Next Article in Journal
MicroRNA as Possible Mediators of the Synergistic Effect of Celecoxib and Glucosamine Sulfate in Human Osteoarthritic Chondrocyte Exposed to IL-1β
Next Article in Special Issue
Androgen Receptor in Hormone Receptor-Positive Breast Cancer
Previous Article in Journal
Zinc Oxide Nanoparticles Affect Early Seedlings’ Growth and Polar Metabolite Profiles of Pea (Pisum sativum L.) and Wheat (Triticum aestivum L.)
Previous Article in Special Issue
Modulation of JAK-STAT Signaling by LNK: A Forgotten Oncogenic Pathway in Hormone Receptor-Positive Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

GPER: An Estrogen Receptor Key in Metastasis and Tumoral Microenvironments

by
Ana Carolina Tirado-Garibay
,
Elba Andrea Falcón-Ruiz
,
Alejandra Ochoa-Zarzosa
and
Joel E. López-Meza
*
Centro Multidisciplinario de Estudios en Biotecnología—FMVZ, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58893, Mexico
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(19), 14993; https://doi.org/10.3390/ijms241914993
Submission received: 30 August 2023 / Revised: 2 October 2023 / Accepted: 5 October 2023 / Published: 8 October 2023
(This article belongs to the Special Issue Hormone Receptor in Breast Cancer)

Abstract

:
Estrogens and their role in cancer are well-studied, and some cancer types are classified in terms of their response to them. In recent years, a G protein-coupled estrogen receptor (GPER) has been described with relevance in cancer. GPER is a pleiotropic receptor with tissue-specific activity; in normal tissues, its activation is related to correct development and homeostasis, while in cancer cells, it can be pro- or anti-tumorigenic. Also, GPER replaces estrogen responsiveness in estrogen receptor alpha (ERα)-lacking cancer cell lines. One of the most outstanding activities of GPER is its role in epithelial–mesenchymal transition (EMT), which is relevant for metastasis development. In addition, the presence of this receptor in tumor microenvironment cells contributes to the phenotypic plasticity required for the dissemination and maintenance of tumors. These characteristics suggest that GPER could be a promising therapeutic target for regulating cancer development. This review focuses on the role of GPER in EMT in tumorigenic and associated cells, highlighting its role in relation to the main hallmarks of cancer and possible therapeutic options.

1. The G Protein-Coupled Estrogen Receptor

In the late 1990s, a novel gene was identified in Burkitt’s lymphoma and breast cancer cells that encoded a heptahelical receptor that seemed to be associated with estrogen receptor alpha (ERα). This sequence showed homology to G protein-coupled receptors (GPCRs) and was named GPCR-Br [1,2]. Further, this novel receptor was identified in animal models such as rats, where the homologous gene was detected in cells lacking ERα. These cells responded to 17β-estradiol (E2) stimulation via activating the mitogen-activated protein kinases (MAPK) pathway, and the receptor was named ER-X or GPR41 [3,4]. Nowadays, this receptor is known as G protein-coupled estrogen receptor 1 (GPER/GPR30) and has been identified as an alternative receptor for ERα that triggers cytoplasmic estrogen-signaling and promotes estrogen gene expression [5,6,7,8].
The human GPER gene is intronless, located on chromosome 7, region p22, and encodes a 375 amino acid protein [9]. The GPER promoter region has estrogen response elements (EREs), which allow the binding of estrogen receptors to promote its transcription; consequently, E2 stimulates GPER expression [10]. GPER is a plasma membrane receptor (Figure 1) but can be internalized via a stimulus (e.g., E2) and localized in membranous organelles such as the endoplasmic reticulum and Golgi apparatus [11,12,13,14,15]. GPER has seven transmembrane domains with extracellular N-terminal region. In contrast, the C-terminal region is cytoplasmic and interacts with hydrolases of nucleotide guanosine triphosphate (GTPases) or trimeric G-protein [1,16,17]. GPER can be activated via E2 and inhibitors of ERα, such as tamoxifen (TMX), fulvestrant (ICI 182,780) (FVT), raloxifene (RLX), synthetic ligands (e.g., G1), aldosterone, and natural compounds such as kaempferol [18,19,20,21,22]. These ligands activate GPER to trigger tissue-specific signaling. Some of these ligands are included in the therapeutic scheme against estrogen response cancer, which could explain the re-incidence or aggressiveness of cancer after treating patients.

2. GPER/GPR30 Signaling in Normal Tissues

In humans, there are four different classes of G-protein alpha subunit (Gαs, Gαi/o, Gαq/11, and Gα12/13). It has been reported that GPER can be coupled to a Gα type s (Gαs) or Gα type i/o (Gαi/o); thereby, activating different routes of signaling and regulation [6,23,24,25]. Once GPER is activated, the G-protein trimer is separated into two subunits, Gαs and Gβγ (Figure 2). The Gαs subunit activates adenylate cyclase and cyclic adenosine monophosphate (cAMP) production to mobilize intracellular calcium (Ca2+). Moreover, GPER decreases plasma membrane Ca2+-ATPase (PMCA) activity, maintaining Ca2+ in the cytoplasm and representing a secondary Ca2+ regulation mechanism [11,18,26,27]. This Ca2+ regulation improves brain development, neuronal growth, and communication [28], and also stimulates potassium (K+) efflux, which promotes nitric oxide (NO) production in the endothelium, resulting in a vasodilator effect [25,29].
On the other hand, GPER also seems to be associated with a Gαi/o because the pertussis toxin inhibits the GPER-mediated ERK ½ signaling [25]. Also, the Gβγ subunit could transactivate the epidermal growth factor receptor (EGFR) via heparin-binding EGF-like growth factor (HB-EGF) activating the phosphatidylinositol 3-kinase (PI3K) and MAPK pathways (Figure 2) [30,31]. Furthermore, these mechanisms have been associated with the proliferation of non-tumorigenic breast cells MCF10A ex vivo [32], dendritic spin density related to spatial learning and memory [33,34,35], and glucose and lipid homeostasis [36].
The binding affinity of E2 to GPER is 3–6 nM, whereas ERα’s is 0.1–1 nM. It is noteworthy that there has been no reported activation of GPER via E2 in epithelial and hippocampus models in vivo [37]. However, other ligands, such as G1, activate the receptor [38], showing the importance of using different study models. Nevertheless, as with other GPCRs which can be allosterically regulated, a GPER ligand-independent activity related to a domain involved in protein–protein interactions in the C-terminal, called PDZ motif, has been reported. This activation is associated with proliferation, migration, and invasion [8,25,39]. These characteristics are related to neoplasia development, which suggests that GPER could be a cancer-relevant regulator.
Figure 2. GPER signaling. Different ligands can activate GPER; some are ERα ligands. Once GPER is activated, the G-protein trimer is separated into two subunits, the Gα is related to cAMP and calcium regulation, and the Gβγ can transactivate the EGFR and its signaling. However, a relationship exists between ERα and GPER, favoring the expression of genes related to cancer development [11,30,40]. Created with BioRender.com (accessed on 25 August 2023).
Figure 2. GPER signaling. Different ligands can activate GPER; some are ERα ligands. Once GPER is activated, the G-protein trimer is separated into two subunits, the Gα is related to cAMP and calcium regulation, and the Gβγ can transactivate the EGFR and its signaling. However, a relationship exists between ERα and GPER, favoring the expression of genes related to cancer development [11,30,40]. Created with BioRender.com (accessed on 25 August 2023).
Ijms 24 14993 g002

3. GPER Expression in Cancer

GPER exhibits a tissue-specific activity which can be pro- or anti-tumoral, depending on the cancer type. The receptor level varies depending on its role; when it shows anti-tumoral activity (e.g., gastric, ovarian, and hepatic cancer), its expression is lower than in normal tissues. Research groups have reported that its activation or overexpression in these cancer types is related to a decrease in cancerous properties, supporting the anti-tumoral role of GPER in these cancer types (Table 1) [31,41,42]. An example of this anti-tumoral activity is that GPER activation inhibits the growth of malignant cells through GPER/ERK signaling in hepatocarcinoma and xenograft models [43]. Additionally, the low expression of this receptor in the hepatocarcinoma model has been related to promoter methylation and histone H3 deacetylation [44]. Likewise, GPER anti-carcinogenic effects have been reported in prostate cells and xenografts involving G2 cell-cycle arrest [45]. Moreover, the anti-proliferative activity of GPER has been observed in ovarian cancer [46] and has been related to epigenetic regulation, such as the H3K4me3 mark [41]. Furthermore, the GPER activation via tamoxifen inhibits proliferation, migration, and invasion in pancreatic cancer and regulates the tumorigenic environment, decreasing the anti-inflammatory macrophage phenotype M2 [47].
However, there are some cancer types where GPER plays a pro-tumorigenic role (Table 1). Although intracellular GPER is mainly located on the endoplasmic reticulum, its cytoplasmic location has been related to the advanced cancer stage [27,48]. A higher GPER expression in cancer cells and tissues favors cancer development, and GPER levels serve as indicators for survival prognoses, where elevated levels are related to bad outcomes. Some examples of the above have been observed in lung, prostate, cervical, glioblastoma, and breast cancer models [49,50,51,52,53]. Moreover, GPER expression is related to stemness features in breast cancer stem cells (BCSC) through phosphorylation of BCL2-associated agonist of cell death (BAD) promoting cell survival, but the silencing of GPER results in a decrease in BCSC in vivo [54,55]. However, in breast cancer cells, GPER is related to the stabilization of the actin cytoskeleton and the up-regulation of Yes-associated protein 1 (YAP) and transcriptional coactivator with a PDZ-binding domain (TAZ) via the activation of Gαq-11, phospholipase C beta (PLCβ)/PKC and Rho/Rho-associated protein kinase (ROCK) signaling [56]. In tumorigenic cells, GPER activation has been related to the expression of cell adhesion molecules (CAMs), metalloproteases (MMPs), focal adhesion (FA) pathway genes, cyclin D1 (CCND1), anti-apoptotic protein B cell lymphoma (Bcl-2), and transcription factors such as c-fos [57,58,59]. Some ERα-target genes demonstrate that GPER can promote the expression of ERE genes as an ERα-alternative receptor [60,61]. These proteins are related to carcinogenic characteristics such as proliferation, energetic deregulation, metastasis, and immune and apoptotic evasion [62], which indicate that GPER is a pro-carcinogenic regulator. However, a dual GPER activity in testicular cancer has been reported. The germ neoplasm group is related to high receptor levels, and a GPER E2-activation induces proliferation through the ERK 1/2 pathway in vitro. On the other hand, GPER activation induces apoptosis in the Leydig cell tumors, showing a specific activity of the receptor upon the type of testicular cancer [63].
Table 1. Role of GPER in different cancer types.
Table 1. Role of GPER in different cancer types.
Anti-Tumorigenic Role of GPER
Cancer TypeReceptor StateActivityMechanism
HepaticOverexpressionAnti-tumoral Through GPER in hepatocarcinoma [43]
ProstateLow expressionAnti-tumoralG2 cell-cycle arrest [45]
OvarianOverexpressionAnti-tumoralEpigenetic regulation, such as the H34me3 mark [42]
PancreaticActivation by tamoxifenInhibits the proliferation, migration, and invasion processRegulates the tumorigenic environment, decreasing the anti-inflammatory macrophage phenotype M2 [47]
Leydig cell tumorsLow expressionAnti-tumoralInduces apoptosis [63]
Pro-Tumorigenic Role of GPER Receptor
Cancer TypeReceptor StateActivityMechanism
LungOverexpressionPro-tumoral, poor prognosis The expression of GPER on the cell surface is related to cell survival [49]
CervicalOverexpressionPro-tumoral Induction of tumor-promoting claudin-1 [50]
ProstateStimulation by estrogenIncreased stromal components and prostatic fibrosis accelerate the clinical progression GPER activates EGFR/ERK and P13k/AKT HIF1α pathways [51]
ER-positive breast tumorOverexpressionPoor prognosisERK1/2 pathway [52]
Triple-negative breast cancerGPER interacts with FAKIncreases the migration, adhesion, and invasion Increasing focal adhesion points [52]
GlioblastomaActivation of GPERPromotes tumor growthInduces the epidermal growth factor receptor, triggering the ERK pathway [53]
Testicular germ neoplasmsOverexpressionPromotes proliferationGPER activates the ERK 1/2 pathway [63]
Another reported example of GPER dual activity occurs in colorectal cancer and depends on gender. In a patient study, men with higher GPER levels had a better prognostic outcome; meanwhile, women showed the opposite effect [64]. In contrast, in vitro studies have demonstrated that GPER activation is related to colorectal cancer, favoring proliferation and migration [65].
On the other hand, the GPER-induced gene expression and the receptor can be epigenetically regulated. For example, a negative relationship between miR-338-3p/miR-124 and GPER has been reported. miR-338 inhibits c-fos translation, and miR-124 targets CD151, decreasing proliferation in hepatocarcinoma and breast cancer, respectively. Nevertheless, the E2-activation of GPER diminishes miR-338-3p/miR-124 expression (Table 2) [66,67]. On the other hand, there is a negative relationship between miR-148a and GPER, where receptor activation increases cell proliferation and inhibits apoptosis. However, antagonists of GPER, such as G15, up-regulate miR-148a and promote cell death in ovarian endometriosis [68] and breast cancer [56]. In the latter, it has been reported that down-regulation of miR-148a is related to an increase in CD151, HOX transcript antisense RNA (HOTAIR), and histocompatibility antigen class I, G (HLA-G) levels, favoring the immune system evasion [56]. Furthermore, an epigenetic regulation of GPER expression in endometrial carcinoma has been reported because miR-424 targets GPER, inhibiting E2-induced proliferation [69]. However, GPER also acquires relevance in the carcinogenic process because it seems critical in metastasis development in some cancer types.

4. Epithelial–Mesenchymal Transition and Metastasis Development

Metastasis is the dissemination of tumorigenic cells to other organs, allowing cancer cells to invade them and initiate a new tumor. This process requires phenotype plasticity to dedifferentiate mature cells back to progenitor states through the loss of linkage proteins and gain of mobilization phenotype, a phenomenon called epithelial–mesenchymal transition (EMT) (Figure 3) [62]. GPER has a relevant role in EMT because it promotes metalloprotease expression through the p38/JNK pathway, favoring the degradation of adherent proteins such as E-cadherin and acquiring a migratory phenotype in several types of cancer, such as breast, lung, colon, and ovarian [31].
GPER levels are directly related to lung carcinoma status. Higher levels are associated with EMT through MMP-2/9 expression via insulin-like growth factor-I (IGF-I)/IGF-IR by ERK, p38, and AKT pathways [72]. The activation of GPER via G1 also promotes the expression MMP-2/9 in renal carcinoma in vitro, improving the acquisition of a metastatic phenotype [73]. However, as mentioned, GPER levels can be regulated via micro RNAs (Table 2), and miR-195 targets the mRNA of the receptor, inhibiting the PI3K/AKT-induced MMP-2/9 expression in endometrial carcinoma; therefore, avoiding the EMT [73]. Additionally, E2-activation of GPER is related to the down-regulation of miR-124, allowing the metastasis progression in breast cancer [67]. Nevertheless, although this carcinoma has a low receptor level, GPER activation is related to MMP-9 expression and metastasis development in ovarian cancer [39] (Figure 3).
Moreover, GPER activation is related to FAK activation in triple-negative breast cancer (TNBC) and triggers ERK1/2/AKT signaling, promoting metastatic characteristics, such as migration and invasion [74]. In the same way, the formation of tubes via endothelium cells exposed to a conditioned medium from GPER-activated TNBC is related to the vessel formation required for migration and tumor sustaining [75]. Indeed, GPER antagonists, such as G15, have demonstrated inhibition of EMT in TNBC through sensitization to doxorubicin, up-regulation of E-cadherin, and down-regulation of vimentin [76]. These changes in expression can be associated with a network between Notch1, hypoxia-inducible factor 1α (HIF-1α), and GPER that improves EMT through increasing HIF-1α recruitment to the Snail promoter via nuclear GPER [77]. Snail is a zinc-finger transcriptional factor, and its expression is related to the deacetylation of the E-cadherin gene promoter (CDH1) and, therefore, the down-regulation of E-cadherin expression [78]. On the other hand, it has been reported that GPER activation inhibits migration and invasion [79] and stimulates miR-199a-3p expression and CD151 down-regulation, inhibiting EMT in TNBC [71]. These reports show contrasting data about GPER’s role in breast cancer. Therefore, more studies are required to elucidate the role of GPER in these events.

