Academic literature on the topic 'G-Protein Coupled Estrogen Receptor (GPER)'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'G-Protein Coupled Estrogen Receptor (GPER).'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "G-Protein Coupled Estrogen Receptor (GPER)"

1

Arterburn, Jeffrey B., and Eric R. Prossnitz. "G Protein–Coupled Estrogen Receptor GPER: Molecular Pharmacology and Therapeutic Applications." Annual Review of Pharmacology and Toxicology 63, no. 1 (January 20, 2023): 295–320. http://dx.doi.org/10.1146/annurev-pharmtox-031122-121944.

Full text
Abstract:
The actions of estrogens and related estrogenic molecules are complex and multifaceted in both sexes. A wide array of natural, synthetic, and therapeutic molecules target pathways that produce and respond to estrogens. Multiple receptors promulgate these responses, including the classical estrogen receptors of the nuclear hormone receptor family (estrogen receptors α and β), which function largely as ligand-activated transcription factors, and the 7-transmembrane G protein–coupled estrogen receptor, GPER, which activates a diverse array of signaling pathways. The pharmacology and functional roles of GPER in physiology and disease reveal important roles in responses to both natural and synthetic estrogenic compounds in numerous physiological systems. These functions have implications in the treatment of myriad disease states, including cancer, cardiovascular diseases, and metabolic disorders. This review focuses on the complex pharmacology of GPER and summarizes major physiological functions of GPER and the therapeutic implications and ongoing applications of GPER-targeted compounds.
APA, Harvard, Vancouver, ISO, and other styles
2

Meyer, Matthias R., Natalie C. Fredette, Matthias Barton, and Eric R. Prossnitz. "G protein-coupled estrogen receptor inhibits vascular prostanoid production and activity." Journal of Endocrinology 227, no. 1 (August 24, 2015): 61–69. http://dx.doi.org/10.1530/joe-15-0257.

Full text
Abstract:
Complications of atherosclerotic vascular disease, such as myocardial infarction and stroke, are the most common causes of death in postmenopausal women. Endogenous estrogens inhibit vascular inflammation-driven atherogenesis, a process that involves cyclooxygenase (COX)-derived vasoconstrictor prostanoids such as thromboxane A2. Here, we studied whether the G protein-coupled estrogen receptor (GPER) mediates estrogen-dependent inhibitory effects on prostanoid production and activity under pro-inflammatory conditions. Effects of estrogen on production of thromboxane A2were determined in human endothelial cells stimulated by the pro-inflammatory cytokine tumour necrosis factor alpha (TNF-α). Moreover,Gper-deficient (Gper−/−) and WT mice were fed a pro-inflammatory diet and underwent ovariectomy or sham surgery to unmask the role of endogenous estrogens. Thereafter, contractions to acetylcholine-stimulated endothelial vasoconstrictor prostanoids and the thromboxane-prostanoid receptor agonist U46619 were recorded in isolated carotid arteries. In endothelial cells, TNF-α-stimulated thromboxane A2production was inhibited by estrogen, an effect blocked by the GPER-selective antagonist G36. In ovary-intact mice, deletion ofGperincreased prostanoid-dependent contractions by twofold. Ovariectomy also augmented prostanoid-dependent contractions by twofold in WT mice but had no additional effect inGper−/−mice. These contractions were blocked by the COX inhibitor meclofenamate and unaffected by the nitric oxide synthase inhibitorl-NG-nitroarginine methyl ester. Vasoconstrictor responses to U46619 did not differ between groups, indicating intact signaling downstream of thromboxane-prostanoid receptor activation. In summary, under pro-inflammatory conditions, estrogen inhibits vasoconstrictor prostanoid production in endothelial cells and activity in intact arteries through GPER. Selective activation of GPER may therefore be considered as a novel strategy to treat increased prostanoid-dependent vasomotor tone or vascular disease in postmenopausal women.
APA, Harvard, Vancouver, ISO, and other styles
3

Qian, Hongyan, Jingxiu Xuan, Yuan Liu, and Guixiu Shi. "Function of G-Protein-Coupled Estrogen Receptor-1 in Reproductive System Tumors." Journal of Immunology Research 2016 (2016): 1–6. http://dx.doi.org/10.1155/2016/7128702.

Full text
Abstract:
The G-protein-coupled estrogen receptor-1 (GPER-1), also known as GPR30, is a novel estrogen receptor mediating estrogen receptor signaling in multiple cell types. The progress of estrogen-related cancer is promoted by GPER-1 activation through mitogen-activated protein kinases (MAPK), phosphoinositide 3-kinase (PI3K), and phospholipase C (PLC) signaling pathways. However, this promoting effect of GPER-1 is nonclassic estrogen receptor (ER) dependent manner. In addition, clinical evidences revealed that GPER-1 is associated with estrogen resistance in estrogen-related cancer patients. These give a hint that GPER-1 may be a novel therapeutic target for the estrogen-related cancers. However, preclinical studies also found that GPER-1 activation of its special agonist G-1 inhibits cancer cell proliferation. This review aims to summarize the characteristics and complex functions of GPER-1 in cancers.
APA, Harvard, Vancouver, ISO, and other styles
4

Filardo, Edward J., and Peter Thomas. "Minireview: G Protein-Coupled Estrogen Receptor-1, GPER-1: Its Mechanism of Action and Role in Female Reproductive Cancer, Renal and Vascular Physiology." Endocrinology 153, no. 7 (April 11, 2012): 2953–62. http://dx.doi.org/10.1210/en.2012-1061.

Full text
Abstract:
Using cDNA cloning strategies commonly employed for G protein-coupled receptors (GPCR), GPCR-30 (GPR30), was isolated from mammalian cells before knowledge of its cognate ligand. GPR30 is evolutionarily conserved throughout the vertebrates. A broad literature suggests that GPR30 is a Gs-coupled heptahelical transmembrane receptor that promotes specific binding of naturally occurring and man-made estrogens but not cortisol, progesterone, or testosterone. Its “pregenomic” signaling actions are manifested by plasma membrane-associated actions familiar to GPCR, namely, stimulation of adenylyl cyclase and Gβγ-subunit protein-dependent release of membrane-tethered heparan bound epidermal growth factor. These facts regarding its mechanism of action have led to the formal renaming of this receptor to its current functional designate, G protein-coupled estrogen receptor (ER) (GPER)-1. Further insight regarding its biochemical action and physiological functions in vertebrates is derived from receptor knockdown studies and the use of selective agonists/antagonists that discriminate GPER-1 from the nuclear steroid hormone receptors, ERα and ERβ. GPER-1-selective agents have linked GPER-1 to physiological and pathological events regulated by estrogen action, including, but not limited to, the central nervous, immune, renal, reproductive, and cardiovascular systems. Moreover, immunohistochemical studies have shown a positive association between GPER-1 expression and progression of female reproductive cancer, a relationship that is diametrically opposed from ER. Unlike ER knockout mice, GPER-1 knockout mice are fertile and show no overt reproductive anomalies. However, they do exhibit thymic atrophy, impaired glucose tolerance, and altered bone growth. Here, we discuss the role of GPER-1 in female reproductive cancers as well as renal and vascular physiology.
APA, Harvard, Vancouver, ISO, and other styles
5

Petrie, Whitney K., Megan K. Dennis, Chelin Hu, Donghai Dai, Jeffrey B. Arterburn, Harriet O. Smith, Helen J. Hathaway, and Eric R. Prossnitz. "G Protein-Coupled Estrogen Receptor-Selective Ligands Modulate Endometrial Tumor Growth." Obstetrics and Gynecology International 2013 (2013): 1–17. http://dx.doi.org/10.1155/2013/472720.

