1
|
Bushi A, Ma Y, Adu-Amankwaah J, Wang R, Cui F, Xiao R, Zhao J, Yuan J, Tan R. G protein-coupled estrogen receptor biased signaling in health and disease. Pharmacol Ther 2025; 269:108822. [PMID: 39978643 DOI: 10.1016/j.pharmthera.2025.108822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
G protein-coupled estrogen receptor (GPER) is now recognized for its pivotal role in cellular signaling, influencing diverse physiological processes and disease states. Unlike classical estrogen receptors, GPER exhibits biased signaling, wherein ligand binding triggers selective pathways over others, significantly impacting cellular responses. This review explores the nuanced mechanisms of biased signaling mediated by GPER, underscoring its relevance in cardiovascular health, neurological function, immune modulation, and oncogenic processes. Despite its critical implications, biased signaling through GPER remains underexplored compared to traditional signaling paradigms. We explore recent progress in understanding GPER signaling specificity and its potential therapeutic implications across various diseases. Future research directions aim to uncover the molecular basis of biased signaling, develop selective ligands, and translate these insights into personalized therapeutic approaches. Exploiting the therapeutic potential of GPER biased signaling represents a promising frontier in precision medicine, offering innovative strategies to address unmet medical needs.
Collapse
Affiliation(s)
- Aisha Bushi
- School international education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yixuan Ma
- First Clinical Medical School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Rong Wang
- The second clinical college, China Medical University, Shenyang, Liaoning 110122, China
| | - Fen Cui
- Research Institution of Behavioral Medicine Education, Jining Medical University, Jining 272067, China
| | - Rui Xiao
- Second Clinical Medical School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jinming Zhao
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China; Department of Pathology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China.
| | - Rubin Tan
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
2
|
Hinton A, Neikirk K, Le H, Harris C, Oliver A, Martin P, Gaye A. Estrogen receptors in mitochondrial metabolism: age-related changes and implications for pregnancy complications. AGING ADVANCES 2024; 1:154-171. [PMID: 39839811 PMCID: PMC11748122 DOI: 10.4103/agingadv.agingadv-d-24-00012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/24/2024] [Indexed: 01/23/2025]
Abstract
Estrogen hormones are primarily associated with their role as female sex hormones responsible for primary and secondary sexual development. Estrogen receptors are known to undergo age-dependent decreases due to age-related changes in hormone production. In the mitochondria, estrogen functions by reducing the production of reactive oxygen species in the electron transport chain, inhibiting apoptosis, and regulating mitochondrial DNA content. Moreover, estrogen receptors may be the key components in maintaining mitochondrial membrane potential and structure. Although estrogen plays a crucial role in the development of pregnancy, our understanding of how estrogen receptors change with aging during pregnancy remains limited. During pregnancy, estrogen levels are significantly elevated, with a corresponding upregulation of estrogen receptors, which play various roles in pregnancy. However, the exact role of estrogen receptors in pregnancy complications remains to be further investigated. The paper reviews the role of estrogen receptors in the regulation of mitochondrial metabolism and in pregnancy complications, with a special focus on the effect of age-related changes on estrogen levels and estrogen receptors function. We also address how estrogen maintains mitochondrial function, including reducing the production of reactive oxygen species in the electron transport chain, inhibiting apoptosis, regulating mitochondrial DNA content, and maintaining mitochondrial membrane potential and structure. However, the effects of estrogen on mitochondria-endoplasmic reticulum contacts have not been well studied. Based on these emergent roles in mitochondria, the differential roles of estrogen receptors in pregnancy complications are of great relevance. The paper emphasizes the association between maternal health and estrogen receptors and indicates the need for future research to elucidate the interdependence of estrogen receptor-regulated maternal health with mitochondrial function and their relationship with the gut microbiome. Overall, we summarize the important role of estrogen receptors during pregnancy and highlight the need for further research to better understand the role of estrogen receptors in aging and pregnancy complications. This not only helps to reveal the mechanism underlying the role of estrogen in maternal health but also has potential clinical implications for the development of new therapies targeting age-related diseases and pregnancy complications.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Han Le
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Chanel Harris
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Ashton Oliver
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Pamela Martin
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Amadou Gaye
- Department of Integrative Genomics and Epidemiology, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
3
|
Zhao Y, Klionsky DJ, Wang X, Huang Q, Deng Z, Xiang J. The Estrogen-Autophagy Axis: Insights into Cytoprotection and Therapeutic Potential in Cancer and Infection. Int J Mol Sci 2024; 25:12576. [PMID: 39684286 PMCID: PMC11641569 DOI: 10.3390/ijms252312576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Macroautophagy, commonly referred to as autophagy, is an essential cytoprotective mechanism that plays a significant role in cellular homeostasis. It has emerged as a promising target for drug development aimed at treating various cancers and infectious diseases. However, the scientific community has yet to reach a consensus on the most effective approach to manipulating autophagy, with ongoing debates about whether its inhibition or stimulation is preferable for managing these complex conditions. One critical factor contributing to the variability in treatment responses for both cancers and infectious diseases is estrogen, a hormone known for its diverse biological effects. Given the strong correlations observed between estrogen signaling and autophagy, this review seeks to summarize the intricate molecular mechanisms that underlie the dual cytoprotective effects of estrogen signaling in conjunction with autophagy. We highlight recent findings from studies that involve various ligands, disease contexts, and cell types, including immune cells. Furthermore, we discuss several factors that regulate autophagy in the context of estrogen's influence. Ultimately, we propose a hypothetical model to elucidate the regulatory effects of the estrogen-autophagy axis on cell fate. Understanding these interactions is crucial for advancing our knowledge of related diseases and facilitating the development of innovative treatment strategies.
Collapse
Affiliation(s)
- Ying Zhao
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (Y.Z.); (X.W.); (Q.H.); (Z.D.)
| | - Daniel J. Klionsky
- Life Sciences Institute, University of Michigan, Mary Sue Coleman Hall, 210 Washtenaw Avenue, Ann Arbor, MI 48109-2216, USA;
| | - Xin Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (Y.Z.); (X.W.); (Q.H.); (Z.D.)
| | - Qiaoying Huang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (Y.Z.); (X.W.); (Q.H.); (Z.D.)
| | - Zixin Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (Y.Z.); (X.W.); (Q.H.); (Z.D.)
| | - Jin Xiang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; (Y.Z.); (X.W.); (Q.H.); (Z.D.)
| |
Collapse
|
4
|
de Alencar AKN, Swan KF, Mahapatra S, Lindsey SH, Pridjian GC, Bayer CL. GPER Stimulation Attenuates Cardiac Dysfunction in a Rat Model of Preeclampsia. Hypertension 2024; 81:e161-e172. [PMID: 39224973 PMCID: PMC11483207 DOI: 10.1161/hypertensionaha.123.22303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Preeclampsia poses a substantial clinical challenge, characterized by maternal hypertension, cardiac dysfunction, and persistent cardiovascular risks for both the mother and offspring. Despite the known roles of the estrogen receptor (GPER [G protein-coupled estrogen receptor]) in placental development, its impact on cardiovascular aspects within a preeclampsia animal model remains unexplored. We propose that G-1, a GPER agonist, could have the potential to regulate not only hypertension but also cardiac dysfunction in rats with preeclampsia. METHODS To explore the influence of G-1 on preeclampsia, we used the reduced uterine perfusion pressure (RUPP) model. RUPP rats were administered either G-1 (100 µg/kg per day) or hydralazine (25 mg/kg per day). We conducted echocardiography to probe the intricate cardiac effects of G-1. RESULTS The RUPP rat model revealed signs of hypertension and cardiac dysfunction and alterations in gene and protein expression within placental and heart tissues. G-1 treatment reduced blood pressure and reversed cardiac dysfunction in rats with preeclampsia. In contrast, administration of the vasodilator hydralazine reduced blood pressure without an improvement in cardiac function. In addition, while G-1 treatment restored the levels of sFLT-1 (soluble fms-like tyrosine kinase-1) in RUPP rats, hydralazine did not normalize this antiangiogenic factor. CONCLUSIONS The therapeutic intervention of G-1 significantly mitigated the cardiovascular dysfunction observed in the RUPP rat model of preeclampsia. This discovery underscores the broader significance of understanding GPER's role in the context of preeclampsia-related cardiovascular complications.
Collapse
Affiliation(s)
| | - Kenneth F. Swan
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Smruti Mahapatra
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, 70112, USA
| | - Gabriella C. Pridjian
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Carolyn L. Bayer
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70118, USA
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| |
Collapse
|
5
|
Almutlaq RN, Pollock DM, Gohar EY. Endothelin receptor B is required for the blood pressure-lowering effect of G protein-coupled estrogen receptor 1 in ovariectomized rats. Am J Physiol Renal Physiol 2024; 327:F599-F609. [PMID: 39143913 PMCID: PMC11483081 DOI: 10.1152/ajprenal.00059.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024] Open
Abstract
Activation of G protein-coupled estrogen receptor 1 (GPER1) elicits antihypertensive actions in different animal models. The endothelin-1 signaling system plays a fundamental role in blood pressure regulation. Lack of functional endothelin receptor B (ETB) evokes hypertension and salt sensitivity. GPER1 and ETB interact to promote urinary sodium excretion in female rats. We hypothesized that activation of GPER1 protects against hypertension and salt sensitivity induced by ETB antagonism in female rats. Female Sprague-Dawley rats were implanted with radiotelemetry. Animals were then subjected to ovariectomy and simultaneously implanted with minipumps to deliver either the GPER1 agonist G1 or its corresponding vehicle. Two weeks post surgery, we initiated treatment of rats with the ETB antagonist A-192621. Animals were maintained on a normal-salt diet and then challenged with a high-salt diet for an additional 5 days. Assessment of mean arterial blood pressure revealed an increase in vehicle-treated, but not G1-treated, rats in response to ovariectomy. A-192621 increased blood pressure in normal-salt diet-fed vehicle- and G1-treated rats. G1 improved the circadian blood pressure rhythms that were disrupted in A-192621-treated ovariectomized rats. Thus, although systemic GPER1 activation did not protect ovariectomized rats from hypertension and salt sensitivity induced by ETB antagonism, it maintained circadian blood pressure rhythms. Functional ETB is required to elicit the antihypertensive actions of GPER1. Additional studies are needed to improve our understanding of the interaction between G protein-coupled receptors in regulating circadian blood pressure rhythm.NEW & NOTEWORTHY Systemic G protein-coupled estrogen receptor 1 (GPER1) activation in rats prevents the increase in blood pressure evoked by ovariectomy. Blockade of endothelin receptor B negates the blood pressure-lowering impact of GPER1 in ovariectomized rats. Endothelin receptor B plays an important role in mediating the blood pressure-lowering action of GPER1 activation in female rats.
Collapse
Affiliation(s)
- Rawan N Almutlaq
- Cardiorenal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - David M Pollock
- Cardiorenal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Eman Y Gohar
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
6
|
Zhang G, Zeng C, Sun X, Zhang Q, Wang Y, Xia R, Mai Q, Xue G, Huang H, Wang F. Zearalenone modulates the function of goat endometrial cells via the mitochondrial quality control system. FASEB J 2024; 38:e23701. [PMID: 38941193 DOI: 10.1096/fj.202302198rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 06/30/2024]
Abstract
Zearalenone (ZEN) is a mycotoxin known for its estrogen-like effects, which can disrupt the normal physiological function of endometrial cells and potentially lead to abortion in female animals. However, the precise mechanism by which ZEN regulates endometrial function remains unclear. In this study, we found that the binding receptor estrogen receptors for ZEN is extensively expressed across various segments of the uterus and within endometrial cells, and a certain concentration of ZEN treatment reduced the proliferation capacity of goat endometrial epithelial cells (EECs) and endometrial stromal cells (ESCs). Meanwhile, cell cycle analysis revealed that ZEN treatment leaded to cell cycle arrest in goat EECs and ESCs. To explore the underlying mechanism, we investigated the mitochondrial quality control systems and observed that ZEN triggered excessive mitochondrial fission and disturbed the balance of mitochondrial fusion-fission dynamics, impaired mitochondrial biogenesis, increased mitochondrial unfolded protein response and mitophagy in goat EECs and ESCs. Additionally, ZEN treatment reduced the activities of mitochondrial respiratory chain complexes, heightened the production of hydrogen peroxide and reactive oxygen species, and caused cellular oxidative stress and mitochondrial dysfunction. These results suggest that ZEN has adverse effects on goat endometrium cells by disrupting the mitochondrial quality control system and affecting cell cycle and proliferation. Understanding the underlying molecular pathways involved in ZEN-induced mitochondrial dysfunction and its consequences on cell function will provide critical insights into the reproductive toxicity of ZEN and contribute to safeguarding the health and wellbeing of animals and humans exposed to this mycotoxin.
