Next Article in Journal
Is It Possible to Find an Antimicrobial Peptide That Passes the Membrane Bilayer with Minimal Force Resistance? An Attempt at a Predictive Approach by Molecular Dynamics Simulation
Next Article in Special Issue
Advanced Maternal Age Impairs Uterine Artery Adaptations to Pregnancy in Rats
Previous Article in Journal
Relative Cation-Anion Diffusion in Alkyltriethylammonium-Based Ionic Liquids
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

G-Protein-Coupled Estrogen Receptor Expression in Rat Uterine Artery Is Increased by Pregnancy and Induces Dilation in a Ca2+ and ERK1/2 Dependent Manner

by
Teresa Tropea
1,2,
Damiano Rigiracciolo
3,
Milena Esposito
4,
Marcello Maggiolini
3 and
Maurizio Mandalà
4,*
1
Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, UK
2
Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary’s Hospital, Manchester M13 9WL, UK
3
Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
4
Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(11), 5996; https://doi.org/10.3390/ijms23115996
Submission received: 26 April 2022 / Revised: 23 May 2022 / Accepted: 23 May 2022 / Published: 26 May 2022
(This article belongs to the Special Issue Molecular Research of Vascular Aspects in Pregnancy)

Abstract

:
Increasing levels of estrogens across gestation are partly responsible for the physiological adaptations of the maternal vasculature to pregnancy. The G protein-coupled estrogen receptor (GPER) mediates acute vasorelaxing effects in the uterine vasculature, which may contribute to the regulation of uteroplacental blood flow. The aim of this study was to investigate whether GPER expression and vasorelaxation may occur following pregnancy. Elucidation of the functional signalling involved was also investigated. Radial uterine and third-order mesenteric arteries were isolated from non-pregnant (NP) and pregnant rats (P). GPER mRNA levels were determined and—concentration–response curve to the GPER-specific agonist, G1 (10−10–10−6 M), was assessed in arteries pre-constricted with phenylephrine. In uterine arteries, GPER mRNA expression was significantly increased and vasorelaxation to G1 was significantly enhanced in P compared with NP rats. Meanwhile, in mesenteric arteries, there was a similar order of magnitude in NP and P rats. Inhibition of L-type calcium channels and extracellular signal-regulated kinases 1/2 significantly reduced vasorelaxation triggered by G1 in uterine arteries. Increased GPER expression and GPER-mediated vasorelaxation are associated with the advancement of gestation in uterine arteries. The modulation of GPER is exclusive to uterine arteries, thus suggesting a physiological contribution of GPER toward the regulation of uteroplacental blood flow during pregnancy.

1. Introduction

Normal pregnancy is associated with physiological adaptations of the maternal vasculature and involves significant changes to both systemic and tissue-specific vascular beds [1,2]. These adaptations are widely triggered by a decrease in vascular tone and an increase in uterine blood flow, which is essential to sustaining sufficient uteroplacental perfusion required by the developing fetus throughout pregnancy [3,4].
Amongst the main mechanisms, vasoactive molecules released from the endothelium, such as nitric oxide (NO) [4,5,6], prostacyclin [7,8,9] and hyperpolarizing factor [10,11], play a key role in modulating pregnancy-associated vascular tone and reactivity, which may be regulated by the mobilization of intracellular calcium (Ca2+) [9,11] and activation of protein kinases, like extracellular signal-regulated kinases (ERK) [9]. Sex hormones, including estrogens, can profoundly influence vascular functions [12,13]. In this regard, it has been shown that natural estrogens stimulate the production of NO and other endothelium-derived factors [13,14,15,16], and L-type Ca2+ channels may be a target of acute estrogenic vasodilatory effects during pregnancy [17].
Estrogen-induced vasodilation has the beneficial role of maintaining arterial health [18,19], with a potentially different degree of vascular effects according to tissue-specific vascular beds. In particular, estrogens exhibit potent vasodilatory properties reaching their greatest effects on the reproductive tissues, where systemic infusion of these hormones increases uterine blood flow up to 10-fold in non-pregnant ewes [20].
Previous studies have established that estrogens exert a functional role in the vascular adaptation to pregnancy and regulation of uterine blood flow [21,22] through binding to the estrogen receptor (ER), Erα, and ERβ [16,23,24]. In addition to the action elicited through these receptors, estrogens activate the G protein-coupled estrogen receptor (GPER) signalling leading to acute vascular effects [25,26]. Notably, we have demonstrated that GPER is expressed in rat uterine arteries, and its activation by the specific agonist G1 induces endothelium-dependent vasorelaxation through the NO-cyclic guanosine monophosphate (cGMP) pathway [27]. Moreover, previous evidence has shown that -plasma levels of estrogens increase progressively in human pregnancy [28] as well as in pregnant rats [29].
However, it is currently unknown whether functional GPER expression changes during pregnancy in the uterine and systemic circulation. Here, we ascertained that pregnancy modulates GPER expression and GPER-mediated vasorelaxation only in uterine arteries. In addition, we investigated mechanisms underlying GPER vasorelaxation.

2. Results

2.1. GPER mRNA Levels Change following Gestational Age in Uterine Arteries

In uterine arteries, mRNA levels of GPER were significantly higher in P7 rats, and this increase was even greater in P14 compared with NP rats (p < 0.05 P7 and P14 vs. NP; Figure 1A). A trend in the increase of GPER expression was assessed by qPCR in uterine arteries following pregnancy progression (p= 0.06, P7 vs. P14; Figure 1A). In contrast, in mesenteric arteries, there were no differences in the mRNA levels of GPER between pregnant and non-pregnant rats or throughout gestation (Figure 1B).

2.2. Pregnancy Modulates Vascular Reactivity Response to G1 in Uterine but Not in Mesenteric Arteries

To provide further insights into the role of GPER, we then ascertained that the GPER agonist, G1, induces vasorelaxation of uterine (Figure 2A) and mesenteric (Figure 2B) arteries in a dose-dependent manner in both pregnant and non-pregnant rats. Notably, in uterine arteries we found a significant effect of pregnancy on G1-mediated vasorelaxation; it was modulated by gestational age (Figure 2A). G1-vasorelaxation was significantly higher in P7 compared with NP rats, with a maximum efficacy of 82.4 ± 6.0% in P7 vs. 66.5 ± 3.7% in NP rats (Figure 2A). The effect of G1 on uterine arteries significantly increased following the progression of pregnancy, with a maximum vasorelaxation of 97.8 ± 2.5% in P14 rats (Figure 2A) at 10−6 M concentration of G1 (Figure 2A). In contrast, in mesenteric arteries, dose-responses to G1 were similar between pregnant and non-pregnant rats and throughout gestation (Figure 2B).

2.3. G1-Induced Vasorelaxation of Uterine Arteries Involves L-Type Calcium Channels

We have previously demonstrated that G1 induces GPER-mediated vasorelaxation via the NO-cGMP pathway in uterine arteries of pregnant rats [27]. To provide further data we investigated uterine artery G1-vasorelaxation in the presence of the inhibitors of L-type Ca2+ channels (verapamil) and of ERK1/2 (PD 098,059). Using verapamil, the vasorelaxation induced by G1 was significantly reduced compared with the control group (18.5 ± 4.9% vs. 68.8 ± 8.4, p < 0.001; Figure 3A). Moreover, the vasorelaxation induced by G1 was also significantly blunted in the presence of PD 098,059 (48.0 ± 5.1% vs. 71.7 ± 5.2%, p < 0.05; Figure 3B).

