1
|
Mishra S, Chaube R. Impact of ovariectomy and estradiol-17β (E2) replacement on the brain steroid levels of the Indian stinging catfish Heteropneustes fossilis. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2022.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
2
|
Molehin D, Filleur S, Pruitt K. Regulation of aromatase expression: Potential therapeutic insight into breast cancer treatment. Mol Cell Endocrinol 2021; 531:111321. [PMID: 33992735 DOI: 10.1016/j.mce.2021.111321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/14/2021] [Accepted: 05/09/2021] [Indexed: 12/13/2022]
Abstract
Estrogen signaling has been implicated in hormone-dependent breast cancer which constitutes >75% of breast cancer diagnosis and other malignancies. Aromatase, the key enzyme involved in the synthesis of estrogen, is often dysregulated in breast cancers. This has led to the administration of aromatase-inhibitors (AIs), commonly used for hormone-dependent breast cancers. Unfortunately, the increasing development of acquired resistance to the current AIs and modulators of estrogen receptors, following initial disease steadiness, has posed a serious clinical challenge in breast cancer treatment. In this review we highlight historical and recent advances on the transcriptional and post-translational regulation of aromatase in both physiological and pathological contexts. We also discuss the different drug combinations targeting various tumor promoting cell signaling pathways currently being developed and tested both in laboratory settings and in the clinic.
Collapse
Affiliation(s)
- Deborah Molehin
- Department of Immunology & Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Stephanie Filleur
- Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, USA
| | - Kevin Pruitt
- Department of Immunology & Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
3
|
Nadal A, Fuentes E, Ripoll C, Villar-Pazos S, Castellano-Muñoz M, Soriano S, Martinez-Pinna J, Quesada I, Alonso-Magdalena P. Extranuclear-initiated estrogenic actions of endocrine disrupting chemicals: Is there toxicology beyond paracelsus? J Steroid Biochem Mol Biol 2018; 176:16-22. [PMID: 28159674 DOI: 10.1016/j.jsbmb.2017.01.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 01/16/2017] [Accepted: 01/20/2017] [Indexed: 12/21/2022]
Abstract
Endocrine Disrupting Chemicals (EDCs), including bisphenol-A (BPA) do not act as traditional toxic chemicals inducing massive cell damage or death in an unspecific manner. EDCs can work upon binding to hormone receptors, acting as agonists, antagonists or modulators. Bisphenol-A displays estrogenic activity and, for many years it has been classified as a weak estrogen, based on the classic transcriptional action of estrogen receptors serving as transcription factors. However, during the last two decades our knowledge about estrogen signaling has advanced considerably. It is now accepted that estrogen receptors ERα and ERβ activate signaling pathways outside the nucleus which may or may not involve transcription. In addition, a new membrane estrogen receptor, GPER, has been proposed. Pharmacological and molecular evidence, along with results obtained in genetically modified mice, demonstrated that BPA, and its substitute BPS, are potent estrogens acting at nanomolar concentrations via extranuclear ERα, ERβ, and GPER. The different signaling pathways activated by BPA and BPS explain the well-known estrogenic effects of low doses of EDCs as well as non-monotonic dose-response relationships. These signaling pathways may help to explain the actions of EDCs with estrogenic activity in the etiology of different pathologies, including type-2 diabetes and obesity.
Collapse
Affiliation(s)
- Angel Nadal
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain.
| | - Esther Fuentes
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain; Departamento de Biología Aplicada, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Cristina Ripoll
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Sabrina Villar-Pazos
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Manuel Castellano-Muñoz
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Sergi Soriano
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Alicante, Spain
| | - Juan Martinez-Pinna
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Alicante, Spain
| | - Ivan Quesada
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain; Departamento de Biología Aplicada, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Paloma Alonso-Magdalena
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Departamento de Biología Aplicada, Universidad Miguel Hernández de Elche, Alicante, Spain
| |
Collapse
|
4
|
Sołtysik K, Czekaj P. ERα36--Another piece of the estrogen puzzle. Eur J Cell Biol 2015; 94:611-25. [PMID: 26522827 DOI: 10.1016/j.ejcb.2015.10.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 09/03/2015] [Accepted: 10/09/2015] [Indexed: 12/22/2022] Open
Abstract
Although the nuclear action of estrogen receptors (ER) is a well-known fact, evidence supporting membrane estrogen receptors is steadily accumulating. New ER variants of unrecognized function have been discovered. ERα is a product of the ESR1 gene. It serves not only as a template for the full-length 66kDa protein, but also for smaller isoforms which exist as independent receptors. The recently discovered ERα36 (36kDa), consisting of 310 amino acids of total 595 ERα66 protein residues, is an example of that group. The transcription initiation site is identified in the first intron of the ESR1 gene. C-Terminal 27 amino acids are encoded by previously unknown exon 9. The presence of this unique C-terminal sequence creates an opportunity for the production of selective antibodies. ERα36 has been shown to have a high affinity to the cell membrane and as much as 90% of the protein can be bound with it. Post-translational palmitoylation is suspected to play a crucial role in ERα36 anchoring to the cell membrane. In silico analysis suggests the existence of a potential transmembrane domain in ERα36. ERα36 was found in most cells of animals at various ages, but its exact physiological function remains to be fully elucidated. It seems that cells traditionally considered as being deprived of ER are able to respond to hormonal stimulation via the ERα36 receptor. Moreover, ERα36 displays unique pharmacological properties and its action may be behind antiestrogen resistance. The use of ERα36 in cancer diagnosis gives rise to great expectations.
Collapse
Affiliation(s)
- Kamil Sołtysik
- Students Scientific Society, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Piotr Czekaj
- Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland.
| |
Collapse
|
5
|
Abstract
Traditionally, steroid hormones such as the vitamin D3 metabolites, testosterone and dihydrotesterone, and 17β-estradiol act through cytosolic and nuclear receptors that directly interact with DNA to alter gene transcription and regulate cellular development. However, recent studies focused on rapid and membrane effects of steroid hormones have given invaluable insight into their non-classical mechanisms of action. In some cases, the traditional receptors were implicated as acting also in the plasma membrane as membrane-associated receptors. However, recent data have demonstrated the presence of an alternative splicing variant to traditional estrogen receptor α known as ERα36, which is present in the plasma membranes of several different cell types including several cancer cell types and even in some normal cells including cartilage and bone cells. The physiological effects that result from the membrane activation of ERα36 may vary from one cell type to another, but the mechanism of action appears to use similar pathways such as the activation of various protein kinases and phospholipases leading to the activation of signaling cascades that result in rapid, non-genomic responses. These rapid responses can affect cell proliferation and apoptotic signaling, indirectly activate downstream genomic signaling through phosphorylation cascades of transcription factors, and crosstalk with classical pathways via interaction with classical receptors. This review describes the data from the last several years and discusses the non-classical, rapid, and membrane-associated cellular responses to steroid hormones, particularly 17β-estradiol, through the classical receptors ERα and ERβ and various non-classical receptors, especially estrogen receptor-α36 (ERα36).
Collapse
Affiliation(s)
- Reyhaan A Chaudhri
- School of Biology, Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA; Atlanta Clinical and Translational Science Institute, Emory University, 1440 Clifton Rd NE, Atlanta, GA 30322, USA
| | - Nofrat Schwartz
- Department of Otolaryngology, Meir Hospital, Tchernichovsky 59, Kfar Saba 44299, Israel
| | - Khairat Elbaradie
- School of Biology, Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA; Department of Zoology, Tanta University, 69 Tout Ankh Amoon St, Tanta 31111, Egypt
| | - Zvi Schwartz
- School of Engineering, Virginia Commonwealth University, 601 West Main Street, Suite 331, Richmond, VA 23284, USA; Department of Periodontics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MSC 7894, San Antonio, TX 78229, USA
| | - Barbara D Boyan
- School of Biology, Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA; School of Engineering, Virginia Commonwealth University, 601 West Main Street, Suite 331, Richmond, VA 23284, USA.
| |
Collapse
|
6
|
Yoon K, Kwack SJ, Kim HS, Lee BM. Estrogenic endocrine-disrupting chemicals: molecular mechanisms of actions on putative human diseases. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2014; 17:127-174. [PMID: 24749480 DOI: 10.1080/10937404.2014.882194] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Endocrine-disrupting chemicals (EDC), including phthalates, bisphenol A (BPA), phytoestrogens such as genistein and daidzein, dichlorodiphenyltrichloroethane (DDT), and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are associated with a variety of adverse health effects in organisms or progeny by altering the endocrine system. Environmental estrogens, including BPA, phthalates, and phytoestrogens, are the most extensively studied and are considered to mimic the actions of endogenous estrogen, 17β-estradiol (E2). Diverse modes of action of estrogen and estrogen receptors (ERα and ERβ) have been described, but the mode of action of estrogenic EDC is postulated to be more complex and needs to be more clearly elucidated. This review examines the adverse effects of estrogenic EDC on male or female reproductive systems and molecular mechanisms underlying EDC effects that modulate ER-mediated signaling. Mechanisms of action for estrogenic EDC may involve both ER-dependent and ER-independent pathways. Recent findings from systems toxicology of examining estrogenic EDC are also discussed.