Collapse
Affiliation(s)
- Guomin Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Cheng Zeng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Xuan Sun
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Qi Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Yifei Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Rongxin Xia
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Qingyang Mai
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Gang Xue
- Yangtze River Delta White Goat Breeding Research Institute, Nantong, China
| | - Han Huang
- Mashan County Centre for Animal Disease Control and Prevention, Nanning, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
7
|
Jandhyam H, Mohanty BP, Parija SC. The vasodilator effect of Eugenol on uterine artery - potential therapeutic applications in pregnancy-associated hypertension. J Recept Signal Transduct Res 2024; 44:54-62. [PMID: 39185770 DOI: 10.1080/10799893.2024.2395301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
Preeclampsia, a gestational associated hypertension, has been reported in 6-8% of pregnant women worldwide leading to premature delivery and low birth weight of newborn due to reduced blood flow to placenta. Although several vasodilators (Methyl dopa, hydralazine, β-blockers and diuretics) are currently in use to treat preeclampsia, still there is a search for safer drugs with better efficacy. Lately, antihypertensive vasodilators from natural sources are gaining importance in treating preeclampsia. Eugenol (Eug), a natural essential oil, has been traditionally used in health and food products without any risk. In the present study, ex vivo experiments were designed to examine the vasorelaxation effect of Eug and its signaling pathways in a middle uterine artery (MUA) of pregnant Capra hircus (Ch). In presence of different blockers (L-NAME, indomethacin, ODQ, Ouabain, glibenclamide, 4-AP, Ba2, Carbenoxolone and 18β Glycyrrhetinic acid), Eug-induced concentration-dependent vasorelaxation response was elicited. The results showed that Eug caused a greater vasorelaxation effect in the MU of pregnant animals, which is mediated by potential activation of eNOS, KATP channels, and Kir channels with moderate activation of Na+- K+- ATPase and sGC and MEGJ. These findings provide a strong basis for developing Eug as a therapeutic candidate in the treatment of pregnancy-associated hypertension.
Collapse
Affiliation(s)
- Harithalakshmi Jandhyam
- Department of Pharmacology & Toxicology, College of Veterinary Science and Animal Husbandry, Orissa University of Agriculture and Technology, Bhubaneswar, India
| | - Bimal Prasanna Mohanty
- Indian Council of Agricultural and Research, Fisheries Science Division, Krishi Anusandhan Bhawan II, New Delhi, India
| | - Subas Chandra Parija
- Department of Pharmacology & Toxicology, College of Veterinary Science and Animal Husbandry, Orissa University of Agriculture and Technology, Bhubaneswar, India
| |
Collapse
|
8
|
Bai J, Li Y, Yan G, Zhou J, Salmeron AG, Fategbe OT, Kumar S, Chen X, Chen DB. ICI 182,780 Attenuates Selective Upregulation of Uterine Artery Cystathionine β-Synthase Expression in Rat Pregnancy. Int J Mol Sci 2023; 24:14384. [PMID: 37762687 PMCID: PMC10532247 DOI: 10.3390/ijms241814384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/16/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Endogenous hydrogen sulfide (H2S) produced by cystathionine β-synthase (CBS) and cystathionine-γ lyase (CSE) has emerged as a novel uterine vasodilator contributing to pregnancy-associated increases in uterine blood flow, which safeguard pregnancy health. Uterine artery (UA) H2S production is stimulated via exogenous estrogen replacement and is associated with elevated endogenous estrogens during pregnancy through the selective upregulation of CBS without altering CSE. However, how endogenous estrogens regulate uterine artery CBS expression in pregnancy is unknown. This study was conducted to test a hypothesis that endogenous estrogens selectively stimulate UA CBS expression via specific estrogen receptors (ER). Treatment with E2β (0.01 to 100 nM) stimulated CBS but not CSE mRNA in organ cultures of fresh UA rings from both NP and P (gestational day 20, GD20) rats, with greater responses to all doses of E2β tested in P vs. NP UA. ER antagonist ICI 182,780 (ICI, 1 µM) completely attenuated E2β-stimulated CBS mRNA in both NP and P rat UA. Subcutaneous injection with ICI 182,780 (0.3 mg/rat) of GD19 P rats for 24 h significantly inhibited UA CBS but not mRNA expression, consistent with reduced endothelial and smooth muscle cell CBS (but not CSE) protein. ICI did not alter mesenteric and renal artery CBS and CSE mRNA. In addition, ICI decreased endothelial nitric oxide synthase mRNA in UA but not in mesenteric or renal arteries. Thus, pregnancy-augmented UA CBS/H2S production is mediated by the actions of endogenous estrogens via specific ER in pregnant rats.
Collapse
Affiliation(s)
- Jin Bai
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| | - Yao Li
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Guofeng Yan
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Jing Zhou
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Alejandra Garcia Salmeron
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| | - Olamide Tolulope Fategbe
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| | - Sathish Kumar
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Xuejin Chen
- Department of Laboratory Animal Sciences, School of Medicine, Shanghai Jiaotong University, 280 South Chongqing Road, Shanghai 200025, China; (Y.L.); (G.Y.); (J.Z.); (X.C.)
| | - Dong-Bao Chen
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, CA 92697, USA; (J.B.); (A.G.S.); (O.T.F.)
| |
Collapse
|
9
|
Xu F, Ma J, Wang X, Wang X, Fang W, Sun J, Li Z, Liu J. The Role of G Protein-Coupled Estrogen Receptor (GPER) in Vascular Pathology and Physiology. Biomolecules 2023; 13:1410. [PMID: 37759810 PMCID: PMC10526873 DOI: 10.3390/biom13091410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
OBJECTIVE Estrogen is indispensable in health and disease and mainly functions through its receptors. The protection of the cardiovascular system by estrogen and its receptors has been recognized for decades. Numerous studies with a focus on estrogen and its receptor system have been conducted to elucidate the underlying mechanism. Although nuclear estrogen receptors, including estrogen receptor-α and estrogen receptor-β, have been shown to be classical receptors that mediate genomic effects, studies now show that GPER mainly mediates rapid signaling events as well as transcriptional regulation via binding to estrogen as a membrane receptor. With the discovery of selective synthetic ligands for GPER and the utilization of GPER knockout mice, significant progress has been made in understanding the function of GPER. In this review, the tissue and cellular localizations, endogenous and exogenous ligands, and signaling pathways of GPER are systematically summarized in diverse physiological and diseased conditions. This article further emphasizes the role of GPER in vascular pathology and physiology, focusing on the latest research progress and evidence of GPER as a promising therapeutic target in hypertension, pulmonary hypertension, and atherosclerosis. Thus, selective regulation of GPER by its agonists and antagonists have the potential to be used in clinical practice for treating such diseases.
Collapse
Affiliation(s)
- Fujie Xu
- Xi’an Medical University, Xi’an 710068, China; (F.X.); (W.F.); (J.S.)
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (J.M.); (X.W.); (X.W.)
| | - Jipeng Ma
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (J.M.); (X.W.); (X.W.)
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (J.M.); (X.W.); (X.W.)
| | - Xiaoya Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (J.M.); (X.W.); (X.W.)
| | - Weiyi Fang
- Xi’an Medical University, Xi’an 710068, China; (F.X.); (W.F.); (J.S.)
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (J.M.); (X.W.); (X.W.)
| | - Jingwei Sun
- Xi’an Medical University, Xi’an 710068, China; (F.X.); (W.F.); (J.S.)
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (J.M.); (X.W.); (X.W.)
| | - Zilin Li
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (J.M.); (X.W.); (X.W.)
| | - Jincheng Liu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (J.M.); (X.W.); (X.W.)
| |
Collapse
|
10
|
Alencar AKN, Swan KF, Pridjian G, Lindsey SH, Bayer CL. Connecting G protein-coupled estrogen receptor biomolecular mechanisms with the pathophysiology of preeclampsia: a review. Reprod Biol Endocrinol 2023; 21:60. [PMID: 37393260 DOI: 10.1186/s12958-023-01112-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND Throughout the course of pregnancy, small maternal spiral arteries that are in contact with fetal tissue undergo structural remodeling, lose smooth muscle cells, and become less responsive to vasoconstrictors. Additionally, placental extravillous trophoblasts invade the maternal decidua to establish an interaction between the fetal placental villi with the maternal blood supply. When successful, this process enables the transport of oxygen, nutrients, and signaling molecules but an insufficiency leads to placental ischemia. In response, the placenta releases vasoactive factors that enter the maternal circulation and promote maternal cardiorenal dysfunction, a hallmark of preeclampsia (PE), the leading cause of maternal and fetal death. An underexplored mechanism in the development of PE is the impact of membrane-initiated estrogen signaling via the G protein-coupled estrogen receptor (GPER). Recent evidence indicates that GPER activation is associated with normal trophoblast invasion, placental angiogenesis/hypoxia, and regulation of uteroplacental vasodilation, and these mechanisms could explain part of the estrogen-induced control of uterine remodeling and placental development in pregnancy. CONCLUSION Although the relevance of GPER in PE remains speculative, this review provides a summary of our current understanding on how GPER stimulation regulates some of the features of normal pregnancy and a potential link between its signaling network and uteroplacental dysfunction in PE. Synthesis of this information will facilitate the development of innovative treatment options.
Collapse
Affiliation(s)
| | - Kenneth F Swan
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Gabriella Pridjian
- Department of Obstetrics & Gynecology, Tulane University, New Orleans, LA, 70112, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, 70112, USA
| | - Carolyn L Bayer
- Department of Biomedical Engineering, Tulane University, 500 Lindy Boggs Center, New Orleans, LA, 70118, USA.
| |
Collapse
|
11
|
Prossnitz ER, Barton M. The G protein-coupled oestrogen receptor GPER in health and disease: an update. Nat Rev Endocrinol 2023:10.1038/s41574-023-00822-7. [PMID: 37193881 DOI: 10.1038/s41574-023-00822-7] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 05/18/2023]
Abstract
Oestrogens and their receptors contribute broadly to physiology and diseases. In premenopausal women, endogenous oestrogens protect against cardiovascular, metabolic and neurological diseases and are involved in hormone-sensitive cancers such as breast cancer. Oestrogens and oestrogen mimetics mediate their effects via the cytosolic and nuclear receptors oestrogen receptor-α (ERα) and oestrogen receptor-β (ERβ) and membrane subpopulations as well as the 7-transmembrane G protein-coupled oestrogen receptor (GPER). GPER, which dates back more than 450 million years in evolution, mediates both rapid signalling and transcriptional regulation. Oestrogen mimetics (such as phytooestrogens and xenooestrogens including endocrine disruptors) and licensed drugs such as selective oestrogen receptor modulators (SERMs) and downregulators (SERDs) also modulate oestrogen receptor activity in both health and disease. Following up on our previous Review of 2011, we herein summarize the progress made in the field of GPER research over the past decade. We will review molecular, cellular and pharmacological aspects of GPER signalling and function, its contribution to physiology, health and disease, and the potential of GPER to serve as a therapeutic target and prognostic indicator of numerous diseases. We also discuss the first clinical trial evaluating a GPER-selective drug and the opportunity of repurposing licensed drugs for the targeting of GPER in clinical medicine.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland.
- Andreas Grüntzig Foundation, Zürich, Switzerland.
| |
Collapse
|
12
|
Dela Justina V, Dos Passos Júnior RR, Lima VV, Giachini FR. Evidence of Nitric Oxide Impairment During Hypertensive Pregnancies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:99-125. [PMID: 37466771 DOI: 10.1007/978-3-031-32554-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Hypertensive disorders of pregnancy complicate up to 10% of pregnancies worldwide, and they can be classified into (1) gestational hypertension, (2) preeclampsia, (3) chronic hypertension and (4) chronic hypertension with preeclampsia. Nitric oxide (NO) plays an essential role in the haemodynamic adaptations observed during pregnancy. It has been shown that the nitric oxide pathway's dysfunction during pregnancy is associated with placental- and vascular-related diseases such as hypertensive disorders of pregnancy. This review aims to present a brief definition of hypertensive disorders of pregnancy and physiological maternal cardiovascular adaptations during pregnancy. We also detail how NO signalling is altered in the (a) systemic vasculature, (b) uterine artery/spiral arteries, (c) implantation and (d) placenta of hypertensive disorders during pregnancy. We conclude by summarizing the anti-hypertensive therapy of hypertensive disorders of pregnancy as a specific management strategy.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Rinaldo Rodrigues Dos Passos Júnior
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| | - Victor Vitorino Lima
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| | - Fernanda Regina Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| |
Collapse
|
13
|
Ortiz-Cerda T, Mosso C, Alcudia A, Vázquez-Román V, González-Ortiz M. Pathophysiology of Preeclampsia and L-Arginine/L-Citrulline Supplementation as a Potential Strategy to Improve Birth Outcomes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:127-148. [PMID: 37466772 DOI: 10.1007/978-3-031-32554-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
In preeclampsia, the shallow invasion of cytotrophoblast cells to uterine spiral arteries, leading to a reduction in placental blood flow, is associated with an imbalance of proangiogenic/antiangiogenic factors to impaired nitric oxide (NO) production. Proangiogenic factors, such as vascular endothelial growth factor (VEGF) and placental growth factor (PlGF), require NO to induce angiogenesis through antioxidant regulation mechanisms. At the same time, there are increases in antiangiogenic factors in preeclampsia, such as soluble fms-like tyrosine kinase type 1 receptor (sFIt1) and toll-like receptor 9 (TLR9), which are mechanism derivates in the reduction of NO bioavailability and oxidative stress in placenta.Different strategies have been proposed to prevent or alleviate the detrimental effects of preeclampsia. However, the only intervention to avoid the severe consequences of the disease is the interruption of pregnancy. In this scenario, different approaches have been analysed to treat preeclamptic pregnant women safely. The supplementation with amino acids is one of them, especially those associated with NO synthesis. In this review, we discuss emerging concepts in the pathogenesis of preeclampsia to highlight L-arginine and L-citrulline supplementation as potential strategies to improve birth outcomes. Clinical and experimental data concerning L-arginine and L-citrulline supplementation have shown benefits in improving NO availability in the placenta and uterine-placental circulation, prolonging pregnancy in patients with gestational hypertension and decreasing maternal blood pressure.
Collapse
Affiliation(s)
- Tamara Ortiz-Cerda
- Departamento de Citología e Histología Normal y Patológica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Constanza Mosso
- Departamento de Nutrición y Dietética, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Ana Alcudia
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Victoria Vázquez-Román
- Departamento de Citología e Histología Normal y Patológica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Marcelo González-Ortiz
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
14
|
Rodríguez-Jaimes SY, Hernández-Hernández GC, Hernández-Aragón LG, Sánchez-García O, Martínez-Gómez M, Cuevas-Romero E, Castelán F. G protein-coupled estrogen receptor (GPER/GPR30) levels in pelvic floor muscles and its association with estrogen status in female rabbits. Gynecol Endocrinol 2022; 38:748-753. [PMID: 35861367 DOI: 10.1080/09513590.2022.2099830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Objective: To assess the relative expression of the G-protein coupled estrogen receptor (GPER) in the bulbospongiosus (Bsm) and pubococcygeus (Pcm) muscles in control, ovariectomized (OVX), and OVX with estradiol benzoate supplementation (OVX + EB) rabbits.Methods: We used tissues from C, 1-month OVX, and OVX plus 15-day EB implanted (OVX + EB) groups. The GPER expression was evaluated by Western blot and immunohistochemistry for both Bsm and Pcm. Results: Both muscles showed a GPER immunoreactivity in blood vessels, inside myofibers next to myonuclei, and in polymorphonuclear cells. Four-week ovariectomy did not modify the GPER expression in the Bsm and Pcm, but two-week estradiol benzoate increased it in the latter muscle alone.Conclusions: We demonstrated that the Bsm and Pcm of female rabbits express GPER. High serum estradiol levels elevate GPER relative expression in the Pcm alone. The present study supports the remarkable estrogen sensitivity of the Pcm.
Collapse
Affiliation(s)
- Sharet Y Rodríguez-Jaimes
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Guadalupe C Hernández-Hernández
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México
- Maestría en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | | | | | - Margarita Martínez-Gómez
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México
- Depto. de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlaxcala, México
| | - Estela Cuevas-Romero
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Francisco Castelán
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, México
- Depto. de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlaxcala, México
| |
Collapse
|
15
|
Peixoto P, Vieira-Alves I, Couto GK, Lemos VS, Rossoni LV, Bissoli NS, Dos Santos RL. Sex differences in the participation of endothelial mediators and signaling pathways involved in the vasodilator effect of a selective GPER agonist in resistance arteries of gonadectomized Wistar rats. Life Sci 2022; 308:120917. [PMID: 36044974 DOI: 10.1016/j.lfs.2022.120917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/16/2022] [Accepted: 08/24/2022] [Indexed: 11/15/2022]
Abstract
AIM Endothelial mechanisms underlying the vascular effects of estrogen modulated by the G protein-coupled estrogen receptor (GPER) are not well understood, especially in gonadal sex hormone deprivation. Thus, we investigated vascular function and endothelial signaling pathways involved in the selective activation of GPER in resistance arteries of gonadectomized rats. METHODS Gonadectomy was performed in Wistar rats of both sexes. After 21 days, the animals were euthanized. Concentration-response curves were obtained by cumulative additions of G-1 in third-order mesenteric arteries. The vasodilatory effects of G-1 were evaluated before and after endothelium removal or incubation with pharmacological inhibitors. Tissue protein expression was measured by western blotting. Assays with 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM) and 2',7' dichlorodihydrofluorescein-diacetate (DCF-DA) were performed in the arteries investigated. Immunolocalization was assessed by immunofluorescence. RESULTS G-1 induced partially endothelium-dependent relaxation in both sexes. The three isoforms of the enzyme nitric oxide synthase contributed to the production and release of nitric oxide in both gonadectomized groups, but the role of inducible nitric oxide synthase is more expressive in males. The mechanistic pathway by which endothelial nitric oxide synthase is phosphorylated appears to differ between sexes, with the rapid signaling pathway phosphatidylinositol-3-kinase/protein kinase B/endothelial nitric oxide synthase (PI3k-Akt-eNOS) being identified for males and mitogen-activated protein kinase/extracellular signal-regulated kinase/endothelial nitric oxide synthase (MEK-ERK-eNOS) for females. The contribution of hydrogen peroxide as an endothelial relaxation mediator seems to be greater in females. CONCLUSION These results provide new insights into the effects of estrogen-induced responses via GPER on vascular function in gonadal sex hormone deprivation.
Collapse
Affiliation(s)
- Pollyana Peixoto
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitoria, ES, Brazil
| | - Ildernandes Vieira-Alves
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gisele Kruger Couto
- Department of Physiology and Biophysics, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Virgínia Soares Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Nazaré Souza Bissoli
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitoria, ES, Brazil
| | - Roger Lyrio Dos Santos
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitoria, ES, Brazil.
| |
Collapse
|
16
|
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:ijms23115996. [PMID: 35682675 PMCID: PMC9180712 DOI: 10.3390/ijms23115996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
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.
Collapse
|
17
|
Singh R, Nasci VL, Guthrie G, Ertuglu LA, Butt MK, Kirabo A, Gohar EY. Emerging Roles for G Protein-Coupled Estrogen Receptor 1 in Cardio-Renal Health: Implications for Aging. Biomolecules 2022; 12:412. [PMID: 35327604 PMCID: PMC8946600 DOI: 10.3390/biom12030412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular (CV) and renal diseases are increasingly prevalent in the United States and globally. CV-related mortality is the leading cause of death in the United States, while renal-related mortality is the 8th. Despite advanced therapeutics, both diseases persist, warranting continued exploration of disease mechanisms to develop novel therapeutics and advance clinical outcomes for cardio-renal health. CV and renal diseases increase with age, and there are sex differences evident in both the prevalence and progression of CV and renal disease. These age and sex differences seen in cardio-renal health implicate sex hormones as potentially important regulators to be studied. One such regulator is G protein-coupled estrogen receptor 1 (GPER1). GPER1 has been implicated in estrogen signaling and is expressed in a variety of tissues including the heart, vasculature, and kidney. GPER1 has been shown to be protective against CV and renal diseases in different experimental animal models. GPER1 actions involve multiple signaling pathways: interaction with aldosterone and endothelin-1 signaling, stimulation of the release of nitric oxide, and reduction in oxidative stress, inflammation, and immune infiltration. This review will discuss the current literature regarding GPER1 and cardio-renal health, particularly in the context of aging. Improving our understanding of GPER1-evoked mechanisms may reveal novel therapeutics aimed at improving cardio-renal health and clinical outcomes in the elderly.
Collapse
Affiliation(s)
- Ravneet Singh
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Medical Research Building IV, Nashville, TN 37232, USA; (R.S.); (V.L.N.)
| | - Victoria L. Nasci
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Medical Research Building IV, Nashville, TN 37232, USA; (R.S.); (V.L.N.)
| | - Ginger Guthrie
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (G.G.); (M.K.B.)
| | - Lale A. Ertuglu
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.A.E.); (A.K.)
| | - Maryam K. Butt
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (G.G.); (M.K.B.)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (L.A.E.); (A.K.)
| | - Eman Y. Gohar
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Medical Research Building IV, Nashville, TN 37232, USA; (R.S.); (V.L.N.)
| |
Collapse
|
18
|
Estrogen-Induced Uterine Vasodilation in Pregnancy and Preeclampsia. MATERNAL-FETAL MEDICINE 2022; 4:52-60. [PMID: 35072088 PMCID: PMC8772435 DOI: 10.1097/fm9.0000000000000132] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/09/2021] [Indexed: 12/02/2022] Open
Abstract
Normal pregnancy is associated with dramatically increased estrogen biosynthesis whose role is believed to raise uterine blood flow to facilitate the bi-directional maternal-fetal exchanges of gases (O2 and CO2), to deliver nutrients, and exhaust wastes to support fetal development and survival. Constrained uterine blood flow in pregnancy is a leading cause of preeclampsia with fetal growth restriction, rendering investigations of uterine hemodynamics to hold a high promise to inform pathways as targets for therapeutic interventions for preeclampsia. The mechanisms of estrogen-induced uterine vasodilation in pregnancy have long been attributed to enhanced endothelium production of nitric oxide, but clinical trials targeting this pathway that dominates uterine hemodynamics have achieved no to little success. Emerging evidence has recently shown a novel proangiogenic vasodilatory role of hydrogen sulfide in regulating uterine hemodynamics in pregnancy and preeclampsia, provoking a new field of perinatal research in searching for alternative pathways for pregnancy disorders especially preeclampsia and intrauterine growth restriction. This minireview is intended to summarize the nitric oxide pathway and to discuss the emerging hydrogen sulfide pathway in modulating estrogen-induced uterine vasodilation in pregnancy and preeclampsia.
Collapse
|
19
|
Favre J, Vessieres E, Guihot AL, Proux C, Grimaud L, Rivron J, Garcia MC, Réthoré L, Zahreddine R, Davezac M, Fébrissy C, Adlanmerini M, Loufrani L, Procaccio V, Foidart JM, Flouriot G, Lenfant F, Fontaine C, Arnal JF, Henrion D. Membrane estrogen receptor alpha (ERα) participates in flow-mediated dilation in a ligand-independent manner. eLife 2021; 10:68695. [PMID: 34842136 PMCID: PMC8676342 DOI: 10.7554/elife.68695] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Estrogen receptor alpha (ERα) activation by estrogens prevents atheroma through its nuclear action, whereas plasma membrane-located ERα accelerates endothelial healing. The genetic deficiency of ERα was associated with a reduction in flow-mediated dilation (FMD) in one man. Here, we evaluated ex vivo the role of ERα on FMD of resistance arteries. FMD, but not agonist (acetylcholine, insulin)-mediated dilation, was reduced in male and female mice lacking ERα (Esr1-/- mice) compared to wild-type mice and was not dependent on the presence of estrogens. In C451A-ERα mice lacking membrane ERα, not in mice lacking AF2-dependent nuclear ERα actions, FMD was reduced, and restored by antioxidant treatments. Compared to wild-type mice, isolated perfused kidneys of C451A-ERα mice revealed a decreased flow-mediated nitrate production and an increased H2O2 production. Thus, endothelial membrane ERα promotes NO bioavailability through inhibition of oxidative stress and thereby participates in FMD in a ligand-independent manner.
Collapse
Affiliation(s)
- Julie Favre
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Emilie Vessieres
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Anne-Laure Guihot
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Coralyne Proux
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Linda Grimaud
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Jordan Rivron
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Manuela Cl Garcia
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France
| | - Léa Réthoré
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France
| | - Rana Zahreddine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Morgane Davezac
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Chanaelle Fébrissy
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Marine Adlanmerini
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Laurent Loufrani
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Vincent Procaccio
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| | - Jean-Michel Foidart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Université de Liège, Liège, Belgium
| | - Gilles Flouriot
- INSERM U1085, IRSET (Institut de Recherche en Santé, Environnement et Travail), University of Rennes, Rennes, France
| | - Françoise Lenfant
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Coralie Fontaine
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Jean-François Arnal
- INSERM U1297, Paul Sabatier University (Toulouse III) , University Hospital (UHC) of Toulouse, Toulouse, France
| | - Daniel Henrion
- Angers University, MITOVASC, CNRS UMR 6015, INSERM U1083, Angers, France.,CARFI facility, Angers University, Angers, France.,University Hospital (CHU) of Angers, Angers, France
| |
Collapse
|
20
|
Hu X, Zhang L. Uteroplacental Circulation in Normal Pregnancy and Preeclampsia: Functional Adaptation and Maladaptation. Int J Mol Sci 2021; 22:8622. [PMID: 34445328 PMCID: PMC8395300 DOI: 10.3390/ijms22168622] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023] Open
Abstract
Uteroplacental blood flow increases as pregnancy advances. Adequate supply of nutrients and oxygen carried by uteroplacental blood flow is essential for the well-being of the mother and growth/development of the fetus. The uteroplacental hemodynamic change is accomplished primarily through uterine vascular adaptation, involving hormonal regulation of myogenic tone, vasoreactivity, release of vasoactive factors and others, in addition to the remodeling of spiral arteries. In preeclampsia, hormonal and angiogenic imbalance, proinflammatory cytokines and autoantibodies cause dysfunction of both endothelium and vascular smooth muscle cells of the uteroplacental vasculature. Consequently, the vascular dysfunction leads to increased vascular resistance and reduced blood flow in the uteroplacental circulation. In this article, the (mal)adaptation of uteroplacental vascular function in normal pregnancy and preeclampsia and underlying mechanisms are reviewed.
Collapse
Affiliation(s)
- Xiangqun Hu
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|
21
|
Harding AT, Goff MA, Froggatt HM, Lim JK, Heaton NS. GPER1 is required to protect fetal health from maternal inflammation. Science 2021; 371:271-276. [PMID: 33446553 DOI: 10.1126/science.aba9001] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/23/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
Type I interferon (IFN) signaling in fetal tissues causes developmental abnormalities and fetal demise. Although pathogens that infect fetal tissues can induce birth defects through the local production of type I IFN, it remains unknown why systemic IFN generated during maternal infections only rarely causes fetal developmental defects. Here, we report that activation of the guanine nucleotide-binding protein-coupled estrogen receptor 1 (GPER1) during pregnancy is both necessary and sufficient to suppress IFN signaling and does so disproportionately in reproductive and fetal tissues. Inactivation of GPER1 in mice halted fetal development and promoted fetal demise, but only in the context of maternal inflammation. Thus, GPER1 is a central regulator of IFN signaling during pregnancy that allows dynamic antiviral responses in maternal tissues while also preserving fetal health.
Collapse
Affiliation(s)
- Alfred T Harding
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Marisa A Goff
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Heather M Froggatt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicholas S Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA. .,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
22
|
Gohar EY, Pollock DM. Functional Interaction of Endothelin Receptors in Mediating Natriuresis Evoked by G Protein-Coupled Estrogen Receptor 1. J Pharmacol Exp Ther 2021; 376:98-105. [PMID: 33127751 PMCID: PMC7788354 DOI: 10.1124/jpet.120.000322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/26/2020] [Indexed: 01/14/2023] Open
Abstract
The G protein-coupled estrogen receptor 1 (GPER1) mediates rapid estrogenic signaling. We recently reported that activation of GPER1 in the renal medulla evokes endothelin-1-dependent natriuresis in female, but not male, rats. However, the involvement of the ET receptors, ETA and ETB, underlying GPER1 natriuretic action remain unclear. In this study, we used genetic and pharmacologic methods to identify the contributions of ETA and ETB in mediating this female-specific natriuretic effect of renal medullary GPER1. Infusion of the GPER1-selective agonist G1 (5 pmol/kg per minute) into the renal medulla for 40 minutes increased Na+ excretion and urine flow in anesthetized female ETB-deficient (ETB def) rats and littermate controls but did not affect blood pressure or urinary K+ excretion in either group. Pretreatment with the selective ETA inhibitor ABT-627 (5 mg/kg, intravenous) abolished G1-induced natriuresis in ETB def rats. To further isolate the effects of inhibiting either receptor alone, we conducted the same experiments in anesthetized female Sprague-Dawley (SD) rats pretreated or not with ABT-627 and/or the selective ETB inhibitor A-192621 (10 mg/kg, intravenous). Neither antagonism of ETA nor antagonism of ETB receptor alone affected the G1-induced increase in Na+ excretion and urine flow in SD rats. However, simultaneous antagonism of both receptors completely abolished these effects. These data suggest that ETA and ETB receptors can mediate the natriuretic and diuretic response to renal medullary GPER1 activation in female rats. SIGNIFICANCE STATEMENT: Activation of G protein-coupled estrogen receptor 1 (GPER1) in the renal medulla of female rats evokes natriuresis via endothelin receptors A and/or B, suggesting that GPER1 and endothelin signaling pathways help efficient sodium excretion in females. Thus, GPER1 activation could be potentially useful to mitigate salt sensitivity in females.
Collapse
Affiliation(s)
- Eman Y Gohar
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Alabama (E.Y.G, D.M.P); and Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (E.Y.G)
| | - David M Pollock
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Alabama (E.Y.G, D.M.P); and Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (E.Y.G)
| |
Collapse
|
23
|
Camilleri C, Buskmiller C, Sammut S. Pregnancy-induced long-term uterine vascular remodeling in the rat. Reprod Biol 2020; 21:100466. [PMID: 33279772 DOI: 10.1016/j.repbio.2020.100466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 10/26/2020] [Accepted: 11/15/2020] [Indexed: 10/22/2022]
Abstract
Despite the available research investigating uterine physiology during and immediately following pregnancy, including at the vascular and muscular levels, knowledge of the potential long-term timeline of such changes is limited. Thus, our study sought to investigate the potential long-term changes in uterine vasculature and horn length in the postpartum rat, following delivery and weaning. Female Long-Evans rats (n = 9-11 rats/group/timepoint) were divided into two groups: a pregnant group and an age-matched virgin control group. Rat weight, food consumption and vaginal impedance measurements were recorded throughout the experiment. Rats in the pregnant group were bred and pregnancy was confirmed using ultrasound imaging. The uterus and its vasculature were collected at various timepoints following weaning: 3 (week of weaning), 8-9 and 13 weeks postpartum, and at age-equivalent timepoints in the virgin group, and the diameters of the main uterine artery and vein, and lengths of the mesometrial segmental vessels (MSV) and uterine horns were recorded. The results indicated a significant difference between the previously-pregnant and virgin rats in both internal and external arterial diameters (but not arterial wall thickness), as well as the uterine horn length, 3 weeks postpartum, but not 8-9 and 13 weeks postpartum. Significant differences were observed in both venous diameter and MSV length at all timepoints measured. Placental scars were also observed in previously-pregnant rats at all timepoints measured. Our study highlights the long-term impact of pregnancy on the uterine vasculature and the necessity for consideration of such changes on subsequent pregnancies, as well as pregnancy-related vascular pathologies.
Collapse
Affiliation(s)
- Christina Camilleri
- Department of Psychology, Franciscan University of Steubenville, 1235 University Blvd, Steubenville, OH, 43952, USA
| | - Cara Buskmiller
- Department of Obstetrics, Gynecology, and Women's Health, Saint Louis University, 6420 Clayton Rd, Ste 240, St. Louis, MO, 63117, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), 6431 Fannin, Suite MSB3.262 Houston, TX 77030, USA
| | - Stephen Sammut
- Department of Psychology, Franciscan University of Steubenville, 1235 University Blvd, Steubenville, OH, 43952, USA.
| |
Collapse
|
24
|
Tropea T, Greenwood SL, Sibley CP, Cottrell EC. Grape Seed Extract Polyphenols Improve Resistance Artery Function in Pregnant eNOS -/- Mice. Front Physiol 2020; 11:588000. [PMID: 33240108 PMCID: PMC7677241 DOI: 10.3389/fphys.2020.588000] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/15/2020] [Indexed: 12/22/2022] Open
Abstract
Hypertension during pregnancy is a leading cause of maternal and fetal morbidity and mortality worldwide, increasing the risk of complications including preeclampsia, intracerebral hemorrhage and fetal growth restriction. Increased oxidative stress is known to contribute to poor vascular function; however, trials of antioxidant supplementation have raised concerns about fetal outcomes, including risk of low birthweight. Grape seed extract polyphenols (GSEP) have been suggested to promote cardiovascular protection, at least in part through antioxidant actions. We tested the hypothesis that administration of GSEP during pregnancy would reduce oxidative stress and improve resistance artery function with no detrimental effects on fetal growth, in an established model of maternal hypertension associated with vascular dysfunction, the endothelial NO synthase knockout (eNOS-/-) mouse. Pregnant C57BL/6J (WT) and eNOS-/- mice received either GSEP (200 mg/kg/day) or drinking water, between gestational (GD) day 10.5 and GD18.5. At GD17.5, maternal systolic blood pressure (SBP) was measured; at GD18.5, maternal malondialdehyde (MDA) concentrations, vascular function of aortic, mesenteric, uterine and posterior cerebral arteries was assessed, and fetal outcome evaluated. GSEP reduced maternal SBP (P < 0.01) and plasma MDA concentrations (P < 0.01) in eNOS-/- mice. Whilst there was no effect of GSEP on vascular reactivity of aortas, GSEP improved endothelial-dependent relaxation in mesenteric and uterine arteries of eNOS-/- mice (P < 0.05 and P < 0.001, respectively) and normalized lumen diameters of pressurized posterior cerebral arteries in eNOS-/- mice (P < 0.001). Supplementation with GSEP had no effect in WT mice and did not affect fetal outcomes in either genotype. Our data suggest that GSEP improve resistance artery function, potentially through antioxidant actions, and provide a basis to further investigate these beneficial effects including in the prevention of intracerebral hemorrhage. Maternal supplementation with GSEP may be a safe intervention to improve outcomes in pregnancies associated with hypertension and vascular dysfunction.
Collapse
Affiliation(s)
- Teresa Tropea
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, United Kingdom
| | - Susan L Greenwood
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, United Kingdom
| | - Colin P Sibley
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, United Kingdom
| | - Elizabeth C Cottrell
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, United Kingdom
| |
Collapse
|
25
|
Chertok VM, Khramova IA, Kotsyuba AE. Dynamics of Organization of the Microcirculatory Bed and Mast Cells of the Uterus in Rats at Different Times of the Day. Bull Exp Biol Med 2020; 169:710-713. [PMID: 32990855 DOI: 10.1007/s10517-020-04961-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Indexed: 10/23/2022]
Abstract
Structural organization of the microcirculatory bed and mast cells in the uterus of mature female Wistar rats (n=60) were examined every 2 h over 24 h. During night hours, many vessels characterized by intensive reaction to NADPH-diaphorase and endothelial NO synthase were detected in the endometrium, and the density of these vessels and mast cells was quite high. During the day and evening hours, the capillaries with high levels of enzymes involved in NO synthesis and mast cells were less numerous. NO directly regulating the diameter of arteries and the intensity of transcapillary exchange can act as a signal molecule in the temporal dynamics of the vasoceptive action of estrogens and their receptors in the uterus.
Collapse
Affiliation(s)
- V M Chertok
- Department of Human Anatomy, Vladivostok, Russia.
| | - I A Khramova
- Department of Obstetrics and Gynecology, Pacific State Medical University, Ministry of Health of the Russian Federation, Vladivostok, Russia
| | - A E Kotsyuba
- Department of Human Anatomy, Vladivostok, Russia
| |
Collapse
|
26
|
Gohar EY. G protein-coupled estrogen receptor 1 as a novel regulator of blood pressure. Am J Physiol Renal Physiol 2020; 319:F612-F617. [PMID: 32893662 DOI: 10.1152/ajprenal.00045.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The mechanisms underlying hypertension are multifaceted and incompletely understood. New evidence suggests that G protein-coupled estrogen receptor 1 (GPER1) mediates protective actions within the cardiovascular and renal systems. This mini-review focuses on recent advancements in our understanding of the vascular, renal, and cardiac GPER1-mediated mechanisms that influence blood pressure regulation. We emphasize clinical and basic evidence that suggests GPER1 as a novel target to aid therapeutic strategies for hypertension. Furthermore, we discuss current controversies and challenges facing GPER1-related research.
Collapse
Affiliation(s)
- Eman Y Gohar
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
27
|
Estrogen Receptors and Estrogen-Induced Uterine Vasodilation in Pregnancy. Int J Mol Sci 2020; 21:ijms21124349. [PMID: 32570961 PMCID: PMC7352873 DOI: 10.3390/ijms21124349] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
Normal pregnancy is associated with dramatic increases in uterine blood flow to facilitate the bidirectional maternal–fetal exchanges of respiratory gases and to provide sole nutrient support for fetal growth and survival. The mechanism(s) underlying pregnancy-associated uterine vasodilation remain incompletely understood, but this is associated with elevated estrogens, which stimulate specific estrogen receptor (ER)-dependent vasodilator production in the uterine artery (UA). The classical ERs (ERα and ERβ) and the plasma-bound G protein-coupled ER (GPR30/GPER) are expressed in UA endothelial cells and smooth muscle cells, mediating the vasodilatory effects of estrogens through genomic and/or nongenomic pathways that are likely epigenetically modified. The activation of these three ERs by estrogens enhances the endothelial production of nitric oxide (NO), which has been shown to play a key role in uterine vasodilation during pregnancy. However, the local blockade of NO biosynthesis only partially attenuates estrogen-induced and pregnancy-associated uterine vasodilation, suggesting that mechanisms other than NO exist to mediate uterine vasodilation. In this review, we summarize the literature on the role of NO in ER-mediated mechanisms controlling estrogen-induced and pregnancy-associated uterine vasodilation and our recent work on a “new” UA vasodilator hydrogen sulfide (H2S) that has dramatically changed our view of how estrogens regulate uterine vasodilation in pregnancy.
Collapse
|
28
|
Gohar EY, Daugherty EM, Aceves JO, Sedaka R, Obi IE, Allan JM, Soliman RH, Jin C, De Miguel C, Lindsey SH, Pollock JS, Pollock DM. Evidence for G-Protein-Coupled Estrogen Receptor as a Pronatriuretic Factor. J Am Heart Assoc 2020; 9:e015110. [PMID: 32390531 PMCID: PMC7660860 DOI: 10.1161/jaha.119.015110] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/02/2020] [Indexed: 12/18/2022]
Abstract
Background The novel estrogen receptor, G-protein-coupled estrogen receptor (GPER), is responsible for rapid estrogen signaling. GPER activation elicits cardiovascular and nephroprotective effects against salt-induced complications, yet there is no direct evidence for GPER control of renal Na+ handling. We hypothesized that GPER activation in the renal medulla facilitates Na+ excretion. Methods and Results Herein, we show that infusion of the GPER agonist, G1, to the renal medulla increased Na+ excretion in female Sprague Dawley rats, but not male rats. We found that GPER mRNA expression and protein abundance were markedly higher in outer medullary tissues from females relative to males. Blockade of GPER in the renal medulla attenuated Na+ excretion in females. Given that medullary endothelin 1 is a well-established natriuretic factor that is regulated by sex and sex steroids, we hypothesized that GPER activation promotes natriuresis via an endothelin 1-dependent pathway. To test this mechanism, we determined the effect of medullary infusion of G1 after blockade of endothelin receptors. Dual endothelin receptor subtype A and endothelin receptor subtype B antagonism attenuated G1-induced natriuresis in females. Unlike males, female mice with genetic deletion of GPER had reduced endothelin 1, endothelin receptor subtype A, and endothelin receptor subtype B mRNA expression compared with wild-type controls. More important, we found that systemic GPER activation ameliorates the increase in mean arterial pressure induced by ovariectomy. Conclusions Our data uncover a novel role for renal medullary GPER in promoting Na+ excretion via an endothelin 1-dependent pathway in female rats, but not in males. These results highlight GPER as a potential therapeutic target for salt-sensitive hypertension in postmenopausal women.
Collapse
MESH Headings
- Animals
- Cyclopentanes/pharmacology
- Endothelin-1/genetics
- Endothelin-1/metabolism
- Estradiol/metabolism
- Estrogens/pharmacology
- Female
- Kidney Medulla/drug effects
- Kidney Medulla/metabolism
- Male
- Mice, Knockout
- Natriuresis/drug effects
- Ovariectomy
- Quinolines/pharmacology
- Rats, Sprague-Dawley
- Receptor, Endothelin A/genetics
- Receptor, Endothelin A/metabolism
- Receptor, Endothelin B/genetics
- Receptor, Endothelin B/metabolism
- Receptors, Estrogen/deficiency
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Sex Factors
- Signal Transduction
Collapse
Affiliation(s)
- Eman Y. Gohar
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| | | | - Jeffrey O. Aceves
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| | - Randee Sedaka
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| | - Ijeoma E. Obi
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| | - J. Miller Allan
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| | - Reham H. Soliman
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| | - Chunhua Jin
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| | - Carmen De Miguel
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| | - Sarah H. Lindsey
- Department of PharmacologySchool of MedicineTulane UniversityNew OrleansLA
| | - Jennifer S. Pollock
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| | - David M. Pollock
- Division of NephrologyDepartment of MedicineUniversity of Alabama at BirminghamAL
| |
Collapse
|
29
|
Abstract
Maternal cardiovascular changes during pregnancy include an expansion of plasma volume, increased cardiac output, decreased peripheral resistance, and increased uteroplacental blood flow. These adaptations facilitate the progressive increase in uteroplacental perfusion that is required for normal fetal growth and development, prevent the development of hypertension, and provide a reserve of blood in anticipation of the significant blood loss associated with parturition. Each woman's genotype and phenotype determine her ability to adapt in response to molecular signals that emanate from the fetoplacental unit. Here, we provide an overview of the major hemodynamic and cardiac changes and then consider regional changes in the splanchnic, renal, cerebral, and uterine circulations in terms of endothelial and vascular smooth muscle cell plasticity. Although consideration of gestational disease is beyond the scope of this review, aberrant signaling and/or maternal responsiveness contribute to the etiology of several common gestational diseases such as preeclampsia, intrauterine growth restriction, and gestational diabetes.
Collapse
Affiliation(s)
- George Osol
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA;
| | - Nga Ling Ko
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA;
| | - Maurizio Mandalà
- Department of Biology, Ecology and Earth Science, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| |
Collapse
|
30
|
Chen Z, Wang L, Ke J, Xiao D. Effects of Estrogen in Gender-dependent Fetal Programming of Adult Cardiovascular Dysfunction. Curr Vasc Pharmacol 2020; 17:147-152. [PMID: 29493455 DOI: 10.2174/1570161116666180301142453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/15/2018] [Accepted: 02/21/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Epidemiological studies and experimental studies have demonstrated that intrauterine adverse environment increases the risk of Cardiovascular Disease (CVD) in adulthood. However, whether an individual develops a cardiovascular dysfunctional phenotype may depend on genetic background, age, and sex. METHODS In this review, we summarize some of the recent experimental animal studies in the developmental programming of adult CVD with an emphasis on sex differences and the potential role of estrogen in mediating sexual dimorphism. RESULTS Few epidemiological studies report the effect of sex on the developmental programming of CVD. However, numerous experimental animal studies have shown a sex difference in fetal programming of adult cardiovascular dysfunction. Most of the animal studies indicate that male offspring develop cardiovascular dysfunction and CVD in adulthood, whereas adult females appear to be protected. Estrogen is one of the key factors that contributes to the sex difference of adult CVD. Estrogen/its Receptor (ER) may interact with the RAS system by changes of DNA methylation patterns at the target gene promoter, serve as an antioxidant to counteract the prenatal insults-induced heightened ROS, and function as an eNOS activator to increase vasodilation, resulting in the protection of female offspring from the development of hypertension and other CVDs. CONCLUSION These studies suggest that estrogen/ER may contribute to sex differences in cardiovascular response to an adverse intrauterine environment and play a significant role in modulating the cardiovascular response in adulthood.
Collapse
Affiliation(s)
- Zewen Chen
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, United States.,Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong, China
| | - Lei Wang
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, United States
| | - Jun Ke
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, United States.,Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong, China
| | - Daliao Xiao
- Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, United States
| |
Collapse
|
31
|
Mandalà M. Influence of Estrogens on Uterine Vascular Adaptation in Normal and Preeclamptic Pregnancies. Int J Mol Sci 2020; 21:ijms21072592. [PMID: 32276444 PMCID: PMC7177259 DOI: 10.3390/ijms21072592] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022] Open
Abstract
During pregnancy, the maternal cardiovascular system undergoes significant changes, including increased heart rate, cardiac output, plasma volume, and uteroplacental blood flow (UPBF) that are required for a successful pregnancy outcome. The increased UPBF is secondary to profound circumferential growth that extends from the downstream small spiral arteries to the upstream conduit main uterine artery. Although some of the mechanisms underlying uterine vascular remodeling are, in part, known, the factors that drive the remodeling are less clear. That higher circulating levels of estrogens are positively correlated with gestational uterine vascular remodeling suggests their involvement in this process. Estrogens binding to the estrogen receptors expressed in cytotrophoblast cells and in the uterine artery wall stimulate an outward hypertrophic remodeling of uterine vasculature. In preeclampsia, generally lower concentrations of estrogens limit the proper uterine remodeling, thereby reducing UPBF increases and restricting the growth of the fetus. This review aims to report estrogenic regulation of the maternal uterine circulatory adaptation in physiological and pathological pregnancy that favors vasodilation, and to consider the underlying molecular mechanisms by which estrogens regulate uteroplacental hemodynamics.
Collapse
Affiliation(s)
- Maurizio Mandalà
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
32
|
Lechuga TJ, Qi QR, Kim T, Magness RR, Chen DB. E2β stimulates ovine uterine artery endothelial cell H2S production in vitro by estrogen receptor-dependent upregulation of cystathionine β-synthase and cystathionine γ-lyase expression†. Biol Reprod 2020; 100:514-522. [PMID: 30277497 DOI: 10.1093/biolre/ioy207] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 08/29/2018] [Accepted: 09/27/2018] [Indexed: 12/31/2022] Open
Abstract
Endogenous hydrogen sulfide (H2S) is a potent vasodilator and proangiogenic second messenger synthesized from L-cysteine by cystathionine β-synthase (CBS) and cystathionine γ-lyase (CTH). Estrogens are potent vasodilators that stimulate H2S biosynthesis in uterine arteries (UA) in vivo; however, the underlying mechanisms are unknown. We hypothesized that estrogens stimulate H2S biosynthesis in UA endothelial cells (UAEC) via specific estrogen receptor (ER)-dependent mechanisms. In cultured primary UAEC, treatment with estradiol-17β (E2β) stimulated CBS and CTH mRNAs and proteins in a time- and concentration-dependent fashion. As little as 0.1 nM E2β was effective in increasing CBS and CTH expressions and these stimulatory effects maximized with 10-100 nM E2β at 48-72 h. E2β also activated CBS and CTH promoters in UAEC, leading to CBS and CTH expression. Treatment with E2β stimulated H2S production, which was blocked by specific inhibitors of either CBS or CTH and their combination and the ER antagonist ICI 182780. Treatment with either specific agonist of ERα or ERβ stimulated both CBS and CTH mRNA and protein expressions and H2S production to levels similar to that of E2β. Specific antagonist of either ERα or ERβ blocked E2β-stimulated CBS and CTH mRNA and protein expressions and H2S production. Combinations of either ERα or ERβ agonists or their antagonists had no additive effects. Thus, E2β stimulates H2S production by upregulating CBS and CTH mRNA and protein expressions through specific ERα or ERβ-dependent CBS and CTH transcription in UAEC in vitro.
Collapse
Affiliation(s)
- Thomas J Lechuga
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, California, USA.,Department of Pathology, University of California Irvine, Irvine, California, USA
| | - Qian-Rong Qi
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, California, USA
| | - Theresa Kim
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, California, USA
| | - Ronald R Magness
- Department of Obstetrics and Gynecology, Perinatal Research Vascular Center, University of South Florida, Tampa, Florida, USA
| | - Dong-Bao Chen
- Department of Obstetrics and Gynecology, University of California Irvine, Irvine, California, USA.,Department of Pathology, University of California Irvine, Irvine, California, USA
| |
Collapse
|
33
|
Hu XQ, Zhang L. MicroRNAs in Uteroplacental Vascular Dysfunction. Cells 2019; 8:E1344. [PMID: 31671866 PMCID: PMC6912833 DOI: 10.3390/cells8111344] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/26/2019] [Accepted: 10/27/2019] [Indexed: 02/06/2023] Open
Abstract
Pregnancy complications of preeclampsia and intrauterine growth restriction (IUGR) are major causes of maternal and perinatal/neonatal morbidity and mortality. Although their etiologies remain elusive, it is generally accepted that they are secondary to placental insufficiency conferred by both failure in spiral artery remodeling and uteroplacental vascular malfunction. MicroRNAs (miRNAs) are small no-coding RNA molecules that regulate gene expression at the post-transcriptional level. Increasing evidence suggests that miRNAs participate in virtually all biological processes and are involved in numerous human diseases. Differentially expressed miRNAs in the placenta are typical features of both preeclampsia and IUGR. Dysregulated miRNAs target genes of various signaling pathways in uteroplacental tissues, contributing to the development of both complications. In this review, we provide an overview of how aberrant miRNA expression in preeclampsia and IUGR impacts the expression of genes involved in trophoblast invasion and uteroplacental vascular adaptation.
Collapse
Affiliation(s)
- Xiang-Qun Hu
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, USA.
| | - Lubo Zhang
- Lawrence D. Longo MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, USA.
| |
Collapse
|
34
|
Ozer A, Tolun F, Aslan F, Hatırnaz S, Alkan F. The role of G protein-associated estrogen receptor (GPER) 1, corin, raftlin, and estrogen in etiopathogenesis of intrauterine growth retardation. J Matern Fetal Neonatal Med 2019; 34:755-760. [PMID: 31088311 DOI: 10.1080/14767058.2019.1615433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Objective: The aim of the present study was to detect the role of G protein-associated estrogen receptor (GPER) 1, corin, raftlin and estrogen in etiopathogenesis of intrauterine growth retardation (IUGR).Materials and methods: The present study was designed prospectively between January 2017 and May 2018. The study group included 32 patients with unexplained IUGR and 32 healthy pregnant women who gave birth at term among the patients who referred to obstetrics clinic of a tertiary reference hospital. Intrauterine growth retardation (IUGR) was accepted as birth weight below 10th percentile according to the estimated fetal weight. Exclusion criteria were as follows: the patients with renal or hepatic dysfunction, presence of any chronic disease, smoker patients, preeclampsia, acute or chronic inflammatory diseases, body mass index as <18 kg/m2 and >25 kg/m2, structural or chromosomal abnormality in fetus Estradiol (E2), estriol (E3), GPER, corin, and raftlin levels were analyzed in maternal serum and placental tissue homogenate through ELISA method.Results: Serum levels of GPER-1, raftlin, and E3 were significantly lower in IUGR group when compared with the control group (p < .05 for all). Serum corin and E2 levels were similar between two groups. GPER-1, E2, E3, raftlin, and corin levels in placental homogenate were found significantly higher in the control group (p < .05 for all).Conclusion: Although maternal, fetal, and placental causes take place in etiopathogenesis of IUGR, exact etiological factor is not revealed in majority of the IUGR cases. The present study serves as the first study revealing the role of the decrease in GPER-1 and raftlin in maternal serum and placental levels on the etiopathogenesis of IUGR. Furthermore, the decrease in placental corin expression of the cases with IUGR was detected first in the literature. The present study reveals a potential therapeutic use of GPER-1, corin, and raftlin for IUGR.
Collapse
Affiliation(s)
- Alev Ozer
- Kahramanmaras Sutcu Imam University Hospital, Kahramanmaras, Turkey
| | - Fatma Tolun
- Kahramanmaras Sutcu Imam University Hospital, Kahramanmaras, Turkey
| | | | | | - Filiz Alkan
- Kahramanmaras Sutcu Imam University Hospital, Kahramanmaras, Turkey
| |
Collapse
|
35
|
Chang Y, Han Z, Zhang Y, Zhou Y, Feng Z, Chen L, Li X, Li L, Si JQ. 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. [PMID: 30784936 DOI: 10.1016/j.biopha.2019.108666] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/24/2019] [Accepted: 02/04/2019] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE This work aimed to investigate whether G protein-coupled estrogen receptor (GPER) can improve the renal interlobular artery vascular function by increasing the NO content, thereby protecting against renal ischemia-reperfusion (IR) injury. METHODS This study classified ovariectomised (OVX) female Sprague-Dawley rats into OVX, OVX + IR, OVX + IR + G1 (the GPER agonist G1), OVX + IR + G1+G15 (GPER blocker) and OVX + IR + G1+L-NAME (eNOS blocker) groups. Enzyme-linked immunosorbent assay was performed to detect the estrogen levels in the body and eliminate interference from endogenous estrogens. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labelling (TUNEL) and HE staining, renal function test and Paller scoring were performed to identify the successful model and detect the degree of renal and renal interlobular arteries injury. The in vitro microvascular pressure diameter measurement technique was used to detect the contraction and diastolic activities of the renal interlobular arteries in each group. Immunofluorescence technique was used to observe the localisation and expression levels of GPER and eNOS in renal interlobular arteries. The GPER and eNOS protein expression levels in each group were detected by Western blot. The NO content in the serum of each group was detected by the nitrate reductase method. RESULT After OVX, the estrogen level in the body decreased significantly (P < 0.01), and TUNEL staining showed a significant increase in the degree of renal tubular epithelial cell apoptosis in the IR group. Serum creatinine (SCr) and blood urea nitrogen (BUN) levels were significantly increased in the IR group (P < 0.01), and the Paller score showed significantly increased kidney damage. When performing drug treatment, the G1 intervention group significantly decreased serum BUN and SCr levels after IR injury (P < 0.01). The Paller score showed significantly decreased the degree of renal injury (P < 0.01). After IR, the renal interlobular artery contraction rate and systolic velocity of blood vessels were significantly decreased (P < 0.01). The G1 intervention group significantly restored contraction rate and systolic velocity of blood vessels (P < 0.01), and G15 and L-NAME partially reversed this effect (P < 0.01). Immunofluorescence technique showed that GPER was expressed in renal interlobular artery smooth muscle and endothelial cells. After IR injury, the GPER protein expression increased, and the eNOS protein expression decreased significantly (P < 0.01). Western blot showed that after IR injury, the GPER protein expression increased, and the eNOS protein expression decreased significantly. After G1 intervention, the GPER content did not change, and the eNOS content increased significantly (P < 0.01). After ischemia and reperfusion, the serum NO content decreased significantly, but it increased after G1 intervention. G15 and L-NAME reversed the effects of G1 to varying degrees (both at P < 0.01). CONCLUSION GPER may improve the renal interlobular artery vascular function by increasing the NO content, thereby protecting against renal IR injury.
Collapse
Affiliation(s)
- Yuechen Chang
- Department of Physiology, Shihezi University Medical College, Shihezi, 832002, China
| | - Ziwei Han
- Department of Physiology, Shihezi University Medical College, Shihezi, 832002, China
| | - Yang Zhang
- Department of Physiology, Shihezi University Medical College, Shihezi, 832002, China
| | - Ying Zhou
- Department of Physiology, Shihezi University Medical College, Shihezi, 832002, China
| | - Ziyi Feng
- Department of Physiology, Shihezi University Medical College, Shihezi, 832002, China
| | - Long Chen
- Department of Physiology, Shihezi University Medical College, Shihezi, 832002, China
| | - XueRui Li
- Department of Physiology, Shihezi University Medical College, Shihezi, 832002, China
| | - Li Li
- Department of Physiology, Shihezi University Medical College, Shihezi, 832002, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi, 832002, China; Department of Physiology, Jiaxing University Medical College, Jiaxing, 314001, China.
| | - Jun-Qiang Si
- Department of Physiology, Shihezi University Medical College, Shihezi, 832002, China; The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University Medical College, Shihezi, 832002, China; Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430070, China; Department of Physiology, Huazhong University of Science and Technology of Basic Medical Sciences, Wuhan, 430070, China.
| |
Collapse
|
36
|
Effect of Oxidative Stress on the Estrogen-NOS-NO-K Ca Channel Pathway in Uteroplacental Dysfunction: Its Implication in Pregnancy Complications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9194269. [PMID: 30881600 PMCID: PMC6387699 DOI: 10.1155/2019/9194269] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/19/2018] [Accepted: 01/14/2019] [Indexed: 12/27/2022]
Abstract
During pregnancy, the adaptive changes in uterine circulation and the formation of the placenta are essential for the growth of the fetus and the well-being of the mother. The steroid hormone estrogen plays a pivotal role in this adaptive process. An insufficient blood supply to the placenta due to uteroplacental dysfunction has been associated with pregnancy complications including preeclampsia and intrauterine fetal growth restriction (IUGR). Oxidative stress is caused by an imbalance between free radical formation and antioxidant defense. Pregnancy itself presents a mild oxidative stress, which is exaggerated in pregnancy complications. Increasing evidence indicates that oxidative stress plays an important role in the maladaptation of uteroplacental circulation partly by impairing estrogen signaling pathways. This review is aimed at providing both an overview of our current understanding of regulation of the estrogen-NOS-NO-KCa pathway by reactive oxygen species (ROS) in uteroplacental tissues and a link between oxidative stress and uteroplacental dysfunction in pregnancy complications. A better understanding of the mechanisms will facilitate the development of novel and effective therapeutic interventions.
Collapse
|
37
|
Han ZW, Chang YC, Zhou Y, Zhang H, Chen L, Zhang Y, Si JQ, Li L. GPER agonist G1 suppresses neuronal apoptosis mediated by endoplasmic reticulum stress after cerebral ischemia/reperfusion injury. Neural Regen Res 2019; 14:1221-1229. [PMID: 30804253 PMCID: PMC6425826 DOI: 10.4103/1673-5374.251571] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Studies have confirmed a strong association between activation of the endoplasmic reticulum stress pathway and cerebral ischemia/reperfusion (I/R) injury. In this study, three key proteins in the endoplasmic reticulum stress pathway (glucose-regulated protein 78, caspase-12, and C/EBP homologous protein) were selected to examine the potential mechanism of endoplasmic reticulum stress in the neuroprotective effect of G protein-coupled estrogen receptor. Female Sprague-Dawley rats received ovariectomy (OVX), and then cerebral I/R rat models (OVX + I/R) were established by middle cerebral artery occlusion. Immediately after I/R, rat models were injected with 100 μg/kg E2 (OVX + I/R + E2), or 100 μg/kg G protein-coupled estrogen receptor agonist G1 (OVX + I/R + G1) in the lateral ventricle. Longa scoring was used to detect neurobehavioral changes in each group. Infarct volumes were measured by 2,3,5-triphenyltetrazolium chloride staining. Morphological changes in neurons were observed by Nissl staining. Terminal dexynucleotidyl transferase-mediated nick end-labeling staining revealed that compared with the OVX + I/R group, neurological function was remarkably improved, infarct volume was reduced, number of normal Nissl bodies was dramatically increased, and number of apoptotic neurons in the hippocampus was decreased after E2 and G1 intervention. To detect the expression and distribution of endoplasmic reticulum stress-related proteins in the endoplasmic reticulum, caspase-12 distribution and expression were detected by immunofluorescence, and mRNA and protein levels of glucose-regulated protein 78, caspase-12, and C/EBP homologous protein were determined by polymerase chain reaction and western blot assay. The results showed that compared with the OVX + I/R group, E2 and G1 treatment obviously decreased mRNA and protein expression levels of glucose-regulated protein 78, C/EBP homologous protein, and caspase-12. However, the G protein-coupled estrogen receptor antagonist G15 (OVX + I/R + E2 + G15) could eliminate the effect of E2 on cerebral I/R injury. These results confirm that E2 and G protein-coupled estrogen receptor can inhibit the expression of endoplasmic reticulum stress-related proteins and neuronal apoptosis in the hippocampus, thereby improving dysfunction caused by cerebral I/R injury. Every experimental protocol was approved by the Institutional Ethics Review Board at the First Affiliated Hospital of Shihezi University School of Medicine, China (approval No. SHZ A2017-171) on February 27, 2017.
Collapse
Affiliation(s)
- Zi-Wei Han
- Department of Physiology, Medical College of Shihezi University; Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Yue-Chen Chang
- Department of Physiology, Medical College of Shihezi University; Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Ying Zhou
- Department of Physiology, Medical College of Shihezi University; Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Hang Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region; Affiliated Teng Zhou Central People's Hospital, Jining Medical University, Jining, Shandong Province, China
| | - Long Chen
- Department of Physiology, Medical College of Shihezi University; Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Yang Zhang
- Department of Physiology, Medical College of Shihezi University; Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Jun-Qiang Si
- Department of Physiology, Medical College of Shihezi University; Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Li Li
- Department of Physiology, Medical College of Shihezi University; Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University School of Medicine, Shihezi, Xinjiang Uygur Autonomous Region; Department of Physiology, Jiaxing College of Medicine, Jiaxing, Zhejiang Province, China
| |
Collapse
|
38
|
Pabbidi MR, Kuppusamy M, Didion SP, Sanapureddy P, Reed JT, Sontakke SP. Sex differences in the vascular function and related mechanisms: role of 17β-estradiol. Am J Physiol Heart Circ Physiol 2018; 315:H1499-H1518. [PMID: 30192631 DOI: 10.1152/ajpheart.00194.2018] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The incidence of cardiovascular disease (CVD) is lower in premenopausal women but increases with age and menopause compared with similarly aged men. Based on the prevalence of CVD in postmenopausal women, sex hormone-dependent mechanisms have been postulated to be the primary factors responsible for the protection from CVD in premenopausal women. Recent Women’s Health Initiative studies, Cochrane Review studies, the Early Versus Late Intervention Trial with Estradiol Study, and the Kronos Early Estrogen Prevention Study have suggested that beneficial effects of hormone replacement therapy (HRT) are seen in women of <60 yr of age and if initiated within <10 yr of menopause. In contrast, the beneficial effects of HRT are not seen in women of >60 yr of age and if commenced after 10 yr of menopause. The higher incidence of CVD and the failure of HRT in postmenopausal aged women could be partly associated with fundamental differences in the vascular structure and function between men and women and in between pre- and postmenopausal women, respectively. In this regard, previous studies from human and animal studies have identified several sex differences in vascular function and associated mechanisms. The female sex hormone 17β-estradiol regulates the majority of these mechanisms. In this review, we summarize the sex differences in vascular structure, myogenic properties, endothelium-dependent and -independent mechanisms, and the role of 17β-estradiol in the regulation of vascular function.
Collapse
Affiliation(s)
- Mallikarjuna R. Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Maniselvan Kuppusamy
- Division of Endocrinology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sean P. Didion
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Padmaja Sanapureddy
- Department of Primary Care and Medicine, G. V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi
| | - Joey T. Reed
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sumit P. Sontakke
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
39
|
Al-Shboul OA, Nazzal MS, Mustafa AG, Al-Dwairi AN, Alqudah MA, Abu Omar A, Alfaqih MA, Alsalem MI. Estrogen relaxes gastric muscle cells via a nitric oxide- and cyclic guanosine monophosphate-dependent mechanism: A sex-associated differential effect. Exp Ther Med 2018; 16:1685-1692. [PMID: 30186388 PMCID: PMC6122185 DOI: 10.3892/etm.2018.6406] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/01/2018] [Indexed: 12/22/2022] Open
Abstract
Various gastrointestinal (GI) disorders have a higher prevalence in women than in men. In addition, estrogen has been demonstrated to have an inhibitory effect on the contractility of GI smooth muscle. Although increased plasma estrogen levels have been implicated in GI disorders, the role of gastric estrogen receptor (ER) in these sex-specific differences remains to be fully elucidated. The present study was designed to investigate the sex-associated differences in the expression of the two ER isoforms, ERα and ERβ, and the effect of estrogen on gastric muscle contraction via the nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) pathway. Experiments were performed on single gastric smooth muscle cells (GSMCs) isolated from male and female Sprague Dawley rats. The effect of acetylcholine (ACh), a muscarinic agonist, on the contraction of GSMCs was measured via scanning micrometry in the presence or absence of 1 µM 17β-estradiol (E2), an agonist to the majority of ERs, 1,3,5-tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole (PPT), an ERα agonist, or diarylpropionitrile (DPN), an ERβ agonist. The protein expression levels of ER subtypes in GSMCs were measured using a specifically designed ELISA. GSMCs from female rats had a higher expression of ERα and ERβ protein compared with GSMCs from males. ACh induced less contraction in female that in male GSMCs. Pre-treatment of GSMCs with E2 reduced the contraction of GSMCs from both sexes, but to a greater extent in those from females. PPT and DPN inhibited ACh-induced contraction in GSMCs from females. Furthermore, E2 increased NO and cGMP levels in GSMCs from males and females; however, higher levels were measured in females. Of note, pre-incubation of female GSMCs with Nω-nitro-L-arginine, a NO synthase inhibitor, or 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, a guanylyl cyclase inhibitor, reduced the inhibitory effect of estrogen on GSMC contraction. In conclusion, estrogen relaxes GSMCs via an NO/cGMP-dependent mechanism, and the reduced contraction in GSMCs from females by estrogen may be associated with the sex-associated increased expression of ERα and ERβ, and greater production of NO and cGMP, compared with that in GSMCs from males.
Collapse
Affiliation(s)
- Othman A Al-Shboul
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mona S Nazzal
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Ayman G Mustafa
- Department of Anatomy, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Ahmed N Al-Dwairi
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammad A Alqudah
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Amal Abu Omar
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mahmoud A Alfaqih
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammad I Alsalem
- Department of Anatomy and Histology, Faculty of Medicine, Jordan University, Amman 11942, Jordan
| |
Collapse
|
40
|
Evanson KW, Goldsmith JA, Ghosh P, Delp MD. 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. [PMID: 29938113 PMCID: PMC6011940 DOI: 10.1002/prp2.409] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/22/2018] [Indexed: 11/07/2022] Open
Abstract
The G protein-coupled estrogen receptor (GPER) is a significant modulator of arterial contractility and blood flow. The GPER-specific activator, G-1, has been widely used to characterize GPER function in a variety of tissue types. Large conductance, calcium (Ca2+)-activated K+ (BK) channels are sensitive to 17β-estradiol (17β-E2) and estrogenic compounds (e.g., tamoxifen, ICI 182 780) that target estrogen receptors. The purpose of this study was to investigate the effects of G-1 on BK channel activation and function in cerebral arterial myocytes. Inside-out and perforated patch clamp were utilized to assess the effects of G-1 (50 nmol·L-1-5 μmol·L-1) on BK channel activation and currents in cerebral arterial myocytes. Pressurized artery myography was used to investigate the effects of G-1 on vasodilatory response and BK channel function of cerebral resistance size arteries. G-1 reduced BK channel activation in cerebral arterial myocytes through elevations in BK channel mean close times. Depressed BK channel activation following G-1 application resulted in attenuated physiological BK currents (transient BK currents). G-1 elicited vasodilation, but reduced BK channel function, in pressurized, endothelium-denuded cerebral arteries. These data suggest that G-1 directly suppresses BK channel activation and currents in cerebral arterial myocytes, BK channels being critically important in the regulation of myocyte membrane potential and arterial contractility. Thus, GPER-mediated vasodilation using G-1 to activate the receptor may underestimate the physiological function and relevance of GPER in the cardiovascular system.
Collapse
Affiliation(s)
- Kirk W. Evanson
- Department of Nutrition, Food, and Exercise SciencesFlorida State UniversityTallahasseeFlorida
| | - Jacob A. Goldsmith
- Department of Nutrition, Food, and Exercise SciencesFlorida State UniversityTallahasseeFlorida
| | - Payal Ghosh
- Department of Nutrition, Food, and Exercise SciencesFlorida State UniversityTallahasseeFlorida
| | - Michael D. Delp
- Department of Nutrition, Food, and Exercise SciencesFlorida State UniversityTallahasseeFlorida
| |
Collapse
|
41
|
Ko NL, Mandalà M, John L, Gelinne A, Osol G. Venoarterial communication mediates arterial wall shear stress-induced maternal uterine vascular remodeling during pregnancy. Am J Physiol Heart Circ Physiol 2018; 315:H709-H717. [PMID: 29775414 DOI: 10.1152/ajpheart.00126.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although expansive remodeling of the maternal uterine circulation during pregnancy is essential for maintaining uteroplacental perfusion and normal fetal growth, the underlying physiological mechanisms are not well understood. Using a rat model, surgical approaches were used to alter uterine hemodynamics and wall shear stress (WSS) to evaluate the effects of WSS and venoarterial communication (e.g., transfer of placentally derived growth signals from postplacental veins to preplacental arteries) on gestational uterine vascular remodeling. Changes in WSS secondary to ligation of the cervical but not the ovarian end of the main uterine artery and vein provoked significant expansive remodeling at the opposite end of both vessels, but only in pregnant animals. The ≈50% increase in lumen diameter (relative to the contralateral horn) was associated with an upregulation of total endothelial nitric oxide (NO) synthase expression and was abolished by in vivo NO synthase inhibition with N-nitro-l-arginine methyl ester. Complete removal of a venous segment adjacent to the uterine artery to eliminate local venous influences significantly attenuated the WSS-induced remodeling by about one-half ( P < 0.05). These findings indicate that, during pregnancy, 1) increased WSS stimulates uterine artery growth via NO signaling and 2) the presence of an adjacent vein is required for arterial remodeling to fully occur. NEW & NOTEWORTHY This study provides the first in vivo evidence for the importance of venous influences on arterial growth within the uteroplacental circulation.
Collapse
Affiliation(s)
- Nga Ling Ko
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Larner College of Medicine, University of Vermont , Burlington, Vermont
| | - Maurizio Mandalà
- Department of Biology, Ecology, and Earth Science, University of Calabria , Cosenza , Italy
| | - Liam John
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Larner College of Medicine, University of Vermont , Burlington, Vermont
| | - Aaron Gelinne
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Larner College of Medicine, University of Vermont , Burlington, Vermont
| | - George Osol
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Larner College of Medicine, University of Vermont , Burlington, Vermont
| |
Collapse
|
42
|
De Francesco EM, Sotgia F, Clarke RB, Lisanti MP, Maggiolini M. G Protein-Coupled Receptors at the Crossroad between Physiologic and Pathologic Angiogenesis: Old Paradigms and Emerging Concepts. Int J Mol Sci 2017; 18:ijms18122713. [PMID: 29240722 PMCID: PMC5751314 DOI: 10.3390/ijms18122713] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) have been implicated in transmitting signals across the extra- and intra-cellular compartments, thus allowing environmental stimuli to elicit critical biological responses. As GPCRs can be activated by an extensive range of factors including hormones, neurotransmitters, phospholipids and other stimuli, their involvement in a plethora of physiological functions is not surprising. Aberrant GPCR signaling has been regarded as a major contributor to diverse pathologic conditions, such as inflammatory, cardiovascular and neoplastic diseases. In this regard, solid tumors have been demonstrated to activate an angiogenic program that relies on GPCR action to support cancer growth and metastatic dissemination. Therefore, the manipulation of aberrant GPCR signaling could represent a promising target in anticancer therapy. Here, we highlight the GPCR-mediated angiogenic function focusing on the molecular mechanisms and transduction effectors driving the patho-physiological vasculogenesis. Specifically, we describe evidence for the role of heptahelic receptors and associated G proteins in promoting angiogenic responses in pathologic conditions, especially tumor angiogenesis and progression. Likewise, we discuss opportunities to manipulate aberrant GPCR-mediated angiogenic signaling for therapeutic benefit using innovative GPCR-targeted and patient-tailored pharmacological strategies.
Collapse
Affiliation(s)
- Ernestina M De Francesco
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria via Savinio, 87036 Rende, Italy.
- Breast Cancer Now Research Unit, Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester M20 4GJ, UK.
| | - Federica Sotgia
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre, University of Salford, Greater Manchester M5 4WT, UK.
| | - Robert B Clarke
- Breast Cancer Now Research Unit, Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester M20 4GJ, UK.
| | - Michael P Lisanti
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre, University of Salford, Greater Manchester M5 4WT, UK.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria via Savinio, 87036 Rende, Italy.
| |
Collapse
|
43
|
Impairment of BKca channels in human placental chorionic plate arteries is potentially relevant to the development of preeclampsia. Hypertens Res 2017; 41:126-134. [DOI: 10.1038/hr.2017.99] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/05/2017] [Accepted: 07/18/2017] [Indexed: 01/02/2023]
|
44
|
Osol G, Ko NL, Mandalà M. Altered Endothelial Nitric Oxide Signaling as a Paradigm for Maternal Vascular Maladaptation in Preeclampsia. Curr Hypertens Rep 2017; 19:82. [PMID: 28942512 DOI: 10.1007/s11906-017-0774-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The goal of this review is to present the newest insights into what we view as a central failure of cardiovascular adaptation in preeclampsia (PE) by focusing on one clinically significant manifestation of maternal endothelial dysfunction: nitric oxide signaling. The etiology, symptoms, and current theories of the PE syndrome are described first, followed by a review of the available evidence, and underlying causes of reduced endothelial nitric oxide (NO) signaling in PE. RECENT FINDINGS PE maladaptations include, but are not limited to, altered physiological stimulatory inputs (e.g., estrogen; VEGF/PlGF; shear stress) and substrates (L-Arg; ADMA), augmented placental secretion of anti-angiogenic and inflammatory factors such as sFlt-1 and Eng, changes in eNOS (polymorphisms, expression), and reduced bioavailability of NO secondary to oxidative stress. PE is a complex obstetrical syndrome that is associated with maternal vascular dysfunction. Diminished peripheral endothelial vasodilator influence in general, and of NO signaling specifically, are key in driving disease progression and severity.
Collapse
Affiliation(s)
- George Osol
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Given H.S.C. C-217A 89 Beaumont Ave, Burlington, VT, 5405, USA.
| | - Nga Ling Ko
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Given H.S.C. C-217A 89 Beaumont Ave, Burlington, VT, 5405, USA
| | - Maurizio Mandalà
- Department of Biology, Ecology and Earth Science, University of Calabria, Cosenza, Italy
| |
Collapse
|
45
|
Peixoto P, Aires RD, Lemos VS, Bissoli NS, Santos RLD. GPER agonist dilates mesenteric arteries via PI3K-Akt-eNOS and potassium channels in both sexes. Life Sci 2017. [DOI: 10.1016/j.lfs.2017.06.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
46
|
|
47
|
De Francesco EM, Rocca C, Scavello F, Amelio D, Pasqua T, Rigiracciolo DC, Scarpelli A, Avino S, Cirillo F, Amodio N, Cerra MC, Maggiolini M, Angelone T. Protective Role of GPER Agonist G-1 on Cardiotoxicity Induced by Doxorubicin. J Cell Physiol 2017; 232:1640-1649. [DOI: 10.1002/jcp.25585] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/06/2016] [Indexed: 01/26/2023]
Affiliation(s)
| | - Carmine Rocca
- Department of Biology, Ecology, and E.S.; University of Calabria; Rende (CS) Italy
| | - Francesco Scavello
- Department of Biology, Ecology, and E.S.; University of Calabria; Rende (CS) Italy
| | - Daniela Amelio
- Department of Biology, Ecology, and E.S.; University of Calabria; Rende (CS) Italy
| | - Teresa Pasqua
- Department of Biology, Ecology, and E.S.; University of Calabria; Rende (CS) Italy
| | - Damiano C. Rigiracciolo
- Department of Pharmacy; Health and Nutritional Sciences; University of Calabria; Rende (CS) Italy
| | - Andrea Scarpelli
- Department of Pharmacy; Health and Nutritional Sciences; University of Calabria; Rende (CS) Italy
| | - Silvia Avino
- Department of Pharmacy; Health and Nutritional Sciences; University of Calabria; Rende (CS) Italy
| | - Francesca Cirillo
- Department of Pharmacy; Health and Nutritional Sciences; University of Calabria; Rende (CS) Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine; University of Catanzaro Magna Graecia; Catanzaro Italy
| | - Maria C. Cerra
- Department of Biology, Ecology, and E.S.; University of Calabria; Rende (CS) Italy
- National Institute of Cardiovascular Research; Bologna Italy
| | - Marcello Maggiolini
- Department of Pharmacy; Health and Nutritional Sciences; University of Calabria; Rende (CS) Italy
| | - Tommaso Angelone
- Department of Biology, Ecology, and E.S.; University of Calabria; Rende (CS) Italy
- National Institute of Cardiovascular Research; Bologna Italy
| |
Collapse
|
48
|
Bonney EA, Howard A, Krebs K, Begin K, Veilleux K, Gokina NI. Impact of Immune Deficiency on Remodeling of Maternal Resistance Vasculature 4 Weeks Postpartum in Mice. Reprod Sci 2017; 24:514-525. [PMID: 27899739 DOI: 10.1177/1933719116678691] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pregnancy manifests changes in the vascular and immune systems that persist postpartum (PP), have important implications for future pregnancies, and may modify responses to cardiovascular stress in late life. The association between immune and vascular function and the generation or progression of cardiovascular disease beg the question of whether altered immunity modifies pregnancy-induced changes in the vasculature. Our objective was to compare changes in the function and remodeling of systemic resistance vessels 4 weeks PP in normal C57BL/6 (B6), and immunodeficient mice recombinase 1-deficient/B6 ( Rag1-/-). Immune deficiency did not change the responsiveness to acetylcholine (ACh) and phenylephrine at baseline but decreased arterial distensibility and increased stiffness PP. Adoptive transfer of CD8 T cells into Rag1-/- mice decreased the response to ACh while increasing distensibility and wall thickness. When compared to PP Rag1-/-, vessels from PP CD4-deficient mice, which have B cells and CD8 T cells, but no CD4 cells, show increased distensibility and decreased responsiveness to ACh in a pattern similar to that seen in Rag1-/- given CD8 T cells prior to mating. These studies suggest a key role for T cell, particularly CD8 T cell, associated factors in the PP remodeling of maternal resistance vessels.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- 1 Division of Reproductive Science Research, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont, Burlington, VT, USA
| | - Ann Howard
- 1 Division of Reproductive Science Research, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont, Burlington, VT, USA
| | - Kendall Krebs
- 1 Division of Reproductive Science Research, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont, Burlington, VT, USA
| | - Kelly Begin
- 1 Division of Reproductive Science Research, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont, Burlington, VT, USA
| | - Kelsey Veilleux
- 1 Division of Reproductive Science Research, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont, Burlington, VT, USA
| | - Natalia I Gokina
- 1 Division of Reproductive Science Research, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont, Burlington, VT, USA
| |
Collapse
|
49
|
Boeldt DS, Bird IM. Vascular adaptation in pregnancy and endothelial dysfunction in preeclampsia. J Endocrinol 2017; 232:R27-R44. [PMID: 27729465 PMCID: PMC5115955 DOI: 10.1530/joe-16-0340] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/11/2016] [Indexed: 12/27/2022]
Abstract
Maternal vascular adaptation to pregnancy is critically important to expand the capacity for blood flow through the uteroplacental unit to meet the needs of the developing fetus. Failure of the maternal vasculature to properly adapt can result in hypertensive disorders of pregnancy such as preeclampsia (PE). Herein, we review the endocrinology of maternal adaptation to pregnancy and contrast this with that of PE. Our focus is specifically on those hormones that directly influence endothelial cell function and dysfunction, as endothelial cell dysfunction is a hallmark of PE. A variety of growth factors and cytokines are present in normal vascular adaptation to pregnancy. However, they have also been shown to be circulating at abnormal levels in PE pregnancies. Many of these factors promote endothelial dysfunction when present at abnormal levels by acutely inhibiting key Ca2+ signaling events and chronically promoting the breakdown of endothelial cell-cell contacts. Increasingly, our understanding of how the contributions of the placenta, immune cells, and the endothelium itself promote the endocrine milieu of PE is becoming clearer. We then describe in detail how the complex endocrine environment of PE affects endothelial cell function, why this has contributed to the difficulty in fully understanding and treating this disorder, and how a focus on signaling convergence points of many hormones may be a more successful treatment strategy.
Collapse
Affiliation(s)
- D S Boeldt
- Department of Ob/GynPerinatal Research Laboratories, University Wisconsin - Madison, Madison, Wisconsin, USA
| | - I M Bird
- Department of Ob/GynPerinatal Research Laboratories, University Wisconsin - Madison, Madison, Wisconsin, USA
| |
Collapse
|
50
|
Li J, Liu CN, Wei N, Li XD, Liu YY, Yang R, Jia YJ. Protective effects of BAY 73-6691, a selective inhibitor of phosphodiesterase 9, on amyloid-β peptides-induced oxidative stress in in-vivo and in-vitro models of Alzheimer's disease. Brain Res 2016; 1642:327-335. [DOI: 10.1016/j.brainres.2016.04.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/06/2016] [Accepted: 04/05/2016] [Indexed: 12/11/2022]
|