5. Tumor Microenvironment Favors EMT through GPER

As aforementioned, EMT consists of re-programming expression patterns that stimuli of the environment can favor [62]. The tumor microenvironment consists of normal cells, tumor cells, tumor stromal cells, including stromal fibroblasts, endothelial cells, immune cells such as macrophages and lymphocytes, and non-cellular components (Figure 4). These elements are involved in a dynamic and bidirectional interaction between tumor cells and surroundings through secreted soluble molecules responsible for the horizontal information transfer between cellular/non-cellular communicating cells [80].
The carcinoma-associated fibroblasts (CAFs) are normal fibroblasts activated via growth factors (e.g., transforming growth factor β (TGFβ), hepatocyte growth factor, fibroblast growth factor), and transcription factors (e.g., nuclear factor-kappa B (NF-κB) and heat shock factor-1 (HSF-1)) [81]. CAFs are a significant component of the tumor stroma and produce MMP-3 and cytokines that favor EMT [82]. In addition, these cells express GPER, and its activation up-regulates IL-6, vascular endothelial growth factor (VEGF), and connective tissue growth factor (CTGF) [27]. Also, the CAF secretome improves the expression of vimentin, ZEB1, and Snail in cancer cells, which are mesenchymal proteins essential for cancer progression [22]. Consequently, the activation of GPER in CAFs stimulates the secretion of proteins that activate the cancer cells, promoting a re-programming of an epithelial expression pattern to a mesenchymal [83,84]. Furthermore, CAFs promote TMX and FLV resistance in breast cancer and EMT in hepatocarcinoma via IL-6/IL-6R/STAT3 pathway [85,86,87]. However, it has been reported that 3-methylcholanthrene activates the EGFR/ERK/c-Fos signaling in CAFs through an aryl hydrocarbon receptor (AhR)-GPER mechanism favoring the tumor growth via the up-regulation of cyclin D1 and CYP1B1 [56]. Also, GPER has a role in this resistance because the receptor levels are higher in drug-resistant cancer cells. However, when cells are exposed to receptor antagonists, cell sensibility is recovered [14]. This resistance is related to an aggressive phenotype and mesenchymal protein expression, which suggests that GPER promotes drug resistance and cancer progression via EMT development.
Figure 4. The tumor microenvironment favors EMT through GPER. GPER overexpression in carcinoma-associated fibroblasts (CAFs) increases the production of MMP3, IL-6, and VEGF. These molecules improve tumor viability via inducing blood vessel formation. Additionally, tumor-associated macrophages (TAMs) increase IL-10 production through GPER activation, generating a protective microenvironment around the tumor mass. The expression of GPER on endothelium has been shown to influence the improvement of tumor angiogenesis. CAFs: carcinoma-associated fibroblasts; TAMs: tumor-associated macrophages; M2 activated: activated macrophages of type M2; ROS: reactive oxygen species; HIF-1α: hypoxia-inducible factor 1α; VEGF: vascular endothelial growth factor. Red arrows indicate un increase in the GPER levels. Modified from references [75,77,82,88]. Created with BioRender.com (accessed on 25 August 2023).
Figure 4. The tumor microenvironment favors EMT through GPER. GPER overexpression in carcinoma-associated fibroblasts (CAFs) increases the production of MMP3, IL-6, and VEGF. These molecules improve tumor viability via inducing blood vessel formation. Additionally, tumor-associated macrophages (TAMs) increase IL-10 production through GPER activation, generating a protective microenvironment around the tumor mass. The expression of GPER on endothelium has been shown to influence the improvement of tumor angiogenesis. CAFs: carcinoma-associated fibroblasts; TAMs: tumor-associated macrophages; M2 activated: activated macrophages of type M2; ROS: reactive oxygen species; HIF-1α: hypoxia-inducible factor 1α; VEGF: vascular endothelial growth factor. Red arrows indicate un increase in the GPER levels. Modified from references [75,77,82,88]. Created with BioRender.com (accessed on 25 August 2023).
Ijms 24 14993 g004
Although immune cells must locate and eradicate tumorigenic cells, they can also contribute to the development of tumors. A chronic inflammatory state is a risk factor for cancer development because prolonged exposure to pro-inflammatory cytokines (e.g., TNF-α) and ROS promotes DNA and tissue damage [89]. This inflammatory state favors the development of cancer cells, and the anti-inflammatory cells (M2) surrounding the neoplasm camouflage the tumor from the immune system, preventing the elimination of affected cells and allowing EMT development [82,90]. Several research groups have reported that GPER is expressed in monocytic cells, such as CD14+ monocytes, in vitro differentiated macrophages, and tissue-resident macrophages [40]. Also, GPER activation is related to expanding regulatory T-cell populations and secretion of the anti-inflammatory cytokine IL-10 from M2-polarized cells [20]. Moreover, it has been reported that exosomes of these macrophages could activate the PI3K-AKT pathway in gastric cancer cells and induce EMT. At the same time, tumorigenic cells stimulate M2 polarization, creating a positive feed-loop [91]. In addition, GPER activation via diethylstilbestrol [92], bisphenols A (BPA), bisphenol AF (BPAF) [93], 17α-ethynylestradiol (EE) [94], and zearalenone (ZLN) [95] have been related to monocyte activation through an increase in the receptor for activated C kinase 1 (RACK1) levels in the cell line THP-1. Also, EE, E2, and ZLN favor the production of LPS-induced pro-inflammatory cytokines, such as TNF-α and IL-8, and the up-regulation of CD86 [95]. However, VIN, atrazine (ATZ), and cypermethrin (CYP) decrease these proteins [96]. On the other hand, the EE and VIN inhibit RACK1 in a primary culture of female donors [97], highlighting the role of GPER in innate immune response regulation.

6. Role of GPER in Angiogenesis

Angiogenesis involves creating new vessels that dispense nutrients and is a mechanism through which malignant cells can disseminate to other tissues. Endothelial cells are critical for tumor progression because vessel formation contributes to cancer dissemination and maintenance. Estrogens are related to vascular protection through receptors ERα, ERβ, and GPER [98]. They are associated with protection in cardiovascular diseases, mainly in pre-menopausal females [88], diminishing the inflammation and the NO production mediated in endothelial cells [99]. The ERβ shows increased expression after vasculature injury in males and females and has been related to a protective effect [100]. In addition, the estrogens are tightly linked to the angiogenesis process, stimulating the ERs [101]. However, information about GPER’s protective activity in the endothelium is scarce. GPER mediates the activation of transduction pathways in different processes related to tumorigenesis, such as angiogenesis favoring tumor growth [102] or stimulating endothelial cell proliferation [103]. Some studies have shown that the activation of GPER via G1 inhibits the proliferation of endothelial cells, inhibiting DNA synthesis and arresting the cells in the S and G2 phases [98]. Also, E2 and G1 activate the GPER/EGFR/ERK/c-fos signaling pathway and increase VEGF via up-regulation of HIF1α [104], which is directly related to vessel formation. Also, E2 regulates endothelial cell tube formation in injury and has protective effects associated with GPER in endothelial cells [101]. For this reason, GPER has been proposed as a possible therapeutic target to control the growth of some tumors, such as breast cancer.

7. Therapeutic Alternatives against Cancer Targeting GPER

Given the pro-tumorigenic role of GPER, it has been proposed as a therapeutic target. There are a variety of GPER antagonists, such as G15, G36, CIMBA, MIBE, PBX1, PBX2, C4PY, and CPT. The abovementioned drugs have been tested in breast cancer and CAF in vitro and in vivo, resulting in a GPER-dependent G1/E2-signaling inhibition [105,106]. Additionally, chimeras made from an E3 ubiquitin ligase ligand called PROTACs degrade the target; in particular, the E2-PROTAC acts against GPER [107]. However, GPER expression is simultaneous with ERα in some estrogen-responsiveness cancers; as a result, anti-estrogenic compounds must be reconsidered because drugs such as TMX and FVS act as a ligand for GPER [20]. As mentioned, CAFs improve EMT; thus, novel therapies have been focused on them, but their use is related to severe secondary effects such as muscle loss and even death [108]. On the other hand, triptolide is a drug reported as impairing the induction of EMT via CAFs in cancer cells. Also, there are novel therapies against signaling pathways related to the development of cancer properties, such as AG490, a drug against the JAK/STAT signaling involved in EMT that can be triggered via GPER activation [109]. These findings suggest that GPER could be a promising therapeutic target that can act at different levels, regulating the cancer cell and its surrounding components.

8. Discussion

The role of GPER in cells is beginning to be understood. GPER has been associated with several physiological functions, such as angiogenesis, brain development, and homeostasis [28,101], but it also participates in several pathological processes, such as carcinogenesis and metastasis development [74,75,76]. Although GPER improves the correct development of neuronal, connective, and epithelial tissues [35,36], changes in its expression can deregulate homeostasis and trigger malignant development [31]. GPER expression levels depend on the cancer type, which could serve as a prognosis marker for the tissue affected [49,50,51]. Because the receptor has a pro-tumorigenic role and its expression is related to cancer status, GPER could become a therapeutic target and complete a drug network to enhance a treatment scheme. Likewise, identifying some miRNAs that can inhibit the receptor and downstream proteins represents a novel therapeutic alternative for the treatment of cancer. When the receptor exhibits an anti-tumoral activity, its ligands, downstream proteins, and associated miRNAs should be well studied to determine their potential as therapeutic alternatives.
Metastasis is a challenge for eradicating cancer because the dissemination of carcinogenic cells affects different tissues and could promote an organic failure; moreover, the number of malignant cells increases, and they acquire diverse phenotypes based on the environment in which they are established [110]. The critical step for metastasis development is the EMT, where the cell attachment decreases and the cells become migratory [62]. GPER plays a relevant role in EMT development because it is expressed in malignant and environmental cells [27,40]. Receptor activation triggers changes in the immunological cells that affect the cancer cells, stimulating the expression of mesenchymal proteins and creating a tumorigenic environment. In contrast, the activation of GPER in the malignant cells allows structural and expression changes needed to invade other tissues [85,86,87]. A remarkable point is that GPER promotes monocyte activation and LPS-induced pro-inflammatory cytokines [56], providing a potential strategy to enhance the immune response. Additionally, GPER has multiple ligands, including some of the drugs used for cancer treatment and xenobiotics; thus, representing an advantage for cancer development and a challenge for medical procedures [20,21,92,93,94,95,96,97].
Nevertheless, all cancer types have a metastasis pattern, which, if it is not compulsory, is a guide of behavior, and this pattern is based on closeness, vessels, and the function of the tissue. For example, breast cancer usually disseminates to the lung, while prostate cancer frequently invades nodes [111,112]. With regards to GPER tissue-specific activity, it is interesting that cancer types with determinate GPER activity develop metastases to organs where the receptor has the same effect, suggesting that if a cancer type is GPER pro-tumorigenic, its dispersion will affect an organ where the new tumor possibly has the same GPER activity, and a focused therapy against this receptor would control primary and metastatic tumors. Furthermore, identifying this receptor in cancer types without therapeutic targets, such as TNBC [76,77], represents an opportunity to develop new drugs against cancers with poor or incomplete treatment. Moreover, whilst there has been considerable research into GPER, there is still contrasting information. This represents an opportunity for future research to elucidate the tissue-specific and ligand-dependent activity in in vitro, ex vivo, and in vivo models, and come to an understanding of the differences reported through the years. Moreover, it will be interesting to evaluate the ligand-independent signaling of the receptor in cancer where GPER is pro-tumorigenic, and, assess if the use of receptor antagonists is enough to inhibit the carcinogenic activity or if the allosteric activation compensates for the lacking functions (Figure 5).

