Open access peer-reviewed chapter

The Research Advances in G-Protein-Coupled Estrogen Receptor

Written By

Hong-Bing Zhang, Yao Wang and Bing Wang

Submitted: 06 June 2022 Reviewed: 13 June 2022 Published: 22 July 2022

DOI: 10.5772/intechopen.105822

From the Edited Volume

Estrogens - Recent Advances

Edited by Courtney Marsh

Chapter metrics overview

81 Chapter Downloads

View Full Metrics

Abstract

Estrogen binds to the typical estrogen receptor (ER) ERα or ERβ and is translocated to the nucleus, where it binds directly to the estrogen response element of the target gene to induce transcription and regulate gene expression, and the whole process is completed in several hours to several days. The G protein-coupled estrogen receptor (GPER), a type that is structurally distinct from typical ERα and ERβ, rapidly induces most non-genomic effects within seconds to minutes. GPER regulates cell growth, migration, and programmed cell death in a variety of tissues and has been associated with the progression of estrogen-associated cancers. Here, the characteristics, cell signal transduction, and the latest research progress of GPER in estrogen-associated tumors and retinal diseases are reviewed.

Keywords

  • estrogen receptor
  • G protein-coupled estrogen receptor
  • G-protein coupled receptor 30
  • GPER
  • GPR30

1. Introduction

Estrogen, the main sex hormone in cisgender women, plays an important role in regulating various physiological and pathological processes in the body and affects mood, bone strength, and even heart health beyond fertility and sex-related functions. Typical estrogen receptor (ER) includes ERα and ERβ. After combined with ERα or ERβ, estrogen transfers in nucleus and directly binds to the estrogen response element in the promoter region of target gene and then induces transcription and regulates gene expression, and this process is called estrogen genomic effect, which usually takes several hours to several days to complete this process [1].

Besides, by combined with a novel type of ER, G protein-coupled ER (GPER), an alternate ER with a structure distinct from the two canonical ERα and ERβ, can also induce rapid non-genomic effects in seconds to minutes and mediate most of the non-genomic effects of estrogen.

GPER triggers several downstream pathways that exert vital biological roles in the regulation of cell growth, migration, and programmed cell death in a variety of tissues. In particular, there is a significant correlation between GPER and the progression of estrogen-related cancers. Therefore, better understanding the role of GPER in physiological function, especially in estrogen-related cancers, may help identify new biological regulatory targets, as well as cancer therapeutic targets. In this review, we summarize the characteristic and cell signaling and detail the functions of GPER, as well as the latest research progress of GPER in estrogen-related cancers.

Advertisement

2. Brief overview of GPER

2.1 G-protein and GPCR

G proteins, also called as guanosine nucleotide-binding proteins, are proteins that are involved in delivering a variety of stimuli from extracellular environment to intracellular organelle. G protein-coupled receptors (GPCRs) are a general name of membrane protein receptors, which locate on cell membranes and are the largest family of proteins encoded by the human genome [2]. The unique feature of this kind of GPCRs is that there are seven transmembrane α helices in the stereo-structure of the receptor, and there are G protein-binding sites on the C-terminal of the peptide chain and the intracellular ring (the third intracellular ring) connecting the fifth and sixth transmembrane helices (starting from the N-terminal of the peptide chain). Endogenous ligands of GPCR include many compounds, such as amines, carbohydrates, lipids, peptides, proteins, hormones, neurotransmitters, chemokines, and even photons and odors. GPCR has been linked to type 2 diabetes, obesity, depression, cancer, Alzheimer’s disease, and other diseases [3].

2.2 ER

Estrogen receptor (ER) is a steroid receptor, belonging to a superfamily of proteins, whose function is to regulate transcription of gene pools in other cells. The estrogen receptors such as ERα, ERβ, and GPER are located in the tissues of the female reproductive tract and breast, as well as in a variety of tissues such as bone, brain, liver, colon, skin, and salivary glands, exerting the effects of estrogen compounds on their target tissues [4].

2.3 GPER

In 1996, the third estrogen receptor, G-protein coupled receptor 30 (GPR30), was discovered in breast cancer tissues for the first time [5, 6]. GPR30 is a member of GPCR family, which can be exclusively bound to by estrogen and estrogen-like molecules, and it is involved in rapid non-genomic estrogen effects [7, 8, 9]. GPR30 also has another name GPER or GPER1 [10]. The function of this receptor provides an important basis for expounding the rapid response of estrogen. The human GPER gene, including a 1128 bp open reading frame that encodes 375-amino acid receptors, is located on chromosome 7 [6, 11].

GPER is expressed in various tissues, such as nerve, reproductive, digestive, and muscle organs [12], as well as in brain pericellular, cortex, dentate gyrus, anterior pituitary, and adrenal medulla cells [13]. The expression of GPER is not limited to the cell surface, and the binding domain of this receptor is also expressed in the endoplasmic reticulum [14].

In normal tissue, GPER was expressed markedly in the cortex and the anterior pituitary, islets and pancreatic ducts, fundic glands of the stomach, the epithelium of the duodenum and gallbladder, hepatocytes, proximal tubules of the kidney, the adrenal medulla, and syncytiotrophoblasts and decidua cells of the placenta [15].

2.4 GPER agonists and antagonists

There are synthetic and natural estrogen compounds that can interact with GPER. GPER agonists can be categorized as follows: (1) natural estrogens including 17β-estradiol (E2, a general ER agonist) and 2-methoxy-estradiol; (2) phytoestrogens including genistein, quercetin, equol, and resveratrol; (3) selective estrogen receptor modulators (SERMs), which includes tamoxifen (TMX), 4-hydroxytamoxifen, and raloxifene; (4) selective estrogen receptor downregulation agents (SERDs), which include fulvestrant and pesticides; (5) plastic compounds as endocrine disruptors, which include atrazine, bisphenol A, zearalenone, nonylphenol, and ketone. GPER antagonists can be categorized as follows: (1) natural estrogens, which include 2-hydroxyoestradiol and estriol; and (2) synthesis of compounds such as MIBE [15, 16, 17].

In addition, there are highly selective GPER ligands, which include GPER agonist G-1 and GPER antagonists G-15 and G-36 [18, 19]. G-1, also known as Tespria, is a nonsteroidal compound and a selective GPER1 agonist. In vitro cell experiments have shown that G-1 has no binding affinity for ERα and ERβ [20, 21]. Moreover, G-1 regulation is involved in intracellular calcium [Ca2+] and phosphoadenosine 3-kinases (PI3Ks) signaling pathways. G-15 is a selective GPER1 antagonist with high binding affinity to GPER1 and less binding to ERα and ERβ [19, 22, 23]. As another selective GPER1 antagonist, G36 also owns poor binding ability to ERα and ERβ as well as G15, whereas G36 is more powerful than G15 in binding to GPER1 and then modulates Ca2+ mobilization and PI3K signaling [18].

