1
|
Niwa T, Takanobu J, Suzuki K, Sato Y, Yamaguchi Y, Hayashi SI. Characterization of a membrane-associated estrogen receptor in breast cancer cells and its contribution to hormone therapy resistance using a novel selective ligand. J Steroid Biochem Mol Biol 2020; 201:105671. [PMID: 32289430 DOI: 10.1016/j.jsbmb.2020.105671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/23/2020] [Accepted: 04/03/2020] [Indexed: 11/24/2022]
Abstract
The estrogen receptor (ER) plays a role in the progression of hormone-dependent breast cancer and is a hormone therapy target. Estrogen acts as a transcription factor (genomic action) and also produces a quick non-genomic reaction through intracellular signaling pathways. The membrane associated ER (mER) may regulate both these signals and hormone therapy resistance. However, the details remain unclear because a reliable method to distinguish the signals induced by the estradiol (E2)-mER and E2-nuclear ER complex has not been established. In the present study, we prepared the novel ligand Qdot-6-E2, selective for mER, by coupling E2 with insoluble quantum dot nano-beads. We investigated the characteristics of mER signaling pathways and its contribution to hormone therapy resistance using different cell lines including estrogen depletion resistant (EDR) cells with different mechanisms. Qdot-6-E2 stimulated proliferation of nuclear ER-positive cells, but nuclear ER-negative cells showed no response. In addition, Qdot-6-E2 indirectly activated nuclear ER and increased mRNA expression of target genes. We confirmed that E2 was not dissociated from Qdot-6-E2 using a mammalian one-hybrid assay. We visually demonstrated that Qdot-6-E2 acts from the outside of cells. The gene expression profile induced by Qdot-6-E2-mER was different from that induced by E2-nuclear ER. The effect of anti-ER antibody, the GFP-ER fusion protein localization, and the effect of palmitoyl acyltransferase inhibitor also indicated the existence of mER. Regarding intracellular phosphorylation signaling pathways, the MAPK (Erk 1/2) and the PI3K/Akt pathways were both activated by Qdot-6-E2. In EDR cells, only nuclear ER-positive cells showed increased cell proliferation with increased localization of ERα to the membrane fraction. These findings suggested that Qdot-6-E2 reacts with ERα surrounding the cell membrane and that mER signals help the cells to survive under estrogen-depleted conditions by re-localizing the ER to use trace amounts of E2 more effectively. We expect that Qdot-6-E2 is a useful tool for studying the mER.
Collapse
Affiliation(s)
- Toshifumi Niwa
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Junko Takanobu
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Kanae Suzuki
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yuta Sato
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yuri Yamaguchi
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Shin-Ichi Hayashi
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
2
|
Blaustein JD. A personal view on traits useful for success in science: Daniel S. Lehrman Award Lecture. Horm Behav 2019; 111:3-6. [PMID: 30578819 DOI: 10.1016/j.yhbeh.2018.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/27/2018] [Accepted: 12/11/2018] [Indexed: 11/24/2022]
Abstract
As the 2018 recipient of the Daniel S. Lehrman Lifetime Achievement Award from the Society for Behavioral Neuroendocrinology, I was asked to give a short lecture in the Young Investigator Symposium of the combined Society for Behavioral Neuroendocrinology/International Congress on Neuroendocrinology meeting in Toronto. This lecture focused on one person's thoughts on what it takes to be successful in an academic science career. In this paper, I elaborate on success, on traits that may be useful for success in science, and on where the field of Behavioral Neuroendocrinology may be going.
Collapse
Affiliation(s)
- Jeffrey D Blaustein
- Department of Psychological and Brain Sciences, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
3
|
Micevych PE, Sinchak K. Extranuclear signaling by ovarian steroids in the regulation of sexual receptivity. Horm Behav 2018; 104:4-14. [PMID: 29753716 PMCID: PMC6240501 DOI: 10.1016/j.yhbeh.2018.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Paul E Micevych
- Dept of Neurobiology, David Geffen School of Medicine at UCLA, Laboratory of Neuroendocrinology of the UCLA Brain Research Institute, United States
| | - Kevin Sinchak
- Dept of Biological Sciences, California State University, Long Beach, United States.
| |
Collapse
|
4
|
Piffanelli A, Giovannini G, Pelizzola D, De Bortoli M. Estrogen and Progesterone Measurement and its Quality Control in Breast Cancer: A Reappraisal. Int J Biol Markers 2018; 1:15-28. [PMID: 3320222 DOI: 10.1177/172460088600100104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This article illustrates the two main methods for routine measurement of cytoplasmic estrogen receptor status in neoplastic biopsy. The first is the Dextran Coated Charcoal Technique (D.C.C. Assay) which is still the method of choice in the majority of clinical laboratories for its simplicity, reproducibility and low cost. The second is a more advanced technique based on the specific binding, enzimatically displayed, of commercially available antiestrogen monoclonal antibodies (Enzyme Immuno Assay - ABBOTT). The sui generis characteristics of endocrine sensitivity assessment on tumor tissues and the importance of decision-making connected with the assay justify rigorous quality assurance schemes. The quality control design proposed by the Italian Committee concerned the evaluation of several lyophilized preparations with scalar receptor content; this permits the identification through linear regression analysis of systematic and non-systematic errors. The Italian Committee has currently connected 50 labs from most regions of the country.
Collapse
|
5
|
Enhancement of the bioavailability of a novel anticancer compound (acetyltanshinone IIA) by encapsulation within mPEG-PLGA nanoparticles: a study of formulation optimization, toxicity, and pharmacokinetics. Oncotarget 2017; 8:12013-12030. [PMID: 28061455 PMCID: PMC5355322 DOI: 10.18632/oncotarget.14481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 12/16/2016] [Indexed: 11/29/2022] Open
Abstract
The Poly (ethylene glycol) methyl ether-block-poly (lactide-co-glycolide) (mPEG-PLGA) nanoparticles carrying acetyltanshinone IIA (ATA), a novel anti-breast cancer agent, were prepared by ultrasonic emulsion method to enhance the bioavailability and reduce the toxicity. Systematic optimization of encapsulation process was achieved using an orthogonal design. Drug efficacy analysis showed that ATA nanoparticles were as effective as free ATA against estrogen receptor positive breast cancer cells, but much less toxic towards human endothelial cells. Furthermore, in zebrafish, ATA nanoparticles displayed much lower toxicity than free ATA. More importantly, the blood concentration of ATA nanoparticles indicated by 24 hour-area under the curve (AUC0-24h) was 10 times higher than free ATA. These results indicated the potential of ATA-loaded mPEG-PLGA nanoparticles for the delivery of ATA in a clinical formulation, and their potential for use in tumor therapy in the future.
Collapse
|
6
|
|
7
|
Bain PA, Ogino Y, Miyagawa S, Iguchi T, Kumar A. Differential ligand selectivity of androgen receptors α and β from Murray-Darling rainbowfish (Melanotaenia fluviatilis). Gen Comp Endocrinol 2015; 212:84-91. [PMID: 25644213 DOI: 10.1016/j.ygcen.2015.01.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 01/20/2015] [Accepted: 01/24/2015] [Indexed: 11/16/2022]
Abstract
Androgen receptors (ARs) mediate the physiological effects of androgens in vertebrates. In fishes, AR-mediated pathways can be modulated by aquatic contaminants, resulting in the masculinisation of female fish or diminished secondary sex characteristics in males. The Murray-Darling rainbowfish (Melanotaenia fluviatilis) is a small-bodied freshwater teleost used in Australia as a test species for environmental toxicology research. We determined concentration-response profiles for selected agonists and antagonists of rainbowfish ARα and ARβ using transient transactivation assays. For both ARα and ARβ, the order of potency of natural agonists was 11-ketotestosterone (11-KT)>5α-dihydrotestosterone>testosterone>androstenedione. Methyltestosterone was a highly potent agonist of both receptors relative to 11-KT. The relative potency of the veterinary growth-promoting androgen, 17β-trenbolone, varied by more than a factor of 5 between ARα and ARβ. The non-steroidal anti-androgen bicalutamide exhibited high inhibitory potency relative to the structurally related model anti-androgen, flutamide. The inhibitory potency of the agricultural fungicide, vinclozolin, was approximately 1.7-fold relative to flutamide for ARα, but over 20-fold in the case of ARβ. Fluorescent protein tagging of ARs showed that the rainbowfish ARα subtype is constitutively localised to the nucleus, while ARβ is cytoplasmic in the absence of ligand, an observation which agrees with the reported subcellular localisation of AR subtypes from other teleost species. Collectively, these data suggest that M. fluviatilis ARα and ARβ respond differently to environmental AR modulators and that in vivo sensitivity to contaminants may depend on the tissue distribution of the AR subtypes at the time of exposure.
Collapse
Affiliation(s)
- Peter A Bain
- Land and Water Flagship, Commonwealth Scientific and Industrial Research Organisation, PMB 2, Glen Osmond, South Australia 5064, Australia.
| | - Yukiko Ogino
- Division of Molecular Environmental Endocrinology, National Institute for Basic Biology, Nishigonaka-38 Myodaijicho, Okazaki, Aichi Prefecture 444-0867, Japan
| | - Shinichi Miyagawa
- Division of Molecular Environmental Endocrinology, National Institute for Basic Biology, Nishigonaka-38 Myodaijicho, Okazaki, Aichi Prefecture 444-0867, Japan
| | - Taisen Iguchi
- Division of Molecular Environmental Endocrinology, National Institute for Basic Biology, Nishigonaka-38 Myodaijicho, Okazaki, Aichi Prefecture 444-0867, Japan
| | - Anupama Kumar
- Land and Water Flagship, Commonwealth Scientific and Industrial Research Organisation, PMB 2, Glen Osmond, South Australia 5064, Australia
| |
Collapse
|
8
|
A novel anti-cancer agent, acetyltanshinone IIA, inhibits oestrogen receptor positive breast cancer cell growth by down-regulating the oestrogen receptor. Cancer Lett 2013; 346:94-103. [PMID: 24374015 DOI: 10.1016/j.canlet.2013.12.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 11/27/2013] [Accepted: 12/10/2013] [Indexed: 02/07/2023]
Abstract
In this paper we show that acetyltanshinone IIA (ATA), a novel anti-cancer agent, preferentially inhibits cell growth of oestrogen receptor positive (ER+) breast cancer cells and that it is more potent than the commonly used anti-breast cancer agent, tamoxifen. The metabolic product of ATA, hydroquinone tanshinone IIA (HTA) binds to the ERα and causes its degradation mainly in the nucleus via an ubiquitin-mediated proteasome-dependent pathway. In addition, ATA also reduced the mRNA levels of the ERα encoding gene, ESR1, distinguishing ATA from another anti-breast cancer drug, fulvestrant. Finally, ATA reduced the transcription of an ER-responsive gene, GREB1.
