1
|
Zhao W, Liu P, Saunders TR, Zhu J. Juvenile hormone induces phosphorylation of insulin/insulin-like growth factor signaling proteins in previtellogenic Aedes aegypti mosquitoes. INSECT SCIENCE 2024. [PMID: 39663731 DOI: 10.1111/1744-7917.13482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 12/13/2024]
Abstract
Juvenile hormone (JH) plays a pivotal role in regulating post-emergence development and metabolism in previtellogenic female Aedes aegypti mosquitoes. In contrast, yolk protein precursor production and egg maturation after a blood meal are regulated by the steroid hormone 20-hydroxyecdysone, the insulin-like growth factor (IGF)/insulin signaling (IIS) pathway, and the mammalian target of rapamycin (mTOR) pathway. The role of IIS/mTOR signaling in female adults prior to blood feeding has not been thoroughly investigated. In this study, we identified a significant increase in the phosphorylation of key effector proteins in the IIS/mTOR signaling pathway, including eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), ribosomal protein S6 kinase (S6K) and forkhead box protein O1 (FoxO1), in previtellogenic females. In vitro fat body culture experiments suggest that JH induces these phosphorylations through rapid nongenomic signaling mediated by the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mTOR network. RNA interference experiments demonstrated that activation of IIS/mTOR signaling in previtellogenic females modulate metabolic gene expression, promoting the accumulation of energy reserves (glycogen and triglycerides), which influence mosquito fecundity. Additionally, depletion of either the insulin receptor (InR) or the JH receptor Methoprene-tolerant (Met) in adult mosquitoes abolished the phosphorylation of these proteins, indicating that both receptors are involved in JH-induced membrane-initiated signal transduction. Although the precise mechanisms remain unclear, this study uncovers a novel function of the IIS/mTOR pathway in adult mosquitoes before blood feeding, as well as a new mode of JH action through its crosstalk with the IIS pathway.
Collapse
Affiliation(s)
- Wenhao Zhao
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
| | - Pengcheng Liu
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
- Laboratory of Bio-Interactions and Crop Health, Nanjing Agricultural University, Nanjing, China
| | - Thomas R Saunders
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
| | - Jinsong Zhu
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
2
|
Gall CM, Le AA, Lynch G. Contributions of site- and sex-specific LTPs to everyday memory. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230223. [PMID: 38853551 PMCID: PMC11343211 DOI: 10.1098/rstb.2023.0223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/27/2024] [Accepted: 03/06/2024] [Indexed: 06/11/2024] Open
Abstract
Commentaries about long-term potentiation (LTP) generally proceed with an implicit assumption that largely the same physiological effect is sampled across different experiments. However, this is clearly not the case. We illustrate the point by comparing LTP in the CA3 projections to CA1 with the different forms of potentiation in the dentate gyrus. These studies lead to the hypothesis that specialized properties of CA1-LTP are adaptations for encoding unsupervised learning and episodic memory, whereas the dentate gyrus variants subserve learning that requires multiple trials and separation of overlapping bodies of information. Recent work has added sex as a second and somewhat surprising dimension along which LTP is also differentiated. Triggering events for CA1-LTP differ between the sexes and the adult induction threshold is significantly higher in females; these findings help explain why males have an advantage in spatial learning. Remarkably, the converse is true before puberty: Females have the lower LTP threshold and are better at spatial memory problems. A mechanism has been identified for the loss-of-function in females but not for the gain-of-function in males. We propose that the many and disparate demands of natural environments, with different processing requirements across ages and between sexes, led to the emergence of multiple LTPs. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Christine M. Gall
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA92697, USA
- Department of Neurobiology and Behavior, University of California at Irvine, Irvine, CA92697, USA
| | - Aliza A. Le
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA92697, USA
| | - Gary Lynch
- Department of Anatomy and Neurobiology, University of California at Irvine, Irvine, CA92697, USA
- Department of Psychiatry and Human Behavior, University of California at Irvine, Irvine, CA92868, USA
| |
Collapse
|
3
|
Prestegui Martel B, Chávez-Blanco AD, Domínguez-Gómez G, Dueñas González A, Gaona-Aguas P, Flores-Mejía R, Somilleda-Ventura SA, Rodríguez-Cortes O, Morales-Bárcena R, Martínez Muñoz A, Mejia Barradas CM, Mendieta Wejebe JE, Correa Basurto J. N-(2-Hydroxyphenyl)-2-Propylpentanamide (HO-AAVPA) Induces Apoptosis and Cell Cycle Arrest in Breast Cancer Cells, Decreasing GPER Expression. Molecules 2024; 29:3509. [PMID: 39124913 PMCID: PMC11314247 DOI: 10.3390/molecules29153509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 08/12/2024] Open
Abstract
In this work, we performed anti-proliferative assays for the compound N-(2-hydroxyphenyl)-2-propylpentanamide (HO-AAVPA) on breast cancer (BC) cells (MCF-7, SKBR3, and triple-negative BC (TNBC) MDA-MB-231 cells) to explore its pharmacological mechanism regarding the type of cell death associated with G protein-coupled estrogen receptor (GPER) expression. The results show that HO-AAVPA induces cell apoptosis at 5 h or 48 h in either estrogen-dependent (MCF-7) or -independent BC cells (SKBR3 and MDA-MB-231). At 5 h, the apoptosis rate for MCF-7 cells was 68.4% and that for MDA-MB-231 cells was 56.1%; at 48 h, that for SKBR3 was 61.6%, that for MCF-7 cells was 54.9%, and that for MDA-MB-231 (TNBC) was 43.1%. HO-AAVPA increased the S phase in MCF-7 cells and reduced the G2/M phase in MCF-7 and MDA-MB-231 cells. GPER expression decreased more than VPA in the presence of HO-AAVPA. In conclusion, the effects of HO-AAVPA on cell apoptosis could be modulated by epigenetic effects through a decrease in GPER expression.
Collapse
Affiliation(s)
- Berenice Prestegui Martel
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional (IPN), Plan de San Luis y Díaz Mirón, Ciudad de México 11340, México; (B.P.M.); (A.M.M.); (C.M.M.B.)
| | - Alma Delia Chávez-Blanco
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, México; (A.D.C.-B.); (G.D.-G.); (A.D.G.); (R.M.-B.)
| | - Guadalupe Domínguez-Gómez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, México; (A.D.C.-B.); (G.D.-G.); (A.D.G.); (R.M.-B.)
| | - Alfonso Dueñas González
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, México; (A.D.C.-B.); (G.D.-G.); (A.D.G.); (R.M.-B.)
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Cancerología, Ciudad de México 04510, México
| | - Patricia Gaona-Aguas
- Laboratorio de Inflamación y Obesidad, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Ciudad de México 11340, México; (P.G.-A.); (R.F.-M.); (O.R.-C.)
| | - Raúl Flores-Mejía
- Laboratorio de Inflamación y Obesidad, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Ciudad de México 11340, México; (P.G.-A.); (R.F.-M.); (O.R.-C.)
| | - Selma Alin Somilleda-Ventura
- Centro de Investigación Biomédica, Fundación Hospital Nuestra Señora de la Luz I.A.P., Ezequiel Montes 135, Tabacalera, Ciudad de México 06030, México;
- Centro Interdisciplinario de Ciencias de la Salud-Instituto Politécnico Nacional (CICS-IPN), Ciudad de México 11340, México
| | - Octavio Rodríguez-Cortes
- Laboratorio de Inflamación y Obesidad, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Ciudad de México 11340, México; (P.G.-A.); (R.F.-M.); (O.R.-C.)
| | - Rocío Morales-Bárcena
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, México; (A.D.C.-B.); (G.D.-G.); (A.D.G.); (R.M.-B.)
| | - Alberto Martínez Muñoz
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional (IPN), Plan de San Luis y Díaz Mirón, Ciudad de México 11340, México; (B.P.M.); (A.M.M.); (C.M.M.B.)
| | - Cesar Miguel Mejia Barradas
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional (IPN), Plan de San Luis y Díaz Mirón, Ciudad de México 11340, México; (B.P.M.); (A.M.M.); (C.M.M.B.)
| | - Jessica Elena Mendieta Wejebe
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional (IPN), Plan de San Luis y Díaz Mirón, Ciudad de México 11340, México; (B.P.M.); (A.M.M.); (C.M.M.B.)
| | - José Correa Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional (IPN), Plan de San Luis y Díaz Mirón, Ciudad de México 11340, México; (B.P.M.); (A.M.M.); (C.M.M.B.)
| |
Collapse
|
4
|
Ma J, Niklewski PJ, Wang HS. Acute exposure to low-dose bisphenol A delays cardiac repolarization in female canine heart - Implication for proarrhythmic toxicity in large animals. Food Chem Toxicol 2023; 172:113589. [PMID: 36584932 PMCID: PMC9852101 DOI: 10.1016/j.fct.2022.113589] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/12/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022]
Abstract
Bisphenol A (BPA) is a common environmental chemical with a range of potential adverse health effects. The impact of environmentally-relevant low dose of BPA on the electrical properties of the hearts of large animals (e.g., dog, human) is poorly defined. Perturbation of cardiac electrical properties is a key arrhythmogenic mechanism. In particular, delay of ventricular repolarization and prolongation of the QT interval of the electrocardiogram is a marker for the risk of malignant arrhythmias. We examined the acute effect of 10-9 M BPA on the electrical properties of female canine ventricular myocytes and tissues. BPA rapidly delayed action potential repolarization and prolonged action potential duration (APD). The dose response curve of BPA on APD was nonmonotonic. BPA rapidly inhibited the IKr K+ current and ICaL Ca2+ current. Computational modeling indicated that the effect of BPA on APD can be accounted for by its suppression of IKr. At the tissue level, BPA acutely prolonged the QT interval in 4 left ventricular wedges. ERβ signaling contributed to the acute effects of BPA on ventricular repolarization. Our results demonstrate that BPA has QT prolongation liability in female canine hearts. These findings have implication for the potential proarrhythmic cardiac toxicity of BPA in large animals.
Collapse
Affiliation(s)
- Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Paul J Niklewski
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
5
|
Segovia-Mendoza M, Mirzaei E, Prado-Garcia H, Miranda LD, Figueroa A, Lemini C. The Interplay of GPER1 with 17β-Aminoestrogens in the Regulation of the Proliferation of Cervical and Breast Cancer Cells: A Pharmacological Approach. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:12361. [PMID: 36231664 PMCID: PMC9566056 DOI: 10.3390/ijerph191912361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 06/16/2023]
Abstract
The G-protein-coupled receptor for estrogen (GPER1) is a transmembrane receptor involved in the progression and development of various neoplasms whose ligand is estradiol (E2). 17β-aminoestrogens (17β-AEs) compounds, analogs to E2, are possible candidates for use in hormone replacement therapy (HRT), but our knowledge of their pharmacological profile is limited. Thus, we explored the molecular recognition of GPER1 with different synthetic 17β-AEs: prolame, butolame, and pentolame. We compared the structure and ligand recognition sites previously reported for a specific agonist (G1), antagonists (G15 and G36), and the natural ligand (E2). Then, the biological effects of 17β-AEs were analyzed through cell viability and cell-cycle assays in two types of female cancer. In addition, the effect of 17β-AEs on the phosphorylation of the oncoprotein c-fos was evaluated, because this molecule is modulated by GPER1. Molecular docking analysis showed that 17β-AEs interacted with GPER1, suggesting that prolame joins GPER1 in a hydrophobic cavity, similarly to G1, G15, and E2. Prolame induced cell proliferation in breast (MCF-7) and cervical cancer (SIHA) cells; meanwhile, butolame and pentolame did not affect cell proliferation. Neither 17β-AEs nor E2 changed the activation of c-fos in MCF-7 cells. Meanwhile, in SIHA cells, E2 and 17β-AEs reduced c-fos phosphorylation. Thus, our data suggest that butolame and pentolame, but not prolame, could be used for HRT without presenting a potential risk of inducing breast- or cervical-cancer-cell proliferation. The novelty of this work lies in its study of compound analogs to E2 that may represent important therapeutic strategies for women in menopause, with non-significant effects on the cell viability of cancer cells. The research focused on the interactions of GPER1, a molecule recently associated with promoting and maintaining various neoplasms.