3. Discussion

The present study demonstrates that (1) expression of GPER is increased, (2) vasorelaxation to G1 is enhanced as pregnancy progresses; GPER-mediated vasorelaxation occurs through (3) L-type Ca2+ channels and (4) ERK1/2 pathway in uterine arteries of pregnant rats; (5) functional expression of GPER does not change through gestation in mesenteric arteries. Activation of GPER has been shown to cause vasorelaxation in rat aorta, carotid, mesenteric, renal, cerebral, and coronary arteries [30,31,32,33,34]. In addition, sex-related controversial effects and stark differences between vascular beds have also been reported [32,35,36,37,38]. Although considerable effort has been made to appreciate the role of GPER in the vasculature in both human and animal models (Supplementary Table S1, in Supplementary Materials), more studies are needed to fully delineate the physiological importance of GPER.
GPER together with the cognate receptors, ERα and ERβ, mediates physiological functions of estrogens, including the crucial role of regulating vascular function [25,26]. The agonist–receptor-coupling with the consequent production and release of NO and other endothelium-derived factors, determine the degree to which extent estrogens may elicit vasorelaxation [39,40]. In the present investigation, pregnancy significantly increased GPER-mediated vasorelaxation in uterine arteries, and this effect was modulated by gestational age.
To elucidate whether the modulating role of pregnancy on GPER-mediated vasorelaxation may be vascular-bed specific, we applied G1 in a cumulative fashion on arteries obtained from the mesenteric circulation. In agreement with other studies [30,34], G1 elicited a vasorelaxing effect in mesenteric arteries. Contrary to our findings in the uterine vasculature, the similarity of responses to G1 (41–52% maximum vasorelaxation) in mesenteric arteries of both -NP and P rats, as well as at different gestational periods, suggests that pregnancy-induced modulation of GPER vascular function is specific of the uterine circulation.
It may be implied that pregnancy-associated changes of the uterine reactivity to G1, could predict an up-regulation of GPER expression in this vascular bed. In fact, evaluation of GPER expression revealed that pregnancy modulates mRNA levels of GPER in uterine arteries, and a trend towards increase in GPER expression was observed as pregnancy progressed. A limitation of this study is that we investigated only mRNA levels of GPER. Further additional data in terms of protein expression are needed to fully support our conclusion. However, we may speculate that an increase in mRNA corresponds with an increase in the relative protein expression of GPER. Our results demonstrated a significant increase in GPER mRNA levels already after 7 days of pregnancy, with GPER expression exceeding a one-fold increase in P14 compared with NP rats. Contrariwise, we found no changes in GPER expression in mesenteric arteries, which could explain our functional results, and suggests again that pregnancy-induced modulation of GPER is specific to uterine arteries.
The lack of modulation in mesenteric arteries may be attributed to the different adaptation of the systemic circulation to pregnancy, in terms of both reactivity and remodelling [2], with the reproductive system undertaking a greater role in both maintaining vascular adaptation [5,24] and regulating uterine blood flow throughout gestation [22]. It is also plausible that the action of estrogens is more powerful at the uterine site than in the systemic circulation through the upregulation of GPER in uterine arteries, which in turn makes this vasculature more sensitive to estrogens. An overexpression of GPER, due to increased sensitivity to estrogens, has been observed in the human melanoma tissues [41].
Although we have previously demonstrated that GPER activation elicits endothelium-dependent vasorelaxation via the NO-cGMP pathway in rat uterine arteries [27], our further elucidation of the functional signalling identified a possible smooth muscle-related mechanism involved in the vascular responses. In our current study, inhibition of L-type Ca2+ channels with verapamil caused about a three-times reduction of the vasorelaxation, thus suggesting that L-type Ca2+ channels participate in the GPER-mediated vasorelaxing effect. This conclusion is supported by other studies that have provided evidence that GPER mediates rapid cell signalling via stimulation of intracellular Ca2+ mobilisation [26,42,43,44,45]. It has been demonstrated that extracellular application of G1 on human aortic vascular smooth muscle cells (VSMCs) increases intracellular Ca2+ concentrations, and this effect is attenuated by GPER silencing [43]. Pre-incubation with the L-type Ca2+ channel agonist Bay K8644, enhanced the vasodilatory estrogenic effects on placental arteries [17]. Furthermore, infusion of estradiol increased perfusion pressure and coronary resistance through a non-genomic molecular mechanism involving activation of the L-type Ca2+ channels in rats [46]. Additionally, inhibition of L-type Ca2+ channels with nifedipine significantly reduced G1-induced intracellular Ca2+ increase in myometrial cells [47], and activation of GPER inhibited the endothelin-1-stimulated increase of intracellular Ca2+ concentrations in VSMCs of murine carotid arteries [45]. Although there is heterogeneity amongst cells on the mechanism through which GPER induces Ca2+ mobilisation [47], both the uptake of extracellular Ca2+ and the release of the ion from intracellular Ca2+ stores in cells may be involved in the GPER molecular signalling.
Studies conducted mainly in cell lines have shown that either estrogens or the GPER-selective agonist G1, can activate a number of protein kinases, including ERK1/2, via a non-ER dependent mechanism that requires increased intracellular concentrations of Ca2+ [48,49,50,51,52] through the L-type Ca2+ channels [51]. It has been demonstrated that ERK 1/2 is a signalling component of the GPER cascade that induces NO production in human endothelial cells [53], with the same mechanism [54] through which estrogen-mediated rapid intracellular signalling regulates NO synthesis [55].
Therefore, once the participation of the L-type Ca2+ channels was established, we sought to investigate the involvement of ERK 1/2 signalling in the vascular effects elicited by GPER in uterine arteries.
Incubation with the ERK 1/2 inhibitor, PD 098059, showed that vasorelaxation in response to G1 was attenuated by 24%, which is suggestive of a partial contribution of ERK1/2 in the mechanism of action of GPER in uterine arteries. The partial inhibition suggests that other protein kinases may be involved in the signalling cascade activated by GPER and warrant further investigation.
Consistent with our data, other studies provided evidence that GPER promotes activation of ERK1/2 in cancer cell lines [50,56], in rat heart [57,58], and in human umbilical vein smooth muscle cells [43].
In conclusion, we have shown that activation of GPER induces vasorelaxation of reproductive (uterine) and systemic (mesenteric) arteries. In uterine arteries, GPER-mediated vasorelaxation increases as pregnancy progresses. This can be explained by an upregulation of GPER gene expression in uterine arteries during pregnancy, which may be of importance in regulating hemodynamic changes of pregnancy in the reproductive system. Moreover, the emerging contribution of L-type Ca2+ channels and ERK 1/2 signalling provides a new perspective for understanding the vascular mechanisms involved in GPER-mediated vasorelaxation of uterine arteries in pregnant rats. The modulating effect of both GPER expression and GPER vasorelaxation, associated with gestational age, is supportive of a physiological role for GPER in the uterine vascular adaptation and may offer a novel therapeutic target through which to selectively improve local uterine circulation in pregnancies with compromised uteroplacental blood flow.

4. Methods

4.1. Experimental Animals

The present study was performed in accordance with the European Guidelines on the protection of animals used for scientific purposes (Directive 2010/63/EU). Arteries were isolated from animals used in studies approved by the local ethical committee (OPBA) at the University of Calabria and the Italian Ministry of Health (Ufficio VI, n. 295/2016-PR, March 2016 and n. 530/2021-PR, July 2021).
Sprague–Dawley rats were housed under controlled conditions on a 12 h light/dark cycle at 20–22 °C; commercial chow and tap water were provided ad libitum. Experiments were performed on age-matched (12–15 weeks old) non-pregnant (NP; n = 8) and pregnant rats at gestational days 7 (P7; n= 8) and 14 (P14; n= 8). Pregnant rats were obtained by mating a female in proestrus with a fertile male overnight and checked the following morning. Detection of spermatozoa in the vaginal smear was used to confirm day 1 of pregnancy. Rats were euthanized with isoflurane, followed by decapitation with a small animal guillotine. Rapidly, the uterus and mesentery were both removed and collected in ice-cold HEPES-physiological saline solution (HEPES-PSS, in mmol/L: sodium chloride 141.8, potassium chloride 4.7, magnesium sulfate 1.7, calcium chloride 2.8, potassium phosphate 1.2, HEPES 10.0, EDTA 0.5, and dextrose 5.0).

4.2. Pressure Myography

Radial uterine and third-order mesenteric arteries (diameter < 300 µm) obtained from NP, P7, and P14 rats were dissected free from perivascular connective and adipose tissue in ice-cold HEPES-PSS. Arterial segments (1–2 mm long) were transferred to the chamber of a small-vessel pressure myograph. One end of the artery was tied onto a glass cannula, any luminal content was flushed off by increasing intraluminal pressure before securing the distal end onto a second glass cannula using a servo-null pressure system (Living Systems Instrumentation, St. Albans City, VT, USA). All arteries were continuously superfused with HEPES-PSS at 37 °C, pressurised to 50 mmHg, and equilibrated for 45 min under no-flow conditions before the experiment started. Lumen diameter was measured by trans-illuminating each arterial segment in conjunction with data-acquisition software (Ionoptix, Westwood, MA, USA) to continuously record lumen diameter. Following equilibration, arteries were pre-constricted with phenylephrine (10−7) to produce a 40–50% reduction in baseline diameter [59]. Once pre-constriction was achieved and remained stable for about 10 min, a concentration–response curve to the specific agonist of GPER, 1-(4-(-6-Bromobenzol(1,3)diodo5-yl)3a,4,5,9b-tetrahidro-3Hcyclopenta(c-)ethenone-8yl)ethenone (G1; 10−10–10−6 M), was obtained (a representative pressure myography trace is reported in Supplementary Materials as Figure S1). To investigate the contribution of L-type calcium channels and ERK1/2 signalling, uterine arteries were pre-incubated with the inhibitors verapamil (10−5 M) or PD 098059 (10−6 M), respectively, for 30 min before pre-constriction with phenylephrine (10−6 M) to produce a 40–50% reduction in baseline diameter, and vasorelaxation to G1 was assessed. HEPES-PSS without Ca2+ plus papaverine (10−4 M) was added to calculate the maximum vessel diameter at the end of each experiment.

4.3. Total RNA Extraction and Quantitative Real-Time Polymerase Chain Reaction (qPCR)

Uterine and mesenteric arteries were isolated as described above and preserved at −80 °C. Frozen samples were homogenised with a motor-driven homogeniser, and total RNA was isolated using Trizol reagent (Life Technologies, Milan, Italy), according to the manufacturer’s instructions. RNA was quantified spectrophotometrically, and quality was checked by electrophoresis, through agarose gels stained with ethidium bromide. Total cDNA was synthesised from RNA by reverse transcription using the murine leukemia virus reverse transcriptase (Life Technologies, Milan, Italy), following the protocol provided by the manufacturer. The expression of GPER was quantified by real-time qPCR using the Step OneTM sequence detection system (Applied Biosystems Inc., Milan, Italy). Specific primers for GAPDH and GPER were designed using Primer Express version 2.0 software (Applied Biosystems). The following sets of primers were used: for GAPDH (internal control), 5′-CAAGGCTGTGGGCAAGGT-3′ (forward), 5′-GGAAGGCCATGCCAGTGA-3′ (reverse); for GPER, 5′-CCTGGACGAGCAGTATTACGATAT-3′ (forward), 5′-CGCTGCTGTACATGTTGATCTG-3′ (reverse). Assays were performed in triplicate.

4.4. Drugs and Chemicals

G1 and PD098059 were purchased from TOCRIS (R&D Systems, Milan, Italy); Verapamil was purchased from Santa Cruz (Heidelberg, Germany). All the other chemicals were purchased from Sigma-Aldrich (Milan, Italy).
G1 stock solution was dissolved in DMSO, and small aliquots were frozen prior to use. All the other drugs tested were prepared daily. Phenylephrine and papaverine were dissolved in HEPES-PSS; verapamil and PD 098059 inhibitors were dissolved in DMSO. All drugs were kept on ice, in lightproof vials, and further diluted in HEPES-PSS as required.

4.5. Statistical Analysis

Vasorelaxation to G1 was expressed as percentage of maximally vasorelaxed diameter. Data are expressed as mean ± SEM, and n is the number of rats used. Differences between groups were determined by one-way ANOVA (for relative mRNA levels), and two-way ANOVA for repeated measures analysis, followed by Tukey’s post hoc test where appropriate (for vasorelaxant responses). Differences in vasorelaxation following incubation with inhibitors were determined using paired Student’s t-test, performed by Prism 8 software (GraphPad Software Inc., La Jolla, CA, USA). Differences were considered significant at p < 0.05.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23115996/s1. References [60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99] are cited in Supplementary Materials.

Author Contributions

Conceptualization, M.M. (Maurizio Mandalà) and T.T.; methodology, M.M. (Maurizio Mandalà) and M.M. (Marcello Maggiolini); formal analysis, T.T., D.R. and M.M. (Maurizio Mandalà); investigation, T.T., D.R. and M.E.; resources, M.M. (Maurizio Mandalà) and M.M. (Marcello Maggiolini); data curation, T.T., D.R., M.M. (Maurizio Mandalà) and M.E.; writing—original draft preparation, T.T. and M.M. (Maurizio Mandalà); writing—review and editing, M.M. (Maurizio Mandalà) and M.M. (Marcello Maggiolini); supervision, M.M. (Maurizio Mandalà); funding acquisition, M.M. (Maurizio Mandalà). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The present study was performed in accordance with the European Guidelines on the protection of animals used for scientific purposes (Directive 2010/63/EU). Arteries were isolated from animals used in studies approved by the local ethical committee (OPBA) at the University of Calabria and the Italian Ministry of Health (Ufficio VI, n. 295/2016-PR, March 2016 and n. 530/2021-PR, July 2021).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Poston, L.; McCarthy, A.L.; Ritter, J.M. Control of vascular resistance in the maternal and feto-placental arterial beds. Pharmacol. Ther. 1995, 65, 215–239. [Google Scholar] [CrossRef]
  2. Osol, G.; Ko, N.L.; Mandalà, M. Plasticity of the Maternal Vasculature During Pregnancy. Annu. Rev. Physiol. 2019, 81, 89–111. [Google Scholar] [CrossRef] [PubMed]
  3. Reynolds, L.P.; Caton, J.S.; Redmer, D.A.; Grazul-Bilska, A.T.; Vonnahme, K.A.; Borowicz, P.P.; Luther, J.S.; Wallace, J.M.; Wu, G.; Spencer, T.E. Evidence for altered placental blood flow and vascularity in compromised pregnancies. J. Physiol. 2006, 572 Pt 1, 51–58. [Google Scholar] [CrossRef] [PubMed]
  4. Osol, G.; Mandala, M. Maternal uterine vascular remodeling during pregnancy. Physiology 2009, 24, 58–71. [Google Scholar] [CrossRef] [Green Version]
  5. van der Heijden, O.W.; Essers, Y.P.; Spaanderman, M.E.; De Mey, J.G.; van Eys, G.J.; Peeters, L.L. Uterine artery remodeling in pseudopregnancy is comparable to that in early pregnancy. Biol. Reprod. 2005, 73, 1289–1293. [Google Scholar] [CrossRef] [Green Version]
  6. Mandala, M.; Osol, G. Physiological remodelling of the maternal uterine circulation during pregnancy. Basic Clin. Pharmacol. Toxicol. 2012, 110, 12–18. [Google Scholar] [CrossRef]
  7. Magness, R.R.; Shideman, C.R.; Habermehl, D.A.; Sullivan, J.A.; Bird, I.M. Endothelial vasodilator production by uterine and systemic arteries. V. Effects of ovariectomy, the ovarian cycle, and pregnancy on prostacyclin synthase expression. Prostaglandins Other Lipid Mediat. 2000, 60, 103–118. [Google Scholar] [CrossRef]
  8. Valdes, G.; Kaufmann, P.; Corthorn, J.; Erices, R.; Brosnihan, K.B.; Joyner-Grantham, J. Vasodilator factors in the systemic and local adaptations to pregnancy. Reprod. Biol. Endocrinol. 2009, 7, 79. [Google Scholar] [CrossRef] [Green Version]
  9. Bird, I.M.; Sullivan, J.A.; Di, T.; Cale, J.M.; Zhang, L.; Zheng, J.; Magness, R.R. Pregnancy-dependent changes in cell signaling underlie changes in differential control of vasodilator production in uterine artery endothelial cells. Endocrinology 2000, 141, 1107–1117. [Google Scholar] [CrossRef]
  10. Fulep, E.E.; Vedernikov, Y.P.; Saade, G.R.; Garfield, R.E. The role of endothelium-derived hyperpolarizing factor in the regulation of the uterine circulation in pregnant rats. Am. J. Obstet. Gynecol. 2001, 185, 638–642. [Google Scholar] [CrossRef]
  11. Gokina, N.I.; Kuzina, O.Y.; Vance, A.M. Augmented EDHF signaling in rat uteroplacental vasculature during late pregnancy. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H1642–H1652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Miller, V.M.; Duckles, S.P. Vascular actions of estrogens: Functional implications. Pharmacol. Rev. 2008, 60, 210–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Mandalà, M. Influence of Estrogens on Uterine Vascular Adaptation in Normal and Preeclamptic Pregnancies. Int. J. Mol. Sci. 2020, 21, 2592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Jaimes, L.; Vinet, R.; Knox, M.; Morales, B.; Benites, J.; Laurido, C.; Martínez, J.L. A Review of the Actions of Endogenous and Exogenous Vasoactive Substances during the Estrous Cycle and Pregnancy in Rats. Animals 2019, 9, 288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Chakrabarti, S.; Morton, J.S.; Davidge, S.T. Mechanisms of estrogen effects on the endothelium: An overview. Can. J. Cardiol. 2014, 30, 705–712. [Google Scholar] [CrossRef]
  16. Bai, J.; Qi, Q.R.; Li, Y.; Day, R.; Makhoul, J.; Magness, R.R.; Chen, D.B. Estrogen Receptors and Estrogen-Induced Uterine Vasodilation in Pregnancy. Int. J. Mol. Sci. 2020, 21, 4349. [Google Scholar] [CrossRef]
  17. Corcoran, J.J.; Nicholson, C.; Sweeney, M.; Charnock, J.C.; Robson, S.C.; Westwood, M.; Taggart, M.J. Human uterine and placental arteries exhibit tissue-specific acute responses to 17β-estradiol and estrogen-receptor-specific agonists. Mol. Hum. Reprod. 2014, 20, 433–441. [Google Scholar] [CrossRef]
  18. Meyer, M.R.; Barton, M. Estrogens and Coronary Artery Disease: New Clinical Perspectives. Adv. Pharmacol. 2016, 77, 307–360. [Google Scholar]
  19. Holm, A.; Nilsson, B.O. Identification and characterization of new mechanisms in vascular oestrogen signalling. Basic Clin. Pharmacol. Toxicol. 2013, 113, 287–293. [Google Scholar] [CrossRef]
  20. Rosenfeld, C.R.; Roy, T.; Cox, B.E. Mechanisms modulating estrogen-induced uterine vasodilation. Vascul. Pharmacol. 2002, 38, 115–125. [Google Scholar] [CrossRef]
  21. Rosenfeld, C.R.; Cox, B.E.; Roy, T.; Magness, R.R. Nitric oxide contributes to estrogen-induced vasodilation of the ovine uterine circulation. J. Clin. Investig. 1996, 98, 2158–2166. [Google Scholar] [CrossRef] [PubMed]
  22. Van Buren, G.A.; Yang, D.S.; Clark, K.E. Estrogen-induced uterine vasodilatation is antagonized by L-nitroarginine methyl ester, an inhibitor of nitric oxide synthesis. Am. J. Obstet. Gynecol. 1992, 167, 828–833. [Google Scholar] [CrossRef]
  23. Mendelsohn, M.E. Genomic and nongenomic effects of estrogen in the vasculature. Am. J. Cardiol. 2002, 90, 3F–6F. [Google Scholar] [CrossRef]
  24. Pastore, M.B.; Jobe, S.O.; Ramadoss, J.; Magness, R.R. Estrogen receptor-a and estrogen receptor-b in the uterine vascular endothelium during pregnancy: Functional implications for regulating uterine blood flow. Semin. Reprod. Med. 2012, 30, 46–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Meyer, M.R.; Prossnitz, E.R.; Barton, M. GPER/GPR30 and Regulation of Vascular Tone and Blood Pressure. Immunol. Endocr. Metab. Agents Med. Chem. 2011, 11, 255–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Revankar, C.M.; Cimino, D.F.; Sklar, L.A.; Arterburn, J.B.; Prossnitz, E.R. A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science 2005, 307, 1625–1630. [Google Scholar] [CrossRef] [Green Version]
  27. Tropea, T.; De Francesco, E.M.; Rigiracciolo, D.; Maggiolini, M.; Wareing, M.; Osol, G.; Mandalà, M. Pregnancy Augments G Protein Estrogen Receptor (GPER) Induced Vasodilation in Rat Uterine Arteries via the Nitric Oxide—cGMP Signaling Pathway. PLoS ONE 2015, 10, e0141997. [Google Scholar] [CrossRef] [Green Version]
  28. Lindberg, B.S.; Johansson, E.D.; Nilsson, B.A. Plasma levels of nonconjugated oestrone, oestradiol-17beta and oestriol during uncomplicated pregnancy. Acta Obstet. Gynecol. Scand. Suppl. 1974, 32, 21–36. [Google Scholar] [CrossRef]
  29. Taya, K.; Greenwald, G.S. In vivo and in vitro ovarian steroidogenesis in the pregnant rat. Biol. Reprod. 1981, 25, 683–691. [Google Scholar] [CrossRef] [Green Version]
  30. Peixoto, P.; Aires, R.D.; Lemos, V.S.; Bissoli, N.S.; Santos, R.L.D. GPER agonist dilates mesenteric arteries via PI3K-Akt-eNOS and potassium channels in both sexes. Life Sci. 2017, 183, 21–27. [Google Scholar] [CrossRef]
  31. Evanson, K.W.; Goldsmith, J.A.; Ghosh, P.; Delp, M.D. The G protein-coupled estrogen receptor agonist, G-1, attenuates BK channel activation in cerebral arterial smooth muscle cells. Pharmacol. Res. Perspect. 2018, 6, e00409. [Google Scholar] [CrossRef] [PubMed]
  32. Debortoli, A.R.; Rouver, W.; Delgado, N.; Mengal, V.; Claudio, E.; Pernomian, L.; Bendhack, L.M.; Moysés, M.R.; Santos, R. GPER modulates tone and coronary vascular reactivity in male and female rats. J. Mol. Endocrinol. 2017, 59, 171–180. [Google Scholar] [CrossRef] [PubMed]
  33. Jang, E.J.; Seok, Y.M.; Arterburn, J.B.; Olatunji, L.A.; Kim, I.K. GPER-1 agonist G1 induces vasorelaxation through activation of epidermal growth factor receptor-dependent signalling pathway. J. Pharm. Pharmacol. 2013, 65, 1488–1499. [Google Scholar] [CrossRef] [PubMed]
  34. Mata, K.M.; Li, W.; Reslan, O.M.; Siddiqui, W.T.; Opsasnick, L.A.; Khalil, R.A. Adaptive increases in expression and vasodilator activity of estrogen receptor subtypes in a blood vessel-specific pattern during pregnancy. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H1679–H1696. [Google Scholar] [CrossRef] [Green Version]
  35. Peixoto, P.; da Silva, J.F.; Aires, R.D.; Costa, E.D.; Lemos, V.S.; Bissoli, N.S.; Dos Santos, R.L. Sex difference in GPER expression does not change vascular relaxation or reactive oxygen species generation in rat mesenteric resistance arteries. Life Sci. 2018, 211, 198–205. [Google Scholar] [CrossRef]
  36. Broughton, B.R.; Miller, A.A.; Sobey, C.G. Endothelium-dependent relaxation by G protein-coupled receptor 30 agonists in rat carotid arteries. Am. J. Physiol. Heart Circ. Physiol. 2010, 298, H1055–H1061. [Google Scholar] [CrossRef] [Green Version]
  37. Murata, T.; Dietrich, H.H.; Xiang, C.; Dacey, R.G. G protein-coupled estrogen receptor agonist improves cerebral microvascular function after hypoxia/reoxygenation injury in male and female rats. Stroke 2013, 44, 779–785. [Google Scholar] [CrossRef]
  38. Lindsey, S.H.; da Silva, A.S.; Silva, M.S.; Chappell, M.C. Reduced vasorelaxation to estradiol and G-1 in aged female and adult male rats is associated with GPR30 downregulation. Am. J. Physiol. Endocrinol. Metab. 2013, 305, E113–E118. [Google Scholar] [CrossRef] [Green Version]
  39. Miller, V.M.; Mulvagh, S.L. Sex steroids and endothelial function: Translating basic science to clinical practice. Trends Pharmacol. Sci. 2007, 28, 263–270. [Google Scholar] [CrossRef]
  40. Smith, R.; Klopper, A.; Hughes, G.; Wilson, G. The compartmental distribution of oestrogens and pregnancy specific beta1 glycoprotein. Br. J. Obstet. Gynaecol. 1979, 86, 119–124. [Google Scholar] [CrossRef]
  41. Fábián, M.; Rencz, F.; Krenács, T.; Brodszky, V.; Hársing, J.; Németh, K.; Balogh, P.; Kárpáti, S. Expression of G protein-coupled oestrogen receptor in melanoma and in pregnancy-associated melanoma. J. Eur. Acad. Dermatol. Venereol. 2017, 31, 1453–1461. [Google Scholar] [CrossRef] [PubMed]
  42. Ariazi, E.A.; Brailoiu, E.; Yerrum, S.; Shupp, H.A.; Slifker, M.J.; Cunliffe, H.E.; Black, M.A.; Donato, A.L.; Arterburn, J.B.; Oprea, T.I.; et al. The G protein-coupled receptor GPR30 inhibits proliferation of estrogen receptor-positive breast cancer cells. Cancer Res. 2010, 70, 1184–1194. [Google Scholar] [CrossRef] [Green Version]
  43. Haas, E.; Bhattacharya, I.; Brailoiu, E.; Damjanović, M.; Brailoiu, G.C.; Gao, X.; Mueller-Guerre, L.; Marjon, N.A.; Gut, A.; Minotti, R.; et al. Regulatory role of G protein-coupled estrogen receptor for vascular function and obesity. Circ. Res. 2009, 104, 288–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Prossnitz, E.R.; Barton, M. The G-protein-coupled estrogen receptor GPER in health and disease. Nat. Rev. Endocrinol. 2011, 7, 715–726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Meyer, M.R.; Field, A.S.; Kanagy, N.L.; Barton, M.; Prossnitz, E.R. GPER regulates endothelin-dependent vascular tone and intracellular calcium. Life Sci. 2012, 91, 623–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Valverde, L.F.; Cedillo, F.D.; Ramos, M.L.; Cervera, E.G.; Quijano, K.; Cordoba, J. Changes induced by estradiol-ethylenediamine derivative on perfusion pressure and coronary resistance in isolated rat heart: L-type calcium channel. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czech Repub. 2011, 155, 27–32. [Google Scholar] [CrossRef] [Green Version]
  47. Han, G.; Li, F.; Yu, X.; White, R.E. GPER: A novel target for non-genomic estrogen action in the cardiovascular system. Pharmacol. Res. 2013, 71, 53–60. [Google Scholar] [CrossRef]
  48. Improta-Brears, T.; Whorton, A.R.; Codazzi, F.; York, J.D.; Meyer, T.; McDonnell, D.P. Estrogen-induced activation of mitogen-activated protein kinase requires mobilization of intracellular calcium. Proc. Natl. Acad. Sci. USA 1999, 96, 4686–4691. [Google Scholar] [CrossRef] [Green Version]
  49. Sharma, G.; Prossnitz, E.R. Mechanisms of estradiol-induced insulin secretion by the G protein-coupled estrogen receptor GPR30/GPER in pancreatic beta-cells. Endocrinology 2011, 152, 3030–3039. [Google Scholar] [CrossRef]
  50. Filardo, E.J.; Quinn, J.A.; Bland, K.I.; Frackelton, A.R. Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein-coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol. Endocrinol. 2000, 14, 1649–1660. [Google Scholar] [CrossRef]
  51. Wu, T.W.; Wang, J.M.; Chen, S.; Brinton, R.D. 17Beta-estradiol induced Ca2+ influx via L-type calcium channels activates the Src/ERK/cyclic-AMP response element binding protein signal pathway and BCL-2 expression in rat hippocampal neurons: A potential initiation mechanism for estrogen-induced neuroprotection. Neuroscience 2005, 135, 59–72. [Google Scholar] [PubMed]
  52. Muchekehu, R.W.; Harvey, B.J. 17beta-estradiol rapidly mobilizes intracellular calcium from ryanodine-receptor-gated stores via a PKC-PKA-Erk-dependent pathway in the human eccrine sweat gland cell line NCL-SG3. Cell Calcium 2008, 44, 276–288. [Google Scholar] [CrossRef] [PubMed]
  53. Fredette, N.C.; Meyer, M.R.; Prossnitz, E.R. Role of GPER in estrogen-dependent nitric oxide formation and vasodilation. J. Steroid Biochem. Mol. Biol. 2018, 176, 65–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Salerno, J.C.; Ghosh, D.K.; Razdan, R.; Helms, K.A.; Brown, C.C.; McMurry, J.L.; Rye, E.A.; Chrestensen, C.A. Endothelial nitric oxide synthase is regulated by ERK phosphorylation at Ser602. Biosci. Rep. 2014, 34, e00137. [Google Scholar] [CrossRef]
  55. Russell, K.S.; Haynes, M.P.; Sinha, D.; Clerisme, E.; Bender, J.R. Human vascular endothelial cells contain membrane binding sites for estradiol, which mediate rapid intracellular signaling. Proc. Natl. Acad. Sci. USA 2000, 97, 5930–5935. [Google Scholar] [CrossRef] [Green Version]
  56. Vivacqua, A.; Bonofiglio, D.; Recchia, A.G.; Musti, A.M.; Picard, D.; Andò, S.; Maggiolini, M. The G protein-coupled receptor GPR30 mediates the proliferative effects induced by 17beta-estradiol and hydroxytamoxifen in endometrial cancer cells. Mol. Endocrinol. 2006, 20, 631–646. [Google Scholar] [CrossRef] [Green Version]
  57. De Francesco, E.M.; Angelone, T.; Pasqua, T.; Pupo, M.; Cerra, M.C.; Maggiolini, M. GPER mediates cardiotropic effects in spontaneously hypertensive rat hearts. PLoS ONE 2013, 8, e69322. [Google Scholar] [CrossRef] [Green Version]
  58. Filice, E.; Recchia, A.G.; Pellegrino, D.; Angelone, T.; Maggiolini, M.; Cerra, M.C. A new membrane G protein-coupled receptor (GPR30) is involved in the cardiac effects of 17beta-estradiol in the male rat. J. Physiol. Pharmacol. 2009, 60, 3–10. [Google Scholar]
  59. Colton, I.; Mandalà, M.; Morton, J.; Davidge, S.T.; Osol, G. Influence of constriction, wall tension, smooth muscle activation and cellular deformation on rat resistance artery vasodilator reactivity. Cell Physiol. Biochem. 2012, 29, 883–892. [Google Scholar] [CrossRef]
  60. Park, J.S.; Lee, G.H.; Jin, S.W.; Pham, T.H.; Thai, T.N.; Kim, J.Y.; Kim, C.Y.; Han, E.H.; Hwang, Y.P.; Choi, C.Y.; et al. G protein-coupled estrogen receptor regulates the KLF2-dependent eNOS expression by activating of Ca. Biochem. Pharmacol. 2021, 192, 114721. [Google Scholar] [CrossRef]
  61. Wang, D.; Wang, M.; Sun, P.; Gao, Q. Eplerenone inhibits oxidized low-density lipoprotein-induced proliferation and migration of vascular smooth muscle cells by downregulating GPER expression. Adv. Clin. Exp. Med. 2021, 30, 405–412. [Google Scholar] [CrossRef] [PubMed]
  62. Feng, Z.; Wang, C.; Yue, J.; Meng, Q.; Wu, J.; Sun, H. Kaempferol-induced GPER upregulation attenuates atherosclerosis via the PI3K/AKT/Nrf2 pathway. Pharm. Biol. 2021, 59, 1106–1116. [Google Scholar] [CrossRef] [PubMed]
  63. Kurmann, L.; Okoniewski, M.; Dubey, R.K. Estradiol Inhibits Human Brain Vascular Pericyte Migration Activity: A Functional and Transcriptomic Analysis. Cells 2021, 10, 2314. [Google Scholar] [CrossRef]
  64. Meyer, M.R.; Fredette, N.C.; Daniel, C.; Sharma, G.; Amann, K.; Arterburn, J.B.; Barton, M.; Prossnitz, E.R. Obligatory role for GPER in cardiovascular aging and disease. Sci. Signal. 2016, 9, ra105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Li, F.; Yu, X.; Szynkarski, C.K.; Meng, C.; Zhou, B.; Barhoumi, R.; White, R.E.; Heaps, C.L.; Stallone, J.N.; Han, G. Activation of GPER Induces Differentiation and Inhibition of Coronary Artery Smooth Muscle Cell Proliferation. PLoS ONE 2013, 8, e64771. [Google Scholar]
  66. Ghaffari, S.; Naderi Nabi, F.; Sugiyama, M.G.; Lee, W.L. Estrogen Inhibits LDL (Low-Density Lipoprotein) Transcytosis by Human Coronary Artery Endothelial Cells via GPER (G-Protein-Coupled Estrogen Receptor) and SR-BI (Scavenger Receptor Class B Type 1). Arterioscler. Thromb. Vasc. Biol. 2018, 38, 2283–2294. [Google Scholar] [CrossRef] [Green Version]
  67. Gao, F.; Huang, Y.; Zhang, L.; Liu, W. Involvement of estrogen receptor and GPER in bisphenol A induced proliferation of vascular smooth muscle cells. Toxicol. In Vitro 2019, 56, 156–162. [Google Scholar] [CrossRef]
  68. Feldman, R.D.; Gros, R.; Ding, Q.; Hussain, Y.; Ban, M.R.; McIntyre, A.D.; Hegele, R.A. A common hypofunctional genetic variant of GPER is associated with increased blood pressure in women. Br. J. Clin. Pharmacol. 2014, 78, 1441–1452. [Google Scholar] [CrossRef] [Green Version]
  69. Arefin, S.; Simoncini, T.; Wieland, R.; Hammarqvist, F.; Spina, S.; Goglia, L.; Kublickiene, K. Vasodilatory effects of the selective GPER agonist G-1 is maximal in arteries of postmenopausal women. Maturitas 2014, 78, 123–130. [Google Scholar] [CrossRef]
  70. Serra, R.; Gallelli, L.; Perri, P.; De Francesco, E.M.; Rigiracciolo, D.C.; Mastroroberto, P.; Maggiolini, M.; de Franciscis, S. Estrogen Receptors and Chronic Venous Disease. Eur. J. Vasc. Endovasc. Surg. 2016, 52, 114–118. [Google Scholar] [CrossRef] [Green Version]
  71. Meyer, M.R.; Fredette, N.C.; Barton, M.; Prossnitz, E.R. G protein-coupled estrogen receptor inhibits vascular prostanoid production and activity. J. Endocrinol. 2015, 227, 61–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Kong, B.S.; Cho, Y.H.; Lee, E.J. G protein-coupled estrogen receptor-1 is involved in the protective effect of protocatechuic aldehyde against endothelial dysfunction. PLoS ONE 2014, 9, e113242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Meyer, M.R.; Rosemann, T.; Barton, M.; Prossnitz, E.R. GPER Mediates Functional Endothelial Aging in Renal Arteries. Pharmacology 2017, 100, 188–193. [Google Scholar] [CrossRef] [PubMed]
  74. Shen, F.; Wang, J.; Gao, F.; Zhu, G. Ginsenoside Rg1 Prevents Cognitive Impairment and Hippocampal Neuronal Apoptosis in Experimental Vascular Dementia Mice by Promoting GPR30 Expression. Neural Plast. 2021, 2021, 2412220. [Google Scholar] [CrossRef] [PubMed]
  75. Ferreira, N.S.; Cau, S.B.; Silva, M.A.; Manzato, C.P.; Mestriner, F.L.; Matsumoto, T.; Carneiro, F.S.; Tostes, R.C. Diabetes impairs the vascular effects of aldosterone mediated by G protein-coupled estrogen receptor activation. Front. Pharmacol. 2015, 6, 34. [Google Scholar] [CrossRef] [Green Version]
  76. Ogola, B.O.; Zimmerman, M.A.; Sure, V.N.; Gentry, K.M.; Duong, J.L.; Clark, G.L.; Miller, K.S.; Katakam, P.; Lindsey, S.H. G Protein-Coupled Estrogen Receptor Protects From Angiotensin II-Induced Increases in Pulse Pressure and Oxidative Stress. Front. Endocrinol. 2019, 10, 586. [Google Scholar] [CrossRef] [Green Version]
  77. Serizawa, I.; Iwasaki, N.; Ishida, H.; Saito, S.Y.; Ishikawa, T. G-protein coupled estrogen receptor-mediated non-genomic facilitatory effect of estrogen on cooling-induced reduction of skin blood flow in mice. Eur. J. Pharmacol. 2017, 797, 26–31. [Google Scholar] [CrossRef]
  78. Luo, P.; Wu, M.M.; Gao, P.; Gao, T.; Dong, L.; Ding, X.W.; Meng, Y.Q.; Qian, J.H.; Zhang, G.H.; Rong, W.F. Stress-related arterial hypertension in Gper-deficient rats. Sheng Li Xue Bao 2017, 69, 532–540. [Google Scholar]
  79. Calfío, C.; Donoso, F.; Huidobro-Toro, J.P. Anthocyanins Activate Membrane Estrogen Receptors With Nanomolar Potencies to Elicit a Nongenomic Vascular Response Via NO Production. J. Am. Heart Assoc. 2021, 10, e020498. [Google Scholar] [CrossRef]
  80. Liu, L.; Kashyap, S.; Murphy, B.; Hutson, D.D.; Budish, R.A.; Trimmer, E.H.; Zimmerman, M.A.; Trask, A.J.; Miller, K.S.; Chappell, M.C.; et al. GPER activation ameliorates aortic remodeling induced by salt-sensitive hypertension. Am. J. Physiol. Heart Circ. Physiol. 2016, 310, H953–H961. [Google Scholar] [CrossRef]
  81. Ding, Q.; Hussain, Y.; Chorazyczewski, J.; Gros, R.; Feldman, R.D. GPER-independent effects of estrogen in rat aortic vascular endothelial cells. Mol. Cell Endocrinol. 2015, 399, 60–68. [Google Scholar] [CrossRef] [PubMed]
  82. Gohar, E.Y.; Daugherty, E.M.; Aceves, J.O.; Sedaka, R.; Obi, I.E.; Allan, J.M.; Soliman, R.H.; Jin, C.; De Miguel, C.; Lindsey, S.H.; et al. Evidence for G-Protein-Coupled Estrogen Receptor as a Pronatriuretic Factor. J. Am. Heart Assoc. 2020, 9, e015110. [Google Scholar] [CrossRef] [PubMed]
  83. Alencar, A.; Montes, G.C.; Costa, D.G.; Mendes, L.; Silva, A.; Martinez, S.T.; Trachez, M.M.; Cunha, V.; Montagnoli, T.L.; Fraga, A.; et al. Cardioprotection Induced by Activation of GPER in Ovariectomized Rats With Pulmonary Hypertension. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 1158–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Chang, Y.; Han, Z.; Zhang, Y.; Zhou, Y.; Feng, Z.; Chen, L.; Li, X.; Li, L.; Si, J.Q. G protein-coupled estrogen receptor activation improves contractile and diastolic functions in rat renal interlobular artery to protect against renal ischemia reperfusion injury. Biomed. Pharmacother. 2019, 112, 108666. [Google Scholar] [CrossRef] [PubMed]
  85. Gros, R.; Ding, Q.; Liu, B.; Chorazyczewski, J.; Feldman, R.D. Aldosterone mediates its rapid effects in vascular endothelial cells through GPER activation. Am. J. Physiol. Cell Physiol. 2013, 304, C532–C540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Ding, Q.; Gros, R.; Limbird, L.E.; Chorazyczewski, J.; Feldman, R.D. Estradiol-mediated ERK phosphorylation and apoptosis in vascular smooth muscle cells requires GPR 30. Am. J. Physiol. Cell Physiol. 2009, 297, C1178–C1187. [Google Scholar] [CrossRef] [Green Version]
  87. Kurt, A.H.; Buyukafsar, K. Vasoconstriction induced by G1, a G-protein-coupled oestrogen receptor1 (GPER-1) agonist, in the isolated perfused rat kidney. Eur. J. Pharmacol. 2013, 702, 71–78. [Google Scholar] [CrossRef]
  88. Delgado, N.T.B.; Rouver, W.D.N.; Freitas-Lima, L.C.; Vieira-Alves, I.; Lemos, V.S.; Dos Santos, R.L. Sex Differences in the Vasodilation Mediated by G Protein-Coupled Estrogen Receptor (GPER) in Hypertensive Rats. Front Physiol. 2021, 12, 659291. [Google Scholar] [CrossRef]
  89. Lindsey, S.H.; Liu, L.; Chappell, M.C. Vasodilation by GPER in mesenteric arteries involves both endothelial nitric oxide and smooth muscle cAMP signaling. Steroids 2014, 81, 99–102. [Google Scholar] [CrossRef] [Green Version]
  90. Gros, R.; Hussain, Y.; Chorazyczewski, J.; Pickering, J.G.; Ding, Q.; Feldman, R.D. Extent of Vascular Remodeling Is Dependent on the Balance Between Estrogen Receptor α and G-Protein-Coupled Estrogen Receptor. Hypertension 2016, 68, 1225–1235. [Google Scholar] [CrossRef] [Green Version]
  91. Yu, X.; Zhang, Q.; Zhao, Y.; Schwarz, B.J.; Stallone, J.N.; Heaps, C.L.; Han, G. Activation of G protein-coupled estrogen receptor 1 induces coronary artery relaxation via Epac/Rap1-mediated inhibition of RhoA/Rho kinase pathway in parallel with PKA. PLoS ONE 2017, 12, e0173085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Yu, X.; Li, F.; Klussmann, E.; Stallone, J.N.; Han, G. G protein-coupled estrogen receptor 1 mediates relaxation of coronary arteries via cAMP/PKA-dependent activation of MLCP. Am. J. Physiol. Endocrinol. Metab. 2014, 307, E398–E407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Yu, X.; Ma, H.; Barman, S.A.; Liu, A.T.; Sellers, M.; Stallone, J.N.; Prossnitz, E.R.; White, R.E.; Han, G. Activation of G protein-coupled estrogen receptor induces endothelium-independent relaxation of coronary artery smooth muscle. Am. J. Physiol. Endocrinol. Metab. 2011, 301, E882–E888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Yu, X.; Stallone, J.N.; Heaps, C.L.; Han, G. The activation of G protein-coupled estrogen receptor induces relaxation via cAMP as well as potentiates contraction via EGFR transactivation in porcine coronary arteries. PLoS ONE 2018, 13, e0191418. [Google Scholar] [CrossRef] [Green Version]
  95. Tran, Q.K.; VerMeer, M.; Burgard, M.A.; Hassan, A.B.; Giles, J. Hetero-oligomeric Complex between the G Protein-coupled Estrogen Receptor 1 and the Plasma Membrane Ca2+-ATPase 4b. J. Biol. Chem. 2015, 290, 13293–13307. [Google Scholar] [CrossRef] [Green Version]
  96. Meyer, M.R.; Baretella, O.; Prossnitz, E.R.; Barton, M. Dilation of epicardial coronary arteries by the G protein-coupled estrogen receptor agonists G-1 and ICI 182,780. Pharmacology 2010, 86, 58–64. [Google Scholar] [CrossRef] [Green Version]
  97. Sarmiento, V.; Ramirez-Sanchez, I.; Moreno-Ulloa, A.; Romero-Perez, D.; Chávez, D.; Ortiz, M.; Najera, N.; Correa-Basurto, J.; Villarreal, F.; Ceballos, G. Synthesis of novel (-)-epicatechin derivatives as potential endothelial GPER agonists: Evaluation of biological effects. Bioorg. Med. Chem. Lett. 2018, 28, 658–663. [Google Scholar] [CrossRef]
  98. Moreno-Ulloa, A.; Mendez-Luna, D.; Beltran-Partida, E.; Castillo, C.; Guevara, G.; Ramirez-Sanchez, I.; Correa-Basurto, J.; Ceballos, G.; Villarreal, F. The effects of (-)-epicatechin on endothelial cells involve the G protein-coupled estrogen receptor (GPER). Pharmacol. Res. 2015, 100, 309–320. [Google Scholar] [CrossRef] [Green Version]
  99. Castelló-Ruiz, M.; Salom, J.B.; Fernández-Musoles, R.; Burguete, M.C.; López-Morales, M.A.; Arduini, A.; Jover-Mengual, T.; Hervás, D.; Torregrosa, G.; Alborch, E. Relaxant Effects of the Selective Estrogen Receptor Modulator, Bazedoxifene, and Estrogen Receptor Agonists in Isolated Rabbit Basilar Artery. J. Cardiovasc. Pharmacol. 2016, 68, 313–321. [Google Scholar] [CrossRef]
Figure 1. Pregnancy increases GPER expression in uterine but not in mesenteric arteries. GPER expression was quantified by qPCR in uterine (A) and mesenteric (B) arteries from non-pregnant (NP) and pregnant rats at 7 (P7) and 14 (P14) days of gestation. GPER mRNA was significantly higher in uterine arteries from both P7 and P14 rats compared with NP rats (A). Expression levels were not affected by pregnancy in mesenteric arteries (B). * p < 0.05, P7, P14 vs. NP, by one-way ANOVA. n = 3 independent experiments per group performed in triplicate.
Figure 1. Pregnancy increases GPER expression in uterine but not in mesenteric arteries. GPER expression was quantified by qPCR in uterine (A) and mesenteric (B) arteries from non-pregnant (NP) and pregnant rats at 7 (P7) and 14 (P14) days of gestation. GPER mRNA was significantly higher in uterine arteries from both P7 and P14 rats compared with NP rats (A). Expression levels were not affected by pregnancy in mesenteric arteries (B). * p < 0.05, P7, P14 vs. NP, by one-way ANOVA. n = 3 independent experiments per group performed in triplicate.
Ijms 23 05996 g001
Figure 2. Pregnancy modulates concentration–responses to G1 in uterine arteries. In uterine arteries (A) of pregnant rats, vasorelaxation to G1 was significantly higher at 7 days (P7, n = 8) compared with non-pregnant (NP, n = 8) rats, and this response was even greater at 14 days of pregnancy (P14, n = 8). In mesenteric arteries (B), pregnancy had no effect on responses to G1 (NP, n = 6; P7, n = 5; P14, n = 6). P7 vs. NP: * p < 0.05, *** p < 0.001, **** p < 0.0001; P14 vs. NP: ### p < 0.001, #### p < 0.0001; P7 vs. P14: ° p < 0.05, °° p < 0.01, °°° p < 0.001, by two-way ANOVA.
Figure 2. Pregnancy modulates concentration–responses to G1 in uterine arteries. In uterine arteries (A) of pregnant rats, vasorelaxation to G1 was significantly higher at 7 days (P7, n = 8) compared with non-pregnant (NP, n = 8) rats, and this response was even greater at 14 days of pregnancy (P14, n = 8). In mesenteric arteries (B), pregnancy had no effect on responses to G1 (NP, n = 6; P7, n = 5; P14, n = 6). P7 vs. NP: * p < 0.05, *** p < 0.001, **** p < 0.0001; P14 vs. NP: ### p < 0.001, #### p < 0.0001; P7 vs. P14: ° p < 0.05, °° p < 0.01, °°° p < 0.001, by two-way ANOVA.
Ijms 23 05996 g002
Figure 3. L-type Ca2+ channels and ERK1/2 signalling contribute to G1-mediated vasorelaxation. In uterine arteries, inhibition of L-type calcium channels with verapamil (10−5 M, (A)) and of ERK1/2 pathway with PD 098,059 (10−6 M, (B)) significantly reduced vasorelaxation to G1 (10−8 M). *** p < 0.001, G1 vs. G1 + verapamil (n = 6); * p < 0.05, G1 vs. G1 + PD 090,859 (n = 5), by t-test.
Figure 3. L-type Ca2+ channels and ERK1/2 signalling contribute to G1-mediated vasorelaxation. In uterine arteries, inhibition of L-type calcium channels with verapamil (10−5 M, (A)) and of ERK1/2 pathway with PD 098,059 (10−6 M, (B)) significantly reduced vasorelaxation to G1 (10−8 M). *** p < 0.001, G1 vs. G1 + verapamil (n = 6); * p < 0.05, G1 vs. G1 + PD 090,859 (n = 5), by t-test.
Ijms 23 05996 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tropea, T.; Rigiracciolo, D.; Esposito, M.; Maggiolini, M.; Mandalà, M. G-Protein-Coupled Estrogen Receptor Expression in Rat Uterine Artery Is Increased by Pregnancy and Induces Dilation in a Ca2+ and ERK1/2 Dependent Manner. Int. J. Mol. Sci. 2022, 23, 5996. https://doi.org/10.3390/ijms23115996

AMA Style

Tropea T, Rigiracciolo D, Esposito M, Maggiolini M, Mandalà M. G-Protein-Coupled Estrogen Receptor Expression in Rat Uterine Artery Is Increased by Pregnancy and Induces Dilation in a Ca2+ and ERK1/2 Dependent Manner. International Journal of Molecular Sciences. 2022; 23(11):5996. https://doi.org/10.3390/ijms23115996

Chicago/Turabian Style

Tropea, Teresa, Damiano Rigiracciolo, Milena Esposito, Marcello Maggiolini, and Maurizio Mandalà. 2022. "G-Protein-Coupled Estrogen Receptor Expression in Rat Uterine Artery Is Increased by Pregnancy and Induces Dilation in a Ca2+ and ERK1/2 Dependent Manner" International Journal of Molecular Sciences 23, no. 11: 5996. https://doi.org/10.3390/ijms23115996

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

Article Metrics

Back to TopTop