation
  • Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S & Gustafsson JA 1997 Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors α and β. Endocrinology 138 863870. (doi:10.1210/en.138.3.863).

    • Search Google Scholar
    • Export Citation
  • Kuiper GG, Lemmen JG, Carlsson B, Corton JC, Safe SH, van der Saag PT, van der Burg B & Gustafsson JA 1998 Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor β. Endocrinology 139 42524263. (doi:10.1210/en.139.10.4252).

    • Search Google Scholar
    • Export Citation
  • Kumar S, Patel R, Moore S, Crawford DK, Suwanna N, Mangiardi M & Tiwari-Woodruff SK 2013 Estrogen receptor β ligand therapy activates PI3K/Akt/mTOR signaling in oligodendrocytes and promotes remyelination in a mouse model of multiple sclerosis. Neurobiological Disorders 56 131144. (doi:10.1016/j.nbd.2013.04.005).

    • Search Google Scholar
    • Export Citation
  • Lai JS, Brown LG, True LD, Hawley SJ, Etzioni RB, Higano CS, Ho SM, Vessella RL & Corey E 2004 Metastases of prostate cancer express estrogen receptor-β. Urology 64 814820. (doi:10.1016/j.urology.2004.05.036).

    • Search Google Scholar
    • Export Citation
  • Lattrich C, Schuler S, Haring J, Skrzypczak M, Ortmann O & Treeck O Effects of a combined treatment with tamoxifen and estrogen receptor β agonists on human breast cancer cell lines Archives of Gynecology and Obstetrics 2013 [in press] doi:10.1007/s00404-013-2977-7).

    • Search Google Scholar
    • Export Citation
  • Lazennec G, Bresson D, Lucas A, Chauveau C & Vignon F 2001 ERβ inhibits proliferation and invasion of breast cancer cells. Endocrinology 142 41204130. (doi:10.1210/en.142.9.4120).

    • Search Google Scholar
    • Export Citation
  • Lechner D, Kallay E & Cross HS 2005 Phytoestrogens and colorectal cancer prevention. Vitamins and Hormones 70 169198.

  • Lee MT, Ouyang B, Ho SM & Leung YK 2013 Differential expression of estrogen receptor β isoforms in prostate cancer through interplay between transcriptional and translational regulation. Molecular and Cellular Endocrinology 376 125135. (doi:10.1016/j.mce.2013.06.023).

    • Search Google Scholar
    • Export Citation
  • Le Romancer M, Poulard C, Cohen P, Sentis S, Renoir JM & Corbo L 2011 Cracking the estrogen receptor's posttranslational code in breast tumors. Endocrine Reviews 32 597622. (doi:10.1210/er.2010-0016).

    • Search Google Scholar
    • Export Citation
  • Leung YK, Gao Y, Lau KM, Zhang X & Ho SM 2006a ICI 182,780-regulated gene expression in DU145 prostate cancer cells is mediated by estrogen receptor-β/NFκB crosstalk. Neoplasia 8 242249. (doi:10.1593/neo.05853).

    • Search Google Scholar
    • Export Citation
  • Leung YK, Mak P, Hassan S & Ho SM 2006b Estrogen receptor (ER)-β isoforms: a key to understanding ER-β signaling. PNAS 103 1316213167. (doi:10.1073/pnas.0605676103).

    • Search Google Scholar
    • Export Citation
  • Leung YK, Lam HM, Wu S, Song D, Levin L, Cheng L, Wu CL & Ho SM 2010 Estrogen receptor β2 and β5 are associated with poor prognosis in prostate cancer, and promote cancer cell migration and invasion. Endocrine-Related Cancer 17 675689. (doi:10.1677/ERC-09-0294).

    • Search Google Scholar
    • Export Citation
  • Li S, Han B, Liu G, Li S, Ouellet J, Labrie F & Pelletier G 2010 Immunocytochemical localization of sex steroid hormone receptors in normal human mammary gland. Journal of Histochemistry and Cytochemistry 58 509515. (doi:10.1369/jhc.2009.954644).

    • Search Google Scholar
    • Export Citation
  • Li W, Winters A, Poteet E, Ryou MG, Lin S, Hao S, Wu Z, Yuan F, Hatanpaa KJ & Simpkins JW et al. 2013 Involvement of estrogen receptor β5 in the progression of glioma. Brain Research 1503 97107. (doi:10.1016/j.brainres.2013.02.004).

    • Search Google Scholar
    • Export Citation
  • Lindberg K, Helguero LA, Omoto Y, Gustafsson JA & Haldosen LA 2011 Estrogen receptor β represses Akt signaling in breast cancer cells via downregulation of HER2/HER3 and upregulation of PTEN: implications for tamoxifen sensitivity. Breast Cancer Research 13 R43. (doi:10.1186/bcr2865).

    • Search Google Scholar
    • Export Citation
  • Love RR & Philips J 2002 Oophorectomy for breast cancer: history revisited. Journal of the National Cancer Institute 94 14331434. (doi:10.1093/jnci/94.19.1433).

    • Search Google Scholar
    • Export Citation
  • Ma Y, Lovekamp-Swan T, Bekele W, Dohi A & Schreihofer DA 2013 Hypoxia-inducible factor and vascular endothelial growth factor are targets of dietary soy during acute stroke in female rats. Endocrinology 154 15891597. (doi:10.1210/en.2012-2120).

