1
|
Torres-Torres J, Espino-y-Sosa S, Martinez-Portilla R, Borboa-Olivares H, Estrada-Gutierrez G, Acevedo-Gallegos S, Ruiz-Ramirez E, Velasco-Espin M, Cerda-Flores P, Ramirez-Gonzalez A, Rojas-Zepeda L. A Narrative Review on the Pathophysiology of Preeclampsia. Int J Mol Sci 2024; 25:7569. [PMID: 39062815 PMCID: PMC11277207 DOI: 10.3390/ijms25147569] [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: 06/15/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Preeclampsia (PE) is a multifactorial pregnancy disorder characterized by hypertension and proteinuria, posing significant risks to both maternal and fetal health. Despite extensive research, its complex pathophysiology remains incompletely understood. This narrative review aims to elucidate the intricate mechanisms contributing to PE, focusing on abnormal placentation, maternal systemic response, oxidative stress, inflammation, and genetic and epigenetic factors. This review synthesizes findings from recent studies, clinical trials, and meta-analyses, highlighting key molecular and cellular pathways involved in PE. The review integrates data on oxidative stress biomarkers, angiogenic factors, immune interactions, and mitochondrial dysfunction. PE is initiated by poor placentation due to inadequate trophoblast invasion and improper spiral artery remodeling, leading to placental hypoxia. This triggers the release of anti-angiogenic factors such as soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng), causing widespread endothelial dysfunction and systemic inflammation. Oxidative stress, mitochondrial abnormalities, and immune dysregulation further exacerbate the condition. Genetic and epigenetic modifications, including polymorphisms in the Fms-like tyrosine kinase 1 (FLT1) gene and altered microRNA (miRNA) expression, play critical roles. Emerging therapeutic strategies targeting oxidative stress, inflammation, angiogenesis, and specific molecular pathways like the heme oxygenase-1/carbon monoxide (HO-1/CO) and cystathionine gamma-lyase/hydrogen sulfide (CSE/H2S) pathways show promise in mitigating preeclampsia's effects. PE is a complex disorder with multifactorial origins involving abnormal placentation, endothelial dysfunction, systemic inflammation, and oxidative stress. Despite advances in understanding its pathophysiology, effective prevention and treatment strategies remain limited. Continued research is essential to develop targeted therapies that can improve outcomes for both mothers and their babies.
Collapse
Affiliation(s)
- Johnatan Torres-Torres
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Salvador Espino-y-Sosa
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Raigam Martinez-Portilla
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Hector Borboa-Olivares
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Guadalupe Estrada-Gutierrez
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Sandra Acevedo-Gallegos
- Clinical Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (R.M.-P.)
| | - Erika Ruiz-Ramirez
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Martha Velasco-Espin
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Pablo Cerda-Flores
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Andrea Ramirez-Gonzalez
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico (P.C.-F.)
| | - Lourdes Rojas-Zepeda
- Maternal-Fetal Medicine Department, Instituto Materno Infantil del Estado de Mexico, Toluca 50170, Mexico
| |
Collapse
|
2
|
Zhang R, Wang H, Cheng X, Fan K, Gao T, Qi X, Gao S, Zheng G, Dong H. High estrogen induces trans-differentiation of vascular smooth muscle cells to a macrophage-like phenotype resulting in aortic inflammation via inhibiting VHL/HIF1a/KLF4 axis. Aging (Albany NY) 2024; 16:9876-9898. [PMID: 38843385 PMCID: PMC11210252 DOI: 10.18632/aging.205904] [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: 12/15/2023] [Accepted: 04/22/2024] [Indexed: 06/22/2024]
Abstract
Estrogen is thought to have a role in slowing down aging and protecting cardiovascular and cognitive function. However, high doses of estrogen are still positively associated with autoimmune diseases and tumors with systemic inflammation. First, we administered exogenous estrogen to female mice for three consecutive months and found that the aorta of mice on estrogen develops inflammatory manifestations similar to Takayasu arteritis (TAK). Then, in vitro estrogen intervention was performed on mouse aortic vascular smooth muscle cells (MOVAS cells). Stimulated by high concentrations of estradiol, MOVAS cells showed decreased expression of contractile phenotypic markers and increased expression of macrophage-like phenotypic markers. This shift was blocked by tamoxifen and Krüppel-like factor 4 (KLF4) inhibitors and enhanced by Von Hippel-Lindau (VHL)/hypoxia-inducible factor-1α (HIF-1α) interaction inhibitors. It suggests that estrogen-targeted regulation of the VHL/HIF-1α/KLF4 axis induces phenotypic transformation of vascular smooth muscle cells (VSMC). In addition, estrogen-regulated phenotypic conversion of VSMC to macrophages is a key mechanism of estrogen-induced vascular inflammation, which justifies the risk of clinical use of estrogen replacement therapy.
Collapse
MESH Headings
- Kruppel-Like Factor 4
- Animals
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Macrophages/metabolism
- Macrophages/drug effects
- Mice
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Female
- Estrogens/pharmacology
- Von Hippel-Lindau Tumor Suppressor Protein/metabolism
- Von Hippel-Lindau Tumor Suppressor Protein/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Cell Transdifferentiation/drug effects
- Phenotype
- Aorta/pathology
- Aorta/drug effects
- Inflammation/metabolism
Collapse
Affiliation(s)
- Ruijing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Heng Wang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xing Cheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Keyi Fan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tingting Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaotong Qi
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Siqi Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guoping Zheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
3
|
Morishita M, Higo S, Iwata K, Ishii H. Sex and interspecies differences in ESR2-expressing cell distributions in mouse and rat brains. Biol Sex Differ 2023; 14:89. [PMID: 38111056 PMCID: PMC10726529 DOI: 10.1186/s13293-023-00574-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND ESR2, a nuclear estrogen receptor also known as estrogen receptor β, is expressed in the brain and contributes to the actions of estrogen in various physiological phenomena. However, its expression profiles in the brain have long been debated because of difficulties in detecting ESR2-expressing cells. In the present study, we aimed to determine the distribution of ESR2 in rodent brains, as well as its sex and interspecies differences, using immunohistochemical detection with a well-validated anti-ESR2 antibody (PPZ0506). METHODS To determine the expression profiles of ESR2 protein in rodent brains, whole brain sections from mice and rats of both sexes were subjected to immunostaining for ESR2. In addition, to evaluate the effects of circulating estrogen on ESR2 expression profiles, ovariectomized female mice and rats were treated with low or high doses of estrogen, and the resulting numbers of ESR2-immunopositive cells were analyzed. Welch's t-test was used for comparisons between two groups for sex differences, and one-way analysis of variance followed by the Tukey-Kramer test were used for comparisons among multiple groups with different estrogen treatments. RESULTS ESR2-immunopositive cells were observed in several subregions of mouse and rat brains, including the preoptic area, extended amygdala, hypothalamus, mesencephalon, and cerebral cortex. Their distribution profiles exhibited sex and interspecies differences. In addition, low-dose estrogen treatment in ovariectomized female mice and rats tended to increase the numbers of ESR2-immunopositive cells, whereas high-dose estrogen treatment tended to decrease these numbers. CONCLUSIONS Immunohistochemistry using the well-validated PPZ0506 antibody revealed a more localized expression of ESR2 protein in rodent brains than has previously been reported. Furthermore, there were marked sex and interspecies differences in its distribution. Our histological analyses also revealed estrogen-dependent changes in ESR2 expression levels in female brains. These findings will be helpful for understanding the ESR2-mediated actions of estrogen in the brain.
Collapse
Affiliation(s)
- Masahiro Morishita
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Shimpei Higo
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Kinuyo Iwata
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Hirotaka Ishii
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan.
| |
Collapse
|
4
|
Cignarella A, Boscaro C, Albiero M, Bolego C, Barton M. Post-Transcriptional and Epigenetic Regulation of Estrogen Signaling. J Pharmacol Exp Ther 2023; 386:288-297. [PMID: 37391222 DOI: 10.1124/jpet.123.001613] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/17/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023] Open
Abstract
Post-translational and epigenetic regulation are important mechanisms controlling functions of genes and proteins. Although the "classic" estrogen receptors (ERs) have been acknowledged to function in mediating estrogen effects via transcriptional mechanisms, estrogenic agents modulate the turnover of several proteins via post-transcriptional and post-translational pathways including epigenetics. For instance, the metabolic and angiogenic action of G-protein coupled estrogen receptor (GPER) in vascular endothelial cells has been recently elucidated. By interacting with GPER, 17β-estradiol and the GPER agonist G1 enhance endothelial stability of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and capillary tube formation by increasing ubiquitin-specific peptidase 19 levels, thereby reducing PFKFB3 ubiquitination and proteasomal degradation. In addition to ligands, the functional expression and trafficking of ERs can be modulated by post-translational modification, including palmitoylation. MicroRNAs (miRNAs), the most abundant form of endogenous small RNAs in humans, regulate multiple target genes and are at the center of the multi-target regulatory network. This review also discusses the emerging evidence of how miRNAs affect glycolytic metabolism in cancer, as well as their regulation by estrogens. Restoring dysregulated miRNA expression represents a promising strategy to counteract the progression of cancer and other disease conditions. Accordingly, estrogen post-transcriptional regulatory and epigenetic mechanisms represent novel targets for pharmacological and nonpharmacological intervention for the treatment and prevention of hormone-sensitive noncommunicable diseases, including estrogen-sensitive cancers of the reproductive system in women. SIGNIFICANCE STATEMENT: The effects of estrogen are mediated by several mechanisms that are not limited to the transcriptional regulation of target genes. Slowing down the turnover of master regulators of metabolism by estrogens allows cells to rapidly adapt to environmental cues. Identification of estrogen-targeted microRNAs may lead to the development of novel RNA therapeutics that disrupt pathological angiogenesis in estrogen-dependent cancers.