Collapse
|
9
|
Panossian A, Hamm R, Kadioglu O, Wikman G, Efferth T. Synergy and Antagonism of Active Constituents of ADAPT-232 on Transcriptional Level of Metabolic Regulation of Isolated Neuroglial Cells. Front Neurosci 2013; 7:16. [PMID: 23430930 PMCID: PMC3576868 DOI: 10.3389/fnins.2013.00016] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 02/01/2013] [Indexed: 12/16/2022] Open
Abstract
Gene expression profiling was performed on the human neuroglial cell line T98G after treatment with adaptogen ADAPT-232 and its constituents – extracts of Eleutherococcus senticosus root, Schisandra chinensis berry, and Rhodiola rosea root as well as several constituents individually, namely, eleutheroside E, schizandrin B, salidroside, triandrin, and tyrosol. A common feature for all tested adaptogens was their effect on G-protein-coupled receptor signaling pathways, i.e., cAMP, phospholipase C (PLC), and phosphatidylinositol signal transduction pathways. Adaptogens may reduce the cAMP level in brain cells by down-regulation of adenylate cyclase gene ADC2Y and up-regulation of phosphodiesterase gene PDE4D that is essential for energy homeostasis as well as for switching from catabolic to anabolic states and vice versa. Down-regulation of cAMP by adaptogens may decrease cAMP-dependent protein kinase A activity in various cells resulting in inhibition stress-induced catabolic transformations and saving of ATP for many ATP-dependant metabolic transformations. All tested adaptogens up-regulated the PLCB1 gene, which encodes phosphoinositide-specific PLC and phosphatidylinositol 3-kinases (PI3Ks), key players for the regulation of NF-κB-mediated defense responses. Other common targets of adaptogens included genes encoding ERα estrogen receptor (2.9–22.6 fold down-regulation), cholesterol ester transfer protein (5.1–10.6 fold down-regulation), heat shock protein Hsp70 (3.0–45.0 fold up-regulation), serpin peptidase inhibitor (neuroserpin), and 5-HT3 receptor of serotonin (2.2–6.6 fold down-regulation). These findings can be reconciled with the observed beneficial effects of adaptogens in behavioral, mental, and aging-associated disorders. Combining two or more active substances in one mixture significantly changes deregulated genes profiles: synergetic interactions result in activation of genes that none of the individual substances affected, while antagonistic interactions result in suppression some genes activated by individual substances. These interactions can have an influence on transcriptional control of metabolic regulation both on the cellular level and the level of the whole organism. Merging of deregulated genes array profiles and intracellular networks is specific to the new substance with unique pharmacological characteristics. Presumably, this phenomenon could be used to eliminate undesirable effects (e.g., toxic effects) and increase the selectivity of pharmacological intervention.
Collapse
|
10
|
Lee HO, Sheen YY. Estrogen modulation of human breast cancer cell growth. Arch Pharm Res 2012; 20:566-71. [PMID: 18982261 DOI: 10.1007/bf02975213] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/1997] [Indexed: 11/30/2022]
Abstract
To gain further insight into how estrogens modulate cell function, the effects of estrogen on cell proliferation were studied in human breast cancer cells. We examined the effects of estrogen on the proliferation of three human breast cancer cell lines that differed in their estrogen receptor contents. Ten nM estradiol markedly stimulated the proliferation of MCF-7 human breast cancer cells that contained high levels of estrogen receptor (1.15+/-0.03 pmole/mg protein) over that of control. In T47D cells that contained low levels of estrogen receptor (0.23+/-0.05 pmole/mg protein), Ten nM estrogen slightly stimulated the proliferation over that of control. MDA-MB-231 cells, that contained no detectable levels of estrogen receptors, had their growth unaffected by estrogen. These results showed their sensitivity to growth stimulation by estrogen correlated well with their estrogen receptor content. Also we examined the effect of estrogen on cellular progesterone receptor level as well as plasminogen activator activity in MCF-7 cells. Ten nM estradiol showed maximal stimulation of progesterone receptor level as well as plasminogen activator activity in MCF-7 cells. It is not clear whether these stimulations of progesterone receptor and plasminogen activator activity by estrogen are related to the estrogen stimulation of cell proliferation of MCF-7 cells. Studies with estrogen in human breast cancer cells in culture indicate that sensitivity to growth stimulation by estrogen correlates well with estrogen receptor contents.
Collapse
Affiliation(s)
- H O Lee
- College of Pharmacy, Ewha Womans University, 120-750, Seoul, Korea
| | | |
Collapse
|
11
|
Lee HO, Sheen YY. Antiestrogen, trans-tamoxifen modulation of human breast cancer cell growth. Arch Pharm Res 2012; 20:572-8. [PMID: 18982262 DOI: 10.1007/bf02975214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/1997] [Indexed: 11/25/2022]
Abstract
To gain further insight into how antiestrogens modulate cell function, the effects of antiestrogen on cell proliferation were studied in human breast cancer cells. We examined the effects of trans-tamoxifen on the proliferation of three human breast cancer cell lines that differed in their estrogen receptor contents. Trans-tamoxifen (1 muM) markedly inhibited the estrogen stimulated proliferation of MCF-7 human breast cancer cells that contained high levels of estrogen receptor (1.15+/-0.03 pmole/mg protein) over that of control. In T47D cells that contained low levels of estrogen receptor (0.23+/-0.05 pmole/mg protein), trans-tamoxifen (1 muM) showed minimal inhibition of estrogen stimulated cell proliferation over that of control. MDA-MB-231 cells, that contained no detectable levels of estrogen receptors, had their growth unaffected by trans-tamoxifen treatment. These results showed their sensitivity to growth inhibition by antiestrogen correlated well with their estrogen receptor content. Also we activator activity in MCF-7 cells. Trans-tamoxifen (1 muM) showed maximal inhibition of estrogen stimulated progestrone receptor level as well as plasminogen activator activity in MCF-7 cells that were stimulated by estrogen. It is not clear whether these inhibitions of progestrone receptor and plasminogen activator activity by estrogen are related to the antiestrogen inhibition of cell proliferation of MCF-7 cells. From the results of this study, it is clearly demonstrated that trans-tamoxifen is an antiestrogen in MCF-7 human breast cancer cells. Our data suggest that the biological effectiveness of trans-tamoxifen appear to result from its affinity of interaction with the estrogen receptor.
Collapse
Affiliation(s)
- H O Lee
- College of Pharmacy, Ewha Womans University, 120-750, Seoul, Korea
| | | |
Collapse
|
12
|
Gintzler AR, Liu NJ. Importance of sex to pain and its amelioration; relevance of spinal estrogens and its membrane receptors. Front Neuroendocrinol 2012; 33:412-24. [PMID: 23036438 PMCID: PMC3778676 DOI: 10.1016/j.yfrne.2012.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 09/13/2012] [Accepted: 09/18/2012] [Indexed: 12/14/2022]
Abstract
Estrogens have a multitude of effects on opioid systems and are thought to play a key role in sexually dimorphic nociception and opioid antinociception. Heretofore, classical genomic actions of estrogens are largely thought to be responsible for the effects of these steroids on nociception and opioid antinociception. The recent discovery that estrogens can also activate estrogen receptors that are located in the plasma membrane, the effects of which are manifest in seconds to minutes instead of hours to days has revolutionized our thinking concerning the ways in which estrogens are likely to modulate pain responsiveness and the dynamic nature of that modulation. This review summarizes parameters of opioid functionality and nociception that are subject to modulation by estrogens, underscoring the added dimensions of such modulation that accrues from rapid membrane estrogen receptor signaling. Implications of this mode of signaling regarding putative sources of estrogens and its degradation are also discussed.
Collapse
Affiliation(s)
- Alan R Gintzler
- State University of New York, Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | | |
Collapse
|
13
|
Alemany M. Do the interactions between glucocorticoids and sex hormones regulate the development of the metabolic syndrome? Front Endocrinol (Lausanne) 2012; 3:27. [PMID: 22649414 PMCID: PMC3355885 DOI: 10.3389/fendo.2012.00027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 02/06/2012] [Indexed: 12/14/2022] Open
Abstract
The metabolic syndrome is basically a maturity-onset disease. Typically, its manifestations begin to flourish years after the initial dietary or environmental aggression began. Since most hormonal, metabolic, or defense responses are practically immediate, the procrastinated response do not seem justified. Only in childhood, the damages of the metabolic syndrome appear with minimal delay. Sex affects the incidence of the metabolic syndrome, but this is more an effect of timing than absolute gender differences, females holding better than males up to menopause, when the differences between sexes tend to disappear. The metabolic syndrome is related to an immune response, countered by a permanent increase in glucocorticoids, which keep the immune system at bay but also induce insulin resistance, alter the lipid metabolism, favor fat deposition, mobilize protein, and decrease androgen synthesis. Androgens limit the operation of glucocorticoids, which is also partly blocked by estrogens, since they decrease inflammation (which enhances glucocorticoid release). These facts suggest that the appearance of the metabolic syndrome symptoms depends on the strength (i.e., levels) of androgens and estrogens. The predominance of glucocorticoids and the full manifestation of the syndrome in men are favored by decreased androgen activity. Low androgens can be found in infancy, maturity, advanced age, or because of their inhibition by glucocorticoids (inflammation, stress, medical treatment). Estrogens decrease inflammation and reduce the glucocorticoid response. Low estrogen (infancy, menopause) again allow the predominance of glucocorticoids and the manifestation of the metabolic syndrome. It is postulated that the equilibrium between sex hormones and glucocorticoids may be a critical element in the timing of the manifestation of metabolic syndrome-related pathologies.