Full text
Abstract:
Endometrial carcinoma is the most common cancer of the female reproductive tract. GPER/GPR30 is a 7-transmembrane spanning G protein-coupled receptor that has been identified as the third estrogen receptor, in addition to ERαand ERβ. High GPER expression is predictive of poor survival in endometrial and ovarian cancer, but despite this, the estrogen-mediated signaling pathways and specific estrogen receptors involved in endometrial cancer remain unclear. Here, employing ERα-negative Hec50 endometrial cancer cells, we demonstrate that GPER mediates estrogen-stimulated activation of ERK and PI3K via matrix metalloproteinase activation and subsequent transactivation of the EGFR and that ER-targeted therapeutic agents (4-hydroxytamoxifen, ICI182,780/fulvestrant, and Raloxifene), the phytoestrogen genistein, and the “ERα-selective” agonist propylpyrazole triol also function as GPER agonists. Furthermore, xenograft tumors of Hec50 cells yield enhanced growth with G-1 and estrogen, the latter being inhibited by GPER-selective pharmacologic antagonism with G36. These results have important implications with respect to the use of putatively ER-selective ligands and particularly for the widespread long-term use of “ER-targeted” therapeutics. Moreover, our findings shed light on the potential mechanisms of SERM/SERD side effects reported in many clinical studies. Finally, our results provide the first demonstration that pharmacological inhibition of GPER activityin vivoprevents estrogen-mediated tumor growth.
APA, Harvard, Vancouver, ISO, and other styles
6

Hsu, Li-Han, Nei-Min Chu, Yung-Feng Lin, and Shu-Huei Kao. "G-Protein Coupled Estrogen Receptor in Breast Cancer." International Journal of Molecular Sciences 20, no. 2 (January 14, 2019): 306. http://dx.doi.org/10.3390/ijms20020306.

Full text
Abstract:
The G-protein coupled estrogen receptor (GPER), an alternate estrogen receptor (ER) with a structure distinct from the two canonical ERs, being ERα, and ERβ, is expressed in 50% to 60% of breast cancer tissues and has been presumed to be associated with the development of tamoxifen resistance in ERα positive breast cancer. On the other hand, triple-negative breast cancer (TNBC) constitutes 15% to 20% of breast cancers and frequently displays a more aggressive behavior. GPER is prevalent and involved in TNBC and can be a therapeutic target. However, contradictory results exist regarding the function of GPER in breast cancer, proliferative or pro-apoptotic. A better understanding of the GPER, its role in breast cancer, and the interactions with the ER and epidermal growth factor receptor will be beneficial for the disease management and prevention in the future.
APA, Harvard, Vancouver, ISO, and other styles
7

Molina, Luis, Felipe A. Bustamante, Kanti D. Bhoola, Carlos D. Figueroa, and Pamela Ehrenfeld. "Possible role of phytoestrogens in breast cancer via GPER-1/GPR30 signaling." Clinical Science 132, no. 24 (December 13, 2018): 2583–98. http://dx.doi.org/10.1042/cs20180885.

Full text
Abstract:
Estrogens generated within endocrine organs and the reproductive system act as ligands for at least three types of estrogen receptors. Estrogen receptors α (ERα) and β (ERβ) belong to the so-called classical family of estrogen receptors, whereas the G protein-coupled receptor GPR30, also known as GPER-1, has been described as a novel estrogen receptor sited in the cell membrane of target cells. Furthermore, these receptors are under stimulation of a family of exogenous estrogens, known as phytoestrogens, which are a diverse group of non-steroidal plant compounds derived from plant food consumed by humans and animals. Because phytoestrogens are omnipresent in our daily diet, they are becoming increasingly important in both human health and disease. Recent evidence indicates that in addition to classical estrogen receptors, phytoestrogens also activate GPER-1 a relevant observation since GPER-1 is involved in several physiopathological disorders and especially in estrogen-dependent diseases such as breast cancer. The first estrogen receptors discovered were the classical ERα and ERβ, but from an evolutionary point of view G protein-coupled receptors trace their origins in history to over a billion years ago suggesting that estrogen receptors like GPER-1 may have been the targets of choice for ancient phytoestrogens and/or estrogens. This review provides a comprehensive and systematic literature search on phytoestrogens and its relationship with classical estrogen receptors and GPER-1 including its role in breast cancer, an issue still under discussion.
APA, Harvard, Vancouver, ISO, and other styles
8

Lu, Dingqiang, Xinqian Wang, Chunlei Feng, Danyang Liu, Yixuan Liu, Yujiao Liu, Jie Li, et al. "Study of the Sensing Kinetics of G Protein-Coupled Estrogen Receptor Sensors for Common Estrogens and Estrogen Analogs." Molecules 28, no. 8 (April 7, 2023): 3286. http://dx.doi.org/10.3390/molecules28083286.

Full text
Abstract:
Endogenous and exogenous estrogens are widely present in food and food packaging, and high levels of natural estrogens and the misuse or illegal use of synthetic estrogens can lead to endocrine disorders and even cancer in humans. Therefore, it is consequently important to accurately evaluate the presence of food-functional ingredients or toxins with estrogen-like effects. In this study, an electrochemical sensor based on G protein-coupled estrogen receptors (GPERs) was fabricated by self-assembly, modified by double-layered gold nanoparticles, and used to measure the sensing kinetics for five GPER ligands. The interconnected allosteric constants (Ka) of the sensor for 17β-estradiol, resveratrol, G-1, G-15, and bisphenol A were 8.90 × 10−17, 8.35 × 10−16, 8.00 × 10−15, 5.01 × 10−15, and 6.65 × 10−16 mol/L, respectively. The sensitivity of the sensor for the five ligands followed the order of 17β-estradiol > bisphenol A > resveratrol > G-15 > G-1. The receptor sensor also demonstrated higher sensor sensitivity for natural estrogens than exogenous estrogens. The results of molecular simulation docking showed that the residues Arg, Glu, His, and Asn of GPER mainly formed hydrogen bonds with -OH, C-O-C, or -NH-. In this study, simulating the intracellular receptor signaling cascade with an electrochemical signal amplification system enabled us to directly measure GPER–ligand interactions and explore the kinetics after the self-assembly of GPERs on a biosensor. This study also provides a novel platform for the accurate functional evaluation of food-functional components and toxins.
APA, Harvard, Vancouver, ISO, and other styles
9

Yu, Xuan, Handong Ma, Scott A. Barman, Alexander T. Liu, Minga Sellers, John N. Stallone, Eric R. Prossnitz, Richard E. White, and Guichun Han. "Activation of G protein-coupled estrogen receptor induces endothelium-independent relaxation of coronary artery smooth muscle." American Journal of Physiology-Endocrinology and Metabolism 301, no. 5 (November 2011): E882—E888. http://dx.doi.org/10.1152/ajpendo.00037.2011.

Full text
Abstract:
Estrogens can either relax or contract arteries via rapid, nongenomic mechanisms involving classic estrogen receptors (ER). In addition to ERα and ERβ, estrogen may also stimulate G protein-coupled estrogen receptor 1 (GPER) in nonvascular tissue; however, a potential role for GPER in coronary arteries is unclear. The purpose of this study was to determine how GPER activity influenced coronary artery reactivity. In vitro isometric force recordings were performed on endothelium-denuded porcine arteries. These studies were augmented by RT-PCR and single-cell patch-clamp experiments. RT-PCR and immunoblot studies confirmed expression of GPER mRNA and protein, respectively, in smooth muscle from either porcine or human coronary arteries. G-1, a selective GPER agonist, produced a concentration-dependent relaxation of endothelium-denuded porcine coronary arteries in vitro. This response was attenuated by G15, a GPER-selective antagonist, or by inhibiting large-conductance calcium-activated potassium (BKCa) channels with iberiotoxin, but not by inhibiting NO signaling. Last, single-channel patch-clamp studies demonstrated that G-1 stimulates BKCa channel activity in intact smooth muscle cells from either porcine or human coronary arteries but had no effect on channels isolated in excised membrane patches. In summary, GPER activation relaxes coronary artery smooth muscle by increasing potassium efflux via BKCa channels and requires an intact cellular signaling mechanism. This novel action of estrogen-like compounds may help clarify some of the controversy surrounding the vascular effects of estrogens.
APA, Harvard, Vancouver, ISO, and other styles
10

Chuang, Shu-Chun, Chung-Hwan Chen, Ya-Shuan Chou, Mei-Ling Ho, and Je-Ken Chang. "G Protein-Coupled Estrogen Receptor Mediates Cell Proliferation through the cAMP/PKA/CREB Pathway in Murine Bone Marrow Mesenchymal Stem Cells." International Journal of Molecular Sciences 21, no. 18 (September 5, 2020): 6490. http://dx.doi.org/10.3390/ijms21186490.