Collapse
Affiliation(s)
- Andrea Cignarella
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Carlotta Boscaro
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Mattia Albiero
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Chiara Bolego
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Matthias Barton
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| |
Collapse
|
5
|
Boscaro C, Carotti M, Albiero M, Trenti A, Fadini GP, Trevisi L, Sandonà D, Cignarella A, Bolego C. Non-genomic mechanisms in the estrogen regulation of glycolytic protein levels in endothelial cells. FASEB J 2020; 34:12768-12784. [PMID: 32757462 DOI: 10.1096/fj.202001130r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/07/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022]
Abstract
Few studies have explored the mechanisms coupling estrogen signals to metabolic demand in endothelial cells. We recently showed that 17β-estradiol (E2) triggers angiogenesis via the membrane G-protein coupled estrogen receptor (GPER) and the key glycolytic protein PFKFB3 as a downstream effector. We herein investigated whether estrogenic agents regulate the stability and/or degradation of glycolytic proteins in human umbilical vein endothelial cells (HUVECs). Similarly to E2, the GPER selective agonist G1 rapidly increased PFKFB3 protein amounts, without affecting mRNA levels. In the presence of cycloheximide, E2 and G1 treatment counteracted PFKFB3 degradation over time, whereas E2-induced PFKFB3 stabilization was abolished by the GPER antagonist G15. Inhibitors of selective SCF E3 ubiquitin ligase (SMER-3) and proteasome (MG132) rapidly increased PFKFB3 protein levels. Accordingly, ubiquitin-bound PFKFB3 was lower in E2- or G1-treated HUVECs. Both agents increased deubiquitinase USP19 levels through GPER signaling. Notably, USP 19 siRNA decreased PFKFB3 levels and abolished E2- and G1-mediated HUVEC tubularization. Finally, E2 and G1 treatments rapidly enhanced glucose transporter GLUT1 levels via GPER independent of transcriptional activation. These findings provide new evidence on mechanisms coupling estrogen signals with the glycolytic program in endothelium and unravel the role of USP19 as a target of the pro-angiogenic effect of estrogenic agents.
Collapse
Affiliation(s)
- Carlotta Boscaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Marcello Carotti
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Annalisa Trenti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Lucia Trevisi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Dorianna Sandonà
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
6
|
Huxley VH, Kemp SS, Schramm C, Sieveking S, Bingaman S, Yu Y, Zaniletti I, Stockard K, Wang J. Sex differences influencing micro- and macrovascular endothelial phenotype in vitro. J Physiol 2018; 596:3929-3949. [PMID: 29885204 DOI: 10.1113/jp276048] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Endothelial dysfunction is an early hallmark of multiple disease states that also display sex differences with respect to age of onset, frequency and severity. Results of in vivo studies of basal and stimulated microvascular barrier function revealed sex differences that are difficult to ascribe to specific cells or environmental factors. The present study evaluated endothelial cells (EC) isolated from macro- and/or microvessels of reproductively mature rats under the controlled conditions of low-passage culture aiming to test the assumption that EC phenotype would be sex independent. The primary finding was that EC, regardless of where they are derived, retain a sex-bias in low-passage culture, independent of varying levels of reproductive hormones. The implications of the present study include the fallacy of expecting a universal set of mechanisms derived from study of EC from one sex and/or one vascular origin to apply uniformly to all EC under unstimulated conditions, and no less in disease. ABSTRACT Vascular endothelial cells (EC) are heterogeneous with respect to phenotype, reflecting at least the organ of origin, location within the vascular network and physical forces. As an independent influence on EC functions in health or aetiology, susceptibility, and progression of dysfunction in numerous disease states, sex has been largely ignored. The present study focussed on EC isolated from aorta (macrovascular) and skeletal muscle vessels (microvascular) of age-matched male and female rats under identical conditions of short-term (passage 4) culture. We tested the hypothesis that genomic sex would not influence endothelial growth, wound healing, morphology, lactate production, or messenger RNA and protein expression of key proteins (sex hormone receptors for androgen and oestrogens α and β; platelet endothelial cell adhesion molecule-1 and vascular endothelial cadherin mediating barrier function; αv β3 and N-cadherin influencing matrix interactions; intracellular adhesion molecule-1 and vascular cell adhesion molecule-1 mediating EC/white cell adhesion). The hypothesis was rejected because the EC origin (macro- vs. microvessel) and sex influenced multiple phenotypic characteristics. Statistical model analysis of EC growth demonstrated an hierarchy of variable importance, recapitulated for other phenotypic characteristics, with predictions assuming EC homogeneity < sex < vessel origin < sex and vessel origin. Furthermore, patterns of EC mRNA expression by vessel origin and by sex did not predict protein expression. Overall, the present study demonstrated that accurate assessment of sex-linked EC dysfunction first requires an understanding of EC function by position in the vascular tree and by sex. The results from a single EC tissue source/species/sex cannot provide universal insight into the mechanisms regulating in vivo endothelial function in health, and no less in disease.
Collapse
Affiliation(s)
- Virginia H Huxley
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Scott S Kemp
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Christine Schramm
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Steve Sieveking
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Susan Bingaman
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Yang Yu
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Isabella Zaniletti
- Department of Statistics, University of Missouri-Columbia, Columbia, MO, USA
| | - Kevin Stockard
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, MO, USA
| | - Jianjie Wang
- National Center for Gender Physiology, University of Missouri-Columbia, Columbia, MO, USA.,Department of Biomedical Sciences, Missouri State University, Springfield, MO, USA
| |
Collapse
|
7
|
Li J, Rao D, Gibbs RB. Effects of Cholinergic Lesions and Cholinesterase Inhibitors on Aromatase and Estrogen Receptor Expression in Different Regions of the Rat Brain. Neuroscience 2018; 384:203-213. [PMID: 29852246 DOI: 10.1016/j.neuroscience.2018.05.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/07/2018] [Accepted: 05/21/2018] [Indexed: 11/28/2022]
Abstract
Cholinergic projections have been shown to interact with estrogens in ways that influence synaptic plasticity and cognitive performance. The mechanisms are not well understood. The goal of this study was to investigate whether cholinergic projections influence brain estrogen production by affecting aromatase (ARO), or influence estrogen signaling by affecting estrogen receptor expression. In the first experiment, ovariectomized rats received intraseptal injection of the selective immunotoxin 192IgG-saporin to destroy cholinergic inputs to the hippocampus. In the second experiment ovariectomized rats received daily intraperitoneal injections of the cholinesterase inhibitors donepezil or galantamine for 1 week. ARO activity and relative levels of ARO, ERα, ERß, and GPR30 mRNAs were quantified in the hippocampus, frontal cortex, amygdala and preoptic area. Results show that the cholinergic lesions effectively removed cholinergic inputs to the hippocampus, but had no significant effect on ARO or on relative levels of ER mRNAs. Likewise, injections of the cholinesterase inhibitors had no effect on ARO or ER expression in most regions of the brain. This suggests that effects of cholinergic inputs on synaptic plasticity and neuronal function are not mediated by effects on local estrogen production or ER expression. One exception was the amygdala where treating with galantamine was associated with a significant increase in ARO activity. The amygdala is a key structure involved in registering fear and anxiety. Hence this finding may be clinically relevant to elderly patients who are treated for memory impairment and who also struggle with fear and anxiety disorders.
Collapse
Affiliation(s)
- Junyi Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Di Rao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Robert B Gibbs
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
8
|
Iyer JK, Kalra M, Kaul A, Payton ME, Kaul R. Estrogen receptor expression in chronic hepatitis C and hepatocellular carcinoma pathogenesis. World J Gastroenterol 2017; 23:6802-6816. [PMID: 29085224 PMCID: PMC5645614 DOI: 10.3748/wjg.v23.i37.6802] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/12/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate gender-specific liver estrogen receptor (ER) expression in normal subjects and patients with hepatitis C virus (HCV)-related cirrhosis and hepatocellular carcinoma (HCC).
METHODS Liver tissues from normal donors and patients diagnosed with HCV-related cirrhosis and HCV-related HCC were obtained from the NIH Liver Tissue and Cell Distribution System. The expression of ER subtypes, ERα and ERβ, were evaluated by Western blotting and real-time RT-PCR. The subcellular distribution of ERα and ERβ was further determined in nuclear and cytoplasmic tissue lysates along with the expression of inflammatory [activated NF-κB and IκB-kinase (IKK)] and oncogenic (cyclin D1) markers by Western blotting and immunohistochemistry. The expression of ERα and ERβ was correlated with the expression of activated NF-κB, activated IKK and cyclin D1 by Spearman’s correlation.
RESULTS Both ER subtypes were expressed in normal livers but male livers showed significantly higher expression of ERα than females (P < 0.05). We observed significantly higher mRNA expression of ERα in HCV-related HCC liver tissues as compared to normals (P < 0.05) and ERβ in livers of HCV-related cirrhosis and HCV-related HCC subjects (P < 0.05). At the protein level, there was a significantly higher expression of nuclear ERα in livers of HCV-related HCC patients and nuclear ERβ in HCV-related cirrhosis patients as compared to normals (P < 0.05). Furthermore, we observed a significantly higher expression of phosphorylated NF-κB and cyclin D1 in diseased livers (P < 0.05). There was a positive correlation between the expression of nuclear ER subtypes and nuclear cyclin D1 and a negative correlation between cytoplasmic ER subtypes and cytoplasmic phosphorylated IKK in HCV-related HCC livers. These findings suggest that dysregulated expression of ER subtypes following chronic HCV-infection may contribute to the progression of HCV-related cirrhosis to HCV-related HCC.
CONCLUSION Gender differences were observed in ERα expression in normal livers. Alterations in ER subtype expression observed in diseased livers may influence gender-related disparity in HCV-related pathogenesis.
