Abstract
The estrogen receptor α (ERα) is a transcription factor that mediates the biological effects of 17β-estradiol (E2). ERα transcriptional activity is also regulated by cytoplasmic signaling cascades. Here, several Gα protein subunits were tested for their ability to regulate ERα activity. Reporter assays revealed that overexpression of a constitutively active Gαo protein subunit potentiated ERα activity in the absence and presence of E2. Transient transfection of the human breast cancer cell line MCF-7 showed that Gαo augments the transcription of several ERα-regulated genes. Western blots of HEK293T cells transfected with ER±Gαo revealed that Gαo stimulated phosphorylation of ERK 1/2 and subsequently increased the phosphorylation of ERα on serine 118. In summary, our results show that Gαo, through activation of the MAPK pathway, plays a role in the regulation of ERα activity.
Introduction
Various agents regulate estrogen receptor α (ERα) activity in addition to 17β-estradiol (E2), including peptide growth factors (PGFs) such as epidermal growth factor (EGF) and insulin-like growth factor 1 (IGF1; Ignar-Trowbridge et al. 1992, 1993, 1996, Aronica & Katzenellenbogen 1993, Curtis et al. 1996, Kenney & Dickson 1996, Lee et al. 2001, Klotz et al. 2002). PGFs and E2 often operate in concert to potentiate/elevate ERα activity to a level above that observed with either factor alone (Ignar-Trowbridge et al. 1993, 1996). Mechanistic studies have revealed that cross talk between the PGF and E2 signaling cascades occurs at the level of ERα, in that PGF activation of cytoplasmic signaling cascades such as the phosphatidylinositol 3-kinase (PI3K)–AKT and MAPK signaling cascades (Dufourny et al. 1997, Bartucci et al. 2001, Dunn et al. 2001, Kato 2001, Brazil et al. 2002, Cantley 2002) ultimately leads to activation of ERα (Martin et al. 2000, Campbell et al. 2001). The MAPKs, including ERK1/2, c-jun N-terminal kinase (JNK), and p38 kinase, are a family of related kinases stimulated by numerous extracellular stimuli including mitogens, peptide hormones, cytokines, and cellular stress (Ballif & Blenis 2001).
Activation of the ERα by cytoplasmic signaling cascades is not limited to PGFs. Recent evidence suggests that factors that activate G-protein-coupled receptors (GPCRs) may also regulate ERα function. Dopamine has been shown to activate ER-mediated transcriptional activity in the absence (Power et al. 1991) or presence (Smith et al. 1993) of E2. Melatonin has been shown to inhibit breast cancer cell proliferation through modulation of ERα function (Ram et al. 1998, 2000b, Rato et al. 1999, Yuan et al. 2002, Girgert et al. 2003, del Rio et al. 2004). Indeed, melatonin may play a role in breast cancer (Sanchez-Barcelo et al. 2003), as high concentrations of melatonin in human breast cancer tissue have been correlated with good prognosis (Maestroni 1999). Also, the growth and proliferation of a number of breast cancers are influenced by β-adrenergic receptors (Cakir et al. 2002), and β-adrenergic stimulation has been shown to elicit classical ‘estrogenic’ responses, such as increases in uterine weight and ER levels (Re et al. 1993, Sukocheva et al. 2003).
Specificity of GPCR signaling is determined by the types of heterotrimeric GTP-binding proteins (G-proteins) that are recruited to, and activated by, an individual receptor (Rockman et al. 2002). The G-proteins consist of α-, β-, and γ-subunits, and they function as second messengers that initiate signaling cascades responsible for the regulation of numerous cellular events. The Gα subunit family includes four distinct subgroups, Gαs, Gαi/o, Gαq, and Gα12 (Downes & Gautam 1999, Radhika & Dhanasekaran 2001), and mutations in Gα subgroups have been linked to human diseases (Lania et al. 2001, Schulte & Fredholm 2003). Classically, Gαi inhibits and Gαs stimulates cAMP production. However, Gα signaling is not limited to the cAMP pathway, as all four Gα subgroups have been shown to regulate kinase-mediated signaling events, including the MAPK and PI3K–AKT pathways (Nava et al. 2002, Marinissen et al. 2003, Radeff-Huang et al. 2004). For example, sphingosine kinase type 1 (SPHK1), the enzyme that phosphorylates sphingosine to form sphingosine 1 phosphate, has been shown to regulate breast cancer cell proliferation and tumorigenesis, possibly through SPHK1's ability to regulate the level of phosphorylated AKT and ERK1/2 (Nava et al. 2002, Sukocheva et al. 2003).
Recent studies have demonstrated pathological Gα signaling in several disease types, including immune system dysfunction, infectious diseases, endocrine disorders, heart disease, and cancer (Kimple et al. 2011, Vassart & Costagliola 2011, Kaur et al. 2012). In the endocrine system, altered Gα has been found in a number of disorders, including those involving the thyroid, hypothalamus, pituitary, pancreas, ovaries, prostate, and breast (Livingstone et al. 1998, Lania et al. 2001, D'Souza et al. 2004, Cotta-Grand et al. 2009, Ogawa et al. 2009, Zhao et al. 2010). In cancer, aberrant Gαo signaling has been found in teratocarcinoma, glioblastoma, neuroblastoma, colon, kidney, lung, gastrointestinal stromaltumors, acute myeloid leukemia, ovarian, pancreas, thyroid, and breast cancers (Hurst & Hooks 2009, Lamba et al. 2009, Kimple et al. 2011). Recent findings have shed light on the role of Gαo signaling in breast carcinoma. Several somatic mutations in Gαo have been identified and characterized in patient breast biopsies (Ram et al. 2000a, Lamba et al. 2009, Kan et al. 2010, Garcia-Marcos et al. 2011). These mutations primarily result in a constitutively active Gαo phenotype in breast tumors, leading to increased Gαo signaling. However, the exact mechanism and consequence of enhanced Gαo signaling in breast cancer remains poorly characterized. A better understanding of the role of Gαo in ERα signaling would increase our understanding of the mechanisms of cancer biology. Given the ability of GPCRs to activate cytoplasmic signaling events that have previously been shown to potentiate ERα transcriptional activity, we hypothesized that G-protein-coupled signaling pathways may target and regulate the transcriptional activity of ERα. The recent identification of naturally occurring Gαo mutants led us to use a constitutively active Gαo vector to simulate the effects of the mutations found in clinical breast tumors. Given the relatively novel discovery of these mutations, in vitro and in vivo models of specific Gαo clinical mutations are currently unavailable. We demonstrated potentiation of ERα activity by Gαo, which culminated in increased expression of the estrogen target genes SDF1 (CXCL12), progesterone receptor (PR (PGR)), and BCL2. Mechanistic studies on Gαo revealed that this subunit activated the ERK1/2 signaling pathway, culminating in the phosphorylation of ERα.