Full text
Abstract:
Estrogen is an important hormone to regulate skeletal physiology via estrogen receptors. The traditional estrogen receptors are ascribed to two nuclear estrogen receptors (ERs), ERα and ERβ. Moreover, G protein-coupled estrogen receptor-1 (GPER-1) was reported as a membrane receptor for estrogen in recent years. However, whether GPER-1 regulated osteogenic cell biology on skeletal system is still unclear. GPER-1 is expressed in growth plate abundantly before puberty but decreased abruptly since the very late stage of puberty in humans. It indicates GPER-1 might play an important role in skeletal growth regulation. GPER-1 expression has been confirmed in osteoblasts, osteocytes and chondrocytes, but its expression in mesenchymal stem cells (MSCs) has not been confirmed. In this study, we hypothesized that GPER-1 is expressed in bone MSCs (BMSC) and enhances BMSC proliferation. The cultured tibiae of neonatal rat and murine BMSCs were tested in our study. GPER-1-specific agonist (G-1) and antagonist (G-15), and GPER-1 siRNA (siGPER-1) were used to evaluate the downstream signaling pathway and cell proliferation. Our results revealed BrdU-positive cell counts were higher in cultured tibiae in the G-1 group. The G-1 also enhanced the cell viability and proliferation, whereas G-15 and siGPER-1 reduced these activities. The cAMP and phosphorylation of CREB were enhanced by G-1 but inhibited by G-15. We further demonstrated that GPER-1 mediates BMSC proliferation via the cAMP/PKA/p-CREB pathway and subsequently upregulates cell cycle regulators, cyclin D1/cyclin-dependent kinase (CDK) 6 and cyclin E1/CDK2 complex. The present study is the first to report that GPER-1 mediates BMSC proliferation. This finding indicates that GPER-1 mediated signaling positively regulates BMSC proliferation and may provide novel insights into addressing estrogen-mediated bone development.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "G-Protein Coupled Estrogen Receptor (GPER)"

1

Attarhaie, Tehrani Mahtab. "Anatomical Expression and Functional Role of the G-Protein Coupled Estrogen Receptor 1 in the Song System of Zebra Finches (Taeniopygia guttata)." Kent State University / OhioLINK, 2018. http://rave.ohiolink.edu/etdc/view?acc_num=kent152416406994131.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Acharya, Kalpana D. Ms. "ROLE OF MEMBRANE BOUND G-PROTEIN COUPLED ESTROGEN RECEPTOR GPR30 AND Z-LINKED RIBOSOMAL GENE S6 (RPS6) IN SEXUALLY DIMORPHIC DEVELOPMENT OF THE ZEBRA FINCH BRAIN." Kent State University / OhioLINK, 2012. http://rave.ohiolink.edu/etdc/view?acc_num=kent1341338394.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Waghulde, Harshal B. "Mapping and CRISPR/Cas9 Gene Editing for Identifying Novel Genomic Factors Influencing Blood Pressure." University of Toledo Health Science Campus / OhioLINK, 2016. http://rave.ohiolink.edu/etdc/view?acc_num=mco1470402637.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

"GPER-1 mediates the inhibitory actions of estrogen on adipogenesis in 3T3-L1 cells through perturbation of mitotic clonal expansion." 2012. http://library.cuhk.edu.hk/record=b5549500.