9. Conclusions

GPER has several activities in metastatic development, which justifies proposing this receptor as a therapeutic target to avoid EMT in various cancer types. Also, it is necessary to rethink the current schemes for cancer treatment where GPER ligands are included.

10. Future Directions

GPER is a novel receptor that must be well studied both in in vitro and in vivo models to determine its role in normal and pathologic processes. It should also be investigated with regard to whether or not it could be an efficient therapeutic target against cancer that would help current therapy schemes. There have been contradictory reports regarding GPER tissue-specific signaling which must be clarified, and the reasons for these discrepancies must be determined. Moreover, the possible activation via other estrogens, drugs, and estrogen-like ligands must be analyzed to determine the signaling cascades activated (Figure 5).

Author Contributions

Conceptualization, A.C.T.-G., E.A.F.-R., A.O.-Z. and J.E.L.-M.; funding acquisition, A.O.-Z. and J.E.L.-M.; project administration, J.E.L.-M.; writing—original draft, A.C.T.-G., E.A.F.-R. and J.E.L.-M.; writing—review and editing, A.C.T.-G., E.A.F.-R., A.O.-Z. and J.E.L.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by Instituto de Ciencia, Tecnología e Innovación del Estado de Michoacán (ICTI, grant number PICIR 022-2022), and CIC-UMSNH (14.5).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Owman, C.; Blay, P.; Nilsson, C.; Lolait, S.J. Cloning of Human CDNA Encoding a Novel Heptahelix Receptor Expressed in Burkitt s Lymphoma and Widely Distributed in Brain and Peripheral Tissues Communication Both in CNS and in Peripheral Tissues. Biochem. Biophys. Res. Commun. 1996, 228, 285–292. [Google Scholar] [CrossRef]
  2. Carmeci, C.; Thompson, D.A.; Ring, H.Z.; Francke, U.; Weigel, R.J. Identification of a Gene (GPR30) With Homology to the G-protein-coupled Receptor Superfamily Associated with Estrogen Receptor Expression in Breast Cancer. Genomics 1997, 45, 607–617. [Google Scholar] [CrossRef]
  3. Kimura, M.; Mizukami, Y.; Miura, T.; Fujimoto, K.; Kobayashi, S.; Matsuzaki, M. Orphan G Protein-Coupled Receptor, GPR41, Induces Apoptosis via a P53/Bax Pathway during Ischemic Hypoxia and Reoxygenation. J. Biol. Chem. 2001, 276, 26453–26460. [Google Scholar] [CrossRef]
  4. Toran-Allerand, C.D.; Guan, X.; MacLusky, N.J.; Horvath, T.L.; Diano, S.; Singh, M.; Connolly, E.S.; Nethrapalli, I.S.; Tinnikov, A.A. ER-X: A Novel, Plasma Membrane-Associated, Putative Estrogen Receptor That Is Regulated during Development and after Ischemic Brain Injury. J. Neurosci. 2002, 22, 8391–8401. [Google Scholar] [CrossRef]
  5. Filardo, E.J.; Quinn, J.; Bland, K.I.; Frackelton, A.R.J. Estrogen-Induced Activation of Erk-1 and Erk-2 Requieres the G Protein-Couple Receptor Homolog, GPER, and Occurs via Trans-Activation of the Epidermal Growth Factor Receptor through Release of HB-EGF. Mol. Endocrinol. 2000, 14, 1649–1660. [Google Scholar] [CrossRef]
  6. Wyckoff, M.H.; Chambliss, K.L.; Mineo, C.; Yuhanna, I.S.; Mendelsohn, M.E.; Mumby, S.M.; Shaul, P.W. Plasma Membrane Estrogen Receptors Are Coupled to Endothelial Nitric-Oxide Synthase through Gαi. J. Biol. Chem. 2001, 276, 27071–27076. [Google Scholar] [CrossRef]
  7. Kanda, N.; Watanabe, S. 17β-Estradiol Stimulates the Growth of Human Keratinocytes by Inducing Cyclin D2 Expression. J. Investig. Dermatol. 2004, 123, 319–328. [Google Scholar] [CrossRef]
  8. Tutzauer, J.; Eilard, I.A.; De Valdivia, E.G.; Sw, K.; Broselid, S.; Kahn, R. Ligand-Independent G Protein—Coupled Estrogen Receptor/G Protein—Coupled Receptor 30 Activity: Lack of Receptor- Dependent Effects of G-1 and 17 b -Estradiol S. Mol. Pharmacol. 2021, 100, 271–282. [Google Scholar] [CrossRef]
  9. O’Dowd, B.F.; Nguyen, T.; Marchese, A.; Cheng, R.; Lynch, K.R.; Heng, H.H.; Kolakowski, L.F., Jr.; George, S.R. Discovery of Three Novel G-protein-coupled Receptor Genes. Genomics 1998, 47, 310–313. [Google Scholar] [CrossRef]
  10. Peña-Gutiérrez, K.M.; Hernández-Ortega, K.; Bello-Alvarez, C.; Camacho-Arroyo, I. Expression and Estrogen Regulation of G Protein-coupled Estrogen Receptor in Human Glioblastoma Cells. Oncol. Lett. 2022, 24, 397. [Google Scholar] [CrossRef]
  11. Thomas, P.; Pang, Y.; Filardo, E.J.; Dong, J. Identity of an Estrogen Membrane Receptor Coupled to a G Protein in Human Breast Cancer Cells. Endocrinology 2005, 146, 624–632. [Google Scholar] [CrossRef]
  12. Filardo, E.; Quinn, J.; Pang, Y.; Graeber, C.; Shaw, S.; Dong, J.; Thomas, P. Activation of the Novel Estrogen Receptor G Protein-Coupled Receptor 30 (GPER) at the Plasma Membrane. Endocrinology 2007, 148, 3236–3245. [Google Scholar] [CrossRef]
  13. Otto, C.; Rohde-Schulz, B.; Schwarz, G.; Fuchs, I.; Klewer, M.; Brittain, D.; Langer, G.; Bader, B.; Prelle, K.; Nubbemeyer, R.G. Protein-Coupled Receptor 30 Localizes to the Endoplasmic Reticulum and Is Not Activated by Estradiol. Endocrinology 2008, 149, 4846–4856. [Google Scholar] [CrossRef] [PubMed]
  14. Mo, Z.; Liu, M.; Yang, F.; Luo, H.; Li, Z.; Tu, G.; Yang, G. GPR30 as an Initiator of Tamoxifen Resistance in Hormone-Dependent Breast Cancer. Breast Cancer Res. 2013, 15, R114. [Google Scholar] [CrossRef] [PubMed]
  15. Funakoshi, T.; Yanai, A.; Shinoda, K.; Kawano, M.M.; Mizukami, Y. G Protein-Coupled Receptor 30 Is an Estrogen Receptor in the Plasma Membrane. Biochem. Biophys. Res. Commun. 2006, 346, 904–910. [Google Scholar] [CrossRef] [PubMed]
  16. Unal, H.; Karnik, S. Domain Coupling in GPCRs: The Engine for Induced Conformational Changes. Trends Pharmacol. Sci. 2012, 33, 79–88. [Google Scholar] [CrossRef] [PubMed]
  17. Christofides, K.; Menon, R.; Jones, C.E. Endocytosis of G Protein-Coupled Receptors and Their Ligands: Is There a Role in Metal Trafficking? Cell Biochem. Biophys. 2018, 76, 329–337. [Google Scholar] [CrossRef]
  18. Revankar, C.M.; Cimino, D.F.; Sklar, L.A.; Arterburn, J.B.; Prossnitz, E.R. A Transmembrane Intracellular Estrogen Receptor Mediates Rapid Cell Signaling. Science 2005, 307, 1625–1630. [Google Scholar] [CrossRef]
  19. Méndez-Luna, D.; Martínez-Archundia, M.; Maroun, R.C.; Ceballos-Reyes, G.; Fragoso-Vázquez, M.J.; González-Juárez, D.E.; Correa-Basurto, J. Deciphering the GPER/GPER-Agonist and Antagonists Interactions Using Molecular Modeling Studies, Molecular Dynamics, and Docking Simulations. J. Biomol. Struct. Dyn. 2015, 33, 2161–2172. [Google Scholar] [CrossRef]
  20. Pepermans, R.A.; Sharma, G.; Prossnitz, E.R. G Protein-Coupled Estrogen Receptor in Cancer and Stromal Cells: Functions and Novel Therapeutic Perspectives. Cells 2021, 10, 672. [Google Scholar] [CrossRef]