Advertisement

3. GPER-related signaling pathway

3.1 Nuclear factor-Kappa B (NF-κB) signaling pathway

When epithelial cells undergo epithelial-mesenchymal transition (EMT), mastitis may be aggravated. GPER1 activation inhibits EMT of goat mammary epithelial cells through NF-κB signaling pathway, thus impeding the occurrence of mastitis [24]. Okamoto et al. [25] delineated that lipopolysaccharide (LPS)-induced interleukin-6 (IL-6) overexpression in mouse macrophages is negatively regulated by GPER agonist G-1 via NF-κB signaling pathway, which is associated with anti-inflammatory effects of estrogen.

3.2 Hippo signaling pathway

Zhou et al. [26] determined that, compared with adjacent normal tissue, the expression of GPER is highly upregulated in the cancer cells of breast invasive ductal carcinoma, and key downstream signals of GPER are associated with the Hippo/YAP (Yes-associated protein 1)/TAZ (transcriptional coactivator with a PDZ-binding domain) pathway, which plays an important role in breast tumorigenesis. YAP is a pivotal effector of Hippo pathway, and Deng et al. [27] exhibited that bisphenol S activates YAP, upgrades YAP nuclear accumulation, and regulates its downstream genes in triple-negative breast cancer cells. By inhibiting GPER, the effect of bisphenol S-induced Hippo/YAP signal pathway can be weakened.

3.3 Mitogen-activated protein kinase (MAPK) signaling pathway

Triclosan can act on GPER to induce estrogen effect. It has been found that triclosan can upregulate the expression of mir-144 by activating GPER, which leads to abnormal regulation of neuro-related genes and neurodevelopmental toxicity, and this process is closely related to the activation of downstream protein kinase C (PKC)/MAPK signaling pathway [28]. In ER-positive breast cancer cells, the activation of GPER and its downstream MAPK/ERK signal pathway increase the expression level of TRIM2 protein, which leads to the reduction of Bim protein in tamoxifen-resistant breast cancer cells, providing a new idea for solving tamoxifen resistance in ER-positive breast cancer cells [29].

3.4 Phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway

The nuclear ER is negative in breast cancer SKBR-3 cells, while GPER is positive. Using the SKBR-3 cells, Shi et al. [30] demonstrated that the expression of PI3K and p-AKT can be downregulated by cryptotanshinone (an active compound of traditional Chinese medicine Danshen), and the inhibitory effect of cryptotanshinone on SKBR-3 cells is carried out by inhibiting GPER-mediated PI3K/AKT pathway. Additionally, cryptotanshinone can mediate the downregulation of PI3K/AKT pathway and induce apoptosis in breast cancer MCF-7 cells by activating GPER [31].

3.5 Extracellular signal-regulated kinase (ERK) signaling pathway

Hepatocellular carcinoma (HCC) is a malignant tumor, whose cell lines HCCLM3 and SMMC-7721 express high levels of GPER. The research by Qiu et al. [32] presented that compared with GPER-negative patients, GPER-positive HCC patients were closely related to the following factors, such as female, negative hepatitis B surface antigen, smaller tumor size, lower serum alpha-fetoprotein level and longer overall survival, and treatment of HCC cell lines HCCLM3 and SMMC-7721 with GPER-specific agonist G1 resulted in the activation of epidermal growth factor receptor (EGFR)/ERK and EGFR/AKT signaling pathways. In a murine Crohn’s disease model induced by trinitrobenzene sulfonic acid, activation of GPER exerts the anti-inflammatory effects and lowers the mortality of mouse and C-reactive protein level, and this activation is accompanied by regulating ERK signaling pathway [33].

Advertisement

4. Physiological effects of GPER

4.1 GPER roles in inflammation

In a mouse model, the GPER selective agonist G-1 was found to reduce circulating levels of inflammatory cytokines tumor necrosis factor α (TNFα), monocyte chemoattractant protein-1 (MCP-1), and IL-6 and to lower the expression of inflammatory genes in a variety of metabolic tissues, when administered over a long period of time [34]. In microglia, GPER is involved in the anti-inflammatory process when genistein was administered to inhibit lipopolysaccharide (LPS)-induced microglia activation [35].

4.2 GPER roles in cardiovascular system

The blood pressure of male Sprague-Dawley (SD) rats can be significantly lowered by GPER1 activation with G1 [36]. Blood pressure is lowered in ovariectomy (OVX) SD rats and OVX mRen2 Lewis rats after chronic systemic administration with G-1 [37, 38]. The downstream of GPER1 activation involves signaling pathways such as nitric oxide (NO), which can mediate vasodilation occurrence and is a well-known vasodilator [39, 40, 41]. Downstream of GPER1 activation is also involved in renin angiotensin aldosterone system (RAAS), which controls blood pressure and glomerular filtration rate through renin and angiotensin I, II and aldosterone [42, 43, 44].

It is GPER1, while not ERα and ERβ, plays an important role in estrogen-responded heart rate regulation [45, 46]. GPER1 activation has a cardioprotective effect on heart failure in male mice and myocardial inflammation in male spontaneously hypertensive rats (SHRs), adriamycin-induced cardiotoxicity in male rats can be alleviated by GPER1 activation, and myocardial cell death can also be protected by GPER1 activation [47, 48, 49, 50]. Moreover, in stress-induced cardiomyopathy, also known as Takotsubo syndrome, GPER activation exerts a protective role through balancing β2 adrenergic receptor (β2AR) with the Gαs and Gαi signaling pathways [51].

In short, it is worth noting that the current studies on the physiological effects of GPER are mainly from GPER knockout (KO) mice, which may have certain limitations. Therefore, when using GPER KO mice to study the mechanism of GPER-mediated action, the possibility of systemic compensation in mice should be taken into account [52].

4.3 GPER roles in stress

Endoplasmic reticulum stress (ERS) has been linked to several diseases such as cancer and diabetes and may be one of the possible inducers of pathological cell death and dysfunction [53, 54]. Icariin, an active constituent of epimedium, can inhibit ERS pathway through promoting the expression of GPER in diabetic kidney disease, and GPER is negatively related to the expression of endoplasmic reticulum response stress protein [55]. ERS pathways can be activated by GPER agonist, which leads to cancer cell death [56, 57]. GPER agonist G-1 can inhibit the increase of colonic crypt cell apoptosis in the colitis model [58] and improve epithelial cells after intestinal injury [59], and the protective effect of GPER is at least partially correlated with the inhibition of ERS.