Full text
Abstract:
G蛋白偶聯雌激素受體(GPER,又名GPR30)乃最近於各種動物包括小鼠、大鼠、人類及斑馬魚中發現之新型跨膜雌激素受體。 GPER表達於脂肪組織及多種器官之中,其已被證明能與雌激素結合並介導各式快速反應及基因轉錄。針對GPER於成脂作用中角色之研究將達致對雌激素作用之更全面了解,且GPER亦有望成為治療肥胖症之一種新型標靶。
脂肪發育調控乃一複雜且精妙之排程,而雌激素已被證明能抑制脂肪形成,是故雌激素替代療法可舒減絶經後婦女之脂肪代謝問題。此項研究發現GPER於小鼠腹部脂肪組織及小鼠前脂肪細胞系3T3-L1中均有表達,且其信使RNA量於受誘導之3T3-L1成脂作用中錄得上調。
3T3-L1細胞分化作用會被名為G1之特異性GPER激動劑阻撓於克隆擴增階段(MCE),此即表明GPER有參與成脂調控之可能。通過油紅O染色發現,受G1處理之3T3-L1細胞於分化後所產生之油滴量實比其對照組為低,但此一效果能被特異性GPER小干擾RNA預處理抹除。另外,本研究以流式細胞儀及西方墨點法對細胞週期及細胞週期因子進行分析後,認為激活GPER能觸發對G1期細胞週期停滯之抑制。另一方面,受G1處理並分化中之3T3-L1細胞出現蛋白激酶B磷酸化效應,意味雌激素與GPER結合對成脂作用有雙向調節之可能性。
總而言之,本研究結果斷定GPER能介導雌激素對脂肪組織發育之影響,並為成脂作用之負調節因子,故此,一系列成果將有助肥胖症藥物之研發。
A novel transmembrane estrogen receptor, G-protein coupled estrogen receptor (GPER, also known as GPR30), is recently identified in various animals including mouse, rat, human and zebrafish. GPER is expressed in many organs including fatty tissues, and has been demonstrated to mediate various rapid responses and transcriptional events upon estrogen binding. The study on the role of GPER in adipogenesis would lead to a more comprehensive understanding of estrogenic actions, with the view of identifying novel therapeutic targets for the treatment of obesity.
Regulation of adipose development is a complex and subtly orchestrated process. Estrogen has been shown to inhibit adipogenesis. Estrogen replacement therapy therefore affects fat metabolism in post-menopausal women. In this study, GPER is identified in mouse abdominal fatty tissues; and there is an up-regulation of GPER in the mouse preadipocyte cell line 3T3-L1 during induced adipogenesis.
Differentiation of 3T3-L1 cells is perturbed by the selective GPER agonist G1 at mitotic clonal expansion (MCE), indicating a possible involvement of GPER in the regulation of adipogenesis. By means of Oil-Red-O staining, the production of oil droplets in the G1-treated, differentiated 3T3-L1 cells is shown to be lower than the untreated control; and such effect is reversed by a specific siRNA knockdown of GPER. FACS analysis and Western blot analysis of cell cycle factors during MCE suggest that GPER activation triggers an inhibition of cell cycle arrest at the G1 stage. On the other hand, phosphorylation of Akt in G1-treated differentiating cells implies a possibility of bi-directional estrogenic regulation of adipogenesis via GPER.
To conclude, it is postulated that GPER mediates estrogenic actions in adipose tissues as a negative regulator of adipogenesis. These results provide insights into the development of therapeutic agents for the treatment of human obesity.
Detailed summary in vernacular field only.
Detailed summary in vernacular field only.
Detailed summary in vernacular field only.
Detailed summary in vernacular field only.
Yuen, Man Leuk.
Thesis (Ph.D.)--Chinese University of Hong Kong, 2012.
Includes bibliographical references (leaves 144-166).
Electronic reproduction. Hong Kong : Chinese University of Hong Kong, [2012] System requirements: Adobe Acrobat Reader. Available via World Wide Web.
Abstract also in Chinese.
Abstract (English version) --- p.I
Abstract (Chinese version) --- p.III
Acknowledgement --- p.V
Table of Contents --- p.VII
List of Abbreviations --- p.XI
List of Tables --- p.XII
List of Figures --- p.XIII
Chapter Chapter 1: --- Introduction --- p.1
Chapter 1.1. --- Obesity and adipose tissue --- p.1
Chapter 1.1.1. --- Obesity --- p.1
Chapter 1.1.2. --- Fat deposition --- p.3
Chapter 1.1.3. --- Origin and development of white adipose tissue --- p.5
Chapter 1.2. --- Adipogenesis --- p.7
Chapter 1.2.1. --- Origins of white adipocytes --- p.7
Chapter 1.2.2. --- Signals for adipogenesis --- p.10
Chapter 1.2.3. --- Regulation of gene expression during adipogenesis --- p.12
Chapter 1.2.4. --- Common adipose cell lines --- p.16
Chapter 1.2.5. --- Mechanism of in vitro adipogenesis --- p.21
Chapter 1.2.5.1. --- Growth arrest --- p.23
Chapter 1.2.5.2. --- Mitotic clonal expansion --- p.23
Chapter 1.2.5.3. --- Early and terminal differentiation --- p.24
Chapter 1.3. --- Estrogen and adipogenesis --- p.28
Chapter 1.4. --- G-protein coupled estrogen receptor-1 --- p.33
Chapter 1.4.1. --- General introduction of GPER --- p.33
Chapter 1.4.2. --- Ligands of GPER --- p.36
Chapter 1.4.3. --- Cellular signaling of GPER --- p.38
Chapter 1.4.4. --- Metabolic actions of GPER: A brief introduction --- p.43
Chapter 1.4.5. --- Metabolic actions of GPER on obesity and glucose metabolism --- p.48
Chapter 1.4.6. --- Study objectives --- p.53
Chapter Chapter 2: --- Expression profiles and cellular localization of Gper/GPER in mouse adipose, 3T3-L1 preadipocytes and 3T3-L1 mature adipocytes --- p.54
Chapter 2.1. --- Introduction --- p.54
Chapter 2.1.1. --- Expression and functional roles of GPER in adipose. --- p.55
Chapter 2.1.2. --- Swiss mouse preadipocytes 3T3-L1 --- p.57
Chapter 2.1.3. --- Study objectives --- p.57
Chapter 2.2. --- Materials and Methods --- p.59
Chapter 2.2.1. --- Reagents --- p.59
Chapter 2.2.2. --- Animal tissues --- p.59
Chapter 2.2.3. --- Cell culture --- p.60
Chapter 2.2.4. --- Reverse transcription polymerase chain reaction (RT-PCR) --- p.62
Chapter 2.2.5. --- Quantitative real-time RT-PCR (qRT-PCR) --- p.66
Chapter 2.2.6. --- SDS-PAGE and Western blot analysis --- p.68
Chapter 2.2.7. --- Immunofluorescence assay --- p.69
Chapter 2.2.8. --- Statistical analysis --- p.70
Chapter 2.3. --- Results --- p.71
Chapter 2.3.1. --- Expression of Gper/GPER in mouse visceral adipose tissues --- p.72
Chapter 2.3.2. --- Expression profiles of Gper/GPER in undifferentiated 3T3-L1 preadipocytes and differentiated 3T3-L1 adipocytes --- p.73
Chapter 2.3.3. --- Cellular localization of GPER in undifferentiated 3T3-L1 preadipocytes and differentiated 3T3-L1 adipocytes --- p.75
Chapter 2.4. --- Discussion --- p.76
Chapter Chapter 3: --- Rapid cellular responses induced by GPER activation in 3T3-L1 preadipocytes --- p.78
Chapter 3.1. --- Introduction --- p.78
Chapter 3.1.1. --- Rapid cellular response of estrogen via GPER --- p.79
Chapter 3.1.2. --- Study objectives --- p.81
Chapter 3.2. --- Materials and Methods --- p.82
Chapter 3.2.1. --- Reagents --- p.82
Chapter 3.2.2. --- Cell culture --- p.82
Chapter 3.2.3. --- SDS-PAGE and Western blot analysis --- p.83
Chapter 3.2.4. --- Statistical analysis --- p.84
Chapter 3.3. --- Results --- p.86
Chapter 3.3.1. --- Phosphorylation of p44/42 MAPK after time-dependent activation of GPER by ICI182,780 and G1 --- p.87
Chapter 3.3.2. --- Phosphorylation of p44/42 MAPK after dose-dependent activation of GPER by a combination of chemical agents --- p.88
Chapter 3.4. --- Discussion --- p.89
Chapter Chapter 4: --- GPER activation on cell viability of 3T3-L1 preadipocytes --- p.90
Chapter 4.1. --- Introduction --- p.90
Chapter 4.1.1. --- Cell proliferation mediated by GPER --- p.90
Chapter 4.1.2. --- Study objectives --- p.92
Chapter 4.2. --- Materials and Methods --- p.93
Chapter 4.2.1. --- Reagents --- p.93
Chapter 4.2.2. --- Cell culture --- p.93
Chapter 4.2.3. --- MTT assay for cell viability --- p.94
Chapter 4.2.4. --- Statistical analysis --- p.95
Chapter 4.3. --- Results --- p.96
Chapter 4.3.1. --- Cell viability of 3T3-L1 after dose-dependent activation of GPER by 17β-estradiol, ICI182,780 and G1 --- p.97
Chapter 4.4. --- Discussion --- p.99
Chapter Chapter 5: --- GPER-mediated estrogenic action on lipid accumulation in the mature 3T3-L1 adipocytes --- p.101
Chapter 5.1. --- Introduction --- p.101
Chapter 5.1.1. --- Induction of differentiation in Swiss mouse preadipocyte 3T3-L1 --- p.101
Chapter 5.1.2. --- Study objectives --- p.102
Chapter 5.2. --- Materials and Methods --- p.103
Chapter 5.2.1. --- Reagents --- p.103
Chapter 5.2.2. --- Cell culture --- p.103
Chapter 5.2.3. --- Oil-Red-O staining and measurement of absorbance --- p.105
Chapter 5.2.4. --- Knockdown of Gper/GPER by siRNA --- p.107
Chapter 5.2.5. --- Reverse transcription polymerase chain reaction (RT-PCR) --- p.110
Chapter 5.2.6. --- SDS-PAGE and Western blot analysis --- p.110
Chapter 5.2.7. --- Statistical analysis --- p.110
Chapter 5.3. --- Results --- p.112
Chapter 5.3.1. --- GPER activation on 3T3-L1 differentiation --- p.114
Chapter 5.3.2. --- Knockdown of Gper/GPER in Swiss mouse preadipocyte 3T3-L1 --- p.114
Chapter 5.3.3. --- Phosphorylation of p44/42 MAPK in Gper/GPER-knockdown 3T3-L1 after time-dependent activation of GPER by G1 --- p.117
Chapter 5.3.4. --- Action of drugs on differentiation of Gper/GPER-knockdown 3T3-L1 --- p.117
Chapter 5.4. --- Discussion --- p.118
Chapter Chapter 6: --- Role of GPER in regulating cell cycle progression during mitotic clonal expansion (MCE) stage in adipogenesis of 3T3-L1 --- p.120
Chapter 6.1. --- Introduction --- p.120
Chapter 6.1.1. --- Differentiation stages of Swiss mouse preadipocyte 3T3-L1 --- p.121
Chapter 6.1.2. --- Apoptosis and cell cycle progression --- p.122
Chapter 6.1.3. --- Study objectives --- p.126
Chapter 6.2. --- Materials and Methods --- p.127
Chapter 6.2.1. --- Reagents --- p.127
Chapter 6.2.2. --- Cell culture --- p.127
Chapter 6.2.3. --- Oil-Red-O staining and measurement of absorbance --- p.129
Chapter 6.2.4. --- Trypan blue exclusion assay for cell viability determination --- p.129
Chapter 6.2.5. --- SDS-PAGE and Western blot analysis --- p.131
Chapter 6.2.6. --- Flow cytometry for analysis of cell cycle progression --- p.132
Chapter 6.2.7. --- Statistical analysis --- p.133
Chapter 6.3. --- Results --- p.134
Chapter 6.3.1. --- Temporal effect of GPER activation on differentiation progress of Swiss mouse preadipocyte 3T3-L1 --- p.137
Chapter 6.3.2. --- Effect of GPER activation on cell viability during adipogenesis --- p.139
Chapter 6.3.3. --- Effect of GPER activation on apoptosis during adipogenesis --- p.139
Chapter 6.3.4. --- Effect of GPER activation on cell cycle distribution during induced adipogenesis --- p.140
Chapter 6.3.5. --- Effect of GPER activation on expression of cell cycle markers during induced adipogenesis --- p.142
Chapter 6.3.6. --- Activation of PI3K/Akt pathway by GPER stimulation during induced adipogenesis --- p.143
Chapter 6.4. --- Discussion --- p.144
Chapter Chapter 7: --- Conclusions and Future Perspectives --- p.148
References --- p.155
APA, Harvard, Vancouver, ISO, and other styles
5

Peyton, Candace Ann. "Involvement of epidermal growth factor receptor (EGFR) signaling in estrogen inhibition of oocyte maturation mediated through G protein-coupled estrogen receptor 1 (GPER) in zebrafish (Danio rerio)." Thesis, 2010. http://hdl.handle.net/2152/ETD-UT-2010-05-1083.