Collapse
Affiliation(s)
- Kyungsil Yoon
- a Lung Cancer Branch , Research Institute, National Cancer Center , Goyang , Gyeonggi-do , South Korea
| | | | | | | |
Collapse
|
7
|
Micevych P, Christensen A. Membrane-initiated estradiol actions mediate structural plasticity and reproduction. Front Neuroendocrinol 2012; 33:331-41. [PMID: 22828999 PMCID: PMC3496015 DOI: 10.1016/j.yfrne.2012.07.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 07/11/2012] [Accepted: 07/13/2012] [Indexed: 12/17/2022]
Abstract
Over the years, our ideas about estrogen signaling have greatly expanded. In addition to estradiol having direct nuclear actions that mediate transcription and translation, more recent experiments have demonstrated membrane-initiated signaling. Both direct nuclear and estradiol membrane signaling can be mediated by the classical estrogen receptors, ERα and ERβ, which are two of the numerous putative membrane estrogen receptors. Thus far, however, only ERα has been shown to play a prominent role in regulating female reproduction and sexual behavior. Because ERα is a ligand-gated transcription factor and not a typical membrane receptor, trafficking to the cell membrane requires post-translational modifications. Two necessary modifications are palmitoylation and association with caveolins, a family of scaffolding proteins. In addition to their role in trafficking, caveolin proteins also serve to determine ERα interactions with metabotropic glutamate receptors (mGluRs). It is through these complexes that ERα, which cannot by itself activate G proteins, is able to initiate intracellular signaling. Various combinations of ERα-mGluR interactions have been demonstrated throughout the nervous system from hippocampus to striatum to hypothalamus to dorsal root ganglion (DRG) in both neurons and astrocytes. These combinations of ER and mGluR allow estradiol to have both facilitative and inhibitory actions in neurons. In hypothalamic astrocytes, the estradiol-mediated release of intracellular calcium stores regulating neurosteroid synthesis requires ERα-mGluR1a interaction. In terms of estradiol regulation of female sexual receptivity, activation of ERα-mGluR1a signaling complex leads to the release of neurotransmitters and alteration of neuronal morphology. This review will examine estradiol membrane signaling (EMS) activating a limbic-hypothalamic lordosis regulating circuit, which involves ERα trafficking, internalization, and modifications of neuronal morphology in a circuit that underlies female sexual receptivity.
Collapse
Affiliation(s)
- Paul Micevych
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1763, United States.
| | | |
Collapse
|
8
|
Zhang Y, Lü N, Zhao ZQ, Zhang YQ. Involvement of estrogen in rapid pain modulation in the rat spinal cord. Neurochem Res 2012; 37:2697-705. [PMID: 22903468 DOI: 10.1007/s11064-012-0859-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 07/17/2012] [Accepted: 07/28/2012] [Indexed: 12/16/2022]
Abstract
The pivotal role of estrogens in the pain sensitivity has been investigated in many ways. Traditionally, it is ascribed to the slow genomic changes mediated by classical nuclear estrogen receptors (ER), ERα and ERβ, depending on peripheral estrogens. Recently, it has become clear that estrogens can also signal through membrane ERs (mERs), such as G-protein-coupled ER1 (GPER1), mediating the non-genomic effects. However, the spinal specific role played by ERs and the underlying cellular mechanisms remain elusive. The present study investigated the rapid estrogenic regulation of nociception at the spinal level. Spinal administration of 17β-estradiol (E2), the most potent natural estrogen, acutely produced a remarkable mechanical allodynia and thermal hyperalgesia without significant differences among male, female and ovariectomized (Ovx) rats. E2-induced the pro-nociceptive effects were partially abrogated by ICI 182,780 (ERs antagonist), and mimicked by E2-BSA (a mER agonist). Inhibition of local E2 synthesis by 1,4,6-Androstatrien-3,17-dione (ATD, a potent irreversible aromatase inhibitor), or blockade of ERs by ICI 182,780 produced an inhibitory effect on the late phase of formalin nociceptive responses. Notably, lumbar puncture injection of G15 (a selective GPER1 antagonist) resulted in similar but more efficient inhibition of formalin nociceptive responses as compared with ICI 182,780. At the cellular level, the amplitude and decay time of spontaneous inhibitory postsynaptic currents were attenuated by short E2 or E2-BSA treatment in spinal slices. These results indicate that estrogen acutely facilitates nociceptive transmission in the spinal cord via activation of membrane-bound estrogen receptors.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, China
| | | | | | | |
Collapse
|
9
|
Ropero AB, Pang Y, Alonso-Magdalena P, Thomas P, Nadal A. Role of ERβ and GPR30 in the endocrine pancreas: A matter of estrogen dose. Steroids 2012; 77:951-8. [PMID: 22306576 DOI: 10.1016/j.steroids.2012.01.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 01/16/2012] [Accepted: 01/19/2012] [Indexed: 12/17/2022]
Abstract
The endocrine pancreas has emerged as a target for estrogens. The functions of pancreatic α-, β- and δ-cells are modulated by the endogenous hormone, 17β-estradiol (E2). Low physiological concentrations (100pM-1nM) of E2 rapidly decrease the activity of the ATP-sensitive potassium channel (K(ATP)) and enhance glucose-induced insulin release in β-cells in an estrogen receptor β (ERβ)-dependent manner. In addition to the insulinotropic action of ERβ, the newly described estrogen receptor, GPR30, is involved in the insulinotropic effects of high doses of E2 (100nM-5μM). The specific GPR30 agonist G1 also increases insulin secretion in β-cells. Low glucose-induced calcium oscillations and glucagon secretion are suppressed by E2. The effects on glucagon secretion may be mediated by GPR30. Somatostatin release is also decreased by E2 and G1. In this review we summarize all the data published up to date on the rapid insulinotropic effects of estrogens in the endocrine pancreas and propose a model to integrate the estrogen actions mediated through both receptors.
Collapse
Affiliation(s)
- Ana B Ropero
- Instituto de Bioingeniería and CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain.
| | | | | | | | | |
Collapse
|
10
|
Bisphenol A interferes with synaptic remodeling. Front Neuroendocrinol 2010; 31:519-30. [PMID: 20609373 PMCID: PMC2964437 DOI: 10.1016/j.yfrne.2010.06.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 06/11/2010] [Accepted: 06/25/2010] [Indexed: 11/21/2022]
Abstract
The potential adverse effects of Bisphenol A (BPA), a synthetic xenoestrogen, have long been debated. Although standard toxicology tests have revealed no harmful effects, recent research highlighted what was missed so far: BPA-induced alterations in the nervous system. Since 2004, our laboratory has been investigating one of the central effects of BPA, which is interference with gonadal steroid-induced synaptogenesis and the resulting loss of spine synapses. We have shown in both rats and nonhuman primates that BPA completely negates the ∼ 70-100% increase in the number of hippocampal and prefrontal spine synapses induced by both estrogens and androgens. Synaptic loss of this magnitude may have significant consequences, potentially causing cognitive decline, depression, and schizophrenia, to mention those that our laboratory has shown to be associated with synaptic loss. Finally, we discuss why children may particularly be vulnerable to BPA, which represents future direction of research in our laboratory.
Collapse
|
11
|
Thomas P, Alyea R, Pang Y, Peyton C, Dong J, Berg AH. Conserved estrogen binding and signaling functions of the G protein-coupled estrogen receptor 1 (GPER) in mammals and fish. Steroids 2010; 75:595-602. [PMID: 19931550 PMCID: PMC2885585 DOI: 10.1016/j.steroids.2009.11.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 11/12/2009] [Indexed: 12/01/2022]
Abstract
Recent studies by several research groups have shown that G protein estrogen receptor-1 (GPER) formerly known as GPR30, mediates 17beta-estradiol (E2) activation of signal transduction pathways in a variety of human cancer cells and displays E2 binding typical of a membrane estrogen receptor. However, the importance of GPER as an estrogen receptor has been questioned by Otto and co-workers. Some of the pitfalls in investigating the functions of recombinant steroid membrane receptors that may explain the negative results of these investigators are discussed. The characteristics of GPER have also been investigated in a teleost fish, Atlantic croaker, where it has been shown to mediate E2 inhibition of oocyte maturation. Investigations on newly discovered homologous proteins from distantly related vertebrate groups are valuable for determining their fundamental, evolutionarily conserved functions. Therefore, the functions of croaker and human GPERs were compared. The comparisons show that croaker and human GPER have very similar estrogen binding characteristics, typical of estrogen membrane receptors, and activate the same estrogen signaling pathways via stimulatory G proteins (Gs) resulting in increased cAMP production. These results suggest that the estrogen binding and estrogen signaling functions of GPER arose early in vertebrate evolution, prior to the divergence of the teleosts from the tetrapods, more than 200 million years ago. The finding that estrogen membrane signaling through GPER has been conserved for such a long period in two distantly related vertebrate groups, mammals and fish, suggests that this is a fundamental function of GPER in vertebrates, and likely its major physiological role.
Collapse
Affiliation(s)
- P Thomas
- University of Texas at Austin Marine Science Institute, 750 Channel View Drive, Port Aransas, TX 78373, USA.
| | | | | | | | | | | |
Collapse
|
12
|
Abstract
The aim of this review was to analyze the potential effects of environmental chemicals on homeostatic control related to glycemia and energy balance. Many of the environmental chemicals can mimic or interfere with the action of hormones and are generally referred to as "endocrine disruptors". Among these compounds, polychlorinated biphenyls, dioxins, phthalates and bisphenol-A have been correlated with alterations in blood glucose homeostasis in humans. In rodents it has been demonstrated that small doses of bisphenol-A have profound effects on glucose metabolism. Therefore, this altered blood glucose homeostasis may enhance the development of type 2 diabetes.