Collapse
Affiliation(s)
- Janaki K Iyer
- Department of Biochemistry and Microbiology, Oklahoma State University-Center for Health Sciences, Tulsa, OK 74107, United States
- (Current Affiliation) Department of Natural Sciences, Northeastern State University, Tahlequah, OK 74464, United States
| | - Mamta Kalra
- Immatics US Inc, Houston, TX 77077, United States
| | - Anil Kaul
- Health Care Administration, Oklahoma State University-Center for Health Sciences, Tulsa, OK 74107, United States
| | - Mark E Payton
- Department of Statistics, Oklahoma State University, Stillwater, OK 74078, United States
| | - Rashmi Kaul
- Department of Biochemistry and Microbiology, Oklahoma State University-Center for Health Sciences, Tulsa, OK 74107, United States
| |
Collapse
|
9
|
Liu KC, Lau SW, Ge W. Spatiotemporal expression analysis of nuclear estrogen receptors in the zebrafish ovary and their regulation in vitro by endocrine hormones and paracrine factors. Gen Comp Endocrinol 2017; 246:218-225. [PMID: 28013034 DOI: 10.1016/j.ygcen.2016.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/11/2016] [Accepted: 12/19/2016] [Indexed: 11/20/2022]
Abstract
Estradiol (E2) stimulates luteinizing hormone receptor (lhcgr) expression via nuclear estrogen receptors (nERs) in the zebrafish ovary. We have demonstrated that endocrine hormones such as gonadotropin (hCG) and paracrine factors such as epidermal growth factor (EGF) and pituitary adenylate cyclase-activating peptide (PACAP) can modulate E2-induced lhcgr expression in vitro. These observations raised a question on whether these hormones and factors exert their effects via regulating the expression of nERs. In this study, we first characterized the spatiotemporal expression profiles of three nER subtypes in the zebrafish ovary, including esr1 (ERα), esr2a (ERβ2) and esr2b (ERβ1). All three nERs increased their expression at the pre-vitellogenic stage and peaked at mid- (esr1 and esr2a) or late vitellogenic (esr2b) stage, followed by a significant decline at the full-grown stage. RT-PCR analysis showed that esr1 and esr2b were exclusively expressed in the follicle layer while esr2a was expressed in both compartments. We then examined how E2, hCG, PACAP and EGF regulated the expression of nERs in cultured zebrafish follicle cells. E2 quickly increased esr1 but reduced esr2a and esr2b expression from 1.5 to 12h of treatment. Similarly, EGF down-regulated esr2a significantly at 1.5h and this effect was further intensified at 24h. hCG decreased the expression of all three nER subtypes with similar potency throughout the 24-h time-course. Interestingly, PACAP exerted a biphasic regulation on esr2a. Our present study suggests that nERs, especially esr2a, provide potential target points for other hormones and factors to modulate E2 activity during folliculogenesis in the zebrafish.
Collapse
Affiliation(s)
- Ka-Cheuk Liu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Shuk-Wa Lau
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China; School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.
| |
Collapse
|
10
|
Bisphenol S negatively affects the meotic maturation of pig oocytes. Sci Rep 2017; 7:485. [PMID: 28352085 PMCID: PMC5428703 DOI: 10.1038/s41598-017-00570-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/01/2017] [Indexed: 01/19/2023] Open
Abstract
Bisphenol A (BPA), a chemical component of plastics, is a widely distributed environmental pollutant and contaminant of water, air, and food that negatively impacts human health. Concerns regarding BPA have led to the use of BPA-free alternatives, one of which is bisphenol S (BPS). However, the effects of BPS are not well characterized, and its specific effects on reproduction and fertility remain unknown. It is therefore necessary to evaluate any effects of BPS on mammalian oocytes. The present study is the first to demonstrate the markedly negative effects of BPS on pig oocyte maturation in vitro, even at doses lower than those humans are exposed to in the environment. Our results demonstrate (1) an effect of BPS on the course of the meiotic cell cycle; (2) the failure of tubulin fibre formation, which controls proper chromosome movement; (3) changes in the supply of maternal mRNA; (4) changes in the protein amounts and distribution of oestrogen receptors α and β and of aromatase; and (5) disrupted cumulus cell expansion. Thus, these results confirm that BPS is an example of regrettable substitution because this substance exerts similar or even worse negative effects than those of the material it replaced.
Collapse
|
11
|
An BH, Jeong H, Zhou W, Liu X, Kim S, Jang CY, Kim HS, Sohn J, Park HJ, Sung NH, Hong CY, Chang M. Evaluation of the Biological Activity of Opuntia ficus indica as a Tissue- and Estrogen Receptor Subtype-Selective Modulator. Phytother Res 2016; 30:971-80. [PMID: 26989859 DOI: 10.1002/ptr.5602] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/07/2016] [Accepted: 02/09/2016] [Indexed: 11/07/2022]
Abstract
Phytoestrogens are selective estrogen receptor modulators (SERMs) with potential for use in hormone replacement therapy (HRT) to relieve peri/postmenopausal symptoms. This study was aimed at elucidating the molecular mechanisms underlying the SERM properties of the extract of Korean-grown Opuntia ficus-indica (KOFI). The KOFI extract induced estrogen response element (ERE)-driven transcription in breast and endometrial cancer cell lines and the expression of endogenous estrogen-responsive genes in breast cancer cells. The flavonoid content of different KOFI preparations affected ERE-luciferase activities, implying that the flavonoid composition likely mediated the estrogenic activities in cells. Oral administration of KOFI decreased the weight gain and levels of both serum glucose and triglyceride in ovariectomized (OVX) rats. Finally, KOFI had an inhibitory effect on the 17β-estradiol-induced proliferation of the endometrial epithelium in OVX rats. Our data demonstrate that KOFI exhibited SERM activity with no uterotrophic side effects. Therefore, KOFI alone or in combination with other botanical supplements, vitamins, or minerals may be an effective and safe alternative active ingredient to HRTs, for the management of postmenopausal symptoms. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Byoung Ha An
- Department of Food and Nutrition, College of Life Sciences, Sookmyung Women's University, 100, Chungparo 47-gil, Seoul, 140-742, Republic of Korea
| | - Hyesoo Jeong
- Graduate School of Biological Sciences, Sookmyung Women's University, 100, Chungparo 47-gil, Seoul, 140-742, Republic of Korea
| | - Wenmei Zhou
- Graduate School of Biological Sciences, Sookmyung Women's University, 100, Chungparo 47-gil, Seoul, 140-742, Republic of Korea
| | - Xiyuan Liu
- Graduate School of Biological Sciences, Sookmyung Women's University, 100, Chungparo 47-gil, Seoul, 140-742, Republic of Korea
| | - Soolin Kim
- Graduate School of Biological Sciences, Sookmyung Women's University, 100, Chungparo 47-gil, Seoul, 140-742, Republic of Korea
| | - Chang Young Jang
- Department of Pharmacy, College of Pharmacy, Sookmyung Women's University, 100, Chungparo 47-gil, Seoul, 140-742, Republic of Korea
| | - Hyun-Sook Kim
- Department of Food and Nutrition, College of Life Sciences, Sookmyung Women's University, 100, Chungparo 47-gil, Seoul, 140-742, Republic of Korea
| | - Johann Sohn
- Natural F&P Corp. 39 Yangcheongsongdae-gil, Ochang-eup, Chongwon-gu, Cheongju-si, Chungbuk, Korea
| | - Hye-Jin Park
- Natural F&P Corp. 39 Yangcheongsongdae-gil, Ochang-eup, Chongwon-gu, Cheongju-si, Chungbuk, Korea
| | - Na-Hye Sung
- Natural F&P Corp. 39 Yangcheongsongdae-gil, Ochang-eup, Chongwon-gu, Cheongju-si, Chungbuk, Korea
| | - Cheol Yi Hong
- Natural F&P Corp. 39 Yangcheongsongdae-gil, Ochang-eup, Chongwon-gu, Cheongju-si, Chungbuk, Korea
| | - Minsun Chang
- Department of Medical and Pharmaceutical Science, College of Science, Sookmyung Women's University, 100, Chungparo 47-gil, Seoul, 140-742, Republic of Korea
| |
Collapse
|
12
|
Pisolato R, Lombardi APG, Vicente CM, Lucas TFG, Lazari MFM, Porto CS. Expression and regulation of the estrogen receptors in PC-3 human prostate cancer cells. Steroids 2016; 107:74-86. [PMID: 26742628 DOI: 10.1016/j.steroids.2015.12.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 10/21/2015] [Accepted: 12/28/2015] [Indexed: 01/02/2023]
Abstract
The aim of this study was to identify the expression, cellular localization and regulation of classic estrogen receptors ERα and ERβ, ER-α36 isoform and GPER in the androgen-independent prostate cancer cell line PC-3. In addition, we evaluated the relative contribution of these receptors to the activation of the ERK1/2 (extracellular signal-regulated protein kinases) signaling pathway. These four estrogen receptors were detected by Western blot assays and were shown by immunofluorescence assays to localize preferentially in extranuclear regions of PC-3 cells. In addition, treatment with 17β-estradiol (E2) (1 μM) for 24 h led to down-regulation of the classic estrogen receptors, whereas E2 at physiological concentration (0.1 nM) for 24h tended to increase the levels of ERα and ERβ. Furthermore, the ERα-selective agonist PPT selectively increased the expression of ERβ and the ERβ-selective agonist DPN increased ERα levels. None of these treatments affected expression of the ER-α36 isoform. The unusual cytoplasmic localization of the classic estrogen receptors in these cells differs from the nuclear localization in the majority of estrogen target cells and suggests that rapid signaling pathways may be preferentially activated. In fact, treatment with selective agonists of ERα, ERβ and GPER induced ERK1/2 phosphorylation that was blocked by the respective antagonists. On the other hand, activation of ERK1/2 induced by E2 may involve additional mechanisms because it was not blocked by the three antagonists. Taken together, the results indicate that there is a crosstalk between ERα and ERβ to regulate the expression of each other, and suggest the involvement of other receptors, such as ER-α36, in the rapid ERK1/2 activation by E2. The identification of new isoforms of ERs, regulation of the receptors and signaling pathways is important to develop new therapeutic strategies for the castration-resistant prostate cancer.
