1
|
Kusewitt DF, Sharma G, Woods CD, Rosas E, Hathaway HJ, Prossnitz ER. GPER expression prevents estrogen-induced urinary retention in obese mice. J Steroid Biochem Mol Biol 2024; 244:106607. [PMID: 39197539 PMCID: PMC11444091 DOI: 10.1016/j.jsbmb.2024.106607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Long-term administration of exogenous estrogen is known to cause urinary retention and marked, often fatal, bladder distention in both male and female mice. Estrogen-treated mice have increased bladder pressure and decreased urine flow, suggesting that urinary retention in estrogen-treated mice is due to infravesicular obstruction to urine outflow. Thus, the condition is commonly referred to as bladder outlet obstruction (BOO). Obesity can also lead to urinary retention. As the effects of estrogen are mediated by multiple receptors, including estrogen receptors ERα and ERβ and the G protein-coupled estrogen receptor (GPER), we sought to determine whether GPER plays a role in estrogen-induced BOO, particularly in the context of obesity. Wild type and GPER knockout (KO) mice fed a high-fat diet were ovariectomized or left ovary-intact (sham surgery) and supplemented with slow-release estrogen or vehicle-only pellets. Supplementing both GPER KO and wild type obese mice with estrogen for 8 weeks resulted in weight loss, splenic enlargement, and thymic atrophy, as expected. However, estrogen-treated obese GPER KO mice developed abdominal distension, debilitation, and ulceration of the skin surrounding the urogenital opening. At necropsy, these mice had prominently distended bladders and hydronephrosis. In contrast, estrogen-treated obese wild type mice only rarely displayed these signs. Our results suggest that, under conditions of obesity, estrogen induces BOO as a result of ERα-driven pathways and that GPER expression is protective against BOO.
Collapse
Affiliation(s)
- Donna F Kusewitt
- Department of Pathology, University of New Mexico Health Science Center, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA.
| | - Geetanjali Sharma
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Christine D Woods
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Emmanuel Rosas
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Helen J Hathaway
- Department of Cell Biology & Physiology, University of New Mexico Health Science Center, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Eric R Prossnitz
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA; Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, University of New Mexico Health Science Center, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA.
| |
Collapse
|
2
|
Bartkowiak-Wieczorek J, Jaros A, Gajdzińska A, Wojtyła-Buciora P, Szymański I, Szymaniak J, Janusz W, Walczak I, Jonaszka G, Bienert A. The Dual Faces of Oestrogen: The Impact of Exogenous Oestrogen on the Physiological and Pathophysiological Functions of Tissues and Organs. Int J Mol Sci 2024; 25:8167. [PMID: 39125736 PMCID: PMC11311417 DOI: 10.3390/ijms25158167] [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: 06/09/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Oestrogen plays a crucial physiological role in both women and men. It regulates reproductive functions and maintains various non-reproductive tissues through its receptors, such as oestrogen receptor 1/oestrogen receptor α (ESR1/Erα), oestrogen receptor 2/oestrogen receptor β (ESR2/Erβ), and G protein-coupled oestrogen receptor 1 (GPER). This hormone is essential for the proper functioning of women's ovaries and uterus. Oestrogen supports testicular function and spermatogenesis in men and contributes to bone density, cardiovascular health, and metabolic processes in both sexes. Nuclear receptors Er-α and Er-β belong to the group of transcription activators that stimulate cell proliferation. In the environment, compounds similar in structure to the oestrogens compete with endogenous hormones for binding sites to receptors and to disrupt homeostasis. The lack of balance in oestrogen levels can lead to infertility, cancer, immunological disorders, and other conditions. Exogenous endocrine-active compounds, such as bisphenol A (BPA), phthalates, and organic phosphoric acid esters, can disrupt signalling pathways responsible for cell division and apoptosis processes. The metabolism of oestrogen and its structurally similar compounds can produce carcinogenic substances. It can also stimulate the growth of cancer cells by regulating genes crucial for cell proliferation and cell cycle progression, with long-term elevated levels linked to hormone-dependent cancers such as breast cancer. Oestrogens can also affect markers of immunological activation and contribute to the development of autoimmune diseases. Hormone replacement therapy, oral contraception, in vitro fertilisation stimulation, and hormonal stimulation of transgender people can increase the risk of breast cancer. Cortisol, similar in structure to oestrogen, can serve as a biomarker associated with the risk of developing breast cancer. The aim of this review is to analyse the sources of oestrogens and their effects on the endogenous and exogenous process of homeostasis.
Collapse
Affiliation(s)
- Joanna Bartkowiak-Wieczorek
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Agnieszka Jaros
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.J.); (A.B.)
| | - Anna Gajdzińska
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Paulina Wojtyła-Buciora
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
- Department of Social Medicine and Public Health, Calisia University, 62-800 Kalisz, Poland
| | - Igor Szymański
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Julian Szymaniak
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Wojciech Janusz
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Iga Walczak
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Gabriela Jonaszka
- Physiology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.G.); (P.W.-B.); (I.S.); (J.S.); (W.J.); (I.W.); (G.J.)
| | - Agnieszka Bienert
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (A.J.); (A.B.)
| |
Collapse
|
3
|
Kilanowski-Doroh IM, McNally AB, Wong T, Visniauskas B, Blessinger SA, Sugi AI, Richard C, Diaz Z, Horton A, Natale CA, Ogola BO, Lindsey SH. Ovariectomy-Induced Arterial Stiffening Differs From Vascular Aging and Is Reversed by GPER Activation. Hypertension 2024; 81:e51-e62. [PMID: 38445498 PMCID: PMC11023783 DOI: 10.1161/hypertensionaha.123.22024] [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: 09/07/2023] [Accepted: 02/16/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Arterial stiffness is a cardiovascular risk factor and dramatically increases as women transition through menopause. The current study assessed whether a mouse model of menopause increases arterial stiffness in a similar manner to aging and whether activation of the G-protein-coupled estrogen receptor could reverse stiffness. METHODS Female C57Bl/6J mice were ovariectomized at 10 weeks of age or aged to 52 weeks, and some mice were treated with G-protein-coupled estrogen receptor agonists. RESULTS Ovariectomy and aging increased pulse wave velocity to a similar extent independent of changes in blood pressure. Aging increased carotid wall thickness, while ovariectomy increased material stiffness without altering vascular geometry. RNA-sequencing analysis revealed that ovariectomy downregulated smooth muscle contractile genes. The enantiomerically pure G-protein-coupled estrogen receptor agonist, LNS8801, reversed stiffness in ovariectomy mice to a greater degree than the racemic agonist G-1. In summary, ovariectomy and aging induced arterial stiffening via potentially different mechanisms. Aging was associated with inward remodeling, while ovariectomy-induced material stiffness independent of geometry and a loss of the contractile phenotype. CONCLUSIONS This study enhances our understanding of the impact of estrogen loss on vascular health in a murine model and warrants further studies to examine the ability of LNS8801 to improve vascular health in menopausal women.
Collapse
Affiliation(s)
| | | | - Tristen Wong
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | - Bruna Visniauskas
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | | | | | - Chase Richard
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| | - Zaidmara Diaz
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | - Alec Horton
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | | | - Benard O. Ogola
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| |
Collapse
|
4
|
Liu J, Niu Y, Zhang B, Sun Q, Li H, Bai L, Su Z. Different Expression Pattern of G Protein-Coupled Estrogen Receptor GPER1 in Esophageal Squamous Cell Carcinoma and Adenocarcinoma. Int J Mol Sci 2023; 24:14055. [PMID: 37762356 PMCID: PMC10531045 DOI: 10.3390/ijms241814055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Esophageal carcinoma is a male-dominant malignancy worldwide, and esophageal adenocarcinoma (EAC) shows more significant sex bias than esophageal squamous cell carcinoma (ESCC) in morbidity and mortality. The G protein-coupled estrogen receptor 1 (GPER1) is involved in several sex-related cancers; however, its expression level in esophageal carcinoma has been poorly investigated and its role is not precisely defined, depending on histological types. In the present study, the mRNA levels of GPER1 in esophageal carcinoma were collected from GEPIA and Oncomine databases for meta-analyses. The protein expression levels of GPER1 were detected by immunohistochemistry in the tissue microarray of EAC and ESCC. The GPER1 selective agonist G1, antagonist G15, and siRNA were applied in vitro to investigate their impacts on esophageal cell lines. Analysis of the RNA levels from the databases showed a decreased expression of GPER1 in overall esophageal carcinoma, and low expression levels of GPER1 were found to be associated with low survival of tumor patients. However, in the subgroup of EAC and its precancerous lesion, Barrett's esophagus, overexpression of GPER1 RNA was increased when compared with the normal tissues. The average staining scores of GPER1 protein in the tissue microarray of EAC were significantly higher than normal esophageal samples, and the rate of positive staining increased with the grade of poor tumor differentiation. The scores of GPER1 protein in ESCC tissues were lower than those in the normal tissues. The results from cell line experiments in vitro showed that the GPER1 agonist G1 inhibited proliferation and promoted apoptosis of ESCC cells EC109 with positive expression of GPER1. G1 had no obvious effect on normal esophageal NE2 cells with weak expression of GPER1. In addition, GPER1 RNA knockdown and application of antagonist G15 reversed the effects of G1 on EC109. The results of this study indicate that the expression levels of GPER1 are higher in EAC than in ESCC, which might be correlated with the dimorphic estrogen signaling pathway in different types of esophageal carcinoma.
Collapse
Affiliation(s)
- Jingshi Liu
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China
| | - Yongdong Niu
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, China
| | - Bin Zhang
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China
| | - Qisi Sun
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China
| | - Haiyi Li
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China
| | - Lu Bai
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China
| | - Zhongjing Su
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
5
|
Kilanowski-Doroh IM, McNally AB, Wong T, Visniauskas B, Blessinger SA, Imulinde Sugi A, Richard C, Diaz Z, Horton A, Natale CA, Ogola BO, Lindsey SH. Ovariectomy-Induced Arterial Stiffening Differs from Vascular Aging and is Reversed by GPER Activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552881. [PMID: 37645992 PMCID: PMC10462036 DOI: 10.1101/2023.08.10.552881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Arterial stiffness is a cardiovascular risk factor and dramatically increases as women transition through menopause. The current study assessed whether a mouse model of menopause increases arterial stiffness in a similar manner to aging, and whether activation of the G protein-coupled estrogen receptor (GPER) could reverse stiffness. Female C57Bl/6J mice were ovariectomized (OVX) at 10 weeks of age or aged to 52 weeks, and some mice were treated with GPER agonists. OVX and aging increased pulse wave velocity to a similar extent independent of changes in blood pressure. Aging increased carotid wall thickness, while OVX increased material stiffness without altering vascular geometry. RNA-Seq analysis revealed that OVX downregulated smooth muscle contractile genes. The enantiomerically pure GPER agonist, LNS8801, reversed stiffness in OVX mice to a greater degree than the racemic agonist G-1. In summary, OVX and aging induced arterial stiffening via potentially different mechanisms. Aging was associated with inward remodeling while OVX induced material stiffness independent of geometry and a loss of the contractile phenotype. This study helps to further our understanding of the impact of menopause on vascular health and identifies LNS8801 as a potential therapy to counteract this detrimental process in women.
Collapse
|
6
|
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
|
7
|
Guthrie GL, Almutlaq RN, Sugahara S, Butt MK, Brooks CR, Pollock DM, Gohar EY. G protein-coupled estrogen receptor 1 regulates renal endothelin-1 signaling system in a sex-specific manner. Front Physiol 2023; 14:1086973. [PMID: 36733911 PMCID: PMC9887121 DOI: 10.3389/fphys.2023.1086973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
Demographic studies reveal lower prevalence of hypertension among premenopausal females compared to age-matched males. The kidney plays a central role in the maintenance of sodium (Na+) homeostasis and consequently blood pressure. Renal endothelin-1 (ET-1) is a pro-natriuretic peptide that contributes to sex differences in blood pressure regulation and Na+ homeostasis. We recently showed that activation of renal medullary G protein-coupled estrogen receptor 1 (GPER1) promotes ET-1-dependent natriuresis in female, but not male, rats. We hypothesized that GPER1 upregulates the renal ET-1 signaling system in females, but not males. To test our hypothesis, we determined the effect of GPER1 deletion on ET-1 and its downstream effectors in the renal cortex, outer and inner medulla obtained from 12-16-week-old female and male mice. GPER1 knockout (KO) mice and wildtype (WT) littermates were implanted with telemetry transmitters for blood pressure assessment, and we used metabolic cages to determine urinary Na+ excretion. GPER1 deletion did not significantly affect 24-h mean arterial pressure (MAP) nor urinary Na+ excretion. However, GPER1 deletion decreased urinary ET-1 excretion in females but not males. Of note, female WT mice had greater urinary ET-1 excretion than male WT littermates, whereas no sex differences were observed in GPER1 KO mice. GPER1 deletion increased inner medullary ET-1 peptide content in both sexes but increased outer medullary ET-1 content in females only. Cortical ET-1 content increased in response to GPER1 deletion in both sexes. Furthermore, GPER1 deletion notably increased inner medullary ET receptor A (ETA) and decreased outer medullary ET receptor B (ETB) mRNA expression in male, but not female, mice. We conclude that GPER1 is required for greater ET-1 excretion in females. Our data suggest that GPER1 is an upstream regulator of renal medullary ET-1 production and ET receptor expression in a sex-specific manner. Overall, our study identifies the role of GPER1 as a sex-specific upstream regulator of the renal ET-1 system.
Collapse
Affiliation(s)
- Ginger L. Guthrie
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rawan N. Almutlaq
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sho Sugahara
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Maryam K. Butt
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Craig R. Brooks
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States
| | - David M. Pollock
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eman Y. Gohar
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, United States,*Correspondence: Eman Y. Gohar,
| |
Collapse
|
8
|
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
|
9
|
Yu X, Nguyen P, Burns NC, Heaps CL, Stallone JN, Sohrabji F, Han G. Activation of G protein-coupled estrogen receptor fine-tunes age-related decreased vascular activities in the aortae of female and male rats. Steroids 2022; 183:108997. [PMID: 35314416 DOI: 10.1016/j.steroids.2022.108997] [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] [Received: 10/13/2021] [Revised: 02/07/2022] [Accepted: 02/22/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hormone replacement therapy was found to be effective in cardiovascular protection only in younger women, not in older women. In this study, we tested whether G protein-coupled estrogen receptor 1 (GPER) activation improves vascular activities in response to ET-1 and ACh in aging rats. METHODS Isometric tension study was applied on aortic rings isolated from young adult (5-7 months) and reproductive senescent middle-aged (10-12 months) female Sprague Dawley rats and age matched males. RESULTS The aortic contractile response to ET-1 and the relaxation response to ACh were reduced in the female middle-aged rats compared to the female young adult rats. The presence of G-1, the GPER agonist, normalized the reduced vascular activities. Cyclooxygenase inhibitor, meclofenamate, blocked the increased constriction effect of G-1, but further enhanced relaxation effect of G-1. There was no significant difference in aortic reactivity to either ET-1 or ACh between the male middle-aged and young adult rats. The contractile response to ET-1 was not different within the same age of the two sex groups, but there was a remarkable difference in relaxation response to ACh between young adult females and males with better response in females. GPER activation greatly improved the aortic relaxation of both young adult and middle-aged females, but not the males. CONCLUSIONS Endothelial dysfunction occurs earlier in males, but in females, dysfunction delays until middle age. GPER activation improves the vascular activities in females, but not males. It is promising to employ GPER as a potential drug target in cardiovascular disease in women.
Collapse
Affiliation(s)
- Xuan Yu
- Women's Health Division, Michael E. DeBakey Institute, Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TX, USA
| | - Peter Nguyen
- Women's Health Division, Michael E. DeBakey Institute, Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TX, USA
| | - Nioka C Burns
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, TX, USA
| | - Cristine L Heaps
- Women's Health Division, Michael E. DeBakey Institute, Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TX, USA
| | - John N Stallone
- Women's Health Division, Michael E. DeBakey Institute, Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TX, USA
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, TX, USA
| | - Guichun Han
- Women's Health Division, Michael E. DeBakey Institute, Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TX, USA; Department of Basic Sciences, Kentucky College of Osteopathic Medicine, University of Pikeville, KY, USA.
| |
Collapse
|
10
|
Sex Differences in Cardiovascular Diseases: A Matter of Estrogens, Ceramides, and Sphingosine 1-Phosphate. Int J Mol Sci 2022; 23:ijms23074009. [PMID: 35409368 PMCID: PMC8999971 DOI: 10.3390/ijms23074009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/30/2022] Open
Abstract
The medical community recognizes sex-related differences in pathophysiology and cardiovascular disease outcomes (CVD), culminating with heart failure. In general, pre-menopausal women tend to have a better prognosis than men. Explaining why this occurs is not a simple matter. For decades, sex hormones like estrogens (Es) have been identified as one of the leading factors driving these sex differences. Indeed, Es seem protective in women as their decline, during and after menopause, coincides with an increased CV risk and HF development. However, clinical trials demonstrated that E replacement in post-menopause women results in adverse cardiac events and increased risk of breast cancer. Thus, a deeper understanding of E-related mechanisms is needed to provide a vital gateway toward better CVD prevention and treatment in women. Of note, sphingolipids (SLs) and their metabolism are strictly related to E activities. Among the SLs, ceramide and sphingosine 1-phosphate play essential roles in mammalian physiology, particularly in the CV system, and appear differently modulated in males and females. In keeping with this view, here we explore the most recent experimental and clinical observations about the role of E and SL metabolism, emphasizing how these factors impact the CV system.
Collapse
|
11
|
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
|
12
|
Tutzauer J, Gonzalez de Valdivia E, Swärd K, Alexandrakis Eilard I, Broselid S, Kahn R, Olde B, Leeb-Lundberg LMF. Ligand-Independent G Protein-Coupled Estrogen Receptor/G Protein-Coupled Receptor 30 Activity: Lack of Receptor-Dependent Effects of G-1 and 17 β-Estradiol. Mol Pharmacol 2021; 100:271-282. [PMID: 34330822 PMCID: PMC8626787 DOI: 10.1124/molpharm.121.000259] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/06/2021] [Indexed: 11/22/2022] Open
Abstract
G protein-coupled receptor 30 (GPR30) is a membrane receptor reported to bind 17β-estradiol (E2) and mediate rapid nongenomic estrogen responses, hence also named G protein-coupled estrogen receptor. G-1 is a proposed GPR30-specific agonist that has been used to implicate the receptor in several pathophysiological events. However, controversy surrounds the role of GPR30 in G-1 and E2 responses. We investigated GPR30 activity in the absence and presence of G-1 and E2 in several eukaryotic systems ex vivo and in vitro in the absence and presence of the receptor. Ex vivo activity was addressed using the caudal artery from wild-type (WT) and GPR30 knockout (KO) mice, and in vitro activity was addressed using a HeLa cell line stably expressing a synthetic multifunctional promoter (nuclear factor κB, signal transducer and activator of transcription, activator protein 1)-luciferase construct (HFF11 cells) and a human GPR30-inducible T-REx system (T-REx HFF11 cells), HFF11 and human embryonic kidney 293 cells transiently expressing WT GPR30 and GPR30 lacking the C-terminal PDZ (postsynaptic density-95/discs-large /zonula occludens-1 homology) motif SSAV, and yeast Saccharomyces cerevisiae transformed to express GPR30. WT and KO arteries exhibited similar contractile responses to 60 mM KCl and 0.3 μM cirazoline, and G-1 relaxed both arteries with the same potency and efficacy. Furthermore, expression of GPR30 did not introduce any responses to 1 μM G-1 and 0.1 μM E2 in vitro. On the other hand, receptor expression caused considerable ligand-independent activity in vitro, which was receptor PDZ motif-dependent in mammalian cells. We conclude from these results that GPR30 exhibits ligand-independent activity in vitro but no G-1- or E2-stimulated activity in any of the systems used. SIGNIFICANCE STATEMENT: Much controversy surrounds 17β-estradiol (E2) and G-1 as G protein-coupled receptor 30 (GPR30) agonists. We used several recombinant eukaryotic systems ex vivo and in vitro with and without GPR30 expression to address the role of this receptor in responses to these proposed agonists. Our results show that GPR30 exhibits considerable ligand-independent activity in vitro but no G-1- or E2-stimulated activity in any of the systems used. Thus, classifying GPR30 as an estrogen receptor and G-1 as a specific GPR30 agonist is unfounded.
Collapse
Affiliation(s)
- Julia Tutzauer
- Department of Experimental Medical Science (J.T., E.G.d.V., K.S., I.A.E., S.B., L.M.F.L.-L.) and Department of Clinical Sciences Lund, Division of Pediatrics and Wallenberg Centre of Molecular Medicine (R.K.) and Division of Cardiology (B.O.), Lund University, Lund, Sweden
| | - Ernesto Gonzalez de Valdivia
- Department of Experimental Medical Science (J.T., E.G.d.V., K.S., I.A.E., S.B., L.M.F.L.-L.) and Department of Clinical Sciences Lund, Division of Pediatrics and Wallenberg Centre of Molecular Medicine (R.K.) and Division of Cardiology (B.O.), Lund University, Lund, Sweden
| | - Karl Swärd
- Department of Experimental Medical Science (J.T., E.G.d.V., K.S., I.A.E., S.B., L.M.F.L.-L.) and Department of Clinical Sciences Lund, Division of Pediatrics and Wallenberg Centre of Molecular Medicine (R.K.) and Division of Cardiology (B.O.), Lund University, Lund, Sweden
| | - Ioannis Alexandrakis Eilard
- Department of Experimental Medical Science (J.T., E.G.d.V., K.S., I.A.E., S.B., L.M.F.L.-L.) and Department of Clinical Sciences Lund, Division of Pediatrics and Wallenberg Centre of Molecular Medicine (R.K.) and Division of Cardiology (B.O.), Lund University, Lund, Sweden
| | - Stefan Broselid
- Department of Experimental Medical Science (J.T., E.G.d.V., K.S., I.A.E., S.B., L.M.F.L.-L.) and Department of Clinical Sciences Lund, Division of Pediatrics and Wallenberg Centre of Molecular Medicine (R.K.) and Division of Cardiology (B.O.), Lund University, Lund, Sweden
| | - Robin Kahn
- Department of Experimental Medical Science (J.T., E.G.d.V., K.S., I.A.E., S.B., L.M.F.L.-L.) and Department of Clinical Sciences Lund, Division of Pediatrics and Wallenberg Centre of Molecular Medicine (R.K.) and Division of Cardiology (B.O.), Lund University, Lund, Sweden
| | - Björn Olde
- Department of Experimental Medical Science (J.T., E.G.d.V., K.S., I.A.E., S.B., L.M.F.L.-L.) and Department of Clinical Sciences Lund, Division of Pediatrics and Wallenberg Centre of Molecular Medicine (R.K.) and Division of Cardiology (B.O.), Lund University, Lund, Sweden
| | - L M Fredrik Leeb-Lundberg
- Department of Experimental Medical Science (J.T., E.G.d.V., K.S., I.A.E., S.B., L.M.F.L.-L.) and Department of Clinical Sciences Lund, Division of Pediatrics and Wallenberg Centre of Molecular Medicine (R.K.) and Division of Cardiology (B.O.), Lund University, Lund, Sweden
| |
Collapse
|
13
|
Dinh QN, Vinh A, Arumugam TV, Drummond GR, Sobey CG. G protein-coupled estrogen receptor 1: a novel target to treat cardiovascular disease in a sex-specific manner? Br J Pharmacol 2021; 178:3849-3863. [PMID: 33948934 DOI: 10.1111/bph.15521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
As an agonist of the classical nuclear receptors, estrogen receptor-α and -β (NR3A1/2), estrogen has been assumed to inhibit the development of cardiovascular disease in premenopausal women. Indeed, reduced levels of estrogen after menopause are believed to contribute to accelerated morbidity and mortality rates in women. However, estrogen replacement therapy has variable effects on cardiovascular risk in postmenopausal women, including increased serious adverse events. Interestingly, preclinical studies have shown that selective activation of the novel membrane-associated G protein-coupled estrogen receptor, GPER, can promote cardiovascular protection. These benefits are more evident in ovariectomised than intact females or in males. It is therefore possible that selective targeting of the GPER in postmenopausal women could provide cardiovascular protection with fewer adverse effects that are caused by conventional 'receptor non-specific' estrogen replacement therapy. This review describes new data regarding the merits of targeting GPER to treat cardiovascular disease with a focus on sex differences.
Collapse
Affiliation(s)
- Quynh Nhu Dinh
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Antony Vinh
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
14
|
The G-Protein-Coupled Estrogen Receptor (GPER) Regulates Trimethylation of Histone H3 at Lysine 4 and Represses Migration and Proliferation of Ovarian Cancer Cells In Vitro. Cells 2021; 10:cells10030619. [PMID: 33799631 PMCID: PMC8001910 DOI: 10.3390/cells10030619] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023] Open
Abstract
Histone H3 lysine 4 trimethylation (H3K4me3) is one of the most recognized epigenetic regulators of transcriptional activity representing, an epigenetic modification of Histone H3. Previous reports have suggested that the broad H3K4me3 domain can be considered as an epigenetic signature for tumor-suppressor genes in human cells. G-protein-coupled estrogen receptor (GPER), a new membrane-bound estrogen receptor, acts as an inhibitor on cell growth via epigenetic regulation in breast and ovarian cancer cells. This study was conducted to evaluate the relationship of GPER and H3K4me3 in ovarian cancer tissue samples as well as in two different cell lines (Caov3 and Caov4). Silencing of GPER by a specific siRNA and two selective regulators with agonistic (G1) and antagonistic (G15) activity were applied for consecutive in vitro studies to investigate their impacts on tumor cell growth and the changes in phosphorylated ERK1/2 (p-ERK1/2) and H3K4me3. We found a positive correlation between GPER and H3K4me3 expression in ovarian cancer patients. Patients overexpressing GPER as well as H3K4me3 had significantly improved overall survival. Increased H3K4me3 and p-ERK1/2 levels and attenuated cell proliferation and migration were observed in Caov3 and Caov4 cells via activation of GPER by G1. Conversely, antagonizing GPER activity by G15 resulted in opposite effects in the Caov4 cell line. In conclusion, interaction of GPER and H3K4me3 appears to be of prognostic significance for ovarian cancer patients. The results of the in vitro analyses confirm the biological rationale for their interplay and identify GPER agonists, such as G1, as a potential therapeutic approach for future investigations.
Collapse
|
15
|
Gurrala R, Kilanowski-Doroh IM, Hutson DD, Ogola BO, Zimmerman MA, Katakam PVG, Satou R, Mostany R, Lindsey SH. Alterations in the estrogen receptor profile of cardiovascular tissues during aging. GeroScience 2021; 43:433-442. [PMID: 33558965 PMCID: PMC8050209 DOI: 10.1007/s11357-021-00331-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 01/31/2021] [Indexed: 12/13/2022] Open
Abstract
Estrogen exerts protective effects on the cardiovascular system via three known estrogen receptors: alpha (ERα), beta (ERß), and the G protein-coupled estrogen receptor (GPER). Our laboratory has previously showed the importance of GPER in the beneficial cardiovascular effects of estrogen. Since clinical studies indicate that the protective effects of exogenous estrogen on cardiovascular function are attenuated or reversed 10 years post-menopause, the hypothesis was that GPER expression may be reduced during aging. Vascular reactivity and GPER protein expression were assessed in female mice of varying ages. Physiological parameters, blood pressure, and estrogen receptor transcripts via droplet digital PCR (ddPCR) were assessed in the heart, kidney, and aorta of adult, middle-aged, and aged male and female C57BL/6 mice. Vasodilation to estrogen (E2) and the GPER agonist G-1 were reduced in aging female mice and were accompanied by downregulation of GPER protein. However, ERα and GPER were the predominant receptors in all tissues, whereas ERß was detectable only in the kidney. Female sex was associated with higher mRNA for both ERα and GPER in both the aorta and the heart. Aging impacted receptor transcript in a tissue-dependent manner. ERα transcript decreased in the heart with aging, while GPER expression increased in the heart. These data indicate that aging impacts estrogen receptor expression in the cardiovascular system in a tissue- and sex-specific manner. Understanding the impact of aging on estrogen receptor expression is critical for developing selective hormone therapies that protect from cardiovascular damage.
Collapse
Affiliation(s)
- Rakesh Gurrala
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | | | - Dillion D Hutson
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Margaret A Zimmerman
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA
| | - Ryousuke Satou
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, 7011, USA
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
- Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, 7011, USA.
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
16
|
Non-genomic actions of sex hormones on pregnant uterine contractility in rats: An in vitro study at term. Life Sci 2020; 263:118584. [PMID: 33058919 DOI: 10.1016/j.lfs.2020.118584] [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] [Received: 09/16/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 11/22/2022]
Abstract
AIMS The non-genomic (prompt) actions of sex steroids on pregnant uterine contractility are not fully explored yet, the aim of our study was to clarify such effects of 17-β estradiol (E2), progesterone (P4) and testosterone (T) on late (22-day) pregnant uterine contractions together with the signaling pathways in rats in vitro. METHODS The uterine effects of sex steroids on KCl-stimulated contractions were examined in the presence of genomic pathway blocker actinomycin D and cycloheximide, sex hormone receptor antagonists (flutamide, fulvestrant, mifepristone) and also after removing the endometrium. The modifications in uterine G-protein activation and cAMP levels were also detected. RESULTS T and E2 both relaxed the uterine contractions in the concentration range of 10-8-10-3 M with an increase in the activated G-protein and cAMP levels of the uterus, while P4 was ineffective. Cycloheximide, actinomycin D, antagonist for T and E2 were not able to modify the responses along with the endothelium removal. Mifepristone blocked the relaxing effects of T and E2 and reduced the activation of G-protein and the formation of cAMP. SIGNIFICANCE T and E2 can inhibit KCl-stimulated contractions in the late pregnant uterus in high concentrations and in a non-genomic manner. Their actions are mediated by a G-protein coupled receptor that can be blocked by mifepristone. A single and high dose of T or E2 might be considered in premature contractions, however, further preclinical and clinical studies are required for the approval of such a therapeutic intervention.
Collapse
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
Arise KK, Kumar P, Garg R, Samivel R, Zhao H, Pandya K, Nguyen C, Lindsey S, Pandey KN. Angiotensin II represses Npr1 expression and receptor function by recruitment of transcription factors CREB and HSF-4a and activation of HDACs. Sci Rep 2020; 10:4337. [PMID: 32152395 PMCID: PMC7062852 DOI: 10.1038/s41598-020-61041-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 02/20/2020] [Indexed: 01/10/2023] Open
Abstract
The two vasoactive hormones, angiotensin II (ANG II; vasoconstrictive) and atrial natriuretic peptide (ANP; vasodilatory) antagonize the biological actions of each other. ANP acting through natriuretic peptide receptor-A (NPRA) lowers blood pressure and blood volume. We tested hypothesis that ANG II plays critical roles in the transcriptional repression of Npr1 (encoding NPRA) and receptor function. ANG II significantly decreased NPRA mRNA and protein levels and cGMP accumulation in cultured mesangial cells and attenuated ANP-mediated relaxation of aortic rings ex vivo. The transcription factors, cAMP-response element-binding protein (CREB) and heat-shock factor-4a (HSF-4a) facilitated the ANG II-mediated repressive effects on Npr1 transcription. Tyrosine kinase (TK) inhibitor, genistein and phosphatidylinositol 3-kinase (PI-3K) inhibitor, wortmannin reversed the ANG II-dependent repression of Npr1 transcription and receptor function. ANG II enhanced the activities of Class I histone deacetylases (HDACs 1/2), thereby decreased histone acetylation of H3K9/14ac and H4K8ac. The repressive effect of ANG II on Npr1 transcription and receptor signaling seems to be transduced by TK and PI-3K pathways and modulated by CREB, HSF-4a, HDACs, and modified histones. The current findings suggest that ANG II-mediated repressive mechanisms of Npr1 transcription and receptor function may provide new molecular targets for treatment and prevention of hypertension and cardiovascular diseases.
Collapse
Affiliation(s)
- Kiran K Arise
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, USA
| | - Prerna Kumar
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, USA
| | - Renu Garg
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, USA
| | - Ramachandran Samivel
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, USA
| | - Hanqing Zhao
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, USA
| | - Krishna Pandya
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, USA
| | - Christian Nguyen
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, USA
| | - Sarah Lindsey
- Department of Pharmacology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, USA
| | - Kailash N Pandey
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
21
|
G-Protein–Coupled Estrogen Receptor Agonist G1 Improves Diastolic Function and Attenuates Cardiac Renin–Angiotensin System Activation in Estrogen-Deficient Hypertensive Rats. J Cardiovasc Pharmacol 2019; 74:443-452. [DOI: 10.1097/fjc.0000000000000721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
22
|
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
|
23
|
Zimmerman MA, Hutson DD, Mauvais-Jarvis F, Lindsey SH. Bazedoxifene-induced vasodilation and inhibition of vasoconstriction is significantly greater than estradiol. Menopause 2019; 26:172-181. [PMID: 30130290 PMCID: PMC6344253 DOI: 10.1097/gme.0000000000001195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE A new strategy for menopausal hormone therapy replaces medroxyprogesterone with the selective estrogen receptor modulator bazedoxifene. While the agonist or antagonist activity of bazedoxifene has been examined in other tissues, the current study explored the impact of bazedoxifene on resistance artery reactivity. We hypothesized that bazedoxifene may induce greater vasoprotective effects than estradiol due to enhanced activation of the G-protein-coupled estrogen receptor. METHODS We measured the vasodilation of mesenteric resistance arteries from adult male and female wild-type and G-protein-coupled estrogen receptor knockout mice (n = 58) in response to increasing concentrations of bazedoxifene, medroxyprogesterone, and estradiol, and also the impact of these compounds on the responses to phenylephrine and sodium nitroprusside. RESULTS Bazedoxifene-induced vasorelaxation was greater than estradiol and blunted phenylephrine-induced contraction-an effect not observed with estradiol. Neither estradiol nor bazedoxifene altered relaxation to sodium nitroprusside. The combination of bazedoxifene + estradiol promoted greater vasodilation than medroxyprogesterone + estradiol, and opposed phenylephrine-induced contraction, whereas medroxyprogesterone + estradiol failed to attenuate this response. Both bazedoxifene + estradiol and medroxyprogesterone + estradiol enhanced sodium nitroprusside-induced relaxation in females. Vascular responses were similar in both sexes in wild-type and G-protein-coupled estrogen receptor knockout mice. CONCLUSION Bazedoxifene and bazedoxifene + estradiol relaxed mesenteric arteries and opposed vasoconstriction to a greater degree than estradiol or medroxyprogesterone + estradiol. These effects were independent of sex and G-protein-coupled estrogen receptor expression. We conclude that bazedoxifene may provide vascular benefits over estrogen alone or estrogen plus progestogen combinations in postmenopausal women.
Collapse
Affiliation(s)
- Margaret A Zimmerman
- Department of Pharmacology, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA
| | - Dillion D Hutson
- Department of Pharmacology, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA
| | - Franck Mauvais-Jarvis
- Department of Medicine, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA
| | - Sarah H Lindsey
- Department of Pharmacology, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
24
|
Roque C, Mendes-Oliveira J, Baltazar G. G protein-coupled estrogen receptor activates cell type-specific signaling pathways in cortical cultures: relevance to the selective loss of astrocytes. J Neurochem 2019; 149:27-40. [PMID: 30570746 DOI: 10.1111/jnc.14648] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/23/2018] [Accepted: 12/11/2018] [Indexed: 12/11/2022]
Abstract
Selective activation of the G protein-coupled estrogen receptor has been proposed to avoid some of the side effects elicited by the activation of classical estrogen receptors α and β. Although its contribution to neuroprotection triggered by estradiol in brain disorders has been explored, the results regarding ischemic stroke are contradictory, and currently, there is no consensus on the role that this receptor may play. The present study aimed to investigate the role of GPER in the ischemic insult. For that, primary cortical cultures exposed to oxygen and glucose deprivation (OGD) were used as a model. Our results demonstrate that neuronal survival was strongly affected by the ischemic insult and concurrent GPER activation with G1 had no further impact. In contrast, OGD had a smaller impact on astrocytes survival but G1, alone or combined with OGD, promoted their apoptosis. This effect was prevented by the GPER antagonist G15. The results also show that ischemia did not change the expression levels of GPER in neurons and astrocytes. In this study, we also demonstrate that selective activation of GPER induced astrocyte apoptosis via the phospholipase C pathway and subsequent intracellular calcium rise, whereas in neurons, this effect was not observed. Taken together, this evidence supports a direct impact of GPER activity on the viability of astrocytes, which seems to be associated with the regulation of different signaling pathways in astrocytes and neurons.
Collapse
Affiliation(s)
- Cláudio Roque
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | - Graça Baltazar
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
25
|
Ogola BO, Zimmerman MA, Sure VN, Gentry KM, Duong JL, Clark GL, Miller KS, Katakam PVG, Lindsey SH. G Protein-Coupled Estrogen Receptor Protects From Angiotensin II-Induced Increases in Pulse Pressure and Oxidative Stress. Front Endocrinol (Lausanne) 2019; 10:586. [PMID: 31507536 PMCID: PMC6718465 DOI: 10.3389/fendo.2019.00586] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022] Open
Abstract
Our previous work showed that the G protein-coupled estrogen receptor (GPER) is protective in the vasculature and kidneys during angiotensin (Ang) II-dependent hypertension by inhibiting oxidative stress. The goal of the current study was to assess the impact of GPER deletion on sex differences in Ang II-induced hypertension and oxidative stress. Male and female wildtype and GPER knockout mice were implanted with radiotelemetry probes for measurement of baseline blood pressure before infusion of Ang II (700 ng/kg/min) for 2 weeks. Mean arterial pressure was increased to the same extent in all groups, but female wildtype mice were protected from Ang II-induced increases in pulse pressure, aortic wall thickness, and Nox4 mRNA. In vitro studies using vascular smooth muscle cells found that pre-treatment with the GPER agonist G-1 inhibited Ang II-induced ROS and NADP/NADPH. Ang II increased while G-1 decreased Nox4 mRNA and protein. The effects of Ang II were blocked by losartan and Nox4 siRNA, while the effects of G-1 were inhibited by adenylyl cyclase inhibition and mimicked by phosphodiesterase inhibition. We conclude that during conditions of elevated Ang II, GPER via the cAMP pathway suppresses Nox4 transcription to limit ROS production and prevent arterial stiffening. Taken together with our previous work, this study provides insight into how acute estrogen signaling via GPER provides cardiovascular protection during Ang II hypertension and potentially other diseases characterized by increased oxidative stress.
Collapse
Affiliation(s)
- Benard O. Ogola
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| | | | - Venkata N. Sure
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| | - Kaylee M. Gentry
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| | - Jennifer L. Duong
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| | - Gabrielle L. Clark
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, United States
| | - Kristin S. Miller
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, United States
| | | | - Sarah H. Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
- *Correspondence: Sarah H. Lindsey
| |
Collapse
|
26
|
Groban L, Tran QK, Ferrario CM, Sun X, Cheng CP, Kitzman DW, Wang H, Lindsey SH. Female Heart Health: Is GPER the Missing Link? Front Endocrinol (Lausanne) 2019; 10:919. [PMID: 31993020 PMCID: PMC6970950 DOI: 10.3389/fendo.2019.00919] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022] Open
Abstract
The G Protein-Coupled Estrogen Receptor (GPER) is a novel membrane-bound receptor that mediates non-genomic actions of the primary female sex hormone 17β-estradiol. Studies over the past two decades have elucidated the beneficial actions of this receptor in a number of cardiometabolic diseases. This review will focus specifically on the cardiac actions of GPER, since this receptor is expressed in cardiomyocytes as well as other cells within the heart and most likely contributes to estrogen-induced cardioprotection. Studies outlining the impact of GPER on diastolic function, mitochondrial function, left ventricular stiffness, calcium dynamics, cardiac inflammation, and aortic distensibility are discussed. In addition, recent data using genetic mouse models with global or cardiomyocyte-specific GPER gene deletion are highlighted. Since estrogen loss due to menopause in combination with chronological aging contributes to unique aspects of cardiac dysfunction in women, this receptor may provide novel therapeutic effects. While clinical studies are still required to fully understand the potential for pharmacological targeting of this receptor in postmenopausal women, this review will summarize the evidence gathered thus far on its likely beneficial effects.
Collapse
Affiliation(s)
- Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
- *Correspondence: Leanne Groban
| | - Quang-Kim Tran
- Department of Physiology & Pharmacology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, United States
| | - Carlos M. Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Physiology-Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Che Ping Cheng
- Department of Internal Medicine, Cardiovascular Medicine Section, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Dalane W. Kitzman
- Department of Internal Medicine, Cardiovascular Medicine Section, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
27
|
Abstract
LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Christopher J Langmead
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| |
Collapse
|
28
|
Mouat MA, Coleman JLJ, Smith NJ. GPCRs in context: sexual dimorphism in the cardiovascular system. Br J Pharmacol 2018; 175:4047-4059. [PMID: 29451687 DOI: 10.1111/bph.14160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/31/2018] [Accepted: 02/09/2018] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease (CVD) remains the largest cause of mortality worldwide, and there is a clear gender gap in disease occurrence, with men being predisposed to earlier onset of CVD, including atherosclerosis and hypertension, relative to women. Oestrogen may be a driving factor for female-specific cardioprotection, though androgens and sex chromosomes are also likely to contribute to sexual dimorphism in the cardiovascular system (CVS). Many GPCR-mediated processes are involved in cardiovascular homeostasis, and some exhibit clear sex divergence. Here, we focus on the G protein-coupled oestrogen receptor, endothelin receptors ETA and ETB and the eicosanoid G protein-coupled receptors (GPCRs), discussing the evidence and potential mechanisms leading to gender dimorphic responses in the vasculature. The use of animal models and pharmacological tools has been essential to understanding the role of these receptors in the CVS and will be key to further delineating their sex-specific effects. Ultimately, this may illuminate wider sex differences in cardiovascular pathology and physiology. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Margaret A Mouat
- Molecular Pharmacology Laboratory, Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, Australia
| | - James L J Coleman
- Molecular Pharmacology Laboratory, Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, Australia
| | - Nicola J Smith
- Molecular Pharmacology Laboratory, Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, Australia
| |
Collapse
|
29
|
Fredette NC, Meyer MR, Prossnitz ER. Role of GPER in estrogen-dependent nitric oxide formation and vasodilation. J Steroid Biochem Mol Biol 2018; 176:65-72. [PMID: 28529128 PMCID: PMC5694388 DOI: 10.1016/j.jsbmb.2017.05.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/11/2022]
Abstract
Estrogens are potent regulators of vasomotor tone, yet underlying receptor- and ligand-specific signaling pathways remain poorly characterized. The primary physiological estrogen 17β-estradiol (E2), a non-selective agonist of classical nuclear estrogen receptors (ERα and ERβ) as well as the G protein-coupled estrogen receptor (GPER), stimulates formation of the vasodilator nitric oxide (NO) in endothelial cells. Here, we studied the contribution of GPER signaling in E2-dependent activation of endothelial NO formation and subsequent vasodilation. Employing E2 and the GPER-selective agonist G-1, we investigated eNOS phosphorylation and NO formation in human endothelial cells, and endothelium-dependent vasodilation in the aortae of wild-type and Gper-deficient mice. Both E2 and G-1 induced phosphorylation of eNOS at the activation site Ser1177 to similar extents. Endothelial NO production to E2 was comparable to that of G-1, and was substantially reduced after pharmacological inhibition of GPER. Similarly, the clinically used ER-targeting drugs 4OH-tamoxifen, raloxifene, and ICI182,780 (faslodex, fulvestrant™) induced NO formation in part via GPER. We identified c-Src, EGFR, PI3K and ERK signaling pathways to be involved in GPER-dependent NO formation. In line with activation of NO formation in cells, E2 and G-1 induced equally potent vasodilation in the aorta of wild-type mice. Gper deletion completely abrogated the vasodilator response to G-1, while reducing the response to E2 by ∼50%. These findings indicate that a substantial portion of E2-induced endothelium-dependent vasodilation and NO formation is mediated by GPER. Thus, selective targeting of vascular GPER may be a suitable approach to activate the endothelial NO pathway, possibly leading to reduced vasomotor tone and inhibition of atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Natalie C Fredette
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; Current address: Department of Pathology, University of Florida, Gainesville, FL, USA
| | - Matthias R Meyer
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; Institute of Primary Care, University of Zurich, Zurich, Switzerland
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
30
|
Prossnitz ER. GPER modulators: Opportunity Nox on the heels of a class Akt. J Steroid Biochem Mol Biol 2018; 176:73-81. [PMID: 28285016 PMCID: PMC5591048 DOI: 10.1016/j.jsbmb.2017.03.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 12/14/2022]
Abstract
The (patho)physiology of estrogen and its receptors is complex. It is therefore not surprising that therapeutic approaches targeting this hormone include stimulation of its activity through supplementation with either the hormone itself or natural or synthetic agonists, inhibition of its activity through the use of antagonists or inhibitors of its synthesis, and tissue-selective modulation of its activity with biased ligands. The physiology of this hormone is further complicated by the existence of at least three receptors, the classical nuclear estrogen receptors α and β (ERα and ERβ), and the 7-transmembrane G protein-coupled estrogen receptor (GPER/GPR30), with overlapping but distinct pharmacologic profiles, particularly of anti-estrogenic ligands. GPER-selective ligands, as well as GPER knockout mice, have greatly aided our understanding of the physiological roles of GPER. Such ligands have revealed that GPER activation mediates many of the rapid cellular signaling events (including Ca2+ mobilization, ERK and PI3K/Akt activation) associated with estrogen activity, as opposed to the nuclear ERs that are traditionally described to function as ligand-induced transcriptional factors. Many of the salutary effects of estrogen throughout the body are reproduced by the GPER-selective agonist G-1, which, owing to its minimal effects on reproductive tissues, can be considered a non-feminizing estrogenic compound, and thus of potential therapeutic use in both women and men. On the contrary, until recently GPER-selective antagonists had predominantly found preclinical application in cancer models where estrogen stimulates cell growth and survival. This viewpoint changed recently with the discovery that GPER is associated with aging, particularly that of the cardiovascular system, where the GPER antagonist G36 reduced hypertension and GPER deficiency prevented cardiac fibrosis and vascular dysfunction with age, through the downregulation of Nox1 and as a consequence superoxide production. Thus, similar to the classical ERs, both agonists and antagonists of GPER may be of therapeutic benefit depending on the disease or condition to be treated.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States.
| |
Collapse
|
31
|
Iorga A, Cunningham CM, Moazeni S, Ruffenach G, Umar S, Eghbali M. The protective role of estrogen and estrogen receptors in cardiovascular disease and the controversial use of estrogen therapy. Biol Sex Differ 2017; 8:33. [PMID: 29065927 PMCID: PMC5655818 DOI: 10.1186/s13293-017-0152-8] [Citation(s) in RCA: 544] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/04/2017] [Indexed: 12/15/2022] Open
Abstract
Epidemiologic studies have previously suggested that premenopausal females have reduced incidence of cardiovascular disease (CVD) when compared to age-matched males, and the incidence and severity of CVD increases postmenopause. The lower incidence of cardiovascular disease in women during reproductive age is attributed at least in part to estrogen (E2). E2 binds to the traditional E2 receptors (ERs), estrogen receptor alpha (ERα), and estrogen receptor beta (ERβ), as well as the more recently identified G-protein-coupled ER (GPR30), and can exert both genomic and non-genomic actions. This review summarizes the protective role of E2 and its receptors in the cardiovascular system and discusses its underlying mechanisms with an emphasis on oxidative stress, fibrosis, angiogenesis, and vascular function. This review also presents the sexual dimorphic role of ERs in modulating E2 action in cardiovascular disease. The controversies surrounding the clinical use of exogenous E2 as a therapeutic agent for cardiovascular disease in women due to the possible risks of thrombotic events, cancers, and arrhythmia are also discussed. Endogenous local E2 biosynthesis from the conversion of testosterone to E2 via aromatase enzyme offers a novel therapeutic paradigm. Targeting specific ERs in the cardiovascular system may result in novel and possibly safer therapeutic options for cardiovascular protection.
Collapse
Affiliation(s)
- Andrea Iorga
- Present address: Department of Medicine, Division of Gastroenterology/Liver, Keck School of Medicine of the University of Southern California, Los Angeles, CA, 90033, USA
| | - Christine M Cunningham
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA
| | - Shayan Moazeni
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA
| | - Gregoire Ruffenach
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA
| | - Soban Umar
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA
| | - Mansoureh Eghbali
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-160CHS, Los Angeles, CA, 90095-7115, USA.
| |
Collapse
|
32
|
Kim SC, Boese AC, Moore MH, Cleland RM, Chang L, Delafontaine P, Yin KJ, Lee JP, Hamblin MH. Rapid estrogen receptor-α signaling mediated by ERK activation regulates vascular tone in male and ovary-intact female mice. Am J Physiol Heart Circ Physiol 2017; 314:H330-H342. [PMID: 28887333 DOI: 10.1152/ajpheart.00841.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Estrogen has been shown to affect vascular reactivity. Here, we assessed the estrogen receptor-α (ERα) dependency of estrogenic effects on vasorelaxation via a rapid nongenomic pathway in both male and ovary-intact female mice. We compared the effect of a primary estrogen, 17β-estradiol (E2) or 4,4',4″-(4-propyl-[1H]pyrazole-1,3,5-triyl)tris-phenol (PPT; selective ERα agonist). We found that E2 and PPT induced greater aortic relaxation in female mice than in male mice, indicating ERα mediation, which was further validated by using ERα antagonism. Treatment with 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole dihydrochloride (MPP dihydrochloride; ERα antagonist) attenuated PPT-mediated vessel relaxation in both sexes. ERα-mediated vessel relaxation was further validated by the absence of significant PPT-mediated relaxation in aortas isolated from ERα knockout mice. Treatment with a specific ERK inhibitor, PD-98059, reduced E2-induced vessel relaxation in both sexes but to a lesser extent in female mice. Furthermore, PD-98059 prevented PPT-induced vessel relaxation in both sexes. Both E2 and PPT treatment activated ERK as early as 5-10 min, which was attenuated by PD-98059 in aortic tissue, cultured primary vascular smooth muscle cells (VSMCs), and endothelial cells (ECs). Aortic rings denuded of endothelium showed no differences in vessel relaxation after E2 or PPT treatment, implicating a role of ECs in the observed sex differences. Here, our results are unique to show estrogen-stimulated rapid ERα signaling mediated by ERK activation in aortic tissue, as well as VSMCs and ECs in vitro, in regulating vascular function by using side-by-side comparisons in male and ovary-intact female mice in response to E2 or PPT. NEW & NOTEWORTHY Here, we assessed the estrogen receptor-α dependency of estrogenic effects in vasorelaxation of both male and ovary-intact female mice by performing side-by-side comparisons. Also, we describe the connection between estrogen-stimulated rapid estrogen receptor-α signaling and downstream ERK activation in regulating vascular function in male and ovary-intact female mice.
Collapse
Affiliation(s)
- Seong Chul Kim
- Department of Pharmacology, Tulane University School of Medicine , New Orleans, Louisiana
| | - Austin C Boese
- Department of Pharmacology, Tulane University School of Medicine , New Orleans, Louisiana
| | - Matthew H Moore
- Department of Pharmacology, Tulane University School of Medicine , New Orleans, Louisiana
| | - Rea M Cleland
- Department of Pharmacology, Tulane University School of Medicine , New Orleans, Louisiana
| | - Lin Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center , Ann Arbor, Michigan
| | - Patrice Delafontaine
- Heart and Vascular Institute, Tulane University School of Medicine , New Orleans, Louisiana
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine , New Orleans, Louisiana.,Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine , New Orleans, Louisiana
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine , New Orleans, Louisiana
| |
Collapse
|
33
|
Debortoli AR, Rouver WDN, Delgado NTB, Mengal V, Claudio ERG, Pernomian L, Bendhack LM, Moysés MR, Santos RLD. GPER modulates tone and coronary vascular reactivity in male and female rats. J Mol Endocrinol 2017; 59:171-180. [PMID: 28733475 DOI: 10.1530/jme-16-0117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 06/23/2017] [Indexed: 12/11/2022]
Abstract
Compared with age-matched men, premenopausal women are largely protected from coronary artery disease, a difference that is lost after menopause. The effects of oestrogens are mediated by the activation of nuclear receptors (ERα and ERβ) and by the G protein-coupled oestrogen receptor (GPER). This study aims to evaluate the potential role of GPER in coronary circulation in female and male rats. The baseline coronary perfusion pressure (CPP) and the concentration-response curve with a GPER agonist (G-1) were evaluated in isolated hearts before and after the blockade of GPER. GPER, superoxide dismutase (SOD-2), catalase and gp91phox protein expression were assessed by Western blotting. Superoxide production was evaluated 'in situ' via dihydroethidium fluorescence (DHE). GPER blockade significantly increased the CPP in both groups, demonstrating the modulation of coronary tone by GPER. G-1 causes relaxation of the coronary bed in a concentration-dependent manner and was significantly higher in female rats. No differences were detected in GPER, SOD-2 and catalase protein expression. However, gp91phox expression and DHE fluorescence were higher in male rats, indicating elevated superoxide production. Therefore, GPER plays an important role in modulating coronary tone and reactivity in female and male rats. The observed differences in vascular reactivity may be related to the higher superoxide production in male rats. These findings help to elucidate the role of GPER-modulating coronary circulation, providing new information to develop a potential therapeutic target for the treatment of coronary heart disease.
Collapse
Affiliation(s)
- Angelina Rafaela Debortoli
- Department of Physiological SciencesHealth Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Wender do Nascimento Rouver
- Department of Physiological SciencesHealth Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | | | - Vinicius Mengal
- Department of Physiological SciencesHealth Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | | | - Laena Pernomian
- Department of Physics and ChemistryFaculty of Pharmaceutical Sciences from Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Lusiane Maria Bendhack
- Department of Physics and ChemistryFaculty of Pharmaceutical Sciences from Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Margareth Ribeiro Moysés
- Department of Physiological SciencesHealth Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Roger Lyrio Dos Santos
- Department of Physiological SciencesHealth Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| |
Collapse
|
34
|
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]
|
35
|
Boese AC, Kim SC, Yin KJ, Lee JP, Hamblin MH. Sex differences in vascular physiology and pathophysiology: estrogen and androgen signaling in health and disease. Am J Physiol Heart Circ Physiol 2017. [PMID: 28626075 DOI: 10.1152/ajpheart.00217.2016] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sex differences between women and men are often overlooked and underappreciated when studying the cardiovascular system. It has been long assumed that men and women are physiologically similar, and this notion has resulted in women being clinically evaluated and treated for cardiovascular pathophysiological complications as men. Currently, there is increased recognition of fundamental sex differences in cardiovascular function, anatomy, cell signaling, and pathophysiology. The National Institutes of Health have enacted guidelines expressly to gain knowledge about ways the sexes differ in both normal function and diseases at the various research levels (molecular, cellular, tissue, and organ system). Greater understanding of these sex differences will be used to steer future directions in the biomedical sciences and translational and clinical research. This review describes sex-based differences in the physiology and pathophysiology of the vasculature, with a special emphasis on sex steroid receptor (estrogen and androgen receptor) signaling and their potential impact on vascular function in health and diseases (e.g., atherosclerosis, hypertension, peripheral artery disease, abdominal aortic aneurysms, cerebral aneurysms, and stroke).
Collapse
Affiliation(s)
- Austin C Boese
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Seong C Kim
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ke-Jie Yin
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jean-Pyo Lee
- Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana; and.,Center for Stem Cell Research and Regenerative Medicine, New Orleans, Louisiana
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana;
| |
Collapse
|
36
|
Clegg D, Hevener AL, Moreau KL, Morselli E, Criollo A, Van Pelt RE, Vieira-Potter VJ. Sex Hormones and Cardiometabolic Health: Role of Estrogen and Estrogen Receptors. Endocrinology 2017; 158:1095-1105. [PMID: 28323912 PMCID: PMC6283431 DOI: 10.1210/en.2016-1677] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 02/14/2017] [Indexed: 01/08/2023]
Abstract
With increased life expectancy, women will spend over three decades of life postmenopause. The menopausal transition increases susceptibility to metabolic diseases such as obesity, diabetes, cardiovascular disease, and cancer. Thus, it is more important than ever to develop effective hormonal treatment strategies to protect aging women. Understanding the role of estrogens, and their biological actions mediated by estrogen receptors (ERs), in the regulation of cardiometabolic health is of paramount importance to discover novel targeted therapeutics. In this brief review, we provide a detailed overview of the literature, from basic science findings to human clinical trial evidence, supporting a protective role of estrogens and their receptors, specifically ERα, in maintenance of cardiometabolic health. In so doing, we provide a concise mechanistic discussion of some of the major tissue-specific roles of estrogens signaling through ERα. Taken together, evidence suggests that targeted, perhaps receptor-specific, hormonal therapies can and should be used to optimize the health of women as they transition through menopause, while reducing the undesired complications that have limited the efficacy and use of traditional hormone replacement interventions.
Collapse
Affiliation(s)
- Deborah Clegg
- Department of Biomedical Sciences, Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Andrea L Hevener
- Department of Medicine, University of California Los Angeles, Los Angeles, California 90095
| | - Kerrie L Moreau
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Denver Veterans Administration Medical Center, Geriatric Research Education and Clinical Center, Denver, Colorado 80220
| | - Eugenia Morselli
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Universidad de Chile, Santiago 8380492, Chile
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 8380492, Chile
| | - Rachael E Van Pelt
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Victoria J Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri 65211
| |
Collapse
|
37
|
GPER-novel membrane oestrogen receptor. Clin Sci (Lond) 2017; 130:1005-16. [PMID: 27154744 DOI: 10.1042/cs20160114] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/02/2016] [Indexed: 12/11/2022]
Abstract
The recent discovery of the G protein-coupled oestrogen receptor (GPER) presents new challenges and opportunities for understanding the physiology, pathophysiology and pharmacology of many diseases. This review will focus on the expression and function of GPER in hypertension, kidney disease, atherosclerosis, vascular remodelling, heart failure, reproduction, metabolic disorders, cancer, environmental health and menopause. Furthermore, this review will highlight the potential of GPER as a therapeutic target.
Collapse
|
38
|
Pang Y, Thomas P. Additive effects of low concentrations of estradiol-17β and progesterone on nitric oxide production by human vascular endothelial cells through shared signaling pathways. J Steroid Biochem Mol Biol 2017; 165:258-267. [PMID: 27369115 DOI: 10.1016/j.jsbmb.2016.06.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/10/2016] [Accepted: 06/28/2016] [Indexed: 01/30/2023]
Abstract
Potential cardiovascular benefits of low-dose formulations of estrogens and progesterone (P4) for treating climacteric symptoms in postmenopausal women remain unclear because information is lacking on their combined vascular effects. Protective effects of low concentrations (5nM) of P4 and estradiol-17β (E2), alone and in combination (P4+E2), were investigated in a nongenomic model of vascular protection which measured acute increases in nitric oxide (NO) production by cultured human umbilical vein endothelial cells (HUVECs). Treatment with 5nM P4+E2 for twenty minutes significantly increased NO production and endothelial NO synthase (eNOS) phosphorylation, whereas 5nM treatments with either steroid alone were ineffective. The 5nM P4+E2 treatment also increased phosphorylation of ERK and Akt, mimicking the effects of higher concentrations of P4 and E2 alone. Pre-treatment with inhibitors of PI3K (wortmannin), Akt (ML-9), and MAP kinase (AZD6244 and U0126) completely blocked the NO response to 5nM P4+E2. Combined 5nM treatments with specific estrogen and progesterone receptor agonists showed an involvement of membrane progesterone receptor alpha (mPRα, also known as PAQR7), G protein-coupled estrogen receptor 1 (GPER), and estrogen receptor alpha (ERα), but not ERβ, in P4+E2 stimulation of NO production. P4+E2 also exerted genomic actions, increasing mPRα, GPER, cyclooxygenase-1, and prostacyclin-synthase mRNA levels. Taken together, the results show that a low concentration of P4+E2 rapidly increases NO production in HUVECs through mPRα, ERα, and GPER and involves common signaling pathways, PI3K/Akt and MAP kinase. These in vitro findings suggest that low doses of E2 and P4 may also have some beneficial cardiovascular effects in vivo when administered as hormone replacement therapy (HRT) for post-menopausal women.
Collapse
Affiliation(s)
- Yefei Pang
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, United States.
| | - Peter Thomas
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, United States.
| |
Collapse
|
39
|
Zimmerman MA, Hutson DD, Trimmer EH, Kashyap SN, Duong JL, Murphy B, Grissom EM, Daniel JM, Lindsey SH. Long- but not short-term estradiol treatment induces renal damage in midlife ovariectomized Long-Evans rats. Am J Physiol Renal Physiol 2016; 312:F305-F311. [PMID: 28153915 DOI: 10.1152/ajprenal.00411.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/04/2016] [Accepted: 11/04/2016] [Indexed: 01/25/2023] Open
Abstract
Clinical recommendations limit menopausal hormone therapy to a few years, yet the impact of a shorter treatment duration on cardiovascular health is unknown. We hypothesized that both short- and long-term estradiol (E2) treatment exerts positive and lasting effects on blood pressure, vascular reactivity, and renal health. This study was designed to mimic midlife menopause, followed by E2 treatment, that either followed or exceeded the current clinical recommendations. Female Long-Evans retired breeders were ovariectomized (OVX) at 11 mo of age and randomized into three groups: 80-day (80d) vehicle (Veh>Veh), 40-day (40d) E2 + 40d vehicle (E2>Veh), and 80d E2 (E2>E2). In comparison to Veh>Veh, both the E2>Veh and E2>E2 groups had lower systolic blood pressure and enhanced mesenteric relaxation in response to estrogen receptor-α stimulation. Despite the reduced blood pressure, E2>E2 induced renal and cardiac hypertrophy, reduced glomerular filtration, and increased proteinuria. Interestingly, kidneys from E2>Veh rats had significantly fewer tubular casts than both of the other groups. In conclusion, long-term E2 lowered blood pressure but exerted detrimental effects on kidney health in midlife OVX Long-Evans rats, whereas short-term E2 lowered blood pressure and reduced renal damage. These findings highlight that the duration of hormone therapy may be an important factor for renal health in aging postmenopausal women.
Collapse
Affiliation(s)
| | - Dillion D Hutson
- Department of Pharmacology, Tulane University, New Orleans, Louisiana
| | - Emma H Trimmer
- Department of Pharmacology, Tulane University, New Orleans, Louisiana
| | - Shreya N Kashyap
- Department of Pharmacology, Tulane University, New Orleans, Louisiana.,Tulane Brain Institute, New Orleans, Louisiana
| | - Jennifer L Duong
- Department of Pharmacology, Tulane University, New Orleans, Louisiana
| | - Brennah Murphy
- Department of Pharmacology, Tulane University, New Orleans, Louisiana.,Tulane Brain Institute, New Orleans, Louisiana
| | - Elin M Grissom
- Department of Psychology, Tulane University, New Orleans, Louisiana; and.,Tulane Brain Institute, New Orleans, Louisiana
| | - Jill M Daniel
- Department of Psychology, Tulane University, New Orleans, Louisiana; and.,Tulane Brain Institute, New Orleans, Louisiana
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University, New Orleans, Louisiana; .,Tulane Brain Institute, New Orleans, Louisiana
| |
Collapse
|
40
|
Feldman RD, Limbird LE. GPER (GPR30): A Nongenomic Receptor (GPCR) for Steroid Hormones with Implications for Cardiovascular Disease and Cancer. Annu Rev Pharmacol Toxicol 2016; 57:567-584. [PMID: 27814026 DOI: 10.1146/annurev-pharmtox-010716-104651] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Although the rapid effects of steroids, such as estrogen and aldosterone, were postulated originally to be nongenomic, it is now appreciated that activation of such signaling pathways via a steroid-acting G protein-coupled receptor, the G protein estrogen receptor (GPER), has important transcription-dependent outcomes in the regulation of cell growth and programmed cell death secondary to GPER-regulated second-messenger pathways. GPER is expressed ubiquitously and has diverse biological effects, including regulation of endocrine, immune, neuronal, and cardiovascular functions. Perhaps the most biologically important consequences of GPER activation are the regulation of cell growth, migration, and apoptotic cell death. These cell growth regulatory effects, important in cancer biology, are also relevant in the regulation of cardiac and vascular hypertrophy and in the response to ischemia. This review provides a summary of relevant findings of the impact of GPER regulation by either estradiol or aldosterone in in vitro model systems and extends those findings to in vivo studies of direct clinical relevance for development of GPER-directed agents for treatment of cancer and cardiovascular diseases associated with cellular proliferation.
Collapse
Affiliation(s)
- Ross D Feldman
- Discipline of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada A1B 3V6;
| | - Lee E Limbird
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee 37208
| |
Collapse
|
41
|
Meyer MR, Barton M. Estrogens and Coronary Artery Disease: New Clinical Perspectives. ADVANCES IN PHARMACOLOGY 2016; 77:307-60. [PMID: 27451102 DOI: 10.1016/bs.apha.2016.05.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In premenopausal women, endogenous estrogens are associated with reduced prevalence of arterial hypertension, coronary artery disease, myocardial infarction, and stroke. Clinical trials conducted in the 1990s such as HERS, WHI, and WISDOM have shown that postmenopausal treatment with horse hormone mixtures (so-called conjugated equine estrogens) and synthetic progestins adversely affects female cardiovascular health. Our understanding of rapid (nongenomic) and chronic (genomic) estrogen signaling has since advanced considerably, including identification of a new G protein-coupled estrogen receptor (GPER), which like the "classical" receptors ERα and ERβ is highly abundant in the cardiovascular system. Here, we discuss the role of estrogen receptors in the pathogenesis of coronary artery disease and review natural and synthetic ligands of estrogen receptors as well as their effects in physiology, on cardiovascular risk factors, and atherosclerotic vascular disease. Data from preclinical and clinical studies using nonselective compounds activating GPER, which include selective estrogen receptor modulators such as tamoxifen or raloxifene, selective estrogen receptor downregulators such as Faslodex™ (fulvestrant/ICI 182,780), vitamin B3 (niacin), green tea catechins, and soy flavonoids such as genistein or resveratrol, strongly suggest that activation of GPER may afford therapeutic benefit for primary and secondary prevention in patients with or at risk for coronary artery disease. Evidence from preclinical studies suggest similar efficacy profiles for selective small molecule GPER agonists such as G-1 which are devoid of uterotrophic activity. Further clinical research in this area is warranted to provide opportunities for future cardiovascular drug development.
Collapse
Affiliation(s)
- M R Meyer
- Triemli City Hospital, Zürich, Switzerland.
| | - M Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
42
|
Feldman RD. Heart Disease in Women: Unappreciated Challenges, GPER as a New Target. Int J Mol Sci 2016; 17:ijms17050760. [PMID: 27213340 PMCID: PMC4881581 DOI: 10.3390/ijms17050760] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 12/30/2022] Open
Abstract
Heart disease in women remains underappreciated, underdiagnosed and undertreated. Further, although we are starting to understand some of the social and behavioral determinants for this, the biological basis for the increased rate of rise in atherosclerosis risk in women after menopause remains very poorly understand. In this review we will outline the scope of the clinical issues related to heart disease in women, the emerging findings regarding the biological basis underlying the increased prevalence of atherosclerotic risk factors in postmenopausal women (vs. men) and the role of the G protein-coupled estrogen receptor (GPER) and its genetic regulation as a determinant of these sex-specific risks. GPER is a recently appreciated GPCR that mediates the rapid effects of estrogen and aldosterone. Recent studies have identified that GPER activation regulates both blood pressure. We have shown that regulation of GPER function via expression of a hypofunctional GPER genetic variant is an important determinant of blood pressure and risk of hypertension in women. Further, our most recent studies have identified that GPER activation is an important regulator of low density lipoprotein (LDL) receptor metabolism and that expression of the hypofunctional GPER genetic variant is an important contributor to the development of hypercholesterolemia in women. GPER appears to be an important determinant of the two major risk factors for coronary artery disease-blood pressure and LDL cholesterol. Further, the importance of this mechanism appears to be greater in women. Thus, the appreciation of the role of GPER function as a determinant of the progression of atherosclerotic disease may be important both in our understanding of cardiometabolic function but also in opening the way to greater appreciation of the sex-specific regulation of atherosclerotic risk factors.
Collapse
Affiliation(s)
- Ross D Feldman
- Discipline of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada.
| |
Collapse
|
43
|
Sen A, Kumar P, Garg R, Lindsey SH, Katakam PVG, Bloodworth M, Pandey KN. Transforming growth factor β1 antagonizes the transcription, expression and vascular signaling of guanylyl cyclase/natriuretic peptide receptor A - role of δEF1. FEBS J 2016; 283:1767-81. [PMID: 26934489 DOI: 10.1111/febs.13701] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 01/20/2016] [Accepted: 02/29/2016] [Indexed: 12/19/2022]
Abstract
The objective of this study was to determine the role of transforming growth factor β1 (TGF-β1) in transcriptional regulation and function of the guanylyl cyclase A/natriuretic peptide receptor A gene (Npr1) and whether cross-talk exists between these two hormonal systems in target cells. After treatment of primary cultured rat thoracic aortic vascular smooth muscle cells and mouse mesangial cells with TGF-β1, the Npr1 promoter construct containing a δ-crystallin enhancer binding factor 1 (δEF1) site showed 85% reduction in luciferase activity in a time- and dose-dependent manner. TGF-β1 also significantly attenuated luciferase activity of the Npr1 promoter by 62%, and decreased atrial natriuretic peptide-mediated relaxation of mouse denuded aortic rings ex vivo. Treatment of cells with TGF-β1 increased the protein levels of δEF1 by 2.4-2.8-fold, and also significantly enhanced the phosphorylation of Smad 2/3, but markedly reduced Npr1 mRNA and receptor protein levels. Over-expression of δEF1 showed a reduction in Npr1 promoter activity by 75%, while deletion or site-directed mutagenesis of δEF1 sites in the Npr1 promoter eliminated the TGF-β1-mediated repression of Npr1 transcription. TGF-β1 significantly increased the expression of α-smooth muscle actin and collagen type I α2 in rat thoracic aortic vascular smooth muscle cells, which was markedly attenuated by atrial natriuretic peptide in cells over-expressing natriuretic peptide receptor A. Together, the present results suggest that an antagonistic cascade exists between the TGF-β1/Smad/δEF1 pathways and Npr1 expression and receptor signaling that is relevant to renal and vascular remodeling, and may be critical in the regulation of blood pressure and cardiovascular homeostasis.
Collapse
Affiliation(s)
- Anagha Sen
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Prerna Kumar
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Renu Garg
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Meaghan Bloodworth
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Kailash N Pandey
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| |
Collapse
|
44
|
Menazza S, Murphy E. The Expanding Complexity of Estrogen Receptor Signaling in the Cardiovascular System. Circ Res 2016; 118:994-1007. [PMID: 26838792 DOI: 10.1161/circresaha.115.305376] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/28/2015] [Indexed: 12/21/2022]
Abstract
Estrogen has important effects on cardiovascular function including regulation of vascular function, blood pressure, endothelial relaxation, and the development of hypertrophy and cardioprotection. However, the mechanisms by which estrogen mediates these effects are still poorly understood. As detailed in this review, estrogen can regulate transcription by binding to 2 nuclear receptors, ERα and ERβ, which differentially regulate gene transcription. ERα and ERβ regulation of gene transcription is further modulated by tissue-specific coactivators and corepressors. Estrogen can bind to ERα and ERβ localized at the plasma membrane as well as G-protein-coupled estrogen receptor to initiate membrane delimited signaling, which enhances kinase signaling pathways that can have acute and long-term effects. The kinase signaling pathways can also mediate transcriptional changes and can synergize with the ER to regulate cell function. This review will summarize the beneficial effects of estrogen in protecting the cardiovascular system through ER-dependent mechanisms with an emphasis on the role of the recently described ER membrane signaling mechanisms.
Collapse
Affiliation(s)
- Sara Menazza
- From the Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD.
| | - Elizabeth Murphy
- From the Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
45
|
Tropea T, De Francesco EM, Rigiracciolo D, Maggiolini M, Wareing M, Osol G, Mandalà M. Pregnancy Augments G Protein Estrogen Receptor (GPER) Induced Vasodilation in Rat Uterine Arteries via the Nitric Oxide - cGMP Signaling Pathway. PLoS One 2015; 10:e0141997. [PMID: 26536245 PMCID: PMC4633123 DOI: 10.1371/journal.pone.0141997] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 10/15/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The regulation of vascular tone in the uterine circulation is a key determinant of appropriate uteroplacental blood perfusion and successful pregnancy outcome. Estrogens, which increase in the maternal circulation throughout pregnancy, can exert acute vasodilatory actions. Recently a third estrogen receptor named GPER (G protein-coupled estrogen receptor) was identified and, although several studies have shown vasodilatory effects in several vascular beds, nothing is known about its role in the uterine vasculature. AIM The aim of this study was to determine the function of GPER in uterine arteries mainly during pregnancy. Uterine arteries were isolated from nonpregnant and pregnant rats. METHODS Vessels were contracted with phenylephrine and then incubated with incremental doses (10-12-10-5 M) of the selective GPER agonist G1. RESULTS G1 induced a dose-dependent vasodilation which was: 1) significantly increased in pregnancy, 2) endothelium-dependent, 3) primarily mediated by NO/cGMP pathway and 4) unaffected by BKca channel inhibition. CONCLUSION This is the first study to show the potential importance of GPER signaling in reducing uterine vascular tone during pregnancy. GPER may therefore play a previously unrecognized role in the regulation of uteroplacental blood flow and normal fetus growth.
Collapse
Affiliation(s)
- Teresa Tropea
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | | | - Damiano Rigiracciolo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Mark Wareing
- Maternal and Fetal Health Research Centre, The University of Manchester, Manchester, United Kingdom
| | - George Osol
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont, Burlington, Vermont, United States of America
| | - Maurizio Mandalà
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
- * E-mail:
| |
Collapse
|
46
|
Ge LC, Wang HS. A commentary on "Involvement of activating ERK1/2 trough G protein coupled receptor 30 and estrogen receptor α/β in low doses of bisphenol A promoting growth of Sertoli TM4 cells". Toxicol Lett 2015; 240:236-7. [PMID: 26427357 DOI: 10.1016/j.toxlet.2015.09.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Li-Chen Ge
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
47
|
Mata KM, Li W, Reslan OM, Siddiqui WT, Opsasnick LA, Khalil RA. Adaptive increases in expression and vasodilator activity of estrogen receptor subtypes in a blood vessel-specific pattern during pregnancy. Am J Physiol Heart Circ Physiol 2015; 309:H1679-96. [PMID: 26408543 DOI: 10.1152/ajpheart.00532.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/09/2015] [Indexed: 11/22/2022]
Abstract
Normal pregnancy is associated with adaptive hemodynamic, hormonal, and vascular changes, and estrogen (E2) may promote vasodilation during pregnancy; however, the specific E2 receptor (ER) subtype, post-ER signaling mechanism, and vascular bed involved are unclear. We tested whether pregnancy-associated vascular adaptations involve changes in the expression/distribution/activity of distinct ER subtypes in a blood vessel-specific manner. Blood pressure (BP) and plasma E2 were measured in virgin and pregnant (day 19) rats, and the thoracic aorta, carotid artery, mesenteric artery, and renal artery were isolated for measurements of ERα, ERβ, and G protein-coupled receptor 30 [G protein-coupled ER (GPER)] expression and tissue distribution in parallel with relaxation responses to E2 (all ERs) and the specific ER agonist 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)-tris-phenol (PPT; ERα), diarylpropionitrile (DPN; ERβ), and G1 (GPER). BP was slightly lower and plasma E2 was higher in pregnant versus virgin rats. Western blots revealed increased ERα and ERβ in the aorta and mesenteric artery and GPER in the aorta of pregnant versus virgin rats. Immunohistochemistry revealed that the increases in ERs were mainly in the intima and media. In phenylephrine-precontracted vessels, E2 and PPT caused relaxation that was greater in the aorta and mesenteric artery but similar in the carotid and renal artery of pregnant versus virgin rats. DPN- and G1-induced relaxation was greater in the mesenteric and renal artery than in the aorta and carotid artery, and aortic relaxation to G1 was greater in pregnant versus virgin rats. The nitric oxide synthase inhibitor N(ω)-nitro-l-arginine methyl ester with or without the cyclooxygenase inhibitor indomethacin with or without the EDHF blocker tetraethylammonium or endothelium removal reduced E2, PPT, and G1-induced relaxation in the aorta of pregnant rats, suggesting an endothelium-dependent mechanism, but did not affect E2-, PPT-, DPN-, or G1-induced relaxation in other vessels, suggesting endothelium-independent mechanisms. E2, PPT, DPN, and G1 caused relaxation of Ca(2+) entry-dependent KCl contraction, and the effect of PPT was greater in the mesenteric artery of pregnant versus virgin rats. Thus, during pregnancy, an increase in ERα expression in endothelial and vascular smooth muscle layers of the aorta and mesenteric artery is associated with increased ERα-mediated relaxation via endothelium-derived vasodilators and inhibition of Ca(2+) entry into vascular smooth muscle, supporting a role of aortic and mesenteric arterial ERα in pregnancy-associated vasodilation. GPER may contribute to aortic relaxation while enhanced ERβ expression could mediate other genomic vascular effects during pregnancy.
Collapse
Affiliation(s)
- Karina M Mata
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wei Li
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ossama M Reslan
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Waleed T Siddiqui
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lauren A Opsasnick
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
48
|
Prossnitz ER, Hathaway HJ. What have we learned about GPER function in physiology and disease from knockout mice? J Steroid Biochem Mol Biol 2015; 153:114-26. [PMID: 26189910 PMCID: PMC4568147 DOI: 10.1016/j.jsbmb.2015.06.014] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 12/16/2022]
Abstract
Estrogens, predominantly 17β-estradiol, exert diverse effects throughout the body in both normal and pathophysiology, during development and in reproductive, metabolic, endocrine, cardiovascular, nervous, musculoskeletal and immune systems. Estrogen and its receptors also play important roles in carcinogenesis and therapy, particularly for breast cancer. In addition to the classical nuclear estrogen receptors (ERα and ERβ) that traditionally mediate predominantly genomic signaling, the G protein-coupled estrogen receptor GPER has become recognized as a critical mediator of rapid signaling in response to estrogen. Mouse models, and in particular knockout (KO) mice, represent an important approach to understand the functions of receptors in normal physiology and disease. Whereas ERα KO mice display multiple significant defects in reproduction and mammary gland development, ERβ KO phenotypes are more limited, and GPER KO exhibit no reproductive deficits. However, the study of GPER KO mice over the last six years has revealed that GPER deficiency results in multiple physiological alterations including obesity, cardiovascular dysfunction, insulin resistance and glucose intolerance. In addition, the lack of estrogen-mediated effects in numerous tissues of GPER KO mice, studied in vivo or ex vivo, including those of the cardiovascular, endocrine, nervous and immune systems, reveals GPER as a genuine mediator of estrogen action. Importantly, GPER KO mice have also demonstrated roles for GPER in breast carcinogenesis and metastasis. In combination with the supporting effects of GPER-selective ligands and GPER knockdown approaches, GPER KO mice demonstrate the therapeutic potential of targeting GPER activity in diseases as diverse as obesity, diabetes, multiple sclerosis, hypertension, atherosclerosis, myocardial infarction, stroke and cancer.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, United States; University of New Mexico Cancer Center, Albuquerque, NM 87131, United States.
| | - Helen J Hathaway
- Department of Cell Biology & Physiology, University of New Mexico, Albuquerque, NM 87131, United States; University of New Mexico Cancer Center, Albuquerque, NM 87131, United States.
| |
Collapse
|
49
|
Moreno-Ulloa A, Mendez-Luna D, Beltran-Partida E, Castillo C, Guevara G, Ramirez-Sanchez I, Correa-Basurto J, Ceballos G, Villarreal F. The effects of (-)-epicatechin on endothelial cells involve the G protein-coupled estrogen receptor (GPER). Pharmacol Res 2015; 100:309-20. [PMID: 26303816 DOI: 10.1016/j.phrs.2015.08.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 02/05/2023]
Abstract
We have provided evidence that the stimulatory effects of (-)-epicatechin ((-)-EPI) on endothelial cell nitric oxide (NO) production may involve the participation of a cell-surface receptor. Thus far, such entity(ies) has not been fully elucidated. The G protein-coupled estrogen receptor (GPER) is a cell-surface receptor that has been linked to protective effects on the cardiovascular system and activation of intracellular signaling pathways (including NO production) similar to those reported with (-)-EPI. In bovine coronary artery endothelial cells (BCAEC) by the use of confocal imaging, we evidence the presence of GPER at the cell-surface and on F-actin filaments. Using in silico studies we document the favorable binding mode between (-)-EPI and GPER. Such binding is comparable to that of the GPER agonist, G1. By the use of selective blockers, we demonstrate that the activation of ERK 1/2 and CaMKII by (-)-EPI is dependent on the GPER/c-SRC/EGFR axis mimicking those effects noted with G1. We also evidence by the use of siRNA the role that GPER has on mediating ERK1/2 activation by (-)-EPI. GPER appears to be coupled to a non Gαi/o or Gαs, protein subtype. To extrapolate our findings to an ex vivo model, we employed phenylephrine pre-contracted aortic rings evidencing that (-)-EPI can mediate vasodilation through GPER activation. In conclusion, we provide evidence that suggests the GPER as a potential mediator of (-)-EPI effects and highlights the important role that GPER may have on cardiovascular system protection.
Collapse
Affiliation(s)
- Aldo Moreno-Ulloa
- University of California, San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA, USA; Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - David Mendez-Luna
- Laboratorio de modelado Molecular y Diseño de Fármacos, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | | | - Carmen Castillo
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Gustavo Guevara
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Israel Ramirez-Sanchez
- University of California, San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA, USA; Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - José Correa-Basurto
- Laboratorio de modelado Molecular y Diseño de Fármacos, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico; Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Guillermo Ceballos
- Laboratorio de Investigación Integral Cardiometabólica, Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Francisco Villarreal
- University of California, San Diego, School of Medicine, 9500 Gilman Drive, La Jolla, CA, USA.
| |
Collapse
|
50
|
Santollo J, Daniels D. Activation of G protein-coupled estrogen receptor 1 (GPER-1) decreases fluid intake in female rats. Horm Behav 2015; 73:39-46. [PMID: 26093261 PMCID: PMC4546888 DOI: 10.1016/j.yhbeh.2015.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 05/19/2015] [Accepted: 05/28/2015] [Indexed: 11/19/2022]
Abstract
Estradiol (E2) decreases fluid intake in the female rat and recent studies from our lab demonstrate that the effect is at least in part mediated by membrane-associated estrogen receptors. Because multiple estrogen receptor subtypes can localize to the cell membrane, it is unclear which receptor(s) is generating the anti-dipsogenic effect of E2. The G protein-coupled estrogen receptor 1 (GPER-1) is a particularly interesting possibility because it has been shown to regulate blood pressure; many drinking-regulatory systems play overlapping roles in the control of blood pressure. Accordingly, we tested the hypothesis that activation of GPER-1 is sufficient to decrease fluid intake in female rats. In support of this hypothesis we found that treatment with the selective GPER-1 agonist G1 reduced AngII-stimulated fluid intake in OVX rats. Given the close association between food and fluid intakes in rats, and previous reports suggesting GPER-1 plays a role in energy homeostasis, we tested the hypothesis that the effect of GPER-1 on fluid intake was caused by a more direct effect on food intake. We found, however, that G1-treatment did not influence short-term or overnight food intake in OVX rats. Together these results reveal a novel effect of GPER-1 in the control of drinking behavior and provide an example of the divergence in the controls of fluid and food intakes in female rats.
Collapse
Affiliation(s)
- Jessica Santollo
- Department of Psychology, University at Buffalo SUNY, Buffalo, NY 14260, United States
| | - Derek Daniels
- Department of Psychology, University at Buffalo SUNY, Buffalo, NY 14260, United States.
| |
Collapse
|