There is emerging evidence linking the ER to nonclassical, rapid signaling events originating at the cell membrane followed by activation of downstream kinase cascades within the cytoplasm. This study focuses on the ability of the G-protein Gαo to modulate the activity of the ER and explores the cellular pathways through which this regulation takes place.
Materials and Methods
Reagents
DMEM, phenol red-free DMEM, fetal bovine serum (FBS), BME amino acids, MEM amino acids, l-glutamine, penicillin, streptomycin, and sodium pyruvate were obtained from GibcoBRL. Porcine insulin was purchased from Sigma, and charcoal-stripped (CS) FBS was obtained from HyClone (Logan, UT, USA). Effectene was purchased from Qiagen. E2 was obtained from Sigma. BrightGlo luciferase assay substrate was purchased from Promega. The lysis buffer used was mammalian protein extraction reagent (M-PER) from Pierce (Rockford, IL, USA). The Berthold AutoLumat Plus luminometer was used for luciferase assays.
Plasmids
Expression constructs for constitutively active Gα subunits (GTPase-deficient mutants of Gαo, Gαi2, Gαq, Gα12, Gα13, and Gα16) were generously provided by Dr Lynn Heasley (Heasley et al. 1996) or obtained through the UMR cDNA Resource Center (Rolla, MO, USA). We have previously described the use of dominant-negative mutant ERK2-DN, which was provided by Drs Melanie Cobb and Roger Davis (Frigo et al. 2004). The pcDNA3.1-ERα plasmids along with pERE-Luc plasmid, which contains three copies of the estrogen response element (ERE) linked to the luciferase gene, were purchased from Panomics (Fremont, CA, USA). ERα-S118A was provided by Benita Katzenellenbogen (Le Goff et al. 1994).
Cell culture
The two cell lines used were the ER-negative human embryonic kidney 293 (HEK293; Le Goff et al. 1994), and the ERα- and ERβ-positive human breast carcinoma (MCF-7) cell lines (Frigo et al. 2002). All were maintained in DMEM supplemented with 10% FBS, BME amino acids, MEM amino acids, l-glutamine, 100 units/ml penicillin, 100 units/ml streptomycin, sodium pyruvate, and 1×10−10 M porcine insulin under mycoplasma-free conditions at 37 °C in humidified 5% CO2. For described studies, cells were grown in phenol red-free DMEM supplemented with 5% CS FBS, and supplements as earlier, but without insulin (5% CS-DMEM) as described previously (Ignar-Trowbridge et al. 1993, Burow et al. 1999).
Transient transfection and luciferase assay
HEK293T and MCF-7 cells were transfected as described previously (Burow et al. 1999). Briefly, cells were plated into 24-well plates at 1×104 cells/well in 5% CS-DMEM and allowed to attach overnight. The next day, the cells were transfected with plasmids for 5 h in the serum/supplement-free DMEM using Effectene according to the manufacturers' protocols. The following amounts of DNA were added to each well: for ERE-luc assays, 0.2 μg ERE-luciferase, 0.3 μg G-proteins, 0.2 μg ER (S118A), and 0.1 μg ERE-luc. After 5 h, the cells were treated with vehicle control or E2 (10 pM). After 24 h, the treatment-containing medium was removed and 1× M-PER mammalian cell lysis buffer (Pierce; 150 μl) was added per well and gently shaken for 5 min at room temperature. Luciferase activity of 100 μl of cell extract was determined using BrightGlo luciferase assay substrate (Promega) in a Berthold luminometer. Data are presented as relative luminescence and are normalized as outlined in each figure. Each data point is the average of three separate measurements. Statistical analysis was performed using the two-sample T test in the Origin graphing program with a P value of 0.05.
Western blot analysis
HEK293 cells were maintained in 5% CS-DMEM and plated in 100×20 mm cell culture dishes at 50–80% confluency overnight. Cells were transfected with 1 μg ERα and either 1 μg empty vector or 1 μg Gαo. For RT-PCR analyses, MCF-7 cells were transfected with either empty vector or 2.0 μg Gαo. After 5 h, either steroid or vehicle was added. The cells were harvested after ∼18 h using M-PER lysis buffer containing a mixture of protease inhibitors (Roche) and phosphatase inhibitors (Sigma) on ice for 10 min. Equal volumes of lysates were added to SDS–PAGE loading buffer (Invitrogen) containing 1% β-mercaptoethanol and then boiled for 5 min. The proteins were electrophoresed on a Bis–Tris 4–12% gradient polyacrylamide gel (Invitrogen) and were subsequently transferred electrophoretically to a nitrocellulose membrane. The membrane was blocked with 5% nonfat dry milk solution in PBS and 0.1% Tween for 1 h at room temperature. The membrane was washed extensively and subsequently incubated overnight at 4 °C with rabbit antibodies to ERα (1:500 dilution; Santa Cruz Biotechnology); ERα-phoshpo-S118 (1:1000 dilution; Cell Signaling, Beverly, MA, USA); phosphorylated forms of ERK1/2, JNK, or p38 (1:1000; Cell Signaling Technology); or Rho-GDI (1:500 dilution; Santa Cruz Biotechnology). All antibodies were diluted in 5% BSA dissolved in 1× PBS+0.1% Tween. The next day, blots were washed in PBS+0.1% Tween and incubated with goat antirabbit antibodies conjugated to IRDye (1:10 000; Li-Cor, Lincoln, NE, USA) for 60 min at room temperature. Following three washes, immunoreactive proteins were scanned using the Li-Cor IR scanner set to 800 nm.