Collapse
Affiliation(s)
- R Pisolato
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, UNIFESP, São Paulo, SP, Brazil
| | - A P G Lombardi
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, UNIFESP, São Paulo, SP, Brazil
| | - C M Vicente
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, UNIFESP, São Paulo, SP, Brazil
| | - T F G Lucas
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, UNIFESP, São Paulo, SP, Brazil
| | - M F M Lazari
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, UNIFESP, São Paulo, SP, Brazil
| | - C S Porto
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina, UNIFESP, São Paulo, SP, Brazil.
| |
Collapse
|
13
|
Nickel S, Mahringer A. The xenoestrogens ethinylestradiol and bisphenol A regulate BCRP at the blood-brain barrier of rats. Xenobiotica 2014; 44:1046-54. [PMID: 24945792 DOI: 10.3109/00498254.2014.922226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
1. Breast cancer resistance protein (BCRP) is an ABC-transporter at the blood-brain barrier (BBB) facilitating efflux of xenobiotics into blood. Expression and function are regulated via estrogen-receptors (ERs). 2. 17α-Ethinylestradiol (EE2) and bisphenol A (BPA) represent two prominent xenoestrogens. We studied whether EE2 and BPA regulate BCRP function and expression upon a 6 h treatment in an ER-dependent manner in a rat BBB-ex-vivo-model. 3. Isolated brain capillaries were incubated with EE2 or BPA. BCRP function and expression were analyzed by confocal microscopy and Western-Blot. ERα-antagonist MPP and ER-antagonist ICI182.780 were used to study involvement of ERs. 4. EE2 and BPA down-regulated BCRP transport function and expression. EE2 effects occurred at pharmacologically relevant doses, BPA exhibited only weak influences. Down-regulation by EE2 was reversed by ICI but not MPP. BPA effects were not reversed by either antagonist. 5. EE2 is a potent regulator of BCRP expression and function acting by ERβ-stimulation. Oral contraception could alter uptake of pharmaceuticals to the brain and might thus be considered as an origin of central nervous system (CNS) side-effects. EE2 could also present a novel co-treatment to improve CNS-pharmacotherapy. BPA is a weak modulator of BCRP expression. Its effects appear not to be caused by ERs.
Collapse
Affiliation(s)
- Sabrina Nickel
- Department of Pharmaceutical Technology and Biopharmaceutics, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg , Heidelberg , Germany
| | | |
Collapse
|
14
|
Age-associated changes in bovine oocytes and granulosa cell complexes collected from early antral follicles. J Assist Reprod Genet 2014; 31:1079-88. [PMID: 24830789 DOI: 10.1007/s10815-014-0251-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/07/2014] [Indexed: 01/07/2023] Open
Abstract
PURPOSE To assess the age-associated changes in oocytes and granulosa cells derived from early antral follicles (EAFs). METHOD Gene expression analysis of granulosa cells of the EAFs using a genome analyzer (Illumina) and in vitro culture of oocyte-granulosa cell complexes (OGCs) of EAFs (400-700 μm in diameter) collected from ovaries of aged (>120 months) and young (<50 months) cows. RESULTS Gene expression profiles in granulosa cells of EAFs of aged cows, which included changes in genes that encode chaperone proteins and antioxidants. In vivo development of EAFs, as determined by oocyte diameter of EAFs and AFs (3-6 mm in diameter), appeared to be impaired in aged cows and the OGCs of aged cows contained low GSH compared to younger counterparts. When the OGCs were cultured in a medium containing low estradiol (E2, 0.1 μg/mL), the ratio of antrum formation was higher for OGCs from aged animals than that from young animals, while higher abnormal fertilization rate and lower total cell number of the blastocysts were observed in the OGCs of aged cows compared with those of young cows. On the contrary, when the OGCs were cultured in a medium containing 10 μg/mL E2, the ratio of antrum formation and fertilization outcome was comparable between the two age groups, whereas the total cell number of the blastocysts was still low in the aged group. CONCLUSION Aging affects the gene expression profiles of the granulosa cells, and impairs in vitro developmental ability of OGCs collected from EAFs.
Collapse
|
15
|
Ishibashi O. Bone Morphogenetic Protein-2 Desensitizes MC3T3-E1 Osteoblastic Cells to Estrogen Through Transcriptional Downregulation of Estrogen Receptor 1. J Bone Metab 2013; 20:83-8. [PMID: 24524062 PMCID: PMC3910307 DOI: 10.11005/jbm.2013.20.2.83] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/16/2013] [Accepted: 10/16/2013] [Indexed: 11/29/2022] Open
Abstract
Background Estrogens exert preferable effects on bone metabolism through two estrogen receptors (ERs), ER1 and ER2, which activate the transcription of a set of genes as ligand-dependent transcription factors. Thus, growth factors and hormones which modulate ER expression in the bone, if any, may possibly modulate the effect of estrogens on bone metabolism. However, research as to which of these molecules regulate the expression of ERs in osteoblasts has not been well documented. Methods A reporter assay system developed in this study was used to explore molecules that modulate ER1 expression in MC3T3-E1 osteoblastic cells. Gene expression was analyzed by reverse transcription-polymerase chain reaction. Results A pilot study using the reporter system revealed that bone morphogenetic protein (BMP)-2 negatively regulated ER1, but not ER2, expression in MC3T3-E1 cells. Consistently, estradiol-induced reporter activity via an estrogen responsive element was strongly suppressed in MC3T3-E1 cells pretreated with BMP-2. Conclusions BMP-2 desensitizes osteoblastic cells to estrogen through downregulation of ER1 expression.
Collapse
Affiliation(s)
- Osamu Ishibashi
- Department of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Japan
| |
Collapse
|
16
|
Edgar AR, Judith PY, Elisa DSM, Rafael CR. Glucocorticoids and estrogens modulate the NF-κB pathway differently in the micro- and macrovasculature. Med Hypotheses 2013; 81:1078-82. [PMID: 24199951 DOI: 10.1016/j.mehy.2013.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/08/2013] [Indexed: 10/26/2022]
Abstract
Estrogens and glucocorticoids have synergistic effects in the micro and macrovasculature of endothelial cells (ECs), having pro-inflammatory effects in the former and inhibiting the expression of adhesion molecules in the latter. The molecular basis of these effects in the endothelium has not yet been clarified. We postulate that the ECs of the micro- and macrovasculature have different non-genomic mechanisms that regulate levels of preexisting complexes of glucocorticoids and estrogens with their respective receptors. Since these receptors are regulated by NF-κB, their expression could be critical to the activation of a pro- or anti-inflammatory response. In the macrovasculature the synergistic effects of estrogens and glucocorticoids on ECs may be through the inhibition of NF-κB, leading to the inhibition of the expression of inflammatory molecules. It seems likely that glucocorticoid-receptor and estrogen-receptor complexes directly bind to NF-κB proteins in the macrovasculature, resulting in the inhibition of an excessive proinflammatory response. Further insights into these processes may help clarify the role of the endothelial cells of different vascular beds during the inflammatory response and chronic inflammation, and thus contribute to the design of more effective therapeutic strategies for the prevention of diseases related to inflammation, including atherosclerosis, systemic lupus erythematosus and rheumatoid arthritis.
Collapse
Affiliation(s)
- Abarca-Rojano Edgar
- Laboratorio de Respiración Celular Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luıis y Díaz Mirón, México, D.F., Mexico.
| | | | | | | |
Collapse
|
17
|
Tirado OM, Selva DM, Toràn N, Suárez-Quian CA, Jansen M, McDonnell DP, Reventós J, Munell F. Increased Expression of Estrogen Receptor β in Pachytene Spermatocytes After Short-Term Methoxyacetic Acid Administration. ACTA ACUST UNITED AC 2013; 25:84-94. [PMID: 14662790 DOI: 10.1002/j.1939-4640.2004.tb02762.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Degeneration of primary spermatocytes by apoptosis occurs during normal spermatogenesis, as well as in several pathological conditions, including exposure to specific testicular toxicants. The mechanisms that regulate the death and survival of primary spermatocytes, however, are still not well understood. The recent localization of estrogen receptor beta (ERbeta) and P450 aromatase in pachytene spermatocytes suggests a role for estrogens in this step of spermatogenesis. Using a well-known model of pachytene spermatocyte apoptosis in adult rats consisting of the administration of methoxyacetic acid (MAA), we investigated the participation of ERbeta during the initial phase of apoptosis, prior to germ cell loss. Adult rats were treated with a single intraperitoneal dose of MAA, and DNA laddering analysis confirmed apoptotic cell death in the testis. In enriched germ cell fractions and testis from MAA-treated animals, ERbeta mRNA increased significantly at 3 and 6 hours, respectively. Next, stage-specific induction of ERbeta mRNA was demonstrated by use of laser capture microdissection of seminiferous tubules in combination with semiquantitative reverse transcription-polymerase chain reaction. The ERbeta protein also increased significantly after 6 hours and was mainly immunolocalized in the cytoplasm of pachytene spermatocytes of afflicted tubules. The cytoplasmic localization was confirmed by Western blot analysis of isolated cytoplasmic and nuclear fractions of testicular extracts. Finally, the MAA activation of ERbeta was tested in vitro in HepG2 cells cotransfected with ERbeta and a reporter construct that contained a consensus estrogen responsive element. Addition of MAA at similar doses used in vivo elicited a similar estrogenic activation as did estradiol at 1 nmol/L concentration. The present results raise the possibility that cytoplasmic ERbeta participates in the apoptotic process of pachytene spermatocytes induced by MAA. Whether MAA interacts with ERbeta in the cytoplasm of primary spermatocytes, preventing the progression of the first meiotic division, however, remains to be determined.