  21. Ding, Q.; Chorazyczewski, J.; Gros, R.; Motulsky, H.J.; Limbird, L.E.; Feldman, R.D. Correlation of Functional and Radioligand Binding Characteristics of GPER Ligands Confirming Aldosterone as a GPER Agonist. Pharmacol. Res. Perspect. 2022, 10, e00995. [Google Scholar] [CrossRef]
  22. Feng, B.; Wu, J.; Shen, B.; Jiang, F.; Feng, J. Cancer-Associated Fibroblasts and Resistance to Anticancer Therapies: Status, Mechanisms, and Countermeasures. Cancer Cell Int. 2022, 22, 166. [Google Scholar] [CrossRef]
  23. Lucas, T.F.G.; Royer, C.; Siu, E.R.; Lazari, M.F.M.; Porto, C.S. Expression and Signaling of G Protein-Coupled Estrogen Receptor 1 (GPER) in Rat Sertoli Cells. Biol. Reprod. 2010, 83, 307–317. [Google Scholar] [CrossRef]
  24. Gaudet, H.M.; Cheng, S.B.; Christensen, E.M.; Filardo, E.J. The G-Protein Coupled Estrogen Receptor, GPER: The inside and inside-out Story. Mol. Cell. Endocrinol. 2015, 418, 207–219. [Google Scholar] [CrossRef]
  25. Tran, Q.K. Reciprocality Between Estrogen Biology and Calcium Signaling in the Cardiovascular System. Front. Endocrinol. 2020, 11, 568203. [Google Scholar] [CrossRef]
  26. Tran, Q.K.; VerMeer, M.; Burgard, M.A.; Hassan, A.B.; Giles, J. Hetero-Oligomeric Complex between the G Protein-Coupled Estrogen Receptor 1 and the Plasma Membrane Ca2+-ATPase 4b. J. Biol. Chem. 2015, 290, 13293–13307. [Google Scholar] [CrossRef]
  27. Xu, S.; Yu, S.; Dong, D.; Lee, L.T.O. G Protein-Coupled Estrogen Receptor: A Potential Therapeutic Target in Cancer. Front. Endocrinol. 2019, 10, 725. [Google Scholar] [CrossRef]
  28. Pemberton, K.; Xu, F.; Rosato, M.; Dedert, C.; Deleon, C.; Arnatt, C. Differential Effects of the G-Protein-Coupled Estrogen Receptor (GPER) on Rat Embryonic (E18) Hippocampal and Cortical Neurons. eNeuro 2022, 9, 1–23. [Google Scholar] [CrossRef]
  29. Fredette, N.C.; Meyer, M.R.; Prossnitz, E.R. Role of GPER in Estrogen-Dependent Nitric Oxide Formation and Vasodilation. J. Steroid Biochem. Mol. Biol. 2018, 176, 65–72. [Google Scholar] [CrossRef]
  30. Prossnitz, E.R.; Maggiolini, M. Mechanisms of Estrogen Signaling and Gene Expression Via GPR30. Mol. Cell. Endocrinol. 2009, 308, 32–38. [Google Scholar] [CrossRef]
  31. Deng, Y.; Miki, Y.; Nakanishi, A. Estradiol/GPER Affects the Integrity of Mammary Duct-like Structures in Vitro. Sci. Rep. 2020, 10, 1386. [Google Scholar] [CrossRef]
  32. Scaling, A.L.; Prossnitz, E.R.; Hathaway, H.J. GPER Mediates Estrogen-Induced Signaling and Proliferation in Human Breast Epithelial Cells and Normal and Malignant Breast. Horm. Cancer 2014, 5, 146–160. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, Y.Y.; Liu, M.Y.; Liu, Z.; Zhao, J.K.; Zhao, Y.G.; He, L.; Li, W.; Zhang, J.Q. GPER-Mediated Estrogenic Regulation of Actin Polymerization and Spatial Memory Involves SRC-1 and PI3K-MTORC2 in the Hippocampus of Female Mice. CNS Neurosci. Ther. 2019, 25, 714–733. [Google Scholar] [CrossRef] [PubMed]
  34. Roque, C.; Mendes-Oliveira, J.; Duarte-Chendo, C.; Baltazar, G. The Role of G Protein-Coupled Estrogen Receptor 1 on Neurological Disorders. Front. Neuroendocrinol. 2019, 55, 100786. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, C.; Liu, Q.; Yu, C.Y.; Wang, F.; Shao, Y.; Sun, K.S.; Sun, T.; Liu, J. G Protein-Coupled Estrogen Receptor 1 Knockout Deteriorates MK-801-Induced Learning and Memory Impairment in Mice. Front. Behav. Neurosci. 2020, 14, 157. [Google Scholar] [CrossRef] [PubMed]
  36. Sharma, G.; Prossnitz, E.R. Targeting the G Protein-Coupled Estrogen Receptor (GPER) in Obesity and Diabetes. Endocr. Metab. Sci. 2021, 2, 100080. [Google Scholar] [CrossRef]
  37. Luo, J.; Liu, D. Does GPER Really Function as a G Protein-Coupled Estrogen Receptor in Vivo? Front. Endocrinol. 2020, 11, 148. [Google Scholar] [CrossRef]
  38. Vo, D.K.H.; Hartig, R.; Weinert, S.; Haybaeck, J.; Nass, N. G-Protein-Coupled Estrogen Receptor (GPER)-Specific Agonist G1 Induces ER Stress Leading to Cell Death in MCF-7 Cells. Biomolecules 2019, 9, 503. [Google Scholar] [CrossRef]
  39. Jung, J. Role of G Protein-Coupled Estrogen Receptor in Cancer Progression. Toxicol. Res. 2019, 35, 209–214. [Google Scholar] [CrossRef]
  40. Notas, G.; Kampa, M.; Castanas, E. G Protein-Coupled Estrogen Receptor in Immune Cells and Its Role in Immune-Related Diseases. Front. Endocrinol. 2020, 11, 579420. [Google Scholar] [CrossRef]
  41. Tian, S.; Zhan, N.; Li, R.; Dong, W. Downregulation of G Protein-Coupled Estrogen Receptor (GPER) Is Associated with Reduced Prognosis in Patients with Gastric Cancer. Med. Sci. Monit. 2019, 25, 3115–3126. [Google Scholar] [CrossRef] [PubMed]
  42. Han, N.; Heublein, S.; Jeschke, U.; Kuhn, C.; Hester, A.; Czogalla, B.; Mahner, S.; Rottmann, M.; Mayr, D.; Schmoeckel, E. The G-Protein-Couple Estrogen Receptor (GPER) Regulates Trimethylation of Histone H3 at Lysine 4 and Represses Migration and Proliferation of Ovarian Cancer Cells in Vitro. Cells 2021, 10, 619. [Google Scholar] [CrossRef]
  43. Qiu, Y.A.; Xiong, J.; Fu, Q.; Dong, Y.; Liu, M.; Peng, M.; Jin, W.; Zhou, L.; Xu, X.; Huang, X. GPER-Induced ERK Signaling Decreases Cell Viability of Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 638171. [Google Scholar] [CrossRef]
  44. Liu, Q.; Chen, Z.; Jiang, G.; Zhou, Y.; Yang, X.; Huang, H.; Liu, H.; Du, J.; Wang, H. Epigenetic down Regulation of G Protein-Coupled Estrogen Receptor (GPER) Functions as a Tumor Suppressor in Colorectal Cancer. Mol. Cancer 2017, 16, 87. [Google Scholar] [CrossRef] [PubMed]
  45. Chan, Q.K.Y.; Lam, H.; Ng, C.; Lee, A.Y.Y.; Chan, E.S.Y.; Ng, K.; Ho, S.; Lau, K. Activation of GPER Inhibits Growth of Prostate Cancer Cells via Sustained Activation of Erk1/2, c-Jun/c-Fos-Dependent Upregulation of P21, and Induction of G2 Cell-Cycle Arrest. Cell Death Differ. 2010, 17, 1511–1523. [Google Scholar] [CrossRef]
  46. Schüler-Toprak, S.; Skrzypczak, M.; Ignatov, T.; Ignatov, A.; Ortmann, O.; Treeck, O.G. Protein-Coupled Estrogen Receptor 1 (GPER-1) and Agonist G-1 Inhibit Growth of Ovarian Cancer Cells by Activation of Anti-Tumoral Transcriptome Responses: Impact of GPER-1 MRNA on Survival. J. Cancer Res. Clin. Oncol. 2020, 146, 3175–3188. [Google Scholar] [CrossRef] [PubMed]
  47. Cortes, E.; Sarper, M.; Robinson, B.; Lachowski, D.; Chronopoulos, A.; Thorpe, S.D.; Lee, D.A.; Río-Hernández, A.E. GPER Is a Mechanoregulator of Pancreatic Stellate Cells and the Tumor Microenvironment. EMBO Rep. 2019, 20, e46556. [Google Scholar] [CrossRef] [PubMed]