Collapse
Affiliation(s)
- Marià Alemany
- Faculty of Biology, Department of Nutrition and Food Science, University of Barcelona Barcelona, Spain.
| |
Collapse
|
14
|
Mani SK, Blaustein JD. Neural progestin receptors and female sexual behavior. Neuroendocrinology 2012; 96:152-61. [PMID: 22538437 PMCID: PMC3498483 DOI: 10.1159/000338668] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 04/02/2012] [Indexed: 01/06/2023]
Abstract
The steroid hormone, progesterone (P), modulates neuroendocrine functions in the central nervous system resulting in integration of reproduction and reproductive behaviors in female mammals. Although it is widely recognized that P's effects on female sex behavior are mediated by the classical neural progestin receptors (PRs) functioning as 'ligand-dependent' transcription factors to regulate genes and genomic networks, additional mechanisms of PR activation also contribute to the behavioral response. Cellular and molecular evidence indicates that PRs can be activated in a ligand-independent manner by neurotransmitters, growth factors, cyclic nucleotides, progestin metabolites and mating stimuli. The rapid responses of P may be mediated by a variety of PR types, including membrane-associated PRs or extranuclear PRs. Furthermore, these rapid nonclassical P actions involving cytoplasmic kinase signaling and/or extranuclear PRs also converge with classical PR-mediated transcription-dependent pathways to regulate reproductive behaviors. In this review, we summarize some of the history of the study of the role of PRs in reproductive behaviors and update the status of PR-mediated mechanisms involved in the facilitation of female sex behavior. We present an integrative model of PR activation via crosstalk and convergence of multiple signaling pathways.
Collapse
Affiliation(s)
- Shaila K Mani
- Department of Molecular and Cellular Biology, Department of Neuroscience, Center on Addiction, Learning and Memory, Baylor College of Medicine, Houston, TX 77030, USA.
| | | |
Collapse
|
15
|
Papaioannou M, Schleich S, Prade I, Degen S, Roell D, Schubert U, Tanner T, Claessens F, Matusch R, Baniahmad A. The natural compound atraric acid is an antagonist of the human androgen receptor inhibiting cellular invasiveness and prostate cancer cell growth. J Cell Mol Med 2010; 13:2210-2223. [PMID: 18627423 DOI: 10.1111/j.1582-4934.2008.00426.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Extracts from Pygeum africanum are used in the treatment of prostatitis, benign prostatic hyperplasia and prostate cancer (Pca), major health problems of men in Western countries. The ligand-activated human androgen receptor (AR) supports the growth of the prostate gland. Inhibition of human AR by androgen ablation therapy and by applying synthetic anti-androgens is therefore the primary goal in treatment of patients. Here, we show that atraric acid (AA) isolated from bark material of Pygeum africanum has anti-androgenic activity, inhibiting the transactivation mediated by the ligand-activated human AR. This androgen antagonistic activity is receptor specific and does not inhibit the closely related glucocorticoid or progesterone receptors. Mechanistically, AA inhibits nuclear transport of AR. Importantly, AA is able to efficiently repress the growth of both the androgen-dependent LNCaP and also the androgen-independent C4-2 Pca cells but not that of PC3 or CV1 cells lacking AR. In line with this, AA inhibits the expression of the endogenous prostate specific antigen gene in both LNCaP und C4-2 cells. Analyses of cell invasion revealed that AA inhibits the invasiveness of LNCaP cells through extracellular matrix. Thus, this study provides a molecular insight for AA as a natural anti-androgenic compound and may serve as a basis for AA derivatives as a new chemical lead structure for novel therapeutic compounds as AR antagonists, that can be used for prophylaxis or treatment of prostatic diseases.
Collapse
Affiliation(s)
- Maria Papaioannou
- Institute of Human Genetics and Anthropology, Friedrich-Schiller-University, Kollegiengasse, Jena, Germany
| | - Sonja Schleich
- Institute for Pharmaceutical Chemistry, Philipps-University Marburg, Marbacher Weg, Marburg, Germany
| | - Ina Prade
- Institute of Human Genetics and Anthropology, Friedrich-Schiller-University, Kollegiengasse, Jena, Germany
| | - Stephanie Degen
- Institute of Human Genetics and Anthropology, Friedrich-Schiller-University, Kollegiengasse, Jena, Germany
| | - Daniela Roell
- Institute of Human Genetics and Anthropology, Friedrich-Schiller-University, Kollegiengasse, Jena, Germany
| | - Undine Schubert
- Institute of Human Genetics and Anthropology, Friedrich-Schiller-University, Kollegiengasse, Jena, Germany
| | - Tamzin Tanner
- Department of Molecular Cell Biology, University of Leuven, Heresraat, Leuven, Belgium
| | - Frank Claessens
- Department of Molecular Cell Biology, University of Leuven, Heresraat, Leuven, Belgium
| | - Rudolf Matusch
- Institute for Pharmaceutical Chemistry, Philipps-University Marburg, Marbacher Weg, Marburg, Germany
| | - Aria Baniahmad
- Institute of Human Genetics and Anthropology, Friedrich-Schiller-University, Kollegiengasse, Jena, Germany.,Department of Molecular Cell Biology, University of Leuven, Heresraat, Leuven, Belgium
| |
Collapse
|
16
|
Orihuela PA, Zuñiga LM, Rios M, Parada-Bustamante A, Sierralta WD, Velásquez LA, Croxatto HB. Mating changes the subcellular distribution and the functionality of estrogen receptors in the rat oviduct. Reprod Biol Endocrinol 2009; 7:139. [PMID: 19948032 PMCID: PMC2789086 DOI: 10.1186/1477-7827-7-139] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 11/30/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mating changes the mode of action of 17beta-estradiol (E2) to accelerate oviductal egg transport from a nongenomic to a genomic mode, although in both pathways estrogen receptors (ER) are required. This change was designated as intracellular path shifting (IPS). METHODS Herein, we examined the subcellular distribution of ESR1 and ESR2 (formerly known as ER-alpha and ER-beta) in oviductal epithelial cells of rats on day 1 of cycle (C1) or pregnancy (P1) using immunoelectron microscopy for ESR1 and ESR2. The effect of mating on intraoviductal ESR1 or ESR2 signaling was then explored comparing the expression of E2-target genes c-fos, brain creatine kinase (Ckb) and calbindin 9 kDa (s100g) in rats on C1 or P1 treated with selective agonists for ESR1 (PPT) or ESR2 (DPN). The effect of ER agonists on egg transport was also evaluated on C1 or P1 rats. RESULTS Receptor immunoreactivity was associated with the nucleus, cytoplasm and plasma membrane of the epithelial cells. Mating affected the subcellular distribution of both receptors as well as the response to E2. In C1 and P1 rats, PPT increased Ckb while both agonists increased c-fos. DPN increased Ckb and s100g only in C1 and P1 rats, respectively. PPT accelerated egg transport in both groups and DPN accelerated egg transport only in C1 rats. CONCLUSION Estrogen receptors present a subcellular distribution compatible with E2 genomic and nongenomic signaling in the oviductal epithelial cells of C1 and P1 although IPS occurs independently of changes in the distribution of ESR1 and ESR2 in the oviductal epithelial cells. Mating affected intraoviductal ER-signaling and induced loss of functional involvement of ESR2 on E2-induced accelerated egg transport. These findings reveal a profound influence on the ER signaling pathways exerted by mating in the oviduct.
Collapse
Affiliation(s)
- Pedro A Orihuela
- Laboratorio de Inmunología de la Reproducción, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Centro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNA, Santiago, Chile
| | - Lidia M Zuñiga
- Millennium Institute for Fundamental and Applied Biology, Santiago, Chile
| | - Mariana Rios
- Unidad de Reproducción y Desarrollo, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Walter D Sierralta
- Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Luis A Velásquez
- Laboratorio de Inmunología de la Reproducción, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Centro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNA, Santiago, Chile
| | - Horacio B Croxatto
- Laboratorio de Inmunología de la Reproducción, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Centro para el Desarrollo en Nanociencia y Nanotecnología-CEDENNA, Santiago, Chile
- Millennium Institute for Fundamental and Applied Biology, Santiago, Chile
| |
Collapse
|
17
|
Boulware MI, Mermelstein PG. Membrane estrogen receptors activate metabotropic glutamate receptors to influence nervous system physiology. Steroids 2009; 74:608-13. [PMID: 19070630 PMCID: PMC2799184 DOI: 10.1016/j.steroids.2008.11.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 11/06/2008] [Accepted: 11/07/2008] [Indexed: 11/21/2022]
Abstract
Until recently, the idea that estradiol could affect cellular processes independent of nuclear estrogen receptors was often dismissed as artifact. This in spite of a large number of carefully controlled studies performed both within and outside the nervous system demonstrating estrogens regulate various intracellular signaling pathways by acting at the membrane surface of cells and/or at biological rates incompatible with the time course of genomic-initiated events. The concept that estradiol can act on surface membrane receptors to regulate nervous system function is now far less controversial. However, there is evidence that there may be multiple types of estrogen receptors on the membrane surface of cells. Determining the physiological relevance of each of these receptors is currently underway. Two important membrane estrogen receptors are in fact the classical estrogen receptor-alpha (ERalpha) and estrogen receptor-beta (ERbeta) proteins, which is somewhat surprising based upon their well-established role in nuclear gene transcription. This review will focus on the mechanism by which surface-localized ERalpha and ERbeta stimulate intracellular signaling events in cells of the nervous system through activation of metabotropic glutamate receptors (mGluRs). This mechanism of estrogen receptor function also requires caveolin proteins, which provide the subcellular compartmentalization of the particular signaling components required for appropriate cell stimulation. The review will conclude with several examples of physiological processes under the apparent regulation of ER/mGluR signaling.