Collapse
Affiliation(s)
- Paloma Alonso-Magdalena
- CIBER of Diabetes and Associated Metabolic Diseases, CIBERDEM, lnstitute of Bioengineering, University Miguel Hernandez of Elche, Elche, Spain.
| | | | | | | | | |
Collapse
|
13
|
Jiang P, Kong Y, Zhang XB, Wang W, Liu CF, Xu TL. Glycine receptor in rat hippocampal and spinal cord neurons as a molecular target for rapid actions of 17-beta-estradiol. Mol Pain 2009; 5:2. [PMID: 19138413 PMCID: PMC2651124 DOI: 10.1186/1744-8069-5-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Accepted: 01/12/2009] [Indexed: 12/20/2022] Open
Abstract
Glycine receptors (GlyRs) play important roles in regulating hippocampal neural network activity and spinal nociception. Here we show that, in cultured rat hippocampal (HIP) and spinal dorsal horn (SDH) neurons, 17-β-estradiol (E2) rapidly and reversibly reduced the peak amplitude of whole-cell glycine-activated currents (IGly). In outside-out membrane patches from HIP neurons devoid of nuclei, E2 similarly inhibited IGly, suggesting a non-genomic characteristic. Moreover, the E2 effect on IGly persisted in the presence of the calcium chelator BAPTA, the protein kinase inhibitor staurosporine, the classical ER (i.e. ERα and ERβ) antagonist tamoxifen, or the G-protein modulators, favoring a direct action of E2 on GlyRs. In HEK293 cells expressing various combinations of GlyR subunits, E2 only affected the IGly in cells expressing α2, α2β or α3β subunits, suggesting that either α2-containing or α3β-GlyRs mediate the E2 effect observed in neurons. Furthermore, E2 inhibited the GlyR-mediated tonic current in pyramidal neurons of HIP CA1 region, where abundant GlyR α2 subunit is expressed. We suggest that the neuronal GlyR is a novel molecular target of E2 which directly inhibits the function of GlyRs in the HIP and SDH regions. This finding may shed new light on premenstrual dysphoric disorder and the gender differences in pain sensation at the CNS level.
Collapse
Affiliation(s)
- Peng Jiang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, PR China
| | | | | | | | | | | |
Collapse
|
14
|
Nott SL, Huang Y, Fluharty BR, Sokolov AM, Huang M, Cox C, Muyan M. Do Estrogen Receptor beta Polymorphisms Play A Role in the Pharmacogenetics of Estrogen Signaling? ACTA ACUST UNITED AC 2008; 6:239-259. [PMID: 19337586 DOI: 10.2174/187569208786733820] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Estrogen hormones play critical roles in the regulation of many tissue functions. The effects of estrogens are primarily mediated by the estrogen receptors (ER) alpha and beta. ERs are ligand-activated transcription factors that regulate a complex array of genomic events that orchestrate cellular growth, differentiation and death. Although many factors contribute to their etiology, estrogens are thought to be the primary agents for the development and/or progression of target tissue malignancies. Many of the current modalities for the treatment of estrogen target tissue malignancies are based on agents with diverse pharmacology that alter or prevent ER functions by acting as estrogen competitors. Although these compounds have been successfully used in clinical settings, the efficacy of treatment shows variability. An increasing body of evidence implicates ERalpha polymorphisms as one of the contributory factors for differential responses to estrogen competitors. This review aims to highlight the recent findings on polymorphisms of the lately identified ERbeta in order to provide a functional perspective with potential pharmacogenomic implications.
Collapse
Affiliation(s)
- Stephanie L Nott
- Department of Biochemistry & Biophysics, University of Rochester Medical School, Rochester, NY, 14642, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
The kinetics of neurosteroid binding to recombinant human microtubule-associated protein 2C (rhMAP2C) and neurosteroid regulation of MAP2C-stimulated tubulin assembly were studied. In a quartz crystal microbalance assay, progesterone-BSA at 1-10 nM showed concentration-dependent binding to rhMAP2C, and this binding was competitively inhibited by pregnenolone or progesterone. However, no progesterone-BSA binding to N-terminal 71 amino acid residues rhMAP2C was found. In an rhMAP2C-stimulated tubulin assembly assay, pregnenolone enhanced the assembly of an rhMAP2C-progesterone-BSA complex in a progesterone-reversible manner, progesterone alone had no effect. Although N-terminal 71 amino acid residues rhMAP2C retains an activity to stimulate this assembly, this effect was not affected by pregnenolone or progesterone. These findings suggest that neurosteroids specifically bind to the N-terminus of rhMAP2 and regulate tubulin assembly.
Collapse
|
16
|
Abstract
Estrogen controls multiple biological functions through binding to estrogen receptors (ERs). Traditionally, ERs have been regarded as transcription factors regulating the expression of target genes. However, growing evidence of rapid estrogen's actions in a number of tissues has been accumulating and alternative mechanisms of signal transduction have been proposed. These so called "extra-nuclear actions" do not require gene expression or protein synthesis and are independent of the nuclear localization of ERs. Indeed, some of these actions are elicited by ERs residing at or near the plasma membrane. Membrane-associated molecules such as ion channels, G proteins, the tyrosine kinase c-Src as well as growth factor receptors are modulated by liganded ERs within the membrane, leading to the activation of downstream cascades such as mitogen-activated protein kinase, phosphatidylinositol 3-OH kinase, protein kinase A, and protein kinase C. These cascades mediate some important rapid actions of estrogen, such as the activation of nitric oxide synthesis or the remodeling of actin cytoskeleton. In addition, these pathways are critical for the regulation of the expression of a number of target proteins implicated in cell proliferation, apoptosis, differentiation, movement, and homeostasis. In this manner, the extra-nuclear pathways are tightly integrated with the genomic pathways to orchestrate the full spectrum of estrogen's biological functions. The recent advancements in the characterization of the molecular basis of the extra-nuclear signaling of estrogen helps to understand the role of estrogen on human cells, and may in future turn out to be of relevance for clinical purposes.
Collapse
Affiliation(s)
- Xiao-Dong Fu
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
| | | |
Collapse
|
17
|
Genomic and nongenomic effects of glucocorticoids. ACTA ACUST UNITED AC 2008; 4:525-33. [PMID: 18762788 DOI: 10.1038/ncprheum0898] [Citation(s) in RCA: 389] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Accepted: 07/18/2008] [Indexed: 12/17/2022]
Abstract
The strong anti-inflammatory and immunosuppressive effects of glucocorticoids are mediated primarily by the cytosolic glucocorticoid receptors. These receptors are members of the steroid hormone receptor family, a superfamily of ligand-inducible transcription factors, and exert genomic effects that can result in increased expression of regulatory-including anti-inflammatory-proteins (transactivation), or decreased production of proinflammatory proteins (transrepression). Transactivation is thought to be responsible for numerous adverse effects of glucocorticoids; transrepression is thought to be responsible for many of the clinically desirable anti-inflammatory and immunosuppressive effects of glucocorticoids. Optimized glucocorticoids, such as selective glucocorticoid receptor agonists, are being developed to try to minimize the adverse effects many patients experience. Glucocorticoids also exert their effects via rapid, nongenomic mechanisms that can be classified as involving nonspecific interactions of glucocorticoids with cellular membranes, nongenomic effects that are mediated by cytosolic glucocorticoid receptors, or specific interactions with membrane-bound glucocorticoid receptors. Increased understanding of these mechanisms of glucocorticoid action could enable the development of novel drugs with which to treat patients with inflammatory and autoimmune disease.
Collapse
|
18
|
Vasudevan N, Pfaff DW. Non-genomic actions of estrogens and their interaction with genomic actions in the brain. Front Neuroendocrinol 2008; 29:238-57. [PMID: 18083219 DOI: 10.1016/j.yfrne.2007.08.003] [Citation(s) in RCA: 262] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Accepted: 08/14/2007] [Indexed: 12/16/2022]
Abstract
Ligands for the nuclear receptor superfamily have at least two mechanisms of action: (a) classical transcriptional regulation of target genes (genomic mechanisms); and (b) non-genomic actions, which are initiated at the cell membrane, which could also impact transcription. Though transcriptional mechanisms are increasingly well understood, membrane-initiated actions of these ligands are incompletely understood. This has led to considerable debate over the physiological relevance of membrane-initiated actions of hormones versus genomic actions of hormones, with genomic actions predominating in the endocrine field. There is good evidence that the membrane-limited actions of hormones, particularly estrogens, involve the rapid activation of kinases and the release of calcium and that these are linked to physiologically relevant scenarios in the brain. We show evidence in this review, that membrane actions of estrogens, which activate these rapid signaling cascades, can also potentiate nuclear transcription in both the central nervous system and in non-neuronal cell lines. We present a theoretical scenario which can be used to understand this phenomenon. These signaling cascades may occur in parallel or in series but subsequently, converge at the modification of transcriptionally relevant molecules such as nuclear receptors and/or coactivators. In addition, other non-cognate hormones or neurotransmitters may also activate cascades to crosstalk with estrogen receptor-mediated transcription, though the relevance of this is less clear. The idea that coupling between membrane-initiated and genomic actions of hormones is a novel idea in neuroendocrinology and provides us with a unified view of hormone action in the central nervous system.
Collapse
Affiliation(s)
- Nandini Vasudevan
- Cell and Molecular Biology Department, Tulane University, LA 70118, USA.
| | | |
Collapse
|
19
|
Zhang Z, Duan L, Du X, Ma H, Park I, Lee C, Zhang J, Shi J. The proliferative effect of estradiol on human prostate stromal cells is mediated through activation of ERK. Prostate 2008; 68:508-16. [PMID: 18213633 DOI: 10.1002/pros.20722] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Estrogen is involved in the development and progression of benign prostatic hyperplasia (BPH). It can stimulate proliferation of prostate stromal cells (PrSCs). However, the exact mechanism remains unclear. METHODS We used the primary cultured human PrSCs and a prostate stromal cell line, WPMY-1, to examine the signaling pathways involved in estrogen-mediated proliferation of PrSCs. Cells were treated with 17beta-estradiol (E(2)) or BSA-E(2). Cell proliferation was assessed by the MTT assay and by cell counting. Western blot analysis was used to determine the status of activation of ERK1/2. RESULTS Results indicated that both E(2) and BSA-E(2) stimulated proliferation of primary PrSCs and WPMY-1 cells. ERK was rapidly activated by E(2) and BSA-E(2). PD98059, which is a selective ERK inhibitor, significantly inhibited estrogen-induced cell proliferation. PrSCs expressed estrogen receptor alpha (ERalpha) and GPR30 but not ERbeta. Small hairpin RNA (shRNA) to ERalpha, but not to GPR30, blocked estrogen-mediated ERK activation and cell proliferation. CONCLUSIONS The results indicated that estrogen could activate ERK pathway through the non-genomic ERalpha pathway, leading to proliferation of PrSCs.
Collapse
Affiliation(s)
- Zhisong Zhang
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin, China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Wang C, Prossnitz ER, Roy SK. Expression of G protein-coupled receptor 30 in the hamster ovary: differential regulation by gonadotropins and steroid hormones. Endocrinology 2007; 148:4853-64. [PMID: 17640985 DOI: 10.1210/en.2007-0727] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The nongenomic actions of estradiol-17beta are mediated by transmembrane estrogen receptors. Recently, G protein-coupled receptor 30 (GPR30) has been suggested to be a transmembrane estrogen receptor that can mediate rapid and transcription-independent estradiol-17beta signaling in different cell types. However, the expression, regulation, or biological relevance of GPR30 in the ovary remains unknown. We examined the expression and hormonal regulation of GPR30 mRNA and protein in hamster ovarian cells during the estrous cycle and after hypophysectomy and hormone replacement. GPR30 protein expression was high in the theca, appreciable in the granulosa, but low in luteal cells. GPR30 protein levels in granulosa and theca cells increased steadily with the development of preantral and antral follicles, respectively. GPR30 mRNA and protein levels increased significantly on diestrous (d 3 of the estrous cycle), but decreased on d 4 at 1600 h after the LH surge. GPR30 mRNA levels increased significantly after hypophysectomy. Although steroid treatment failed to alter ovarian GPR30 mRNA levels, either FSH or LH effectively reduced the levels. Interestingly, the decrease in GPR30 mRNA corresponded to a marked increase in the receptor protein levels. FSH treatment, either alone or together with LH, resulted in a marked increase in GPR30 immunostaining in granulosa cells. LH alone significantly increased immunostaining in theca cells. These results suggest that GPR30 is expressed in the membrane of hamster granulosa and theca cells, and the expression is regulated by gonadotropins. The unique pattern of GPR30 expression suggests that gonadotropin-regulated follicular cell functions may involve GPR30 activity.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 984515 Nebraska Medical Center, Omaha, NE 68198-4515, USA
| | | | | |
Collapse
|
21
|
Belcher SM. Rapid signaling mechanisms of estrogens in the developing cerebellum. ACTA ACUST UNITED AC 2007; 57:481-92. [PMID: 17931703 DOI: 10.1016/j.brainresrev.2007.07.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Revised: 07/03/2007] [Accepted: 07/18/2007] [Indexed: 11/25/2022]
Abstract
The steroid hormone 17beta-estradiol regulates the normal function and development of the mammalian nervous system. Many of estradiol's effects are mediated via the nuclear hormone estrogen receptors ERalpha and ERbeta. In addition to regulating estrogen-responsive gene expression, estradiol also acts in an immediate and cell-specific fashion to regulate various intracellular signal transduction pathways. The goal of this review is to develop a contextual framework to understand the generalized function of estrogen during development of brain regions not known to be sexually specialized. However, it is first important to build this framework on the more well-developed foundation of estrogen's gonad-driven sex-specific actions. As a result, a discussion of known and proposed mechanisms of estrogen actions in reproductive and other tissues will be presented. Building upon this information, a review of our research group's recent in vitro and in vivo studies that have focused on elucidating the mechanisms of estrogen actions in neurons of the non-sexually specialized cerebellum will be presented. While the full spectrum of estrogen action during normal cerebellar development remains unresolved, results of recent studies have revealed a pathologic role for estrogen and estrogen receptors in medulloblastoma, common pediatric brain tumors that arise from cerebellar granule cell-like precursors. The potential use of anti-estrogen signaling agents as adjuvant therapy for medulloblastoma is proposed based on those finding.
Collapse
Affiliation(s)
- Scott M Belcher
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, 231 Albert Sabin Way, PO Box 670575, Cincinnati, OH 45267-0575, USA.
| |
Collapse
|
22
|
Marin R, Ramírez CM, González M, González-Muñoz E, Zorzano A, Camps M, Alonso R, Díaz M. Voltage-dependent anion channel (VDAC) participates in amyloid beta-induced toxicity and interacts with plasma membrane estrogen receptor alpha in septal and hippocampal neurons. Mol Membr Biol 2007; 24:148-60. [PMID: 17453421 DOI: 10.1080/09687860601055559] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Voltage-dependent anion channel (VDAC) is a porin known by its role in metabolite transport across mitochondria and participation in apoptotic processes. Although traditionally accepted to be located within mitochondrial outer membrane, some data has also reported its presence at the plasma membrane level where it seems to participate in regulation of normal redox homeostasis and apoptosis. Here, exposure of septal SN56 and hippocampal HT22 cells to specific anti-VDAC antibodies prior to amyloid beta (Abeta) peptide was observed to prevent neurotoxicity. In these cell lines, we identified a VDAC form associated with the plasma membrane that seems to be particularly abundant in caveolae. The two membrane-related isoforms of estrogen receptor alpha (mERalpha) (80 and 67 kDa), known in SN56 cells to participate in estrogen-induced neuroprotection against Abeta injury, were also observed to be present in caveolae. Interestingly, we demonstrated for the first time that both VDAC and mERalpha interact at the plasma membrane of these neurons as well as in microsomal fractions of the corresponding murine septal and hippocampal tissues. These proteins were also shown to associate with caveolin-1, thereby corroborating their presence in caveolar microdomains. Taken together, these results suggest that VDAC-mERalpha association at the plasma membrane level may participate in the modulation of Abeta-induced cell death.
Collapse
Affiliation(s)
- Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Physiology & Institute of Biomedical Technologies, University of La Laguna, School of Medicine, Santa Cruz de Tenerife, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Hormonal ligands for the nuclear receptor superfamily have at least two interacting mechanisms of action: 1) classical transcriptional regulation of target genes (genomic mechanisms); and 2) nongenomic actions that are initiated at the cell membrane, which could impact transcription. Although transcriptional mechanisms are increasingly well understood, membrane-initiated actions of these ligands are incompletely understood. Historically, this has led to a considerable divergence of thought in the molecular endocrine field. We have attempted to uncover principles of hormone action that are relevant to membrane-initiated actions of estrogens. There is evidence that the membrane-limited actions of hormones, particularly estrogens, involve the rapid activation of kinases and the release of calcium. Membrane actions of estrogens, which activate these rapid signaling cascades, can also potentiate nuclear transcription. These signaling cascades may occur in parallel or in series but subsequently converge at the level of modification of transcriptionally relevant molecules such as nuclear receptors and/or coactivators. In addition, other hormones or neurotransmitters may also activate cascades to crosstalk with estrogen receptor-mediated transcription. The idea of synergistic coupling between membrane-initiated and genomic actions of hormones fundamentally revises the paradigms of cell signaling in neuroendocrinology.
Collapse
Affiliation(s)
- Nandini Vasudevan
- Department of Biology, Pennsylvania State University, University Park, Pennsylvania 16802, USA.
| | | |
Collapse
|
24
|
Abstract
Contrary to the view that embryos and larvae are the most fragile stages of life, development is stable under real-world conditions. Early cleavage embryos are prepared for environmental vagaries by having high levels of cellular defenses already present in the egg before fertilization. Later in development, adaptive responses to the environment either buffer stress or produce alternative developmental phenotypes. These buffers, defenses, and alternative pathways set physiological limits for development under expected conditions; teratology occurs when embryos encounter unexpected environmental changes and when stress exceeds these limits. Of concern is that rapid anthropogenic changes to the environment are beyond the range of these protective mechanisms.
Collapse
Affiliation(s)
- Amro Hamdoun
- Hopkins Marine Station of Stanford University, Pacific Grove, CA 93950
- *To whom correspondence may be addressed at:
Hopkins Marine Station 120 Oceanview Boulevard, Pacific Grove, CA 93950. E-mail:
or
| | - David Epel
- Hopkins Marine Station of Stanford University, Pacific Grove, CA 93950
- *To whom correspondence may be addressed at:
Hopkins Marine Station 120 Oceanview Boulevard, Pacific Grove, CA 93950. E-mail:
or
| |
Collapse
|
25
|
Guo SW, Olive DL. Two Unsuccessful Clinical Trials on Endometriosis and a Few Lessons Learned. Gynecol Obstet Invest 2007; 64:24-35. [PMID: 17202821 DOI: 10.1159/000098413] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In 1999, a phase II clinical trial on the use of fulvestrant to treat endometriosis was launched; yet after 7 years there is still no report on its outcome. In 2005, another trial on the use of raloxifene to treat endometriosis was terminated early due to unfavorable outcome. The two apparently unsuccessful clinical trials on endometriosis have taught us a few important lessons. First, we need to understand endometriosis through more basic research. We have also been reminded that human endometriosis trials differ from animal studies; anatomy and physiology are often divergent, and outcome measures are certainly different. Ectopic endometrium can differ significantly from eutopic tissue, and this issue needs to be more thoroughly explored. We believe human cell lines will prove to be an inexpensive and valuable tool for future preliminary evaluation of medical therapies as well as discerning pathophysiologic processes of the disease. Based on our current understanding of endometriosis, some concrete benchmarks can be established for testing or screening potential compounds in vitro. Finally, estrogen receptor modulators are often tissue-, cell-, and context-specific in their actions; they should not be simplistically grouped together nor should extrapolations from one compound to another be undertaken in a cavalier manner.
Collapse
Affiliation(s)
- Sun-Wei Guo
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226-0509, USA.
| | | |
Collapse
|
26
|
Abstract
In humans, structural and functional changes attributable to aging are more visibly evident in the skin than in any other organ. Estrogens have significant effects on skin physiology and modulate epidermal keratinocytes, dermal fibroblasts and melanocytes, in addition to skin appendages including the hair follicle and the sebaceous gland. Importantly, skin aging can be significantly delayed by the administration of estrogen. This paper reviews the effects of estrogens on skin and the mechanisms by which estrogens can alleviate the changes due to aging that occur in human skin. The relevance of estrogen replacement therapy (HRT) in postmenopausal women and the potential value of selective estrogen receptor modulators (SERMs) as a therapy for diminishing skin aging are also highlighted.
Collapse
Affiliation(s)
| | - Julie Thornton
- Cutaneous Research, Medical Biosciences, School of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
27
|
Ropero AB, Alonso-Magdalena P, Ripoll C, Fuentes E, Nadal A. Rapid endocrine disruption: environmental estrogen actions triggered outside the nucleus. J Steroid Biochem Mol Biol 2006; 102:163-9. [PMID: 17084624 DOI: 10.1016/j.jsbmb.2006.09.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An exogenous substance is defined as an endocrine disrupter chemical (EDC) if it alters the function of the endocrine system provoking adverse health effects. Environmental estrogens are the most studied EDCs. They follow the same mechanisms of action as the gonadal hormone 17beta-estradiol. Up to now, the estrogenicity of environmental estrogenic pollutants has been based on the property of these compounds to bind to estrogen receptors (ERs), either ERalpha or ERbeta, and to act subsequently as transcription factors when binding to the estrogen response element (ERE) in the DNA. All the estrogenic bioassays currently used are based on this mechanism of action. New evidence indicates that the definition of estrogenicity for a chemical should take into account other estrogen receptors as well as new signaling pathways. These include the activation of additional transcription factors as well as the action of xenoestrogens through estrogen receptors located outside the nucleus: in the plasma membrane, mitochondria and probably the cytosol. Therefore, new estrogenic bioassays should be developed to include the novel concept of rapid endocrine disruption.
Collapse
Affiliation(s)
- Ana B Ropero
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain
| | | | | | | | | |
Collapse
|
28
|
Philp KL, Hussain M, Byrne NF, Diver MJ, Hart G, Coker SJ. Greater antiarrhythmic activity of acute 17beta-estradiol in female than male anaesthetized rats: correlation with Ca2+ channel blockade. Br J Pharmacol 2006; 149:233-42. [PMID: 16940993 PMCID: PMC2014275 DOI: 10.1038/sj.bjp.0706850] [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] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Female sex hormones may protect pre-menopausal women from sudden cardiac death. We therefore investigated the effects of the main female sex hormone, 17beta-estradiol, on ischaemia-induced cardiac arrhythmias and on the L-type Ca2+ current (ICaL). EXPERIMENTAL APPROACH In vivo experiments were performed in pentobarbital-anaesthetized rats subjected to acute coronary artery occlusion. ICaL was measured by the whole-cell patch-clamp technique, in rat isolated ventricular myocytes. KEY RESULTS Acute intravenous administration of 17beta-estradiol as a bolus dose followed by a continuous infusion, commencing 10 min before coronary artery occlusion, had dose-dependent antiarrhythmic activity. In female rats 300 ng kg(-1) + 30 ng kg(-1) min(-1) 17beta-estradiol significantly reduced the number of ventricular premature beats (VPBs) and the incidence of ventricular fibrillation (VF). A ten fold higher dose of 17beta-estradiol was required to cause similar effects in male rats. In vitro 17beta-estradiol reduced peak ICaL in a concentration-dependent manner. The EC50 was ten-fold higher in male myocytes (0.66 microM) than in females (0.06 microM). CONCLUSIONS AND IMPLICATIONS These results indicate that 17beta-estradiol has marked dose-dependent antiarrhythmic activity that is greater in female rats than in males. A similar differential potency in blocking ICaL in myocytes from female and male rats can account for this effect. This provides an explanation for the antiarrhythmic activity of 17beta-estradiol and gender-selective protection against sudden cardiac death.
Collapse
Affiliation(s)
- K L Philp
- Department of Pharmacology and TherapeuticsUK
| | - M Hussain
- Division of Clinical Science, The University of LiverpoolUK
| | - N F Byrne
- Department of Pharmacology and TherapeuticsUK
| | - M J Diver
- Division of Clinical Chemistry, The University of Liverpool Liverpool, UK
| | - G Hart
- Division of Clinical Science, The University of LiverpoolUK
| | - S J Coker
- Department of Pharmacology and TherapeuticsUK
- Author for correspondence:
| |
Collapse
|
29
|
Dubey S, Siegfried JM, Traynor AM. Non-small-cell lung cancer and breast carcinoma: chemotherapy and beyond. Lancet Oncol 2006; 7:416-24. [PMID: 16648046 DOI: 10.1016/s1470-2045(06)70693-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Early screening, adjuvant and sequential systemic treatment, and hormonal therapy have benefits in treatment of breast cancer. Management of non-small-cell lung cancer (NSCLC) is progressing and will hopefully follow in the same footsteps as that of breast cancer. Only recently have clinical trials established adjuvant treatment as the standard of care in lung cancer. A growing number of effective cytotoxic and targeted agents have resulted in increased survival when used as sequential treatment in both breast cancer and NSCLC. The interaction between oestrogen receptors (ER) in the lung and epidermal growth factor receptor (EGFR) suggests a potential role for endocrine manipulation in the treatment of NSCLC. This complex interaction involves several types of ER receptors and different signalling pathways. Interactions between tobacco and oestrogen confound the effects of exogenous oestrogens on risk of lung cancer, but not on that of breast cancer. The optimum application of hormonal manipulation to prevent or treat lung cancer will depend on a more-complete understanding of lung-specific ER signalling. Early trials have assessed the interaction between the ER and EGFR signalling.
Collapse
Affiliation(s)
- Sarita Dubey
- Division of Hematology/Oncology, University of California, San Francisco, CA 94115, USA.
| | | | | |
Collapse
|
30
|
|
31
|
Sarkar DK. Genesis of prolactinomas: studies using estrogen-treated animals. FRONTIERS OF HORMONE RESEARCH 2006; 35:32-49. [PMID: 16809921 PMCID: PMC2882189 DOI: 10.1159/000094307] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Prolactin-secreting adenomas (prolactinomas) are the most prevalent form of pituitary tumors in humans. Our knowledge of the formation of these tumors is limited. Experimental work in animal has uncovered that estradiol exposure leads to prolactinoma formation via orchestrated events involving dopamine D2 receptors, transforming growth factor-beta(TGF-beta) isoforms and their receptors, as well as factors secondary to TGF-beta action. Additionally, these studies determined that TGF-beta and b-FGF interact to facilitate the communication between lactotropes and folliculo-stellate cells that is necessary for the mitogenic action of estradiol. The downstream signaling that governs lactotropic cell proliferation involves activation of the MAP kinase p44/42-dependent pathway.
Collapse
Affiliation(s)
- Dipak K Sarkar
- Endocrine Program, Biomedical Division of the Center of Alcohol Studies and Department of Animal Sciences, Rutgers, State University of New Jersey, New Brunswick, N.J., USA
| |
Collapse
|
32
|
Mizota K, Ueda H. Endocrine Disrupting Chemical Atrazine Causes Degranulation through Gq/11 Protein-Coupled Neurosteroid Receptor in Mast Cells. Toxicol Sci 2005; 90:362-8. [PMID: 16381660 DOI: 10.1093/toxsci/kfj087] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We studied the effects of representative endocrine-disrupting chemicals on beta-hexosaminidase release from mast cells and their putative neurosteroid receptor involvement. Some endocrine-disrupting chemicals, such as amitrol, benzophenon, bisphenol A, pentachlorophenol, and tetrabromophenol A did not cause hexosaminidase release from RBL-2H3 cells, but they blocked the release by dehydroepiandrosterone sulfate, a representative neurosteroid agonist. On the contrary, atrazine, which is a widely used herbicide, caused a rapid and concentration-dependent degranulation in the range between 10 nM and 1 microM in RBL-2H3 and peritoneal mast cells. Atrazine-induced degranulation was also evaluated by Alexa 488-annexin V binding to the phosphatidylserine, which is externalized during degranulation, and these actions were blocked by BSA-conjugated (membrane-impermeable) progesterone (PROG-BSA). The atrazine-induced beta-hexosaminidase release was characterized by various inhibitors including antisense-oligodeoxynucleotide for Galpha(q/11), pertussis toxin, phospholipase C inhibitor U-73122, inositol 1,4,5-triphosphate receptor inhibitor xestospongin C and Ca(2+) channel blocker lanthanum chloride. These analyses revealed that the degranulation is mediated by putative metabotropic neurosteroid receptor, G(q/11), phospholipase C and Ca(2+) mobilization from intracellular stores. Having documented progesterone receptor-modulation of atrazine-induced mast cell degranulation in vitro, this response was evaluated in mice. Atrazine caused pain responses when injected in the foot pads of mice, and they were antagonized by local administration of PROG-BSA or diphenhydramine. Atrazine also caused PROG-BSA-reversible plasma extravasation. All these findings strongly suggest that herbicide atrazine exerts inflammatory activity through activation of putative G(q/11)-coupled neurosteroid receptor and phospholipase C.