Collapse
Affiliation(s)
- Oscar M Tirado
- Unitat de Recerca Biomèdica, Hospital Materno-Infantil Vall d'Hebron, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Marino M, Pellegrini M, La Rosa P, Acconcia F. Susceptibility of estrogen receptor rapid responses to xenoestrogens: Physiological outcomes. Steroids 2012; 77:910-7. [PMID: 22410438 DOI: 10.1016/j.steroids.2012.02.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 02/13/2012] [Accepted: 02/24/2012] [Indexed: 02/06/2023]
Abstract
17β-Estradiol (E2) binding induces rapid modification in the conformation of its cognate receptors (i.e., ERα and ERβ). These allosteric changes allow the association of ERs with cell specific transcriptional cofactors, thus determining cellular contexts specific variations in gene expression. In addition, E2-ER complexes could also interact with membrane and cytosolic signal molecules triggering extra-nuclear signalling pathways. The synergy between these mechanisms is necessary for E2-induced pleiotropic actions in target tissues. Besides E2, the ER ligand binding domains can accommodate many other natural and synthetic ligands. Several of these compounds act as agonist or antagonist of ER transcriptional activity due to their ability to modify the interactions between ERs and transcriptional co-regulators. However, the ability of natural or manmade ER ligands to affect the extra-nuclear interactions of the ERs has been rarely evaluated. Here, the ability of two diet-derived flavonoids (i.e., naringenin and quercetin) and of the synthetic food-contaminant bisphenol A to modulate specifically ER extra-nuclear signalling pathways will be reported. All the tested compounds bind to both ER subtypes even if lesser than E2 activating divergent signal transduction pathways. In fact, in the presence of ERα, both naringenin and quercetin decouple ERα activities by specifically interfering with ERα membrane initiating signals. On the other hand, bisphenol A, but not flavonoids, maintains ERβ at the membrane thus impairing the activation of the downstream kinases. As a whole, extra-nuclear ER signals are highly susceptible to different ligands that, by unbalancing E2-induced cell functions drive cells to different functional endpoints.
Collapse
Affiliation(s)
- Maria Marino
- Department of Biology, University Roma TRE, viale G. Marconi, 446, I-00146 Rome, Italy.
| | | | | | | |
Collapse
|
19
|
Simard M, Drolet R, Blomquist CH, Tremblay Y. Human type 2 17beta-hydroxysteroid dehydrogenase in umbilical vein and artery endothelial cells: differential inactivation of sex steroids according to the vessel type. Endocrine 2011; 40:203-11. [PMID: 21877158 DOI: 10.1007/s12020-011-9519-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 07/29/2011] [Indexed: 10/17/2022]
Abstract
The human placenta produces high amounts of estradiol. 17β-hydroxysteroid dehydrogenase type 2 (17βHSD2) is expressed by placental endothelial cells and was proposed to regulate sex hormone levels. Previous results obtained in term placenta suggested that 17βHSD2 expression and activity differ among umbilical cord vessels. In this study, 17βHSD2 expression level and enzymatic activity, and estrogen receptor α and β expression levels, were measured in endothelial cell cultures from umbilical arteries (HUAEC) and vein (HUVEC) using real-time quantitative PCR, western blot, and radiolabeled steroids. 17βHSD2-specific activities were also measured in proximal and distal segments of freshly isolated umbilical cord arteries and vein. 17βHSD2 mRNA level and activity were higher in HUAEC than in HUVEC. Activity was higher in umbilical arteries than in the umbilical vein. In arteries, enzymatic activity was higher near the placenta, suggesting a gradient of expression. No difference was found in ERα expression, whereas ERβ was expressed at a higher level in HUAEC than in HUVEC. Expression profiles of estrogen receptors and 17βHSD2 suggest a vessel type-specific response to estrogens. Our data support a differential modulation of biologically active sex steroid levels according to the vessel type in the foeto-placental unit, with apparent higher inactivation in the arterial system.
Collapse
Affiliation(s)
- Marc Simard
- Reproduction, Perinatal Health, and Child Health Axis, CHUQ Research Center, 2705 Laurier Blvd., Rm T-1-49, Québec, QC G1V 4G2, Canada
| | | | | | | |
Collapse
|
20
|
Mahringer A, Fricker G. BCRP at the blood-brain barrier: genomic regulation by 17β-estradiol. Mol Pharm 2010; 7:1835-47. [PMID: 20735085 DOI: 10.1021/mp1001729] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
At the blood-brain barrier (BBB), the ABC transporter breast cancer resistance protein (BCRP) actively extrudes a variety of therapeutic drugs, including cytostatics, and diminishes their pharmacological efficacy in the brain. Consequently, new strategies to circumvent BCRP-mediated multidrug resistance in the CNS are required. One major approach to increase brain drug levels is to manipulate signaling mechanisms that control transporter expression and function. In the present study, we investigated the long-term effect of 17β-estradiol on BCRP in an ex vivo model of isolated rat brain capillaries. BCRP function and protein expression were decreased after 6 h of incubation with nanomolar concentrations of 17β-estradiol in capillaries from male and female rats. Concomitantly, levels of BCRP mRNA were also reduced by 17β-estradiol suggesting that the transporter is down-regulated via a genomic pathway. Additionally, we identified the presence of both estrogen receptor (ER) subtypes α and β at the rat BBB. Experiments using selective ER agonists and antagonists revealed that ER subtype β is responsible for the hormone-induced reduction of BCRP function and protein expression. These findings were confirmed by the use of ERKO mice. Blocking the proteasome-dependent degradation by lactacystin reversed the 17β-estradiol-mediated decrease of BCRP supposing that transcriptional down-regulation of the efflux transporter is paralleled by protein degradation. This study demonstrates that 17β-estradiol induces the down-regulation of BCRP on transcriptional and translational levels via the activation of ERβ in rat brain capillaries after 6 h. These results could help to improve brain targeting of BCRP substrates in the treatment of CNS diseases such as brain tumors and also contribute to an enlarged understanding of BCRP-drug interactions at a chronic intake of phytoestrogens and oral contraceptives.
Collapse
Affiliation(s)
- Anne Mahringer
- Institute of Pharmacy and Molecular Biotechnology, Department of Pharmaceutical Technology and Biopharmaceutics, University of Heidelberg, 69120 Heidelberg, Germany
| | | |
Collapse
|
21
|
Liu H, Du J, Hu C, Qi H, Wang X, Wang S, Liu Q, Li Z. Delayed activation of extracellular-signal-regulated kinase 1/2 is involved in genistein- and equol-induced cell proliferation and estrogen-receptor-alpha-mediated transcription in MCF-7 breast cancer cells. J Nutr Biochem 2009; 21:390-6. [PMID: 19427779 DOI: 10.1016/j.jnutbio.2009.01.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 12/25/2008] [Accepted: 01/16/2009] [Indexed: 11/17/2022]
Abstract
The aim of this study was to determine whether the extracellular-signal-regulated kinase 1/2 (ERK1/2) pathway is involved in genistein- and equol-induced cell proliferation and estrogen receptor (ER) alpha transactivation. For MCF-7 human breast cells, low concentrations of genistein and equol enhanced proliferation and induced MCF-7 cells to enter the S-phase. Genistein- and equol-induced cell proliferation and S-phase entry were blocked by the ERalpha antagonists 4-hydroxytamoxifen and ICI 182,780 and by the mitogen-activated protein kinase 1/2 inhibitor U0126. These data indicated that ERalpha and mitogen-activated protein extracellular kinase/ERK signaling were required for the effects of genistein/equol on cell growth and cell cycle progression. Genistein and equol induced delayed and prolonged activation of ERK1/2. Inhibition of ERK1/2 phosphorylation by U0126 led to complete suppression of genistein- and equol-induced estrogen response element reporter activity and to suppression of the estrogen-responsive gene pS2. The anti-estrogen ICI had no effect on genistein- and equol-induced ERK1/2 phosphorylation. These results suggest that activation of ERK1/2 lies upstream of ER-mediated transcription, and that ERK1/2 activation is necessary for the transactivation of ERalpha. In conclusion, genistein and equol elicit a delayed activation of ERK1/2, and this activation appears to be involved in the proliferation of breast cancer cells and estrogen-dependent transcriptional activation.
Collapse
Affiliation(s)
- Huaqing Liu
- The Key Laboratory of Reproductive Medicine of Jiangsu Province, Institute of Toxicology, Nanjing Medical University, Jiangsu, Nanjing, China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Hu C, Liu H, Du J, Mo B, Qi H, Wang X, Ye S, Li Z. Estrogenic activities of extracts of Chinese licorice (Glycyrrhiza uralensis) root in MCF-7 breast cancer cells. J Steroid Biochem Mol Biol 2009; 113:209-16. [PMID: 19167497 DOI: 10.1016/j.jsbmb.2008.12.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 10/01/2008] [Accepted: 12/22/2008] [Indexed: 02/07/2023]
Abstract
Despite the wide use of Chinese licorice root (Glycyrrhiza uralensis) for the treatment of menopausal complaints, little is known on its potential estrogenic properties, and available information relative to its effects on cell proliferation is contradictory. In this study, the estrogenic properties of licorice root were evaluated in vitro by use of several assays. The effects of increasing concentrations of a DMSO extract of licorice root on the growth of MCF-7 breast cancer cells were biphasic. The extract showed an ER-dependent growth-promoting effect at low concentrations and an ER-independent anti-proliferative activity at high concentrations. In further experiments, licorice root was sequentially extracted to yield four fractions: hexane, EtOAc, methanol and H(2)O. Only the EtOAc extract had effects on cell proliferation similar to the DMSO extract. The hexane extract had no effect on cell growth. In contrast, the methanol and water extracts showed an ER-independent, growth-promoting effect. Similar to its effects on cell proliferation, the EtOAc extract had a biphasic effect on S phase cell cycle distribution and the level of PCNA protein. This extract-induced transactivation of endogenous ERalpha in MCF-7 cells, supported by inducing down-regulation of ERalpha protein and mRNA levels, and up-regulation of ERalpha target genes pS2 and GREB1. These results suggest that the activity of licorice root and the balance between increased risk for cancer and prevention of estrogen-dependent breast cancer may depend on the amount of dietary intake.
Collapse
Affiliation(s)
- Chunyan Hu
- Department of Nutrition & Food Hygiene, Nanjing Medical University, Jiangsu, Nanjing, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Pregnancy is a physiological state that involves a significant decrease in uterine vascular tone and an increase in uterine blood flow, which is mediated in part by steroid hormones, including estrogen, progesterone, and cortisol. Previous studies have demonstrated the involvement of these hormones in the regulation of uterine artery contractility through signaling pathways specific to the endothelium and the vascular smooth muscle. Alterations in endothelial nitric oxide synthase expression and activity, nitric oxide production, and expression of enzymes involved in PGI(2) production contribute to the uterine artery endothelium-specific responses. Steroid hormones also have an effect on calcium-activated potassium channel activity, PKC signaling pathway and myogenic tone, and alterations in pharmacomechanical coupling in the uterine artery smooth muscle. This review addresses current understanding of the molecular mechanisms by which steroid hormones including estrogen, progesterone, and cortisol modulate uterine artery contractility to alter uterine blood flow during pregnancy with an emphasis on the pregnant ewe model.
Collapse
Affiliation(s)
- Katherine Chang
- Center for Perinatal Biology, Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California 62350, USA.
| | | |
Collapse
|
24
|
Wood CE. Cerebral hypoperfusion increases estrogen receptor abundance in the ovine fetal brain and pituitary. Neuroendocrinology 2008; 87:216-22. [PMID: 18160819 PMCID: PMC2793328 DOI: 10.1159/000112844] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 11/14/2007] [Indexed: 12/30/2022]
Abstract
BACKGROUND/AIMS Estrogen is an important component of fetal neuroendocrine function in late-gestation fetal sheep; however, little is known about the regulation of estrogen receptor abundance in the brain and pituitary of fetuses. The present study was performed to test the hypotheses that estrogen receptor abundance in the fetal brain and pituitary are influenced by circulating estradiol concentrations and that they are acutely regulated after cerebral hypoperfusion. METHODS We studied 16 time-dated fetal sheep (124-128 days gestation) that were chronically catheterized and instrumented at least 5 days before study. Four groups (n = 4 each) were studied in which fetuses received estradiol (0.25 mg/day, producing physiological increases in fetal plasma estradiol concentrations) or placebo implants, and in which fetuses received a 10-min period of brachiocephalic occlusion (BCO) or sham-BCO. One hour after BCO or sham-BCO, fetuses were euthanized and tissues rapidly removed for analysis of estrogen receptors (ER)-alpha and -beta at the mRNA and protein levels. RESULTS Both BCO and estradiol treatment were effective in changing ER expression, although the effects were region-specific. BCO dramatically increased ER-alpha in the pituitary and both ER-alpha and ER-beta in the brainstem, while decreasing ER-alpha expression in the hypothalamus. Estradiol treatment decreased ER-alpha expression in the hypothalamus, whereas it increased ER-alpha expression in the brainstem, cerebral cortex and hippocampus. CONCLUSIONS We conclude that the expression of ER-alpha and ER-beta in the brain and pituitary of fetal sheep are influenced by circulating estrogen concentrations and acutely regulated in response to cerebral hypoperfusion.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Fla. 32610-0274, USA.
| |
Collapse
|
25
|
Park S, Heo MK, Lee MJ, Kim JH, Park BW. Changes of Coregulators, MAP Kinase Activity and p27/kip1 with Estrogen or Antiestrogen Treatment in Breast Cancer Cell Line. J Breast Cancer 2008. [DOI: 10.4048/jbc.2008.11.2.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Seho Park
- Department of Surgery, Yonsei University College of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Min Kyu Heo
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Jeong Lee
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Joo-hee Kim
- Department of Surgery, Yonsei University College of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Byeong-Woo Park
- Department of Surgery, Yonsei University College of Medicine, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Ardley HC, Robinson PA. The role of ubiquitin-protein ligases in neurodegenerative disease. NEURODEGENER DIS 2006; 1:71-87. [PMID: 16908979 DOI: 10.1159/000080048] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2003] [Accepted: 02/19/2004] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease and Parkinson's disease are the most common neurodegenerative conditions associated with the ageing process. The pathology of these and other neurodegenerative disorders, including polyglutamine diseases, is characterised by the presence of inclusion bodies in brain tissue of affected patients. In general, these inclusion bodies consist of insoluble, unfolded proteins that are commonly tagged with the small protein, ubiquitin. Covalent tagging of proteins with chains of ubiquitin generally targets them for degradation. Indeed, the ubiquitin/proteasome system (UPS) is the major route through which intracellular proteolysis is regulated. This strongly implicates the UPS in these disease-associated inclusions, either due to malfunction (of specific UPS components) or overload of the system (due to aggregation of unfolded/mutant proteins), resulting in subsequent cellular toxicity. Protein targeting for degradation is a highly regulated process. It relies on transfer of ubiquitin molecules to the target protein via an enzyme cascade and specific recognition of a substrate protein by ubiquitin-protein ligases (E3s). Recent advances in our knowledge gained from the Human Genome Mapping Project have revealed the presence of potentially hundreds of E3s within the human genome. The discovery that parkin, mutations in which are found in at least 50% of patients with autosomal recessive juvenile parkinsonism, is an E3 further highlights the importance of the UPS in neurological disease. To date, parkin is the only E3 confirmed to have a direct causal role in neurodegenerative disorders. However, a number of other (putative) E3s have now been identified that may cause disease directly or interact with neurological disease-associated proteins. Many of these are either lost or mutated in a given disease or fail to process disease-associated mutant proteins correctly. In this review, we will discuss the role(s) of E3s in neurodegenerative disorders.
Collapse
Affiliation(s)
- Helen C Ardley
- Molecular Medicine Unit, University of Leeds, St. James's University Hospital, Leeds, UK.
| | | |
Collapse
|
27
|
Dietrich W, Haitel A, Holzer G, Huber JC, Kolbus A, Tschugguel W. Estrogen receptor-beta is the predominant estrogen receptor subtype in normal human synovia. ACTA ACUST UNITED AC 2006; 13:512-7. [PMID: 16990033 DOI: 10.1016/j.jsgi.2006.07.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Joint pain increases after menopause with more than 50% of woman suffering from arthralgies. Since pain and inflammation of joints originate from synovial tissue, we aimed to discover whether estrogen receptors are present in the human synovia. METHODS This in vitro study was performed on samples of human synovial tissue, obtained from pre- (n = 8) and postmenopausal woman (n = 11) and men (n = 5) following surgery due to traumatic lesions. Fresh synovial tissue specimens were assessed for the localization as well as the presence of estrogen receptor-alpha (ER alpha) and estrogen receptor-beta (ER beta) by means of immunohistochemistry, as well as Western blot and reverse transcriptase-polymerase chain reaction (RT-PCR), respectively. RESULTS ER beta protein and mRNA were found to be equally and highly expressed in synovial stroma and lining cells of all explants independent of sex or menopausal status. In contrast, weak ER alpha staining was localized in the synovial lining cells in only three of 24 explants. ER alpha protein was found to be weakly expressed in three of ten explants. ER alpha mRNA was found with highly variable amounts in seven of ten explants. CONCLUSION In view of our observation that ER beta but not ER alpha is expressed regularly in normal human synovia in high amounts, we propose that estrogen could play a significant role in synovial membrane function in women and men, operating preferably via the ER beta isoform.
Collapse
Affiliation(s)
- Wolf Dietrich
- Department of Obstetrics and Gynecology, Division of Gynecologic Endocrinology and Reproductive Medicine, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
28
|
Affiliation(s)
- Matthias R Meyer
- Department of Internal Medicine, Medical Policlinic, University Hospital Zurich, Switzerland
| | | | | |
Collapse
|
29
|
Ducza E, Kormányos Z, Resch BE, Falkay G. Correlation between the alterations in the mRNA expressions of the α1-adrenoceptor and estrogen receptor subtypes in the pregnant human uterus and cervix. Eur J Pharmacol 2005; 528:183-7. [PMID: 16325176 DOI: 10.1016/j.ejphar.2005.10.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Revised: 10/21/2005] [Accepted: 10/26/2005] [Indexed: 11/26/2022]
Abstract
Our present aim was to determine the association between the mRNA expressions of the estrogen and adrenoceptor subtypes in the pregnant human uterus and cervix. The presence of the mRNA expressions of all the alpha1-adrenoceptor and estrogen receptor subtypes in the uterus and cervix was proved by means of a reverse transcription polymerase chain reaction method, with a predominance of the mRNAs of the alpha1B-adrenoceptor and estrogen alpha receptors, respectively. The change in the mRNA expression of the estrogen receptor alpha correlated strongly with the change in mRNA level of the alpha1B-adrenoceptors. We presume that the expression of the alpha1B-adrenoceptors at 33-34 weeks in the pregnant human uterus is regulated by estrogen through the estrogen receptor alpha subtypes.
Collapse
Affiliation(s)
- Eszter Ducza
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, H-6720 Szeged, Eötvös u. 6, Hungary
| | | | | | | |
Collapse
|
30
|
Dimayuga FO, Reed JL, Carnero GA, Wang C, Dimayuga ER, Dimayuga VM, Perger A, Wilson ME, Keller JN, Bruce-Keller AJ. Estrogen and brain inflammation: effects on microglial expression of MHC, costimulatory molecules and cytokines. J Neuroimmunol 2005; 161:123-36. [PMID: 15748951 DOI: 10.1016/j.jneuroim.2004.12.016] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2004] [Revised: 11/29/2004] [Accepted: 12/20/2004] [Indexed: 02/07/2023]
Abstract
To model the effects of estrogen on adaptive immunity in the brain, we examined the effects of 17beta-estradiol on microglial parameters related to antigen presentation and T cell activation. Specifically, the effects of 17beta-estradiol on basal and LPS-induced surface staining of Class I and II MHC, as well as CD40, CD80, CD86, CD152, CD28, CD8, CD11b, Fas, FasL, and also ERalpha and ERbeta, were examined in N9 microglial cells. Additionally, the effects of 17beta-estradiol on basal and LPS-induced release of cytokines (TNF-alpha, IFN-gamma, IL-2, IL-4, and IL-10) were determined. Data indicate that estrogen increases IL-10 while decreasing TNFalpha and IFNgamma release from resting and LPS-stimulated N9 cells. Additionally, LPS-induced surface staining of MHC Class I, CD40, and CD86 was significantly attenuated by estrogen pretreatment. The basal percentage of cells positive for MHC Class I and II, CD40, and CD152, Fas, and FasL was significantly decreased by estrogen exposure. However, CD8, CD86, CD11b, and CD28 were unaffected by estrogen, and CD80 cell surface staining significantly increased following estrogen exposure. Taken together, these data indicate that estrogen can significantly decrease components of adaptive immunity in microglial cells, and highlight the multi-faceted regulatory effects of estrogen on microglial parameters related to antigen presentation and T cell interaction.
Collapse
Affiliation(s)
- Filomena O Dimayuga
- Department of Anatomy and Neurobiology, University of Kentucky, MN 222 Chandler Medical Center, Lexington, KY 40536-0298, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Myasthenia gravis (MG) is an autoimmune disease associated with thymic hyperplasia and is much more prevalent in women than men. In this study we investigated potential changes in estrogen receptor (ER) expression in thymic hyperplasia. We first quantified by real-time PCR the relative expression of ER alpha and ER beta in normal thymus and found that the ER beta to ER alpha ratio was inverted in thymocytes (8.6 +/- 1.2), compared with thymic epithelial cells (0.18 +/- 0.05). The ER transcript number gradually decreased in thymic epithelial cells during culture, indicating that the thymic environment influences ER expression. CD4+ helper T cells expressed higher level of ERs, compared with CD8+ cells, as assessed by flow cytometry in thymocytes and peripheral blood mononuclear cells. In MG patients, we found an increased expression of ER alpha on thymocytes and both ERs on T cells from peripheral blood mononuclear cells, indicating that the signals provided by thymic and peripheral microenvironments are distinct. Finally, activation of normal thymocytes by proinflammatory cytokines induced increased expression of ERs especially in the CD4+ subset, suggesting that an excess of proinflammatory cytokines could explain the increase of ERs expression on MG lymphocytes. The dysregulation of ER expression in MG lymphocytes could affect the maintenance of the homeostatic conditions and might influence the progression of the autoimmune response.
Collapse
Affiliation(s)
- Patrice Nancy
- Centre National de la Recherche Scientifique-Unité Mixte de Recherche-8078, Institut Paris-Sud Cytokines, Université Paris XI, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | | |
Collapse
|
32
|
Magness RR, Phernetton TM, Gibson TC, Chen DB. Uterine blood flow responses to ICI 182 780 in ovariectomized oestradiol-17beta-treated, intact follicular and pregnant sheep. J Physiol 2005; 565:71-83. [PMID: 15774510 PMCID: PMC1464500 DOI: 10.1113/jphysiol.2005.086439] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Oestrogen dramatically increases uterine blood flow (UBF) in ovariectomized (Ovx) ewes. Both the follicular phase and pregnancy are normal physiological states with elevated levels of circulating oestrogen. ICI 182 780 is a pure steroidal oestrogen receptor (ER) antagonist that blocks oestrogenic actions in oestrogen-responsive tissue. We hypothesized that an ER-mediated mechanism is responsible for in vivo rises in UBF in physiological states of high oestrogen. The purpose of the study was to examine the effect of an ER antagonist on exogenous and endogenous oestradiol-17beta (E2beta)-mediated elevations in UBF. Sheep were surgically instrumented with bilateral uterine artery blood flow transducers, and uterine and femoral artery catheters. Ovx animals (n = 8) were infused with vehicle (35% ethanol) or ICI 182 780 (0.1-3.0 microg min(-1)) into one uterine artery for 10 min before and 50 min after E2beta was given (1 microg kg(-1) I.V. bolus) and UBF was recorded for an additional hour. Intact, cycling sheep were synchronized to the follicular phase using progesterone, prostaglandin F2alpha(PGF2alpha) and pregnant mare serum gonadotrophin (PMSG). When peri-ovulatory rises in UBF reached near peak levels, ICI 182 780 (1 or 2 microg (ml uterine blood flow)-1) was infused unilaterally (n = 4 sheep). Ewes in the last stages of pregnancy (late pregnant ewes) were also given ICI 182 780 (0.23-2.0 microg (ml uterine blood flow)-1; 60 min infusion) into one uterine artery (n = 8 sheep). In Ovx sheep, local infusion of ICI 182 780 did not alter systemic cardiovascular parameters, such as mean arterial blood pressure or heart rate; however, it maximally decreased ipsilateral, but not contralateral, UBF vasodilatory responses to exogenous E2beta by approximately 55-60% (P < 0.01). In two models of elevated endogenous E2beta, local ICI 182 780 infusion inhibited the elevated UBF seen in follicular phase and late pregnant ewes in a time-dependent manner by approximately 60% and 37%, respectively; ipsilateral >> contralateral effects (P < 0.01). In late pregnant sheep ICI 182 780 also mildly and acutely (for 5-30 min) elevated mean arterial pressure and heart rate (P < 0.05). We conclude that exogenous E2beta-induced increases in UBF in the Ovx animal and endogenous E2beta-mediated elevations of UBF during the follicular phase and late pregnancy are partially mediated by ER-dependent mechanisms.
Collapse
Affiliation(s)
- Ronald R Magness
- Department of Obstetrics and Gynecology, Perinatal Research Laboratories, University of Wisconsin-Madison, Atrium B Meriter Hospital, Madison, WI 53715, USA.
| | | | | | | |
Collapse
|
33
|
Liao WX, Magness RR, Chen DB. Expression of estrogen receptors-alpha and -beta in the pregnant ovine uterine artery endothelial cells in vivo and in vitro. Biol Reprod 2004; 72:530-7. [PMID: 15564597 DOI: 10.1095/biolreprod.104.035949] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Estrogen is recognized to be one of the driving forces in increases in uterine blood flow through both rapid and delayed actions via binding to its receptors, ER alpha and ER beta at the uterine artery (UA) wall, and especially in UA endothelium (UAE). However, information regarding estrogen receptor (ER) expression in UAE is limited. This study was designed to test whether ERs are expressed in UAE in vivo, and if they are, whether these receptors are maintained in cultured UA endothelial cells (UAECs) in vitro. By using immunohistochemical and Western blot analyses, we clearly demonstrated ER alpha and ER beta protein expression in pregnant (Days 120-130) sheep UA and UAE in vivo and as well as cultured UAECs in vitro. Reverse transcription-polymerase chain reaction (RT-PCR) amplified both ER alpha and ER beta mRNAs in UA, UAE, and UAECs. Of interest, a truncated ER beta (ER beta2) variant due to a splicing deletion of exon 5 of the ER beta gene was detected in these cells. Quantitative RT-PCR analysis revealed that ER alpha mRNA levels are approximately 8-fold (P < 0.01) higher than that of ER beta in UAECs, indicating that ER alpha may play a more important role than ER beta in the UAEC responses to estrogen. Fluorescence immunolabeling analysis showed that ER alpha is present in both nuclei and plasma membranes in UAECs, and the latter is also colocalized with caveolin-1. The membrane and nuclear ER alpha presumably participate in rapid and delayed responses, respectively, to estrogen on UAE. Taken together, our data demonstrated that UAE is a direct target of estrogen actions and that the UAEC culture model we established is suitable for dissecting estrogen actions on UAE.
Collapse
Affiliation(s)
- Wu Xiang Liao
- Department of Reproductive Medicine, University of California San Diego, La Jolla, California 92093-0802, USA
| | | | | |
Collapse
|
34
|
Baker AE, Brautigam VM, Watters JJ. Estrogen modulates microglial inflammatory mediator production via interactions with estrogen receptor beta. Endocrinology 2004; 145:5021-32. [PMID: 15256495 DOI: 10.1210/en.2004-0619] [Citation(s) in RCA: 175] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogens are well known to exert antiinflammatory effects outside the central nervous system (CNS). They have also been shown to exert neuroprotective effects in the CNS after several types of injury, including neurodegeneration. However, the molecular mechanisms by which these effects occur remain unclear. Because microglial hyperactivation and their production of neurotoxins is associated with many types of brain injury for which estrogens are beneficial, we sought to investigate the ability of estrogen to modulate microglial function. Furthermore, because little is known regarding the role of each of the two known estrogen receptors (ERs) in microglia, our studies were designed to test the hypothesis that 17beta-estradiol (E(2)) exerts antiinflammatory effects in microglia, specifically via interactions with ERbeta. We tested this hypothesis using the murine microglial cell line BV-2, which naturally expresses only ERbeta. Our results indicate that not only does E(2) decrease lipopolysaccharide (LPS)-stimulated nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) expression, it also reduces the expression of cyclooxygenase-2, a target for estrogen that has not previously been reported for ERbeta. We also observed that LPS-stimulated TNFalpha mRNA was increased by estrogen. E(2) exerts these effects within 30 min compared with typical estrogen transcriptional responses. Tamoxifen and ICI 182,780 differentially blocked the inhibitory effects of E(2) on LPS-stimulated iNOS and cyclooxygenase-2. In addition, we show that E(2) alters LPS-stimulated MAPK pathway activation, supporting the idea that alterations in the MAPKs may be a potential mechanism by which ERbeta mediates decreased microglial activation.