Oxidative stress refers to the imbalance between oxidation and antioxidant effects in the body. Oxidative stress can occur when overexposed to reactive oxygen and nitrogen species, leading to damage of proteins, lipids, and DNA [60, 61]. Icariin also inhibits oxidative stress and promotes experimental diabetic nephropathy repair through GPER-mediated degradation of Keap1 protein and activation of Nrf2 protein [62]. Studies using a male rat model with traumatic brain injury-induced liver injury found that oxidative stress was significantly inhibited by 17β-estradiol [63, 64, 65]. In human skin fibroblasts, keratinocytes, and hepatic cells’ 17 β-estradiol is also involved in inhibiting oxidative stress through the participation of GPER [66, 67, 68].

The range of cytoskeletal variation can be modulated by GPER, which is thought to be involved in mechanical transduction [69, 70]. When mechanical stress-mediated chondrocyte apoptosis occurs, chondrocyte apoptosis in osteoarthritis can be attenuated by GPER via causing the inhibition of Piezo 1 protein [71].

Advertisement

5. GPER in cancer

5.1 GPER in gynecological tumors

Bubb et al. [15] found that GPER is expressed in hepatocellular, pancreatic, renal and endometrial cancers, pancreatic neuroendocrine tumors, and pheochromocytomas by using GPER antibody 20H15L21. GPER is found to be expressed in 50–60% of breast cancer tissues [72, 73, 74], and is thought to be involved in the development of tamoxifen resistance in ERα-positive breast cancer [75, 76]. GPER can inhibit YAP1 phosphorylation by shutting down the Hippo pathway in breast cancer [27]. An immunohistochemical study of 1245 patients with primary invasive breast cancer showed that low expression of GPER was significantly related not only to clinicopathological and molecular characteristics of invasive behavior but also to poor survival in patients with breast cancer [77]. Chan et al. [78] showed that GPER induces phosphorylation of PKA and BAD-Ser118 to maintain breast cancer stem cells through activation of its ligands, which include tamoxifen (TMX) and GPER agonists increase cell proliferation and the number of breast cancer stem cells through the p-PKA/p-BAD signaling pathway.

GPER is expressed in cell lines such as human ovarian cancer cell line SKOV3, OVCAR-3, and OVCAR5 [79, 80], in both malignant and benign ovarian tumors, and overexpressed in some malignant ovarian tumors [81]. GPER is found in serous and mucinous ovarian adenocarcinoma biopsies. G-1 controls ovarian cancer cell proliferation, inhibits G2/M cell cycle progression, and promotes ovarian cancer cell apoptosis [79]. Zhu et al. showed that nuclear GPER is an independent negative marker for the prognosis of ovarian cancer patients, particularly for patients with highly malignant ovarian cancer [82].

Fujiwara et al. [83, 84] found that high GPER expression is related to poor prognosis in ovarian cancer patients, and lower GPER expression was positively correlated with overall survival time. However, in contrast to the results mentioned earlier, the study by Ignatov et al. found that high GPER expression positively affected 2-year disease-free survival in ovarian cancer patients [79]. The relation between GPER and ovarian cancer needs more studies to clarify.

5.2 GPER in tumor of male reproductive system

Concentrations of 17B-estradiol (E2) in the testis are 10 to 100 times higher than in the blood [85]. Although the molecular mechanisms by which estrogen and its receptors play a role in spermatogenesis are not fully understood, it is no doubt that estrogen is crucial in spermatogenesis [86]. GPER regulates the balance between testosterone and estrogen and is associated with the male reproductive system. GPER is predominantly located in the basal epithelium and mediates estrogen action, and the level of GPER expression is low in prostate tumor cells and in benign prostate tissue [87]. The expression of GPER was negatively correlated with the differentiation of prostate tumor cells [88]. In addition, it has been reported that GPER is a tumor suppressor that activates ERK1/2 and C-Jun/C-FOS signaling and thus inhibits PC3 cells, a cell line characteristic of prostatic small cell carcinoma and from arresting in G2 phase [89]. Lau et al. reported that GPER-specific agonist G1 binds to GPER to maintain ErK1/2 activation, thereby inhibiting prostate cancer cell growth and regulating metastasis characteristics [90].

Testicular germ cell carcinoma (TGCC) is a malignant solid tumor and is account for the main reason of death in young men. 17β-estradiol inhibits the proliferation of human seminoma cell lines by an ERβ-dependent mechanism, which is stimulated in vitro by GPER activation of ERK1/2 and protein kinase A. GPER is not expressed in non-seminoma, whereas GPER is exclusively overexpressed in seminoma, which is linked to the downregulation of ERβ, and may also be associated with genetic variations such as single-nucleotide polymorphisms [91].

It is worth mentioning that, regarding the potential sex-specific effects of GPER-1, there was no significant prognostic value of GPER-1 in the group of male breast cancers, which is contrary to what was expected in the studies of female breast cancers [92].

5.3 GPER in other cancer

Liu et al. [93, 94, 95] demonstrated that both cytoplasm-GPER (80.49%) and nucleus-GPER (53.05%) were detected in non-small-cell lung cancer (NSCLC) samples through immunohistochemical analysis, GPER activation promotes the growth of NSCLC cells via YAP1/QKI/circNOTCH1/m6A-methylated NOTCH1 pathway, and GPER antagonist G15 administration can reverse estrogen-induced progress of NSCLC cells, and these results suggest that blocking GPER signaling by G15 may be a new therapeutic target in NSCLC. In addition, the role of GPER in adrenal cortical carcinoma is related to mitochondria-related signal transduction, and GPER positively regulates mitochondrial apoptotic pathways through the EGR-1 pathway and ROS/EGR-1/BAX pathway, thereby inhibiting adrenocortical cancer cell growth [96].

Advertisement

6. GPER in retinal disease

Retinopathy of prematurity (ROP), characterized by abnormal growth of immature retinal blood vessels, is one of the leading causes of blindness and visual impairment in children worldwide. High incidence of ROP is intimately related to improved perinatal life care and poor ophthalmological special management in many developing countries, which lifts the survival rate of infants with ROP, and simultaneously, total infants with ROP [97].

Our research group has studied the role of GPER in ROP. The expression of GPER was found in endoplasmic reticulum of mouse retinal microglia, retinal ganglion cells, and retinal astrocytes. By GPER activation, hyperoxia-induced autophagy and apoptosis in these cells were reduced markedly, hyperoxia-induced decrease of the viability in these cells was reversed significantly, the high activity of inositol-1,4, 5-triphosphate receptor in these cells was reduced significantly also, and simultaneously, high calcium levels induced by hyperoxia in the endoplasmic reticulum was lowered again in these cells. These changes were reversed after the use of GPER antagonist G15. Thus, ERS irritated by hyperoxia was mitigated by G1 administration. These results suggest that GPER agonists may have therapeutic potential in the early stage of ROP [98, 99, 100].

