51
|
Centrella M, McCarthy TL. Estrogen receptor dependent gene expression by osteoblasts - direct, indirect, circumspect, and speculative effects. Steroids 2012; 77:174-84. [PMID: 22093482 DOI: 10.1016/j.steroids.2011.10.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 10/31/2011] [Indexed: 12/15/2022]
Abstract
Hormone activated estrogen receptors (ERs) have long been appreciated as potent mediators of gene expression in female reproductive tissues. These highly targeted responses likely evolved from more elemental roles in lower organisms, in agreement with their widespread effects in the cardiovascular, immunological, central nervous, and skeletal tissue systems. Still, despite intense investigation, the multiple and often perplexing roles of ERs retain significant attention. In the skeleton, this in part derives from apparently opposing effects by ER agonists on bone growth versus bone remodeling, and in younger versus older individuals. The complexity associated with ER activation can also derive from their interactions with other hormone and growth factor systems, and their direct and indirect effects on gene expression. We propose that part of this complexity results from essential interactions between ERs and other transcription factors, each with their own biochemical and molecular intricacies. Solving some of the many questions that persist may help to achieve better, or better directed, use of agents that can drive ER activation in focused and possibly tissue restricted ways.
Collapse
Affiliation(s)
- Michael Centrella
- Department of Surgery, Yale University School of Medicine, 310 Cedar Street, New Haven, CT 06520-8041, United States.
| | | |
Collapse
|
52
|
Hofmeister MV, Damkier HH, Christensen BM, Olde B, Fredrik Leeb-Lundberg LM, Fenton RA, Praetorius HA, Praetorius J. 17β-Estradiol induces nongenomic effects in renal intercalated cells through G protein-coupled estrogen receptor 1. Am J Physiol Renal Physiol 2011; 302:F358-68. [PMID: 21993891 DOI: 10.1152/ajprenal.00343.2011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Steroid hormones such as 17β-estradiol (E2) are known to modulate ion transporter expression in the kidney through classic intracellular receptors. Steroid hormones are also known to cause rapid nongenomic responses in a variety of nonrenal tissues. However, little is known about renal short-term effects of steroid hormones. Here, we studied the acute actions of E2 on intracellular Ca(2+) signaling in isolated distal convoluted tubules (DCT2), connecting tubules (CNT), and initial cortical collecting ducts (iCCD) by fluo 4 fluorometry. Physiological concentrations of E2 induced transient increases in intracellular Ca(2+) concentration ([Ca(2+)](i)) in a subpopulation of cells. The [Ca(2+)](i) increases required extracellular Ca(2+) and were inhibited by Gd(3+). Strikingly, the classic E2 receptor antagonist ICI 182,780 also increased [Ca(2+)](i), which is inconsistent with the activation of classic E2 receptors. G protein-coupled estrogen receptor 1 (GPER1 or GPR30) was detected in microdissected DCT2/CNT/iCCD by RT-PCR. Stimulation with the specific GPER1 agonist G-1 induced similar [Ca(2+)](i) increases as E2, and in tubules from GPER1 knockout mice, E2, G-1, and ICI 182,780 failed to induce [Ca(2+)](i) elevations. The intercalated cells showed both E2-induced concanamycin-sensitive H(+)-ATPase activity by BCECF fluorometry and the E2-mediated [Ca(2+)](i) increment. We propose that E2 via GPER1 evokes [Ca(2+)](i) transients and increases H(+)-ATPase activity in intercalated cells in mouse DCT2/CNT/iCCD.
Collapse
|
53
|
Simard M, Drolet R, Blomquist CH, Tremblay Y. Human type 2 17beta-hydroxysteroid dehydrogenase in umbilical vein and artery endothelial cells: differential inactivation of sex steroids according to the vessel type. Endocrine 2011; 40:203-11. [PMID: 21877158 DOI: 10.1007/s12020-011-9519-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 07/29/2011] [Indexed: 10/17/2022]
Abstract
The human placenta produces high amounts of estradiol. 17β-hydroxysteroid dehydrogenase type 2 (17βHSD2) is expressed by placental endothelial cells and was proposed to regulate sex hormone levels. Previous results obtained in term placenta suggested that 17βHSD2 expression and activity differ among umbilical cord vessels. In this study, 17βHSD2 expression level and enzymatic activity, and estrogen receptor α and β expression levels, were measured in endothelial cell cultures from umbilical arteries (HUAEC) and vein (HUVEC) using real-time quantitative PCR, western blot, and radiolabeled steroids. 17βHSD2-specific activities were also measured in proximal and distal segments of freshly isolated umbilical cord arteries and vein. 17βHSD2 mRNA level and activity were higher in HUAEC than in HUVEC. Activity was higher in umbilical arteries than in the umbilical vein. In arteries, enzymatic activity was higher near the placenta, suggesting a gradient of expression. No difference was found in ERα expression, whereas ERβ was expressed at a higher level in HUAEC than in HUVEC. Expression profiles of estrogen receptors and 17βHSD2 suggest a vessel type-specific response to estrogens. Our data support a differential modulation of biologically active sex steroid levels according to the vessel type in the foeto-placental unit, with apparent higher inactivation in the arterial system.