Collapse
Affiliation(s)
- Marissa I. Boulware
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | - Paul G. Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| |
Collapse
|
18
|
Affiliation(s)
- R Dreicer
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City 52242
| | | |
Collapse
|
19
|
Lu IF, Hasio AC, Hu MC, Yang FM, Su HM. Docosahexaenoic acid induces proteasome-dependent degradation of estrogen receptor alpha and inhibits the downstream signaling target in MCF-7 breast cancer cells. J Nutr Biochem 2009; 21:512-7. [PMID: 19369047 DOI: 10.1016/j.jnutbio.2009.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 02/04/2009] [Accepted: 02/16/2009] [Indexed: 02/07/2023]
Abstract
About two thirds of breast cancers in women are hormone-dependent and require estrogen for growth, its effects being mainly mediated through estrogen receptor alpha (ERalpha). Docosahexaenoic acid (DHA, 22:6n-3) and arachidonic acid (AA, 20:4n-6) have opposite effects on carcinogenesis, with DHA suppressing and AA promoting tumor growth both in vitro and in vivo. However, the mechanism is not clear. Here, we examined whether the effect is mediated through changes in ERalpha distribution. MCF-7 cells, an ERalpha-positive human breast cancer cell line, was cultured in estrogen-free medium containing 0, 10 or 60 microM DHA or AA, then were stimulated with estradiol. DHA supplementation resulted in down-regulation of ERalpha expression (particularly in the extranuclear fraction), a reduction in phosphorylated MAPK, a decrease in cyclin D1 levels and an inhibition in cell viability. In contrast, AA had no such effects. The DHA-induced decrease in ERalpha expression resulted from proteasome-dependent degradation and not from decreased ERalpha mRNA expression. We propose that breast cancer cell proliferation is inhibited by DHA through proteasome-dependent degradation of ERalpha, reduced cyclin D1 expression and inhibition of MAPK signaling.
Collapse
Affiliation(s)
- I-Fen Lu
- Department of Physiology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | | | | | | | | |
Collapse
|
20
|
Mermelstein PG. Membrane-localised oestrogen receptor alpha and beta influence neuronal activity through activation of metabotropic glutamate receptors. J Neuroendocrinol 2009; 21:257-62. [PMID: 19207809 PMCID: PMC2805164 DOI: 10.1111/j.1365-2826.2009.01838.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Until recently, the idea that oestradiol could affect cellular processes independent of nuclear oestrogen receptors (ERs) was controversial. This was despite the large number of carefully controlled studies performed both within and outside the nervous system demonstrating that oestrogens regulate various intracellular signalling pathways by acting at the membrane surface of cells and/or at biological rates incompatible with the time course of genomic-initiated events. At present, it is far less controversial that oestradiol acts at surface membrane receptors to regulate nervous system function. Recent studies have demonstrated that the classical intracellular ERs, ERalpha and ERbeta, are major players in mediating the actions of oestradiol on the membrane surface. This review focuses on one potential mechanism by which surface-localised ERalpha and ERbeta stimulate intracellular signalling events in cells of the nervous system. After oestradiol treatment, both ERalpha and ERbeta are capable of activating different classes of metabotropic glutamate receptors (mGluRs). Oestradiol activation of mGluRs is independent of glutamate, but requires expression of several different caveolin proteins to compartmentalise the different ERs with mGluRs into functional signalling microdomains. ER/mGluR signalling is a potential means by which oestrogens can both rapidly and for extended periods, influence a variety of intracellular signalling processes and behaviours.
Collapse
Affiliation(s)
- P G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
21
|
Shi H, Clegg DJ. Sex differences in the regulation of body weight. Physiol Behav 2009; 97:199-204. [PMID: 19250944 DOI: 10.1016/j.physbeh.2009.02.017] [Citation(s) in RCA: 188] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 02/12/2009] [Accepted: 02/20/2009] [Indexed: 10/21/2022]
Abstract
Obesity and its associated health disorders and costs are increasing. Males and females differ in terms of how and where body fat is stored, the hormones they secrete in proportion to their fat, and the way their brains respond to signals that regulate body fat. Fat accumulation in the intra-abdominal adipose depot is associated with the risk for developing cardiovascular problems, type-2 diabetes mellitus, certain cancers and other disorders. Men and postmenopausal women accumulate more fat in the intra-abdominal depot than do pre-menopausal women, and therefore have a greater risk of developing metabolic complications associated with obesity. The goal of this review is to explore what we know about sexual dimorphisms in adipose tissue accrual and deposition. Elucidating the mechanisms by which sex hormones may modulate the way in which fat is accumulated and stored is a critical area of research due to the prevalence of obesity and the metabolic syndrome, and the rapid increase in propensity for these diseases following menopause.
Collapse
Affiliation(s)
- H Shi
- Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, OH 45237, USA
| | | |
Collapse
|
22
|
Mylonas I, Makovitzky J, Friese K, Jeschke U. Immunohistochemical labelling of steroid receptors in normal and malignant human endometrium. Acta Histochem 2009; 111:349-59. [PMID: 19195687 DOI: 10.1016/j.acthis.2008.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
For several years it was generally believed that only a single estrogen receptor (ER) and progesterone receptor (PR) existed. However, the discovery of a new ER (ERbeta) with specificity for estrogens has induced new insights in the estrogen signalling system. Moreover, PR is expressed as two major isoforms, PR-A and PR-B that arise from alternative transcriptional starting sites within the same gene. Although PR-A and PR-B were thought to occur in similar amounts, it is now clear that they are differentially expressed and thus have distinct functions in several human tissues, including human endometrium. The ER and PR expression and distribution pattern might play an important role in normal endometrial function and pathogenesis and the expression and relationship of the two distinct ER's and PR's could be of essential clinical implications. Moreover, the imbalance in ERalpha/ERbeta expression and the PR-A/PR-B ratio might play an important role in endometrial transition and subsequently influence endometrial pathogenesis. The knowledge of the pattern of steroid receptors in human endometrial tissue is of extreme importance, since it might start a new field in hormone therapy of endometrial cancer.
Collapse
|
23
|
Möller FJ, Zierau O, Hertrampf T, Bliedtner A, Diel P, Vollmer G. Long-term effects of dietary isoflavones on uterine gene expression profiles. J Steroid Biochem Mol Biol 2009; 113:296-303. [PMID: 19429436 DOI: 10.1016/j.jsbmb.2009.01.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 01/22/2009] [Accepted: 01/22/2009] [Indexed: 10/21/2022]
Abstract
Isoflavones (ISOs) are bioactive food ingredients of the traditional East Asian diet and currently discussed as alternatives to classical hormone replacement therapies and for reducing the prevalence of hormone-dependent cancers. Although there are many studies on ISOs, not much is known about their long-term effects. Therefore, we performed an animal experiment analyzing the effects of three different diets: a phytoestrogen-free diet, a diet supplemented with genistein (700 microg/g diet) and an ISO-high diet (232 microg daidzein and 240 microg genistein/g) at two distinct time points, juvenile (21 days) and adult (97 days). Exposure started prior to mating of the parents and throughout the life of the offspring. We observed a stronger increase of uterine wet weights in juvenile offspring with genistein exposure (1018+/-350 mg/kg BW) than with ISO-high diet (497+/-133 mg/kg BW). Whereas the expression of proliferation related genes (PCNA; Ki67; IGF-1; IGF-1R), analyzed by real-time-qPCR and Western blot, were significantly down-regulated in juvenile animals exposed to genistein. Additionally, genistein exposure led to estrogenic responses, observed upon increase of complement C3 and decrease of estrogen receptors gene expressions, while the exposure to ISO-high diet did not show these effects. In conclusion, both the time point on which phytoestrogen exposure starts together with the composition of the ingested phytoestrogen containing diet are of great importance for the biological response of the offspring.
Collapse
Affiliation(s)
- Frank Josef Möller
- Institute for Zoology, Molecular Cell Physiology and Endocrinology, Technical University Dresden, Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
24
|
Affiliation(s)
- Brandon C Wadas
- Colorado State University, Department of Biomedical Sciences, 1617 Campus Delivery, Fort Collins, Colorado 80523, USA
| | | |
Collapse
|
25
|
Micevych PE, Mermelstein PG. Membrane estrogen receptors acting through metabotropic glutamate receptors: an emerging mechanism of estrogen action in brain. Mol Neurobiol 2008; 38:66-77. [PMID: 18670908 DOI: 10.1007/s12035-008-8034-z] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Accepted: 06/18/2008] [Indexed: 12/27/2022]
Abstract
It has been over 60 years since the first studies have been published describing the effects of steroid hormones on brain function. For over 30 years, estrogen has been presumed to directly affect gene expression and protein synthesis through a specific receptor. More than 20 years ago, the first estrogen receptor was cloned and identified as a transcription factor. Yet, throughout their course of study, estrogens have also been observed to affect nervous system function via mechanisms independent of intracellular receptor regulation of gene expression. Up until recently, the membrane estrogen receptors responsible for these rapid actions have remained elusive. Recent studies have demonstrated that a large number of these rapid, membrane-initiated actions of estradiol are due to surface expression of classical estrogen receptors. This review focuses on the importance of membrane estrogen receptor interactions with metabotropic glutamate receptors for understanding rapid estradiol signaling mechanisms and downstream effectors, as well as their significance in a variety of physiological processes.