Collapse
Affiliation(s)
- Kaori Mizota
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | |
Collapse
|
33
|
Belcher SM, Le HH, Spurling L, Wong JK. Rapid estrogenic regulation of extracellular signal- regulated kinase 1/2 signaling in cerebellar granule cells involves a G protein- and protein kinase A-dependent mechanism and intracellular activation of protein phosphatase 2A. Endocrinology 2005; 146:5397-406. [PMID: 16123167 DOI: 10.1210/en.2005-0564] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In neonatal rat cerebellar neurons, 17beta-estradiol (E(2)) rapidly stimulates ERK1/2 phosphorylation through a membrane-associated receptor. Here the mechanism of rapid E(2)-induced ERK1/2 signaling in primary cultured granule cells was investigated in more detail. The results of these studies show that E(2) and ICI182,780, a steroidal antagonist of estrogen receptor transactivation, rapidly increased ERK signaling with a time course similar to the transient activation induced by epidermal growth factor (EGF). However, EGF receptor (EGFR) autophosphorylation was not increased by E(2), and blockade of EGFR tyrosine kinase activity did not abrogate the rapid actions of E(2). The involvement of Src-tyrosine kinase activity was demonstrated by detection of increased c-Src phosphorylation in response to E(2) and by blockade of E(2)-induced ERK1/2 activation by inhibition of Src-family tyrosine kinase activity. Inhibition of Galphai signaling or protein kinase A (PKA) activity blocked the ability of ICI182,780 to rapidly stimulate ERK signaling. Under those conditions, E(2) treatment induced a rapid and transient suppression of basal ERK1/2 phosphorylation. Protein phosphatase 2A (PP2A) activity was rapidly increased by E(2) but not by E(2) covalently linked to BSA. Rapid E(2)-induced increases in PP2A activity were insensitive to pertussis toxin. The presented evidence indicates that the rapid effects of estrogens on ERK signaling in cerebellar granule cells are induced through a novel G protein-coupled receptor mechanism that requires PKA and Src-kinase activity to link E(2) to the ERK/MAPK signaling module. Along with stimulating ERK signaling, E(2) rapidly activates PP2A via an independent signaling mechanism that may serve as a cell-specific regulator of signal duration.
Collapse
Affiliation(s)
- Scott M Belcher
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Ohio 45267-0575, USA.
| | | | | | | |
Collapse
|
34
|
Viso-León MC, Ripoll C, Nadal A. Oestradiol rapidly inhibits Ca2+ signals in ciliary neurons through classical oestrogen receptors in cytoplasm. Pflugers Arch 2005; 449:33-41. [PMID: 15258764 DOI: 10.1007/s00424-004-1308-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Oestrogen plays a key role in a great variety of actions in the nervous system, either through classical or alternative pathways. The classical pathways are initiated after oestrogen binding to the oestrogen receptors ERalpha or ERbeta, which translocate from the cytoplasm to the nucleus and act there as transcription factors. Alternative pathways are initiated at the plasma membrane and cytoplasm, via binding to classical or non-classical ERs. Using isolated ciliary ganglion neurons from the chick embryo and Ca2+ imaging, we demonstrated that a 10-min exposure to 17beta-oestradiol reduces Ca2+ influx through the plasma membrane. This effect was not reproduced by oestradiol conjugated to bovine serum albumin, which does not cross the plasma membrane, indicating that 17beta-oestradiol was acting intracellularly. ERalpha was detected in the cytoplasm by immunostaining and its involvement in the regulation of Ca2+ influx by ICI182,780 inhibition. The phosphatidylinositol-3 kinase (Pi3-kinase) inhibitor wortmannin and the nitric oxide synthase (NOS) inhibitor Nomega-nitro-L-arginine methyl ester (L-NAME) both blocked the oestradiol effect. The oestradiol effect was reproduced by 8Br-cGMP and abolished in the presence of the cGMP-dependent protein kinase (PKG) inhibitor KT5823. Our study indicates that 17beta-oestradiol can regulate Ca2+ influx via PI3-kinase, NOS and PKG after activation of cytoplasmic ER.
Collapse
Affiliation(s)
- M Carmen Viso-León
- Institut de Bioenginyeria, Universitat Miguel Hernández d'Elx, Campus de Sant Joan, Carretera Alacant-Valéncia Km 87, 03550 Sant Joan d'Alacant, Spain
| | | | | |
Collapse
|
35
|
Abstract
The female sex steroid hormones 17beta-estradiol and progesterone mediate their biological effects on development, differentiation, and maintenance of reproductive tract and other target tissues through gene regulation by nuclear steroid receptors that function as ligand-dependent transcription factors. However, not all effects of 17beta-estradiol and progesterone are mediated by direct control of gene expression. These hormones also have rapid stimulatory effects on the activities of a variety of signal transduction molecules and pathways and, in many cases, these effects appear to be initiated from the plasma cell membrane. There is growing evidence that a subpopulation of the conventional nuclear steroid receptor localized at the cell membrane mediates many of the rapid signaling actions of steroid hormones; however, novel membrane receptors unrelated to conventional steroid receptors have also been implicated. This chapter reviews the nature of the receptors that mediate rapid signaling actions of estrogen and progesterone and describes the signaling molecules and pathways involved, the mechanisms by which receptors couple with components of signaling complexes and trigger responses, and the target tissues and cell functions regulated by this mode of steroid hormone action.
Collapse
Affiliation(s)
- Dean P Edwards
- University of Colorado Health Sciences Center, Department of Pathology and Program in Molecular Biology, Aurora, Colorado 80045, USA.
| |
Collapse
|
36
|
Thomas P, Pang Y, Filardo EJ, Dong J. Identity of an estrogen membrane receptor coupled to a G protein in human breast cancer cells. Endocrinology 2005; 146:624-32. [PMID: 15539556 DOI: 10.1210/en.2004-1064] [Citation(s) in RCA: 1076] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Although nonclassical estrogen actions initiated at the cell surface have been described in many tissues, the identities of the membrane estrogen receptors (mERs) mediating these actions remain unclear. Here we show that GPR30, an orphan receptor unrelated to nuclear estrogen receptors, has all the binding and signaling characteristics of a mER. A high-affinity (dissociation constant 2.7 nm), limited capacity, displaceable, single binding site specific for estrogens was detected in plasma membranes of SKBR3 breast cancer cells that express GPR30 but lack nuclear estrogen receptors. Progesterone-induced increases and small interfering RNA-induced decreases in GPR30 expression in SKBR3 cells were accompanied by parallel changes in specific estradiol-17beta (E2) binding. Plasma membranes of human embryonic kidney 293 cells transfected with GPR30, but not those of untransfected cells, and human placental tissues that express GPR30 also displayed high-affinity, specific estrogen binding typical of mERs. E2 treatment of transfected cell membranes caused activation of a stimulatory G protein that is directly coupled to the receptor, indicating GPR30 is a G protein-coupled receptor (GPCR), and also increased adenylyl cyclase activity. The finding that the antiestrogens tamoxifen and ICI 182,780, and an environmental estrogen, ortho,para-dichlorodiphenyldichloroethylene (o,p'-DDE), have high binding affinities to the receptor and mimic the actions of E2 has important implications for both the development and treatment of estrogen-dependent breast cancer. GPR30 is structurally unrelated to the recently discovered family of GPCR-like membrane progestin receptors. The identification of a second distinct class of GPCR-like steroid membrane receptors suggests a widespread role for GPCRs in nonclassical steroid hormone actions.
Collapse
Affiliation(s)
- P Thomas
- University of Texas Marine Science Institute, 750 Channel View Drive, Port Aransas, Texas 78373, USA.
| | | | | | | |
Collapse
|
37
|
Zhao X, MacBride MM, Peterson BR, Pfaff DW, Vasudevan N. Calcium flux in neuroblastoma cells is a coupling mechanism between non-genomic and genomic modes of estrogens. Neuroendocrinology 2005; 81:174-82. [PMID: 16020926 DOI: 10.1159/000087000] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2004] [Accepted: 03/19/2005] [Indexed: 11/19/2022]
Abstract
Estrogens have been demonstrated to rapidly modulate calcium levels in a variety of cell types. However, the significance of estrogen-mediated calcium flux in neuronal cells is largely unknown. The relative importance of intra- and extracellular sources of calcium in estrogenic effects on neurons is also not well understood. Previously, we have demonstrated that membrane-limited estrogens, such as E-BSA given before an administration of a 2-hour pulse of 17beta-estradiol (E2), can potentiate the transcription mediated by E2 from a consensus estrogen response element (ERE)-driven reporter gene. Inhibitors to signal transduction cascades given along with E-BSA or E2 demonstrated that calcium flux is important for E-BSA-mediated potentiation of transcription in a transiently transfected neuroblastoma cell line. In this report, we have used inhibitors to different voltage-gated calcium channels (VGCCs) and to intracellular store receptors along with E-BSA in the first pulse or with E2 in the second pulse to investigate the relative importance of these channels to estrogen-mediated transcription. Neither L- nor P-type VGCCs seem to play a role in estrogen action in these cells; while N-type VGCCs are important in both the non-genomic and genomic modes of estrogen action. Specific inhibitors also showed that the ryanodine receptor and the inositol trisphosphate receptor are important to E-BSA-mediated transcriptional potentiation. This report provides evidence that while intracellular stores of calcium are required to couple non-genomic actions of estrogen initiated at the membrane to transcription in the nucleus, extracellular sources of calcium are also important in both non-genomic and genomic actions of estrogens.
Collapse
Affiliation(s)
- Xing Zhao
- Department of Biology, Pennsylvania State University, University Park, PA 16802 , USA
| | | | | | | | | |
Collapse
|
38
|
Buttgereit F, Straub RH, Wehling M, Burmester GR. Glucocorticoids in the treatment of rheumatic diseases: an update on the mechanisms of action. ACTA ACUST UNITED AC 2004; 50:3408-17. [PMID: 15529366 DOI: 10.1002/art.20583] [Citation(s) in RCA: 216] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Schumannstrasse 20/21, 10117 Berlin, Germany.
| | | | | | | |
Collapse
|
39
|
Nadal A, Alonso-Magdalena P, Ripoll C, Fuentes E. Disentangling the molecular mechanisms of action of endogenous and environmental estrogens. Pflugers Arch 2004; 449:335-43. [PMID: 15517344 DOI: 10.1007/s00424-004-1343-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Accepted: 08/30/2004] [Indexed: 01/07/2023]
Abstract
The gonadal hormone 17beta-estradiol is involved in numerous cellular processes. In many cases, 17beta-estradiol actions are imitated by synthetic and natural chemicals in the environment. Their actions differ depending on the target tissue, the receptors involved and the molecular pathways activated. The plethora of estrogenic actions is triggered by different receptors and other specific structures that activate different signalling pathways. This amount of information may lead to a maze of effects triggered by endogenous and environmental estrogens that we intend to clarify in this review. Understanding the variety of estrogen receptors, their different locations and the signalling pathways activated by estrogenic ligands is fundamental for understanding the diversity of actions that estrogens have in different tissues and cells.