    • Search Google Scholar
    • Export Citation
  • Maggiolini M, Bonofiglio D, Marsico S, Panno ML, Cenni B, Picard D & Ando S 2001 Estrogen receptor α mediates the proliferative but not the cytotoxic dose-dependent effects of two major phytoestrogens on human breast cancer cells. Molecular Pharmacology 60 595602.

    • Search Google Scholar
    • Export Citation
  • Mak P, Leav I, Pursell B, Bae D, Yang X, Taglienti CA, Gouvin LM, Sharma VM & Mercurio AM 2010 ERβ impedes prostate cancer EMT by destabilizing HIF-1α and inhibiting VEGF-mediated snail nuclear localization: implications for Gleason grading. Cancer Cell 17 319332. (doi:10.1016/j.ccr.2010.02.030).

    • Search Google Scholar
    • Export Citation
  • Mak P, Chang C, Pursell B & Mercurio AM 2013 Estrogen receptor β sustains epithelial differentiation by regulating prolyl hydroxylase 2 transcription. PNAS 110 47084713. (doi:10.1073/pnas.1221654110).

    • Search Google Scholar
    • Export Citation
  • Malamas MS, Manas ES, McDevitt RE, Gunawan I, Xu ZB, Collini MD, Miller CP, Dinh T, Henderson RA & Keith JC Jr et al. 2004 Design and synthesis of aryl diphenolic azoles as potent and selective estrogen receptor-β ligands. Journal of Medicinal Chemistry 47 50215040. (doi:10.1021/jm049719y).

    • Search Google Scholar
    • Export Citation
  • Markey GC, Cullen R, Diggin P, Hill AD, Mc Dermott EW, O'Higgins NJ & Duffy MJ 2009 Estrogen receptor-β mRNA is associated with adverse outcome in patients with breast cancer. Tumour Biology 30 171175. (doi:10.1159/000236409).

    • Search Google Scholar
    • Export Citation
  • McCarthy M 2013 Finasteride for prostate cancer prevention has no effect on survival, study finds. BMJ 347 f5203. (doi:10.1136/bmj.f5203).

    • Search Google Scholar
    • Export Citation
  • McGaha TL, Huang L, Lemos H, Metz R, Mautino M, Prendergast GC & Mellor AL 2012 Amino acid catabolism: a pivotal regulator of innate and adaptive immunity. Immunological Reviews 249 135157. (doi:10.1111/j.1600-065X.2012.01149.x).

    • Search Google Scholar
    • Export Citation
  • McGlynn LM, Tovey S, Bartlett JM, Doughty J, Cooke TG & Edwards J 2013 Interactions between MAP kinase and oestrogen receptor in human breast cancer. European Journal of Cancer 49 11761186. (doi:10.1016/j.ejca.2012.11.020).

    • Search Google Scholar
    • Export Citation
  • McPherson SJ, Hussain S, Balanathan P, Hedwards SL, Niranjan B, Grant M, Chandrasiri UP, Toivanen R, Wang Y & Taylor RA et al. 2010 Estrogen receptor-β activated apoptosis in benign hyperplasia and cancer of the prostate is androgen independent and TNFα mediated. PNAS 107 31233128. (doi:10.1073/pnas.0905524107).

    • Search Google Scholar
    • Export Citation
  • Miller TW, Balko JM, Fox EM, Ghazoui Z, Dunbier A, Anderson H, Dowsett M, Jiang A, Smith RA & Maira SM et al. 2011 ERα-dependent E2F transcription can mediate resistance to estrogen deprivation in human breast cancer. Cancer Discovery 1 338351. (doi:10.1158/2159-8290.CD-11-0101).

    • Search Google Scholar
    • Export Citation
  • Moore JT, McKee DD, Slentz-Kesler K, Moore LB, Jones SA, Horne EL, Su JL, Kliewer SA, Lehmann JM & Willson TM 1998 Cloning and characterization of human estrogen receptor β isoforms. Biochemical and Biophysical Research Communications 247 7578. (doi:10.1006/bbrc.1998.8738).

    • Search Google Scholar
    • Export Citation
  • Murphy LC & Watson PH 2006 Is oestrogen receptor-β a predictor of endocrine therapy responsiveness in human breast cancer? Endocrine-Related Cancer 13 327334. (doi:10.1677/erc.1.01141).

    • Search Google Scholar
    • Export Citation
  • Murphy LC & Leygue E 2012 The role of estrogen receptor-β in breast cancer. Seminars in Reproductive Medicine 30 513. (doi:10.1055/s-0031-1299592).

    • Search Google Scholar
    • Export Citation
  • Musatov S, Chen W, Pfaff DW, Mobbs CV, Yang XJ, Clegg DJ, Kaplitt MG & Ogawa S 2007 Silencing of estrogen receptor α in the ventromedial nucleus of hypothalamus leads to metabolic syndrome. PNAS 104 25012506. (doi:10.1073/pnas.0610787104).

    • Search Google Scholar
    • Export Citation
  • Muschamp JW & Hull EM 2007 Melanin concentrating hormone and estrogen receptor-α are coexstensive but not coexpressed in cells of male rat hypothalamus. Neuroscience Letters 427 123126. (doi:10.1016/j.neulet.2007.09.031).

    • Search Google Scholar
    • Export Citation
  • Musgrove EA & Sutherland RL 2009 Biological determinants of endocrine resistance in breast cancer. Nature Reviews. Cancer 9 631643. (doi:10.1038/nrc2713).