Collapse
Affiliation(s)
- Marc Simard
- Reproduction, Perinatal Health, and Child Health Axis, CHUQ Research Center, 2705 Laurier Blvd., Rm T-1-49, Québec, QC G1V 4G2, Canada
| | | | | | | |
Collapse
|
54
|
Chen J, Liu L, Hou R, Shao Z, Wu Y, Chen X, Zhou L. Calycosin promotes proliferation of estrogen receptor-positive cells via estrogen receptors and ERK1/2 activation in vitro and in vivo. Cancer Lett 2011; 308:144-51. [DOI: 10.1016/j.canlet.2011.04.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 04/21/2011] [Accepted: 04/28/2011] [Indexed: 12/23/2022]
|
55
|
Hwang YP, Kim HG, Hien TT, Jeong MH, Jeong TC, Jeong HG. Puerarin activates endothelial nitric oxide synthase through estrogen receptor-dependent PI3-kinase and calcium-dependent AMP-activated protein kinase. Toxicol Appl Pharmacol 2011; 257:48-58. [PMID: 21884717 DOI: 10.1016/j.taap.2011.08.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 08/02/2011] [Accepted: 08/15/2011] [Indexed: 01/14/2023]
Abstract
The cardioprotective properties of puerarin, a natural product, have been attributed to the endothelial nitric oxide synthase (eNOS)-mediated production of nitric oxide (NO) in EA.hy926 endothelial cells. However, the mechanism by which puerarin activates eNOS remains unclear. In this study, we sought to identify the intracellular pathways underlying eNOS activation by puerarin. Puerarin induced the activating phosphorylation of eNOS on Ser1177 and the production of NO in EA.hy926 cells. Puerarin-induced eNOS phosphorylation required estrogen receptor (ER)-mediated phosphatidylinositol 3-kinase (PI3K)/Akt signaling and was reversed by AMP-activated protein kinase (AMPK) and calcium/calmodulin-dependent kinase II (CaMKII) inhibition. Importantly, puerarin inhibited the adhesion of tumor necrosis factor (TNF)-α-stimulated monocytes to endothelial cells and suppressed the TNF-α induced expression of intercellular cell adhesion molecule-1. Puerarin also inhibited the TNF-α-induced nuclear factor-κB activation, which was attenuated by pretreatment with N(G)-nitro-L-arginine methyl ester, a NOS inhibitor. These results indicate that puerarin stimulates eNOS phosphorylation and NO production via activation of an estrogen receptor-mediated PI3K/Akt- and CaMKII/AMPK-dependent pathway. Puerarin may be useful for the treatment or prevention of endothelial dysfunction associated with diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Yong Pil Hwang
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | | | | | | | | | | |
Collapse
|
56
|
Wenner MM, Taylor HS, Stachenfeld NS. Endothelin B receptor contribution to peripheral microvascular function in women with polycystic ovary syndrome. J Physiol 2011; 589:4671-9. [PMID: 21825025 DOI: 10.1113/jphysiol.2011.216218] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Endothelin-1 is elevated in women with polycystic ovary syndrome (PCOS), and may play a role in the endothelial dysfunction associated with PCOS. Endothelin-1 binds two receptor subtypes, endothelin A (ET-A) and endothelin B (ET-B). We hypothesized that ET-A mediates vasoconstriction in the cutaneous microvasculature in women with and without PCOS. We further hypothesized that while the ET-B receptors mediate vasodilatation in both groups of women, this response would be blunted in women with PCOS. During local skin warming, we used laser Doppler flowmetry combined with intradermal microdialysis to measure skin blood flow (SkBF) during graded ET-A (BQ-123) and ET-B (BQ-788) antagonist infusions in women with (n = 6) and without (n = 8) PCOS. In both groups, SkBF increased during local heating. The percentage of maximal SkBF-[BQ123] sigmoidal dose-response curve indicated a vasodilatory response as the concentration of the antagonist increased (Hill slope 4.96 ± 4.77, 4.74 ± 5.01; logED(50) 2.53 ± 0.09, 2.49 ± 0.09 nm, for PCOS and Control, respectively). In contrast, the % max SkBF-[BQ788] curve indicated a vasoconstrictive response (Hill slope -4.69 ± 3.85, -4.03 ± 3.85; logED(50), 2.56 ± 0.09, 2.41 ± 0.12 nm, in PCOS and Control). Moreover, the SkBF-[BQ788] curve shifted to the right in women with PCOS, suggesting attenuated ET-B receptor mediated vasodilatation during local skin warming compared to Controls. Thus, the endothelium located ET-B receptors function similarly in women with and without PCOS, although with blunted responsiveness in women with PCOS. Our studies suggest that the lower ET-B receptor responsiveness associated with PCOS may reflect lower endothelial-mediated vasodilatation independent of generally lower vascular reactivity.
Collapse
Affiliation(s)
- Megan M Wenner
- The John B. Pierce Laboratory, and Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, 290 Congress Avenue, New Haven, CT 06519, USA.
| | | | | |
Collapse
|
57
|
He H, Yang F, Wang X, Zeng X, Hu Q, Huang J, Feng J. Complete estrogen receptor blocker ICI182,780 promotes the proliferation of vascular smooth muscle cells. Acta Biochim Biophys Sin (Shanghai) 2011; 43:118-23. [PMID: 21193431 DOI: 10.1093/abbs/gmq119] [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: 11/14/2022] Open
Abstract
ICI182,780 is used in adjuvant therapies of breast cancer. As a complete estrogen receptor (ER) blocker, ICI182,780 may antagonize the effects of estrogen on the cardiovascular system. Estrogen inhibits the proliferation of vascular smooth muscle cells (VSMCs), which is one of the mechanisms that estrogen can exert cardioprotective effects. In the present study, to assess the effects of ICI182,780 on the proliferation of VSMCs, we cultured VSMCs isolated from rat aorta with or without the ER antagonist ICI182,780. The results of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, bromodeoxyuridine incorporation assay, viable cell count, immunochemical staining for proliferating cell nuclear antigen (PCNA), and S-phase ratio determined by flow cytometry revealed a remarkable proliferation of VSMCs after ICI182,780 treatment. ICI182,780 significantly enhanced cell growth in a dose-dependent manner (10(-8)-10(-5) M). Furthermore, the number of PCNA-positive cells and the S-phase progression of VSMCs increased after treatment with ICI182,780. Reverse transcriptase-polymerase chain reaction (RT-PCR) and western blot analysis showed that the mRNA and protein level of cyclin D1 in VSMCs increased under the treatment of ICI182,780. These data suggested that ICI182,780 can promote the growth of VSMCs, which might produce some adverse effects on the cardiovascular system.
Collapse
Affiliation(s)
- Hong He
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University, Jinan, China
| | | | | | | | | | | | | |
Collapse
|
58
|
Tan Z, Zhou LJ, Li Y, Cui YH, Xiang QL, Lin GP, Wang TH. E₂-BSA activates caveolin-1 via PI₃K/ERK1/2 and lysosomal degradation pathway and contributes to EPC proliferation. Int J Cardiol 2011; 158:46-53. [PMID: 21255851 DOI: 10.1016/j.ijcard.2010.12.106] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 11/03/2010] [Accepted: 12/30/2010] [Indexed: 01/18/2023]
Abstract
BACKGROUND The mechanism that estrogen (E(2)) increases the number of endothelial progenitor cells (EPC) is largely unknown. Here we used E(2)-conjugated bovine serum albumin (E(2)-BSA, membrane impermeable) to investigate whether the membrane estrogen receptor (mER) and its related protein caveolin-1 (CAV-1) are involved in these processes. METHODS AND RESULTS E(2)-BSA promoted [(3)H]-thymidine incorporation of EPC through increasing CAV-1 expression via mER (ERα, but not ERβ or GPR30). Both cholesterol depletion and CAV-1 knockdown with use of CAV-1 siRNA significantly attenuated E(2)-BSA-induced [(3)H]-thymidine incorporation. Western blot showed that E(2)-BSA increased membrane CAV-1 protein expression 12h after treatment, whereas mRNA levels of CAV-1 were augmented until 24h after E(2)-BSA treatment. Furthermore, pre-incubated EPC with ICI 182780 (a specific ER antagonist), LY 294002 (a selective PI(3)K inhibitor) or PD 98059 (a specific ERK1/2 inhibitor) before E(2)-BSA inhibited the late-stage effect of E(2)-BSA (≥24 h) on up-regulation of CAV-1 mRNA and protein expression. Pulse chase results demonstrated that E(2)-BSA inhibited lysosome-mediated degradation of CAV-1 protein at the early stage (≤12 h), and then resulted in the increased CAV-1 protein. CONCLUSION In the present work we demonstrated that E(2)-BSA promotes EPC proliferation through mER (ERα) in CAV-1-dependent manner: prolonging the stability of CAV-1 protein through quick inhibition of the lysosomal degradation pathway at the early stage (≤12 h) and up-regulating CAV-1 at transcription levels through PI(3)K/ERK1/2 signaling pathway at the late stage (≥24 h). These data indicated that a there is a novel mechanism of E(2)-BSA in the regulation of EPC proliferation through CAV-1.
Collapse
Affiliation(s)
- Zhi Tan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, PR China.
| | | | | | | | | | | | | |
Collapse
|
59
|
Nanni S, Grasselli A, Benvenuti V, Aiello A, Pantisano V, Re A, Gaetano C, Capogrossi MC, Bacchetti S, Pontecorvi A, Farsetti A. The role of nuclear endothelial nitric oxide synthase in the endothelial and prostate microenvironments. Horm Mol Biol Clin Investig 2011; 5:91-6. [DOI: 10.1515/hmbci.2011.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 11/15/2022]
Abstract
AbstractThis review is based on novel observations from our laboratory on the nuclear translocation and functional role of endothelial nitric oxide synthase (eNOS) in endothelial and prostate cancer (PCa) epithelial cells. Nitric oxide (NO), the product of eNOS, is a free radical involved in the physiology and pathophysiology of living organisms and in a variety of biological processes including the maintenance of vascular homeostasis. Of relevance in this context is the role that estrogens play in the apoptotic process and the migration of endothelial cells through the regulation of target genes such as eNOS itself. It has been shown that both estrogen and NO signaling, mediated respectively by the estrogen receptors (ERs) and eNOS, can strongly counteract endothelial senescence through a common effector, the catalytic subunit of human telomerase. Therefore, this protein has been identified as a key molecule in the aging process which, intriguingly, is considered the only risk factor in the development of PCa and one of the major determinants of cardiovascular diseases. Indeed, in both these contexts we have defined a molecular mechanism involving activation of eNOS and hypoxia-inducible factors in association with ERβ that characterizes the most aggressive form of PCa or influences endothelial cell differentiation. Altogether these data led us to postulate that activation of eNOS is a crucial requirement for the delaying of endothelial senescence as well as for the acquisition of androgen-independence and for tumor progression in the prostate microenvironment.
Collapse
|
60
|
Thomas P, Alyea R, Pang Y, Peyton C, Dong J, Berg AH. Conserved estrogen binding and signaling functions of the G protein-coupled estrogen receptor 1 (GPER) in mammals and fish. Steroids 2010; 75:595-602. [PMID: 19931550 PMCID: PMC2885585 DOI: 10.1016/j.steroids.2009.11.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 11/12/2009] [Indexed: 12/01/2022]
Abstract
Recent studies by several research groups have shown that G protein estrogen receptor-1 (GPER) formerly known as GPR30, mediates 17beta-estradiol (E2) activation of signal transduction pathways in a variety of human cancer cells and displays E2 binding typical of a membrane estrogen receptor. However, the importance of GPER as an estrogen receptor has been questioned by Otto and co-workers. Some of the pitfalls in investigating the functions of recombinant steroid membrane receptors that may explain the negative results of these investigators are discussed. The characteristics of GPER have also been investigated in a teleost fish, Atlantic croaker, where it has been shown to mediate E2 inhibition of oocyte maturation. Investigations on newly discovered homologous proteins from distantly related vertebrate groups are valuable for determining their fundamental, evolutionarily conserved functions. Therefore, the functions of croaker and human GPERs were compared. The comparisons show that croaker and human GPER have very similar estrogen binding characteristics, typical of estrogen membrane receptors, and activate the same estrogen signaling pathways via stimulatory G proteins (Gs) resulting in increased cAMP production. These results suggest that the estrogen binding and estrogen signaling functions of GPER arose early in vertebrate evolution, prior to the divergence of the teleosts from the tetrapods, more than 200 million years ago. The finding that estrogen membrane signaling through GPER has been conserved for such a long period in two distantly related vertebrate groups, mammals and fish, suggests that this is a fundamental function of GPER in vertebrates, and likely its major physiological role.
Collapse
Affiliation(s)
- P Thomas
- University of Texas at Austin Marine Science Institute, 750 Channel View Drive, Port Aransas, TX 78373, USA.
| | | | | | | | | | | |
Collapse
|
61
|
Tang JY, Li S, Li ZH, Zhang ZJ, Hu G, Cheang LCV, Alex D, Hoi MPM, Kwan YW, Chan SW, Leung GPH, Lee SMY. Calycosin promotes angiogenesis involving estrogen receptor and mitogen-activated protein kinase (MAPK) signaling pathway in zebrafish and HUVEC. PLoS One 2010; 5:e11822. [PMID: 20686605 PMCID: PMC2912279 DOI: 10.1371/journal.pone.0011822] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 06/16/2010] [Indexed: 01/01/2023] Open
Abstract
Background Angiogenesis plays an important role in a wide range of physiological processes, and many diseases are associated with the dysregulation of angiogenesis. Radix Astragali is a Chinese medicinal herb commonly used for treating cardiovascular disorders and has been shown to possess angiogenic effect in previous studies but its active constituent and underlying mechanism remain unclear. The present study investigates the angiogenic effects of calycosin, a major isoflavonoid isolated from Radix Astragali, in vitro and in vivo. Methodology Tg(fli1:EGFP) and Tg(fli1:nEGFP) transgenic zebrafish embryos were treated with different concentrations of calycosin (10, 30, 100 µM) from 72 hpf to 96 hpf prior morphological observation and angiogenesis phenotypes assessment. Zebrafish embryos were exposed to calycosin (10, 100 µM) from 72 hpf to 78 hpf before gene-expression analysis. The effects of VEGFR tyrosine kinase inhibitor on calycosin-induced angiogenesis were studied using 72 hpf Tg(fli1:EGFP) and Tg(fli1:nEGFP) zebrafish embryos. The pro-angiogenic effects of calycosin were compared with raloxifene and tamoxifen in 72 hpf Tg(fli1:EGFP) zebrafish embryos. The binding affinities of calycosin to estrogen receptors (ERs) were evaluated by cell-free and cell-based estrogen receptor binding assays. Human umbilical vein endothelial cell cultures (HUVEC) were pretreated with different concentrations of calycosin (3, 10, 30, 100 µM) for 48 h then tested for cell viability and tube formation. The role of MAPK signaling in calycosin-induced angiogenesis was evaluated using western blotting. Conclusion Calycosin was shown to induce angiogenesis in human umbilical vein endothelial cell cultures (HUVEC) in vitro and zebrafish embryos in vivo via the up-regulation of vascular endothelial growth factor (VEGF), VEGFR1 and VEGFR2 mRNA expression. It was demonstrated that calycosin acted similar to other selective estrogen receptor modulators (SERMs), such as raloxifene and tamoxifen, by displaying selective potency and affinity to estrogen receptors ERα and ERβ. Our results further indicated that calycosin promotes angiogenesis via activation of MAPK with the involvement of ERK1/2 and ER. Together, this study revealed, for the first time, that calycosin acts as a selective estrogen receptor modulator (SERM) to promote angiogenesis, at least in part through VEGF-VEGFR2 and MAPK signaling pathways.
Collapse
Affiliation(s)
- Jing Yan Tang
- Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Endothelial-mediated microcirculatory responses to an acute estradiol test are influenced by time since menopause, cumulative hormone exposure, and vasomotor symptoms. Menopause 2010; 17:749-57. [DOI: 10.1097/gme.0b013e3181cde2bd] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
63
|
Kumar S, Lata K, Mukhopadhyay S, Mukherjee TK. Role of estrogen receptors in pro-oxidative and anti-oxidative actions of estrogens: a perspective. Biochim Biophys Acta Gen Subj 2010; 1800:1127-35. [PMID: 20434525 DOI: 10.1016/j.bbagen.2010.04.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 03/23/2010] [Accepted: 04/21/2010] [Indexed: 01/07/2023]
Abstract
BACKGROUND Estrogens are steroid hormones responsible for the primary and secondary sexual characteristics in females. While pre-menopausal women use estrogens as the main constituents of contraceptive pills, post-menopausal women use the same for Hormone Replacement Therapy. Estrogens produce reactive oxygen species by increasing mitochondrial activity and redox cycling of estrogen metabolites. The phenolic hydroxyl group present at the C3 position of the A ring of estrogens can get oxidized either by accepting an electron or by losing a proton. Thus, estrogens might act as pro-oxidant in some settings, resulting in complicated non-communicable diseases, namely, cancer and cardiovascular disorders. However, in some other settings the phenolic hydroxyl group of estrogens may be responsible for the anti-oxidative beneficial functions and thus protect against cardiovascular and neurodegenerative diseases. SCOPE OF REVIEW To date, no single review article has mentioned the implication of estrogen receptors in both the pro-oxidative and anti-oxidative actions of estrogens. MAJOR CONCLUSION The controversial role of estrogens as pro-oxidant or anti-oxidant is largely dependent on cell types, ratio of different types of estrogen receptors present in a particular cell and context specificity of the estrogen hormone responses. Both pro-oxidant and anti-oxidant effects of estrogens might involve different estrogen receptors that can have either genomic or non-genomic action to manifest further hormonal response. GENERAL SIGNIFICANCE This review highlights the role of estrogen receptors in the pro-oxidative and anti-oxidative actions of estrogens with special emphasis on neuronal cells.
Collapse
Affiliation(s)
- Sukhdeep Kumar
- Department of Biology, Indiana Institute of Science Eduaction and Research (IISER), Transit Campus, Chandigarh, India
| | | | | | | |
Collapse
|
64
|
Herrmann JL, Abarbanell AM, Weil BR, Manukyan MC, Poynter JA, Wang Y, Coffey AC, Meldrum DR. Gender dimorphisms in progenitor and stem cell function in cardiovascular disease. J Cardiovasc Transl Res 2010; 3:103-13. [PMID: 20376198 PMCID: PMC2850109 DOI: 10.1007/s12265-009-9149-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Differences in cardiovascular disease outcomes between men and women have long been recognized and attributed, in part, to gender and sex steroids. Gender dimorphisms also exist with respect to the roles of progenitor and stem cells in post-ischemic myocardial and endothelial repair and regeneration. Understanding how these cells are influenced by donor gender and the recipient hormonal milieu may enable researchers to further account for the gender-related disparities in clinical outcomes as well as utilize the beneficial effects of these hormones to optimize transplanted cell function and survival. This review discusses (1) the cardiovascular effects of sex steroids (specifically estradiol and testosterone); (2) the therapeutic potentials of endothelial progenitor cells, mesenchymal stem cells, and embryonic stem cells; and (3) the direct effect of sex steroids on these cell types.
Collapse
Affiliation(s)
- Jeremy L. Herrmann
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Aaron M. Abarbanell
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brent R. Weil
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mariuxi C. Manukyan
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeffrey A. Poynter
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yue Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arthur C. Coffey
- Clarian Cardiovascular Surgery, Methodist Hospital, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Daniel R. Meldrum
- Clarian Cardiovascular Surgery, Methodist Hospital, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN, USA
- 635 Barnhill Drive, Van Nuys Medical Science Bldg Rm. #2017, Indianapolis, IN 46202, USA
| |
Collapse
|
65
|
Townsend EA, Thompson MA, Pabelick CM, Prakash YS. Rapid effects of estrogen on intracellular Ca2+ regulation in human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2010; 298:L521-30. [PMID: 20097735 DOI: 10.1152/ajplung.00287.2009] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The severity of asthma, a disease characterized by airway hyperresponsiveness and inflammation, is enhanced in some women during the menstrual cycle and during pregnancy but relieved in others. These clinical findings suggest that sex steroids modulate airway tone. Based on well-known relaxant effects of estrogens on vascular smooth muscle, we hypothesized that estrogens relax airway smooth muscle (ASM), thus facilitating bronchodilation. In ASM tissues from female patients, Western and immunocytochemical analyses confirmed the presence of both estrogen receptor (ER) isoforms, ERalpha and ERbeta. In fura 2-loaded, dissociated ASM cells maintained in culture, acute exposure to physiological concentrations of 17beta-estradiol (E(2); 100 pM to 10 nM) decreased the intracellular Ca(2+) ([Ca(2+)](i)) response to 1 muM histamine, an effect reversed by the ER antagonist ICI-182,780. The ERalpha-selective agonist (R,R)-THC had a greater reducing effect on [Ca(2+)](i) responses to histamine and 1 muM ACh compared with the ERbeta-selective agonist (DPN). The effects of E(2) on [Ca(2+)](i) were mediated, at least in part, via decreased Ca(2+) influx through l-type channels and store-operated Ca(2+) entry but not via Ca(2+)-activated K(+) channels, receptor-operated entry, or sarcoplasmic reticulum reuptake. Overall, these data support our hypothesis that estrogens relax ASM and suggest a potentially novel therapeutic target in airway hyperresponsiveness.
Collapse
Affiliation(s)
- Elizabeth A Townsend
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | |
Collapse
|
66
|
Chalopin M, Tesse A, Martínez MC, Rognan D, Arnal JF, Andriantsitohaina R. Estrogen receptor alpha as a key target of red wine polyphenols action on the endothelium. PLoS One 2010; 5:e8554. [PMID: 20049322 PMCID: PMC2796721 DOI: 10.1371/journal.pone.0008554] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 12/09/2009] [Indexed: 11/18/2022] Open
Abstract
Background A greater reduction in cardiovascular risk and vascular protection associated with diet rich in polyphenols are generally accepted; however, the molecular targets for polyphenols effects remain unknown. Meanwhile evidences in the literature have enlightened, not only structural similarities between estrogens and polyphenols known as phytoestrogens, but also in their vascular effects. We hypothesized that alpha isoform of estrogen receptor (ERα) could be involved in the transduction of the vascular benefits of polyphenols. Methodology/Principal Findings Here, we used ERα deficient mice to show that endothelium-dependent vasorelaxation induced either by red wine polyphenol extract, Provinols™, or delphinidin, an anthocyanin that possesses similar pharmacological profile, is mediated by ERα. Indeed, Provinols™, delphinidin and ERα agonists, 17-beta-estradiol and PPT, are able to induce endothelial vasodilatation in aorta from ERα Wild-Type but not from Knock-Out mice, by activation of nitric oxide (NO) pathway in endothelial cells. Besides, silencing the effects of ERα completely prevented the effects of Provinols™ and delphinidin to activate NO pathway (Src, ERK 1/2, eNOS, caveolin-1) leading to NO production. Furthermore, direct interaction between delphinidin and ERα activator site is demonstrated using both binding assay and docking. Most interestingly, the ability of short term oral administration of Provinols™ to decrease response to serotonin and to enhance sensitivity of the endothelium-dependent relaxation to acetylcholine, associated with concomitant increased NO production and decreased superoxide anions, was completely blunted in ERα deficient mice. Conclusions/Significance This study provides evidence that red wine polyphenols, especially delphinidin, exert their endothelial benefits via ERα activation. It is a major breakthrough bringing new insights of the potential therapeutic of polyphenols against cardiovascular pathologies.
Collapse
Affiliation(s)
| | - Angela Tesse
- INSERM, U771, CNRS UMR, 6214, Université d'Angers, Angers, France
| | | | - Didier Rognan
- Bioinformatics of the Drug, UMR 7175 CNRS-ULP (Université Louis Pasteur-Strasbourg I), Illkirch, France
| | - Jean-François Arnal
- INSERM U858, Université Toulouse III Paul Sabatier, CHU (Centre Hospitalier Universitaire), Toulouse, France
| | | |
Collapse
|
67
|
Wang Y, Wang Q, Zhao Y, Gong D, Wang D, Li C, Zhao H. Protective Effects of Estrogen Against Reperfusion Arrhythmias Following Severe Myocardial Ischemia in Rats. Circ J 2010; 74:634-43. [DOI: 10.1253/circj.cj-09-0223] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yan Wang
- Laboratory Center, Second Affiliated Hospital of Dalian Medical University
| | - Qi Wang
- Respiratory Department, Second Affiliated Hospital of Dalian Medical University
| | - Yingnan Zhao
- Center for Disease Control of Heilongjiang Province
| | - Dezheng Gong
- Department of Functional Experiment, Dalian Medical University
| | - Dongmei Wang
- Department of Functional Experiment, Dalian Medical University
| | - Cong Li
- Department of Pathophysiology, Dalian Medical University
| | - Henan Zhao
- Department of Pathophysiology, Dalian Medical University
| |
Collapse
|
68
|
Cussons AJ, Watts GF, Stuckey BGA. Dissociation of endothelial function and arterial stiffness in nonobese women with polycystic ovary syndrome (PCOS). Clin Endocrinol (Oxf) 2009; 71:808-14. [PMID: 19508597 DOI: 10.1111/j.1365-2265.2009.03598.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is associated with cardiovascular risk but it is not clear if this is independent of obesity and insulin resistance. This study therefore investigates endothelial function and arterial stiffness in nonobese, noninsulin resistant women with PCOS. DESIGN This is cross-sectional case-control study. PATIENTS A total of 19 young women with PCOS, with body mass index (BMI) <30 kg/m(2), and 19 healthy controls matched for age and BMI were included in the study. MEASUREMENTS Endothelial function was assessed with flow mediated dilatation (FMD) of the brachial artery, while arterial stiffness was assessed with pulse wave velocity (PWV) and augmentation index (AI). RESULTS There were no significant differences between PCOS and control subjects when assessing the following clinical and biochemical variables: blood pressure, homeostasis model assessment insulin-resistance index, lipids and oestradiol. Women with PCOS had higher free androgen index scores (5.14 ± 3.47 vs. 3.25 ± 1.42, P = 0.036). The PCOS subjects had significantly lower FMD of the brachial artery compared with the controls (6.5 ± 2.9%vs. 10.5 ± 4.0%, P < 0.01). There were no significant differences in markers of arterial stiffness (PWV 5.8 ± 1.1 vs. 6.0 ± 1.0, P = 0.58, AI 16.5 ± 10.2 vs. 20.3 ± 10.2, P = 0.25). CONCLUSIONS Women with polycystic ovary syndrome who are young, nonobese, and have no biochemical evidence of insulin resistance, have abnormal vascular function, but normal arterial stiffness, when compared with age and weight matched control subjects. Whether this leads to a greater risk of cardiovascular disease requires further investigation.
Collapse
Affiliation(s)
- Andrea J Cussons
- Keogh Institute for Medical Research, Nedlands, Western Australia.
| | | | | |
Collapse
|
69
|
Pierce JP, Kievits J, Graustein B, Speth RC, Iadecola C, Milner TA. Sex differences in the subcellular distribution of angiotensin type 1 receptors and NADPH oxidase subunits in the dendrites of C1 neurons in the rat rostral ventrolateral medulla. Neuroscience 2009; 163:329-38. [PMID: 19501631 PMCID: PMC2740659 DOI: 10.1016/j.neuroscience.2009.06.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Revised: 06/02/2009] [Accepted: 06/03/2009] [Indexed: 02/07/2023]
Abstract
The rostral ventrolateral medulla (RVLM), a region critical for the tonic and reflex control of arterial pressure, contains a group of adrenergic (C1) neurons that project to the spinal cord and directly modulate pre-ganglionic sympathetic neurons. Epidemiological data suggest that there are gender differences in the regulation of blood pressure. One factor that could be involved is angiotensin II signaling and the associated production of reactive oxygen species (ROS) by NADPH oxidase, which is emerging as an important molecular substrate for central autonomic regulation and dysregulation. In this study dual electron microscopic immunolabeling was used to examine the subcellular distribution of the angiotensin type 1 (AT(1)) receptor and two NADPH oxidase subunits (p47 and p22) in C1 dendritic processes, in tissue from male, proestrus (high estrogen) and diestrus (low estrogen) female rats. Female dendrites displayed significantly more AT(1) labeling and significantly less p47 labeling than males. While elevations in AT(1) labeling primarily resulted from higher levels of receptor on the plasma membrane, p47 labeling was reduced both on the plasma membrane and in the cytoplasm. Across the estrous cycle, proestrus females displayed significantly higher levels of AT(1) labeling than diestrus females, which resulted exclusively from plasma membrane density differences. In contrast, p47 labeling did not change across the estrous cycle, indicating that ROS production might reflect AT(1) receptor membrane density. No significant differences in p22 labeling were observed. These findings demonstrate that both sex and hormonal levels can selectively affect the expression and subcellular distribution of components of the angiotensin II signaling pathway within C1 RVLM neurons. Such effects could reflect differences in the capacity for ROS production, potentially influencing short term excitability and long term gene expression in a cell group which is critically involved in blood pressure regulation, potentially contributing to gender differences in the risk of cardiovascular disease.
Collapse
Affiliation(s)
- J P Pierce
- Division of Neurobiology, Department of Neurology and Neuroscience, Weill Cornell Medical College, 407 East 61st Street, New York, NY 10065, USA.
| | | | | | | | | | | |
Collapse
|
70
|
Gavin KM, Seals DR, Silver AE, Moreau KL. Vascular endothelial estrogen receptor alpha is modulated by estrogen status and related to endothelial function and endothelial nitric oxide synthase in healthy women. J Clin Endocrinol Metab 2009; 94:3513-20. [PMID: 19509105 PMCID: PMC2741709 DOI: 10.1210/jc.2009-0278] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
CONTEXT AND OBJECTIVE Estrogen receptor alpha (ER alpha), a potent transcription factor expressed in vascular endothelial cells, plays a key role in regulating vascular function and health. We determined whether vascular endothelial cell expression of ER alpha is influenced by estrogen status and is related to vascular endothelial function in healthy women. METHODS ER alpha protein expression was measured (quantitative immunofluorescence) in endothelial cells from peripheral veins of 16 healthy, premenopausal women during the early follicular (EF) and late follicular (LF) phases of the menstrual cycle and 17 estrogen-deficient postmenopausal women. Endothelial-dependent dilation (EDD; brachial artery flow-mediated dilation) and endothelial nitric oxide synthase (eNOS) expression and activation were also measured in a subgroup of women. RESULTS In premenopausal women (n = 10), ER alpha expression was 30% lower (P < 0.001) during the EF (low estrogen) compared with the LF (high estrogen) phase of the menstrual cycle. In postmenopausal women, ER alpha expression was 33% lower (P < 0.001) compared with the LF phase of the menstrual cycle in premenopausal women. ER alpha expression was strongly related (r = 0.67; P < 0.001) to EDD, which was reduced in postmenopausal women. ER alpha abundance was positively related to expression of eNOS (r = 0.54; P = 0.009; n = 21) and ser1177 phosphorylated eNOS (r = 0.59; P = 0.006; n = 20). CONCLUSIONS These results provide the first evidence that expression of ER alpha in vascular endothelial cells is modulated by estrogen status and may be a key determinant of vascular endothelial function in healthy pre- and postmenopausal women. ER alpha expression may influence vascular endothelial function in women by affecting protein levels and activation of eNOS.
Collapse
Affiliation(s)
- Kathleen M Gavin
- Department of Integrative Physiology, University of Colorado Denver, Aurora, Colorado 80045, USA
| | | | | | | |
Collapse
|
71
|
The Effects of CD133-Positive Cells to a Nonvascularized Fasciocutaneous Free Graft in the Rat Model. Ann Plast Surg 2009; 63:331-5. [DOI: 10.1097/sap.0b013e3181934951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
72
|
Mao H, Zhang H, Wang H, Wang Y, Zhao F, Hu L, Yanagihara N, Gao X. Dual effects of lipophilic extract of Salvia miltiorrhiza (Danshen) on catecholamine secretion in cultured bovine adrenal medullary cells. JOURNAL OF ETHNOPHARMACOLOGY 2009; 125:59-67. [PMID: 19549564 DOI: 10.1016/j.jep.2009.06.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 06/03/2009] [Accepted: 06/14/2009] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Salvia miltiorrhiza (Danshen) is a well known traditional Chinese herb, which has been used widely in China for the treatment of cardiovascular diseases in clinic. AIM OF THIS STUDY The aim of the present study is to clarify the effects of lipophilic extract of Salvia miltiorrhiza (LESM) on catecholamine (CA) secretion, a traditional Chinese medicine used widely for the treatment of cardiovascular diseases in China. MATERIALS AND METHODS LESM was evaluated for its effects on CA secretion using HPLC-ECD method. The effects of LESM on 22Na+ influx and intracellular calcium ([Ca2+]i) were also investigated. RESULTS Our results showed that LEMS directly stimulated basal CA secretion in an extracellular Ca2+-dependent manner. And the stimulation was not affected by combination of hexamethonium (Hex),an inhibitor of nAChR. LESM also directly elevated [Ca2+]i. In addition, using selective blockers of voltage-dependent Ca2+ channels, such as nitrendipine (for L-type), omega-agatoxin-IVA (for P-type) and -conotoxin-GVIA (for N-type), it was found that nitrendipine suppressed the elevation of [Ca2+]i induced by LESM, but not omega-agatoxin-IVA or omega-conotoxin-GVIA. Compared with acetylcholine (ACh) only, however, combination of LESM with ACh inhibited the raise of CA secretion, 22Na+ influx and [Ca2+]i in a concentration-dependent manner. Furthermore, LESM also inhibited CA secretion induced by veratridine (Ver), and 56 mM K+ at concentrations similar to those for [Ca2+]i rise. One of the lipophilic active compounds, cryptotanshione (Cryp), also had the same effects on CA secretion with LESM. CONCLUSIONS All these findings suggest that LESM exerts dual effects on CA secretion in cultured bovine adrenal medullary cells. LESM exerts antagonistic effects on nAChR, voltage-dependent Na+ and Ca2+ channels, whereas it is an agonist of L-type Ca2+ channel when it used alone.
Collapse
Affiliation(s)
- Haoping Mao
- Tianjin University of Traditional Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 300-193, China
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Sun X, Kumar S, Tian J, Black SM. Estradiol increases guanosine 5'-triphosphate cyclohydrolase expression via the nitric oxide-mediated activation of cyclic adenosine 5'-monophosphate response element binding protein. Endocrinology 2009; 150:3742-52. [PMID: 19389836 PMCID: PMC2717883 DOI: 10.1210/en.2008-1464] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A number of studies have demonstrated that estradiol can stimulate endothelial nitric oxide synthase expression and activity, resulting in enhanced nitric oxide (NO) generation. However, its effect on the NO synthase cofactor, tetrahydrobiopterin are less clear. Cellular tetrahydrobiopterin levels are regulated, at least in part, by GTP cyclohydrolase 1 (GCH1). Thus, the purpose of this study was to determine the effect of estradiol on GCH1 expression and the regulatory mechanisms in pulmonary arterial endothelial cells. Our data indicate that 17beta-estradiol (E2) increases GCH1 transcription in a dose- and time-dependent manner, whereas estrogen receptor antagonism or NO synthase inhibition attenuated E2-stimulated GCH1 expression. Analysis of the GCH1 promoter fragment responsive to E2 revealed the presence of a cAMP response element, and we found that E2 triggers a rapid but transient elevation of phospho-cAMP response element-binding protein (CREB; <1 h) followed by a second sustained rise after 6 h. EMSA analysis revealed an increase in the binding of CREB during E2 treatment and mutation of the cAMP response element in the GCH1 promoter attenuated the E2-mediated increase in transcription. Furthermore, inhibition of the cAMP-dependent kinase, protein kinase A (PKA) completely abolished the E2-stimulated GCH1 promoter activity, whereas the stimulation of cAMP levels with forskolin increased GCH1 promoter activity, indicating the key role of cAMP in regulating GCH1 promoter activity. In conclusion, our results demonstrate that estradiol can modulate GCH1 expression via NO-mediated activation of CREB in pulmonary arterial endothelial cells. These findings provide new insight into the vascular protective effect of estradiol.
Collapse
Affiliation(s)
- Xutong Sun
- Program in Pulmonary Vascular Disease, Vascular Biology Center, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | | | |
Collapse
|
74
|
Miller TW, Isenberg JS, Roberts DD. Molecular regulation of tumor angiogenesis and perfusion via redox signaling. Chem Rev 2009; 109:3099-124. [PMID: 19374334 PMCID: PMC2801866 DOI: 10.1021/cr8005125] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
| | | | - David D. Roberts
- To whom correspondence should be addressed: NIH, Building 10, Room 2A33, 10 Center Dr, MSC1500, Bethesda, Maryland 20892,
| |
Collapse
|
75
|
Han G, Ma H, Chintala R, Fulton DJR, Barman SA, White RE. Essential role of the 90-kilodalton heat shock protein in mediating nongenomic estrogen signaling in coronary artery smooth muscle. J Pharmacol Exp Ther 2009; 329:850-5. [PMID: 19293389 PMCID: PMC2683768 DOI: 10.1124/jpet.108.149112] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 03/16/2009] [Indexed: 12/29/2022] Open
Abstract
Under normal physiological conditions, estrogen is a coronary vasodilator, and this response involves production of NO from endothelial cells. In addition, estrogen also stimulates NO production in coronary artery smooth muscle (CASM); however, the molecular basis for this nongenomic effect of estrogen is unclear. The purpose of this study was to investigate a potential role for the 90-kDa heat shock protein (Hsp90) in estrogen-stimulated neuronal nitric-oxide synthase (nNOS) activity in coronary artery smooth muscle. 17Beta-estradiol produced a concentration-dependent relaxation of endothelium-denuded porcine coronary arteries in vitro, and this response was attenuated by inhibiting Hsp90 function with 1 microM geldanamycin (GA) or 100 microg/ml radicicol (RAD). These inhibitors also prevented estrogen-stimulated NO production in human CASM cells and reversed the stimulatory effect of estrogen on calcium-activated potassium (BK(Ca)) channels. These functional studies indicated a role for Hsp90 in coupling estrogen receptor activation to NOS stimulation in CASM. Furthermore, coimmunoprecipitation studies demonstrated that estrogen stimulates bimolecular interaction of immunoprecipitated nNOS with Hsp90 and that either GA or RAD could inhibit this association. Blocking estrogen receptors with ICI182780 (fulvestrant) also prevented this association. These findings indicate an essential role for Hsp90 in nongenomic estrogen signaling in CASM and further suggest that Hsp90 might represent a prospective therapeutic target to enhance estrogen-stimulated cardiovascular protection.
Collapse
Affiliation(s)
- Guichun Han
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, Georgia, USA.
| | | | | | | | | | | |
Collapse
|
76
|
Abstract
Protective role of estrogens (E2) against cardiovascular disease has been appreciated for many years until the equivocal results of cardiovascular outcomes in clinical trials on hormone replacement therapy were reported. Although new ongoing trials aim to resolve these discrepancies, it is obvious that cardiovascular effects of E(2) are complex and diverse. To understand further the cardiovascular effects of E(2), the detailed knowledge on the specific role of both classical estrogen receptor (ER) subtypes and G protein-coupled receptor-30 in the vasculature are of importance. In this article, we review the current knowledge about the pattern of ERalpha and ERbeta expression in human vasculature, the genomic and non-genomic cardiovascular effects of E(2)versus subtype selective ERalpha and ERbeta stimulation on isolated arteries and in different knockout animal models. The results indicate that although ERalpha and ERbeta are expressed in the endothelium and media of human arteries, there is no definite evidence for predominant expression of one over another, the pattern depends on vascular bed, sex and diseased condition. Data from the experiments on isolated arteries and in ER knockout animal models may indicate that activation of specific ER subtypes could provide additional cardiovascular protective effects. However, a clear role for each ERs have to be finalised with focus on mechanisms and by exploring the potential of ERs-selective agonists for clinical utility.
Collapse
Affiliation(s)
- Leanid Luksha
- Department of Obstetrics and Gynecology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | | |
Collapse
|
77
|
Abstract
Sufficient uteroplacental blood flow is essential for normal pregnancy outcome and is accomplished by the coordinated growth and remodeling of the entire uterine circulation, as well as the creation of a new fetal vascular organ: the placenta. The process of remodeling involves a number of cellular processes, including hyperplasia and hypertrophy, rearrangement of existing elements, and changes in extracellular matrix. In this review, we provide information on uterine blood flow increases during pregnancy, the influence of placentation type on the distribution of uterine vascular resistance, consideration of the patterns, nature, and extent of maternal uterine vascular remodeling during pregnancy, and what is known about the underlying cellular mechanisms.
Collapse
Affiliation(s)
- George Osol
- Department of Obstetrics and Gynecology, University of Vermont College of Medicine, Burlington, Vermont, USA.
| | | |
Collapse
|
78
|
Arnal JF, Gourdy P, Simoncini T. Interference of progestins with endothelial actions of estrogens: a matter of glucocorticoid action or deprivation? Arterioscler Thromb Vasc Biol 2009; 29:441-3. [PMID: 19179642 DOI: 10.1161/atvbaha.108.182337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
79
|
Dong B, Matsumura F. The conversion of rapid TCCD nongenomic signals to persistent inflammatory effects via select protein kinases in MCF10A cells. Mol Endocrinol 2009; 23:549-58. [PMID: 19147701 DOI: 10.1210/me.2008-0317] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Previously we found that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces rapid inflammatory cellular responses in MCF10A mammary epithelial cells through a distinct nongenomic pathway by activating cytosolic phospholipase A2 and Src kinase within 30 min. In the current study we investigated how such an initial, seemingly transient signaling induced by TCDD is subsequently converted into more stable long-term messages. We found that TCDD causes prolonged activation of the binding activity of nuclear proteins to the oligonucleotide probes representing consensus activator protein 1 and CCAAT enhancer binding protein response element sequences, followed by later induction of some diagnostic marker including cyclooxgenase-2, matrix metalloproteinase-2, colony stimulating factor-1, and cytochrome P450 19 (or aromatase). Blocking the early steps of the nongenomic pathway inhibits this action of TCDD. It was also found that Src kinase is mainly responsible for the increase of binding activity to the activator protein 1 probe, and another kinase, protein kinase A (PKA), is accountable for most of the increase of binding activity to the CCAAT enhancer binding protein probe. The induction of those diagnostic markers is also affected by 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (a Src kinase inhibitor) or H89 (a PKA inhibitor). These results indicate that Src kinase and PKA act as the second messengers in propagating the initial nongenomic signaling of TCDD.
Collapse
Affiliation(s)
- Bin Dong
- Department of Environmental Toxicology, University of California, Davis, California 95616, USA
| | | |
Collapse
|
80
|
Ouyang J, Hong H, Shen C, Zhao Y, Ouyang C, Dong L, Zhu J, Guo Z, Zeng K, Chen J, Zhang C, Zhang J. A novel fluorescent probe for the detection of nitric oxide in vitro and in vivo. Free Radic Biol Med 2008; 45:1426-36. [PMID: 18804530 DOI: 10.1016/j.freeradbiomed.2008.08.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 08/07/2008] [Accepted: 08/08/2008] [Indexed: 11/30/2022]
Abstract
Fluorescence imaging of nitric oxide (NO) in vitro and in vivo is essential to developing our understanding of the role of nitric oxide in biology and medicine. Current probes such as diaminofluorescein depend on reactions with oxidized NO products, but not with nitric oxide directly, and this limits their applicability. Here we report the formation of an imaging probe for nitric oxide by coordinating the highly fluorescent chemical 4-methoxy-2-(1H-naphtho[2,3-d]imidazol-2-yl)phenol (MNIP) with Cu(II). The coordination compound MNIP-Cu reacts rapidly and specifically with nitric oxide to generate a product with blue fluorescence that can be used in vitro and in vivo. In the present study MNIP-Cu was used to reveal nitric oxide produced by inducible nitric oxide synthase in lipopolysaccharide (LPS)-activated macrophages (Raw 264.7 cells) and by endothelial nitric oxide synthase in endothelial cells (HUVEC). MNIP-Cu was also used to evaluate the distribution of nitric oxide synthesis in a model of acute liver injury induced by LPS and d-galactosamine in mice. The results demonstrate that MNIP-Cu can act as a novel fluorescent probe for nitric oxide and has many potential applications in biomedical research.
Collapse
Affiliation(s)
- Jie Ouyang
- The School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300191, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|