Collapse
Affiliation(s)
- Angel Nadal
- Institute of Bioengineering, School of Medicine, Miguel Hernández University, Sant Joan Campus, Carretera Alicante-Valencia Km 87, 03550 Alicante, Spain.
| | | | | | | |
Collapse
|
40
|
Bartholome B, Spies CM, Gaber T, Schuchmann S, Berki T, Kunkel D, Bienert M, Radbruch A, Burmester GR, Lauster R, Scheffold A, Buttgereit F. Membrane glucocorticoid receptors (mGCR) are expressed in normal human peripheral blood mononuclear cells and up-regulated after in vitro stimulation and in patients with rheumatoid arthritis. FASEB J 2004; 18:70-80. [PMID: 14718388 DOI: 10.1096/fj.03-0328com] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Glucocorticoids mediate their therapeutic actions mostly by genomic effects via cytosolic receptors, but some effects are too rapid to be mediated by changes at the genomic level. The detailed mechanisms of these nongenomic actions are still unclear. Membrane-bound glucocorticoid receptors (mGCR) have been suggested to be involved, although their physiological existence in humans so far is hypothetical. For the first time we demonstrate the existence of mGCR on monocytes and B cells obtained from healthy blood donors using high-sensitivity immunofluorescent staining. Immunostimulation with lipopolysaccharide increases the percentage of mGCR-positive monocytes, which can be prevented by inhibiting the secretory pathway. Overexpression of the human glucocorticoid receptor alpha alone is not sufficient to enhance mGCR expression. These in vitro findings are consistent with our clinical observation that in patients with rheumatoid arthritis the frequency of mGCR positive monocytes is increased and positively correlated with disease activity. We conclude that mGCR are 1) indeed physiologically present in healthy blood donors, but remained unidentified by conventional techniques due to their small number per cell and 2) actively up-regulated and transported through the cell after immunostimulation. These receptors may reflect a feedback mechanism of the organism upon immunostimulation and/or play a role in pathogenesis.
Collapse
Affiliation(s)
- Burkhard Bartholome
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chaturvedi K, Sarkar DK. Involvement of protein kinase C-dependent mitogen-activated protein kinase p44/42 signaling pathway for cross-talk between estradiol and transforming growth factor-beta3 in increasing basic fibroblast growth factor in folliculostellate cells. Endocrinology 2004; 145:706-15. [PMID: 14605008 PMCID: PMC2895416 DOI: 10.1210/en.2003-1063] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We have recently shown that TGF-beta3, in the presence of estradiol, increases the release of basic fibroblast growth factor (bFGF) from folliculostellate (FS) cells in the pituitary. We determined the interactive effects of TGF-beta3 and estradiol on bFGF production and release from FS cells, and the role of the MAPK pathway in TGF-beta3 and estradiol interaction. We found that TGF-beta3 and estradiol alone moderately increased cell content and release of bFGF from FS cells; but together, they markedly increased the peptide. Estradiol and TGF-beta3 alone moderately activated MAPK p44/42; together they produced marked activation of MAPK p44/42. Pretreatment of FS cells with an MAPK kinase 1/2 inhibitor or with protein kinase C inhibitors suppressed the activation of MAPK p44/42, bFGF release, and protein level increases, all of which were induced by TGF-beta3 and estradiol. Estradiol and TGF-beta3, either alone or in combination, increased the levels of active Ras. Furthermore, bFGF induction by TGF-beta3 and estradiol was blocked by overexpression of Ras N17, a dominant negative mutant of Ras p21. Estrogen receptor blocker ICI 182,780 failed to prevent estrogen's and TGF-beta3's effects on bFGF. These data suggest that an estradiol receptor-independent protein kinase C- activated Ras-dependent MAPK pathway is involved in the cross-talk between TGF-beta3 and estradiol to increase bFGF production and/or release from FS cells.
Collapse
Affiliation(s)
- Kirti Chaturvedi
- Endocrinology Program, Biomedical Division of the Center of Alcohol Studies and Department of Animal Sciences, Rutgers, The State University of New Jersey, 84 Lipman Drive, New Brunswick, NJ 08901, USA
| | | |
Collapse
|
42
|
Soria B, Quesada I, Ropero AB, Pertusa JA, Martín F, Nadal A. Novel players in pancreatic islet signaling: from membrane receptors to nuclear channels. Diabetes 2004; 53 Suppl 1:S86-91. [PMID: 14749271 DOI: 10.2337/diabetes.53.2007.s86] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Glucose and other nutrients regulate many aspects of pancreatic islet physiology. This includes not only insulin release, but also insulin synthesis and storage and other aspects of beta-cell biology, including cell proliferation, apoptosis, differentiation, and gene expression. This implies that in addition to the well-described signals for insulin release, other intracellular signaling mechanisms are needed. Here we describe the role of global and local Ca(2+) signals in insulin release, the regulation of these signals by new membrane receptors, and the generation of nuclear Ca(2+) signals involved in gene expression. An integrated view of these pathways should improve the present description of the beta-cell biology and provide new targets for novel drugs.
Collapse
Affiliation(s)
- Bernat Soria
- Institute of Bioengineering, Miguel Hernandez University, Alicante, Spain.
| | | | | | | | | | | |
Collapse
|
43
|
Chen ZJ, Vetter M, Chang GD, Liu S, Ding Y, Chang CH. Non-genomic effects of tamoxifen on the activation of membrane-bound guanylate cyclase GC-A. J Pharm Pharmacol 2004; 55:1539-45. [PMID: 14713365 DOI: 10.1211/0022357022089] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Oestrogen is known to exert both genomic and non-genomic effects on target tissues. Unlike the genomic effects, the identity of receptors mediating the non-genomic effects of oestrogen remains controversial. 17beta-estradiol has been shown to activate membrane-bound guanylate cyclase GC-A in PC12 cells in a non-genomic manner. To examine whether 17beta-estradiol exerts a similar effect in other cell types, we measured the effect of 17beta-estradiol and tamoxifen, an anti-oestrogen, on guanylate cyclase activity in porcine kidney proximal tubular LLC-PK1 cells. 17beta-estradiol increased cGMP levels in LLC-PK1 cells. Interestingly, addition of tamoxifen also increased cGMP levels in a concentration-dependent manner in LLC-PK1 cells. The effects of both 17beta-estradiol and tamoxifen on guanylate cyclase activity were not additive, suggesting that oestrogen and tamoxifen activate the same enzyme. Similar phenomena were also observed in LLC-PK1 cell membrane preparation. LLC-PK1 cells do not express membrane-bound guanylate cyclase GC-B and express low levels of membrane-bound guanylate cyclase GC-C. Tamoxifen inhibited the activation of GC-A by atrial natriuretic factor (ANF). However, it did not affect membrane-bound guanylate cyclase GC-C stimulated by guanylin or Escherichia coli heat-stable toxin STa. These results indicate that 17beta-estradiol and tamoxifen activate GC-A in LLC-PK1 cells. Thus, tamoxifen functions as an agonist rather than an antagonist for the membrane oestrogen receptor coupled to the activation of GC-A.
Collapse
Affiliation(s)
- Zi-Jiang Chen
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | | | |
Collapse
|
44
|
Booth EA, Marchesi M, Kilbourne EJ, Lucchesi BR. 17Beta-estradiol as a receptor-mediated cardioprotective agent. J Pharmacol Exp Ther 2003; 307:395-401. [PMID: 12893838 DOI: 10.1124/jpet.103.054205] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cardiac tissue that undergoes an ischemic episode exhibits irreversible alterations that become more extensive upon reperfusion. Estrogen treatment has been reported to protect against reperfusion injury, but the mechanism remains unknown. The cardioprotective effects of 17beta-estradiol, a biologically active form of the hormone, and 17alpha-estradiol were assessed in an in vivo occlusion-reperfusion model. Anesthetized, ovariectomized rabbits were administered 17beta-estradiol (20 microg), 17alpha-estradiol (1 mg), or vehicle intravenously 30 min before a 30-min occlusion of the left anterior descending (LAD) coronary artery followed by 4 h of reperfusion. Infarct size as a percentage of area at risk decreased in the 17beta-estradiol-treated group (18.8 +/- 1.7) compared with 17alpha-estradiol (41.9 +/- 4.8; P < 0.01) or vehicle groups (48 +/- 5.5; P < 0.001). Similar results were obtained when infarct size was expressed as a percentage of total left ventricle. The second objective of the study was to assess fulvestrant (Faslodex, ICI 182,780), an estrogen receptor antagonist, for its effects on infarct size in ovariectomized female rabbits treated with 17beta-estradiol. ICI 182,780 was administered intravenously 1 h before the administration of 17beta-estradiol (20 microg) or vehicle. The hearts were subjected to 30-min LAD coronary artery occlusion and 4 h of reperfusion. Pretreatment with ICI 182,780 significantly limited the infarct size sparing effect of 17beta-estradiol when expressed as a percentage of the risk region (53.0 +/- 5.0). The results indicate that 17beta-estradiol protects the heart against ischemia-reperfusion injury and that the observed cardioprotection is mediated by the estrogen receptor.