    • Search Google Scholar
    • Export Citation
  • Naftolin F 2008 Sex steroids and the brain. Gynecological Endocrinology 24 533534. (doi:10.1080/09513590802296104).

  • Nahon JL 2006 The melanocortins and melanin-concentrating hormone in the central regulation of feeding behavior and energy homeostasis. Comptes Rendus Biologies 329 623638.(discussion 653-655) (doi:10.1016/j.crvi.2006.03.021).

    • Search Google Scholar
    • Export Citation
  • Nakajima Y, Akaogi K, Suzuki T, Osakabe A, Yamaguchi C, Sunahara N, Ishida J, Kako K, Ogawa S & Fujimura T et al. 2011 Estrogen regulates tumor growth through a nonclassical pathway that includes the transcription factors ERβ and KLF5. Science Signaling 4 ra22. (doi:10.1126/scisignal.2001551).

    • Search Google Scholar
    • Export Citation
  • Nehra R, Riggins RB, Shajahan AN, Zwart A, Crawford AC & Clarke R 2010 BCL2 and CASP8 regulation by NF-κB differentially affect mitochondrial function and cell fate in antiestrogen-sensitive and -resistant breast cancer cells. FASEB Journal 24 20402055. (doi:10.1096/fj.09-138305).

    • Search Google Scholar
    • Export Citation
  • Nilsson S, Koehler KF & Gustafsson JA 2011 Development of subtype-selective oestrogen receptor-based therapeutics. Nature Reviews. Drug Discovery 10 778792. (doi:10.1038/nrd3551).

    • Search Google Scholar
    • Export Citation
  • Njiaju UO & Olopade OI 2012 Genetic determinants of breast cancer risk: a review of current literature and issues pertaining to clinical application. Breast Journal 18 436442. (doi:10.1111/j.1524-4741.2012.01274.x).

    • Search Google Scholar
    • Export Citation
  • Ogawa S, Inoue S, Watanabe T, Orimo A, Hosoi T, Ouchi Y & Muramatsu M 1998 Molecular cloning and characterization of human estrogen receptor βcx: a potential inhibitor of estrogen action in human. Nucleic Acids Research 26 35053512. (doi:10.1093/nar/26.15.3505).

    • Search Google Scholar
    • Export Citation
  • Omoto Y, Imamov O, Warner M & Gustafsson JA 2005 Estrogen receptor α and imprinting of the neonatal mouse ventral prostate by estrogen. PNAS 102 14841489. (doi:10.1073/pnas.0409168102).

    • Search Google Scholar
    • Export Citation
  • Osterlund MK 2010 Underlying mechanisms mediating the antidepressant effects of estrogens. Biochimica et Biophysica Acta 1800 11361144. (doi:10.1016/j.bbagen.2009.11.001).

    • Search Google Scholar
    • Export Citation
  • Paige LA, Christensen DJ, Gron H, Norris JD, Gottlin EB, Padilla KM, Chang CY, Ballas LM, Hamilton PT & McDonnell DP et al. 1999 Estrogen receptor (ER) modulators each induce distinct conformational changes in ERα and ERβ. PNAS 96 39994004. (doi:10.1073/pnas.96.7.3999).

    • Search Google Scholar
    • Export Citation
  • Paoloni-Giacobino A, Chen H, Peitsch MC, Rossier C & Antonarakis SE 1997 Cloning of the TMPRSS2 gene, which encodes a novel serine protease with transmembrane, LDLRA, and SRCR domains and maps to 21q22.3. Genomics 44 309320. (doi:10.1006/geno.1997.4845).

    • Search Google Scholar
    • Export Citation
  • Paruthiyil S, Parmar H, Kerekatte V, Cunha GR, Firestone GL & Leitman DC 2004 Estrogen receptor β inhibits human breast cancer cell proliferation and tumor formation by causing a G2 cell cycle arrest. Cancer Research 64 423428. (doi:10.1158/0008-5472.CAN-03-2446).

    • Search Google Scholar
    • Export Citation
  • Pettersson K & Gustafsson JA 2001 Role of estrogen receptor β in estrogen action. Annual Review of Physiology 63 165192. (doi:10.1146/annurev.physiol.63.1.165).

    • Search Google Scholar
    • Export Citation
  • Pike AC, Brzozowski AM, Hubbard RE, Bonn T, Thorsell AG, Engstrom O, Ljunggren J, Gustafsson JA & Carlquist M 1999 Structure of the ligand-binding domain of oestrogen receptor β in the presence of a partial agonist and a full antagonist. EMBO Journal 18 46084618. (doi:10.1093/emboj/18.17.4608).

    • Search Google Scholar
    • Export Citation
  • Pillot B, Duraffourd C, Begeot M, Joly A, Luquet S, Houberdon I, Naville D, Vigier M, Gautier-Stein A & Magnan C et al. 2011 Role of hypothalamic melanocortin system in adaptation of food intake to food protein increase in mice. PLoS ONE 6 e19107. (doi:10.1371/journal.pone.0019107).

    • Search Google Scholar
    • Export Citation
  • Pravettoni A, Mornati O, Martini PG, Marino M, Colciago A, Celotti F, Motta M & Negri-Cesi P 2007 Estrogen receptor β (ERβ) and inhibition of prostate cancer cell proliferation: studies on the possible mechanism of action in DU145 cells. Molecular and Cellular Endocrinology 263 4654. (doi:10.1016/j.mce.2006.08.008).