Advertisement

7. Future directions

The key scientific issues that GPER needs to be further addressed as a key node are as follows: (1) the relationship between drugs (such as phytoestrogens, active ingredients of phytochemicals) and GPER and its signaling pathways needs to be systematically studied; (2) the interaction parameters and kinetics between GPER and ligands need to be further studied; (3) more work needs to be done to fully understand the potential of GPER1 in health regulatory mechanisms; and (4) the relationship between GPER and estrogen-related cancers, and the research on GPER as a therapeutic target for estrogen-related cancers needs to be carried out, which may unveil new therapies aimed at improving clinical outcomes of the diseases related to estrogen.

Advertisement

Acknowledgments

This work was supported by grants from the Key Project of Natural Science Basic Research Plan of Shaanxi Province (China, No. 2021JZ-60).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Abbreviations

EGFRepidermal growth factor receptor
E217-β-estradiol
ERestrogen receptor
GPCRG protein-coupled receptor
GPERG protein-coupled estrogen receptor
GPR30G protein-coupled receptor 30
HCChepatocellular carcinoma
KOknockout
SHRspontaneously hypertensive rats
SD ratSprague-Dawley rats
SERMselective estrogen receptor modulator
TGCCtesticular germ cell cancers
NSCLCnon-small-cell lung cancer