RT-PCR assays
Cell lysates were obtained as explained earlier. RNA was isolated using the RNAeasy miniprep kit (Qiagen). Second-strand cDNA synthesis was performed using 2 μg total RNA and the cDNA synthesis kit (Bio-Rad) according to the manufacturer's instructions. RT-PCR assays were assembled in 96-well plates using 5 μl of 1:10 dilution of the synthesized cDNA, 0.1 μg of 1:1 mixture of forward and reverse primers, and 1× SYBR-Green solution (Bio-Rad).
Results
G-proteins potentiate ERα activity
Activation of G-protein signaling can occur in response to classic GPCRs, growth factor/cytokine receptors, and steroid hormones (Rockman et al. 2002, Radeff-Huang et al. 2004). There are four major families of Gα signaling proteins: Gαi, Gαo, Gαq, and Gα12/13. Therefore, we chose the primary members of these four major families to screen for effects on ERα transcriptional activity. To examine the role of specific Gα subunits on ERα activity, expression vectors containing Gα subunits mutated to function in a constitutively active manner were used. MCF-7 breast carcinoma cells, which contain both the isoforms of ER, were transfected with empty vector (VEC), Gαi2, Gαo, Gαq, Gα12, Gα13, and Gα16, along with an estrogen-responsive luciferase reporter construct. The ability of 1 nM E2 to activate the ER in VEC cells was normalized to 1.0. The expression of Gαo, Gαq, Gα12, and Gα13 potentiated estrogen-bound ERα activity (Fig. 1). Interestingly, only Gαo overexpression resulted in both a ligand-independent and a ligand-dependent increase in ERα activity.
Gαo potentiates ER-mediated gene transcription
We focused the remaining studies on Gαo because of its ability to augment ER activity, both in the presence and absence of estrogen. To determine whether Gαo overexpression affects transcription of endogenously ER-regulated genes, we transfected MCF-7 cells with either VEC or constitutively active Gαo in the absence or presence of 1 nM E2 followed by real-time PCR. In the vector cells, the addition of 1 nM E2 caused an expected increase in the transcription of ERα-regulated genes pS2 (TFF1; fourfold), SDF1 (fivefold), PR (33-fold), and BCL2 (1.8-fold) (Fig. 2A). Transfection of Gαo resulted in an increase in the transcription of PR (2.7-fold), SDF1 (1.9-fold), and BCL2 (twofold) in the absence of estrogen; however, no change was seen in the expression of pS2 after overexpression of Gαo (Fig. 2A). We believe that the latter effect is due, in part, to altered promoter contexts of each ER-mediated gene. Curiously, the addition of Gαo did not increase the transcription of PR, pS2, or SDF1 beyond the level induced by estrogen. It is possible that, with these particular genes, treatment with E2 resulted in a maximum transcriptional response in this cell context. It is important to note that in the cases of PR and SDF1, Gαo alone was able to induce transcription, although to a lesser level than that seen with E2. Finally, we found that the Gαo-mediated increase in expression of these ER-regulated genes was not due to an increase in ERα transcription levels in cells that were transfected with Gαo (Fig. 2B).
Gαo activates the ERK pathway
Several cell signaling cascades have been shown to cross talk with ERα (Dufourny et al. 1997, Martin et al. 2000, Bartucci et al. 2001, Campbell et al. 2001, Dunn et al. 2001, Kato 2001, Brazil et al. 2002, Cantley 2002). To determine whether Gαo activates ERK1/2, p38, or JNK kinases, western blot analysis was performed using phosphorylation-specific antibodies to detect the active forms of these kinases. Transfection of HEK293T cells with constitutively active Gαo resulted in an increase in the phosphorylation of ERK1/2 but not JNK or p38 (Fig. 3). Similar results were seen in MCF-7 cells (data not shown).
Gαo affects ERα's phosphorylation status
Our results indicate that Gαo activates the ERK1/2 pathway. Because ERα contains an ERK phosphorylation site at serine 118, we tested the ability of Gαo overexpression to increase phosphorylation of the ER at this site. We used western blot analysis of HEK293T cells that were transfected with ERα, with or without Gαo. Using densitometry, we divided the overall increase in ER phosphorylation at serine 118 by the total amount of ER within the cell. Our results indicate that Gαo caused an increase in ERα phosphorylation at Ser118 of approximately twofold (Fig. 4A and B).
We validated these findings using a serine 118 to alanine point mutant of ERα in ERE reporter assays. Transfection of the mutant into HEK293T cells showed that alteration of this residue resulted in loss of the Gαo effect on ERα activity (Fig. 5A). We also used a dominant-negative mutant of ERK2 and found that this mutant was able to decrease Gαo's potentiation of ligand-bound ERα by ∼30% (Fig. 5B). Finally, we showed that when ERK was pharmacologically inhibited by UO126, Gαo's potentiation of ligand-bound ERα activity fell by ∼30% while the inhibitor had no effect on E2-stimulated ERα activity (Fig. 5C). These results confirm that an intact ERK1/2 pathway is necessary for the maximal Gαo effect on ER activity.
Discussion
Rapid, ‘nongenomic’ signaling of estrogen has been the focus of intense research in recent years. E2 has been shown to rapidly (seconds to minutes) activate many signaling molecules (Cheskis 2004, Shupnik 2004, Levin 2005), such as i) IGF1 receptor and EGF receptor, ii) p21ras and Raf, iii) MAPK and Akt, iv) protein kinase C, v) intracellular calcium transients, vi) nitric oxide and prolactin secretion, and vii) Maxi-K channels. However, the mechanisms of these activations remain unclear. Although several groups have proposed that alternative E2-binding proteins exist within the plasma membrane, accumulating evidence supports the classical full-length ERα as the membrane E2-binding protein. Current studies are underway to determine what accessory proteins are associated with and transmit the signal of ERα at the membrane to intracellular signaling cascades and ultimately to the nucleus.