  48. Zhu, C.X.; Xiong, W.; Wang, M.L.; Yang, J.; Shi, H.J.; Chen, H.Q.; Niu, G. Nuclear G Protein-Coupled Oestrogen Receptor (GPER) Predicts Poor Survival in Patients with Ovarian Cancer. J. Int. Med. Res. 2018, 46, 723–731. [Google Scholar] [CrossRef]
  49. Jala, V.R.; Radde, B.N.; Haribabu, B.; Klinge, C.M. Enhanced Expression of G-Protein Coupled Estrogen Receptor (GPER/GPER) in Lung Cancer. BMC Cancer 2012, 12, 624. [Google Scholar] [CrossRef]
  50. Akimoto, T.; Takasawa, A.; Takasawa, K.; Aoyama, T.; Murata, M.; Osanai, M.; Saito, T.; Sawada, N. Estrogen/GPER Signaling Contributes to the Malignant Potentials of ER-Negative Cervical Adenocarcinoma via Regulation of Claudin-1 Expression. Neoplasia 2018, 20, 1083–1093. [Google Scholar] [CrossRef]
  51. Yang, Y.; Sheng, J.; Hu, S.; Cui, Y.; Xiao, J.; Yu, W.; Peng, J.; Han, W.; He, Q.; Fan, Y. Estrogen and G Protein-Coupled Estrogen Receptor Accelerate the Progression of Benign Prostatic Hyperplasia by Inducing Prostatic Fibrosis. Cell Death Dis. 2022, 13, 533. [Google Scholar] [CrossRef] [PubMed]
  52. Xu, T.; Ma, D.; Chen, S.; Tang, R.; Yang, J.; Meng, C.; Feng, Y.; Liu, L.; Wang, J.; Luo, H. High GPER Expression in Triple-Negative Breast Cancer Is Linked to pro-Metastatic Pathways and Predicts Poor Patient Outcomes. NPJ Breast Cancer 2022, 8, 100. [Google Scholar] [CrossRef]
  53. Gutierrez-Almeida, C.E.; Santerre, A.; Leon-Moreno, L.C.; Aguilar-Garcia, I.G.; Castaneda-Arellano, R.; Duenas-Jimenez, S.H.; Duenas-Jimenez, J.M. Proliferation and Apoptosis Regulation by G Protein-Coupled Estrogen Receptor in Glioblastoma C6 Cells. Oncol. Lett. 2022, 24, 217. [Google Scholar] [CrossRef] [PubMed]
  54. Bui, N.L.C.; Pandey, V.; Zhu, T.; Ma, L.; Basappa; Lobie, P.E. Bad Phosphorylation as a Target of Inhibition in Oncology. Cancer Lett. 2018, 415, 177–186. [Google Scholar] [CrossRef] [PubMed]
  55. Chan, Y.T.; Lai, A.C.Y.; Lin, R.J.; Wang, Y.H.; Wang, Y.T.; Chang, W.W.; Wu, H.Y.; Lin, Y.J.; Chang, W.Y.; Wu, J.C. GPER-Induced Signaling Is Essential for the Survival of Breast Cancer Stem Cells. Int. J. Cancer 2020, 147, 1674–1685. [Google Scholar] [CrossRef]
  56. Masi, M.; Racchi, M.; Travelli, C.; Corsini, E.; Buoso, E. Molecular Characterization of Membrane Steroid Receptors in Hormone-Sensitive Cancers. Cells. 2021, 10, 2999. [Google Scholar] [CrossRef]
  57. Zhou, X.; Wang, S.; Wang, Z.; Feng, X.; Liu, P.; Lv, X.B.; Li, F.; Yu, F.X.; Sun, Y.; Yuan, H. Estrogen Regulates Hippo Signaling via GPER in Breast Cancer. J. Clin. Investig. 2015, 125, 2123–2135. [Google Scholar] [CrossRef]
  58. Chuang, S.C.; Chen, C.H.; Chou, Y.S.; Ho, M.L.; Chang, J.K. G Protein-Coupled Estrogen Receptor Mediates Cell Proliferation through the Camp/Pka/Creb Pathway in Murine Bone Marrow Mesenchymal Stem Cells. Int. J. Mol. Sci. 2020, 21, 6490. [Google Scholar] [CrossRef]
  59. Talia, M.; De Francesco, E.M.; Rigiracciolo, D.C.; Muoio, M.G.; Muglia, L.; Belfiore, A.; Maggiolini, M.; Sims, A.H.; Lappano, R. The G Protein-Coupled Estrogen Receptor (GPER) Expression Correlates with Pro-Metastatic Pathways in ER-Negative Breast Cancer: A Bioinformatics Analysis. Cells 2020, 9, 622. [Google Scholar] [CrossRef]
  60. Weisz, A.; Rosales, R. Identification of an Estrogen Response Element Upstream of the Human C-Fos Gene That Binds the Estrogen Receptor and the AP-1 Transcription Factor. Nucleic Acids Res. 1990, 18, 5097–5106. [Google Scholar] [CrossRef]
  61. Welboren, W.J.; Van Driel, M.A.; Janssen-Megens, E.M.; Van Heeringen, S.J.; Sweep, F.C.; Span, P.N.; Stunnenberg, H.G. ChIP-Seq of ERα and RNA Polymerase II Defines Genes Differentially Responding to Ligands. EMBO J. 2009, 28, 1418–1428. [Google Scholar] [CrossRef]
  62. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  63. Chimento, A.; De Luca, A.; Nocito, M.C.; Avena, P.; La Padula, D.; Zavaglia, L.; Pezzi, V. Role of GPER-Mediated Signaling in Testicular Functions and Tumorigenesis. Cells 2020, 9, 2115. [Google Scholar] [CrossRef] [PubMed]
  64. Jacenik, D.; Krajewska, W.M. Significance of G Protein-Coupled Estrogen Receptor in the Pathophysiology of Irritable Bowel Syndrome, Inflammatory Bowel Diseases and Colorectal Cancer. Front. Endocrinol. 2020, 11, 390. [Google Scholar] [CrossRef]
  65. Jacenik, D.; Beswick, E.J.; Krajewska, W.M.; Prossnitz, E.R. G protein-coupled estrogen receptor in colon function, immune regulation and carcinogenesis. World J. Gastroenterol. 2019, 25, 4092–4104. [Google Scholar] [CrossRef] [PubMed]
  66. Vivacqua, A.; Sebastiani, A.; Miglietta, A.M.; Rigiracciolo, D.C.; Cirillo, F.; Galli, G.R.; Talia, M.; Santolla, M.F.; Lappano, R.; Giordano, F. MiR-338-3p Is Regulated by Estrogens through GPER in Breast Cancer Cells and Cancer-Associated Fibroblasts (CAFs). Cells 2018, 7, 203. [Google Scholar] [CrossRef]
  67. Yang, H.; Wang, C.; Liao, H.; Wang, Q. Activation of GPER by E2 Promotes Proliferation, Invasion and Migration of Breast Cancer Cells by Regulating the MiR-124/CD151 Pathway. Oncol. Lett. 2021, 21, 432. [Google Scholar] [CrossRef]
  68. He, S.Z.; Li, J.; Bao, H.C.; Wang, M.M.; Wang, X.R.; Huang, X.; Li, F.H.; Zhang, W.; Xu, A.L.; Fang, H.C. G Protein-Coupled Estrogen Receptor/MiR-148a/Human Leukocyte Antigen-G Signaling Pathway Mediates Cell Apoptosis of Ovarian Endometriosis. Mol. Med. Rep. 2018, 18, 1141–1148. [Google Scholar] [CrossRef]
  69. Zhang, H.; Wang, X.; Chen, Z.; Wang, W. MicroRNA-424 Suppresses Estradiol-Induced Cell Proliferation via Targeting GPER in Endometrial Cancer Cells. Cell. Mol. Biol. 2015, 61, 96–101. [Google Scholar] [CrossRef]
  70. Deng, J.; Wang, W.; Yu, G.; Ma, X. MicroRNA-195 Inhibits Epithelial-mesenchymal Transition by Targeting G Protein-coupled Estrogen Receptor 1 in Endometrial Carcinoma. Mol. Med. Rep. 2019, 20, 4023–4032. [Google Scholar] [CrossRef]
  71. Huang, R.; Li, J.; Pan, F.; Zhang, B.; Yao, Y. The Activation of GPER Inhibits Cells Proliferation, Invasion and EMT of Triple-Negative Breast Cancer via CD151/MiR-199a-3p Bio-Axis. Am. J. Transl. Res. 2020, 12, 32–44. [Google Scholar] [PubMed]
  72. Maitra, R.; Malik, P.; Mukherjee, T.K. Targeting Estrogens and Various Estrogen-Related Receptors against Non-Small Cell Lung Cancers: A Perspective. Cancers 2022, 14, 80. [Google Scholar] [CrossRef]
  73. Guan, B.Z.; Yan, R.L.; Huang, J.W.; Li, F.L.; Zhong, Y.X.; Chen, Y.; Liu, F.N.; Hu, B.; Huang, S.B.; Yin, L.H. Activation of G Protein Coupled Estrogen Receptor (GPER) Promotes the Migration of Renal Cell Carcinoma via the PI3K/AKT/MMP-9 Signals. Cell Adhes. Migr. 2018, 12, 109–117. [Google Scholar] [CrossRef] [PubMed]