Collapse
Affiliation(s)
- Mariana Segovia-Mendoza
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad No. 3000, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Elahe Mirzaei
- Instituto Nacional de Medicina Genómica, Col. Arenal Tepepan, Ciudad de México 14610, Mexico
| | - Heriberto Prado-Garcia
- Laboratorio de Onco-Inmunobiologia, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México 14080, Mexico
| | - Luis D. Miranda
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior S.N., Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Alejandra Figueroa
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad No. 3000, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Cristina Lemini
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad No. 3000, Ciudad Universitaria, Ciudad de México 04510, Mexico
| |
Collapse
|
6
|
Koukoulis GN, Filiponi M, Gougoura S, Befani C, Liakos P, Bargiota Α. Testosterone and dihydrotestosterone modulate the redox homeostasis of endothelium. Cell Biol Int 2022; 46:660-670. [DOI: 10.1002/cbin.11768] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 12/26/2021] [Accepted: 01/02/2022] [Indexed: 12/08/2022]
Affiliation(s)
- George N Koukoulis
- Research Laboratory, Department of Endocrinology and Metabolic Diseases, Larissa University Hospital, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Maria Filiponi
- Research Laboratory, Department of Endocrinology and Metabolic Diseases, Larissa University Hospital, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Sofia Gougoura
- Research Laboratory, Department of Endocrinology and Metabolic Diseases, Larissa University Hospital, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Christina Befani
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Panagiotis Liakos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| | - Αlexandra Bargiota
- Research Laboratory, Department of Endocrinology and Metabolic Diseases, Larissa University Hospital, Faculty of Medicine, University of Thessaly41500BiopolisLarissaGreece
| |
Collapse
|
7
|
Gur S, Alzweri L, Yilmaz‐Oral D, Kaya‐Sezginer E, Abdel‐Mageed AB, Dick B, Sikka SC, Volkan Oztekin C, Hellstrom WJG. Testosterone positively regulates functional responses and nitric oxide expression in the isolated human corpus cavernosum. Andrology 2020; 8:1824-1833. [DOI: 10.1111/andr.12866] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 04/30/2020] [Accepted: 07/08/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Serap Gur
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
- Department of Pharmacology Faculty of Pharmacy Ankara University Ankara Turkey
| | - Laith Alzweri
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
- Division of Urology, Department of Surgery University of Texas Medical Branch Galveston TX USA
| | - Didem Yilmaz‐Oral
- Department of Pharmacology Faculty of Pharmacy Cukurova University Adana Turkey
| | - Ecem Kaya‐Sezginer
- Department of Biochemistry Faculty of Pharmacy Ankara University Ankara Turkey
| | - Asim B Abdel‐Mageed
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
| | - Brian Dick
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
| | - Suresh C. Sikka
- Department of Urology Tulane University Health Sciences Center New Orleans LA USA
| | - Cetin Volkan Oztekin
- Department of Urology Faculty of Medicine University of KyreniaTurkish Republic of North Cyprus Girne, Mersin 10 Turkey
| | | |
Collapse
|
8
|
Loffredo LF, Coden ME, Berdnikovs S. Endocrine Disruptor Bisphenol A (BPA) Triggers Systemic Para-Inflammation and is Sufficient to Induce Airway Allergic Sensitization in Mice. Nutrients 2020; 12:nu12020343. [PMID: 32012983 PMCID: PMC7071314 DOI: 10.3390/nu12020343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 12/18/2022] Open
Abstract
Allergic airway diseases are accompanied by increased permeability and an inflammatory state of epithelial barriers, which are thought to be susceptible to allergen sensitization. Although exogenous drivers (proteases, allergens) of epithelial barrier disruption and sensitization are well studied, endogenous contributors (diet, xenobiotics, hormones, and metabolism) to allergic sensitization are much less understood. Xenoestrogens are synthetic or natural chemical compounds that have the ability to mimic estrogen and are ubiquitous in the food and water supply of developed countries. By interfering with the estrogen produced by the endocrine system, these compounds have the systemic potential to disrupt the homeostasis of multiple tissues. Our study examined the potential of prototypical xenoestrogen bisphenol A (BPA) to disrupt epithelial homeostasis in vitro and promote allergic responses in vivo. We found that BPA exposure in epithelial cultures in vitro significantly inhibited epithelial cell proliferation and wound healing, as well as promoted the expression of the innate alarmin cytokine TSLP in a time-and dose-dependent manner. In vivo, the exposure to BPA through water supply or inhalation induced a systemic para-inflammatory response by promoting the expression of innate inflammatory mediators in the skin, gut, and airway. In a murine tolerogenic antigen challenge model, chronic systemic exposure to BPA was sufficient to induce airway sensitization to innocuous chicken egg ovalbumin in the complete absence of adjuvants. Mechanistic studies are needed to test conclusively whether endocrine disruptors may play an upstream role in allergic sensitization via their ability to promote a para-inflammatory state.
Collapse
|
9
|
Luo J, Liu D. Does GPER Really Function as a G Protein-Coupled Estrogen Receptor in vivo? Front Endocrinol (Lausanne) 2020; 11:148. [PMID: 32296387 PMCID: PMC7137379 DOI: 10.3389/fendo.2020.00148] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/03/2020] [Indexed: 12/25/2022] Open
Abstract
Estrogen can elicit pleiotropic cellular responses via a diversity of estrogen receptors (ERs)-mediated genomic and rapid non-genomic mechanisms. Unlike the genomic responses, where the classical nuclear ERα and ERβ act as transcriptional factors following estrogen binding to regulate gene transcription in estrogen target tissues, the non-genomic cellular responses to estrogen are believed to start at the plasma membrane, leading to rapid activation of second messengers-triggered cytoplasmic signal transduction cascades. The recently acknowledged ER, GPR30 or GPER, was discovered in human breast cancer cells two decades ago and subsequently in many other cells. Since its discovery, it has been claimed that estrogen, ER antagonist fulvestrant, as well as some estrogenic compounds can directly bind to GPER, and therefore initiate the non-genomic cellular responses. Various recently developed genetic tools as well as chemical ligands greatly facilitated research aimed at determining the physiological roles of GPER in different tissues. However, there is still lack of evidence that GPER plays a significant role in mediating endogenous estrogen action in vivo. This review summarizes current knowledge about GPER, including its tissue expression and cellular localization, with emphasis on the research findings elucidating its role in health and disease. Understanding the role of GPER in estrogen signaling will provide opportunities for the development of new therapeutic strategies to strengthen the benefits of estrogen while limiting the potential side effects.
Collapse
Affiliation(s)
- Jing Luo
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
- Department of Human Nutrition, Foods and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Dongmin Liu
- Department of Human Nutrition, Foods and Exercise, College of Agricultural and Life Sciences, Virginia Tech, Blacksburg, VA, United States
- *Correspondence: Dongmin Liu
| |
Collapse
|
10
|
Roque C, Mendes-Oliveira J, Duarte-Chendo C, Baltazar G. The role of G protein-coupled estrogen receptor 1 on neurological disorders. Front Neuroendocrinol 2019; 55:100786. [PMID: 31513775 DOI: 10.1016/j.yfrne.2019.100786] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 02/06/2023]
Abstract
G protein-coupled estrogen receptor 1 (GPER) is a membrane-associated estrogen receptor (ER) associated with rapid estrogen-mediated effects. Over recent years GPER emerged has a potential therapeutic target to induce neuroprotection, avoiding the side effects elicited by the activation of classical ERs. The putative neuroprotection triggered by GPER selective activation was demonstrated in mood disorders, Alzheimer's disease or Parkinson's disease of male and female in vivo rodent models. In others, like ischemic stroke, the results are contradictory and currently there is no consensus on the role played by this receptor. However, it seems clear that sex is a biological variable that may impact the results. The major objective of this review is to provide an overview about the physiological effects of GPER in the brain and its putative contribution in neurodegenerative disorders, discussing the data about the signaling pathways involved, as well as, the diverse effects observed.
Collapse
Affiliation(s)
- C Roque
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - J Mendes-Oliveira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - C Duarte-Chendo
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - G Baltazar
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
11
|
Weidner K, Behnes M, Rusnak J, Schupp T, Hoppner J, Taton G, Reiser L, Bollow A, Reichelt T, Ellguth D, Engelke N, Kuche P, Ansari U, El‐Battrawy I, Lang S, Nienaber CA, Akin M, Mashayekhi K, Ferdinand D, Weiß C, Borggrefe M, Akin I. Male sex increases mortality in ventricular tachyarrhythmias. Intern Med J 2019; 49:711-721. [DOI: 10.1111/imj.14170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Kathrin Weidner
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Michael Behnes
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Jonas Rusnak
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Tobias Schupp
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Jorge Hoppner
- Clinic for Diagnostic and Interventional Radiology HeidelbergUniversity of Heidelberg Heidelberg Germany
| | - Gabriel Taton
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Linda Reiser
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Armin Bollow
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Thomas Reichelt
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Dominik Ellguth
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Niko Engelke
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Philipp Kuche
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Uzair Ansari
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Ibrahim El‐Battrawy
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Siegfried Lang
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | | | - Muharrem Akin
- Department of Cardiology and AngiologyHannover Medical School Hannover Germany
| | - Kambis Mashayekhi
- Department of Cardiology and Angiology IIUniversity Heart Center Freiburg Bad Krozingen Germany
| | - Dennis Ferdinand
- Institute of Biomathematics and Medical Statistics, Faculty of Medicine MannheimUniversity Medical Center Mannheim Germany
| | | | - Martin Borggrefe
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| | - Ibrahim Akin
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine MannheimUniversity of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim Mannheim Germany
| |
Collapse
|
12
|
Zheng S, Sun P, Liu H, Li R, Long L, Xu Y, Chen S, Xu J. 17β-estradiol upregulates striatin protein levels via Akt pathway in human umbilical vein endothelial cells. PLoS One 2018; 13:e0202500. [PMID: 30138337 PMCID: PMC6107185 DOI: 10.1371/journal.pone.0202500] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/04/2018] [Indexed: 11/19/2022] Open
Abstract
17β-estradiol (E2) has been shown to have beneficial effects on the cardiovascular system. We previously demonstrated that E2 increases striatin levels and inhibits migration in vascular smooth muscle cells. The objective of the present study was to investigate the effects of E2 on the regulation of striatin expression in human umbilical vein endothelial cells (HUVECs). We demonstrated that E2 increased striatin protein expression in a dose- and time-dependent manner in HUVECs. Pretreatment with ICI 182780 or the phosphatidylinositol-3 kinase inhibitor, wortmannin, abolished E2-mediated upregulation of striatin protein expression. Treatment with E2 resulted in Akt phosphorylation in a time-dependent manner. Moreover, silencing striatin significantly inhibited HUVEC migration, while striatin overexpression significantly promoted HUVEC migration. Finally, E2 enhanced HUVEC migration, which was inhibited by silencing striatin. In conclusion, our results demonstrated that E2-mediated upregulation of striatin promotes cell migration in HUVECs.
Collapse
Affiliation(s)
- Shuhui Zheng
- Research Center of Translational Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Peng Sun
- Department of Pathology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative innovation Center for Cancer Medicine, Guangzhou, China
| | - Haimei Liu
- Department of Physiology, Basic Medical College, Guangzhou University of Chinese Medicine, University Town, Guangzhou, China
| | - Runmei Li
- School of Chinese Pharmaceutical Science, Guangzhou University of Chinese Medicine, University Town, Guangzhou, China
| | - Lingli Long
- Research Center of Translational Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yuxia Xu
- Research Center of Translational Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Suiqing Chen
- Department of Physiology, Basic Medical College, Guangzhou University of Chinese Medicine, University Town, Guangzhou, China
| | - Jinwen Xu
- Department of Physiology, Basic Medical College, Guangzhou University of Chinese Medicine, University Town, Guangzhou, China
- * E-mail:
| |
Collapse
|
13
|
Memory-Related Synaptic Plasticity Is Sexually Dimorphic in Rodent Hippocampus. J Neurosci 2018; 38:7935-7951. [PMID: 30209204 DOI: 10.1523/jneurosci.0801-18.2018] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/22/2018] [Accepted: 07/15/2018] [Indexed: 12/22/2022] Open
Abstract
Men are generally superior to women in remembering spatial relationships, whereas the reverse holds for semantic information, but the neurobiological bases for these differences are not understood. Here we describe striking sexual dimorphism in synaptic mechanisms of memory encoding in hippocampal field CA1, a region critical for spatial learning. Studies of acute hippocampal slices from adult rats and mice show that for excitatory Schaffer-commissural projections, the memory-related long-term potentiation (LTP) effect depends upon endogenous estrogen and membrane estrogen receptor α (ERα) in females but not in males; there was no evident involvement of nuclear ERα in females, or of ERβ or GPER1 (G-protein-coupled estrogen receptor 1) in either sex. Quantitative immunofluorescence showed that stimulation-induced activation of two LTP-related kinases (Src, ERK1/2), and of postsynaptic TrkB, required ERα in females only, and that postsynaptic ERα levels are higher in females than in males. Several downstream signaling events involved in LTP were comparable between the sexes. In contrast to endogenous estrogen effects, infused estradiol facilitated LTP and synaptic signaling in females via both ERα and ERβ. The estrogen dependence of LTP in females was associated with a higher threshold for both inducing potentiation and acquiring spatial information. These results indicate that the observed sexual dimorphism in hippocampal LTP reflects differences in synaptic kinase activation, including both a weaker association with NMDA receptors and a greater ERα-mediated kinase activation in response to locally produced estrogen in females. We propose that male/female differences in mechanisms and threshold for field CA1 LTP contribute to differences in encoding specific types of memories.SIGNIFICANCE STATEMENT There is good evidence for male/female differences in memory-related cognitive function, but the neurobiological basis for this sexual dimorphism is not understood. Here we describe sex differences in synaptic function in a brain area that is critical for learning spatial cues. Our results show that female rodents have higher synaptic levels of estrogen receptor α (ERα) and, in contrast to males, require membrane ERα for the activation of signaling kinases that support long-term potentiation (LTP), a form of synaptic plasticity thought to underlie learning. The additional requirement of estrogen signaling in females resulted in a higher threshold for both LTP and hippocampal field CA1-dependent spatial learning. These results describe a synaptic basis for sexual dimorphism in encoding spatial information.
Collapse
|
14
|
Guivarc'h E, Buscato M, Guihot AL, Favre J, Vessières E, Grimaud L, Wakim J, Melhem NJ, Zahreddine R, Adlanmerini M, Loufrani L, Knauf C, Katzenellenbogen JA, Katzenellenbogen BS, Foidart JM, Gourdy P, Lenfant F, Arnal JF, Henrion D, Fontaine C. Predominant Role of Nuclear Versus Membrane Estrogen Receptor α in Arterial Protection: Implications for Estrogen Receptor α Modulation in Cardiovascular Prevention/Safety. J Am Heart Assoc 2018; 7:e008950. [PMID: 29959137 PMCID: PMC6064913 DOI: 10.1161/jaha.118.008950] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/20/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Although estrogen receptor α (ERα) acts primarily as a transcription factor, it can also elicit membrane-initiated steroid signaling. Pharmacological tools and transgenic mouse models previously highlighted the key role of ERα membrane-initiated steroid signaling in 2 actions of estrogens in the endothelium: increase in NO production and acceleration of reendothelialization. METHODS AND RESULTS Using mice with ERα mutated at cysteine 451 (ERaC451A), recognized as the key palmitoylation site required for ERα plasma membrane location, and mice with disruption of nuclear actions because of inactivation of activation function 2 (ERaAF20 = ERaAF2°), we sought to fully characterize the respective roles of nuclear versus membrane-initiated steroid signaling in the arterial protection conferred by ERα. ERaC451A mice were fully responsive to estrogens to prevent atheroma and angiotensin II-induced hypertension as well as to allow flow-mediated arteriolar remodeling. By contrast, ERαAF20 mice were unresponsive to estrogens for these beneficial vascular effects. Accordingly, selective activation of nuclear ERα with estetrol was able to prevent hypertension and to restore flow-mediated arteriolar remodeling. CONCLUSIONS Altogether, these results reveal an unexpected prominent role of nuclear ERα in the vasculoprotective action of estrogens with major implications in medicine, particularly for selective nuclear ERα agonist, such as estetrol, which is currently under development as a new oral contraceptive and for hormone replacement therapy in menopausal women.
Collapse
Affiliation(s)
- Emmanuel Guivarc'h
- From the institut des maladies des mitochondries, du coeur et des vaisseaux (MITOVASC) Institute, Cardiovascular Functions investigation (CARFI) Facility, Institut National de la Sante et de la Recherche Medicale (INSERM) U1083, Unité mixte de Recherche du Centre national de la recherche scientifique (UMR CNRS) 6015, University of Angers, France
| | - Mélissa Buscato
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France
| | - Anne-Laure Guihot
- From the institut des maladies des mitochondries, du coeur et des vaisseaux (MITOVASC) Institute, Cardiovascular Functions investigation (CARFI) Facility, Institut National de la Sante et de la Recherche Medicale (INSERM) U1083, Unité mixte de Recherche du Centre national de la recherche scientifique (UMR CNRS) 6015, University of Angers, France
| | - Julie Favre
- From the institut des maladies des mitochondries, du coeur et des vaisseaux (MITOVASC) Institute, Cardiovascular Functions investigation (CARFI) Facility, Institut National de la Sante et de la Recherche Medicale (INSERM) U1083, Unité mixte de Recherche du Centre national de la recherche scientifique (UMR CNRS) 6015, University of Angers, France
| | - Emilie Vessières
- From the institut des maladies des mitochondries, du coeur et des vaisseaux (MITOVASC) Institute, Cardiovascular Functions investigation (CARFI) Facility, Institut National de la Sante et de la Recherche Medicale (INSERM) U1083, Unité mixte de Recherche du Centre national de la recherche scientifique (UMR CNRS) 6015, University of Angers, France
| | - Linda Grimaud
- From the institut des maladies des mitochondries, du coeur et des vaisseaux (MITOVASC) Institute, Cardiovascular Functions investigation (CARFI) Facility, Institut National de la Sante et de la Recherche Medicale (INSERM) U1083, Unité mixte de Recherche du Centre national de la recherche scientifique (UMR CNRS) 6015, University of Angers, France
| | - Jamal Wakim
- From the institut des maladies des mitochondries, du coeur et des vaisseaux (MITOVASC) Institute, Cardiovascular Functions investigation (CARFI) Facility, Institut National de la Sante et de la Recherche Medicale (INSERM) U1083, Unité mixte de Recherche du Centre national de la recherche scientifique (UMR CNRS) 6015, University of Angers, France
| | - Nada-Joe Melhem
- From the institut des maladies des mitochondries, du coeur et des vaisseaux (MITOVASC) Institute, Cardiovascular Functions investigation (CARFI) Facility, Institut National de la Sante et de la Recherche Medicale (INSERM) U1083, Unité mixte de Recherche du Centre national de la recherche scientifique (UMR CNRS) 6015, University of Angers, France
| | - Rana Zahreddine
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France
| | - Marine Adlanmerini
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France
| | - Laurent Loufrani
- From the institut des maladies des mitochondries, du coeur et des vaisseaux (MITOVASC) Institute, Cardiovascular Functions investigation (CARFI) Facility, Institut National de la Sante et de la Recherche Medicale (INSERM) U1083, Unité mixte de Recherche du Centre national de la recherche scientifique (UMR CNRS) 6015, University of Angers, France
| | - Claude Knauf
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France
| | - John A Katzenellenbogen
- Department of Chemistry and Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Benita S Katzenellenbogen
- Department of Chemistry and Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Jean-Michel Foidart
- Groupe Interdisciplinaire de Génoprotéomique Appliquée, Université de Liège, Belgium
| | - Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France
| | - Françoise Lenfant
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France
| | - Jean-François Arnal
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France
| | - Daniel Henrion
- From the institut des maladies des mitochondries, du coeur et des vaisseaux (MITOVASC) Institute, Cardiovascular Functions investigation (CARFI) Facility, Institut National de la Sante et de la Recherche Medicale (INSERM) U1083, Unité mixte de Recherche du Centre national de la recherche scientifique (UMR CNRS) 6015, University of Angers, France
| | - Coralie Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, University of Toulouse 3, France
| |
Collapse
|
15
|
Pedersen AL, Saldanha CJ. Reciprocal interactions between prostaglandin E2- and estradiol-dependent signaling pathways in the injured zebra finch brain. J Neuroinflammation 2017; 14:262. [PMID: 29284502 PMCID: PMC5747085 DOI: 10.1186/s12974-017-1040-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 12/12/2017] [Indexed: 01/19/2023] Open
Abstract
Background Astrocytic aromatization and consequent increases in estradiol are neuroprotective in the injured brain. In zebra finches, cyclooxygenase-activity is necessary for injury-induced aromatase expression, and increased central estradiol lowers neuroinflammation. The mechanisms underlying these influences are unknown. Here, we document injury-induced, cyclooxygenase-dependent increases in glial aromatase expression and replicate previous work in our lab showing increases in central prostaglandin E2 and estradiol following brain damage. Further, we describe injury-dependent changes in E-prostanoid and estrogen receptor expression and reveal the necessity of E-prostanoid and estrogen receptors in the injury-dependent, reciprocal interactions of neuroinflammatory and neurosteroidogenic pathways. Methods Adult male and female birds were shams or received bilateral injections of the appropriate drug or vehicle into contralateral telencephalic lobes. Results Injuries sustained in the presence of indomethacin (a cyclooxygenase inhibitor) had fewer aromatase-expressing reactive astrocytes relative to injuries injected with vehicle suggesting that cyclooxygenase activity is necessary for the induction of glial aromatase around the site of damage. Injured hemispheres had higher prostaglandin E2 and estradiol content relative to shams. Importantly, injured hemispheres injected with E-prostanoid- or estrogen receptor-antagonists showed elevated prostaglandin E2 and estradiol, respectively, but lower prostaglandin E2 or estradiol-dependent downstream activity (protein kinase A or phosphoinositide-3-kinase mRNA) suggesting that receptor antagonism did not affect injury-induced prostaglandin E2 or estradiol, but inhibited the effects of these ligands. Antagonism of E-prostanoid receptors 3 or 4 prevented injury-induced increases in neural estradiol in males and females, respectively, albeit this apparent sex-difference needs to be tested more stringently. Further, estrogen receptor-α, but not estrogen receptor-β antagonism, exaggerated neural prostaglandin E2 levels relative to the contralateral lobe in both sexes. Conclusion These data suggest injury-induced, sex-specific prostaglandin E2-dependent estradiol synthesis, and estrogen receptor-α dependent decreases in neuroinflammation in the vertebrate brain.