Collapse
Affiliation(s)
- Paul E Micevych
- Department of Neurobiology and Laboratory of Neuroendocrinology of the Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | | |
Collapse
|
26
|
Milanesi L, de Boland AR, Boland R. Expression and localization of estrogen receptor α in the C2C12 murine skeletal muscle cell line. J Cell Biochem 2008; 104:1254-73. [DOI: 10.1002/jcb.21706] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
27
|
Lichti U, Yuspa SH. Inhibition of epidermal terminal differentiation and tumour promotion by retinoids. CIBA FOUNDATION SYMPOSIUM 2008; 113:77-89. [PMID: 2863095 DOI: 10.1002/9780470720943.ch6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Retinoids are physiological regulators of growth and differentiation for a number of epithelial tissues. In several of these, retinoids also act as pharmacological anticarcinogens. Retinoids are most effective as anticarcinogens in the post-initiation portion of carcinogenesis. In mouse skin, retinoids are inhibitors of phorbol ester-mediated tumour promotion and can cause regression of pre-existing benign tumours. Studies in vivo and in vitro have indicated that phorbol ester-mediated skin tumour promotion results from selective clonal expansion of initiated cells. We have proposed that the biological basis for selection resides in the induction of terminal differentiation in subpopulations of keratinocytes while other keratinocytes, including initiated cells, are stimulated to proliferate. Terminal differentiation is accelerated by phorbol esters through the induction of epidermal transglutaminase and consequent cornification. Retinoids inhibit terminal differentiation of keratinocytes. Retinoids also induce transglutaminase in epidermis, but they inhibit cornification. Recent results suggest a biochemical basis for this paradox. The phorbol ester-induced transglutaminase is primarily particulate but the retinoid-induced enzyme is cytosolic. The induced enzymes differ in kinetic parameters, thermal stability and in elution from ion-exchange columns. Induction of the retinoid enzyme is associated with suppression of the induction of transglutaminase by phorbol esters. The retinoid-induced epidermal transglutaminase could interfere with normal or promoter-induced differentiation by inappropriately cross-linking precursor proteins before their assembly at the cell periphery. This could explain one aspect of the inhibitory action of retinoids on tumour promotion.
Collapse
|
28
|
Davey JC, Bodwell JE, Gosse JA, Hamilton JW. Arsenic as an Endocrine Disruptor: Effects of Arsenic on Estrogen Receptor–Mediated Gene Expression In Vivo and in Cell Culture. Toxicol Sci 2007; 98:75-86. [PMID: 17283378 DOI: 10.1093/toxsci/kfm013] [Citation(s) in RCA: 179] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Arsenic (As) contamination of drinking water is considered a serious worldwide environmental health threat that is associated with increased disease risks including skin, lung, bladder, and other cancers; type 2 diabetes; vascular and cardiovascular diseases; reproductive and developmental effects; and neurological and cognitive effects. Increased health risks may occur at as low as 10-50 ppb, while biological effects have been observed in experimental animal and cell culture systems at much lower levels. We previously reported that As is a potent endocrine disruptor, altering gene regulation by the closely related glucocorticoid, mineralocorticoid, progesterone, and androgen steroid receptors (SRs) at concentrations as low as 0.01 microM ( approximately 0.7 ppb). Very low doses enhanced hormone-mediated gene transcription, whereas slightly higher but still noncytotoxic doses were suppressive. We report here that As also disrupts the more distally related estrogen receptor (ER) both in vivo and in cell culture. At noncytotoxic doses (1-50 micromol/kg arsenite) As strongly suppressed ER-dependent gene transcription of the 17beta-estradiol (E2)-inducible vitellogenin II gene in chick embryo liver in vivo. In cell culture, noncytotoxic levels (0.25-3 microM, approximately 20-225 ppb) of As significantly inhibited E2-mediated gene activation of an ER-regulated reporter gene and the native ER-regulated GREB1 gene in human breast cancer MCF-7 cells. While the effects of As on ER-dependent gene regulation were generally similar to As effects on the other SRs, there were specific differences, particularly the lack of significant enhancement at the lowest doses, that may provide insights into possible mechanisms.
Collapse
Affiliation(s)
- Jennifer C Davey
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755-3835, USA
| | | | | | | |
Collapse
|
29
|
Estrogen-induced modification of uterine RNA polymerase activity depends on localization of the estrogen receptor. ARCH BIOL SCI 2007. [DOI: 10.2298/abs0702105z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The aim of this study was to examine the effects of estradiol (E2) on activity of RNA polymerase I and RNA polymerase II in uterine nuclei of ovariectomized (OVX) female rats. The obtained results show that estrogen-receptor (E-R) complexes in 30 min induced an increase of polymerase II activity. A second increase of polymerase II activity was observed after 3 h-incubation of nuclei with the E-R complex formed in the cytosol fraction. However, activity of polymerase I was increased 2 h after the start of incubation, with highest activity detected at 3 h in nuclei incubated with E-R complexes. On the contrary, no stimulatory effect on either polymerase I or polymerase II activity was observed in nuclei incubated with E2 alone. These results indicate that E2 stimulates the cytosolic estrogen receptor (ER), which in turn causes uterotrophic responses in OVX rats. In addition, they suggest that in order to provoke uterotrophic responses E-R complexes formed in the cytosol need to be retained in the nucleus for a longer period of time. .
Collapse
|
30
|
Damodaran KM, Epperly MW, Pillai KMR, Bloomer WD. A facile and improved synthesis of 17α-{2-(E)-[1 2 5 I]-iodovinyl}-19-nortestosterone, a no-carrier-added ligand for progesterone receptor analyses. J Labelled Comp Radiopharm 2006. [DOI: 10.1002/jlcr.2580340104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
31
|
Márquez DC, Chen HW, Curran EM, Welshons WV, Pietras RJ. Estrogen receptors in membrane lipid rafts and signal transduction in breast cancer. Mol Cell Endocrinol 2006; 246:91-100. [PMID: 16388889 DOI: 10.1016/j.mce.2005.11.020] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Regulation of breast cancer growth by estrogen is mediated by estrogen receptors (ER) in nuclear and extranuclear compartments. We assessed the structure and functions of extranuclear ER that initiate downstream signaling to the nucleus. ER, including full-length 66-kDa ER and a 46-kDa ER splice variant, are enriched in lipid rafts from MCF-7 cells with (MCF-7/HER-2) or without (MCF-7/PAR) HER-2 gene overexpression and co-localize with HER-1 and HER-2 growth factor receptors, as well as with lipid raft marker flotillin-2. In contrast, ER-negative MCF-7 cells do not express nuclear or lipid raft ER. ER knockdown with siRNA also elicits a marked loss of ER in MCF-7 cell rafts. In MCF-7/PAR cells, estrogen enhances ER association with membrane rafts and induces rapid phosphorylation of nuclear receptor coactivator AIB1, actions not detected in ER-negative cells. Thus, nuclear and lipid raft ER derive from the same transcript, and extranuclear ER co-localizes with HER receptors in membrane signaling domains that modulate downstream nuclear events leading to cell growth.
Collapse
Affiliation(s)
- Diana C Márquez
- UCLA School of Medicine, Department of Medicine, Division of Hematology-Oncology, Los Angeles, CA 90095-1678, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
This contribution will trace some of the many seminal studies on the female uterus (endometrium) over the centuries and conclude with a description of some current research initiatives in our laboratory. Numerous contributions from many investigators over the years have contributed to our current understanding of endometrial function. The historical section of this chapter is intended to be a brief overall description of some of these efforts and not exhaustive. Additional information can be found in the review articles and books cited herein.
Collapse
Affiliation(s)
- William C Okulicz
- Department of Physiology, UMass Medical School, 55 Lake Avenue N, Worcester, MA 01655, USA.
| |
Collapse
|
33
|
Saradhi M, Sengupta A, Mukhopadhyay G, Tyagi RK. Pregnane and Xenobiotic Receptor (PXR/SXR) resides predominantly in the nuclear compartment of the interphase cell and associates with the condensed chromosomes during mitosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1746:85-94. [PMID: 16297466 DOI: 10.1016/j.bbamcr.2005.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2004] [Revised: 09/29/2005] [Accepted: 10/07/2005] [Indexed: 10/25/2022]
Abstract
Pregnane and Xenobiotic Receptor (PXR) is a transcription factor that is activated by a diverse range of xenobiotics and endogenous metabolites including steroids, bile acids and about 50% of the prescription drugs. In specific cell types (e.g. liver and intestine) it serves as a 'xenosensor' by regulating expression of a network of genes involved in xenobiotic clearance from the body. PXR expression in several cancerous tissues and its regulated expression of multi-drug resistance proteins highlight its significance in prognosis of malignancies. The view that subcellular localization and ligand induced movements of transcription factors is one of the major phenomena in regulating transcriptional activity, we used a green fluorescent protein tagged PXR chimera to study its dynamic behaviour in living cells. Under all experimental conditions, PXR was observed to be a predominantly nuclear protein maintaining a dynamic equilibrium between the nuclear and cytoplasmic compartments of the interphase cells. Interestingly, for the first time, a member of the nuclear receptor superfamily, PXR, has been observed to be associated with condensed chromosomes during all the mitotic stages of cell division. The significance of PXR association with mitotic chromosomes is discussed.