  74. Rigiracciolo, D.C.; Santolla, M.F.; Lappano, R.; Vivacqua, A.; Cirillo, F.; Galli, G.R.; Talia, M.; Muglia, L.; Pellegrino, M.; Nohata, N. Focal Adhesion Kinase (FAK) Activation by Estrogens Involves GPER in Triple-Negative Breast Cancer Cells. J. Exp. Clin. Cancer Res. 2019, 38, 58. [Google Scholar] [CrossRef] [PubMed]
  75. Avena, P.; Casaburi, I.; Zavaglia, L.; Nocito, M.C.; La Padula, D.; Rago, V.; Dong, J.; Thomas, P.; Mineo, C.; Sirianni, R. 27-Hydroxycholesterol Binds GPER and Induces Progression of Estrogen Receptor-Negative Breast Cancer. Cancers 2022, 14, 1521. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, Y.; Du, F.Y.; Chen, W.; Fu, P.F.; Yao, M.Y.; Zheng, S.S. G15 Sensitizes Epithelial Breast Cancer Cells to Doxorubicin by Preventing Epithelial-Mesenchymal Transition through Inhibition of GPER. Am. J. Transl. Res. 2015, 7, 967–975. [Google Scholar]
  77. Qin, S.; Jiang, J.; Lu, Y.; Nice, E.C.; Huang, C.; Zhang, J.; He, W. Emerging Role of Tumor Cell Plasticity in Modifying Therapeutic Response. Signal Transduct. Target 2020, 5, 228. [Google Scholar]
  78. De Francesco, E.M.; Maggiolini, M.; Musti, A.M. Crosstalk between Notch, HIF-1α and GPER in Breast Cancer EMT. Int. J. Mol. Sci. 2018, 19, 2011. [Google Scholar] [CrossRef]
  79. Chen, Z.J.; Wei, W.; Jiang, G.M.; Liu, H.; Wei, W.D.; Yang, X.; Wu, Y.M.; Liu, H.; Wong, C.K.C.; Du, J. Activation of GPER Suppresses Epithelial Mesenchymal Transition of Triple Negative Breast Cancer Cells via NF-ΚB Signals. Mol. Oncol. 2016, 10, 775–788. [Google Scholar] [CrossRef]
  80. Baghban, R.; Roshangar, L.; Jahanban-Esfahlan, R.; Seidi, K.; Ebrahimi-Kalan, A.; Jaymand, M.; Kolahian, S.; Javaheri, T.; Zare, P. Tumor Microenvironment Complexity and Therapeutic Implications at a Glance. Cell Commun. Signal. 2020, 18, 59. [Google Scholar] [CrossRef]
  81. Ping, Q.; Yan, R.; Cheng, X.; Wang, W.; Zhong, Y.; Hou, Z.; Shi, Y.; Wang, C.; Li, R. Cancer-Associated Fibroblasts: Overview, Progress, Challenges, and Directions. Cancer Gene Ther. 2021, 28, 984–999. [Google Scholar] [CrossRef] [PubMed]
  82. Anderson, N.M.; Simon, M.C. Tumor Microencironment. Physiol. Behav. 2020, 30, R921–R925. [Google Scholar] [CrossRef]
  83. Cortés, M.; Sanchez-Moral, L.; de Barrios, O.; Fernández-Aceñero, M.J.; Martínez-Campanario, M.; Esteve-Codina, A.; Darling, D.S.; Győrffy, B.; Lawrence, T.; Dean, D.C. Tumor-associated Macrophages (TAMs) Depend on ZEB1 for Their Cancer-promoting Roles. EMBO J. 2017, 36, 3336–3355. [Google Scholar] [CrossRef]
  84. Wu, H.T.; Zhong, H.T.; Li, G.W.; Shen, J.X.; Ye, Q.Q.; Zhang, M.L.; Liu, J. Oncogenic Functions of the EMT-Related Transcription Factor ZEB1 in Breast Cancer. J. Transl. Med. 2020, 18, 51. [Google Scholar] [CrossRef]
  85. Sun, X.; Mao, Y.; Wang, J.; Zu, L.; Hao, M.; Cheng, G.; Qu, Q.; Cui, D.; Keller, E.T.; Chen, X. IL-6 Secreted by Cancer-Associated Fibroblasts Induces Tamoxifen Resistance in Luminal Breast Cancer. Oncogene 2014, 33, 4450. [Google Scholar] [CrossRef]
  86. Dittmer, A.; Lange, T.; Leyh, B.; Dittmer, J. Protein- And Growth-Modulatory Effects of Carcinoma-Associated Fibroblasts on Breast Cancer Cells: Role of Interleukin-6. Int. J. Oncol. 2020, 56, 258–272. [Google Scholar] [CrossRef] [PubMed]
  87. Wu, F.; Yang, J.; Liu, J.; Wang, Y.; Mu, J.; Zeng, Q.; Deng, S.; Zhou, H. Signaling Pathways in Cancer-Associated Fibroblasts and Targeted Therapy for Cancer. Signal Transduct. Target. Ther. 2021, 6, 218. [Google Scholar] [CrossRef]
  88. Iorga, A.; Cunningham, C.M.; Moazeni, S.; Ruffenach, G.; Umar, S.; Eghbali, M. The Protective Role of Estrogen and Estrogen Receptors in Cardiovascular Disease and the Controversial Use of Estrogen Therapy. Biol. Sex. Differ. 2017, 8, 33. [Google Scholar] [CrossRef]
  89. Singh, N.; Baby, D.; Rajguru, J.P.; Patil, P.B.; Thakkannavar, S.S.; Pujari, V.B. Inflammation and Cancer. Ann. Afr. Med. 2019, 1, 121–126. [Google Scholar] [CrossRef]
  90. Cendrowicz, E.; Sas, Z.; Bremer, E.; Rygiel, T.P. The Role of Macrophages in Cancer Development and Therapy. Cancers 2021, 13, 1946. [Google Scholar] [CrossRef]
  91. Zhou, J.; Tang, Z.; Gao, S.; Li, C.; Feng, Y.; Zhou, X. Tumor-Associated Macrophages: Recent Insights and Therapies. Front. Oncol. 2020, 10, 188. [Google Scholar] [CrossRef] [PubMed]
  92. Buoso, E.; Masi, M.; Racchi, M.; Corsini, E. Endocrine-Disrupting Chemicals’ (EDCs) Effects on Tumour Microenvironment and Cancer Progression: Emerging Contribution of RACK1. Int. J. Mol. Sci. 2020, 21, 9229. [Google Scholar] [CrossRef]
  93. Buoso, E.; Kenda, M.; Masi, M.; Linciano, P.; Galbiati, V.; Racchi, M.; Dolenc, M.S.; Corsini, E. Effects of Bisphenols on RACK1 Expression and Their Immunological Implications in THP-1 Cells. Front. Pharmacol. 2021, 12, 743991. [Google Scholar] [CrossRef] [PubMed]
  94. Masi, M.; Maddalon, A.; Iulini, M.; Linciano, P.; Galbiati, V.; Marinovich, M.; Racchi, M.; Corsini, E.; Buoso, E. Effects of endocrine disrupting chemicals on the expression of RACK1 and LPS-induced THP-1 cell activation. Toxicology 2022, 480, 153321. [Google Scholar] [CrossRef]
  95. Buoso, E.; Masi, M.; Galbiati, V.; Maddalon, A.; Iulini, M.; Kenda, M.; Sollner, D.M.; Marinovich, M.; Racchi, M.; Corsini, E. Effect of estrogen-active compounds on the expression of RACK1 and immunological implications. Arch. Toxicol. 2020, 94, 2081–2095. [Google Scholar] [CrossRef]
  96. Maddalon, A.; Masi, M.; Iulini, M.; Linciano, P.; Galbiati, V.; Marinovich, M.; Racchi, M.; Buoso, E.; Corsini, E. Effects of endocrine active contaminating pesticides on RACK1 expression and immunological consequences in THP-1 cells. Environ. Toxicol. Pharmacol. 2022, 95, 103971. [Google Scholar] [CrossRef]
  97. Maddalon, A.; Cari, L.; Iulini, M. Impact of endocrine disruptors on peripheral blood mononuclear cells in vitro: Role of gender. Arch. Toxicol. 2023. Online ahead of print. [Google Scholar] [CrossRef]
  98. Holm, A.; Baldetorp, B.; Olde, B.; Leeb-Lundberg, L.M.F.; Nilsson, B.O. The GPER1 Agonist G-1 Attenuates Endothelial Cell Proliferation by Inhibiting DNA Synthesis and Accumulating Cells in the S and G2 Phases of the Cell Cycle. J. Vasc. Res. 2011, 48, 327–335. [Google Scholar] [CrossRef]
  99. Chen, Z.; Yuhanna, I.S.; Galcheva-Gargova, Z.; Karas, R.H.; Mendelsohn, M.E.; Shaul, P.W. Estrogen Receptor α Mediates the Nongenomic Activation of Endothelial Nitric Oxide Synthase by Estrogen. J. Clin. Investig. 1999, 103, 401–406. [Google Scholar] [CrossRef] [PubMed]
  100. Lindner, V.; Kim, S.K.; Karas, R.H.; Kuiper, G.G.J.M.; Mendelsohn, M.E. Increased Expression of Estrogen Receptor-b MRNA in Male Blood Vessels After Vascular Injury. Circ. Res. 1998, 83, 224–229. [Google Scholar] [CrossRef]