References

  1. 1. Filardo EJ, Thomas P. Minireview: G protein-coupled estrogen receptor-1, GPER-1: Its mechanism of action and role in female reproductive cancer, renal and vascular physiology. Endocrinology. 2012;153(7):2953-2962. DOI: 10.1210/en.2012-1061
  2. 2. Insel PA, Sriram K, Gorr MW, Wiley SZ, Michkov A, Salmerón C, et al. GPCRomics: An approach to discover GPCR drug targets. Trends in Pharmacological Sciences. 2019;40(6):378-387. DOI: 10.1016/j.tips.2019.04.001
  3. 3. Sriram K, Insel PA. G protein-coupled receptors as targets for approved drugs: How many targets and how many drugs? Molecular Pharmacology. 2018;93(4):251-258. DOI: 10.1124/mol.117.111062
  4. 4. Eyster KM. The estrogen receptors: An overview from different perspectives. Methods in Molecular Biology. 2016;1366:1-10. DOI: 10.1007/978-1-4939-3127-9_1
  5. 5. Takada Y, Kato C, Kondo S, Korenaga R, Ando J. Cloning of cDNAs encoding G protein-coupled receptor expressed in human endothelial cells exposed to fluid shear stress. Biochemical and Biophysical Research Communications. 1997;240(3):737-741. DOI: 10.1006/bbrc.1997.7734
  6. 6. Carmeci C, Thompson DA, Ring HZ, Francke U, Weigel RJ. 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(3):607-617. DOI: 10.1006/geno.1997.4972
  7. 7. Revankar CM, Cimino DF, Sklar LA, Arterburn JB, Prossnitz ER. A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science. 2005;307(5715):1625-1630. DOI: 10.1126/science.1106943
  8. 8. Filardo EJ, Quinn JA, Frackelton AR Jr, Bland KI. Estrogen action via the G protein-coupled receptor, GPR30: Stimulation of adenylyl cyclase and cAMP-mediated attenuation of the epidermal growth factor receptor-to-MAPK signaling axis. Molecular Endocrinology. 2002;16(1):70-84. DOI: 10.1210/mend.16.1.0758
  9. 9. Filardo E, Quinn J, Pang Y, Graeber C, Shaw S, Dong J, et al. Activation of the novel estrogen receptor G protein-coupled receptor 30 (GPR30) at the plasma membrane. Endocrinology. 2007;148(7):3236-3245. DOI: 10.1210/en.2006-1605
  10. 10. Ariazi EA, Brailoiu E, Yerrum S, Shupp HA, Slifker MJ, Cunliffe HE, et al. The G protein-coupled receptor GPR30 inhibits proliferation of estrogen receptor-positive breast cancer cells. Cancer Research. 2010;70(3):1184-1194. DOI: 10.1158/0008-5472.Can-09-3068
  11. 11. O'Dowd BF, Nguyen T, Marchese A, Cheng R, Lynch KR, Heng HH, et al. Discovery of three novel G-protein-coupled receptor genes. Genomics. 1998;47(2):310-313. DOI: 10.1006/geno.1998.5095
  12. 12. Olde B, Leeb-Lundberg LM. GPR30/GPER1: Searching for a role in estrogen physiology. Trends in Endocrinology and Metabolism. 2009;20(8):409-416. DOI: 10.1016/j.tem.2009.04.006
  13. 13. Isensee J, Meoli L, Zazzu V, Nabzdyk C, Witt H, Soewarto D, et al. Expression pattern of G protein-coupled receptor 30 in LacZ reporter mice. Endocrinology. 2009;150(4):1722-1730. DOI: 10.1210/en.2008-1488
  14. 14. Funakoshi T, Yanai A, Shinoda K, Kawano MM, Mizukami Y. G protein-coupled receptor 30 is an estrogen receptor in the plasma membrane. Biochemical and Biophysical Research Communications. 2006;346(3):904-910. DOI: 10.1016/j.bbrc.2006.05.191
  15. 15. Bubb M, Beyer A-SL, Dasgupta P, Kaemmerer D, Sänger J, Evert K, et al. Assessment of G protein-coupled oestrogen receptor expression in normal and neoplastic human tissues using a novel rabbit monoclonal antibody. International Journal of Molecular Sciences. 2022 May 6;23(9):5191. DOI: 10.3390/ijms23095191
  16. 16. Prossnitz ER, Arterburn JB. International union of basic and clinical pharmacology. XCVII. G protein-coupled estrogen receptor and its pharmacologic modulators. Pharmacological Reviews. 2015;67(3):505-540. DOI: 10.1124/pr.114.009712
  17. 17. Barton M, Prossnitz ER. Emerging roles of GPER in diabetes and atherosclerosis. Trends in Endocrinology and Metabolism. 2015;26(4):185-192. DOI: 10.1016/j.tem.2015.02.003
  18. 18. Dennis MK, Field AS, Burai R, Ramesh C, Petrie WK, Bologa CG, et al. Identification of a GPER/GPR30 antagonist with improved estrogen receptor counterselectivity. The Journal of Steroid Biochemistry and Molecular Biology. 2011;127(3-5):358-366. DOI: 10.1016/j.jsbmb.2011.07.002
  19. 19. Dennis MK, Burai R, Ramesh C, Petrie WK, Alcon SN, Nayak TK, et al. In vivo effects of a GPR30 antagonist. Nature Chemical Biology. 2009;5(6):421-427. DOI: 10.1038/nchembio.168
  20. 20. Bologa CG, Revankar CM, Young SM, Edwards BS, Arterburn JB, Kiselyov AS, et al. Virtual and biomolecular screening converge on a selective agonist for GPR30. Nature Chemical Biology. 2006;2(4):207-212. DOI: 10.1038/nchembio775
  21. 21. Albanito L, Madeo A, Lappano R, Vivacqua A, Rago V, Carpino A, et al. G protein-coupled receptor 30 (GPR30) mediates gene expression changes and growth response to 17beta-estradiol and selective GPR30 ligand G-1 in ovarian cancer cells. Cancer Research. 2007;67(4):1859-1866. DOI: 10.1158/0008-5472.Can-06-2909
  22. 22. Di Mattia RA, Mariángelo JIE, Blanco PG, Jaquenod De Giusti C, Portiansky EL, Mundiña-Weilenmann C, et al. The activation of the G protein-coupled estrogen receptor (GPER) prevents and regresses cardiac hypertrophy. Life Sciences. 2020;242:117211. DOI: 10.1016/j.lfs.2019.117211
  23. 23. Groban L, Tran QK, Ferrario CM, Sun X, Cheng CP, Kitzman DW, et al. Female heart health: Is GPER the missing link? Frontiers in Endocrinology (Lausanne). 2019;10:919. DOI: 10.3389/fendo.2019.00919
  24. 24. Zhao Y, Yang Z, Miao Y, Fan M, Zhao X, Wei Q , et al. G protein-coupled estrogen receptor 1 inhibits the epithelial-mesenchymal transition of goat mammary epithelial cells via NF-κB signalling pathway. Reproduction in Domestic Animals. 2021;56(8):1137-1144. DOI: 10.1111/rda.13957
  25. 25. Okamoto M, Suzuki T, Mizukami Y, Ikeda T. The membrane-type estrogen receptor G-protein-coupled estrogen receptor suppresses lipopolysaccharide-induced interleukin 6 via inhibition of nuclear factor-kappa B pathway in murine macrophage cells. Animal Science Journal. 2017;88(11):1870-1879. DOI: 10.1111/asj.12868
  26. 26. Zhou X, Wang S, Wang Z, Feng X, Liu P, Lv XB, et al. Estrogen regulates Hippo signaling via GPER in breast cancer. The Journal of Clinical Investigation. 2015;125(5):2123-2135. DOI: 10.1172/jci79573