Collapse
Affiliation(s)
- Mallampati Saradhi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi - 110067, India
| | | | | | | |
Collapse
|
34
|
Mukherjee K, Syed V, Ho SM. Estrogen-induced loss of progesterone receptor expression in normal and malignant ovarian surface epithelial cells. Oncogene 2005; 24:4388-400. [PMID: 15806153 DOI: 10.1038/sj.onc.1208623] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
While estrogens are suspected risk factors for epithelial ovarian cancer (OCa), progesterone (P4) has been shown to exert protective effects. The biological actions of P4 in target cells are mediated by progesterone receptors (PRs) that exist principally as A- and B-isoforms. We observed overexpression of PR-A and PR-B protein in two lines of OCa cells when compared to two lines of nontumorigenic, normal human ovarian surface epithelial (HOSE) cells. Treatment of HOSE or OCa cells with estrone or 17beta-estradiol at 10(-8) M for a period of 72 h induced significant loss of PR-A and PR-B mRNA and protein expression, with the regulation primarily controlled at the transcriptional level. In contrast, breast cancer cells (line MCF-7) exposed to estrogens upregulated PR-A and PR-B expression. Of significance, both the inhibitory and stimulatory actions of estrogens were blocked by the specific ER-antagonist ICI 182,780 (ICI, 10(-5) M), confirming estrogen specificity. Co-treatment of estrogen-exposed HOSE, OCa, and MCF-7 cell lines with inhibitors of type 1- and type 2-17beta hydroxysteroid dehydrogenase did not affect the previously observed changes in PR expression, suggesting that the action of each estrogen is direct and not mediated via conversion to its metabolic counterpart. Green fluorescence protein (GFP)-PR-A and GFP-PR-B were localized in the cytoplasmic compartment of untreated HOSE cells and translocated to the nucleus after P4 treatment, while both chimera PRs resided in the nuclei of OCa cells in a ligand-independent manner. In OCa cell cultures, P4 (10(-6) M), but not RU486 (10(-5) M), induced apoptosis that was blocked by co-treatment with the antiprogestin but enhanced by co-treatment with ICI. In sharp contrast, P4 induced proliferation, while ICI and RU486 caused cell death in MCF-7 cells. In conclusion, this study is first to demonstrate estrogens as negative regulators of PR expression in HOSE/OCa cells and to provide a mechanistic basis upon which to explain the antagonism of estrogens on the anti-OCa action of progestins. It also raises the possibility of using progestin and ICI as a combinational therapy for OCa treatment.
Collapse
Affiliation(s)
- Kasturi Mukherjee
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | |
Collapse
|
35
|
Oenga GN, Spink DC, Carpenter DO. TCDD and PCBs inhibit breast cancer cell proliferation in vitro. Toxicol In Vitro 2005; 18:811-9. [PMID: 15465647 DOI: 10.1016/j.tiv.2004.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Accepted: 04/09/2004] [Indexed: 11/28/2022]
Abstract
The effects on cell proliferation of arylhydrocarbon receptor (AhR) agonists in estrogen-responsive T47D and ZR-75-1 cells were investigated. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and the non-ortho-substituted polychlorinated biphenyl (PCB) congeners, PCB 77, PCB 81, PCB 126, and PCB 169 each inhibited 17beta-estradiol (E(2))-stimulated cell proliferation in a dose-responsive manner. In the absence of added E(2), TCDD, PCB 77, PCB 81, and PCB 169 had no significant effect on cell proliferation, while PCB 126 at high concentrations caused slight elevations. The order of effective inhibition of E(2)-stimulated cell proliferation by the PCB congeners was: PCB 81>PCB 126 approximately = PCB 169>PCB 77. In the comparative literature, mammalian TEFs for these congeners toxic potency are in the order: PCB 126>PCB 169>PCB 81 approximately = PCB 77 [Organohalogen Compd. 34 (1997) 237]. Our results thus show an unexpected different pattern for the inhibitory effects of PCBs congeners on E(2)-mediated cell proliferation.
Collapse
Affiliation(s)
- Gideon N Oenga
- Wadsworth Center, New York State Department of Health, Albany, NY 12201-0509, USA
| | | | | |
Collapse
|
36
|
Wang Y, Cheng CHK. ERalpha and STAT5a cross-talk: interaction through C-terminal portions of the proteins decreases STAT5a phosphorylation, nuclear translocation and DNA-binding. FEBS Lett 2004; 572:238-44. [PMID: 15304355 DOI: 10.1016/j.febslet.2004.06.098] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2004] [Revised: 06/19/2004] [Accepted: 06/23/2004] [Indexed: 01/12/2023]
Abstract
Cross-talk between ERalpha and STAT5a was demonstrated to mediate through a direct physical association between the two proteins. By GST pull-down assays and functional assays with various constructs of ERalpha and STAT5a, it was shown that the C-termini of these two proteins were mainly responsible for this interaction. Furthermore, the interaction between ERalpha and STAT5a was demonstrated to give rise to functional changes in their signaling events. In cell transfection studies, it was shown that ERalpha activation could attenuate PRLR signaling through STAT5a. This ERalpha-mediated attenuation of PRLR signaling was substantiated by observed decreases in the phosphorylation of JAK2 and STAT5a, reduced translocation of STAT5a into the nucleus, and reduced binding of STAT5a onto a GAS-containing nucleotide. Apart from transfected cells, the interaction between ERalpha and STAT5a could also be observed in established breast cancer cell lines of MCF-7 and T-47D in co-immunoprecipitation studies. However, the functional consequence of the interaction in these cancer cells was very different from the transfected HEK293 cells. ER activation could lead to potentiation of PRLR signaling in MCF-7 cells but not in T-47D cells. Conversely, in both MCF-7 and T-47D cells, PRLR activation could lead to attenuation of ER signaling. These data serve to elucidate the mechanisms underlying the ERalpha-STAT5a cross-talk and in demonstrating that the functional consequence of this cross-talk depends on the precise milieus of the intracellular environment.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, PR China
| | | |
Collapse
|
37
|
Baxter JD, Funder JW, Apriletti JW, Webb P. Towards selectively modulating mineralocorticoid receptor function: lessons from other systems. Mol Cell Endocrinol 2004; 217:151-65. [PMID: 15134814 DOI: 10.1016/j.mce.2003.10.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Although there is clinical utility in blocking mineralocorticoid receptor (MR) action, the usefulness of available MR antagonists is limited because of cross-reactivity with the androgen and progesterone receptors (spironolactone) or possibly by low affinity for MR (eplerenone). MR binds aldosterone and physiologic glucocorticoids, such as cortisol, which both can act as MR agonists in epithelial tissues. However, in preliminary studies aldosterone and cortisol appear to induce different conformations in non-epithelial tissues; in the cardiomyocyte, cortisol usually acts as an MR antagonist, whereas in vascular smooth muscle cortisol mimics aldosterone actions if it can access MR, just as it does in the kidney. Thus, there are needs for improved MR antagonists with higher selectivity and potency and, if possible, for compounds that lock MR into specific desirable conformations. Efforts are underway to modulate selectively the action of many nuclear receptors, and insights from one nuclear receptor may be applicable to others given the similarities in structure and function. We have used traditional approaches aided by X-ray crystallography to obtain several classes of selective ligands that modulate thyroid receptor (TR) action. We describe the properties of these selective TR modulators here, and discuss the possibility that similar approaches to ligand design may yield MR interacting compounds with improved specificity and, possibly, tissue specificity.
Collapse
MESH Headings
- Aldosterone/metabolism
- Anti-Inflammatory Agents/administration & dosage
- Anti-Inflammatory Agents/metabolism
- Crystallography, X-Ray
- Epithelium/physiology
- Eplerenone
- Hydrocortisone/administration & dosage
- Hydrocortisone/metabolism
- Mineralocorticoid Receptor Antagonists/chemistry
- Myocytes, Cardiac/metabolism
- Protein Binding
- Protein Structure, Tertiary
- Receptors, Glucocorticoid/agonists
- Receptors, Glucocorticoid/analysis
- Receptors, Glucocorticoid/chemistry
- Receptors, Glucocorticoid/metabolism
- Receptors, Mineralocorticoid/agonists
- Receptors, Mineralocorticoid/chemistry
- Receptors, Mineralocorticoid/metabolism
- Receptors, Thyroid Hormone/antagonists & inhibitors
- Receptors, Thyroid Hormone/chemistry
- Receptors, Thyroid Hormone/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Spironolactone/administration & dosage
- Spironolactone/analogs & derivatives
- Spironolactone/metabolism
- Structure-Activity Relationship
Collapse
Affiliation(s)
- John D Baxter
- Diabetes Center and Metabolic Research Unit, University of California-San Francisco, San Francisco, CA 94143-0540, USA.
| | | | | | | |
Collapse
|
38
|
Blaustein JD. Progestin receptors: neuronal integrators of hormonal and environmental stimulation. Ann N Y Acad Sci 2004; 1007:238-50. [PMID: 14993057 DOI: 10.1196/annals.1286.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although it originally was believed that neuronal steroid hormone receptors require binding to cognate ligand for activation, more recent evidence suggests that the receptors can be activated indirectly by other compounds, such as neurotransmitters and growth factors, acting through their own membrane receptors and specific intracellular signaling pathways. For example, as is the case with facilitation of sexual behavior by progesterone, facilitation of sexual behavior by D(1)/D(5) dopamine receptor agonists is blocked by disruption of progestin receptors. Therefore, some dopamine agonists facilitate sexual behavior at least in part by a progestin receptor-dependent mechanism, as does progesterone. This "ligand-independent activation" of neuronal progestin receptors is not limited to dopamine agonists; a variety of other compounds, as well as mating stimulation, facilitate sexual receptivity by a progestin receptor-dependent process. Steroid hormone receptors also can be regulated by afferent input in another way. Various neurotransmitters upregulate or downregulate steroid hormone receptors in some neurons. This, in turn, presumably confers greater or decreased sensitivity to the particular factors that can activate the particular steroid receptor in those particular neurons. Therefore, steroid hormones are but one class of factors that can regulate and activate steroid hormone receptors. Some additional factors that activate steroid hormone receptors have been identified, as have some factors that can regulate concentrations of receptors. Relatively little is known at this time about the range of neurotransmitters, humoral factors, and intracellular signaling pathways that are involved.