  101. Zhou, L.; Chen, H.; Mao, X.; Qi, H.; Baker, P.N.; Hua, Z. G-Protein-Coupled Receptor 30 Mediates the Effects of Estrogen on Endothelial Cell Tube Formation in Vitro. Int. J. Mol. Med. 2017, 39, 1461–1467. [Google Scholar] [CrossRef]
  102. Lappano, R.; Rigiracciolo, D.; De Marco, P.; Avino, S.; Cappello, A.R.; Rosano, C.; Maggiolini, M.; De Francesco, E.M. Recent Advances on the Role of G Protein-Coupled Receptors in Hypoxia-Mediated Signaling. AAPS J. 2016, 18, 305–310. [Google Scholar] [CrossRef] [PubMed]
  103. Holm, A.; Grände, P.O.; Ludueña, R.F.; Olde, B.; Prasad, V.; Leeb-Lundberg, L.M.F.; Nilsson, B.O. The G Protein-Coupled Oestrogen Receptor 1 Agonist G-1 Disrupts Endothelial Cell Microtubule Structure in a Receptor-Independent Manner. Mol. Cell. Biochem. 2012, 366, 239–249. [Google Scholar] [CrossRef] [PubMed]
  104. De Francesco, E.M.; Pellegrino, M.; Santolla, M.F.; Lappano, R.; Ricchio, E.; Abonante, S.; Maggiolini, M. GPER Mediates Activation of HIF1α/VEGF Signaling by Estrogens. Cancer Res. 2014, 74, 4053–4064. [Google Scholar] [CrossRef]
  105. Maggiolini, M.; Santolla, M.F.; Avino, S.; Aiello, F.; Rosano, C.; Garofalo, A.; Grande, F. Identification of Two Benzopyrroloxazines Acting as Selective GPER Antagonists in Breast Cancer Cells and Cancer-Associated Fibroblasts. Futur. Med. Chem. 2015, 7, 437–448. [Google Scholar] [CrossRef]
  106. Rouhimoghadam, M.; Lu, A.S.; Salem, A.K.; Filardo, E.J. Therapeutic Perspectives on the Modulation of G-Protein Coupled Estrogen Receptor, GPER, Function. Front. Endocrinol. 2020, 11, 591217. [Google Scholar] [CrossRef]
  107. Arterburn, J.B.; Prossnitz, E.R. G Protein-Coupled Estrogen Receptor GPER: Molecular Pharmacology and Therapeutic Applications. Annu. Rev. Pharmacol. Toxicol. 2023, 63, 295–320. [Google Scholar] [CrossRef]
  108. Geng, X.; Chen, H.; Zhao, L.; Hu, J.; Yang, W.; Li, G.; Cheng, C.; Zhao, Z.; Zhang, T.; Sun, B. Cancer-Associated Fibroblast (CAF) Heterogeneity and Targeting Therapy of CAFs in Pancreatic Cancer. Front. Cell Dev. Biol. 2021, 9, 655152. [Google Scholar] [CrossRef]
  109. Rihawi, K.; Ricci, A.D.; Rizzo, A.; Brocchi, S.; Marasco, G.; Pastore, L.V.; Llimpe, F.L.R.; Golfieri, R.; Renzulli, M. Tumor-Associated Macrophages and Inflammatory Microenvironment in Gastric Cancer: Novel Translational Implications. Int. J. Mol. Sci. 2021, 22, 3805. [Google Scholar] [CrossRef] [PubMed]
  110. Qian, C.N.; Mei, Y.; Zhang, J. Cancer Metastasis: Issues and Challenges. Chin. J. Cancer 2017, 36, 36–39. [Google Scholar] [CrossRef]
  111. Jin, L.; Han, B.; Siegel, E.; Cui, Y.; Giuliano, A.; Cui, X. Breast Cancer Lung Metastasis: Molecular Biology and Therapeutic Implications. Cancer Biol. Ther. 2018, 19, 858–868. [Google Scholar] [CrossRef] [PubMed]
  112. Datta, K.; Muders, M.; Zhang, H.; Tindall, D.J. Mechanism of Lymph Node Metastasis in Prostate Cancer. Futur. Oncol. 2010, 6, 823–836. [Google Scholar] [CrossRef] [PubMed]
Figure 1. GPER localization and amino acid sequence. (a) The sequence of the 375 amino acids of the human protein is shown here. The seven transmembrane regions are indicated in yellow. (b) Three-dimensional representation of GPER. The modeling was performed using Swiss Model with the template Q99527.1.A. Modified from [9].
Figure 1. GPER localization and amino acid sequence. (a) The sequence of the 375 amino acids of the human protein is shown here. The seven transmembrane regions are indicated in yellow. (b) Three-dimensional representation of GPER. The modeling was performed using Swiss Model with the template Q99527.1.A. Modified from [9].
Ijms 24 14993 g001
Figure 3. GPER’s relationship with EMT in cancer. Overexpression of GPER in breast, lung, kidney, and ovarian cancer cells generates a kinase activation pathway that stimulates MMP-3, MMP-2/9, and MMP-9 activation, which degrades the adhesive proteins, favoring the promotion of metastasis. Additionally, specific miRNAs, like miRNA195, block GPER activation in ovarian cancer cells; consequently, MMP-2/9 activation via AKT/PI3K is diminished, preventing the degradation of adhesive proteins and interrupting the promotion of metastasis. Modified from references [31,49,52]. Created with BioRender.com (accessed on 25 August 2023).
Figure 3. GPER’s relationship with EMT in cancer. Overexpression of GPER in breast, lung, kidney, and ovarian cancer cells generates a kinase activation pathway that stimulates MMP-3, MMP-2/9, and MMP-9 activation, which degrades the adhesive proteins, favoring the promotion of metastasis. Additionally, specific miRNAs, like miRNA195, block GPER activation in ovarian cancer cells; consequently, MMP-2/9 activation via AKT/PI3K is diminished, preventing the degradation of adhesive proteins and interrupting the promotion of metastasis. Modified from references [31,49,52]. Created with BioRender.com (accessed on 25 August 2023).
Ijms 24 14993 g003
Figure 5. Future trends in GPER study.
Figure 5. Future trends in GPER study.
Ijms 24 14993 g005
Table 2. Role of miRNAs in GPER signaling in different cancer types.
Table 2. Role of miRNAs in GPER signaling in different cancer types.
miRNATargetGPER RegulationEffect
miR338-3pc-fosNegativeProliferation decreases in hepatocarcinoma [66]
miR-124CD151NegativeProliferation decreases in luminal breast cancer [67]
miR148-aHuman leukocyte antigen-GNegativeCell death in ovarian endometriosis [68]
miR-424GPERNegativeE2-induced proliferation in endometrial carcinoma [69]
miR-195GPERNegativePI3K/AKT-induced MMP-2/9 expression in endometrial carcinoma [70]
miR-199a-3pCD151PositivePromotes EMT in triple-negative breast cancer [71]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tirado-Garibay, A.C.; Falcón-Ruiz, E.A.; Ochoa-Zarzosa, A.; López-Meza, J.E. GPER: An Estrogen Receptor Key in Metastasis and Tumoral Microenvironments. Int. J. Mol. Sci. 2023, 24, 14993. https://doi.org/10.3390/ijms241914993

AMA Style

Tirado-Garibay AC, Falcón-Ruiz EA, Ochoa-Zarzosa A, López-Meza JE. GPER: An Estrogen Receptor Key in Metastasis and Tumoral Microenvironments. International Journal of Molecular Sciences. 2023; 24(19):14993. https://doi.org/10.3390/ijms241914993

Chicago/Turabian Style

Tirado-Garibay, Ana Carolina, Elba Andrea Falcón-Ruiz, Alejandra Ochoa-Zarzosa, and Joel E. López-Meza. 2023. "GPER: An Estrogen Receptor Key in Metastasis and Tumoral Microenvironments" International Journal of Molecular Sciences 24, no. 19: 14993. https://doi.org/10.3390/ijms241914993

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop