1
|
Al-Karagholi MAM, Gram C, Nielsen CAW, Ashina M. Targeting BK Ca Channels in Migraine: Rationale and Perspectives. CNS Drugs 2020; 34:325-335. [PMID: 32060729 DOI: 10.1007/s40263-020-00706-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Large (big)-conductance calcium-activated potassium (BKCa) channels are expressed in migraine-related structures such as the cranial arteries, trigeminal ganglion and trigeminal spinal nucleus, and they play a substantial role in vascular tonus and neuronal excitability. Using synthetic BKCa channels openers was associated with headache as a frequent adverse effect in healthy volunteers. Additionally, BKCa channels are downstream molecules in migraine signalling pathways that are activated by several compounds known to provoke migraine, including calcitonin gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP) and glyceryl trinitrate (GTN). Also, there is a high affinity and a close coupling between BKCa channels and ATP-sensitive potassium (KATP) channels, the role of which has recently been established in migraine pathophysiology. These observations raise the question as to whether direct BKCa channel activation can provoke migraine in migraine patients, and whether the BKCa channel could be a potential novel anti-migraine target. Hence, randomized and placebo-controlled clinical studies on BKCa channel openers or blockers in migraine patients are needed.
Collapse
Affiliation(s)
- Mohammad Al-Mahdi Al-Karagholi
- Danish Headache Center, Department of Neurology, University of Copenhagen, Valdemar Hansen Vej 5, 2600, Glostrup, Denmark
| | - Christian Gram
- Danish Headache Center, Department of Neurology, University of Copenhagen, Valdemar Hansen Vej 5, 2600, Glostrup, Denmark
| | - Cherie Amalie Waldorff Nielsen
- Danish Headache Center, Department of Neurology, University of Copenhagen, Valdemar Hansen Vej 5, 2600, Glostrup, Denmark
| | - Messoud Ashina
- Danish Headache Center, Department of Neurology, University of Copenhagen, Valdemar Hansen Vej 5, 2600, Glostrup, Denmark. .,Glostrup Research Park, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Glostrup, Denmark.
| |
Collapse
|
2
|
Filgueira FP, Lobato NS, Nascimento DL, Ceravolo GS, Giachini FRC, Lima VV, Dantas AP, Fortes ZB, Webb RC, Tostes RC, Carvalho MHC. Equilin displays similar endothelium-independent vasodilator potential to 17β-estradiol regardless of lower potential to inhibit calcium entry. Steroids 2019; 141:46-54. [PMID: 30458188 PMCID: PMC6984400 DOI: 10.1016/j.steroids.2018.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/21/2018] [Accepted: 11/14/2018] [Indexed: 12/24/2022]
Abstract
Conjugated equine estrogens (CEE) have been widely used by women who seek to relieve symptoms of menopause. Despite evidence describing protective effects against risk factors for cardiovascular diseases by naturally occurring estrogens, little is known about the vascular effects of equilin, one of the main components of CEE and not physiologically present in women. In this regard, the present study aims to compare the vascular effects of equilin in an experimental model of hypertension with those induced by 17β-estradiol. Resistance mesenteric arteries from female spontaneously hypertensive rats (SHR) were used for recording isometric tension in a small vessel myograph. As effectively as 17β-estradiol, equilin evoked a concentration-dependent relaxation in mesenteric arteries from female SHRs contracted with KCl, U46619, PDBu or ET-1. Equilin-induced vasodilation does not involve classical estrogen receptor activation, since the estrogen receptor antagonist (ICI 182,780) failed to inhibit relaxation in U46619-precontracted mesenteric arteries. Vasorelaxation was not affected by either endothelium removal or by inhibiting the release or action of endothelium-derived factors. Incubation with L-NAME (NOS inhibitor), ODQ (guanylyl cyclase inhibitor) or KT5823 (inhibitor of protein kinase G) did not affect equilin-induced relaxation. Similarly, indomethacin (COX inhibitor) or blockage of potassium channels with tetraethylammonium, glibenclamide, 4-aminopyridine, or ouabain did not affect equilin-induced relaxation. Inhibitors of adenylyl cyclase SQ22536 or protein kinase A (KT5720) also had no effects on equilin-induced relaxation. While 17β-estradiol inhibited calcium (Ca2+) -induced contractions in high-K+ depolarization medium in a concentration-dependent manner, equilin induced a slight rightward-shift in the contractile responses to Ca2+. Comparable pattern of responses were observed in the concentration-response curves to (S)-(-)-Bay K 8644, a L-type Ca2+ channel activator. Equilin was unable to block the transitory contraction produced by caffeine-induced Ca2+ release from intracellular stores. In conclusion, equilin blocks L-type Ca2+ channels less effectively than 17β-estradiol. Despite its lower effectiveness, equilin equally relaxes resistance mesenteric arteries by blocking Ca2+ entry on smooth muscle.
Collapse
Affiliation(s)
- Fernando P Filgueira
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Department of Physiology, Augusta University, Augusta, GA, USA; Faculty of Medicine, Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil.
| | - Núbia S Lobato
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Department of Physiology, Augusta University, Augusta, GA, USA; Faculty of Medicine, Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil
| | - Denise L Nascimento
- Faculty of Medicine, Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil
| | - Graziela S Ceravolo
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Department of Physiological Sciences, Biological Sciences Center, State University of Londrina, Londrina, PR, Brazil
| | - Fernanda R C Giachini
- Department of Physiology, Augusta University, Augusta, GA, USA; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, MT, Brazil
| | - Victor V Lima
- Department of Physiology, Augusta University, Augusta, GA, USA; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, MT, Brazil; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Ana Paula Dantas
- Experimental Cardiology, Institut Clínic Cardiovascular, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Zuleica B Fortes
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Maria Helena C Carvalho
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
3
|
Zhang Q, Tian P, Zhai M, Lei X, Yang Z, Liu Y, Liu M, Huang H, Zhang X, Yang X, Zhao Y, Meng Z. Formaldehyde regulates vascular tensions through nitric oxide-cGMP signaling pathway and ion channels. CHEMOSPHERE 2018; 193:60-73. [PMID: 29126066 DOI: 10.1016/j.chemosphere.2017.11.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 10/26/2017] [Accepted: 11/03/2017] [Indexed: 06/07/2023]
Abstract
Formaldehyde (FA) has been linked to the detrimental cardiovascular effects. Here, we explored the effects and mechanisms of FA on rat aortas both in vivo and in vitro. The results presented that FA evidently lowered the blood pressures of rats. The expression levels of BKCa subunits α and β1 and iNOS of the aortas were up-regulated by FA in vivo. However, FA markedly reduced the levels of Cav1.2 and Cav1.3, which are the subunits of L-Ca2+ channel. Furthermore, the contents of NO, cGMP and iNOS in the aortas were augmented by FA. To further confirm these findings, the mechanisms accredited to these effects were examined in vitro. The data showed that FA contracted the isolated aortic rings at low concentrations (<300 μM), while it relaxed the rings at high concentrations (>500 μM). The FA-induced vasoconstriction at low concentrations was blocked partly by an inhibitor of ACE. The relaxation caused by FA at high concentrations was attenuated by the inhibitors of NO-cGMP pathway, L-Ca2+ channel and BKCa channel, respectively. Similarly, the expression of iNOS was strongly enhanced by FA in vitro. The effects of FA on the aortic rings with endothelium were significantly greater than those on the rings without endothelium. Our results indicate that the vasoconstriction of FA at low concentrations might be partially pertinent to endothelin, and the FA-caused vasorelaxation at high concentrations is possibly associated with the NO-cGMP pathway, L-Ca2+ channel and BKCa channel. This study will improve our understanding of the pathogenic mechanisms for FA-related cardiovascular diseases.
Collapse
Affiliation(s)
- Quanxi Zhang
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China.
| | - Peiru Tian
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Miaomiao Zhai
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Xiaodong Lei
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Zhenhua Yang
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Yan Liu
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Mengting Liu
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Hao Huang
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Xiri Zhang
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Xu Yang
- Laboratory of Environmental Biomedicine, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Huazhong Normal University, Wuhan 430079, China.
| | - Yun Zhao
- Laboratory of Environmental Biomedicine, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Huazhong Normal University, Wuhan 430079, China
| | - Ziqiang Meng
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
4
|
Wong CM, Au CL, Tsang SY, Lau CW, Yao X, Cai Z, Chung ACK. Role of inducible nitric oxide synthase in endothelium-independent relaxation to raloxifene in rat aorta. Br J Pharmacol 2017; 174:718-733. [PMID: 28138957 DOI: 10.1111/bph.13733] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/04/2016] [Accepted: 01/18/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Raloxifene can induce both endothelium-dependent and -independent relaxation in different arteries. However, the underlying mechanisms by which raloxifene triggers endothelium-independent relaxation are still incompletely understood. The purpose of present study was to examine the roles of NOSs and Ca2+ channels in the relaxant response to raloxifene in the rat isolated, endothelium-denuded aorta. EXPERIMENTAL APPROACH Changes in isometric tension, cGMP, nitrite, inducible NOS protein expression and distribution in response to raloxifene in endothelium-denuded aortic rings were studied by organ baths, radioimmunoassay, Griess reaction, western blot and immunohistochemistry respectively. KEY RESULTS Raloxifene reduced the contraction to CaCl2 in a Ca2+ -free, high K+ -containing solution in intact aortic rings. Raloxifene also acutely relaxed the aorta primarily through an endothelium-independent mechanism involving NO, mostly from inducible NOS (iNOS) in vascular smooth muscle layers. This effect of raloxifene involved the generation of cGMP and nitrite. Also, it was genomic in nature, as it was inhibited by a classical oestrogen receptor antagonist and inhibitors of RNA and protein synthesis. Raloxifene-induced stimulation of iNOS gene expression was partly mediated through activation of the NF-κB pathway. Raloxifene was more potent than 17β-estradiol or tamoxifen at relaxing endothelium-denuded aortic rings by stimulation of iNOS. CONCLUSIONS AND IMPLICATIONS Raloxifene-mediated vasorelaxation in rat aorta is independent of a functional endothelium and is mediated by oestrogen receptors and NF-κB. This effect is mainly mediated through an enhanced production of NO, cGMP and nitrite, via the induction of iNOS and inhibition of calcium influx through Ca2+ channels in rat aortic smooth muscle.
Collapse
Affiliation(s)
- Chi Ming Wong
- Partner State Key Laboratory of Environmental and Biological Analysis and Department of Chemistry, Hong Kong Baptist University (HKBU), Hong Kong.,HKBU Institute for Research and Continuing Education, Shenzhen, China.,School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Chak Leung Au
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Suk Ying Tsang
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, Chinese University of Hong Kong, Hong Kong
| | - Chi Wai Lau
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Xiaoqiang Yao
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Zongwei Cai
- Partner State Key Laboratory of Environmental and Biological Analysis and Department of Chemistry, Hong Kong Baptist University (HKBU), Hong Kong.,HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Arthur Chi-Kong Chung
- Partner State Key Laboratory of Environmental and Biological Analysis and Department of Chemistry, Hong Kong Baptist University (HKBU), Hong Kong.,HKBU Institute for Research and Continuing Education, Shenzhen, China
| |
Collapse
|
5
|
Bai Y, Zhang Q, Yang Z, Meng Z, Zhao Q. The vasorelaxant mechanisms of methanol on isolated rat aortic rings: Involvement of ion channels and signal transduction pathways. Hum Exp Toxicol 2016; 36:1031-1038. [PMID: 27852936 DOI: 10.1177/0960327116678301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It is reported that methanol is generally used as an industrial solvent, antifreeze, windshield washer fluid, cooking fuel and perfume. Methanol ingestion can lead to severe metabolic disturbances, blindness, or even death. So far, few studies about its negative effects on cardiovascular system have been reported. The purpose of this study was to determine the vasoactive effect of methanol and roles of ion channels and signal transduction pathways on isolated rat aorta. The results suggested that the mechanism of methanol-induced vasorelaxation at low concentrations (<500 mM) was mediated by ATP-sensitive K+ (KATP) and L-type Ca2+ channels, but the mechanism at high concentrations (>600 mM) was related to KATP, voltage-dependent K+, big-conductance Ca2+-activated K+, L-type Ca2+ channels as well as prostacyclin, protein kinase C, β-adrenoceptors pathways. In addition, methanol induced a dose-dependent inhibition of vasoconstrictions caused by calcium chloride, potassium chloride, or norepinephrine. Further work is needed to investigate the relative contribution of each channel and pathway in methanol-induced vasoactive effect.
Collapse
Affiliation(s)
- Y Bai
- 1 Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan, People's Republic of China
| | - Q Zhang
- 1 Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan, People's Republic of China
| | - Z Yang
- 1 Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan, People's Republic of China
| | - Z Meng
- 1 Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan, People's Republic of China
| | - Q Zhao
- 2 State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
6
|
Maekawa H, Serrone JC, Tjahjadi M, Hernesniemi J. RETRACTED ARTICLE: The role of estrogen on the pathology of cerebral aneurysms. Expert Rev Neurother 2016; 16:927-35. [DOI: 10.1080/14737175.2016.1189827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
7
|
Han G, White RE. G-protein-coupled estrogen receptor as a new therapeutic target for treating coronary artery disease. World J Cardiol 2014; 6:367-375. [PMID: 24976908 PMCID: PMC4072826 DOI: 10.4330/wjc.v6.i6.367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 03/06/2014] [Accepted: 04/29/2014] [Indexed: 02/06/2023] Open
Abstract
Coronary heart disease (CHD) continues to be the greatest mortality risk factor in the developed world. Estrogens are recognized to have great therapeutic potential to treat CHD and other cardiovascular diseases; however, a significant array of potentially debilitating side effects continues to limit their use. Moreover, recent clinical trials have indicated that long-term postmenopausal estrogen therapy may actually be detrimental to cardiovascular health. An exciting new development is the finding that the more recently discovered G-protein-coupled estrogen receptor (GPER) is expressed in coronary arteries-both in coronary endothelium and in smooth muscle within the vascular wall. Accumulating evidence indicates that GPER activation dilates coronary arteries and can also inhibit the proliferation and migration of coronary smooth muscle cells. Thus, selective GPER activation has the potential to increase coronary blood flow and possibly limit the debilitating consequences of coronary atherosclerotic disease. This review will highlight what is currently known regarding the impact of GPER activation on coronary arteries and the potential signaling mechanisms stimulated by GPER agonists in these vessels. A thorough understanding of GPER function in coronary arteries may promote the development of new therapies that would help alleviate CHD, while limiting the potentially dangerous side effects of estrogen therapy.
Collapse
|
8
|
Khalil RA. Estrogen, vascular estrogen receptor and hormone therapy in postmenopausal vascular disease. Biochem Pharmacol 2013; 86:1627-42. [PMID: 24099797 DOI: 10.1016/j.bcp.2013.09.024] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/25/2013] [Accepted: 09/25/2013] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) is less common in premenopausal women than men of the same age or postmenopausal women, suggesting vascular benefits of estrogen. Estrogen activates estrogen receptors ERα, ERβ and GPR30 in endothelium and vascular smooth muscle (VSM), which trigger downstream signaling pathways and lead to genomic and non-genomic vascular effects such as vasodilation, decreased VSM contraction and growth and reduced vascular remodeling. However, randomized clinical trials (RCTs), such as the Women's Health Initiative (WHI) and Heart and Estrogen/progestin Replacement Study (HERS), have shown little vascular benefits and even adverse events with menopausal hormone therapy (MHT), likely due to factors related to the MHT used, ER profile, and RCT design. Some MHT forms, dose, combinations or route of administration may have inadequate vascular effects. Age-related changes in ER amount, distribution, integrity and post-ER signaling could alter the vascular response to MHT. The subject's age, preexisting CVD, and hormone environment could also reduce the effects of MHT. Further evaluation of natural and synthetic estrogens, phytoestrogens, and selective estrogen-receptor modulators (SERMs), and the design of appropriate MHT combinations, dose, route and 'timing' could improve the effectiveness of conventional MHT and provide alternative therapies in the peri-menopausal period. Targeting ER using specific ER agonists, localized MHT delivery, and activation of specific post-ER signaling pathways could counter age-related changes in ER. Examination of the hormone environment and conditions associated with hormone imbalance such as polycystic ovary syndrome may reveal the causes of abnormal hormone-receptor interactions. Consideration of these factors in new RCTs such as the Kronos Early Estrogen Prevention Study (KEEPS) could enhance the vascular benefits of estrogen in postmenopausal CVD.
Collapse
Affiliation(s)
- Raouf A Khalil
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Reslan OM, Khalil RA. Vascular effects of estrogenic menopausal hormone therapy. Rev Recent Clin Trials 2012; 7:47-70. [PMID: 21864249 DOI: 10.2174/157488712799363253] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Revised: 07/22/2011] [Accepted: 07/29/2011] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease (CVD) is more common in men and postmenopausal women (Post-MW) than premenopausal women (Pre-MW). Despite recent advances in preventive measures, the incidence of CVD in women has shown a rise that matched the increase in the Post-MW population. The increased incidence of CVD in Post-MW has been related to the decline in estrogen levels, and hence suggested vascular benefits of endogenous estrogen. Experimental studies have identified estrogen receptor ERα, ERβ and a novel estrogen binding membrane protein GPR30 (GPER) in blood vessels of humans and experimental animals. The interaction of estrogen with vascular ERs mediates both genomic and non-genomic effects. Estrogen promotes endothelium-dependent relaxation by increasing nitric oxide, prostacyclin, and hyperpolarizing factor. Estrogen also inhibits the mechanisms of vascular smooth muscle (VSM) contraction including [Ca2+]i, protein kinase C and Rho-kinase. Additional effects of estrogen on the vascular cytoskeleton, extracellular matrix, lipid profile and the vascular inflammatory response have been reported. In addition to the experimental evidence in animal models and vascular cells, initial observational studies in women using menopausal hormonal therapy (MHT) have suggested that estrogen may protect against CVD. However, randomized clinical trials (RCTs) such as the Heart and Estrogen/ progestin Replacement Study (HERS) and the Women's Health Initiative (WHI), which examined the effects of conjugated equine estrogens (CEE) in older women with established CVD (HERS) or without overt CVD (WHI), failed to demonstrate protective vascular effects of estrogen treatment. Despite the initial set-back from the results of MHT RCTs, growing evidence now supports the 'timing hypothesis', which suggests that MHT could increase the risk of CVD if started late after menopause, but may produce beneficial cardiovascular effects in younger women during the perimenopausal period. The choice of an appropriate MHT dose, route of administration, and estrogen/progestin combination could maximize the vascular benefits of MHT and minimize other adverse effects, especially if given within a reasonably short time after menopause to women that seek MHT for the relief of menopausal symptoms.
Collapse
Affiliation(s)
- Ossama M Reslan
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | |
Collapse
|
10
|
Meng Z, Yang Z, Li J, Zhang Q. The vasorelaxant effect and its mechanisms of sodium bisulfite as a sulfur dioxide donor. CHEMOSPHERE 2012; 89:579-584. [PMID: 22763331 DOI: 10.1016/j.chemosphere.2012.05.056] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 04/30/2012] [Accepted: 05/16/2012] [Indexed: 06/01/2023]
Abstract
To study the biological role of bisulfite on vascular contractility and its underlying cellular and molecular mechanisms, to explore whether bisulfite can be used as a sulfur dioxide (SO(2)) donor in the biological experiments, the vasorelaxant effects of sodium bisulfite and sodium sulfite on isolated rat thoracic aortic rings were compared; and the signal transduction pathways and the ion channels involved in the vascular effects of bisulfite were investigated. The results show that: (1) Sodium bisulfite relaxed rat thoracic aortic rings in a concentration-dependent manner (from 100 to 4000 μM); however, sodium sulfite at 500 and 1000 μM caused vasoconstriction, and only at higher concentrations (from 2000 to 4000 μM) it caused vasorelaxation in a concentration-dependent manner. (2) The vasorelaxation caused by the bisulfite at low concentrations (≤500 μM) was endothelium-dependent, but at high concentrations (≥1000 μM) it was endothelium-independent. (3) The vasorelaxation by the bisulfite at the low concentrations was partially mediated by the cGMP pathway and the vasorelaxation was related to big-conductance Ca(2+)-activated K(+) (BK(Ca)) channel, but not due to prostaglandin, protein kinase C (PKC) and cAMP pathways. (4) The vasorelaxation by the bisulfite at high concentrations was partially inhibited by tetraethylammonium (TEA) and glibenclamide, suggesting that the vasorelaxation was related to ATP-sensitive K(+) channel (K(ATP)) and L-type calcium-channel. These results led to the conclusion that bisulfite (HSO(3)(-)) might be a vasoactive factor and sodium bisulfite can be used as a SO(2) donor for the study of SO(2) biology.
Collapse
Affiliation(s)
- Ziqiang Meng
- Institute of Environmental Medicine and Toxicology, Shanxi University, Taiyuan 030006, China.
| | | | | | | |
Collapse
|
11
|
Yang Z, Zhang Y, Meng Z. The vasodilator mechanisms of sodium metabisulfite on precontracted isolated aortic rings in rats: signal transduction pathways and ion channels. Food Chem Toxicol 2012; 50:3114-9. [PMID: 22727858 DOI: 10.1016/j.fct.2012.06.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Revised: 05/30/2012] [Accepted: 06/12/2012] [Indexed: 11/24/2022]
Abstract
Sodium metabisulfite (SMB) is most commonly used as a food additives, however few study was performed on the vasodilator effect of SMB. In the present paper, the vasodilator effects of SMB and roles of Ca(2+) and K(+) channels as well as the cGMP pathway on isolated rat aortic rings were studied. The results show that: (1) SMB could relax isolated aortic rings precontracted by norepinephrine in a concentration-dependent manner. The maximal endothelium-dependent vasorelaxation was approximately 20% whereas that not depending on the presence of the endothelium was more than 90%. (2) The vasorelaxant effects induced by 50 or 200 μM SMB were partially inhibited by iberiotoxin, NS-2028 or l-NNA. The vasorelaxation of 1000 μM SMB was partially inhibited by nifedipine or glibenclamide. The SMB induced vasorelaxation was partially inhibited by tetraethylammonium. These results led to the conclusions that the vasorelaxation of SMB at low concentrations (<400 μM) was endothelium-dependent and mediated by the cGMP pathway and BK(Ca) channel, but at high concentrations (>500 μM) was endothelium-independent and mediated by K(ATP) channel and L-type Ca(2+) channel. The maximal allowable concentration from China and the acceptable daily intake level from WHO of SMB as a food additive should be revised.
Collapse
Affiliation(s)
- Zhenhua Yang
- Institute of Environmental Medicine and Toxicology, Research Center of Environmental Science and Engineering, Shanxi University, Taiyuan 030006, China
| | | | | |
Collapse
|
12
|
Meyer MR, Prossnitz ER, Barton M. GPER/GPR30 and Regulation of Vascular Tone and Blood Pressure. ACTA ACUST UNITED AC 2011; 11:255-261. [PMID: 24999376 DOI: 10.2174/1871522211108040255] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Natural estrogens such as 17β-estradiol are endogenous vasodilators and have been implicated in the gender differences of hypertension. These hormones activate estrogen receptors ERα and ERβ, which mediate part of estrogen-dependent vasodilation. In addition, a novel G protein-coupled estrogen-binding receptor termed GPER/GPR30 has been identified that is expressed in the cardiovascular system. Using knock-out animals or drugs selectively targeting GPER/GPR30, a significant role for this receptor as a mediator of acute estrogen-dependent vasodilation involving nitric oxide (NO) and blood pressure-lowering activity has been demonstrated. The accumulating evidence that GPER/GPR30 is responsible for control of vascular tone indicates that this receptor may represent a novel drug target for pharmacologic treatment of hypertension in postmenopausal women and possibly also men.
Collapse
Affiliation(s)
- Matthias R Meyer
- Molecular Internal Medicine, University of Zurich, Zurich, Switzerland ; Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Eric R Prossnitz
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Matthias Barton
- Molecular Internal Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Meyer MR, Prossnitz ER, Barton M. The G protein-coupled estrogen receptor GPER/GPR30 as a regulator of cardiovascular function. Vascul Pharmacol 2011; 55:17-25. [PMID: 21742056 PMCID: PMC3216677 DOI: 10.1016/j.vph.2011.06.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 06/19/2011] [Accepted: 06/25/2011] [Indexed: 12/29/2022]
Abstract
Endogenous estrogens are important regulators of cardiovascular homeostasis in premenopausal women and delay the development of hypertension and coronary artery disease. These hormones act via three different estrogen receptors affecting both gene transcription and rapid signaling pathways in a complex interplay. In addition to the classical estrogen receptors ERα and ERβ, which are known mediators of estrogen-dependent vascular effects, a G protein-coupled estrogen receptor termed GPER that is expressed in the cardiovascular system has recently been identified. Endogenous human 17β-estradiol, selective estrogen receptor modulators (SERMs) including tamoxifen and raloxifene, and selective estrogen receptor downregulators (SERDs) such as ICI 182,780 are all agonists of GPER, which has been implicated in the regulation of vasomotor tone and protection from myocardial ischemia/reperfusion injury. As a result, understanding the individual role of ERα, ERβ, and GPER in cardiovascular function has become increasingly complex. With accumulating evidence that GPER is responsible for a variety of beneficial cardiovascular effects of estrogens, this receptor may represent a novel target to develop effective strategies for the treatment of cardiovascular diseases by tissue-specific, selective activation of estrogen-dependent molecular pathways devoid of side effects seen with conventional hormone therapy.
Collapse
Affiliation(s)
- Matthias R. Meyer
- Molecular Internal Medicine, University of Zurich, Zurich, Switzerland
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Eric R. Prossnitz
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Matthias Barton
- Molecular Internal Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
Differential vasoactive effects of oestrogen, oestrogen receptor agonists and selective oestrogen receptor modulators in rat middle cerebral artery. Neurosci Res 2011; 71:78-84. [PMID: 21624404 DOI: 10.1016/j.neures.2011.05.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 05/13/2011] [Accepted: 05/16/2011] [Indexed: 12/29/2022]
Abstract
Cerebrovascular disorders are less common in pre-menopausal than post-menopausal women and in females than males. This protection may be due, in part at least, to direct effects of oestrogens on blood vessels. Oestrogen's vasodilatory mechanisms have been reported to be via the endothelium, vascular smooth muscle and extracellular matrix, depending on the vascular bed studied. Herein we investigated the vasoactive effects of oestrogen, oestrogen receptor (ER) and GPR30 agonists and selective ER modulators (SERMs) in the rat middle cerebral artery(MCA), an artery affected in focal ischaemia. MCAs isolated from male Sprague Dawley rats were mounted on a wire myograph. Concentration response curves were constructed to 17β-oestradiol, ERα agonist-PPT, ERβ agonist-DPN, GPR30 agonist-G1 and novel SERMs (LY362321 and LY2120310) in pre-constricted vessels, in the presence and absence of endothelium, blocking agents for nitric oxide synthase (L-NAME), classic ER antagonist (ICI182,780) or plasma membrane specific ERα (ERα-36) antibody. 17β-oestradiol induced rapid vasorelaxation of the MCA which was not affected by endothelium removal, L-NAME or ICI182,780. Vasorelaxation was mimicked by PPT, DPN and G1 but not by the SERMs. Using ERα-36 antibody, effects of oestrogen were partially blocked. PPT had a greater vasorelaxation, while DPN and G1 had a lesser effect than 17β-oestradiol. These findings indicate that activation of plasma membrane bound ERα, β and GPR30 elicits rapid, endothelial-nitric oxide-independent relaxation of the rat MCA.
Collapse
|
15
|
Masood DEN, Roach EC, Beauregard KG, Khalil RA. Impact of sex hormone metabolism on the vascular effects of menopausal hormone therapy in cardiovascular disease. Curr Drug Metab 2011; 11:693-714. [PMID: 21189141 DOI: 10.2174/138920010794233477] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 10/25/2010] [Indexed: 12/24/2022]
Abstract
Epidemiological studies have shown that cardiovascular disease (CVD) is less common in pre-menopausal women (Pre-MW) compared to men of the same age or post-menopausal women (Post-MW), suggesting cardiovascular benefits of estrogen. Estrogen receptors (ERs) have been identified in the vasculature, and experimental studies have demonstrated vasodilator effects of estrogen/ER on the endothelium, vascular smooth muscle (VSM) and extracellular matrix. Several natural and synthetic estrogenic preparations have been developed for relief of menopausal vasomotor symptoms. However, whether menopausal hormone therapy (MHT) is beneficial in postmenopausal CVD remains controversial. Despite reports of vascular benefits of MHT from observational and experimental studies, randomized clinical trials (RCTs), such as the Heart and Estrogen/progestin Replacement Study (HERS) and the Women's Health Initiative (WHI), have suggested that, contrary to expectations, MHT may increase the risk of CVD. These discrepancies could be due to agerelated changes in sex hormone synthesis and metabolism, which would influence the effective dose of MHT and the sex hormone environment in Post-MW. Age-related changes in the vascular ER subtype, structure, expression, distribution, and post-ER signaling pathways in the endothelium and VSM, along with factors related to the design of RCTs, preexisting CVD condition, and structural changes in the blood vessels architecture have also been suggested as possible causes of MHT failure in CVD. Careful examination of these factors should help in identifying the causes of the changes in the vascular effects of estrogen with age. The sex hormone metabolic pathways, the active versus inactive estrogen metabolites, and their effects on vascular function, the mitochondria, the inflammatory process and angiogenesis should be further examined. Also, the genomic and non-genomic effects of estrogenic compounds should be viewed as integrated rather than discrete responses. The complex interactions between these factors highlight the importance of careful design of MHT RCTs, and the need of a more customized approach for each individual patient in order to enhance the vascular benefits of MHT in postmenopausal CVD.
Collapse
Affiliation(s)
- Durr-e-Nayab Masood
- Vascular Surgery Research Laboratory, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
16
|
Xu Z, Wang X, Dai Y, Kong L, Wang F, Xu H, Lu D, Song J, Hou Z. (+/-)-Praeruptorin A enantiomers exert distinct relaxant effects on isolated rat aorta rings dependent on endothelium and nitric oxide synthesis. Chem Biol Interact 2010; 186:239-46. [PMID: 20433815 DOI: 10.1016/j.cbi.2010.04.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 04/16/2010] [Accepted: 04/19/2010] [Indexed: 01/04/2023]
Abstract
Praeruptorin A is a coumarin compound naturally occurring in the roots of Peucedanum praeruptorum Dunn., a commonly used traditional Chinese medicine for the treatment of certain respiratory diseases and hypertension. Although previous studies indicated the relaxant effects of (+/-)-praeruptorin A on tracheal and arterial preparations, little is known about the functional characteristics of the enantiomers. In the present study, the two enantiomers were successfully isolated and identified by using a preparative Daicel Chiralpak AD-H column, and their relaxant effects on aorta rings were observed and compared. (+)-Praeruptorin A showed more potent relaxation than (-)-praeruptorin A against KCl- and phenylephrine-induced contraction of rat isolated aortic rings with intact endothelium. Removal of the endothelium remarkably reduced the relaxant effect of (+)-praeruptorin A but not that of (-)-praeruptorin A. Pretreatment of aortic rings with N(omega)-nitro-L-arginine methyl ester (L-NAME, an inhibitor of nitric oxide synthase) or methylene blue (MB, a soluble guanylyl cyclase inhibitor) resulted in similar changes of the relaxant effects of the two enantiomers to endothelium removal. Molecular docking studies also demonstrated that (+)-praeruptorin A was in more agreement to nitric oxide synthase pharmacophores than (-)-praeruptorin A. On the other hand, the two enantiomers of praeruptorin A could slightly attenuate the contraction of rat aortic rings induced by internal Ca(2+) release from sarcoplasmic reticulum (SR). These findings indicated that (+)-praeruptorin A and (-)-praeruptorin A exerted distinct relaxant effects on isolated rat aorta rings, which might be mainly attributed to nitric oxide synthesis catalyzed by endothelial nitric oxide synthase.
Collapse
Affiliation(s)
- Zhao Xu
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, Jiangsu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Chan YC, Leung FP, Wong WT, Tian XY, Yung LM, Lau CW, Tsang SY, Yao X, Chen ZY, Huang Y. Therapeutically relevant concentrations of raloxifene dilate pressurized rat resistance arteries via calcium-dependent endothelial nitric oxide synthase activation. Arterioscler Thromb Vasc Biol 2010; 30:992-9. [PMID: 20185791 DOI: 10.1161/atvbaha.110.203935] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Selective estrogen receptor modulators (SERMs) inhibit constriction of mammalian conduit arteries. However, it is unknown whether SERMs at therapeutically achievable concentrations could reduce vascular tone in resistance arteries. The present study aimed to examine roles of Ca(2+) influx in endothelium and endothelial nitric oxide synthase (eNOS) activation in dilatations induced by raloxifene, a second-generation SERM in myogenically active arteries. METHODS AND RESULTS Small mesenteric arteries from Sprague-Dawley rats were isolated and mounted in a pressure myograph for measurement of changes in vessel diameter. [Ca(2+)](i) images on native endothelial cells of intact arteries were determined by the fluorescence imaging technique, and phosphorylation of eNOS was assayed by Western blotting. Raloxifene (0.3 to 10 nmol/L) produced dilatations on established steady myogenic constriction. Female rat arteries dilated significantly more in response to raloxifene than male arteries. Raloxifene-induced dilatations of female arteries were blunted by N(G)-nitro-l-arginine methyl ester but unaffected by 1400W, charybdotoxin plus apamin, wortmannin, or LY294002. Raloxifene (3 nmol/L) triggered rises in endothelial cell [Ca(2+)](i) and increased eNOS phosphorylation at Ser1177. Both effects were greater in arteries from female rats than in arteries from male rats. Increases in endothelial cell [Ca(2+)](i) and in eNOS phosphorylation were prevented by removal of extracellular Ca(2+) ions. Finally, ICI 182,780 did not affect the raloxifene-stimulated rise in endothelial cell [Ca(2+)](i), eNOS phosphorylation, and vasodilatations. Chronic raloxifene treatment reduced myogenic constriction in arteries from female but not male rats. CONCLUSION Raloxifene at therapeutically relevant concentrations inhibits myogenic constriction by an NO-dependent mechanism that causally involves the elevated [Ca(2+)](i) in endothelial cells and subsequent eNOS activation. Raloxifene dilates resistance arteries more effectively in female rats, indicating its significant gender-related action on endothelial cells in microcirculation.
Collapse
Affiliation(s)
- Yau Chi Chan
- Institute of Vascular Medicine, Hong Kong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
do Nascimento GRA, Barros YVR, Wells AK, Khalil RA. Research into Specific Modulators of Vascular Sex Hormone Receptors in the Management of Postmenopausal Cardiovascular Disease. Curr Hypertens Rev 2009; 5:283-306. [PMID: 20694192 PMCID: PMC2915874 DOI: 10.2174/157340209789587717] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cardiovascular disease (CVD) is more common in men and postmenopausal women than premenopausal women, suggesting vascular benefits of female sex hormones. Studies on the vasculature have identified estrogen receptors ERα, ERβ and a novel estrogen binding membrane protein GPR30, that mediate genomic and/or non-genomic effects. Estrogen promotes endothelium-dependent relaxation by inducing the production/activity of nitric oxide, prostacyclin, and hyperpolarizing factor, and inhibits the mechanisms of vascular smooth muscle contraction including [Ca(2+)](i), protein kinase C, Rho kinase and mitogen-activated protein kinase. Additional effects of estrogen on the cytoskeleton, matrix metalloproteinases and inflammatory factors contribute to vascular remodeling. However, the experimental evidence did not translate into vascular benefits of menopausal hormone therapy (MHT), and the HERS, HERS-II and WHI clinical trials demonstrated adverse cardiovascular events. The discrepancy has been partly related to delayed MHT and potential changes in the vascular ER amount, integrity, affinity, and downstream signaling pathways due to the subjects' age and preexisting CVD. The adverse vascular effects of MHT also highlighted the need of specific modulators of vascular sex hormone receptors. The effectiveness of MHT can be improved by delineating the differences in phramcokinetics and pharmacodynamics of natural, synthetic, and conjugated equine estrogens. Estriol, "hormone bioidenticals" and phytoestrogens are potential estradiol substitutes. The benefits of low dose MHT, and transdermal or vaginal estrogens over oral preparations are being evaluated. Specific ER modulators (SERMs) and ER agonists are being developed to maximize the effects on vascular ERs. Also, the effects of estrogen are being examined in the context of the whole body hormonal environment and the levels of progesterone and androgens. Thus, the experimental vascular benefits of estrogen can be translated to the outcome of MHT in postmenopausal CVD, as more specific modulators of sex hormone receptors become available and are used at the right dose, route of administration and timing, depending on the subject's age and preexisting cardiovascular condition.
Collapse
|
19
|
Prossnitz ER, Barton M. Signaling, physiological functions and clinical relevance of the G protein-coupled estrogen receptor GPER. Prostaglandins Other Lipid Mediat 2009; 89:89-97. [PMID: 19442754 PMCID: PMC2740807 DOI: 10.1016/j.prostaglandins.2009.05.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 05/01/2009] [Accepted: 05/03/2009] [Indexed: 12/29/2022]
Abstract
GPR30, now named GPER1 (G protein-coupled estrogen receptor1) or GPER here, was first identified as an orphan 7-transmembrane G protein-coupled receptor by multiple laboratories using either homology cloning or differential expression and subsequently shown to be required for estrogen-mediated signaling in certain cancer cells. The actions of estrogen are extensive in the body and are thought to be mediated predominantly by classical nuclear estrogen receptors that act as transcription factors/regulators. Nevertheless, certain aspects of estrogen function remain incompatible with the generally accepted mechanisms of classical estrogen receptor action. Many recent studies have revealed that GPER contributes to some of the actions of estrogen, including rapid signaling events and rapid transcriptional activation. With the introduction of GPER-selective ligands and GPER knockout mice, the functions of GPER are becoming more clearly defined. In many cases, there appears to be a complex interplay between the two receptor systems, suggesting that estrogen-mediated physiological responses may be mediated by either receptor or a combination of both receptor types, with important medical implications.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Cell Biology & Physiology and UNM Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA.
| | | |
Collapse
|
20
|
The vasodilator mechanism of sulfur dioxide on isolated aortic rings of rats: Involvement of the K+ and Ca2+ channels. Eur J Pharmacol 2009; 602:117-23. [DOI: 10.1016/j.ejphar.2008.11.030] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 11/14/2008] [Accepted: 11/17/2008] [Indexed: 11/22/2022]
|
21
|
Wong CM, Yung LM, Leung FP, Tsang SY, Au CL, Chen ZY, Yao X, Cheng CHK, Lau CW, Gollasch M, Huang Y. Raloxifene protects endothelial cell function against oxidative stress. Br J Pharmacol 2008; 155:326-34. [PMID: 18574454 DOI: 10.1038/bjp.2008.262] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE Maintaining a delicate balance between the generation of nitric oxide (NO) and removal of reactive oxygen species (ROS) within the vascular wall is crucial to the physiological regulation of vascular tone. Increased production of ROS reduces the effect and/or bioavailability of NO, leading to an impaired endothelial function. This study tested the hypothesis that raloxifene, a selective oestrogen receptor modulator, can prevent endothelial dysfunction under oxidative stress. EXPERIMENTAL APPROACH Changes in isometric tension were measured in rat aortic rings. The content of cyclic GMP in aortic tissue was determined by radioimmunoassay. Phosphorylation of endothelial NOS (eNOS) and Akt was assayed by Western blot analysis. KEY RESULTS In rings with endothelium, ACh-induced relaxations were attenuated by a ROS-generating reaction (hypoxanthine plus xanthine oxidase, HXXO). The impaired relaxations were ameliorated by acute treatment with raloxifene. HXXO suppressed the ACh-stimulated increase in cyclic GMP levels; this effect was antagonized by raloxifene. The improved endothelial function by raloxifene was abolished by ICI 182,780, and by wortmannin or LY294002. Raloxifene also protected endothelial cell function against H2O2. Raloxifene increased the phosphorylation of eNOS at Ser-1177 and Akt at Ser-473; this effect was blocked by ICI 182,780. Finally, raloxifene was not directly involved in scavenging ROS, and neither inhibited the activity of xanthine oxidase nor stimulated that of superoxide dismutase. CONCLUSION AND IMPLICATIONS Raloxifene is effective against oxidative stress-induced endothelial dysfunction in vitro through an ICI 182,780-sensitive mechanism that involves the increased phosphorylation and activity of Akt and eNOS in rat aortae.
Collapse
Affiliation(s)
- C M Wong
- Institute of Vascular Medicine and Department of Physiology, Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Leung FP, Yung LM, Leung HS, Au CL, Yao X, Vanhoutte PM, Laher I, Huang Y. Therapeutic concentrations of raloxifene augment nitric oxide-dependent coronary artery dilatation in vitro. Br J Pharmacol 2007; 152:223-9. [PMID: 17618301 PMCID: PMC1978259 DOI: 10.1038/sj.bjp.0707387] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND PURPOSE Raloxifene improves cardiovascular function. This study examines the hypothesis that therapeutic concentrations of raloxifene augment endothelium-dependent relaxation via up-regulation of eNOS expression and activity in porcine coronary arteries. EXPERIMENTAL APPROACH Isometric tension was measured in rings from isolated arteries. Intracellular Ca(2+) concentrations ([Ca(2+)](i)) in arterial endothelial cells were detected by Ca(2+) fluorescence imaging. Phosphorylation of eNOS at Ser-1177 was assayed by Western blot analysis. KEY RESULTS In arterial rings pre-contracted with 9,11-dideoxy-11alpha,9alpha-epoxy-methano-prostaglandin F(2alpha) (U46619), treatment with raloxifene (1-3 nM) augmented bradykinin- or substance P-induced relaxation and this effect was antagonized by ICI 182,780, an estrogen receptor antagonist. The enhanced relaxation was abolished in rings treated with inhibitors of nitric oxide/cyclic GMP-dependent dilation, N(G)-nitro-L-arginine methyl ester (L-NAME) plus 1H-[1,2,4]oxadizolo[4,3-a]quinoxalin-1-one (ODQ). In contrast, effects of raloxifene were unaffected after inhibition of endothelium-derived hyperpolarizing factors by charybdotoxin plus apamin. Raloxifene (3 nM) did not influence endothelium-independent relaxation to sodium nitroprusside. 17beta-Estradiol (3-10 nM) also enhanced bradykinin-induced relaxation, which was inhibited by ICI 182,780. Treatment with raloxifene (3 nM) did not affect bradykinin-stimulated rise in endothelial cell [Ca(2+)](i). Raloxifene, 17beta-estradiol, and bradykinin increased eNOS phosphorylation at Ser-1177 and ICI 182,780 prevented effects of raloxifene or 17beta-estradiol but not that of bradykinin. Raloxifene had neither additive nor antagonistic effects on 17beta-estradiol-induced eNOS phosphorylation. CONCLUSIONS AND IMPLICATIONS Raloxifene in therapeutically relevant concentrations augmented endothelial function in porcine coronary arteries in vitro through ICI 182,780-sensitive mechanisms that were associated with increased phosphorylation of eNOS but independent of changes in endothelial cell [Ca(2+)](i).
Collapse
Affiliation(s)
- F P Leung
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong Hong Kong SAR, China
- Department of Physiology, Chinese University of Hong Kong Hong Kong SAR, China
| | - L M Yung
- Department of Physiology, Chinese University of Hong Kong Hong Kong SAR, China
| | - H S Leung
- Department of Physiology, Chinese University of Hong Kong Hong Kong SAR, China
| | - C L Au
- Department of Physiology, Chinese University of Hong Kong Hong Kong SAR, China
| | - X Yao
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong Hong Kong SAR, China
- Department of Physiology, Chinese University of Hong Kong Hong Kong SAR, China
- Institute of Vascular Medicine, Chinese University of Hong Kong Hong Kong SAR, China
| | - P M Vanhoutte
- Department of Pharmacology, University of Hong Kong Hong Kong, China
| | - I Laher
- Department of Pharmacology and Therapeutics, University of British Columbia Vancouver, Canada
| | - Y Huang
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong Hong Kong SAR, China
- Department of Physiology, Chinese University of Hong Kong Hong Kong SAR, China
- Institute of Vascular Medicine, Chinese University of Hong Kong Hong Kong SAR, China
- Author for correspondence:
| |
Collapse
|
23
|
Moritz A, Gust R, Pertz HH. Characterization of the relaxant response to N,N'-dipropyl-1,2-bis(2,6-dichloro-4-hydroxyphenyl)ethylenediamine in porcine coronary arteries. J Pharmacol Exp Ther 2007; 321:699-706. [PMID: 17322023 DOI: 10.1124/jpet.107.120337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
N,N'-Dialkyl-1,2-bis(2,6-dichloro-4-hydroxyphenyl)ethylenediamines show structural analogy with estrogens and selective estrogen receptor modulators. Because the vasodilator properties of these compounds are unknown, we investigated their potential to relax porcine coronary arteries and determined the mechanism(s) of relaxation. Isolated porcine coronary arterial rings were suspended in organ chambers, precontracted with KCl (30 mM), and the relaxant response was determined by measurement of changes in isometric force. Dependent on the chemical structure, the drugs induced concentration-dependent relaxation in rings with and without endothelium. N,N'-Dipropyl-1,2-bis(2,6-dichloro-4-hydroxyphenyl)ethylenediamine (8) was most potent and showed a 12- to 15-fold higher vasodilatory effect than 17beta-estradiol (E2). The vasorelaxation was independent of endothelium. Calcium concentration-dependent contractions in high-potassium depolarizing medium were insurmountably inhibited by 8. The effect of the L-type Ca2+ channel activator (S)-(-)-Bay K 8644 [(S)-(-)-1,4-dihydro-2,6-dimethyl-5-nitro-4-[2-(trifluoromethyl)phenyl]-3-pyridine-carboxylic acid methyl ester], which induced a leftward shift of Ca2+ contraction, was blocked by 8. The relaxant response to 8 was unaffected by the estrogen receptor antagonist ICI 182,780 (7alpha-[9-[(4,4,5,5,5-pentafluoropentyl]-sulfinyl]nonyl]-estra-1,3,5(10)-triene-3,17beta-diol) and K+ channel blockers, i.e., TEA, glibenclamide, and 4-aminopyridine. Furthermore, the vasodilatory effect of 8 was unaffected by the adenylyl cyclase inhibitor SQ 22536 [9-(tetrahydro-2-furanyl)-9H-purin-6-amine], the guanylyl cyclase inhibitor ODQ [1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one], the protein kinase A inhibitor KT 5720 [(9S,10S,12R)-2,3,9,10,11,12-hexahydro-10-hydroxy-9-methyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg: 3',2',1'-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid hexyl ester], the protein kinase G inhibitor KT 5823 [(9S,10R,12R)-2,3,9,10,11,12-hexahydro-10-methoxy-2,9-dimethyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid methyl ester], and the p38 mitogen-activated protein kinase (MAPK) inhibitor SB 203580 [4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)-1H-imidazole]. Western blot analysis demonstrated that 8, unlike E2, raloxifene, and tamoxifen, failed to stimulate p38 MAPK. It is concluded that N,N'-dipropyl-1,2-bis(2,6-dichloro-4-hydroxyphenyl)ethylenediamine induces endothelium-independent relaxation of coronary arteries; the mechanism apparently involves inhibition of L-type Ca2+ channels. The drug may be protective against cardiovascular diseases.
Collapse
Affiliation(s)
- Alkje Moritz
- Institute of Pharmacy, Free University of Berlin, Königin-Luise-Strasse 2 + 4, 14195 Berlin, Germany
| | | | | |
Collapse
|