Collapse
Affiliation(s)
- Alyssa L Pedersen
- Department of Biology, Program in Behavior, Cognition and Neuroscience, and the Center for Behavioral Neuroscience, American University, 4400 Massachusetts Avenue NW, Washington, DC, 20016, USA
| | - Colin J Saldanha
- Department of Biology, Program in Behavior, Cognition and Neuroscience, and the Center for Behavioral Neuroscience, American University, 4400 Massachusetts Avenue NW, Washington, DC, 20016, USA.
| |
Collapse
|
16
|
Kalo D, Roth Z. Low level of mono(2-ethylhexyl) phthalate reduces oocyte developmental competence in association with impaired gene expression. Toxicology 2016; 377:38-48. [PMID: 27989758 DOI: 10.1016/j.tox.2016.12.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 12/24/2022]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) and its metabolite, mono-(2-ethylhexyl) phthalate (MEHP), are reproductive toxicants. However, disruptive effects of MEHP at low concentrations on the oocyte and developing blastocyst are unknown. Previously, we detected low levels of MEHP in follicular fluid aspirated from DEHP-treated cows associated with reduced estradiol levels. Moreover, the MEHP concentrations found were similar to those reported for follicular fluid aspirated from women who have undergone IVF cycles. In the current study, we used an in vitro embryo production model to examine the effect of MEHP at low levels on oocyte developmental competence. We set up several experiments to mimic the follicular fluid content, i.e., low MEHP level and low estradiol. For all experiments, cumulus oocyte complexes (COCs) were aspirated from bovine ovaries, then matured in vitro in standard oocyte maturation medium (OMM) supplemented with: MEHP at a range levels (20-1000nM) or with estradiol at a range levels (0-2000ng/ml). Then, oocytes were fertilized and cultured for an additional 7days to allow blastocyst development. Findings revealed that MEHP at low levels impairs oocyte developmental competence in a dose-dependent manner (P<0.05) and that estradiol by itself does not impair it. Accordingly, in another set of experiments, COCs were matured in vitro with MEHP at two choosen concentrations (20 or 1000nM) with or without estradiol, fertilized and cultured for 7days. Samples of mature oocytes and their derived blastocysts were subjected to quantitative real-time PCR to examine the profiles of selected genes (CYC1, MT-CO1, ATP5B, POU5F1, SOX2 and DNMT3b). Maturation of COCs with MEHP (20 or 1000nM) affected gene expression in the mature oocyte. Maturation of COCs with MEHP (20 or 1000nM) in the absence of estradiol reduced oocyte developmental competence (P<0.05). A differential carryover effect on transcript abundance was recorded in blastocysts developed from MEHP-treated oocytes. In the presence of estradiol, increased expression was recorded for CYC1, ATP5B, SOX2 and DNMT3b. In the absence of estradiol, decreased expression was recorded, with a significant effect for 1000nM MEHP (P<0.05). Taken together, the findings suggest that low levels of phthalate must be taken into consideration in risk assessments.
Collapse
Affiliation(s)
- D Kalo
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel; Center of Excellence in Agriculture and Environmental Health, The Hebrew University, Rehovot 76100, Israel
| | - Z Roth
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel; Center of Excellence in Agriculture and Environmental Health, The Hebrew University, Rehovot 76100, Israel.
| |
Collapse
|
17
|
L-Type Calcium Channels Modulation by Estradiol. Mol Neurobiol 2016; 54:4996-5007. [PMID: 27525676 DOI: 10.1007/s12035-016-0045-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 08/08/2016] [Indexed: 01/29/2023]
Abstract
Voltage-gated calcium channels are key regulators of brain function, and their dysfunction has been associated with multiple conditions and neurodegenerative diseases because they couple membrane depolarization to the influx of calcium-and other processes such as gene expression-in excitable cells. L-type calcium channels, one of the three major classes and probably the best characterized of the voltage-gated calcium channels, act as an essential calcium binding proteins with a significant biological relevance. It is well known that estradiol can activate rapidly brain signaling pathways and modulatory/regulatory proteins through non-genomic (or non-transcriptional) mechanisms, which lead to an increase of intracellular calcium that activate multiple kinases and signaling cascades, in the same way as L-type calcium channels responses. In this context, estrogens-L-type calcium channels signaling raises intracellular calcium levels and activates the same signaling cascades in the brain probably through estrogen receptor-independent modulatory mechanisms. In this review, we discuss the available literature on this area, which seems to suggest that estradiol exerts dual effects/modulation on these channels in a concentration-dependent manner (as a potentiator of these channels in pM concentrations and as an inhibitor in nM concentrations). Indeed, estradiol may orchestrate multiple neurotrophic responses, which open a new avenue for the development of novel estrogen-based therapies to alleviate different neuropathologies. We also highlight that it is essential to determine through computational and/or experimental approaches the interaction between estradiol and L-type calcium channels to assist these developments, which is an interesting area of research that deserves a closer look in future biomedical research.
Collapse
|
18
|
Seleit I, Bakry OA, El Repey HS, Ali R. Intrinsic versus Extrinsic Aging: A Histopathological, Morphometric and Immunohistochemical Study of Estrogen Receptor β and Androgen Receptor. Skin Pharmacol Physiol 2016; 29:178-89. [PMID: 27399919 DOI: 10.1159/000446662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/28/2016] [Indexed: 11/19/2022]
Abstract
Skin is a target organ of sex steroids which play important roles in skin health and disease. The aim of this study is to investigate the expression of estrogen receptor β (ERβ) and androgen receptor (AR) in human skin from different age groups for a better understanding of the hormonal regulation of skin aging. Using standard immunohistochemical techniques, biopsies of sun-unprotected and sun-protected skin were taken from 60 normal subjects. Sun-protected skin showed significantly higher immunoreactivity for ERβ and AR compared to sun-unprotected skin of all age groups. Significantly higher ERβ H score and percent of expression were associated with the 20-35 years age group compared to the groups that were 35-50 years and >50 years old (p < 0.02, p = 0.03, respectively) in sun-unprotected and sun-protected skin (p < 0.001, p = 0.01, respectively). AR H score showed a negative correlation with age (p = 0.04) with no significant difference in immunoreactivity in different age groups, either in sun-unprotected or sun-protected skin. There was also a significant correlation between ERβ H score and epidermal thickness in sun-unprotected (p = 0.04) and sun-protected skin (p = 0.04) in studied subjects regardless of age. The same relationships did not reach significance with AR expression. However, a significant positive correlation was detected between H scores and percent of expression of ERβ and AR in sun-unprotected (p = 0.01, p = 0.02, respectively) and sun-protected skin (p = 0.005, p = 0.02, respectively) regardless of age. In conclusion, both ERβ and AR decline gradually with intrinsic and extrinsic aging. This decline is more obvious with extrinsic aging. Further large-scaled studies are recommended to expand, validate and translate current findings to clinically significant, diagnostic and therapeutic applications. Molecular studies to investigate the probable ligand-independent action of both receptors are warranted. In addition, their gene expression patterns and associated signaling and metabolic pathways can also be tackled to provide a basis for further interventions in pathological processes that involve their dysregulation.
Collapse
Affiliation(s)
- Iman Seleit
- Department of Dermatology, Andrology and STDs, Faculty of Medicine, Menoufiya University, Shibin El Koom, Egypt
| | | | | | | |
Collapse
|
19
|
Rossetti MF, Cambiasso MJ, Holschbach MA, Cabrera R. Oestrogens and Progestagens: Synthesis and Action in the Brain. J Neuroendocrinol 2016; 28. [PMID: 27306650 DOI: 10.1111/jne.12402] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/14/2016] [Accepted: 06/14/2016] [Indexed: 12/25/2022]
Abstract
When steroids, such as pregnenolone, progesterone and oestrogen, are synthesised de novo in neural tissues, they are more specifically referred to as neurosteroids. These neurosteroids bind specific receptors to promote essential brain functions. Pregnenolone supports cognition and protects mouse hippocampal cells against glutamate and amyloid peptide-induced cell death. Progesterone promotes myelination, spinogenesis, synaptogenesis, neuronal survival and dendritic growth. Allopregnanolone increases hippocampal neurogenesis, neuronal survival and cognitive functions. Oestrogens, such as oestradiol, regulate synaptic plasticity, reproductive behaviour, aggressive behaviour and learning. In addition, neurosteroids are neuroprotective in animal models of Alzheimer's disease, Parkinson's disease, brain injury and ageing. Using in situ hybridisation and/or immunohistochemistry, steroidogenic enzymes, including cytochrome P450 side-chain cleavage, 3β-hydroxysteroid dehydrogenase/Δ5-Δ4 isomerase, cytochrome P450arom, steroid 5α-reductase and 3α-hydroxysteroid dehydrogenase, have been detected in numerous brain regions, including the hippocampus, hypothalamus and cerebral cortex. In the present review, we summarise some of the studies related to the synthesis and function of oestrogens and progestagens in the central nervous system.
Collapse
Affiliation(s)
- M F Rossetti
- Departamento de Bioquímica Clínica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
- Instituto de Salud y Ambiente del Litoral, CONICET-Universidad Nacional del Litoral, Santa Fe, Argentina
| | - M J Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M A Holschbach
- Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - R Cabrera
- Instituto de Investigaciones Biomédicas, INBIOMED-IMBECU-CONICET, Universidad de Mendoza, Mendoza, Argentina
| |
Collapse
|
20
|
Selina AK, Murat K, Emre K, Ismail M, Bunyami U, Cemal G. The relationship between estrogen receptors and microtubule dynamics in post-menopausal rat brain. Acta Histochem 2015; 117:747-51. [PMID: 26319549 DOI: 10.1016/j.acthis.2015.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/16/2015] [Accepted: 08/19/2015] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Estrogen is one of the most important regulators of neuron function. There is a broad consensus that a loss of estrogen is associated with neurodegeneration in the hippocampus which leads to cognitive impairment. Hematopoietic-Pbx-interaction-protein (HPIP) is a novel scaffolding protein which interacts with microtubules and estrogen receptors. In this study, we investigated the presence and role of HPIP in hippocampal neurons and examined the relationship between estrogen receptors and microtubule damage in post-menopausal rat brains. METHOD Eighty female Wistar albino rats, 12 weeks old, were divided into 10 groups: control, control+17-β-estradiol, control+tamoxifen, control+mitogen-activated protein kinases (MAPK) inhibitor, control+phosphoinositide 3-kinase (PI3-K) inhibitor, ovariectomized, ovariectomized+17-β-estradiol, ovariectomized+tamoxifen, ovariectomized+MAPK inhibitor, and ovariectomized+PI3-K inhibitor. Light and electron microscopic examinations were performed. Real-time polymerase chain reaction (PCR) was used to determine the expression level of HPIP in experimental groups. RESULTS Light and electron microscopic examinations revealed morphological changes in hippocampal neuron axons. Axonal fluctuations and shrinkage were detected in all ovariectomized groups. HPIP was detected in all neurons with difference expression levels. CONCLUSION Proof that the HPIP protein can be found on hippocampal neurons may give rise to a new focus on neurodegeneration in post-menopausal women. Future molecular and pharmacological studies should be performed to reduce the rate of cognitive symptoms resulting from hippocampal neurodegeneration.
Collapse
|
21
|
Almey A, Milner TA, Brake WG. Estrogen receptors in the central nervous system and their implication for dopamine-dependent cognition in females. Horm Behav 2015; 74:125-38. [PMID: 26122294 PMCID: PMC4820286 DOI: 10.1016/j.yhbeh.2015.06.010] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 12/12/2022]
Abstract
This article is part of a Special Issue "Estradiol and cognition". Over the past 30 years, research has demonstrated that estrogens not only are important for female reproduction, but also play a role in a diverse array of cognitive functions. Originally, estrogens were thought to have only one receptor, localized exclusively to the cytoplasm and nucleus of cells. However, it is now known that there are at least three estrogen receptors (ERs): ERα, ERβ and G-protein coupled ER1 (GPER1). In addition to being localized to nuclei, ERα and ERβ are localized to the cell membrane, and GPER1 is also observed at the cell membrane. The mechanism through which ERs are associated with the membrane remains unclear, but palmitoylation of receptors and associations between ERs and caveolin are implicated in membrane association. ERα and ERβ are mostly observed in the nucleus using light microscopy unless they are particularly abundant. However, electron microscopy has revealed that ERs are also found at the membrane in complimentary distributions in multiple brain regions, many of which are innervated by dopamine inputs and were previously thought to contain few ERs. In particular, membrane-associated ERs are observed in the prefrontal cortex, dorsal striatum, nucleus accumbens, and hippocampus, all of which are involved in learning and memory. These findings provide a mechanism for the rapid effects of estrogens in these regions. The effects of estrogens on dopamine-dependent cognition likely result from binding at both nuclear and membrane-associated ERs, so elucidating the localization of membrane-associated ERs helps provide a more complete understanding of the cognitive effects of these hormones.
Collapse
Affiliation(s)
- Anne Almey
- Centre for Studies in Behavioral Neurobiology (CSBN), Department of Psychology, Concordia University, Montreal, QC, Canada.
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY USA; Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA.
| | - Wayne G Brake
- Centre for Studies in Behavioral Neurobiology (CSBN), Department of Psychology, Concordia University, Montreal, QC, Canada.
| |
Collapse
|
22
|
Oróstica ML, Lopez J, Rojas I, Rocco J, Díaz P, Reuquén P, Cardenas H, Parada-Bustamante A, Orihuela PA. Estradiol increases cAMP in the oviductal secretory cells through a nongenomic mechanism. Reproduction 2015; 148:285-94. [PMID: 25038866 DOI: 10.1530/rep-14-0128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the rat oviduct, estradiol (E2) accelerates egg transport by a nongenomic action that requires previous conversion of E2 to methoxyestrogens via catechol-O-methyltranferase (COMT) and activation of estrogen receptor (ER) with subsequent production of cAMP and inositol triphosphate (IP3). However, the role of the different oviductal cellular phenotypes on this E2 nongenomic pathway remains undetermined. The aim of this study was to investigate the effect of E2 on the levels of cAMP and IP3 in primary cultures of secretory and smooth muscle cells from rat oviducts and determine the mechanism by which E2 increases cAMP in the secretory cells. In the secretory cells, E2 increased cAMP but not IP3, while in the smooth muscle cells E2 decreased cAMP and increased IP3. Suppression of protein synthesis by actinomycin D did not prevent the E2-induced cAMP increase, but this was blocked by the ER antagonist ICI 182 780 and the inhibitors of COMT OR 486, G protein-α inhibitory (Gαi) protein pertussis toxin and adenylyl cyclase (AC) SQ 22536. Expression of the mRNA for the enzymes that metabolizes estrogens, Comt, Cyp1a1, and Cyp1b1 was found in the secretory cells, but this was not affected by E2. Finally, confocal immunofluorescence analysis showed that E2 induced colocalization between ESR1 (ERα) and Gαi in extranuclear regions of the secretory cells. We conclude that E2 differentially regulates cAMP and IP3 in the secretory and smooth muscle cells of the rat oviduct. In the secretory cells, E2 increases cAMP via a nongenomic action that requires activation of COMT and ER, coupling between ESR1 and Gαi, and stimulation of AC.
Collapse
Affiliation(s)
- María L Oróstica
- Laboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, ChileLaboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, Chile
| | - John Lopez
- Laboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, ChileLaboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, Chile
| | - Israel Rojas
- Laboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, ChileLaboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, Chile
| | - Jocelyn Rocco
- Laboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, ChileLaboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, Chile
| | - Patricia Díaz
- Laboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, ChileLaboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, Chile
| | - Patricia Reuquén
- Laboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, ChileLaboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, Chile
| | - Hugo Cardenas
- Laboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, ChileLaboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, Chile
| | - Alexis Parada-Bustamante
- Laboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, Chile
| | - Pedro A Orihuela
- Laboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, ChileLaboratorio de Inmunología de la ReproducciónFacultad de Química y Biología, Universidad de Santiago de Chile, Alameda 3363, Casilla 40, Correo 33, Santiago, ChileCentro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNASantiago, ChileInstituto de Investigaciones Materno-InfantilUniversidad de Chile, Santiago, Chile
| |
Collapse
|
23
|
Montani C, Steimberg N, Boniotti J, Biasiotto G, Zanella I, Diafera G, Biunno I, Caimi L, Mazzoleni G, Di Lorenzo D. Fibroblasts maintained in 3 dimensions show a better differentiation state and higher sensitivity to estrogens. Toxicol Appl Pharmacol 2014; 280:421-33. [DOI: 10.1016/j.taap.2014.08.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 07/24/2014] [Accepted: 08/12/2014] [Indexed: 01/07/2023]
|
24
|
Rational modification of estrogen receptor by combination of computational and experimental analysis. PLoS One 2014; 9:e102658. [PMID: 25075862 PMCID: PMC4116177 DOI: 10.1371/journal.pone.0102658] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/21/2014] [Indexed: 11/19/2022] Open
Abstract
In this manuscript, we modulate the binding properties of estrogen receptor protein by rationally modifying the amino acid composition of its ligand binding domain. By combining sequence alignment and structural analysis of known estrogen receptor-ligand complexes with computational analysis, we were able to predict estrogen receptor mutants with altered binding properties. These predictions were experimentally confirmed by producing single point variants with up to an order of magnitude increased binding affinity towards some estrogen disrupting chemicals and reaching an half maximal inhibitory concentration (IC50) value of 2 nM for the 17α-ethinylestradiol ligand. Due to increased affinity and stability, utilizing such mutated estrogen receptor instead of the wild type as bio-recognition element would be beneficial in an assay or biosensor.
Collapse
|
25
|
Arefin S, Simoncini T, Wieland R, Hammarqvist F, Spina S, Goglia L, Kublickiene K. Vasodilatory effects of the selective GPER agonist G-1 is maximal in arteries of postmenopausal women. Maturitas 2014; 78:123-30. [DOI: 10.1016/j.maturitas.2014.04.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 03/29/2014] [Accepted: 04/01/2014] [Indexed: 01/02/2023]
|
26
|
Kiyama R, Zhu Y. DNA microarray-based gene expression profiling of estrogenic chemicals. Cell Mol Life Sci 2014; 71:2065-82. [PMID: 24399289 PMCID: PMC11113397 DOI: 10.1007/s00018-013-1544-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/06/2013] [Accepted: 12/16/2013] [Indexed: 12/31/2022]
Abstract
We summarize updated information about DNA microarray-based gene expression profiling by focusing on its application to estrogenic chemicals. First, estrogenic chemicals, including natural/industrial estrogens and phytoestrogens, and the methods for detection and evaluation of estrogenic chemicals were overviewed along with a comprehensive list of estrogenic chemicals of natural or industrial origin. Second, gene expression profiling of chemicals using a focused microarray containing estrogen-responsive genes is summarized. Third, silent estrogens, a new type of estrogenic chemicals characterized by their estrogenic gene expression profiles without growth stimulative or inhibitory effects, have been identified so far exclusively by DNA microarray assay. Lastly, the prospect of a microarray assay is discussed, including issues such as commercialization, future directions of applications and quality control methods.
Collapse
Affiliation(s)
- Ryoiti Kiyama
- Signaling Molecules Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan,
| | | |
Collapse
|
27
|
Prossnitz ER, Barton M. Estrogen biology: new insights into GPER function and clinical opportunities. Mol Cell Endocrinol 2014; 389:71-83. [PMID: 24530924 PMCID: PMC4040308 DOI: 10.1016/j.mce.2014.02.002] [Citation(s) in RCA: 289] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 02/04/2014] [Indexed: 12/16/2022]
Abstract
Estrogens play an important role in the regulation of normal physiology, aging and many disease states. Although the nuclear estrogen receptors have classically been described to function as ligand-activated transcription factors mediating genomic effects in hormonally regulated tissues, more recent studies reveal that estrogens also mediate rapid signaling events traditionally associated with G protein-coupled receptors. The G protein-coupled estrogen receptor GPER (formerly GPR30) has now become recognized as a major mediator of estrogen's rapid cellular effects throughout the body. With the discovery of selective synthetic ligands for GPER, both agonists and antagonists, as well as the use of GPER knockout mice, significant advances have been made in our understanding of GPER function at the cellular, tissue and organismal levels. In many instances, the protective/beneficial effects of estrogen are mimicked by selective GPER agonism and are absent or reduced in GPER knockout mice, suggesting an essential or at least parallel role for GPER in the actions of estrogen. In this review, we will discuss recent advances and our current understanding of the role of GPER and the activity of clinically used drugs, such as SERMs and SERDs, in physiology and disease. We will also highlight novel opportunities for clinical development towards GPER-targeted therapeutics, for molecular imaging, as well as for theranostic approaches and personalized medicine.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Cell Biology and Physiology, UNM Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87120, USA.
| | - Matthias Barton
- Molecular Internal Medicine, University of Zurich, Switzerland.
| |
Collapse
|
28
|
Bakry OA, Samaka RM, Shoeib MAM, Maher A. Immunolocalization of androgen receptor and estrogen receptors in skin tags. Ultrastruct Pathol 2014; 38:344-57. [PMID: 24830664 DOI: 10.3109/01913123.2014.911788] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Skin tags (STs) are benign connective tissue tumors of the dermis. Several clinical observations suggested the involvement of sex steroids in their development. This study aimed at investigating the possible role of androgen receptor (AR) and estrogen receptors (ERs) in STs pathogenesis through their immunohistochemical (IHC) localization in skin biopsies of this disease and to correlate their expression with different clinical and histopathological parameters. Using IHC techniques, we examined 62 cases with STs and 30 gender- and age-matched, healthy subjects, representing the control group. ERα, ERβ, and AR were upregulated in STs compared to normal skin in epidermis and dermis (p < .001 for all). Higher AR H score was significantly associated with axillary STs (p = .02), skin colored tags (p = .03), acanthosis, and papillomatosis (p = .04 for both). Higher ERα H score was significantly associated with hyperpigmented tags (p < .001) and positive family history (p = .003). Higher ERβ H score was significantly associated with female gender and obesity (p = .004 for both). Higher ERα and AR H scores were significantly associated with loose collagen arrangement (p = .02, p = .004, respectively). Higher AR, ERα, and ERβ H scores were significantly associated with the presence of mast cells (p = .01, p = .04, p = .002, respectively) and dilated blood vessels (p = .006, p = .04, p = .04, respectively). In conclusion, AR and ERs may share in STs pathogenesis through their effect on keratinocytes, fibroblasts, and mast cells.
Collapse
|
29
|
Stournaras C, Gravanis A, Margioris AN, Lang F. The actin cytoskeleton in rapid steroid hormone actions. Cytoskeleton (Hoboken) 2014; 71:285-93. [DOI: 10.1002/cm.21172] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/20/2014] [Accepted: 02/26/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Christos Stournaras
- Department of Biochemistry; University of Crete Medical School; Heraklion Greece
- Department of Physiology; University of Tübingen; Tübingen Germany
| | - Achilles Gravanis
- Department of Pharmacology; University of Crete Medical School; Heraklion Greece
| | - Andrew N. Margioris
- Department of Clinical Chemistry; University of Crete Medical School; Heraklion Greece
| | - Florian Lang
- Department of Physiology; University of Tübingen; Tübingen Germany
| |
Collapse
|
30
|
Actions of 17β-estradiol and testosterone in the mitochondria and their implications in aging. Ageing Res Rev 2013; 12:907-17. [PMID: 24041489 DOI: 10.1016/j.arr.2013.09.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 09/06/2013] [Indexed: 02/02/2023]
Abstract
A decline in the mitochondrial functions and aging are two closely related processes. The presence of estrogen and androgen receptors and hormone-responsive elements in the mitochondria represents the starting point for the investigation of the effects of 17β-estradiol and testosterone on the mitochondrial functions and their relationships with aging. Both steroids trigger a complex molecular mechanism that involves crosstalk between the mitochondria, nucleus, and plasma membrane, and the cytoskeleton plays a key role in these interactions. The result of this signaling is mitochondrial protection. Therefore, the molecular components of the pathways activated by the sexual steroids could represent targets for anti-aging therapies. In this review, we discuss previous studies that describe the estrogen- and testosterone-dependent actions on the mitochondrial processes implicated in aging.
Collapse
|
31
|
Jiang K, Yang Z, Cheng L, Wang S, Ning K, Zhou L, Lin J, Zhong H, Wang L, Li Y, Huang J, Zhang H, Ye Q. Mediator of ERBB2-driven cell motility (MEMO) promotes extranuclear estrogen receptor signaling involving the growth factor receptors IGF1R and ERBB2. J Biol Chem 2013; 288:24590-9. [PMID: 23861392 DOI: 10.1074/jbc.m113.467837] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In addition to nuclear estrogen receptor (ER) acting as a transcription factor, extranuclear ER also plays an important role in cancer cell growth regulation through activation of kinase cascades. However, the molecular mechanisms by which extranuclear ER exerts its function are still poorly understood. Here, we report that mediator of ERBB2-driven cell motility (MEMO) regulates extranuclear functions of ER. MEMO physically and functionally interacted with ER. Through its interaction with the growth factor receptors IGF1R and ERBB2, MEMO mediated extranuclear functions of ER, including activation of mitogen-activated protein kinase (MAPK) and protein kinase B/AKT, two important growth regulatory protein kinases, and integration of function with nuclear ER. Activation of MAPK and AKT was responsible for MEMO modulation of ER phosphorylation and estrogen-responsive gene expression. Moreover, MEMO increased anchorage-dependent and -independent growth of ER-positive breast cancer cells in vitro and was required for estrogen-induced breast tumor growth in nude mice. Together, our studies identified MEMO as a new component of extranuclear ER signalosome and suggest an essential role for MEMO in the regulation of ER-positive breast cancer cell growth.
Collapse
Affiliation(s)
- Kai Jiang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Tamoxifen Elicits Atheroprotection through Estrogen Receptor α AF-1 But Does Not Accelerate Reendothelialization. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:304-12. [DOI: 10.1016/j.ajpath.2013.03.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/18/2013] [Accepted: 03/07/2013] [Indexed: 01/01/2023]
|
33
|
Zhou K, Gao Q, Zheng S, Pan S, Li P, Suo K, Simoncini T, Wang T, Fu X. 17β-estradiol induces vasorelaxation by stimulating endothelial hydrogen sulfide release. Mol Hum Reprod 2012; 19:169-76. [PMID: 23041593 DOI: 10.1093/molehr/gas044] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Estrogen exerts vascular protective effects, but the underlying mechanisms remain to be understood fully. In recent years, hydrogen sulfide (H(2)S) has increasingly been recognized as an important signaling molecule in the cardiovascular system. Vascular H(2)S is produced from L-cysteine, catalyzed by cystathionine γ-lyase (CSE). In our study, apolipoprotein E (ApoE)-deficient mice were ovariectomized and implanted with placebo (OVX mice) or 17β-estradiol (E(2)) pellets (OVX + E(2) mice). Compared with OVX mice, OVX + E(2) mice showed increased plasma H(2)S levels (P = 0.012) and decreased aortic lesion area (P = 0.028). These effects were largely reversed when supplementing with the irreversible CSE inhibitor DL-propargylglycine (PPG) in the OVX + E(2) + PPG mice. Meanwhile, the nitric oxide and prostacyclin-resistant responses to cumulative application of acetylcholine (ACh) were studied among all the three groups of femoral arteries. Compared with the arteries in the OVX group, the vasodilator sensitivity of arteries to ACh was increased in the OVX + E(2) group and attenuated in the OVX + E(2) + PPG group. E(2) and estrogen receptor (ER) α agonist 4',4″,4'″-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol rapidly increased H(2)S release in human endothelial cells, but not partially selective ERβ agonist 2,3-bis-(4-hydroxyphenyl)-propionitrile. These effects were inhibited by ER antagonist ICI 182780 or by protein kinase G (PKG) inhibitor KT5823. Furthermore, endothelial PKG activity was increased by E(2) (P = 0.003) and E(2)-induced vasodilation was inhibited by KT5823 (P = 0.009). In conclusion, the endothelial CSE/H(2)S pathway is activated by E(2) through PKG, which leads to vasodilation. These actions may be relevant to estrogen's anti-atherogenic effect.
Collapse
Affiliation(s)
- Kewen Zhou
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Vassalle C, Simoncini T, Chedraui P, Pérez-López FR. Why sex matters: the biological mechanisms of cardiovascular disease. Gynecol Endocrinol 2012; 28:746-51. [PMID: 22329808 DOI: 10.3109/09513590.2011.652720] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading determinant of mortality and morbidity in women. However, a full understanding of the basic and clinical aspects of CVD in women is far from being accomplished. Sexual dimorphism in CVD has been reported both in humans and experimental animals. Menopause is a risk factor for CVD due to the reduction of endogenous estrogen, although the mechanisms underlying are poorly understood. Estrogens act through binding to vascular estrogen receptors and by non-genomic mechanisms. Advances in this field are essential to improve CVD diagnostic and clinical strategies in women, and to develop sex-specific prevention plans as much as female-oriented treatment algorithms. This paper reviews pathophysiology of CVD in women and its potential clinical implications. Particular emphasis is given to biochemical markers and to indicators of cardiovascular dysfunction and damage. Estimation of these parameters, central to cardiovascular pathophysiology, could represent a particularly relevant tool in female patients. More research is needed to identify women who will profit most of early intervention.
Collapse
Affiliation(s)
- Cristina Vassalle
- G. Monasterio Foundation of the Toscana Region and Institute of Clinical Physiology-CNR, Pisa, Italy
| | | | | | | |
Collapse
|
35
|
Zheng S, Huang J, Zhou K, Xiang Q, Zhang Y, Tan Z, Simoncini T, Fu X, Wang T. Progesterone enhances vascular endothelial cell migration via activation of focal adhesion kinase. J Cell Mol Med 2012; 16:296-305. [PMID: 21418517 PMCID: PMC3823293 DOI: 10.1111/j.1582-4934.2011.01305.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 03/01/2011] [Indexed: 11/28/2022] Open
Abstract
The mechanisms of progesterone on endothelial cell motility are poorly investigated. Previously we showed that progesterone stimulated endothelial cell migration via the activation of actin-binding protein moesin, leading to actin cytoskeleton remodelling and the formation of cell membrane structures required for cell movement. In this study, we investigated the effects of progesterone on the formation of focal adhesion complexes, which provide anchoring sites for cell movement. In cultured human umbilical endothelial cells, progesterone enhanced focal adhesion kinase (FAK) phosphorylation at Tyr(397) in a dose- and time-dependent manner. Several signalling inhibitors interfered with progesterone-induced FAK activation, including progesterone receptor (PR) antagonist ORG 31710, specific c-Src kinase inhibitor PP2, phosphatidylinosital-3 kinase (PI3K) inhibitor wortmannin as well as ρ-associated kinase (ROCK-2) inhibitor Y27632. It suggested that PR, c-Src, PI3K and ROCK-2 are implicated in this action. In line with this, we found that progesterone rapidly promoted c-Src/PI3K/Akt activity, which activated the small GTPase RhoA/ρ-associated kinase (ROCK-2) complex, resulting in FAK phosphorylation. In the presence of progesterone, endothelial cells displayed enhanced horizontal migration, which was reversed by small interfering RNAs abrogating FAK expression. In conclusion, progesterone promotes endothelial cell movement via the rapid regulation of FAK. These findings provide new information on the biological actions of progesterone on human endothelial cells that are relevant for vascular function.
Collapse
Affiliation(s)
- Shuhui Zheng
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Jinghe Huang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Kewen Zhou
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Qiuling Xiang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Yaxing Zhang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Zhi Tan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Tommaso Simoncini
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of PisaPisa, Italy
| | - Xiaodong Fu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Tinghuai Wang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
36
|
Sanchez AM, Flamini MI, Polak K, Palla G, Spina S, Mannella P, Genazzani AD, Simoncini T. Actin cytoskeleton remodelling by sex steroids in neurones. J Neuroendocrinol 2012; 24:195-201. [PMID: 22103470 DOI: 10.1111/j.1365-2826.2011.02258.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cell morphology and its interaction with the extracellular environment are integrated processes involving a number of intracellular controllers orchestrating cytoskeletal proteins and their interaction with the cell membrane and anchorage proteins. Sex steroids are effective regulators of cell morphology and tissue organisation, and recent evidence indicates that this is obtained through the regulation of the actin cytoskeleton. Intriguingly, many of these regulatory actions related to cell morphology are achieved through the rapid, nonclassical signalling of sex steroid receptors to kinase cascades, independently from nuclear alteration of gene expression or protein synthesis. The identification of the mechanistic basis for these rapid actions on cell cytoskeleton has special relevance for the characterisation of the effects of sex steroids under physiological conditions, such as for the development of neurone/neurone interconnections and dendritic spine density. This is considered to be critical for gender-specific differences in brain function and dysfunction. Recent advancements in the characterisation of the molecular basis of the extranuclear signalling of sex steroids help to clarify the role of oestrogen and progesterone in the brain, and may turn out to be of relevance for clinical purposes. This review highlights the regulatory effects of oestrogens and progesterone on actin cytoskeleton and neurone morphology, as well as recent progresses in the characterisation of these mechanisms, providing insights and working hypotheses on possible clinical applications for the modulation of these pathways in the central nervous system.
Collapse
Affiliation(s)
- A M Sanchez
- Institute of Medicine and Experimental Biology of Cuyo, CCT-CONICET Mendoza, National University of Cuyo, Parque General San Martin s/n, Mendoza, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Hofmeister MV, Damkier HH, Christensen BM, Olde B, Fredrik Leeb-Lundberg LM, Fenton RA, Praetorius HA, Praetorius J. 17β-Estradiol induces nongenomic effects in renal intercalated cells through G protein-coupled estrogen receptor 1. Am J Physiol Renal Physiol 2011; 302:F358-68. [PMID: 21993891 DOI: 10.1152/ajprenal.00343.2011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Steroid hormones such as 17β-estradiol (E2) are known to modulate ion transporter expression in the kidney through classic intracellular receptors. Steroid hormones are also known to cause rapid nongenomic responses in a variety of nonrenal tissues. However, little is known about renal short-term effects of steroid hormones. Here, we studied the acute actions of E2 on intracellular Ca(2+) signaling in isolated distal convoluted tubules (DCT2), connecting tubules (CNT), and initial cortical collecting ducts (iCCD) by fluo 4 fluorometry. Physiological concentrations of E2 induced transient increases in intracellular Ca(2+) concentration ([Ca(2+)](i)) in a subpopulation of cells. The [Ca(2+)](i) increases required extracellular Ca(2+) and were inhibited by Gd(3+). Strikingly, the classic E2 receptor antagonist ICI 182,780 also increased [Ca(2+)](i), which is inconsistent with the activation of classic E2 receptors. G protein-coupled estrogen receptor 1 (GPER1 or GPR30) was detected in microdissected DCT2/CNT/iCCD by RT-PCR. Stimulation with the specific GPER1 agonist G-1 induced similar [Ca(2+)](i) increases as E2, and in tubules from GPER1 knockout mice, E2, G-1, and ICI 182,780 failed to induce [Ca(2+)](i) elevations. The intercalated cells showed both E2-induced concanamycin-sensitive H(+)-ATPase activity by BCECF fluorometry and the E2-mediated [Ca(2+)](i) increment. We propose that E2 via GPER1 evokes [Ca(2+)](i) transients and increases H(+)-ATPase activity in intercalated cells in mouse DCT2/CNT/iCCD.
Collapse
|
38
|
Abstract
Aging of the skin is associated with skin thinning, atrophy, dryness, wrinkling, and delayed wound healing. These undesirable aging effects are exacerbated by declining estrogen levels in postmenopausal women. With the rise in interest in long-term postmenopausal skin management, studies on the restorative benefits that estrogen may have on aged skin have expanded. Systemic estrogen replacement therapy (ERT) has been shown to improve some aspects of skin. Estrogen restores skin thickness by increasing collagen synthesis while limiting excessive collagen degradation. Wrinkling is improved following estrogen treatment since estrogen enhances the morphology and synthesis of elastic fibers, collagen type III, and hyaluronic acids. Dryness is also alleviated through increased water-holding capacity, increased sebum production, and improved barrier function of the skin. Furthermore, estrogen modulates local inflammation, granulation, re-epithelialization, and possibly wound contraction, which collectively accelerates wound healing at the expense of forming lower quality scars. Despite its promises, long-term ERT has been associated with harmful systemic effects. In the search for safe and effective alternatives with more focused effects on the skin, topical estrogens, phytoestrogens, and tissue-specific drugs called selective estrogen receptor modulators (SERMs) have been explored. We discuss the promises and challenges of utilizing topical estrogens, SERMs, and phytoestrogens in postmenopausal skin management.
Collapse
|
39
|
Yan S, Chen Y, Dong M, Song W, Belcher SM, Wang HS. Bisphenol A and 17β-estradiol promote arrhythmia in the female heart via alteration of calcium handling. PLoS One 2011; 6:e25455. [PMID: 21980463 PMCID: PMC3181279 DOI: 10.1371/journal.pone.0025455] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 09/05/2011] [Indexed: 12/25/2022] Open
Abstract
Background There is wide-spread human exposure to bisphenol A (BPA), a ubiquitous estrogenic endocrine disruptor that has been implicated as having potentially harmful effects on human heart health. Higher urine BPA concentrations have been shown to be associated with cardiovascular diseases in humans. However, neither the nature nor the mechanism(s) of BPA action on the heart are understood. Methodology/Principal Findings The rapid (<7 min) effects of BPA and 17β-estradiol (E2) in the heart and ventricular myocytes from rodents were investigated in the present study. In isolated ventricular myocytes from young adult females, but not males, physiological concentrations of BPA or E2 (10−9 M) rapidly induced arrhythmogenic triggered activities. The effects of BPA were particularly pronounced when combined with estradiol. Under conditions of catecholamine stimulation, E2 and BPA promoted ventricular arrhythmias in female, but not male, hearts. The cellular mechanism of the female-specific pro-arrhythmic effects of BPA and E2 were investigated. Exposure to E2 and/or BPA rapidly altered myocyte Ca2+ handling; in particular, estrogens markedly increased sarcoplasmic reticulum (SR) Ca2+ leak, and increased SR Ca2+ load. Ryanodine (10−7 M) inhibition of SR Ca2+ leak suppressed estrogen-induced triggered activities. The rapid response of female myocytes to estrogens was abolished in an estrogen receptor (ER) β knockout mouse model. Conclusions/Significance Physiologically-relevant concentrations of BPA and E2 promote arrhythmias in a female-specific manner in rat hearts; the pro-arrhythmic actions of estrogens are mediated by ERβ-signaling through alterations of myocyte Ca2+ handling, particularly increases in SR Ca2+ leak. Our study provides the first experimental evidence suggesting that exposure to estrogenic endocrine disrupting chemicals and the unique sensitivity of female hearts to estrogens may play a role in arrhythmogenesis in the female heart.
Collapse
Affiliation(s)
- Sujuan Yan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Yamei Chen
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Min Dong
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Weizhong Song
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Scott M. Belcher
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Hong-Sheng Wang
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
40
|
Zheng S, Huang J, Zhou K, Zhang C, Xiang Q, Tan Z, Wang T, Fu X. 17β-Estradiol enhances breast cancer cell motility and invasion via extra-nuclear activation of actin-binding protein ezrin. PLoS One 2011; 6:e22439. [PMID: 21818323 PMCID: PMC3144228 DOI: 10.1371/journal.pone.0022439] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 06/27/2011] [Indexed: 12/21/2022] Open
Abstract
Estrogen promotes breast cancer metastasis. However, the detailed mechanism remains largely unknown. The actin binding protein ezrin is a key component in tumor metastasis and its over-expression is positively correlated to the poor outcome of breast cancer. In this study, we investigate the effects of 17β-estradiol (E2) on the activation of ezrin and its role in estrogen-dependent breast cancer cell movement. In T47-D breast cancer cells, E2 rapidly enhances ezrin phosphorylation at Thr567 in a time- and concentration-dependent manner. The signalling cascade implicated in this action involves estrogen receptor (ER) interaction with the non-receptor tyrosine kinase c-Src, which activates the phosphatidylinositol-3 kinase/Akt pathway and the small GTPase RhoA/Rho-associated kinase (ROCK-2) complex. E2 enhances the horizontal cell migration and invasion of T47-D breast cancer cells in three-dimensional matrices, which is reversed by transfection of cells with specific ezrin siRNAs. In conclusion, E2 promotes breast cancer cell movement and invasion by the activation of ezrin. These results provide novel insights into the effects of estrogen on breast cancer progression and highlight potential targets to treat endocrine-sensitive breast cancers.
Collapse
Affiliation(s)
- Shuhui Zheng
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinghe Huang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kewen Zhou
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chengxi Zhang
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiuling Xiang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi Tan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tinghuai Wang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XDF); (THW)
| | - Xiaodong Fu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XDF); (THW)
| |
Collapse
|
41
|
Danesh SM, Kundu P, Lu R, Stefani E, Toro L. Distinct transcriptional regulation of human large conductance voltage- and calcium-activated K+ channel gene (hSlo1) by activated estrogen receptor alpha and c-Src tyrosine kinase. J Biol Chem 2011; 286:31064-71. [PMID: 21757754 DOI: 10.1074/jbc.m111.235457] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Estrogen receptor α (ERα) regulates gene transcription via "genomic" (binding directly or indirectly, typically via Sp1 or AP-1 sites, to target genes) and/or "nongenomic" (signaling) mechanisms. ERα activation by estrogen up-regulates the murine Ca(2+)-activated K(+) channel α subunit gene (mSlo1) via genomic mechanisms. Here, we investigated whether ERα also drives transcription of the human (hSlo1) gene. Consistent with this view, estrogen increased hSlo1 transcript levels in primary human smooth muscle cells. Promoter studies revealed that estrogen/hERα-mediated hSlo1 transcription was nearly 6-fold more efficient than for mSlo1 (EC(50), 0.07 versus 0.4 nM). Unlike the genomic transcriptional mechanism employed by mSlo1, hSlo1 exhibits a nongenomic hERα-mediated regulatory mechanism. This is supported by the following: 1) efficient hSlo1 transcription after disruption of the DNA-binding domain of hERα or knockdown of Sp1, and 2) lack of AP-1 sites in the hSlo1 promoter. Three nongenomic signaling pathways were explored: Src, Rho, and PI3K. Inhibition of Src with 10 μM PP2, and reported downstream ERK with 25 μM PD98059 did not prevent estrogen action but caused an increase in hSlo1 basal transcription; conversely, constitutively active c-Src (Y527F) decreased hSlo1 basal transcription even preventing its estrogen/hERα-mediated transcriptional activation. Rho inhibition by coexpressed Clostridium botulinum C3 transferase did not alter estrogen action. In contrast, inhibition of PI3K activity with 10 μM LY294002 decreased estrogen-stimulated hSlo1 transcription by ∼40%. These results indicate that the nongenomic PI3K signaling pathway plays a role in estrogen/hERα-stimulated hSlo1 gene expression; whereas c-Src activity leads to hSlo1 gene tonic repression independently of estrogen, likely through ERK activation.
Collapse
Affiliation(s)
- Shahab M Danesh
- Division of Molecular Medicine, Department of Anesthesiology, UCLA, Los Angeles, California 90095-1778, USA
| | | | | | | | | |
Collapse
|
42
|
Koricanac G, Tepavcevic S, Zakula Z, Milosavljevic T, Stojiljkovic M, Isenovic ER. Interference between insulin and estradiol signaling pathways in the regulation of cardiac eNOS and Na+/K+-ATPase. Eur J Pharmacol 2011; 655:23-30. [DOI: 10.1016/j.ejphar.2011.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 12/10/2010] [Accepted: 01/07/2011] [Indexed: 11/16/2022]
|
43
|
Manning EE, Ransome MI, Burrows EL, Hannan AJ. Increased adult hippocampal neurogenesis and abnormal migration of adult-born granule neurons is associated with hippocampal-specific cognitive deficits in phospholipase C-β1 knockout mice. Hippocampus 2010; 22:309-19. [PMID: 21080410 DOI: 10.1002/hipo.20900] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2010] [Indexed: 12/12/2022]
Abstract
Schizophrenia is a devastating psychiatric illness with a complex pathophysiology. We have recently documented schizophrenia-like endophenotypes in phospholipase C-β1 knockout (PLC-β1(-/-)) mice, including deficits in prepulse inhibition, hyperlocomotion, and cognitive impairments. PLC-β1 signals via multiple G-protein coupled receptor pathways implicated in neural cellular plasticity; however, adult neurogenesis has yet to be explored in this knockout model. In this study, we employed PLC-β1(-/-) mice to elucidate possible correlates between aberrant adult hippocampal neurogenesis (AHN) and schizophrenia-like behaviors. Using stereology and bromodeoxyuridine (BrdU) immunohistochemistry we demonstrated a significant increase in the density of adult-generated cells in the granule cell layer (GCL) of adult PLC-β1(-/-) mice compared with wild-type littermates. Cellular phenotype analysis using confocal microscopy revealed these cells to be mature granule neurons expressing NeuN and calbindin. Increased neuronal survival occurred concomitant with reduced caspase-3(+) cells in the GCL of PLC-β1(-/-) mice. Stereological analysis of Ki67(+) cells in the subgranular zone suggested that neural precursor proliferation is unchanged in PLC-β1(-/-) mice. We further showed aberrant migration of mature granule neurons within the GCL of adult PLC-β1(-/-) mice with excessive adult-generated mature neurons residing in the middle and outer GCL. PLC-β1(-/-) mice exhibited specific behavioral deficits in location recognition, a measure of hippocampal-dependent memory, but not novel object recognition. Overall, we have shown that PLC-β1(-/-) mice have a threefold increase in net AHN, and have provided further evidence to suggest a specific deficit in hippocampal-dependent cognition. We propose that abnormal cellular plasticity in these mice may contribute to their schizophrenia-like behavioral endophenotypes.
Collapse
Affiliation(s)
- Elizabeth E Manning
- Howard Florey Institute, Florey Neuroscience Institutes, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | |
Collapse
|
44
|
Ali I, Penttinen-Damdimopoulou PE, Mäkelä SI, Berglund M, Stenius U, Åkesson A, Håkansson H, Halldin K. Estrogen-like effects of cadmium in vivo do not appear to be mediated via the classical estrogen receptor transcriptional pathway. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:1389-94. [PMID: 20525538 PMCID: PMC2957917 DOI: 10.1289/ehp.1001967] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 06/04/2010] [Indexed: 05/25/2023]
Abstract
BACKGROUND Cadmium (Cd), a ubiquitous food contaminant, has been proposed to be an endocrine disruptor by inducing estrogenic responses in vivo. Several in vitro studies suggested that these effects are mediated via estrogen receptors (ERs). OBJECTIVE We performed this study to clarify whether Cd-induced effects in vivo are mediated via classical ER signaling through estrogen responsive element (ERE)-regulated genes or if other signaling pathways are involved. METHODS We investigated the estrogenic effects of cadmium chloride (CdCl2) exposure in vivo by applying the Organisation for Economic Co-operation and Development (OECD) rodent uterotrophic bioassay to transgenic ERE-luciferase reporter mice. Immature female mice were injected subcutaneously with CdCl2 (5, 50, or 500 µg/kg body weight) or with 17α-ethinylestradiol (EE2) on 3 consecutive days. We examined uterine weight and histology, vaginal opening, body and organ weights, Cd tissue retention, activation of mitogen-activated protein kinase (MAPK) pathways, and ERE-dependent luciferase expression. RESULTS CdCl2 increased the height of the uterine luminal epithelium in a dose-dependent manner without increasing the uterine wet weight, altering the timing of vaginal opening, or affecting the luciferase activity in reproductive or nonreproductive organs. However, we observed changes in the phosphorylation of mouse double minute 2 oncoprotein (Mdm2) and extracellular signal-regulated kinase (Erk1/2) in the liver after CdCl2 exposure. As we expected, EE2 advanced vaginal opening and increased uterine epithelial height, uterine wet weight, and luciferase activity in various tissues. CONCLUSION Our data suggest that Cd exposure induces a limited spectrum of estrogenic responses in vivo and that, in certain targets, effects of Cd might not be mediated via classical ER signaling through ERE-regulated genes.
Collapse
Affiliation(s)
- Imran Ali
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Sari I. Mäkelä
- Functional Foods Forum and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Marika Berglund
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ulla Stenius
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Åkesson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Helen Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Krister Halldin
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
Sanchez AM, Simoncini T. Extra-nuclear signaling of ERalpha to the actin cytoskeleton in the central nervous system. Steroids 2010; 75:528-32. [PMID: 20018203 DOI: 10.1016/j.steroids.2009.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 11/30/2009] [Accepted: 12/02/2009] [Indexed: 11/19/2022]
Abstract
Cell morphology is controlled by a complex and redundant array of intracellular signaling pathways devoted to the regulation of the actin cytoskeleton and of its relationship with the cell membrane and the extracellular matrix. Sex steroids are effective regulators of cell morphology and tissue organization, and recent evidence indicates that this is obtained through the regulation of the cytoskeleton. Intriguingly, many of these regulatory actions related to cell morphology are achieved through rapid, non-classical signaling of sex steroid receptors to kinase cascades, independently from nuclear alteration of gene expression or protein synthesis. The identification of the mechanistic basis for these rapid actions on cell cytoskeleton has special relevance for the characterization of the effects of sex steroids in physiological conditions, such as their role in the control of brain cell remodeling. Brain cell morphology is controlled by estrogens that regulate the development of neuron/neuron interconnections and dendritic spine density. This is thought to be critical for gender-specific differences in brain function and dysfunction. The recent advancements in the characterization of the molecular basis of the extra-nuclear signaling of estrogen helps to understand the role of estrogen in the brain, and may in the future turn out to be of relevance for clinical purposes. This review highlights the regulatory effects on the cytoskeleton and cell morphology of estrogens as well as the recent advances in the characterization of these mechanisms, providing insights and working hypotheses on possible clinical applications for the modulation of these pathways in the central nervous system.
Collapse
Affiliation(s)
- Angel Matias Sanchez
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa, Pisa 56100, Italy
| | | |
Collapse
|
46
|
Goglia L, Tosi V, Sanchez AM, Flamini MI, Fu XD, Zullino S, Genazzani AR, Simoncini T. Endothelial regulation of eNOS, PAI-1 and t-PA by testosterone and dihydrotestosterone in vitro and in vivo. Mol Hum Reprod 2010; 16:761-9. [DOI: 10.1093/molehr/gaq049] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
47
|
Morkuniene R, Arandarcikaite O, Ivanoviene L, Borutaite V. Estradiol-induced protection against ischemia-induced heart mitochondrial damage and caspase activation is mediated by protein kinase G. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:1012-7. [DOI: 10.1016/j.bbabio.2010.03.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 03/28/2010] [Accepted: 03/29/2010] [Indexed: 10/19/2022]
|
48
|
Kang L, Zhang X, Xie Y, Tu Y, Wang D, Liu Z, Wang ZY. Involvement of estrogen receptor variant ER-alpha36, not GPR30, in nongenomic estrogen signaling. Mol Endocrinol 2010; 24:709-21. [PMID: 20197310 PMCID: PMC2852353 DOI: 10.1210/me.2009-0317] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 01/22/2010] [Indexed: 02/08/2023] Open
Abstract
Accumulating evidence suggested that an orphan G protein-coupled receptor (GPR)30, mediates nongenomic responses to estrogen. The present study was performed to investigate the molecular mechanisms underlying GPR30 function. We found that knockdown of GPR30 expression in breast cancer SK-BR-3 cells down-regulated the expression levels of estrogen receptor (ER)-alpha36, a variant of ER-alpha. Introduction of a GPR30 expression vector into GPR30 nonexpressing cells induced endogenous ER-alpha36 expression, and cotransfection assay demonstrated that GPR30 activated the promoter activity of ER-alpha36 via an activator protein 1 binding site. Both 17beta-estradiol (E2) and G1, a compound reported to be a selective GPR30 agonist, increased the phosphorylation levels of the MAPK/ERK1/2 in SK-BR-3 cells, which could be blocked by an anti-ER-alpha36-specific antibody against its ligand-binding domain. G1 induced activities mediated by ER-alpha36, such as transcription activation activity of a VP16-ER-alpha36 fusion protein and activation of the MAPK/ERK1/2 in ER-alpha36-expressing cells. ER-alpha36-expressing cells, but not the nonexpressing cells, displayed high-affinity, specific E2 and G1 binding, and E2- and G1-induced intracellular Ca(2+) mobilization only in ER-alpha36 expressing cells. Taken together, our results demonstrated that previously reported activities of GPR30 in response to estrogen were through its ability to induce ER-alpha36 expression. The selective G protein-coupled receptor (GPR)30 agonist G1 actually interacts with ER-alpha36. Thus, the ER-alpha variant ER-alpha36, not GPR30, is involved in nongenomic estrogen signaling.
Collapse
Affiliation(s)
- Lianguo Kang
- Department of Medical Microbiology and Immunology, Creighton University Medical School, Omaha, Nebraska 68178, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Grzegorzewski WJ, Chłopek J, Tabecka-Łonczyńska A, Stefańczyk-Krzymowska S. The influence of steroids on vascular tension of isolated superficial veins of the nose and face during the estrous cycle of gilts. Theriogenology 2010; 73:215-24. [PMID: 19878982 DOI: 10.1016/j.theriogenology.2009.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 09/11/2009] [Accepted: 09/11/2009] [Indexed: 11/18/2022]
Abstract
The arrangement of the superficial facial veins enables blood flow from the nasal cavity into the peripheral circulation by two pathways: through the frontal vein into the cavernous sinus and through the facial vein into the external jugular vein. The current study was designed to determine whether estradiol and progesterone affect the vascular tone of the superficial veins of the nose and face in cycling gilts (Sus scrofa f. domestica) and to analyze the immunolocalization of progesterone receptors and estradiol receptors in these veins. The influence of hormones on vascular tension differed depending on the type of vessel and the phase of the estrous cycle. Estradiol decreased vascular tension in the nasal vein during the follicular phase (P<0.05) and increased tension in the frontal vein during the luteal phase (P<0.05). Progesterone increased the vascular tension of the frontal vein (P<0.05) and decreased the tension of the other veins (P<0.05) in both phases of the cycle. Expression of estradiol receptor beta but not of progesterone receptor was observed in the superficial veins of the nose and face. In conclusion, the effect of ovarian steroid hormones on the vascular tension of the superficial veins of the nose and face in female pigs as well as the reactivity of these veins to steroid boar pheromones can affect the blood supply from the nasal cavity to the venous cavernous sinus. We propose that the ovarian steroid hormones that modulate the vascular tension of the nasal and facial veins may also influence the action of boar pheromones absorbed into the nasal mucosa in gilts and may reach the brain via local destination transfer.
Collapse
Affiliation(s)
- W J Grzegorzewski
- Department of Local Physiological Regulation, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Poland.
| | | | | | | |
Collapse
|
50
|
Simoncini T. Mechanisms of action of estrogen receptors in vascular cells: relevance for menopause and aging. Climacteric 2010; 12 Suppl 1:6-11. [PMID: 19811233 DOI: 10.1080/13697130902986385] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Estrogen exerts pleiotropic functions on the cardiovascular system through binding to estrogen receptors (ERs). Traditionally, ERs have been recognized as transcription factors regulating the expression of target genes. In the past decades, however, numerous studies have revealed rapid actions of estrogen in different systems, especially in non-reproductive tissues such as the cardiovascular system. At this level, estrogen triggers rapid vasodilatation, exerts anti-inflammatory effects, regulates vascular cell growth and migration, and confers protection to cardiomyocytes. These so-called 'extranuclear actions' do not require gene expression or protein synthesis and are independent of the nuclear localization of ERs. Indeed, some of these actions are elicited by ERs residing at or near the plasma membrane. Through complex interactions with membrane-associated signaling molecules such as ion channels, G proteins and the tyrosine kinase c-Src, liganded extranuclear ERs lead to the activation of downstream cascades such as mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-OH kinase (PI3K). These cascades are responsible for important cardiovascular actions of estrogen, for instance, the activation of nitric oxide synthesis or the remodeling of the endothelial actin cytoskeleton. Moreover, these cascades play crucial roles in regulating the expression of target proteins implicated in cell proliferation, apoptosis, differentiation, movement and homeostasis. Recent advancements in the characterization of the molecular basis of the extranuclear signaling of estrogen help us to understand the biological functions of estrogen and would be beneficial in elucidating current controversies on estrogen's clinical efficacy in the cardiovascular system.
Collapse
Affiliation(s)
- T Simoncini
- Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
| |
Collapse
|