    • Search Google Scholar
    • Export Citation
  • Price RH Jr, Butler CA, Webb P, Uht R, Kushner P & Handa RJ 2001 A splice variant of estrogen receptor β missing exon 3 displays altered subnuclear localization and capacity for transcriptional activation. Endocrinology 142 20392049. (doi:10.1210/en.142.5.2039).

    • Search Google Scholar
    • Export Citation
  • Prins GS & Birch L 1997 Neonatal estrogen exposure up-regulates estrogen receptor expression in the developing and adult rat prostate lobes. Endocrinology 138 18011809. (doi:10.1210/en.138.5.1801).

    • Search Google Scholar
    • Export Citation
  • Raval AP, Borges-Garcia R, Javier Moreno W, Perez-Pinzon MA & Bramlett H 2013 Periodic 17β-estradiol pretreatment protects rat brain from cerebral ischemic damage via estrogen receptor-β. PLoS ONE 8 e60716. (doi:10.1371/journal.pone.0060716).

    • Search Google Scholar
    • Export Citation
  • Rice S, Patel B, Bano G, Ugwumadu A & Whitehead SA 2012 Aromatase expression in abdominal omental/visceral and subcutaneous fat depots: a comparison of pregnant and obese women. Fertility and Sterility 97 14601466.(e1461) (doi:10.1016/j.fertnstert.2012.03.008).

    • Search Google Scholar
    • Export Citation
  • Riggio M, Polo ML, Blaustein M, Colman-Lerner A, Luthy I, Lanari C & Novaro V 2012 PI3K/AKT pathway regulates phosphorylation of steroid receptors, hormone independence and tumor differentiation in breast cancer. Carcinogenesis 33 509518. (doi:10.1093/carcin/bgr303).

    • Search Google Scholar
    • Export Citation
  • Roepke TA 2009 Oestrogen modulates hypothalamic control of energy homeostasis through multiple mechanisms. Journal of Neuroendocrinology 21 141150. (doi:10.1111/j.1365-2826.2008.01814.x).

    • Search Google Scholar
    • Export Citation
  • Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV & Johnson KC et al. 2002 Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. Journal of the American Medical Association 288 321333. (doi:10.1001/jama.288.3.321).

    • Search Google Scholar
    • Export Citation
  • Rudolph A, Toth C, Hoffmeister M, Roth W, Herpel E, Jansen L, Marx A, Brenner H & Chang-Claude J 2012 Expression of oestrogen receptor β and prognosis of colorectal cancer. British Journal of Cancer 107 831839. (doi:10.1038/bjc.2012.323).

    • Search Google Scholar
    • Export Citation
  • Saji S, Jensen EV, Nilsson S, Rylander T, Warner M & Gustafsson JA 2000 Estrogen receptors α and β in the rodent mammary gland. PNAS 97 337342. (doi:10.1073/pnas.97.1.337).

    • Search Google Scholar
    • Export Citation
  • Saji S, Omoto Y, Shimizu C, Warner M, Hayashi Y, Horiguchi S, Watanabe T, Hayashi S, Gustafsson JA & Toi M 2002 Expression of estrogen receptor (ER) (β)cx protein in ER(α)-positive breast cancer: specific correlation with progesterone receptor. Cancer Research 62 48494853.

    • Search Google Scholar
    • Export Citation
  • Saleiro D, Murillo G, Lubahn DB, Kopelovich L, Korach KS & Mehta RG 2010 Enhanced induction of mucin-depleted foci in estrogen receptor {β} knockout mice. Cancer Prevention Research 3 11981204. (doi:10.1158/1940-6207.CAPR-10-0044).

    • Search Google Scholar
    • Export Citation
  • Saleiro D, Murillo G, Benya RV, Bissonnette M, Hart J & Mehta RG 2012 Estrogen receptor-β protects against colitis-associated neoplasia in mice. International Journal of Cancer 131 25532561. (doi:10.1002/ijc.27578).

    • Search Google Scholar
    • Export Citation
  • Sanchez ER 2012 Chaperoning steroidal physiology: lessons from mouse genetic models of Hsp90 and its cochaperones. Biochimica et Biophysica Acta 1823 722729. (doi:10.1016/j.bbamcr.2011.11.006).

    • Search Google Scholar
    • Export Citation
  • Schulze H & Barrack ER 1987 Immunocytochemical localization of estrogen receptors in the normal male and female canine urinary tract and prostate. Endocrinology 121 17731783. (doi:10.1210/endo-121-5-1773).

    • Search Google Scholar
    • Export Citation
  • Schulze H & Claus S 1990 Histological localization of estrogen receptors in normal and diseased human prostates by immunocytochemistry. Prostate 16 331343. (doi:10.1002/pros.2990160408).

    • Search Google Scholar
    • Export Citation
  • Setlur SR, Mertz KD, Hoshida Y, Demichelis F, Lupien M, Perner S, Sboner A, Pawitan Y, Andren O & Johnson LA et al. 2008 Estrogen-dependent signaling in a molecularly distinct subclass of aggressive prostate cancer. Journal of the National Cancer Institute 100 815825. (doi:10.1093/jnci/djn150).

    • Search Google Scholar
    • Export Citation
  • Shaaban AM, O'Neill PA, Davies MP, Sibson R, West CR, Smith PH & Foster CS 2003 Declining estrogen receptor-β expression defines malignant progression of human breast neoplasia. American Journal of Surgical Pathology 27 15021512. (doi:10.1097/00000478-200312000-00002).

