252 results on '"G protein-coupled estrogen receptor"'
Search Results
2. The activation of the G-protein-coupled estrogen receptor promotes the aggressiveness of MDA-MB231 cells by targeting the IRE1α/TXNIP pathway
- Author
-
Maryam Mohammad-Sadeghipour, Mohammad Hadi Nematollahi, Hassan Ahmadinia, Mohammad Reza Hajizadeh, and Mehdi Mahmoodi
- Subjects
breast cancer ,drug resistance ,g protein-coupled estrogen receptor ,mir-17-5p ,thioredoxin interacting protein ,unfolded protein response ,Pharmacy and materia medica ,RS1-441 - Abstract
Background and purpose: This study investigated modulating the G protein-coupled estrogen receptor (GPER) on the IRElα/TXNIP pathway and its role in drug resistance in MDA-MB231 cells. Experimental approach: To determine the optimal concentrations of G1 and 4-hydroxytamoxifen (TAM), GPER expression and ERK1/2 phosphorylation were analyzed using qRT-PCR and western blotting, respectively. Cells were treated with individual concentrations of G1 (1000 nM), G15 (1000 nM), and TAM (2000 nM), as well as combinations of these treatments (G1 + G15, TAM + G15, and G1 + TAM) for 24 and 48 h. The expression levels of GPER, IRE1α, miR-17-5p, TXNIP, ABCB1, and ABCC1 genes and TXNIP protein expression were evaluated. Finally, apoptosis and cell migration were examined using flow cytometry and the wound-healing assay, respectively. Findings/Results: Activating GPER with its specific agonist G1 and TAM significantly increased IRE1α levels in MDA-MB231 cells. IRE1α through splicing XBP1 led to unfolded protein response. In addition, decreased TXNIP gene and protein expression reduced apoptosis, increased migration, and upregulated the genes associated with drug resistance. Conclusion and implication: Our investigation revealed that blocking the GPER/IRE1α/TXNIP pathway in MDA-MB231 cells could enhance treatment efficacy and improve chemotherapy responsiveness. The distinct unfolded protein response observed in MDA-MB231 cells may stem from the unique characteristics of these cells, which lack receptors for estrogen, progesterone, and HER2/neu hormones, possessing only the GPER receptor (ER-/PR-/HER2-/GPER+). This study introduced a new pathway in TNBC cells, indicating that targeting GPER could be crucial in comprehensive therapeutic strategies in TNBC cells.
- Published
- 2024
- Full Text
- View/download PDF
3. The activation of the G-protein-coupled estrogen receptor promotes the aggressiveness of MDA-MB231 cells by targeting the IRE1α/TXNIP pathway.
- Author
-
Mohammad-Sadeghipour, Maryam, Nematollahi, Mohammad Hadi, Ahmadinia, Hassan, Hajizadeh, Mohammad Reza, and Mahmoodi, Mehdi
- Subjects
THIOREDOXIN-interacting protein ,ESTROGEN receptors ,UNFOLDED protein response ,DRUG resistance in cancer cells ,G protein coupled receptors ,PROGESTERONE receptors - Abstract
Background and purpose: This study investigated modulating the G protein-coupled estrogen receptor (GPER) on the IRE1α/TXNIP pathway and its role in drug resistance in MDA-MB231 cells. Experimental approach: To determine the optimal concentrations of G
1 and 4-hydroxytamoxifen (TAM), GPER expression and ERK1/2 phosphorylation were analyzed using qRT-PCR and western blotting, respectively. Cells were treated with individual concentrations of G1 (1000 nM), G15 (1000 nM), and TAM (2000 nM), as well as combinations of these treatments (G1 + G15 , TAM + G15 , and G1 + TAM) for 24 and 48 h. The expression levels of GPER, IRE1α, miR-17-5p, TXNIP, ABCB1, and ABCC1 genes and TXNIP protein expression were evaluated. Finally, apoptosis and cell migration were examined using flow cytometry and the wound-healing assay, respectively. Findings/Results: Activating GPER with its specific agonist G1 and TAM significantly increased IRE1α levels in MDA-MB231 cells. IRE1α through splicing XBP1 led to unfolded protein response. In addition, decreased TXNIP gene and protein expression reduced apoptosis, increased migration, and upregulated the genes associated with drug resistance. Conclusion and implication: Our investigation revealed that blocking the GPER/IRE1α/TXNIP pathway in MDA-MB231 cells could enhance treatment efficacy and improve chemotherapy responsiveness. The distinct unfolded protein response observed in MDA-MB231 cells may stem from the unique characteristics of these cells, which lack receptors for estrogen, progesterone, and HER2/neu hormones, possessing only the GPER receptor (ER- /PR- /HER2- /GPER+ ). This study introduced a new pathway in TNBC cells, indicating that targeting GPER could be crucial in comprehensive therapeutic strategies in TNBC cells. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
4. GPER-mediated inhibition of astrocyte activation mitigates retinal neovascularization in oxygen-induced retinopathy mice
- Author
-
WANG Xuan, YE Jian, and LIU Wei
- Subjects
retinopathy of prematurity ,newborn mouse model of oxygen-induced retinopathy ,astrocytes ,inflammatory factors ,g protein-coupled estrogen receptor ,Medicine (General) ,R5-920 - Abstract
Objective To investigate the mechanism by which G protein-coupled estrogen receptor (GPER) reduces retinal neovascularisation in oxygen-induced retinopathy (OIR) in newborn mice. Methods A total of 42 newborn mice were randomly divided into normoxic control group (n=11), OIR group (n=11), G-1(GPER agonist) group (n=10), and solvent control group (n=10).On postnatal day 17(P17), the distribution of GPER in the retina of mice in the normoxic control group was observed by immunofluorescence staining on frozen sections of the eyeballs.The mice of the G-1 group and solvent control group were given by intraperitoneal injection 50 μg/(kg·d) G-1 or corn oil solvent from P12 to P15.Immunofluorescence staining of retinal spreads at P17 was performed to observe the expression of retinal vascular marker IB4 and astrocyte marker glia fibrilary acidic protein (GFAP).Western blotting was used to quantify the expression of retinal vascular endothelial growth factor A (VEGFA), GFAP, and inflammatory factors TNF-α, IGF-1, and IL1-β. Results GPER was present throughout the retina and co-stained with the astrocyte marker GFAP in the ganglion cell layer (GCL).Compared with the normoxic control group, the retinas of mice in the OIR group showed neovascular and avascular areas, and the expression of VEGFA and GFAP was significantly increased (P < 0.05).Compared with the solvent control group, the retinal neovascularisation was reduced in the G-1 group, and the expression levels of VEGFA, GFAP, TNF-α, IGF-1, and IL1-β proteins were decreased significantly (P < 0.05). Conclusion GPER inhibits astrocyte activity, reduces VEGFA expression and release of inflammatory factors, and then decreases retinal neovascularisation in OIR mice.
- Published
- 2024
- Full Text
- View/download PDF
5. Genistein Alleviates Dextran Sulfate Sodium-induced Ulcerative Colitis in Mice by Regulating Th17/Treg Cell Balance: Implication for the G Protein-coupled Estrogen Receptor.
- Author
-
Shu, Baolian, Wu, Youxing, Wang, Xiaoyang, Hu, Jianlan, Zhang, Danxia, Gong, Xianghao, He, Saiqi, Xie, Yangzhi, Chen, Xuan, and Gui, Ruohu
- Subjects
- *
G protein coupled receptors , *ULCERATIVE colitis , *DEXTRAN sulfate , *GENISTEIN , *REGULATORY T cells , *T helper cells , *SODIUM channels - Abstract
Objectives: This study aimed to evaluate the therapeutic effects and mechanism of genistein on dextran sulfate sodium-induced mouse model of ulcerative colitis (UC). Materials and Methods: A DSS-induced mouse model for UC was used in this study. Mice were then treated with genistein or a combination of genistein and the G protein-coupled estrogen receptor (GPER) antagonist G15. Colon length, disease activity index (DAI), spleen index (SI), histopathological alterations, and integrity of the colonic barrier were evaluated. The expression of inflammatory cytokines and antioxidant capacity were detected. Populations of T helper 17 cells (Th17) and Regulatory T cells (Treg) in the mesenteric lymph nodes (MLN) were analyzed. The expression of a transcription factor for Th17 and Treg in the colonic tissues was detected. Results: Genistein treatment significantly alleviated DSS-induced colitis, summarized as increased colonic length, decreased DAI, SI, improved histopathological alterations, and colonic barrier integrity. Genistein treatment restrained pro-inflammatory cytokines and oxidative enzyme release while promoting anti-inflammatory and anti-oxidative enzyme release. Flow cytometry indicated that genistein significantly reduced the Th17 population while boosting Treg populations. Furthermore, genistein inhibited the expression of transcription factors associated with Th17 and promoted the expression of transcription factors associated with Treg in the colonic tissue. Intriguingly, these observed effects of genistein were abolished when UC mice were treated with a combination of genistein and GPER antagonist G15. Conclusion: This study suggests that genistein is potent in protecting against DSS-induced colonic injuries by rebalancing Th17/Treg and that GPER may be a vital target for genistein-mediated immunomodulatory effects in UC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Analysis of Binding Modes between Three Perfluorosulfonates and GPER Based on Computational Simulation and Multiple Spectral Methods.
- Author
-
Liang, Wenhui, Chen, Yanting, Wei, Yuchen, Song, Zeyu, Li, Cancan, Zheng, Yanhong, and Yi, Zhongsheng
- Subjects
BINDING energy ,POLLUTANTS ,G protein coupled receptors ,LIFE sciences ,MOLECULAR dynamics ,FOURIER transform infrared spectroscopy - Abstract
Perfluorinated compounds (PFCs) belong to a significant category of global environmental pollutants. Investigating the toxicological effects of PFCs within biological systems is of critical significance in various disciplines such as life sciences, environmental science, chemistry, and ecotoxicology. In this study, under simulated human physiological conditions (pH = 7.4), a combination of multiple spectroscopic techniques and computational simulations was employed to investigate the impact of perfluorinated compounds (PFCs) on the G protein-coupled estrogen receptor (GPER). Additionally, the research focused on exploring the binding modes and toxicological mechanisms between PFCs and GPER at the molecular level. All three perfluorinated sulfonic acids (PFSAs) can induce quenching of GPER fluorescence through static quenching and non-radiative energy transfer. Steady-state fluorescence calculations at different temperatures revealed apparent binding constants in the order of 10
6 , confirming a strong binding affinity between the three PFSAs and GPER. Molecular docking studies indicated that the binding sites of PFSAs are located within the largest hydrophobic cavity in the head region of GPER, where they can engage in hydrogen bonding and hydrophobic interactions with amino acid residues within the cavity. Fourier transform infrared spectroscopy, three-dimensional fluorescence, and molecular dynamics simulations collectively indicate that proteins become more stable upon binding with small molecules. There is an overall increase in hydrophobicity, and alterations in the secondary structure of the protein are observed. This study deepens the comprehension of the effects of PFCs on the endocrine system, aiding in evaluating their potential impact on human health. It provides a basis for policy-making and environmental management while also offering insights for developing new pollution monitoring methods and drug therapies. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
7. 17β-Estradiol, through activating the G protein-coupled estrogen receptor, exacerbates the complication of benign prostatic hyperplasia in type 2 diabetes mellitus patients by inducing prostate proliferation
- Author
-
Tingting Yang, Zhen Qiu, Jiaming Shen, Yutian He, Longxiang Yin, Li Chen, Jiayu Yuan, Junjie Liu, Tao Wang, Zhenzhou Jiang, Changjiang Ying, Sitong Qian, Jinfang Song, Xiaoxing Yin, and Qian Lu
- Subjects
Sex steroid hormone homeostasis ,Proliferation ,17β-Estradiol ,G protein-coupled estrogen receptor ,T2DM complicated with BPH ,Hippo-YAP1 signaling ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Benign prostatic hyperplasia (BPH) is one of the major chronic complications of type 2 diabetes mellitus (T2DM), and sex steroid hormones are common risk factors for the occurrence of T2DM and BPH. The profiles of sex steroid hormones are simultaneously quantified by LC–MS/MS in the clinical serum of patients, including simple BPH patients, newly diagnosed T2DM patients, T2DM complicated with BPH patients and matched healthy individuals. The G protein-coupled estrogen receptor (GPER) inhibitor G15, GPER knockdown lentivirus, the YAP1 inhibitor verteporfin, YAP1 knockdown/overexpression lentivirus, targeted metabolomics analysis, and Co-IP assays are used to investigate the molecular mechanisms of the disrupted sex steroid hormones homeostasis in the pathological process of T2DM complicated with BPH. The homeostasis of sex steroid hormone is disrupted in the serum of patients, accompanying with the proliferated prostatic epithelial cells (PECs). The sex steroid hormone metabolic profiles of T2DM patients complicated with BPH have the greatest degrees of separation from those of healthy individuals. Elevated 17β-estradiol (E2) is the key contributor to the disrupted sex steroid hormone homeostasis, and is significantly positively related to the clinical characteristics of T2DM patients complicated with BPH. Activating GPER by E2 via Hippo-YAP1 signaling exacerbates high glucose (HG)-induced PECs proliferation through the formation of the YAP1-TEAD4 heterodimer. Knockdown or inhibition of GPER-mediated Hippo-YAP1 signaling suppresses PECs proliferation in HG and E2 co-treated BPH-1 cells. The anti-proliferative effects of verteporfin, an inhibitor of YAP1, are blocked by YAP1 overexpression in HG and E2 co-treated BPH-1 cells. Inactivating E2/GPER/Hippo/YAP1 signaling may be effective at delaying the progression of T2DM complicated with BPH by inhibiting PECs proliferation.