Collapse
Affiliation(s)
- Erin A Booth
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632, USA
| | | | | | | |
Collapse
|
45
|
Estrogens and ICI182,780 (Faslodex) modulate mitosis and cell death in immature cerebellar neurons via rapid activation of p44/p42 mitogen-activated protein kinase. J Neurosci 2003. [PMID: 12832521 DOI: 10.1523/jneurosci.23-12-04984.2003] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Estrogen influences the development and function of the nervous system through estrogen receptor-dependent changes in gene expression and by rapidly influencing diverse intracellular signaling pathways. We have investigated the influence of estradiol on developing neonatal rat cerebellar neurons in primary culture and found that low concentrations of 17beta-estradiol (17beta-E2), 17alpha-E2, 17beta-E2-BSA, and ICI182,780 stimulated phosphorylation of the extracellular signal-regulated kinases 1/2 (ERK1/2) mitogen-activated protein kinases (MAPK). Neither testosterone nor progesterone increased ERK1/2 phosphorylation. The effects of the estrogens were specific to the ERK1/2 MAPK pathway and were blocked by U0126, an inhibitor of the ERK1/2 MAPK kinase (MEK1/2). Compared with control cultures, significant MAPK-dependent decreases in viable granule cell numbers were observed in dissociated explant cultures of developing cerebellar neurons 24-96 hr after pulse treatment with 10 pm 17beta-E2 or 10 nm ICI182,780. In contrast, continuous exposure to 10 pm 17beta-E2 significantly increased granule cell numbers. Analysis of bromodeoxyuridine incorporation revealed that a 15 min pulsed treatment with 10 pm 17beta-E2 increased mitogenesis, whereas continuous exposure to the same concentration of 17beta-E2 was anti-mitotic. Estradiol did not increase caspase activity; however, significant increases in cellular permeability and lysis were observed. Cell lysis and death were independent of the pan-caspase inhibitor zVAD-fmk but were blocked fully by the irreversible calpain inhibitor PD150606. These results indicate that rapid activation of the ERK1/2 MAPK pathway by low concentrations of 17beta-E2 induces oncotic/necrotic, but not apoptotic, programmed cell death in a subpopulation of developing granule cells and increased mitogenesis of the granule cell neuroblasts refractory to estrogen-induced neurotoxicity.
Collapse
|
46
|
Fiorini S, Ferretti ME, Biondi C, Pavan B, Lunghi L, Paganetto G, Abelli L. 17Beta-eEstradiol stimulates arachidonate release from human amnion-like WISH cells through a rapid mechanism involving a membrane receptor. Endocrinology 2003; 144:3359-67. [PMID: 12865314 DOI: 10.1210/en.2002-221106] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
17beta-Estradiol (17beta-E(2)) greatly and dose-dependently stimulates [(3)H]arachidonic acid (AA) release from the human amnion-like Wistar Institute Susan Hayflick (WISH) cells. This action is abolished by the phospholipase A(2) inhibitor AACOCF(3), significantly reduced by the estrogen receptor (ER) antagonist ICI 182,780, and uninfluenced by cycloheximide. The estradiol-BSA conjugate E(2)coBSA, which binds putative membrane ERs and is unable to enter the cell, also highly stimulates [(3)H]AA release from WISH cells, although to a lesser extent compared with 17beta-E(2). The fluorescent conjugate E(2)coBSA-FITC specifically binds to the surface of a subset of intact WISH cells, and labeling intensity appears dose and time dependent. Cell permeabilization results in a dense intracellular staining, mainly in the peripheral cytoplasm. H-150, an antibody against the N terminus of human ERbeta, also labels the plasma membrane of intact WISH cells and the cytoplasm of permeabilized cells. Almost no labeling is observed using ER-21, an antibody against the N terminus of human ERalpha. RT-PCR evidences the presence of mRNA for ERbeta, not for ERalpha. Our data suggest that 17beta-E(2) stimulates [(3)H]AA release from WISH cells through an apparently nongenomic pathway and interaction with membrane binding sites. These last are, at least in part, similar if not identical to ERbeta.
Collapse
Affiliation(s)
- Sara Fiorini
- Department of Biology, University of Ferrara, 44100-I Ferrara, Italy
| | | | | | | | | | | | | |
Collapse
|
47
|
Marino M, Acconcia F, Trentalance A. Biphasic estradiol-induced AKT phosphorylation is modulated by PTEN via MAP kinase in HepG2 cells. Mol Biol Cell 2003; 14:2583-91. [PMID: 12808053 PMCID: PMC194905 DOI: 10.1091/mbc.e02-09-0621] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We reported previously in HepG2 cells that estradiol induces cell cycle progression throughout the G1-S transition by the parallel stimulation of both PKC-alpha and ERK signaling molecules. The analysis of the cyclin D1 gene expression showed that only the MAP kinase pathway was involved. Here, the presence of rapid/nongenomic, estradiol-regulated, PI3K/AKT signal transduction pathway, its modulation by the levels of the tumor suppressor PTEN, its cross-talk with the ERK pathway, and its involvement in DNA synthesis and cyclin D1 gene promoter activity have all been studied in HepG2 cells. 17beta-Estradiol induced the rapid and biphasic phosphorylation of AKT. These phosphorylations were independent of each other, being the first wave of activation independent of the estrogen receptor (ER), whereas the second was dependent on ER. Both activations were dependent on PI3K activity; furthermore, the ERK pathway modulated AKT phosphorylation by acting on the PTEN levels. The results showed that the PI3K pathway, as well as ER, were strongly involved in both G1-S progression and cyclin D1 promoter activity by acting on its proximal region (-254 base pairs). These data indicate that in HepG2 cells, different rapid/nongenomic estradiol-induced signal transduction pathways modulate the multiple steps of G1-S phase transition.
Collapse
Affiliation(s)
- Maria Marino
- Dipartimento di Biologia, Università Roma Tre, V. le G. Marconi, 446, Italy.
| | | | | |
Collapse
|
48
|
Chesnoy-Marchais D. Potentiation of glycine responses by dideoxyforskolin and tamoxifen in rat spinal neurons. Eur J Neurosci 2003; 17:681-91. [PMID: 12603258 DOI: 10.1046/j.1460-9568.2003.02481.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dideoxyforskolin, a forskolin analogue unable to stimulate adenylate cyclase, and tamoxifen, an antioestrogen widely used against breast cancer, are both known to block some Cl- channels. Their effects on Cl- responses to glycine or GABA have been tested here by using whole-cell recording from cultured spinal neurons. Dideoxyforskolin (4 or 16 microm) and tamoxifen (0.2-5 microm) both potentiate responses to low glycine concentrations. They also induce blocking effects, predominant at high glycine concentrations. At 5 microm, tamoxifen increased responses to 15 microm glycine by a factor >4.5, reaching 20 in some neurons. Potentiation by extracellular dideoxyforskolin or tamoxifen persisted after intracellular application of the modulator and was not due to Zn2+ contamination. Potentiation by tamoxifen also persisted in a Ca2+-free extracellular solution, after intracellular Ca2+ buffering and protein kinase C blockade. Thus, the critical sites of action are not intracellular. The EC50 for glycine was lowered 6.6-fold by 5 microm tamoxifen. The kinetics and voltage-dependence of the effects of tamoxifen on glycine responses support the idea that this hydrophobic drug may act from a site located within the membrane. Tamoxifen (5 micro m) also increased responses to 2 micro m GABA by a factor of 3.5, but barely affected peak responses to 20 microm GABA. The demonstration that tamoxifen affects some of the main inhibitory receptors should be useful for better evaluating its neurological effects. Furthermore, the results identify a new class of molecules that potentiate glycine receptor function.
Collapse
Affiliation(s)
- Dominique Chesnoy-Marchais
- Laboratoire de Neurobiologie Moléculaire et Cellulaire, CNRS UMR-8544, Ecole Normale Supérieure, 46 rue d'Ulm, 75005, Paris, France.
| |
Collapse
|
49
|
Abstract
There is still extensive disparity in our understanding of how estrogens exert their actions, particularly in non-reproductive tissues such as the skin. Although it has been recognized for some time that estrogens have significant effects on many aspects of skin physiology and pathophysiology, studies on estrogen action in skin have been limited. However, estrogens clearly have an important function in many components of human skin including the epidermis, dermis, vasculature, hair follicle and the sebaceous, eccrine and apocrine glands, having significant roles in skin aging, pigmentation, hair growth, sebum production and skin cancer. The recent discovery of a second intracellular estrogen receptor (ERbeta) with different cell-specific roles to the classic estrogen receptor (ERalpha), and the identification of cell surface estrogen receptors, has provided further challenges to understanding the mechanism of estrogen action. It is now time to readdress many of the outstanding questions regarding the role of estrogens in skin and improve our understanding of the physiology and interaction of steroid hormones and their receptors in human skin. Not only will this lead to a better understanding of estrogen action, but may also provide a basis for further interventions in pathological processes that involve dysregulation of estrogen action.
Collapse
Affiliation(s)
- M J Thornton
- Department of Biomedical Sciences, University of Bradford, Bradford, West Yorkshire, UK.
| |
Collapse
|
50
|
Dick GM. The pure anti-oestrogen ICI 182,780 (Faslodex) activates large conductance Ca(2+)-activated K(+) channels in smooth muscle. Br J Pharmacol 2002; 136:961-4. [PMID: 12145095 PMCID: PMC1573435 DOI: 10.1038/sj.bjp.0704807] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Oestrogen and tamoxifen activate large conductance Ca(2+)-activated K(+) (BK(Ca)) channels in smooth muscle through a non-genomic mechanism that depends on the regulatory beta1 subunit and an extracellular binding site. It is unknown whether a "pure" anti-oestrogen such as ICI 182,780 (Faslodex), that has no known oestrogenic properties, would have any effect on BK(Ca) channels. Using single channel patch clamp techniques on canine colonic myocytes, the hypothesis that ICI 182,780 would activate BK(Ca) channels was tested. ICI 182,780 increased the open probability of BK(Ca) channels in inside-out patches with an EC(50) of 1 microM. These data suggest that molecules with the ability to bind nuclear oestrogen receptors, regardless of oestrogenic or anti-oestrogenic nature, activate BK(Ca) channels through this nongenomic, membrane-delimited mechanism. The identity and characteristics of this putative binding site remain unclear; however, it has pharmacological similarity to oestrogen receptors alpha and beta, as ICI 182,780 interacts with it.
Collapse
Affiliation(s)
- Gregory M Dick
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, NV 89557, U.S.A.
| |
Collapse
|