Collapse
Affiliation(s)
- Jeffrey D Blaustein
- Center for Neuroendocrine Studies, University of Massachusetts, Amherst, Massachusetts 01003-9271, USA.
| |
Collapse
|
39
|
Catalano S, Mauro L, Marsico S, Giordano C, Rizza P, Rago V, Montanaro D, Maggiolini M, Panno ML, Andó S. Leptin induces, via ERK1/ERK2 signal, functional activation of estrogen receptor alpha in MCF-7 cells. J Biol Chem 2004; 279:19908-15. [PMID: 14985328 DOI: 10.1074/jbc.m313191200] [Citation(s) in RCA: 191] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Leptin is a hormone with multiple biological actions, produced predominantly by adipose tissue. In humans, plasma levels correlate with total body fat, and high concentrations occur in obese women. Among its functions, leptin is able to stimulate normal and tumor cell growth. We demonstrated that leptin induces aromatase activity in MCF-7 cells evidencing its important role in enhancing in situ estradiol production and promoting estrogen-dependent breast cancer progression. Estrogen receptor alpha (ERalpha), which plays an essential role in breast cancer development, can be transcriptionally activated in a ligand-independent manner. Taking into account that unliganded ERalpha is an effector of mitogen-activated protein kinase (MAPK) signal and that leptin is able, via Janus kinase, to activate the Ras-dependent MAPK pathway, in the present study we investigate the ability of leptin to transactivate ERalpha. We provided evidence that leptin is able to reproduce the classic features of ERalpha transactivation in a breast cancer cell line: nuclear localization, down-regulation of its mRNA and protein levels, and up-regulation of a classic estrogen-dependent gene such as pS2. Transactivation experiments with a transfected reporter gene for nuclear ER showed an activation of ERalpha either in MCF-7 or in HeLa cells. Using a dominant negative ERK2 or the MAPK inhibitor PD 98059, we showed that leptin activates the ERalpha through the MAPK pathway. The N-terminal transcriptional activation function 1 appears essential for the leptin response. Finally, it is worth noting that leptin exposure potentates also the estradiol-induced activation of ERalpha. Thus, we are able to demonstrate that the amplification of estrogen signal induced by leptin occurs through an enhancing in situ E(2) production as well as a direct functional activation of ERalpha.
Collapse
Affiliation(s)
- Stefania Catalano
- Centro Sanitario, University of Calabria, Arcavacata di Rende, Cosenza 87030, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Fowler AM, Solodin N, Preisler-Mashek MT, Zhang P, Lee AV, Alarid ET. Increases in estrogen receptor-alpha concentration in breast cancer cells promote serine 118/104/106-independent AF-1 transactivation and growth in the absence of estrogen. FASEB J 2004; 18:81-93. [PMID: 14718389 DOI: 10.1096/fj.03-0038com] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A common phenotype in breast cancer is the expansion of the estrogen receptor-alpha (ER+) cell population and an inappropriate elevation of ERalpha protein, the latter predisposing patients for a poorer prognosis than those with lower levels of the receptor. A tetracycline-inducible ERalpha overexpression model was developed in the MCF-7 cell line to assess induction of endogenous gene activation and growth in response to elevations in ERalpha protein. Heightened levels of ERalpha resulted in aberrant promoter occupancy and gene activation in the absence of hormone, which was independent of ligand and AF-2 function. This increased receptor activity required the amino-terminal A/B domain and was not inhibited by tamoxifen, which supports an enhancement of AF-1 function, yet was independent of serine-104, 106, and 118 phosphorylation. Ligand-independent transcription was accompanied by an increase in growth in the absence of hormonal stimulation. The results suggest that elevated levels of ERalpha in breast cancer cells can result in activation of receptor transcriptional function in a manner distinct from classical mechanisms that involve ligand binding or growth factor-induced phosphorylation. Further, they describe a potential mechanism whereby increases in ERalpha concentration may provide a proliferative advantage by augmenting ERalpha function regardless of ligand status.
Collapse
Affiliation(s)
- Amy M Fowler
- Department of Physiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
One of the difficulties associated with commercial layer production is the development of osteoporosis in hens late in the production cycle. In light of this fact and because of hens' unique requirements for Ca, many studies have focused on the regulation of Ca and the role of estrogen in this process. The time course of estrogen synthesis over the productive life of hens has been well documented; increased circulating estrogen accompanies the onset of sexual maturity while decreases signal a decline in egg production prior to a molt. Numbers of estrogen receptors decrease with age in numerous tissues. The parallel changes in calcium-regulating proteins, primarily Calbindin D28K, and in the ability of duodenal cells to transport Ca, are thought to occur as a result of the changes in estrogen, and are also reversible by the molt process. In addition to the traditional model of estrogen action, evidence now exists for a possible nongenomic action of estrogen via membrane-bound receptors, demonstrated by extremely rapid surges of ionized Ca in chicken granulosa cells in response to 17beta-estradiol. Estrogen receptors have also been discovered in duodenal tissue, and tamoxifen, which binds to the estrogen receptor, has been shown to cause a rapid increase in Ca transport in the duodenum. In addition, recent evidence also suggests that mineralization of bone per se may not explain entirely the etiology of osteoporosis in the hen but that changes in the collagen matrix may contribute through decreases in bone elasticity. Taken together, these studies suggest that changes in estrogen synthesis and estrogen receptor populations may underlie the age-related changes in avian bone. As with postmenopausal women, dietary Ca and vitamin D are of limited benefit as remedies for osteoporosis in the hen.
Collapse
Affiliation(s)
- M M Beck
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska 68583, USA.
| | | |
Collapse
|
42
|
Lyttle CR, Komm BS, Cheskis BJ. Estrogens: from classical endocrine action to tissue selective action. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2004:1-21. [PMID: 15248501 DOI: 10.1007/978-3-662-05386-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- C R Lyttle
- Wyeth Research, Women's Health Research Institute, Collegeville, PA 19426, USA.
| | | | | |
Collapse
|
43
|
Gélinas S, Bureau G, Valastro B, Massicotte G, Cicchetti F, Chiasson K, Gagne B, Blanchet J, Martinoli MG. Alpha and beta estradiol protect neuronal but not native PC12 cells from paraquat-induced oxidative stress. Neurotox Res 2004; 6:141-8. [PMID: 15325966 DOI: 10.1007/bf03033216] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oxidative stress is currently considered a mediator of cell death in several neurodegenerative diseases. Notably, it may play an important role in the degeneration of dopamine neurons of the substantia nigra in Parkinson's disease. We examined the effect of a strong oxidant, the herbicide paraquat, on cell distress using native and neuronal pheochromocytoma PC12 cells. Paraquat administration for 8 hours induced a significant cellular death in both native and in neuronal PC12 cells. Since the anti-oxidant properties of estrogens may promote neuroprotection in vitro and in vivo, we then investigated the ability of estradiol stereoisomers, 17alpha-estradiol and 17- beta-estradiol, to rescue PC12 cells submitted to paraquat-induced oxidative stress. Our results show a protective effect of both estradiol stereoisomers in neuronal PC12 cells treated with paraquat, whereas this effect could not be observed in native PC12 cells. We also demonstrate that estrogen receptor beta protein expression is modulated by paraquat administration in native PC12 cells, while paraquat does not change estrogen receptor beta ?expression in neuronal PC12 cells. Paraquat also decreases estrogen receptor alpha in neuronal PC12 cells, thus suggesting new routes for paraquat to collapse cellular metabolism. Besides, the oxidation of dihydrodhodamine-123 into fluorescent rhodamine in the presence of paraquat but not in presence of paraquat and 17 alpha-estradiol or 17 beta-estradiol, sustain a possible direct scavenging role of both estradiol stereoisomers.
Collapse
Affiliation(s)
- Sylvie Gélinas
- Neurosciences Research Group, Department of Biochemistry, Université du Québec á Trois-Riviéres, Trois-Riviéres, G9A 5H7, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Rotchell JM, Ostrander GK. Molecular markers of endocrine disruption in aquatic organisms. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2003; 6:453-496. [PMID: 12888444 DOI: 10.1080/10937400306476] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
A wide range of organic contaminant compounds prevalent in the aquatic environment has been shown to exhibit hormone-disrupting activity. The actual potency of such compounds are low compared with endogenous hormones, such as 17beta-estradiol, but may still produce detrimental biological effects. Induced hormone levels are routinely measured using commercial testing kits, though these fail to relate to actual effects. Field and laboratory studies on the biological effects of environmental estrogens have, in the past, largely relied on assays of vitellogenin (vtg) induction in male fish, reduced growth in testes formation, and intersex incidence. Here, we critically review the current and potential application of molecular techniques in assessing the adverse biological reproductive effects of endocrine-disrupting chemicals in aquatic organisms. The role of fish (estrogen, androgen, and progestogen) hormone receptors and invertebrate (ecdysone) hormone receptor, egg production (vtg and chorion) proteins, steroid biosynthesis enzymes (aromatase, sulfotransferase, and hydroxysteroid dehydrogenase), DNA damage, apoptosis, and their potential development as biomarkers are discussed in turn. In each case, the sequences characterized are presented and homologies across species are highlighted. Molecular methods of gauging vtg and zona radiata (ZR) expression and protein concentrations have included immunoassay and reverse transcription polymerase chain reaction (RT-PCR). Suggestions for the isolation for key gene expression products (aromatase, ZR, and vtg, for instance), from a wider range of fish species using degenerate primers, are given. Endocrine disruption in invertebrates has received less attention compared with fish, partly because the knowledge regarding invertebrate endocrinology is limited. Here we review and suggest alternate isolation strategies for key players in the imposex induction process: vitellin (Vn), aromatase, and Ala-Pro-Gly-Trp (APGW) amide neurohormone. Current molecular-level techniques rely on ligand-binding assays, enzyme-linked immunosorbent assay (ELISA), and, more recently, gene expression. In the future, more reliance will be placed on the development of gene expression assays using reporter systems combined with cross-species PCR-based or polyclonal antibody-based assays. We discuss the use of recombinant receptors as a means of primary screening of environmental samples for estrogenicity and antiestrogenicity, which avoids species and seasonal variation in receptor response to ligand binding, a recognized problem of earlier bioassays. Most exciting is the potential that microarray and proteomics approaches have to offer. Such techniques are now used routinely in medical research to identify specific genes and proteins affected by treatment with endocrine disruptors, including estradiol. The technique has yet to be used to screen aquatic organisms, but it has the potential to implicate previously unsuspected estradiol-sensitive genes that may later become molecular markers of endocrine disruption.