Collapse
Affiliation(s)
- Ann E Baker
- Department of Comparative Biosciences, 2015 Linden Drive, Madison, Wisconsin 53706, USA.
| | | | | |
Collapse
|
35
|
Liang M, Nilsson BO. Proteasome-dependent degradation of ERalpha but not ERbeta in cultured mouse aorta smooth muscle cells. Mol Cell Endocrinol 2004; 224:65-71. [PMID: 15353181 DOI: 10.1016/j.mce.2004.06.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2004] [Revised: 06/01/2004] [Accepted: 06/29/2004] [Indexed: 02/07/2023]
Abstract
Here we investigate ERalpha and ERbeta expression and regulation in vascular smooth muscle cells from mouse aorta. Immunocytochemistry showed nuclear staining for both ERalpha and ERbeta. Double stainings revealed co-expression of ERalpha and ERbeta in vascular smooth muscle cells. ERalpha (66 kDa) and ERbeta (54 kDa) expression determined by Western blotting was unchanged within 7 h after inhibition of protein synthesis with cycloheximide in the absence of 17beta-estradiol (E(2)), showing that both proteins are stable without ligand-binding. Treatment with 10 nM E(2) for 7 h in the presence of cycloheximide increased ERalpha, suggesting that E(2) causes a conformational change in the ERalpha protein. The ERbeta was not affected by E(2). Treatment with the proteasome inhibitor epoxomicin (100 nM) for 3 days caused a prominent upregulation of ERalpha both in the absence and in the presence of E(2), while ERbeta was unaffected, suggesting that ERalpha but not ERbeta is degraded by ubiquitin-proteasome system in vascular smooth muscle cells. In summary, we disclose a short-term regulation of ERalpha protein by estrogen and that ERalpha but not ERbeta is degraded via the ubiquitin-proteasome pathway in vascular smooth muscle cells.
Collapse
Affiliation(s)
- Min Liang
- Department of Physiological Sciences, Lund University, BMC F12, SE-221 84 Lund, Sweden.
| | | |
Collapse
|
36
|
Förster C, Kietz S, Hultenby K, Warner M, Gustafsson JA. Characterization of the ERbeta-/-mouse heart. Proc Natl Acad Sci U S A 2004; 101:14234-9. [PMID: 15375213 PMCID: PMC521141 DOI: 10.1073/pnas.0405571101] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although the heart responds to estrogen, it is not clear whether estrogen acts directly on heart muscle or indirectly by means of the vascular, immune, or nervous system. No role for estrogen receptor (ER) beta in the heart has been established, but ERbeta(-/-) mice are hypertensive, and as they age, their hearts become enlarged. Histological and ultrastructural analysis of the heart revealed a disarray of myocytes, a disruption of intercalated discs, an increase in the number and size of gap junctions, and a profound alteration in nuclear structure, concomitantly with a loss of expression of lamin A/C from the nuclear envelope. In the lungs of ERbeta(-/-) mice, lamin A/C was located in the nuclear membrane, indicating that lamin A/C is not an ERbeta-regulated gene. Immunohistochemical studies with ERbeta antibodies failed to detect ERbeta in the myocardium. We conclude that abnormalities in heart morphology in ERbeta(-/-) mice are likely due to stress on the nuclear envelope as a result of the chronic sustained systolic and diastolic hypertension observed in ERbeta(-/-) mice. Because neither ERalpha nor ERbeta could be detected in heart muscle, the effects of estrogen on the myocardium seem to be indirect.
Collapse
Affiliation(s)
- Carola Förster
- Department of Medical Nutrition, Karolinska Institutet, Novum, S-141 86 Huddinge, Sweden
| | | | | | | | | |
Collapse
|
37
|
Salli U, Reddy AP, Salli N, Lu NZ, Kuo HC, Pau FKY, Wolf DP, Bethea CL. Serotonin neurons derived from rhesus monkey embryonic stem cells: similarities to CNS serotonin neurons. Exp Neurol 2004; 188:351-64. [PMID: 15246835 DOI: 10.1016/j.expneurol.2004.04.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2003] [Revised: 04/01/2004] [Accepted: 04/15/2004] [Indexed: 11/24/2022]
Abstract
We sought an in vitro primate model for serotonin neurons. Rhesus monkey embryonic stem (ES) cell colonies were isolated and differentiated into embryoid bodies (EBs), then transferred to serum-free medium with 1% insulin-transferrin-selenium for 7 days to induce neural precursor cell (NPC) formation. NPCs were cultured in medium with 1% N-2 neural supplement and human fibroblast growth factor 2 (FGF2, 10 ng/ml) for 7 days to stimulate cell proliferation. Lastly, NPCs were dispersed into single cells and cultured without FGF2 for another 7 days to obtain terminal differentiation. Terminal cells were characterized for neuronal and serotonergic markers. Over 95% of the NPCs were immunopositive for nestin and Musashi1. Terminally differentiated cells appeared in both small and large morphologies. Most (>95%) of the mature cells (both small and large) were immunopositive for neuron-specific nuclear protein (NeuN), synaptophysin, microtubule-associated protein (MAP2C), Tau-1, neurofilament 160 (NF-160), beta-tubulin (TujIII), tryptophan hydroxylase (TPH), serotonin, the serotonin reuptake transporter (SERT), estrogen receptor-beta (ERbeta), and progestin receptor (PR), but not estrogen receptor-alpha (ERalpha). Less than 2-3% of cells were positive for tyrosine hydroxylase (TH). Reverse transcriptase polymerase chain reaction (RT-PCR) detected mRNA transcripts for TPH-1, TPH-2, SERT, 5-HT1A-autoreceptor, ERbeta, and PR in the differentiated population. A low level of expression of ERalpha mRNA was also detected. Quantitative RT-PCR indicated that the relative abundance of TPH-2 mRNA was greater than TPH-1 mRNA. Serotonin as measured by ELISA increased 3-fold in the mature stage compared to the selection and expansion stages. In summary, a remarkably high percentage of cells derived from monkey ES cells exhibited neuronal plus serotonergic markers as well as nuclear steroid receptors similar to primate CNS serotonin neurons, suggesting that these cells may serve as a useful primate model for serotonergic neurons.
Collapse
Affiliation(s)
- Ugur Salli
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton 97006, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Suzuki S, Handa RJ. Regulation of estrogen receptor-beta expression in the female rat hypothalamus: differential effects of dexamethasone and estradiol. Endocrinology 2004; 145:3658-70. [PMID: 15087431 DOI: 10.1210/en.2003-1688] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogen and glucocorticoids interact in multiple aspects of endocrine regulation by exerting opposing influences on the expression of selective genes. In rats, estrogen receptor (ER)-beta is the predominant form of ER present in the hypothalamic paraventricular (PVN) and supraoptic (SON) nuclei, suggesting its involvement in neuroendocrine regulation. To date, the hormonal regulatory profile of the ERbeta gene in the rat central nervous system has not been closely elucidated. In the present study, we first examined the effects of dexamethasone (DEX) and estradiol benzoate (EB) on the ERbeta protein expression in the PVN and SON of ovariectomized female rats. In the SON and parvocellular and magnocellular parts of the PVN, the number of ERbeta immunoreactive nuclei significantly increased after DEX treatment, compared with the control group, whereas EB treatment caused a significant decrease. The effect of EB was consistent across other brain nuclei such as the anteroventral periventricular nucleus and medial preoptic nucleus. To determine the molecular level at which DEX and EB control ERbeta expression, we examined the effects of these steroids on ERbeta mRNA levels using real-time RT-PCR. EB significantly decreased the expression of ERbeta mRNA in the PVN (P = 0.0006) and SON (P < 0.01). In contrast, DEX did not change ERbeta mRNA levels. These results indicate that glucocorticoids and estrogen exert opposing regulatory influences on the ERbeta gene expression. This may represent a mechanism by which these steroids can alter the cellular sensitivity of ERbeta-expressing neurons to subsequent steroidal activation.
Collapse
Affiliation(s)
- Shotaro Suzuki
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | | |
Collapse
|
39
|
Pinzone JJ, Stevenson H, Strobl JS, Berg PE. Molecular and cellular determinants of estrogen receptor alpha expression. Mol Cell Biol 2004; 24:4605-12. [PMID: 15143157 PMCID: PMC416410 DOI: 10.1128/mcb.24.11.4605-4612.2004] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Joseph J Pinzone
- The George Washington University School of Medicine, Department of Biochemistry and Molecular Biology, Ross Hall, Washington, D.C. 20037, USA
| | | | | | | |
Collapse
|
40
|
|
41
|
Korovkina VP, Brainard AM, Ismail P, Schmidt TJ, England SK. Estradiol binding to maxi-K channels induces their down-regulation via proteasomal degradation. J Biol Chem 2003; 279:1217-23. [PMID: 14555652 DOI: 10.1074/jbc.m309158200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Estrogens exert their biological action via both genomic and non-genomic mechanisms. Proteins different from classical estradiol receptors are believed to mediate the latter effects. Here we demonstrate that the maxi-K channel functions as an estrogen-binding protein in transfected HEK293 cells. Whole-cell maxi-K channel currents and protein expression were attenuated by exposure to either 17alpha- or 17beta-estradiol. This effect was dose-dependent for 17beta-estradiol at concentrations ranging from 10 nm to 1 microm, while 17alpha-estradiol inhibited channel expression only at 1 microm. These effects were mediated by direct low affinity binding of estradiol to the maxi-K channel but not to its accessory beta1-subunit, as revealed by cell membrane estradiol binding assays. However, specific binding of estradiol to the channel was facilitated by the presence of the beta1 subunit. Addition of MG-132, a blocker of proteasomal degradation, stabilized channel expression. These data suggest that channel down-regulation is mediated by estrogen-induced proteasomal degradation, similar to the pathway used for estrogen receptor degradation. Membrane expression of endogenous maxi-K channels in cultured vascular smooth muscle cells was also attenuated by prolonged exposure to 17alpha- and 17beta-estradiol. Thus our studies demonstrate that estrogen binds to maxi-K channels and may directly regulate channel expression and function. These results will have important implications in understanding estradiol-induced effects in multiple tissues including vascular smooth muscle.
Collapse
Affiliation(s)
- Victoria P Korovkina
- Department of Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|