- Published
- 2024
- Full Text
- View/download PDF
8. The G protein‐coupled estrogen receptor of the trigeminal ganglion regulates acute and chronic itch in mice.
- Author
-
Li, Jun, Gao, Po, Zhang, Siyu, Lin, Xiaoqi, Chen, Junhui, Zhang, Song, Jiao, Yingfu, Yu, Weifeng, Xia, Xiaoqiong, and Yang, Liqun
- Subjects
- *
ITCHING , *G protein coupled receptors , *SENSORY neurons , *GANGLIA , *PAIN perception - Abstract
Aims: Itch is an unpleasant sensation that severely impacts the patient's quality of life. Recent studies revealed that the G protein‐coupled estrogen receptor (GPER) may play a crucial role in the regulation of pain and itch perception. However, the contribution of the GPER in primary sensory neurons to the regulation of itch perception remains elusive. This study aimed to investigate whether and how the GPER participates in the regulation of itch perception in the trigeminal ganglion (TG). Methods and Results: Immunofluorescence staining results showed that GPER‐positive (GPER+) neurons of the TG were activated in both acute and chronic itch. Behavioral data indicated that the chemogenetic activation of GPER+ neurons of the TG of Gper‐Cre mice abrogated scratching behaviors evoked by acute and chronic itch. Conversely, the chemogenetic inhibition of GPER+ neurons resulted in increased itch responses. Furthermore, the GPER expression and function were both upregulated in the TG of the dry skin‐induced chronic itch mouse model. Pharmacological inhibition of GPER (or Gper deficiency) markedly increased acute and chronic itch‐related scratching behaviors in mouse. Calcium imaging assays further revealed that Gper deficiency in TG neurons led to a marked increase in the calcium responses evoked by agonists of the transient receptor potential ankyrin A1 (TRPA1) and transient receptor potential vanilloid V1 (TRPV1). Conclusion: Our findings demonstrated that the GPER of TG neurons is involved in the regulation of acute and chronic itch perception, by modulating the function of TRPA1 and TRPV1. This study provides new insights into peripheral itch sensory signal processing mechanisms and offers new targets for future clinical antipruritic therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Role of G-protein-coupled estrogen receptor in the pathogenesis of chronic asthma.
- Author
-
Itoga, Masamichi, Ishioka, Yoshiko, Makiguchi, Tomonori, Tanaka, Hisashi, Taima, Kageaki, Saito, Norihiro, Tomita, Hirofumi, and Tasaka, Sadatomo
- Subjects
- *
ESTROGEN receptors , *G protein coupled receptors , *ASTHMA , *REGULATORY T cells , *CELL membranes , *WHEEZE - Abstract
• GPER-specific agonist (G-1) reduces number of inflammatory cells in BALF. • G-1 administration increases anti-inflammatory cytokines (IL-10, TGF-β). • G-1 administration suppresses allergic airway inflammation in chronic asthma. • GPER may be a potential therapeutic target for the treatment of asthma. G protein-coupled estrogen receptor (GPER) is an estrogen receptor located on the plasma membrane. We previously reported that the administration of G-1, a GPER-specific agonist, suppressed development of acute ovalbumin (OVA)-induced asthma in a mouse model. Herein, we evaluate the involvement of GPER in a mouse model of chronic OVA asthma. G-1 or saline was administered subcutaneously to BALB/c mice with chronic OVA asthma, and pathological and immunological evaluation was performed. In addition, Foxp3-expressing CD4-positive T-cells in the spleen and ILC2 in the lungs were measured using flow cytometry. We observed a significant decrease in the number of inflammatory cells in the bronchoalveolar lavage fluid (BALF) in the G-1 treated group. In the airways, inflammatory cell accumulation, Th2 cytokines (IL-4, IL-5, IL-13, and eotaxin) and epithelial cytokine TSLP were suppressed, while in the BALF, anti-inflammatory cytokines (IL-10 and TGF-β) were increased. Furthermore, in splenic mononuclear cells, Foxp3-expressing CD4-positive T-cells were increased in the G-1 group, whereas treatment with G-1 did not change the percentage of ILC2 in the lungs. G-1 administration suppressed allergic airway inflammation in mice with chronic OVA asthma. GPER may be a potential therapeutic target for chronic allergic asthma. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Current progress and prospects for G protein-coupled estrogen receptor in triple-negative breast cancer
- Author
-
Duo Zhang, Hong Chen, Jinpeng Wang, Jiale Ji, Murshid Imam, Zhijie Zhang, and Shunchao Yan
- Subjects
G protein-coupled estrogen receptor ,triple-negative breast cancer ,tumor development ,mechanism ,prognostic ,Biology (General) ,QH301-705.5 - Abstract
Triple-negative breast cancer (TNBC) is a biologically and clinically heterogeneous disease. The G protein-coupled estrogen receptor (GPER) plays a crucial role in mediating the effect of estrogen and estrogen-like compounds in TNBC cells. Compared with other subtypes, GPER has a higher expression in TNBC. The GPER mechanisms have been thoroughly characterized and analyzed in estrogen receptor α (ERα) positive breast cancer, but not in TNBC. Our previous work revealed that a higher expression of GPER mRNA indicates a better prognosis for ERα-positive breast cancer; however, its effects in TNBC differ. Whether GPER could serve as a predictive prognostic marker or therapeutic target for TNBC remains unclear. In this review, we provide a detailed introduction to the subcellular localization of GPER, the different effects of various ligands, and the interactions between GPER and closely associated factors in TNBC. We focused on the internal molecular mechanisms specific to TNBC and thoroughly explored the role of GPER in promoting tumor development. We also discussed the interaction of GPER with specific cytokines and chemokines, and the relationship between GPER and immune evasion. Additionally, we discussed the feasibility of using GPER as a therapeutic target in the context of existing studies. This comprehensive review highlights the effects of GPER on TNBC, providing a framework and directions for future research.
- Published
- 2024
- Full Text
- View/download PDF
11. G Protein-Coupled Estrogen Receptor (GPER) and ERs Are Modulated in the Testis–Epididymal Complex in the Normal and Cryptorchid Dog.
- Author
-
Liguori, Giovanna, Tafuri, Simona, Pelagalli, Alessandra, Ali', Sabrina, Russo, Marco, Mirabella, Nicola, and Squillacioti, Caterina
- Subjects
G protein coupled receptors ,ESTROGEN receptors ,GENE expression ,GENITALIA ,DOGS ,ANDROGEN receptors - Abstract
Simple Summary: The testicular function as well as the maintenance and control of spermatogenesis are regulated by a delicate balance between androgens and estrogens. Cryptorchidism, largely diffused in canine species, is a congenital abnormality of the genitourinary tract, due to the failure to descend by one or both testes into the scrotal sac. In consideration of the possible role of estrogenic molecules affecting testicular descent, the aim of this study was to determine the distribution and expression (proteins and relative mRNAs levels) of two nuclear estrogen receptors (ERs), ER-alpha and ER-beta and a trans-membrane G protein-coupled estrogen receptor (GPER), in the testis–epididymal complex of the dog. In addition, in these tissues the expression level of two proteins as SOD1 and Nrf2 normally associated with oxidative stress was investigated to evaluate possible relation with ERs. Collectively, the results obtained by using Immunohistochemistry, Western blot and qRT-PCR showed changes in the distribution and expression of the GPER and ERs between the normal and cryptorchid dog. In detail, an upregulation of GPER and ER-alpha and a downregulation of ER-beta in the canine cryptorchid reproductive tract was observed in association with a modulation of SOD1 and Nrf2 expression. There is growing evidence by the literature that the unbalance between androgens and estrogens is a relevant condition associated with a common canine reproductive disorder known as cryptorchidism. The role of estrogens in regulating testicular cell function and reproductive events is supposedly due to the wide expression of two nuclear estrogen receptors (ERs), ER-alpha and ER-beta and a trans-membrane G protein-coupled estrogen receptor (GPER) in the testis. In this study, immunohistochemistry, Western blotting and qRT-PCR were used to assess the distribution and expression of GPER in the testis–epididymal complex in the normal and cryptorchid dog. ER-alpha and ER-beta were also evaluated to better characterize the relative abundances of all three receptors. In addition, in these tissues, the expression level of two proteins as SOD1 and Nrf2 normally associated with oxidative stress was investigated to evaluate a possible relationship with ERs. Our data revealed changes in the distribution and expression of the GPER between the normal and cryptorchid dog. In particular, dogs affected by cryptorchidism showed an upregulation of GPER at level of the examined reproductive tract. Also considering the obtained result of a modulation of SOD1 and Nrf2 expression, we could hypothesize the involvement of GPER in the cryptorchid condition. Further studies are, however, necessary to characterize the role of GPER and its specific signaling mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Genistein alleviates chronic heat stress-induced lipid metabolism disorder and mitochondrial energetic dysfunction by activating the GPR30-AMPK-PGC-1α signaling pathways in the livers of broiler chickens
- Author
-
Longlong Li, Ze Lu, Yulei Wang, Ying Yang, Huihui Wang, and Haitian Ma
- Subjects
genistein ,heat stress ,lipid metabolism disorder ,G protein-coupled estrogen receptor ,broiler chicken ,Animal culture ,SF1-1100 - Abstract
ABSTRACT: The objective of this study was to investigate the preventive effects and mechanisms of genistein (GEN) on production performance and metabolic disorders in broilers under chronic heat stress (HS). A total of 120 male 3-wk-old Ross broilers were randomly assigned to 5 groups: a thermoneutral zone (TN) group maintained at normal temperature (21°C ± 1°C daily), an HS group subjected to cyclic high temperature (32°C ± 1°C for 8 h daily), and 3 groups exposed to HS with varying doses of GEN (50, 100, or 150 mg/kg diet). The experimental period lasted for 3 wk. Here, HS led to a decline in growth performance parameters and hormone secretion disorders (P < 0.05), which were improved by 100 and 150 mg/kg GEN treatment (P < 0.05). Moreover, the HS-induced increases in the liver index (P < 0.01) and abdominal fat rate (P < 0.05) were attenuated by 150 mg/kg GEN (P < 0.05). The HS-induced excessive lipid accumulation in the liver and serum (P < 0.01) was ameliorated after 100 and 150 mg/kg GEN treatment (P < 0.05). Furthermore, the HS-induced decreases in lipolysis-related mRNA levels and increases in lipid synthesis-related mRNA levels in the liver (P < 0.01) were effectively blunted after 100 and 150 mg/kg GEN treatment (P < 0.05). Importantly, the HS-stimulated hepatic mitochondrial energetic dysfunction and decreases in the mRNA or protein levels of peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α), nuclear respiratory factor 1, and mitochondrial transcription factor A in the liver were ameliorated by 150 mg/kg GEN (P < 0.05). Moreover, 50 to 150 mg/kg GEN treatment resulted in a significant increase in the mRNA or protein levels of G protein-coupled estrogen receptor (GPR30), AMP-activated protein kinase (AMPK) α1, phosphorylated AMPKα, and phosphorylated acetyl-CoA carboxylase α. Collectively, GEN alleviated metabolic disorders and hepatic mitochondrial energetic dysfunction under HS, possibly through the activation of GPR30-AMPM-PGC-1α pathways. These data provide a sufficient basis for GEN as an additive to alleviate HS in broilers.