Full text
Abstract:
Oocyte maturation (OM) in teleosts is under precise hormonal control by estrogens and progestins. We show here that estrogens activate an epidermal growth factor receptor (EGFR) signaling pathway through the G protein-coupled estrogen receptor (GPER) to maintain meiotic arrest of full-grown zebrafish (Danio rerio) oocytes in an in vitro germinal vesicle breakdown (GVBD) bioassay. A GPER- specific agonist decreased OM and a GPER-specific antagonist increased spontaneous OM, whereas specific nuclear estrogen receptor (ERα and ERβ) agonists did not affect OM, which suggests the inhibitory action of estrogens on OM are solely mediated through GPER. Furthermore, a peptide-bound estrogen, which cannot enter the oocyte, decreased GVBD, showing that these estrogen actions are mediated through a membrane receptor. Treatment of oocytes with actinomycin D, a transcription inhibitor, did not block the inhibitory effects of estrogens on OM, indicating that estrogens act via a nongenomic mechanism to maintain oocyte meiotic arrest. EGFR mRNA was detected in denuded zebrafish oocytes by reverse transcription polymerase chain reaction (RT-PCR). Therefore, the potential role of transactivation of EGFR in estrogen inhibition of OM was investigated. The matrix metalloproteinase inhibitor, ilomastat, which prevents the release of heparin-bound epidermal growth factor (HB-EGF), increased spontaneous OM. Moreover, specific EGFR1 (ErbB1) inhibitors and inhibitors of extracellular-related kinase 1 and 2 (ERK1/2) increased spontaneous OM. Previously, estrogens have been shown to increase 3’-5’-cyclic adenosine mono phosphate (cAMP) levels through GPER in zebrafish oocytes during meiotic arrest. Taken together these present results suggest that estrogens also act through GPER to maintain meiotic arrest through a second signaling pathway involving transactivation of EGFR and activation of ERK 1 and 2.
text
APA, Harvard, Vancouver, ISO, and other styles
6

"Expression of G protein-coupled estrogen receptor (GPER) and its effects on P2Y receptor-mediated Ca²⁺ signalling and cytokine secretion in human bronchial epithelia." 2014. http://repository.lib.cuhk.edu.hk/en/item/cuhk-1290692.

Full text
Abstract:
The airway epithelium plays a central role in respiratory physiology through its transport and immunological functions. Our previous study suggested that P2Y receptors are expressed in airway epithelia and play a significant role in regulating transepithelial ion transport. P2Y receptors belong to the family of purinergic receptors, which can be stimulated by nucleotides such as UTP and UDP. P2Y receptors are G protein-coupled receptors and classically signal through G[subscript q], resulting in an increase in intracellular Ca²⁺ concentration ([Ca²⁺]ᵢ) and thereby in the activation of Ca²⁺-dependent ion channels and downstream signalling pathway(s). Furthermore, P2Y receptors are involved in asthmatic inflammation.
Estrogen (or E₂) is an important hormone in human physiology. In addition to the classical nuclear hormone receptors ERα and ERβ, a novel estrogen receptor, G protein-coupled estrogen receptor (GPER), was recently identified and found to be involved in both rapid signalling and transcriptional regulations. The action of GPER is unclear, but it has been implicated in mediating anti-inflammatory responses.
In our experiments, both human bronchial epithelial cell line, 16HBE14o-, and primary normal human bronchial epithelial cells expressed GPER at mRNA and protein levels, as demonstrated by RT-PCR and western blotting, respectively. ERα and ERβ expression were also detected at mRNA and protein level. Expression of GPER receptors was localized in the human bronchial epithelial cells by immunofluorescence staining and western blotting of fractionated cell lysates.
[Ca²⁺]ᵢ induced by nucleotides were monitored by calcium imaging technique using MetaFluor fluorescence ratio imaging system. Stimulation of epithelial cells with E₂ or with the specific agonist of GPER, G1, rapidly attenuated a UDP-, UTP- or ATPyS- evoked increase in [Ca²⁺]ᵢ in both 16HBE14o- cell line and primary cells. This inhibitory effect of E₂ and G1 were concentration dependent, while this effect was reversed by GPER specific antagonist, G15. To examine the effect of E₂ and G1 on UDP-activated intracellular Ca²⁺ release and influx, the epithelia were exposed to nominally Ca²⁺ -free solution in the presence or absence of G1 or E₂, and then stimulated with UDP. Subsequently, Ca²⁺ was added to the perfusate. Both E₂ and G1 could inhibit UDP-induced Ca²⁺ release. However, only E₂ but not G1 could inhibit UDP-induced Ca²⁺ influx.
E₂ or G1 inhibited the secretion of two pro-inflammatory cytokines, interleukin (IL)-6 or IL-8, in cells stimulated by different nucleotides or the cationic protein, poly-L-arginie, as quantified by ELISA. CFP-Epac-YPF, an Epac-based polypeptide FRET reporter was used to monitor the real-time cAMP changes in 16HBE14o- cells. Both G1 and E₂ induced an increase in cAMP production. The transepithelial chloride (Cl⁻) secretion was measured using short circuit current technique in cells grown on permeable support. Cl⁻ secretion induced by apical UDP was partially inhibited by G1 in a concentration dependent manner.
Our results provide the first evidence that human bronchial epithelia express GPER, which interact with the P2Y receptor-mediated calcium signalling pathway and cytokine secretion. Moreover, the anti-inflammatory role of GPER may be due to its opposing effect on the pro-inflammatory pathway activated by the P2Y receptors in inflamed airway epithelia.
气道上皮具有调节运输以及参与免疫反应等功能,在呼吸生理学研究中有着十分关键的意义。我们曾经的研究发现P2Y受体在气道上皮中表达并调节上皮细胞离子运输过程。P2Y受体属于嘌呤受体,可被三磷酸尿苷(UTP),二磷酸尿苷(UDP)等核苷酸激活。同时,P2Y受体也是一类G蛋白偶联受体,可通过活化G[subscript q]蛋白调控细胞内钙离子浓度而激活钙依赖性离子通道及其他下游信号通路。此外P2Y受体还参与哮喘炎症的调控。
雌激素(或雌二醇,E₂)是人体一类十分重要的激素。除传统的核受体ERα与ERβ外,一类新型雌激素受体GPER已被发现和鉴定。GPER属于G蛋白偶联受体,可同时参与转录调控和非基因依赖的快速信号调节。尽管具体机理尚不明确,但研究发现GPER可介导抗炎症反应。
实验结果显示,在支气管上皮细胞株16HBE14o-和原代人支气管上皮细胞中GPER都被检测到基因和蛋白水平的表达。GPER在人支气管上皮细胞中的定位也通过免疫荧光染色(immunofluorescence)和亚细胞组分蛋白质印迹(western blot of fractionated cells)得到鉴定。
本研究中,荧光显微技术(fluorescence microscopy)被用于测定核苷酸介导的细胞内钙离子浓度([Ca²⁺]ᵢ)。在16HBE14o- 和原代培养人支气管上皮细胞中,E₂和GPER特异性激动剂G1都可抑制核苷酸介导的 [Ca²⁺]ᵢ增加,且这种抑制作用呈浓度依赖。GPER特异性拮抗剂G15可抵消G1的抑制作用。进一步研究表明,E₂和G1都可抑制UTP诱导的胞内钙库释放,然而只有E₂抑制UTP诱导的胞外钙离子内流。
除钙离子调节外,E₂和G1还可抑制支气管上皮细胞中核苷酸或聚精氨酸(poly-L-arginine)刺激介导的两种促炎症细胞因子,白介素6(IL-6)和白介素8(IL-8)的分泌。酶联免疫法(ELISA)被用于细胞因子的定量。同时,CFP-Epac-YPF作为一类多肽荧光共振能量转移(FRET)探针被转染入16HBE14o- ,探测细胞内腺苷-3',5'-环化一磷酸(cAMP)的实时变化。结果显示在人支气管上皮细胞中E₂和G1都可引导cAMP生成。此外,我们使用短路电流(short-circuit current, Isc)技术测定单层上皮细胞的氯离子(Cl⁻)分泌,并发现人支气管上皮顶膜面UDP诱导的Cl⁻ 分泌可被G1部分抑制,且抑制效果呈浓度依赖。
本研究首次证明GPER表达于人支气管上皮, 且激活GPER对P2Y受体介导的钙离子信号通路以及细胞因子生成起到抑制作用。这些结果表明在气道炎症反应中,GPER可通过反向调节P2Y受体激活的促炎症作用,达到抗炎症的效果。
Hao, Yuan.
Thesis (Ph.D.)--Chinese University of Hong Kong, 2014.
Includes bibliographical references (leaves 187-211).
Abstracts also in Chinese.
Title from PDF title page (viewed on 03, November, 2016).
Detailed summary in vernacular field only.
Detailed summary in vernacular field only.
Detailed summary in vernacular field only.
Detailed summary in vernacular field only.
Detailed summary in vernacular field only.
Detailed summary in vernacular field only.
APA, Harvard, Vancouver, ISO, and other styles
7

Rago, Vittoria, Diego Sisci, and Amalia Carpino. "Identificazione del recettore strogenico GPER (G-protein-couple estrogen receptor) nella ghiandola prostatica umana: valutazione del GPER nei tessuti prostateci benigni e neoplastici." Thesis, 2015. http://hdl.handle.net/10955/1549.

Full text
Abstract:
Dottorato di Ricerca in Biochimica Cellulare ed Attività dei Farmaci in Oncologia, XXVIII Ciclo,a.a. 2015-2016
Gli estrogeni sono coinvolti nella crescita, differenziazione e patogenesi della prostata umana attraverso la mediazione dei classici recettori estrogenici (ER e ER). D'altro canto, il meccanismo non genomico degli estrogeni sembra esercitare un ruolo importante su segnalazioni di percorsi che dirigono o indirettamente modulano l'espressione genica. In questo scenario, il G protein-coupled receptor, GPER (precedentemente chiamato GPR30), è stato implicato nella mediazione rapida degli eventi trascrizionali in risposta agli estrogeni. Alcuni studi supportano l'ipotesi che GPER rappresenta un recettore estrogeno-sensibile e la sua iper-espressione sembra essere fondamentale i diverse patologie neoplasiche. L’espressione di GPER è stata recentemente evidenziata in alcuni tessuti riproduttivi umani, ma la sua espressione a livello prostatico è ancora sconosciuta. In questo studio, abbiamo valutato, l’espressione di GPER in 5 pazienti affetti da patologie prostatiche non neoplastiche, e in 50 pazienti affetti da adenocarcinoma, mediante analisi immunoistochimica e Western blot.. Le aree normali della prostata benigna hanno mostrato una forte immunoreattività del GPER nel citoplasma delle cellule epiteliali basali insieme ad una debole colorazione nel citoplasma delle cellule stromali. Nessuna immunocolorazione è stata invece osservata nelle cellule epiteliali luminali secretorie. L’analisi immunoistochimica ha evidenziato l’espressione cellulare di GPER in tutti i campioni di adenocarcinoma esaminati ma con una variabilità correlata alle diverse architetture delle cellule tumorali (Gleason patterns). Le regioni che presentavano lesioni pre-neoplastiche HGPIN (high-grade prostatic intraepithelial neoplasia) hanno evidenziato una intensa immunoreattività per il recettore, mentre nelle aree tumorali la positività al GPER è stata correlata ai “Gleason patterns” e valutata con il metodo di Allred. Una intensa immunoposititvità al GPER è stata evidenziata nelle aree tumorali Gleason pattern 2 e pattern Gleason 3 (leggermente ridotta in queste ultime), mentre debolmente colorate apparivano le aree con Gleason pattern 4. L’analisi Western blot degli estratti proteici benigni e tumorali ha confermato questo risultato. È stato inoltre osservato un aumento delle forme fosforilate dei livelli di Akt e di CREB nei campioni di pazienti affetti da adenocarcinoma scarsamente differenziato rispetto alle altre categorie. In conclusione, nel presente lavoro, per la prima volta, abbiamo identificato GPER nelle cellule basali epiteliali della prostata umana non neoplastica, con una diversa localizzazione rispetto ai classici recettori estrogenici. Abbiamo inoltre evidenziato l'espressione di GPER nelle cellule di adenocarcinoma prostatico ma con una modulazione della sua intensità dipendente dall’organizzazione delle cellule neoplastiche. La immunoreattività al GPER appare quindi inversamente correlata al grado di differenziazione tumoral
Università della Calabria
APA, Harvard, Vancouver, ISO, and other styles
8

Thangavel, Hariprasad, Bartolo Gabriele, Giovanni Sindona, and Anna Napoli. "Advanced mass spectrometry-based strategies for the isolation and characterization of G protein-coupled estrogen receptor 1(GPR)." Thesis, 2014. http://hdl.handle.net/10955/1269.

Full text
Abstract:
Dottorato di Ricerca in Organic Materials of Pharmaceutical interest (OMPI) Ciclo XXVI, a.a. 2011-2014
Estrogen signaling plays a vital role in breast, ovarian and endometrial cancers. The actions of estrogen are mainly mediated by classical estrogen receptors, ERα and ERβ that belongs to the nuclear receptor superfamily. In recent years, a class of membrane-associated estrogen receptors are found to mimic the functions of classical ERs, including genomic as well as non-genomic signaling. These non-genomic signaling events include pathways that are usually thought of as arising from transmembrane growth factor receptors and G protein-coupled receptors (GPCRs). GPCRs belong to a superfamily of cell surface signaling proteins. GPCRs represent the most significant family of validated pharmacological targets in medical biology. A member of the GPCR family, named GPER, mediates rapid biological responses to estrogen in diverse normal and cancer cells, as well as transformed cell types. The identification and characterization of GPER will lead to understand the mechanisms underlying complex biological pathways and identify potentially new drug targets. Here, we proposed a novel gel-free method to isolate and enrich GPER from crude lysate using home-made hydroxyapatite column (HTP). The HTP eluate was subjected to cellulose acetate (CA) filteration, followed by on-membrane protein digestion with different proteases and analyzed by MALDI MS. GPER was identified by peptide mass fingerprinting (PMF) after intensive data analysis. Sequence analysis reports 3 potential N-glycosylation in GPER. We manually validated 2 out of 3 glycosylation sites in GPER from the obtained MS/MS data and also validated the glycan moieties predicted by Glycomod. This approach is the first of its kind to identify GPER and characterize post-translational modifications (PTMs) by MS-based proteomic analysis. The proposed method is simple, robust and unique with great reproducibility. Finally, we designed and synthesized polymer nanoparticles (NPs) in an effort to capture GPER with high affinity and selectivity from crude lysate. PNIPAm-based NPs were synthesized by a free radical precipitation polymerization method with no control over the functional monomer sequence. The NP binding affinity was evaluated against both truncated-GPER (short peptide epitopes) and GPER (whole protein). As the NPs were designed with complementary functionality against the peptides/protein, the NPspeptide/ protein binding will be through multipoint interactions. The initial qualitative results obtained by immunoblotting analysis revealed interesting hints on GPER’s competitive affinity towards NPs when probed against multiple antibodies. We anticipate to use this strategy as a sample purification step prior to MS-based proteomic analysis
Università della Calabria
APA, Harvard, Vancouver, ISO, and other styles
9

Bernardino, Ana Carolina de Matos. "Evaluation of the antioxidant action of the G protein-coupled estrogen receptor." Master's thesis, 2020. http://hdl.handle.net/10400.6/10803.

Full text
Abstract:
The brain is characterized by a high metabolism and contains several easily oxidizable substances such as amines and lipids, resulting in exposure to high levels of oxidative stress. In Parkinson's disease (PD), oxidative stress has been shown to be correlated with lipid peroxidation, inflammation, mitochondrial dysfunction and aggregation of a-synuclein (asyn). This demonstrates that oxidative stress can be one of the triggers of Parkinson's disease, as it is capable of inducing a series of pathogenic mechanisms characteristic of the disease, contributing to its progression. In this sense, the identification of mechanisms that help reducing oxidative stress may be an interesting strategy for controlling the progression of the disease. Since 17ß-estradiol exerts neuroprotective functions and has proved beneficial effects on several mechanisms such as neuroinflammation, excitotoxicity, among others, we assessed whether the selective activation of the G protein-coupled estrogen receptor (GPER), characterized by being involved in rapid non-genomic actions of 17ßestradiol, can exert a neuroprotective effect associated with the modulation of oxidative stress. With this objective, we developed an in vivo study with mice injected with 6-OHDA, which were later submitted to subcutaneous or intranasal treatment with the GPER agonist, G1. We evaluated how the selective activation of the receptor can contribute to the reversion of oxidative stress. To this end, several behavioral tests were performed to evaluate motor function, such as Grip Test, Rotarod and Open Field Test, and relative mRNA levels of antioxidant enzymes were measured by real-time PCR (RT-PCR). From the behavioral tests, it was possible to conclude that the 6-OHDA-injection was not capable of affecting motor behavior, since the results obtained with the Rotarod test, and the total distance travelled obtained with the Open field Test did not present significant differences. On the other hand, it was possible to observe that the parameters related with anxious behavior were increased in animals injected with 6-OHDA, when compared with the control group. Therefore, it can be concluded that the toxin had no effect at the level of motor behavior, but induced changes in non-motor domains. Regarding the expression of antioxidant enzymes, although not significant, an increase in the mRNA levels of Gpx4 and Nrf2 was observed in 6-OHDAinjected mice. This increase suggests a protective mechanism aiming to limit oxidative stress. However, further studies are needed to confirm this hypothesis. Our results have shown effects exercised by the G1, when administered by the two delivery approaches. However, it was not possible to conclude whether the two types of G1 delivery have an antioxidant effect in the presence of a dopaminergic insult. In this sense, further studies would be necessary to confirm whether GPER activation is capable of modulating oxidative stress and whether this effect is related to its currently recognized neuroprotective effects.
O cérebro caracteriza-se por apresentar um elevado metabolismo, e contém várias substâncias facilmente oxidáveis, tais como aminas e lípidos, o que resulta numa exposição a elevados níveis de stress oxidativo. Foi demonstrado que na doença de Parkinson (DP), o stress oxidativo está correlacionado com a peroxidação lipídica, inflamação, disfunção mitocondrial e agregação da a-synucleína (a-syn). Isto demonstra que o stress oxidativo pode ser um dos desencadeadores da doença de Parkinson, por ser capaz de induzir uma série de mecanismos patogénicos característicos da doença, contribuindo de forma crucial para a sua progressão. Neste sentido, a identificação de mecanismos que ajudem a reduzir o stress oxidativo podem ser estratégias interessantes para o controlo da progressão da doença. Uma vez que o 17ß-estradiol foi classificado como neuroprotetor e já demonstrou efeitos benéficos em diversos mecanismos como neuroinflamação, excitotoxicidade, entre outros, fomos avaliar se a ativação seletiva do recetor de estrogénios acoplado à proteína G (GPER), caracterizado por estar envolvido em ações não genómicas rápidas do 17ßestradiol, pode exercer um efeito neuroprotetor associado à modulação do stress oxidativo na DP. Com este objetivo, desenvolvemos um estudo in vivo com murganhos injetados com 6-OHDA, que foram, posteriormente, submetidos a tratamento subcutâneo ou intranasal com um agonista do recetor, o G1. Assim, avaliámos de que forma a ativação seletiva do recetor pode contribuir para a reversão do stress oxidativo. Para isso, foram efetuados vários testes comportamentais para avaliar a função motora, como o Grip Test, o Rotarod e o Open Field Test, e foram medidos os níveis de mRNA de enzimas antioxidantes, por PCR em tempo real (RT-PCR). A partir dos testes comportamentais, foi possível concluir que a injeção da toxina não afetou o comportamento motor uma vez que os resultados obtidos no Rotarod, e distância total percorrida obtida no Open Field Test não mostraram diferenças significativas. Por outro lado, foi possível observar que a injeção com 6-OHDA aumentou os parâmetros relacionados com o comportamento ansioso. Desta forma, é possível concluir que a toxina não exerceu efeito ao nível do comportamento motor, porém, induziu alterações a nível não-motor. Relativamente à expressão das enzimas antioxidantes, observou-se um aumento, ainda que sem significância estatística, dos níveis de mRNA da Gpx4 e do Nrf2 em animais injetados com 6-OHDA. Este aumento, pode querer evidenciar um mecanismo de proteção desencadeado por estas enzimas para lidar com o stress oxidativo. No entanto, mais estudos seriam necessários para conseguir comprovar esta hipótese. Os nossos resultados evidenciaram efeitos exercidos pelo G1, quando entregue pelos dois tipos de administração. No entanto, não foi possível concluir se os dois tipos de entrega do G1 têm um efeito antioxidante na presença de um insulto dopaminérgico. Neste sentido, mais estudos seriam necessários para perceber se a ativação do GPER é capaz de modular o stress oxidativo e, se este efeito está relacionado com os seus efeitos neuroprotetores atualmente reconhecidos.
APA, Harvard, Vancouver, ISO, and other styles
10

Albanito, Lidia, Marcello Maggiolini, and Benedictis Giovanna De. "The G protein-coupled receptor GPR30 mediates estrogen signaling in cancer cells." Thesis, 2013. http://hdl.handle.net/10955/349.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "G-Protein Coupled Estrogen Receptor (GPER)"

1

Sharma, Geetanjali, and Eric R. Prossnitz. "G-Protein-Coupled Estrogen Receptor (GPER) and Sex-Specific Metabolic Homeostasis." In Sex and Gender Factors Affecting Metabolic Homeostasis, Diabetes and Obesity, 427–53. Cham: Springer International Publishing, 2017. http://dx.doi.org/10.1007/978-3-319-70178-3_20.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Thekkumkara, Thomas, Russell Snyder, and Vardan T. Karamyan. "Competitive Binding Assay for the G-Protein-Coupled Receptor 30 (GPR30) or G-Protein-Coupled Estrogen Receptor (GPER)." In Methods in Molecular Biology, 11–17. New York, NY: Springer New York, 2016. http://dx.doi.org/10.1007/978-1-4939-3127-9_2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Arnatt, Christopher K., and Yan Zhang*. "Chapter 7. A Nuclear G Protein-coupled Estrogen Receptor, GPER. Homology Modeling Studies Toward Its Ligand-binding Mode Characterization." In Drug Discovery, 117–37. Cambridge: Royal Society of Chemistry, 2012. http://dx.doi.org/10.1039/9781849735353-00117.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Filardo, Edward J., Jeffrey A. Quinn, and C. Thomas Graeber. "Evidence Supporting a Role for Gpr30, an Orphan Member of the G-Protein-Coupled Receptor Superfamily, in Rapid Estrogen Signaling." In The Identities of Membrane Steroid Receptors, 139–46. Boston, MA: Springer US, 2003. http://dx.doi.org/10.1007/978-1-4615-0339-2_17.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Zhang, Hong-Bing, Yao Wang, and Bing Wang. "The Research Advances in G-Protein-Coupled Estrogen Receptor." In Estrogens - Recent Advances [Working Title]. IntechOpen, 2022. http://dx.doi.org/10.5772/intechopen.105822.

Full text
Abstract:
Estrogen binds to the typical estrogen receptor (ER) ERα or ERβ and is translocated to the nucleus, where it binds directly to the estrogen response element of the target gene to induce transcription and regulate gene expression, and the whole process is completed in several hours to several days. The G protein-coupled estrogen receptor (GPER), a type that is structurally distinct from typical ERα and ERβ, rapidly induces most non-genomic effects within seconds to minutes. GPER regulates cell growth, migration, and programmed cell death in a variety of tissues and has been associated with the progression of estrogen-associated cancers. Here, the characteristics, cell signal transduction, and the latest research progress of GPER in estrogen-associated tumors and retinal diseases are reviewed.
APA, Harvard, Vancouver, ISO, and other styles
6

Torres-Reveron, Annelyn, Wayne G. Brake, and Teresa A. Milner. "Estrogen Receptor Distribution in the Hippocampus and Prefrontal Cortex." In Estrogens and Memory, 11–23. Oxford University Press, 2020. http://dx.doi.org/10.1093/oso/9780190645908.003.0002.

Full text
Abstract:
This chapter presents anatomical evidence for the distribution of estrogen receptors in the brain. First, the chapter presents a brief discussion of the historical findings that led to the discovery of nuclear and extranuclear estrogen receptors in the brain. A distribution pattern for each one of the receptors, estrogen receptor alpha (ERα‎), estrogen receptor beta (ERβ‎), and G-protein coupled estrogen receptor 1 (GPER1), is presented in sequential subsections. The discussion focuses on the hippocampus and prefrontal cortex areas, as these are largely involved in memory and cognitive behaviors, further discussed in other chapters in this book. In addition, co-localization studies with other neurotransmitter systems and molecules important for the functional activity of estrogen receptors is reviewed.
APA, Harvard, Vancouver, ISO, and other styles
7

Shi, Haifei, Shiva Priya Dharshan Senthil Kumar, and Xian Liu. "G Protein-Coupled Estrogen Receptor in Energy Homeostasis and Obesity Pathogenesis." In Progress in Molecular Biology and Translational Science, 193–250. Elsevier, 2013. http://dx.doi.org/10.1016/b978-0-12-386933-3.00006-6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Srivastava, Deepak P., Katherine J. Sellers, and Peter D. Evans. "Rapid Modulation of Spinogenesis by Estradiol in the Neocortex." In Estrogens and Memory, 48–68. Oxford University Press, 2020. http://dx.doi.org/10.1093/oso/9780190645908.003.0005.

Full text
Abstract:
This chapter explores our recent advances in our understanding of how estrogens can modulate spinogenesis within the cortex and its relevance for estrogenic-regulation of cognition. It describes how estrogens, including 17β‎-estradiol and estrogen receptor modulators, rapidly modify dendritic spine density concurrently with influencing cognitive behaviors that require cortical processing. Furthermore, it reviews the evidence that these effects are not limited to female animals but may represent a relevant mechanism in the male brain. This chapter will also explore the emerging role for a novel estrogen receptor, G-protein estrogen receptor (GPER), in mediating the rapid effects of estrogens on dendritic spines. Finally, the chapter also reviews the potential molecular mechanisms that underlie rapid estrogenic signaling, linking this signaling to the modulation of spinogenesis, which may ultimately provide a cellular model by which estrogens can produce long-lasting changes in neural circuitry.
APA, Harvard, Vancouver, ISO, and other styles
9

Li, XY, Y. Lu, HY Sun, MY Liu, J. Yang, and G. Ning. "G Protein Coupled Receptor 48 Up-Regulates Estrogen Receptor Alpha Expression Via cAMP/PKA Signaling in Male Reproductive Tract." In The Endocrine Society's 92nd Annual Meeting, June 19–22, 2010 - San Diego, P1–321—P1–321. Endocrine Society, 2010. http://dx.doi.org/10.1210/endo-meetings.2010.part1.p7.p1-321.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Conference papers on the topic "G-Protein Coupled Estrogen Receptor (GPER)"

1

Jala, Venkatakrishna R., Haribabu Bodduluri, Brandie Radde, and Carolyn M. Klinge. "Abstract 4548: The role of GPR30/G-protein coupled estrogen receptor (GPER) in lung cancer development." In Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL. American Association for Cancer Research, 2011. http://dx.doi.org/10.1158/1538-7445.am2011-4548.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Castillo, Maryann, Angelique M. Wimbley, Jacob J. Mayfield, Jenifer C. Lascano, and Kevin D. Houston. "Abstract 1305: Activation of G-protein coupled estrogen receptor (GPER) inhibits ELT-3 uterine leiomyoma cell proliferation." In Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC. American Association for Cancer Research, 2013. http://dx.doi.org/10.1158/1538-7445.am2013-1305.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Lv, Xiangmin, Guohua Hua, Chunbo He, John S. Davis, and Cheng Wang. "Abstract B82: G-protein coupled estrogen receptor (GPER) agonist G-1 inhibits growth of human granulosa cell tumor cells via blocking microtubule assembly." In Abstracts: AACR Special Conference on Advances in Ovarian Cancer Research: From Concept to Clinic; September 18-21, 2013; Miami, FL. American Association for Cancer Research, 2013. http://dx.doi.org/10.1158/1078-0432.ovca13-b82.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Wang, Cheng, Chao Jiang, Xiangmin Lv, Lan Fu, and John S. Davis. "Abstract 3920: Off-target effects of the putative G-protein coupled estrogen receptor 1 (GPER) agonist G1 in ovarian and breast cancer cells." In Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL. American Association for Cancer Research, 2012. http://dx.doi.org/10.1158/1538-7445.am2012-3920.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Natale, Chris, Tina Garyantes, and Todd Ridky. "Abstract 1225: LNS8801: A novel, enantiomerically pure, small molecule agonist of the G protein-coupled estrogen receptor (GPER) for the treatment of cancer." In Proceedings: AACR Annual Meeting 2021; April 10-15, 2021 and May 17-21, 2021; Philadelphia, PA. American Association for Cancer Research, 2021. http://dx.doi.org/10.1158/1538-7445.am2021-1225.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Meng, Ran, Ying Xiong, Yuan Zhao, Yan Wang, Tao Tao, Qiqi Wang, Hua Liu, et al. "Abstract 1149: Interaction with NHERF1 enhances protein stability of G protein-coupled estrogen receptor." In Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-1149.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Natale, Christopher, Jinyang Li, Tzvete Dentchev, Brian Capell, John Seykora, Ben Stanger, and Todd Ridky. "Abstract B21: Pharmacologic activation of G protein-coupled estrogen receptor inhibits pancreatic ductal adenocarcinoma." In Abstracts: AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; September 6-9, 2019; Boston, MA. American Association for Cancer Research, 2019. http://dx.doi.org/10.1158/1538-7445.panca19-b21.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Gohar, Eman, Ravneet Singh, Rawan Almutlaq, and Victoria Nasci. "G Protein-coupled Estrogen Receptor 1 and Pregnancy Confer Protection against Hypertension in Aged Female Mice." In ASPET 2023 Annual Meeting Abstracts. American Society for Pharmacology and Experimental Therapeutics, 2023. http://dx.doi.org/10.1124/jpet.122.140420.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Deming-Halverson, Sandra L., Carl Graeber, Jason Machan, Edmond Sabo, Wei Zheng, and Edward J. Filardo. "Abstract 2272: Association of G-protein estrogen receptor (GPER) in primary breast cancer in Caucasian and Black women by tumor immunophenotype and menopausal status." In Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-2272.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Leeb-Lundberg, FLM, M. Sjöström, S. Broselid, K. Jirström, M. Belting, B. Olde, K. Lövgren, et al. "P4-09-02: G Protein-Coupled Estrogen Receptor 1 Positively Correlates with Estrogen Receptor a Expression and Increased Distant Disease-Free Survival of Breast Cancer Patients." In Abstracts: Thirty-Fourth Annual CTRC‐AACR San Antonio Breast Cancer Symposium‐‐ Dec 6‐10, 2011; San Antonio, TX. American Association for Cancer Research, 2011. http://dx.doi.org/10.1158/0008-5472.sabcs11-p4-09-02.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography