1
|
Tokiwa H, Ueda K, Takimoto E. The emerging role of estrogen's non-nuclear signaling in the cardiovascular disease. Front Cardiovasc Med 2023; 10:1127340. [PMID: 37123472 PMCID: PMC10130590 DOI: 10.3389/fcvm.2023.1127340] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
Sexual dimorphism exists in the epidemiology of cardiovascular disease (CVD), which indicates the involvement of sexual hormones in the pathophysiology of CVD. In particular, ample evidence has demonstrated estrogen's protective effect on the cardiovascular system. While estrogen receptors, bound to estrogen, act as a transcription factor which regulates gene expressions by binding to the specific DNA sequence, a subpopulation of estrogen receptors localized at the plasma membrane induces activation of intracellular signaling, called "non-nuclear signaling" or "membrane-initiated steroid signaling of estrogen". Although the precise molecular mechanism of non-nuclear signaling as well as its physiological impact was unclear for a long time, recent development of genetically modified animal models and pathway-selective estrogen receptor stimulant bring new insights into this pathway. We review the published experimental studies on non-nuclear signaling of estrogen, and summarize its role in cardiovascular system, especially focusing on: (1) the molecular mechanism of non-nuclear signaling; (2) the design of genetically modified animals and pathway-selective stimulant of estrogen receptor.
Collapse
Affiliation(s)
- Hiroyuki Tokiwa
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazutaka Ueda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
2
|
Adlanmerini M, Fontaine C, Gourdy P, Arnal JF, Lenfant F. Segregation of nuclear and membrane-initiated actions of estrogen receptor using genetically modified animals and pharmacological tools. Mol Cell Endocrinol 2022; 539:111467. [PMID: 34626731 DOI: 10.1016/j.mce.2021.111467] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022]
Abstract
Estrogen receptor alpha (ERα) and beta (ERβ) are members of the nuclear receptor superfamily, playing widespread functions in reproductive and non-reproductive tissues. Beside the canonical function of ERs as nuclear receptors, in this review, we summarize our current understanding of extra-nuclear, membrane-initiated functions of ERs with a specific focus on ERα. Over the last decade, in vivo evidence has accumulated to demonstrate the physiological relevance of this ERα membrane-initiated-signaling from mouse models to selective pharmacological tools. Finally, we discuss the perspectives and future challenges opened by the integration of extra-nuclear ERα signaling in physiology and pathology of estrogens.
Collapse
Affiliation(s)
- Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Jean-François Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France.
| |
Collapse
|
3
|
McMillin SL, Minchew EC, Lowe DA, Spangenburg EE. Skeletal muscle wasting: the estrogen side of sexual dimorphism. Am J Physiol Cell Physiol 2022; 322:C24-C37. [PMID: 34788147 PMCID: PMC8721895 DOI: 10.1152/ajpcell.00333.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The importance of defining sex differences across various biological and physiological mechanisms is more pervasive now than it has been over the past 15-20 years. As the muscle biology field pushes to identify small molecules and interventions to prevent, attenuate, or even reverse muscle wasting, we must consider the effect of sex as a biological variable. It should not be assumed that a therapeutic will affect males and females with equal efficacy or equivalent target affinities under conditions where muscle wasting is observed. With that said, it is not surprising to find that we have an unclear or even a poor understanding of the effects of sex or sex hormones on muscle wasting conditions. Although recent investigations are beginning to establish experimental approaches that will allow investigators to assess the impact of sex-specific hormones on muscle wasting, the field still needs rigorous scientific tools that will allow the community to address critical hypotheses centered around sex hormones. The focus of this review is on female sex hormones, specifically estrogens, and the roles that these hormones and their receptors play in skeletal muscle wasting conditions. With the overall review goal of assembling the current knowledge in the area of sexual dimorphism driven by estrogens with an effort to provide insights to interested physiologists on necessary considerations when trying to assess models for potential sex differences in cellular and molecular mechanisms of muscle wasting.
Collapse
Affiliation(s)
- Shawna L. McMillin
- 1Division of Rehabilitation Science, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota,2Division of Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Everett C. Minchew
- 3Department of Physiology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Dawn A. Lowe
- 1Division of Rehabilitation Science, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota,2Division of Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Espen E. Spangenburg
- 3Department of Physiology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| |
Collapse
|
4
|
Oh TR, Han KD, Choi HS, Kim CS, Bae EH, Ma SK, Kim SW. Hypertension as a risk factor for retinal vein occlusion in menopausal women: A nationwide Korean population-based study. Medicine (Baltimore) 2021; 100:e27628. [PMID: 34713852 PMCID: PMC8556045 DOI: 10.1097/md.0000000000027628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/07/2021] [Indexed: 01/05/2023] Open
Abstract
Retinal vein occlusion (RVO) is an important cause of blindness. Hypertension is a well-known risk factor for RVO. Although the prevalence of hypertension increases in women after menopause, the relationship between blood pressure and RVO in women before and after menopause has not been studied in detail.We retrospectively analyzed 2,619,206 patients from the Korean National Health Insurance System database. A Cox proportional hazard regression model was used to evaluate the independent association between blood pressure and the risk of RVO development and identify differences between premenopausal and postmenopausal women.The incidence of RVO was higher among postmenopausal women than in premenopausal women. In the model adjusted for socioeconomic and clinical variables, there was an association between blood pressure and RVO development in premenopausal and postmenopausal women; however, this was stronger than premenopausal women.Both systolic and diastolic blood pressure are associated with an increased risk of RVO, and their effects are more potent in premenopausal women than postmenopausal women. Thus, comprehensive management of hypertension in premenopausal women is essential to reduce the risk of RVO.
Collapse
Affiliation(s)
- Tae Ryom Oh
- Department of Internal Medicine, Chonnam National University Medical School, and Chonnam National University Hospital, Gwangju, Korea
| | - Kyung-Do Han
- Department of Statistics and Actuarial Science, The Soongsil University of Korea, Seoul, Korea
| | - Hong Sang Choi
- Department of Internal Medicine, Chonnam National University Medical School, and Chonnam National University Hospital, Gwangju, Korea
| | - Chang Seong Kim
- Department of Internal Medicine, Chonnam National University Medical School, and Chonnam National University Hospital, Gwangju, Korea
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School, and Chonnam National University Hospital, Gwangju, Korea
| | - Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, and Chonnam National University Hospital, Gwangju, Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, and Chonnam National University Hospital, Gwangju, Korea
| |
Collapse
|
5
|
Effects of sex and estrous cycle on the brain and plasma arginine metabolic profile in rats. Amino Acids 2021; 53:1441-1454. [PMID: 34245369 DOI: 10.1007/s00726-021-03040-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
L-arginine is a versatile amino acid with a number of bioactive metabolites. Increasing evidence implicates altered arginine metabolism in the aging and neurodegenerative processes. The present study, for the first time, determined the effects of sex and estrous cycle on the brain and blood (plasma) arginine metabolic profile in naïve rats. Female rats displayed significantly lower levels of L-arginine in the frontal cortex and three sub-regions of the hippocampus when compared to male rats. Moreover, female rats had significantly higher levels of L-arginine and γ-aminobutyric acid, but lower levels of L-ornithine, agmatine and putrescine, in plasma relative to male rats. The observed sex difference in brain L-arginine appeared to be independent of the enzymes involved in its metabolism, de novo synthesis and blood-to-brain transport (cationic acid transporter 1 protein expression at least), as well as circulating L-arginine. While the estrous cycle did not affect L-arginine and its metabolites in the brain, there were estrous cycle phase-dependent changes in plasma L-arginine. These findings demonstrate the sex difference in brain L-arginine in the estrous cycle-independent manner. Since peripheral blood has been increasingly used to identify biomarkers of brain pathology, the influences of sex and estrous cycle on blood arginine metabolic profile need attention when experimental research involves female rodents.
Collapse
|
6
|
The Complex Interplay between Endocannabinoid System and the Estrogen System in Central Nervous System and Periphery. Int J Mol Sci 2021; 22:ijms22020972. [PMID: 33478092 PMCID: PMC7835826 DOI: 10.3390/ijms22020972] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
The endocannabinoid system (ECS) is a lipid cell signaling system involved in the physiology and homeostasis of the brain and peripheral tissues. Synaptic plasticity, neuroendocrine functions, reproduction, and immune response among others all require the activity of functional ECS, with the onset of disease in case of ECS impairment. Estrogens, classically considered as female steroid hormones, regulate growth, differentiation, and many other functions in a broad range of target tissues and both sexes through the activation of nuclear and membrane estrogen receptors (ERs), which leads to genomic and non-genomic cell responses. Since ECS function overlaps or integrates with many other cell signaling systems, this review aims at updating the knowledge about the possible crosstalk between ECS and estrogen system (ES) at both central and peripheral level, with focuses on the central nervous system, reproduction, and cancer.
Collapse
|
7
|
Pastore MB, Landeros RV, Chen DB, Magness RR. Structural analysis of estrogen receptors: interaction between estrogen receptors and cav-1 within the caveolae†. Biol Reprod 2020; 100:495-504. [PMID: 30137221 DOI: 10.1093/biolre/ioy188] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/13/2018] [Accepted: 08/20/2018] [Indexed: 01/12/2023] Open
Abstract
Pregnancy is a physiologic state of substantially elevated estrogen biosynthesis that maintains vasodilator production by uterine artery endothelial cells (P-UAECs) and thus uterine perfusion. Estrogen receptors (ER-α and ER-β; ESR1 and ESR2) stimulate nongenomic rapid vasodilatory responses partly through activation of endothelial nitric oxide synthase (eNOS). Rapid estrogenic responses are initiated by the ∼4% ESRs localized to the plasmalemma of endothelial cells. Caveolin-1 (Cav-1) interactions within the caveolae are theorized to influence estrogenic effects mediated by both ESRs. Hypothesis: Both ESR1 and ESR2 display similar spatial partitioning between the plasmalemma and nucleus of UAECs and have similar interactions with Cav-1 at the plasmalemma. Using transmission electron microscopy, we observed numerous caveolae structures in UAECs, while immunogold labeling and subcellular fractionations identified ESR1 and ESR2 in three subcellular locations: membrane, cytosol, and nucleus. Bioinformatics approaches to analyze ESR1 and ESR2 transmembrane domains identified no regions that facilitate ESR interaction with plasmalemma. However, sucrose density centrifugation and Cav-1 immunoisolation columns uniquely demonstrated very high protein-protein association only between ESR1, but not ESR2, with Cav-1. These data demonstrate (1) both ESRs localize to the plasmalemma, cytosol and nucleus; (2) neither ESR1 nor ESR2 contain a classic region that crosses the plasmalemma to facilitate attachment; and (3) ESR1, but not ESR2, can be detected in the caveolar subcellular domain demonstrating ESR1 is the only ESR bound in close proximity to Cav-1 and eNOS within this microdomain. Lack of protein-protein interaction between Cav-1 and ESR2 demonstrates a novel independent association of these proteins at the plasmalemma.
Collapse
Affiliation(s)
- Mayra B Pastore
- Department of Obstetrics and Gynecology Perinatal Research Labs, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Cellular and Molecular Pharmacology, University of California-San Francisco, San Francisco, California, USA
| | - Rosalina Villalon Landeros
- Department of Obstetrics and Gynecology Perinatal Research Labs, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Dong-Bao Chen
- Department of Obstetrics and Gynecology University of California Irvine, Irvine, California, USA
| | - Ronald R Magness
- Department of Obstetrics and Gynecology Perinatal Research Labs, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Obstetrics and Gynecology University of South Florida, Tampa, Florida, USA
| |
Collapse
|
8
|
Filiponi M, Gougoura SG, Befani C, Bargiota Α, Liakos P, Koukoulis GN. 17-β estradiol attenuates the pro-oxidant activity of corticotropin-releasing hormone in macroendothelial cells. Cell Biol Int 2019; 43:1407-1415. [PMID: 31141240 DOI: 10.1002/cbin.11188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 05/25/2019] [Indexed: 01/24/2023]
Abstract
Corticotropin-releasing hormone, which is the predominant regulator of neuroendocrine responses to stress, attenuates inflammation through stimulation of glucocorticoid release. Enhanced corticotropin-releasing hormone expression has been detected in inflammatory cells of the vascular endothelium, where it acts as a local regulator of endothelial redox homeostasis. Estrogens have beneficial effects on endothelial integrity and function, though the mechanism underlying their antioxidative effect remains as yet largely unknown. We therefore investigated the effect of 17β-estradiol on pro-oxidant action of corticotropin-releasing hormone in vitro in macroendothelial cells, and, more specifically, the role of 17β-estradiol on corticotropin-releasing hormone-induced activities/release of the antioxidant enzymes namely, endothelial nitric oxide synthase, superoxide dismutase, catalase, and glutathione. We observed that 17β-estradiol abolished the stimulatory effect of corticotropin-releasing hormone on intracellular reactive oxygen species levels and counteracted its inhibitory effect on endothelial nitric oxide synthase activity and nitric oxide release. In addition, 17β-estradiol significantly induced superoxide dismutase and catalase activity, an effect that was not significantly influenced by corticotropin-releasing hormone. Finally, 17β-estradiol significantly increased glutathione levels and the glutathione/glutathione + glutathione disulfide ratio, an action that was partially blocked by corticotropin-releasing hormone. Our results reveal that 17β-estradiol counterbalances corticotropin-releasing hormone-mediated pro-inflammatory action and thereby maintains the physiological threshold of the endothelial cell redox environment. These observations may be of importance, considering the protective role of estrogen in the development of atherosclerosis.
Collapse
Affiliation(s)
- Maria Filiponi
- Department of Endocrinology and Metabolic Diseases, Research Laboratory, Larissa University Hospital, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Sofia G Gougoura
- Department of Endocrinology and Metabolic Diseases, Research Laboratory, Larissa University Hospital, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Christina Befani
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Αlexandra Bargiota
- Department of Endocrinology and Metabolic Diseases, Research Laboratory, Larissa University Hospital, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Panagiotis Liakos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - George N Koukoulis
- Department of Endocrinology and Metabolic Diseases, Research Laboratory, Larissa University Hospital, Faculty of Medicine, University of Thessaly, Biopolis, 41500, Larissa, Greece
| |
Collapse
|
9
|
Fortini F, Vieceli Dalla Sega F, Caliceti C, Lambertini E, Pannuti A, Peiffer DS, Balla C, Rizzo P. Estrogen-mediated protection against coronary heart disease: The role of the Notch pathway. J Steroid Biochem Mol Biol 2019; 189:87-100. [PMID: 30817989 DOI: 10.1016/j.jsbmb.2019.02.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/05/2019] [Accepted: 02/20/2019] [Indexed: 12/28/2022]
Abstract
Estrogen regulates a plethora of biological processes, under physiological and pathological conditions, by affecting key pathways involved in the regulation of cell proliferation, fate, survival and metabolism. The Notch receptors are mediators of communication between adjacent cells and are key determinants of cell fate during development and in postnatal life. Crosstalk between estrogen and the Notch pathway intervenes in many processes underlying the development and maintenance of the cardiovascular system. The identification of molecular mechanisms underlying the interaction between these types of endocrine and juxtacrine signaling are leading to a deeper understanding of physiological conditions regulated by these steroid hormones and, potentially, to novel therapeutic approaches to prevent pathologies linked to reduced levels of estrogen, such as coronary heart disease, and cardiotoxicity caused by hormone therapy for estrogen-receptor-positive breast cancer.
Collapse
Affiliation(s)
| | | | - Cristiana Caliceti
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Antonio Pannuti
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Daniel S Peiffer
- Oncology Research Institute, Loyola University Chicago: Health Sciences Division, Maywood, Illinois, USA; Department of Microbiology and Immunology, Loyola University Chicago: Health Sciences Division, Maywood, Illinois, USA
| | - Cristina Balla
- Cardiovascular Center, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, RA, Italy; Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy; Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
10
|
Stefano GB, Esch T, Kream RM. Augmentation of Whole-Body Metabolic Status by Mind-Body Training: Synchronous Integration of Tissue- and Organ-Specific Mitochondrial Function. Med Sci Monit Basic Res 2019; 25:8-14. [PMID: 30631032 PMCID: PMC6505060 DOI: 10.12659/msmbr.913264] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The objective of our concise review is to elaborate an evidence-based integrative medicine model that incorporates functional linkages of key aspects of cortically-driven mind-body training procedures to biochemical and molecular processes driving enhanced cellular bioenergetics and whole-body metabolic advantage. This entails the adoption of a unified biological systems approach to selectively elucidate basic biochemical and molecular events responsible for achieving physiological relaxation of complex cellular structures. We provide accumulated evidence in support of the potential synergy of voluntary breathing exercises in combination with meditation and/or complementary cognitive tasks to promote medically beneficial enhancements in whole-body relaxation, anti-stress mechanisms, and restorative sleep. Accordingly, we propose that the widespread metabolic and physiological advantages emanating from a sustained series of complementary mind-body exercises will ultimately engender enhanced functional integration of cortical and limbic areas controlling voluntary respiratory processes with autonomic brainstem neural pattern generators. Finally, a unified mechanism is proposed that links behaviorally-mediated enhancements of whole-body metabolic advantage to optimization of synchronous regulation of mitochondrial oxygen utilization via recycling of nitrite and nitric oxide by iron-sulfur centers of coupled respiratory complexes and nitrite reductases.
Collapse
Affiliation(s)
- George B Stefano
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Prague, Czech Republic
| | - Tobias Esch
- School of Medicine, Faculty of Health, Witten/Herdecke University, Institute for Integrative Health Care, Witten, Germany
| | - Richard M Kream
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Prague, Czech Republic
| |
Collapse
|
11
|
Puglisi R, Mattia G, Carè A, Marano G, Malorni W, Matarrese P. Non-genomic Effects of Estrogen on Cell Homeostasis and Remodeling With Special Focus on Cardiac Ischemia/Reperfusion Injury. Front Endocrinol (Lausanne) 2019; 10:733. [PMID: 31708877 PMCID: PMC6823206 DOI: 10.3389/fendo.2019.00733] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
Abstract
This review takes into consideration the main mechanisms involved in cellular remodeling following an ischemic injury, with special focus on the possible role played by non-genomic estrogen effects. Sex differences have also been considered. In fact, cardiac ischemic events induce damage to different cellular components of the heart, such as cardiomyocytes, vascular cells, endothelial cells, and cardiac fibroblasts. The ability of the cardiovascular system to counteract an ischemic insult is orchestrated by these cell types and is carried out thanks to a number of complex molecular pathways, including genomic (slow) or non-genomic (fast) effects of estrogen. These pathways are probably responsible for differences observed between the two sexes. Literature suggests that male and female hearts, and, more in general, cardiovascular system cells, show significant differences in many parameters under both physiological and pathological conditions. In particular, many experimental studies dealing with sex differences in the cardiovascular system suggest a higher ability of females to respond to environmental insults in comparison with males. For instance, as cells from females are more effective in counteracting the ischemia/reperfusion injury if compared with males, a role for estrogen in this sex disparity has been hypothesized. However, the possible involvement of estrogen-dependent non-genomic effects on the cardiovascular system is still under debate. Further experimental studies, including sex-specific studies, are needed in order to shed further light on this matter.
Collapse
Affiliation(s)
- Rossella Puglisi
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gianfranco Mattia
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Carè
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giuseppe Marano
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Walter Malorni
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
- School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Matarrese
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
- *Correspondence: Paola Matarrese
| |
Collapse
|
12
|
Speth RC, D'Ambra M, Ji H, Sandberg K. A heartfelt message, estrogen replacement therapy: use it or lose it. Am J Physiol Heart Circ Physiol 2018; 315:H1765-H1778. [PMID: 30216118 PMCID: PMC6336974 DOI: 10.1152/ajpheart.00041.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 12/24/2022]
Abstract
The issue of cardiovascular and cognitive health in women is complex. During the premenopausal phase of life, women have healthy blood pressure levels that are lower than those of age-matched men, and they have less cardiovascular disease. However, in the postmenopausal stage of life, blood pressure in women increases, and they are increasingly susceptible to cardiovascular disease, cognitive impairments, and dementia, exceeding the incidence in men. The major difference between pre- and postmenopausal women is the loss of estrogen. Thus, it seemed logical that postmenopausal estrogen replacement therapy, with or without progestin, generally referred to as menopausal hormone treatment (MHT), would prevent these adverse sequelae. However, despite initially promising results, a major randomized clinical trial refuted the benefits of MHT, leading to its falling from favor. However, reappraisal of this study in the framework of a "critical window," or "timing hypothesis," has changed our perspective on the benefit-to-risk ratio of MHT, and this review discusses the historical, current, and future approaches to MHT.
Collapse
Affiliation(s)
- Robert C Speth
- College of Pharmacy, Nova Southeastern University , Fort Lauderdale, Florida
- Department of Pharmacology and Physiology, College of Medicine, Georgetown University , Washington, District of Columbia
| | | | - Hong Ji
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University , Washington, District of Columbia
| | | |
Collapse
|
13
|
Landeros RV, Pastore MB, Magness RR. Effects of the Catechol and Methoxy Metabolites of 17β-Estradiol on Nitric Oxide Production by Ovine Uterine Artery Endothelial Cells. Reprod Sci 2018; 26:459-468. [PMID: 29929429 DOI: 10.1177/1933719118783265] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Nitric oxide (NO) production is essential to facilitate rises in uterine blood flow (UBF) during pregnancy. It has been proposed that the metabolites of E2β, 2-hydroxyestradiol (2-OHE2), 4-hydroxyestradiol (4-OHE2), 2-methoxyestradiol (2-ME2), and 4-methoxyestradiol (4-ME2) play a role in mediating vasodilation and rises in UBF during pregnancy. We previously showed that the E2β metabolites stimulate prostacyclin production in pregnancy-derived ovine uterine artery endothelial cells (P-UAECs); however, it is unknown whether the E2β metabolites also induce NO production. Herein, UAECs derived from nonpregnant and pregnant ewes were used to test the hypothesis that E2β metabolites stimulate NO production in a pregnancy-specific manner. Specific estrogen receptor (ER) and adrenergic receptor (AR) antagonists were used to determine the roles of ERs or ARs in E2β metabolite-induced NO production. E2β and its metabolites increased total nitric oxide metabolites (NOx) levels (NO2 + NO3) in P-UAECs, but not in NP-UAECs. Pretreatment with combined 1 µmol/L 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole dihydrochloride (MPP; ER-α antagonist) and 1 µmol/L 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl]phenol (PHTPP; ER-β antagonist) inhibited the rises in NOx levels stimulated by E2β and 2-ME2, but had no effect on 2-OHE2-, 4-OHE2-, or 4-ME2-stimulated rises in NOx levels. Pretreatment with yohimbine (α2-AR antagonist) and propranolol (β2,3-AR antagonist) inhibited the rises in NOx levels stimulated by 2-OHE2, but not by E2β, 4-OHE2, 2-ME2, or 4-ME2. These data demonstrate that E2β metabolites stimulate NO synthesis via ERs or ARs in UAECs in a pregnancy-specific manner, suggesting that these metabolites contribute to rises in vasodilation and UBF during pregnancy.
Collapse
Affiliation(s)
- Rosalina Villalon Landeros
- 1 Department of Obstetrics and Gynecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI, USA
| | - Mayra B Pastore
- 2 Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Ronald R Magness
- 1 Department of Obstetrics and Gynecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Madison, WI, USA.,3 Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA.,4 Department of Animal Science, University of Wisconsin-Madison, Madison, WI, USA.,5 Department of Obstetrics and Gynecology, Morsani College of Medicine, Perinatal Research Vascular Center, University of South Florida, Tampa, FL, USA
| |
Collapse
|
14
|
Gourdy P, Guillaume M, Fontaine C, Adlanmerini M, Montagner A, Laurell H, Lenfant F, Arnal JF. Estrogen receptor subcellular localization and cardiometabolism. Mol Metab 2018; 15:56-69. [PMID: 29807870 PMCID: PMC6066739 DOI: 10.1016/j.molmet.2018.05.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In addition to their crucial role in reproduction, estrogens are key regulators of energy and glucose homeostasis and they also exert several cardiovascular protective effects. These beneficial actions are mainly mediated by estrogen receptor alpha (ERα), which is widely expressed in metabolic and vascular tissues. As a member of the nuclear receptor superfamily, ERα was primarily considered as a transcription factor that controls gene expression through the activation of its two activation functions (ERαAF-1 and ERαAF-2). However, besides these nuclear actions, a pool of ERα is localized in the vicinity of the plasma membrane, where it mediates rapid signaling effects called membrane-initiated steroid signals (MISS) that have been well described in vitro, especially in endothelial cells. SCOPE OF THE REVIEW This review aims to summarize our current knowledge of the mechanisms of nuclear vs membrane ERα activation that contribute to the cardiometabolic protection conferred by estrogens. Indeed, new transgenic mouse models (affecting either DNA binding, activation functions or membrane localization), together with the use of novel pharmacological tools that electively activate membrane ERα effects recently allowed to begin to unravel the different modes of ERα signaling in vivo. CONCLUSION Altogether, available data demonstrate the prominent role of ERα nuclear effects, and, more specifically, of ERαAF-2, in the preventive effects of estrogens against obesity, diabetes, and atheroma. However, membrane ERα signaling selectively mediates some of the estrogen endothelial/vascular effects (NO release, reendothelialization) and could also contribute to the regulation of energy balance, insulin sensitivity, and glucose metabolism. Such a dissection of ERα biological functions related to its subcellular localization will help to understand the mechanism of action of "old" ER modulators and to design new ones with an optimized benefit/risk profile.
Collapse
Affiliation(s)
- Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France; Service de Diabétologie, Maladies Métaboliques et Nutrition, CHU de Toulouse, Toulouse, France.
| | - Maeva Guillaume
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France; Service d'Hépatologie et Gastro-Entérologie, CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Marine Adlanmerini
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Henrik Laurell
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Françoise Lenfant
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Jean-François Arnal
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| |
Collapse
|
15
|
Abstract
Membrane-initiated steroid signaling (MISS) is a recently discovered aspect of steroidal control over cell function that has proved highly challenging to study due to its rapidity and ultrasensitivity to the steroid trigger [Chow RWY, Handelsman DJ, Ng MKC (2010) Endocrinology 151:2411-2422]. Fundamental aspects underlying MISS, such as receptor binding, kinetics of ion-channel opening, and production of downstream effector molecules remain obscure because a pristine molecular technology that could trigger the release of signaling steroids was not available. We have recently described a prototype DNA nanocapsule which can be programmed to release small molecules upon photoirradiation [Veetil AT, et al. (2017) Nat Nanotechnol 12:1183-1189]. Here we show that this DNA-based molecular technology can now be programmed to chemically trigger MISS, significantly expanding its applicability to systems that are refractory to photoirradiation.
Collapse
|
16
|
Chen CH, Wu SH, Tseng YM, Hou MF, Tsai LY, Tsai SM. Distinct role of endothelial nitric oxide synthase gene polymorphisms from menopausal status in the patients with sporadic breast cancer in Taiwan. Nitric Oxide 2018; 72:1-6. [PMID: 29102546 DOI: 10.1016/j.niox.2017.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/30/2022]
Abstract
Breast cancer has a high incidence in Taiwanese women and worldwide. Previous studies have indicated that NO has multiple independent roles in carcinogenesis; genetic polymorphisms in the endothelial nitric oxide synthase (eNOS) gene could modify its transcription and endogenous NO production. Previous studies have reported conflicting results for the relationship between polymorphisms in the eNOS gene and breast cancer risk. Estrogen levels are associated with eNOS expression; accordingly, variation in estrogen levels may contribute to the discordant results. Therefore, in this study, the effects of eNOS polymorphisms on breast cancer susceptibility were examined in terms of menopausal status in Taiwanese women. Three eNOS polymorphisms (-786T > C, VNTR, and 894G > T) were genotyped in 283 patients with breast cancer (139 premenopausal and 144 postmenopausal) and 200 cancer-free controls (100 premenopausal and 100 postmenopausal) by PCR-RFLP. There was a significantly higher breast cancer risk in premenopausal women carrying 894G > T T than in those with the 894G > T GG genotype; however, postmenopausal women carrying 894G > T T had a lower risk of developing breast cancer. In addition, based on a binary logistic regression analysis, interaction effects of these polymorphisms differed according to menopausal status. The relationship between eNOS polymorphisms and breast cancer hazard depended on menopause status, especially for the 894G > T polymorphism, which may provide an explanation for previous conflicting results.
Collapse
Affiliation(s)
- Chien-Hung Chen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Szu-Hsien Wu
- Division of Plastic Surgery, Department of Surgery, Taipei Veterans General Hospital and Yang Ming University, Taipei, Taiwan
| | - Yang-Ming Tseng
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ming-Feng Hou
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Li-Yu Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Shih-Meng Tsai
- Department of Public Health and Environmental Medicine, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
17
|
Pócs L, Janovszky Á, Garab D, Terhes G, Ocsovszki I, Kaszaki J, Boros M, Piffkó J, Szabó A. Estrogen-dependent efficacy of limb ischemic preconditioning in female rats. J Orthop Res 2018; 36:97-105. [PMID: 28561381 DOI: 10.1002/jor.23621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 05/19/2017] [Indexed: 02/04/2023]
Abstract
Our aim was to examine the effects of ischemic preconditioning (IPC) on the local periosteal and systemic inflammatory consequences of hindlimb ischemia-reperfusion (IR) in Sprague-Dawley rats with chronic estrogen deficiency (13 weeks after ovariectomy, OVX) in the presence and absence of chronic 17beta-estradiol supplementation (E2, 20 µg kg-1 , 5 days/week for 5 weeks); sham-operated (non-OVX) animals served as controls. As assessed by intravital fluorescence microscopy, rolling and the firm adhesion of polymorphonuclear neutrophil leukocytes (PMNs) gave similar results in the Sham + IR and OVX + IR groups in the tibial periosteal microcirculation during the 3-h reperfusion period after a 60-min tourniquet ischemia. Postischemic increases in periosteal PMN adhesion and PMN-derived adhesion molecule CD11b expressions, however, were significantly reduced by IPC (two cycles of 10'/10') in Sham animals, but not in OVX animals; neither plasma free radical levels (as measured by chemiluminescence), nor TNF-alpha release was affected by IPC. E2 supplementation in OVX animals restored the IPC-related microcirculatory integrity and PMN-derived CD11b levels, and TNF-alpha and free radical levels were reduced by IPC only with E2. An enhanced estrogen receptor beta expression could also be demonstrated after E2 in the periosteum. Overall, the beneficial periosteal microcirculatory effects of limb IPC are lost in chronic estrogen deficiency, but they can be restored by E2 supplementation. This suggests that the presence of endogenous estrogen is a necessary facilitating factor of the anti-inflammatory protection provided by limb IPC in females. The IPC-independent effects of E2 on inflammatory reactions should also be taken into account in this model. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:97-105, 2018.
Collapse
Affiliation(s)
- Levente Pócs
- Department of Traumatology and Hand Surgery, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
| | - Ágnes Janovszky
- Department of Oral and Maxillofacial Surgery, University of Szeged, Szeged, Hungary
| | - Dénes Garab
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Gabriella Terhes
- Institute of Clinical Microbiology, University of Szeged, Szeged, Hungary
| | - Imre Ocsovszki
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - József Kaszaki
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Mihály Boros
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - József Piffkó
- Department of Oral and Maxillofacial Surgery, University of Szeged, Szeged, Hungary
| | - Andrea Szabó
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| |
Collapse
|
18
|
Arnal JF, Lenfant F, Metivier R, Flouriot G, Henrion D, Adlanmerini M, Fontaine C, Gourdy P, Chambon P, Katzenellenbogen B, Katzenellenbogen J. Membrane and Nuclear Estrogen Receptor Alpha Actions: From Tissue Specificity to Medical Implications. Physiol Rev 2017; 97:1045-1087. [DOI: 10.1152/physrev.00024.2016] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/19/2016] [Accepted: 01/18/2017] [Indexed: 12/22/2022] Open
Abstract
Estrogen receptor alpha (ERα) has been recognized now for several decades as playing a key role in reproduction and exerting functions in numerous nonreproductive tissues. In this review, we attempt to summarize the in vitro studies that are the basis of our current understanding of the mechanisms of action of ERα as a nuclear receptor and the key roles played by its two activation functions (AFs) in its transcriptional activities. We then depict the consequences of the selective inactivation of these AFs in mouse models, focusing on the prominent roles played by ERα in the reproductive tract and in the vascular system. Evidence has accumulated over the two last decades that ERα is also associated with the plasma membrane and activates non-nuclear signaling from this site. These rapid/nongenomic/membrane-initiated steroid signals (MISS) have been characterized in a variety of cell lines, and in particular in endothelial cells. The development of selective pharmacological tools that specifically activate MISS and the generation of mice expressing an ERα protein impeded for membrane localization have begun to unravel the physiological role of MISS in vivo. Finally, we discuss novel perspectives for the design of tissue-selective ER modulators based on the integration of the physiological and pathophysiological roles of MISS actions of estrogens.
Collapse
Affiliation(s)
- Jean-Francois Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Raphaël Metivier
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Gilles Flouriot
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Daniel Henrion
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Chambon
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Benita Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - John Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| |
Collapse
|
19
|
Ahmed B, Creager MA. Alternative causes of myocardial ischemia in women: An update on spontaneous coronary artery dissection, vasospastic angina and coronary microvascular dysfunction. Vasc Med 2017; 22:146-160. [DOI: 10.1177/1358863x16686410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Bina Ahmed
- Dartmouth-Hitchcock Heart and Vascular Center, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Mark A Creager
- Dartmouth-Hitchcock Heart and Vascular Center, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
20
|
Berkane N, Liere P, Oudinet JP, Hertig A, Lefèvre G, Pluchino N, Schumacher M, Chabbert-Buffet N. From Pregnancy to Preeclampsia: A Key Role for Estrogens. Endocr Rev 2017; 38:123-144. [PMID: 28323944 DOI: 10.1210/er.2016-1065] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 02/28/2017] [Indexed: 02/08/2023]
Abstract
Preeclampsia (PE) results in placental dysfunction and is one of the primary causes of maternal and fetal mortality and morbidity. During pregnancy, estrogen is produced primarily in the placenta by conversion of androgen precursors originating from maternal and fetal adrenal glands. These processes lead to increased plasma estrogen concentrations compared with levels in nonpregnant women. Aberrant production of estrogens could play a key role in PE symptoms because they are exclusively produced by the placenta and they promote angiogenesis and vasodilation. Previous assessments of estrogen synthesis during PE yielded conflicting results, possibly because of the lack of specificity of the assays. However, with the introduction of reliable analytical protocols using liquid chromatography/mass spectrometry or gas chromatography/mass spectrometry, more recent studies suggest a marked decrease in estradiol levels in PE. The aim of this review is to summarize current knowledge of estrogen synthesis, regulation in the placenta, and biological effects during pregnancy and PE. Moreover, this review highlights the links among the occurrence of PE, estrogen biosynthesis, angiogenic factors, and cardiovascular risk factors. A close link between estrogen dysregulation and PE occurrence might validate estrogen levels as a biomarker but could also reveal a potential approach for prevention or cure of PE.
Collapse
Affiliation(s)
- Nadia Berkane
- Department of Gynecology and Obstetrics of University Hospital of Geneva, 1205, Genève, Switzerland.,U1195, INSERM and University Paris Sud, 94276 Kremlin Bicêtre, France
| | - Philippe Liere
- U1195, INSERM and University Paris Sud, 94276 Kremlin Bicêtre, France
| | - Jean-Paul Oudinet
- U1195, INSERM and University Paris Sud, 94276 Kremlin Bicêtre, France
| | - Alexandre Hertig
- Department of Nephrology, Tenon Hospital, APHP, 75020 Paris, France.,University of Pierre and Marie Curie, Sorbonne University, Paris 06, 75005 Paris, France.,Unité Mixte de Recherche Scientifique 1155, F-75020 Paris, France
| | - Guillaume Lefèvre
- University of Pierre and Marie Curie, Sorbonne University, Paris 06, 75005 Paris, France.,Department of Biochemistry and Hormonology, Tenon Hospital, APHP, F-75020 Paris, France
| | - Nicola Pluchino
- Department of Gynecology and Obstetrics of University Hospital of Geneva, 1205, Genève, Switzerland
| | | | - Nathalie Chabbert-Buffet
- University of Pierre and Marie Curie, Sorbonne University, Paris 06, 75005 Paris, France.,Department of Obstetrics, Gynecology and Reproductive Medicine, Tenon Hospital, APHP, F-75020 Paris, France.,INSERM, UMR-S938, Centre de Recherche Saint-Antoine, F-75012 Paris, France
| |
Collapse
|
21
|
Hirose K, Daimon M, Miyazaki S, Chiang SJ, Morimoto-Ichikawa R, Maruyama M, Kawata T, Ohmura H, Daida H. Estrogen variation during the menstrual cycle does not influence left ventricular diastolic function and untwisting rate in premenopausal women. J Cardiol 2017; 69:389-393. [DOI: 10.1016/j.jjcc.2016.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/06/2016] [Accepted: 09/16/2016] [Indexed: 01/31/2023]
|
22
|
Nuclear and Membrane Actions of Estrogen Receptor Alpha: Contribution to the Regulation of Energy and Glucose Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:401-426. [PMID: 29224105 DOI: 10.1007/978-3-319-70178-3_19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Estrogen receptor alpha (ERα) has been demonstrated to play a key role in reproduction but also to exert numerous functions in nonreproductive tissues. Accordingly, ERα is now recognized as a key regulator of energy homeostasis and glucose metabolism and mediates the protective effects of estrogens against obesity and type 2 diabetes. This chapter attempts to summarize our current understanding of the mechanisms of ERα activation and their involvement in the modulation of energy balance and glucose metabolism. We first focus on the experimental studies that constitute the basis of the understanding of ERα as a nuclear receptor and more specifically on the key roles played by its two activation functions (AFs). We depict the consequences of the selective inactivation of these AFs in mouse models, which further underline the prominent role of nuclear ERα in the prevention of obesity and diabetes, as on the reproductive tract and the vascular system. Besides these nuclear actions, a fraction of ERα is associated with the plasma membrane and activates nonnuclear signaling from this site. Such rapid effects, called membrane-initiated steroid signals (MISS), have been characterized in a variety of cell lines and in particular in endothelial cells. The development of selective pharmacological tools that specifically activate MISS as well as the generation of mice expressing an ERα protein impeded for membrane localization has just begun to unravel the physiological role of MISS in vivo and their contribution to ERα-mediated metabolic protection. Finally, we discuss novel perspectives for the design of tissue-selective ER modulators.
Collapse
|
23
|
Kow LM, Pfaff DW. Rapid estrogen actions on ion channels: A survey in search for mechanisms. Steroids 2016; 111:46-53. [PMID: 26939826 PMCID: PMC4929851 DOI: 10.1016/j.steroids.2016.02.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 12/31/2022]
Abstract
A survey of nearly two hundred reports shows that rapid estrogenic actions can be detected across a range of kinds of estrogens, a range of doses, on a wide range of tissue, cell and ion channel types. Striking is the fact that preparations of estrogenic agents that do not permeate the cell membrane almost always mimic the actions of the estrogenic agents that do permeate the membrane. All kinds of estrogens, ranging from natural ones, through receptor modulators, endocrine disruptors, phytoestrogens, agonists, and antagonists to novel G-1 and STX, have been reported to be effective. For actions on specific types of ion channels, the possibility of opposing actions, in different cases, is the rule, not the exception. With this variety there is no single, specific action mechanism for estrogens per se, although in some cases estrogens can act directly or via some signaling pathways to affect ion channels. We infer that estrogens can bind a large number of substrates/receptors at the membrane surface. As against the variety of subsequent routes of action, this initial step of the estrogen's binding action is the key.
Collapse
Affiliation(s)
- Lee-Ming Kow
- The Rockefeller University, New York, NY 10065, USA.
| | | |
Collapse
|
24
|
Kow LM, Pataky S, Dupré C, Phan A, Martin-Alguacil N, Pfaff DW. Analyses of rapid estrogen actions on rat ventromedial hypothalamic neurons. Steroids 2016; 111:100-112. [PMID: 27017919 PMCID: PMC4965276 DOI: 10.1016/j.steroids.2016.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 02/26/2016] [Accepted: 03/11/2016] [Indexed: 10/22/2022]
Abstract
Rapid estrogen actions are widely diverse across many cell types. We conducted a series of electrophysiological studies on single rat hypothalamic neurons and found that estradiol (E2) could rapidly and independently potentiate neuronal excitation/depolarizations induced by histamine (HA) and N-Methyl-d-Aspartate (NMDA). Now, the present whole-cell patch study was designed to determine whether E2 potentiates HA and NMDA depolarizations - mediated by distinctly different types of receptors - by the same or by different mechanisms. For this, the actions of HA, NMDA, as well as E2, were investigated first using various ion channel blockers and then by analyzing and comparing their channel activating characteristics. Results indicate that: first, both HA and NMDA depolarize neurons by inhibiting K(+) currents. Second, E2 potentiates both HA and NMDA depolarizations by enhancing the inhibition of K(+) currents, an inhibition caused by the two transmitters. Third, E2 employs the very same mechanism, the enhancement of K(+) current inhibition, thus to rapidly potentiate HA and NMDA depolarizations. These data are of behavioral importance, since the rapid E2 potentiation of depolarization synergizes with nuclear genomic actions of E2 to facilitate lordosis behavior, the primary female-typical reproductive behavior.
Collapse
Affiliation(s)
- Lee-Ming Kow
- Laboratory of Neurobiology & Behavior, The Rockefeller University, New York, NY 10065, USA.
| | - Stefan Pataky
- Laboratory of Neurobiology & Behavior, The Rockefeller University, New York, NY 10065, USA.
| | - Christophe Dupré
- Laboratory of Neurobiology & Behavior, The Rockefeller University, New York, NY 10065, USA.
| | - Anna Phan
- Laboratory of Neurobiology & Behavior, The Rockefeller University, New York, NY 10065, USA.
| | - Nieves Martin-Alguacil
- Laboratory of Neurobiology & Behavior, The Rockefeller University, New York, NY 10065, USA.
| | - Donald W Pfaff
- Laboratory of Neurobiology & Behavior, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
25
|
Menazza S, Murphy E. The Expanding Complexity of Estrogen Receptor Signaling in the Cardiovascular System. Circ Res 2016; 118:994-1007. [PMID: 26838792 DOI: 10.1161/circresaha.115.305376] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/28/2015] [Indexed: 12/21/2022]
Abstract
Estrogen has important effects on cardiovascular function including regulation of vascular function, blood pressure, endothelial relaxation, and the development of hypertrophy and cardioprotection. However, the mechanisms by which estrogen mediates these effects are still poorly understood. As detailed in this review, estrogen can regulate transcription by binding to 2 nuclear receptors, ERα and ERβ, which differentially regulate gene transcription. ERα and ERβ regulation of gene transcription is further modulated by tissue-specific coactivators and corepressors. Estrogen can bind to ERα and ERβ localized at the plasma membrane as well as G-protein-coupled estrogen receptor to initiate membrane delimited signaling, which enhances kinase signaling pathways that can have acute and long-term effects. The kinase signaling pathways can also mediate transcriptional changes and can synergize with the ER to regulate cell function. This review will summarize the beneficial effects of estrogen in protecting the cardiovascular system through ER-dependent mechanisms with an emphasis on the role of the recently described ER membrane signaling mechanisms.
Collapse
Affiliation(s)
- Sara Menazza
- From the Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD.
| | - Elizabeth Murphy
- From the Systems Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
26
|
Abstract
Estrogen receptors (ER) classically function as transcription factors regulating gene expression. More recently, evidence has continued to accumulate that ER additionally serve numerous important functions remote from the nucleus in a variety of cell types, particularly in non-reproductive tissues. The identification of post-translational modifications of ERα and protein-protein interactions with the receptor that are critical to its non-nuclear functions has afforded opportunities to gain greater insights into these novel non-genomic roles of the receptor. The development of a stable ligand that selectively activates non-nuclear ER has also been invaluable. In this review focused on ERα, recent new understanding of the processes underlying non-nuclear ER action and their in vivo consequences will be discussed. Further research into the non-nuclear capacities by which ER modulate cellular behavior is essential to ultimately harnessing these processes for therapeutic gain in numerous disease contexts.
Collapse
Affiliation(s)
- Subhashis Banerjee
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| | - Ken L Chambliss
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| | - Chieko Mineo
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| | - Philip W Shaul
- Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA.
| |
Collapse
|
27
|
Caliceti C, Aquila G, Pannella M, Morelli MB, Fortini C, Pinton P, Bonora M, Hrelia S, Pannuti A, Miele L, Rizzo P, Ferrari R. 17β-estradiol enhances signalling mediated by VEGF-A-delta-like ligand 4-notch1 axis in human endothelial cells. PLoS One 2013; 8:e71440. [PMID: 23967210 PMCID: PMC3742772 DOI: 10.1371/journal.pone.0071440] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 06/30/2013] [Indexed: 01/22/2023] Open
Abstract
Estrogens play a protective role in coronary artery disease. The mechanisms of action are still poorly understood, although a role for estrogens in stimulation of angiogenesis has been suggested. In several cell types, estrogens modulate the Notch pathway, which is involved in controlling angiogenesis downstream of vascular endothelial growth factor A (VEGF-A). The goal of our study was to establish whether estrogens modulate Notch activity in endothelial cells and the possible consequences on angiogenesis. Human umbilical vein endothelial cells (HUVECs) were treated with 17β-estradiol (E2) and the effects on Notch signalling were evaluated. E2 increased Notch1 processing as indicated by i) decreased levels of Notch1 transmembrane subunit ii) increased amount of Notch1 in nuclei iii) unaffected level of mRNA. Similarly, E2 increased the levels of the active form of Notch4 without altering Notch4 mRNA. Conversely, protein and mRNA levels of Notch2 were both reduced suggesting transcriptional repression of Notch2 by E2. Under conditions where Notch was activated by upregulation of Delta-like ligand 4 (Dll4) following VEGF-A treatment, E2 caused a further increase of the active form of Notch1, of the number of cells with nuclear Notch1 and of Hey2 mRNA. Estrogen receptor antagonist ICI 182.780 antagonized these effects suggesting that E2 modulation of Notch1 is mediated by estrogen receptors. E2 treatment abolished the increase in endothelial cells sprouting caused by Notch inhibition in a tube formation assay on 3D Matrigel and in mouse aortic ring explants. In conclusion, E2 affects several Notch pathway components in HUVECs, leading to an activation of the VEGF-A-Dll4-Notch1 axis and to a modulation of vascular branching when Notch signalling is inhibited. These results contribute to our understanding of the molecular mechanisms of cardiovascular protection exerted by estrogens by uncovering a novel role of E2 in the Notch signalling-mediated modulation of angiogenesis.
Collapse
Affiliation(s)
- Cristiana Caliceti
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Giorgio Aquila
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Micaela Pannella
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Marco Bruno Morelli
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Cinzia Fortini
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Surgeon, Morphology and Experimental Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Massimo Bonora
- Department of Surgeon, Morphology and Experimental Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Antonio Pannuti
- University of Mississippi, Medical Center Cancer Institute, Jackson, Mississippi, United States of America
| | - Lucio Miele
- University of Mississippi, Medical Center Cancer Institute, Jackson, Mississippi, United States of America
| | - Paola Rizzo
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Department of Medical Sciences, Cardiovascular Section, Azienda Ospedaliero-Universitaria, Arcispedale Sant’Anna, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| |
Collapse
|
28
|
Ueda K, Karas RH. Emerging evidence of the importance of rapid, non-nuclear estrogen receptor signaling in the cardiovascular system. Steroids 2013; 78:589-96. [PMID: 23276634 DOI: 10.1016/j.steroids.2012.12.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 12/07/2012] [Accepted: 12/13/2012] [Indexed: 11/20/2022]
Abstract
Estrogen receptors are classically known as ligand-activated transcription factors that regulate gene transcription in cells in response to hormone binding. In addition to this "genomic" signaling pathway, a "rapid, non-nuclear" signaling pathway mediated by cell membrane-associated estrogen receptors also has been recognized. Although for many years there was little evidence to support any physiological relevance of rapid-signaling, very recently evidence has been accumulating supporting the importance of the rapid, non-nuclear signaling as potentially critical for the protective effects of estrogen in the cardiovascular system. Better understanding of the rapid, non-nuclear signaling potentially provides an opportunity to design "pathway-specific" selective estrogen receptor modulators capable of differentially regulating non-nuclear vs. genomic effects that may prove useful ultimately as specific therapies for cardiovascular diseases.
Collapse
Affiliation(s)
- Kazutaka Ueda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | | |
Collapse
|
29
|
Roney JR, Simmons ZL. Hormonal predictors of sexual motivation in natural menstrual cycles. Horm Behav 2013; 63:636-45. [PMID: 23601091 DOI: 10.1016/j.yhbeh.2013.02.013] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/15/2013] [Accepted: 02/20/2013] [Indexed: 11/23/2022]
Abstract
Little is known regarding which hormonal signals may best predict within- and between-women variance in sexual motivation among naturally cycling women. To address this, we collected daily saliva samples across 1-2 menstrual cycles from a sample of young women; assayed samples for estradiol, progesterone, and testosterone; and also collected daily diary reports of women's sexual behavior and subjective sexual desire. With respect to within-cycle, day-to-day fluctuations in subjective desire, we found evidence for positive effects of estradiol and negative effects of progesterone. Desire exhibited a mid-cycle peak, similar to previous findings; measured progesterone concentrations statistically mediated the fall in desire from mid-cycle to the luteal phase, but no combination of hormone measures substantially mediated the follicular phase rise in desire, which suggests that other signals may be implicated in this effect. Hormonal predictors of within-cycle fluctuations in sexual behavior generally reached only trend levels of statistical significance, though the patterns again suggested positive effects of estradiol and negative effects of progesterone. Between-women and within-women, between-cycle effects of hormone concentrations were generally absent, although statistical power was more limited at these higher levels of analysis. There were no significant effects of testosterone concentrations when controlling for the effects of estradiol and progesterone, which raises questions regarding the importance of this hormone for the regulation of sexual motivation in natural cycles. Our study is among the first to identify hormonal predictors of within-cycle fluctuations in sexual motivation, and thus adds novel evidence regarding the endocrine correlates of human sexuality.
Collapse
Affiliation(s)
- James R Roney
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA 93106-9660, USA.
| | | |
Collapse
|
30
|
Heydarpour P, Salehi-Sadaghiani M, Javadi-Paydar M, Rahimian R, Fakhfouri G, Khosravi M, Khoshkish S, Gharedaghi MH, Ghasemi M, Dehpour AR. Estradiol reduces depressive-like behavior through inhibiting nitric oxide/cyclic GMP pathway in ovariectomized mice. Horm Behav 2013; 63:361-369. [PMID: 23262264 DOI: 10.1016/j.yhbeh.2012.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 12/05/2012] [Accepted: 12/07/2012] [Indexed: 12/26/2022]
Abstract
Estradiol decline has been associated with depressive-like behavior in female mice and NO has been suggested to play a major role in the pathogenesis of major depression. This study was conducted to investigate the antidepressant-like effects of acute estradiol administration in female ovariectomized (OVX) mice and the possible role of nitric oxide (NO)/cyclic GMP (cGMP) pathway. To this end, bilateral ovariectomy was performed in female mice and different doses of estradiol were injected alone or in combination with non-specific NO synthase (NOS) inhibitor (L-NAME), selective neural NOS (nNOS) inhibitor (7-NI), an NO precursor (L-arginine) or selective phosphodiesterase type 5 inhibitor (sildenafil). The duration of immobility was recorded in the forced swimming test (FST) to assess the depressive behavior. Moreover, hippocampal levels of NO were determined in select groups. 10 days following the procedure, OVX mice showed significantly prolonged immobility time in comparison with the sham group. Estradiol (3, 10, and 30 μg/kg, s.c.), when injected 1 h prior to FST, exerted antidepressant-like effects in OVX mice. Both L-NAME (30 mg/kg, i.p.), and 7-NI (50 mg/kg, i.p.) significantly reduced the immobility times of OVX mice. Administration of a sub-effective dose of L-NAME (10mg/kg), 15 min after a sub-effective dose of estradiol (1 μg/kg, s.c.) had a robust antidepressant-like effect in OVX mice. Also a sub-effective dose of 7-NI (25 mg/kg), 30 min after a sub-effective dose of estradiol (1 μg/kg, s.c.) showed antidepressant-like effect in OVX mice. Both the NO precursor L-arginine (750 mg/kg, i.p.) and the cGMP-specific phosphodiesterase type 5 inhibitor sildenafil (5 mg/kg, i.p.), 30 min before estradiol treatment, prevented the antidepressant-like effect of a potent dose of estradiol (10 μg/kg, s.c.) in OVX mice. The present findings suggest that suppression of the NO synthase/NO/cGMP pathway may be involved in the antidepressant-like effects of estradiol in OVX mice.
Collapse
Affiliation(s)
- Pouria Heydarpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Sadeghian R, Fereidoni M, Soukhtanloo M, Azizi-Malekabadi H, Hosseini M. Decreased nitric oxide levels in the hippocampus may play a role in learning and memory deficits in ovariectomized rats treated by a high dose of estradiol. ARQUIVOS DE NEURO-PSIQUIATRIA 2012; 70:874-879. [PMID: 23175201 DOI: 10.1590/s0004-282x2012001100010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 07/18/2012] [Indexed: 12/15/2022]
Abstract
The effects of a high estradiol dose on memory and on nitric oxide metabolites in hippocampal tissues were investigated. Sham-Est and OVX-Est Groups were treated with 4 mg/kg of estradiol valerate for 12 weeks. Time latency and path length were significantly higher in the Sham-Est and OVX-Est Groups than in the Sham and OVX Groups, respectively (p<0.001). The animals in the Sham-Est and OVX-Est Groups spent lower time in the target quadrant (Q1) than those of the Sham and OVX Groups during the probe trial test (p<0.05 and <0.001, respectively). Significantly lower nitric oxide metabolite levels in the hippocampi of the Sham-Est and OVX-Est Groups were observed than in the Sham and OVX ones (p<0.001). These results suggest that decreased nitric oxide levels in the hippocampus may play a role in the learning and memory deficits observed after treatment with a high dose of estradiol, although the precise underlying mechanisms remain to be elucidated.
Collapse
Affiliation(s)
- Reihaneh Sadeghian
- Department of Biology, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | | | | | | |
Collapse
|
32
|
Azizi-Malekabadi H, Hosseini M, Soukhtanloo M, Sadeghian R, Fereidoni M, Khodabandehloo F. Different effects of scopolamine on learning, memory, and nitric oxide metabolite levels in hippocampal tissues of ovariectomized and Sham-operated rats. ARQUIVOS DE NEURO-PSIQUIATRIA 2012; 70:447-452. [PMID: 22699543 DOI: 10.1590/s0004-282x2012000600012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Accepted: 02/07/2012] [Indexed: 11/21/2022]
Abstract
Different effects of scopolamine on learning, memory, and nitric oxide (NO) metabolites in hippocampal tissues of ovariectomized (OVX) and sham-operated rats were investigated. The animals in the Sham-Scopolamine (Sham-Sco) and OVX-Scopolamine (OVX-Sco) Groups were treated with 2 mg/kg scopolamine before undergoing the Morris water maze, while the animals in the Sham and OVX Groups received saline. The time latency and path length were significantly higher in both the Sham-Sco and the OVX-Sco Groups, in comparison with the Sham and OVX Groups, respectively (p<0.001). Significantly lower NO metabolite levels in the hippocampi of the Sham-Sco Group were observed, compared with the Sham Group (p<0.001), while there was no significant difference between the OVX-Sco and OVX Groups. The decreased NO level in the hippocampus may play a role in the learning and memory deficits induced by scopolamine. However, it seems that the effect of scopolamine on hippocampal NO differs between situations of presence and absence of ovarian hormones.
Collapse
|
33
|
The impaired fibrinolytic capacity in hypertension is unaffected by acute blood pressure lowering. J Thromb Thrombolysis 2012; 32:399-404. [PMID: 21614456 DOI: 10.1007/s11239-011-0595-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The endogenous fibrinolytic system and the ability of the endothelium to release tissue-plasminogen activator (t-PA) play a pivotal role to protect humans from atherothrombotic events. We have recently reported that the decreased capacity for t-PA release in hypertension is restored with chronic blood pressure lowering. Thus, we explored if acute blood pressure lowering has the same effect. The capacity for acute t-PA release was investigated in the perfused-forearm model during stimulation by intra-arterial substance P 8 pmol/min in hypertensive subjects. The procedure was then repeated during acute blood pressure lowering (n = 9) induced by sodium nitroprusside (SNP) infusion or during placebo infusion (n = 3). SNP lowered mean arterial pressure from 108.6 (2.6) to 83.0 (2.6) (mean and SEM) mmHg (P < 0.001). Substance P induced significant increase in t-PA release during both high- and low-pressure conditions (P < 0.01, ANOVA). Peak t-PA release rate was 199 (77) and 167 (41) (mean and SEM) ng/min/l tissue, and accumulated t-PA release was 2,395 (750) and 2,394 (473) ng, during high- and low-pressure conditions, respectively. t-PA release and hemodynamic responses were almost identical during high- and low-pressure conditions (P = ns, for all). Acute blood pressure lowering does not restore stimulated t-PA release from the endothelium in hypertensive subjects. These findings are in contrast to previously described effects of chronic blood pressure treatment. Although data need to be confirmed in a larger study, they suggest that high blood pressure decreases the cellular t-PA pool rather than interferes with release mechanisms of the protein.
Collapse
|
34
|
Karami R, Hosseini M, Khodabandehloo F, Khatami L, Taiarani Z. Different effects of L-arginine on morphine tolerance in sham and ovariectomized female mice. J Zhejiang Univ Sci B 2011; 12:1016-1023. [PMID: 22135151 PMCID: PMC3232435 DOI: 10.1631/jzus.b1100029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Accepted: 04/29/2011] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The roles of gonadal hormones and nitric oxide (NO) on the analgesic effects of morphine, tolerance to morphine, and their interactions have been widely investigated. In the present study, the effect of L-arginine (an NO precursor) on morphine tolerance in sham and ovariectomized (OVX) female mice was investigated. METHODS Forty mice were divided into sham and OVX groups. On the first day, a hot plate test ((55±0.2) °C; cut-off 30 s) was carried out as a base record 15 min before injection of morphine (10 mg/kg, subcutaneously (s.c.)) and was repeated every 15 min after injection. The sham group was then divided into two subgroups: sham-tolerance-L-arginine (Sham-Tol-LA) and sham-tolerance-saline (Sham-Tol-Sal) which received either L-arginine 50 mg/kg (intraperitoneally (i.p.)) or saline 10 ml/kg (i.p.), respectively, three times in a day for three consecutive days. Morphine tolerance was induced in animals by injecting 30 mg/kg morphine (s.c.) three times/day for three days. This treatment was also used for OVX subgroups. On the fifth day, the hot plate test was repeated. The analgesic effect of morphine was calculated as the maximal percent effect (MPE). The results were compared using repeated measure analysis of variance (ANOVA). RESULTS There was no significant difference in MPE between the OVX and sham groups. The MPEs in both the Sham-Tol-Sal and OVX-Tol-Sal groups were lower than those in both the sham and OVX groups (P<0.01). The MPE in the OVX-Tol-Sal group was greater than that in the Sham-Tol-Sal group (P<0.01). The MPE in the Sham-Tol-LA group was higher than that in the Sham-Tol-Sal group (P<0.01). However, there was no significant difference between the Sham-Tol-LA and sham groups or between the OVX-Tol-LA and OVX-Tol-Sal groups. CONCLUSIONS The results of the present study showed that repeated administration of morphine causes tolerance to the analgesic effect of morphine. L-arginine could prevent tolerance to morphine but its effect was different in the presence of ovarian hormones.
Collapse
Affiliation(s)
- Reza Karami
- Neuroscience Research Center and Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Neuroscience Research Center and Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmacological Research Center of Medicinal Plants, Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatimeh Khodabandehloo
- Neuroscience Research Center and Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Khatami
- Neuroscience Research Center and Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Taiarani
- Pharmacological Research Center of Medicinal Plants, Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
35
|
Hosseini M, Nemati Karimooy HA, Hadjzadeh MAR, Safari V. Inducible nitric oxide synthase inhibitor aminoguanidine, differently affects Morris water maze tasks of ovariectomized and naïve female rats. ACTA PHYSIOLOGICA HUNGARICA 2011; 98:421-432. [PMID: 22173023 DOI: 10.1556/aphysiol.98.2011.4.5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The role of ovarian hormones, nitric oxide, and their interaction on learning and memory has been widely investigated. The objective of present study was to investigate different effects of chronic administration of inducible nitric oxide synthase inhibitor, aminoguanidine (AM) on learning and memory of ovariectomized (OVX) and naïve (Sham) female rats. Thirty-two rats were divided into four groups: 1) Sham, 2) OVX, 3) Sham-AM and 4) OVX-AM. The animals of Sham-AM and OVX-AM chronically received 100 mg/kg/day of aminoguanidine during 8 weeks before 5 test days. The animals in Sham and OVX groups received 1 ml/kg saline instead of aminoguanidine. The animals were tested in Morris water maze and the escape latency and traveled path to reach the platform were compared between groups. On the fifth day, the platform was removed, and the animals were allowed to swim for 60 s ( prob trial). The time spent in the target quadrant (Q1) was compared between groups.Results showed that the escape latency and traveled path in OVX group were significantly higher than in the Sham group (p<0.01). Both escape latency and traveled path in the Sham-AM group was significantly higher than in the Sham group (p<0.01) however, there was no significant difference between OVX-AM and OVX groups.The time spent by the animals of OVX group in the target quadrant (Q1) during the probe trial was significantly lower than that in the Sham group (p<0.01). The animals of the Sham-AM group spent shorter times in the target quadrant in comparison with the Sham group (p<0.01). There was no significant difference between the OVX and OVX-AM groups in the time spent in tarthe get quadrant. It is concluded that the effect of aminoguanidine on learning and memory is different in the presence or absence of ovarian hormones but it needs further investigation.
Collapse
Affiliation(s)
- Mahmoud Hosseini
- Department of Physiology, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | | | | |
Collapse
|
36
|
Direct vasorelaxation by a novel phytoestrogen tanshinone IIA is mediated by nongenomic action of estrogen receptor through endothelial nitric oxide synthase activation and calcium mobilization. J Cardiovasc Pharmacol 2011; 57:340-7. [PMID: 21383591 DOI: 10.1097/fjc.0b013e31820a0da1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Salvia miltiorrhiza (Danshen) has been widely used in China and other Asian countries for treating various cardiovascular diseases resulting from its ability to improve coronary microcirculation and increase coronary blood flow. Tanshinone IIA (Tan IIA), the major active lipophilic ingredient responsible for the beneficial actions of Salvia miltiorrhiza, has been shown to induce vasodilation in coronary arteries. Because our recent study identified Tan IIA as a new member of the phytoestrogens, we hypothesized that its action might be mediated by estrogen receptor (ER) in vascular endothelial cells. The aim of the present study was to assess whether cardiovascular protection exerted by Tan IIA is mediated by the ER signal pathway and whether the genomic or nongenomic action of ER is involved within arteries and vascular endothelial cells. The effect of Tan IIA on blood vessels was investigated by vascular ring assay using endothelium-intact and endothelium-denuded rat aortas. Similar to estrogen, Tan IIA caused an nitric oxide- and endothelium-dependent relaxation, which was blocked by ER antagonist ICI 182,780. Primary cardiac microvascular endothelial cells were used as a model to study the cellular and molecular mechanisms of Tan IIA-induced vasorelaxation. We demonstrate that Tan IIA is capable of activating the estrogen receptor signal pathway, leading to increased endothelial nitric oxide synthase gene expression, nitric oxide production, ERK1/2 phosphorylation, and Ca mobilization. Collectively, these effects contribute to Tan IIA's vasodilative activity effects of y ER antagonist Cnt of cardiovascular diseases. Our findings support a continued effort in discovering and developing novel phytoestrogens as an alternative hormone replacement therapy for safer and more effective treatment of cardiovascular diseases.
Collapse
|
37
|
Keung W, Man RYK. Circulating sex hormones modulate vascular contractions and acute response to 17β-estradiol in rat mesenteric arteries. Pharmacology 2011; 88:55-64. [PMID: 21791946 DOI: 10.1159/000329426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 05/09/2011] [Indexed: 11/19/2022]
Abstract
AIMS We investigated how modification of levels of the sex hormones 17β-estradiol and testosterone affects vascular contraction and nongenomic vascular effects of 17β-estradiol. METHODS Male and female rats were treated with vehicle, 17β-estradiol (25 μg/kg/day) or testosterone (1 mg/kg/day) for 14 consecutive days after sham gonadectomy or gonadectomy was performed. Isometric tensions were then measured from mesenteric arteries of each group of rats. RESULTS Contraction to phenylephrine was increased in mesenteric arteries from rats with or without gonadectomy treated with testosterone for 14 days compared to their intact controls. Contraction to phenylephrine was reduced in mesenteric arteries of rats with or without gonadectomy treated with 17β-estradiol for 14 days compared to their intact controls. Incubation of mesenteric arteries with 17β-estradiol (1 nmol/l) for 30 min reduced contraction to phenylephrine in mesenteric arteries of rats that were treated with testosterone for 14 days. This acute incubation of 17β-estradiol had no effect on arteries from rats that were treated with 17β-estradiol for 14 days. The acute effect of 17β-estradiol (1 nmol/l) is preserved in arteries without endothelium. CONCLUSION Our results suggest that 14 days' testosterone treatment enhances while 14 days' 17β-estradiol treatment suppresses contraction as well as the nongenomic effects of 17β-estradiol in the vascular smooth muscles.
Collapse
Affiliation(s)
- Wendy Keung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, SAR, China.
| | | |
Collapse
|
38
|
Abstract
Lung cancer is the leading cause of cancer mortality in both women and men worldwide but gender differences exist in their clinical and biological manifestations. In particular, among life time non-smoker, female are far more likely to develop lung carcinoma than male. Recent studies demonstrated that estrogens are synthesized in situ in both male and female lung cancers through aromatase, suggesting that sex steroid may contribute to the pathogenesis and development of lung carcinoma. In addition, human lung carcinomas have been recently demonstrated to be frequently associated with expression of estrogen receptors in both male and female patients and a lower expression of aromatase was reported to be associated with better prognosis. Preclinical studies further demonstrated that aromatase inhibitor (AI) suppressed the lung tumor growth both in vitro and in vivo. These findings all suggest a potential role of intratumoral aromatase in biological behavior of non-small cell lung cancer (NSCLC), the most common form of human lung malignancy. Therefore, AIs may become viable therapeutic options for disease management in NSCLC patients but further studies are definitely required to obtain a better understanding of the potential roles of intratumoral aromatase expression as a predictive biomarker for clinical outcome in these NSCLC patients.
Collapse
Affiliation(s)
- Mohit K Verma
- Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seriyo-machi, Aoba-ku, Sendai 980-8575, Miyagi-ken, Japan
| | | | | |
Collapse
|
39
|
Wu Q, Chambliss K, Umetani M, Mineo C, Shaul PW. Non-nuclear estrogen receptor signaling in the endothelium. J Biol Chem 2011; 286:14737-43. [PMID: 21343284 PMCID: PMC3083154 DOI: 10.1074/jbc.r110.191791] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In addition to the classical function of estrogen receptors (ER) as transcription factors, evidence continues to accumulate that they mediate non-nuclear processes in numerous cell types, including the endothelium, in which they activate endothelial NO synthase. Non-nuclear ER signaling entails unique post-translational modifications and protein-protein interactions of the receptor with adaptor molecules, kinases, and G proteins. Recent in vitro and in vivo studies in mice using an estrogen-dendrimer conjugate that is excluded from the nucleus indicate that non-nuclear ER activation underlies the migration and growth responses of endothelial cells to estrogen but not the growth responses of endometrial or breast cancer cells to the hormone. In this minireview, the features of ERα and protein-protein interactions that enable it to invoke extranuclear signaling in the endothelium and the consequences of that signaling are discussed.
Collapse
Affiliation(s)
- Qian Wu
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Ken Chambliss
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Michihisa Umetani
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Chieko Mineo
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Philip W. Shaul
- From the Division of Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
40
|
Hosseini M, Taiarani Z, Hadjzadeh MAR, Salehabadi S, Tehranipour M, Alaei HA. Different responses of nitric oxide synthase inhibition on morphine-induced antinociception in male and female rats. PATHOPHYSIOLOGY 2011; 18:143-149. [DOI: 10.1016/j.pathophys.2010.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2009] [Revised: 11/16/2009] [Accepted: 05/18/2010] [Indexed: 01/28/2023] Open
|
41
|
Lee DY, Kim JY, Kim JH, Choi DS, Kim DK, Koh KK, Yoon BK. Effects of hormone therapy on ambulatory blood pressure in postmenopausal Korean women. Climacteric 2011; 14:92-9. [DOI: 10.3109/13697137.2010.491924] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
42
|
Azizi-Malekabadi H, Hosseini M, Saffarzadeh F, Karami R, Khodabandehloo F. Chronic treatment with the nitric oxide synthase inhibitor, L-NAME, attenuates estradiol-mediated improvement of learning and memory in ovariectomized rats. Clinics (Sao Paulo) 2011; 66:673-679. [PMID: 21655764 PMCID: PMC3093799 DOI: 10.1590/s1807-59322011000400024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Revised: 10/21/2010] [Accepted: 12/07/2010] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION The role of ovarian hormones and nitric oxide in learning and memory has been widely investigated. OBJECTIVE The present study was carried out to evaluate the effect of the nitric oxide synthase (NOS) inhibitor, N (G)-nitro-L-arginine methyl ester (L-NAME), on the ability of estradiol to improve learning in OVX rats using the Morris water maze. METHODS Forty rats were divided into five groups: (1) ovariectomized (OVX), (2) ovariectomized-estradiol (OVX-Est), (3) ovariectomized-L-NAME 10 (OVX-LN 10), (4) ovariectomized-L-NAME 50 (OVX-LN 50) and (5) ovariectomized-estradiol-L-NAME 50 (OVX-Est-LN 50). The animals in the OVX-Est group were treated with a weekly injection of estradiol valerate (2 mg/kg; i.m.). The OVX-LN 10 and OVX-LN 50 groups were treated with daily injections of 10 and 50 mg/kg L-NAME (i.p.), respectively. The animals in the OVX-Est-LN 50 group received a weekly injection of estradiol valerate and a daily injection of 50 mg/kg L-NAME. After 8 weeks, all animals were tested in the Morris water maze. RESULTS The animals in the OVX-Est group had a significantly lower latency in the maze than the OVX group (p<0.001). There was no significant difference in latency between the OVX-LN 10 and OVX-LN 50 groups in comparison with the OVX group. The latency in the OVX-Est-LN 50 group was significantly higher than that in the OVX-Est group (p<0.001). CONCLUSION These results show that L-NAME treatment attenuated estradiol-mediated enhancement of spatial learning and memory in OVX rats, but it had no significant effect in OVX rats without estrogen, suggesting an interaction of nitric oxide and estradiol in these specific brain functions.
Collapse
Affiliation(s)
- Hamid Azizi-Malekabadi
- Dept. of Biology, Faculty of Basic Science, Islamic Azad University, Khuraskan Branch, Isfahan, Iran
| | | | | | | | | |
Collapse
|
43
|
Bellefontaine N, Hanchate NK, Parkash J, Campagne C, de Seranno S, Clasadonte J, d'Anglemont de Tassigny X, Prevot V. Nitric oxide as key mediator of neuron-to-neuron and endothelia-to-glia communication involved in the neuroendocrine control of reproduction. Neuroendocrinology 2011; 93:74-89. [PMID: 21335953 DOI: 10.1159/000324147] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 01/04/2011] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) is a peculiar chemical transmitter that freely diffuses through aqueous and lipid environments and plays a role in major aspects of brain function. Within the hypothalamus, NO exerts critical effects upon the gonadotropin-releasing hormone (GnRH) network to maintain fertility. Here, we review recent evidence that NO regulates major aspects of the GnRH neuron physiology. Far more active than once thought, NO powerfully controls GnRH neuronal activity, GnRH release and structural plasticity at the neurohemal junction. In the preoptic region, neuronal nitric oxide synthase (nNOS) activity is tightly regulated by estrogens and is found to be maximal at the proestrus stage. Natural fluctuations of estrogens control both the differential coupling of this Ca²+-activated enzyme to glutamate N-methyl-D-aspartic acid receptor channels and phosphorylation-mediated nNOS activation. Furthermore, NO endogenously produced by neurons expressing nNOS acutely and directly suppresses spontaneous firing in GnRH neurons, which suggests that neuronal NO may serve as a synchronizing switch within the preoptic region. At the median eminence, NO is spontaneously released from an endothelial source and follows a pulsatile and cyclic pattern of secretion. Importantly, GnRH release appears to be causally related to endothelial NO release. NO is also highly involved in mediating the dialogue set in motion between vascular endothelial cells and tanycytes that control the direct access of GnRH neurons to the pituitary portal blood during the estrous cycle. Altogether, these data raise the intriguing possibility that the neuroendocrine brain uses NO to coordinate both GnRH neuronal activity and GnRH release at key stages of reproductive physiology.
Collapse
Affiliation(s)
- Nicole Bellefontaine
- Inserm, Jean-Pierre Aubert Research Center, Development and Plasticity of the Postnatal Brain, U837, Lille, France
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Chistyakov VA, Serezhenkov VA, Alexandrova AA, Milyutina NP, Prokof’ev VN, Mashkina EV, Gutnikova LV, Dem’yanenko SV. Effect of Plastoquinone Derivative 10-(6′-Plastoquinonyl)decyltriphenylphosphonium (SkQ1) on Contents of Steroid Hormones and NO Level in Rats. BIOCHEMISTRY (MOSCOW) 2010; 75:1383-7. [DOI: 10.1134/s0006297910110106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Dias DPM, Oliveira M, Salgado HC, Fazan R. Ovariectomy does not affect the cardiac sympathovagal balance of female SHR but estradiol does. Braz J Med Biol Res 2010; 43:969-75. [PMID: 20945035 DOI: 10.1590/s0100-879x2010007500105] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Accepted: 09/23/2010] [Indexed: 11/22/2022] Open
Abstract
The low incidence of cardiovascular diseases, including hypertension, in premenopausal women has led to the conclusion that ovarian hormones may have a protective effect on the cardiovascular system. We evaluated the effects of ovariectomy and/or estradiol on sympathovagal balance and heart rate variability (HRV) in female spontaneously hypertensive rats (SHR) with tachycardia and compared them to Wistar rats (12 weeks old; N = 8-12). Ovariectomy (OVX) and/or estradiol (10 µg/kg) did not affect basal arterial pressure in either rat strain, but estradiol increased basal heart rate (HR) in OVX SHR (454 ± 18 vs 377 ± 9 bpm). HR changes elicited by methylatropine and propranolol were used to evaluate the sympathovagal balance. Ovariectomy did not affect the cardiac sympathovagal balance of any group, while estradiol increased sympathetic tone in OVX SHR (120 ± 8 vs 56 ± 10 bpm) and sham-operated Wistar rats (57 ± 7 vs 28 ± 4 bpm), and decreased the parasympathetic tone only in OVX SHR (26 ± 7 vs 37 ± 5 bpm). HRV was studied in the frequency domain (Fast Fourier Transformation). Spectra of HR series were examined at low frequency (LF: 0.2-0.75 Hz) and high frequency (HF: 0.75-3 Hz) bands. The power of LF, as well as the LF/HF ratio, was not affected by ovariectomy, but estradiol increased both LF (29 ± 4 vs 18 ± 3 nu in Wistar sham-operated, 26 ± 5 vs 15 ± 3 nu in Wistar OVX, 50 ± 3 vs 38 ± 4 nu in SHR sham-operated, and 51 ± 3 vs 42 ± 3 nu in SHR OVX) and LF/HF (0.48 ± 0.08 vs 0.23 ± 0.03 nu in Wistar sham-operated, 0.41 ± 0.14 vs 0.19 ± 0.05 nu in Wistar OVX, 0.98 ± 0.11 vs 0.63 ± 0.11 nu in SHR sham-operated, and 1.10 ± 0.11 vs 0.78 ± 0.1 nu in SHR OVX). Thus, we suggest that ovariectomy did not affect the cardiac sympathovagal balance of SHR or Wistar rats, while estradiol increased the sympathetic modulation of HR.
Collapse
Affiliation(s)
- D P M Dias
- Departamento de Fisiologia, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | | | | | | |
Collapse
|
46
|
Saffarzadeh F, Eslamizade MJ, Nemati Karimooy HA, Hadjzadeh MA, Khazaei M, Hosseini M. The effect of L-arginine on Morris water maze tasks of ovariectomized rats. ACTA PHYSIOLOGICA HUNGARICA 2010; 97:216-223. [PMID: 20511131 DOI: 10.1556/aphysiol.97.2010.2.8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The role of ovarian hormones, nitric oxide, and their interaction on learning and memory has been widely investigated. The present study was carried out to evaluate the effect of acute (A) and chronic (C) L-arginine (L-Arg) administration on learning and memory in ovariectomized (OVx) rats. Forty-five rats were divided into six groups: 1) sham, 2) OVx, 3) sham-L-Arg-A, 4) OVx-L-Arg-A, 5) sham-L-Arg-C, 6) OVx-L-Arg-C. The animals of sham-L-Arg-A and OVx-L-Arg-A were acutely treated with 500 mg/kg of L-Arg during 5 test days. Sham-L-Arg-C and OVx-L-Arg-C chronically received 500 mg/kg/day of L-Arg during 8 weeks before 5 test days. The animals in sham and OVx groups received 1 ml/kg saline instead of L-Arg. At the end of the experiment, the animals were tested in Morris water maze and the escape latency and traveled path to reach the platform were compared between groups. Results showed that the escape latency and traveled path in OVx group were significantly higher than in sham group ( p <0.05). The animals in OVx-L-Arg-A group had significantly lower traveled path length and escape latency compared to OVx group ( p <0.001). Chronic treatment by L-Arg had no significant effect on learning and memory in OVx and sham-operated animals. It seems that L-Arg has a role in Morris water maze tasks disturbances in OVx rats but it needs to be further investigated.
Collapse
Affiliation(s)
- F Saffarzadeh
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | | | | | | |
Collapse
|
47
|
Thor D, Uchizono JA, Lin-Cereghino GP, Rahimian R. The effect of 17 beta-estradiol on intracellular calcium homeostasis in human endothelial cells. Eur J Pharmacol 2010; 630:92-9. [PMID: 20044991 DOI: 10.1016/j.ejphar.2009.12.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 11/17/2009] [Accepted: 12/17/2009] [Indexed: 10/20/2022]
Abstract
The cardiovascular effects of estrogen are mediated in part by augmenting the function of endothelial nitric oxide synthase. Endothelial nitric oxide synthase activity is dependent on many cofactors including Ca(2+). Hence, we investigated the effect of chronic 17 beta-estradiol treatment on the intracellular Ca(2+) concentration and endothelial nitric oxide synthase protein expression in the human endothelial cell line, EA.hy926, using spectrofluorometry and Western blot, respectively. Inhibiting the sarco(endo)plasmic reticulum Ca(2+) ATPase with thapsigargin caused an increase in the intracellular Ca(2+) concentration, which was higher in chronically 17 beta-estradiol-treated (1muM, 24h) cells loaded with Fura-2-acetoxymethyl ester compared to vehicle-treated cells, suggesting a higher endoplasmic reticulum Ca(2+) content in 17 beta-estradiol-treated cells. An enhanced Ca(2+) influx pathway in chronically 17 beta-estradiol-treated cells was also observed. In addition, 17 beta-estradiol-treated cells expressed higher levels of endothelial nitric oxide synthase protein in comparison to vehicle-treated cells. The chronic effect of 17 beta-estradiol on Ca(2+) homeostasis and endothelial nitric oxide synthase expression was attenuated with the nonselective estrogen receptor inhibitor, ICI 182,780 (10muM, 7alpha, 17beta-[9-[(4,4,5,5,5-Pentafluoropentyl)sulfinyl]nonyl] estra-1,3,5(10)-triene-3,17-diol). Furthermore, analysis of the thapsigargin-evoked Ca(2+) response in chronically 17 beta-estradiol-treated estrogen receptor alpha-knockdown cells showed no significant difference in Ca(2+) response compared to vehicle-treated estrogen receptor alpha-knockdown cells, indicating that the regulation of Ca(2+) homeostasis by 17 beta-estradiol is mediated through an estrogen receptor alpha-dependent pathway. These data revealed an estrogen receptor alpha-dependent modulation of Ca(2+) homeostasis accompanying the enhancement of endothelial nitric oxide synthase expression in 17 beta-estradiol-treated human endothelial cells.
Collapse
Affiliation(s)
- Der Thor
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy and Health Sciences, 751 Brookside Rd, University of the Pacific, Stockton, CA 95211, USA
| | | | | | | |
Collapse
|
48
|
Galbraith H. Hormones in international meat production: biological, sociological and consumer issues. Nutr Res Rev 2009; 15:293-314. [PMID: 19087409 DOI: 10.1079/nrr200246] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Beef and its products are an important source of nutrition in many human societies. Methods of production vary and include the use of hormonal compounds ('hormones') to increase growth and lean tissue with reduced fat deposition in cattle. The hormonal compounds are naturally occurring in animals or are synthetically produced xenobiotics and have oestrogenic (oestradiol-17beta and its esters; zeranol), androgenic (testosterone and esters; trenbolone acetate) or progestogenic (progesterone; melengestrol acetate) activity. The use of hormones as production aids is permitted in North American countries but is no longer allowed in the European Union (EU), which also prohibits the importation of beef and its products derived from hormone-treated cattle. These actions have resulted in a trade dispute between the two trading blocs. The major concern for EU authorities is the possibility of adverse effects on human consumers of residues of hormones and metabolites. Methods used to assess possible adverse effects are typical of those used by international agencies to assess acceptability of chemicals in human food. These include analysis of quantities present in the context of known biological activity and digestive, absorptive, post-absorptive and excretory processes. Particular considerations include the low quantities of hormonal compounds consumed in meat products and their relationships to endogenous production particularly in prepubertal children, enterohepatic inactivation, cellular receptor- and non-receptor-mediated effects and potential for interference with growth, development and physiological function in consumers. There is particular concern about the role of oestradiol-17beta as a carcinogen in certain tissues. Now subject to a 'permanent' EU ban, current evidence suggests that certain catechol metabolites may induce free-radical damage of DNA in cell and laboratory animal test systems. Classical oestrogen-receptor mediation is considered to stimulate proliferation in cells maintaining receptivity. Mathematical models describing quantitative relationships between consumption of small amounts of oestrogens in meat in addition to greater concentrations from endogenous production, chemical stoichiometry at cellular level and human pathology have not been developed. Such an approach will be necessary to establish 'molecular materiality' of the additional hormone intake as a component of relative risk assessment. The other hormones, although generally less well researched, are similarly subject to a range of tests to determine potentially adverse effects. The resulting limited international consensus relates to the application of the 'precautionary principle' and non-acceptance by the European Commission of the recommendations of the Codex Alimentarius Commission, which determined that meat from cattle, hormone-treated according to good practice, was safe for human consumers. The present review considers the hormone issue in the context of current international social methodology and regulation, recent advances in knowledge of biological activity of hormones and current status of science-based evaluation of food safety and risk for human consumers.
Collapse
Affiliation(s)
- Hugh Galbraith
- Department of Agriculture and Forestry University of Aberdeen 581 King Street Aberdeen AB24 5UA, UK.
| |
Collapse
|
49
|
Hosseini M, Sadeghnia HR, Salehabadi S, Alavi H, Gorji A. The effect of L-arginine and L-NAME on pentylenetetrazole induced seizures in ovariectomized rats, an in vivo study. Seizure 2009; 18:695-698. [PMID: 19854076 DOI: 10.1016/j.seizure.2009.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 09/19/2009] [Accepted: 09/24/2009] [Indexed: 11/22/2022] Open
Abstract
The role of ovarian hormones and nitric oxide (NO) on seizure and their interaction have been widely investigated. The present study carried out to evaluate the effect of chronic administration of L-arginine (LA) and L-NAME (LN) on pentylenetetrazole (PTZ) induced epilepsy in ovariectomized (OVX) and naïve female rats. Fourty-eight female rats were randomly divided into six groups (n=8) as follows: (1) sham, (2) ovarectomized (OVX), (3) sham-LA, (4) sham-LN, (5) OVX-LA, and (6) OVX-LN. The animals of sham-LA and OVX-LA received daily injection of 500 mg/kg L-arginine (i.p.) during 4 weeks. Sham-LN and OVX-LN were treated by 10 mg/kg L-NAME (i.p.) daily for 4 weeks. The animals of sham and OVX groups received 1 ml/kg saline (i.p.) instead of L-arginine and L-NAME. The latencies to minimal clonic seizures (MCS) and generalized tonic-clonic seizures (GTCS) after intraperitoneal injection of penetylenetetrazole (PTZ, 90 mg/kg) was recorded and compared between groups. A significant increase in the GTCS, but not MCS, latency was seen in OVX rats in comparison with sham-operated animals. Pretreatment of animals with L-NAME resulted in a significant increase in the GTCS and MCS latencies in sham group while no significant effects were seen in OVX rats. On the contrary, while pretreatment with L-arginine had no effects on MCS and GTCS latencies in sham group, a significant decrease in GTCS latency was observed in OVX rats. It is concluded that ovarian sex hormones affect seizure thresholds induced by PTZ and NO has a role on seizures susceptibility following PTZ administration. This NO effect might be differing in the presence or absence of ovarian hormones, but further investigations need to be done.
Collapse
Affiliation(s)
- Mahmoud Hosseini
- Dept. of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | | | | | | |
Collapse
|
50
|
Vornehm ND, Wang M, Abarbanell A, Herrmann J, Weil B, Tan J, Wang Y, Kelly M, Meldrum DR. Acute postischemic treatment with estrogen receptor-alpha agonist or estrogen receptor-beta agonist improves myocardial recovery. Surgery 2009; 146:145-54. [PMID: 19628068 DOI: 10.1016/j.surg.2009.04.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 04/17/2009] [Indexed: 11/24/2022]
Abstract
BACKGROUND After ischemia/reperfusion (I/R) injury, female hearts demonstrate improved functional recovery compared with male, which suggests a protective role for estrogen. Acute postischemic treatment with 17-beta-estradiol (E2) attenuates myocardial dysfunction. However, it is unknown by which estrogen receptor (ER) E2 mediates this acute cardioprotection during I/R. Therefore, we hypothesize that postischemic infusion of the selective ER-alpha agonist (4,4',4''-[4-propyl-(1H)-pyrazole-1,3,5-triyl]tris-phenol [PPT]) or the selective ER-beta agonist (2,3-bis(4-hydroxyphenyl)-propionitrile [DPN]) will improve myocardial function after I/R injury. METHODS Isolated, perfused hearts (Langendorff) from adult male rats were subjected to 25 minutes of ischemia followed by 40 minutes of reperfusion. Hearts (n = 4-6 per group) were randomly infused with either perfusate, PPT or DPN at 1, 10, or 100 nmol/L throughout reperfusion. After I/R, heart tissue was analyzed for tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, vascular endothelial growth factor (VEGF), and lactate dehydrogenase (LDH). RESULTS Postischemic treatment with 10 nmol/L of PPT significantly improved myocardial function. Additionally, 10 or 100 nmol/L of DPN significantly increased myocardial functional recovery after I/R injury, with maximum benefit at the 10 nmol/L dose. A trend toward lower levels of LDH was noted in DPN- and PPT-treated groups after I/R injury. Neither PPT nor DPN affected myocardial production of TNF-alpha or IL-1beta. However, higher levels of myocardial VEGF were noted in the PPT-treated group compared with controls. CONCLUSION Both ER-alpha and ER-beta are involved in mediating E2-induced rapid cardioprotection after I/R injury. Advancing our understanding of both ER subtypes may be useful for the development of novel strategies that may benefit both males and females in response to myocardial ischemia.
Collapse
Affiliation(s)
- Nicholas D Vornehm
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|