- Published
- 2024
- Full Text
- View/download PDF
13. GPER 调控缺血性脑卒中相关分子机制的 研究进展.
- Author
-
张梦杰, 方世才, and 黄志华
- Subjects
- *
G protein coupled receptors , *CEREBRAL ischemia , *ISCHEMIC stroke , *CEREBROVASCULAR disease , *DRUG development - Abstract
As an acute cerebrovascular disease, ischemic stroke is one of the leading causes of death and disability worldwide. It involves in fast developing and irreversible brain damage. Effective treatment is an important strategy to save patients' life and maintain their health. G protein-coupled estrogen receptor (GPER) mediates a rapid effect of estrogen, which can protect the nerves after ischemic brain injury. This review aims to summarize the multiple pathophysiological processes and signaling pathways of the involvement of GPER in cerebral ischemia, so as to provide theoretical reference for further study of the pathogenesis of ischemic stroke and new ideas for the development of drug targets. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. Role of Estrogen Receptor β, G-Protein Coupled Estrogen Receptor and Estrogen-Related Receptors in Endometrial and Ovarian Cancer.
- Author
-
Schüler-Toprak, Susanne, Skrzypczak, Maciej, Gründker, Carsten, Ortmann, Olaf, and Treeck, Oliver
- Subjects
- *
DISEASE progression , *PROTEINS , *OVARIAN tumors , *CELL receptors , *ESTROGEN receptors , *ENDOMETRIAL tumors , *CELL proliferation - Abstract
Simple Summary: Despite new therapeutic approaches, ovarian cancer is still the most lethal gynecological cancer that is mainly diagnosed in its advanced stages. Contrarily, endometrial cancer is often detected in its early stages. However, in the cases of recurrence or advanced disease, treatment options are still limited. Both the ovary and endometrium are affected by estrogens and their receptors. The well-known estrogen receptor α (ERα) mediates estrogen effects such as the activation of cell proliferation. In contrast, the functions of the later discovered ERs, ERβ and GPER1, and of estrogen-related receptors (ERRs), are less understood. Increasing evidence suggests them to be involved in tumor development, progression, and metastasis. This article provides a summary and update of the current findings on the role of these receptors in ovarian and endometrial cancer to show at which points further research is reasonable and might change the future of their treatment. Ovarian and endometrial cancers are affected by estrogens and their receptors. It has been long known that in different types of cancers, estrogens activate tumor cell proliferation via estrogen receptor α (ERα). In contrast, the role of ERs discovered later, including ERβ and G-protein-coupled ER (GPER1), in cancer is less well understood, but the current state of knowledge indicates them to have a considerable impact on both cancer development and progression. Moreover, estrogen related receptors (ERRs) have been reported to affect pathobiology of many tumor types. This article provides a summary and update of the current findings on the role of ERβ, GPER1, and ERRs in ovarian and endometrial cancer. For this purpose, original research articles on the role of ERβ, GPER1, and ERRs in ovarian and endometrial cancers listed in the PubMed database have been reviewed. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. Analysis of Binding Modes between Three Perfluorosulfonates and GPER Based on Computational Simulation and Multiple Spectral Methods
- Author
-
Wenhui Liang, Yanting Chen, Yuchen Wei, Zeyu Song, Cancan Li, Yanhong Zheng, and Zhongsheng Yi
- Subjects
G protein-coupled estrogen receptor ,perfluorochemical ,spectroscopy ,molecular dynamics simulation ,molecular docking ,Chemical technology ,TP1-1185 - Abstract
Perfluorinated compounds (PFCs) belong to a significant category of global environmental pollutants. Investigating the toxicological effects of PFCs within biological systems is of critical significance in various disciplines such as life sciences, environmental science, chemistry, and ecotoxicology. In this study, under simulated human physiological conditions (pH = 7.4), a combination of multiple spectroscopic techniques and computational simulations was employed to investigate the impact of perfluorinated compounds (PFCs) on the G protein-coupled estrogen receptor (GPER). Additionally, the research focused on exploring the binding modes and toxicological mechanisms between PFCs and GPER at the molecular level. All three perfluorinated sulfonic acids (PFSAs) can induce quenching of GPER fluorescence through static quenching and non-radiative energy transfer. Steady-state fluorescence calculations at different temperatures revealed apparent binding constants in the order of 106, confirming a strong binding affinity between the three PFSAs and GPER. Molecular docking studies indicated that the binding sites of PFSAs are located within the largest hydrophobic cavity in the head region of GPER, where they can engage in hydrogen bonding and hydrophobic interactions with amino acid residues within the cavity. Fourier transform infrared spectroscopy, three-dimensional fluorescence, and molecular dynamics simulations collectively indicate that proteins become more stable upon binding with small molecules. There is an overall increase in hydrophobicity, and alterations in the secondary structure of the protein are observed. This study deepens the comprehension of the effects of PFCs on the endocrine system, aiding in evaluating their potential impact on human health. It provides a basis for policy-making and environmental management while also offering insights for developing new pollution monitoring methods and drug therapies.
- Published
- 2024
- Full Text
- View/download PDF
16. GPER: a key player in prostate cancer and its interactions with signaling pathways.
- Author
-
Rico-Fuentes, Cecilia, López-Pulido, Edgar I., Moreno-Ortíz, José M., Villegas-Pineda, Julio C., Pereira-Suárez, Ana L., Ramírez-de-Arellano, Adrián, and Sierra-Diaz, Erick
- Abstract
This review explores the role of G protein-coupled estrogen receptor (GPER) and its interaction with the mitogen-activated protein kinase (MAPK)/extracellular regulated protein kinase (ERK), phosphatidylinositol 3-kinase (PI3K)/AKT, and hedgehog (Hh) signaling pathways in prostate cancer (PCa) progression. PCa is a significant health problem globally, and GPER has been shown to play a crucial role in the regulation of cell proliferation and survival in PCa cells. The MAPK/ERK pathway, activated by GPER, is involved in cell growth and differentiation, while the PI3K/AKT pathway influences cell cycle progression and survival. The Hh pathway is important for prostate development and has been implicated in the formation and progression of PCa. However, the specific contributions of GLI1 and GLI2 factors and their correlation with Gleason scores and GPER expression in PCa are not yet fully understood. Understanding the interactions between GPER and these signaling pathways provides valuable insights for the development of targeted therapies for PCa. Further research is needed to explore the potential of GLI1 and GLI2 factors and their relationship with GPER expression in PCa progression and metastasis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Estrogen receptors as potential therapeutic target in endometrial cancer.
- Author
-
Guha, Payel, Sen, Koushik, Chowdhury, Piyali, and Mukherjee, Dilip
- Abstract
Endometrial cancer (EC) is one of the most common gynecological carcinomas in both developed and developing countries. Majority of the gynecological malignancies are hormonally driven where estrogen signaling acts as an oncogenic signal. Estrogen's effects are mediated via classical nuclear estrogen receptors; estrogen receptor alpha and beta (ERα and ERβ) and a trans-membrane G protein-coupled estrogen receptor (GPR30 and GPER). ERs and GPER through ligand binding triggers multiple downstream signaling pathways causing cell cycle regulation, cell differentiation, migration, and apoptosis in various tissues including endometrium. Although the molecular aspect of estrogen function in ER-mediated signaling is now partly understood, the same is not true for GPER-mediated signaling in endometrial malignancies. Understanding the physiological roles of ERα and GPER in EC biology therefore leads to the identification of some novel therapeutic targets. Here we review the effect of estrogen signaling through ERα-and GPER in EC, major types, and some affordable treatment approaches for endometrial tumor patients which has interesting implications in understanding uterine cancer progression. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
18. G Protein-Coupled Estrogen Receptor (GPER) and ERs Are Modulated in the Testis–Epididymal Complex in the Normal and Cryptorchid Dog
- Author
-
Giovanna Liguori, Simona Tafuri, Alessandra Pelagalli, Sabrina Ali’, Marco Russo, Nicola Mirabella, and Caterina Squillacioti
- Subjects
estrogen receptors ,G protein-coupled estrogen receptor ,dog male genital tract ,cryptorchidism ,oxidative stress ,Veterinary medicine ,SF600-1100 - Abstract
There is growing evidence by the literature that the unbalance between androgens and estrogens is a relevant condition associated with a common canine reproductive disorder known as cryptorchidism. The role of estrogens in regulating testicular cell function and reproductive events is supposedly due to the wide expression of two nuclear estrogen receptors (ERs), ER-alpha and ER-beta and a trans-membrane G protein-coupled estrogen receptor (GPER) in the testis. In this study, immunohistochemistry, Western blotting and qRT-PCR were used to assess the distribution and expression of GPER in the testis–epididymal complex in the normal and cryptorchid dog. ER-alpha and ER-beta were also evaluated to better characterize the relative abundances of all three receptors. In addition, in these tissues, the expression level of two proteins as SOD1 and Nrf2 normally associated with oxidative stress was investigated to evaluate a possible relationship with ERs. Our data revealed changes in the distribution and expression of the GPER between the normal and cryptorchid dog. In particular, dogs affected by cryptorchidism showed an upregulation of GPER at level of the examined reproductive tract. Also considering the obtained result of a modulation of SOD1 and Nrf2 expression, we could hypothesize the involvement of GPER in the cryptorchid condition. Further studies are, however, necessary to characterize the role of GPER and its specific signaling mechanisms.