    • Search Google Scholar
    • Export Citation
  • Shao TC, Castaneda E, Rosenfield RL & Liao S 1975 Selective retention and formation of a Δ5-androstenediol-receptor complex in cell nuclei of the rat vagina. Journal of Biological Chemistry 250 30953100.

    • Search Google Scholar
    • Export Citation
  • Shiota M, Yokomizo A, Masubuchi D, Tada Y, Inokuchi J, Eto M, Uchiumi T, Fujimoto N & Naito S 2010 Tip60 promotes prostate cancer cell proliferation by translocation of androgen receptor into the nucleus. Prostate 70 540554. (doi:10.1002/pros.21088).

    • Search Google Scholar
    • Export Citation
  • Siegel R, Naishadham D & Jemal A 2013 Cancer statistics, 2013. CA: A Cancer Journal for Clinicians 63 1130. (doi:10.3322/caac.21166).

  • Slusarz A, Jackson GA, Day JK, Shenouda NS, Bogener JL, Browning JD, Fritsche KL, MacDonald RS, Besch-Williford CL & Lubahn DB 2012 Aggressive prostate cancer is prevented in ERαKO mice and stimulated in ERβKO TRAMP mice. Endocrinology 153 41604170. (doi:10.1210/en.2012-1030).

    • Search Google Scholar
    • Export Citation
  • de Souza FS, Nasif S, Lopez-Leal R, Levi DH, Low MJ & Rubinsten M 2011 The estrogen receptor α colocalizes with proopiomelanocortin in hypothalamic neurons and binds to a conserved motif present in the neuron-specific enhancer nPE2. European Journal of Pharmacology 660 181187. (doi:10.1016/j.ejphar.2010.10.114).

    • Search Google Scholar
    • Export Citation
  • Speirs V, Skliris GP, Burdall SE & Carder PJ 2002 Distinct expression patterns of ERα and ERβ in normal human mammary gland. Journal of Clinical Pathology 55 371374. (doi:10.1136/jcp.55.5.371).

    • Search Google Scholar
    • Export Citation
  • Stertz L, Magalhaes PV & Kapczinski F 2013 Is bipolar disorder an inflammatory condition? The relevance of microglial activation. Current Opinion in Psychiatry 26 1926. (doi:10.1097/YCO.0b013e32835aa4b4).

    • Search Google Scholar
    • Export Citation
  • Stewart ME, Pochi PE, Strauss JS, Wotiz HH & Clark SJ 1977 In-vitro metabolism of [3H]testosterone by scalp and back skin: conversion of testosterone into 5α-androstane-3β, 17β-diol. Journal of Endocrinology 72 385390. (doi:10.1677/joe.0.0720385).

    • Search Google Scholar
    • Export Citation
  • Stocco C 2012 Tissue physiology and pathology of aromatase. Steroids 77 2735. (doi:10.1016/j.steroids.2011.10.013).

  • Strom A, Hartman J, Foster JS, Kietz S, Wimalasena J & Gustafsson JA 2004 Estrogen receptor β inhibits 17β-estradiol-stimulated proliferation of the breast cancer cell line T47D. PNAS 101 15661571. (doi:10.1073/pnas.0308319100).

    • Search Google Scholar
    • Export Citation
  • Sugiura H, Toyama T, Hara Y, Zhang Z, Kobayashi S, Fujii Y, Iwase H & Yamashita H 2007 Expression of estrogen receptor β wild-type and its variant ERβcx/β2 is correlated with better prognosis in breast cancer. Japanese Journal of Clinical Oncology 37 820828. (doi:10.1093/jjco/hym114).

    • Search Google Scholar
    • Export Citation
  • Sun PM, Gao M, Wei LH, Mustea A, Wang JL, Konsgen D, Lichtenegger W & Sehouli J 2006 An estrogen receptor α-dependent regulation of estrogen receptor-related receptor α in the proliferation of endometrial carcinoma cells. International Journal of Gynecological Cancer 16 (Suppl 2) 564568. (doi:10.1111/j.1525-1438.2006.00697.x).

    • Search Google Scholar
    • Export Citation
  • Sun Y, Jiang X & Price BD 2010 Tip60: connecting chromatin to DNA damage signaling. Cell Cycle 9 930936. (doi:10.4161/cc.9.5.10931).

  • Suzuki H, Barros RP, Sugiyama N, Krishnan V, Yaden BC, Kim HJ, Warner M & Gustafsson JA 2013 Involvement of estrogen receptor β in maintenance of serotonergic neurons of the dorsal raphe. Molecular Psychiatry 18 674680. (doi:10.1038/mp.2012.62).

    • Search Google Scholar
    • Export Citation
  • Taplin ME & Ho SM 2001 Clinical review 134: The endocrinology of prostate cancer. Journal of Clinical Endocrinology and Metabolism 86 34673477. (doi:10.1210/jc.86.8.3467).

    • Search Google Scholar
    • Export Citation
  • Taylor JA, Richter CA, Suzuki A, Watanabe H, Iguchi T, Coser KR, Shioda T & vom Saal FS 2012 Dose-related estrogen effects on gene expression in fetal mouse prostate mesenchymal cells. PLoS ONE 7 e48311. (doi:10.1371/journal.pone.0048311).

    • Search Google Scholar
    • Export Citation
  • Teng J, Wang ZY, Jarrard DF & Bjorling DE 2008 Roles of estrogen receptor α and β in modulating urothelial cell proliferation. Endocrine-Related Cancer 15 351364. (doi:10.1677/erc.1.01255).

    • Search Google Scholar
    • Export Citation
  • Theoharides TC, Asadi S & Patel AB 2013 Focal brain inflammation and autism. Journal of Neuroinflammation 10 46. (doi:10.1186/1742-2094-10-46).

    • Search Google Scholar
    • Export Citation
  • Thomas C & Gustafsson JA 2011 The different roles of ER subtypes in cancer biology and therapy. Nature Reviews. Cancer 11 597608. (doi:10.1038/nrc3093).

    • Search Google Scholar
    • Export Citation
  • Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, Varambally S, Cao X, Tchinda J & Kuefer R et al. 2005 Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310 644648. (doi:10.1126/science.1117679).

    • Search Google Scholar
    • Export Citation
  • Tsurusaki T, Aoki D, Kanetake H, Inoue S, Muramatsu M, Hishikawa Y & Koji T 2003 Zone-dependent expression of estrogen receptors α and β in human benign prostatic hyperplasia. Journal of Clinical Endocrinology and Metabolism 88 13331340. (doi:10.1210/jc.2002-021015).

    • Search Google Scholar
    • Export Citation
  • Vinayagam R, Sibson DR, Holcombe C, Aachi V & Davies MP 2007 Association of oestrogen receptor β2 (ERβ2/ERβcx) with outcome of adjuvant endocrine treatment for primary breast cancer – a retrospective study. BMC Cancer 7 131. (doi:10.1186/1471-2407-7-131).

    • Search Google Scholar
    • Export Citation
  • Wada-Hiraike O, Imamov O, Hiraike H, Hultenby K, Schwend T, Omoto Y, Warner M & Gustafsson JA 2006 Role of estrogen receptor β in colonic epithelium. PNAS 103 29592964. (doi:10.1073/pnas.0511271103).

    • Search Google Scholar
    • Export Citation
  • Walter P, Green S, Greene G, Krust A, Bornert JM, Jeltsch JM, Staub A, Jensen E, Scrace G & Waterfield M et al. 1985 Cloning of the human estrogen receptor cDNA. PNAS 82 78897893. (doi:10.1073/pnas.82.23.7889).

    • Search Google Scholar
    • Export Citation
  • Wang L, Andersson S, Warner M & Gustafsson JA 2003 Estrogen receptor (ER)β knockout mice reveal a role for ERβ in migration of cortical neurons in the developing brain. PNAS 100 703708. (doi:10.1073/pnas.242735799).

    • Search Google Scholar
    • Export Citation
  • Wang C, Mayer JA, Mazumdar A, Fertuck K, Kim H, Brown M & Brown PH 2011 Estrogen induces c-myc gene expression via an upstream enhancer activated by the estrogen receptor and the AP-1 transcription factor. Molecular Endocrinology 25 15271538. (doi:10.1210/me.2011-1037).

    • Search Google Scholar
    • Export Citation
  • Wang H, Li Q & Chen H 2012 Genistein affects histone modifications on Dickkopf-related protein 1 (DKK1) gene in SW480 human colon cancer cell line. PLoS ONE 7 e40955. (doi:10.1371/journal.pone.0040955).

    • Search Google Scholar
    • Export Citation
  • Webb P, Nguyen P, Valentine C, Lopez GN, Kwok GR, McInerney E, Katzenellenbogen BS, Enmark E, Gustafsson JA & Nilsson S et al. 1999 The estrogen receptor enhances AP-1 activity by two distinct mechanisms with different requirements for receptor transactivation functions. Molecular Endocrinology 13 16721685. (doi:10.1210/me.13.10.1672).

    • Search Google Scholar
    • Export Citation
  • Webb P, Valentine C, Nguyen P, Price RH Jr, Marimuthu A, West BL, Baxter JD & Kushner PJ 2003 ERβ binds N-CoR in the presence of estrogens via an LXXLL-like motif in the N-CoR C-terminus. Nuclear Receptor 1 4. (doi:10.1186/1478-1336-1-4).

    • Search Google Scholar
    • Export Citation
  • Weihua Z, Lathe R, Warner M & Gustafsson JA 2002a An endocrine pathway in the prostate, ERβ, AR, 5α-androstane-3β,17β-diol, and CYP7B1, regulates prostate growth. PNAS 99 1358913594. (doi:10.1073/pnas.162477299).

    • Search Google Scholar
    • Export Citation
  • Weihua Z, Warner M & Gustafsson JA 2002b Estrogen receptor β in the prostate. Molecular and Cellular Endocrinology 193 15. (doi:10.1016/S0303-7207(02)00089-8).

    • Search Google Scholar
    • Export Citation
  • White R, Lees JA, Needham M, Ham J & Parker M 1987 Structural organization and expression of the mouse estrogen receptor. Molecular Endocrinology 1 735744. (doi:10.1210/mend-1-10-735).

    • Search Google Scholar
    • Export Citation
  • Wu WF, Tan XJ, Dai YB, Krishnan V, Warner M & Gustafsson JA 2013 Targeting estrogen receptor β in microglia and T cells to treat experimental autoimmune encephalomyelitis. PNAS 110 35433548. (doi:10.1073/pnas.1300313110).

    • Search Google Scholar
    • Export Citation
  • Xu B, Goulding EH, Zang K, Cepoi D, Cone RD, Jones KR, Tecott LH & Reichardt LF 2003 Brain-derived neurotrophic factor regulates energy balance downstream of melanocortin-4 receptor. Nature Neuroscience 6 736742. (doi:10.1038/nn1073).

    • Search Google Scholar
    • Export Citation
  • Xu J, Wu RC & O'Malley BW 2009 Normal and cancer-related functions of the p160 steroid receptor co-activator (SRC) family. Nature Reviews. Cancer 9 615630. (doi:10.1038/nrc2695).

    • Search Google Scholar
    • Export Citation
  • Xue B, Zhang Z, Beltz TG, Johnson RF, Guo F, Hay M & Johnson AK 2013 Estrogen receptor-β in the paraventricular nucleus and rostroventrolateral medulla plays an essential protective role in aldosterone/salt-induced hypertension in female rats. Hypertension 61 12551262. (doi:10.1161/HYPERTENSIONAHA.111.00903).

    • Search Google Scholar
    • Export Citation
  • Yang GS, Wang Y, Wang P & Chen ZD 2007 Expression of oestrogen receptor-α and oestrogen receptor-β in prostate cancer. Chinese Medical Journal 120 16111615.

    • Search Google Scholar
    • Export Citation
  • Yu M, Yu J, Mani RS, Cao Q, Brenner CJ, Cao X, Wang X, Wu L, Li J & Hu M et al. 2010 An integrated network of androgen receptor, polycomb, and TMPRSS2–ERG gene fusions in prostate cancer progression. Cancer Cell 17 443454. (doi:10.1016/j.ccr.2010.03.018).

    • Search Google Scholar
    • Export Citation
  • Zang YC, Halder JB, Hong J, Rivera VM & Zhang JZ 2002 Regulatory effects of estriol on T cell migration and cytokine profile: inhibition of transcription factor NF-κB. Journal of Neuroimmunology 124 106114. (doi:10.1016/S0165-5728(02)00016-4).

    • Search Google Scholar
    • Export Citation
  • Zhao C, Gao H, Liu Y, Papoutsi Z, Jaffrey S, Gustafsson JA & Dahlman-Wright K 2010 Genome-wide mapping of estrogen receptor-β-binding regions reveals extensive cross-talk with transcription factor activator protein-1. Cancer Research 70 51745183. (doi:10.1158/0008-5472.CAN-09-4407).

    • Search Google Scholar
    • Export Citation
  • Zhou R, Lai Y, Yamabe N, Fukui M & Zhu BT 2011 Estriol has different effects from 17β-estradiol in modulating mouse splenocyte function under inflammatory conditions. Journal of Immunotoxicology 8 346358. (doi:10.3109/1547691X.2011.617791).

    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • View in gallery

    Schematic of the structure of the estrogen receptor (ER). The ER is composed of three main domains: the N-terminal domain (NTD), which contains the activation function 1, the DNA-binding domain (DBD), and the ligand-binding domain (LBD), containing the activation function 2. The DBDs of ERα and ERβ are highly conserved and the NTDs are the least conserved. The NTD of ERα is shorter than that of ERβ.

  • View in gallery

    Pathways regulated by ERβ. ERβ upon binding to its ligand 3β-Adiol/8β-VE2/DPN gets activated and transcriptionally upregulates downstream target genes such as FOXO3a and PHD2. The target genes carry ERβ binding elements such as ERE or AP1-ERE-half-site upstream or downstream of the transcriptional start site. The activated FOXO3a in turn transcriptionally upregulates PUMA, p21, and p27. PHD2 inhibits HIF1α by prolyl hydroxylation of the oxygen-dependent degradation domain (ODDD), which targets HIF1α proteins for proteasomal degradation by promoting their interaction with von Hippel–Lindau (VHL). ERβ also inhibits proliferation of genes such as c-MYC and p45Skp2 by an as yet unknown mechanism. Moreover, some of the EMT and bone metastasis genes, such as β-catenin, SLUG/SNAIL and TWIST, are opposed by ERβ.

  • Antal MC, Krust A, Chambon P & Mark M 2008 Sterility and absence of histopathological defects in nonreproductive organs of a mouse ERβ-null mutant. PNAS 105 24332438. (doi:10.1073/pnas.0712029105).

    • Search Google Scholar
    • Export Citation
  • Armstrong CM, Billimek AR, Allred KF, Sturino JM, Weeks BR & Allred CD 2013 A novel shift in estrogen receptor expression occurs as estradiol suppresses inflammation-associated colon tumor formation. Endocrine-Related Cancer 20 515525. (doi:10.1530/ERC-12-0308).

    • Search Google Scholar
    • Export Citation
  • Attia DM & Ederveen AG 2012 Opposing roles of ERα and ERβ in the genesis and progression of adenocarcinoma in the rat ventral prostate. Prostate 72 10131022. (doi:10.1002/pros.21507).

    • Search Google Scholar
    • Export Citation
  • Barros RP & Gustafsson JA 2011 Estrogen receptors and the metabolic network. Cell Metabolism 14 289299. (doi:10.1016/j.cmet.2011.08.005).

    • Search Google Scholar
    • Export Citation
  • Barros RP, Gabbi C, Morani A, Warner M & Gustafsson JA 2009 Participation of ERα and ERβ in glucose homeostasis in skeletal muscle and white adipose tissue. American Journal of Physiology. Endocrinology and Metabolism 297 E124E133. (doi:10.1152/ajpendo.00189.2009).

    • Search Google Scholar
    • Export Citation
  • Benjamins JA 2013 Direct effects of secretory products of immune cells on neurons and glia. Journal of Neurological Sciences 333 3036. (doi:10.1016/j.jns.2013.06.001).

    • Search Google Scholar
    • Export Citation
  • Bielecki A, Roberts J, Mehta R & Raju J 2011 Estrogen receptor-β mediates the inhibition of DLD-1 human colon adenocarcinoma cells by soy isoflavones. Nutrition and Cancer 63 139150. (doi:10.1080/01635581.2010.516867).

    • Search Google Scholar
    • Export Citation
  • Boon WC, Chow JD & Simpson ER 2010 The multiple roles of estrogens and the enzyme aromatase. Progress in Brain Research 181 209232. (doi:10.1016/S0079-6123(08)81012-6).

    • Search Google Scholar
    • Export Citation
  • Bosland MC 2000 The role of steroid hormones in prostate carcinogenesis. Journal of the National Cancer Institute. Monographs 2000 (27) 3966. (doi:10.1093/oxfordjournals.jncimonographs.a024244).

    • Search Google Scholar
    • Export Citation
  • Brenner RM, West NB & McClellan MC 1990 Estrogen and progestin receptors in the reproductive tract of male and female primates. Biology of Reproduction 42 1119. (doi:10.1095/biolreprod42.1.11).

    • Search Google Scholar
    • Export Citation
  • Briganti A 2009 Oestrogens and prostate cancer: novel concepts about an old issue. European Urology 55 543545. (doi:10.1016/j.eururo.2008.12.004).

    • Search Google Scholar
    • Export Citation
  • Butt AJ, McNeil CM, Musgrove EA & Sutherland RL 2005 Downstream targets of growth factor and oestrogen signalling and endocrine resistance: the potential roles of c-Myc, cyclin D1 and cyclin E. Endocrine-Related Cancer 12 (Suppl 1) S47S59. (doi:10.1677/erc.1.00993).

    • Search Google Scholar
    • Export Citation
  • Carroll JS, Liu XS, Brodsky AS, Li W, Meyer CA, Szary AJ, Eeckhoute J, Shao W, Hestermann EV & Geistlinger TR et al. 2005 Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1. Cell 122 3343. (doi:10.1016/j.cell.2005.05.008).

    • Search Google Scholar
    • Export Citation
  • Carson-Jurica MA, Schrader WT & O'Malley BW 1990 Steroid receptor family: structure and functions. Endocrine Reviews 11 201220. (doi:10.1210/edrv-11-2-201).

    • Search Google Scholar
    • Export Citation
  • Chen Y, Chen L, Li JY, Mukaida N, Wang Q, Yang C, Yin WJ, Zeng XH, Jin W & Shao ZM 2011 ERβ and PEA3 co-activate IL-8 expression and promote the invasion of breast cancer cells. Cancer Biology & Therapy 11 497511. (doi:10.4161/cbt.11.5.14667).

    • Search Google Scholar
    • Export Citation
  • Chen P, Hover CV, Lindberg D & Li C 2012 Central urocortin 3 and type 2 corticotropin-releasing factor receptor in the regulation of energy homeostasis: critical involvement of the ventromedial hypothalamus. Frontiers in Endocrinology 3 180. (doi:10.3389/fendo.2012.00180).

    • Search Google Scholar
    • Export Citation
  • Cheng J, Lee EJ, Madison LD & Lazennec G 2004 Expression of estrogen receptor β in prostate carcinoma cells inhibits invasion and proliferation and triggers apoptosis. FEBS Letters 566 169172. (doi:10.1016/j.febslet.2004.04.025).

    • Search Google Scholar
    • Export Citation
  • Cheng G, Butler R, Warner M, Gustafsson JA, Wilczek B & Landgren BM 2013 Effects of short-term estradiol and norethindrone acetate treatment on the breasts of normal postmenopausal women. Menopause 20 496503. (doi:10.1097/GME.0b013e318276c4ea).

    • Search Google Scholar
    • Export Citation
  • Clark JA, Alves S, Gundlah C, Rocha B, Birzin ET, Cai SJ, Flick R, Hayes E, Ho K & Warrier S et al. 2012 Selective estrogen receptor-β (SERM-β) compounds modulate raphe nuclei tryptophan hydroxylase-1 (TPH-1) mRNA expression and cause antidepressant-like effects in the forced swim test. Neuropharmacology 63 10511063. (doi:10.1016/j.neuropharm.2012.07.004).

    • Search Google Scholar
    • Export Citation
  • Clarke RB, Howell A, Potten CS & Anderson E 1997 Dissociation between steroid receptor expression and cell proliferation in the human breast. Cancer Research 57 49874991.

    • Search Google Scholar
    • Export Citation
  • Cleveland AG, Oikarinen SI, Bynote KK, Marttinen M, Rafter JJ, Gustafsson JA, Roy SK, Pitot HC, Korach KS & Lubahn DB et al. 2009 Disruption of estrogen receptor signaling enhances intestinal neoplasia in Apc(Min/+) mice. Carcinogenesis 30 15811590. (doi:10.1093/carcin/bgp132).

    • Search Google Scholar
    • Export Citation
  • Couse JF & Korach KS 1999 Estrogen receptor null mice: what have we learned and where will they lead us? Endocrine Reviews 20 358