Collapse
Affiliation(s)
- Jeanette M Rotchell
- Centre for Environmental Research, School of Chemistry, Physics, and Environmental Science, University of Sussex, Falmer, Brighton, UK
| | | |
Collapse
|
45
|
Konstantinidou AE, Korkolopoulou P, Mahera H, Kotsiakis X, Hranioti S, Eftychiadis C, Patsouris E. Hormone receptors in non-malignant meningiomas correlate with apoptosis, cell proliferation and recurrence-free survival. Histopathology 2003; 43:280-90. [PMID: 12940781 DOI: 10.1046/j.1365-2559.2003.01712.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIMS A retrospective immunohistochemical and statistical analysis of patients with non-malignant meningiomas was undertaken to determine the correlation of steroid hormone receptor status with apoptosis, tumour cell proliferation, clinicopathological characteristics and prediction of recurrence. METHODS AND RESULTS Paraffin sections from 51 primary intracranial totally resected benign and atypical meningiomas were immunohistochemically evaluated for the expression of progesterone (PR), oestrogen (ER) and androgen (AR) receptors, apoptotic rate, Bcl-2, p53 and Ki67 antigens. In addition to the above parameters, the mitotic index and the patients' clinicopathological data were statistically correlated and entered in a recurrence-free survival analysis. A high level of apoptotic cell death was associated with loss of PR expression by logistic regression analysis (P = 0.016). An inverse correlation existed between the mitotic index and PR counts (P = 0.009), while high Ki67 values correlated with increased ARs (P = 0.041). Atypical meningiomas had a lower ER staining score (P = 0.036). Multivariate analysis indicated that the absence of PR and large tumour size were significant factors for shorter disease-free intervals. CONCLUSIONS The results suggest that ER expression is lost or reduced in atypical meningiomas, whereas loss of PR expression is an indicator of increased apoptosis and early recurrence. PRs and ARs may also influence tumour cell proliferation.
Collapse
Affiliation(s)
- A E Konstantinidou
- Department of Pathology, Faculty of Medicine, National Kapodistrian University of Athens, Greece.
| | | | | | | | | | | | | |
Collapse
|
46
|
Graham M, Cossette L, Gélinas S, Martinoli MG. In vitro modulation of prolactin mRNA by toxaphene and 3,3',4,4'-tetrachlorobiphenyl. ENVIRONMENTAL RESEARCH 2003; 92:207-212. [PMID: 12804517 DOI: 10.1016/s0013-9351(02)00093-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
It is now well recognized that many environmental contaminants are capable of disrupting endocrine processes in a variety of species, including birds, mammals, reptiles, and fish. Among these contaminants are toxaphene and polychlorinated biphenyls (PCBs), two of the most prevalent contaminants present in aquatic food chains of the Great Lakes and the Canadian Arctic region. We set out to investigate the possible endocrine-modulating activities of toxaphene, the PCB congener 3,3',4,4'-tetrachlorobiphenyl (TeCB), an equimolar mixture of both compounds (toxaphene/TeCB), and estradiol (E(2)) (E(2)/toxaphene, E(2)/TeCB) on prolactin (PRL) mRNA expression. Concentrations ranging from 10(-7) to 10(-11)M for both toxaphene and TeCB were assayed but only toxaphene modulated PRL mRNA levels, as determined by relative quantitative reverse transcriptase-polymerase chain reaction. Maximal induction by toxaphene was seen at 10(-7)M, resulting in a fourfold increase in PRL mRNA levels. No interactions were observed for combinations of the test substances. Our study demonstrates that toxaphene may exhibit estrogen-like activity by modulating PRL mRNA levels in GH(3) cells.
Collapse
Affiliation(s)
- Meghan Graham
- Department of Biochemistry, Université du Québec à Trois-Rivières, Trois-Rivières, Qué., Canada G9A 5H7
| | | | | | | |
Collapse
|
47
|
Gagné B, Gélinas S, Bureau G, Lagacé B, Ramassamy C, Chiasson K, Valastro B, Martinoli MG. Effects of estradiol, phytoestrogens, and Ginkgo biloba extracts against 1-methyl-4-phenyl-pyridine-induced oxidative stress. Endocrine 2003; 21:89-95. [PMID: 12777708 DOI: 10.1385/endo:21:1:89] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2003] [Revised: 01/02/2003] [Accepted: 02/06/2003] [Indexed: 11/11/2022]
Abstract
Oxidative stress has been recently considered as a mediator of nerve cell death in several neurodegenerative diseases. We studied the effect of the parkinsonism-inducing toxine 1-methyl-4-phenyl-pyridine (MPP+) on several parameters of cell distress using native and neuronal PC12 cells. Then, since estrogens have been reported to prevent neuronal degeneration caused by oxidative damage, we investigated the ability of 17beta- estradiol (E2); two Ginkgo biloba extracts, EGb 761 and Cp 202; as well as two flavonoids, quercetin and kaempferol, to rescue PC12 cells submitted to MPP+- induced oxidative stress. Our results consistently show that both Ginkgo biloba extracts could prevent cell death in native and neuronal PC12 cells, while in neuronal PC12 cells also quercetin and E2 could reverse MPP+ neurotoxic effet. Western blot analysis demonstrated that MPP+ injuries might modulate dopamine transporter (DAT) protein expression but not estrogen receptor beta (ERbeta) protein expression. EGb 761 and Cp 202 also modulate DAT and ERbeta protein expression in neuronal cells. From these studies, we outline the importance of testing estrogen-like plant-derived molecules as potent antioxidants and examine their effect on protein expression.
Collapse
Affiliation(s)
- Benoit Gagné
- Department of Biochemistry, Research Group in Neuroscience, Université du Québec à Trois-Rivières, Trois-Rivières, Canada
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Ray S, Rastogi R, Kumar A. Current status of estrogen receptors. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2003; 59:201-32. [PMID: 12458968 DOI: 10.1007/978-3-0348-8171-5_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Increasing knowledge on structure and function of estrogen receptors is providing information on the mechanism of action of estrogen agonists, as well as antagonists, and in understanding their tissue-selective action. However, there are still many factors associated with estrogen response which are poorly understood. Therefore, the task of designing a tissue-selective estrogen for use as a pharmaceutical in estrogen-dependent disorders remains an uncertain game. This review provides information on the current status of estrogen receptors for a better understanding.
Collapse
Affiliation(s)
- Suprabhat Ray
- Medicinal Chemistry Division, Central Drug Research Institute, Lucknow 226001, India.
| | | | | |
Collapse
|
49
|
Maruvada P, Baumann CT, Hager GL, Yen PM. Dynamic shuttling and intranuclear mobility of nuclear hormone receptors. J Biol Chem 2003; 278:12425-32. [PMID: 12506123 DOI: 10.1074/jbc.m202752200] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We expressed green fluorescent protein (GFP) chimeras of estrogen, retinoic acid, and thyroid hormone receptors (ERs, RARs, and TRs, respectively) in HeLa cells to examine nucleocytoplasmic shuttling and intranuclear mobility of nuclear hormone receptors (NRs) by confocal microscopy. These receptors were predominantly in the nucleus and, interestingly, underwent intranuclear reorganization after ligand treatment. Nucleocytoplasmic shuttling was demonstrated by heterokaryon experiments and energy-dependent blockade of nuclear import and leptomycin-dependent blockade of nuclear export. Ligand addition decreased shuttling by GFP-ER, whereas heterodimerization with retinoid X receptor helped maintain TR and RAR within the nucleus. Intranuclear mobility of the GFP-NRs was studied by fluorescence recovery after photo-bleaching +/- cognate ligands. Both GFP-TR and GFP-RAR moved rapidly in the nucleus, and ligand binding did not significantly affect their mobility. In contrast, estrogen binding decreased the mobility of GFP-ER and also increased the fraction of GFP-ER that was unable to diffuse. These effects were even more pronounced with tamoxifen. Co-transfection of the co-activator, SRC-1, further slowed the mobility of liganded GFP-ER. Our findings suggest estradiol and tamoxifen exert differential effects on the intranuclear mobility of GFP-ER. They also show that ligand-binding and protein-protein interactions can affect the intracellular mobility of some NRs and thereby may contribute to their biological activity.
Collapse
Affiliation(s)
- Padma Maruvada
- Molecular Regulation and Neuroendocrinology Section, Clinical Endocrinology Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
50
|
Sipilä S, Poutamo J. Muscle performance, sex hormones and training in peri-menopausal and post-menopausal women. Scand J Med Sci Sports 2003; 13:19-25. [PMID: 12535313 DOI: 10.1034/j.1600-0838.2003.20210.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Age-related deterioration in muscle performance is one of the major reasons for decreased functional capacity and disability in older people. In women, impaired muscle performance has already been observed during peri-menopause in concert with rapid and dramatic decrease in ovarian hormone production. This observation suggests that female sex steroids may have an important role among other agents in regulating muscle performance in middle-aged and older women. Previous experimental studies have shown that hormone replacement therapy (HRT) and intensive physical training have positive effects on muscle force and explosive power in healthy post-menopausal women. Hormone replacement therapy in combination with physical training may exert even greater gains in muscle performance than HRT and training alone. Despite the significant mean increase in muscle force and power by HRT and/or training, a considerable variability in the individual responses is observed. The mechanism, by which female sex steroids act on muscle performance, is still unclear. Therefore, more research is needed in order to explore all the pathways, by which these steroids could act on skeletal muscle in peri-menopausal and post-menopausal women.
Collapse
Affiliation(s)
- S Sipilä
- Department of Health Sciences, University of Jyväskylä, Finland
| | | |
Collapse
|