- Published
- 2024
- Full Text
- View/download PDF
19. The effects of GPER on age-associated memory impairment induced by decreased estrogen levels
- Author
-
Wenyu Luo, Yudie Yan, Yunpeng Cao, Yanbo Zhang, and Zhen Zhang
- Subjects
G protein-coupled estrogen receptor ,estrogen ,aging ,memory impairment ,neurogenesis ,synaptic plasticity ,Biology (General) ,QH301-705.5 - Abstract
Estrogen, as a pleiotropic endocrine hormone, not only regulates the physiological functions of peripheral tissues but also exerts vital neuroregulatory effects in the central nervous system (CNS), such as the development of neurons and the formation of neural network connections, wherein rapid estrogen-mediated reactions positively stimulate spinogenesis and regulate synaptic plasticity and synaptic transmission to facilitate cognitive and memory performance. These fast non-genomic effects can be initiated by membrane-bound estrogen receptors (ERs), three best known of which are ERα, ERβ, and G protein-coupled estrogen receptor (GPER). To date, the effects of ERα and ERβ have been well studied in age-associated memory impairment, whereas there is still a lack of attention to the role of GPER in age-associated memory impairment, and there are still disputes about whether GPER indeed functions as an ER to enhance learning and memory. In this review, we provide a systematic overview of the role of GPER in age-associated memory impairment based on its expression, distribution, and signaling pathways, which might bring some inspiration for translational drugs targeting GPER for age-related diseases and update knowledge on the role of estrogen and its receptor system in the brain.
- Published
- 2023
- Full Text
- View/download PDF
20. Estrogen deficiency promotes neurodegeneration in female hemi-parkinsonian mice: The role of regulatory T cells.
- Author
-
Xie Y, Chen J, Xu S, and Cheng O
- Abstract
Background: Circulating levels of the female hormone estrogen has been associated with the development of Parkinson's disease (PD), although the underlying mechanism remains unclear. Immune homeostasis mediated by peripheral regulatory T cells (Treg) is a crucial factor in PD. The aim of this study was to explore the effects of estrogen deficiency on neuroinflammation and neurodegeneration in a rodent model of PD, with particular reference to Treg., Methods: Estrogen deficiency was established in a mouse model by bilateral ovariectomy (OVX). PD was modeled by the injection of LPS into the striatum. Motor performance was assessed in each experimental group. Dopaminergic degeneration was evaluated using tyrosine-hydroxylase (Th) immunohistochemical staining of the substantia nigra (SN) and striatum. Dopamine and dopamine metabolite levels in the striatum were also evaluated, together with the infiltration of CD4 T cells into the SN. Neuroinflammation was assessed by evaluating the mRNA level of microglial and M1/M2 phenotype markers, as well as the abundance of pro-inflammatory cytokines in the midbrain. The frequency of peripheral Treg cells was evaluated using flow cytometry., Results: OVX prior to LPS injection markedly aggravated neurodegeneration, neuroinflammation, motor performance, and CD4 T cell infiltration compared to the LPS-only group. Estradiol treatment or activation of the G protein-coupled estrogen receptor (GPER) in OVX mice prior to LPS injection induced functional improvement and reduced the levels of neurodegeneration, neuroinflammation, and CD4 T cell infiltration. OVX prior to LPS injection also led to a decreased frequency of Treg compared to the LPS-only group. Moreover, estradiol treatment or GPER activation of OVX mice prior to LPS injection significantly increased the Treg frequency. Antibody-mediated depletion of Treg after GPER activation counteracted the ability of GPER to alleviate neurodegeneration, CD4 T cell infiltration, the release of pro-inflammatory cytokines, and motor performance in the PD model., Conclusion: Estrogen deficiency can disrupt Treg-mediated immune homeostasis and thus further aggravate microglial inflammation and dopaminergic degeneration in PD model. The effects of estrogen on Treg may be partially mediated by GPER signaling, and thus GPER is a promising target for PD, especially in estrogen-deficient women., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2025. Published by Elsevier B.V.)
- Published
- 2025
- Full Text
- View/download PDF
21. β-Blockers are associated with increased B-type natriuretic peptide levels differently in men and women in heart failure with preserved ejection fraction.
- Author
-
Eisaku Harada, Yuji Mizuno, Masanobu Ishii, Toshifumi Ishida, Toshihiro Yamada, Fumihito Kugimiya, and Hirofumi Yasue
- Subjects
- *
VENTRICULAR ejection fraction , *HEART failure , *SYSTOLIC blood pressure , *LOGISTIC regression analysis , *PEPTIDES , *ALDOSTERONE antagonists , *BRAIN natriuretic factor - Abstract
β-Blocker (BB) use is a mainstay for the treatment of heart failure (HF) with reduced ejection fraction (HFrEF), whereas its efficacy for heart failure with preserved ejection fraction (HFpEF) remains controversial. Women outnumber men in HFpEF, whereas men outnumber women in HFrEF. Plasma B-type natriuretic peptide (BNP) is established as a biomarker for HF. We examined whether BB use is associated with plasma BNP levels differently in men and women with HFpEF. The study subjects comprised 721 patients with HFpEF [left ventricular ejection fraction (LVEF) ≤ 50%] (184 men, mean age 78.2± 9.2 yr and 537 women, mean age 83.1 ±8.8 yr), 179 on BB (66 men and 113 women) and 542 no BB (118 men and 424 women), 583 in sinus rhythm (SR) and 138 in atrial fibrillation (AF). A multivariable logistic regression test was used. Plasma BNP levels were higher (P = 0.0005), systolic blood pressure and LVEF lower (P = 0.0003, and P = 0.0059, respectively) on BBs than on no BBs in women, whereas in men, plasma BNP levels, systolic blood pressure, and LVEF were not altered significantly (P = 0.0849, P = 0.9129, and P = 0.4718, respectively) on BBs compared with no BBs in patients with SR. Multivariable logistic regression analysis revealed that BB use and women were a positive and a negative predictor for high BNP levels (P = 0.003 and P = 0.032, respectively) in SR but not in AF. BB use was associated with high-plasma BNP levels and lower LVEF in women but not in men with HFpEF and SR, suggesting that the pathogenesis and treatment of HFpEF may differ in men and women in SR. NEW & NOTEWORTHY Pathogenesis and treatment for heart failure with preserved ejection fraction (HFpEF) may differ in men and women in sinus rhythm (SR). [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. 17β-Estradiol, through activating the G protein-coupled estrogen receptor, exacerbates the complication of benign prostatic hyperplasia in type 2 diabetes mellitus patients by inducing prostate proliferation.
- Author
-
Yang, Tingting, Qiu, Zhen, Shen, Jiaming, He, Yutian, Yin, Longxiang, Chen, Li, Yuan, Jiayu, Liu, Junjie, Wang, Tao, Jiang, Zhenzhou, Ying, Changjiang, Qian, Sitong, Song, Jinfang, Yin, Xiaoxing, and Lu, Qian
- Subjects
TYPE 2 diabetes ,SEX hormones ,G protein coupled receptors ,BENIGN prostatic hyperplasia ,YAP signaling proteins - Abstract
Benign prostatic hyperplasia (BPH) is one of the major chronic complications of type 2 diabetes mellitus (T2DM), and sex steroid hormones are common risk factors for the occurrence of T2DM and BPH. The profiles of sex steroid hormones are simultaneously quantified by LC-MS/MS in the clinical serum of patients, including simple BPH patients, newly diagnosed T2DM patients, T2DM complicated with BPH patients and matched healthy individuals. The G protein-coupled estrogen receptor (GPER) inhibitor G15, GPER knockdown lentivirus, the YAP1 inhibitor verteporfin, YAP1 knockdown/overexpression lentivirus, targeted metabolomics analysis, and Co-IP assays are used to investigate the molecular mechanisms of the disrupted sex steroid hormones homeostasis in the pathological process of T2DM complicated with BPH. The homeostasis of sex steroid hormone is disrupted in the serum of patients, accompanying with the proliferated prostatic epithelial cells (PECs). The sex steroid hormone metabolic profiles of T2DM patients complicated with BPH have the greatest degrees of separation from those of healthy individuals. Elevated 17β-estradiol (E2) is the key contributor to the disrupted sex steroid hormone homeostasis, and is significantly positively related to the clinical characteristics of T2DM patients complicated with BPH. Activating GPER by E2 via Hippo-YAP1 signaling exacerbates high glucose (HG)-induced PECs proliferation through the formation of the YAP1-TEAD4 heterodimer. Knockdown or inhibition of GPER-mediated Hippo-YAP1 signaling suppresses PECs proliferation in HG and E2 co-treated BPH-1 cells. The anti-proliferative effects of verteporfin, an inhibitor of YAP1, are blocked by YAP1 overexpression in HG and E2 co-treated BPH-1 cells. Inactivating E2/GPER/Hippo/YAP1 signaling may be effective at delaying the progression of T2DM complicated with BPH by inhibiting PECs proliferation. [Display omitted] • Sex steroid hormone compositions were dysregulated in the serum of BPH patients complicated with T2DM. • E2 exacerbated HG-induced PECs proliferation via activating GPER. • GPER knockdown inhibited the activation of Hippo-YAP1 signaling in HG and E2 co-cultured BPH-1 cells. • Knockdown or inhibition of GPER-mediated Hippo-YAP1 signaling inhibited PECs proliferation in vitro. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Exploring the physiological role of the G protein-coupled estrogen receptor (GPER) and its associations with human diseases.
- Author
-
Żabińska, Magdalena, Wiśniewska, Karolina, Węgrzyn, Grzegorz, and Pierzynowska, Karolina
- Subjects
- *
G protein coupled receptors , *ESTROGEN receptors , *CENTRAL nervous system , *HUMAN physiology , *NERVOUS system - Abstract
Estrogen is a group of hormones that collaborate with the nervous system to impact the overall well-being of all genders. It influences many processes, including those occurring in the central nervous system, affecting learning and memory, and playing roles in neurodegenerative diseases and mental disorders. The hormone's action is mediated by specific receptors. Significant roles of classical estrogen receptors, ERα and ERβ, in various diseases were known since many years, but after identifying a structurally and locationally distinct receptor, the G protein-coupled estrogen receptor (GPER), its role in human physiology and pathophysiology was investigated. This review compiles GPER-related information, highlighting its impact on homeostasis and diseases, while putting special attention on functions and dysfunctions of this receptor in neurobiology and biobehavioral processes. Understanding the receptor modulation possibilities is essential for therapy, as disruptions in receptors can lead to diseases or disorders, irrespective of correct estrogen levels. We conclude that studies on the GPER receptor have the potential to develop therapies that regulate estrogen and positively impact human health. [Display omitted] • GPER functions in physiology and disease are reviewed. • Recent data on GPER association with neurological disorders are discussed. • Agonistic and antagonistic compounds for GPER are summarized and characterized. • Deeper GPER receptor analysis for diagnosing estrogen-related diseases is recommended. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. A novel GPER antagonist protects against the formation of estrogen-induced cholesterol gallstones in female mice
- Author
-
Chelsea DeLeon, Helen H. Wang, Joseph Gunn, McKenna Wilhelm, Aidan Cole, Stacy Arnett, David Q-H. Wang, and Christopher K. Arnatt
- Subjects
bile ,bile salts ,crystallization ,Lith ,mucin ,G protein-coupled estrogen receptor ,Biochemistry ,QD415-436 - Abstract
Many clinical studies and epidemiological investigations have clearly demonstrated that women are twice as likely to develop cholesterol gallstones as men, and oral contraceptives and other estrogen therapies dramatically increase that risk. Further, animal studies have revealed that estrogen promotes cholesterol gallstone formation through the estrogen receptor (ER) α, but not ERβ, pathway. More importantly, some genetic and pathophysiological studies have found that G protein-coupled estrogen receptor (GPER) 1 is a new gallstone gene, Lith18, on chromosome 5 in mice and produces additional lithogenic actions, working independently of ERα, to markedly increase cholelithogenesis in female mice. Based on computational modeling of GPER, a novel series of GPER-selective antagonists were designed, synthesized, and subsequently assessed for their therapeutic effects via calcium mobilization, cAMP, and ERα and ERβ fluorescence polarization binding assays. From this series of compounds, one new compound, 2-cyclohexyl-4-isopropyl-N-(4-methoxybenzyl)aniline (CIMBA), exhibits superior antagonism and selectivity exclusively for GPER. Furthermore, CIMBA reduces the formation of 17β-estradiol-induced gallstones in a dose-dependent manner in ovariectomized mice fed a lithogenic diet for 8 weeks. At 32 μg/day/kg CIMBA, no gallstones are found, even in ovariectomized ERα (−/−) mice treated with 6 μg/day 17β-estradiol and fed the lithogenic diet for 8 weeks. In conclusion, CIMBA treatment protects against the formation of estrogen-induced cholesterol gallstones by inhibiting the GPER signaling pathway in female mice. CIMBA may thus be a new agent for effectively treating cholesterol gallstone disease in women.
- Published
- 2020
- Full Text
- View/download PDF
25. Chlorobisphenol A activated kisspeptin/GPR54-GnRH neuroendocrine signals through ERα and GPER pathway in neuronal GT1-7 cells
- Author
-
Bingli Lei, Lanbing Xu, Yaoyao Huang, Yun Liu, Mengjie Yu, and Qianqian Tang
- Subjects
Chlorobisphenol A ,GT1–7 neuronal cells ,Kisspeptin/GPR54-GnRH neuroendocrine system ,G protein-coupled estrogen receptor ,Molecular mechanisms ,Environmental pollution ,TD172-193.5 ,Environmental sciences ,GE1-350 - Abstract
Chlorobisphenol A (ClxBPA) is a kind of novel estrogenic compounds. The present study aims to investigate the effects of three ClxBPA compounds on the kisspeptin/G protein-coupled receptor 54 (GPR54, also named KissR1)-gonadotropin-releasing hormone (GnRH) (KGG) system in neuronal GT1–7 cells with mechanistic insights by estrogen receptor signaling pathways. The study demonstrated that low-concentration ClxBPA induced the cell proliferation, promoted GnRH secretion, upregulated the expression of KGG neuroendocrine signal-related proteins (KissR1, GnRH1 and kisspeptin) and genes including Kiss1, GnRH1, KissR1, luteinizing hormone receptor (Lhr) and follicle-stimulating hormone receptor (Fshr) in GT1–7 cells. Additionally, ClxBPA activated nuclear estrogen receptor alpha (ERα) and member estrogen receptor G protein-coupled estrogen receptor (GPER)-regulated phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) and extracellular signal-regulated kinase (Erk1/2) signaling pathways. Pretreatment of GT1–7 cells with GPER inhibitor G15 and ERα inhibitor ICI reduced the expression of KissR1, GnRH1 and kisspeptin proteins, attenuated mRNA levels of Kiss1, GnRH1, KissR1, Fshr and Lhr genes, and decreased ClxBPA-induced GT1–7 cell proliferation. The results suggested that ClxBPA activated the KGG neuroendocrine signals and induced the proliferation of GT1–7 cells via ERα and GPER signaling pathways. This study provides a new perspective to explore the neuroendocrine toxicity mechanism of ClxBPA. Capsule: ClxBPA activated KGG neuroendocrine signaling pathway via ERα and GPER and induced the proliferation of GT1–7 cells.
- Published
- 2022
- Full Text
- View/download PDF
26. G protein-coupled estrogen receptor signaling dependent epidermal growth-like factor expression is required for NPR2 inhibition and meiotic resumption in goat oocytes.
- Author
-
Lu, Sihai, Zhang, Hui, Tang, Yaju, Xu, Rui, Liu, Jie, Yao, Ru, Wei, Juncai, Li, Chan, Zhao, Xiaoe, Wei, Qiang, and Ma, Baohua
- Subjects
- *
G protein coupled receptors , *ESTROGEN receptors , *CYCLIC guanylic acid , *CELLULAR signal transduction , *EPIDERMAL growth factor receptors , *PEPTIDE receptors - Abstract
G protein-coupled estrogen receptor (GPER), which is different from traditional estrogen nuclear receptors (ERs), mediates the rapid transduction of nongenomic signals in cells, and works by regulating transcription and intracellular second messengers. Studies have shown that GPER may regulate oocyte maturation, but the relevant mechanism is not entirely clear. Here, goat cumulus-oocyte complexes (COCs) were used as a model to explore the regulation and mechanism of GPER on oocyte maturation. Our study showed that 17β-estradiol (E 2) significantly reduced cyclic guanosine monophosphate (cGMP) synthesis in COCs and accelerated the meiotic resumption of goat oocytes via GPER. Further investigation found that GPER mediated the downregulation of natriuretic peptide receptor 2 (NPR2) protein expression in goat cumulus cells by E 2. In addition, we found that E 2 significantly upregulated the mRNA levels of epidermal growth (EGF)-like factors in goat cumulus cells through GPER, and activated the downstream EGF receptor (EGFR) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathways. Both AG1478 (EGFR inhibitor) and U0126 (ERK1/2 inhibitor) abolished the inhibitory effect of E 2 on the protein expression of NPR2. These results indicate that, through GPER, E 2 upregulates the mRNA levels of EGF-like factors in goat cumulus cells and activates the downstream EGF signaling network to suppress the expression of NPR2 protein, which results in a decrease in cGMP synthesis and acceleration of meiotic resumption in goat oocytes. • 17 β-Estradiol accelerated the meiotic resumption of oocytes via GPER. • 17 β-Estradiol downregulated the protein expression of NPR2 and cGMP synthesis in goat cumulus cells through GPER. • GPER mediated the upregulation of EGF-like factor and activation of the EGFR signaling pathway induced by 17 β-estradiol. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
27. G protein‐coupled estrogen receptor in the rostral ventromedial medulla contributes to the chronification of postoperative pain.
- Author
-
Xu, Jia‐Jia, Gao, Po, Wu, Ying, Yin, Su‐Qing, Zhu, Ling, Xu, Sai‐Hong, Tang, Dan, Cheung, Chi‐Wai, Jiao, Ying‐Fu, Yu, Wei‐Feng, Li, Yuan‐Hai, and Yang, Li‐Qun
- Subjects
- *
G protein coupled receptors , *POSTOPERATIVE pain , *CHRONIC pain , *OPTOGENETICS , *SURGICAL site , *PAIN threshold - Abstract
Aims: Chronification of postoperative pain is a common clinical phenomenon following surgical operation, and it perplexes a great number of patients. Estrogen and its membrane receptor (G protein‐coupled estrogen receptor, GPER) play a crucial role in pain regulation. Here, we explored the role of GPER in the rostral ventromedial medulla (RVM) during chronic postoperative pain and search for the possible mechanism. Methods and Results: Postoperative pain was induced in mice or rats via a plantar incision surgery. Behavioral tests were conducted to detect both thermal and mechanical pain, showing a small part (16.2%) of mice developed into pain persisting state with consistent low pain threshold on 14 days after incision surgery compared with the pain recovery mice. Immunofluorescent staining assay revealed that the GPER‐positive neurons in the RVM were significantly activated in pain persisting rats. In addition, RT‐PCR and immunoblot analyses showed that the levels of GPER and phosphorylated μ‐type opioid receptor (p‐MOR) in the RVM of pain persisting mice were apparently increased on 14 days after incision surgery. Furthermore, chemogenetic activation of GPER‐positive neurons in the RVM of Gper‐Cre mice could reverse the pain threshold of pain recovery mice. Conversely, chemogenetic inhibition of GPER‐positive neurons in the RVM could prevent mice from being in the pain persistent state. Conclusion: Our findings demonstrated that the GPER in the RVM was responsible for the chronification of postoperative pain and the downstream pathway might be involved in MOR phosphorylation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
28. The sex steroid precursor dehydroepiandrosterone prevents nonalcoholic steatohepatitis by activating the AMPK pathway mediated by GPR30
- Author
-
Longlong Li, Hongjun Wang, Yao Yao, Ji Cao, Zhihao Jiang, Weiyuan Yan, Xu Chu, Qian Li, Miaomiao Lu, and Haitian Ma
- Subjects
Dehydroepiandrosterone ,Biotransformation ,Nonalcoholic steatohepatitis ,AMP-Activated protein kinase ,G protein-coupled estrogen receptor ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
The prevalence of nonalcoholic steatohepatitis (NASH) caused by estrogen deficiency increased sharply in recent decades and has become a major threat to liver health in postmenopausal women. There is no effective strategy to control the incidence and development of NASH. Dehydroepiandrosterone (DHEA) is the most abundant circulating steroid with immune and metabolic regulatory properties, and its level markedly declines with increasing age in humans. Importantly, DHEA can convert into active sex hormones depending on the local needs of target tissues with little diffusion, which serves to avoid systemic side-effects from other tissues’ exposure to estrogen. Here, we found that DHEA prevented the incidence and development of NASH, which is characterized by the reduction of hepatic steatosis, fibrosis, and inflammation in female mice fed with high-fat/high-cholesterol diets and effectively attenuated lipid accumulation, inflammatory response, and oxidative stress in palmitic acid-challenged hepatocytes. Mechanistically, in vitro and in vivo studies showed that the anti-NASH function of DHEA depended on its biotransformation into estrogen rather than androgen, and which up-regulates the expression of G protein-coupled estrogen receptor (GPR30), a non-classical estrogen receptor. The activation of GPR30-mediated AMP-activated protein kinase signaling is a necessary prerequisite for the alleviative effects of DHEA on NASH. Collectively, our data show the mechanisms of DHEA treatment and its effects on NASH that were previously overlooked; the data also show that GPR30 can be used as a target for treating lipid metabolism disorders and related diseases, such as NASH. Furthermore, these findings have the potential to help researchers develop new strategies for preventing NASH in postmenopausal women.
- Published
- 2021
- Full Text
- View/download PDF
29. Editorial: GPER and Human Pathologies
- Author
-
Yves Jacquot, Marilena Kampa, and Sarah H. Lindsey
- Subjects
G protein-coupled estrogen receptor ,human pathologies ,cancer ,inflammation ,nociception ,brain ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Published
- 2021
- Full Text
- View/download PDF
30. Editorial: GPER: Control and Functions
- Author
-
Yves Jacquot, Marilena Kampa, and Sarah H. Lindsey
- Subjects
G protein-coupled estrogen receptor ,sexual dimorphism ,central nervous system ,heart tissue ,signaling cascade ,endocrine-disrupting chemicals ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Published
- 2021
- Full Text
- View/download PDF
31. Triclocarban and triclosan promote breast cancer progression in vitro and in vivo via activating G protein-coupled estrogen receptor signaling pathways.
- Author
-
He, Ting-Ting, Li, Xin, Ma, Jie-Zhi, Yang, Yuan, Zhu, Shiye, Zeng, Jianhua, Luo, Lin, Yin, Yu-Long, and Cao, Lin-Ying
- Published
- 2024
- Full Text
- View/download PDF
32. Targeting the crosstalk between estrogen receptors and membrane growth factor receptors in breast cancer treatment: Advances and opportunities.
- Author
-
Yan, Shunchao, Ji, Jiale, Zhang, Zhijie, Imam, Murshid, Chen, Hong, Zhang, Duo, and Wang, Jinpeng
- Subjects
- *
ESTROGEN , *ESTROGEN receptors , *BREAST cancer , *EPIDERMAL growth factor , *TRIPLE-negative breast cancer , *G protein coupled receptors - Abstract
Estrogens play a critical role in the initiation and progression of breast cancer. Estrogen receptor (ER)α, ERβ, and G protein-coupled estrogen receptor are the primary receptors for estrogen in breast cancer. These receptors are mainly activated by binding with estrogens. The crosstalk between ERs and membrane growth factor receptors creates additional pathways that amplify the effects of their ligands and promote tumor growth. This crosstalk may cause endocrine therapy resistance in ERα-positive breast cancer. Furthermore, this may explain the resistance to anti-human epidermal growth factor receptor-2 (HER2) treatment in ERα-/HER2-positive breast cancer and chemotherapy resistance in triple-negative breast cancer. Accordingly, it is necessary to understand the complex crosstalk between ERs and growth factor receptors. In this review, we delineate the crosstalk between ERs and membrane growth factor receptors in breast cancer. Moreover, this review highlights the current progress in clinical treatment and discusses how pharmaceuticals target the crosstalk. Lastly, we discuss the current challenges and propose potential solutions regarding the implications of targeting crosstalk via pharmacological inhibition. Overall, the present review provides a landscape of the crosstalk between ERs and membrane growth factor receptors in breast cancer, along with valuable insights for future studies and clinical treatments using a chemotherapy-sparing regimen to improve patient quality of life. [Display omitted] • Crosstalk between estrogen receptors and membrane growth factor receptors is crucial in breast cancer development and progression. • Targeting this crosstalk has yielded preliminary preclinical and clinical results; however, further research is essential. • This review discusses crosstalk between estrogen receptors and membrane growth factor receptors in breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. GPER-Induced ERK Signaling Decreases Cell Viability of Hepatocellular Carcinoma
- Author
-
Yu-an Qiu, Jianping Xiong, Qin Fu, Yun Dong, Manran Liu, Meixi Peng, Wenjian Jin, Lixia Zhou, Xue Xu, Xianming Huang, Airong Fu, Guohui Xu, Gang Tu, and Tenghua Yu
- Subjects
G protein-coupled estrogen receptor ,cell viability ,hepatocellular carcinoma ,ERK signaling ,therapeutic target ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Hepatocellular carcinoma (HCC) is an aggressive malignancy with a poor prognosis. Effective biomarkers and specific therapeutic targets for HCC are therefore urgently needed. G protein-coupled estrogen receptor (GPER) plays a crucial role in numerous cancer types; however, its functions in HCC require further exploration. In the present study, we found a remarkable difference in GPER staining between tumor tissue (100/141, 70.9%) and matched non-tumor tissue (27/30, 90.0%). Compared with the GPER-negative patients, the GPER-positive patients with HCC were closely associated with female sex, negative hepatitis B surface antigen, small tumor size, low serum alpha fetoprotein level, and longer overall survival. Treatment with GPER-specific agonist G1 led to the sustained and transient activation of the EGFR/ERK and EGFR/AKT signaling pathways, respectively, in the HCC cell lines HCCLM3 and SMMC-7721, which express high levels of GPER. Interestingly, G1-induced EGFR/ERK signaling, rather than EGFR/AKT signaling mediated by GPER, was involved in decreasing cell viability by blocking cell cycle progression, thereby promoting apoptosis and inhibiting cell growth. Clinical analysis indicated that simultaneous high expression of GPER and phosphorylated-ERK (p-ERK) predicted improved prognosis for HCC. Finally, the activation of GPER/ERK signaling remarkably suppressed tumor growth in an HCC xenograft model, and this result was consistent with the in vitro data. Our findings suggest that specific activation of the GPER/ERK axis may serve as a novel tumor-suppressive mechanism and that this axis could be a therapeutic target for HCC.
- Published
- 2021
- Full Text
- View/download PDF
34. G Protein-Coupled Estrogen Receptor Correlates With Dkk2 Expression and Has Prognostic Impact in Ovarian Cancer Patients
- Author
-
Patricia Fraungruber, Till Kaltofen, Sabine Heublein, Christina Kuhn, Doris Mayr, Alexander Burges, Sven Mahner, Philipp Rathert, Udo Jeschke, and Fabian Trillsch
- Subjects
Dickkopf 2 ,G protein-coupled estrogen receptor ,Wnt signaling ,estrogen ,epithelial ovarian cancer ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
PurposeWnt pathway modulator Dickkopf 2 (Dkk2) and signaling of the G protein-coupled estrogen receptor (GPER) seem to have essential functions in numerous cancer types. For epithelial ovarian cancer (EOC), it has not been proven if either Dkk2 or the GPER on its own have an independent impact on overall survival (OS). So far, the correlation of both factors and their clinical significance has not systematically been investigated before.MethodsExpression levels of Dkk2 were immunohistochemically analyzed in 156 patient samples from different histologic subtypes of EOC applying the immune-reactivity score (IRS). Expression analyses were correlated with clinical and pathological parameters to assess for prognostic relevance. Data analysis was performed using Spearman’s correlations, Kruskal-Wallis-test and Kaplan-Meier estimates.ResultsHighest Dkk2 expression of all subtypes was observed in clear cell carcinoma. In addition, Dkk2 expression differed significantly (p4) and GPER (IRS>8), had a significantly better overall survival compared to patients with low expression (61 months vs. 33 months; p=0.024).ConclusionDkk2 and GPER expression correlates in EOC and combined expression of both is associated with improved OS. These findings underline the clinical significance of both pathways and indicate a possible prognostic impact as well as a potential for treatment strategies addressing interactions between estrogen and Wnt signaling in ovarian cancer.
- Published
- 2021
- Full Text
- View/download PDF
35. GPER-Induced ERK Signaling Decreases Cell Viability of Hepatocellular Carcinoma.
- Author
-
Qiu, Yu-an, Xiong, Jianping, Fu, Qin, Dong, Yun, Liu, Manran, Peng, Meixi, Jin, Wenjian, Zhou, Lixia, Xu, Xue, Huang, Xianming, Fu, Airong, Xu, Guohui, Tu, Gang, and Yu, Tenghua
- Subjects
CELL survival ,HEPATOCELLULAR carcinoma ,HEPATITIS associated antigen ,G protein coupled receptors ,EPIDERMAL growth factor receptors ,HEPATITIS B - Abstract
Hepatocellular carcinoma (HCC) is an aggressive malignancy with a poor prognosis. Effective biomarkers and specific therapeutic targets for HCC are therefore urgently needed. G protein-coupled estrogen receptor (GPER) plays a crucial role in numerous cancer types; however, its functions in HCC require further exploration. In the present study, we found a remarkable difference in GPER staining between tumor tissue (100/141, 70.9%) and matched non-tumor tissue (27/30, 90.0%). Compared with the GPER-negative patients, the GPER-positive patients with HCC were closely associated with female sex, negative hepatitis B surface antigen, small tumor size, low serum alpha fetoprotein level, and longer overall survival. Treatment with GPER-specific agonist G1 led to the sustained and transient activation of the EGFR/ERK and EGFR/AKT signaling pathways, respectively, in the HCC cell lines HCCLM3 and SMMC-7721, which express high levels of GPER. Interestingly, G1-induced EGFR/ERK signaling, rather than EGFR/AKT signaling mediated by GPER, was involved in decreasing cell viability by blocking cell cycle progression, thereby promoting apoptosis and inhibiting cell growth. Clinical analysis indicated that simultaneous high expression of GPER and phosphorylated-ERK (p-ERK) predicted improved prognosis for HCC. Finally, the activation of GPER/ERK signaling remarkably suppressed tumor growth in an HCC xenograft model, and this result was consistent with the in vitro data. Our findings suggest that specific activation of the GPER/ERK axis may serve as a novel tumor-suppressive mechanism and that this axis could be a therapeutic target for HCC. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
36. G Protein-Coupled Estrogen Receptor Correlates With Dkk2 Expression and Has Prognostic Impact in Ovarian Cancer Patients.
- Author
-
Fraungruber, Patricia, Kaltofen, Till, Heublein, Sabine, Kuhn, Christina, Mayr, Doris, Burges, Alexander, Mahner, Sven, Rathert, Philipp, Jeschke, Udo, and Trillsch, Fabian
- Subjects
G protein coupled receptors ,OVARIAN cancer ,OVARIAN epithelial cancer ,CANCER patients ,ESTROGEN receptors - Abstract
Purpose: Wnt pathway modulator Dickkopf 2 (Dkk2) and signaling of the G protein-coupled estrogen receptor (GPER) seem to have essential functions in numerous cancer types. For epithelial ovarian cancer (EOC), it has not been proven if either Dkk2 or the GPER on its own have an independent impact on overall survival (OS). So far, the correlation of both factors and their clinical significance has not systematically been investigated before. Methods: Expression levels of Dkk2 were immunohistochemically analyzed in 156 patient samples from different histologic subtypes of EOC applying the immune-reactivity score (IRS). Expression analyses were correlated with clinical and pathological parameters to assess for prognostic relevance. Data analysis was performed using Spearman's correlations, Kruskal-Wallis-test and Kaplan-Meier estimates. Results: Highest Dkk2 expression of all subtypes was observed in clear cell carcinoma. In addition, Dkk2 expression differed significantly (p<0.001) between low and high grade serous ovarian cancer. A significant correlation of Dkk2 with the cytoplasmic GPER expression was noted (p=0.001) but not for the nuclear estrogen receptor alpha (ERα) or beta (ERβ). Patients exhibiting both, high expression Dkk2 (IRS>4) and GPER (IRS>8), had a significantly better overall survival compared to patients with low expression (61 months vs. 33 months; p=0.024). Conclusion: Dkk2 and GPER expression correlates in EOC and combined expression of both is associated with improved OS. These findings underline the clinical significance of both pathways and indicate a possible prognostic impact as well as a potential for treatment strategies addressing interactions between estrogen and Wnt signaling in ovarian cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
37. Reciprocality Between Estrogen Biology and Calcium Signaling in the Cardiovascular System
- Author
-
Quang-Kim Tran
- Subjects
estrogen ,G protein—coupled estrogen receptor ,calcium ,calmodulin ,calmodulin-binding proteins ,cardiomyocytes ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
17β-Estradiol (E2) is the main estrogenic hormone in the body and exerts many cardiovascular protective effects. Via three receptors known to date, including estrogen receptors α (ERα) and β (ERβ) and the G protein-coupled estrogen receptor 1 (GPER, aka GPR30), E2 regulates numerous calcium-dependent activities in cardiovascular tissues. Nevertheless, effects of E2 and its receptors on components of the calcium signaling machinery (CSM), the underlying mechanisms, and the linked functional impact are only beginning to be elucidated. A picture is emerging of the reciprocality between estrogen biology and Ca2+ signaling. Therein, E2 and GPER, via both E2-dependent and E2-independent actions, moderate Ca2+-dependent activities; in turn, ERα and GPER are regulated by Ca2+ at the receptor level and downstream signaling via a feedforward loop. This article reviews current understanding of the effects of E2 and its receptors on the cardiovascular CSM and vice versa with a focus on mechanisms and combined functional impact. An overview of the main CSM components in cardiovascular tissues will be first provided, followed by a brief review of estrogen receptors and their Ca2+-dependent regulation. The effects of estrogenic agonists to stimulate acute Ca2+ signals will then be reviewed. Subsequently, E2-dependent and E2-independent effects of GPER on components of the Ca2+ signals triggered by other stimuli will be discussed. Finally, a case study will illustrate how the many mechanisms are coordinated to moderate Ca2+-dependent activities in the cardiovascular system.
- Published
- 2020
- Full Text
- View/download PDF
38. Upregulation of G Protein-Coupled Estrogen Receptor by Chrysin-Nanoparticles Inhibits Tumor Proliferation and Metastasis in Triple Negative Breast Cancer Xenograft Model
- Author
-
Kyoung Mee Kim and Joohee Jung
- Subjects
chrysin-nanoparticle ,triple-negative breast cancer ,metastasis ,tumor progression ,G protein-coupled estrogen receptor ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Triple-negative breast cancer (TNBC) is associated with a high mortality rate among women globally. TNBC shows a high rate of recurrence and distant metastasis. Particularly, the chemotherapy is limited because hormone therapy of breast cancer is ineffective. Thus, an effective chemotherapeutic agent is needed for tumor suppression. Chrysin-nanoparticles (chrysin-NPs) were investigated for their inhibitory effect on a MDA-MB-231-derived xenograft model. To gain insight into the underlying mechanisms, we conducted human matrix metalloproteinase (MMP) array, western blot, and immunohistochemistry analysis. Furthermore, in vivo imaging was used to monitor the chemotherapeutic efficacy of chrysin-NPs in a metastasis mouse model. Chrysin-NPs significantly inhibited the proliferation of MDA-MB-231 cells via the PI3K/JNK pathway and induced cell death through the p53-apoptosis pathway, leading to delayed MDA-MB-231-derived tumor growth. Interestingly, chrysin-NPs significantly induced G protein-coupled estrogen receptor (GPER) expression, which suppresses MMPs and NF-κB expression. Chrysin-NPs acted as effective metastasis inhibitors. Our results suggest that chrysin-NPs may be used as an effective adjuvant formulation to inhibit TNBC progression.
- Published
- 2020
- Full Text
- View/download PDF
39. Expression and Role of the G Protein-Coupled Estrogen Receptor (GPR30/GPER) in the Development and Immune Response in Female Reproductive Cancers
- Author
-
Christian David Hernández-Silva, Julio César Villegas-Pineda, and Ana Laura Pereira-Suárez
- Subjects
female reproductive cancers ,estrogen receptor ,G protein-coupled estrogen receptor ,GPER ,GPR30 ,estrogen ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Cancer is a major public health issue and represents the second leading cause of death in women worldwide, as female reproductive-related neoplasms are the main cause of incidence and mortality. Female reproductive cancers have a close relationship to estrogens, the principal female sex steroid hormones. Estrogens exert their actions by the nuclear estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ). ERα, and ERβ act as transcription factors mediating genomic effects. Besides, the G protein-coupled estrogen receptor (GPER, formerly known as GPR30) was recently described as a seven-transmembrane receptor that mediates non-genomic estrogenic signaling, including calcium mobilization, cAMP synthesis, cleavage of matrix metalloproteinases, transactivation of epidermal growth factor receptor (EGFR), and the subsequent activation of PI3K and MAPK signaling pathways, which are the reasons why it is related to cellular processes, such as cell-cycle progression, cellular proliferation, differentiation, apoptosis, migration, and invasion. Since its discovery, selective agonists and antagonists have been found and developed. GPER has been implicated in a variety of hormone-responsiveness tumors, such as breast, endometrial, ovarian, cervical, prostate, and testicular cancer as well as lung, hepatic, thyroid, colorectal, and adrenocortical cancers. Nevertheless, GPER actions in cancer are still debatable due to the conflicting information that has been reported to date, since many reports indicate that activation of this receptor can modulate carcinogenesis. In contrast, many others show that its activation inhibits tumor activity. Besides, estrogens play an essential role in the regulation of the immune system, but little information exists about the role of GPER activation on its modulation within cancer context. This review focuses on the role that the stimulation of GPER plays in female reproductive neoplasms, specifically breast, endometrial, ovarian, and cervical cancers, in its tumor activity and immune response regulation.
- Published
- 2020
- Full Text
- View/download PDF
40. Significance of G Protein-Coupled Estrogen Receptor in the Pathophysiology of Irritable Bowel Syndrome, Inflammatory Bowel Diseases and Colorectal Cancer
- Author
-
Damian Jacenik and Wanda M. Krajewska
- Subjects
G protein-coupled estrogen receptor ,irritable bowel syndrome ,inflammatory bowel diseases ,Crohn's disease ,ulcerative colitis ,colorectal cancer ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
The regulatory role of estrogens and nuclear estrogen receptors, i. e., estrogen receptor α and β has been reported in gastrointestinal diseases. However, the contribution of G protein-coupled estrogen receptor, the membrane-bound estrogen receptor, is still poorly understood. Unlike nuclear estrogen receptors, which are responsible for the genomic activity of estrogens, the G protein-coupled estrogen receptor affects the “rapid” non-genomic activity of estrogens, leading to modulation of many signaling pathways and ultimately changing gene expression. Recently, the crucial role of G protein-coupled estrogen receptor in intestinal pathogenesis has been documented. It has been shown that the G protein-coupled estrogen receptor can modulate the progression of irritable bowel syndrome, inflammatory bowel diseases such as Crohn's disease and ulcerative colitis as well as colorectal cancer. The G protein-coupled estrogen receptor appears to be a potent factor regulating abdominal sensitivity and pain, intestinal peristalsis, colitis development, proliferation and migration potential of colorectal cancer cells and seems to be a useful target in gastrointestinal diseases. In this review, we present the current state of knowledge about the contribution of the G protein-coupled estrogen receptor to irritable bowel syndrome, inflammatory bowel diseases and colorectal cancer.
- Published
- 2020
- Full Text
- View/download PDF
41. GPER agonist G1 suppresses neuronal apoptosis mediated by endoplasmic reticulum stress after cerebral ischemia/reperfusion injury
- Author
-
Zi-Wei Han, Yue-Chen Chang, Ying Zhou, Hang Zhang, Long Chen, Yang Zhang, Jun-Qiang Si, and Li Li
- Subjects
nerve regeneration ,cerebral ischemia/reperfusion injury ,estrogen ,G protein-coupled estrogen receptor ,G1 ,G15 ,endoplasmic reticulum stress ,glucose-regulated protein 78 ,caspase-12 ,C/EBP homologous protein ,neuronal apoptosis ,neural regeneration ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Studies have confirmed a strong association between activation of the endoplasmic reticulum stress pathway and cerebral ischemia/reperfusion (I/R) injury. In this study, three key proteins in the endoplasmic reticulum stress pathway (glucose-regulated protein 78, caspase-12, and C/EBP homologous protein) were selected to examine the potential mechanism of endoplasmic reticulum stress in the neuroprotective effect of G protein-coupled estrogen receptor. Female Sprague-Dawley rats received ovariectomy (OVX), and then cerebral I/R rat models (OVX + I/R) were established by middle cerebral artery occlusion. Immediately after I/R, rat models were injected with 100 μg/kg E2 (OVX + I/R + E2), or 100 μg/kg G protein-coupled estrogen receptor agonist G1 (OVX + I/R + G1) in the lateral ventricle. Longa scoring was used to detect neurobehavioral changes in each group. Infarct volumes were measured by 2,3,5-triphenyltetrazolium chloride staining. Morphological changes in neurons were observed by Nissl staining. Terminal dexynucleotidyl transferase-mediated nick end-labeling staining revealed that compared with the OVX + I/R group, neurological function was remarkably improved, infarct volume was reduced, number of normal Nissl bodies was dramatically increased, and number of apoptotic neurons in the hippocampus was decreased after E2 and G1 intervention. To detect the expression and distribution of endoplasmic reticulum stress-related proteins in the endoplasmic reticulum, caspase-12 distribution and expression were detected by immunofluorescence, and mRNA and protein levels of glucose-regulated protein 78, caspase-12, and C/EBP homologous protein were determined by polymerase chain reaction and western blot assay. The results showed that compared with the OVX + I/R group, E2 and G1 treatment obviously decreased mRNA and protein expression levels of glucose-regulated protein 78, C/EBP homologous protein, and caspase-12. However, the G protein-coupled estrogen receptor antagonist G15 (OVX + I/R + E2 + G15) could eliminate the effect of E2 on cerebral I/R injury. These results confirm that E2 and G protein-coupled estrogen receptor can inhibit the expression of endoplasmic reticulum stress-related proteins and neuronal apoptosis in the hippocampus, thereby improving dysfunction caused by cerebral I/R injury. Every experimental protocol was approved by the Institutional Ethics Review Board at the First Affiliated Hospital of Shihezi University School of Medicine, China (approval No. SHZ A2017-171) on February 27, 2017.
- Published
- 2019
- Full Text
- View/download PDF
42. GPER was associated with hypertension in post-menopausal women
- Author
-
Liu Shichao, Ding Tongbin, Liu Hang, and Jian Liguo
- Subjects
hypertension ,g protein-coupled estrogen receptor ,menopause ,estrogen receptor ,case-control ,Medicine - Abstract
To explore the relationship between G protein-coupled estrogen receptor (GPER) and hypertension in post-menopausal women.
- Published
- 2018
- Full Text
- View/download PDF
43. Mechanisms of Estradiol-induced EGF-like Factor Expression and Oocyte Maturation via G Protein-coupled Estrogen Receptor.
- Author
-
Zhang, Hui, Lu, Sihai, Xu, Rui, Tang, Yaju, Liu, Jie, Li, Chan, Wei, Juncai, Yao, Ru, Zhao, Xiaoe, Wei, Qiang, and Ma, Baohua
- Subjects
G protein coupled receptors ,CYCLIC adenylic acid ,EPIDERMAL growth factor ,EPIDERMAL growth factor receptors ,ESTRADIOL ,ESTROGEN - Abstract
Estrogen is an important modulator of reproductive activity through nuclear receptors and G protein–coupled estrogen receptor (GPER). Here, we observed that both estradiol and the GPER-specific agonist G1 rapidly induced cyclic adenosine monophosphate (cAMP) production in cumulus cells, leading to transient stimulation of phosphorylated cAMP response element binding protein (CREB), which was conducive to the transcription of epidermal growth factor (EGF)-like factors, amphiregulin, epiregulin, and betacellulin. Inhibition of GPER by G15 significantly reduced estradiol-induced CREB phosphorylation and EGF-like factor gene expression. Consistently, the silencing of GPER expression in cultured cumulus cells abrogated the estradiol-induced CREB phosphorylation and EGF-like factor transcription. In addition, the increase in EGF-like factor expression in the cumulus cells is associated with EGF receptor (EFGR) tyrosine kinase phosphorylation and extracellular signal–regulated kinase 1/2 (ERK1/2) activation. Furthermore, we demonstrated that GPER-mediated phosphorylation of EGFR and ERK1/2 was involved in reduced gap junction communication, cumulus expansion, increased oocyte mitochondrial activity and first polar body extrusion. Overall, our study identified a novel function for estrogen in regulating EGFR activation via GPER in cumulus cells during oocyte maturation. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
44. Reciprocality Between Estrogen Biology and Calcium Signaling in the Cardiovascular System.
- Author
-
Tran, Quang-Kim
- Subjects
CARDIOVASCULAR system ,ESTROGEN receptors ,G protein coupled receptors ,CALCIUM ,ESTROGEN - Abstract
17β-Estradiol (E
2 ) is the main estrogenic hormone in the body and exerts many cardiovascular protective effects. Via three receptors known to date, including estrogen receptors α (ERα) and β (ERβ) and the G protein-coupled estrogen receptor 1 (GPER, aka GPR30), E2 regulates numerous calcium-dependent activities in cardiovascular tissues. Nevertheless, effects of E2 and its receptors on components of the calcium signaling machinery (CSM), the underlying mechanisms, and the linked functional impact are only beginning to be elucidated. A picture is emerging of the reciprocality between estrogen biology and Ca2+ signaling. Therein, E2 and GPER, via both E2 -dependent and E2 -independent actions, moderate Ca2+ -dependent activities; in turn, ERα and GPER are regulated by Ca2+ at the receptor level and downstream signaling via a feedforward loop. This article reviews current understanding of the effects of E2 and its receptors on the cardiovascular CSM and vice versa with a focus on mechanisms and combined functional impact. An overview of the main CSM components in cardiovascular tissues will be first provided, followed by a brief review of estrogen receptors and their Ca2+ -dependent regulation. The effects of estrogenic agonists to stimulate acute Ca2+ signals will then be reviewed. Subsequently, E2 -dependent and E2 -independent effects of GPER on components of the Ca2+ signals triggered by other stimuli will be discussed. Finally, a case study will illustrate how the many mechanisms are coordinated to moderate Ca2+ -dependent activities in the cardiovascular system. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
45. Upregulation of G Protein-Coupled Estrogen Receptor by Chrysin-Nanoparticles Inhibits Tumor Proliferation and Metastasis in Triple Negative Breast Cancer Xenograft Model.
- Author
-
Kim, Kyoung Mee and Jung, Joohee
- Subjects
TRIPLE-negative breast cancer ,G protein coupled receptors ,METASTASIS ,CANCER hormone therapy ,ESTROGEN antagonists - Abstract
Triple-negative breast cancer (TNBC) is associated with a high mortality rate among women globally. TNBC shows a high rate of recurrence and distant metastasis. Particularly, the chemotherapy is limited because hormone therapy of breast cancer is ineffective. Thus, an effective chemotherapeutic agent is needed for tumor suppression. Chrysin-nanoparticles (chrysin-NPs) were investigated for their inhibitory effect on a MDA-MB-231-derived xenograft model. To gain insight into the underlying mechanisms, we conducted human matrix metalloproteinase (MMP) array, western blot, and immunohistochemistry analysis. Furthermore, in vivo imaging was used to monitor the chemotherapeutic efficacy of chrysin-NPs in a metastasis mouse model. Chrysin-NPs significantly inhibited the proliferation of MDA-MB-231 cells via the PI3K/JNK pathway and induced cell death through the p53-apoptosis pathway, leading to delayed MDA-MB-231-derived tumor growth. Interestingly, chrysin-NPs significantly induced G protein-coupled estrogen receptor (GPER) expression, which suppresses MMPs and NF-κB expression. Chrysin-NPs acted as effective metastasis inhibitors. Our results suggest that chrysin-NPs may be used as an effective adjuvant formulation to inhibit TNBC progression. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
46. Expression and Role of the G Protein-Coupled Estrogen Receptor (GPR30/GPER) in the Development and Immune Response in Female Reproductive Cancers.
- Author
-
Hernández-Silva, Christian David, Villegas-Pineda, Julio César, and Pereira-Suárez, Ana Laura
- Subjects
G protein coupled receptors ,TESTICULAR cancer ,ESTROGEN antagonists ,EPIDERMAL growth factor receptors ,IMMUNOREGULATION ,ESTROGEN receptors ,CYCLIC-AMP-dependent protein kinase ,SEX hormones - Abstract
Cancer is a major public health issue and represents the second leading cause of death in women worldwide, as female reproductive-related neoplasms are the main cause of incidence and mortality. Female reproductive cancers have a close relationship to estrogens, the principal female sex steroid hormones. Estrogens exert their actions by the nuclear estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ). ERα, and ERβ act as transcription factors mediating genomic effects. Besides, the G protein-coupled estrogen receptor (GPER, formerly known as GPR30) was recently described as a seven-transmembrane receptor that mediates non-genomic estrogenic signaling, including calcium mobilization, cAMP synthesis, cleavage of matrix metalloproteinases, transactivation of epidermal growth factor receptor (EGFR), and the subsequent activation of PI3K and MAPK signaling pathways, which are the reasons why it is related to cellular processes, such as cell-cycle progression, cellular proliferation, differentiation, apoptosis, migration, and invasion. Since its discovery, selective agonists and antagonists have been found and developed. GPER has been implicated in a variety of hormone-responsiveness tumors, such as breast, endometrial, ovarian, cervical, prostate, and testicular cancer as well as lung, hepatic, thyroid, colorectal, and adrenocortical cancers. Nevertheless, GPER actions in cancer are still debatable due to the conflicting information that has been reported to date, since many reports indicate that activation of this receptor can modulate carcinogenesis. In contrast, many others show that its activation inhibits tumor activity. Besides, estrogens play an essential role in the regulation of the immune system, but little information exists about the role of GPER activation on its modulation within cancer context. This review focuses on the role that the stimulation of GPER plays in female reproductive neoplasms, specifically breast, endometrial, ovarian, and cervical cancers, in its tumor activity and immune response regulation. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
47. Towards understanding leydigioma: do G protein-coupled estrogen receptor and peroxisome proliferator–activated receptor regulate lipid metabolism and steroidogenesis in Leydig cell tumors?
- Author
-
Kotula-Balak, M., Gorowska-Wojtowicz, E., Milon, A., Pawlicki, P., Tworzydlo, W., Płachno, B. J., Krakowska, I, Hejmej, A., Wolski, J. K., and Bilinska, B.
- Subjects
- *
STEROIDOGENIC acute regulatory protein , *G protein coupled receptors , *LEYDIG cells , *PEROXISOME proliferator-activated receptors , *LIPID metabolism , *TRANSLOCATOR proteins , *ESTROGEN - Abstract
Leydig cell tumors (LCT) are the most common type of testicular stromal tumor. Herein, we investigate the G protein-coupled estrogen receptor (GPER) and peroxisome proliferator–activated receptor (PPAR) implication in regulation of lipid homeostasis including the expression of steroidogenesis-controlling molecules in clinical specimens of LCTs and tumor Leydig cells (MA-10). We showed the general structure and morphology of LCTs by scanning electron and light microscopy. In LCTs, mRNA and protein analyses revealed increased expression of GPER and decreased expression of PPARα, β, and γ. Concomitantly, changes in expression pattern of the lutropin receptor (LHR), protein kinase A (PKA), perilipin (PLIN), hormone sensitive lipase (HSL), steroidogenic acute regulatory protein (StAR), translocator protein (TSPO), HMG-CoA synthase, and reductase (HMGCS, HMGCR) were observed. Using MA-10 cells treated with GPER and PPAR antagonists (alone and in combination), we demonstrated GPER-PPAR–mediated control of estradiol secretion via GPER-PPARα and cyclic guanosine monophosphate (cGMP) concentration via GPER-PPARγ. It is assumed that GPER and PPAR can crosstalk, and this can be altered in LCT, resulting in a perturbed lipid balance and steroidogenesis. In LCTs, the phosphatidylinositol-3-kinase (PI3K)-Akt-mTOR pathway was disturbed. Thus, PI3K-Akt-mTOR with cGMP can play a role in LCT outcome and biology including lipid metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
48. Oestrogen receptors and hypoxia inducible factor 1 alpha expression in abdominal wall endometriosis.
- Author
-
Zhang, Ling, Xiong, Wenqian, Fu, Tian, Long, Xuefeng, Zhang, Zhibing, Liu, Yi, and Lv, Gang
- Subjects
- *
ABDOMINAL wall , *G protein coupled receptors , *ESTROGEN , *EPITHELIAL cells , *HYPOXEMIA - Abstract
Research question: What are the protein levels and localization of oestrogen receptors (including ERa, ERb and G protein-coupled oestrogen receptor [GPER]) and hypoxia-inducible factor-1alpha (HIF-1a) in normal control endometrium (COEM) and ectopic endometrium from abdominal wall endometriosis (AWE). Design: AWE (n = 20) were obtained during surgery; COEM (n = 40) were collected by curettage. All tissues were obtained during the proliferative or secretory phase. Formalin-fixed paraffin-embedded tissues were used for immunohistochemical study for oestrogen receptors and HIF-1a proteins. Result(s): The expression of oestrogen receptors and HIF-1a in AWE differed from that in the corresponding menstrual cycle phase of COEM. Compared with COEM, ERa and HIF-1a were decreased whereas ERb and GPER were increased in AWE. The greatest difference was in GPER, with increased protein expression in both the cytoplasm and nucleus of endometrial epithelial and stromal cells, as well as a distinct change in localization from cytoplasmic expression to nuclear and cytoplasmic expression, compared with COEM. Conclusions: Our data suggest that the expression changes of oestrogen receptors and HIF-1a, especially GPER, are associated with AWE, which may provide new clues to understanding the cause of endometriosis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
49. Significance of G Protein-Coupled Estrogen Receptor in the Pathophysiology of Irritable Bowel Syndrome, Inflammatory Bowel Diseases and Colorectal Cancer.
- Author
-
Jacenik, Damian and Krajewska, Wanda M.
- Subjects
INFLAMMATORY bowel diseases ,G protein coupled receptors ,COLON diseases ,CROHN'S disease ,COLORECTAL cancer - Abstract
The regulatory role of estrogens and nuclear estrogen receptors, i. e., estrogen receptor α and β has been reported in gastrointestinal diseases. However, the contribution of G protein-coupled estrogen receptor, the membrane-bound estrogen receptor, is still poorly understood. Unlike nuclear estrogen receptors, which are responsible for the genomic activity of estrogens, the G protein-coupled estrogen receptor affects the "rapid" non-genomic activity of estrogens, leading to modulation of many signaling pathways and ultimately changing gene expression. Recently, the crucial role of G protein-coupled estrogen receptor in intestinal pathogenesis has been documented. It has been shown that the G protein-coupled estrogen receptor can modulate the progression of irritable bowel syndrome, inflammatory bowel diseases such as Crohn's disease and ulcerative colitis as well as colorectal cancer. The G protein-coupled estrogen receptor appears to be a potent factor regulating abdominal sensitivity and pain, intestinal peristalsis, colitis development, proliferation and migration potential of colorectal cancer cells and seems to be a useful target in gastrointestinal diseases. In this review, we present the current state of knowledge about the contribution of the G protein-coupled estrogen receptor to irritable bowel syndrome, inflammatory bowel diseases and colorectal cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
50. The Effects of 17α‐Ethinylestradiol on the Heart Rate of Embryonic Japanese Medaka (Oryzias latipes).
- Author
-
Anderson, Jordan Christopher, Beyger, Lindsay, Guchardi, John, and Holdway, Douglas Alan
- Subjects
- *
HEART beat , *ORYZIAS latipes , *G protein coupled receptors , *BIOLOGICAL systems , *ESTROGEN receptors , *ESTROGEN - Abstract
Estrogen toxicity has been an area of priority in aquatic toxicology over the last 20 yr. Currently, estrogen toxicity is primarily linked to classical estrogen signaling, the interaction of estrogen receptors alpha and beta (ERα and ERβ). Recent evidence has indicated that a rapid, nongenomic, nonclassical estrogen signaling pathway exists via the G protein–coupled estrogen receptor (GPER), which is expressed in many biological systems, with roles in the cardiovascular system. The objective of the present study was to investigate the effect of 17α‐ethinylestradiol (EE2) on the heart rate of embryonic Japanese medaka (Oryzias latipes). A significant decrease (bradycardia) in embryonic heart rate was observed at all treatment concentrations (0.1, 1, 10, 100, and 1000 ng/L EE2) at 144, 168, and 192 h postfertilization (hpf; p ≤ 0.05), whereas 120 and 216 hpf embryos experienced a significant decrease from the control at 10, 100, and 1000 ng/L EE2 and 0.1, 100, and 1000 ng/L EE2, respectively (p ≤ 0.05). In addition, using select estrogen receptor modulators, it was demonstrated that estrogen‐induced bradycardia appears to be linked to GPER and not ERα and ERβ. The present study highlights GPER as a novel and alternative mode of action for EE2 toxicity at environmentally relevant concentrations. Environ Toxicol Chem 2020;39:904–912. © 2020 SETAC [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.