Several laboratories have evidence suggesting that G-proteins provide this link between the ER at the plasma membrane and estrogen-induced rapid kinase activation within the cell. Wyckoff et al. (2001) showed that endothelial nitric oxide synthase (eNOS) stimulation was mediated by plasma membrane-tethered E2-bound ERα and that this stimulation was dependent on ER coupling with Gαi. Razandi et al. (2003) demonstrated that ER interaction with Gαi controls rapid signaling through Src–ERK. In addition, ER has been shown to interact with Gα13, and this interaction stimulates the small GTPase RhoA and the downstream effector Rho-associated kinase (Simoncini et al. 2006).
Here, we investigated the role of the G-protein subunit Gαo as a mediator of ERα transcriptional activity. We have shown using ERE-luciferase assays that a constitutively active Gαo is able to potentiate ERα activity in the absence and presence of E2 (Fig. 1). Using RT-PCR analysis of MCF-7 cells transfected with the constitutively active Gαo, we showed a Gαo-mediated potentiation of transcription of the ERα-regulated genes BCL2, SDF1, and PR (Fig. 2). The increase in transcription of these genes was not the result of a Gαo-mediated increase in ERα transcription.
Because several laboratories have previously shown that G-proteins mediate phosphorylation of cellular signaling kinases and that these phosphorylation events can impact ERα activity, we used western blot analysis of HEK293T cells transfected with either pCDNA or constitutively active Gαo to probe the phosphorylation status of several major signaling kinases. We showed that overexpression of Gαo resulted in increased phosphorylation and hence activation of ERK1/2 kinase but not p38 kinase or JNK kinase (Fig. 3). This result led us to explore the phosphorylation status of the ER in the presence and absence of constitutively active Gαo. Again, we used western blot analysis of HEK293T cells that were transfected with ERα±Gαo. Our results indicated an increase in phosphorylation of ERα at serine 118, a known ERK1/2 site (Fig. 4A). Our densitometric analysis showed that serine 118 was in fact hyperphosphorylated as a result of Gαo overexpression (Fig. 4B). In addition, ERE-luciferase assays using an ERK-DN construct, a specific ERK pharmacological inhibitor, or a serine 118 to alanine point mutant of ERα, we confirmed that serine 118 is necessary for a maximal Gαo effect (Fig. 5A, B and C). Previous laboratories have shown a direct link between ERα phosphorylation at serine 118 and an increased ability of the receptor to dimerize and bind coactivators (Sheeler et al. 2003). Therefore, the increased activity of ERα in the presence of Gαo can be partially explained by our evidence linking Gαo to activation of ERK1/2, followed by hyperphosphorylation of ER at serine 118, and an increase in ER dimerization and coactivator recruitment.
Given that there are multiple physiological mutations resulting in increased Gαo activity, we chose to use a constitutively active Gαo variant to mimic increased Gαo activity in our cell systems. The Gαo mutant used in our study is GTPase deficient, resulting in a constitutively active Gαo phenotype, similar to those found in human tumors. There are several known downstream effectors of Gαo, including protein kinase C, Src, Stat3, and c-mos. Gαo uses these various effectors based on initiating signaling event and tissue type (Kroll et al. 1991, Diverse-Pierluissi et al. 1997, Ram et al. 2000a). While Gαo has been shown to use MAPKs to exert its effects, the role of MAPKs in Gαo signaling is less clear. There are varying reports in the literature regarding Gαo activation of MAPKs. For example, some studies have reported that Gαo can activate p38 and Erk1/2 signaling while others have shown that Gαo inhibits Erk1/2 activation (Yamauchi et al. 1997, Liu et al. 2002). Conversely, several studies have demonstrated a relationship between Gαo and JNK and Akt activity (Goel et al. 2004, He et al. 2006). To date, the majority of these studies have excluded breast tissue in their analysis and ability of Gαo to regulate MAPK activity in ER-positive cells has not been well characterized.
The vast majority of breast cancers are ERα positive, and pathological ERα signaling is the primary growth cascade in these cancers. Therefore, we chose to investigate the relationship between Gαo and ER pathways. Our findings demonstrate that Gαo can alter ERα signaling. Furthermore, we report a relationship between MAPK signaling and Gαo. MAPKs are known to promote hormone-independent tumor growth and endocrine therapy resistance (Musgrove & Sutherland 2009). Furthermore, ligand-independent tumor growth is a hallmark of breast cancer progression (Musgrove & Sutherland 2009). Several mechanisms of ligand-independent activation of the ER have been identified, including increased endogenous activation of the ER through MAPK-specific phosphorylation (Kato et al. 1995). In breast cancer, ERK1/2 is known to activate the ER at the S118 phosphorylation site, which results in enhanced proliferation and tumorigenesis (Kato et al. 1995, Chen et al. 2000). Furthermore, phosphorylation of the ER by ERK1/2 promotes hormone-independent tumor growth and endocrine therapy resistance (Campbell et al. 2001). In the clinic, increased S118 phosphorylation is associated with decreased survival and the efficacy of therapeutic agents may be correlated with activation at this site (Yamashita et al. 2008, Zoubir et al. 2008). Our findings that Gαo activates ERK1/2, but not p38 or JNK, are similar to previously published studies in prostate but contrast those found in pituitary, kidney, and neuronal cells (Yamauchi et al. 1997, Liu et al. 2002, 2011, He et al. 2006). The fact that mutation of S118 to alanine blocks the Gαo effect entirely while pharmacological or molecular inhibition of ERK1/2 only partially does so implies that Gαo may influence other kinases within the cell that ultimately affect phosphorylation of ER at serine 118. These results provide evidence for a tissue-type-specific role of Gαo activation of MAPK in ER-positive cells. Identification of Gαo-induced ER activity mediated by ERK1/2 could provide a novel target for ligand-independent tumors, as well as endocrine therapy-resistant breast cancer. Although further study is needed to determine the therapeutic potential of this pathway, identification and characterization of secondary ER growth pathways are necessary for the development of novel anticancer agents. Our findings that Gαo promotes ER signaling through the ERK signaling cascade may account for the previously published findings of increased tumorigenesis and tumor growth associated with Gαo (Nguyen et al. 2002, Prevost et al. 2006, Garcia-Marcos et al. 2011, Lin et al. 2011). Increased understanding of the interplay between Gαo and the ER may also identify novel breast cancer treatments that target ER signaling. For example, one recent study used a Gα inhibitor as an anticancer agent across several cancer types, including ER-positive breast cancer (Prevost et al. 2006). Given the high rate of endocrine therapy resistance, anticancer therapies directed against Gαo, and indirectly targeting the ER, may be an effective therapeutic strategy. In conclusion, we have shown that Gαo plays a significant role in the regulation of ERα activity, both in the presence and absence of E2. Specifically, Gαo can enhance ERK1/2 phosphorylation. This activation leads to specific phosphorylation of the ER at serine 118, culminating in a marked increase in ER-mediated transcriptional activity. Our results also indicated that Gαo can induce ER phosphorylation in the absence of E2, which is especially relevant considering the clinical phenomenon of estrogen-independent proliferation of breast cancer cells. It is important to note that because of the constitutively active nature of the Gαo construct, we are circumventing the involvement of the GPCR, which is endogenously associated with Gαo. Further studies using wild-type Gαo overexpression will explore the role, if any, of a GPCR in the Gαo-ER mechanism. In any case, our results showing a direct link between an activated G-protein- and estrogen-independent phosphorylation and activation of the ER could open a new avenue of research regarding treatment of estrogen-independent and/or tamoxifen-resistant breast cancer.