  27. 27. Deng Q , Jiang G, Wu Y, Li J, Liang W, Chen L, et al. GPER/Hippo-YAP signal is involved in Bisphenol S induced migration of triple negative breast cancer (TNBC) cells. Journal of Hazardous Materials. 2018;355:1-9. DOI: 10.1016/j.jhazmat.2018.05.013
  28. 28. Diao W, Qian Q , Sheng G, He A, Yan J, Dahlgren RA, et al. Triclosan targets miR-144 abnormal expression to induce neurodevelopmental toxicity mediated by activating PKC/MAPK signaling pathway. Journal of Hazardous Materials. 2022;431:128560. DOI: 10.1016/j.jhazmat.2022.128560
  29. 29. Yin H, Zhu Q , Liu M, Tu G, Li Q , Yuan J, et al. GPER promotes tamoxifen-resistance in ER+ breast cancer cells by reduced Bim proteins through MAPK/Erk-TRIM2 signaling axis. International Journal of Oncology. 2017;51(4):1191-1198. DOI: 10.3892/ijo.2017.4117
  30. 30. Shi D, Zhao P, Cui L, Li H, Sun L, Niu J, et al. Inhibition of PI3K/AKT molecular pathway mediated by membrane estrogen receptor GPER accounts for cryptotanshinone induced antiproliferative effect on breast cancer SKBR-3 cells. BMC. Pharmacology and Toxicology. 2020;21(1):32. DOI: 10.1186/s40360-020-00410-9
  31. 31. Shi D, Li H, Zhang Z, He Y, Chen M, Sun L, et al. Cryptotanshinone inhibits proliferation and induces apoptosis of breast cancer MCF-7 cells via GPER mediated PI3K/AKT signaling pathway. PLoS One. 2022;17(1):e0262389. DOI: 10.1371/journal.pone.0262389
  32. 32. Qiu Y-A, Xiong J, Fu Q , Dong Y, Liu M, Peng M, et al. GPER-induced ERK signaling decreases cell viability of hepatocellular carcinoma. Frontiers in Oncology. 2021;11:638171. DOI: 10.3389/fonc.2021.638171
  33. 33. Jacenik D, Zielińska M, Mokrowiecka A, Michlewska S, Małecka-Panas E, Kordek R, et al. G protein-coupled estrogen receptor mediates anti-inflammatory action in Crohn's disease. Scientific Reports. 2019;9(1):6749. DOI: 10.1038/s41598-019-43233-3
  34. 34. Sharma G, Hu C, Staquicini DI, Brigman JL, Liu M, Mauvais-Jarvis F, et al. Preclinical efficacy of the GPER-selective agonist G-1 in mouse models of obesity and diabetes. Science Translational Medicine. 2020;12(528). DOI: 10.1126/scitranslmed.aau5956
  35. 35. Du ZR, Feng XQ , Li N, Qu JX, Feng L, Chen L, et al. G protein-coupled estrogen receptor is involved in the anti-inflammatory effects of genistein in microglia. Phytomedicine. 2018;43:11-20. DOI: 10.1016/j.phymed.2018.03.039
  36. 36. Haas E, Bhattacharya I, Brailoiu E, Damjanović M, Brailoiu GC, Gao X, et al. Regulatory role of G protein-coupled estrogen receptor for vascular function and obesity. Circulation Research. 2009;104(3):288-291. DOI: 10.1161/circresaha.108.190892
  37. 37. Gohar EY, Daugherty EM, Aceves JO, Sedaka R, Obi IE, Allan JM, et al. Evidence for G-protein-coupled estrogen receptor as a pronatriuretic factor. Journal of the American Heart Association. 2020;9(10):e015110. DOI: 10.1161/jaha.119.015110
  38. 38. Lindsey SH, Cohen JA, Brosnihan KB, Gallagher PE, Chappell MC. Chronic treatment with the G protein-coupled receptor 30 agonist G-1 decreases blood pressure in ovariectomized mRen2.Lewis rats. Endocrinology. 2009;150(8):3753-3758. DOI: 10.1210/en.2008-1664
  39. 39. Lindsey SH, Liu L, Chappell MC. Vasodilation by GPER in mesenteric arteries involves both endothelial nitric oxide and smooth muscle cAMP signaling. Steroids. 2014;81:99-102. DOI: 10.1016/j.steroids.2013.10.017
  40. 40. Tropea T, De Francesco EM, Rigiracciolo D, Maggiolini M, Wareing M, Osol G, et al. Pregnancy augments G protein estrogen receptor (GPER) induced vasodilation in rat uterine arteries via the nitric oxide—cGMP signaling pathway. PLoS One. 2015;10(11):e0141997. DOI: 10.1371/journal.pone.0141997
  41. 41. Lindsey SH, da Silva AS, Silva MS, Chappell MC. Reduced vasorelaxation to estradiol and G-1 in aged female and adult male rats is associated with GPR30 downregulation. American Journal of Physiology. Endocrinology and Metabolism. 2013;305(1):E113-E118. DOI: 10.1152/ajpendo.00649.2012
  42. 42. Gros R, Ding Q , Liu B, Chorazyczewski J, Feldman RD. Aldosterone mediates its rapid effects in vascular endothelial cells through GPER activation. American Journal of Physiology. Cell Physiology. 2013;304(6):C532-C540. DOI: 10.1152/ajpcell.00203.2012
  43. 43. Ferreira NS, Cau SB, Silva MA, Manzato CP, Mestriner FL, Matsumoto T, et al. Diabetes impairs the vascular effects of aldosterone mediated by G protein-coupled estrogen receptor activation. Frontiers in Pharmacology. 2015;6:34. DOI: 10.3389/fphar.2015.00034
  44. 44. Rigiracciolo DC, Scarpelli A, Lappano R, Pisano A, Santolla MF, Avino S, et al. GPER is involved in the stimulatory effects of aldosterone in breast cancer cells and breast tumor-derived endothelial cells. Oncotarget. 2016;7(1):94-111. DOI: 10.18632/oncotarget.6475
  45. 45. Anderson JC, Beyger L, Guchardi J, Holdway DA. The effects of 17α-ethinylestradiol on the heart rate of embryonic Japanese medaka (Oryzias latipes). Environmental Toxicology and Chemistry. 2020;39(4):904-912. DOI: 10.1002/etc.4691
  46. 46. Romano SN, Edwards HE, Souder JP, Ryan KJ, Cui X, Gorelick DA. G protein-coupled estrogen receptor regulates embryonic heart rate in zebrafish. PLoS Genetics. 2017;13(10):e1007069. DOI: 10.1371/journal.pgen.1007069
  47. 47. Zhang X, Li T, Cheng HJ, Wang H, Ferrario CM, Groban L, et al. Chronic GPR30 agonist therapy causes restoration of normal cardiac functional performance in a male mouse model of progressive heart failure: Insights into cellular mechanisms. Life Sciences. 2021;285:119955. DOI: 10.1016/j.lfs.2021.119955
  48. 48. Li WL, Xiang W, Ping Y. Activation of novel estrogen receptor GPER results in inhibition of cardiocyte apoptosis and cardioprotection. Molecular Medicine Reports. 2015;12(2):2425-2430. DOI: 10.3892/mmr.2015.3674
  49. 49. Deschamps AM, Murphy E. Activation of a novel estrogen receptor, GPER, is cardioprotective in male and female rats. American Journal of Physiology. Heart and Circulatory Physiology. 2009;297(5):H1806-H1813. DOI: 10.1152/ajpheart.00283.2009
  50. 50. De Francesco EM, Rocca C, Scavello F, Amelio D, Pasqua T, Rigiracciolo DC, et al. Protective role of GPER agonist G-1 on cardiotoxicity induced by doxorubicin. Journal of Cellular Physiology. 2017;232(7):1640-1649. DOI: 10.1002/jcp.25585
  51. 51. Fu L, Zhang H, Ong'achwa Machuki J, Zhang T, Han L, Sang L, et al. GPER mediates estrogen cardioprotection against epinephrine-induced stress. The Journal of Endocrinology. 2021;249(3):209-222. DOI: 10.1530/joe-20-0451
  52. 52. Sharma G, Prossnitz ER. Targeting the G protein-coupled estrogen receptor (GPER) in obesity and diabetes. Endocrine and Metabolic Science. 2021;2. DOI: 10.1016/j.endmts.2021.100080
  53. 53. Xu C, Bailly-Maitre B, Reed JC. Endoplasmic reticulum stress: Cell life and death decisions. The Journal of Clinical Investigation. 2005;115(10):2656-2664. DOI: 10.1172/JCI26373
  54. 54. Shen X, Zhang K, Kaufman RJ. The unfolded protein response—A stress signaling pathway of the endoplasmic reticulum. Journal of Chemical Neuroanatomy. 2004;28(1-2):79-92. DOI: 10.1016/j.jchemneu.2004.02.006
  55. 55. Su B, Cheng D, Chen G, Zhang S, Wang L, Wu X, et al. Icariin attenuation of diabetic kidney disease through inhibition of endoplasmic reticulum stress via G protein-coupled estrogen receptors. Journal of Biomedical Nanotechnology. 2022;18(2):488-497. DOI: 10.1166/jbn.2022.3242
  56. 56. Vo DH, 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(9). DOI: 10.3390/biom9090503
  57. 57. Lee SJ, Kim TW, Park GL, Hwang YS, Cho HJ, Kim JT, et al. G protein-coupled estrogen receptor-1 agonist induces chemotherapeutic effect via ER stress signaling in gastric cancer. BMB Reports. 2019;52(11):647-652. DOI: 10.5483/BMBRep.2019.52.11.007
  58. 58. Wang Q , Li Z, Liu K, Liu J, Chai S, Chen G, et al. Activation of the G protein-coupled estrogen receptor prevented the development of acute colitis by protecting the crypt cell. The Journal of Pharmacology and Experimental Therapeutics. 2021;376(2):281-293. DOI: 10.1124/jpet.120.000216
  59. 59. Chai S, Liu K, Feng W, Liu T, Wang Q , Zhou R, et al. Activation of G protein-coupled estrogen receptor protects intestine from ischemia/reperfusion injury in mice by protecting the crypt cell proliferation. Clinical Science (London, England). 2019;133(3):449-464. DOI: 10.1042/cs20180919
  60. 60. Bowler RP, Crapo JD. Oxidative stress in allergic respiratory diseases. The Journal of Allergy and Clinical Immunology. 2002;110(3):349-356. DOI: 10.1067/mai.2002.126780
  61. 61. Pizzino G, Irrera N, Cucinotta M, Pallio G, Mannino F, Arcoraci V, et al. Oxidative stress: Harms and benefits for human health. Oxidative Medicine and Cellular Longevity. 2017;2017:8416763, 10.1155/2017/8416763
  62. 62. Wang K, Zheng X, Pan Z, Yao W, Gao X, Wang X, et al. Icariin prevents extracellular matrix accumulation and ameliorates experimental diabetic kidney disease by inhibiting oxidative stress via GPER mediated p62-dependent Keap1 degradation and Nrf2 activation. Frontiers in Cell and Development Biology. 2020;8:559. DOI: 10.3389/fcell.2020.00559
  63. 63. Amiresmaili S, Khaksari M, Shahrokhi N, Abolhassani M. Evolution of TLR4 role in mediating the hepatoprotective effects of estradiol after traumatic brain injury in male rats. Biochemical Pharmacology. 2020;178:114044. DOI: 10.1016/j.bcp.2020.114044
  64. 64. Amiresmaili S, Shahrokhi N, Khaksari M, AsadiKaram G, Aflatoonian MR, Shirazpour S, et al. The Hepatoprotective mechanisms of 17β-estradiol after traumatic brain injury in male rats: Classical and non-classical estrogen receptors. Ecotoxicology and Environmental Safety. 2021;213:111987. DOI: 10.1016/j.ecoenv.2021.111987
  65. 65. Amirkhosravi L, Khaksari M, Soltani Z, Esmaeili-Mahani S, Asadi Karam G, Hoseini M. E2-BSA and G1 exert neuroprotective effects and improve behavioral abnormalities following traumatic brain injury: The role of classic and non-classic estrogen receptors. Brain Research. 2021;1750:147168. DOI: 10.1016/j.brainres.2020.147168
  66. 66. Savoia P, Raina G, Camillo L, Farruggio S, Mary D, Veronese F, et al. Anti-oxidative effects of 17 β-estradiol and genistein in human skin fibroblasts and keratinocytes. Journal of Dermatological Science. 2018;92(1):62-77. DOI: 10.1016/j.jdermsci.2018.07.007
  67. 67. Surico D, Ercoli A, Farruggio S, Raina G, Filippini D, Mary D, et al. Modulation of oxidative stress by 17 β-estradiol and genistein in human hepatic cell lines in vitro. Cellular Physiology and Biochemistry. 2017;42(3):1051-1062. DOI: 10.1159/000478752
  68. 68. Farruggio S, Raina G, Cocomazzi G, Librasi C, Mary D, Gentilli S, et al. Genistein improves viability, proliferation and mitochondrial function of cardiomyoblasts cultured in physiologic and peroxidative conditions. International Journal of Molecular Medicine. 2019;44(6):2298-2310. DOI: 10.3892/ijmm.2019.4365
  69. 69. Lachowski D, Cortes E, Matellan C, Rice A, Lee DA, Thorpe SD, et al. G protein-coupled estrogen receptor regulates actin cytoskeleton dynamics to impair cell polarization. Frontiers in Cell and Development Biology. 2020;8:592628. DOI: 10.3389/fcell.2020.592628
  70. 70. Wang Z, Sun L, Liang S, Liu ZC, Zhao ZY, Yang J, et al. GPER stabilizes F-actin cytoskeleton and activates TAZ via PLCβ-PKC and Rho/ROCK-LIMK-cofilin pathway. Biochemical and Biophysical Research Communications. 2019;516(3):976-982. DOI: 10.1016/j.bbrc.2019.06.132
  71. 71. Sun Y, Leng P, Guo P, Gao H, Liu Y, Li C, et al. G protein coupled estrogen receptor attenuates mechanical stress-mediated apoptosis of chondrocyte in osteoarthritis via suppression of Piezo1. Molecular Medicine. 2021;27(1):96. DOI: 10.1186/s10020-021-00360-w
  72. 72. Aiello F, Carullo G, Giordano F, Spina E, Nigro A, Garofalo A, et al. Identification of breast cancer inhibitors specific for G protein-coupled estrogen receptor (GPER)-expressing cells. ChemMedChem. 2017;12(16):1279-1285. DOI: 10.1002/cmdc.201700145
  73. 73. Molina L, Figueroa CD, Bhoola KD, Ehrenfeld P. GPER-1/GPR30 a novel estrogen receptor sited in the cell membrane: Therapeutic coupling to breast cancer. Expert Opinion on Therapeutic Targets. 2017;21(8):755-766. DOI: 10.1080/14728222.2017.1350264
  74. 74. Jacenik D, Cygankiewicz AI, Krajewska WM. The G protein-coupled estrogen receptor as a modulator of neoplastic transformation. Molecular and Cellular Endocrinology. 2016;429:10-18. DOI: 10.1016/j.mce.2016.04.011
  75. 75. Nass N, Kalinski T. Tamoxifen resistance: From cell culture experiments towards novel biomarkers. Pathology, Research and Practice. 2015;211(3):189-197. DOI: 10.1016/j.prp.2015.01.004
  76. 76. Rondón-Lagos M, Villegas VE, Rangel N, Sánchez MC, Zaphiropoulos PG. Tamoxifen resistance: Emerging molecular targets. International Journal of Molecular Sciences. 2016;17(8). DOI: 10.3390/ijms17081357
  77. 77. Martin SG, Lebot MN, Sukkarn B, Ball G, Green AR, Rakha EA, et al. Low expression of G protein-coupled oestrogen receptor 1 (GPER) is associated with adverse survival of breast cancer patients. Oncotarget. 2018;9(40):25946-25956. DOI: 10.18632/oncotarget.25408
  78. 78. Chan Y-T, Lai ACY, Lin R-J, Wang Y-H, Wang Y-T, Chang W-W, et al. GPER-induced signaling is essential for the survival of breast cancer stem cells. International Journal of Cancer. 2020;146(6):1674-1685. DOI: 10.1002/ijc.32588
  79. 79. Ignatov T, Modl S, Thulig M, Weißenborn C, Treeck O, Ortmann O, et al. GPER-1 acts as a tumor suppressor in ovarian cancer. Journal of Ovarian Research. 2013;6(1):51. DOI: 10.1186/1757-2215-6-51
  80. 80. Yan Y, Jiang X, Zhao Y, Wen H, Liu G. Role of GPER on proliferation, migration and invasion in ligand-independent manner in human ovarian cancer cell line SKOV3. Cell Biochemistry and Function. 2015;33(8):552-559. DOI: 10.1002/cbf.3154
  81. 81. Kolkova Z, Casslén V, Henic E, Ahmadi S, Ehinger A, Jirström K, et al. The G protein-coupled estrogen receptor 1 (GPER/GPR30) does not predict survival in patients with ovarian cancer. Journal of Ovarian Research. 2012;5:9. DOI: 10.1186/1757-2215-5-9
  82. 82. Zhu CX, Xiong W, Wang ML, Yang J, Shi HJ, Chen HQ , et al. Nuclear G protein-coupled oestrogen receptor (GPR30) predicts poor survival in patients with ovarian cancer. The Journal of International Medical Research. 2018;46(2):723-731. DOI: 10.1177/0300060517717625
  83. 83. Fujiwara S, Terai Y, Kawaguchi H, Takai M, Yoo S, Tanaka Y, et al. GPR30 regulates the EGFR-Akt cascade and predicts lower survival in patients with ovarian cancer. Journal of Ovarian Research. 2012;5(1):35. DOI: 10.1186/1757-2215-5-35
  84. 84. Heublein S, Mayr D, Vrekoussis T, Friese K, Hofmann SS, Jeschke U, et al. The G-protein coupled estrogen receptor (GPER/GPR30) is a gonadotropin receptor dependent positive prognosticator in ovarian carcinoma patients. PLoS One. 2013;8(8):e71791. DOI: 10.1371/journal.pone.0071791
  85. 85. Carreau S, Bourguiba S, Lambard S, Galeraud-Denis I, Genissel C, Levallet J. Reproductive system: Aromatase and estrogens. Molecular and Cellular Endocrinology. 2002;193(1-2):137-143. DOI: 10.1016/s0303-7207(02)00107-7
  86. 86. Carreau S, Bouraima-Lelong H, Delalande C. Role of estrogens in spermatogenesis. Frontiers in Bioscience (Elite Edition). 2012;4(1):1-11. DOI: 10.2741/356
  87. 87. Rago V, Romeo F, Giordano F, Ferraro A, Carpino A. Identification of the G protein-coupled estrogen receptor (GPER) in human prostate: Expression site of the estrogen receptor in the benign and neoplastic gland. Andrology. 2016;4(1):121-127. DOI: 10.1111/andr.12131
  88. 88. Xu S, Yu S, Dong D, Lee LTO. G protein-coupled estrogen receptor: A potential therapeutic target in cancer. Frontiers in endocrinology. 2019;10:725. DOI: 10.3389/fendo.2019.00725
  89. 89. Chan QK, Lam HM, Ng CF, Lee AY, Chan ES, Ng HK, et al. Activation of GPR30 inhibits the growth of prostate cancer cells through sustained activation of Erk1/2, c-jun/c-fos-dependent upregulation of p21, and induction of G(2) cell-cycle arrest. Cell Death and Differentiation. 2010;17(9):1511-1523. DOI: 10.1038/cdd.2010.20
  90. 90. Lau KM, Ma FM, Xia JT, Chan QKY, Ng CF, To KF. Activation of GPR30 stimulates GTP-binding of Gαi1 protein to sustain activation of Erk1/2 in inhibition of prostate cancer cell growth and modulates metastatic properties. Experimental Cell Research. 2017;350(1):199-209. DOI: 10.1016/j.yexcr.2016.11.022
  91. 91. Chevalier N, Hinault C, Clavel S, Paul-Bellon R, Fenichel P. GPER and testicular germ cell cancer. Frontiers in Endocrinology. 2021;11:600404. DOI: 10.3389/fendo.2020.600404
  92. 92. Maiwald JH, Sprung S, Czapiewski P, Lessel W, Scherping A, Schomburg D, et al. The impact of G protein-coupled oestrogen receptor 1 on male breast cancer: A retrospective analysis. Contemporary Oncology (Pozn). 2021;25(3):204-212. DOI: 10.5114/wo.2021.110010
  93. 93. Liu C, Liao Y, Fan S, Tang H, Jiang Z, Zhou B, et al. G protein-coupled estrogen receptor (GPER) mediates NSCLC progression induced by 17β-estradiol (E2) and selective agonist G1. Medical Oncology. 2015;32(4):104. DOI: 10.1007/s12032-015-0558-2
  94. 94. Liu C, Liao Y, Fan S, Fu X, Xiong J, Zhou S, et al. G-protein-coupled estrogen receptor antagonist G15 decreases estrogen-induced development of non-small cell lung cancer. Oncology Research. 2019;27(3):283-292. DOI: 10.3727/096504017x15035795904677
  95. 95. Shen Y, Li C, Zhou L, Huang JA. G protein-coupled oestrogen receptor promotes cell growth of non-small cell lung cancer cells via YAP1/QKI/circNOTCH1/m6A methylated NOTCH1 signalling. Journal of Cellular and Molecular Medicine. 2021;25(1):284-296. DOI: 10.1111/jcmm.15997
  96. 96. Casaburi I, Avena P, De Luca A, Sirianni R, Rago V, Chimento A, et al. GPER-independent inhibition of adrenocortical cancer growth by G-1 involves ROS/Egr-1/BAX pathway. Oncotarget. 2017;8(70):115609-115619. DOI: 10.18632/oncotarget.23314
  97. 97. Romo-Aguas JC, González HLA, Meraz-Gutiérrez MP, Martínez-Castellanos MA. Retinopathy of prematurity: Incidence report of outliers based on international screening guidelines. International Journal of Retina Vitreous. 2019;5(Suppl. 1):53. DOI: 10.1186/s40942-019-0203-x
  98. 98. Li R, Wang Y, Chen P, Meng J, Zhang H. G-protein coupled estrogen receptor activation protects the viability of hyperoxia-treated primary murine retinal microglia by reducing ER stress. Aging (Albany NY). 2020;12(17):17367-17379. DOI: 10.18632/aging.103733
  99. 99. Li R, Wang Y, Chen P, Meng J, Zhang H. Inhibiting endoplasmic reticulum stress by activation of G-protein-coupled estrogen receptor to protect retinal astrocytes under hyperoxia. Journal of Biochemical and Molecular Toxicology. 2021;35(2):e22641. DOI: 10.1002/jbt.22641
  100. 100. Li R, Wang Y, Chen P, Meng J, Zhang H. G-protein-coupled estrogen receptor protects retinal ganglion cells via inhibiting endoplasmic reticulum stress under hyperoxia. Journal of Cellular Physiology. 2021;236(5):3780-3788. DOI: 10.1002/jcp.30149

Written By

Hong-Bing Zhang, Yao Wang and Bing Wang

Submitted: 06 June 2022 Reviewed: 13 June 2022 Published: 22 July 2022