Declaration of interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
Funding
This work was supported by National Institutes of Health Grant DK 059389 (to M E B), Department of Energy Grant DE-FC26-00NT40843 (to J A M and M E B), Center for Disease Control Grant RO6/CCR419466-02 (to J A M), the Center for Bioenvironmental Research at Tulane and Xavier Universities (to J A M), the Office of Naval Research Grant N00014-99-1-0763 (J A M and M E B), the Louisiana Cancer Research Consortium (M R B), and the Susan G Komen Breast Cancer Foundation Grant BCTR0601198 (M E B) and National Institutes of Health grant K01 DK084205 (DEF).
Acknowledgements
The authors would like to thank Drs Roger Davis, Pierre Chambon, Donald P McDonnell, Jan-Ake Gustaffson, Benita Katzenellenbogen, Melanie Cobb, and Diane M Klotz for the generous provision of expression constructs and cell lines.
References
Aronica SM & Katzenellenbogen BS 1993 Stimulation of estrogen receptor-mediated transcription and alteration in the phosphorylation state of the rat uterine estrogen receptor by estrogen, cyclic adenosine monophosphate, and insulin-like growth factor-I. Molecular Endocrinology 7 743–752. doi:10.1210/me.7.6.743.
Ballif BA & Blenis J 2001 Molecular mechanisms mediating mammalian mitogen-activated protein kinase (MAPK) kinase (MEK)–MAPK cell survival signals. Cell Growth & Differentiation 12 397–408.
Bartucci M, Morelli C, Mauro L, Ando S & Surmacz E 2001 Differential insulin-like growth factor I receptor signaling and function in estrogen receptor (ER)-positive MCF-7 and ER-negative MDA-MB-231 breast cancer cells. Cancer Research 61 6747–6754.
Brazil DP, Park J & Hemmings BA 2002 PKB binding proteins. Getting in on the Akt. Cell 111 293–303. doi:10.1016/S0092-8674(02)01083-8.
Burow ME, Tang Y, Collins-Burow BM, Krajewski S, Reed JC, McLachlan JA & Beckman BS 1999 Effects of environmental estrogens on tumor necrosis factor alpha-mediated apoptosis in MCF-7 cells. Carcinogenesis 20 2057–2061. doi:10.1093/carcin/20.11.2057.
Cakir Y, Plummer HK III, Tithof PK & Schuller HM 2002 Beta-adrenergic and arachidonic acid-mediated growth regulation of human breast cancer cell lines. International Journal of Oncology 21 153–157.
Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S & Nakshatri H 2001 Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: a new model for anti-estrogen resistance. Journal of Biological Chemistry 276 9817–9824. doi:10.1074/jbc.M010840200.
Cantley LC 2002 The phosphoinositide 3-kinase pathway. Science 296 1655–1657. doi:10.1126/science.296.5573.1655.
Chen D, Riedl T, Washbrook E, Pace PE, Coombes RC, Egly JM & Ali S 2000 Activation of estrogen receptor alpha by S118 phosphorylation involves a ligand-dependent interaction with TFIIH and participation of CDK7. Molecular Cell 6 127–137.
Cheskis BJ 2004 Regulation of cell signalling cascades by steroid hormones. Journal of Cellular Biochemistry 93 20–27. doi:10.1002/jcb.20180.
Cotta-Grand N, Rovere C, Guyon A, Cervantes A, Brau F & Nahon JL 2009 Melanin-concentrating hormone induces neurite outgrowth in human neuroblastoma SH-SY5Y cells through p53 and MAPKinase signaling pathways. Peptides 30 2014–2024. doi:10.1016/j.peptides.2009.06.015.
Curtis SW, Washburn T, Sewall C, DiAugustine R, Lindzey J, Couse JF & Korach KS 1996 Physiological coupling of growth factor and steroid receptor signaling pathways: estrogen receptor knockout mice lack estrogen-like response to epidermal growth factor. PNAS 93 12626–12630. doi:10.1073/pnas.93.22.12626.
Diverse-Pierluissi M, Remmers AE, Neubig RR & Dunlap K 1997 Novel form of crosstalk between G protein and tyrosine kinase pathways. PNAS 94 5417–5421. doi:10.1073/pnas.94.10.5417.
Downes GB & Gautam N 1999 The G protein subunit gene families. Genomics 62 544–552. doi:10.1006/geno.1999.5992.
D'Souza DN, Zhang Y, Damjanoska KJ, Carrasco GA, Sullivan NR, Garcia F, Battaglia G, Doncarlos LL, Muma NA & Van de Kar LD 2004 Estrogen reduces serotonin-1A receptor-mediated oxytocin release and Galpha(i/o/z) proteins in the hypothalamus of ovariectomized rats. Neuroendocrinology 80 31–41. doi:10.1159/000080795.
Dufourny B, Alblas J, van Teeffelen HA, van Schaik FM, van der Burg B, Steenbergh PH & Sussenbach JS 1997 Mitogenic signaling of insulin-like growth factor I in MCF-7 human breast cancer cells requires phosphatidylinositol 3-kinase and is independent of mitogen-activated protein kinase. Journal of Biological Chemistry 272 31163–31171. doi:10.1074/jbc.272.49.31163.
Dunn SE, Torres JV, Oh JS, Cykert DM & Barrett JC 2001 Up-regulation of urokinase-type plasminogen activator by insulin-like growth factor-I depends upon phosphatidylinositol-3 kinase and mitogen-activated protein kinase kinase. Cancer Research 61 1367–1374.
Frigo DE, Burow ME, Mitchell KA, Chiang TC & McLachlan JA 2002 DDT and its metabolites alter gene expression in human uterine cell lines through estrogen receptor-independent mechanisms. Environmental Health Perspectives 110 1239–1245. doi:10.1289/ehp.021101239.
Frigo DE, Tang Y, Beckman BS, Scandurro AB, Alam J, Burow ME & McLachlan JA 2004 Mechanism of AP-1-mediated gene expression by select organochlorines through the p38 MAPK pathway. Carcinogenesis 25 249–261. doi:10.1093/carcin/bgh009.
Garcia-Marcos M, Ghosh P & Farquhar MG 2011 Molecular basis of a novel oncogenic mutation in GNAO1. Oncogene 30 2691–2696. doi:10.1038/onc.2010.645.
Girgert R, Bartsch C, Hill SM, Kreienberg R & Hanf V 2003 Tracking the elusive antiestrogenic effect of melatonin: a new methodological approach. Neuro Endocrinology Letters 24 440–444.
Goel R, Phillips-Mason PJ, Gardner A, Raben DM & Baldassare JJ 2004 Alpha-thrombin-mediated phosphatidylinositol 3-kinase activation through release of Gbetagamma dimers from Galphaq and Galphai2. Journal of Biological Chemistry 279 6701–6710. doi:10.1074/jbc.M308753200.
He JC, Neves SR, Jordan JD & Iyengar R 2006 Role of the Go/i signaling network in the regulation of neurite outgrowth. Canadian Journal of Physiology and Pharmacology 84 687–694. doi:10.1139/y06-025.
Heasley L, Storey B, Fanger G, Butterfield L, Zamarripa J, Blumberg D & Maue R 1996 GTPase-deficient G alpha 16 and G alpha q induce PC12 cell differentiation and persistent activation of cJun NH2-terminal kinases. Molecular and Cellular Biology 16 648–656.
Hurst JH & Hooks SB 2009 Regulator of G-protein signaling (RGS) proteins in cancer biology. Biochemical Pharmacology 78 1289–1297. doi:10.1016/j.bcp.2009.06.028.
Ignar-Trowbridge DM, Nelson KG, Bidwell MC, Curtis SW, Washburn TF, McLachlan JA & Korach KS 1992 Coupling of dual signaling pathways: epidermal growth factor action involves the estrogen receptor. PNAS 89 4658–4662. doi:10.1073/pnas.89.10.4658.
Ignar-Trowbridge DM, Teng CT, Ross KA, Parker MG, Korach KS & McLachlan JA 1993 Peptide growth factors elicit estrogen receptor-dependent transcriptional activation of an estrogen-responsive element. Molecular Endocrinology 7 992–998. doi:10.1210/me.7.8.992.
Ignar-Trowbridge DM, Pimentel M, Parker MG, McLachlan JA & Korach KS 1996 Peptide growth factor cross-talk with the estrogen receptor requires the A/B domain and occurs independently of protein kinase C or estradiol. Endocrinology 137 1735–1744. doi:10.1210/en.137.5.1735.
Kan Z, Jaiswal BS, Stinson J, Janakiraman V, Bhatt D, Stern HM, Yue P, Haverty PM, Bourgon R & Zheng J et al. 2010 Diverse somatic mutation patterns and pathway alterations in human cancers. Nature 466 869–873. doi:10.1038/nature09208.
Kato S 2001 Estrogen receptor-mediated cross-talk with growth factor signaling pathways. Breast Cancer 8 3–9. doi:10.1007/BF02967472.
Kato S, Endoh H, Masuhiro Y, Kitamoto T, Uchiyama S, Sasaki H, Masushige S, Gotoh Y, Nishida E & Kawashima H et al. 1995 Activation of the estrogen receptor through phosphorylation by mitogen-activated protein kinase. Science 270 1491–1494. doi:10.1126/science.270.5241.1491.
Kaur K, Parra S, Chen R, Charbeneau RA, Wade SM, Jay PY & Neubig RR 2012 Galpha(i2) signaling: friend or foe in cardiac injury and heart failure? Naunyn-Schmiedeberg's Archives of Pharmacology 385 443–453. doi:10.1007/s00210-011-0705-z.
Kenney NJ & Dickson RB 1996 Growth factor and sex steroid interactions in breast cancer. Journal of Mammary Gland Biology and Neoplasia 1 189–198. doi:10.1007/BF02013642.
Kimple AJ, Bosch DE, Giguere PM & Siderovski DP 2011 Regulators of G-protein signaling and their Galpha substrates: promises and challenges in their use as drug discovery targets. Pharmacological Reviews 63 728–749. doi:10.1124/pr.110.003038.
Klotz DM, Hewitt SC, Ciana P, Raviscioni M, Lindzey JK, Foley J, Maggi A, DiAugustine RP & Korach KS 2002 Requirement of estrogen receptor-alpha in insulin-like growth factor-1 (IGF-1)-induced uterine responses and in vivo evidence for IGF-1/estrogen receptor cross-talk. Journal of Biological Chemistry 277 8531–8537. doi:10.1074/jbc.M109592200.
Kroll SD, Omri G, Landau EM & Iyengar R 1991 Activated alpha subunit of Go protein induces oocyte maturation. PNAS 88 5182–5186. doi:10.1073/pnas.88.12.5182.
Lamba S, Felicioni L, Buttitta F, Bleeker FE, Malatesta S, Corbo V, Scarpa A, Rodolfo M, Knowles M & Frattini M et al. 2009 Mutational profile of GNAQQ209 in human tumors. PLoS ONE 4 e6833 doi:10.1371/journal.pone.0006833.
Lania A, Mantovani G & Spada A 2001 G protein mutations in endocrine diseases. European Journal of Endocrinology 145 543–559. doi:10.1530/eje.0.1450543.
Lee AV, Cui X & Oesterreich S 2001 Cross-talk among estrogen receptor, epidermal growth factor, and insulin-like growth factor signaling in breast cancer. Clinical Cancer Research 7 4429s–4435s.(discussion 4411s–4412s)
Le Goff P, Montano MM, Schodin DJ & Katzenellenbogen BS 1994 Phosphorylation of the human estrogen receptor. Identification of hormone-regulated sites and examination of their influence on transcriptional activity. Journal of Biological Chemistry 269 4458–4466.
Levin ER 2005 Integration of the extranuclear and nuclear actions of estrogen. Molecular Endocrinology 19 1951–1959. doi:10.1210/me.2004-0390.
Lin C, Ear J, Pavlova Y, Mittal Y, Kufareva I, Ghassemian M, Abagyan R, Garcia-Marcos M & Ghosh P 2011 Tyrosine phosphorylation of the Galpha-interacting protein GIV promotes activation of phosphoinositide 3-kinase during cell migration. Science Signaling 4 ra64 doi:10.1126/scisignal.2002049.
Liu JC, Baker RE, Sun C, Sundmark VC & Elsholtz HP 2002 Activation of Go-coupled dopamine D2 receptors inhibits ERK1/ERK2 in pituitary cells. A key step in the transcriptional suppression of the prolactin gene. Journal of Biological Chemistry 277 35819–35825. doi:10.1074/jbc.M202920200.
Liu J, Youn H, Yang J, Du N, Liu J, Liu H & Li B 2011 G-protein alpha-s and -12 subunits are involved in androgen-stimulated PI3K activation and androgen receptor transactivation in prostate cancer cells. Prostate 71 1276–1286.
Livingstone JD, Lerant A & Freeman ME 1998 Ovarian steroids modulate responsiveness to dopamine and expression of G-proteins in lactotropes. Neuroendocrinology 68 172–179. doi:10.1159/000054363.
Maestroni GJ 1999 Therapeutic potential of melatonin in immunodeficiency states, viral diseases, and cancer. Advances in Experimental Medicine and Biology 467 217–226.
Marinissen MJ, Servitja JM, Offermanns S, Simon MI & Gutkind JS 2003 Thrombin protease-activated receptor-1 signals through Gq- and G13-initiated MAPK cascades regulating c-Jun expression to induce cell transformation. Journal of Biological Chemistry 278 46814–46825. doi:10.1074/jbc.M305709200.
Martin MB, Franke TF, Stoica GE, Chambon P, Katzenellenbogen BS, Stoica BA, McLemore MS, Olivo SE & Stoica A 2000 A role for Akt in mediating the estrogenic functions of epidermal growth factor and insulin-like growth factor I. Endocrinology 141 4503–4511. doi:10.1210/en.141.12.4503.
Musgrove EA & Sutherland RL 2009 Biological determinants of endocrine resistance in breast cancer. Nature Reviews Cancer 9 631–643. doi:10.1038/nrc2713.
Nava VE, Hobson JP, Murthy S, Milstien S & Spiegel S 2002 Sphingosine kinase type 1 promotes estrogen-dependent tumorigenesis of breast cancer MCF-7 cells. Experimental Cell Research 281 115–127. doi:10.1006/excr.2002.5658.
Nguyen QD, Faivre S, Bruyneel E, Rivat C, Seto M, Endo T, Mareel M, Emami S & Gespach C 2002 RhoA- and RhoD-dependent regulatory switch of Galpha subunit signaling by PAR-1 receptors in cellular invasion. FASEB Journal 16 565–576. doi:10.1096/fj.01-0525com.
Ogawa T, Forero M, Burgon PG, Kuroski de Bold ML, Georgalis T & de Bold AJ 2009 Role of potassium channels in stretch-promoted atrial natriuretic factor secretion. Journal of the American Society of Hypertension 3 9–18. doi:10.1016/j.jash.2008.07.002.
Power RF, Mani SK, Codina J, Conneely OM & O'Malley BW 1991 Dopaminergic and ligand-independent activation of steroid hormone receptors. Science 254 1636–1639. doi:10.1126/science.1749936.
Prevost GP, Lonchampt MO, Holbeck S, Attoub S, Zaharevitz D, Alley M, Wright J, Brezak MC, Coulomb H & Savola A et al. 2006 Anticancer activity of BIM-46174, a new inhibitor of the heterotrimeric Galpha/Gbetagamma protein complex. Cancer Research 66 9227–9234. doi:10.1158/0008-5472.CAN-05-4205.
Radeff-Huang J, Seasholtz TM, Matteo RG & Brown JH 2004 G protein mediated signaling pathways in lysophospholipid induced cell proliferation and survival. Journal of Cellular Biochemistry 92 949–966. doi:10.1002/jcb.20094.
Radhika V & Dhanasekaran N 2001 Transforming G proteins. Oncogene 20 1607–1614. doi:10.1038/sj.onc.1204274.
Ram PT, Kiefer T, Silverman M, Song Y, Brown GM & Hill SM 1998 Estrogen receptor transactivation in MCF-7 breast cancer cells by melatonin and growth factors. Molecular and Cellular Endocrinology 141 53–64. doi:10.1016/S0303-7207(98)00095-1.
Ram PT, Horvath CM & Iyengar R 2000a Stat3-mediated transformation of NIH-3T3 cells by the constitutively active Q205L Galphao protein. Science 287 142–144. doi:10.1126/science.287.5450.142.
Ram PT, Yuan L, Dai J, Kiefer T, Klotz DM, Spriggs LL & Hill SM 2000b Differential responsiveness of MCF-7 human breast cancer cell line stocks to the pineal hormone, melatonin. Journal of Pineal Research 28 210–218. doi:10.1034/j.1600-079X.2000.280403.x.
Rato AG, Pedrero JG, Martinez MA, del Rio B, Lazo PS & Ramos S 1999 Melatonin blocks the activation of estrogen receptor for DNA binding. FASEB Journal 13 857–868.
Razandi M, Alton G, Pedram A, Ghonshani S, Webb P & Levin ER 2003 Identification of a structural determinant necessary for the localization and function of estrogen receptor alpha at the plasma membrane. Molecular and Cellular Biology 23 1633–1646. doi:10.1128/MCB.23.5.1633-1646.2003.
Re G, Badino P, Dacasto M, Di Carlo F & Girardi C 1993 Regulation of uterine estrogen receptors (ER) by beta-adrenergic stimulation in immature rats. Journal of Veterinary Pharmacology and Therapeutics 16 328–334. doi:10.1111/j.1365-2885.1993.tb00179.x.
del Rio B, Garcia Pedrero JM, Martinez-Campa C, Zuazua P, Lazo PS & Ramos S 2004 Melatonin, an endogenous-specific inhibitor of estrogen receptor alpha via calmodulin. Journal of Biological Chemistry 279 38294–38302. doi:10.1074/jbc.M403140200.
Rockman HA, Koch WJ & Lefkowitz RJ 2002 Seven-transmembrane-spanning receptors and heart function. Nature 415 206–212. doi:10.1038/415206a.
Sanchez-Barcelo EJ, Cos S, Fernandez R & Mediavilla MD 2003 Melatonin and mammary cancer: a short review. Endocrine-Related Cancer 10 153–159. doi:10.1677/erc.0.0100153.
Schulte G & Fredholm BB 2003 The G(s)-coupled adenosine A(2B) receptor recruits divergent pathways to regulate ERK1/2 and p38. Experimental Cell Research 290 168–176. doi:10.1016/S0014-4827(03)00324-0.
Sheeler CQ, Singleton DW & Khan SA 2003 Mutation of serines 104, 106, and 118 inhibits dimerization of the human estrogen receptor in yeast. Endocrine Research 29 237–255. doi:10.1081/ERC-120022321.
Shupnik MA 2004 Crosstalk between steroid receptors and the c-Src-receptor tyrosine kinase pathways: implications for cell proliferation. Oncogene 23 7979–7989. doi:10.1038/sj.onc.1208076.
Simoncini T, Scorticati C, Mannella P, Fadiel A, Giretti MS, Fu XD, Baldacci C, Garibaldi S, Caruso A & Fornari L et al. 2006 Estrogen receptor alpha interacts with Galpha13 to drive actin remodeling and endothelial cell migration via the RhoA/Rho kinase/moesin pathway. Molecular Endocrinology 20 1756–1771. doi:10.1210/me.2005-0259.
Smith CL, Conneely OM & O'Malley BW 1993 Modulation of the ligand-independent activation of the human estrogen receptor by hormone and antihormone. PNAS 90 6120–6124. doi:10.1073/pnas.90.13.6120.
Sukocheva OA, Wang L, Albanese N, Pitson SM, Vadas MA & Xia P 2003 Sphingosine kinase transmits estrogen signaling in human breast cancer cells. Molecular Endocrinology 17 2002–2012. doi:10.1210/me.2003-0119.
Vassart G & Costagliola S 2011 G protein-coupled receptors: mutations and endocrine diseases. Nature Reviews Endocrinology 7 362–372. doi:10.1038/nrendo.2011.20.
Wyckoff MH, Chambliss KL, Mineo C, Yuhanna IS, Mendelsohn ME, Mumby SM & Shaul PW 2001 Plasma membrane estrogen receptors are coupled to endothelial nitric-oxide synthase through Galpha(i). Journal of Biological Chemistry 276 27071–27076. doi:10.1074/jbc.M100312200.
Yamashita H, Nishio M, Toyama T, Sugiura H, Kondo N, Kobayashi S, Fujii Y & Iwase H 2008 Low phosphorylation of estrogen receptor alpha (ERalpha) serine 118 and high phosphorylation of ERalpha serine 167 improve survival in ER-positive breast cancer. Endocrine-Related Cancer 15 755–763. doi:10.1677/ERC-08-0078.
Yamauchi J, Nagao M, Kaziro Y & Itoh H 1997 Activation of p38 mitogen-activated protein kinase by signaling through G protein-coupled receptors. Involvement of Gbetagamma and Galphaq/11 subunits. Journal of Biological Chemistry 272 27771–27777. doi:10.1074/jbc.272.44.27771.
Yuan L, Collins AR, Dai J, Dubocovich ML & Hill SM 2002 MT(1) melatonin receptor overexpression enhances the growth suppressive effect of melatonin in human breast cancer cells. Molecular and Cellular Endocrinology 192 147–156. doi:10.1016/S0303-7207(02)00029-1.
Zhao A, Ohara-Imaizumi M, Brissova M, Benninger RK, Xu Y, Hao Y, Abramowitz J, Boulay G, Powers AC & Piston D et al. 2010 Gαo represses insulin secretion by reducing vesicular docking in pancreatic beta-cells. Diabetes 59 2522–2529. doi:10.2337/db09-1719.
Zoubir M, Mathieu MC, Mazouni C, Liedtke C, Corley L, Geha S, Bouaziz J, Spielmann M, Drusche F & Symmans WF et al. 2008 Modulation of ER phosphorylation on serine 118 by endocrine therapy: a new surrogate marker for efficacy. Annals of Oncology 19 1402–1406. doi:10.1093/annonc/mdn151.